09.01.2013 Views

Oral Presentations - Federation of Clinical Immunology Societies

Oral Presentations - Federation of Clinical Immunology Societies

Oral Presentations - Federation of Clinical Immunology Societies

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>Clinical</strong> <strong>Immunology</strong> (2007) 123, S3–S128<br />

ABSTRACTS<br />

<strong>Oral</strong> <strong>Presentations</strong>: Friday, June 8<br />

#1201- Therapies to Induce Tolerance<br />

Prevention and Treatment <strong>of</strong> Autoimmune Arthritis<br />

Using IL-12 Gene-Silenced Dendritic Cells<br />

Friday, June 8<br />

10:45 am−11:05 am<br />

Wei-Ping Min, Scientist, University <strong>of</strong> Western Ontario,<br />

London, ON, Canada, Xiufen Zheng, Fellow, University <strong>of</strong><br />

Western Ontario, London, ON, Canada, Igor Popov, Fellow,<br />

University <strong>of</strong> Western Ontario, London, ON, Canada, Xusheng<br />

Zhang, Fellow, University <strong>of</strong> Western Ontario, London, ON,<br />

Canada, Mu Li, Fellow, University <strong>of</strong> Western Ontario,<br />

London, ON, Canada, Hongtao Sun, Fellow, University <strong>of</strong><br />

Western Ontario, London, ON, Canada, Motohiko Suzuki,<br />

Fellow, University <strong>of</strong> Western Ontario, London, ON, Canada,<br />

Costin Vladau, Student, University <strong>of</strong> Western Ontario,<br />

London, ON, Canada, Bertha Garcia, Pr<strong>of</strong>essor, University <strong>of</strong><br />

Western Ontario, London, ON, Canada, Robert Inman,<br />

Pr<strong>of</strong>essor, Toronto Western Hospital, Toronto, ON, Canada<br />

We have recently demonstrated that dendritic cell(DC)mediated<br />

immune modulation and deviation can be accomplished<br />

through RNA interference (RNAi), highlighting the<br />

therapeutic potential <strong>of</strong> RNAi-modified DC as antigenspecific<br />

tolerogenic vaccines. To date, an RNAi-based<br />

immune therapy for autoimmune diseases has not been<br />

reported. The current study was designed to develop siRNAmodified<br />

DC as antigen-specific, tolerogenic vaccines for<br />

prevention and intervention <strong>of</strong> autoimmune arthritis. Using<br />

small interfering RNA (siRNA) that specifically targets IL-<br />

12p35 gene (IL-12 siRNA), we have generated a type <strong>of</strong> DC<br />

that exhibits multiple tolerogenic characteristics. After<br />

inducation <strong>of</strong> RNAi in DC using siRNA, the expression <strong>of</strong> IL-<br />

12 was inhibited as determined by real time PCR and flow<br />

cytometry. IL-12 silenced DC suppressed Tcell response in an<br />

MLR, and impaired Th1 differentiation in vitro. Immunization<br />

with IL-12 gene-silenced and type II collagen (CII)-pulsed DC<br />

(CII-pulsed/gene-silenced DC) resulted in antigen-specific<br />

non-responsiveness. Vaccination with CII-pulsed/genesilenced<br />

DC prevented collagen-induced arthritis (CIA)<br />

onset in a murine rheumatoid arthritis model. Furthermore,<br />

administration <strong>of</strong> CII-pulsed/gene-silenced DC was sufficient<br />

doi:10.1016/j.clim.2007.03.179<br />

available at www.sciencedirect.com<br />

www.elsevier.com/locate/yclim<br />

to inhibit progression <strong>of</strong> CIA. The therapeutic effects were<br />

further evidenced by decreased clinical score in CIA,<br />

inhibited inflammatory infiltrates in joints, and suppressed<br />

T cell and B cell responses to CII. In conclusion, this study is<br />

the first to demonstrate the therapeutic utilization <strong>of</strong> RNAimodified<br />

DC as antigen-specific tolerogenic vaccines for<br />

autoimmune arthritis.<br />

doi:10.1016/j.clim.2007.03.180<br />

#1202- Commensal Flora and the Immune Response<br />

Helicobacter Pylori in the Pathogenesis <strong>of</strong> Chronic<br />

Autoimmune Pancreatitis<br />

Friday, June 8<br />

10:45 am−11:05 am<br />

Antonio Puccetti, Pr<strong>of</strong>essor <strong>of</strong> Medicine, Institute G. Gaslini,<br />

Genova, Italy, Claudio Lunardi, Pr<strong>of</strong>essor <strong>of</strong> Medicine,<br />

University <strong>of</strong> Verona-Department <strong>of</strong> Internal Medicine,<br />

Verona, Italy, Luca Frullloni, Pr<strong>of</strong>essor <strong>of</strong> Gastroenterology,<br />

Department <strong>of</strong> Gastroenterology, University <strong>of</strong> Verona,<br />

Verona, Italy, Caterina Bason, Research Fellow, University <strong>of</strong><br />

Verona-Department <strong>of</strong> Internal Medicine, Verona, Italy,<br />

Dimitri Peterlana, PhD Student, University <strong>of</strong> Verona-<br />

Department <strong>of</strong> Internal Medicine, Verona, Italy<br />

Autoimmune chronic pancreatitis (ACP) is a type <strong>of</strong> pancreatitis<br />

characterized by a chronic inflammatory process<br />

with prominent lymphocyte infiltration leading to fibrosis <strong>of</strong><br />

the pancreas and eventually to organ dysfunction. The cause<br />

<strong>of</strong> the disease is still unknown. Current evidence suggests an<br />

autoimmune origin for ACP, based on its frequent association<br />

with other autoimmune diseases (rheumatoid arthritis,<br />

Sjogren’s syndrome, and inflammatory bowel disease) and<br />

on the presence <strong>of</strong> immunological abnormalities, including<br />

hypergammaglobulinemia, elevated serum IgG4 levels and the<br />

presence <strong>of</strong> autoantibodies, such as antibodies against<br />

carbonic anhydrase II. However, little is known about its<br />

actual pathogenesis. In order to clarify the pathogenesis <strong>of</strong> the<br />

disease, we screened a random peptide library with pooled<br />

IgG immunoglobulins derived from 20 patients with ACP.


S4 Abstracts<br />

Among the identified peptides, one was recognized by the<br />

majority <strong>of</strong> patients' sera, but not by sera <strong>of</strong> normal donors and<br />

<strong>of</strong> patients with other autoimmune diseases. The peptide<br />

showed homology with an Helicobacter pylori derived protein<br />

and with carbonic anhydrase 5B, a protein highly expressed in<br />

pancreas, kidney and salivary glands, and with lact<strong>of</strong>errin,<br />

considered an important autoantigen target in ACP. Antipeptide<br />

antibodies, affinity purified from patients' sera<br />

recognize the helicobacter derived protein, carbonic anhydrase<br />

5B and lact<strong>of</strong>errin. Moreover antibodies against the<br />

bacterial epitope can be detected in the vast majority <strong>of</strong><br />

patients with ACP. Our findings suggest that Helicobacter<br />

pylori infection may be linked to the pathogenesis <strong>of</strong> ACP<br />

through a molecular mimicry mechanism and that carbonic<br />

anhydrase 5B can be considered a novel autoantigen in ACP.<br />

doi:10.1016/j.clim.2007.03.181<br />

OR.1 Foxp3 (Scurfy) Mutation Greatly Accelerates<br />

Diabetes in Insulin-specific TCR Transgenic Mice<br />

Jean Jasinski, University <strong>of</strong> Colorado Health Sciences Center,<br />

Aurora, CO, Maki Nakayama, Fellow, University <strong>of</strong> Colorado<br />

Health Sciences Center, Barbara Davis Center, Aurora, CO,<br />

Kelly Johnson, Pr<strong>of</strong>essional Research Assistant, University <strong>of</strong><br />

Colorado Health Sciences Center, Barbara Davis Center,<br />

Aurora, CO, George Eisenbarth, Executive Director, Barbara<br />

Davis Center, Aurora, CO, Liping Yu, Senior Research<br />

Assistant, University <strong>of</strong> Colorado Health Sciences Center,<br />

Barbara Davis Center, Aurora, CO<br />

Previously we developed and described the development<br />

<strong>of</strong> a transgenic mouse expressing an insulin-specific (ins2<br />

B:9–23) T cell receptor mouse model (BDC12-4.1). This<br />

pathogenic model exhibited spontaneous diabetes development<br />

on a backcross-1 (FVB×NOD)×NOD background. Disease<br />

development was heterogeneous and dependent on both I-<br />

Ag7 and RAG-deficient genotypes with relatively slow<br />

development <strong>of</strong> diabetes such that the median age <strong>of</strong> onset<br />

<strong>of</strong> diabetes was 49 weeks, and less than 10% <strong>of</strong> mice<br />

developed diabetes prior to 10 weeks <strong>of</strong> age. To test the<br />

hypothesis that the 12-4.1 transgenic T cells, even on a<br />

RAG−/− background, were heterogeneous with development <strong>of</strong><br />

both regulatory and pathogenic T cells, we crossed NOD<br />

congenic BDC12-4.1 mice with C57BL/6 Foxp3 mutant mice.<br />

Expression <strong>of</strong> the single BDC12-4.1 TCR on a RAG-deficient<br />

background rescues the mouse from the scurfy phenotype at<br />

the BC1 (C57BL/6×NOD)×NOD generation. All RAG-deficient<br />

Foxp3 mutant mice (8/8) expressing the BDC12-4.1 TCR develop<br />

diabetes between 5 and 10 weeks <strong>of</strong> age with aggressive<br />

destruction <strong>of</strong> essentially all beta cells whereas prediabetic<br />

mice exhibit invasive insulitis. Disease develops equally in<br />

homozygous (I-Ag7/g7) and heterozygous (I-Ag7/b) mice<br />

significantly earlier and with greater uniformity than previously<br />

described (Pb0.0001). Disease can be adoptively transferred<br />

from diabetic animals to NOD.scid mice. These studies provide<br />

evidence that a single transgenic TCR generates both pathogenic<br />

and Foxp3-dependent regulatory T cells and provides an<br />

experimental model to test therapies <strong>of</strong> diabetes prevention<br />

without Foxp3 dependent regulatory T cells.<br />

doi:10.1016/j.clim.2007.03.182<br />

OR.2 Regulatory T Cells Require Serum for<br />

Suppression <strong>of</strong> Effector T Cell Proliferation and<br />

Express Stable Membrane-bound CD25<br />

Todd Brusko, Postdoctoral Associate, University <strong>of</strong> Florida,<br />

College <strong>of</strong> Medicine, Gainesville, FL, Clive Wasserfall,<br />

Biological Scientist, University <strong>of</strong> Florida, College <strong>of</strong><br />

Medicine, Department <strong>of</strong> Pathology, Gainesville, FL, Kieran<br />

McGrail, Laboratory Technician, University <strong>of</strong> Florida,<br />

College <strong>of</strong> Medicine, Department <strong>of</strong> Pathology, Gainesville,<br />

FL, Marcus Moore, Laboratory Technician, University <strong>of</strong><br />

Florida, College <strong>of</strong> Medicine, Department <strong>of</strong> Pathology,<br />

Gainesville, FL, Alyssa Huegel, Laboratory Technician,<br />

University <strong>of</strong> Florida, College <strong>of</strong> Medicine, Department <strong>of</strong><br />

Pathology, Gainesville, FL, Desmond Schatz, Pr<strong>of</strong>essor <strong>of</strong><br />

Pediatric Medicine, University <strong>of</strong> Florida, College <strong>of</strong><br />

Medicine, Department <strong>of</strong> Pediatrics, Gainesville, FL, Mark<br />

Atkinson, Pr<strong>of</strong>essor, University <strong>of</strong> Florida, College <strong>of</strong><br />

Medicine, Department <strong>of</strong> Pathology, Gainesville, FL<br />

CD4+CD25+ regulatory T cells (Treg) suppress effector T<br />

cell (Teff) functions and are essential for maintaining<br />

peripheral tolerance. Recent work suggests that IL-2 signaling<br />

is crucial for proper Treg development and function. In this<br />

study, we assessed the production <strong>of</strong> the soluble form <strong>of</strong> CD25<br />

(sCD25) in human Treg or Teff cells, alone and in combination,<br />

following polyclonal activation during an in vitro suppression<br />

assay. We found that surface CD25 is relatively stable on Treg<br />

in vitro, whereas CD25 expression on isolated Teff cells is<br />

upregulated following activation; subsequently resulting in<br />

high levels <strong>of</strong> sCD25 detected in culture supernatants (mean +<br />

SD, 682.2+367.1 vs. 5369.5+ 2575.6 pg/ml for Treg vs. Teff<br />

cells respectively; N=7, P=0.004). The production <strong>of</strong> sCD25<br />

can occur in an autocrine fashion and correlates with T cell<br />

proliferation (r=0.76, Pb0.0001). To eliminate the influence<br />

<strong>of</strong> serum sCD25, our studies were also conducted under<br />

serum-free conditions. Surprisingly, under these conditions,<br />

Treg fail to suppress Teff cell proliferation (% suppression at a<br />

1:1 Treg to Teff ratio; −197.8+ 270.6 for serum-free medium<br />

vs. 70.4+17.3 with 1.0% serum; P=0.04). Despite this<br />

functional defect, Treg cells still maintained their capacity<br />

to inhibit Teff cell cytokine production (e.g., IFN-γ responses<br />

were suppressed 75.6% in co-culture vs. Teff alone, Pb0.05).<br />

These data highlight a mechanism by which Treg may obtain<br />

qualitatively greater IL-2 signals than Teff cells and suggest<br />

that serum is required for full Treg activity.<br />

doi:10.1016/j.clim.2007.03.183<br />

OR.3 TGF-β-induced TCR-Tg/NOD Regulatory T Cells<br />

Suppress Both Diabetes Transfer and Islet Graft<br />

Destruction in an Antigen-dependent Manner<br />

Daniel Tonkin, Graduate Student, University <strong>of</strong> Colorado<br />

Health Sciences Center, Department <strong>of</strong> <strong>Immunology</strong>, Denver,<br />

CO, Brenda Bradley, Senior Pr<strong>of</strong>essional Research Assistant,<br />

University <strong>of</strong> Colorado Health Sciences Center, Department<br />

<strong>of</strong> <strong>Immunology</strong>, Denver, CO, Gene Barbour, Pr<strong>of</strong>essional<br />

Research Assistant, University <strong>of</strong> Colorado Health Sciences<br />

Center, Department <strong>of</strong> <strong>Immunology</strong>, Denver, CO, Kathryn<br />

Haskins, Pr<strong>of</strong>essor, University <strong>of</strong> Colorado Health Sciences<br />

Center, Department <strong>of</strong> <strong>Immunology</strong>, Denver, CO


Abstracts<br />

Regulatory T cells (Tregs) are understood to fall into<br />

two categories: natural Tregs and induced Tregs. Induced<br />

Tregs can be produced by activating CD4 T cells in the<br />

presence <strong>of</strong> TGF- 2 . Such induced Tregs have great promise<br />

for immunotherapy, but it has been unclear if they<br />

require stimulation by antigen in order to suppress in<br />

vivo. We have investigated induced Treg protection in the<br />

NOD mouse model <strong>of</strong> autoimmune diabetes. We produced<br />

TGF- 2 -induced Tregs from the 6.9 TCR-Tg/NOD mouse that<br />

are specific for islet autoantigen. These Tregs express<br />

Foxp3 and produce little IFN- 3 . We have shown that<br />

induced Tregs can prevent transfer <strong>of</strong> diabetes by effector<br />

T cells in NOD mice, where they decrease inflammatory<br />

cytokine production and macrophage infiltration in the<br />

pancreas. In some experiments we transferred cells into a<br />

congenic mouse, the NOD.C6, that specifically lacks the<br />

antigen for the 6.9 TCR-Tg T cells but not for other isletspecific<br />

T cells. The 6.9 TCR-Tg Tregs fail to prevent<br />

diabetes transfer in the NOD.C6 mice, demonstrating that<br />

their function is antigen dependent. In human diabetes,<br />

induced Tregs might protect islet grafts from immune<br />

destruction. We grafted NOD.scid islets into NOD.scid.C6<br />

recipients after streptozotocin treatment, and demonstrated<br />

that induced Tregs can prevent islet graft<br />

destruction by effector T cells. When we grafted NOD.<br />

scid.C6 islets, the 6.9 TCR-Tg islets failed to protect.<br />

With these antigen-free islets, we have demonstrated that<br />

induced Tregs require activation by their antigen in the<br />

islet graft in order to protect the graft from autoimmune<br />

destruction.<br />

doi:10.1016/j.clim.2007.03.184<br />

OR.4 Leptin Modulates Treg Cell Activity in Human<br />

SLE<br />

Antonio La Cava, Associate Pr<strong>of</strong>essor, University <strong>of</strong><br />

California, Los Angeles, Los Angeles, CA, Giuseppe<br />

Matarese, Assistant Researcher, University <strong>of</strong> Naples,<br />

Naples, Bevra Hahn, Pr<strong>of</strong>essor and Chief <strong>of</strong> Rheumatology,<br />

University <strong>of</strong> California, Los Angeles, Los Angeles, CA<br />

Leptin is a cytokine/hormone that links nutritional<br />

status with neuroendocrine and immune functions. We<br />

and others have recently shown that leptin can contribute<br />

to the onset and progression <strong>of</strong> organ-specific autoimmunity.<br />

Here, we measured serum leptin in systemic lupus<br />

erythematosus (SLE) patients (n=66) and healthy controls<br />

(n=50) matched for gender, age and ethnicity. Parallel flow<br />

cytometry studies correlated the numbers <strong>of</strong> CD4+<br />

CD25highFoxp3+ T (Treg) cells with serum leptin in the<br />

SLE patients vs. controls. Serum leptin concentration was<br />

significantly higher in SLE patients than in controls<br />

(Pb0.0001) and did not relate to body mass index and/or<br />

therapy. Interestingly, a reduced number <strong>of</strong> CD4+<br />

CD25highFoxp3+ T cells significantly correlated with the<br />

elevated serum leptin in SLE patients (n=9, P=0.02, r 2 =<br />

0.51) but not in controls (n=10, P=0.3, r 2 =0.2). Because<br />

<strong>of</strong> this inverse correlation between CD4+CD25highFoxp3+ T<br />

cells and serum leptin levels, we tested whether leptin<br />

could influence the number and/or activity <strong>of</strong> human Treg<br />

cells. We found that neutralization <strong>of</strong> leptin with antileptin<br />

Ab reversed the hyporesponsiveness <strong>of</strong> human Tregs<br />

in a dose-dependent fashion and Foxp3-expressing Treg<br />

cells expressed elevated levels <strong>of</strong> leptin receptor. Additionally,<br />

anti-leptin Ab promoted both the upregulation <strong>of</strong><br />

Foxp3 and the suppressive in vitro activity <strong>of</strong> the Tregs<br />

cells on effector CD4+CD25− T cells. These data suggest<br />

that leptin can influence the number and activity <strong>of</strong> Treg<br />

cells and may have implication for leptin-based intervention<br />

on Treg cells in SLE.<br />

doi:10.1016/j.clim.2007.03.185<br />

OR.5 Gene Related to Anergy in Lymphocytes (GRAIL)<br />

Expression in CD4+ CD25+ T Regulatory Cells and<br />

Correlation with T:APC Conjugate Formation<br />

Jill Schartner, Graduate Student, University <strong>of</strong> Wisconsin,<br />

Department <strong>of</strong> Pediatrics, Madison, WI, Amanda Timmel,<br />

Technician, University <strong>of</strong> Wisconsin, Department <strong>of</strong><br />

Pediatrics, Madison, WI, William Simonson, Graduate<br />

Student, University <strong>of</strong> Wisconsin, Madison, WI, Christine<br />

Seroogy, Assistant Pr<strong>of</strong>essor, University <strong>of</strong> Wisconsin,<br />

Department <strong>of</strong> Pediatrics, Madison, WI, Anna Huttenlocher,<br />

Associate Pr<strong>of</strong>essor, University <strong>of</strong> Wisconsin, Department <strong>of</strong><br />

Pediatrics, Madison, WI<br />

It has been reported that CD4+CD25+ regulatory T cells<br />

(CD25+ Tregs) have an anergic phenotype and disrupted actin<br />

cap formation in vitro. Recent reports suggest that strong<br />

activation can abrogate the suppressor activity <strong>of</strong> CD25+<br />

Tregs. We have found that the anergy-related E3 ubiquitin<br />

ligase, GRAIL is differentially expressed in CD25+ Tregs. We<br />

sought to correlate the expression pr<strong>of</strong>ile <strong>of</strong> an anergyrelated<br />

gene, GRAIL, with immunologic synapse formation<br />

and suppressor activity in CD25+ Tregs. GRAIL mRNA was<br />

quantitated by real-time QPCR in CD25+ Tregs activated with<br />

anti-CD3/CD28 or anti-CD3 conjugated beads. DO11.10 CD4+<br />

Tcell lines were transduced with GRAIL-IRES-GFP or GFP-only<br />

expressing retrovirus. The ability <strong>of</strong> these cells to form<br />

conjugates with OVA323–339 loaded antigen presenting cells<br />

(APCs) was evaluated via FACS. Microscopy was used to<br />

evaluate localization <strong>of</strong> LFA-1, actin, and GRAIL during<br />

conjugate formation. Strong activation <strong>of</strong> CD25+ Tregs with<br />

anti-CD3/CD28 beads resulted in a 10-fold reduction in GRAIL<br />

mRNA. In contrast, activation <strong>of</strong> CD25+ Tregs with anti-CD3<br />

beads resulted in stable expression <strong>of</strong> GRAIL mRNA. Forced<br />

expression <strong>of</strong> GRAIL in a DO11.10 T cells sufficiently converts<br />

these cells to a suppressor phenotype and resulted in a 2-fold<br />

reduction in their ability to form conjugates with OVA323–<br />

339 loaded APCs. Following conjugation with anti-CD3coated<br />

beads GRAIL expressing T cells exhibited impaired<br />

LFA-1 localization and disrupted actin cap formation. These<br />

data suggest that expression <strong>of</strong> GRAIL in CD25+ Tregs is<br />

differentially regulated and dependent on the activation<br />

context and may correlate with suppressor function.<br />

Furthermore, ectopic expression <strong>of</strong> GRAIL correlates with a<br />

suppressor phenotype and disrupted synapse formation.<br />

doi:10.1016/j.clim.2007.03.186<br />

S5


S6 Abstracts<br />

OR.6 Islet Antigen Specific Regulatory T Cells<br />

Isolated From Non-Diabetic Individuals Suppress<br />

Pro-Inflammatory Responses via Cytotoxic Mechanisms<br />

Timothy Tree, Lecturer, Department <strong>of</strong> Immunobiology,<br />

King’s College London, London, Jenifer Lawson, Research<br />

Assistant, Department <strong>of</strong> Immunobiology, King’s College<br />

London, London, Bart Roep, Pr<strong>of</strong>essor, Department <strong>of</strong><br />

Immunohaematology and Blood Transfusion, Leiden<br />

University Medical Center, Leiden, Hannah Edwards,<br />

Research Technician, Department <strong>of</strong> Immunobiology,<br />

King’s College London, London, England, Mark Peakman,<br />

Pr<strong>of</strong>essor <strong>of</strong> <strong>Clinical</strong> <strong>Immunology</strong>, Department <strong>of</strong><br />

Immunobiology, King’s College London, London,<br />

England<br />

Autoreactive T cells recognizing islet ? cell antigens<br />

could represent both pathogenic effectors in the development<br />

<strong>of</strong> type 1 diabetes (T1DM) and mediators <strong>of</strong><br />

tolerance in non-diabetic individuals depending on the<br />

quality <strong>of</strong> response they produce. We have previously<br />

demonstrated that, whereas autoreactive T cells in<br />

patients with T1DM exhibit polarization towards a proinflammatory<br />

T helper 1 (Th1) phenotype, the majority <strong>of</strong><br />

non-diabetic subjects also manifest a response against<br />

islet peptides, but one that shows T regulatory cell (Treg),<br />

IL-10 secreting, bias. We therefore proposed that isletspecific<br />

IL-10 producing T cells may be involved in the<br />

maintenance <strong>of</strong> tolerance to islets by the suppression <strong>of</strong><br />

proinflammatory Th1 cells. To test this hypothosis, we<br />

isolated T cell clones from healthy individuals that secrete<br />

high levels <strong>of</strong> IL-10 in response to islet antigens directly<br />

ex vivo. These CD4+CD25+Foxp3+ Tregs are potent<br />

suppressors <strong>of</strong> proinflammatory Th1 effector cells. Suppression<br />

is not mediated by soluble factors but is cellcontact<br />

dependent, and dependent upon presentation <strong>of</strong><br />

stimulating antigens for both the Treg and effector Th1<br />

cells by the same antigen presenting cell (APC), thus<br />

providing exquisite specificity <strong>of</strong> suppression. These Tregs<br />

express the cytotoxic molecules perforin and granzymes A<br />

and B directly ex vivo and mediate their suppression via<br />

the specific lysis <strong>of</strong> APC presenting high levels <strong>of</strong> islet<br />

antigens. This hitherto un-described population <strong>of</strong> antigen<br />

specific Tregs, which appear to be constitutively present<br />

in the majority <strong>of</strong> non-diabetic individuals, may provide a<br />

suitable target to strengthen tolerance to islets by clinical<br />

immunointervention.<br />

doi:10.1016/j.clim.2007.03.187<br />

OR.7 Rapamycin Promotes Induction <strong>of</strong> CD4+Foxp3+<br />

T Cells that Proliferate in Response to Common<br />

Gamma Chain Cytokines<br />

S. Alice Long, Scientist, Benaroya Research Institute,<br />

Seattle, WA, Megan Van Landeghen, RA II, Benaroya<br />

Research Institute, Seattle, WA, Jane Buckner, Pr<strong>of</strong>essor,<br />

Benaroya Research Institute, Seattle, WA<br />

We and others have shown that CD4+CD25+Foxp3+ T<br />

cells can be induced from CD4+CD25−Foxp3− T cells<br />

(iTReg). Using intracellular staining for Foxp3, we consistently<br />

observe that the CD4+CD25+ population that arises<br />

upon activation <strong>of</strong> CD4+CD25− T cells with polyclonal or<br />

antigenic stimulation is composed <strong>of</strong> both Foxp3− and Foxp3+<br />

T cells. Thus, determining factors that influence the<br />

frequency <strong>of</strong> iTReg may lead to development <strong>of</strong> targeted<br />

therapies. IL-2 is a known growth factor for natural Treg.<br />

Others have observed that treatment <strong>of</strong> patients with<br />

Rapamycin leads to an increased number <strong>of</strong> CD4+CD25+<br />

Foxp3+ Tcells. Here, we demonstrate that in vitro activation<br />

<strong>of</strong> CD4+CD25− T cells in the presence <strong>of</strong> IL-2 or Rapamycin<br />

results in generation <strong>of</strong> functional iTReg. However, combination<br />

<strong>of</strong> both Rapamycin and IL-2 results in a significantly<br />

higher frequency and absolute number <strong>of</strong> iTReg. Analysis <strong>of</strong><br />

Foxp3 over time demonstrates that up-regulation <strong>of</strong> Foxp3<br />

occurs prior to cell division and increases during the course<br />

<strong>of</strong> cell division. Foxp3− and Foxp3+ T cells proliferate in<br />

parallel suggesting that Rapamycin is not significantly<br />

inhibiting the proliferation <strong>of</strong> Foxp3− T cells, but instead,<br />

is preferentially inducing Foxp3 expression in a subset <strong>of</strong><br />

CD25− T cells. The addition <strong>of</strong> IL-2, but also IL-4, IL-7, and<br />

IL-15, facilitates proliferation <strong>of</strong> iTReg. Thus, we demonstrate<br />

that Rapamycin plus IL-2 and/or common gamma<br />

chain cytokines promotes induction and proliferation <strong>of</strong><br />

Foxp3+ T cells. These studies provide rationale for using<br />

Rapamycin in combination with IL-2 to enhance expansion<br />

<strong>of</strong> regulatory T cells in vivo.<br />

doi:10.1016/j.clim.2007.03.188<br />

OR.8 IL-2Rβ Links IL-2R Signaling with Foxp3<br />

Expression<br />

David Soper, Graduate Student, University <strong>of</strong> Washington,<br />

Department <strong>of</strong> <strong>Immunology</strong>, Seattle, WA<br />

Immunological tolerance to self-antigens is a tightly<br />

regulated process. Recent work has demonstrated that<br />

the forkhead family member Foxp3 is a critical element in<br />

the differentiation and function <strong>of</strong> mouse CD4+CD25+<br />

regulatory T (TR) cells. Recent work has suggested an<br />

important role for IL-2 in the development and maintenance<br />

<strong>of</strong> TR cells. To directly assess the effect <strong>of</strong> IL-2<br />

signaling on TR development and function, we analyzed<br />

mice that were genetically deficient in components <strong>of</strong> the<br />

IL-2 receptor (IL-2R). Mice lacking CD25 (IL-2Rα) displayed<br />

a slight decrease in TR cells within the thymus, while<br />

peripheral numbers are unchanged. In contrast, we found<br />

that mice deficient in CD122 (IL-2Rβ) had a pr<strong>of</strong>ound<br />

reduction in both thymic and peripheral TR cells,<br />

coinciding with more rapid development <strong>of</strong> a fatal<br />

lymphoproliferative disease. Expression <strong>of</strong> a Foxp3 transgene<br />

restored TR cells and protected against the onset <strong>of</strong><br />

autoimmunity. Thus, a signal mediated by IL-2Rβ is essential<br />

for the development and homeostasis <strong>of</strong> Foxp3+ TR cells<br />

in vivo.<br />

doi:10.1016/j.clim.2007.03.189


Abstracts<br />

OR.9 High Affinity T Cell Receptors as Therapeutic<br />

Agents<br />

Rebecca Ashfield, Senior Research Project Manager, Avidex<br />

Ltd, Abingdon, Oxfordshire, England, Deborah Sutton,<br />

Group Leader, Cell Biology, Avidex Ltd, Abingdon,<br />

Oxfordshire, England, Giovanna Bossi, Senior Scientist,<br />

Avidex Ltd, Abingdon, Oxfordshire, England, Joanna Brewer,<br />

Group Leader, Cell biology, Avidex Ltd, Abingdon,<br />

Oxfordshire, England, Marco Purbhoo, Research Associate,<br />

Division <strong>of</strong> Molecular and Cell Biology, London, Rebecca<br />

Dennis, Senior Scientist, Avidex Ltd, Abingdon, Oxfordshire,<br />

England, Julia Karbach, Researcher, Ludwig Institute,<br />

Frankfurt, Germany, Tara Mahon, Senior Scientist, Avidex<br />

Ltd, Abingdon, Oxfordshire, England, Brian Cameron, Senior<br />

Scientist, Avidex Ltd, Abingdon, Oxfordshire, England, Bent<br />

Jakobsen, Senior Vice President, Research, Avidex Ltd,<br />

Abingdon, Oxfordshire, England<br />

T cell receptors (TCRs) recognise peptides derived from<br />

disease related antigens, many <strong>of</strong> them intracellular in origin,<br />

which are presented on the cell surface by HLA molecules. The<br />

development <strong>of</strong> TCRs as therapeutic agents has until recently<br />

been impeded by their low natural affinity, and the difficulty <strong>of</strong><br />

expressing membrane-bound receptors as soluble molecules.<br />

We have engineered TCRs to produce stable, soluble proteins<br />

that can be made in large quantities by expression in E. coli.<br />

These monoclonal TCRs (mTCRs) can be affinity matured by<br />

phage display technology to sub-nM affinity, and can be fused to<br />

toxins or immuno-modulatory effector functions to produce<br />

therapeutic proteins suitable for targeting tumours, virally<br />

infected cells or tissues under autoimmune attack. Using this<br />

technology, high affinity mTCRs have been produced which<br />

recognise HLA-A2 specific epitopes from HIVgag (amino acids<br />

77–85) and two tumour associated antigens, NY-ESO (amino<br />

acids 157–165) and Melan-A (amino acids 26–35). Each <strong>of</strong> these<br />

mTCRs has sub-nM affinity and a binding half life <strong>of</strong> several<br />

hours, whilst retaining specificity for the target HLA–epitope<br />

complex. Naturally processed epitopes on the surface <strong>of</strong> antigen<br />

presenting cells are targeted by the high affinity mTCRs, and<br />

can be detected using flow cytometry and fluorescence<br />

microscopy. We present data comparing the antigen density <strong>of</strong><br />

the two tumour associated epitopes, NY-ESO157–165 and<br />

Melan-A26–35, on the surface <strong>of</strong> melanoma cell lines. Furthermore,<br />

we show that the NY-ESO specific mTCR binds specifically<br />

to freshly isolated NY-ESO+, HLA-A2+ myeloma cells.<br />

doi:10.1016/j.clim.2007.03.190<br />

Regulatory T Cells I<br />

Friday, June 8<br />

2:45 pm−4:45 pm<br />

OR.10 From EAE to an MS <strong>Clinical</strong> Trial:<br />

DR2/MOG-35−55 Recombinant T Cell Receptor<br />

Ligand (RTL) Treats Relapse <strong>of</strong> Experimental<br />

Encephalomyelitis in DR2 Transgenic Mice<br />

Halina Offner, Pr<strong>of</strong>essor, Oregon Health and Science<br />

University, Neurology, Portland, OR, Gregory Burrows,<br />

Research Associate Pr<strong>of</strong>essor, Oregon Health and Science<br />

University, Neurology, Portland, OR, Arthur Vandenbark,<br />

Pr<strong>of</strong>essor, Portland VA Medical Center, Neuroimmunology<br />

Research, Portland, OR, Jason Link, Associate Investigator,<br />

Portland VA Medical Center, Neuroimmunology Research,<br />

Portland, OR<br />

To evaluate the ability <strong>of</strong> a newly designed monomeric<br />

recombinant TCR ligand, RTL342M, containing HLA-DR2<br />

peptide-binding domains covalently linked to MOG-35–55<br />

peptide, to prevent and treat both the initial episode and<br />

subsequent relapses <strong>of</strong> experimental autoimmune encephalomyelitis<br />

(EAE) in HLA-DR2 transgenic mice, in preparation<br />

for a safety trial in patients with multiple sclerosis (MS),<br />

single or multiple doses <strong>of</strong> RTL342M were given i.v. or s.c. to<br />

HLA-DR2 mice prior to, at onset, or during relapses <strong>of</strong><br />

paralytic EAE. RTL342M produced rapid (within 24 h), dosedependent<br />

reversal <strong>of</strong> clinical signs <strong>of</strong> paralytic EAE; even a<br />

single dose could produce significant treatment effect.<br />

Multiple daily doses were even more effective than the<br />

same total amount <strong>of</strong> RTL given as a single dose. By<br />

establishing the minimal effective dose, RTLs may be 50<br />

times more potent than molar equivalent doses <strong>of</strong> myelin<br />

peptide alone. RTL342M given prior to induction <strong>of</strong> EAE<br />

prevented disease in most mice, and the remainder could be<br />

successfully re-treated with RTL. Most important for clinical<br />

application, RTL342M was highly effective for treating EAE<br />

relapses when given periodically prior to the relapse or even<br />

after relapses had occurred. These data demonstrate rapid<br />

and potent clinical effects <strong>of</strong> RTL342M at disease onset and<br />

during relapses in EAE, establishing important principles<br />

governing application <strong>of</strong> this novel, highly selective therapeutic<br />

approach for regulating pathogenic Tcells. Data from<br />

this study provide key preclinical information for a phase I<br />

safety study in patients with MS (outline to be presented) that<br />

is now in progress.<br />

doi:10.1016/j.clim.2007.03.191<br />

OR.11 Immunological Efficacy <strong>of</strong> hsp60 Peptide<br />

DiaPep277 Therapy in Human Type 1 Diabetes<br />

Bart Roep, Immunologist, Leiden University Medical Center,<br />

Leiden<br />

An immunogenic peptide (p277) from the 60-kDa heat-shock<br />

protein (hsp60) arrested beta-cell destruction in non-obese<br />

diabetic (NOD) mice. A randomised, double blind, phase Ib/II<br />

clinical trial <strong>of</strong> DiaPep277 peptide treatment was performed in<br />

recent onset type 1 diabetes patients with remaining insulin<br />

production. We studied the immunological efficacy <strong>of</strong> this<br />

peptide therapy and correlated this with clinical outcome.<br />

Forty-eight C-peptide positive patients were assigned subcutaneous<br />

injections <strong>of</strong> 0.2, 1.0 or 2.5 mg p277 (n=12perdosage)at<br />

entry, and 1, 6 and 12 months, or four placebo injections (n=12).<br />

T cell autoimmunity to hsp60, DiaPep277, GAD and tetanus<br />

toxoid (recall response control) were assayed by proliferation<br />

and cytokine secretion assays (ELIspot) on regular intervals until<br />

18 months after first injection. All treated patients at each<br />

dosage <strong>of</strong> peptide demonstrated an altered immune response to<br />

DiaPep277 while the majority <strong>of</strong> placebo-treated patient<br />

remained non-responsive to treatment (p=0.00001), indicating<br />

a 100% efficacy <strong>of</strong> immunization. Cytokine production in<br />

S7


S8 Abstracts<br />

response to therapy was dominated by IL-10, but this pr<strong>of</strong>ile did<br />

not correlate with better preserved beta-cell function. Instead,<br />

patients developing immunological tolerance to Diapep277<br />

showed less decreased C-peptide production than patients<br />

that kept a reactive phenotype. Third party control immune<br />

responses were unaffected by therapy. No potentially adverse<br />

immunological side effects were noted.<br />

DiaPep277 is immunogenic in type 1 diabetic subjects and<br />

has immune modulating properties. Immunological monitoring<br />

allowed to distinguish therapy from placebo treatment<br />

and could determine immunological efficacy. Tolerance to<br />

peptide DiaPep277 may serve as immunological biomarker<br />

measure for clinical efficacy.<br />

doi:10.1016/j.clim.2007.03.192<br />

OR.12 Somatic B-Cell Gene Vaccination with<br />

Anti-DNA Ig Consensus Peptide Protects (NZB × NZW)<br />

F1 Lupus Mice From Renal Disease<br />

Francesca Ferrera, Postdoctoral Fellow, University <strong>of</strong><br />

Genova, Genova, Bevra Hahn, Pr<strong>of</strong>essor and Chief <strong>of</strong><br />

Rheumatology, University <strong>of</strong> California, Los Angeles, Los<br />

Angeles, CA, Marta Rizzi, Postdoctoral Fellow, University<br />

<strong>of</strong> Genova, Genova, Gilberto Filaci, Associate Pr<strong>of</strong>essor,<br />

University <strong>of</strong> Genova, Genova, Francesco Indiveri,<br />

Pr<strong>of</strong>essor, University <strong>of</strong> Genova, Genova, Antonio La Cava,<br />

Associate Pr<strong>of</strong>essor, University <strong>of</strong> California, Los Angeles,<br />

Los Angeles, CA, Antonio La Cava, Associate Pr<strong>of</strong>essor<br />

<strong>of</strong> Medicine, University <strong>of</strong> California, Los Angeles,<br />

Los Angeles, CA<br />

Ig molecules that participate in humoral immune responses<br />

contain epitopes that induce T cell-mediated immune<br />

responses. B cells process and present such epitopes and<br />

activate T cells. We hypothesized that T cells recognizing an Ig<br />

consensus sequence presented by B cells will modulate lupuslike<br />

disease in mice. To test this hypothesis, NZB/W F1 lupus<br />

mice received B cell somatic gene transfer <strong>of</strong> DNA plasmid<br />

encoding an artificial consensus sequence mimicking T cell<br />

determinants <strong>of</strong> murine anti-DNA IgG, or control plasmids.<br />

Treated animals were monitored for production <strong>of</strong> antibody,<br />

development <strong>of</strong> renal disease and phenotype, number, and<br />

function <strong>of</strong> consensus gene-induced Tcells. It was found that the<br />

treatment <strong>of</strong> mice with Ig consensus gene induced TGFβproducing<br />

CD8+CD28− T cells that: (1) suppressed antigenspecific<br />

stimulation <strong>of</strong> CD4+ Tcells in a cell-contact independent<br />

manner; (2) reduced autoantibody production; (3) retarded the<br />

development <strong>of</strong> nephritis, and (4) improved survival <strong>of</strong> treated<br />

animals. Significantly, the adoptive transfer <strong>of</strong> CD8+CD28− T<br />

cells from Ig consensus gene-protected mice into hypergammaglobulinemic<br />

NZB/W F1 mice effectively protected the transferred<br />

mice from development <strong>of</strong> renal disease. We conclude<br />

that genetic expression <strong>of</strong> anti-DNA Ig consensus can induce<br />

immunoregulatory circuits capable <strong>of</strong> delaying development <strong>of</strong><br />

lupus nephritis via the suppression <strong>of</strong> hypergammaglobulinemia<br />

and renal disease.<br />

doi:10.1016/j.clim.2007.03.193<br />

OR.13 Therapeutic Vaccination with a Trivalent<br />

T Cell Receptor Peptide Vaccine Induces<br />

Peptide-specific IL-10-secreting T Cells, Restores<br />

Deficient Foxp3 Expression, and Induces Bystander<br />

Immunomodulation in Multiple Sclerosis Subjects<br />

Arthur Vandenbark, Pr<strong>of</strong>essor, Oregon Health and Science<br />

University, Neurology, Portland, OR, Richard Bartholomew,<br />

Executive Director <strong>of</strong> Research and Development, The<br />

Immune Response Corporation, Carlsbad, CA, Halina<br />

Offner, Pr<strong>of</strong>essor, Oregon Health and Science University,<br />

Neurology, Portland, OR, Dennis Bourdette, Pr<strong>of</strong>essor and<br />

Chairman, Oregon Health and Science University,<br />

Neurology, Portland, OR<br />

We are developing a therapeutic vaccine containing<br />

CDR2 peptides from BV5S2, BV6S5, and BV13S1 emulsified<br />

in IFA as a V gene-specific therapy for RR-MS. This TCR<br />

vaccine is currently being tested in a blinded, placebocontrolled,<br />

200 patient study with reduction <strong>of</strong> new<br />

gadolinium enhancing lesions as the primary endpoint. We<br />

here report new mechanistic data from a recently<br />

completed open-label study. Immunological and clinical<br />

parameters were followed in 27 treatment naïve and 9<br />

previously vaccinated subjects with MS who received<br />

monthly injections <strong>of</strong> the trivalent peptide vaccine over<br />

1 year. TCR vaccination induced high frequencies <strong>of</strong> IL-10secreting<br />

T cells specific for the injected TCR peptides as<br />

well as an apparently broader pattern <strong>of</strong> anti-TCR<br />

specificities. These regulatory specificities induced biphasic<br />

immunomodulation that involved an initial rise<br />

after 9 weeks <strong>of</strong> IL-10-secreting neuroantigen-specific T<br />

cells, in conjunction with potent induction <strong>of</strong> Foxp3+ Treg<br />

cells to normal levels after 12 weeks <strong>of</strong> treatment. These<br />

findings are consistent with vaccine-induced stimulation <strong>of</strong><br />

Foxp3+ Treg cells originating from both CD25(−) and<br />

conventional CD25(+) T cells. Restoration <strong>of</strong> previously<br />

deficient Treg cells could explain the broad reduction <strong>of</strong><br />

TCR-dependent responses <strong>of</strong> both cognate T cells expressing<br />

V genes targeted by the vaccine as well as bystander T<br />

cells expressing different V genes, including those specific<br />

for neuroantigens. These findings demonstrate that therapeutic<br />

vaccination using only three commonly expressed<br />

BV gene determinants can induce a broad immunoregulatory<br />

network in vivo that may help control autoreactive<br />

responses that characterize the inflammatory phase <strong>of</strong> MS.<br />

doi:10.1016/j.clim.2007.03.194<br />

OR.14 Mechanistic Insights Into the Differential<br />

Efficacy <strong>of</strong> GAD65 DNA Vaccine and Anti-CD3<br />

Combination Therapy in Treating New-Onset<br />

Autoimmune Diabetes in RIP-LCMV and NOD Mice<br />

Damien Bresson, Postdoctoral Fellow, La Jolla Institute for<br />

Allergy and <strong>Immunology</strong>, La Jolla, CA, Diane Rottembourg,<br />

Postdoctoral Fellow, La Jolla Institute for Allergy and<br />

<strong>Immunology</strong>, La Jolla, CA, Lisa Togher, Technician, La Jolla<br />

Institute for Allergy and <strong>Immunology</strong>, La Jolla, CA, Matthias<br />

von Herrath, Pr<strong>of</strong>essor, La Jolla Institute for Allergy and<br />

<strong>Immunology</strong>, La Jolla, CA


Abstracts<br />

Type 1 diabetes (T1D) results from a progressive<br />

destruction <strong>of</strong> insulin-producing pancreatic beta cells by<br />

autoaggressive T cells. In this study, we evaluated the<br />

efficacy <strong>of</strong> a combination therapy (CT) (non-Fc binding<br />

anti-CD3 and human GAD65-expressing DNA vaccine) to<br />

reverse new-onset T1D in two animal models (NOD and<br />

RIP-LCMV mice). CT enhanced efficacy only in the RIP-<br />

LCMV model (40% improvement compared to anti-CD3 or<br />

GAD65 vaccine given alone). We evaluated three factors<br />

that could account for such a discrepancy: (i) amount <strong>of</strong><br />

GAD65 protein within the islets <strong>of</strong> Langerhans, (ii) level<br />

and functionality <strong>of</strong> toll-like receptor-9 (TLR9) expressed<br />

by immune cells and (iii) epitopes recognized by GAD65specific<br />

Tregs expanded after CT vs. anti-CD3 alone.<br />

Although similar levels <strong>of</strong> pancreatic GAD65 levels were<br />

found in both animal models, TLR9 expression was slighlty<br />

enhanced in antigen-presenting cells derived from RIP-<br />

LCMV vs. NOD mice. By using a peptide library the GAD65specific<br />

Tregs epitopes were mapped in the C-terminal<br />

domain <strong>of</strong> the protein after CT in the RIP-LCMV but not<br />

NOD mice, thus emphasizing the importance <strong>of</strong> genetic<br />

background in the therapeutic efficacy <strong>of</strong> antigen-specific<br />

immuno-interventions. Furthermore, these CD4+CD25+<br />

Foxp3+GITR+ Tregs also expressed the co-stimulatory<br />

molecule OX40 and protected 50% <strong>of</strong> mice when transferred<br />

in vivo into immunocompetent recipients. The<br />

involvement <strong>of</strong> OX40 in this bystander suppression mechanism<br />

will be studied by antibody blockade and siRNA<br />

technology.<br />

This work was supported by a NIH DK51091, AI44451 and a<br />

U-19 prevention center grant to MVH. DB is supported by a<br />

European Marie-Curie Outgoing Fellowship.<br />

doi:10.1016/j.clim.2007.03.195<br />

OR.15 Mimotopes for Active Immunotherapy <strong>of</strong><br />

Tumors: Allergooncology<br />

Erika Jensen-Jarolim, Pr<strong>of</strong>essor, Medical University Vienna,<br />

Vienna, Angelika B. Riemer, Medicine University Vienna,<br />

Vienna, Regina Knittelfelder, MSc, Medicine University Vienna,<br />

Vienna, Panos Karagiannis, Student, Med University <strong>of</strong> Vienna,<br />

Vienna, Josef Singer, Student, Medicine University Vienna,<br />

Vienna, Eva Untersmayr, Medical University Vienna, Vienna,<br />

Kira Braemswig, Medicine University <strong>of</strong> Vienna, Vienna<br />

Antibodies recognize 3D-epitopes and have the great<br />

potential to sense viable, malignant cells and to destroy<br />

them by various mechanisms. Several chimeric or humanized<br />

antibodies are, therefore, in clinical use today.<br />

However, the effects <strong>of</strong> these passive immunotherapy<br />

treatments are limited by the antibody’s halflife, and<br />

repeated applications have to be given to the patient. In<br />

contrast, active induction <strong>of</strong> these desired antibody<br />

specificities in the patient could be pursued by the<br />

generation <strong>of</strong> peptide epitope mimics – mimotopes –<br />

which are suitable tools for vaccination. By virtue <strong>of</strong><br />

molecular mimicry, these mimotopes induced anti-tumor<br />

antibodies in mice able to attack tumors not only in vitro<br />

but also in vivo in murine transgenic, and in syngenic<br />

transplant models. Interestingly, besides IgG also IgE<br />

antibodies could be induced when mimotopes were<br />

gavaged by the oral route. These IgE antibodies sensitized<br />

mast cells and could be triggered by tumor cells in a<br />

specific fashion. Most importantly, the released mediators<br />

were cytotoxic for tumor cells. Indeed, immunohistochemical<br />

analyses <strong>of</strong> human tumor sections showed that IgE<br />

antibodies are present in tumor lesions, pointing towards<br />

a natural surveillance function <strong>of</strong> this antibody class.<br />

Conclusion: Taken together, we suggest that mimotopes<br />

are candidates for active immunization <strong>of</strong> patients for<br />

tumor prophylaxis or therapy, especially in settings <strong>of</strong><br />

minimal residual disease. Studies supported by BioLife-<br />

Science and grant P-18238-B13 <strong>of</strong> the Austrian Science<br />

Fund FWF.<br />

doi:10.1016/j.clim.2007.03.196<br />

OR.16 Gene Expression Analysis by Microarray<br />

Following Anti-CD3 Antibody Therapy <strong>of</strong> NOD Mice<br />

Hideyuki Iwai, Postdoctoral Fellow, Stanford University,<br />

Department <strong>of</strong> Medicine Division <strong>of</strong> <strong>Immunology</strong> and<br />

Rheumatology, Stanford, CA, Keichi Kodama, Postdoctoral<br />

Fellow, Stanford University, Department <strong>of</strong> Medicine<br />

Division <strong>of</strong> <strong>Immunology</strong> and Rheumatology, Stanford, CA,<br />

Demi Dang, Technician, Stanford University, Department <strong>of</strong><br />

Medicine Division <strong>of</strong> <strong>Immunology</strong> and Rheumatology,<br />

Stanford, CA, C. Garrison Fathman, Pr<strong>of</strong>essor, Stanford<br />

University, Department <strong>of</strong> Medicine Division <strong>of</strong> <strong>Immunology</strong><br />

and Rheumatology, Stanford, CA, Jeffrey A. Bluestone,<br />

Pr<strong>of</strong>essor, Diabetes Center, University <strong>of</strong> California,<br />

San Francisco, San Francisco, CA<br />

Anti-CD3-specific antibodies have the demonstrable<br />

capacity to induce long-term remission <strong>of</strong> overt diabetes<br />

in nonobese diabetic (NOD) mice and have had some<br />

success in C1-peptide preservation when used to treat<br />

recent onset human type I diabetics. The underlying<br />

mechanisms <strong>of</strong> action are unclear. To begin to study<br />

mechanism <strong>of</strong> action, we examined gene expression<br />

following 5 days <strong>of</strong> anti-CD3 antibody therapy <strong>of</strong> NOD<br />

mice, using 41K Agilent mouse genome oligo-microarrays.<br />

This treatment successfully restored euglycemia in NOD<br />

mice treated after the onset <strong>of</strong> hyperglycemia (blood<br />

glucose N250) with 10 μg <strong>of</strong> either conventional anti-CD-3,<br />

IgG1 2C11, or FcR nonbinding IgG3 anti-CD3 mAb, which<br />

cannot cross-link surface CD3 molecules on T cells and is<br />

therefore non-mitogenic. Both antibodies had similar<br />

effect in disease remission. RNA from islets and pancreatic<br />

lymph nodes <strong>of</strong> treated and untreated NOD mice was<br />

isolated on day 12. Microarray analysis was performed<br />

comparing treated mouse tissues to untreated control.<br />

Several hundred genes were dysregulated following therapy.<br />

We hypothesized that genes specifically affected by<br />

anti-CD3 antibody would be in overlapping groups among<br />

those dysregulated by these two antibodies. We will<br />

present data on these overlapping genes and discuss<br />

potential mechanism <strong>of</strong> action <strong>of</strong> anti-CD3 therapy.<br />

doi:10.1016/j.clim.2007.03.197<br />

S9


S10 Abstracts<br />

OR.17 C-Peptide Reactive T Cells Induce<br />

Autoimmune Diabetes: A Novel Mechanism <strong>of</strong> Beta<br />

Cell Autoimmunity<br />

Matteo Levisetti, Assistant Pr<strong>of</strong>essor, Washington<br />

University, Saint Louis, MO<br />

There is extensive experimental evidence supporting an<br />

important role for proinsulin as an autoantigen in the<br />

pathogenesis <strong>of</strong> autoimmune diabetes in both humans and<br />

the NOD mouse model <strong>of</strong> the disease. We identified<br />

spontaneously occurring C-peptide reactive T cells in the<br />

islet-infiltrate and peri-pancreatic lymph nodes <strong>of</strong> prediabetic<br />

mice. CD4 Tcells lines were generated by immunization<br />

with C-peptide in adjuvant and characterized. These T cells<br />

are reactive with full length C-peptide 1 and 2 and are<br />

reactive with freshly isolated mouse beta cells. Furthermore,<br />

these T cells are activated by fixed splenocytes,<br />

demonstrating that C-peptide can be presented in the<br />

absence <strong>of</strong> antigen processing by I-Ag7 bearing antigen<br />

presenting cells. C-peptide reactive T cells transferred<br />

disease into NOD.Scid recipient mice and accelerated<br />

disease in neonatal NOD recipients. These data identify a<br />

new T cell epitope within the proinsulin molecule that gives<br />

rise to diabetogenic T cells in the NOD mouse. In addition,<br />

these data define a novel mechanism for the loss <strong>of</strong> tolerance<br />

to secreted self proteins.<br />

doi:10.1016/j.clim.2007.03.198<br />

Autoantigens<br />

Friday, June 8<br />

2:45 pm−4:45 pm<br />

OR.18 Cytotoxic Preproinsulin-specific CD8 T Cells<br />

in Type 1 Diabetes in Man<br />

Anna Skowera, Postdoctoral Fellow, King’s College London,<br />

Department <strong>of</strong> Immunobiology, London, England, Richard<br />

Ellis, Postdoctoral Fellow, King’s College London,<br />

Department <strong>of</strong> Immunobiology, London, England, Timothy<br />

Tree, Lecture, King’s College London, Department <strong>of</strong><br />

Immunobiology, London, England, Mark Peakman, Pr<strong>of</strong>essor<br />

<strong>of</strong> <strong>Clinical</strong> <strong>Immunology</strong>, King’s College London, Department<br />

<strong>of</strong> Immunobiology, London, England, Sefina Arif,<br />

Postdoctoral Fellow, King’s College London, Department <strong>of</strong><br />

Immunobiology, London, England<br />

Type 1 diabetes (T1D) is a T cell mediated autoimmune<br />

disease in which the insulin producing pancreatic islet<br />

beta-cells are selectively eliminated. The mechanism <strong>of</strong><br />

this precisely targeted destruction is unknown, but<br />

cytotoxic T cells (CTLs) recognizing beta cell-specific<br />

targets are good candidates. To date, no beta cellspecific<br />

CTLs have been characterised in T1D. In this<br />

study, we identified novel epitopes originating from<br />

preproinsulin (PPI) that were naturally processed and<br />

presented by HLA-A2.1. Using these epitopes deployed in<br />

sensitive IFN-γ ELISPOTs a significantly increased prevalence<br />

<strong>of</strong> PPI-reactive CD8 T cells in the blood <strong>of</strong> HLA-A2.1<br />

+ T1D patients was detected, compared with A2.1+<br />

healthy controls in which these cells were extremely<br />

rare. CD8 T cell clones were generated from T1D patients<br />

using PPI epitope-loaded HLA-A2.1 tetramers, but could<br />

not be raised in controls subjects. Such clones were<br />

highly cytotoxic against HLA-A2.1+ PPI-transfected target<br />

cells and express NKG2D and CXCR3, which may be<br />

important both in beta cell targeting and migration to<br />

inflamed islets. The identification <strong>of</strong> beta cell restricted<br />

CD8 T cell epitopes and isolation <strong>of</strong> relevant CTLs in T1D<br />

patients suggests that this pathway may be important in<br />

targeted beta cell destruction.<br />

doi:10.1016/j.clim.2007.03.199<br />

OR.19 Anti-Thymocyte Globulin Prevents<br />

Autoimmune Encephalomyelitis by Expanding<br />

Myelin Antigen Specific Foxp3+ Regulatory T Cells<br />

Denise Chung, Postdoctoraltoral Fellow, Brigham and<br />

Women’s Hospital, Boston, MA, Thomas Korn, Research<br />

Fellow, Brigham and Women’s Hospital, Boston, MA, Julie<br />

Richard, Scientist, Genzyme Corporation, Framingham, MA,<br />

Adam Kohm, Research Fellow, Feinberg School <strong>of</strong> Medicine,<br />

Northwestern University, Chicago, IL, Melanie Ruzek,<br />

Scientist, Genzyme Corporation, Framingham, MA,<br />

Mohamed Oukka, Instructor, Brigham and Women’s<br />

hospital, Boston, MA, Stephen Miller, Pr<strong>of</strong>essor, Feinberg<br />

School <strong>of</strong> Medicine, Northwestern University, Chicago, IL,<br />

Sharon Nahill, Director <strong>of</strong> Research, Genzyme Corporation,<br />

Framingham, MA, Vijay Kuchroo, Pr<strong>of</strong>essor <strong>of</strong> Neurology,<br />

Brigham and Women’s Hospital, Boston, MA<br />

The T cell-depleting polyclonal antibody, anti-thymocyte<br />

globulin (ATG) has been used in organ transplantation<br />

to treat acute rejection episodes. However, its mechanism<br />

has not been fully understood. It is assumed that ATG<br />

treatment deletes all T cells equally resulting in generalized<br />

inhibition <strong>of</strong> T cell response. We hypothesized that<br />

ATG might selectively delete effector T cells and spare<br />

regulatory T cells and thereby provide an attractive<br />

option for the treatment <strong>of</strong> autoimmune diseases. In<br />

order to test this, we used Foxp3gfp “knock-in” mice in<br />

combination with a myelin oligodendrocyte glycoprotein<br />

(MOG)35–55/IAb tetramer to study more closely the<br />

effect <strong>of</strong> ATG treatment on antigen-specific T cell<br />

responses in vivo during MOG-induced experimental<br />

autoimmune encephalomyelitis (EAE). Administration <strong>of</strong><br />

ATG in vivo resulted in preferential depletion <strong>of</strong> T-eff<br />

with a marked expansion <strong>of</strong> MOG-specific tetramerreactive<br />

T-reg (CD4+Foxp3+) upon MOG immunization. In<br />

both acute and relapsing remitting disease models, ATG<br />

treatment resulted in the protection from EAE, both in a<br />

preventive and therapeutic setting. Analysis <strong>of</strong> purified Teff<br />

from control and ATG treated animals revealed that<br />

on a per cell basis, the effector function <strong>of</strong> residual T-eff<br />

was not compromised by ATG. Thus, ATG tips the balance<br />

<strong>of</strong> T-eff and T-reg and skews an autoantigen specific<br />

immune reaction from a pathogenic T cell response to a<br />

protective T-reg response that maintains immunological<br />

tolerance and prevents autoimmune inflammation. We<br />

conclude that ATG treatment enforces the development <strong>of</strong>


Abstracts<br />

a dominant immunoregulatory environment which may be<br />

advantageous for the treatment <strong>of</strong> T cell driven autoimmune<br />

diseases.<br />

doi:10.1016/j.clim.2007.03.200<br />

OR.20 Regulation Through Mimicry: The Autologous<br />

Milk Protein Butyrophilin Determines Susceptibility<br />

to Myelin Oligodendrocyte Glycoprotein-induced<br />

Experimental Autoimmune Encephalomyelitis<br />

Kerstin Berer, PhD Student, University <strong>of</strong> Aberdeen,<br />

Department <strong>of</strong> Medicine and Therapeutics, Aberdeen, Maria<br />

Storch, Pr<strong>of</strong>essor, Universitätsklinikum Graz,<br />

Universitätsklinik für Neurologie, Graz, Ian Mather,<br />

Pr<strong>of</strong>essor, University <strong>of</strong> Maryland, Department <strong>of</strong> Animal<br />

and Avian Sciences, Washington, MD, Christopher Linington,<br />

Pr<strong>of</strong>essor, University <strong>of</strong> Aberdeen, Department <strong>of</strong> Medicine<br />

and Therapeutics, Aberdeen<br />

Sensitization to the autologous milk protein butyrophilin<br />

(BTN) expands a pre-existing, regulatory T cell<br />

response which cross-reacts with myelin oligodendrocyte<br />

glycoprotein (MOG), an important candidate autoantigen<br />

in multiple sclerosis (MS). This regulatory T cell population<br />

is associated with the production <strong>of</strong> high levels <strong>of</strong> IL-10<br />

and is able to suppress proliferation <strong>of</strong> MOG-specific T<br />

cells in vitro, and to abrogate disease severity in C57BL/6<br />

mice with MOG-induced experimental autoimmune encephalomyelitis<br />

(EAE). These findings led us to speculate<br />

that post-natal induction <strong>of</strong> regulatory T cell responses by<br />

BTN in the mother’s milk is a factor that may determine<br />

susceptibility to MOG-induced EAE in adulthood. We<br />

investigated this hypothesis using BTN-deficient (BTN−/−)<br />

C57BL/6 mice and wild-type (WT) littermates. Earlier<br />

onset <strong>of</strong> disease and higher disease severity clearly<br />

indicate an increased susceptibility to MOG-induced EAE<br />

in BTN−/− mice. This was associated with higher inflammatory<br />

and demyelinating indices in BTN−/− mice<br />

compared to their WT littermates. Moreover, functional<br />

analysis <strong>of</strong> MOG-specific T cells in BTN−/− mice revealed a<br />

marked increase in the production <strong>of</strong> encephalitogenic<br />

cytokines (IL-17, IFN-γ and IL-6), while the synthesis <strong>of</strong> IL-<br />

10 was consistently decreased. Collectively, our data<br />

suggest that regulatory T cell responses induced by<br />

autologous BTN play an important role in determining<br />

susceptibility to MOG-induced EAE.<br />

doi:10.1016/j.clim.2007.03.201<br />

OR.21 NOD Islet Autoimmunity Dependence on<br />

Single Amino Acid Difference in Insulin B:9−23<br />

Peptide<br />

Maki Nakayama, Fellow, Barbara Davis Center for Childhood<br />

Diabetes, University <strong>of</strong> Colorado Health Sciences Center,<br />

Aurora, CO, Jean Jasinski, Graduate Student, Barbara Davis<br />

Center for Childhood Diabetes, University <strong>of</strong> Colorado<br />

Health Sciences Center, Aurora, CO, Dongmei Miao, PRA,<br />

Barbara Davis Center for Childhood Diabetes, University <strong>of</strong><br />

Colorado Health Sciences Center, Aurora, CO, Kelly<br />

Johnson, PRA, Barbara Davis Center for Childhood Diabetes,<br />

University <strong>of</strong> Colorado Health Sciences Center, Aurora, CO,<br />

Edwin Liu, Assistant Pr<strong>of</strong>essor, Barbara Davis Center for<br />

Childhood Diabetes, University <strong>of</strong> Colorado Health Sciences<br />

Center, Aurora, CO, John Elliott, Pr<strong>of</strong>essor, Department <strong>of</strong><br />

Medical Microbiology and <strong>Immunology</strong>, University <strong>of</strong><br />

Alberta, Edmonton, AB, Canada, Canada, George<br />

Eisenbarth, Executive Director, Barbara Davis Center for<br />

Childhood Diabetes, University <strong>of</strong> Colorado Health Sciences<br />

Center, Aurora, CO<br />

NOD mice lacking native insulin genes but bearing<br />

mutated preproinsulin transgene (alanine [A] rather than<br />

tyrosine [Y] at B chain position 16) do not develop diabetes<br />

(Nakayama et al., Nature 2005). In the current study, we<br />

explore whether the initiation <strong>of</strong> anti-islet autoimmunity is<br />

dependent on this one amino acid difference and demonstrate<br />

that peripheral exposure to native B:9–23 sequence<br />

restores anti-islet autoimmunity. We created multiple<br />

double insulin knockout NOD mice bearing a normal<br />

preproinsulin 2 transgene (B16:Y-dKO mice), instead <strong>of</strong> the<br />

mutated B16:A transgene (B16:A-dKO mice). Eleven out <strong>of</strong><br />

twenty five B16:Y-dKO mice developed insulin autoantibodies,<br />

whereas less than 5% <strong>of</strong> B16:A-dKO mice (1/31) did<br />

(Pb0.01). B16:Y-dKO mice express insulin message in thymus<br />

as high as B16:A-dKO and wild-type NOD mice. These results<br />

indicate that the native B16:Y sequence in a transgene<br />

determines development <strong>of</strong> anti-insulin autoimmunity not<br />

because <strong>of</strong> less thymic insulin expression. We next investigated<br />

where the normal insulin sequence acts to induce<br />

autoimmunity. Transplantation <strong>of</strong> NOD islets, but not bone<br />

marrow, with native insulin sequences [B16:Y] into B16:AdKO<br />

mice rapidly restored development <strong>of</strong> insulin autoantibodies<br />

and insulitis. Splenocytes from B16:A-dKO mice<br />

transplanted with B16:Y islets induced diabetes when<br />

transferred into either wild-type or B16:A dKO NOD.SCID<br />

mice. Splenocytes from mice immunized with native insulin<br />

B:9–23 peptide induced diabetes on transfer if the recipients<br />

expressed the native B:9–23 [B16:Y] in their pancreas. These<br />

studies document dependence upon insulin B16:Tyrosine <strong>of</strong><br />

the insulin B:9–23 peptide for both the initial priming and<br />

effector phase <strong>of</strong> NOD anti-islet autoimmunity.<br />

doi:10.1016/j.clim.2007.03.202<br />

S11<br />

OR.22 Use <strong>of</strong> Random Peptide Display and Novel<br />

Bioinformatics Algorithm to Detect Conformational<br />

Epitopes <strong>of</strong> Jun a 1, The Major Allergen <strong>of</strong> Mountain<br />

Cedar Pollen<br />

Terumi Midoro-Horiuti, Assistant Pr<strong>of</strong>essor, Pediatrics,<br />

Galveston, TX, Ruby Tiwari, Postdoctoraltoral Fellow,<br />

Pediatrics, Galveston, TX, Surendra Negi, Postdoctoral<br />

Fellow, Biochemistry and Molecular Biology, Galveston, TX,<br />

Catherine Schein, Assistant Pr<strong>of</strong>essor, Biochemistry and<br />

Molecular Biology, Galveston, TX, Bo Ning, Research<br />

Assistant, Pediatrics, Galveston, TX, Randall Goldblum,<br />

Pr<strong>of</strong>essor, Pediatrics, Galveston, TX, Werner Braun,<br />

Pr<strong>of</strong>essor, Biochemistry and Molecular Biology, Galveston, TX


S12 Abstracts<br />

Rationale: Pollen hypersensitivity is a major cause <strong>of</strong><br />

seasonal airway disease world wide. We recently resolved<br />

the crystal structure and mapped the linear IgE epitopes <strong>of</strong><br />

Jun a 1. Heat and chemical denaturation <strong>of</strong> Jun a 1<br />

reduced the binding <strong>of</strong> human IgE, suggesting the importance<br />

<strong>of</strong> conformational epitopes for reactivity. Methods:<br />

To identify conformational IgE epitopes, we developed a<br />

panel <strong>of</strong> monoclonal antibodies (mAbs) against Jun a 1 and<br />

selected those that were heat sensitive and inhibited the<br />

binding <strong>of</strong> IgE from patient sera. These mAbs were used to<br />

screen a random peptide, phage display library. After<br />

repeated panning, phage clones were isolated and their<br />

peptide sequences determined. To compare the affinity<br />

and specificity <strong>of</strong> phage-expressed peptides the binding <strong>of</strong><br />

the phage to mAbs and patient IgE, and the ability <strong>of</strong> the<br />

phage to inhibit binding <strong>of</strong> Jun a 1 to the mAb were tested<br />

by ELISA. Consensus amino acids from selected phage were<br />

matched with clusters <strong>of</strong> amino acid residues exposed on<br />

the surface <strong>of</strong> Jun a 1, using GETAREA (http://www.scsb.<br />

utmb.edu/cgi-bin/get_a_form.tcl) and a program specifically<br />

designed to analyze similarity between the specific<br />

phage residues and surface patches on allergens. Results:<br />

Consensus amino acids from phage clones revealed common<br />

patterns <strong>of</strong> amino acids which were identical or<br />

similar to compact patches on the 3-D structure <strong>of</strong> Jun a 1<br />

and represent putative conformational epitopes. Conclusion:<br />

Bioinformatics search tools can play an important role<br />

in identifying the IgE epitopes on allergens with known<br />

structures or reliable 3D models.<br />

doi:10.1016/j.clim.2007.03.203<br />

OR.23 Antibodies Against Human Cytomegalovirus<br />

in the Pathogenesis <strong>of</strong> Atherosclerosis: A Gene Array<br />

Approach<br />

Antonio Puccetti, Pr<strong>of</strong>essor <strong>of</strong> Medicine, Gaslini<br />

Institute-Department <strong>of</strong> <strong>Immunology</strong>, Genova, Marzia Dolci,<br />

England Research Fellow, Gaslini Institute-Department <strong>of</strong><br />

<strong>Immunology</strong>, Genova, Dimitri Peterlana, PhD Student,<br />

University <strong>of</strong> Verona-Department <strong>of</strong> Internal Medicine,<br />

Verona, Riccardo Navone, Research Fellow, University <strong>of</strong><br />

Verona-Department <strong>of</strong> Internal Medicine, Verona, Caterina<br />

Bason, Research Fellow, University <strong>of</strong> Verona-Department <strong>of</strong><br />

Internal Medicine, Verona, Claudio Lunardi, Pr<strong>of</strong>essor <strong>of</strong><br />

Medicine, University <strong>of</strong> Verona-Department <strong>of</strong> Internal<br />

Medicine, Verona<br />

Human cytomegalovirus (hCMV) is involved in the<br />

pathogenesis <strong>of</strong> atherosclerosis. We have shown in patients<br />

with atherosclerosis that antibodies directed against the<br />

hCMV-derived proteins US28 and UL122 are able to induce<br />

endothelial cell damage and apoptosis <strong>of</strong> non-stressed<br />

endothelial cells through cross-rection with normally<br />

expressed surface molecules. Our aim was to dissect the<br />

molecular basis <strong>of</strong> such interaction and to investigate<br />

mechanisms linking innate immunity to atherosclerosis. We<br />

analysed the gene expression pr<strong>of</strong>iles in endothelial cells<br />

stimulated with antibodies affinity purified against either<br />

the UL122 or the US28 peptides. Real-time polymerase<br />

chain reaction was used to validate the microarray results.<br />

Supernatant <strong>of</strong> endothelial cells incubated with antibodies<br />

was analysed for the presence <strong>of</strong> Heat Shock Protein (HSP)<br />

60 and was used to assess stimulation <strong>of</strong> toll-like receptor-<br />

4 (TLR4). Antibodies against UL122 and US28 induced the<br />

expression <strong>of</strong> genes encoding for adhesion molecules,<br />

chemokines, growth factors and molecules involved in the<br />

apoptotic process together with other genes involved in<br />

the initiation and progression <strong>of</strong> atherosclerosis. HSP60 was<br />

released in the medium <strong>of</strong> cells incubated with anti-US28<br />

antibodies and was able to engage TLR4. Antibodies<br />

directed against hCMV modulate the expression <strong>of</strong> genes<br />

coding for molecules involved in the pathogenesis <strong>of</strong><br />

atherosclerosis. Moreover, endothelial cells exposed to<br />

such antibodies express HSP60 on the cell surface and<br />

release HSP60 in the medium able to activate TLR4. These<br />

data confirm that hCMV plays a crucial role in mediating<br />

the atherosclerotic process and that HSP60 is an endogenous<br />

ligand for TLR4 signaling.<br />

doi:10.1016/j.clim.2007.03.204<br />

OR.24 Differential Effects <strong>of</strong> Aire Gene Deficiency<br />

on the Development <strong>of</strong> Autoimmune<br />

Encephalomyelitis Induced by Two Representative<br />

Myelin Peptides<br />

Asako Tagawa, Research Fellow, National Institute <strong>of</strong><br />

Neuroscience, NCNP, Kodaira, Tokyo, Japan, Toshimasa<br />

Aranami, Section chief, Department <strong>of</strong> <strong>Immunology</strong>,<br />

National Institute <strong>of</strong> Neuroscience, NCNP, Kodaira, Tokyo,<br />

Japan, Mitsuru Matsumoto, Director, Division <strong>of</strong> Molecular<br />

<strong>Immunology</strong>, Institute for Enzyme Research, University <strong>of</strong><br />

Tokushima, Japan, Tokushima, Japan, Takashi Yamamura,<br />

Director, Department <strong>of</strong> <strong>Immunology</strong>, National Institute <strong>of</strong><br />

Neuroscience, NCNP, Kodaira, Tokyo, Japan<br />

Hazardous autoimmunity is prevented partly by clonal<br />

deletion <strong>of</strong> autoimmune T cells in the thymus. In the process<br />

referred to as central tolerance, autoimmune regulator<br />

(Aire) gene plays a pivotal role through transcriptional<br />

control <strong>of</strong> ectopic antigen expression. In fact, thymic<br />

expression <strong>of</strong> myelin antigens was shown to be involved in<br />

shaping Tcell repertoire that would mediate development <strong>of</strong><br />

EAE. Here we asked if Aire gene deficiency might alter<br />

manifestations <strong>of</strong> EAE induced with representative, encephalitogenic<br />

peptides myelin oligodendrocyte glycoprotein<br />

(MOG)35–55 and proteolipid protein (PLP)178–191 by using<br />

Aire−/− mice. It is thought that MOG is not expressed in the<br />

thymus (JCI 12: 544, 2003), whereas thymic expression <strong>of</strong><br />

PLP178–191 was previously proven (Nat Med 6:56, 2000).<br />

First, we assessed recall responses to the peptides in the<br />

immunized mice, and found that sensitized T cells from the<br />

Aire−/− produced a higher amount <strong>of</strong> IL-17 than from wildtype<br />

(WT) mice regardless <strong>of</strong> the peptides used for<br />

immunization. Anti-MOG35–55 response was also associated<br />

with a higher IFN-ã production in Aire−/−. However, there<br />

was no significant difference between Aire−/− and WT in<br />

IFN-ã production in response to PLP178–191, which resulted<br />

in an augmented Th17/Th1 ratio in PLP178–191-immunized<br />

mice. PLP178–191 immunization induced more serious EAE in<br />

Aire−/− than WT, whereas such a difference could not be


Abstracts<br />

seen after sensitization with MOG35–55. As such, Aire gene<br />

deficiency would differentially alter autoimmune responses<br />

according to the presence or absence <strong>of</strong> the directed epitope<br />

in the thymus.<br />

doi:10.1016/j.clim.2007.03.205<br />

Stem Cell/Immunodeficiency/HIV<br />

Friday, June 8<br />

2:45 pm−4:45 pm<br />

OR.25 A Novel RAG-1 Null Mutant Causes T-B-NK+<br />

SCID in Native Americans<br />

Morton Cowan, Pr<strong>of</strong>essor, Pediatric Bone Marrow Transplant<br />

Division, University <strong>of</strong> California, San Francisco Children’s<br />

Hospital, San Francisco, CA<br />

Severe combined immunodeficiency disease (SCID) is the<br />

most serious inherited immunological deficit. Ubiquitous<br />

DNA double strand repair factors in the non-homologous<br />

ending joining (NHEJ) pathway resolve DNA double-strand<br />

breaks (DSB) during V(D)J recombination <strong>of</strong> T and B<br />

lymphocyte receptor genes. We have described a null<br />

mutation <strong>of</strong> the Artemis gene that has relatively high<br />

occurrence among Athabascan-speaking Native Americans,<br />

which results in a complete absence <strong>of</strong> T and B lymphocytes<br />

and increased cellular sensitivity to ionizing radiation<br />

causing radiosensitive-SCID (RS-SCID). Recently we identified<br />

a novel RAG-1 null mutation, R776W, in three related<br />

children from the Dine Indian Tribe in the Canadian<br />

Northwest Territories who manifest a classic T-B-NK+ SCID<br />

phenotype. We found a normal pattern <strong>of</strong> radiation<br />

sensitivity in skin fibroblast cell lines from these patients,<br />

indicating that the NHEJ pathway is intact. The impaired<br />

activity <strong>of</strong> this RAG-1 mutant in V(D)J recombination was<br />

revealed by an enhanced green fluorescent protein (EGFP)<br />

based assay. Additional studies <strong>of</strong> the RAG-1 null mutant<br />

will evaluate DNA binding and cleavage activity, hairpin<br />

formation in vitro as well as catalysis <strong>of</strong> other DNA strand<br />

transfer reactions such as transposition. Our current results<br />

suggest that the T-B-NK+ SCID phenotype is triggered by this<br />

novel RAG-1 mutant.<br />

doi:10.1016/j.clim.2007.03.206<br />

OR.26 HIV-1 Infectivity Inhibited by Binding <strong>of</strong> the<br />

Green Tea Catechin, Epigallocatechin Gallate, to<br />

CD4<br />

Christina Nance, Instructor, Baylor College <strong>of</strong> Medicine,<br />

Houston, TX, Edward Siwak, Assistant Pr<strong>of</strong>essor, Baylor<br />

College <strong>of</strong> Medicine, Virology, Houston, TX, Aarthi Ram,<br />

Research Technician, Baylor College <strong>of</strong> Medicine, Pediatrics,<br />

Houston, TX, Van Willis, Research Technician, Baylor College<br />

<strong>of</strong> Medicine, Pediatrics, Houston, TX, Susan Westerfield,<br />

Research Technician, Baylor College <strong>of</strong> Medicine, Pediatrics,<br />

Houston, TX, William Shearer, Pr<strong>of</strong>essor, Baylor College <strong>of</strong><br />

Medicine, Pediatrics, Houston, TX<br />

Binding <strong>of</strong> HIV-1 envelope glycoprotein, gp120, to the CD4<br />

T cell receptor results in HIV-1 infection. Previously, we have<br />

presented evidence <strong>of</strong> high affinity binding <strong>of</strong> the green tea<br />

catechin, epigallocatechin gallate (EGCG), to the CD4<br />

molecule at the gp120 binding pocket. We now present<br />

evidence that EGCG inhibits the binding <strong>of</strong> gp120 on human<br />

CD4+ T cells and PBMC and prevents the HIV-1 infectivity <strong>of</strong><br />

human macrophages and PBMC. Binding studies utilized flow<br />

cytometry. HIV-1 infectivity was assessed by HIV-1 p24 EIA. Mtropic<br />

(R5) (strains SF162, Bal), T-tropic (X4) (strain IIIB), and<br />

dual tropic (R5X4) (strain 89.6) HIV-1 isolates were used.<br />

EGCG markedly inhibited the binding <strong>of</strong> HIV-1-gp120IIIB to<br />

CD4+ T cells in a dose-dependent manner at physiologically<br />

relevant levels (32% at 0.2 μM pb0.05, 42% at 2 μM and 47%<br />

at 20 μM pb0.01) and higher (55% at 50 μM and 71% at 100 μM<br />

pb0.001). EGCG significantly inhibited the infectivity <strong>of</strong><br />

human macrophages by M- and dual-tropic HIV-1 strains at<br />

200–400 TCID50 in a dose-dependent manner. There was<br />

100% inhibition <strong>of</strong> p24 production by EGCG (25–100 μM)<br />

(pb0.0001), 95% at 12 μM, and 79% at 6 μM (pb0.001). The<br />

response by human PBMC was similar. The control catechin<br />

did not alter gp120 binding nor inhibit HIV-1 infectivity. We<br />

conclude that EGCG is able to significantly reduce the<br />

attachment <strong>of</strong> gp120 to CD4, along with a decrease in the<br />

infectivity <strong>of</strong> HIV-1 to target cells. The competitive binding<br />

properties <strong>of</strong> EGCG for the CD4 binding sites by gp120 may<br />

translate to an HIV-1 preventative strategy.<br />

doi:10.1016/j.clim.2007.03.207<br />

S13<br />

OR.27 Impaired Dendritic Cell Function in<br />

Ectodermal Dysplasia with Immune Deficiency is<br />

Linked to Defective NEMO Ubiquitination<br />

Stephan Temmerman, Postdoctoral Fellow, NIH-NIAID-LHD,<br />

Bethesda, MD, Chi Ma, Senior Biologist, NIH-NIAID-LHD,<br />

Bethesda, MD, Ashish Jain, PI, NIH-NIAID-LHD, Bethesda, MD<br />

NF-kappaB essential modulator (NEMO) regulates the<br />

activation <strong>of</strong> the transcription factors NF-kappaB. Alterations<br />

in NEMO cause ectodermal dysplasia with immunodeficiency<br />

(EDI), a disorder that is characterized by<br />

defects in innate and adaptive immunity as well as<br />

abnormal development <strong>of</strong> ectoderm-derived tissues.<br />

Because the biochemical mechanism by which NEMO<br />

mutations cause immune dysfunction remains undefined,<br />

we investigated the effect <strong>of</strong> a cysteine to arginine<br />

substitution found in the NEMO zinc finger domain on<br />

dendritic cell (DC) function. Following CD40 ligand<br />

(CD40L) stimulation <strong>of</strong> DCs from two EDI patients, we<br />

found that lysine 63-linked polyubiquitination <strong>of</strong> NEMO,<br />

which promotes activation <strong>of</strong> the IKK complex, was<br />

absent. We associated this defect with preserved RelA<br />

but absent c-Rel activity. Therefore, CD40L-stimulated EDI<br />

DCs failed to synthesize the c-Rel dependant cytokine IL-<br />

12, displayed reduced cell aggregates and membrane<br />

extensions, and failed to support allogeneic lymphocyte<br />

proliferation. We utilized gene expression pr<strong>of</strong>iling to<br />

compare the genes induced by CD40L in normal DCs but<br />

not in EDI DCs. In this gene set, we identified a number


S14 Abstracts<br />

<strong>of</strong> molecules that are likely to be critical to the<br />

maturation and function <strong>of</strong> DCs. In contrast, downstream<br />

NF-kappaB activity, DC maturation, and NEMO polyubiquitination<br />

were normal in EDI DCs following stimulation with<br />

the TLR4 ligand. These results show for the first time that<br />

CD40 and TLR4 use different signaling pathways to<br />

ubiquitinate NEMO, and <strong>of</strong>fer insight into how a mutation<br />

in the zinc finger domain <strong>of</strong> NEMO leads to pathway<br />

specific defects in NF-kappaB signaling and thus immune<br />

deficiency.<br />

doi:10.1016/j.clim.2007.03.208<br />

OR.28 Treatment <strong>of</strong> HIV-infected Patients with<br />

Vitamin D-binding Protein Derived Macrophage<br />

Activating Factor (GcMAF) Eradicates HIV-Infection<br />

Nobuto Yamamoto, Director, Socrates Institute for<br />

Therapeutic <strong>Immunology</strong>, Philadelphia, PA, Masumi Ueda,<br />

<strong>Immunology</strong>, Chief, Microbiology, Philadelphia, PA,<br />

Charles Benson, Head, Infectious Diseases, School <strong>of</strong><br />

Veterinary Medicine, University <strong>of</strong> Pennsylvania, Kennett<br />

Square, PA<br />

Serum vitamin D-binding protein (known as Gc protein) is<br />

the precursor for the principal macrophage activating factor<br />

(MAF). The MAF precursor activity <strong>of</strong> serum Gc protein <strong>of</strong><br />

HIV-infected patients was lost or reduced because Gc<br />

protein is deglycosylated by serum α-N-acetylgalactosaminidase<br />

(Nagalase) secreted from HIV-infected cells. Since<br />

Nagalase is the intrinsic component <strong>of</strong> gp120, serum<br />

Nagalase activity is the sum <strong>of</strong> enzyme activities expressed<br />

in both HIV virions and envelope proteins released from HIVinfected<br />

cells. Because <strong>of</strong> Nagalase being an HIV viral<br />

protein and immunogenic, serum Nagalase was already<br />

complexed with anti-HIV immunoglobulin G (IgG) in patient<br />

blood stream. These antibodies, however, were largely not<br />

neutralizing antibodies. The IgG-bound viral proteins<br />

retained Nagalase activity that deglycosylates Gc protein.<br />

Therefore, macrophages <strong>of</strong> HIV-infected patients having<br />

deglycosylated Gc protein cannot be activated, leading to<br />

immunosuppression. Stepwise treatment <strong>of</strong> purified Gc<br />

protein with immobilized beta-galactosidase and sialidase<br />

generated the most potent macrophage activating factor<br />

(termed GcMAF), which produces no side effect in humans.<br />

Macrophages activated by administration <strong>of</strong> GcMAF<br />

(100 ng/patient) develop a large amount <strong>of</strong> Fc receptors<br />

as well as enormous variation <strong>of</strong> receptors that recognize<br />

IgG bound and unbound HIV virions. Thus, macrophages<br />

activated by GcMAF preferentially phagocytize IgG-bound<br />

HIV virions via Fc receptor mediation. Cells latently<br />

infected with HIV are unstable and spontaneously release<br />

the virions at a high rate. After less than 18 weekly<br />

administrations <strong>of</strong> 100 ng GcMAF for twenty-one nonanemic<br />

patients, they exhibited low serum Nagalase activities<br />

equivalent to healthy controls, indicating eradication <strong>of</strong> HIV<br />

infection.<br />

doi:10.1016/j.clim.2007.03.209<br />

OR.29 Impaired In Vitro Regulatory T Cell Function<br />

Associated with Wiskott-Aldrich Syndrome<br />

Marsilio Adriani, Visiting Fellow, NIH/NHGRI, Bethesda, MD,<br />

Joseph Aoki, Visiting Fellow, NIH/NHGRI, Bethesda, MD,<br />

Reiko Horai, Postdoctoral Fellow, NIH/NHGRI, Bethesda,<br />

MD, Akihiro Kon, England Postdoctoral, Fellow, Bethesda,<br />

MD, Angela M. Thornton, Staff Scientist, NIH/NIAID,<br />

Bethesda, MD, Martha Kirby, Senior Research Assistant,<br />

NIH/NHGRI, Bethesda, MD, Stacie M. Anderson, Senior<br />

Research Assistant, NIH/NHGRI, Bethesda, MD, Richard M.<br />

Siegel, Investigator, NIH/NIAMS, Bethesda, MD, Pamela L.<br />

Schwartzberg, Investigator, NIH/NHGRI, Bethesda, MD,<br />

Fabio Candotti, Senior Investigator, NIH/NHGRI, Bethesda,<br />

MD<br />

Wiskott-Aldrich syndrome (WAS) is a primary immunodeficiency<br />

characterized by thrombocytopenia, eczema, recurrent<br />

infections and high incidence <strong>of</strong> malignancy. This disease is<br />

caused by mutations <strong>of</strong> the WAS protein (WASP) gene, a key<br />

regulator <strong>of</strong> actin polymerization. WAS patients show high<br />

incidence <strong>of</strong> autoimmune disorders (∼40–70%) the occurrence<br />

<strong>of</strong> which, however, does not correlate with the severity <strong>of</strong> the<br />

other classical hallmarks <strong>of</strong> the disease. A central question in<br />

understanding the pathophysiology <strong>of</strong> WAS is why immunodeficient<br />

patients develop symptoms suggestive <strong>of</strong> hyperactivation<br />

<strong>of</strong> immune compartments, such as eczema and autoimmune<br />

diseases. Since defects in CD4+CD25+ regulatory T cells (Treg)<br />

have been associated with autoimmunity, we examined the<br />

presence and function <strong>of</strong> these cells in WAS patients and Waspdeficient<br />

mice. Wasp-deficient patients and mice develop Treg<br />

cells that express Foxp3 with no detectable differences in<br />

frequency compared to controls. However, in vitro suppression<br />

assays showed that these Treg cells have impaired inhibitory<br />

function in vitro,which,inmice,couldonlybepartiallyrescued<br />

by pre-activation with exogenous IL-2. The demonstration <strong>of</strong><br />

impaired in vitro suppressor function in WASp-deficient Treg<br />

cells suggests that defective regulatory Tcell function may be an<br />

important factor contributing to immune dysregulation in WAS.<br />

doi:10.1016/j.clim.2007.03.210<br />

OR.30 Regulatory T Cell Abnormalities Associated<br />

with Aberrant CD4+ T Cell Responses in Patients<br />

with Immune Reconstitution Disease (IRD)<br />

Nabila Seddiki, Research Fellow, Centre For <strong>Immunology</strong><br />

and National Centre in HIV Epidemiology and <strong>Clinical</strong><br />

Research (UNSW), Darlinghurst<br />

Up to 30% <strong>of</strong> patients with HIV commencing antiretroviral<br />

therapy (ART) late in the disease restore a pathogen-specific<br />

cellular immune response that is immunopathological and<br />

causes disease referred as immune reconstitution disease or<br />

IRD. The immunopathogenesis <strong>of</strong> IRD is not well understood and<br />

is likely to be a consequence <strong>of</strong> an aberrant reconstitution <strong>of</strong><br />

certain T cell subsets. In a cross-sectional study, using HIV<br />

patients who developed mycobacterial IRD, we show that in<br />

comparison to those starting ARTand not developing IRD, CD4+ T<br />

cells specific for MTB and M. avium complex (MAC) antigens,<br />

produce high levels <strong>of</strong> IFN-g and IL-2 (Pb0.01) and proliferate<br />

strongly when compared to controls. We postulated that deficits<br />

in regulatory T cells (Tregs) numbers may play a central role in


Abstracts<br />

the pathogenesis <strong>of</strong> IRD. When Tregs numbers were examined by<br />

using our recently described marker CD127 (IL-7R), we found a<br />

significant expansion <strong>of</strong> memory CD127loFoxp3+CD25+ Tregs in<br />

IRD patients compared to controls. To further investigate<br />

abnormalities in T cell responses, we examined a range <strong>of</strong><br />

cytokines in the plasma <strong>of</strong> patients and controls and found<br />

increased levels <strong>of</strong> IL-4, IL-6, IL-7 and IL-21. Interestingly, we<br />

show that some <strong>of</strong> these cytokines inhibit strongly Treg<br />

suppression. Furthermore, we show that suppressors and<br />

responders from these patients present defect to regulate CD4<br />

T cell immune responses. Altogether, these data suggest that<br />

despite the high Treg expansion in IRD, their ability to induce<br />

suppression and turn <strong>of</strong>f these aberrant immune responses is<br />

compromised.<br />

doi:10.1016/j.clim.2007.03.211<br />

OR.31 Liver Tissue Cells Inhibit Immune Responses<br />

by Inducing T Cell Apoptosis: An Application in Islet<br />

Transplantation<br />

Lina Lu, Associate Pr<strong>of</strong>essor, Cleveland Clinic, Cleveland,<br />

OH, Cheng-Hsu Chen, Research Fellow, Department <strong>of</strong><br />

<strong>Immunology</strong>, Cleveland Clinic, Cleveland, OH, Zhenyu Yin,<br />

Research Fellow, Department <strong>of</strong> <strong>Immunology</strong>, Cleveland<br />

Clinic, Cleveland, OH, John J. Fung, Pr<strong>of</strong>essor, Department<br />

<strong>of</strong> General Surgery, Cleveland Clinic, Cleveland, OH,<br />

Liang-Mou Kuo, Research Fellow, Department <strong>of</strong><br />

<strong>Immunology</strong>, Cleveland Clinic, Cleveland, OH, Shiguang<br />

Qian, Associate Pr<strong>of</strong>essor, Department <strong>of</strong> <strong>Immunology</strong>,<br />

General Surgery, Cleveland Clinic, Cleveland, OH<br />

Organ transplantation has been successful for decades, but<br />

outcome <strong>of</strong> cell transplants remains poor. This is true in animal<br />

models. Liver transplants are spontaneously accepted in mice,<br />

but hepatocyte allografts are acutely rejected, suggesting an<br />

immunosuppressive effect <strong>of</strong> liver tissue cells. In this study, we<br />

demonstrated a pr<strong>of</strong>ound in vitro immune inhibitory activity <strong>of</strong><br />

activated (but not quiescent) hepatic stellate cells (HSC) that<br />

are well known to be involved in liver fibrosis. Activation <strong>of</strong> HSC<br />

by exposure to IFN-γ or directly contact with activated T cells<br />

resulted in upregulation <strong>of</strong> B7-H1, inhibitory cytokines (IL-6, IL-<br />

10, TGF-β), and enhanced T cell apoptotic death (annexin<br />

V + 7AAD− ), which is largely mediated by B7-H1, since B7-H1−/− HSC triggered significantly less apoptosis. To explore potential<br />

clinical application, BALB/c (H2d ) islets (300) mixed with<br />

activated HSC (3×10 5 )<strong>of</strong>B6(H2b )miceweretransplanted<br />

under renal capsule <strong>of</strong> chemically diabetic B6 recipients,<br />

showing a marked prolongation <strong>of</strong> islet graft survival [from<br />

median survival time (MST) <strong>of</strong> 13 days in control to N60 days<br />

(pb0.01)] with 55% <strong>of</strong> recipients being euglycemia for N60 days.<br />

None <strong>of</strong> islet-only recipients remained euglycemia for N17 days.<br />

Removal <strong>of</strong> islet-grafted kidney resulted in quick glycemia.<br />

Immunochemistry showed that prolongation <strong>of</strong> islet allografts by<br />

co-transplanted HSC was associated with upregulated apoptotic<br />

activity (TUNEL) and less T cell infiltration (CD3), which<br />

appeared to be mediated by B7-H1, as co-transplant with B7-<br />

H1 −/− HSC markedly reduced their protection to islet allografts<br />

(MST 26 days). These scientific observations may lead to<br />

substantial improvement <strong>of</strong> islet transplantation.<br />

doi:10.1016/j.clim.2007.03.212<br />

OR.32 BAFF Receptor Expression in CVID Patients<br />

Marta Rizzi, MD PhD, <strong>Clinical</strong> Research Unit for<br />

Rheumatology, Freiburg/Br., Ulrich Salzer, PhD, Division <strong>of</strong><br />

Rheumatology and <strong>Clinical</strong> <strong>Immunology</strong>, Freiburg/Br., Klaus<br />

Warnatz, MD PhD, Division <strong>of</strong> Rheumatology and <strong>Clinical</strong><br />

<strong>Immunology</strong>, Freiburg/Br., Sigune Goldacker, MD, Division <strong>of</strong><br />

Rheumatology and <strong>Clinical</strong> <strong>Immunology</strong>, Freiburg/Br.,<br />

Stefanie Hamm, Student, <strong>Clinical</strong> Research Unit for<br />

Rheumatology, Freiburg/Br., Hans Hartmut Peter, Pr<strong>of</strong>essor<br />

MD PhD, Division <strong>of</strong> Rheumatology and <strong>Clinical</strong> <strong>Immunology</strong>,<br />

Freiburg/Br., Hermann Eibel, PhD, <strong>Clinical</strong> Research Unit<br />

for Rheumatology, Freiburg/Br.<br />

BAFF receptor (BAFF-R) is a member <strong>of</strong> the TNF-R superfamily<br />

and is expressed mainly on mature B cells. Common<br />

variable immune deficiency (CVID) is a primary immune<br />

deficiency characterized by hypogammaglobulinemia. Several<br />

genetic defects contributing to this phenotype have been<br />

identified (mutations in ICOS gene, TACI gene, CD19 gene). Our<br />

group recently identified a deletion mutation within the BAFF-<br />

R gene, removing the transmembrane domain <strong>of</strong> the protein.<br />

The homozygous mutation precludes the syntheses <strong>of</strong> functional<br />

BAFF-R. Therefore, B cell development in the BAFF-R<br />

deficient patient is arrested at the transitional B cell stage. As<br />

a result, the patient lacks IgG-producing plasma cells and is<br />

severely B lymphopenic. Searching for additional BAFF-R mutations<br />

we screened a subgroup <strong>of</strong> our cohort <strong>of</strong> CVID patients<br />

with very low number <strong>of</strong> B cells in peripheral blood (b5%) for<br />

BAFF-R expression. We found that 8 out <strong>of</strong> the 13 patients<br />

presented low BAFF-R expression on the B cells surface (mean<br />

fluorescence 21.9 SD 4.3, control mean fluorescence 58.9 SD<br />

3.6), and low BAFF binding activity. Low BAFF-R expression<br />

significantly correlates with a low number <strong>of</strong> detectable<br />

memory B cells (CD19+CD27+). Sequencing <strong>of</strong> the BAFF-R<br />

alleles excluded known mutations in the BAFF-R gene. We then<br />

investigated if low BAFF-R expression might result from<br />

regulatory defects. Stimulation <strong>of</strong> the BCR, TLR 9 and/or<br />

CD40 increased BAFF-R expression. Therefore, low levels <strong>of</strong><br />

BAFF-R expression in CVID patients may result from regulatory<br />

defects and contribute to antibody deficiency.<br />

doi:10.1016/j.clim.2007.03.213<br />

S15


S16 Abstracts<br />

F.1 Thyroid Function and Prevalence <strong>of</strong><br />

Anti-Thyroid Antibodies in Iranian Patients with<br />

Type 1 Diabetes Mellitus: Influences <strong>of</strong> Age and Sex<br />

Faranak Sharifi, Endocrinologist, Zanjan University <strong>of</strong><br />

Medical Sciences, Zanjan, Leila Ghasemi Hashtroodi,<br />

Resident for Internal Medicine, Zanjan University <strong>of</strong> Medical<br />

Sciences, Zanjan, Yahya Jaberi, Assistant Pr<strong>of</strong>essor, Zanjan<br />

University <strong>of</strong> Medical Sciences, Zanjan<br />

Objective: Type 1 diabetes mellitus is frequently associated<br />

with autoimmune thyroid disease (ATD). Genetic<br />

susceptibility to autoantibody formation in association with<br />

ATD and type 1 diabetes mellitus has been described with<br />

varying frequencies, but there is still debate about the<br />

situation in Iran. We have, therefore, investigated the<br />

prevalence <strong>of</strong> anti-thyroid peroxidase (anti-TPO) and antithyroglobulin<br />

(anti-TG) antibodies in type 1 diabetic patients,<br />

and compared the effect <strong>of</strong> age and sex on the thyroid<br />

autoimmunity in patients with type 1 diabetes mellitus in<br />

Iran. Subjects and Methods: Ninety one subjects with type 1<br />

diabetes mellitus and one hundred and sixty three unrelated<br />

normal controls under age thirty were recruited for the<br />

detection <strong>of</strong> anti-TPO and anti-TG. Radioimmunoassay was<br />

used for anti-TPO and anti-TG detection respectively.<br />

Results: Among 91 type 1 diabetic patients, 36 (39.6%)<br />

were positive for anti-TPO and 27 (30%) were positive for<br />

anti-TG. Anti-TPO antibodies were detected only in 6.7% <strong>of</strong><br />

control group. Compared with those without thyroid autoimmunity,<br />

there was a female preponderance for the type 1<br />

diabetic patients with thyroid autoimmunity (female: male,<br />

28:14 vs. 28:20 respectively). Among the type 1 diabetic<br />

patients those with thyroid autoimmunity tended to have<br />

older age (p: 0.04) and higher TSH concentration (p: 0.03).<br />

Patients with high anti-TPO levels had longer duration <strong>of</strong><br />

diabetes (p: 0.02). Conclusion: The presence <strong>of</strong> anti-TPO in<br />

39.6% <strong>of</strong> our type 1 diabetic patients compared with 8.5% <strong>of</strong><br />

normal subjects confirmed the strong association <strong>of</strong> ATD and<br />

type 1 diabetes mellitus.<br />

doi:10.1016/j.clim.2007.03.214<br />

Poster Sessions<br />

Friday, June 8<br />

7:30 am−7:00 pm<br />

Authors Present: 5:45 pm−7:00 pm<br />

F.2 Reduced Foxp3 and Interleukin 10 Expression<br />

During the Partial Remission Phase <strong>of</strong> Type 1<br />

Diabetes in Children<br />

Srinath Sanda, <strong>Clinical</strong> Fellow, University <strong>of</strong> California, San<br />

Diego School <strong>of</strong> Medicine, San Diego, CA, Bart Roep,<br />

Associate Pr<strong>of</strong>essor, Department <strong>of</strong> Immunohematology and<br />

Blood Transfusion Leiden University Medical Center, Leiden,<br />

Matthias von Herrath, Pr<strong>of</strong>essor, Developmental<br />

<strong>Immunology</strong> at the La Jolla Institute for Allergy and<br />

<strong>Immunology</strong>, La Jolla, CA<br />

Background: Some children undergo a reduction in insulin<br />

requirements after diagnosis and initiation <strong>of</strong> insulin<br />

therapy. The exact mechanism underlying this partial<br />

remission phase (also known as the honeymoon phase) <strong>of</strong><br />

type 1 diabetes in children is not known. Study Design: We<br />

conducted a cross-sectional study comparing peripheral<br />

blood T cell responses by ELISPOT and flow cytometry<br />

between a cohort <strong>of</strong> newly diagnosed children with type 1<br />

diabetes at initiation <strong>of</strong> insulin therapy and a cohort <strong>of</strong><br />

children in the honeymoon phase. Children were classified<br />

in the honeymoon phase if they were between 3 and<br />

9 months from diagnosis, had a total daily insulin requirement<br />

<strong>of</strong> b0.5 U/kg/day, and had experienced a N50%<br />

reduction in their total daily insulin dose since diagnosis. Six<br />

children were recruited in each group. Results: Honeymoon<br />

patients were on average 7.4 months from diagnosis and had<br />

significantly reduced insulin doses and hemoglobin A1c<br />

levels compared to new onset patients. Both cohorts had<br />

similar numbers <strong>of</strong> peripheral CD4+, CD8+, and CD4+CD25+<br />

cells. Honeymoon patients had fewer numbers <strong>of</strong> peripheral<br />

CD4+CD25+Foxp3+ cells compared to newly diagnosed<br />

patients (37.3% ±23% vs. 57.8±27.8% p=0.054). ELISPOT<br />

assays using epitopes to GAD65, IA2, and proinsulin,<br />

demonstrated a trend for increased interferon-gamma<br />

production and reduced interleukin-10 production in the<br />

honeymoon patients. Conclusion: Reduced numbers <strong>of</strong><br />

peripheral blood CD4+CD25+Foxp3+ and interleukin 10<br />

producing cells are seen in type 1 diabetes patients during<br />

their honeymoon phase compared to patients at diagnosis.<br />

doi:10.1016/j.clim.2007.03.215<br />

F.3 A Novel Cell-based Therapeutic Approach to<br />

Prevent and Cure Autoimmune Type I Diabetes in<br />

NOD Mice<br />

Dong Zhang, Postdoctoral Research Fellow, Harvard Medical<br />

School, Beth Israel Deaconess Medical Center, Boston, MA,<br />

Yan Tian, Research Assistant, Harvard Medical School, Beth<br />

Israel Deaconess Medical Center, Boston, MA, Nicolas<br />

Degauque, Research Fellow, Harvard Medical School, Beth<br />

Israel Deaconess Medical Center, Boston, MA, Wei Yang,<br />

Research Fellow, Harvard Medical School, Beth Israel<br />

Deaconess Medical Center, Boston, MA, Allison Mikita,<br />

Research Assistant, Harvard Medical School, Beth Israel<br />

Deaconess Medical Center, Boston, MA, Xin Xiao Zheng,<br />

Assistant Pr<strong>of</strong>essor, Harvard Medical School, Beth Israel<br />

Deaconess Medical Center, Boston, MA


Abstracts<br />

Regulatory T cells play vital roles in maintaining peripheral<br />

tolerance to auto- and alloantigens. Further characterization<br />

<strong>of</strong> the function and development <strong>of</strong> these regulatory<br />

T cells will contribute to understanding <strong>of</strong> the intrinsic and<br />

extrinsic mechanisms for peripheral tolerance. Our study<br />

showed that proliferated CD4+ T cells could convert to CD4−<br />

CD8−CD3+ (DN) regulatory T cells after either allogeneic or<br />

syngeneic DC triggered proliferation, and IL-2 and IL-15<br />

enhanced the conversion. Converted DN T cells were<br />

resistant to AICD and expressed unique cell markers and<br />

gene pr<strong>of</strong>ile. Converted DN T cells exerted powerful<br />

inhibition on the same, but not third party, alloantigen<br />

triggered proliferation <strong>of</strong> naive CD4+CD25− T effectors. It<br />

was notable that at a per cell base comparison, converted DN<br />

T cells were more potent in suppressing alloantigen trigged<br />

naive T cells proliferation than naive and proliferated CD4+<br />

CD25+ Tregs. Perforin, a cytotoxic cytokine, which was highly<br />

expressed by DN T cells, played a role in DN T cell mediated<br />

suppression. Adoptive transferring <strong>of</strong> T cells from new onset<br />

diabetic NOD mice precipitated a rapid onset <strong>of</strong> diabetes in<br />

NOD/SCID recipients. Co-transferring <strong>of</strong> DN T cells (in vitro<br />

converted from CD4+ T cells from new onset diabetic NOD)<br />

with diabetogenic T cells significantly delay the onset <strong>of</strong><br />

diabetes in NOD/SCID mice (P=0.0012). Moreover, the small<br />

number <strong>of</strong> converted DN can reverse diabetes after disease<br />

onset in NOD mice. The results <strong>of</strong> using ex vivo CD4+ T cells<br />

converted DN T cells in NOD mouse model support the<br />

concept and the feasibility <strong>of</strong> potentially utilizing this novel<br />

cell-based therapeutic approach clinically for the treatment<br />

<strong>of</strong> autoimmune diseases.<br />

doi:10.1016/j.clim.2007.03.216<br />

F.4 Innate Immune Modulation <strong>of</strong> Type 1 Diabetes<br />

with Glycosphingolipids<br />

Dalam Ly, Graduate Student, University <strong>of</strong> Western Ontario,<br />

Microbiology and <strong>Immunology</strong>, London, ON, Canada, Terry<br />

Delovitch, Senior Scientist/Pr<strong>of</strong>essor, Robarts Research<br />

Institute/University <strong>of</strong> Western Ontario, London, ON,<br />

Canada<br />

The pathogenesis <strong>of</strong> T1D may be mediated by functional<br />

deficiencies in CD4+CD25+Foxp3+ Tregs and iNKT cells. As<br />

self-reactive Tregs and iNKT cells both protect NOD mice<br />

from T1D, we investigated whether iNKT-Treg collaboration<br />

is required for this protection. While α-galactosylceramide<br />

(α-GalCer) therapy does not alter the proliferation, clonal<br />

expansion or function <strong>of</strong> Tregs, protection from T1D by Tregs<br />

does not require iNKT cell activity. However, Treg activity is<br />

required for α-GalCer induced protection from T1D by iNKT<br />

cells. Significant decreases in IFN-γ and IL-4 secretion and<br />

cellular (T, B, NK, DC) transactivation by iNKT cells were<br />

detected in the presence <strong>of</strong> functional Tregs. Thus, during<br />

α-GalCer therapy <strong>of</strong> T1D, activated iNKT cells transactivate<br />

Tregs that may then downregulate iNKT cell responses and<br />

return iNKT cells to a homeostatic level <strong>of</strong> activation. Such<br />

Treg-dependent homeostatic control <strong>of</strong> iNKT cell activity<br />

may explain how Treg/iNKT collaboration protects from<br />

T1D. In contrast, we found that Treg activity is not required<br />

for protection from T1D induced by the C20:2 analog <strong>of</strong> α-<br />

GalCer, which possesses a shortened (C20) fatty acyl chain<br />

and cis-unsaturations at carbons 11 and 14. This may arise<br />

from the ability <strong>of</strong> C20:2 to elicit significantly reduced IFN-γ<br />

and enhanced IL-4 secretion by iNKT cells relative to α-<br />

GalCer. Thus, α-GalCer and C20:2 may differ in their<br />

requirement <strong>of</strong> Tregs for iNKT mediated protection from<br />

T1D. Ongoing studies <strong>of</strong> the mechanism(s) <strong>of</strong> this differential<br />

requirement <strong>of</strong> Tregs for protection from T1D may<br />

have significant implications for innate immunity focused<br />

T1D therapeutic approaches.<br />

doi:10.1016/j.clim.2007.03.217<br />

F.5 Targeted Cellular Gene Therapy Using IL-4<br />

Secreting DCs Corrects Immune Dysregulations and<br />

Prevents Diabetes in NOD Mice<br />

Remi J. Creusot, Postdoctoral Fellow, Stanford University,<br />

Department <strong>of</strong> Medicine, Stanford, CA, Shahriar Yaghoubi,<br />

Research Associate, Stanford University, Department <strong>of</strong><br />

Radiology, Stanford, CA, Keiichi Kodama, Postdoctoral<br />

Fellow, Stanford University, Department <strong>of</strong> Medicine,<br />

Stanford, CA, Sam S. Gambhir, Pr<strong>of</strong>essor, Stanford<br />

University, Department <strong>of</strong> Radiology, Stanford, CA, Demi<br />

Dang, Research Assistant, Stanford University, Department<br />

<strong>of</strong> Medicine, Stanford, CA, C. Garrison Fathman, Pr<strong>of</strong>essor,<br />

Stanford University, Department <strong>of</strong> Medicine, Stanford, CA<br />

A deficit in IL-4 has been associated with the development<br />

<strong>of</strong> type 1 diabetes in man and in the non-obese diabetic<br />

(NOD) mouse. We set out to correct this deficiency by<br />

delivering IL-4 using bone marrow-derived dendritic cells<br />

transduced to express IL-4 (DC/IL-4). DC/IL/4 were injected<br />

into 12-week-old female NOD mice with advanced prediabetic<br />

insulitis. After either intravenous (iv) or intraperitoneal<br />

(ip) injection <strong>of</strong> DC/IL-4, the onset <strong>of</strong> the disease was<br />

delayed for ∼5 weeks and the majority <strong>of</strong> the treated mice<br />

were protected until at least 30 weeks <strong>of</strong> age. Using<br />

bioluminescence imaging and biodistribution assays, we<br />

demonstrated that iv-injected DCs trafficked primarily to<br />

the spleen and pancreatic lymph nodes (PLN), with little or<br />

no homing to other lymph nodes. While equally effective, ipinjected<br />

DCs showed preferential migration to the PLN, with<br />

some homing to the pancreas. We then asked what effect<br />

DC/IL-4 had in the PLN by performing cDNA microarray<br />

analysis. Initial studies demonstrated that many genes were<br />

significantly dysregulated in the PLN <strong>of</strong> 12-week-old female<br />

NOD mice, compared to non-diabetic congenic NOD.B10<br />

mice. Interestingly, the expression level <strong>of</strong> most <strong>of</strong> these<br />

dysregulated genes (N200), in addition to IL-4 itself, was<br />

brought towards normal levels as early as 3 days after DC/IL-<br />

4 treatment. MHC-deficient DC/IL-4 had no protective<br />

effect, demonstrating that interaction <strong>of</strong> the transduced<br />

DCs with T cells was required for the therapeutic effect. In<br />

conclusion, the effects <strong>of</strong> DC/IL-4 were localized, rapid, long<br />

lasting and characterized by the normalization <strong>of</strong> many<br />

dysregulated genes.<br />

doi:10.1016/j.clim.2007.03.218<br />

S17


S18 Abstracts<br />

F.6 Plasmacytoid Dendritic Cells Induction by a<br />

Self-peptide Ep1.B Derived from Apolipoprotein E<br />

Prevent Autoimmune Diabetes<br />

Enayat Nikoopour, Postdoctoral Fellow, Department <strong>of</strong><br />

Microbiology and <strong>Immunology</strong> and Robarts Research<br />

Institute, London, ON, Canada, Tracey A. Stephens,<br />

Graduate Student, Department <strong>of</strong> Microbiology and<br />

<strong>Immunology</strong> and Robarts Research Institute, London, ON,<br />

Canada, Beverley J. Rider, Graduate Student, Department<br />

<strong>of</strong> Microbiology and <strong>Immunology</strong> and Robarts Research<br />

Institute, London, ON, Canada, Edwin Lee-Chan, Lab<br />

Technician/Lab Manager, Department <strong>of</strong> Microbiology and<br />

<strong>Immunology</strong> and Robarts Research Institute, London, ON,<br />

Canada, Bhagirath Singh, Pr<strong>of</strong>essor, Department <strong>of</strong><br />

Microbiology and <strong>Immunology</strong> and Robarts Research<br />

Institute, London, ON, Canada<br />

Dendritic cells (DC) are potent inducers <strong>of</strong> T cell<br />

tolerance. The mechanism by which this is achieved is not<br />

well understood. We postulated that in vivo induction <strong>of</strong><br />

plasmacytoid dendritic cells (PDC) could prevent autoimmunity<br />

through induction <strong>of</strong> T cell tolerance. We report that a<br />

novel self-peptide Ep1.B <strong>of</strong> mouse Apolipoprotein E (ApoE)<br />

induced differentiation <strong>of</strong> bone marrow derived monocytes<br />

(BMM) into PDC. In vitro Ep1.B induced PDCs are B220+, Ly6-<br />

C+, CD11c+, mPDCA+, CD62L+ and CD8a+. Footpad injection<br />

<strong>of</strong> Ep1.B in diabetes prone NOD mice increased the level <strong>of</strong><br />

CD11c+, mPDCA and B220+ cells in the draining lymph nodes<br />

(LN) and these CD11c+ cells have an increased expression <strong>of</strong><br />

tolerogenic programmed death ligand (PD-L1). Since PD-1-<br />

PD-L1 pathway has been shown to be important in maintaining<br />

tolerance, PDC induced by Ep1.B may inhibit the effector<br />

T cells in disease process. In addition, we found increased<br />

number <strong>of</strong> CD4+CD25+ T cells with elevated glucocorticoidinduced<br />

tumor necrosis factor receptor related protein<br />

(GITR) in the LN <strong>of</strong> these animals. We found that injection<br />

<strong>of</strong> Ep1.B in young NOD mice significantly prevented type 1<br />

diabetes development in these mice. In summary, we suggest<br />

that in vivo Ep1.B induced differentiation <strong>of</strong> BMM into PDC<br />

that in turn prevented autoimmunity and induced regulatory<br />

T cells in type 1 diabetes prone NOD mice.<br />

doi:10.1016/j.clim.2007.03.219<br />

F.7 Generation and Characterization <strong>of</strong> Insulin<br />

Peptide-specific Regulatory T Cells<br />

Marianne Martinic, Postdoctoral Fellow, Immune<br />

Regulation Lab DI-3, La Jolla Institute for Allergy and<br />

<strong>Immunology</strong>, La Jolla, CA, Lisa Togher, Technician, Immune<br />

Regulation Lab DI-3, La Jolla Institute for Allergy and<br />

<strong>Immunology</strong>, La Jolla, CA, Christophe Filippi, Postdoctoral<br />

Fellow, Immune Regulation Lab DI-3, La Jolla Institute for<br />

Allergy and <strong>Immunology</strong>, La Jolla, CA, Jean Jasinski, PhD<br />

Student, Barbara Davis Center for Childhood Diabetes,<br />

Aurora, CO, Damien Bresson, Postdoctoral Fellow, Immune<br />

Regulation Lab DI-3, La Jolla Institute for Allergy and<br />

<strong>Immunology</strong>, La Jolla, CA, George Eisenbarth, Executive<br />

Director, Barbara Davis Center for Childhood Diabetes,<br />

Aurora, CO, Matthias von Herrath, Division Head, Immune<br />

Regulation Lab DI-3, La Jolla Institute for Allergy and<br />

<strong>Immunology</strong>, La Jolla, CA<br />

Our goal is to generate insulin peptide-specific regulatory<br />

T cells (Tregs) in vitro, which suppress overt diabetes in<br />

prediabetic and reverse already ongoing disease in diabetic<br />

mice. Furthermore we aim to analyze the suppressive<br />

properties and mechanisms <strong>of</strong> these Tregs in vitro and in<br />

vivo. In order to generate insulin peptide-specific Tregs, we<br />

took advantage <strong>of</strong> insTCR transgenic mice, which express T<br />

cell receptor (TCR) transgenic CD4+ T cells specific for<br />

the insulinB9–23 peptide (insB) presented on MHC class<br />

II H-2IAg7/d molecules. The in vitro generation <strong>of</strong> insBspecific<br />

Tregs involved purification <strong>of</strong> either CD4+CD25+ or<br />

CD4+CD25− insTCR T cells, which were cultured for 1–<br />

2 weeks with the insB peptide, syngeneic antigen presenting<br />

cells and high doses <strong>of</strong> IL-2 yielding 25+ and 25− cultures,<br />

respectively. Using the classical in vitro suppression assay,<br />

only cells derived from the 25+ cultures were able to<br />

suppress while cells from the 25− cultures enhanced<br />

proliferation and cytokine secretion <strong>of</strong> CD8+ effector T<br />

cells. In vivo, however, cells from both cultures were unable<br />

to suppress lymphocytic choriomeningitis virus-induced<br />

diabetes. Interestingly, freshly isolated as well as IL-10cultured<br />

CD4+CD25− but not freshly isolated CD4+CD25+<br />

insTCR T cells suppressed spontaneous diabetes in NOD<br />

females. We are currently investigating the mechanisms<br />

underlying the in vivo suppressive potential <strong>of</strong> these CD4<br />

+CD25− T cells.<br />

doi:10.1016/j.clim.2007.03.220<br />

F.8 The Insulitis Reporter Mouse<br />

Jennifer Ondr, Postdoctoral Fellow, Cincinnati Children’s<br />

Hospital Medical Center, Division <strong>of</strong> Endocrinology, Diabetes<br />

Research Center, Cincinnati, OH, Robert Opoka, Research<br />

Assistant, Cincinnati Children’s Hospital Medical Center,<br />

Division <strong>of</strong> Endocrinology, Diabetes Research Center,<br />

Cincinnati, OH, Sankarannand Vukkadapu, Postdoctoral<br />

Fellow, Cincinnati Children’s Research Foundation,<br />

Cincinnati, OH, Jonathan Katz, Associate Pr<strong>of</strong>essor,<br />

Cincinnati Children’s Hospital Medical Center, Division <strong>of</strong><br />

Endocrinology, Diabetes Research Center, Cincinnati, OH<br />

Type 1 diabetes mellitus (T1DM) is caused by the<br />

autoimmune destruction <strong>of</strong> insulin producing beta cells by<br />

leukocytes that infiltrate the pancreas in a prolonged and<br />

clinically silent process termed insulitis. Inability to detect<br />

insulitis prior to the onset <strong>of</strong> overt disease symptoms stymies<br />

progress in T1DM research and treatment. In humans,<br />

detection <strong>of</strong> antibody to islet cell antigens is the only means<br />

to diagnose pre-diabetic insulitis, however, no relationship<br />

between sero-conversion and insulitis progression is established.<br />

In NOD mice, insulitis can be measured, but only as an<br />

end-stage pancreatectomy. An early, accurate diagnosis <strong>of</strong><br />

insulitis would allow the possibility <strong>of</strong> therapeutic intervention,<br />

thus there remains an urgent need for non-invasive and<br />

real-time measures <strong>of</strong> insulitis. To this end, we are generating<br />

a gene-targeted NOD mouse in which production <strong>of</strong> a


Abstracts<br />

molecular reporter, soluble human alkaline phosphatase<br />

(sHPAP), is controlled by a gene, Reg3γ, expressed exclusively<br />

by islets cells <strong>of</strong> pancreata undergoing insulitis. Our insulitis<br />

reporter (InsuRe) mouse will self-indicate the onset and<br />

severity <strong>of</strong> insulitis when Reg3γ expression, triggered by the<br />

infiltration <strong>of</strong> pancreatic islets, induces rapid and measurable<br />

secretion <strong>of</strong> sHPAP into the blood. We will validate the fidelity<br />

<strong>of</strong> sHPAP expression in the InsuRe mouse with multiple<br />

established NOD mouse models <strong>of</strong> insulitis and diabetes.<br />

The InsuRe mouse will allow the identification <strong>of</strong> biomarkers<br />

that appear in the blood <strong>of</strong> NOD mice throughout insulitis. The<br />

identification <strong>of</strong> these biomarkers will aid in the non-invasive<br />

detection <strong>of</strong> occult insulitis, or diagnosis <strong>of</strong> pre-diabetes, in<br />

both mice and humans.<br />

doi:10.1016/j.clim.2007.03.221<br />

F.9 Identification <strong>of</strong> Candidate IDDM Disease<br />

Susceptibility Genes in the Idd Regions <strong>of</strong> NOD Mice<br />

by Temporal Microarray Gene Expression Data<br />

Analysis<br />

Keiichi Kodama, Postdoctoral Fellow, Stanford University,<br />

Stanford, CA, Demi Dang, Tech Person, Stanford University,<br />

Stanford, CA, Claire Holness, PhD, Stanford University,<br />

Stanford, CA, Hideyuki Iwai, MD. PhD, Stanford<br />

University, Stanford, CA, Mark Hartnett, Engineer,<br />

Agilent Technologies, Inc, Santa Clara, CA, Atul Butte,<br />

Assistant Pr<strong>of</strong>essor, Stanford University, Stanford, CA,<br />

C. Garrison Fathman, Pr<strong>of</strong>essor, Stanford University,<br />

Stanford, CA<br />

Using Agilent 41K mouse genome oligo-microarrays to<br />

study temporal gene expression, we identified candidate<br />

genes in the Idd disease susceptibility regions <strong>of</strong> NOD mice<br />

dysregulated by the pathophysiology <strong>of</strong> disease, not as<br />

mutations or polymorphic variants <strong>of</strong> disease susceptibility<br />

genes. Pancreatic lymph nodes (PLN) and spleen (SPL) were<br />

removed from 10-day, 4-week, 8-week, 12-week, 16-week,<br />

and 20-week-old NOD female mice, or as control from 10 day<br />

and 20-week-old genetically identical (except the MHC),<br />

NOD.B10Sn-H2b/J (NOD.B10) female mice (n¡Ü10). At 10 days<br />

to 4 weeks <strong>of</strong> age, coincident with the initiation <strong>of</strong> disease,<br />

there were ∼500 significantly dysregulated genes detected in<br />

the NOD PLN but none in the spleen, supporting the possibility<br />

that the PLN may be an important site for disease initiation.<br />

∼450 significantly dysregulated genes were seen in the PLN<br />

and ∼400 in splenic tissues from 12- to 20-week-old mice,<br />

suggesting that both tissues may be important in disease<br />

progression. b100 genes were simultaneously dysregulated in<br />

both the PLN and SPL during this period. Two <strong>of</strong> these, Ptpn22<br />

and Chi3l3, are located within the Idd 18.2 region. Expression<br />

changes for these two genes were confirmed by qPCR using<br />

the same mouse RNA samples as were used for the<br />

microarrays. This strategy may allow identification <strong>of</strong> Idd<br />

candidate genes that play critical roles in the induction or<br />

progression <strong>of</strong> autoimmune type 1 diabetes that are the<br />

consequence, not the cause <strong>of</strong> disease.<br />

doi:10.1016/j.clim.2007.03.222<br />

F.10 Peptide-based Immunotherapy for Type 1<br />

Diabetes; The Antigen, the Dose, the Immunization<br />

Route and the State <strong>of</strong> the Disease Can Make a<br />

Difference<br />

Georgia Fousteri, Postdoctoral Fellow, La Jolla Institute<br />

for Allergy and <strong>Immunology</strong>, La Jolla, CA, Amy Dave,<br />

Student, University <strong>of</strong> California, San Diego, La Jolla, CA,<br />

Michael Cr<strong>of</strong>t, Pr<strong>of</strong>essor, La Jolla Institute for Allergy and<br />

<strong>Immunology</strong>, La Jolla, CA, Matthias von Herrath,<br />

Pr<strong>of</strong>essor, La Jolla Institute for Allergy and <strong>Immunology</strong>,<br />

La Jolla, CA<br />

Immunization with beta-cell derived antigens such as<br />

insulin has been under intense investigation for treating or<br />

preventing T1D. This strategy is <strong>of</strong> particular interest since<br />

the administration <strong>of</strong> immunosuppressive drugs may be<br />

circumvented. We compared several islet-antigen derived<br />

peptides (insulin and IGRP) given alone or in combination<br />

by two different routes (intranasally [i.n.] or subcutaneously<br />

[s.c.]) to 10-week-old prediabetic NOD mice.<br />

Surprisingly, and in contrast to previous studies, we found<br />

that neither intranasal nor subcutaneous administration <strong>of</strong><br />

human pro-insulin II peptide B24–C36 (hpII) affected T1D,<br />

whereas insB-chain 9–23 (B9–23) and its altered-peptide<br />

ligand (Ala 16, 19) (APL) accelerated diabetes development<br />

significantly following intranasal administration. Combination<br />

<strong>of</strong> the three CD4 epitopes did not affect T1D when<br />

administered by either route. When insB-chain 15–23 (B15–<br />

23) and mouse insB24–36 (B24–C36) CD8 epitopes were<br />

tested (s.c. route), B15–23 delayed and B24–36 accelerated<br />

diabetes. Overall the diabetes incidence reached<br />

similar levels in both groups. Lastly, when two IGRPderived<br />

altered peptide ligands NRPI4 and NRPV7 were<br />

compared for their protective effect following s.c. immunization,<br />

NRPV7 provided the greatest delay but without a<br />

long-term protection. In contrast to our present findings,<br />

mucosal immunization with whole oral or intranasal insulin<br />

or insB9–23 peptide combined with suitable adjuvants (i.<br />

e., IFA) was able to prevent diabetes efficiently in previous<br />

studies, especially when given at a much earlier stage (4to<br />

6-week-old NODs). It will be important to consider the<br />

issues <strong>of</strong> age, route <strong>of</strong> administration and adjuvants for the<br />

design <strong>of</strong> future clinical trials.<br />

doi:10.1016/j.clim.2007.03.223<br />

S19<br />

F.11 Monocyte Immunophenotypes in Type 1<br />

Diabetes<br />

Erik Fung, DRF Fellow, Department <strong>of</strong> Medical Genetics,<br />

University <strong>of</strong> Cambridge, England, Cambridge, England, John<br />

A. Todd, Pr<strong>of</strong>essor and Director <strong>of</strong> Laboratory, Department<br />

<strong>of</strong> Medical Genetics, University <strong>of</strong> Cambridge, England,<br />

Cambridge, England, Linda S. Wicker, Pr<strong>of</strong>essor and<br />

Co-Director <strong>of</strong> Laboratory, Department <strong>of</strong> Medical Genetics,<br />

University <strong>of</strong> Cambridge, England, Cambridge, England<br />

Type 1 diabetes (T1D) is a complex-trait disease that<br />

results from interaction <strong>of</strong> genes and environmental<br />

exposures leading to immune destruction <strong>of</strong> pancreatic<br />

islet cells. Previous studies have indicated that monocytes


S20 Abstracts<br />

may be altered in chronic T1D. However, it is still unclear if<br />

this immune phenotypic variation is a cause or effect <strong>of</strong><br />

T1D. Using polychromatic flow cytometry to analyze whole<br />

blood, we have extended to pediatric T1D patients the<br />

findings by others that adult T1D cases have increased<br />

expression <strong>of</strong> CD11b on monocytes as compared to<br />

apparently healthy non-T1D adults. We examined 37 T1D<br />

patients (age 9.2–18.3 years, median 14.4 years; median<br />

time past diagnosis for non-newly diagnosed cases=4.3<br />

years) and 63 healthy volunteers (age 9.9–60 years, median<br />

34 years). CD11b expression (MFI) in HLA-DR+ monocyte<br />

subsets were as follows: on CD16+ monocytes, 265±20 vs.<br />

218±10 (pediatric T1D cases vs. non-T1D adults); CD16+<br />

CD14+ monocytes, 1108 ±51 vs. 840±27; CD14++ ‘inflammatory’<br />

monocytes, 1479±38 vs. 1146±54. Expression<br />

levels <strong>of</strong> CCR2 on CD14++ monocytes were lower in T1D<br />

patients than in healthy volunteers (MFI 62±2 vs. 75±2),<br />

consistent with an activated monocyte phenotype. CD11b<br />

and CCR2 expression differences were independent <strong>of</strong> ages<br />

at diagnosis or at immunophenotyping. In order to help<br />

distinguish between cause and effect and begin to explore<br />

the possibility that extremes <strong>of</strong> these phenotypes may be<br />

precursors to disease, we will analyze age-matched<br />

subjects and unaffected siblings, and test the association<br />

<strong>of</strong> alleles <strong>of</strong> known T1D susceptibility genes (e.g., HLA<br />

haplotypes, INS, PTPN22, CD25, IFIH1, CTLA4) with monocyte-subset<br />

phenotypes.<br />

doi:10.1016/j.clim.2007.03.224<br />

F.12 A High Specificity Competitive Europium Based<br />

Assay for Autoantibodies <strong>of</strong> APS1 Patients Reacting<br />

with Interferon Alpha<br />

Li Zhang, Fellow, Barbara Davis Center, Denver, CO,<br />

Raffaele Badolato, MD, PhD, Department <strong>of</strong> Pediatrics,<br />

University <strong>of</strong> Brescia, Brescia, Jennifer Barker, Assistant<br />

Pr<strong>of</strong>essor <strong>of</strong> Pediatrics, Barbara Davis Center for Childhood<br />

Diabetes, Denver, CO, Maureen Su Pra, University <strong>of</strong><br />

California, San Francisco Diabetes Center, San Francisco,<br />

CA, Sunanda Babu, Research Associate, Barbara Davis<br />

Center, Denver, CO, Mickie Cheng, MD, PhD, University <strong>of</strong><br />

California, San Francisco Diabetes Center, San Francisco,<br />

CA, Tony Shum, MD, University <strong>of</strong> California, San Francisco<br />

Diabetes Center, San Francisco, CA, Ehud Zamir, MD, The<br />

Royal Victorian Eye and Ear Hospital, Victoria, BC, Canada,<br />

Adam Law, MD, Ithaca Medical School, Ithaca, NY, George<br />

Eisenbarth, Executive Director, Barbar Davis Center, Denver,<br />

CO, Mark Anderson, Assistant Pr<strong>of</strong>essor, University <strong>of</strong><br />

California, San Francisco Diabetes Center, San Francisco, CA<br />

IFN-α autoantibodies have been described in autoimmune<br />

polyglandular syndrome type 1 (APS1) patients. We have<br />

established a competitive Europium time resolved fluorescence<br />

assay for autoantibodies to IFN-α and have tested it in<br />

a cohort <strong>of</strong> APS1 patients. Methods: The europium-ELISA<br />

method utilizes plate bound IF α incubated with or without<br />

competition with fluid phase IFN-α and sera. Anti-IgG<br />

biotinylated antibody is added, followed by incubation with<br />

streptavidin–europium and detection with time resolved<br />

fluorescence (measured in counts per second, CPS). Seven<br />

APS1 patients, 6 relatives, 71 non-APS1 Addison patients and<br />

141 T1DM patients were tested as well as 100 normal<br />

controls. The APS1 patients had multiple different mutations<br />

in the AIRE gene. Results: Normal control CPS without<br />

competition was 31,237∼17,328 CPS and delta with competition<br />

was −6563∼10,303 CPS. The initial APS1 patient (used<br />

to create the index, index 1.0) gave 394,063 CPS without<br />

competition and a delta <strong>of</strong> 363,662∼31,587 CPS with<br />

competition. Scatchard plot analysis revealed a high avidity<br />

(Kd <strong>of</strong> 0.5 nM). The CPS, delta and index for 6/7 APS1<br />

patients were strongly positive and above 3 standard<br />

deviations <strong>of</strong> controls. Relatives were negative as were 71<br />

Addison disease (non-APS-1) and 141 T1DM patients. The one<br />

negative APS1 patient is presumed APS1 based on clinical<br />

history. The assay has a sensitivity <strong>of</strong> 86% or greater and<br />

specificity greater than 99.5%. Conclusion: IFN-α autoantibodies<br />

are detected specifically in APS1 patients with<br />

specificity greater than 99% using a competitive Europium<br />

time resolved fluorescence assay.<br />

doi:10.1016/j.clim.2007.03.225<br />

F.13 Validation <strong>of</strong> Anti-Idiotypic Bridging ELISA to<br />

Quantitate TRX4 (Anti-Human CD3) Mab Levels in<br />

Human Serum<br />

Francis Carmody, Preclinical Scientist III, Preclinical<br />

Development, Cambridge, MA, Michael Slavonic, Preclinical<br />

Scientist II, Tolerx, Preclinical Development, Cambridge, MA,<br />

Adam O’Shea, Research Scientist II, Tolerx, Applied Research,<br />

Cambridge, MA, Michael Paglia, Senior Manager Manufacturing,<br />

Tolerx, Manufacturing, Cambridge, MA, Reema Gulati,<br />

Research Scientist II, Tolerx, Applied Research, Cambridge, MA,<br />

Sharon Li, Former Tolerx Employee, Applied Research,<br />

Cambridge, MA, Herman Waldmann, Pr<strong>of</strong>essor <strong>of</strong> Pathology,<br />

Head <strong>of</strong> Department, University <strong>of</strong> Oxford, Sir William Dunn<br />

School <strong>of</strong> Pathology, Cambridge, MA, Michale Rosenzweig,<br />

Director Preclinical Development, Tolerx, Preclinical<br />

Development, Cambridge, MA<br />

TRX4 is an aglycosyl anti-human CD3μ monoclonal antibody<br />

(Mab) that is being evaluated as a therapy for autoimmune<br />

disorders such as new-onset type 1 diabetes. Our goal was to<br />

develop a sensitive, high throughput assay to quantitate TRX4<br />

serum levels that could be used as an alternative to a cell-based<br />

assay that had been used in previous studies. Historically, TRX4<br />

was detected with a polyclonal anti-human IgG after it bound to<br />

the human T cell line, HuT 78. This method was both laborious<br />

and lacked sufficient sensitivity. Two different approaches were<br />

used to raise monoclonal antibodies to TRX4 complementarity<br />

determining regions (CDRs). First, transgenic human CD3 mice<br />

were tolerized to human IgG by two 1 mg IV doses and then<br />

subsequently immunized with 100 μg IP <strong>of</strong> TRX4. For the second<br />

strategy, BALB/c mice were administered 100 μg SC <strong>of</strong> TRX4 F<br />

(ab)′2 fragments in incomplete Freund’s adjuvant. These<br />

independent immunization strategies produced two non-competing<br />

anti-idiotypic TRX4 Mabs that satisfied specificity<br />

criteria. Significant serum interference was observed with<br />

both the Mabs when used individually in ELISAs. However,<br />

when used together, these anti-idiotypic antibodies were able to<br />

capture and detect TRX4 in whole sera with no detectable


Abstracts<br />

enhancement or inhibition due to serum alone. Assay validation<br />

has confirmed the accuracy (±20%), precision (±20%), specificity<br />

and robustness necessary to fulfil the ng/mL detection goal<br />

sought. Once validated, the ELISA was utilized as a crossover<br />

analysis between preclinical and clinical programs in support <strong>of</strong><br />

our TRX4 development program.<br />

doi:10.1016/j.clim.2007.03.226<br />

F.14 Regulation <strong>of</strong> Insulin Expression in Thymic<br />

Epithelial Cells<br />

Dina Levi, PhD Candidate, McGill University, Human<br />

Genetics, Pointe-Claire, QC, Canada, Michael Palumbo, PhD<br />

Candidate, McGill University, Experimental Medicine,<br />

Montreal, QC, Canada, Constantin Polychronakos, Principal<br />

Investigator, Montreal Children’s Hospital Research<br />

Institute, Montreal, QC, Canada<br />

Thethymushasanessentialfunctionintheselection<strong>of</strong>T<br />

cells with a properly formed TCR and the elimination <strong>of</strong> selfreactive<br />

ones. It has been found that tissue specific selfantigens<br />

are expressed in thymic epithelial cells, specifically<br />

in the medulla, for the purpose <strong>of</strong> presentation to thymocytes.<br />

Insulin is an important self-antigen that is implicated in the<br />

destruction <strong>of</strong> the pancreatic beta-cells in type 1 diabetes. We<br />

have isolated and cultured insulin-expressing (as well as<br />

insulin-non-expressing) epithelial clones from the medulla <strong>of</strong><br />

the thymus (mTECs), which provide an exclusive in vitro<br />

model to study self-antigen expression in the thymus.<br />

Preliminary studies show a number <strong>of</strong> tissue specific selfantigens<br />

overexpressed by microarray gene expression pr<strong>of</strong>iling,<br />

in both insulin-expressing and non-expressing clones, with<br />

the majority <strong>of</strong> tissues being represented. Stimulation <strong>of</strong><br />

mTECs with IFN- 3 and TNF-± cytokines demonstrates a pattern<br />

<strong>of</strong> decrease in insulin expression by real-time RT-PCR,<br />

indicating a possible regulation mechanism. Furthermore,<br />

co-culturing mTECs with isolated thymocytes results in an<br />

increase in insulin expression when cells are in direct contact.<br />

This interaction is supposed essential for the proper selection<br />

<strong>of</strong> non-autoreactive T lymphocytes in order to prevent<br />

autoimmune disease. Therefore, the regulation <strong>of</strong> insulin<br />

self-antigen expression in the thymus is an essential mechanism<br />

involved in negative selection and central tolerance and<br />

necessitates further studies.<br />

doi:10.1016/j.clim.2007.03.227<br />

F.15 Dysregulation <strong>of</strong> the TIM-3-Galectin-9 Pathway<br />

in Type I Diabetic T Cells<br />

William Hastings, Postdoctoral Fellow, Brigham and<br />

Women’s Hospital/Harvard Medical School, Center for<br />

Neurologic Diseases, Boston, MA, Allison Greer, Research<br />

Technician, Brigham and Women’s Hospital/Harvard<br />

Medical School, Center for Neurologic Diseases, Boston, MA,<br />

Vijay Kuchroo, Pr<strong>of</strong>essor, Brigham and Women’s Hospital/<br />

Harvard Medical School, Center for Neurologic Diseases,<br />

Boston, MA, David Hafler, Pr<strong>of</strong>essor, Brigham and Women’s<br />

Hospital/Harvard Medical School, Center for Neurologic<br />

Diseases, Boston, MA, David Anderson, Instructor, Brigham<br />

and Women’s Hospital/Harvard Medical School, Center for<br />

Neurologic Diseases, Boston, MA, Sally Kent, Pr<strong>of</strong>essor,<br />

Brigham and Women’s Hospital/Harvard Medical School,<br />

Center for Neurologic Diseases, Boston, MA<br />

Our laboratory has generated, in a non-biased manner, a<br />

large panel <strong>of</strong> T cell clones from the pancreatic lymph nodes<br />

(PLN) <strong>of</strong> type 1 diabetic (T1D) subjects and we asked if there<br />

were alterations in function <strong>of</strong> T cells from the PLN <strong>of</strong> T1D<br />

subjects versus those from controls. In this regard, we have<br />

recently shown that TIM-3, a molecule associated with negative<br />

regulation <strong>of</strong> fully differentiated Th1 but not Th2 cells, is<br />

dysregulated on T cell clones isolated from the CSF <strong>of</strong> MS<br />

patients. In light <strong>of</strong> this, we analyzed Tcell clones from the PLN<br />

<strong>of</strong> T1D as well as from normal controls to determine if there are<br />

similar alterations in expression and/or function <strong>of</strong> TIM-3 and its<br />

ligand Galectin-9 in these cells. We report that PLN clones from<br />

T1D subjects express lower levels <strong>of</strong> Galectin-9 but not TIM-3<br />

mRNA than clones from normal controls, as analyzed by Taqman<br />

PCR. In addition, we show that blockade <strong>of</strong> TIM-3 signaling with<br />

antibody increased proliferation and IFNg secretion in PLN Tcell<br />

clones. Interestingly, FACS analysis <strong>of</strong> PLN cells from controls and<br />

T1D subjects showed that TIM-3 is more highly expressed than on<br />

comparable subpopulations <strong>of</strong> T cells present in peripheral<br />

blood. Gene chip analysis has identified key differences in gene<br />

expression between TIM-3+ and TIM-3− T cell populations.<br />

Collectively, these data contribute to our understanding <strong>of</strong> how<br />

TIM-3 may regulate human T cell function and impact type 1<br />

diabetes.<br />

doi:10.1016/j.clim.2007.03.228<br />

S21<br />

F.16 huFlt3-L Treatment Does Not Protect RIP-GP<br />

and RIP-NP C57B1/6 Mice Against LCMV-induced<br />

Diabetes<br />

Tom Van Belle, Postdoctoral Fellow, La Jolla Institute for<br />

Allergy and <strong>Immunology</strong>, La Jolla, CA, Eleanor Ling, PhD, La<br />

Jolla Institute for Allergy and <strong>Immunology</strong>, La Jolla, CA, Lisa<br />

Togher, Research Assistant, La Jolla Institute for Allergy and<br />

<strong>Immunology</strong>, La Jolla, CA, Matthias von Herrath, PI, La Jolla<br />

Institute for Allergy and <strong>Immunology</strong>, La Jolla, CA<br />

Human Fms-like Tyrosine Kinase3-L (huFlt3-L) treatment <strong>of</strong><br />

prediabetic NOD mice has been shown to increase the<br />

intrinsically low numbers <strong>of</strong> myeloid dendritic cells in these<br />

mice, while decreasing insulitis and delaying the progression <strong>of</strong><br />

diabetes. In this study, we investigated the influence <strong>of</strong> huFlt3-L<br />

treatment on insulitis and diabetes onset in the LCMV-induced<br />

RIP-GP (fast onset) or RIP-NP (slow onset) C57Bl/6 mouse models<br />

<strong>of</strong> T1D. We showed that these mice have no intrinsic abnormality<br />

in DC subset development during LCMV infection. As expected,<br />

huFlt3-L treatment dramatically increased the total DC fraction<br />

and numbers in both spleen and pancreatic draining LN, with a<br />

preferential outgrowth <strong>of</strong> myeloid DCs (CD11c+B220−CD11b+)<br />

over plasmacytoid DCs (CD11c+B220+CD11b−). Moreover, NK<br />

cells and NK-DC cells were also increased, but CD4 and CD8 Tcell<br />

fractions were reduced. Strikingly, repeated huFlt3-L administrations,<br />

initiated in the prediabetic phase, did not protect<br />

against diabetes onset in this model. It is possible that this is


S22 Abstracts<br />

correlated with the observed reduced fraction <strong>of</strong> CD4+CD25+ T<br />

cells in the huFlt3-L-treated mice. Translation <strong>of</strong> these data<br />

suggests that huFlt3-L might only be a useful therapeutic agent<br />

for T1D patients with a demonstrated DC (subset) defect.<br />

doi:10.1016/j.clim.2007.03.229<br />

F.17 The BDC 12-4.1 TCR Alpha Chain is Sufficient to<br />

Generate Anti-Insulin Autoantibodies; α+β Chain<br />

for Insulitis<br />

Masakazu Kobayashi, Fellow, Barbara Davis Center, Aurora,<br />

CO, Jean Jasinski, Graduate Student, Barbara Davis Center,<br />

Aurora, CO, Maki Nakayama, Fellow, Barbara Davis Center,<br />

Aurora, CO, Dongmei Miao, PRA, Barbara Davis Center,<br />

Aurora, CO, Marcella Li, PRA, Barbara Davis Center, Aurora,<br />

CO, Liping Yu, PRA, Barbara Davis Center, Aurora, CO, Edwin<br />

Liu, Associate Pr<strong>of</strong>essor, Barbara Davis Center, Aurora, CO,<br />

George Eisenbarth, PI, Barbara Davis Center, Aurora, CO<br />

There is a marked T cell receptor (TCR) α chain restriction<br />

<strong>of</strong> T cell clones that target the insulin peptide B:9–23 in NOD<br />

mice. We are pursuing the hypothesis that this non-stringent<br />

TCR with conserved V α (TRAV5D-4*04) and J α (TRAJ53*01)<br />

chains (not N <strong>of</strong> α or β chain) underlies insulin autoimmunity<br />

and diabetes propensity. To test this hypothesis, we created<br />

TCR transgenic mice bearing only the BDC12-4.1 α chain,<br />

combined with C α knockout. Mice with the BDC 12-4.1 α<br />

chain, regardless <strong>of</strong> the presence or absence <strong>of</strong> the BDC 12-4.1<br />

TCR β chain, produced insulin autoantibodies, whereas mice<br />

bearing BDC 12-4.1 TCR β chain but not α chain did not. In<br />

contrast to production <strong>of</strong> insulin autoantibodies, we found<br />

insulitis in only mice with both 12-4.1 α and β chains (C α TCR<br />

knockout). Our results indicate that provision <strong>of</strong> only the<br />

BDC12-4.1 α chain results in expression <strong>of</strong> insulin autoantibodies<br />

(with endogenous β chains) and the ββ chain<br />

contributes to development <strong>of</strong> insulitis. At present we are<br />

producing mice with BDC12-4.1 α chain (Cα knockout) and β<br />

chain from anti-ovalbumin DO.11.10 T cell clone, which also<br />

produce insulin autoantibodies, but we need to combine Vβ<br />

knockout. A simple TCR motif and single insulin peptide may<br />

be essential for anti-insulin/anti-islet autoimmunity <strong>of</strong> the<br />

NOD mouse.<br />

doi:10.1016/j.clim.2007.03.230<br />

F.18 HLA-DR3/4 Heterozygosity is a Risk Factor for<br />

Autoantibody Conversion and Type 1 Diabetes<br />

Recurrence After Simultaneous Pancreas-Kidney<br />

Transplantation<br />

Stavros Diamantopoulos, Fellow, Diabetes Research<br />

Institute and Department <strong>of</strong> Pediatrics, University <strong>of</strong><br />

Miami, Miami, FL, Gloria Allende, Research Associate,<br />

Diabetes Research Institute, Miller School <strong>of</strong> Medicine,<br />

University <strong>of</strong> Miami, Miami, FL, George W. Burke, Pr<strong>of</strong>essor<br />

and Co-Director, Division <strong>of</strong> Transplantation, Department <strong>of</strong><br />

Surgery, Miller School <strong>of</strong> Medicine, University <strong>of</strong> Miami,<br />

Miami, FL, Gaetano Ciancio, Pr<strong>of</strong>essor and Director <strong>of</strong><br />

Transplant Education, Department <strong>of</strong> Surgery, Miller School<br />

<strong>of</strong> Medicine, University <strong>of</strong> Miami, Miami, FL, Alberto<br />

Pugliese, Director, Division <strong>of</strong> Immunogenetics, Diabetes<br />

Research Institute, Miller School <strong>of</strong> Medicine, University <strong>of</strong><br />

Miami, Miami, FL<br />

We studied 252 patients with type 1 diabetes (T1D) who<br />

received a simultaneous pancreas-kidney (SPK) transplant.<br />

Patients were tested for GAD and IA-2 autoantibodies, wellknown<br />

T1D risk markers. One-hundred-thirty-one patients (52%)<br />

remained GAD/IA-2 autoantibody-negative; 44 (17.4%) converted<br />

to GAD and/or IA-2 positivity while 77 (30.6%) had<br />

autoantibodies before the transplant that persisted on followup.<br />

We then analyzed recipients' HLA-DR types in relation to<br />

autoantibody conversion and T1D recurrence (T1DR) on followup<br />

(range 0.08-15.9 years, mean 5.6+3.6 years) in the 175<br />

patients who remained autoantibody-negative or converted.<br />

Twenty <strong>of</strong> 44 (45%) converters carried the HLA-DR3/4 genotype<br />

vs. 22/131 (17%) autoantibody-negative patients (p=0.0001,<br />

OR=4.5). Conversion occurred in 20/42 (47.6%) patients with the<br />

HLA-DR3/4 genotype vs. 24/133 (18%) patients without it<br />

(p=0.0001, Kaplan-Meier lifetable analysis; p=0.0002. OR=4.2,<br />

Chi-square). The non-heterozygotes included 53 patients with<br />

HLA-DR3 alone, 53 with HLA-DR4 alone and 23 with neither<br />

types. Excluding patients that did not carry HLA-DR3 or HLA-<br />

DR4, HLA-DR3/4 heterozygotes converted more <strong>of</strong>ten than<br />

patients carrying HLA-DR3 or HLA-DR4 alone (p=0.0001, Kaplan-<br />

Meier lifetable analysis; p=0.0002, OR=4.4, Chi-square). Ten<br />

patients developed T1DR on follow-up, <strong>of</strong> whom 60% were HLA-<br />

DR3/4 heterozygotes: 6/42 (14%) patients with the HLA-DR3/4<br />

genotype developed T1DR vs. 4/133 (3%) without it (p=0.01,<br />

Kaplan-Meier lifetables analysis; p=0.01, OR=5.37, Chi-square).<br />

Our data suggest that the HLA-DR3/4 genotype is a risk factor for<br />

autoantibody conversion and for the development <strong>of</strong> disease<br />

recurrence in SPK recipients. Patients with this genotype<br />

represent a high-risk group that should be monitored for<br />

markers <strong>of</strong> recurrent autoimmunity.<br />

doi:10.1016/j.clim.2007.03.231<br />

F.19 Kinetics <strong>of</strong> Regulatory T Cells Upon Specific<br />

Diabetogeneic Activation in a Group <strong>of</strong> High Risk<br />

Relatives <strong>of</strong> Type 1 Diabetes Mellitus Patients in<br />

Comparison with Healthy Controls<br />

Zuzana Vrabelova, PhD Student, University Hospital Motol,<br />

Children’s Neurology Department, Prague, Zuzana<br />

Hladikova, Statist, Czech Agriculture University,<br />

Department <strong>of</strong> Statistics, Prague, Katerina Stechova, Lab<br />

Chief, University Hospital Motol, Pediatric Department,<br />

Prague, Kristyna Bohmova, PhD Student, University Hospital<br />

Motol, Pediatric Department, Prague, Jaroslav Michalek,<br />

Head <strong>of</strong> Cell Immunotherapy Center, Masaryk University Br<br />

England Cell Immunotherapy Center, England<br />

Abnormalities in CD4+CD25+ regulatory Tcells (Tregs) may<br />

contribute to type 1 diabetes (T1D) development. We have<br />

studied immunoregulatory populations <strong>of</strong> CD4+CD25+ T cells<br />

in a group <strong>of</strong> high-risk relatives <strong>of</strong> T1D patients. We have<br />

evaluated kinetics <strong>of</strong> Tregs (using specific markers CD127−<br />

and Foxp3+) after specific stimulation with diabetogenic<br />

autoantigens in 11 high-risk (according to HLA-linked T1D<br />

genetic risk) relatives <strong>of</strong> T1D patients and 14 healthy


Abstracts<br />

controls. For stimulation specific amino acid sequences <strong>of</strong><br />

glutamic acid decarboxylase 65 (GAD65), thyrosine phosphatase<br />

(IA2), beta-proinsulin chain and whole molecule <strong>of</strong><br />

insulin were used. Cell activation was measured by surface<br />

expression <strong>of</strong> interferon gamma (IFN-g) on activated CD4+ T<br />

cells. High-risk relatives <strong>of</strong> T1D patients have significantly<br />

lower pre- and post-stimulatory numbers <strong>of</strong> Tregs than<br />

healthy controls (pb0.05). The autoantigen activation <strong>of</strong><br />

diabetogenic T cells was significantly higher in high-risk<br />

relatives <strong>of</strong> T1D patients than in healthy controls (pb0.02).<br />

In conclusion, defects <strong>of</strong> CD4+CD25+ regulatory T cells have<br />

been proved in individuals at increased genetic risk for T1D.<br />

Supported by IGA MZCR NR 9355-3.<br />

doi:10.1016/j.clim.2007.03.232<br />

F.20 Differential Immune Recognition <strong>of</strong> Heat-Shock<br />

Protein 60 Epitopes in Type 1 Diabetes<br />

Sylvia Kamphuis, Pediatric Immunologist, Department <strong>of</strong><br />

Paediatric <strong>Immunology</strong>, Erasmus MC Sophia Children’sHospital,<br />

Rotterdam,GijsTeklenburg,MD,Pediatric<strong>Immunology</strong>,UMC<br />

Utrecht, Utrecht, Annemarie Verrijn Stuart, Pediatric<br />

Endocrinologist, Department <strong>of</strong> Pediatric Endocrinology, UMC<br />

Utrecht, Utrecht, Irun Cohen, MD PhD, Department <strong>of</strong><br />

<strong>Immunology</strong>, Weizmann Institute <strong>of</strong> Science, Rehovot, Wilco de<br />

Jager, Msc, Department <strong>of</strong> <strong>Immunology</strong>, UMC Utrecht, Utrecht,<br />

Wietse Kuis, MD PhD, Department <strong>of</strong> <strong>Immunology</strong>, UMC Utrecht,<br />

Utrecht, Salvatore Albani, MD PhD, Department <strong>of</strong> Pediatrics<br />

and Medicine, University <strong>of</strong> California, San Diego, CA, Berent<br />

Prakken, MD PhD, Department <strong>of</strong> <strong>Immunology</strong>, UMC Utrecht,<br />

Utrecht, Mark Klein, Msc, Department <strong>of</strong> <strong>Immunology</strong>, UMC<br />

Utrecht, Utrecht<br />

Aim: Evidence is cumulating for an immunoregulatory role <strong>of</strong><br />

heat-shock proteins (HSP) in an increasing number <strong>of</strong> autoimmune<br />

diseases. Previously we identified pan-DR binding<br />

HSP60 epitopes that were recognized in juvenile idiopathic<br />

arthritis, an autoimmune disease characterized by chronic<br />

joint inflammation. The present study was performed to test<br />

whether these HSP60 epitopes were recognized in children<br />

with type 1 diabetes as well. Methods: We analyzed the<br />

pattern <strong>of</strong> HSP60 peptide-specific cytokine and chemokine<br />

responses in peripheral blood mononuclear cells <strong>of</strong> 18 type 1<br />

diabetes patients with primarily longstanding disease. In<br />

addition, a subgroup <strong>of</strong> newly diagnosed patients was followed<br />

over time for HSP60 peptide-specific immune responses.<br />

Results: We found peptide-specific induction <strong>of</strong> 8 cytokines<br />

(IL-1a, IL-1b, IL-6, IL-10, IL-17, IL-18, TNF-a, IFN-g) and 5<br />

chemokines (MIP-1a, MDC, IL-8, IP10, and OSM). Considerable<br />

variation in peptide-induced cytokine and chemokine pr<strong>of</strong>iles<br />

however was seen in the individual patients, irrespective <strong>of</strong><br />

age and disease duration. When following newly diagnosed<br />

patients over time, we found that the presence <strong>of</strong> T cell<br />

proliferative responses was almost exclusively seen in the<br />

patients that experienced a partial remission phase, with<br />

concomitant induction <strong>of</strong> IL-10. Conclusion: The identified<br />

HSP60 epitopes are immunogenic in children with type 1<br />

diabetes. The recorded correlation <strong>of</strong> peptide-specific T cell<br />

responses with (temporary) disease remission underscores the<br />

alleged immunomodulating role <strong>of</strong> HSP in chronic inflammation.<br />

This makes these HSP60 epitopes promising candidates<br />

for antigen-specific immunotherapy in not only juvenile<br />

idiopathic arthritis, but also in type 1 diabetes and possibly<br />

other autoimmune diseases.<br />

doi:10.1016/j.clim.2007.03.233<br />

F.21 Blocking the Toll Like Receptor 9 Signal Inhibits<br />

Activation <strong>of</strong> Diabetogenic CD8 T Cells and Delays<br />

Autoimmune Diabetes in NOD Mice<br />

Yiqun Zhang, Research Associate, Child and Family Research<br />

Institute, University <strong>of</strong> British Columbia, Vancouver, BC,<br />

Canada, Xuan Geng, Research Assistant, Kinesiology, Simon<br />

Fraser University, Burnaby, BC, Canada, Pere Santamaria,<br />

Pr<strong>of</strong>essor, Microbiology and Infectious Diseases, The University<br />

<strong>of</strong> Calgary, Calgary, AB, Canada, Diane Finegood, Pr<strong>of</strong>essor,<br />

Kinesiology, Simon Fraser University, Burnaby, BC, Canada, Jan<br />

P. Dutz, Pr<strong>of</strong>essor, CFRI, University <strong>of</strong> British Columbia,<br />

Vancouver, BC, Canada<br />

Type 1 diabetes is an autoimmune disease in which pancreatic<br />

beta cells are destroyed by CD8 T cells. Dendritic cells<br />

(DC)s are capable <strong>of</strong> priming CD8 Tcells. Toll-like receptor (TLR)<br />

triggering can lead to DC maturation. The contributions <strong>of</strong> TLR<br />

signals to spontaneous diabetes development are still largely<br />

unknown. We found that non-obese diabetic (NOD) mouse bone<br />

marrow derived DCs (BMDC)s pulsed with freeze–thawed NIT1<br />

insulinoma cells in the presence <strong>of</strong> TLR9 agonist CpG and CD40<br />

ligation were able to fully prime 8.3 TCR transgenic and<br />

diabetogenic CD8 T cells. Addition <strong>of</strong> the TLR9 inhibitors ODN<br />

2088 or chloroquine potently inhibited CD8 T cell CD25<br />

upregulation and intracellular IFN-γ production in response to<br />

CPG but not LPS. Furthermore, ODN 2088 or chloroquine<br />

inhibited CPG-induced BMDC CD40 upregulation and IL-12 p70<br />

production. In vivo, CPGaloneorwithanti-CD40induced8.3<br />

CD8 T cell CD69 upregulation and CTL activity that triggered<br />

rapid diabetes development in 8.3 NOD mice. Pre-treatment<br />

with ODN 2088 inhibited CPG-induced diabetes and treatment<br />

with either ODN 2088 or chloroquine delayed spontaneous<br />

diabetes in 8.3 NOD mice. Finally, administration <strong>of</strong> chloroquine<br />

inhibited spontaneous diabetes in NOD mice, down-regulated<br />

CD40 expression on pancreatic DCs, and inhibited serum rises <strong>of</strong><br />

IL-12 p70, IL-6, IFN-γ andMCP-1inducedbyCPGbutnotLPS.<br />

Thus, TLR9 activation may contribute to the spontaneous onset<br />

<strong>of</strong> CD8 T cell autoimmunity in NOD mice and TLR9 family<br />

inhibitors may be used to prevent and delay autoimmune<br />

diabetes in disease-prone animals.<br />

doi:10.1016/j.clim.2007.03.234<br />

S23<br />

F.22 Genome-Wide Association Scan for Type 1<br />

Diabetes Susceptibility Genes in a Danish Population<br />

Caroline Brorsson, PhD Student, Steno Diabetes Center,<br />

Gent<strong>of</strong>te, Elzbieta Swiergala, Lab Manager, Department <strong>of</strong><br />

Medical Genetics, University <strong>of</strong> British Columbia,<br />

Vancouver, BC, Canada, Krist<strong>of</strong>fer Rapacki, Computer<br />

Scientist, Center for Biological Sequence Analysis, Technical<br />

University <strong>of</strong> Denmark, Lyngby, Regine Bergholdt, Research<br />

Fellow, Steno Diabetes Center, Gent<strong>of</strong>te, Shaun Purcell,<br />

Assistant Pr<strong>of</strong>essor, Center for Human Genetic Research,


S24 Abstracts<br />

Massachusetts General Hospital, Boston, MA, Leigh Field,<br />

Pr<strong>of</strong>essor, Department <strong>of</strong> Medical Genetics, University <strong>of</strong><br />

British Columbia, Vancouver, BC, Canada, Flemming Pociot,<br />

Pr<strong>of</strong>essor, Steno Diabetes Center, Gent<strong>of</strong>te<br />

Type 1 diabetes (T1D) is a complex disease believed to<br />

result from interactions between multiple risk-conferring<br />

genes and environmental factors. In order to further<br />

investigate the genetic contribution to T1D we genotyped a<br />

case–control material for 58,000 SNP and tested these for<br />

association to disease. The first phase <strong>of</strong> the study comprised<br />

<strong>of</strong> thorough quality tests <strong>of</strong> the data for issues such as sample<br />

contamination, inbreeding, low genotyping rates, relatedness<br />

and population stratification which led to samples being<br />

excluded. SNPs were removed for failing HWE in controls, low<br />

genotyping rate, differences in missing genotyping rates<br />

between cases and controls and being non-polymorphic,<br />

which contributed to significant false positive findings. After<br />

ensuring the highest quality <strong>of</strong> the data-set the remaining<br />

52,000 SNPs and 605 individuals were analysed using basic<br />

allelic and model-based tests <strong>of</strong> association corrected for<br />

multiple testing and using permutation with empirically<br />

corrected p-values. Furthermore we performed genomewide<br />

sliding-window haplotype tests and full pair-wise test<br />

for epistasis across the genome. We also performed the same<br />

analyses for markers in 12 linkage regions previously reported<br />

in T1D. Every individual test confirmed the importance <strong>of</strong> the<br />

HLA region on chromosome 6p21.3 for developing T1D. In<br />

combination these tests point to other chromosomal regions<br />

contributing to T1D susceptibility. The significant findings<br />

will be followed up in a larger case–control material to<br />

confirm these present results. In addition this study has<br />

demonstrated the importance <strong>of</strong> high quality data for<br />

avoiding false positive findings when performing association<br />

analyses in complex diseases.<br />

doi:10.1016/j.clim.2007.03.235<br />

F.23 iNKT Cell Regulation <strong>of</strong> Type 1 Diabetes<br />

Dalam Ly, PhD Candidate, Robarts Research Institute,<br />

London, ON, Canada, Qing Sheng Mi, Research Associate,<br />

Robarts Research Institute, London, ON, Canada, Shabbir<br />

Hussain, Research Associate, Robarts Reseach Institute,<br />

London, ON, Canada, Terry L. Delovitch, Pr<strong>of</strong>essor, Robarts<br />

Research Institute, London, ON, Canada, Steven A. Porcelli,<br />

Pr<strong>of</strong>essor, Albert Einstein College <strong>of</strong> Medicine, Bronx, NY<br />

The pathogenesis <strong>of</strong> type 1 diabetes (T1D) in NOD mice is<br />

mediated by numerical and functional deficiencies in both<br />

CD4+CD25+FoxP3+ regulatory T cells (Tregs) and invariant<br />

natural killer T (iNKT) cells. Previously, we reported that<br />

protection <strong>of</strong> NOD mice from T1D can be achieved by<br />

activation <strong>of</strong> iNKT cells upon treatment with a multi-dose agalactosylceramide<br />

(a-GalCer) protocol that promotes preferential<br />

IL-4 secretion by iNKT cells. Since activated Tregs<br />

and iNKT cells are both self-reactive and protect NOD mice<br />

from T1D, we investigated whether iNKT-Treg collaboration<br />

is required for this protection. We recently reported that<br />

while a-GalCer therapy does not alter the proliferation or<br />

regulatory activity <strong>of</strong> Tregs, Treg activity is required for a-<br />

GalCer induced protection from T1D. This requirement <strong>of</strong><br />

Treg/iNKT collaboration for iNKT-mediated protection from<br />

T1D may not apply for the therapy <strong>of</strong> T1D provided by certain<br />

analogs <strong>of</strong> a-GalCer. Of particular interest is C20:2, an a-<br />

GalCer analog with a shortened (C20) acyl chain and two<br />

unsaturated bonds. When compared to a-GalCer, C20:2 yields<br />

significantly reduced IFN-g and enhanced IL-4 secretion by<br />

iNKT cells. Interestingly, we found that Treg activity is not<br />

required for C20:2 mediated protection from T1D by iNKT<br />

cells. Thus, our current results indicate that a-GalCer and<br />

C20:2 may differ in their requirement <strong>of</strong> Tregs for iNKT<br />

mediated protection from T1D. Ongoing studies will address<br />

the mechanism(s) that underlies this differential requirement<br />

<strong>of</strong> Tregs for protection from T1D.<br />

doi:10.1016/j.clim.2007.03.236<br />

F.24 Absence <strong>of</strong> Beta Cells in Long-Term Type 1a<br />

Diabetic Patients<br />

Travis Still, Pr<strong>of</strong>essional Research Assistant, Barbara Davis<br />

Center, Aurora, CO, Sally Kent, Instructor, Brigham and<br />

Women’s Hospital, Boston, MA, Alberto Pugliese, Diabetes<br />

Research Institute, University <strong>of</strong> Miami, Miami, FL, Piero<br />

Marchetti, Full Pr<strong>of</strong>essor, Ospedale di Cisanello, Pisa, Italy,<br />

Francesco Dotta, Full Pr<strong>of</strong>essor, University <strong>of</strong> Siena, Siena,<br />

Bernhard Hering, Full Pr<strong>of</strong>essor, Diabetes Institute for<br />

<strong>Immunology</strong> and Transplantation, Department <strong>of</strong> Surgery,<br />

University <strong>of</strong> Minnesota, Minneapolis, MN, Norma Kenyon,<br />

Full Pr<strong>of</strong>essor, Diabetes Research Institute, Cell Transplant<br />

Center, University <strong>of</strong> Miami, Miami, FL, Camillo Ricordi,<br />

Full Pr<strong>of</strong>essor, Diabetes Research Institute, Cell Transplant<br />

Center, University <strong>of</strong> Miami, FL, Miami, FL, Roberto<br />

Gianani, Assistant Pr<strong>of</strong>essor, Barbara Davis Center For<br />

Childhood Diabetes, Aurora, CT<br />

A recent report indicates that residual beta cells can be<br />

identified in the pancreas <strong>of</strong> long-term diabetic subjects by<br />

insulin immunostaining and in a subset <strong>of</strong> individuals by<br />

detection <strong>of</strong> C-peptide. We have analyzed pancreata from 20<br />

normal controls and 4 long-term diabetic subjects (1.5, 5, 22<br />

and 30 years post diagnosis) by immunohistochemistry for<br />

both insulin and the beta cell specific marker VMAT-2. We<br />

confirmed the beta cell specificity <strong>of</strong> VMAT-2 by triple<br />

immunostaining with glucagon, somatostatin and VMAT-2 <strong>of</strong><br />

normal human pancreas.<br />

In the pancreata from long-term diabetic subjects we did<br />

not find any VMAT-2 or insulin positive cells. In contrast<br />

abundant insulin (as expected) and VMAT-2 positive cells<br />

were present in all 20 normal human pancreata, in one new<br />

onset type 1a and in a type 2 diabetic patient. There was<br />

significant heterogeneity <strong>of</strong> staining intensity for VMAT-2<br />

amongst the normal control pancreata suggesting individual<br />

variability in VMAT-2 islet expression. Furthermore, there<br />

was also variability in VMAT-2 expression between different<br />

areas within the same pancreas with a zonal distribution <strong>of</strong><br />

strongly stained islets. These data indicate that in subjects<br />

with long-term type 1A diabetes, there are no residual beta<br />

cells by immunohistochemistry. Further studies <strong>of</strong> a larger<br />

series <strong>of</strong> patients are needed to establish whether beta cells<br />

continue to be present in type 1a diabetic subjects. In


Abstracts<br />

particular, it will be important to study pancreata from longterm<br />

diabetic subjects with documented C-peptide secretion<br />

(not available for these subjects).<br />

doi:10.1016/j.clim.2007.03.237<br />

F.25 Regulation <strong>of</strong> Pathogenicity Through CD40<br />

Expressed on CD4 T Cells<br />

Rocky Baker, Postdoctoral Fellow, Department <strong>of</strong><br />

<strong>Immunology</strong>, University <strong>of</strong> Colorado Health Sciences Center,<br />

Denver, CO, David Wagner, Assistant Pr<strong>of</strong>essor, Department<br />

<strong>of</strong> <strong>Immunology</strong>, University <strong>of</strong> Colorado Health Sciences<br />

Center, Denver, CO, Kathryn Haskins, Pr<strong>of</strong>essor, NJMRC,<br />

Denver, CO<br />

We have reported previously that the costimulatory<br />

molecule CD40 is a marker <strong>of</strong> autoreactive T cells found on<br />

diabetogenic T cell clones and that CD40+ T cells from the<br />

NOD mouse transfer diabetes. Although there is strong<br />

evidence that CD40 is functional on T cells, CD40 expression<br />

on T cells remains controversial because <strong>of</strong> typically low and<br />

variable levels <strong>of</strong> CD40 on primary T cells. We have<br />

investigated the conditions under which T cells express the<br />

CD40 molecule and how CD40 expression contributes to<br />

pathogenicity <strong>of</strong> autoreactive T cells. First, we have<br />

demonstrated that CD40 levels on primary T cells can be<br />

induced on 90% <strong>of</strong> CD4+CD40− T cells from NOD mice upon<br />

activation. Furthermore, comparing naive and primed T cells<br />

from different TCR-Tg NOD mouse models, we have found<br />

that the ability to express CD40 depends on the activation<br />

status <strong>of</strong> the T cell. Second, we investigated how CD40<br />

costimulation influences the effector function <strong>of</strong> CD4 T cells<br />

by comparing CD28 and CD40 costimulation in NOD mice. We<br />

have found that CD40 engagement can replace or synergize<br />

with CD28 costimulation in terms <strong>of</strong> T cell activation and<br />

proliferation. However, there is a major difference when<br />

costimulation occurs through CD40: our results indicate that<br />

CD40 costimulation influences the Th1/Th2 balance differently<br />

and that CD40 engagement on autoreactive T cells<br />

enhances a Th1 effector phenotype. Taken together, the data<br />

indicate that CD40 is an effective alternative pathway to<br />

CD28 costimulation and might regulate the pathogenicity <strong>of</strong><br />

CD4 T cells in T1D.<br />

doi:10.1016/j.clim.2007.03.238<br />

F.26 TRECS and Telomerase Analysis <strong>of</strong> Lymphocytes<br />

from Autoimmune Thyroid Disease Patients Point to<br />

the Migration <strong>of</strong> Recent Thymic Emigrants to the<br />

Thyroid Gland at the First Stages <strong>of</strong> Disease<br />

Ricardo Pujol Borrell, Pr<strong>of</strong>essor, Blood and Tissue Bank<br />

(LIRAD), Health Science Research Institut, Badalona, Marco<br />

Antonio Fernandez, Head Cytometry Facility, Health Science<br />

Research Institut, Badalona, Maria Pilar Armengol,<br />

Posdoctoral Fellow, Blood and Tissue Bank (LIRAD), Health<br />

Science Research Institut, Badalona, Manel Juan, Head HLA<br />

Laboratory, Blood and Tissue Bank (LIRAD), Health Science<br />

Research Institut, Badalona, Lidia Sabater, Research Fellow,<br />

Blood and Tissue Bank (LIRAD), Health Science Research<br />

Institut, Badalona, Ricardo Pujol Borrell, Head <strong>Immunology</strong><br />

Laboratory, Blood and Tissue Bank (LIRAD), Health Science<br />

Research Institut, Badalona<br />

According to the threshold hypothesis <strong>of</strong> autoimmunity,<br />

the abundance and affinity <strong>of</strong> autoreactive T cells in the<br />

repertoire determine the probability for the triggering <strong>of</strong><br />

autoimmune diseases. We have investigated how recent<br />

thymic emigrants may contribute the autoreactive repertoire<br />

in autoimmune thyroid disease (AITD) patients, by measuring<br />

TRECs in PBLs from AITD patients (n=50) and controls (n=55)<br />

and in intrathyroidal lymphocytes from autoimmune thyroid<br />

glands. TRECS were higher in AITD patients, especially in the<br />

N45-year-old patients. The analysis <strong>of</strong> TRECS in intrathyroidal<br />

lymphocytes (ITL) (n=45) in relation to disease duration<br />

revealed a distinct pattern <strong>of</strong> distribution: TRECS were higher<br />

in ITL than in PBL in b30-month-disease-duration patients<br />

(fourfold excess in average), and vice-versa N30-month<br />

patients (tenfold excess). Interestingly there was a correlation<br />

between the levels <strong>of</strong> TRECS in ITLs and PBLs in both<br />

groups but <strong>of</strong> different sign and intensity: it was stronger in<br />

the N30 months (r=0.94) than in the b30 months (r =0.535).<br />

This may indicate that while in the N30 months group the<br />

TRECs in ITLs is dependent on the TRECs in PBLs, this is not the<br />

case in the b30 months. The study <strong>of</strong> the telomere length did<br />

not reveal a relation between TRECs and telomere length in<br />

the T cells but was significantly shorter in ITLs cells than in<br />

PBLs. These results suggest that early in disease there is a<br />

migration to AITD glands <strong>of</strong> T lymphocytes newly generated in<br />

the thymus that then undergoes a local expansion.<br />

doi:10.1016/j.clim.2007.03.239<br />

S25<br />

F.27 Nramp1 Enhances Autoimmune Diabetogenic<br />

T Cell Response by Altering the Processing and<br />

Presentation <strong>of</strong> Pancreatic Islet Antigens in<br />

Dendritic Cells<br />

Yang Dai, Research Scientist, Torrey Pines Institute for<br />

Molecular Studies, San Diego, CA, Babak Shirdel, Student,<br />

Torrey Pines Institute for Molecular Studies, San Diego, CA,<br />

Sandra Chau, Student, Torrey Pines Institute for Molecular<br />

Studies, San Diego, CA, Linda Wicker, Pr<strong>of</strong>essor, Cambridge<br />

Institute for Medical Research, Cambridge, Philippe Gros,<br />

Pr<strong>of</strong>essor, McGill University, Montreal, QC, Canada, Eli<br />

Sercarz, Pr<strong>of</strong>essor, Torrey Pines Institute for Molecular<br />

Studies, San Diego, CA<br />

Dendritic cells are essential for initiating adaptive immune<br />

responses and establishing self tolerance. We hypothesize<br />

that the antigen processing machinery differs between<br />

diabetes-susceptible and -resistant, MHC-matched individuals,<br />

and that this difference contributes to the generation<br />

<strong>of</strong> pathogenic effectors rather than regulatory T cells in<br />

diabetes prone individuals. The Idd5.2 (insulin-dependent<br />

diabetes 5.2) region contributes to the genetic susceptibility<br />

<strong>of</strong> NOD (non-obese diabetes) mice to autoimmune diabetes<br />

and contains the gene encoding Nramp1 (natural resistanceassociated<br />

macrophage protein 1). We found that Nramp1<br />

expression is not restricted to macrophages; importantly,


S26 Abstracts<br />

dendritic cells (DC) can upregulate Nramp1 expression<br />

dramatically after stimulation. Nramp1-expressing DC exhibit<br />

an accelerated pH change (acidification) within endocytotic<br />

vesicles. Interestingly, when Nramp1 function is present, both<br />

bone marrow-derived DC and adherent splenic DC are more<br />

potent in stimulating autoreactive Tcells following processing<br />

<strong>of</strong> a diabetes-associated antigen, GAD (glutamic acid decarboxylase)<br />

65. Furthermore, an islet-infiltrating Tcell clone,<br />

BDC2.5, reacts much more vigorously to the islets when<br />

Nramp1-functional DC are used as antigen presenting cells.<br />

These data suggest that Nramp1 may alter the processing <strong>of</strong><br />

self antigens in antigen processing compartments to increase<br />

the availability <strong>of</strong> determinants involved in inducing pathogenic<br />

T cells (supported by grants to EES from Juvenile<br />

Diabetes Research Foundation and Diabetes National<br />

Research Group).<br />

doi:10.1016/j.clim.2007.03.241<br />

F.28 Perturbations in the Function and Composition<br />

<strong>of</strong> the B Cell Compartment are Present in Type 1<br />

Diabetes<br />

Mary Rieck, Research Technician, Benaroya Research<br />

Institute, Seattle, WA, Adrian Arechiga, Research Associate,<br />

Benaroya Research Institute, Seattle, WA, Catherine<br />

Pihoker, Associate Pr<strong>of</strong>essor, Department <strong>of</strong> Pediatrics,<br />

Seattle, WA, Jane Buckner, Associate Member, Benaroya<br />

Research Institute, Seattle, WA, Carla Greenbaum, Member,<br />

Benaroya Research Institute, Seattle, WA<br />

Disturbances in B cell populations have been identified in<br />

autoimmune diseases where autoantibodies are present. A<br />

role for B cells in the pathogenesis <strong>of</strong> type 1 diabetes (T1D) is<br />

suggested by studies in animal models and by the autoantibodies<br />

present in T1D. To address whether alterations in<br />

the B cell compartment are present in individuals with T1D we<br />

examined PBMC from healthy, T1D, and type 2 diabetic (T2D)<br />

subjects. The relative frequency <strong>of</strong> the naïve, memory and<br />

plasmablast subsets were significantly different between<br />

healthy and T1D subjects, while total B cells were no<br />

different. The plasmablast (CD19+CD27high) population was<br />

increased in T1D subjects compared to controls (pb0.006),<br />

and was most pronounced in subjects within 2 years <strong>of</strong><br />

diagnosis. The memory B cell (CD19+CD27+) subset was<br />

decreased in diabetic subjects (p =0.0006). The B cell<br />

populations in T2D subjects were comparable to healthy<br />

controls; however, first degree relatives <strong>of</strong> T1D subjects, like<br />

individuals with T1D, displayed a decrease in memory B cells<br />

(p=0.02). Thus the decrease in memory B cells is not due to<br />

the metabolic abnormalities associated with T1D but may be<br />

genetically influenced. In addition, we have identified a<br />

defect in BCR signaling within memory B cells <strong>of</strong> T1D subjects.<br />

This suggests that a B cell intrinsic mechanism may contribute<br />

to alterations in the B cell compartment. We hypothesize that<br />

this alteration <strong>of</strong> B cell signal transduction may contribute to<br />

pathogenesis <strong>of</strong> T1D by tipping the balance <strong>of</strong> long-term B cell<br />

memory toward high affinity antibody production.<br />

doi:10.1016/j.clim.2007.03.242<br />

F.30 Tracking Putative Pathogenic Members <strong>of</strong> the<br />

T Cell Repertoire in NOD Mice<br />

Idania Marrero, Postdoctoral Fellow, Torrey Pines Institute<br />

for Molecular Studies, San Diego, CA, Yang Dai, Postdoctoral<br />

Fellow, Torrey Pines Institute for Molecular Studies, San<br />

Diego, CA, Eli Sercarz, Pr<strong>of</strong>essor, Torrey Pines Institute for<br />

Molecular Studies, San Diego, CA<br />

To define the major components <strong>of</strong> the autoreactive Tcell<br />

repertoire that arise spontaneously in the diabetic NOD<br />

mouse, we used CDR3 length spectroscopy to (1) study the<br />

entire Tcell repertoire in pancreatic lymph nodes (PLN) from<br />

pre-diabetic NOD mice and (2) to identify potentially<br />

pathogenic clones in islet-infiltrating cells that persist after<br />

cyclophosphamide (Cyp) treatment <strong>of</strong> female NOD mice,<br />

which is designed to destroy regulatory T cells. Any public or<br />

quasi-public T cell expansions were identified in PLN <strong>of</strong><br />

female NOD mice at 4 and 10 weeks <strong>of</strong> age. We identified two<br />

expansions, Vb5.2/Jb2.5 and Vb10/Jb1.5, which were found<br />

in 60% and 50% <strong>of</strong> the 4-week-old female NOD mice. These<br />

expansions can be useful in the study <strong>of</strong> how the T cell<br />

repertoire can be modified after islet transplantation. After<br />

Cyp treatment, the Vb4, Vb5.2, Vb10 and Vb12 families<br />

showed dominant expansions. Interestingly, we found a<br />

unique Vb4-“195 nucleotide” peak that was found in 10 out<br />

<strong>of</strong> 11 (91%) Cyp-treated mice. In contrast, this peak was not<br />

detected in untreated NOD mice. Another interesting result<br />

was seen in the Vb12 family, in which a Vb12-“200” peak was<br />

prominent in 6 out <strong>of</strong> 10 (60%) Cyp-treated mice. These<br />

expansions may represent the more pathogenic clones.<br />

Currently, we are sequencing these expansions to establish<br />

whether the Cyp-induced expansions represent truly unique<br />

clones. We believe that these Cyp experiments will provide<br />

the possibility <strong>of</strong> distinguishing between regulatory and aggressive<br />

clones. This work was supported by JDRF 3-2005-336.<br />

doi:10.1016/j.clim.2007.03.243<br />

F.31 Reduced CD4+ T Cell-specific Gene Expression<br />

in Human Type 1 Diabetes Mellitus<br />

Tihamer Orban, Researcher, Joslin Diabetes Center, Boston,<br />

MA, Janos Kis, Diabetologist, Department <strong>of</strong> Internal<br />

Medicine and Gastroenterology, Polyclinic <strong>of</strong> the Hospitaller<br />

Brothers, Budapest, Laszlo Szereday, PhD, Reproductive and<br />

Tumor <strong>Immunology</strong> Research Group Hungarian Academy <strong>of</strong><br />

Science, Pecs, Klara Farkas, Joslin Diabetes Center, Boston,<br />

MA, Peter Engelmann, PhD, Department <strong>of</strong> <strong>Immunology</strong> and<br />

Biotechnology, Faculty <strong>of</strong> Medicine, University <strong>of</strong> Pecs,<br />

Pecs, Heyam Jalahej, Joslin Diabetes Center, Boston, MA,<br />

Andras Treszl, Pediatrician, SOTE First Department <strong>of</strong><br />

Paediatrics, Budapest<br />

Type 1 diabetes mellitus (T1DM) in humans is characterized<br />

by the T cell dependent destruction <strong>of</strong> the insulin<br />

producing pancreatic beta cells; however, the precise<br />

pathogenesis <strong>of</strong> the disease, especially the initiation <strong>of</strong><br />

pathologic immune response, is still largely unknown. We<br />

hypothesized that the function <strong>of</strong> human CD4+ T cells is<br />

altered in T1DM and analyzed unstimulated human peripheral<br />

blood CD4+ T cell gene expression. We used a novel<br />

three-way comparison <strong>of</strong> DNA microarray data <strong>of</strong> CD4+ Tcells


Abstracts<br />

isolated from new onset T1DM, patients with long-term type<br />

2 diabetes (T2DM), and from healthy controls in order to<br />

eliminate any possible influence <strong>of</strong> glucose homeostasis on<br />

our findings. We analyzed the T1DM specific gene expression<br />

changes and their functional relevance to T1DM autoimmunity.<br />

Our genetic and functional data show that T1DM CD4+ T<br />

cells are down-regulated specifically affecting key immune<br />

functions and cell cycle. Histone deacetylase gene expression,<br />

a key regulator <strong>of</strong> epigenetic modification, is also<br />

reduced. The CD4+ T cells showed impaired function,<br />

including an abnormal immune response, which may be a<br />

key element that leads to the breakdown <strong>of</strong> self-tolerance.<br />

doi:10.1016/j.clim.2007.03.244<br />

F.32 Protective Versus aggressive T Cell Responses<br />

to the Major Proinsulin73−90 Epitope in Beta-Islet<br />

Cell Autoimmunity<br />

Ivana Durinovic-Bello, Staff Scientist, Visiting Associate<br />

Pr<strong>of</strong>essor, Benaroya Research Institute, Virginia Mason<br />

Research Center, Seattle, WA, Nicol Pratt, Research<br />

Technician, Benaroya Research Institute, Virginia Mason<br />

Research Center, Seattle, WA, Susan A. Masewicz, Research<br />

Technician, Benaroya Research Institute, Virginia Mason<br />

Research Center, Seattle, WA, Gerald Nepom, Director,<br />

Virginia Mason Research Center, Benaroya Research<br />

Institute, Seattle, WA<br />

Proinsulin (P-Ins)73–90 is a major T cell epitope <strong>of</strong><br />

DRB1*0401 subjects with beta-islet cell autoimmunity. Subset<br />

<strong>of</strong> P-Ins73–90 specific T cells <strong>of</strong> the recent onset type 1<br />

diabetic (T1D) expressed Foxp3, a marker for regulatory Tcell<br />

lineages, secreted high IL-5 and IL-10 and low levels <strong>of</strong> IFNg,<br />

and predominantly used Tcell receptor Vb14 (Durinovic-Belló<br />

I et al. PNAS 103; 11,683, 2006). Addition <strong>of</strong> N- or C-terminal<br />

amino acids to P-Ins73–90 drastically changed their reactivity<br />

and cytokine phenotype, suggesting an immunomodulatory<br />

potential for this P-Ins epitope. To test the hypothesis that in<br />

subjects with beta-islet cell autoimmunity subsets <strong>of</strong> P-<br />

Ins73–90 specific T cells with potential down-regulatory<br />

phenotypes still exist, we have isolated P-Ins73–90 specific T<br />

cells <strong>of</strong> additional DRB1*0401 subjects (n=17) by depletion <strong>of</strong><br />

CD25+ Tcells, stimulation with P-Ins73–90 and IL-2, and Vb14<br />

and tetramer sorting. Both effector and down-regulatory<br />

cells show characteristics <strong>of</strong> activated T cells and express<br />

CD25 and OX40; in addition effector cells up-regulate CD154<br />

(CD40L), CD80/CD86, Fas (CD95) and FasL (CD178) and have<br />

high proliferative capacity, and down-regulatory cells<br />

express Foxp3 and suppress cognate and bystander responses.<br />

In T1D patients T cells secreting high levels <strong>of</strong> IFNg and IL-2<br />

predominate, whereas T cells <strong>of</strong> the control individuals and<br />

prediabetic subjects are characterized by high IL-4 and IL-5<br />

secretion and low IFNg and IL-10. Subsets <strong>of</strong> P-Ins73–90<br />

specific Tcells with down-regulatory phenotype may prove as<br />

a suitable targets for therapeutic intervention in DRB1*0401<br />

positive humans with beta-islet cell autoimmunity or recent<br />

onset T1D.<br />

doi:10.1016/j.clim.2007.03.245<br />

F.33 The Programmed Death-1 (pd-1) Pathway<br />

RegulatesPeripheralTCellToleranceDuring<br />

Autoimmune Diabetes in Nonobese Diabetic (NOD)<br />

Mice<br />

Brian T. Fife, Postdoctoral Fellow, University <strong>of</strong> California, San<br />

Francisco, San Francisco, CA, Indira Guleria, PhD,<br />

Transplantation Research Center, Brigham and Women’s<br />

Hospital and Children’s Hospital, Boston, MA, Melanie Bupp,<br />

PhD, University <strong>of</strong> California, San Francisco Diabetes Center,<br />

San Francisco, CA, Qizhi Tang, PhD, University <strong>of</strong> California, San<br />

Francisco Diabetes Center, San Francisco, CA, Todd Eagar, PhD,<br />

University <strong>of</strong> California, San Francisco Diabetes Center, San<br />

Francisco, CA, Helene Bour-Jordan, PhD, University <strong>of</strong><br />

California, San Francisco Diabetes Center, San Francisco, CA,<br />

Hideo Yagita, Pr<strong>of</strong>essor, Department <strong>of</strong> <strong>Immunology</strong>, Juntendo<br />

University School <strong>of</strong> Medicine, Tokyo, Japan, Miyuki Azuma,<br />

Pr<strong>of</strong>essor, Department <strong>of</strong> Molecular <strong>Immunology</strong>, Tokyo Medical<br />

and Dental University, Tokyo, Japan, Mohamed H. Sayegh,<br />

Pr<strong>of</strong>essor, Transplantation Research Center, Brigham and<br />

Women’s Hospital and Children’s Hospital, Boston, MA,<br />

Jeffrey Bluestone, Pr<strong>of</strong>essor, University <strong>of</strong> California,<br />

San Francisco Diabetes Center, San Francisco, CA<br />

Programmed death-1 (PD-1), an inhibitory costimulatory<br />

molecule on activated T cells, down-regulates immune<br />

responses. We investigated the role <strong>of</strong> this pathway in<br />

peripheral tolerance in the NOD mouse model <strong>of</strong> autoimmune<br />

diabetes. In this study, tolerance was induced using<br />

either antigen-coupled ethylene carbodiimide-fixed splenocytes<br />

or FcR non-binding anti-CD3 mAb administration.<br />

Antigen-coupled splenocyte treatment conferred complete<br />

and long lasting protection from diabetes transfer using islet<br />

reactive BDC2.5 TCR transgenic T cells. In addition, insulincoupled<br />

splenocytes were used to treat new onset spontaneously<br />

diabetic mice. Using this approach we demonstrate<br />

that disease remission is associated with inhibition <strong>of</strong><br />

pathogenic T cell proliferation, decreased cytokine production,<br />

and T cell anergy induction. Moreover, we show that<br />

robust long-term tolerance depends on the PD-1/PD-L1<br />

pathway in both systems. Anti-PD-1 and PD-L1, but not anti-<br />

PD-L2, reversed tolerance weeks after tolerogenic therapy<br />

by promoting antigen-specific T cell proliferation and<br />

inflammatory cytokines directly in infiltrated tissues. PD-<br />

1/PD-L1 blockade did not limit Treg activity suggesting<br />

direct effects on pathogenic T cells. Finally, we describe a<br />

critical role for PD-1/PD-L1 in another powerful immunotherapy<br />

model using anti-CD3, currently being tested in<br />

clinical trials. Anti-CD3 treatment <strong>of</strong> new onset spontaneously<br />

diabetic NOD mice results in disease remission. PD-<br />

1/PD-L1 blockade reverses tolerance and rapidly precipitates<br />

diabetes. These findings suggest that PD-1/PD-L1<br />

interactions form a common pathway to selectively maintain<br />

tolerance directly in the target tissues. Taken together,<br />

these results provide important insights in the regulation <strong>of</strong><br />

autoimmune responses and may lead to novel immunotherapeutic<br />

approaches for peripheral tolerance and treatment<br />

<strong>of</strong> autoimmune diabetes. Supported by NIH AI35297 and<br />

JDRF 10-2006-799.<br />

doi:10.1016/j.clim.2007.03.248<br />

S27


S28 Abstracts<br />

F.34 Gleevec: A New Therapeutic for Type I<br />

Diabetes?<br />

Cedric Louvet, Postdoctoral Research Fellow, Diabetes<br />

Center at University <strong>of</strong> California, San Francisco, San<br />

Francisco, CA, Gregory L. Szot, Islet Transplant Facility<br />

Manager, Diabetes Center, San Francisco, CA, Jiena Lang,<br />

Staff Research Associate, Diabetes Center at University <strong>of</strong><br />

California, San Francisco, San Francisco, CA, Jeffrey A.<br />

Bluestone, Diabetes Center Director, Diabetes Center at<br />

University <strong>of</strong> California, San Francisco, San Francisco, CA,<br />

Michael R. Lee, Staff Research Assistant, Diabetes Center at<br />

University <strong>of</strong> California, San Francisco, San Francisco, CA<br />

Gleevec (Imatinib, formerly STI571) is the first FDA-approved<br />

drug to directly turn <strong>of</strong>f a specific protein known to cause a<br />

cancer. Although targeted to the Bcr-Abl kinase, Gleevec has<br />

been shown to modestly affect related kinases including PDGF,<br />

c-fms and c-kit potentially involved in various cell types critical<br />

to the development and progression <strong>of</strong> autoimmune diseases.<br />

Based on these fundamental properties, we embarked on a<br />

study to determine the clinical effects <strong>of</strong> Gleevec in T1D in NOD<br />

mice.<br />

NOD mice were treated by gavage daily with 1.5 mg/<br />

mouse <strong>of</strong> commercially available Gleevec suspended in<br />

peanut oil. Pre-diabetic mice treated with Gleevec, but not<br />

with peanut oil only, were protected from spontaneous<br />

diabetes. More importantly, when Gleevec treatment was<br />

initiated at the time <strong>of</strong> disease onset (blood sugars between<br />

250 and 350 mg/dl), the majority <strong>of</strong> mice went into remission<br />

and b10% relapsed during the course <strong>of</strong> the therapy. When<br />

therapy was discontinued at 2 weeks, the majority <strong>of</strong> the<br />

mice became hyperglycemic within 1–2 weeks. However,<br />

longer therapy (8–10 weeks) resulted in long-term normoglycemia<br />

in a majority <strong>of</strong> treated animals even after drug<br />

cessation suggesting that Gleevec does not have to be given<br />

continuously to promote long-lasting protection. The possibility<br />

that short-term therapy with Gleevec can lead to longterm<br />

effects makes it a very attractive potential therapeutic<br />

for the treatment <strong>of</strong> patients with new onset T1D as well as<br />

potentially patients undergoing an islet transplant.<br />

doi:10.1016/j.clim.2007.03.249<br />

F.38 Characterization <strong>of</strong> Peptide Agonists and<br />

Antagonists for the Interaction <strong>of</strong> CD200 with<br />

CD200R1 and Their Efficacy in Modulation <strong>of</strong><br />

LPS-induced TNFα Production and Mortality In Vivo<br />

Reginald Gorczynski, Pr<strong>of</strong>essor, University Health Network,<br />

Toronto, ON, Canada, Ivo Boudakov, PDF, University Health<br />

Network, Toronto, ON, Canada, Ismat Khatri, PDF,<br />

University Health Network, Toronto, ON, Canada<br />

After engagement <strong>of</strong> the Ig-supergene family members<br />

CD200R1 and CD200, immunosuppressive and/or antiinflammatory<br />

signals augment allograft survival and<br />

decrease production <strong>of</strong> inflammatory cytokines. The primary<br />

domains <strong>of</strong> both CD200 and CD200R1 implicated in<br />

these interactions are found in the NH2-terminal region <strong>of</strong><br />

each molecule. We investigated the capacity <strong>of</strong> 10–15 mer<br />

synthetic peptides defining the three complementaritydetermining<br />

(CDR1–3) regions <strong>of</strong> mouse CD200 to reproduce<br />

the function <strong>of</strong> full-length CD200, or antagonize that<br />

function, assaying the suppression <strong>of</strong> LPS-induced TNFα<br />

production (in vivo/in); suppression <strong>of</strong> MLC responses in<br />

vitro; and LPS-induced mortality (d7) in vivo. Peptides<br />

located in the CDR2 region, but not CDR1 or CDR3 were<br />

potent agonists in vitro and in vivo, recapitulating the<br />

effects seen with the full-length molecule (CD200Fc). These<br />

small molecular weight agonists <strong>of</strong> CD200 might have<br />

efficacy in vivo in models <strong>of</strong> sepsis (and TNFα production).<br />

In a further analysis we explored the ability <strong>of</strong> the same<br />

peptides to block the suppressive and anti-inflammatory<br />

effect <strong>of</strong> the full-length CD200Fc which was seen in MLCs<br />

and after LPS administration both in vitro and in vivo. In<br />

these studies peptides located in the CDR1 and CDR3<br />

regions <strong>of</strong> the NH2-terminal <strong>of</strong> CD200 were found to be<br />

antagonists <strong>of</strong> the biological effects <strong>of</strong> CD200. Our data<br />

confirm that discrete small molecular weight agonists and<br />

antagonists can be defined for the interactions <strong>of</strong> CD200<br />

and the CD200R1 receptor. Since these interactions result in<br />

regulation <strong>of</strong> both acquired immune responses, and<br />

inflammatory responses, these peptides may have clinical<br />

utility.<br />

doi:10.1016/j.clim.2007.03.250<br />

F.39 Calculating the Number <strong>of</strong> Ordered Mutations<br />

Necessary to Cause a Specific Cancer in a Specific<br />

Risk Group and the Fraction <strong>of</strong> the Group that is<br />

Immune to the Cancer<br />

Ivan Kramer, Associate Pr<strong>of</strong>essor, UMBC, Canada Physics,<br />

Catonsville, MD<br />

The series <strong>of</strong> ordered mutations that cause a cell to<br />

become cancerous is modeled so that the fraction <strong>of</strong> a risk<br />

group (e.g., white men) that has developed a specific cancer<br />

(e.g., melanoma) at any age can be computed. The saturated<br />

model constructed here allows the possibility that a fraction<br />

<strong>of</strong> a risk group may be immune to developing a specific<br />

cancer but is otherwise isomorphic to the physical model<br />

describing an ordered chain <strong>of</strong> radioactive nuclear decays.<br />

The model’s cancer incidence function depends on only<br />

three independent parameters: the number <strong>of</strong> mutations<br />

necessary for a cell to become cancerous, the fraction <strong>of</strong> the<br />

group that is immune to developing the cancer, and the time<br />

required for 50% <strong>of</strong> the group to experience an average<br />

mutation (defined as the mutation half-life). These three<br />

parameters are determined by fitting the model’s cancer<br />

incidence function to the cancer incidence data. For<br />

example, the modeling predicts that all white males in the<br />

USA are vulnerable to developing melanoma, five ordered<br />

mutations are required to develop it, and the mutation halflife<br />

is 33.5 years. By contrast, the modeling also predicts that<br />

80.7% <strong>of</strong> white females in the USA are immune to developing<br />

melanoma, three ordered mutations are required to develop<br />

it, and the mutation half-life is 54.7 years. Thus, different<br />

risk groups can develop the same cancer through different<br />

pathways. The mechanism underlying female immunity to<br />

melanoma should become a research priority. Stimulating


Abstracts<br />

the immune system to eliminate mutated, pre-cancerous<br />

cells would prevent cancer.<br />

doi:10.1016/j.clim.2007.03.255<br />

F.41 The State <strong>of</strong> Immune System in the Pregnants<br />

with Untreated Syphilis<br />

Tatiana Yaremtchuk, Doucent, Lviv National Medical<br />

University, Department <strong>of</strong> Obstetrics, Gynecology and<br />

Perinatology <strong>of</strong> PGE, Lviv, Ukraine, Olga Korchynska,<br />

Assistant, Department <strong>of</strong> Obstetrics, Gynecology and<br />

Perinatology <strong>of</strong> PGE <strong>of</strong> Lviv National Medical University,<br />

Lviv, Ukraine, Yaroslav Korinets, Doctor, Scientific<br />

Collaborator, Department <strong>of</strong> Antenatal Diagnostics and<br />

Perinatology <strong>of</strong> Institute <strong>of</strong> Hereditary Pathology, Lviv,<br />

Ukraine, Natalia Prokoptchuk, Genetics and USG Diagnostics<br />

Doctor, Department <strong>of</strong> Antenatal Diagnostics and<br />

Perinatology <strong>of</strong> Institute <strong>of</strong> Hereditary Pathology, Lviv,<br />

Ukraine, Angela Misiura, Assistant, Department <strong>of</strong><br />

Obstetrics, Gynecology and Perinatology <strong>of</strong> PGE <strong>of</strong> Lviv<br />

National Medical University, Lviv, Ukraine<br />

Objective <strong>of</strong> Study: The state <strong>of</strong> immune system in the<br />

pregnants with untreated syphilis. Materials: Vein blood <strong>of</strong> 5<br />

pregnants with untreated early latent syphilis within 26-33<br />

weeks and 30 healthy pregnants in the same terms. Methods:<br />

Designation <strong>of</strong> quantity <strong>of</strong> CD3, CD4, CD8, CD16, CD56, CD19,<br />

CD4/CD8 was carried out with test-systems <strong>of</strong> monoclonal<br />

antibodies. The concentrations <strong>of</strong> IgA, IgM, IgG were<br />

designated by method <strong>of</strong> radial immune diffusion. The level<br />

<strong>of</strong> circulating immune complexes was investigated by<br />

spectrophotometral method. Results: Initial investigations<br />

showed the quantity <strong>of</strong> CD3 composed 59.4 % in comparison<br />

with 62,53+/–1,23 % in control group (P>0,05). The level <strong>of</strong><br />

CD4 trustworthy was lower – 16,2 % contra 35,6+/–0,86 %<br />

(Pb0,01). The quantity <strong>of</strong> CD8, CD16, CD56 correspondingly<br />

had tendency to reduce. These indicators accordingly made<br />

up – 25,0 % in comparison with 27,6+/–1,72 % (P>0.05), 3,0 %<br />

contra 6,51+/–0,88 % (P>0,05), 12,2 % in comparison with<br />

14,68+/–1,71 % (P>0,05). The immuneregulative index was<br />

0,65+/–0,22 in comparison 1,29+/–0,06 (Pb0,05). The concentration<br />

CD19 was trustworthy lower 5,2 % contra 12,83+/–<br />

0,88 % (Pb0,05). The levels <strong>of</strong> IgA and IgG were accordingly<br />

1,56+/–0,2 g/l in comparison with 1,98+/–0,06 g/l (Pb0,05)<br />

and 10,29+/–0,54 g/l contra 12,38+/–0,43 g/l (Pb0,05). The<br />

concentration <strong>of</strong> IgM had tendency to decrease – 1,37+/–0,32<br />

g/l contra 1,76+/–0,11 g/l (P>0,05). The level <strong>of</strong> circulating<br />

immune complexes also was trustworthy lower 59,8+/–11,4<br />

ODU contra 93,5+/–3,58 ODU (Pb0,01). Conclusions: The<br />

suppression <strong>of</strong> immune system has been found out in the<br />

pregnants with untreated early latent syphilis.<br />

doi:10.1016/j.clim.2007.03.256<br />

F.44 Human Norovirus Infection: Genetic and<br />

Immune Determinants <strong>of</strong> Susceptibility and<br />

Resistance<br />

Juan Leon, Postdoctoral Fellow, Emory University, Atlanta,<br />

GA, Lisa Lindesmith, Laboratory Technician, Department <strong>of</strong><br />

Epidemiology, University <strong>of</strong> North Carolina at Chapel Hill,<br />

Chapel Hill, NC, Erin-Joi McNeal, Laboratory Technician,<br />

Department <strong>of</strong> Global Health, Rollins School <strong>of</strong> Public<br />

Health, Atlanta, GA, Ralph Baric, Pr<strong>of</strong>essor, Department <strong>of</strong><br />

Epidemiology, University <strong>of</strong> North Carolina at Chapel Hill,<br />

Chapel Hill, NC, Elizabeth Ailes, Graduate Student,<br />

Department <strong>of</strong> Epidemiology, Emory University, Atlanta,<br />

GA, Christine Moe, Associate Pr<strong>of</strong>essor, Department <strong>of</strong><br />

Global Health, Rollins School <strong>of</strong> Public Health, Atlanta, GA<br />

Infection with Norovirus (NoV) is the main cause <strong>of</strong><br />

epidemic worldwide. NoV are classified by the CDC and NIAID<br />

as Bioterrorism Category B Priority Pathogens based on their<br />

high transmissibility, low infectious dose, and serious<br />

economic impact. Little is known on what factors protect<br />

individuals from NoV infection. To meet this need, our goal is<br />

to identify genetic and immunologic determinants <strong>of</strong><br />

protection from NoV infection. Because animal models are<br />

not available and these viruses do not replicate in culture,<br />

we developed expertise in human challenge models. We<br />

challenged 109 human volunteers with varying doses <strong>of</strong> the<br />

Norwalk virus (NV) strain <strong>of</strong> NoV. We found that gender,<br />

ethnicity, blood group, levels <strong>of</strong> pre-challenge serum NVspecific<br />

IgG or levels <strong>of</strong> pre-challenge saliva NV-specific IgA<br />

were not significant predictors <strong>of</strong> infection. Presence <strong>of</strong><br />

serum NV-specific IgG and presence <strong>of</strong> a putative NV-binding<br />

receptor, H type 1, were significantly associated with<br />

infection. Dose <strong>of</strong> inoculum received and age <strong>of</strong> the<br />

individual may also be predictors <strong>of</strong> infection. We observed<br />

a significant correlation in levels <strong>of</strong> salivary NV-specific IgG<br />

with salivary NV-specific IgA and serum NV-specific IgG.<br />

Lastly, in genetically susceptible individuals, infected individuals<br />

exhibited a rise in NV-specific serum and salivary<br />

antibodies while uninfected individuals did not. These<br />

results suggest that only infected individuals develop<br />

activation <strong>of</strong> the humoral response. These data will be<br />

used to better understand the immune response to NV<br />

infection and design effective vaccines against NoV to<br />

protect high risk populations including children, food<br />

handlers, and the elderly.<br />

doi:10.1016/j.clim.2007.03.257<br />

F.45 Polymorphism <strong>of</strong> IL-6 gene and Its Association<br />

with Susceptibility to Brucellosis<br />

Manoochehr Rasouli, <strong>Immunology</strong> Department, <strong>Clinical</strong><br />

Microbiology Research Center-Shiraz University <strong>of</strong> Medical<br />

Sciences, Shiraz, Simin Kiany, MSc, <strong>Immunology</strong> Department,<br />

<strong>Clinical</strong> Microbiology Research Center-Shiraz University <strong>of</strong><br />

Medical Sciences, Shiraz<br />

S29<br />

Background: Brucellosis is a highly contagious bacterial<br />

zoonosis that affects millions <strong>of</strong> people worldwide. Polymorphonuclear<br />

cells (PMNs) are the most abundant type <strong>of</strong><br />

circulating white blood cells (WBCs) and are the major cell<br />

type mediating acute inflammation response to bacterial<br />

infection. Since IL-6 plays a major role in inflammation and<br />

stimulates production <strong>of</strong> neutrophils from BM progenitors,<br />

this supports the involvement <strong>of</strong> IL-6 in the process and<br />

pathogenesis <strong>of</strong> brucellosis. It has been shown that the


S30 Abstracts<br />

production level <strong>of</strong> IL-6 has been correlated with allele<br />

inherited in individuals. Therefore, the aim <strong>of</strong> current study<br />

was to assess whether IL-6 (−174) polymorphism was<br />

associated with susceptibility to brucellosis. Method: One<br />

hundred and seventy-five patients with brucellosis and 77<br />

healthy animal husbandmen who had infected animals and<br />

consumed their dairy products, joined this study. DNAs<br />

extracted from samples were genotyped for IL-6 (−174 G/C)<br />

polymorphism using AS-PCR method. Results: Our results<br />

showed that IL-6 (−174) gene polymorphism was distributed<br />

similarly in the patient and control groups (P =0.3).<br />

Discussion: Although we found no association between IL-6<br />

(−174) gene polymorphism and susceptibility to brucellosis<br />

we cannot exclude that other polymorphisms within this<br />

gene or its receptor may donate susceptibility to brucellosis.<br />

Moreover these findings need confirmation in other<br />

population groups.<br />

doi:10.1016/j.clim.2007.03.259<br />

F.46 Interleukin-1β(+3953) Gene Polymorphism and<br />

Susceptibility to Helicobacter Pylori Associated<br />

Gastritis<br />

Ayda Hosseinkhani, Pharmacist, <strong>Clinical</strong> Microbiology<br />

Research Center, Shiraz University <strong>of</strong> Medical Sciences,<br />

Shiraz, Simin Kiany, MSc, <strong>Clinical</strong> Microbiology Research<br />

Center, Shiraz University <strong>of</strong> Medical Sciences, Shiraz,<br />

Manoochehr Rasouli, PhD, <strong>Clinical</strong> Microbiology Research<br />

Center, Shiraz University <strong>of</strong> Medical Sciences, Shiraz, Akram<br />

Jamshidzadeh, PhD, Faculty <strong>of</strong> Pharmacy-Shiraz University<br />

<strong>of</strong> Medical Sciences, Shiraz, Shohreh Farshad, PhD, <strong>Clinical</strong><br />

Microbiology Research Center, Shiraz University <strong>of</strong> Medical<br />

Sciences, Shiraz<br />

Introduction: It has been speculated that IL-1 genes<br />

play a crucial role in the genetic predisposition to gastric<br />

problems upon H. pylori infection by modulating the host<br />

immune response. Three biallelic polymorphisms have<br />

been reported in IL-1 β all representing CNT base<br />

transition at position −511, −31 and +3953 from the<br />

transcriptional site. In the present study we attempted to<br />

determine the IL-1 β (+3953) risk genotypes to H. pylori<br />

mediated gastritis. Methods: The subjects were 373<br />

individuals with no gastrointestinal problems and 49 H.<br />

pylori positive patients suffering from gastritis which was<br />

confirmed on the basis <strong>of</strong> endoscopic findings. All subjects<br />

were genotyped for IL-1 β+3953 gene polymorphism using<br />

polymerase chain reaction-restriction fragment length<br />

polymorphism (PCR-RFLP). Results: There was no significant<br />

difference in the frequency <strong>of</strong> IL-1 β +3953 C/T<br />

genotypes between two groups.<br />

Discussion: The lack <strong>of</strong> significant difference in the<br />

frequency <strong>of</strong> IL-1 β+3953 C/T between the studied groups<br />

might be the result <strong>of</strong> our small patient population. Thus we<br />

suggest that this study be performed on larger group <strong>of</strong><br />

patients; also investigation <strong>of</strong> other IL-1 functional gene<br />

polymorphism on Iranian population is recommended.<br />

doi:10.1016/j.clim.2007.03.260<br />

F.48 The −251 AA Genotype <strong>of</strong> the Interleukin-8<br />

Promoter is Associated with Susceptibility to<br />

Brucellosis<br />

Manoochehr Rasouli, PhD, <strong>Immunology</strong> Department, <strong>Clinical</strong><br />

Microbiology Research Center, Shiraz University <strong>of</strong> Medical<br />

Sciences, Shiraz, Simin Kiany, MSc, <strong>Immunology</strong> Department,<br />

<strong>Clinical</strong> Microbiology Research Center, Shiraz University <strong>of</strong><br />

Medical Sciences, Shiraz<br />

Background: Interleukin-8 (IL-8), a member <strong>of</strong> CXC chemokine<br />

family, is a chemoattractant <strong>of</strong> neutrophils and lymphocytes.<br />

IL-8 promoter is estimated to be 1500 bp. Several reports<br />

have shown relationship between IL-8 gene polymorphism and<br />

several human diseases such as bronchial asthma, Parkinson’s<br />

disease and Helicobacter pylori infection. The aim <strong>of</strong> this study<br />

was to evaluate the relationship between IL-8 (−251 A/T)<br />

polymorphism and brucellosis. Methods: One hundred and<br />

eighty eight patients with brucellosis and 77 healthy farmers<br />

who consumed contaminated row milk and dairy products <strong>of</strong><br />

animals with brucellosis were included in this study. All<br />

individuals were genotyped for IL-8 (−251 A/T) promoter<br />

polymorphism using polymerase chain reaction-restriction fragment<br />

length polymorphism (PCR-RFLP). Results: The frequency<br />

<strong>of</strong> AA genotype was significantly higher in patient group than<br />

healthy individuals (14.3% vs. 2.5%, P=0.005). Discussion: As<br />

data revealed that the frequency <strong>of</strong> AA genotype was lower in<br />

healthy controls than patients. Hence, our study provides<br />

evidence that the presence <strong>of</strong> AA genotype is significantly<br />

associated with susceptibility to brucellosis.<br />

doi:10.1016/j.clim.2007.03.261<br />

F.49 Interferon Gamma Producing T Lymphocytes in<br />

Bacterial Bi<strong>of</strong>ilm Infection: Response Modifiers for<br />

Polymorphonuclear Neutrophils (PMN)<br />

Christ<strong>of</strong> Wagner, Associate Pr<strong>of</strong>essor, University <strong>of</strong><br />

Heidelberg, Heidelberg, Germany, Christ<strong>of</strong> Iking-Konert,<br />

Assisant Pr<strong>of</strong>essor, University <strong>of</strong> Duesseldorf, Duesseldorf,<br />

Germany, Andreas Wentzensen, Head <strong>of</strong> Clinic, Klinik fur<br />

Unfall-und Weiderherstellungschirurgie, Ludwigshafen,<br />

Germany, Gertrud Maria Haensch, Full Pr<strong>of</strong>essor, University<br />

<strong>of</strong> Heidelberg Department <strong>of</strong> <strong>Immunology</strong>, Heidelberg,<br />

Germany, Volkmar Heppert, Head <strong>of</strong> Department, Septic<br />

Traumatology, Ludwigshafen, Germany<br />

Bacterial infection with bi<strong>of</strong>ilm formation on orthopaedic<br />

implants causes a progressive, destructive inflammatory<br />

process, causing massive tissue destruction, bone resorption,<br />

and loosening <strong>of</strong> the implant. In most cases, this so-called<br />

implant-associated posttraumatic osteomyelitis requires the<br />

removal <strong>of</strong> the implant, allowing recovery and analysis <strong>of</strong> the<br />

local cellular infiltrate. By cyt<strong>of</strong>luorometry, the majority (60–<br />

85%) <strong>of</strong> the infiltrated cells were identified as highly activated<br />

polymorphonuclear neutrophils (PMN); the next largest cell<br />

population (5–35%) were T-lymphocytes. CD4 and CD8 positive T<br />

cells were found, the latter prevailing compared to the<br />

peripheral blood. The majority <strong>of</strong> the CD8+ cells were CD28<br />

negative; about 26% expressed CD11b, 76% CD57. The latter two<br />

receptors were co-expressed on less than 5% <strong>of</strong> the cells. The


Abstracts<br />

CD11b positive Tcells produced interferon (IFN) gamma. In vitro<br />

experiments provided a link between activated Tcells and PMN:<br />

IFN γ, supernatants <strong>of</strong> the Tcells, or co-incubation <strong>of</strong> PMN with<br />

activated T cells induced expression on PMN <strong>of</strong> CD64, the high<br />

affinity receptor for immunoglobulin, and <strong>of</strong> MHC class II<br />

antigens. The fact that IFN γ is among the few cytokines that<br />

can stimulate PMN to produce CD64 and MHC class II, and the<br />

observation that CD64 and MHC class II positive PMN is found in<br />

the cellular infiltrate suggest that CD8+ T cells via synthesis <strong>of</strong><br />

IFN γ modulatethefunction<strong>of</strong>infiltratedPMNandpointtoa<br />

novel, not yet appreciated role <strong>of</strong> Tcells in the defence against<br />

bacterial bi<strong>of</strong>ilm infections.<br />

doi:10.1016/j.clim.2007.03.262<br />

F.50 Server Hepatitis Related Gene mfgl2 shRNA<br />

Plasmid Construction and Its RNA Interference<br />

In Vitro<br />

Qin Ning, Chief Physician, Department <strong>of</strong> Infectious Disease,<br />

Tongji Hospital Huazhong University <strong>of</strong> Science and<br />

Technology, Wuhan, China, Zhimo Wang, Doctor in Charge,<br />

Department <strong>of</strong> Infectious Disease, Tongji Hospital, Huazhong<br />

University <strong>of</strong> Science and Technology, Wuhan, China, Dong Xi,<br />

Assistant Researcher, Department <strong>of</strong> Infectious Disease, Tongji<br />

Hospital, Huazhong University <strong>of</strong> Science and Technology,<br />

Wuhan, China, Chuanlong Zhu, Doctor in Charge, Department<br />

<strong>of</strong> Infectious Disease, Tongji Hospital, Huazhong University <strong>of</strong><br />

ScienceandTechnology,Wuhan,China,SuiGao,Postgraduate<br />

Student, Department <strong>of</strong> Infectious Disease, Tongji Hospital,<br />

Huazhong University <strong>of</strong> Science and Technology, Wuhan,<br />

China, Weiming Yan, Associate Researcher, Department <strong>of</strong><br />

Infectious Disease, Tongji Hospital, Huazhong University <strong>of</strong><br />

Science and Technology, Wuhan, China, Xiaoping Luo, Chief<br />

Physician, Department <strong>of</strong> Infectious Disease, Tongji Hospital,<br />

Huazhong University <strong>of</strong> Science and Technology, Wuhan, China<br />

Objective: To determine the viral and host factors involved<br />

in the transcription <strong>of</strong> hfgl2 gene which was demonstrated to<br />

play a pivotal role in human and experiment fulminant viral<br />

hepatitis. Methods: HBc, HBs or HBx expression plasmids were<br />

constructed and cotransfected with a hfgl2 luciferase report<br />

construct into eukaryotic cells. Results: Luciferase assay<br />

showed that HBc or HBx protein, but not HBs protein<br />

significantly enhanced hfgl2 transcription in both CHO and<br />

HepG2 cells. Series deletion assay <strong>of</strong> hfgl2 gene promoter<br />

demonstrated that a strong regulatory domain from ¨C 712 to<br />

−568 was responsible for hfgl2 gene transcription in response<br />

to HBc or HBx proteins. By site-directed mutagenesis, the<br />

overlapped cis-elements LEF/c-Ets in domain −712/−568 was<br />

demonstrated to play an important role in the regulation <strong>of</strong><br />

hfgl2 gene in response to HBc protein, while another two ciselements<br />

LEF/c-Ets and HSTF in the same domain were found<br />

to participate in hfgl2 transcription regulation in response to<br />

HBx protein. EMSA assays showed that HBc and HBx proteins<br />

did not directly induce hfgl2 gene activation, while an Ets<br />

family member c-Ets-2 bound to the cognate cis-element in<br />

hfgl2 promoter was responsible for induction <strong>of</strong> hfgl2<br />

activation in response to viral proteins. Conclusion: Our<br />

study provides new insights in understanding the pathology <strong>of</strong><br />

fulminant hepatic failure and the interaction between HBV<br />

virus and host gene expression. This work was supported by<br />

NSFC 30225040, 30571643, 30672380, National Key Basic<br />

Research Program <strong>of</strong> China (2005CB522901, 2005CB522507)<br />

and <strong>Clinical</strong> Subject Key Project from the Ministry <strong>of</strong> Health.<br />

doi:10.1016/j.clim.2007.03.263<br />

F.51 TLR2 and TLR4 Expression on PBMC and Ascites<br />

Fluid from Hepatic Cirrhotic Patients with SPB<br />

Carmen Contreras, MD, <strong>Immunology</strong> Universidad de<br />

Guadalajara, Guadalajara, Mexico, Jorge Segura,<br />

Gastroenterologist, Gastroenterology Hospital Civil,<br />

Guadalajara, Mexico, Cecilia Guillen, Pr<strong>of</strong>essor,<br />

<strong>Immunology</strong> Universidad de Guadalajara, Guadalajara,<br />

Mexico, Mary Fafutis, Immunologist Researcher,<br />

<strong>Immunology</strong>, Universidad de Guadalajara, Guadalajara,<br />

Mexico, Margarita Montoya, Immunologist, <strong>Immunology</strong><br />

Universidad de Guadalajara, Guadalajara, Mexico<br />

Justification: The cirrhosis <strong>of</strong> the liver is a pathology with a<br />

high morbid-mortality, being one <strong>of</strong> the main complication’s<br />

cause <strong>of</strong> spontaneous bacterial peritonitis (SBP) due to an<br />

alteration in the immunologic balance, observed like a<br />

deficiency in the bactericide action <strong>of</strong> serum, opsonins, and<br />

complement; an increase in the proinflammatory cytokines;<br />

changes in the endoplasmatic reticulum system; and a<br />

neutrophil’s functional alteration. It has been demonstrated<br />

that the toll-like receptors (TLR) are essentials for the<br />

pathogenic recognition that starts with the activation <strong>of</strong> NFkB<br />

a transcriptional factor necessary for the biosynthesis <strong>of</strong><br />

proinflammatory cytokines. At the moment the role played by<br />

these membrane receptors in the development <strong>of</strong> SBP is<br />

unknown. The aim <strong>of</strong> this work is to study the TLR2 and 4<br />

expression on PBMC and ascites fluid <strong>of</strong> cirrhotic patients (CP)<br />

with or without an SPB development. Methodology: Venous<br />

peripheral blood and ascites fluid was obtained from CP with<br />

or without SPB, as well as venous peripheral blood from<br />

healthy individuals (HI). The expression <strong>of</strong> TLR2, TLR4 and<br />

CD14 by flow cytometer was measured. Results: The<br />

peripheral blood’s cells from HI show a higher expression <strong>of</strong><br />

TLR2 and TLR4 than CP and SPB, however the differences<br />

were not significant. The TLR2 and TLR4 expression in<br />

patient’s cells with SPB was higher than the cirrhotic patients<br />

(pb0.05) in ascites fluid. Conclusion: In this work it was found<br />

that the TLR2 and TLR4 expression was higher in SPB than<br />

cirrhotic patients in ascites fluid.<br />

doi:10.1016/j.clim.2007.03.264<br />

S31<br />

F.52 Persistently Deficient In Vitro<br />

Anti-Mycobacterial Immunity Despite Highly<br />

Successful Long-Term (>4 years) HAART:<br />

Differential Impact <strong>of</strong> Latent Tuberculosis Infection<br />

and Previous Active Disease<br />

Gil Benard, Medical Researcher, Medical School <strong>of</strong> the<br />

University <strong>of</strong> São Paulo, São Paulo, Brazil, Marcelo<br />

Mendonça, Laboratory <strong>of</strong> Dermatology and<br />

Immunodeficiencies, Medical School <strong>of</strong> the University <strong>of</strong>


S32 Abstracts<br />

São Paulo, São Paulo, Brazil, Léia C. Rodrigues Silva, PhD<br />

Student, Laboratory <strong>of</strong> Dermatology and<br />

Immunodeficiencies, Medical School <strong>of</strong> the University <strong>of</strong><br />

São Paulo, São Paulo, Brazil, Guilherme G. Silveira,<br />

Student, Laboratory <strong>of</strong> Dermatology and<br />

Immunodeficiencies, Medical School <strong>of</strong> the University <strong>of</strong><br />

São Paulo, São Paulo, Brazil, Maury M. Tanji, Laboratory <strong>of</strong><br />

Dermatology and Immunodeficiencies, Medical School <strong>of</strong><br />

the University <strong>of</strong> São Paulo, São Paulo, Brazil, Denise<br />

Schout, Epidemiology Service, Hospital das Clínicas,<br />

Medical School <strong>of</strong> the University <strong>of</strong> São Paulo, São Paulo,<br />

Brazil, Alberto J.S. Duarte, Pr<strong>of</strong>essor, Laboratory <strong>of</strong><br />

Dermatology and Immunodeficiencies, Medical School <strong>of</strong><br />

the University <strong>of</strong> São Paulo, São Paulo, Brazil<br />

Background: The degree Mycobacterium tuberculosis (Mtb)<br />

immune reconstitution provided by long-term (N4 years)<br />

successful HAART and whether a past tuberculosis (TB) history<br />

influences this reconstitution are not known. Methods: We<br />

compared the lymphocyte proliferative response (LPR) and IFNã<br />

secretion to phytohemagglutinin (PHA) and Mtb antigens<br />

(ESAT6, Ag85B and whole Mtb lysate [SMtb]) <strong>of</strong> immunereconstituted<br />

(CD4 N500 cells/ìl) HIV+ patients with a past history <strong>of</strong><br />

cured TB (HIV+ CTB group) or latent infection as presumed by<br />

positive purified protein derivative skin test (PPD+) (HIV+ PPD+<br />

group) with HIV− controls either cured from TB (CTB group) or<br />

PPD reactors (PPD+ group). The databank on TB incidence<br />

among HIV+ and HIV− patients at our services during 1999–2005<br />

was also analyzed. Results: Most HIV+ patients presented normal<br />

PHA responses. However, Mtb immune reconstitution was<br />

incomplete, especially to ESAT6 and Ag85B. SMtb reactivity<br />

was fully restored in the HIV+ CTB group, while in HIV+ PPD+<br />

patients it remained lower than in PPD+ controls. Comparison<br />

between the two HIV groups also suggested better immune<br />

reconstitution in HIV+ CTB patients. Databank analysis revealed<br />

that some HIV+ patients with N360 CD4 cells/ìl still developed<br />

TB, but not those with previous TB history. Conclusions: Mtb<br />

immune reconstitution is incomplete in HIV+ CTB and HIV+ PPD<br />

+ patients even after highly successful HAART. However,<br />

reactivity to whole mycobacteria antigens is restored,<br />

especially in HIV+ CTB patients, possibly due to survival <strong>of</strong><br />

higher numbers <strong>of</strong> mycobacteria-specific T cell clones throughout<br />

immunosuppression, probably allowing sufficient protection<br />

against new Mtb challenges.<br />

doi:10.1016/j.clim.2007.03.266<br />

F.53 Human CD8+ Cytotoxic T Lymphocyte Epitopes<br />

Identified from Herpes Simplex Virus Type 1<br />

Glycoprotein D<br />

Aziz Alami Chentoufi, Assistant Specialist, Cellular and<br />

Molecular <strong>Immunology</strong> Laboratory, The Eye Institute,<br />

University <strong>of</strong> California, Irvine, Orange, CA, Xiuli Zhang,<br />

Postdoctoral Fellow, Cellular and Molecular <strong>Immunology</strong><br />

Laboratory, The Eye Institute, University <strong>of</strong> California,<br />

Irvine, Orange, CA, Kasper Lamberth, Researcher, Institute<br />

<strong>of</strong> Medical Microbiology and <strong>Immunology</strong> (IMMI), University<br />

<strong>of</strong> Copenhagen, 2200 Copenhagen, Denmark, Xiaoming Zhu,<br />

Research Associate, Cellular and Molecular <strong>Immunology</strong><br />

Laboratory, The Eye Institute, University <strong>of</strong> California,<br />

Irvine, Orange, CA, Gargi Dasgupta, Research Associate,<br />

Cellular and Molecular <strong>Immunology</strong> Laboratory, The Eye<br />

Institute, University <strong>of</strong> California, Irvine, orange, CA,<br />

Michele Wu, Student, Cellular and Molecular <strong>Immunology</strong><br />

Laboratory, The Eye Institute, University <strong>of</strong> California,<br />

Irvine, Orange, CA, Alex Nguyen, Bio-199 Student, Cellular<br />

and Molecular <strong>Immunology</strong> Laboratory, The Eye Institute,<br />

University <strong>of</strong> California, Irvine, Orange, CA, Ilham Bettahi,<br />

Postdoctoral Fellow, Cellular and Molecular <strong>Immunology</strong><br />

Laboratory, The Eye Institute, University <strong>of</strong> California,<br />

Irvine, Orange, CA, Søren Buus, Pr<strong>of</strong>essor, Institute <strong>of</strong><br />

Medical Microbiology and <strong>Immunology</strong> (IMMI), University <strong>of</strong><br />

Copenhagen, 2200 Copenhagen, Denmark, Lbachir<br />

BenMohamed, Assistant Pr<strong>of</strong>essor, Cellular and Molecular<br />

<strong>Immunology</strong> Laboratory, The Eye Institute, University <strong>of</strong><br />

California, Irvine, Orange, CA<br />

Cytolystic T cells (CTLs) play a major role in herpes simplex<br />

virus type 1 and type 2 (HSV-1 and HSV-2) infections and<br />

diseases. Among some eleven HSV-1 and HSV-2 coat glycoproteins,<br />

glycoprotein D (gD) induces immunodominant T cells and<br />

is a leading vaccine candidate antigen. However, little is known<br />

about human CD8+ T cell epitopes <strong>of</strong> gD. Based on predictive<br />

computational algorithms and peptide binding affinity to HLA-<br />

A*0201 molecules, we have identified ten regions within the<br />

HSV-1 gD, each 9 to 10 amino acids in length, exhibiting high<br />

affinity CD8+ Tcell epitope(s). T2 cell stabilization assay showed<br />

peptide gD53-61, gD70-78 and gD278-286 significantly, and in<br />

dose-dependent manner, upregulates HLA-A*0201 molecules<br />

expression.Toobtainanobjectiveenumeration<strong>of</strong>CD8+Tcells<br />

induced by each gD peptide epitope, we employed the HLA-<br />

A*0201 tetramer staining procedure. A relatively high percentages<br />

<strong>of</strong> CD8+ Tcells positive for gD-18-10, gD53-61, gD70-78 and<br />

gD278-286/HLA-A*0201 tetramers were detected in PBMC from<br />

HLA-A*0201 seropositive patients, either directly ex vivo or<br />

following a single in vitro stimulation. Accordingly, strong HLA-<br />

A*0201-restricted CD8+ CTLs, assessed by INF-γ-ELISPOT and<br />

CD107a/b cytotoxic assays, were mapped to gD53-61, gD70-78<br />

and gD278-286 peptides. However, only gD53-61 and gD278-286<br />

peptides were able to generate CD8+ T cells that recognize<br />

infected target cells. Collectively, these findings suggest that<br />

high-affinity HLA-A*0201-binding gD53-61 and gD278-286 epitopes<br />

are naturally processed and are recognized by HSVspecific<br />

CD8+ CTLs in the infected human population, providing<br />

important information for the development <strong>of</strong> subunit vaccine<br />

strategies against herpes.<br />

doi:10.1016/j.clim.2007.03.267<br />

F.55 Protection Against SEB-Induced Toxic Shock by<br />

Anti-TCR Vb8 Antibody<br />

Manisha Singh, Postdoctoral Associate, Baylor College <strong>of</strong><br />

Medicine, <strong>Immunology</strong> Department, Houston, TX<br />

Intraperitoneal injection <strong>of</strong> the bacterial superantigen,<br />

Staphylococcus enterotoxin B (SEB) in HLA-DR3 transgenic<br />

mice leads to toxic shock and death within 72 h. In vivo,<br />

this is characterized by a marked expansion <strong>of</strong> splenic TCR<br />

Vb8+ CD4 and CD8 T cells compared to PBS treated mice. It


Abstracts<br />

appears that TCR Vb8-bearing CD4 and CD8 cells and the<br />

cytokines they secrete might be responsible for the<br />

pathogenesis <strong>of</strong> toxic shock caused by SEB. Therefore, we<br />

reasoned that depletion <strong>of</strong> TCR Vb8 bearing T cells by anti-<br />

Vb8 antibody might protect mice from SEB-induced shock.<br />

To test this hypothesis, we administered 200 μg <strong>of</strong> purified<br />

anti-TCR Vb8 antibody (clone 23.1) either prior to or 2 h<br />

after SEB injection. Irrespective <strong>of</strong> timing <strong>of</strong> anti-TCR Vb8<br />

administration, all anti-Vb8-treated mice (six <strong>of</strong> six)<br />

remained healthy while those injected with control antibody<br />

were either sick (n=2) or died (n=3). This salutary<br />

anti-TCR Vb8 effect correlated with markedly diminished<br />

SEB-induced serum levels <strong>of</strong> IL-6 and IL-2; however IFN-g<br />

and TNF release was not affected. Anti-TCR Vb8 Abs<br />

treatment also reduced infiltration in various organs like<br />

liver, lungs and kidney, those normally affected by SEB.<br />

These findings suggest that the pathogenesis <strong>of</strong> SEB induced<br />

toxic shock is primarily mediated by T cells and anti-TCR<br />

Vb8 antibody therapy may have therapeutic implications for<br />

SEB mediated shock.<br />

doi:10.1016/j.clim.2007.03.268<br />

F.56 Anti-Mouse GITR Monoclonal Antibody (Mab)<br />

Augments Humoral and Cellular Responses to H5N1<br />

and H3N2 Avian Influenza Hemagglutinin<br />

Joe Ponte, Senior Scientist, Tolerx, Inc, Cambridge, MA,<br />

Reema Gulati, Scientist, Tolerx, Cambridge, MA, Adam<br />

O’Shea, Scientist, Tolerx, Cambridge, MA, Paul Ponath,<br />

Immunologist, Tolerx, Cambridge, MA, Michael Paglia,<br />

Senior Manager, Tolerx, Cambridge, MA, Michael<br />

Rosenzweig, Senior Director, Tolerx, Cambridge, MA<br />

Adjuvants, including antibodies to tumor necrosis factor<br />

receptor superfamily (TNFRSF) members, augment immune<br />

responses to vaccines. One member <strong>of</strong> this family, glucocorticoid-induced<br />

tumor necrosis factor receptor (GITR), is<br />

expressed on regulatory and effector T cells. The aim <strong>of</strong> this<br />

study was to assess the ability <strong>of</strong> an anti-mouse GITR Mab,<br />

2F8, to stimulate murine humoral and cellular immunity to<br />

H3N2 and H5N1 viral hemagglutanins (HA). 2F8 enhanced<br />

anti-HA titers compared with controls and this enhancement<br />

was equal to or greater than titers obtained with incomplete<br />

Freund’s adjuvant or aluminum hydroxide. 2F8 F(ab(E))2<br />

fragments were also effective in boosting humoral immunity,<br />

suggesting that Fc interactions were not required. Moreover,<br />

the enhanced response was durable and specific for the<br />

antigen administered at the time <strong>of</strong> antibody treatment, and<br />

2F8-treated mice required less antigen to obtain titers<br />

equivalent to IFA-treated mice. 2F8 also enhanced cellular<br />

immunity as measured by EliSPOT. These results were<br />

recapitulated in cynomolgus monkeys using an anti-human<br />

GITR Mab (6C8). Thus, anti-GITR Mabs augment humoral and<br />

cellular immunity in mice and cynomolgus monkeys. Ongoing<br />

studies are evaluating the effect <strong>of</strong> anti-GITR Mabs on<br />

regulatory and effector T cells.<br />

doi:10.1016/j.clim.2007.03.269<br />

F.57 The B Cell Superantigen Peptostreprococcus<br />

Magnus Protein L Induces Lung Inflammation<br />

Through a MyD88-dependent Mechanism<br />

Amy Anderson, Research Associate, University <strong>of</strong> Pennsylvania<br />

School <strong>of</strong> Medicine, Philadelphia, PA, David LaRosa,<br />

Postdoctoral Fellow, University <strong>of</strong> Pennsylvania, Medicine,<br />

Philadelphia, PA, Arnold Levinson, Pr<strong>of</strong>essor <strong>of</strong> Medicine,<br />

University <strong>of</strong> Pennsylvania School <strong>of</strong> Medicine, Medicine,<br />

Philadelphia, PA<br />

Peptostreptococcus magnus protein L functions as a B cell<br />

superantigen by interacting with V kappa light chain<br />

determinants on many human and mouse immunoglobulins,<br />

irrespective <strong>of</strong> their antigen specificities or heavy chain<br />

isotype. We sought to determine if this property endowed<br />

protein L with the capacity to elicit lung inflammation.<br />

Following intratracheal (i.t.) administration <strong>of</strong> recombinant<br />

protein L or BSA to C57/BL6 mice, broncheoalveolar lavage<br />

fluid (BALF) was recovered at varying time points and<br />

analyzed for total neutrophil counts and concentrations <strong>of</strong><br />

MIP-2, KC and TNF. Lung tissue was harvested for histological<br />

examination. Protein L-treated mice accumulated neutrophils<br />

in their BALF as early as 4 h and showed an alveolar and<br />

peribronchial infiltrate <strong>of</strong> inflammatory cells peaking<br />

between 8 and 24 h. MIP-2, TNF, and KC levels in protein L<br />

challenged mice peaked at 4 h. These findings are consistent<br />

with those reported in conventional immune-complex models<br />

<strong>of</strong> lung inflammation. Studies were repeated in JHT mice to<br />

examine the importance <strong>of</strong> B cells/immunoglobulins in these<br />

reactions. Surprisingly, both the BALF response and lung<br />

histopathology were preserved. By contrast, both types <strong>of</strong><br />

reactions were abrogated in MyD88 knockout mice, which<br />

have defective toll-like receptor signaling. These results<br />

indicate that protein L-induced lung inflammation is not<br />

dependent on its immunoglobulin binding (B cell superantigenic)<br />

activity but does appear to rely on signaling<br />

through a toll-like receptor. Thus, the present study reveals a<br />

novel, unanticipated pro-inflammatory mechanism initiated<br />

by a microbial protein with known B cell superantigenic<br />

properties.<br />

doi:10.1016/j.clim.2007.03.272<br />

S33<br />

F.58 Regulation <strong>of</strong> the Epstein−Barr Virus<br />

LMP1-mediated NF-kappaB Signaling Pathway<br />

by a Novel Adaptor Protein STAP-2<br />

Osamu Ikeda, Undergraduate, Department <strong>of</strong> <strong>Immunology</strong>,<br />

Graduate School <strong>of</strong> Pharmaceutical Sciences Hokkaido<br />

University, Sapporo, Japan, Yuichi Sekine, Assistant,<br />

Department <strong>of</strong> <strong>Immunology</strong>, Graduate School <strong>of</strong> Pharmaceutical<br />

Sciences Hokkaido University, Sapporo, Japan, Teruhito Yasui,<br />

Assistant, Department <strong>of</strong> Molecular <strong>Immunology</strong>, Research<br />

Institute for Microbial Diseases, Osaka University, Japan, Suita<br />

Kenji Sugiyma, PhD, Nippon Boehringer Ingelheim Co. Ltd.<br />

Kawanishi Pharma Research Institute, Kawanishi, Japan, Kenji<br />

Oritani, Assistant, Department <strong>of</strong> Hematology and Oncology,<br />

Graduate School <strong>of</strong> Medicine, Osaka University, Japan, Suita<br />

Akihiko Yoshimura, Pr<strong>of</strong>essor, Division <strong>of</strong> Molecular and Cellular<br />

<strong>Immunology</strong>, Medical Institute <strong>of</strong> Bioregulation, Kyushu


S34 Abstracts<br />

University, Maidashi, Japan, Tadashi Matsuda, Pr<strong>of</strong>essor,<br />

Department <strong>of</strong> <strong>Immunology</strong>, Graduate School <strong>of</strong> Pharmaceutical<br />

Sciences Hokkaido University, Sapporo, Japan<br />

Signal transducing adaptor protein-2 (STAP-2) is a recently<br />

identified adaptor protein, which contains pleckstrin homology<br />

(PH) and Src homology 2 (SH2)-like domains as well as a prolinerich<br />

domain in its C-terminal region. Our previous studies have<br />

demonstrated that STAP-2 binds to MyD88 and IKK-α/β, and<br />

modulates NF-kappaB signaling in macrophages. In this study,<br />

STAP-2 was induced by expression <strong>of</strong> the Epstein–Barr virus LMP1<br />

in B cells, while overexpression <strong>of</strong> STAP-2 inhibited LMP1mediated<br />

NF-kappaB signaling. Furthermore, STAP-2 associated<br />

with LMP1 in EBV-positive B cells. These results strongly suggest<br />

that STAP-2 acts as an endogenous negative inhibitor <strong>of</strong> the EBV<br />

LMP1-mediated signaling, which is critical for the EBV persistent<br />

infection and EBV-associated pathogenesis.<br />

doi:10.1016/j.clim.2007.03.273<br />

F.61 Defensins Block Bacterial Toxin-induced<br />

Interleukin-1β Production<br />

Jishu Shi, Assistant Pr<strong>of</strong>essor, Anatomy, Physiology and<br />

Pharmacology, Auburn, AL, Shelly Ao, Research Assistant,<br />

APP, Auburn, AL, Charalabos Pathoulakis, Pr<strong>of</strong>essor,<br />

Gasterenterology, Boston, MA<br />

This study was designed to test the hypothesis that<br />

defensins can block the release <strong>of</strong> IL-1β induced by<br />

Clostridium difficile toxin A (TxA) and Staphylococcus<br />

aureus α-toxin (a-Tox). LPS-activated (20 ng/ml, 2 h)<br />

human monocytes were treated with 3 nM TxA in the<br />

presence or absence <strong>of</strong> human neutrophil defensin HNP-1<br />

(25–50 μg/ml) or human Paneth cell defensin HD-5 (25–50<br />

μg/ml) for 6 h. In the absence <strong>of</strong> defensins, TxAstimulated<br />

monocytes released a large amount <strong>of</strong> mature<br />

IL-1β (17 kDa) into the extracellular milieu. The release<br />

<strong>of</strong> mature IL-1β from TxA-stimulated monocytes was<br />

blocked by HNP-1 and HD-5 in a dose-dependent manner.<br />

Different from ATP, TxA-induced IL-1β release was not<br />

inhibited by KN-62, a P2X7 receptor antagonist. In<br />

contrast to TxA, a-Tox (250 ng/ml) induced the release<br />

<strong>of</strong> both proIL-1β (31 kDa) and mature IL-1β from LPSactivated<br />

monocytes. In the presence <strong>of</strong> 20 μg/ml <strong>of</strong><br />

HNP-1 or HD-5, the amount <strong>of</strong> mature IL-1β released from<br />

a-Tox-treated monocytes was reduced by 70%, while a-<br />

Tox-induced release <strong>of</strong> proIL-1β was completely blocked.<br />

Similar to TxA, a-Tox-induced IL-1β release was not<br />

inhibited by KN-62. Furthermore, the release <strong>of</strong> mature<br />

IL-1β was caspase-1-dependent for both toxins. These<br />

data suggest that TxA- and a-Tox-induced IL-1β release is<br />

independent <strong>of</strong> the ATP/P2X7 receptor pathway, and that<br />

defensins can block bacterial toxin-induced posttranslational<br />

processing and release <strong>of</strong> IL-1β. In addition to their<br />

antimicrobial activity, defensins may play an important<br />

role in host inflammatory response to bacterial infection<br />

by controlling the production <strong>of</strong> IL-1β.<br />

doi:10.1016/j.clim.2007.03.274<br />

F.62 Lack <strong>of</strong> Association <strong>of</strong> Interleukin-8 Gene<br />

Polymorphism with Helicobacter Pylori-induced<br />

Gastritis in Iranian Patients<br />

Ayda Hosseinkhani, Pharmacist, <strong>Clinical</strong> Microbiology<br />

Research Center, Shiraz University <strong>of</strong> Medical Sciences,<br />

Shiraz, Iran, Farshad Shohreh, Microbiologist, <strong>Clinical</strong><br />

Microbiology Research Center, Shiraz University <strong>of</strong> Medical<br />

Sciences, Shiraz, Iran, Akram Jamshidzadeh, PhD, Faculty <strong>of</strong><br />

Pharmacy-Shiraz University <strong>of</strong> Medical Sciences, Shiraz,<br />

Iran, Manoochehr Rasouli, PhD, <strong>Clinical</strong> Microbiology<br />

Research Center, Shiraz University <strong>of</strong> Medical Sciences,<br />

Shiraz, Iran, Simin Kiany, MSc, <strong>Clinical</strong> Microbiology<br />

Research Center, Shiraz University <strong>of</strong> Medical Sciences,<br />

Shiraz, Iran<br />

Background: Helicobacter pylori is a major cause <strong>of</strong><br />

neoplastic and inflammatory gastroduodenal diseases <strong>of</strong> the<br />

stomach. H. pylori infection and associated gastric diseases are<br />

common in developing countries. Cytokine gene polymorphisms<br />

and H. pylori have been linked to gastric disease. We determined<br />

the role <strong>of</strong> host interleukin-8 (−251 A/T) gene polymorphism in<br />

the development <strong>of</strong> gastritis in south Iranian population.<br />

Methods: We genotyped IL-8 (−251 A/T) gene polymorphism in<br />

54 H. pylori infected individuals with gastritis and 337 infected<br />

individuals with normal mucosa using polymerase chain reaction-restriction<br />

fragment length polymorphism (PCR-RFLP). The<br />

diagnosis <strong>of</strong> gastritis was established on the basis <strong>of</strong> endoscopic<br />

findings.<br />

Results: We did not find any significant differences in<br />

allele and genotype frequencies <strong>of</strong> IL-8 (−251A/T) among our<br />

study groups. Discussion: Our analysis did not reveal a<br />

significant difference between the frequencies <strong>of</strong> IL-8<br />

(−251) genotypes and alleles which might be the result <strong>of</strong><br />

our limited number <strong>of</strong> patient population. We suggest this<br />

study be continued on larger population <strong>of</strong> the patients. Also,<br />

analysis <strong>of</strong> polymorphism in other positions <strong>of</strong> IL-8 gene is<br />

recommended.<br />

doi:10.1016/j.clim.2007.03.275<br />

F.63 The Impact <strong>of</strong> HLA Polymorphisms on<br />

Measles Virus-specific T-Cell Memory Responses<br />

in Vaccinated Subjects<br />

Inna Ovsyannikova, Senior Research Associate, Mayo Clinic,<br />

Mayo Vaccine Research Group, Rochester, MN, Jenna Ryan,<br />

Senior Research Technologist, Mayo Clinic, Mayo Vaccine<br />

Research Group, Rochester, MN, Norman Pinsky, Senior<br />

Research Technologist, Mayo Clinic, Mayo Vaccine Research<br />

Group, Rochester, MN, Robert Jacobson, Pr<strong>of</strong>essor <strong>of</strong><br />

Pediatrics, Mayo Clinic, Department <strong>of</strong> Pediatric and<br />

Adolescent Medicine, Rochester, MN, Robert Vierkant,<br />

Senior Statistician, Mayo Clinic, Division <strong>of</strong> Biostatistics,<br />

Rochester, MN, Gregory Poland, Pr<strong>of</strong>essor <strong>of</strong> Medicine, Mayo<br />

Clinic, Department <strong>of</strong> Internal Medicine, Rochester, MN<br />

The humoral and cellular immune responses to measles virus<br />

(MV) are genetically influenced. We hypothesized that the<br />

antigen-presenting function <strong>of</strong> HLA molecules could play a role<br />

in the variability in measles-specific T cell memory responses in


Abstracts<br />

vaccinated subjects. We studied the association between IFN-g<br />

producing T cells specific for MV and the alleles <strong>of</strong> HLA genes<br />

among 336 children (12−18 years <strong>of</strong> age) who previously<br />

received two doses <strong>of</strong> the measles vaccine. PBMCs (2x100,000<br />

cells/well; HLA typed) were cultured with Edmonston strain <strong>of</strong><br />

measles (moi 0.5) and spot-forming cells (SFC) were analyzed by<br />

ELISPOT. Linear-regression analysis was used to study allelic<br />

associations. Measles-induced IFN-g responses (median 6 SFC;<br />

interquartile range 2, 17 SFC per 2x100,000 T cells) were<br />

detected in subjects up to 12 years after vaccination. Univariate<br />

analyses identified that class I HLA-A*1101 (median 6 SFC,<br />

p=0.04) and B*3701 (median 34 SFC, p=0.07) alleles were<br />

suggestive <strong>of</strong> an association with higher memory IFN-g<br />

responses, while the Cw*0802 (median SFC 3, p=0.08) allele<br />

wassuggestive<strong>of</strong>anassociationwithadecreaseintherelative<br />

frequency <strong>of</strong> IFN-g producing T cells. Class II HLA alleles with<br />

potential associations with increased measles-specific T cell<br />

responses included DRB1*1303 (median SFC 17, p=0.01),<br />

DPB1*0301 (median SFC 11, p=0.02) and DPB1*1501 (median<br />

SFC 10, p=0.07). A suggestive association was observed between<br />

DQB1*0303 (median SFC 4, p=0.07) alleles and decreased<br />

frequency <strong>of</strong> MV memory IFN-g response. Thus, the presence<br />

or absence <strong>of</strong> certain HLA alleles may influence long-term<br />

measles immunity and the dynamics <strong>of</strong> virus-specific T cell<br />

memory outcome in vaccinated subjects.<br />

doi:10.1016/j.clim.2007.03.276<br />

F.64 Loss <strong>of</strong> Interferon Gamma Receptor Alpha<br />

(IFNGR α) in Peripheral Blood Mononuclear Cells<br />

from Patients with Chronic Hepatitis C<br />

Lucia Cristina Jamli Abel, Pr<strong>of</strong>essor, Albert Einstein<br />

Research Institute-Albert Einstein Hospital, São Paulo,<br />

Brazil, Mário Pessoa, Albert Einstein Research<br />

Institute-IEP-Albert Einstein Hospital, São Paulo, Brazil,<br />

Hoel Sette Jr., Albert Einstein Research Institute-IEP-<br />

Albert Einstein Hospital, São Paulo, Brazil, Carlos<br />

Moreira-Filho, Albert Einstein Research Institute-IEP-<br />

Albert Einstein Hospital, São Paulo, Brazil, Ben-Hur Ferraz<br />

Neto, Albert Einstein Research Institute-IEP-Albert Einstein<br />

Hospital, São Paulo, Brazil, Luciana Marti, Albert Einstein<br />

Research Institute-IEP-Albert Einstein Hospital, São Paulo,<br />

Brazil, Denise Rodrigues, Infectious Diseases Division,<br />

Federal University <strong>of</strong> São Paulo, São Paulo SP, Brazil, Esper<br />

Kallas, Pr<strong>of</strong>essor, Infectious Diseases Division, Federal<br />

University <strong>of</strong> São Paulo, São Paulo, Brazil, Venancio Alves,<br />

Pr<strong>of</strong>essor, Department <strong>of</strong> Pathology, University <strong>of</strong> São<br />

Paulo School <strong>of</strong> Medicine, São Paulo, Brazil<br />

The mechanisms underlying the pathogenesis <strong>of</strong> chronic liver<br />

disease due to HCV are not fully understood. IFN-γ plays an<br />

important role in viral clearance, where IFN-γ either exogenous<br />

or endogenously produced by virus-specific CD8+ Tcells inhibits<br />

the replication <strong>of</strong> HCV. The alpha chain receptor (IFNGR α) isa<br />

high affinity receptor that binds to IFN-γ, therefore its<br />

expression can be important in the control <strong>of</strong> HCV infection.<br />

Using flow cytometric analysis, we found that the number <strong>of</strong><br />

peripheral lymphocytes expressing alpha chain receptor (IFNGR<br />

α) in patients with chronic hepatitis C was lower than the normal<br />

controls (N) (p=0.018) and significantly decreased in patients<br />

with abnormal ALT (p=0.05). In chronic patients the expression<br />

<strong>of</strong> IFNGR α was inversely correlated to viral load (r=−0.362,<br />

p=0.038). In the liver, 50% <strong>of</strong> T cell lines obtained from biopsy<br />

isolated from patients with end stage liver disease expressed<br />

IFNGR α and when stimulated with staphylococcal enterotoxin B<br />

(SEB)producedhighlevels<strong>of</strong>IFN-γ. No significant difference<br />

was found for IFN-γ production by peripheral blood mononuclear<br />

cells in the presence <strong>of</strong> PHA between chronic HCV patients and<br />

normal controls, but the IFN-γ production was weaker in viremic<br />

chronic hepatitis C patients than in subjects who are able to<br />

clear the virus (p=0.033). The results suggest that HCV infection<br />

may down-regulate the number <strong>of</strong> lymphocytes expressing<br />

IFNGR α in the peripheral blood but not in liver and that the<br />

intrahepatic response contributes at least in part to the control<br />

<strong>of</strong> disease.<br />

doi:10.1016/j.clim.2007.03.277<br />

S35<br />

F.65 Normal Adult Ranges and Diversity <strong>of</strong> Serotype<br />

Specific Anti-Pneumococcal Antibodies (SSAPAbs)<br />

after Immunisation with 23-Valent Polysaccharide<br />

Vaccine<br />

Robert Hallam, Senior Biomedical Scientist, Papworth Hospital/<br />

<strong>Immunology</strong> Department, Cambridgeshire, England, Paul<br />

Balmer, <strong>Clinical</strong> Scientist, HPA, Vaccine Evaluation Department,<br />

Manchester, England, Ray Borrow, <strong>Clinical</strong> Scientist, HPA-<br />

Vaccine Evaluation Department, Manchester, England, Diana<br />

Bilton, Consultant Respiratory Physician, Papworth Hospital,<br />

Respiratory Infection, Inflammation and <strong>Immunology</strong> (RIII)<br />

Department, Cambridgeshire, England, Charles Haworth,<br />

Consultant Respiratory Physician, Papworth Hospital,<br />

Respiratory Infection, Inflammation and <strong>Immunology</strong> (RIII)<br />

Department, Cambridgeshire, England, Andrew Exley,<br />

Consultant Immunologist, Papworth Hospital/<strong>Immunology</strong><br />

Department, Cambridge<br />

Background: <strong>Clinical</strong> and experimental studies identify<br />

SSAPAbs as key elements <strong>of</strong> pneumococcal immunity reflecting<br />

interactions with antigen specific and pathogen recognition<br />

receptors on APCs, B and T cells. New assays for SSAPAbs<br />

incorporating multiplex bead arrays with CPS and 22F<br />

absorption enable analysis <strong>of</strong> large data sets to determine<br />

normal adult ranges and diversity <strong>of</strong> responses. Earlier<br />

studies indicating diversity <strong>of</strong> responses and potential for<br />

immunologic refractoriness caution against standard<br />

approaches in healthy volunteers. Method: We investigated<br />

pneumococcal immunity in 250 adults with recurrent lower<br />

respiratory tract infections, measuring serum SSAPAbs pre<br />

and 4–6 weeks post-immunization with 23-valent polysaccharide<br />

vaccine. We tested the hypothesis that cases include<br />

a mixture <strong>of</strong> normal and low responders to standard<br />

immunization, on a background <strong>of</strong> natural exposure to<br />

antigen through carriage and infection. SSAPAb levels pre,<br />

post, and post/pre were log transformed and examined for<br />

goodness <strong>of</strong> fit using mixture modeling, with censoring <strong>of</strong><br />

outlying data according to post-immunization SSAPAb levels.<br />

We then investigated for hierarchy and diversity <strong>of</strong> SSAPAb<br />

levels after immunization. Results: Mixture modeling indicated<br />

serotype specific normal and low adult ranges.<br />

Confidence intervals were resolved to determine threshold


S36 Abstracts<br />

values for antibody levels post-immunization. Serotypes 14,<br />

9V, and 4 appear high to low in hierarchy <strong>of</strong> responses.<br />

Discussion: Third generation multiplex assays for SSAPAb<br />

levels, without cross-reacting antibodies as confounders,<br />

indicate normal ranges after immunization are serotype<br />

specific with evidence <strong>of</strong> hierarchy and diversity. These data<br />

are consistent with serotype specific and non-specific pathways<br />

in antibody responses to pneumococcal capsular<br />

polysaccharides.<br />

doi:10.1016/j.clim.2007.03.278<br />

F.66 Differential Cytokine Response Induced by<br />

M. Avium and M. Abscessus in Human Macrophages<br />

is Mediated Through p38 MapKinase Signalling<br />

Pathway and Partially Dependent on TLR2<br />

Activation<br />

Elizabeth Sampaio, Senior Visiting Investigator, NIAID,<br />

NIH, Bethesda, MD, Houda Elloumi, Postdoctoral Fellow,<br />

NIH/NIAID, Bethesda, MD, Adrian Zelazny, Staff Scientist,<br />

NIH/NIAID, Bethesda, MD, Yvonne Shea, Laboratory<br />

Supervisor, NIAID/NIH, Bethesda, MD, Li Ding, Lab<br />

Manager, NIAID/NIH, Bethesda, MD, Steven Holland, Lab<br />

Chief, NIH/NIAID, Bethesda, MD<br />

Non-tuberculous mycobacteria (NTM) are ubiquitous<br />

environmental organisms that can cause chronic lung<br />

infection associated with primary or acquired immune<br />

deficiencies or in otherwise apparently normal individuals.<br />

Among NTM, M. avium is the most common cause <strong>of</strong><br />

mycobacterial infection. Moreover, M. abscessus is an<br />

emerging pulmonary pathogen responsible for the majority<br />

<strong>of</strong> infections by rapidly growing mycobacteria. It has been<br />

shown that the ability <strong>of</strong> mycobacteria to induce TNFα<br />

secretion is inversely related to their virulence. In this<br />

work, cytokine response and signaling pathways triggered<br />

by reference and clinical isolates <strong>of</strong> M. abscessus and M.<br />

avium (5 smooth and 5 rough morphotypes) were assessed<br />

in human PBMCs and monocytes. Mycobacteria-induced<br />

TNFα response is enhanced for M. abscessus (8535<br />

±1010 pg/ml) as compared to M. avium (2400±244 pg/<br />

ml, pb0.017) and no major differences were noted when<br />

compared colony morphologies within the same species.<br />

All mycobacterial strains were able to activate p38 MAP<br />

kinase phosphorylation and NF-kB translocation. Induction<br />

<strong>of</strong> TNFα was dependent on p38 MAPK signaling pathway<br />

since pre-incubation <strong>of</strong> cells with the p38 signaling<br />

inhibitor (SB203580) led to N80% reduction in cytokine<br />

secretion. In addition, treatment <strong>of</strong> cells with anti-human<br />

TLR2 antibodies showed TLR2 to be at least partially<br />

involved in signaling for M. abscessus as well. The present<br />

data indicate that M. avium is a more virulent pathogen<br />

than M. abscessus and elicits limited activation <strong>of</strong> cellular<br />

effector mechanisms, thereby escaping elimination.<br />

Accordingly, lung disease induced by NTM in nonpredisposing<br />

individuals may be associated with a yet uncharacterized<br />

underlying genetic defect.<br />

doi:10.1016/j.clim.2007.03.279<br />

F.67 Characterization <strong>of</strong> the Cellular Immunity in<br />

Patients Presenting Extensive Dermatophytoses Due<br />

to Trichophyton Rubrum<br />

Dewton Moraes Vasconcelos, MD, PhD, Department <strong>of</strong><br />

Dermatology, University <strong>of</strong> São Paulo Medical School, São Paulo,<br />

Brazil, Anna Cristina Collanieri, BSc, Department <strong>of</strong><br />

Dermatology, University <strong>of</strong> São Paulo Medical School, São Paulo,<br />

Brazil, Mauricio Domingues Ferreira, MD, PhD, Department <strong>of</strong><br />

Dermatology, University <strong>of</strong> São Paulo Medical School, São Paulo,<br />

Brazil, Tatiana Negri Santi-BSc, Department <strong>of</strong> Dermatology,<br />

University <strong>of</strong> São Paulo Medical School, São Paulo, Brazil, Anete<br />

S. Grumach, MD, PhD, Department <strong>of</strong> Dermatology, University<br />

<strong>of</strong> São Paulo Medical School, São Paulo, Brazil, Alexandre<br />

Almeida, MD, MSc, Department <strong>of</strong> Dermatology, University <strong>of</strong><br />

São Paulo Medical School, São Paulo, Brazil, Alberto Jose da<br />

Silva Duarte, MD, PhD, Department <strong>of</strong> Dermatology, University<br />

<strong>of</strong> São Paulo Medical School, São Paulo, Brazil<br />

Background: Dermatophytes cause infection in humans<br />

independent <strong>of</strong> the immunological status <strong>of</strong> the patient. In<br />

common to other infections the clinical features differ in<br />

immunodeficient patients. Dermatophytoses in cellular immunodeficient<br />

patients are usually less inflammatory, but some<br />

patients present pustular extensive lesions, frequently with<br />

follicular involvement.<br />

Objectives: Obtaining a reliable antigen by growth and<br />

purification <strong>of</strong> T. rubrum and to evaluate the immune<br />

reactivity “in vitro”, and quantify the immune response to<br />

the peptide YIIDTGIDID <strong>of</strong> T. rubrum. Methods: The fungal<br />

samples were obtained from the fungal library <strong>of</strong> the Institute<br />

<strong>of</strong> Tropical Medicine for antigen preparation, and the peptide<br />

was synthesized by EvoQuest. The lymphoproliferation assay<br />

was performed by tritiated thymidine incorporation and the<br />

cytokine quantifications by ELISA <strong>of</strong> culture supernatants.<br />

Results: The antigenic extract was efficient in the stimulation<br />

<strong>of</strong> cell cultures. The response to the peptide was also efficient<br />

and highly specific in sensitized controls. Despite most<br />

patients presented deficient responses, some presented<br />

normal lymphoproliferation. There was no difference<br />

between the cytokine secretion among patients and controls.<br />

doi:10.1016/j.clim.2007.03.280<br />

F.68 Evaluation <strong>of</strong> Cellular Responses to Rotavirus<br />

and NSP4 in Children during Natural Rotavirus<br />

Infection<br />

Jyoti Logani, Research Officer, Department <strong>of</strong> Pediatrics, All<br />

India Institute <strong>of</strong> Medical Sciences, New Delhi, India, Santosh<br />

Gupta, Research Associate, Department <strong>of</strong> Pediatrics, All India<br />

Institute <strong>of</strong> Medical Sciences, New Delhi, India, Shinjini<br />

Bhatnagar, Scientist, Department <strong>of</strong> Pediatrics, All India<br />

Institute <strong>of</strong> Medical Sciences, New Delhi, India, Pratima Ray,<br />

Scientist, Department <strong>of</strong> Pediatrics, All India Institute <strong>of</strong><br />

Medical Sciences, New Delhi, India, M.K. Bhan, Department <strong>of</strong><br />

Pediatrics, All India Institute <strong>of</strong> Medical Sciences, New Delhi,<br />

India<br />

Rotavirus (RV) is a major cause <strong>of</strong> gastroenteritis in young<br />

children leading to ∼611,000 deaths annually. Further


Abstracts<br />

development and evaluation <strong>of</strong> effective vaccines require better<br />

understanding <strong>of</strong> immune effectors involved in protective<br />

immunity. We examined IFN-γ and/or IL-4 responses in PBMC <strong>of</strong><br />

children suffering from RV (n=25), non-RV (n=10) gastroenteritis<br />

and healthy adults (n=10) by ELISPOT assay. We detected IFN-γ<br />

but no IL-4 response to NSP4 and RV in children with RV<br />

gastroenteritis and adults. IFN-γ responses in adults were<br />

persistent and significantly higher than children (pb0.01). The<br />

responses to the RV were stronger in magnitude in comparison to<br />

NSP4, but were positively correlated (pb0.01, rs=0.666). During<br />

RV gastroenteritis, 8/25 children elicited IFN-γ response to NSP4<br />

between 1 and 30 days after onset <strong>of</strong> diarrhea; in the control<br />

uninfected children, no response to NSP4 was observed. The %<br />

responders with positive IFN-γ response to NSP4 detected<br />

between 1 and 6 days <strong>of</strong> illness were 11.1%, 66.6% between 7<br />

and 15 days and 30% between 16 and 30 days <strong>of</strong> illness. No IFN-γ<br />

responses were detected beyond 30 days <strong>of</strong> diarrhea. On<br />

screening <strong>of</strong> three samples from same subject, it was further<br />

validatedthatatransientriseintheIFN-γresponsetoNSP4occurs<br />

between 7 and 25 days <strong>of</strong> symptomatic RV diarrhea. Stool RV+<br />

children depicted similar kinetics <strong>of</strong> IFN-γ response to RV. Thus<br />

significant IFN-γ response to NSP4 and RV indicates Th1 response<br />

duringnaturalRVinfection.IFN-γresponseistransientinchildren<br />

whereas in adults, responses are persistent and may play a role in<br />

protection.<br />

doi:10.1016/j.clim.2007.03.281<br />

F.69 Differential NF-kappaB Responses Induced by<br />

Bordetella Pertussis Filamentous-Hemagglutinin<br />

(FHA) in Macrophages and Bronchial Epithelial Cells<br />

Tzvia Abramson, Pr<strong>of</strong>esor, SJSU, Palo Alto, CA, David<br />

Relman, Pr<strong>of</strong>essor, Stanford University, Palo Alto, CA,<br />

Hassya Kedem, Research Associate, Stanford University,<br />

Palo Alto, CA<br />

Filamentous hemagglutinin (FHA), an adhesin and secreted<br />

factor <strong>of</strong> Bordetella pertussis, induces both inflammatory and<br />

apoptotic responses. Given the role <strong>of</strong> NF-kappaB transcription<br />

factor family in both these functions, we investigated the FHA<br />

interference with this pathway. FHA (5 μg/ml) induces a rapid<br />

degradation <strong>of</strong> IkappaB (within 30 min) in human monocytes<br />

and U-937 macrophages. IkappaB cytosolic levels are restored<br />

and accumulated within 2 h. This activation <strong>of</strong> NF-kappaB<br />

pathway is confirmed by NF-kappaB DNA binding as well as by<br />

inflammatorycytokinesecretion.None<strong>of</strong>thoseactivitieswas<br />

observed in BEAS-2B bronchial epithelial cells. Surprisingly, 2hour<br />

exposure <strong>of</strong> both epithelial cells and macrophages to FHA<br />

blocked the ability <strong>of</strong> TNF-α to induce proteasomal degradation<br />

<strong>of</strong> IkappaB as well as the nuclear translocation <strong>of</strong> p65.<br />

Immunoprecipitation analysis revealed the ubiquitination and<br />

phosphorylation <strong>of</strong> IkappaB, implicating that 2-hour treatment<br />

with FHA interferes with the proteasomal activity rather than<br />

an upstream activity. Attenuated proteasome activity is<br />

revealed at 2–8 h FHA treatment in both cell types. However,<br />

no significant inhibitory activity was observed at less than 1 h<br />

exposure to FHA. These results imply that the accumulated<br />

levels <strong>of</strong> IkappaB at 2 h exposure <strong>of</strong> cells to FHA may result in<br />

proteasomal inhibition. This study suggests that despite the<br />

immediate strong activation <strong>of</strong> inflammatory responses by FHA<br />

in macrophages, longer exposure <strong>of</strong> immune and host cells to<br />

this secreted virulent factor may trigger anti-inflammatory<br />

activity which may interfere with the development <strong>of</strong> an<br />

effective immune response that will resolve the infection<br />

promptly.<br />

doi:10.1016/j.clim.2007.03.282<br />

F.70 Cytokine Activated Human Neutrophils<br />

Simultaneously Express T Cell Co-Stimulatory and<br />

Negative Regulatory Molecules<br />

Paul Bankey, Associate Pr<strong>of</strong>essor, University <strong>of</strong> Rochester<br />

Medical Center; Surgery, Rochester, NY, Mita De, Research<br />

Associate, University <strong>of</strong> Rochester Medical Center; Surgery,<br />

Rochester, NY, Andrea Zucchiatti, Research Fellow,<br />

University <strong>of</strong> Rochester Medical Center; Surgery, Rochester,<br />

NY, Asit De, Assistant Pr<strong>of</strong>essor, University <strong>of</strong> Rochester<br />

Medical Center, Rochester, NY, Carol Miller-Grazia,<br />

Pr<strong>of</strong>essor <strong>of</strong> <strong>Immunology</strong>, University <strong>of</strong> Rochester Medical<br />

Center; Surgery, Rochester, NY<br />

The role <strong>of</strong> neutrophils (PMN) in innate immunity is well<br />

established. However, their role in adaptive immunity is not<br />

well characterized. Initiation <strong>of</strong> adaptive immunity by pr<strong>of</strong>essional<br />

antigen presenting cells (APCs) is critically dependent on<br />

expression <strong>of</strong> MHC class II molecules (e.g., HLA-DR) and costimulatory<br />

molecules (e.g., CD86). APCs also regulate T cell<br />

mediated adaptive immunity through expression <strong>of</strong> negative<br />

regulatory molecules such as program death ligands (PDL) and<br />

immunoglobulin like transcript (ILT) molecules. Our purpose<br />

was to assess the surface expression <strong>of</strong> HLA-DR, CD86, PD-L1<br />

and 2 and ILT-2 and 4 by flow cytometry in fresh and cytokine<br />

(GM-CSF+IFN-γ: G+I) treated neutrophils isolated from healthy<br />

controls (n=22). Untreated neutrophils expressed low levels<br />

(b2%) <strong>of</strong> HLA-DR and CD86; however, in vitro culture <strong>of</strong><br />

neutrophils with cytokines G+I significantly increased the<br />

expression <strong>of</strong> both molecules (HLA-DR: 5.61±1.16% at 24 h<br />

and 17.94±2.95% at 48h; CD86: 2.57±0.45% at 24 h and 8.11±<br />

1.59% at 48 h). Concurrently, cytokines G+I also significantly<br />

induce PMN expression <strong>of</strong> the T cell negative regulatory<br />

molecules: PD-L1 and 2 (PD-L1: 0.11±0.06% in fresh cells,<br />

26.49±2.47% at 24 h and 39.55±4.72% at 48 h; PD-L2: 0.25±<br />

0.0.25% in fresh cells, 6.12±1.37% at 24 h and 14.94±4.58% at<br />

48 h). ILT4 is constitutively expressed on over 90% <strong>of</strong> human<br />

neutrophils and expression is unchanged after culture with G+I.<br />

ILT-2 is not expressed on either fresh or G+I treated neutrophils.<br />

Simultaneous expression <strong>of</strong> T cell negative signaling molecules<br />

(PD-L1 and 2, ILT4) and MHC/co-stimulatory molecule CD86 in<br />

neutrophils supports an expanded role for PMN in both initiation<br />

and regulation <strong>of</strong> adaptive immunity.<br />

doi:10.1016/j.clim.2007.03.283<br />

S37<br />

F.71 An Analysis <strong>of</strong> HCV and CMV Specific Effector T<br />

Cells in Peripheral Blood, Perihepatic Lymph Nodes<br />

and Liver in HCV Infected and Uninfected Patients<br />

Dilip Moonka, Medical Director <strong>of</strong> Liver Transplantation,<br />

Henry Ford Health Systems, Division <strong>of</strong> Gastroenterology,


S38 Abstracts<br />

Detroit, MI, Kimberly Milkovich, Research Associate,<br />

Division <strong>of</strong> Rheumatology, Case Western Reserve University,<br />

Cleveland, OH, Benig Rodriguez, Assistant Pr<strong>of</strong>essor, Center<br />

for AIDS Research, Case Western Reserve University,<br />

Cleveland, OH, Michael Lederman, Pr<strong>of</strong>essor, Case Western<br />

Reserve University, Center for AIDS Research, Cleveland,<br />

OH, Marwan Abouljoud, Director <strong>of</strong> Transplant Institute,<br />

Henry Ford Health Systems, Transplant Institute, Detroit,<br />

MI, Donald Anthony, Assistant Pr<strong>of</strong>essor, Case Western<br />

Reserve University, Division <strong>of</strong> Rheumatology and Infectious<br />

Disease, Cleveland, OH<br />

It is difficult to detect HCV-specific T cells in the peripheral<br />

blood <strong>of</strong> chronic HCV patients, while T cells specific<br />

for other viruses appear intact. One explanation for this is a<br />

compartmentalization <strong>of</strong> the immune response. Here we<br />

performed phenotypic analysis <strong>of</strong> T cells isolated from liver,<br />

perihepatic lymph nodes (LN), and peripheral blood <strong>of</strong> 23<br />

HCV infected and 12 uninfected subjects undergoing liver<br />

transplantation. Results: HCV specific IFN-γ and proliferative<br />

responses were commonly observed in the LN <strong>of</strong> HCV infected<br />

subjects (40% and 46% respectively), while they were<br />

uncommonly observed in the peripheral blood and liver<br />

(IFN-γ 16% and 25% respectively; proliferation 11% and 8%<br />

respectively). In contrast, CMV specific IFN-γ producing<br />

responses were not as commonly observed in the LN (30%)<br />

compared to the periphery (58%) and liver (60%) <strong>of</strong> these<br />

same HCV patients. Conclusions: These data are among the<br />

first to look at HCV-specific responses in perihepatic lymph<br />

nodes. The data indicate a selective defect in HCV, but not<br />

CMV specific, T cell effector function in the periphery <strong>of</strong><br />

chronic HCV patients. The relatively common presence <strong>of</strong><br />

HCV-reactive T cells in the LN is consistent with T cell<br />

activation at this site. Finally, we do not detect HCVspecific<br />

T cells more readily in the liver compared to the<br />

periphery or LN, indicating that hepatic lymphocytes are<br />

defective in effector function similar to those in the<br />

peripheral blood. The frequency <strong>of</strong> HCV-specific T cells did<br />

not clearly correlate with the severity <strong>of</strong> recurrent HCV<br />

after transplant.<br />

doi:10.1016/j.clim.2007.03.284<br />

F.72 Carbohydrate Analysis <strong>of</strong> HIV Envelope<br />

Glycoprotein<br />

Ingrid D. Cruzado-Park, Senior Development Scientist,<br />

Beckman Coulter, Inc. Fullerton, CA, Munir Alam, Duke<br />

University Medical Center, Durham, NC, Hua-Xin Liao, Duke<br />

University Medical Center, Durham, NC, Barton Haynes,<br />

Director, Duke University Medical Center, Human Vaccine<br />

Institute and CHAVI, Durham, NC, Heather Desaire, Duke<br />

University Medical Center, Durham, NC, Enrique Rabelli,<br />

Director, Custom BioPharma Solutions, Beckman Coulter,<br />

Inc. Miami, FL, Sybil D’Costa, Staff Advanced Research<br />

Scientist, Beckman Coulter, Inc. Miami, FL, Michael H.<br />

Simonian, Manager, Biomarker Discovery and Automation<br />

Center, Beckman Coulter, Inc. Fullerton, CA, Chitra K.<br />

Ratnayake, Team Leader, Beckman Coulter, Inc. Biomarker<br />

Applications and Chemistry Development, Fullerton, CA<br />

To date over 65 million people have been infected with the<br />

human immunodeficiency virus (HIV) which continues to<br />

claim several million lives a year. There is an urgent need to<br />

design and develop vaccines that generate broad neutralizing<br />

antibody enabling preventative vaccination strategies.<br />

Although the virus has evolved to circumvent the immune<br />

response, for example, carbohydrates on the envelope<br />

glycoprotein <strong>of</strong> HIV serve as a strong defense against antibody<br />

mediated responses, there is evidence that these oligomannose<br />

components can also serve as ideal vaccine candidates<br />

due to their ability to generate efficacious neutralizing<br />

antibodies in HIV infected individuals (2G12 antibody) albeit<br />

infrequently. To address the role <strong>of</strong> glycosylation in HIV<br />

pathogenesis and immunogenicity, carbohydrate pr<strong>of</strong>iles<br />

from wild type and consensus modeled, genetically engineered<br />

envelope glycoproteins were compared. We present<br />

data showing the N-linked oligosaccharide pr<strong>of</strong>iles for two<br />

types <strong>of</strong> HIV gp140 envelope glycoproteins; JRFL is a primary<br />

isolate and the other from a genetically engineered strain<br />

containing a consensus sequence (ConS), which is more<br />

effective at eliciting neutralizing antibodies. Following<br />

treatment with PNGase F, released N-linked oligosaccharides<br />

were labeled with fluorescent 8-aminopyrene-1,3,6-trisulfonate<br />

and pr<strong>of</strong>iled using the ProteomeLab PA 800 Protein<br />

Characterization System with laser-induced fluorescence<br />

(LIF) detection (488 nm excitation, 520 nm emission). We<br />

observed significant differences in the carbohydrate pr<strong>of</strong>iles<br />

<strong>of</strong> these gp140 envelopes and will present and discuss those<br />

differences. This strategy for oligosaccharide characterization<br />

and subsequent identification may help evaluate role <strong>of</strong><br />

glycosylation in generating broadly neutralizing antibody<br />

responses and enable intelligent design <strong>of</strong> carbohydratebased<br />

epitope vaccines.<br />

doi:10.1016/j.clim.2007.03.285<br />

F.73 Defining the Role <strong>of</strong> BmIL-5 in Tropical<br />

Pulmonary Eosinophilia<br />

Sung (Steve) Kwon, University <strong>of</strong> Illinois College <strong>of</strong> Medicine<br />

at Rockford, Rockford, IL, Gnanasekar Munirathinam,<br />

University <strong>of</strong> Illinois College <strong>of</strong> Medicine at Rockford,<br />

Rockford, IL, Anand Setty Balakrishnan, University <strong>of</strong> Illinois<br />

College <strong>of</strong> Medicine at Rockford, Rockford, IL, Ramaswamy<br />

Kalyanasundaram, University <strong>of</strong> Illinois College <strong>of</strong> Medicine<br />

at Rockford, Rockford, IL<br />

This study describes characterization <strong>of</strong> a novel pathogenderived<br />

human IL-5 receptor binding molecule designated<br />

BmIL-5. This molecule was identified in our laboratory by<br />

screening a phage displayed cDNA expression library <strong>of</strong> the<br />

infective larvae <strong>of</strong> the human lymphatic filarial parasite,<br />

Brugia malayi. Soluble human IL-5 receptor (huIL-5R) was<br />

used as the bait. This screening procedure yielded a single<br />

clone and sequence analyses <strong>of</strong> the insert from this clone<br />

showed a novel protein designated as “B. malayi-derived IL-<br />

5-like molecule” (BmIL-5). Recombinant BmIL-5 was then<br />

expressed and purified. ELISA-based binding assays confirmed<br />

binding <strong>of</strong> rBmIL-5 to huIL-5R. Functional significance<br />

<strong>of</strong> this binding was then evaluated using a human eosinophil<br />

cell line, named Hs 454.T. Initial studies showed that BmIL-5


Abstracts<br />

binds to eosinophil surface. To test whether this binding<br />

initiates any signaling events, we evaluated STAT1 phosphorylation<br />

status in the eosinophil. We also performed a series <strong>of</strong><br />

experiments to identify the receptor binding region <strong>of</strong> BmIL-5<br />

by peptide mapping. In these studies, we designed overlapping<br />

20 mer peptides and evaluated the binding pattern <strong>of</strong><br />

these peptides to huIL-5R. These studies identified peptide 1<br />

and peptide 11 as the domains binding to huIL-5R. An<br />

interesting observation was that both these peptides interfered<br />

with the binding <strong>of</strong> huIL-5 to huIL-5R in vitro. Thus, we<br />

believe that BmIL-5 interferes with IL-5 function by preventing<br />

IL-5 binding to IL-5R. Our findings demonstrate a novel<br />

method <strong>of</strong> how pathogens potentially manipulate the human<br />

immune system.<br />

doi:10.1016/j.clim.2007.03.286<br />

F.74 What Triggers Transient AIDS in the Acute<br />

Phase <strong>of</strong> HIV Infection and Chronic AIDS at the End<br />

<strong>of</strong> the Incubation Period?<br />

Ivan Kramer, Associate Pr<strong>of</strong>essor, UMBC, Canada Physics,<br />

Catonsville, MD<br />

Novel dynamical models are introduced demonstrating<br />

that the T helper cell (THC) density drops in the acute<br />

infection phase <strong>of</strong> HIV infection, sometimes causing<br />

transient AIDS, and at the end <strong>of</strong> the incubation period<br />

causing chronic AIDS have a common dynamical cause. The<br />

immune systems’ inability to produce enough uninfected<br />

THCs to replace the infected ones it is destroying causes a<br />

drop in the THC density at any stage <strong>of</strong> HIV infection.<br />

Increases in viral infectivity, probably caused by random<br />

mutation <strong>of</strong> HIV, are shown to drive the progression <strong>of</strong> the<br />

infection. The existence <strong>of</strong> transient AIDS and this model<br />

which explains it are inconsistent with the antigenic<br />

diversity thresholds model <strong>of</strong> AIDS. The minimum incubation<br />

period for the long-term non-progressors (LTNPs) was<br />

calculated from a novel physical model: 0.3% <strong>of</strong> infected<br />

people have incubation periods <strong>of</strong> 23.1 years or more, and<br />

there is no biomedical difference between LTNPs and<br />

progressors. If this model is correct, then there is no<br />

unique, special anti-viral substance secreted by the CD8+ T<br />

cells <strong>of</strong> LTNPs that accounts for their unusually long<br />

incubation periods; indeed, to date, no such substance has<br />

been found although researchers have looked for it for<br />

years. The model also predicts that chronic AIDS results<br />

from 3 random transitions linking 4 clinically distinct stages<br />

<strong>of</strong> HIV infection following seroconversion, a result that<br />

agrees with the World Health Organization’s staging system<br />

based on clinical manifestations <strong>of</strong> the disease.<br />

doi:10.1016/j.clim.2007.03.287<br />

F.75 Characterization <strong>of</strong> a CD21low B Cell<br />

Subpopulation in Patients with CVID<br />

Mirzokhid Rakhmanov, Postdoctoral Fellow, Department <strong>of</strong><br />

Rheumatology and <strong>Clinical</strong> <strong>Immunology</strong>, University Hospital<br />

Freiburg, Freiburg i. Breisgau, Germany, Sylvia Gutenberger,<br />

Research Assistant, Department <strong>of</strong> Rheumatology and<br />

<strong>Clinical</strong> <strong>Immunology</strong>, University Hospital Freiburg, Freiburg i.<br />

Breisgau, Germany, Baerbel Keller, Graduate Student,<br />

Department <strong>of</strong> Rheumatology and <strong>Clinical</strong> <strong>Immunology</strong>,<br />

University Hospital Freiburg, Freiburg i. Breisgau, Germany,<br />

Klaus Warnatz, Assistant Medical Director, Department <strong>of</strong><br />

Rheumatology and <strong>Clinical</strong> <strong>Immunology</strong>, University Hospital<br />

Freiburg, Freiburg, Germany, Hans-Hartmut Peter, Director<br />

<strong>of</strong> the Department, Department <strong>of</strong> Rheumatology and<br />

<strong>Clinical</strong> <strong>Immunology</strong>, University Hospital Freiburg, Freiburg,<br />

Germany<br />

Common variable immunodeficiency (CVID) is a primary<br />

immune deficiency disorder characterized by hypogammaglobulinemia<br />

and recurrent bacterial infections. Analyzing B<br />

cell homeostasis in a subgroup <strong>of</strong> CVID patients with<br />

splenomegaly and autoimmune cytopenia revealed a B cell<br />

population with unusually low expression <strong>of</strong> CD21. We<br />

therefore termed these cells “CD21low” B cells. The low<br />

expression <strong>of</strong> CD21 is the effect <strong>of</strong> a reduced transcription.<br />

However, coexisting B cells expressing normal levels <strong>of</strong> CD21<br />

in these patients suggest that the low expression is not due to<br />

genetic dysregulation <strong>of</strong> CD21 expression, but rather the<br />

result <strong>of</strong> an unusual cellular differentiation. The phenotyping<br />

<strong>of</strong> these cells did not identify a stage along the current<br />

understanding <strong>of</strong> B cell differentiation, but the increased<br />

cell size and expression <strong>of</strong> CD86 suggest rather a recent<br />

activation status. The frequency <strong>of</strong> somatic hypermutations<br />

was increased compared to naïve CD21+ B cells, but below<br />

memory B cells <strong>of</strong> the same patient. We and others had<br />

previously described a similar B cell population in patients<br />

with SLE or chronic HIV infection. Interestingly, CD21low B<br />

cells express increased levels <strong>of</strong> MxA message, suggesting<br />

recent exposure to type I interferons. The expansion <strong>of</strong><br />

CD21low B cells is associated with severe shifts within the<br />

CD4+ T cell compartment <strong>of</strong> these patients. Therefore, we<br />

suggest an inflammatory dysregulation <strong>of</strong> the immune system<br />

underlying the disturbed B and T cell homeostasis in CVID<br />

patients with Freiburg type Ia.<br />

doi:10.1016/j.clim.2007.03.288<br />

S39<br />

F.76 Use <strong>of</strong> Prophylactic Antibiotics and Adjunctive<br />

Therapies in Primary Immunodeficiency Diseases<br />

Pierre Yong, Fellow, Hospital <strong>of</strong> the University <strong>of</strong><br />

Pennsylvania, Division <strong>of</strong> Allergy and <strong>Immunology</strong>,<br />

Philadelphia, PA, John Boyle, Founder, Immune Deficiency<br />

Foundation, Towson, MD, Jordan Orange, Assistant<br />

Pr<strong>of</strong>essor, Children’s Hospital <strong>of</strong> Philadelphia, Division <strong>of</strong><br />

Allergy and <strong>Immunology</strong>, Philadelphia, PA<br />

Background: There is limited data supporting the use <strong>of</strong><br />

prophylactic antibiotics and other adjunctive therapies in<br />

primary immunodeficiency diseases (PIDD). Methods: A webbased<br />

survey regarding therapy in PIDD was designed jointly by<br />

the Immune Deficiency Foundation and the American Academy<br />

<strong>of</strong> Allergy, Asthma & <strong>Immunology</strong> (AAAAI), and <strong>of</strong>fered to AAAAI<br />

members. The response rate was 13.5%, n = 405. Responses<br />

were analyzed with a basic statistical program. Those devoting


S40 Abstracts<br />

more than 10% <strong>of</strong> their clinical practice to PIDD were defined as<br />

experts. Results: Use <strong>of</strong> prophylactic antibiotics in PIDD is<br />

widespread and perceived to be efficacious. Significant<br />

differences, however, between expert and non-expert immunologists<br />

were found in frequency <strong>of</strong> use as well as in perceived<br />

effectiveness in multiple PIDD. Experts find prophylaxis more<br />

useful and use it more <strong>of</strong>ten. They also use prophylaxis in<br />

conjunction with IVIG more frequently. Live viral vaccines were<br />

most frequently avoided in severe combined immunodeficiency<br />

and DiGeorge syndrome. Additionally, differences in each<br />

group’s vaccine use were seen in several PIDD, including<br />

hyper-IgM syndrome. The only hygiene intervention felt<br />

beneficial was hand-washing with soap and water. Complementary<br />

and alternative medicine (CAM) was not believed to<br />

enhance immunity or prevent infections. Discussion: These<br />

results highlight the significant use <strong>of</strong> prophylactic antibiotics<br />

for PIDD by immunologists. Important differences were found<br />

between experts and non-experts. Surprisingly, many immunologists<br />

do not avoid live viral vaccines in many PIDD despite<br />

labeling recommending avoidance. Most hygiene and CAM<br />

interventions were not seen as beneficial, although some were<br />

more popular than others.<br />

doi:10.1016/j.clim.2007.03.289<br />

F.77 Common Variable Immunodeficiency:<br />

Presentation <strong>of</strong> 7 Cases and Review <strong>of</strong> Literature<br />

Carlos Torres-Loza, England Immunoallergist, West National<br />

Medical Center and Department <strong>of</strong> Allergy and <strong>Clinical</strong><br />

<strong>Immunology</strong>, Guadalajara, Jalisco, Mexico<br />

Aim <strong>of</strong> our work is to present a global view <strong>of</strong> this syndrome<br />

and analyze our experience <strong>of</strong> 7 adult patients bearing this<br />

syndrome which were seen at our department. It is a<br />

retrospective work for which it was designed a sheet to<br />

recollect data and also we reviewed literature <strong>of</strong> all possible<br />

electronic ways from the last 7 years, six are alive and one is<br />

now dead. The patients were diagnosed in most cases through<br />

air view infections such as pneumonia, chronic sinusitis and<br />

chronic middle otitis. In one <strong>of</strong> the cases an autoimmune<br />

process was the clinical problem before we made the<br />

diagnosis <strong>of</strong> this syndrome. During their evolution in our<br />

department, only three <strong>of</strong> them have had intestinal clinical<br />

manifestations characterized by several periods <strong>of</strong> chronic<br />

diarrhea and in three we found evidence <strong>of</strong> autoimmunity. All<br />

<strong>of</strong> the cases had low levels <strong>of</strong> serum immunoglobulin, neither<br />

<strong>of</strong> them had high serum level <strong>of</strong> IgE and all <strong>of</strong> our patients have<br />

shown normal lymphocytes subpopulations except in two<br />

where we found low levels <strong>of</strong> CD4.<br />

doi:10.1016/j.clim.2007.03.290<br />

F.78 Severe Agammaglobulinemia in an Adult Male<br />

with Nomal Number <strong>of</strong> Circulating B Cells<br />

Daniel Suez, Assistant <strong>Clinical</strong> Pr<strong>of</strong>essor at UTSW Dallas, TX,<br />

Daniel Suez, MD, AAIC, PA, Irving, TX, Hans Ochs, Pr<strong>of</strong>essor<br />

<strong>of</strong> Pediatrics, University <strong>of</strong> Washington School <strong>of</strong> Medicine,<br />

Seattle, WA<br />

We report here a case <strong>of</strong> a 34-year-old male with a history<br />

<strong>of</strong> recurrent pneumonia since early childhood with no other<br />

significant family history who presented to our clinic with<br />

severe agammaglobulinemia: IgG b7 mg/dl, IgM b5 mg/dl,<br />

IgA b13 mg/dl. Flow cytometry revealed normal numbers <strong>of</strong><br />

circulating B cells −9.5% <strong>of</strong> CD20 (221/mm 3 ) and 12.4% <strong>of</strong><br />

CD19 (322/mm 3 ) respectively. <strong>Clinical</strong> examination was<br />

remarkable for the paucity <strong>of</strong> lymphoid tissue. Further<br />

evaluation revealed normal numbers <strong>of</strong> memory B cells by<br />

CD27 expression yet very few switched memory B cells<br />

(1.36% vs. 23.7 in controls). The patient had normal BTK<br />

expression in B cells and had normal sequence analysis <strong>of</strong><br />

gDNA for CD40L. Flow cytometry analysis <strong>of</strong> CD3 lymphocyte<br />

subsets revealed CD4/CD8 inversion with total CD4 16.9%<br />

(439/mm 3 ) and CD8 <strong>of</strong> 85.5% (2220/mm 3 ) while NK cell<br />

number was at 2% (52/mm 3 ). Other laboratory findings<br />

included normal kidney and liver functions. There is no<br />

history <strong>of</strong> significant gut disease and the patient’s ability to<br />

maintain stable through IgG levels on intravenous immunoglobulin<br />

therapy rules out the possibility <strong>of</strong> protein loss or<br />

high catabolic rate to explain the severe agammaglobulinemia.<br />

The differential diagnosis considered: all hyper-IgM<br />

syndromes can be ruled out due to the severe IgM deficiency,<br />

XLP syndrome may be considered yet the patient has no<br />

history <strong>of</strong> mononucleosis. To rule out XLP the patient will be<br />

sequenced for SAP and EBV by PCR will be requested.<br />

Autosomal recessive agammaglobulinemia can be also considered<br />

yet unlikely with normal circulating B cell numbers.<br />

doi:10.1016/j.clim.2007.03.291<br />

F.79 Severe, Steroid Resistant, Numular Atopic<br />

Dermatitis with Hypogammaglobulinemia<br />

Daniel Suez, Assistant <strong>Clinical</strong> Pr<strong>of</strong>essor at UTSW Dallas,<br />

Daniel Suez, MD, AAIC, PA, Irving, TX, Hans Ochs, Pr<strong>of</strong>essor<br />

<strong>of</strong> Pediatrics, University <strong>of</strong> Washington School <strong>of</strong> Medicine,<br />

Seattle, WA<br />

We report here the case <strong>of</strong> a 26-month-old white male with<br />

severe, steroid resistant numular atopic dermatitis since early<br />

infancy. Allergy evaluation revealed severe IgE mediated<br />

sensitivities to food and perennial allergens with elevated<br />

total serum IgE N8900 IU/ml. The patient failed to respond to<br />

numerous attempts <strong>of</strong> aggressive steroid therapy, both topical<br />

(under closed dressings) and systemic treatment along with<br />

comprehensive supportive therapy. Laboratory evaluation<br />

revealed hypogammaglobulinemia with a serum IgG level <strong>of</strong><br />

342 mg/dl (normal range for age 453–916 mg/dl), and normal<br />

IgA (40 mg/dl) and IgM (28 mg/dl). CBC and differential revealed<br />

eosinophilia (N20%) and flow cytometry analysis was normal<br />

(CD19, CD3, CD4, CD8, CD16/56, CD45RO, and CD45RA). The<br />

patient was placed on high dose intravenous immunoglobulin<br />

therapy with dramatic improvement. Differential diagnosis<br />

considered is: Netherton syndrome (currently being evaluated<br />

by sequence analysis), Wiskott-Aldrich syndrome (which can be<br />

ruled out with normal platelet count), Omenn syndrome (yet not<br />

likely because he has normal numbers <strong>of</strong> circulating B cells),<br />

IPEX syndrome (yet not likely since the patient does not have any<br />

evidence for autoimmune disease such as polyendocrinopathy,


Abstracts<br />

neutropenia, or hemolytic anemia), and hyper IgE syndrome<br />

(yet unlikely as well since he does not have any <strong>of</strong> the clinical<br />

features).<br />

doi:10.1016/j.clim.2007.03.292<br />

F.80 Lymphocytic Interstitial Pneumonitis in<br />

Common Variable Immunodeficiency Disorders:<br />

A Comparison with HIV<br />

Dilani Arnold, Specialist Registrar, Department <strong>of</strong><br />

<strong>Immunology</strong>, John Radcliffe Hospital, Oxford, England,<br />

Gleeson Fergus, Consultant Radiologist, Department <strong>of</strong><br />

Radiology, Churchill Hospital, Oxford, England, Helen<br />

Chapel, Consultant in <strong>Immunology</strong>, Department <strong>of</strong><br />

<strong>Immunology</strong>, John Radcliffe Hospital, Oxford, England,<br />

Robert Davies, Consultant in Chest Medicine, Department<br />

<strong>of</strong> Chest Medicine, Churchill Hospital, Oxford, England,<br />

Siraj Misbah, Consultant in <strong>Immunology</strong>, Department <strong>of</strong><br />

<strong>Immunology</strong>, John Radcliffe Hospital, Oxford, England<br />

Background: The objective <strong>of</strong> this study was to assess the<br />

presentation <strong>of</strong> Lymphocytic Interstitial Pneumonitis (LIP) in<br />

Common Variable Immunodeficiency Disorders (CVID). Methods:<br />

The medical records <strong>of</strong> all CVID patients followed up in<br />

our department were examined retrospectively. Results: In<br />

our institution, between 1970 and 2006, 96 patients met the<br />

diagnostic criteria for CVID. Of these, six (four women, two<br />

men, mean age, 45 years, range 42−65) had biopsy-proven<br />

LIP (microscopic evidence <strong>of</strong> polyclonal lymphoid cell<br />

infiltrate). HIV PCR was negative in all patients. Two patients<br />

were asymptomatic; four presented with cough, fatigue and<br />

exertional dyspnoea (mean time 2.75 months (range 1−7)<br />

from initial presentation to LIP diagnosis). Five patients had<br />

elevated C-reactive protein (11−31); four had raised beta-2<br />

microglobulin (3.8−4.7). Two had T-cell lymphocytosis (2.5−<br />

8.09) with elevated CD4 counts (1.82−3.43). One patient had<br />

an elevated CD8 count <strong>of</strong> 5.14 (0.2−0.7) and another had B<br />

cell lymphopenia (CD19 0.06). Two had falling IgG levels<br />

(4.8−5.87) despite immunoglobulin therapy. The predominant<br />

radiological abnormalities on CT were ground-glass<br />

opacification and mediastinal lymphadenopathy (present<br />

in four patients). Two had subpleural nodules. Immunohistology<br />

showed either an excess <strong>of</strong> CD4 cells or CD8 cells in a minority <strong>of</strong><br />

patients. No viral antigen was identified. Conclusion: The<br />

presentation <strong>of</strong> LIP is highly variable and diagnostically<br />

challenging. Comparison with LIP in immunodeficiency states<br />

such as HIV shows similar clinical and radiological features. The<br />

aetiology <strong>of</strong> LIP in CVID and HIV is unknown but is presumed to be<br />

viral, though no specific virus was identified in our small cohort.<br />

doi:10.1016/j.clim.2007.03.293<br />

F.81 Defective CD107a Surface Expression Heralds<br />

Munc13-4 Defect and Discriminates Between<br />

Genetic Subtypes <strong>of</strong> Familial Hemophagocytic<br />

Lymphohistiocytosis (FHL)<br />

Daniela Pende, Senior Investigator, Istituto Nazionale per la<br />

Ricerca sul Cancro, Genova, Stefania Marcenaro, Fellow,<br />

DIMES, Genova, Stefania Martini, Technician, Istituto<br />

Nazionale per la Ricerca sul Cancro, Genova, Alessandra<br />

Santoro, Fellow, Ospedale “G. Di Cristina”, Palermo, Gillian<br />

Griffiths, Senior Investigator, Sir William Dunn School <strong>of</strong><br />

Pathology, Oxford, England, Maurizio Aricò, Physician,<br />

Ospedale dei Bambini “G. Di Cristina”, Palermo, Lorenzo<br />

Moretta, Full Pr<strong>of</strong>essor, Istituto G. Gaslini, Genova<br />

Familial hemophagocytic lymphohistiocytosis (FHL) is a<br />

rare, heterogeneous fatal disease <strong>of</strong> early infancy characterized<br />

by an hyperinflammatory syndrome. Defective cellular<br />

cytotoxicity results in pathogen persistence and hypercytokinemia<br />

with disseminated infiltration <strong>of</strong> lymphocytes and<br />

histiocytes and hemophagocytosis. Differential diagnosis<br />

between FHL due to PRF1 (FHL2) or Munc13-4 (FHL3) and<br />

additional still unclassified genetic subtypes is not easy<br />

unless mutation analysis is performed. We investigated<br />

natural killer (NK) cells function and phenotype in patients<br />

with FHL2 (n=5) or FHL3 (n=8). NK cells were cultured in IL-2<br />

prior to their use in the various assays. Here we report on the<br />

surface CD107a expression as a novel rapid tool for<br />

identification <strong>of</strong> patients with Munc13-4 defect. Upon target<br />

interaction and degranulation, FHL3 NK cells displayed low<br />

levels <strong>of</strong> surface CD107a staining, in contrast to normal<br />

controls or perforin-deficient NK cells. B-EBV cell lines and<br />

dendritic cell targets reveal the FHL3 NK cell defect while<br />

highly susceptible tumor targets were partially lysed by FHL3<br />

NK cells expressing only trace amounts <strong>of</strong> Munc13-4 protein.<br />

Perforin-deficient NK cells were completely devoid <strong>of</strong> any<br />

ability to lyse target cells. Cytokine production induced by<br />

mAb-crosslinking <strong>of</strong> triggering receptors was comparable in<br />

patients and normal controls. However, when cytokine<br />

production was induced by co-culture with 721.221 B-EBV<br />

cells, FHL NK cells resulted high producers, whereas control<br />

cells were almost ineffective. This could reflect survival<br />

versus elimination <strong>of</strong> B-EBV cells i.e., the source <strong>of</strong> NK cell<br />

stimulation, in patients vs. healthy controls, thus mimicking<br />

the pathophysiological scenario <strong>of</strong> FHL.<br />

doi:10.1016/j.clim.2007.03.294<br />

S41<br />

F.82 Autoimmune Polyendocrinopathy-Candidiasis-<br />

Ectodermal Dystrophy Syndrome (APECED) Caused<br />

by Homozygous P326L AIRE Mutations in a Brazilian<br />

Family<br />

Patrícia de Campos Pieri, Biologist, Laboratory <strong>of</strong> Medical<br />

Investigation 36, Pediatrics Department FMUSP, São Paulo,<br />

Brazil, Joao B. Oliveira, Associate Researcher, Molecular<br />

<strong>Immunology</strong> and Genetics Section, Laboratory <strong>of</strong> Medical<br />

Investigation #56, USP, São Paulo, Brazil, Cristina Miuki Abe<br />

Jacob, Head <strong>of</strong> Unit, Department <strong>of</strong> Pediatrics, Allergy and<br />

<strong>Immunology</strong> Unit FMUSP, São Paulo, Brazil, Alberto J.S.<br />

Duarte, Lab Director, Laboratory <strong>of</strong> Medical Investigation<br />

#56 (LIM-56), USP, São Paulo, Brazil, Antonio Carlos Pastori,<br />

Assistant Pr<strong>of</strong>essor, Pediatrics Department, Allergy and<br />

<strong>Immunology</strong> Unit FMUSP, São Paulo, Brazil, Magda Maria<br />

Carneiro-Sampaio, Head <strong>of</strong> Department, Department <strong>of</strong><br />

Pediatrics, Allergy and <strong>Immunology</strong> Unit, FMUSP, São Paulo,<br />

Brazil


S42 Abstracts<br />

Introduction: APECED is characterized by the early onset<br />

<strong>of</strong> multiple organ directed autoimmunity, chronic mucocutaneous<br />

candidiasis and ectodermal dystrophy, and caused<br />

by recessive mutations in AIRE gene. The vast majority <strong>of</strong><br />

affected patients are <strong>of</strong> northern European descent,<br />

Sardinian or Iranian Jews. We describe here a Brazilian<br />

family affected by the disease. Material and methods: All<br />

14 exons and flanking intronic regions <strong>of</strong> AIRE were<br />

amplified by PCR, purified and sequenced using an<br />

automated capillary sequencer. Results: The index patient,<br />

LGM, is a 15-year-old boy from São Paulo. At 7 months he<br />

developed recurrent thrush and at the age <strong>of</strong> 2 years<br />

presented ungueal candidiasis. At 9 years his height was<br />

119 cm (pb3), levels <strong>of</strong> IGF1 and IGFBP3 were low, and a<br />

diagnosis <strong>of</strong> GH deficiency was made. At 12 years he<br />

developed anti-adrenal antibodies (1/40), and at 15 years<br />

he showed high levels <strong>of</strong> ACTH and PTH. His mother<br />

presents oral thrush, megaloblastic anemia and hypoparathyroidism,<br />

and his sister has recurrent candidiasis, without<br />

signs <strong>of</strong> autoimmunity. Parents deny consanguinity.<br />

Genomic DNA sequencing <strong>of</strong> AIRE identified a homozygous<br />

missense mutation in exon 8 (Pro326Leu). This mutation<br />

affects the first PHD finger domain, probably interfering<br />

with adequate subcellular protein localization and functioning.<br />

Conclusion: This is to our knowledge the first report<br />

<strong>of</strong> AIRE mutations causing APECED in a Brazilian family, and<br />

<strong>of</strong> a P326L AIRE mutation in a homozygous state. Molecular<br />

diagnosis is important for genetic counseling and institution<br />

<strong>of</strong> adequate therapy early in life.<br />

doi:10.1016/j.clim.2007.03.295<br />

F.83 NOD2 Polymorphisms in CVID: Investigation<br />

into Associations with Gastrointestinal Pathology<br />

and Granulomatous Disease<br />

Kerri Packwood, Grade A <strong>Clinical</strong> Scientist in<br />

<strong>Immunology</strong>, <strong>Clinical</strong> <strong>Immunology</strong>, Oxford Radcliffe<br />

Hospitals NHS Trust, Oxford, OXON, England, Berne Ferry,<br />

Principle Scientist, <strong>Clinical</strong> <strong>Immunology</strong>, Oxford Radcliffe<br />

Hospitals NHS Trust, Oxford, OXON, England, Divya<br />

Punwani, PhD Student, Wetherall Institute <strong>of</strong> Molecular<br />

Medicine, Human <strong>Immunology</strong> Unit, Oxford, OXON,<br />

England, Eduardo Lopez-Granados, Consultant<br />

Immunologist, <strong>Clinical</strong> <strong>Immunology</strong>, Oxford Radcliffe<br />

Hospitals NHS Trust, Oxford, OXON, England, Helen<br />

Chapel, Consultant Immunologist, <strong>Clinical</strong> <strong>Immunology</strong>,<br />

Oxford Radcliffe Hospitals NHS Trust, Oxford, OXON,<br />

England<br />

Common Variable Immunodeficiency Disorders (CVID)<br />

are a heterogeneous group <strong>of</strong> diseases characterised by<br />

hypogammaglobulinaemia. Associated complications<br />

include enteropathy and granulomatous disease. These<br />

conditions have diverse immunopathogeneses including<br />

primary defects in B cells, T cells, macrophages, dendritic<br />

cells, NK cells and cytokine production yet <strong>of</strong>ten the<br />

underlying aetiology is unknown. Prediction <strong>of</strong> complications<br />

would aid clinical management and we are<br />

investigating possible disease modifier genes as a potentially<br />

powerful tool. Classification <strong>of</strong> heterophenotypic<br />

CVID subgroups would also enable more focused and<br />

productive research studies. Mutations <strong>of</strong> the NOD2 gene<br />

including gly908arg, leu1007finsc and arg702trp polymorphisms<br />

are associated with Crohn'€s disease. We<br />

hypothesised that NOD2 may also be a disease modifier<br />

gene towards an enteropathic or granulomatous CVID<br />

phenotype. Methods to examine the above three polymorphisms<br />

were established and investigated in a cohort<br />

<strong>of</strong> 52 CVID patients and 26 non-patient controls. Five<br />

heterozygous NOD2 leu1007finsc mutations, two heterozygous<br />

NOD2 gly908arg mutations and four heterozygous<br />

NOD2 arg702trp mutations were found amongst CVID<br />

patients giving allele frequencies <strong>of</strong> 0.048, 0.019 and<br />

0.038 respectively. No polymorphisms were found amongst<br />

non-patient controls. Although to date we are unable to<br />

demonstrate a significant genotype/phenotype association,<br />

results from this study show this as reliable<br />

screening method for gly908arg, leu1007finsc and<br />

arg702trp NOD2 polymorphisms. More patients are<br />

required to conclude whether these and/or additional<br />

NOD2 polymorphisms can define a clinical subgroup <strong>of</strong><br />

CVID patients likely to develop enteropathic or granulomatous<br />

complications.<br />

doi:10.1016/j.clim.2007.03.296<br />

F.84 Development <strong>of</strong> Regulatory T Cells After<br />

Thymus Transplantation in Subjects with Complete<br />

DiGeorge Syndrome<br />

Ivan Chinn, Fellow-In-Training, Duke University Medical<br />

Center, Pediatrics, Durham, NC, Blythe Devlin, Assistant<br />

Research Pr<strong>of</strong>essor, Duke University Medical Center,<br />

Pediatrics, Durham, NC, M. Louise Markert, Associate<br />

Pr<strong>of</strong>essor, Duke University Medical Center, Pediatrics,<br />

Durham, NC<br />

Purpose: Characterize the development <strong>of</strong> CD4+Foxp3+<br />

regulatory Tcells in subjects with complete DiGeorge syndrome<br />

after thymus transplantation. Background: Forty-four subjects<br />

with complete DiGeorge syndrome, including athymia (b50<br />

naïveTcells/cumm),havereceivedculturedpostnatalallogeneic<br />

HLA nonmatched thymus grafts. Twenty-seven <strong>of</strong> 32<br />

survivors are currently at 1-year or greater post-transplantation.<br />

All 1-year survivors have naïve T cells with normal<br />

proliferative responses to phytohemagglutinin and normal T<br />

cell receptor repertoires. We now report the percentages <strong>of</strong><br />

Foxp3+ CD4+ T cells in the peripheral blood <strong>of</strong> 11 subjects who<br />

are 1-year survivors after thymus transplantation. Five <strong>of</strong> the<br />

subjects received peri-transplantation immunosuppression.<br />

Five <strong>of</strong> the 11 subjects have autoimmune thyroid disease,<br />

including one subject in the group who received immunosuppression.<br />

Results: All eleven subjects had percentages <strong>of</strong> CD4<br />

+Foxp3+ Tcells in the expected range compared to healthy adult<br />

controls at various time points post-transplantation. Absolute<br />

numbers <strong>of</strong> CD4+Foxp3+ T cells were low (12−65 cells/cumm),<br />

reflecting the subjects’ low total T cell counts for age. Similar<br />

percentages <strong>of</strong> regulatory T cells were found in subjects who<br />

received immunosuppression (median 3.85%, range 3.48−5.33%)<br />

compared with those who did not (median 4.58%, range 2.96<br />

−7.35%). The percentages <strong>of</strong> CD4+Foxp3+ Tcells did not appear


Abstracts<br />

to correlate with the development <strong>of</strong> autoimmune thyroid<br />

disease (median 5.2% [range 3.05−6.96%] in subjects with<br />

thyroid disease vs. 3.8% [range 2.96−7.35%] without thyroid<br />

disease). Discussion: Subjects with complete DiGeorge syndrome<br />

develop normal percentages <strong>of</strong> regulatory T cells in the<br />

peripheral blood after thymus transplantation.<br />

doi:10.1016/j.clim.2007.03.297<br />

F.85 Identification <strong>of</strong> a Mutation in the Gene Encoding<br />

the Endosomal Adaptor Protein p14 (MAPBPIP) in a<br />

Novel Primary Immunodeficiency Syndrome<br />

Jens Thiel, MD, University Hospital Freiburg, Freiburg,<br />

Germany<br />

The study <strong>of</strong> human primary immunodeficiency disorders<br />

has greatly advanced our understanding <strong>of</strong> basic<br />

host defense. We have investigated the molecular etiology<br />

<strong>of</strong> a novel autosomal recessive primary immunodeficiency<br />

syndrome. In a pedigree <strong>of</strong> 15 members <strong>of</strong> a Caucasian<br />

Mennonite family, four children suffered from congenital<br />

neutropenia, B cell and cytotoxic T cell deficiency. These<br />

children additionally showed a characteristic clinical<br />

phenotype associating small stature, hypopigmented<br />

skin, coarse facial features, and recurrent bronchopulmonary<br />

infections. In an attempt to identify a genetic<br />

basis for this novel immunodeficiency syndrome, we<br />

carried out a genetic linkage study. Since the family<br />

belongs to a religious isolate <strong>of</strong> Mennonite background,<br />

we expected that the disease would be associated with a<br />

homozygous mutation in a single gene. We identified four<br />

adjacent, perfectly segregating markers on chromosome<br />

1q21: D1S498, D1S2346, D1S305, and D1S1153. Within this<br />

minimal critical region <strong>of</strong> the linkage interval, there were<br />

a total <strong>of</strong> 87 genes or predicted genes and within the<br />

maximal possible linkage region, there were an additional<br />

105 genes or predicted genes. Combining the results <strong>of</strong><br />

the linkage analysis with a transcriptional pr<strong>of</strong>iling<br />

approach, only one gene located within the critical region<br />

on chromosome 1q21 was significantly underexpressed in<br />

the patients, namely MAPBPIP. By genomic sequencing a<br />

homozygous mutation in the 3′-UTR <strong>of</strong> MAPBPIP, a recently<br />

identified adaptor molecule controlling late endosomal<br />

signaling by the MP1–MAPK complex was identified in the<br />

four index family members. Neutrophils from all patients<br />

showed decreased RNA and protein levels <strong>of</strong> p14.<br />

doi:10.1016/j.clim.2007.03.298<br />

F.86 Foxp3 Expression Following Bone Marrow<br />

Transplantation for IPEX Syndrome After<br />

Reduced-Intensity Conditioning<br />

Morna J. Dorsey, Assistant Pr<strong>of</strong>essor, University <strong>of</strong> South<br />

Florida, St. Petersburg, FL, Aleksandra Petrovic, Assistant<br />

Pr<strong>of</strong>essor, All Children’s Hospital, St. Petersburg, FL,<br />

Matthew R. Morrow, Director, Flow Cytometry Core Facility,<br />

University <strong>of</strong> South Florida, Pediatrics, St. Petersburg, FL,<br />

John W. Sleasman, Pr<strong>of</strong>essor, University <strong>of</strong> South Florida,<br />

Pediatrics, St. Petersburg, FL, Larry J. Dishaw, Instructor,<br />

All Children’s Hospital, St. Petersburg, FL<br />

Objectives: To determine if immune reconstitution <strong>of</strong> Foxp3+<br />

regulatory T cells correlates with clinical improvement <strong>of</strong> IPEX<br />

syndrome following allogeneic HSCT. Methods: 8-month-old<br />

male infant with a mutation in the polyadenylation site <strong>of</strong> Foxp3<br />

gene, absence <strong>of</strong> Foxp3 protein expression and clinical<br />

manifestations <strong>of</strong> IPEX syndrome, including eczema, colitis,<br />

failure to thrive, TPN requirement, and elevated serum IgE,<br />

underwent matched unrelated HSCT following reduced-intensity<br />

conditioning regimen. Donor chimerism was determined by<br />

molecular analysis <strong>of</strong> blood or bone marrow. Blood samples were<br />

collected pre- and post-transplant. Regulatory Tcell population<br />

was determined by flow cytometry staining for Foxp3+<br />

CD25bright CD4+ T cells. Semi-quantitative PCR <strong>of</strong> Foxp3+<br />

mRNA was conducted as well. Results: Patient underwent<br />

reduced-intensity conditioning with alemtuzumab followed by<br />

fludarabine and melphalan. GVHD prophylaxis included tacrolimus,<br />

methotrexate, and prednisone. Neutrophils engrafted<br />

day +15, and platelets day+29. Patient was a full donor chimera<br />

day +28 and +60. Foxp3 protein expression was absent pre-<br />

HSCT. Post-transplantation, percentage CD4+ T cells expressing<br />

Foxp3+ CD25bright phenotype increased from 4.5% (day +29) to<br />

23% (day +90) and continued in this trend. Foxp3 mRNA<br />

expression confirmed flow cytometry data. Serum IgE levels<br />

decreased from 5000 IU/mL pre-transplant to 6 IU/mL on day<br />

+90, eczema resolved, and secretory diarrhea and feeding<br />

intolerance improved. Conclusions: Regulatory T cell reconstitution<br />

is evident soon after HSCT following reduced-intensity<br />

conditioning correlating with development <strong>of</strong> full donor<br />

chimerism. Increased Foxp3 expression correlates with correction<br />

<strong>of</strong> clinical and laboratory manifestations <strong>of</strong> IPEX syndrome<br />

providing evidence that HSCT is a curative procedure for this<br />

disorder.<br />

doi:10.1016/j.clim.2007.03.299<br />

S43<br />

F.87 Monoclonal Gammapathy Following Cord Blood<br />

Transplantation for Chediak−Higashi Syndrome<br />

Kevin YehSheng Wang, FIT, Pediatrics, University <strong>of</strong><br />

California, Los Angeles, Los Angeles, CA, Robert Roberts,<br />

Pr<strong>of</strong>essor <strong>of</strong> Pediatrics, Department <strong>of</strong> Pediatrics,<br />

University <strong>of</strong> California, Los Angeles, Los Angeles, CA<br />

Introduction: Chediak–Higashi syndrome (CHS) is a rare<br />

autosomal recessive disorder which <strong>of</strong>ten undergoes progression<br />

to a life-threatening accelerated phase with<br />

lymphohistiocytic infiltration in multiple organs. We report<br />

a case <strong>of</strong> CHS in the accelerated phase which was successfully<br />

treated with IVIG and high dose corticosteroids. He<br />

underwent cord blood transplantation and then developed a<br />

monoclonal gammapathy. Case report: Our patient was<br />

diagnosed with CHS at 16 months <strong>of</strong> age by peripheral<br />

blood smear which revealed giant granules in his white blood<br />

cells. At 6 years <strong>of</strong> age, he presented to us with<br />

pancytopenia, fever, respiratory stress, mental status<br />

changes, and seizure activity. On physical examination, he


S44 Abstracts<br />

had significant hepatosplenomegaly. Bone marrow biopsy<br />

showed cytoplasmic inclusions granule and hemophagocytic<br />

cells. For treatment, he received 3 courses <strong>of</strong> IVIG (4.5 g/kg<br />

total). He was also given methlyprednisolone. He was then<br />

placed on dexamethasone until the time <strong>of</strong> transplantation.<br />

Over the course <strong>of</strong> several weeks, his mental status returned<br />

to baseline, seizure activity ceased, hepatomegaly resolved,<br />

and pancytopenia improved. Therefore his accelerated<br />

phase had undergone remission. He then received a 10/10<br />

match cord blood transplantation. The patient appeared to<br />

engraft successfully following the transplantation and<br />

routine infusions <strong>of</strong> IVIG were stopped. Approximately<br />

15 months after transplantation, the patient was clinically<br />

stable but serum IgG level was found to be 4000 mg/dL and<br />

his CD 4/8 ratio was 0.55. Immunoelectrophoresis revealed<br />

that he had developed a monoclonal IgG gammapathy. Bone<br />

marrow and repeat RFLP studies are being done to further<br />

investigate this abnormality.<br />

doi:10.1016/j.clim.2007.03.300<br />

F.88 Distribution, Infections, Treatments and<br />

Molecular Analysis in a Large Cohort <strong>of</strong> Patients with<br />

Primary Immunodeficiency Diseases (PIDD) in<br />

Taiwan<br />

Wen-I Lee, Assistant Pr<strong>of</strong>essor, Immunodeficiency Diagnosis<br />

and Research Institute, Chang Gung University and<br />

Children’s Hospital, Taoyuan, China, Tang-Her Jaing,<br />

Assistant Pr<strong>of</strong>essor, Department <strong>of</strong> Pediatric Hematology<br />

and Oncology, Chung Gung Children’s Hospital, Taoyuan,<br />

China, Meng-Ying Hsieh, MD, Department <strong>of</strong> Pediatric<br />

Neurology, Chang Gung University and Children’s Hospital,<br />

Taoyuan, China, Syh-Jae Lin, Associated Pr<strong>of</strong>essor,<br />

Department <strong>of</strong> Pediatric Allergy, <strong>Immunology</strong> and<br />

Rheumatology, Chang Gung University, Taoyuan, China,<br />

Jing-Long Huang, Pr<strong>of</strong>essor, Department <strong>of</strong> Pediatric<br />

Allergy, <strong>Immunology</strong> and Rheumatology, Chang Gung<br />

University, Taoyuan, China, Li-Chen Chen, Assistant<br />

Pr<strong>of</strong>essor, Department <strong>of</strong> Pediatric Allergy, <strong>Immunology</strong> and<br />

Rheumatology, Chang Gung University, Taoyuan, China,<br />

Ming-Ling Kuo, Pr<strong>of</strong>essor, Department <strong>of</strong> Microbiology and<br />

<strong>Immunology</strong>, Graduate Institute <strong>of</strong> Basic Medical Sciences,<br />

Taoyuan, China<br />

One hundred and twenty-four patients (from 120<br />

families) to date diagnosed as primary immunodeficiency<br />

diseases were enrolled from five tertiary medical centers.<br />

The distribution by an update eight categories showed 45<br />

patients (13 females/32 males; 36.3%) with predominant<br />

antibody deficiencies, 27 patients (6/21; 21.8%) with T and<br />

B cell immunodeficiency, 25 patients (9/16; 20.2%) with<br />

congenital defects <strong>of</strong> phagocyte, 25 patients (4/21; 20.2%)<br />

with other well-defined immunodeficiency syndromes, one<br />

boy (0.8%) with disease in immune deregulation (Chediak–<br />

Higashi syndrome) and another with complement 3 deficiency.<br />

None had defects in innate immunity or auto<br />

inflammatory disorders. Pseudomonas and Salmonella spp.<br />

were the two most identified microorganisms in septicemia<br />

(39.7%; 27/68 episodes). Twenty-three patients (18.5%) had<br />

mortality. Stem cell transplantation succeeded in 7 <strong>of</strong> 12<br />

patients. In addition to nine patients with DiGerge<br />

syndrome recognized by FISH, direct sequencing identified<br />

12 unique mutations from 20 families, reflecting distinct<br />

Taiwan geography, although a selection bias may exist. The<br />

study is still ongoing.<br />

doi:10.1016/j.clim.2007.03.301<br />

F.89 Expression <strong>of</strong> FoxP3 and CD25 in T Cells is<br />

Altered in ICOS-/- Common Variable<br />

Immunodeficiency (CVID) patients<br />

Hans Hartmut Peter, Head <strong>of</strong> Department,University<br />

Hospital Freiburg, Freiburg, Germany, Julia Horn, Assistant<br />

Doctor, University Hospital Freiburg, Department <strong>of</strong> <strong>Clinical</strong><br />

<strong>Immunology</strong> and Rheumatology, Freiburg, Germany, Ulrich<br />

Salzer, Senior Postdoctoral Fellow, University Hospital<br />

Freiburg, Department <strong>of</strong> <strong>Clinical</strong> <strong>Immunology</strong> and<br />

Rheumatology, Freiburg, Germany, Jennifer Birmelin, MTA,<br />

Royal Free Hospital Department <strong>of</strong> <strong>Immunology</strong>, London,<br />

England, Klaus Warnatz, Consultant, University Hospital<br />

Freiburg, Department <strong>of</strong> <strong>Clinical</strong> <strong>Immunology</strong> and<br />

Rheumatology, Freiburg, Germany, Michael Schlesier, Head<br />

<strong>of</strong> Laboratory, University Hospital Freiburg, Department <strong>of</strong><br />

<strong>Clinical</strong> Immunolgy and Rheumatology, Freiburg,<br />

Germany, Bodo Grimbacher, Consultant, Royal Free<br />

Hospital, Department <strong>of</strong> <strong>Immunology</strong>,<br />

London, England<br />

Objective: FoxP3+CD25+ high regulatory T cells play a<br />

central role in suppressing immune responses to self<br />

antigens. To determine whether CVID patients have an<br />

altered frequency <strong>of</strong> FoxP3+CD25+ high regulatory T cells<br />

(Tregs), we studied this cell type in 48 CVID patients<br />

including four ICOS−/− CVID patients from the Freiburg<br />

CVID cohort by FACS analysis. Results: First, we found that<br />

the frequency <strong>of</strong> FoxP3+CD25+ Tregs (median +/− SD; p>=<br />

Wilcoxon rank sum test) was not significantly different in<br />

CVID patients (1.9% +/− 0.9%, n = 48; p>= 0.05) as compared<br />

to healthy controls (3.41% +/− 0.9%, n = 17). Second, CVID<br />

patients with autoimmune phenomena (n = 17 <strong>of</strong> 48) do not<br />

have lower frequencies <strong>of</strong> Tregs than CVID patients without<br />

autoimmune phenomena except one patient who presents<br />

with immune thrombocytopenia (ITP). Interestingly, ICOS-/-<br />

CVID patients (n = 4 <strong>of</strong> 48) have normal or increased Treg<br />

frequencies. In addition, in ICOS−/− CVID patients an<br />

increased FoxP3+CD25− T cell population in comparison to<br />

other CVID patients and healthy controls is observed.<br />

However, culturing ICOS−/− CD4+ T cells with IL-2 for 72 h<br />

results in decreased FoxP3 expression (16.4% to 4.68%) <strong>of</strong><br />

CD25- T cells due to activation. Conclusion: Our data suggest<br />

that CVID patients with autoimmune phenomena, do not<br />

have reduced numbers <strong>of</strong> Treg cells. However, ICOS−/− CVID<br />

patients who have no autoimmune phenomena show an<br />

increased FoxP3+CD25+ T cell population and an increased<br />

Foxp3 expression <strong>of</strong> CD25− T cells.<br />

doi:10.1016/j.clim.2007.03.302


Abstracts<br />

F.90 Clusters Differentiation in HIV Patients<br />

According to Measurements <strong>of</strong> T Cells<br />

Subpopulations in Whole Blood After In Vitro<br />

Activation with Mitogen<br />

Marie-Paule Guillaume, Physician, MD, Brugmann Hospital,<br />

Brussels<br />

Absolute counts <strong>of</strong> CD4+ lymphocytes are considered as the<br />

most important surrogate marker for the risk <strong>of</strong> AIDS. However,<br />

contrasts between patient evolutions and their presumed<br />

risk category are not unusual. We formulated the hypothesis<br />

that underlying regulation patterns <strong>of</strong> T cell responses affect<br />

the actual personal outcome despite similar levels <strong>of</strong><br />

circulating CD4+ T cells. We used a standardized PHA stimulation<br />

<strong>of</strong> whole blood cultures as a simplified model <strong>of</strong> T cell<br />

interactions. Relative proportions <strong>of</strong> different subpopulations<br />

were assessed by cyt<strong>of</strong>luorometry, characterized by combined<br />

expressions <strong>of</strong> CD4, CD25, CD8, CD28, and CD69 molecules.<br />

Prospective data from 175 HIV+ patients with different disease<br />

status, and 235 blood donors as controls, were evaluated.<br />

Three different clusters were found: controls and 2 subgroups<br />

<strong>of</strong> patients. Hierarchical analysis <strong>of</strong> these parameters allowed<br />

the provisional definition <strong>of</strong> an algorithm classifying over 95%<br />

<strong>of</strong> the patients according to their cluster. This was based on<br />

levels <strong>of</strong> CD4+25++CD69+ under PHA stimulation, basal levels<br />

<strong>of</strong> CD4+CD25+CD69+ and CD8+CD28negCD69+. Involvement <strong>of</strong><br />

regulatory or suppressive cells was independently confirmed<br />

by expression <strong>of</strong> GITR markers. Repeated measures on 735<br />

successive observations show that patients tend to stick to<br />

their cluster despite changes in CD4 absolute levels or PHA<br />

response in vitro. Confirmation <strong>of</strong> these data would support<br />

the existence <strong>of</strong> different patterns <strong>of</strong> functional immune<br />

regulations in HIV patients.<br />

doi:10.1016/j.clim.2007.03.303<br />

F.91 HIV Treatment Reveals<br />

Hypogammaglobulinemia in an Individual with<br />

Common Variable Immune Deficiency<br />

Lulu Sun, Immunodeficiency Treatment Centre, McGill<br />

University Health Centre, Montreal, QC, Canada,<br />

Christos M Tsoukas, Immunodeficiency Treatment Centre,<br />

McGill University Health Centre, Montreal, QC, Canada<br />

While common variable immune deficiency (CVID) is<br />

characterized by hypogammaglobulinemia, HIV-1 infection<br />

is associated with elevated immunoglobulin levels. There<br />

have been four CVID patients reported in the literature who<br />

recovered antibody production after contracting HIV-1. Here<br />

we describe an HIV patient with concomitant CVID whose<br />

immunoglobulin levels dramatically decreased upon successful<br />

antiretroviral therapy (ART), and investigate a plausible<br />

mechanism by which HIV is able to modulate circulating<br />

immunoglobulin through regulation <strong>of</strong> the B cell activation<br />

factor (BAFF)/BAFF-receptor pathway. Four cohorts were<br />

compared: our HIV/CVID patient, HIV patients, CVID<br />

patients, and healthy controls. Phenotypic and functional<br />

analyses <strong>of</strong> the Tand B cells <strong>of</strong> these cohorts were performed<br />

using flow cytometry, and BAFF serum levels were measured<br />

by ELISA. In addition to marked hypogammaglobulinemia,<br />

the patient has a complete defect <strong>of</strong> memory IgD+/−<br />

CD27+CD19+ B cells that was not altered by ART, consistent<br />

with severe CVID. When viral load briefly increased, a<br />

transient normalization <strong>of</strong> immunoglobulins was also<br />

observed. At the same time, the percentage <strong>of</strong> BAFF-R+ B<br />

cells was boosted from low to normal levels. Plasma BAFF<br />

levels were high compared to controls during viral suppression,<br />

due primarily to the lack <strong>of</strong> memory B cells. Finally, T<br />

cell phenotypes remained abnormal, with a low CD4+ T cell<br />

count that is characteristic <strong>of</strong> rigorous HIV disease, and<br />

impaired in vitro cellular proliferation. Our findings suggest<br />

that high HIV viraemia was required to maintain normal<br />

immunoglobulin levels in the CVID patient, possibly through<br />

upregulating BAFF-R.<br />

doi:10.1016/j.clim.2007.03.304<br />

S45<br />

F.92 Novel IL12RB1 Compound Heterozygous<br />

Mutation in a Brazilian Family with Disseminated<br />

Bacille Calmette-Guérin (BCG) Infection<br />

Joao B. Oliveira, Associate Researcher, Molecular<br />

<strong>Immunology</strong> and Genetics Section, LIM56, USP, São Paulo,<br />

Brazil, Ana Nishiya, Technician, Molecular <strong>Immunology</strong> and<br />

Genetics Section, LIM-56, USP, São Paulo, Brazil, Ludovic de<br />

Beaucoudrey, Researcher, Laboratory <strong>of</strong> Human Genetics <strong>of</strong><br />

Infectious Diseases, University <strong>of</strong> Paris Rene Descartes,<br />

INSERM U550, Paris, Jean-Laurent Casanova, Director,<br />

Laboratoire de Genetique Humaine des Maladies<br />

Infectieuses, INSERM Unite 550, Paris, Anete Grumach,<br />

Associate Pr<strong>of</strong>essor, Primary Immunodeficiency Outpatient<br />

Unit ADEE-3003, LIM-56, USP, São Paulo, Brazil, Alberto J.S.<br />

Duarte, Lab Director, Laboratory <strong>of</strong> Medical Investigation<br />

Unit 56 (LIM-56), University <strong>of</strong> São Paulo, Brazil, São Paulo,<br />

Brazil, Dewton Moraes-Vasconcelos, Associate Pr<strong>of</strong>essor,<br />

2Primary Immunodeficiency Outpatient Unit ADEE-3003,<br />

LIM-56, USP, São Paulo, Brazil<br />

Introduction. Mendelian inheritance to mycobacterial<br />

disease (MSMD) refers to a group <strong>of</strong> rare diseases characterized<br />

by predisposition to clinical disease caused by<br />

environmental non-tuberculous or poorly virulent mycobaterial<br />

species such as Bacille Calmette-Guérin (BCG)<br />

vaccine. Mutations in 5 components <strong>of</strong> the IFN-γ/IL-12 axis<br />

underlie MSMD: IFNR1, IFNR2, STAT1, IL12B and IL12RB1. We<br />

describe here a Brazilian family with a novel compound<br />

heterozygous mutation in IL12RB1. Material and methods.<br />

The 17 exons <strong>of</strong> IL12RB1 and flanking intronic regions were<br />

amplified by PCR, purified and sequenced using an automated<br />

capillary sequencer. Results. LMS, a 2.7-year-old girl,<br />

presented disseminated BCG infection 1 month after<br />

vaccination. Therapeutics was instituted 4 times with<br />

partial response, with relapse <strong>of</strong> the disease immediately<br />

after withdrawal <strong>of</strong> the drugs. She had normal blood cell<br />

counts and lymphoproliferation to mitogens and Candida,<br />

but low response to PPD. Surface expression <strong>of</strong> IL-12 RB1, IL-<br />

12 p40 and IFN-γ R1 was normal. IFN-γ production was low,<br />

with no improvement after stimulation by BCG plus IL-12.<br />

Genomic DNA sequencing <strong>of</strong> IL12RB1 demonstrated a<br />

compound heterozygous pattern, with a substitution in


S46 Abstracts<br />

exon 5 (P173W) and a deletion <strong>of</strong> 17 bp in exon 9. Analysis <strong>of</strong><br />

parents and relatives showed the deletion to be <strong>of</strong> paternal<br />

origin and the substitution <strong>of</strong> maternal origin, but present<br />

also in a maternal uncle. Conclusion. A novel heterozygous<br />

mutation in IL12RB1 causes susceptibility to BCG infection<br />

after vaccination. In suspected cases, mutational screening<br />

should be performed even with normal expression <strong>of</strong> the<br />

IL12Rb1 receptor.<br />

doi:10.1016/j.clim.2007.03.305<br />

F.93 Modulation <strong>of</strong> In Vivo T Cell Activation by an<br />

Acetylcholine-Esterase Inhibitor in Patients<br />

Chronically Infected with HIV<br />

Carlos Cantu-Brito, Investigator, Department <strong>of</strong> Neurology,<br />

Instituto Nacional de Ciencias Medicas y Nutricion SZ,<br />

Mexico City, Mexico, Sergio Ivan Valdes-Ferrer, Resident,<br />

Department <strong>of</strong> Neurology, Instituto Nacional de Ciencias<br />

Medicas y Nutricion SZ, Mexico City, Mexico, Jose Crispin,<br />

PhD Student, Department <strong>of</strong> <strong>Immunology</strong> and<br />

Rheumatology, Instituto Nacional de Ciencias Medicas y<br />

Nutricion SZ, Mexico City, Mexico, Francisco Belaunzaran,<br />

Resident, Department <strong>of</strong> Infectious Diseases, Instituto<br />

Nacional de Ciencias Medicas y Nutricion SZ, Mexico City,<br />

Mexico, Maria Ines Vargas Rojas, PhD Student, Department<br />

<strong>of</strong> <strong>Immunology</strong> and Rheumatology, Instituto Nacional de<br />

Ciencias Medicas y Nutricion SZ, Mexico City, Mexico, Juan<br />

Sierra, Investigator, Department <strong>of</strong> Infectious Diseases,<br />

Instituto Nacional de Ciencias Medicas y Nutricion SZ,<br />

Mexico City, Mexico, Jorge Alcocer Varela, Investigator,<br />

Department <strong>of</strong> <strong>Immunology</strong> and Rheumatology, Instituto<br />

Nacional de Ciencias Medicas y Nutricion SZ, Mexico City,<br />

Mexico<br />

The immune system <strong>of</strong> patients with HIV is overstimulated.<br />

An increased fraction <strong>of</strong> in vivo activated CD4+<br />

Tcells coupled with a decrease in regulatory Tcell numbers is<br />

a cardinal feature commonly observed. Vagal stimulation can<br />

modulate inflammatory response through T cell and macrophages<br />

by down-regulating their production <strong>of</strong> pro-inflammatory<br />

cytokines, namely TNF-α, IL-1, and IL-6. The aim <strong>of</strong><br />

this work was to analyze if the administration <strong>of</strong> an<br />

acetylcholine-esterase (Ach-E) inhibitor is capable <strong>of</strong> diminishing<br />

immune over-stimulation in patients chronically<br />

infected with HIV. Methods: Nineteen HIV-infected patients<br />

with more than 250 CD4+ T cells, devoid <strong>of</strong> antiretroviral<br />

therapy, were assigned to pyridostigmine sulfate 90 mg/day<br />

or matching placebo. In every case, two peripheral blood<br />

samples were obtained: one before and one after 1 week <strong>of</strong><br />

treatment. In vivo activated CD4+ T cells (HLA-DR+, CD69+)<br />

and regulatory T cells (CD4+CD25+Foxp3+) were quantified<br />

with flow cytometry. CD4+ Tcell proliferation was quantified<br />

after stimulation with a polyclonal stimulus (either PHA or<br />

PMA+ionomycin). Results: Nine patients received pyridostigmine,<br />

10 placebo. There were no basal differences<br />

between treatment and placebo groups. In vivo activated<br />

CD4+ T cells diminished significantly (p=0.025) in patients<br />

who received pyridostigmine. Conversely, regulatory T cells<br />

increased in treated patients. Finally, CD4+ T cell proliferation<br />

was significantly lower in patients who received<br />

pyridostigmine (p=0.026). Discussion: We found that the<br />

administration <strong>of</strong> a low dose <strong>of</strong> Ach-E inhibitors can<br />

successfully diminish CD4+ Tcell over-stimulation in patients<br />

with chronic HIV infection.<br />

doi:10.1016/j.clim.2007.03.306<br />

F.94 CD4/CD8 T Cell Ratio Predicts HIV Infection in<br />

Infants: The NHLBI P2C2 HIV Study<br />

William Shearer, Pr<strong>of</strong>essor, Baylor College <strong>of</strong> Medicine,<br />

Texas Children’s Hospital, Pediatrics, Houston, TX, Savita<br />

Pahwa, Pr<strong>of</strong>essor, University <strong>of</strong> Miami, Miller School <strong>of</strong><br />

Medicine, Miami, FL, Jennifer Read, Medical Officer,<br />

National Institute <strong>of</strong> Child Health and Human Development,<br />

Bethesda, MD, Sameera Wijayawardana, Biostatistician<br />

Student, Emory University Rollins School <strong>of</strong> Medicine,<br />

Atlanta, GA, Jing Chen, Assistant Pr<strong>of</strong>essor, Emory<br />

University Rollins School <strong>of</strong> Medicine, Antanta, GA, Kirk<br />

Easley, Senior Associate, Emory University Rollins School <strong>of</strong><br />

Medicine, Atlanta, GA, Paul Palumbo, Pr<strong>of</strong>essor, Dartmouth<br />

Medical School, Hanover, NH, Elaine Abrams, Associate<br />

Pr<strong>of</strong>essor, Columbia University, New York, NY, Steven<br />

Nesheim, Associate Pr<strong>of</strong>essor, Emory University School <strong>of</strong><br />

Medicine, Atlanta, GA<br />

Background: Although CD4 and CD8 T cell counts are<br />

available in many resource-poor settings, HIV DNA PCR tests<br />

are not. We analyzed data from the P2C2 HIV and CDC PACTS<br />

studies (overall HIV transmission rates: 17% and 18%,<br />

respectively) to evaluate the CD4/CD8 T cell ratio as a<br />

predictor <strong>of</strong> HIV infection among HIV-exposed infants.<br />

Methods: Data from the 3-month visit (45–150 days) for<br />

infants born to HIV-infected mothers enrolled in the P2C2<br />

HIV study (79 HIV-infected, 409 uninfected) were analyzed.<br />

The CDC PACTS cohort (224 HIV-infected, 1015<br />

uninfected) was used for validation. Results: The area<br />

under the ROC curve was higher for the CD4/CD8 ratio<br />

compared to the CD4 cell count (AUC=0.83 and 0.75,<br />

P=0.03). The mean CD4/CD8 ratio at the 3-month visit<br />

was 1.7 for HIV-infected infants and 3.0 for uninfected<br />

infants. A CD4/CD8 ratio <strong>of</strong> 2.4 was almost 2.5 times (95%<br />

CI for the likelihood ratio: 2.1–2.9) more likely to occur in<br />

an HIV-infected infant compared to an uninfected infant (test<br />

sensitivity 81%; posttest probability <strong>of</strong> HIV 33%). Model<br />

performance in the CDC PACTS validation sample was equally<br />

good (AUC=0.78 for the CD4/CD8 ratio; good agreement<br />

between the predicted and observed risk <strong>of</strong> HIV). Conclusion:<br />

We conclude that the CD4/CD8 Tcell ratio is a more sensitive<br />

predictor <strong>of</strong> HIV infection in infants than the CD4 Tcell count<br />

and, when necessary, the CD4/CD8 T cell ratio may be used<br />

with caution to diagnose HIV infection.<br />

doi:10.1016/j.clim.2007.03.307<br />

F.95 First Case <strong>of</strong> Human CD21 Deficiency—<br />

Association with Hypogammaglobulinemia<br />

Jens Thiel, MD, University Hospital Freiburg, Freiburg


Abstracts<br />

Complement receptor type 2 (CR2/CD21) is mainly<br />

expressed by mature B cells and FDCs. CD21 functions as<br />

receptor for C3d-opsonized immune complexes and forms a<br />

coreceptor signalling complex together with CD19, and CD81.<br />

We report a case <strong>of</strong> human CD21 deficiency associated with<br />

hypogammaglobulinemia. The 28-year-old patient presented<br />

with flu-like symptoms and reduced serum IgG and IgA.<br />

Leucocytes and lymphocytes were within normal range, as<br />

were the cell counts for T cell subpopulations and CD19+ B<br />

cells. However, class switched CD27+ memory B cells were<br />

reduced and CD21 expression was absent from all patient B<br />

cells. Moreover, no soluble CD21 was detectable in the<br />

patient’s serum, ruling out increased CD21 shedding from the<br />

B cell surface. Complement receptor type 1 was expressed<br />

normally and an immature B cell phenotype could be ruled<br />

out. Thus, the hypogammaglobulinemia and mild immunodeficiency<br />

in this patient may be caused by insufficient<br />

formation <strong>of</strong> switched memory B cell and antibodies. Looking<br />

for the underlying molecular defect, no mutation could be<br />

identified within the coding regions <strong>of</strong> CD21 by genomic<br />

sequencing. CD21 mRNA levels in the patient’s B cells were<br />

not reduced. Sequencing <strong>of</strong> intronic regions adjacent to the<br />

19 exons <strong>of</strong> CD21 revealed a heterozygous point mutation<br />

within the 3′ splice site <strong>of</strong> CD21 exon 6 leading to one<br />

shortened mRNA allele that lacks exon 6. Further experiments<br />

are necessary to test whether this heterozygous splice<br />

site mutation is sufficient to explain the absence <strong>of</strong> CD21 on<br />

the patient’s B cells.<br />

doi:10.1016/j.clim.2007.03.308<br />

F.96 Warning Signs for Primary Immunodeficiences<br />

Investigation in Hospitalized Patients<br />

Anete Sevciovic Grumach, Department <strong>of</strong> Dermatology,<br />

University <strong>of</strong> São Paulo, Brazil, São Paulo, Brazil, Dewton<br />

Moraes-Vasconcelos, Department <strong>of</strong> Dermatology,<br />

University <strong>of</strong> São Paulo, Brazil, São Paulo, Brazil, Joao Bosco<br />

Oliveira, Department <strong>of</strong> Dermatology, University de São<br />

Paulo, Brazil, São Paulo, Brazil, Alexandre Correa, BSc,<br />

Department <strong>of</strong> Dermatology, University <strong>of</strong> São Paulo, Brazil,<br />

São Paulo, Brazil, Rosemeire Navickas Constantino-Silva,<br />

BSc, Department <strong>of</strong> Dermatology, University <strong>of</strong> São Paulo,<br />

Brazil, São Paulo, Brazil, Noemia Orii, BSc, Department <strong>of</strong><br />

Dermatology, University <strong>of</strong> São Paulo, Brazil, São Paulo,<br />

Brazil, Noac Chuffi Barros, Department <strong>of</strong> Dermatology,<br />

University <strong>of</strong> São Paulo, Brazil, São Paulo, Brazil, Maria do<br />

Socorro Ferrao, Department <strong>of</strong> Dermatology, University <strong>of</strong><br />

São Paulo, Brazil, São Paulo, Brazil, Marinella Dellanegra,<br />

Instituto de Infectologia Emilio Ribas, São Paulo, Brazil,<br />

Alberto Jose da Silva Duarte, Pr<strong>of</strong>essor, Department <strong>of</strong><br />

Dermatology, University <strong>of</strong> São Paulo, Brazil, São Paulo,<br />

Brazil<br />

Background: It has been published a higher prevalence <strong>of</strong><br />

PID in hospitalized patients. Nevertheless, most <strong>of</strong> the<br />

recommendations for the identification <strong>of</strong> PID are based on<br />

retrospective reports or in outpatients groups. Objective: To<br />

select the main signs dealing with PID diagnosis based on a<br />

prospective evaluation <strong>of</strong> hospitalized patients. Methodology:<br />

A prospective evaluation <strong>of</strong> inpatients was performed<br />

for a 6 year period. All the patients with clinical symptomatology<br />

not well elucidated were referred to the immunologist.<br />

A comprehensive immunological evaluation was<br />

available and the laboratorial tests were performed according<br />

to the clinical manifestations. Results: 462 patients (1.4<br />

M:1 F) were evaluated and the main symptoms were:<br />

prolonged fever (3.5%), hepatosplenomegaly (2%), uncommon<br />

infections (67%), recurrent infections (4.3%), unexpected<br />

complications (18.2%), suggestive auto-immunity<br />

(3.7%), complications due to vaccines (0.7%), and allergy<br />

(2.2%). PIDs were identified in 18 patients. No PID was<br />

diagnosed among patients complaining <strong>of</strong> prolonged fever<br />

and hepatosplenomegaly. Conclusions: The following criteria<br />

could be used as warning signs for hospitalized patients:<br />

recurrent infections, meningococcal meningitis above 5<br />

years old, complicated infections caused by common pathogens,<br />

uncommon pathogens in HIV negative patients and<br />

vaccinal complications.<br />

doi:10.1016/j.clim.2007.03.309<br />

S47<br />

F.97 Single Cell Evaluation <strong>of</strong> the Production <strong>of</strong><br />

T Cell Cytokines by Chronic Mucocutaneous<br />

Candidiasis Patients<br />

Dewton Moraes Vasconcelos, MD, PhD, Department <strong>of</strong><br />

Dermatology, University <strong>of</strong> São Paulo Medical School, São<br />

Paulo, Brazil, Alexandre de Almeida, MD, MSc, Department<br />

<strong>of</strong> Dermatology, University <strong>of</strong> São Paulo Medical School, São<br />

Paulo, Brazil, Dewton Moraes Vasconcelos, MD, PhD,<br />

Department <strong>of</strong> Dermatology, University <strong>of</strong> São Paulo<br />

Medical School, São Paulo, Brazil, Anete S. Grumach, MD,<br />

PhD, Department <strong>of</strong> Dermatology, University <strong>of</strong> São Paulo<br />

Medical School, São Paulo, Brazil, Mauricio Domingues<br />

Ferreira, MD, PhD, Department <strong>of</strong> Dermatology, University<br />

<strong>of</strong> São Paulo Medical School, São Paulo, Brazil, Tatiana Negri<br />

Santi, BSc, Department <strong>of</strong> Dermatology, University <strong>of</strong> São<br />

Paulo Medical School, São Paulo, Brazil, Alberto Jose da<br />

Silva Duarte, MD, PhD, Department <strong>of</strong> Dermatology,<br />

University <strong>of</strong> São Paulo Medical School, São Paulo, Brazil<br />

Background: Chronic mucocutaneous candidiasis (CMC) is<br />

a rare primary immune deficiency characterized by chronic<br />

or recurrent infection by Candida in mucous membranes,<br />

nails and skin. Impaired cell mediated immunity to Candida<br />

as well as dysregulation <strong>of</strong> the production <strong>of</strong> Th1 and Th2<br />

cytokines is an important mechanism in CMC. Objective:<br />

Evaluate the number <strong>of</strong> Th1 (IL-2, IFN-γ) and Th2 (IL-10, IL-<br />

4) cytokines producing cells <strong>of</strong> a group <strong>of</strong> patients with CMC<br />

to understand the relationship <strong>of</strong> these cytokines and CMC.<br />

Methods: We studied the production <strong>of</strong> cytokines <strong>of</strong> 15<br />

patients with CMC and 13 controls. We evaluated the<br />

production <strong>of</strong> IL-2, IL-4, IL-10 and IFN-γ by total T cells, CD4<br />

+ and CD8+ subsets by intracellular cytokine flow cytometry.<br />

Results: The patients presented lower number <strong>of</strong> IL-2, IFNγ,<br />

IL-10 and IL-4 producing CD4+ cells than controls, when<br />

stimulated with Candida and tetanus toxoid. The patients’<br />

presented less CD8+ T cells producing Th1 cytokines when<br />

compared to controls. There were two distinct subgroups <strong>of</strong><br />

patients. One presented less IL-2 and IFN-γ producing T cells<br />

than the other. The second subgroup presented a lower


S48 Abstracts<br />

number <strong>of</strong> IFN-γ and a similar number <strong>of</strong> IL-2 producing T<br />

cells compared to control group. Conclusion: Our patients<br />

presented a decreased number <strong>of</strong> T cells producing both Th1<br />

and Th2 cytokines. Despite the presence <strong>of</strong> 2 different<br />

groups in relation to cytokine production there was no<br />

correlation to the clinical features.<br />

doi:10.1016/j.clim.2007.03.310<br />

F.98 Immunomodulation by Cytokines in Chronic<br />

Mucocutaneous Candidiasis Patients: Evaluation <strong>of</strong><br />

the Production <strong>of</strong> T Cell Cytokines at a Single Cell<br />

Level<br />

Mauricio Domingues Ferreira, MD, PhD, Department <strong>of</strong><br />

Dermatology, University <strong>of</strong> São Paulo Medical School, São<br />

Paulo, Brazil, Alberto Jose da Silva Duarte, MD, PhD,<br />

Department <strong>of</strong> Dermatology, University <strong>of</strong> São Paulo<br />

Medical School, São Paulo, Brazil, Dewton Moraes<br />

Vasconcelos, MD, PhD, Department <strong>of</strong> Dermatology,<br />

University <strong>of</strong> São Paulo Medical School, São Paulo, Brazil,<br />

Tatiana Negri Santi-BSc, Department <strong>of</strong> Dermatology,<br />

University <strong>of</strong> São Paulo Medical School, São Paulo, Brazil,<br />

Anete S. Grumach, MD, PhD, Department <strong>of</strong> Dermatology,<br />

University <strong>of</strong> São Paulo Medical School, São Paulo, Brazil,<br />

Alexandre Almeida, MD, MSc, Department <strong>of</strong> Dermatology,<br />

University <strong>of</strong> São Paulo Medical School, São Paulo, Brazil<br />

Background: Chronic mucocutaneous candidiasis (CMC)<br />

is a rare primary immune deficiency characterized by<br />

chronic or recurrent Candida infection in mucous membranes,<br />

nails and skin. Impaired cell mediated response<br />

and dysregulation <strong>of</strong> the production <strong>of</strong> Th1 and Th2<br />

cytokines to Candida are important in CMC. We previously<br />

found a lower number <strong>of</strong> IL-2, IL-10, IL-4 and IFN-γ<br />

producing T cells and its CD4+ and CD8+ subsets compared<br />

to the control group when stimulated with Candida and<br />

tetanus toxoid. Objective: Modulate the T cell response by<br />

adding to the cell culture medium IL-2, or IL-12, or anti-IL-<br />

10. Methods: We studied the production <strong>of</strong> cytokines <strong>of</strong> 15<br />

patients with CMC and 13 controls stimulated by Candida<br />

and tetanus toxoid. We evaluated IL-2, IL-4, IL-10 and IFN-γ<br />

in total T cells, as well as CD4+ and CD8+ subsets by an<br />

intracellular cytokine flow cytometric assay. We tried to<br />

immunomodulate with recombinant human IL-2, or IL-12, or<br />

a monoclonal antibody anti-IL-10. Results: We did not find a<br />

significant response to the immunomodulation trials, but<br />

there was a trend <strong>of</strong> improvement in the synthesis <strong>of</strong> IL-2<br />

from the CD4+ cells <strong>of</strong> patients when stimulated by Candida<br />

and modulated by IL-2, IL-12 or anti-IL-10. Conclusion:<br />

There was no significant improvement by the immunomodulation<br />

by cytokines, but there was a tendency <strong>of</strong><br />

improvement. It is possible that if we have used a<br />

combination <strong>of</strong> cytokines, the results could be better. Our<br />

data suggest a new possible way <strong>of</strong> treatment to patients<br />

with CMC.<br />

doi:10.1016/j.clim.2007.03.311<br />

F.99 Hereditary Angiodema (HAE) in Brazil: Registry<br />

<strong>of</strong> 100 Cases<br />

Anete Sevciovic Grumach, Department <strong>of</strong> Dermatology,<br />

University <strong>of</strong> São Paulo, Brazil, São Paulo, Brazil, Jorge<br />

Andrade Pinto, Department <strong>of</strong> Pediatrics, Federal<br />

University <strong>of</strong> Minas Gerais, Belo Horizonte, Alexandre Pires<br />

Correa, BSc, Department <strong>of</strong> Dermatology, University <strong>of</strong><br />

São Paulo, Brazil, São Paulo, Brazil, Dewton<br />

Moraes-Vasconcelos, MD, PhD, Department <strong>of</strong> Dermatology,<br />

University <strong>of</strong> São Paulo, Brazil, São Paulo, Brazil, Rosemeire<br />

Navickas Constabtino-Silva, BSc, Department <strong>of</strong><br />

Dermatology, University <strong>of</strong> São Paulo, Brazil, São Paulo,<br />

Brazil, Sollange Valle, Federal University <strong>of</strong> Rio de Janeiro,<br />

Rio de Janeiro, Eli Mansour, Department <strong>of</strong> Pediatrics,<br />

University <strong>of</strong> Campinas, Campinas, Maria Marluce Vilela,<br />

Department <strong>of</strong> Pediatrics, University <strong>of</strong> Campinas,<br />

Campinas, Ricardo Zollner, <strong>Clinical</strong> Medicine, University <strong>of</strong><br />

Campinas, Campinas, Alberto Jose da Silva Duarte,<br />

Pr<strong>of</strong>essor, Department <strong>of</strong> Dermatology, University <strong>of</strong> São<br />

Paulo, Brazil, São Paulo, Brazil, Evandro Rivitti, Pr<strong>of</strong>essor,<br />

Department <strong>of</strong> Dermatology, University <strong>of</strong> São Paulo, Brazil,<br />

São Paulo, Brazil<br />

HAE is a primary quantitative or functional defect <strong>of</strong> C1<br />

inhibitor. <strong>Clinical</strong> manifestations include subcutaneous,<br />

respiratory and gastrointestinal edema. Asphyxia could<br />

occur in 25–40% <strong>of</strong> the non-treated cases. There was no<br />

registry in Brazil until now. Objective: Report the clinical and<br />

laboratorial characteristics <strong>of</strong> HAE in Brazilians. Methods:<br />

Collaborative work groups were established among specialized<br />

services. The following parameters were evaluated:<br />

gender, age, age <strong>of</strong> diagnosis, main clinical manifestations,<br />

time for diagnosis, triggering factor, treatment, familial<br />

history and C1 inhibitor and C4 levels and CH50. Laboratorial<br />

diagnosis <strong>of</strong> C1 INH deficiency was confirmed in all patients.<br />

Results: HAE affected 67 F:33 M, 1–70 years <strong>of</strong> age and the<br />

first symptoms were reported during childhood in most <strong>of</strong> the<br />

cases. One episode <strong>of</strong> laryngeal edema was present in 17%<br />

and the triggering factors were trauma (31/100), stress (11/<br />

100), and menses (6/100). Familial history was positive in<br />

53/100. Conclusion: Familial history was determinant for<br />

diagnosis in half <strong>of</strong> the cases. Late diagnosis had been<br />

established but the first symptoms occurred during childhood.<br />

HAE diagnosis is still not well recognized in our<br />

country.<br />

doi:10.1016/j.clim.2007.03.312<br />

F.100 Autosomal Dominant Combined Immune<br />

Deficiency: A Unique Family with Absent B-Cells and<br />

Decreased T-Cells<br />

Michael Land, Physician, University <strong>of</strong> California, Los<br />

Angeles, Pediatrics, Los Angeles, CA, Raffi Tachdjian,<br />

Physician, University <strong>of</strong> California, Los Angeles, Pediatrics,<br />

Los Angeles, CA, Erina Lin, Physician, University <strong>of</strong><br />

California, Los Angeles, Pediatrics, Los Angeles, CA, Robert<br />

Roberts, Physician, University <strong>of</strong> California, Los Angeles,<br />

Pediatrics, Los Angeles, CA, Marc Riedl, Physician,<br />

University <strong>of</strong> California, Los Angeles, Department <strong>of</strong><br />

Medicine, Los Angeles, CA


Abstracts<br />

Rationale: We present a family <strong>of</strong> four individuals with<br />

immunodeficiency. The father and daughter were initially<br />

diagnosed with Common Variable Immune Deficiency (CVID)<br />

and were found to have absent B cells and low T cells. The<br />

father’s brother and mother also had immunodeficiency but<br />

died years ago. This may represent a unique form <strong>of</strong><br />

Combined Immunodeficiency.<br />

Methods/Results: The father is a 35-year-old man with a<br />

history <strong>of</strong> CVID diagnosed at age 12 following multiple upper<br />

respiratory infections. He was started on intravenous<br />

immunoglobulin (IVIG) for hypogammaglobulinemia, but<br />

never had any severe infections. His mother and older<br />

brother also received gamma globulin for presumed CVID,<br />

but both died from infections at age 36. He had one episode<br />

<strong>of</strong> pneumonia at age 19, with occasional sinus infections. His<br />

trough IgG level recently was 162, but increased to 724 with<br />

higher dosing. His IgA was b7, IgM b0.5, IgE b0.1. The<br />

patient’s 3-year-old daughter has been receiving IVIG since<br />

age 8 months for hypogammaglobulinemia. She has a history<br />

<strong>of</strong> sinusitis and has never been hospitalized. Her IgA is b7,<br />

IgM 6, IgE 0.1. Interestingly, both father and daughter have<br />

undetectable B cells (CD19 b1%) and CD4 lymphopenia<br />

(father: 202, daughter: 608). Both patients also have low<br />

CD8 counts (father: 162, daughter: 189). They have normal<br />

responses to mitogens and antigens. Conclusion: This family,<br />

along with the deceased relatives, exhibits an unusual<br />

pattern <strong>of</strong> inheritance for B cell and T cell deficiency<br />

consistent with combined immunodeficiency, suggesting<br />

autosomal dominance. Further characterization and investigation<br />

<strong>of</strong> this disease are warranted.<br />

doi:10.1016/j.clim.2007.03.313<br />

F.101 Circulating Class-Switched Memory B Cells<br />

are Markedly Reduced in Some Patients with<br />

Specific Antibody Deficiency (SAD) and/or Mild IgG<br />

Subclass Deficiency<br />

Lily E. Leiva, Associate Pr<strong>of</strong>essor, Louisiana State University<br />

Health Sciences Center and Children’s Hospital, Department<br />

<strong>of</strong> Pediatrics, New Orleans, LA, Kenneth Paris, Assistant<br />

Pr<strong>of</strong>essor, Louisiana State University Health Sciences Center<br />

and Children’s Hospital, Department <strong>of</strong> Pediatrics, New<br />

Orleans, LA, Hanh Monjure, Research Associate, Louisiana<br />

State University Health Sciences Center and Children’s<br />

Hospital, Department <strong>of</strong> Pediatrics, New Orleans, LA,<br />

Ricardo U. Sorensen, Pr<strong>of</strong>essor and Chair, Louisiana State<br />

University Health Sciences Center and Children’s Hospital,<br />

Department <strong>of</strong> Pediatrics, New Orleans, LA, Dana Minor,<br />

Research Technician, Research Institute for Children,<br />

Children’s Hospital, New Orleans, LA<br />

Some patients with recurrent respiratory infections are<br />

affected by specific antibody deficiency (SAD) and/or IgG<br />

subclass deficiencies. In this study we investigated if the<br />

inability <strong>of</strong> patients with SAD and/or IgG subclass deficiency to<br />

respond to pneumococcal polysaccharide antigens (PP) was<br />

associated with reduced numbers <strong>of</strong> circulating class-switched<br />

memory B cells. We studied 7 children with recurrent respiratory<br />

infections with SAD and/or mild IgG subclass deficiency and 8<br />

healthy controls. All patients were evaluated with total<br />

immunoglobulin and IgG subclass concentrations. We measured<br />

IgG and IgM anti-PP antibody levels by a standardized ELISA test.<br />

Purified PBMC were stained for the expression <strong>of</strong> CD27 and IgD<br />

on CD19+ B cells. Percentages <strong>of</strong> naïve (CD27NIgD+), nonswitched<br />

memory (CD27+IgD+) and class-switched memory<br />

(CD27+IgDN) B cells were determined by flow cytometry.<br />

Results: In patients in whom IgG anti-PP antibody levels were<br />

low, IgM levels were within the normal ranges for each <strong>of</strong> the 8<br />

serotypes tested. The B cell compartment in these patients<br />

showed a marked reduction in the percentage <strong>of</strong> class-switched<br />

memory B cells in 2 patients (6% and 2%, respectively) in<br />

comparison to the other 5 patients (range: 32%–90%) and to<br />

controls (range: 22%–87%). These results suggest that a deficient<br />

IgM to IgG antibody switch is associated with a lack <strong>of</strong> circulating<br />

class-switched memory B cells in some patients, and that this<br />

may be the explanation for the failure <strong>of</strong> some patients to<br />

respond to PP. Studies are in progress to determine the<br />

prognostic implications <strong>of</strong> these differences.<br />

doi:10.1016/j.clim.2007.03.314<br />

F.102 HIV Treatment Reveals<br />

Hypogammaglobulinemia in an Individual with<br />

Common Variable Immune Deficiency<br />

Lulu Sun, Immunodeficiency Treatment Centre, McGill<br />

University Health Centre, Montreal, QC, Canada, Christos<br />

M. Tsoukas, Immunodeficiency Treatment Centre, McGill<br />

University Health Centre, Montreal, QC, Canada<br />

While common variable immune deficiency (CVID) is characterized<br />

by hypogammaglobulinemia, HIV-1 infection is associated<br />

with elevated immunoglobulin levels. There have been<br />

four CVID patients reported in the literature who recovered<br />

antibody production after contracting HIV-1. Here we describe<br />

an HIV patient with concomitant CVID whose immunoglobulin<br />

levels dramatically decreased upon successful antiretroviral<br />

therapy (ART), and investigate a plausible mechanism by which<br />

HIV is able to modulate circulating immunoglobulin through<br />

regulation <strong>of</strong> the B cell activation factor (BAFF)/BAFF-receptor<br />

pathway. Four cohorts were compared: our HIV/CVID patient,<br />

HIV patients, CVID patients, and healthy controls. Phenotypic<br />

and functional analyses <strong>of</strong> the Tand B cells <strong>of</strong> these cohorts were<br />

performed using flow cytometry, and BAFF serum levels were<br />

measured by ELISA. In addition to marked hypogammaglobulinemia,<br />

the patient has a complete defect <strong>of</strong> memory IgD+/−<br />

CD27+ CD19+ B cells that was not altered by ART, consistent with<br />

severe CVID. When viral load briefly increased, a transient<br />

normalization <strong>of</strong> immunoglobulins was also observed. At the<br />

same time, the percentage <strong>of</strong> BAFF-R+B cells was boosted from<br />

low to normal levels. Plasma BAFF levels were high compared to<br />

controls during viral suppression, due primarily to the lack <strong>of</strong><br />

memory B cells. Finally, T cell phenotypes remained abnormal,<br />

with a low CD4+ Tcell count that is characteristic <strong>of</strong> rigorous HIV<br />

disease, and impaired in vitro cellular proliferation. Our findings<br />

suggest that high HIV viraemia was required to maintain normal<br />

immunoglobulin levels in the CVID patient, possibly through<br />

upregulating BAFF-R.<br />

doi:10.1016/j.clim.2007.03.315<br />

S49


S50 Abstracts<br />

F.103 Differences in Telomerase Expression by the<br />

CD1a+ Cells in Langerhans Cell Histiocytosis Reflects<br />

the Diverse <strong>Clinical</strong> Presentation <strong>of</strong> the Disease<br />

Cristiana Costa, PhD Student, Leiden University Medical<br />

Center, Department <strong>of</strong> Pediatrics, Leiden, The Netherlands,<br />

Maarten Egeler, MD PhD, Leiden University Medical Center,<br />

Leiden, The Netherlands, Manja Hoogeboom, Leiden<br />

University Medical Center, Leiden, The Netherlands, Ramses<br />

Forsyth, N. Goormaghtigh Institute <strong>of</strong> Pathology, University<br />

Hospital Gent, Gent, Karoly Szuhai, PhD, Leiden University<br />

Medical Center, Leiden, The Netherlands, Marieke Niesters,<br />

Leiden University Medical Center, Department <strong>of</strong> Pediatrics,<br />

Leiden, The Netherlands, Ronald de Krijger, Erasmus<br />

Medical Center, Department <strong>of</strong> Pathology, Rotterdam,<br />

Abdellatif Tati, Service de Pneumonologie, Hospital Saint<br />

Louis, Paris, Pancras Hogendoorn, Leiden University Medical<br />

Center, Department <strong>of</strong> Pathology, Leiden, The Netherlands,<br />

Nicola Annels, PhD, Leiden University Medical Center,<br />

Department <strong>of</strong> Pediatrics, Leiden, The Netherlands<br />

Langerhans cell histiocytosis (LCH) is a disease characterised<br />

by an uncontrolled clonal proliferation <strong>of</strong> Langerhans<br />

cells, whose aetiology is still unclear. The clonal nature <strong>of</strong><br />

LCH could support the hypothesis that it is a neoplastic<br />

disease with unlimited growth potential. One requirement<br />

for unlimited proliferation is the maintenance <strong>of</strong> telomere<br />

length. In a group <strong>of</strong> 70 patients we investigated whether a<br />

telomere maintenance mechanism is active by LCH cells.<br />

Results showed that LCH cells from all restricted skin LCH<br />

lesions (6/6) expressed telomerase as assessed by human<br />

telomere reverse transcriptase (hTERT) immunohistochemistry,<br />

whereas LCH cells from the majority <strong>of</strong> bone lesions<br />

analysed did not (26/34). Interestingly, in contrast to solitary<br />

bone lesions, LCH cells from lesions <strong>of</strong> multisystem patients<br />

always expressed telomerase (11/11), regardless <strong>of</strong> the<br />

lesional site. In situ telomeric repeat amplification protocol<br />

(TRAP) assay performed on different lesional sites showed<br />

that telomerase was active. In addition, the telomere length<br />

<strong>of</strong> LCH cells from a hTERT positive skin multisystem lesion<br />

was long and homogeneous when compared to the LCH cells<br />

from hTERT negative bone single system LCH lesions, which<br />

were heterogeneous in length. No evidence for an alternative<br />

lengthening <strong>of</strong> telomeres mechanism was found in<br />

hTERT negative lesions. The difference in telomerase<br />

expression and telomere length in the different lesional<br />

sites and in biopsies from patients with solitary versus<br />

multisystem disease appears to be reflective <strong>of</strong> the diverse<br />

clinical presentation and course <strong>of</strong> LCH. The results from this<br />

study have important implications for understanding the<br />

nature <strong>of</strong> this disease.<br />

doi:10.1016/j.clim.2007.03.316<br />

F.104 Multiplex Capabilities for Serological<br />

Immunoassays Using Luminex xMAP ® Technology<br />

Harold Baker, Senior Scientist, Luminex Corporation,<br />

Austin, TX<br />

The Luminex xMAP technology incorporates fluorescently<br />

encoded microspheres (xMAP microspheres) to perform<br />

multiplexed bioassays in conjunction with advanced digital<br />

signal processing and proprietary identification techniques.<br />

The advantage <strong>of</strong> the multiplexing technology is the<br />

capability to measure several antigens within one sample.<br />

The combination <strong>of</strong> multiple results and reduced sample<br />

volume increases the efficiency for testing and sample<br />

consumption. Another advantage is that the serological<br />

assays can be multiplexed with other immunoassays. Five<br />

specific antigens were coupled to xMAP microspheres<br />

representing several individual regions. BSA coated microspheres<br />

were prepared to function as internal assay controls<br />

for non-specific reactivity. A total <strong>of</strong> five regions were used<br />

with each antigen to develop a 30-plex assay. The xMAP<br />

serological assay format we used was a 60 minute, no wash<br />

assay. Diluted patient serum, 10 ƒÝL, was combined with the<br />

microsphere mixture, 50 ƒÝL, and incubated 30 min. Diluted<br />

PE conjugated antibody, 150 ƒÝL, was added and incubated<br />

30 min before being analyzed with the Luminex system.<br />

Equivalent results were obtained with the replicate antigen<br />

coated regions. Analytical sensitivity was defined by the<br />

maximum dilution before an appreciable decrement in MFI<br />

response was observed. This was observed to be at a dilution<br />

<strong>of</strong> 1:1000 representing the use <strong>of</strong> 0.01 ƒÝL serum for the<br />

measurement <strong>of</strong> antibodies in the serum sample. These<br />

results demonstrate that the Luminex xMAP technology can<br />

be used to develop multiplex assays such as those required<br />

for serological assessment <strong>of</strong> autoimmune diseases.<br />

doi:10.1016/j.clim.2007.03.319<br />

F.105 Immun<strong>of</strong>luorometrical Exploring <strong>of</strong> FSH<br />

Levels in the Serum <strong>of</strong> Patients with Histologically<br />

Verified Macrocellular Lung Cancer<br />

Ivan Milosevic, Primary Care Physician, Dispensary 2, ZZZZR,<br />

Kragujevac<br />

It is already known in that the cells <strong>of</strong> macrocellular lung<br />

cancer can produce gonadotropins. By following the levels <strong>of</strong><br />

FSH as one <strong>of</strong> gonadotropins, in the serum <strong>of</strong> macrocellular<br />

lung cancer patients, we could make certain statistical<br />

conclusions how significant these levels would be in eventual<br />

future early diagnostic procedures, besides already existing<br />

tumor markers and other methods. This work would connect<br />

the Oncology, Endocrinology and <strong>Immunology</strong> fields, containing<br />

very interesting immun<strong>of</strong>luorometrical procedures,<br />

hormonal theories and statistical estimates. As an immunological<br />

part in this theoretical model, it is represented<br />

through figure, time-resolved fluoroimmunoassay for quantitative<br />

detection <strong>of</strong> FSH. By following <strong>of</strong> FSH levels in the<br />

serum <strong>of</strong> microcellular lung cancer patients and making<br />

determinate statistical conclusions about its importance,<br />

that procedure could be eventually used as one <strong>of</strong> early or<br />

advanced diagnostic methods concerning mentioned disease.<br />

What would we get with that? The histological verification is<br />

very slow and painful, when the macrocellular lung cancer is<br />

in the question. This histological type gives the metastases<br />

rapidly and it needs to be diagnosed in the fastest possible<br />

way in order to be prevented by adequate reaction and<br />

therapy. At the same time the biopsy as a very invasive<br />

method would be avoided. When the tumor markers are in


Abstracts<br />

question as a diagnostic method supplemented and supported<br />

by the FSH levels exploring could be more reliable.<br />

Concerning all above presented, in future establishing <strong>of</strong> this<br />

procedure, we would get a rapid, comfortable and safe<br />

diagnostic method.<br />

doi:10.1016/j.clim.2007.03.320<br />

F.108 Distinct Regulation <strong>of</strong> Individual Tyrosine<br />

Phosphorylation <strong>of</strong> the Cytoplasmic Domain <strong>of</strong> CD19<br />

Nobuko Ishiura, MD, Department <strong>of</strong> Dermatology, Graduate<br />

School <strong>of</strong> Medicine, University <strong>of</strong> Tokyo, Tokyo, Japan,<br />

Manabu Fujimoto, PhD, Department <strong>of</strong> Dermatology,<br />

Kanazawa University Graduate School <strong>of</strong> Medical Science,<br />

Kanazawa Ishikawa, Japan, Kunihiko Tamaki, PhD,<br />

Department <strong>of</strong> Dermatology, Graduate School <strong>of</strong> Medicine,<br />

University <strong>of</strong> Tokyo, Tokyo, Japan<br />

Cell surface molecules called co-receptors on lymphocytes<br />

positively or negatively modulate the antigen receptor<br />

signaling. CD19 is a B lymphocyte-specific co-receptor which<br />

regulates constitutive thresholds and enhances antigen<br />

receptor-induced signaling through its cytoplasmic domain.<br />

The cytoplasmic region <strong>of</strong> CD19 contains nine conserved<br />

tyrosine residues, which become phosphorylated upon B cell<br />

activation. Tyrosine phosphorylation <strong>of</strong> CD19 mediates<br />

transmembrane signals by recruiting multiple signaling<br />

molecules including Src family protein tyrosine kinases<br />

(PTKs), especially Lyn, Vav, and PI 3-kinase. Of nine tyrosine<br />

residues, Y513, Y482, and Y391 are supposed to play critical<br />

roles in CD19-mediated signal transduction, although the<br />

precise regulation <strong>of</strong> individual tyrosine phosphorylation<br />

remains unknown. Herein, we have developed phosphospecific<br />

antibodies against these tyrosine residues, CD19pY513,<br />

CD19-pY482 and CD19-pY391, and have assessed the<br />

regulation <strong>of</strong> tyrosine phosphorylation <strong>of</strong> CD19 following<br />

various stimulations. BCR ligation induced Y513 phosphorylation<br />

preceded by Y482 and Y391 phosphorylation, suggesting<br />

Y513 as the primary phosphorylation site.<br />

Phosphorylations <strong>of</strong> three tyrosine residues were much<br />

enhanced and prolonged by simultaneous stimulation <strong>of</strong><br />

BCR and CD40 ligation. Intriguingly, CD19 containing phosphorylated<br />

Y513 was exclusively located within the lipid rafts<br />

following the BCR ligation, while majority <strong>of</strong> CD19 containing<br />

phosphorylated Y482 and Y391 stayed out <strong>of</strong> the lipid rafts.<br />

Therefore, individual tyrosines <strong>of</strong> CD19 undergo distinct<br />

regulation <strong>of</strong> phosphorylation, which in turn determines<br />

different distribution on the cell surface.<br />

doi:10.1016/j.clim.2007.03.321<br />

F.109 Vitamin D Receptor Activators Calcitriol and<br />

Paricalcitol Modulate Dendritic Cell Phenotype and<br />

Function In Vitro Study<br />

Klara Sochorova, Pharm Department <strong>of</strong> <strong>Immunology</strong>, 2nd<br />

Medical School, Charles University Prague, Prague, Vit<br />

Budinsky, Manager. Department <strong>of</strong> immunology, 2nd Medical<br />

School, Charles University, Prague, Zuzana Tobiasova,<br />

Manager. Department <strong>of</strong> <strong>Immunology</strong>, 2nd Medical School,<br />

Charles University, Prague, Jirina Bartunkova, Pr<strong>of</strong>essor,<br />

Department <strong>of</strong> <strong>Immunology</strong>, 2nd Medical School, Charles<br />

University, Prague, Sylva Dusilova Sulkova, Pr<strong>of</strong>essor,<br />

Departments <strong>of</strong> Gerontology and Metabolism, Medical<br />

School and Teaching Hospital, Hradec Kralove<br />

Vitamin D is known as an important immunomodulatory<br />

agent. Its potential to suppress immune response in autoimmunity<br />

is intensively studied. Use <strong>of</strong> active metabolite <strong>of</strong><br />

vit. D3, calcitriol, in vivo, however, is limited due to its<br />

hypercalcemic effect. Paricalcitol, the synthetic analogue <strong>of</strong><br />

calcitriol, is characterized by reduced hypercalcemic effect,<br />

but its immunosupressive properties have not been evaluated<br />

yet. In our study we compared the effect <strong>of</strong> calcitriol and<br />

paricalcitol on morphological and functional characteristics<br />

<strong>of</strong> dendritic cells (DC) in vitro. DC were differentiated from<br />

monocytes with IL4 and GM-CSF for 5 days and then induced to<br />

maturate with viral or bacterial products (poly-IC, LPS).<br />

Calcitriol or paricalcitol were added during the period <strong>of</strong><br />

differentiation or maturation. We studied the expression <strong>of</strong><br />

DC-relevant surface molecules. DC function was tested by<br />

endocytosis, T-stimulatory capacity and cytokine production.<br />

DC differentiated in the presence <strong>of</strong> calcitriol or paricalcitol<br />

remain at an immature state (low expression <strong>of</strong> CD80, CD83,<br />

CD86 and HLA-DR after stimulation by both poly-IC and LPS,<br />

high expression <strong>of</strong> CD14 and PD-L1, high endocytic activity,<br />

low T cell activation and low IL-6 and TNF production). DC<br />

maturated in the presence <strong>of</strong> both drugs were also impaired,<br />

but less than in previous case. The immunosuppressive effect<br />

<strong>of</strong> both calcitriol and paricalcitol on DC is comparable.<br />

Paricalcitol thus seems to be a perspective drug for<br />

influencing the pathological immune response in autoimmunity.<br />

This project was supported by MSM 0021620812.<br />

doi:10.1016/j.clim.2007.03.322<br />

S51<br />

F.110 Significance <strong>of</strong> IgG4 in the Diagnosis <strong>of</strong> Mucous<br />

Membrane Pemphigoid<br />

Vijay Kumar, Director, IMMCO Diagnostics Inc., Buffalo, NY,<br />

Lakshmanan Suresh, IMMCO Diagnostics Inc., Buffalo, NY<br />

Background: Mucous membrane pemphigoid (MMP) is an<br />

immune mediated vesiculo-bullous disorder predominantly<br />

affecting the mucous membranes <strong>of</strong> the oropharynx. Direct<br />

immun<strong>of</strong>luorescence (IF) studies <strong>of</strong> the presence <strong>of</strong> immunoglobulins<br />

and/or complement are considered as the gold<br />

standard in the diagnosis <strong>of</strong> MMP. The tissue-bound IgG and<br />

other immunoreactants are seen as a linear band in the<br />

basement membrane zone (BMZ). However immunodeposition<br />

<strong>of</strong> IgG and/or C3 is not observed in all cases <strong>of</strong> MMP. One <strong>of</strong> the<br />

reasons <strong>of</strong> this lack <strong>of</strong> immune deposits may be that certain IgG<br />

subclass <strong>of</strong> immunoglobulins may be the autoreactants and that<br />

the total IgG conjugates fail to detect this subclass <strong>of</strong> IgG.<br />

Objective: The purpose <strong>of</strong> our study was to determine the<br />

diagnostic value and frequency <strong>of</strong> tissue deposition <strong>of</strong> IgG4 in<br />

comparison to polyclonal IgG, other immunoglobulins and C3.<br />

Methods: <strong>Oral</strong> mucosal biopsies <strong>of</strong> 82 patients clinically<br />

suspected to have MMP were analyzed by direct IF using<br />

polyclonal anti-human IgG, IgM, IgA, fibrin, complement C3,<br />

and anti-human IgG4 subclass monoclonal antibodies. Results:


S52 Abstracts<br />

Based on H&E and direct IF studies, 34 cases were diagnosed as<br />

MMP. The most common antibody deposited was IgG (90%)<br />

followed by C3 (82%) and IgG4 (71%). Strikingly, IgG4 was the<br />

sole antibody detected in two cases (6%). Conclusion: Our<br />

results suggest that the use <strong>of</strong> monoclonal IgG4 is important in<br />

the diagnosis <strong>of</strong> MMP. We suggest adding monoclonal IgG4 to the<br />

routine panel <strong>of</strong> antibodies used in studies <strong>of</strong> cases suspected to<br />

have MMP to avoid false-negatives.<br />

doi:10.1016/j.clim.2007.03.324<br />

F.111 Immuophenotyping <strong>of</strong> PBL and B.M Markers in<br />

Patients with Leukemia by Flowcytometry<br />

Fereshteh Alesahebfosoul, Academic Member <strong>of</strong> Isfahan<br />

University, Isfahan University Medical School, <strong>Immunology</strong><br />

Department, Isfahan<br />

Introduction: Peripheral blood lymphocytes (PBL) and bone<br />

marrow (B.M.) immunophenotyping is widely used for diagnosis<br />

and prognosis <strong>of</strong> leukemia. Flow cytometry technique provided<br />

facilities to assess the frequency <strong>of</strong> malignant cell population<br />

and the protein expression in each group <strong>of</strong> cells which have<br />

been gated. Method and material: Fresh peripheral blood and<br />

bone marrow <strong>of</strong> patient were collected and tested for following<br />

examination: total white blood cells and leukocyte percentage<br />

rate were determined by H1. Machine: The samples were<br />

treatedwithconjugatedMoAbwithFITCand/orphycoErythrine<br />

and then dual staining <strong>of</strong> cells was performed. The following<br />

monoclonal antibodies were used: CD10, CD19, CD20, CD5,<br />

CD3, CD22, CD7, CD33, anti-HLA DR, and CD13. After direct<br />

staining <strong>of</strong> cell samples, 30,000 cells were analyzed by flow<br />

cytometry. Result and discussion: The results <strong>of</strong> the first case <strong>of</strong><br />

the study were WBC=780 and LUC=2.2%, CD7=60%, CD=57%,<br />

CD5=56%<strong>of</strong>thelymphocytepopulation<strong>of</strong>peripheralblood.But<br />

the same markers in bone marrow sample were determined for<br />

D7=5%, CD3=5%, and CD5=6.5%. The results <strong>of</strong> the second case<br />

<strong>of</strong> the study were CD19=90%, CD20=89%, CD22=82%, and<br />

CD5=95% <strong>of</strong> the lymphocyte population <strong>of</strong> peripheral blood. In<br />

addition the same markers in bone marrow samples were<br />

determined for CD19=76%, CD20==85%, CD22=75.9%, and<br />

CD5=93%. HLA-DR marker was shown to be over 80% in both<br />

peripheral blood and bone marrow for this case. The result <strong>of</strong><br />

cell counting for the third case <strong>of</strong> the study showed WBC=4800<br />

and LUC=6.3%, CD19=41.5%, CD20=29.5%, CD22=39% and<br />

CD5=43.12%, CD3=40.3%, and CD7=41.1% in peripheral blood.<br />

But the same markers from bone marrow sample were<br />

determined for CD19=85.6%, CD22=81.2% and CD5=2.08%<br />

and CD3=1.88%. The results <strong>of</strong> this study show that the<br />

lymphocyte protein expression in leukemic samples is the value<br />

in diagnostic and classification <strong>of</strong> leukemia.<br />

doi:10.1016/j.clim.2007.03.325<br />

F.113 An All-Human Culture Model Supporting<br />

Human B Lymphopoesis<br />

Mercy Kagoda, Graduate Research Assistant/Student, Loma<br />

Linda University School <strong>of</strong> Medicine Department <strong>of</strong> Pathology<br />

and Human Anatomy, Loma Linda, CA, Yasmin Parrish,<br />

Research Specialist II, Childrens Hospital Los Angeles Research<br />

<strong>Immunology</strong> Bone Marrow Transplant, Los Angeles, CA,<br />

Jaqueline Rogerio, Post Doctoral Fellow, Childrens Hospital Los<br />

Angeles Research <strong>Immunology</strong> Bone Marrow Transplant, Los<br />

Angeles, CA, Ineavely Baez, Senior Research Assistant, Loma<br />

Linda University School <strong>of</strong> Medicine Department <strong>of</strong> Pathology<br />

and Human Anatomy, Loma Linda, CA, Qian-Lin Hao, Research<br />

Specialist IV, Childrens Hospital Los Angeles Research<br />

<strong>Immunology</strong> Bone Marrow Transplant, Los Angeles, CA, Lora<br />

Barsky, Flow Technologist, Childrens Hospital Los Angeles<br />

Research <strong>Immunology</strong> Bone Marrow Transplant, Los Angeles,<br />

CA, Ewa Zielinska, Flow Technologist, Childrens Hospital Los<br />

Angeles Research <strong>Immunology</strong> Bone Marrow Transplant, Los<br />

Angeles, CA, Monika Smogorzewska, Flow Technologist,<br />

Childrens Hospital Los Angeles Research <strong>Immunology</strong> Bone<br />

Marrow Transplant, Los Angeles, CA, Kimberly Payne, Assistant<br />

Pr<strong>of</strong>essor, Loma Linda University School <strong>of</strong> Medicine<br />

Department <strong>of</strong> Pathology and Human Anatomy, Loma Linda,<br />

CA<br />

Traditionally, nonfetal models <strong>of</strong> human B cell development<br />

have been based on co-culturing hematopoietic stem cells<br />

(HSCs) from cord blood (CB) and bone marrow (BM) on murine<br />

stromal cell lines. Here we describe the progressive replacement<br />

<strong>of</strong> murine components to generate a totally human<br />

culture model that supports human hematopoiesis, including B<br />

cell development. First, we replaced murine stromal cell lines<br />

with primary human BM stroma, next human serum was<br />

substituted for nonhuman sera. It has been reported that<br />

human B cell precursors selectively interact with CD10+ stromal<br />

cells during in vivo B cell development. In our cultures, human<br />

stroma express CD10 and secrete IL-7 at levels comparable to<br />

primary murine BM stroma. We found that human stroma bind<br />

IL-7 independently <strong>of</strong> the IL-7 receptor thereby having the<br />

potential for presenting IL-7 to developing B cell precursors.<br />

This is important as we have recently shown that IL-7 expands<br />

human B cell precursors by ∼50 fold and that IL-7 is essential for<br />

production <strong>of</strong> B cells from HSCs in adult human BM. While<br />

human stroma in our cultures express the human pre-BCR<br />

ligand, galectin-1 (Gal-1), the presence <strong>of</strong> Gal-1 was not<br />

required for IL-7-induced expansion <strong>of</strong> human B cell precursors.<br />

HSCs in co-cultures with human stroma and serum showed a<br />

generative capacity for CD19+ and CD19− progeny that was at<br />

least equal to that seen on murine stroma with nonhuman sera.<br />

Planned experiments will assess the ability <strong>of</strong> this culture model<br />

to support primary B cell malignancies.<br />

doi:10.1016/j.clim.2007.03.326<br />

F.114 Expression <strong>of</strong> CD3 and T Cell Receptor (TCR)<br />

on a Minor Population <strong>of</strong> Human CD14 Positive Cells<br />

Daron Forman, Senior Scientist, Tolerx, Cambridge, MA,<br />

Michael Rosenzweig, Senior Director <strong>of</strong> Preclinical<br />

Research, Tolerx, Cambridge, MA, Lou Vaickus, Chief<br />

Medical Officer, Tolerx, Cambridge, MA<br />

The TCR associates with CD3 polypeptides including CD3<br />

delta, gamma, epsilon, and zeta. It was commonly believed<br />

that TCR and CD3 expression occur exclusively on T cells.<br />

Recently however, TCR expression was reported on a minor<br />

population <strong>of</strong> both mouse and human neutrophils indicating


Abstracts<br />

that other populations may express CD3 and/or TCR. We<br />

evaluated whether CD3 and TCR-ab expression could be<br />

detected on non-T cell lineages in human peripheral<br />

blood. Cells were analyzed for CD3 and TCR-ab expression<br />

using monoclonal antibodies (Mabs) and flow cytometry. In<br />

addition to the expected distribution on lymphocytes, CD3<br />

and TCR-ab were detected on 0.3–1.0% population <strong>of</strong><br />

cells that expressed CD11b, CD11c, and CD14. This<br />

population expressed CD3 and TCR with an intensity<br />

similar to CD4+ and CD8+ T cells. CD14 is expressed at<br />

high levels on Fc-receptor (FcR) bearing monocytes. To<br />

exclude FcR binding as an explanation for the detection<br />

<strong>of</strong> anti-CD3 and anti-TCR-ab Mabs on CD14+ cells, studies<br />

were repeated with human serum or with pre-incubation<br />

with anti-FcR Mabs (anti-CD16 and anti-CD32). Staining<br />

was equivalent with and without FcR block, suggesting<br />

that anti-CD3 and anti-TCR-ab Mab binding was specific.<br />

These data suggest that a minor population <strong>of</strong> nonlymphoid<br />

cells that express CD11b, CD11c, and CD14 also<br />

express CD3 and TCR-ab. Further studies are ongoing to<br />

fully characterize this population.<br />

doi:10.1016/j.clim.2007.03.327<br />

F.115 Haplotype-specific Use <strong>of</strong> Lysosomal<br />

Proteases Can Modulate the Class II MHC Peptide<br />

Repertoire but Does Not Impair Invariant Chain<br />

Degradation in Human Antigen Presenting Cells<br />

Cristina Costanti-England PhD Candidate, Harvard Medical<br />

School/Brigham and Women’s Hospital, Center for<br />

Neurologic Diseases, Boston, MA, Howard Hang,<br />

Postdoctoral Fellow, Whitehead Institute, Massachusetts<br />

Institute <strong>of</strong> Technology, Cambridge, MA, David Hafler,<br />

Jack, Sadie and David Breakstone, Pr<strong>of</strong>essor <strong>of</strong> Neurology,<br />

Harvard Medical School/Brigham and Women’s Hospital,<br />

Center for Neurologic Diseases, Boston, MA, Hidde Ploegh,<br />

Pr<strong>of</strong>essor <strong>of</strong> Biology, Whitehead Institute, Massachusetts<br />

institute <strong>of</strong> Technology, Cambridge, MA<br />

The lysosomal protease asparagine endopeptidase (AEP)<br />

has been implicated in both the destructive processing <strong>of</strong><br />

self-antigen and the regulation <strong>of</strong> class II MHC maturation.<br />

In this study, we explore the role <strong>of</strong> human AEP in<br />

invariant chain processing in the maturation <strong>of</strong> several<br />

distinct allotypes <strong>of</strong> human class II MHC products. We find<br />

that, as is the case in mice, AEP activity is not necessary<br />

for class II maturation in human antigen-presenting cells.<br />

Our data demonstrate that Ii cleavage is regulated by<br />

many lysosomal proteases, including both aspartyl and<br />

cysteine proteases. Furthermore, we find that the<br />

repertoire <strong>of</strong> active proteases in EBV-transformed B cells<br />

differs between lines generated from different donors.<br />

These results suggest a unique pr<strong>of</strong>ile <strong>of</strong> lysosomal<br />

protease activity within each antigen-presenting cell<br />

that regulates Ii degradation. Our findings underscore<br />

the complexity <strong>of</strong> the class II MHC-restricted pathway <strong>of</strong><br />

antigen presentation. While allelic variation in MHC<br />

products is a decisive factor in determining the repertoire<br />

<strong>of</strong> presented peptides, the identity and level <strong>of</strong> activity<br />

<strong>of</strong> processing proteases present in antigen-presenting cells<br />

are contributing factors as well. As these proteases also<br />

generate the peptide cargo loaded into the binding<br />

pocket <strong>of</strong> class II MHC, it is certain that the repertoire<br />

<strong>of</strong> self and antigenic peptides presented to CD4+ T cells<br />

differs between individuals. Thus, altered presentation <strong>of</strong><br />

immunogenic self-peptides due to alternate protease<br />

pr<strong>of</strong>iles may be a factor contributing to the susceptibility<br />

to and severity <strong>of</strong> autoimmune diseases such as type 1<br />

diabetes and multiple sclerosis.<br />

doi:10.1016/j.clim.2007.03.330<br />

S53<br />

F.116 Induction <strong>of</strong> Antigen-specific <strong>Oral</strong> Tolerance<br />

by Genetically Modified Lactococcus Lactis<br />

Delivering DQ8-specific Immunodominant Gliadin<br />

Epitopes to Gluten-sensitized Class II Transgenic<br />

Mice<br />

Inge Huibregtse, Academic Medical Center, Amsterdam,<br />

Eric Marietta, Mayo Clinic, Department <strong>of</strong> <strong>Immunology</strong>,<br />

Rochester, MN, Shadi Rashtak, Drs, Mayo Clinic,<br />

Department <strong>of</strong> <strong>Immunology</strong>, Rochester, MN, Chella David,<br />

Mayo Clinic, Department <strong>of</strong> <strong>Immunology</strong>, Rochester, MN,<br />

Pieter Rottiers, VIB, Department for Molecular Biomedical<br />

Research, Zwijnaarde, Ghent, Sander van Deventer,<br />

Pr<strong>of</strong>essor Academic Medical Center, Center for<br />

Experimental and Molecular Medicine, Amsterdam,<br />

Joseph Murray, Mayo Clinic, Department <strong>of</strong> <strong>Immunology</strong>,<br />

Rochester, MN<br />

Antigen-specific immune suppression is an attractive<br />

therapy for the treatment <strong>of</strong> several gastro-intestinal<br />

diseases especially celiac disease. Active delivery <strong>of</strong><br />

recombinant autoantigens or allergens at the intestinal<br />

mucosa by genetically modified Lactococcus lactis (LL)<br />

provides a novel therapeutic approach for the induction<br />

<strong>of</strong> tolerance. For this purpose we genetically engineered<br />

deamidated DQ8 epitope secreting LL (LL-eDQ8d) and<br />

evaluated local and systemic immune responses in glutensensitized<br />

NOD ABo DQ8 class II transgenic mice after oral<br />

supplementation. Mice were sensitized with either 10 1/4g<br />

deamidated DQ8 epitopes or 50 ng pertussis toxin and 100<br />

1/4 g crude gluten and fed for 10 or 21 days, respectively<br />

with LL-pT1NX (empty vector) or LL-eDQ8d (1×10 9 CFU/<br />

day). Tolerance induction was assessed by DTH responses,<br />

ex vivo proliferation assays and cytokine measurements.<br />

Daily intragastric administration <strong>of</strong> LL-eDQ8d in sensitized<br />

transgenic mice led to a significant decrease in DTH response<br />

and proliferative capacity <strong>of</strong> the splenocytes and inguinal<br />

lymph node cells compared to the negative controls. LLpT1NX<br />

was also able to reduce the DTH response and the<br />

proliferation <strong>of</strong> splenocytes although not as much as the LLeDQ8d.<br />

Moreover, LL-eDQ8d significantly reduced the IFN- 3<br />

production in the draining lymph nodes and the spleen<br />

compared to the LL-pT1NX. Mucosal delivery <strong>of</strong> immunodominant<br />

gliadin epitopes by genetically modified LL induces<br />

suppression <strong>of</strong> local and systemic specific T cell responses in<br />

NOD ABo DQ8 transgenic mice. Moreover our data provide<br />

promise for the development <strong>of</strong> effective therapeutics for<br />

treatment <strong>of</strong> several common autoimmune, inflammatory


S54 Abstracts<br />

and/or allergic diseases by autoantigen- and/or allergensecreting<br />

L. lactis.<br />

doi:10.1016/j.clim.2007.03.331<br />

F.120 Evaluation <strong>of</strong> an Enhanced-sensitivity<br />

Interferon-γ ELISpot Assay Kit Using Cryopreserved<br />

PBMC Donor Bank Panels<br />

Keith Moskowitz, Director, SeraCare Life Sciences,<br />

Gaithersburg, MD, Janet Lathey, Director, Virology/<br />

<strong>Immunology</strong>, Gaithersburg, MD, Lisa Jeffrey, Research<br />

Associate, Virology/<strong>Immunology</strong>, Gaithersburg, MD, Scott<br />

Hickman, Product Manager, Marketing, Gaithersburg, MD,<br />

Rodshawn Branch, Research Associate, SeraCare Life<br />

Sciences, Gaithersburg, MD, Dmitry Moshk<strong>of</strong>f, Project<br />

Scientist, Virology/<strong>Immunology</strong>, Gaiithersburg, MD, Mark<br />

Manak, CSO, SeraCare, Gaithersburg, MD<br />

Human interferon-gamma (IFN-γ) ELISpot assays detect<br />

single IFN-γ secreting cells. We compared a novel ELISpot<br />

(SC) to a commercial kit (CK). Three lots <strong>of</strong> SC and one <strong>of</strong> CK<br />

were evaluated using PHA, CEF, CMV and mock stimulation <strong>of</strong><br />

cryopreserved PBMC from 24 independent donors. In<br />

response to PHA, SC and CK showed significant differences<br />

between frequency distributions (KS 0.36). SC median was<br />

479, 25th–75th percentiles (quartiles), and 275–975 SFC/<br />

well. The CK median was 200 (quartiles 45–343). Means were<br />

also different: SC=474, CK=152, Pb0.0001. Average CVs<br />

were 11% for SC and 18% for CK. For CEF stimulus, SC median<br />

was 64 (quartiles 27–179), mean was 129, CV 20%; CK median<br />

was 54 (quartiles 23–174), mean 139, and CV 28%. CMV<br />

stimulus median for SC was 100 (quartiles 12–200), mean<br />

149, and CV 27%; CK median was 87 (quartiles 7–222), mean<br />

112, and CV 38%. All backgrounds averaged b2. SC lots<br />

performed equivalently with all agonists. Some CK wells<br />

were unreadable (30/216), while 100% <strong>of</strong> 648 SC wells gave<br />

SFC values, Pb0.000001. CK averaged ∼14% failures, with no<br />

readings in 4 <strong>of</strong> 24 subjects. Thus, SeraCare has validated an<br />

IFN-γ ELISpot kit demonstrating superior performance in<br />

response to PHA, lower CV to all agonists and statistically<br />

fewer well failures relative to a commercial kit.<br />

doi:10.1016/j.clim.2007.03.332<br />

F.121 Involvement <strong>of</strong> Hsp90β but Not Hsp90α in<br />

Anti-Apoptotic Effect <strong>of</strong> CpG-B ODN<br />

Cheng-Chin Kuo, Postdoctoral Fellow, Agricultural<br />

Biotechnology Research Center, Academia Sinica, Taipei,<br />

Chi-Ming Liang, Pr<strong>of</strong>essor, Institute <strong>of</strong> Biological Chemistry,<br />

Academia Sinica, Taipei, Chen-Yen Lai, Research Assistant,<br />

Agricultural Biotechnology Research Center, Academia<br />

Sinica, Taipei, Shu-Mei Liang, Pr<strong>of</strong>essor, Agricultural<br />

Biotechnology Research Center, Academia Sinica, Taipei<br />

Unmethylated CpG oligodeoxynucleotides (CpG ODNs)<br />

activate immune responses in a toll-like receptor 9 (TLR9)dependent<br />

manner. In this study, we found that stimulation<br />

<strong>of</strong> mouse macrophages and dendritic cells with B-type CpG<br />

ODN (CpG-B ODN) increased cellular heat shock protein<br />

(Hsp) 90β but not Hsp90α and prevented apoptosis induced<br />

by serum starvation or staurosporine treatment. The CpG-B<br />

ODN-induced Hsp90β expression depended on TLR9, myeloid<br />

differentiation factor 88 and PI3K. Inhibition <strong>of</strong> Hsp90β<br />

by expressing small interfering RNA suppressed not only<br />

Hsp90β expression but also PI3K-dependent phosphorylation<br />

<strong>of</strong> Akt and CpG-B ODN-mediated anti-apoptosis. Additional<br />

studies demonstrated that as described by other group,<br />

Hsp90β but not Hsp90α was associated with Bcl-2. Inhibition<br />

<strong>of</strong> Hsp90β suppressed the CpG-B ODN-induced association <strong>of</strong><br />

Hsp90β with Bcl-2 and impaired the inhibitory effect <strong>of</strong><br />

CpG-B ODN on the release <strong>of</strong> cytochrome c as well as the<br />

activation <strong>of</strong> caspase-3. This study thus reveals the<br />

involvement <strong>of</strong> Hsp90β but not Hsp90α in CpG-B ODNmediated<br />

anti-apoptotic responses and shows that Hsp90β is<br />

distinct from Hsp90α in its regulation <strong>of</strong> the cellular<br />

function <strong>of</strong> immune cells.<br />

doi:10.1016/j.clim.2007.03.333<br />

F.122 Immunoassays for Detection <strong>of</strong> Soluble Fc<br />

Alpha Receptor (CD89) in Plasma <strong>of</strong> IgA<br />

Nephropathy Patients<br />

Mirjana Hahn-Zoric, Research and Development Engineer,<br />

Sahlgrenska University Hospital, <strong>Clinical</strong> <strong>Immunology</strong>,<br />

Gothenburg, Sweden, Neda Tahmasebifar, Student,<br />

Sahlgrenska University Hospital, <strong>Clinical</strong> <strong>Immunology</strong>,<br />

Gothenburg, Sweden, Cees van Kooten, Department <strong>of</strong><br />

Nephrology, Leiden, The Netherlands, Sweden, Jarl Ahlmen,<br />

Nephrology Clinics, Skövde, Per-Anton Westerberg,<br />

Nephrology Clinics, Skövde, Sweden, Svante Swerkersson,<br />

Pediatric Uro-Nephrologic Center, Gothenburg, Sweden,<br />

Sverker Hansson, Pediatric Uro-Nephrologic Center,<br />

Gothenburg, Sweden, Ulla Berg, Karolinska University<br />

Hospital, Stockholm, Sweden, Lars Åke Hanson, Pr<strong>of</strong>essor<br />

Emeritus, Sahlgrenska Academy, Gothenburg University,<br />

<strong>Clinical</strong> <strong>Immunology</strong>, Gothenburg, Sweden, Leonid<br />

Padyukov, Senior Researcher, Rheumatology Unit,<br />

Department <strong>of</strong> Medicine, Stockholm, Sweden, Stefan H.<br />

Jacobson, Department <strong>of</strong> Nephrology, Stockholm, Sweden<br />

Background: Fc±RI, or CD89, is a receptor binding the Fc<br />

portion <strong>of</strong> IgA. Recently it has been shown that CD89 is involved<br />

in IgA–immune complex formation. CD89–IgA complexes were<br />

suggested to be pathogenic in several diseases, one <strong>of</strong> which is<br />

IgA nephropathy (IgAN). IgAN is characterized by increased<br />

serum IgA levels paralleled by the deposition <strong>of</strong> IgA in renal<br />

tissue. IgAN is the most common form <strong>of</strong> glomerulonephritis in<br />

the Western world and is the second most common cause <strong>of</strong><br />

dialysis or kidney transplantation after diabetes. The pathogenesis<br />

and the mechanisms <strong>of</strong> this disease are not well understood<br />

and there are no reliable prognostic factors that can predict<br />

which patients are <strong>of</strong> high risk to develop severe kidney damage.<br />

Aim <strong>of</strong> this study was to develop immunoassay(s) for detection <strong>of</strong><br />

soluble IgA receptor (sCD89) in human plasma. Methods and<br />

material: By using CD89-specific MIP8a and A3 monoclonal<br />

antibodies and polyclonal anti-CD89 IgG, a Western blot method<br />

as well as a direct and an indirect ELISA for quantification <strong>of</strong><br />

sCD89 were developed. 45 IgAN patients (32 adults and 13


Abstracts<br />

teenagers) and 45 matched healthy controls were analyzed.<br />

Results: A single sCD89 band <strong>of</strong> around 30 kDa was identified in<br />

Western blot with polyclonal IgG. Significantly higher levels <strong>of</strong><br />

sCD89 were found in adult IgAN patients compared to controls in<br />

monoclonal antibody-based ELISA. Conclusion: We developed<br />

assays that provide helpful tools for detection <strong>of</strong> sCD89, which<br />

may be a useful diagnostic marker for further studies <strong>of</strong> IgAN.<br />

doi:10.1016/j.clim.2007.03.335<br />

F.124 Resistin Like Molecule Beta Regulates<br />

Intestinal Inflammation in Chronic Colitis<br />

Luqman Seidu, Allergy/<strong>Immunology</strong> Fellow, Cincinnati<br />

Children’s Hospital, Cincinnati, OH, Elizabeth Forbes, PhD<br />

Student, Cincinnati Children’s Hospital, Cincinnati, OH,<br />

Richard Aherns, Rheumatoid Arthritis, Cincinnati Children’s<br />

Hospital, Cincinnati, OH, Margaret Karow, PhD, Regeneron<br />

Pharma, Tarrytown, NY, Carine Blanchard, PhD, Cincinnati<br />

Children’s Hospital, Cincinnati, OH, Andrew Murphy, PhD,<br />

Regeneron Pharma, Tarrytown, NY, David Valenzuela, PhD,<br />

Regeneron Pharma, Tarrytown, NY, George Yancopoulos,<br />

MD/PhD, Regeneron Pharma, Tarrytown, NY, Marc<br />

Rothenberg, MD/PhD, Cincinnati Children’s Hospital,<br />

Cincinnati, OH, Simon Hogan, PhD, Cincinnati Children’s<br />

Hospital, Cincinnati, OH<br />

ResistinlikemoleculeRELM-β, a member <strong>of</strong> resistin family,<br />

has been implicated in glucose metabolism and intestinal<br />

immunity. We demonstrate that RELM-β is constitutively<br />

expressed in the colonic epithelium and is upregulated during<br />

chronic intestinal inflammation. To examine the contribution<br />

<strong>of</strong> RELM-β to chronic intestinal inflammation, we utilized<br />

RELM-β deficient mice and models <strong>of</strong> colitis. We show that<br />

RELM-β deficient mice have decreased susceptibility to<br />

chemical (DSS [dextran sodium sulfate])-induced colitis<br />

(disease activity index, 1.53±1.53 vs. 7.37±2.4 respectively<br />

mean±SE; survival at day 8, 100% vs. 75% respectively n=4–<br />

5 mice per group). We show that the link between colitis<br />

susceptibility and RELM β was associated with the expression<br />

<strong>of</strong> the NFkappaB signaling inhibitor, REG3β. Moreover,<br />

REG3β mRNA expression was increased 500-fold in DSStreated<br />

RELM-β deficient mice vs. wild type mice (616.8±<br />

138.1 vs. 26.35±11.69, fold increase [REG3β expression/<br />

GAPDH ratio], respectively n=4–5 mice per group, mean±<br />

SE). Collectively this work suggests an interaction among<br />

RELM-β, REG3β and intestinal inflammation.<br />

doi:10.1016/j.clim.2007.03.336<br />

F.125 Immune Parameters in Normal Humans with<br />

Different Behavior Patterns: Type A and B Behavior<br />

Pattern<br />

Fereshteh Alesahebfosoul, Academic Member <strong>of</strong> Isfahan<br />

University, Medical School, <strong>Immunology</strong> DEP, Isfahan,<br />

Bahareh Zzarkeshfard, Food Science Engineering, Health<br />

Faculty, Isfahan, Farzad Oreizi, Immunologist, Medical<br />

School, <strong>Immunology</strong> DEP, Isfahan, Minoo Adib, Pr<strong>of</strong>essor,<br />

medical school, <strong>Immunology</strong> DEP, Isfahan<br />

Introduction: Numerous studies have described various<br />

aspects <strong>of</strong> behavioral influences on immune function. Behavior<br />

and emotions modulate immune function. In this study<br />

distinctive immunological findings in normal humans with type<br />

A and B behavior pattern were described. Method: In cross<br />

sectional study 105 participants (P) admitted to be in the first<br />

phase (20–30 years old). 6 Ps were excluded because <strong>of</strong><br />

abnormal CBC. We used standard questioner (Bortne scale) to<br />

separate type A and B behavior patterns. The samples <strong>of</strong><br />

peripheral blood were referred to immunology laboratory for<br />

measurement <strong>of</strong> immune factors. SPSS (ver.10) and t test were<br />

used in statistical analysis. Results: 40 Ps were type A and 59<br />

type B. There were no significant differences between two<br />

groups on serum B cell, T cell, and NK cell. There was a<br />

significant increase (P valueb0.05) on serum CD4 positive Tcells<br />

in type A but no significant increase was viewed in CD8 positive T<br />

cells. There were significant increases on lymphocytes and<br />

granulocytes in type A. It was significant in male Ps with type A.<br />

CD4 positive T cells increased in counter female with type B,<br />

however no significant difference was seen in male with type A<br />

in contrast to female. Discussion: The finding <strong>of</strong> this study was<br />

that type A behavior pattern with specific characteristics (time<br />

emergency) has no significant effect on immune system and the<br />

effect <strong>of</strong> gender <strong>of</strong> immune parameters has to be considered.<br />

doi:10.1016/j.clim.2007.03.337<br />

S55<br />

F.126 Multi-Parameter Analysis (MPA) <strong>of</strong> Cells and<br />

Serum Using a Standard Flow Cytometer<br />

Robert Hallam, Senior Biomedical Scientist, Papworth<br />

Hospital, <strong>Immunology</strong> Department, Cambridgeshire,<br />

England, Paul Balmer, <strong>Clinical</strong> Scientist, HPA, Vaccine<br />

Evaluation Department, Manchester, England, Ray Borrow,<br />

<strong>Clinical</strong> Scientist, HPA, Vaccine Evaluation Department,<br />

Manchester, England, Andrew Exley, Consultant<br />

Immunologist, Papworth Hospital/<strong>Immunology</strong> Department,<br />

Cambridge, England<br />

Background: Flow cytometric peripheral blood MPA identifies<br />

cell populations by size, granularity, and expression pattern<br />

<strong>of</strong> cell surface or intracellular antigens using fluorophore<br />

conjugated Abs. Alternative platforms for serum MPA measure<br />

concentrations <strong>of</strong> secreted proteins including cytokines and<br />

pathogen specific Abs in multiplex, liquid phase capture assays<br />

using antigen or antibody coated beads identified by size,<br />

fluorescence intensity and wavelength. Fresh blood samples<br />

preferred for cellular analysis generate peaks and troughs in<br />

workload contrasting with batch analysis <strong>of</strong> sera. Methods: B<br />

cell populations were defined by CD45/ss pan-leucogating, then<br />

differential expression <strong>of</strong> CD19, CD27, IgD or IgM using a<br />

Beckman-Coulter FC-500 flow cytometer. 10-plex serotype<br />

specific anti-pneumococcal antibody assays were established<br />

using carboxylated microspheres coated with pneumococcal<br />

capsular polysaccharide, CPS and 22F absorption, PE-conjugated<br />

mouse anti-human IgG/IgM, and reference standard 89SF.<br />

Results: These serum MPAs have 2–3 log dynamic linear ranges,<br />

b6% intraassay and 16% interassay CVs, 1.88 ng/ml median<br />

limits <strong>of</strong> quantification, and correlate well with Bioplex systems<br />

and ELISAs. The complex data set is presented as an assay


S56 Abstracts<br />

report highlighted by a “traffic light system” and pictorial<br />

patient reports in MS-WORD, after analysis by especially<br />

designed s<strong>of</strong>tware incorporating MS-EXCEL macros enhanced<br />

by Visual Basic programming. B cell phenotyping correlated<br />

with pre versus post-immunisation serum pneumococcal antibody<br />

levels in cases investigated for primary and secondary<br />

immunodeficiency. Discussion: MPA <strong>of</strong> cells and serum on a<br />

single standard flow cytometer with powerful s<strong>of</strong>tware applies<br />

existing expertise and resources in a flexible cost-effective<br />

approach suitable for high-throughput testing and clinical<br />

laboratories.<br />

doi:10.1016/j.clim.2007.03.338<br />

F.127 Catalytic Anti-dsDNA Activity from SLE Sera is<br />

Associated with Remission Phases <strong>of</strong> the Disease<br />

Genevieve Servais, Responsible Autoimmunity LAB, Canada,<br />

Canada, CHU Brugmann, <strong>Immunology</strong>, Bruxelles, Belgium,<br />

Mimouna El Baz, Pharmacist, CHU Brugmann <strong>Immunology</strong>,<br />

Bruxelles, Belgium, Rafik Karmali, MD, CHU Brugmann<br />

Internal Medicine, Bruxelles, Belgium, Marie Paule<br />

Guillaume, MD, CHU Brugmann Internal Medicine,<br />

Bruxelles, Belgium, Jean Duchateau, MD PHD, CHU<br />

Brugmann <strong>Immunology</strong>, Bruxelles, Belgium<br />

SLE is characterized by the presence <strong>of</strong> circulating anti-<br />

DNA antibodies, one <strong>of</strong> the eleven ACR criteria for the<br />

classification <strong>of</strong> SLE, and they play an important role in the<br />

pathogenesis <strong>of</strong> SLE. Anti-DNA antibodies display different<br />

specificities, some being restricted to nuclear double<br />

stranded DNA (dsDNA), or single stranded DNA (ssDNA), or<br />

membrane associated DNA (mDNA) or nucleosomes. Some<br />

are sharing dual specificities. Autoantibodies displaying a<br />

catalytic activity, abzymes, were reported in patient’s sera,<br />

including catalytic anti-DNA antibodies. They have not been<br />

reported in healthy control subjects. Catalytic activity varies<br />

in different pathologies and changes during the different<br />

phases <strong>of</strong> the disease. The biological role <strong>of</strong> the catalytic<br />

anti-DNA present in serum <strong>of</strong> SLE is complex, as it seems that<br />

some may play positive and others negative roles, from a<br />

clearance role to a lowering <strong>of</strong> DNAse activity in SLE<br />

serum. We show here that circulating catalytic anti-DNA is<br />

found in SLE and RA patients but also in healthy controls<br />

though with a higher percentage in SLE than in controls,<br />

where they can be unraveled when the sera are diluted<br />

at 1/10. Their presence is inversely correlated with the<br />

clinical activity <strong>of</strong> the disease and with the titer <strong>of</strong><br />

circulating autoantibodies to dsDNA, mDNA but not antinucleosomes<br />

antibodies.<br />

doi:10.1016/j.clim.2007.03.339<br />

F.128 Fungal Hypersensitivity is Not Type I (IgE)<br />

Allergy<br />

Vincent A. Marinkovich, MD, Inc., Redwood City, CA<br />

One hundred adult patients undergoing clinical evaluations<br />

for symptoms developing during chronic exposure to<br />

high ambient fungal antigen levels in water damaged<br />

homes were tested for specific IgE (RAST) and specific IgG<br />

(ELISA) levels to a number <strong>of</strong> fungi commonly found in<br />

mold-amplified environments. The tests were standard<br />

runs in a State licensed Physician’s Office Laboratory<br />

using test materials provided by Hitachi Chemical Diagnostics<br />

for specific IgE and specific IgG in a multi antigen<br />

panel format. The panels contain aspergillus, penicillium,<br />

alternaria, cladosporium and mucor. Results read as<br />

positive when greater than 2 standard deviations greater<br />

than the mean using reference ranges previously determined<br />

by Hitachi. Seven patients had elevated specific<br />

IgE levels to at least one <strong>of</strong> the fungi tested (7%). Eighty<br />

nine patients showed elevated IgG titers to at least one<br />

<strong>of</strong> the molds. The sera were then exposed to an<br />

additional eight common molds, botrytis, candida, epicoccum,<br />

fusarium, helminthosporium, pullularia, rhizopus<br />

and stemphylium, on an IgG panel and all patients (100%)<br />

showed elevated titers to at least one <strong>of</strong> the molds. The<br />

common symptoms reported were remarkably consistent<br />

among patients: fatigue, nasal congestion, headaches,<br />

cough, memory loss and chronic flu-like symptoms. All the<br />

patients showed signs <strong>of</strong> chronic rhinosinusitis suggesting<br />

fungal colonization and eosinophilic degranulation. Systemic<br />

symptoms are likely the result <strong>of</strong> circulating<br />

immune complexes under conditions <strong>of</strong> functional asplenia<br />

(i.e., IC overload). The neurological symptoms may be the<br />

result <strong>of</strong> local uptake <strong>of</strong> eosinophilic neurotoxin via the<br />

cribriform plate.<br />

doi:10.1016/j.clim.2007.03.340<br />

F.129 Automation for Isolation <strong>of</strong> Peripheral Blood<br />

Mononuclear Cells (PBMC)<br />

Carlos Aparicio, Scientist, Beckman Coulter, Inc., Miami, FL,<br />

Enrique Rabelli, Director, Beckman Coulter, Inc., Miami, FL,<br />

Edward Jachimowicz, Scientist, Beckman Coulter, Inc.,<br />

Miami, FL, Sybil D’Costa, Scientist, Beckman Coulter, Inc.,<br />

Miami, FL, Mahsa Aspsater, Scientist, Beckman Coulter,<br />

Inc., Miami, FL, Julie Wilkinson, Scientist, Beckman<br />

Coulter, Inc., Miami, FL, Wade Bolton, VP, Beckman<br />

Coulter, Inc., Miami, FL, Enrique Rabelli, Director,<br />

Beckman Coulter, Inc., Miami, FL<br />

Peripheral blood mononuclear cells (PBMC) are considered<br />

appropriate target populations to immunologically<br />

assess the various subsets <strong>of</strong> blood lymphocytes. Purification<br />

<strong>of</strong> PBMC by density gradient centrifugation is a<br />

laborious and time consuming manual process and, thus,<br />

limiting factor for processing large number specimens.<br />

Generally, the media–gradient interface is generated by<br />

carefully pipetting the blood cell suspension onto a<br />

density gradient solution without disturbing the interface.<br />

Disruption <strong>of</strong> the interface can jeopardize the overall<br />

yield, purity and viability <strong>of</strong> the mononuclear cell<br />

preparation. The study described herein focuses on an<br />

alternative semi-automated strategy to harvest PBMC<br />

using the Ficoll-Paque density gradient. This process<br />

integrates the use <strong>of</strong> an automated liquid handler and a<br />

centrifuge to minimize the number <strong>of</strong> steps and time


Abstracts<br />

requiring operator intervention in current laboratory<br />

procedures. PBMC harvested by this automated process<br />

had a viability over 95%, and when compared to the<br />

manual procedure showed: (1) cell recovery (N95%) with<br />

no selective loss as judged by phenotyping and functional<br />

response by the various lymphocytes. The goal <strong>of</strong> this<br />

work is to provide a fully automated and standardized<br />

method for preparing PBMC that can facilitate support<br />

cell based-assay testing strategies in clinical trials by<br />

improving overall quality and efficiency <strong>of</strong> the process as<br />

well as reducing labor and cost.<br />

doi:10.1016/j.clim.2007.03.341<br />

F.130 Interleukin 32 Expression is Associated with a<br />

Unique Subset <strong>of</strong> Human Dendritic Cells Receptive<br />

to the Venezuelan Equine Encephalitis<br />

Virus-derived Replicon Vector<br />

Kevin Nishimoto, Graduate student, University <strong>of</strong> California<br />

Irvine, Department <strong>of</strong> Microbiology and Biochemistry,<br />

Irvine, CA, Charles Dinarello, Pr<strong>of</strong>essor <strong>of</strong> Medicine,<br />

University <strong>of</strong> Colorado Health Sciences Center, Department<br />

<strong>of</strong> Medicine, Denver, CO, Stephen Hou, PhD, University <strong>of</strong><br />

California Irvine, Department <strong>of</strong> Medicine, Division <strong>of</strong><br />

Hematology/Oncology, School <strong>of</strong>, Irvine, CA, Edward<br />

Nelson, Assistant Pr<strong>of</strong>essor <strong>of</strong> Medicine, University <strong>of</strong><br />

California Irvine Department <strong>of</strong> Medicine, Division <strong>of</strong><br />

Hematology/Oncology, School <strong>of</strong> Medicine, Irvine, CA<br />

Establishing successful immunotherapeutic strategies<br />

utilizing dendritic cells (DC), the most potent antigen<br />

presenting cell, largely depends on understanding the<br />

requirements necessary for optimal DC function. We and<br />

others have reported that the Venezuelan Equine Encephalitis<br />

(VEE) replicon particle (VRP) vector system has<br />

conserved tropism for murine, rat, and human DCs.<br />

However the DC subset(s) and functional capacities are<br />

not fully understood. The VRP vector system has in vitro<br />

tropism for a subset <strong>of</strong> human immature monocytederived<br />

DCs (M-DCs); therefore, we developed a novel<br />

technique to isolate this VRP receptive DC subset for<br />

further characterization. An evaluation between VRP<br />

receptive and non-receptive M-DC subsets by microarray<br />

revealed distinct differences in gene expression pr<strong>of</strong>iles<br />

with N700 significantly expressed genes (0.001 e P value,<br />

0.98 d PPDE) between cellular subsets. Notably in the<br />

receptive DC population, the NK4 transcript was significantly<br />

increased and accompanied by protein expression.<br />

The NK4 gene has recently been identified as encoding a<br />

recently described cytokine, interleukin 32. These data<br />

provide the initial characterization <strong>of</strong> a new subset <strong>of</strong><br />

myeloid DCs targeted by this vector, and provide insights<br />

into human DC biology allowing explicit study <strong>of</strong> the<br />

mechanisms employed by this exceptionally potent immunotherapeutic<br />

vector system.<br />

doi:10.1016/j.clim.2007.03.342<br />

F.131 Retinoic Acid Promotes Development <strong>of</strong><br />

Human Macrophages Over Dendritic Cell<br />

Differentiation<br />

Juliana Sousa-Canavez, Researcher, Genoa Biotech, São<br />

Paulo, Brazil, Cristina Massoco, Researcher, Genoa<br />

Biotech, São Paulo, Brazil, Elaine Corneta, Student,<br />

Genoa Biotech, São Paulo, Brazil, Katia Leite, Pathologist,<br />

Genoa Biotech, São Paulo, Brazil, Tatiane Marisis, Student,<br />

Genoa Biotech, São Paulo, Brazil, Luiz Camara-Lopes,<br />

Pathologist, Genoa Biotech, São Paulo, Brazil, Antonio<br />

Misiara, MD, Genoa Biotech, São Paulo, Brazil, Dewton<br />

Moraes-Vasconcelos, Pr<strong>of</strong>essor, Genoa Biotech, São Paulo,<br />

Brazil<br />

Previous studies showed that all vitamins including<br />

vitamin A are fundamental for immune system properly<br />

functioning. All trans retinoic acid (atRA) at physiological<br />

or near-physiological concentrations appears to affect<br />

Th1–Th2 differentiation and it is believed that effects<br />

<strong>of</strong> retinoids on immune responses might also be mediated<br />

by dendritic cells (DC). Nonetheless, effects <strong>of</strong> atRA in DC<br />

differentiation have been showing contradictory results<br />

since it was observed either induction or inhibition <strong>of</strong><br />

differentiation. Our work shows effects <strong>of</strong> atRA on human<br />

monocyte derived DC by phenotype and phagocytosis<br />

studies. DC (positive control group) was differentiated<br />

with cytokines (GM-CSF and IL-4) and maturation induced<br />

by TNF-α. We have tested pharmacological (10 1/4M and<br />

1 1/4M) and physiological (10 nM) doses <strong>of</strong> atRA with or<br />

without cytokines. Cell phenotype was analyzed by flow<br />

cytometry using CD80, CD86 and CD14 antibodies. We also<br />

investigated functional performance analyzing phagocytosis<br />

and respiratory burst (ROS) in all culture conditions<br />

mentioned above. Our data demonstrate that effects <strong>of</strong><br />

atRA depend on the dose used, as pharmacological doses<br />

inhibited expression <strong>of</strong> all phenotypic markers tested,<br />

while a physiological one caused cell differentiation.<br />

Physiologic concentration <strong>of</strong> atRA especially in combination<br />

with GM-CSF blocked differentiation <strong>of</strong> DC population<br />

corroborating a macrophage preferable transformation.<br />

Moreover phagocytosis and ROS were approximately three<br />

times greater in cells cultivated with atRA compared to<br />

positive control group. On the other hand, IL-4 and atRA<br />

apparently induced differentiation to an immature DC<br />

phenotype when TNF-α stimulus was added.<br />

doi:10.1016/j.clim.2007.03.343<br />

S57<br />

F.132 OTC Allergy Testing<br />

Chris Brown, ImmuneTech, Inc., Menlo Park, CA, Vincent A.<br />

Marinkovich, MD, Inc. Redwood City, CA<br />

Quantitative, comprehensive in vitro, specific IgE tests<br />

for allergy have not hitherto been introduced into the<br />

over the counter (OTC) market because <strong>of</strong> their high costs<br />

and the need for a large sample <strong>of</strong> blood requiring a<br />

venapuncture. Recently the U.S. Food and Drug Administration<br />

cleared for OTC distribution a new allergenspecific<br />

IgE test which has two major advantages over


S58 Abstracts<br />

its predecessors: low blood volume and economy. This<br />

new system, utilizing a flow cytometry platform, is<br />

currently configured to provide quantitative specific IgE<br />

levels to each <strong>of</strong> ten allergens simultaneously. It utilizes<br />

just 5 μl <strong>of</strong> capillary or venous serum in a homogenous<br />

assay, and can be completed in under 3 h. The system<br />

compares favorably to the current laboratory testing<br />

methods:<br />

Sensitivity/specificity/accuracy data in %:<br />

– Mountain cedar 88/93/90<br />

– Timothy grass 94/97/90<br />

– Bermuda grass 86/97/91<br />

– Short ragweed 90/95/92<br />

– House dust mite 96/93/95<br />

– Cat hair 96/89/93<br />

– Cow’s milk 81/92/89<br />

– Egg 80/94/92<br />

– Wheat 85/93/90<br />

– Alternaria 87/94/90<br />

The coefficient <strong>of</strong> variation between test runs on<br />

different days ranged from 3.4 to 7.2% and within the<br />

same day from 2.9 to 8.3%. The test’s lowest level <strong>of</strong><br />

detection is 0.35 IU/ml. This accurate, low cost, low blood<br />

volume test with fingerstick convenience can expand the<br />

availability <strong>of</strong> accurate allergy diagnosis to a patient<br />

population not currently served by existing in vitro systems<br />

or by skin testing.<br />

doi:10.1016/j.clim.2007.03.344<br />

F.133 Standardization and Quality Assurance <strong>of</strong><br />

Cytokine Flow Cytometry Assays<br />

Maria Jaimes, Scientist, BD Biosciences, San Jose, CA,<br />

Janice Darden, CRS Team Leader, DAIDS/NIAID/NIH,<br />

Bethesda, MD, Patricia D’Souza, Program Officer, DAIDS/<br />

NIAID/NIH, Bethesda, MD, Holden Maecker, Group<br />

Manager-Immune Function, BD Biosciences, San Jose, CA<br />

Cytokine flow cytometry (CFC) is used to measure<br />

antigen-specific T cell responses in settings such as<br />

experimental HIV vaccination. Standardization <strong>of</strong> CFC<br />

among different laboratories would provide a common<br />

endpoint assay for comparing the immunogenicity <strong>of</strong><br />

vaccine candidates in clinical trials. For this purpose, a<br />

Quality Assurance Program has been developed in a<br />

collaborative effort between BD Biosciences, the Division<br />

<strong>of</strong> AIDS, NIAID, NIH and SeraCare BioServices, in which 15<br />

sites currently participate. Three rounds <strong>of</strong> the program<br />

have been completed. In each round, IFN-ƒ× and IL-2<br />

responses in CD4+ and CD8+ T cells to CEF or CMV pp65<br />

lyophilized peptide mixes were tested using cryopreserved<br />

PBMC from CMV+ donors. Each site analyzed its data<br />

independently and provided, through the program’s<br />

website (http://bdicsqa.webbasix.com), their raw data<br />

for centralized analysis. In addition, the data generated<br />

by each laboratory were compared to a standard. The<br />

first two rounds <strong>of</strong> the program yielded inter-laboratory<br />

variability from 12 to 60%, for cytokine+ responses higher<br />

than 0.2%. Instrument setup and analysis (gating strategy)<br />

played a critical role in this variation. This variability was<br />

decreased in the third round by providing feedback to the<br />

laboratories, guidelines for data analysis, and a protocol<br />

and reagents for instrument setup. Of note, the interlaboratory<br />

variability (18–32%) was comparable to the<br />

intra-laboratory variability (4–25%) in the third round.<br />

Critical to success was the provision <strong>of</strong> standard operating<br />

procedures, centrally supplied reagents, and laboratory<br />

adherence to GCLP. The implementation <strong>of</strong> this Quality<br />

Assurance Program will ensure greater uniformity <strong>of</strong> data<br />

from multiple laboratories.<br />

doi:10.1016/j.clim.2007.03.345<br />

F.134 A Novel Cell Contact-dependent Requirement<br />

for Human CD40 Ligand Expression<br />

Jack Ragheb, Senior <strong>Clinical</strong> Investigator, Laboratory <strong>of</strong><br />

<strong>Immunology</strong>, NEI, NIH, Bethesda, MD, James Snyder,<br />

Research Fellow, NIH, Bethesda, MD, Jijia Shen, Research<br />

Fellow, NIH, Bethesda, MD, Paul Hu, Research Fellow, nIH,<br />

Bethesda, MD, Hooman Azmi, Research Fellow, NIH,<br />

Bethesda, MD, Qian Chen, Research Fellow, NIH, Bethesda,<br />

MD<br />

CD40L (CD154), an inducible costimulatory molecule<br />

expressed on CD4+ T cells, plays an essential role in the<br />

activation <strong>of</strong> B cells. Through the CD40L mediated<br />

induction <strong>of</strong> IL-12 and upregulation <strong>of</strong> MHC class II,<br />

CD80/86, and other accessory molecules on antigen<br />

presenting cells (APC), CD40L also catalyzes a positive<br />

feedback loop for T cell activation. Despite the pivotal<br />

juxtaposition <strong>of</strong> CD40L between APC and T cell activation,<br />

it is thought that only a TCR stimulus is required to<br />

initiate early CD40L expression. We demonstrate that<br />

unlike its mouse counterpart, human CD40L is not<br />

constitutively expressed on circulating T cells, nor do<br />

human CD4+ T cells contain an intracellular pool <strong>of</strong> CD40L<br />

protein. While a TCR stimulus is necessary for CD40L<br />

expression on primary human T cells, we show for the<br />

first time that expression <strong>of</strong> CD40L is highly dependent<br />

upon a cell–cell interaction with CD14hi monocytes that<br />

does not require LFA-3, ICAM-1, or CD80/86. Like CD28dependent<br />

IL-2 expression, this monocyte signal upregulates<br />

the transcription <strong>of</strong> CD40L. Thus the inducible<br />

expression <strong>of</strong> CD40L on CD4+ T cells, which provides a<br />

critical costimulatory signal for monocyte and B cell<br />

activation, is itself dependent upon a costimulatory signal<br />

delivered by monocytes but not B cells. The latter<br />

finding implies that a human B cell cannot activate its<br />

cognate helper T cell to deliver CD40L-mediated help.<br />

Therapeutics targeted at this monocyte costimulatory<br />

factor for CD40L may lead to new means <strong>of</strong> modulating<br />

the immune response and inducing long-term graft<br />

acceptance.<br />

doi:10.1016/j.clim.2007.03.346


Abstracts<br />

F.135 Endocytosis <strong>of</strong> Hsp-chaperoned Proteins and<br />

Peptides is Essential for MHC II Dependent Peptide<br />

Presentation<br />

Irena Straub, Medical Candidate, University Children’s<br />

Hospital Tübingen, Tuebingen, Markus Haug, University<br />

Children’s Hospital Tübingen, Tuebingen, Dorothee Wernet,<br />

Pr<strong>of</strong>essor, Department <strong>of</strong> Transfusion Medicine University<br />

Hospital Tübingen, Tuebingen, Ursula Holzer, Dr. Medicine,<br />

University Children’s Hospital Tübingen, Tuebingen,<br />

Guenther E. Dannecker, Pr<strong>of</strong>essor, Olgahospital Stuttgart,<br />

Stuttgart<br />

Heat shock protein Hsp70 complexed to antigenic peptides<br />

enhances the activation and proliferation <strong>of</strong> CD8+ and CD4+ T<br />

cells in vitro. However the mechanism for HSP70-dependent<br />

antigen presentation by MHC II molecules is not yet clear. It is<br />

thought that after internalization <strong>of</strong> the HSP–peptide<br />

complexes by APCs the complex is trafficking into the<br />

cytoplasm or endosomal vesicles to be represented by MHC I<br />

and/or MHC II, although the necessity <strong>of</strong> an uptake for<br />

presentation by MHC II is not proven. Here we investigated<br />

the need <strong>of</strong> endocytosis <strong>of</strong> HSP–peptide complexes for<br />

sufficient antigen presentation in a human antigen-specific<br />

system. PBMCs <strong>of</strong> healthy donors immunized against tetanus<br />

or influenza with known HLA-DR haplotype (DRB*1101, 0401)<br />

were used. Stimulation <strong>of</strong> Tcells was induced by tetanus toxin<br />

C-fragment (TT944-966), influenza hemagglutinin (HA307–<br />

319) or the whole tetanus toxid protein either with peptide/<br />

protein alone or with the peptide/protein complexed to<br />

Hsp70. Antigen specificity was detected by HLA-DR-tetramers.<br />

For blocking the endocytosis mechanism <strong>of</strong> CD4− APCs,<br />

the cells were pre-treated with Cytochalasin, Genistein or<br />

Chloroquine, resulting in a marked decline in generating<br />

specific T cells. These results argue for an internalization <strong>of</strong><br />

the complexes by APCs and uptake into the endosomal<br />

vesicles for presentation by MHC II. Furthermore HSP–protein<br />

complexes are able to enhance the proliferation <strong>of</strong> antigenspecific<br />

T cells affirming an intracellular pathway <strong>of</strong> HSPmediated<br />

peptide presentation. Taken together, we conclude<br />

that for MHC class II dependent activation <strong>of</strong> CD4+ antigenspecific<br />

T cells uptake <strong>of</strong> Hsp-chaperoned proteins and<br />

peptides into APCs and intracellular processing is necessary.<br />

doi:10.1016/j.clim.2007.03.347<br />

F.136 Development and Validation <strong>of</strong> a Novel Ig<br />

Isotyping Assay on Magnetic Microspheres<br />

Candice Reyes, Scientist, Bio-Rad Laboratories Protein<br />

Function Division, Research and Development, Hercules,<br />

CA, Joe Fedynyshyn, Staff Scientist, Bio-Rad Laboratories<br />

Protein Function Division, Research and Development,<br />

Hercules, CA, Sophie Allauzen, Research and<br />

Development Manager, Bio-Rad Laboratories Protein<br />

Function Division, Research and Development,<br />

Hercules, CA<br />

The Bio-Plex Suspension Array system provides a<br />

flexible and efficient platform for multiplex analysis <strong>of</strong><br />

biological samples. Monitoring the levels <strong>of</strong> immunoglobulins<br />

is a valuable tool for evaluating the immune<br />

response in many different fields <strong>of</strong> research. Bio-Rad<br />

has developed a new multiplex immunoglobulin isotyping<br />

assay on magnetic beads that simultaneously quantifies<br />

the levels <strong>of</strong> IgG1, IgG2, IgG3, IgG4, IgM, IgA, and IgE in<br />

human serum and plasma. The levels <strong>of</strong> immunoglobulins<br />

can vary for each class and subclass depending on factors<br />

like genetics, disease state, and drug treatment. Using<br />

the Bio-Plex Pro Human Ig Isotyping assay kit, a complete<br />

pr<strong>of</strong>ile <strong>of</strong> isotype levels can be created from as little as<br />

5 μL <strong>of</strong> sample in about 3 h. Sensitivity and upper and<br />

lower limits <strong>of</strong> quantitation (LLOQ and ULLOQ) will be<br />

discussed. Additional validation data obtained from control<br />

human serum samples and World Health Organization<br />

(WHO) Reference Standards will also be presented.<br />

doi:10.1016/j.clim.2007.03.348<br />

F.137 Utility <strong>of</strong> Lyophilized PMA and Ionomycin to<br />

Stimulate Lymphocytes in Whole Blood for<br />

Immunological Assays<br />

Shelley Belouski, Senior Scientist, Amgen <strong>Clinical</strong><br />

<strong>Immunology</strong>, Thousand Oaks, CA, John Ferbas, Director<br />

Medical Sciences, Amgen <strong>Clinical</strong> <strong>Immunology</strong>, Thousand<br />

Oaks, CA, Keith Kelley, Senior Associate Scientist, Amgen<br />

<strong>Clinical</strong> <strong>Immunology</strong>, Thousand Oaks, CA, Julie Wilkinson,<br />

Staff Adv Research Scientist, Beckman Coulter, Inc., Custom<br />

BioPharma Solutions, Miami, FL, Sid Suggs, Director, Amgen<br />

Medical Sciences, Thousand Oaks, CA, John Thomas, Senior<br />

Associate Scientist, Amgen <strong>Clinical</strong> <strong>Immunology</strong>, Thousand<br />

Oaks, CA, Shen-Wu Wang, Senior Associate Scientist, Amgen<br />

Molecular Sciences, Thousand Oaks, CA<br />

Background: The need to implement robust biomarkers in<br />

clinical trials has never been greater, and such efforts can be<br />

easily compromised by reagent instability or simple human<br />

error during assay set-up. Many biotechnology and pharmaceutical<br />

companies are introducing efforts to conduct<br />

biomarker studies under more rigorous settings, and use <strong>of</strong><br />

plates or tubes pre-loaded with stimulation or staining<br />

reagents could be <strong>of</strong> value for studies that involve flow<br />

cytometry. Results: Our laboratory took a biomarker analytical<br />

method that relied on liquid formulations <strong>of</strong> phorbol 12myristate<br />

13-acetate (PMA) and ionomycin and demonstrates<br />

here that tubes pre-loaded with lyophilized versions <strong>of</strong> the<br />

liquid reagents can provide equivalent results. The value <strong>of</strong><br />

this approach is that it safeguards against omission or<br />

erroneous addition <strong>of</strong> bulk liquid formulations <strong>of</strong> PMA and<br />

ionomycin to the reaction vessel (i.e., plate or tube) and also<br />

lends itself to extended stability/shelf-life <strong>of</strong> these<br />

reagents. Conclusions: On the basis <strong>of</strong> this initial success,<br />

we plan to expand our evaluation <strong>of</strong> lyophilized reagents so<br />

that they can be incorporated into our clinical biomarker<br />

campaigns as appropriate.<br />

doi:10.1016/j.clim.2007.03.349<br />

S59


S60 Abstracts<br />

#2201- Inflammation Good or Bad for Tregs<br />

Saturday, June 9<br />

10:45 am−11:05 am<br />

Viral Infection Enables CD4+CD25+ Regulatory T<br />

Cells to Protect From Autoimmune Diabetes<br />

Christophe Filippi, Postdoctoral Fellow, La Jolla Institute<br />

for Allergy and <strong>Immunology</strong>, La Jolla, CA, Janine Oldham,<br />

Research Assistant, La Jolla Institute for Allergy and<br />

<strong>Immunology</strong>, La Jolla, CA, Tom Wolfe, Research Assistant,<br />

La Jolla Institute for Allergy and <strong>Immunology</strong>, La Jolla,<br />

CA, Evelyn Rodrigo, Research Assistant, La Jolla Institute<br />

for Allergy and <strong>Immunology</strong>, La Jolla, CA, Urs Christen,<br />

Research Scientist, La Jolla Institute for Allergy and<br />

<strong>Immunology</strong>, La Jolla, CA, Lisa Togher, Research Assistant,<br />

La Jolla Institute for Allergy and <strong>Immunology</strong>, La Jolla,<br />

CA, Marianne Martinic, Postdoctoral Fellow, La Jolla<br />

Institute for Allergy and <strong>Immunology</strong>, La Jolla, CA,<br />

Matthias von Herrath, Pr<strong>of</strong>essor, La Jolla Institute for<br />

Allergy and <strong>Immunology</strong>, La Jolla, CA<br />

Type 1 Diabetes (T1D) is an autoimmune disease that<br />

results from the selective destruction <strong>of</strong> insulin-producing<br />

beta cells in the pancreas. While viral infections may be<br />

capable <strong>of</strong> triggering autoimmunity in genetically susceptible<br />

individuals, accumulating evidence indicates that viruses<br />

can also prevent T1D. Furthermore, viruses have been<br />

suggested to be potent inducers <strong>of</strong> CD4+CD25+ regulatory T<br />

cells (Tregs), which are known to play a crucial role in the<br />

prevention <strong>of</strong> autoimmunity. However, how viral infections<br />

may protect from autoimmune diabetes is still under<br />

investigation. Here, we show that activation <strong>of</strong> CD4+CD25+<br />

Tregs by acute lymphocytic choriomeningitis virus (LCMV)<br />

infection provides these cells with the capacity to prevent<br />

both spontaneous and virally induced T1D in the mouse. We<br />

found that CD4+CD25+ Tregs activated by LCMV in vivo or in<br />

vitro produce the regulatory cytokine TGF-β and are capable<br />

<strong>of</strong> protecting NOD and RIP-LCMV mice from spontaneous and<br />

LCMV-induced diabetes, respectively. Our results suggest<br />

that LCMV-modulated CD4+CD25+ Tregs act in a bystander<br />

fashion independent <strong>of</strong> beta-cell antigens and suppress TNFα<br />

production by autoreactive CD8+ T cells, leading to<br />

prevention <strong>of</strong> T1D. These findings provide a novel mechanistic<br />

explanation for the previously reported ability <strong>of</strong> viral<br />

infections to prevent autoimmune diabetes. Furthermore,<br />

our results support a model explaining the differential ability<br />

<strong>of</strong> viral infections to modulate diabetes. Activation <strong>of</strong> Tregs<br />

during viral infections may be a general feature accounting<br />

for the ability <strong>of</strong> viruses to prevent T1D as well as other<br />

autoimmune disorders.<br />

doi:10.1016/j.clim.2007.03.350<br />

<strong>Oral</strong> <strong>Presentations</strong>: Saturday, June 9<br />

Saturday, June 9<br />

2:45 pm−4:45 pm<br />

OR.33 Cytotoxic Human IL-22-expressing Th17<br />

Lymphocytes Promote Immune Cell Migration Into<br />

the Central Nervous System<br />

Hania Kebir, PhD Student, CHUM-Notre-Dame Hospital,<br />

Neuroimmunology Department, Montreal, QC, Canada, Igal<br />

Ifergan, PhD Student, CHUM-Notre-Dame Hospital,<br />

Neuroimmunology Department, Montreal, QC, Canada,<br />

Aurore Dodelet-Devillers, M.Sc. Student, CHUM-Notre-Dame<br />

Hospital, Neuroimmunolgy Department, Montreal, QC,<br />

Canada, Fabrizio Giuliani, Assistant Pr<strong>of</strong>essor <strong>of</strong> Medicine,<br />

University <strong>of</strong> Alberta, Department <strong>of</strong> Medicine, Edmonton,<br />

AB, Canada, Romain Cayrol, PhD Student, CHUM-Notre-Dame<br />

Hospital, Neuroimmunolgy Department, Montreal, QC,<br />

Canada, Nathalie Arbour, Assistant Pr<strong>of</strong>essor <strong>of</strong> Medicine,<br />

CHUM-Notre-Dame Hospital, Neuroimmunology<br />

Department, Montreal, QC, Canada, Alexandre Prat,<br />

Assistant Pr<strong>of</strong>essor <strong>of</strong> Medicine and Doctor,<br />

CHUM-Notre-Dame Hospital, Neuroimmunolgy Department<br />

and MS Clinic, Montreal, QC, Canada<br />

Interleukin (IL)-17-secreting lymphocytes (Th17) appear<br />

essential in the pathogenesis <strong>of</strong> numerous models <strong>of</strong><br />

inflammatory diseases, including experimental autoimmune<br />

encephalomyelitis (EAE). As survival <strong>of</strong> Th17 cells<br />

is attributed to IL-23, recent evidence suggests that<br />

transforming growth factor (TGF)-β is required for the<br />

expression <strong>of</strong> IL-23 receptor (R) by lymphocytes, thereby<br />

acting upstream in the development <strong>of</strong> Th17. However,<br />

while IL-23 and TGF-β R null animals are resistant to<br />

autoimmune diseases, IL-17 neutralization only partially<br />

protects against EAE and colitis, suggesting that additional<br />

Th17-derived factors contribute to tissue inflammation. We<br />

demonstrate that human Th17 lymphocytes originate from<br />

memory CD4+CD45RO+ cells and express IL-22 under the<br />

influence <strong>of</strong> IL-23, while TGF-β acts as a negative regulator<br />

<strong>of</strong> IL-22 production. We show that IL-17R and IL-22R are upregulated<br />

on blood–brain barrier (BBB)-endothelial cells<br />

(ECs) in situ during inflammation, and that IL-17 and IL-22<br />

disrupt the human BBB and promote CD4+ T lymphocyte<br />

recruitment in vitro. Furthermore, Th17 lymphocytes transmigrate<br />

more readily across brain-ECs than Th1 and<br />

express high levels <strong>of</strong> granzyme B. Thus, IL-23 induces<br />

the formation <strong>of</strong> a unique Th subset expressing cytotoxic<br />

factors and promoting leukocyte recruitment across the<br />

BBB, through the concerted and redundant effects <strong>of</strong> IL-17<br />

and IL-22.<br />

doi:10.1016/j.clim.2007.03.351


Abstracts<br />

OR.34 CNS Myeloid Dendritic Cells Drive Epitope<br />

Spreading and Th-17 Differentiation in Relapsing<br />

Experimental Autoimmune Encephalomyelitis<br />

(R-EAE)<br />

Stephen Miller, Pr<strong>of</strong>essor, Northwestern University Medical<br />

School, Department <strong>of</strong> Microbiology-<strong>Immunology</strong>, Chicago,<br />

IL, Samantha Bailey, Postdoctoral Fellow, Northwestern<br />

University Medical School, Department <strong>of</strong><br />

Microbiology-<strong>Immunology</strong>, Chicago, IL, Bettina Schreiner,<br />

Postdoctoral Fellow, Northwestern University Medical<br />

School, Department <strong>of</strong> Microbiology-<strong>Immunology</strong>, Chicago, IL<br />

Chronic progression <strong>of</strong> relapsing experimental autoimmune<br />

encephalomyelitis (R-EAE) is dependent on the activation<br />

<strong>of</strong> T cells to released endogenous myelin epitopes, i.e.,<br />

epitope spreading. Using transfer <strong>of</strong> naive CFSE-labeled TCR<br />

transgenic T cells, we have shown that activation <strong>of</strong> naive<br />

PLP139–151-specific T cells in SJL mice undergoing PLP178-<br />

191-induced R-EAE occurs directly in the CNS. Flow cytometric<br />

and histologic examination <strong>of</strong> the R-EAE CNS revealed<br />

the infiltration <strong>of</strong> significant numbers <strong>of</strong> CD11c+ dendritic<br />

cells (DCs) (including myeloid, lymphoid and plasmacytoid<br />

subsets) which are not seen in the healthy CNS. Functional<br />

examination <strong>of</strong> the antigen presentation capacity <strong>of</strong> CNS APC<br />

populations shows that only F4/80-CD11c+CD45hi DCs, not<br />

infiltrating macrophages or resident microglia, efficiently<br />

present endogenous antigen resulting in the activation <strong>of</strong><br />

naive PLP139–151-specific Tg T cells. Bone marrow chimeras<br />

indicate that the vast majority <strong>of</strong> CNS DCs are <strong>of</strong> peripheral<br />

origin. Among the DC subsets, CD11b+ CD11c+ myeloid DCs<br />

(mDCs) cluster with CNS-resident PLP139–151-specific Tg T<br />

cells and are by far the most efficient endogenous activators<br />

<strong>of</strong> naumlautve T cells both in vivo and in vitro. In contrast,<br />

lymphoid and plasmacytoid DCs are less effective. mDCs<br />

uniquely biased Th-17 and not Th1 differentiation, correlating<br />

with their enhanced expression <strong>of</strong> TGF- 2 1 and IL-6 and IL-<br />

23. These findings indicate that the inflamed CNS can<br />

function as a neo-lymphoid organ and inhibition <strong>of</strong> DC<br />

migration may be a viable target for treatment <strong>of</strong> MS<br />

(supported by NIH Grant NS-030871, NMSS Grant RG-3793-A-<br />

7 and a grant from the Myelin Repair Foundation).<br />

doi:10.1016/j.clim.2007.03.352<br />

OR.35 IL-23 and IL-17 in Pathogenesis <strong>of</strong><br />

Experimental Ocular Autoimmunity: Requirement<br />

for IL-23 May Extend Beyond Its Role in Sustaining<br />

the IL-17 Effector Response<br />

Dror Luger, Postdoctoral Fellow, National Institutes <strong>of</strong><br />

Health, NEI Laboratory, Phyllis Silver, Biologist, National<br />

Institutes <strong>of</strong> Health, NEI Laboratory, Daniel Cua, Senior<br />

Principal Scientist, Schering Plough Biopharma, DNAX, Palo<br />

Alto, CA, Zoe Chen, Scientist, Schering Plough Biopharma,<br />

Palo Alto, CA, Yoichiro Iwakura, Pr<strong>of</strong>essor, University <strong>of</strong><br />

Tokyo, Tokyo, Japan, Edward Bowman, Principal Scientist,<br />

Schering-Plough Biopharma, DNAX, Palo Alto, CA, Chi-Chao<br />

Chan, Senior Scientist, National Institutes <strong>of</strong> Health, NEI,<br />

Laboratory <strong>Immunology</strong>, Bethesda, MD, Rachel Caspi, Senior<br />

Investigator, National Institutes <strong>of</strong> Health, NEI, Laboratory<br />

<strong>Immunology</strong>, Bethesda, MD<br />

Experimental autoimmune uveitis (EAU) represents autoimmune<br />

uveitis in humans. Here we examined the role <strong>of</strong> the<br />

IL-23/IL-17 pathway in EAU. We immunized IL-23p19, IL-<br />

12p35, IL-12p40, IFN-g and IL-17 KO mice with the<br />

uveitogenic Ag IRBP. Alternatively, we treated wild type<br />

mice immunized for EAU with Abs to IL-23p19 or to IL-17<br />

throughout the disease course (prevention), or only during<br />

the effector phase (reversal). IL-23p19 KO were resistant to<br />

EAU, showing reduced Ag-specific DTH, IL-2, IFN-g, IL-17, IL-<br />

1b, IL-6 and IL-5. In contrast, IL-12p35 and IFN-g KO mice<br />

developed exacerbated EAU, DTH and IL-17 responses.<br />

Treatment with anti-IL-23p19 prevented EAU and reduced<br />

immunological responses, but was unable to reverse an<br />

ongoing disease process, indicating an inductive role for IL-<br />

23. In contrast, anti-IL-17 prevented as well as reversed EAU,<br />

indicating a role for IL-17 in effector mechanisms. Interestingly,<br />

severe EAU could be induced with a Th1 cell line that<br />

does not produce IL-17. EAU in recipients <strong>of</strong> the line could be<br />

inhibited with anti-IFN-g or anti-TNF-a, but not with anti-IL-<br />

17, speaking against a need for host-derived IL-17 and<br />

suggesting that under some conditions an antigen-specific<br />

Th1 effector is sufficient to induce EAU. In keeping with this,<br />

IL-17 KO mice were still susceptible to disease. These data<br />

raise the possibility that the nonredundant need for IL-23 in<br />

EAU may extend beyond its role in promoting the Th17<br />

effector response and point to IL-23 and IL-17 as new<br />

therapeutic targets for uveitis.<br />

doi:10.1016/j.clim.2007.03.353<br />

S61<br />

OR.36 The Role <strong>of</strong> IL-17 and IFN-γ in Defining<br />

Pathogenic Potential <strong>of</strong> T Cells in EAE<br />

Ingunn Stromnes, Graduate Student, University <strong>of</strong><br />

Washington, Department <strong>of</strong> <strong>Immunology</strong>, Seattle, WA, Joan<br />

Goverman, Pr<strong>of</strong>essor, University <strong>of</strong> Washington,<br />

Department <strong>of</strong> <strong>Immunology</strong>, Seattle, WA, Denny Liggitt,<br />

Chair, University <strong>of</strong> Washington, Department <strong>of</strong><br />

Comparative Medicine, Seattle, WA<br />

Multiple sclerosis (MS) is a demyelinating disease <strong>of</strong> the<br />

central nervous system (CNS) in which inflammatory infiltrates<br />

localize in the white matter <strong>of</strong> the brain and spinal<br />

cord. The mechanisms that determine specific sites <strong>of</strong><br />

inflammation are not understood. We developed a novel<br />

model <strong>of</strong> experimental allergic encephalomyelitis, an animal<br />

model for MS, in which T cells specific for one epitope <strong>of</strong> a<br />

myelin oligodendrocyte glycoprotein (MOG) preferentially<br />

induce inflammation in the brain, while T cells specific for<br />

two different MOG epitopes preferentially induce inflammation<br />

in the spinal cord, resulting in pr<strong>of</strong>oundly distinct<br />

clinical signs. T cells responding to all three epitopes<br />

generate both IL-17- and IFN-γ-producing non-overlapping<br />

populations, and both the IL-17- and IFN-γ-producing T cells<br />

induce disease upon adoptive transfer. We found that the<br />

ability to induce inflammation preferentially in the brain or<br />

spinal cord is not determined by the absolute number <strong>of</strong><br />

either IL-17- or IFN-γ-producing T cells, but rather by the<br />

ratio <strong>of</strong> the two populations. Changing the IL-17 to IFN-γ<br />

ratio for Tcells <strong>of</strong> each specificity by culturing them in either<br />

IL-12 or IL-23 prior to adoptive transfer altered the


S62 Abstracts<br />

inflammation sites in the CNS, which resulted in different<br />

clinical signs in recipient mice. These data show that the<br />

encephalitogenic potential <strong>of</strong> Tcells is not defined solely by a<br />

production <strong>of</strong> a single cytokine, but by the relative frequency<br />

<strong>of</strong> Tcells producing either IFN-γ or IL-17. These findings have<br />

important implications for cytokine-targeted therapies <strong>of</strong><br />

CNS autoimmune disease.<br />

doi:10.1016/j.clim.2007.03.354<br />

OR.37 The Role <strong>of</strong> IL-22, a TH17 Cytokine, in<br />

Autoimmune Diseases and Bacteria Infection<br />

Yan Zheng, Postdoctoral Research Fellow, Department <strong>of</strong><br />

<strong>Immunology</strong>, Genentech Inc, South San Francisco, CA,<br />

Dimitry Danilenko, Pathologist, Department <strong>of</strong> Pathology,<br />

Genentech Inc, South San Francisco, CA, Patricia Valdez,<br />

Senior Research Associate, Department <strong>of</strong> <strong>Immunology</strong>,<br />

Genentech Inc, South San Francisco, CA, Jeffrey<br />

Eastham-Anderson, Department <strong>of</strong> Pathology, Genentech<br />

Inc, South San Francisco, CA, Ian Kasman, Department <strong>of</strong><br />

Pathology, Genentech Inc, South San Francisco, CA, Jianfeng<br />

Wu, Research Associate, Department <strong>of</strong> <strong>Immunology</strong>,<br />

Genentech Inc, South San Francisco, CA, Wenjun Ouyang,<br />

Scientist, Department <strong>of</strong> <strong>Immunology</strong>, Genentech Inc, South<br />

San Francisco, CA<br />

Recently, interleukin (IL)-23, a cytokine involved in the<br />

development <strong>of</strong> IL-17-producing T helper cells (TH17 cells),<br />

was found to have a potential function in the pathogenesis <strong>of</strong><br />

autoimmune diseases and the host defense against infectious<br />

agents. We identified that IL-22 is preferentially produced by<br />

TH17 cells. However, the production <strong>of</strong> IL-22 and IL-17 from<br />

TH17 cells is differentially regulated. Transforming growth<br />

factor-beta (TGF-β), although crucial for IL-17 production,<br />

actually inhibits IL-22 production. Furthermore, IL-22 mediates<br />

IL-23-induced acanthosis and dermal inflammation<br />

through the activation <strong>of</strong> Stat3 (signal transduction and<br />

activators <strong>of</strong> transcription 3) in vivo. Our preliminary data<br />

also showed a protective role <strong>of</strong> IL-22 in host defense against<br />

bacteria infection. Our results suggest that TH17 cells,<br />

through the production <strong>of</strong> both IL-22 and IL-17, might have<br />

essential functions both in host defense and in the pathogenesis<br />

<strong>of</strong> autoimmune diseases such as psoriasis. IL-22, as an<br />

effector cytokine produced by Tcells, mediates the crosstalk<br />

between the immune system and the tissues.<br />

doi:10.1016/j.clim.2007.03.355<br />

OR.38 Endogenous IL-6 is Required for Inducing and<br />

Sustaining an Antigen-specific IL-17 Memory<br />

Response in Experimental Autoimmune Uveitis<br />

(EAU), but Not for Innate IL-17 Production<br />

Aleksandra V. Rachitskaya, Medical Student, Laboratory <strong>of</strong><br />

<strong>Immunology</strong>, National Eye Institute, NIH; HHMI-NIH<br />

Research Scholars Program, Bethesda, MD, Anna M. Hansen,<br />

Postdoctoraltoral Fellow, Laboratory <strong>of</strong> <strong>Immunology</strong>,<br />

National Eye Institute, National Institutes <strong>of</strong> Health (NIH),<br />

Bethesda, MD, Rajeev K. Agarwal, Staff Scientist,<br />

Laboratory <strong>of</strong> <strong>Immunology</strong>, National Eye Institute, National<br />

Institutes <strong>of</strong> Health (NIH), Bethesda, MD, Dror Luger,<br />

Postdoctoraltoral Fellow, Laboratory <strong>of</strong> <strong>Immunology</strong>,<br />

National Eye Institute, National Institutes <strong>of</strong> Health (NIH),<br />

Bethesda, MD, Phyllis B. Silver, Biologist, Laboratory <strong>of</strong><br />

<strong>Immunology</strong>, National Eye Institute, National Institutes <strong>of</strong><br />

Health (NIH), Bethesda, MD, Chi-Chao Chan, Senior<br />

Investigator, Laboratory <strong>of</strong> <strong>Immunology</strong>, National Eye<br />

Institute, National Institutes <strong>of</strong> Health (NIH), Bethesda, MD,<br />

Rachel R. Caspi, Senior Investigator, Laboratory <strong>of</strong><br />

<strong>Immunology</strong>, National Eye Institute, National Institutes <strong>of</strong><br />

Health (NIH), Bethesda, MD<br />

We examined the role <strong>of</strong> IL-6 in production <strong>of</strong> the<br />

proinflammatory cytokine IL-17 under conditions <strong>of</strong> a primary<br />

immune response, as represented by T cell receptor ligation<br />

in vitro, and under conditions <strong>of</strong> antigen-specific recall<br />

response in the model <strong>of</strong> experimental autoimmune uveitis<br />

(EAU) in vivo. To mimic a primary response, naive splenocytes<br />

from IL-6 deficient mice (IL-6 KO) or from their C57BL/6 wildtype<br />

counterparts (WT) were stimulated with anti-CD3 and IL-<br />

23. EAU was induced in IL-6 KO and WT mice by immunization<br />

with the retinal antigen IRBP in complete Freund’s adjuvant<br />

(CFA). EAU development was evaluated by fundus examination<br />

and confirmed by eye histology. Eyes and lymphoid organs<br />

were harvested on day 21. Responses were examined by IL-17<br />

ELISA, intracellular cytokine staining combined with immunophenotyping,<br />

DTH and lymphocyte proliferation. IRBPimmunized<br />

IL-6 KO mice were completely resistant to EAU and<br />

had significantly reduced DTH and proliferative responses to<br />

IRBP as compared to their WT counterparts. Notably, their<br />

IRBP-specific IL-17 production was conspicuously decreased<br />

as compared to WT. In contrast, during an in vitro<br />

primary response, IL-17 production, observed at approximately<br />

24 hours after stimulation, was the same in both<br />

genotypes and required IL-23, but not IL-6. Our results<br />

indicate that IL-6 plays an essential role in inducing and<br />

sustaining the IL-17 memory and recall responses. However,<br />

an initial innate IL-17 production occurs early during<br />

a primary response and is IL-6 independent. By immunophenotyping,<br />

the cellular source <strong>of</strong> this early IL-17<br />

response appears to be NK/NKT cells.<br />

doi:10.1016/j.clim.2007.03.356<br />

OR.39 Evidence for a Role <strong>of</strong> the Interleukin-23<br />

Pathway in the Pathogenesis <strong>of</strong> Psoriasis<br />

Frank Nestle, Pr<strong>of</strong>essor, Kings College London, London,<br />

England, Francesca Capon, Doctor, Department <strong>of</strong> Genetics,<br />

King’s College London School <strong>of</strong> Medicine, London, England,<br />

Jonathan Barker, Pr<strong>of</strong>essor, St. Johns Institute <strong>of</strong><br />

Dermatology, London, England, Robert Kastelein, Staff,<br />

Schering-Plough Biopharma, Palo Alto, CA, Richard<br />

Trembath, Pr<strong>of</strong>essor, Division <strong>of</strong> Genetics and Molecular<br />

Medicine, London, England, Giulia Tonel, PhD Student,<br />

Department <strong>of</strong> Dermatology (F14), University <strong>of</strong> Zurich,<br />

Zurich, Switzerland, Paola Di Meglio, Research Associate,<br />

Cutaneous Medicine and Immunotherapy Unit, St. John’s<br />

Institute <strong>of</strong> Dermatology, London, England, Elizabeth<br />

Oldham, Staff, Schering-Plough Biopharma, Palo Alto, CA,<br />

Jerry Lanchbury, Staff, Myriad Genetics, Salt Lake City, UT


Abstracts<br />

To identify genes relevant to disease pathogenesis we<br />

performed a genome-wide association study in psoriasis, one<br />

<strong>of</strong> the most common chronic inflammatory disorders. We<br />

detected a highly significant association between psoriasis<br />

and genetic markers in the interleukin-23 receptor (IL-23R)<br />

gene on chromosome 1p31, a finding replicated in an independent<br />

dataset. The most significantly associated polymorphism<br />

(combined p value=1.6 ×10 −5 ) results in an amino<br />

acid substitution (Arg381Gln) located in the IL-23R cytoplasmic<br />

domain. The same variant has recently been implicated<br />

in the pathogenesis <strong>of</strong> inflammatory bowel disease, supporting<br />

a critical role <strong>of</strong> IL-23 signaling in epithelial inflammation.<br />

As a necessary step to determine the importance <strong>of</strong> this<br />

pathway in the pathogenesis <strong>of</strong> psoriasis, we undertook functional<br />

investigations using patient samples and a clinically<br />

relevant model system. We found an increased expression <strong>of</strong><br />

IL-23R on psoriatic helper T cells compared to normal controls.<br />

Functional dissection <strong>of</strong> the IL-23 pathway using immunosuppressed<br />

AGR mice grafted with psoriatic skin<br />

revealed a key role for the IL-23 pathways in the disease<br />

process. Administration <strong>of</strong> a neutralizing anti-human IL-23<br />

antibody inhibited psoriasis development comparable to the<br />

use <strong>of</strong> anti-TNF-α blockers. These data provide genetic and<br />

functional evidence for a crucial role <strong>of</strong> the IL-23 pathway<br />

in cutaneous inflammation and lay the foundation for new<br />

treatment strategies in psoriasis and potentially other chronic<br />

epithelial inflammatory disorders.<br />

doi:10.1016/j.clim.2007.03.357<br />

OR.40 The Fully Human Antibody CNTO1275<br />

Effectively Inhibits Human IL-12 and IL-23 Mediated<br />

Th1 and Th17 Cell Function and Provides <strong>Clinical</strong><br />

Benefit to Patients with Plaque Psoriasis<br />

Jacqueline Benson, Assistant Director, Centocor Research<br />

and Development, Discovery Research, Radnor, PA,<br />

Yevgeniya Orlovsky, Senior Associate Scientist, Centocor<br />

Research and Development, Radnor, PA, Kim Staquet,<br />

Manager, Centocor Research and Development, Radnor,<br />

PA, Jinquan Luo, Principal Research Scientist, Centocor<br />

Research and Development, Radnor, PA, Patrick Branigan,<br />

Research Scientist, Centocor Research and Development,<br />

Radnor, PA, Roberta Lamb, Senior Associate Scientist,<br />

Centocor Research and Development, Radnor, PA, Jian Li,<br />

Research Scientist, Centocor Research and Development,<br />

Radnor, PA, Renold Capocasale, Research Scientist,<br />

Centocor, Radnor, PA, Janine Huber, Senior Associate<br />

Scientist, Therakos, Exton, PA, Bernie Scallon, Sr<br />

Research Fellow, Centocor, Radnor, PA, David Peritt,<br />

Director, Therakos, Exton, PA, David Shealy, Senior<br />

Research Fellow, Centocor Research and Development,<br />

Radnor, PA, George Heavner, Distinguished Research<br />

Fellow, Centocor Research and Development, Radnor, PA,<br />

Jill Giles-Komar, Director, Centocor Research and<br />

Development, Radnor, PA, Tom Nesspor, Senior Associate<br />

Scientist, Centocor, Radnor, PA<br />

Interleukins (IL)-12 and IL-23 are potent contributors to<br />

CD4+ T cell differentiation to Th1 and Th17 cell types which<br />

are associated with several immune-mediated human dis-<br />

eases, including psoriasis, Crohn’s disease, rheumatoid<br />

arthritis, multiple sclerosis, and others. Unique TLR agonist<br />

combinations will elicit IL-12 versus IL-23 production from<br />

antigen presenting cells and these cytokines demonstrate<br />

distinct effects on human and mouse T cell activation.<br />

CNTO1275 is a fully human monoclonal antibody that<br />

potently inhibits the bioactivity <strong>of</strong> human IL-12 and IL-23<br />

by binding to the shared p40 subunit and preventing<br />

interaction with the IL-12Rb1 receptor on the surface on<br />

NK and CD4+ T cells. Thus, CNTO1275 inhibits IL-12 and IL-23<br />

mediated activation and cytokine production <strong>of</strong> receptorbearing<br />

cells. CNTO1275 binds human and primate, but not<br />

rodent, IL-12 and IL-23 and suppressed disease in a primate<br />

model <strong>of</strong> multiple sclerosis and in a human skin transplant<br />

mouse model <strong>of</strong> psoriasis. CNTO1275 has also provided a<br />

significant level <strong>of</strong> efficacy in the majority <strong>of</strong> subjects in<br />

phase I and II clinical studies <strong>of</strong> plaque psoriasis. These data<br />

support a central role for IL-12/23p40 in psoriasis pathogenesis.<br />

CNTO1275 was generally well-tolerated and adverse<br />

events observed in these studies did not indicate dose–<br />

response safety relationships. Similarly, studies in cynomolgus<br />

monkeys have shown that even high doses <strong>of</strong> CNTO1275<br />

are generally well-tolerated. Collectively, these results<br />

suggest that CNTO1275 may represent an exciting first-inclass<br />

therapy with significant therapeutic potential for<br />

immune-mediated diseases such as psoriasis.<br />

doi:10.1016/j.clim.2007.03.358<br />

Therapy- From Gene Silencing to Costimulation<br />

Saturday, June 9<br />

2:45 pm–4:45 pm<br />

S63<br />

OR.41 Effects <strong>of</strong> Long-Term Fingolimod Therapy on<br />

Circulating Mononuclear Cell Populations in Multiple<br />

Sclerosis<br />

Amit Bar-Or, Associate Pr<strong>of</strong>essor, Montreal Neurological<br />

Institute, McGill University, Montreal, QC, Canada, Nathalie<br />

Arbour, Associate Researcher, University <strong>of</strong> Montreal,<br />

Faculty <strong>of</strong> Medicine, Montreal, QC, Canada, Ellie McCrae,<br />

Technician, Montreal Neurological Institute, Montreal, QC,<br />

Canada, Jack Antel, Pr<strong>of</strong>essor, Montreal Neurological<br />

Institute, Montreal, QC, Canada, Tarik Touil, Postdoctoral<br />

Fellow, Montreal Neurological Institute, Montreal, QC,<br />

Canada, Karen Newell, Visiting Pr<strong>of</strong>essor, Montreal<br />

Neurological Institute, Montreal, QC, Canada<br />

Fingolimod (FTY720) is an orally administered sphingosine-1-phosphate<br />

(S1P) receptor modulator. In animal and<br />

short-term human studies it down-regulates lymphocyte (but<br />

not myeloid cell) receptor expression. Fingolimod reduced<br />

clinical and MRI activity in relapsing MS patients. Objectives:<br />

To examine the pr<strong>of</strong>ile <strong>of</strong> circulating mononuclear cells<br />

(MNC) in MS patients (n=6) who continue daily use <strong>of</strong> 1.25 mg<br />

<strong>of</strong> oral fingolimod for N18 months (open label extension<br />

phase <strong>of</strong> original phase II trial). This represents the longest<br />

experience using fingolimod as a single agent in humans.<br />

Methods: Whole peripheral blood samples obtained from the<br />

patient cohort were immunostained for leukocyte markers


S64 Abstracts<br />

and analyzed by flow cytometry. Results: Absolute lymphocyte<br />

number (mean 0.48+0.06 ×10 3 per microliter) was<br />

reduced with treatment (normal range 0.91–4.28×103);<br />

while CD14+ monocyte counts (0.47 +0.07×103) were within<br />

normal range (0.12–0.92×103). CD4+ T cells were relatively<br />

more depleted (2.3+0.8% <strong>of</strong> total MNCs) (control 28.0+6.4%,<br />

n=6) than CD8+ T cells (10.4 +2.5%) (control 22.0+4.2%),<br />

resulting in reversed CD4:CD8 ratio (0.2). CD19+ B cells were<br />

also markedly reduced (1.1+0.2% <strong>of</strong> total MNCs) (control<br />

11.1+2.1%). The proportion <strong>of</strong> monocytes (29.4+4.4%) and<br />

CD56+ NK cells (32.2+9.3%) among MNC was increased in<br />

treated patients compared to controls (7.0+1.8%) and (14.5 +<br />

4.3%) respectively — although their absolute number did not.<br />

Conclusions: MS patients receiving fingolimod monotherapy<br />

show sustained decreases in circulating B cells and CD4 T<br />

cells, relative less depletion <strong>of</strong> CD8 T cells, and maintained<br />

numbers <strong>of</strong> innate immune cells. Correlations with clinical<br />

measures will help define the basis for the efficacy, and<br />

safety pr<strong>of</strong>ile <strong>of</strong> this type <strong>of</strong> immuno-modulation.<br />

doi:10.1016/j.clim.2007.03.359<br />

OR.42 In Vivo Dendritic Cell-specific Gene Silencing<br />

Using a Novel and Selective siRNA Delivery Method<br />

to Induce Immune Modulation<br />

Costin Vladau, Graduate Student, University <strong>of</strong> Western<br />

Ontario, Department <strong>of</strong> Microbiology and <strong>Immunology</strong>,<br />

London, ON, Canada, Dong Chen, Postdoctoral Fellow,<br />

University <strong>of</strong> Western Ontario, Department <strong>of</strong> Microbiology<br />

and <strong>Immunology</strong>, London, ON, Canada, Motohiko Suzuki,<br />

Postdoctoral Fellow, University <strong>of</strong> Western Ontario,<br />

Department <strong>of</strong> Microbiology and <strong>Immunology</strong>, London,<br />

England, Xusheng Zhang, Postdoctoral Fellow, University <strong>of</strong><br />

Western Ontario, Department <strong>of</strong> Microbiology and<br />

<strong>Immunology</strong>, London, ON, Canada, Xiufen Zheng,<br />

Postdoctoral Fellow, London Health Sciences Centre,<br />

University Hospital, Department <strong>of</strong> Surgery, London, ON,<br />

Canada, Wei-Ping Min, Principle Investigator, University <strong>of</strong><br />

Western Ontario, Department <strong>of</strong> Microbiology and<br />

<strong>Immunology</strong>, London, ON, Canada<br />

Silencing immune molecules, such as the costimulatory<br />

molecule CD40, using siRNA was shown to have therapeutic<br />

potential and promise in immune modulation. However, a<br />

major barrier to the clinical application <strong>of</strong> siRNA is the current<br />

lack <strong>of</strong> an effective and cell-specific delivery system. Herein,<br />

we present a new method <strong>of</strong> selectively delivering siRNA to DC<br />

in vivo using stealth immunoliposomes (SILs). CD40 siRNAcontaining<br />

SILs were generated using 4 types <strong>of</strong> lipids and<br />

decorated with surface-bound, DC-specific mAbs. DC-specific<br />

binding capacity <strong>of</strong> SILs was demonstrated by fluorescence<br />

microscopy and flow cytometry. Upon treatment with CD40<br />

siRNA-SILs, DC expression <strong>of</strong> CD40 was successfully suppressed<br />

in vitro. In vivo administration <strong>of</strong> CD40 siRNA-SILs resulted in<br />

DC-specific tissue targeting, as evidenced by increased uptake<br />

<strong>of</strong> fluorescence-tagged siRNA. Additionally, DC from mice<br />

treated with CD40 siRNA-SILs exhibited CD40-specific gene<br />

silencing in vivo. Tolerogenic properties <strong>of</strong> DC treated with<br />

CD40 siRNA-SILs were determined by inhibition <strong>of</strong> allogeneic T<br />

cell proliferation in MLR. Furthermore, a strong in vivo<br />

immune modulation was observed in mice treated with CD40<br />

siRNA-SILs. In conclusion, this is the first demonstration <strong>of</strong> DCspecific<br />

siRNA delivery and gene silencing in vivo, which<br />

highlights the potential <strong>of</strong> DC-mediated immune modulation<br />

and the feasibility <strong>of</strong> siRNA based clinical therapy.<br />

doi:10.1016/j.clim.2007.03.360<br />

OR.43 Immunotherapy <strong>of</strong> Solid Tumors Utilizing<br />

PLGA/Tumor Lysate Nanoparticles to Stimulate TIL<br />

and Circulating CD8+ Tumor-specific T Cells<br />

Douglas Hanlon, Associate Research Scientist, Yale<br />

Dermatology, New Haven, CT, Virginia Cody, Research<br />

Assistant, Yale Dermatology, New Haven, CT, Shashi Prasad,<br />

Postdoctoraltoral Fellow, Yale Dermatology, New Haven, CT,<br />

Mark Saltzman, Pr<strong>of</strong>essor, Yale Biomedical Engineering, New<br />

Haven, CT, Camille Solbrig, Associate Research Scientist,<br />

Yale Biomedical Engineering, New Haven, CT<br />

Dendritic cell (DC)-based therapies for solid tumors have<br />

failed to achieve large-scale clinical utilization because at<br />

present no unified technology exists to overcome two fundamental<br />

obstacles. These are the lack <strong>of</strong> known tumor-associated<br />

antigens (TAA) from most solid malignancies and the lack <strong>of</strong><br />

efficient methodologies to transfer antigen from tumor to DC.<br />

Here we describe the development <strong>of</strong> an alternative technology<br />

for antigen delivery that appears to transfer a broad spectrum <strong>of</strong><br />

tumor antigens to DC with extremely high efficiency. We used<br />

this methodology, termed “nanoparticle (NP)-mediated tumor<br />

delivery,” to encapsulate whole melanoma protein lysates into<br />

PLGA biodegradable microspheres and targeted these conjugates<br />

to DC. Optimization <strong>of</strong> Ag delivery was achieved by<br />

experimental determination <strong>of</strong> which polymer formulation, Ag<br />

source and release characteristics led to most potent DC antigen<br />

presentation. In both murine and human T cell stimulation<br />

assays, DC loaded with NP/tumor conjugates were superior to<br />

other Ag forms in stimulating anti-tumor T cells — including<br />

murine gp100-specific naive T cells from pmel transgenics and<br />

human TIL and blood CD8+ T cells from melanoma patients.<br />

Analysis <strong>of</strong> cytokine production by DC and T cell by cytometric<br />

bead array (CBA) assays indicated that NP-loaded DC drove<br />

increased production <strong>of</strong> Th1 and proinflammatory cytokines<br />

such as IFN-g and IL-2. Although melanoma Ags were utilized as<br />

controls, a major potential advantage <strong>of</strong> NP-based antigen<br />

delivery is the fact that antigens need not be defined prior to DC<br />

loading, opening the door to vaccine targeting <strong>of</strong> virtually any<br />

solid tumor.<br />

doi:10.1016/j.clim.2007.03.361<br />

OR.44 Safety and In Vivo Efficacy <strong>of</strong> Innate (Class R)<br />

and Broadly Reactive (Class B) Inhibitory<br />

Oligonucleotides (INH-ODN) in MRL-Fas/lpr Lupus<br />

Mice<br />

Petar Lenert, Assistant Pr<strong>of</strong>essor, Department <strong>of</strong> Internal<br />

Medicine, The University <strong>of</strong> Iowa, Iowa City, IA, Sarah<br />

Seyfer, BS, Internal Medicine, Iowa City, IA, Elizabeth R.<br />

Pareigis, BS, Internal Medicine, The University <strong>of</strong> Iowa, Iowa<br />

City, IA, Robert F. Ashman, Pr<strong>of</strong>essor, Internal Medicine, The<br />

University <strong>of</strong> Iowa, Iowa City, IA


Abstracts<br />

Several lines <strong>of</strong> evidence indicate that intracytoplasmic<br />

TLR receptors 7 and 9 play an important role in the<br />

pathogenesis <strong>of</strong> SLE, both in vitro and in vivo. For example,<br />

MRL-Fas/lpr mice lacking TLR9 have a shorter life span and<br />

develop more aggressive kidney disease. On contrary, mice<br />

lacking TLR7 have a milder disease. INH-ODNs containing<br />

nuclease-resistant phosphorothioate backbone (PS) were<br />

recently developed that can block TLR7 and/or TLR9<br />

activation at low nanomolar concentrations. Here we show<br />

that under certain stimulatory conditions prototypic TLR9specific<br />

INH-ODNs can also block TLR7 activation <strong>of</strong> mouse<br />

macrophages induced by several (but not all) TLR7 ligands<br />

like imiquimod, CL075, CL097, CL087 and loxoribine. Interestingly,<br />

in primary B cells, certain TLR7/8 ligands, but not<br />

phosphodiester (PO) CpG-ODNs, could escape inhibition<br />

when INH-ODNs were made with the natural PO backbone.<br />

Such INH-ODNs could actually enhance B cell G1–M entry,<br />

survival and polyclonal IgM secretion induced by suboptimal<br />

TLR7/8 stimulation. Both TLR7/8 inhibition and enhancement<br />

were independent <strong>of</strong> the TLR9 expression. We have<br />

recently modified these INH-ODNs to allow formation <strong>of</strong><br />

stable secondary structures. In vitro, these modified innate<br />

INH-ODNs (class R) showed higher potency for macrophages,<br />

dendritic cells and marginal zone B cells over follicular B<br />

cells. However, in vivo, both linear and Class R INH-ODNs<br />

promoted better survival and ameliorated spontaneous lupus<br />

disease in MRL-Fas/lpr mice. It remains to be determined<br />

whether this effect required TLR7/8 or TLR9. This work was<br />

supported by the NIH grant 1R56AI064736-01A2 and by a<br />

grant from the Carver Trust fund.<br />

doi:10.1016/j.clim.2007.03.362<br />

OR.45 Tumor Cell Lysate-specific DTH Reaction<br />

Correlates with Longer Survival and Disease<br />

Stability in Vaccinated Melanoma Patients<br />

Flavio Salazar-Onfray, PhD, Universidad de Chile, Santiago<br />

de Chile, Chile<br />

Dendritic cell (DCs)-based therapy has proven to be<br />

effective in patients with malignancies. However, an optimal<br />

immunization protocol using DCs and the best means for<br />

delivering antigens has not been yet described. In this study, 37<br />

patients with malignant melanoma in stage IV were vaccinated<br />

with autologous DCs pulsed with a melanoma cell lysate, to<br />

assess toxicity, immunological and clinical responses. The<br />

responses <strong>of</strong> the patients were analyzed by ELISPOT and<br />

Delayed type IV hypersensitivity reaction (DTH) against the<br />

melanoma cell lysate. After vaccination, 50% <strong>of</strong> tested<br />

patients (18 out <strong>of</strong> 37) showed a positive DTH against the<br />

melanoma cell lysate. All patients showed a positive DTH<br />

reaction against the used adjuvant KLH demonstrating that all<br />

patients were immunocompetents. Significant correlations<br />

were found between positive DTH responses against tumor<br />

lysate and both disease stability and post-vaccination survival<br />

on stage IV patients. In fact, the mean <strong>of</strong> post-vaccination<br />

survival was 15 months for all the vaccinated patients, double<br />

than the historical survival for stage IV melanoma patients.<br />

More important, positive DTH patients showed a postvaccination<br />

survival <strong>of</strong> 25 months compared to 8 months<br />

observed for the negative DTH group <strong>of</strong> patients. There<br />

were no toxicities associated with the vaccines or evidence<br />

<strong>of</strong> autoimmunity including vitiligo. Our data suggest that<br />

autologous DCs pulsed with tumour lysate may provide a<br />

standardized and widely applicable source <strong>of</strong> melanoma<br />

specific antigens for clinical use, it is safe and causes no<br />

significant side effects, demonstrating to be partially efficient<br />

at triggering effective anti-melanoma immunity.<br />

doi:10.1016/j.clim.2007.03.363<br />

OR.46 In Vivo CTLA4Ig Treatment Substantially<br />

Inhibits CD4+CD25+ Regulatory T Cell Turnover but<br />

Not Function<br />

Anita Tang, Surgical Resident, Division <strong>of</strong> Transplantation,<br />

Department <strong>of</strong> Surgery, University <strong>of</strong> Maryland School <strong>of</strong><br />

Medicine, Baltimore, MD, Modesta Ndejembi, Graduate<br />

Student, Department <strong>of</strong> Microbiology and <strong>Immunology</strong>,<br />

University <strong>of</strong> Maryland School <strong>of</strong> Medicine, Baltimore, MD,<br />

Donna Farber, Associate Pr<strong>of</strong>essor, Division <strong>of</strong><br />

Transplantation, Department <strong>of</strong> Surgery, University <strong>of</strong><br />

Maryland School <strong>of</strong> Medicine, Baltimore, MD<br />

CTLA4Ig and its variant, LEA29Y, target the CD28/B7<br />

costimulatory pathway and are highly effective treatments<br />

in rheumatoid arthritis and transplantation, respectively.<br />

Since CD28/B7 signaling has been implicated in regulatory T<br />

cell (Treg) generation, we investigated the effects <strong>of</strong><br />

CTLA4Ig on Treg phenotype, proliferation and function.<br />

BALB/c mice were administered CTLA4Ig (250 μg/dose)<br />

every other day for 3 doses, with IgG2a and PBS as controls.<br />

Splenic CD4 T cells were purified 3–14 days later and<br />

analyzed for CD25, CTLA4 and Foxp3 expression. Following<br />

CTLA4Ig treatment, CD4+Foxp3+ Tregs decreased 60% and<br />

CTLA4 expression by 30% compared to controls, with the<br />

most significant depletion (80%) in the CTLA4+Foxp3+ subset.<br />

Additionally, CD25 expression and CD25+Foxp3+ Tregs<br />

dropped 50%. Similar findings were observed in thymectomized<br />

mice, suggesting that CTLA4Ig mediates its effects on<br />

peripheral Tregs. To assess the mechanistic effects <strong>of</strong><br />

CTLA4Ig, CFSE-labeled CD4+CD25+ and CD4+ CD25− T cells<br />

were transferred into syngeneic mice receiving CTLA4Ig and<br />

harvested 6 days later. Interestingly, we noted extensive and<br />

rapid in vivo proliferation <strong>of</strong> CD4+CD25+ Tregs (62%)<br />

compared to the CD4+CD25− subset (10%) in controltreated<br />

mice, suggesting that Treg proliferation is an<br />

antigen-driven phenomenon. Moreover, we observed that<br />

this rapid turnover <strong>of</strong> Tregs was almost completely blocked<br />

by CTLA4Ig. Despite a decreased yield, expression <strong>of</strong> CTLA4<br />

and turnover after CTLA4Ig treatment, the remaining Tregs<br />

were functionally competent and suppressed CD4+CD25− T<br />

cell proliferation comparable to controls in co-culture<br />

assays. These findings may have important implications<br />

regarding the constitution <strong>of</strong> Treg subsets and their balance<br />

within non-regulatory populations in patients receiving<br />

CTLA4Ig and LEA29Y.<br />

doi:10.1016/j.clim.2007.03.364<br />

S65


S66 Abstracts<br />

OR.47 A Novel Anti-Tumor Vaccine Using IDO<br />

Silenced DC<br />

Xiufen Zheng, Postdoctoraltoral Fellow, University <strong>of</strong><br />

Western Ontario, London, ON, Canada, Bertha Garcia,<br />

Pr<strong>of</strong>essor, Departments <strong>of</strong> Surgery, Microbiology, Pathology,<br />

University <strong>of</strong> Western Ontario, London, ON, Canada, Costin<br />

Vladau, MS Student, Departments <strong>of</strong> Microbiology and<br />

<strong>Immunology</strong>, University <strong>of</strong> Western Ontario, London, ON,<br />

Canada, James Koropatnick, Pr<strong>of</strong>essor, Departments <strong>of</strong><br />

Microbiology and <strong>Immunology</strong>, Pathology, Oncology,<br />

University <strong>of</strong> Western Ontario, London, ON, Canada, Dong<br />

Chen, PDF, Departments <strong>of</strong> Surgery, University <strong>of</strong> Western<br />

Ontario, London, ON, Canada, Motohiko Suzuki1, PDF,<br />

Departments <strong>of</strong> Surgery, University <strong>of</strong> Western Ontario,<br />

London, ON, Canada, Mu Li, PDF, Departments <strong>of</strong> Surgery,<br />

University <strong>of</strong> Western Ontario, London, ON, Canada,<br />

Wei-ping Min, Pr<strong>of</strong>essor, Departments <strong>of</strong> Surgery,<br />

Microbiology and <strong>Immunology</strong>, Pathology, Oncology,<br />

University <strong>of</strong> Western Ontario, London, ON, Canada, Xusheng<br />

Zhang, Postdoctoral Fellow, Department <strong>of</strong> Surgery,<br />

University <strong>of</strong> Western Ontario, London, Canada<br />

Hyporesponsiveness is a major hallmark in dendritic cell<br />

(DC)-mediated anticancer immunotherapy. Indoleanime 2,3dioxygenase<br />

(IDO), an immunosuppressive molecule<br />

expressed by DC, is a critical factor mediating hyporesponsiveness<br />

to cancer immune therapy. We hypothesized that<br />

antisense silencing <strong>of</strong> IDO in DCs would enhance anticancer<br />

therapy. In this study, DC were cultured in vitro, exposed to<br />

melanoma B16 lysate, transfected with IDO siRNA, and<br />

injected into tail veins <strong>of</strong> C57/BL6 mice. Mice were then<br />

challenged with B16 tumor cells. The anticancer effects <strong>of</strong><br />

IDO-silenced DC therapy, as compared with non-silenced,<br />

conventional control DC vaccine, were evidenced by (i)<br />

postponed melanoma tumor onset time, and (ii) decreased<br />

tumor size. In addition, after immunization with IDOsilenced<br />

DC, the number <strong>of</strong> CD8+ T cells was significantly<br />

greater than in animals immunized with control RNA-treated<br />

DC and CD4+ and CD8+ T cell apoptosis in draining lymph<br />

nodes was significantly lower. Furthermore, immunization<br />

with IDO-silenced DC enhanced tumor antigen-specific T cell<br />

proliferation and CTL activity and decreased numbers <strong>of</strong> CD4<br />

+CD25+ Foxp3+ regulatory T cells (Treg). In conclusion, this<br />

study is the first to demonstrate a novel anti-tumor vaccine<br />

by silencing an immunosuppressive gene (IDO) in DC, which<br />

enhanced anti-tumor immunity, reduced T cell apoptosis and<br />

Treg cell formation, and prevented tumor growth. IDOsilenced<br />

DC has clinical potential as an immune-based therapy.<br />

doi:10.1016/j.clim.2007.03.365<br />

OR.48 Post-Transcriptional Regulation <strong>of</strong> T Cell<br />

Receptor zeta Chain in Systemic Lupus<br />

Erythematosus<br />

Vaishali R. Moulton, Postdoctoral Fellow, Beth Israel<br />

Deaconess Medical Center, Medicine, Boston, MA, Vasileios<br />

C. Kyttaris, Instructor, Beth Israel Deaconess Medical<br />

Center, Medicine, Boston, MA, Yuang-Taung Juang,<br />

Instructor, Beth Israel Deaconess Medical Center, Medicine,<br />

Boston, MA, George C. Tsokos, Pr<strong>of</strong>essor, Beth Israel<br />

Deaconess Medical Center, Medicine, Boston, MA<br />

Systemic lupus erythematosus (SLE) is a complex autoimmune<br />

disease characterized by the production <strong>of</strong> autoantibodies,<br />

immune complex deposition, T cell infiltration<br />

and abnormal production <strong>of</strong> cytokines all leading to<br />

inflammatory damage <strong>of</strong> target organs (joints, kidneys<br />

and brain). SLE T cells are deficient in their expression <strong>of</strong><br />

the T cell receptor (TCR)/CD3 zeta chain protein (CD3Z), a<br />

critical molecule for the initiation <strong>of</strong> intracellular signaling<br />

upon antigen encounter. This deficiency is partially due to<br />

differential splicing <strong>of</strong> the CD3Z mRNA; SLE T cells express<br />

an alternatively spliced form <strong>of</strong> CD3Z mRNA which is missing<br />

562 nucleotides (672 to 1233) within its 3′ untranslated<br />

region (UTR). Studies by our group have shown that this<br />

alternatively spliced CD3Z mRNA is unstable and has<br />

reduced translation efficiency. We have also demonstrated<br />

that two adenosine-uridine-rich elements (AREs) at positions<br />

705 and 985 in the 3′ UTR are independently essential<br />

for its stability and translation. We hypothesized that<br />

proteins binding to these two AREs may stabilize the CD3Z<br />

mRNA. Electrophoretic mobility shift assays showed several<br />

protein complexes in jurkat cell nuclear extracts binding to<br />

32P-labeled oligonucleotides containing the ARE (705)<br />

sequence, which were outcompeted in the presence <strong>of</strong><br />

similar unlabeled oligonucleotides. Using an oligonucleotide<br />

pulldown assay, mass spectrophotometry and peptide<br />

sequencing, we identified HuR as one <strong>of</strong> the putative<br />

proteins binding to the ARE (705) oligonucleotide. Immunoblotting<br />

with specific antibody confirmed this finding.<br />

These results reveal molecular mechanisms that may<br />

contribute to CD3Z mRNA stability and altered TCRZ chain<br />

expression in lupus.<br />

doi:10.1016/j.clim.2007.03.366<br />

Biomarkers & New Technologies<br />

Saturday, June 9<br />

2:45 pm–4:45 pm<br />

OR.49 Autoimmune Development in Phenotypic<br />

Type 2 Diabetes Patients<br />

Barbara Brooks-Worrell, Research Scientist, University <strong>of</strong><br />

Washington/VA Puget Sound Health Care System, Seattle,<br />

WA, Heba Ismail, Research Fellow, VA Puget Sound Health<br />

Care System, Seattle, WA, Michael Wotring, Research<br />

Scientist, University <strong>of</strong> Washington, Seattle, WA, A.U.<br />

Liemin, Data Processor, University <strong>of</strong> Washington/VA Puget<br />

Sound Health Care System, Seattle, WA, Crystal Kimmie,<br />

Research Scientist, VA Puget Sound Health Care System,<br />

Seattle, WA, Jamie Felton, Research Associate, VA Puget<br />

Sound Health Care System, Seattle, WA, Jerry Palmer,<br />

Pr<strong>of</strong>essor <strong>of</strong> Medicine, University <strong>of</strong> Washington/VA Puget<br />

Sound Health Care System, Seattle, WA<br />

Type 1 (T1DM) diabetes is an autoimmune disease,<br />

whereas type 2 (T2DM) diabetes is considered nonautoimmune.<br />

However, cross-sectional studies have identified<br />

autoimmune diabetes in 10–15% <strong>of</strong> phenotypic T2DM<br />

patients. In this study, we evaluated phenotypic autoimmune<br />

negative T2DM patients prospectively to determine<br />

the importance <strong>of</strong> repeated testing to detect autoimmune


Abstracts<br />

diabetes. In 40 phenotypic T2DM patients we determined<br />

autoantibody (IA-2/IAA/GAD), T cell status (cellular immunoblotting),<br />

cytokine responses (ELISPOT), fasting C-peptide,<br />

and glucagon-stimulated C-peptide responses at<br />

baseline and every 3 months for an average follow-up <strong>of</strong><br />

23.3 months (range 6–36 months). Of the 40 patients, 14/<br />

40 (35%) were positive for autoantibody (Ab) and/or T cell<br />

(T) reactivity at baseline while 26/40 (65%) were Ab-T−. Of<br />

the 26 patients Ab-T− at baseline, 8/26 (30.7%) remained<br />

Ab-T− stable (persistently negative), 4/26 patients demonstrated<br />

transient Ab or T cell positivity (1 positive time<br />

point during follow-up), while 14/26 developed stable<br />

(persistent positivity after conversion) Ab+ and/or T+. We<br />

have categorized the patients with stable negative or<br />

transient Ab and/or T reactivity as non-autoimmune nonprogressors<br />

(NANP) and the patients developing stable Ab<br />

and/or T cell positivity as progressors. We observed that<br />

the mean fasting and glucagon-stimulated C-peptide<br />

responses were significantly (pb0.0001) lower, the INF-g<br />

responses to sonicated islets and GAD were significantly<br />

higher (pb0.05), and the IL-10 responses (pb0.05) to islets<br />

were significantly lower in the progressors compared to<br />

NANP. These results demonstrate that continued follow-up<br />

<strong>of</strong> T2DM patients is needed to identify development <strong>of</strong><br />

autoimmunity in patients previously classified as nonautoimmune.<br />

doi:10.1016/j.clim.2007.03.367<br />

OR.50 Derangements in Cytokine-induced STAT<br />

Protein Phosphorylation in Human Systemic Lupus<br />

Erythematosus<br />

Veronika Sharp, Fellow, Stanford University Medical School,<br />

Palo Alto, CA, Paul J. Utz, Stanford University Medical<br />

School, Palo Alto, CA, G.P. Nolan, The Baxter Laboratory for<br />

Genetic Pharmacology, CA, OD Perez, The Baxter Laboratory<br />

for Genetic Pharmacology, CA<br />

Systemic lupus erythematosus (SLE) is an autoimmune<br />

disease with variable clinical manifestations, many <strong>of</strong> which<br />

are devastating to patients. Currently it is difficult to<br />

predict disease onset, severity, or response to therapy. New<br />

and better markers are needed in clinical practice for<br />

improved predictability. Phospho-specific flow cytometry is<br />

a powerful technique that allows for the analysis <strong>of</strong> several<br />

intracellular signaling networks in multiple cellular subsets<br />

simultaneously in response to functionally relevant stimuli.<br />

Using this technique we interrogated signaling through<br />

different members <strong>of</strong> the Jak-STAT and MAPK pathways in<br />

response to cytokines (including IFN α, IFN γ, IL-4, IL-6 and<br />

IL-10) in peripheral blood mononuclear cells (PBMC)<br />

isolated from 14 patients with SLE and 11 healthy controls.<br />

CD3, CD4, CD8 and CD33 were used as surface markers to<br />

differentiate T cells subsets, B cells and monocytes. While<br />

PBMC from healthy and diseased individuals had similar<br />

phosphorylation pr<strong>of</strong>iles in the resting state, there were<br />

remarkable differences in cellular response once cells were<br />

stimulated. Among other differential responses, hyperphosphorylation<br />

<strong>of</strong> some STAT proteins was observed in<br />

response to IFN α. This is consistent with prior studies<br />

implicating a role for type I interferons in SLE pathogenesis.<br />

Our data demonstrate the feasibility <strong>of</strong> analyzing several<br />

signaling networks simultaneously in viable PBMC subsets in<br />

response to an array <strong>of</strong> cytokines. These differences may<br />

not be visible when only whole and resting PBMC are used.<br />

This study takes us one step closer to defining individual<br />

biosignatures for SLE patients that would allow for more<br />

customized immunotherapy.<br />

doi:10.1016/j.clim.2007.03.368<br />

OR.51 Evidence for an Increase in the Functional<br />

Avidity <strong>of</strong> the GAD-reactive CD4+ T Cell Population<br />

Prior to Diabetes Onset<br />

Nathan Standifer, Postdoctoral Research Associate,<br />

Benaroya Research Institute, Seattle, WA, Gerald Nepom,<br />

Director, Benaroya Research Institute, Seattle, WA<br />

Type 1 diabetes (T1D) is an autoimmune disease characterized<br />

by the cell-mediated destruction <strong>of</strong> insulin-producing<br />

β cells <strong>of</strong> the pancreatic islets. Immunologically, islet<br />

destruction is marked by increased auto-antibody titers and<br />

the expansion <strong>of</strong> glutamic acid decarboxylase (GAD)-reactive<br />

CD4+ T cells in many pre-diabetic subjects. We hypothesized<br />

that the functional avidity <strong>of</strong> the GAD-reactive T cell<br />

population increases over time in pre-diabetic subjects as<br />

the islet β cell mass decreases. To test this, we cultured<br />

longitudinal peripheral blood lymphocyte samples drawn<br />

from auto-antibody+, at-risk, pre-diabetic or diabetic subjects<br />

and calculated the GAD-reactive CD4+ Tcell population<br />

ED50 values: the concentration <strong>of</strong> peptide that induces onehalf<br />

maximal interferon-γ secretion. Longitudinal samples<br />

from an at-risk subject exhibited the highest Tcell population<br />

ED50 values (4.26 1/4M and 8.76 1/4M) while the ED50 values<br />

<strong>of</strong> samples from a T1D patient drawn 4 or 5 years after<br />

diagnosis were substantially lower (2.24 nM and 48.30 nM<br />

respectively). Interestingly, the ED50 value <strong>of</strong> the GADreactive<br />

T cell population cultured from a pre-diabetic<br />

subject 6 years prior to diagnosis was significantly higher<br />

than that <strong>of</strong> a sample drawn 2 years prior to diagnosis (5.04<br />

¼M and 15.38 nM respectively). We also observed that the<br />

frequency <strong>of</strong> GAD-reactive, central-memory cells increased<br />

over time in the pre-diabetic samples. Our data imply that, as<br />

the islet β cell mass decreases, the GAD-reactive T cell<br />

population undergoes a phenotypic shift from effector to<br />

central memory cells concomitant with an increase in the<br />

functional avidity as measured by interferon-γ secretion.<br />

doi:10.1016/j.clim.2007.03.369<br />

S67<br />

OR.52 Peripheral Blood Flow Cytometric Evaluation<br />

<strong>of</strong> a Large Cohort <strong>of</strong> Hyper Immunoglobulin E<br />

Syndrome Patients<br />

Nina N. Brodsky, Researcher, NIH/NIAID/LCID, Bethesda,<br />

MD, Margaret R. Brown, Biologist, NIH/CC/DLM, Bethesda,<br />

MD, Steven M. Holland, Chief, LCID, NIH/NIAID/LCID,<br />

Bethesda, MD, Gulbu Uzel, Staff Clinician, NIH/NIAID/LCID,<br />

Bethesda, MD


S68 Abstracts<br />

Job’s Syndrome, or the Hyper-IgE Syndrome (HIES), is an<br />

autosomal dominant disorder characterized by elevated<br />

serum IgE levels, eosinophilia, dermatitis, skeletal abnormalities,<br />

staphylococcal skin infections, and lung cysts. An in<br />

vitro immunologic phenotype has not been established, and<br />

earlier findings have included only a small number <strong>of</strong> HIES<br />

patients. We have performed flow cytometric analysis <strong>of</strong><br />

lymphocyte subsets and eosinophils by using relevant cell<br />

surface markers in a cohort <strong>of</strong> thirty HIES patients and<br />

compared them to healthy controls using a non-parametric<br />

t test (Mann–Whitney U). Absolute lymphocyte counts did<br />

not differ between the two groups (pN0.3). We have found<br />

that patients’ eosinophils have significantly higher expression<br />

<strong>of</strong> CD23, HLA-DR, and CD69 compared to controls<br />

(p=0.0002, 0.0001 and b0.0001, respectively). We have<br />

also found that percentages <strong>of</strong> natural killer cells<br />

(pb0.0001) and CD3+ CD54+ T cells (p b0.0001) were<br />

lower in HIES. We have detected significantly smaller<br />

number <strong>of</strong> CD8 and CD4 memory T cells (CD8+CD45RO+ and<br />

CD4+CD45RO+) (pb0.0001 and pb0.04 respectively), more<br />

naive CD4 T cells (CD4+ CD45RA+) (p=0.001) and CD8+CD57+<br />

cells (p=0.0007) in patients. HIES patients had significantly<br />

more T cells expressing CD147 (a plasma membrane glycoprotein<br />

shown to induce extracellular matrix metalloproteinases<br />

[MMPs], pb0.04). Our findings <strong>of</strong> decreased number <strong>of</strong><br />

memory T cells in HIES, similar to Hyper IgM syndrome and<br />

ectodermal dysplasia with immunodeficiency (EDID), suggest a<br />

potential defect in the NFkB pathway. On the other hand,<br />

increased CD147 expression points at pathways responsible for<br />

regulation <strong>of</strong> MMPs, which may be critical for tissue<br />

remodeling and regeneration defects in HIES.<br />

doi:10.1016/j.clim.2007.03.370<br />

OR.53 Global Assessment <strong>of</strong> B Cell Alloimmunity<br />

Using Human Protein Microarrays<br />

Persis P. Wadia, Postdoctoral Fellow, Stanford University,<br />

Miklos Lab, Stanford, CA, Marc Coram, Stanford University,<br />

Stanford, CA, Marina Sirota, Stanford University, Stanford,<br />

CA, David B. Miklos, Assistant Pr<strong>of</strong>essor, Stanford<br />

University, Stanford, CA, Atul Butte, Stanford University,<br />

Stanford, CA<br />

Allogeneic hematopoietic cell transplantation (HCT) can<br />

cure hematologic malignancies through beneficial graft-vleukemia<br />

(GVL) allo-immune responses, but is limited by<br />

graft-versus-host disease (GVHD). Previous studies demonstrate<br />

that allogeneic antibodies (Ab) develop against minor<br />

histocompatibility antigens (mHA) encoded on the Y-chromosome<br />

(H-Y antigens) after sex-mismatch HCT in association<br />

with chronic GVHD and persistent disease remission. We<br />

hypothesize that novel mHA can be serologically identified as<br />

targets <strong>of</strong> allo-Ab responses that develop post-transplant.<br />

ProtoArray displays 5000 full-length human proteins with Nterminal<br />

GSTepitopes. Technical replicates <strong>of</strong> plasma measured<br />

at 1:50, 1:150, 1:500 confirmed technical feasibility with R 2<br />

N0.95. In this study, plasma collected from four AML patients<br />

1 year post-transplant, pre-transplant, and their donors were<br />

detected for antibody specificity against 5000 human proteins<br />

displayed on the ProtoArray. Pre-post fluorescence differences<br />

ranging from 0.5 to 3 logs identified 60–75 targets <strong>of</strong> allo-Ab.<br />

Over 90% <strong>of</strong> allo-Ab targets have known non-synonymous<br />

SNPs. Chromatin Assembly Factor 1, subunit B (p60)<br />

(CHAF1B), and Nucleolar and Spindle Associated Protein 1<br />

(NuSAP1) were recognized by two <strong>of</strong> the four patients after<br />

HCT. These allo-Abs were absent pre and 2 months post-HCT,<br />

developed in association with cGVHD at 11 months, and<br />

persisted through 16 months. CHAF1B and NuSAP have been<br />

recombinantly expressed and specifically detected by post-<br />

HCT patient sera by western blot validating their discovery as<br />

allo-antigens. Ongoing studies are characterizing CHAF1B and<br />

NuSAP1 Ab by ELISA in HCT patient samples, and normals. In<br />

summary, protein microarrays are innovative tools for highthroughput<br />

global assessment <strong>of</strong> B cell alloimmunity and mHA<br />

discovery.<br />

doi:10.1016/j.clim.2007.03.371<br />

OR.54 Improving Targeted Immunotherapy Through<br />

Direct Validation <strong>of</strong> Peptide–MHC Epitopes Using<br />

TCRm Antibodies<br />

Tiffany Nguyen, Senior Research Technician, Receptor<br />

Logic, Ltd, Amarillo, TX, Tito Woodburn, Research<br />

Associate, Receptor Logic, Ltd, Amarillo, TX, Francisca<br />

Neethling, Scientist, Receptor Logic, Ltd, Amarillo, TX,<br />

Jon A. Weidanz, Assistant Pr<strong>of</strong>essor, Texas Tech<br />

University Health Sciences Center, Amarillo, TX<br />

Adoptive T cell immunotherapy represents a promising<br />

approach for the treatment <strong>of</strong> cancer and chronic infections.<br />

Improved response rates using this approach might occur if<br />

the specific peptide–MHC targets on the diseased cells were<br />

known. We have developed technology to directly assess the<br />

presentation <strong>of</strong> specific peptide–MHC complexes on tumor<br />

cells using antibodies that mimic the binding specificity <strong>of</strong> T<br />

cell receptors designated as TCRmimics (TCRms). A pro<strong>of</strong>-<strong>of</strong>concept<br />

validation assay was developed to directly examine<br />

the presentation and density <strong>of</strong> MHC class I peptides derived<br />

from the processing <strong>of</strong> a model tumor antigen, hCGb, on the<br />

surface <strong>of</strong> various human tumor cell lines. In this study we<br />

used previously characterized TCRm to three peptide–HLA-<br />

A*0201 epitopes derived from hCGb and designated as TMT<br />

(60–68), VLQ (64–72), and GVL (67–75). For the assessment<br />

<strong>of</strong> antigen presentation function, hCGb positive tumor cell<br />

lines were stained for detection and quantitation <strong>of</strong> each<br />

peptide–HLA epitope. To our surprise, presentation <strong>of</strong> each<br />

peptide–epitope varied widely with no clear dominant<br />

peptide–epitope being expressed on the different tumor<br />

cell lines. A human breast carcinoma cell line expressed all<br />

three peptide–HLA-A2 epitopes with GVL peptide being<br />

dominant. In contrast, only GVL peptide–epitope was<br />

detected on an ovarian carcinoma cell line while only TMT<br />

peptide–epitope was found expressed on a colon carcinoma<br />

cell line. Taken together, these findings illustrate a heterogeneous<br />

pattern <strong>of</strong> peptide–HLA expression and signify the<br />

need for development <strong>of</strong> reagents that can directly assess<br />

specific peptide–HLA expression and lead to better outcomes<br />

with T cell immunotherapy.<br />

doi:10.1016/j.clim.2007.03.372


Abstracts<br />

OR.55 Transcription Pr<strong>of</strong>iles <strong>of</strong> Rheumatoid Arthritis<br />

Patients Reveal Genes Characterizing Different<br />

Response to Anti-TNF Therapy<br />

Franak Batliwalla, Assistant Investigator, Feinstein Institute<br />

for Medical Research, Manhasset, NY, Peter Gregersen,<br />

Investigator, Feinstein Institute for Medical Research,<br />

Manhasset, NY, Normand Allaire, Scientist, BiogenIdec, Drug<br />

Discovery, Cambridge, NY, Wentian Li, Assistant Investigator,<br />

Feinstein Institute for Medical Research, Manhasset, NY,<br />

Houman Khalili, Steve Perrin, Associate Director, BiogenIdec,<br />

Drug Discovery, Cambridge, MA, Marlena Kern, Research<br />

Associate, Feinstein Institute for Medical Research,<br />

Manhasset, NY, Aarti Damle, Research Associate, Feinstein<br />

Institute for Medical Research, Manhasset, NY, John Carulli,<br />

Principal Scientist, BiogenIdec, Drug Discovery, Cambridge,<br />

MA, Jadwiga Bienkowska, Principal Scientist, Biogenidec,<br />

Drug Discovery, Cambridge, MA<br />

The Autoimmune Biomarkers Collaborative Network<br />

(ABCoN) has enrolled a longitudinal cohort <strong>of</strong> RA patients<br />

beginning anti-TNF treatment in order to identify biomarkers<br />

influencing response to anti-TNF therapy. 116 patients<br />

beginning anti-TNF therapy (54 etaneracept, 25 adalimumab,<br />

37 infliximab) were followed for 14 weeks, with DAS28<br />

measurements and RNA collection at three time points: pretreatment,<br />

6 weeks and 14 weeks post-treatment. Using the<br />

hgu133plus2 Affymetrix chips we have completed genomewide<br />

transcript pr<strong>of</strong>iling for these 116 patients as well as 65<br />

healthy controls. Defining response as a DDAS28 N40% and<br />

non-response as DDAS28 b20%, analysis <strong>of</strong> the gene expression<br />

differences between patients and controls indicates that the<br />

overall number <strong>of</strong> differentially expressed genes is different<br />

for responders and non-responders. Furthermore, the responders<br />

are characterized by a unique list <strong>of</strong> genes differentially<br />

expressed as compared to controls at 14 weeks posttreatment.<br />

This observation suggests that anti-TNF therapy<br />

recruits specific biological pathways in responders. In order<br />

to identify the biomarkers that predict the response to anti-<br />

TNF therapy we have analyzed the gene expression pr<strong>of</strong>iles <strong>of</strong><br />

responders and non-responders using blood collected at the<br />

pre-treatment visit. Using the machine learning technique<br />

Random Forest we have identified a set <strong>of</strong> over 100 genes that<br />

are predictive <strong>of</strong> the anti-TNF response. Using a selected set<br />

<strong>of</strong> 25 candidate biomarkers we can distinguish the responders<br />

versus non-responders with 75% accuracy. We are validating<br />

the proposed biomarkers using an independent cohort <strong>of</strong><br />

patients as well as low-density RT-PCR arrays.<br />

doi:10.1016/j.clim.2007.03.373<br />

New Animal Models <strong>of</strong> Disease<br />

Saturday, June 9<br />

2:45 pm−4:45 pm<br />

OR.56 Development <strong>of</strong> Latest-generation<br />

HU-PBMC-NOD/SCID Mice to Study Human Islet<br />

Allo-reactivity<br />

Todd Pearson, Postdoctoral Fellow, University <strong>of</strong> Massachusetts<br />

Medical School, Diabetes Division, Worcester, MA, Marie King,<br />

MD/PhD Student, University <strong>of</strong> Massachusetts Medical School,<br />

Diabetes Division, Worcester, MA, Leonard Shultz, Senior Staff<br />

Scientist, The Jackson Laboratory, Bar Harbor, ME, Jean Leif,<br />

Lab Manager, University <strong>of</strong> Massachusetts Medical School,<br />

Diabetes Division, Worcester, MA, Dale Greiner, Pr<strong>of</strong>essor,<br />

University <strong>of</strong> Massachusetts Medical School, Diabetes Division,<br />

Worcester, MA, John Mordes, Pr<strong>of</strong>essor, University <strong>of</strong><br />

Massachusetts Medical School, Diabetes Division, Worcester,<br />

MA, Aldo Rossini, Pr<strong>of</strong>essor, University <strong>of</strong> Massachusetts<br />

Medical School, Diabetes Division, Worcester, MA, Mark<br />

Atkinson, Pr<strong>of</strong>essor, University <strong>of</strong> Florida College <strong>of</strong> Medicine,<br />

Department <strong>of</strong> Pathology, <strong>Immunology</strong> and Laboratory<br />

Medicine, Gainesville, FL, Clive Wasserfall, Assistant in<br />

Pathology, University <strong>of</strong> Florida College <strong>of</strong> Medicine,<br />

Department <strong>of</strong> Pathology, <strong>Immunology</strong> and Laboratory<br />

Medicine, Gainesville, FL, Massimo Trucco, Pr<strong>of</strong>essor,<br />

University <strong>of</strong> Pittsburgh School <strong>of</strong> Medicine, Pediatrics,<br />

Pittsburgh, PA, Kevan Herold, Pr<strong>of</strong>essor, Yale University School<br />

<strong>of</strong> Medicine, Department <strong>of</strong> Internal Medicine, New Haven, CT,<br />

Rita Botti, Assistant Pr<strong>of</strong>essor, University <strong>of</strong> Pittsburgh,<br />

Pediatrics, Pittsburgh, PA<br />

Small animal models have been used to study a number <strong>of</strong><br />

human diseases, including autoimmune diseases such as type<br />

1 diabetes (T1D). Unfortunately, translating therapies from<br />

animal models to human patients has been hindered by<br />

differences in rodent and human immune systems. “Humanized”<br />

mouse models hold great promise in the development<br />

<strong>of</strong> efficacious therapies to treat a wide array <strong>of</strong> human<br />

immune-mediated conditions, without putting human subjects<br />

at risk during protocol development. However, developing<br />

a system that faithfully recapitulates human immunity<br />

in a murine host has proven difficult. Recently, the generation<br />

<strong>of</strong> a new stock <strong>of</strong> immunodeficient hosts, the NOD.Cg-<br />

Prkdcscid Il2rgtm1Wjl/Sz (NOD-scid Il2rgnull) strain, has<br />

overcome many <strong>of</strong> the previous limitations <strong>of</strong> humanized<br />

mice. We have characterized the engraftment <strong>of</strong> human<br />

PBMC into NOD-scid Il2rgnull hosts and document that this<br />

stock supports higher human cell engraftment at lower cell<br />

input levels. Importantly, we further demonstrate that<br />

human PBMC-engrafted NOD-scid Il2rgnull mice allow for<br />

allogeneic rejection <strong>of</strong> transplanted HLA-mismatched human<br />

islets, even when the islets are allowed to heal-in prior to<br />

PBMC engraftment. Collectively, these data suggest that<br />

humanized NOD-scid Il2rgnull mice may be superior to<br />

previous immunodeficient recipients for generation <strong>of</strong><br />

humanized mice for studies <strong>of</strong> in vivo human immune<br />

function.<br />

doi:10.1016/j.clim.2007.03.374<br />

S69<br />

OR.57 TSLP-dependent Induction <strong>of</strong> Airway<br />

Inflammatory Disease is Antigen-driven in an Acute<br />

Model <strong>of</strong> Allergic Airway Inflammation<br />

Mark Headley, Graduate Student, University <strong>of</strong> Washington<br />

<strong>Immunology</strong>, Seattle, WA, Baohua Zhou, Post Doctoral<br />

Fellow, Benaroya Research Institute, Ziegler Lab, Seattle,<br />

WA, Steve Ziegler, Director <strong>of</strong> <strong>Immunology</strong>, Benaroya<br />

Research Institute, Ziegler Lab, Seattle, WA


S70 Abstracts<br />

The cytokine, Thymic Stromal Lymphopoietin (TSLP), has<br />

been shown to play an important role in the mediation <strong>of</strong> Th2biased<br />

inflammatory diseases in particular the atopic diseases:<br />

asthma and atopic dermatitis. Mice expressing a TSLP transgene<br />

driven by the lung-specific Surfactant Protein C promoter (SPC-<br />

TSLP) develop a severe and chronic asthma-like disease<br />

characterized by airway hyperresponsiveness, leukocytic infiltration<br />

<strong>of</strong> the lung, eosinophilia, goblet cell hyperplasia, subepithelial<br />

fibrosis, and elevated serum IgE. Previous studies have<br />

indicated that TSLP alone is sufficient to induce all <strong>of</strong> the<br />

hallmark features <strong>of</strong> asthma in mice. Here we show that an<br />

acute airway inflammatory disease, similar both to that seen in<br />

asthmatics as well as SPC-TSLP mice, can be induced in BALB/c<br />

mice through direct intranasal administration <strong>of</strong> TSLP in<br />

conjunction with a foreign antigen, sans adjuvant. This has<br />

been observed with multiple antigens, including ovalbumin,<br />

chicken gamma globulin, and bovine serum albumin. In contrast,<br />

mice treated with either TSLP alone, the antigens alone, or TSLP<br />

in conjunction with the self-protein, mouse serum albumin, fail<br />

to develop disease. These data demonstrate for the first time<br />

that in an acute model <strong>of</strong> TSLP-mediated airway inflammation,<br />

both thymic stromal lymphopoietin and antigen are required in<br />

order to develop full disease symptoms.<br />

doi:10.1016/j.clim.2007.03.375<br />

OR.58 Disrupting Mer Receptor Tyrosine Kinase<br />

Expression Prevents Autoimmune Chronic<br />

Graft-versus-host Disease<br />

Wenha Shao, Postdoctoral Research Associate,<br />

Rheumatology Division, Department <strong>of</strong> Medicine,<br />

Philadelphia, PA, Robert A. Eisenberg, Pr<strong>of</strong>essor <strong>of</strong><br />

Medicine, Rheumatology Division, Department <strong>of</strong> Medicine,<br />

University <strong>of</strong> Pennsylvania, Philadelphia, PA, Philip L.<br />

Cohen, Pr<strong>of</strong>essor <strong>of</strong> Medicine, Rheumatology Division,<br />

Department <strong>of</strong> Medicine, University <strong>of</strong> Pennsylvania,<br />

Philadelphia, PA<br />

The Mer receptor tyrosine kinase mediates apoptotic cell<br />

phagocytosis and modulates macrophage cytokine production.<br />

Mer knockout mice (Mer-KO) have defective clearance <strong>of</strong><br />

apoptotic debris and develop SLE-like autoimmune disease.<br />

Because <strong>of</strong> their spontaneous autoimmune propensity, we<br />

wondered if Mer-KO mice (backcrossed 10 generations onto<br />

C57BL/6) might be particularly susceptible to the SLE-like<br />

autoimmunity induced by chronic GVH. Using the wellestablished<br />

cGVH model (bm12′B6 mice), we were surprised<br />

to observe that the Mer-KO mice were protected from the<br />

development <strong>of</strong> GVHD. Mer-KO recipients <strong>of</strong> bm12 spleen cells<br />

failed to develop detectable anti-dsDNA and anti-chromatin<br />

autoantibodies, while the B6 hosts produced significant<br />

amounts <strong>of</strong> these antibodies that peaked at week 2. Slightly<br />

increased levels <strong>of</strong> rheumatoid factor were found in Mer-KO<br />

mice, though much lower than B6 control hosts. Mer-KO mice<br />

did not develop splenomegaly. The lack <strong>of</strong> autoantibody<br />

formation was not due to absence <strong>of</strong> alloreactivity because<br />

spleen cells from bm12 mice proliferated equally in response<br />

to irradiated WT and Mer-KO irradiated spleen cells. These<br />

findings indicate a hitherto unrecognized requirement for Mer<br />

in the genesis <strong>of</strong> alloreactivity-induced autoimmunity. Future<br />

experiments will determine whether this reflects the role <strong>of</strong><br />

this kinase in recognition and phagocytosis <strong>of</strong> apoptotic cells,<br />

its function in regulating cytokine production, or perhaps a<br />

new function in the immune response.<br />

doi:10.1016/j.clim.2007.03.376<br />

OR.59 In Vivo and In Silico Modeling the Dynamics <strong>of</strong><br />

Autoimmune Diabetes<br />

Qizhi Tang, Assistant Adjunct Pr<strong>of</strong>essor, University <strong>of</strong><br />

California, San Francisco Diabetes Center, San Francisco,<br />

CA, Yanan Zheng, Engineer, Biological Systems, Entelos, In<br />

Silico Research and Development, Foster City, CA, Jason<br />

Adams, MD, Stanford University, Stanford, CA, Huub<br />

Kreuwel, Scientist, Immunologic Diseases, Entelos, In Silico<br />

Research and Development, Foster City, CA, Kapil Gadkar,<br />

Biosystems Engineer, Entelos, In Silico Research and<br />

Development, Foster City, CA, Cristina Penaranda,<br />

Graduate Student, University <strong>of</strong> California, San Francisco<br />

Diabetes Center, San Francisco, CA, Lisl Shoda, Senior<br />

Scientist, Entelos, In Silico Research and Development,<br />

Foster City, CA, Chan C. Whiting, Scientist, Immunologic<br />

Diseases, Entelos, In Silico Research and Development,<br />

Foster City, CA, Daniel Young, Dynamics Engineer, Entelos,<br />

In Silico Research and Development, Foster City, CA, Jeffrey<br />

Bluestone, Pr<strong>of</strong>essor, University <strong>of</strong> California, San Francisco<br />

Diabetes Center, San Francisco, CA<br />

Progression <strong>of</strong> autoimmune diabetes results as a consequence<br />

<strong>of</strong> an imbalance <strong>of</strong> Teff and suppressive regulatory T<br />

cells (Tregs). A novel mathematical model <strong>of</strong> type 1 diabetes<br />

(T1D) in the non-obese diabetes (NOD) mouse (T1D Physio-<br />

Lab® platform) was developed that encompasses the<br />

dynamics <strong>of</strong> key immune components in the pancreatic<br />

lymph nodes (PLNs) and islets, and beta cell physiology.<br />

Research using this platform predicted that autoimmunity<br />

was controlled in the PLN, as shown by an increase in Tregs<br />

over the course <strong>of</strong> disease. This unexpected behavior was<br />

confirmed experimentally by functional and histological<br />

data. Additionally, laboratory analysis <strong>of</strong> individual islets<br />

demonstrated an inverse relationship between Teff and Treg<br />

infiltration <strong>of</strong> the tissue. Specifically, as Teff infiltration<br />

increases, the Treg fraction decreases. However, differential<br />

proliferation <strong>of</strong> Teffs and Tregs in vivo was found to be<br />

insufficient to explain this relationship although evidence <strong>of</strong><br />

reduced expression <strong>of</strong> CD25 on the resident pancreatic Tregs<br />

could alter Treg survival accounting for the observed effects.<br />

Based on these in vivo results, the relative turnover and<br />

recruitment <strong>of</strong> Tregs and Teffs were tested in the T1D<br />

platform. Results <strong>of</strong> these in silico investigations demonstrated<br />

that the ultimate determination <strong>of</strong> disease activity<br />

depends on dynamic changes in intra-islet Treg turnover and<br />

changes in the Teff/Treg balance. These combined experimental<br />

and mathematical modeling results emphasize the<br />

central importance <strong>of</strong> both PLN and intra-islet Treg function<br />

in regulating disease pathogenesis and the power <strong>of</strong><br />

integrating in vivo and in silico modeling to dissect mechanisms<br />

leading to disease pathogenesis.<br />

doi:10.1016/j.clim.2007.03.377


Abstracts<br />

OR.60 Antigen Identification in a Novel Model <strong>of</strong><br />

Spontaneous Autoimmune Ovarian Disease<br />

Mickie Cheng, <strong>Clinical</strong> Fellow in Endocrinology, University <strong>of</strong><br />

California, San Francisco Diabetes Center, San Francisco,<br />

CA, Kellsey Johannes, Staff Research Associate, University<br />

<strong>of</strong> California, San Francisco Diabetes Center, San Francisco,<br />

CA, Wen Lu, Staff Research Associate, University <strong>of</strong><br />

California, San Francisco Diabetes Center, San Francisco,<br />

CA, Mark Anderson, Assistant Pr<strong>of</strong>essor <strong>of</strong> Medicine in<br />

Residence, University <strong>of</strong> California, San Francisco Diabetes<br />

Center, San Francisco, CA<br />

Autoimmune ovarian disease (AOD) is a poorly understood<br />

clinical entity wherein immune-mediated destruction <strong>of</strong><br />

oocytes leads to premature ovarian failure and infertility.<br />

We have generated a novel spontaneous mouse model to<br />

study AOD using mice deficient for the AutoImmune<br />

REgulator (aire), a gene first identified in the human<br />

Autoimmune Polyglandular Syndrome (APS) type 1. Like<br />

patients with APS 1, aire-deficient mice develop disease <strong>of</strong><br />

multiple organs, including the ovary, due to a breakdown in<br />

central tolerance. In aire KO mice, thymic epithelial cells are<br />

defective in their ability to tolerize developing Tcells to selfantigens,<br />

thus allowing autoreactive cells to escape into the<br />

periphery and induce self tissue destruction. Autoimmunity<br />

is manifested by the development <strong>of</strong> antigen-specific<br />

autoantibodies, organ infiltration, and transfer <strong>of</strong> disease<br />

by lymphocytes from affected aire KO mice. aire KO females<br />

exhibit 100% incidence <strong>of</strong> histologic oophoritis as well as<br />

circulating autoantibodies to ovarian tissue in a strain<br />

specific manner, presenting a valuable spontaneous disease<br />

model to identify ovarian antigens. Initial characterization <strong>of</strong><br />

AOD in these mice implicates a central role for T cell<br />

mediated disease with predominantly Th1 CD4+ T cell<br />

infiltrate. Furthermore, autoantibodies from aire KO females<br />

target the oocyte cytoplasm and follicular layer and<br />

recognize a 70 kDa autoantigen in ovarian lysates. Identification<br />

<strong>of</strong> the disease-inciting antigen could enable improved<br />

testing for the diagnosis <strong>of</strong> autoimmune ovarian disease and<br />

may provide a potential therapeutic target for treatment<br />

and prevention <strong>of</strong> a significant cause <strong>of</strong> human ovarian<br />

failure and infertility.<br />

doi:10.1016/j.clim.2007.03.378<br />

OR.61 Delineating the Roles <strong>of</strong> CD4+ and CD8+ T Cell<br />

Subsets in the Pathogenesis <strong>of</strong> Spontaneous<br />

Autoimmune Diabetes Using HLA-DQ8 Transgenic<br />

Mice<br />

Govindarajan Rajagopalan, Department <strong>of</strong> <strong>Immunology</strong>,<br />

Mayo Clinic, Rochester, MN, Ashutosh Mangalam,<br />

Department <strong>of</strong> <strong>Immunology</strong>, Mayo Clinic, Rochester, MN,<br />

Yogish Kudva, Department <strong>of</strong> Endocrinology, Mayo Clinic,<br />

Rochester, MN, Chella David, Department <strong>of</strong> <strong>Immunology</strong>,<br />

Mayo Clinic, Rochester, MN<br />

Certain MHC class II molecules show a strong association<br />

with T1D implying an important role for CD4+ T cells. MHC<br />

class I-restricted CD8+ T cells also play a crucial effector<br />

role. The roles <strong>of</strong> CD4+ and CD8+ T cell subsets in the<br />

pathogenesis <strong>of</strong> spontaneous autoimmune diabetes were<br />

studied using HLA-DQ8 transgenic mouse models. CD8sufficient<br />

or CD8-deficient Abo.DQ8 transgenic mice expressing<br />

either the proinflammatory cytokine, tumor necrosis<br />

factor (TNF)-α or the costimulatory molecule, B7.1 or both<br />

in the pancreatic beta cells were used in this study. In the<br />

RIP-TNF model, the incidence <strong>of</strong> diabetes was class II or<br />

CD4+ T cell independent as 75% <strong>of</strong> mice lacking HLA-DQ8<br />

(and there by CD4+ T cells) developed T1D. However, it was<br />

absolutely class I or CD8+ T cell dependent as none <strong>of</strong> the<br />

CD8-deficient mice developed T1D. In the RIP-B7 model, the<br />

incidence <strong>of</strong> diabetes was class II or CD4+ T cell dependent,<br />

as none <strong>of</strong> the mice lacking HLA-DQ8 (and there by CD4+ T<br />

cells) developed T1D when compared to 35% incidence<br />

when HLA-DQ8 (and there by CD4+ T cells) is expressed.<br />

Nonetheless, in RIP-B7 model, CD8-deficient HLA-DQ8<br />

transgenic mice were still susceptible to T1D albeit at a<br />

lower rate (Incidence ¡V 17%). In the RIP-B7.RIP-TNF double<br />

transgenic mouse model, 100% <strong>of</strong> the animals developed<br />

diabetes irrespective <strong>of</strong> whether they lacked CD4+ or CD8+<br />

T cells. Thus, CD4+ and CD8+ T cells contribute differentially<br />

to the pathogenesis <strong>of</strong> T1D according to the local immunological<br />

insult.<br />

doi:10.1016/j.clim.2007.03.379<br />

S71<br />

OR.62 The CYP450 Mouse Model for Autoimmune<br />

Hepatitis: Breaking <strong>of</strong> Self-Tolerance in Transgenic<br />

CYP2D6 and Wildtype FVB-Mice by Viral Infection<br />

Urs Christen, Assistant Pr<strong>of</strong>essor, Pharmazentrum, JWG<br />

University Frankfurt, Frankfurt am Main, Germany,<br />

Martin Holdener, PhD Student, Pharmazentrum, JWG<br />

University Frankfurt, Frankfurt am Main, Germany,<br />

Edith Hintermann, Senior Postdoctoral Fellow,<br />

Pharmazentrum, JWG University Frankfurt, Frankfurt<br />

am Main, Germany, Matthias von Herrath, Pr<strong>of</strong>essor, La<br />

Jolla Institute for Allergy and <strong>Immunology</strong>, La Jolla,<br />

CA, Michael Manns, Pr<strong>of</strong>essor, Hannover Medical School,<br />

Hannover, Germany<br />

The etiology <strong>of</strong> autoimmune hepatitis (AIH) is poorly<br />

understood although the major autoantigen, cytochrome<br />

P450 2D6 (CYP2D6), has been identified. We generated an<br />

animal model for human AIH using the natural autoantigen<br />

CYP2D6. We infected transgenic mice expressing human<br />

CYP2D6 in the liver with an Adenovirus-CYP2D6 vector (Ad-<br />

2D6) to break self-tolerance. Upon infection with Ad-2D6<br />

not only transgenic CYP2D6 mice but also wildtype FVB<br />

mice showed persistent features <strong>of</strong> severe liver damage<br />

associated with AIH (hepatic fibrosis, fused liver lobules,<br />

cellular infiltrations, elevated serum ALT levels, confluent<br />

necrosis). Ad-2D6-infected mice (CYP2D6 and FVB) generated<br />

high titers <strong>of</strong> anti-CYP2D6 antibodies. Epitope mapping<br />

revealed that anti-CYP2D6 antibodies predominantly<br />

recognized the same immunodominant linear epitope<br />

recognized by sera <strong>of</strong> AIH patients (AQPPRD aa 265–270).<br />

In contrast, mice infected with a control Adenovirus<br />

expressing green fluorescence protein did neither develop


S72 Abstracts<br />

liver damage nor generated anti-CYP2D6 antibodies. The<br />

severity <strong>of</strong> liver damage as well as antibody formation was<br />

enhanced in FVB mice compared to transgenic CYP2D6<br />

mice indicating a stronger tolerance to human CYP2D6 in<br />

CYP2D6 mice. In FVB mice, due to the homology <strong>of</strong> the<br />

mouse isoenzymes <strong>of</strong> the CYP superfamily to human<br />

CYP2D6, autoimmune liver damage by Ad-2D6 infection<br />

was possibly induced via true molecular mimicry.<br />

doi:10.1016/j.clim.2007.03.380<br />

Immunoregulatory Mechanisms<br />

Saturday, June 9<br />

2:45 pm−4:45 pm<br />

OR.63 Modification <strong>of</strong> Host Cellular DNA by<br />

Parvovirus B19 Nonstructural Protein 1 (NS1):<br />

Implications for Induction <strong>of</strong> Autoimmunity by DNA<br />

Viruses<br />

Leona Gilbert, University <strong>of</strong> Jyvaskala, University <strong>of</strong><br />

Jyvaskala, Jyvaskala, Finland, Brian D. Poole, Oklahoma<br />

Medical Research Foundation, Oklahoma Medical Research<br />

Foundation, Oklahoma City, OK, Violetta Kivovich,<br />

University <strong>of</strong> Jyvaskala, University <strong>of</strong> Jyvaskala, Jyvaskala,<br />

Finland, Stanley Naides, Medical Director, <strong>Immunology</strong>,<br />

Quest Diagnostics Nichols Institute, San Juan Capistra, CA,<br />

Jing Zhou, Pennsylvania State University College <strong>of</strong><br />

Medicine/Milton S. Hershey Medical Center, Pennsylvania<br />

State University College <strong>of</strong> Medicine/Milton S. Hershey<br />

Medical Center, Hershey, PA<br />

Autoantibodies cross-reactive to EBV proteins precede<br />

SLE onset in a subset <strong>of</strong> patients. However, the event that<br />

links viral infection to autoimmunity, the “original sin,”<br />

remains unknown. We studied the role <strong>of</strong> parvovirus B19<br />

infection in host DNA modification. B19 infection in nonerythroid<br />

cells (e.g., Hep G2 and primary hepatocytes) is<br />

restricted to expression <strong>of</strong> NS1. NS1 is a superfamily 3 (SF3)<br />

helicase, typical <strong>of</strong> small DNA viruses, that plays a key role<br />

in viral genome replication. In hepatocytes, B19 induces<br />

apoptosis through mitochondrial stress pathways. We<br />

examined NS1 expression in Hep G2 cells using an enhanced<br />

green fluorescent fusion protein (eGFP/NS1). Expressed<br />

eGFP/NS1 localized to the nucleus. The majority <strong>of</strong> cells<br />

expressing eGFP/NS1 underwent apoptosis with PARP<br />

activation. Inhibitors <strong>of</strong> PARP or ATR abrogated apoptosis.<br />

eGFP/NS1 colocalized with RPA and PCNA. Western blot<br />

analysis and autoradiographs demonstrated that NS1<br />

formed dimers under reducing conditions and covalently<br />

bound to cellular DNA. NS1 was polyADP ribosylated.<br />

Apoptotic subcellular particles from cells expressing<br />

eGFP/NS1 fluoresced. Our data demonstrated that B19<br />

NS1 forms bulky adducts with cellular DNA and nicks<br />

cellular DNA, initiating pathways that lead to mitochondrial<br />

stress and apoptosis. NS1 with covalently linked self-DNA is<br />

extruded into the extracellular milieu in apoptotic bodies.<br />

We propose that modification <strong>of</strong> cellular DNA by viral SF3<br />

helicase breaks tolerance to self DNA when presented to<br />

the immune system during apoptosis. These findings suggest<br />

a general model in which modification <strong>of</strong> host DNA by viral<br />

SF3 helicase induces or accelerates autoimmunity, the<br />

“original sin.”<br />

doi:10.1016/j.clim.2007.03.381<br />

OR.64 Long-term Deposition <strong>of</strong> Inhaled Antigen in<br />

Lung Resident CD11b−CD11c+ Cells<br />

Kate E. Matthews, Student, Malaghan Institute, Wellington<br />

South, New Zealand, Adela Karabeg, Student, Medical<br />

University <strong>of</strong> Vienna, Department Dermatology, Vienna,<br />

Austria, Gerhard Dekan, Pr<strong>of</strong>essor, Medical University <strong>of</strong><br />

Vienna, <strong>Clinical</strong> Pathology, Vienna, Austria, Franca<br />

Roncheses, Principal Investigator, Malaghan Institute,<br />

Wellington South, New Zealand, Michelle Epstein,<br />

Laboratory Leader, Medical University <strong>of</strong> Vienna,<br />

Department <strong>of</strong> Dermatology, Vienna, Austria<br />

We report the characterization <strong>of</strong> a population <strong>of</strong> lung<br />

resident CD11b−CD11c+ cells that are able to take up inhaled<br />

antigen and retain it for extended periods <strong>of</strong> time.<br />

Ovalbumin conjugated to fluorescein isothiocyanate administered<br />

intranasally to mice was taken up by two main<br />

populations <strong>of</strong> cells in the lung, a migratory CD11c+CD11b+<br />

population consisting <strong>of</strong> DC, which rapidly transported<br />

antigen to the draining lymph node (LN), and a resident<br />

CD11b−CD11c+ population that retained engulfed antigen<br />

without apparently degrading it for up to 8 weeks after<br />

administration. The FITC+CD11b−CD11c+ cells did not<br />

migrate to draining LN and did not upregulate expression <strong>of</strong><br />

costimulatory molecules in response to LPS treatment.<br />

FITC+CD11b−CD11c+ cells found in the airway were large<br />

and were consistent with macrophages. Additionally, a<br />

population <strong>of</strong> FITC+CD11b−CD11c+ seen in lung sections<br />

were situated along alveolar walls with a distribution also<br />

consistent with macrophages. Although FITC+CD11b−<br />

CD11c+ cells expressed molecules associated with APC<br />

function, they did not induce proliferation <strong>of</strong> antigenspecific<br />

CD4+ T cells in vitro or acute cytokine production<br />

by activated CD4+ Tcells in vivo. We propose that these longlived,<br />

lung resident cells might provide a depot <strong>of</strong> antigen for<br />

the indirect reactivation or retention <strong>of</strong> antigen-specific T<br />

cells in the lung.<br />

doi:10.1016/j.clim.2007.03.382<br />

OR.65 Proteinase-activated Receptor-2 (PAR-2)<br />

Activation Promotes Allergic Sensitization and<br />

Prevents the Development <strong>of</strong> Immune Tolerance to<br />

Inhaled Antigens Through a TNF Mediated Pathway<br />

Cory Ebeling, PhD candidate, University <strong>of</strong> Alberta,<br />

Department <strong>of</strong> Medicine, Edmonton, AB, Canada, Tong Lam,<br />

Medical Student, University <strong>of</strong> Alberta, Edmonton, AB,<br />

Canada, John Gordon, Associate Pr<strong>of</strong>essor, University <strong>of</strong><br />

Saskatchewan, Department <strong>of</strong> Veterinary Microbiology,<br />

Saskatoon, SK, Canada, Harissios Vliag<strong>of</strong>tis, Associate<br />

Pr<strong>of</strong>essor, University <strong>of</strong> Alberta, Department <strong>of</strong> Medicine,<br />

Edmonton, AB, Canada, Morley Hollenberg, Associate


Abstracts<br />

Pr<strong>of</strong>essor, University <strong>of</strong> Calgary, Department <strong>of</strong><br />

Pharmacology and Therapeutics, Calgary, AB, Canada<br />

The reason why particular inhaled antigens induce<br />

allergic sensitization and prevent the development <strong>of</strong><br />

immune tolerance is unclear. Intrinsic characteristics <strong>of</strong><br />

these antigens must be <strong>of</strong> importance. A common<br />

characteristic <strong>of</strong> many potent allergens is that they either<br />

possess serine proteinase activity or are inhaled in particles<br />

rich in serine proteinases. Many allergens, such as house<br />

dust mite, have the potential to activate the Proteinaseactivated<br />

Receptor-2 (PAR-2). We hypothesized that PAR-2<br />

activation by such allergens is crucial to facilitate our<br />

allergic sensitization to them. To test our hypothesis we<br />

used a Balb/c murine system with mucosal exposure to<br />

OVA, as an antigen, with a PAR-2 activating peptide (PAR-<br />

2AP) to mimic the potential <strong>of</strong> a proteolytic allergen, or<br />

other inhaled proteinases, to activate PAR-2. Upon allergen<br />

re-exposure mice initially administered OVA with the PAR-<br />

2AP developed airway inflammation, airway hyperresponsiveness<br />

(AHR), produced OVA-specific IgE as well as OVAspecific<br />

T cells with a Th2 cytokine pr<strong>of</strong>ile. Conversely,<br />

mice given OVA alone or with a control peptide (PAR-2CP)<br />

developed tolerance. These immune tolerant mice did not<br />

develop airway inflammation, AHR, or OVA-specific IgE, but<br />

developed OVA-specific T cells that secreted high levels <strong>of</strong><br />

IL-10 indicative <strong>of</strong> Treg cell function. Finally, we showed<br />

that this PAR-2-mediated allergic sensitization was TNF<br />

dependent. Thus, PAR-2 activation in the airways could be<br />

a critical factor in the development <strong>of</strong> allergic sensitization<br />

following mucosal exposure to allergens with serine<br />

proteinase activity. Interfering with this pathway may<br />

prove to be useful for the prevention or treatment <strong>of</strong><br />

asthma.<br />

doi:10.1016/j.clim.2007.03.383<br />

OR.66 Downregulation <strong>of</strong> Innate B Cells, B-1a and<br />

MZB, by iNKT Cells<br />

Xiangshu Wen, Postdoctoral Fellow, University <strong>of</strong> California,<br />

Los Angeles, Medicine, Los Angeles, CA, Jun-Qi Yang,<br />

Assistant Research Pr<strong>of</strong>essor, UC, Medicine, Cincinnati, OH,<br />

Ram Raj Singh, Pr<strong>of</strong>essor, University <strong>of</strong> California, Los<br />

Angeles, Medicine, Los Angeles, CA<br />

B cells are essential for the development <strong>of</strong> autoimmune<br />

diseases such as lupus. Although all mature B cell subsets<br />

can produce autoantibodies, several lines <strong>of</strong> evidence<br />

implicate innate B cells, namely B-1a and marginal zone B<br />

(MZB) cells, in the development <strong>of</strong> such diseases. Here, we<br />

investigated the role <strong>of</strong> interaction between innate B cells<br />

that express high levels <strong>of</strong> CD1d and CD1d-reactive iNKT<br />

cells in the regulation <strong>of</strong> autoantibodies. We found that<br />

whereas activated iNKT cells increase activation markers<br />

CD69 and CD86 on all B cell subsets, they selectively reduce<br />

the frequency <strong>of</strong> B-1a and MZB cells. In resonance with such<br />

regulatory role, the proportions <strong>of</strong> MZB cells are increased<br />

in iNKT cell deficient (Ja18−/−) mice, whereas MZB cells<br />

are reduced in iNKT cell transgenic (Va14Tg) mice.<br />

Furthermore, whereas iNKT cells increase expression <strong>of</strong><br />

activation markers and production <strong>of</strong> normal Ig via<br />

cytokines secreted by iNKT cells in transwell cultures,<br />

they selectively suppress the production <strong>of</strong> IgG anti-DNA<br />

antibody and rheumatoid factor and IL-10 by B cells in a<br />

contact-dependent manner. Such regulation <strong>of</strong> B cells by<br />

iNKT cells must be important for in vivo regulation <strong>of</strong><br />

autoantibodies and lupus disease, as the in vivo transfer <strong>of</strong><br />

iNKT cells in B cell-reconstituted SCID mice or in Ja18−/−<br />

mice suppresses autoantibody production. Treatment with<br />

an iNKT cell ligand also delays the onset <strong>of</strong> lupus in BWF1<br />

mice. Thus, iNKT cells suppress autoreactive B cells and can<br />

prevent systemic autoimmunity. They do so via regulating<br />

innate B cells.<br />

doi:10.1016/j.clim.2007.03.384<br />

OR.67 CD40 Is<strong>of</strong>orm Differences and Microdomain<br />

Localization Explain Preferential Survival <strong>of</strong><br />

CD4+CD40+ T Cells in Autoimmunity<br />

Gisela M. Vaitaitis, Pr<strong>of</strong>essional Research Assistant,<br />

University <strong>of</strong> Colorado Health Sciences Center, Webb-<br />

Waring, Denver, CO, David H. Wagner, Assistant Pr<strong>of</strong>essor,<br />

University <strong>of</strong> Colorado Health Sciences Center, Webb-<br />

Waring, Denver, CO<br />

CD40 plays a critical role in autoimmunity. CD40expressing<br />

CD4 T cells (TCD40) are expanded in autoimmunity<br />

and are necessary and sufficient in transferring disease<br />

in mouse type I diabetes. CD40 on TCD40 can have<br />

costimulatory effect and induces Nf-kB and recombinase<br />

protein expression with subsequent alteration <strong>of</strong> TCR. CD40<br />

exists as five is<strong>of</strong>orms in mice with little known-about<br />

differences in is<strong>of</strong>orm expression between cell types,<br />

between autoimmune and non-autoimmune conditions and<br />

differences in outcome <strong>of</strong> CD40 is<strong>of</strong>orm signaling. Here we<br />

show that the major is<strong>of</strong>orm expressed in TCD40 is is<strong>of</strong>orm-<br />

V with is<strong>of</strong>orm-I expressed at lower levels. This differs<br />

distinctly from antigen presenting cells which predominantly<br />

express is<strong>of</strong>orm-I. In autoimmune NOD the ratio <strong>of</strong><br />

TCD40 is<strong>of</strong>orm-V to -I is exaggerated and overall expression<br />

is greater compared to non-autoimmune BALB/c. It was<br />

shown in lymphocyte cell lines that CD40 and TRAF2 are not<br />

associated with detergent-insoluble microdomains (rafts)<br />

until CD40 is engaged. However, untreated NOD TCD40 have<br />

the CD40 is<strong>of</strong>orms and TRAF2 associated with this fraction<br />

while BALB/c TCD40 do not. A functional outcome <strong>of</strong> CD40<br />

signals to NOD TCD40 is increased survival through induction<br />

<strong>of</strong> survival proteins Bcl-XL and cFLIPp43. In addition, CD40<br />

signals to NOD TCD40 alter the outcome <strong>of</strong> Fas engagement<br />

such that Fas instead <strong>of</strong> inducing apoptosis induces<br />

increased survival beyond that <strong>of</strong> CD40 alone. Therefore<br />

TCD40 from NOD may be poised for survival even when<br />

encountering death promoting conditions explaining how<br />

this population gains the upper hand in autoimmune<br />

conditions to thwart T cell homeostasis.<br />

doi:10.1016/j.clim.2007.03.385<br />

S73


S74 Abstracts<br />

OR.68 An Immunoregulatory Network <strong>of</strong> Natural<br />

Killer T Cells and CD4+CD25+Foxp3+ T Cells<br />

Protects Against Graft-versus-host Disease<br />

Asha Pillai, Postdoctoraltoral Research Fellow, Stanford,<br />

Stanford, CA, Suparna Dutt, Research Associate,<br />

Department <strong>of</strong> Medicine, Stanford, CA, Tracy George,<br />

Assistant Pr<strong>of</strong>essor, Department <strong>of</strong> Pathology, Stanford, CA,<br />

Samuel Strober, Pr<strong>of</strong>essor <strong>of</strong> Medicine, Stanford University,<br />

Department <strong>of</strong> Medicine, Stanford, CA<br />

To investigate acute graft-versus-host disease (GVHD)<br />

protection in the murine regimen <strong>of</strong> total lymphoid irradiation<br />

and anti-thymocyte serum (TLI/ATS), we performed transplants<br />

<strong>of</strong> 50×10 6 bone marrow cells and 60×10 6 splenocytes (BMT)<br />

from wild-type (WT) C57BL/6 donors into WT BALB/c hosts<br />

treated with TLI/ATS or control BALB/c hosts given 800 cGy total<br />

body irradiation (TBI) and ATS. TLI/ATS-conditioned WT hosts<br />

given BMT from WT donors all survived 100 days without GVHD.<br />

BMT from WT donors into TBI/ATS treated WT hosts resulted in<br />

lethal GVHD. TLI/ATS-treated natural killer (NK) Tcell deficient<br />

Jα18−/− hosts also died <strong>of</strong> GVHD. TLI/ATS conditioned WT hosts<br />

receiving WT BMT demonstrated a significant (pb0.01) increase<br />

in the absolute number <strong>of</strong> donor CD4+CD25+Foxp3+ cells (CD4+<br />

Tregs) in the spleen at day 6 after BMT relative to TBI/ATS<br />

controls or TLI/ATS-treated Jα18−/− hosts. All BMT groups<br />

developing GVHD demonstrated dramatic donor CD8+ T cell<br />

accumulation in the mesenteric lymph nodes (MLN) and colon at<br />

day 6 after BMT. Adoptive transfer <strong>of</strong> sorted BALB/c NK T cells<br />

protected TLI/ATS-treated Jα18−/− hosts from donor CD8+ T<br />

cell accumulation, with a significant (pb0.001) increase in the<br />

absolute number <strong>of</strong> donor CD4+ Tregs in the host spleen at day 6.<br />

Depletion <strong>of</strong> CD25+ T cells before BMT into TLI/ATS-treated WT<br />

hosts resulted in loss <strong>of</strong> donor CD4+ Tregs in the host spleen and<br />

lethal GVHD. The present studies show that both host NK Tcells<br />

and donor CD4+CD25+Foxp3+ Tregs are required to protect<br />

against GVHD after TLI/ATS conditioning and allogeneic BMT.<br />

doi:10.1016/j.clim.2007.03.386<br />

OR.69 Peroxisome Poliferator-activated Receptor-α<br />

(PPARα) Expression in T Cells Mediates Gender<br />

Differences in Development <strong>of</strong> T Cell-mediated<br />

Autoimmunity<br />

Shannon Dunn, Postdoctoral Fellow, Stanford University,<br />

Department <strong>of</strong> Neurology, Stanford, CA, Shalina Ousman,<br />

Postdoctoral Fellow, Stanford University, Department <strong>of</strong><br />

Neurology, Stanford, CA, Raymond Sobel, Pr<strong>of</strong>essor,<br />

Department <strong>of</strong> Pathology, Stanford, CA, Sergio Baranzini,<br />

Research Associate, University <strong>of</strong> California, San Francisco,<br />

Department <strong>of</strong> Neurology, San Francisco, CA, Luis Zuniga,<br />

Student, Stanford University, Stanford, CA, Sawsan Youssef,<br />

Postdoctoral Fellow, Stanford University, Department <strong>of</strong><br />

Neurology, Stanford, CA, Jorge Oksenberg, Associate<br />

Pr<strong>of</strong>essor, University <strong>of</strong> California, San Francisco,<br />

Department <strong>of</strong> Neurology, San Francisco, CA, Lawrence<br />

Steinman, Pr<strong>of</strong>essor, Departmental Chair in <strong>Immunology</strong>,<br />

Stanford University, Department <strong>of</strong> Neurology,<br />

Stanford, CA<br />

PPAR is a nuclear receptor that mediates gender<br />

differences in lipid metabolism. PPAR also functions to<br />

control inflammatory responses by repressing the activity <strong>of</strong><br />

NFkappaB and c-jun in immune cells. Since PPAR is situated<br />

at the crossroads <strong>of</strong> gender and immune regulation, we<br />

hypothesized that this gene may mediate sex differences in<br />

the development <strong>of</strong> T cell-mediated autoimmune disease.<br />

We show that PPAR is more abundant in male as compared<br />

to female CD4 + cells and is sensitive to androgen levels.<br />

Genetic ablation <strong>of</strong> this gene selectively removed the brake<br />

on NFkappaB and c-jun activity in male T lymphocytes<br />

resulting in higher production <strong>of</strong> IFN-γ and TNF (but not IL-<br />

17), and lower production <strong>of</strong> Th2 cytokines. Upon induction<br />

<strong>of</strong> experimental autoimmune encephalomyelitis (EAE), male<br />

but not PPAR −/− mice developed more severe clinical signs<br />

that were restricted to the acute phase <strong>of</strong> disease. To<br />

determine the extent to which changes in PPAR expression<br />

explain the androgen sensitivity <strong>of</strong> T helper responses, we<br />

contrasted the effects <strong>of</strong> castration on male WT versus<br />

PPAR −/− mice. This surgical intervention lowered PPAR<br />

expression in T cells, and enhanced the production <strong>of</strong> IFNγ,<br />

TNF, and IL-17 by WT male T cells in response to anti-CD3<br />

and anti-CD28 stimulation. Castration also enhanced the<br />

severity <strong>of</strong> acute EAE in male WT mice. These effects <strong>of</strong><br />

castration were significantly blunted in PPAR −/− mice. These<br />

results suggest that males may be less prone to develop<br />

Th1-mediated autoimmunity because they have higher<br />

levels <strong>of</strong> PPAR.<br />

doi:10.1016/j.clim.2007.03.390


Abstracts<br />

Sa.1 Nitric Oxide Metabolites and Nitrotyrosine in<br />

Asthma and COPD<br />

Jenny Garmendia, <strong>Clinical</strong> Immunologist and Internal<br />

Medicine, Instituto de Inmunología, UCV, Caracas,<br />

Venezuela, Dolores Moreno, Pulmonologist, Servicio de<br />

Neumonología, Hospital Universitario de Caracas, Caracas,<br />

Venezuela, Nancy Larocca, <strong>Clinical</strong> Immunologist and<br />

Pediatrician, Instituto de Inmunología, UCV, Caracas,<br />

Venezuela, Juan De Sanctis, Biochemistry, Instituto de<br />

Inmunología, UCV, Caracas, Venezuela, Carlos Tálamo,<br />

Pulmonologist, Servicio de Neumonología, Hospital<br />

Universitario de Caracas, Caracas, Venezuela<br />

Asthma and chronic obstructive pulmonary disease (COPD)<br />

are two different inflammatory airway diseases. Serum nitric<br />

oxide (NO) metabolites and nitrotyrosine may be useful<br />

parameters <strong>of</strong> inflammation. We studied 150 Asthma patients<br />

(24 ± 4.5 years), 185 COPD patients (62 ± 9 years), and 300<br />

healthy subjects (45 ± 19 years). The controls were divided in<br />

twogroups<strong>of</strong>150individualseach(meanage25±5and62±6<br />

years, respectively). Serum nitrites, nitrates (NO metabolites)<br />

and nitrotyrosine levels were determined by colorimetric,<br />

enzymatic assay and ELISA, respectively. No significant differences<br />

were recorded among the two groups <strong>of</strong> controls and thus<br />

we used the whole group for comparison. Nitrite levels were<br />

significantly reduced (pb 0.001) only en COPD patients (COPD<br />

4.1±1.9, Asthma 6.1 ± 1.3 vs. 6.5 ± 1.7 µM). Nitrates levels were<br />

significantly reduced in both groups <strong>of</strong> patients (COPD 16.2 ±<br />

4.1, Asthma 18.6 ± 3.2 vs controls 24.9 ± 3.5 µM, p b 0.001).<br />

Nevertheless, nitrotyrosine levels were significantly increased<br />

in the patient groups (controls: 4.2 ± 2.5; COPD: 11.3 ± 8.5<br />

pb0.001; Asthma: 6.7 ± 4.3 uM pb0.01). Nitrotyrosine levels<br />

were higher in COPD patients as compared to asthmatics (pb<br />

0.01). Moreover, we observed an inverse correlation between<br />

nitrates and nitrotyrosine only in patients (r = 0.62, n = 335). We<br />

conclude that metabolism <strong>of</strong> NO is modified in patients with<br />

asthma and COPD with a bypass <strong>of</strong> normal NO metabolites to a<br />

reactive species <strong>of</strong> nitrogen as peroxynitrites assessed partially<br />

by nitrotyrosine. FONACIT G2005000389.<br />

doi:10.1016/j.clim.2007.03.391<br />

Poster Session<br />

Saturday, June 9<br />

7:30 am−7:00 pm<br />

Authors Present: 5:45 pm−7:00 pm<br />

Sa.5 Effect <strong>of</strong> Omalizumab on Pulmonary Function<br />

and Asthma-related Outcome Measures in Patients<br />

Allergic to Cat Dander<br />

Marc Massanari, Associate Medical Director, Novartis<br />

Pharmaceuticals Corporation, East Hanover, NJ, Robert<br />

Maykut, Medical Director, Novartis Pharmaceuticals<br />

Corporation, East Hanover, NJ, Robert Zeldin, Executive<br />

Medical Director, Novartis Pharmaceuticals Corporation,<br />

East Hanover, NJ, Farid Kianifard, Statistician, Novartis<br />

Pharmaceuticals Corporation, East Hanover, NJ, Gregory<br />

Geba, Therapeutic Area Head, Novartis Pharmaceuticals<br />

Corporation, Verona, NJ<br />

Rationale: Exposure to cat dander, which if <strong>of</strong>ten<br />

unexpected, can worsen control in susceptible asthmatic<br />

patients. Add-on omalizumab (OMA) has been shown to help<br />

protect patients with perennial allergen sensitivity from<br />

exacerbations. We evaluated the effect <strong>of</strong> OMA on FEV1 and<br />

asthma-related outcome measures in patients with asthma<br />

and cat dander sensitivity. Methods: Data were pooled from 5<br />

randomized, double-blind, 28–32 week, placebo (PBO)controlled<br />

studies <strong>of</strong> patients with moderate–severe persistent<br />

IgE-mediated asthma and perennial allergen sensitivity<br />

inadequately controlled on inhaled corticosteroids (ICS). Cat<br />

sensitivity was determined by skin prick or RAST testing. The<br />

effect <strong>of</strong> add-on OMA on mean changes from baseline in FEV1,<br />

total rescue puffs <strong>of</strong> beta-agonist, and total asthma symptom<br />

score were analyzed using analysis <strong>of</strong> covariance; least<br />

squares means (LSM) and 95% confidence intervals (CI) were<br />

calculated. Results: 1589 patients (811 OMA, 778 PBO) were<br />

sensitive to cat dander. Mean age =39.7 years, 60% were<br />

female, mean IgE=219.1 IU/mL and FEV1 percent predicted<br />

was 60–79% in 46% and b60% in 26% <strong>of</strong> patients at baseline.<br />

End-<strong>of</strong>-study mean FEV1 increased by 100.3 mL (5.4%) and<br />

33.1 mL (2.5%) in OMA- and PBO-treated patients, respectively.<br />

LSM treatment difference was 63.11 (26.88, 99.35;<br />

pb0.001) in favor <strong>of</strong> OMA. The LSM difference (OMA-PBO)<br />

for change in rescue beta-agonist puffs was −0.47 (95% CI<br />

−0.72, −0.22; pb0.001) and was −0.40 (−0.55, −0.25;<br />

pb0.001) for change in total asthma symptom score. Conclusion:<br />

Omalizumab improved pulmonary function and asthmarelated<br />

outcome measures in cat allergic patients with<br />

moderate–severe persistent asthma inadequately controlled<br />

with ICS.<br />

doi:10.1016/j.clim.2007.03.392<br />

S75<br />

Sa.6 Hypersensitivity Pneumonitis <strong>of</strong> Unidentifiable<br />

Cause<br />

Viktor Hanak, MD, Mayo Clinic Foundation, Rochester, MN,<br />

Jason Golbin, MD, Mayo Clinic Foundation, Rochester,<br />

MN, Jay Ryu, MD, Mayo Clinic Foundation, Rochester, MN<br />

Identification <strong>of</strong> a specific antigen is important in disease<br />

management since antigen avoidance is necessary. Frequency<br />

<strong>of</strong> HP cases where causative antigen is not identifiable<br />

despite a thorough medical evaluation is not known.<br />

Objective: To assess the proportion <strong>of</strong> HP cases with no<br />

identifiable inciting antigen. Methods: Consecutive cases <strong>of</strong><br />

HP diagnosed at Mayo Clinic Rochester, MN between 1997 and


S76 Abstracts<br />

2002 were reviewed. Diagnostic criteria included: (1)<br />

respiratory symptoms, (2) compatible radiologic changes,<br />

(3) known exposure or a positive serologic test to an inciting<br />

antigen, and (4) no other identifiable cause for the lung<br />

disease. If there was no identifiable inciting antigen, one <strong>of</strong><br />

following two criteria was required: (1) diagnostic lung biopsy<br />

or (2) bronchoalveolar lavage lymphocytosis and compatible<br />

chest CTchanges. Results: We identified 85 consecutive cases<br />

<strong>of</strong> HP. The mean age (±SD) was 53±14 years; 53 (62%) patients<br />

were women. Cause was identified in 64 patients (75%). In 21<br />

patients (25%) the inciting antigen could not be identified by<br />

exposure history or serologic testing. In all 21 <strong>of</strong> these<br />

patients the diagnosis <strong>of</strong> HP was supported by histopathologic<br />

findings with diagnostic samples obtained in 18 patients by<br />

surgical and in 3 patients by bronchoscopic lung biopsy.<br />

Conclusions: The inciting antigen was not identifiable in 25%<br />

<strong>of</strong> patients evaluated for HP at a tertiary referral care center<br />

in the Midwest region <strong>of</strong> U.S.<br />

doi:10.1016/j.clim.2007.03.393<br />

Sa.7 Comparison <strong>of</strong> Influences <strong>of</strong> Three Types <strong>of</strong><br />

Music (Western, Classical, Traditional Persian and<br />

Pop) on Immunological Parameters<br />

Alireza Salekmoghaddam, Pr<strong>of</strong>essor, Iran University <strong>of</strong><br />

Medical Sciences, <strong>Immunology</strong>, Tehran, Iran, Alireza Salek<br />

Moghddam, Pr<strong>of</strong>essor, <strong>Immunology</strong>, Tehran, Iran, Saba Arshi,<br />

Assistant Pr<strong>of</strong>essor, <strong>Immunology</strong>, Tehran, Iran, Mohammad<br />

Kamali, Assistant Pr<strong>of</strong>essor, Statistics, Tehran, Iran, Hassan<br />

Ashayeri, Pr<strong>of</strong>essor, Neuropsychology, Tehran, Iran,<br />

Gholomreza Azadi, Technologist, <strong>Immunology</strong>, Tehran, Iran<br />

28 allergic rhinitis patients who had inclusion, exclusion<br />

criteria for this intervention were divided in four groups, three<br />

interventional groups and one control group. In each there were<br />

seven subjects related to their favorite music. Methods: Blood<br />

samples were taken before and after 1 month interventional<br />

period. Samples were examined immediately by flow cytometry<br />

and six cellular parameters (CD3, CD4, CD8, CD16, CD19 and<br />

HLA-DR) were measured and were analyzed by t test and<br />

between group by ANOVA. Conclusion: Evaluation <strong>of</strong> immune<br />

cells in atopic patients that listened to pop music showed<br />

significant increases in CD3 T cells (p=0.03) and CD4+ cells<br />

(p=0.03), CD8+ cells (p=0.05), and NK cells (p=0.014) that show<br />

immune response shifts to Th1 immune responses. As a result<br />

pop music had positive effects on atopic subjects. Possibly<br />

regular listening to favorite music can modulate immune<br />

parameters and improve signs and symptoms <strong>of</strong> allergic rhinitis<br />

in atopic patients.<br />

doi:10.1016/j.clim.2007.03.394<br />

Sa.8 Emergency Department (ED) Evaluation;<br />

Treatment, Hospital Admission and sIgE Levels in<br />

African American (AA) and Hispanic (H) Pediatric<br />

Asthmatics<br />

Ray Mun Loo, Resident in Pediatrics, Nassau University<br />

Medical Center, East Meadow, NY, Katarzyna Koscielna,<br />

Resident, Nassau University Medical Center, East Meadow,<br />

NY, Krishan Kumar, Attending in Pediatric Emergency<br />

Medicine, Nassau University Medical Center, East Meadow,<br />

NY, Marianne Frieri, Attending in Allergy <strong>Immunology</strong>,<br />

Nassau University Medical Center, East Meadow, NY<br />

Introduction: Asthma is higher in AA and 20–25% higher in<br />

the ED. We evaluated 66 ED adult asthmatics with class II<br />

allergens to cat, dust mite, mold and cockroach. This study<br />

evaluated pediatric patients on either nebulized Budesonide (B)<br />

or normal saline (NS) prior to admission. Methods: Eleven<br />

moderately severe asthmatic patients, age 8–16 years (9 AA; 2<br />

H) were clinically evaluated in the ED over 4 months and<br />

randomized to receive either 1 mg <strong>of</strong> nebulized (B) or (NS) as<br />

control. Improvement was based on pulmonary index score<br />

(PIS), peak flow (PF), and spirometry. FENO (Aerocrine) was<br />

measured in 3 patients. All non-responsive patients were<br />

admitted; sIgE levels were determined by sensitive ELISA’s<br />

(Quest; Hitachi Chemical Diagnostics). Results: Six received (B)<br />

with higher PSI <strong>of</strong> 6–9 vs.5on(NS).PF=59–267L/min. FEV1<br />

levels reversed post bronchodilation in 9 <strong>of</strong> 10, and B increased<br />

FEV1 relative to baseline (P=0.024). Increased FNO levels in 2<br />

had PSI <strong>of</strong> 8. sIgE levels in 9 <strong>of</strong> 11 tested=class III–IV to mite in 8;<br />

IV–Vtocockroach;II–IV to animal dander; II–IV to grass in 7; I–<br />

IV to mold; II–IV to rice and wheat in 6; I–III to ragweed in 5; II–<br />

VI to trees; II–IV to fish in 4. Conclusion: Polysensitization<br />

developed in 89% to inhalants and various foods but higher<br />

levels compared to our adult ED population. Education <strong>of</strong><br />

allergenic exposure, environmental and dietary control is<br />

essential to prevent hospitalization <strong>of</strong> pediatric asthmatics.<br />

doi:10.1016/j.clim.2007.03.395<br />

Sa.9 Association <strong>of</strong> the Polymorphisms <strong>of</strong> IL-4 Gene<br />

Promoter (−590CNT), IL-13 Coding Region (r130q)<br />

and IL-16 Gene Promoter (−295TNC) with Allergic<br />

Asthma<br />

Sara Hosseini-Farahabadi, Researcher, Bu-Ali Research<br />

Institute (BARI), Department <strong>of</strong> Immunogenetics, Mashhad,<br />

Iran, Jalil Tavakkol-Afshari, Vice President <strong>of</strong> Research,<br />

Head, Department <strong>of</strong> Immunogenetics and Cell Culture,<br />

Bu-Ali Research Institute (BARI), Department <strong>of</strong><br />

Immunogenetics and Cell Culture, Mashhad, Iran, Houshang<br />

Rafatpanah, Ghaem Medical Center, Department <strong>of</strong> Allergy<br />

and <strong>Immunology</strong>, Mashhad, Iran, Mohammad Khaje Daluei,<br />

Ghaem Medical Center, Department <strong>of</strong> Epidemiology and<br />

Statistics, Mashhad, Iran, Rez Farid-Hosseini, Head,<br />

Department <strong>of</strong> Allergy and <strong>Immunology</strong>, Ghaem Medical<br />

Center, Department <strong>of</strong> Allergy and <strong>Immunology</strong>, Mashhad<br />

Allergic asthma is a multifactorial disease, influenced by<br />

genetic and environmental factors. Recent family-based studies<br />

have revealed evidence for linkage <strong>of</strong> human chromosomes<br />

5q31–33, 12q15–24, 11q13 and 15q23.6 as regions likely to<br />

contain genes related to asthma. Among the candidate genes in<br />

these regions are the genes encoding for human interleukin-4,<br />

interleukin-13 and interleukin-16. To test this linkage, we<br />

examined an Iranian population <strong>of</strong> patients with asthma. A total<br />

<strong>of</strong> 30 patients with allergic asthma and 50 normal subjects were<br />

studied. Allergic asthma was confirmed using skin prick test and


Abstracts<br />

spirometry. DNA was extracted from whole blood and IL-4<br />

(−590CNT), IL-13 (R130Q) and IL-16 (−295TNC) polymorphisms<br />

were determined by PCR-RFLP method. Out <strong>of</strong> 30 patients with<br />

allergic asthma, 17 (56.7%) had CC, 8 (26.7%) had CT and 5<br />

(16.7%) had TT genotypes for IL-4 polymorphism, 11 (36.7%) had<br />

GG, 13 (43.3%) had GA and 6 (20%) had AA genotypes for IL-13<br />

polymorphism, 23 (76.7%) had TT, 7 (23.3%) had CT and no<br />

patient had CC genotypes for IL-16 polymorphism. A higher<br />

proportion <strong>of</strong> case subjects with the C allele for the IL-4, G<br />

allele for the IL-13 and T allele for the IL-16 polymorphisms<br />

were found compared with the T, A and C alleles respectively.<br />

These results do suggest an influence <strong>of</strong> genetic variability at<br />

the promoter <strong>of</strong> IL-4 gene (−590CNT) and a coding region <strong>of</strong> IL-<br />

13gene(R130Q)ontheoccurrence<strong>of</strong>allergicasthmaandno<br />

relationship between IL-16 promoter polymorphism (−295TNC)<br />

and this disease.<br />

doi:10.1016/j.clim.2007.03.396<br />

Sa.10 Lymphotactin Improves Treg Function in<br />

Asthmatics via the STAT5 Pathway<br />

Alison Fohner, Undergraduate Student, Stanford University,<br />

Pediatrics and <strong>Immunology</strong>, Stanford, CA, Khoa Nguyen, Lab<br />

Specialist, Stanford University, Pediatrics and <strong>Immunology</strong>,<br />

Stanford, CA, Alan M. Krensky, Doctor, Stanford University,<br />

Department <strong>of</strong> Pediatrics and <strong>Immunology</strong>, Stanford, CA,<br />

Kari C. Nadeau, Doctor, Stanford University, Department <strong>of</strong><br />

Pediatrics and <strong>Immunology</strong>, Stanford, CA<br />

Lymphotactin is a chemokine (XCL1) associated with the<br />

function <strong>of</strong> regulatory T cells (Treg) and with allergic airway<br />

inflammation. Compared to healthy controls (HC) (n=10),<br />

allergic asthma patients (AA) (n=10) have fewer Tregs and<br />

more invariant natural killer T cells (iNKT) in the periphery,<br />

and iNKT kill Treg. We reported that Treg in asthmatics are<br />

dysfunctional but that XCL1 increases their suppressive<br />

function in vitro. We hypothesized that incubation <strong>of</strong> Treg<br />

with XCL1 could increase Treg absolute numbers and/or<br />

improve Treg function in AA subjects. Therefore, we added<br />

200 ng/mL XCL1 to HC Treg (Foxp3+CD127−CD25+CD4+) vs.<br />

non-Treg (Foxp3−CD127−CD25−CD4+) and AA Treg vs. non-<br />

Treg. Incubation with XCL1 for 4 days increased the ratio <strong>of</strong><br />

Treg to non-Treg by 2–3×. Via a CFSE uptake assay,<br />

increased proliferation was not detected, suggesting that<br />

non-Treg were becoming Treg. We examined biochemical<br />

differences that could cause this conversion, including<br />

STAT1, STAT3, STAT5, and granzyme B expression since they<br />

have been implicated in Treg function. STAT5 phosphorylation<br />

increased in AA Treg and non-Treg, but not in HC or<br />

non-allergic asthmatics (NA). However, after XCL1 stimulation<br />

for 10 min, phosphorylated STAT5 increased significantly<br />

in non-Treg, but not Treg, <strong>of</strong> AA compared to HC<br />

(pb0.02) and NA (pb0.02). Since STAT5 phosphorylation<br />

plays a role in normal Treg function, these data support the<br />

conversion <strong>of</strong> non-Treg into Treg after incubation with<br />

XCL1. Thus, XCL1 functions through a STAT5 pathway to<br />

enhance Treg function in asthmatics. STAT5 could be a<br />

target for asthma therapy in the future.<br />

doi:10.1016/j.clim.2007.03.397<br />

Sa.11 Long-Term Tolerance in a Murine Model <strong>of</strong><br />

Type I Allergy Through Transplantation <strong>of</strong><br />

Genetically Modified Hematopoietic Stem Cells<br />

Ulrike Baranyi, PhD, Medical University <strong>of</strong> Vienna,<br />

Department <strong>of</strong> Surgery, Division <strong>of</strong> Transplantation, Vienna,<br />

Austria, Birgit Linhart, PhD, Division <strong>of</strong> Immunopathology,<br />

Department <strong>of</strong> Pathophysiology, Center <strong>of</strong> Physiology and<br />

Pathophysiology, Vienna, Austria, Nina Pilat, Division <strong>of</strong><br />

Transplantation, Department <strong>of</strong> Surgery, Medical University<br />

<strong>of</strong> Vienna, Vienna, Austria, John Iacomini, PhD,<br />

Transplantation Research Center, Brigham and Women’s<br />

Hospital and Women’s Hospital and Children’s Hospital,<br />

Boston, MA, Jessamyn Bagely, PhD, Transplantation<br />

Research Center, Brigham and Women’s Hospital and<br />

Children’s Hospital, Harvard Medical School, Boston, MA,<br />

Rudolf Valenta, MD, Division <strong>of</strong> Immunopathology, Center <strong>of</strong><br />

Physiology and Pathophysiology, Vienna, Austria, Thomas<br />

Wekerle, MD, Division <strong>of</strong> Transplantation, Department <strong>of</strong><br />

Surgery, Vienna, Austria<br />

Introduction: Current therapies for treating allergy are<br />

associated with various limitations. We thus investigated<br />

whether tolerance can be induced through transplantation <strong>of</strong><br />

syngeneic bone marrow retrovirally transduced to express an<br />

allergen. Methods: Balb/c bone marrow cells (BMC) were<br />

retrovirally transduced in vitro to express Phl p 5 (a major<br />

grass pollen allergen) in a membrane-anchored fashion<br />

(transduction efficiency: 35–55%). Myeloablated Balb/c<br />

mice received 2–4×10 6 transduced BMC iv and were thereafter<br />

repeatedly injected sc with recombinant Phl p 5 and<br />

rBet v 1 (an unrelated control allergen) (0.5 μg plus aluminum<br />

hydroxide, weeks 6, 9, 12 and 22). Allergen-specific antibody<br />

levels were determined in sera by ELISA, Phl p 5 expression by<br />

FACS. Results: All mice (n=10) transplanted with Phl p 5transduced<br />

BMC developed high levels <strong>of</strong> multi-lineage<br />

molecular chimerism which remained stable for the length<br />

<strong>of</strong> follow-up (up to 9 months) (e.g., 11 mean% Phl p 5+B cells<br />

and 22% T cells, 25 weeks post-BMT). Serum levels <strong>of</strong> Phl p 5specific<br />

IgE and IgG1 remained undetectable in all chimeras<br />

throughout follow-up while high levels <strong>of</strong> Bet v 1-specific IgE<br />

and IgG1 were measured. Basophil degranulation assays<br />

revealed the absence <strong>of</strong> Phl p 5-specific degranulation in<br />

chimeras, while in contrast Bet v 1-specific degranulation was<br />

preserved. In T cell proliferation assays chimeras showed<br />

specific non-responsiveness to Phl p 5 (n=3; pb0.01 vs. nontransduced<br />

mice). Conclusion: These pro<strong>of</strong>-<strong>of</strong>-principle<br />

experiments demonstra5.5e for the first time that molecular<br />

chimerism induces robust and long-lasting tolerance in<br />

allergy at the B cell, T cell and effector cell levels.<br />

doi:10.1016/j.clim.2007.03.398<br />

S77<br />

Sa.12 Lichen Planus (LP) Associated with Xolair<br />

Administration<br />

Filiz Seeborg, Instructor, Baylor College <strong>of</strong> Medicine and<br />

Texas Children’s Hospital, Section <strong>of</strong> Allergy and<br />

<strong>Immunology</strong>, Houston, TX, Pardeep S. Rihal, MD,<br />

West Houston Allergy and Asthma, Katy, TX, Adam Czelusta,<br />

MD, Katy Dermatology, PA, Katy, TX, Imelda C. Hanson,


S78 Abstracts<br />

Pr<strong>of</strong>essor, Baylor College <strong>of</strong> Medicine and Texas Childrens<br />

Hospital, Section <strong>of</strong> Allergy and <strong>Immunology</strong>, Houston, TX<br />

Background: Omalizumab (Xolair) is a recombinant<br />

monoclonal anti-immunoglobulin E (IgE) antibody that<br />

binds to free IgE, clearing it from the circulation. Xolair is<br />

approved for moderate to severe persistent asthma in<br />

adolescents and adults. Contraindication to use includes<br />

prior anaphylaxis to Xolair. Case report: We describe a 63year-old<br />

female who developed LP following Xolair administration.<br />

She had steroid-dependent asthma, allergic rhinitis,<br />

osteoarthritis, and hypertension. Her serum IgE level was<br />

55 KU/L. She developed a brief, generalized pruritus<br />

following first Xolair administration (300 μg/month). With<br />

second dosing, she had a generalized pruritic rash with lacy,<br />

violaceous facial papules lasting 2 weeks. Xolair and<br />

Dicl<strong>of</strong>enac were discontinued. A skin biopsy revealed LP.<br />

Following third dosing (6 months later), she developed white<br />

oral lesions in addition to the previously experienced pruritic<br />

rash. Discussion: LP is a T cell mediated inflammatory<br />

mucocutaneous condition with pruritic violaceous papules<br />

and plaques. <strong>Oral</strong> LP may predispose to development <strong>of</strong><br />

squamous cell carcinoma. LP may develop secondary to<br />

certain systemic medications and hepatitis C infection. No<br />

association between Xolair and LP is in the published<br />

literature. Malignant neoplasms have been reported during<br />

Xolair treatment, but a causal relationship has not been<br />

documented. Our case demonstrates a clear temporal<br />

association between Xolair use/cessation and the development/disappearance<br />

<strong>of</strong> LP. Per the manufacturer, malignancy<br />

risk for Xolair is unknown in individuals with existing<br />

malignant risk (oral LP). Conclusion: Repeated exposure to<br />

immunoregulatory therapy using Xolair may have potential<br />

to produce a T cell mediated immune activation leading to<br />

LP.<br />

doi:10.1016/j.clim.2007.03.401<br />

Sa.13 Inducible Expression <strong>of</strong> the Proallergic<br />

Cytokine TSLP in Airway Epithelial Cells is<br />

Controlled by NFkappaB<br />

Hai-Chon Lee, Postdoctoral Fellow, <strong>Immunology</strong> Program,<br />

Benaroya Research Institute, Seattle, WA, Steven F. Ziegler,<br />

Program Reader, <strong>Immunology</strong> Program, Benaroya Research<br />

Institute, Seattle, WA<br />

The epithelial-derived cytokine thymic stromal lymphopoietin<br />

(TSLP) is important for the initiation <strong>of</strong> allergic<br />

airway inflammation through a dendritic cell-mediated Th2<br />

response. To identify the factors that control TSLP expression,<br />

we examined the ability <strong>of</strong> inflammatory mediators to<br />

regulate TSLP production in human airway epithelial cells.<br />

We found that both IL-12 and TNF-± were capable <strong>of</strong><br />

inducing rapid TSLP production in primary human bronchial<br />

airway epithelial cells. We further characterized the human<br />

TSLP gene promoter using two human epithelial cell lines,<br />

16HBEo and A549, and showed that IL-12- and TNF-±mediated<br />

human TSLP promoter activation in these cells<br />

was mediated by an upstream NFkB site. Mutation <strong>of</strong> this<br />

NFkB site completely abolished activation, as did overexpression<br />

<strong>of</strong> a dominant-negative version <strong>of</strong> IKK 2 . Interestingly,<br />

human TSLP mRNA levels were also increased<br />

following exposure to TLR2, 8, and 9 ligands, further<br />

supporting an important role for NFkB in TSLP gene<br />

regulation. In a similar manner, analysis <strong>of</strong> the mouse<br />

TSLP gene promoter revealed the presence <strong>of</strong> a similar<br />

situated NFkB site that was also critical for IL-12-inducible<br />

expression <strong>of</strong> mouse TSLP. Taken together, these results<br />

demonstrate that the inflammatory mediators IL-12 and<br />

TNF-± regulate human TSLP gene expression in an NFkBdependent<br />

manner.<br />

doi:10.1016/j.clim.2007.03.402<br />

Sa.16 CpG Induces Th1 Response in Neonatal Mice<br />

but Does Not Suppress Anaphylactic IgG1 Response:<br />

Possible Role <strong>of</strong> Low TLR-9 Expression<br />

Cyro Alves Brito, PharmD, University <strong>of</strong> São Paulo, Brazil,<br />

Department <strong>of</strong> Dermatology, São Paulo, Brazil, Ana Elisa<br />

Fusaro, PharmD, University <strong>of</strong> São Paulo, Brazil,<br />

Department <strong>of</strong> Dermatology, São Paulo, Brazil, Jeferson<br />

Russo Victor, BSc, University <strong>of</strong> São Paulo, Brazil,<br />

Department <strong>of</strong> Dermatology, São Paulo, Brazil, Adriana<br />

Leticia Goldoni, BSc, University <strong>of</strong> São Paulo, Brazil,<br />

Department <strong>of</strong> Dermatology, São Paulo, Brazil, Paula<br />

Ordonhez Rigato, BSc, University <strong>of</strong> São Paulo, Brazil,<br />

Department <strong>of</strong> Dermatology, São Paulo, Brazil, Alberto Jose<br />

Silva Duarte, MD, University <strong>of</strong> São Paulo, Brazil,<br />

Department <strong>of</strong> Dermatology, São Paulo, Brazil, Maria<br />

Notomi Sato, BSc, University <strong>of</strong> São Paulo, Brazil,<br />

Department <strong>of</strong> Dermatology, São Paulo, Brazil<br />

It has been described that there are several differences<br />

between immune responses in adults and newborns. The<br />

aim <strong>of</strong> this work was to evaluate in early life and adult<br />

stage <strong>of</strong> mice, the immune modulatory effects <strong>of</strong> oligodeoxynucleotides<br />

containing CpG motif (CpG-ODN) in<br />

immunization with ovalbumin (OVA) and Blomia tropicalis<br />

(Bt). Three-day-old and 8–10-week-old A/Sn mice were<br />

immunized with OVA or Bt in alum, associated with CpG-<br />

ODN or control-ODN. Spleen cells from non-immunized mice<br />

were cultured with CpG or control-ODN for TLR-9 expression<br />

analysis. Neonate and adult co-administration <strong>of</strong> CpG<br />

with OVA or Bt was able to significantly decrease IgE<br />

response in parallel to an enhancement <strong>of</strong> IgG2a antibody<br />

levels compared to control mice. Furthermore, spleen cells<br />

from newborns immunized with OVA associated to CpG<br />

showed increased IFN-g production after anti-CD3 or LPS<br />

stimuli compared to control group. However, the negative<br />

regulation <strong>of</strong> IgE response was more evident in adult- than<br />

in neonate-immunized mice, also CpG down-regulates<br />

anaphylactic anti-OVA IgG1 production only in adult mice.<br />

Next we investigated whether an altered TLR-9 expression<br />

could be involved in impaired CpG effect in neonates. It<br />

was observed that after 48 hours <strong>of</strong> CpG stimulus TLR-9<br />

expression was already up-regulated in adult but not in<br />

neonate B cells. The pattern <strong>of</strong> antibody and cytokine<br />

production suggests a Th1-type response induced by CpG-<br />

ODN. Administration <strong>of</strong> CpG in newborns may be less


Abstracts<br />

effective than in adults due, in part, to a decreased B cell<br />

sensitivity to CpG stimulus. Supported by FAPESP, LIM-56,<br />

and HC-FMUSP.<br />

doi:10.1016/j.clim.2007.03.403<br />

Sa.17 IgE-reactive 60S Ribosomal Protein P2 in<br />

Almond (Prunus Dulcis) and Walnut (Juglans Regia),<br />

a New Class <strong>of</strong> Food Allergen Cross-reactive with<br />

Fungal Aeroallergens<br />

Pallavi Tawde, PhD Candidate, Florida State University,<br />

Department <strong>of</strong> Biological Science and Institute <strong>of</strong> Molecular<br />

Biophysics, Tallahassee, FL, Mohsen Abolhassani, Visiting<br />

Scholar, Florida State University, Department <strong>of</strong> Biology,<br />

Tallahassee, FL, Vanessa Seamon, Research Technician,<br />

Florida State University, Department <strong>of</strong> Biology,<br />

Tallahassee, FL, Suzanne Teuber, Associate Pr<strong>of</strong>essor,<br />

University <strong>of</strong> California, Davis, Internal Medicine,<br />

Rheumatology and Allergy, Davis, CA, Fang Wang,<br />

Postdoctoraltoral Research Fellow, Florida State University,<br />

Department <strong>of</strong> Biology, Tallahassee, FL, Sandra Uratsu,<br />

Staff Research Associate, University <strong>of</strong> California,<br />

Davis, Department <strong>of</strong> Plant Sciences, Davis, CA, Abhya<br />

Dandekar, Pr<strong>of</strong>essor, University <strong>of</strong> California, Davis,<br />

Department <strong>of</strong> Plant Sciences, Davis, CA, Stefan Vieths,<br />

Pr<strong>of</strong>essor, Paul-Ehrlich-Institut, Department <strong>of</strong><br />

Allergology, Langen, Germany, Shridhar Sathe,<br />

Pr<strong>of</strong>essor, Florida State University, Department <strong>of</strong><br />

Food Science, Tallahassee, FL, Kenneth Roux, Pr<strong>of</strong>essor,<br />

Florida State University, Department <strong>of</strong> Biological<br />

Science and Institute <strong>of</strong> Molecular Biophysics,<br />

Tallahassee, FL<br />

Introduction: Tree nuts are a source <strong>of</strong> food allergens<br />

<strong>of</strong>ten associated with life-threatening reactions in susceptible<br />

individuals. Molecular characterization <strong>of</strong> the protein<br />

allergens is essential for a better understanding <strong>of</strong> the<br />

pathogenesis <strong>of</strong> atopic diseases, and in the development<br />

<strong>of</strong> more efficacious diagnostic and therapeutic modalities.<br />

We employed molecular cloning and expression to identify<br />

the 60S acidic ribosomal protein P2 (60S RP) as IgEreactive<br />

proteins in almonds and walnuts. 60S RPs have<br />

been described as aeroallergens in some molds. Results:<br />

Immunoscreening identified IgE-reactive almond and walnut<br />

cDNA clones, each encoding an 11,450 Da 60S acidic<br />

ribosomal protein P2 (60S RP), designated as Pru du 5 and<br />

Jug r 5, respectively, and sharing 91% sequence similarity.<br />

Of 27 sera from patients reporting allergy to almond and/<br />

or walnut, 9 (33%) had IgE reactive with the recombinant<br />

proteins: 8 reacted to both proteins and 1 only with<br />

walnut Jug r 5. The 9 positive patient sera, but no<br />

others, also reacted with, and were inhibited by,<br />

recombinant Fus c 1, a fungal (Fusarium culmorum) 60S<br />

RP aeroallergen. Native 60S RP was detected in both<br />

almond and walnut crude extracts by reaction with rabbit<br />

anti-Pru du 5 antibodies. Conclusions: 60S RP represents a<br />

new class <strong>of</strong> food allergen in plants. Over a third <strong>of</strong><br />

tested patients had IgE that bound almond (Pru du 5)<br />

and/or walnut (Jug r 5) homologues in in vitro assays.<br />

The data indicate considerable cross-reactivity between<br />

the almond, walnut, and fungal homologues.<br />

doi:10.1016/j.clim.2007.03.404<br />

Sa.18 Pistachio Vicilin, Pis v 1, is Allergenic and<br />

Cross-reactive with the Homologous Cashew<br />

Allergen, Ana o 1<br />

LeAnna N. Willison, Graduate Student, Florida State<br />

University, Tallahasee, FL, Pallavi Tawde, Graduate<br />

Student, Florida State University, Department <strong>of</strong> Biological<br />

Sciences, Tallahasee, FL, Jason M. Robotham,<br />

Postdoctoraltoral Fellow, Florida State University,<br />

Department <strong>of</strong> Biological Sciences, Tallahasee, FL, Suzanne<br />

S. Teuber, Associate Pr<strong>of</strong>essor, University <strong>of</strong> California<br />

Davis, Internal Medicine, Rheumatology and Allergy, Davis,<br />

CA, Richard Penny, Graduate Student, Florida State<br />

University, Department <strong>of</strong> Biological Sciences, Tallahasee,<br />

FL, Shridhar K. Sathe, Pr<strong>of</strong>essor, Florida State University,<br />

Food Sciences and Nutrition Department, Tallahasee, FL,<br />

Kenneth H. Roux, Pr<strong>of</strong>essor, Florida State University,<br />

Department <strong>of</strong> Biological Sciences, Tallahasee, FL<br />

Introduction: Several studies have noted potential crossreactivity<br />

between certain tree nuts, especially those<br />

within the same botanical family. A potential example<br />

includes pistachio and cashew, which are both from the<br />

Anacardiaceae family. Results: A pistachio cDNA library was<br />

screened to detect potential pistachio allergens. An isolate<br />

was sequenced, cloned, and expressed in E. coli. The<br />

isolate coded for a 7S vicilin-like protein designated Pis v 1.<br />

Reactivity to the allergen was screened using 18 patients’<br />

sera: 9 cashew- and pistachio-allergic, 6 cashew-allergic<br />

but had never eaten pistachios, 1 pistachio-allergic only,<br />

and 2 cashew-allergic only. IgE reactivity to Pis v 1 was<br />

found in the serum <strong>of</strong> 5 patients, all <strong>of</strong> which had histories<br />

<strong>of</strong> allergy to both tree nuts or had never eaten pistachio.<br />

Cross-reactivity between Pis v 1 and the cashew homologue,<br />

Ana o 1, was investigated using an inhibition dot blot<br />

assay. Of the tested sera, the 5 (28%) patients that<br />

recognized Pis v 1 also recognized Ana o 1 and IgE binding<br />

was completely inhibited after pre-incubation with either<br />

cloned allergen. Conclusion: Patients allergic to cashew<br />

<strong>of</strong>ten report allergy to pistachio, which could be a result <strong>of</strong><br />

cross-reactivity between the two. This study revealed that<br />

28% <strong>of</strong> tested patients recognized the pistachio allergen Pis<br />

v 1 and these same patients also showed complete crossreactivity<br />

between the vicilin-like allergens from pistachio<br />

and cashew. Therefore, the pistachio allergen Pis v 1 is a<br />

likely contributor to the observed co-sensitivity to pistachio<br />

and cashew in some patients.<br />

doi:10.1016/j.clim.2007.03.405<br />

Sa.19 Infusion Reactions from Human<br />

Plasma-derived Products<br />

Hon-Sum Ko, Medical Officer, Food and Drug<br />

Administration/Division <strong>of</strong> Hematology, Potomac, MD<br />

S79


S80 Abstracts<br />

Introduction: Although human plasma-derived products<br />

are generally considered to have a favorable safety<br />

pr<strong>of</strong>ile, intravenous infusion <strong>of</strong> these products is occasionally<br />

associated with local or systemic reactions,<br />

varying from local discomfort to anaphylaxis-like manifestations.<br />

This study is an attempt to identify the factors<br />

that may affect the risks <strong>of</strong> such reactions. Materials and<br />

methods: A review was made in the clinical and<br />

laboratory data found in the package inserts or license<br />

applications <strong>of</strong> fifteen randomly chosen human plasmaderived<br />

products available in the U.S. The products<br />

included albumin, plasma protein fraction, intravenous<br />

immunoglobulins, coagulation factors, and enzyme inhibitors<br />

in plasma. The adverse effects from administration <strong>of</strong><br />

the products, including the frequency, severity, and<br />

potential attribution <strong>of</strong> local and systemic reactions<br />

were correlated with patient age, sex, ethnicity, allergic<br />

history, concomitant medications, previous infusion reactions,<br />

infusion rate, product class, product manufacture<br />

process, serum immunoglobulin levels, and other immune<br />

parameters whenever available. Results: Aside from<br />

infusion rate, which appears to correlate positively with<br />

the frequency <strong>of</strong> systemic reactions upon administration<br />

<strong>of</strong> immunoglobulin products, no consistent relationship<br />

between reaction frequency or severity and the above<br />

clinical/laboratory parameters was observed. A protective<br />

effect <strong>of</strong> premedication use before infusion was not<br />

observed. Conclusions: Much remains to be learned<br />

concerning the risk factors for infusion reactions to<br />

human plasma-derived products. Although current clinical<br />

and laboratory parameters may not be adequate to<br />

predict such risks, the development <strong>of</strong> newer biomarkers<br />

might <strong>of</strong>fer an opportunity to help mitigate potentially<br />

serious infusion reactions.<br />

doi:10.1016/j.clim.2007.03.406<br />

Sa.20 Tolerance-Inducing Capacity is Preserved in<br />

Dendritic Cells from Patients with SLE<br />

Jose Crispin, PhD Student, Department <strong>of</strong> <strong>Immunology</strong> and<br />

Rheumatology, Instituto Nacional de Ciencias Medicas y<br />

Nutricion SZ, Mexico City, Mexico, Leonardo Limon,<br />

Resident, Department <strong>of</strong> <strong>Immunology</strong> and Rheumatology,<br />

Instituto Nacional de Ciencias Medicas y Nutricion SZ,<br />

Mexico City, Mexico, Maria Ines Vargas Rojas, PhD Student,<br />

Department <strong>of</strong> <strong>Immunology</strong> and Rheumatology, Instituto<br />

Nacional de Ciencias Medicas y Nutricion SZ, Mexico City,<br />

Mexico, Jorge Alcocer Varela, Investigator, Department <strong>of</strong><br />

<strong>Immunology</strong> and Rheumatology, Instituto Nacional de<br />

Ciencias Medicas y Nutricion SZ, Mexico City, Mexico<br />

Granted with the faculty <strong>of</strong> inducing T cell activation<br />

and regulation, dendritic cell (DC) function is pivotal for<br />

the maintenance <strong>of</strong> tolerance. DC are able to promote<br />

anti-nuclear oriented responses and worsen SLE in animal<br />

models. The aim <strong>of</strong> this project was to analyze the<br />

phenotype and function <strong>of</strong> SLE-derived DC and explore<br />

their capacity to induce T cell tolerance. 23 patients with<br />

SLE and 11 controls were included. The phenotype <strong>of</strong><br />

circulating DC was analyzed by flow cytometry. Phenotype<br />

and cytokine production <strong>of</strong> monocyte-derived DC (MD-DC)<br />

was studied at the end <strong>of</strong> the differentiation period (basal)<br />

and after stimulation (LPS, TNF + PGE2 or anti-CD40).<br />

Ability to stimulate T cell proliferation was tested in<br />

allogeneic MLR. Phenotype and function <strong>of</strong> tolerogenic DC<br />

(grown in the presence <strong>of</strong> IL-10) was analyzed. The<br />

capacity <strong>of</strong> DC to induce tolerance towards allogeneic<br />

antigens, was analyzed in a recall assay. A higher<br />

proportion <strong>of</strong> DC from patients with SLE displayed<br />

costimulatory molecules in vivo (Pb0.05). Conversely,<br />

phenotype and T cell stimulatory capacity did not differ<br />

between MD-DC from patients and controls, neither in<br />

basal conditions, nor after stimulation. However, DC from<br />

patients with SLE showed a diminished response to<br />

stimulation via CD40. MD-DC from patients with SLE<br />

exhibited a normal capacity to induce tolerance towards<br />

alloantigens. DC from patients with SLE have an overstimulated<br />

phenotype when studied in vivo. However,such<br />

abnormality is absent when cells are grown in vitro.<br />

Tolerogenic MD-DC from SLE patients are capable <strong>of</strong><br />

inducing T cell tolerance.<br />

doi:10.1016/j.clim.2007.03.407<br />

Sa.21 The Largest International Collaborative<br />

Studies on Ocular Lesions in Behcet Disease<br />

Nobuyoshi Kitaichi, Ophthalmologist, Department <strong>of</strong><br />

Ophthalmology and Visual Sciences, Hokkaido University<br />

Graduate School <strong>of</strong> Medicine, Sapporo, Japan,<br />

Shigeaki Oh, Ophthalmologist, Department <strong>of</strong><br />

Ophthalmology and Visual Sciences, Hokkaido University<br />

Graduate School <strong>of</strong> Medicine, Sapporo, Japan<br />

Purpose: Behcet disease is predominantly found<br />

between East Asia and Mediterranean basin along the<br />

ancient Silk Road. In order to investigate the clinical<br />

features <strong>of</strong> ocular lesions in Behcet disease, international<br />

collaborative studies were performed by utilizing the<br />

same questionnaire. Methods: Descriptive questionnaires<br />

were sent to 132 ophthalmology centers in the world. Answers<br />

<strong>of</strong> 1465 patients were collected from 25 eye centers in 14<br />

countries; Australia, Germany, Greece, India, Iran, Italy,<br />

Japan, Jordan, Morocco, Portugal, Turkey, Saudi Arabia,<br />

Tunisia, and United Kingdom. Results: Men were seen in<br />

68.3% <strong>of</strong> the patients. The average age <strong>of</strong> onset was 27.5 years<br />

old. The frequency <strong>of</strong> HLA-B51 was detected in 62.0%.<br />

Recurrent oral aphthous ulcers were observed in 94.5%, skin<br />

lesions in 69.5% and genital ulcers in 61.4%. Allergic diseases<br />

were recognized in only 4.3% <strong>of</strong> the patients. Bilaterality and<br />

recurrency <strong>of</strong> intraocular inflammation were seen in 85.5%<br />

and 95.7%, respectively. Regarding visual prognosis, 23.3% had<br />

poor vision less than 0.1 (20/200) at the final visit. The<br />

percentage <strong>of</strong> poor vision was significantly higher in India,<br />

Iran, and Japan, and the lowest in Italy (pb0.01). Combined<br />

anterior and posterior segment intraocular inflammation<br />

(CAPSII), more serious than anterior segment intraocular<br />

inflammation, was seen more frequently in men (95.4%) than<br />

in women (89.9%, pb0.01). Conclusions: We performed the<br />

largest world-scale investigation <strong>of</strong> ocular lesions in Behcet


Abstracts<br />

disease. Th2-dominant allergic diseases seem quite rare in<br />

Th1-dominant Behcet disease. Men tend to have worse visual<br />

prognosis than women. <strong>Clinical</strong> pictures <strong>of</strong> Behcet disease<br />

vary regionally.<br />

doi:10.1016/j.clim.2007.03.408<br />

Sa.22 Splenic Macrophages Promotes Responses to<br />

Nucleosomes in NZB/W F1 Mice<br />

Akiko Okamoto, Department <strong>of</strong> Allergy and Rheumatology,<br />

School <strong>of</strong> Medicine, The University <strong>of</strong> Tokyo, Tokyo, Japan,<br />

Keishi Fujio, Department <strong>of</strong> Allergy and Rheumatology,<br />

School <strong>of</strong> Medicine, The University <strong>of</strong> Tokyo, Tokyo, Japan,<br />

Nico van Rooijen, Department <strong>of</strong> Cell biology and<br />

<strong>Immunology</strong>, Faculty <strong>of</strong> Medicine, Free University,<br />

Amsterdam, Netherlands, Kazuhiko Yamamoto, Pr<strong>of</strong>essor<br />

and Chairman <strong>of</strong> the Department <strong>of</strong> Allergy and<br />

Rheumatology, Department <strong>of</strong> Allergy and Rheumatology,<br />

School <strong>of</strong> Medicine, The University <strong>of</strong> Tokyo, Tokyo, Japan,<br />

Keith B. Elkon, Pr<strong>of</strong>essor <strong>of</strong> Medicine, Head, Division <strong>of</strong><br />

Rheumatology, Division <strong>of</strong> Rheumatology, University <strong>of</strong><br />

Washington, Seattle, WA<br />

Systemic lupus erythematosus (SLE) is characterized by<br />

autoantibody production and organ damage. Autoantigen<br />

presentation to T cells is important for the development <strong>of</strong><br />

lupus. Nucleosomes are major autoantigens in SLE. In this<br />

study, we examined nucleosome presentation in lupus-prone<br />

NZB×NZW F1 (NZB/W F1) mice. Nucleosome-specific T cells<br />

were generated by retroviral transfer <strong>of</strong> nucleosome-specific<br />

TCR. We found that nucleosome-specific T cells were<br />

stimulated dominantly in the spleen, compared to lymph<br />

nodes, lung, and thymus. Among splenic antigen-presenting<br />

cells (APCs), F4/80+ macrophages and CD11c+CD11b+<br />

dendritic cells stimulated nucleosome-specific T cells<br />

strongly. F4/80+ macrophage was the most abundant APC<br />

subset in the spleen. Then we investigated whether splenic<br />

phagocytes were responsible for nucleosome presentation<br />

and disease progression in vivo. Splenic phagocytes, mostly<br />

F4/80+ macrophages, were depleted in NZB/W F1 mice by<br />

intravenous injection <strong>of</strong> Cl2MDP (dichloromethylene diphosphonate)-liposomes.<br />

Cl2MDP-liposome treatment dramatically<br />

suppressed nucleosome presentation in the spleen.<br />

Moreover, Cl2MDP-liposome treatment at 20 and 22 weeks <strong>of</strong><br />

age significantly suppressed anti-nucleosome antibody (Ab)<br />

and anti-double stranded DNA (dsDNA) Ab production.<br />

Proteinuria progression was delayed and survival was<br />

prolonged in phagocyte-depleted mice. The numbers <strong>of</strong><br />

autoantibody secreting cells were decreased in the spleen<br />

from phagocyte-depleted mice. Multiple injections <strong>of</strong><br />

splenic F4/80+ macrophages, not CD11c+ dendritic cells,<br />

induced autoantibody production and proteinuria progression<br />

in NZB/W F1 mice. These results indicate that<br />

autoantigen presentation by splenic macrophages significantly<br />

contributes to autoantibody production and disease<br />

progression in lupus-prone mice.<br />

doi:10.1016/j.clim.2007.03.409<br />

Sa.23 A Comparison <strong>of</strong> TNF Dependent and<br />

Independent Features <strong>of</strong> Murine<br />

Spondyloarthropathy<br />

Peggy Jacques, MD, Ugent, University Hospital, Internal<br />

Medicine, Ghent, Belgium, Rik Lories, MD, PhD, University<br />

Hospitals Leuven, Internal Medicine, Leuven, Belgium, Ken<br />

Coppieters, M. Sc, Ugent, University Hospital, Internal<br />

Medicine, Ghent, Belgium, Katrien Van Beneden, PhD,<br />

Ugent, University Hospital, Internal Medicine, Ghent,<br />

Belgium, Herman Mielants, MD, PhD, Ugent, University<br />

Hospital, Internal Medicine, Gent, Belgium, Sylvie Seeuws,<br />

PhD, Ugent, University Hospital, Internal Medicine, Ghent,<br />

Belgium, Gust Verbruggen, MD, PhD, Ugent, University<br />

Hospital, Internal Medicine, Gent, Belgium, Martine De Vos,<br />

MD, PhD, Ugent, University Hospital, Internal Medicine,<br />

Gent, Belgium, Dirk Elewaut, MD, PhD, Ugent, University<br />

Hospital, Internal Medicine, Gent, Belgium<br />

Rheumatoid arthritis (RA) and spondyloarthropathies<br />

(SpA) are frequently occurring inflammatory disorders<br />

with distinct clinical features in which tumour necrosis<br />

factor (TNF) plays a predominant role. The focus <strong>of</strong> this<br />

study was to compare the typical features <strong>of</strong> three distinct<br />

arthritis models with special reference to synovitis and<br />

enthesitis. Collagen induced arthritis (CIA) is a well<br />

established model for RA in which peripheral joints are<br />

characterized by inflamed synovium and exudate in the<br />

synovial cavity. Proteoglycan depletion, cartilage destruction<br />

and bone loss are other features <strong>of</strong> this disease, which<br />

can be restored by TNF neutralisation. We also evaluated<br />

the TNF&DeltaARE mouse model, characterized by an<br />

increased TNF mRNA stability, where Crohn's disease is<br />

accompanied by peripheral arthritis. We demonstrated that<br />

this model, unlike CIA, is also characterized by dactylitis,<br />

enthesitis and axial involvement with sacroiliitis and<br />

spondylitis (histologically and by in vivo imaging (MRI)).<br />

Strikingly, severe bone demineralisation was observed but<br />

no signs <strong>of</strong> new bone formation, a common feature <strong>of</strong><br />

human SpA, were evident. Spontaneous ankylosing dactylitis<br />

(SpAD) in ageing male DBA/1 mice, which develop<br />

ankylosing enthesitis with enthesial cartilage and bone<br />

formation is an established SpA model characterized by<br />

dactylitis, onychoperiostitis, and joint space bridging,<br />

without axial involvement or bowel inflammation. Contrary<br />

to the inflammatory features in the first models, the new<br />

bone formation was not found to be enhanced by TNF<br />

neutralisation. Conclusion: These findings indicate that<br />

inflammatory features <strong>of</strong> murine SpA are largely dependent<br />

upon TNF, but other mechanisms control bone new<br />

formation frequently occurring in SpA.<br />

doi:10.1016/j.clim.2007.03.410<br />

S81<br />

Sa.24 The Presence <strong>of</strong> Anti-La Autoantibody is<br />

Associated with a Lower Risk <strong>of</strong> Nephritis and<br />

Seizures in Lupus Patients<br />

Sanober Malik, Medicine Resident, Department <strong>of</strong> Medicine,<br />

Oklahoma University Health Sciences Center, Oklahoma<br />

City, OK, Gail R. Bruner, <strong>Clinical</strong> Coordinator, Arthritis and


S82 Abstracts<br />

<strong>Immunology</strong>/Oklahoma Medical Research Foundation,<br />

Oklahoma City, OK, Lourdes Feo, Recruiter, Oklahoma<br />

Medical Research Foundation, Oklahoma City, OK, Courtney<br />

Williams-Weese, Recruiter, Oklahoma Medical Research<br />

Foundation, Oklahoma City, OK, R. Hal Sc<strong>of</strong>ield, Pr<strong>of</strong>essor,<br />

Department <strong>of</strong> Medicine, Oklahoma University Health<br />

Sciences Center/VA Medical Center/Oklahoma Medical<br />

Research Foundation, Oklahoma City, OK, John B. Harley,<br />

Pr<strong>of</strong>essor and Section Chief, Program Head, Department <strong>of</strong><br />

Medicine, Oklahoma University Health Sciences Center/VA<br />

Medical Center/Oklahoma Medical Research Foundation,<br />

Oklahoma City, OK, Amr H. Sawalha, Assistant Pr<strong>of</strong>essor,<br />

Department <strong>of</strong> Medicine, Arthritis and <strong>Immunology</strong><br />

Program, Oklahoma University Health Sciences Center/VA<br />

Medical Center/Oklahoma Medical Research Foundation,<br />

Oklahoma City, OK<br />

Background: Previous reports suggest a protective role for<br />

anti-La autoantibody against the development <strong>of</strong> lupus<br />

nephritis. We studied the effect <strong>of</strong> anti-La on the prevalence<br />

<strong>of</strong> nephritis in a large cohort <strong>of</strong> lupus patients. In addition, we<br />

determined the association between anti-La and the presence<br />

<strong>of</strong> the various lupus manifestations. Methods: We<br />

studied 1100 lupus patients enrolled in the Lupus Multiplex<br />

Registry and Repository. Only one lupus patient per family<br />

was selected to exclude intrafamilial correlation. Since anti-<br />

La is present in patients who also have anti-Ro autoantibody,<br />

we compared anti-Ro positive lupus patients in the presence<br />

or absence <strong>of</strong> anti-La. <strong>Clinical</strong> data were obtained from<br />

medical records, interviews, and participant questionnaires.<br />

Tests for autoantibodies against extractable nuclear antigens<br />

were performed using immunodiffussion assays. Results:<br />

There is no difference in the age, sex, or race between the<br />

anti-La positive and anti-La negative lupus patients. The<br />

presence <strong>of</strong> anti-La is associated with a significantly reduced<br />

risk <strong>of</strong> lupus nephritis (proteinuria: 29.3% versus 46.3%,<br />

OR=0.48, p =0.023; cellular casts: 8.6% versus 20.6%,<br />

OR=0.36, p=0.038). In addition, lupus patients with anti-La<br />

have a reduced risk for seizures (0% versus 11%, p=0.0096).<br />

The presence <strong>of</strong> anti-nRNP autoantibody is significantly<br />

reduced in anti-La positive compared to anti-La negative<br />

lupus patients (10.3% versus 27.4%, OR=0.30, p=0.0075).<br />

Conclusion: Anti-La autoantibody is associated with less<br />

severe lupus. Patients with anti-La have a lower risk <strong>of</strong> renal<br />

and CNS involvement compared to anti-La negative patients.<br />

doi:10.1016/j.clim.2007.03.411<br />

Sa.25 Establishment <strong>of</strong> an Animal Model for Allergic<br />

Granulomatous Vasculitis by IgE-mediated<br />

Cutaneous Reverse Passive Arthus Reaction:<br />

Contribution <strong>of</strong> Selectins and ICAM-1<br />

Minoru Hasegawa, Assistant Pr<strong>of</strong>essor, Dermatology,<br />

Kanazawa University, Kanazawa, Tomoyuki Fujita, Graduate<br />

Student, Dermatology, Kanazawa University, Kanazawa,<br />

Manabu Fujimoto, Associate Pr<strong>of</strong>essor, Dermatology,<br />

Kanazawa University, Kanazawa, Shinichi Sato, Pr<strong>of</strong>essor,<br />

Dermatology, Nagasaki University, Nagasaki, Kazuhiko<br />

Takehara, Pr<strong>of</strong>essor, Dermatology, Kanazawa University,<br />

Kanazawa<br />

Allergic granulomatous vasculitis (Churg–Strauss syndrome)<br />

occurs in patients with a history <strong>of</strong> asthma and is<br />

characterized by necrotizing vasculitis <strong>of</strong> small arteries and<br />

veins, with extravascular granulomas and a marked eosinophilia<br />

in the perivascular tissue <strong>of</strong> multiple organs and in the<br />

peripheral blood. IgE deposition in the lesion has been<br />

demonstrated, which suggests the involvement <strong>of</strong> IgE-containing<br />

immune complex (IC) in the pathomechanism. The classical<br />

and standard animal model for the inflammatory response in<br />

IC-mediated vasculitis is the reverse Arthus reaction. In the<br />

current study, we have established a mouse model mimicking<br />

allergic granulomatous vasculitis using cutaneous reverse<br />

passive Arthus reaction by IgE injection. IgE-mediated IC<br />

challenge induced hemorrhage and the accumulation <strong>of</strong><br />

eosinophil, mast cell, and macrophage, which similar to<br />

allergic granulomatous vasculitis. Remarkably, infiltration <strong>of</strong><br />

eosinophils and mast cells was observed, which was associated<br />

with the increased expression <strong>of</strong> MCP-3 and TNF-α. Furthermore,<br />

to assess the relative contribution <strong>of</strong> adhesion molecules,<br />

including selectins and ICAM-1, this model was<br />

examined in mice lacking E-selectin, P-selectin, L-selectin or<br />

ICAM-1. Eosinophil and mast cell numbers were significantly<br />

reduced in all mutant mice compared with wild type mice.<br />

Furthermore, both E- and P-selectin blockade in mice lacking<br />

for both L-selectin and ICAM-1 completely abrogated hemorrhage.<br />

These results indicate that E-, P-, L-selectin and ICAM-1<br />

contribute to the IgE-mediated cutaneous Arthus reaction by<br />

regulating eosinophil and mast cell recruitment and suggest<br />

that these adhesion molecules would be potential therapeutic<br />

targets for allergic granulomatous vasculitis.<br />

doi:10.1016/j.clim.2007.03.412<br />

Sa.26 Innate Immune Signal in New Murine Models <strong>of</strong><br />

Autoimmunity Induced by U1 RNA and the RNA<br />

Binding Domain <strong>of</strong> the U1-70KD Ribonucleoprotein<br />

Antigen<br />

Annette Abril, Resident in Medicine, University <strong>of</strong> Miami/<br />

Department <strong>of</strong> Medicine/Division <strong>of</strong> Rheumatology, Miami,<br />

FL, Kimberly Jaimes, Research Specialist, University <strong>of</strong><br />

Miami/Department <strong>of</strong> Medicine/Division <strong>of</strong> Rheumatology,<br />

Miami, FL, Robert W. H<strong>of</strong>fman, Pr<strong>of</strong>essor and Chief,<br />

University <strong>of</strong> Miami/Department <strong>of</strong> Medicine, Division<br />

Rheumatology and Department Microbiology and<br />

<strong>Immunology</strong>, Miami, FL<br />

We have recently developed two complementary murine<br />

models <strong>of</strong> systemic autoimmunity that are induced following<br />

a single exposure to U1 RNA and a polypeptide<br />

encompassing the RNA binding domain <strong>of</strong> the 70 kDa<br />

polypeptide <strong>of</strong> U1 RNP. In mice deficient for the toll-like<br />

receptor (TLR3−/−), tissue targeting in the model is shifted<br />

from the lungs to the kidneys, and anti-double stranded<br />

DNA (dsDNA) antibodies emerge. This study examines the<br />

influence <strong>of</strong> TLR3 and innate immune signaling on the<br />

induction <strong>of</strong> antiphospholipid antibodies in these new<br />

models. Methods: Mice transgenic for the human major<br />

histocompatibility complex gene HLA-DR4 and TLR3−/−<br />

mice were immunized with U1 RNA and U1-70kDa


Abstracts<br />

polypeptide, control fusion protein, with or without U1-<br />

RNA, or saline. Sera from control mice <strong>of</strong> the background<br />

strains C57BL/6 or B6/129 were also studied. Complete<br />

serologic and histologic analyses were performed. Anticardiolipin,<br />

anti-dsDNA and anti-U1-70kDa antibodies were<br />

measured by ELISA. Results: We found that the mice<br />

deficient for TLR3 failed to produce anticardiolipin antibodies<br />

despite the development <strong>of</strong> high titers <strong>of</strong> antidsDNA,<br />

anti-U1-70kDa and other antibodies, as well as<br />

developing proteinuria and proliferative glomerulonephritis.<br />

In fact, TLR3 deficient mice had reduced levels <strong>of</strong><br />

anticardiolipin reactivity in their sera as measured by ELISA<br />

compared to control mice (pb0.05). Conclusions: We have<br />

developed two new models <strong>of</strong> systemic autoimmunity that<br />

are induced following a single exposure to U1-RNA and its<br />

associated RNA binding protein. These models allow for<br />

genetic dissection <strong>of</strong> differences in tissue targeting,<br />

autoantibody development and the influences <strong>of</strong> innate<br />

immune signaling.<br />

doi:10.1016/j.clim.2007.03.413<br />

Sa.27 Urinary Lipocalin-2 is Upregulated in Murine<br />

and Human Lupus Nephritis<br />

Milena Pitashny, Posdoctoral Fellow, Albert Einstein College<br />

<strong>of</strong> Medicine, Division <strong>of</strong> Rheumatology, Bronx, NY<br />

Lipocalin-2, a member <strong>of</strong> the lipocalin family <strong>of</strong> iron<br />

binding proteins, is upregulated in the kidney following a<br />

variety <strong>of</strong> insults that lead to renal injury. We recently<br />

reported that in vitro exposure <strong>of</strong> kidney cells to monoclonal<br />

anti-double stranded (ds) DNA antibodies significantly upregulates<br />

Lipocalin-2. We hypothesized that polyclonal pathogenic<br />

anti-dsDNA antibodies induce kidney overexpression <strong>of</strong><br />

Lipocalin-2, and the latter may represent a marker <strong>of</strong> renal<br />

involvement in lupus-prone mice and SLE patients. Kidney<br />

and serum Lipocalin-2 from old lupus-prone MRL lpr/lpr<br />

(MRL/lpr), NZB×NZW F1 (B/W) and non-autoimmune control<br />

mouse strains were studied. B/W and MRL/lpr mice had<br />

significantly higher kidney RNA expression <strong>of</strong> Lipocalin-2 than<br />

age-matched BALB/c mice (pb0.01). Furthermore, MRL/lpr<br />

had higher levels <strong>of</strong> serum Lipocalin-2 than MRL+/+ and<br />

BALB/c mice (pb0.05). Kidney Lipocalin-2 correlated with<br />

serum Lipocalin-2 (pb0.005), proteinuria (pb0.05) and antidsDNA<br />

antibody titers (pb0.05). We measured Lipocalin-2<br />

levels in urine from patients with or without active lupus<br />

nephritis (LN). SLE patients had significantly higher levels <strong>of</strong><br />

urinary (u) Lipocalin-2 than normal controls (pb0.005).<br />

Moreover, patients with active LN had significantly higher<br />

levels <strong>of</strong> uLipocalin-2 than patients without LN (pb0.05).<br />

Finally, in LN patients we found a significant correlation<br />

between uLipocalin-2 levels and the renal SLE disease<br />

activity index (SLEDAI) (r=0.45, pb0.01). Upregulation <strong>of</strong><br />

Lipocalin-2 in the kidneys <strong>of</strong> lupus prone mice and SLE<br />

patients with nephritis strongly suggests that Lipocalin-2 may<br />

serve as a valuable marker <strong>of</strong> nephritis in SLE, although its<br />

role in the pathogenesis <strong>of</strong> LN has yet to be determined.<br />

doi:10.1016/j.clim.2007.03.414<br />

Sa.28 CD4+ T Cells Reactive with U1-70kD<br />

Autoantigen Can Transfer Disease and are Central to<br />

Pathogenesis in New Murine Model <strong>of</strong> Lupus<br />

Robert W. H<strong>of</strong>fman, Pr<strong>of</strong>essor and Chief, University <strong>of</strong><br />

Miami/Department <strong>of</strong> Medicine, Division Rheumatology and<br />

Department Microbiology and <strong>Immunology</strong>, Miami, FL, Eric<br />

L. Greidinger, Assistant Pr<strong>of</strong>essor, University <strong>of</strong> Miami/<br />

Department <strong>of</strong> Medicine, Division Rheumatology and<br />

Department Microbiology and <strong>Immunology</strong>, Miami, FL, Yun<br />

Zang, Research Scientist, University <strong>of</strong> Miami/Department<br />

<strong>of</strong> Medicine, Division Rheumatology and Department<br />

Microbiology and <strong>Immunology</strong>, Miami, FL<br />

C57BL/6 mice transgenic for the human major histocompatibility<br />

complex gene HLA-DR4 (DR4-Tg) were immunized<br />

with U1 RNA and the U1-70kDa polypeptide, control fusion<br />

protein, with or without U1-RNA, or saline. Complete serologic<br />

and histologic analyses were performed on all mice. CD4+ T<br />

cells and APCs were isolated from immunized mice using<br />

immunoaffinity columns and tested for proliferation. Either<br />

freshly isolated CD4+ T cells from immunized mice or T cell<br />

lines were adoptively transferred to unmanipulated HLA-DR4<br />

Tg or RAG−/− mice. Freshly isolated CD4+ T cell from<br />

immunized DR4 Tg mice proliferated in response to U1-<br />

70kDa and to synthetic peptides from the RNA binding domain<br />

region <strong>of</strong> the protein in the presence <strong>of</strong> irradiated autologous<br />

APCs. We found that CD4+ T cells specific for U1-70kDa were<br />

able to transfer disease to unmanipulated syngeneic mice.<br />

Furthermore, using purified CD4+ Tcells or Tcell lines specific<br />

for U1-70kDa we were able to transfer disease to RAG−/− mice<br />

in the absence <strong>of</strong> detectable B cells or autoantibodies. In<br />

conclusion, we have developed a new model <strong>of</strong> systemic<br />

autoimmunity that is induced following a single exposure to<br />

U1-RNA and its associated RNA binding protein. Anti-U1-70kDa<br />

autoantigen-specific CD4+ T cells appear to medicate pathogenesis<br />

<strong>of</strong> disease in this model and can do so in the absence <strong>of</strong><br />

detectable B cells or autoantibodies. This work was supported<br />

by NIH award AR43308 (RWH), the Lupus Foundation <strong>of</strong><br />

American (RWH) and Department <strong>of</strong> Veterans Affairs Merit<br />

Review awards (RWH and ELG).<br />

doi:10.1016/j.clim.2007.03.415<br />

S83<br />

Sa.29 Nasal Delivery <strong>of</strong> Anti-CD3 Antibody Suppresses<br />

Murine Lupus by Inducing IL-10 Producing<br />

Foxp3+CD4+CD25−LAP+ Regulatory T Cells<br />

Henry Yim Wu, Instructor, Harvard Medical School, Boston, MA,<br />

Howard Weiner, Pr<strong>of</strong>essor, Harvard Medical School, Boston, MA<br />

One <strong>of</strong> the major contributors to the development <strong>of</strong> SLE<br />

is a functionally defective T regulatory cell pool in the<br />

periphery <strong>of</strong> susceptible individuals. Therefore the induction<br />

<strong>of</strong> novel T regulatory cells that can control autoaggressive<br />

immune responses in patients holds promise as<br />

immunotherapy for SLE. We previously reported that oral<br />

administration <strong>of</strong> anti-CD3 monoclonal antibody suppresses<br />

experimental autoimmune encephalomyelitis in a TGF-?<br />

dependent fashion both in vitro and in vivo. We have now<br />

discovered that nasally administered murine anti-CD3 F(ab′)


S84 Abstracts<br />

2 monoclonal antibody (clone 2C-11) is biologically active<br />

and reduced the incidence <strong>of</strong> lupus nephritis and autoantibody<br />

production in the SNF1 accelerated lupus model. In<br />

addition nasal anti-CD3 arrested on-going lupus in NZB strain<br />

<strong>of</strong> lupus prone mice. Animals were given 5 treatments per<br />

week every other week for 12 weeks. Nasal administration<br />

<strong>of</strong> anti-CD3 antibody induced Foxp3+CD4+CD25−LAP+ T<br />

regulatory cells that mediate suppression in an IL-10<br />

dependent but contact independent manor in vitro.<br />

Adoptive transfer studies demonstrated that LAP+ T<br />

regulatory cells required IL-10 and TGF-? to suppress disease<br />

in vivo. We hypothesize that LAP+ T regulatory cells control<br />

the activation CD4+ICOS+CXCR5+ T follicular helper cells<br />

which in turn downregulate the differentiation <strong>of</strong> B cells<br />

into IgG+CD38+ plasma cells and inhibit the production <strong>of</strong><br />

anti-nuclear autoantibodies. Our findings identify a novel<br />

immunotherapeutic approach for the treatment <strong>of</strong> SLE that<br />

is applicable to humans.<br />

doi:10.1016/j.clim.2007.03.416<br />

Sa.30 Reverse Phase Protein Lysate Microarrays in<br />

the Analysis <strong>of</strong> Activated B Lymphocytes<br />

Alvina D. Chu, Rheumatology Fellow, Stanford University,<br />

Division <strong>of</strong> <strong>Immunology</strong> and Rheumatology, Palo Alto, CA,<br />

Andrzej J. Chruscinski, Cardiology Fellow, Stanford<br />

University, Division <strong>of</strong> Cardiovascular Medicine, Stanford,<br />

CA, Harvir Singh, Research Assistant, Stanford University,<br />

Division <strong>of</strong> <strong>Immunology</strong> and Rheumatology, Stanford, CA,<br />

Paul J. Utz, Associate Pr<strong>of</strong>essor, Stanford University,<br />

Division <strong>of</strong> <strong>Immunology</strong> and Rheumatology, Stanford, CA<br />

Systemic lupus erythematosus (SLE) is an autoimmune<br />

inflammatory disease characterized by destruction <strong>of</strong> multiple<br />

organ systems. The pathophysiology <strong>of</strong> SLE involves<br />

activated B cells that produce antibodies directed against<br />

self-antigens. Our laboratory has previously demonstrated<br />

that reverse phase protein (RPP) lysate microarrays can be<br />

used to detect multiple intracellular phosphorylation events<br />

from T cell populations maintained in cell culture in vitro.<br />

Here we show a novel application <strong>of</strong> RPP lysate microarray<br />

technology to analyze activated B lymphocytes taken ex<br />

vivo from MRL/MpJ-Faslpr (MRL/lpr) mice. Splenocytes<br />

were harvested from MRL/lpr mice at ∼25 weeks <strong>of</strong> age,<br />

toward the end <strong>of</strong> their lifespan. B lymphocytes were<br />

isolated by negative selection using magnetic beads and<br />

then briefly stimulated with B cell activating factors,<br />

including phorbol 12-myristate 13-acetate (PMA). Using a<br />

robotic microarrayer, nanoliter amounts <strong>of</strong> lysate were<br />

deposited onto nitrocellulose-coated slides. Slides were<br />

probed with phosphorylation state-dependent antibodies<br />

targeting key intracellular phosphorylation events involved<br />

with B cell activation, including phosphorylation <strong>of</strong> ERK,<br />

Akt, Jak, and STATs. Bound antibodies were detected using<br />

secondary antibodies conjugated to fluorophores, and mean<br />

fluorescence intensity for each sample was calculated. We<br />

found differential activation <strong>of</strong> kinase substrates within<br />

stimulated cells compared to basal levels within unstimulated<br />

B cell populations. Results were validated by Western<br />

blot analysis. Reverse phase protein microarrays provide a<br />

powerful tool for quantifying and comparing the level <strong>of</strong><br />

phosphorylation events in activated B cells isolated from<br />

secondary lymphoid organs, and show promise for elucidating<br />

signal transduction pathways involved in the pathogenesis<br />

<strong>of</strong> SLE.<br />

doi:10.1016/j.clim.2007.03.417<br />

Sa.31 Complement C4d Deposition on Circulating<br />

Blood Cells in Systemic Lupus Erythematosus (SLE)<br />

Chau-Ching Liu, Assistant Pr<strong>of</strong>essor, University <strong>of</strong> Pittsburgh<br />

School <strong>of</strong> Medicine, Department <strong>of</strong> Medicine, Pittsburgh,<br />

PA, Jean Lin, Medical Student, University <strong>of</strong> Pittsburgh<br />

School <strong>of</strong> Medicine, Pittsburgh, PA, Jeannine S. Navratil,<br />

Research Associate, University <strong>of</strong> Pittsburgh School <strong>of</strong><br />

Medicine, Department <strong>of</strong> Medicine, Pittsburgh, PA, Susan<br />

Manzi, Associate Pr<strong>of</strong>essor, University <strong>of</strong> Pittsburgh School<br />

<strong>of</strong> Medicine, Department <strong>of</strong> Medicine, Pittsburgh, PA, Amy<br />

H. Kao, Assistant Pr<strong>of</strong>essor, University <strong>of</strong> Pittsburgh School<br />

<strong>of</strong> Medicine, Department <strong>of</strong> Medicine, Pittsburgh, PA,<br />

Joseph M. Ahearn, Associate Pr<strong>of</strong>essor, University <strong>of</strong><br />

Pittsburgh School <strong>of</strong> Medicine, Department <strong>of</strong> Medicine,<br />

Pittsburgh, PA<br />

Immune dysregulation in SLE is characterized by polyclonal<br />

lymphocyte activation, autoantibody production, and<br />

complement activation. Recently, we discovered that significant<br />

levels <strong>of</strong> complement C4d are present on surfaces <strong>of</strong><br />

erythrocytes, platelets, and lymphocytes <strong>of</strong> SLE patients and<br />

that C4d-bearing T lymphocytes exhibited functional<br />

abnormalities. These observations led to the current study<br />

in which circulating blood cells from SLE patients were<br />

investigated for C4d deposition and for the underlying<br />

mechanism(s). Specifically, we performed flow cytometry<br />

to measure C4d levels on various circulating cells from 307<br />

SLE patients, 187 patients with other inflammatory diseases,<br />

and 95 healthy controls. The results demonstrated that<br />

significantly elevated levels <strong>of</strong> C4d were present on T<br />

lymphocytes, B lymphocytes, and monocytes <strong>of</strong> SLE patients<br />

(mean fluorescence intensity (MFI) =13.5, 50.6, and 9.7,<br />

respectively), compared to patients with other diseases<br />

(MFI=3.2, 6.9, 4.7, respectively; pb0.0001) and healthy<br />

controls (MFI=1.8, 8.0, and 3.7, respectively; pb0.0001).<br />

However, only modest levels <strong>of</strong> C4d were detected on<br />

granulocytes from a small number <strong>of</strong> SLE patients (MFI=2.9).<br />

Furthermore, high C4d levels were not necessarily concurrently<br />

present on erythrocytes, platelets, lymphocytes, and<br />

monocytes <strong>of</strong> a given SLE patient on a particular day. These<br />

findings, combined, indicate that the C4d-bearing circulating<br />

cell phenotype is mediated by a patient-dependent, cell<br />

lineage-specific mechanism rather than the result <strong>of</strong><br />

complement activation that simply affects all circulating<br />

cells simultaneously. Ongoing studies investigating the<br />

association <strong>of</strong> the differential leukocyte-C4d binding patterns<br />

with clinical features <strong>of</strong> SLE may lead to additional<br />

clues to SLE pathogenesis and may identify targets for<br />

therapeutic intervention.<br />

doi:10.1016/j.clim.2007.03.418


Abstracts<br />

Sa.32 Cell-Type and Stimulus Determine the Impact<br />

<strong>of</strong> the Serine/Threonine Kinase Cot/Tpl2 in Acute<br />

and Chronic Inflammation<br />

Anwar Murtaza, Senior Scientist, Abbott Bioresearch<br />

Center, Pharmacology, Worcester, MA, Shaughn Bryant,<br />

Research Associate, Abbott Bioresearch Center,<br />

Pharmacology, Worcester, MA, Suzanne Mathieu, Research<br />

Associate, Abbott Bioresearch Center, Pharmacology,<br />

Worcester, MA, Hamish Allen, Director, Abbott Bioresearch<br />

Center, Molecular and Cellular Biology, Worcester, MA,<br />

Catherine Tripp, Senior Director, Abbott Bioresearch<br />

Center, Worcester, MA, Neil Wishart, Associate Director,<br />

Abbott Bioresearch Center, Chemistry, Worcester, MA<br />

TNF has been identified as a key pro-inflammatory<br />

cytokine in the pathogenesis <strong>of</strong> acute and chronic inflammatory<br />

diseases such as septic shock and rheumatoid<br />

arthritis (RA). Anti-TNF therapy has been effective in the<br />

treatments <strong>of</strong> RA, Crohn’s disease (CD) and psoriasis (Ps).<br />

Binding <strong>of</strong> TNF to its receptors induces a number <strong>of</strong> signaling<br />

cascades leading to the activation <strong>of</strong> MAP kinases and NF-kB.<br />

Targeting Cot/Tpl2 a serine/threonine kinase in the MEK/ERK<br />

MAP kinase pathway <strong>of</strong>fers an attractive approach for<br />

developing an oral therapeutic that inhibits TNF production<br />

and hence an alternative strategy to anti-TNF therapy. Here<br />

we have used Tpl2 deficient mice to elucidate its role in<br />

acute and chronic inflammation. Our results demonstrate<br />

that Tpl2 deficiency decreases the production <strong>of</strong> a panel <strong>of</strong><br />

pro-inflammatory cytokines such as TNF, IL-1, IFN-g IL-6, and<br />

IL-18 following LPS challenge in vivo. We further demonstrate<br />

for the first time that Tpl2 knockout mice are only<br />

partially protected to collagen-induced arthritis (CIA).<br />

Moreover, the treatment <strong>of</strong> Tpl2 deficient mice with an<br />

anti-TNF antibody dramatically inhibited the progression and<br />

severity <strong>of</strong> disease suggesting a Tpl2 independent production<br />

<strong>of</strong> TNF in rodent arthritis. Moreover, we show that the impact<br />

<strong>of</strong> Tpl2 deficiency in LPS and arthritis models may be due to a<br />

differential activity <strong>of</strong> the kinase in different cell types.<br />

Thus, our data demonstrate that the Cot/Tpl2 pathway is<br />

critical in the response to endotoxin but may have a limited<br />

role in arthritis.<br />

doi:10.1016/j.clim.2007.03.419<br />

Sa.33 Bosentan Synergizes with Iloprost in Treating<br />

Pulmonary Hypertension in Scleroderma Patients<br />

Marta Sessarego, Doctor, Switzerland, Department <strong>of</strong><br />

Internal Medicine-University <strong>of</strong> Genova, Genova,<br />

Switzerland, Marta Rizzi, Department <strong>of</strong> Internal<br />

Medicine-University <strong>of</strong> Genova, Genova, Switzerland,<br />

Massimo Ghio, Department <strong>of</strong> Internal Medicine-University<br />

<strong>of</strong> Genova, Genova, Francesco Indiveri, Pr<strong>of</strong>essor,<br />

Department <strong>of</strong> Internal Medicine-University <strong>of</strong> Genova,<br />

Genova, Switzerland<br />

Pulmonary arterial hypertension (PAH) is a devastating<br />

complication <strong>of</strong> systemic sclerosis (SSc) characterized by<br />

endothelial dysfunction (lesser production <strong>of</strong> prostacyclin<br />

associated with increased secretion <strong>of</strong> endotelin), for which<br />

no curative treatments are available. On this basis it has<br />

been postulated that the oral dual endothelin receptors<br />

antagonist, bosentan, might strengthen the activity <strong>of</strong> prostacyclin<br />

therapy. Patients and methods: 8 patients affected<br />

by SSc with increased PAH (PA systolic pressure PAPS ¡Ý 45 mm<br />

Hg by echocardiogram or PA pressure ¡Ý 35 mm Hg at rest by<br />

cardiac catheterism, WHO functional classes III–IV) were<br />

reclutated, all <strong>of</strong> them had shown a poor response to 1 year<br />

treatment with prostacyclin e.v. infusion (iloprost, 0.5–2 ng/<br />

kg/min 6 h a day — 5 days a month). Bosentan (62.5 mg bid<br />

for 1 month, then 125 mg bid) and iloprost (0.5–2 ng/kg/min)<br />

in continuous (24h/24 h) were administered. The Doppler<br />

echocardiogram, the six-minute walking test (6MWT) and the<br />

complete pulmonary tests were performed at baseline and<br />

after 3, 6 and 12 months. Results: After 3 months PAPS was<br />

decreased and 6MWT was improved at least 25 min in all<br />

patients; at the end <strong>of</strong> the study an improvement <strong>of</strong> WHO<br />

class and pulmonary function was observed in 7 and 3<br />

patients respectively. All the subjects tolerated the therapy,<br />

thereafter no one discontinued therapy. Conclusion: The<br />

association <strong>of</strong> bosentan with iloprost infused continuously<br />

may improve PAP and hemodynamics in SSc patients.<br />

doi:10.1016/j.clim.2007.03.421<br />

S85<br />

Sa.35 Anti-lymphocyte Autoantibodies in Systemic<br />

Lupus Erythematosus (SLE) Revisited<br />

Jean Lin, Medical Student, University <strong>of</strong> Pittsburgh,<br />

Pittsburgh, PA, Joseph M. Ahearn, Associate Pr<strong>of</strong>essor,<br />

University <strong>of</strong> Pittsburgh School <strong>of</strong> Medicine, Department <strong>of</strong><br />

Medicine, Pittsburgh, PA, Susan Manzi, Associate Pr<strong>of</strong>essor,<br />

University <strong>of</strong> Pittsburgh School <strong>of</strong> Medicine, Department <strong>of</strong><br />

Medicine, Pittsburgh, PA, Chau-Ching Liu, Assistant<br />

Pr<strong>of</strong>essor, University <strong>of</strong> Pittsburgh School <strong>of</strong> Medicine,<br />

Department <strong>of</strong> Medicine, Pittsburgh, PA<br />

Complement, anti-lymphocyte autoantibodies (ALA), and<br />

lymphocyte dysfunction have previously been implicated in<br />

the pathogenesis <strong>of</strong> SLE. However, the causal relationships<br />

between complement, ALA, and lymphocyte function are<br />

largely unexplored. Our recent identification <strong>of</strong> complement<br />

C4d on T cells <strong>of</strong> SLE patients has led us to hypothesize that<br />

binding <strong>of</strong> ALA to T cells may trigger complement activation<br />

and C4d deposition on these cells, which in turn causes<br />

functional dysregulation <strong>of</strong> these cells and contributes to SLE<br />

pathogenesis. The current study was performed to characterize<br />

C4d deposition on T cells (T-C4d) and to investigate<br />

the role <strong>of</strong> ALA in T-C4d deposition. Briefly, T-C4d and ALA<br />

were determined by flow cytometry in 225 SLE patients, 135<br />

patients with other inflammatory diseases, and 15 healthy<br />

controls. Significantly elevated levels <strong>of</strong> C4d were detected<br />

on T cells <strong>of</strong> SLE patients (median fluorescence intensity<br />

(MFI)=14.8), compared with T cells from patients with other<br />

diseases (MFI=3.4; pb0.001) or healthy controls (MFI=0.8;<br />

pb0.001). IgM and/or IgG were detected concomitantly<br />

with C4d on T cells in a significant fraction <strong>of</strong> SLE patients<br />

(73/225), but not in healthy controls or patients with other<br />

diseases. Moreover, high levels <strong>of</strong> T-C4d could be induced on<br />

normal T cells after incubation with selected SLE plasma or<br />

immunoglobulins. Such in vitro T-C4d inducing activity could<br />

be eliminated by pre-absorption <strong>of</strong> SLE plasma or


S86 Abstracts<br />

immunoglobulins with lymphocytes, indicating that ALA is<br />

primarily responsible for triggering C4d deposition on Tcells.<br />

These results, together, support a previously unrecognized<br />

ALA-mediated pathogenic mechanism in SLE.<br />

doi:10.1016/j.clim.2007.422<br />

Sa.36 Treg Cells Directly Suppress Anti-DNA<br />

Ig-producing B Cells Through TGFβ-mediated<br />

Mechanisms in Murine Lupus<br />

Antonio La Cava, Associate Pr<strong>of</strong>essor, University <strong>of</strong><br />

California, Los Angeles, Los Angeles, CA, Chung Lee,<br />

Pr<strong>of</strong>essor, Northwestern University, Chicago, IL, Bevra<br />

Hahn, Pr<strong>of</strong>essor and Chief <strong>of</strong> Rheumatology, University <strong>of</strong><br />

California, Los Angeles, Los Angeles, CA<br />

In SLE, production <strong>of</strong> autoantibodies (autoAb) including<br />

anti-DNA Ig is central to pathogenesis <strong>of</strong> the disease. We<br />

reported that regulatory Foxp3+CD4+CD25+ T (Treg) cells in<br />

(NZB×NZW) F1 (NZB/W F1) lupus mice suppress the production<br />

<strong>of</strong> anti-DNA Ig by B cells in vitro. To discriminate between<br />

the possibility that Treg cells suppressed Tcells providing help<br />

to Ig-producing B cells and the possibility that Treg cells<br />

directly suppressed autoAb-producing B cells, we sublethally<br />

irradiated NZB/W F1 mice for adoptive transfer with<br />

syngeneic Treg cells together with B cells only or with B<br />

cells plus helper T cells. Additional experiments consisted <strong>of</strong><br />

transfer <strong>of</strong> these combinations but membrane expression <strong>of</strong><br />

TGFβ receptor II (TBR) in B cells was disrupted using a<br />

retrovirally based vector that expresses a dominant negative<br />

(DN) TBR. It was found that Treg suppression <strong>of</strong> anti-DNA Ig in<br />

vivo occurred without requirement <strong>of</strong> intermediate regulation<br />

<strong>of</strong> CD4+ Tcells; suppression <strong>of</strong> anti-DNA Ab was observed<br />

in the absence <strong>of</strong> CD4+ T cells (Pb0.03 vs. anti-DNA Ab from<br />

transfer <strong>of</strong> B cells only). Importantly, an intact TGFβ signaling<br />

pathway was required for B cell suppression by Treg.<br />

Disruption <strong>of</strong> B cell surface expression <strong>of</strong> TGFβ receptor via<br />

the dominant negative (DN) TBR permitted escape <strong>of</strong> B cells<br />

from the Treg-controlled suppression and led to dysregulated<br />

production <strong>of</strong> autoAb in vitro and in vivo. The findings<br />

identify events controlling production <strong>of</strong> autoAb in SLE and<br />

envision new possibilities for targeted intervention.<br />

doi:10.1016/j.clim.2007.03.424<br />

Sa.38 Agalactosyl Immunoglobulins are a Useful<br />

Marker for <strong>Clinical</strong> Progression and Successful<br />

Therapy <strong>of</strong> Collagen-induced Arthritis<br />

Katrien Van Beneden, PhD, Ghent University, Ghent,<br />

Belgium, Ken Coppieters, PhD Student, Ghent University,<br />

Ghent, Belgium, Wouter Laroy, PhD, Ghent University and<br />

Flanders Interuniversity Institute for Biotechnology,<br />

Zwijnaarde, Belgium, Pieter Rottiers, PhD, Ghent<br />

University and Flanders Interuniversity Institute for<br />

Biotechnology, Zwijnaarde, Belgium, Gust Verbruggen, MD<br />

PhD, Ghent University, Ghent, Belgium, Nico Callewaert,<br />

PhD, Ghent University and Flanders Interuniversity Institute<br />

for Biotechnology, Zwijnaarde, Belgium, Dirk Elewaut, MD<br />

PhD, Ghent University, Ghent, Belgium<br />

The purpose <strong>of</strong> this study was to evaluate the validity <strong>of</strong><br />

serum agalactosyl Ig as a biomarker for murine collagen-induced<br />

arthritis(CIA).Wesoughttoclarifythecorrelationbetween<br />

arthritic index and agalactosyl Ig fraction, the kinetics <strong>of</strong><br />

agalactosyl Ig levels upon development and progression <strong>of</strong> CIA,<br />

and the effect <strong>of</strong> antigen specific versus non-specific treatment.<br />

CIA was induced in susceptible mice, serum was collected from<br />

several arthritic and non-arthritic animals and agalactosyl Ig<br />

content was determined using a sensitive, improved technology<br />

for DNA sequencer aided analysis <strong>of</strong> N-linked glycans. Prophylactic<br />

tolerization with collagen type II was performed and the<br />

clinical effect was monitored; agalactosyl IgG was analyzed in<br />

serum derived from endpoint bleeding. Semi-therapeutic<br />

treatment with either dexamethasone or etanercept was<br />

performed and compared with PBS controls. All mice were<br />

sequentially bled at various time points during treatment.<br />

Agalactosyl Ig ratios were quantified and compared with agematched,<br />

unimmunized DBA/1 mice. Arthritic animals could be<br />

distinguished from non-arthritic animals with high sensitivity<br />

and specificity. Improvement <strong>of</strong> clinical arthritis scores by<br />

antigen specific tolerance induction correlated with downregulated<br />

levels <strong>of</strong> agalactosyl Ig. Finally, increased serum<br />

agalactosyl Ig levels correlate with CIA progression and are<br />

reversed by successful semi-therapeutic treatment. These<br />

findings indicate that agalactosyl determination may be a very<br />

useful serum marker for monitoring the clinical outcome <strong>of</strong><br />

experimental treatment in CIA, by use <strong>of</strong> a sensitive albeit<br />

simple methodology. This observation may be valuable for<br />

longitudinal CIA studies or for a more detailed judgment <strong>of</strong><br />

outcome upon treatment.<br />

doi:10.1016/j.clim.2007.03.426<br />

Sa.40 NKT Cells are Novel Accelerator and Producer<br />

<strong>of</strong> IL-17 in the Pathogenesis <strong>of</strong> Collagen-induced<br />

Arthritis<br />

Yohei Yoshiga, Graduate Student, Tsukuba University,<br />

Tsukuba, Japan, Goto Daisuke, MD, PhD, Tsukuba<br />

University, Tsukuba, Japan, Mizuko Mamura, MD, PhD,<br />

Tsukuba University, Tsukuba, Satoshi Ito, MD, PhD, Tsukuba<br />

University, Tsukuba, Japan, Isao Matsumoto, MD, PhD,<br />

Tsukuba University, Tsukuba, Japan, Akito Tsutsumi, MD,<br />

PhD, Tsukuba University, Tsukuba, Japan, Takayuki Sumida,<br />

MD, PhD, Tsukuba University, Tsukuba, Japan<br />

Objective: To clarify the role <strong>of</strong> NKTcells in CIA. Methods: (1)<br />

CIA was induced in C57BL/6 (B6) and NKT-KO (Jƒ¿281−/−)mice.<br />

Mice were immunized with type II collagen (CII) emulsified in<br />

CFA intradermally at day 0 and day 21, and then the incidence<br />

and severity <strong>of</strong> arthritis were evaluated. (2) To analyze the CIIspecific<br />

T cell response, splenocytes and draining lymph node<br />

(DLN) cells from B6 and NKT-KO mice immunized with CII/CFA<br />

were stimulated with CII. The culture supernatants were used<br />

for cytokine ELISA, and these cells were used for intracellular<br />

cytokine staining. (3) To identify the association <strong>of</strong> NKTcells and<br />

IL-17 production, splenocytes from naive B6 and NKT-KO mice<br />

were stimulated with ƒ¿-GalCer, ligand for NKT cells. The IL-17<br />

production was assayed by intracellular cytokine staining.<br />

Results: (1) The incidence <strong>of</strong> arthritis in NKT-KO mice was<br />

suppressed compared with that in B6 mice. The severity <strong>of</strong>


Abstracts<br />

arthritis in NKT-KO mice was also milder than that in B6 mice.<br />

(2) The production <strong>of</strong> IFN-ƒÁ and IL-4 was not significantly<br />

different between NKT-KO and B6 mice, but IL-17 production<br />

was reduced in NKT-KO mice. IL-17-producing T helper (Th17)<br />

cells were reduced in DLN cells from NKT-KO mice. (3) NKTcells<br />

produced IL-17 in B6 splenocytes stimulated with ƒ¿-GalCer,<br />

although there are no IL-17-producing cells in NKT-KO<br />

splenocytes. Conclusion: These findings support the notion<br />

that NKT cells are a novel producer and accelerator <strong>of</strong> IL-17,<br />

indicating NKTcells might play a crucial role in the pathogenesis<br />

<strong>of</strong> CIA via IL-17.<br />

doi:10.1016/j.clim.2007.03.427<br />

Sa.41 Endogenous CD4 T Cells Program B Cells to<br />

Respond in the Chronic GVH Model <strong>of</strong> SLE<br />

Arpita Choudhury, Post Doctoral Fellow, Division <strong>of</strong><br />

Rheumatology, School <strong>of</strong> Medicine, University <strong>of</strong><br />

Pennsylvania, Philadelphia, PA, Philip Cohen, Pr<strong>of</strong>essor,<br />

Division <strong>of</strong> Rheumatology, School <strong>of</strong> Medicine, University <strong>of</strong><br />

Pennsylvania, Philadelphia, PA, Robert Eisenberg, Pr<strong>of</strong>essor,<br />

Division <strong>of</strong> Rheumatology, School <strong>of</strong> Medicine, University <strong>of</strong><br />

Pennsylvania, Philadelphia, PA<br />

The murine chronic GVH (cGVH) model <strong>of</strong> SLE is induced by<br />

allo-recognition <strong>of</strong> foreign major histocompatibility complex<br />

(MHC) class II determinants. Our previous studies suggested that<br />

B cells in CD4KO mice have intrinsic defects that prevent them<br />

from responding to allo-help (JI 174, 7600, 2005). We have<br />

further explored the role <strong>of</strong> host CD4 T cells in this model by<br />

using post-irradiation auto-reconstitution and cell transfer<br />

experiments. C57Bl/6 (B6)-CD4KO mice were irradiated (5<br />

Gy), and 3–5 million CD4 Tcells from various B6 congenic strains<br />

were transferred. After allowing the B cells to auto-reconstitute<br />

over 3 weeks, 10 8 bm12 spleen cells were injected to initiate<br />

the cGVH. Purified CD4 T cells from IL-6KO, IFN- 3 KO and IL-10<br />

KO, but not IL-4 KO, mice were able to restore B cell response to<br />

cGVH in CD4 KO mice, signifying a critical role for IL-4.<br />

Treatment with recombinant IL-4 (rIL-4) also restored the<br />

ability <strong>of</strong> B cells from CD4 KO mice to secrete auto-abs in cGVH.<br />

Furthermore, the treatment <strong>of</strong> CD4KO recipients with an<br />

agonistic anti-CD40 (FGK-115, rat IgG2a) mAb also restored B<br />

cell function in cGVH. Our studies show that the endogenous<br />

CD4 T cell help needed for B cells to mount an effective cGVH<br />

response depends on IL-4, but not IL-6, IL-10 or IFN- 3 .Thus,<br />

autoreactive B cells need to have developed in a particular<br />

cytokine milieu in order to be receptive to alloreactive stimuli.<br />

Understanding the intracellular changes that condition B cells<br />

to become activated in cGVH may yield clues to their function<br />

in SLE-like autoimmunity.<br />

doi:10.1016/j.clim.2007.03.428<br />

Sa.42 The Mer Receptor Tyrosine Kinase Influences<br />

Toll-like Receptor Stimulation <strong>of</strong> Dendritic Cells<br />

Benjamin Scott, Postdoctoral Fellow, University <strong>of</strong><br />

Pennsylvania, Division <strong>of</strong> Rheumatology, Philadelphia, PA,<br />

Joudy-Ann Dinnall, Research Associate, Children’s Hospital,<br />

Department <strong>of</strong> Pediatrics, Philadelphia, PA, Stefania<br />

Gallucci, Assistant Pr<strong>of</strong>essor, Children’s Hospital <strong>of</strong><br />

Philadelphia, Department <strong>of</strong> Pediatrics, Philadelphia, PA,<br />

Philip Cohen, Pr<strong>of</strong>essor, University <strong>of</strong> Pennsylvania, Division<br />

<strong>of</strong> Rheumatology, Philadelphia, PA, Robert Eisenberg,<br />

Pr<strong>of</strong>essor, University <strong>of</strong> Pennsylvania, Division <strong>of</strong><br />

Rheumatology, Philadelphia, PA<br />

The mer receptor tyrosine kinase, which is expressed on<br />

macrophages and dendritic cells (DC), is known to promote<br />

apoptotic cell disposal and inhibit inflammatory cytokine<br />

production by macrophages, but its role in DCs is unclear.<br />

Mice lacking functional mer (mer kd mice) develop autoimmunity<br />

characterized by antibodies to DNA and chromatin,<br />

presumably due to their accumulation <strong>of</strong> apoptotic cells. To<br />

explore the role <strong>of</strong> mer on splenic DCs, mer kd and B6 mice<br />

were injected i.p. with toll-like receptor (TLR) ligands, and<br />

the mice were sacrificed 1 day later for analysis by flow<br />

cytometry. The numbers <strong>of</strong> splenic DCs and degree <strong>of</strong><br />

activation <strong>of</strong> splenic DCs in control mer kd mice are comparable<br />

to control B6 mice. However, following LPS (TLR-4<br />

ligand) injection, merkd splenic DCs displayed an abnormally<br />

activated pr<strong>of</strong>ile compared to B6 controls, characterized by<br />

the increased expression <strong>of</strong> the costimulatory molecules<br />

CD40, CD80, and CD86. This activated phenotype was highly<br />

pronounced in the plasmacytoid subset <strong>of</strong> DC (pDC) and was<br />

present to a lesser extent in the CD8α + and myeloid subsets.<br />

In contrast, after poly(I:C) (TLR-3 ligand) injection, mer kd DC<br />

appeared to have suppressed costimulatory molecule expression<br />

compared to B6 controls, whereas CpG-ODN (TLR-9<br />

ligand) stimulation <strong>of</strong> DC did not seem affected by the mer<br />

deficiency. Thus, mer may differentially regulate DC TLR 3<br />

and 4 signals and may negatively regulate pDC during<br />

inflammation. Further studies may provide important insight<br />

into SLE pathogenesis since pDC have been linked to lupus in<br />

humans and animal models.<br />

doi:10.1016/j.clim.2007.03.429<br />

S87<br />

Sa.43 Membranous Glomerulopathy Associated with<br />

Rheumatoid Arthritis may Respond to Rituximab<br />

Bryan J. Wolf, Resident Physician, NV, Medical Center/<br />

University <strong>of</strong> Nevada, Reno Department <strong>of</strong> Internal<br />

Medicine, NV, William M. O’Neil, <strong>Clinical</strong> Pr<strong>of</strong>essor,<br />

University <strong>of</strong> Nevada School <strong>of</strong> Medicine, V, John S. Pixley,<br />

Associate Pr<strong>of</strong>essor <strong>of</strong> Medicine, VA Medical Center/<br />

University <strong>of</strong> Nevada School <strong>of</strong> Medicine, NV<br />

Membranous glomerulonephritis is an uncommon extraarticular<br />

manifestation <strong>of</strong> rheumatoid arthritis, which need<br />

not be associated with prior DMARD (disease-modifying<br />

antirheumatic drug) therapies (Clin Rheum 1996, 15, 385).<br />

We observed a patient with an 8-year history <strong>of</strong> seropositive<br />

erosive rheumatoid arthritis complicated by the development<br />

<strong>of</strong> refractory nephrotic syndrome secondary to<br />

biopsy-proven membranous glomerulonephritis. Signs and<br />

symptoms <strong>of</strong> his disabling kidney disease included renal<br />

insufficiency, massive proteinuria (25 g), edema and a<br />

hypercoaguable state (deep venous thrombosis and pulmonary<br />

emboli). Alkylating agents, antimetabolites and


S88 Abstracts<br />

glucocorticoids were ineffective in reducing his proteinuria<br />

over a 2 year period. The patient subsequently received 2<br />

courses (1 g twice over a 2 week period) <strong>of</strong> rituximab (a<br />

monoclonal antibody to CD20 that specifically depletes B<br />

cells), 6 months apart. Serum albumin at initial treatment<br />

was 2.0 g/dl and normalized (3.7 g/dl) 1 month after the<br />

second course <strong>of</strong> therapy. This was accompanied by a<br />

corresponding decrease excretion <strong>of</strong> urinary protein (22 g<br />

to 7.4 g/24 h) and a reduction in diuretic requirements. B<br />

cell directed therapy may prove useful in treating membranous<br />

glomerulopathies.<br />

doi:10.1016/j.clim.2007.03.430<br />

Sa.44 Design and Elaboration <strong>of</strong> Truncated Mutants<br />

<strong>of</strong> the Autoantigen hSRP72, Present in<br />

Dermatomyositis, in Order to Identify the Sites <strong>of</strong><br />

Phosphorylation<br />

Victor Ermilo Arana-Argaez, Master in Science, <strong>Immunology</strong>,<br />

University <strong>of</strong> Guadalajara, Institute <strong>of</strong> Rheumatology,<br />

Guadalajara, Mexico, Victor Ermilo Arana-Argaez, Master in<br />

Science, University <strong>of</strong> Guadalajara, Guadalajara, Mexico,<br />

Beatriz Martin-Marquez, Master in Science, University <strong>of</strong><br />

Guadalajara, Guadalajara, Mexico, Muñoz-Valle Jose<br />

Francisco, PhD, PhD, Guadalajara, Mexico, Erika<br />

Martínez-Garcia, Master in Science, University <strong>of</strong><br />

Guadalajara, Institute <strong>of</strong> Rheumatology, Guadalajara,<br />

Mexico, Oscar Leon-Murguia, MD, Institute <strong>of</strong><br />

Rheumatology, Guadalajara, Mexico, Monica Vazquez-Del<br />

Mercado, PhD, MD, University <strong>of</strong> Guadalajara, Guadalajara,<br />

Mexico<br />

Dermatomyositis (DM) is an idiopathic inflammatory<br />

myopathy (IIM) with characteristic muscle-cutaneous findings.<br />

Using sera from patients with DM the SRP72<br />

component <strong>of</strong> the signal recognition particle (SRP) as an<br />

autoantigen phosphorylated on serine (S) residues was<br />

identified. The mechanisms to explain the lost <strong>of</strong> tolerance<br />

against the hSRP72 remain without explanation. To determine<br />

whether the phosphorylation state <strong>of</strong> this particle<br />

could affect the antigenicity, we cloned the autoantigen<br />

hSRP72 into a pRS314-TRP shuttle vector and we designed<br />

truncated mutants to identify the possible site/sites <strong>of</strong><br />

phosphorylation <strong>of</strong> the serine residues and its further<br />

implication on the break <strong>of</strong> tolerance against this protein.<br />

Aim: To truncate mutants <strong>of</strong> the hSRP72 to further identify<br />

the site or sites <strong>of</strong> phosphorylation for the application on<br />

the study <strong>of</strong> the induction mechanisms <strong>of</strong> autoantigenicity<br />

in DM. Material and methods: Based on the map <strong>of</strong> serine<br />

residues in the full length SRP72 sequence, we cloned the<br />

autoantigen hSRP72, designed and elaborated 13 truncated<br />

mutants to identify the region <strong>of</strong> the protein where<br />

phosphorylation might occur. We designed primers to<br />

amplify 13 mutants truncated by means <strong>of</strong> PCR using as a<br />

template the construct pRS314hSRP72. Next, in vitro<br />

translation and metabolic labeling with 35 S and 32P-g ATP<br />

will be done. Results and conclusions: We cloned successfully<br />

the autoantigen hSRP72 and the design and elaboration<br />

<strong>of</strong> truncated mutants according to the phosphorylation<br />

prediction map was correct based on its PCR product length<br />

for later use in in vitro translation phosphorylation tests<br />

and studies on antigenicity induction.<br />

doi:10.1016/j.clim.2007.03.431<br />

Sa.45 Activated Interstitial Macrophages are<br />

Important Mediators <strong>of</strong> SLE Nephritis<br />

Meera Ramanujam, Research Scientist, Feinstein Medical<br />

Research Institute, Manhasset, NY, Ramalingam<br />

Bethunaickan, Postdoctoral Fellow, Feinstein Medical<br />

Research Institute, Manhasset, NY, Anne Davidson,<br />

Pr<strong>of</strong>essor <strong>of</strong> Medicine, Feinstein Medical Research Center,<br />

Manhasset, NY<br />

SLE is an autoimmune disease that is complicated by<br />

autoantibody mediated glomerulonephritis. In NZB/W SLEprone<br />

mice recruitment <strong>of</strong> inflammatory cells and mediators<br />

is associated with early expression <strong>of</strong> a limited panel <strong>of</strong><br />

chemokines followed by spreading to involve multiple other<br />

inflammatory molecules. Furthermore, inflammatory cell<br />

subsets home to different areas <strong>of</strong> the NZB/W kidney. B<br />

cells, T cells and dendritic cells are found in disorganized<br />

aggregates in the perihilar region and around blood vessels,<br />

whereas macrophages invade the interstitium <strong>of</strong> the kidney<br />

and form a layer around the glomeruli. In NZW/BXSB mice,<br />

despite a similar expression pr<strong>of</strong>ile <strong>of</strong> inflammatory genes<br />

in the kidney, there is marked invasion <strong>of</strong> the interstitium<br />

by sheets <strong>of</strong> F4/80 macrophages whereas dendritic cells are<br />

found in large numbers only inside the glomeruli. NZM2410<br />

mice that develop a sclerotic form <strong>of</strong> glomerulonephritis<br />

express few inflammatory mediators in the kidney; the<br />

onset <strong>of</strong> glomerulonephritis is associated with invasion only<br />

by macrophages that are confined to the renal medulla.<br />

Both NZB/W and NZW/BXSB mice have large numbers <strong>of</strong><br />

circulating CD11bhi/Gr1lo monocytes; the kidneys <strong>of</strong> both<br />

these strains contain numerous F4/80+/Gr1lo/Ly6Clo/<br />

CD62L− macrophages. These cells appear concomitant<br />

with proteinuria onset and produce TNF-±, IL-1, BAFF and<br />

CXCL13; thus although phenotypically they resemble alternatively<br />

activated macrophages, they migrate to inflamed<br />

tissue and produce pro-inflammatory mediators. Further<br />

study <strong>of</strong> these cells should help us understand what directs<br />

traffic and local differentiation <strong>of</strong> monocytes to macrophages<br />

and dendritic cells, and how interactions between<br />

the differentiated macrophages and the kidney microenvironment<br />

promote injury.<br />

doi:10.1016/j.clim.2007.03.432<br />

Sa.46 Optimization <strong>of</strong> Autoantigen Microarrays to<br />

Study Autoimmunity<br />

Imelda Balboni, Postdoctoral Fellow, Stanford University,<br />

Medicine and Pediatrics, Stanford, CA, Cindy Limb,<br />

Research Assistant, Stanford University, Medicine, Stanford,<br />

CA, Paul Utz, Associate Pr<strong>of</strong>essor, Stanford University,<br />

Medicine, Stanford, CA, Jessica Tenenbaum, Graduate<br />

Student, Stanford University, Medicine, Stanford, CA


Abstracts<br />

Autoantigen microarrays are used to study autoimmune<br />

diseases including systemic lupus erythematosus, rheumatoid<br />

arthritis, and multiple sclerosis. Hundreds <strong>of</strong> autoantigens<br />

can be analyzed with microliter volumes <strong>of</strong><br />

serum in a high throughput manner. Various slide surfaces,<br />

printing methods and arraying conditions have been<br />

reported, and we sought to determine the optimal<br />

conditions for printing microarrays based on the initial<br />

protocol developed by the Robinson and Utz laboratories.<br />

We analyzed 22 slide surfaces for overall background,<br />

uniformity, streaking, and smearing <strong>of</strong> features. Based on<br />

these characteristics, coefficient <strong>of</strong> variance (CV) was<br />

determined for eight surfaces, and ultimately FAST®,<br />

poly-L-lysine, and SuperEpoxy slides were chosen for<br />

further studies. Several histidine-tagged autoantigens<br />

were spotted onto microarrays using a robotic microarrayer.<br />

Slides were probed with a histidine-specific<br />

monoclonal antibody and fluorophore-labeled secondary<br />

antibody. Arrays were scanned with a GenePix 4000<br />

scanner and analyzed using GenePix Pro 6.0 s<strong>of</strong>tware.<br />

CV was calculated based on the fluorescence intensity<br />

minus background for each feature. Similar studies using<br />

human sera were performed using the FAST® surface to<br />

demonstrate applicability to the study <strong>of</strong> human autoimmune<br />

disease. FAST® slides had the lowest CV in all<br />

studies, with interslide CV as low as 9% and intraslide CV<br />

as low as 15% when probing with a histidine-specific<br />

monoclonal antibody. CV was more variable when probing<br />

with human autoimmune sera, ranging from 5 to 36% with<br />

a median <strong>of</strong> 15%. Additional parameters for printing,<br />

probing, and analyzing microarrays were also investigated,<br />

and optimal conditions have been established to study<br />

autoantibody pr<strong>of</strong>iles in autoimmune disease.<br />

doi:10.1016/j.clim.2007.03.434<br />

Sa.47 Over-expression <strong>of</strong> Interferon Alpha in Lupus<br />

Families: Evidence for a Complex Heritable Trait<br />

Timothy Niewold, Fellow, Mary Kirkland Center for Lupus<br />

Research, Hospital for Special Surgery, New York, NY, John<br />

Harley, Pr<strong>of</strong>essor, Oklahoma Medical Research Foundation,<br />

Oklahoma City, OK, Jing Hua, Research Fellow, Mary<br />

Kirkland Center for Lupus Research, Hospital for Special<br />

Surgery, New York, NY, Mary Crow, Pr<strong>of</strong>essor, Mary Kirkland<br />

Center for Lupus Research, Hospital for Special Surgery,<br />

New York, NY, Thomas Lehman, Pr<strong>of</strong>essor, Mary Kirkland<br />

Center for Lupus Research, Hospital for Special Surgery,<br />

New York, NY<br />

Background: Interferon alpha (IFN-α) levels are elevated<br />

in many patients with systemic lupus erythematosus<br />

(SLE), however it is not known whether increased IFN-α is<br />

a cause or a result <strong>of</strong> SLE. A haplotype <strong>of</strong> the IFN-α<br />

pathway gene IRF5 has been associated with risk <strong>of</strong> SLE.<br />

Methods: Serum IFN-α activity was measured in samples<br />

from the Hospital for Special Surgery Family Lupus<br />

Registry (n=226, 111 SLE) and the Lupus Multiplex Registry<br />

and Repository (n=445, 155 SLE) using a reporter cell<br />

assay and normalized to unrelated healthy donor samples<br />

(n=106). The rs2004640 and rs2070197 SNPs in IRF5 were<br />

genotyped using ABI Taqman probes. Results: 47% <strong>of</strong> SLE<br />

patients had high serum IFN-a activity, and 18% <strong>of</strong> healthy<br />

relatives <strong>of</strong> SLE patients also had high IFN-α activity<br />

compared to unrelated donors (p=0.0002). High IFN-α<br />

expression is clustered in certain families. 54% <strong>of</strong> high<br />

IFN-α SLE patients have a healthy first degree relative<br />

with high IFN-α (p=2x10-7), and SLE-affected sib pairs had<br />

concordant IFN-α expression in most cases (p=0.023).<br />

There was a trend toward higher IFN-α expression in SLE<br />

patients with the IRF5 SLE risk haplotype. There were no<br />

ethnic differences in IFN-α expression, but the IRF5 risk<br />

haplotype was much more common in European American<br />

and Hispanic populations (p=0.0018). Conclusions: Many<br />

SLE families demonstrate abnormally high IFN-α expression<br />

in both healthy and affected individuals compared to<br />

healthy unrelated donors, suggesting that high endogenous<br />

IFN-α expression is a heritable SLE risk factor. It is<br />

likely that many genetic factors underlie this complex<br />

trait.<br />

doi:10.1016/j.clim.2007.03.435<br />

Sa.48 Evaluation <strong>of</strong> the Polymorphism <strong>of</strong> IL-1RN<br />

Gene in Patients with SLE (Systemic Lupus<br />

Erythematosus)<br />

Rashin Ganjali, Researcher, Immunogenetics Department,<br />

Bu-Ali Research Institute, Mashhad University <strong>of</strong> Medical<br />

Sciences, Mashhad, Iran, Jalil Tavakkol Afshari, Vice<br />

President for Research, Immunogenetics Department,<br />

Bu-Ali Research Institute, Mashhad University <strong>of</strong> Medical<br />

Sciences, Mashhad, Iran, Maryam Mazhani, Researcher,<br />

Immunogenetics Department, Bu-Ali Research Institute,<br />

Mashhad University <strong>of</strong> Medical Sciences, Mashhad, Iran<br />

Systemic lupus erythematosus (SLE) is an autoimmune<br />

disease with unknown etiology. SLE is characterized by<br />

malregulation <strong>of</strong> the immune system, with hyperactive B cells<br />

synthesizing excessive amounts <strong>of</strong> different autoantibodies.<br />

Enhanced IL-1 gene expression and IL-1 protein secretion are<br />

detected in the pathogenesis <strong>of</strong> SLE. IL-1RN has a downregulatory<br />

effect; therefore it is supposed that its gene<br />

polymorphisms may contribute to the pathogenesis <strong>of</strong> SLE. A<br />

total <strong>of</strong> 34 patients with SLE and 26 healthy subjects were<br />

studied. DNA was extracted from whole blood by salting out<br />

method and IL-1RN genotype was determined after PCR. The<br />

data were analyzed by Chi-square and Fisher’s exact tests.<br />

There were no statistical differences between genotype<br />

frequencies in both groups. Except A4 and A5 alleles, there<br />

were no statistically differences between allele frequencies,<br />

too; while A4 and A5 had been increased in healthy subjects<br />

(p=0.07). In the present population, polymorphic genotypes <strong>of</strong><br />

IL-1RN did not increase the susceptibility to SLE; while<br />

increased two polymorphic alleles in healthy subjects confirm<br />

this idea. However for/against relations between gene polymorphisms<br />

and such diseases require more studies with more<br />

individuals.<br />

doi:10.1016/j.clim.2007.03.436<br />

S89


S90 Abstracts<br />

Sa.49 TNF Receptor-associated Periodic Fever<br />

Syndrome (TRAPS) Caused by a Novel C30F Mutation<br />

Associated with the Common R92Q Low-Penetrance<br />

Mutation in TNFRSF1A<br />

Joao B. Oliveira, Associate Researcher, Molecular<br />

<strong>Immunology</strong> and Genetics Section, Laboratory <strong>of</strong> Medical<br />

Investigation #56, USP, São Paulo, Brazil, Adriana Jesus,<br />

Pediatric Rheumatology Second Year Fellow, Pediatrics<br />

Department, University <strong>of</strong> São Paulo, Brazil, São Paulo,<br />

Brazil, Ivona Aksentijevich, Staff Scientist, NIH/NIAMS/<br />

GGB, Bethesda, MD, Clovis Silva, Head <strong>of</strong> Pediatric<br />

Rheumatology Unit, Pediatrics Department, University <strong>of</strong><br />

São Paulo, Brazil, São Paulo, Brazil, Alberto J.S. Duarte,<br />

Lab Director, Laboratory <strong>of</strong> Medical Investigation #56<br />

(LIM_56), USP, São Paulo, Brazil<br />

Introduction: TRAPS is the most common <strong>of</strong> the autosomal<br />

dominant periodic fever syndromes. It is caused by<br />

mutations in the TNFRSF1A gene, which encodes for the<br />

type 1 TNF-receptor (TNFR1). We describe here a Brazilian<br />

patient with TRAPS and a novel TNFRSF1A mutation.<br />

Material and methods: The exons 2 to 5 and flanking<br />

intronic regions were amplified by PCR, purified and<br />

sequenced using an automated capillary sequencer. Results.<br />

The patient is a 9-year-old girl from São Paulo, Brazil. She<br />

presents recurrent fevers since age <strong>of</strong> 3 years, usually<br />

lasting 3 to 7 days, and recurring every other week. These<br />

episodes are associated with a mild abdominal pain, nausea,<br />

vomiting and generalized myalgia. Recurrent conjunctivitis<br />

and erysipela-like skin lesions in lower limbs are also<br />

present. Laboratory studies show persistent normocytic<br />

normochromic anemia, thrombocytosis, elevated erythrocyte<br />

sedimentation rate and C-reactive protein. Antinuclear<br />

antibody is positive (1:80). Physical examination is unremarkable,<br />

except for height in percentile 3 and weight in<br />

percentile 10–25. IgD level is within normal range. Genetic<br />

testing for Familial Mediterranean Fever (FMF) excluded five<br />

most common FMF mutations (M680I, M694V, M694I, V726A,<br />

E148Q). Mutational screening <strong>of</strong> TNFRSF1A revealed the<br />

novel C30F mutation and the common R92Q low-penetrance<br />

mutation. The R92Q is seen in 5% <strong>of</strong> the general population,<br />

and is associated with atypical inflammatory phenotypes.<br />

Conclusion: We report a TRAPS patient with the novel<br />

TNFRSF1 mutation, C30F, in conjunction with the low-penetrance<br />

mutation R92Q. Our findings expand the diversity <strong>of</strong><br />

TNFRSF1A mutations associated with TRAPS.<br />

doi:10.1016/j.clim.2007.03.437<br />

Sa.50 Cryopyrin-associated Periodic Syndromes<br />

(CAPS): Report <strong>of</strong> Two Brazilian Cases, Including a<br />

Rare Non-Exon 3 CIAS1 Mutation<br />

Adriana Jesus, Second Year Pediatric Rheumatology Fellow,<br />

Pediatrics Department, University <strong>of</strong> São Paulo, Brazil, São<br />

Paulo, Brazil, Brazil Joao Oliveira, Associate Researcher,<br />

Molecular <strong>Immunology</strong> and Genetics Section, Laboratory <strong>of</strong><br />

Medical Investigation #56, USP, São Paulo, Brazil, Ivona<br />

Aksentijevich, Staff Scientist, NIH/NIAMS/GGB, Bethesda,<br />

MD, Clovis Silva, Head <strong>of</strong> Pediatric Rheumatology Unit,<br />

Pediatrics Department, São Paulo, Brazil, Alberto J.S.<br />

Duarte, Lab Director, Laboratory <strong>of</strong> Medical Investigation<br />

#56 (LIM-56), USP, São Paulo, Brazil<br />

Introduction: Cryopyrin-associated periodic syndromes<br />

(CAPS) are autoinflammatory diseases that include three<br />

clinical syndromes linked to mutations in CIAS1 gene: familial<br />

cold autoinflammatory syndrome (FCAS), Muckle–Wells syndrome<br />

(MWS), and neonatal onset multisystem inflammatory<br />

disease (NOMID). We describe here 2 CAPS patients with the<br />

corresponding molecular defect. Material and methods: Exons<br />

and flanking intronic regions <strong>of</strong> CIAS1 were amplified by PCR,<br />

purified and sequenced using an automated capillary sequencer.<br />

Results: Patient 1 is an 8 year-old girl who presented at<br />

20 days <strong>of</strong> age with an erythematous and macular rash<br />

involving face, chest, abdomen, limbs, palms and soles. At<br />

3 months <strong>of</strong> age, she developed recurrent fever, exacerbated<br />

by cold weather. Elevated acute phase reactants were present.<br />

Arthritis <strong>of</strong> large joints and unilateral anterior uveitis developed<br />

during follow-up. DNA sequencing identified the T436I<br />

CIAS1 mutation. Patient 2 is a 7-year-old boy who soon after<br />

birth developed a papular exanthema involving chest, abdomen<br />

and extremities with daily fever episodes. Persistent<br />

anemia, leukocytosis, thrombocytosis, and high acute phase<br />

reactants were seen. Later, he presented with growth failure,<br />

frontal bossing and marked patellae enlargement. Cognitive<br />

and motor impairments and chronic intracranial hypertension<br />

were noted. Molecular studies identified a rare non-exon 3<br />

mutation, G755R, located in exon 4 <strong>of</strong> CIAS1. Conclusion: Two<br />

Brazilian patients with clinically and genetically related<br />

entities known as FCAS and NOMID were found to have<br />

disease-causing mutations in the CIAS1 gene, including one<br />

outside exon 3. Our result reinforces that CAPS patients should<br />

be screened for mutations in all exons <strong>of</strong> CIAS1.<br />

doi:10.1016/j.clim.2007.03.438<br />

Sa.51 Dickkopf-1 is a Link Between the Immune<br />

System and Bone Formation<br />

Georg Schett, Pr<strong>of</strong>essor, University <strong>of</strong> Erlangen-Nuremberg,<br />

Erlangen, Danielle Diarra, Fellow, Medical University <strong>of</strong><br />

Vienna, Vienna, Austria, William Richards, PhD, Amgen,<br />

Thousand Oaks, CA, Marina Stolina, PhD, Amgen, Thousand<br />

Oaks, CA<br />

Inflammatory joint disease can either lead to bone<br />

erosion or to bone formation. In rheumatoid arthritis,<br />

immune-mediated inflammation leads to joint destruction<br />

with virtually no signs <strong>of</strong> bone repair. In contrast, diseases<br />

such as ankylosing spondylitis are characterized by dramatic<br />

bone formation. The molecular signals determine whether<br />

inflammatory tissue induces bone loss or bone formation. By<br />

inhibiting Dickkopf-1 (DKK-1), a regulatory molecule <strong>of</strong> the<br />

Wnt pathway, we show that a bone destructive pattern <strong>of</strong><br />

arthritis can be reversed into a bone-forming pattern. Thus,<br />

treatment <strong>of</strong> tumor necrosis factor transgenic mice by an<br />

antibody against DKK-1 completely inhibited bone erosions<br />

and induced the formation <strong>of</strong> bony nodules called osteophytes.<br />

Tumor necrosis factor α (TNF) was identified as key<br />

inducer <strong>of</strong> DKK-1 in mouse inflammatory arthritis as well as


Abstracts<br />

human rheumatoid arthritis. This suggests that the Wnt<br />

pathway is a key regulator <strong>of</strong> joint remodeling and a link<br />

between the immune system and bone formation.<br />

doi:10.1016/j.clim.2007.03.439<br />

Sa.52 Biosimulations Predict that Rituximab is More<br />

Efficacious than Other Standards <strong>of</strong> Care in<br />

Rheumatoid Arthritis Patients with Severe Disease<br />

Kosmas Kretsos, Engineer, Entelos, Inc., Foster City, CA,<br />

Daniel L. Young, Engineer, Entelos Inc., Foster City, CA,<br />

Chan C. Whiting, Scientist, Entelos, Inc., Foster City, CA,<br />

Katherine Kudrycki, Scientist, Entelos, Inc., Foster City, CA,<br />

Sirid-Aimee Kellermann, Senior Scientist, Entelos, Inc.,<br />

Foster City, CA, Nadine A. Derfanoux, Senior Scientist,<br />

Entelos, Inc., Foster City, CA<br />

The clinical efficacy <strong>of</strong> the B cell-depleting therapy<br />

rituximab in patients with rheumatoid arthritis (RA) highlights<br />

the critical contribution <strong>of</strong> B cells to the disease<br />

process. To explore the efficacy <strong>of</strong> rituximab in RA patients<br />

with different phenotypes, the Entelos® RA PhysioLab®<br />

platform, a mathematical model representing an inflamed<br />

joint in an RA patient, was used to simulate the effects <strong>of</strong><br />

rituximab and other therapeutic approaches on key clinical<br />

outcomes. The platform dynamically integrates the contributions<br />

<strong>of</strong> multiple cell types and mediators involved in synovial<br />

hyperplasia and joint structural damage. B cell development,<br />

recruitment, activation and effector processes (e.g., antigen<br />

presentation, cytokine synthesis and autoantibody production)<br />

are also represented in the platform. Biosimulation in a<br />

virtual patient with severe RA predicts an ACR response<br />

almost identical (within a 95% confidence interval) to that<br />

reported in rituximab clinical trials. Furthermore, the<br />

predicted level <strong>of</strong> B cell depletion in the virtual patient is<br />

consistent with published reports. These results suggest that<br />

rituximab will be a highly effective single-agent treatment<br />

for severe RA. However, similar analyses in a virtual patient<br />

with moderate RA show that rituximab is less effective than<br />

anti-TNF-α therapies. In both phenotypes, biosimulations<br />

predict that rituximab induces sustained benefits in joint<br />

structure, namely decreases in cartilage degradation and<br />

bone erosion, which persist for months after cessation <strong>of</strong><br />

rituximab treatment, even after joint inflammation returns.<br />

Findings such as these can be used to help develop more<br />

targeted RA therapies and improve drug discovery by<br />

identifying prospective responder patient phenotypes.<br />

doi:10.1016/j.clim.2007.03.440<br />

Sa.53 Gene-environment Interactions Between<br />

Heavy Cigarette Smoking and HLA-DRB1 Shared<br />

Epitope in Predicting Incident RA, Data from a Two<br />

Large Prospective Cohort Studies<br />

Elizabeth W. Karlson, Associate Pr<strong>of</strong>essor <strong>of</strong> Medicine,<br />

Brigham and Women’s Hospital, Department <strong>of</strong> Medicine,<br />

Dover, MA, Shun-Chiao Chang, Doctoral Student, Harvard<br />

School <strong>of</strong> Public Health, Epidemiology, Boston, MA, Karen H.<br />

Costenbader, Assistant Pr<strong>of</strong>essor <strong>of</strong> Medicine, Brigham and<br />

Women’s Hospital, Department <strong>of</strong> Medicine, Boston, MA,<br />

Lori B. Chibnik, Statistician, Brigham and Women’s Hospital,<br />

Medicine, Boston, MA, Patricia Fraser, Medical Director,<br />

Pharmacovigilance/Medical Affairs, Cambridge, MA<br />

Background: Heavy smoking (N10 pack years) increases<br />

risk <strong>of</strong> RA. HLA-DRB1 demonstrates gene–environment<br />

interaction (GEI) with smoking when comparing current to<br />

never smokers. We studied this GEI in the Nurses’ Health<br />

Studies. Methods: RA diagnosis was validated by chart review<br />

for ACR criteria. Controls were matched on age, menopausal<br />

status, and postmenopausal hormone use. High resolution<br />

HLA-DRB1 genotyping was performed for SE alleles. HLA-SE,<br />

smoking, and HLA-SE×smoking interaction, and risk <strong>of</strong> RA<br />

were assessed using conditional logistic regression models,<br />

adjusted for age and reproductive factors. We tested for<br />

multiplicative and additive interaction. Results: 244 Caucasian<br />

matched pairs were included. Mean age at diagnosis was<br />

54 years; 59% <strong>of</strong> cases were RF+. There was no interaction<br />

between never/ever smoking and HLA-SE and risk <strong>of</strong> RA. We<br />

found no multiplicative interaction (p=0.10) but a strong<br />

additive interaction (p=0.006, attributable proportion (AP)<br />

0.49) with smoking categorized as b10 pack years vs. N10<br />

pack years. The highest risk was in heavy smokers with<br />

double copy HLA-SE (OR, 21.5; 95% CI, 2.8–167.9). There was<br />

evidence for additive interaction in RF+ (p=0.01, AP 0.47)<br />

and RF− cases (p=0.04, AP 0.53), each compared with<br />

controls. Models using pack years smoked as a continuous<br />

variable had no evidence for multiplicative interaction<br />

(p=0.33). Conclusion: GEI between HLA-SE and smoking in<br />

RA risk is strongest with N10 pack years smoked. Future<br />

studies should assess pack years smoked when testing for<br />

GEI. Additive interaction suggests a pathway that requires<br />

involvement <strong>of</strong> both risk factors.<br />

doi:10.1016/j.clim.2007.03.441<br />

S91<br />

Sa.54 Anti-fibrinbogen Autoimmunity in<br />

Rheumatoid Arthritis<br />

William Robinson, Assistant Pr<strong>of</strong>essor, Division <strong>of</strong><br />

<strong>Immunology</strong> and Rheumatology, Stanford University, Palo<br />

Alto, CA, Wolfgang Hueber, Research Associate, Division <strong>of</strong><br />

<strong>Immunology</strong> and Rheumatology, Stanford University, Palo<br />

Alto, CA, Peggy P. Ho, Research Associate, Department<br />

Neurology, Stanford University, Stanford, CA, Xiaoyan Zhao,<br />

Postdoctoral Fellow, Division <strong>of</strong> <strong>Immunology</strong> and<br />

Rheumatology, Stanford University, Palo Alto, CA<br />

Rheumatoid arthritis (RA) is an autoimmune synovitis<br />

affecting 0.5% <strong>of</strong> the world population, yet the mechanisms<br />

underlying the initiation and perpetuation <strong>of</strong> RA remain<br />

elusive. A subset <strong>of</strong> RA patients exhibit anti-citrulline<br />

autoimmunity and citrullinated proteins are generated<br />

through post-transnational conversion <strong>of</strong> arginine to citrulline<br />

by peptidyl-arginine deiminase. The pathogenesis <strong>of</strong> RA<br />

involves the deposition and excessive local generation <strong>of</strong><br />

fibrin in the inflamed joint, and recent studies identified<br />

citrullinated fibrin as a candidate autoantigen in RA. We<br />

performed direct immunoprecipitation <strong>of</strong> immune


S92 Abstracts<br />

complexes from RA and control synovial fluid and tissue<br />

lysates, followed by mass spectrometry (MS) analysis. MS/<br />

MS analysis revealed previously described and novel<br />

candidate autoantigens, including native and citrullinated<br />

forms <strong>of</strong> fibrinogen, fibronectin, HSPs, vimentin, and<br />

elongation factor. We added peptides and proteins representing<br />

these candidates to synovial antigen arrays to<br />

screen samples derived from RA and control patient<br />

cohorts. Synovial mircroarray and ELISA analyses demonstrated<br />

specific autoreactive B cell targeting <strong>of</strong> citrullinated<br />

fibrinogen peptides and proteins in approximately<br />

30% <strong>of</strong> RA patients. Anti-citrullinated fibrinogen antibodies<br />

were present almost exclusively in patients possessing anti-<br />

CCP antibodies. We also detect fibrinogen-containing<br />

immune complexes in blood derived from patients possessing<br />

anti-fibrinogen autoantibodies. Based on these results<br />

we developed a fibrinogen-induced arthritis (FIA) mouse<br />

model using human fibrinogen as the immunizing antigen<br />

(see abstract presented by Ho et al.). These data support<br />

our hypothesis that citrullinated fibrinogen is a pathogenic<br />

autoantigen in RA, and that autoimmunity against fibrinogen<br />

defines a molecular subtype representing approximately<br />

30% <strong>of</strong> RA patients.<br />

doi:10.1016/j.clim.2007.03.442<br />

Sa.55 Genome-Wide SNP Scan Identifies<br />

Polymorphisms Associated with Differential<br />

Response to Anti-TNF Treatment Among<br />

Rheumatoid Arthritis Patients in the ABCoN Cohort<br />

John Carulli, Principal Scientist, BiogenIdec, Cambridge,<br />

MA, Franak Batliwalla, Assistant Investigator, Feinstein<br />

Institute for Medical Research, Manhasset, NY, Chunyu Liu,<br />

Scientist, BiogenIdec, Drug Discovery, Cambridge, MA, Aarti<br />

Damle, Assistant Investigator, Feinstein Institute for<br />

Medical Research, Manhasset, NY, Jadwiga Bienkowska,<br />

Principal Scientist, Biogenidec, Drug Discovery, Cambridge,<br />

MA, Annette Lee, Assistant Investigator, Feinstein Institute<br />

for Medical Research, Manhasset, NY, Wentian Li, Assistant<br />

Investigator, Feinstein Institute for Medical Research,<br />

Manhasset, NY, Peter Gregersen, Investigator, Feinstein<br />

Institute for Medical Research, Manhasset, NY<br />

The Autoimmune Biomarkers Collaborative Network<br />

(ABCoN) has enrolled a longitudinal cohort <strong>of</strong> RA patients<br />

beginning anti-TNF treatment in order to identify biomarkers<br />

influencing response to anti-TNF therapy. Genomewide<br />

(Illumina 317K) SNP data were collected on 116 <strong>of</strong><br />

these patients (54 etaneracept, 25 adalimumab, 37<br />

infliximab). DAS28 scores for these 116 patients ranged<br />

from 2.72 to 7.08 (mean =5.23) prior to starting anti-TNF<br />

therapy, decreasing to 1.4 to 6.68 (mean=3.77) at 14 weeks<br />

after enrollment. SNP genotypes were tested for association<br />

with the change in DAS28 score at 6 weeks and<br />

14 weeks after initiation <strong>of</strong> anti-TNF therapy. While no<br />

individual SNPs show genome-wide significance (pb10 −7 )in<br />

this small data set, preliminary analysis shows 36 SNPs<br />

associated (pb10 −4 ) with DDAS28 at 6 weeks, and 41 SNPs<br />

associated with DDAS28 at 14 weeks. Nine regions <strong>of</strong> the<br />

genome have two or more SNPs within 100 kb with pb10 −5 ,<br />

and genes in these regions represent candidates for anti-<br />

TNF pharmacogenetic markers. Permutation tests on these<br />

data indicate a low type I error rate. Haplotype analysis<br />

may reveal additional evidence for association. RNA<br />

samples (PaxGene) from peripheral blood <strong>of</strong> the same<br />

patients were analyzed by genome-wide transcript pr<strong>of</strong>iling.<br />

Cross-referencing <strong>of</strong> genes comprising expression-based<br />

classifiers for anti-TNF responders and non-responders (see<br />

FOCIS 2007 abstract by Batliwalla et al.) with genes showing<br />

nominal genetic association with anti-TNF response represents<br />

a powerful approach to identifying pharmacogenomic<br />

markers for anti-TNF response among rheumatoid arthritis<br />

patients.<br />

doi:10.1016/j.clim.2007.03.443<br />

Sa.56 Oncogenic Properties <strong>of</strong> Rheumatoid Synovial<br />

Cells<br />

Toshihiro Nakajima, Pr<strong>of</strong>essor, Institute <strong>of</strong> Medical Science,<br />

St. Marianna University School <strong>of</strong> Medicine, Kawasaki,<br />

Japan, Naoko Yagishita, Senior Assistant Pr<strong>of</strong>essor, Institute<br />

<strong>of</strong> Medical Science, St. Marianna University School <strong>of</strong><br />

Medicine, Kawasaki, Japan, Satoshi Yamasaki, Senior<br />

Assistant Pr<strong>of</strong>essor, Institute <strong>of</strong> Medical Science, St.<br />

Marianna University School <strong>of</strong> Medicine, Kawasaki, Japan,<br />

Kusuki Nishioka, Pr<strong>of</strong>essor, I, Kawasaki, Japan<br />

Rheumatoid arthritis (RA) is one <strong>of</strong> the most common<br />

and critical articular diseases with synovial hyperplasia.<br />

However, the mechanism that regulates synovial cell outgrowth<br />

is not fully understood. To clarify the mechanism,<br />

we carried out immunoscreening using anti-rheumatoid<br />

synovial cell antibody and cloned a putative RING finger<br />

protein, designated Synoviolin (G&D, 2003). Synoviolin is an<br />

E3 ubiquitin ligase and located in endoreticulum. It was<br />

highly expressed in the rheumatoid synovium, and mice<br />

overexpressing this molecule developed spontaneous<br />

arthropathy. In this regard, like rheumatoid synovial cells,<br />

the anti-apoptotic effect <strong>of</strong> Synoviolin was observed even<br />

for its expressed ectopically in NIH3T3 cells, which resulted<br />

in enhanced cell overgrowth in these cells (MCB, 2005).<br />

These results were confirmed also in fly system. Next, to<br />

identify the substrates for Synoviolin that may be involved<br />

in cell growth, we analyzed the comprehensive pr<strong>of</strong>ile <strong>of</strong><br />

protein expression in Synoviolin null cells (JBC, 2005).<br />

Then, we found that Synoviolin targets tumor suppressor<br />

gene p53 for ubiquitination (EMBO J., 2007). Synoviolin<br />

sequestrated and metabolized p53 in the cytoplasm and<br />

negatively regulated its cellular level and biological<br />

functions, including transcription, cell cycle regulation<br />

and apoptosis. Furthermore, these p53 regulatory functions<br />

<strong>of</strong> Synoviolin were irrelevant to other E3 ubiquitin ligases<br />

for p53, such as MDM2, Pirh2 and Cop1, which form<br />

autoregulatory feedback loops. Our results provide novel<br />

insights into p53 signaling mediated by Synoviolin and<br />

clarify the molecular basis <strong>of</strong> synovial hyperplasia in RA.<br />

doi:10.1016/j.clim.2007.03.444


Abstracts<br />

Sa.57 T Lymphocyte Clonal Alterations in<br />

Anti-Citrullinated Protein Antibody Positive<br />

Synovitis<br />

Tineke Cantaert, PhD Student, University <strong>of</strong> Amsterdam,<br />

Amsterdam, The Netherlands, Sophie Brouard, PhD, INSERM<br />

U643, Nantes, France, Cristophe Braud, PhD, INSERM U643,<br />

Nantes, France, Jean-Paul Soulillou, Pr<strong>of</strong>essor, INSERM<br />

U643, Nantes, France, Niek de Vries, PhD, <strong>Clinical</strong><br />

<strong>Immunology</strong> and Rheumatology, Academic Medical Center,<br />

University <strong>of</strong> Amsterdam, Amsterdam, The Netherlands,<br />

Paul-Peter Tak, Pr<strong>of</strong>essor, <strong>Clinical</strong> <strong>Immunology</strong> and<br />

Rheumatology, Academic Medical Center, University <strong>of</strong><br />

Amsterdam, Amsterdam, The Netherlands, Dominique<br />

Baeten, Pr<strong>of</strong>essor, <strong>Clinical</strong> <strong>Immunology</strong> and Rheumatology,<br />

Academic Medical Center/University <strong>of</strong> Amsterdam,<br />

Amsterdam, The Netherlands<br />

Objective: Increasing evidence suggests that the pathophysiology<br />

<strong>of</strong> rheumatoid arthritis (RA) is fundamentally<br />

different in patients with or without anti-citrullinated<br />

protein antibodies (ACPA). The association <strong>of</strong> RA with HLA-<br />

DR shared epitope exclusively in the ACPA+ subset supports<br />

this concept and suggests that T cell help may be involved in<br />

the ACPA response. We assessed the potential involvement<br />

<strong>of</strong> T lymphocytes in ACPA+ synovitis. Methods: Synovial<br />

biopsies were obtained from inflamed knee joints <strong>of</strong> 54 RA<br />

patients. 37 were ACPA+ (anti-CCP2 ELISA). mRNA was<br />

isolated from 7 ACPA+ RA synovia, 4 ACPA− RA synovia, and<br />

10 control spondyloarthritis (SpA) synovia with similar levels<br />

<strong>of</strong> T and B cell infiltration. T lymphocyte clonality in the<br />

synovium was studied by combined quantitative and<br />

qualitative TCR CDR3 analysis by the TcLandscape technology.<br />

Results: Comparing ACPA+ with the ACPA− RA patients,<br />

there were no significant differences in clinical and<br />

histological parameters. Alterations <strong>of</strong> the normal TCR<br />

length distribution were analyzed for all Vbeta families.<br />

TCR analysis showed a significantly higher degree <strong>of</strong><br />

alteration <strong>of</strong> the TCR length distribution in ACPA+ (31.2±<br />

3.7%) compared to ACPA− (18.5±1.4%; p=0.012) and to SpA<br />

synovitis (22.4 ±1.3%; p=0.014). These alterations reflecting<br />

oligo- or monoclonal expansions were observed in several<br />

Vbeta families and were different in each individual sample.<br />

Conclusion: We demonstrate an increased alteration <strong>of</strong> the<br />

TCR length distribution in ACPA+ but not in ACPA− RA or<br />

control synovitis. These data suggest a specific contribution<br />

<strong>of</strong> T lymphocytes in the local disease process and possibly<br />

the ACPA response in this RA subset.<br />

doi:10.1016/j.clim.2007.03.445<br />

Sa.58 Synovial T/B Cell Lymphoid Aggregates<br />

Regulate the Production <strong>of</strong> Rheumatoid<br />

Arthritis-specific Autoantibodies<br />

Tineke Cantaert, PhD Student, University <strong>of</strong> Amsterdam,<br />

Amsterdam, The Netherlands, Trieneke Timmer, PhD<br />

Student, Molecular Cell Biology and <strong>Immunology</strong>, VU<br />

Medical Center, Amsterdam, The Netherlands, Bernard<br />

Vandooren, PhD Student, Rheumatology, Ghent University<br />

Hospital, Ghent, Belgium, Carla Wijbrandts, PhD Student,<br />

<strong>Clinical</strong> <strong>Immunology</strong> and Rheumatology, Academic Medical<br />

Center/University <strong>of</strong> Amsterdam, Amsterdam, The<br />

Netherlands, Rogier Thurlings, PhD Student, <strong>Clinical</strong><br />

<strong>Immunology</strong> and Rheumatology, Academic Medical Center/<br />

University <strong>of</strong> Amsterdam, Amsterdam, The Netherlands,<br />

Leen De Rycke, PhD, <strong>Clinical</strong> <strong>Immunology</strong> and<br />

Rheumatology, Academic Medical Center, University <strong>of</strong><br />

Amsterdam, Amsterdam, The Netherlands, Tineke van der<br />

Pouw-Kraan, PhD, Molecular Cell Biology and <strong>Immunology</strong>,<br />

VU Medical Center, Amsterdam, The Netherlands, Theo Out,<br />

PhD, Experimental <strong>Immunology</strong>, Academic Medical Center/<br />

University <strong>of</strong> Amsterdam, The Netherlands, Paul-Peter Tak,<br />

Pr<strong>of</strong>essor, <strong>Clinical</strong> <strong>Immunology</strong> and Rheumatology, Academic<br />

Medical Center, University <strong>of</strong> Amsterdam, Amsterdam, The<br />

Netherlands, Dominique Baeten, Pr<strong>of</strong>essor, <strong>Clinical</strong><br />

<strong>Immunology</strong> and Rheumatology, Academic Medical Center,<br />

University <strong>of</strong> Amsterdam, Amsterdam, The Netherlands<br />

Ectopic lymphoid neogenesis occurs in synovial membrane<br />

in rheumatoid arthritis (RA) and inflamed tissues.<br />

Partial structural resemblance with follicles in secondary<br />

lymphoid organs suggests a role in the maturation <strong>of</strong><br />

humoral immune and autoimmune responses. We investigated<br />

the relation between synovial T/B cell lymphoid<br />

aggregates, inflammation, and autoantibody production.<br />

Synovial biopsies were obtained from inflamed joint <strong>of</strong> 130<br />

patients with RA or spondyloarthritis (SpA), a chronic<br />

arthritis devoid <strong>of</strong> known autoantibodies. B–T cell aggregates<br />

were analyzed by immunohistochemistry and realtime<br />

PCR. Autoantibodies were measured in synovial fluid by<br />

ELISA, corrected for total IgG and IgM. The presence <strong>of</strong> large<br />

lymphoid aggregates was observed in 1/3 <strong>of</strong> the rheumatoid<br />

synovia, but also in SpA. This was associated with synovial<br />

inflammation in both diseases, with infiltrating memory B<br />

lymphocytes observed almost exclusively within these<br />

aggregates. TNFα blockade induced the disintegration <strong>of</strong><br />

the follicles (pb0.05). Levels <strong>of</strong> CXCL13 and CCL19 were<br />

elevated in follicular versus diffuse synovitis (pb0.05). We<br />

could not observe a dark and light zone nor detect naive B<br />

cells by CD23/IgD staining. These structures occasionally<br />

displayed germinal centres characterized by high endothelial<br />

venules, T/B cell segregation, and follicular dendritic<br />

cells (b10%). This was confirmed by the low amount <strong>of</strong><br />

CD21L mRNA. Both rheumatoid factor (p=0.033) and anticitrullinated<br />

protein antibodies (p=0.09) were lower in<br />

follicular versus diffuse RA synovitis.<br />

Ectopic lymphoid neogenesis is an inducible and reversible<br />

phenomenon in chronic synovitis. Germinal centre reactions<br />

can occasionally be observed in synovial follicles, which<br />

regulate rather than support local humoral autoimmunity in<br />

RA.<br />

doi:10.1016/j.clim.2007.03.447<br />

S93<br />

Sa.59 Non-Pathogenic Antinuclear Antibodies<br />

Contribute to the Clearance <strong>of</strong> Apoptotic Antigens<br />

Released During TNFα Blockade in Spondyloarthritis<br />

(SpA)<br />

Tineke Cantaert, PhD Student, University <strong>of</strong> Amsterdam,<br />

Amsterdam, The Netherlands, Leen De Rycke, PhD, <strong>Clinical</strong>


S94 Abstracts<br />

<strong>Immunology</strong> and Rheumatology, Academic Medical Center/<br />

University <strong>of</strong> Amsterdam, Amsterdam, The Netherlands,<br />

Bernard Vandooren, PhD Student, Rheumatology, Ghent<br />

University Hospital, Ghent, Belgium, Carla Wijbrandts, PhD<br />

Student, <strong>Clinical</strong> <strong>Immunology</strong> and Rheumatology, Academic<br />

Medical Center, University <strong>of</strong> Amsterdam, Amsterdam, The<br />

Netherlands, Elli Kruith<strong>of</strong>, PhD, Rheumatology, Ghent<br />

University Hospital, Ghent, Belgium, Filip De Keyser,<br />

Pr<strong>of</strong>essor, Rheumatology, Ghent University Hospital, Ghent,<br />

Belgium, Eric M. Veys, Pr<strong>of</strong>essor, Rheumatology, Ghent<br />

University Hospital, Ghent, Belgium, Paul-Peter Tak,<br />

Pr<strong>of</strong>essor, <strong>Clinical</strong> <strong>Immunology</strong> and Rheumatology,<br />

Academic Medical Center, University <strong>of</strong> Amsterdam,<br />

Amsterdam, The Netherlands, Dominique Baeten, Pr<strong>of</strong>essor,<br />

<strong>Clinical</strong> <strong>Immunology</strong> and Rheumatology, Academic Medical<br />

Center, University <strong>of</strong> Amsterdam, Amsterdam, The<br />

Netherlands<br />

Infliximab, not etanercept, treatment in arthritis induces<br />

non-pathogenic, IgM antinuclear antibodies. In SLE, antinuclear<br />

antibodies have been related to increased BAFF and<br />

type I-interferon signaling and increased presence <strong>of</strong><br />

apoptotic blebs. We assessed the mechanisms leading to<br />

autoantibody induction by TNFα blockade in spondyloarthritis<br />

(SpA), a disease without humoral autoimmunity.<br />

Serum and synovial biopsies were obtained from SpA<br />

patients treated with infliximab (n=20) or etanercept<br />

(n=20). Serum levels <strong>of</strong> BAFF, IFNα, nucleosomes, and<br />

anti-nucleosomes were assessed (ELISA). Synovial apoptosis<br />

was assessed by TUNEL and active caspase3. Nucleosome<br />

levels increased at 2 (+89%; p =0.002) and 6 (+26%;<br />

p=0.014) weeks <strong>of</strong> infliximab but not etanercept treatment.<br />

This was not related to cell death in the synovium at<br />

weeks 1 and 12 <strong>of</strong> treatment. Infliximab induced an<br />

upregulation <strong>of</strong> IFNα at 2 (p=0.090) and 6 (p=0.042)<br />

weeks. BAFF levels remained stable during treatment with<br />

both biologicals. Anti-nucleosome antibodies were<br />

increased at week 12 (+94%; pb0.001) and 34 (+124%;<br />

p b0.001) <strong>of</strong> infliximab treatment. This increase was<br />

restricted to IgM isotype and correlated with the rise <strong>of</strong><br />

nucleosomes at week 12 (r =0.383;p =0.008) and 34<br />

(r =0.453; p=0.014). The continuous requirement <strong>of</strong> nuclear<br />

antigens for the autoantibody response was indicated by the<br />

disappearance <strong>of</strong> the antibodies upon interruption <strong>of</strong> anti-<br />

TNFα. However, nucleosome levels dropped to normal upon<br />

appearance <strong>of</strong> the autoantibodies. Infliximab induces nonpathogenic<br />

IgM antinuclear antibodies by upregulation <strong>of</strong><br />

serum nucleosomes, suggesting induction <strong>of</strong> apoptosis in<br />

vivo, and the associated increase in type I IFN. This antibody<br />

response could represent a physiological mechanism which<br />

contributes to the normal clearance <strong>of</strong> nuclear antigens. TC<br />

and LDR contributed equally.<br />

doi:10.1016/j.clim.2007.03.448<br />

Sa.61 Proteomic Signatures <strong>of</strong> Secreted Proteins for<br />

the Prediction <strong>of</strong> Treatment Response to TNF<br />

Blockers in RA<br />

Wolfgang Hueber, Postdoctoral Fellow, Stanford University,<br />

Medicine, Palo Alto, CA, Beren Tomooka, Research<br />

Assistant, Stanford University, Medicine, Palo Alto, CA,<br />

Franak Batliwalla, Research Scientist, North Shore Long<br />

Island Jewish Health Care, Manhasset, NY, Robert<br />

Tibshirani, Pr<strong>of</strong>essor, Stanford University, Stanford, CA,<br />

Peter Gregersen, Pr<strong>of</strong>essor, North Shore Long Island Jewish<br />

Health Care System, Manhasset, NY, Lars Klareskog,<br />

Pr<strong>of</strong>essor, Karolinska Institutet, Stockholm, Sweden,<br />

William Robinson, Pr<strong>of</strong>essor, Stanford University, Medicine,<br />

Palo Alto, CA<br />

We pr<strong>of</strong>iled autoantibodies and cytokines in pre-treatment<br />

and 3-month post-treatment sera from RA patients to<br />

delineate secreted proteins predictive <strong>of</strong> response to<br />

therapy with etanercept. Subgroups from two cohorts <strong>of</strong><br />

Enbrel-treated RA patients were analyzed: 29 patients<br />

enrolled in the ABCoN cohort, and 28 patients treated at a<br />

Swedish tertiary care hospital. Serum autoantibodies were<br />

determined using synovial antigen microarrays, and 14<br />

cytokines were measured using a bead-based multiplex<br />

assay. Serum was collected before and 3 months after<br />

initiation <strong>of</strong> therapy. Differential expression <strong>of</strong> autoantibodies<br />

and cytokines in serum from responders and nonresponders<br />

was analyzed. Prediction analysis <strong>of</strong> microarrays<br />

(PAM) was applied to identify panels <strong>of</strong> secreted molecules<br />

predictive <strong>of</strong> response. Pre-treatment samples from etanercept<br />

responders with ACR 50 or greater response but not<br />

non-responders demonstrated higher reactivity against<br />

peptides derived from RA candidate autoantigens including<br />

fibrinogen, calpastatin, heat shock proteins, proteoglycans,<br />

and histones (FDRb0.1). The cytokines IL-6, IL12p40 and<br />

Eotaxin showed a trend towards higher mean levels in pretreatment<br />

samples (pb0.1) in the Swedish but not in the<br />

ABCoN cohort. Pair-wise comparisons <strong>of</strong> pre-treatment with<br />

post-treatment pr<strong>of</strong>iles demonstrated that decreases in<br />

certain autoantibody reactivities and the cytokine IL-6 were<br />

associated with ACR 50 or greater responses. PAM identified<br />

a panel <strong>of</strong> secreted molecules that correctly predicted 73%<br />

<strong>of</strong> patients who subsequently responded to etanercept<br />

therapy with ACR 50 responses or greater. Our data suggest<br />

that pr<strong>of</strong>iles <strong>of</strong> secreted molecules in pre-treatment sera<br />

are associated with RA patients more likely to respond to<br />

etanercept.<br />

doi:10.1016/j.clim.2007.03.449<br />

Sa.62 Distinct Molecular and Cellular Aspects <strong>of</strong><br />

Systemic Juvenile Idiopathic Arthritis (SJIA) and<br />

Polyarticular (PolyJIA)<br />

Claudia Macaubas, Research Associate, Stanford University<br />

Department <strong>of</strong> Pediatrics, Stanford, CA, Khoa Nguyen, BS,<br />

Stanford University Department <strong>of</strong> Pediatrics, Stanford, CA,<br />

Kuang-Hung Pan, PhD Student, Stanford University<br />

Department <strong>of</strong> Genetics, Stanford, CA, Tzielan Lee, <strong>Clinical</strong><br />

Assistant Pr<strong>of</strong>essor, Stanford University Department <strong>of</strong><br />

Pediatrics, Stanford, CA, Chetan Deshpande, Lab Manager,<br />

Stanford University Department <strong>of</strong> Pediatrics, Stanford, CA,<br />

Christy Sandborg, Pr<strong>of</strong>essor and Chief, Stanford<br />

University Department <strong>of</strong> Pediatrics, Stanford, CA,<br />

Stanley Cohen, Pr<strong>of</strong>essor, Stanford University Department<br />

<strong>of</strong> Genetics, Stanford, CA, Elizabeth Mellins, Associate


Abstracts<br />

Pr<strong>of</strong>essor, Stanford University Department <strong>of</strong> Pediatrics,<br />

Stanford, CA<br />

Juvenile idiopathic arthritis (JIA) is a family <strong>of</strong> chronic<br />

inflammatory arthritides that includes distinct subtypes:<br />

systemic (SJIA), polyarticular (polyJIA) and pauciarticular.<br />

The subtypes differ in epidemiology, genetic risk factors,<br />

clinical features, prognosis and response to therapy.<br />

However, the specific molecular mechanisms that distinguish<br />

these diseases are not well understood. We performed<br />

a comparative analysis <strong>of</strong> molecular and cellular aspects <strong>of</strong><br />

SJIA and polyJIA. Blood samples and associated clinical<br />

information were obtained from children with SJIA (n=24),<br />

polyJIA (n=17), and age-matched children without immunological<br />

health problems (n =14). Gene expression in<br />

freshly isolated PBMCs was studied by quantitative PCR<br />

(qPCR) <strong>of</strong> 81 immune related genes. Cell types were<br />

enumerated by FACS. Using GABRIEL analysis s<strong>of</strong>tware, we<br />

identified genes whose expression correlates with erythrocyte<br />

sedimentation rate (ESR). In SJIA, ESR is positively<br />

correlated with transcripts expressed by activated monocytes,<br />

whereas in polyJIA, ESR correlates with transcripts<br />

for cytolytic proteins and Th1-related genes. Expression <strong>of</strong><br />

distinct genes correlates with number <strong>of</strong> active joints in<br />

SJIA and polyJIA, including IL2R and MIF vs. IL-12,<br />

respectively. At the cellular level, we observed expansion<br />

<strong>of</strong> monocytes and altered monocyte subset distribution<br />

(CD14hi vs. CD16hi) in SJIA, but not polyJIA subjects,<br />

compared to controls. Samples from poly JIA subjects<br />

showed a reduced percentage <strong>of</strong> NK cells, and lower<br />

percentage <strong>of</strong> T regulatory cells. Our findings argue that<br />

SJIA is associated with dysregulation <strong>of</strong> innate cells,<br />

whereas poly JIA is associated with altered adaptive<br />

immunity.<br />

doi:10.1016/j.clim.2007.03.450<br />

Sa.63 The Alternative Pathway is Necessary and<br />

Sufficient for Complement Involvement in the<br />

Effector Phase <strong>of</strong> Anti-Collagen Antibody Induced<br />

Arthritis (ACAIA)<br />

Nirmal Banda, Associate Pr<strong>of</strong>essor, University <strong>of</strong> Colorado at<br />

Denver Health Sciences Center, Aurora, CO, Allyson Wood,<br />

Research Associate, University <strong>of</strong> Colorado at Denver Health<br />

Sciences Center, Aurora, CO, Kazue Takahashi, Assistant<br />

Pr<strong>of</strong>essor, Pediatrics, Massachusetts General Hospital,<br />

Boston, MA, William P. Arend, Pr<strong>of</strong>essor <strong>of</strong> Medicine,<br />

University <strong>of</strong> Colorado at Denver Health Sciences Center,<br />

Aurora, CO, Alan Ezekowitz, Pr<strong>of</strong>essor, Pediatrics,<br />

Massachusetts General Hospital, Boston, MA, V. Michael<br />

Holers, Pr<strong>of</strong>essor <strong>of</strong> Medicine and <strong>Immunology</strong>, University <strong>of</strong><br />

Colorado at Denver and Health Sciences Center, Aurora, CO<br />

We reported that ACAIA is unchanged in classical/lectin<br />

pathway C4-deficient, mice but 90% decreased in alternative<br />

pathway factor-B-deficient mice (J. Immunol. 2006;<br />

177: 1904). We have further examined the role <strong>of</strong><br />

complement pathways in ACAIA using C57BL/6 mice<br />

genetically deficient in components C3 (C3−/−) or C1q<br />

(C1q−/−), deficient in both mannose-binding lectins A and<br />

C (MBL−/−), and doubly deficient in both C1q and MBL-A/C<br />

(C1q−/−/MBL−/−), with only the alternative pathway<br />

intact. Arthritis was induced by a mixture <strong>of</strong> 4 anti-type II<br />

collagen monoclonal antibodies, with an IP injection at day<br />

3 <strong>of</strong> LPS to cycle arthritis development. Mice were scored<br />

daily for arthritis in the paws using a scale <strong>of</strong> 0–3<br />

(maximum score <strong>of</strong> 12 per mouse) and sacrificed on day<br />

10. There were no significant differences in the clinical<br />

disease activity scores (CDAS) between C1q−/− mice and<br />

wild type (WT, normal C57BL/6 mice), MBL−/− and WT<br />

mice, or C1q−/−/MBL−/− and WT mice. In contrast, similar<br />

to factor B-deficient mice, CDAS in C3−/− mice were 66%<br />

decreased in comparison to WT mice (3.75 ±1.03 and 11.75<br />

±0.25, mean±SEM, n=4, respectively). The histological<br />

scores for inflammation, pannus formation, bone damage<br />

and cartilage damage, as well as immunohistochemical<br />

scores for the deposition <strong>of</strong> C3, were all consistent with the<br />

CDAS. These results indicate that the alternative pathway<br />

<strong>of</strong> complement is unique in being both necessary and<br />

sufficient for complement involvement in the effector<br />

phase <strong>of</strong> ACAIA. Potential mechanism(s) underlying this key<br />

role are under investigation.<br />

doi:10.1016/j.clim.2007.03.451<br />

S95<br />

Sa.64 Anti-Inflammatory Effect <strong>of</strong> Modified-<br />

Extracellular Matrix Proteins (MECMP) in CIA<br />

Perla Macip-Rodríguez, Physician, Instituto Nacional de<br />

Ciencias Medicas y Nutricion SZ, Department <strong>of</strong><br />

<strong>Immunology</strong> and Rheumatology, Mexico, Janette Furuzawa-<br />

Carballeda, Chemistry, Instituto Nacional de Ciencias<br />

Medicas y Nutricion SZ, Department <strong>of</strong> <strong>Immunology</strong> and<br />

Rheumatology, Mexico, Ma. Inés Vargas-Rojas, MD, Instituto<br />

Nacional de Ciencias Medicas y Nutricion SZ, Department <strong>of</strong><br />

<strong>Immunology</strong> and Rheumatology, Mexico, Virginia<br />

Soto-Abraham, Physician, Instituto Nacional de Cardiología<br />

ICh. Department <strong>of</strong> Pathology, Mexico, David Cruz-Robles,<br />

Biologist, Instituto Nacional de Cardiología ICh,<br />

Department <strong>of</strong> Pathology, Mexico, Leonardo Limón-<br />

Camacho, Physician, Instituto Nacional de Ciencias Medicas<br />

y Nutricion SZ, Department <strong>of</strong> <strong>Immunology</strong> and<br />

Rheumatology, Mexico<br />

The aim <strong>of</strong> this study was to evaluate the effect <strong>of</strong><br />

intradermal MECMP administration on a type II collagen<br />

(CII)-induced arthritis (CIA). Age-matched mice were<br />

immunized intradermally at the base <strong>of</strong> the tail with 100<br />

μg <strong>of</strong> chicken CII emulsified in complete Freund’s adjuvant.<br />

Mice were then boosted with 100 μg <strong>of</strong> CII in incomplete<br />

Freund’s adjuvant at 21 days. Mice were treated 1 week<br />

after boost with 100 μl <strong>of</strong> (a) placebo, (b) matrikines<br />

(hydrolyzed-collagen and elastin), (c) collagen-PVP, (d) b+c<br />

and (e) methotrexate (2.5 mg/kg) and every week during<br />

1 month. <strong>Clinical</strong> scores were assessed immediately before<br />

immunization (day 0) and thereafter weekly. Inflammation<br />

<strong>of</strong> the four paws was cored as follows: 0, no inflammation;<br />

1, swelling or redness <strong>of</strong> one joint; 2, swelling or redness <strong>of</strong><br />

more than one joint or mild inflammation <strong>of</strong> the whole<br />

paw; 3, severe inflammation <strong>of</strong> whole paw or ankylosis. The<br />

incidence <strong>of</strong> CIA was 100% by day 28 in CII challenged mice.


S96 Abstracts<br />

Weight and temperature were also determined. Macroscopical<br />

analysis showed a down-regulation <strong>of</strong> inflammation<br />

post administration <strong>of</strong> MECMP and methotrexate treatments.<br />

The histological analysis showed that CIA-mice<br />

group had an extensive bone erosion, pannus and severe<br />

focal inflammatory infiltrates. MECMP mice-treated group<br />

ameliorated the clinical signs. Bone erosion and inflammation<br />

were moderate compared to CIA-mice group. Concluding,<br />

MECMP exerts a down-regulation <strong>of</strong> inflammation on<br />

established inflammation and is able to ameliorate the<br />

tissue damage associated with CIA.<br />

doi:10.1016/j.clim.2007.03.452<br />

Sa.65 Established Murine Collagen-induced Arthritis<br />

and IL-17 Production are Specifically Inhibited by a<br />

Single Inoculation <strong>of</strong> Lipopolysaccharide-stimulated<br />

Dendritic Cells<br />

Juan C. Aguillon, Biochemist, Universidad de Chile,<br />

Santiago, Chile, Lorena Salazar, Biochemist, Universidad<br />

de Chile, <strong>Immunology</strong> Program, Santiago, Chile, Octavio<br />

Aravena, Biochemist, Universidad de Chile, <strong>Immunology</strong><br />

Program, Santiago, Chile, Carlos Gonzalez, DVM,<br />

Universidad de Chile, Facultad de Ciencias Veterinarias,<br />

Santiago, Chile, Paula Abello, DVM, Universidad de Chile,<br />

<strong>Immunology</strong> Program, Santiago, Juan Contreras-Levicoy,<br />

MD, Universidad de Chile, <strong>Immunology</strong> Program, Santiago,<br />

Chile, Barbara Pesce, Biochemist, Universidad de Chile,<br />

<strong>Immunology</strong> Program, Santiago, Chile, Flavi Salazar-<br />

Onfray, Biologist, Universidad de Chile, <strong>Immunology</strong><br />

Program, Santiago, Chile, Miguel Cuchacovich, MD,<br />

Hospital Clinico Universidad de Chile, Santiago, Chile<br />

Dendritic cells (DCs), crucial in immune responses, are<br />

also involved in peripheral tolerance induction. TNFmodulated<br />

DCs have demonstrated to restore tolerance<br />

in experimental multiple sclerosis and collagen induced<br />

arthritis (CIA). We investigated the capacity <strong>of</strong> short-term<br />

lipopolysaccharide (LPS)-stimulated DCs pulsed with type II<br />

collagen (CII) to induce tolerance against established CIA.<br />

Bone marrow-derived DCs were generated in the presence<br />

<strong>of</strong> GM-CSF. After CIA induction with CII, mice were<br />

injected at day 35 with a single dose <strong>of</strong> immature DCs, 4<br />

or 24 h LPS-stimulated DCs that had been loaded with CII<br />

(CII/DCs, 4 h LPS/CII/DCs, or 24 h LPS/CII/DCs). Arthritis<br />

progression was monitored by clinical and histologic<br />

evaluations. Flow cytometry <strong>of</strong> 4 h LPS/CII/DCs showed<br />

intermediate CD40 and CD86 expression, lower than that<br />

<strong>of</strong> 24 h LPS/CII/DCs (fully mature), and higher than that <strong>of</strong><br />

CII/DCs (immature). A functional assay showed that 4 h<br />

LPS/CII/DCs display increased endocytosis ability with<br />

respect to 24 h LPS/CII/DCs, indicating a semi-mature<br />

state. The single inoculation <strong>of</strong> 4 h LPS/CII/DCs in mice<br />

with established CIA reduced significantly disease severity<br />

at day 70. Histologic evaluation <strong>of</strong> mice treated with 4 h<br />

LPS/CII/DCs revealed diminished inflammatory synovitis,<br />

cartilage damage, and fibrosis. Unlike CII-stimulated<br />

splenocytes from CIA control mice, those from mice<br />

inoculated with 4 h LPS/CII/DCs showed diminished IL-17<br />

and increased IL-10 levels in culture supernatants. The<br />

high IL-10 production is consistent with the induction <strong>of</strong><br />

TR1 regulatory T cells responsible for inhibiting to effector<br />

IL-17-producing CD4+ T cells. Conclusion: Short-term LPSmodulated<br />

DCs inoculation interferes with CIA progression<br />

and IL-17 production when loaded with CII. Supported by<br />

Fondecyt-1070553, Millennium Nucleus-P04/030-F and<br />

Mecesup-UCH 0115.<br />

doi:10.1016/j.clim.2007.03.453<br />

Sa.66 Kinetics <strong>of</strong> Immune Tolerization in a <strong>Clinical</strong><br />

Trial on Epitope-specific Therapy in Rheumatoid<br />

Arthritis (RA)<br />

Eva K<strong>of</strong>feman, University <strong>of</strong> California San Diego,<br />

Department <strong>of</strong> Medicine and Pediatrics, Utrecht, The<br />

Netherlands, Diane Amox, University <strong>of</strong> California San<br />

Diego, Department <strong>of</strong> Medicine and Pediatrics, La Jolla, CA,<br />

Adriana Tremoulet, University <strong>of</strong> California San Diego,<br />

Department <strong>of</strong> Medicine and Pediatrics, La Jolla, CA, Elissa<br />

Keogh, University <strong>of</strong> California San Diego, Department <strong>of</strong><br />

Medicine and Pediatrics, La Jolla, CA, Peter Zeiseniss,<br />

University <strong>of</strong> California San Diego, Department <strong>of</strong> Medicine<br />

and Pediatrics, La Jolla, CA, Courtney Gosch, University <strong>of</strong><br />

California San Diego, Department <strong>of</strong> Medicine and<br />

Pediatrics, La Jolla, CA, Samantha Friend, University <strong>of</strong><br />

California San Diego, Department <strong>of</strong> Medicine and<br />

Pediatrics, La Jolla, CA, Xiao Zhang, University <strong>of</strong> Alabama<br />

at Birmingham, School <strong>of</strong> Public Health Department <strong>of</strong><br />

Biostatistics, Birmingham, AL, Gary Cutter, University <strong>of</strong><br />

Alabama at Birmingham, School <strong>of</strong> Public Health,<br />

Department <strong>of</strong> Biostatistics, Birmingham, AL, Salvatore<br />

Albani, Pr<strong>of</strong>essor, University <strong>of</strong> California San Diego,<br />

Department <strong>of</strong> Medicine and Pediatrics, La Jolla, CA<br />

Background: In a phase II trial on mucosal tolerization<br />

in 160 RA-patients, dnaJp1 (E. coli heat shock protein<br />

dnaJ-peptide) 25 mg 1dd for 168 days was safe and<br />

clinically effective. Objectives: (i) Study immunological<br />

kinetics underlying clinical response. (ii) Identify biological<br />

markers predicting clinical response. Methods: PBMCs<br />

collected on days 0, 112 and 168 were cultured with<br />

dnaJP1, Tetanus Toxoid and controls and FACS-stained for<br />

CD3 and intracellular cytokines. <strong>Clinical</strong> response is ≥1<br />

ACR20 (American College <strong>of</strong> Rheumatology) response<br />

between day 112 and 1-month follow-up. Results: High<br />

CD69 expression upon dnaJp1 in vitro on day 0 associated<br />

with later clinical amelioration (ACR20 112–168 p=0.06,<br />

ACR50 p=0.02). TNFa production to dnaJp1 decreased<br />

before day 112 (0–112 pb0.0001) in the dnaJp1 group,<br />

remaining low on day 168. In the clinical responders<br />

subgroup, trends were seen <strong>of</strong> an increase in IL10 (0–168<br />

p=0.09) and initial decrease (0–112 p=0.17) and late<br />

increase in IFNg (112–168 p=0.15). IFNg increase was<br />

associated with the IL10 increase in dnaJP1 patients<br />

(p=0.10). Reactions to Tetanus Toxoid and reactions in<br />

the placebo group did not change. Discussion: CD69<br />

expression to dnaJp1 in vitro may be a predictor <strong>of</strong><br />

clinical effect. This suggests that antigen-specific T cells<br />

play an important role in mucosal tolerization. Cytokine<br />

data suggest that tolerization started with deviation <strong>of</strong>


Abstracts<br />

antigen-specific T-effectors (TNFa decrease IFNg decrease,<br />

IL10 increase), followed by induction <strong>of</strong> regulatory T cell<br />

mechanisms (IL10+/IFNg+ Tr1) and clinical amelioration.<br />

doi:10.1016/j.clim.2007.03.454<br />

Sa.67 Nasal Co-Administration <strong>of</strong> ISS Increases the<br />

Arthritis-Protective Effect <strong>of</strong> an HSP60-derived<br />

Peptide<br />

Evelien Zonneveld-Huijssoon, University Medical Center<br />

Utrecht, Pediatric <strong>Immunology</strong>, Utrecht, The Netherlands,<br />

Femke van Wijk, University Medical Center Utrecht,<br />

Pediatric <strong>Immunology</strong>, Utrecht, The Netherlands, Wilco de<br />

Jager, Mr, University Medical Center Utrecht, Pediatric<br />

<strong>Immunology</strong>, Utrecht, The Netherlands, Sarah Roord,<br />

University Medical Center Utrecht, Pediatric <strong>Immunology</strong>,<br />

Utrecht, The Netherlands, Salvatore Albani, University <strong>of</strong><br />

California San Diego, Medicine and Pediatrics, La Jolla, CA,<br />

Wietse Kuis, University Medical Center Utrecht, Pediatric<br />

<strong>Immunology</strong>, Utrecht, The Netherlands, Berent Prakken,<br />

University Medical Center Utrecht, Pediatric <strong>Immunology</strong>,<br />

Utrecht, The Netherlands<br />

Peptide-based immunotherapy is a promising way to treat<br />

autoimmune diseases. In earlier studies we identified T cell<br />

epitopes derived from human and mycobacterial HSP60 that<br />

are recognized by a majority <strong>of</strong> patients with rheumatoid or<br />

juvenile idiopathic arthritis. Nasal administration <strong>of</strong> one <strong>of</strong><br />

these epitopes (p1) partially prevented adjuvant arthritis in<br />

the rat. To further enhance this arthritis-protective effect we<br />

tested two types <strong>of</strong> ISS and peptidoglycan (PGN) as adjuvants.<br />

Rats were treated with three nasal doses <strong>of</strong> p1, adjuvant or a<br />

combination <strong>of</strong> p1 and adjuvant prior to arthritis induction<br />

with CFA. Sixty days after arthritis induction, mandibular and<br />

inguinal lymph nodes and spleens were harvested. We<br />

determined phenotype, proliferative responses and cytokine<br />

production <strong>of</strong> fresh and in vitro stimulated cells. P1+ISS cotreatment<br />

enhanced the arthritis-protective effect <strong>of</strong> p1. ISS<br />

treatment alone did not have any effect on arthritis scores,<br />

nor did P1+PGN co-treatment or PGN alone. P1+ISS cotreatment<br />

led to increased p1-induced IFNg production by<br />

spleen cells. Co-treatment with one <strong>of</strong> the tested ISS led to<br />

increased IL-10 production as well. No clear differences could<br />

be detected between treatment groups in percentage <strong>of</strong> CD4<br />

+IL10+IFNg+ Tcells or CD4+CD25+Foxp3+ Tcells. <strong>Clinical</strong> data<br />

so far imply that ISS may be a suitable adjuvant for peptidespecific<br />

immune therapy in arthritis. Future plans include<br />

adoptive transfer <strong>of</strong> p1+ISS treated spleen cells and a further<br />

analysis <strong>of</strong> the induced p1 specific T cells and dendritic cells.<br />

doi:10.1016/j.clim.2007.03.455<br />

Sa.68 Abrogation <strong>of</strong> Autoimmune-mediated<br />

Inflammation by the Natural Plant Product,<br />

Honokiol via GABA(A)-mediated Alteration <strong>of</strong><br />

CD40 and LMP1 Signaling in B Cells<br />

Melissa E. Munroe, Postdoctoral Research Fellow, University<br />

<strong>of</strong> Iowa Department <strong>of</strong> Microbiology, Iowa City, IA, Gail A.<br />

Bishop, Pr<strong>of</strong>essor, University <strong>of</strong> Iowa Department <strong>of</strong> Internal<br />

Medicine and Microbiology and the VAMC, Iowa City, IA<br />

Our lab has extensively studied signaling in B cells by<br />

CD40 and its viral mimic, LMP1, which signals in an<br />

amplified and sustained manner, and has been implicated<br />

in pathogenesis <strong>of</strong> lymphoma and autoimmune disease.<br />

Honokiol (HNK), a phenolic compound isolated and purified<br />

from magnolia, has been found to have a number <strong>of</strong><br />

pharmacologic benefits, including anti-inflammatory properties,<br />

without the toxicity found in vivo with current<br />

treatments. HNK stabilized already established inflammatory<br />

arthritis, with a decrease in TNF-α, IL-6, and collagenspecific,<br />

pro-inflammatory mediated, antibodies. We evaluated<br />

potential molecular mechanisms leading to the antiinflammatory<br />

effects <strong>of</strong> HNK using B cell lines expressing<br />

either hCD40 or the hCD40-LMP1 chimeric receptor. CD40<br />

and LMP1 mediated NFκB and AP-1 pathways were<br />

abrogated, but not eliminated by HNK, with a dosedependent<br />

decrease in nuclear translocation <strong>of</strong> NFκB1,<br />

NFκB2, and AP-1 associated proteins. Furthermore, the<br />

anti-inflammatory effects <strong>of</strong> HNK could be reversed using<br />

inhibitors <strong>of</strong> the neurotransmitter GABA(A), a previously<br />

reported target for HNK interaction. We are currently<br />

investigating potential downstream mediators <strong>of</strong> GABA<br />

activation which may affect CD40 and LMP1 signaling,<br />

including kinases and proteosome function. These findings<br />

are particularly exciting and suggest that the nontoxic antiinflammatory<br />

properties <strong>of</strong> HNK could be a means for<br />

blocking the autoimmune response.<br />

doi:10.1016/j.clim.2007.03.456<br />

S97<br />

Sa.69 Reversal <strong>of</strong> CD4:CD8 Ratio in Pediatric<br />

Patients with Macrophage Activation Syndrome/<br />

Reactive Hemophagocytic Lymphohistiocytosis<br />

(MAS/HLH): A Potential Marker for Active Disease<br />

Process<br />

Paivi Miettunen, Doctor, Division <strong>of</strong> Pediatric Rheumatology<br />

and University <strong>of</strong> Calgary, Calgary, Alberta, AB, Canada<br />

Background: MAS/HLH in pediatric rheumatology refers<br />

to excessive activation and proliferation <strong>of</strong> mature macrophages<br />

leading to an overwhelming inflammatory reaction.<br />

While abnormal T cell activation (TCA) has been postulated<br />

to be at the center <strong>of</strong> this process, the details <strong>of</strong> the<br />

immunological mechanisms that initiate and maintain the<br />

abnormal macrophage functions are unclear. Objective: To<br />

test the hypothesis that alterations in CD4:CD8 ratios<br />

would provide an early marker for abnormal TCA in MAS/<br />

HLH. Methods: All patients met Histiocytosis Society’s 2004<br />

criteria for HLH. Peripheral blood was collected at the time<br />

<strong>of</strong> diagnosis and at remission (n=7, 4 F; 3 M: ages 4 to 16<br />

years). Blood from age and sex matched healthy children<br />

was used as controls. Peripheral blood CD4:CD8 ratios were<br />

analyzed by flow cytometry. Results: Significant reversal <strong>of</strong><br />

CD4:CD8 ratio was seen in four <strong>of</strong> seven patients with<br />

active MAS/HLH. An infectious process was ruled out<br />

(negative HIV, EBV and CMV serology). At the time <strong>of</strong>


S98 Abstracts<br />

remission CD4:CD8 ratio had normalized. In one patient<br />

with reactivation <strong>of</strong> MAS/HLH, a reversal <strong>of</strong> the CD4:CD8<br />

ratio recurred, and normalized with cyclosporin, corticosteroids<br />

and interleukin-1 antagonist, anakinra treatment.<br />

Conclusions: Our preliminary studies indicate that reversal<br />

<strong>of</strong> CD4:CD8 can be an important marker for active MAS/<br />

HLH in a significant proportion <strong>of</strong> patients, supporting role<br />

<strong>of</strong> abnormal T cell activation. CD4:CD8 ratio can also be<br />

used to follow treatment response in selected patients.<br />

Studies are in progress to identify additional immunological<br />

markers to diagnose this condition accurately and to<br />

delineate the potential mechanisms involved in this<br />

process.<br />

doi:10.1016/j.clim.2007.03.457<br />

Sa.70 Quantitative Biomarker Analysis <strong>of</strong> Anti-CCP,<br />

Rheumatoid Factor (RF) and Immunoglobulin Levels<br />

in Synovial Biopsies Indicate Enrichment and Local<br />

Production in Rheumatoid Arthritis (RA)<br />

Sanna Rosengren, Associate Project Scientist, University <strong>of</strong><br />

California, San Diego, Medicine, La Jolla, CA, Nathan Wei,<br />

<strong>Clinical</strong> Director, Arthritis and Osteoporosis Center,<br />

Frederick, MD, David Boyle, Associate Research<br />

Immunologist, University <strong>of</strong> California, San Diego,<br />

Medicine, La Jolla, CA, Nathan Zvaifler, Pr<strong>of</strong>essor Emeritus,<br />

University <strong>of</strong> California, San Diego, Medicine, La Jolla, CA<br />

Circulating autoantibodies such as RF and anti-CCP are<br />

associated with RA. B cells and plasma cells accumulate in RA<br />

synovium, yet their contribution to autoantibody production<br />

is unclear. We developed ELISA-based methods to quantify<br />

autoantibodies and total Ig (immunoglobulin) G and IgM in<br />

extracts <strong>of</strong> RA and osteoarthritis synovial tissues prepared<br />

using 1% Brij-35 in PBS. Synovial tissue and matched serum<br />

were obtained at arthroplasty or arthroscopic biopsy from RA<br />

(n=11) and OA (n=6) patients. RF-IgM, anti-CCP and antitetanus<br />

IgG, total IgM and IgG, and albumin were determined<br />

by ELISA in synovial extracts and serum. Specific activity was<br />

calculated as the ratio <strong>of</strong> synovial to serum antibody/<br />

albumin. IgG1 and IgM heavy constant mRNA levels were<br />

determined by qPCR as a measure <strong>of</strong> synovial Ig synthesis.<br />

Interestingly, anti-CCP IgG, but not RF-IgM, was significantly<br />

enriched in RA synovia compared to serum. OA synovial<br />

extracts contained no detectable autoantibodies. Total IgG<br />

specific activity was significantly higher in RA synovia than in<br />

OA (median (range): RA 2.58 (1.22–2.99); OA 0.76 (0.73–<br />

0.87), p=0.0077). Similar results were obtained for total IgM<br />

and tetanus IgG. Notably, mRNA content and specific activity<br />

correlated significantly for both IgM and IgG. Synovia<br />

containing lymphocyte aggregates (n=3) contained significantly<br />

higher total IgG but not total IgM. In conclusion,<br />

correlations between immunoglobulin message and protein<br />

indicate that local synthesis contributes to synovial Ig<br />

content. Reliable determinations <strong>of</strong> autoantibodies, antitetanus<br />

IgG, and total IgM and IgG in synovial biopsy extracts<br />

will be <strong>of</strong> value in pro<strong>of</strong>-<strong>of</strong>-concept clinical trials.<br />

doi:10.1016/j.clim.2007.03.458<br />

Sa.71 Thrombin-activatable Carboxypeptidase B<br />

Prevents Anti-Collagen Antibody Induced Arthritis<br />

Jason Song, Rheumatology Fellow, Department <strong>of</strong> Medicine<br />

Division <strong>of</strong> Rheumatology Stanford University, Palo Alto,<br />

CA, Toshihiko Nishimura, Life Science Research Assistant,<br />

Department <strong>of</strong> Medicine, Division <strong>of</strong> Pulmonary and Critical<br />

Care Medicine, Stanford University, Menlo Park, CA, Michael<br />

Garcia, Undergraduate Student, Department <strong>of</strong> Medicine<br />

Division <strong>of</strong> Rheumatology Stanford University, Palo Alto,<br />

CA, Timothy Myles, Senior Research Scientist, Department<br />

<strong>of</strong> Medicine, Division <strong>of</strong> Hematology, Stanford University,<br />

Stanford, CA, Peggy Ho, Basic Life Science Research<br />

Associate, Department <strong>of</strong> Neurology, Stanford University,<br />

Stanford, CA, Xiaoyan Du, Post Doc, Department <strong>of</strong><br />

Medicine, Division <strong>of</strong> Hematology, Stanford University,<br />

Stanford, CA, Shadi Sharif, Life Science Research Assistant,<br />

Department <strong>of</strong> Medicine, Division <strong>of</strong> Hematology, Stanford<br />

University, Stanford, CA, Lawrence Leung, Pr<strong>of</strong>essor <strong>of</strong><br />

Medicine, Department <strong>of</strong> Medicine, Division <strong>of</strong> Hematology,<br />

Stanford University, Stanford, CA, William Robinson,<br />

Assistant Pr<strong>of</strong>essor in Medicine, Department <strong>of</strong> Medicine<br />

Division <strong>of</strong> Rheumatology Stanford University, Palo Alto, CA<br />

Thrombin-activatable carboxypeptidase B (CPB) is well<br />

established to play an anti-fibrinolytic role by removing Cterminal<br />

lysine residues from fibrin, thereby preventing its<br />

cleavage by plasmin. Recently, C3a, C5a, thrombin-cleaved<br />

osteopontin and bradykinin have been identified as additional<br />

substrates <strong>of</strong> CPB. CPB removes arginine residues from<br />

the C-terminus <strong>of</strong> these inflammatory mediators, thereby<br />

altering the biologic functions. We investigated the role <strong>of</strong><br />

CPB in arthritis using proCPB deficient mice. Arthritis was<br />

induced by an intravenous injection <strong>of</strong> anti-collagen antibodies<br />

on day 0, followed by intraperitoneal injection <strong>of</strong><br />

lipopolysaccharide on day 3. Arthritis was assessed by visual<br />

score and paw thickness. On day 10 post-injection, joints<br />

were collected for histology. Arthritis was also studied in<br />

bradykinin receptor−/− mice and C5−/− mice. As compared<br />

to controls, proCPB−/− mice exhibited significantly more<br />

severe arthritis (on day 10 arthritis score 12.3 vs. 0.4*; paw<br />

thickness 3.02 vs. 2.21 mm*, *pb0.01). Histology, graded 0–<br />

4, demonstrated severe arthritis in proCPB −/− mice versus<br />

controls (synovitis 3.7 vs. 1.2*, pannus 2.8 vs. 1.1*, bone<br />

erosion 3.2 vs. 0.7*, *pb0.01). C5−/− mice were resistant to<br />

induction <strong>of</strong> arthritis, while bradykinin receptor−/− mice<br />

exhibited a similar severity <strong>of</strong> arthritis to controls. These<br />

results suggest that CPB plays a critical role in regulating<br />

inflammation in anti-collagen antibody induced arthritis,<br />

and that this effect could be in part mediated by its downregulation<br />

<strong>of</strong> C5a activity. Studies are underway to determine<br />

the impact <strong>of</strong> CPB on C5a, C3a, and thrombin-cleaved<br />

osteopontin in the pathogenesis <strong>of</strong> this arthritis model.<br />

doi:10.1016/j.clim.2007.03.459<br />

Sa.72 Kinetics <strong>of</strong> Cytokine Gene Expression After<br />

Antigen Stimulation in Frozen Human PBMCs Using<br />

Quantitative Real-Time PCR and Flow Cytometry<br />

Annick van de Ven, MD Student, University <strong>of</strong> California, San<br />

Diego, La Jolla, CA, Pediatrics/University <strong>of</strong> Utrecht, The


Abstracts<br />

Netherlands, Elissa Keogh, Senior Research Associate,<br />

Supervisor, University <strong>of</strong> California, San Diego, La Jolla, CA,<br />

Negar Ghahramani, Staff Research Associate, Pediatrics,<br />

University <strong>of</strong> California, San Diego, La Jolla, CA, Salvatore<br />

Albani, Pr<strong>of</strong>essor <strong>of</strong> Pediatrics, University <strong>of</strong> California, San<br />

Diego, Department <strong>of</strong> Pediatrics, La Jolla, CA<br />

Background: Understanding <strong>of</strong> antigen-induced kinetics <strong>of</strong><br />

gene expression facilitates the analysis <strong>of</strong> immune responses<br />

using RT-PCR and DNA microarrays. Therefore, cytokine<br />

expression was investigated using 2 distinct antigenic stimuli:<br />

a common recall antigen (influenza peptide HA307–319) and<br />

the peptide epitope dnaJP1 associated with the pathogenesis<br />

<strong>of</strong> rheumatoid arthritis. Method: Frozen PBMCs from 5<br />

dnaJP1-responsive RA patients and fresh and frozen PBMCs<br />

from one healthy, influenza-vaccinated donor were stimulated<br />

for up to 96 hours with or without dnaJP1 and HA307–<br />

319, respectively. At designated intervals, aliquots were<br />

removed for measurement <strong>of</strong> cytokines by TaqMan. Additionally,<br />

intracellular IL-2 and TNFα were measured by FACS.<br />

Results: Following dnaJP1 stimulation, both IL-2 and IFNγ<br />

expressions were detectable by TaqMan at 12–36 hours while<br />

TNFα was undetectable. Intracellular IL-2 exhibited a<br />

biphasic pr<strong>of</strong>ile (48 and 96 hours) and TNFα expression was<br />

detected at 48 hours. It should be noted that gene expression<br />

at 6 hours was highly nonspecific. When fresh PBMCs were<br />

stimulated with HA307–319, IL-2 expression was biphasic (24<br />

and 48 hours) while TNFα and INFγ peaked at 12 and 48 hours,<br />

respectively. Gene expression in frozen PBMCs appeared to be<br />

delayed by about 12 hours. Conclusions: Peptide antigen<br />

stimulation induces cytokine expression more slowly than<br />

that observed for mitogen stimulation. Gene expression also<br />

depends, to some extent, on the gene <strong>of</strong> interest. As<br />

expected, intracellular detection follows gene expression<br />

by 24–36 hours. Additionally, PBMCs require a 12 hour<br />

recovery period after cryopreservation.<br />

doi:10.1016/j.clim.2007.03.460<br />

Sa.73 Epitope-specific Immune Tolerance Can<br />

Maintain the Disease Control Induced by<br />

Conventional Methotrexate—Etanercept<br />

Combination Therapy in Rat Adjuvant Arthritis<br />

Negar Ghahramani, Staff Research Associate, University <strong>of</strong><br />

California, San Diego, Department <strong>of</strong> Pediatrics, La Jolla,<br />

CA, Elissa Keogh, Staff Research Associate, Supervisor,<br />

University <strong>of</strong> California, San Diego Department <strong>of</strong><br />

Pediatrics, La Jolla, CA, Annick van de Ven, MD Student,<br />

University <strong>of</strong> California, San Diego Pediatrics/University <strong>of</strong><br />

Utrecht, The Netherlands, La Jolla, CA, Salvatore Albani,<br />

Pr<strong>of</strong>essor, University <strong>of</strong> California, San Diego, Department<br />

<strong>of</strong> Pediatrics, La Jolla, CA<br />

Tools to maintain remission that allow tapering from<br />

aggressive therapies are still lacking for the majority <strong>of</strong><br />

patients with rheumatoid arthritis. The success for longterm<br />

disease control with minimal background therapy<br />

could lie in the ability to establish tolerance. This approach<br />

can be complementary to the blend <strong>of</strong> blockade and<br />

suppression which characterizes current therapies. We<br />

have demonstrated that in rats with adjuvant arthritis<br />

(AA), mucosal tolerization to an arthritogenic peptide<br />

(hsp60 p180–188) can lead to significant improvement and<br />

immune deviation. In this study, the effect <strong>of</strong> nasal hsp60<br />

p180–188 peptide treatment (0.1 mg, days 10, 13 and 16) in<br />

combination with oral methotrexate (0.3 mg/kg, days 9 and<br />

13) and s.c. etanercept (0.3 mg/kg, day 9) was compared<br />

with standard etanercept–methotrexate treatment regimen.<br />

Results clearly indicate that the combination <strong>of</strong><br />

mucosal tolerization to hsp60 p180–188 with single dose<br />

<strong>of</strong> etanercept can lead to significant disease control, to a<br />

degree comparable to full dose etanercept and tangibly<br />

better than the other treatment regimens (average clinical<br />

improvement <strong>of</strong> 42.3% for hsp60 p180–188 +single dose<br />

etanercept+methotrexate, 36.7% for hsp60 p180–188+<br />

methotrexate; and 14% for full dose etanercept–methotrexate<br />

therapy). The marked increase in the percentage <strong>of</strong> CD4<br />

+ CD25hiFoxp3+ T cells following epitope-specific combination<br />

therapy suggested induction <strong>of</strong> Treg cells and restoration<br />

<strong>of</strong> tolerance. Such changes were restricted to the<br />

draining site <strong>of</strong> the nasal mucosa <strong>of</strong> animals receiving the<br />

combined treatment.<br />

doi:10.1016/j.clim.2007.03.461<br />

S99<br />

Sa.74 Independent Signals Within the MHC are<br />

Associated with Systemic Lupus Erythematosus—A<br />

Family-based SNP Study<br />

Michelle Fernando, ARC <strong>Clinical</strong> Research Fellow, Imperial<br />

College London, Section <strong>of</strong> Molecular Genetics and<br />

Rheumatology, London, England, Christine Stevens, Project<br />

Coordinator, The Broad Institute <strong>of</strong> MIT and Harvard,<br />

Inflammatory Genetics Group, Cambridge, MA, Emily Walsh,<br />

Cancer Research Institute Postdoctoral Fellow, Broad<br />

Institute <strong>of</strong> MIT and Harvard, Cambridge, MA, Todd Green,<br />

Project Coordinator, Program in Medical and Population<br />

Genetics, Broad Institute <strong>of</strong> Harvard and MIT, Cambridge,<br />

MA, Pardis Sabeti, Postdoctoral Fellow, Infectious Disease<br />

Initiative, The Broad Institute <strong>of</strong> MIT and Harvard,<br />

Cambridge, MA, John Rioux, Canada Research Chair in<br />

Genetics and Genomic Medicine <strong>of</strong> Inflammation, Universite<br />

de Montreal, Montreal Heart Institute, Montreal, QC,<br />

Canada, Tim Vyse, Reader and Honorary Consultant in<br />

Rheumatology/Medicine, Imperial College London, Section<br />

<strong>of</strong> Molecular Genetics and Rheumatology, London, England<br />

The major histocompatibility complex (MHC) has been<br />

associated with SLE in numerous case–control studies. The<br />

main associated alleles are HLA-DR3 (0301) and HLA-DR2<br />

(1501) and their respective haplotypes in white Caucasian<br />

populations. MHC class III alleles have also shown association<br />

with lupus but extensive linkage disequilibrium (LD) in<br />

the region has prevented the discovery <strong>of</strong> the causal<br />

alleles. We therefore set out to investigate whether we<br />

could delineate separate class II and class III signals in our<br />

UK SLE cohort using a family-based SNP association study.<br />

67 SNPs were successfully typed using the Sequenom<br />

MassARRAYTM platform across 1.7 mb <strong>of</strong> genome (HLA-B<br />

to HLA-DPA2) in 314 Caucasian UK SLE trios. 4-digit HLA-<br />

DRB1 typing was performed using oligo-hybridization. Our


S100 Abstracts<br />

findings confirm previously published data <strong>of</strong> an association<br />

with HLA-DRB1*0301 (p=2.1×10 −8 ). We find no association<br />

with HLA-DRB1*1501 or HLA-DRB1*0801. 15 out <strong>of</strong> 67 SNP<br />

markers show evidence <strong>of</strong> association with WHAP TDT<br />

(permutated pb0.05, 10,000 permutations). Conditional<br />

analysis (WHAP) reveals association signals independent <strong>of</strong><br />

HLA-DRB1*0301 in class I (HLA-B) and class III (BAT3,<br />

SLC44A4, RDBP) with the latter signal being the strongest.<br />

These independent markers are not in LD with any other<br />

HLA-DRB1 allele (TRANSMIT) but do arise from the B8-<br />

DRB1*0301 extended haplotype shown by Extended Haplotype<br />

Homozygosity analysis (SWEEP).<br />

HLA-DRB1*0301 is associated with SLE in our UK cohort.<br />

We have also demonstrated a separate class III signal arising<br />

from recombination on the B8-DRB1*0301 extended haplotype.<br />

The haplotype block from which the class III signal<br />

arises encompasses the complement C4-containing RCCX<br />

module.<br />

doi:10.1016/j.clim.2007.03.462<br />

Sa.75 Quantitation <strong>of</strong> Total and Unoccupied BAFF-R<br />

in SLE<br />

Seth Knight, University <strong>of</strong> Alabama, Birmingham,<br />

Department <strong>of</strong> Medicine, Birmingham, AL, Hong Zhao,<br />

University <strong>of</strong> Alabama, Birmingham, Department <strong>of</strong><br />

Medicine, Birmingham, AL, Tong Zhou, University <strong>of</strong><br />

Alabama, Birmingham, Department <strong>of</strong> Medicine,<br />

Birmingham, AL, Robert Carter, University <strong>of</strong> Alabama,<br />

Birmingham, Department <strong>of</strong> Medicine, Birmingham, AL<br />

Background: Systemic lupus erythematosus (SLE) is an<br />

autoimmune disease characterized by abnormally active B<br />

lymphocytes. B lymphocyte stimulator (BLyS) has been<br />

shown to be an important regulator <strong>of</strong> B cell activity in SLE.<br />

The serum concentration <strong>of</strong> BLyS is increased in SLE<br />

patients, and in mice, increased BLyS induces a lupus-like<br />

syndrome. We have previously demonstrated that BAFF-R,<br />

the primary receptor for BlyS on B cells, is occupied to a<br />

greater extent on PBMCs in SLE patients than in healthy<br />

controls. We have now developed a robust assay for<br />

measuring total BAFF-R and occupied BAFF-R using quantitative<br />

flow cytometry. Methods/Results: Anti-BAFF-R monoclonal<br />

Ab-producing hybridoma clones were screened by<br />

ELISA. We identified clone 4A4 and performed a BLyS<br />

competition assay on tonsillar B cells and show that BlyS<br />

and 4A4 Ab competitively bind to BAFF-R. 4A4 Ab and a<br />

second anti-BAFF-R Ab (11C1) were then used to quantitate<br />

total and occupied BAFF-R on peripheral blood B cells in SLE<br />

and control cohorts using bead-based, quantitative flow<br />

cytometry to derive absolute values for antibody binding<br />

based on a standard curve. We show that both total and<br />

unoccupied BAFF-R are decreased on peripheral blood B<br />

cells in SLE patients, in some cases to 10% <strong>of</strong> healthy<br />

controls. These data demonstrate a more robust system for<br />

BAFF-R analysis in SLE and confirm the engagement <strong>of</strong><br />

BAFF-R in most lupus subjects.<br />

doi:10.1016/j.clim.2007.03.464<br />

Sa.76 Role <strong>of</strong> CD8+ Ti cells in Suppression <strong>of</strong><br />

Autoimmunity Depends on Foxp3 Expression<br />

Ram Singh, Assistant Pr<strong>of</strong>essor, David Geffen School <strong>of</strong><br />

Medicine at University <strong>of</strong> California, Los Angeles, Los<br />

Angeles, CA, Antonio La Cava, Associate Pr<strong>of</strong>essor, David<br />

Geffen School <strong>of</strong> Medicine at University <strong>of</strong> California, Los<br />

Angeles, Medicine, Los Angeles, CA, Bevra Hahn, Pr<strong>of</strong>essor<br />

and Chief, David Geffen School <strong>of</strong> Medicine at University <strong>of</strong><br />

California, Los Angeles (Medicine), Los Angeles, CA<br />

Systemic lupus erythematosus (SLE) is an autoimmune<br />

disease caused by autoantibodies including IgG anti-DNA.<br />

NZB/NZW Fl female (BWF1) mice, a model <strong>of</strong> spontaneous<br />

polygenic SLE, tolerized with an artificial peptide (pCONSEN-<br />

SUS, pCons) based on anti-DNA IgG sequences containing MHC<br />

class I and class iI T cell determinants, develop regulatory<br />

CD4+CD25+ T cells and inhibitory CD8+ T cells (CD8+ Ti), both<br />

<strong>of</strong> which suppress autoAb production. In the present study,<br />

using various cellular and molecular approaches, we show that<br />

inhibitory function <strong>of</strong> CD8+ T cells from tolerized mice is<br />

sustained for up to 8 weeks and at all times depends on<br />

expression <strong>of</strong> Foxp3. Both CD28-positive and -negative CD8+ T<br />

cells contain inhibitory cells, but the expression <strong>of</strong> Foxp3 and<br />

<strong>of</strong> mRNA for TGFb is higher and lasts longer in the CD28−<br />

subset. In vitro addition <strong>of</strong> TGFb (in the presence <strong>of</strong> IL-2)<br />

induces Foxp3 expression in a dose–response manner. Gene<br />

inhibition or blockade with siRNA <strong>of</strong> Foxp3 abrogates the<br />

ability <strong>of</strong> the CD8+ Ti to inhibit anti-DNA production and the<br />

proliferation <strong>of</strong> CD4+ helper T cells. Moreover, we found a<br />

significant correlation between expression <strong>of</strong> Foxp3 and<br />

ability <strong>of</strong> CD8+ Ti cells to secrete TGFb. Therefore, CD8+ Ti<br />

in this system <strong>of</strong> tolerance is dependent on expression <strong>of</strong><br />

Foxp3, and there may be a bi-directional Foxp3/TGFb<br />

autocrine loop that determines the ability <strong>of</strong> the CD8+ T<br />

cells to control autoimmunity. Supported by NIH grants.<br />

R37 AI 46776, AI63515AR53239, Tina C Foundation, and<br />

gifts from Jeanne Rappaport and the Horchow Family<br />

Trust.<br />

doi:10.1016/j.clim.2007.03.465<br />

Sa.78 T Cell Epitope Molecular Mimicry Can Initiate<br />

Immune Response Against Lupus-associated SmD<br />

Autoantigen<br />

Davis Sim, MD/PhD Student, University <strong>of</strong> Virginia Division<br />

<strong>of</strong> Rheumatology and <strong>Immunology</strong>, Charlottesville, VA,<br />

Govind Rajagopolan, Research Associate, Mayo Clinic<br />

Department <strong>of</strong> <strong>Immunology</strong>, Rochester, MN, Shu Man Fu,<br />

Pr<strong>of</strong>essor, University <strong>of</strong> Virginia Division <strong>of</strong> Rheumatology<br />

and <strong>Immunology</strong>, Charlottesville, VA, Chella David,<br />

Pr<strong>of</strong>essor, Mayo Clinic Department <strong>of</strong> <strong>Immunology</strong>,<br />

Rochester, MN, Umesh Deshmukh, Assistant Pr<strong>of</strong>essor,<br />

University <strong>of</strong> Virginia Division <strong>of</strong> Rheumatology and<br />

<strong>Immunology</strong>, Charlottesville, VA<br />

Autoantibodies reactive with Sm proteins are specific<br />

for systemic lupus erythematosus. One <strong>of</strong> the hypotheses<br />

behind the initiation <strong>of</strong> an immune response against Sm<br />

autoantigens is molecular mimicry between foreign and<br />

self proteins. However, clear evidence for this at T cell


Abstracts<br />

level is lacking. This study was designed to determine the<br />

role <strong>of</strong> T cell epitope mimicry in the initiation <strong>of</strong> immune<br />

response against SmD1.Using HLA-DR3 transgenic mice as<br />

experimental model system, a dominant T cell epitope<br />

was identified between amino acid SmD71 and 95. Using a<br />

T cell hybridoma (C1P2) reactive against this epitope,<br />

critical amino acid residues were localized within SmD81–<br />

88. Screening <strong>of</strong> databases with pattern recognition and<br />

MHC binding s<strong>of</strong>tware identified 20 putative peptide<br />

mimics. Among these, a peptide from galactoside ABC<br />

transporter protein from Vibrio activated C1P2. Immunization<br />

<strong>of</strong> HLA-DR3 mice with this peptide induced both T and<br />

B cell responses against SmD. To further characterize T<br />

cell responses against this mimic, additional T cell<br />

hybridomas were generated. Interestingly a T hybridoma<br />

P20 not only reacted with SmD79–93 and the Vibrio<br />

peptide but also with peptides from a human La-related<br />

protein and methyltransferase protein from Streptococcus<br />

agalactiae. TCR characterization <strong>of</strong> both C1P2 and P20<br />

revealed different V 2 usage, V 2 8.3 and V 2 8.1 respectively.<br />

Our study demonstrates the potential <strong>of</strong> T cell<br />

epitope mimicry to initiate autoimmune responses against<br />

lupus-associated antigens. In addition our data suggest<br />

that based on the TCR usage, different T cells reactive<br />

against the same region <strong>of</strong> an autoantigen can be<br />

activated by multiple molecular mimics.<br />

doi:10.1016/j.clim.2007.03.466<br />

Sa.79 Autoantibody Onset Prior to SLE Diagnosis<br />

Follows Predictable Patterns <strong>of</strong> Development<br />

Latisha Heinlen, Graduate Student, Oklahoma Medical<br />

Research Foundation, Oklahoma City, OK, Micah McClain,<br />

Scientist, Oklahoma Medical Research Foundation,<br />

Oklahoma City, OK, Tony Prestigiacomo, Scientist, BioRad<br />

Laboratories, Benecia, CA, Ben Bruner, Graduate Student,<br />

Oklahoma Medical Research Foundation, Oklahoma City,<br />

OK, Steven Binder, Scientist, Bio-Rad Laboratories,<br />

Benecia, CA, Colin Edgerton, Chief, Rheumatology Service,<br />

US Army Center for Health Promotion and Prevention,<br />

Washington, DC, Mark Rubertone, Active Duty Military, US<br />

Army Center for Health Promotion and Preventive<br />

Medicine, Washington, DC, Judith James, Member,<br />

Oklahoma Medical Research Foundation, Oklahoma City,<br />

OK, John Harley, Member, Oklahoma Medical Research<br />

Foundation, Oklahoma City, OK<br />

Systemic lupus erythematosus (SLE) is a clinically<br />

diverse humoral autoimmune disorder. We sought to<br />

determine the earliest known protein autoantigens targeted<br />

prior to SLE diagnosis, and to define the order in<br />

which specific protein autoantibodies arise. Previously<br />

collected and stored serum samples were obtained from<br />

the United States Department <strong>of</strong> Defense Serum Repository.<br />

All available patient and selected control samples were<br />

tested for autoantibodies to lupus autoantigens. Using<br />

these prospective samples we identified patterns <strong>of</strong><br />

autoantibody onset. In 33 patients the initial single<br />

autoantibody specificities were identified and were most<br />

commonly against 60 kDa Ro, and nRNP A. Surprisingly,<br />

none <strong>of</strong> these patients initiated lupus autoimmunity with<br />

anti-nRNP 70K or anti-Sm. Also, no patient had antibodies to<br />

nRNP 70K without antibodies to nRNP A. Anti-A appeared<br />

before or simultaneously with anti-70K in 96% <strong>of</strong> patients that<br />

developed both under observation. Anti-60 kDa Ro antibodies<br />

appeared before or simultaneously with anti-La or anti-52kD<br />

Ro in nearly all patients (98% and 95% respectively). These<br />

data suggest that the autoantibody response in SLE begins<br />

simply, <strong>of</strong>ten with a single protein years before disease onset,<br />

followed by the accumulation <strong>of</strong> additional autoantigenic<br />

specificities in partly predictable patterns.<br />

doi:10.1016/j.clim.2007.03.467<br />

Sa.80 Osteopontin Gene Polymorphism and<br />

Systemic Lupus Erythematosus in Chinese<br />

Maida Wong, <strong>Clinical</strong> Fellow, University <strong>of</strong> California, Los<br />

Angeles, School <strong>of</strong> Medicine, Los Angeles, CA, Joseph Yeung,<br />

Research Associate, University <strong>of</strong> Hong Kong, Faculty <strong>of</strong><br />

Medicine, Hong Kong, Chak-Sing Lau, Pr<strong>of</strong>essor, University<br />

<strong>of</strong> Hong Kong, Faculty <strong>of</strong> Medicine, Hong Kong<br />

Osteopontin (OPN) is a secreted glycosylated and phosphorylated<br />

extracellular matrix protein that influences autoimmune<br />

disease. Its serum level is known to be elevated in<br />

patients with systemic lupus erythematosus (SLE). In this<br />

study, we analyzed a minor Tallele (C707T SNP, corresponding<br />

to rs1126616) <strong>of</strong> OPN in Chinese SLE patients and healthy<br />

controls by polymerase chain reaction. Serum OPN levels were<br />

measured by ELISA, and its correlation with OPN polymorphism<br />

was analyzed statistically based on their genotypes, renal<br />

involvement, and disease activity (active disease defined as<br />

SLEDAI = or > 4). 162/253 SLE patients and 91/168 controls had<br />

one or more allelic mutations at 707T <strong>of</strong> the OPN gene. OPN<br />

mutation is associated with the development <strong>of</strong> SLE, OR = 1.51<br />

(95% CI 1.012-2.253). The OPN genotype is also associated with<br />

SLE (p=0.0003). Serum OPN is elevated in SLE patients with<br />

history <strong>of</strong> renal disease, irrespective <strong>of</strong> their disease activity<br />

(pb0.005 by ANOVA and post-hoc test, p= 0.013 by Mann-<br />

Whitney U test). There was no association between the allelic<br />

mutation and CNS disease, hematological disorders, malar or<br />

discoid rash, low complements, or elevated dsDNA antibody.<br />

The C707Tallelic frequency <strong>of</strong> the control population followed<br />

the Hardy-Weinberg equilibrium. Although the allelic frequencies<br />

between SLE patients (0.340) and controls (0.327) were<br />

not significantly different, they were significantly higher than<br />

the healthy Caucasian controls as published by Forton et al<br />

(p=0.004), which may explain the higher prevalence <strong>of</strong> SLE<br />

in the Chinese population, especially those with renal<br />

involvement.<br />

doi:10.1016/j.clim.2007.03.468<br />

S101<br />

Sa.81 Quantitative and Qualitatively Normal<br />

Regulatory T Cells are Not Capable <strong>of</strong> Inducing<br />

Suppression in SLE Patients Due to T Cell Resistance<br />

María Inés Vargas-Rojas, Medical Doctor, Instituto Nacional<br />

de Ciencias Médicas y Nutrición Salvador Zubiran/


S102 Abstracts<br />

Rheumatology and <strong>Immunology</strong>, Mexico, José Carlos Crispín,<br />

Medical Doctor, Instituto Nacional de Ciencias Médicas y<br />

Nutrición Salvador Zubiran/Rheumatology and <strong>Immunology</strong>,<br />

Mexico, Jorge Alcocer-Varela, Medical Doctor, Instituto<br />

Nacional de Ciencias Médicas y Nutrición Salvador Zubiran/<br />

Rheumatology and <strong>Immunology</strong>, Mexico<br />

Regulatory T cells (Treg) are considered an essential<br />

component <strong>of</strong> the mechanisms that protect and maintain<br />

peripheral tolerance. Their absence leads inevitably to lifethreatening<br />

autoimmune disease. Numerous works have<br />

studied their numbers and behavior in patients with<br />

autoimmune conditions; several defects that include diminished<br />

numbers and lack <strong>of</strong> suppressive capacity have been<br />

documented in various conditions including systemic lupus<br />

erythematosus. The aim <strong>of</strong> this study was to further<br />

characterize the regulatory defect documented in Treg <strong>of</strong><br />

patients with SLE. Twenty patients with SLE (10 with active<br />

disease) and 20 age- and sex-matched controls were<br />

included. Patients were not receiving treatment. Treg<br />

cells, defined as CD4+Foxp3+, were quantified by flow<br />

cytometry. CD4+CD25+ and CD4+CD25− cells were isolated<br />

by magnetic beads. Suppressive capacity was quantified in<br />

autologous and allogeneic co-cultures. No quantitative<br />

difference was found in Treg numbers between patients<br />

and controls (2.8±2.1 vs. 3.2±2.2, respectively). Suppression<br />

<strong>of</strong> cell proliferation was not observed in autologous cocultures<br />

<strong>of</strong> SLE cells. Conversely, normal Treg greatly<br />

suppressed autologous T cell proliferation (∼80%). Surprisingly,<br />

when SLE-derived Treg were added to healthy T cell<br />

cultures suppression was observed. In contrast, T cells<br />

obtained from SLE patients were not susceptible to suppression<br />

by normal Treg. Our findings suggest that even though a<br />

T cell suppression defect is present in patients with SLE, it<br />

cannot be explained by defects in Treg. Our data indicate<br />

that SLE-derived T cells are resistant to Treg-mediated<br />

suppression.<br />

doi:10.1016/j.clim.2007.03.469<br />

Sa.82 Increased Regulatory T Cell Prevalence and<br />

Retained Capacity to Suppress Effector T Cells<br />

During Disease in NZB/W Lupus Mice<br />

Kenneth Scalapi, Adjunct Assistant Pr<strong>of</strong>essor <strong>of</strong> Medicine,<br />

University <strong>of</strong> California, San Francisco and San Francisco VA<br />

Medical Center, San Francisco, CA, David Daikh, Associate<br />

Pr<strong>of</strong>essor <strong>of</strong> Medicine, University <strong>of</strong> California, San<br />

Francisco and San Francisco VA Medical Center, San<br />

Francisco, CA<br />

Purpose: SLE is characterized by loss <strong>of</strong> tolerance to<br />

multiple self-antigens. Regulatory T cells (Tregs) serve to<br />

maintain peripheral tolerance by inhibiting autoreactive<br />

effector Tcells (Teff). Studies have correlated low peripheral<br />

blood Treg prevalence and/or abnormal Treg function with<br />

several autoimmune diseases including SLE. To assess Treg<br />

frequency and function in lupus prone NZB/W (B/W) mice,<br />

we evaluated CD4+Foxp3+ T cells during the course <strong>of</strong><br />

disease. Cellular function in young and old B/W mice was<br />

assessed utilizing the CD4+CD25+ Treg subset, young and old<br />

Teffs and antigen-presenting cells (APCs) in mixed suppression<br />

assays. Methods: CD4+Foxp3+ Treg prevalence was<br />

measured by FACS. Function <strong>of</strong> purified Tregs, Teffs and<br />

APCs was evaluated in mixed suppression assays. Results:<br />

Frequency <strong>of</strong> CD4+Foxp3+ Tregs as a percentage <strong>of</strong> CD4+ cells<br />

is low only in pre-diseased B/W mice. During active lupus,<br />

Tregs prevalence significantly increases such that it equals or<br />

exceeds that <strong>of</strong> age-matched control mice. A significant<br />

fraction <strong>of</strong> Tregs in sick mice loses CD25− expression. The<br />

Treg prevalence in peripheral blood did not correlate with<br />

tissue prevalence or disease state. Mixed suppression assays<br />

demonstrated that the function <strong>of</strong> CD4+CD25+ Tregs is not<br />

affected by disease state and Teffs do not become resistant<br />

to Treg suppression as mice age and develop disease.<br />

Conclusion: A significant expansion <strong>of</strong> functional Tregs, not<br />

evident in peripheral blood, occurs in lupus prone B/W with<br />

disease onset, emphasizing the unreliability <strong>of</strong> peripheral<br />

blood to assess these cells. Furthermore, Tregs retain the<br />

capacity to suppress Teffs in mice with active lupus.<br />

doi:10.1016/j.clim.2007.03.470<br />

Sa.84 Immune Opsonins Modulate BLyS/BAFF<br />

Release in a Receptor-specific Fashion<br />

Xinrui Li, Graduate Student, Division <strong>of</strong> <strong>Clinical</strong> <strong>Immunology</strong><br />

and Rheumatology, University <strong>of</strong> Alabama at Birmingham,<br />

Birmingham, AL, Jeffrey Edberg, Associate Pr<strong>of</strong>essor,<br />

Division <strong>of</strong> <strong>Clinical</strong> <strong>Immunology</strong> and Rheumatology,<br />

University <strong>of</strong> Alabama at Birmingham, Birmingham, AL,<br />

Kaihong Su, Assistant Pr<strong>of</strong>essor, Division <strong>of</strong> <strong>Clinical</strong><br />

<strong>Immunology</strong> and Rheumatology, University <strong>of</strong> Alabama at<br />

Birmingham, Birmingham, AL, Tong Zhou, Associate<br />

Pr<strong>of</strong>essor, Division <strong>of</strong> <strong>Clinical</strong> <strong>Immunology</strong> and<br />

Rheumatology, University <strong>of</strong> Alabama at Birmingham,<br />

Birmingham, AL, Alexander Szalai, Associate Pr<strong>of</strong>essor,<br />

Division <strong>of</strong> <strong>Clinical</strong> <strong>Immunology</strong> and Rheumatology,<br />

University <strong>of</strong> Alabama at Birmingham, Birmingham, AL,<br />

Robert Kimberly, Pr<strong>of</strong>essor, Division <strong>of</strong> <strong>Clinical</strong> <strong>Immunology</strong><br />

and Rheumatology, University <strong>of</strong> Alabama at Birmingham,<br />

Birmingham, AL<br />

TNF ligand superfamily member 13B (B lymphocyte<br />

stimulator (BLyS), B cell activating factor (BAFF)) promotes<br />

primary B cell proliferation and immunoglobulin production.<br />

It exists in both membrane-bound and soluble forms. Because<br />

the soluble form is thought to be the primary biologically<br />

active form, we investigated factors that might regulate its<br />

cleavage and processing. In both myeloid cell lines (U937,<br />

THP-1and HL-60) and primary human monocytes, the<br />

shedding <strong>of</strong> membrane BLyS can be regulated by CRP and<br />

IgG. Within 10 minites, both <strong>of</strong> these opsonins trigger<br />

significant shedding <strong>of</strong> BLyS (up to 52.2+/-3.0%, p=0.003)<br />

by engaging FcgRs. In U937 cells, a primary role <strong>of</strong> FcgRI is<br />

demonstrated both by direct receptor cross-linking (FcgRI<br />

45.69+/-2.05% verses FcgRIIa 2.11+/-1.33%) and by the lack<br />

<strong>of</strong> enhanced cleavage when FcgRI and FcgRIIa are cocrosslinked.<br />

The shed BLyS is biologically functional since it<br />

promotes B cell survival. The generation <strong>of</strong> a B cell<br />

proliferation and survival factor by Fc gamma receptor<br />

engagement, especially FcgRI, which can also enhance


Abstracts<br />

antigen uptake and presentation mediated by ligands <strong>of</strong> both<br />

innate and adaptive immune systems, provides a unique<br />

opportunity to facilitate antibody production. These functions<br />

suggest that Fc gamma receptors may provide a link between<br />

immune complex mediated autoimmune diseases and elevations<br />

in circulating BLyS in autoimmune disease patients.<br />

doi:10.1016/j.clim.2007.03.471<br />

Sa.85 The CD4+CD25+Foxp3+ T Cells in Lupus<br />

Nephritis<br />

Mercedes Zabaleta-Lanz, Immunologist, Institute <strong>of</strong><br />

<strong>Immunology</strong>, Central University School <strong>of</strong> Medicine,<br />

Caracas, Venezuela, Ronak Seyeddi, MD, Institute <strong>of</strong><br />

<strong>Immunology</strong>, Caracas, Venezuela, Maria Elena Marquez,<br />

Bbiologist, Institute <strong>of</strong> <strong>Immunology</strong>, Caracas, Venezuela,<br />

Perla Chirinos, Bioanalist, Instituto de Inmunologia, UCV,<br />

Caracas, Venezuela, Isaac Blanca, Biologist, Institute <strong>of</strong><br />

<strong>Immunology</strong>, Caracas, Venezuela, Nicolás Bianco,<br />

Immunologist, Institute <strong>of</strong> <strong>Immunology</strong>, Caracas, Venezuela,<br />

Mercedes Zabaleta-Lanz, Immunologist, Institute <strong>of</strong><br />

<strong>Immunology</strong>, Caracas, Venezuela<br />

The role <strong>of</strong> CD4+CD25+Foxp3+ regulatory T cells (Tregs)<br />

are critical in maintaining self tolerance. The levels <strong>of</strong> Treg<br />

in patient with systemic lupus erythematosus (SLE), a<br />

chronic autoimmune disorder, have been reported<br />

depressed. Here we have determined the levels <strong>of</strong> Treg<br />

and the CTLA-4 CD4 T cells in 21 patients with SLE, 7 with<br />

active disease and 15 with inactive disease and 25 healthy<br />

controls. Treg and CTLA-4 were evaluated in highly purified<br />

CD4 T cells from patients and controls by flow cytometry<br />

using a tri-color analysis protocol. The 7 SLE patients with<br />

active disease, 4 with overt lupus nephritis (OLN) and 3<br />

with silent lupus nephritis (SLN) show a significant reduction<br />

<strong>of</strong> Treg in comparison to controls (SLN, 0.23±0.18%,<br />

OLN 1.35±1.21%, controls, 3,69±3%¸ pb0.05 and pb0.02<br />

respectively). A significant reduction <strong>of</strong> Treg was also<br />

observed between the inactive SLE patients versus controls<br />

(pb0.02). A higher expression <strong>of</strong> Foxp3 was observed in<br />

Treg cells from patients with active SLE disease (88.74 ±<br />

5.92%) vs. inactive disease (71.39 ±20.15%; p=0.008). Nonsignificant<br />

differences were observed in the levels <strong>of</strong> CD4<br />

+CD25+CTLA-4+ T cells, and no correlation between Tregs<br />

and serological parameters (anti-DNA, anti-C1q, antinucleosome,<br />

C3 and C4) was observed. Ours results suggest<br />

that Tregs are significantly decreased in active lupus even<br />

before the installation <strong>of</strong> an OLN. However, the increased<br />

expression <strong>of</strong> Foxp3 in the Tregs cells from patients with<br />

active disease suggests a possible mechanism <strong>of</strong> compensatory<br />

function <strong>of</strong> these molecules.<br />

doi:10.1016/j.clim.2007.03.472<br />

Sa.86 Rapamycin Induces Tolerance by Increasing<br />

Regulatory T Cells in Chronic Graft Versus Host<br />

Disease Model<br />

Jeanne Palmer, Postdoctoral Fellow, Duke University<br />

Medical Center, Department <strong>of</strong> Hematology, Oncology and<br />

Cellular Therapy, Durham, NC, Benny Chen, Assistant<br />

Pr<strong>of</strong>essor <strong>of</strong> Medicine, Duke University Medical Center,<br />

Division <strong>of</strong> Cellular Therapy/BMT, Durham, NC, Nelson<br />

Chao, Director, Division <strong>of</strong> Cellular Therapy/BMT, Pr<strong>of</strong>essor<br />

<strong>of</strong> Medicine and <strong>Immunology</strong>, Duke University Medical<br />

Center, Durham, NC, Ngocdiep Le, Medical Sciences<br />

Director, Amgen Inc., Thousand Oaks, CA<br />

Rapamycin is an immunosuppressant that has been found<br />

to prevent/treat acute and chronic graft versus host disease<br />

after allogeneic hematopoeitic stem cell transplant (HSCT)<br />

in both murine models and humans. One possible mechanism<br />

is through induction <strong>of</strong> tolerance, which may be due to<br />

increased number or enhanced survival <strong>of</strong> regulatory T cells.<br />

In two experiments B10.D2 bone marrow and splenocytes<br />

were injected into lethally irradiated BALB/c mice. They<br />

were then given IP injections from days 1-28 with rapamycin<br />

1.5 mg/kg or 5 mg/kg, which induced tolerance in this<br />

chronic graft versus host disease model. We noted however a<br />

dense cellular infiltrate in the animals who did not have<br />

clinical GVHD. We have documented many these infiltrating<br />

cells as in situ regulatory T cells by intracellular foxP3<br />

staining. These T regs are increased in tissues <strong>of</strong> rapamycin<br />

treated mice. This effect appears to decrease following<br />

discontinuation <strong>of</strong> rapamycin. To understand the kinetics <strong>of</strong><br />

this effect, we will repeat the experiments, and sacrifice the<br />

mice at 2, 4, and 6 weeks. At that point, serum cytokine<br />

analysis, histologic evaluation <strong>of</strong> multiple organs and FACS<br />

analysis on splenocytes will be performed.<br />

doi:10.1016/j.clim.2007.03.473<br />

S103<br />

Sa.87 Elimination <strong>of</strong> Insulitis and Augmentation <strong>of</strong><br />

Islet β Cell Regeneration via Induction <strong>of</strong> Chimerism<br />

in Overtly Diabetic NOD Mice<br />

Chunyan Zhang, Postdoctoral Research Fellow, The Beckman<br />

Research Institute, City <strong>of</strong> Hope National Medical Center,<br />

Duarte, CA, Fouad Kandeel, Physician, The Beckman<br />

Research Institute, City <strong>of</strong> Hope National Medical Center,<br />

Duarte, CA, Ivan Todorov, Associate Research Scientist, The<br />

Beckman Research Institute, City <strong>of</strong> Hope National Medical<br />

Center, Duarte, CA, Mark Atkinson, Pr<strong>of</strong>essor, University <strong>of</strong><br />

Florida College <strong>of</strong> Medicine, Gainesville, FL, Chia-Lei Lin,<br />

Research Associate I, The Beckman Research Institute, City<br />

<strong>of</strong> Hope National Medical Center, Duarte, CA, Stephen<br />

Forman, Physician, The Beckman Research Institute, City <strong>of</strong><br />

Hope National Medical Center, Duarte, CO, Defu Zeng,<br />

Assistant Pr<strong>of</strong>essor, The Beckman Research Institute, City <strong>of</strong><br />

Hope National Medical Center, Duarte, CA<br />

Type 1 diabetes in both humans and NOD mice results from<br />

autoreactive T cell destruction <strong>of</strong> insulin-producing beta<br />

cells. Cure <strong>of</strong> type 1 diabetes may require both reversal <strong>of</strong><br />

autoimmunity and regeneration <strong>of</strong> beta cells. Induction <strong>of</strong><br />

chimerism via allogeneic hematopoietic transplantation<br />

(HCT) has been shown to re-establish tolerance in both<br />

prediabetic and diabetic NOD mice. However, it is unclear<br />

whether this therapy augments beta cell regeneration.<br />

Furthermore, this procedure usually requires total body


S104 Abstracts<br />

irradiation (TBI) conditioning <strong>of</strong> recipients. The toxicity <strong>of</strong><br />

TBI conditioning and potential for graft-versus-host disease<br />

(GVHD) limit the application <strong>of</strong> allogeneic HCT for treating<br />

type 1 diabetes. Here, we report that injection <strong>of</strong> donor bone<br />

marrow and CD4+ T-depleted spleen cells induced chimerism<br />

without causing GVHD in overtly diabetic NOD mice conditioned<br />

with anti-CD3/CD8, and that induction <strong>of</strong> chimerism<br />

in new-onset diabetic NOD mice led to elimination <strong>of</strong><br />

insulitis, replication <strong>of</strong> host beta cells, and reversal <strong>of</strong><br />

hyperglycemia. Although induction <strong>of</strong> chimerism in latestage<br />

<strong>of</strong> diabetic NOD mice did not reverse diabetes, the<br />

chimeras were tolerant to donor islets, and donor islet<br />

transplantation normalized blood glucose. Furthermore, we<br />

found that induction <strong>of</strong> mixed chimerism in autoreactive<br />

BDC2.5 TCR transgenic NOD mice deleted 99% <strong>of</strong> the BDC2.5<br />

T cells among host CD4+CD8+ thymocytes. Therefore, this<br />

radiation-free GVHD preventive approach for induction <strong>of</strong><br />

chimerism may represent a viable means for reversing type 1<br />

diabetes.<br />

doi:10.1016/j.clim.2007.03.474<br />

Sa.88 Purified MHC-matched Hematopoietic Stem<br />

Cell Transplantation Following Total Lymphoid<br />

Irradiation and Anti-thymocyte Globulin (TLI+ATG)<br />

Blocks EAE Pathogenesis<br />

Li-Fen Lee, Instructor, Blood and Marrow Transplantation,<br />

Stanford, CA, Matthew Burge, Research Assistant, Blood<br />

and Marrow Transplantation, Stanford, CA, Rosa Landa,<br />

Research Assistant, Blood and Marrow Transplantation,<br />

Stanford, CA, Raymond Sobels, Pr<strong>of</strong>essor, Pathology,<br />

Stanford, CA, Peggy Ho, Basic Life Science Research<br />

Associate, Neurology, Stanford, CA, Judith Shizuru,<br />

Associate Pr<strong>of</strong>essor, Blood and Marrow Transplantation,<br />

Stanford, CA<br />

In prior studies, we demonstrated that transplantation <strong>of</strong><br />

major histocompatibility complex (MHC) matched and mismatched<br />

purified hematopoietic cells (HSCs) can block<br />

diabetes pathogenesis in pre-diabetic NOD mice. Here we<br />

applied this treatment to experimental allergic encephalomyelitis<br />

(EAE), an animal model for the human autoimmune<br />

disease multiple sclerosis (MS). C57BL/6 (B6) strains congenic<br />

for the CD45 locus were immunized with MOG 35–55 to induce<br />

disease. Similar to our studies in NOD mice we observed that<br />

disease response after transplantation <strong>of</strong> MHC-mismatched<br />

hematopoietic cells in EAE affected mice was superior to those<br />

that received congenic cells. Importantly, mice transplanted<br />

with MHC-matched HSC or BM showed no significant improvement<br />

in disease symptoms when compared with congenic<br />

recipients or disease controls. This result was markedly<br />

different from the positive response observed in NOD mice<br />

transplanted with MHC-matched HSC. We then sought to<br />

optimize translation <strong>of</strong> this approach by giving total lymphoid<br />

irradiation (TLI) plus anti-thymocyte globulin (ATG), a nonmyeloablative<br />

regimen, and permitting allogeneic hematopoietic<br />

cell engraftment with significant protective effect<br />

against GVHD. We found that TLI/ATG treatment in wild type<br />

C57BL/6 mice permits engraftment <strong>of</strong> purified HSC isolated<br />

from MHC-matched AKR/B donors and improves disease<br />

severity significantly. The level <strong>of</strong> stable mixed chimerism<br />

correlated with the clinical score. A predominance <strong>of</strong> host<br />

NK1.1 T cells and CD4+CD25+ regulatory T cells was observed<br />

after TLI/ATG regimen. We hope an MHC-matched hematopoietic<br />

graft in conjunction with TLI/ATG conditioning shown<br />

here to successfully ameliorate EAE can be translated to the<br />

treatment <strong>of</strong> human autoimmune disease.<br />

doi:10.1016/j.clim.2007.03.475<br />

Sa.89 Two Types <strong>of</strong> Immune Regulatory Mechanisms<br />

are Developed In Vivo for the Control <strong>of</strong> Chronic<br />

Graft-versus-Host Disease<br />

Magali Noval Rivas, PhD Student, Institute for Medical<br />

<strong>Immunology</strong> (IMI), Gosselies, Brussels, Marc Hazzan, Doctor,<br />

Department <strong>of</strong> Nephrology and Hemodialysis, CHR Lille,<br />

Lille, France, Michel Braun, Institute for Medical<br />

<strong>Immunology</strong> (IMI), Gosselies, Brussels<br />

We developed a graft-versus-host disease (GVHD) model<br />

based on the adoptive transfer <strong>of</strong> RAG1 −/− TcR-trangenic H-Yspecific<br />

Marilyn T cells (1×10 6 cells) into male recipients.<br />

Adoptive transfer into sub-lethally irradiated RAG1 +/+ IL-<br />

2Rgc +/+ , RAG1 −/− IL-2Rgc +/+ or RAG1 −/− IL-2Rgc −/− male<br />

recipients induced 100% mortality after 1 week. On the<br />

contrary, non-irradiated mice <strong>of</strong> the three types survived<br />

indefinitely after transfer and did not develop clinical sign <strong>of</strong><br />

acute GVHD. Marilyn T cells transferred into non-irradiated<br />

RAG1 +/+ IL-2Rgc +/+ or RAG1 −/− IL-2Rgc +/+ did not expand or<br />

were rapidly eliminated (b45×10 3 in the spleen after<br />

30 days). Remarkably, transgenic T cells transferred into<br />

non-irradiated RAG1 −/− IL-2Rgc −/− hosts expanded extensively<br />

(N15×10 6 in the spleen after 30 days) and were present<br />

in many organs (spleen, lung, liver, kidney, lymph nodes).<br />

Histology revealed their capacity to mediate chronic GVHD<br />

lesions (grade 1 after 30 days) in these hosts. The T cells<br />

did not develop a regulatory phenotype since they<br />

remained Foxp3-negative and produced high amounts <strong>of</strong><br />

IFN-g when stimulated in vitro. However, they were unable<br />

to promote acute GVHD after transfer into irradiated<br />

recipients. Our model suggests that, in vivo, there are two<br />

types <strong>of</strong> mechanisms involved into the regulation <strong>of</strong> T cell<br />

responses to persistent antigenic stimulation: one is selfcontained<br />

by the T cells and involves partial anergy,<br />

probably maintained by antigenic persistence. The other<br />

one involves the activity <strong>of</strong> existing populations <strong>of</strong><br />

regulatory cells. Both <strong>of</strong> them appear to be necessary for<br />

full T cell tolerance.<br />

doi:10.1016/j.clim.2007.03.476<br />

Sa.90 NK1.1+ Cells Promote Human Cell<br />

Repopulation via an NKG2D Dependent Mechanism<br />

Hye-Youn Son, Research Pr<strong>of</strong>essor, Samsung Biomedical<br />

Research Institute, Seoul, Republic <strong>of</strong> Korea, Sung-Yeon Joo,<br />

MS, Samsung Biomedical Research Institute, Seoul, Republic<br />

<strong>of</strong> Korea, Mi Jin Kang, MS, Samsung Biomedical Research<br />

Institute, Seoul, Republic <strong>of</strong> Korea, Sung-Joo Kim,<br />

Prodessor, Samsung Medical Center, Seoul, Republic <strong>of</strong>


Abstracts<br />

Korea, Bong-Kum Choi, MS, Samsung Biomedical Research<br />

Institute, Seoul, Republic <strong>of</strong> Korea<br />

To achieve donor-specific immune tolerance in Rag2−/−<br />

f×C−/− mice to xenogeneic human CD34+ hematopoietic<br />

stem cells (HSCs), it is imperative to understand the cell<br />

types involved in xenograft rejection. In this study, we<br />

examined whether NK1.1+ cells in Rag2−/−f×C−/− mice play<br />

a direct role in the tolerance against transplanted HSCs, if<br />

thus which molecules are important. We used CD34+<br />

hematopoietic stem cells to reconstitute human cells from<br />

either cord blood or fetal liver. From 2 weeks posttransplantation,<br />

we have monitored human cells repopulation<br />

(referred as % <strong>of</strong> CD45) in peripheral blood every 2 or<br />

4 weeks. Besides, anti-mouse CD3, CD19, NK1.1, Gr-1, and<br />

NKG2D were used to determine the mouse cells involved<br />

in tolerance induction. In our system, hCD45 was<br />

significantly increased following human HSC’s transplantation<br />

as well as successfully developed multi-lineage. We<br />

found that NK1.1+ cells, especially Gr-1− subset, were<br />

directly co-related with human cell reconstitution. Interestingly,<br />

NKG2D expression on NK1.1+ cells was significantly<br />

increased in the group highly repopulated with<br />

human cells. Finally, CD3 population appeared after<br />

transplantation was shown to be inversely related with<br />

NKG2D expression. Taken together, our data strikingly<br />

demonstrate that NK1.1+ cells play a direct role to regulate<br />

human cell reconstitution as well as HSCs engraftment in<br />

Rag2−/−f×C−/− mice. Moreover, our data show that NKG2D<br />

expression significantly contributes to tolerance induction<br />

which could be involved in donor-specific cytotoxic cell<br />

killing mechanism. We are planning to down-regulate and/<br />

or delete specific NKG2D+ subsets in mixed chimeras in<br />

vivo to clear the results.<br />

doi:10.1016/j.clim.2007.03.477<br />

Sa.91 Human Cell Reconstitution from Umbilical<br />

Cord Blood-derived Stem Cells in Rag1−/−gC−/− Mice<br />

by Co-Transplantation <strong>of</strong> Unrelated Fetal<br />

Liver/Thymus Tissues<br />

Mi Jin Kang, MS, Samsung Biomedical Research Institute,<br />

Seoul, Republic <strong>of</strong> Korea, Hye-Youn Son, Research pr<strong>of</strong>essor,<br />

Samsung Biomedical Research Institute, Seoul, Republic <strong>of</strong><br />

Korea, Bong-Kum Choi, MS, Samsung Biomedical Research<br />

Institute, Seoul, Republic <strong>of</strong> Korea, Sung-Joo Kim,<br />

Pr<strong>of</strong>essor, Samsung Medical Center, Seoul, Republic <strong>of</strong><br />

Korea, Sung-Yeon Joo, MS, Samsung Biomedical Research<br />

Institute, Seoul, Republic <strong>of</strong> Korea<br />

To establish human immune and hematopoietic development<br />

and function in vivo, a number <strong>of</strong> studies have<br />

been tried to reproduce human hematopoiesis as human<br />

hematopoietic tissue and cell transplantation in immunodeficient<br />

mice. We report a model co-transplanted <strong>of</strong><br />

human fetal thymus/liver (Thy/Liv) tissues with cord blood<br />

(CB) CD34+ cells into Rag2−/−gC−/− mice. To solve the<br />

limitation <strong>of</strong> cell dose, we also used ex vivo expansion with<br />

cytokines combination; Flt-3 ligand (FL), thrombopoietin<br />

(TPO), stem cell factor (SCF), erythropoietin (EPO), G-CSF,<br />

GM-CSF or interleukin-3 (IL-3). Rag2−/−gC−/− mice were<br />

transplanted with CB CD34+ hematopoietic progenitor cells<br />

with HLA unrelated fetal Thy/Liv. MHC-mismatched CB<br />

CD34+ cell transplantation with fetal tissues resulted in<br />

measurable engraftment levels (about 5% <strong>of</strong> human CD45)<br />

in PBL more than 4 months after transplantation. Also, we<br />

observed that implanted human fetal liver and thymus<br />

under the kidney capsule were significantly grown. We<br />

found T cell and NK cell activity after in vitro restimulation<br />

at 20 weeks after transplantation. Especially, CD3+ T cell<br />

development was remarkable in a part <strong>of</strong> ex vivo cultured<br />

cell transplanted group (included with FL, TPO, and SCF).<br />

We also found human IgG in serum 20 weeks after<br />

transplantation even though we have never seen reasonable<br />

CD19+ cells in PBL, BM, or spleen. These results<br />

demonstrate that functional human T cell can be reconstituted<br />

by transfusion <strong>of</strong> MHC mismatched CB derived<br />

CD34+ cells with co-implantation <strong>of</strong> fetal Thy/Liv and that<br />

might provide a useful tool to study human disease like<br />

viral infection.<br />

doi:10.1016/j.clim.2007.03.478<br />

S105<br />

Sa.92 Fetal Liver Cells as Corrector <strong>of</strong> Immune and<br />

Hemopoietic Activity <strong>of</strong> Allogeneic Bone Marrow<br />

Anatoliy Goltsev, Pr<strong>of</strong>essor, Institute for Problems <strong>of</strong><br />

Cryobiology and Cryomedicine <strong>of</strong> the National Academy <strong>of</strong><br />

Science <strong>of</strong> Ukraine, Kharkov, Ukraine, Irina Matsevitaya,<br />

Institute for Problems <strong>of</strong> Cryobiology and Cryomedicine <strong>of</strong><br />

the National Academcy <strong>of</strong> Science <strong>of</strong> Ukraine, Kharkov,<br />

Ukraine, Elena Lutsenko, Institute for Problems <strong>of</strong><br />

Cryobiology and Cryomedicine <strong>of</strong> the National Academy <strong>of</strong><br />

Science <strong>of</strong> Ukraine, Kharkov, Ukraine, Yulia Kozlova,<br />

Institute for Problems <strong>of</strong> Cryobiology and Cryomedicine <strong>of</strong><br />

the National Academy <strong>of</strong> Science <strong>of</strong> Ukraine, Kharkov,<br />

Ukraine, Tatyana Dubrava, Institute for Problems <strong>of</strong><br />

Cryobiology and Cryomedicine <strong>of</strong> the National Academy <strong>of</strong><br />

Science <strong>of</strong> Ukraine, Kharkov, Ukraine, Maxim Ostankov,<br />

Institute for Problems <strong>of</strong> Cryobiology and Cryomedicine <strong>of</strong><br />

the National Academy <strong>of</strong> Science <strong>of</strong> Ukraine, Kharkov,<br />

Ukraine<br />

We have shown the possibility to reduce immune reactivity <strong>of</strong><br />

allogeneic bone marrow (BM) as graft-versus-host reaction<br />

(GVHR) by the change <strong>of</strong> the content <strong>of</strong> myelotransplant<br />

hemopoietic microenvironment cells. These changes may be<br />

achieved by additional introduction with BM <strong>of</strong> cells possessing<br />

regulatory activity. There is accumulated information about<br />

manifested immune modulating ability <strong>of</strong> fetal liver cells (FLCs).<br />

Research aim was the substantiation <strong>of</strong> the possible use <strong>of</strong> FLCs<br />

as the preparation: modulators <strong>of</strong> immune and hemopoietic<br />

activity <strong>of</strong> allogeneic bone marrow and a rise in its protective<br />

potential. Lethally irradiated Balb/c mice were intravenously<br />

injected BM <strong>of</strong> CBA ones (5×10 6 /mouse) together with FLC<br />

(5×10 5 /mouse). GVHR development intensity was assessed<br />

according to traditional parameters. Content <strong>of</strong> stem hemopoietic<br />

cells in BM <strong>of</strong> recipients was examined by cloning <strong>of</strong><br />

hemopoietic precursors in vivo (CFUs). Obtained results testify<br />

to the fact that co-transplanted FLCs rendered manifested<br />

regulatory effect on immune reactivity and hemopoietic


S106 Abstracts<br />

function <strong>of</strong> cryopreserved allogeneic BM. Character and extent<br />

<strong>of</strong> its manifestation have dose-dependent effect. FLC cotransplantation<br />

contributed to minimization <strong>of</strong> hypoplasia <strong>of</strong><br />

recipient’s thymus, increase <strong>of</strong> content in BM <strong>of</strong> recipients <strong>of</strong><br />

CFUs and finally to a rise in the survival percentage <strong>of</strong> the<br />

animals to the 70th day. Therefore the effect <strong>of</strong> FLC may be<br />

stipulated not only by the limitation <strong>of</strong> the extent <strong>of</strong><br />

allomyelotransplant GVHR activity but also by an increase in<br />

its hemopoietic potential. Mechanisms <strong>of</strong> FLC modulating<br />

activity in respect to co-transplanted BM are under discussion.<br />

doi:10.1016/j.clim.2007.03.479<br />

Sa.93 Suppression <strong>of</strong> Acute Graft-versus-host<br />

Disease by Regulatory T Cells Induced by Toxic<br />

Shock Syndrome Toxin-1<br />

Haowei Li, PhD Candidate, University <strong>of</strong> British Columbia,<br />

Department <strong>of</strong> Medicine, Vancouver, BC, Canada, Anthony<br />

W. Chow, Pr<strong>of</strong>essor Emeritus, University <strong>of</strong> British<br />

Columbia, Department <strong>of</strong> Medicine, Vancouver, BC, Canada<br />

Allogeneic bone marrow transplantation can cure multiple<br />

hematopoietic malignancies, but acute graft-versus-host<br />

disease (aGVHD) is a major complication. Previous studies in<br />

our laboratory have shown that chronic exposure to the<br />

staphylococcal superantigen, toxic shock syndrome toxin-1<br />

(TSST-1), can induce regulatory T cells (Tregs) in C57BL/6<br />

(B6) mice. These TSST-1-induced Tregs have potent suppressive<br />

ability to inhibit TSST-1 or SEA-induced pro-inflammatory<br />

cytokine responses in vitro and in vivo. However, it is<br />

unclear if TSST-1-induced Tregs can be applied clinically to<br />

control aGVHD. In this report, using a murine aGVHD model<br />

(B6 to B6D2F1), we showed that transplantation <strong>of</strong> an<br />

allogeneic graft containing TSST-1-induced Tregs alone did<br />

not confer protection against aGVHD. However, reactivation<br />

<strong>of</strong> TSST-1-induced Tregs by post-transplant exposure to TSST-<br />

1 led to attenuation <strong>of</strong> aGVHD. While one dose <strong>of</strong> TSST-1 only<br />

prolonged the survival time but not the survival rate <strong>of</strong><br />

recipients <strong>of</strong> TSST-1-induced Tregs, two doses <strong>of</strong> TSST-1 both<br />

prolonged the survival time and increased the survival rate <strong>of</strong><br />

the recipients. Activation <strong>of</strong> TSST-1-induced Tregs did not<br />

reduce engraftment or expansion <strong>of</strong> donor T cells, nor<br />

enhance the elimination <strong>of</strong> host antigen presenting cells, but<br />

was associated with decreased production <strong>of</strong> pro-inflammatory<br />

cytokines. Blockade <strong>of</strong> IL-10 did not reverse the<br />

suppression. More importantly, control <strong>of</strong> aGVHD by activation<br />

<strong>of</strong> TSST-1-induced Tregs did not interfere with graft-vs.tumor<br />

(GVT) effects. In summary, we conceptually demonstrated<br />

that Tregs induced by TSST-1 were able to control<br />

aGVHD in vivo via bystander suppression while sparing GVT<br />

effects.<br />

doi:10.1016/j.clim.2007.03.480<br />

Sa.94 Bone Marrow Derived Mesenchymal Stromal<br />

Cells Promote Treg Cell Development Ex Vivo<br />

Qingping Yao, Rheumatology Fellow, Medicine, University <strong>of</strong><br />

California, Los Angeles David Geffen School <strong>of</strong> Medicine, Los<br />

Angeles, CA, James Wang, Researcher, Medicine, University<br />

<strong>of</strong> California, Los Angeles David Geffen School <strong>of</strong> Medicine,<br />

Los Angeles, CA, Ram Raj Singh, Pr<strong>of</strong>essor, Medicine,<br />

University <strong>of</strong> California, Los Angeles David Geffen School <strong>of</strong><br />

Medicine, Los Angeles, CA<br />

Objective: Bone marrow derived mesenchymal stromal<br />

cells (BMSCs) provide niche for the development <strong>of</strong> hematopoietic<br />

cells. The BMSCs likely accomplish this via multiple<br />

cytokines. Here, we asked whether BMSCs affect the<br />

development <strong>of</strong> regulatory T (Treg) cells that can downregulate<br />

autoimmune diseases. Methods: Bone marrow cells<br />

isolated from lupus-prone (NZB×NZW) F1 (BWF1) and normal<br />

BALB/c mice were cultured in 24 well plates for 6 days. After<br />

removal <strong>of</strong> nonadherent cells in these cultures, adherent<br />

BMSCs were co-incubated with freshly isolated splenocytes<br />

from either BWF1 or BALB/c mice for an additional 6 days.<br />

Splenic cells harvested from these cultures were analyzed by<br />

flow cytometry (FACS) for CD4+CD25+ T lymphocytes. Cell<br />

culture supernatants were assayed for cytokines by ELISA.<br />

Results: We found a significantly higher number <strong>of</strong> CD4+<br />

CD25+ T lymphocytes in co-cultures <strong>of</strong> splenocytes plus<br />

BMSCs than in cultures <strong>of</strong> splenocyte alone. CD4+CD25+ T<br />

cells were found by FACS to be 8.5 and 178 fold higher in cocultures<br />

<strong>of</strong> splenocytes +BMSCs from BWF1 and BALB/c mice,<br />

respectively, over cultures <strong>of</strong> splenocytes alone. The<br />

increase in Treg cells was associated with increased IL-10<br />

production. Conclusion: BMSCs promote the development <strong>of</strong><br />

Treg cells, which is more efficient in normal mice than in<br />

autoimmune-prone mice. Thus, BMSC impairments might<br />

contribute to abnormalities in Treg cells in autoimmune<br />

conditions. Ongoing study will further characterize the Treg<br />

cell functions in our model, determine mechanisms by which<br />

BMSCs interact with Treg cells and examine the in vivo<br />

effects <strong>of</strong> BMSCs on Treg cells.<br />

doi:10.1016/j.clim.2007.03.481<br />

Sa.95 Selective Depletion <strong>of</strong> Alloreactive T Cells and<br />

Study <strong>of</strong> Anti-Tumor Activity <strong>of</strong> Specific T Cell<br />

Clones in Patients with Leukemia and Renal<br />

Carcinoma<br />

Eva Matejkova, PhD Student, Masaryk University, Zuzana<br />

Hrotekova, MSc., Cell Immunotherapy Center, Drahomira<br />

Kyjovska, Medical Laboratory Technician, Masaryk<br />

University, Czech Republic, Jaroslav Michalek, Head <strong>of</strong> Cell<br />

Immunotherapy Center, Masaryk University, Czech<br />

Republic, Petra Vidlakova, Lab Chief, Masaryk University,<br />

Czech Republic<br />

A major challenge in the field <strong>of</strong> hematopoietic stem<br />

cell transplantation (HSCT) is to prevent the alloreactivity<br />

<strong>of</strong> donor T cells which leads to acute graft-versus-host<br />

disease (GVHD) while preserving a graft-versus-tumor (GVT)<br />

effect. Selective depletion using anti-CD25 immunotoxin<br />

(IT) can eliminate harmful alloreactive T cells while<br />

preserving other donor T cells with antileukemic/antitumor<br />

reactivity. We have used irradiated peripheral blood<br />

mononuclear cells (PBMC) from cancer patients and healthy


Abstracts<br />

donor PBMC as responder cells in primary mixed leukocyte<br />

reaction (MLR). To prepare GVL/GVT-specific T cells,<br />

alloreactive T cells in primary MLR were depleted with<br />

anti-CD25 IT. The remaining T cells had insignificant<br />

alloreactivity in secondary MLR. Allodepleted donor cells<br />

were then repeatedly stimulated using purified leukemia/<br />

tumor cells from the same cancer patient. Leukemia/<br />

tumor-reactive donor T cells were purified by immunomagnetic<br />

separation on the basis <strong>of</strong> INF-g production. 17 MLRs<br />

(10 with leukemic and 7 with renal carcinoma cells) were<br />

performed. Selective depletion <strong>of</strong> alloreactive donor T cells<br />

with anti-CD25 IT led to more than 2log depletion. Graftversus-leukemia<br />

(GVL) effect <strong>of</strong> donor T cells was well<br />

preserved while the graft-versus-host (GVH) reactivation <strong>of</strong><br />

donor cells was negligible even after repeated stimulation<br />

with patient’s non-leukemic PBMC. In the case <strong>of</strong> renal<br />

carcinoma GVT effect was less dominant and GVH<br />

reactivation <strong>of</strong> donor cells was significant. In conclusion,<br />

it is possible to selectively deplete donor alloreactive T<br />

cells with anti-CD25 IT. In the case <strong>of</strong> patients with<br />

leukemia, the GVL effect can be separated from GVHD, but<br />

in case <strong>of</strong> renal carcinoma severe GVHD effect reappeared.<br />

Supported by the Ministry <strong>of</strong> Education <strong>of</strong> the<br />

Czech Republic, NPVII-2B06058.<br />

doi:10.1016/j.clim.2007.03.482<br />

Sa.96 CD28 Costimulates Suppression in an In Vitro<br />

Model <strong>of</strong> the Tumor Microenvironment<br />

Daniel Silberman, Student, Biology Department, Rider<br />

University, Lawrenceville, NJ, Amanda Bucknum, Student,<br />

Biology Department, Rider University, Lawrenceville, NJ,<br />

Tiffany Bloomfield, Student, Biology Department, Rider<br />

University, Lawrenceville, NJ, John Somerville, Associate<br />

Scientist, Bristol-Myers Squibb Pharmaceutical Research<br />

Institute, Princeton, NJ, Jessica Reid, Student, Biology<br />

Department, Rider University, Lawrenceville, NJ, James<br />

Riggs, Pr<strong>of</strong>essor <strong>of</strong> Biology, Biology Department, Rider<br />

University, Lawrenceville, NJ<br />

Although the cellular components essential for an<br />

effective immune response are <strong>of</strong>ten found in the tumor<br />

microenvironment they fail to prevent cancer progression.<br />

T cell activation, dependent upon costimulation provided<br />

by antigen-presenting cells (APCs), is restrained in tumors.<br />

Macrophages are <strong>of</strong>ten found in large numbers in tumors.<br />

An unbalanced macrophage:T cell ratio fosters T cell<br />

suppression. We have developed an in vitro model that<br />

mimics this characteristic <strong>of</strong> the tumor microenvironment.<br />

Murine peritoneal cavity (PerC) macrophages suppress T<br />

cell activation by indoleamine 2,3-dioxygenase (IDO) and<br />

inducible nitric oxide synthase (iNOS) catalyzed consumption<br />

<strong>of</strong> tryptophan and arginine. 1-Methyl tryptophan and<br />

NG-monomethyl-L-arginine block these enzymes and permit<br />

T cell activation. We are testing various costimulatory<br />

ligand–receptor combinations to determine if T cell<br />

activation can be rescued by treatment with an immunobiologic.<br />

Rather than promoting T cell activation, CD28<br />

ligation was found to promote macrophage-mediated<br />

suppression. The suppression was triggered by T cell<br />

production <strong>of</strong> IFN 3 . Treatment with CD28-Ig did not release<br />

suppression. These data will be discussed in the context <strong>of</strong><br />

strategies to curb the suppressive activity evident in tissues<br />

with aberrant macrophage:T cell ratios. Supported by NIH<br />

AREA grant R15-AI060356-01.<br />

doi:10.1016/j.clim.2007.03.483<br />

Sa.97 Development and In Vitro Validation <strong>of</strong><br />

Anti-Mesothelin Biobodies that Prevent CA125/<br />

Mesothelin-dependent Cell Attachment<br />

Nathalie Scholler, Senior Staff Scientist, Fred Hutchinson<br />

Cancer Research Center, Seattle, WA, Lindsay Bergan,<br />

Research Technician, Fred Hutchinson Cancer Research<br />

Center, Seattle, WA, Jennifer Gross, Research Technician,<br />

Fred Hutchinson Cancer Research Center, Seattle, WA, Barry<br />

Nevin, Research Technician, Fred Hutchinson Cancer<br />

Research Center, Seattle, WA, Barbara Garvik, Research<br />

Technician, Fred Hutchinson Cancer Research Center,<br />

Seattle, WA, Nicole Urban, Department head, Fred<br />

Hutchinson Cancer Research Center, Seattle, WA<br />

Preventing peritoneal implantation <strong>of</strong> carcinoma cells<br />

could prolong ovarian cancer patient remission and<br />

survival. Peritoneal cells constitutively express mesothelin,<br />

a ligand for CA125 that is expressed by tumor cells.<br />

Blocking CA125/mesothelin-dependent cell attachment<br />

may then prevent or delay peritoneal metastatic recurrence.<br />

We previously developed an in vitro cell adhesion<br />

assay combining cells expressing CA125 and mesothelintransfected<br />

cells. We demonstrated that CA125/mesothelin-dependent<br />

cell adhesion could be blocked with<br />

mesothelin recombinant protein fused to an Ig domain<br />

(meso-Ig) and cross-linked with anti-Ig antibodies, or with<br />

anti-CA125 mouse monoclonal antibodies (mAb) which<br />

suggests that new therapies based upon affinity agents<br />

able to compete with or to block the CA125/mesothelin<br />

interaction could prevent or delay the development <strong>of</strong><br />

peritoneal metastasis. However, mAbs are highly immunogenic<br />

in vivo. Recombinant antibodies are emerging as an<br />

attractive alternative to mouse antibodies for in vivo<br />

imaging and therapy. We have developed a new class <strong>of</strong><br />

recombinant antibodies, referred to as biobodies (Bbs)<br />

that are secreted by yeast as in vivo biotinylated proteins.<br />

Building on our previous work, we generated Bbs that<br />

specifically bind to mesothelin. Anti-mesothelin recognition<br />

sequences were isolated from a yeast-display scFv<br />

library by magnetic and flow sorting, using an in vivo<br />

biotinylated mesothelin recombinant protein also secreted<br />

by yeast. Anti-mesothelin yeast-display scFv were transformed<br />

into Bbs and validated by ELISA and flow<br />

cytometry. We demonstrated that the anti-mesothelin<br />

Bbs corresponding to the yeast-display scFv pool <strong>of</strong> highest<br />

affinity could recognize both membrane-bound and soluble<br />

mesothelins, and block CA125/mesothelin-dependent cell<br />

adhesion.<br />

doi:10.1016/j.clim.2007.03.484<br />

S107


S108 Abstracts<br />

Sa.98 Visualization and Characterisation <strong>of</strong> Melan A<br />

Epitope (25–35)/HLA-A2 Complex by High Affinity<br />

Soluble Monoclonal T Cell Receptors (mTCRs)<br />

Samantha Paston, Avidex Ltd, Abingdon, Oxfordshire,<br />

England, Katherine Adams, Ms, Avidex Ltd, Abingdon,<br />

Oxfordshire, England, Giovanna Bossi, Avidex Ltd,<br />

Abingdon, Oxfordshire, England, Nathaniel Liddy, Avidex<br />

Ltd, Abingdon, Oxfordshire, England, Deborah Sutton,<br />

Avidex Ltd, Abingdon, Oxfordshire, England, Tara Mahon,<br />

Avidex Ltd, Abingdon, Oxfordshire, England, Peter Molloy,<br />

Avidex Ltd, Abingdon, Oxfordshire, England, Emma<br />

Gostick, Avidex Ltd, Abingdon, Oxfordshire, England, Andy<br />

Sewell, Department <strong>of</strong> Medical Biochemistry and<br />

<strong>Immunology</strong>, Cardiff, England, Bent Jakobsen, Avidex Ltd,<br />

Abingdon, Oxfordshire, England<br />

Cytotoxic T cells (CTL) use TCRs to detect tumor and viral<br />

antigens. Soluble TCRs are potential tools for therapeutic<br />

treatment <strong>of</strong> cancer, viral infections and autoimmune diseases.<br />

The Melan A protein (MART-1) was one <strong>of</strong> the first tumour<br />

associated antigens to be identified, and is over-expressed in<br />

80–100% <strong>of</strong> melanomas. CTL’s recognising the heterolytic<br />

ELAGIGILTV peptide can be found in both healthy donors and<br />

melanoma patients. Adoptive therapy and vaccination trials<br />

using the ELAGIGILTV peptide have resulted in some good<br />

clinical responses with partial or complete regression <strong>of</strong> some<br />

metastates. In healthy donors, up to 0.1% <strong>of</strong> Tcells in peripheral<br />

blood can be stained with the Melan A tetramer; patients have a<br />

higher frequency <strong>of</strong> Melan A specific Tcells where 1–15% <strong>of</strong> the T<br />

cellscanbestainedwiththetetramer.TheMelanAmTCRwas<br />

affinity matured to bind HLA-A2 presenting the heteroclytic<br />

ELAGIGILTVpeptidewithakDa<strong>of</strong>10pMandahalflife<strong>of</strong>67h.<br />

Here we describe the generation <strong>of</strong> a Tcell clone specific for the<br />

HLA-A2/Melan A epitope (26–35) derived from a healthy blood<br />

donor. Using this Melan A (26–35) mTCR we can specifically block<br />

cytokineproductionfromaMelanATcellclone.Inadditionwe<br />

are also able to detect and quantify the endogenous peptide<br />

processed and presented by melanoma cell lines using 3D<br />

fluoresence microscopy and flow cytometry. Visualisation and<br />

quantification <strong>of</strong> these specific antigen/MHC complexes enable<br />

the potential <strong>of</strong> mTCRs recognising these complexes as anticancer<br />

therapeutics to be evaluated.<br />

doi:10.1016/j.clim.2007.03.486<br />

Sa.100 AdCD40L Immunogene Therapy for Bladder<br />

Carcinoma<br />

Moa Fransson, PhD Student, <strong>Clinical</strong> <strong>Immunology</strong> Division,<br />

Uppsala, Sweden, Angelica Loskog, Scientist, <strong>Clinical</strong><br />

<strong>Immunology</strong> Division, Uppsala, Sweden, Thomas Tötterman,<br />

Pr<strong>of</strong>esor, <strong>Clinical</strong> <strong>Immunology</strong> Division, Uppsala, Sweden<br />

Recently regulatory T cells (Treg) have made a comeback in<br />

the immunological arena and the role <strong>of</strong> these cells in cancer is<br />

in focus. <strong>Clinical</strong> protocols that effectively counteract Tregmediated<br />

immunosuppression are in high demand. We developed<br />

an immunostimulatory gene therapy for urinary bladder<br />

cancer based on CD40L gene transfer into the tumor site. The<br />

efficacy <strong>of</strong> this therapy was evaluated in mouse and human<br />

experimental models <strong>of</strong> bladder cancer. The CD40L gene was<br />

transferred to the bladder wall <strong>of</strong> tumor-bearing mice by<br />

adenoviral vector instillation. AdCD40L gene therapy cured 60%<br />

<strong>of</strong> mice with pre-established aggressive tumors. Cured mice<br />

were completely resistant to s.c. challenge with wt tumor. The<br />

mRNA levels <strong>of</strong> the Treg transcription factor Foxp3 were<br />

measured in tumor biopsies and lymph nodes. There were no<br />

differences within the tumors <strong>of</strong> the different treatment groups.<br />

However, Foxp3 mRNA levels were down-regulated in the lymph<br />

nodes <strong>of</strong> AdCD40L treated mice. Human bladder cancer cell line<br />

cells were transduced with AdCD40L vector and used to<br />

stimulate immune cells. Cytokine and immune cell pr<strong>of</strong>iling<br />

indicated a Th1 type response. The AdCD40L-modified cell lines<br />

stimulated maturation <strong>of</strong> dendritic cells and cytotoxic Tcells as<br />

opposed to wt tumor cells. In conclusion, CD40L gene transfer<br />

evokes Th1 cytokine responses and counteracts Tregulatory cell<br />

development and/or function in the preclinical setting. We are<br />

currently conducting a phase I/II trial with AdCD40L in patients<br />

with bladder cancer.<br />

doi:10.1016/j.clim.2007.03.487<br />

Sa.101 Functional T Cell Responses to Tumor<br />

Antigens in Breast Cancer Patients Have a Unique<br />

Phenotype and Cytokine Signature<br />

Margaret Inokuma, Scientist, BD Biosciences, Immune<br />

Function, San Jose, CA, Corazon dela Rosa, Research<br />

Associate, University <strong>of</strong> Washington, Seattle, WA, Perry<br />

Haaland, BD Fellow, BD Technologies, Research Triangle<br />

Park, NC, Douglas Petry, patent attorney, BD Biosciences,<br />

San Jose, CA, Janet Siebert, president, CytoAnalytics,<br />

Denver, CO, MengXiang Tang, Senior Algorithm Developer,<br />

BD Biosciences, San Jose, CA, John Dunne, Associate<br />

Director, BD Biosciences, San Jose, CA, Vernon Mai, Director,<br />

BD Biosciences, San Jose, CA, Mary Disis, Associate<br />

Pr<strong>of</strong>essor, University <strong>of</strong> Washington, Department <strong>of</strong><br />

Oncology, Seattle, WA, Holden Maecker, Research Grp<br />

Manager, BD Biosciences, San Jose, CA<br />

The prevalence with which endogenous tumor antigens<br />

induce host T cell responses is unclear. Even when such<br />

responses are detected, they do not usually result in<br />

spontaneous remission <strong>of</strong> the cancer. We hypothesized that<br />

this might be associated with a predominant phenotype and/or<br />

cytokine pr<strong>of</strong>ile <strong>of</strong> tumor-specific responses that is different<br />

from protective Tcell responses to other chronic antigens, such<br />

as cytomegalovirus (CMV). We detected significant T cell<br />

responses to CEA, HER-2/neu, and/or MAGE-3 in 17 <strong>of</strong> 21<br />

breast cancer patients naive to immunotherapy. The pattern <strong>of</strong><br />

T cell cytokines produced in response to tumor-associated<br />

antigens (TAAs) in breast cancer patients was significantly<br />

different from that produced in response to CMV in the same<br />

patients. Specifically, there was a higher proportion <strong>of</strong> IL-2<br />

producing CD8+ T cells, and a lower proportion <strong>of</strong> IFN γproducing<br />

CD4+ and CD8+ T cells responding to TAAs compared<br />

to CMV antigens. Finally, the phenotype <strong>of</strong> TAA-responsive CD8+<br />

T cells in breast cancer patients was almost completely CD28<br />

+CD45RA− (central memory phenotype). CMV-responsive CD8+<br />

Tcellsinthesamepatientswerebroadlydistributedamong<br />

phenotypes, and contained a high proportion <strong>of</strong> terminal


Abstracts<br />

effector cells (CD27−CD28−CD45RA+) that were absent in the<br />

TAA responses. Taken together, these results suggest that TAAresponsive<br />

T cells are induced in breast cancer patients, but<br />

those Tcells are phenotypically and functionally different from<br />

CMV-responsive Tcells. Immunotherapies directed against TAAs<br />

may need to alter these T cell signatures in order to be<br />

effective.<br />

doi:10.1016/j.clim.2007.03.488<br />

Sa.102 Dendritic Cell-based Therapy for Mantle Cell<br />

Lymphoma<br />

Corey Munger, Graduate Student, Nebraska Medical Center,<br />

Genetics, Cell Biology, and Anatomy, Omaha, NE, Ganapati<br />

Hegde, Postdoctoral Fellow, Nebraska Medical Center,<br />

Genetics, Cell Biology, and Anatomy, Omaha, NE, Julie Vose,<br />

Pr<strong>of</strong>essor, Internal Medicine, Omaha, NE, Shantaram Joshi,<br />

Pr<strong>of</strong>essor, Nebraska Medical Center, Genetics Cell Biology,<br />

and Anatomy, Omaha, NE, Dennis Weisenburger, Pr<strong>of</strong>essor,<br />

Pathology and Microbiology, Omaha, NE<br />

Mantle cell lymphoma (MCL) is an incurable B cell<br />

malignancy due to the development <strong>of</strong> therapy-resistant<br />

cells. The ability <strong>of</strong> dendritic cells (DC) to stimulate tumorspecific<br />

cytotoxic T lymphocytes (CTL) makes DC-based<br />

therapy a promising strategy to treat therapy-resistant MCL.<br />

Therefore, we investigated the efficacy <strong>of</strong> DC-based immunotherapy<br />

to treat resistant MCL. Specifically, DCs loaded<br />

with MCL lysate, DCs fused with MCL cells, and DCs<br />

electroporated with MCL RNA were used to generate<br />

tumor-specific CTLs against the MCL cell line, Granta 519.<br />

CTL-mediated cytotoxicities at a 50:1 effector-to-target<br />

ratio using each method <strong>of</strong> DC priming are as follows: 58.4<br />

±12.7% (MCL lysate-pulsed), 54.0±8.3% (MCL RNA-transfection),<br />

and 59.3±8.4% (DC-MCL hybrid). Cytotoxicity toward<br />

the irrelevant HLA-matched cell line MDA-231 was 28.7±<br />

3.6%. To assess the in vivo cytotoxic abilities <strong>of</strong> CTLs<br />

stimulated by DC-MCL hybrids, NOD/SCID mice transplanted<br />

intravenously with 1×106 Granta 519 cells were treated with<br />

four weekly injections <strong>of</strong> 3×106 HLA-matched CTLs. Histological<br />

analysis revealed that 66% <strong>of</strong> the control mice at week<br />

five displayed significant tumor growth in the liver and<br />

kidneys versus only 16% in the treatment group. In addition to<br />

reducing the number <strong>of</strong> tumor nodules, the average size <strong>of</strong><br />

the nodules was decreased in treated mice versus the<br />

untreated control mice. These studies demonstrate each<br />

method <strong>of</strong> DC priming stimulated tumor-specific CTLs in vitro<br />

and administration <strong>of</strong> DC-MCL hybrid-stimulated CTLs inhibited<br />

tumor formation in vivo. This study was supported by<br />

the Lymphoma Research Foundation, New York, NY.<br />

doi:10.1016/j.clim.2007.03.489<br />

Sa.103 Targetting Sonic Hedgehog-GLI Signaling for<br />

the Treatment <strong>of</strong> Mantle Cell Lymphoma<br />

Ganapati Hegde, Postdoctoral Research Associate,<br />

Department <strong>of</strong> Genetics, Cell Biology and Anatomy, Omaha,<br />

NE, Corey Munger, Graduate Student, Department <strong>of</strong><br />

Genetics, Cell Biology and Anatomy, Omaha, NE, Katy<br />

Emanuel, Summer Student, Department <strong>of</strong> Genetics, Cell<br />

Biology and Anatomy, Omaha, NE, Dennis Weisenburger,<br />

Pr<strong>of</strong>essor, Pathology and Microbiology, Omaha, NE, Avadhut<br />

Joshi, Graduate student, Department <strong>of</strong> Genetics, Cell<br />

Biology and Anatomy, Omaha, NE, Julie Vose, Pr<strong>of</strong>essor,<br />

Internal Medicine, Oncology/Hematology, Omaha, NE,<br />

Shantaram Joshi, Pr<strong>of</strong>essor, Department <strong>of</strong> Genetics, Cell<br />

Biology and Anatomy, Omaha, NE<br />

Mantle cell lymphoma (MCL) has one <strong>of</strong> the worst clinical<br />

outcomes among the B cell lymphomas with a median<br />

survival <strong>of</strong> only 3–4 years. Particularly, the blastoid variant<br />

has a poorer prognosis compared to classical MCL. Therefore,<br />

a better understanding <strong>of</strong> the underlying mechanisms that<br />

regulate MCL proliferation and survival is clearly needed.<br />

Since, sonic hedgehog (Shh)-GLI signaling has been shown to<br />

be important in the proliferation/survival <strong>of</strong> several cancers,<br />

and no such information is available in any B cell malignancies<br />

including MCL, this study was undertaken. Our<br />

results demonstrate that the molecules associated with Shh<br />

signaling (PTCH, SMO, GLI) are considerably expressed in<br />

classical, blastoid and patients’ primary MCL. Overexpression<br />

<strong>of</strong> PTCH and GLI1, the Shh-GLI signaling target genes, in<br />

blastoid versus classical MCL suggests constitutive expression<br />

<strong>of</strong> these genes in blastoid variant. Classical, but not the<br />

blastoid MCL cells responded (pb0.01) to perturbation <strong>of</strong> Shh<br />

signaling in the presence <strong>of</strong> exogenous Shh/cyclopamine.<br />

Furthermore, down-regulation <strong>of</strong> GLI transcription factors<br />

using anti-sense oligonucleotides resulted in significantly<br />

(pb0.001) decreased proliferation <strong>of</strong> both blastoid and<br />

classical MCL, and significantly (pb0.001) increased their<br />

susceptibility to chemotherapy. In addition, down-regulation<br />

<strong>of</strong> GLI by anti-sense ODN decreased Cyclin D1 (CCND1) and<br />

BCL2 transcript levels, which suggests that these key<br />

molecules might be regulated by GLI in MCL. Thus, these<br />

results suggest a significant role for Shh-GLI signaling in the<br />

proliferation <strong>of</strong> MCL, and targeting GLI as therapeutic<br />

approach to treat MCL effectively is very promising (this<br />

study was supported by the Lymphoma Research Foundation,<br />

New York).<br />

doi:10.1016/j.clim.2007.03.490<br />

S109<br />

Sa.104 The Role <strong>of</strong> Thrombospondin-1 in Driving<br />

Regulatory T Cell Generation Following<br />

Extracorporeal Photopheresis<br />

Amy Krutsick, Senior Scientist, Therakos, Inc. RandCD,<br />

Exton, PA, Peter O’Brien, Principal Scientist, Therakos, Inc.<br />

RandCD, Exton, PA, Ryan Gailey, Research Associate I,<br />

Therakos, Inc. RandCD, Exton, PA, Kim Campbell, Principal<br />

Research Scientist, Therakos, Inc. RandCD, Exton, PA, Frank<br />

Strobl, Director <strong>of</strong> Scientific Affairs, Therakos, Inc. RandCD,<br />

Exton, PA<br />

Extracorporeal photopheresis (ECP) is an immune cell<br />

therapy approved for the palliative treatment <strong>of</strong> cutaneous<br />

T cell lymphoma and has demonstrated activity in<br />

immune-mediated inflammatory diseases. During ECP,


S110 Abstracts<br />

peripheral blood leukocytes are treated ex vivo with 8methoxypsoralen<br />

and UVA light, rendering cells apoptotic.<br />

Reinfusion <strong>of</strong> ECP-treated cells modulates immune<br />

responses through the generation <strong>of</strong> tolerogenic dendritic<br />

cells and T regulatory cells (Tregs). We previously<br />

demonstrated that ECP-treated peripheral blood mononuclear<br />

cells (PBMCs) co-cultured directly with T cells<br />

result in the generation <strong>of</strong> a regulatory T cell population.<br />

ECP-generated Tregs can suppress a syngeneic T cell<br />

response in a contact-dependent fashion by greater than<br />

70%. These Tregs demonstrate a modest increase in Foxp3<br />

expression and exhibit an anergic phenotype upon in vitro<br />

re-stimulation. We found that monocytes are required for<br />

ECP-mediated Treg generation and apoptotic ECP-treated<br />

monocytes secrete a significant level (N500 ng/ml) <strong>of</strong><br />

Thrombospondin-1 (TSP-1). TSP-1 is an extracellular<br />

matrix protein with anti-angiogenic properties shown by<br />

others to have immunomodulatory effects on T cells<br />

through interactions with CD47. We hypothesized that<br />

TSP-1 is involved in the generation <strong>of</strong> Tregs via ECP cells.<br />

To confirm this hypothesis, TSP-1 activity was blocked<br />

through addition <strong>of</strong> anti-CD47 to cultures containing ECPtreated<br />

PBMCs and T cells. Blocking this TSP-1 receptor on<br />

T cells inhibited ECP-mediated Treg generation. Additional<br />

studies are in progress to further evaluate the role <strong>of</strong> TSP-<br />

1 in the mechanism <strong>of</strong> action <strong>of</strong> ECP. TSP-1 may be<br />

important for the clinical benefit <strong>of</strong> ECP in a variety <strong>of</strong><br />

pathophysiological disorders.<br />

doi:10.1016/j.clim.2007.03.491<br />

Sa.105 Cytotocix Effect <strong>of</strong> Total Saffron Extract on<br />

Human Hepatocell Carcinoma (HCC)<br />

Jalil Tavakkol-Afshari, Vice President for Research, Head,<br />

Department <strong>of</strong> Immunogenetics and Cell Culture, Bu-Ali<br />

Research Institute (BARI), Department <strong>of</strong> Immunogenetics<br />

and Cell Culture, Mashhad, Iran, Khadijeh Nejad<br />

Shahrokhabadi, Researcher, Bu-Ali Research Institute<br />

(BARI), Department <strong>of</strong> Immunogenetics, Mashhad, Iran,<br />

Hassan Rakhshandeh, Pharmacologist, School <strong>of</strong><br />

Pharmocology, Mashhad University <strong>of</strong> Medical Sciences,<br />

Mashhad, Iran, Azam Brook, Researcher, Bu-Ali Research<br />

Institute (BARI), Department <strong>of</strong> Immunogenetics, Mashhad,<br />

Iran<br />

Saffron – dried stigma <strong>of</strong> Crocus Sativus L. – is used as a<br />

nutritional spice and dry additive in cooking. The antitumor<br />

effect <strong>of</strong> saffron extract has been proved both in vitro and<br />

in vivo during recent years. In this research, cytotoxic<br />

effect <strong>of</strong> total saffron aqueous extract on human Hepatocell<br />

Carcinoma (HepG2) and mouse fibroblastic cells (L929)<br />

(as controls) in vitro has been investigated. Effects <strong>of</strong><br />

extract on quantitative proliferation <strong>of</strong> each cell line were<br />

determined using (MTT) colorimetric assay. MTT assay is a<br />

fast, sensitive and quantitative method for all kinds <strong>of</strong> cell<br />

proliferation by spectrophotometry. Different amounts <strong>of</strong><br />

extract were used and results showed that the 50%<br />

inhibition occurred in tumoral cells at the concentration<br />

<strong>of</strong> 400 microgram/milliliter that had no inhibitory effects<br />

on normal cells. According to our findings, the effect <strong>of</strong><br />

saffron extract may be useful in vivo against hepatic tumoral<br />

cells.<br />

doi:10.1016/j.clim.2007.03.492<br />

Sa.106 Optimizing Peptide Loading <strong>of</strong> Dendritic<br />

Cells Using TCRm Antibodies<br />

Tffany Nguyen, Senior Research Technician, Receptor Logic,<br />

Ltd. Amarillo, TX, Olivier Faure, Scientist, IDM, Inc. Irvine,<br />

CA, Francisca Neethling, Scientist, Receptor Logic, Ltd.<br />

Amarillo, TX, Roy Guillermo, Scientist, IDM, Inc. Irvine, CA,<br />

Sun min Lee, Scientist, IDM, Inc. Irvine, CA, James Bender,<br />

Scientist, IDM, Inc. Irvine, CA, Jon A. Weidanz, Assistant<br />

Pr<strong>of</strong>essor, Texas Tech University Health Sciences Center,<br />

Amarillo, TX<br />

Dendritic cells (DCs) represent potential vaccine vehicles<br />

for the treatment <strong>of</strong> cancer. Peptide-loaded DC’s require no<br />

protein processing and are amendable for development <strong>of</strong><br />

“personalized” vaccines. Conditions for peptide-loading DC<br />

are poorly described and have not yet been optimized. We<br />

examined peptide-loading conditions for DC using a TCRm<br />

antibody that specifically recognizes a peptide<br />

(TMTRVLQGV40-48) presented by HLA-A*0201 derived from<br />

the tumor-associated antigen human chorionic gonadotropin<br />

beta (hCGb). Characterization <strong>of</strong> the 3F9 TCRm included<br />

ELISA and flow cytometric analysis and demonstrated specific<br />

binding to peptide–HLA-A2 tetramer and T2 cells loaded with<br />

the TMT peptide. Moreover, the 3F9 TCRm displayed<br />

detection sensitivity in the picomolar range. TCRm staining<br />

<strong>of</strong> DCs was shown to be dependent on peptide concentration<br />

with maximum and minimum mean fluorescence intensity<br />

(MFI) reported at 80 μM and 10 μM peptide, respectively. We<br />

next examined the relationship between pulsing time and<br />

peptide presentation. Immature DCs were pulsed with 20 μM<br />

peptide for 1, 2, 4, 6, 8 and 22 h followed by cytokine-induced<br />

maturation during the final six hours. DC pulsed for shorter<br />

periods <strong>of</strong> time displayed TCRm staining with higher MFI<br />

values, suggesting that longer pulse times decrease peptide-<br />

HLA presentation. DCs were then pulsed with a peptide<br />

cocktail (TMT+6 other peptides) to determine TMT peptide<br />

loading efficiency. Similar TCRm staining intensities were<br />

observed for DC pulsed with TMT peptide alone and cells<br />

pulsed with the peptide cocktail. These findings suggest that<br />

TCRm may be useful tools for the development and<br />

standardization <strong>of</strong> peptide-pulsed DC vaccines.<br />

doi:10.1016/j.clim.2007.03.493<br />

Sa.107 Phase I Study <strong>of</strong> the Anti-MUC-1 Antibody,<br />

huHMFG1 (AS1402), in Patients with Advanced<br />

Breast Cancer<br />

Nigel Courtenay-Luck, Chief Scientific Officer, Antisoma<br />

Research Ltd, London, England, Mark Pegram, Associate<br />

Pr<strong>of</strong>essor <strong>of</strong> Medicine, David Geffen School <strong>of</strong> Medicine, Los<br />

Angeles, CA<br />

Background: A 20 amino acid MUC-1 core tandem repeat<br />

is aberrantly glycosylated in N90% <strong>of</strong> epithelial tumors,


Abstracts<br />

exposing the peptide sequence PDTRP. Humanized IgG1 K<br />

monoclonal antibody huHMFG1 (AS1402, formerly R1550),<br />

binds to PDTRP (Kd ∼3 nM) and, in vitro, induces potent<br />

antibody dependent cellular cytotoxicity (ADCC). This study<br />

was designed to evaluate the safety, tolerability and<br />

recommended dose <strong>of</strong> huHMFG1 in patients with advanced<br />

breast cancer. Methods: 26 patients were enrolled with<br />

locally advanced or metastatic breast cancer, which was<br />

progressive following up to 3 prior chemotherapy regimens.<br />

Patients were randomly assigned to 4 cohorts, each<br />

allocated to receive huHMFG1 at a dose <strong>of</strong> 1, 3, 9, or<br />

16 mg/kg, which was administered via I.V. infusion with<br />

repeated weekly dosing until disease progression. <strong>Clinical</strong><br />

and laboratory analysis was performed regularly. Results:<br />

The maximum tolerated dose (MTD) <strong>of</strong> huHMFG1 was not<br />

reached in this study and no serious adverse events were<br />

reported. No anti-huHMFG1 antibodies were detected.<br />

Response data are available from 22 patients with the<br />

best recorded clinical response, using RECIST criteria, being<br />

stable disease. Time to tumor progression ranged between<br />

28 and 119 days. Conclusion: huHMFG1 appears to be well<br />

tolerated with the MTD exceeding the doses tested. This<br />

study showed cases <strong>of</strong> prolonged stable disease among<br />

breast cancer patients who had relapsed following chemotherapy,<br />

suggesting that evaluation <strong>of</strong> huHMFG1 in phase<br />

II trials is warranted.<br />

doi:10.1016/j.clim.2007.03.494<br />

Sa.108 30 Years from Creation <strong>of</strong> the First<br />

Cytochemical Method Analysing RNP, DNP and<br />

Cationic Proteins in Circulating Leukocytes <strong>of</strong><br />

Cancer Patients as a Tool for Early Diagnostics <strong>of</strong><br />

Malignancies<br />

Elissaveta Borissova Zvetkova, Associated Pr<strong>of</strong>essor, IEMAM,<br />

S<strong>of</strong>ia, Bulgaria, Georgi Stefanov Kostov, Head <strong>of</strong> Surgery<br />

Department, Oncological Hospital, Veliko Tarnovo, Yordanka<br />

Georgieva Gluhcheva, Research Associate, IEMAM, Cell<br />

Differentiation, S<strong>of</strong>ia, Bulgaria<br />

Background: In our efforts to analyse cellular features <strong>of</strong><br />

peripheral blood leukocytes in cancer patients and tumorbearing<br />

animals, we developed (30 years ago) an accurate<br />

method for early detection <strong>of</strong> malignancies (1–4). Methods:<br />

We combined our cytological/cytochemical method for<br />

simultaneous visualization <strong>of</strong> nucleoproteins (RNP and DNP)<br />

and cationic proteins with ultrastructural and cytophotometrical<br />

techniques. Results and Discussion: Examining a panel<br />

<strong>of</strong> activation/deactivation cell markers in peripheral blood<br />

leukocytes <strong>of</strong> cancer and precancerous patients, we observed<br />

specific abnormalities: (1) Quantitative reduction <strong>of</strong> the<br />

cytoplasmic (ribosomal) RNP-content in the peripheral blood<br />

mononuclears T-lymphocytes and monocytes, as an additional<br />

tool for early cancer diagnostics; (2) Changes in the<br />

quantity, condensation and distribution <strong>of</strong> DNP in the nuclear<br />

chromatin <strong>of</strong> mono-and polymorphonuclear leukocytes as<br />

cytological signs for tumor-microenvironment-induced transcriptional<br />

arrest and apoptosis. (3) Polymorphism in size,<br />

staining properties and high extractibility <strong>of</strong> cytoplasmic<br />

(cationic proteins’ containing) granules <strong>of</strong> circulating poly-<br />

morphonuclear granulocytes—neutrophils and eosinophils.<br />

Today, 30 years later, our old hypothesis that cancer is not<br />

local, but systemic nuclear chromatin disease altering<br />

peripheral blood cell genome expression is in very good<br />

agreement with recent data based on gene expression<br />

patterns in peripheral blood cells <strong>of</strong> cancer patients (5) as<br />

monitors for early detection <strong>of</strong> malignant disease elsewhere<br />

in the body. References: (1) Acta histochem., 57, 1976, 1. (2)<br />

Folia Haematol., 106/2, 1979, 205. (3) Arch. Union Med Balk.,<br />

XX/1–2, 1984, 140. (4) Acta morphol. and anthropol., 5,<br />

2000, 3. (5) Breast Cancer Res., 7/5, 2005, R634.<br />

doi:10.1016/j.clim.2007.03.495<br />

Sa.109 Human Lung Tumor Cells Modifies Rates <strong>of</strong><br />

CD4+CD25+ T Regulatory, CD19+CD23+ B Regulatory<br />

Cells, CD3+CD95+ Cells, NK Cells and B Lymphocytes<br />

Bayram Kiran, Tip Fakultesi, Temel Bilimler Binasi, Istanbul,<br />

Turkey, Akif Turna, Yedikule Hospital for Chest Diseases and<br />

Thoracic Surgery, Kadikoy, Istanbul, Turkey, Alper Yener,<br />

Istanbul Tip Fakultesi, Istanbul, Turkey, Atilla Gürses,<br />

Yedikule Gogus Hastaliklari Hastanesi, Istanbul, Turkey,<br />

Andac Salman, Istanbul Tip Fakultesi, Istanbul, Turkey,<br />

Selim Badur, Istanbul Tip Fakultesi, Temel Bilimler Binasi,<br />

Istanbul, Turkey<br />

The role <strong>of</strong> T regulatory cells and NK cells in human lung<br />

cancer has not yet been clarified. Our aim was to evaluate<br />

how the microenvironment <strong>of</strong> a tumor mass induced phenotypical<br />

changes in lymphocyte subsets. Our subjects were<br />

10 patients with resectable non-small cell lung cancer. We<br />

evaluated 47 different phenotypically different lymphocyte<br />

subsets in blood samples taken from the pulmonary artery,<br />

pulmonary vein <strong>of</strong> a tumor bearing pulmonary lobe and<br />

peripheral blood during pulmonary resectional surgery. We<br />

showed that, tumor cells boosted CD25high+CD4high+T<br />

lymphocytes (Treg cells), B lymphocytes, NK and CD19+<br />

CD23+ cells (Breg cells) and CD3+CD95+ (FasL+T lymphocytes)<br />

(p=0.015, p=0.001, p=0.02, p=0.04, p=0.03 respectively).<br />

However, Mac-1 cells and HLADR+T lymphocytes<br />

were found to be decreased by tumor mass (p=0.04 and<br />

p=0.04), whereas only Breg, NK cell and B lymphocyte<br />

subsets were found to be expansed. In addition, the patients<br />

showed expansions <strong>of</strong> CD45R0+ activated T lymphocytes and<br />

CD18+CD11b+(Mac-1) cell subsets without significant alteration<br />

by tumor microenvironment. In conclusion, subsets <strong>of</strong><br />

Treg, Breg cells, FasL+ T lymphocytes, B lymphocytes were<br />

modified by lung cancer microenvironment itself. This study<br />

also showed that, peripheral blood might not be good source<br />

for investigating the anti-tumor immunity since only Breg, NK<br />

cell and Treg cell subsets were found expansed peripherally.<br />

T lymphocytes and Mac-1 cells may be modified by systemic<br />

antitumor response rather than by local tumoral microenvironment.<br />

This study provides important insight into how<br />

tumor infiltrating lymphocytes and peripheral immune<br />

systems may be different in terms <strong>of</strong> lymphocyte subset<br />

expansion dynamics.<br />

doi:10.1016/j.clim.2007.03.496<br />

S111


S112 Abstracts<br />

Sa.110 ABCB5 Gene is Expressed in Acral Melanoma<br />

<strong>of</strong> Poor Prognosis<br />

Norma Herrera-Gonzàlez, Pharmaceutical Industrial<br />

Chemest, Postgraduate Division, Mexico, Ismael Vasquez,<br />

Medical Doctor, Postgraduate Division, Mexico, Marco<br />

Antonio Meraz-Rios, Pharmacobiology Chemist, Biomedicine<br />

Department, CINVESTAV, Mexico, Cleva Villanueva, Medical<br />

Doctor, Postgraduate Division, Mexico<br />

Cutaneous melanomais a malignant neoplasm characterized<br />

by a high risk <strong>of</strong> metastasis disease and death.<br />

Metastatic melanoma displays strong resistance against<br />

antineoplastic drugs. Combination <strong>of</strong> chemotherapy and<br />

radiotherapy has been tried in patients and the best response<br />

rates have been less than 20%. The dramatic increase in the<br />

incidence and mortality <strong>of</strong> melanoma highlights the need for<br />

improved methods <strong>of</strong> diagnosis as well as a better understanding<br />

<strong>of</strong> basic concepts <strong>of</strong> some types <strong>of</strong> melanoma that<br />

have been scarcely studied as Acral Melanoma (AM), this<br />

work describes a molecular screen for differentially<br />

expressed genes in AM, the most common melanoma in<br />

Hispanic population. We used differential display reverse<br />

transcription-polymerase chain reaction and compared the<br />

gene expression pattern between primary melanomas and<br />

normal skin. In this work we fond an overexpression <strong>of</strong> the<br />

ABCB5 gene in melanoma tissue. ABCB5 has been recently<br />

characterized in normal human melanocytes and has been<br />

implicated in the regulation <strong>of</strong> progenitor cell fusion. ABCB5<br />

amplification bands were detected in nine <strong>of</strong> ten melanoma<br />

tissues by using RT-PCR. In situ hybridization using specific<br />

probes showed the presence <strong>of</strong> this gene in paraffin<br />

melanoma tissues. We suggest that this event might be<br />

related to metastatic melanoma behavior.<br />

doi:10.1016/j.clim.2007.03.497<br />

Sa.111 MHC Class I and MICA Expressions in Liver<br />

Fluke Associated Cholangiocarcinoma<br />

Wachanan Wongsena, Lecturer, Biomedical Sciences<br />

Program, Graduate School, Khon Kaen University, Khon<br />

Kaen, Thailand, Solda Ferrone, Educator/Research Director,<br />

Roswell Park Cancer Institute, Buffalo, NY, Richard Cheney,<br />

Pathologist/Dermatopathologist, Roswell Park Cancer<br />

Institute, Buffalo, NY, Chanvit Leelayuwat, Educator, The<br />

Centre for Research and Development <strong>of</strong> Medical Diagnostic<br />

Laboratories, Khon Kaen University, Khon Kaen, Thailand,<br />

Banchob Sripa, Educator, Liver Fluke and<br />

Cholangiocarcinoma Research Center, Khon Kaen, Thailand<br />

Alterations <strong>of</strong> HLA class I antigens and MHC class I chain<br />

related gene A (MICA), a ligand for activating receptor,<br />

NKG2D, have been described in several types <strong>of</strong> tumors. Such<br />

a study in cholangiocarcinoma has not been reported. A total<br />

<strong>of</strong> 102 paraffin-embedded cholangiocarcinoma lesions were<br />

stained by mouse anti-HLA class I heavy chain, anti-β-2microglobulin,<br />

anti-Tapasin, anti-TAP1, anti-TAP2, anti-<br />

LMP2, anti-LMP7, and three <strong>of</strong> anti-MICA monoclonal antibodies<br />

using immunoperoxidase reaction. HLA class I heavy<br />

chain, β-2-microglobulin, Tapasin, TAP1, TAP2, LMP2 and<br />

LMP7 were lost in 12.7, 5.9, 6.9, 2.0, 1.0, 6.9 and 8.8% <strong>of</strong> the<br />

lesions tested, respectively. Although most <strong>of</strong> the lesions<br />

expressed HLA class I heavy chain and β-2-microglobulin, the<br />

expression was restricted to the cytoplasm in 68.5 and 64.6%<br />

<strong>of</strong> the lesions, respectively. Loss <strong>of</strong> HLA class I heavy chain<br />

expression was associated with late stage (p=0.034) and<br />

poor differentiation <strong>of</strong> tumors (p=0.014). Loss <strong>of</strong> LMP7<br />

expression was also associated with poor differentiation <strong>of</strong><br />

tumors and Adenosquamous/squamous type (p =0.038).<br />

Multivariate analysis showed that Tapasin and TAP1 losses<br />

were associated with a high risk <strong>of</strong> death. Although 85% <strong>of</strong><br />

CCA lesions were stained with anti-MICA, most <strong>of</strong> them were<br />

restricted in the cytoplasm. Thus, MICA expression was not<br />

associated with any parameters even in cases <strong>of</strong> HLA class I<br />

loss. Our results demonstrated several abnormalities in HLA<br />

class I antigens and MICA expressions suggesting the potential<br />

mechanisms utilized by cholangiocarcinoma for immune<br />

evasion.<br />

doi:10.1016/j.clim.2007.03.498<br />

Sa.112 CD8+CD28− T Suppressor Lymphocytes<br />

Inhibiting T Cell Proliferative and Cytotoxic<br />

Functions Infiltrate Human Cancers<br />

Gilberto Filaci, Associate Pr<strong>of</strong>essor <strong>of</strong> Internal Medicine,<br />

University <strong>of</strong> Genoa, Department <strong>of</strong> Internal Medicine and<br />

Centre <strong>of</strong> Excellence for Biomedical Research, Genoa, Italy,<br />

Daniela Fenoglio, Assistant Pr<strong>of</strong>essor, University <strong>of</strong> Genoa,<br />

Centre <strong>of</strong> Excellence for Biomedical Research, Genoa, Italy,<br />

Marco Fravega, Fellow, University <strong>of</strong> Genoa, Centre <strong>of</strong><br />

Excellence for Biomedical Research, Genoa, Italy, Giacomo<br />

Borgonovo, Associate Pr<strong>of</strong>essor, University <strong>of</strong> Genoa,<br />

Department <strong>of</strong> Surgical and Morphological Disciplines and<br />

Integrated Methodologi, Genoa, Italy, Gianluca Ansaldo,<br />

Assistant Pr<strong>of</strong>essor, University <strong>of</strong> Genoa, Department <strong>of</strong><br />

Surgical and Morphological Disciplines and Integrated<br />

Methodologi, Genoa, Italy, Paolo Traverso, Assistant<br />

Pr<strong>of</strong>essor, University <strong>of</strong> Genoa, Urology Department, Genoa,<br />

Barbara Villaggio, Technician, University <strong>of</strong> Genoa,<br />

Department <strong>of</strong> Internal Medicine, Genoa, Italy, Jean Louis<br />

Ravetti, Director <strong>of</strong> Anatomic Pathology, San Martino<br />

Hospital, Anatomic Pathology, Genoa, Italy, Giorgio<br />

Carmignani, Full Pr<strong>of</strong>essor, University <strong>of</strong> Genoa, Urology<br />

Department, Genoa, Giancarlo Torre, Full pr<strong>of</strong>essor,<br />

Department <strong>of</strong> Surgical and Morphological Disciplines and<br />

Integrated Methodologies, University <strong>of</strong> Gen, Genoa, Italy,<br />

Francesco Indiveri, Full Pr<strong>of</strong>essor <strong>of</strong> Internal Medicine,<br />

University <strong>of</strong> Genoa, Department <strong>of</strong> Internal Medicine and<br />

Centre <strong>of</strong> Excellence for Biomedical Research, Genoa, Italy<br />

Since an important role in determining tumor evasion<br />

from immune control might be played by tumor infiltrating<br />

regulatory lymphocytes we performed a study aimed at<br />

characterizing phenotype and function <strong>of</strong> CD8+CD28− T<br />

suppressor cells infiltrating human cancer. Lymphocytes<br />

infiltrating primitive tumor lesion and/or satellite lymph<br />

node from a series <strong>of</strong> 42 human cancers were phenotypically<br />

studied and functionally analyzed by suppressor assays. The<br />

unprecedented observation was made that CD8+CD28− T<br />

suppressor lymphocytes are almost constantly present and<br />

functional in human tumors, being able to inhibit both T cell


Abstracts<br />

proliferation and cytotoxicity. CD4+CD25+ T regulatory<br />

lymphocytes associate with CD8+CD28− T suppressor cells<br />

so that the immunosuppressive activity <strong>of</strong> tumor infiltrating<br />

regulatory T cell subsets, altogether considered, may<br />

become predominant. The infiltration <strong>of</strong> regulatory T cells<br />

seems tumor-related, being present in metastatic but not in<br />

metastasis free satellite lymph nodes; it likely depends on<br />

both in situ generation (via cytokine production) and<br />

recruitment from the periphery (via chemokine secretion).<br />

Collectively, these results have pathogenic relevance and<br />

implication for immunotherapy <strong>of</strong> cancer.<br />

doi:10.1016/j.clim.2007.03.499<br />

Sa.113 GCN2 Kinase: An Evolutionarily Conserved<br />

Mechanism <strong>of</strong> Human T Lymphocyte Suppression<br />

Upon Arginine Depletion<br />

Markus Munder, Instructor in Medicine, University <strong>of</strong><br />

Heidelberg. Department <strong>of</strong> Hematology, Oncology and<br />

Rheumatology, Heidelberg, Germany, Michelle Bhairo,<br />

Research Assistant, University <strong>of</strong> Heidelberg. Institute <strong>of</strong><br />

<strong>Immunology</strong>, Heidelberg, Germany, Thomas Giese, Group<br />

Leader, University <strong>of</strong> Heidelberg. Institute <strong>of</strong> <strong>Immunology</strong>,<br />

Heidelberg, Germany, Claudia Luckner, Research Assistant,<br />

University <strong>of</strong> Heidelberg, Department <strong>of</strong> Hematology,<br />

Oncology and Rheumatology, Heidelberg, Germany, Anthony<br />

Ho, Head <strong>of</strong> Department, University <strong>of</strong> Heidelberg.<br />

Department <strong>of</strong> Hematology, Oncology and Rheumatology,<br />

Heidelberg, Germany, Stefan Meuer, Head <strong>of</strong> Department,<br />

University <strong>of</strong> Heidelberg. Institute <strong>of</strong> <strong>Immunology</strong>,<br />

Heidelberg, Germany<br />

Impaired T cell immunity is a cardinal feature <strong>of</strong> chronic<br />

inflammation and tumor growth. We have recently demonstrated<br />

that human polymorphonuclear leukocytes (PMN)<br />

constitutively express high activities <strong>of</strong> the arginine-metabolising<br />

enzyme arginase. Upon PMN cell death (e.g., during<br />

purulent inflammation) this enzyme is liberated and metabolizes<br />

arginine to ornithine and urea in the microenvironment.<br />

This arginine depletion completely inhibits<br />

proliferation and effector functions <strong>of</strong> activated human T<br />

lymphocytes while their viability remains unaltered. Specifically,<br />

the T cell cytokine response is impaired at a<br />

posttranscriptional level by arginine depletion. Arginasemediated<br />

arginine depletion is likely a key mechanism <strong>of</strong> T<br />

cell immunosuppression in the inflammatory or tumor<br />

environment. We now wanted to further clarify how T cell<br />

reactivity is turned <strong>of</strong>f in the face <strong>of</strong> arginine depletion. Here<br />

we demonstrate for the first time that human T lymphocytes<br />

respond to arginine depletion by phosphorylation <strong>of</strong> the<br />

kinase GCN2. This is analogous to the general control<br />

response <strong>of</strong> lower eukaryotes upon amino acid depletion.<br />

We also analysed phosphorylation <strong>of</strong> the α-subunit <strong>of</strong><br />

eukaryotic translation initiation factor 2 (eIF2α) and ATF-2<br />

as well as expression <strong>of</strong> transcription factors ATF-4 and CHOP,<br />

known elements in lower eukaryotes in their response to<br />

amino acid depletion. We show that these mammalian<br />

homologues are less stringently regulated in human lymphocytes<br />

upon arginine depletion than phosphorylation <strong>of</strong><br />

GCN2K. In summary, our data demonstrate an evolutionarily<br />

conserved mechanism <strong>of</strong> cellular response towards amino<br />

acid depletion in human T lymphocytes.<br />

doi:10.1016/j.clim.2007.03.500<br />

Sa.114 Characterization <strong>of</strong> the B Cell Infiltrate<br />

Within Intracranial Germinomas<br />

Shannon L. McArdel, Research Technician, Brigham and<br />

Women’s Hospital, Boston, MA, Katharine Cronk,<br />

Neurosurgery Resident, Columbia Medical School, New York,<br />

NY, Kimberly L. Shampain, Student Researcher, Brigham and<br />

Women’s Hospital, Boston, MA, Richard C.E. Anderson,<br />

Assistant Pr<strong>of</strong>essor <strong>of</strong> Neurosurgery and Pediatric<br />

Neurosurgery, Columbia University Medical Center, New<br />

York, NY, David E. Anderson, Instructor in Neurology,<br />

Harvard Medical School, Boston, MA, Jeffrey N. Bruce,<br />

Pr<strong>of</strong>essor <strong>of</strong> Neurosurgery, Columbia University Medical<br />

Center, New York, NY, David A. Hafler, Pr<strong>of</strong>essor <strong>of</strong><br />

Neurology, Harvard Medical School, Brigham and Women’s<br />

Hospital, Boston, MA, Kevin C. O’Connor, Instructor in<br />

Neurology, Harvard Medical School, Boston, MA<br />

Intracranial germinomas arise primarily in children, from<br />

neoplastic transformation <strong>of</strong> embryonic germ cells in the<br />

CNS, most <strong>of</strong>ten in the pineal and suprasellar regions. These<br />

malignant tumors are currently treated with surgery followed<br />

by radiation therapy. Many children however, are too<br />

young to receive radiation therapy or present with tumor<br />

disseminated throughout the cerebrospinal fluid pathways,<br />

making treatment difficult. Thus, germinomas present an<br />

attractive target for antibody-based immunotherapy. Intracranial<br />

germinomas commonly include an immune cell<br />

infiltrate, including a prominent population <strong>of</strong> B cell subsets<br />

that have not yet been characterized. We began to<br />

investigate these B cells by examining their antibody<br />

repertoire. We constructed immunoglobulin variable region<br />

(V) gene libraries from consecutive sections and from laser<br />

capture microdissected B cells from resected germinoma<br />

tumors. Comparison <strong>of</strong> the V-gene sequences to their<br />

corresponding germline sequences revealed that somatic<br />

mutation, oligoclonal expansion and isotype switching were<br />

prominent within the tumor infiltrate. These are cardinal<br />

features <strong>of</strong> an antigen driven B cell response, indicating that<br />

a local inflammatory response may be occurring within the<br />

CNS. This response is likely to be driven by tumor-associated<br />

antigens, the identification <strong>of</strong> which will provide insight into<br />

the immune-related tumor microenvironment.<br />

doi:10.1016/j.clim.2007.03.501<br />

S113<br />

Sa.115 Wnt5a Regulates Melanoma Antigen<br />

Recognized by T Cells 1 (MART-1), a Predominant<br />

Antigen in Melanoma Cells<br />

Samudra Dissanayake, Post doctoral Fellow, National<br />

Institute on Aging, National Institute <strong>of</strong> Health, Baltimore,<br />

MD, Devin Rosenthal, Student, Laboratory <strong>of</strong> <strong>Immunology</strong>,<br />

National Institute on Aging, National Institute <strong>of</strong> Health,<br />

Baltimore, MD, Kyle Hewitt, Student, Laboratory <strong>of</strong>


S114 Abstracts<br />

<strong>Immunology</strong>, National Institute on Aging, National Institute<br />

<strong>of</strong> Health, Baltimore, MD, Michael Wade, Student,<br />

Laboratory <strong>of</strong> <strong>Immunology</strong>, National Institute on Aging,<br />

National Institute <strong>of</strong> Health, Baltimore, MD, Sherry Yang,<br />

Student, Laboratory <strong>of</strong> <strong>Immunology</strong>, National Institute on<br />

Aging, National Institute <strong>of</strong> Health, Baltimore, MD, Poloko<br />

Leotlela, Post doctoral Fellow, Laboratory <strong>of</strong> <strong>Immunology</strong>,<br />

National Institute on Aging National Institute <strong>of</strong> Health,<br />

Baltimore, MD, Dennis Taub, Chief, Laboratory <strong>of</strong><br />

<strong>Immunology</strong>, Laboratory <strong>of</strong> <strong>Immunology</strong>, National Institute<br />

on Aging, National Institute <strong>of</strong> Health, Baltimore, MD, Adam<br />

Riker, Chief <strong>of</strong> Surgical Oncology, Mitchell cancer Institute,<br />

University <strong>of</strong> South Alabama, North Mobile, AL, Brian<br />

Nickol<strong>of</strong>f, Pr<strong>of</strong>essor pathology, Loyola University Medical<br />

Center, Maywood, IL, Ashani Weeraratna, Staff Scientist,<br />

Laboratory <strong>of</strong> <strong>Immunology</strong>, National Institute on Aging,<br />

National Institute <strong>of</strong> Health, Baltimore, MD<br />

Wnt5a increases melanoma cell motility via activation <strong>of</strong><br />

Protein Kinase C (PKC). Microarray analysis showed an<br />

inverse relationship between Wnt5a and MART-1. Using PKC<br />

activation and inhibition studies, as well as recombinant<br />

Wnt5a and Wnt5a RNAi, we demonstrate here that Wnt5aactivated<br />

PKC affects the expression <strong>of</strong> MART-1 via the<br />

phosphorylation <strong>of</strong> STAT3. STAT3 is a transcriptional factor<br />

known to inhibit the binding <strong>of</strong> MART-1 transcriptional<br />

activators MITF, SOX10 and PAX3. These effects are mediated<br />

by Wnt5a upstream <strong>of</strong> PKC, as overexpression <strong>of</strong> Wnt5a<br />

increases pSTAT3 and Wnt5a RNAi treatment decreases<br />

pSTAT3 levels. The decrease in pSTAT3 corresponds to an<br />

increase in MART1 and vice versa. Translocation <strong>of</strong> pSTAT3<br />

from the cytosol to the nucleus upon transfection <strong>of</strong> Wnt5a,<br />

indicated activation <strong>of</strong> STAT3 by Wnt5a, while the reverse<br />

was observed for cells knocked down for Wnt5a. Further,<br />

steady increases in PAX3 and MART-1 protein levels were<br />

observed in cells treated with Wnt5a RNAi, possibly due to<br />

suppressing the effects <strong>of</strong> STAT3. MART1 positive melanoma<br />

cells are able to activate CTL, while addition <strong>of</strong> recombinant<br />

Wnt5a to these cells could not, presumably due to the downregulation<br />

<strong>of</strong> MART-1. RNA from human melanoma biopsies<br />

analyzed for Wnt5a by real-time PCR shows an inverse<br />

relationship between Wnt5a levels and patient survival time,<br />

suggesting that cells with higher MART-1 have a better<br />

outcome. Collectively, our data suggest that Wnt5a, via PKC,<br />

results in the phosphorylation <strong>of</strong> STAT-3 and a subsequent<br />

down-regulation MART1, thereby rendering melanoma cells<br />

able to escape immune detection.<br />

doi:10.1016/j.clim.2007.03.502<br />

Sa.116 Discovery <strong>of</strong> Cancer and Autoimmune<br />

Biomarkers Utilizing Serum Antibody Pr<strong>of</strong>iling on<br />

Protein Microarrays<br />

Jennifer Cannon, Scientist, Invitrogen Corporation,<br />

Carlsbad, CA, Michael Snyder, Pr<strong>of</strong>essor, Yale University,<br />

New Haven, CT, Michael Hudson, Scientist, Yale University,<br />

New Haven, CT, Gregory Michaud, Scientist, Invitrogen<br />

Corporation, Carlsbad, CA, Michael Smith, Scientist,<br />

Invitrogen Corporation, Carlsbad, CA, Barry Schweitzer,<br />

Research and Development Director, Invitrogen<br />

Corporation, Carlsbad, CA, Guillermo Mor, Associate<br />

Pr<strong>of</strong>esor, OB/GYN, New Haven, CT<br />

It has been well established that serum autoantibodies<br />

have important diagnostic value for many diseases, including<br />

cancer and autoimmune diseases. Identifying the antigens<br />

which elicit an autoimmune response can yield sets <strong>of</strong><br />

biomarkers that provide classifiers for particular diseases<br />

and disease stages as well as predictors <strong>of</strong> patient outcomes<br />

and patient responses to therapeutics. Protein microarrays<br />

are valuable tools that have been successfully applied to<br />

investigate the circulating antibody pr<strong>of</strong>ile in several disease<br />

states. To discover potential autoantibody biomarkers<br />

specific to ovarian cancer, sera from 30 ovarian cancer<br />

patients and 30 healthy patients were pr<strong>of</strong>iled on protein<br />

microarrays. Patient and control sera were diluted and<br />

applied to protein microarrays containing over 5000 purified<br />

proteins expressed in a baculovirus expression system. Over<br />

95 candidate biomarkers were identified that exhibited<br />

enhanced reactivity in sera from cancer patients relative to<br />

that <strong>of</strong> the control individuals. Antibodies to four antigens<br />

were tested for differential reactivity in tissue samples <strong>of</strong><br />

cancer patients relative to healthy patients using immunoblot<br />

analysis and tissue microarrays. Three biomarkers<br />

exhibited elevated expression in the cancer tissue relative<br />

to controls. Signal observed for the biomarker antigens<br />

appeared significantly stronger than signal corresponding to<br />

the existing ovarian cancer antigen, CA-125. Additional<br />

studies focused on identifying biomarkers associated with<br />

autoimmune diseases, such as Rheumatoid Arthritis and<br />

Systemic Lupus Erythematosus, have recently utilized a<br />

higher content protein microarray including over 8000<br />

unique human proteins. The application <strong>of</strong> protein microarray<br />

technology for serum antibody pr<strong>of</strong>iling will be<br />

discussed along with data from on-going validation studies.<br />

doi:10.1016/j.clim.2007.03.503<br />

Sa.117 Phenotype and Function Analysis <strong>of</strong><br />

Dendritic Cells From Patients with Prostate<br />

Carcinoma After Radical Prostatectomy<br />

Ivo Minarik, PhD Student, Department <strong>of</strong> <strong>Immunology</strong>, 2nd<br />

Medical School, Charles University, Prague 5, Czech<br />

Republic, Zuzana Tobiasova, PhD Student, Department <strong>of</strong><br />

<strong>Immunology</strong>, 2nd Medical School, Charles University, Prague<br />

5, Czech Republic, Jirina Bartunkova, Head <strong>of</strong> Department,<br />

Department <strong>of</strong> <strong>Immunology</strong>, 2nd Medical School, Charles<br />

University, Prague 5, Czech Republic, Ivan Kawaciuk, Head<br />

<strong>of</strong> Department, Department <strong>of</strong> Urology, 2nd Medical School,<br />

Charles University, Prague 5, Czech Republic<br />

Prostate carcinoma represents the 3rd most frequent<br />

malignancy in the Czech male population with the<br />

incidence <strong>of</strong> 68.3/100000 and mortality 28.2/100000.<br />

About 35% <strong>of</strong> patients experience biochemical relapse<br />

(PSA increase) any time after radical prostatectomy.<br />

Hence, alternative treatment is highly required. Dendritic<br />

cell (DC)-based immunotherapy seems to be a promising<br />

approach for the elimination <strong>of</strong> relapse. In our study we aim<br />

to compare function and phenotype <strong>of</strong> dendritic cells from


Abstracts<br />

patients with prostate carcinoma, who underwent radical<br />

prostatectomy, to controls without tumor. We included 10<br />

patients (age 53-76) with prostate carcinoma who fulfilled<br />

at least one <strong>of</strong> the following risk factors <strong>of</strong> biochemical<br />

relapse: preoperative PSA> 10ng/ml, postoperative Gleason<br />

score >7 or penetration through prostate capsule. The<br />

control group comprised 10 patients (age 53-73) with<br />

benign prostate hyperplasia. We examined the phenotype<br />

(HLA-DR, CD11c, CD83, CD80, and CD86) and functionality<br />

(endocytosis, production <strong>of</strong> IL-12, IL-6, IL-1beta, IL-10, and<br />

TNF-alpha) <strong>of</strong> dendritic cells (DC) and compared the same<br />

features on DC from controls. Immature DCs were prepared<br />

from monocytes after 6 days <strong>of</strong> cultivation in the presence<br />

<strong>of</strong> GM-CSF and IL-4. Their maturation was achieved by 24<br />

hours co-incubation with either poly (I:C), or LPS. Immature<br />

DCs demonstrated higher phagocytic activity, while mature<br />

DCs produced more inflammatory cytokines and expressed<br />

CD83, HLA-DR and costimulatory molecules in higher<br />

amounts. The difference between patients and healthy<br />

donors was insignificant. In conclusion, dendritic cells<br />

prepared from monocytes <strong>of</strong> patients are functionally and<br />

phenotypically competent and so can be used for development<br />

<strong>of</strong> DC vaccines.<br />

doi:10.1016/j.clim.2007.03.505<br />

Sa.119 Parallel Mechanisms <strong>of</strong> Activation by Human<br />

and Mouse Dendritic Cells Treated with the IgM<br />

Immune Modulator B7-DC XAb<br />

Larry Pease, Pr<strong>of</strong>essor, Mayo Clinic College <strong>of</strong> Medicine,<br />

<strong>Immunology</strong>, Rochester, MN, Erin Schenk, Graduate Student,<br />

Mayo Clinic College <strong>of</strong> Medicine, Department <strong>of</strong><br />

<strong>Immunology</strong>, Rochester, MN, Svetomir Markovic, Associate<br />

Pr<strong>of</strong>essor, Mayo Clinic College <strong>of</strong> Medicine, Rochester, MN<br />

Systemic treatment <strong>of</strong> mice with the human antibody B7-<br />

DC XAb modulates dendritic cell (DC) function, protecting<br />

animals from experimental inflammatory airway disease and<br />

established syngeneic tumor implants. Protection in both<br />

models involves modulation <strong>of</strong> T cell function by activated<br />

DCs. B7-DC XAb (1) activates mouse and human DCs in a B7-<br />

DC XAb dependent fashion, (2) induces very similar macromolecular<br />

complexes on the cell surface <strong>of</strong> mouse and human<br />

DCs, and (3) initiates similar signaling cascades, patterns <strong>of</strong><br />

gene expression, and DC functions in these antigen presenting<br />

cells derived from the two species. Furthermore, human<br />

DCs activated with B7-DC XAb are strong inducers <strong>of</strong> antigenspecific<br />

T cell immune responses by peripheral blood T cells<br />

in vitro. The similar response pattern found in mouse and<br />

human DCs is even more remarkable since bone marrowderived<br />

DCs from the mouse and monocyte-derived DCs from<br />

human peripheral blood do not represent closely related<br />

differentiation states prior to activation with the immune<br />

modulator. Nonetheless, these strong parallels in mouse and<br />

human responsiveness to B7-DC XAb treatment provide<br />

justification for our ongoing efforts to develop this human<br />

antibody for use in clinical trials.<br />

doi:10.1016/j.clim.2007.03.506<br />

Sa.120 Human Glioblastoma Multiforme (GBM)<br />

Tumors are Enriched for Strongly Suppressive,<br />

Apoptosis Resistant, DR+ Tregs<br />

Charles Ashley, Research Technician, Harvard Medical<br />

School, Brigham and Women’s Hospital, Department <strong>of</strong><br />

Neurology, Boston, MA, Richard Anderson, Assistant<br />

Pr<strong>of</strong>essor, Columbia University, Department <strong>of</strong> Pediatrics,<br />

New York, NY, Jeffrey Bruce, Pr<strong>of</strong>essor, Neurological<br />

Institute <strong>of</strong> New York, New York, NY, David Anderson,<br />

Instructor <strong>of</strong> Neurology, Harvard Medical School, Brigham<br />

and Women’s Hospital, Department <strong>of</strong> Neurology, Boston,<br />

MA, David Hafler, Breakstone Chair <strong>of</strong> Neurology, Harvard<br />

Medical School, Brigham and Women’s Hospital,<br />

Department <strong>of</strong> Neurology, Boston, MA, Clare Baecher-Allan,<br />

Instructor <strong>of</strong> Neurology, Harvard Medical School, Brigham<br />

and Women’s Hospital, Department <strong>of</strong> Neurology,<br />

Boston, MA<br />

Glioblastoma Multiforme (GBM) is a CNS tumor <strong>of</strong> astrocytic<br />

origin, that is highly refractory to current therapies<br />

and associated with survival <strong>of</strong> less than 15 months. Using<br />

post surgical resection GBM samples to study the pr<strong>of</strong>ile <strong>of</strong><br />

the infiltrating regulatory T cells, we have found marked<br />

alterations in subset frequency, activation state, and gene<br />

expression in GBM-derived Tregs. In GBM, not only are<br />

there a large number <strong>of</strong> tumor-infiltrating Tregs, but<br />

importantly, these Tregs are strikingly enriched for the<br />

most suppressive Treg subset that are classified by HLA-DR<br />

expression. In a large panel <strong>of</strong> samples, flow cytometric<br />

analyses indicated that GBM tumors contained a significantly<br />

greater Treg infiltrate than meningiomas, or<br />

metastases <strong>of</strong> non-CNS tumors. Subsequently, using six<br />

color FACS sorting, specific DR+ and DR− Treg cell subsets<br />

were quantified and isolated from matched tumor and<br />

peripheral blood samples from the same individual with<br />

either GBM or other CNS tumors, and from blood <strong>of</strong> healthy<br />

controls. Not only did these analyses demonstrate an<br />

overwhelming prevalence <strong>of</strong> DR+ Tregs in the GBM tumor,<br />

but also that these tumor derived DR+ Tregs exhibit<br />

extremely high levels <strong>of</strong> Foxp3 and extremely low levels<br />

<strong>of</strong> the pro-apoptotic gene, Bax, as compared to peripheral<br />

blood derived DR+ Tregs. These data indicate that the GBM<br />

associated increase in Tregs may be due to increased Treg<br />

survival, possibly due to the actions <strong>of</strong> IL-10 which we have<br />

found reduces Treg expression <strong>of</strong> both Bax protein and<br />

mRNA.<br />

doi:10.1016/j.clim.2007.03.507<br />

S115<br />

Sa.121 Therapeutic Dendritic Cell Vaccine<br />

Preparation Using Tumor RNA Transfection<br />

Juliana Sousa-Canavez, Researcher, Genoa Biotech, São<br />

Paulo, Brazil, Flavio Canavez, Researcher, Genoa Biotech,<br />

São Paulo, Brazil, Cristina Massoco, Researcher, Genoa<br />

Biotech, São Paulo, Brazil, Katia Leite, Pathologist,<br />

Genoa Biotech, São Paulo, Brazil, Luiz Camara-Lopes,<br />

Pathologist, Genoa Biotech, São Paulo, Brazil, Dewton<br />

Moraes Vasconcelos, Pr<strong>of</strong>essor, Genoa Biotech, São Paulo,<br />

Brazil


S116 Abstracts<br />

Dendritic cells (DC) are potent antigen-presenting cells<br />

that induce and maintain primary immune responses. In the<br />

last years, many studies have used this property to initiate<br />

antigen T cell responses against neoplasic cells. This was<br />

achieved by vaccinating patients with DC carrying tumorassociated<br />

antigens prepared in vitro. A promising method<br />

for DC vaccine preparation is the transfection <strong>of</strong> tumor<br />

cells RNA. Preliminary clinical studies using RNA transfection<br />

have been shown encouraging results and minor side<br />

effects. Many aspects <strong>of</strong> this method, however, are not<br />

understood and further investigations are needed, especially<br />

regarding vaccine preparation. Here, we present our<br />

data obtained by DC transfection with total RNA isolated<br />

from a prostate tumor lineage LNCAP that overexpresses<br />

PSA antigen. PBMC were collected from healthy volunteers<br />

through apheresis and mononuclear cells were separated in<br />

Ficoll gradient. DC was differentiated with GM-CSF and IL-<br />

4, and maturation induced by TNFα DC phenotypes were<br />

confirmed by flow-cytometry using CD80, CD86, CD14,<br />

CD11c, CD1a and CCR7 antibodies. Immature cells were<br />

transfected with 4 1/4 and 16 1/4 g <strong>of</strong> RNA using either<br />

300 V or 400 V pulses. Mature cells were transfected with 4<br />

1/4 μg and 5 1/4 μg <strong>of</strong> RNA using 300 V, 400 V or 500 V<br />

pulses. In all conditions, we detected PSA expression by<br />

immunohistochemistry indicating that RNA penetrated in<br />

DC. For mature cells, we have also co-incubated total RNA<br />

(4 1/4 μg and 5 1/4 μg) and DC with poor results. Our<br />

experiments point to RNA transfection (5 1/4 μg total RNA)<br />

in mature DC using 400 V pulse as the most efficient<br />

condition.<br />

doi:10.1016/j.clim.2007.03.508<br />

Sa.122 Regulatory T Cells Affect the DC-mediated<br />

Immunotherapy Outcome in Melanoma Patients<br />

Mercedes N. López, Medical Doctor, Research Support<br />

Office, <strong>Clinical</strong> Hospital, University <strong>of</strong> Chile, Santiago de<br />

Chile, Chile, Gabriela Segal, Biotech, Program <strong>of</strong><br />

<strong>Immunology</strong>, Institute <strong>of</strong> Biomedical Sciences, University <strong>of</strong><br />

Chile, Santiago de Chile, Chile, Cristian Pereda, Biotech,<br />

Program <strong>of</strong> <strong>Immunology</strong>, Institute <strong>of</strong> Biomedical Sciences,<br />

University <strong>of</strong> Chile, Santiago de Chile, Chile, Raquel<br />

Aguilera, Medical Doctor, Program <strong>of</strong> <strong>Immunology</strong>, Institute<br />

<strong>of</strong> Biomedical Sciences, University <strong>of</strong> Chile, Santiago de<br />

Chile, Chile, Alexandra Ginesta, Medical Doctor, Program<br />

<strong>of</strong> <strong>Immunology</strong>, Institute <strong>of</strong> Biomedical Sciences, University<br />

<strong>of</strong> Chile, Santiago de Chile, Chile, Diego Reyes, Medical<br />

Doctor, Program <strong>of</strong> <strong>Immunology</strong>, Institute <strong>of</strong> Biomedical<br />

Sciences, University <strong>of</strong> Chile, Santiago de Chile, Chile,<br />

Carlos Ferrada, Medical Doctor, Surgery department,<br />

<strong>Clinical</strong> Hospital, University <strong>of</strong> Chile, Santiago de Chile,<br />

Chile, Flavio Salazar-Onfray, PhD <strong>of</strong> Sciences, Program <strong>of</strong><br />

<strong>Immunology</strong>, Institute <strong>of</strong> Biomedical Sciences, University <strong>of</strong><br />

Chile, Santiago de Chile, Chile<br />

Dendritic cell (DCs)-based therapy has proven to be<br />

effective in patients with malignancies. A Chilean phase I<br />

clinical study for the treatment <strong>of</strong> advanced malignant<br />

melanoma performed in our laboratory, indicates that<br />

autologous human dendritic cells (hDCs) pulsed with a<br />

melanoma cell lysate acquired a mature phenotype and<br />

were able to trigger specific immune responses to tumor<br />

antigens in vivo. In this study, 37 patients with malignant<br />

melanoma stage IV were vaccinated with autologous DCs<br />

pulsed with a melanoma cell lysate. After vaccination, 50%<br />

tested patients (18 out <strong>of</strong> 37) showed a positive Delayed type<br />

IV hypersensitivity reaction (DTH) against the melanoma cell<br />

lysate. All patients showed a positive DTH against the used<br />

adjuvant KLH demonstrating that all patients were immunocompetents.<br />

Significant correlations were found between<br />

DTH positive responses against tumour cell lysate and both<br />

disease stability and post-vaccination survival on the stage IV<br />

patients. However, there is a group <strong>of</strong> patients that did not<br />

show immunological responses. We compared a number <strong>of</strong><br />

DTH responder patients (n=4) with a group <strong>of</strong> DTH noresponder<br />

patients (n=4) and analyzed the number and<br />

proportion <strong>of</strong> regulatory T cells (Treg). Our results showed<br />

that DTH non-responder patients showed an increase <strong>of</strong> Treg<br />

cells after the DC vaccination compared with DTH responder<br />

patients that did not show changes in the number or<br />

proportion <strong>of</strong> Treg cells. These observations should be<br />

important in the study <strong>of</strong> the role <strong>of</strong> regulatory T cells in<br />

the clinical responses <strong>of</strong> cancer patients treated with<br />

immunotherapy.<br />

doi:10.1016/j.clim.2007.03.509<br />

Sa.123 Dendritic Cell-Tumor Cell Fusion Vaccine for<br />

the Treatment <strong>of</strong> Advanced Tumors: Evaluation <strong>of</strong><br />

the Quality <strong>of</strong> Life by Questionnaires<br />

Dewton Moraes Vasconcelos, MD, PhD, Genoa Biotecnologia,<br />

São Paulo, Brazil, Antonio Carlos Misiara, MD, PhD, Genoa<br />

Biotecnologia S.A., São Paulo, Brazil, Juliana Moreira<br />

Sousa-Canavez, BSc, PhD, Genoa Biotecnologia S.A., São<br />

Paulo, Brazil, Paloma Aguiar, BSc, Genoa Biotecnologia S.A.,<br />

São Paulo, Brazil, Elaine Cristina Corneta, MSc, Genoa<br />

Biotecnologia S.A., São Paulo, Brazil, Katia Ramos Moreira<br />

Leite, MD, PhD, Genoa Biotecnologia S.A., São Paulo, Brazil,<br />

Luiz Heraldo Arouche Camara Lopes, MD, PhD, Genoa<br />

Biotecnologia S.A., São Paulo, Brazil<br />

Dendritic cells are the most potent antigen-presenting<br />

cells, and the possibility <strong>of</strong> their use for cancer vaccination<br />

has renewed the interest in this therapeutic modality.<br />

Nevertheless, the ideal immunization protocol with these<br />

cells has not been described yet. In this report we describe<br />

the preliminary results <strong>of</strong> a protocol using autologous tumor<br />

and allogeneic dendritic hybrid cell vaccination every<br />

4 weeks, for metastatic melanoma and renal cell carcinoma<br />

(RCC) patients, and a few patients with other types <strong>of</strong><br />

neoplasia. Thirty-six patients were evaluated by a questionnaire<br />

searching for information <strong>of</strong> the quality <strong>of</strong> life<br />

(QoL) during the treatment with the vaccine. Though all<br />

patients included presented with large tumor burdens and<br />

progressive diseases, 77% <strong>of</strong> them experienced stability after<br />

vaccination, 9% related improvement and 14% presented<br />

deterioration <strong>of</strong> the clinical features. Among RCC patients 3/<br />

10 (30%) presented deterioration, 1/10 improvement (10%)<br />

and 6/10 (60%) stabilization. Among melanoma patients 1/15<br />

(6.6%) deteriorated, 1/15 (6.6%) improved and 13/15 (86.6%)


Abstracts<br />

stabilized the disease. These data indicate that dendritic<br />

cell-tumor cell hybrid vaccination affects the natural history<br />

<strong>of</strong> advanced cancer and provides support for its study in less<br />

advanced patients, who should, more likely, benefit even<br />

more from this approach.<br />

doi:10.1016/j.clim.2007.03.510<br />

Sa.124 Rice Bran Supplement (MGN-3/Biobran)<br />

Suppresses Tumor Growth via Modulating Cytokine<br />

Production and Increasing Apoptotic Level in Ehrlich<br />

Carcinoma-bearing Mice<br />

Nariman Badr El-Din, Pr<strong>of</strong>essor, University <strong>of</strong> Mansoura,<br />

Faculty <strong>of</strong> Science, Department <strong>of</strong> Zoology, Mansoura, Eman<br />

Noaman, Pr<strong>of</strong>essor, National Center for Radiation and<br />

Technology, Department <strong>of</strong> Radiation Biology, Cairo, Alia<br />

Ghoneum, Student, Charles R. Drew University <strong>of</strong> Medicine<br />

and Science, Department <strong>of</strong> Otolaryngology, Los Angeles,<br />

CA, Mamdooh Ghoneum, Associate Pr<strong>of</strong>essor III, Charles R.<br />

Drew University <strong>of</strong> Medicine and Science, Department <strong>of</strong><br />

Otolaryngology, Los Angeles, CA<br />

This study was undertaken to investigate the anti-tumor<br />

activity in vivo by MGN-3/Biobran and the mechanisms<br />

underlying its effect. MGN-3 is an arabinoxylan extracted<br />

from rice bran that is treated with hydrolyzing enzymes from<br />

shiitake mushrooms. Female Swiss albino mice were inoculated<br />

intramuscularly in the right thigh with Ehrlich Ascites<br />

Carcinoma (EAC) cells. At day 8, mice bearing Solid Ehrlich<br />

Carcinoma tumor (SEC) were treated with MGN-3 (40 mg/kg<br />

body weight) via intraperitoneal injection, 3 times/week,<br />

until day 35. Tumor growth (volume and weight), plasma<br />

cytokine production, and apoptotic effect <strong>of</strong> MGN-3 were<br />

examined. Injection with MGN-3 caused a highly significant<br />

delay in both tumor volume (63.27%) and tumor weight<br />

(45.2%) from controls (pb0.01). The mechanisms by which<br />

MGN-3 exerts its anti-tumor effect seem to involve its ability<br />

to influence plasma cytokine production via increasing the<br />

levels <strong>of</strong> IFNγ (184.4%, pb0.01) and TNFα (11%, pb0.01)<br />

over control mice bearing SEC, while down-regulating levels<br />

<strong>of</strong> the immune suppressing cytokine IL-10. In addition, MGN-3<br />

induced 1.8 fold increase in the percentage <strong>of</strong> apoptotic SEC<br />

cells as determined by flow cytometry and the histopathological<br />

examination. No adverse side effects due to MGN-3<br />

treatment were observed; all animals displayed normal<br />

feeding/drinking and life activity patterns with a significant<br />

body weight gain <strong>of</strong> 7.32%, pb0.025. These data may have<br />

clinical implications for the treatment <strong>of</strong> solid cancers. MGN-<br />

3/Biobran was <strong>of</strong>fered by Daiwa Pharmaceuticals Co., Ltd.,<br />

Tokyo, Japan.<br />

doi:10.1016/j.clim.2007.03.511<br />

Sa.125 Therapeutic Efficacy <strong>of</strong> the Most Potent<br />

Macrophage Activating Factor (GcMAF) for Prostate<br />

and Breast Cancers<br />

Nobuto Yamamoto, Director, Socrates Institute for<br />

Therapeutic <strong>Immunology</strong>, Philadelphia, PA, Masumi Ueda,<br />

Chief, Microbiology Department, Socrates Institute for<br />

Therapeutic <strong>Immunology</strong>, Philadelphia, PA, Charles Benson,<br />

Head, Infectious Diseases, School <strong>of</strong> Veterinary Medicine,<br />

University <strong>of</strong> Pennsylvania, Kennett Square, PA<br />

Inflammation-primed macrophage activation is the principal<br />

macrophage activation process that requires serum<br />

vitamin D-binding protein (known as Gc protein) and<br />

participation <strong>of</strong> B and T lymphocytes. A trisaccharide<br />

composed <strong>of</strong> N-acetylgalactosamine with dibranched galactose<br />

and sialic acid termini at 420 threonine residue <strong>of</strong> Gc<br />

protein is hydrolyzed by the β-galactosidase (Bgl) <strong>of</strong><br />

inflammation-primed B cells and the Neu-1 sialidase <strong>of</strong> T<br />

cells to yield the macrophage activating factor (MAF). Thus,<br />

Gc protein is the precursor for the principal MAF. However,<br />

the MAF precursor activity <strong>of</strong> serum Gc protein <strong>of</strong> cancer<br />

patients was lost or reduced because Gc protein is<br />

deglycosylated by serum α-N-acetylgalactosaminidase<br />

(Nagalase) secreted from cancerous cells. Serum Nagalase<br />

activity is proportional to tumor burden and serves as a<br />

prognostic index. Stepwise treatment <strong>of</strong> highly purified Gc<br />

protein with immobilized β-galactosidase and sialidase<br />

generated the most potent macrophage activating factor<br />

(GcMAF) that produces no side effect in humans. When<br />

human macrophages were treated in vitro with GcMAF (100<br />

pg/ml) for 3 h, the macrophages developed enormous<br />

variation <strong>of</strong> receptors that recognize a variety <strong>of</strong> cellular<br />

abnormalities, i.e., tumor associated antigens, in cancerous<br />

cell surface. Thus, the activated macrophages were<br />

highly tumoricidal to a variety <strong>of</strong> cancerous cells. Administration<br />

<strong>of</strong> 100 ng GcMAF/human results in the maximal<br />

activation <strong>of</strong> systemic macrophages. When nonanemic<br />

prostate and breast cancer patients (total <strong>of</strong> 36 patients)<br />

were treated with 14–25 weekly administrations <strong>of</strong> GcMAF<br />

(100 ng/week), all cancer patients exhibited healthy<br />

control levels <strong>of</strong> the serum Nagalase activity, indicating<br />

eradication <strong>of</strong> tumors.<br />

doi:10.1016/j.clim.2007.03.512<br />

S117<br />

Sa.126 Activation <strong>of</strong> Hepatic Stellate Cells During<br />

Fibrosis: Comparison <strong>of</strong> the CYP2D6 Model for<br />

Autoimmune Hepatits and CCl4 Injection<br />

Urs Christen, Assistant Pr<strong>of</strong>essor, Pharmazentrum, JWG<br />

University Frankfurt, Frankfurt am Main, Germany, Edith<br />

Hintermann, Senior Postdoctoral Fellow, Pharmazentrum,<br />

JWG University Frankfurt, Frankfurt am Main, Germany,<br />

Monika Bayer, Res Tech, Pharmazentrum, JWG University<br />

Frankfurt, Frankfurt am Main, Germany, Selina Christen,<br />

PhD Student, Pharmazentrum, JWG University Frankfurt,<br />

Frankfurt am Main, Germany<br />

Only little is known about the mechanisms <strong>of</strong> periportal<br />

fibrosis in the pathogenesis <strong>of</strong> human autoimmune hepatitis.<br />

We used the virus induced CYP2D6 model system to<br />

investigate the activation <strong>of</strong> hepatic stellate cells (HSC)<br />

and the kinetics <strong>of</strong> fibrosis in comparison with the CCl4induced<br />

fibrosis model. CYP2D6 transgenic mice express the<br />

human Cytochrome P450 in the liver and develop liver<br />

damage upon Adenovirus-CYP2D6 infection. In the CYP2D6


S118 Abstracts<br />

model we found mostly subcapsular fibrosis resulting in the<br />

fusion <strong>of</strong> individual lobules (Sirius Red, Collagen I). At 10–<br />

12 weeks post-infection, weak periportal fibrosis became<br />

apparent. In contrast, in CCl4-treated mice the kinetic <strong>of</strong><br />

extracellular matrix deposition was accelerated resulting in<br />

periportal fibrosis after 3–4 week <strong>of</strong> CCl4 administration. At<br />

later times, fibrosis was much more pronounced in CCl4treated<br />

mice compared to virus-infected CYP2D6 mice but<br />

subcapsular fibrosis was not as dominant. Activation <strong>of</strong> HSCs<br />

could be detected by staining for a-smooth muscle actin<br />

(aSMA) in liver sections <strong>of</strong> both CCl4-treated mice and virusinfected<br />

CYP2D6 mice. In addition, isolation <strong>of</strong> HSCs<br />

revealed an enhanced activation status (decreased amount<br />

<strong>of</strong> oil droplets, de novo aSMA expression) in CCl4-treated<br />

mice and virus-infected CYP2D6 mice. Our data indicate<br />

that virus-infected CYP2D6 mice display subcapsular and<br />

periportal fibrosis that correlates with an activation <strong>of</strong> HSCs<br />

similar to CCl4-induced fibrosis. Thus, the CYP2D6 mouse is<br />

a good model system to further investigate the molecular<br />

mechanisms involved in fibrotic events during autoimmune<br />

hepatitis.<br />

doi:10.1016/j.clim.2007.03.513<br />

Sa.127 Bone-Marrow Derived DCs From CD200R1<br />

KO Mice Stimulated with the Ligand CD200Fc<br />

Preferentially Induce Populations <strong>of</strong><br />

CD4+CD25+ Treg<br />

Reg Gorczynski, Pr<strong>of</strong>essor, University Health Network,<br />

Toronto, ON, Canada, Ivo Boudakov, PDF, University Health<br />

Network, Toronto, ON, Canada<br />

CD200: CD200R1 interaction causes suppression <strong>of</strong><br />

inflammation and graft rejection. The functional activity<br />

induced by alternate CD200R is<strong>of</strong>orms remains unclear. We<br />

generated a BL/6 mouse with deletion <strong>of</strong> the gene for<br />

CD200R1. Marrow cells from KO and wt mice were<br />

cultured for 8 days in the presence <strong>of</strong> (GMCSF +IL-4),<br />

with/without addition <strong>of</strong> a soluble form <strong>of</strong> CD200<br />

(CD200Fc). DCs were stimulated with LPS and used to<br />

activate C3H splenocytes. CTL for BL/6 targets was<br />

assayed at 5 days, or cells were harvested at 72 h,<br />

enriched for CD4+CD25+ cells, and added to fresh cultures<br />

<strong>of</strong> C3H splenocytes stimulated with either BL/6 or BALB/c<br />

mitomycin-c treated DCs. CTL specific for H2b or H2d<br />

targets was assayed 5d later.<br />

DCs <strong>of</strong> wt or CD200R1KO mice cultured without<br />

CD200Fc induced allospecific (anti-H2b) CTL in vitro.<br />

After culture in the presence <strong>of</strong> CD200Fc, DCs from<br />

CD200R1KO mice were inefficient inducers <strong>of</strong> CTL, but<br />

promoted development <strong>of</strong> CD4+CD25+Foxp3+ Treg which<br />

suppressed C3H anti-BL/6 CTL induction in fresh responder/stimulator<br />

cells. To characterize an in vivo equivalent<br />

<strong>of</strong> this phenomenon CD200Fc or mouseIgG2a was infused<br />

iv into wt or CD200R1KO mice for 14 days. Splenic DCs<br />

were isolated by conventional techniques. DCs from<br />

control mice (wt or CD200R1KO) receiving IgG2a only<br />

were equivalent in induction <strong>of</strong> CTL. DCs isolated after<br />

infusion <strong>of</strong> CD200Fc into CD200R1KO mice were impaired<br />

for CTL induction, but induced antigen-specific Treg in<br />

culture. We conclude that immunoregulation induced by<br />

CD200 binding <strong>of</strong> non-CD200R1 is<strong>of</strong>orms <strong>of</strong> the CD200R<br />

receptor family reflects the preferential induction <strong>of</strong><br />

“tolerogenic” DCs.<br />

doi:10.1016/j.clim.2007.03.514<br />

Sa.128 High Molecular Weight Hyaluronan Promotes<br />

the Suppressive Effects <strong>of</strong> CD4+CD25+ Treg<br />

Paul Bollyky, Benaroya Research Institute, Seattle, WA,<br />

Gerald Nepom, Benaroya Research Institute, Seattle, WA,<br />

Jane Buckner, Benaroya Research Institute, Seattle, WA,<br />

Susan Masewicz, Benaroya Research Institute, Seattle, WA,<br />

James Lord, Benaroya Research Institute, Seattle, WA,<br />

Stephen Evanko, Benaroya Research Institute, Seattle,<br />

WA, Thomas Wight, Benaroya Research Institute, Seattle,<br />

WA<br />

Hyaluronan (HA) is a glycosaminoglycan present in the<br />

extra-cellular matrix. When HA is degraded during infection<br />

and injury low-molecular-weight (LMW-HA) forms are<br />

generated whose interactions influence inflammation and<br />

angiogenesis. Intact high-molecular-weight hyaluronan<br />

(HMW-HA), conversely, has been reported to convey antiinflammatory<br />

signals. Here we demonstrate that HMW-HA<br />

acts on human CD4+CD25+ regulatory T cells (Treg) to<br />

promote their suppressive effects while LMW-HA does not.<br />

HMW-HA enhances regulatory T cell functional suppression<br />

<strong>of</strong> responder cell proliferation and up-regulates the<br />

transcription factor Foxp3 on Treg. These effects are only<br />

seen with activated Treg and are associated with expression<br />

<strong>of</strong> CD44 isomers which more highly bind HMW-HA. At<br />

higher concentrations HMW-HA also has direct suppressive<br />

effects on T cells. We propose that the state <strong>of</strong> HA in the<br />

matrix environment provides contextual cues to Treg and T<br />

cells, thereby providing a link between the innate<br />

inflammatory network and the regulation <strong>of</strong> adaptive<br />

immune responses.<br />

doi:10.1016/j.clim.2007.03.515<br />

Sa.129 The Forkhead Transcription Factor, Foxq1,<br />

Enhances NF-kB Activity and is Critical for Robust<br />

T Cell Activation and Autoimmunity<br />

Melanie Gubbels Bupp, Postdoctoral Fellow, Roche Palo<br />

Alto, Palo Alto, CA, Stanford Peng, Director, Arthritis<br />

Research, Roche Palo Alto, Palo Alto, CA<br />

Essential immunoregulatory functions have recently<br />

been ascribed to several members <strong>of</strong> the forkhead<br />

transcription factor family. For example, Foxo3a and<br />

Foxj1 inhibit NF-kB activity and Foxp3 regulates lineage<br />

commitment and function <strong>of</strong> regulatory T cells. Here, we<br />

report that another family member, Foxq1, is critical for<br />

robust helper T cell activation. CD4+ T cells from mice<br />

bearing a mutant allele <strong>of</strong> Foxq1 displayed a significantly<br />

reduced proliferative response with impaired secretion <strong>of</strong><br />

IL-2 and IFN-gamma after TCR ligation as compared to


Abstracts<br />

wild-type (WT) T cells. This defect is largely rescued by<br />

the addition <strong>of</strong> recombinant human IL-2, suggesting that<br />

Foxq1 is required for optimal production <strong>of</strong> IL-2 and<br />

consequently, robust T cell proliferation. Accordingly, in in<br />

vitro luciferase assays, Foxq1 augmented transcriptional<br />

activity <strong>of</strong> the IL-2 transactivator NF-kB while mutant<br />

Foxq1 did not retain this function. Finally, Foxq1-mutant<br />

mice exhibited reduced susceptibility to the progression <strong>of</strong><br />

experimental autoimmune encephalomyelitis than their<br />

WT counterparts. Thus, Foxq1 is essential for helper T cell<br />

activation and autoaggressive T cell responses in vivo,<br />

likely via its ability to potentiate NF-kB activity and IL-2<br />

production.<br />

doi:10.1016/j.clim.2007.03.516<br />

Sa.130 Human Bone Marrow Mesenchymal Stem<br />

Cells Support Polyclonal Stimulation <strong>of</strong> Human B<br />

Cells<br />

Elisabetta Traggiai, PhD, Institute G Gaslini, Genova, Italy,<br />

Alberto Martini, MD/PhD, Institute G Gaslini, Genova, Italy,<br />

Antonio Uccelli, MD/PhD, Department <strong>of</strong> Neurosciences,<br />

Ophthalmology and Genetics, Genova, Italy, Lorenzo<br />

Moretta, MD/PhD, Institute G Gaslini, Genova, Italy,<br />

Federica Benvenuto, PhD, Department <strong>of</strong> Neurosciences,<br />

Ophthalmology and Genetics, Genova, Italy, Marco<br />

Gattorno, MD, Institute G Gaslini, Genova, Italy<br />

To investigate the possible application <strong>of</strong> bone marrow<br />

(BM) mesenchymal stem cells (MSCs) in Systemic Lupus<br />

Erythematosus (SLE), an autoimmune disease in which B<br />

cells play a pivotal role, we studied the interaction <strong>of</strong> B<br />

cell subsets isolated from healthy donor and pediatric SLE<br />

patients with human BM MSCs. We isolated from peripheral<br />

blood <strong>of</strong> healthy donors: a) immature transitional B<br />

cells, b) naive B cells, c) IgM memory and d) switch<br />

memory B cells. BM MSCs promoted proliferation and<br />

differentiation into immunoglobulin secreting cells <strong>of</strong><br />

transitional and naive B cells stimulated with CpG 2006<br />

in the absence <strong>of</strong> BCR triggering, and strongly enhanced<br />

proliferation and differentiation <strong>of</strong> both memory B cell<br />

populations. Furthermore, both proliferation and differentiation<br />

into plasma cells <strong>of</strong> CD19+ B cells isolated from<br />

pediatric SLE patients were enhanced by BM MSCs upon<br />

polyclonal stimulation. Inhibition <strong>of</strong> T cell proliferation by<br />

MSCs was suggested to be dependent on INF-g€ . To test<br />

whether INF-g€ could have an effect on the B cell<br />

response under the influence <strong>of</strong> MSCs, we co-cultured B<br />

cell subsets with MSC in the presence <strong>of</strong> human<br />

recombinant INF-ï §ï€®ï€ After 4 days <strong>of</strong> culture both<br />

proliferation and differentiation into plasma cells <strong>of</strong> all B<br />

cell subsets stimulated with CpG, soluble CD40L and anti-<br />

Ig were inhibited in both healthy donors and SLE patients.<br />

These data show the complexity <strong>of</strong> the cross-talk between<br />

MSCs and B cell and how the microenviroment in which<br />

the interaction take place could influence the outcome <strong>of</strong><br />

the immune response.<br />

doi:10.1016/j.clim.2007.03.517<br />

Sa.131 Examination <strong>of</strong> a Unique T Cell Subset TCD40<br />

in Type 1 Diabetes<br />

David Wagner, Assistant Pr<strong>of</strong>essor, Webb-Waring Institute,<br />

Denver, CO<br />

The identification <strong>of</strong> autoaggressive T cells in human<br />

disease has proven very difficult. We have discovered a<br />

unique T cell subset described as CD4lo and expressing the<br />

CD40 receptor termed TCD40. These T cells are significantly<br />

expanded in T1D but not T2D or control subjects. A portion<br />

<strong>of</strong> TCD40 cells respond to pre-pro-insulin, GAD peptides,<br />

and human islets. These T cells are expanded in diabetes<br />

high susceptibility HLA (DR3, DR4, DQ8) subjects, but are<br />

also expanded when T1D subjects do not possess any <strong>of</strong> the<br />

high susceptibility HLAs. TCD40 cells remain at expanded<br />

percentages in long-term diabetic subjects, suggesting that<br />

CD40 is not merely a T cell activation marker. Interestingly,<br />

non-autoimmune subjects carrying DR3, DR4 or DQ8 do not<br />

have expanded TCD40 cells. Mechanistic differences in<br />

induction <strong>of</strong> RAG1 and RAG2 are seen between T1D and<br />

control subjects. Examination <strong>of</strong> TRAV usage in T1D and<br />

controls demonstrate prominent differences including the<br />

detection <strong>of</strong> TRAV8 in the periphery. TCD40 cells achieve<br />

effector status synthesizing predominantly Th1 cytokines.<br />

These data suggest a unique T cell population that may be<br />

predictive <strong>of</strong> autoimmunity.<br />

doi:10.1016/j.clim.2007.03.518<br />

S119<br />

Sa.132 Effects <strong>of</strong> Class II/CLIP Affinity on the Class II<br />

Antigen Presentation Pathway in the Context <strong>of</strong><br />

Autoimmunity<br />

Cornelia Rinderknecht, Graduate Student, Stanford<br />

University, Department <strong>of</strong> Pediatrics, San Carlos, CA,<br />

Tatiana Catanzarite, Student, Stanford University,<br />

Department <strong>of</strong> Pediatrics, Stanford, CA, Michael Belmares,<br />

Scientist, Arbor Vita Corporation, Sunnyvale, CA, Elizabeth<br />

Mellins, Associate Pr<strong>of</strong>essor, Stanford University,<br />

Department <strong>of</strong> Pediatrics, Stanford, CA, Susan Kovats,<br />

Assistant Member, Oklahoma Medical Research Foundation,<br />

Arthritis and <strong>Immunology</strong> Research Program, Oklahoma<br />

City, OK<br />

Several MHC class II alleles linked with autoimmune<br />

diseases form unusually low-stability complexes with class IIassociated<br />

invariant chain peptides (CLIP), leading us to<br />

hypothesize that this is an important feature contributing to<br />

disease pathogenesis. To investigate cellular consequences<br />

<strong>of</strong> altering class II/CLIP affinity, we evaluated invariant chain<br />

(Ii) mutants with increased CLIP affinity for two murine class<br />

II alleles, I-Ed and I-Ag7, which have low affinity for wt CLIP.<br />

I-Ed is associated with a mouse model <strong>of</strong> spontaneous,<br />

autoimmune joint inflammation, and I-Ag7 is associated with<br />

models <strong>of</strong> diabetes (NOD) and arthritis (KRN). Single amino<br />

acid mutations were made in murine Ii. Pulse-chase/coimmunoprecipitation<br />

analysis <strong>of</strong> I-Ed or I-Ag7 and CLIP was<br />

used to identify physiologically relevant CLIP mutants that<br />

are <strong>of</strong> high affinity but remain DM-sensitive. An increase in<br />

CLIP affinity for I-Ed or I-Ag7 resulted in increased cell-


S120 Abstracts<br />

surface and total cellular levels <strong>of</strong> class II, suggesting post-ER<br />

chaperoning by Ii via its CLIP peptides. Quantitative effects<br />

on class II were less robust in H-2M-expressing cells,<br />

suggesting complementary effects mediated by Ii and H-<br />

2M, and implying that the impact <strong>of</strong> varied CLIP affinity on<br />

immune responses will be highest in cells with limited H-2M<br />

activity. Functionally, cell lines expressing wt or high CLIPaffinity<br />

mutant Ii have differential capacity to present<br />

peptide or whole antigen to a T cell hybridoma, suggesting<br />

a model in which increased CLIP affinity for class II serves to<br />

restrict peptide loading to H-2M-containing compartments,<br />

ensuring proper editing <strong>of</strong> antigenic peptides.<br />

doi:10.1016/j.clim.2007.03.519<br />

Sa.133 Treg Potency Assay: Suppression <strong>of</strong> T Cell<br />

Cytokine Expression in Response to T Cell Specific<br />

Antigenic Stimulation <strong>of</strong> Autologous PBMC<br />

Smita Ghanekar, Senior Research Scientist, BD Biosciences,<br />

Research and Development, San Jose, CA, Joyce Ruitenberg,<br />

RAII, BD Biosciences, Research and Development, San Jose,<br />

CA<br />

CD4+CD25+ regulatory Tcells (Tregs) play a critical role in<br />

the induction and maintenance <strong>of</strong> peripheral immune<br />

tolerance. Naturally occurring Tregs are crucial for the<br />

control <strong>of</strong> auto-reactive T cells and <strong>of</strong> the effector function<br />

<strong>of</strong> allo-reactive CD4+ and CD8+ T cells in transplantation<br />

models in vivo. Tregs have been shown to be potent<br />

suppressors <strong>of</strong> activated T cells in vitro. Phenotypically,<br />

Tregs are characterized as T cells that are CD4+, CD25++,<br />

CD127− and Foxp3+. Suppressive ability <strong>of</strong> Tregs is usually<br />

assessed by applying long-term (5 days) assays such as<br />

proliferation, but the therapeutic potential <strong>of</strong> these cells<br />

demands a quicker analysis method. Therefore, in this study,<br />

the ability <strong>of</strong> Tregs from healthy donors to inhibit or suppress<br />

cytokine expression in short-term assays has been examined.<br />

PBMC enriched for CD4+ cells were purified as CD4+CD25++<br />

Tregs by sorting with a BD FACSAria. These highly purified<br />

Tregs were then cultured with rIL2 in vitro for 5–7 days and<br />

used in 6-h intracellular cytokine expression assays <strong>of</strong><br />

autologous PBMC. Activation <strong>of</strong> PBMC with SEB and/or<br />

CD3+CD28 results in the expression <strong>of</strong> inflammatory cytokines<br />

(IFNg, TNFa, and IL2) by CD4+ and CD8+ T cells. The<br />

results suggest that the addition <strong>of</strong> Tregs to the cytokine<br />

assay <strong>of</strong> autologous PBMC reduces the frequency <strong>of</strong> cytokine<br />

expressing T cells. Since the intracellular T cell cytokine<br />

expression assay has been validated extensively, this shortterm<br />

assay would be a valuable tool to qualify Tregs for<br />

immunotherapy.<br />

doi:10.1016/j.clim.2007.03.520<br />

Sa.134 β-Tubulin-induced Autoimmune<br />

Hearing Loss Exacerbates in IL-10-deficient Mice<br />

and Restores by IL-10 Gene Transfer<br />

Bin Zhou, Research Associate, University <strong>of</strong> Tennessee,<br />

Department <strong>of</strong> Medicine, Memphis, TN, Mohammad Habiby<br />

Kermany, Postdoctoral Fellow, University <strong>of</strong> Tennessee,<br />

Department <strong>of</strong> Medicine, Memphis, TN, Yixuan Zhou,<br />

Postdoctoral Fellow, University <strong>of</strong> Tennessee, Department<br />

<strong>of</strong> Medicine, Memphis, TN, Qing Cai, Postdoctoral Fellow,<br />

University <strong>of</strong> Tennessee, Department <strong>of</strong> Medicine, Memphis,<br />

TN, Chun Cai, Postdoctoral Fellow, University <strong>of</strong> Tennessee,<br />

Department <strong>of</strong> Medicine, Memphis, TN, Junwoo Kim,<br />

Postdoctoral Fellow, University <strong>of</strong> Tennessee, Department<br />

<strong>of</strong> Medicine, Memphis, TN, Patrick Kim, Postdoctoral<br />

Fellow, University <strong>of</strong> Tennessee, Department <strong>of</strong> Medicine,<br />

Memphis, TN, Wenxi Liu, Postdoctoral Fellow, University <strong>of</strong><br />

Tennessee, Department <strong>of</strong> Medicine, Memphis, TN, Tai June<br />

Yoo, Pr<strong>of</strong>essor, University <strong>of</strong> Tennessee, Department <strong>of</strong><br />

Medicine, Memphis, TN<br />

We hypothesized that endogenous levels <strong>of</strong> IL-10 function<br />

to regulate the severity <strong>of</strong> βtubulin-induced experimental<br />

autoimmune hearing loss (EAHL), and exogenous <strong>of</strong><br />

IL-10 would abrogate βtubulin-induced EAHL. BALB/c wildtype<br />

(WT) and homozygous (IL-10−/−) IL-10-deficient mice<br />

underwent βtubulin immunization, development <strong>of</strong> EAHL<br />

was monitored over time. Additional groups <strong>of</strong> IL-10deficient<br />

mice immunized βtubulin to induce EAHL; IL-10deficient<br />

mice were intramuscularly injected with plasmid<br />

DNA encoding IL-10 on the climax <strong>of</strong> EAHL. Auditory<br />

brainstem responses (ABR) were examined over time.<br />

EAHL developed progressively in both BALB/c WT and IL-<br />

10-deficient mice immunized with βtubulin. However, the<br />

severity <strong>of</strong> hearing loss in the IL-10−/− mice was significantly<br />

greater than that in WT animals. Disease severity was<br />

associated with significantly enhanced IFNγ and TNFα level<br />

from stimulated splenocyte cultures, increased IgG2a antiβtubulin<br />

antibody responses and degeneration <strong>of</strong> Spiral<br />

ganglion cells <strong>of</strong> the Cochlea. αtubulin immunized IL-10−/−<br />

mice receiving intramuscular injection <strong>of</strong> plasmid DNA<br />

encoding IL-10 developed high serum levels <strong>of</strong> IL-10 yet<br />

the hearing function was significantly improved in the IL-10treated<br />

groups than that in the vector control group.<br />

Moreover, the ABR thresholds in the IL-10-treated groups<br />

are equivalent to naive controls and the histological<br />

analysis <strong>of</strong> the cochlear cross section was similar to those<br />

<strong>of</strong> naive controls. Furthermore, IL-10-treated mice exhibited<br />

significant suppression <strong>of</strong> βtubulin-induced IFNγ and<br />

TNFγ, but increased IgG1 production compared to levels<br />

exhibited by control mice. Our data demonstrate that lack<br />

<strong>of</strong> IL-10 exacerbates hearing loss and exogenous administration<br />

<strong>of</strong> IL-10 improved hearing function.<br />

doi:10.1016/j.clim.2007.03.521<br />

Sa.135 Galectin-1 Selectively Suppresses<br />

Plasmacytoid DC<br />

James Chen, Student, University <strong>of</strong> California, Los Angeles,<br />

Medicine, Los Angeles, CA, Xiangshu Wen, Postdoctoral<br />

Fellow, University <strong>of</strong> California, Los Angeles, Medicine, Los<br />

Angeles, CA, Jennifer Fulcher, MD, PhD Student, University<br />

<strong>of</strong> California, Los Angeles, Microbiology <strong>Immunology</strong> and<br />

Molecular Genetics, Los Angeles, CA, Benhur Lee, Assistant<br />

Pr<strong>of</strong>essor, University <strong>of</strong> California, Los Angeles,<br />

Microbiology <strong>Immunology</strong> and Molecular Genetics, Los


Abstracts<br />

Angeles, CA, Anna Eriksson, Postdoctoral Fellow, University<br />

<strong>of</strong> California, Los Angeles, Medicine, Los Angeles, CA, Ram<br />

Raj Singh, Pr<strong>of</strong>essor, University <strong>of</strong> California, Los Angeles,<br />

Medicine, Los Angeles, CA<br />

We have recently reported that galectin-1, a β-galactoside-binding<br />

lectin with a broad range <strong>of</strong> immunomodulatory<br />

properties, binds to and activates human monocyte-derived<br />

dendritic cells (DC). To evaluate the implications <strong>of</strong> this<br />

finding in vivo, we have investigated the role <strong>of</strong> galectin-1 in<br />

the modulation <strong>of</strong> DC in mice. We first show that galectin-1<br />

strongly binds murine DC. To examine the effect <strong>of</strong> galectin-1<br />

on DC subsets, we cultured spleen cells from BALB/c mice<br />

with LPS or galectin-1. We found that whereas LPS induces<br />

CD40 expression on all DC subsets including myeloid<br />

(CD11c+CD11b+), lymphoid (CD11c+CD8a+) and plasmacytoid<br />

(pDC; CD11c+B220+) DC, galectin-1 induces CD40 expression<br />

only in a fraction <strong>of</strong> DC. Strikingly, galectin-1 reduces the<br />

percentage and numbers <strong>of</strong> pDC and their activation as<br />

determined by CD40 expression, whereas LPS causes their<br />

expansion and activation. To assess the effect <strong>of</strong> galectin-1 on<br />

pDC in vivo, we used autoimmune-prone MRL-lpr mice that<br />

have increased numbers <strong>of</strong> pDC accumulating in inflamed<br />

organs. A single s.c. injection <strong>of</strong> galectin-1 in these mice<br />

reduced the proportion <strong>of</strong> pDC in cutaneous lymph nodes.<br />

These data suggest a role <strong>of</strong> galectin-1 in the regulation <strong>of</strong><br />

pDC. B220 is a spliced is<strong>of</strong>orm <strong>of</strong> CD45 and thus, our data are<br />

consistent with the known ability <strong>of</strong> galectin-1 to trigger<br />

apoptotic cell death in T cells via cross-linking <strong>of</strong> CD45.<br />

Ongoing studies will investigate the implication <strong>of</strong> this finding<br />

on the development <strong>of</strong> systemic inflammation in MRL-lpr<br />

mice, and the possible therapeutic consequences <strong>of</strong> galecin-1<br />

administration in the murine lupus model.<br />

doi:10.1016/j.clim.2007.03.522<br />

Sa.136 T Cell Responsiveness to Complementary<br />

PR3 Protein Supports a Pathogenic Role <strong>of</strong><br />

Autoantigen Complementarity in PR3-ANCA<br />

Autoimmune Disease<br />

Jiajin Yang, Assistant Pr<strong>of</strong>essor, University <strong>of</strong> North Carolina<br />

at Chapel Hill Kidney Center, Chapel Hill, NC, David Bautz,<br />

Graduate Student, University <strong>of</strong> North Carolina at Chapel<br />

Hill Kidney Center, Chapel Hill, NY, John Smitz, Associate<br />

Pr<strong>of</strong>essor, University <strong>of</strong> North Carolina at Chapel Hill<br />

Department <strong>of</strong> Pathology and Laboratory Medicine, Chapel<br />

Hill, NC, Hyunsook Chin, Statistician II, University <strong>of</strong> North<br />

Carolina at Chapel Hill Kidney Center, Chapel Hill, NC,<br />

Barrrak Pressler, Assistant Pr<strong>of</strong>essor <strong>of</strong> Internal Medicine,<br />

Purdue University, Lafayette, IN, Susan Hogan, Research<br />

Assistant Pr<strong>of</strong>essor, Medicine, University <strong>of</strong> North Carolina<br />

at Chapel Hill Kidney Center, Chapel Hill, NC, Roland Tisch,<br />

Assoc Pr<strong>of</strong>essor, Microbiology and <strong>Immunology</strong>, Chapel Hill,<br />

NC, J. Charles Jennette, Distinguished Pr<strong>of</strong>essor,<br />

Department Chair, Pathology and Laboratory Medicine,<br />

Chapel Hill, NC, Ronald Falk, Distinguished Pr<strong>of</strong>essor/<br />

Division Chief/Director, University <strong>of</strong> North Carolina at<br />

Chapel Hill Kidney Center, Chapel Hill, NC, Gloria Preston,<br />

Pr<strong>of</strong>essor, University <strong>of</strong> North Carolina at Chapel Hill Kidney<br />

Center, Chapel Hill, NC<br />

We discovered that patients with PR3-ANCA vasculitis<br />

who have autoantibodies against the autoantigen proteinase<br />

3 (PR3) also have a subset <strong>of</strong> antibodies reactive<br />

against a protein complementary – or antisense – to the<br />

autoantigen, as detected using recombinant, complementary-PR3<br />

protein, cPR3. Investigations into the etiology and<br />

consequences <strong>of</strong> these anti-cPR3 antibodies led to the<br />

proposal that complementary proteins are involved in<br />

inciting autoantibody production, as detailed in the theory<br />

<strong>of</strong> autoantigen complementarity (Nat Med 2004, 10: 72–79).<br />

The present studies indicate that CD4+TH1 cells from<br />

patients (n=26) with PR3-ANCA vasculitis versus healthy<br />

controls (n=34) exhibit increased proliferation (Wilcoxon<br />

rank sum test, p=0.0014) and IFN-γ expression (p=b0.0001)<br />

when stimulated with cPR3-peptide (32 aa, 138–169) but<br />

not a scrambled peptide (p=0.60). These recall responses<br />

indicate that the responding T cells were activated prior in<br />

vivo. Reactivity to smaller, overlapping fragments <strong>of</strong> cPR3<br />

ruled out the possibility that cPR3-peptide functions as a<br />

superantigen. Interestingly, the HLA-DRB1*15 allele group<br />

was overrepresented in the patient group, as compared to<br />

frequencies from a Caucasian population US database<br />

(Fisher’s exact test, p=0.006). This allele group is predicted<br />

to bind cPR3 peptide with high affinity (based on the<br />

Immune Epitope and Analysis Resource database). Ranked<br />

linear regression analysis indicated a likelihood <strong>of</strong> p=0.009<br />

that anti-cPR3 antibodies and cPR3-specific T cells coexist<br />

within an individual. The T cell responses observed are<br />

consistent with a history <strong>of</strong> complementary-protein encounter<br />

in these patients. Consideration <strong>of</strong> potential contributions<br />

<strong>of</strong> complementary protein pairs in autoimmune<br />

diseases could revolutionize the approach for exploring<br />

pathogenic mechanisms.<br />

doi:10.1016/j.clim.2007.03.523<br />

S121<br />

Sa.137 Enhanced CD16 Mediated Depletion <strong>of</strong><br />

Activated Lymphocytes by a Non-Fucosylated Fully<br />

Human Anti-CD70 IgG1<br />

Hadia Lemar, Research Associate III, Medarex, Inc., Milpitas,<br />

CA, Diane Feingersh, Scientist I, Medarex, Inc., Milpitas,<br />

CA, Alison Witte, Scientist III, Medarex, Inc., Milpitas, CA,<br />

Jon Terrett, Director, Medarex, Inc., Milpitas, CA, Ben<br />

Preston, Research Assotciate III, Medarex, Inc., Milpitas,<br />

CA, Mark Selby, Director, Medarex, Inc., Milpitas, CA, Alan<br />

Korman, VP, Medarex, Inc., Milpitas, CA, Marco Coccia,<br />

Senior Scientist II, Medarex, Inc., Milpitas, CA<br />

CD70 is the only known ligand for CD27 mediated<br />

costimulation <strong>of</strong> B and T memory and effector responses.<br />

CD70 expression in normal tissues is restricted to activated<br />

lymphocytes and dendritic cells. Patients with autoimmune<br />

disorders express high levels <strong>of</strong> CD70 on chronically activated<br />

T cells and lymphocytes. Furthermore, CD70 blocking<br />

antibodies were shown to delay onset <strong>of</strong> experimental<br />

autoimmune encephalomyelitis and cardiac allograft rejection<br />

in mice. We demonstrate here that antibody dependent<br />

cellular cytotoxicity (ADCC) mediated depletion <strong>of</strong> hyperactivated<br />

CD70+ leukocytes may be an effective therapeutic<br />

strategy for autoimmunity. We previously reported that a


S122 Abstracts<br />

fully human, non-fucoslyated anti-human CD70 IgG1 (anti-<br />

CD70nf) mediated superior ADCC <strong>of</strong> CD70+ tumor cells<br />

compared to parental fucosylated IgG1 (anti-CD70p). Anti-<br />

CD70nf also stimulated ADCC <strong>of</strong> purified T cells activated by<br />

anti-CD3/anti-CD28 coated beads with ∼10-fold greater<br />

potency and ∼50% greater efficacy than anti-CD70p.<br />

Stimulation <strong>of</strong> PBMC with cytomegalovirus (CMV) peptide<br />

induced CD70 expression on responsive CD8+/CMV pentamer+<br />

cells. Anti-CD70p and anti-CD70nf decreased CD8+/CMV<br />

pentamer+ cells but depletion by anti-CD70nf was both<br />

more potent and efficacious. Anti-CD16 blocking was effective<br />

in reversing depletion by both antibodies, but 1000-fold<br />

greater concentration <strong>of</strong> anti-CD16 was required to inhibit<br />

depletion by anti-CD70nf. CD8+/CMV pentamer− cells were<br />

unaffected by either CD70 antibody. These CD70 antibodies<br />

also inhibited CD70-Fc binding to CD27+/CD70− RL cells and<br />

CD70 costimulation <strong>of</strong> T cells activated by anti-CD3 on CD32/<br />

CD70 CHO transfectants. Further evaluation <strong>of</strong> enhanced<br />

ADCC mediated depletion <strong>of</strong> activated leukocytes and anti-<br />

CD70 functional blockade as therapeutic strategies for<br />

autoimmunity is in progress.<br />

doi:10.1016/j.clim.2007.03.524<br />

Sa.138 Evidence for DC:Treg Paracrine IL-2 Delivery<br />

Katarina Kulhankova, Postdoctoral Scholar, Carver College<br />

<strong>of</strong> Medicine and Veterans Affairs Medical Center,<br />

Department <strong>of</strong> Internal Medicine, Iowa City, IA, Mohamed<br />

Nasr, Research Scientist, Carver College <strong>of</strong> Medicine and<br />

Veterans Affairs Medical Center, Department <strong>of</strong> Internal<br />

Medicine, Iowa City, IA, Michael E. Dailey, Pr<strong>of</strong>essor,<br />

University <strong>of</strong> Iowa, Department <strong>of</strong> Biological Sciences, Iowa<br />

City, IA, Todd Rouse, Research Assistant, Carver College <strong>of</strong><br />

Medicine and Veterans Affairs Medical Center, Department<br />

<strong>of</strong> Internal Medicine, Iowa City, IA, Elizabeth H. Field,<br />

Pr<strong>of</strong>essor, Carver College <strong>of</strong> Medicine and Veterans Affairs<br />

Medical Center, Department <strong>of</strong> Internal Medicine,<br />

Iowa City, IA<br />

Regulatory CD4+CD25+ Tcells (Tregs) play a key role in the<br />

maintenance <strong>of</strong> immune tolerance, and understanding the<br />

precise mechanism <strong>of</strong> their function can help guide<br />

therapeutic strategies for achieving transplant tolerance.<br />

Once activated, Tregs inhibit CD4+ cell proliferation and IL-2<br />

production in vitro. Treg cell function is blocked by culturing<br />

Tregs and effectors in the presence <strong>of</strong> anti-CD25 or<br />

separately in transwell system. This indicates that both IL-<br />

2 and cell:cell contact is necessary for the gain or execution<br />

<strong>of</strong> Treg-mediated suppression. However, the source for IL-2<br />

and the context, in which it is delivered, remains unknown.<br />

To address this question we generated the CD4+DC25+<br />

hybridoma RD6 from a mouse with acquired tolerance to<br />

foreign MHCs. Like natural Tregs, RD6 inhibition <strong>of</strong> CD4+<br />

effectors requires CD25 and cell:cell contact. Neither Treg<br />

nor RD6 showed inhibition when IL-2KO DCs were substituted<br />

for WT DCs in Treg assays. We utilized four-dimensional<br />

multi-channel fluorescent live-cell confocal microscopy to<br />

define the CD25 expression on RD6 cells during cell:cell<br />

interactions between RD6 and semi-allogeneic DCs or<br />

allogeneic membrane-labeled DC cells, JAWSII. RD6 engaged<br />

themselves and DCs in a variety <strong>of</strong> cell:cell interactions that<br />

included long-lasting conjugate formation as well as formation<br />

<strong>of</strong> stable cell:cell membrane bridges <strong>of</strong> various lengths.<br />

RD6 CD25 expression localized to points <strong>of</strong> contact with<br />

dendritic processes over 24 hours. RD6 cells acquired patches<br />

<strong>of</strong> DC membranes and internalized them. These data are<br />

consistent with a model, in which DCs deliver IL-2 to Tregs via<br />

tight membrane contacts.<br />

doi:10.1016/j.clim.2007.03.525<br />

Sa.139 Admixture Mapping, a New Tool for Disease<br />

Gene Finding in Autoimmune and Other Diseases<br />

Alicja Waliszewska, Research Specialist, Broad Institute,<br />

Cambridge, MA, Phillip De Jager, Assistant Pr<strong>of</strong>essor,<br />

Department <strong>of</strong> Genetics, Harvard Medical School, Boston<br />

MA, David Hafler, Pr<strong>of</strong>essor, Department <strong>of</strong> Genetics,<br />

Harvard Medical School, Boston MA, David Reich, Assistant<br />

Pr<strong>of</strong>essor, Department <strong>of</strong> Genetics, Harvard Medical School,<br />

Boston, MA<br />

Admixture mapping is a powerful technique for finding<br />

genes for complex diseases. The practical requirements<br />

include (a) a set <strong>of</strong> markers spanning the genome with large<br />

allele-frequency differences between the parental ethnicities<br />

contributing to the admixed population and (b) an<br />

understanding <strong>of</strong> the extent <strong>of</strong> admixture in the study<br />

population. African American and Latino American populations<br />

are the product <strong>of</strong> gene flow (genetic admixture) during<br />

the last 500 years, which has produced new genotypes from<br />

once-isolated ancestral groups. Admixture mapping attempts<br />

to associate African, European, or Native American to disease<br />

risk. We have performed admixture scans in African Americans<br />

to find genes affecting Multiple Sclerosis (MS),<br />

Rheumatoid Arthritis (RA), markers <strong>of</strong> inflammation, and<br />

prostate cancer. We have also been building resources for<br />

admixture mapping in Latinos. We have detected loci that are<br />

significantly associated to MS on chromosome 1 (LOD=5.2), to<br />

prostate cancer on chromosome 8 (LOD=7.1), and to<br />

interleukin 6 soluble receptor (IL6 SR) on chromosome 1<br />

(LOD=4.59). For IL6 SR and prostate cancer this has led to<br />

cloning <strong>of</strong> disease risk alleles. In African Americans with<br />

rheumatoid arthritis, the scan was negative, and identified<br />

no loci for disease risk.<br />

doi:10.1016/j.clim.2007.03.526<br />

Sa.140 Comparison <strong>of</strong> Membrane-bound and<br />

Secreted TGFβ by IL-2 Activated Natural and<br />

Adaptive CD4+CD25+ Regulatory Cells from Healthy<br />

and Lupus-prone Mice<br />

Song Guo Zheng, Assistant Pr<strong>of</strong>essor <strong>of</strong> Research Medicine,<br />

University <strong>of</strong> Southern California Keck School <strong>of</strong> Medicine,<br />

Los Angeles, CA, Julie Wang, Research Lab Specialist,<br />

University <strong>of</strong> Southern California Keck School <strong>of</strong> Medicine,<br />

Los Angeles, CA, Shuang Ye, Postdoctoral Fellow, University<br />

<strong>of</strong> Southern California Keck School <strong>of</strong> Medicine, Los Angeles,<br />

CA, David Sehy, Director, New Technologies, eBioscience/<br />

Department <strong>of</strong> New Technologies, San Diego, CA, Beth


Abstracts<br />

Palian, Postdoctoral Student, University <strong>of</strong> Southern<br />

California Keck School <strong>of</strong> Medicine, Los Angeles, CA, David<br />

A. Horwitz, Pr<strong>of</strong>essor Chief, Division <strong>of</strong> Rheumatology and<br />

<strong>Immunology</strong>, University <strong>of</strong> Southern California Keck School<br />

<strong>of</strong> Medicine, Los Angeles, CA<br />

CD4+CD25+Foxp3+ regulatory T cells can be divided into<br />

natural, thymus-derived cells and adaptive Tregs induced<br />

from CD25− precursors in the periphery. Although TGFβ<br />

induced, adaptive Tregs produce TGFβ, whether natural<br />

CD4+CD25+ Tregs also produce this cytokine has been<br />

controversial. Mouse CD4+CD25+ cells were stimulated via<br />

TCR and/or IL-2 and naive CD4+CD25− cells were stimulated<br />

with TGFβ to induce adaptive Th3 cells. Although<br />

freshly isolated murine CD4+CD25+ cells did not express<br />

mature TGFβ, when TCR activated with IL-2, these Foxp3+<br />

Treg cells displayed similar numbers <strong>of</strong> membrane-bound<br />

TGFβ under appropriate staining conditions as Th3 cells<br />

induced with TGFβ ex vivo. Both natural and adaptive Treg<br />

subsets also secreted similar amounts <strong>of</strong> active TGFβ. Since<br />

TGFβ induces CD8+ cells to express α-E integrin (CD103),<br />

we found the activated CD25+ Tregs induced CD103<br />

expression on CD8+ cells to levels equivalent to that <strong>of</strong><br />

activated Th3 cells. ALK5 (TGFβ receptor I inhibitor), but<br />

not anti-TGFβ almost completely abolished this CD103<br />

expression. Transwell experiments revealed that surface<br />

contact was not required. Thus, activated natural CD4+<br />

CD25+ Treg cells can produce similar levels <strong>of</strong> mature TGFβ<br />

as adaptive TGFβ induced Tregs. Finally, we studied<br />

activated CD4+CD25+ cells from lupus-prone (NZBxNZW)<br />

F1 young and old mice and found that cells from old mice<br />

with established disease expressed decreased membranebound<br />

and produced less soluble TGFβ. Thus, defective<br />

TGFβ production by CD4+CD25+ Tregs may contribute to<br />

their decreased functional activity and to the development<br />

and progression <strong>of</strong> systemic lupus.<br />

doi:10.1016/j.clim.2007.03.527<br />

Sa.141 Thymic Organogenesis Controlled by<br />

NF-kappaB Activating Factors<br />

Masashi Ya, Graduate Student, Institute for Enzyme<br />

Research, University <strong>of</strong> Tokushima, Tokushima, Japan,<br />

Yasuhiro Mouri, Graduate Student, Institute for Enzyme<br />

Research, University <strong>of</strong> Tokushima, Tokushima, Japan,<br />

Mitsuru Matsumoto, Pr<strong>of</strong>essor, Institute for Enzyme<br />

Research, University <strong>of</strong> Tokushima, Tokushima, Japan<br />

Thymic epithelial cells (TEC) play pivotal roles in the<br />

establishment <strong>of</strong> self-tolerance through critical dialogue<br />

with developing thymocytes. Unique actions <strong>of</strong> two NFkappaB<br />

activating factors within TECs, NF-kappaB-inducing<br />

kinase (NIK) and IkappaB kinase α (IKKα), for the establishment<br />

<strong>of</strong> self-tolerance have recently been highlighted by<br />

studies using a strain <strong>of</strong> mouse bearing a natural mutation<br />

<strong>of</strong> the NIK gene (aly mice) and gene-targeted mice,<br />

respectively. Previous studies have demonstrated essential<br />

roles <strong>of</strong> NIK-IKKα downstream <strong>of</strong> the lymphotoxin-beta<br />

receptor (LTβR), which is essential for the development <strong>of</strong><br />

secondary lymphoid organs; aly mice lack all lymph nodes<br />

and Peyer’s patches because <strong>of</strong> the defective LTβR<br />

signaling. Now additional roles <strong>of</strong> NIK-IKKα in thymic<br />

organogenesis, mainly through the developmental regulation<br />

<strong>of</strong> TECs, have emerged, although the corresponding<br />

upstream receptor(s) and ligand(s) participating in this<br />

action have not been fully characterized. Previous studies<br />

have suggested that LTβR regulates thymic organogenesis.<br />

In order to investigate the contribution <strong>of</strong> LTβR signaling for<br />

the thymic organogenesis in NIK-IKKα-dependent pathways,<br />

we have analyzed thymic architecture together with Aire<br />

expression from both LTα-deficient mice and LTβR-deficient<br />

mice. We found that Aire-expressing cells were largely<br />

retained from those mice, whereas they were dramatically<br />

reduced from both NIK-mutant mice and IKKα-deficient<br />

mice. Because NIK-mutant mice and IKKα-deficient mice<br />

show more pr<strong>of</strong>ound thymic disorganization <strong>of</strong> TECs than<br />

that from LTβR-deficient mice, it is likely that NIK-IKKα is<br />

additionally acting downstream on other receptor(s) beyond<br />

LTβR in this process.<br />

doi:10.1016/j.clim.2007.03.528<br />

S123<br />

Sa.142 A FLIPR-based Assay to Assess Potency <strong>of</strong><br />

Inhibitors <strong>of</strong> the TEC Family Kinases Itk and Btk<br />

John Douhan III, Principal Research Scientist, Wyeth<br />

Research, Inflammation, Cambridge, MA, Joy Miyashiro,<br />

Research Scientist, Wyeth Research, Inflammation,<br />

Cambridge, MA, Xiaochuan Zhou, Research Scientist, Wyeth<br />

Research, Inflammation, Cambridge, MA, Paul Wu, Research<br />

Scientist, Wyeth Research, Inflammation, Cambridge, MA,<br />

Derek Cole, Principal Research Scientist, Wyeth Research,<br />

Chemical and Screening Sciences, Pearl River, NY, Mary<br />

Collins, Vice President, Wyeth Research, Inflammation,<br />

Cambridge, MA, Kyriaki Dunussi-Joannopoulos, Associate<br />

Director, Wyeth Research, Inflammation, Cambridge, MA<br />

Bruton’s tyrosine kinase (Btk) and interleukin-2-inducible<br />

Tcell kinase (Itk), members <strong>of</strong> the TEC family <strong>of</strong> nonreceptor<br />

protein tyrosine kinases, are expressed primarily in B and T<br />

cells respectively and are critically involved in lymphocyte<br />

development and signal transduction. In particular, both Btk<br />

and Itk regulate calcium mobilization subsequent to antigen<br />

receptor stimulation which in turn regulates downstream<br />

effector functions such as proliferation, antibody secretion<br />

and cytokine production. Small molecule antagonists <strong>of</strong> Btk<br />

and Itk may therefore be useful in treating inflammatory and<br />

autoimmune conditions. We have developed a FLIPR-based<br />

assay which takes advantage <strong>of</strong> Btk-deficient DT40 chicken B<br />

cells. It has been reported that these cells are unable to<br />

mobilize calcium in response to cross-linking <strong>of</strong> their B cell<br />

receptor and that ectopic expression <strong>of</strong> human Btk or Itk can<br />

restore signaling upon B cell receptor stimulation. Here we<br />

report that not only the expression <strong>of</strong> wild type human Btk,<br />

but also the expression <strong>of</strong> a chimeric Btk–Itk kinase molecule<br />

can restore calcium responses in Btk-deficient DT40 cells. We<br />

have generated stable cell lines expressing either full length<br />

human Btk or a chimeric human Btk–Itk molecule—a Btk<br />

protein whose kinase domain has been replaced by that <strong>of</strong><br />

Itk. Calcium responses in both cell lines can be inhibited by<br />

the tyrosine kinase inhibitor staurosporine, but only the


S124 Abstracts<br />

chimeric Btk-Itk DT40 line is inhibited by an Itk-specific<br />

kinase inhibitor. We demonstrate that potency and selectivity<br />

<strong>of</strong> inhibitors <strong>of</strong> Itk and Btk can be assessed in this FLIPR<br />

cell-based assay.<br />

doi:10.1016/j.clim.2007.03.529<br />

Sa.143 Anti-Inflammatory Activities <strong>of</strong> Stilbene<br />

Analogs for Targeting Autoimmune Diseases<br />

Liren Tang, Scientist, Welichem Biotech Inc., Burnaby, BC,<br />

Canada, Genhui Chen, Celestial Pharmaceuticals Ltd,<br />

Shenzhen, China, Bin Li, Scientist, Welichem Biotech Inc.,<br />

Burnaby, BC, Canada, Quanhai Liu, Pr<strong>of</strong>essor, Department <strong>of</strong><br />

Pharmacology, Shanghai Instititue <strong>of</strong> Pharmaceutical<br />

Industry, Shanghai, China, Michael Lyle, Postdoctoral<br />

Fellow, Welichem Biotech Inc., Burnaby, BC, Canada, John<br />

Webster, Pr<strong>of</strong>essor and CSO, Welichem Biotech Inc., Simon<br />

Fraser University, Burnaby, BC, Canada<br />

Certain species <strong>of</strong> symbiotic bacteria (Enterobacteriaceae),<br />

when released into an insect by their nematode<br />

vector, released metabolites that modulate the insect’s<br />

innate immune response. These metabolites (stilbene<br />

analogs, in the WBI-1000 series) possess unique antiinflammatory<br />

properties. In general, they are weakly<br />

cytotoxic to mammalian cells, being more active on<br />

activated than on quiescent T cells or other normal cells.<br />

When tested in vitro, the WBI-1000 series selectively<br />

inhibited IL-2 and IFN-γ production in human T cells, and<br />

significantly inhibited TNF-α production in mice, but IL-4 was<br />

not affected. As well, they significantly inhibited T cell<br />

migration towards LTB4 in vitro. When topically applied in<br />

the TPA-induced ear edema mouse model, WBI-1000 compounds<br />

significantly reduced both skin redness and thickness.<br />

To further explore the clinical applications <strong>of</strong> these<br />

compounds one <strong>of</strong> them, WBI-1001 was tested for its efficacy<br />

on different animal models for autoimmune diseases. WBI-<br />

1001 showed dose-related efficacy in both the vaginal<br />

mucosa and mouse tail models for psoriasis as a topical<br />

treatment, significant clinical improvement in dextran<br />

sodium sulphate (DSS)-induced inflammatory bowel disease<br />

(IBD) in mice and decreased inflammation-induced arthritis<br />

in rats when compared with indomathecin. In conclusion,<br />

due to its unique anti-inflammatory properties, including<br />

selectivity on activated T cells, strong inhibition <strong>of</strong> proinflammatory<br />

cytokines and T cell migration, the WBI-1000<br />

series <strong>of</strong> compounds could be developed into effective, novel<br />

treatment modalities for autoimmune diseases.<br />

doi:10.1016/j.clim.2007.03.551<br />

Sa.145 Functional Knockout <strong>of</strong> Kv1.3 Channels<br />

Suppresses Effector Memory Phenotype Acquisition<br />

in Dominant Negative Kv1.3-expressing Human<br />

CD4+ T Cells<br />

Lina Hu, Research Associate, Department <strong>of</strong> Neurology,<br />

Johns Hopkins School <strong>of</strong> Medicine, Baltimore, MD, Katharine<br />

Whartenby, Assistant Pr<strong>of</strong>essor, Sidney Kimmel Cancer<br />

Center, Johns Hopkins School <strong>of</strong> Medicine, Baltimore, MD,<br />

Rameeza Allie, Research Specialist, Department <strong>of</strong><br />

Neurology, Johns Hopkins School <strong>of</strong> Medicine Baltimore, MD,<br />

Peter Calabresi, Associate Pr<strong>of</strong>essor, Department <strong>of</strong><br />

Neurology, Johns Hopkins School <strong>of</strong> Medicine, Baltimore, MD<br />

Activated effector memory T cells (TEM) that highly<br />

express the voltage-gated potassium channel Kv1.3 have<br />

the potential <strong>of</strong> migrating to the CNS and are hypothesized<br />

to play a pathogenic role in multiple sclerosis (MS). The<br />

pharmacological effects <strong>of</strong> Kv1.3 channel blockade on TEM<br />

cell activation and proliferation are well documented.<br />

However, the functional relevance <strong>of</strong> Kv1.3 in the homeostatic<br />

maintenance <strong>of</strong> TEM is poorly understood. Herein,<br />

using a system in which Kv1.3-channel function was blocked<br />

by transduction <strong>of</strong> CD4+ T cells with a GFP-tagged,<br />

lentiviral vector expressing dominant-negative Kv1.x<br />

sequence, we studied the kinetic changes in TEM subset<br />

within dominant negative Kv1.3-expressing CD4+ T cells<br />

following anti-CD3/CD28 stimulation. The interference <strong>of</strong><br />

endogenous Kv1.3 by misexpression <strong>of</strong> dominant-negative<br />

Kv1.x markedly attenuated the wild-type current. Analysis<br />

<strong>of</strong> GFP expression in CD4+ T cell subsets demonstrated that<br />

while a substantial reservoir <strong>of</strong> TCM was maintained, TEM<br />

cells expressing the dominant-negative Kv1.3 product were<br />

significantly less abundant than that infected with GFP<br />

control viruses at 2, 3 and 4 weeks after transduction. In<br />

contrast, the ratio between TEM and TCM cells in GFP<br />

control transfected cells is strongly biased toward the TEM.<br />

Thus, the generation and maintenance <strong>of</strong> TEM cells are<br />

markedly impaired by the inactivation <strong>of</strong> Kv1.3 channel<br />

function. These results <strong>of</strong>fer new insights into the<br />

functional dependence <strong>of</strong> TEM cells on Kv1.3, and have<br />

important clinical implications for using Kv1.3 blockers to<br />

prevent the generation <strong>of</strong> TEM cells, thereby abrogating<br />

the pathologic consequences <strong>of</strong> this subset in autoimmune<br />

diseases, such as MS.<br />

doi:10.1016/j.clim.2007.03.532<br />

Sa.146 Tim-4 Expressed on Antigen-presenting Cells<br />

Induces T Cell Expansion and Survival<br />

Roselynn Rodriguez Manzanet, Graduate Student, Brigham<br />

and Women’s Hospital, Boston, MA, Jennifer Hartt-Meyers,<br />

Post doctoral Fellow, LIR/NIAID/NIH, Bethesda, MD,<br />

Jacqueline Slavik, Administrative Director, Brigham and<br />

Women’s Hospital, Biomedical Research Institute, Boston,<br />

MA, Valerie Dardalhon, Post doctoral Fellow, Brigham and<br />

Women’s Hospital, Department <strong>of</strong> Neurology, Boston, MA,<br />

Nasim Kassam, Research specialist, Brigham and Women’s<br />

Hospital, Department <strong>of</strong> Neurology, Boston, MA, Edward A.<br />

Greenfield, Consultant, Brigham and Women’s Hospital,<br />

Department <strong>of</strong> Neurology, Boston, MA, Raymond A. Sobel,<br />

Pr<strong>of</strong>essor, Stanford School <strong>of</strong> Medicine, Department <strong>of</strong><br />

Pathology, Palo Alto, CA, Terry B. Strom, Physician<br />

Pr<strong>of</strong>essor, Beth Israel Deaconess Medical Center, Boston,<br />

MA, David A. Hafler, Pr<strong>of</strong>essor, Brigham and Women’s<br />

Hospital, Department <strong>of</strong> Neurology, Boston, MA, Vijay K.<br />

Kuchroo, Pr<strong>of</strong>essor, Brigham and Women’s Hospital,<br />

Department <strong>of</strong> Neurology, Boston, MA


Abstracts<br />

The TIM (T cell, immunoglobulin, mucin) proteins characterized<br />

to date have been shown to regulate Tcell immune<br />

responses. Tim-4 has been shown to be a ligand for Tim-1 and<br />

Tim-4.Ig fusion protein could both inhibit and activate T cell<br />

proliferation in in vitro assays. Further studies <strong>of</strong> this<br />

molecule have been hindered by the lack <strong>of</strong> a specific<br />

monoclonal antibody. Thus, we have generated three<br />

monoclonal anti-Tim-4 antibodies and show that Tim-4<br />

protein expression is restricted to the antigen-presenting<br />

cell population, specifically CD11c+ and CD11b+ cells. We<br />

demonstrate that Tim-4 expression is upregulated upon<br />

cellular activation, and that ectopic expression <strong>of</strong> Tim-4 on<br />

artificial APCs induces massive T cell proliferation. Additionally,<br />

in vitro crosslinking <strong>of</strong> Tim-4 ligand on the surface <strong>of</strong> T<br />

cells directly induced cell division and anti-apoptotic protein<br />

Bcl-2. We conclude that Tim-4 is a costimulatory molecule<br />

capable <strong>of</strong> promoting T cell expansion and show that it can<br />

enhance both T cell proliferation and survival.<br />

doi:10.1016/j.clim.2007.03.533<br />

Sa.147 Interleukin-2 Mastering Regulation in Cancer<br />

and Autoimmunity<br />

Enrique Montero, MD, PhD, Center <strong>of</strong> Molecular<br />

<strong>Immunology</strong>, Department <strong>of</strong> Experimental Immunotherapy,<br />

Havana, Cuba, Livan Alonso, BSc in Physics, Center <strong>of</strong><br />

Molecular <strong>Immunology</strong>, Department <strong>of</strong> Experimental<br />

Immunotherapy, Havana, Cuba, Rolando Perez, PhD, Center<br />

<strong>of</strong> Molecular <strong>Immunology</strong>, Havana, Cuba, Agustin Lage, MD,<br />

PhD, Center <strong>of</strong> Molecular <strong>Immunology</strong>, Havana, Cuba<br />

Autoimmunity and tumor immunity have evolved as two<br />

well distant arenas in immunological research. However, the<br />

identification <strong>of</strong> self-antigens as the major components <strong>of</strong><br />

malignant cells may define a central role for autoimmunity in<br />

cancer control tuned by peripheral immunoregulatory<br />

mechanisms avoiding self-aggression. Interleukin-2 (IL-2)<br />

paradoxical effects after in vitro or in vivo manipulations<br />

may reflect the complex interplay between immunoregulatory<br />

mechanisms. Emerging evidences support the contribution<br />

<strong>of</strong> IL-2 to the maintenance <strong>of</strong> natural immunological<br />

tolerance. It encourages the systematic evaluation in vivo <strong>of</strong><br />

formulations with IL-2 neutralization capacity compared<br />

with IL-2 exogenous administration to explore their influence<br />

on immunobiology in cancer and autoimmunity. IL-2 chemically<br />

conjugated to the carrier protein P64k from Neisseria<br />

Meningitides in Montanide ISA51 adjuvant was used for active<br />

immunization. We found that BALB/c, C57BL/6 and NMRI<br />

mice developed a long-lasting immunoresponse to IL-2 after<br />

immunization, without signs <strong>of</strong> autoimmune diseases.<br />

Immune sera had a similar IL-2 blocking effect in vitro to a<br />

specific anti-IL-2 monoclonal antibody (mAb). Interestingly,<br />

IL-2 deprivation did not affect the immunoresponse to<br />

therapeutic vaccines; however, it restores the response to<br />

nominal antigens in immunosuppressed hosts. Surprisingly,<br />

we found a differential effect <strong>of</strong> anti-IL-2 and anti-CD25 mAb<br />

on anti-tumor immunoresponse that may represent a broader<br />

impact <strong>of</strong> IL-2 on immunoregulation beyond its effect on<br />

CD4+CD25+ regulatory T cells. Moreover, the main effect <strong>of</strong><br />

IL-2 administration was associated with an enhancement <strong>of</strong><br />

immunoregulation. We conclude that IL-2 plays a major role<br />

in the interplay between cancer and autoimmunity with<br />

therapeutic implications.<br />

doi:10.1016/j.clim.2007.03.534<br />

Sa.148 Decrease in Memory B Cell Frequency and<br />

Activation is Associated with PTPN22 1858T Variant<br />

in Healthy Subjects<br />

Mary Rieck, Research Technician II, Benaroya Research<br />

Institute, Seattle, WA, Patric Concannon, Pr<strong>of</strong>essor, Center<br />

for Public Health Genomic, University <strong>of</strong> Virginia,<br />

Charlotte, SC, Adrian Arechiga, Research Associate,<br />

Benaroya Research Institute, Seattle, WA, Jane Buckner,<br />

Associate Member, Benaroya Research Institute, Seattle,<br />

WA, Suna Onengut-Gumuscu, Research Associate, Center for<br />

Health Genomic, Charlotte, SC<br />

It is known that the 1858T variant <strong>of</strong> the PTPN22 gene is<br />

associated with multiple autoimmune disorders, yet the<br />

functional consequences <strong>of</strong> this variant and how they<br />

contribute to autoimmunity are still unknown. PTPN22<br />

encodes the protein tyrosine phosphatase, Lyp, which<br />

participates in the proximal events <strong>of</strong> TCR and BCR signaling.<br />

The 1858T mutations results in an R and #61664;W amino acid<br />

change at residue 620 <strong>of</strong> Lyp. In Tcells Lyp620W dampens TCR<br />

signaling, however, its effects on the B cell compartment are<br />

not known. In this study we address this question by<br />

examining B cell function and phenotype in control subjects<br />

possessing the 1858T allele (1858C/T) as compared to<br />

subjects without the variant (1858C/C). These studies reveal<br />

no difference in percentage or number <strong>of</strong> total B cells in the<br />

periphery. However, within the B cell pool there was a<br />

decrease in the percent <strong>of</strong> memory B cells, defined as<br />

CD19+CD27+, in 1858C/T versus 1858C/C subjects (p=0.01).<br />

This decrease in the memory pool was even more pr<strong>of</strong>ound in<br />

autoimmune subjects homozygous for the 1858T variant<br />

(p=0.002). In addition, we find that the response <strong>of</strong> B cells to<br />

BCR stimulation, as measured by calcium flux is diminished in<br />

subjects carrying the 1858T allele, this was most pr<strong>of</strong>ound in<br />

the memory subset. These studies demonstrate that the<br />

PTPN22 1858T variant leads to altered B cell subsets and<br />

function. This suggests the possibility that 1858Tacts through<br />

B cell intrinsic mechanisms that contribute to the pathogenesis<br />

<strong>of</strong> autoimmunity in individuals with this variant.<br />

doi:10.1016/j.clim.2007.03.535<br />

S125<br />

Sa.149 Deconvolution <strong>of</strong> Blood Microarray Data<br />

Elucidates Cellular Activation Patterns in SLE<br />

Alexander R. Abbas, Genentech Inc., South San Francisco,<br />

CA, Kristen Wolslegel, Genentech Inc., South San Francisco,<br />

CA, Dhaya Seshasayee, Genentech Inc., South San Francisco,<br />

CA, Hilary F. Clark, Genentech Inc., South San Francisco, CA<br />

Understanding the biology <strong>of</strong> a disease <strong>of</strong>ten depends on<br />

knowing the different types and states <strong>of</strong> cells in biological


S126 Abstracts<br />

samples from relevant organs. The proportions <strong>of</strong> cells in<br />

blood or tissue are traditionally determined using methods<br />

that rely on one or two genes' expression or protein products,<br />

such as FACS or immunohistochemistry. These techniques<br />

detect only a few cell types at once and <strong>of</strong>ten cannot<br />

determine the state <strong>of</strong> cells. Here we present a method for<br />

using gene expression pr<strong>of</strong>iles <strong>of</strong> purified leukocyte cell types<br />

to probe the levels <strong>of</strong> cell types and cell states in whole blood<br />

samples. We demonstrate that this method accurately<br />

deconvolves mixed samples <strong>of</strong> known composition. We<br />

deconvolve blood samples from healthy donors and systemic<br />

lupus erythematosus patients to reveal patterns <strong>of</strong> cellular<br />

activation that apparently underlie this disease's prominent<br />

interferon signature.<br />

doi:10.1016/j.clim.2007.03.536<br />

Sa.150 Characterization <strong>of</strong> the Psoriasis-associated<br />

IL12B and IL23R Genes<br />

Ann Begovich, Director, Celera, Alameda, CA, Monica<br />

Chang, Senior Associate Scientist, Celera, Alameda, CA,<br />

Mark Leppert, Pr<strong>of</strong>essor, Department <strong>of</strong> Human Genetics,<br />

University <strong>of</strong> Utah, Salt Lake City, UT, Steven Schrodi,<br />

Senior Scientist, Celera, Alameda, CA, Gerald Krueger,<br />

Pr<strong>of</strong>essor, Department <strong>of</strong> Dermatology, University <strong>of</strong> Utah,<br />

Salt Lake City, UT<br />

Common genetic variants in IL12B and IL23R have been<br />

associated with psoriasis risk. To further investigate the<br />

role <strong>of</strong> these genes in susceptibility to psoriasis and other<br />

autoimmune diseases, we have resequenced both genes in<br />

96 individuals with psoriasis, identified novel SNPs and<br />

genotyped a subset <strong>of</strong> these and others from public<br />

databases in three independent white North American<br />

case–control sample sets (1446 cases and 1432 controls).<br />

To date, allelic and genotypic data for 51 IL12B-region and<br />

46 IL23R-region SNPs have been analyzed and preliminary<br />

haplotype analyses have been performed. The data suggest<br />

that association <strong>of</strong> IL12B with psoriasis is driven by two<br />

SNPs, rs3212227 and rs6887695, or SNPs in significant LD<br />

with these two markers. A sliding-window <strong>of</strong> haplotype<br />

association co-localizes psoriasis susceptibility within the<br />

boundaries <strong>of</strong> IL23R and not IL12RB2, which lies directly<br />

adjacent. Further genotyping in this region is ongoing to<br />

better define the IL23R genetic variants responsible for the<br />

association with psoriasis. We have also assessed whether<br />

the two psoriasis-associated IL12B SNPs are associated with<br />

specific clinical characteristics including age <strong>of</strong> onset,<br />

family history, psoriatic arthritis, and the effect <strong>of</strong><br />

infection on psoriasis severity in the two sample sets<br />

with complete phenotype data. The only interesting<br />

replicated finding was that the effect <strong>of</strong> rs6887695 may<br />

be significantly different when stratifying by response to<br />

infection (Breslow-Day P=0.04 and 0.043). Finally, the role<br />

<strong>of</strong> these SNPs in susceptibility to rheumatoid arthritis and<br />

multiple sclerosis will be addressed.<br />

doi:10.1016/j.clim.2007.03.537<br />

Sa.151 Elifacs: A Multiplex Assay to Detect Antigen<br />

Specific T Cells and to Monitor the Immune<br />

Response<br />

Khadir Raddassi, Research Instructor, Brigham and Women’s<br />

Hospital, Neurology, Boston, MA, Kasia Bouchier, Scientist,<br />

Immune Tolerance Network, San Francisco, CA, Jose<br />

Estevam, Research Assistant, Brigham and Women’s<br />

Hospital, Neurology, Boston, MA, Vicki Seyfert-Margolis,<br />

Scientist, Immune Tolerance Network, San Francisco, CA,<br />

David Hafler, Lab Director, Brigham and Women’s Hospital,<br />

Neurology, Boston, MA<br />

As part <strong>of</strong> the Immune Tolerance Network assay development<br />

group, we optimized a multiplex cell based assay to<br />

detect antigen reactive T cells. Cell assays to detect antigen<br />

specific T cells require large cell numbers. The Elispot assay<br />

is fast and sensitive but provides limited information<br />

regarding individual cells. In contrast, flow cytometry allows<br />

better characterization <strong>of</strong> antigen reactive T cells. Similarly,<br />

measuring thymidine incorporation provides no information<br />

regarding T cell function. Here, we combined the three to<br />

monitor T cell response to antigens using the same sample.<br />

Fresh or cryopreserved PBMCs were stained with CFSE, and<br />

exposed to tetanus toxoid or myelin antigens. After 42 h the<br />

cells were transferred to Elispot plates for 16 h; the Elispot<br />

plates were developed for cytokine expression, and the cells<br />

were collected and cultured for an additional 5 days before<br />

measuring intracellular cytokines and proliferation by flow<br />

cytometry. The combination <strong>of</strong> Elispot and flow cytometry<br />

data did not alter the specificity or sensitivity <strong>of</strong> these<br />

techniques. The recovery and viability <strong>of</strong> the cells from the<br />

Elispot plate were above 90%. There was a tight correlation<br />

(r 2 =0.960) between these data obtained by Elispot (cytokine<br />

positive cells) and by flow cytometry (CFSE and the cytokine<br />

production). We were able to measure IFNg, IL-10, IL-4,<br />

TGFβ and proliferative responses from the same sample. This<br />

technique allows us to extract more information from the<br />

same sample and thus provides a useful tool in monitoring<br />

the immune response.<br />

doi:10.1016/j.clim.2007.03.538<br />

Sa.152 Population-based Studies <strong>of</strong> Risk Factors in<br />

Autoimmunity: The Kaiser Permanente<br />

Autoimmune Disease Research Group (KPADRG)<br />

Lisa Barcellos, Assistant Pr<strong>of</strong>essor, UC Berkeley, School <strong>of</strong><br />

Public Health, Berkeley, CA, Ling Shen, Programmer/<br />

Analyst, Kaiser Permanente Division <strong>of</strong> Research, Oakland,<br />

CA, Lisa Herrinton, Epidemiologist/Senior Researcher,<br />

Kaiser Permanente Division <strong>of</strong> Research, Oakland, CA, Allan<br />

L. Bernstein, Chief <strong>of</strong> Neurology, Kaiser Permanente Santa<br />

Rosa, Kaiser Permanente Division <strong>of</strong> Research, Oakland, CA,<br />

James E. Allison, Adjunct Investigator, Kaiser Permanente<br />

Division <strong>of</strong> Research, Oakland, CA, John Citron,<br />

Endocrinologist, Kaiser Permanente Walnut Creek, Kaiser<br />

Permanente Division <strong>of</strong> Research, Oakland, CA, Stanford<br />

Shoor, Kaiser Permanente Santa Clara, Kaiser Permanente<br />

Division <strong>of</strong> Research, Oakland, CA, Andrew Karter,<br />

Epidemiologist/Senior Researcher, Kaiser Permanente<br />

Division <strong>of</strong> Research, Oakland, CA, De-Kun Li,


Abstracts<br />

Epidemiologist/Senior Researcher, Kaiser Permanente<br />

Division <strong>of</strong> Research, Oakland, CA, Catherine Schaefer,<br />

Epidemiologist/Senior Researcher, Kaiser Permanente<br />

Division <strong>of</strong> Research, Oakland, CA, Erica Gunderson,<br />

Epidemiologist/Senior Researcher, Kaiser Permanente<br />

Division <strong>of</strong> Research, Oakland, CA, Joe Selby, Director,<br />

Kaiser Permanente Division <strong>of</strong> Research, Oakland, CA, Lisa<br />

Croen, Epidemiologist/Senior Researcher, Kaiser<br />

Permanente Division <strong>of</strong> Research, Oakland, CA, Neil Risch,<br />

Adjunct Investigator, Kaiser Permanente Division <strong>of</strong><br />

Research, Oakland, CA<br />

Autoimmune disorders, collectively, include more than<br />

60 chronic and <strong>of</strong>ten disabling conditions that result from<br />

underlying defects in the immune system. As a group they<br />

affect approximately 15–20 million people in the United<br />

States, resulting in significant morbidity and mortality.<br />

Large, well-characterized, clinical populations in an environment<br />

that supports a broad research program are needed<br />

to advance our understanding <strong>of</strong> disease etiology. The<br />

Kaiser Permanente Medical Care Program (Northern California<br />

Region) or KPNC membership includes 3.3 million<br />

people, representing about 30% <strong>of</strong> the population <strong>of</strong><br />

northern California. The membership is sociodemographically<br />

and culturally diverse, while highly representative <strong>of</strong><br />

the general population. Comprehensive electronic medical<br />

records, including outpatient, pharmacy, laboratory, imaging<br />

and hospitalization data maintained since 1995 were<br />

used to construct a large registry <strong>of</strong> over 200,000<br />

individuals with diagnoses <strong>of</strong> one or more autoimmune<br />

conditions. Conditions for the study were derived from<br />

over 50 ICD-9 codes, broadly categorized into the following<br />

groups: endocrine, gastrointestinal, hematologic, kidney,<br />

liver, skin, neurologic/muscular, vascular, and connective<br />

tissue/joints. Planned additions to this registry include<br />

biospecimens and survey data on environmental and<br />

behavioral risk factors. This resource will be used to<br />

conduct the large epidemiologic investigations needed to<br />

advance our understanding <strong>of</strong> autoimmunity, including<br />

studies <strong>of</strong> genetic, social, and environmental contributions<br />

to risk, progression, and response to treatment. Results<br />

from our current investigations <strong>of</strong> autoimmune disease<br />

incidence/prevalence rates and patterns <strong>of</strong> co-morbidity<br />

will be presented, including results from our focused<br />

research efforts in multiple sclerosis and inflammatory<br />

bowel disease.<br />

doi:10.1016/j.clim.2007.03.539<br />

Sa.153 Functional Characterization <strong>of</strong> the<br />

Autoimmune-associated LYP-W620 Variant<br />

Lei Zhao, PhD Student, University <strong>of</strong> Southern California,<br />

Los Angeles, CA, Yingge Liu, Postdoctoral Fellow, University<br />

<strong>of</strong> Southern California, Los Angeles, CA, Edoardo Fiorillo,<br />

Postdoctoral Fellow, University <strong>of</strong> Southern California, Los<br />

Angeles, CA, Stephanie Stanford, PhD Student, University <strong>of</strong><br />

Southern California, Los Angeles, CA, Valeria Orru,<br />

Postdoctoral Fellow, University <strong>of</strong> Southern California, Los<br />

Angeles, CA, Nunzio Bottini, Assistant Pr<strong>of</strong>essor, University<br />

<strong>of</strong> Southern California, Los Angeles, CA<br />

A missense single-nucleotide polymorphism, C1858T in<br />

the PTPN22 gene is associated with multiple human<br />

autoimmune diseases, including type 1 diabetes, rheumatoid<br />

arthritis, systemic lupus erythematosus, Graves disease,<br />

juvenile idiopathic arthritis, generalized vitiligo, and<br />

others. Genetic studies have shown that the PTPN22<br />

C1858T polymorphism is primarily associated with autoimmunity<br />

in different populations. The PTPN22 gene<br />

encodes the lymphoid tyrosine phosphatase LYP, which is<br />

expressed only in white blood cells and acts as a<br />

gatekeeper <strong>of</strong> T lymphocyte activation. The molecular<br />

mechanism by which LYP tempers T lymphocyte activation<br />

involves the formation <strong>of</strong> a complex between LYP and the<br />

negative regulatory kinase Csk. When compared to the<br />

common LYP-R620 variant, the autoimmune-predisposing<br />

LYP-W620 variant shows reduced binding to Csk, and more<br />

recently we found that LYP-W620 is a gain-<strong>of</strong>-function form<br />

<strong>of</strong> the phosphatase. In order to analyze the molecular<br />

mechanism <strong>of</strong> the gain-<strong>of</strong>-function phenotype <strong>of</strong> LYP-W620,<br />

we isolated recombinant Csk-free LYP-R620 and W620 using<br />

an insect cell expression system. When we reconstituted in<br />

vitro the complex between LYP and Csk, we found that<br />

recombinant LYP-R620 binds Csk more efficiently than LYP-<br />

W620. We also analyzed the effect <strong>of</strong> Csk on the enzymatic<br />

activity <strong>of</strong> LYP. Our data obtained on in vitro reconstituted<br />

LYP–Csk complexes suggest that reduced binding to Csk<br />

cannot fully explain the gain-<strong>of</strong>-function phenotype <strong>of</strong> the<br />

LYP-W620 variant.<br />

doi:10.1016/j.clim.2007.03.540<br />

S127<br />

Sa.154 Spontaneous Myocarditis in Transgenic Mice<br />

Needs Appropriate MHC and Non-MHC Genes<br />

Veena Taneja, Assistant Pr<strong>of</strong>essor, Department <strong>of</strong><br />

<strong>Immunology</strong>, Mayo Clinic, Rochester, MN, Marshall Behrens,<br />

Senior Technician, Department <strong>of</strong> <strong>Immunology</strong>, Mayo Clinic,<br />

Rochester, MN, Leslie Cooper, Consultant, Mayo Clinic,<br />

Department <strong>of</strong> Cardiovascular Disease, Rochester, MN,<br />

Chella David, Pr<strong>of</strong>essor, Department <strong>of</strong> <strong>Immunology</strong>, Mayo<br />

Clinic, Rochester, MN<br />

Viral infection has been associated with the development<br />

<strong>of</strong> myocarditis in humans. Myocarditis is characterized by<br />

mononuclear infiltrate with attendant myocyte necrosis.<br />

Animal models <strong>of</strong> induced myocarditis have been generated<br />

by using coxsackie virus and immunization with cardiac<br />

myosin. We have recently generated a spontaneous model <strong>of</strong><br />

myocarditis. HLA-DQ8 and DR3 were expressed as transgenes<br />

in NOD mice lacking endogenous class II molecules, Abo.DQ8.<br />

NOD and Abo.DR3.NOD. Also mice expressing both transgenes<br />

in C57\B10 mice lacking endogenous class II molecules were<br />

generated. All mice were bred to congenic backgrounds. Only<br />

mice expressing HLA-DQ8 in NOD background developed<br />

spontaneous myocarditis. Abo.DR3.NOD, Abo.DQ8.B10 and<br />

Abo.DR3.B10 and transgene negative littermates did develop<br />

any gross or microscopic cardiac pathology. The autoimmunity<br />

was associated with the presence <strong>of</strong> spontaneous<br />

autoreactive T cells and antibodies to cardiac myosin. Only<br />

DQ8.NOD mice mounted a strong spontaneous in vitro T cell<br />

response and in vivo DTH response to cardiac myosin. These


S128 Abstracts<br />

data suggest that DQ8.NOD mice have lost tolerance to the<br />

self-cardiac myosin leading to spontaneous myocarditis and<br />

dilated cardiomyopathy. HLA-DQ8 is required for predisposition<br />

to the spontaneous autoreactivity while NOD background<br />

influences onset and progression <strong>of</strong> disease. This model <strong>of</strong><br />

myocarditis occurs predominantly in female mice and may<br />

provide insight into the pathogenesis <strong>of</strong> heart disease in<br />

women.<br />

doi:10.1016/j.clim.2007.03.554<br />

Sa.155 Human Lung Tumor Cells Modifies Rates <strong>of</strong><br />

CD4+CD25+ T Regulatory, CD19+CD23+ B Regulatory<br />

Cells, CD3+CD95+ Cells, NK Cells and B Lymphocytes<br />

Bayram Kiran, Tip Fakultesi, Temel Bilimler Binasi, Istanbul,<br />

Turkey, Akif Turna, Yedikule Hospital for Chest Diseases and<br />

Thoracic Surgery, Kadikoy, Istanbul, Turkey, Alper Yener,<br />

Istanbul Tip Fakultesi, Istanbul, Turkey, Atilla Gürses,<br />

Yedikule Gogus Hastaliklari Hastanesi, Istanbul, Turkey,<br />

Andac Salman, Istanbul Tip Fakultesi, Istanbul, Turkey,<br />

Selim Badur, Istanbul Tip Fakultesi, Temel Bilimler Binasi,<br />

Istanbul, Turkey<br />

The role <strong>of</strong> T regulatory cells and NK cells in human lung<br />

cancer has not yet been clarified. Our aim was to evaluate<br />

how the microenvironment <strong>of</strong> a tumor mass induced<br />

phenotypical changes in lymphocyte subsets. Our subjects<br />

were 10 patients with resectable non-small cell lung cancer.<br />

We evaluated 47 different phenotypically different lymphocyte<br />

subsets in blood samples taken from the pulmonary<br />

artery, pulmonary vein <strong>of</strong> a tumor bearing pulmonary lobe<br />

and peripheral blood during pulmonary resectional surgery.<br />

We showed that, tumor cells boosted CD25high+CD4high+T<br />

lymphocytes (Treg cells), B lymphocytes, NK and CD19+CD23<br />

+ cells (Breg cells) and CD3+CD95+ (FasL+T lymphocytes)<br />

(p=0.015, p=0.001, P=0.02, p=0.04, p=0.03 respectively).<br />

However, Mac-1 cells and HLADR+T lymphocytes were found<br />

to be decreased by tumor mass(p=0.04 and p=0.04), whereas<br />

only Breg , NK cell and B lymphocyte subsets were found to<br />

be expansed. In addition, the patients showed expansions <strong>of</strong><br />

CD45R0+ activated T lymphocytes and CD18+CD11b+(Mac-1)<br />

cell subsets without significant alteration by tumor microenvironment.<br />

In conclusion, subsets <strong>of</strong> Treg, Breg cells, FasL+<br />

T lymphocytes, B lymphocytes were modified by lung cancer<br />

microenvironment itself. This study also showed that,<br />

peripheral blood might not be good source for investigating<br />

the anti-tumor immunity since only Breg, NK cell and Treg<br />

cell subsets were found expansed peripherally. T lymphocytes<br />

and Mac-1 cells may be modified by systemic antitumor<br />

response rather than by local tumoral microenvironment.<br />

This study provides important insight into how tumor<br />

infiltrating lymphocytes and peripheral immune systems<br />

may be different in terms <strong>of</strong> lymphocyte subset expansion<br />

dynamics.<br />

doi:10.1016/j.clim.2007.03.552


<strong>Clinical</strong> <strong>Immunology</strong> (2007) 123, S129–S188<br />

ABSTRACTS<br />

<strong>Oral</strong> <strong>Presentations</strong>: Sunday, June 10<br />

#3201- From Genes to Treatment<br />

Sunday, June 10<br />

10:45 am−11:05 am<br />

Immune Reconstitution in X-Linked Hyper-IgM<br />

Syndrome With Recombinant CD40 Ligand<br />

Ashish Jain, Tenure- Track Investigator, Laboratory<br />

<strong>of</strong> Host Defenses, NIAID, NIH, Bethesda, MD, David<br />

Nelson, Senior Investigator, NCI, Bethesda, MD, Jospeh<br />

Kovacs, Senior Investigator, <strong>Clinical</strong> Center, Bethesda,<br />

MD, Shuying Liu, Study Coordinator, NIAID, Bethesda, MD,<br />

Thomas Fleisher, Senior Investigator, <strong>Clinical</strong> Center,<br />

Bethesda, MD, William Fanslow, Group Leader, Amgen<br />

Seattle, Seattle, WA, Hans Ochs, Pr<strong>of</strong>essor, Department<br />

<strong>of</strong> Pediatrics, Seattle, WA, Warren Strober, Senior<br />

Investigator, Laboratory <strong>of</strong> Host Defenses, NIAID, NIH,<br />

Bethesda, MD<br />

Purpose: X-linked hyper IgM syndrome (XHIM) is a<br />

combined immune deficiency disorder caused by mutations<br />

in the gene encoding CD40 ligand. The purpose <strong>of</strong><br />

this study was to investigate the safety and efficacy<br />

recombinant human CD40 ligand (rCD40L) for patients<br />

with XHIM. Methods: The study was designed as a phase I/<br />

II, open-label, single-dose study <strong>of</strong> rCD40L administered<br />

subcutaneously three times per week for 24 weeks in<br />

three children with XHIM. Adverse effects and efficacy <strong>of</strong><br />

rCD40L were evaluated during 6- month trial period.<br />

Results: The results <strong>of</strong> more than 230 subcutaneous<br />

injections showed that rCD40L was generally well tolerated.<br />

With rCD40L treatment, patients developed for the<br />

first time delayed type hypersensitivity reactions on skin<br />

testing that disappeared during the drug free follow up<br />

period. Analysis <strong>of</strong> patients’ T cells demonstrated first<br />

time capacity to synthesize IFN-g and TNF-a when<br />

stimulated with anti-CD3, or SEB, or SEA in vitro. Studies<br />

<strong>of</strong> cytokine production by intracellular staining demonstrate<br />

that rCD40L was able to prime both the CD4 and<br />

CD8 T cell populations with in vivo administration.<br />

Improvements <strong>of</strong> lymph node size and architecture were<br />

also noted; patients showed the development <strong>of</strong> primary<br />

follicles and follicular dendritic cells, as well as an<br />

doi:10.1016/j.clim.2007.03.006<br />

available at www.sciencedirect.com<br />

www.elsevier.com/locate/yclim<br />

expansion <strong>of</strong> B and T cell populations. Conclusions: This<br />

phase I/II first study <strong>of</strong> recombinant human CD40 ligand<br />

showed that rCD40L is capable <strong>of</strong> restoring immune<br />

functions in patients with XHIM. Further study will be<br />

needed to assess the overall potential <strong>of</strong> this therapy.<br />

doi:10.1016/j.clim.2007.03.007<br />

#3202- Antigen Specific Immune Modulation<br />

Sunday, June 10<br />

10:45 am−11:05 am<br />

Neur<strong>of</strong>ascin-specific Autoantibodies Mediate Axonal<br />

Injury in Inflammatory Demyelinating Diseases <strong>of</strong><br />

the Central Nervous System<br />

Christopher Linington, Pr<strong>of</strong>essor <strong>of</strong> Immunobiology,<br />

University <strong>of</strong> Aberdeen, Aberdeen, UK, Emily Mathey,<br />

Research Associate, University <strong>of</strong> Aberdeen, Aberdeen,<br />

UK, Tobias Derfuss, Max Planck Institute for<br />

Neurobiology, Martinsried, Germany, Matthew Rasband,<br />

Assistant Pr<strong>of</strong>essor, University <strong>of</strong> Connecticut Health<br />

Center, Farmington, CT, Maria Storch, Pr<strong>of</strong>essor, Medical<br />

University Graz, General Neurology, Graz, Germany,<br />

Reinhard Hohlfeld, Pr<strong>of</strong>essor, LMU University Hospital,<br />

<strong>Clinical</strong> Neuroimmunology, Munich, Germany, Edgar<br />

Meinl, Pr<strong>of</strong>essor, Max Planck Institute for Neurobiology,<br />

Martinsried, Germany<br />

We report that multiple sclerosis is associated with<br />

elevated autoantibody responses to neur<strong>of</strong>ascin, as<br />

compared to other inflammatory neurological diseases.<br />

The two is<strong>of</strong>orms <strong>of</strong> neur<strong>of</strong>ascin play essential roles in<br />

maintaining the molecular organization <strong>of</strong> myelinated<br />

fibers: NF186 is a neuronal protein located at the node<br />

<strong>of</strong> Ranvier and axonal initial segment where it interacts<br />

with voltage gated sodium channels, whilst NF155 is an<br />

oligodendroglial product sequestered at junctional complexes<br />

formed where paranodal loops <strong>of</strong> the myelin<br />

sheath contact the axonal surface. Analysis <strong>of</strong> neur<strong>of</strong>ascin-specific<br />

autoantibodies immunopurified from


S130 Abstracts<br />

seropositive donors demonstrates that they recognise the<br />

extracellular domain <strong>of</strong> both is<strong>of</strong>orms <strong>of</strong> the native<br />

protein indicating that this response may participate in<br />

disease pathogenesis. We investigated this hypothesis in<br />

experimental autoimmune encephalomyelitis using a pan<br />

NF155/186 mouse mAb to mimic the human autoantibody<br />

response. Passive transfer (i.p.) <strong>of</strong> mAb induced a rapid<br />

exacerbation <strong>of</strong> disease that was associated with a<br />

corresponding increase in acute axonal injury. Intriguing<br />

this occurred in the absence <strong>of</strong> any concomitant increase<br />

in the local inflammatory response or demyelination.<br />

Immun<strong>of</strong>luorescence microscopy revealed that binding <strong>of</strong><br />

the transferred neur<strong>of</strong>ascin-specific antibody was<br />

restricted nodes <strong>of</strong> Ranvier where it co-localised with<br />

voltage gated sodium channels. These results identify<br />

NF186 as a primary target for neur<strong>of</strong>ascin-specific autoantibodies<br />

and indicate antibody-dependent effector<br />

mechanisms are involved in the pathogenesis <strong>of</strong> axonal<br />

injury and loss in multiple sclerosis.<br />

doi:10.1016/j.clim.2007.03.008<br />

#3203- Innate <strong>Immunology</strong><br />

Sunday, June 10<br />

10:45 am−11:05 am<br />

IL-23 is Required for Induction <strong>of</strong> an Innate Immune<br />

Response to Citrobacter Rodentium<br />

Darrell O’Quinn, Postdoctoral Fellow, University <strong>of</strong><br />

Alabama, Department <strong>of</strong> Pathology, Birmingham, AL,<br />

Trenton Schoeb, Pr<strong>of</strong>essor, University <strong>of</strong> Alabama,<br />

Department <strong>of</strong> Genetics, Birmingham, AL, Casey Weaver,<br />

Pr<strong>of</strong>essor, University <strong>of</strong> Alabama, Department <strong>of</strong><br />

Pathology, Birmingham, AL<br />

We have previously shown that infection <strong>of</strong> IL-23deficient<br />

mice with the enteric murine pathogen Citrobacter<br />

rodentium results in complete mortality within 7-10<br />

days following inoculation despite the presence <strong>of</strong> CD4+ IL-<br />

17-competent cells. Using a mutant strain <strong>of</strong> bioluminescent<br />

C. rodentium, we here demonstrate that colonization<br />

<strong>of</strong> the cecum and mid- and distal colon <strong>of</strong> IL-23-deficient<br />

mice occurs earlier and in greater numbers than in IFNγ-,<br />

IFNγR-, IL-12-deficient, or wild-type controls. Histopathological<br />

examination <strong>of</strong> the lesions in IL-23-deficient mice<br />

confirms the presence <strong>of</strong> massive numbers <strong>of</strong> bacteria<br />

attached to intestinal epithelial cells <strong>of</strong> the mid- and distal<br />

colon, and multi-focal areas <strong>of</strong> ulcerative necrosis indicate<br />

a limited or absent inflammatory response. Analysis <strong>of</strong> C.<br />

rodentium colonization in IL-17R-/- mice reveal no significant<br />

differences from wild-type controls suggesting that<br />

IL-23, not IL-17, may be critical for the maintenance <strong>of</strong><br />

innate defenses in the distal colon.<br />

doi:10.1016/j.clim.2007.03.009<br />

OR.70 Intestinal Epithelial Cell Derived Thymic<br />

Stromal Lymphopoeitin Controls Dendritic<br />

Cell Function and T Regulatory Cell<br />

Homeostasis<br />

Iliyan D. Iliev, PhD Student, European Institute <strong>of</strong><br />

Oncology, Milan, Italy, Erika Mileti, Laboratory Assistant,<br />

European Institute <strong>of</strong> Oncology, Milan, Italy, Maria<br />

Rescig, Group Leader, European Institute <strong>of</strong> Oncology,<br />

Milan, Italy<br />

The mucosal immune system has developed mechanisms<br />

to tolerate beneficial micr<strong>of</strong>lora, but in the same time to<br />

initiate immunity against invading pathogens. How these<br />

mechanisms function is still not completely understood. In<br />

our current study we investigated the possible role <strong>of</strong><br />

Epithelial Cells (EC) as a “tolerance keepers” in the gut<br />

driving the development <strong>of</strong> non-inflammatory Dendritic<br />

Cells (DC) able to promote CD4+CD25+Foxp3+ T regulatory<br />

cells (Treg) development. We developed a co-culture<br />

system, in which EC (Caco2 or primary intestinal EC)<br />

supernatants were used to condition monocyte-derived DC.<br />

We found that in both, human and mice, EC-conditioned DC<br />

were producing less inflammatory cytokines in response to<br />

bacterial stimuli and favored the development <strong>of</strong> functionally<br />

active Treg. These tolerogenic DC were involved in<br />

driving Treg also in vivo in mice. Thymic stromal<br />

lymphopoeitin (TSLP) produced by EC, together with<br />

additional factors, was involved in this process since DC<br />

conditioned with supernatants from Caco2 cell line<br />

“silenced” for TSLP, were unable to expand Treg. Finally,<br />

mice immunized with Salmonella loaded EC-conditioned DC<br />

were less resistant to lethal Salmonella dose than control<br />

mice, confirming their tolerogenic potential in vivo.<br />

Therefore, intestinal EC released TSLP, can influence gut<br />

DC function and can control the homeostasis <strong>of</strong> peripheral<br />

Treg. We propose that failure in this mechanism can<br />

participate in the pathology <strong>of</strong> Inflammatory Bowel<br />

Disease.<br />

doi:10.1016/j.clim.2007.03.010<br />

Regulatory T Cells II<br />

Sunday, June 10<br />

2:45 pm−4:45 pm<br />

OR.71 Critical Role <strong>of</strong> IFNg in Allograft Tolerance<br />

Mediated by Foxp3+CD4+CD25+ Regulatory T Cells<br />

and the Production <strong>of</strong> Indoleamine 2, 3-dioxygenase<br />

by Graft Endothelial Cells<br />

Pamela Thebault, PhD, INSERM U643, Nantes, France,<br />

Michele Heslan, IE, INSERM U643, Nantes, France, Thomas<br />

Condamine, PhD, INSERM U643, Nantes, France, Abdelhadi<br />

Saoudi, CR1, INSERM U563, Toulouse, France, Marcello Hill,<br />

Postdoctoral Fellow, INSERM U643, Nantes, France, Regis<br />

Josien, CR1, INSERM U643, Nantes, France, Ignacio Anegon,<br />

DR2, INSERM U643, Nantes, France, Maria-Cristina Cuturi,<br />

DR2, INSERM U643, Nantes, France, Elise Chiffoleau, CR 2,<br />

INSERM U643, Nantes, France


Abstracts<br />

Functionally specialized subsets <strong>of</strong> regulatory CD4+T cells<br />

have been shown to play an important role in the regulation <strong>of</strong><br />

both T cell-mediated and innate immune responses. In particular,<br />

the CD4+CD25+ subpopulation <strong>of</strong> T cells has been<br />

shown to be crucial in self-tolerance and also to prevent<br />

allograft rejection. As CD25 is not uniquely expressed by<br />

regulatory T cells, but can also be expressed by activated<br />

effector cells, the identification <strong>of</strong> Foxp3 has provided an<br />

opportunity to specifically identify regulatory T cells and to<br />

track them in vivo so as to better define the mechanisms<br />

involved in their regulation <strong>of</strong> immune responses. We previously<br />

demonstrated, in a fully MHC mismatched rat cardiac<br />

allograft combination, that a short-term treatment with a<br />

deoxyspergualine analogue, LF15-0195, induces long-term<br />

allograft tolerance with a specific expansion <strong>of</strong> regulatory<br />

CD4+CD25+T cells that accumulate within the graft. In this<br />

study, we show that following transfer to a secondary irradiated<br />

recipient, regulatory CD4+CD25+T cells are able to<br />

rapidly home to the graft, induce accumulation <strong>of</strong> Foxp3+ and<br />

Tr1/IL10+ regulatory CD4+Tcells and induce graft endothelial<br />

cell expression <strong>of</strong> Indoleamine 2, 3-dioxygenase (IDO),<br />

inducible Nitric Oxide synthase (iNOS) and Heme-Oxygenase-1<br />

(HO-1). Moreover, in vivo transfer <strong>of</strong> tolerance can be<br />

abrogated by blocking IFNg or IDO and in vitro, anti-IFNg<br />

reduces the survival/function <strong>of</strong> alloantigen-induced Foxp3+<br />

CD4+ regulatory T cells. Together, our results demonstrate<br />

interrelated mechanisms between regulatory CD4+Tcells and<br />

the graft endothelial cells in this local immune privilege and a<br />

key role for IFNg and IDO in this process.<br />

doi:10.1016/j.clim.2007.03.011<br />

OR.72 Amino-Terminal Region <strong>of</strong> Foxp3 Performs<br />

Multiple Functions in Regulating Transcription<br />

Jared E. Lopes, Graduate Students, University <strong>of</strong><br />

Washington, Seattle, WA, Jianguang Du, Postdoctoral Fellow,<br />

Benaroya Research Institute, Seattle, WA, Xiaocui Sun,<br />

Research Technician, Benaroya Research Institute, Seattle,<br />

WA, Mark Chong, Postdoctoral Fellow, Molecular<br />

Pathogenesis Program, Skirball Institute <strong>of</strong> Biomolecular<br />

Medicine, New York, NY, Liang Zhou, Fellow, Molecular<br />

Pathogenesis Program, Skirball Institute <strong>of</strong> Biomolecular<br />

Medicine, New York, NY, Dan R. Littman, Director <strong>of</strong><br />

Molecular Pathogenesis Program, Skirball Institute <strong>of</strong><br />

Biomolecular Medicine and Howard Hughes Medical<br />

Institute, New York, NY, Steven F. Ziegler, <strong>Immunology</strong><br />

Program Director, Benaroya Research Institute, <strong>Immunology</strong><br />

Program, Seattle, WA<br />

Foxp3 has been shown to be both necessary and sufficient<br />

for the development and function <strong>of</strong> naturally-arising CD4+<br />

CD25+ regulatory Tcells (Tregs) in mice. Mutation <strong>of</strong> Foxp3 in<br />

scurfy mice and Foxp3 in humans with IPEX results in fatal,<br />

early onset autoimmune disease and demonstrates the<br />

critical role <strong>of</strong> Foxp3 in maintaining immune homeostasis.<br />

The Foxp3 protein encodes several functional domains<br />

including a C2H2 zinc finger, a leucine zipper, and a wingedhelix/forkhead<br />

(FKH) domain. We have previously shown that<br />

Foxp3 functions as a transcriptional repressor and inhibits<br />

activation-induced IL-2 gene transcription. We recently<br />

identified a novel functional domain within the aminoterminal<br />

half <strong>of</strong> Foxp3, which is required for Foxp3-mediated<br />

repression <strong>of</strong> transcription from both a constitutively active<br />

and an NFAT-inducible promoter. In order to determine the<br />

mechanism by which Foxp3 regulates transcription, we<br />

performed a yeast-2-hybrid analysis searching for proteins<br />

that interact with the amino-terminal region <strong>of</strong> Foxp3. One<br />

target was RORgammat (RORgt), which is required for the<br />

differentiation <strong>of</strong> naïve CD4+ T cells into pro-inflammatory,<br />

IL-17-producing T cells (Th17). Interestingly, CD4+ T cells<br />

express Foxp3 and RORgt in response to TCR stimulation in the<br />

presence <strong>of</strong> TGF-&#946; and differentiate into either Tregs or<br />

Th17 cells, depending on the presence <strong>of</strong> IL-6. We show that<br />

Foxp3 directly inhibits RORgt-mediated transcription. Future<br />

studies will address the mechanism by which Foxp3 inhibits<br />

RORgt function and how this process is regulated by IL-6.<br />

doi:10.1016/j.clim.2007.03.012<br />

S131<br />

OR.73 Phenotypic and Functional Characteristics <strong>of</strong><br />

Different Regulatory T Cell Subsets (Tregs) in<br />

Patients with Cancer<br />

Christoph Bergmann, Postdoctoral Fellow, University <strong>of</strong><br />

Pittsburgh Cancer Institute, Department <strong>of</strong> Pathology,<br />

Pittsburgh, PA, Laura Strauss, Postdoctoral Fellow,<br />

University <strong>of</strong> Pittsburgh Cancer Institute, Department <strong>of</strong><br />

Pathology, Pittsburgh, PA, Jonas Johnson, Pr<strong>of</strong>essor and<br />

Chair, University <strong>of</strong> Pittsburgh Cancer Institute, Department<br />

<strong>of</strong> Otolaryngology, Pittsburgh, PA, Theresa L. Whiteside,<br />

Director, University <strong>of</strong> Pittsburgh Cancer Institute,<br />

Department <strong>of</strong> Pathology, Pittsburgh, PA<br />

Immune suppression mediated by Tregs is a feature <strong>of</strong><br />

cancer. To compare phenotype and function <strong>of</strong> Tregs present<br />

in the tumor (TIL) and blood (PBMC) <strong>of</strong> patients with head and<br />

neck cancer (HNC), we obtained samples from 40 patients and<br />

15 normal controls (NC). The phenotype and frequency <strong>of</strong><br />

CD4 + CD25 high and CD4 + CD25 − T cell subsets were evaluated.<br />

These cells were separated by single-cell sorting, tested for<br />

suppressor function and also cultured +/− cytokines and/or<br />

rapamycin (1 nM). IL-10 or TGF- 2 1 mAbs were used to block<br />

suppression <strong>of</strong> responder cell (R) proliferation. TIL were<br />

enriched in CD4 + CD25 high (13%±3) cells (Foxp3 + GITR + IL-<br />

10 + TGF− 2 1 + ). In PBMC, Tregs (CD25 high Foxp3 + CD62L + CCR7 + GI-<br />

TR + IL-10 + TGF− 2 1 + ) represented 5%±3 among CD4 + cells. In<br />

contrast, PBMC in NC contained fewer (pb0.001) Tregs (2%±<br />

1.5). In PBMC <strong>of</strong> patients and NC, CD4 + CD25 - IL-10 + TGF- 2 1 +<br />

(Tr1) cells were detected. In TIL, Tr1 cells represented 23%±3<br />

<strong>of</strong> CD4 + cells. Tr1 cells expanded from PBMC and TIL were<br />

CD25 − IL-10 + IL2R 3+ TGF- 2 1 + IL-4 − with low levels <strong>of</strong> Foxp3 and<br />

CTLA-4 expression. TIL Tregs mediated stronger suppression<br />

(p b0.001) than PBMC Tregs. NC Tregs mediated weak<br />

suppression. Neutralizing mAbs to IL-10 or TGF- 2 1 abolished<br />

suppression <strong>of</strong> R proliferation by Tregs. Cell contact <strong>of</strong> Tregs,<br />

but not Tr1, with R enhanced suppression. Rapamycin<br />

promoted selective expansion <strong>of</strong> Tregs and Tr1 cells by<br />

eliminating non-Tregs in patients and NC. Characteristics <strong>of</strong><br />

rapamycin-expanded Tregs and Tregs in freshly-isolated<br />

lymphocytes were similar. Tregs mediating suppression in<br />

the tumor and circulation <strong>of</strong> HNC patients are heterogeneous


S132 Abstracts<br />

populations distinct from Tregs in NC. To mediate immune<br />

escape, tumor activates and/or induces unique Treg subsets.<br />

doi:10.1016/j.clim.2007.03.013<br />

OR.74 Hepatic Stellate Cells Expand<br />

CD4 + CD25 + Foxp3 + Tregs and Prevent Rejection <strong>of</strong><br />

Allogeneic Islet Transplants<br />

Shiguang Qian, Associate Pr<strong>of</strong>essor, Cleveland Clinic,<br />

Cleveland, OH, Guoping Jiang, Research Fellow,<br />

Department <strong>of</strong> <strong>Immunology</strong>, Cleveland Clinic, Cleveland,<br />

OH, Zhenyu Yin, Research Fellow, Department <strong>of</strong><br />

<strong>Immunology</strong>, Cleveland Clinic, Cleveland, OH, John J. Fung,<br />

Pr<strong>of</strong>essor, Department <strong>of</strong> General Surgery, Cleveland, OH,<br />

Liang-Mou Kuo, Research Fellow, Department <strong>of</strong><br />

<strong>Immunology</strong>, Cleveland Clinic, Cleveland, OH, Lina Lu,<br />

Associate Pr<strong>of</strong>essor, Department <strong>of</strong> <strong>Immunology</strong>, General<br />

Surgery, Cleveland Clinic, Cleveland, OH<br />

Liver tolerance was demonstrated by spontaneous acceptance<br />

<strong>of</strong> liver allografts in many species, but hepatocyte<br />

transplants were acutely rejected, suggesting an immunosuppressive<br />

effect <strong>of</strong> liver tissue cells. In this study, we<br />

demonstrated immunosuppressive activity <strong>of</strong> hepatic stellate<br />

cells (HSC) through expansion <strong>of</strong> CD4 + CD25 + Foxp3 +<br />

Tregs. HSC isolated from B6 mice (H2 b ) constitutively expressed<br />

low MHC class I and CD80. Expression <strong>of</strong> MHC class I, II<br />

and B7-H1 was markedly upregulated in IFN-γ activated-HSC<br />

(aHSC), which rendered them to stimulate allogeneic T cell<br />

proliferation in both CD4 + and CD8 + cells (CFSE). aHSC<br />

cultured with allogenetic CD4 + T cells at 1:10 ratio for 5 days<br />

markedly expanded CD4 + CD25+ + Foxp3 + cells (from 2.5% to<br />

10.5%), which were obviously from naïve CD4 + CD25 + population,<br />

since depletion <strong>of</strong> CD4 + CD25 + , only small number <strong>of</strong><br />

CD4 + CD25 + cells were expanded, and were all Foxp3 − .<br />

Addition <strong>of</strong> aHSC expanded CD4 + CD25 + cells into CD4 + cells<br />

either stimulated by anti-CD3 or allo-antigens, resulted in<br />

marked inhibition <strong>of</strong> T cell proliferation in a dose dependent<br />

manner. To explore clinical application, 300 BALB/c (H2 d )<br />

islets mixed with aHSC (3×10 5 ) transplanted under renal<br />

capsule <strong>of</strong> chemically diabetic B6 recipients, achieving<br />

markedly prolonged islet graft survival [median survival<br />

time (MST) from 13 days to N60 days (pb0.01)] with 55%<br />

being euglycemia for N60d. None <strong>of</strong> islet-only recipients<br />

remained euglycemia for N17d. Removal <strong>of</strong> islet-grafted<br />

kidney resulted in quick glycemia. Prolongation <strong>of</strong> islet<br />

allografts by co-transplanted HSC was associated with<br />

upregulated apoptotic activity (TUNEL) and less T cell<br />

infiltration (CD3), indicating potential application in improving<br />

islet transplantation.<br />

doi:10.1016/j.clim.2007.03.014<br />

OR.75 Polyclonal CD4+CD25+ Regulatory T Cells<br />

Induced With TGF-β Prevent a Stimulatory<br />

Graft-Versus-Host Disease With a Lupus-Like Syndrome<br />

Song Guo Zheng, Assistant Pr<strong>of</strong>essor <strong>of</strong> Research Medicine,<br />

University <strong>of</strong> Southern California Keck School <strong>of</strong> Medicine, Los<br />

Angeles, CA, Julie Wang, Research Lab Specialist, University <strong>of</strong><br />

Southern California Keck School <strong>of</strong> Medicine, Los Angeles, CA,<br />

Shuang Ye, Postdoctoraltoral Fellow, University <strong>of</strong> Southern<br />

California Keck School <strong>of</strong> Medicine, Los Angeles, CA, David A.<br />

Horwitz, Pr<strong>of</strong>essor, University <strong>of</strong> Southern California Keck<br />

School <strong>of</strong> Medicine, Los Angeles, CA<br />

We have previously reported that the adoptive transfer <strong>of</strong><br />

antigen-specific Tregs generated by allogeneic antigen stimulation<br />

with TGF-β prevents the appearance <strong>of</strong> lupus syndrome<br />

induced by injection <strong>of</strong> DBA/2 mouse spleen cells into (DBA/<br />

2×C57BL/6) F1 mice. Here we have considered whether<br />

polyclonal CD4+ Tregs induced ex vivo may have similar<br />

suppressive effects in this model. Naïve CD4+CD25- cells<br />

isolated from DBA/2 mice were stimulated with anti-CD3/<br />

CD28 coated beads+TGF-β without APC. TGF-β was able to<br />

directly induce the majority <strong>of</strong> activated CD25+ cells to express<br />

Foxp3, develop cell markers characteristic <strong>of</strong> Tregs, and acquire<br />

suppressive activity in vitro. The remaining CD25- cells did not<br />

express Foxp3 or develop suppressive activity. TGF-β -induced<br />

CD4+ Tregs not only suppressed the Tcell proliferative response<br />

to anti-CD3, but also inhibited the T cell response to allogeneic<br />

cells. Substituting soluble anti-CD3 and antigen-presenting cells<br />

(APCs) for anti-CD3/28 beads resulted in decreased Foxp3+<br />

expression, an effect that was reversed by the addition <strong>of</strong> anti-<br />

IL-6. These levels <strong>of</strong> IL-6 in cultures containing TGF-β did not<br />

result in the production <strong>of</strong> IL-17. Finally, (D2xB6)F1 mice<br />

injected with D2 cells and 5 million TGF-β -induced CD4+ CD25+<br />

Tregs did not develop increased polyclonal IgG, anti-dsDNA<br />

autoantibodies, or nephritis. Since SLE is a generalized<br />

autoimmune disease with many autoantibodies, and production<br />

<strong>of</strong> IL-2 and TGF-β is decreased in this disease, the<br />

generation <strong>of</strong> large numbers <strong>of</strong> autologous, polyclonal CD4+<br />

CD25+Foxp3+ Treg cells with these cytokines ex vivo may have<br />

considerable therapeutic potential.<br />

doi:10.1016/j.clim.2007.03.015<br />

OR.76 Low Dose Antigen Promotes Induction <strong>of</strong><br />

Both Foreign and Self Reactive Human<br />

CD25+Foxp3+ Regulatory T Cells from<br />

CD4+CD25-Foxp3- Peripheral T Cells<br />

S. Alice Long, Benaroya Research Institute, Seattle, WA,<br />

Mindi Walker, Scientist, Therakos, Exton, PA, Isabelle<br />

Mueller, Graduate Student, Benaroya Research Institute,<br />

Seattle, WA, Jane Buckner, Pr<strong>of</strong>essor, Benaroya Research<br />

Institute, Seattle, WA, Mary Rieck, RA II, Benaroya Research<br />

Institute, Seattle, WA<br />

Upon activation, a subpopulation <strong>of</strong> human CD4+CD25-<br />

Foxp3- T cells upregulate Foxp3 and become regulatory. These<br />

cells are referred to as induced TR (iTR) and have similar<br />

properties to TR isolated directly from peripheral blood. We<br />

focus here on properties <strong>of</strong> stimulation that influence generation<br />

<strong>of</strong> iTR and characteristics <strong>of</strong> the CD4+ CD25- T cells from<br />

which they are derived. We consistently observe that stimulation<br />

<strong>of</strong> CD4+CD25- T cells with foreign antigen results in two<br />

populations <strong>of</strong> antigen specific tetramer positive (Tmr+) Tcells;<br />

Tmr+Foxp3+ and Tmr− Foxp3− cells. The Foxp3+ population can<br />

arise from cells which express low or high levels <strong>of</strong> CD127, with<br />

the greatest number <strong>of</strong> which originate from CD127mid/hi


Abstracts<br />

populations. We demonstrate similar results with islet antigens<br />

in frequency, source and potency. Generation <strong>of</strong> iTR is enhanced<br />

in the presence <strong>of</strong> low dose antigen relative to high dose. Thus,<br />

low level TCR stimulation results in enhanced frequency <strong>of</strong> Tmr+<br />

CD25+Foxp3+ T cells. These results demonstrate that iTR are<br />

generated as a consequence <strong>of</strong> normal immune responses in<br />

settings where antigen is in limited quantities, which may, in<br />

part, explain the phenomenon <strong>of</strong> low dose tolerance. Studies<br />

with T1D samples demonstrate the induction <strong>of</strong> islet and<br />

foreign iTR with similar functional potency. However, dose <strong>of</strong><br />

antigen has no effect on the induction <strong>of</strong> iTR in T1D. Thus, T1D<br />

subjects have the ability to generate islet specific iTR in the<br />

periphery, but defects in the response to activation or the<br />

frequency <strong>of</strong> iTR, may contribute to disease progression.<br />

doi:10.1016/j.clim.2007.03.016<br />

OR.77 IL-10 Both Inhibits Treg Suppressive Activity<br />

and is a Survival Factor for Human Natural Tregs<br />

Clare Baecher-Allan, Instructor <strong>of</strong> Neurology, Harvard<br />

Medical School, Brigham and Women’s Hospital,<br />

Department <strong>of</strong> Neurology, Boston, MA, Charles Ashley,<br />

Research Technician, Harvard Medical School, Brigham and<br />

Women's Hospital, Department <strong>of</strong> Neurology, Boston, MA,<br />

David Hafler, Breakstone Chair <strong>of</strong> Neurology, Harvard<br />

Medical School, Brigham and Women’s Hospital,<br />

Department <strong>of</strong> Neurology, Boston, MA<br />

Human natural Tregs (CD4+CD25high) are comprised <strong>of</strong><br />

two functionally distinct populations that differentially<br />

express HLA Class II proteins directly ex vivo. The MHC<br />

class II+ Tregs, referred to as ‘DR+Tregs’, are a highly suppressive<br />

population that rapidly inhibit the activation <strong>of</strong> cocultured<br />

responder T cells. The DRnegTregs, on the other<br />

hand, exhibit a delayed effector function as they allow an<br />

initial period <strong>of</strong> responder T cell proliferation before they<br />

shut down the response. While the DR+Tregs suppress solely<br />

by cell contact and do not produce IL-10, IL-10 is <strong>of</strong>ten (but<br />

not always) produced by the responder T cells and by the<br />

DR-Tregs in these accessory-cell-free assays. Interestingly,<br />

in those experiments in which the responder T cells did<br />

produce IL-10, the DR+Treg co-cultures exhibit less suppression<br />

than the duplicate co-cultures that had been given<br />

neutralizing IL-10 antibody from the outset. Thus, DR+Treg<br />

rapid suppression proceeds unimpeded in the absence <strong>of</strong> IL-<br />

10, while the presence <strong>of</strong> IL-10 inhibits their function. In<br />

order to understand this effect <strong>of</strong> IL-10, the two different<br />

cellular components <strong>of</strong> the co-culture were assayed to<br />

determine if the presence <strong>of</strong> IL-10 modulated the expression<br />

<strong>of</strong> regulatory (Foxp3, IL-10, TGFb), or apoptosis (BAX,<br />

BCL-2, and BCL-XL) related products. The results indicate<br />

that blocking IL-10 in DR+Treg co-cultures increases the<br />

frequency <strong>of</strong> AnnexinV+ responder T cells, reflecting the<br />

increase in suppression. Within the DR+Treg, however,<br />

blocking IL-10 triggered a reproducible decrease in BCL-XL<br />

and an increase in BAX, indicating increased Treg<br />

sensitivity to apoptosis that may affect long-term maintenance<br />

<strong>of</strong> suppression.<br />

doi:10.1016/j.clim.2007.03.017<br />

OR.78 High Density SNP Analysis <strong>of</strong> the MHC Region<br />

Reveals Multiple Loci for Type 1A Diabetes<br />

Theresa A. Aly, Graduate Student, University <strong>of</strong> Colorado<br />

Health Sciences Center, Barbara Davis Center, Aurora, CO,<br />

Erin E. Baschal, Graduate Student, University <strong>of</strong> Colorado<br />

Health Sciences Center, Barbara Davis Center, Aurora, CO,<br />

Mohamed M. Jahromi, Fellow, University <strong>of</strong> Colorado Health<br />

Sciences Center, Barbara Davis Center for Childhood<br />

Diabetes, Aurora, CO, Adam Kretowski, Physician,<br />

University <strong>of</strong> Colorado Health Sciences Center, Barbara<br />

Davis Center for Childhood Diabetes, Aurora, CO, Sunanda<br />

R. Babu, Research Associate, University <strong>of</strong> Colorado Health<br />

Sciences Center, Barbara Davis Center for Childhood<br />

Diabetes, Aurora, CO, Marian J. Rewers, Physician,<br />

University <strong>of</strong> Colorado Health Sciences Center, Barbara<br />

Davis Center for Childhood Diabetes, Aurora, CO, George S.<br />

Eisenbarth, Physician-Scientist, University <strong>of</strong> Colorado<br />

Health Sciences Center, Barbara Davis Center, Aurora, CO<br />

Haplotype sharing studies combined with HLA genotyping<br />

demonstrate that loci linked to the MHC region in addition to<br />

HLA-DR/DQ loci contribute major risk for type 1A diabetes. The<br />

hunt for these additional major loci is underway. In our survey<br />

study, we analyzed 656 single nucleotide polymorphisms (SNPs)<br />

in the MHC region and compared their allele frequencies in case<br />

versus control chromosomes from 45 families participating in<br />

the Diabetes Autoimmunity Study <strong>of</strong> the Young (DAISY). We<br />

included significant SNPs (pb0.01) from our survey study and<br />

additional SNPs extending 6 Mb further telomeric <strong>of</strong> the MHC in<br />

a follow-up study <strong>of</strong> 342 additional DAISY, HBDI, and Polish<br />

families. The most significant SNPs with distinct diabetesassociated<br />

peaks centromeric or within the MHC were at the<br />

ITPR3 (inositol-triphosphate receptor 3), HLA-DPB1, -DQB1, -<br />

DRB1, -DRA, and MICA loci. In addition to confirming these<br />

associations, our follow-up studies revealed a strong association<br />

<strong>of</strong> a 4 SNP DRA haplotype in 422/461 case chromosomes (92%)<br />

versus 437/579 control chromosomes (75%, p=1.8×10–11). The<br />

DRA haplotype remained significant when specific high-risk<br />

(DR3, DR4) and protective (DR2) chromosomes were excluded<br />

(p=0.05). The most significant regions telomeric <strong>of</strong> the MHC<br />

were at the Mas1L/UBD (Mas-related G protein coupled<br />

receptor/diubiquitin, p=0.00001) and PRSS16 loci (thymusspecific<br />

serine protease, p=0.00005). An HLA-DRA haplotype is<br />

strongly associated with type 1A diabetes; however, the<br />

biological basis for this association may be due to linked<br />

genes (e.g. HLA-DRB1-DQB1). Two novel regions 3100 kb and<br />

5400 kb telomeric <strong>of</strong> the HLA-DR and -DQ loci are also<br />

associated with diabetes.<br />

doi:10.1016/j.clim.2007.03.018<br />

Genetics<br />

Sunday, June 10<br />

2:45 pm−4:45 pm<br />

OR.79 Uniquely Designed Candidate Gene<br />

Identification Platform Discovers Novels Genes in<br />

Childhood Onset SLE<br />

Don Armstrong, FOCIS Center, University <strong>of</strong> Southern<br />

California School <strong>of</strong> Medicine, Los Angeles, CA, Andreas<br />

S133


S134 Abstracts<br />

Reiff, Children’s Hospital <strong>of</strong> Los Angeles and University <strong>of</strong><br />

Southern California School <strong>of</strong> Medicine, Los Angeles, CA,<br />

Chaim Jacob, FOCIS Center, University <strong>of</strong> Southern<br />

California School <strong>of</strong> Medicine, Los Angeles, CA, Raphael<br />

Zidovetzki, FOCIS Center, University <strong>of</strong> Southern California<br />

School <strong>of</strong> Medicine, Los Angeles, CA<br />

We present a novel methodology to maximize the ability <strong>of</strong><br />

microarray based single nucleotide polymorphism (SNP) typing<br />

platforms to discover genes which are associated with complex<br />

polygenic diseases when complete genomic coverage is infeasible.<br />

This methodology was used to identify genetic variants<br />

which are associated with childhood onset SLE. A platform <strong>of</strong><br />

9412 SNPs corresponding to 1204 genes was selected using<br />

bioinformatics and expert knowledge and validated. Molecular<br />

inversion probes and high throughput techniques were used to<br />

type SNP alleles. A cohort <strong>of</strong> 753 subjects, corresponding to 251<br />

full trios (both parents and affected SLE child) were genotyped.<br />

Utilizing the Transmission Disequilibrium Test (TDT) and appropriate<br />

multiple comparison corrections, a panel <strong>of</strong> several novel<br />

childhood onset SLE genes were determined to be significant.<br />

Among these, the N673S polymorphism in SELP (P-Selectin) and<br />

the C203S polymorphism in IRAK1 (Interleukin 1 receptorassociated<br />

kinase 1) were found with a high degree <strong>of</strong><br />

significance. These results suggest new directions in which to<br />

expand the understanding <strong>of</strong> the pathogenesis <strong>of</strong> SLE. The<br />

results <strong>of</strong> this study demonstrate that the novel methodology<br />

provides a powerful approach which is generally applicable to<br />

the identification <strong>of</strong> the genetic foundations <strong>of</strong> complex<br />

diseases.<br />

doi:10.1016/j.clim.2007.03.019<br />

OR.80 The Molecular Basis by Which Polymorphism<br />

in the OX40L Gene Predisposes to SLE<br />

Harinder Manku, Research Fellow, Imperial College London,<br />

Molecular Genetics and Rheumatology, London, England,<br />

Deborah Cunninghame Graham, Research Fellow, Imperial<br />

College London, Molecular Genetics and Rheumatology,<br />

London, England, Robert R. Graham, Research Fellow, Broad<br />

Institute <strong>of</strong> Harvard and MIT, Cambridge, MA, Timothy W.<br />

Behrens, Genentech Inc., San Francisco, CA, Timothy W.<br />

Behrens, Adjunct Pr<strong>of</strong>essor, University <strong>of</strong> Minnesota,<br />

Department <strong>of</strong> Medicine, Minneapolis, MN, David<br />

Althshuler, Associate Pr<strong>of</strong>essor, Broad Institute <strong>of</strong> Harvard<br />

and MIT, Cambridge, MA, Timothy J. Vyse, Reader and<br />

Honorary Consultant, Imperial College London, Department<br />

<strong>of</strong> Molecular Genetics and Rheumatology, London, England<br />

SLE is a complex polygenic disease where immune dysregulation<br />

results in chronic activation <strong>of</strong> B lymphocytes and<br />

autoantibody production against a variety <strong>of</strong> nuclear antigens.<br />

OX40L, a type II membrane glycoprotein found on the surface <strong>of</strong><br />

activated B lymphocytes, interacts with OX40 on T lymphocytes<br />

to amplify T-dependent B cell proliferation and differentiation.<br />

A family-based association analysis using a set <strong>of</strong> 45 SNP<br />

markers across OX40L was conducted in 472 UK and 429 US SLE<br />

parent-<strong>of</strong>fspring trios. We identified an associated promoter<br />

haplotype block (32 kb) that was preferentially transmitted to<br />

affected <strong>of</strong>fspring (P=0.003 UK, P=0.0005 US, P=6.7×10 −6<br />

combined). A tagging SNP (rs2205960) from this haplotype<br />

showed significant association with SLE in an independent set <strong>of</strong><br />

310UKcasesand642controls(P= 1.3×10 −5 OR=1.55, 95%<br />

CI=1.27–1.89). In order to investigate the molecular basis <strong>of</strong> the<br />

genetic association, we studied OX40L expression in individuals<br />

selected on the basis <strong>of</strong> allelic composition at the OX40L<br />

promoter. The promoter polymorphisms associated with SLE<br />

correlated with increased expression levels <strong>of</strong> OX40L. This was<br />

shown by three-colour flow cytometry <strong>of</strong> 16 EBV-transformed<br />

cell lines: those homozygous for the over-transmitted haplotype<br />

exhibited increased expression relative to the under-transmitted<br />

homozygote. We further show that the disease susceptibility<br />

alleles in the OX40L promoter were associated with an<br />

eight-fold increase in RNA expression (P=0.008). The mechanism<br />

by which increased OX40L expression increases the<br />

susceptibility to SLE remains to be established, but is the<br />

subject <strong>of</strong> ongoing investigation.<br />

doi:10.1016/j.clim.2007.03.020<br />

OR.81 Genetic Association <strong>of</strong> IL-21 Polymorphisms<br />

with Systemic Lupus Erythematosus<br />

Amr H. Sawalha, Assistant Pr<strong>of</strong>essor, Department <strong>of</strong><br />

Medicine, Oklahoma University Health Sciences Center, VA<br />

Medical Center, Oklahoma Medical Research Foundation,<br />

Oklahoma City, OK, Kenneth Kaufman, Senior Research<br />

Scientist, Oklahoma Medical Research Foundation,<br />

Oklahoma City, OK, Jennifer Kelly, Senior Research<br />

Assistant, Oklahoma Medical Research Foundation,<br />

Oklahoma City, OK, Teresa Aberle, PA-C, Oklahoma Medical<br />

Research Foundation, Oklahoma City, OK, Adam Adler,<br />

Research Associate, Oklahoma Medical Research<br />

Foundation, Oklahoma City, OK, Jeff Kilpatrick, Computer<br />

Programmer/Data Analyst, Oklahoma Medical Research<br />

Foundation, Oklahoma City, OK, Edward Wakeland,<br />

Pr<strong>of</strong>essor, UT Southwestern, Center <strong>of</strong> <strong>Immunology</strong>, Dallas,<br />

TX, Quan-Zhen Li, Assistant Pr<strong>of</strong>essor, Center for<br />

<strong>Immunology</strong>, University <strong>of</strong> Texas Southwestern Medical<br />

Center, Dallas, TX, Amy Wandstrat, Instructor, UT<br />

Southwestern, Rheumatology, Dallas, TX, Judith James,<br />

Pr<strong>of</strong>essor/Member, Oklahoma Medical Research<br />

Foundation/ Arthritis and <strong>Immunology</strong>, Oklahoma City, OK,<br />

Joan Merrill, Member, Oklahoma Medical Research<br />

Foundation, <strong>Clinical</strong> Pharmacology, Oklahoma City, OK,<br />

Peter Lipsky, Chief, NIAMS, Bethesda, MD, John B. Harley,<br />

Pr<strong>of</strong>essor and Department Head, Oklahoma University<br />

Health Sciences Center/Oklahoma Medical Research<br />

Foundation, Oklahoma City, OK<br />

Purpose: The etiology <strong>of</strong> systemic lupus erythematosus (SLE)<br />

is incompletely understood. Both genetic and environmental<br />

factors are implicated in the pathogenesis <strong>of</strong> the disease.<br />

Herein, we describe genetic association between SLE and<br />

polymorphisms in the IL-21 gene. The reported effects <strong>of</strong> IL-21<br />

on B cell differentiation into plasma cells, as well as its effect<br />

on dendritic cell maturation and T cell response, make IL-21 an<br />

attractive SLE candidate gene. Methods: A genetic association<br />

study was performed by genotyping three single nucleotide<br />

polymorphisms (SNPs) in the IL-21 gene in a total <strong>of</strong> 2636<br />

samples (1318 independent SLE cases and 1318 unrelated<br />

controls matched for age, sex and race). Genotyping was<br />

performed on the Illumina BeadStation 500GX system at the


Abstracts<br />

University <strong>of</strong> Texas Southwestern Microarray Core Facility<br />

(Dallas, TX). Population-based case-control association designs<br />

were employed. Results: Two SNPs located within intronal<br />

regions <strong>of</strong> IL-21 are associated with SLE (rs907715: chi2=11.55,<br />

p=0.00068; rs2221903: chi2=5.49, p=0.019). Furthermore,<br />

genotypes homozygous for the risk alleles were more frequent<br />

than genotypes homozygous for the non-risk alleles in<br />

European-American patients as compared to controls<br />

(rs907715 (GG versus AA): Odds ratio=1.66, p=0.0049;<br />

rs2221903 (GG versus AA): Odds ratio=1.60, p=0.025). Conclusion:<br />

We report an association between polymorphisms in the<br />

IL-21 gene and SLE. The functional effects <strong>of</strong> IL-21 polymorphisms,<br />

when revealed, might improve our understanding <strong>of</strong> SLE<br />

and provide new therapeutic targets.<br />

doi:10.1016/j.clim.2007.03.021<br />

OR.82 Both Shared and Strain-specific Genetic Loci<br />

Control Susceptibility to Virus-induced Autoimmune<br />

Diabetes in Two Rat Models<br />

Elizabeth Blankenhorn, Pr<strong>of</strong>essor, Drexel University College<br />

<strong>of</strong> Medicine, Philadelphia, PA, Laura Cort, Research<br />

Assistant, Department <strong>of</strong> Microbiology and <strong>Immunology</strong>,<br />

Philadelphia, PA, Daniel Cheeran, Research Assistant,<br />

Department <strong>of</strong> Microbiology and <strong>Immunology</strong>, Philadelphia,<br />

PA, Rebecca Tirabasi, Research Scientist, BRM, Inc.,<br />

Worcester, MA, Elaine Norowski, Research Assistant,<br />

Diabetes Division, Worcester, MA, Dennis Guberski,<br />

President, BRM, Inc., Worcester, MA, John Mordes,<br />

Pr<strong>of</strong>essor, Diabetes Division, Worcester, MA<br />

Viral infection may be important in the pathogenesis <strong>of</strong><br />

human type 1 diabetes (T1DM). MHC congenic (RT1u/u/a)<br />

LEW.1WR1 rats develop spontaneous T1DM at a low<br />

frequency (∼2%) and are also susceptible to the induction<br />

<strong>of</strong> diabetes in response to infection with a parvovirus, Kilham<br />

rat virus (KRV). An injection <strong>of</strong> 1×10 7 PFU KRV induces<br />

diabetes in ∼40% <strong>of</strong> animals, and co-injection <strong>of</strong> a low dose<br />

<strong>of</strong> poly I:C with the KRV increases disease penetrance to<br />

100%. KRV does not infect beta cells. In their susceptibility to<br />

KRV-T1DM, LEW.1WR1 rats resemble BBDR (RT1u/u/u) rats,<br />

but WF strain rats (also RT1u/u/u) are resistant to the<br />

induction <strong>of</strong> diabetes by this virus. In crosses <strong>of</strong> (BBDR×WF)<br />

rats, we have mapped two quantitative trait loci (QTL) that<br />

determine susceptibility to KRV-poly I:C induced T1DM,<br />

Iddm14 and Iddm20. We have now mapped the QTL required<br />

for diabetes in (LEW.1WR1 ×WF) rats. We show that Iddm14<br />

allele from the diabetes-susceptible LEW.1WR1 strain is<br />

required for both insulitis and diabetes (46/47 rats with<br />

diabetes had Iddm14d/−). However, Iddm20 is not a determinant<br />

<strong>of</strong> either insulitis or diabetes in this strain combination.<br />

Instead, markers on chromosome 20 near RT1 show<br />

significance. These data demonstrate that virus-specific<br />

diabetogenic responses differ between LEW.1WR1 and BBDR<br />

rats, even though both are susceptible to KRV. A genome scan<br />

for LEW.1WR1-specific QTL will be reported. These data shed<br />

light on light on the factors responsible for the variable<br />

penetrance <strong>of</strong> T1D in high risk human populations.<br />

doi:10.1016/j.clim.2007.03.022<br />

OR.83 An Association Between the Autoimmune<br />

Susceptibility Locus CTLA-4 and Alterations in<br />

Proximal TCR Signaling in Healthy Human<br />

Volunteers<br />

Lisa Maier, Neurology Fellow, Center for Neurologic<br />

Diseases, Boston, MA, David Anderson, Instructor, Center<br />

for Neurologic Diseases, Boston, MA, Philip de Jager,<br />

Assistant Pr<strong>of</strong>essor, Center for Neurologic Diseases, Boston,<br />

MA, David Hafler, Pr<strong>of</strong>essor, Center for Neurologic Diseases,<br />

Boston, MA, Linda Wicker, Pr<strong>of</strong>essor, JDRF/WT Diabetes and<br />

Inflammation Laboratory, Cambridge<br />

With the rapid advancement in high-throughput genotyping<br />

technologies, an increasing number <strong>of</strong> genetic variants have<br />

been associated with susceptibility to human autoimmune<br />

disease. However, little is known about the functional effects <strong>of</strong><br />

susceptibility variants on human immune cells. The single<br />

nucleotide polymorphism (SNP) called CT60 (rs3087243),<br />

located in the 3′ untranslated region <strong>of</strong> CTLA-4, has been<br />

associated with many autoimmune diseases, including type 1<br />

diabetes. The susceptible genotype at this SNP has previously<br />

been shown to result in a 2-fold lower production <strong>of</strong> the soluble<br />

CTLA-4mRNAis<strong>of</strong>orminnaïveCD4+Tcells.Wewerethus<br />

interested in whether this or other CT60-associated functional<br />

consequences would affect TCR signaling, given that CTLA-4 is a<br />

negative T cell regulator. We have applied a flow-cytometric<br />

technique that measures phosphoproteins to the study <strong>of</strong><br />

proximal TCR signaling in naïve and memory CD4+ T cells using<br />

fresh whole blood stimulated with anti-CD3 monoclonal antibody,<br />

and have measured CD3z (pY142), LAT (pY171), LCK<br />

(pY505), ZAP70 (pY292), ZAP70 (pY319/Syk (pY352)) and SLP-76<br />

(pY128) in healthy human volunteers. By categorizing results<br />

obtained from TCR stimulation into the three genotype classes;<br />

AA, AG and GG, we observed no differences in TCR responses<br />

with CD4+CD45RA+ T cells. However, we observed that relative<br />

to the CD45RA+ subset, CD4+CD45RA- T cells from individuals<br />

with the susceptibility allele are less responsive in all TCR<br />

signaling molecules examined. These data contribute to our<br />

understanding <strong>of</strong> the link between the susceptibility genotype<br />

at CTLA4 and the mechanisms involved that may result in<br />

autoimmunity.<br />

doi:10.1016/j.clim.2007.03.023<br />

S135<br />

OR.84 Variants <strong>of</strong> CARD15/NOD2 Associated with<br />

Crohn’s Disease and Blau Syndrome Differ from Wild<br />

Type NOD2 in Regulating Cytokine Secretion<br />

Induced by TLR2 and TLR4 Agonists in a Transduced<br />

Macrophage Cell Line<br />

Michael Davey, Pr<strong>of</strong>essor <strong>of</strong> Medicine, Portland VA Medical<br />

Center and OHSU, Portland, OR, Zili Zhang, Assistant<br />

Pr<strong>of</strong>essor, OHSU, Pediatrics, Portland, OR, Tammy Martin,<br />

Associate Pr<strong>of</strong>essor, OHSU and CEI, Portland, OR, Holly<br />

Rosenzweig, Postdoctoral Fellow, OHSU, Casey Eye<br />

Institute, Portland, OR, Steven Planck, Pr<strong>of</strong>essor, OHSU and<br />

CEI, Portland, OR, James Rosenbaum, Pr<strong>of</strong>essor, OHSU and<br />

CEI, Portland, OR<br />

NOD2 mutations are associated with Crohn's disease and<br />

Blau syndrome. NOD2 senses muramyl dipeptide (MDP) and


S136 Abstracts<br />

activates NF-kB. Studies in NOD2 deficient mice found that<br />

NOD2 suppresses TLR2 cytokine responses. Using cells overexpressing<br />

NOD2, this study further explored the regulatory<br />

role <strong>of</strong> NOD2. Murine mononuclear cells (J774 line) were<br />

transduced with constructs containing murine wild type (WT)<br />

NOD2, the murine equivalent <strong>of</strong> common Blau (BL) (R314Q)<br />

and Crohn's mutations (frame shift [fs] at L980) or an empty<br />

retroviral vector control (VC). Cells were stimulated with<br />

TLR2 (PAM3CSK4) or TLR4 (LPS) agonists with or without MDP,<br />

and secretion <strong>of</strong> IL-6, IL-10, IL-12p40 and TNF-α was<br />

measured. Over-expression <strong>of</strong> WT, BL and fs variants <strong>of</strong><br />

NOD2 suppressed IL-10 secretion and enhanced TNF-α<br />

secretion in response to TLR2 and TLR4 agonists. Both<br />

agonists suppressed IL-6 and IL-12p40 secretion in WT cells<br />

but enhanced secretion from BL and fs cells. MDP and TLR<br />

ligands given simultaneously to VC cells enhanced secretion<br />

<strong>of</strong> all cytokines studied. Synergy did not occur or was<br />

suppressed in WT cells, while it was enhanced in BL and fs<br />

cells. These data confirm the regulatory role <strong>of</strong> NOD2 on TLR2<br />

signaling and show TLR4 pathways can also be regulated. The<br />

differences observed between suppression and enhancement<br />

<strong>of</strong> cytokine secretion with WTcompared to BL and fs variants<br />

<strong>of</strong> NOD2 may be in vitro correlates <strong>of</strong> processes linking these<br />

mutations with disease. Elucidating mechanisms responsible<br />

for these different patterns may enhance our understanding<br />

<strong>of</strong> Crohn's disease and Blau syndrome.<br />

doi:10.1016/j.clim.2007.03.024<br />

OR.85 A Genome-wide Assessment <strong>of</strong> the Genetic<br />

Basis <strong>of</strong> Type 1 Diabetes<br />

Vincent Plagnol, Mr, JDRF/WT DIL, University <strong>of</strong> Cambridge,<br />

Cambridge, England, David Clayton, Pr<strong>of</strong>essor, JDRF/WT<br />

Diabetes and Inflammation Laboratory, Cambridge,<br />

England, David Dunger, Department <strong>of</strong> Pediatric, University<br />

<strong>of</strong> Cambridge, Cambridge, England, Kate Downes, JDRF/WT<br />

Diabetes and Inflammation Laboratory, Cambridge,<br />

England, Hin-Tak Leung, JDRF/WT Diabetes and<br />

Inflammation Laboratory, Cambridge, England, Deborah<br />

Smyth, JDRF/WT Diabetes and Inflammation Laboratory,<br />

Cambridge, England, Helen Stevens, JDRF/WT Diabetes and<br />

Inflammation Laboratory, Cambridge, England, Neil Walker,<br />

Mr, JDRF/WT Diabetes and Inflammation Laboratory,<br />

Cambridge, England, WTCCC, Wellcome Trust Case Control<br />

Consortium, http://www.wtccc.org.uk/, Wellcome Trust,<br />

Cambridge, England, John Todd, Pr<strong>of</strong>essor, JDRF/WT<br />

Diabetes and Inflammation Laboratory, Cambridge, England<br />

The strong familial clustering <strong>of</strong> autoimmune type 1 diabetes<br />

(T1D) in families remains only partially explained. The six<br />

confirmed genes or chromosome regions with convincing<br />

statistical support in large, and multiple, populations, namely<br />

the major histocompatibility complex (MHC), the insulin gene<br />

(INS), CTLA-4, PTPN22, IL2RA/CD25, and IFIH1/MDA5 can<br />

explain only about 50% <strong>of</strong> familial aggregation. Nonetheless,<br />

their identification has provided many insights into the biology<br />

and pathways <strong>of</strong> T1D. Recent expansions in sample collections,<br />

including the Juvenile Diabetes Research Foundation/Wellcome<br />

Trust Diabetes and Inflammation Laboratory (JDRF/WT DIL)<br />

British T1D case sample (n= 8000), and advances in affordable,<br />

genome-wide, high throughput single nucleotide polymorphism<br />

(SNP) genotyping technologies (Affymetrix), has allowed an<br />

association analysis <strong>of</strong> 500,000 SNPs across the genome in 2000<br />

T1D cases and 3000 controls as part <strong>of</strong> the Wellcome Trust Case-<br />

Control Consortium (WTCCC). Attesting to the quality <strong>of</strong> the<br />

SNP map, positive results were obtained for all six susceptibility<br />

loci reported previously. However, follow-up genotyping studies<br />

<strong>of</strong> 14 other chromosome regions showing Pb10 −5 in the WTCCC<br />

scan in over 6000 cases and 6000 controls, and in approximately<br />

2000 families, supported in a convincing way (Pb10 −15 and odds<br />

ratios approximately 1.2) four regions associated with T1D in<br />

both the family and case-control samples, on chromosomes<br />

16p13, 18p11, 12q13 and 12q24. These results illustrate the<br />

power <strong>of</strong> genome-wide association approach to characterise<br />

the genetic basis <strong>of</strong> T1D.<br />

doi:10.1016/j.clim.2007.03.025<br />

Cytokine/Chemocine Regulators <strong>of</strong><br />

Inflammation & Allergy<br />

Sunday, June 10<br />

2:45 pm−4:45 pm<br />

OR.86 Human B Cell Cytokine Regulation and<br />

Multiple Sclerosis (MS)<br />

Christine Ghorayeb, McGill University/<strong>Immunology</strong>,<br />

Montreal, QC, Masaaki Nii Hokkaido, University Hospital/<br />

Neurology, Sapporo, Japan, Claudia Calder, McGill<br />

University/Neuroimmunology, Montreal, QC, Canada, Amit<br />

Bar-Or, McGill University/Neurology and Neurosurgery,<br />

Montreal, QC, Canada<br />

B cells are increasingly recognized for roles in both normal<br />

immune responses and in autoimmune diseases including MS.<br />

We recently reported that normal human B cells can produce<br />

either pro-inflammatory (TNFá; Lymphotoxin/LT) or regulatory<br />

(IL-10) effector cytokines, depending on their sequence<br />

<strong>of</strong> activation. We further identified that MS patient B cells<br />

exhibit deficient expression <strong>of</strong> IL-10, implicating a B cell<br />

abnormality in failed immune regulation. Here, we asked (i)<br />

whether and how the local cytokine milieu may modulate the<br />

pr<strong>of</strong>ile <strong>of</strong> the normal effector B cell cytokine network, and<br />

(ii) whether such modulation is different for MS B cells.<br />

Specifically, we added either IFNγ (Th1 milieu) or IL-4 (Th2<br />

milieu) during our established ex vivo paradigm <strong>of</strong> B cell<br />

activation. The presence <strong>of</strong> IFNγ induced normal B cells to<br />

secrete increased levels <strong>of</strong> LT (429±44; versus 259±28 pg/<br />

ml; n=15; p=0.0002) and TNFá (274±44 versus 144±30 pg/<br />

ml; p=0.0003). IL-4 also induced increased levels <strong>of</strong> LT (419±<br />

47 pg/ml, p=0.0001) and <strong>of</strong> TNFá (366±51, pb0.0001),<br />

while significantly suppressing secretion <strong>of</strong> IL-10 (37±11<br />

versus 145 ±26 pg/ml; pb0.0001). We further discovered<br />

that in the ‘Th1 milieu’, MS B cells produced significantly<br />

higher levels <strong>of</strong> LT (average increase 28%; n=22, p=0.0069)<br />

and TNFá (26%, p=0.0049), compared to normal B cells. We<br />

demonstrate that a novel regulatory network <strong>of</strong> effector<br />

cytokines in normal B cells is significantly modulated<br />

depending on the local cytokine milieu. Furthermore, in a<br />

'Th1 milieu', MS B cells secrete abnormally high levels <strong>of</strong> TNFá


Abstracts<br />

and LT, which would contribute to the local inflammatory<br />

response.<br />

doi:10.1016/j.clim.2007.03.026<br />

OR.87 Immune Mediated Protection in Multiple<br />

Sclerosis: A Role for Neurokines in Oligodendrocyte<br />

Survival and Macrophage Modulation<br />

Niels Hellings, PhD, Hasselt University, Diepenbeek,<br />

Belgium, Niels Hellings, PhD, Hasselt University, BIOMED,<br />

Diepenbeek, Belgium, Jerome JJA Hendriks, PhD, Hasselt<br />

University, BIOMED, Diepenbeek, Belgium, S<strong>of</strong>ie Carmans,<br />

MsC, Hasselt University, BIOMED, Diepenbeek, Belgium,<br />

Leen Slaets, MsC, Hasselt University, BIOMED, Diepenbeek,<br />

Belgium, Debora Dumont, PhD, Hasselt University, BIOMED,<br />

Diepenbeek, Belgium, Joris Vanderlocht, PhD, Hasselt<br />

University, BIOMED, Diepenbeek, Belgium, Piet Stinissen,<br />

PhD, Hasselt University, BIOMED, Diepenbeek, Belgium<br />

In multiple sclerosis (MS), immune mediated destruction<br />

<strong>of</strong> the myelin sheath, oligodendrocytes and axons leads to<br />

irreversible neurological deficits. Recent data show that<br />

immune responses in the central nervous system (CNS) may<br />

also confer protective effects. We recently demonstrated<br />

that autoreactive T cells and macrophages produce<br />

leukemia inhibitory factor (LIF), a member <strong>of</strong> the IL-6<br />

family <strong>of</strong> neurokines. CD4+ T cells from relapsing remitting<br />

MS-patients show a reduced LIF production. Still, LIF<br />

immunoreactivity is detected among T cells and perivascular<br />

macrophages in MS lesions. In rat oligodendrocyte<br />

cultures, LIF protects against TNF-α and IFN-γ induced<br />

apoptosis, but not against other cell death mediators<br />

(oxidative stress, staurosporin). LIF receptor signalling in<br />

oligodendrocytes does not induce the expression <strong>of</strong><br />

suppressors <strong>of</strong> cytokine signalling (SOCS) or the antiapoptotic<br />

molecules Bcl-2/Bcl-XL. Using quantitative proteomics,<br />

we demonstrate that LIF leads to upregulation <strong>of</strong><br />

a panel <strong>of</strong> proteins that have pro-survival functions. Future<br />

experiments are needed to study their role in LIF mediated<br />

oligodendrocyte protection. Since macrophages, but not T<br />

cells, express functional LIF-receptor complexes we studied<br />

whether LIF also exerts immunomodulatory functions.<br />

LIF significantly upregulates myelin phagocytosis and<br />

reduces the production <strong>of</strong> reactive oxygen species and<br />

TNF-α in vitro. In summary, neurokines may act on various<br />

levels during CNS inflammation: they may modulate<br />

immune responses and protect CNS resident cells under<br />

attack. Further clarification in the actions <strong>of</strong> different<br />

neurokine members during neuroinflammation may lead to<br />

new therapeutic strategies for MS.<br />

doi:10.1016/j.clim.2007.03.027<br />

OR.88 ALCAM is a Novel Adhesion Molecule <strong>of</strong> the<br />

Inflammed Endothelium Involved in Leukocyte<br />

Trafficking to the Central Nervous System<br />

Romain Cayrol, PhD Student, Université de Montreal,<br />

Montreal, QC, Canada, Aurore Dodelet-Devillers, M.Sc.<br />

Student, Université de Montreal, Montreal, QC, Canada, Igal<br />

Ifergan, PhD Student, Université de Montreal, Montreal, QC,<br />

Canada, Arsalan Haqqani, PhD Student, National Research<br />

Council, Institute for Biological Sciences, Ottawa, ON,<br />

Canada, Hania Kebir, PhD Student, Université de Montreal,<br />

Montreal, QC, Canada, Danica Stanimirovic, Pr<strong>of</strong>essor,<br />

Institute for Biological Sciences; National Research Council <strong>of</strong><br />

Canada, Ottawa, ON, Canada, Alexandre Prat, Clinician and<br />

Pr<strong>of</strong>essor, Université de Montreal, Montreal, QC, Canada<br />

Leukocyte trafficking from the blood to local inflammatory<br />

sites is essential for the initiation and maintenance <strong>of</strong> tissue<br />

specific immune responses. In autoimmune diseases such as<br />

multiple sclerosis (MS), leukocyte transmigration from the<br />

blood to the target organ is dependent on intercellular cell<br />

adhesion molecule 1 (ICAM-1) and vascular cell adhesion<br />

molecule 1 (VCAM-1) expressed by the endothelial cells (ECs).<br />

In this study we describe CD166/activated leukocyte cell<br />

adhesion molecule (ALCAM) as a novel adhesion molecule <strong>of</strong><br />

the human blood–brain barrier (BBB). We demonstrate that<br />

inflammatory cytokines up-regulate the expression <strong>of</strong> ALCAM<br />

on the surface <strong>of</strong> BBB-ECs in vitro and that endothelial ALCAM<br />

co-localizes with leukocyte expressed-CD6 in the transmigratory<br />

cup during migration. We further show that ALCAM<br />

blocking antibody restricts the transmigration <strong>of</strong> CD4, CD14<br />

and CD19 immune cells across human BBB-ECs and that ALCAM<br />

expression is increased on ECs within active MS lesions, as<br />

compared to normal appearing white matter and to non-MS<br />

brains. These results suggest an important role for ALCAM in<br />

the recruitment <strong>of</strong> leukocytes to the CNS and identify ALCAM<br />

as a potential target to modulate CNS inflammatory reactions.<br />

doi:10.1016/j.clim.2007.03.028<br />

S137<br />

OR.89 Circulation <strong>of</strong> Membrane-bound TNFa by Way<br />

<strong>of</strong> Plasma-derived Exosomes<br />

Nicole Bianco, Postdoctoral Scholar, University <strong>of</strong><br />

Pittsburgh, Molecular Genetics and Biochemistry,<br />

Pittsburgh, PA, Seon-Hee Kim, Director <strong>of</strong> Research, Seoul<br />

National University, Seoul, South Korea, Paul Robbins,<br />

Pr<strong>of</strong>essor, University <strong>of</strong> Pittsburgh, Molecular Genetics and<br />

Biochemistry, Pittsburgh, PA, Teresa Hennon, Research<br />

Fellow, University <strong>of</strong> Pittsburgh, Molecular Genetics and<br />

Biochemistry, Pittsburgh, PA<br />

Exosomes are membrane nanovesicles (50–100 nm diameter)<br />

generated by reverse budding <strong>of</strong> the limiting membrane<br />

<strong>of</strong> multivesicular late endosomes. Exosomes originating<br />

from immune cells, such as B cells, T cells, DC, and mast<br />

cells, are thought to play a role in communicating immunoregulatory<br />

signals to other cells in either an immuno-stimulating<br />

or suppressive manner. Since we have demonstrated<br />

the presence <strong>of</strong> FasL, a TNF family member, in exosomes, we<br />

examined whether exosomes also can carry TNFa. To establish<br />

this, we first generated exosomes from a murine tumor<br />

cell line overexpressing murine TNFa and demonstrated the<br />

presence <strong>of</strong> membrane-bound TNFa (mbTNFa) in exosomes by<br />

FACS and Western blot analysis. The mbTNFa in exosomes was<br />

also functional in the L929 cytotoxicity assay. To determine<br />

whether exosomes containing mbTNFa might circulate<br />

systemically during active disease, we collected serum from<br />

WT or IL10−/− mice with inflammatory bowel disease. There


S138 Abstracts<br />

was significantly more mbTNFa in IL10−/− serum exosomes.<br />

Mice with collagen-induced arthritis also contained elevated<br />

levels <strong>of</strong> mbTNFa in their serum exosomes. This research<br />

implies that mbTNFa, in addition to functioning in a local<br />

manner by cell-to-cell contact, can also travel either locally<br />

or systemically to regulate inflammation at distant sites. This<br />

research also suggests that the bioactive mbTNFa in the<br />

vesicles may play a role in the pathology <strong>of</strong> various TNFdependent<br />

autoimmune diseases and that anti-TNF therapies<br />

might target mbTNFa carried on microvesicles. Furthermore,<br />

the level <strong>of</strong> TNFa in the serum exosomes might be useful as a<br />

diagnostic marker <strong>of</strong> certain autoimmune diseases.<br />

doi:10.1016/j.clim.2007.03.029<br />

OR.90 Ccr2 Deficiency Impairs Microglial<br />

Accumulation and Accelerates Progression <strong>of</strong><br />

Alzheimer’s Disease<br />

Joseph El Khoury, Assistant Pr<strong>of</strong>essor <strong>of</strong> Medicine, Harvard<br />

Medical School, Charlestown, MA, Michelle T<strong>of</strong>t, Laboratory<br />

Technician, Massachusetts General Hospital, Charlestown,<br />

MA, Suzanne Hickman, Scientist, Massachusetts General<br />

Hospital, Charlestown, MA, Changiz Geula, Pr<strong>of</strong>essor,<br />

Harvard Medical School, Charlestown, MA, Terry Means,<br />

Instructor in Medicine, Massachusetts General Hospital,<br />

Charlestown, MA, Andrew Luster, Pr<strong>of</strong>essor <strong>of</strong> Medicine,<br />

Harvard Medical School, Charelstown, MA<br />

Microglia are the principal immune cell <strong>of</strong> the brain. In<br />

Alzheimer's disease (AD), microglia accumulate in senile<br />

plaques and may in part be recruited from peripheral blood.<br />

The role <strong>of</strong> microglia in AD pathogenesis has not been<br />

resolved. Microglia may play a neuroprotective role by phagocytosing<br />

beta amyloid, a pathogenic protein deposited in<br />

AD brains. But their activation and subsequent secretion <strong>of</strong><br />

neurotoxins may also lead to neurodegeneration. Chemokine<br />

receptor 2 (Ccr2) is a chemokine receptor expressed on<br />

microglia that mediates accumulation <strong>of</strong> leukocytes at sites<br />

<strong>of</strong> inflammation. Transgenic mice expressing a mutated form<br />

<strong>of</strong> the human amyloid precursor protein (APP) are a robust<br />

model <strong>of</strong> AD used to study the role <strong>of</strong> microglia in AD. To<br />

determine the role <strong>of</strong> Ccr2 in microglial accumulation in AD,<br />

we generated transgenic APP mice deficient in Ccr2 and<br />

analyzed these mice for AD-like pathology. We found that<br />

Ccr2 deficiency accelerates early disease progression and<br />

markedly reduces microglial recruitment and accumulation<br />

to baseline in this transgenic mouse model <strong>of</strong> AD. APP mice<br />

deficient in Ccr2 died prematurely in correlation with Ccr2<br />

gene dosage, and accumulated beta amyloid in their brains<br />

much earlier than APP mice with normal Ccr2 levels,<br />

indicating that absence <strong>of</strong> early microglial recruitment<br />

leads to decreased beta amyloid clearance. Furthermore,<br />

Ccr2-deficient mice microinjected with beta amyloid into<br />

their brains failed to recruit microglia to the sites <strong>of</strong><br />

injection. Thus, Ccr2-dependent microglial accumulation<br />

plays a protective role in the early stages <strong>of</strong> AD by promoting<br />

clearance <strong>of</strong> beta amyloid.<br />

doi:10.1016/j.clim.2007.03.030<br />

OR.91 Chemokine-like Receptor-1 (CMKLR1) Regulates<br />

Experimental Autoimmune Encephalomyelitis<br />

Kareem L. Graham, Postdoctoral Scholar, Department <strong>of</strong><br />

Pathology, Stanford, CA, Luis A. Zuniga, Graduate Student,<br />

Department <strong>of</strong> Pathology, Stanford, CA, Brian A. Zabel,<br />

Postdoctoral Fellow, Department <strong>of</strong> Pathology, Stanford,<br />

CA, Eugene C. Butcher, Pr<strong>of</strong>essor <strong>of</strong> Pathology, Department<br />

<strong>of</strong> Pathology, Stanford, CA, Raymond A. Sobel, Pr<strong>of</strong>essor <strong>of</strong><br />

Pathology, Department <strong>of</strong> Pathology, Stanford, CA<br />

The pathology <strong>of</strong> multiple sclerosis (MS) involves leukocyte<br />

extravasation <strong>of</strong> the blood–brain barrier and associated myelin<br />

damage, which leads to impaired nerve function and paralysis.<br />

Chemokines, adhesion molecules, and their receptors have been<br />

implicated in recruitment <strong>of</strong> inflammatory cells to the central<br />

nervous system (CNS) during MS. However, the mechanisms that<br />

regulate migration <strong>of</strong> various leukocyte subsets to the CNS<br />

remain poorly understood. Chemokine-like receptor-1 (CMKLR1,<br />

also known as ChemR23 or Dez) is a recently de-orphaned<br />

chemoattractant receptor that has emerging roles in macrophage<br />

migration during inflammatory processes. In this study,<br />

we examined the role <strong>of</strong> CMKLR1 in experimental autoimmune<br />

encephalomyelitis (EAE), a mouse model <strong>of</strong> human MS. We found<br />

that mice deficient in CMKLR1 are resistant to progressive EAE.<br />

CMKLR1-deficient mice had reduced inflammatory infiltrates in<br />

the brain and spinal cord, with especially marked differences in<br />

the CNS parenchyma. While transcripts for CMKRL1 have been<br />

detected in a variety <strong>of</strong> tissues, experiments with bone marrow<br />

chimeric mice indicate that CMKLR1 expression on cells <strong>of</strong><br />

hematopoietic origin is required for maximal expression <strong>of</strong> EAE.<br />

Together, the data demonstrate that CMKLR1 regulates a model<br />

<strong>of</strong> autoimmune demyelinating disease and suggest that CMKLR1<br />

may be a potential target in blocking development <strong>of</strong><br />

progressive EAE/MS.<br />

doi:10.1016/j.clim.2007.03.031<br />

OR.92 Cell-type Specific Changes in Cytokine-STAT<br />

Signal Transduction Stage Systemic Lupus<br />

Erythematosus<br />

Matthew, Postdoctoral Fellow, Stanford University,<br />

Department <strong>of</strong> Microbiology and <strong>Immunology</strong>, Stanford, CA,<br />

Peter Krutzik, Research Associate, Stanford University,<br />

Department <strong>of</strong> Microbiology and <strong>Immunology</strong>, Stanford, CA,<br />

Garry Nolan, Pr<strong>of</strong>essor, Stanford University, Department <strong>of</strong><br />

Microbiology and <strong>Immunology</strong>, Stanford, CA<br />

The regulation <strong>of</strong> cytokine signal transduction is thought to<br />

be critical in shaping the character and duration <strong>of</strong> immune<br />

responses, but little is known <strong>of</strong> its role in autoimmune disease.<br />

We applied phospho-specific flow cytometry to generate a<br />

highly multiplexed view <strong>of</strong> how Systemic Lupus Erythematosus<br />

(SLE) disturbs cytokine signaling through the STAT family <strong>of</strong><br />

transcription factors. Using a panel <strong>of</strong> 10 cytokines previously<br />

suggested to have causal roles in the disease, we measured<br />

signaling responses at the single-cell level in multiple immune<br />

cell types from human SLE patients with a range <strong>of</strong> disease<br />

activity, as well as followed disease progression in the MRLlpr<br />

murine model. Unexpected changes in subset responses to IFNα,<br />

IFNγ, IL-4, IL-6, IL-15 and IL-21 were observed. Suppressor <strong>of</strong><br />

cytokine signaling 1 (SOCS1) levels were elevated in PBMC from


Abstracts<br />

human patients as well as in lymphocytes from the murine model<br />

where SOCS1 was implicated in pathway-specific inhibition <strong>of</strong><br />

responses to IL-6 in T cell subsets. Not only did aberrant signal<br />

transduction distinguish SLE patients from normal donors, but<br />

robustly significant differences in certain subsets discriminated<br />

patients experiencing flare from those with low levels <strong>of</strong> disease<br />

activity. Importantly, cell-type specific regulation <strong>of</strong> cytokine<br />

signaling may mark an important transition between flare and<br />

remission in humans. These results provide unique mechanistic<br />

insights into the signaling disruptions that occur during SLE,<br />

demonstrate that this approach can rapidly relate disease status<br />

to signaling network changes, and suggest it will be possible to<br />

stratify patients with greater accuracy.<br />

doi:10.1016/j.clim.2007.03.032<br />

OR.93 Accelerated Development <strong>of</strong><br />

Glomerulonephritis in TNF Receptor 1 and 2<br />

Double-knockout Lupus-mice<br />

Noam Jacob, Research Fellow, University <strong>of</strong> Southern<br />

California Keck School <strong>of</strong> Medicine, Los Angeles, CA,<br />

Luminita Pricop, Associate Pr<strong>of</strong>essor <strong>of</strong> Medicine, Hospital<br />

for Special Surgery, New York, NY, Chaim Putterman,<br />

Associate Pr<strong>of</strong>essor, Albert Einstein College <strong>of</strong> Medicine/<br />

Department <strong>of</strong> Medicine, Bronx, NY, Chaim O. Jacob,<br />

Associate Pr<strong>of</strong>essor <strong>of</strong> Medicine, University <strong>of</strong> Southern<br />

California Keck School <strong>of</strong> Medicine/Department <strong>of</strong><br />

Medicine, Los Angeles, CA, Michael Koss, Pr<strong>of</strong>essor <strong>of</strong><br />

Pathology, University <strong>of</strong> Southern California Keck School <strong>of</strong><br />

Medicine/Department <strong>of</strong> Pathology, Los Angeles, CA<br />

We have developed lupus-prone (NZB×NZW)F1-derived<br />

congenic New Zealand Mixed (NZM) 2328 lines that are deficient<br />

in TNF receptor 1 (TNFR1), TNF receptor 2 (TNFR2) or in both<br />

TNFR1 and TNFR2. While the development <strong>of</strong> lupus-nephritis in<br />

TNFR2 deficient mice was very similar to wild type, TNFR1<br />

deficient mice had only a slightly delayed disease development<br />

compared to TNF receptors intact NZM 2328 controls. On the<br />

other hand, mice that lacked both TNFR1 and TNFR2 developed<br />

a significantly accelerated lupus-nephritis and increased<br />

mortality associated with increased levels <strong>of</strong> IgG anti-doublestranded<br />

DNA autoantibodies both in the periphery and in the<br />

glomerular deposits. Double knockout mice have significantly<br />

enlarged spleens compared to all other lines, which is<br />

associated with increased number <strong>of</strong> B and T lymphocytes<br />

mostly <strong>of</strong> CD4+ subsets. Most notably, double KO mice have a 3–<br />

5 fold increase in the number <strong>of</strong> activated memory Tcells (CD4+<br />

CD25− CD44high CD62Llow) while TNFR1 deficient and TNFR2<br />

deficient are not different from each other. Immunostaining <strong>of</strong><br />

spleens shows spontaneous germinal center formation in wild<br />

type NZM 2328 and TNFR2 deficient mice. TNFR1 deficient mice<br />

have reduced germinal center formation at 2 months and still at<br />

6 months <strong>of</strong> age, while double KO mice have reduced GC<br />

formation at 2 months but by 6 months have as much or more<br />

than the control mice. These results suggest a fine and complex<br />

balance between the two TNF receptors and underscore the<br />

need for both receptors in the immune development <strong>of</strong><br />

autoimmune kidney disease.<br />

doi:10.1016/j.clim.2007.03.033<br />

OR.94 Protective and Therapeutic Role for α<br />

B-Crystallin in Autoimmune Demyelination<br />

Shalina Ousman, Stanford University, Stanford, CA, Beren<br />

Tomooka, Mr. Stanford University, Division <strong>of</strong> <strong>Immunology</strong><br />

and Rheumatology, Palo Alto, CA, Johannes Van Noort,<br />

Department <strong>of</strong> Biosciences, Leiden, The Netherlands, Kevin<br />

O’Conner, Harvard Medical School, Brigham and Women’s<br />

Hospital, Boston, MA, Eric Wawrousek, National Eye<br />

Institute, Bethesda, MD, David Hafler, Harvard Medical<br />

School, Brigham and Women's Hospital, Boston, MA,<br />

Raymond Sobel, Department <strong>of</strong> Pathology, Stanford<br />

University, Palo Alto, CA, William Robinson, Division <strong>of</strong><br />

<strong>Immunology</strong> and Rheumatology, Stanford University,<br />

Stanford, Palo Alto, CA, Lawrence Steinman, Pr<strong>of</strong>essor,<br />

Department <strong>of</strong> Neurology and Neurological Studies,<br />

Stanford University, Stanford, CA<br />

Alpha B-crystallin (±BC) is the major target <strong>of</strong> the immune<br />

response to myelin from multiple sclerosis (MS) and control<br />

brain. It is the most abundant transcript uniquely present in<br />

MS lesions and, recent studies have identified anti-apoptotic<br />

and anti-inflammatory roles for±BC. We hypothesize that<br />

this crystallin plays a key protective role in demyelinating<br />

disease. ±BC−/− mice displayed worse symptoms <strong>of</strong> experimental<br />

allergic encephalomyelitis (EAE), the animal model <strong>of</strong><br />

MS, with higher Th1 and Th-17 cytokine secretion, proliferation<br />

and p38 signaling by their T cells and macrophages and,<br />

more intense CNS inflammation and demyelination compared<br />

to their WT counterparts. Glial cells from ±BC−/− mice also<br />

expressed more cleaved caspase-3, TUNEL staining and<br />

enhanced ERK and NFkB signaling. These results indicated<br />

that ±BC plays a protective role in curbing both glial cell<br />

death and immune cell activation. Of interest, myelin antigen<br />

arrays identified antibodies to native ±BC and to p21–40<br />

and p116–135 <strong>of</strong> ±BC in the cerebrospinal fluid <strong>of</strong> patients<br />

with relapsing remitting MS (RRMS). These antibodies could<br />

neutralize ±BC function. We then assessed whether recombinant<br />

±BC itself could resolve disease in mice with ongoing<br />

EAE. Remarkably, clinical disease was ameliorated in ±BCtreated<br />

animals along with decreased Th1 and Th17 cytokine<br />

production and glial cell death. The immune response against<br />

a negative regulator <strong>of</strong> inflammation in demyelinating<br />

disease, would promote inflammation and demyelination,<br />

and this can be countered therapeutically by giving ±BC itself.<br />

Supported by NIH, National Multiple Sclerosis Society and<br />

Multiple Sclerosis Society <strong>of</strong> Canada.<br />

doi:10.1016/j.clim.2007.03.034<br />

Regulating Inflammation<br />

Sunday, June 10<br />

2:45 pm−4:45 pm<br />

S139<br />

OR.95 Defect in TIM-3 Regulation <strong>of</strong> CD4+ T Cells in<br />

a Human Autoimmune Disease<br />

Li Yang, Postdoctoral Fellow, Center for Neurologic<br />

Diseases, Harvard Medical School, Boston, MA, David E.<br />

Anderson, Instructor, Center for Neurologic Diseases,<br />

Harvard Medical School, Boston, MA, Vijay K. Kuchroo,<br />

Pr<strong>of</strong>essor, Center for Neurologic Diseases, Harvard Medical


S140 Abstracts<br />

School, Boston, MA, David A. Hafler, Pr<strong>of</strong>essor, Center for<br />

Neurologic Diseases, Harvard Medical school, Boston, MA<br />

T cell immunoglobulin- and mucin-domain-containing molecules-3<br />

(Tim-3), a T helper type 1 (Th1)-specific cell surface<br />

molecule, functions as an inhibitory molecule in mice. However,<br />

lessisknownaboutthefunction<strong>of</strong>TIM-3inhumans.Wehave<br />

previously shown that reduction <strong>of</strong> TIM-3 expression on ex vivo<br />

human CD4+ T cells by small interfering RNA (siRNA) oligos<br />

increases cell proliferation and IFN-γ secretion, which confirms<br />

a negative role <strong>of</strong> TIM-3 in regulation <strong>of</strong> human T cell function.<br />

We now report the results <strong>of</strong> a study that interrogated the<br />

function <strong>of</strong> TIM-3 regulation <strong>of</strong> CD4+ T cells in the autoimmune<br />

diseasemultiplesclerosis(MS).Westimulatedex vivo CD4+ T<br />

cellsfrompatientswithMS(n=54) and healthy subjects (n=37)<br />

with anti-CD3/CD28 monoclonal antibodies in the presence or<br />

absence <strong>of</strong> anti-TIM-3 blocking antibody. Our data indicate that<br />

in the presence <strong>of</strong> anti-TIM-3 monoclonal antibody treatment<br />

there is a significant increase <strong>of</strong> IFN-γ secretion from CD4+ T<br />

cells in healthy subjects but not in untreated patients with MS.<br />

Analysis <strong>of</strong> the kinetics <strong>of</strong> TIM-3 expression demonstrates lower<br />

steady state levels <strong>of</strong> TIM-3 and delayed kinetics <strong>of</strong> upregulation<br />

on T cells present in patients with MS relative to<br />

healthy subjects. Interestingly, T cells obtained from patents<br />

treated with interferons restored TIM-3 function, as IFN-γ<br />

secretion in interferons-treated patients is increased with TIM3<br />

blockade when activated at lower doses <strong>of</strong> anti-CD3. Taken<br />

together, our results demonstrate that there is a significant<br />

defect in TIM-3 regulation <strong>of</strong> CD4 Tcells in a human autoimmune<br />

disease.<br />

doi:10.1016/j.clim.2007.03.035<br />

OR.96 Increasing GABA Activity Prevents<br />

Autoimmune Neuroinflammation<br />

Roopa Bhat, Post Doctoral Fellow, Stanford University,<br />

Neurology and Neurological Sciences, Stanford, CA,<br />

Christopher Lock, Vice President, Research and Development,<br />

Carantech BioSciences, Inc. San Francisco, CA, Lawrence<br />

Steinman, Pr<strong>of</strong>essor and Chair, Stanford University,<br />

Interdepartmental Program in <strong>Immunology</strong>, Stanford, CA<br />

Gamma-amino butyric acid (GABA) is the principal inhibitory<br />

neurotransmitter in the central nervous system (CNS). GABA is<br />

made by glutamic acid decarboxylase (GAD) and catabolized by<br />

GABA transaminase (GABAT). Using transcriptional microarrays,<br />

we showed that, in acute/active inflammatory brain lesions in<br />

multiple sclerosis (MS) patients, GABAT is upregulated 38-fold<br />

while specific GABA receptor subunits and GAD are downregulated<br />

(Lock et al., Nat Med 8: 500–508, 2002). GABA<br />

inhibits the development <strong>of</strong> autoimmunity, pro-inflammatory T<br />

cell responses, and disease progression in the mouse model <strong>of</strong><br />

autoimmune type1 diabetes (Tian et al., JImmunol173: 5298–<br />

5304, 2004). We, therefore, hypothesized that GABA ameliorates<br />

autoimmune inflammation in MS. To test this hypothesis,<br />

we used the mouse model <strong>of</strong> MS, experimental autoimmune<br />

encephalomyelitis, and pre-treated daily with vigabatrin,<br />

which irreversibly inhibits GABAT. We found that vigabatrin<br />

ameliorated paralysis significantly: disease incidence<br />

decreased by 64%, while the severity and number <strong>of</strong> relapses<br />

diminished by 10-fold and 72±14% (SEM) respectively. Topi-<br />

ramate, which binds the GABA-A receptor, also prevented CNS<br />

autoimmunity in a similar manner. This indicates a common<br />

mechanism that acts by increasing GABA-ergic function<br />

whether that be through the GABA metabolic or signaling<br />

pathway. Both agents suppressed myelin-specific T cell<br />

proliferation and inhibited the production <strong>of</strong> TNF, IFN- 3 ,IL-17<br />

and IL-6. Inherent abnormalities in the GABA metabolic and<br />

signaling pathway may play a previously unrecognized role in<br />

the pathogenesis <strong>of</strong> MS. Increasing GABA-ergic function using<br />

agents such as those in our study could be beneficial in the<br />

prevention <strong>of</strong> autoimmune disorders such as MS.<br />

doi:10.1016/j.clim.2007.03.036<br />

OR.97 Cyclooxygenase-2 Deficiency has a Protective<br />

Function in Liver Ischemia/Reperfusion Injury<br />

Takashi Hamada, Postdoctoraltoral Fellow, The<br />

Dumont-University <strong>of</strong> California, Los Angeles Transplant<br />

Center, Los Angeles, CA, Armine Avanesyan, Medical<br />

Student, The Dumont-University <strong>of</strong> California, Los Angeles<br />

Transplant Center, Los Angeles, CA, Sei-ichiro Tsuchihashi,<br />

Postdoctoral Fellow, The Dumont-University <strong>of</strong> California,<br />

Los Angeles Transplant Center, Los Angeles, CA, Carolina<br />

Moore, Student, The Dumont-University <strong>of</strong> California, Los<br />

Angeles Transplant Center, Los Angeles, CA, Ana Coito,<br />

Associate Pr<strong>of</strong>essor, The Dumont University <strong>of</strong> California,<br />

Los Angeles Transplant Center, Los Angeles, CA<br />

Cyclooxygenase-2 (COX-2) is a potent mediator <strong>of</strong> inflammation.<br />

In liver ischemia/reperfusion (I/R) injury, COX-2<br />

expression is highly correlated with the degree <strong>of</strong> tissue<br />

damage. This work tests the hypothesis that COX-2 expression<br />

has a critical function in the pathogenesis <strong>of</strong><br />

hepatic I/R injury. Methods and Results: COX-2 −/− mice<br />

(KO) and matched wild-type (WT) (COX-2 +/+) controls<br />

(n=8/g) were submitted to partial warm ischemia in the<br />

left/medium hepatic lobes for 90 minutes, followed by<br />

reperfusion. H&E staining <strong>of</strong> liver sections in COX-2 −/−<br />

mice at 24 h post-reperfusion revealed overall good<br />

histological preservation with modest signs <strong>of</strong> vascular<br />

congestion and necrosis, contrasting with significant<br />

edema, centrilobular ballooning, hepatocellular necrosis<br />

(N60%), and sinusoidal congestion observed in the WT<br />

counterparts (score: 1.5 ±1 vs. 3.5 ±0.6; pb0.03). Liver<br />

function, as evidenced by sGOT levels (IU/L), was improved<br />

in COX-2 (−/−) mice as compared with WT mice (206±45 vs.<br />

1244 ±290; pb0.01). MPO activity (U/gm) was also significantly<br />

lower in COX-2 (−/−) mice (1.2±0.4 vs. 3.4 ±1.5;<br />

pb0.01). Moreover, TUNEL staining showed reduced levels<br />

<strong>of</strong> apoptosis in COX-2 KO livers as compared with controls<br />

(16.2 ±3.3 vs. 73.6± 7.9, pb0.01). This was correlated with<br />

significant higher levels <strong>of</strong> BclXL, an anti-apoptotic member<br />

<strong>of</strong> the Bcl-2 family, in the COX-2 (−/−) livers. Conclusion:<br />

Our findings evidence an important function for COX-2 in<br />

the pathogenesis <strong>of</strong> hepatic I/R injury. They provide also<br />

the rationale for identification <strong>of</strong> improved therapeutic<br />

approaches to prevent hepatic I/R injury, and consequently<br />

for improving the outcome <strong>of</strong> liver transplantation.<br />

doi:10.1016/j.clim.2007.03.037


Abstracts<br />

OR.98 Crosstalk Between Leptin and LPS Signaling<br />

Pathways to Upregulate CD40 Expression via Akt in<br />

Dendritic Cells<br />

Queenie Lai Kwan Lam, PhD Candidate, University <strong>of</strong> Hong<br />

Kong, Department <strong>of</strong> Pathology, Hong Kong, Liwei Lu,<br />

Pr<strong>of</strong>essor, University <strong>of</strong> Hong Kong, Department <strong>of</strong><br />

Pathology, Hong Kong<br />

A body <strong>of</strong> evidence has demonstrated the regulatory role <strong>of</strong><br />

leptin in the immune system over the past decade but little is<br />

clear about how it regulates dendritic cell (DC) function. CD40, a<br />

cell-surface receptor and a costimulatory molecule, is a key<br />

determinant for the activity <strong>of</strong> DC in immunity and tolerance.<br />

We have previously shown that leptin signaling is critically<br />

involved in DC maturation and survival. In this study, we showed<br />

that leptin induced CD40 expression synergistically with LPS. In<br />

an Akt-dependent manner, both leptin and LPS activated the<br />

downstream transcription factors Nuclear Factor (NF)-kappaBp65<br />

and Signal Transducer and Activator <strong>of</strong> Transcription-<br />

1α (STAT-1α) in DC. Using dominant negative forms <strong>of</strong> Akt and<br />

IKK, as well as small interfering RNA (siRNA) for STAT-1α, we<br />

showed that Akt, STAT-1α and NF-kappaBp65 were important for<br />

CD40 expression induced by leptin, LPS and their synergistic<br />

action. Blocking studies demonstrated a crucial role for Akt in<br />

the translocation <strong>of</strong> STAT-1α and NF-kappaBp65 to the nucleus<br />

and activation <strong>of</strong> the CD40 promoter. Further analysis with<br />

chromatin immunoprecipitation assay confirmed that leptin<br />

recruited STAT-1α, NF-kappaBp65, acetylated histone 4 and RNA<br />

polymerase II to the CD40 promoter in a time-dependent<br />

manner. Administration <strong>of</strong> leptin into normal mice significantly<br />

augmented LPS-induced CD40 expression in DC in the lymph<br />

nodes. Thus, this study identifies a novel function for leptin to<br />

act an adjuvant to modulate DC function and provides new<br />

insight into the new role <strong>of</strong> leptin in modulating inflammatory<br />

immune response.<br />

doi:10.1016/j.clim.2007.03.038<br />

OR.99 The prolyl isomerase Pin1 regulates ECM<br />

mRNA expression in primary lung fibroblasts<br />

Zhong-Jian Shen, PhD, University <strong>of</strong> Wisconsin Madison,<br />

Madison, WI, James Malter, Pr<strong>of</strong>essor, University <strong>of</strong><br />

Wisconsin Madison, Department <strong>of</strong> Pathology and<br />

Laboratory Medicine, Madison, WI<br />

A key pathological feature <strong>of</strong> asthma is excessive airway and<br />

parenchymal deposition <strong>of</strong> extracellular matrix (ECM) protein,<br />

possibly resulting from reduced catabolism or increased<br />

production or both. TGF-β1 playsacentralroleintheairway<br />

remodeling, mediating ECM gene expression by resident<br />

fibroblasts. The molecular mechanisms underlying this dynamic<br />

process remain poorly understood. Here, we examined the role<br />

<strong>of</strong> the peptidyl-prolyl isomerase (PPIase) Pin1 in ECM expression<br />

using lung fibroblasts from Pin1 knockout mice. Cells were<br />

starved before TGF-β1 treatment, and the level <strong>of</strong> ECM mRNA<br />

was measured by RT/qPCR. Treatment with TGF-β1 consistently<br />

increased the level <strong>of</strong> collagen I, III and V in wild type cells.<br />

Knockout cells opposed to TGF-β1 however were unable to<br />

upregulate collagen III mRNA. As matrix metalloproteinase<br />

(MMP)2and3aremajoris<strong>of</strong>ormsinthelung,whichdegrade<br />

collagen I/V and collagen III/V, respectively, the level <strong>of</strong> MMPs<br />

was also evaluated. In the presence <strong>of</strong> TGF-β1, the level <strong>of</strong><br />

MMP2 and 3 was considerably increased in Pin1 knockout cells.<br />

Among the three tissue inhibitor <strong>of</strong> matrix metalloproteinase<br />

(TIMP) is<strong>of</strong>orms (1, 2 and 3), TIMP1 levels were significantly<br />

reduced in Pin1 knockout cells despite TGF-β1 stimulation.<br />

Therefore, the decreased expression <strong>of</strong> type III collagen and<br />

TIMP1 along with increased MMP2 and 3 in Pin1 knockout<br />

fibroblasts support our previous finding that in vivo administration<br />

<strong>of</strong> Pin1 inhibitors reduced airway fibrosis and suggest that<br />

Pin1 may be a target for the treatment <strong>of</strong> asthma and other<br />

fibrotic disorders as well.<br />

doi:10.1016/j.clim.2007.03.039<br />

OR.100 Genetic Evidence for CD8 + T Cell<br />

Immunoregulation in Murine CD4 + T Cell Colitis<br />

Bo Wei, Assistant Pr<strong>of</strong>essor, Department Pathology and Lab<br />

Medicine, University <strong>of</strong> California, Los Angeles David Geffen<br />

School <strong>of</strong> Medicine, Los Angeles, CA, Michael Macpherson,<br />

Graduate Student, Department Pathology and Lab Medicine,<br />

University <strong>of</strong> California, Los Angeles David Geffen School <strong>of</strong><br />

Medicine, Los Angeles, CA, Daisuke Fujiwara, Postgraduate<br />

Researcher, Department Pathology and Lab Medicine, University<br />

<strong>of</strong> California, Los Angeles David Geffen School <strong>of</strong> Medicine, Los<br />

Angeles, CA, Sarah Brewer, Graduate Student, University <strong>of</strong><br />

California, Los Angeles David Geffen School <strong>of</strong> Medicine, Los<br />

Angeles, CA, Jonathan Braun, Pr<strong>of</strong>essor, Department Pathology<br />

and Lab Medicine, University <strong>of</strong> California, Los Angeles David<br />

Geffen School <strong>of</strong> Medicine, Los Angeles, CA<br />

Abnormal CD4 + Tcell activity and consequent tissue damage<br />

are major pathogenic features <strong>of</strong> inflammatory bowel disease<br />

(IBD) and many systemic and tissue restricted autoimmune<br />

diseases. Disease susceptibility may involve deficiency <strong>of</strong> normal<br />

mucosal immunoregulation that attenuates CD4 + T cell activity<br />

targeting the mucosa. CD8 + T cells have emerged as one such<br />

regulatory population, but their modes <strong>of</strong> induction and<br />

targeting are poorly understood. We have demonstrated that<br />

B cells attenuate immune colitis through the interaction with<br />

CD8 + Tcells.Inthisstudy,weevaluatedthegenetics<strong>of</strong>Bcell<br />

traits involved in immunoregulation <strong>of</strong> G±i2−/− Tcellsinduced<br />

colitis. Transfer <strong>of</strong> G±i2−/− Tcells from 4 to 5 weeks mice (prior<br />

to colitis onset) induced progressive colitis, with additional<br />

features <strong>of</strong> systemic immune inflammation. Mesenteric node B<br />

cells from wild type mice inhibited colitis and systemic<br />

inflammation, and blocked the attendant CD4 + Tcellexpansion<br />

expressing TNF- 2 ,IFN- 3 and IL-17. Surprisingly, the protective<br />

function <strong>of</strong> B cells did not require genetic sufficiency for MHC<br />

class II, indicating that they did not directly interact with<br />

colitigenic or regulatory CD4 + T cells. However, protection<br />

required both MHC class I and TAP1 sufficiency, indicating that<br />

they augmented immunoregulation through direct interaction<br />

with classical peptide-restricted CD8 + T cells. These findings<br />

provide new evidence for CD8 + T cell immunoregulation in<br />

colitis, and suggest that functional or numerical deficiencies <strong>of</strong><br />

cooperative B or CD8 + Tcell subpopulations may be susceptibility<br />

traits, and targets for compensatory therapy, in inflammatory<br />

bowel disease. Supported by NIH DK4673 and DK69434.<br />

doi:10.1016/j.clim.2007.03.040<br />

S141


S142 Abstracts<br />

OR.101 The Galectin-9/TIM-3 Pathway and Human<br />

Glioma Pathogenesis<br />

Chengjie Zheng, Student, Harvard University, Boston, MA,<br />

Juhi Kuchroo, Student, Brigham and Women’s Hospital and<br />

Harvard Medical School, Boston, MA, David E. Anderson,<br />

Instructor in Neurology, Brigham and Women's Hospital and<br />

Harvard Medical School, Boston, MA<br />

Approximately 15,000 patients die each year in the United<br />

States from GBMs. Two well-documented aspects <strong>of</strong> malignant<br />

gliomas are that they are highly invasive and that immunity<br />

directed against them is ineffective. We hypothesize that<br />

glioma expression <strong>of</strong> galectin-9 (Gal-9) regulates both <strong>of</strong> these<br />

aspects <strong>of</strong> glioma pathogenesis. Gal-9 expression by peripheral<br />

human tumors has a favorable clinical prognosis and limits<br />

tumor cell metastasis/invasiveness. Thus, one prediction is<br />

that down-modulation <strong>of</strong> Gal-9 in gliomas may contribute to<br />

tumor cell invasiveness. Indeed, using FACS to isolate purified<br />

tumor cells or reactive astrocytes from ex vivo tumor speci-<br />

mens and quantitative RT-PCR, we have found that GBM tumor<br />

cells express little/no Gal-9 whereas Gal-9 is readily detected<br />

in reactive astrocytes. Inhibition <strong>of</strong> TGF-&#946; signaling using<br />

soluble receptor or siRNA approaches indicates that TGF-<br />

&#946; is responsible for down-modulation <strong>of</strong> Gal-9, which<br />

provides a molecular explanation for the long-standing<br />

observation that levels <strong>of</strong> TGF-&#946; increase with grade <strong>of</strong><br />

malignancy and invasiveness. On the other hand, Gal-9 has<br />

recently been identified as a ligand <strong>of</strong> TIM-3 and been shown to<br />

kill IFN-&#947;-secreting Th1 cells. To this end, co-culture <strong>of</strong><br />

ex vivo CD4+ Tcells with a primary GBM cell line in the presence<br />

<strong>of</strong> blocking anti-TIM-3 monoclonal antibody enhances T cell<br />

proliferation and IFN-&#947; secretion. Collectively, the data<br />

suggest a paradigm in which Gal-9 expression by low-grade<br />

gliomas inactivates infiltrating TIM-3+ CD4+ and CD8+ T cells,<br />

and that with increasing stages <strong>of</strong> malignancy, down-modulation<br />

<strong>of</strong> Gal-9 promotes tumor cell invasiveness.<br />

doi:10.1016/j.clim.2007.03.042


Abstracts<br />

Su.1 Multiple Sclerosis Exacerbations:<br />

Simultaneous Activation <strong>of</strong> the Immune System by<br />

Multiple Immunogens<br />

Frederick Westall, President and Research Pr<strong>of</strong>essor,<br />

Institute for Disease Research, Temecula, CA<br />

Multiple Sclerosis (MS)is considered to be an autoimmune<br />

response against central nervous system myelin components.<br />

Immunogenic mimics on a surface protein <strong>of</strong> a microorganism<br />

initiate the immune process. Because <strong>of</strong> recent data indicating<br />

that such mimics are plentiful, it is suggested that in MS<br />

multiple mimics actually are simultaneously processed. Multiple<br />

simultaneous mimic processing can explain much <strong>of</strong> the<br />

confusing immunological data in MS and would parallel what is<br />

observed in chronic relapsing allergic encephalomyelitis, a<br />

clinical and histological model for MS.<br />

doi:10.1016/j.clim.2007.03.043<br />

Su.2 Comprehensive Molecular Explanation <strong>of</strong><br />

Recent MRI Multiple Sclerosis Studies<br />

Frederick Westall, President and Research Pr<strong>of</strong>essor,<br />

Institute for Disease Research, Temecula, CA<br />

Recent MRI studies have indicated that MS is a two part<br />

disease. First, diffuse lesions, apparently initiated by inflammatory<br />

processes, appear. Some <strong>of</strong> these then develop into the<br />

classical immunologically involved focal events. Inflammatory<br />

agents appear in the CSF and serum as much as a month before<br />

the focal lesions are seen. One <strong>of</strong> the earliest <strong>of</strong> these substances<br />

observed is metalloproteinase 9. This is followed by caspases and<br />

plasminogen and eventually various cytokines. Since there is no<br />

evidence <strong>of</strong> a chronic microbiological invasion <strong>of</strong> the CNS in MS,<br />

what is initiating this early inflammation? The immunological<br />

component <strong>of</strong> MS is thought to be caused by a microorganism<br />

possessing a protein sequence, which mimics an immunological<br />

active region in myelin. These mimics are not rare. Many<br />

potential mimics appear in the proteins <strong>of</strong> normal gut bacteria.<br />

In fact gut bacteria also contain the adjuvant molecules shown<br />

essential for autoimmune disease. The initiation <strong>of</strong> clinical<br />

autoimmune disease takes weeks. However, it has been clearly<br />

shown that the adjuvant molecules themselves can quickly<br />

inititate an inflammatory response. Many <strong>of</strong> these adjuvant<br />

molecules, for example lipopolysaccharides, can pass the gut<br />

barrier. An abnormal digestion in the gut could initiate the<br />

production <strong>of</strong> both immunogens and adjuvant molecules. These<br />

adjuvants could initially cause a general inflammatory response<br />

and later a specific autoimmune response in the CNS. These<br />

actions would explain the MRI studies, i.e. a diffuse inflammatory<br />

response with an occasional autoimmune involvement.<br />

doi:10.1016/j.clim.2007.03.044<br />

Poster Session<br />

Sunday, June 10<br />

7:30 am−7:00 pm<br />

Authors Present: 5:45 pm−7:00 pm<br />

Su.3 <strong>Oral</strong> Therapy with FTY720 Inhibits<br />

Angiogenesis in the Spinal Cord <strong>of</strong> Lewis Rats in the<br />

Relapse Phase <strong>of</strong> Experimental Autoimmune<br />

Encephalomyelitis<br />

Peter Hiestand, Senior Research Scientist, Novartis<br />

Institutes for BioMedical Research, Autoimmune Diseases<br />

and Transplantation, Basel, Switzerland, Christian Schnell,<br />

Senior Research Investigator, Novartis Institutes for<br />

BioMedical Research, Autoimmune Diseases and<br />

Transplantation, Basel, Switzerland<br />

Fingolimod (FTY720) is a sphingosine-1-phosphate receptor<br />

modulator that retards the egress <strong>of</strong> lymphocytes from<br />

peripheral lymph nodes. FTY720 has been shown to affect all<br />

stages <strong>of</strong> experimental autoimmune encephalomyelitis (EAE)<br />

in rats as well as in mice either induced with neuronal tissue<br />

or specific myelin protein antigens. While we have demonstrated<br />

that monocyte migration into the CNS <strong>of</strong> rats<br />

affected by EAE is blocked in the acute phase, the pronounced<br />

effects observed with FTY720 in the relapse phase<br />

<strong>of</strong> EAE seem to be linked to additional mechanisms. One <strong>of</strong><br />

the proposed factors involved in the induction <strong>of</strong> relapses <strong>of</strong><br />

EAE in the Lewis rats could be an increase in blood vessel<br />

density in the spinal cord as it has been described in EAE in<br />

guinea pigs. Using a casting procedure <strong>of</strong> blood vessels <strong>of</strong> the<br />

brain and spinal cord <strong>of</strong> Lewis rats going through acute and<br />

relapsing phases <strong>of</strong> EAE, we demonstrate an increase in<br />

vessel density induced in relapsing EAE and a FTY720mediated<br />

blockade <strong>of</strong> the induced angiogenesis. A similar<br />

protective effect was seen using a specific VEGF-receptor<br />

kinase inhibitor, demonstrating that a blockade <strong>of</strong> diseaseinduced<br />

angiogenesis leads to complete amelioration <strong>of</strong><br />

clinical symptoms <strong>of</strong> relapsing EAE.<br />

This neo-angiogenesis was mainly observed in the lumbar<br />

section <strong>of</strong> the spinal cord suggesting that this region be <strong>of</strong><br />

particular importance to the paralytic episodes observed in<br />

EAE in rats.<br />

doi:10.1016/j.clim.2007.03.045<br />

S143<br />

Su.5 Enhanced Glucocorticoid Signaling Impacts<br />

Thymocyte Apoptosis and Adaptive Immune<br />

Responses<br />

Fred Lühder, PhD, Institute for Multiple Sclerosis Research,<br />

Göttingen, Germany, Jens van den Brandt, PhD, Department<br />

<strong>of</strong> Cellular and Molecular <strong>Immunology</strong>, Göttingen, Germany,<br />

Holger Reichardt, PhD, Department <strong>of</strong> Cellular and<br />

Molecular <strong>Immunology</strong>, Göttingen, Germany, Ralf Gold, MD,<br />

St. Josef-Hospital/Ruhr-University Bochum, Bochum,<br />

Germany<br />

Glucocorticoids (GC) are synthesized by the adrenal gland<br />

and exert pleiotropic effects ranging from the regulation <strong>of</strong>


S144 Abstracts<br />

energy metabolism and the control <strong>of</strong> cognitive functions to the<br />

modulation <strong>of</strong> the immune system. Therapeutically they are<br />

used as potent anti-inflammatory and immunosuppressive drugs<br />

in a variety <strong>of</strong> conditions, but endogenous GCs rather exert<br />

positive as well as negative effects on the immune system. To<br />

study the effects <strong>of</strong> enhanced GC signaling on T cells we<br />

generated transgenic rats expressing a glucocorticoid receptor<br />

(GR) with increased ligand affinity. This causes massive<br />

thymocyte apoptosis resulting in a dramatic loss <strong>of</strong> thymic<br />

cellularity, which could be reverted by adrenalectomy. In the<br />

periphery, mature T lymphocytes accumulate that respond<br />

normally to costimulation and whose expansion is probably due<br />

to homeostatic proliferation, but have a skewed Tcell receptor<br />

repertoire. The transgenic rats are partially protected from<br />

Experimental Autoimmune Encephalomyelitis (EAE) induced by<br />

immunization with gpMBP, a predominantly TH1 mediated<br />

autoimmune model. The onset <strong>of</strong> EAE in transgenic rats was<br />

delayed by 5 days and the disease course was significantly<br />

milder. This difference appears to be the consequence <strong>of</strong> an<br />

impaired leukocyte infiltration into the CNS and a distinct<br />

cytokine pr<strong>of</strong>ile. In contrast, the ability <strong>of</strong> the transgenic rats to<br />

mount a predominantly TH2 mediated allergic airway response<br />

to ovalbumin was unaffected, although isotype switching <strong>of</strong><br />

antigen-specific immunoglobulins was altered. Collectively, our<br />

findings suggest that endogenous glucocorticoids impact T cell<br />

development and favor the selection <strong>of</strong> TH2 over TH1 dominated<br />

adaptive immune responses.<br />

doi:10.1016/j.clim.2007.03.046<br />

Su.6 Microarray Analysis Identifies<br />

Interferon-β-responsive Genes in Human Microglia<br />

Hiroko Tabunoki, Assistant, Meiji Pharmaceutical<br />

University, Bioinformatics, Kiyose, Japan, Tamako Misawa,<br />

Graduate Student, Meiji Pharmaceutical University,<br />

Bioinformatics, Kiyose, Japan, Jun-ichi Sato, Pr<strong>of</strong>essor,<br />

Meiji Pharmaceutical University, Bioinformatics, Kiyose,<br />

Japan<br />

Objective/Background: MS is mediated by autoreactive Th1<br />

cells. When activated Th1 cells enter the CNS across the blood–<br />

brain barrier (BBB), they are reactivated by microglia (MCG), the<br />

pr<strong>of</strong>essional antigen-presenting cell type in the CNS. Consequently,<br />

MCG activated by Th1 cytokines release mediators <strong>of</strong><br />

demyelination. Although interferon-beta (IFNB) reduces the<br />

frequency <strong>of</strong> relapses in MS patients, the molecular mechanism<br />

responsible for beneficial effects <strong>of</strong> IFNB in MS remains<br />

unknown. Because IFNB could pass through the BBB disrupted<br />

in active lesions <strong>of</strong> MS, it might directly modulate the biological<br />

function <strong>of</strong> MCG. Methods: To identify IFNB-responsive genes<br />

(IRGs) in human MCG, we studied the gene expression pr<strong>of</strong>ile <strong>of</strong> a<br />

human MCG cell line HMO6 (Neurobiol Dis 8:1057, 2001)<br />

following treatment with 50 ng/ml IFNB for 3 hours, by using<br />

an oligonucleotide microarray (Hitach), verified by Northern<br />

blot and real-time RT-PCR. Results: Among total 1606 genes on<br />

the array, IFNB elevated the expression <strong>of</strong> 39 genes. Top10<br />

included IFI56, IFI60, MX1, MX2, ISG15, IFI44, IRF7, STAT1, IFI6,<br />

and TAP1, all <strong>of</strong> which were known IRGs. The common upstream<br />

search <strong>of</strong> 39 genes on KeyMolnet (Curr Drug Discov Technol 2:89,<br />

2005), the knowledge-based data mining tool <strong>of</strong> bioinformatics,<br />

indicated a molecular network most relevant to gene regulation<br />

by IRF and NF-kB. Conclusions: IFNB induces the expression <strong>of</strong> a<br />

battery <strong>of</strong> IRGs in human microglia, including IRF7 and ISG15, a<br />

central regulator <strong>of</strong> innate and adaptive immunity (Nature Rev<br />

Immunol 6:644, 2006), suggesting an immunomodulatory effect<br />

<strong>of</strong> IFNB on human microglia.<br />

doi:10.1016/j.clim.2007.03.047<br />

Su.7 The Complement-scavenging Capacity <strong>of</strong> IVIG<br />

Products<br />

Martin O. Spycher, PhD, CSL Behring AG, Research and<br />

Development, Bern 22, Switzerland, Katja Matozan,<br />

University <strong>of</strong> Bern, Department <strong>of</strong> <strong>Clinical</strong> Research, Bern,<br />

Switzerland, Kathrin Minnig, PhD, CSL Behring AG, QA, Bern<br />

22, Switzerland, Sylvia Miescher, PhD, CSL Behring AG,<br />

Research and Development, Bern 22, Switzerland, Roland<br />

Zehnder, CSL Behring AG, Research and Development, Bern<br />

22, Switzerland, Liane Hoefferer, PhD, CSL Behring AG,<br />

Research and Development, Bern 22, Switzerland, Robert<br />

Rieben, PhD, University <strong>of</strong> Bern, Department <strong>of</strong> <strong>Clinical</strong><br />

Research, Bern, Switzerland<br />

IVIG induced complement scavenging describes one antiinflammatory<br />

effect <strong>of</strong> IVIG. This mechanism <strong>of</strong> action is<br />

considered important in inflammatory autoimmune disorders<br />

such as multifocal motor neuropathy or dematomyositis. The<br />

complement scavenging properties <strong>of</strong> eight IVIG products were<br />

compared. Constant amounts <strong>of</strong> aggregated human IgG coated<br />

to microtiterplates served as complement activators. They were<br />

incubated with constant amounts <strong>of</strong> human serum as a<br />

complementsourceinthepresenceorabsence<strong>of</strong>increasing<br />

concentrations <strong>of</strong> IVIGs. Complement activation was quantified<br />

by measuring bound activation products C3b/iC3b/C3c using a<br />

peroxidase-conjugated anti-C3c antibody.<br />

All IVIG tested exhibited dose-dependent inhibition <strong>of</strong> C3<br />

deposition. This effect was associated with reduced C3a<br />

generation, confirming that it was due to complement inhibition,<br />

not complement activation and consumption. Inhibition <strong>of</strong><br />

complement deposition was paralleled by an increase <strong>of</strong> C3<br />

activation product binding to IgG in the fluid phase. The extent<br />

<strong>of</strong> complement inhibition was dependent on the IVIG tested,<br />

with up to 3.5 fold differences between products. An IVIG<br />

currently in development, named IgPro10, manufactured with<br />

gentle procedures was the most potent product. Differences <strong>of</strong><br />

inhibition between products did not correlate with differences<br />

in C3 activation product binding to IgG in the fluid phase at given<br />

IVIG concentrations suggesting additional mechanisms being<br />

functional besides complement deviation. Complement scavenging<br />

is a common feature <strong>of</strong> IVIGs, however the extent <strong>of</strong><br />

complement scavenging depends on the product. IgPro10, a new<br />

IVIG product in development and manufactured with a gentle<br />

chromatographic process, is a more potent complement<br />

scavenger than currently used IVIGs.<br />

doi:10.1016/j.clim.2007.03.048<br />

Su.8 Influence <strong>of</strong> Interferon-β Therapy Switching on<br />

Neutralizing Antibody Titers: Results from the<br />

Austrian Switch Study (ASS)<br />

Florian Deisenhammer, Pr<strong>of</strong>essor, Department <strong>of</strong> Neurology,


Abstracts<br />

Innsbruck Medical University, Innsbruck, Austria, Alban<br />

Millonig, Assistant, Department <strong>of</strong> Neurology, Innsbruck<br />

Medical University, Innsbruck, Austria, Claudia Gneiss,<br />

Assistant, Department <strong>of</strong> Neurology, Innsbruck Medical<br />

University, Innsbruck, Austria<br />

Continuous treatment with interferon beta (IFNb) leads<br />

to neutralizing antibody (NAb) negativity in many previously<br />

NAb positive patients. To date no strategies are<br />

available to accelerate NAb reversion. We investigated the<br />

effect <strong>of</strong> switching between IFNb products on the development<br />

<strong>of</strong> NAb titers. Twenty-four NAb positive MS patients<br />

on IFNb-1a s.c. or IFNb-1b were included. At baseline IFNb<br />

treatment was interrupted for 3 months in all patients and<br />

2 infusions with 1 g methylprednisolone were given. Then<br />

patients were randomized either to their previous treatment<br />

or to IFNb-1a im. Twelve months later NAb titers were<br />

measured. Twelve patients were switched to IFNb-1a im<br />

30 μg (7 from IFNb-1a sc, 5 from IFNb-1b). Currently, final<br />

NAb titers (given in neutralizing units) are available in 18<br />

patients. The median baseline NAb titer was 846, 293 after<br />

corticosteroids, 1239 3 months after switching, and 393 at<br />

the end <strong>of</strong> study. The median change <strong>of</strong> NAb titers between<br />

baseline and end <strong>of</strong> study was −317 in patients who<br />

switched and −94 in patients who did not switch therapy<br />

(not significant). Baseline and end <strong>of</strong> study NAb titers<br />

correlated significantly (r=0.73). NAb can be regarded as a<br />

model <strong>of</strong> autoimmunity by breaking and re-inducing<br />

immune tolerance. The main influencing factor on the<br />

final NAb titer was the baseline titer. Many factors must be<br />

considered when choosing therapy for an individual patient.<br />

However, as far as NAb are concerned no strategy for<br />

reversion could be established leaving prevention the only<br />

option as yet.<br />

doi:10.1016/j.clim.2007.03.049<br />

Su.9 IL-21 Receptor Modulates Autoimmune<br />

Responses and Expression <strong>of</strong> Experimental<br />

Autoimmune Encephalomyelitis<br />

Ruolan Liu, Postdoctoral, Barrow Neurological Institute,<br />

Neurology, Phoenix, AZ, Antoni La Cava, Associate<br />

Pr<strong>of</strong>essor, David Geffen School <strong>of</strong> Medicine, Los Angeles,<br />

CA, Debbie Young, Pr<strong>of</strong>essor, Wyeth Research,<br />

Inflammation, Cambridge, MA, Mary Collins, Scientist,<br />

Wyeth Research, Cambridge, MA, Denise Campagnolo,<br />

Neurologist, Barrow Neurological Institute, Neurology,<br />

Phoenix, AZ, Timothy Vollmer, Neurologist, Barrow<br />

Neurological Institute, Neurology, Phoenix, MA, Fu-Dong<br />

Shi, Scientist, Barrow Neurological Institute, Neurology,<br />

Phoenix, AZ<br />

IL-21 receptor (IL-21R) consists <strong>of</strong> a unique IL-21R<br />

subunit and also shares the common gamma chain (γ c).<br />

IL-21R ligand IL-21 plays a plethora <strong>of</strong> roles in innate and<br />

adaptive immune responses. In this study we examined<br />

the influence <strong>of</strong> IL-21R deficiency in the development <strong>of</strong><br />

myelin oligodendrocyte glycoprotein peptide 35–55<br />

(MOG35–55)-induced experimental autoimmune encephalomyelitis<br />

(EAE), an animal model for human multiple<br />

sclerosis (MS). Upon immunization, IL-21R−/− mice devel-<br />

oped an exacerbated form <strong>of</strong> EAE with marked inflammatory<br />

infiltration into the central nervous system (CNS).<br />

Both Th1 and Th2 responses to MOG were augmented in<br />

IL-21R−/− mice. Although IL-21R deficiency did not<br />

dramatically alter the numbers <strong>of</strong> NK cells, NKT cells,<br />

CD11c, CD11b cells, and CD4+CD25+ regulatory T cells<br />

(Treg) during EAE, the expression <strong>of</strong> Foxp3 on Treg cells<br />

was reduced in MOG-primed IL-21R−/− mice. Our results<br />

suggest that IL-21R/ligand pathways may affect Treg cells,<br />

which may subsequently affect the magnitude <strong>of</strong> CNS<br />

autoimmunity.<br />

doi:10.1016/j.clim.2007.03.050<br />

S145<br />

Su.10 Targeted Delivery <strong>of</strong> Immunomodulating<br />

Agents to Dendritic Cells for Treatment <strong>of</strong><br />

Autoimmune Disease Using Biodegradable<br />

Microspheres<br />

Tali Brunner, PhD Student, Biotechnology Engineering,<br />

Ben-Gurion University <strong>of</strong> the Negev, Beer Sheva, Israel,<br />

Alon Monsonego, Researcher, Department <strong>of</strong> Microbiology<br />

and <strong>Immunology</strong>, Faculty <strong>of</strong> Health Sciences and The<br />

National Institute, Beer Sheva, Israel, Smadar Cohen,<br />

Researcher, Department <strong>of</strong> Biotechnology Engineering,<br />

Ben Gurion University <strong>of</strong> the Negev,<br />

Beer Sheva, Israel<br />

Background: Dendritic cells (DCs) are a central element<br />

for drug targeting in autoimmune diseases, due to their<br />

function in the onset and progression <strong>of</strong> destructive<br />

autoimmunity and in maintaining the Treg repertoire.<br />

When injected intradermally, microspheres sized 1–10<br />

microns are preferentially uptaken by DCs. Use <strong>of</strong> such<br />

microspheres has been investigated for delivery <strong>of</strong> antigens<br />

and DNA to DCs for vaccination; however this delivery<br />

system has not been implemented for delivery <strong>of</strong> siRNAs<br />

capable <strong>of</strong> regulating autoimmunity. In vivo delivery <strong>of</strong><br />

siRNA is a great challenge, since these RNA duplexes are<br />

rapidly degraded and require appropriate stabilization.<br />

Goal: To use biodegradable microspheres for delivering<br />

siRNA and other immunomodulating agents to DCs, for<br />

treatment <strong>of</strong> autoimmune disease. Results: Microspheres<br />

composed <strong>of</strong> poly-lactic-acid better target CD11c positive<br />

cells in a bone marrow DC (BMDC) culture, in comparison to<br />

a poly-lactic-co-glycolic composition. siRNA, stabilized<br />

using poly-ethylene-imine and encapsulated in microspheres<br />

using a double-emulsion procedure, remained<br />

active after release from microspheres in BMDCs. Importantly,<br />

the microspheres did not have an adjuvant effect<br />

upon the BMDCs compared with LPS-induced DC-mediated<br />

T cell proliferation, meaning that the BMDCs maintained<br />

their immature state, which is crucial for creating an<br />

immunoregulatory environment which can ameliorate the<br />

course <strong>of</strong> autoimmune disease.<br />

Summary: This work combines biotechnology engineering,<br />

assessing microsphere behavior and distribution, with<br />

basic immunological research, assessing activity <strong>of</strong> immunomodulating<br />

agents. We believe that this delivery platform<br />

can be used for delivering not only various targeting<br />

siRNAs, but also other immunomodulating agents such as


S146 Abstracts<br />

peptides which can interfere with intracellular signaling<br />

pathways.<br />

doi:10.1016/j.clim.2007.03.051<br />

Su.11 Neur<strong>of</strong>ascin-specific Autoantibodies Mediate<br />

Axonal Injury in Inflammatory Demyelinating<br />

Diseases <strong>of</strong> the Central Nervous System<br />

Christopher Linington, Pr<strong>of</strong>essor <strong>of</strong> Immunobiology,<br />

University <strong>of</strong> Aberdeen, Aberdeen, UK, Emily Mathey,<br />

Research Associate, University <strong>of</strong> Aberdeen, Aberdeen, UK,<br />

Tobias Derfuss, Max Planck Institute for Neurobiology,<br />

Martinsried, Germany, Matthew Rasband, Assistant<br />

Pr<strong>of</strong>essor, University <strong>of</strong> Connecticut Health Center,<br />

Farmington, CT, Maria Storch, Pr<strong>of</strong>essor, Medical University<br />

Graz, General Neurology, Graz, Germany, Reinhard<br />

Hohlfeld, Pr<strong>of</strong>essor, LMU University Hospital, <strong>Clinical</strong><br />

Neuroimmunology, Munich, Germany, Edgar Meinl,<br />

Pr<strong>of</strong>essor, Max Planck Institute for Neurobiology,<br />

Martinsried, Germany<br />

We report that multiple sclerosis is associated with elevated<br />

autoantibody responses to neur<strong>of</strong>ascin, as compared to other<br />

inflammatory neurological diseases. The two is<strong>of</strong>orms <strong>of</strong><br />

neur<strong>of</strong>ascin play essential roles in maintaining the molecular<br />

organization <strong>of</strong> myelinated fibers: NF186 is a neuronal protein<br />

located at the node <strong>of</strong> Ranvier and axonal initial segment where<br />

it interacts with voltage gated sodium channels, whilst NF155 is<br />

an oligodendroglial product sequestered at junctional complexes<br />

formed where paranodal loops <strong>of</strong> the myelin sheath<br />

contact the axonal surface. Analysis <strong>of</strong> neur<strong>of</strong>ascin-specific<br />

autoantibodies immunopurified from seropositive donors<br />

demonstrates that they recognise the extracellular domain <strong>of</strong><br />

both is<strong>of</strong>orms <strong>of</strong> the native protein indicating that this response<br />

may participate in disease pathogenesis. We investigated this<br />

hypothesis in experimental autoimmune encephalomyelitis<br />

using a pan NF155/186 mouse mAb to mimic the human<br />

autoantibody response. Passive transfer (i.p.) <strong>of</strong> mAb induced<br />

a rapid exacerbation <strong>of</strong> disease that was associated with a<br />

corresponding increase in acute axonal injury. Intriguing this<br />

occurred in the absence <strong>of</strong> any concomitant increase in the local<br />

inflammatory response or demyelination. Immun<strong>of</strong>luorescence<br />

microscopy revealed that binding <strong>of</strong> the transferred neur<strong>of</strong>ascin-specific<br />

antibody was restricted nodes <strong>of</strong> Ranvier where it<br />

co-localised with voltage gated sodium channels. These results<br />

identify NF186 as a primary target for neur<strong>of</strong>ascin-specific<br />

autoantibodies and indicate antibody-dependent effector<br />

mechanisms are involved in the pathogenesis <strong>of</strong> axonal injury<br />

andlossinmultiplesclerosis.<br />

doi:10.1016/j.clim.2007.03.052<br />

Su.12 CD11c on NK Cells Mirrors the Temporal<br />

Disease Activity <strong>of</strong> Multiple Sclerosis<br />

Toshimasa Aranami, Section Chief, Department <strong>of</strong><br />

<strong>Immunology</strong>, National Institute <strong>of</strong> Neuroscience, NCNP,<br />

Kodaira, Japan, Sachiko Miyake, Section Chief, Department<br />

<strong>of</strong> <strong>Immunology</strong>, National Institute <strong>of</strong> Neuroscience, NCNP,<br />

Kodaira, Japan, Masafumi Ogawa, Section Chief,<br />

Department <strong>of</strong> Neurology, National Center Hospital for<br />

Mental Nervous and Muscular Disorders, NCNP, Kodaira,<br />

Japan, Takashi Yamamura, Director, Department <strong>of</strong><br />

<strong>Immunology</strong>, National Institute <strong>of</strong> Neuroscience, NCNP,<br />

Kodaira, Japan<br />

Multiple sclerosis (MS) is an autoimmune disease, showing a<br />

greatdegree<strong>of</strong>varianceindiseaseactivity.Wehaverecently<br />

demonstrated that NK cells biased for producing IL-5 (bias<br />

towards type2 NK cells, NK2 bias) are associated with remission<br />

state <strong>of</strong> MS. Here we report that upregulation <strong>of</strong> CD11c on NK<br />

cells would characterize a subgroup <strong>of</strong> MS in remission whose NK<br />

cells are lacking NK2 bias. When we compared this group<br />

(CD11chigh) with the other group <strong>of</strong> patients (CD11clow)<br />

concerning NK cell expression <strong>of</strong> IL-5 and GATA-3, we found<br />

NK2 bias to be a selective character <strong>of</strong> CD11clow patients. In<br />

contrast, CD11chigh group showed an obvious trend for a larger<br />

proportion <strong>of</strong> HLA-DR+ cells within NK cells. Since NK cell<br />

stimulatory cytokine such as IL-15 was found to upregulate<br />

CD11c expression on NK cells in vitro, we postulate that<br />

inflammatory cytokine milieu may play a role in inducing<br />

CD11chigh NK cell phenotype. Although there was no clinical<br />

difference between CD11chigh and CD11clow at blood sampling,<br />

CD11chigh MS showed significantly higher relapsing rate than<br />

CD11clow during 120 days follow-up. These data point to an<br />

interpretation that CD11chigh patients are at a higher risk for<br />

developing relapse, which is probably due to the loss <strong>of</strong> NK2<br />

phenotype. Thus, CD11c on NK cells mirrors the temporal<br />

disease activity <strong>of</strong> MS and may be used as a disease biomarker.<br />

doi:10.1016/j.clim.2007.03.053<br />

Su.13 Functional Involvement <strong>of</strong> NR4A2 in the<br />

Pathogenesis <strong>of</strong> Multiple Sclerosis<br />

Yoshimitsu Doi, Physician, Department <strong>of</strong> <strong>Immunology</strong>,<br />

National Institute <strong>of</strong> Neuroscience, NCNP, Kodaira City,<br />

Tokyo, Japan, Shinji Oki, Scientist, Department <strong>of</strong><br />

<strong>Immunology</strong>, National Institute <strong>of</strong> Neuroscience, NCNP,<br />

Kodaira City, Tokyo, Japan, Sachiko Miyake, Physician,<br />

Department <strong>of</strong> <strong>Immunology</strong>, National Institute <strong>of</strong><br />

Neuroscience, NCNP, Kodaira City, Tokyo, Japan, Takashi<br />

Yamamura, Physician, Department <strong>of</strong> <strong>Immunology</strong>, National<br />

Institute <strong>of</strong> Neuroscience, NCNP, Kodaira City, Tokyo, Japan<br />

Objective: To study a role <strong>of</strong> the NR4A2 in T cells <strong>of</strong><br />

multiple sclerosis (MS). Background: NR4A2, a transcription<br />

factor <strong>of</strong> the steroid/thyroid receptor superfamily, is rapidly<br />

induced by exposure to growth factors and cytokines,<br />

suggesting a biological relevance to various cell functions.<br />

Through a cDNA microarray analysis, higher expression <strong>of</strong><br />

NR4A2 was observed in CD3+ T cells <strong>of</strong> 57 MS patients<br />

compared with 19 healthy control subjects (Satoh et al.<br />

Neurobiol Dis 18:537–50, 2005). However, the functional<br />

involvement <strong>of</strong> NR4A2 expression and its transcriptional<br />

targets in MS Tcells remain unknown. Methods: Expression <strong>of</strong><br />

NR4A2 mRNA was analyzed by quantitative RT-PCR in PBMCderived<br />

human T cells or murine T cells infiltrated in CNS<br />

after EAE induction. Activities <strong>of</strong> cytokine gene promoters<br />

were analyzed by reporter gene analysis. Cytokine production<br />

in primary T cells was measured after retroviral<br />

transduction <strong>of</strong> NR4A2 gene or silencing <strong>of</strong> NR4A2 gene by<br />

using siRNA. Results: Higher expression <strong>of</strong> NR4A2 was


Abstracts<br />

demonstrated both in MS Tcells and in CNS-infiltrating Tcells<br />

in EAE mice. NR4A2 augmented promoter activities <strong>of</strong><br />

inflammatory cytokine genes. Overexpression <strong>of</strong> NR4A2<br />

induced up-regulation <strong>of</strong> IL-17 and IFN-f× production. Meanwhile,<br />

silencing <strong>of</strong> NR4A2 expression by siRNA resulted in<br />

down-regulation <strong>of</strong> the inflammatory cytokines. Conclusions:<br />

NR4A2 was upregulated in T cells <strong>of</strong> MS patients and in CNSinfiltrating<br />

T cells <strong>of</strong> EAE mice. Inflammatory cytokines<br />

including IL-17 and IFN-f× were the possible targets <strong>of</strong> NR4A2<br />

in T cells, suggesting that NR4A2 may play an important role<br />

in immunopathogenesis <strong>of</strong> MS.<br />

doi:10.1016/j.clim.2007.03.054<br />

Su.14 Functional Analysis <strong>of</strong> Carcinoembryonic<br />

Antigen-related Cellular Adhesion Molecule 1 in<br />

Experimental Autoimmune Encephalomyelitis<br />

Shinji Oki, Section Chief, Department <strong>of</strong> <strong>Immunology</strong>,<br />

National Institute <strong>of</strong> Neuroscience, NCNP, Tokyo, Japan,<br />

Mayumi Fujita, Postdoctoral Researcher, Department <strong>of</strong><br />

<strong>Immunology</strong>, National Institute <strong>of</strong> Neuroscience, NCNP,<br />

Tokyo, Japan, Takao Ootsuka, Graduate Student, Department<br />

<strong>of</strong> <strong>Immunology</strong>, National Institute <strong>of</strong> Neuroscience, NCNP,<br />

Tokyo, Japan, Chiharu Tomi, Researcher, Department <strong>of</strong><br />

<strong>Immunology</strong>, National Institute <strong>of</strong> Neuroscience, NCNP,<br />

Tokyo, Japan, Miho Mizu, Researcher, Department <strong>of</strong><br />

<strong>Immunology</strong>, National Institute <strong>of</strong> Neuroscience, NCNP,<br />

Tokyo, Japan, Shinjiro Kaieda, Graduate Student,<br />

Department <strong>of</strong> <strong>Immunology</strong>, National Institute <strong>of</strong><br />

Neuroscience, NCNP, Tokyo, Japan, Takashi Yamamura,<br />

Director, Department <strong>of</strong> <strong>Immunology</strong>, National Institute <strong>of</strong><br />

Neuroscience, NCNP, Tokyo, Japan, Sachiko Miyake, Section<br />

Chief, Department <strong>of</strong> <strong>Immunology</strong>, National Institute <strong>of</strong><br />

Neuroscience, NCNP, Tokyo, Japan<br />

Introduction: Carcinoembryonic antigen-related cellular<br />

adhesion molecule 1 (CEACAM1) is one <strong>of</strong> the CEA-family<br />

members and is demonstrated to play an important role in<br />

immune regulation. In this study, we investigated the role <strong>of</strong><br />

CEACAM1 in experimental autoimmune encephalomyelitis<br />

(EAE). [Methods] C57BL/6J mice immunized with MOG peptide<br />

were treated with anti-CEACAM1 mAb (AgB10) or CEACAM1-Fc<br />

fusion protein twice a week. The clinical signs <strong>of</strong> each group<br />

were scored to evaluate the effect <strong>of</strong> CEACAM1-mediated<br />

signals in EAE. MOG-specific cytokine production by inguinal<br />

lymph node cells derived from AgB10-treated or CEACAM1-Fctreated<br />

mice were analyzed by ELISA 10 days after the<br />

immunization. MOG-specific antibody production in serum<br />

was analyzed for AgB10-treated mice. Results and Discussion:<br />

The repetitive treatment with AgB10 enhanced the clinical sign<br />

<strong>of</strong> EAE, whereas the treatment with CEACAM1-Fc suppressed<br />

EAE as compared with control animals, indicating that CEACAM1<br />

exerts suppressive signal for the development <strong>of</strong> EAE. The<br />

production <strong>of</strong> inflammatory cytokines such as IFN-fÁ andIL-17<br />

in T cells derived from AgB10-treated mice was significantly<br />

enhanced, whereas the production <strong>of</strong> these cytokines from<br />

CEACAM1-Fc-treated mice was significantly decreased. These<br />

results suggest that CEACAM1-mediated signal is inhibitory for<br />

MOG-specific Th1 or Th17 responses. Accordingly, MOG-specific<br />

antibody production from AgB10-treated mice showed the<br />

reduced level <strong>of</strong> IgG1 and enhanced level <strong>of</strong> IgG2a compared<br />

with control mice. Taken together, CEACAM1 signal holds an<br />

important suppressive role in EAE by inhibiting both Th1 and<br />

Th17 responses to tissue-specific antigens. Mechanistic analysis<br />

for the role <strong>of</strong> CEACAM1 signal is currently under investigation.<br />

doi:10.1016/j.clim.2007.03.055<br />

Su.15 Gene Expression Pr<strong>of</strong>ile Modulated by IVIG in<br />

Healthy Donors and Multiple Sclerosis Patients<br />

Christian Jacobi, MD, University <strong>of</strong> Heidelberg, Department<br />

<strong>of</strong> Neurology, Institute <strong>of</strong> <strong>Immunology</strong>, Heidelberg,<br />

Germany, Jürgen, Römisch, PhD, Octapharma PPmbH,<br />

Vienna, Austria, Stefan, Meuer, MD, University <strong>of</strong><br />

Heidelberg, Institute <strong>of</strong> <strong>Immunology</strong>, Heidelberg, Germany,<br />

Thomas Giese, MD, University <strong>of</strong> Heidelberg, Institute <strong>of</strong><br />

<strong>Immunology</strong>, Heidelberg, Germany<br />

Intravenous immunoglobulin G (IVIG) has become an<br />

established first- and second-line line treatment in a<br />

number <strong>of</strong> immune mediated diseases during recent years.<br />

IVIG is now proposed to modulate a wide range <strong>of</strong> molecules<br />

(e.g. bacterial/viral antigens, toxins, Fc receptors, complement,<br />

autoantibodies), to act on different cell types (e.<br />

g. B cells, T cells, macrophages, dendritic cells, endothelial<br />

cells), and to modulate various immunological pathways at<br />

both the humoral and cellular levels including inflammation,<br />

antigen presentation, cell growth and apoptosis. Using<br />

a variety <strong>of</strong> specific assays many distinct effects <strong>of</strong> IVIG<br />

have been demonstrated in vitro. To provide further insight<br />

into the mechanisms involved in IVIG-dependent immunmodulation<br />

under physiological conditions, we have established<br />

a human whole blood gene expression assay in vitro.<br />

There, we analyzed the effect <strong>of</strong> IVIG on CD2-, CD3-, LPSand<br />

PMA/Ionomycin stimulated leukocytes in whole blood <strong>of</strong><br />

20 healthy donors and 12 untreated MS patients. IVIG<br />

induces among others the expression and release <strong>of</strong><br />

Interferon-γ, MCP1 and IP10. Interestingly, IVIG reduces<br />

the MCP1 and IP10 expression and release upon LPS<br />

stimulation, whereas Interferon-γ expression is further<br />

enhanced. Since both chemokines are physiologically<br />

induced by Interferon, this effect <strong>of</strong> IVIG is unanticipated<br />

and deserves further investigation for its possible role in<br />

anti-inflammatory properties <strong>of</strong> IVIG. No significant differences<br />

in the IVIG induced expression pr<strong>of</strong>ile between<br />

healthy donors and MS patients could be observed.<br />

doi:10.1016/j.clim.2007.03.056<br />

S147<br />

Su.16 IL-15 and IL-15Rα Expressed in Human<br />

Central Nervous System by Astrocytes Contribute to<br />

CD8 T Lymphocyte Activation and Persistence:<br />

Implications for Multiple Sclerosis<br />

Nathalie Arbour, Assistant Pr<strong>of</strong>essor, University <strong>of</strong><br />

Montreal-Medicine, Montreal, QC, Canada, Philippe Saikali,<br />

PhD Student, McGill University-Neurology, Montreal, QC,<br />

Canada, Jack Antel, Pr<strong>of</strong>essor<br />

Increasing experimental evidences suggest that CD8 T<br />

lymphocytes partake in multiple sclerosis (MS) related central<br />

nervous system (CNS) damage. CD8 T lymphocytes are


S148 Abstracts<br />

prominent cells in MS brain infiltrates and could injure<br />

oligodendrocytes and axons, both damaged in MS, as they are<br />

observed touching these structures. Activated memory CD8 T<br />

cells were shown to clonally expand and persist in the CNS <strong>of</strong> MS<br />

patients for years. Interleukin-15 (IL-15) is pivotal in the<br />

generation and maintenance <strong>of</strong> memory CD8 T cells. IL-15<br />

bound to IL-15Rα onitscell<strong>of</strong>originefficientlyactivatesIL-<br />

15Rβ/γ bearing cells. We determined whether a local source <strong>of</strong><br />

IL-15 and IL-15Rα can supply this cytokine to infiltrating CD8 T<br />

lymphocytes in the MS CNS. Human astrocytes expressed surface<br />

bound IL-15 and IL-15Rα in vitro; such expression was upregulated<br />

in response to pro-inflammatory cytokines. We<br />

detected astrocytes expressing IL-15 in MS brain supporting its<br />

in vivo presence. Human CD8 T cells cultured in the presence<br />

<strong>of</strong> IL-15 were more activated and cytotoxic (high amounts <strong>of</strong><br />

cytolytic enzymes) and acquired a memory phenotype that was<br />

not observed in the presence <strong>of</strong> IL-2. CD8 T lymphocytes bearing<br />

a similar activated memory phenotype were observed in the<br />

cerebrospinal fluid <strong>of</strong> MS patients. The localization <strong>of</strong> astrocytes<br />

within the brain architecture suggests that infiltrating CD8 T<br />

cells would readily encounter this source <strong>of</strong> IL-15+IL15Rα.<br />

These results underline a novel potential role for IL-15/IL-15Rα<br />

in the survival <strong>of</strong> memory CD8 T cells in the CNS during<br />

inflammatory diseases such as MS.<br />

doi:10.1016/j.clim.2007.03.057<br />

Su.18 CD4+CD28null T Cells in Autoimmune Disease:<br />

Pathogenic Features and Decreased Susceptibility to<br />

Immunoregulation<br />

Marielle Thewissen, Hasselt University, Biomedical Research<br />

Institute, Diepenbeek, Belgium, Veerle Somers, Hasselt<br />

University, Biomedical Research Institute, Diepenbeek,<br />

Belgium, Niels Hellings, Hasselt University, Biomedical<br />

Research Institute, Diepenbeek, Belgium, Piet Stinissen,<br />

Director, Hasselt University, Diepenbeek, Belgium, Koen<br />

Venken, Hasselt University, Biomedical Research Institute,<br />

Diepenbeek, Belgium<br />

CD4+CD28null T cells have been described to be<br />

generated as a consequence <strong>of</strong> persistent immune stimulation.<br />

These CD4+ T cells are incapable <strong>of</strong> providing help for<br />

B cell proliferation and antibody production, but have<br />

acquired cytolytic function. To date, the triggers <strong>of</strong><br />

activation <strong>of</strong> CD4+CD28null T cells have not been fully<br />

elucidated. An expansion <strong>of</strong> this cell subset has been<br />

described in several pathological conditions including<br />

multiple sclerosis (MS) and rheumatoid arthritis (RA). In<br />

this study the role <strong>of</strong> the expanded CD4+CD28null T cell<br />

subset in RA and MS pathology was further investigated.<br />

CD4+CD28null T cells were found to be terminally differentiated<br />

effector memory cells. They are equipped to<br />

infiltrate tissues and could be detected in the synovial<br />

tissue <strong>of</strong> RA patients. Whereas no reactivity to candidate<br />

autoantigens (myelin, collagen) was observed within the<br />

CD4+CD28null T cell subset, cytomegalovirus (CMV) reactivity<br />

was prominent in 4/4 HC, 4/4 RA patients and 3/4<br />

MS patients. Of note was that the intensity <strong>of</strong> the CMV<br />

induced proliferative response was related to the number<br />

<strong>of</strong> CMV epitopes recognized. Interestingly, our results<br />

illustrated that CD4+CD28null T cells were not susceptible<br />

to the suppressive actions <strong>of</strong> CD4+CD25 high regulatory T<br />

cells. In conclusion, this study provides several indications<br />

for a role <strong>of</strong> CD4+CD28null T cells in autoimmune pathology.<br />

These cells display pathogenic features, fill up immunological<br />

space and are less vulnerable to regulatory mechanisms.<br />

However, our results do not support a direct<br />

autoreactive role <strong>of</strong> CD4+CD28null T cells in the pathogenesis<br />

<strong>of</strong> autoimmune diseases.<br />

doi:10.1016/j.clim.2007.03.058<br />

Su.19 Intravenous TCV-CD4 Treatment for Multiple<br />

Sclerosis<br />

Gustavo A. Moviglia, MD, Regina Mater Foundation and<br />

Maimonides University, Buenos Aires, Argentina, Gabriela<br />

Varela, PhD, Regina Mater Foundation, Buenos Aires,<br />

Argentina, J. Hirsch, MD, Regina Mater Foundation, Buenos<br />

Aires, Argentina, J.A. Brizuela, MD, Regina Mater<br />

Foundation and Maimonides University, Buenos Aires,<br />

Argentina, C.A. Palleros, Regina Mater Foundation, Buenos<br />

Aires, Argentina, R. Moya, MD, Regina Mater Foundation,<br />

Buenos Aires, Argentina, Fabiana Bastos, MD, Regina Mater<br />

Foundation and Maimonides University, Buenos Aires,<br />

Argentina, M.T. Moviglia, MD, Regina Mater Foundation and<br />

Maimonides University, Buenos Aires, Argentina<br />

Background: T cell vaccine (TCV) is a proven pre clinical and<br />

clinical cell therapy for MS. The original technique, developed<br />

by Irum Cohen and collaborators, was as monoclonal or<br />

oligoclonal antimyelin and selective cell suspension to be<br />

activated, irradiated and administered to MS stable patients.<br />

Recent clinical trials showed that 92% <strong>of</strong> treated patients were<br />

stabilized. Methods: 82 CPMS patients were divided into 3<br />

groups: Group 1 (42 patients) received 10 subcutaneous<br />

immunizations <strong>of</strong> polyclonal TCV (1 every 4 weeks). Group 2<br />

(24 patients) received 10 IV polyclonal TCV. Group 3 (16<br />

patients) received 10 IV polyclonal TCV-CD4. Experienced<br />

neurologist evaluated the EDSS index from the beginning up to<br />

3 years <strong>of</strong> follow-up. The TCV-CD4 vaccine was prepared<br />

selecting only CD4+ cells from polyclonal TCV (Stem Sep for<br />

CD4, Vancouver) Results: Group 1, 19% <strong>of</strong> patients improved an<br />

average <strong>of</strong> 1 point <strong>of</strong> original EDSS index, 65% remained stable<br />

and 16% deteriorated only 0.5 points. 83% developed local<br />

aseptic abscesses at immunization site that took 1–6monthsto<br />

be solved. Group 2, 50% improved 2 points; 29% improved 1 point<br />

and 21% remained stable. No abscesses on the 24 patients but all<br />

developed fever (39.5 C) during first 24 hours after immunization.<br />

Group 3, 69% improved 2 points, 25%, 1 point and 6%<br />

remained stable. No fever nor abscesses observed. Conclusion:<br />

IV TCV-CD4 treatment is safe and shows therapeutic efficacy for<br />

CPMS. The spleen seems to be where antiergotype regulator<br />

cells are produced.<br />

doi:10.1016/j.clim.2007.03.059<br />

Su.20 Ingested (<strong>Oral</strong>) Soluble Immune Response<br />

Suppressor (SIRS) Peptide 1−21 Inhibits Acute EAE<br />

Staley Brod, Pr<strong>of</strong>essor, University <strong>of</strong> Texas Houston, MSRG,<br />

Houston, TX, Zach Hood, Technician, University <strong>of</strong> Texas<br />

Houston, MSRG, Houston, TX


Abstracts<br />

Background: Ingested type I IFN inhibits clinical attacks,<br />

relapses and inflammation in murine EAE. Type I IFN activate<br />

human suppressor T cells that produce SIRS. Objectives: We<br />

examined whether SIRS peptide would have similar effects to<br />

ingested IFN-α in EAE. Methods: C57BL/6 mice were actively<br />

immunized with myelin oligodendroglial (MOG) peptide 35–<br />

55 (MEVGWYRSPFSRVVHLYRNGK) in IFA and followed for<br />

disease. <strong>Clinical</strong> severity was graded daily. For parenteral<br />

treatment, B6 mice were injected with control saline<br />

(mock), 0.1, 1, or 10 mg SIRS peptide 1–21 (NH–MET–Thr–<br />

Glu–Glu–Asn–Gln–Gln–Ser–Ser–Gln–Pro–Lys–Thr–Thr–Ile–<br />

Asn–Asn–Ala–Gly–Asp–Ser–CysOH). For oral treatment, B6<br />

mice were fed with 0.1 ml <strong>of</strong> saline (mock) or 1, 10 or 100 mg<br />

SIRS peptide 1– 21 daily. Following sacrifice, spinal cords<br />

were evaluated for foci <strong>of</strong> inflammation. Splenocytes from<br />

grouped saline (mock) fed or 100 mcg SIRS peptide fed mice<br />

were stimulated with Con A and examined using mouse<br />

cytokine inflammatory antibody array. Results: S.C. 10 mcg<br />

SIRS peptide 1–21 showed significant inhibition <strong>of</strong> EAE<br />

(pb0.001). Ingested SIRS peptide at 10 and 100 mcg SIRS<br />

peptide inhibited EAE compared to placebo (pb0.001).<br />

There were significantly less inflammatory foci in the SIRS<br />

peptide fed group compared to the control mock saline group<br />

(pb0.03). Splenocytes from SIRS peptide 1–21 fed mice<br />

showed increased production <strong>of</strong> CD30L, IL-6, IL-13, I-TAC,<br />

TCA-3/CCL1, TNF-α. Conclusion: Ingested SIRS peptide<br />

inhibits both clinical EAE and inflammation via counterregulatory<br />

type 2-like cytokines and chemokines IL-13,<br />

CD30L and TCA-3.<br />

doi:10.1016/j.clim.2007.03.060<br />

Su.21 Identifying New Immunodominant Myelin<br />

Peptides in Relapsing Remitting Multiple Sclerosis<br />

Patients<br />

Brian Newsom, Senior Director <strong>of</strong> Project Development,<br />

Opexa Therapeutics, Inc. The Woodlands, TX, Kathrin von<br />

Gynz Rekowski, Research Scientist, Opexa Therapeutics,<br />

Inc. The Woodlands, TX, Mitzi Montgomery, Consultant,<br />

Opexa Therapeutics, Inc. The Woodlands, TX, Jim Williams,<br />

Chief Operating Officer, Opexa Therapeutics, The<br />

Woodlands, TX, Edward Fox, <strong>Clinical</strong> Assistant Pr<strong>of</strong>essor,<br />

University <strong>of</strong> Texas Medical Branch, MS Clinic <strong>of</strong> Central<br />

Texas, Round Rock, TX<br />

T cell reactivity to peptides found to be instrumental in<br />

Multiple Sclerosis (MS) pathology and experimental animal<br />

models has centered on the major myelin proteins; Myelin<br />

Basic Protein (MBP), Myelin Proteolipid Protein (PLP), and<br />

Myelin Oligodendrocyte Glycoprotein (MOG). With recent<br />

structural studies on these proteins and discoveries <strong>of</strong><br />

additional myelin proteins, there are questions about<br />

potential interactions between these proteins and T cells in<br />

MS. Using synthetic peptides <strong>of</strong> 16 amino acid residues and<br />

overlaps <strong>of</strong> 12 residues, we have surveyed the resulting 163<br />

peptides from these three proteins for their proliferative<br />

reactivity in 16 healthy subjects and 63 MS patients,<br />

including 22 from our Phase I trials <strong>of</strong> the T cell vaccine<br />

Tovaxin. Using a Tcell Epitope Analysis Assay (EAA), peptides<br />

were screened using peripheral blood mononuclear cells and<br />

a tritiated thymidine incorporation assay. The results from<br />

this assay generated a pattern <strong>of</strong> individual, patient specific<br />

reactivity by calculating the stimulation index <strong>of</strong> T cells in<br />

the presence <strong>of</strong> peptide antigens when compared to media<br />

controls. Each assay also included controls <strong>of</strong> non-specific or<br />

superantigenic stimuli, such as phytohemagglutin (PHA). The<br />

results <strong>of</strong> 173 assays have allowed us to identify several<br />

peptides as being immunodominant, including some never<br />

before reported, and others as being non-reactive. The EAA<br />

can be utilized to screen additional peptides or combinations<br />

that have biological significance. The results have permitted<br />

us to reduce the number <strong>of</strong> peptides to 109, in the screening<br />

assay for Tovaxin vaccine production, to qualify subjects for<br />

our current Phase IIb clinical trial.<br />

doi:10.1016/j.clim.2007.03.061<br />

Su.22 Differential Gene Expression Identifies Novel<br />

Markers <strong>of</strong> Myelin-Specific Murine Memory CD4+<br />

T Cells<br />

Wassim Elyaman, Research Fellow, Brigham and Women’s<br />

Hospital-Harvard, Boston, MA, Pia Kivisakk, Research<br />

Fellow, Brigham and Women's Hospital, Boston, MA, Jaime<br />

Imitola, Instructor, Brigham and Women’s Hospital, Boston,<br />

MA, Samia Khoury, Associate Pr<strong>of</strong>essor, Brigham and<br />

Women’s Hospital -Harvard, Boston, MA, Mohamed Sayegh,<br />

Pr<strong>of</strong>essor, Brigham and Women's Hospital -Children's<br />

Hospital-Harvard, Boston, MA<br />

Memory CD4+ T cells function as a dynamic repository <strong>of</strong><br />

antigen-experienced T lymphocytes that tend to resist<br />

immunomodulatory and tolerance strategies. Their activation<br />

in response to a secondary antigenic stimulation involves<br />

changes in the expression level <strong>of</strong> a large number <strong>of</strong> genes.<br />

Recently, we have shown that myelin specific CD4 memory<br />

cells express high levels <strong>of</strong> ICOS and lower amounts <strong>of</strong> CD28 in<br />

comparison to effector T cells. This was associated with<br />

differential response to costimulation blockade in a mouse<br />

model <strong>of</strong> autoimmune encephalomyelitis. Here, we have<br />

used cDNA array technology to characterize the differences<br />

in gene expression between murine myelin-specific memory<br />

and effector CD4+ T cells. To generate myelin-specific<br />

memory CD4+ T cells, MOG-TCR transgenic mice were<br />

immunized with MOG/CFA for 2 weeks and CD4+ T cells<br />

were parked for 3 months in Wild-type naïve recipients.<br />

Effector CD4+ T cells were generated by immunizing MOG<br />

transgenic mice for 2 weeks. The gene expression pr<strong>of</strong>iles <strong>of</strong><br />

mouse donors <strong>of</strong> CD4+ T cells <strong>of</strong> each group (n=3) were<br />

analyzed. 204 genes were consistently differentially<br />

expressed (3 fold change) between memory and effector T<br />

cells, which included 23 genes that are only expressed in<br />

CD4+ memory cells. These differentially expressed genes<br />

include a combination <strong>of</strong> well-known, previously characterized<br />

genes with a range <strong>of</strong> biological functions and unknown<br />

in silico predicted hypothetical genes. These observed<br />

differences in the gene expression pr<strong>of</strong>ile <strong>of</strong> autoantigen<br />

memory and effector CD4+ T cells have important clinical<br />

implications for designing new therapies for autoimmune<br />

diseases in humans.<br />

doi:10.1016/j.clim.2007.03.063<br />

S149


S150 Abstracts<br />

Su.23 Reduced Number <strong>of</strong> Blood Circulating Foxp3+<br />

CD25highCD4+ Regulatory T Cells and a Decreased<br />

Foxp3 Expression at the Single-cell Level in Patients<br />

with Relapsing-remitting Multiple Sclerosis<br />

Koen Venken, MSc, Hasselt University, Biomedisch<br />

Onderzoeksinstituut, Diepenbeek, Belgium, Niels Hellings,<br />

PhD, Hasselt University, Biomedisch Onderzoeksinstituut,<br />

Diepenbeek, Belgium, Veerle Somers, PhD, Hasselt<br />

University, Biomedisch Onderzoeksinstituut, Diepenbeek,<br />

Belgium, Pieter Meuwissen, Student, Hasselt University,<br />

Biomedisch Onderzoeksinstituut, Diepenbeek, Belgium,<br />

Marielle Thewissen, MSc, Hasselt University, Biomedisch<br />

Onderzoeksinstituut, Diepenbeek, Belgium, Karen Hensen,<br />

PhD, <strong>Clinical</strong> Laboratory <strong>of</strong> Experimental Hematology, Virga<br />

Jesse Hospital, Hasselt, Belgium, Jean-Luc Rummens, MD,<br />

<strong>Clinical</strong> Laboratory <strong>of</strong> Experimental Hematology, Virga<br />

Jesse Hospital, Hasselt, Belgium, Piet Stinissen, PhD,<br />

Hasselt University, Biomedisch Onderzoeksinstituut,<br />

Diepenbeek, Belgium<br />

CD4+CD25high regulatory T cells (Tregs) <strong>of</strong> patients with<br />

relapsing–remitting (RR), but not secondary-progressive (SP)<br />

multiple sclerosis (MS), show a reduced suppressive function<br />

and FoxP3 mRNA expression. In this study we analyzed FoxP3<br />

protein expression by means <strong>of</strong> flow cytometry in PBMC <strong>of</strong> 55<br />

RR-MS, 15 SP-MS patients and 40 healthy controls (HC). RR-MS<br />

patients displayed a significant reduced number <strong>of</strong> CD4+<br />

CD25highFoxp3+ T cells as compared to HC and SP-MS. In<br />

addition, a significant lower Foxp3 expression per cell was<br />

detected in RR-MS patients. Interestingly, IFN-β treated RR-<br />

MS patients (n=15) showed a higher frequency <strong>of</strong> CD4+ CD25<br />

highFoxp3+ cells as compared to untreated RR-MS patients<br />

(n=40). Furthermore, a correlation was observed between<br />

Foxp3 levels and suppressive capacity <strong>of</strong> Tregs as measured in<br />

Treg-Tresponder coculture experiments. We further phenotypically<br />

analyzed Tregs <strong>of</strong> HC and untreated MS patients by<br />

measuring the expression <strong>of</strong> adhesion molecules by flow<br />

cytometry. Whereas no differences were detected in percentage<br />

<strong>of</strong> L-selectin+ (CD62L) and hyaluronate receptor+ (CD44)<br />

cells, a significant higher percentage <strong>of</strong> integrin αE+ (CD103)<br />

and α4+ (CD49d, VLA-4) cells was observed within CD4+<br />

CD25high Foxp3+ Tregs <strong>of</strong> RR-MS as compared to HC and SP-<br />

MS. Taken together, a lower number <strong>of</strong> CD4+ CD25highFoxp3+<br />

T cells and a reduced Foxp3 expression level was detected in<br />

RR-MS patients further demonstrating Treg dysfunction in<br />

these patients. The detection <strong>of</strong> a higher percentage <strong>of</strong><br />

CD103+ and VLA-4+ in the Treg population in RR-MS patients<br />

could reflect an increased migration capacity <strong>of</strong> Tregs<br />

towards inflammatory lesions in the central nervous system.<br />

doi:10.1016/j.clim.2007.03.064<br />

Su.24 Upregulation <strong>of</strong> Water Channel Aquaporin-4<br />

in Experimental Autoimmune Encephalomyeritis<br />

Katsuichi Miyamoto, MD, PhD, Kinki University School <strong>of</strong><br />

Medicine, Osaka-sayama, Japan, Kazuo Kataoka, MD, PhD,<br />

Department <strong>of</strong> Neurosurgery, Kinki University School <strong>of</strong><br />

Medicine, Osaka-sayama, Japan, Susumu Kusunoki, MD,<br />

PhD, Department <strong>of</strong> Neurology, Kinki University School <strong>of</strong><br />

Medicine, Osaka-sayama, Japan<br />

Aquaporin-4 (AQP4) is a water channel protein, which plays<br />

an important role in water movement in the central nervous<br />

system. Involvement <strong>of</strong> AQP4 in the pathogenesis <strong>of</strong> brain edema<br />

in the acute ischemic brain model was reported. Recently<br />

presence <strong>of</strong> the antibody against AQP4 has been reported in the<br />

patients affected with neuromyelitis optica (NMO) or opticospinal<br />

type multiple sclerosis (MS). AQP4 therefore may be<br />

involved in the pathophysiological mechanism <strong>of</strong> inflammatory<br />

demyelinating disorder. Here we analyzed AQP4 expression in<br />

the central nervous system <strong>of</strong> experimental autoimmune<br />

encephalomyelitis (EAE), an animal model <strong>of</strong> MS. We performed<br />

semi-quantitative analysis <strong>of</strong> AQP4-mRNA in brain and spinal<br />

cord from EAE-induced mice by using real-time PCR. Furthermore<br />

immuno-histochemical analysis was performed. The brain<br />

and spinal cord from myelin oligodendrocyte glycoprotein<br />

(MOG)-induced EAE mice were removed on 7, 14, 21, 28 and<br />

35 days after first inoculation. The increased levels <strong>of</strong> AQP4mRNA<br />

expression were seen; that in the spinal cord reached the<br />

peak on 14 days, and that in the brain on 21 days after first<br />

inoculation, respectively. The histopathological analysis showed<br />

that demyelination and cell infiltration were most severe on<br />

21 days after first inoculation, when the clinical manifestations<br />

also were most severe. This is the first study to show the<br />

upregulation <strong>of</strong> AQP4 in the inflammatory demyelinating disease<br />

<strong>of</strong> CNS. Further investigation is necessary to investigate the<br />

possibility that anti-AQP4 antibody binds to the upregulated<br />

AQP4 within CNS and is directly involved in the pathogenesis <strong>of</strong><br />

NMO or optico-spinal type MS.<br />

doi:10.1016/j.clim.2007.03.065<br />

Su.26 Role <strong>of</strong> IFN-γ in Virus-induced Demyelinating<br />

Disease<br />

Julie Olson, Assistant Pr<strong>of</strong>essor, Department <strong>of</strong> Neurological<br />

Surgery, University <strong>of</strong> Wisconsin-Madison, Madison, WI<br />

Multiple sclerosis is a demyelinating disease in humans<br />

which has been associated with an inflammatory component.<br />

Epidemiological evidence has suggested that a virus infection<br />

may be involved in disease initiation and disease exacerbation.<br />

Theiler's murine encephalomyelitis virus (TMEV) serves as a<br />

mouse model <strong>of</strong> MS. Susceptible mice (SJL strain) infected with<br />

TMEV, BeAn strain, develop demyelinating disease beginning<br />

with clinical signs <strong>of</strong> disease around 35 days post infection.<br />

TMEV induces a chronic progressive demyelinating disease<br />

associated with a myelin-specific CD4+ Tcell response which is<br />

a Th1 type Tcell response. However, some strains <strong>of</strong> mice, such<br />

as C57BL6, are resistant to development <strong>of</strong> TMEV- induced<br />

demyelinating disease. IFN-γ is a proinflammatory cytokine<br />

associated with demyelinating disease and the myelin- specific<br />

autoimmune response. In these studies, the role <strong>of</strong> IFN-γ was<br />

examined during the initiation and progression <strong>of</strong> demyelinating<br />

disease using resistant C57BL6 mice deficient in IFN-γ.<br />

Interestingly, the IFN-γ deficient mice infected with TMEV<br />

developed demyelinating disease with clinical disease similar<br />

to the susceptible (SJL) mice. TMEV- infected IFN γ deficient<br />

mice developed a virus- specific CD4+ T cell response and a<br />

myelin- specific T cell response late during demyelinating<br />

disease. The demyelinating disease was associated with<br />

infiltration <strong>of</strong> immune cells into the CNS, most interestingly,<br />

CD4+ T cells and activated macrophage. These results suggest


Abstracts<br />

that IFN-γ was protective against development <strong>of</strong> virusinduced<br />

demyelinating disease and that IFN-γ was not required<br />

for development <strong>of</strong> autoimmune demyelinating disease. This<br />

research was supported by the National Multiple Sclerosis<br />

Society RG3625.<br />

doi:10.1016/j.clim.2007.03.066<br />

Su.27 Large Scale Immunopr<strong>of</strong>iling Uncovers<br />

Population Structure in Untreated MS Subjects and<br />

Highlights the Role <strong>of</strong> NK Cells<br />

Elizabeth Rossin, Data Analyst, The Broad Institute <strong>of</strong> MIT<br />

and Harvard, The Program in Medical and Population<br />

Genetics, Cambridge, MA, Vissia Viglietta, Instructer,<br />

Brigham and Women’s Hospital, Department <strong>of</strong> Neurology,<br />

Boston, MA, Dulce Soler-Ferran, Research Scientist,<br />

Millenium Pharmaceuticals, Cambridge, MA, Alex Parker,<br />

Research Scientist, Millenium Pharmaceuticals, Cambridge,<br />

MA, Mira Weiner, Project Manager, Brigham and Women’s<br />

Hospital, Department <strong>of</strong> Neurology, Boston, MA, Guy<br />

Buckle, Associate Neurologist, Department <strong>of</strong> Neurology,<br />

Brigham and Women's Hospital, Brookline, MA, Samia<br />

Khoury, Associate Pr<strong>of</strong>essor, Brigham and Women’s Hospital,<br />

Department <strong>of</strong> Neurology, Boston, MA, David Hafler,<br />

Pr<strong>of</strong>essor, Brigham and Women’s Hospital, Department <strong>of</strong><br />

Neurology, Boston, MA, Howard Weiner, Pr<strong>of</strong>essor, Brigham<br />

and Women’s Hospital, Department <strong>of</strong> Neurology, Boston,<br />

MA, Philip De Jager, Assistant Pr<strong>of</strong>essor, Brigham and<br />

Women’s Hospital, Department <strong>of</strong> Neurology, Boston, MA<br />

As part <strong>of</strong> the Multiple Sclerosis Registry, we acquired an<br />

immunological pr<strong>of</strong>ile from healthy subjects and subjects<br />

with remitting relapsing multiple sclerosis (RRMS) who were<br />

untreated. Fresh blood from each subject was screened ex<br />

vivo using a panel <strong>of</strong> 50 fluorescently labeled monoclonal<br />

antibodies (Ab) distributed amongst 56 pools; flow cytometry<br />

was performed to quantitate the binding <strong>of</strong> each Ab.<br />

Longitudinal analysis <strong>of</strong> 15 healthy control subjects identified<br />

the “% positive” parameters as the most robust to<br />

fluctuation over time; after further quality control processing,<br />

1018 individual “% positive” parameters or “features”<br />

were selected for the analysis. The exploratory screen<br />

consisted <strong>of</strong> 17 healthy control subjects and 23 untreated<br />

RRMS subjects. It was followed by an extension analysis in<br />

which 15 additional healthy controls and 15 additional RRMS<br />

subjects were added to the dataset. This analysis identified a<br />

CD8lowCD56+CD3− population (NK cells) as being reduced in<br />

frequency in untreated RRMS subjects (P=10–4). In analyzing<br />

the 38 cases by themselves, clustering analysis revealed that<br />

there were 3 populations <strong>of</strong> RRMS subjects with different<br />

immunological pr<strong>of</strong>iles and that disease duration (range 3–<br />

42 years) might explain some <strong>of</strong> this population structure<br />

(P=0.03). In prediction analyses, our Ab panel was unable to<br />

accurately predict control and RRMS subjects but was able to<br />

accurately segregate 11 cases <strong>of</strong> clinically isolated syndrome<br />

from controls. These data suggest that disease duration is an<br />

important factor to consider in the analysis and future<br />

development <strong>of</strong> immunological pr<strong>of</strong>iling in MS.<br />

doi:10.1016/j.clim.2007.03.068<br />

Su.28 Treatment with a Fullerene Derivative<br />

(ABS-75) Reduces Disease Progression and Axonal<br />

Loss in MOG-induced Progressive EAE in NOD Mice<br />

Alexandre S. Basso, Research Fellow, Center for Neurologic<br />

Disease, Harvard Medical School, Boston, MA, Dan Frenkel,<br />

Research Fellow, Center for Neurologic Diseases, Harvard<br />

Medical School, Boston, MA, Sanja Petrovic-Stojkovic,<br />

Research Assistant, Center for Neurologic Diseases, Harvard<br />

Medical School, Boston, MA, Alon Monsonego, Research<br />

Fellow, Center for Neurologic Diseases, Harvard Medical<br />

School, Boston, MA, Lindsay Puckett, Research Assistant,<br />

Center for Neurologic Diseases, Harvard Medical School,<br />

Boston, MA, Michael Gozin, Pr<strong>of</strong>essor, School <strong>of</strong> Chemistry,<br />

Faculty <strong>of</strong> Exact Sciences, Tel Aviv University, Tel Aviv,<br />

Israel, Howard Weiner, Robert L. Kroc Pr<strong>of</strong>essor <strong>of</strong><br />

Neurology, Center for Neurologic Diseases, Harvard Medical<br />

School, Boston, MA<br />

Inflammation-induced oxidative stress can lead to axonal<br />

degeneration, which is felt to be a major determinant <strong>of</strong><br />

progressive neurological disability in Multiple Sclerosis (MS).<br />

Water-soluble derivatives <strong>of</strong> fullerenes are a unique class <strong>of</strong><br />

allotropic form <strong>of</strong> carbon compounds with potent antioxidant<br />

properties. 10 week old mice were immunized with 150 μg<strong>of</strong><br />

MOG 35–55 peptide in CFA followed by pertussis toxin.<br />

Disease is characterized by an attack followed by a<br />

progressive phase with chronic clinical impairment. Following<br />

the first attack, animals were distributed into different<br />

groups with similar disease courses and treated intraperitoneally<br />

either with 200 μl<strong>of</strong>a1μM fullerene solution or vehicle<br />

every day until termination <strong>of</strong> the experiment (day 63). We<br />

tested three different fullerene derivatives (ABS-75, ABS-16,<br />

and the C60 fullerene core) and found that ABS-75 treatment<br />

initiated after disease onset reduced the clinical progression<br />

<strong>of</strong> chronic EAE in NOD mice (treated vs. control, pb0.05).<br />

Fullerene ABS-75 consists <strong>of</strong> a C60 carbon fullerene core to<br />

which a known NMDA receptor ligand was attached. ABS-75<br />

treatment also reduced axonal loss, demyelination (as<br />

evaluated by silver and Luxol fast blue staining, respectively),<br />

and CD11b+ infiltration in the white matter <strong>of</strong> mice.<br />

In vitro, ABS-75 was able to protect neurons from glutamateinduced<br />

toxicity and to reverse reduced glutamine synthetase<br />

expression in astrocytes under inflammatory insult. Our<br />

data demonstrate a neuroprotective effect <strong>of</strong> a treatment<br />

with a fullerene compound combined with a NMDA receptor<br />

ligand that may have applicability in the treatment <strong>of</strong><br />

progressive MS and other neurodegenerative diseases.<br />

doi:10.1016/j.clim.2007.03.069<br />

S151<br />

Su.29 Human Blood−brain Barrier-associated DCs<br />

Originate from Blood Monocytes and Polarize CD4+<br />

Lymphocytes into Th17 or Th1<br />

Igal Ifergan, PhD Student, CHUM-Hopital Notre-Dame,<br />

Neuroimmunology Department, Montreal, QC, Canada,<br />

Hania, Kebir, PhD student, CHUM-Hopital Notre-Dame,<br />

Neuroimmunology Department, Montreal, QC, Canada,<br />

Nathalie Arbour, PhD, CHUM-Hopital Notre-Dame,<br />

Neuroimmunology Department, Montreal, QC, Canada,<br />

Alexandre Prat, MD, PhD, CHUM-Hopital Notre-Dame,<br />

Neuroimmunology Department, Montreal, QC, Canada


S152 Abstracts<br />

Trafficking <strong>of</strong> antigen presenting cells (APC) into the<br />

central nervous system (CNS) is essential for lymphocyte<br />

reactivation within the CNS compartment. The origin and<br />

function <strong>of</strong> the APC found in CNS autoimmune or inflammatory<br />

lesions remain, however, controversial. We demonstrate<br />

that blood CD14+ monocytes migrate across the<br />

inflammed blood–brain barrier (BBB) and differentiate into<br />

CD209+CD83+ (myeloid) or CD123+ (plasmacytoid) dendritic<br />

cells (DCs) under the influence <strong>of</strong> CNS endothelial-secreted<br />

transforming growth factor-beta (TGF-β) and granulocyte<br />

macrophage colony stimulating factor (GM-CSF). We also<br />

demonstrate that while CD123+ DCs secrete interleukin-12<br />

p70 (IL-12p70) and promote the proliferation and expansion<br />

<strong>of</strong> interferon-γ-secreting Th1 CD4+ lymphocytes, CD209+<br />

CD83+ DCs secrete high levels <strong>of</strong> TGF-β and IL-6 and skew<br />

CD4+ lymphocytes towards a Th17 phenotype. Using multiple<br />

sclerosis as a prototypical human CNS autoimmune<br />

disease, we confirmed by immunohistochemistry the abundance<br />

<strong>of</strong> such perivascular CD83+ and CD123+ DCs within<br />

acute lesions, closely associated with microvascular BBBendothelial<br />

cells. Our data support the notion that<br />

functional perivascular myeloid and plasmacytoid CNS DCs<br />

arise as a consequence <strong>of</strong> migration <strong>of</strong> blood CD14+<br />

monocytes across the human BBB, through the concerted<br />

actions <strong>of</strong> TGF-β and GM-CSF.<br />

doi:10.1016/j.clim.2007.03.071<br />

Su.31 Characterization <strong>of</strong> Single B Cell<br />

Immunoglobulin Variable Region Genes Derived<br />

from Infiltrated Muscle Tissue <strong>of</strong> Subjects with<br />

Inflammatory Myopathies<br />

Elizabeth Bradshaw, Postdoctoral Fellow, Laboratory <strong>of</strong><br />

Molecular <strong>Immunology</strong>, Center for Neurologic Diseases,<br />

Brigham and Women’s Hospital, Boston, MA, Ana Orihuela,<br />

Research Technician, Children’s Hospital Informatics<br />

Program at the Harvard-MIT Division <strong>of</strong> Health Sciences and<br />

Technology, Boston, MA, Shannon McArdel, Research<br />

Technician, Laboratory <strong>of</strong> Molecular <strong>Immunology</strong>, Center for<br />

Neurologic Diseases, Brigham and Women’s Hospital,<br />

Boston, MA, David Hafler, Pr<strong>of</strong>essor, Laboratory <strong>of</strong><br />

Molecular <strong>Immunology</strong>, Center for Neurologic Diseases,<br />

Brigham and Women’s Hospital, Boston, MA, Anthony<br />

Amato, Associate Pr<strong>of</strong>essor, Department <strong>of</strong> Neurology,<br />

Division <strong>of</strong> Neuromuscular Disease, Brigham and Women’s<br />

Hospital and Harvard, Boston, MA, Kevin O’Connor,<br />

Assistant Pr<strong>of</strong>essor, Laboratory <strong>of</strong> Molecular <strong>Immunology</strong>,<br />

Center for Neurologic Diseases, Brigham and Women's<br />

Hospital, Boston, MA<br />

The inflammatory myopathies (IM) are putative autoimmune<br />

disorders characterized by muscle weakness and the<br />

presence <strong>of</strong> inflammatory infiltrates in skeletal muscle.<br />

Varying numbers <strong>of</strong> both B cells and plasma cells are among<br />

the muscle tissue infiltrate found in these diseases. Through<br />

examination <strong>of</strong> immunoglobulin heavy chain variable regions,<br />

we previously demonstrated that there was significant<br />

oligoclonal expansion <strong>of</strong> B cells found in IM muscle. To extend<br />

these findings, we isolated single B cells from IM muscle by<br />

laser capture microdissection or FACS, then examined the<br />

paired immunoglobulin heavy and light chain variable region<br />

sequences. B cell immunoglobulin variable region cDNAs were<br />

sequenced affording identification <strong>of</strong> the isotype, CDR3 and<br />

the degree <strong>of</strong> somatic mutation in the entire V-region. A<br />

pattern <strong>of</strong> B cell affinity maturation was evident in clones<br />

that had accumulated varying numbers <strong>of</strong> common and<br />

unique somatic mutations. These data suggested that a<br />

local inflammatory response occurs within the muscle tissue<br />

<strong>of</strong> patients with IM that may indicate the presence <strong>of</strong> a B cell<br />

antigen-specific response.<br />

doi:10.1016/j.clim.2007.03.072<br />

Su.32 Myelin-specific Regulatory T Cells Accumulate<br />

in the Central Nervous System, but Fail to Suppress<br />

Pathogenic Effector T Cells at the Peak <strong>of</strong><br />

Autoimmune Inflammation<br />

Thomas Korn, Postdoctoral Fellow, Harvard Medical School,<br />

Center for Neurologic Diseases, Boston, MA, Mohamed<br />

Oukka, Instructor, Harvard Medical School, Center for<br />

Neurologic Diseases, Boston, MA, Jay, Reddy, Instructor,<br />

Harvard Medical School, Center for Neurologic Diseases,<br />

Boston, MA, Estelle Bettelli, Instructor, Harvard Medical<br />

School, Center for Neurologic Diseases, Boston, MA, Amit<br />

Awasthi, Postdoctoral Fellow, Harvard Medical School,<br />

Center for Neurologic Diseases, Boston, MA, Raymond Sobel,<br />

Associate Pr<strong>of</strong>essor <strong>of</strong> Pathology, Stanford University,<br />

Department <strong>of</strong> Pathology, Stanford, CA, Kai Wucherpfennig,<br />

Associate Pr<strong>of</strong>essor <strong>of</strong> Neurology, Harvard Medical School,<br />

Dana Farber Cancer Institute, Department <strong>of</strong> Cancer<br />

<strong>Immunology</strong> and AIDS, Boston, MA, Vijay Kuchroo, Pr<strong>of</strong>essor<br />

<strong>of</strong> Neurology, Harvard Medical School, Center for Neurologic<br />

Diseases, Boston, MA<br />

Treatment with ex vivo generated regulatory T cells (Treg)<br />

has been regarded as highly attractive therapeutic<br />

approach for autoimmune diseases. However, the dynamics<br />

and function <strong>of</strong> T-reg in autoimmunity are not well understood.<br />

Thus, we developed Foxp3gfp “knock-in” mice and<br />

myelin oligodendrocyte glycoprotein (MOG)35–55/IAb tetramers<br />

to track autoantigen-specific effector T cells (T-eff)<br />

and T-reg in vivo during experimental autoimmune encephalomyelitis,<br />

an animal model for Multiple Sclerosis.<br />

Following immunization with the encephalitogenic peptide<br />

MOG35–55 emulsified in complete Freund's adjuvant,<br />

MOG35–55-tetramer-reactive, Foxp3+ T-reg expanded in<br />

the peripheral lymphoid compartment and readily accumulated<br />

in the central nervous system (CNS), but did not<br />

prevent the onset <strong>of</strong> disease. During disease onset, the MOGtetramer+<br />

T-eff population in the CNS increased faster than<br />

the population <strong>of</strong> antigen-specific T-reg. At the peak <strong>of</strong><br />

disease, the ratio <strong>of</strong> T-reg vs. T-eff was 1:17 which dramatically<br />

changed into 1:2 at the beginning <strong>of</strong> recovery.<br />

Foxp3+ T-reg isolated from the CNS were fully competent in<br />

suppressing naive MOG-specific T cells. However, Foxp3+ Treg<br />

failed to control encephalitogenic T-eff which in contrast<br />

to T-eff from the peripheral immune compartment, secreted<br />

IL-6 and TNF when they were isolated from the CNS at the<br />

peak <strong>of</strong> disease. Our data suggest that in the face <strong>of</strong> inflammation,<br />

the regulation <strong>of</strong> autoimmunity by CD4+Foxp3+<br />

T-reg in situ may not be accomplished simply by changing<br />

the numerical balance <strong>of</strong> antigen-specific pathogenic vs.


Abstracts<br />

regulatory T cells, but may require the control <strong>of</strong> tissue<br />

inflammation as well.<br />

doi:10.1016/j.clim.2007.03.073<br />

Su.34 Proteomic Pr<strong>of</strong>iling <strong>of</strong> Multiple Sclerosis<br />

Lesions Identify Potential Targets for Therapy<br />

May H. Han, Postdoctoral Fellow, Department <strong>of</strong> Neurology<br />

and Neurological Sciences Stanford University, Stanford,<br />

CA, Sunil Hwang, Postdoctoral Fellow, Department <strong>of</strong><br />

Physiology, University <strong>of</strong> Conneticut Health Center,<br />

Farmington, CT, Dolly Roy, Neurologist, Department <strong>of</strong><br />

Neurology and Neurological Sciences, Stanford University,<br />

Stanford, CA, Cedric Raine, Pr<strong>of</strong>essor, Department <strong>of</strong><br />

Pathology, Albert Einstein College <strong>of</strong> Medicine, Bronx, NY,<br />

William H. Robinson, Assistant Pr<strong>of</strong>essor, Departments <strong>of</strong><br />

Medicine and <strong>Immunology</strong>, Stanford University, Stanford,<br />

CA, Raymond A. Sobel, Pr<strong>of</strong>essor <strong>of</strong> Pathology, Department<br />

<strong>of</strong> Pathology, Stanford University, Stanford, CA, David Han,<br />

Associate Pr<strong>of</strong>essor, Department <strong>of</strong> Physiology, University <strong>of</strong><br />

Conneticut Health Center, Farmington, CT, Lawrence<br />

Steinman, Pr<strong>of</strong>essor, Department <strong>of</strong> Neurology and<br />

Neurological Sciences, Stanford, CA<br />

Demyelinating lesions or plaques in multiple sclerosis (MS)<br />

have distinct histological features that reflect their stage <strong>of</strong><br />

evolution. To identify key molecules involved at different stages<br />

during lesion progression, we performed proteomic pr<strong>of</strong>iling <strong>of</strong><br />

three types <strong>of</strong> MS plaques using mass spectrometry and bioinformatics.<br />

Fresh frozen autopsy brain samples from MS<br />

patients (n=6) and age-matched controls (n=2)wereclassified<br />

as 1) acute plaque (AP, characterized by presence <strong>of</strong> inflammatory<br />

cells, ongoing demyelination), 2) chronic active plaque<br />

(CAP, characterized by chronic demyelination, astrogliosis with<br />

active rim) or 3) chronic plaque (CP, characterized by lack <strong>of</strong><br />

inflammatory cells, dense astrogliosis). Samples from plaques<br />

were isolated by laser capture microdissection and global<br />

protein identification was carried out by SDS-PAGE, in-gel<br />

protease digestion, liquid chromatography and mass spectrometry.<br />

A total <strong>of</strong> 3894 proteins were identified out <strong>of</strong> which 2184<br />

proteins were unique to MS lesions. Then we identified proteins<br />

uniquetoeachtype<strong>of</strong>lesion(AP(N=172), CAP (N=248) and CP<br />

(N=252), and proteins common to all three types (N=89) using<br />

bio-informatics s<strong>of</strong>tware INTERSECT. Analysis by s<strong>of</strong>tware<br />

PROTEOME 3D identified proteins <strong>of</strong> coagulation cascade and<br />

neural regeneration which may be potential targets for therapy.<br />

These results provide the first and most comprehensive<br />

information on global protein expression in MS plaques, enhance<br />

our understanding <strong>of</strong> the molecular pathogenesis <strong>of</strong> MS lesions<br />

and identify potential key molecules involved in disease<br />

progression that <strong>of</strong>fer possibilities for therapeutic targets.<br />

doi:10.1016/j.clim.2007.03.075<br />

Su.35 A Genome-wide Screen for Genetic Variants<br />

Affecting the Expression <strong>of</strong> Immunologically<br />

Relevant Cell Surface Markers<br />

Philip De Jager, Assistant Pr<strong>of</strong>essor, Department <strong>of</strong><br />

Neurology, Brigham and Women’s Hospital, Boston, MA,<br />

Roman Yelensky, Student, Medical and Population Genetics<br />

Program, Broad Institute, Cambridge, MA, Elizabeth Rossin,<br />

Data Analyst, Medical and Population Genetics Program,<br />

Broad Institute, Boston, MA, Sasha Bonadkar, Research<br />

Assistant, Medical and Population Genetics Program, Broad<br />

Institute, Cambridge, MA, Edwin Choy, Director, Sarcoma<br />

Research, Medical and Population Genetics Program, Broad<br />

Institute, Cambridge, MA, Mark Daly, Assistant Pr<strong>of</strong>essor,<br />

Medical and Population Genetics Program, Broad Institute,<br />

Cambridge, MA, David Altshuler, Associate Pr<strong>of</strong>essor,<br />

Medical and Population Genetics Program, Broad Institute,<br />

Cambridge, MA, David Hafler, Pr<strong>of</strong>essor, Department <strong>of</strong><br />

Neurology, Brigham and Women's Hospital, Boston, MA<br />

While a few alleles affecting the expression level <strong>of</strong> immunologically<br />

relevant molecules have been identified and<br />

validated, our knowledge <strong>of</strong> human polymorphisms associated<br />

with differences in immune function remains very<br />

limited. We have therefore evaluated the use <strong>of</strong> the HapMap<br />

lymphoblastic cell lines (LCLs) as a platform to discover such<br />

polymorphisms on a large scale using a test panel <strong>of</strong> cell<br />

surface molecules that includes 15 cell-surface markers:<br />

CD19, CD20, CD21, CD40, CD58, CD80, CD86, CD95, CD227,<br />

HLA DQ, HLA DR, IgD, IgG, IgM, and IL6R. We then correlated<br />

the mean expression level <strong>of</strong> a particular molecule in each<br />

LCL with the over 2 million genotypes available in the<br />

HapMap database. Amongst several positive results, this<br />

analysis reveals that certain SNPs in the vicinity <strong>of</strong> the HLA<br />

DQA1 gene are correlated with both the level <strong>of</strong> expression <strong>of</strong><br />

HLA DQ on the LCL surface and the level <strong>of</strong> mRNA expression<br />

for HLA DQA1. One <strong>of</strong> these SNPs, rs9272346, is a null allele<br />

<strong>of</strong> HLA DQA1 and is associated to this immunophenotype at a<br />

level that exceeds our predetermined level <strong>of</strong> genome-wide<br />

significance (P b2.6×10 −8 ), validating our approach. In<br />

exploring the mRNA data further within the MHC class I and<br />

class II clusters, it is clear that several different polymorphisms<br />

exist that affect the level <strong>of</strong> expression <strong>of</strong> these<br />

molecules. We are currently assessing the role <strong>of</strong> these<br />

functional variants in the association <strong>of</strong> the MHC with MS<br />

susceptibility.<br />

doi:10.1016/j.clim.2007.03.076<br />

S153<br />

Su.36 Novel Computational Methods to Enhance the<br />

Analysis <strong>of</strong> Cytometric Immunophenotypes<br />

Philip De Jager, Assistant Pr<strong>of</strong>essor, Department <strong>of</strong><br />

Neurology, Brigham and Women’s Hospital, Boston, MA,<br />

Saumyadipta Pyne, Postdoctoral Fellow, Cancer Program,<br />

Broad Institute, Cambridge, MA, Elizabeth Rossin, Data<br />

Analyst, Medical and Population Genetics Program, Broad<br />

Institute, Cambridge, MA, Jill Mesirov, Director, Cancer<br />

Program, Broad Institute, Cambridge, MA, Pablo Tamayo,<br />

Staff Scientist, Cancer Program, Broad Institute,<br />

Cambridge, MA, David Hafler, Pr<strong>of</strong>essor, Department <strong>of</strong><br />

Neurology, Brigham and Women’s Hospital,<br />

Boston, MA<br />

The current method for analyzing raw flow cytometry<br />

information involves gating, the process <strong>of</strong> first visualizing<br />

cell-surface marker expression values for a population <strong>of</strong><br />

cells and then manually selecting distinct cell populations on


S154 Abstracts<br />

which to report statistics. Thus, manual processing is timeconsuming<br />

and is limited to 2-dimensional projections. Furthermore,<br />

these cell populations are described by statistics<br />

that fail to characterize their full distribution and shape. We<br />

propose two automated methods to improve these shortcomings:<br />

(1) histogram mean shape comparison: this method<br />

creates an n+1-dimensional histogram for n-dimensional<br />

data. Expression data is then parsed into n+1-dimensional<br />

bins, and individual bin values can be compared across<br />

categories <strong>of</strong> subjects or samples to discover cell populations<br />

that differ across sample categories. (2) Hierarchical mixture<br />

model: this method characterizes the expression values <strong>of</strong><br />

each cell population in n dimensions using a hierarchical<br />

mixture model. The mixture model clusters individual cells<br />

by assigning them to distributions with maximum likelihood.<br />

A mixture <strong>of</strong> distributions that make up a model thus<br />

characterizes the n dimensional data. This method allows<br />

the cells to exist in distinct distributions rather than be<br />

collapsed to a statistic, and the distribution <strong>of</strong> cells can then<br />

be compared across samples. Both methods have been<br />

implemented to characterize the expression <strong>of</strong> 15 molecules<br />

on the 269 HapMap lymphoblastic cell lines. We present a<br />

comparison <strong>of</strong> both methods against manually processed<br />

data to demonstrate the most robust pipeline with which to<br />

characterize cell population structure and calculate expression<br />

levels using cytometric data.<br />

doi:10.1016/j.clim.2007.03.077<br />

Su.37 Improving Elispot to Detect Antigen Specific<br />

T Cells<br />

Khadir Raddassi, Research Instructor, Brigham and Women’s<br />

Hospital, Neurology, Boston, MA, Kasia Bourcier, Scientist,<br />

Immune Tolerance Network, San Francisco, CA, Jose<br />

Estevam, Research Assistant, Brigham and Women's<br />

Hospital, Neurology, Boston, MA, Vicki, Seyfert-Margolis,<br />

Scientist, Immune Tolerance Network, San Francisco, CA,<br />

David Hafler, Lab Director, Brigham and Women’s Hospital,<br />

Neurology, Boston, MA<br />

A major interest <strong>of</strong> our Immune Tolerance Network assay<br />

group is to develop and validate assays to detect auto- and<br />

allo-antigen reactive cells. In collaboration with Dr. Peakman<br />

(Kings College, London) we have developed a modified<br />

elispot protocol. We tested the sensitivity <strong>of</strong> this improved<br />

elispot to detect myelin-reactive T cells in multiple sclerosis<br />

patients (MS) and healthy controls (HC). Fresh or cryopreserved<br />

PBMCs were exposed to myelin antigens or tetanus<br />

toxoid (TT). After 42 hours the cells were transferred to<br />

elispot plates for 16 hours; the elispot plates were developed<br />

to detect the frequency <strong>of</strong> IFNg secreting cells. Comparing<br />

side by side elispot and flow cytometry regarding IFNg<br />

production in response to TT showed similar results, but<br />

elispot was more sensitive at low doses stimulation. The<br />

detection <strong>of</strong> antigen reactive cells by elispot could be<br />

performed on fresh and cryopreserved PBMCs. We validated<br />

this method between different labs, the data were well<br />

correlated (r2=0.987). Using IL-7 enhanced the response to<br />

weak stimuli. Taking advantage <strong>of</strong> this improved elispot and<br />

IL-7, we examined the reactivity <strong>of</strong> PBMCs from 13 HC and 11<br />

MS. In the presence <strong>of</strong> IL-7 PBMCs from MS showed high<br />

reactivity to human MBP (30±5 spots for MS vs. 17±4 for HC)<br />

and to a pool <strong>of</strong> myelin peptides (46 ±8 spots for MS vs. 15±4<br />

for HC). This modified elispot would provide a quick method<br />

to monitor immune reaction in clinical research. The fast<br />

read out allows the cells to be recovered and used for further<br />

testing.<br />

doi:10.1016/j.clim.2007.03.078<br />

Su.38 TIM-3 is a Negative Regulator <strong>of</strong> Human Th1<br />

Cells<br />

David E. Anderson, PhD, Brigham and Women’s Hospital and<br />

Harvard Medical School, Boston, MA, Juhi Kuchroo, Womens<br />

Hospital and Harvard Medical School, Boston, MA, Nasim<br />

Kassam, Brigham and Women’s Hospital and Harvard<br />

Medical School, Boston, MA, David Hafler, Jack, Sadie and<br />

David Breakstone Pr<strong>of</strong>essor <strong>of</strong> Neurology (Neuroscience),<br />

Brigham and Womens Hospital and Harvard Medical School,<br />

Boston, MA<br />

The relevance and utility <strong>of</strong> the Th1/Th2 paradigm has<br />

been well established in murine models <strong>of</strong> a many human<br />

immune-mediated diseases. However, data on the role and<br />

regulation <strong>of</strong> Th1 and Th2 cells in modulating immune<br />

responses in humans is less clear. TIM-3 is a molecule<br />

selectively expressed on a subset <strong>of</strong> murine IFN- 3 -secreting<br />

Th1 cells but not Th2 cells, and regulates Th1 immunity and<br />

tolerance in vivo. To determine if TIM-3 similarly identifies<br />

and regulates Th1 cells in humans, we generated a panel <strong>of</strong><br />

monoclonal antibodies specific for human TIM-3. We report<br />

that TIM-3 is preferentially expressed on the surface <strong>of</strong> in<br />

vitro polarized human Th1 cells. TIM-3 is not readily<br />

apparent on the surface <strong>of</strong> CD4+ T cells examined ex vivo,<br />

but T cell activation rapidly induces cell surface expression<br />

<strong>of</strong> TIM-3 on a subset <strong>of</strong> IFN- 3 -secreting CD4+ T cells. More<br />

specifically, highest expression <strong>of</strong> TIM-3 is present on T cells<br />

secreting the greatest amounts <strong>of</strong> IFN- 3 . Functionally,<br />

human T cells secrete elevated levels <strong>of</strong> IFN- 3 when<br />

stimulated in the presence <strong>of</strong> TIM-3-specific antibodies,<br />

with no effect on other cytokines including TNF-α and IL-<br />

13. These results demonstrate that TIM-3 preferentially<br />

regulates IFN- 3 -secretion from human Th1 cells and may<br />

influence a variety <strong>of</strong> human diseases including cancer and<br />

autoimmunity.<br />

doi:10.1016/j.clim.2007.03.079<br />

Su.39 Rapamycin Inhibits Autocrine Signaling<br />

(IL-6, IL-10) in Viral Lymphomas<br />

Sang-Hoon Sin, Postdoctoral Research Associate, University<br />

<strong>of</strong> North Carolina Chapel Hill, Chapel Hill, NC, Debasmita<br />

Roy, Graduate Student, University <strong>of</strong> North Carolina Chapel<br />

Hill, Microbiology & <strong>Immunology</strong>, Chapel Hill, NC,<br />

Dhavalkumar Patel, Pr<strong>of</strong>essor, University <strong>of</strong> North Carolina,<br />

Chapel Hill, Chapel Hill, NC, Dirk Dittmer, Assistant<br />

Pr<strong>of</strong>essor, University <strong>of</strong> North Carolina, Chapel Hill,<br />

Microbiology & <strong>Immunology</strong>, Chapel Hill, NC<br />

The mTOR inhibitor, rapamycin (sirolimus) is known for its<br />

immunosuppressive activity and more recently has been


Abstracts<br />

subject <strong>of</strong> intense investigations as an anti-cancer agent.<br />

Here we show that a group <strong>of</strong> viral malignancies, which<br />

critically dependent in inflammatory cytokines (and thus<br />

have sometimes be described as viral inflammatory lesions)<br />

are susceptible to rapamycin. We found that rapamycin was<br />

efficacious against primary effusion lymphoma in culture and<br />

in a murine xenograft model; that rapamycin inhibited mTOR<br />

signaling and that this correlated with inhibition <strong>of</strong> IL-10, IL-<br />

6, IFNgamma, IP-10, and IL-12p40 secretion (as measured by<br />

Luminex and ELISA). Moreover, addition <strong>of</strong> exogenous IL-10 or<br />

IL-6 could be reversed the rapamycin growth arrest. This<br />

validates sirolimus as a new treatment option for viral<br />

malignancies that depend on inflammatory cytokines.<br />

doi:10.1016/j.clim.2007.03.080<br />

Su.40 The Transcription Factor GATA-3 Regulates<br />

IL-4 Responses in B cells<br />

Dee Aud, Research Scientist, Roche Palo Alto, Palo Alto, CA,<br />

Sung-Yun Pai, Instructor, Dana Farber Cancer Institute,<br />

Boston, MA, Stanford Peng, Director, Arthritis Research,<br />

Roche Palo Alto, Palo Alto, CA, I-Cheng Ho, Associate<br />

Pr<strong>of</strong>essor, Brigham and Women's Hospital, Boston, MA<br />

The GATA-3 transcription factor plays a central role in T<br />

helper type 2 development, but its role in B cells has not been<br />

well described, even though GATA-3 is present in naïve B cells.<br />

To study the role <strong>of</strong> GATA-3 in B cells, mice with a floxed Gata3<br />

allele (Gata3fl/fl) mice were crossed to CD19-Cre transgenic<br />

mice, allowing B cell-specific deletion <strong>of</strong> GATA-3. B cell<br />

development was grossly normal in Gata3fl/fl CD19-Cre, but<br />

surprisingly, Gata3fl/fl CD19-Cre B cells showed a decreased<br />

ability to proliferate (in an IL-4 dose dependent manner) but<br />

an increased ability to produce IgG1 and IgE in response to<br />

anti-CD40 and/or IL-4 in vitro. These findings may reflect an<br />

interaction between GATA-3 and type 2 Ig-regulating transcription<br />

factors such as NF-kB or STAT6, because GATA-3 can<br />

modulate the activities <strong>of</strong> these factors in vitro. These<br />

findings demonstrate a critical yet surprising role for GATA-3<br />

in the regulation <strong>of</strong> IL-4-mediated responses in B cells.<br />

doi:10.1016/j.clim.2007.03.081<br />

Su.41 Natural and Synthetic TLR7 Ligands Inhibit<br />

CpG-A- and CpG-C-ODN-induced IFN-α Production<br />

by Plasmacytoid Dendritic Cells<br />

Holger Hackstein, Resident, University <strong>of</strong> Giessen, Giessen,<br />

Germany, Beate Berghöfer, PhD Student, University <strong>of</strong><br />

Giessen, Giessen, Germany, Gregor Bein, Pr<strong>of</strong>essor,<br />

University <strong>of</strong> Giessen, Giessen, Germany<br />

Plasmacytoid dendritic cells (pDC) are unique with respect<br />

to their capacity to produce unsurpassed amounts <strong>of</strong><br />

IFN-α and coexpress TLR7 and TLR9, mediating IFN-α<br />

production. Although TLRs are critical receptors <strong>of</strong> innate<br />

immunity, little is known about the immunological effects <strong>of</strong><br />

TLR7/TLR9 costimulation. We have analyzed the effects <strong>of</strong><br />

TLR7/TLR9 costimulation on IFN-α production by leukocytes<br />

and pDCs. Our experiments revealed that both synthetic<br />

(resiquimod, loxoribine) and natural (ssRNA40) TLR7 ligands<br />

abrogate CpG-A- and CpG-C-ODN-induced IFN-α production<br />

by human leukocytes. Since TLR7 ligands themselves<br />

represent important IFN-α inducers, we demonstrated that<br />

substimulatory TLR7 ligand concentrations significantly<br />

inhibited CpG-A-induced IFN-α. Delayed addition <strong>of</strong> TLR7<br />

ligands still resulted in complete suppression <strong>of</strong> CpG-A-ODNinduced<br />

IFN-α production, suggesting that the inhibition is<br />

unlikely to be caused by a kinetic uptake advantage. Unlike<br />

for CpG-A and CpG-C, TLR7 ligands did not inhibit CpG-B-<br />

ODN-induced IFN-α production. Experiments with purified<br />

human pDCs demonstrated that the inhibitory effects <strong>of</strong><br />

TLR7/TLR9 costimulation were mediated directly by pDCs.<br />

Suppression <strong>of</strong> IFN-α production was not related to increased<br />

cell death and was also detectable in enriched mouse pDCs.<br />

Analyses <strong>of</strong> pDCs suggested that the TLR7 signal regulates the<br />

outcome <strong>of</strong> TLR7 ligand/CpG-A-ODN costimulation and can<br />

either inhibit (IFN-α) or promote (IL-8/CD40) cytokine and<br />

surface marker expression. Our data reveal for the first time<br />

a strong inhibitory effect <strong>of</strong> TLR7 stimulation on IFN-α<br />

production induced by CpG-A- and CpG-C-ODNs. These<br />

findings provide novel insight into the effects <strong>of</strong> TLR7/TLR9<br />

costimulation and may support the development <strong>of</strong> novel<br />

TLR9 inhibitors.<br />

doi:10.1016/j.clim.2007.03.082<br />

S155<br />

Su.42 Association <strong>of</strong> MHC Class II Haplotypes and<br />

Immunogenicity <strong>of</strong> a Therapeutic Biological Protein<br />

Danika Wullner, Senior Associate, Amgen, Thousand Oaks,<br />

CA, Erica Yue, Associate, <strong>Clinical</strong> <strong>Immunology</strong>, Thousand<br />

Oaks, CA, Steven J. Swanson, Executive Director, <strong>Clinical</strong><br />

<strong>Immunology</strong>, Thousand Oaks, CA, Vibha Jawa, Senior<br />

Scientist, <strong>Clinical</strong> <strong>Immunology</strong>, Thousand Oaks, CA<br />

A mature high affinity antibody response that is T cell<br />

driven can be elicited during administration <strong>of</strong> biological<br />

therapeutics in humans. The processing <strong>of</strong> the protein<br />

antigen by antigen presenting cells and their binding to<br />

Class II HLA molecules is necessary for the antibody response<br />

to mature. In this study, the predisposition <strong>of</strong> certain MHC<br />

haplotypes to present peptides derived after processing a<br />

recombinant protein (FPX-protein) is discussed. Ex vivo T<br />

cell priming assays were used to predict immunogenicity <strong>of</strong><br />

Class II restricted T cell epitopes contained within the FPXprotein.<br />

The immunogenic, promiscuous epitope(s) are<br />

contained within the 14-amino acid carboxy-terminal region<br />

<strong>of</strong> the FPX-peptide, but none are found in the aminoterminal<br />

region comprising 10 amino acids as observed by in<br />

silico prediction and in vivo data. Peripheral blood mononuclear<br />

cells (PBMCs) from haplotyped donors were challenged<br />

with multiple rounds <strong>of</strong> FPX peptides (C-terminal and<br />

N-terminal) for 7–14 days followed by ELISPOT assays for T<br />

lymphocyte activation. HLA haplotype DRB1*0701/1301 was<br />

associated with an increased number <strong>of</strong> spot forming cells<br />

when challenged with the immunogenic C-terminal FPXpeptide<br />

(10.15 fold increase) and negligible with the nonimmunogenic<br />

N-terminal (1.38 fold increase). The same<br />

haplotype was associated with a high antibody response and<br />

a memory T cell response when the protein was administered<br />

in vivo as well as by in silico prediction. Ex vivo T cell<br />

priming assays can be potential tools for prediction <strong>of</strong>


S156 Abstracts<br />

immunogenicity <strong>of</strong> T cell epitopes. These assays enable<br />

selection <strong>of</strong> the right therapeutic in early development and<br />

determine which MHC haplotypes are predisposed to an<br />

immune response.<br />

doi:10.1016/j.clim.2007.03.083<br />

Su.43 Hepatitis B Virus Proteins Induce Activation <strong>of</strong><br />

hfgl2 Prothrombinase Gene Through c-Ets-2<br />

Transcription Factors<br />

Meifang Han, Doctor in Charge, Tongji Hospital, Huazhong<br />

University <strong>of</strong> Science and Technology, Department <strong>of</strong><br />

Infectious Disease, Wuhan, China, Dong Xi, Assistant<br />

Researcher, Department <strong>of</strong> Infectious Disease, Tongji<br />

Hospital, Huazhong University <strong>of</strong> Science and Technology,<br />

Wuhan, China, Weiming Yan, Assistant Researcher,<br />

Department <strong>of</strong> Infectious Disease, Tongji Hospital,<br />

Huazhong University <strong>of</strong> Science and Technology, Wuhan,<br />

China, Gary Levy, Pr<strong>of</strong>essor, Chief Physician, University<br />

Health Network, University <strong>of</strong> Toronto, Toronto, ON,<br />

Canada, Mingfeng Liu, Postdoctoral Researcher and<br />

Associate, University Health Network, University <strong>of</strong><br />

Toronto, Toronto, ON, Canada, Xiaoping Luo, Chief<br />

Physician, Department <strong>of</strong> Pediatrics <strong>of</strong> Tongji Hospital,<br />

Huazhong University <strong>of</strong> Science and Technology, Wuhan,<br />

China, Qin Ning, Chief Physician, Department <strong>of</strong> Infectious<br />

Disease, Tongji Hospital, Huazhong University <strong>of</strong> Science<br />

and Technology, Wuhan, China<br />

Objective: To determine the viral and host factors<br />

involved in the transcription <strong>of</strong> hfgl2 gene which was<br />

demonstrated to play pivoral role in human and experiment<br />

fulminant viral hepatitis. Methods. HBc, HBs or HBx expression<br />

plasmids were constructed and cotransfected with a<br />

hfgl2 luciferase report construct into eukaryotic cells.<br />

Results. Luciferase assay showed that HBc or HBx protein,<br />

but not HBs protein significantly enhanced hfgl2 transcription<br />

in both CHO and HepG2 cells. Series deletion assay <strong>of</strong><br />

hfgl2 gene promoter demonstrated that a strong regulatory<br />

domain from −712 to −568 was responsible for hfgl2 gene<br />

transcription in response to HBc or HBx proteins. By sitedirected<br />

mutagenesis, the overlapped cis-elements LEF/c-<br />

Ets in domain −712/−568 was demonstrated to play an<br />

important role in the regulation <strong>of</strong> hfgl2 gene in response<br />

to HBc protein, while another two cis-elements LEF/c-Ets<br />

and HSTF in the same domain were found to participate in<br />

hfgl2 transcription regulation in response to HBx protein.<br />

EMSA assays showed that HBc and HBx proteins did not<br />

directly induce hfgl2 gene activation, while an Ets family<br />

member c-Ets-2 bound the cognate cis-element in hfgl2<br />

promoter was responsible for induction <strong>of</strong> hfgl2 activation<br />

in response to viral proteins. Conclusion. Our study provides<br />

new insights in understanding the pathology <strong>of</strong> fulminant<br />

hepatic failure and the interaction between HBV virus and<br />

host gene expression. This work was supported by NSFC<br />

30225040, 30571643, 30672380, National Key Basic Research<br />

Program <strong>of</strong> China (2005CB522901, 2005CB522507) and <strong>Clinical</strong><br />

Subject Key Project from the Ministry <strong>of</strong> Health.<br />

doi:10.1016/j.clim.2007.03.084<br />

Su.44 Arthritogenic Simulation <strong>of</strong> Human Synovial<br />

Fibroblasts by TWEAK<br />

Timothy Zheng, Senior Scientist, Biogen Idec, Inc.,<br />

Cambridge, MA, Shawn Weng, Associate Scientist, Biogen<br />

Idec, Inc., Cambridge, MA, Suzanne Szak, Scientist II, Biogen<br />

Idec, Inc., Cambridge, MA, Linda Burkly, Distinguished<br />

Investigator, Biogen Idec, Inc., Cambridge, MA<br />

Synovial fibroblasts are critically involved in propagating<br />

joint inflammation in rheumatoid arthritis (RA) through the<br />

production <strong>of</strong> numerous arthritogenic mediators. The proinflammatory<br />

TNF ligand superfamily cytokine TWEAK has<br />

previously been shown to induce several chemokine/cytokines<br />

in human fibroblast cell types including synovciocytes. In this<br />

study, we seek to understand the involvement <strong>of</strong> TWEAK in<br />

human RA pathogenesis by studying its broad effect on human<br />

synovial fibroblasts. We found that synovial fluid TWEAK levels<br />

were elevated in RA patients when compared to those in OA<br />

patients, consistent with the notion that TWEAK may play a role<br />

in driving inflammatory response in the joints <strong>of</strong> RA patients.<br />

Using transcription pr<strong>of</strong>iling, we demonstrated that TWEAK<br />

consistently induced a large panel <strong>of</strong> genes, including many<br />

known arthritogenic mediators such as IL-1, IL-6, RANTES, ENA-<br />

78, IP-10 and MMPs. Interestingly, the potency <strong>of</strong> gene<br />

regulation by TWEAK when compared to TNF varied greatly<br />

amongst the four different donor cells, perhaps reflecting<br />

patient heterogeneity regarding the involvement <strong>of</strong> TWEAK visa-vie<br />

TNF in driving synoviocyte-mediated joint inflammation.<br />

Importantly, the production <strong>of</strong> TWEAK and TNF appeared to be<br />

independent <strong>of</strong> each other as neither TWEAK nor TNF induced<br />

or inhibited the expression <strong>of</strong> the other. When combined<br />

however, TWEAK and TNF synergically or additively induced the<br />

production <strong>of</strong> many other well-known arthritogenic mediators.<br />

Taken together, these results suggest that TWEAK and TNF may<br />

represent two concurrent upstream cytokines that regulate<br />

joint inflammation and damage in RA patients.<br />

doi:10.1016/j.clim.2007.03.085<br />

Su.45 IL-6/TH-17 Axis Plays a Crucial Role in the<br />

Generation <strong>of</strong> GPI-induced Arthritis<br />

Keiichi Iwanami, PhD Student, Graduate School <strong>of</strong><br />

Comprehensive Human Science, University <strong>of</strong> Tsukuba,<br />

Tsukuba, Japan, Asuka Inoue, Master Student, Graduate<br />

School <strong>of</strong> Comprehensive Human Science, University <strong>of</strong><br />

Tsukuba, Tsukuba, Japan, Isao Matsumoto, Instructor,<br />

Graduate School <strong>of</strong> Comprehensive Human Science,<br />

University <strong>of</strong> Tsukuba, Tsukuba, Japan, Mizuko Mamura,<br />

Research Fellow, Graduate School <strong>of</strong> Comprehensive Human<br />

Science, University <strong>of</strong> Tsukuba, Tsukuba, Japan, Yoko<br />

Watanabe, PhD Student, Graduate School <strong>of</strong> Comprehensive<br />

Human Science, University <strong>of</strong> Tsukuba, Tsukuba, Japan,<br />

Daisuke Goto, Instructor, Graduate School <strong>of</strong><br />

Comprehensive Human Science, University <strong>of</strong> Tsukuba,<br />

Tsukuba, Japan, Satoshi Ito, Instructor, Graduate School <strong>of</strong><br />

Comprehensive Human Science, University <strong>of</strong> Tsukuba,<br />

Tsukuba, Japan, Akito Tsutsumi, Assistant Pr<strong>of</strong>essor,<br />

Graduate School <strong>of</strong> Comprehensive Human Science,<br />

University <strong>of</strong> Tsukuba, Tsukuba, Japan, Takayuki Sumida,<br />

Pr<strong>of</strong>essor, Graduate School <strong>of</strong> Comprehensive Human<br />

Science, University <strong>of</strong> Tsukuba, Tsukuba, Japan


Abstracts<br />

Backgrounds: Glucose-6-phosphate isomerase (GPI)induced<br />

arthritis is a murine model <strong>of</strong> rheumatoid arthritis<br />

(RA). Although anti-CD4 antibodies (Abs) had therapeutic<br />

effect in this model, the pathogenicity <strong>of</strong> each CD4+ T cell<br />

lineage was uncertain. Here we undertook the present study<br />

to clarify GPI-specific T cell lineages and investigate their<br />

pathological and regulatory roles in the generation <strong>of</strong><br />

arthritis. [Methods] DBA/1 mice were immunized with<br />

300fÊg <strong>of</strong> GPI to induce arthritis. 1) CD4+ T cells and APCs<br />

were co-cultured with GPI for 24 h, and the supernatant was<br />

analyzed by cytokine ELISA. 2) Anti-IFNfÁ mAb or anti IL-17<br />

mAb was injected on day 7 to investigate arthritis development.<br />

3) A mAb to murine IL-6 receptor (MR16-1) was<br />

injected on days 0, 3 or 8 to investigate arthritis development.<br />

4) After the injection <strong>of</strong> MR16-1 on days 0 or 3,<br />

draining lymph nodes (DLNs) were harvested on day 7, and<br />

TH-17 cells were analyzed by flow cytometry. Results: 1)<br />

IFNfÁ and IL-17 were produced by GPI-specific CD4+ T cells.<br />

2) The administration <strong>of</strong> anti-IL-17 mAb on day 7<br />

significantly ameliorated arthritis (Pb0.01), whereas that<br />

<strong>of</strong> anti-IFNfÁ mAb exacerbated. 3) The administration <strong>of</strong><br />

MR16-1 on day 0 or 3 protected the induction <strong>of</strong> arthritis,<br />

and that <strong>of</strong> MR16-1 on day 8 significantly ameliorated<br />

arthritis (Pb0.05). 4) The differentiation <strong>of</strong> TH-17 in DLNs<br />

was markedly suppressed by MR16-1. Conclusions: IL-6 and<br />

TH-17 play an essential role in GPI-induced arthritis. Our<br />

study suggests that IL-6/TH-17 axis might be also involved<br />

in RA.<br />

doi:10.1016/j.clim.2007.03.086<br />

Su.46 The Serum Cytokine Pr<strong>of</strong>ile in Low-Stage<br />

Chronic Lymphocytic Leukemia is a 7th Hallmark <strong>of</strong><br />

Cancer<br />

Catherine Spier, Associate Pr<strong>of</strong>essor, Pathology, University<br />

<strong>of</strong> Arizona College <strong>of</strong> Medicine, Department <strong>of</strong> Pathology,<br />

Tucson, AZ, Christopher Riley, Graduate Student, University<br />

<strong>of</strong> Arizona College <strong>of</strong> Medicine, Arizona Cancer Center,<br />

Tucson, AZ, James Choi, Fellow, University <strong>of</strong> Arizona<br />

College <strong>of</strong> Medicine, Arizona Cancer Center, Tucson, AZ,<br />

Lawrence Cooke, Research Specialist, University <strong>of</strong> Arizona<br />

College <strong>of</strong> Medicine, Arizona Cancer Center, Tucson, AZ,<br />

Thomas Klinkhammer, Fellow, University <strong>of</strong> Arizona College<br />

<strong>of</strong> Medicine, Arizona Cancer Center, Tucson, AZ, Daruka<br />

Mahadevan, Associate Pr<strong>of</strong>essor, Medicine, University <strong>of</strong><br />

Arizona College <strong>of</strong> Medicine, Arizona Cancer Center,<br />

Tucson, AZ<br />

Background: In B cell Chronic Lymphocytic Leukemia (B-<br />

CLL) there are no validated screening, diagnostic or<br />

prognostic serum markers for early detection. Several<br />

aberrant cytokines have previously been identified individually<br />

in patients with B-CLL that are thought to enhance<br />

malignant cell survival. Methods: An IRB approved protocol<br />

was used to collect and examine 120 serum cytokines from<br />

26 Rai Stage 0/1 patients who were treatment naive and<br />

also from 4 normal volunteers (RayBiotech Cytokine Array,<br />

#AAH-CYT-G1000). Fold change was calculated for each<br />

patient, corrected for absolute lymphocyte count and a<br />

mean ± SD obtained for each cytokine for all patients.<br />

Results: This cytokine pr<strong>of</strong>ile identified all <strong>of</strong> the<br />

published cytokines associated with B-CLL cell survival<br />

(IL-1B, IL-2, IL-4, IL-6, IL-8, IL-10, TNFa, INFa or g, G-CSF<br />

or GM-CSF) or inhibition (IL-5, TGF-B). In this pr<strong>of</strong>ile the<br />

1st ranked is INF-g (14.46 fold) which is known to prevent<br />

apoptosis <strong>of</strong> B-CLL cells. The 2nd ranked is IGFBP-4 (14.22<br />

fold) and 3rd ranked is G-CSF (10.89 fold) which is known<br />

to decrease B-CLL cell apoptosis. Other cytokines <strong>of</strong> note<br />

are Flt-3 ligand (7.82 fold), SCF (6.76 fold) and SDF-1<br />

(6.54 fold). Conclusion: Thus, our cytokine pr<strong>of</strong>ile, that<br />

sampled 120 cytokines simultaneously, validates the existing<br />

data, lists the ctyokines by order <strong>of</strong> expression from<br />

highest to lowest and significantly adds to the list <strong>of</strong><br />

cytokines identified as important for B-CLL cell survival in<br />

low stage patients.<br />

doi:10.1016/j.clim.2007.03.087<br />

Su.47 High Sensitivity Multiplexed Immunoassays<br />

for Simultaneous Quantification <strong>of</strong> Key Cytokines in<br />

Human Samples<br />

Jehangir Mistry, PhD, LINCO (now part <strong>of</strong> Millipore), St.<br />

Charles, MO, Jiaxin Dong, PhD, LINCO (now part <strong>of</strong><br />

Millipore), St. Charles, MO, Terry Whitehead, NA, LINCO<br />

(now part <strong>of</strong> Millipore), St. Charles, MO, Shaoquan Ji,<br />

Research and Development Manager, LINCO (now part <strong>of</strong><br />

Millipore), St. Charles, MO, Hank Hwang, PhD, LINCO (now<br />

part <strong>of</strong> Millipore), St. Charles, MO<br />

Low levels <strong>of</strong> inflammation play a key role in etiology <strong>of</strong><br />

many disease states (e.g. atherosclerosis, allergy, cancer,<br />

diabetes). Understanding the role <strong>of</strong> inflammation in pathogenesis<br />

has been impeded because currently available cytokine<br />

assays are not sensitive enough to detect the<br />

differences between normal and subclinically inflammed<br />

states. Using the Luminex xMAP technology, we developed a<br />

highly sensitive, multiplexed immunoassay panel for simultaneously<br />

quantifying 13 human cytokines commonly involved<br />

in Th1/Th2 and inflammatory responses (IL-1β, IL-2,<br />

IL-4, IL-5, IL-6, IL-7, IL-8, IL-10, IL-12p70, IL-13, GMCSF,<br />

IFNγ and TNFα). The assays follow the typical bead-based<br />

sandwich format and use a simple protocol. Each antibody<br />

pair is highly specific, with no or negligible cross-reactivity<br />

to other analytes within the panel. All assays in the panel<br />

have a common dynamic range <strong>of</strong> 0.128 to 2000 pg/ml,<br />

wide enough for almost all sample types. Sensitivities <strong>of</strong> the<br />

assays are between 0.01 to 0.48 pg/ml with most analytes<br />

at approximately 0.1 pg/ml. Normal serum samples are 80<br />

to 100% detectable for most cytokines. The assay robustness<br />

is demonstrated by acceptable precisions (CV=2.2–<br />

14.3% for inter-assay; CV=3.1–5.9% for intra-assay), accuracy<br />

(93.0 to 112%), and linearity <strong>of</strong> dilution (102 to 113%)<br />

for serum or plasma samples. No serum/plasma dilution is<br />

required. The assays may be used for other sample types (e.<br />

g. cell culture supernatant, tissue/cell lysate from human<br />

origin). This novel, robust and highly sensitive assay panel<br />

serves as an accurate, reproducible and economic tool for<br />

simultaneously quantifying multiple cytokines in human<br />

samples.<br />

doi:10.1016/j.clim.2007.03.088<br />

S157


S158 Abstracts<br />

Su.48 Use <strong>of</strong> a 10-Cytokine Multiplex RT-PCR in<br />

Conjunction with a Corresponding 10-plex<br />

Fluorescent Bead Quantitative Protein<br />

Immunoassay to Reveal Expression Patterns in<br />

Mitogen-Stimulated Human Peripheral Leukocytes<br />

Cynthia Boardman, Development Scientist, Beckman<br />

Coulter Inc. Nucleic Acid Testing Business, Fullerton, CA,<br />

Cynthia Boardman, Development Scientist, Beckman<br />

Coulter Inc. Nucleic Acid Testing Business, Fullerton, CA, Yu<br />

Suen, Staff Development Scientist, Beckman Coulter Inc.<br />

Biomarker Discovery and Automation Center, Fullerton, CA,<br />

Scott Boyer, Genomics Group Manager, Beckman Coulter Inc.<br />

Nucleic Acid Testing Business, Fullerton, CA, Keith Roby,<br />

Product Manager, Beckman Coulter Inc. Biomarker Discovery<br />

and Automation Center, Fullerton, CA<br />

The ability to monitor the expression <strong>of</strong> mediators <strong>of</strong><br />

inflammation and immune response is critical to the study <strong>of</strong><br />

disease and pharmacodynamics. Quantitative protein immunoassays<br />

are one <strong>of</strong> the standard tools for the examination<br />

<strong>of</strong> cellular responses but reveal only part <strong>of</strong> the cellular<br />

story. We show here how the GenomeLab GeXP Genetic<br />

Analysis System can be used to corroborate protein studies<br />

and provide a more complete picture <strong>of</strong> cellular responses<br />

to effectors. Specifically, we have developed a multiplexed<br />

RT-PCR† assay which can analyze the mRNA expression<br />

pr<strong>of</strong>ile <strong>of</strong> ten Th1/Th2 cytokines simultaneously. The<br />

eXpress Designer s<strong>of</strong>tware included in the GeXP System<br />

was used to design multiplex PCR primers for the following:<br />

IFN-g, IL-1B, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, TNF-α and<br />

TNF-B; and the resulting multiplex was used to analyze<br />

changes in the mRNA expression pr<strong>of</strong>iles for these ten<br />

products in human peripheral leukocytes upon stimulation<br />

with LPS or PMA/PHA. The data were then used to corroborate<br />

results <strong>of</strong> a quantitative protein analysis performed<br />

on the matching cell supernatant samples using the<br />

FlowCytomix Multiplex Human Th1/Th2 10-Plex fluorescent<br />

bead immunoassay kit and analyzed on the Cytomics FC500<br />

MPL flow cytometry system. This dual-platform approach<br />

can be used to monitor the immune response effected by<br />

diseases and treatments at the level <strong>of</strong> both mRNA and<br />

protein expression.<br />

doi:10.1016/j.clim.2007.03.089<br />

Su.49 Dysregulated Inflammatory Cytokine<br />

Response in Lipopolysaccharide- and<br />

Peptidoglycan-Stimulated Monouclear Cells <strong>of</strong> a<br />

Patient with Blau Syndrome<br />

Sang Wook Son, Pr<strong>of</strong>essor, Department <strong>of</strong> Dermatology,<br />

Korea University College <strong>of</strong> Medicine, Ansan-si, Korea, Jin<br />

Lee, Doctor, Department <strong>of</strong> Pediatrics, Korea University<br />

College <strong>of</strong> Medicine, Ansan-si, Korea, Jae Eun Choi, Doctor,<br />

Department <strong>of</strong> Dermatology, Korea University College <strong>of</strong><br />

Medicine, Ansan-si, Korea, Il Hwan Kim, Pr<strong>of</strong>essor,<br />

Department <strong>of</strong> Dermatology, Korea University College <strong>of</strong><br />

Medicine, Ansan-si, Korea, Jeongwon Sohn, Pr<strong>of</strong>essor,<br />

Department <strong>of</strong> Biochemistry, Korea University College <strong>of</strong><br />

Medicine, Seoul, South Korea, Young Chul Kye, Pr<strong>of</strong>esor,<br />

Department <strong>of</strong> Dermatology, Korea University College <strong>of</strong><br />

Medicine, Ansan-si, Korea, Kwang Chul Lee, Pr<strong>of</strong>essor,<br />

Department <strong>of</strong> Pediatrics, Korea University College <strong>of</strong><br />

Medicine, Ansan-si, Korea, JungHwa Lee, Pr<strong>of</strong>essor,<br />

Department <strong>of</strong> Pediatrics, Korea University College <strong>of</strong><br />

Medicine, Ansan-si, Korea<br />

Inheritable Blau syndrome (BS) and sporadic early-onset<br />

sarcoidosis (EOS) share characteristic clinical features <strong>of</strong><br />

multi-organ granulomatous inflammation involving joints,<br />

skin and eyes. Multiple mutations in the caspase recruitment<br />

domain gene CARD15 were described in BS. EOS was<br />

diagnosed in a 5-month-old girl who initially had biopsyproven<br />

chronic non-caseating granulomatous papules on a<br />

whole body and later developed joint abnormalities and<br />

steroid-responsive hypercalcemia. Her 32-year-old mother<br />

was blind since she was about 5 years old and showed<br />

camptodactyly and eczematous skin lesions. All <strong>of</strong> the other<br />

mother side family members were healthy with no evidence<br />

<strong>of</strong> any skin, joint and eye abnormalities. DNA analysis<br />

demonstrated a missense mutation, 1000CNT (R334W), in<br />

the CARD15 gene in both the mother and the child,<br />

suggesting sporadic incidence <strong>of</strong> the mutation in the mother<br />

and its inheritance to her child. When peripheral blood<br />

mononuculear cells <strong>of</strong> the mother were stimulated by<br />

lipopolysaccharide and peptidoglycan, production <strong>of</strong> an<br />

anti-inflammatory cytokine, IL-10 was reduced compared<br />

to the normal control. In addition, induction <strong>of</strong> TNF-α<br />

production was reduced when stimulated with peptidoglycan<br />

alone. Basal levels <strong>of</strong> both cytokines in the mother were not<br />

different compared to the normal control. These findings<br />

indicate that BS and EOS are diseases <strong>of</strong> the same spectrum<br />

sharing the common etiology <strong>of</strong> CARD15 mutation and that a<br />

dysregulated anti- and pro-inflammatory cytokine response<br />

to the stimulation may lead to the chronic inflammatory<br />

manifestations in BS.<br />

doi:10.1016/j.clim.2007.03.090<br />

Su.50 Quantitative Evaluation <strong>of</strong> Interleukin-6,<br />

Interleukin-11 and Interleukin-17 in Healthy and<br />

Inflammatory Tissues <strong>of</strong> Gingival<br />

Hamid Reza, ArAB, Dentist, Periodontology Department,<br />

School <strong>of</strong> Dentistry and Mashhad Dental Research Center,<br />

Mashhad, Iran, Jalil Tavakkol-Afshari, Vice President for<br />

Research; Head, Department <strong>of</strong> Immunogenetics and Cell<br />

Culture, Bu-Ali Research Institute (BARI), Department <strong>of</strong><br />

Immunogenetics and Cell Culture, Mashhad, Iran, Zahra<br />

Baghani, Dentist, Periodontology Department, School <strong>of</strong><br />

Dentistry and Mashhad Dental Research Center, Mashhad,<br />

Iran, Nasrin Moheghi, Researcher, Bu-Ali Research Institute<br />

(BARI), Department <strong>of</strong> Immunogenetics, Mashhad, Iran<br />

Interleukin 11(IL-11), IL-17 and IL-6 are cytokines that<br />

modulate the inflammatory process and have not been<br />

assessed within normal or inflammed gingival tissues. Our<br />

purpose was comparing the concentrations <strong>of</strong> human IL-6, IL-<br />

11 and IL-17 within healthy and diseased human gingival<br />

tissues to determine their possible role in the initiation or<br />

progression <strong>of</strong> periodontal disease. Biopsies from healthy<br />

(non-hemorrhagic gingiva adjacent to the 3 mm, 4–5 mm and<br />

≥ 6 mm periodontal pockets) were studied. Interleukin-<br />

11, IL-17 and IL-6 concentrations were assessed within


Abstracts<br />

solubilized gingival biopsies by enzyme-linked immunosorbent<br />

assay. Data were analyzed by Kruskal–Wallis and Mann–<br />

Whitney U tests for finding significant correlation between<br />

groups. Interleukin 11, IL-17 and IL-6 concentrations were<br />

decreased within gingival adjacent to 4–5 mm sulcular depth<br />

but later increased within gingiva adjacent to ≥ 6mm<br />

pocket depths. The amount <strong>of</strong> IL-17 between healthy tissue<br />

and gingiva adjacent to ≥ 6 mm pockets and 4–5 mm and â<br />

‰¥ 6 mm pockets was significant (pb0.05). Interleukin-11,<br />

IL-6 and IL-17 concentrations were greatly correlated with<br />

sulcular depth. However, in most cases this difference was<br />

not statistically significant. Interleukin-6, IL-17, IL-11 concentrations<br />

were increased in the inflammatory gingival<br />

tissue adjacent to ≥ 6 mm pockets in comparison to<br />

healthy gingiva but only the concentration <strong>of</strong> IL-17 was<br />

significant (p=0.03).<br />

doi:10.1016/j.clim.2007.03.091<br />

Su.51 Inhibition <strong>of</strong> Experimental Tumor Metastasis<br />

by CpG Oligodeoxynucleotide-Pulsed Dendritic Cell<br />

Han-A Kim, Assistant, Department <strong>of</strong> Biological Sciences,<br />

Kwangju, South Korea, Hyun-Mi Ko, Department <strong>of</strong><br />

Biological Sciences, Kwangju, South Korea, Hern-Ku Lee,<br />

Pr<strong>of</strong>essor, Department <strong>of</strong> <strong>Immunology</strong>, Chonju, South Korea,<br />

Suhn-young Im, Pr<strong>of</strong>essor, Department <strong>of</strong> Biological<br />

Sciences, Kwangju, South Korea<br />

Dendritic cell (DC) pulsed with tumor antigen induced<br />

tumor-specific immune response. CpG ODN has been recognized<br />

as a powerful stimulant <strong>of</strong> DC. This study was<br />

designed to investigate whether CpG oligodeoxynucleotide<br />

(ODN)-pulsed DC could inhibit the experimental pulmonary<br />

metastasis <strong>of</strong> B16F10 murine melanoma. Treatment <strong>of</strong> CpG<br />

ODN intraperitoneally either before or after B16F10, it<br />

inhibited lung colonization <strong>of</strong> B16F10. Bone marrow-derived<br />

DC pulsed with B16F10 lysate inhibited the tumor metastasis.<br />

In this case, the inhibitory effect <strong>of</strong> mature DC was<br />

much more prominent than <strong>of</strong> immature DC. In addition, the<br />

inhibitory effect DC was further augmented when DC was<br />

stimulated with CpG ODN. The magnitude <strong>of</strong> experimental<br />

pulmonary metastasis was increased in IL-12 knock out<br />

(IL-12−/−) mice compared with their littermates. CpG ODN<br />

did not exert its metastasis-inhibiting activity in IL-12−/−<br />

mice. Moreover, DC originated from IL-12−/− mice do not<br />

inhibited metastasis even when the cells were stimulated<br />

with B16F10 lysate plus CpG ODN. In vitro, CpG ODN induced<br />

the gene expression and protein synthesis <strong>of</strong> IL-12 in DC.<br />

Tumor metastasis was inhibited by adoptive transfer <strong>of</strong><br />

splenic T cells from CpG ODN-treated mice. These results<br />

suggest that CpG ODN inhibited tumor metastasis through<br />

activating DC to produce IL-12, which resulted in the<br />

development <strong>of</strong> effector T cells.<br />

doi:10.1016/j.clim.2007.03.092<br />

Su.52 Regulation <strong>of</strong> the STAT3-Mediated Signaling by<br />

Daxx<br />

Kenji Sugiyama, Researcher, Department Molecular Biology,<br />

Nippon Boehringer Ingelheim Co. Kawanishi, Japan, Ryuta<br />

Muromoto, Postdoctoral Fellow, Department <strong>Immunology</strong>,<br />

Graduate School Pharmaceutical Science, Hokkaido<br />

University, Sapporo, Japan, Masato Ishida, Researcher,<br />

Department Molecular Biology, Nippon Boehringer<br />

Ingelheim Co, Kawanishi, Japan, Yuichi Sekine, Assosiate<br />

Pr<strong>of</strong>essor, Department <strong>Immunology</strong>, Graduate School<br />

Pharmaceutical Science, Hokkaido University, Sapporo,<br />

Japan, Kenji Oritani, Assistant Pr<strong>of</strong>essor, Department <strong>of</strong><br />

Hematol. and Oncology, Graduate School Med, Osaka<br />

University, Suita, Japan, Tadashi Matsuda, Pr<strong>of</strong>essor,<br />

Department <strong>Immunology</strong>, Graduate, School Pharmaceutical<br />

Science, Hokkaido University, Sapporo, Japan<br />

Signal transducer and activator <strong>of</strong> transcription 3 (STAT3)<br />

plays key roles in the intracellular signaling pathways <strong>of</strong> the<br />

interleukin (IL)-6 family <strong>of</strong> cytokines, which exhibits a<br />

diverse set <strong>of</strong> cellular responses, including cell proliferation<br />

and differentiation. Dysregulated IL-6/STAT3 signaling is<br />

involved in the pathogenesis <strong>of</strong> several diseases, for<br />

examples autoimmune diseases and tumors. Type I interferon<br />

(IFN) induces the expression <strong>of</strong> proapoptotic genes<br />

and has been used in the clinical treatment <strong>of</strong> several<br />

tumors. In the present study, we found that type I IFN<br />

suppressed IL-6/STAT3-mediated transcription and gene<br />

expression. Furthermore, a type I IFN-induced protein,<br />

Daxx, also suppressed STAT3-mediated transcriptional activation,<br />

while overexpression <strong>of</strong> Daxx inhibited IL-6/STAT3mediated<br />

gene expression. Importantly, small-interfering<br />

RNA-mediated reduction <strong>of</strong> Daxx expression enhanced IL-6/<br />

leukemia inhibitory factor (LIF)-induced STAT3-dependent<br />

transcription. Co-immunoprecipitation studies revealed a<br />

physical interaction between Daxx and STAT3 in transiently<br />

transfected 293T cells. We further found that Daxx and<br />

STAT3 were co-localized in the nucleus. These results<br />

indicate that Daxx may serve as a transcriptional regulator<br />

<strong>of</strong> type I IFN-mediated suppression <strong>of</strong> the IL-6/STAT3<br />

signaling pathway.<br />

doi:10.1016/j.clim.2007.03.093<br />

S159<br />

Su.53 Immunological Relationships between Heart<br />

Tissue and Non-Cardiac Organs in Murine<br />

Eosinophilc and Lymphocytic Myocarditis<br />

Masao Hirasawa, Doctor <strong>of</strong> Medicine, Third Division <strong>of</strong><br />

Internal Medicine, Osaka Medical College, Takatsuki-city,<br />

Osaka, Japan, Minoru Miyashita, Assistant, Department <strong>of</strong><br />

Neurosurgery, Osaka Medical College, Takatsuki-city, Osaka,<br />

Japan<br />

Background: Eosinophilic disorders usually develop into<br />

one <strong>of</strong> the factors leading to myocarditis. However,<br />

immunological interactions between heart tissue and noncardiac<br />

organs in such diseases are still unclear. In this study,<br />

the JAK/STAT signaling pathway in acute myocarditis was<br />

investigated, considering the immunological mechanism in<br />

non-cardiac organs. Methods: Adoptive transfer and Coxsackie<br />

B3 viral infection were performed to initiate<br />

eosinophilic and lymphocytic myocarditis in DBA/2 mice.<br />

Eosinophils, T cells, and JAK3/STAT6 mRNAs were examined<br />

by histological methods and RT-PCR. In the spleen, the ratios<br />

<strong>of</strong> CD4 to CD8 were compared between OVA-sensitized and


S160 Abstracts<br />

control mice. In the bone marrow, we attempted to detect<br />

CD34 by FACS analysis. Results: Eosinophilic myocarditis was<br />

observed in adoptive transferred mice, and marked lymphocytic<br />

myocarditis was developed in virus-infected mice. In<br />

both cases, JAK3s were usually seen in inflammatory heart<br />

lesions; in particular, the parallel expression <strong>of</strong> JAK3 and<br />

STAT6 mRNAs was seen in eosinophilic myocarditis. In the<br />

spleen, CD4/CD8 ratios were elevated in OVA-sensitized<br />

cases compared with controls (pb0.05), and the quantity <strong>of</strong><br />

JAK3 mRNA was higher in adoptive transfer cases than<br />

controls (pb0.05). Bone marrow cell analysis showed the<br />

role <strong>of</strong> CD34 in OVA-sensitized and normal control mice.<br />

Conclusion: These results suggested that (1) JAK/STAT<br />

signaling plays a role in myocardial damage in eosinophilic<br />

and viral myocarditis, (2) JAK3 also conveys immunological<br />

tolerance within a CD4-dominant environment in noncardiac<br />

organs, and (3) bone marrow cells <strong>of</strong> donor mice<br />

may have potential for regeneration therapy.<br />

doi:10.1016/j.clim.2007.03.094<br />

Su.54 Combined Pharmacodynamic and<br />

Transcriptome Pr<strong>of</strong>iling <strong>of</strong> Recombinant Human<br />

Interleukin-21 Responses in Patients with Stage IV<br />

Malignant Melanoma<br />

Dorthe Lundsgaard, Research Scientist, Project Manager,<br />

Novo Nordisk A/S, Maaloev, Denmark, Klaus Stensgaard<br />

Frederiksen, Senior Scientist, Novo Nordisk A/S, Bagsvaerd,<br />

Denmark, Lasse Tengbjerg Hansen, <strong>Clinical</strong> Scientist, Novo<br />

Nordisk A/S, Bagsvaerd, Denmark, Birte K. Skrumsager,<br />

Senior <strong>Clinical</strong> Pharmacologist, Novo Nordisk A/S,<br />

Bagsvaerd, Denmark, Ulrik Mouritzen, International Medical<br />

Officer, Novo Nordisk A/S, Bagsvaerd, Denmark, Grant<br />

McArthur, Pr<strong>of</strong>essor, Cancer Trials Australia, Peter<br />

MacCallum Cancer Centre, Melbourne, Australia, Ian D.<br />

Davis, Pr<strong>of</strong>essor, Cancer Trials Australia, Austin Hospital,<br />

Melbourne, Australia, Kresten Skak, Senior Scientist, Novo<br />

Nordisk A/S, Bagsvaerd, Denmark<br />

Interleukin-21 is a pleiotropic class I cytokine that<br />

activates CD8+ T cells and NK cells. In a phase 1 dose<br />

escalation trial <strong>of</strong> intravenous IL-21 administered in two<br />

different dose regimens, the pharmacodynamic effects<br />

were monitored by multi-analyte pr<strong>of</strong>iling <strong>of</strong> 90 serum<br />

proteins and global transcriptional pr<strong>of</strong>iling <strong>of</strong> peripheral<br />

CD8+ T cells. A total <strong>of</strong> 17 serum proteins were observed to<br />

increase at least 2-fold upon IL-21 treatment whereas 4<br />

proteins were down-regulated. Each dose regimen displayed<br />

a distinct pattern <strong>of</strong> regulation, reflecting differences<br />

in pharmacodynamic effects. Among the up-regulated<br />

proteins were acute-phase proteins, chemokines, and<br />

cytokines, e.g. CRP, TNF-α, MCP-1, and IL-18. In CD8+ T<br />

cells, isolated within the first week <strong>of</strong> treatment, a total <strong>of</strong><br />

471 probe sets were found to be significantly differentially<br />

regulated. In particular, effector genes such as granzyme A<br />

and interferon-gamma as well as chemokine receptor<br />

genes, such as CXCR3 and CXCR6, were clearly up-regulated<br />

upon IL-21 treatment. These observations may indicate<br />

stimulation <strong>of</strong> both effector functions and motility <strong>of</strong> CD8+<br />

T cells. Moreover, suggestive <strong>of</strong> increased proliferation <strong>of</strong><br />

CD8+ T cells, levels <strong>of</strong> several DNA replication and cell cycle<br />

related genes, including PCNA and Ki-67, were induced in a<br />

dose-dependent manner. The presented data demonstrate<br />

that administration <strong>of</strong> IL-21 in patients with stage IV<br />

malignant melanoma induces a variety <strong>of</strong> pharmacodynamic<br />

responses related to immunological activation that warrants<br />

further clinical investigations <strong>of</strong> this cytokine.<br />

doi:10.1016/j.clim.2007.03.095<br />

Su.55 Differential Expression on the TNF-RI, IL-1RI,<br />

IL-6R Membrane and Soluble Receptors and NF-kB,<br />

AP-1 Activation by In Vitro Proteasome Inhibition in<br />

U937 cells<br />

Alejandro Bravo-Cuéllar, MD, Instituto Mexicano del Seguro<br />

Social, Centro de Investigacion Biomedica de Occidente<br />

Division de Inmunologia, Guadalajara, Mexico, Pablo C.<br />

Ortiz-Lazare, Chemistry Biologist and Pharmacology,<br />

Instituto Mexicano del Seguro Social, Centro de<br />

Investigacion Biomedica de Occidente Division de<br />

Inmunologia, Guadalajara, Mexico, Jorge R. Dominguez-<br />

Rodriguez, Biologist, Instituto Mexicano del Seguro Social,<br />

Centro de Investigacion Biomedica de Occidente Division de<br />

Inmunologia, G, Mexico, Jose M. Lerma-Diaz, MD,<br />

Universidad de Guadalajara, CUALTOS, Tepatitlan de<br />

Morelos, Mexico, Georgina Hernandez-Flores, Biologist,<br />

Instituto Mexicano del Seguro Social, Centro de<br />

Investigacion Biomedica de Occidente Division de<br />

Inmunologia, Guadalajara, Mexico, Piedad C. Gomez-<br />

Contreras, Chemistry Biologist and Pharmacology, Instituto<br />

Mexicano del Seguro Social, Centro de Investigacion<br />

Biomedica de Occidente Division de Inmunologia,<br />

Guadalajara, Mexico, Martha Barba-Barajas, Chemistry<br />

Biologist and Pharmacology, Instituto Mexicano del Seguro<br />

Social, Centro de Investigacion Biomedica de Occidente<br />

Division de Inmunologia, Guadalajara, Mexico,<br />

Luis F. Jave-Suarez, Biologist, Instituto Mexicano del Seguro<br />

Social, Centro de Investigacion Biomedica de Occidente<br />

Division de Inmunologia, Guadalajara, Mexico, Adriana C.<br />

Aguilar-Lemarrroy, Biologist, Instituto Mexicano del Seguro<br />

Social, Centro de Investigacion Biomedica de Occidente<br />

Division de Inmunologia, Guadalajara, Mexico<br />

Proteasome is a large multimeric protease complex,<br />

which plays a vital role in several cellular functions, such<br />

as: proteins degradation, regulation <strong>of</strong> cyclins and transcription<br />

factors, its also related with antigens degradation. On<br />

the other hand several stimuli such as proinflammatory<br />

cytokines, free oxygen radicals, components <strong>of</strong> the bacteria<br />

cellular wall, can trigger NF-kB activation, which normally<br />

exits as an inactive form, characterized by its union at the<br />

trimolecular complex called IkB. NF-kB activation begins by<br />

phosphorylation, ubiquitination and degradation <strong>of</strong> IkB<br />

complex in the proteasome. Once NF-kB transcriptional<br />

factor is liberated from its natural IkB inhibitor, it can reach<br />

the nucleus and link to promoters regions <strong>of</strong> genes that codify<br />

for TNF-α, IL-1β, IL-6, membrane receptors, and adhesion<br />

molecules. Monocyte/macrophages, lymphocytes and others<br />

cells generate these cytokines that increase the inflammatory<br />

process. TNF-α, IL-1β, IL-6 have been associated with<br />

different pathologies such as rheumatoid arthritis, inflammatory<br />

intestinal disease, shock septic. In this study, we


Abstracts<br />

investigated the in vitro effects <strong>of</strong> the proteasome inhibitor<br />

MG132 on TNF-RI, IL-1RI and IL-6R, the activation <strong>of</strong> NF-kB<br />

and AP-1 in U937 cells. Cell viability was evaluated by flow<br />

cytometry using cellular annexin V binding. Proteasome<br />

inhibition increases the release <strong>of</strong> TNF-RI and IL-1RI from<br />

U937 cells and decreases their cell surface expression. In<br />

other way, the MG132 increases the cell surface expression <strong>of</strong><br />

IL-6R and decreases their liberation. The proteasome<br />

inhibitor, also, led to increased LPS+PMA-induced AP-1<br />

activation, and attenuated LPS+PMA-induced IkB degradation<br />

resulting in abolished NF-kB activation.<br />

doi:10.1016/j.clim.2007.03.096<br />

Su.56 Mucosal and Systemic T Cell Responses to<br />

Cry1Ac Protoxin from Bacillus Thuringiensis<br />

Aldo Arturo Reséndiz Albor, Cell Biology, Universidad<br />

Nacional Autonoma de Mexico, Mexico, Leticia<br />

Moreno-Fierros, Cell Biology, Inmunidad en Mucosas<br />

UBIMED, FES-Iztacala, Universidad Nacional Autonoma de<br />

Mexico, Mexico<br />

Cry1Ac protoxin (pCry1Ac) from Bacillus thuringiensis is a<br />

potent immunogen with mucosal and systemic adjuvant<br />

properties when administered to mice by systemic or<br />

mucosal routes although its mechanism <strong>of</strong> action remains<br />

unknown. Thus, in this work, we investigated in vivo and in<br />

vitro the proportion <strong>of</strong> T cells producing IL-2, IL-4, IL-10,<br />

and IFN-g by intracellular cytokine staining on CD3+ T<br />

lymphocytes freshly isolated from spleen and Peyer's<br />

patches (PP) following intraperitoneal immunization <strong>of</strong><br />

pCry1Ac. Furthermore, we evaluated the induction <strong>of</strong><br />

CD69 and CD25 in T cell subsets after a single intraperitoneal<br />

administration <strong>of</strong> pCry1Ac in spleen and PP as a<br />

parameter for detecting T cell activation following by in<br />

vitro stimuli with pCry1Ac. In control mice we observed that<br />

PP present T cells spontaneously producing IFN-g while<br />

cytokine production was not detected in spleen lymphocytes.<br />

Addition <strong>of</strong> pCry1Ac to splenic and PP T cell cultures<br />

promoted a Th1 pr<strong>of</strong>ile <strong>of</strong> cytokines with high levels <strong>of</strong> IL-2<br />

and IFN-g without IL-4. We also found that intraperitoneal<br />

immunization with pCry1Ac increase <strong>of</strong> T lymphocytes<br />

expressing CD69, IL-4 in the spleen and CD69, IL-2 and<br />

IFN-g in PP. pCry1Ac stimulation led to rapid expression <strong>of</strong><br />

CD69 on both TCD4+ and B cells between 4 h post incubation<br />

in spleen and PP which remained stable throughout the 72 h<br />

culture period. The effects elicited by pCry1Ac on cytokine<br />

pr<strong>of</strong>iles are different between lymphocytes from the spleen<br />

and PP suggesting that their phenotypic and functional<br />

differences may influence the immunomodulatory effects <strong>of</strong><br />

the protein.<br />

doi:10.1016/j.clim.2007.03.097<br />

Su.57 Effects <strong>of</strong> a Novel Mucosal Adjuvant AT-1002<br />

on Immune Responses in Mice Immunized<br />

Intranasally with Ovalbumin<br />

Amir Tamiz, Director <strong>of</strong> Chemistry, Alba Therapeutics,<br />

Baltimore, MD, Niranjan Pandey, Director <strong>of</strong> Biology, Alba<br />

Therapeutics, Baltimore, MD, Blake Paterson, CEO, Alba<br />

Therapeutics, Baltimore, MD, Sefik Alkan, Vice President,<br />

Alba Therapeutics, Baltimore, MD<br />

Antigens, adjuvants and delivery systems are the main<br />

essential components to the development <strong>of</strong> efficient<br />

vaccines. The purpose <strong>of</strong> this study was to evaluate AT-<br />

1002 HCl, a six-mer synthetic peptide, FCIGRL, derived<br />

from Zonula Occludens Toxin (cholera's second toxin) as<br />

an effective mucosal delivery agent that is expected to<br />

enhance antigen passage through the mucosal/endothelial<br />

barriers and hence stimulate antigen-specific immune<br />

responses. In this study, female Balb/c mice were<br />

immunized intranasally with Ova and AT-1002 simultaneously,<br />

four times in 21-day intervals and complete<br />

antibody responses to Ova were measured in the serum<br />

and in vaginal washes using quantitative Ova-specific<br />

ELISA. Effects <strong>of</strong> AT-1002 stimulation on IgG subclasses<br />

(IgG1, IgG2a, IgG2b, IgA and IgE) were studied. Ovaspecific<br />

T cell responses were also measured at 5–8 days<br />

after the last immunization in both the spleen and the<br />

lung <strong>of</strong> all mice. We found that mice immunized with Ova<br />

in the presence <strong>of</strong> AT-1002 exhibit significantly higher<br />

antibody titers than those induced by immunization with<br />

antigen alone. In addition, we found that AT-1002 and<br />

Cholera Toxin induced similar patterns <strong>of</strong> Ova-specific IgG<br />

subclasses. In summary, our study suggests that the tight<br />

junction modulator AT-1002 might provide a novel<br />

approach to vaccination.<br />

doi:10.1016/j.clim.2007.03.098<br />

S161<br />

Su.58 Dual Targeting <strong>of</strong> CCR2 and CX3CR1 in an<br />

Arterial Injury Model <strong>of</strong> Vascular Inflammation<br />

Maya Jerath, Fellow, University <strong>of</strong> North Carolina,<br />

Department <strong>of</strong> Medicine, Chapel Hill, NC, Peng Liu,<br />

Research Associate, University <strong>of</strong> North Carolina, Thurston<br />

Arthritis Research Center, Chapel Hill, NC, Mauricio Rojas,<br />

Research Instructor, University <strong>of</strong> North Carolina,<br />

Department <strong>of</strong> Medicine, Chapel Hill, NC, Mary Struthers,<br />

Research Fellow, Merck Research Laboratories, Department<br />

<strong>of</strong> <strong>Immunology</strong>, Rahway, NJ, Julie Demartino, Senior<br />

Director, Merck Research Laboratories, Department <strong>of</strong><br />

<strong>Immunology</strong>, Rahway, NJ, Kathryn Lyons, Senior Research<br />

Associate, Merck Research Laboratories, Department <strong>of</strong><br />

Medicinal Chemistry, Rahway, NJ, Ruoqi Peng, Senior<br />

Research Immunologist, Merck Research Laboratories,<br />

Department <strong>of</strong> <strong>Immunology</strong>, Rahway, NJ, Lihu Yang,<br />

Director, Merck Research Laboratories, Department <strong>of</strong><br />

Medicinal Chemistry, Rahway, NJ, Dhaval Patel, Pr<strong>of</strong>essor,<br />

The University <strong>of</strong> North Carolina, Department <strong>of</strong> Medicine,<br />

Chapel Hill, NC<br />

Rationale: CCR2 and CX3CR1 are chemokine receptors<br />

differentially expressed on monocyte subsets in diverse<br />

species from rodents to man, and each receptor has been<br />

individually hypothesized to play a role in inflammatory<br />

cell trafficking to atherosclerotic lesions. We hypothesized<br />

that CCR2 and CX3CR1 had non-redundant effects in<br />

vascular inflammation, and sought to determine their<br />

combined effect. Methods: We used a murine vascular<br />

injury model in which both CCR2 KO and CX3CR1 KO have


S162 Abstracts<br />

shown a partial block in the injury response. Experimental<br />

arms consisted <strong>of</strong> C57Bl/6 WT mice, CX3CR1 KO mice, WT<br />

mice given a small molecule CCR2 antagonist (MRL-677,<br />

Merck & Co.) and CX3CR1 KO mice given the same<br />

antagonist. Vessels were harvested at 14 days analyzed<br />

by histology, morphometry and immunohistochemistry.<br />

Results: Blocking CCR2 with MRL-677 resulted in a 39%<br />

decrease in the vascular injury response (n=10, p=NS).<br />

CX3CR1 deficient mice had similar injury to WT animals<br />

(n=8, p=NS). Mice in which both CCR2 and CX3CR1<br />

pathways were targeted (CX3CR1 KO mice given MRL-677)<br />

had an 86% decrease in the injury response (n=6, p=0.002).<br />

Conclusions: In this study, we have shown that blocking<br />

CCR2 with a low molecular weight antagonist ameliorates<br />

the inflammatory response to vascular injury. Simultaneous<br />

targeting <strong>of</strong> both CCR2 and CX3CR1 has complementary and<br />

perhaps synergistic effects on the development <strong>of</strong> the<br />

vascular inflammatory response. Our results imply that<br />

CCR2 and CX3CR1 have independent, non-overlapping roles<br />

in vascular inflammation and that approaches to target<br />

both may have more beneficial effects than targeting each<br />

one separately.<br />

doi:10.1016/j.clim.2007.03.099<br />

Su.59 Cytokine and Chemokine Production by NK<br />

Cells Activated by Monokines IL-12, IL-18<br />

Following Crosslinking <strong>of</strong> CD16 and Modulation<br />

by KIR<br />

Zaheed Husain, Junior Investigator/Instructor, CBR<br />

Institute for Biomedical Research, Harvard Medical<br />

School, Boston, MA, Devendra Dubey, Investigator, CBR<br />

Institute for Biomedical Research, Harvard Medical<br />

School, Boston, MA, Shampa Das, Research Fellow, CBR<br />

Institute for Biomedical Research, Harvard Medical<br />

School, Boston, MA, Chester Alper, Pr<strong>of</strong>essor, CBR<br />

Institute for Biomedical Research, Harvard Medical<br />

School, Boston, MA<br />

NK cells express both activation and inhibitory receptors.<br />

Activating receptors play a central role in cytokine and<br />

chemokine production that is modulated by inhibitory<br />

receptors. We used high throughput, antibody-based cytokine<br />

chips to study the levels <strong>of</strong> secreted cytokines/<br />

chemokines. Data was collected to study the rate <strong>of</strong><br />

cytokines/chemokines production and secretion by human<br />

NK cells activated by monokines IL-12 and IL-18 following<br />

crosslinking <strong>of</strong> CD16. We also determined the effects <strong>of</strong> KIR<br />

on the production <strong>of</strong> these cytokines/chemokines. Several<br />

characteristics were observed: (1) IFN-g and IL-8 showed a<br />

linear increase with time, (2) the level <strong>of</strong> chemokines<br />

peaked within 2 hrs and reverted back to control levels<br />

within 24 hrs. The synthesis <strong>of</strong> chemokines was significantly<br />

higher (3.5 to 4.5 fold) than cytokines such as IFN-γ. Several<br />

cytokines and growth factors were inhibited by KIR3DL1. The<br />

mechanism <strong>of</strong> the increase in the levels <strong>of</strong> IFN-γ and IL-8 was<br />

further investigated by analyzing intracellular cytokine<br />

production using 4-color FACS analysis. Exposure <strong>of</strong> IFN-γ<br />

to NK cells induced a rapid increase in the intracellular<br />

expression <strong>of</strong> IL-8 suggesting that increase in IL-8 production<br />

could be due to the de novo synthesis <strong>of</strong> IL-8 by NK cells. This<br />

study shows that IL-12/IL-18/CD16 induced activation <strong>of</strong> NK<br />

cells triggers several signaling pathways that regulate<br />

adaptive immunity.<br />

doi:10.1016/j.clim.2007.03.100<br />

Su.60 Differential Effect <strong>of</strong> NK Activation Molecules<br />

CD16 and 2B4 on Cytokine Production: Significant<br />

Inhibition <strong>of</strong> GM-CSF but not IFN-g, TNF-α or<br />

Cytotoxic Activity<br />

Zaheed Husain, Junior Investigator/Instructor, CBR Institute<br />

for Biomedical Research, Harvard Medical School, Boston,<br />

MA, Shampa Das, Research Fellow, CBR Institute for<br />

Biomedical Research, Harvard Medical School, Boston, MA,<br />

Chester Alper, Pr<strong>of</strong>essor, CBR Institute for Biomedical<br />

Research, Harvard Medical School, Boston, MA, Devendra<br />

Dubey, Investigator, CBR Institute for Biomedical Research,<br />

Harvard Medical School, Boston, MA<br />

NK cells express several activation and inhibitory<br />

receptors which are involved in cytokine, chemokine and<br />

growth factor production and cytotoxic activity against<br />

virus infected cells, tumor or cells deficient in HLA class I<br />

expression. Crosslinking <strong>of</strong> CD16 or 2B4 significantly<br />

increases IFN-γ, TNF-α and GM-CSF production, and<br />

cytotoxic activity in redirected lysis assay. It is not known<br />

how co-crosslinking <strong>of</strong> these two receptors affect cytokine<br />

production and cytotoxic activity. To investigate this<br />

question we used antibody mediated crosslinking <strong>of</strong> CD16<br />

and 2B4 molecules on IL-2 activated NK cells activity in<br />

redirected lysis assay. Separate engagement <strong>of</strong> CD16 or 2B4<br />

strongly enhanced cytotoxic activity, IFN-γ, TNF-α and GM-<br />

CSF production. These are inhibited by co-crosslinking <strong>of</strong><br />

KIR receptors. Co-engagement <strong>of</strong> these activation receptors<br />

strongly inhibited the production <strong>of</strong> GM-CSF (N90%) but not<br />

IFN-γ or TNF-α production or cytotoxic activity suggesting<br />

that the pathway for GM-CSF production is separate from<br />

IFN-γ or TNF-α production. This further suggests that CD16<br />

and 2B4 may function together to optimize cytokine<br />

production.<br />

doi:10.1016/j.clim.2007.03.101<br />

Su.61 Augmenting CD8+ T Cell Responses with<br />

IL-15/sIL-15Rα Complexes<br />

Mark Rubinstein, Postdoctoral Fellow, University <strong>of</strong><br />

California, San Diego, La Jolla, CA, Jonathan Sprent,<br />

Pr<strong>of</strong>essor, Garvan Institute <strong>of</strong> Medical Research,<br />

Darlinghurst, Australia, Ananda Goldrath, Assistant<br />

Pr<strong>of</strong>essor, University <strong>of</strong> California, San Diego,<br />

La Jolla, CA<br />

Administration <strong>of</strong> IL-15 has shown efficacy in augmenting<br />

CD8+ T cell responses following vaccination. However,<br />

soluble IL-15 is limited in vivo by half life and poor biological<br />

activity. Recently, we showed that the biological activity <strong>of</strong><br />

soluble IL-15 is much improved after interaction with<br />

recombinant soluble IL-15Rα. After injection, soluble IL-<br />

15/IL-15Rα complexes rapidly induce strong and selective<br />

expansion <strong>of</strong> memory CD8+ cells and natural killer cells.


Abstracts<br />

Surprisingly, we also find that soluble IL-15/IL-15Rα complexes<br />

can drive conversion <strong>of</strong> naïve Tcells to memory Tcells<br />

in vivo, without administration <strong>of</strong> antigen. This conversion is<br />

associated with an acquisition <strong>of</strong> effector function. Our<br />

results suggest the therapeutic potential <strong>of</strong> IL-15 could be<br />

considerably enhanced by administration <strong>of</strong> preformed<br />

soluble IL-15/IL-15Rα complexes.<br />

doi:10.1016/j.clim.2007.03.102<br />

Su.62 Evaluation <strong>of</strong> IL-1 Beta Secretion Level <strong>of</strong><br />

Human Osteoblasts and Morphology <strong>of</strong> These Cells<br />

Adjunct to Gray MTA, White MTA, Portland Cement<br />

and IRM as Retr<strong>of</strong>illing<br />

Jalil Tavakkol Afshari, Vice President for Research,<br />

Immunogenetics Department, Bu-Ali Research Institute,<br />

Mashhad University <strong>of</strong> Medical Sciences, Mashhad, Iran,<br />

Navid Aghasizadeh, Dentist, School <strong>of</strong> Dentistry, Mashhad<br />

University <strong>of</strong> Medical Sciences, Mashhad, Iran, Maryam<br />

Bidar, Dentist, School <strong>of</strong> Dentistry, Mashhad University<br />

<strong>of</strong> Medical Sciences, Mashhad, Iran, Mohammad Ali<br />

Fahmidekar, Researcher, Immunogenetics Department,<br />

Bu-Ali Research Institute, Mashhad University <strong>of</strong><br />

Medical Sciences, Mashhad, Iran, Mohammad Zarabi,<br />

Dentist, School <strong>of</strong> Dentistry, Mashhad University <strong>of</strong><br />

Medical Sciences, Mashhad, Iran, Azam Brook,<br />

Researcher, Immunogenetics Department, Bu-Ali Research<br />

Institute, Mashhad University <strong>of</strong> Medical Sciences,<br />

Mashhad, Iran<br />

Ostetoblasts and ligament periodontal cells are the<br />

essential cells for wound repair after root-end resections<br />

and perforation repair. Cell attachment and spread on the<br />

surface materials and evaluation <strong>of</strong> the cell secretory<br />

function are primary phases in normal cell function. The<br />

aim <strong>of</strong> this study was evaluating osteoblasts (MG-63) function<br />

morphologically and IL-1 β level adjacent to gray MTA, white<br />

MTA, Portland cement and IRM, as filling materials for rootend<br />

fillings and repairing perforations. Human osteoblasts<br />

(MG-63) were grown in RPMI-1640 medium. Materials were<br />

mixed and seeded in appropriate plates. Cells were added<br />

after primary setting <strong>of</strong> materials. They were surveyed in<br />

days 1, 3 and 7. The amount <strong>of</strong> IL-1 β in each specimen was<br />

measured by ELISA. Morphological outcomes showed that<br />

after 7 days, a large amount <strong>of</strong> osteoblasts adjacent to gray<br />

and white MTA had a nice attachment. Cells adjacent to<br />

Portland cement were round and mostly separated from the<br />

surface. All cells were round and separated from the plate<br />

surface adjacent to IRM. Levels <strong>of</strong> IL-1 β adjacent to gray and<br />

white MTA were significantly more than to IRM and Portland<br />

cement (P=0.00). Amount <strong>of</strong> IL-1 β was not significantly<br />

different adjacent to gray and white MTA. Osteoblasts<br />

adjacent to gray and white MTA perform more appropriate<br />

response in comparison to Portland cement and IRM. Therefore,<br />

noticing the color <strong>of</strong> white MTA, it can be substituted by<br />

gray MTA in places with more cosmetic effects. It also<br />

appears that Portland cement is not a good substitute for<br />

MTA.<br />

doi:10.1016/j.clim.2007.03.103<br />

Su.63 Blockade <strong>of</strong> CD40−CD40 Ligand Interactions<br />

Attenuates Skin Fibrosis and Autoimmunity in the<br />

Tight-skin Mouse<br />

Kazuhiro Komura, MD, Department <strong>of</strong> Dermatology,<br />

Nagasaki University Graduate School <strong>of</strong> Biomedical<br />

Sciences, Nagasaki, Japan, Manabu Fujimoto, MD,<br />

Department <strong>of</strong> Dermatology, Kanazawa University Graduate<br />

School <strong>of</strong> Medical Science, Kanazawa, Japan, Minoru<br />

Hasegawa, MD, Department <strong>of</strong> Dermatology, Kanazawa<br />

University Graduate School <strong>of</strong> Medical Science, Kanazawa,<br />

Japan, Shinichi Sato, MD, Department <strong>of</strong> Dermatology,<br />

Nagasaki University Graduate School <strong>of</strong> Biomedical<br />

Sciences, Nagasaki, Japan<br />

The tight-skin (TSK/+) mouse, a mouse model for human<br />

systemic sclerosis (SSc), develops cutaneous fibrosis and<br />

autoimmunity. Molecular mechanisms <strong>of</strong> fibrosis and autoimmune<br />

responses in both TSK/+ mice and SSc remain<br />

unknown. In this study, we investigated the role <strong>of</strong> CD40/<br />

CD40 ligand (CD40L) interactions in TSK/+ mice and SSc. The<br />

blockade <strong>of</strong> CD40/CD40L interactions by anti-CD40L mAb<br />

reduced the development <strong>of</strong> cutaneous fibrosis (65%) and<br />

autoantibody production in TSK/+ mice. Furthermore, abnormal<br />

CD40/CD40L interactions in TSK/+ mice was suggested<br />

by up-regulated CD40L expression on CD4+ T<br />

lymphocytes, elevated plasma CD40L levels, and hyperresponsiveness<br />

to CD40 stimulation <strong>of</strong> B cells and fibroblasts. In<br />

addition, augmented CD40/CD40L signaling in human SSc was<br />

also suggested by elevated levels <strong>of</strong> circulating soluble<br />

CD40L and circulating soluble CD40. In addition to the<br />

reports from other groups, the results <strong>of</strong> the current studies<br />

suggest that CD40/CD40L interactions can be therapeutical<br />

targets in SSc.<br />

doi:10.1016/j.clim.2007.03.104<br />

S163<br />

Su.64 Intracellular Expression <strong>of</strong> TNF-α and IFN-γ<br />

by Peripheral Blood Mononuclear Cells in a Family<br />

with Rosacea<br />

Gabriel Andres Luna-Baca, MD, Institute <strong>of</strong> Ophthalmology<br />

Conde de Valenciana, Mexico City, Mexico, Concepcion<br />

Santacruz-Valdes, MD, Cornea and Refractive Surgery<br />

Department, Institute <strong>of</strong> Ophthalmology Conde de<br />

Valenciana, Mexico City, Mexico, Maria Carmen<br />

Jimenez-Martinez, MD, PhD, Research Unit, Institute <strong>of</strong><br />

Ophthalmology Conde de Valenciana, Mexico City, Mexico<br />

Introduction: Rosacea is a chronic dermatologic condition<br />

that predominantly affects the central aspect <strong>of</strong> the face.<br />

Although the pathophysiology <strong>of</strong> rosacea remains unknown,<br />

there is increasing agreement that inflammatory mediators<br />

are operative in this disorder. Recent studies have shown the<br />

role <strong>of</strong> oxidative stress and antioxidative system disorders in<br />

Rosacea. Despite IFN-γ and TNF-α have been involved in the<br />

pathogenesis <strong>of</strong> many dermatologic diseases driving to the<br />

exacerbation <strong>of</strong> inflammatory and prooxidative responses,<br />

there is no evidence that these mediators are involved in<br />

rosacea. Objective: To evaluate the expression <strong>of</strong> IFN-γ and<br />

TNF-α by peripheral blood mononuclear cells (PBMC) in a<br />

family with Rosacea (RF). Methods: CD4, CD8, CD19, CD56,<br />

IFN-γ, TNF-α, IL-1β, IL-4 and IL-6 expression was determined


S164 Abstracts<br />

by flow cytometry on PBMC from RF and also on PBMC from<br />

healthy controls (HC). Results. IFN-γ+cells in RF-1=38.5 vs.<br />

15 HC (p=0.0016); TNF-α+cells in RF-1=55 vs. 3.2 HC (p=<br />

0.0015); IFN-γ+cells in RF-2=32.6 vs. 8.1 HC (p=0.026);<br />

TNF-α+cells in RF-2=34.6 vs. 1.5 HC (p=0.0056); IFN-γ+cells<br />

in RF-2=32.6 vs. 8.1 HC (p=0.026); TNF-α+cells in RF-2=34.6<br />

vs. 1.5 HC (p=0.0056); IFN-γ+cells in RF-3=24.2 vs. 8.3 HC<br />

(p=0.04); TNF-α+cells in RF-3=32.2 vs. 3.1 HC (p=0.0046).<br />

CONCLUSIONS. Peripheral blood mononuclear cells from a<br />

Rosacea family have significant increase in the expression <strong>of</strong><br />

IFN-γ+ and TNF-α+ cells. IFN-γ and TNF-α may have a role in<br />

the inflammatory process <strong>of</strong> rosacea stimulating the release<br />

<strong>of</strong> reactive oxygen species in macrophages and neutrophils<br />

causing damage to facial follicles in persons with Rosacea.<br />

doi:10.1016/j.clim.2007.03.105<br />

Su.65 Does Quality <strong>of</strong> Life in Cutaneous Lupus<br />

Erythematosus Correlate with Disease Severity?<br />

Elizabeth Gaines, Pre-doctoral Research Fellow,<br />

University <strong>of</strong> Pennsylvania, Department <strong>of</strong> Dermatology,<br />

Philadelphia, PA<br />

The purpose <strong>of</strong> this study was to assess the correlation<br />

between cutaneous lupus erythematosus (CLE) disease<br />

severity, as measured by the CLASI, and quality-<strong>of</strong>-life<br />

(QoL), as measured by the Skindex-29. This was a prospective,<br />

longitudinal study from baseline (Day 0) to Day 56, done<br />

at a University hospital cutaneous autoimmunity clinic. Eight<br />

patients with biopsy-confirmed CLE (4 DLE, 2 SCLE, 2 DLE/<br />

SLE) completed the study. At each visit, patients completed<br />

the CLE-modified Skindex-29. Using scales from 0-10, they<br />

also assessed their general and skin health, and their pain,<br />

itch, and fatigue. The PI evaluated disease severity, with the<br />

CLASI, and the patient s global skin health using a scale from<br />

0−10. There was a significant improvement in total Skindex<br />

score over time (t=2.36, pb0.05). There was a moderate<br />

correlation (r=0.59, p=0.12, n=8) between change in activity<br />

(CLASI), and change in skin symptoms (Skindex). Neither<br />

change in emotion or function correlated with change in<br />

activity. There was no correlation between any Skindex<br />

subscale with damage. Skindex scores for patients with SCLE<br />

were, on average, lower and changed less than Skindex<br />

scores for patients with DLE. While both the CLASI activity<br />

score and the modified Skindex total score improved<br />

significantly over time, the correlation between the two<br />

scores was poor. Different elements <strong>of</strong> skin health may be<br />

captured by each index. Given the different disease courses<br />

and likelihood <strong>of</strong> damage, skin-related QoL may differ across<br />

different subtypes <strong>of</strong> CLE. Preliminary analysis <strong>of</strong> data from<br />

25 additional CLE patients supports these conclusions.<br />

doi:10.1016/j.clim.2007.03.106<br />

Su.66 Association Between the Polymorphism <strong>of</strong><br />

TGF-β1 Gene Promoter (−509CNT) and Idiopathic<br />

Chronic Urticaria<br />

Sara Hosseini-Farahabadi, Dr.; Researcher, Bu-Ali Research<br />

Institute (BARI), Department <strong>of</strong> Immunogenetics, Mashhad,<br />

Iran, Jalil Tavakkol-Afshari, Vice President <strong>of</strong> Research;<br />

Head, Department <strong>of</strong> Immunogenetics and Cell Culture,<br />

Bu-Ali Research Institute (BARI), Department <strong>of</strong><br />

Immunogenetics and Cell Culture, Mashhad, Iran, Rashin<br />

Ganjali, Researcher, Bu-Ali Research Institute (BARI),<br />

Department <strong>of</strong> Immunogenetics, Mashhad, Iran, Javad<br />

Ghaffari, Mazandaran University <strong>of</strong> Medical Sciences,<br />

Department <strong>of</strong> Pediatrics, Sari, Iran, Houshang Rafatpanah,<br />

Ghaem Medical Center, Department <strong>of</strong> Allergy and<br />

<strong>Immunology</strong>, Mashhad, Iran, Reza Farid-Hosseini, Head,<br />

Department <strong>of</strong> Allergy and <strong>Immunology</strong>, Ghaem Medical<br />

Center, Department <strong>of</strong> Allergy and <strong>Immunology</strong>,<br />

Mashhad, Iran<br />

Background: Idiopathic Chronic Urticaria (ICU), the most<br />

common form (70–80%) <strong>of</strong> chronic urticaria is supposed to<br />

have immune basis causes. It is speculated that the promoter<br />

polymorphism <strong>of</strong> TGF-β1 gene may be involved in ICU. This<br />

condition is thought to affect at least 0.1% <strong>of</strong> the population<br />

and <strong>of</strong>ten it can be severe and difficult to treat. Materials<br />

and Methods: A total <strong>of</strong> 40 patients with ICU and 41 normal<br />

subjects were studied. DNA was extracted from whole blood<br />

and TGF-β1 promoter −509CNT polymorphism was determined<br />

by PCR-RFLP method. Results: Out <strong>of</strong> the 40 patients<br />

with ICU, 11 (27.5%) had CC, 26 (65%) had CTand 3 (7.5%) had<br />

TT genotypes. A higher proportion <strong>of</strong> case subjects with the C<br />

allele (CT type or CC type) was found compared with the T<br />

allele. Conclusion: These results do suggest an influence <strong>of</strong><br />

genetic variability at the promoter <strong>of</strong> TGF-β1 gene<br />

(−509CNT) on the occurrence <strong>of</strong> ICU. This polymorphism<br />

has been shown as a useful genetic change in our study.<br />

Further work is required to confirm this result.<br />

doi:10.1016/j.clim.2007.03.107<br />

Su.67 Investigation <strong>of</strong> the Mechanisms <strong>of</strong> Pressure<br />

Ulcers Using a Cyclical Cutaneous<br />

Ischemic−reperfusion Injury Model<br />

Yuki Saito, Graduate Student, Kanazawa University,<br />

Dermatology, Kanazawa, Japan, Minoru Hasegawa, Assistant<br />

pr<strong>of</strong>essor, Kanazawa University, Dermatology, Kanazawa,<br />

Japan, Manabu Fujimoto, Associate pr<strong>of</strong>essor, Kanazawa<br />

university, Dermatology, Kanazawa, Japan, Kazuhiko<br />

Takehara, Pr<strong>of</strong>essor, Kanazawa university, Dermatology,<br />

Kanazawa, Japan<br />

The formation <strong>of</strong> pressure ulcers is considered to be<br />

derived from multifactorial factors. Among them, the occurrence<br />

<strong>of</strong> cycles <strong>of</strong> ischemic–reperfusion is an especially<br />

significant contributing factor. This study assessed immunological<br />

mechanism <strong>of</strong> a reproducible murine model <strong>of</strong><br />

ischemic–reperfusion injury by the external application <strong>of</strong><br />

magnets. Dorsal skin was gently pulled and placed between<br />

two round ceramic magnetic plates. A single ischemic–<br />

reperfusion cycle (I–R cycle) consisted <strong>of</strong> a 12 hours <strong>of</strong><br />

magnet placement followed by a 12 hours <strong>of</strong> release or rest.<br />

Three cycles were performed to induce pressure ulcer formation.<br />

A marked edema was observed after an I–R cycle (day<br />

1) and skin ulcer developed until day 6 (3 days post<br />

treatment). Then, skin ulcers were healed until day 20 in<br />

all mice. In the lesional skin, the infiltration <strong>of</strong> neutrophils<br />

and macrophages, and tumor necrosis factor-alpha expression


Abstracts<br />

were most prominent at day 2. Then, inducible nitric oxide<br />

synthase (iNOS) expression was augmented at day 3. While<br />

nitric oxide is considered to be involved in ischemic–<br />

reperfusion injury, iNOS is a critical enzyme for the synthesis<br />

<strong>of</strong> nitric oxide. iNOS is produced by various cells including<br />

neutrophils and macrophages stimulated with proinflammatory<br />

cytokines such as tumor necrosis factor-alpha. Therefore,<br />

our findings suggest that iNOS production from<br />

infiltrated neutrophils and macrophages stimulated by<br />

tumor necrosis factor-alpha contributes to the development<br />

<strong>of</strong> skin ulcer in this model. We propose that this cyclical<br />

cutaneous ischemic–reperfusion injury model is useful for<br />

investigating the mechanism or developing novel therapies <strong>of</strong><br />

pressure ulcers.<br />

doi:10.1016/j.clim.2007.03.109<br />

Su.68 CD19 Expression in B cells is Important for<br />

Suppression <strong>of</strong> Contact Hypersensitivity<br />

Manabu Fujimoto, Associate Pr<strong>of</strong>essor, Department <strong>of</strong><br />

Dermatology, Kanzawa University, Kanazawa Ishikawa,<br />

Japan, Rei Watanabe, Student, Department <strong>of</strong> Dermatology,<br />

University <strong>of</strong> Tokyo, Bunkyo Tokyo, Japan, Thomas Tedder,<br />

Pr<strong>of</strong>essor, Department <strong>of</strong> <strong>Immunology</strong>, Duke University<br />

Medical Center, Durham, NC, Kunihiko Tamaki, Pr<strong>of</strong>essor,<br />

Department <strong>of</strong> Dermatology, University <strong>of</strong> Tokyo, Bunkyo<br />

Tokyo, Japan<br />

Contact hypersensitivity (CHS) is a cutaneous immune<br />

reaction mediated mainly by Ag-specific effector Tcells, and<br />

regarded as a model for Th1/Tc1-mediated inflammation.<br />

However, recent reports have suggested pivotal roles <strong>of</strong> B<br />

cells in CHS. CD19 is a member <strong>of</strong> the Ig superfamily expressed<br />

on B cells and follicular dendritic cells, and serves as a<br />

positive B cell response regulator which defines signaling<br />

thresholds critical for B cell responses. In the current study,<br />

we assessed the role <strong>of</strong> the B cell-specific surface molecule<br />

CD19 on the development <strong>of</strong> CHS through examining CD19deficient<br />

mice. Although CD19-deficient mice are hyposensitive<br />

to a variety <strong>of</strong> transmembrane signals, CD19 loss resulted<br />

in increased and prolonged reaction <strong>of</strong> CHS, suggesting an<br />

inhibitory role <strong>of</strong> CD19 expression in the etiology <strong>of</strong> CHS.<br />

Sensitized draining lymph nodes and elicited ear lesions from<br />

CD19-deficient mice exhibited Th1/Tc1-shifted cytokine<br />

pr<strong>of</strong>ile with increased IFN-fÁ expression and decreased IL-<br />

10 expression. Adoptive transfer experiments revealed that<br />

CD19 expression in recipient mice was required for optimal<br />

suppression <strong>of</strong> CHS response, indicating its role in elicitation<br />

phase. Furthermore, spleen B-2 cells, especially marginal<br />

zone B cells, from wild type mice were able to normalize<br />

exaggerated CHS reactions in CD19-deficient mice. Thus,<br />

CD19 expression in B cells is critical for termination <strong>of</strong> CHS<br />

responses, possibly through the function <strong>of</strong> regulatory B cells.<br />

doi:10.1016/j.clim.2007.03.111<br />

Su.69 Automated Enumeration <strong>of</strong> Skin Mast Cells by<br />

Laser Scanning Cytometry<br />

Esther Trueblood, Director <strong>of</strong> Pathology, Amgen, Inc./<br />

Pathology, Seattle, WA, Stephen Z., Scientist, Amgen, Inc./<br />

<strong>Clinical</strong> <strong>Immunology</strong>, Thousand Oaks, CA, Andrea Ita, Senior<br />

Scientist, Amgen, Inc./Inflammation, Thousand Oaks, CA,<br />

John Ferbas, Director Medical Sciences, Amgen, Inc./<br />

<strong>Clinical</strong> <strong>Immunology</strong>, Thousand Oaks, CA, James Chung,<br />

Medical Sciences Director, Amgen, Inc./Early development,<br />

Thousand Oaks, CA, Gordon Ng, Scientific Director, Amgen,<br />

Inc./Inflammation, Thousand Oaks, CA, Gloria Juan,<br />

Senior Scientist, Amgen, Inc./<strong>Clinical</strong> <strong>Immunology</strong>,<br />

Thousand Oaks, CA<br />

Mast cells (MCs) are bone marrow-derived immune competent<br />

cells dependent on stem cell factor for survival,<br />

chemotaxis, functional activation, and adhesion to connective<br />

tissue matrix. After activation, MCs can release a<br />

variety <strong>of</strong> mediators including histamine, tryptase, leukotrienes,<br />

prostaglandins, contributing to anaphylactoid,<br />

allergic and inflammatory processes. MCs are also associated<br />

with wound repair together with fibroblasts and<br />

other immune cells. The current effort presents a novel<br />

application <strong>of</strong> Laser Scanning Cytometry (LSC) to study the<br />

time course <strong>of</strong> mast cell recruitment in a skin woundhealing<br />

model. A full-thickness incisional wound model was<br />

developed in the mouse. A skin biopsy was taken on day 8<br />

after wounding in animals treated with control or an antimurine<br />

c-Kit antibody (ACK2) that blocks SCF. A two-step<br />

protocol on the LSC was created to automate quantitation<br />

<strong>of</strong> wound-adjacent MCs. The first step executed a lowresolution<br />

optical scan that defined the position and area <strong>of</strong><br />

each biopsy on the slide. The second step consisted <strong>of</strong> a<br />

high-resolution scan that captured sequential 20× image<br />

fields <strong>of</strong> the entire biopsy and stitched the images together<br />

into a mosaic. Computer analysis segregated stored image<br />

events into discrete populations, and ultimately MCs. MCs<br />

counts were obtained from a defined region around the<br />

wound site. ACK2 treatment reduced wound-activated Mast<br />

Cell expansion as measured by LSC (T-test; P=0.0094) and<br />

consistent with the manual counts. This method <strong>of</strong><br />

quantitation allows implementation <strong>of</strong> a more efficient,<br />

objective and precise method to score effects on MCs which<br />

are tissue resident and difficult to analyze.<br />

doi:10.1016/j.clim.2007.03.112<br />

S165<br />

Su.70 Kawasaki Disease: Presentation <strong>of</strong> One Case<br />

Carlos Torres-Loza, Immunoallergist, West National Medical<br />

Center, UMAE-IMSS, Guadalajara, Mexico<br />

Kawasaki disease (KD) is the most common vasculitis <strong>of</strong><br />

childhood and may well be the most common vasculitis at any<br />

age. The aim <strong>of</strong> this work is to present a case with an atypical<br />

clinical presentation or perhaps to consider other immunoinflamatory<br />

syndrome and to share this clinical experience<br />

with other immunologists. Ketzalli is a girl <strong>of</strong> 5 years old who<br />

started abruptly with abdominal pain, remittent fever <strong>of</strong>ten<br />

up 38.5 °C and scarlatiniform and multiform cutaneous rash<br />

and irritability by 2–3 days. After that acute period, we<br />

treated with metamizole as antithermic. After that period,<br />

we realized that our patient started to feel better but, with<br />

changes in her lips characterized by dry, swollen and vertically<br />

cracked lips and diffusely without strawberry tongue.<br />

Actually, by that time we realized both erythema <strong>of</strong> palms


S166 Abstracts<br />

and soles and skin desquamation. However, after 1–2 days<br />

more we see how her fingertips begin to peel and how one<br />

typical lesion <strong>of</strong> vasculitis appear on skin <strong>of</strong> her right<br />

fingertip. Ketzalli was seen by several specialist in pediatrics<br />

including, infectologist, dermatologist, immunoallergist and<br />

not all <strong>of</strong> them were in agreement about diagnosis <strong>of</strong><br />

Kawasaki disease. Giving benefit <strong>of</strong> the doubt, Ketzalli was<br />

treated with IVIgG to a doses <strong>of</strong> 1.5 g/kg in one doses. After<br />

two weeks <strong>of</strong> such clinical event a normal echocardiogram<br />

was reported. As far as we know does not exist other clinical<br />

problem with such skin desquamation and peeling hence to<br />

share this case with other immunologists.<br />

doi:10.1016/j.clim.2007.03.113<br />

Su.72 CD4+CD25+ Regulatory T Cells Abrogate<br />

Psoriatic-like Skin Lesions in a Murine Model <strong>of</strong><br />

Psoriasis, Whereas TNFa Blockade Exacerbates the<br />

Lesions<br />

Hak-Ling Ma, Senior Research Scientist II, Wyeth Research,<br />

Inflammation Department, Cambridge, MA<br />

Psoriasis is a chronic inflammatory skin disease that is due<br />

to rapid epidermal proliferation with mononuclear cell infiltrates.<br />

Recent studies demonstrated that transferring<br />

naïve CD4+ T cells into scid/scid mice was able to induce<br />

psoriatic like lesions in the recipient mice. This indicates<br />

that T cells play a central role in the pathogenesis <strong>of</strong> the<br />

disease. By adoptive transferring naïve T cells that lacked<br />

Tregs, we found that recipient mice experienced close to<br />

100% incidence <strong>of</strong> disease with clinical symptoms (i.e. skin<br />

erythema and scaling) and histological features (i.e. epidermal<br />

thickening, rete pegs and hyperplasia <strong>of</strong> epidermis and<br />

keratinocytes) resembling that <strong>of</strong> human psoriasis. We also<br />

found that co-administration <strong>of</strong> Tregs with naïve T cells<br />

significantly decreased the incidence and severity <strong>of</strong><br />

psoriatic-like skin lesions. It has been shown that cytokines<br />

such as IL-12, TNFa, IL-22, and IL-17 play important roles in<br />

disease progression. TNFa antagonists have been used in<br />

treatment <strong>of</strong> psoriasis, however, they can also exacerbate<br />

skin lesions in some individuals. Interestingly, in our studies,<br />

soluble murine TNFRIIFc exacerbated disease in comparison<br />

to isotype control treated mice. Overall, our results suggest<br />

that Tregs inhibit whereas soluble murine TNFRIIFc exacerbates<br />

skin inflammation in this model.<br />

doi:10.1016/j.clim.2007.03.114<br />

Su.74 Investigator-Initiated Trial <strong>of</strong> Efalizumab for<br />

Atopic Dermatitis: A Pro<strong>of</strong>-<strong>of</strong>-Concept Study in<br />

Adults<br />

Melissa Magliocco, Assistant Pr<strong>of</strong>essor <strong>of</strong> Medicine,<br />

UMDNJ-Robert Wood Johnson Medical School, Department<br />

<strong>of</strong> Medicine, New Brunswick, NJ, Irina Lipets, Assistant<br />

Program Manager, UMDNJ-Robert Wood Johnson Medical<br />

School, New Brunswick, NJ, Viktor Dombrovskiy, Assistant<br />

Pr<strong>of</strong>essor <strong>of</strong> Medicine, UMDNJ-Robert Wood Johnson<br />

Medical School, New Brunswick, NJ, Alice Gottlieb,<br />

Pr<strong>of</strong>essor and Chair <strong>of</strong> the Department <strong>of</strong> Dermatology,<br />

Tufts-New England Medical Center, Boston, MA<br />

Background: Atopic dermatitis (AD) is an autoimmune<br />

skin disorder mediated acutely by predominantly activated<br />

Th2 T cells and in chronic stages by activated Th1 cells.<br />

There is a need for safe, effective AD treatment. Efalizumab<br />

is a humanized monoclonal IgG1 antibody that binds the<br />

CD11a subunit <strong>of</strong> LFA-1, inhibiting T cell activation,<br />

reactivation, and emigration into skin. Its FDA approval for<br />

psoriasis suggests that it would be effective and safe for AD,<br />

because both diseases exhibit Th1 cell predominance in<br />

lesional skin. Objective: To determine whether targeting<br />

CD11a on T cells decreases the clinical activity <strong>of</strong> AD.<br />

Methods: This was a single-arm, open-label, pilot study.<br />

Fifteen subjects with at least a “moderate” Investigator's<br />

Global Assessment (IGA) score were recruited. Subjects<br />

were excluded if they received photo/systemic therapy<br />

within 1 week <strong>of</strong> baseline. Subjects received a subcutaneous<br />

injection <strong>of</strong> efalizumab weekly. Efficacy was assessed<br />

monthly for 6 months using EASI (Eczema Area and Severity<br />

Index), IGA, a subjective pruritus assessment scale, and<br />

photography. Safety was evaluated by physical examination<br />

and laboratory tests. Results: Three subjects completed the<br />

study. Changes in EASI, IGA, and pruritus scores at 6 months<br />

compared with baseline were analyzed. The mean decrease<br />

in EASI was 15.3% (95% CI −3.4%–34.0%), whereas that <strong>of</strong> the<br />

IGA was 10.6% (95% CI 1.0%–20.2%). Reduction in mean<br />

pruritus score was 29.5% (95% CI 45.0%–104.1%) Few adverse<br />

events were noted, including infection. Conclusion: Treatment<br />

<strong>of</strong> AD with efalizumab resulted in a significant<br />

improvement in IGA, but did not significantly affect EASI<br />

or pruritus.<br />

doi:10.1016/j.clim.2007.03.115<br />

Su.75 Demodex Folliculorum and Chronic<br />

Granulomatous Disease<br />

Anete Sevciovic Grumach, University <strong>of</strong> Sao Paulo, São<br />

Paulo, Brazil, Rosemeire Navickas Constantino-Silva, BSc,<br />

Department <strong>of</strong> Dermatology, São Paulo, Brazil, Marcelo<br />

Mentha Nico, Department <strong>of</strong> Dermatology, University <strong>of</strong> São<br />

Paulo, São Paulo, Brazil, Alexandre Correa, BSc,<br />

Department <strong>of</strong> Dermatology, University <strong>of</strong> Sao Paulo, Sao<br />

Paulo, MRF, Correa, Hospital Mandaqui, Sao Paulo, Brazil,<br />

Israel Zeckcer, Department <strong>of</strong> Dermatology, University <strong>of</strong><br />

Sao Paulo, São Paulo, Brazil, Joao Bosco Oliveira,<br />

Department <strong>of</strong> Dermatology, University <strong>of</strong> Sao Paulo, São<br />

Paulo, Brazil, Alberto Jose da Silva Duarte, Pr<strong>of</strong>essor,<br />

Department <strong>of</strong> Dermatology, São Paulo, Brazil, Dewton<br />

Moraes-Vasconcelos, Department <strong>of</strong> Dermatology, São<br />

Paulo, Brazil<br />

Background: Chronic Granulomatous Disease (CGD) is a<br />

primary immunodeficiency with a defect on oxidative<br />

metabolism <strong>of</strong> phagocytes. Demodex are also recognized as<br />

mites <strong>of</strong> the face and they live in the human pillous follicles.<br />

They affect aging people or immunosuppressed patients. No<br />

previous report has found those mites in primary immunodeficiencies.<br />

Objective: To report the clinical and laboratorial<br />

manifestations <strong>of</strong> a patient with CGD and Demodex<br />

folliculorum. Case report: A 1 year and 4 months old, male,<br />

born to a non-consanguineous family presenting with facial<br />

“suppurative” lesions was referred to our outpatient group in


Abstracts<br />

order to evaluate the recurrent localized lesions on his face.<br />

He was treated with several antibiotics with no clinical<br />

improvement. He had been hospitalized twice for bronchopneumonia<br />

(2X) and severe anemia receiving blood transfusions.<br />

The facial secretion was evaluated in the microscope<br />

and Demodex was identified. Considering the cause <strong>of</strong><br />

referral, DHR was performed and CGD was confirmed.<br />

Conclusion: The follicular pitiriasis or demodicidosis was<br />

first described in secondary immunodeficiencies such as<br />

leukemia or after kidney transplantation. This patient<br />

represents the first case report associating Demodex with a<br />

primary immunodeficiency (CGD).<br />

doi:10.1016/j.clim.2007.03.116<br />

Su.76 Immune Response Using Dialyzable Leukocyte<br />

Extract in the Treatment <strong>of</strong> Herpetic Keratitis<br />

Gabriel Andres Luna-Baca, MD, Research Unit, Institute <strong>of</strong><br />

Ophthalmology Conde de Valenciana, Mexico City, Mexico,<br />

Marisela Linares, Master <strong>of</strong> Science, Research Unit, Institute<br />

<strong>of</strong> Ophthalmology Conde de Valenciana, Mexico City,<br />

Mexico, Concepcion Santacruz-Valdes, MD, Cornea and<br />

Refractive Surgery Department, Institute <strong>of</strong> Ophthalmology<br />

Conde de Valenciana, Mexico City, Mexico, Raul Chavez, MD,<br />

PhD, Laboratorio de Inmunologia, Departamento de<br />

Bioquimica, Universidad Nacional Autonoma de Mexico,<br />

Mexico City, Mexico, Gustavo Aguilar-Velazquez, MD,<br />

Research Unit, Institute <strong>of</strong> Ophthalmology Conde de<br />

Valenciana, Mexico City, Mexico, Mayra Perez-Tapia, PhD,<br />

Department <strong>of</strong> <strong>Immunology</strong>, National School <strong>of</strong> Biological<br />

Sciences, ENCB-IPN, Mexico City, Mexico, Iris Estrada-<br />

Garcia, PhD, Department <strong>of</strong> <strong>Immunology</strong>, National School <strong>of</strong><br />

Biological Sciences, ENCB-IPN, Mexico City, Mexico, Sergio<br />

Estrada-Parra, PhD, Department <strong>of</strong> <strong>Immunology</strong>, National<br />

School <strong>of</strong> Biological Sciences, ENCB-IPN, Mexico City,<br />

Mexico, Maria Carmen Jimenez-Martinez, MD, PhD,<br />

Research Unit, Institute <strong>of</strong> Ophthalmology Conde de<br />

Valenciana, Mexico City, Mexico<br />

Introduction: T lymphocytes are involved in herpetic<br />

keratitis (HK) producing IFNg. Transfer factor (TF) has the<br />

ability to modulate the immune response, probably due to<br />

IFNg production. TF has been used in the treatment <strong>of</strong> viral<br />

infections like herpes simplex and zoster. Objective: To<br />

evaluate the cytokine expression by peripheral blood mononuclear<br />

cells (PBMC) from HK patients (p) treated with TF.<br />

Methods. CD4, CD8, IFNg, IL-4, perforin and granzyme expression<br />

was determined by flow cytometry on PBMC from<br />

HK-p, before (BTF) and after (ATF) treatment with TF and<br />

also from healthy controls (HC). Results: Percentage (%) CD4+<br />

IFNg+T cells in HK-p BTF=15 vs. 9 HC (p=0.028) vs. 33 ATF; %<br />

CD4+IL-4+Tcells in HK-p BTF=20 vs. 12 HC vs. 6.5 ATF; %CD8+<br />

IFNg+T cells in HK-p BTF=23 vs. 25 HC vs. 50 ATF (p=0.0002<br />

vs. ATF, p=0.005 vs. HC); %CD8+IL-4+T cells in HK-p BTF=7<br />

vs. 10 HC vs. 10 ATF; %CD8+IL-4+T cells=7 vs. 23 CD8+IFNg+T<br />

cells in HK-p BTF (p=0.005); %CD8+IL-4+T cells=10 vs. 50<br />

CD8+IFNg+T cells in HK-p ATF (p=0.003); %CD4+Granzyme+<br />

T cells in HK-p BTF=9.5 vs. 9.4 HC vs. 10.7 ATF; %CD4+<br />

Perforin+T cells in HK-p BTF=5.7 vs. 1.1 HC vs. 9 ATF; %CD4+<br />

Granzyme+Perforin+T cells in HK-p BTF=8.1 vs. 2.7 HC vs.<br />

10.8 ATF; %CD8+Granzyme+T cells in HK-p BTF=12 vs. 26 HC<br />

(p=0.012) vs. 10.7 ATF (p=0.008 vs. IS); %CD8+Perforin+Tcells<br />

in HK-p BTF=8.5 vs. 4.8 HC vs. 13.2 ATF; %CD8+ Granzyme+<br />

Perforin+T cells in HK-p BTF=32.8 vs. 33.5 HC vs. 37.1 ATF.<br />

Conclusions: CD8+lymphocytes from HK-p predominantly produce<br />

IFNg after treatment with TF. This could explain the better<br />

outcomes by a cytotoxic Tcell response in the studied patients.<br />

doi:10.1016/j.clim.2007.03.117<br />

Su.78 Gene Expression Pr<strong>of</strong>iling <strong>of</strong> Non-infectious<br />

Uveitis Patients Using Pathway Specific cDNA<br />

Microarray Analysis<br />

Zhuqing Li, Staff Scientist, Lab <strong>Immunology</strong>, NEI, NIH,<br />

Bethesda, MD, Sankaranarayana Mahesh, Postdoctoral<br />

Fellow, Laboratory <strong>of</strong> <strong>Immunology</strong>, NEI, NIH, Bethesda, MD,<br />

Baoying Liu, Fellow, <strong>Immunology</strong>, NEI, NIH, Bethesda, MD,<br />

Grace Clarke, Physician, <strong>Immunology</strong> Lab, NEI, NIH,<br />

Bethesda, MD, Wee Kiak Lim, Fellow, <strong>Immunology</strong>, NEI, NIH,<br />

Bethesda, MD, Robert Nussenblatt, Physician, <strong>Immunology</strong><br />

Lab, NEI, NIH, Bethesda, MD<br />

To study gene expression pr<strong>of</strong>iling <strong>of</strong> leukocytes from<br />

peripheral blood <strong>of</strong> patients with non-infectious uveitis,<br />

peripheral blood mononuclear cells (PBMCs) were isolated<br />

from 50 patients with clinically characterized non-infectious<br />

intermediate uveitis. Total RNA from PBMCs were extracted<br />

and quantified. Biotinylated probes were generated from<br />

total RNAs by incorporating biotinylated dUTP into synthesized<br />

cDNAs using a T7-polymerase linear amplification<br />

strategy. RNAs isolated from 20 healthy donors were pooled<br />

and served as normal control. Probes were hybridized to a<br />

pathway-specific cDNA microarray chip that contains some<br />

400 autoimmune and inflammatory response related genes.<br />

Signals for specific binding were recorded by a CCD camera<br />

and gene expression pr<strong>of</strong>iling was analyzed using GEArray<br />

Suite s<strong>of</strong>tware. Despite tremendous heterogeneity <strong>of</strong> gene<br />

expression among patients and normal controls, there were<br />

clear differential gene expression patterns comparing those<br />

from patients to those from normal control. Although there<br />

was more than 80% <strong>of</strong> overlap <strong>of</strong> gene expression, some 15% <strong>of</strong><br />

the genes were differentially expressed among patients<br />

compared to the normal donors. Several cytokine, chemokine<br />

or chemokine receptor genes were consistently more highly<br />

expressed among uveitis patients compared to the normal<br />

control. There seems less consistency in terms <strong>of</strong> downregulated<br />

genes among patients when compared to normal<br />

control. We show here that pathway specific cDNA microarray<br />

can be an efficient and economic strategy to pr<strong>of</strong>iling uveitis<br />

patients at the genomic level. Although preliminary and<br />

needing further confirmation, our data revealed several<br />

immune response genes that may be implicated in presumably<br />

autoimmune originated non-infectious uveitis.<br />

doi:10.1016/j.clim.2007.03.118<br />

S167<br />

Su.79 A Novel Model <strong>of</strong> MDP-induced Ocular<br />

Inflammation is Dependent on CARD15/NOD2 But<br />

Not on Interleukin-1<br />

Holly Rosenzweig, Postdoctoral Fellow, Oregon Health and<br />

Science University, Portland, OR, Tammy Martin, Research


S168 Abstracts<br />

Assistant Pr<strong>of</strong>essor, Oregon Health and Science University,<br />

Department <strong>of</strong> Ophthalmology, Portland, OR, Michael<br />

Davey, Pr<strong>of</strong>essor, VA Medical Center, Portland, OR, Monica<br />

Jann, Research Assistant, VA Medical Center, Portland, OR,<br />

Steve Planck, Research Associate Pr<strong>of</strong>essor, Oregon Health<br />

and Science University, Department <strong>of</strong> Ophthalmology,<br />

Portland, OR, Kelley Goodwin, Student Research Assistant,<br />

Oregon Health and Science University, Department <strong>of</strong><br />

Ophthalmology, Portland, OR, Koichi Kobayashi, Assistant<br />

Pr<strong>of</strong>essor, Harvard Medical School, Department <strong>of</strong><br />

Pathology, Boston, MA, Richard Flavell, Pr<strong>of</strong>essor, Yale<br />

University School Med, HHMI, New Haven, CT, James<br />

Rosenbaum, Pr<strong>of</strong>essor, Oregon Health and Science<br />

University, Department <strong>of</strong> Ophthalmology, Portland, OR<br />

Mutations in the human CARD15/NOD2 gene are associated<br />

with Crohn's disease or Blau syndrome, an autosomal<br />

dominant form <strong>of</strong> uveitis, arthritis, and dermatitis. NOD2 is<br />

responsible for sensing the bacterial product, muramyl<br />

dipeptide (MDP). Interestingly, mutations in a related gene,<br />

CIAS1, are associated with autoinflammatory syndromes in<br />

which interleukin (IL)-1 β plays a critical role in disease<br />

pathogenesis. Since the in vivo effects <strong>of</strong> MDP are<br />

incompletely studied and the susceptibility <strong>of</strong> the eye to<br />

mutations in NOD2 is unexplained, we developed a novel<br />

murine model <strong>of</strong> MDP-dependent uveitis. Intraocularly<br />

injected MDP induced a significant, transient inflammatory<br />

response. We characterized several salient features <strong>of</strong> this<br />

model such as the effect <strong>of</strong> dose and timing on the rolling,<br />

sticking and infiltrating leukocytes within the iris vasculature<br />

and tissue by intravital microscopy and histology. The<br />

increase in rolling leukocytes in MDP treated mice was<br />

dependent on the adhesion molecule L-selectin, as Lselectin<br />

knockout mice showed a significant decrease in the<br />

number <strong>of</strong> rolling cells within the iris vasculature in<br />

response to MDP. Importantly, NOD2 plays an essential role<br />

in ocular inflammation induced by MDP, as NOD2 knockout<br />

mice failed to develop uveitis in response to MDP.<br />

Unexpectedly, IL-1 receptor knockout mice did not show<br />

reduced MDP-induced uveitis. Thus, NOD2 may participate<br />

in the development <strong>of</strong> uveitis in response to bacterial<br />

products through its ability to enhance intravascular<br />

interactions between leukocytes and the endothelium in<br />

the eye in an IL-1 receptor independent fashion.<br />

doi:10.1016/j.clim.2007.03.119<br />

Su.81 Aut<strong>of</strong>luorescence Can be Used to<br />

Differentiate Between Normal, Activated, or<br />

Malignant Lymphocyte Populations<br />

Seth Pantanelli, Guest Researcher, National Institutes <strong>of</strong><br />

Health, NEI, Bethesda, MD, Zhuqing Li, Staff Scientist,<br />

National Institutes <strong>of</strong> Health, NEI, Bethesda, MD, Rober<br />

Fariss, Researcher, National Institutes <strong>of</strong> Health, NEI,<br />

Bethesda, MD, Matthew Cunningham, Guest Researcher,<br />

National Institutes <strong>of</strong> Health, NEI, Bethesda, MD, Baoying<br />

Liu, Researcher, National Institutes <strong>of</strong> Health, NEI,<br />

Bethesda, MD, Sankara Mahesh, Researcher, National<br />

Institutes <strong>of</strong> Health, NEI, Bethesda, MD, Robert<br />

Nussenblatt, Principal Investigator, National Institutes <strong>of</strong><br />

Health, NEI, Bethesda, MD<br />

The goal <strong>of</strong> this study was to investigate whether aut<strong>of</strong>luorescent<br />

properties <strong>of</strong> normal, activated or malignant<br />

lymphocyte populations could facilitate clinical diagnosis <strong>of</strong><br />

ocular diseases. Peripheral blood mononuclear cells<br />

(PBMCs) were isolated from normal human donor buffy<br />

coat blood products. T cells were purified from PBMCs and<br />

cultured with or without anti-CD3 and anti-CD28 stimulation.<br />

A B-lymphoma cell line (CA46) was cultured separately.<br />

Five experimental groups were prepared:<br />

unstimulated T cell, stimulated T cell, CA46 cell, stimulated<br />

T cell mixed with CA46 cell at a ratio <strong>of</strong> 1:4, or mixed<br />

at a ratio <strong>of</strong> 4:1. At 0, 1, and 3 days after culturing,<br />

0.6 ×106 cells from each <strong>of</strong> the five cell groups were cytospun<br />

onto a slide and imaged with a confocal microscope.<br />

Samples were excited with six excitation wavelengths: 351,<br />

364, 458, 476, 488, and 514 nm. For each excitation<br />

condition, the aut<strong>of</strong>luorescence intensity emitted from the<br />

sample was measured over a broad range <strong>of</strong> wavelengths.<br />

Pure T and B lymphoma populations were clearly distinguishable<br />

based on aut<strong>of</strong>luorescence intensity spectra. B<br />

lymphoma cells were the least fluorescent when excited<br />

with 351 nm light, but most fluorescent when excited with<br />

longer wavelengths like 488 nm. Mixed populations <strong>of</strong> T and<br />

B lymphoma cells had emission intensities that fell inbetween<br />

those <strong>of</strong> the pure populations. We conclude that<br />

normal, activated and malignant lymphocyte populations<br />

can be distinguished based on their aut<strong>of</strong>luorescent<br />

properties in vitro. Future work with in vivo models may<br />

prove useful in facilitating the diagnosis <strong>of</strong> uveitis and<br />

other ocular diseases.<br />

doi:10.1016/j.clim.2007.03.120<br />

Su.82 Developmental Expression <strong>of</strong> PD1 in the<br />

Retina<br />

Ling Chen, PhD Student, Jules Stein Eye Institute, Los<br />

Angeles, CA, Vicky Chang, Medical Student, Jules Stein Eye<br />

Institute, Los Angeles, CA, Ralph Levinson, Assistant<br />

Pr<strong>of</strong>essor, Jules Stein Eye Institute, Los Angeles, CA, Lynn<br />

Gordon, Associate Pr<strong>of</strong>essor, Jules Stein Eye Institute, Los<br />

Angeles, CA, Jonathan Braun, Pr<strong>of</strong>essor, Department <strong>of</strong><br />

Pathology and Laboratory Medicine, Los Angeles, CA<br />

Purpose: Programmed cell death 1 (PD-1) has a major<br />

function as a negative immune regulator. We recently<br />

identified PD-1 expression in neuronal cells <strong>of</strong> the retina.<br />

One potential role for PD-1 expression is in retinal<br />

maturation. The purpose <strong>of</strong> this study was to identify<br />

neuronal cell types that express retinal PD-1 and determine<br />

the time course <strong>of</strong> expression in the developing<br />

retina. Methods: PD-1 protein expression in the mouse<br />

retina was detected by immunohistochemistry at multiple<br />

embryonic and adult stages. Co-localization experiments<br />

were performed to detect PD-1 and Brn3a, Thy-1, AP2,<br />

GFAP, calbindin, or islet. RT-PCR and in situ hybridization<br />

were performed to quantify PD-1 mRNA expression.<br />

Results: Constitutive expression <strong>of</strong> PD-1 protein and<br />

mRNA was observed in the retinal ganglion cell. Confocal<br />

microscopy revealed that 68% <strong>of</strong> the PD-1 cells were <strong>of</strong> the<br />

retinal ganglion cell lineage and 27% were amacrine cells.<br />

PD-1 expression levels changed markedly during development.


Abstracts<br />

PD-1 expression was first observed at E14, 2 days after the<br />

first retinal ganglion cells appear in the retina. Expression<br />

levels <strong>of</strong> PD-1 significantly increased during development,<br />

reaching a peak at P13 with a subsequent decrease to an<br />

intermediate expression level at P24. Conclusions: PD-1 is<br />

expressed in retinal ganglion cells and amacrine cells and<br />

its expression dynamically changes during retinal development.<br />

Maximal PD-1 expression is noted at the most critical<br />

time period in postnatal visual plasticity. This observation<br />

raises the possibility <strong>of</strong> a developmental role for PD-1 in<br />

maturation <strong>of</strong> the ganglion cell layer and retinal remodeling<br />

process<br />

doi:10.1016/j.clim.2007.03.121<br />

Su.83 Human Lung Tumor Cells Modifies Rates <strong>of</strong><br />

CD4+CD25+ T Regulatory, CD19+CD23+ B Regulatory<br />

Cells, CD3+CD95+ Cells, NK Cells and B Lymphocytes<br />

Bayram Kiran, Tip Fakultesi, Temel Bilimler Binasi, Istanbul,<br />

Turkey, Akif Turna, Yedikule Hospital for Chest Diseases and<br />

Thoracic Surgery, Kadikoy, Istanbul, Turkey, Alper Yener,<br />

Istanbul Tip Fakultesi, Istanbul, Turkey, Atilla Gürses,<br />

Yedikule Gogus Hastaliklari Hastanesi, Istanbul, Turkey,<br />

Andac Salman, Istanbul Tip Fakultesi, Istanbul, Turkey,<br />

Selim Badur, Istanbul Tip Fakultesi, Temel Bilimler Binasi,<br />

Istanbul, Turkey<br />

The role <strong>of</strong> T regulatory cells and NK cells in human lung<br />

cancer has not yet been clarified. Our aim was to evaluate<br />

how the microenvironment <strong>of</strong> a tumor mass induced phenotypical<br />

changes in lymphocyte subsets. Our subjects were<br />

10 patients with resectable non-small cell lung cancer. We<br />

evaluated 47 different phenotypically different lymphocyte<br />

subsets in blood samples taken from the pulmonary artery,<br />

pulmonary vein <strong>of</strong> a tumor bearing pulmonary lobe and<br />

peripheral blood during pulmonary resectional surgery. We<br />

showed that, tumor cells boosted CD25high+CD4high+T<br />

lymphocytes (Treg cells), B lymphocytes, NK and CD19+<br />

CD23+ cells (Breg cells) and CD3+CD95+ (FasL+T lymphocytes)<br />

(p=0.015, p=0.001, p=0.02, p=0.04, p=0.03 respectively).<br />

However, Mac-1 cells and HLADR+T lymphocytes<br />

were found to be decreased by tumor mass (p=0.04 and<br />

p=0.04), whereas only Breg, NK cell and B lymphocyte<br />

subsets were found to be expansed. In addition, the patients<br />

showed expansions <strong>of</strong> CD45R0+ activated T lymphocytes and<br />

CD18+CD11b+(Mac-1) cell subsets without significant alteration<br />

by tumor microenvironment. In conclusion, subsets <strong>of</strong><br />

Treg, Breg cells, FasL+ T lymphocytes, B lymphocytes were<br />

modified by lung cancer microenvironment itself. This study<br />

also showed that, peripheral blood might not be good source<br />

for investigating the anti-tumor immunity since only Breg, NK<br />

cell and Treg cell subsets were found expansed peripherally.<br />

T lymphocytes and Mac-1 cells may be modified by systemic<br />

antitumor response rather than by local tumoral microenvironment.<br />

This study provides important insight into how<br />

tumor infiltrating lymphocytes and peripheral immune<br />

systems may be different in terms <strong>of</strong> lymphocyte subset<br />

expansion dynamics.<br />

doi:10.1016/j.clim.2007.03.122<br />

Su.84 Gene Expression Patterns <strong>of</strong> Intestinal<br />

Epithelial Cells in Murine Models <strong>of</strong> Colitis<br />

Ken Flanagan, Postdoctoral Researcher, Genentech,<br />

Department <strong>of</strong> Pathology, South San Francisco, CA, Jennine<br />

Cornelius, Research Associate, Genentech, Department <strong>of</strong><br />

Pathology, South San Francisco, CA, Zora Modrusan,<br />

Scientist, Genentech, Department <strong>of</strong> Molecular Biology,<br />

South San Francisco, CA, Lian Mo, Senior Research<br />

Associate, Genentech, Department <strong>of</strong> Pathology, South San<br />

Francisco, CA, Arvind Chavali, Intern, Genentech,<br />

Department <strong>of</strong> Pathology, South San Francisco, CA, Ian<br />

Kasman, Research Associate, Genentech, Department <strong>of</strong><br />

Pathology, South San Francisco, CA, Laszlo Komuves,<br />

Scientist, Genentech, Department <strong>of</strong> Pathology, South San<br />

Francisco, CA, Lauri Diehl, Scientist, Genentech,<br />

Department <strong>of</strong> Pathology, South San Francisco, CA<br />

In the healthy colon, intestinal epithelial cells (IEC) form<br />

a physical barrier separating the myriad <strong>of</strong> gut antigens from<br />

the cells <strong>of</strong> the immune system. Simultaneously, IEC employ<br />

several mechanisms to actively maintain immunologic<br />

tolerance to non-pathogenic antigens, including commensal<br />

bacteria. However, during inflammatory bowel disease (IBD)<br />

the line <strong>of</strong> defense provided by IEC is breached, resulting in<br />

uncontrolled immune responses. As IEC are a principal mediator<br />

<strong>of</strong> immune responses in the gut, we were interested in<br />

discerning the gene expression pattern <strong>of</strong> IEC during<br />

development and progression <strong>of</strong> IBD. Laser capture microdissection<br />

and microarray analysis were combined to identify<br />

genes with altered expression patterns in two models <strong>of</strong><br />

murine colitis. Genes involved in processes <strong>of</strong> the immune<br />

system, including lymphocyte migration, apoptosis and<br />

antigen presentation were affected in the IEC <strong>of</strong> mice with<br />

colitis indicating that IEC function to attract and activate<br />

cells <strong>of</strong> the immune system.<br />

doi:10.1016/j.clim.2007.03.123<br />

S169<br />

Su.85 TNF Binding by Certolizumab Pegol,<br />

Adalimumab, and Infliximab-Stoichiometry,<br />

Complex Formation, and the Biologic Effects <strong>of</strong><br />

Complexes<br />

Alistair Henry, Senior Principal Scientist, UCB Celltech,<br />

Research Biologicals, Slough, UK, Jeff Kennedy, Senior<br />

Scientist, UCB Celltech, Enzymology, Slough, UK, Gianluca<br />

Fossati, Principal Scientist, UCB Celltech, Slough, UK,<br />

Andrew Nesbitt, Senior Group Leader, UCB Celltech,<br />

Inflammation Discovery, Slough, UK<br />

Immune complexes can have various unwanted consequences.<br />

We determined the binding stoichiometry <strong>of</strong> certolizumab<br />

pegol, adalimumab, and infliximab to the tumor<br />

necrosis factor (TNF) trimer, and the size and in vitro biologic<br />

consequences <strong>of</strong> the resulting immune complexes. Isothermal<br />

titration calorimetry was used to assess the stoichiometry <strong>of</strong><br />

binding through calculation <strong>of</strong> the number <strong>of</strong> anti-TNF<br />

molecules bound to a TNF trimer when no more heat <strong>of</strong><br />

binding was released. Dynamic light scatter assessed the size<br />

<strong>of</strong> complexes formed over a range <strong>of</strong> antigen:antibody ratios.<br />

Effect <strong>of</strong> the complexes on peripheral blood neutrophil<br />

degranulation (myeloperoxidase release) and superoxide


S170 Abstracts<br />

production (oxidation <strong>of</strong> cytochrome c) were measured. At<br />

optimum antigen:antibody ratio, adalimumab and infliximab<br />

formed huge complexes N30 nm in diameter, suggesting that<br />

these bivalent antibodies crosslink TNF trimers. Certolizumab<br />

pegol complexes were b20 nm in diameter, consistent<br />

with its univalent structure preventing crosslinking. Certolizumab<br />

pegol bound 2.9 monomers at saturation, whereas<br />

infliximab and adalimumab both bound around 2.5 monomers.<br />

This suggests that steric or allosteric restrictions<br />

prevent the bivalent antibodies binding to all monomers in<br />

a trimer. The large immune complexes formed by adalimumab<br />

and infliximab caused degranulation and superoxide<br />

production by neutrophils. The smaller certolizumab pegol<br />

complexes had only marginal effects. The bivalent structures<br />

<strong>of</strong> infliximab and adalimumab form enormous complexes with<br />

TNF trimers, which have proinflammatory effects on neutrophils<br />

in vitro. Certolizumab pegol does not form large<br />

complexes with TNF trimers due to its univalent structure<br />

preventing crosslinking, giving it a unique mode <strong>of</strong> action in<br />

this regard.<br />

doi:10.1016/j.clim.2007.03.124<br />

Su.86 Certolizumab Pegol and Adalimumab<br />

Accumulation in the Inflammed Paws <strong>of</strong> Mice with<br />

Collagen-induced Arthritis Compared to<br />

Noninflammed Tissue In Vivo Bi<strong>of</strong>luorescence<br />

Imaging <strong>of</strong> Alexa 680-labeled Antibodies<br />

Roger Palframan, Senior Group Leader, UCB Celltech,<br />

Pharmacology, Slough, UK, Alex Vugler, Research Scientist,<br />

UCB Celltech, Pharmacology, Slough, UK, Adrian Moore,<br />

Senior Group Leader, UCB Celltech, Pharmacology, Slough,<br />

UK, Mark Baker, Research Scientist, UCB Celltech, <strong>Clinical</strong><br />

Pharmacology and Experimental Medicine, Slough, UK,<br />

Daniel Lightwood, Group Leader, UCB Celltech, Antibody<br />

Biology, Slough, UK, Andrew Nesbitt, Senior Group Leader,<br />

UCB Celltech, Inflammation Discovery, Slough, UK, Roly<br />

Foulkes, Director, UCB Celltech, Non-clinical Development/<br />

Inflammation Therapeutic Area, Slough, UK, Neil Gozzard,<br />

Director, UCB Celltech, Pharmacology, Slough, UK<br />

Variability in disposition <strong>of</strong> monoclonal antibodies results<br />

in different exposure time courses in inflammed and noninflammed<br />

tissues. We investigated the disposition <strong>of</strong> certolizumab<br />

pegol and adalimumab in normal and inflammed<br />

tissue in vivo in mice, using a novel noninvasive bi<strong>of</strong>luorescence<br />

technique. Certolizumab pegol or adalimumab labeled<br />

with the low-molecular weight dye Alexa 680 was administered<br />

intravenously (2 mg/kg) in naïve DBA/1 mice and in<br />

DBA/1 mice with ongoing collagen-induced arthritis. The<br />

accumulation <strong>of</strong> certolizumab pegol and adalimumab was<br />

measured in the hind paws at multiple points up to 26 hours<br />

using a Xenogen IVIS200 bi<strong>of</strong>luorescence imager. Tail blood<br />

samples were taken for determination <strong>of</strong> serum levels <strong>of</strong> the<br />

reagents by ELISA. Both reagents penetrated inflammed<br />

tissue more than noninflammed tissue. The penetration <strong>of</strong><br />

certolizumab pegol into inflammed arthritic paws was<br />

greater and more prolonged than for adalimumab (inflammed:noninflammed<br />

tissue ratios were 3.9:1 for certolizumab<br />

pegol and 1.9:1 for adalimumab; elimination half-lives<br />

were 27.8 and 5.5 hours, respectively). The plasma time<br />

courses reflected known differences in exposure, with<br />

certolizumab pegol maintaining higher plasma concentrations<br />

than adalimumab. In this model, certolizumab pegol<br />

disposition was more responsive to inflammation, with<br />

prolonged tissue infiltration occurring primarily in inflammed<br />

tissue. Certolizumab pegol, in contrast to adalimumab,<br />

appeared to readily enter inflammed tissue but not noninflammed<br />

tissue. This feature <strong>of</strong> certolizumab pegol may be<br />

conferred on the molecule by PEGylation. Increased drug<br />

exposure at the site <strong>of</strong> inflammation might be an important<br />

consideration for the treatment <strong>of</strong> inflammatory disorders<br />

such as rheumatoid arthritis and Crohn's disease.<br />

doi:10.1016/j.clim.2007.03.126<br />

Su.87 Comparison <strong>of</strong> Naturally Occurring Human<br />

Immunoglobulin Sequences with the Anti-TNF<br />

Agents Certolizumab Pegol, Adalimumab, and<br />

Infliximab<br />

Andrew Martin, Senior Lecturer, University College London,<br />

Biochemistry and Molecular Biology, London, UK, Kerry<br />

Tyson, Principal Scientist, UCB Celltech, Antibody Biology,<br />

Slough, UK, Matt Page, Research Associate Informatics, UCB<br />

Celltech, Therapeutic Project Informatics, Slough, UK,<br />

Gianluca Fossati, Principal Scientist, UCB Celltech,<br />

Inflammation Discovery, Slough, UK, James Snowden, Senior<br />

Scientist, UCB Celltech, Informatics, Slough, UK, Raghavan<br />

Abhinandan, Lecturer, University College London,<br />

Biochemistry and Molecular Biology, London, UK, Alastair<br />

Lawson, Director <strong>of</strong> Antibody Biology, UCB Celltech,<br />

Antibody Biology, Slough, UK, Andrew Nesbitt, Senior Group<br />

Leader, UCB Celltech, Inflammation Discovery, Slough, UK,<br />

Andrew Popplewell, Associate Director, UCB Celltech,<br />

Antibody Biology, Slough, UK<br />

V-region sequences <strong>of</strong> certolizumab pegol, adalimumab,<br />

and infliximab were compared with a database <strong>of</strong> normal<br />

human IgG V-region sequences. A database <strong>of</strong> rearranged<br />

human variable heavy [VH] and variable kappa light [VK]<br />

amino acid sequences was obtained by sequencing 243<br />

unique VH chains and 312 unique VK chains from healthy<br />

donors. Each sequence was compared to the closest human<br />

“germline” sequence and also scored against every other<br />

sequence <strong>of</strong> the same chain type, using percentage identity<br />

to measure sequence similarity. Z-scores were calculated<br />

from a frequency distribution plot and compared between<br />

agents. A positive Z-score indicates that a sequence is more<br />

typical than average <strong>of</strong> a human sequence. The mean<br />

number <strong>of</strong> nongermline framework residues in rearranged<br />

human antibodies was 4.3% for VK and 8.8% for VH. Of the<br />

three agents, certolizumab pegol had a V-region most typical<br />

<strong>of</strong> the rearranged human sequences with 3.8% nongermline<br />

residues for VK and 8.5% for VH. Adalimumab had 1.3% and<br />

1.2% and infliximab 31.3% and 22.2%, respectively. Positive Zscores<br />

were obtained for the VK and VH sequences <strong>of</strong><br />

certolizumab pegol (0.15 and 1.46) and adalimumab (0.67<br />

and 1.34). However, infliximab V-region sequences gave<br />

negative Z-scores (−2.37 and −0.58) indicating they are less<br />

typical <strong>of</strong> human sequences. The certolizumab pegol Vregion<br />

framework sequence has a similar mean number <strong>of</strong><br />

nongermline residues as rearranged human IgG V-region


Abstracts<br />

frameworks. The complete VH and VK sequences were also<br />

more human-like than average. The certolizumab pegol Fab'<br />

sequence can be considered typical <strong>of</strong> naturally occurring<br />

human IgG.<br />

doi:10.1016/j.clim.2007.03.127<br />

Su.89 Role <strong>of</strong> Enteric Lumenal TLR Ligand Sampling<br />

in the Formation <strong>of</strong> Novel Resident Mucosal<br />

Dendritic Cells<br />

Daisuke Fujiwara, Postdoctoral Fellow, Department <strong>of</strong><br />

Pathology and Laboratory Medicine, David Geffen School <strong>of</strong><br />

Medicine at University <strong>of</strong> California, Los Angeles, Los<br />

Angeles, CA, Bo Wei, Assistant Pr<strong>of</strong>essor, Department <strong>of</strong><br />

Pathology and Laboratory Medicine, David Geffen School <strong>of</strong><br />

Medicine at University <strong>of</strong> California, Los Angeles, Los<br />

Angeles, CA, Michael McPherson, PhD Student, Department<br />

<strong>of</strong> Pathology and Laboratory Medicine, David Geffen School<br />

<strong>of</strong> Medicine at University <strong>of</strong> California, Los Angeles, Los<br />

Angeles, CA, Jonathan Braun, Pr<strong>of</strong>essor and Chair,<br />

Department <strong>of</strong> Pathology and Laboratory Medicine, David<br />

Geffen School <strong>of</strong> Medicine at University <strong>of</strong> California, Los<br />

Angeles, Los Angeles, CA<br />

Resident intestinal dendritic cells (DCs) play important<br />

immunoregulatory roles through sensing and antigen handling<br />

<strong>of</strong> enteric microbiota. However, intestinal DCs have not been<br />

well characterized. In this study, we found that CD11c+ cells<br />

isolated from the small intestine superficial and deep lamina<br />

propria were unusual (compared to lymph node and spleen<br />

DC's) due to their graduated expression <strong>of</strong> CD11b or B220, and<br />

the predominance <strong>of</strong> B220+CD11b+ double-positive (DP) DC's.<br />

Since lamina propria DC's engage in lumenal sampling and thus<br />

encounter TLR ligands, we wondered whether TLR signaling<br />

might induce the unusual DP phenotype. Using bone marrowderived<br />

DC's differentiated with Flt-3L, we evaluated the<br />

effect <strong>of</strong> co-culture with various TLR ligands. Whereas Flt-3L<br />

alone induced single-positive CD11b+ or B220+ DCs, DP DCs<br />

predominated after TLR2, TLR4, and TLR9 ligands. The<br />

response was abrogated in myd88−/− mice, suggesting that<br />

DP DC's are induced through TLR-MyD88 pathway. Fecal<br />

extracts also induced DP DC formation; because this response<br />

was absent in TLR4−/− mice, it appeared that the predominant<br />

enteric bioactivity for DP DC induction was LPS. Among<br />

lamina propria DCs, we also observed a probable immature<br />

mDC subset (CD11c-B220-CD11blow). This subset was deficient<br />

in germ-free (GF) mice or cd8 knockout mice, suggesting an<br />

important TLRL role in differentiation stage <strong>of</strong> DCs in vivo,and<br />

a novel requirement for CD8+ T cells. These findings indicate<br />

that lumenal TLR4 ligands shape the differentiation <strong>of</strong> mucosal<br />

resident DCs, resulting in a novel phenotype associated with<br />

distinct immunoregulatory traits. Supported by NIH DK46763<br />

and DK69434.<br />

doi:10.1016/j.clim.2007.03.128<br />

Su.90 Up-Regulation <strong>of</strong> the PI3-Kinase Pathway in<br />

Lamina Propria T Lymphocytes<br />

Jutta Braunstein, Research Scientist, University Hospital<br />

Heidelberg, Heidelberg, Germany, Frank Autschbach,<br />

Deputy Head <strong>of</strong> Pathology, University Hospital Heidelberg,<br />

Heidelberg, Germany, Felix Lasitschka, Postdoctoral Fellow,<br />

University Hospital Heidelberg, Heidelberg, Germany,<br />

Thomas Giese, Group Leader, University Hospital<br />

Heidelberg, Heidelberg, Germany, Antje Heidtmann,<br />

Technician, University Hospital Heidelberg, Heidelberg,<br />

Germany, Bernd Sido, Senior Physician, University Hospital<br />

Heidelberg, Heidelberg, Germany, Benjamin Funke,<br />

Physician, University Hospital Heidelberg, Heidelberg,<br />

Germany, Andreas Schroeder, Medical Advisor, Merck Sero,<br />

Darmstadt, Germany, Yvonne Samstag, Group Leader,<br />

University Hospital Heidelberg, Heidelberg, Germany,<br />

Stefan Meuer, Head <strong>of</strong> Institute <strong>of</strong> <strong>Immunology</strong>, University<br />

Hospital Heidelberg, Heidelberg, Germany<br />

Understanding the regulation <strong>of</strong> immune responses in the<br />

normal intestinal mucosa is critical for defining its alterations<br />

in inflammatory bowel disease. Importantly, intestinal lamina<br />

propria T lymphocytes (LPT) when investigated ex vivo exhibit<br />

functional properties pr<strong>of</strong>oundly different from those <strong>of</strong><br />

peripheral blood T lymphocytes (PBT). In particular, following<br />

CD2 stimulation they are able to produce markedly higher<br />

levels <strong>of</strong> cytokines than PBT. This study provides insight into<br />

signaling events associated with the high CD2 responsiveness <strong>of</strong><br />

LPT. CD2 stimulation results in enhanced phosphorylation <strong>of</strong><br />

the PI3-kinase dependent kinase Akt and its down-stream<br />

target GSK-3 in LPTwhen compared to PBT. That up-regulation<br />

<strong>of</strong> PI3-kinase pathway activation in LPTcontributes to the high<br />

IL-2 expression in response to CD2 stimulation is demonstrated<br />

by the fact that inhibition <strong>of</strong> this pathway by the PI3-kinase<br />

specific inhibitor Ly294002 to levels induced in PBT clearly<br />

reduces IL-2 gene expression (and TNF-α, CD40L gene<br />

expression) in LPT. Immunohistochemical analysis reveals<br />

that Akt phosphorylation occurs in few lamina propria mononuclear<br />

cells in the normal mucosa in vivo; moreintense<br />

phospho-Akt staining <strong>of</strong> larger numbers <strong>of</strong> lamina propria<br />

mononuclear cells is detectable in the inflammed intestine<br />

(ulcerative colitis) indicating that activation <strong>of</strong> the PI3-kinase<br />

pathway plays an important role in intestinal inflammation.<br />

Enhanced responsiveness <strong>of</strong> these pathways to costimulatory<br />

stimuli may allow for rapid and vigorous T cell responses and<br />

could therefore be fundamental to intestinal mucosal immune<br />

responses and homeostasis.<br />

doi:10.1016/j.clim.2007.03.129<br />

S171<br />

Su.91 Differential Levels <strong>of</strong> Intercellular Glutathion<br />

(GSH) Dictate the Shift From Adaptive to Innate T<br />

Cell Function in the Human Intestinal Mucosa<br />

Bernd Sido, Senior Physician, Department <strong>of</strong> Surgery,<br />

Heidelberg, Germany, Frank Autschbach, Senior Physician,<br />

Institute <strong>of</strong> Pathology, Heidelberg, Germany, Jutta<br />

Schroeder-Braunstein, Scientist, Institute <strong>of</strong> <strong>Immunology</strong>,<br />

Heidelberg, Germany, Thomas Giese, Group Leader,<br />

Institute <strong>of</strong> <strong>Immunology</strong>, Heidelberg, Germany, Felix<br />

Lasitschka, Postdoctoral Fellow, Institute <strong>of</strong> <strong>Immunology</strong>,<br />

Heidelberg, Germany, Stefan Meuer, Director, Institute <strong>of</strong><br />

<strong>Immunology</strong>, Heidelberg, Germany<br />

Isolated human Tcells from the healthy human mucosa are<br />

non-responsive to T cell receptor engagement yet mount


S172 Abstracts<br />

vigorous responses to non-specific activation through CD2 or<br />

CD28 triggering with regard to both proliferation and cytokine<br />

secretion. Since resting T lymphocytes do not express<br />

the cystine transporter (xCT), which is critical for the import<br />

<strong>of</strong> cystine across the plasma membrane and since cystine is a<br />

critical precursor for GSH synthesis, they are dependent on an<br />

extracellular source <strong>of</strong> cysteine. Under physiologic conditions,<br />

cysteine is not available to T cells in the intestinal<br />

mucosa since the local myeloid cell population is unable to<br />

secrete cysteine, unlike circulating monocytes from peripheral<br />

blood. As a consequence, mucosal T lymphocytes contain<br />

significantly lower GSH concentrations than blood T lymphocytes.<br />

Enhancement <strong>of</strong> GSH levels in mucosal T cells by<br />

providing cysteine either directly or by adding blood<br />

monocytes restores their antigen receptor reactivity. This<br />

mechanism appears to be relevant for their behaviour in<br />

inflammatory bowel diseases where GSH levels in mucosal T<br />

cells from inflammatory sites are at least as high as in<br />

circulating blood T cells. Here, myeloid cells with the capacity<br />

to secrete cysteine are frequently detected. Importantly,<br />

this is not only due to their influx from blood but, at least in<br />

part, to a functional and phenotypic change under altered<br />

microenvironmental conditions. Supported by a grant from<br />

DFG/SFB405/B6.<br />

doi:10.1016/j.clim.2007.03.130<br />

Su.92 Whole Genome Association Identifies Novel<br />

Susceptibility Genes for Crohn's Disease and<br />

Implicates a Crucial Role for Autophagy<br />

John Rioux, Associate Pr<strong>of</strong>essor, Universite de Montreal,<br />

Department <strong>of</strong> Medicine, Montreal, QC, Canada, Ramnik<br />

Xavier, Associate Pr<strong>of</strong>essor, Massachusetts General Hospital,<br />

Harvard Medical School, Boston, MA, Kent Taylor, Associate<br />

Pr<strong>of</strong>essor, Cedars Sinai Medical Center, Los Angeles, CA,<br />

Philippe Goyette, Scientist, Montreal Heart Institute,<br />

Montreal, QC, Canada, Mark Silverberg, Associate Pr<strong>of</strong>essor,<br />

Mount Sinai Hospital, Toronto, ON, Canada, Alan Huett,<br />

Postdoctoral Fellow, Massachusetts General Hospital,<br />

Harvard Medical School, Boston, MA, Todd Green, Project<br />

Manager, Broad Institute <strong>of</strong> MIT and Harvard, Cambridge,<br />

MA, Petric Kuballa, Postdoctoral Fellow, Massachusetts<br />

General Hospital, Harvard Medical School, Boston, MA,<br />

Michael Barmada, Associate Pr<strong>of</strong>essor, University <strong>of</strong><br />

Pittsburgh, Pittsburgh, PA, Lisa Datta, John Hopkins<br />

University School <strong>of</strong> Medicine, Baltimore, MD, Yin Yao<br />

Shugart, Associate Pr<strong>of</strong>essor, John Hopkins University,<br />

Bloomberg School <strong>of</strong> Public Health, Baltimore, MD, Edmond<br />

Jean Bernard, Assistant Pr<strong>of</strong>essor, Universite de Montreal,<br />

Montreal, QC, Canada, Ling Mei, Cedars Sinai Medical<br />

Center, Los Angeles, CA, Dan Nicolae, Associate Pr<strong>of</strong>essor,<br />

University <strong>of</strong> Chicago, Departments <strong>of</strong> Medicine and<br />

Statistics, Chicago, IL, Hillary Steinhart, Associate<br />

Pr<strong>of</strong>essor, Mount Sinai Hospital and University <strong>of</strong> Toronto,<br />

Toronto, ON, Canada, Jerome Rotter, Pr<strong>of</strong>essor, Cedars Sinai<br />

Medical Center, Los Angeles, CA, Judy Cho, Yale University,<br />

New Haven, CT, Mark Daly, The Broad Institute <strong>of</strong> MIT and<br />

Harvard, Cambridge, MA, Miguel Regueiro, Associate<br />

Pr<strong>of</strong>essor <strong>of</strong> Medicine, University <strong>of</strong> Pittsburgh, Division<br />

<strong>of</strong> Gastroenterology, Hepatology and Nutrition, Pittsburgh,<br />

PA, Philip Schumm, University <strong>of</strong> Chicago, Department<br />

<strong>of</strong> Health Studies, Chicago, IL, Richard Duerr, Associate<br />

Pr<strong>of</strong>essor, University <strong>of</strong> Pittsburgh, School <strong>of</strong> Medicine,<br />

Pittsburgh, PA, Steven Brant, Meyerh<strong>of</strong>f Inflammatory<br />

Bowel Disease Center, John Hopkins University School<br />

<strong>of</strong> Medicine, Baltimore, MD<br />

In 2002, the NIDDK IBD Genetics Consortium was founded<br />

by six Genetic Research Centers located in Canada and the<br />

US. Our first stage genome-wide association (GWA) study <strong>of</strong><br />

308,332 autosomal single nucleotide polymorphisms (SNPs)<br />

in 567 ileal CD samples and 571 controls identified genetic<br />

variants within the IL23R gene that influence an individual's<br />

risk for developing IBD (Duerr et al. Science 2006). In the<br />

current study, we typed an independent set 401 patients with<br />

ileal CD and 433 controls for the exact same set <strong>of</strong> SNPs. We<br />

have since performed a combined analysis <strong>of</strong> all cases and<br />

controls as well as an independent replication study that<br />

establish five regions <strong>of</strong> unambiguously confirmed association<br />

<strong>of</strong> genome-wide significance. Specifically, in addition to<br />

the established CARD15 and IL23R associations, we report<br />

three novel confirmed associations with variation in the<br />

PHOX2B and ATG16L1 genes and in an intergenic region on<br />

10q21.1. Moreover, we also identify a statistically significant<br />

excess <strong>of</strong> additional regions showing nominal replication <strong>of</strong><br />

association that likely contain additional gene(s). We further<br />

demonstrate that the ATG16L1 gene is expressed in intestinal<br />

epithelial cells and that functional knock down <strong>of</strong> this gene<br />

abrogates autophagy <strong>of</strong> Salmonella typhimurium. Together<br />

these findings implicate that autophagy and other host<br />

responses to intra-cellular microbes are involved in the pathogenesis<br />

<strong>of</strong> CD.<br />

doi:10.1016/j.clim.2007.03.131<br />

Su.93 A High Density Association Study <strong>of</strong> the<br />

Extended Major Histocompatibility Locus in<br />

Ulcerative Colitis<br />

Philippe Goyette, Research Associate, Montreal Heart<br />

Institute, Montreal, QC, Canada, Todd Green, The Broad<br />

Institute <strong>of</strong> MIT & Harvard, Cambridge, MA, Paul de Bakker,<br />

The Broad Institute <strong>of</strong> MIT & Harvard, Cambridge, MA,<br />

Daniel Mirel, The Broad Institute <strong>of</strong> MIT & Harvard,<br />

Cambridge, MA, Christine Stevens, The Board Institute<br />

<strong>of</strong> MIT & Harvard, Cambridge, MA, Anna Latiano, IRCCS<br />

Casa Sollievo della S<strong>of</strong>ferenza, Italy, Jorge Oksenberg,<br />

University <strong>of</strong> California, San Francisco, San Francisco, CA,<br />

S. Hauser, University <strong>of</strong> California, San Francisco, San<br />

Francisco, CA, Vitto Annese, Medical School and University<br />

<strong>of</strong> Foggia, Italy, John Rioux, Montreal, QC, Canada,<br />

Mark Daly, The Broad Institute <strong>of</strong> MIT & Harvard,<br />

Cambridge, MA<br />

Ulcerative colitis (UC) and Crohn's disease (CD) are chronic<br />

inflammatory diseases <strong>of</strong> the gastrointestinal tract. While<br />

the study <strong>of</strong> genetic risk factors in CD has led to the identification<br />

<strong>of</strong> multiple risk factors, few have been convincingly<br />

identified in UC. Several independent genome-wide<br />

linkage scans, however, have suggested that the major<br />

histocompatibility complex (MHC) region plays a role in the<br />

genetic susceptibility to UC. While several putative association<br />

results have been reported for UC in this region, findings


Abstracts<br />

have not been consistent due to the limited number <strong>of</strong> alleles<br />

tested, to small sample sizes and to extensive LD in the<br />

region. We have recently reported the creation <strong>of</strong> a high<br />

density genetic map, including 7500 SNPs and DIPs, as well as<br />

the classical HLA genes. In the current study, we have<br />

selected 1500 <strong>of</strong> the most informative SNPs in order to<br />

capture the common variability in this region. Using this<br />

panel, we performed an association study in a cohort <strong>of</strong> 643<br />

UC cases and 1056 controls. Single marker association testing<br />

identified 35 SNPs with significant association (b4.5×10 − 5 )<br />

to UC. The peak <strong>of</strong> association, supported by multiple SNPs,<br />

is localized in the HLA-DRA and HLA-DQA gene regions. HLA<br />

tagging SNPs also identify this region as containing a UC<br />

susceptibility locus. Our initial analyses suggest that a<br />

common variant (∼30% frequency) in this region can explain<br />

the observed association. Higher density association mapping<br />

<strong>of</strong> this region and replication in an independent cohort<br />

will be necessary to identify causal allele(s).<br />

doi:10.1016/j.clim.2007.03.132<br />

Su.94 TRAM-34, a Blocker <strong>of</strong> the Calcium-activated<br />

Potassium Channel KCa3.1, Prevents Acute<br />

Transplant Rejection<br />

Heike Wulff, Assistant Pr<strong>of</strong>essor, University <strong>of</strong> California,<br />

Davis, Davis, CA, Yi-Je Chen, Graduate Student, University<br />

<strong>of</strong> California, Davis, Surgical and Radiological Sciences,<br />

Davis, CA, Girija Raman, Postdoctoral Researcher,<br />

University <strong>of</strong> California, Davis, Medical Pharmacology,<br />

Davis, CA, Clare Gregory, Pr<strong>of</strong>essor, University <strong>of</strong> California,<br />

Davis, Surgical and Radiological Sciences, Davis, CA<br />

Rejection <strong>of</strong> transplanted organs is caused by an immune<br />

response <strong>of</strong> recipient T cells against the donor MHC. One<br />

emerging new target for the suppression <strong>of</strong> T cell activation<br />

is the calcium-activated potassium channel KCa3.1. In<br />

addition to T and B cells, KCa3.1 is also expressed in<br />

macrophages, proliferating vascular smooth muscle and<br />

endothelial cells and has been shown to play an important<br />

role in the proliferation and migration <strong>of</strong> these cells. In<br />

2000, we designed a selective small molecule KCa3.1<br />

blocker called TRAM-34 (EC50=20 nM) and showed that it<br />

suppressed T cell proliferation and synergized with cyclosporine<br />

(PNAS 2000, 97: 8151). TRAM-34 has a half-life <strong>of</strong><br />

1.8 hours in rats and <strong>of</strong> 2.1 hours in rhesus macaques,<br />

prevents restenosis in rats and exhibits no signs <strong>of</strong> acute <strong>of</strong><br />

long-term toxicity. Encouraged by these results we are here<br />

testing TRAM-34 alone or in combination with cyclosporine<br />

in a model <strong>of</strong> acute transplant rejection. Hearts are<br />

heterotopically transplanted from male Brown Norway to<br />

male Lewis rats. While transplanted hearts in vehicle<br />

treated animals failed within 9 days (n=6), transplanted<br />

hearts in TRAM-34 treated rats (10 mg/kg twice daily)<br />

remained functional for an average <strong>of</strong> 25 days (n=6). In<br />

comparison, treatment with low dose cyclosporine (2 mg/kg<br />

Neoral®) resulted in a graft survival time <strong>of</strong> 18 days (n=6).<br />

Combinations <strong>of</strong> TRAM-34 with low dose cyclosporine are<br />

currently being tested. Supported by NIH and UC Davis.<br />

doi:10.1016/j.clim.2007.03.133<br />

Su.95 Low Doses IVIG with Plasmapheresis and<br />

Rituximab in Positive Cross Match Patients<br />

Miguel Rodriguez, Immunoallergist, Hospital<br />

Especialidades, IMSS, Guadalajara, Mexico, Luis Vales-<br />

Alberto, MD, Hospital Especialidades IMSS, Departmento de<br />

Nefrologia, Guadalajara, Mexico, Felicia Castro-Ramos,<br />

Biologist, Hospital Especialidades IMSS, Nephrology Unit,<br />

Guadalajara, Mexico, Francisco Monteon-Ramos,<br />

Nephrologist, Hospital de Especialidades IMSS, Nephrology<br />

Unit, Guadalajara, Efrain Montano-Gonzales,<br />

Immunoallergologist, Hospital Especialidades UMAE, IMSS<br />

Department <strong>of</strong> <strong>Immunology</strong> and Allergy, Guadalajara,<br />

Mexico, Alejandro Bautista-Vazquez, Nephrologist, Hospital<br />

de Especialidades, CMENGLANDUMAE, IMSS, Unit <strong>of</strong><br />

Nephrology, Guadalajara, Mexico, Salvador Chavez, MD,<br />

Hospital Especialidades IMSS, Departamento de Nefrologia,<br />

Guadalajara, Mexico<br />

Introduction: Renal Transplant (RT) is the best option for<br />

End Stage Renal Disease (ESRD) Positive Cross Match (+XM) is<br />

formal contraindication to RT. Protocols have been developed<br />

to overcome this problem. Methods and Materials: Simple<br />

not ramdomized sample. Patients ABO compatible with<br />

+XM (up to 30%) by FlowCytometry (FC) to living related<br />

donor. Patients scheme was: PP after that IVIG 100 mg/kg 3<br />

times every other day. If XM becomes negative we administrated<br />

Rituximab and go to RT. After transplant 3 more<br />

sessions <strong>of</strong> PP and IVIG were given. Results: We treated 6<br />

patients, 3 in 2nd RT. One persisted positive after scheme<br />

and was not transplanted. Average PRA Class I 91.83% (SD<br />

6.1) Class II 24.14 (SD 31.7. Five transplanted patients with<br />

satisfactory evolution after 7.6 Mo (SD 3.9 Mo) with excellent<br />

renal function: creatinine 0.9 (SD 0.07 mg/dl) and creatinine<br />

depuration 84 (SD 10 ml/min/1.73 m2. One case presented<br />

cellular rejection (percutaneous renal biopsy with negative<br />

c4d) 10 weeks after RT that respond to IV steroids. We do not<br />

detect any infectious complication. Discussion: It is possible<br />

to desensitize positive cross match patients with this<br />

schedule <strong>of</strong> very low IVIG doses, diminishing costs and<br />

without side effects. Until now results are very satisfactory,<br />

we need more cases to obtain experience.<br />

doi:10.1016/j.clim.2007.03.135<br />

S173<br />

Su.96 Semi-mature Bone Marrow Derived DC Expand<br />

Foxp3+CD4+CD25+CD127- Regulatory T Cells with<br />

Suppressive Activity in the Macaque<br />

Aureli Moreau, PhD student, INSERM U643- ITERT/U649,<br />

Nantes, France, Gaelle Beriou, PhD, Center <strong>of</strong> Neurologic<br />

Diseases, Boston, MA, Jack-Yves Deschamps, DVM, ENVN,<br />

Nantes, France, Joanna Ashton-Chess, PhD, INSERM<br />

U643- ITERT, Nantes, France, Heslan Michele, Engineer,<br />

INSERM U643- ITERT, Nantes, France, Elise Chiffoleau, CR2<br />

INSERM, INSERM U643- ITERT, Nantes, France, Regis Josien,<br />

CR1, INSERM U643- ITERT, Nantes, France, Philippe Moullier,<br />

DR2 INSERM, U649, Nantes, France, Brigitte Alliot-Licht,<br />

PhD, INSERM U643- ITERT, Nantes, France, Maria-Cristina<br />

Cuturi, DR2, INSERM U643- ITERT, Nantes, France<br />

Over the past 10 years, several types <strong>of</strong> tolerogenic dendritic<br />

cells (DC) and T regulatory cells (Treg) have been


S174 Abstracts<br />

identified that play a pivotal role in the control <strong>of</strong> autoimmunity<br />

and transplantation tolerance in rodents. Recent<br />

studies have shown that immature DC and ex vivo expanded<br />

Treg could be potential reagents to promote antigen-specific<br />

tolerance in vivo. To date, these works have been carried out<br />

mostly in rodents and will need to be validated in primates<br />

before being applied in the clinic. In order to better<br />

characterise tolerogenic DC and Treg in primates we first<br />

generated and characterised different types <strong>of</strong> DC from<br />

macaque bone marrow (BMDC). Among these cells, the most<br />

immature BMDC (adherent cells resulting from culture in the<br />

presence <strong>of</strong> GM-CSF alone) could be good candidates for in<br />

vivo testing in protocols <strong>of</strong> tolerance induction. We were also<br />

able to demonstrate that in the presence <strong>of</strong> IL-2, semimature<br />

BMDC (cultured with GM-CSF and IL-4) have the<br />

ability to expand CD4+CD25+CD127-Foxp3+ macaque Treg<br />

cells (25 fold in 7 days) and additionally increase their suppressive<br />

activity. These results represent an important step<br />

towards the potential use <strong>of</strong> tolerogenic DC and/or Treg as an<br />

adoptive cell therapy to induce antigen-specific tolerance in<br />

pre-clinical, non-human primate models.<br />

doi:10.1016/j.clim.2007.03.136<br />

Su.98 Low Doses <strong>of</strong> Intravenous Immunoglobulin in<br />

Renal Transplant Patients with High Levels <strong>of</strong><br />

Anti-HLA Panel Reactive Antibodies (PRA)<br />

Miguel Rodriguez, Immunologist, Hospital Especialidades,<br />

IMSS Department Inmunologia, Guadalajara, Mexico,<br />

Alejandro Bautista, Nephrologist, Hospital Especialidades<br />

IMSS, Department Nephrology, Guadalajara, Mexico,<br />

Francisco Monteon, MD, Hospital Especialidades Division <strong>of</strong><br />

Transplantes, Guadalajara, Mexico, Felicia Castro,<br />

Pharmacobiology Chemist, Hospital Especialidades, Division<br />

<strong>of</strong> Transplantes, Guadalajara, Mexico, Efrain Montaño, MD,<br />

Hospital Especialidades IMSS, Department Inmunologia<br />

Clínica, Guadalajara, Mexico, Antonio Flores, MD, Hospital<br />

Especialidades, IMSS Department Nefrologia, Guadalajara,<br />

Mexico<br />

Introduction: The renal transplant (RT) is the best<br />

treatment for the patients with End Stage Renal Disease<br />

(ESRD). Patient with anti-HLA antibodies has a short graft life.<br />

Different studies shown that intravenous immunoglobulin<br />

(IVIG) diminishes anti-HLA antibodies. Methods and materials:<br />

An open, not randomized clinical trial. We used 100 mg/kg<br />

<strong>of</strong> IVIG in patient with PRA higher than 305 with negative<br />

crossmatch to donor (XM−) by flow cytometry (FC). We<br />

administered 3 doses <strong>of</strong> IVIG 100 mg/kg every other day. One<br />

week after the PRA was repeated. Patients with PRA lower<br />

than 30%, were considered desensitized and proceeded to RT,<br />

monitoring the occurrence <strong>of</strong> rejections and graft survival.<br />

Results: We reported 8 patients (3M/5F), age <strong>of</strong> 22.8 SD<br />

2.4 years. Four patients with second RT (50%). PRA previous<br />

88.65% (SD 5.65%) Class I and 50.09% (SD 40.32%) Class II. After<br />

treatment 7.66% (SD 12.32%) Class I and 11.39% (SD25.37%)<br />

Class II. PRA diminished in 7 patients and in 1 patient<br />

remained elevated. Five received a RT from living related<br />

donor, ABO compatible (71.4%) and two <strong>of</strong> them from cadaver<br />

(28.6). The monitoring <strong>of</strong> this group is 46+–25.7 weeks. One<br />

patient (from cadaver) presented two rejection crises both<br />

resolved favorably. Creatinine being 0.95 mg/dl (SD 0.4).<br />

Creatinine depuration (MDRD)88.6 SD 22.05 ml/min/1.73m2.<br />

We did not document any infectious process within the group.<br />

Discussion: IVIG in low doses confer a protective effect, in<br />

patients with high PRA. We need more monitoring to evaluate<br />

the impact in the long term.<br />

doi:10.1016/j.clim.2007.03.137<br />

Su.99 Efficient Ex Vivo Priming <strong>of</strong> Naïve Precursors<br />

into EBV-specific Type-1 CD8+ T Cell Responses from<br />

EBV Seronegative Pediatric HTx Patients Requires<br />

IL-12 and IL-27<br />

Diana Metes, Assistant Pr<strong>of</strong>essor, University <strong>of</strong> Pittsburgh,<br />

Transplantation Surgery, Pittsburgh, PA, Camila Macedo,<br />

Postdoctoral Associate, University <strong>of</strong> Pittsburgh,<br />

Transplantation Surgery, Pittsburgh, PA, Walter Storkus,<br />

Pr<strong>of</strong>essor, University <strong>of</strong> Pittsburgh, Dermatology,<br />

Pittsburgh, PA, Iulia Popescu, Research Instructor,<br />

University <strong>of</strong> Pittsburgh, Transplantation Surgery,<br />

Pittsburgh, PA, Steve Webber, Pr<strong>of</strong>essor, University <strong>of</strong><br />

Pittsburgh, Pediatrics, Pittsburgh, PA<br />

The risk <strong>of</strong> post-transplant lymphoproliferative disorders<br />

(PTLD) is increased in pediatric recipients, and cellular<br />

immunotherapy with EBV specific cytotoxic T lymphocytes<br />

(CTLs) represents a promising therapeutic strategy to restore<br />

cellular immunity in PTLD patients. Although we were<br />

effective at priming strong EBV-specific CTL responses from<br />

EBV-seronegative healthy adults using DC-based protocols,<br />

similar attempts were unsuccessful for EBV-seronegative<br />

pediatric Tx patients. Peripheral blood lymphocytes were<br />

obtained from 5 EBV seronegative HTx patients and were<br />

stimulated ex vivo for 10 days with autologous mature DC<br />

pulsed with a cocktail <strong>of</strong> MHC class I-restricted EBV peptides<br />

as follows: (i)DC/peptide only (ii)DC/peptide +IL-12 (an<br />

adjuvant cytokine for promoting Type-1 immunity-IFNgama<br />

secretion) (iii)DC/peptide +IL-27 (a potent early cytokine<br />

regulator <strong>of</strong> Type-1 commitment) (IV)DC/peptide+IL-12+IL-27.<br />

EBV-specific CD8+ T cell responses were screened by flow<br />

cytometry and IFNgama ELISPOT assays. The DC/peptide +<br />

IL-12 +IL-27 approach yielded the best yields (1.7X), the<br />

optimal EBV-specific CD8+ T cell expansion (from undetectable<br />

to 1.5% +0.5 EBV-tetramer+ CD8 T cells), and optimal<br />

EBV specificity (from 3+2 spots/105 to 45+15 spots/105<br />

CD8+ T cells in ELISPOT assays). DC/peptide (9+2 spots/105)<br />

or the DC/peptide+IL-12 (12 +4 spots/105) protocols were<br />

unsuccessful in this setting. These results suggest that<br />

priming <strong>of</strong> functional EBV-specific CD8+ Tcells from pediatric<br />

EBV seronegative HTx patients is feasible, but EBV-Ag presentation<br />

by DC may require IL-27 in addition to IL-12 to<br />

promote Tc1 cells from naive CD8+ T cell precursors ex vivo.<br />

doi:10.1016/j.clim.2007.03.138<br />

Su.100 Pharmacodynamic Controlled Tapering <strong>of</strong><br />

Cyclosporine A: A New Step Towards Individualized<br />

Immunosuppressive Therapy in Transplanted Patients<br />

Thomas Giese, Group Leader, Institute <strong>of</strong> <strong>Immunology</strong>,<br />

Heidelberg, Germany, Claudia Sommerer, Physician,


Abstracts<br />

Department <strong>of</strong> Nephrology, Heidelberg, Germany, Martin<br />

Zeier, Medical Director, Department <strong>of</strong> Nephrology,<br />

Heidelberg, Germany, Stefan Meuer, Director, Institute <strong>of</strong><br />

<strong>Immunology</strong>, Heidelberg, Germany<br />

Background: At present it is unclear which dose <strong>of</strong> Cyclosporine<br />

A (CsA) is optimal with respect to immunosuppressive<br />

efficacy and drug specific side effects at the level <strong>of</strong> the<br />

individual patient. Recently we proposed the quantitative<br />

assessment <strong>of</strong> NFAT regulated gene expression in peripheral<br />

lymphocytes as a new tool to measure the individual degree<br />

<strong>of</strong> immunosuppression by CsA and demonstrated that a<br />

significant correlation exists between suppression <strong>of</strong> NFATregulated<br />

genes and clinical complications such as infections<br />

and malignancies. The reduction <strong>of</strong> CsA dosage under close<br />

pharmacodynamic monitoring may lead to reduced drug<br />

specific side effects while maintaining optimal graft protection.<br />

Methods: Eight stable renal transplant recipients were<br />

enrolled and longitudinally followed for 27 (12–44) months.<br />

Median CsA dose was 1.83 mg/kg/day at the time <strong>of</strong><br />

enrolment and was tapered to 1.19 mg/kg/day. NFATregulated<br />

gene expression was measured in peripheral<br />

blood lymphocytes stimulated with PMA and ionomycin ex<br />

vivo. All patients had an ultrasound guided allograft biopsy.<br />

Results: The stepwise reduction <strong>of</strong> CsA was inversely<br />

correlated to the residual NFAT-regulated gene expression.<br />

In seven patients there were no signs <strong>of</strong> acute rejection in<br />

the whole study period. One patient had a BANFF Ia<br />

rejection. In this patient the mean residual NFAT-regulated<br />

gene expression had increased up to 47% in 6 months prior to<br />

rejection. All patients without rejection had a median NFATregulated<br />

gene expression <strong>of</strong> 25%. Conclusion: The measurement<br />

<strong>of</strong> residual NFAT-related gene expression is a helpful<br />

and reliable tool in individualizing the immunosuppressive<br />

therapy in long-term allograft recipients.<br />

doi:10.1016/j.clim.2007.03.139<br />

Su.101 A Non-allogeneic Stimulus Triggers the<br />

Production <strong>of</strong> De Novo HLA Antibodies in Healthy<br />

Adults<br />

Jose Crispin, PhD Student, Instituto Nacional de Ciencias<br />

Medicas y Nutricion SZ, Mexico City, Mexico, Luis Morales<br />

Buenrostro, Investigator, Department <strong>of</strong> Nephrology,<br />

Instituto Nacional de Ciencias Medicas y Nutricion SZ,<br />

Mexico City, Mexico, Maria Ines Vargas Rojas, PhD Student,<br />

Department <strong>of</strong> <strong>Immunology</strong> and Rheumatology, Instituto<br />

Nacional de Ciencias Medicas y Nutricion SZ, Mexico City,<br />

Mexico, Lluvia Marino Vazquez, Undergraduate Student,<br />

Department <strong>of</strong> Nephrology, Instituto Nacional de Ciencias<br />

Medicas y Nutricion SZ, Mexico City, Mexico, Claudia de Leo,<br />

Chemist, Department <strong>of</strong> Transplantation, Instituto Nacional<br />

de Ciencias Medicas y Nutricion SZ, Mexico City, Mexico,<br />

Josefina Alberu, Head <strong>of</strong> Department, Department <strong>of</strong><br />

Transplantation, Instituto Nacional de Ciencias Medicas y<br />

Nutricion SZ, Mexico City, Mexico<br />

Transplant recipients develop antibodies against a wide<br />

array <strong>of</strong> HLA specificities, and not only against the antigens<br />

to which they have been exposed. The aim <strong>of</strong> the present<br />

study was to establish if HLA-unrelated immune stimulation<br />

is capable <strong>of</strong> triggering the production <strong>of</strong> HLA antibodies. We<br />

determined the presence <strong>of</strong> HLA antibodies in 20 healthy<br />

adults before and after the administration <strong>of</strong> an immune<br />

stimulus (hepatitis B vaccine plus tuberculin skin test). HLA<br />

antibodies were assessed with a flow-cytometry based<br />

system. At baseline, HBsAg antibodies were detected in<br />

protective titers in 75% <strong>of</strong> the subjects; HLA antibodies were<br />

negative in all but one. One week after the antigenic<br />

stimulus, HBsAg antibody titers increased significantly,<br />

without any detectable changes in HLA antibodies. Surprisingly,<br />

HLA antibodies developed in 8 participants 1 month<br />

after the application <strong>of</strong> the stimulus. One additional subject<br />

developed HLA antibodies 1 month later. Therefore 9/20<br />

subjects became PRA positive during the observation period.<br />

Antibody specificities were identified by single-antigen<br />

assay. The alloantibody response elicited by the immune<br />

stimulus was not consistent with memory cell stimulation.<br />

The findings <strong>of</strong> this study demonstrate that a non-HLA<br />

antigenic stimulus is capable <strong>of</strong> eliciting the development <strong>of</strong><br />

HLA antibodies in healthy adults.<br />

doi:10.1016/j.clim.2007.03.140<br />

S175<br />

Su.102 Activation <strong>of</strong> Transcription Factor<br />

PPARγ In Vivo Protects Against Alloreactive Vascular<br />

Remodeling <strong>of</strong> Human Artery<br />

Ivana Kawikova, Associate Research Scientist, Yale School <strong>of</strong><br />

Medicine, New Haven, CT, Yi Tai, Associate Research<br />

Scientist, Transplantation Surgery, New Haven, CT, Marc<br />

Lorber, Pr<strong>of</strong>essor, Transplantation Surgery, Yale School <strong>of</strong><br />

Medicine, New Haven, CT, George Tellides, Associate<br />

Pr<strong>of</strong>essor, Cardiothoracic Surgery, New Haven, CT, Jordan<br />

Pober, Pr<strong>of</strong>essor, Immunobiology, Yale School <strong>of</strong> Medicine,<br />

New Haven, CT, Alfred Bothwell, Pr<strong>of</strong>essor, Immunobiology,<br />

New Haven, CT<br />

PPARγ is a ligand-activated transcription factor that<br />

controls lipogenesis and plays a role in regulation <strong>of</strong> immune<br />

responses. PPARγ ligands inhibited inflammation in several<br />

animal disease models. Here we tested effects <strong>of</strong> PPARγ<br />

ligands in the in vivo model <strong>of</strong> human graft arteriosclerosis<br />

(GA) in humanized (SCID)/beige mice. Human artery was<br />

inserted into mouse abdominal aorta, allowed to heal and<br />

then the mice were injected with alloreactive PBMC (3×108<br />

ip/mouse). This led to the development <strong>of</strong> GA in human<br />

artery within 4 weeks, while mouse vessels remained intact.<br />

Treatment with endogenous PPARγ ligands, 15-deoxy-prostaglandin<br />

J2(15-d-PGJ2; 50 μg/kg ip for 4 weeks) or<br />

pharmacological PPARγ ligand, ciglitazone (2 mg/kg for 4<br />

weeks ip.) prevents the vascular remodeling. PPARN=antagonist,<br />

GW9662 (300 μg/kg ip for 4 weeks), significantly<br />

reduced the protective effects <strong>of</strong> PPARγ ligands suggesting<br />

that the protection is mainly mediated via PPARγ activation.<br />

Since T cells infiltrating GA lesion produce IFNγ that can on<br />

its own induce GA <strong>of</strong> human artery, we tested the effects <strong>of</strong><br />

15-d-PGJ2(50 μg/kg ip. for 4 weeks) also in this model and<br />

found that the treatment inhibited also IFNγ-induced GA. In<br />

summary, activation <strong>of</strong> PPARN=protects human artery<br />

against alloreactive GA and the mechanisms involve effects<br />

on pathways downstream <strong>of</strong> IFNγ. These studies may open<br />

new possibilities for the treatment <strong>of</strong> human graft rejection


S176 Abstracts<br />

since glitazons are PPARγ ligands that are already FDA<br />

approved drugs used for treatment <strong>of</strong> diabetes type 2.<br />

doi:10.1016/j.clim.2007.03.141<br />

Su.103 Potential Role <strong>of</strong> Donor-derived Regulatory<br />

T Cells in the Suppression <strong>of</strong> Alloimmune Responses<br />

Fleur Samantha Benghiat, MD, Institute for Medical<br />

<strong>Immunology</strong>, Gosselies, Belgium<br />

Naturally occurring CD4+CD25+FoxP3+ regulatory cells<br />

(Tregs) are essential for the regulation <strong>of</strong> allogeneic responses.<br />

In view <strong>of</strong> the small numbers <strong>of</strong> circulating Treg cells,<br />

many protocols nowadays attempt to expand recipient Tregs<br />

ex vivo in order to achieve allograft tolerance by infusing<br />

them back to the recipient. But these are tedious procedures.<br />

We instead hypothesized that Tregs coming from the<br />

same donor as the allograft could be easily extracted in large<br />

numbers and could target the graft by recognizing selfantigens<br />

on the graft. For this purpose, in vitro experiments<br />

were performed with C57BL/6 CD4+CD25neg cells as<br />

responders and bm12 bone marrow derived immature<br />

dendritic cells as stimulators. The efficacy <strong>of</strong> syngeneic<br />

(bm12) versus allogeneic (C57BL/6) Tregs in regulating Th1<br />

and Th2 alloresponses were compared. Our results show that<br />

syngeneic Tregs are as efficient as allogeneic Tregs to<br />

suppress IL-2, IFN-g and IL-13 in a dose dependent manner.<br />

Nevertheless, the more Tregs were added, the more the<br />

proliferation – tested by thymidine incorporation – was<br />

enhanced. Using CFSE staining and Thy1.1–Thy1.2 congenic<br />

markers, it was confirmed that proliferating cells were<br />

Foxp3+ Tregs originally coming from naturally occurring Tregs<br />

added to the culture. Treg proliferation was still observed<br />

when CD4+CD25neg alloreactive cells were substituted by<br />

recombinant IL-2. Similar results were observed with human<br />

cells. In conclusion, allogeneic and syngeneic Tregs are<br />

equally efficient in regulating Th1 and Th2 alloresponses in<br />

vitro and proliferate under the cover <strong>of</strong> IL-2. Ongoing experiments<br />

are investigating the role <strong>of</strong> donor Tregs in allograft<br />

acceptance.<br />

doi:10.1016/j.clim.2007.03.142<br />

Su.104 HMGB1 and IL-1 are Endogenous Mediators<br />

Linking Cell Injury to the Adaptive Immune<br />

Response<br />

Deepak A. Rao, MD/PhD Student, Immunobiology, Yale<br />

University School <strong>of</strong> Medicine, New Haven, CT, Jordan S.<br />

Pober, Pr<strong>of</strong>essor and Vice-Chair, Immunobiology, Yale<br />

University School <strong>of</strong> Medicine, New Haven, CT<br />

Pre- or peri-operative allograft injury predisposes to<br />

acute and chronic allograft rejection; however, the mediators<br />

released by injured tissues that promote rejection<br />

remain unclear. We and others have demonstrated that IFNgamma-producing<br />

T cells play a key role in the development<br />

<strong>of</strong> chronic vascular rejection. Here, we report that freezethaw<br />

lysates <strong>of</strong> human umbilical vein endothelial cells (EC)<br />

increase the amount <strong>of</strong> IFN-gamma produced by T cells when<br />

added to co-cultures <strong>of</strong> CD4+ T cells with HLA-DR+ allogeneic<br />

EC. Immunoadsorption <strong>of</strong> HMGB1, a mediator released by<br />

necrotic cells, partly removes activity from the lysates.<br />

Recombinant HMGB1 has no effect on EC but increases IFNgamma<br />

production by CD4+ Tcells activated by allogeneic EC<br />

or by anti-CD3 and anti-CD28 mAbs. However, HMGB1enhanced,<br />

but not basal, IFN-gamma production is markedly<br />

reduced in co-cultures thoroughly depleted <strong>of</strong> monocytes.<br />

HMGB1 preparations rigorously depleted <strong>of</strong> endotoxin act on<br />

monocytes via TLR4 and CD14 to induce IL-1beta release,<br />

and neutralization <strong>of</strong> IL-1beta significantly reduces the<br />

effect <strong>of</strong> HMGB1 on EC-T cell co-cultures containing<br />

monocytes. EC lysates enhance IFN-gamma production in<br />

the absence <strong>of</strong> monocytes, and neutralization <strong>of</strong> IL-1alpha<br />

blocks almost all <strong>of</strong> this activity. Memory T cells express IL-<br />

1R1, and both IL-1alpha and IL-1beta increase IFN-gamma<br />

production from memory CD4+ T cells activated by allogeneic<br />

EC or anti-CD3 and anti-CD28 mAbs. We conclude that<br />

damaged EC may release HMGB1 and IL-1alpha, both <strong>of</strong><br />

which directly co-stimulate T cell production <strong>of</strong> IFN-gamma.<br />

In addition, HMGB1 induces monocyte secretion <strong>of</strong> IL-1beta,<br />

further promoting IFN-gamma production. Supported by the<br />

NIH.<br />

doi:10.1016/j.clim.2007.03.144<br />

Su.105 Alloantigen-specific Treg: Repertoire<br />

Selection, TH17 Cell Differentiation and<br />

Prolongation <strong>of</strong> Organ Allograft Survival<br />

Giorgio Raimondi, Postdoctoral Scholar, Starzl<br />

Transplantation Institute, University <strong>of</strong> Pittsburgh,<br />

Department <strong>of</strong> Surgery, Pittsburgh, PA, Tokita Daisuke,<br />

Postdoctoral Associate, Starzl Transplantation Institute,<br />

University <strong>of</strong> Pittsburgh, Department <strong>of</strong> Surgery,<br />

Pittsburgh, PA, Zhiliang Wang, Research Assistant Pr<strong>of</strong>essor,<br />

Starzl Transplantation Institute, University <strong>of</strong> Pittsburgh,<br />

Department <strong>of</strong> Surgery, Pittsburgh, PA, Tina Supter,<br />

Postdoctoral Associate, Starzl Transplantation Institute,<br />

University <strong>of</strong> Pittsburgh, Department <strong>of</strong> Surgery,<br />

Pittsburgh, PA, Angus Thomson, Pr<strong>of</strong>essor <strong>of</strong> Surgery, Starzl<br />

Transplantation Institute, University <strong>of</strong> Pittsburgh,<br />

Department <strong>of</strong> Surgery, Pittsburgh, PA<br />

Little information exists regarding the selection and<br />

functional evaluation <strong>of</strong> CD4+CD25+Foxp3+ naturally-arising<br />

regulatory T cells (Treg) for promotion <strong>of</strong> transplant<br />

tolerance. We have investigated/optimized procedures for<br />

the selection <strong>of</strong> murine alloantigen (Ag)-specific Treg<br />

(AAsTreg), and tested their function in a MHC-mismatched<br />

heart allograft model. When naïve Treg were stimulated with<br />

mature (LPS-stimulated), allogeneic DC, with a high IL-2<br />

concentration (500 U/ml), significant numbers <strong>of</strong> Foxp3cells<br />

quickly accumulated. Intracellular flow analysis identified<br />

these Foxp3- cells as IL-17 producers (TH17). The<br />

expansion <strong>of</strong> Treg during an MLR supported then the use <strong>of</strong><br />

immature allogeneic DC as stimulators. However, determination<br />

<strong>of</strong> Treg responder frequency (based on CFSE dilution<br />

analysis) during the MLR (immature DC:CD4+ T cells),<br />

revealed that an unexpected high frequency <strong>of</strong> Treg was<br />

induced to proliferate (approximately 27% <strong>of</strong> Foxp3+ cells,<br />

compared with the anticipated 5–8% <strong>of</strong> non-Treg). The high<br />

levels <strong>of</strong> IL-2 accounted for most <strong>of</strong> what we determined to


Abstracts<br />

be an ‘Ag-non-specific’ stimulation, causing the high<br />

precursor frequency measured. Based on these results, we<br />

used immature allogeneic DC and a mixture <strong>of</strong> stimulatory<br />

cytokines (low concentration) to select AAsTreg that proved<br />

extremely powerful Ag-specific inhibitors <strong>of</strong> alloreactive T<br />

cells proliferation (several-fold superior to freshly-isolated<br />

Treg). Moreover, 50% <strong>of</strong> heart transplants in normal animals<br />

given a short course <strong>of</strong> sub-therapeutic rapamycin, and<br />

injected with AAsTreg on day 7 post-transplant, are currently<br />

beating N70 days. Accounting for <strong>of</strong> all these observations<br />

and further optimization <strong>of</strong> the AAsTreg adoptive transfer<br />

strategy will be key to successful implementation <strong>of</strong> this<br />

clinically relevant regimen.<br />

doi:10.1016/j.clim.2007.03.145<br />

Su.107 Polyclonal Anti-thymocyte Globulin Exhibits<br />

Consistent Immunosuppressive Capabilities Beyond<br />

Cell Depletion<br />

Gina LaCorcia, Principal Research Associate, Genzyme Corp.<br />

IMD Biology and Transplant Research, Framingham, MA,<br />

Mark Swistak, Senior Research Associate, Genzyme Corp.<br />

IMD Biology and Transplant Research, Framingham, MA,<br />

Carla Lawendowski, Principal Research Associate, Genzyme<br />

Corp. IMD Biology and Transplant Research, Framingham,<br />

MA, Tim Weeden, Senior Research Associate, Genzyme Corp.<br />

IMD Biology and Transplant Research, Framingham, MA, Su<br />

Duan, Principal Research Associate, Genzyme Corp. IMD<br />

Biology and Transplant Research, Framingham, MA, Sharon<br />

Nahill, Scientific Director, Genzyme Corp. IMD Biology and<br />

Transplant Research, Framingham, MA, John Williams, Vice<br />

President, Genzyme Corp. IMD Biology and Transplant<br />

Research, Framingham, MA, John Dzuris, Senior Scientist,<br />

Genzyme Corp. IMD Biology and Transplant Research,<br />

Framingham, MA<br />

Thymoglobulin® [Anti-thymocyte Globulin (Rabbit)] is a<br />

purified, gamma globulin produced by immunization <strong>of</strong><br />

rabbits with human thymocytes. It is an FDA approved therapy<br />

for acute organ transplant rejection. Although the<br />

mechanism <strong>of</strong> action <strong>of</strong> Thymoglobulin® is not fully understood,<br />

its efficacy is thought to be the result <strong>of</strong> lymphocyte<br />

depletion via cytotoxic antibodies directed against antigens<br />

expressed on human T lymphocytes. Thymoglobulin® also<br />

contains antigen reactivities that may account for downmodulation<br />

<strong>of</strong> T cell activation and lymphocyte trafficking.<br />

The in vitro studies presented here demonstrate some <strong>of</strong> the<br />

immunosuppressive effects <strong>of</strong> different manufactured<br />

batches <strong>of</strong> Thymoglobulin®. We show that culture <strong>of</strong><br />

human PBMC with Thymoglobulin® results in the emergence<br />

<strong>of</strong> an increased population <strong>of</strong> Foxp3+ regulatory T cells (T<br />

reg) which are functional in that they potently suppress in<br />

vitro T cell activation. All 14 retained batches <strong>of</strong> manufactured<br />

Thymoglobulin® tested induced T reg that were<br />

immunosuppressive. Thymoglobulin® also interacts with<br />

many different cell surface receptors in a consistent and<br />

reproducible manner. Flow cytometric analysis demonstrated<br />

that all batches <strong>of</strong> Thymoglobulin® tested were<br />

comparable in binding ability to several cell surface<br />

receptors. One receptor to which Thymoglobulin® has<br />

reactivity is the chemokine receptor CXCR4. We demonstrate<br />

that 14 manufactured batches <strong>of</strong> Thymoglobulin® tested<br />

inhibit chemotaxis <strong>of</strong> the Jurkat Tcell line in response to SDF-1.<br />

These studies provide additional insights into the mechanisms<br />

by which Thymoglobulin® contributes to prevention and<br />

treatment <strong>of</strong> transplant rejection episodes and underscores<br />

the overall consistency <strong>of</strong> the immunosuppressive capability<br />

among different manufactured batches <strong>of</strong> Thymoglobulin®.<br />

doi:10.1016/j.clim.2007.03.146<br />

Su.108 KIR Genotyping <strong>of</strong> the Venezuelan Mestizo<br />

Population by PCR-SSP<br />

Angela Conesa, Bioanalist, Institute <strong>of</strong> <strong>Immunology</strong>,<br />

Caracas, Venezuela, Felix Toro, Biologist, Institute <strong>of</strong><br />

<strong>Immunology</strong>, Caracas, Venezuela, Nubia Silva, Biologist,<br />

Institute <strong>of</strong> <strong>Immunology</strong>, Caracas, Venezuela, Maria-Elena<br />

Marquez, Biologist, Institute <strong>of</strong> <strong>Immunology</strong>, Caracas,<br />

Venezuela, Paolo Tassinari, MD, Institute <strong>of</strong> <strong>Immunology</strong>,<br />

Caracas, Venezuela, Isaac Blanca, Biologist, Institute <strong>of</strong><br />

<strong>Immunology</strong>, Caracas, Venezuela<br />

Killer-cell-Immunoglobulin-like Receptors (KIR) belong to a<br />

family <strong>of</strong> NK cell receptors that recognize MHC-I molecules. 17<br />

KIR genes have been identified and clustered in 2 major<br />

haplotypes (A and B). These haplotypes show variations in<br />

geographical distribution among different human populations.<br />

Goals: a) To standardize the PCR technique for KIR genotyping<br />

in DNA samples, b) To determine KIR gene frequency in the<br />

Venezuelan mestizo population. Methods: Genomic DNA was<br />

isolated from Peripheral Blood Leucocytes. KIR genotyping was<br />

determined by PCR-SSP technique (1). Different experimental<br />

conditions were assessed: primers and DNA sample concentration,<br />

time and temperature <strong>of</strong> annealing and buffer composition.<br />

A commercial kit for KIR genotyping (Invitrogen) was used<br />

as reference assay. Results: We accomplished gene amplification<br />

<strong>of</strong> 14 KIR genes and the pseudogenes 2DP1 and 3DP1A/B.<br />

From 40 DNA samples analyzed, 100% (f=1) were positive for<br />

KIR2DL1, 2DL4, 2DS4, 3DL1 and pseudogenes 2DP1 and 3DP1.<br />

Other KIR showed a gene frequency b1: 2DL2(.28), 2DL3(.78),<br />

2DL5(.5), 2DS1(.39), 2DS2(.28), 2DS3(.18), 2DS5(.29),<br />

3DL2(.78), 3DL3(.73) and 3DS1(.33). Conclusions: a) Our<br />

preliminary results showed that frequency <strong>of</strong> inhibitory KIR<br />

genes was N0,73, except for KIR2DL2 and 2DL5, whereas<br />

frequency <strong>of</strong> the activating KIR2DS4 gene was equal to 1,<br />

showing an A haplotype similar to the Caucasian population,<br />

b) The PCR-SSP technique represents an efficient and low<br />

cost method for KIR genotyping. Key words, KIR, ADN, PCR-<br />

SSP. Supported by FONACIT G-2005000395. (1)Tissue Antigen<br />

2002; 59:184–193.<br />

doi:10.1016/j.clim.2007.03.147<br />

S177<br />

Su.109 Immunological Properties <strong>of</strong> Human<br />

Embryonic Stem Cell-derived Oligodendrocyte<br />

Progenitor Cells<br />

Ross Okamura, Scientist, Geron Corporation, Cell Biology,<br />

Menlo Park, CA, Melinda Au, Senior Research Associate,<br />

Geron Corporation, Cell Biology, Menlo Park, CA, Catherine<br />

Priest, Associate Director, Geron Corporation, Cell Biology,<br />

Menlo Park, CA, Anish Majumdar, Senior Director, Geron


S178 Abstracts<br />

Corporation, Cell Biology, Menlo Park, CA, Jerrod Denham,<br />

Senior Manager, Geron Corporation, Product Development,<br />

Menlo Park, CA<br />

A major concern in the use <strong>of</strong> allotransplantation <strong>of</strong> human<br />

embryonic stem cell (hESC)-derived cell therapies is the<br />

possibility <strong>of</strong> rejection by the host's immune system. We have<br />

previously reported that the undifferentiated hESC lines H1,<br />

H7 and H9 possess unique immunological properties which<br />

may make them less susceptible to rejection. To determine if<br />

differentiation alters the immunogenicity <strong>of</strong> hESCs, we chose<br />

to study hESC-derived oligodendrocyte progenitor cells (OPC)<br />

that have the potential for clinical application to treat<br />

patients with spinal cord injury. Undifferentiated H1 and H1derived<br />

OPCs express low to moderate levels <strong>of</strong> surface HLA-A,<br />

B,C antigens and lack surface expression <strong>of</strong> HLA-DR,DP,DQ<br />

antigens. In an in vitro mixed lymphocyte reaction (MLR)<br />

assay, both hESCs and OPCs stimulated weak T cell proliferation<br />

compared to allogeneic dendritic cells. Both undifferentiated<br />

hESCs (uhESCs) and their differentiated OPC<br />

derivatives were largely resistant to NK cell-mediated lysis.<br />

We have also investigated whether hESC-derived OPCs are<br />

susceptible to the presence <strong>of</strong> anti-Neu5Gc antibodies in<br />

normal human sera. We observed that <strong>of</strong> ten sera from normal<br />

healthy individuals representing all four blood groups, uhESCs<br />

showed no susceptibility to lysis and eight sera did not show<br />

measurable cytotoxicity against OPCs. The same sera when<br />

tested against murine embryonic fibroblasts (MEFs) demonstrated<br />

significant complement-dependent lysis. Taken<br />

together, these data show that the hESC-derived OPCs retain<br />

some <strong>of</strong> the unique immunological properties <strong>of</strong> the parental<br />

cell line from which they were differentiated.<br />

doi:10.1016/j.clim.2007.03.148<br />

Su.110 Adhesion Molecules in Pancreatitisassociated<br />

Lung Injury<br />

Roman Vatseba, Medical University, Lviv, Ukraine, Serge<br />

Chooklin, Medical University, Lviv, Ukraine, Myroslav Lyba,<br />

Medical University, Lviv, Ukraine<br />

Respiratory failure is typical for acute necrotizing pancreatitis.<br />

Unfortunately, the pathogenesis <strong>of</strong> these changes is<br />

fully unknown. The role <strong>of</strong> the endothelium in the human<br />

acute pancreatitis is still unclear. Applying the ELISA<br />

technique, plasma levels <strong>of</strong> soluble E-selectin (sE-selectin)<br />

and sICAM-1 were studied in 51 patients with acute<br />

pancreatitis. Twenty-five <strong>of</strong> them had respiratory complications.<br />

In this study enrolled patients who admitted to clinic<br />

within 48 hours after disease onset. The control group<br />

compiled 10 healthy volunteers. Already after admission the<br />

increased levels <strong>of</strong> adhesion molecules were noted in all<br />

patients. By that, the highest levels were observed in<br />

patients with lung injury. During first 7 days plasma levels<br />

<strong>of</strong> sE-selectin and sICAM-1 practically did not change in<br />

patients with necrotizing pancreatitis, while in patients with<br />

the pancreatitis-associated lung injury its levels gradually<br />

increased starting from the third day. There was a clear<br />

correlation between sE-selectin, sICAM-1 and severity <strong>of</strong><br />

hemodynamic compromise was established. The intensity <strong>of</strong><br />

endothelial activation, measured by adhesion molecules<br />

concentration, considered to be key features <strong>of</strong> systemic<br />

inflammation in severe pancreatitis. These data pointed on<br />

the role <strong>of</strong> adhesion molecules in the pathogenesis <strong>of</strong><br />

pancreatitis-associated lung injury. Blockade <strong>of</strong> ICAM-1 and<br />

E-selectin synthesis and its receptors may be a novel target<br />

in the management <strong>of</strong> pancreatitis-associated lung injury.<br />

doi:10.1016/j.clim.2007.03.149<br />

Su.111 Aire Deficient Mice—A Model for<br />

Autoimmune Lung Disease<br />

Anthony Shum, <strong>Clinical</strong> Fellow, University <strong>of</strong> California, San<br />

Francisco Department <strong>of</strong> Medicine, San Francisco, CA, Jason<br />

DeVoss, Postdoctoral Fellow, University <strong>of</strong> California, San<br />

Francisco Diabetes Center, San Francisco, CA, Mark<br />

Anderson, Assistant Pr<strong>of</strong>essor, University <strong>of</strong> California, San<br />

Francisco Diabetes Center and Department <strong>of</strong> Medicine, San<br />

Francisco, CA<br />

Aire deficient mice have a mutation for the gene encoding<br />

AIRE, a transcription factor that drives the ectopic expression<br />

<strong>of</strong> organ-specific antigens in the thymus. A deficiency in AIRE<br />

results in a breakdown <strong>of</strong> central tolerance toward selfantigens<br />

resulting in organ-specific autoimmune disease. The<br />

AIRE knockout mouse is a model for Autoimmune Polyglandular<br />

Syndrome Type 1, a disorder characterized by polyglandular<br />

autoimmune disease. Pulmonary manifestations,<br />

while rare, have been reported, including lymphocytic<br />

interstitial pneumonitis and bronchiolitis obliterans organizing<br />

pneumonia. Our goals are to 1) understand the mechanism<br />

<strong>of</strong> autoimmune-mediated attack and its relationship to the<br />

thymus 2) identify lung specific auto-antigens. Methods: AIRE<br />

knockout mice were examined for lung histology and<br />

immunohistochemistry. Serum from knockout mice was used<br />

for immunoblotting and indirect immun<strong>of</strong>luorescence. Flow<br />

cytometry and intracellular cytokine staining was performed<br />

on lymphocytes isolated from AIRE knockout lungs. Results:<br />

Lung histology reveals perivascular and peribronchiolar<br />

mononuclear inflammation. Immunoblots reveal a predominant<br />

antigen at 84 kilodaltons. The presence <strong>of</strong> autoantibody<br />

to this antigen correlates with the presence <strong>of</strong> histologic<br />

infiltrates. Indirect immun<strong>of</strong>luorescence on lung sections<br />

from unaffected mice show staining <strong>of</strong> bronchiolar epithelium.<br />

Immune cell subtypes visualized on sections <strong>of</strong> knockout<br />

mice reveal a predominance <strong>of</strong> CD4+ cells. Flow cytometric<br />

analysis <strong>of</strong> lymphocytes from lungs <strong>of</strong> knockout mice confirm<br />

the predominance <strong>of</strong> CD4+ T cells. Intracellular cytokine<br />

staining <strong>of</strong> lymphocytes reveal the predominance <strong>of</strong> the TH1<br />

cytokine IFN-γ. Conclusions: AIRE deficient mice are a<br />

promising model <strong>of</strong> spontaneous autoimmune lung disease<br />

<strong>of</strong>fering the potential to identify lung auto-antigens.<br />

doi:10.1016/j.clim.2007.03.150<br />

Su.112 The XX Sex Chromosome Complement, as<br />

compared to the XY, Confers Greater Susceptibility to<br />

Experimental Autoimmune Diseases — EAE and SLE<br />

Deborah Smith, Doctoral Candidate, University <strong>of</strong> California,<br />

Los Angeles Neuroscience, Los Angeles, CA, Sienmi Du,<br />

Research Assistant, University <strong>of</strong> California, Los Angeles


Abstracts<br />

Neurology, Los Angeles, CA, Seema Tiwari-Woodruff,<br />

Associate Pr<strong>of</strong>essor, University <strong>of</strong> California, Los Angeles<br />

Neurology, Los Angeles, CA, Arthur P. Arnold, Chair,<br />

Distinguished Pr<strong>of</strong>essor, University <strong>of</strong> California, Los Angeles<br />

Physiological Science, Los Angeles, CA, Jennifer K. King,<br />

Post-Residency Fellow, University <strong>of</strong> California, Los Angeles,<br />

Rheumatology, Los Angeles, CA, Ram Raj Singh, Pr<strong>of</strong>essor<br />

<strong>of</strong> Medicine, University <strong>of</strong> California, Los Angeles Medicine-<br />

Rheumatology, Los Angeles, CA, Rhonda R. Voskuhl,<br />

Pr<strong>of</strong>essor in Residence, University <strong>of</strong> California, Los<br />

Angeles Neurology, Los Angeles, CA<br />

The majority <strong>of</strong> autoimmune diseases are more common<br />

in women as compared to men. This may be attributed to<br />

differences in sex hormones, sex chromosomes or both. To<br />

determine the contribution <strong>of</strong> sex chromosomes to sex<br />

differences in susceptibility to autoimmune disease, we used<br />

animal models characterized by a known female preponderance,<br />

experimental autoimmune encephalomyelitis (EAE)<br />

and pristane-induced lupus, each in SJL mice. Mice which<br />

have Sry, the gene that encodes for testicular development,<br />

deleted from the Y chromosome were backcrossed onto the<br />

SJL strain, resulting in EAE and lupus-susceptible mice which<br />

differ in their complement <strong>of</strong> sex chromosomes, while having<br />

the same gonadal type, thereby permitting assessment <strong>of</strong><br />

sex chromosome effects not confounded by differences in<br />

sex hormones (ovary bearing XX vs. XY- mice). When<br />

comparing EAE in ovariectomized XX vs. XY- SJL mice,<br />

disease was more severe and there was more inflammation in<br />

the spinal cords <strong>of</strong> mice with the XX sex chromosome<br />

complement. Also, when lupus was induced in ovariectomized<br />

XX vs. XY- SJL mice, mortality, nephritis and autoantibody<br />

levels were each greater in mice with the XX sex<br />

chromosome complement. The differences in susceptibility<br />

to EAE and lupus also occurred when comparing castrated XX<br />

Sry versus XY- Sry mice (testis bearing mice). This is the first<br />

evidence that XX sex chromosome complement, as compared<br />

to XY, confers greater susceptibility to two immunopathologically<br />

distinct diseases. This may be relevant to the<br />

increased susceptibility <strong>of</strong> women to a variety <strong>of</strong> distinct<br />

autoimmune diseases.<br />

doi:10.1016/j.clim.2007.03.151<br />

Su.113 Expression <strong>of</strong> the Renal Antigen Neprhin by<br />

Human Medullary Thymic Epithelial Cells<br />

Michael Tasch, Senior Fellow, University <strong>of</strong> Washington,<br />

Department <strong>of</strong> Medicine, Seattle, WA, Kimberly Muczynski,<br />

Associate Pr<strong>of</strong>essor, University <strong>of</strong> Washington, Department<br />

<strong>of</strong> Medicine, Seattle, WA, Anne Stevens, Assistant Pr<strong>of</strong>essor,<br />

University <strong>of</strong> Washington, Department <strong>of</strong> Pediatrics,<br />

Seattle, WA, J. Lee Nelson, Pr<strong>of</strong>essor, Fred Hutchinson<br />

Cancer Research Center, Division <strong>of</strong> <strong>Clinical</strong> Research,<br />

Seattle, WA<br />

Idiopathic nephrotic syndrome, a set <strong>of</strong> glomerular pathologies<br />

<strong>of</strong>ten leading to end-stage renal disease, is associated<br />

with disturbances in Tcell function. This has led to the<br />

view that these diseases are a consequence <strong>of</strong> T cell<br />

mediated autoimmunity, though the relevant autoantigen(s)<br />

remain unidentified. Nephrin is a possible target for<br />

autoimmunity in the kidney. The protein product <strong>of</strong> the<br />

NPHS1 gene and an essential component <strong>of</strong> the glomerular<br />

filtration barrier, nephrin is expressed proximal to autoimmune<br />

renal lesions and conforms to the definition <strong>of</strong> a<br />

tissue restricted antigen (TRA). Since intrathymic expression<br />

<strong>of</strong> other TRAs has been implicated in the establishment <strong>of</strong><br />

central tolerance and protection from organ-specific autoimmunity,<br />

we hypothesized that nephrin is expressed by cells<br />

<strong>of</strong> the human thymus. RT-PCR analysis showed that NPHS1<br />

mRNA is present in all human thymi (N=12) tested. Using<br />

primer sets flanking sequences encoding immunoglobulinlike<br />

domains, we showed that the thymic-specific transcript<br />

appears identical to that from kidney. FACS-purified CD326+,<br />

HLA-DR+ medullary thymic epithelial cells (mTEC) exhibited<br />

the highest concentration <strong>of</strong> NPHS1 transcripts. Immun<strong>of</strong>luorescence<br />

analysis showed that nephrin protein is<br />

expressed in thymus sections. Nephrin exhibited a predominantly<br />

medullary pattern <strong>of</strong> expression, co-localizing with<br />

HLA-DR and cytokeratin, consistent with mTEC, although<br />

infrequent nephrin-positive cells were seen in the cortex and<br />

capsule also. Nephrin staining also showed distinct intracellular<br />

patterns including surface, intracellular granules and<br />

punctate structures. These data provide a strong rationale<br />

for exploring nephrin's role in negative selection and the role<br />

<strong>of</strong> nephrin-specific T cells in human nephrotic syndrome.<br />

doi:10.1016/j.clim.2007.03.153<br />

S179<br />

Su.114 Low-dose <strong>of</strong> Local Radiation Treatment Can<br />

Inhibit the Progression <strong>of</strong> Experimental<br />

Autoimmune Nephritis by Promoting Apoptosis<br />

M. S. Razzaque, Research Scientist, Department <strong>of</strong><br />

Pathology, Nagasaki University, Nagasaki, Japan, L. Diange,<br />

Research Scientist, Department <strong>of</strong> Pathology, Nagasaki<br />

University, Nagasaki, Japan, A. Nazneen, Research<br />

Scientist, Department <strong>of</strong> Pathology, Nagasaki University,<br />

Nagasaki, Japan, T. Taguchi, Pr<strong>of</strong>essor, Department <strong>of</strong><br />

Pathology, Nagasaki University, Nagasaki, Japan<br />

Autoimmune crescentic glomerulonephritis (CrGN) is a<br />

rapidly progressive form <strong>of</strong> nephritis and usually resistant to<br />

therapeutic intervention. Apoptosis plays important role in<br />

resolution <strong>of</strong> CrGN. We investigated the effects <strong>of</strong> local<br />

radiation treatment in the progression <strong>of</strong> CrGN in rats. Three<br />

experimental groups were studied; Group-I: sham-operated<br />

control (n=12); Group-II: intravenous injection <strong>of</strong> rabbit<br />

anti-rat GBM antibody (nephrotoxic serum, NTS) (n=23); and<br />

Group-III: a single low-dose <strong>of</strong> 0.5 Gy X-ray radiation<br />

treatment to local bilateral kidneys at 6, 13, 20, and<br />

27 days after NTS injection (n=55). In Group-III, the level<br />

<strong>of</strong> BUN and serum creatinine was significantly decreased<br />

after radiation treatment compared with age-matched<br />

Group-II nephritic rats (pN0.05 or 0.001). Glomerular<br />

hypercellularity, crescents, global sclerosis and tubulointerstitial<br />

damage gradually developed throughout the time<br />

course in Group-II rats, were significantly less (PN 0.05 or<br />

0.001) after radiation treatment in Group-III. The expression<br />

<strong>of</strong> active caspases-3 and -7 was increased for irradiated<br />

kidneys, compared with their corresponding Group-II rats.<br />

Western blot analysis showed that 17 kDa active caspase-3<br />

and 35 kDa active caspase-7 expressions markedly increased


S180 Abstracts<br />

in Group-III kidneys, compared with the Group-II. Increased<br />

expression <strong>of</strong> p53 protein was also observed in radiation<br />

treated kidneys compared with groups Group-II. Low-dose <strong>of</strong><br />

local radiation to the kidneys, can delay the progression <strong>of</strong><br />

CrGN in rats by p53-dependent radiation-induced apoptosis.<br />

Caspases-3 and -7 are the key mediators in such apoptosis.<br />

This study shows that radiation treatment is effective in<br />

preventing acute glomerular inflammation and crescent<br />

formation in the rat model <strong>of</strong> autoimmune CrGN.<br />

doi:10.1016/j.clim.2007.03.154<br />

Su.116 IL-1beta, MMP-1, and MMP-3 Transcript<br />

Levels are Associated with CD3delta Expression in<br />

the Osteoarthritic Synovial Membrane<br />

Carla Scanzello, Rheumatology Fellow, Department <strong>of</strong><br />

Rheumatology, Hospital for Special Surgery, New York, NY,<br />

Andrew Pearle, Assistant Attending Orthopedic Surgeon,<br />

Department <strong>of</strong> Orthopedics, Hospital for Special Surgery,<br />

New York, NY, Mary Crow, Attending Physician, Department<br />

<strong>of</strong> Rheumatology, Hospital for Special Surgery, New York,<br />

NY, Edward DiCarlo, Associate Attending Pathologist,<br />

Department <strong>of</strong> Laboratory Medicine, Hospital for Special<br />

Surgery, New York, NY<br />

Many patients with osteoarthritis (OA) develop inflammatory<br />

infiltrates containing T lymphocytes within the<br />

synovial membrane (SM). These infiltrates have the<br />

potential to augment cytokine (i.e.IL-1beta and TNFalpha)<br />

and degradative enzyme (i.e. MMPs) production by<br />

macrophages or synovial fibroblasts. Purpose: The aim <strong>of</strong><br />

this study was to determine if lymphocytic synovial<br />

infiltration in patients with OA is associated with increased<br />

cytokines and enzyme expression implicated in cartilage<br />

catabolism. Methods: SM specimens from 37 OA patients<br />

undergoing total hip or knee arthroplasty (n=31) or<br />

arthroscopy (n=6) were obtained. Total RNA was extracted<br />

and cDNA synthesized. Expression levels <strong>of</strong> CD3delta, IL-<br />

1beta, TNF-alpha, IL-15, IL-6, IL-10, TGF-beta, MMP-1, and<br />

MMP-3, were measured by real-time PCR. Synovial inflammation<br />

was graded in H&E sections on a four-point scale.<br />

Correlation coefficients were calculated to determine if<br />

cytokine and enzyme mRNA levels varied with CD3delta<br />

expression or histologic score. Results: CD3delta transcript<br />

levels correlated most significantly with IL-1beta (r=0.677,<br />

pb0.0001), but were also associated with TNF-alpha<br />

(r=0.487, p=0.003), IL-15 (r=0.524, p=0.001), IL-10<br />

(r=0.570, p=0.0004), and TGF-beta (r=0.381, p=0.022).<br />

CD3delta levels were also associated with MMP-1<br />

(r=0.432, p=0.008) and MMP-3 (r=0.506, r=0.0014) transcripts.<br />

In addition, MMP-1 expression was significantly<br />

associated with increasing histologic inflammatory score<br />

(r=0.412, p=0.011). Conclusions: mRNA levels <strong>of</strong> IL-1beta,<br />

MMP-1 and MMP-3 correlated with levels <strong>of</strong> CD3delta<br />

transcripts within the SM <strong>of</strong> patients with both knee and<br />

hip OA.These relationships suggest that synovial infiltrating<br />

T cells may contribute to disease pathogenesis in OA by<br />

promoting synthesis <strong>of</strong> these mediators <strong>of</strong> joint damage.<br />

doi:10.1016/j.clim.2007.03.155<br />

Su.117 An In Silico Model <strong>of</strong> Regulatory T Cell<br />

Function<br />

Paul Taylor, Postdoctoral Researcher, La Jolla Institute for<br />

Allergy and <strong>Immunology</strong>, San Diego, CA, Matthias von<br />

Herrath, Primary Investigator, La Jolla Institute for Allergy<br />

and <strong>Immunology</strong>, San Diego, CA<br />

Regulatory T (Treg) cells participate in response to<br />

infection reported so far involve chronic or persistent viral<br />

infections. The findings that Treg cells influence the functional<br />

immunity during viral infections however, might<br />

indicate that, in some cases, virus-specific Treg cells not<br />

only influence immune pathology or prevent pathogen elimination<br />

but also can promote a generalized state <strong>of</strong><br />

immuno-suppression in vivo such that the host is more<br />

susceptible to secondary infections with other pathogens or<br />

has reduced tumour resistance. The regulatory mechanism by<br />

which Tregs act has yet to be definitively characterized.<br />

There is opposing evidence to suggest either cell-to-cell<br />

contact being required with the cell being suppressed or a<br />

cytokine mediated mechanism, either through immunosuppressive<br />

cytokines or by acting as a growth factor ‘sink’. We<br />

have developed a cellular automaton that is an in silico representation<br />

<strong>of</strong> the cells <strong>of</strong> the immune system. The<br />

automaton aims to faithfully simulate these cells and models<br />

their interaction with cognate rule sets derived from in vitro<br />

and in vivo experimentation. The automaton has been<br />

developed to provide a stochastic model <strong>of</strong> a Treg suppression<br />

assay which can be used to investigate the possible mechanisms<br />

behind their immuno-suppressive effect. The model can<br />

rapidly simulate a large number <strong>of</strong> assays under a wide range<br />

<strong>of</strong> parameters and the detailed results allow for the analysis<br />

<strong>of</strong> the parameters at a detail level currently impossible in<br />

vitro. The insights obtained by using the automaton provide a<br />

powerful tool for directing in vitro research into Treg<br />

function.<br />

doi:10.1016/j.clim.2007.03.156<br />

Su.118 Data Analysis Protocols for Identifying<br />

Cytokine Signatures in Breast Cancer and Type 1<br />

Diabetes<br />

Janet Siebert, Data Architect, CytoAnalytics, Denver, CO,<br />

Margaret Inokuma, Scientist, BD Biosciences, San Jose, CA,<br />

Nathan Pennock, Pr<strong>of</strong>essional Research Assistant, Webb-<br />

Waring Institute, The University <strong>of</strong> Colorado Denver and<br />

Health Sciences Center, Denver, CO, Dan Waid, Researcher,<br />

Webb-Waring Institute, Denver, CO, Corazon dela Rosa,<br />

Research Scientist, Medicine/Oncology/Tumor Vaccine<br />

Group, University <strong>of</strong> Washington, Seattle, WA, Mary Disis,<br />

Pr<strong>of</strong>essor, Medicine/Oncology/Tumor Vaccine Group,<br />

University <strong>of</strong> Washington, Seattle, WA, Jack Dunne,<br />

Research Scientist, BD Biosciences, San Jose, CA, David<br />

Wagner, Assistant Pr<strong>of</strong>essor, Webb-Waring Institute, The<br />

University <strong>of</strong> Colorado Denver and Health Sciences Center,<br />

Denver, CO, Holden Maecker, Research Scientist, BD<br />

Biosciences, San Jose, CA<br />

Large clinical studies <strong>of</strong> cytokine expression generate<br />

complex data sets which can be difficult to analyze. With the<br />

identification <strong>of</strong> cytokine signatures and biomarkers a goal <strong>of</strong>


Abstracts<br />

much immunodiagnostic research, fast and effective methods<br />

for extracting meaning from the data are needed. This<br />

interdisciplinary study presents several data analysis protocols<br />

for identfying cytokine signatures. Two different data<br />

sets are considered. The first examines cytokine signatures<br />

<strong>of</strong> T cell responses to tumor antigens in breast cancer<br />

patients (IFNγ, IL-2, and TNFα as expressed by CD4+ and<br />

CD8+ T cells). The second explores cytokine signatures <strong>of</strong><br />

Type 1 diabetes patients (IFNγ, IL-1b, IL-2, IL-4, IL-5, IL-6, IL-<br />

8, IL-10, TNFα and TNFβ; on T cells). In the breast cancer<br />

study, a successful response to a foreign antigen (CMV<br />

antigen in CMV seropositive donors) was compared to an<br />

unsuccessful response to a self antigen (CEA, Her2/neu, and<br />

MAGE-3). The differing magnitude <strong>of</strong> the cytokine responses<br />

required data normalization techniques to support visual and<br />

statistical comparison <strong>of</strong> the cytokine responses. In the Type<br />

1 diabetes study, measurements were collected for 10<br />

cytokines and 4 activation environments, for a total <strong>of</strong> 40<br />

different measurements per donor. High-throughput data<br />

analysis techniques allowed quick identification <strong>of</strong> significant<br />

differences between healthy and diseased cohorts, and<br />

the exploration <strong>of</strong> differing donor-level signature patterns.<br />

Taken together, these protocols demonstrate effective<br />

methods for identifying signature patterns <strong>of</strong> multiple<br />

cytokines in complex data sets.<br />

doi:10.1016/j.clim.2007.03.157<br />

Su.119 Carbon Monoxide Generated by Heme<br />

Oxygenase-1 Activity Confers Tolerogenic Capacity<br />

to Dendritic Cells<br />

Christine Chauveau, Postdoctoral Fellow, ITERT-INSERM<br />

U643, Nantes, France, Régis Brion, Technician, ITERT-INSERM<br />

U643, New York, AK, Séverine Remy, Engineer, ITERT-INSERM<br />

U643, Nantes, France, Marcelo Hill, Postdoctoral Fellow,<br />

ITERT-INSERM U643, Nantes, France, Pierre-Joseph Royer,<br />

Postdoctoral Fellow, INSERM U601, Nantes, France, Séverine<br />

Tanguy-Royer, PhD, ITERT-INSERM U643, Nantes, France,<br />

Laurent Tesson, Engineer, ITERT-INSERM U643, Nantes,<br />

France, Roberto Motterlini, GroupLeader, Northwick Park<br />

Hospital, Harrow, Roberta Foresti, Group Leader, Northwick<br />

Park Hospital, Harrow, Ignacio Anegon, Group Leader,<br />

ITERT-INSERM U643, Nantes, France<br />

Heme oxygenase -1 (HO-1) is the inducible heme oxygenase<br />

that catabolizes the degradation <strong>of</strong> heme into<br />

biliverdin Fe 2+ and carbon monoxide (CO). Biliverdin is<br />

subsequently reduced into bilirubin by biliverdin reductase,<br />

and Fe2+ induces the expression <strong>of</strong> ferritin. We have<br />

recently described that immature DCs express HO-1 and<br />

that the expression <strong>of</strong> HO-1 confers tolerogenic capacity to<br />

DCs. We now demonstrate that treatment <strong>of</strong> human<br />

monocyte-derived DCs by exogenous CO blocks LPS-induced<br />

increases in MHC II, CD83, CD80, CD86 expression and proinflammatory<br />

IL-12 cytokine production associated with DC<br />

maturation, while preserving the expression <strong>of</strong> the antiinflammatory<br />

cytokine IL-10. CO treated DCs also display a<br />

reduced capacity to induce T cell allogeneic proliferation in<br />

vitro. In contrast, treatment <strong>of</strong> DCs with bilirubin, biliverdin<br />

and deferoxamine (which mimics the action <strong>of</strong> ferritin) had<br />

no effect on DC phenotype or cytokine production. These<br />

observations indicate that the effect <strong>of</strong> HO-1 on DC function<br />

is mediated through CO. HO-1 induced the expression <strong>of</strong> IL-<br />

10 and the inhibition <strong>of</strong> DC maturation by HO-1 was partially<br />

prevented by anti-IL-10 antibodies. Preliminary results show<br />

that HO-1 expression by DCs inhibits autoimmune diabetes<br />

in a transgenic model. In conclusion, we show the capacity<br />

<strong>of</strong> CO generated by HO-1 activity to block DC maturation<br />

and to inhibit pro-inflammatory and allogeneic immune<br />

responses while preserving IL-10 production. This novel<br />

immune function for CO may be <strong>of</strong> interest for the inhibition<br />

<strong>of</strong> immune responses in autoimmune diseases, transplantation,<br />

and other conditions involving activation <strong>of</strong> the<br />

immune system.<br />

doi:10.1016/j.clim.2007.03.158<br />

Su.120 Real-time Characterization <strong>of</strong> Antibodies<br />

and Biomarker Detection in the Study <strong>of</strong><br />

Immune-based Disease<br />

Jean-Francois Houle, Director, Axela Biosensors, Toronto,<br />

ON, Canada<br />

We describe a versatile platform for more informed decisions<br />

in assay development for biomarker detection and<br />

characterization <strong>of</strong> the etiological agents <strong>of</strong> some immune<br />

disorders and monitoring responses to therapeutic interventions.<br />

Diffraction-based sensing is a simple, sensitive technique<br />

for detecting real-time biomolecular interactions in<br />

complex media. Briefly, capture molecules or substrates are<br />

immobilized on a flat surface in a specific optimized grating<br />

that produces a strong diffraction pattern when illuminated.<br />

Binding <strong>of</strong> biomolecules to the patterned capture molecules<br />

causes a change in the diffractive properties <strong>of</strong> the pattern,<br />

which increases the diffracted signal intensity. Conversely,<br />

release <strong>of</strong> the substrate or interacting species leads to a<br />

measurable decrease in signal. We have used this ability to<br />

continuously observe antibody–antigen interactions as a<br />

means to significantly accelerate immunoassay development.<br />

Representative examples include antibody characterization,<br />

activity measurements, antibody mapping and<br />

pairing studies, buffer optimization for assays as well as<br />

quantitative analyte measurements. Furthermore, we have<br />

demonstrated simultaneous measurement <strong>of</strong> biomarkers<br />

from high micromolar through single-digit picomolar levels<br />

in a single sample.<br />

doi:10.1016/j.clim.2007.03.159<br />

S181<br />

Su.121 Inhibition <strong>of</strong> CXCR2 Expression in HL60 Cells<br />

by CXCR2 Antisense RNA<br />

Lingjie Liao, Postdoctoral Researcher, Department <strong>of</strong><br />

Pediatrics, Tongji Hospital, Huazhong University <strong>of</strong> Science<br />

and Technology, Wuhan, China, Wei Wang, Postgraduate<br />

Student, Department <strong>of</strong> Pediatrics, Tongji Hospital,<br />

Huazhong University <strong>of</strong> Science and Technology, Wuhan,<br />

China, Qin Ning, Chief Physician, Department <strong>of</strong> Infectious<br />

Disease, Tongji Hospital, Huazhong University <strong>of</strong> Science<br />

and Technology, Wuhan, China, Xiaoping Luo, Chief<br />

Physician, Department <strong>of</strong> Pediatrics, Tongji Hospital,<br />

Huazhong University <strong>of</strong> Science and Technology, Wuhan,


S182 Abstracts<br />

China, Guanxin Shen, Pr<strong>of</strong>essor, Department <strong>of</strong> <strong>Immunology</strong>,<br />

Tongji Medical College, Huazhong University <strong>of</strong> Science and<br />

Technology, Wuhan, China<br />

Summary: CXC chemokine receptor 2 (CXCR2) is the<br />

common receptor <strong>of</strong> several potent chemokines, which plays<br />

an important role in the neutrophil-mediated inflammation.<br />

In this study, we investigated the effect <strong>of</strong> CXCR2 antisense<br />

RNA on the gene expression and chemotactic activity in<br />

promyelocytic HL60 cells. A human CXCR2 antisense RNA<br />

recombinant vector, pcDNA-ASCXCR2 complementary to nt<br />

−21∼ nt 1080 <strong>of</strong> CXCR2 was constructed and transfected into<br />

HL60 cells by electroporation. The stable transfected clones<br />

were screened by G418. The expression <strong>of</strong> CXCR2 gene was<br />

measured by RT-PCR and immunohistochemistry. Chemotactic<br />

response to IL-8 was determined using Boyden chamber<br />

procedure. CXCR2 expression in pcDNA-ASCXCR2 transfected<br />

HL60 cells was significantly decreased at mRNA and protein<br />

levels, as compared with those <strong>of</strong> untransfected cells.<br />

Furthermore, chemotactic indexes <strong>of</strong> HL60 cells were<br />

markedly decreased by pcDNA-ASCXCR2 transfection. In<br />

conclusion, CXCR2 antisense RNA can suppress chemotactic<br />

activity through depleting CXCR2 expression on plasma<br />

membrane <strong>of</strong> targeted cells, which would be a new<br />

therapeutic strategy for inflammatory diseases. This work<br />

was supported by the NSFC National Science Fund<br />

£¨No.30571643£No.30672380£©, and National Key Basic<br />

Research Program <strong>of</strong> China (2005CB522901, 2005CB522507)<br />

and <strong>Clinical</strong> Subject Key Project from the Ministry <strong>of</strong> Health.<br />

doi:10.1016/j.clim.2007.03.160<br />

Su.122 Functional Polymorphisms <strong>of</strong> MICA and MICB<br />

in NK Cell Activation<br />

Amonrat Jumnainsong, PhD Candidate, Faculty <strong>of</strong> Medical<br />

Technology, Mahidol University, Bangkok, Thailand, Khon<br />

Kaen, Thailand, Hugh Reyburn, Department <strong>of</strong> Pathology,<br />

University <strong>of</strong> Cambridge, Cambridge, MA, Mar Vales-Gomez,<br />

Department <strong>of</strong> Pathology, University <strong>of</strong> Cambridge,<br />

Cambridge, MA, Chanvit Leelayuwat, Assistant Pr<strong>of</strong>essor,<br />

Department <strong>of</strong> <strong>Clinical</strong> <strong>Immunology</strong>, Khon Kaen University,<br />

Khon Kaen, Thailand<br />

MICA and MICB are ligands <strong>of</strong> NKG2D that expressed on the<br />

cell surface <strong>of</strong> many immune cells. The MIC expressions are<br />

increased in response to cellular stress including tumor<br />

transformation and viral infection. Although MICA and MICB<br />

genes are highly polymorphic and associated with some<br />

diseases, the evidence demonstrating different capability <strong>of</strong><br />

diverse alleles to activate NK cells is rare. In this study, we<br />

investigated the effect <strong>of</strong> various MICA and MICB alleles in NK<br />

cell activation. C1R stable transfected cells expressing<br />

different MICA or MICB alleles were tested for NK cell<br />

activation by chromium releasing assay and IFN-f× production.<br />

MICA*004 transfectants expressing MICA at a higher level<br />

could induce NK cells less than transfectants expressing<br />

MICA*01801 and 045. It is known that MICA amino acid at<br />

position 129 affects NKG2D binding. The methionine at this<br />

position can bind to NKG2D stronger than that <strong>of</strong> valine.<br />

MICA*004 carries valine whereas MICA*01801 and 045 carry<br />

methionine at this position. Thus, it is conceivable that this<br />

position may also affect NK cell activation. MICB*002 and<br />

MICB*004 transfectants could express MICB at the same level<br />

but MICB*002 expressing cells could activate NK cells<br />

stronger than those <strong>of</strong> MICB*004. From crystal structure <strong>of</strong><br />

NKG2D-MIC binding, position 52 in the alpha 1 domain is<br />

located near the NKG2D binding site. Thus, the alteration <strong>of</strong><br />

charge between aspartic acid and asparagine (in MICB*004)<br />

may affect NK cell activation. These results suggested that<br />

the MICA/B polymorphisms affecting NK cell activation<br />

could be relevant to diseases and transplantations.<br />

doi:10.1016/j.clim.2007.03.161<br />

Su.123 The Role <strong>of</strong> Regulatory T Cells in the<br />

Pathogenesis <strong>of</strong> Experimental Biliary Atresia<br />

Alexander Miethke, <strong>Clinical</strong> Fellow, Cincinnati Children’s<br />

Hospital Medical Center, Pediatric Gastroenterology,<br />

Cincinnati, OH, Pranavkumar Shivakumar, Research<br />

Associate, Cincinnati Children’s Hospital Medical Center,<br />

Division <strong>of</strong> Pediatric Gastroenterology, Cincinnati, OH,<br />

Jorge Bezerra, Pr<strong>of</strong>essor <strong>of</strong> Pediatrics, Cincinnati Children’s<br />

Hospital Medical Center, Division <strong>of</strong> Pediatric<br />

Gastroenterology, Cincinnati, OH<br />

In rotavirus induced experimental biliary atresia (BA) the<br />

extraheaptic bile ducts <strong>of</strong> newborn mice undergo a Th1regulated<br />

fibroinflammatory obstruction similar to BA in<br />

children. Here we investigate the role <strong>of</strong> regulatory T (Treg)<br />

cells in the pathogenesis <strong>of</strong> experimental BA. Using 3 color<br />

flow cytometry identifying CD4+CD25+Foxp3+, CD4+CD25+<br />

CD103+ and CD4+CD25+CTLA-4+ populations, we found that<br />

Treg cells had low abundance in livers <strong>of</strong> 3-day-old mice as<br />

compared to adult mice. Infection <strong>of</strong> neonatal mice with<br />

rotavirus did not induce an emergence <strong>of</strong> Treg cells in the liver<br />

within 3 days post infection. However, 7 days after infection,<br />

at a time when neonatal mice develop jaundice, acholic<br />

stools and bilirubinuria, CD4+CD25+ lymphocytes co-staining<br />

for Foxp3+, CD103+ and CTLA-4+ increased by 3-, 10- and 43fold,<br />

respectively in the liver when compared to age-matched<br />

uninfected controls. Real-time PCR analysis <strong>of</strong> purified<br />

lymphocyte subsets revealed a 970 fold increase <strong>of</strong> IL-10<br />

mRNA expression in CD4+CD25+ Treg cells from livers <strong>of</strong> RRV<br />

infected mice compared to saline controls; however, similar<br />

increase in TGF β1 expression was not found. In conclusion,<br />

postnatal viral challenge did not induce a rise in Treg cells<br />

within 3 days <strong>of</strong> life. While there was an increased expression<br />

<strong>of</strong> functional markers (CTLA-4 and IL-10), activated Treg cells<br />

failed to surge TGF β1 mRNA expression at the time <strong>of</strong> duct<br />

obstruction. We speculate that a deficit in number and<br />

selective function <strong>of</strong> neonatal Treg cells may be responsible<br />

for the susceptibility <strong>of</strong> neonates to biliary atresia.<br />

doi:10.1016/j.clim.2007.03.162<br />

Su.124 Association <strong>of</strong> HLA DRB1*04 and HLA<br />

DQB1*06 with Severe Early Childhood Caries<br />

Hossein Neamatollahi, Dentist, Pediatric Department,<br />

School <strong>of</strong> Dentistry, Mashhad, University <strong>of</strong> Medical<br />

Sciences, Mashhad, Iran, Jalil Tavakkol-Afshari, Vice<br />

President for Research, Head, Department <strong>of</strong>


Abstracts<br />

Immunogenetics and Cell Culture, Bu-Ali Research Institute<br />

(BARI), Department <strong>of</strong> Immunogenetics and Cell Culture,<br />

Mashhad, Iran, Alireza Sarraf, Dentist, Pediatric<br />

Department, School <strong>of</strong> Dentistry, Mashhad University <strong>of</strong><br />

Medical Sciences, Mashhad, Iran, Ali Bagherian, Dentist,<br />

School <strong>of</strong> Dentistry, Mashhad University <strong>of</strong> Medical<br />

Sciences, Mashhad, Iran, Habibollah Esmaili,<br />

Epidemiologist, Epidemiology and Statistics Department,<br />

Ghaem Medical Center, Mashhad University <strong>of</strong> Medical<br />

Sciences, Mashhad, Iran, Nasrin Moheghi, Researcher, Bu-<br />

Ali Research Institute (BARI), Department <strong>of</strong><br />

Immunogenetics, Mashhad, Iran<br />

Severe early childhood caries (SECC) is one <strong>of</strong> the most<br />

common diseases in childhood. Etiology <strong>of</strong> SECC is multifactorial<br />

and both genetic and environmental factors play<br />

important roles in the pathogenesis <strong>of</strong> disease. Genetic<br />

variation <strong>of</strong> the host may contribute to the different risks <strong>of</strong><br />

dental caries. Genetic factors such as the Human Leukocyte<br />

Antigen (HLA) have been recently introduced as predisposing<br />

factors. The aim <strong>of</strong> this study was looking for an association<br />

between HLA-DRB1*04 and HLADQB1*06 with SECC for early<br />

diagnosis as well as prevention <strong>of</strong> the disease. In this study<br />

we extracted the genomic DNAs from the whole blood<br />

samples <strong>of</strong> 44 patients with SECC and 35 caries-free children<br />

(control group) by salting out method. We amplified the<br />

genomic DNA by PCR Sequence Specific Primer (PCR-SSP) and<br />

HLA-typing was performed for both alleles. The results<br />

revealed a significant increase in the frequency <strong>of</strong><br />

HLADRB1*04 in the patient group (P-Value =0.019). The<br />

odds ratio for this allele was detected to be 10. Frequency<br />

<strong>of</strong> HLA-DQB1*06 allele was not significantly different<br />

between two groups (P-Value=0.37).<br />

The above results suggest that HLA-DRB1*04 is associated<br />

with the susceptibility to SECC. Therefore, the detection <strong>of</strong><br />

HLA-DRB1*04 allele as a molecular marker for early diagnosis<br />

<strong>of</strong> SECC is recommended.<br />

doi:10.1016/j.clim.2007.03.163<br />

Su.125 The Genetic Relationship Among Eleven<br />

Persian Ethnic Groups: An Anthropological View<br />

Based on HLA Class II Gene Polymorphism<br />

Shirin Farjadian, Assistant Pr<strong>of</strong>essor, Shiraz University<br />

Medical Sciences, <strong>Immunology</strong> Department, Shiraz, Iran,<br />

Abbas Ghaderi, Full Pr<strong>of</strong>essor, <strong>Immunology</strong> Department,<br />

Shiraz, Iran<br />

Besides being considered by immunologists for its pivotal<br />

role in the immune response, highly polymorphic HLA genes<br />

are still regarded as useful markers by anthropologists for<br />

determination <strong>of</strong> genetic relationship and interaction among<br />

different populations. Knowledge <strong>of</strong> the HLA allele distributions<br />

in various populations is also critical for establishing<br />

bone marrow donor registries; study <strong>of</strong> HLA associated<br />

disease and designing <strong>of</strong> peptide vaccines against infections,<br />

tumors, or autoimmune diseases. In this study, HLA class II<br />

pr<strong>of</strong>iles were determined by molecular methods in 816 DNA<br />

samples from eleven ethnic groups <strong>of</strong> Persia and the genetic<br />

relationship <strong>of</strong> Persians to Asians and Europeans were<br />

investigated. DRB1*1103=04, DQA1*0501, and DQB1*0301<br />

were the most frequent alleles and DRB1*1103=04-<br />

DQA1*0501-DQB1*0301 was the most common haplotype in<br />

Persia. Six samples typed as DRB1*1605 by direct sequencing<br />

<strong>of</strong> exon 2 and all <strong>of</strong> them were heterozygote for DRB1 locus<br />

and belonged to DRB1*1605-DQA1*0101/2-DQB1*0502 haplotype.<br />

The results <strong>of</strong> neighbor-joining and correspondence<br />

analysis revealed a close genetic relationship among Persian<br />

subpopulations that were well separated from other Asians<br />

and European populations. The results <strong>of</strong> AMOVA also<br />

revealed that the main variation component (96.9%) was<br />

contributed to by the within-population level and genetic<br />

differentiation (FST) was 0.03 when all Persian subpopulations<br />

considered as one group.<br />

doi:10.1016/j.clim.2007.03.164<br />

Su.126 IFN-g Increases Neurogenesis in Aging and<br />

Alzheimer's-like Disease<br />

Shai Abutbul, PhD Student, The National Institute <strong>of</strong><br />

Biotechnology in the Negev, Department <strong>of</strong> Microbiology<br />

and <strong>Immunology</strong>, Beer-Sheva, Israel, Rona Baron, MSc, The<br />

National Institute <strong>of</strong> Biotechnology in the Negev,<br />

Department <strong>of</strong> Microbiology and <strong>Immunology</strong>, Beer Sheva,<br />

Israel, Anna Nemirovsky, MSc, The National Institute <strong>of</strong><br />

Biotechnology in the Negev, Department <strong>of</strong> Microbiology<br />

and <strong>Immunology</strong>, Beer sheva, Alon Monsonego, PhD, The<br />

National Institute <strong>of</strong> Biotechnology in the Negev,<br />

Department <strong>of</strong> Microbiology and <strong>Immunology</strong>, Beer Sheva,<br />

Israel, Yair Fisher, BSc, The National Institute <strong>of</strong><br />

Biotechnology in the Negev, Department <strong>of</strong> Microbiology<br />

and <strong>Immunology</strong>, Beer Sheva, Israel<br />

The birth <strong>of</strong> new neurons is maintained in the adult central<br />

nervous system (CNS) within restricted areas in the brain,<br />

primarily the dentate gyrus (DG) and the subventricular zone<br />

(SVZ). Neurogenesis significantly declines with aging, possibly<br />

to a greater extent with the progression <strong>of</strong> Alzheimer's disease<br />

(AD). The primary cause <strong>of</strong> this decline is unclear, but brainendogenous<br />

and peripheral immune mechanisms associated<br />

with aging and the progression <strong>of</strong> AD appear to affect stem cell<br />

proliferation and differentiation in the brain. In contrast to<br />

the suppression <strong>of</strong> neurogenesis by proinflammatory cytokines<br />

(such as IL-1b, TNF-a, and IL-6) associated with chronic glial<br />

activation and neurotoxicity in the aging or AD brain, our<br />

studies in IFNg-transgenic mice with AD-like pathology show<br />

that the T cell-derived cytokine IFN-g, expressed under the<br />

myelin basic protein promoter, induces an approximately<br />

tw<strong>of</strong>old increase in the quantity <strong>of</strong> neuronal progenitor cells in<br />

the DG <strong>of</strong> aged, but not young, mice. Limited expression <strong>of</strong><br />

IFN-g in the periphery also resulted in a significant decrease in<br />

the portion <strong>of</strong> CD4+CD25+ and CD4+Foxp3+ regulatory T cells<br />

among the CD4+ T cell population in the spleens <strong>of</strong> the<br />

transgenic mice (vs. control). IFN-g has <strong>of</strong>ten been implicated<br />

in brain inflammation and neuronal damage, although recent<br />

studies suggest that it plays a neuroprotective role. Our<br />

findings also demonstrate a novel role for IFN-g in neuronal<br />

recovery by acting directly on brain endogenous cells and/or<br />

indirectly by increasing peripheral immunity.<br />

doi:10.1016/j.clim.2007.03.165<br />

S183


S184 Abstracts<br />

Su.127 Astrocyte-Mediated Regulation <strong>of</strong> Human<br />

CNS Inflammation<br />

Alex Kostianovsky, MD, Center for Neurologic Diseases,<br />

Harvard Medical School, Boston, MA, David E. Anderson,<br />

PhD, Center for Neurologic Diseases, Harvard Medical<br />

School, Boston, MA<br />

Microglial activation, or a lack there<strong>of</strong>, is associated with<br />

numerous central nervous system (CNS) neurodegenerative<br />

and inflammatory diseases, including Alzheimer's disease,<br />

multiple sclerosis, and CNS tumors. We are interested in<br />

better understanding the extent to which astrocytes modulate<br />

microglial and T cell activation within the CNS. Using<br />

primary human astrocytes co-cultured with ex vivo human<br />

monocytes or primary human microglia, we report that<br />

astrocytes potently inhibit monocyte/microglial activation<br />

induced with pathogen-derived (TLR agonists) but not T cellderived<br />

(CD40L) inflammatory signals. More specifically,<br />

human astrocytes inhibit TNF-± secretion and induce IL-10<br />

secretion. Comparisons between primary human astrocytes<br />

and primary human malignant glioma cell lines (transformed<br />

astrocytes) have demonstrated that gliomas inhibit TNF-±<br />

and induce IL-10 to greater extents than do primary astrocytes;<br />

moreover, they can inhibit monocyte activation<br />

induced with both TLR and CD40L signals. Trans-well assays<br />

demonstrate that inhibition <strong>of</strong> TNF-± secretion is mediated<br />

by both soluble and contact-dependent mechanisms, whereas<br />

induction <strong>of</strong> IL-10 is contact-dependent. Candidate molecules<br />

such as TGF- 2 , IL-10, COX2, and CD200 do not appear to<br />

be responsible for the astrocyte-associated suppression <strong>of</strong><br />

TNF-±. Preliminary data suggest indicate that up-regulation<br />

<strong>of</strong> members <strong>of</strong> the STAT gene family in transformed<br />

astrocytes is associated with the immunosuppressive phenotype.<br />

Collectively, these results indicate that astrocytes<br />

serve an immunoregulatory role within the CNS, which may<br />

be amplified or suppressed in different CNS diseases. In the<br />

context <strong>of</strong> malignant gliomas, amplification <strong>of</strong> the normal<br />

immunoregulatory function <strong>of</strong> astrocytes may explain microglial<br />

defects and impaired T cell immunity associated with<br />

these tumors.<br />

doi:10.1016/j.clim.2007.03.166<br />

Su.128 Peripheral Markers <strong>of</strong> Encephalitis on<br />

Monocytes from SIV-infected Rhesus Monkeys<br />

Maria Cecilia Marcondes, Staff Scientist, The Scripps<br />

Research Institute, Molecular and Integrative Neurosciences<br />

Department, La Jolla, CA, Tricia Burdo, Research Associate,<br />

The Scripps Research Institute, Molecular and Integrative<br />

Neurosciences Department, La Jolla, CA, Caroline Lanigan,<br />

Scientific Associate, The Scripps Research Institute,<br />

Molecular and Integrative Neurosciences Department, La<br />

Jolla, CA, Howard Fox, Associate Pr<strong>of</strong>essor, The Scripps<br />

Research Institute, Molecular and Integrative Neurosciences<br />

Department, La Jolla, CA, Debbie Watry, Research<br />

Assistant, The Scripps Research Institute, Molecular and<br />

Integrative Neurosciences Department, La Jolla, CA<br />

In HIV-1 infected people, the accumulation <strong>of</strong> macrophages<br />

(MØs) in the brain correlates with dementia and<br />

encephalitis. We hypothesized that a pattern <strong>of</strong> monocyte<br />

surface marker expression may serve as a peripheral marker<br />

for CNS disease. Using the SIV/rhesus monkey model, we first<br />

analyzed functionally relevant surface markers on monocytes<br />

and macrophages from blood and organs upon<br />

termination in groups <strong>of</strong> animals that later did (SIVE) or did<br />

not (SIVnoE) develop encephalitis. Cluster analysis revealed<br />

that the pattern <strong>of</strong> staining <strong>of</strong> cells from the brain was<br />

closest to that found in blood, compared to spleen, liver,<br />

bone marrow and bronchioalveolar lavage. In the blood, the<br />

predominant (CD16low) population <strong>of</strong> monocytes from SIVE<br />

animals had higher levels <strong>of</strong> both CD44v6 and CCR5 compared<br />

to SIVnoE animals. Furthermore the inflammatory CD16+<br />

subpopulation <strong>of</strong> blood monocytes from SIVE animals<br />

expressed high levels <strong>of</strong> CD44v6, but not CCR5, compared<br />

to those from SIVnoE animals. In the brain, only CCR5<br />

expression on CD16+ macrophages correlated with encephalitis.<br />

A longitudinal analysis <strong>of</strong> blood monocyte markers<br />

revealed that, beginning at 3 weeks prior to termination,<br />

both CD16low and CD16+ monocytes indeed upregulated<br />

CD44v6. This provides a potential biomarker to determine<br />

individuals who may develop the central nervous system<br />

complications <strong>of</strong> HIV infection. Furthermore, given its role in<br />

cellular adhesion and as a receptor for osteopontin, CD44v6<br />

upregulation on monocytes <strong>of</strong>fers functional clues to the<br />

pathogenesis <strong>of</strong> such complications, and provides a target for<br />

preventative and therapeutic measures. (NIH Grant 1R01<br />

NS045534-04).<br />

doi:10.1016/j.clim.2007.03.169<br />

Su.129 Polymerized-Type I Collagen a Biodrug for<br />

the Treatment <strong>of</strong> Patients with Knee Osteoarthritis<br />

Janette Furuzawa-Carballeda, Chemistry, Instituto Nacional<br />

de Ciencias Medicas y Nutricion SZ, Department <strong>of</strong><br />

<strong>Immunology</strong> and Rheumatology, Mexico, Olga Muñoz-<br />

Chable, Physician, Instituto Nacional de Ciencias Medicas y<br />

Nutricion SZ, Department <strong>of</strong> <strong>Immunology</strong> and<br />

Rheumatology, Mexico, Israel Macias-Hernandez, Physician,<br />

Instituto Nacional de Ciencias Medicas y Nutricion SZ,<br />

Department <strong>of</strong> <strong>Immunology</strong> and Rheumatology, Mexico,<br />

Andres Agualimpia-Janning, Physician, Instituto Nacional de<br />

Ciencias Medicas y Nutricion SZ, Department <strong>of</strong> <strong>Immunology</strong><br />

and Rheumatology, Mexico<br />

Polymerized-Type I Collagen (Collagen-PVP) is an antiinflammatory<br />

and a tissue regenerator biodrug. In vivo<br />

study: Patients (n=53) were treated with 12 intraarticular<br />

injections <strong>of</strong> 2 ml <strong>of</strong> collagen-PVP (16.6 mg <strong>of</strong> collagen)<br />

(n=27) or 2 ml <strong>of</strong> placebo (n=26) during 6 months. The<br />

follow up was done during the next 6 months. The primary<br />

endpoints included WOMAC, Lequesne index and pain<br />

intensity on a VAS. Secondary outcomes were patient and<br />

investigator global score and biodrug evaluation. <strong>Clinical</strong><br />

improvement was determined if the decrease in pain<br />

exceeds 20 mm on VAS and patients achieved at least<br />

20% <strong>of</strong> improvement in primary endpoints from baseline to<br />

week 24. In vitro study: Cartilage and synovial tissue cocultures<br />

from 5 patients with OA were performed. Tissues<br />

were cultured during 7 days with/without 1% Collagen-PVP<br />

and they were stained with Alcian Blue technique. IL-1β,<br />

IL-8, IL-10, IL-12, TNF-α and IFN-γ were measured in


Abstracts<br />

supernatants by ELISA. COMP, type II collagen, TNF-α, IL-10<br />

and Ki-67 expression was determined by immunohistochemistry.<br />

Patients had a statistically significant improvement<br />

(pb0.05) in Collagen-PVP-treated versus baseline and versus<br />

placebo in Lequesne Index, WOMAC, VAS, patient global score<br />

and physician drug evaluation. The histological analysis<br />

showed that Collagen-PVP increased 3 to 6-fold proteoglycans,<br />

Ki-67, COMP and type II collagen. IL-1β and TNF-α<br />

production was inhibited; meanwhile IL-10 levels were upregulated<br />

treated versus untreated-cultures. Collagen-PVP<br />

was safe and more effective than placebo for the treatment<br />

<strong>of</strong> knee OA, probably due to the induction <strong>of</strong> tissue<br />

regeneration and the down-regulation <strong>of</strong> inflammation.<br />

doi:10.1016/j.clim.2007.03.170<br />

Su.132 Antibody-oligo Arrays for Multiplex Surface<br />

Marker Pr<strong>of</strong>iling <strong>of</strong> Immune Cells Subsets<br />

Michael Kattah, MSTP Student, Stanford University<br />

<strong>Immunology</strong> and Rheumatology, Stanford, CA, John Coller,<br />

Research and Development Engineer, School <strong>of</strong> Medicine,<br />

MDRP’S, Stanford Functional Genomics Facility, Stanford,<br />

CA, Golnaz Alemi, Medical Student, Drexel University,<br />

Stanford, CA, Neekaan Oshidary, Undergraduate Student,<br />

Stanford University <strong>Immunology</strong> and Rheumatology,<br />

Stanford, CA, Paul J. Utz, Associate Pr<strong>of</strong>essor <strong>of</strong> Medicine<br />

(<strong>Immunology</strong> and Rheumatology), Medicine, <strong>Immunology</strong><br />

and Rheumatology and Center for <strong>Clinical</strong> <strong>Immunology</strong> at<br />

Stanford, Stanford, CA<br />

While the field <strong>of</strong> proteomics holds great promise for the<br />

study <strong>of</strong> immune-related disorders, methods for analyzing<br />

specific proteins in a high-throughput manner are still needed.<br />

In an effort to develop a multi-analyte proteomics platform<br />

using monoclonal antibodies, we are developing a new assay<br />

termed antibody-oligo arrays (AOAs). This method employs a<br />

panel <strong>of</strong> monoclonal antibodies, each tagged with a unique<br />

oligonucleotide sequence identifier. After staining a sample,<br />

the complex mixture <strong>of</strong> tags is amplified and hybridized to a<br />

custom DNA microarray, providing an indirect measure <strong>of</strong> the<br />

relative levels <strong>of</strong> each analyte in the original sample. In an<br />

effort to identify cell-surface markers that are either up- or<br />

down-regulated in a given cell population, we have developed<br />

a cell-surface AOA to pr<strong>of</strong>ile 1–2×106 cells with a panel <strong>of</strong> 90<br />

surface markers and 4 isotype controls in two 48-plex<br />

reactions. Although there are many potential applications for<br />

this technology, we have performed pilot experiments using<br />

both cancer cell lines and primary human lymphocytes.<br />

Preliminary results have identified changes on the surface <strong>of</strong><br />

primary human naïve CD4+CD45RA+CD45RO-CD25- Tcells upon<br />

polyclonal activation in vitro with anti-CD3 and anti-CD28<br />

coated beads. The markers identified in this screen agree with<br />

previously described activation markers and were all validated<br />

by conventional flow cytometry. In the future we hope to<br />

pr<strong>of</strong>ile cell-surface markers on Tregulatory and IL-17 secreting<br />

Th17 cells with our newly developed cell-surface AOA in order<br />

to identify targets for future functional studies and for<br />

potential therapeutic interventions.<br />

doi:10.1016/j.clim.2007.03.171<br />

Su.133 Industrial Science: Biomarker Discovery<br />

from Plasma: Maximizing Proteomic Breath and<br />

Depth Using Protein Partitioning and Fractionation<br />

Followed by Shotgun Mass Spectrometry<br />

Shijun Sheng, Senior Research Technologist, Johns Hopkins<br />

Bayview NHLBI Proteomics Center, Baltimore, MD, Qin Fu,<br />

Research Associate, Johns Hopkins Bayview NHLBI<br />

Proteomics Center, Baltimore, MD, Jeff Chapman, Director,<br />

Beckman Coulter, Biomarker Discovery and Automation<br />

Business Center, Fullerton, CA, Jennifer Van Eyk, Associate<br />

Pr<strong>of</strong>essor Medicine, Johns Hopkins University-Bayview<br />

Campus, Baltimore, MD, Jerald Feitelson, Manager<br />

Strategic Marketing, Beckman Coulter, Biomarker Discovery<br />

and Automation Business Center, Fullerton, CA<br />

The potential <strong>of</strong> proteomics to identify new diagnostic<br />

markers was widely touted but has met with limited success<br />

due to technical issues related to reproducibility, sample<br />

requirements, availability <strong>of</strong> well characterized clinical<br />

cohorts for discovery and validation, and ability to obtain<br />

sufficient proteome depth and coverage. To discover novel<br />

biomarkers while overcoming previous limitations in proteomic<br />

methodology, we have developed a robust biomarker<br />

discovery pipeline that allows the detection and quantification<br />

<strong>of</strong> proteins below 100 pg/ml. The proteomic pipeline<br />

involves two key sample preparation steps at a large scale:<br />

(a) partitioning (removal) <strong>of</strong> the top 12 most abundant<br />

plasma proteins using immunoaffinity chromatography, and<br />

(b) subsequent two-dimensional liquid chromatography that<br />

fractionates intact proteins based on two <strong>of</strong> their key<br />

intrinsic properties, pI and hydrophobicity. Fractions are<br />

split allowing the third intrinsic protein parameter, intact<br />

mass, to be obtained using MALDI-TOF mass spectrometry.<br />

When combined with analysis <strong>of</strong> proteome maps based on<br />

aligned second dimension chromatograms, we can identify<br />

fractions that differ between individual samples in large data<br />

sets. To push the proteomics envelope, we carried out<br />

multiple analyses on each individual sample prior to pooling<br />

and quantitative shotgun LC-MS/MS to identify and characterize<br />

proteins. This biomarker discovery pipeline is being<br />

applied to unprecedented broad and deep analyses <strong>of</strong> 100<br />

individuals attempting to obtain quantitative protein characterization<br />

(is<strong>of</strong>orm and PTM) data. We have shown that this<br />

scale <strong>of</strong> discovery proteomics (1 mL plasma per patient, 100<br />

patients and controls) was able to identify candidate<br />

biomarkers <strong>of</strong> a crucially important disease.<br />

doi:10.1016/j.clim.2007.03.173<br />

S185<br />

Su.134 Human C-Reactive Protein Augments the Gut<br />

Ischemia/Reperfusion Injury in Mice<br />

Xinyue Lu, Research Fellow, Department Cellular Injury,<br />

Walter Reed Army Institute <strong>of</strong> Research, Silver Spring, MD,<br />

Russell Peckham, Principle Investigator, Corresponding<br />

Author, Department Cellular Injury, Walter Reed Army<br />

Institute <strong>of</strong> Research, Silver Spring, MD, Michael Falabella,<br />

Research Assistant, Department Cellular Injury, Walter Reed<br />

Army Institute <strong>of</strong> Research, Silver Spring, MD, George C.<br />

Tsokos, Principle Investigator, Pr<strong>of</strong>essor, Department<br />

Cellular Injury, Walter Reed Army Institute <strong>of</strong> Research,<br />

Silver Spring, MD


S186 Abstracts<br />

C-reactive protein (CRP) has been shown to increase<br />

ischemia/reperfusion (IR) injury in models <strong>of</strong> acute myocardial<br />

infarction and stroke. The role <strong>of</strong> CRP on intestinal IR which<br />

results in serious local and systemic inflammatory reactions is<br />

unknown. We adapted a murine superior mesenteric artery IR<br />

model which we used to study the effect <strong>of</strong> CRP. Human<br />

purified CRP (250, 500, and 1000 μg per mouse) was<br />

administered prior to the ischemia period <strong>of</strong> 30 min and tissue<br />

injury was evaluated after 2 hours <strong>of</strong> reperfusion. Intestinal<br />

injury damage, estimated using an established score, was<br />

statistically significantly higher in all CRP-treated groups<br />

compared to non-treated animals. The increased injury score<br />

was associated with increased intestinal myeloperoxidase<br />

activity only in the CRP-treated animals. In addition, the levels<br />

<strong>of</strong> the proinflammatory cytokines IL-6, IL-1α,TNF-α mRNA, as<br />

determined by real-time RT-PCR, were increased in the<br />

intestines <strong>of</strong> CRP-treated animals compared to the nontreated<br />

animals. Immun<strong>of</strong>luorescent staining revealed that<br />

human CRP was deposited in the intestinal tissues and that it<br />

co-localized with complement C3, factor H, MBL-A and C5b-9.<br />

Our results indicate CRP enhances intestinal IR injury probably<br />

by recruiting and activating the complement pathway.<br />

doi:10.1016/j.clim.2007.03.174<br />

Su.136 Expression, Histological Localization <strong>of</strong> hfgl2<br />

and Its Gene Regulation in Cancer<br />

Kai Su, Postgraduate Student, Department <strong>of</strong> Infectious<br />

Disease, Tongji Hospital, Huazhong University <strong>of</strong> Science<br />

and Technology, Wuhan, China, Fang Chen, Resident,<br />

Department <strong>of</strong> Infectious Disease, Tongji Hospital,<br />

Huazhong University <strong>of</strong> Science and Technology, Wuhan,<br />

China, Ping Zhang, Graduate Student, Department <strong>of</strong><br />

Infectious Disease, Tongji Hospital, Huazhong University <strong>of</strong><br />

Science and Technology, Wuhan, China, Xiaomin Qin, Doctor<br />

in Charge, Department <strong>of</strong> Infectious Disease, Tongji<br />

Hospital, Huazhong University <strong>of</strong> Science and Technology,<br />

Wuhan, China, Meifang Han, Doctor in Charge, Department<br />

<strong>of</strong> Infectious Disease, Tongji Hospital, Huazhong University<br />

<strong>of</strong> Science and Technology, Wuhan, China, Weiming Yan,<br />

Assistant Researcher, Department <strong>of</strong> Infectious Disease,<br />

Tongji Hospital, Huazhong University <strong>of</strong> Science and<br />

Technology, Wuhan, China, Bin Pi, Assistant Researcher,<br />

Department <strong>of</strong> Infectious Disease, Tongji Hospital,<br />

Huazhong University <strong>of</strong> Science and Technology, Wuhan,<br />

China, Dong Xi, Assistant Research, Department <strong>of</strong><br />

Infectious Disease, Tongji Hospital, Huazhong University <strong>of</strong><br />

Science and Technology, Wuhan, China, Xiaoping Luo, Chief<br />

Physician, Department <strong>of</strong> Pediatrics <strong>of</strong> Tongji Hospital,<br />

Huazhong University <strong>of</strong> Science and Technology, Wuhan,<br />

China, Qin Ning, Chief Physician, Department <strong>of</strong> Infectious<br />

Disease, Tongji Hospital, Huazhong University <strong>of</strong> Science<br />

and Technology, Wuhan, China<br />

To study the role <strong>of</strong> fibrinogen-like protein 2/fibroleukin<br />

(fgl2) in tumor development, human malignant tumor tissues<br />

from 133 patients with various kinds <strong>of</strong> cancers and the<br />

animal tumor tissues from human hepatocellular carcinoma<br />

(HCC) model on nude mice established by using different HCC<br />

cell lines were used to detect the fgl2 expression through an<br />

immunohistological method. Antibodies against different<br />

cellular markers (CD8, CD57, CD68 and vWF) were used to<br />

determine the cellular types in which the fgl2 was expressed.<br />

Fgl2 was detected in 127 out <strong>of</strong> 133 patients' samples and<br />

human HCC nude mice model. Fibrin (nogen) co-localization<br />

with fgl2 expression was determined by double staining.<br />

Particularly, it was evidenced that fgl2 was highly expressed<br />

not just in cancer cells, but also in interstitial infiltrated cells<br />

including macrophages, NK cells, CD8+T lymphocytes and<br />

vascular endothelial cells. In situ hybridization shows fgl2<br />

mRNA stained in associated location. Furthermore, IL-2, IFN-γ<br />

and TNF-a can dramatically up-regulate fgl2 mRNA (increased<br />

within the range <strong>of</strong> 10–100 fold) and protein expression in<br />

both THP-1 and HUVEC cell lines. One-stage clotting assay<br />

demonstrated that the significantly increased procoagulant<br />

activity after cytokines stimulation <strong>of</strong> both cell lines.<br />

The present study shows that fg12 contribute to the<br />

hypercoagulability in cancer, and thus in turn induces tumor<br />

angiogenesis and metastasis. This work was supported by<br />

Natural Science Foundation <strong>of</strong> China (NSFC 30225040,<br />

30571643, 30672380), National Key Basic Research Program<br />

<strong>of</strong> China (2005CB522901, 2005CB522507) and <strong>Clinical</strong> Subject<br />

Key Project from the Ministry <strong>of</strong> Health.<br />

doi:10.1016/j.clim.2007.03.175<br />

Su.137 CpG ODN Reduced Amyloid-β<br />

Accumulation Through Up-Regulated F-Spondin on<br />

Mouse BV2 Microglial Cell<br />

Yung Chih Cheng, PhD Student, Institute <strong>of</strong> Biochemical<br />

Science, National Taiwan University, Taipei, Taiwan, Cheng-<br />

Chin Kuo, Postdoctoral Fellowtor, Agricultural<br />

Biotechnology Research Center, Academia Sinica, Taipei,<br />

Taiwan, Shu-Mei Liang, Pr<strong>of</strong>essor, Agricultural<br />

Biotechnology Research Center, Academia Sinica, Taipei,<br />

Taiwan<br />

Alzheimer's disease, the most common senile dementia, is<br />

characterized by amyloid plaques, vascular amyloid, neur<strong>of</strong>ibrillary<br />

tangles, and progressive neurodegeneration. Amyloid<br />

plaques are mainly composed <strong>of</strong> amyloid-beta (A-β)<br />

peptides, which are derived from processing <strong>of</strong> the betaamyloid<br />

precursor protein (APP) by secretases. In this study,<br />

we found that BV2 microglial cells stimulated with unmethylated<br />

CpG-ODN not only increased the level <strong>of</strong> F-spondin but<br />

also resistant to A-β formation. Similar results were observed<br />

in CpG ODN stimulated RAW264.7 cells. Results from both real<br />

time PCR and western blotting demonstrated that CpG ODN<br />

regulated F-spondin expression at both transcriptional and<br />

translational level. Inhibition <strong>of</strong> TLR9 and MyD88 by inhibitors<br />

or dominant negative mutants indicated that CpG ODN upregulated<br />

F-spondin was TLR9/MyD88 dependent. In addition,<br />

we also found that a downstream kinase <strong>of</strong> TLR9, PI3K,<br />

was essential for CpG ODN-induced F-spondin expression.<br />

Depletion <strong>of</strong> F-spondin by small interfering RNA attenuated<br />

the prevention <strong>of</strong> CpG ODN on APP degradation by secretases,<br />

thereby increasing the level <strong>of</strong> A-β and soluble APP in culture<br />

medium. Collectively, these findings suggest that CpG ODN<br />

increases F-spondin via a TLR9/MyD88/PI3K pathway. The<br />

enhancement <strong>of</strong> F-spondin plays a critical role in the<br />

reduction <strong>of</strong> A-β formation mediated by CpG ODN, thereby<br />

reducing Alzheimer disease occurrence. These results may


Abstracts<br />

provide an insight for developing a potential drug for the<br />

treatment <strong>of</strong> Alzheimer's disease.<br />

doi:10.1016/j.clim.2007.03.176<br />

Su.138 Loss <strong>of</strong> Th1 and Effector T Cell Function in<br />

Chronic Versus Subacute Hypersensitivity<br />

Pneumonitis<br />

Lourdes Barrera, Biological Sciences Doctor, National<br />

Institute <strong>of</strong> Respiratory Diseases, Mexico City, Mexico,<br />

Felipe Mendoza, MsC, National Institute <strong>of</strong> Respiratory<br />

Diseases, Mexico City, Mexico, Moises Selman, National<br />

Insitute <strong>of</strong> Respiratory Diseases, Mexico City, Mexico<br />

Hypersensitivity pneumonitis (HP) represents an immunologically-mediated<br />

inflammation caused by a variety <strong>of</strong><br />

antigens. 21 patients with Sub-HP, 20 with Chr-HP and 8<br />

healthy subjects were studied. T cells from bronchoalveolar<br />

lavage (BAL) were analyzed by flow cytometry. Cytokine<br />

expression and memory functional pr<strong>of</strong>ile were evaluated in<br />

cell cultures. Chr-HP patients showed lower BAL lymphocytosis,<br />

higher CD4+/CD8+ ratio (3.5–3.3 versus 1.2–0.4 and<br />

1.7–1.5 from controls and Sub-HP; pb0.05), and a decrease<br />

<strong>of</strong> f× fÔT cells (2.5+0.9 versus 13.3+6.3 and 10.4+5.6 from<br />

controls and Sub-HP; pb0.01). Chr-HP had an increase in the<br />

terminally differentiated memory CD4+ and CD8+ T cells<br />

subsets compared with the subacute group (p b0.05).<br />

Memory cells from Chr-HP showed lower IFN-f× production,<br />

and decreased cytotoxicity <strong>of</strong> CD8+ T-lymphocytes. Chr-HP<br />

displayed a Th2-like phenotype with decreased CXCR3/CCR4<br />

ratio on T cells (2.5–1.8 versus 14.8–8.8; pb0.01) and lower<br />

levels <strong>of</strong> the CXCR3 ligands, IP-10/CXCL10 and MIG/CXCL9 in<br />

BAL supernatants. BAL lymphocytes from Chr-HP, stimulated<br />

with the specific antigen expressed lower levels <strong>of</strong> Th1<br />

cytokines and higher levels <strong>of</strong> Th2 cytokines (IFN-f× /IL-4<br />

ratio: 0.7–0.3% versus 2.1–0.6% from Sub-HP; pb0.01).<br />

Differences between chronic and subacute patients include<br />

an increase in CD4+/CD8+ ratio, a decrease <strong>of</strong> f× fÔT cells, a<br />

skewing towards Th2 as opposed to Th1 and exhaustion <strong>of</strong><br />

effector CD8+ T cells. These findings may provide insight in<br />

the development <strong>of</strong> new therapeutic strategies e.g. the<br />

inhibition <strong>of</strong> GATA-3 transcription factor to modify the Th1/<br />

Th2 cell balance and the blocking <strong>of</strong> PD-1 receptor to restore<br />

the Tcell function may be useful in the treatment for chronic HP.<br />

doi:10.1016/j.clim.2007.03.558<br />

Su.139 Analysis <strong>of</strong> Natural Killer Cells Isolated from<br />

Human Decidua: Evidence that 2B4 (CD244)<br />

Functions as an Inhibitory Receptor and Blocks NK<br />

Cell Function<br />

Gabriella Pietra, PhD, DIMES-University <strong>of</strong> Genoa,<br />

Genova, Italy, Paola Vacca, PhD Student, DIMES-University<br />

<strong>of</strong> Genoa, Genova, Italy, Lorenzo Moretta, MD, G Gaslini<br />

Institute, Genova, Italy, Federico Prefumo, MD, G<br />

Gaslini Institute, Genova, Italy, Maria Cristina Mingari,<br />

PhD, National Cancer Institute, Genova, Italy<br />

During the first trimester <strong>of</strong> pregnancy, CD56bright NK<br />

cells constitute more than 50% <strong>of</strong> the lymphocytes present<br />

in the decidua, a tissue in which B and T lymphocytes are<br />

rare. The role they have at this site is still unclear. It is<br />

currently believed that in the early stages <strong>of</strong> gestation,<br />

decidual NK (dNK) cells possess the ability to regulate<br />

trophoblastic differentiation and invasion. In this study we<br />

analyzed dNK cells for their phenotype and functional<br />

capability. We show that dNK cells express normal surface<br />

levels <strong>of</strong> certain activating receptors, including NKp46,<br />

NKG2D and 2B4, as well as <strong>of</strong> killer-cell immunoglobulin-like<br />

receptors (KIRs) and CD94/NKG2A receptor. In addition,<br />

high levels <strong>of</strong> cytoplasmic granules despite their CD56bright<br />

CD16- surface phenotype characterize them. Moreover, we<br />

provide evidence that in dNK cells, activating NK receptors<br />

display normal triggering capability while 2B4 functions as<br />

an inhibitory receptor. Thus, crosslinking <strong>of</strong> 2B4 resulted in<br />

inhibition <strong>of</strong> both cytolytic activity and interferon-gamma<br />

production. Clonal analysis revealed that, in the majority <strong>of</strong><br />

dNK cell clones, the 2B4 inhibitory function is related to the<br />

deficient expression <strong>of</strong> SLAM-associated protein (SAP)<br />

mRNA. Moreover biochemical analysis revealed low levels<br />

<strong>of</strong> SAP protein in dNK polyclonal population. Thus, in the<br />

absence <strong>of</strong> SAP, SHP-1 phosphatase can bind to 2B4 and<br />

mediate inhibition <strong>of</strong> the dNK-mediated cytolysis CD48+<br />

target cells (eg EBV+ B cells). This suggests that dNK cells,<br />

although potentially capable <strong>of</strong> killing, are inhibited in their<br />

function when interacting with cells expressing CD48 (the<br />

2B4 ligand).<br />

doi:10.1016/j.clim.2007.03.555<br />

S187<br />

Su.140 Population <strong>of</strong> T, T Reg, NK Cells and Their<br />

Activation and Inhibitory Receptors in Peripheral<br />

Blood <strong>of</strong> Healthy Women During the Menstrual<br />

Cycle<br />

Erika Aurora Martínez García, Master in Science<br />

(<strong>Immunology</strong>), University <strong>of</strong> Guadalajara, Guadalajara,<br />

Mexico, Victor Ermilo Arana Argaez, Master in Science<br />

(<strong>Immunology</strong>), University <strong>of</strong> Guadalajara, Guadalajara,<br />

Mexico, Beatriz Teresita Martín Márquez, Master in<br />

Science (<strong>Immunology</strong>), University <strong>of</strong> Guadalajara,<br />

Guadalajara, Mexico, Pedro Ernesto Sánchez Hernández,<br />

PhD, University <strong>of</strong> Guadalajara, Guadalajara, Mexico, Luz<br />

Maria Adriana Balderas Peña, PhD, Unidad de<br />

Investigación Medica en Epidemiología Clínica, UMAE, HE,<br />

CMNO Instituto Mexicano del Segu, Guadalajara, Mexico,<br />

Trinidad García Iglesias, PhD, University <strong>of</strong> Guadalajara,<br />

Guadalajara, Mexico, Alicia Del Toro Arreola, Master in<br />

Science, University <strong>of</strong> Guadalajara, Guadalajara, Mexico,<br />

Oscar Javier López León Murguía, MD, University <strong>of</strong><br />

Guadalajara, Guadalajara, Mexico, José Francisco Muñoz<br />

Valle, PhD, University <strong>of</strong> Guadalajara, Guadalajara,<br />

Mexico, Adrián Daneri Navarro, PhD, University <strong>of</strong><br />

Guadalajara, Guadalajara, Mexico, Mónica Vázquez Del<br />

Mercado Espin, PhD, University <strong>of</strong> Guadalajara,<br />

Guadalajara, Mexico<br />

There is evidence that the maternal immune system is<br />

influence by changes in the hormonal levels during the<br />

menstrual cycle. So far, the information related to the levels<br />

<strong>of</strong> T, T reg, NK cells and their activation and inhibitory<br />

receptors is scarce. Aim: To analyze the populations <strong>of</strong> T, T


S188 Abstracts<br />

reg, NK cells and their activation and inhibitory receptors <strong>of</strong><br />

peripheral blood <strong>of</strong> healthy women during the menstrual<br />

cycle. Material and Methods: We studied 10 women not using<br />

hormonal contraceptives in the day 5 <strong>of</strong> the follicular phase<br />

and 21st <strong>of</strong> the luteal phase <strong>of</strong> the menstrual cycle.<br />

Peripheral blood lymphocyte subsets and their receptors<br />

were determined by flow cytometry. Results: We do not<br />

observe changes in the percentages <strong>of</strong> CD4+ and CD8+ Tcells.<br />

With respect to ILT-2 and CD94/NKG2 receptors expressed in<br />

CD4+ T cells and at the levels <strong>of</strong> T reg cells (CD4+CD25+<br />

Foxp3+) we noted an increased in the follicular phase. On<br />

the other hand, the levels <strong>of</strong> NK cells remain constant in the<br />

two menstrual phases as well as their receptors NKG2D and<br />

ILT-2. According to the receptors NKp30 and NKp46 we<br />

identified major fluctuations in the follicular phase and the<br />

opposite for CD94/NKG2. Conclusions: We observed an<br />

increase in day 5 and decay in day 21 <strong>of</strong> the menstrual<br />

cycle in the majority <strong>of</strong> the cellular populations and their<br />

receptors. Perhaps these results are product <strong>of</strong> the necessary<br />

mechanisms in menstrual cycle to prepare the<br />

processes <strong>of</strong> implantation in early pregnancy.<br />

doi:10.1016/j.clim.2007.03.556


<strong>Clinical</strong> <strong>Immunology</strong> (2007) 123, S189–S206<br />

FOCIS 2007 Abstract Index by Author<br />

Abbas, Alexander R. Sa.149<br />

Abel, Lucia Cristina Jamli F.64<br />

Abello, Paula Sa.65<br />

Aberle, Teresa Sa.27<br />

Abhinandan, Raghavan Su.87<br />

Abolhassani, Mohsen Sa.17<br />

Abouljoud, Marwan F.71<br />

Abrams, Elaine F.94<br />

Abramson, Tzvia F.69<br />

Abril, Annette Sa.26<br />

Abutbul, Shai Su.126<br />

Adams, Jason OR.59<br />

Adams, Katherine Sa.98<br />

Adib, Minoo F.125<br />

Adler, Adam OR.81<br />

Adriani, Marsilio OR.29<br />

Agarwal, Rajeev K. OR.38<br />

Aghasizadeh, Navid Su.62<br />

Agualimpia-Janning, Andres Su.129<br />

Aguiar, Paloma Sa.134<br />

Aguilar-Lemarrroy, Adriana C. Su.55<br />

Aguilar-Velazquez, Gustavo Su.76<br />

Aguilera, Raquel Sa.133<br />

Aguillon, Juan C. Sa.65<br />

Ahearn, Joseph M. Sa.31, Sa.35<br />

Aherns, Richard F.124<br />

Ahlmen, Jarl F.129<br />

Ailes, Elizabeth F.44<br />

Aksentijevich, Ivona Sa.49, Sa.50<br />

Alam, Munir F.72<br />

Alami Chentoufi, Aziz F.53<br />

Albani, Salvatore F.20, Sa.66, Sa.67, Sa.72,<br />

Sa.73<br />

Alberu, Josefina Su.101<br />

Alcocer Varela, Jorge F.93, Sa.81<br />

Alemi, Golnaz<br />

Alesahebfosoul, Fereshteh F.111, F.125<br />

Alkan, Sefik Su.57<br />

Allaire, Normand OR.55<br />

Allauzen, Sophie F.136<br />

Allen, Hamish Sa.40<br />

Allende, Gloria F.18<br />

Allie, Rameeza Sa.145<br />

Alliot-Licht, Brigitte Su.96<br />

doi:10.1016/j.clim.2007.03.541<br />

available at www.sciencedirect.com<br />

www.elsevier.com/locate/yclim<br />

Allison, James E. Sa.152<br />

Almeida, Alexandre F.67, F.98<br />

Alonso, Liv an Sa.147<br />

Alper, Chester Su.59, Su.60<br />

Althshuler, David OR.80<br />

Altshuler, David Su.35<br />

Alves, Venancio F.64<br />

Aly, Theresa A. OR.78<br />

Amato, Anthony Su.31<br />

Amox, Diane Sa.66<br />

Anderson, Amy F.57<br />

Anderson, David E. OR.95, OR.101, Sa.114,<br />

Su.127, Su.38<br />

Anderson, David F.15, OR.83, Sa.131<br />

Anderson, Mark F.12, OR.60, Su.111<br />

Anderson, Richard C.E. OR.58<br />

Anderson, Richard Sa.120<br />

Anderson, Stacie M. OR.29<br />

Anegon, Ignacio OR.71, Su.119<br />

Annels, Nicola F.103<br />

Annese, Vitto Su.93<br />

Ansaldo, Gianluca Sa.112<br />

Antel, Jack Su.10, Su.16<br />

Anthony, Donald F.71<br />

Aoki, Joseph OR.29<br />

Aono, Shelly F.61<br />

Aparicio, Carlos F.129<br />

Arab, Hamid Reza Su.50<br />

Arana Argaez, Victor Ermilo Su.140, Sa.44<br />

Aranami, Toshimasa OR.24, Su.12<br />

Aravena, Octavio Sa.65<br />

Arbour, Nathalie OR.33, OR.41, Su.16,<br />

Su.29<br />

Arechiga, Adrian F.28, Sa.148<br />

Arend, William P. Sa.71<br />

Aricò, Maurizio F.81<br />

Arif, Sefina OR.18<br />

Armengol, Maria Pilar F.26<br />

Armstrong, Don OR.79<br />

Arnold, Arthur P. Su.134<br />

Arnold, Dilani F.80<br />

Arouche Camara Lopes,<br />

Luiz Heraldo<br />

Sa.123<br />

Arshi, Saba Sa.7


S190 FOCIS 2007 Abstract Index by Author<br />

Ashayeri, Hassan Sa.7<br />

Ashfield, Rebecca OR.9<br />

Ashley, Charles OR.77, Sa.120<br />

Ashman, Robert F. OR.44<br />

Ashton-Chess, Joanna Su.113<br />

Ashutosh, Mangalam OR.61<br />

Aspsater, Mahsa F.129<br />

Atkinson, Mark OR.15, OR.2, OR.56<br />

Au, Liemin OR.49<br />

Au, Melinda Su.109<br />

Aud, Dee Su.40<br />

Autschbach, Frank Su.90, Su.91<br />

Avanesyan, Armine OR.97<br />

Awasthi, Amit Su.32<br />

Azadi, Gholomreza Sa.7<br />

Azmi, Hooman F.134<br />

Azuma, Miyuki F.33<br />

Azzari, Chiara F.91<br />

B. Elkon, Keith Sa.22<br />

Babu, Sunanda R. OR.78, F.12<br />

Bacchetta, Rosa F.91<br />

Badolato, Raffaele F.12, F.91<br />

Badr El-Din, Nariman Sa.124<br />

Badur, Selim Sa.109<br />

Baecher-Allan, Clare OR.77, Sa.120<br />

Baeten, Dominique Sa.57, Sa.58, Sa.59<br />

Baez, Ineavely F.113<br />

Bagely, Jessamyn Sa.11<br />

Baghani, Zahra<br />

Bagherian, Ali:<br />

Su.50<br />

Bailey, Samantha OR.34<br />

Baker, Harold F.104<br />

Baker, Mark Su.101<br />

Baker, Rocky F.25<br />

Balboni, Imelda Sa.46<br />

Balderas Peña, Luz Maria Su.140<br />

Adriana<br />

Balmer, Paul F.126, F.65<br />

Banda, Nirmal Sa.63<br />

Bankey, Paul F.70<br />

Baranyi, Ulrike Sa.11<br />

Baranzini, Sergio Su.15<br />

Barba-Barajas, Martha Su.55<br />

Barbour, Gene OR.3<br />

Barcellos, Lisa Sa.152<br />

Baric, Ralph F.46<br />

Barker, Jennifer F.12<br />

Barker, Jonathan OR.39<br />

Barmada, Michael Su.92<br />

Baron, Rona Su.126<br />

Bar-Or, Amit OR.41, OR.86<br />

Barrera, Lourdes Su.138<br />

Barros, Noac Chuffi F.96<br />

Barsky, Lora F.113<br />

Bartholomew, Richard OR.13<br />

Bartunkova, Jirina F.116, Sa.117<br />

Baschal, Erin E. OR.78<br />

Bason, Caterina F.65, OR.23, 1202<br />

Basso, Alexandre S. Su.28<br />

Bastos, Fabiana Su.19<br />

Batliwalla, Franak OR.55, Sa.55, Sa.61<br />

Bautista, Alejandro Su.98<br />

Bautista-Vazquez, Alejandro Su.95<br />

Bautz, David Sa.136<br />

Bayer, Monika Sa.126<br />

Begovich, Ann Sa.150<br />

Behrens, Marshall Sa.154<br />

Behrens, Timothy W. OR.80<br />

Bein, Gregor Su.41<br />

Belaunzaran, Francisco F.97<br />

Belmares, Michael Sa.132<br />

Belouski, Shelley F.137<br />

Benard, Gil F.52<br />

Bender, James Sa.106<br />

Benghiat, Fleur Samantha Su.103, Su.130<br />

BenMohamed, Lbachir F.53<br />

Benson, Charles OR.28, Sa.125<br />

Benson, Jacqueline OR.40<br />

Benvenuti, Luiz A. Su.124<br />

Benvenuto, Federica Sa.130<br />

Berer, Kerstin OR.20<br />

Berg, Ulla F.122<br />

Bergan, Lindsay Sa.97<br />

Berghöfer, Beate Su.41<br />

Bergholdt, Regine F.22<br />

Bergmann, Christoph OR.73<br />

Beriou, Gaelle Su.96<br />

Bernstein, Allan L. Su.2<br />

Bethunaickan, Ramalingam Sa.45<br />

Bettahi, Ilham F.53<br />

Bettelli, Estelle Su.42<br />

Bezerra, Jorge Su.123<br />

Bhairo, Michelle Sa.113<br />

Bhan, M.K. F.68<br />

Bhat, Roopa OR.96<br />

Bhatnagar, Shinjini F.68<br />

Bianchi, Lucia F.91<br />

Bianco, Nicolás Sa.85<br />

Bianco, Nicole OR.89<br />

Bidar, Maryam Su.62<br />

Biedermann, Tilo Sa.153<br />

Bienkowska, Jadwiga OR.55, Sa.55<br />

Bilton, Diana F.67<br />

Binder, Steven Sa.79<br />

Birmelin, Jennifer F.89<br />

Bishop, Gail A. Sa.68<br />

Blanca, Isaac Sa.85, Su.108<br />

Blanchard, Carine F.124<br />

Blankenhorn, Elizabeth OR.82<br />

Bloomfield, Tiffany Sa.96<br />

Bluestone, Jeffrey A. OR.16, OR.59, F.33, F.34<br />

Boardman, Cynthia Su.48<br />

Bohmova, Kristyna F.19<br />

Bollyky, Paul Sa.128<br />

Bolton, Wade F.129<br />

Bonadkar, Sasha Su.45<br />

Borgonovo, Giacomo OR.62<br />

Borrow, Ray F.126, F.65<br />

Bossi, Giovanna OR.9, Sa.98<br />

Bothwell, Alfred Su.102<br />

Bottini, Nunzio Sa.153<br />

Boudakov, Ivo F.38, Sa.127<br />

Bourcier, Kasia Sa.151, Su.37


FOCIS 2007 Abstract Index by Author<br />

Bourdette, Dennis OR.13<br />

Bour-Jordan, Helene F.33<br />

Bowman, Edward Su.91<br />

Boyer, Scott Su.63<br />

Boyle, John F.76<br />

Bradley, Brenda OR.3<br />

Bradshaw, Elizabeth Su.31<br />

Braemswig, Kira OR.15<br />

Branch, Rodshawn F.120<br />

Branigan, Patrick OR.40<br />

Braud, Cristophe Sa.57<br />

Braun, Jonathan OR.100, Su.82, Su.89<br />

Braun, Michel Sa.89<br />

Braun, Werner OR.22<br />

Braunstein, Jutta Su.90<br />

Bravo-Cuéllar, Alejandro Su.55<br />

Bresson, Damien OR.14, F.7<br />

Brewer, Joanna Sa.107<br />

Brion, Régis Su.119<br />

Brito, Cyro Alves Sa.16<br />

Brizuela, J.A. Su.32<br />

Brod, Staley Su.20<br />

Brodsky, Nina N. OR.52<br />

Brook, Azam Sa.105, Su.62<br />

Brooks-Worrell, Barbara OR.49<br />

Brorsson, Caroline F.22<br />

Brouard, Sophie Sa.57<br />

Brown, Chris F.132<br />

Brown, Margaret R. OR.52<br />

Bruce, Jeffrey N. Sa.114<br />

Bruce, Jeffrey Sa.120<br />

Bruner, Ben OR.89<br />

Bruner, Gail R. Sa.24<br />

Brunner, Tali Su.10<br />

Brusko, Todd OR.2<br />

Bryant, Shaughn Sa.32<br />

Buckle, Guy Su.27<br />

Buckner, Jane OR.19, OR.7, OR.76, F.28,<br />

Sa.128<br />

Bucknum, Amanda Sa.96<br />

Budinsky, Vit F.109<br />

Bupp, Melanie F.33<br />

Burdo, Tricia Su.128<br />

Burge, Matthew Sa.88<br />

Burke, George W. F.18<br />

Burrows, Gregory OR.10<br />

Butcher, Eugene C. OR.37<br />

Butte, Atul OR.53, F.9<br />

Buus, Søren F.53<br />

Cai, Chun Sa.134<br />

Cai, Qing Sa.150<br />

Calabresi, Peter Sa.145<br />

Calder, Claudia OR.86<br />

Callewaert, Nico Sa.38<br />

Camara-Lopes, Luiz F.131, Sa.121<br />

Cameron, Brian OR.9<br />

Campagnolo, Denise Su.9<br />

Canavez, Flavio Sa.121<br />

Candotti, Fabio OR.29<br />

Cannon, Jennifer Sa.116<br />

Cantaert, Tineke Sa.57, Sa.58, Sa.59<br />

Cantu-Brito, Carlos F.93<br />

Capocasale, Renold OR.40<br />

Capon, Francesca OR.39<br />

Carmans, S<strong>of</strong>ie OR.94<br />

Carmignani, Giorgio Sa.112<br />

Carmody, Francis F.13<br />

Carneiro-Sampaio, Magda Maria F.82<br />

Carter, Robert Sa.75<br />

Carulli, John OR.55, Sa.55<br />

Casanova, Jean-Laurent F.96<br />

Caspi, Rachel R. OR.38, OR.35<br />

Castro-Ramos, Feliciano Su.95<br />

Catanzarite, Tatiana Sa.132<br />

Cayrol, Romain OR.33, OR.88<br />

Chaim, Jacob OR.79<br />

Chan, Chi-Chao OR.35, OR.38<br />

Chang, Monica Sa.150<br />

Chang, Shun-Chiao Sa.53<br />

Chang, Vicky Su.82<br />

Chao, Nelson Sa.86<br />

Chapel, Helen F.80, F.83<br />

Chapman, Jeff Su.133<br />

Chau, Sandra F.27<br />

Chauveau, Christine Su.119<br />

Chavali, Arvind Su.84<br />

Chavez, Raul Su.93<br />

Chavez, Salvador Su.95<br />

Cheeran, Daniel OR.82<br />

Chella, David OR.61<br />

Chen, Benny Sa.86<br />

Chen, Cheng-Hsu OR.31<br />

Chen, Dong OR.42, OR.47<br />

Chen, Fang Su.136<br />

Chen, Genhui Sa.143<br />

Chen, James Sa.135<br />

Chen, Jing F.94<br />

Chen, Li-Chen F.88<br />

Chen, Ling Su.82<br />

Chen, Qian F.134<br />

Chen, Yi-Je Su.94<br />

Chen, Zoe OR.35<br />

Cheney, Richard Sa.111<br />

Cheng, Mickie OR.60, F.12<br />

Cheng, Yung Chih Su.137<br />

Chiffoleau, Elise OR.71, Su.96<br />

Chin, Hyunsook Sa.152<br />

Chini, Loredana F.93<br />

Chinn, Ivan F.84<br />

Chirinos, Perla Sa.90<br />

Cho, Judy Su.92<br />

Choi, Bong-Kum Sa.90, Sa.91<br />

Choi, Jae Eun Su.49<br />

Choi, James Su.62<br />

Chong, Mark Su.3<br />

Chooklin, Serge Su.110<br />

Choudhury, Arpita Sa.41<br />

Chow, Anthony W. Sa.93<br />

Choy, Edwin Su.35<br />

Christen, Selina Sa.126<br />

Christen, Urs 2201, OR.62, Sa.126<br />

Chruscinski, Andrzej J. Sa.30<br />

Chu, Alvina D. Sa.30<br />

S191


S192 FOCIS 2007 Abstract Index by Author<br />

Chung, Denise OR.19<br />

Chung, James Su.69<br />

Ciancio, Gaetano F.18<br />

Citron, John Sa.152<br />

Ciullini Mannurita, Sara F.91<br />

Clarke, Grace Su.94<br />

Clayton, David OR.85<br />

Coccia, Marco Sa.137<br />

Cody, Virginia OR.43<br />

Cohen, Irun F.20<br />

Cohen, Philip L. OR.58<br />

Cohen, Philip Sa.41, Sa.50<br />

Cohen, Smadar Su.10<br />

Cohen, Stanley Sa.62<br />

Coito, Ana OR.97<br />

Cole, Derek Sa.142<br />

Collanieri, Anna Cristina F.67<br />

Coller, John Su.132<br />

Collins, Mary Sa.142, Su.9<br />

Concannon, Patric Sa.148<br />

Condamine, Thomas OR.71<br />

Conesa, Angela Su.108<br />

Constabtino-Silva, Rosemeire F.99, F.96, Su.75<br />

Navickas<br />

Contreras, Carmen F.51<br />

Contreras-Levicoy, Juan Sa.65<br />

Cooke, Lawrence Su.46<br />

Cooper, Leslie Sa.154<br />

Coppieters, Ken Sa.38, Sa.95, Sa.23<br />

Coram, Marc OR.53<br />

Cornelius, Jennine Su.84<br />

Corneta, Elaine Cristina Sa.123<br />

Corneta, Elaine F.131<br />

Correa, Alexandre OR.100, F.100<br />

Correa, MRF Su.75<br />

Cort, Laura OR.82<br />

Costa, Cristiana F.103<br />

Costantino, Cristina F.115<br />

Costenbader, Karen H. Sa.53<br />

Courtenay-Luck, Nigel Sa.107<br />

Cowan, Morton OR.25<br />

Creusot, Remi J. F.5<br />

Crispín, José Carlos Sa.81<br />

Crispin, Jose F.93, Su.101, Sa.20<br />

Croen, Lisa Sa.152<br />

Cr<strong>of</strong>t, Michael F.10<br />

Cronk, Katharine Sa.114<br />

Crow, Mary Sa.47, Sa.92, Su.116<br />

Cruzado-Park, Ingrid D. F.72<br />

Cruz-Robles, David Sa.64<br />

Cua, Daniel OR.35<br />

Cuchacovich, Miguel Sa.65<br />

Cunningham, Matthew Su.96<br />

Cunninghame Graham, Deborah OR.80<br />

Cutter, Gary Sa.66<br />

Cuturi, Maria-Cristina OR.71, Su.96<br />

Czelusta, Adam Sa.12<br />

da Silva Duarte, Alberto Jose F.67, F.97, F.98<br />

Dai, Yang F.27, F.30<br />

Daikh, David Sa.82<br />

Dailey, Michael E. Sa.138, Su.5<br />

Daisuke, Goto Sa.40<br />

Daisuke, Tokita Su.105<br />

Daly, Mark Su.35, Su.93<br />

Damle, Aarti OR.55, Sa.63<br />

Dan, Nicolae Su.92<br />

Dandekar, Abhya Sa.17<br />

Daneri Navarro, Adrián Su.140<br />

Dang, Demi F.5, F.9, OR.16<br />

Danilenko, Dimitry OR.37<br />

Dannecker, Guenther E. F.135<br />

Dardalhon, Valerie OR.34<br />

Darden, Janice F.133<br />

Das, Shampa Su.59, Su.60<br />

Dasgupta, Gargi F.53<br />

Datta, Lisa Su.92<br />

Dave, Amy F.10<br />

Davey, Michael OR.84, Su.79<br />

David, Chella Sa.154, Sa.78<br />

Davidson, Anne Sa.45<br />

Davies, Robert F.80<br />

Davis, Ian D. Su.54<br />

D’Costa, Sybil F.136, F.72<br />

De, Asit F.72<br />

De, Mita F.70<br />

de Almeida, Alexandre F.97<br />

de Bakker, Paul Su.93<br />

de Beaucoudrey, Ludovic F.92<br />

De Jager, Philip OR.83, Su.27, Su.35,<br />

Su.36, Sa.139<br />

de Jager, Wilco F.20, Sa.75<br />

De Keyser, Filip Sa.59<br />

de Krijger, Ronald F.103<br />

de Leo, Claudia Su.101<br />

De Rycke, Leen Sa.58, Sa.59<br />

De Sanctis, Juan Sa.1, OR.81<br />

De Vos, Martine Sa.23<br />

de Vries, Niek Sa.57<br />

Degauque, Nicolas F.3<br />

Deisenhammer, Florian Su.8<br />

Dekan, Gerhard OR.64<br />

Del Toro Arreola, Alicia Su.140<br />

dela Rosa, Corazon Sa.101, Su.118<br />

Dellanegra, Marinella F.96<br />

Delovitch, Terry L. OR.30, F.4, F.23<br />

Demartino, Julie Su.58<br />

Denham, Jerrod Su.109<br />

Dennis, Rebecca OR.9<br />

Derfanoux, Nadine A. Sa.52<br />

Derfuss, Tobias 3202<br />

Desaire, Heather F.72<br />

Deschamps, Jack-Yves Su.96<br />

Deshmukh, Umesh Sa.78<br />

Deshpande, Chetan Sa.62<br />

Devlin, Blythe F.84<br />

DeVoss, Jason Su.111<br />

Di Meglio, Paola OR.39<br />

Di Mitri, Diletta Su.43<br />

Diamantopoulos, Stavros F.18<br />

Diange, L. Su.114<br />

Diarra, Danielle Sa.51<br />

DiCarlo, Edward Su.116<br />

Diehl, Lauri Su.84


FOCIS 2007 Abstract Index by Author<br />

Dinarello, Charles F.130<br />

Ding, Li F.66<br />

Dinnall, Joudy-Ann Sa.42<br />

Dishaw, Larry J. F.86<br />

Disis, Mary Sa.101, Su.118<br />

Dissanayake, Samudra Sa.115<br />

Dittmer, Dirk Su.39<br />

Dodelet-Devillers, Aurore OR.33, OR.88<br />

Doi, Yoshimitsu Su.13<br />

Dolcino, Marzia OR.23<br />

Dombrovskiy, Viktor Su.74<br />

Domingues Ferreira, Mauricio F.67, F.97, F.98<br />

Dominguez-Rodriguez, Su.55<br />

Jorge R.<br />

Dong, Jiaxin Su.47<br />

Dorsey, Morna J. F.86<br />

Dotta, Francesco F.24<br />

Douhan III, John Sa.142<br />

Downes, Kate OR.85<br />

D'Souza, Patricia F.133<br />

Du, Jianguang OR.72<br />

Du, Sienmi Su.112<br />

Du, Xiaoyan Sa.71<br />

Duan, Su Su.107<br />

Duarte, Alberto J.S. Sa.49, F.52, F.86, Sa.50,<br />

F.96, F.99, Su.75, Sa.16,<br />

F.92<br />

Dubey, Devendra Su.59, Su.60<br />

Dubrava, Tatyana Sa.92<br />

Duchateau, Jean<br />

Dugast, Anne-Sophie<br />

F.127<br />

Dumont, Debora OR.87<br />

Dunger, David OR.85<br />

Dunn, Shannon OR.69<br />

Dunne, Jack Su.118<br />

Dunne, John Sa.101<br />

Dunussi-Joannopoulos, Sa.142<br />

Kyriaki<br />

Durinovic-Bello, Ivana F.32<br />

Dusilova Sulkova, Sylva F.109<br />

Dutt, Suparna OR.68<br />

Dutz, Jan P. F.21<br />

Dzuris, John Su.107<br />

Eagar, Todd F.33<br />

Easley, Kirk F.94<br />

Eastham-Anderson, Jeffrey Su.55<br />

Ebeling, Cory OR.65<br />

Edberg, Jeffrey Sa.84<br />

Edgerton, Colin Sa.79<br />

Edmond, Jean Su.92<br />

Edwards, Hannah OR.6<br />

Egeler, Maarten F.103<br />

Eibel, Hermann OR.32<br />

Eisenbarth, George S. OR.78, F.12, F.17, F.7,<br />

OR.1, OR.21<br />

Eisenberg, Robert A. OR.58, Sa.41, Sa.42<br />

El Baz, Mimouna F.127<br />

El Khoury, Joseph OR.90<br />

Elewaut, Dirk Sa.23, Sa.38<br />

Elliott, John OR.21<br />

Ellis, Richard OR.18<br />

Elloumi, Houda F.66<br />

Elmets, Craig Su.89<br />

Elyaman, Wassim Su.22<br />

Emanuel, Katy Sa.103<br />

Engelmann, Peter F.31<br />

Epstein, Michelle OR.64<br />

Eriksson, Anna Sa.135<br />

Esmaili, Habibollah Su.124<br />

Estevam, Jose Sa.151, Su.37<br />

Estrada-Garcia, Iris Su.76<br />

Estrada-Parra, Sergio Su.76<br />

Evanko, Stephen Sa.128<br />

Exley, Andrew F.126, F.65<br />

Ezekowitz, Alan Sa.63<br />

Fafutis, Mary F.53<br />

Fahmidekar, Mohammad Ali Su.78<br />

Falabella, Michael Su.134<br />

Falk, Ronald Sa.136<br />

Fanslow, William 3201<br />

Farber, Donna OR.46<br />

Farid-Hosseini, Reza Sa.9, Su.66<br />

Fariss, Robert Su.81<br />

Farjadian, Shirin Su.125<br />

Farkas, Klara F.31<br />

Farshad, Shohreh F.46<br />

Fathman, C. Garrison F.5, F.9, OR.16<br />

Faure, Olivier Sa.106<br />

Fedynyshyn, Joe F.136<br />

Feingersh, Diane Sa.137<br />

Feitelson, Jerald Su.133<br />

Felton, Jamie OR.49<br />

Fenoglio, Daniela Sa.112<br />

Feo, Lourdes Sa.32<br />

Ferbas, John F.137, Su.86<br />

Fergus, Gleeson F.80<br />

Fernandez, Marco Antonio F.26<br />

Fernando, Michelle Sa.74<br />

Ferrada, Carlos Sa.122<br />

Ferrao, Maria do Socorro F.96<br />

Ferraz Neto, Ben-Hur F.64<br />

Ferrera, Francesca OR.12<br />

Ferrone, Soldano Sa.111<br />

Ferry, Berne F.83<br />

Field, Elizabeth H. Sa.138<br />

Field, Leigh F.22<br />

Fife, Brian T. F.33<br />

Filaci, Gilberto Sa.112<br />

Filippi, Christophe 2201, F.7<br />

Finegood, Diane F.21<br />

Fiorillo, Edoardo Sa.153<br />

Fisher, Yair Su.126<br />

Flanagan, Ken Su.84<br />

Flavell, Richard Su.79<br />

Fleisher, Thomas 3201<br />

Flores, Antonio Su.98<br />

Fohner, Alison Sa.10<br />

Forbes, Elizabeth F.124<br />

Foresti, Roberta Su.119<br />

Forman, Daron F.114<br />

Forman, Stephen Sa.87<br />

Forsyth, Ramses F.105<br />

S193


S194 FOCIS 2007 Abstract Index by Author<br />

Fossati, Gianluca Su.102, Su.85<br />

Foulkes, Roly Su.86<br />

Fousteri, Georgia F.10<br />

Fox, Edward Su.21<br />

Fransson, Moa Sa.100<br />

Fraser, Patricia Sa.53<br />

Fravega, Marco Sa.112<br />

Frederiksen,<br />

Su.54<br />

Klaus Stensgaard<br />

Frenkel, Dan Su.28<br />

Friend, Samantha Sa.66<br />

Frieri, Marianne Sa.8<br />

Frulloni, Luca 1202<br />

Fu, Qin Su.133<br />

Fujimoto, Manabu F.108, Sa.25, Su.63,<br />

Su.67, Su.68, Su.68<br />

Fujio, Keishi Sa.22<br />

Fujita, Mayumi Su.14<br />

Fujita, Tomoyuki Sa.25<br />

Fujiwara, Daisuke OR.100, Su.89<br />

Fulcher, Jennifer Sa.135<br />

Fung, Erik F.11<br />

Fung, John J. OR.31, Su.121<br />

Funke, Benjamin Su.90<br />

Furuzawa-Carballeda, Janette Sa.64, Su.129<br />

Fusaro, Ana Elisa Sa.16<br />

Gadkar, Kapil OR.59<br />

Gailey, Ryan Sa.104<br />

Gaines, Elizabeth Su.65<br />

Gallucci, Stefania Sa.42<br />

Gambhir, Sam S. F.5<br />

Gambineri, Eleonora F.91<br />

Ganjali, Rashin Sa.48, Su.66<br />

Gao, Sui F.50<br />

García Iglesias, Trinidad Su.140<br />

Garcia, Bertha OR.47, 1201<br />

Garcia, Michael Sa.71<br />

Garmendia, Jenny Sa.1<br />

Garvik, Barbara Sa.97<br />

Gattorno, Marco Sa.130<br />

Geba, Gregory Sa.5<br />

Gelli, Anna Maria Grazia<br />

Geng, Xuan F.21<br />

George, Tracy OR.68<br />

George, Heavner OR.40<br />

Gailey, Ryan Sa.104<br />

Ghaderi, Abbas Su.125<br />

Ghaffari, Javad Sa.109<br />

Ghahramani, Negar Sa.72, Sa.73<br />

Ghanekar, Smita Sa.133<br />

Ghasemi Hashtroodi, Leila F.1<br />

Ghio, Massimo Sa.33<br />

Ghoneum, Alia Sa.124<br />

Ghoneum, Mamdooh Sa.124<br />

Ghorayeb, Christine OR.86<br />

Giese, Thomas OR.46, Su.100, Su.107,<br />

Su.15, Su.90<br />

Gilbert, Leona OR.63<br />

Ginesta, Alexandra Sa.122<br />

Giuliani, Fabrizio Su.14<br />

Gluhcheva, Yordanka Georgieva Sa.108<br />

Gneiss, Claudia Su.8<br />

Golbin, Jason Sa.6<br />

Gold, Ralf Su.5<br />

Goldblum, Randall OR.22<br />

Goldoni, Adriana Leticia OR.48<br />

Goldrath, Ananda Su.61<br />

Goltsev, Anatoliy Sa.92<br />

Gomez-Contreras, Piedad C. Su.55<br />

Gonzalez, Carlos Sa.73<br />

Goodwin, Kelley Su.79<br />

Gorczynski, Reg Sa.127<br />

Gorczynski, Reginald F.38<br />

Gordon, John OR.65<br />

Gorelik, Elieser Sa.123<br />

Gosch, Courtney Sa.66<br />

Gostick, Emma Sa.98<br />

Goto, Daisuke Su.45<br />

Gottlieb, Alice Su.74<br />

Goverman, Joan OR.36<br />

Govindarajan, Rajagopalan OR.61<br />

Goyette, Philippe Su.108, Su.93<br />

Gozin, Michael Su.28<br />

Gozzard, Neil Su.86<br />

Graham, Kareem L. OR.91<br />

Graham, Robert R. OR.80<br />

Green, Todd Su.92, Su.93<br />

Greenbaum, Carla F.28<br />

Greenfield, Edward A. Sa.146<br />

Greer, Allison F.15<br />

Gregersen, Peter OR.55, Sa.55, Sa.61<br />

Gregory, Clare Su.94<br />

Greidinger, Eric L. Sa.28<br />

Greiner, Dale OR.56<br />

Griffiths, Gillian F.81<br />

Grimbacher, Bodo F.89<br />

Gros, Philippe F.27<br />

Gross, Jennifer Sa.97<br />

Grumach, Anete Sevciovic F.96, F.99, Su.75, F.92<br />

Gubbels Bupp, Melanie Sa.129<br />

Guberski, Dennis OR.82<br />

Guillaume, Marie Paule F.90, F.134<br />

Guillen, Cecilia F.51<br />

Guillermo, Roy Sa.120<br />

Gulati, Reema F.13, F.56<br />

Guleria, Indira F.33<br />

Gunderson, Erica Sa.152<br />

Gupta, Santosh F.68<br />

Gürses, Atilla Su.98<br />

Gutenberger, Sylvia F.75<br />

Haaland, Perry Sa.101<br />

Habiby Kermany,<br />

Sa.134<br />

Mohammad<br />

Hackstein, Holger Su.41<br />

Haensch, Gertrud Maria OR.98<br />

Hafler, David A. OR.77, OR.83, OR.94,<br />

OR.95, F.15, F.115,<br />

Sa.114, Sa.120, Sa.139,<br />

Sa.146, Sa.151, Su.27,<br />

Su.35, Su.36, Su.37,<br />

Su.38, Su.41<br />

Hahn, Bevra OR.4, OR.12, Sa.36, Sa.76


FOCIS 2007 Abstract Index by Author<br />

Hahn-Zoric, Mirjana F.122<br />

Hale, Matthew OR.92<br />

Hallam, Robert F.65, F.126<br />

Hamada, Takashi OR.97<br />

Hamm, Stefanie OR.32<br />

Han, David Su.34<br />

Han, May H. Su.34<br />

Han, Meifang Su.136, Su.43<br />

Hanak, Viktor Sa.6<br />

Hang, Howard F.115<br />

Hanlon, Douglas OR.43<br />

Hansen, Anna M. OR.38<br />

Hansen, Lasse Tengbjerg Su.54<br />

Hanson, Imelda C. Sa.12<br />

Hanson, Lars Åke F.122<br />

Hansson, Sverker F.122<br />

Hao, Qian-Lin F.113<br />

Haqqani, Arsalan OR.2<br />

Harley, John B. Sa.24, Sa.79<br />

Harley, John Sa.47, OR.81<br />

Hartnett, Mark F.9<br />

Hartt-Meyers, Jennifer Sa.146<br />

Hasegawa, Minoru Sa.25, Su.63, Su.67<br />

Haskins, Kathryn F.25, OR.3<br />

Hastings, William F.15<br />

Haug, Markus F.135<br />

Hauser, S. Su.93<br />

Haworth, Charles F.65<br />

Haynes, Barton OR.25<br />

Hazzan, Marc Sa.89<br />

Headley, Mark OR.57<br />

Hegde, Ganapati Sa.102, Sa.103<br />

Heidtmann, Antje Su.90<br />

Heinlen, Latisha Sa.79<br />

Hellings, Niels OR.87, Su.18, Su.23<br />

Hendriks, Jerome JJA OR.87<br />

Hennon, Teresa OR.89<br />

Henry, Alistair Su.85<br />

Hensen, Karen Su.23<br />

Heppert, Volkmar F.49<br />

Hering, Bernhard F.24<br />

Hernandez-Flores, Georgina Su.55<br />

Herold, Kevan OR.56<br />

Herrera-Gonzàlez, Norma Sa.110<br />

Herrinton, Lisa Sa.152<br />

Heslan, Michele OR.71, Su.96<br />

Hewitt, Kyle Sa.115<br />

Hickman, Scott F.127<br />

Hickman, Suzanne OR.90<br />

Hiestand, Peter Su.3<br />

Highton, John Sa.88<br />

Hill, Marcello OR.71<br />

Hillary, Steinhart Su.92<br />

Hintermann, Edith OR.62, Sa.126<br />

Hirasawa, Masao Su.53<br />

Hirsch, J. Su.19<br />

Hladikova, Zuzana F.19<br />

Ho, Anthony Sa.113<br />

Ho, I-Cheng Su.40<br />

Ho, Peggy P. Sa.54, Sa.71, Sa.88<br />

Hoefferer, Liane Su.7<br />

H<strong>of</strong>fman, Robert W. Sa.26, Sa.28<br />

Hogan, Simon F.124<br />

Hogan, Susan Sa.136<br />

Hogendoorn, Pancras F.103<br />

3202<br />

Hojaili, Bernard OR.9<br />

Holdener, Martin OR.62<br />

Holers, V Michael Sa.63<br />

Holland, Steven M. OR.52, F.66<br />

Hollenberg, Morley OR.65<br />

Holness, Claire F.9<br />

Holzer, Ursula OR.65<br />

Hood, Zach Su.20<br />

Hoogeboom, Manja F.103<br />

Horai, Reiko OR.29<br />

Horn, Julia F.89<br />

Horwitz, David A. OR.75, Sa.140<br />

Hosseini-Farahabadi, Sara Sa.9, Su.66<br />

Hosseinkhani, Ayda F.46, F.62<br />

Hou, Stephen F.130<br />

Houle, Jean-Francois Su.120<br />

Hrotekova, Zuzana F.19, Sa.95<br />

Hsieh, Meng-Ying F.88<br />

Hu, Lina Sa.145<br />

Hu, Paul OR.64<br />

Hua, Jing Sa.47<br />

Huang, Jing-Long F.88<br />

Huber, Janine OR.40<br />

Hudson, Michael Sa.116<br />

Hueber, Wolfgang Sa.54, Sa.61<br />

Huegel, Alyssa OR.2<br />

Huett, Alan Su.92<br />

Huibregtse, Inge F.116<br />

Husain, Zaheed Su.59, Su.60<br />

Hussain, Shabbir F.23<br />

Huttenlocher, Anna OR.5<br />

Hwang, Hank Su.47<br />

Hwang, Sunil Su.34<br />

Iacomini, John Sa.18<br />

Ifergan, Igal OR.33, OR.88, Su.29<br />

Ikeda, Osamu F.58<br />

Iking-Konert, Christ<strong>of</strong> F.49<br />

Iliev, Iliyan D. OR.70<br />

Im, Suhn-young Su.51<br />

Imitola, Jaime Su.22<br />

Indiveri, Francesco OR.12, Sa.112, Sa.33<br />

Inman, Robert Sa.47, 1201<br />

Inokuma, Margaret Sa.101, Su.118<br />

Inoue, Asuka Su.45<br />

Ishida, Masato Su.52<br />

Ishiura, Nobuko F.108<br />

Ismail, Heba OR.49<br />

Israel, David OR.74<br />

Itano, Andrea Su.69<br />

Ito, Satoshi Sa.48, Su.45<br />

Iwai, Hideyuki F.9, OR.16<br />

Iwakura, Yoichiro OR.35<br />

Iwanami, Keiichi Su.45<br />

Jaberi, Yahya F.1<br />

Jachimowicz, Edward F.129<br />

Jacob, Chaim O. Sa.94<br />

S195


S196 FOCIS 2007 Abstract Index by Author<br />

Jacob, Cristina Miuki Abe F.82<br />

Jacob, Noam OR.93<br />

Jacobi, Christian Su.15<br />

Jacobson, Robert OR.26, F.63<br />

Jacobson, Stefan H. F.122<br />

Jacques, Peggy Sa.23<br />

Jahromi, Mohamed M. OR.78<br />

Jaimes, Kimberly Sa.26<br />

Jaimes, Maria F.133<br />

Jain, Ashish 3201, OR.27<br />

Jaing, Tang-Her F.88<br />

Jakobsen, Bent OR.9, Sa.98<br />

Jalahej, Heyam F.31<br />

James, Judith OR.81, Sa.79<br />

Jamshidzadeh, Akram F.48, F.62<br />

Jann, Monica OR.71<br />

Jasinski, Jean F.17, F.7, OR.1, OR.21<br />

Jave-Suarez, Luis F. Su.55<br />

Jawa, Vibha Su.42<br />

Jeffrey, Lisa F.120<br />

Jennette, J. Charles Sa.136<br />

Jensen-Jarolim, Erika OR.15<br />

Jerath, Maya Su.58<br />

Jerome, Rotter Su.92<br />

Jesus, Adriana Sa.49, Sa.50<br />

Ji, Shaoquan OR.39, Su.47<br />

Jiang, Guoping OR.74<br />

Jill, Giles OR.40<br />

Jimenez-Martinez, Maria Su.64, Su.76<br />

Carmen<br />

Joe, Selby Sa.152<br />

Johannes, Kellsey OR.60<br />

John, Eigth Author Su.107<br />

Johnson, Jonas OR.73<br />

Johnson, Kelly OR.1, OR.21<br />

Joo, Sung-Yeon Sa.90, Sa.91<br />

Jose Francisco,<br />

Sa.52<br />

Muñoz-Valle<br />

Joshi, Avadhut Sa.103<br />

Joshi, Shantaram Sa.103, Sa.116<br />

Josien, Regis OR.71, Su.96<br />

Juan, Gloria Su.69<br />

Juan, Manel F.26<br />

Juang, Yuang-Taung OR.48<br />

Jumnainsong, Amonrat Su.122<br />

Kagoda, Mercy F.113<br />

Kaieda, Shinjiro Su.14<br />

Kallas, Esper F.64<br />

Kalyanasundaram,<br />

F.73<br />

Ramaswamy<br />

Kamphuis, Sylvia F.20<br />

Kandeel, Fouad Sa.87<br />

Kang, Mi Jin Sa.90, Sa.91<br />

Kao, Amy H. Sa.31<br />

Karabeg, Adela OR.64<br />

Karagiannis, Panos Sa.112<br />

Karbach, Julia OR.9<br />

Karlson, Elizabeth W. Sa.53<br />

Karmali, Rafik F.127<br />

Karow, Margaret F.131<br />

Karter, Andrew Sa.152<br />

Kasman, Ian OR.37, Su.84<br />

Kassam, Nasim Sa.146, Su.38<br />

Kastelein, Robert Su.79<br />

Kataoka, Kazuo Su.24<br />

Kattah, Michael Su.132<br />

Katz, Jonathan F.8<br />

Kaufman, Kenneth OR.81<br />

Kawaciuk, Ivan Sa.117<br />

Kawikova, Ivana Su.102<br />

Kebir, Hania OR.33, OR.88, Su.29<br />

Kedem, Hassya F.69<br />

Keller, Baerbel F.75<br />

Kellermann, Sirid-Aimee Sa.52<br />

Kelley, Keith F.137<br />

Kelly, Jennifer OR.81<br />

Kennedy, Jeff Su.85<br />

Kent, Sally F.15, F.24<br />

Kenyon, Norma F.24<br />

Keogh, Elissa Sa.66, Sa.72, Sa.73<br />

Kern, Marlena OR.55<br />

Khaje Daluei, Mohammad Sa.16<br />

Khalili, Houman OR.55<br />

Khatri, Ismat F.38<br />

Khoury, Samia Su.27, Su.35<br />

Kianifard, Farid Sa.5<br />

Kiany, Simin F.45, F.46, F.48, F.62<br />

Kilpatrick, Jeff OR.81<br />

Kim, Han-A Su.51<br />

Kim, Il Hwan Su.64<br />

Kim, Junwoo Sa.134<br />

Kim, Patrick Sa.134<br />

Kim, Seon-Hee OR.89<br />

Kim, Sung-Joo Sa.100<br />

Kimberly, Robert Sa.84<br />

Kimmie, Crystal OR.49<br />

King, Jennifer K. Su.112<br />

King, Marie OR.56<br />

Kiran, Bayram Sa.109<br />

Kirby, Martha OR.29<br />

Kis, Janos F.31<br />

Kitaichi, Nobuyoshi Sa.21<br />

Kivisakk, Pia Su.22<br />

Kivovich, Violetta OR.63<br />

Klareskog, Lars Sa.61<br />

Klein, Mark F.20<br />

Klinkhammer, Thomas Su.46<br />

Knight, Seth Sa.75<br />

Knittelfelder, Regina OR.15<br />

Ko, Hon-Sum Sa.19<br />

Ko, Hyun-Mi Su.51<br />

Kobayashi, Koichi Su.79<br />

Kobayashi, Masakazu F.17<br />

Kodama, Keichi OR.16, F.5, F.9<br />

K<strong>of</strong>feman, Eva Sa.66<br />

Kohlmoos, Cassidy OR.93<br />

Kohm, Adam Su.13<br />

Komura, Kazuhiro Su.63<br />

Komuves, Laszlo Su.84<br />

Korchynska, Olga F.41<br />

Korinets, Yaroslav F.41<br />

Korman, Alan Sa.137<br />

Korn, Thomas OR.19, Su.32


FOCIS 2007 Abstract Index by Author<br />

Koscielna, Katarzyna Sa.8<br />

Koss, Michael OR.93<br />

Kostianovsky, Alex Su.127<br />

Kostov, Georgi Stefanov Sa.108<br />

Kovacs, Jospeh 3201<br />

Kovats, Susan Sa.132<br />

Kozlova, Yulia Sa.101<br />

Kramer, Ivan F.39, F.74<br />

Krensky, Alan M. Sa.10<br />

Kretowski, Adam OR.78<br />

Kretsos, Kosmas Sa.52<br />

Kreuwel, Huub OR.59<br />

Krienke, Stefan Sa.47, 1201<br />

Krueger, Gerald Sa.150<br />

Kruith<strong>of</strong>, Elli Sa.59<br />

Krutsick, Amy Sa.104<br />

Krutzik, Peter OR.92<br />

Kuballa, Petric Su.92<br />

Kuchroo, Juhi OR.101, Su.38<br />

Kuchroo, Vijay K. OR.19, OR.95, F.15,<br />

Sa.146, Su.32<br />

Kudrycki, Katherine Sa.59<br />

Kudva, Yogish OR.61<br />

Kühne, Ronald Sa.129<br />

Kuis, Wietse F.20, Sa.67<br />

Kumar, Krishan Sa.8<br />

Kumar, Vijay F.110<br />

Kulhankova, Katarina Sa.138<br />

Kuo, Cheng-Chin F.121, Su.137<br />

Kuo, Liang-Mou OR.31, OR.74<br />

Kuo, Ming-Ling F.88<br />

Kusunoki, Susumu Su.24<br />

Kwon, Sung(Steve) F.73<br />

Kye, Young Chul Su.49<br />

Kyjovska, Drahomira Sa.95<br />

Kyttaris, Vasileios C. OR.48<br />

La Cava, Antonio OR.12, OR.4, Sa.36,<br />

Sa.76, Su.9<br />

LaCorcia, Gina Su.107<br />

Lage, Agustin Sa.147<br />

Lai, Chen-Yen F.121<br />

Lam, Queenie Lai Kwan OR.98<br />

Lam, Tong OR.65<br />

Lamb, Roberta OR.40<br />

Lamberth, Kasper F.53<br />

Lanchbury, Jerry OR.39<br />

Land, Michael F.100<br />

Landa, Rosa Sa.88<br />

Lang, Jiena F.34<br />

Lanigan, Caroline Su.128<br />

Larocca, Nancy Sa.1<br />

LaRosa, David F.57<br />

Laroy, Wouter Sa.38<br />

Lasitschka, Felix Su.105, Su.91<br />

Lathey, Janet F.120<br />

Latiano, A. Su.93<br />

Lau, Chak-Sing Sa.80<br />

Law, Adam F.12<br />

Lawendowski, Carla Su.107<br />

Lawson, Alastair Su.87<br />

Lawson, Jenifer OR.6<br />

Lederman, Michael F.73<br />

Le, Ngocdiep Sa.86<br />

Lee, Annette Sa.55<br />

Lee, Benhur Sa.151<br />

Lee, Chung Sa.36<br />

Lee, Hai-Chon Sa.13<br />

Lee, Hern-Ku Su.51<br />

Lee, Jin Su.49<br />

Lee, JungHwa Su.49<br />

Lee, Kwang Chul Su.49<br />

Lee, Li-Fen Sa.88<br />

Lee, Michael R. F.34<br />

Lee, Sun min Sa.106<br />

Lee, Tzielan Sa.70<br />

Lee, Wen-I F.88<br />

Lee-Chan, Edwin F.6<br />

Leelayuwat, Chanvit Sa.125, Su.122<br />

Lehman, Thomas Sa.47<br />

Leif, Jean OR.97<br />

Leite, Katia Sa.132<br />

Leiva, Lily E. F.101<br />

Lemar, Hadia Sa.137<br />

Lenert, Petar OR.44<br />

Leon, Juan F.44<br />

Leon-Murguia, Oscar Sa.44<br />

Leotlela, Poloko Sa.115<br />

Leppert, Mark Sa.150<br />

Lerma-Diaz, Jose M. Su.70<br />

Leung, Hin-Tak OR.85<br />

Leung, Lawrence Sa.71<br />

Levi, Dina F.14<br />

Levinson, Arnold F.57<br />

Levinson, Ralph Su.82<br />

Levisetti, Matteo OR.17<br />

Levy, Gary Su.59<br />

Li, Bin Sa.143<br />

Li, De-Kun Sa.152<br />

Li, Haowei Sa.93<br />

Li, Jian OR.40<br />

Li, Marcella F.17<br />

Li, Mu OR.12, OR.47, 1201<br />

Li, Quan-Zhen OR.81<br />

Li, Sharon F.13<br />

Li, Wentian Sa.25, Sa.55<br />

Li, Xinrui Sa.84<br />

Li, Zhuqing Su.78, Su.81<br />

Liang, Chi-Ming F.121<br />

Liang, Shu-Mei F.121, Su.137<br />

Liao, Hua-Xin F.72<br />

Liao, Lingjie Su.121<br />

Liddy, Nathaniel Sa.114<br />

Lierl, Michelle B. Su.131<br />

Liggitt, Denny OR.36<br />

Lightwood, Daniel Su.86<br />

Lim, Wee Kiak Su.78<br />

Limb, Cindy Sa.46<br />

Limón-Camacho, Leonardo Sa.20, Sa.64<br />

Lin, Chia-Lei Sa.87<br />

Lin, Erina F.100<br />

Lin, Jean Sa.31, Sa.35<br />

Lin, Syh-Jae F.92<br />

Linares, Marisela Su.76<br />

S197


S198 FOCIS 2007 Abstract Index by Author<br />

Lindesmith, Lisa F.44<br />

Ling, Eleanor F.16<br />

Ling, Mei Su.92<br />

Linhart, Birgit Sa.11<br />

Linington, Christopher OR.20, 3202<br />

Link, Jason OR.10<br />

Lipets, Irina Su.74<br />

Littman, Dan R. OR.72<br />

Liu, Baoying Su.78, Su.81<br />

Liu, Chau-Ching Sa.31, Sa.35<br />

Liu, Chunyu Sa.55<br />

Liu, Edwin F.17, OR.21<br />

Liu, Mingfeng Su.43<br />

Liu, Peng Su.58<br />

Liu, Quanhai OR.41<br />

Liu, Ruolan Su.9, Su.9<br />

Liu, Shuying F.87, 3201<br />

Liu, Wenxia Sa.134<br />

Liu, Yingge Sa.153<br />

Lock, Christopher OR.96<br />

Logani, Jyoti F.68<br />

Long, S. Alice OR.7, OR.76<br />

Loo, Ray Mun Sa.8<br />

Lopes, Jared E. OR.72<br />

Lopez-Granados, Eduardo F.83<br />

López León Murguía,<br />

Su.140<br />

Oscar Javier<br />

López, Mercedes N. Sa.122<br />

Lorber, Marc Su.102<br />

Lord, James Sa.128<br />

Lories, Rik Sa.23<br />

Loskog, Angelica Sa.100<br />

Louvet, Cedric F.34<br />

Lu, Lina OR.31, OR.74<br />

Lu, Liwei OR.98<br />

Lu, Wen OR.60<br />

Lu, Xinyue Su.134<br />

Luckner, Claudia Sa.113<br />

Luger, Dror OR.35, Su.54<br />

Lühder, Fred Su.5, Su.5<br />

Luna-Baca, Gabriel Andres Su.64, Su.76<br />

Lunardi, Claudio 1202, OR.23<br />

Lundsgaard, Dorthe Su.54<br />

Luo, Jinquan Sa.141<br />

Luo, Xiaoping OR.86, F.50, Su.43,<br />

Su.136<br />

Luster, Andrew OR.90<br />

Lutsenko, Elena Sa.92<br />

Ly, Dalam F.4, F.23<br />

Lyba, Myroslav Su.110<br />

Lyle, Michael Sa.143<br />

Lyons, Kathryn Su.58<br />

Ma, Chi OR.27<br />

Ma, Hak-Ling Su.72<br />

Macaubas, Claudia Sa.62<br />

Macedo, Camila Su.99<br />

Macias-Hernandez, Israel Su.129<br />

Macip-Rodríguez, Perla Sa.64<br />

Macpherson, Michael OR.100<br />

Maecker, Holden F.133, Sa.101, Su.118<br />

Magliocco, Melissa Su.74<br />

Mahadevan, Daruka Su.46<br />

Mahesh, Sankara Su.81<br />

Mahesh, Sankaranarayana Su.78<br />

Mahon, Tara OR.9, Sa.98<br />

Maier, Lisa OR.83<br />

Maino, Vernon Sa.101<br />

Majumdar, Anish Su.127<br />

Malik, Sanober Sa.24<br />

Malter, James OR.99<br />

Mamura, Mizuko Sa.40, Su.61<br />

Manak, Mark F.120<br />

Manku, Harinder OR.80<br />

Manns, Michael OR.62<br />

Mansour, Eli F.99<br />

Manzi, Susan Sa.35<br />

Marcenaro, Stefania F.81<br />

Marchetti, Piero F.24<br />

Marcondes, Maria Cecilia Su.128<br />

Marietta, Eric F.116<br />

Marinkovich, Vincent A. F.128, F.132<br />

Marino Vazquez, Lluvia Su.118<br />

Marisis, Tatiane F.131<br />

Mark, Daly Su.92<br />

Markert, Louise F.84<br />

Markovic, Svetomir Sa.119<br />

Marquez, Maria Elena Sa.85, Su.126<br />

Marrero, Idania F.30<br />

Marti, Luciana F.64<br />

Martín Márquez, Beatriz Teresita Sa.44, Su.140<br />

Martin, Andrew Su.87<br />

Martin, Tammy Su.79<br />

Martínez García, Erika Su.140, Sa.44<br />

Martini, Alberto Sa.130, Su.137<br />

Martini, Stefania F.85<br />

Martinic, Marianne 2201, F.7<br />

Maryam, Tadayon F.106<br />

Masewicz, Susan A. F.32<br />

Massanari, Marc Sa.5<br />

Massoco, Cristina F.131, Sa.121<br />

Matarese, Giuseppe OR.4<br />

Matejkova, Eva Sa.95<br />

Mather, Ian OR.20<br />

Mathey, Emily 3202<br />

Mathieu, Suzanne Sa.32<br />

Matozan, Katja Su.7<br />

Matsevitaya, Irina Sa.92<br />

Matsuda, Tadashi F.58, Su.52<br />

Matsumoto, Isao Sa.40, Su.45<br />

Matsumoto, Mitsuru OR.24, Sa.141<br />

Matthews, Kate OR.64<br />

Maykut, Robert Sa.5<br />

Mazhani, Maryam Sa.48<br />

McArdel, Shannon Sa.114, Su.31<br />

McArthur, Grant Su.54<br />

McClain, Micah Sa.79<br />

McCrae, Ellie OR.41<br />

McGrail, Kieran OR.2<br />

McNeal, Erin-Joi F.44<br />

McPherson, Michael Su.89<br />

Means, Terry OR.90<br />

Meinl, Edgar 3202<br />

Mellins, Elizabeth Sa.148, Sa.62


FOCIS 2007 Abstract Index by Author<br />

Mendonça, Marcelo F.52<br />

Mendoza, Felipe Su.138<br />

Meraz-Rios, Marco Antonio Sa.110<br />

Merrill, Joan OR.81<br />

Mesirov, Jill Su.46<br />

Metes, Diana Su.99<br />

Meuer, Stefan Sa.113, Su.15, Su.90,<br />

Su.91, Su.100<br />

Meuwissen, Pieter Su.36<br />

Miao, Dongmei OR.21, F.17<br />

Michalek, Jaroslav F.19, Sa.104<br />

Michaud, Gregory Sa.127<br />

Midoro-Horiuti, Terumi OR.22<br />

Mielants, Herman Sa.23<br />

Miescher, Sylvia Su.22<br />

Miethke, Alexander Su.123<br />

Miettunen, Paivi Sa.69<br />

Miguel, Regueiro Su.92<br />

Mikita, Allison F.3<br />

Miklos, David B. OR.43<br />

Mileti, Erika OR.70<br />

Milkovich, Kimberly F.71<br />

Miller, Stephen OR.19, OR.34<br />

Miller-Graziano, Carol F.70<br />

Millonig, Alban Su.8<br />

Milosevic, Ivan F.105<br />

Min, Wei-Ping OR.42, OR.47, 1201<br />

Minarik, Ivo Sa.117<br />

Mingari, Maria Cristina Su.139<br />

Minnig, Kathrin Su.7<br />

Minor, Dana F.101<br />

Mi Qing, Sheng F.23<br />

Mirel, Daniel Su.109<br />

Misawa, Tamako Su.6<br />

Misbah, Siraj F.80<br />

Misiara, Antonio Sa.123, F.131<br />

Misiura, Angela F.41<br />

Mistry, Jehangir Su.47<br />

Miyake, Sachiko Su.12, Su.13, Su.14<br />

Miyamoto, Katsuichi Su.24<br />

Miyashiro, Joy Sa.142<br />

Miyashita, Minoru Su.53<br />

Mizuno, Miho Su.14<br />

Mo, Lian Su.99<br />

Modrusan, Zora Su.84<br />

Moe, Christine F.44<br />

Moheghi, Nasrin Su.50, Su.124<br />

Molloy, Peter Sa.98<br />

Monjure, Hanh F.101<br />

Monsonego, Alon OR.38, Su.10<br />

Montaño, Efrain Su.98<br />

Montano-Gonzales, Efrain Su.95<br />

Monteon, Francisco Su.98<br />

Monteon-Ramos, Francisco Su.112<br />

Montero, Enrique Sa.147<br />

Montgomery, Mitzi Su.21<br />

Montoya, Margarita F.51<br />

Moonka, Dilip F.71<br />

Moore, Adrian Su.86<br />

Moore, Carolina OR.97<br />

Moore, Marcus Su.52<br />

Mor, Guillermo Sa.116<br />

Moraes Vasconcelos, Dewton F.67, F.92, F.96, F.97,<br />

F.98, F.103, F.131, Sa.121,<br />

Sa.123, Su.75<br />

Morales Buenrostro, Luis Su.101<br />

Mordes, John OR.56, OR.82<br />

Moreau, Aurelie Su.96<br />

Moreira-Filho, Carlos F.66<br />

Moreno, Dolores Sa.1<br />

Moreno-Fierros, Leticia Su.56<br />

Moretta, Lorenzo F.81, Sa.130, Su.139<br />

Morrow, Matthew R. F.86<br />

Moshk<strong>of</strong>f, Dmitry F.120<br />

Moskowitz, Keith F.120<br />

Motterlini, Roberto Su.119<br />

Moullier, Philippe Su.96<br />

Moulton, Vaishali R. OR.48<br />

Mouri, Yasuhiro Sa.141<br />

Mouritzen, Ulrik Su.54<br />

Moviglia, Gustavo A. Su.19<br />

Moviglia, M.T. Su.19<br />

Moya, R. Su.19<br />

Muczynski, Kimberly Su.113<br />

Mueller, Isabelle OR.76<br />

Munder, Markus Sa.113<br />

Munger, Corey Sa.102, Sa.103<br />

Munirathinam, Gnanasekar F.73<br />

Muñoz Valle, José Francisco Su.140<br />

Muñoz-Chable, Olga Su.129<br />

Munroe, Melissa E. Sa.68<br />

Muromoto, Ryuta Su.52<br />

Murphy, Andrew F.124<br />

Murray, Joseph F.116<br />

Murtaza, Anwar Sa.32<br />

Myles, Timothy Sa.78<br />

Nadeau, Kari C. Sa.10<br />

Nahill, Sharon OR.19, Su.107<br />

Nakajima, Toshihiro Sa.56<br />

Nakayama, Maki OR.1, OR.21, F.17<br />

Nance, Christina OR.26<br />

Nasr, Mohamed Sa.138<br />

Navratil, Jeannine S. Sa.31<br />

Nazneen, A. Su.114<br />

Ndejembi, Modesta OR.46<br />

Neamatollahi, Hossein Su.124<br />

Neethling, Francisca OR.54, Sa.106<br />

Negi, Surendra OR.22<br />

Negri Santi, Tatiana F.69, F.97<br />

Neil, Risch Sa.152<br />

Nejad Shahrokhabadi, Khadijeh Sa.105<br />

Nelson, David 3201<br />

Nelson, Edward F.130<br />

Nelson, J. Lee Su.113<br />

Nemirovsky, Anna Su.126<br />

Nepom, Gerald OR.51, F.32, Sa.128<br />

Nesbitt, Andrew Su.85, Su.86<br />

Nesheim, Steven F.94<br />

Nesspor, Tom OR.40<br />

Nestle, Frank OR.39<br />

Nevin, Barry Sa.113<br />

Newell, Karen OR.41<br />

Newsom, Brian Su.21<br />

S199


S200 FOCIS 2007 Abstract Index by Author<br />

Ng, Gordon Su.69<br />

Nguyen, Alex F.53<br />

Nguyen, Khoa Sa.10, Sa.62<br />

Nguyen, Tffany OR.54, Sa.106<br />

Nickol<strong>of</strong>f, Brian Sa.115<br />

Nico, Marcelo Mentha Su.75<br />

Niesters, Marieke F.103<br />

Niewold, Timothy Sa.47<br />

Nikoopour, Enayat F.6<br />

Ning, Bo OR.22<br />

Ning, Qin F.50, Su.43, Su.121,<br />

Su.136<br />

Nishimoto, Kevin F.130<br />

Nishimura, Toshihiko Sa.71<br />

Nishioka, Kusuki Sa.56<br />

Nishiya, Ana F.92<br />

Noaman, Eman Sa.124<br />

Nolan, Garry OR.92<br />

Nolan, GP OR.50<br />

Norowski, Elaine OR.82<br />

Notarangelo, Luigi F.91<br />

Noval Rivas, Magali Sa.89<br />

Nussenblatt, Robert Su.78, Su.81<br />

O’Brien, Peter Sa.104<br />

Ochs, Hans F.78, F.79, 3201<br />

O’Conner, Kevin Sa.114, Su.31, Su.47<br />

Offner, Halina OR.10, OR.13<br />

Ogawa, Masafumi Su.12<br />

Ohno, Shigeaki Sa.21<br />

Okamoto, Akiko Sa.22<br />

Okamura, Ross Su.109<br />

Oki, Shinji Su.13, Su.14<br />

Oksenberg, Jorge OR.69, Su.93<br />

Oldham, Elizabeth OR.39<br />

Oldham, Janine 2201<br />

Oliveira, Joao B. F.82, F.92, Sa.49, F.96,<br />

Su.75, Sa.50<br />

Olson, Julie Su.26<br />

Ondr, Jennifer F.8<br />

O’Neil, William M. Sa.43<br />

Onengut-Gumuscu, Suna Sa.148<br />

Ootsuka, Takao Su.14<br />

Opoka, Robert F.8<br />

O’Quinn, Darrell 3203<br />

Orange, Jordan F.76<br />

Orban, Tihamer F.31<br />

Oreizi, Farzad F.125<br />

Orihuela, Ana Su.31<br />

Orii, Noemia F.96<br />

Oritani, Kenji F.58, Su.52<br />

Orlovsky, Yevgeniya OR.40<br />

Orru, Valeria Sa.153<br />

Ortiz-Lazareno, Pablo C. Su.55<br />

O’Shea, Adam F.13, F.56<br />

Oshidary, Neekaan Su.132<br />

Ostankov, Maxim Sa.92<br />

Oukka, Mohamed OR.19, Su.32<br />

Ousman, Shalina OR.69, OR.94<br />

Out, Theo Sa.58<br />

Ouyang, Wenjun OR.37<br />

Ovsyannikova, Inna F.63<br />

Packwood, Kerri F.83<br />

Padyukov, Leonid F.122<br />

Page, Matt Su.87<br />

Paglia, Michael F.13, F.56<br />

Pahwa, Savita F.94<br />

Pai, Sung-Yun Su.40<br />

Palframan, Roger Su.86<br />

Palian, Beth Sa.140<br />

Palleros, C.A. Su.19<br />

Palmer, Jeanne Sa.86<br />

Palmer, Jerry OR.49<br />

Palumbo, Michael F.14<br />

Palumbo, Paul F.94<br />

Pan, Kuang-Hung Sa.62<br />

Pandey, Niranjan Su.57<br />

Pantanelli, Seth Su.81<br />

Pareigis, Elizabeth R. OR.44<br />

Paris, Kenneth F.101<br />

Parker, Alex OR.8<br />

Parrish, Yasmin F.113<br />

Pasalar, Mehdi F.114<br />

Passerini, Laura F.91<br />

Paston, Samantha Sa.98<br />

Pastorino, Antonio Carlos F.82<br />

Patel, Dhavalkumar Su.39, Su.58<br />

Paterson, Blake Su.57<br />

Pathoulakis, Charalabos F.61<br />

Patil, Tanvi F.38<br />

Payne, Kimberly F.113<br />

Peakman, Mark OR.6, OR.18<br />

Pearle, Andrew Su.116<br />

Pearson, Todd OR.56<br />

Pease, Larry Sa.119<br />

Peckham, Russell Su.134<br />

Pegram, Mark Sa.107<br />

Penaranda, Cristina OR.59<br />

Pende, Daniela F.81<br />

Peng, Stanford Sa.129, Su.57<br />

Peng, Ruoqi Su.58<br />

Penny, Richard Sa.18<br />

Pereda, Cristian Sa.122<br />

Perez, OD OR.50<br />

Perez, Rolando Sa.147<br />

Perez-Tapia, Mayra Su.76<br />

Peritt, David OR.40<br />

Perkins, Izabella F.44<br />

Perrin, Steve OR.55<br />

Perroni, Lucia Beatrice F.91<br />

Pesce, Barbara Sa.65<br />

Pessoa, Mário F.64<br />

Peter, Hans Hartmut OR.32, F.75, F.89<br />

Peter, Lipsky OR.81<br />

Peterlana, Dimitri OR.23, 1202<br />

Petrovic, Aleksandra F.86<br />

Petrovic-Stojkovic, Sanja Su.28<br />

Petry, Douglas OR.101<br />

Pi, Bin Su.136<br />

Pieri, Patrícia de Campos F.82<br />

Pietra, Gabriella Su.139<br />

Pihoker, Catherine F.28<br />

Pilat, Nina Sa.11<br />

Pillai, Asha OR.68


FOCIS 2007 Abstract Index by Author<br />

Pinsky, Norman F.63<br />

Pinto, Jorge Andrade F.99<br />

Pires Correa, Alexandre F.99<br />

Pitashny, Milena Sa.27<br />

Pixley, John S. Sa.43<br />

Plagnol, Vincent OR.85<br />

Planck, Steve Su.79<br />

Planck, Steven OR.84<br />

Ploegh, Hidde F.115<br />

Pober, Jordan Su.102, Su.104<br />

Pociot, Flemming F.22<br />

Poland, Gregory F.63<br />

Polychronakos, Constantin F.14<br />

Ponath, Paul F.58<br />

Ponte, Joe F.56<br />

Poole, Brian D. OR.63<br />

Popescu, Iulia Su.99<br />

Popov, Igor Sa.47, 1201<br />

Popplewell, Andrew Su.87<br />

Porcelli, A. F.23<br />

Prakken, Berent F.20, Sa.67<br />

Prasad, Shashi OR.43<br />

Prat, Alexandre OR.33, OR.88, Su.29<br />

Pratt, Nicol F.32<br />

Prefumo, Federico Su.139<br />

Pressler, Barrrak Sa.136<br />

Prestigiacomo, Tony Sa.79<br />

Preston, Ben Sa.137<br />

Preston, Gloria Sa.136<br />

Pricop, Luminita OR.93<br />

Priest, Catherine Su.109<br />

Prokoptchuk, Natalia F.41<br />

Puccetti, Antonio OR.23, 1202<br />

Puckett, Lindsay Su.28<br />

Pugliese, Alberto F.18, F.24<br />

Pujol Borrell, Ricardo F.26<br />

Punwani, Divya F.83<br />

Purbhoo, Marco OR.9<br />

Purcell, Shaun F.22<br />

Putterman, Chaim OR.93<br />

Pyne, Saumyadipta Su.36<br />

Qian, Shiguang OR.31, OR.74<br />

Rabellino, Enrique F.72, F.129<br />

Rachitskaya, Aleksandra V. OR.38<br />

Rafatpanah, Houshang Sa.9, Su.66<br />

Ragheb, Jack F.134<br />

Raimondi, Giorgio Su.105<br />

Rajagopolan, Govind Sa.78<br />

Rakhmanov, Mirzokhid F.75<br />

Rakhshandeh, Hassan Sa.105<br />

Ram, Aarthi OR.26<br />

Raman, Girija Su.94<br />

Ramanujam, Meera Sa.45<br />

Ramos Moreira Leite, Katia Sa.123<br />

Rao, Deepak A. Su.104<br />

Rapacki, Krist<strong>of</strong>fer F.22<br />

Rasband, Matthew Su.26, 3202<br />

Rashtak, Shadi F.116<br />

Rasouli, Manoochehr F.45, F.46, F.48, F.64<br />

Rassassi, Khadir Sa.151, Su.37<br />

Ratnayake, Chitra K. F.72<br />

Ravetti, Jean Louis Sa.112<br />

Ray, Pratima F.70<br />

Razzaque, M.S. Su.114<br />

Read, Jennifer F.94<br />

Reddy, Jay Su.32<br />

Reich, David Sa.139<br />

Reichardt, Holger Su.20<br />

Reid, Jessica Sa.96<br />

Reiff, Andreas OR.79<br />

Relman, David F.69<br />

Remy, Séverine Su.119<br />

Rescigno, Maria OR.70<br />

Reséndiz Albor, Aldo Arturo Su.56<br />

Rewers, Marian J. OR.78<br />

Reyburn, Hugh Su.122<br />

Reyes, Candice F.136<br />

Reyes, Diego Sa.122<br />

Richard, Julie OR.19<br />

Richards, William Sa.51<br />

Ricordi, Camillo F.24<br />

Rider, Beverley J. OR.23, F.6<br />

Rieben, Robert Su.7<br />

Rieck, Mary F.28, OR.76, Sa.148<br />

Riedl, Marc F.100<br />

Riemer, Angelika B. OR.15<br />

Rigato, Paula Ordonhez Sa.16<br />

Riggs, James Sa.96<br />

Rihal, Pardeep S. Sa.12<br />

Riker, Adam Sa.115<br />

Riley, Christopher Su.46<br />

Rinderknecht, Cornelia Sa.132<br />

Rioux, John Sa.74, Su.92, Su.93<br />

Rita, Bottino OR.56<br />

Rivitti, Evandro F.99<br />

Rizzi, Marta OR.12, OR.32, Sa.33<br />

Robbins, Paul Sa.91<br />

Roberto, Eigth Author F.24<br />

Roberts, Robert OR.42, F.87<br />

Robinson, William OR.94, Sa.54, Sa.61,<br />

Sa.71, Su.34<br />

Robotham, Jason M. Sa.18<br />

Roby, Keith Su.48<br />

Rodrigo, Evelyn 2201<br />

Rodrigues, Denise F.64<br />

Rodriguez Manzanet, Roselynn Sa.146<br />

Rodriguez, Benigno F.71<br />

Rodriguez, Miguel Su.95, Su.98<br />

Roep, Bart F.2, OR.11, OR.6<br />

Rogerio, Jaqueline F.120<br />

Rojas, Mauricio Su.58<br />

Romeo, Elisa<br />

Römisch, Jürgen Su.15<br />

Roncarolo, Maria Grazia F.91<br />

Ronchese, Franca Sa.6<br />

Roord, Sarah Sa.67<br />

Rosenbaum, James OR.84, Su.79<br />

Rosengren, Sanna Sa.70<br />

Rosenthal, Devin Sa.115<br />

Rosenzweig, Holly OR.84, Su.79<br />

Rosenzweig, Michael F.114, F.56, F.13<br />

Rossin, Elizabeth Su.27, Su.35, Su.36<br />

S201


S202 FOCIS 2007 Abstract Index by Author<br />

Rossini, Aldo OR.56<br />

Rothenberg, Marc F.124<br />

Rottembourg, Diane OR.14<br />

Rottiers, Pieter F.116, Sa.46<br />

Rouse, Todd Sa.138<br />

Roux, Kenneth H. Sa.17, Sa.18<br />

Roy, Debasmita Su.39<br />

Roy, Dolly Su.34<br />

Royer, Pierre-Joseph Su.119<br />

Rubertone, Mark Sa.79<br />

Rubinstein, Mark Su.61<br />

Ruitenberg, Joyce Sa.133<br />

Rummens, Jean-Luc Su.23<br />

Ruzek, Melanie OR.19<br />

Ryan, Jenna F.63<br />

Ryu, Jay Sa.6<br />

S. Grumach, Anete F.67, F.98, F.101<br />

Sabater, Lidia F.26<br />

Sabeti, Pardis Sa.74<br />

Saikali, Philippe Su.16<br />

Saito, Yuki Su.67<br />

Salazar, Lorena Sa.65<br />

Salazar-Onfray, Flavio OR.45, Sa.65, Sa.122<br />

Salek Moghddam, Alireza Sa.7<br />

Salman, Andac Sa.109<br />

Saltzman, Mark Sa.110<br />

Salzer, Ulrich OR.32, F.89<br />

Sampaio, Elizabeth F.66<br />

Samstag, Yvonne<br />

Sánchez Hernández,<br />

Pedro Ernesto<br />

Su.90<br />

Sanda, Srinath F.2<br />

Sandborg, Christy Sa.62<br />

Santacruz-Valdes,<br />

Su.64, Su.76<br />

Concepcion<br />

Santamaria, Pere F.21<br />

Santoro, Alessandra F.81<br />

Saoudi, Abdelhadi Su.111<br />

Sarraf, Alireza Su.124<br />

Sathe, Shridhar Sa.17, Sa.18<br />

Sato, Jun-ichi Su.6<br />

Sato, Maria Notomi Sa.16<br />

Sato, Shinichi Sa.33, Su.63<br />

Sawalha, Amr H. OR.81, Sa.24<br />

Sayegh, Mohamed Su.22<br />

Scalapino, Kenneth Sa.82<br />

Scallon, Bernie OR.40<br />

Scanzello, Carla Su.116<br />

Schaefer, Catherine Sa.152<br />

Schartner, Jill OR.5<br />

Schatz, Desmond OR.2<br />

Schein, Catherine Sa.5<br />

Schenk, Erin Sa.119<br />

Schett, Georg Sa.51<br />

Schlesier, Michael F.89<br />

Schnell, Christian Su.3<br />

Schoeb, Trenton 3203<br />

Scholler, Nathalie Sa.97<br />

Schout, Denise F.52<br />

Schreiner, Bettina OR.34<br />

Schrodi, Steven OR.69<br />

Schroeder, Andreas Su.90<br />

Schroeder-Braunstein, Jutta Su.91<br />

Schwartzberg, Pamela L. OR.29<br />

Schweitzer, Barry Sa.116<br />

Sc<strong>of</strong>ield, R Hal Sa.24<br />

Scott, Benjamin Sa.42<br />

Seamon, Vanessa Sa.17<br />

Seddiki, Nabila OR.30<br />

Seeborg, Filiz Sa.12<br />

Seeuws, Sylvie Sa.23<br />

Segal, Gabriela Sa.122<br />

Segura, Jorge F.51<br />

Seidu, Luqman F.124<br />

Sekine, Yuichi F.58, Su.67<br />

Selby, Mark Sa.137<br />

Selman, Moises Su.138<br />

Sercarz, Eli F.27, F.30<br />

Seroogy, Christine F.110<br />

Servais, Genevieve F.127<br />

Seshasayee, Dhaya Sa.149<br />

Sessarego, Marta Sa.33<br />

Sette, Jr., Hoel F.64<br />

Setty Balakrishnan, Anand F.73<br />

Sewell, Andy Sa.98<br />

Seyeddi, Ronak Sa.85<br />

Seyfer, Sarah OR.44<br />

Seyfert-Margolis, Vicki Su.1, Su.50<br />

Shampain, Kimberly L. Sa.114<br />

Shao, Wenhai OR.58<br />

Sharif, Shadi Sa.71<br />

Sharifi, Faranak F.1<br />

Sharma, Meera F.79<br />

Sharp, Veronika OR.50<br />

Shea, Yvonne F.68<br />

Shealy, David OR.40<br />

Shearer, William OR.26, F.94<br />

Shen, Guanxin Su.121<br />

Shen, Jijia F.134<br />

Shen, Ling Sa.152<br />

Shen, Zhong-Jian OR.99<br />

Sheng, Shijun Su.133<br />

Shi, Fu-Dong Su.9<br />

Shi, Jishu F.61<br />

Shirdel, Babak F.27<br />

Shivakumar, Pranavkumar Su.123<br />

Shizuru, Judith Sa.88<br />

Shoda, Lisl OR.59<br />

Shohreh, Farshad F.62<br />

Shoor, Stanford Sa.152<br />

Shugart, Yin Yao Su.92<br />

Shultz, Leonard OR.56<br />

Shum, Anthony Su.111<br />

Shum, Tony F.12<br />

Sido, Bernd Su.90, Su.91<br />

Siebert, Janet Sa.101, Su.118<br />

Siegel, Richard M. OR.29<br />

Sierra, Juan F.93<br />

Silberman, Daniel Sa.96<br />

Silva, Clovis Sa.56, Sa.57<br />

Silva, Léia C. Rodrigues F.52<br />

Silva, Nubia Su.108<br />

Silver, Phyllis OR.35, OR.38


FOCIS 2007 Abstract Index by Author<br />

Silverberg, Mark Su.92<br />

Sim, Davis Sa.78<br />

Simonian, Michael H. F.72<br />

Simonson, William OR.5<br />

Sims, Martin Sa.42<br />

Sin, Sang-Hoon Su.39<br />

Singer, Josef OR.15<br />

Singh, Bhagirath F.6<br />

Singh, Harvir Sa.30<br />

Singh, Manisha F.55<br />

Singh, Ram Raj OR.66, Sa.76, Sa.94,<br />

Su.112, Sa.135<br />

Sirota, Marina OR.53<br />

Siwak, Edward OR.26<br />

Skak, Kresten Su.54<br />

Skowera, Anna OR.18<br />

Skrumsager, Birte K. Su.69<br />

Slaets, Leen OR.87<br />

Slavik, Jacqueline Sa.146<br />

Slavonic, Michael F.13<br />

Sleasman, John W. F.90<br />

Smit, Helga Su.114<br />

Smith, Deborah Su.112<br />

Smith, Michael Sa.116<br />

Smitz, John Sa.136<br />

Smogorzewska, Monika F.113<br />

Smyth, Deborah OR.85<br />

Snowden, James Su.87<br />

Snyder, James F.134<br />

Snyder, Michael Sa.116<br />

Sobel, Raymond A. Sa.146, OR.69, OR.91,<br />

OR.94, Sa.97, Su.32,<br />

Su.34<br />

Sochorova, Klara F.109<br />

Sohn, Jeongwon Su.49<br />

Solbrig, Camille OR.43<br />

Soler-Ferran, Dulce Su.27<br />

Somers, Veerle Su.18, Su.23<br />

Somerville, John Sa.106<br />

Sommerer, Claudia Su.100<br />

Son, Hye-Youn Sa.90, Sa.91<br />

Son, Sang Wook Su.49<br />

Song, Jason Sa.71<br />

Soper, David OR.8<br />

Sorensen, Ricardo U. OR.5<br />

Soto-Abraham, Virginia Sa.72<br />

Soulillou, Jean-Paul Sa.65<br />

Sousa-Canavez, Juliana F.131, Sa.121, Sa.123<br />

Spier, Catherine Su.46<br />

Sprent, Jonathan Su.61<br />

Spycher, Martin O. Su.7<br />

Sripa, Banchob Sa.111<br />

Standifer, Nathan OR.51<br />

Stanimirovic, Danica OR.88<br />

Staquet, Kim OR.40<br />

Stechova, Katerina F.19<br />

Steinman, Lawrence OR.69, OR.94, OR.96,<br />

Su.34<br />

Stephens, Tracey A. F.6<br />

Sternjak, Alexander OR.10<br />

Stevens, Anne Su.113<br />

Stevens, Christine Sa.74, Su.93<br />

Stevens, Helen OR.85<br />

Still, Travis F.24<br />

Stinissen, Piet OR.87, Su.18, Su.23,<br />

Su.31<br />

Stolina, Marina Sa.51<br />

Storch, Maria OR.20, 3202<br />

Stourac, Petr F.62<br />

Straub, Irena F.135<br />

Strauss, Laura OR.73<br />

Strober, Samuel OR.68<br />

Strober, Warren 3201<br />

Strobl, Frank Sa.104<br />

Strom, Terry B. Sa.146<br />

Stromnes, Ingunn OR.36<br />

Struthers, Mary Su.58<br />

Su, Kai Su.136<br />

Su, Kaihong Sa.84<br />

Su, Maureen F.12<br />

Suen, Yu Su.48<br />

Suez, Daniel F.78, F.79<br />

Suggs, Sid F.137<br />

Sugiyama, Kenji Su.52<br />

Sumida, Takayuki Sa.40, Su.45<br />

Sun, Hongtao 1201<br />

Sun, Lulu F.102<br />

Sun, Xiaocui OR.72<br />

Supter, Tina Su.105<br />

Suresh, Lakshmanan F.110<br />

Sutton, Deborah OR.9, Sa.98<br />

Suzuki, Motohiko OR.42, OR.47, 1201<br />

Swanson, Steven J. Su.42<br />

Swerkersson, Svante F.122<br />

Swiergala, Elzbieta F.22<br />

Swistak, Mark Su.107<br />

Szalai, Alexander Sa.84<br />

Szereday, Laszlo F.31<br />

Szot, Gregory L. F.34<br />

Szuhai, Karoly F.103<br />

Tabunoki, Hiroko Su.6<br />

Tachdjian, Raffi F.100<br />

Tagawa, Asako OR.24<br />

Taguchi, T. Su.114<br />

Tahmasebifar, Neda F.122<br />

Tai, Yi Su.102<br />

Tak, Paul-Peter Sa.57, Sa.58, Sa.59<br />

Takahashi, Kazue Sa.63<br />

Takehara, Kazuhiko Sa.25, Su.67<br />

Tálamo, Carlos Sa.1<br />

Tamaki, Kunihiko F.108, Su.68<br />

Tamayo, Pablo Su.36<br />

Tamiz, Amir Su.57<br />

Taneja, Veena Sa.154<br />

Tang, Anita OR.46<br />

Tang, Liren Sa.143<br />

Tang, MengXiang Sa.101<br />

Tang, Qizhi OR.59, F.33<br />

Tanguy-Royer, Séverine Su.119<br />

Tanji, Maury M. F.52<br />

Tasch, Michael Su.113<br />

Tassinari, Paolo Su.108<br />

Tati, Abdellatif F.103<br />

S203


S204 FOCIS 2007 Abstract Index by Author<br />

Taub, Dennis Sa.115<br />

Tavakkol Afshari, Jalil Sa.9, Sa.48, Sa.105,<br />

Su.50, Su.62, Su.66,<br />

Su.124<br />

Tavares, R.C. F.104<br />

Tawde, Pallavi Sa.17, Sa.18<br />

Taylor, Kent Su.92<br />

Taylor, Paul Su.117<br />

Tedder, Thomas Su.85<br />

Teklenburg, Gijs F.20<br />

Tellides, George Su.119, Su.129<br />

Temmerman, Stephane OR.27<br />

Tenenbaum, Jessica Sa.46<br />

Terrett, Jon OR.72<br />

Tesson, Laurent Su.119<br />

Teuber, Suzanne OR.50, OR.63<br />

Thebault, Pamela OR.71<br />

Thewissen, Marielle Su.18, Su.23<br />

Thiel, Jens F.85, F.95<br />

Thomas, John F.137<br />

Thomson, Angus Su.105<br />

Thornton, Angela M. OR.29<br />

Thurlings, Rogier Sa.58<br />

Tian, Yan F.3<br />

Tibshirani, Robert Sa.69<br />

Timmel, Amanda OR.5<br />

Timmer, Trieneke Sa.58<br />

Tirabasi, Rebecca OR.82<br />

Tisch, Roland Sa.136<br />

Tiwari, Ruby OR.22<br />

Tiwari-Woodruff, Seema Su.112<br />

Tobiasova, Zuzana F.109, Sa.117<br />

Todd, John A. F.11<br />

Todd, John OR.85<br />

Todorov, Ivan Sa.87<br />

T<strong>of</strong>t, Michelle OR.90<br />

Togher, Lisa OR.14, 2201, F.7, F.16<br />

Tomi, Chiharu Su.28<br />

Tommasini, Alberto F.95<br />

Tomooka, Beren OR.94, Sa.61<br />

Tonel, Giulia OR.39<br />

Tonkin, Daniel OR.3<br />

Toro, Felix Su.108<br />

Torre, Giancarlo Sa.112<br />

Torres-Lozano, Carlos F.77, Su.70<br />

Tötterman, Thomas Sa.100<br />

Touil, Tarik OR.41<br />

Traggiai, Elisabetta Sa.130<br />

Traverso, Paolo Sa.112<br />

Tree, Timothy OR.6, OR.18<br />

Trembath, Richard OR.39<br />

Tremoulet, Adriana Sa.74<br />

Treszl, Andras F.31<br />

Tripp, Catherine Sa.32<br />

Trucco, Massimo OR.56<br />

Trueblood, Esther Su.69<br />

Tsokos, George C. OR.48, Su.134<br />

Tsoukas, Christos M. F.91<br />

Tsuchihashi, Sei-ichiro Su.128<br />

Tsutsumi, Akito Sa.40, Su.45<br />

Turna, Akif Sa.109<br />

Tyson, Kerry Su.87<br />

Uccelli, Antonio Sa.130<br />

Ueda, Masumi OR.28, Sa.125<br />

Untersmayr, Eva OR.15<br />

Uratsu, Sandra Sa.17<br />

Urban, Nicole Sa.97<br />

Utz, Paul J. OR.50, OR.75, Sa.30,<br />

Su.132<br />

Uzel, Gulbu OR.52<br />

Vacca, Paola Su.139<br />

Vaickus, Lou F.114<br />

Vaitaitis, Gisela M. OR.67<br />

Valdes-Ferrer, Sergio Ivan F.93<br />

Valdez, Patricia OR.37<br />

Valenta, Rudolf Sa.11<br />

Valenzuela, David F.124<br />

Vales-Alberto, Luis Su.95<br />

Vales-Gomez, Mar Su.122<br />

Valle, Sollange F.99<br />

Van Belle, Tom F.16<br />

Van Beneden, Katrien Sa.23, Sa.38<br />

van de Ven, Annick Sa.72, Sa.73<br />

van den Brandt, Jens Su.5<br />

van der Pouw-Kraan, Tineke Sa.58<br />

van Deventer, Sander<br />

Van Eyk, Jennifer<br />

F.116<br />

van Kooten, Cees F.122<br />

Van Landeghen, Megan OR.7<br />

Van Noort, Johannes OR.94<br />

van Rooijen, Nico Sa.22<br />

van Wijk, Femke Sa.67<br />

Vandenbark, Arthur OR.10, OR.13<br />

Vanderlocht, Joris OR.87<br />

Vandooren, Bernard Sa.58, Sa.59<br />

Varela, Gabriela Su.19<br />

Varela, Jorge Alcocer Sa.20<br />

Vargas Rojas, Maria Ines F.93, Sa.20, Sa.64, Sa.81,<br />

Su.101<br />

Vasquez, Ismael Sa.110<br />

Vatseba, Roman Su.110<br />

Vázquez Del Mercado Espin, Sa.44, Su.140<br />

Mónica<br />

Venken, Koen Su.18, Su.23<br />

Verbruggen, Gust Sa.23, Sa.38<br />

Verrijn Stuart, Annemarie F.20<br />

Veys, Eric M. Sa.59<br />

Victor, Jeferson Russo Sa.16<br />

Vidlakova, Petra Sa.95<br />

Vierkant, Robert F.63<br />

Vieths, Stefan Sa.17<br />

Viglietta, Vissia Su.27<br />

Vilela, Maria Marluce F.99<br />

Villaggio, Barbara Sa.112<br />

Villanueva, Cleva Sa.110<br />

Vladau, Costin F.124, OR.42, OR.47, 1201<br />

Vliag<strong>of</strong>tis, Harissios Sa.7<br />

Vollmer, Timothy Su.9<br />

von Gynz Rekowski, Kathrin Su.21<br />

von Herrath, Matthias OR.14, F.2, 2201, F.7, F.10,<br />

F.16, Sa.137, Su.117<br />

Vose, Julie Sa.102, Sa.103<br />

Voskuhl, Rhonda R. Su.112


FOCIS 2007 Abstract Index by Author<br />

Vrabelova, Zuzana F.19<br />

Vugler, Alex Su.86<br />

Vyse, Timothy J. OR.80, Sa.74<br />

Wade, Michael OR.7<br />

Wadia, Persis P. OR.53<br />

Wagner, Christ<strong>of</strong> F.49<br />

Wagner, David OR.67, F.25, Sa.131,<br />

Su.118<br />

Waid, Dan Su.118<br />

Wakeland, Edward OR.81<br />

Waldmann, Herman F.13<br />

Waliszewska, Alicja Sa.139<br />

Walker, Mindi OR.76<br />

Walker, Neil OR.85<br />

Walsh, Emily Sa.74<br />

Wandstrat, Amy OR.81<br />

Wang, Fang Sa.17<br />

Wang, James Sa.94<br />

Wang, Julie OR.75, Sa.140<br />

Wang, Kevin YehSheng F.87<br />

Wang, Shen-Wu F.137<br />

Wang, Wei Su.121<br />

Wang, Zhiliang Su.123<br />

Wang, Zhimo F.50<br />

Warnatz, Klaus OR.32, F.75, F.89<br />

Wasserfall, Clive OR.2, OR.56<br />

Watanabe, Rei Su.68<br />

Watanabe, Yoko Su.45<br />

Watry, Debbie Su.128<br />

Wawrousek, Eric OR.94<br />

Weaver, Casey 3203<br />

Webber, Steve Su.99<br />

Webster, John Sa.143<br />

Weeraratna, Ashani Sa.115<br />

Wei, Bo OR.100, Su.89<br />

Wei, Nathan Sa.70<br />

Weidanz, Jon A. OR.54, Sa.106, Sa.108<br />

Weiner, Howard Su.27, Su.28<br />

Weiner, Mira Su.27<br />

Weisenburger, Dennis Sa.102<br />

Wekerle, Thomas Sa.11<br />

Wellcome Trust Case Contr OR.85<br />

Wen, Xiangshu OR.66, Sa.135<br />

Wentzensen, Andreas F.49<br />

Wernet, Dorothee F.135<br />

Westall, Frederick Su.1, Su.2<br />

Westerberg, Per-Anton F.122<br />

Westerfield, Susan OR.26<br />

Whartenby, Katharine Sa.145<br />

Whitehead, Terry Su.47<br />

Whiteside, Theresa L. OR.73<br />

Whiting, Chan C. OR.59, Sa.52<br />

Wicker, Linda F.11, F.27, OR.83<br />

Wight, Thomas Sa.128<br />

Wijayawardana, Sameera F.98<br />

Wijbrandts, Carla Sa.66, Sa.67<br />

Wilkinson, Julie F.129, Sa.2<br />

Williams, Jim Su.34<br />

Williams, John Su.107<br />

Williams-Weese, Courtney Sa.24<br />

Willis, Van OR.52<br />

Willison, LeAnna N. Sa.18<br />

Wishart, Neil Sa.32<br />

Witte, Alison Sa.137<br />

Wolf, Bryan J. Sa.43<br />

Wolslegel, Kristen Sa.149<br />

2201<br />

Wong, Maida Sa.80<br />

Wongsena, Wachanan Sa.111<br />

Wood, Allyson Sa.63<br />

Woodburn, Tito OR.54<br />

Wotring, Michael OR.49<br />

Wu, Jianfeng OR.37<br />

Wu, Michele F.53<br />

Wu, Paul OR.13<br />

Wucherpfennig, Kai Su.32<br />

Wulff, Heike Su.94<br />

Wullner, Danika Su.42<br />

Xavier, Ramnik Su.92<br />

Xi, Dong F.50, Su.43, Su.136<br />

Yaghoubi, Shahriar F.5<br />

Yagishita, Naoko Sa.56<br />

Yagita, Hideo F.33<br />

Yamamoto, Kazuhiko Sa.30<br />

Yamamoto, Nobuto OR.28, Sa.125<br />

Yamamura, Takashi OR.24, Su.12, Su.13,<br />

Su.14<br />

Yamasaki, Satoshi Sa.56<br />

Yan, Weiming F.50, Su.43, Su.136<br />

Yancopoulos, George F.124<br />

Yang, Jiajin Sa.136<br />

Yang, Jun-Qi OR.66<br />

Yang, Li OR.95<br />

Yang, Lihu Su.58<br />

Yang, Sherry Sa.115<br />

Yang, Wei F.3<br />

Yano, Masashi Sa.141<br />

Yao, Qingping Sa.94<br />

Yaremtchuk, Tatiana F.41<br />

Yasui, Teruhito F.58<br />

Ye, Shuang OR.75, Sa.140<br />

Yelensky, Roman Su.35<br />

Yener, Alper Sa.109<br />

Yeung, Joseph Sa.80<br />

Yin, Zhenyu OR.31, OR.74<br />

Yong, Pierre F.76<br />

Yoo, Tai June Sa.134<br />

Yoshiga, Yohei Sa.40<br />

Yoshimura, Akihiko F.58<br />

Young, Daniel OR.59, Sa.52<br />

Young, Debbie OR.87<br />

Youssef, Sawsan OR.69<br />

Yu, Liping OR.1, F.17<br />

Yue, Erica Su.42<br />

Z, Stephen Su.69<br />

Zabaleta-Lanz, Mercedes Sa.85<br />

Zabel, Brian A. OR.91<br />

Zamir, Ehud F.12<br />

Zang, Yun Sa.28<br />

Zarabi, Mohammad Su.62<br />

S205


S206 FOCIS 2007 Abstract Index by Author<br />

Zarkeshfard, Bahareh F.125<br />

Zeckcer, Israel Su.75<br />

Zehnder, Roland Su.7<br />

Zeier, Martin Su.100<br />

Zeiseniss, Peter Sa.66<br />

Zelazny, Adrian F.66<br />

Zeldin, Robert Sa.12<br />

Zeng, Defu Sa.87<br />

Zhang, Chunyan Sa.87<br />

Zhang, Dong F.3<br />

Zhang, Li F.12<br />

Zhang, Ping Su.136<br />

Zhang, Xiao Sa.66<br />

Zhang, Xiuli F.53<br />

Zhang, Xusheng OR.47, F.109, Sa.54<br />

Zhang, Yiqun F.21<br />

Zhang, Zili OR.84<br />

Zhao, Hong Sa.75<br />

Zhao, Lei Sa.153<br />

Zhao, Xiaoyan Sa.54<br />

Zheng, Chengjie OR.101<br />

Zheng, Song Guo OR.75, Sa.140<br />

Zheng, Xin Xiao F.3<br />

Zheng, Xiufen OR.42, OR.47, 1201<br />

Zheng, Yan OR.37<br />

Zheng, Yanan OR.59<br />

Zhou, Baohua OR.57<br />

Zhou, Bin Sa.134<br />

Zhou, Jing OR.63<br />

Zhou, Liang OR.72<br />

Zhou, Tong OR.47, Sa.75, Sa.84<br />

Zhou, Xiaochuan Sa.142<br />

Zhou, Yixuan Sa.134<br />

Zhu, Chuanlong F.51<br />

Zhu, Xiaoming F.55, OR.57<br />

Zidovetzki, Raphael OR.79<br />

Ziegler, Steven F. OR.72, Sa.13<br />

Zielinska, Ewa F.113<br />

Zollner, Ricardo F.99<br />

Zonneveld-Huijssoon, Evelien Sa.67<br />

Zucchiatti, Andrea F.70<br />

Zuniga, Luis OR.91, OR.69<br />

Zvaifler, Nathan Sa.70<br />

Zvetkova, Elissaveta Borissova Sa.108


<strong>Clinical</strong> <strong>Immunology</strong> (2007) 123, S207–S208<br />

Abstract Index by Category<br />

Diabetes and other autoimmune endocrine diseases<br />

2201, OR.1, OR.3, OR.6, OR.11, OR.14, OR.16, OR.17, OR.18, OR.21, OR.31, OR.49, OR.51, OR.59, OR.61, OR.67, OR.76,<br />

OR.78, OR.82, OR.83, OR.85, F.1, F.2, F.3, F.4, F.5, F.6, F.7, F.8, F.9, F.10, F.11, F.12, F.13, F.14, F.15, F.16, F.17, F.18, F.19,<br />

F.20, F.21, F.22, F.23, F.24, F.25, F.26, F.27, F.28, F.30, F.31, F.32, F.33, F.34<br />

Immunity and infection<br />

1202, 3203, OR.23, OR.30, OR.98, F.38, F.39, F.44, F.45, F.46, F.48, F.49, F.50, F.51, F.52, F.53, F.55, F.56, F.57, F.58, F.61,<br />

F.62, F.63; F.64, F.65, F.66, F.67, F.68, F.69, F.70, F.71, F.72, F.73<br />

Immunodeficiency: primary or acquired<br />

2201, 3201, OR.25, OR.26, OR.27, OR.28, OR.29, OR.32, OR.52, F.74, F.75, F.76, F.77, F.78, F.79, F.80, F.81, F.82, F.83, F.84,<br />

F.85, F.86, F.87, F.88, F.89, F.90, F.91, F.92, F.93, F.94, F.95, F.96, F.97, F.98, F.99, F.100, F.101, F.102, F.103<br />

Laboratory immunology<br />

OR.5, OR.42, F.104, F.105, F.108, F.109, F.110, F.111, F.113, F.114, F.115, F.116, F.120, F.121, F.122, F.124, F.125, F.126, F.127,<br />

F.128, F.129, F.130, F.131, F.132, F.133, F.134, F.135, F.136, F.137<br />

Allergy/Asthma<br />

OR.22, OR.57, OR.64, OR.65, OR.99, Sa.1, Sa.5, Sa.6, Sa.7, Sa.8, Sa.9, Sa.10, Sa.11, Sa.12, Sa.13, Sa.16, Sa.17, Sa.18, Sa.19<br />

Autoimmune rheumatologic diseases<br />

1201, OR.12, OR.44, OR.48, OR.50, OR.55, OR.63, OR.66, OR.75, OR. 80, OR.81, OR.89, OR.92, OR.93, Sa.20, Sa.21, Sa.22,<br />

Sa.23, Sa.24, Sa.25, Sa.26, Sa.27, Sa.28, Sa.29, Sa.30, Sa.31, Sa.32, Sa.33, Sa.35, Sa.36, Sa.38, Sa.40, Sa.41, Sa.42, Sa.43,<br />

Sa.44, Sa.45, Sa.46, Sa.47, Sa.48, Sa.49, Sa.50, Sa.51, Sa.52, Sa.53, Sa.54, Sa.55, Sa.56, Sa.57, Sa.58, Sa.59, Sa.61, Sa.62,<br />

Sa.63, Sa.64, Sa.65, Sa.66, Sa.67, Sa.68, Sa.69, Sa.70, Sa.71, Sa.72, Sa.73, Sa.74, Sa.75, Sa.76, Sa.78, Sa.79, Sa.80, Sa.81,<br />

Sa.82, Sa.84, Sa.85, Sa.86<br />

Bone marrow or stem cell transplantation<br />

OR.53, OR.68, Sa.87, Sa.88, Sa.89, Sa.90, Sa.91, Sa.92, Sa.93, Sa.94, Sa.95<br />

Immuno-oncology<br />

OR.9, OR.15, OR.43, OR.45, OR.47, OR.54, OR.73, OR.101, Sa.96, Sa.97, Sa.98, Sa.100, Sa.101, Sa.102, Sa.103, Sa.104,<br />

Sa.105, Sa.106, Sa.107, Sa.108, Sa.109, Sa.110, Sa.111, Sa.112, Sa.113, Sa.114, Sa.115, Sa.116, Sa.117, Sa.119, Sa.120,<br />

Sa.121, Sa.122, Sa.123, Sa.124, Sa.125, Su.83<br />

General autoimmunity<br />

OR.7, OR.40, OR.46, OR.58, OR.62, OR.72, OR.79, Sa.126, Sa.127, Sa.128, Sa.129, Sa.130, Sa.131, Sa.132, Sa.133, Sa.134,<br />

Sa.135, Sa.136, Sa.137, Sa.138, Sa.139, Sa.140, Sa.141, Sa.142, Sa.143, Sa.145, Sa.146, Sa.147, Sa.148, Sa.149, Sa.150,<br />

Sa.151, Sa.152, Sa.153, Sa.154, Sa.155<br />

doi:10.1016/j.clim.2007.03.542<br />

available at www.sciencedirect.com<br />

www.elsevier.com/locate/yclim


S208 Abstract Index by Category<br />

Autoimmune neurologic diseases<br />

3202, OR.10, OR.13, OR.19, OR.20, OR.24, OR.33, OR.34, OR.36, OR.41, OR.69, OR.77, OR.86, OR.87, OR.88, OR.90, OR.91,<br />

OR.94, OR.95, OR.96, Su. 1, Su. 2, Su. 3, Su. 5, Su. 6, Su. 7, Su. 8, Su. 9, Su. 10, Su.11, Su.12, Su.13, Su.14, Su.15, Su.16,<br />

Su.18, Su.19, Su.20, Su.21, Su.22, Su.23, Su.24, Su.26, Su.27, Su.28, Su.29, Su.31, Su.32, Su.34, Su.35, Su.36, Su.37, Su.38<br />

Cytokines/Chemokines<br />

OR.2, OR.4, OR.8, OR.37, OR.38, OR.84, Su.39, Su.40, Su.41, Su.42, Su.43, Su.44, Su.45, Su.46, Su.47, Su.48, Su.49, Su.50,<br />

Su.51, Su.52, Su.53, Su.54, Su.55, Su.56, Su.57, Su.58, Su.59, Su.60, Su.61, Su.62<br />

Immuno-dermatology<br />

OR.39, Su.63, Su.64, Su.65, Su.66, Su.67, Su.68, Su.69, Su.70, Su.72, Su.74, Su.75<br />

<strong>Immunology</strong> <strong>of</strong> the eye<br />

OR.35, Su.76, Su.78, Su.79, Su.81, Su.82, Su.83<br />

Inflammatory bowel disease<br />

OR.70, OR.100, Su.84, Su.85, Su.86, Su.87, Su.89, Su.90, Su.91, Su.92, Su.93<br />

Organ transplantation<br />

OR.56, OR.71, OR.74, OR.97, Su.94, Su.95, Su.96, Su.98, Su.99, Su.100, Su.101, Su.102, Su.103, Su.104, Su.105, Su.107,<br />

Su.108, Su.109<br />

Other<br />

Su.110, Su.111, Su.112, Su.113, Su.114, Su.116, Su.117, Su.118, Su.119, Su.120, Su.121, Su.122, Su.123, Su.124, Su.125,<br />

Su.126, Su.127, Su.128, Su.129, Su.132, Su.133, Su.134, Su.136, Su.137, Su.138<br />

Reproductive immunology<br />

OR.60, Su.139, Su.140

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!