US20070248580A1 - Identification and Isolation of Multipotent Cells From Non-Osteochondral Mesenchymal Tissue - Google Patents

Identification and Isolation of Multipotent Cells From Non-Osteochondral Mesenchymal Tissue Download PDF

Info

Publication number
US20070248580A1
US20070248580A1 US11/576,573 US57657305A US2007248580A1 US 20070248580 A1 US20070248580 A1 US 20070248580A1 US 57657305 A US57657305 A US 57657305A US 2007248580 A1 US2007248580 A1 US 2007248580A1
Authority
US
United States
Prior art keywords
cells
cell
multipotent adult
isolated
population
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/576,573
Inventor
Rosa Garcia Castro
Maria Fernadez Miguel
Mariano Garcia Arranz
Manuel Gonzalez De La Pena
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Universidad Autonoma de Madrid
Tigenix SA
Original Assignee
Universidad Autonoma de Madrid
Cellerix SA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=35457282&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20070248580(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Universidad Autonoma de Madrid, Cellerix SA filed Critical Universidad Autonoma de Madrid
Assigned to UNIVERSIDAD AUTONOMA DE MADRID reassignment UNIVERSIDAD AUTONOMA DE MADRID ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GARCIA ARRANZ, MARIANO, GARCIA OLMO, DAMIAN
Assigned to CELLERIX, S.L. reassignment CELLERIX, S.L. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GONZALEZ DE LA PENA, MANUEL ANGEL, FERNANDEZ MIGUEL, MARIA GEMA, GARCIA CASTRO, ROSA ANA
Publication of US20070248580A1 publication Critical patent/US20070248580A1/en
Assigned to TIGENIX, S.A.U reassignment TIGENIX, S.A.U CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: CELLERIX, S.A.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0607Non-embryonic pluripotent stem cells, e.g. MASC
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0618Cells of the nervous system
    • C12N5/0619Neurons
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0654Osteocytes, Osteoblasts, Odontocytes; Bones, Teeth
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0658Skeletal muscle cells, e.g. myocytes, myotubes, myoblasts
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0667Adipose-derived stem cells [ADSC]; Adipose stromal stem cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/13Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells
    • C12N2506/1346Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells from mesenchymal stem cells
    • C12N2506/1384Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells from mesenchymal stem cells from adipose-derived stem cells [ADSC], from adipose stromal stem cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells

Definitions

  • the present invention relates to isolated multipotent adult cells which are isolated from non-osteochondral mesenchymal tissue and are characterized by the presence and absence of a set of cell surface markers.
  • the invention also relates to a method for identifying and isolating a population of said cells, as well as to the applications thereof, for example, in the manufacture of a pharmaceutical composition for the repair and regeneration of tissues.
  • Stem cells show differential characteristics as they are able to sustain themselves and differentiate into one or more cell type. Although research into stem cells and their applications is still in its early stages, adult stem cells in bone marrow have been used in transplants for more than 30 years. Nevertheless, in recent years, stem cell technology has made large advances such that stem cells are currently considered as a promising source of tissue and organs, with an important therapeutic potential for repair and regeneration of tissues.
  • stem cells are an alternative therapy for several human diseases, particularly those in which there is a loss of functional cells, including chondral, bone and muscular lesions, neurodegenerative diseases, immunologic rejection, heart disease and skin disorders (see U.S. Pat. No. 5,811,094, U.S. Pat. No. 5,958,767, U.S. Pat. No. 6,328,960, U.S. Pat. No. 6,379,953, U.S. Pat. No. 6,497,875).
  • stem cells In addition to cell therapy applications, stem cells have potential applications in the research and development of new drugs.
  • the study of mechanisms implicated in the proliferation and differentiation of stem cells is of great value in the process of searching for and characterizing new genes involved in a wide range of biological processes, including cell development and differentiation and neoplastic processes (Phillips et al., 2000; Ramalho-Santos et al., 2002; Ivanova et al., 2002).
  • stem cell technology allows specialized cells to be generated and the development of cell models for human and animal diseases, in which the efficacy and toxicity of new active ingredients can be determined in the preclinical phase (U.S. Pat. No. 6,294,346).
  • An adult somatic stem cell is an undifferentiated cell which is found in differentiated tissue and which has the capacity to proliferate and differentiate into one or more cell types.
  • Adult stem cells are present in different adult tissue, their presence being extensively reported in bone marrow, blood, cornea, retina, brain, muscle, skeleton, dental pulp, gastrointestinal epithelium, liver and skin (Jiang et al., 2002).
  • adult stem cells can be used in an autologous setting, and as such, they are immunologically compatibles and their use does not raise any ethical concerns.
  • phenotype refers to observable characteristics of the cell, such as characteristic morphology, interactions with other cells and with the extracellular matrix, cell surface proteins (surface markers) and characteristic functions.
  • the ideal source of adult stem cells is one in which they can be obtained by an easy, non-invasive process and one that allows a sufficient number of cells to be isolated.
  • a source should provide stem cells that can be easily isolated from a living subject without significant risks and discomfort and the source should allow a high yield to be obtained with minimal contamination from other cell types, without excessive cost of isolation and culture.
  • multipotent adult cells can be obtained from skeletal muscle and other connective tissue of mammals (Young et al. 1993, Rogers et al. 1995). Multipotent cells have also been obtained from human lipoaspirated tissue (Zuk et al., 2001). Another example of multipotent cells isolated from adult connective tissue is the so-called Multipotent Adult Progenitor Cells (MAPC) obtained from bone marrow (Jiang et al., 2002).
  • MPC Multipotent Adult Progenitor Cells
  • phenotype characterization of stem cells comprises determination of markers such as cell surface receptors, among others; and the determination of their capacity for differentiation in in vitro cultures.
  • markers such as cell surface receptors, among others; and the determination of their capacity for differentiation in in vitro cultures.
  • Each cell type has a certain combination of surface markers, that is, it has a certain profile of expression that characterizes that particular cell type, distinguishing it from others.
  • CD44 hyaluronic acid receptor
  • a significant disadvantage in using multipotent adult cells resides in the fact that most of the current sources for obtaining multipotent adult cells are contaminated with other cell types, complicating the process of identification, isolation and characterization of the populations of multipotent adult cells with the objective of using them for therapeutic or other ends. Thus, there is an interest in obtaining a population of multipotent adult cells isolated in a substantially pure form.
  • the characterization of a multipotent adult cell population from non-osteochondral mesenchymal tissue will allow a method for identification and isolation to be designed, as well as the identification of growth factors associated with self-regeneration. Moreover, there may be growth factors associated with the initial phases of differentiation, knowledge of which would allow more efficient in vivo and ex vivo differentiation, as well as for exercising control over the proliferation of stem cells.
  • the present invention provides a multipotent adult cell population from non-osteochondral mesenchymal tissue, preferable from adipose tissue, isolated and characterized by means of immunophenotype markers present on the cell surface, showing their multipotent nature.
  • the present invention provides a method for the identification and isolation of a population of multipotent adult cells from non-osteochondral mesenchymal tissue, dependent on a pattern of characteristic immunophenotype markers, allowing a composition of substantially homogeneous multipotent stem cell markers to be obtained.
  • the invention relates to an isolated multipotent adult cell, hereinafter referred to as the multipotent adult cell of the invention, which (a) is isolated from non-osteochondral mesenchymal tissue; (b) expresses CD9 ⁇ +>, CD10 ⁇ +>, CD13 ⁇ +>, CD29 ⁇ +>, CD44 ⁇ +>, CD49a ⁇ +>, CD51 ⁇ +>, CD54 ⁇ +>, CD55 ⁇ +>, CD58 ⁇ +>, CD59 ⁇ +>, CD90 ⁇ +> and CD105 ⁇ +>; and (c) lacks expression of CD11b, CD14, CD15, CD16, CD31, CD34, CD45, CD49f, CD102, CD104, CD106 and CD133.
  • the multipotent adult cells of the invention present the capacity to proliferate and to be differentiated into different cell lineages.
  • the multipotent adult cells of the invention can be differentiated into bone phenotype cells, muscle phenotype cells and neuronal phenotype cells.
  • the invention relates to an isolated cell population that comprises, or consists of, the multipotent adult cells of the invention.
  • said cell population is nearly, i.e., substantially, homogeneous.
  • the invention in another aspect, relates to a substantially homogeneous cell composition which comprises a multipotent adult cell of the invention or a cell population of multipotent adult cells of the invention.
  • the invention relates to a cell that expresses at least one characteristic of a specialized cell, wherein the cell is derived from an isolated multipotent adult cell of the invention.
  • the invention relates to a cell that expresses at least one characteristic of a specialized cell, wherein the at least one characteristic is that of a cell selected from the group consisting of an epithelial cell, an endothelial cell, an adipocyte, a myocyte, a chondrocyte, an osteocyte, a neuron, an astrocyte, an oligodendrocyte, a hepatocyte, a cardiomyocyte, and a pancreatic cell.
  • An isolated cell population that comprises said cells that express at least one characteristic of a specialized cell, wherein the cells are derived from isolated multipotent adult cells of the invention, constitutes a further aspect of the invention.
  • the invention relates to a method for obtaining the isolated multipotent adult cells of the invention which comprises:
  • the cells obtained and isolated according to said method contain the characteristics of the multipotent adult cells of the invention, i.e., they (a) have been isolated from non-osteochondral mesenchymal tissue; (b) are positive to the following markers: CD9, CD10, CD13, CD29, CD44, CD49A, CD51, CD54, CD55, CD58, CD59, CD90 and CD105; and (c) lack expression of the following markers: CD11b, CD14, CD15, CD16, CD31, CD34, CD45, CD49f, CD102, CD104, CD106 and CD133.
  • said cells present the capacity to proliferate and to be differentiated into different cell lineages.
  • the non-osteochondral mesenchymal tissue is a connective tissue, preferably adipose tissue.
  • the multipotent adult cells of the invention can be genetically modified.
  • the invention relates to a method for identifying a population of multipotent adult cells, wherein said population comprises, or consists of, isolated multipotent adult cells of the invention, the method comprising:
  • said specific binding compound is an antibody.
  • the present invention relates to a method for isolating a population of multipotent adult cells of the invention, which comprises:
  • the presence or absence of said surface markers characterizes said cells, thus, being characteristic for said cell population.
  • Cells that have the profile of expression of characteristic markers of the multipotent adult cells of the invention are finally selected.
  • said method of isolation consists of performing a negative selection, whereby cells that show binding to labelled compounds that bind specifically to a marker selected from the group consisting of CD11b, CD14, CD15, CD16, CD31, CD34, CD45, CD49f, CD102, CD104, CD106 and CD133 are excluded, and a subsequent positive selection, whereby cells that bind to labelled compounds that bind specifically to a marker selected from the group consisting of CD9, CD10, CD13, CD29, CD44, CD49a, CD51, CD54, CD55, CD58, CD59, CD90 and CD105 are selected.
  • the labelled compound of specific binding is an antibody.
  • the invention relates to a multipotent adult cell of the invention, or a population of multipotent adult cells of the invention, for therapeutic use, e.g., for use as a medicament.
  • the invention relates to a multipotent adult cell of the invention, or to a population of multipotent adult cells of the invention for use in the repair and regeneration of tissues.
  • the invention relates to a pharmaceutical composition that comprises a multipotent adult cell of the invention, or a population of multipotent adult cells of the invention, and a pharmaceutically acceptable carrier.
  • said pharmaceutical composition is useful for the repair and regeneration of tissues.
  • the invention relates to the use of a multipotent adult cell of the invention, or a cell population of multipotent adult cells of the invention, for the manufacture of a pharmaceutical composition for the repair and regeneration of tissues.
  • the invention relates to a therapeutic method comprising administering said pharmaceutical composition to a patient in need thereof.
  • said therapeutic method is for tissue repair or regeneration.
  • the invention relates to a method for assessing in vitro or in vivo cell response to a biological or pharmacological agent, or to a combinatorial library of said agents, which comprises:
  • the population of multipotent adult cells of the invention of step (a) is isolated from an individual or from a statistically significant population thereof.
  • the cells of the cell population of multipotent adult cells of the invention of step (a) are nearly, i.e., substantially, homogeneous.
  • the cells prior to step (c), the cells are allowed to differentiate into a specific type of cells.
  • FIGS. 1 a - 1 d show histograms of fluorescence immunocytometry corresponding to the profile of surface markers obtained from cells isolated from liposuction samples of a healthy donor. The results show the evolution over time of the markers studied in the cell cultures, indicating in each case the particular time period to which the analyzed cells belong.
  • FIG. 1 a shows the expression of markers on Day 0.
  • FIG. 1 b shows the expression of markers on Day 7 of culturing.
  • FIG. 1 c shows the expression of markers after 4 weeks of culturing and
  • FIG. 1 d shows the expression of markers after 3 months of culturing.
  • FIGS. 2 a - 2 d show the histograms of fluorescence immunocytometry corresponding to the profile of surface markers obtained from cells isolated from liposuction samples from a second healthy donor. The results show the evolution over time of the markers studied in the cell cultures, indicating in each case the particular time period to which the analyzed cells belong.
  • FIG. 2 a shows the expression of markers on Day 0.
  • FIG. 2 b shows the expression of markers on Day 7 of culturing.
  • FIG. 2 c shows the expression of markers after 4 weeks of culturing and
  • FIG. 2 d shows the expression of markers after 3 months of culturing.
  • FIGS. 3 a - 3 d show the histograms of fluorescence immunocytometry corresponding to the profile of surface markers obtained from cells isolated from liposuction samples from a third healthy donor. The results show the evolution over time of the markers studied in the cell cultures, indicating in each case the particular time period to which the analyzed cells belong.
  • FIG. 3 a shows the expression of markers on Day 0.
  • FIG. 3 b shows the expression of markers on Day 7 of culturing.
  • FIG. 3 c shows the expression of markers after 4 weeks of culturing and
  • FIG. 3 d shows the expression of markers after 3 months of culturing.
  • FIGS. 4 a - 4 d show microphotographs of cells incubated in osteogenic medium for 3 weeks.
  • FIG. 4 a shows mesenchymal stem cells from human bone marrow (positive control).
  • FIG. 4 b shows the cells of the invention, multipotent adult cells from non-osteochondral mesenchymal tissue, incubated in osteogenic medium for the first week.
  • FIG. 4 c shows the multipotent adult cells of the invention, incubated in osteogenic medium for the second week.
  • FIG. 4 d shows the multipotent adult cells of the invention, incubated in osteogenic medium during the third week.
  • the characterization of multipotent adult cells from non-osteochondral mesenchymal tissue by means of determining their immunophenotype profile allows to define said population in terms of the presence or absence of a certain set of surface markers. These markers are epitopes that can be identified using specific antibodies, constituting a valuable tool that allows to identify the population, as well as to design a strategy for isolation or purification thereof.
  • the isolated cells were subjected to differentiation assays with the objective of showing their multipotent nature.
  • the isolated and characterized cells were induced to in vitro differentiate into cells that express at least one characteristic of a specialized cell.
  • the methods that can be used to induce differentiation of the multipotent adult cells of the present invention into different specific cell types are known by those skilled in the art and some of them are explained in detail in Examples 2, 3 and 4, which show in vitro differentiation of the multipotent adult cells of the invention into bone phenotype cells, muscle phenotype cells and neuronal phenotype cells, respectively.
  • the invention relates to an isolated multipotent adult cell, hereinafter referred to as the multipotent adult cell of the invention, which (a) is isolated from non-osteochondral mesenchymal tissue; (b) expresses CD9 ⁇ +>, CD10 ⁇ +>, CD13 ⁇ +>, CD29 ⁇ +>, CD44 ⁇ +>, CD49a ⁇ +>, CD51 ⁇ +>, CD54 ⁇ +>, CD55 ⁇ +>, CD58 ⁇ +>, CD59 ⁇ +>, CD90 ⁇ +> and CD105 ⁇ +>; and (c) lacks expression of CD11b, CD14, CD15, CD16, CD31, CD34, CD45, CD49f, CD102, CD104, CD106 and CD133.
  • An isolated cell population that comprises, or consists of, the multipotent adult cells of the invention constitutes a further aspect of the present invention.
  • said cell population is nearly, i.e., substantially, homogeneous.
  • the multipotent adult cells of the invention are isolated from non-osteochondral mesenchymal tissue.
  • non-osteochondral mesenchymal tissue refers to a mesenchymal tissue other than cartilage and bone marrow.
  • Non-osteochondral mesodermal tissues are also referred in this description to as “soft tissues”.
  • Illustrative, non-limitative, examples of non-osteochondral mesodermal tissues include skin, fat and muscle tissue.
  • the multipotent adult cells of the invention are isolated from sub-dermal adipose tissue.
  • the multipotent adult cells of the invention can be obtained from any suitable source of non-osteochondral mesenchymal tissue from any suitable animal, including human beings. In general, said cells are obtained from non-pathological post-natal mammalian non-osteochondral mesenchymal tissues. In a preferred embodiment, the multipotent adult cells of the invention are obtained from a source of non-osteochondral mesenchymal tissue, such as sub-dermal adipose tissue. Also, in a particular embodiment, the multipotent adult cells of the invention are isolated from a mammal, e.g., a rodent, primate, etc., preferably, from a human being.
  • a mammal e.g., a rodent, primate, etc.
  • the multipotent adult cells of the invention are also characterized by the presence and absence of a set of markers, namely, said cells are characterized in that they (i) are positive for some markers [CD9 ⁇ +>, CD10 ⁇ +>, CD13 ⁇ +>, CD29 ⁇ +>, CD44 ⁇ +>, CD49a ⁇ +>, CD51 ⁇ +>, CD54 ⁇ +>, CD55 ⁇ +>, CD58 ⁇ +>, CD59 ⁇ +>, CD90 ⁇ +> and CD105 ⁇ +>], and (ii) are negative for some markers [CD11b, CD14, CD15, CD16, CD31, CD34, CD45, CD49f, CD102, CD104, CD106 and CD133].
  • the phenotype characterization of the multipotent adult cells of the invention, or of a cell population comprising said cells, by surface markers can be conventionally performed either by individual staining of the cells (flow cytometry) or by making histological cuts of the population in situ, in accordance with conventional methods.
  • the expression of said surface markers on the multipotent adult cells of the invention may be monitored by flow cytometry.
  • the characterization of the multipotent adult cells of the invention, or a cell population comprising, or consisting of, said cells, by their immuno-phenotype profile can be used to define said cells or cell population in terms of the presence or absence of a certain set of surface markers.
  • These markers are epitopes that can be identified with specific antibodies, constituting a valuable tool that allows to identify the population, as well as to design a strategy for isolation or purification thereof.
  • Monoclonal antibodies against said surface markers can be used to identify the multipotent adult cells of the invention.
  • the determination of the profile of surface markers by antibodies may be direct, using a labelled antibody, or indirect, using a second labelled antibody against the primary specific antibody of the cell marker, thus achieving signal amplification.
  • the presence or absence of binding of the antibody may be determined by different methods that include but are not limited to immunofluorescence microscopy and radiography.
  • specific reagent refers to a member of a specific binding pair; members of a specific binding pair, include but are not limited to, binding pairs of antigens and antibodies, pairs comprising MHC antigens and T-cell receptors, complementary nucleotide sequences, as well as pairs of peptide ligands and their receptor.
  • the specific binding pairs include analogues, fragments and derivatives of the specific member of the binding pair.
  • antibodies as reagents with affinity is of particular interest.
  • the production of specific monoclonal antibodies will be evident to any ordinary skilled person in the art.
  • the antibodies are labelled.
  • markers that are used include but are not limited to: magnetic particles, biotin and fluorochromes that will allow identification or separation of the cell type to which the antibody has bound.
  • the analysis of the cell population comprising the multipotent adult cells of the invention by flow cytometry allows different antibodies labelled with fluorochromes that emit at different wavelengths to be used in the same sample.
  • the separation of the populations that present the phenotype of interest can be carried out by affinity separation techniques, which include magnetic separation (using magnetic particles coated with specific antibodies), affinity chromatography, cytotoxic agents bound to monoclonal antibodies or used along with monoclonal antibodies and panning with the antibody attached to a solid support, as well as by other techniques that are appropriate.
  • affinity separation techniques include magnetic separation (using magnetic particles coated with specific antibodies), affinity chromatography, cytotoxic agents bound to monoclonal antibodies or used along with monoclonal antibodies and panning with the antibody attached to a solid support, as well as by other techniques that are appropriate.
  • a more precise separation would be obtained by flow cytometry, a technique that allows the separation of cell populations according to the intensity of staining, along with other parameters such as cell size and cell complexity.
  • the multipotent adult cells of the invention present the capacity to proliferate and to be differentiated into different cell lineages.
  • Illustrative, non-limitative examples of cell lineages in which the multipotent adult cells of the invention can be differentiated include, e.g., bone phenotype cells, muscle phenotype cells and neuronal phenotype cells.
  • the multipotent adult cells of the invention can proliferate and differentiate into cells of other lineages by conventional methods. Methods of identifying and subsequently isolating differentiated cells from their undifferentiated counterparts can be also carried out by methods well known in the art.
  • the multipotent adult cells of the invention are also capable of being expanded ex vivo. That is, after isolation, the multipotent adult cells of the invention can be maintained and allowed to proliferate ex vivo in culture medium.
  • culture medium comprises, for example, Dulbecco's Modified Eagle's Medium (DMEM), antibiotics, and glutamine, and it is usually supplemented with 2-20% fetal bovine serum (FBS). It is within the skill of one in the art to modify or modulate concentrations of media and/or media supplements as necessary for the cells used. Sera often contain cellular factors and components that are necessary for viability and expansion.
  • sera examples include FBS, bovine serum (BS), calf serum (CS), fetal calf serum (FCS), newborn calf serum (NCS), goat serum (GS), horse serum (HS), porcine serum, sheep serum, rabbit serum, rat serum (RS), etc.
  • BS bovine serum
  • CS calf serum
  • FCS fetal calf serum
  • NCS newborn calf serum
  • GS goat serum
  • HS horse serum
  • porcine serum sheep serum
  • rabbit serum rat serum
  • RS rat serum
  • the multipotent adult cells of the invention are of human origin, supplementation of cell culture medium with a human serum, preferably of autologous origin. It is understood that sera can be heat-inactivated at 55-65° C. if deemed necessary to inactivate components of the complement cascade. Modulation of serum concentrations, withdrawal of serum from the culture medium can also be used to promote survival of one or more desired cell types.
  • multipotent adult cells of the invention will benefit from FBS concentrations of about 2% to about 25%.
  • the multipotent adult cells of the invention can be expanded in a culture medium of definite composition, in which the serum is replaced by a combination of serum albumin, serum transferrin, selenium, and recombinant proteins including but not limited to: insulin, platelet-derived growth factor (PDGF), and basic fibroblast growth factor (bFGF).
  • PDGF platelet-derived growth factor
  • bFGF basic fibroblast growth factor
  • amino acids include, but are not limited to, L-alanine, L-arginine, L-aspartic acid, L-asparagine, L-cysteine, L-cystine, L-glutamic acid, L-glutamine, L-glycine, and the like.
  • Antimicrobial agents are also typically used in cell culture to mitigate bacterial, mycoplasmal, and fungal contamination.
  • antibiotics or anti-mycotic compounds used are mixtures of penicillin/streptomycin, but can also include, but are not limited to amphotericin (Fungizone®), ampicillin, gentamicin, bleomycin, hygromacin, kanamycin, mitomycin, etc.
  • Hormones can also be advantageously used in cell culture and include, but are not limited to, D-aldosterone, diethylstilbestrol (DES), dexamethasone, b-estradiol, hydrocortisone, insulin, prolactin, progesterone, somatostatin/human growth hormone (HGH), etc.
  • DES diethylstilbestrol
  • HGH somatostatin/human growth hormone
  • the maintenance conditions of the multipotent adult cells of the invention can also contain cellular factors that allow cells to remain in an undifferentiated form. It is apparent to those skilled in the art that prior to differentiation supplements that inhibit cell differentiation must be removed from the culture medium. It is also apparent that not all cells will require these factors. In fact, these factors may elicit unwanted effects, depending on the cell type.
  • the multipotent adult cells of the invention can be transfected or genetically engineered to express, at least, one polypeptide of interest.
  • the polypeptide of interest is a product capable of inducing or increasing the expression of genes involved in the repair or regeneration of a tissue.
  • the invention relates to a method for obtaining the isolated multipotent adult cells of the invention which comprises:
  • the cells obtained according to said method contain the characteristics of the multipotent adult cells of the invention.
  • solid surface refers to any material that allow the multipotent adult cells of the invention to adhere.
  • said material is a plastic material treated to promote the adhesion of mammalian cells to its surface.
  • Steps a)-d) can be carried out by conventional techniques known by those skilled in the art.
  • the multipotent adult cells of the invention can be obtained by conventional means from any suitable source of non-osteochondral mesenchymal tissue from any suitable animal, including human beings, e.g., from human adipose tissue.
  • the animal can be alive or dead, so long as non-osteochondral mesenchymal tissue cells within the animal are viable.
  • human adipose cells are obtained from living donors, using well-recognized protocols such as surgical or suction lipectomy.
  • liposuction effluent is a particularly preferred source from which the cells of the invention can be derived.
  • the multipotent adult cells of the invention are from human sub-dermal adipose tissue obtained by liposuction.
  • the sample of non-osteochondral mesenchymal tissue is, preferably, washed before being processed.
  • the sample of non-osteochondral mesenchymal tissue is washed with physiologically-compatible saline solution (e.g., phosphate buffered saline (PBS)) and then vigorously agitated and left to settle, a step that removes loose matter (e.g., damaged tissue, blood, erythrocytes, etc) from the tissue.
  • physiologically-compatible saline solution e.g., phosphate buffered saline (PBS)
  • PBS phosphate buffered saline
  • the washing and settling steps generally are repeated until the supernatant is relatively clear of debris.
  • the remaining cells generally will be present in lumps of various sizes, and the protocol proceeds using steps gauged to degrade the gross structure while minimizing damage to the cells themselves.
  • One method of achieving this end is to treat the washed lumps of cells with an enzyme that weakens or destroys bonds between cells (e.g., collagenase, dispase, trypsin, etc.).
  • an enzyme that weakens or destroys bonds between cells e.g., collagenase, dispase, trypsin, etc.
  • the amount and duration of such enzymatic treatment will vary, depending on the conditions employed, but the use of such enzymes is generally known in the art.
  • the lumps of cells can be degraded using other treatments, such as mechanical agitation, sonic energy, thermal energy, etc. If degradation is accomplished by enzymatic methods, it is desirable to neutralize the enzyme following a suitable period, to minimize deleterious effects on the cells.
  • the degradation step typically produces a slurry or suspension of aggregated cells and a fluid fraction containing generally free stromal cells (e.g., red blood cells, smooth muscle cells, endothelial cells, fibroblast cells, and stem cells).
  • stromal cells e.g., red blood cells, smooth muscle cells, endothelial cells, fibroblast cells, and stem cells.
  • the next stage in the separation process is to separate the aggregated cells from the cells of the invention. This can be accomplished by centrifugation, which forces the cells into a pellet covered by a supernatant. The supernatant then can be discarded and the pellet suspended in a physiologically-compatible fluid.
  • the suspended cells typically include erythrocytes, and in most protocols it is desirable to lyse them.
  • erythrocytes Methods for selectively lysing erythrocytes are known in the art, and any suitable protocol can be employed (e.g., incubation in a hyper-or hypotonic medium, by lysis using ammonium chloride, etc.). Of course, if the erythrocytes are lysed, the remaining cells should then be separated from the lysate, for example by filtration, sedimentation, or density fractionation.
  • the suspended cells can be washed, re-centrifuged, and resuspended one or more successive times to achieve greater purity.
  • the cells can be separated on the basis of cells surface markers profile or on the basis of cell size and granularity.
  • the cells can be cultured and, if desired, assayed for number and viability to assess the yield.
  • the cells will be cultured without differentiation, on a solid surface, using a suitable cell culture media, at the appropriate cell densities and culture conditions.
  • a suitable cell culture medium e.g., DMEM, typically supplemented with 5-15% (e.g., 10%) of a suitable serum, such as fetal bovine serum or human serum
  • cells are washed in order to remove non-adhered cells and cell fragments.
  • the cells are maintained in culture in the same medium and under the same conditions until they reach the adequate confluence, typically, about 80% cell confluence, with replacement of the cell culture medium when necessary.
  • the cells can be expanded by means of consecutive passages using a detachment agent such as trypsin and seeding onto a bigger cell culture surface at the appropriate cell density (usually 2,000-10,000 cells/cm 2 ).
  • the cells can be passaged some times without losing their developmental phenotype.
  • the cells are plated at a desired density such as between about 100 cells/cm 2 to about 100,000 cells/cm 2 (such as about 500 cells/cm 2 to about 50,000 cells/cm 2 , or, more particularly, between about 1,000 cells/cm 2 to about 20,000 cells/cm 2 ). If plated at lower densities (e.g., about 300 cells/cm 2 ), the cells can be more easily clonally isolated. For example, after a few days, cells plated at such densities will proliferate into an homogeneous population. In a particular embodiment, the cell density is between 2,000-10,000 cells/cm 2 .
  • Example 1 describes in a detailed manner the isolation of multipotent adult cells of the invention from human sub-dermal adipose tissue.
  • Cell-surface markers can be identified by any suitable conventional technique, usually based on a positive/negative selection; for example, monoclonal antibodies against cell-surface markers, which presence/absence in the cells has to be confirmed, can be used; although other techniques can also be used.
  • monoclonal antibodies against CD9, CD10, CD13, CD29, CD44, CD49a, CD51, CD54, CD55, CD58, CD59, CD90 and CD105 are used in order to confirm the absence of said markers in the selected cells; and monoclonal antibodies against CD11b, CD14, CD15, CD16, CD31, CD34, CD45, CD49f, CD102, CD104, CD106 and CD133 are used in order to confirm the absence of said markers in the selected cells.
  • Said monoclonal antibodies are known or can be obtained by a skilled person in the art by conventional methods.
  • the capacity of the selected cells to differentiate into different cell lineages can be assayed by conventional methods as previously disclosed.
  • the multipotent adult cells of the invention and cell populations provided by the instant invention can be clonally expanded, if desired, using a suitable method for cloning cell populations.
  • a proliferated population of cells can be physically picked and seeded into a separate plate (or the well of a multi-well plate).
  • the cells can be subcloned onto a multi-well plate at a statistical ratio for facilitating placing a single cell into each well (e.g., from about 0.1 to about 1 cell/well or even about 0.25 to about 0.5 cells/well, such as 0.5 cells/well).
  • the cells can be cloned by plating them at low density (e.g., in a Petri dish or other suitable substrate) and isolating them from other cells using devices such as a cloning rings.
  • the production of a clonal population can be expanded in any suitable culture medium.
  • the isolated cells can be cultured to a suitable point when their developmental phenotype can be assessed.
  • any of the steps and procedures for isolating the cells of the cell population of the invention can be performed manually, if desired.
  • the process for isolating such cells can be facilitated through a suitable device, many of which are known in the art.
  • the invention relates to a method for identifying a population of multipotent adult cells, wherein said population comprises, or consists of, multipotent adult cells of the invention, the method comprising:
  • said specific binding compound is an antibody.
  • the present invention relates to a method for isolating a population of multipotent adult cells of the invention, which comprises:
  • the presence or absence of said surface markers characterizes said cells, thus, being characteristic for said cell population.
  • Cells that have the profile of expression of characteristic markers of the multipotent adult cells of the invention are finally selected.
  • said method of isolation consists of performing a negative selection, whereby cells are excluded that show binding to labelled compounds that bind specifically to a marker selected from the group consisting of CD11b, CD14, CD15, CD16, CD31, CD34, CD45, CD49f, CD102, CD104, CD106 and CD133, and a subsequent positive selection, whereby cells are selected that bind to labelled compounds that bind specifically to a marker selected from the group consisting of CD9, CD10, CD13, CD29, CD44, CD49a, CD51, CD54, CD55, CD58, CD59, CD90 and CD105.
  • the labelled compound of specific binding is an antibody.
  • This method can be carried out as previously mentioned in connection with the method for obtaining the multipotent adult cells of the invention.
  • the multipotent adult cells of the invention can be found in a cell composition. Therefore, in another aspect, the invention relates to a substantially homogeneous cell composition which comprises a multipotent adult cell of the invention or a cell population of multipotent adult cells of the invention.
  • the multipotent adult cells of the invention can be used for repairing and regenerating tissues.
  • the invention relates to a multipotent adult cell of the invention, or a population of multipotent adult cells of the invention, for therapeutic use, e.g., for use as a medicament.
  • the invention relates to a multipotent adult cell of the invention, or to a population of multipotent adult cells of the invention for use in the repair and regeneration of tissues.
  • the invention in another aspect, relates to a pharmaceutical composition that comprises a multipotent adult cell of the invention, or a population of multipotent adult cells of the invention, and a pharmaceutically acceptable carrier.
  • said pharmaceutical composition is useful for the repair and regeneration of tissues.
  • the pharmaceutical composition of the invention comprises a prophylactically or therapeutically effective amount of a multipotent adult cell of the invention, or a cell population of multipotent adult cells of the invention, and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia, or European Pharmacopeia, or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic agent is administered.
  • the composition if desired, can also contain minor amounts of pH buffering agents.
  • compositions will contain a prophylactically or therapeutically effective amount of a multipotent adult cell of the invention, or a cell population of multipotent adult cells of the invention preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the subject.
  • the formulation should suit the mode of administration.
  • the pharmaceutical compositions are sterile and in suitable form for administration to a subject, preferably an animal subject, more preferably a mammalian subject, and most preferably a human subject.
  • the pharmaceutical composition of the invention may be in a variety of forms. These include, for example, solid, semi-solid, and liquid dosage forms, such as lyophilized preparations, liquids solutions or suspensions, injectable and infusible solutions, etc.
  • the preferred form depends on the intended mode of administration and therapeutic application.
  • the administration of the cells or cell population of the invention, or the pharmaceutical composition comprising same, to the subject in need thereof can be carried out by conventional means.
  • said cells or cell population is administered to the subject by a method which involves transferring the cells to the desired tissue, either in vitro or in vivo, to the animal tissue directly.
  • the cells can be transferred to the desired tissue by any appropriate method, which generally will vary according to the tissue type. For example, cells can be seeded onto the desired site within the tissue to establish a population, etc. Cells can be transferred to sites in vivo using devices such as catherters, trocars, cannulae, stents (which can be seeded with the cells), etc.
  • the pharmaceutical composition of the invention can be used in a combination therapy.
  • the combination therapy is administered to a subject in need of treatment, such as a patient in need of repair or regeneration of a tissue.
  • the combination therapy is used in conjunction with other types of treatments to repair or regenerate tissues.
  • the combination therapies of the invention can be used prior to, concurrently or subsequent to the administration of the multipotent adult cells of the invention.
  • the invention relates to the use of a multipotent adult cell of the invention, or a cell population of multipotent adult cells of the invention, for the manufacture of a pharmaceutical composition for the repair and regeneration of tissues.
  • the invention further relates, in another aspect, to a therapeutic method comprising administering said pharmaceutical composition to a patient in need thereof.
  • said therapeutic method is for tissue repair or regeneration.
  • the invention relates to a method for assessing in vitro or in vivo cell response to a biological or pharmacological agent, or to a combinatorial library of said agents, which comprises:
  • the population of multipotent adult cells of the invention of step (a) is isolated from an individual or from a statistically significant population thereof.
  • the cells of the cell population of multipotent adult cells of the invention of step (a) are nearly, i.e., substantially, homogeneous.
  • the cells prior to step (c), the cells are allowed to differentiate into a specific type of cells.
  • the invention relates to a cell that expresses at least one characteristic of a specialized cell, wherein the cell is derived from an isolated multipotent adult cell of the invention.
  • These cells are also multipotent adult cells having a differentiation stage more advanced than that of the multipotent adult cells of the invention.
  • the invention relates to a cell that expresses at least one characteristic of a specialized cell, wherein the at least one characteristic is that of a cell selected from the group consisting of an epithelial cell, an endothelial cell, an adipocyte, a myocyte, a chondrocyte, an osteocyte, a neuron, an astrocyte, an oligodendrocyte, a hepatocyte, a cardiomyocyte, and a pancreatic cell.
  • An isolated cell population that comprises said cells that express at least one characteristic of a specialized cell, wherein the cells are derived from the isolated multipotent adult cells of the invention, constitutes a further aspect of the invention.
  • the isolation of multipotent adult cells from soft tissue was performed by selecting those cells with a capacity for proliferation and differentiation, characterized in that they show adhesion to the plastic container of the cell culture. Then, the cells were characterized by monitoring by flow cytometry the expression of a series of surface markers on the recently isolated cells and during the course of the culture development in vitro.
  • the isolation of the multipotent adult cells was carried out from sub-dermal adipose tissue, obtained by liposuction from 3 healthy human donors (donors 1, 2 and 3).
  • the sample from the sub-dermal adipose tissue was washed with phosphate buffered saline solution (PBS).
  • PBS phosphate buffered saline solution
  • an enzymatic digestion was performed with type 11 collagenase in saline solution (5 mg/ml) at 37° C. for 30 minutes.
  • the collagenase was deactivated by adding an equivalent volume of DMEM medium, with 10% fetal bovine serum. This cell suspension was centrifuged at 250 g for 10 minutes to obtain a cell deposit.
  • NH 4 Cl was added at an end concentration of 0.16 M and the mixture was incubated for 10 minutes at room temperature to induce the lysis of the erythrocytes present.
  • the suspension was centrifuged at 250-400 g and resuspended in DMEM-10% FBS with 1% ampicillin-streptomycin. Finally, the cells were plated, inoculating 2 ⁇ 10 4 -3 ⁇ 10 4 cells per cm 2 .
  • the cells were cultured for 20-24 hours at 37° C., under an atmosphere with 5% CO 2 . After 24 hours, the culture was washed with PBS to remove the cells and the remains of the tissue in suspension.
  • the cells selected by adhesion to the plastic container were cultured in DMEM+10% fetal bovine serum (FBS).
  • the multipotent adult cells isolated were characterized from one of the donors, in function of the presence/absence of a series of surface markers. To do this, the expression of the following surface markers was monitored by flow cytometry:
  • the immunophenotype characterization of the cells was performed on recently isolated cells and also on day 7, after 4 weeks and after 3 months of culture, of the samples from the 3 healthy human donors. Taking into account that the selection is performed by adhesion to the plastic container of the cell culture, cells from the cell fraction adhered after less than 24 hours in the culture since isolation are considered as “recently isolated cells”.
  • the cells to be characterized were collected by means of gentle digestion with trypsin, washed with PBS and incubated for 30 minutes at 4° C. with fluorescein (FITC) or phycoerythrin (PE) labelled antibodies against each one of the cell surface markers to be analyzed.
  • the cell markers were washed and immediately analyzed using the Epics-XL cytometer (Coulter). As controls, cells stained with unspecific, FITC or PE labelled, antibodies of the corresponding isotypes were used.
  • FIGS. 1 a - 1 d, 2 a - 2 d and 3 a - 3 d show the histograms grouped by donor for a better visualization of the evolution of the markers studied during the culturing, indicating in each case what time in the culture period the analyzed cells belong.
  • the multipotent adult cells isolated from non-osteochondral mesenchymal tissue provided by the present invention is characterized by being positive for CD9 ⁇ +>, CD10 ⁇ +>, CD13 ⁇ +>, CD29 ⁇ +>, CD44 ⁇ +>, CD49A ⁇ +>, CD51 ⁇ +>, CD54 ⁇ +>, CD55 ⁇ +>, CD58 ⁇ +>, CD59 ⁇ +>, CD90 ⁇ +> and CD105 ⁇ +>; and for lacking expression of CD11b, CD14, CD15, CD16, CD31, CD34, CD45, CD49f, CD102, CD104, CD106 and CD133.
  • characterized human multipotent adult cells of the present invention were used.
  • the cells were isolated from the 3 samples of analyzed lipoaspirates, each corresponding to a healthy donor (Example 1).
  • the multipotent adult cells were isolated and characterized as mentioned in Example 1.
  • a sample of mesenchymal stem cells (MSC) of human bone marrow was used as positive control.
  • the isolated cells were seeded at a density of 10,000 cells/cm 2 onto 6-well plates (one plate per sample), and were incubated in standard culture medium (DMEM, 10% FBS, L-Glutamine 2 mM and antibiotic). After two days of culturing, the culture medium of one of the wells (control) is replaced with fresh medium, and the remaining wells with osteogenesis inducing medium, which contains the standard culture medium with the following added:
  • the cells were cultured for 3 weeks in normal conditions, changing the medium every 2-3 days. After three weeks, the presence of mineralized deposits of calcium phosphate can be seen, which indicates the presence of osseus nodules. These nodules are detected by staining with Alizarin red (Standford et al., 1995). Specifically, the medium was eliminated, the cells were washed twice with PBS and fixed with 70% cold ethanol for 30 minutes at room temperature. The fixed wells were then washed with PBS and stained with Alizarin red (40 mM, pH 4.1) for 10 minutes at room temperature. The stained cells were washed with abundant water, and the precipitates of calcium phosphate, which appear strongly stained red, were examined under the microscope.
  • Alizarin red Alizarin red
  • FIGS. 4 a - 4 d show microphotographs of the osteo-induced cells stained with Alizarin red. Although the formation of calcium phosphate is quicker in the sample corresponding to MSC from bone marrow which acts as a positive control ( FIG. 4 a ), in the 3 samples from the adipose tissue, the formation of large quantities of bone matrix can be discerned, although with differing intensity in each of the samples. All wells in which osteogenesis was induced showed the same behaviour and in the control wells (not submitted to osteogenic stimuli) the formation of bone matrix was not detected. No relationship was seen between the amount of bone matrix formed and the time that each sample was being cultured after isolation from the tissue (between 3 and 9 weeks).
  • characterized human multipotent adult cells of the present invention were used.
  • the cells were isolated from the 3 samples of analyzed lipoaspirates, each corresponding to a healthy donor (Example 1).
  • the multipotent adult cells were isolated and characterized as mentioned in Example 1.
  • a sample of MSC of human bone marrow was used as positive control.
  • the isolated cells were seeded at a density of 10,000 cells/cm 2 into standard culture medium (DMEM, 10% FBS, L-Glutamine 2 mM and antibiotic). After two days of culturing, the culture medium of one of the wells (control) was replaced with fresh medium, and the remaining wells with myogenesis inducing medium (Wakitani et al., 1995), which contains the standard culture medium with the following added:
  • the medium was replaced by standard culture medium, and the cells were cultured for 2-3 weeks, changing the medium every 2-3 days. After this time, the cells acquired an elongated phenotype, formed fibrillar structures and some cell fusions can be seen.
  • the cells obtained were fixed with paraformaldehyde (PFA) at 4% and incubated with an antibody against the heavy chain of myosin, which is the specific antigen for muscle. The results confirm the differentiation of the human multipotent adult cells of the invention into muscle phenotype cells.
  • PFA paraformaldehyde
  • characterized human multipotent adult cells of the present invention were used.
  • the cells were isolated from the 3 samples of analyzed lipoaspirates, each corresponding to a healthy donor (Example 1).
  • the multipotent adult cells were isolated and characterized as mentioned in Example 1.
  • a sample of MSC from human bone marrow was used as positive control.
  • the isolated cells were seeded at low density, 3 ⁇ 10 3 cells/cm 2 , into standard culture medium (DMEM, 10% FBS, L-Glutamine 2 mM and antibiotic), supplemented with 10 ng/ml bFGF and incubated for 24-36 hours to yield a large number of cells.
  • DMEM 10% FBS, L-Glutamine 2 mM and antibiotic
  • 10 ng/ml bFGF 10 ng/ml bFGF

Abstract

Identification and isolation of multipotent cells from non-osteochondral mesenchymal tissue. This invention relates to the identification and isolation of multipotent cells from non-osteochondral mesenchymal tissue. Specifically, it relates to an adult multipotent cell or a cell population or composition comprising said cell, isolated from non-osteochondral mesenchymal tissue, characterized in that it is positive for the following markers: CD9, CD10, CD13, CD29, CD44, CD49A, CD51, CD54, CD55, CD58, CD59, CD90 and CD105 and because it lacks expression of the following markers: CD11b, CD14, CD15, CD16, CD31, CD34, CD45, CD49f, CD102, CD104, CD106 and CD133.

Description

    FIELD OF THE INVENTION
  • The present invention relates to isolated multipotent adult cells which are isolated from non-osteochondral mesenchymal tissue and are characterized by the presence and absence of a set of cell surface markers. The invention also relates to a method for identifying and isolating a population of said cells, as well as to the applications thereof, for example, in the manufacture of a pharmaceutical composition for the repair and regeneration of tissues.
  • BACKGROUND OF THE INVENTION
  • Stem cells show differential characteristics as they are able to sustain themselves and differentiate into one or more cell type. Although research into stem cells and their applications is still in its early stages, adult stem cells in bone marrow have been used in transplants for more than 30 years. Nevertheless, in recent years, stem cell technology has made large advances such that stem cells are currently considered as a promising source of tissue and organs, with an important therapeutic potential for repair and regeneration of tissues.
  • The use of stem cells is an alternative therapy for several human diseases, particularly those in which there is a loss of functional cells, including chondral, bone and muscular lesions, neurodegenerative diseases, immunologic rejection, heart disease and skin disorders (see U.S. Pat. No. 5,811,094, U.S. Pat. No. 5,958,767, U.S. Pat. No. 6,328,960, U.S. Pat. No. 6,379,953, U.S. Pat. No. 6,497,875).
  • In addition to cell therapy applications, stem cells have potential applications in the research and development of new drugs. On the one hand, the study of mechanisms implicated in the proliferation and differentiation of stem cells is of great value in the process of searching for and characterizing new genes involved in a wide range of biological processes, including cell development and differentiation and neoplastic processes (Phillips et al., 2000; Ramalho-Santos et al., 2002; Ivanova et al., 2002). On the other hand, stem cell technology allows specialized cells to be generated and the development of cell models for human and animal diseases, in which the efficacy and toxicity of new active ingredients can be determined in the preclinical phase (U.S. Pat. No. 6,294,346).
  • An adult somatic stem cell is an undifferentiated cell which is found in differentiated tissue and which has the capacity to proliferate and differentiate into one or more cell types. Adult stem cells are present in different adult tissue, their presence being extensively reported in bone marrow, blood, cornea, retina, brain, muscle, skeleton, dental pulp, gastrointestinal epithelium, liver and skin (Jiang et al., 2002). By their nature, adult stem cells can be used in an autologous setting, and as such, they are immunologically compatibles and their use does not raise any ethical concerns.
  • An adult stem cell should be able to give rise to fully differentiated cells with mature phenotypes which are integrated into the tissue where they are found and which are able to carry out the specialized functions of the given tissue. The term “phenotype” refers to observable characteristics of the cell, such as characteristic morphology, interactions with other cells and with the extracellular matrix, cell surface proteins (surface markers) and characteristic functions.
  • Different populations of adult stem cells capable to contribute to the repair of different tissues have been described. Among these populations, those of mesodermic origin are of particular interest because they offer the theoretical possibility of regenerating a large number of clinically very relevant connective tissues such as bone, cartilage, tendons, skeletal muscle, heart muscle, vascular endothelium, sub-dermal fat and bone marrow stroma. The first cell population of this type isolated was the so-called mesenchymal stem cells (MSC), which are found in bone marrow stroma (Friedenstein et al., 1976; Caplan et al., 1991; Pittenger et al., 1999). These cells have been extensively characterized and studies performed with these cells have shown that they can differentiate into different mesenchymal cell lines such as adipocytes (Beresford et al., 1992), chondrocytes (Johnstone et al., 1998), myoblasts (Wakitani et al., 1995) and osteoblasts (Haynesworth et al., 1992). Likewise, they also have the capacity to differentiate into neurons (Sanchez-Ramos et al., 2000).
  • The ideal source of adult stem cells is one in which they can be obtained by an easy, non-invasive process and one that allows a sufficient number of cells to be isolated. In particular, a source should provide stem cells that can be easily isolated from a living subject without significant risks and discomfort and the source should allow a high yield to be obtained with minimal contamination from other cell types, without excessive cost of isolation and culture.
  • The process of obtaining bone marrow is painful and the yield is very low, a substantial increase in the number of cells being necessary by ex vivo expansion, to obtain clinically relevant amount. This step increases cost and makes the procedure time consuming, as well as increases the risk of contamination and loss of material. For these reasons, it would be very desirable to be able to isolate multipotent cells from mesenchymal tissues other than bone marrow. In particular, given their surgical accessibility, it would be convenient to be able to isolate cells from non-osteochondral mesodermal tissues such as, but not limited to, skin, fat and muscle tissue.
  • The presence of different populations of multipotent adult cells in soft tissues derived from the embryonic mesoderm has been reported by several authors. For example, it has been reported that multipotent cells can be obtained from skeletal muscle and other connective tissue of mammals (Young et al. 1993, Rogers et al. 1995). Multipotent cells have also been obtained from human lipoaspirated tissue (Zuk et al., 2001). Another example of multipotent cells isolated from adult connective tissue is the so-called Multipotent Adult Progenitor Cells (MAPC) obtained from bone marrow (Jiang et al., 2002). In principle, all these isolated cell populations could be used in the repair and regeneration of connective tissue in a similar fashion to the MSC of bone marrow (Caplan et al., 2001). However, except for MAPC, none of these populations has been, until present, sufficiently characterized at the phenotype level. Therefore, although the presence of multipotent adult cells has been described in different connective tissues, in the current state of the art, it is not possible to identify and unequivocally distinguish between different multipotent cell types obtained from soft tissue, or to obtain a substantially pure population.
  • Currently, phenotype characterization of stem cells comprises determination of markers such as cell surface receptors, among others; and the determination of their capacity for differentiation in in vitro cultures. Each cell type has a certain combination of surface markers, that is, it has a certain profile of expression that characterizes that particular cell type, distinguishing it from others.
  • Different combinations of surface markers have been used for identifying and isolating substantially pure populations of hematopoietic stem cells from the bone marrow of mice, such as: [Linneg/low, Thy1.1low, c-Kithigh, Sca-1+j, [Lin, Thy1.1low, Sca-1+, rhodamine 123low] (Morrison, S. J. et al., 1995) or [Lin, CD34−/int, c-Kit+, Sca-1+] (Osawa, M. et al., 1996). Likewise, similar combinations of markers have been used for enriching populations of human hematopoietic stem cells [Lin, Thy1+, CD34+, CD38neg/low] (Morrison, S. J. et al., 1995).
  • Currently, it is not known how many markers associated with compromised and differentiated cells are also present in the different multipotent adult mesenchymal cell populations. For example, a commonly used marker for enriching multipotent adult mesenchymal cells is CD44 (hyaluronic acid receptor). Nevertheless, CD44 is also present in different types of compromised and differentiated cell types. The uncertainty about which markers are associated with the stem cells to allow them to be distinguished from those cells that show a greater degree of differentiation, along with the low percentage of stem cells present in adult cells, has made it difficult to identify and purify populations of multipotent adult mesenchymal cells.
  • A significant disadvantage in using multipotent adult cells resides in the fact that most of the current sources for obtaining multipotent adult cells are contaminated with other cell types, complicating the process of identification, isolation and characterization of the populations of multipotent adult cells with the objective of using them for therapeutic or other ends. Thus, there is an interest in obtaining a population of multipotent adult cells isolated in a substantially pure form.
  • The characterization of a multipotent adult cell population from non-osteochondral mesenchymal tissue will allow a method for identification and isolation to be designed, as well as the identification of growth factors associated with self-regeneration. Moreover, there may be growth factors associated with the initial phases of differentiation, knowledge of which would allow more efficient in vivo and ex vivo differentiation, as well as for exercising control over the proliferation of stem cells.
  • The present invention provides a multipotent adult cell population from non-osteochondral mesenchymal tissue, preferable from adipose tissue, isolated and characterized by means of immunophenotype markers present on the cell surface, showing their multipotent nature.
  • Similarly, the present invention provides a method for the identification and isolation of a population of multipotent adult cells from non-osteochondral mesenchymal tissue, dependent on a pattern of characteristic immunophenotype markers, allowing a composition of substantially homogeneous multipotent stem cell markers to be obtained.
  • BRIEF DESCRIPTION OF THE INVENTION
  • In an aspect, the invention relates to an isolated multipotent adult cell, hereinafter referred to as the multipotent adult cell of the invention, which (a) is isolated from non-osteochondral mesenchymal tissue; (b) expresses CD9<+>, CD10<+>, CD13<+>, CD29<+>, CD44<+>, CD49a<+>, CD51<+>, CD54<+>, CD55<+>, CD58<+>, CD59<+>, CD90<+> and CD105<+>; and (c) lacks expression of CD11b, CD14, CD15, CD16, CD31, CD34, CD45, CD49f, CD102, CD104, CD106 and CD133. The multipotent adult cells of the invention present the capacity to proliferate and to be differentiated into different cell lineages. In a particular embodiment, the multipotent adult cells of the invention can be differentiated into bone phenotype cells, muscle phenotype cells and neuronal phenotype cells.
  • In another aspect, the invention relates to an isolated cell population that comprises, or consists of, the multipotent adult cells of the invention. In an embodiment, said cell population is nearly, i.e., substantially, homogeneous.
  • In another aspect, the invention relates to a substantially homogeneous cell composition which comprises a multipotent adult cell of the invention or a cell population of multipotent adult cells of the invention.
  • In another aspect, the invention relates to a cell that expresses at least one characteristic of a specialized cell, wherein the cell is derived from an isolated multipotent adult cell of the invention. In an embodiment, the invention relates to a cell that expresses at least one characteristic of a specialized cell, wherein the at least one characteristic is that of a cell selected from the group consisting of an epithelial cell, an endothelial cell, an adipocyte, a myocyte, a chondrocyte, an osteocyte, a neuron, an astrocyte, an oligodendrocyte, a hepatocyte, a cardiomyocyte, and a pancreatic cell. An isolated cell population that comprises said cells that express at least one characteristic of a specialized cell, wherein the cells are derived from isolated multipotent adult cells of the invention, constitutes a further aspect of the invention.
  • In another aspect, the invention relates to a method for obtaining the isolated multipotent adult cells of the invention which comprises:
      • a) collecting a non-osteochondral mesenchymal tissue;
      • b) obtaining a cell suspension by enzymatic digestion;
      • c) sedimentating the cells and resuspending the cells in an appropriate culture medium; and
      • d) culturing the cells, and eliminating cells that show no adhesion.
  • The cells obtained and isolated according to said method contain the characteristics of the multipotent adult cells of the invention, i.e., they (a) have been isolated from non-osteochondral mesenchymal tissue; (b) are positive to the following markers: CD9, CD10, CD13, CD29, CD44, CD49A, CD51, CD54, CD55, CD58, CD59, CD90 and CD105; and (c) lack expression of the following markers: CD11b, CD14, CD15, CD16, CD31, CD34, CD45, CD49f, CD102, CD104, CD106 and CD133. In addition, said cells present the capacity to proliferate and to be differentiated into different cell lineages.
  • In a preferred embodiment, the non-osteochondral mesenchymal tissue is a connective tissue, preferably adipose tissue. In a still another preferred embodiment, the multipotent adult cells of the invention can be genetically modified.
  • In another aspect, the invention relates to a method for identifying a population of multipotent adult cells, wherein said population comprises, or consists of, isolated multipotent adult cells of the invention, the method comprising:
  • (a) incubating the cells with labelled specific binding compounds for one or more characteristic markers for said population; and
  • (b) detecting the presence or absence of binding by the cells to these specific binding compounds.
  • In a preferred embodiment, said specific binding compound is an antibody.
  • In another aspect, the present invention relates to a method for isolating a population of multipotent adult cells of the invention, which comprises:
      • (a) collecting a non-osteochondral mesenchymal tissue;
      • (b) obtaining a cell suspension from the tissue by enzymatic digestion;
      • (c) incubating the cell suspension with a labelled compound that binds specifically to one or more of the surface markers characteristic for said population; and
      • (d) selecting those cells that have the profile of expression of markers.
  • The presence or absence of said surface markers characterizes said cells, thus, being characteristic for said cell population. Cells that have the profile of expression of characteristic markers of the multipotent adult cells of the invention are finally selected.
  • In a preferred embodiment, said method of isolation consists of performing a negative selection, whereby cells that show binding to labelled compounds that bind specifically to a marker selected from the group consisting of CD11b, CD14, CD15, CD16, CD31, CD34, CD45, CD49f, CD102, CD104, CD106 and CD133 are excluded, and a subsequent positive selection, whereby cells that bind to labelled compounds that bind specifically to a marker selected from the group consisting of CD9, CD10, CD13, CD29, CD44, CD49a, CD51, CD54, CD55, CD58, CD59, CD90 and CD105 are selected. Preferably, the labelled compound of specific binding is an antibody.
  • In another aspect, the invention relates to a multipotent adult cell of the invention, or a population of multipotent adult cells of the invention, for therapeutic use, e.g., for use as a medicament. In an embodiment, the invention relates to a multipotent adult cell of the invention, or to a population of multipotent adult cells of the invention for use in the repair and regeneration of tissues.
  • Thus, in another aspect, the invention relates to a pharmaceutical composition that comprises a multipotent adult cell of the invention, or a population of multipotent adult cells of the invention, and a pharmaceutically acceptable carrier. In a preferred embodiment, said pharmaceutical composition is useful for the repair and regeneration of tissues.
  • Further, in another aspect, the invention relates to the use of a multipotent adult cell of the invention, or a cell population of multipotent adult cells of the invention, for the manufacture of a pharmaceutical composition for the repair and regeneration of tissues.
  • Also, in other aspect, the invention relates to a therapeutic method comprising administering said pharmaceutical composition to a patient in need thereof. In an embodiment, said therapeutic method is for tissue repair or regeneration.
  • In another aspect, the invention, relates to a method for assessing in vitro or in vivo cell response to a biological or pharmacological agent, or to a combinatorial library of said agents, which comprises:
      • (a) isolating a cell population of multipotent adult cells of the invention, wherein the cells are nearly homogeneous,
      • (b) expanding the cell population via culturing;
      • (c) applying a biological agent or a pharmacological agent or a combinatorial library of said agents to said cell population, and assessing the effects of said agents on the cultured cells.
  • In an embodiment, the population of multipotent adult cells of the invention of step (a) is isolated from an individual or from a statistically significant population thereof. In other embodiment, the cells of the cell population of multipotent adult cells of the invention of step (a) are nearly, i.e., substantially, homogeneous. In another embodiment, prior to step (c), the cells are allowed to differentiate into a specific type of cells.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIGS. 1 a-1 d show histograms of fluorescence immunocytometry corresponding to the profile of surface markers obtained from cells isolated from liposuction samples of a healthy donor. The results show the evolution over time of the markers studied in the cell cultures, indicating in each case the particular time period to which the analyzed cells belong. FIG. 1 a shows the expression of markers on Day 0. FIG. 1 b shows the expression of markers on Day 7 of culturing. FIG. 1 c shows the expression of markers after 4 weeks of culturing and FIG. 1 d shows the expression of markers after 3 months of culturing.
  • FIGS. 2 a-2 d show the histograms of fluorescence immunocytometry corresponding to the profile of surface markers obtained from cells isolated from liposuction samples from a second healthy donor. The results show the evolution over time of the markers studied in the cell cultures, indicating in each case the particular time period to which the analyzed cells belong. FIG. 2 a shows the expression of markers on Day 0. FIG. 2 b shows the expression of markers on Day 7 of culturing. FIG. 2 c shows the expression of markers after 4 weeks of culturing and FIG. 2 d shows the expression of markers after 3 months of culturing.
  • FIGS. 3 a-3 d show the histograms of fluorescence immunocytometry corresponding to the profile of surface markers obtained from cells isolated from liposuction samples from a third healthy donor. The results show the evolution over time of the markers studied in the cell cultures, indicating in each case the particular time period to which the analyzed cells belong. FIG. 3 a shows the expression of markers on Day 0. FIG. 3 b shows the expression of markers on Day 7 of culturing. FIG. 3 c shows the expression of markers after 4 weeks of culturing and FIG. 3 d shows the expression of markers after 3 months of culturing.
  • FIGS. 4 a-4 d show microphotographs of cells incubated in osteogenic medium for 3 weeks. FIG. 4 a shows mesenchymal stem cells from human bone marrow (positive control). FIG. 4 b shows the cells of the invention, multipotent adult cells from non-osteochondral mesenchymal tissue, incubated in osteogenic medium for the first week. FIG. 4 c shows the multipotent adult cells of the invention, incubated in osteogenic medium for the second week. FIG. 4 d shows the multipotent adult cells of the invention, incubated in osteogenic medium during the third week.
  • DETAILED DESCRIPTION OF THE INVENTION
  • With the objective of designing an identification and isolation method useful for obtaining a defined population of multipotent adult cells from non-osteochondral mesenchymal tissue (“soft tissue”), the phenotype of recently isolated human mesenchymal cells obtained from sub-dermal adipose tissue was analyzed and also the evolution of surface markers was studied during the expansion of the cells in vitro, as well as their capacity for differentiating into different cell lineages.
  • Firstly, expression of a series of surface markers on the adult cells from sub-dermal adipose tissue was monitored by flow cytometry when cells were recently isolated and also during the development of the culture in vitro. To do this, a series of commonly used markers were used to identify stem cells, as well as to characterize differentiated cells, including but not limited to: integrins, hematopoietic markers, growth factor receptors and extracellular matrix receptors (Example 1).
  • The characterization of multipotent adult cells from non-osteochondral mesenchymal tissue by means of determining their immunophenotype profile allows to define said population in terms of the presence or absence of a certain set of surface markers. These markers are epitopes that can be identified using specific antibodies, constituting a valuable tool that allows to identify the population, as well as to design a strategy for isolation or purification thereof.
  • Subsequently, once characterized, the isolated cells were subjected to differentiation assays with the objective of showing their multipotent nature. To do this, the isolated and characterized cells were induced to in vitro differentiate into cells that express at least one characteristic of a specialized cell. The methods that can be used to induce differentiation of the multipotent adult cells of the present invention into different specific cell types are known by those skilled in the art and some of them are explained in detail in Examples 2, 3 and 4, which show in vitro differentiation of the multipotent adult cells of the invention into bone phenotype cells, muscle phenotype cells and neuronal phenotype cells, respectively.
  • Therefore, in an aspect, the invention relates to an isolated multipotent adult cell, hereinafter referred to as the multipotent adult cell of the invention, which (a) is isolated from non-osteochondral mesenchymal tissue; (b) expresses CD9<+>, CD10<+>, CD13<+>, CD29<+>, CD44<+>, CD49a<+>, CD51<+>, CD54<+>, CD55<+>, CD58<+>, CD59<+>, CD90<+> and CD105<+>; and (c) lacks expression of CD11b, CD14, CD15, CD16, CD31, CD34, CD45, CD49f, CD102, CD104, CD106 and CD133.
  • An isolated cell population that comprises, or consists of, the multipotent adult cells of the invention constitutes a further aspect of the present invention. In a particular embodiment, said cell population is nearly, i.e., substantially, homogeneous.
  • The multipotent adult cells of the invention are isolated from non-osteochondral mesenchymal tissue. The term “non-osteochondral mesenchymal tissue” refers to a mesenchymal tissue other than cartilage and bone marrow. Non-osteochondral mesodermal tissues are also referred in this description to as “soft tissues”. Illustrative, non-limitative, examples of non-osteochondral mesodermal tissues include skin, fat and muscle tissue. In a particular embodiment, the multipotent adult cells of the invention are isolated from sub-dermal adipose tissue.
  • The multipotent adult cells of the invention can be obtained from any suitable source of non-osteochondral mesenchymal tissue from any suitable animal, including human beings. In general, said cells are obtained from non-pathological post-natal mammalian non-osteochondral mesenchymal tissues. In a preferred embodiment, the multipotent adult cells of the invention are obtained from a source of non-osteochondral mesenchymal tissue, such as sub-dermal adipose tissue. Also, in a particular embodiment, the multipotent adult cells of the invention are isolated from a mammal, e.g., a rodent, primate, etc., preferably, from a human being.
  • The multipotent adult cells of the invention are also characterized by the presence and absence of a set of markers, namely, said cells are characterized in that they (i) are positive for some markers [CD9<+>, CD10<+>, CD13<+>, CD29<+>, CD44<+>, CD49a<+>, CD51<+>, CD54<+>, CD55<+>, CD58<+>, CD59<+>, CD90<+> and CD105<+>], and (ii) are negative for some markers [CD11b, CD14, CD15, CD16, CD31, CD34, CD45, CD49f, CD102, CD104, CD106 and CD133].
  • The phenotype characterization of the multipotent adult cells of the invention, or of a cell population comprising said cells, by surface markers can be conventionally performed either by individual staining of the cells (flow cytometry) or by making histological cuts of the population in situ, in accordance with conventional methods. In a particular embodiment, the expression of said surface markers on the multipotent adult cells of the invention may be monitored by flow cytometry.
  • The characterization of the multipotent adult cells of the invention, or a cell population comprising, or consisting of, said cells, by their immuno-phenotype profile can be used to define said cells or cell population in terms of the presence or absence of a certain set of surface markers. These markers are epitopes that can be identified with specific antibodies, constituting a valuable tool that allows to identify the population, as well as to design a strategy for isolation or purification thereof. Monoclonal antibodies against said surface markers can be used to identify the multipotent adult cells of the invention.
  • The determination of the profile of surface markers by antibodies (immunophenotype characterization) may be direct, using a labelled antibody, or indirect, using a second labelled antibody against the primary specific antibody of the cell marker, thus achieving signal amplification.
  • On the other hand, the presence or absence of binding of the antibody may be determined by different methods that include but are not limited to immunofluorescence microscopy and radiography. Similarly, it is possible to carry out the monitoring of the levels of binding of the antibody by flow cytometry, a technique that allows the levels of fluorochrome to be correlated with the quantity of antigens present on the cell surface bound specifically to the labelled antibodies.
  • In the assay of identification and isolation, the cell population comes into contact with a specific reagent, labelled or not, depending on whether the assay is performed by a direct or indirect detection method, respectively. The term “specific reagent” refers to a member of a specific binding pair; members of a specific binding pair, include but are not limited to, binding pairs of antigens and antibodies, pairs comprising MHC antigens and T-cell receptors, complementary nucleotide sequences, as well as pairs of peptide ligands and their receptor. The specific binding pairs include analogues, fragments and derivatives of the specific member of the binding pair.
  • The use of antibodies as reagents with affinity is of particular interest. The production of specific monoclonal antibodies will be evident to any ordinary skilled person in the art. In experiments of identification or separation of cell populations, the antibodies are labelled. For this purpose, markers that are used include but are not limited to: magnetic particles, biotin and fluorochromes that will allow identification or separation of the cell type to which the antibody has bound. Thus, for example, the analysis of the cell population comprising the multipotent adult cells of the invention by flow cytometry allows different antibodies labelled with fluorochromes that emit at different wavelengths to be used in the same sample. Thus, it is possible to know the specific profile of the population for these surface markers, as well as carry out a separation for the set of markers used.
  • The separation of the populations that present the phenotype of interest can be carried out by affinity separation techniques, which include magnetic separation (using magnetic particles coated with specific antibodies), affinity chromatography, cytotoxic agents bound to monoclonal antibodies or used along with monoclonal antibodies and panning with the antibody attached to a solid support, as well as by other techniques that are appropriate. A more precise separation would be obtained by flow cytometry, a technique that allows the separation of cell populations according to the intensity of staining, along with other parameters such as cell size and cell complexity.
  • The multipotent adult cells of the invention present the capacity to proliferate and to be differentiated into different cell lineages. Illustrative, non-limitative examples of cell lineages in which the multipotent adult cells of the invention can be differentiated include, e.g., bone phenotype cells, muscle phenotype cells and neuronal phenotype cells.
  • The multipotent adult cells of the invention can proliferate and differentiate into cells of other lineages by conventional methods. Methods of identifying and subsequently isolating differentiated cells from their undifferentiated counterparts can be also carried out by methods well known in the art.
  • The multipotent adult cells of the invention are also capable of being expanded ex vivo. That is, after isolation, the multipotent adult cells of the invention can be maintained and allowed to proliferate ex vivo in culture medium. Such medium comprises, for example, Dulbecco's Modified Eagle's Medium (DMEM), antibiotics, and glutamine, and it is usually supplemented with 2-20% fetal bovine serum (FBS). It is within the skill of one in the art to modify or modulate concentrations of media and/or media supplements as necessary for the cells used. Sera often contain cellular factors and components that are necessary for viability and expansion. Examples of sera include FBS, bovine serum (BS), calf serum (CS), fetal calf serum (FCS), newborn calf serum (NCS), goat serum (GS), horse serum (HS), porcine serum, sheep serum, rabbit serum, rat serum (RS), etc. Also it is contemplated, if the multipotent adult cells of the invention are of human origin, supplementation of cell culture medium with a human serum, preferably of autologous origin. It is understood that sera can be heat-inactivated at 55-65° C. if deemed necessary to inactivate components of the complement cascade. Modulation of serum concentrations, withdrawal of serum from the culture medium can also be used to promote survival of one or more desired cell types. Preferably, multipotent adult cells of the invention will benefit from FBS concentrations of about 2% to about 25%. In another embodiment, the multipotent adult cells of the invention can be expanded in a culture medium of definite composition, in which the serum is replaced by a combination of serum albumin, serum transferrin, selenium, and recombinant proteins including but not limited to: insulin, platelet-derived growth factor (PDGF), and basic fibroblast growth factor (bFGF).
  • Many cell culture media already contain amino acids; however some require supplementation prior to culturing cells. Such amino acids include, but are not limited to, L-alanine, L-arginine, L-aspartic acid, L-asparagine, L-cysteine, L-cystine, L-glutamic acid, L-glutamine, L-glycine, and the like.
  • Antimicrobial agents are also typically used in cell culture to mitigate bacterial, mycoplasmal, and fungal contamination. Typically, antibiotics or anti-mycotic compounds used are mixtures of penicillin/streptomycin, but can also include, but are not limited to amphotericin (Fungizone®), ampicillin, gentamicin, bleomycin, hygromacin, kanamycin, mitomycin, etc.
  • Hormones can also be advantageously used in cell culture and include, but are not limited to, D-aldosterone, diethylstilbestrol (DES), dexamethasone, b-estradiol, hydrocortisone, insulin, prolactin, progesterone, somatostatin/human growth hormone (HGH), etc.
  • The maintenance conditions of the multipotent adult cells of the invention can also contain cellular factors that allow cells to remain in an undifferentiated form. It is apparent to those skilled in the art that prior to differentiation supplements that inhibit cell differentiation must be removed from the culture medium. It is also apparent that not all cells will require these factors. In fact, these factors may elicit unwanted effects, depending on the cell type.
  • The multipotent adult cells of the invention can be transfected or genetically engineered to express, at least, one polypeptide of interest. In an embodiment, the polypeptide of interest is a product capable of inducing or increasing the expression of genes involved in the repair or regeneration of a tissue.
  • In another aspect, the invention relates to a method for obtaining the isolated multipotent adult cells of the invention which comprises:
      • a) collecting a non-osteochondral mesenchymal tissue;
      • b) obtaining a cell suspension by enzymatic digestion;
      • c) sedimentating the cells and resuspending the cells in an appropriate culture medium; and
      • d) culturing the cells in a solid surface, and eliminating cells that show no adhesion to said solid surface.
  • The cells obtained according to said method contain the characteristics of the multipotent adult cells of the invention.
  • As used herein, the term “solid surface” refers to any material that allow the multipotent adult cells of the invention to adhere. In a particular embodiment said material is a plastic material treated to promote the adhesion of mammalian cells to its surface.
  • Steps a)-d) can be carried out by conventional techniques known by those skilled in the art. Briefly, the multipotent adult cells of the invention can be obtained by conventional means from any suitable source of non-osteochondral mesenchymal tissue from any suitable animal, including human beings, e.g., from human adipose tissue. The animal can be alive or dead, so long as non-osteochondral mesenchymal tissue cells within the animal are viable. Typically, human adipose cells are obtained from living donors, using well-recognized protocols such as surgical or suction lipectomy. Indeed, as liposuction procedures are so common, liposuction effluent is a particularly preferred source from which the cells of the invention can be derived. Thus, in a particular embodiment, the multipotent adult cells of the invention are from human sub-dermal adipose tissue obtained by liposuction.
  • The sample of non-osteochondral mesenchymal tissue is, preferably, washed before being processed. In a protocol, the sample of non-osteochondral mesenchymal tissue is washed with physiologically-compatible saline solution (e.g., phosphate buffered saline (PBS)) and then vigorously agitated and left to settle, a step that removes loose matter (e.g., damaged tissue, blood, erythrocytes, etc) from the tissue. Thus, the washing and settling steps generally are repeated until the supernatant is relatively clear of debris. The remaining cells generally will be present in lumps of various sizes, and the protocol proceeds using steps gauged to degrade the gross structure while minimizing damage to the cells themselves. One method of achieving this end is to treat the washed lumps of cells with an enzyme that weakens or destroys bonds between cells (e.g., collagenase, dispase, trypsin, etc.). The amount and duration of such enzymatic treatment will vary, depending on the conditions employed, but the use of such enzymes is generally known in the art. Alternatively or in conjunction with such enzymatic treatment, the lumps of cells can be degraded using other treatments, such as mechanical agitation, sonic energy, thermal energy, etc. If degradation is accomplished by enzymatic methods, it is desirable to neutralize the enzyme following a suitable period, to minimize deleterious effects on the cells.
  • The degradation step typically produces a slurry or suspension of aggregated cells and a fluid fraction containing generally free stromal cells (e.g., red blood cells, smooth muscle cells, endothelial cells, fibroblast cells, and stem cells). The next stage in the separation process is to separate the aggregated cells from the cells of the invention. This can be accomplished by centrifugation, which forces the cells into a pellet covered by a supernatant. The supernatant then can be discarded and the pellet suspended in a physiologically-compatible fluid. Moreover, the suspended cells typically include erythrocytes, and in most protocols it is desirable to lyse them. Methods for selectively lysing erythrocytes are known in the art, and any suitable protocol can be employed (e.g., incubation in a hyper-or hypotonic medium, by lysis using ammonium chloride, etc.). Of course, if the erythrocytes are lysed, the remaining cells should then be separated from the lysate, for example by filtration, sedimentation, or density fractionation.
  • Regardless of whether the erythrocytes are lysed, the suspended cells can be washed, re-centrifuged, and resuspended one or more successive times to achieve greater purity. Alternatively, the cells can be separated on the basis of cells surface markers profile or on the basis of cell size and granularity.
  • Following the final isolation and resuspension, the cells can be cultured and, if desired, assayed for number and viability to assess the yield. Desirably, the cells will be cultured without differentiation, on a solid surface, using a suitable cell culture media, at the appropriate cell densities and culture conditions. Thus, in a particular embodiment, cells are cultured without differentiation on a solid surface, usually made of a plastic material, such as Petri dishes or cell culture flasks, in the presence of a suitable cell culture medium [e.g., DMEM, typically supplemented with 5-15% (e.g., 10%) of a suitable serum, such as fetal bovine serum or human serum], and incubated under conditions which allow cells to adhere to the solid surface and proliferate. After incubation, cells are washed in order to remove non-adhered cells and cell fragments. The cells are maintained in culture in the same medium and under the same conditions until they reach the adequate confluence, typically, about 80% cell confluence, with replacement of the cell culture medium when necessary. After reaching the desired cell confluence, the cells can be expanded by means of consecutive passages using a detachment agent such as trypsin and seeding onto a bigger cell culture surface at the appropriate cell density (usually 2,000-10,000 cells/cm2). The cells can be passaged some times without losing their developmental phenotype. Typically, the cells are plated at a desired density such as between about 100 cells/cm2 to about 100,000 cells/cm2 (such as about 500 cells/cm2 to about 50,000 cells/cm2, or, more particularly, between about 1,000 cells/cm2 to about 20,000 cells/cm2). If plated at lower densities (e.g., about 300 cells/cm2), the cells can be more easily clonally isolated. For example, after a few days, cells plated at such densities will proliferate into an homogeneous population. In a particular embodiment, the cell density is between 2,000-10,000 cells/cm2.
  • Cells which remain adhered to the solid surface are selected and the phenotype thereof is analyzed by conventional methods in order to confirm the identity of the multipotent adult cells of the invention as will be mentioned below. Cells which remain finally adhered to the solid surface constitute a homogeneous cell population of multipotent adult cells of the invention. Example 1 describes in a detailed manner the isolation of multipotent adult cells of the invention from human sub-dermal adipose tissue.
  • Usually, cells which remain adhered to the solid surface show the desired phenotype, although it has to be confirmed so that the cells can be used according to the invention. Therefore, the adhesion of cells to the solid surface constitutes a criteria for selecting the multipotent adult cells of the invention. Confirmation of the phenotype of interest can be carried out by using conventional means.
  • Cell-surface markers can be identified by any suitable conventional technique, usually based on a positive/negative selection; for example, monoclonal antibodies against cell-surface markers, which presence/absence in the cells has to be confirmed, can be used; although other techniques can also be used. Thus, in a particular embodiment, monoclonal antibodies against CD9, CD10, CD13, CD29, CD44, CD49a, CD51, CD54, CD55, CD58, CD59, CD90 and CD105 are used in order to confirm the absence of said markers in the selected cells; and monoclonal antibodies against CD11b, CD14, CD15, CD16, CD31, CD34, CD45, CD49f, CD102, CD104, CD106 and CD133 are used in order to confirm the absence of said markers in the selected cells. Said monoclonal antibodies are known or can be obtained by a skilled person in the art by conventional methods.
  • The capacity of the selected cells to differentiate into different cell lineages can be assayed by conventional methods as previously disclosed.
  • The multipotent adult cells of the invention and cell populations provided by the instant invention can be clonally expanded, if desired, using a suitable method for cloning cell populations. For example, a proliferated population of cells can be physically picked and seeded into a separate plate (or the well of a multi-well plate). Alternatively, the cells can be subcloned onto a multi-well plate at a statistical ratio for facilitating placing a single cell into each well (e.g., from about 0.1 to about 1 cell/well or even about 0.25 to about 0.5 cells/well, such as 0.5 cells/well). Of course, the cells can be cloned by plating them at low density (e.g., in a Petri dish or other suitable substrate) and isolating them from other cells using devices such as a cloning rings. The production of a clonal population can be expanded in any suitable culture medium. In any event, the isolated cells can be cultured to a suitable point when their developmental phenotype can be assessed.
  • Further assays carried out by the inventors have shown that ex vivo expansion of the cells of the invention without inducing differentiation can be accomplished for extended time periods by using specially screened lots of suitable serum (such as fetal bovine serum or human serum). Methods for measuring viability and yield are known in the art (e. g., trypan blue exclusion).
  • Any of the steps and procedures for isolating the cells of the cell population of the invention can be performed manually, if desired. Alternatively, the process for isolating such cells can be facilitated through a suitable device, many of which are known in the art.
  • In another aspect, the invention relates to a method for identifying a population of multipotent adult cells, wherein said population comprises, or consists of, multipotent adult cells of the invention, the method comprising:
  • (a) incubating the cells with labelled specific binding compounds for one or more characteristic markers for said population; and
  • (b) detecting the presence or absence of binding by the cells to these specific binding compounds.
  • This method can be carried out as previously mentioned in connection with the immunophenotype characterization of the cells of the invention. In a preferred embodiment, said specific binding compound is an antibody.
  • In another aspect, the present invention relates to a method for isolating a population of multipotent adult cells of the invention, which comprises:
      • (a) collecting a non-osteochondral mesenchymal tissue;
      • (b) obtaining a cell suspension from the tissue by enzymatic digestion;
      • (c) incubating the cell suspension with a labelled compound that binds specifically to one or more of the surface markers characteristic for said population; and
      • (d) selecting those cells that have the profile of expression of markers.
  • The presence or absence of said surface markers characterizes said cells, thus, being characteristic for said cell population. Cells that have the profile of expression of characteristic markers of the multipotent adult cells of the invention are finally selected.
  • In a preferred embodiment, said method of isolation consists of performing a negative selection, whereby cells are excluded that show binding to labelled compounds that bind specifically to a marker selected from the group consisting of CD11b, CD14, CD15, CD16, CD31, CD34, CD45, CD49f, CD102, CD104, CD106 and CD133, and a subsequent positive selection, whereby cells are selected that bind to labelled compounds that bind specifically to a marker selected from the group consisting of CD9, CD10, CD13, CD29, CD44, CD49a, CD51, CD54, CD55, CD58, CD59, CD90 and CD105. Preferably, the labelled compound of specific binding is an antibody.
  • This method can be carried out as previously mentioned in connection with the method for obtaining the multipotent adult cells of the invention.
  • The multipotent adult cells of the invention, or a cell population of multipotent adult cells of the invention, can be found in a cell composition. Therefore, in another aspect, the invention relates to a substantially homogeneous cell composition which comprises a multipotent adult cell of the invention or a cell population of multipotent adult cells of the invention.
  • The multipotent adult cells of the invention can be used for repairing and regenerating tissues. Thus, in another aspect, the invention relates to a multipotent adult cell of the invention, or a population of multipotent adult cells of the invention, for therapeutic use, e.g., for use as a medicament. In an embodiment, the invention relates to a multipotent adult cell of the invention, or to a population of multipotent adult cells of the invention for use in the repair and regeneration of tissues.
  • In another aspect, the invention relates to a pharmaceutical composition that comprises a multipotent adult cell of the invention, or a population of multipotent adult cells of the invention, and a pharmaceutically acceptable carrier. In a preferred embodiment, said pharmaceutical composition is useful for the repair and regeneration of tissues.
  • Combinations of two or more of type of multipotent adult cells of the invention are included within the scope of the pharmaceutical compositions provided by the instant invention.
  • The pharmaceutical composition of the invention comprises a prophylactically or therapeutically effective amount of a multipotent adult cell of the invention, or a cell population of multipotent adult cells of the invention, and a pharmaceutically acceptable carrier. In a specific embodiment, the term “pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia, or European Pharmacopeia, or other generally recognized pharmacopeia for use in animals, and more particularly in humans. The term “carrier” refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic agent is administered. The composition, if desired, can also contain minor amounts of pH buffering agents. Examples of suitable pharmaceutical carriers are described in “Remington's Pharmaceutical Sciences” by E. W. Martin. Such compositions will contain a prophylactically or therapeutically effective amount of a multipotent adult cell of the invention, or a cell population of multipotent adult cells of the invention preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the subject. The formulation should suit the mode of administration. In a preferred embodiment, the pharmaceutical compositions are sterile and in suitable form for administration to a subject, preferably an animal subject, more preferably a mammalian subject, and most preferably a human subject.
  • The pharmaceutical composition of the invention may be in a variety of forms. These include, for example, solid, semi-solid, and liquid dosage forms, such as lyophilized preparations, liquids solutions or suspensions, injectable and infusible solutions, etc. The preferred form depends on the intended mode of administration and therapeutic application.
  • The administration of the cells or cell population of the invention, or the pharmaceutical composition comprising same, to the subject in need thereof can be carried out by conventional means. In a particular embodiment, said cells or cell population is administered to the subject by a method which involves transferring the cells to the desired tissue, either in vitro or in vivo, to the animal tissue directly. The cells can be transferred to the desired tissue by any appropriate method, which generally will vary according to the tissue type. For example, cells can be seeded onto the desired site within the tissue to establish a population, etc. Cells can be transferred to sites in vivo using devices such as catherters, trocars, cannulae, stents (which can be seeded with the cells), etc.
  • The pharmaceutical composition of the invention can be used in a combination therapy. In a specific embodiment, the combination therapy is administered to a subject in need of treatment, such as a patient in need of repair or regeneration of a tissue. In an embodiment, the combination therapy is used in conjunction with other types of treatments to repair or regenerate tissues. In accordance with the above embodiment, the combination therapies of the invention can be used prior to, concurrently or subsequent to the administration of the multipotent adult cells of the invention.
  • Also, in another aspect, the invention relates to the use of a multipotent adult cell of the invention, or a cell population of multipotent adult cells of the invention, for the manufacture of a pharmaceutical composition for the repair and regeneration of tissues.
  • The invention further relates, in another aspect, to a therapeutic method comprising administering said pharmaceutical composition to a patient in need thereof. In an embodiment, said therapeutic method is for tissue repair or regeneration.
  • In another aspect, the invention, relates to a method for assessing in vitro or in vivo cell response to a biological or pharmacological agent, or to a combinatorial library of said agents, which comprises:
      • (a) isolating a cell population of multipotent adult cells of the invention, wherein the cells are nearly homogeneous,
      • (b) expanding the cell population via culturing;
      • (c) applying a biological agent or a pharmacological agent or a combinatorial library of said agents to said cell population, and assessing the effects of said agents on the cultured cells.
  • In an embodiment, the population of multipotent adult cells of the invention of step (a) is isolated from an individual or from a statistically significant population thereof. In other embodiment, the cells of the cell population of multipotent adult cells of the invention of step (a) are nearly, i.e., substantially, homogeneous. In another embodiment, prior to step (c), the cells are allowed to differentiate into a specific type of cells.
  • Further, in another aspect, the invention relates to a cell that expresses at least one characteristic of a specialized cell, wherein the cell is derived from an isolated multipotent adult cell of the invention. These cells are also multipotent adult cells having a differentiation stage more advanced than that of the multipotent adult cells of the invention. In an embodiment, the invention relates to a cell that expresses at least one characteristic of a specialized cell, wherein the at least one characteristic is that of a cell selected from the group consisting of an epithelial cell, an endothelial cell, an adipocyte, a myocyte, a chondrocyte, an osteocyte, a neuron, an astrocyte, an oligodendrocyte, a hepatocyte, a cardiomyocyte, and a pancreatic cell. An isolated cell population that comprises said cells that express at least one characteristic of a specialized cell, wherein the cells are derived from the isolated multipotent adult cells of the invention, constitutes a further aspect of the invention.
  • The following examples are presented to illustrate the invention, but they in no way limit it.
  • EXAMPLE 1 Isolation of Multipotent Adult Cells from Soft Tissue and Characterization of Surface Markers
  • The isolation of multipotent adult cells from soft tissue was performed by selecting those cells with a capacity for proliferation and differentiation, characterized in that they show adhesion to the plastic container of the cell culture. Then, the cells were characterized by monitoring by flow cytometry the expression of a series of surface markers on the recently isolated cells and during the course of the culture development in vitro.
  • The isolation of the multipotent adult cells was carried out from sub-dermal adipose tissue, obtained by liposuction from 3 healthy human donors ( donors 1, 2 and 3). First, the sample from the sub-dermal adipose tissue was washed with phosphate buffered saline solution (PBS). To achieve destruction of the extracellular matrix and the isolation of the cells, an enzymatic digestion was performed with type 11 collagenase in saline solution (5 mg/ml) at 37° C. for 30 minutes. The collagenase was deactivated by adding an equivalent volume of DMEM medium, with 10% fetal bovine serum. This cell suspension was centrifuged at 250 g for 10 minutes to obtain a cell deposit.
  • NH4Cl was added at an end concentration of 0.16 M and the mixture was incubated for 10 minutes at room temperature to induce the lysis of the erythrocytes present. The suspension was centrifuged at 250-400 g and resuspended in DMEM-10% FBS with 1% ampicillin-streptomycin. Finally, the cells were plated, inoculating 2×104-3×104 cells per cm2.
  • The cells were cultured for 20-24 hours at 37° C., under an atmosphere with 5% CO2. After 24 hours, the culture was washed with PBS to remove the cells and the remains of the tissue in suspension. The cells selected by adhesion to the plastic container were cultured in DMEM+10% fetal bovine serum (FBS).
  • After isolation, the multipotent adult cells isolated were characterized from one of the donors, in function of the presence/absence of a series of surface markers. To do this, the expression of the following surface markers was monitored by flow cytometry:
      • Integrin: CD11 b, CD18, CD29, CD49a, CD49b, CD49c, CD49d, CD49e, CD49f, CD51, CD61, CD104.
      • Hematopoietic markers: CD3, CD9, CD10, CD13, CD14, CD16, CD19, CD28, CD34, CD38, CD45, CD90, CD133 and glycophorin.
      • Growth factor receptors: CD105, NGFR.
      • Extracellular matrix receptors: CD15, CD31, CD44, CD50, CD54, CD62E, CD62L, CD62P, CD102, CD106, CD166.
      • Others: CD36, CD55, CD56, CD58, CD59, CD95, HLA-I, HLA-II, β2-microglobuline.
  • The immunophenotype characterization of the cells was performed on recently isolated cells and also on day 7, after 4 weeks and after 3 months of culture, of the samples from the 3 healthy human donors. Taking into account that the selection is performed by adhesion to the plastic container of the cell culture, cells from the cell fraction adhered after less than 24 hours in the culture since isolation are considered as “recently isolated cells”.
  • The cells to be characterized were collected by means of gentle digestion with trypsin, washed with PBS and incubated for 30 minutes at 4° C. with fluorescein (FITC) or phycoerythrin (PE) labelled antibodies against each one of the cell surface markers to be analyzed. The cell markers were washed and immediately analyzed using the Epics-XL cytometer (Coulter). As controls, cells stained with unspecific, FITC or PE labelled, antibodies of the corresponding isotypes were used.
  • FIGS. 1 a-1 d, 2 a-2 d and 3 a-3 d show the histograms grouped by donor for a better visualization of the evolution of the markers studied during the culturing, indicating in each case what time in the culture period the analyzed cells belong.
  • The analysis of surface markers at different times allowed their presence or absence to be determined, as well as their behaviour during the culture process. The results obtained show that the cell populations isolated from the different healthy donors show a homogeneous behaviour in their phenotype characterization.
  • From the analysis of the expression profile of surface markers (FIGS. 1 a-1 d, 2 a-2 d and 3 a-3 d), 3 criteria were used to determine which markers define the cell population and allow it to be identified and differentiated with respect to other types of cell populations. These criteria were:
      • 1. Discard those markers that vary from one sample to the other or over time during culturing.
      • 2. Verify that those that are positive are also positive at time zero (recently isolated cells).
      • 3. Select them as a function of their biological relevance, discarding markers characteristic of specific cell types (for example, CD3 is a marker exclusive to lymphocytes).
  • Applying these criteria, the multipotent adult cells isolated from non-osteochondral mesenchymal tissue provided by the present invention is characterized by being positive for CD9<+>, CD10<+>, CD13<+>, CD29<+>, CD44<+>, CD49A<+>, CD51<+>, CD54<+>, CD55<+>, CD58<+>, CD59<+>, CD90<+> and CD105<+>; and for lacking expression of CD11b, CD14, CD15, CD16, CD31, CD34, CD45, CD49f, CD102, CD104, CD106 and CD133.
  • EXAMPLE 2 In Vitro Differentiation of Multipotent Adult Cells from Human Non-Osteochondral Mesenchymal Tissue into Bone Phenotype Cells
  • In the differentiation assay, characterized human multipotent adult cells of the present invention were used. The cells were isolated from the 3 samples of analyzed lipoaspirates, each corresponding to a healthy donor (Example 1). The multipotent adult cells were isolated and characterized as mentioned in Example 1. A sample of mesenchymal stem cells (MSC) of human bone marrow was used as positive control.
  • The isolated cells were seeded at a density of 10,000 cells/cm2 onto 6-well plates (one plate per sample), and were incubated in standard culture medium (DMEM, 10% FBS, L-Glutamine 2 mM and antibiotic). After two days of culturing, the culture medium of one of the wells (control) is replaced with fresh medium, and the remaining wells with osteogenesis inducing medium, which contains the standard culture medium with the following added:
  • Dexamethasone 100 M
  • Ascorbic acid 50 μM
  • p-Glycerophosphate 10 mM
  • The cells were cultured for 3 weeks in normal conditions, changing the medium every 2-3 days. After three weeks, the presence of mineralized deposits of calcium phosphate can be seen, which indicates the presence of osseus nodules. These nodules are detected by staining with Alizarin red (Standford et al., 1995). Specifically, the medium was eliminated, the cells were washed twice with PBS and fixed with 70% cold ethanol for 30 minutes at room temperature. The fixed wells were then washed with PBS and stained with Alizarin red (40 mM, pH 4.1) for 10 minutes at room temperature. The stained cells were washed with abundant water, and the precipitates of calcium phosphate, which appear strongly stained red, were examined under the microscope.
  • FIGS. 4 a-4 d show microphotographs of the osteo-induced cells stained with Alizarin red. Although the formation of calcium phosphate is quicker in the sample corresponding to MSC from bone marrow which acts as a positive control (FIG. 4 a), in the 3 samples from the adipose tissue, the formation of large quantities of bone matrix can be discerned, although with differing intensity in each of the samples. All wells in which osteogenesis was induced showed the same behaviour and in the control wells (not submitted to osteogenic stimuli) the formation of bone matrix was not detected. No relationship was seen between the amount of bone matrix formed and the time that each sample was being cultured after isolation from the tissue (between 3 and 9 weeks).
  • EXAMPLE 3 In Vitro Differentiation of Multipotent Adult Cells from Human Non-Osteochondral Mesenchymal Tissue into Muscle Phenotype Cells
  • In the differentiation assay, characterized human multipotent adult cells of the present invention were used. The cells were isolated from the 3 samples of analyzed lipoaspirates, each corresponding to a healthy donor (Example 1). The multipotent adult cells were isolated and characterized as mentioned in Example 1. A sample of MSC of human bone marrow was used as positive control.
  • The isolated cells were seeded at a density of 10,000 cells/cm2 into standard culture medium (DMEM, 10% FBS, L-Glutamine 2 mM and antibiotic). After two days of culturing, the culture medium of one of the wells (control) was replaced with fresh medium, and the remaining wells with myogenesis inducing medium (Wakitani et al., 1995), which contains the standard culture medium with the following added:
  • Ascorbate-2-phosphate 0.1 mM
  • Dexamethasone 0.01 μM
  • ITS+1 (Sigma-Aldrich)
  • 5-Azacytidine 3 μM
  • After 24 hours, the medium was replaced by standard culture medium, and the cells were cultured for 2-3 weeks, changing the medium every 2-3 days. After this time, the cells acquired an elongated phenotype, formed fibrillar structures and some cell fusions can be seen. To detect the myoblast phenotype, the cells obtained were fixed with paraformaldehyde (PFA) at 4% and incubated with an antibody against the heavy chain of myosin, which is the specific antigen for muscle. The results confirm the differentiation of the human multipotent adult cells of the invention into muscle phenotype cells.
  • EXAMPLE 4 In Vitro Differentiation of Multipotent Adult Cells from Human Non-Osteochondral Mesenchymal Tissue into Neuronal Phenotype Cells
  • In the differentiation assay, characterized human multipotent adult cells of the present invention were used. The cells were isolated from the 3 samples of analyzed lipoaspirates, each corresponding to a healthy donor (Example 1). The multipotent adult cells were isolated and characterized as mentioned in Example 1. A sample of MSC from human bone marrow was used as positive control.
  • The isolated cells were seeded at low density, 3×103 cells/cm2, into standard culture medium (DMEM, 10% FBS, L-Glutamine 2 mM and antibiotic), supplemented with 10 ng/ml bFGF and incubated for 24-36 hours to yield a large number of cells. The wells were then washed and neuron-inducing medium was added (Black and Woodbury, 2001), comprising:
  • αMEM
  • BHA 200 μM
  • Penicillin/streptomycin
  • L-Glutamine 2 mM
  • Forskolin 10 μM
  • 2% DMSO
  • Hydrocortisone 1 μM
  • Insulin 5 μg/ml
  • CIK 25 mM
  • Valproic acid 2 mM
  • A few hours after induction, a morphological change could be observed; the cells acquired a rounded shape and very refringent, with prolongations with a similar appearance to axons and dendrites of nerve cells. After 3 days, the cells obtained were fixed with PFA at 4% and incubated with antibodies against neuron specific antigens NF-200 and TuJ1. The results confirm the differentiation of the human multipotent adult cells of the invention into muscle phenotype cells.
  • REFERENCES
    • Osawa M., Hanada K., Hanada H. and Nakauchi H. (1996) Science 273, 242-245.
    • Morrison S. J., Uchida N. and Weissman I. L. (1995) Annu. Rev. Cell Dev. Biol. 11, 35-71.
    • Ivanova N. B., Dimos J. T., Schaniel C., Hackney J. A., Moore K. A., Lemischka I. R. (2002) Science 298, 601-604.
    • Phillips R L. (2000) Curr Top Microbiol Immunol. 251, 13-19.
    • Ramalho-Santos M, Yoon S, Matsuzaki Y, Mulligan R C, Melton D A. (2002) Science 298, 597-600.
    • Friedenstein A J, Gorskaja J F, Kulagina N N, Exp Hematol. September 1976;4(5):267-74.
    • Caplan A I J Orthop Res. September 1991;9(5):641-50
    • Pittenger, M. F. et al. (1999) Science 284: 143-147
    • Beresford J N, Bennett J H, Devlin C, Leboy P S, Owen M E, J Cell Sci. June 1992;102 (Pt 2):341-51
    • Yoo J U, Johnstone B, Clin Orthop. October 1998;(355 Suppl):S73-81
    • Wakitani S. et al. (1995) Muscle Nerve 18: 1417-1426.
    • Haynesworth S E, Goshima J, Goldberg V M, Caplan A I, Bone. 1992; 13(1):81-8.
    • Sanchez-Ramos J, Song S, Cardozo-Pelaez F, Hazzi C, Stedeford T, Willing A, Freeman T B, Saporta S, Janssen W, Patel N, Cooper D R, Sanberg P R, Exp Neurol. August 2000; 164(2):247-56.
    • Rogers J J, Young H E, Adkison L R, Lucas P A, Black A C Jr, Am Surg. March 1995; 61 (3):231-6.
    • Zuk, P. A. et al. (2001) Tissue Eng 7: 211-228.
    • Jiang Y, Vaessen B, Lenvik T, Blackstad M, Reyes M, Verfaillie C M, Exp Hematol. August 2002; 30(8):896-904.
    • Caplan A I, Bruder S P, Trends Mol Med. June 2001; 7(6):259-64. 5
    • Stanford. C. M. et al. (1995) J Biol Chem 270: 9420-9428.
    • Young H E, Ceballos E M, Smith J C, Mancini M L, Wright R P, Ragan B L, Bushell I, Lucas P A. In vitro Cell Dev Biol Anim. September 1993; 29 A(9):723-36.

Claims (24)

1. An isolated multipotent adult cell, which (a) is isolated from non-osteochondral mesenchymal tissue, (b) expresses CD9<+>, CD10<+>, CD13<+>, CD29<+>, CD44<+>, CD49A<+>, CD51<+>, CD54<+>, CD55<+>, CD58<+>, CD59<+>, CD90<+> and CD105<+>; and (c) lacks expression of CD11b, CD14, CD15, CD16, CD31, CD34, CD45, CD49f, CD102, CD104, CD106 and CD133.
2. An isolated multipotent adult cell according to claim 1, which is isolated by a method that comprises:
(a) collecting a non-osteochondral mesenchymal tissue;
(b) obtaining a cell suspension by enzymatic digestion;
(c) sedimenting and resuspending the cells in a culture medium; and
(d) culturing the cells on a solid surface, and eliminating cells that show no adhesion to said solid surface.
3. The isolated multipotent adult cell according to claim 1, wherein the non-osteochondral mesenchymal tissue is connective tissue.
4. The isolated multipotent adult cell according to claim 3, wherein said connective tissue is adipose tissue.
5. The isolated multipotent adult cell according to claim 1, wherein the cell is a genetically modified cell.
6. A cell that expresses at least one characteristic of a specialized cell, wherein the cell is derived from an isolated multipotent adult cell according to claim 1.
7. The cell according to claim 6, wherein the at least one characteristic is that of a cell selected from the group consisting of an epithelial cell, an endothelial cell, an adipocyte, a myocyte, a chondrocyte, an osteocyte, a neuron, an astrocyte, an oligodendrocyte, a hepatocyte, a cardiomyocyte, and a pancreatic cell.
8. An isolated cell population that comprises cells according to claim 1.
9. The isolated cell population according to claim 8, wherein said cell population is nearly homogeneous.
10. A method for identifying a population of multipotent adult cells, wherein said population comprises isolated cells according to claim 1, the method comprising: (a) incubating the cells with labelled specific binding compounds for one or more characteristic markers for said population; and (b) detecting the presence or absence of binding by the cells to these specific binding compounds.
11. The method according to claim 10, wherein the specific binding compound is an antibody.
12. A method for isolating a population of multipotent adult cells according to claim 1, which comprises:
(a) collecting a non-osteochondral mesenchymal tissue;
(b) obtaining a cell suspension from the tissue by enzymatic digestion;
(c) incubating the cell suspension with a labelled compound that binds specifically to one or more of the surface makers of said multipotent adult cells; and
(d) selecting those cells that have the desired profile of expression of markers.
13. The method according to claim 12, wherein a negative selection is performed, whereby cells are excluded that show binding to labelled compounds that bind specifically to a marker selected from the group consisting of CD11b, CD14, CD15, CD16, CD31, CD34, CD45, CD49f, CD102, CD104, CD106 and CD133.
14. The method according to claim 12, wherein a positive selection is performed, whereby cells are selected that bind to labelled compounds that bind specifically to a marker selected from the group consisting of CD9, CD10, CD13, CD29, CD44, CD49a, CD51, CD54, CD55, CD58, CD59, CD90, and CD105.
15. The method according to claim 12, wherein the labelled compound of specific binding is an antibody.
16. A substantially homogeneous cell composition which comprises a multipotent adult cell according to claim 1.
17. A pharmaceutical composition that comprises a multipotent adult cell according to claim 1, and a pharmaceutically acceptable carrier.
18. An isolated multipotent adult cell according to claim 1 for use selected from the group consisting of: in therapy; and in the repair and regeneration of tissues.
19. (canceled)
20. (canceled)
21. A therapeutic method comprising administering a pharmaceutical composition according to claim 17 to a patient in need thereof.
22. Method according to claim 21, wherein said therapeutic method is for tissue repair or regeneration.
23. A method for assessing in vitro cell response to a biological or pharmacological agent, or to a combinatorial library of said agents, the method comprising:
a) isolating a cell population according to claim 8,
b) expanding the cell population via culturing, and
c) applying a biological agent or pharmacological agent or a combinatorial library of said agents to the cell population, and assessing effects of said agents on the cultured cells.
24. Method according to claim 23, wherein prior to step (c) the cells are allowed to differentiate into a specific type of cells.
US11/576,573 2004-10-04 2005-10-04 Identification and Isolation of Multipotent Cells From Non-Osteochondral Mesenchymal Tissue Abandoned US20070248580A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
ES200402355A ES2313805B1 (en) 2004-10-04 2004-10-04 IDENTIFICATION AND ISOLATION OF MULTIPOTENT CELLS OF NON-OSTEOCONDRAL MESENQUIMAL FABRIC.
ES200402355 2004-10-04
PCT/EP2005/010811 WO2006037649A1 (en) 2004-10-04 2005-10-04 Identification and insolation of multipotent cells from non-osteochondral mesenchymal tissue

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2005/010811 A-371-Of-International WO2006037649A1 (en) 2004-10-04 2005-10-04 Identification and insolation of multipotent cells from non-osteochondral mesenchymal tissue

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/834,006 Continuation US10729726B2 (en) 2004-10-04 2015-08-24 Identification and isolation of multipotent cells from non-osteochondral mesenchymal tissue

Publications (1)

Publication Number Publication Date
US20070248580A1 true US20070248580A1 (en) 2007-10-25

Family

ID=35457282

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/065,461 Abandoned US20060073124A1 (en) 2004-08-25 2005-02-25 Identification and isolation of multipotent cells from non-osteochondral mesenchymal tissue
US11/576,573 Abandoned US20070248580A1 (en) 2004-10-04 2005-10-04 Identification and Isolation of Multipotent Cells From Non-Osteochondral Mesenchymal Tissue
US14/834,006 Active 2025-12-28 US10729726B2 (en) 2004-10-04 2015-08-24 Identification and isolation of multipotent cells from non-osteochondral mesenchymal tissue

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/065,461 Abandoned US20060073124A1 (en) 2004-08-25 2005-02-25 Identification and isolation of multipotent cells from non-osteochondral mesenchymal tissue

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/834,006 Active 2025-12-28 US10729726B2 (en) 2004-10-04 2015-08-24 Identification and isolation of multipotent cells from non-osteochondral mesenchymal tissue

Country Status (17)

Country Link
US (3) US20060073124A1 (en)
EP (5) EP2862925A1 (en)
JP (2) JP2008515413A (en)
KR (1) KR20070085294A (en)
CN (1) CN101056974B (en)
AU (1) AU2005291353A1 (en)
CA (1) CA2583151C (en)
CY (1) CY1116198T1 (en)
DK (1) DK2292736T3 (en)
ES (2) ES2313805B1 (en)
HU (1) HUE025155T2 (en)
IL (1) IL182441A (en)
PL (1) PL2292736T3 (en)
PT (1) PT2292736E (en)
SG (2) SG158853A1 (en)
SI (1) SI2292736T1 (en)
WO (1) WO2006037649A1 (en)

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070020758A1 (en) * 2003-07-31 2007-01-25 Universita Degli Studi Di Roma "La Sapienza" Method for the isolation and expansion of cardiac stem cells from biopsy
US20080267921A1 (en) * 2004-11-08 2008-10-30 Johns Hopkins University Cardiac Stem Cells
US20100112694A1 (en) * 2006-11-09 2010-05-06 The Johns Hopkins University Dedifferentiation of Adult Mammalian Cardiomyocytes into Cardiac Stem Cells
US8834928B1 (en) 2011-05-16 2014-09-16 Musculoskeletal Transplant Foundation Tissue-derived tissugenic implants, and methods of fabricating and using same
US8883210B1 (en) 2010-05-14 2014-11-11 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US9249392B2 (en) 2010-04-30 2016-02-02 Cedars-Sinai Medical Center Methods and compositions for maintaining genomic stability in cultured stem cells
US9352003B1 (en) 2010-05-14 2016-05-31 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US9828603B2 (en) 2012-08-13 2017-11-28 Cedars Sinai Medical Center Exosomes and micro-ribonucleic acids for tissue regeneration
US9845457B2 (en) 2010-04-30 2017-12-19 Cedars-Sinai Medical Center Maintenance of genomic stability in cultured stem cells
US9884076B2 (en) 2012-06-05 2018-02-06 Capricor, Inc. Optimized methods for generation of cardiac stem cells from cardiac tissue and their use in cardiac therapy
US9943550B2 (en) 2005-09-23 2018-04-17 Tigenix, S.A.U. Cell populations having immunoregulatory activity, method for isolation and uses
US10092600B2 (en) 2013-07-30 2018-10-09 Musculoskeletal Transplant Foundation Method of preparing an adipose tissue derived matrix
US10130736B1 (en) 2010-05-14 2018-11-20 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US10531957B2 (en) 2015-05-21 2020-01-14 Musculoskeletal Transplant Foundation Modified demineralized cortical bone fibers
US10548924B2 (en) 2004-08-25 2020-02-04 Tigenix, S.A.U. Use of adipose tissue-derived stromal stem cells in treating fistula
US10729726B2 (en) 2004-10-04 2020-08-04 Tigenix, S.A.U. Identification and isolation of multipotent cells from non-osteochondral mesenchymal tissue
US10912864B2 (en) 2015-07-24 2021-02-09 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for preparing same
US11052175B2 (en) 2015-08-19 2021-07-06 Musculoskeletal Transplant Foundation Cartilage-derived implants and methods of making and using same
US11253551B2 (en) 2016-01-11 2022-02-22 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of heart failure with preserved ejection fraction
US11273182B2 (en) 2016-03-14 2022-03-15 Takeda Pharmaceutical Company Limited Adipose tissue-derived stromal stem cells for use in treating refractory complex perianal fistulas in Crohn's disease
US11351200B2 (en) 2016-06-03 2022-06-07 Cedars-Sinai Medical Center CDC-derived exosomes for treatment of ventricular tachyarrythmias
US11357799B2 (en) 2014-10-03 2022-06-14 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of muscular dystrophy
US11541078B2 (en) 2016-09-20 2023-01-03 Cedars-Sinai Medical Center Cardiosphere-derived cells and their extracellular vesicles to retard or reverse aging and age-related disorders
US11660355B2 (en) 2017-12-20 2023-05-30 Cedars-Sinai Medical Center Engineered extracellular vesicles for enhanced tissue delivery
US11660317B2 (en) 2004-11-08 2023-05-30 The Johns Hopkins University Compositions comprising cardiosphere-derived cells for use in cell therapy
US11759482B2 (en) 2017-04-19 2023-09-19 Cedars-Sinai Medical Center Methods and compositions for treating skeletal muscular dystrophy

Families Citing this family (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004069172A2 (en) 2003-01-30 2004-08-19 The Government of the United States of America as represented by the Department of Veterans Affairs Multilineage-inducible cells and uses thereof
CN100377165C (en) * 2003-04-24 2008-03-26 皇家飞利浦电子股份有限公司 Non-invasive left ventricular volume determination
US7266519B2 (en) * 2003-06-30 2007-09-04 Qualcomm Incorporated Billing system with authenticated wireless device transaction event data
CA2640185A1 (en) * 2006-01-24 2007-08-02 Christopher J. Centeno Mesenchymal stem cell isolation and transplantation method and system to be used in a clinical setting
US20080025953A1 (en) * 2006-07-25 2008-01-31 Kiminobu Sugaya Vigor Enhancement of Animals Via Administration of Stem Cells
KR100827660B1 (en) 2006-10-25 2008-05-07 대한민국 (식품의약품안전청장) Methods for selection and culture of mesenchymal stem cell expressing CD9
CA2668330C (en) * 2006-11-03 2016-02-02 Aastrom Biosciences, Inc. Mixed cell populations for tissue repair and separation technique for cell processing
FR2918073B1 (en) * 2007-06-27 2012-10-19 Univ Paris Curie CELLS FIXING TUMOR CELLS.
US9095562B2 (en) * 2007-07-05 2015-08-04 Regenerative Sciences, Inc. Methods and compositions for optimized expansion and implantation of mesenchymal stem cells
WO2009085969A2 (en) * 2007-12-19 2009-07-09 Regenerative Sciences, Llc Compositions and methods to promote implantation and engrafment of stem cells
CN102026546A (en) 2008-03-14 2011-04-20 再生科学有限责任公司 Compositions and methods for cartilage repair
GB0814249D0 (en) * 2008-08-04 2008-09-10 Cellerix Sa Uses of mesenchymal stem cells
AU2015268704B2 (en) * 2008-08-04 2017-08-10 Consejo Superior De Investigaciones Cientificas Uses of mesenchymal stem cells
AU2009284692B2 (en) 2008-08-18 2016-02-25 Mesoblast, Inc. Monoclonal antibody STRO-4
WO2010065854A1 (en) 2008-12-05 2010-06-10 Regenerative Sciences, Llc Methods and compositions to facilitate repair of avascular tissue
US20100168022A1 (en) * 2008-12-11 2010-07-01 Centeno Christopher J Use of In-Vitro Culture to Design or Test Personalized Treatment Regimens
US20110054929A1 (en) * 2009-09-01 2011-03-03 Cell Solutions Colorado Llc Stem Cell Marketplace
JP5810435B2 (en) * 2009-09-30 2015-11-11 国立大学法人 熊本大学 Method for detecting endoderm cells, intestinal cells or pancreatic cells
US9113950B2 (en) 2009-11-04 2015-08-25 Regenerative Sciences, Llc Therapeutic delivery device
JP6243839B2 (en) 2011-06-29 2017-12-06 バイオリストーラティブ セラピーズ, インコーポレイテッド Brown adipocyte composition and method
EP2634195B1 (en) * 2012-03-01 2017-02-01 Miltenyi Biotec GmbH Separation of living untouched neurons
WO2014172561A1 (en) 2013-04-19 2014-10-23 Biorestorative Therapies, Inc. Human brown adipose derived stem cells and uses
BR102013033827B1 (en) * 2013-12-27 2021-09-08 Regenera - Medicina Veterinária Avançada Ltda. MULTIPOTENT AND IMMUNOCOMPATIBLE CONCENTRATE OF STEM CELLS AND BIOPHARMACEUTICALS AND DOSAGE FORM THAT INCLUDE IT
EP3381458B1 (en) 2015-11-24 2023-06-07 Rohto Pharmaceutical Co., Ltd. Therapeutic agent for liver disease containing stromal cells derived from adipose tissue and method for producing the same
EP3385372B1 (en) * 2015-12-01 2023-08-09 AdipoSeeds, Inc. Method for manufacturing mesenchymal cell line derived from vertebrate animal adipose tissue
ES2890850T3 (en) * 2016-08-26 2022-01-24 Juno Therapeutics Inc Methods for enumerating particles present in a cellular composition
EP3417888A1 (en) * 2017-06-25 2018-12-26 co.don AG Method for producing transplantable cartilage tissue
CN111405898B (en) 2018-01-29 2022-09-13 中国科学院动物研究所 Method for destroying cell mechanical homeostasis and promoting tissue and organ regeneration and repair and application thereof
CN110090300A (en) * 2018-01-29 2019-08-06 中国科学院动物研究所 A kind of method and its application for the Regeneration and Repair promoting histoorgan
ES2957507A1 (en) * 2022-06-09 2024-01-19 Fundacion Instituto De Investig Sanitaria Fundacion Jimenez Diaz TREATMENT OF MEIBOMIAL GLAND DYSFUNCTION (Machine-translation by Google Translate, not legally binding)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030082152A1 (en) * 1999-03-10 2003-05-01 Hedrick Marc H. Adipose-derived stem cells and lattices
US20050019911A1 (en) * 1999-07-07 2005-01-27 Medvet Science Pty Ltd Mesenchymal precursor cell
US20050153442A1 (en) * 1999-03-10 2005-07-14 Adam Katz Adipose-derived stem cells and lattices
US20050244963A1 (en) * 2004-04-09 2005-11-03 Teplyashin Alexander S Method for obtaining mesenchymal stem cells
US20120027730A1 (en) * 2008-08-04 2012-02-02 Consejo Superior De Investigaciones Cientificas Uses of mesenchymal stem cells

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5811094A (en) 1990-11-16 1998-09-22 Osiris Therapeutics, Inc. Connective tissue regeneration using human mesenchymal stem cell preparations
US6294346B1 (en) 1991-07-08 2001-09-25 Neurospheres Holdings, Ltd. Use of multipotent neural stem cells and their progeny for the screening of drugs and other biological agents
JP2000508922A (en) 1996-04-26 2000-07-18 ケース ウエスターン リザーブ ユニバーシティ Skin regeneration using mesenchymal stem cells
WO1999011287A1 (en) 1997-09-04 1999-03-11 Osiris Therapeutics, Inc. Ligands that modulate differentiation of mesenchymal stem cells
JP4740452B2 (en) 1998-03-18 2011-08-03 オシリス セラピューティクス,インコーポレイテッド Methods, compositions and methods of using mesenchymal stem cells for the prevention and treatment of immune responses in transplantation
US5958767A (en) 1998-08-14 1999-09-28 The Children's Medical Center Corp. Engraftable human neural stem cells
KR100968165B1 (en) * 1999-03-10 2010-07-06 더 리전츠 오브 더 유니버시티 오브 캘리포니아 Adipose-derived stem cells and lattices
ATE384124T1 (en) * 2000-02-26 2008-02-15 Artecel Inc PLURIPOTENT STEM CELLS CREATED FROM ADIATIVE TISSUE STROMAL CELLS AND THEIR USES
CA2489212C (en) * 2002-06-14 2012-08-14 Cartela Ab Marker for stem cells and its use
AU2003278212B2 (en) * 2002-07-31 2009-07-09 Centre National De La Recherche Scientifique Stem cells derived from adipous tissue and differentiated cells derived from said cells
ES2265199B1 (en) * 2003-06-12 2008-02-01 Cellerix, S.L. MOTHER CELLS MULTIPOTENT ADULTS FROM DEDIFFERENTIATED DRIVERS AND ITS APPLICATIONS.
JP4749331B2 (en) * 2003-10-07 2011-08-17 株式会社バイオマスター Cell differentiation of adipose-derived progenitor cells
JP4217262B2 (en) * 2003-11-04 2009-01-28 株式会社バイオマスター Method and system for preparing stem cells from adipose tissue
JP2007530543A (en) 2004-03-22 2007-11-01 オシリス セラピューティクス,インコーポレイテッド Mesenchymal stem cells and use thereof
ES2313805B1 (en) 2004-10-04 2009-12-23 Cellerix, S.L. IDENTIFICATION AND ISOLATION OF MULTIPOTENT CELLS OF NON-OSTEOCONDRAL MESENQUIMAL FABRIC.
ES2264862B8 (en) 2004-08-25 2017-01-20 Cellerix, S.L. BIOMATERIAL FOR SUTURE.

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030082152A1 (en) * 1999-03-10 2003-05-01 Hedrick Marc H. Adipose-derived stem cells and lattices
US20050153442A1 (en) * 1999-03-10 2005-07-14 Adam Katz Adipose-derived stem cells and lattices
US20050019911A1 (en) * 1999-07-07 2005-01-27 Medvet Science Pty Ltd Mesenchymal precursor cell
US20050244963A1 (en) * 2004-04-09 2005-11-03 Teplyashin Alexander S Method for obtaining mesenchymal stem cells
US20120027730A1 (en) * 2008-08-04 2012-02-02 Consejo Superior De Investigaciones Cientificas Uses of mesenchymal stem cells

Cited By (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8846396B2 (en) 2003-07-31 2014-09-30 Universita Degli Studi Di Roma “La Sapienza” Methods for the isolation of cardiac stem cells
US8772030B2 (en) 2003-07-31 2014-07-08 Universita Degli Studi Di Roma “La Sapienza” Cardiac stem cells and methods for isolation of same
US20070020758A1 (en) * 2003-07-31 2007-01-25 Universita Degli Studi Di Roma "La Sapienza" Method for the isolation and expansion of cardiac stem cells from biopsy
US8268619B2 (en) 2003-07-31 2012-09-18 Universita Degli Studi Di Roma “La Sapienza” Method for the isolation and expansion of cardiac stem cells from biopsy
US10548924B2 (en) 2004-08-25 2020-02-04 Tigenix, S.A.U. Use of adipose tissue-derived stromal stem cells in treating fistula
US10780132B2 (en) 2004-08-25 2020-09-22 Tigenix, S.A.U. Use of adipose tissue-derived stromal stem cells in treating fistula
US10729726B2 (en) 2004-10-04 2020-08-04 Tigenix, S.A.U. Identification and isolation of multipotent cells from non-osteochondral mesenchymal tissue
US20080267921A1 (en) * 2004-11-08 2008-10-30 Johns Hopkins University Cardiac Stem Cells
US11660317B2 (en) 2004-11-08 2023-05-30 The Johns Hopkins University Compositions comprising cardiosphere-derived cells for use in cell therapy
US11672831B2 (en) 2005-06-24 2023-06-13 Takeda Pharmaceutical Company Limited Use of adipose tissue-derived stromal stem cells in treating fistula
US11660318B2 (en) 2005-06-24 2023-05-30 Takeda Pharmaceutical Company Limited Use of adipose tissue-derived stromal stem cells in treating fistula
US10758575B2 (en) 2005-06-24 2020-09-01 Tigenix, S.A.U. Use of adipose tissue-derived stromal stem cells in treating fistula
US9943550B2 (en) 2005-09-23 2018-04-17 Tigenix, S.A.U. Cell populations having immunoregulatory activity, method for isolation and uses
US20100112694A1 (en) * 2006-11-09 2010-05-06 The Johns Hopkins University Dedifferentiation of Adult Mammalian Cardiomyocytes into Cardiac Stem Cells
US9845457B2 (en) 2010-04-30 2017-12-19 Cedars-Sinai Medical Center Maintenance of genomic stability in cultured stem cells
US9249392B2 (en) 2010-04-30 2016-02-02 Cedars-Sinai Medical Center Methods and compositions for maintaining genomic stability in cultured stem cells
US9352003B1 (en) 2010-05-14 2016-05-31 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US8883210B1 (en) 2010-05-14 2014-11-11 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US10130736B1 (en) 2010-05-14 2018-11-20 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US11305035B2 (en) 2010-05-14 2022-04-19 Musculoskeletal Transplant Foundatiaon Tissue-derived tissuegenic implants, and methods of fabricating and using same
US8834928B1 (en) 2011-05-16 2014-09-16 Musculoskeletal Transplant Foundation Tissue-derived tissugenic implants, and methods of fabricating and using same
US9884076B2 (en) 2012-06-05 2018-02-06 Capricor, Inc. Optimized methods for generation of cardiac stem cells from cardiac tissue and their use in cardiac therapy
US10457942B2 (en) 2012-08-13 2019-10-29 Cedars-Sinai Medical Center Exosomes and micro-ribonucleic acids for tissue regeneration
US9828603B2 (en) 2012-08-13 2017-11-28 Cedars Sinai Medical Center Exosomes and micro-ribonucleic acids for tissue regeneration
US11220687B2 (en) 2012-08-13 2022-01-11 Cedars-Sinai Medical Center Exosomes and micro-ribonucleic acids for tissue regeneration
US11191788B2 (en) 2013-07-30 2021-12-07 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for preparing same
US11779610B2 (en) 2013-07-30 2023-10-10 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for using same
US10596201B2 (en) 2013-07-30 2020-03-24 Musculoskeletal Transplant Foundation Delipidated, decellularized adipose tissue matrix
US10092600B2 (en) 2013-07-30 2018-10-09 Musculoskeletal Transplant Foundation Method of preparing an adipose tissue derived matrix
US11357799B2 (en) 2014-10-03 2022-06-14 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of muscular dystrophy
US10531957B2 (en) 2015-05-21 2020-01-14 Musculoskeletal Transplant Foundation Modified demineralized cortical bone fibers
US11596517B2 (en) 2015-05-21 2023-03-07 Musculoskeletal Transplant Foundation Modified demineralized cortical bone fibers
US10912864B2 (en) 2015-07-24 2021-02-09 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for preparing same
US11524093B2 (en) 2015-07-24 2022-12-13 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for preparing same
US11806443B2 (en) 2015-08-19 2023-11-07 Musculoskeletal Transplant Foundation Cartilage-derived implants and methods of making and using same
US11938245B2 (en) 2015-08-19 2024-03-26 Musculoskeletal Transplant Foundation Cartilage-derived implants and methods of making and using same
US11052175B2 (en) 2015-08-19 2021-07-06 Musculoskeletal Transplant Foundation Cartilage-derived implants and methods of making and using same
US11253551B2 (en) 2016-01-11 2022-02-22 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of heart failure with preserved ejection fraction
US11872251B2 (en) 2016-01-11 2024-01-16 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of heart failure with preserved ejection fraction
US11273182B2 (en) 2016-03-14 2022-03-15 Takeda Pharmaceutical Company Limited Adipose tissue-derived stromal stem cells for use in treating refractory complex perianal fistulas in Crohn's disease
US11351200B2 (en) 2016-06-03 2022-06-07 Cedars-Sinai Medical Center CDC-derived exosomes for treatment of ventricular tachyarrythmias
US11541078B2 (en) 2016-09-20 2023-01-03 Cedars-Sinai Medical Center Cardiosphere-derived cells and their extracellular vesicles to retard or reverse aging and age-related disorders
US11759482B2 (en) 2017-04-19 2023-09-19 Cedars-Sinai Medical Center Methods and compositions for treating skeletal muscular dystrophy
US11660355B2 (en) 2017-12-20 2023-05-30 Cedars-Sinai Medical Center Engineered extracellular vesicles for enhanced tissue delivery

Also Published As

Publication number Publication date
JP2008515413A (en) 2008-05-15
DK2292736T3 (en) 2015-04-20
ES2313805A1 (en) 2009-03-01
JP2013013411A (en) 2013-01-24
PT2292736E (en) 2015-04-29
SG158853A1 (en) 2010-02-26
EP3342857A1 (en) 2018-07-04
EP2292736A2 (en) 2011-03-09
EP2292736A3 (en) 2011-06-15
EP2292736B1 (en) 2015-01-21
HUE025155T2 (en) 2016-03-29
CY1116198T1 (en) 2017-02-08
ES2535042T3 (en) 2015-05-04
PL2292736T3 (en) 2015-07-31
ES2313805B1 (en) 2009-12-23
EP2862925A1 (en) 2015-04-22
WO2006037649A1 (en) 2006-04-13
KR20070085294A (en) 2007-08-27
SG192459A1 (en) 2013-08-30
EP3165601A1 (en) 2017-05-10
SI2292736T1 (en) 2015-05-29
JP5732011B2 (en) 2015-06-10
CN101056974B (en) 2015-11-25
AU2005291353A1 (en) 2006-04-13
CN101056974A (en) 2007-10-17
US10729726B2 (en) 2020-08-04
IL182441A (en) 2015-08-31
CA2583151A1 (en) 2006-04-13
EP1812558A1 (en) 2007-08-01
IL182441A0 (en) 2007-07-24
US20170296585A1 (en) 2017-10-19
CA2583151C (en) 2015-09-15
US20060073124A1 (en) 2006-04-06

Similar Documents

Publication Publication Date Title
US10729726B2 (en) Identification and isolation of multipotent cells from non-osteochondral mesenchymal tissue
Romanov et al. Mesenchymal stem cells from human bone marrow and adipose tissue: isolation, characterization, and differentiation potentialities
Trivedi et al. Derivation and immunological characterization of mesenchymal stromal cells from human embryonic stem cells
Kramer et al. In vivo matrix-guided human mesenchymal stem cells
Krampera et al. Mesenchymal stem cells: from biology to clinical use
Chong et al. Human peripheral blood derived mesenchymal stem cells demonstrate similar characteristics and chondrogenic differentiation potential to bone marrow derived mesenchymal stem cells
Jackson et al. Mesenchymal progenitor cells derived from traumatized human muscle
US7915039B2 (en) Method for obtaining mesenchymal stem cells
US20020045260A1 (en) Method of isolating mesenchymal stem cells
US20140341863A1 (en) Adult mesenchymal stem cell (msc) compositions and methods for preparing the same
JP2003517259A (en) Method for isolating bone and precursor chondrocytes
Croft et al. Mesenchymal stem cells from the bone marrow stroma: basic biology and potential for cell therapy
AU2011253985B2 (en) Identification and isolation of multipotent cells from non-osteochondral mesenchymal tissue
KR20140016841A (en) A novel stem cell source derived from fetal cartilage tissue and cellular therapeutic agent
US20080299656A1 (en) Isolation of multi-lineage stem cells
Kadivar et al. Isolation, culture and characterization of postnatal human umbilical vein-derived mesenchymal stem cells
Umran et al. Comparative Study of Expansion and Proliferation of Adult Mice Mesenchymal Stem Cells Derived from Bone Marrow and Adipose Tissue
Singh et al. In vitro Culture and Morphometry of Porcine Adipose Derived Mesenchymal Stem Cells (pAD-MSCs)
Kramera et al. Research Article In vivo matrix-guided human mesenchymal stem cells

Legal Events

Date Code Title Description
AS Assignment

Owner name: CELLERIX, S.L., SPAIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GARCIA CASTRO, ROSA ANA;FERNANDEZ MIGUEL, MARIA GEMA;GONZALEZ DE LA PENA, MANUEL ANGEL;REEL/FRAME:019556/0503;SIGNING DATES FROM 20070507 TO 20070516

Owner name: UNIVERSIDAD AUTONOMA DE MADRID, SPAIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GARCIA ARRANZ, MARIANO;GARCIA OLMO, DAMIAN;REEL/FRAME:019556/0467

Effective date: 20070503

AS Assignment

Owner name: TIGENIX, S.A.U, SPAIN

Free format text: CHANGE OF NAME;ASSIGNOR:CELLERIX, S.A.;REEL/FRAME:030987/0894

Effective date: 20120131

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION