WO2006013109A1 - Method for testing a substance interacting with a target molecule - Google Patents

Method for testing a substance interacting with a target molecule Download PDF

Info

Publication number
WO2006013109A1
WO2006013109A1 PCT/EP2005/008510 EP2005008510W WO2006013109A1 WO 2006013109 A1 WO2006013109 A1 WO 2006013109A1 EP 2005008510 W EP2005008510 W EP 2005008510W WO 2006013109 A1 WO2006013109 A1 WO 2006013109A1
Authority
WO
WIPO (PCT)
Prior art keywords
target molecule
pattern
diffusion coefficients
cell
diffusion
Prior art date
Application number
PCT/EP2005/008510
Other languages
French (fr)
Inventor
Hanna Jankevics
Michael Prummer
Horst Vogel
Original Assignee
Epfl Swiss Federal Institute Of Technology-Lausanne
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
Application filed by Epfl Swiss Federal Institute Of Technology-Lausanne filed Critical Epfl Swiss Federal Institute Of Technology-Lausanne
Publication of WO2006013109A1 publication Critical patent/WO2006013109A1/en

Links

Classifications

    • 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
    • G01N33/5091Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing the pathological state of an organism
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N21/00Investigating or analysing materials by the use of optical means, i.e. using sub-millimetre waves, infrared, visible or ultraviolet light
    • G01N21/62Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light
    • G01N21/63Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light optically excited
    • G01N21/64Fluorescence; Phosphorescence
    • G01N21/6428Measuring fluorescence of fluorescent products of reactions or of fluorochrome labelled reactive substances, e.g. measuring quenching effects, using measuring "optrodes"
    • 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
    • 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
    • G01N33/502Chemical 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 for testing non-proliferative effects
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/72Assays involving receptors, cell surface antigens or cell surface determinants for hormones

Definitions

  • the present invention relates to a method for testing a substance interacting with a target molecule in a cell by fluorescence correlation spectroscopy (FCS).
  • the target molecule is a nuclear hormone receptor, e.g. the estrogen receptor.
  • US 2002/0072076 describes a determination of the interaction of a substance with a hormone receptor in a cell by FCS wherein alterations of the diffusion coefficient of a fluorescently labelled hormone receptor in the absence or in the presence of a test compound is determined.
  • the method allows determination whether or not the test substance is a substance interacting with the hormone receptor. The method, however, does not allow a clarification of the effect of the test substance on the hormone receptor.
  • the present invention provides a method for testing a substance interacting with a target molecule in a cell, comprising: a) providing a cell containing a target molecule carrying a fluorescent label, b) adding a test substance to the cell and determining a pattern or distribution of diffusion coefficients for the fluorescently labelled target molecule by FCS, c) optionally comparing the pattern of diffusion coefficients obtained in step
  • step (b) with a reference pattern/distribution and d) optionally classifying the effect of the test substance on the target molecule based on the pattern of diffusion coefficients obtained in step
  • the target molecule may be any molecule, which can carry a fluorescent label, e.g. a peptide, polypeptide or nucleic acid, e.g. DNA or RNA.
  • a fluorescent label e.g. a peptide, polypeptide or nucleic acid, e.g. DNA or RNA.
  • the target molecule is a polypeptide or protein, e.g. a hormone receptor such as the estrogen receptor (ER).
  • the target molecule may be located in the cytoplasm, in the nucleus, in an organelle or on and/or adjacent to a cell membrane.
  • the cell is preferably a living cell, e.g. a prokaryotic, or eukaryotic, e.g. an animal or plant cell. More preferably, the cell is a mammalian, e.g. human cell.
  • the target molecule carries a fluorescent label.
  • Fluorescent labels can be added externally and subsequently bound to the target molecule, e.g. chemical compounds like ligands which specifically interact with biological macromolecules or macromolecular complexes or macromolecular structures on the cell surface, inside the cell membrane, or inside of the cell.
  • cellular components might be labeled either chemically, biochemically, by expressing suitable mutant proteins, e.g. fusion proteins between the protein of interest and different fluorescent proteins such as GFP and YFP or, fusion proteins between the protein of interest and human alkylguanin-DNA-alkyltransferase which subsequently are selectively modified with a fluorescent label (Keppler et al.
  • labelled target molecules are fusion proteins of a fluorescent protein, e.g. GFP or YFP with nuclear hormone receptor.
  • fluorescent protein e.g. GFP or YFP with nuclear hormone receptor.
  • suitable labelled target molecules are:
  • YFP-labeled full length steroid receptor coactivator-3 SRC-3
  • [YFP-FL] YFP-labeled receptor interaction domain (RID) of SRC-3
  • [YFP-RID] YFP-labeled point mutated YFP-ER (not interacting with SRCs)
  • further test systems may comprise the testing of specific nuclear hormone receptor interaction partners such as DNA, co- activator proteins, co-repressor proteins, chaperone proteins, proteasomes and proteins involved in the general transcription machinery.
  • specific nuclear hormone receptor interaction partners such as DNA, co- activator proteins, co-repressor proteins, chaperone proteins, proteasomes and proteins involved in the general transcription machinery.
  • the present invention is based on the finding that the determination of diffusion coefficients for the fluorescently labelled target molecule by FCS in the presence of a test substance under defined test conditions in a heterogeneous environment gives a pattern or distribution of diffusion coefficients which is characteristic for the effect of the test substance on the target molecule.
  • the diffusion coefficient is preferably determined by comparing a measured intensity autocorrelation function with a model function assuming one or more, e.g. two diffusive components.
  • the determination of a pattern of diffusion coefficients requires a plurality of measurements. The measurements may be carried out under identical and/or different test conditions. In addition, a number of distributions may be determined under different test conditions, e.g.
  • test substance by varying the concentration of the test substance, physical parameters such as temperature, pressure, irradiation (e.g. UV, VIS 1 IR), stress (e.g. chemical or mechanical stress) or environmental conditions (e.g. medium composition, presence of chemical compounds or surfaces with particular composition and/or structure) or any combination thereof, and a part of the whole set of distributions of the target molecule may be used to assess the effect of a test substance.
  • physical parameters e.g. UV, VIS 1 IR
  • stress e.g. chemical or mechanical stress
  • environmental conditions e.g. medium composition, presence of chemical compounds or surfaces with particular composition and/or structure
  • the distribution of the diffusion coefficients for a test compound may be compared with one or several reference patterns or distributions of diffusion coefficients. These reference patterns may be obtained by determining a pattern of diffusion coefficients with a substance having an already known effect on the target molecule.
  • the present invention it may be determined, if a test substance binds or interacts with the target molecule at all. Additionally, the present invention allows a classification of the effect of the test substance on the target molecule, e.g. by determining the class of ligands the test compounds belong to.
  • the test compound may be. a partial or complete agonist or a partial or complete antagonist of the target molecule.
  • the method of the present invention is particularly useful for clinical diagnostics and high-throughput drug screening.
  • HEK293 Endothelial breast cancer cells
  • HEK293 Human Embryo Kidney cells 293
  • CHO Chinese Hamster Ovary cells
  • HEK293, HeLa, CHO and MCF-7 cells were regularly maintained in DMEM/F-12 with Glutamax-I (Invitrogen Corporation, UK) with 2.2 % Fetal Calf Serum in 5 % CO 2 incubator at 37C.
  • Glutamax-I Invitrogen Corporation, UK
  • 2.2 % Fetal Calf Serum in 5 % CO 2 incubator at 37C.
  • FCS measurements were performed using the Confocor2-LSM510 microscope (Zeiss, Jena, Germany) equipped with an C-Apochromat 40x 1.2 water immersion objective.
  • the YFP fluorescence was excited with a 488-nm line from an argon ion laser and detected through a pinhole with a diameter of 70 ⁇ m, and a 530-600 nm band pass filter. A laser power of below 3 kW/cm 2 for the FCS experiments.
  • the LSM510 was used to localize cells with a low expression level, and to position the laser beam for the FCS measurement in different areas of the cell.
  • the coordinates of the laser beam in the LSM510 images for different zooms and scan speeds were determined by recording the coordinates of the bleached spot seen on a cover glass slide containing fluorescent glue, after a short pulse of the FCS laser at maximum intensity settings.
  • the cover slide was a kind gift from Zeiss (Zeiss, Jena, Germany).
  • Rhodamine 6G Rho ⁇ G
  • PBS solution pH 7.4.
  • Rho ⁇ G diffusion was obtained as an average of 20 consecutive measurements, at the experimental conditions used for the subsequent study.
  • the confocal volume changed slightly for every measurement day, and in turn also the characteristic diffusion times. Therefore it was necessary to normalize the diffusion times retrieved in experiments made at different days to one standard rhodamine diffusion time. All diffusion times given in this work are scaled to a Rho ⁇ G diffusion time of 21 ⁇ s.
  • FCS data analysis In the data analysis the raw data are described by a suitable correlation function. To fit the correlation function to the raw data, an iterative procedure was performed with the Levenberg-Marquardt algorithm to minimize chi 2 .
  • the mean BF and the standard deviation of the mean were calculated for the different sample conditions and used in further evaluations. Normally, at least ten cells were investigated of the same experimental conditions.
  • the mean BF for the different ligands was plotted versus the ligand concentration to get dose-response curves. These dose-response curves were used to evaluate the potency of the different ligands to induce immobile ERs.
  • a half-maximal value of the effective concentration (EC50) for the bleaching was calculated from these curves for each ligand in order to separate their individual potency to immobilize the ER inside the nuclei.
  • "immobile” corresponds to the timescale of FCS, i.e. to those particles which move so slowly that their fluorophores are bleached during passage across the detection volume.
  • the autocorrelation functions of YFP chimeras were fitted to one- , two-, or three-component models of free 3D diffusion, plus two dark-states for all in vivo measurements, to derive the translational diffusion coefficients and the number of molecules diffusing through the confocal volume.
  • the second model will be preferred over the first.
  • the F-distribution can be found tabulated in a number of books, and it is also available from different software packages (e.g. Mathematica 4.2, Wolfram Research Inc., Champaign, IL, USA).
  • Weighted histograms are mean histograms that are constructed in the following manner: the amplitude of the bin in these weighted histograms is made by adding the fraction of each diffusion coefficient that falls into that bin. The fraction of the diffusion coefficient is retrieved from the 2P* fit.
  • Diffusion coefficient histograms at different ligand concentrations In order to compare the distributions of diffusion coefficients caused by the different ER ligands in vivo and to compare data from different ligand concentrations, the 2P* model with two dark-states was used. The reason for keeping the same model for all ligand concentrations was that the change of model will change the diffusion coefficients retrieved, and hence to be able to directly compare the data they all have to be fitted with the same model.
  • the first diffusive particle was kept fixed to increase the number of successful fits. This is due to the sensitivity of the data fitting procedure.
  • the fixed value was chosen based on a first run through the data leaving the two diffusion parameters free. This value was then kept the same for all ligand concentrations, because the choice of this value will affect the second diffusion coefficient as well.
  • Figure 1 shows a discrimination of different concentrations of an added ligand (E2) by comparison of the measured distributions of the diffusion coefficient.
  • Figure 2 shows a discrimination of different added ligands by comparing the measured distributions of the diffusion coefficient of the receptor.
  • Antagonists (ICI), partial agonists (4OHT), and agonists (E2, BPA) exhibit different diffusion patterns. Even though the partial agonist 4OHT acts as an antagonist in MCF-7 cells its diffusive behavior differs significantly from that of the full antagonist ICI. This detailed discrimination goes far beyond a pure detection of binding events (Fig. 2A ⁇ D). A determination of the ligand binding curves of the different ligands to the receptor from the bleaching (Fig. 2E) and the mobility distributions (Fig. 2F).
  • Figure 3 shows a discrimination of the effect of different ligands on the estrogen receptor and 2 different coactivator segments, RID and FL.

Abstract

The present invention relates to a method for testing a substance interacting with a target molecule in a cell by fluorescence correlation spectroscopy (FCS) by adding a test substance to the cell and determining a distribution of diffusion coefficients for the fluorescently labelled target molecule. Preferably, the target molecule is a nuclear hormone receptor, e.g. the estrogen receptor.

Description

Method for testing a substance interacting with a target molecule
Description
The present invention relates to a method for testing a substance interacting with a target molecule in a cell by fluorescence correlation spectroscopy (FCS). Preferably, the target molecule is a nuclear hormone receptor, e.g. the estrogen receptor.
US 2002/0072076 describes a determination of the interaction of a substance with a hormone receptor in a cell by FCS wherein alterations of the diffusion coefficient of a fluorescently labelled hormone receptor in the absence or in the presence of a test compound is determined. The method allows determination whether or not the test substance is a substance interacting with the hormone receptor. The method, however, does not allow a clarification of the effect of the test substance on the hormone receptor.
In order to overcome the deficiencies of the prior art, the present invention provides a method for testing a substance interacting with a target molecule in a cell, comprising: a) providing a cell containing a target molecule carrying a fluorescent label, b) adding a test substance to the cell and determining a pattern or distribution of diffusion coefficients for the fluorescently labelled target molecule by FCS, c) optionally comparing the pattern of diffusion coefficients obtained in step
(b) with a reference pattern/distribution and d) optionally classifying the effect of the test substance on the target molecule based on the pattern of diffusion coefficients obtained in step
(b).
The target molecule may be any molecule, which can carry a fluorescent label, e.g. a peptide, polypeptide or nucleic acid, e.g. DNA or RNA.
Preferably, the target molecule is a polypeptide or protein, e.g. a hormone receptor such as the estrogen receptor (ER). The target molecule may be located in the cytoplasm, in the nucleus, in an organelle or on and/or adjacent to a cell membrane. The cell is preferably a living cell, e.g. a prokaryotic, or eukaryotic, e.g. an animal or plant cell. More preferably, the cell is a mammalian, e.g. human cell.
The target molecule carries a fluorescent label. Fluorescent labels can be added externally and subsequently bound to the target molecule, e.g. chemical compounds like ligands which specifically interact with biological macromolecules or macromolecular complexes or macromolecular structures on the cell surface, inside the cell membrane, or inside of the cell.
Alternatively cellular components might be labeled either chemically, biochemically, by expressing suitable mutant proteins, e.g. fusion proteins between the protein of interest and different fluorescent proteins such as GFP and YFP or, fusion proteins between the protein of interest and human alkylguanin-DNA-alkyltransferase which subsequently are selectively modified with a fluorescent label (Keppler et al. 2003, Nature Biotechnol 21 , 86-89), by introducing non-natural fluorescent amino acids by suppressor t- RNA technology, by using biotinylated proteins which can be labeled with fluorescent avidin/streptavidin, by introducing fluorescent bases into DNA/RNA molecules, by using fluorescent complementary DNA segments to label DNA and/or by introducing fluorescent antibodies which bind to the protein, DNA, RNA, or organelles of interest.
Preferred examples of labelled target molecules are fusion proteins of a fluorescent protein, e.g. GFP or YFP with nuclear hormone receptor. Specific examples of suitable labelled target molecules are:
Yellow fluorescent protein-labeled estrogen receptor, [YFP-ER],
YFP-labeled full length steroid receptor coactivator-3 (SRC-3), [YFP-FL], YFP-labeled receptor interaction domain (RID) of SRC-3, [YFP-RID], YFP-labeled point mutated YFP-ER (not interacting with SRCs), [YFP-ER L539A]
In the experiments of the present invention, the effect of unlabelled steroid receptor coactivator-1 (SRC-1) and agonist on the mobility of YFP-ER and YFP-ERL539A was tested. Further, the effect of unlabelled ligands on YFP- ER, YFP-FL and YFP-RID was tested.
According to the invention, further test systems may comprise the testing of specific nuclear hormone receptor interaction partners such as DNA, co- activator proteins, co-repressor proteins, chaperone proteins, proteasomes and proteins involved in the general transcription machinery.
The present invention is based on the finding that the determination of diffusion coefficients for the fluorescently labelled target molecule by FCS in the presence of a test substance under defined test conditions in a heterogeneous environment gives a pattern or distribution of diffusion coefficients which is characteristic for the effect of the test substance on the target molecule. The diffusion coefficient is preferably determined by comparing a measured intensity autocorrelation function with a model function assuming one or more, e.g. two diffusive components. The determination of a pattern of diffusion coefficients requires a plurality of measurements. The measurements may be carried out under identical and/or different test conditions. In addition, a number of distributions may be determined under different test conditions, e.g. by varying the concentration of the test substance, physical parameters such as temperature, pressure, irradiation (e.g. UV, VIS1 IR), stress (e.g. chemical or mechanical stress) or environmental conditions (e.g. medium composition, presence of chemical compounds or surfaces with particular composition and/or structure) or any combination thereof, and a part of the whole set of distributions of the target molecule may be used to assess the effect of a test substance.
The distribution of the diffusion coefficients for a test compound (a compound for which the effect on the target molecule is unknown) may be compared with one or several reference patterns or distributions of diffusion coefficients. These reference patterns may be obtained by determining a pattern of diffusion coefficients with a substance having an already known effect on the target molecule.
According to the present invention, it may be determined, if a test substance binds or interacts with the target molecule at all. Additionally, the present invention allows a classification of the effect of the test substance on the target molecule, e.g. by determining the class of ligands the test compounds belong to. For example, the test compound may be. a partial or complete agonist or a partial or complete antagonist of the target molecule. Thus, the method of the present invention is particularly useful for clinical diagnostics and high-throughput drug screening.
Further, the present invention shall be explained in more detail by the following examples:
Examples
1. Methods
1.1 Sample preparations
Cell culture and transfection The different cell lines used in this study were Endothelial breast cancer cells (MCF-7), HeLa cancer cells, Human Embryo Kidney cells 293 (HEK293), and Chinese Hamster Ovary cells (CHO). HEK293, HeLa, CHO and MCF-7 cells were regularly maintained in DMEM/F-12 with Glutamax-I (Invitrogen Corporation, UK) with 2.2 % Fetal Calf Serum in 5 % CO2 incubator at 37C. Three days before measurements the medium was changed for D-MEM/F-12 without phenol red (Invitrogen Corporation, UK) with 2.2 % charcoal treated Fetal Calf Serum. Two days before measurements the cells were seeded in Lab-Tek chambered cover glass (Nalge Nunc International Corp, Naperville, IL, USA) at density of 10,000 cells/ml. The vectors were transiently expressed using Effectene transfection kit (QIAGEN GmbH, Germany).
1.2 FCS experiments
FCS setup
FCS measurements were performed using the Confocor2-LSM510 microscope (Zeiss, Jena, Germany) equipped with an C-Apochromat 40x 1.2 water immersion objective.
All measurements were performed at room temperature. The YFP fluorescence was excited with a 488-nm line from an argon ion laser and detected through a pinhole with a diameter of 70 μm, and a 530-600 nm band pass filter. A laser power of below 3 kW/cm2for the FCS experiments. The LSM510 was used to localize cells with a low expression level, and to position the laser beam for the FCS measurement in different areas of the cell. The coordinates of the laser beam in the LSM510 images for different zooms and scan speeds were determined by recording the coordinates of the bleached spot seen on a cover glass slide containing fluorescent glue, after a short pulse of the FCS laser at maximum intensity settings. The cover slide was a kind gift from Zeiss (Zeiss, Jena, Germany).
FCS measurements
Autocorrelation curves were derived from fluctuations of fluorescence intensity measured over 5-seconds intervals.
For the intracellular experiments, normally 19 measurements were taken per sample spot and nucleus. The first measurement was skipped due to possible bleaching effects. In the intranuclear FCS measurements, care was taken in choosing the measurement position to avoid the presence of large speckles and nucleosomes in the laser beam. Measurements that were directly on speckles showed a high degree of initial bleaching, indicating that the speckles are relatively immobile components. Hence, all diffusion complexes seen in this study, are those that are located in between the larger speckles, but they may still be interacting with the speckles or exchanging with partners in the speckles.
To evaluate the variance between measurements at different locations inside the nucleus, four measurements were performed at each location. The first measurement was skipped due to possible bleaching effects and an average autocorrelation curve of the three following measurements was evaluated for the variations to improve the statistics.
In a typical FCS experiment there were approximately 10,000 to 100,000 receptors inside the nucleus. The numbers were estimated based on the number of molecules retrieved in the confocal volume in the initial FCS experiment, compared to the initial fluorescence intensity signal keeping in mind the counts per molecule (CPM) of the fluorophores.
Calibration of FCS setup
To calibrate the FCS set-up and to enable the comparison of data measured at different days, at the start of each series of measurements a calibration measurement was done using Rhodamine 6G (RhoδG) in PBS solution at pH 7.4. The RhoδG diffusion was obtained as an average of 20 consecutive measurements, at the experimental conditions used for the subsequent study. The confocal volume changed slightly for every measurement day, and in turn also the characteristic diffusion times. Therefore it was necessary to normalize the diffusion times retrieved in experiments made at different days to one standard rhodamine diffusion time. All diffusion times given in this work are scaled to a RhoδG diffusion time of 21 μs.
A value of 280 μnfVs was used for the diffusion coefficient of RhoδG in PBS solution in all further calculations. Hence, to calculate a particular diffusion coefficient the corresponding diffusion time (in seconds) replaces τD D=(21μs*280 μnτ7s )/τD [μm2/s]
The same calibration method was used for in vitro and in vivo experiments assuming that the cellular structures will not change the confocal volume dramatically. The assumption was tested by measuring the diffusion time of Rhodamine 6G in PBS above cells growing on the cover slip; the same diffusion time was measured as for RhoθG in PBS buffer in the presence and absence of cells, indicating that the assumption is valid. This is in agreement with a literature report where no changes in diffusion time and spot size were detected in measurements done on the basal or apical side of cells, indicating that t:he passage of the light through the cell does not change the confocal volume (Licht et al. 2003, Biochemistry 42, 2916-25).
FCS data analysis In the data analysis the raw data are described by a suitable correlation function. To fit the correlation function to the raw data, an iterative procedure was performed with the Levenberg-Marquardt algorithm to minimize chi2.
Bleaching analysis of initial FCS experiment To estimate the amount of immobile molecules in the sample, the first intensity trace was fitted with a simple exponential decay. The amount of bleached molecules was calculated from the initial and the end point of the fit according to following relation: BF =((lrlΘ)/li)*100 [%], where BF is the bleached fraction, Ii the initial intensity, and le the end intensity of the fit.
The mean BF and the standard deviation of the mean were calculated for the different sample conditions and used in further evaluations. Normally, at least ten cells were investigated of the same experimental conditions. The mean BF for the different ligands was plotted versus the ligand concentration to get dose-response curves. These dose-response curves were used to evaluate the potency of the different ligands to induce immobile ERs. A half-maximal value of the effective concentration (EC50) for the bleaching was calculated from these curves for each ligand in order to separate their individual potency to immobilize the ER inside the nuclei. Here, "immobile" corresponds to the timescale of FCS, i.e. to those particles which move so slowly that their fluorophores are bleached during passage across the detection volume.
Diffusion analysis of FCS experiments
The autocorrelation functions of YFP chimeras were fitted to one- , two-, or three-component models of free 3D diffusion, plus two dark-states for all in vivo measurements, to derive the translational diffusion coefficients and the number of molecules diffusing through the confocal volume.
The dark-states were introduced to cover for the laser- and pH-induced photophysical phenomena in the YFP (Schwille et al. 2000, Proc Natl Acad Sci U S A 97, 151-6). Both dark-states were kept fixed during the fitting procedure to values (12 μs and 80 μs) retrieved from calibration measurements with YFP alone. The different models used have been described in more detail in general reviews (Krichevsky and Bonnet 2002, Reports on Progress in Physics 65, 251-297; Editor Lakowicz, J.R 1991, Topics in Fluorescence spectroscopy, Plenum Press). All data were primarily fitted to a one diffusing particle model (1P*) to get an indication of mobility changes under different conditions, since the 1P* model corresponds to a population average of the different diffusing components weighted by their relative fractions contributing to the autocorrelation curve.
As a second step, all data were fitted with a two diffusing particle model (2P*). In the 2P* model one diffusion coefficient was kept to a fixed value, to increase the number of successful fits. The fixed value was chosen by first letting the two diffusion components free and determining the average diffusion coefficient of the fast and slow component. Thereafter the fast component was fixed at 4-5 different values around the average diffusion coefficient and the value finally chosen was the one that minimized the oscillations of the fit residues. The choice between 1P* or 2P* model was made by F-test as described in the next section.
Goodness of fit with F-test
In a first step, a visual inspection of the residuals from the fit to avoid oscillating residues around the baseline was used as a check for the quality of the fit, the residues are defined as res=(y~yϊ)/σi.
Second, to compare two different fits and decide which of the two different models is the most appropriate one, the reduced chi2 (chi2r) was calculated according to chi2r= chi2/(n-p), where n is the number of points used, and p the number of free parameters in the fit and then used in the F-test (Bevington and Robinson 1992, Data reduction and error analysis for the physical sciences). Depending on the chosen confidence level and on chPr of the two models (chi2r1 , chi2r2), the F-distribution gives a limit for the ratio F = chi2r1/ chi2r2 if the two fits that can be assumed equally well for the data. If this ratio is higher than the given limit, the second model will be preferred over the first. The F-distribution can be found tabulated in a number of books, and it is also available from different software packages (e.g. Mathematica 4.2, Wolfram Research Inc., Champaign, IL, USA).
Creation of averaged diffusion coefficient histograms
In order to estimate the distributions from experimentally obtained histograms, and to avoid misleading peaks or shoulders arising from the choice of starting point of the histograms, the method of creating averaged histograms was chosen (Venables and Ripley 1997, Modern applied statistics with S-plus, Springer Verlag). An averaged histogram of 5 different realizations of the same data set was created. The five different realizations were made by changing the starting point of the binning by a fifth of the bin width, and then average over the 5 different realizations. The bin width was chosen to be at least two times the error in each data point.
When the data were analyzed with 2P* fits, the histograms are showing the free diffusion coefficient and the histograms are weighted histograms. Weighted histograms are mean histograms that are constructed in the following manner: the amplitude of the bin in these weighted histograms is made by adding the fraction of each diffusion coefficient that falls into that bin. The fraction of the diffusion coefficient is retrieved from the 2P* fit.
Diffusion coefficient histograms at different ligand concentrations In order to compare the distributions of diffusion coefficients caused by the different ER ligands in vivo and to compare data from different ligand concentrations, the 2P* model with two dark-states was used. The reason for keeping the same model for all ligand concentrations was that the change of model will change the diffusion coefficients retrieved, and hence to be able to directly compare the data they all have to be fitted with the same model.
The first diffusive particle was kept fixed to increase the number of successful fits. This is due to the sensitivity of the data fitting procedure. The fixed value was chosen based on a first run through the data leaving the two diffusion parameters free. This value was then kept the same for all ligand concentrations, because the choice of this value will affect the second diffusion coefficient as well.
Evaluation of ligand potency in affecting ER mobility
In order to estimate the different ligands1 efficacy in affecting the ERs mobility inside the nucleus a single parameter was wanted as an estimator of the effect on the mobility. Here, the choice fell on the diffusion coefficient from the 1P* model since it is a population average of the ensemble of molecules contributing to the auto-correlation function (ACF) and their individual contribution is weighted by their relative fraction. From these data the median of the distribution of diffusion coefficients was taken as a robust estimator of the distribution and to estimate the error in this estimation we used the standard deviation of the mean in lack of better options. These values were plotted for each ligand concentration to achieve dose-response curves for the different ligands, and the EC50 values of the different ligands were retrieved from the fits to these dose-response curves. 2. Results
Figure 1 shows a discrimination of different concentrations of an added ligand (E2) by comparison of the measured distributions of the diffusion coefficient.
Figure 2 shows a discrimination of different added ligands by comparing the measured distributions of the diffusion coefficient of the receptor.
Antagonists (ICI), partial agonists (4OHT), and agonists (E2, BPA) exhibit different diffusion patterns. Even though the partial agonist 4OHT acts as an antagonist in MCF-7 cells its diffusive behavior differs significantly from that of the full antagonist ICI. This detailed discrimination goes far beyond a pure detection of binding events (Fig. 2A~D). A determination of the ligand binding curves of the different ligands to the receptor from the bleaching (Fig. 2E) and the mobility distributions (Fig. 2F).
Figure 3 shows a discrimination of the effect of different ligands on the estrogen receptor and 2 different coactivator segments, RID and FL.
By comparing the effect of the different ligands not only on the mobility of the estrogen receptor but also on the mobility of coactivators, another dimension of information is added to discriminate between the. ligands.

Claims

Claims
1. A method for testing a substance interacting with a target molecule in a cell, comprising: a) providing a cell containing a target molecule carrying a fluorescent label, b) adding a test substance to the cell and determining a distribution or pattern of diffusion coefficients for the fluorescently labelled target molecule by FCS assuming one or more diffusive components, c) optionally comparing the pattern of diffusion coefficients obtained in step (b) with a reference pattern and d) optionally classifying the effect of the test substance on the target molecule based on the pattern of diffusion coefficients obtained in step (b).
2. The method according to claim 1 wherein the target molecule is selected from proteins, polypeptides, nucleic acids, e.g. DNA or RNA, and other molecules which can carry a fluorescent label.
3. The method according to claim 1 or 2, wherein the target molecule is a hormone receptor.
4. The method according to claim 1 or 3, wherein the target molecule is located in the cytoplasm, in the nucleus, in an organelle, or on and/or adjacent to a membrane.
5. The method of any one of claims 1 -4, wherein the distribution or pattern of diffusion coefficients is determined for different test conditions, e.g. the concentration of the test substance, physical parameters such as temperature, pressure, irradiation (e.g. UV, VlS, IR), stress (e.g. chemical or mechanical stress) or environmental conditions (e.g. medium composition, presence of chemical compounds or surfaces with particular composition and/or structure) or any combination thereof.
6. The method of any one of claims 1-5, wherein the reference pattern has been obtained by determining a pattern/distribution of diffusion coefficients with a substance having an already known effect on the target molecule.
PCT/EP2005/008510 2004-08-06 2005-08-05 Method for testing a substance interacting with a target molecule WO2006013109A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP04018731.2 2004-08-06
EP04018731 2004-08-06

Publications (1)

Publication Number Publication Date
WO2006013109A1 true WO2006013109A1 (en) 2006-02-09

Family

ID=35106877

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2005/008510 WO2006013109A1 (en) 2004-08-06 2005-08-05 Method for testing a substance interacting with a target molecule

Country Status (1)

Country Link
WO (1) WO2006013109A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2186827A1 (en) 2008-11-14 2010-05-19 HS LifeSciences Ltd. Surrogate marker directed cDNA cloning of selectively induced mRNAs
US7892756B2 (en) 2006-10-19 2011-02-22 Monell Chemical Senses Center Human salty taste receptor and methods of modulating salty taste perception

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030022224A1 (en) * 2001-07-19 2003-01-30 Olympus Optical Co., Ltd. Method of detecting binding reaction between protein and test substance
US6582907B1 (en) * 1999-12-09 2003-06-24 Pharmacia & Upjohn Company Use of fluorescence correlation spectroscopy to identify compounds that bind to target species under isothermal denaturing conditions
US20040142386A1 (en) * 1993-01-18 2004-07-22 Evotec Biosystems Gmbh Method and a device for the evaluation of biopolymer fitness

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040142386A1 (en) * 1993-01-18 2004-07-22 Evotec Biosystems Gmbh Method and a device for the evaluation of biopolymer fitness
US6582907B1 (en) * 1999-12-09 2003-06-24 Pharmacia & Upjohn Company Use of fluorescence correlation spectroscopy to identify compounds that bind to target species under isothermal denaturing conditions
US20030022224A1 (en) * 2001-07-19 2003-01-30 Olympus Optical Co., Ltd. Method of detecting binding reaction between protein and test substance

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
MODOS K ET AL: "Maximum-entropy decomposition of fluorescence correlation spectroscopy data: application to liposome-human serum albumin association", EUROPEAN BIOPHYSICS JOURNAL SPRINGER-VERLAG GERMANY, vol. 33, no. 1, February 2004 (2004-02-01), pages 59 - 67, XP002350974, ISSN: 0175-7571 *
N. KAHYA ET AL: "Lipid domain formation and dynamics in giant unilamellar vesicles explored by fluorescence correlation spectroscopy", JOURNAL OF STRUCTURAL BIOLOGY, vol. 147, July 2004 (2004-07-01), pages 77 - 89, XP002350975 *
WACHSMUTH MALTE ET AL: "Analyzing intracellular binding and diffusion with continuous fluorescence photobleaching.", BIOPHYSICAL JOURNAL, vol. 84, no. 5, May 2003 (2003-05-01), pages 3353 - 3363, XP002352320, ISSN: 0006-3495 *
WEIDEMANN T ET AL: "Counting Nucleosomes in Living Cells with a Combination of Fluorescence Correlation Spectroscopy and Confocal Imaging", JOURNAL OF MOLECULAR BIOLOGY, LONDON, GB, vol. 334, no. 2, 21 November 2003 (2003-11-21), pages 229 - 240, XP004470971, ISSN: 0022-2836 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7892756B2 (en) 2006-10-19 2011-02-22 Monell Chemical Senses Center Human salty taste receptor and methods of modulating salty taste perception
EP2186827A1 (en) 2008-11-14 2010-05-19 HS LifeSciences Ltd. Surrogate marker directed cDNA cloning of selectively induced mRNAs

Similar Documents

Publication Publication Date Title
JP4443407B2 (en) Detection of receptor oligomer formation
JP4511363B2 (en) FRET probe and method for detecting interacting molecules
Seuma et al. Combination of immunohistochemistry and laser ablation ICP mass spectrometry for imaging of cancer biomarkers
JP2002525603A (en) System for cell-based screening
JP2002520595A (en) System for cell-based screening
Lin et al. Rapid measurements of intracellular calcium using a fluorescence plate reader
AU2004267420A1 (en) Detecting and profiling molecular complexes
Hintersteiner et al. A two-channel detection method for autofluorescence correction and efficient on-bead screening of one-bead one-compound combinatorial libraries using the COPAS fluorescence activated bead sorting system
JP2004532978A (en) Automated method for detecting receptor activity
EP2700947B1 (en) Method for analyzing protein-protein interaction on single-molecule level within the cellular environment
US20140199704A1 (en) Multiplex cell signalling assays
Filby et al. The analysis of cell cycle, proliferation, and asymmetric cell division by imaging flow cytometry
WO2006013109A1 (en) Method for testing a substance interacting with a target molecule
JP2012223104A (en) Method for discriminating induction multipotent stem cell
Lugli et al. Polychromatic analysis of mitochondrial membrane potential using JC‐1
AU2005241246B2 (en) Method for characterising compounds
Kodippili et al. DARC, Glycophorin A, Band 3, and GLUT1 Diffusion in Erythrocytes: Insights into Membrane Complexes
Chatzimichail et al. Absolute quantification of protein copy number in single cells with immunofluorescence microscopy calibrated using single-molecule microarrays
Pick et al. Distribution plasticity of the human estrogen receptor α in live cells: distinct imaging of consecutively expressed receptors
CN110268108B (en) Methods and systems for screening using microcapillary arrays
Orlova et al. A nonfitting method using a spatial sine window transform for inhomogeneous effective-diffusion measurements by FRAP
JP4509702B2 (en) Method for measuring intracellular granular structure
JP2005207823A (en) Spectrofluorometric analyzing method
JP2009250652A (en) Screening method for functional material by measuring cell migration speed
Götte et al. Image-based high-content screening in drug discovery

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 69(1) EPC (EPO FORM 1205A DATED 29.06.07)

122 Ep: pct application non-entry in european phase

Ref document number: 05769779

Country of ref document: EP

Kind code of ref document: A1