WO2000025134A1 - Assays for ligands for nuclear receptors - Google Patents

Assays for ligands for nuclear receptors Download PDF

Info

Publication number
WO2000025134A1
WO2000025134A1 PCT/US1999/024956 US9924956W WO0025134A1 WO 2000025134 A1 WO2000025134 A1 WO 2000025134A1 US 9924956 W US9924956 W US 9924956W WO 0025134 A1 WO0025134 A1 WO 0025134A1
Authority
WO
WIPO (PCT)
Prior art keywords
nuclear receptor
component
marking
binding domain
interaction
Prior art date
Application number
PCT/US1999/024956
Other languages
French (fr)
Inventor
Steven Gerard Blanchard
Derek J. Parks
Julie Beth Stimmel
Original Assignee
Glaxo Group Limited
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 Glaxo Group Limited filed Critical Glaxo Group Limited
Priority to AU12290/00A priority Critical patent/AU1229000A/en
Priority to JP2000578657A priority patent/JP2002528721A/en
Priority to EP99971090A priority patent/EP1137940A4/en
Publication of WO2000025134A1 publication Critical patent/WO2000025134A1/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
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/566Immunoassay; Biospecific binding assay; Materials therefor using specific carrier or receptor proteins as ligand binding reagents where possible specific carrier or receptor proteins are classified with their target compounds
    • 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
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/536Immunoassay; Biospecific binding assay; Materials therefor with immune complex formed in liquid phase
    • G01N33/542Immunoassay; Biospecific binding assay; Materials therefor with immune complex formed in liquid phase with steric inhibition or signal modification, e.g. fluorescent quenching
    • 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
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6875Nucleoproteins

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Cell Biology (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Microbiology (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Peptides Or Proteins (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

The present invention includes new nuclear receptor heterodimer and nuclear receptor-coactivator peptide assays for identifying ligands for nuclear receptors, utilizing scintillation proximity and fluorescence resonance energy transfer (FRET).

Description

ASSAYS FOR LIGANDS FOR NUCLEAR RECEPTORS
Cross reference to Related Applications
This application claims benefit under 35 U.S.C.119(e) of US provisional applications 60/150,390 filed 23 October 1998; 60/135,097 filed 23 December 1998; and 60/134,836 filed 19 May 1999.
Background of the Invention
Identification of ligands for orphan nuclear receptors has traditionally made use of either cell-based reporter gene assays or, in those cases where some ligand is known, competition binding assays. Both the transactivation and ligand-binding assays typically used to determine activity of putative nuclear receptor activators assess the effect of test ligands on an isolated receptor. The available evidence, however, indicates that a large proportion of the known orphan nuclear receptors, including the LXRs and FXR (1 ,2,8), function as heterodimeric receptor complexes with the common dimerization partner, RXR (reviewed in 6). Recently, it has been demonstrated that certain synthetic RXR ligands exhibit preferential activation of RXR heterodimers, and that this preference is determined by the receptor partner bound to RXR (7). These findings suggested the possibility that ligand binding to, and therefore subsequent activation of, nuclear receptors may be modulated by the receptor's dimerization state. Further, microscopic reversibility implies that ligand binding should modulate heterodimer affinity. Therefore, the ability of a ligand to induce changes in the degree of receptor dimerization could be used as the basis for a novel assay for the discovery of nuclear receptor ligands. FXR is an orphan nuclear receptor initially identified from a rat liver cDNA library (8) that is most closely related to the insect ecdysone receptor. The ligand binding domain of the receptor was cloned and expressed in support of an effort to develop a robust assay to identify a novel ligand. The availability of ligands for FXR will aid in the elucidation of the physiological role of this receptor. In addition, the information gained will further increase understanding of nuclear receptors as a target class.
For both FXR and LXR, a basal level of nuclear receptor-RXRα heterodimer formation is observed in the absence of added ligand. Ligands that promote heterodimer formation induce a concentration-dependent increase in time-resolved fluorescent signal. Compounds which bind equally well to both monomeric receptor and to the receptor: RXRα heterodimer would be expected to give no change in signal, whereas ligands which bind preferentially to the monomeric receptor would be expected to induce a concentration-dependent decrease in the observed signal.
Summary of the Invention
The present invention includes a generic approach to assay development for nuclear receptors, utilizing purified ligand binding domains. The concept of generic assay development has been extended to develop in vitro assays that detect ligand binding by monitoring ligand induced changes in receptor heterodimerization. This approach has been demonstrated using both scintillation proximity and homogenous time-resolved fluorimetry (HTRF) in the accompanying examples but it is not restricted to these methods. Other marking and measuring techniques may also be used. However, the use of scintillation proximity or HTRF provides a simpler and more practical methodology.
Another aspect of the invention is a new nuclear receptor-peptide assay for identifying ligands. This assay utilizes fluorescence resonance energy transfer (FRET) and was used to test whether putative ligands bound to FXR. The FRET assay is based upon the principle that ligands induce conformational changes in nuclear receptors that facilitate interactions with coactivator proteins required for transcriptional activation. In FRET, a fluorescent donor molecule transfers energy via a non-radiative dipole-dipole interaction to an acceptor molecule (which is usually a fluorescent molecule). FRET is a standard spectroscopic technique for measuring distances in the 10-70A range. Upon energy transfer, which depends on the R"6 distance between the donor and acceptor, the donor's fluorescence is reduced, and the acceptor fluorescence is increased, or sensitized. FRET is frequently used in both polymer science and structural biology and has recently been used to study macromolecular complexes of DNA, RNA, and proteins. In addition, Mathis has used europium cryptates with the multichromophoric Allophycocanin to achieve an extremely large R0 of 9θA Mathis, G. (1993) Clin. Chem. 39, 1953-1959.
Brief Description of the Drawings
Fig. 1. As shown in Fig 1 , ligand binding to LXRβ measured by modulation of LXRβ:RXR heterodimer formation.
Fig.2. Fig. 2 shows ligand binding to FXR measured by modulation of
FXR:RXR heterodimer formation.
Detailed Description of the Invention
In the assay of the present invention a method is provided for the rapid and simple determination of a ligand for a nuclear receptor which comprises contacting a component to be tested with an isolated nuclear receptor ligand binding domain which may be associated with a marking component, and a dimerization partner for the nuclear receptor ligand binding domain which is also associated with a marker; and measuring the interaction between the marking components to determine whether the component to be tested modifies heterodimerization. Various known markers may be used in the process of the present invention such as radioactive markers. The marker could also be a fluorescent dye. When the marker is radioactive, scintillation proximity may be used to measure the marker. When the marker used is a fluorescent dye, homogenous time-resolved fluorimetry may be used to detect the marker. Other known marking and measuring techniques may be used depending on the marker. However, the markers need to be in close proximity to indicate heterodimerization. That is, to indicate that the component to be tested functions as a ligand for the dimerization pair.
RXRs, such as RXRα, RXRβ, and RXRy, PPARs, such as PPARα, PPARy, and PPARδ, LXR α, β, ERα, ERβ, CAR α, HNF4 α,β,γ, NGFIB α,β,γ, PXR, PHR, EAR-1 , EAR-2, TR, RAR, and ERRs are examples of nuclear receptors that may be used as the dimerization partners and/or the nuclear receptors in the process of the present invention. RXRα is exemplified in some of the examples. Any nuclear receptor can be selected for use in an assay. More dimerization partners may be known or later discovered which can readily be utilized in the assay. It is preferable that the dimerization partners and the nuclear receptor ligand binding domains are recombinant proteins and preferably are bacterial expressed.
This method for the rapid determination of a ligand for a nuclear receptor comprises contacting a component to be tested with an isolated nuclear receptor ligand binding domain which is associated with a first marking component, and a heterodimeric partner for the nuclear receptor ligand binding domain associated with a second marking component, and measuring the interaction between the marking components to determine whether the component to be tested modifies hetero-dimerization.
The first marking component may be a radioactive marker and the second marking component (or second marker) may be a SPA bead. The interaction of the markers in this case is determined by scintillation proximity. Alternatively, the first marking component may be a first fluorescent dye emitting at an emitting wavelength which excites the second marking component which may be a second fluorescent dye. The interaction of the markers in this case is determined by homogenous time-resolved fluorimetry.
The interaction of the marking components in either case is measured by comparing a signal produced by a combination of the heterodimeric partner, the isolated nuclear receptor binding domain and the component to be tested with a signal produced by a combination of the heterodimeric partner and the isolated nuclear receptor ligand binding domain in the absence of the component to be tested. Example 1
This example describes the use of ligand mediated heterodimerization to quantify ligand binding to the nuclear receptor, Liver X Receptor (LXR). Liver X receptor alpha (LXRα) is an orphan nuclear receptor initially identified from a rat liver cDNA library (1 ). Human LXRα (2) and LXR β (3) have also been identified. The ligand binding domains of these receptors were cloned and expressed in support of an effort to develop a robust assay to identify a novel ligand. Oxysterols, including 24(S),25- epoxycholesterol have been identified as weak activators for these receptors (4,5). The availability of more potent and selective ligands for the LXRs may aid in the elucidation of the physiological role(s) of these receptors. In addition, the information gained will further increase understanding of nuclear receptors as a target class.
This example describes the use of ligand mediated heterodimerization to quantify ligand binding to the nuclear receptor Liver X Receptor beta (LXRβ). The method measures the ability of putative ligands to mediate the heterodimerization between the purified bacterial expressed LXRβ, and RXRα, ligand binding domains (LBD). Detection of the associated LBD's are measured by time resolved fluorimetry (TRF). The purified LBD of LXRβ is labeled with biotin then mixed with stoichiometric amounts of europium labeled streptavidin (Wallac Inc). The purified LBD of RXRα is labeled with CY5Tm. Equimolar amounts of each modified LBD are mixed together and allowed to equilibrate for at least one hour prior to the addition to either variable or constant concentrations of the sample for which the affinity is to be determined. After equilibration, the time-resolved fluorescent signal is quantitated using a fluorescent plate reader. The affinity of the test compound is estimated from a plot of fluorescence versus concentration of test compound added. A basal level of LXRβ:RXRα heterodimer formation is observed in the absence of added ligand. Ligands that promote heterodimer formation induce a concentration-dependent increase in time-resolved fluorescent signal. Compounds which bind equally well to both monomeric LXRβ and to the LXRβ:RXRα, heterodimer would be expected to give no change in signal, whereas ligands which bind preferentially to the monomeric receptor would be expected to induce a concentration-dependent decrease in the observed signal. METHODS & MATERIALS Advance Preparation:
Human LXRβ Ligand Binding Domain (LXR β LBD; Genbank accession number U 07132, amino acids 185-461 ) was expressed in E.coli strain BL21 (DE3) as an amino-terminal polyhistidine tagged fusion protein. Expression was under the control of an IPTG inducible T7 promoter. DNA encoding this recombinant protein and a modified polyhistidine tag was subcloned into the expression vector pRSETa (Invitrogen).
Ten-liter fermentation batches were grown in Rich PO4 media with 0.1 mg/mL Ampicillin at 25°C for 12 hours, cooled to 9°C and held at that temperature for 36 hours to a density of OD600=14. At this cell density, 0.25 mL IPTG was added and induction proceeded for 24 hours at 9°C, to a final OD600 =16. Cells were harvested by centrifugation (20 minutes, 3500g, 4°C), and concentrated cell slurries were stored in PBS at -8°C.
Purification of Receptor Ligand Binding Domain Routinely, 30-40 g cell paste (equivalent to 2-3 liters of the fermentation batch) was resuspended in 300-400 mL TBS, pH 8.5 (25mM Tris, 150 mM NaCI). Cells were lysed by passing three times through a homogenizer (Rannie) and cell debris was removed by centrifugation (30 minutes, 20,000g, 4°C). The cleared supernatant was filtered through coarse pre-filters, and TBS, pH 8.5, containing 500 mM imidazole was added to obtain a final imidazole concentration of 50mM. This lysate was loaded onto a column (6 x 8 cm) packed with Sepharose (Ni ++charged) Chelation resin (Pharmacia) and pre-equilibrated with TBS pH 8.5/ 50mM imidazole. After washing to baseline absorbance with equilibration buffer, the column was developed with a linear gradient of 50 to 275 mM imidazole in TBS, pH 8.5. Column fractions were pooled and dialyzed against TBS, pH 8.5, containing 5% 1 ,2-propanediol, 5mM DTT and 0.5mM EDTA. The protein sample was concentrated using Centri-prep 10K (Amicon) and subjected to size exclusion, using a column (3 x 90 cm) packed with Sepharose S-75 resin (Pharmacia) pre-equilibrated with TBS, pH 8.5, containing 5% 1 ,2-propanediol, 5mM DTT and 0.5mM EDTA. Biotinylation of LXRβ
Purified LXRβ, LBD was desalted/buffer exchanged using PD-10 gel filtration columns into PBS [100mM Na Phosphate, pH 7.2, 150 mM NaCI]. LXRβ LBD was diluted to approximately 10μM in PBS and five-fold molar excess of NHS-LC-Biotin (Pierce) was added in a minimal volume of PBS. This solution was incubated with gentle mixing for 30 minutes at room temperature. The biotinylation modification reaction was stopped by the addition of 2000x molar excess of Tris-HCI, pH 8. The modified LXR β LBD was dialyzed against 4 buffer changes, each of at least 50 volumes, PBS containing 5mM DTT 2mM EDTA and 2% sucrose. The biotinylated LXRβ LBD was subjected to mass spectrometric analysis to reveal the extent of modification by the biotinylation reagent. In general, approximately 95% of the protein had at least a single site of biotinylation; and the overall extent of biotinylation followed a normal distribution of multiple sites, ranging from one to nine.
RXRα LBD Human Retinoid X Receptor alpha Ligand Binding Domains: RXR-alpha LBD (amino acids 225-462) was expressed in E. coli strain BL21 (DE3) as an amino-terminal polyHistidine tagged fusion protein. Expression was under the control of an IPTG inducible T7 promoter. DNA encoding this recombinant protein and a modified histidine tag was subcloned into the expression vector pRSETa (Invitrogen). The sequence used in the construction of RXR-alpha LBD was derived from Genbank accession number X52773. Ten-liter fermentation batches were grown in Rich P04 media with 0.1 mg/mL Ampicillin at 25°C for 12 hours, cooled to 9°C and held at that temperature for 36 hours to a density of OD600 =14. At this cell density, 0.25 mM IPTG was added and induction proceeded for 24 hours at 9°C, to a final OD600 =16. Cells were harvested by centrifugation (20 minutes, 3500g, 4°C), and concentrated cell slurries were stored in PBS at -8°C.
Protein purification
Routinely, 40-50 g frozen cell paste (equivalent to 2-3 liters of the fermentation batch) was thawed and resuspended in 300mL TBS, pH 7.2, (25mM Tris, 150mM NaCI). Cells were lysed by three passages through a homogenizer (Rannie) and cell debris was removed by centrifugation (30 minutes, 20,000g, 4°C). The cleared supernatant was filtered through coarse pre-filters and TBS, pH 7.2, containing 500 mM Imidazole was added to obtain a final imidazole concentration of 50 mM. This lysate was loaded onto a column (3 x 8 cm) packed with Sepharose [Ni++ charged] Chelation resin (Pharmacia) and pre-equilibrated with TBS, pH 7.2, containing 50mM imidazole. After washing to baseline absorbance, the column was developed with a linear gradient of 50 to 500 mM imidazole in TBS, pH 7.2. Column fractions were pooled and dialyzed against TBS, pH 7.2, containing 5 mM DTT and .5mM EDTA. After dialysis the sample was concentrated using
Centri-prep 10K (Amicon) and subjected to size exclusion with a column (3 x 90 cm) packed with Sepharose S-75 resin (Pharmacia) pre-equilibrated with the same buffer.
Biotinylation of Human Retinoid-X Receptor Ligand Binding Domain Purified RXRα LBD biotinylation was carried out in a manner similar to that described for LXRβ LBD. Labeling of RXRα with CY5™
Purified RXRα LBD was diluted to approximately 10μM in PBS and approximately five-fold molar excess of Cy5™ monofunctional reactive dye [NHS ester] (Amersham Life Sciences) was added in a minimal volume of PBS. This solution was incubated in the dark with mixing for 30 minutes at ambient room temperature (approximately 23°C). The modification reaction was stopped by the addition of an excess of Tris-HCI, pH 8. Fluorescent dye modified RXRα LBD was dialyzed at 4°C, with minimal exposure to light, against 4 buffer changes, each of at least 50 volumes, PBS containing 5mM DTT, 2mM EDTA, and 2% (w/v) sucrose. Aliquots were frozen on dry ice and stored at -80°C.
Preparation of CY5™ -RXRα:Streptavdin-(Europium Chelate)-LXRβ Complex Equimolar concentrations of biotinylated LXRβ, and streptavidin- conjugated europium chelate were incubated in assay buffer containing 10 mM DTT for at least 10 minutes. To this solution equimolar concentrations of Cy5™ labeled RXRα was added and allowed to equilibrate for at least 30 minutes. The premixed receptor was then added in a one-step addition to the compound plate, utilizing e.g., a Titertek Multidrop 384.
Materials:
Cy5Tm RXRα and Europium labeled streptavidin LXRβ Complex
Assay Buffer: 50 mM KCI, 0.1 mg/mL BSA, 10 mM DTT and 50 mM Tris (pH 8) The stock buffer is made by dissolving 2.853g Tris base, 4.167 g Tris hydrochloride, 3.73 g KCI, and 0.1 g fatty acid free bovine serum albumin, in 1 L of deionized water. The pH is checked and adjusted to 8.0, if necessary, before adjusting to final volume. 0.154 g of solid DTT is added per 100 mL of buffer just before the start of an experiment.
BSA, fatty acid free DTT KCI
Europium labeled Streptavidin: (Wallac CR28-100) Tris Hydrochloride
96 well plates: polypropylene for intermediate dilutions (Costar #3794) and either a clear-bottomed white SPA plates (Costar #3632) or a black Polyfiltronics plate (UP350 PSB) for assays. Methods: Experimental Details:
Each well to be assayed contained a previously prepared solution of CY5 ™ RXRα and Europium labeled LXR and the desired concentration of test samples or controls (100μL total volume). In general, the total volume was held constant by varying the concentration and volume of premixed receptors to compensate for any changes in the volume of a particular set of samples. The plates were incubated for at least 2 hours at room temperature and the fluorescent signal determined in a Wallac Victor Multilabel Fluorescence Reader. Data Reduction:
For single concentration assays, the results of each test well were expressed as % of control, C, calculated according to eq. 1.
' sample " '"basal C = 100 * (1 )
F 1 std - F ' basal
where Fsample is the signal observed in a particular sample well, Ftota, is the signal observed in the presence of control inhibitor and Fbas_| is the count rate observed in the presence of no ligand. The values used for Fstd and Fbasa| were averages of the corresponding control wells included on every plate. For Dose response assays, the data were first normalized to % of control using eq. (1 ). A plot of CL, the % of control observed at ligand concentration L, versus ligand concentration, L was constructed. The data were fit to equation (2) to obtain best-fit parameters for the EC50, Fmax and ' basal-
F 1 max * L "- CL = F basal + (2)
EC50 + L
Note that Fmax, the maximal amplitude observed at saturating ligand concentrations, can be either a positive or negative value. The sign of this parameter indicates whether a particular test compound favors binding to the LXR:RXR complex (positive Fmax) or to either of the component receptors in a non-heterodimeric state (negative Fmax). Furthermore, both Fmax and Fbasa) are expressed in units of % of a standard compound.
RESULTS
Both the magnitude and sign of Fmax (the maximal ligand-induced amplitude) must be considered. Note that the definition of the maximal response observed for 24(S), 25-epoxycholesterol = 100% is an arbitrary assignment. The purpose of the normalization is only to allow comparison of values obtained for different compounds on different days and/or using different fluorescence plate readers. The results are shown in Figure 1.
Example 2
Determination of Ligand Binding to Famasoid X Receptor: Retinoid X Receptor Heterodimer utilizing Time Resolved Fluorimetry.
This example describes the use of ligand mediated heterodimerization to quantify ligand binding to the nuclear receptor Farnasoid X Receptor (FXR).
The method measures the ability of putative ligands to mediate the heterodimerization between the purified bacterial expressed FXR and RXRα ligand binding domains (LBD). Detection of the associated LBD's are measured by time resolved fluorimetry (TRF). The purified LBD of FXR is labeled with biotin then mixed with stoichiometric amounts of europium labeled streptavidin (Wallac Inc). The purified LBD of RXRα is labeled with CY5™ Equimolar amounts of each modified LBD are mixed together and allowed to equilibrate for at least 1 hour prior to the addition to either variable or constant concentrations of the sample for which the affinity is to be determined. After equilibration, the time-resolved fluorescent signal is quantitated using a fluorescent plate reader. The affinity of the test compound is estimated from a plot of fluorescence versus concentration of test compound added.
A basal level of FXR:RXRα heterodimer formation is observed in the absence of added ligand. Ligands that promote heterodimer formation induce a concentration-dependent increase in time-resolved fluorescent signal. Compounds which bind equally well to both monomeric FXR and to the FXR:RXRα heterodimer would be expected to give no change in signal whereas ligands which bind preferentially to the monomeric receptor would be expected to induce a concentration-dependent decrease in the observed signal. METHODS & MATERIALS Advance Preparation:
Human Famasoid X Receptor alpha Ligand Binding Domain Human FXRα Ligand Binding Domain (FXRα LBD) was expressed in E.coli strain BL21 (DE3) as an amino-terminal polyhistidine tagged fusion protein. Expression was under the control of an IPTG inducible T7 promoter. DNA encoding this recombinant protein was subcloned into the pRSET-A expression vector (Invitrogen). The coding sequence of Human FXRα LBD was derived from Genbank accession number U 68233 (amino acids 222 to 472). Ten-liter fermentation batches were grown in Rich P04 media with 0.1 mg/mL Ampicillin at 25°C for 12 hours, cooled to 9°C and held at that temperature for 36 hours to a density of OD600 =14. At this cell density, 0.25 mM IPTG was added and induction proceeded for 24 hours at 9°C, to a finalOD600 = 16. Cells were harvested by centrifugation (20 minutes, 3500g, 4°C), and concentrated cell slurries were stored in PBS at -8°C. Purification of Receptor Ligand Binding Domain
Routinely, 30-40 g cell paste (equivalent to 2-3 liters of the fermentation batch) was resuspended in 200-250 mL TBS, pH 7.2 (25mM Tris, 150 mM NaCI). Cells were lysed by passing 3 times through a French Press and cell debris was removed by centrifugation (30 minutes, 20,000g, 4°C). The cleared supernatant was filtered through course pre-filters, and TBS, pH 7.2, containing 500 mM imidazole was added to obtain a final imidazole concentration of 50mM. This lysate was loaded onto a column (6 x 8 cm) packed with Sepharose [Ni++charged] Chelation resin (Pharmacia) and pre- equilibrated with TBS pH 7.2/ 50mM imidazole. After washing to baseline absorbance with equilibration buffer, the column was washed with one column volume of TBS pH 7.2 containing 90mM imidazole. FXRαLBD was eluted directly with 365 mM imidazole. Column fractions were pooled and dialyzed against TBS, pH 7.2, containing 0.5mM EDTA and 5mM DTT. The dialyzed protein sample was concentrated using Centri-prep 10 K (Amicon) and subjected to size exclusion, using a column (3 x 90 cm) packed with Sepharose S-75 resin (Pharmacia) pre-equilibrated with TBS, pH 7.2, containing 0.5mM EDTA and 5mM DTT. Biotinylation of FXR Purified FXRα LBD was desalted/buffer exchanged using PD-10 gel filtration columns into PBS [100mM NaPhosphate, pH 7.2, 150mM NaCI]. FXRα LBD was diluted to approximately 10 μM in PBS and five-fold molar excess of NHS-LC-Biotin (Pierce) was added in a minimal volume of PBS. This solution was incubated with gentle mixing for 30 minutes at room temperature. The biotinylation modification reaction was stopped by the addition of 2000x molar excess of Tris-HCI, pH 8. The modified FXRα LBD was dialyzed against 4 buffer changes, each of at least 50 volumes, PBS containing 5mM DTT, 2mM EDTA and 2% sucrose. The biotinylated FXRα LBD was subjected to mass spectrometric analysis to reveal the extent of modification by the biotinylation reagent. In general, approximately 95% of the protein had at least a single site of biotinylation; and the overall extent of biotinylation followed a normal distribution of multiple sites, ranging from one to nine.
RXRα LBD
RXRα LBD was prepared and labeled with CY5 Tm in accordance with the procedures set forth in example 1.
Preparation of CY5 ™ -RXR:Streptavdin-(Eu.opium Chelate)-FXR Complex
Equimolar concentrations of biotinylated FXR and streptavidin- conjugated europium chelate were incubated in assay buffer containing 10 mM DTT for at least 10 minutes. To this solution an equimolar concentrations of Cy5™ labeled RXRα was added and allowed to equilibrate for at least 30 min. The premixed receptor was then added in a one-step addition to the compound plate, utilizing e.g., a Titertek Multidrop 384.
Materials: Assay Buffer: 50 mM KCI, 0. 1 mg/mL BSA, 10 mM DTT, and 50 mM Tris
(pH 8) The stock buffer is made by dissolving 2.853g Tris base, 4.167 g Tris hydrochloride, 3.73 g KCI, and 0. 1 g fatty acid free bovine serum albumin, in 1 L of deionized water. The pH is checked and adjusted to 8.0, if necessary, before adjusting to final volume. 0.154 g of solid DTT is added per 100 mL of buffer just before the start of an experiment.
BSA, fatty acid free
DTT
KCI
Europium labeled Streptavidin: (Wallac CR28-1 00) Tris Hydrochloride
96 well plates: polypropylene for intermediate dilutions (Costar #3794) and either a clear-bottomed white SPA plates (Costar #3632) or a black Polyfiltronics plate (UP350 PSB) for assays.
Methods: The methods, data reduction and interpretation of results are as described in example 1 and the results are shown in Figure 2. Example 3
A novel fluorescence resonance energy transfer (FRET) assay as described further infra, was used to test whether putative ligands bound to a nuclear receptor, using FXR as an example. The FRET assay is based upon the principle that ligands induce conformational changes in nuclear receptors that facilitate interactions with nuclear receptor coactivator proteins required for transcriptional activation. In an example, the ligand binding domain of FXR labeled with fluorophore allophycocyanin (APC) was incubated with a peptide containing the nuclear receptor interaction domain from the SRC-1 coactivator labeled with europium cryptate.
Both the transactivation and ligand-binding assays typically used to determine activity of putative nuclear receptor activators assess the effect of test ligands on isolated receptor. However, a large proportion of the known orphan nuclear receptors interact with cofactor or coactivator proteins as a complex. Therefore, the present invention suggests the possibility that ligand binding to nuclear receptors may be modulated by the receptor's complexation with a cofactor peptide. The ability of ligand to induce changes in the degree of this complex was then used as a basis for an inventive assay for the discovery of nuclear receptor ligands. Certain sequences of the cofactor may only be required to interact with the nuclear receptor. Various sequences of the two cofactor proteins SRC-1 and CBP were synthesized and tested in HTRF and Biacore to determine the best sequences to use. The peptide, CPSSHSSLTERHKILHRLLQEGSPS-CONH2 (SEQ ID NO.:1 ), i.e., SRC-1 (LCD2,676-700) was used in screening efforts with FXR and this forms a further aspect of this invention.
Coactivator proteins interact with nuclear receptors in a ligand- dependent manner and augment transcription (9). A short amphipathic α- helical domain that includes the amino acid motif LXXLL (L is Leu and X is any other amino acid) serves as the interaction interface between these coactivator molecules and the ligand-dependent activation function (AF-2) located in the COOH-terminus of the nuclear receptor LBD (10). To test whether ligands would induce a conformation of FXR that favors coactivator binding, a cell-free ligand-sensing assay utilizing fluorescence resonance energy transfer (FRET) to monitor allosteric interaction of a peptide based on the sequence of the steroid receptor coactivator 1 (SRC1 ) with the receptor was established. The use of FRET to monitor macromolecular complex formation is well established, particularly for immunoassays (11 ), and this detection methodology has recently been extended to characterize ligand binding to nuclear receptors (12).
Human FXR LBD was prepared and fluorescently labeled as described in Example 2. The LBD of human FXR was labeled with the fluorophore allophycocyanin and incubated with a peptide derived from the second LXXLL (SEQ ID NO.:1 ) motif of SRC1 (amino acids 676 to 700) that was labeled with europium chelate. The FRET ligand-sensing assay was performed by incubating 10 nM of the biotinylated FXR LBD that was labeled with streptavidin-conjugated allophycocyanin (Molecular Probes) and 10 nM of the SRC1 peptide [amino acids 676 to 700, 5'-biotin-
CPSSHSSLTERHKILHRLLQEGSPS-CONHJ (SynPEP) that was labeled with streptavidin-conjugated europium chelate (Wallac), in 50 mM Tris pH 8, 50 mM KCI, 0.1 mg/ml BSA, 1 mM EDTA, and 10 mM DTT, in the presence of test compound for 2 hours at 22°C. Data were collected using a Wallac Victor™ fluorescence reader in a time-resolved mode. The relative fluorescence was measured at 665nM and the data reduction was as described in Example 1. Preparation of Streptavidin-(Europium Chelate)-SRC1 :Steptavidin-(APC)-FXR Complex Biotinylated SRC-1 (LDC2,676-700) peptide and a Y2 stoichiometric amount of streptavidin-conjugated europium chelate were incubated in assay buffer containing 10mM DTT for at least 30 minutes. A second solution of stoichiometric amounts of biotinylated FXR and streptavidin-conjugated APC were incubated in assay buffer containing 10mM DTT for at least 30 minutes. Each solution was then blocked with a 5 fold molar excess of biotin and allowed to equilibrate for at least 15 minutes. The labeled receptor and labeled peptide were mixed and again allowed to equilibrate for at least 30 minutes, then added in a one-step addition to the compound plate, utilizing, e.g., a Titertek Multidrop 384. Materials: APC-labeled streptavidin FXR and Europium labeled streptavidin SRC- 1 (LDC2,676-700) SRC-1 (LDC2,676-700:(SynPEP) APC-Labeled streptavidin FXR and Europium labeled streptavidin
SRC1 (LCD2,676-700)
Biotinylated Human Farnasoid-X receptor LBD:
Biotinylated SRC-1 (LDC2,676-700):Biotin-CPSSHSSLTERHKILHRLL- QEGSPS-CONH2 (SynPEP) Assay Buffer: 50 mM KCI, 2mM EDTA, 0.1 mg/mL BSA, 10 mM DTT, and 50 mM Tris (pH 8). The stock buffer is made by dissolving 2.853g Tris base, 4.167 g Tris hydrochloride, 3.73 g KCI, 0.74 g EDTA (disodium salt, dihydrate) and 0.1 g fatty acid free bovine serum albumin, in 1 L of deionized water. The pH is checked and adjusted to 8.0, if necessary, before adjusting to final volume. 0.154 g of solid DTT is added per 100 mL of buffer just before the start of an experiment. BSA, fatty acid free DTT
EDTA, disodium salt dihydrate KCI
Allophycocyanin labeled streptavidin: (Molecular Probes S-868) Europium labeled Streptavidin: (Wallac CR28-100) Tris Hydrochloride
96 well plates: polypropylene for intermediate dilutions (Costar #3794) and either a clear-bottomed white SPA plates (Costar #3632) or a black Polyfiltronics plate (UP350 PSB) for assays.
RESULTS
Ligands increased the interaction between FXR and the SRC1 peptide as determined with time-resolved FRET. Dose response analysis showed that the ligands increased the amount of SRC1 peptide bound to the FXR
LBD. A typical saturable concentration response curve characteristic of specific interaction was observed.
As used in the claims for this assay, "nuclear receptor coactivator peptide" means a peptide whose affinity for the receptor is changed in the presence of ligand and which has a LXXLL motif. Examples of coactivator peptides useful for ligand identification by this method that have been demonstrated to interact with FXR include SRC-1 and those listed below:
1. B-QEQLSPKKKENNALLRYLLDRDDPS-CONH2 (SEQ ID NO.: 2), ACTR (734-758), RAC3 (724-748), SRC-3 (724-748), AIB1 (724-748), pCIP (716- 740)
2. B-QEPVSPKKKENALLRYLLDKDDTKD-CONH2 (SEQ ID NO.:3), TIF2 (732-756)
3. B-GSTHGTSLKEKHKILHRLLQDSSSPVD-CONH2 (SEQ ID NO.:4), TIF2
(676-702) 4. B-SNMHGSLLQEKHRILHKLLQNGNSPAE-CONH2 (SEQ ID N0..5), pCIP (664-690), RAC3 (671-697), ACTR (681-707), AIB1 (671-697)
The sequences of peptides 1 and 4 appear in a number of coactivators, hence the multiple names. The "B" in the sequences stands for biotinylated, which is a modification that allows attachment of the peptide during the analysis. The abbreviations used in the examples are included below.
ACTR Activator for Thyroid Hormone and Retinoid Receptors
AIB1 Amplified in Breast Cancer
APC Allophycocyanin APMSF p-Amidinophenylmethylsulfonylfluoride, HCI
Bestatin [(2S,3R)-3-Amino-2-hydroxy-4-phenylbutanoyl]-Leucine
BSA bovine serum albumin
CHAPS (3-[3-Cholamidopropyl)-dimethylammonio]-1-propanesulfonate
CPM counts per minute DMSO dimethylsulfoxide
DTT dithiothreitol
EDTA Ethylenediaminetetraacetic acid
FXR Farnasoid X Receptor
IBTGisopropyl-β-D-thiogalactopyranoside LBD ligand binding domain
LXR Liver X Receptor
OD600 optical density at 600 nm
PBS phosphate buffered saline [100mM NaPhosphate, pH 7.2, 50mM
NaCI] pCIP Co-Integrator Protein
RAC Receptor Activated Cofactor
RPM revolutions per minute
RXR Retinoid X Receptor
SA-APC Streptavidin Crosslinked Allophycocyanin SPA Scintillation Proximity Assay
SRC Steroid Receptor Cofactor
TIF Transcriptional Intermediary Factor
Tris tris-(Hydroxymethyl)-aminomethane The following references are noted and the entire disclosure of each is herein incorporated by reference
1. Apfel., R.H., Benbrook; D., Lernhardt, E., Ortiz, M.A., Salbert, G. And Pfahl, M. "A novel orphan receptor specific for a subset of TREs and its interaction with the retinoid/thyroid hormone receptor superfamily" Mol. Cell. Biol. 14, 7025-7035 (1994)
2. Willy PJ. Umesono K. Ong ES. Evans RM. Heyman RA. Mangelsdorf DJ. "LXR, a nuclear receptor that defines a distinct retinoid response pathway" Genes & Development. 9:1033-45 (1995)
3. Shinar, D.M., Endo.N., Rutledge, S.J., Vogel, R., Rodan, G.A. and Schmidt, A. "NER, a new member of the gene family encoding the human steroid hormone nuclear receptor" Gene 147, 273-276 (1994)
4. Janowski BA. Willy PJ. Devi TR. Falck JR. Mangelsdorf DJ. "An oxysterol signalling pathway mediated by the nuclear receptor LXR alpha" Nature.383: 728-31 , (1996)
5. Lehmann JM. Kliewer SA. Moore LB. Smith-Oliver TA. Oliver BB. Su JL. Sundseth SS. Winegar DA. Blanchard DE. Spencer TA. Willson TM. "Activation of the nuclear receptor LXR by oxysterols defines a new hormone response pathway" Journal of Biological Chemistry.272: 3137-40 (1997)
6. Mangelsdorf, D.J. and Evans, R.M. "The RXR Heterodimers and Orphan Receptors" Cell 83: 841-850 (1995)
7. Mukherjee R, Davies, P.J., Crombie, D.L., Bischoff, E.D., Cesario, R.M., Jow, L., Hamann, L.G., Boehm, M.F., Mondon, C.E., Nadzan, A.M., Paterniti Jr., J.R., and
Heyman R.A. Sensitization of diabetic and obese mice to insulin by retinoid X receptor agonists. Nature 386: 407-410 (1997)
8. Forman BM. Goode E. Chen J. Oro AE. Bradley DJ. Perlmann T. Noonan DJ. Burka LT. McMorris T. Lamph WW. et al "Identification of a nuclear receptor that is activated by farnesol metabolites" Cell 81 :687-693 (1995) 9. C. K. Glass, D. W. Rose, M. G. Rosenfeld, Curr. Opin. Cell Biol. 9, 222 (1997); D. Moras and H. Gronemeyer, Curr. Opin. Cell Biol. 10, 384 (1998).
10. B. Le Douarin et al., EMBO J. 15, 6701 (1996); D. M. Heery, E. Kalkhoven, S. Hoare, M. G. Parker, Nature 387, 733 (1997); G. Krey et al.,
Mol. Endochnol. 11 , 779 (1997).
11. E. Soini, I. Hemmila, P. Dahlen, Ann. Biol. Clin. 48, 567 (1990); E. F. Gudgin Dickson, A. Pollak, E. P. Diamandis, J. Photochem. Photobiol. 27, 3 (1995). 12. G. Zhou et al., Mol. Endochnol. 12, 1594 (1998); L. Paige et al., Proc.
Natl. Acad. Sci USA. 96, 3999 (1999).

Claims

We claim:
1. A method for the rapid determination of a ligand for a nuclear receptor which comprises contacting a component to be tested with an isolated nuclear receptor ligand binding domain which is associated with a first marking component, and a nuclear receptor coactivator peptide associated with a second marking component, and measuring the interaction between the marking components to determine whether the component to be tested modifies binding between the nuclear receptor ligand binding domain and the nuclear receptor coactivator peptide.
2. The method of claim 1 , wherein the first marking component is a radioactive marker and the second marking component is a SPA bead.
3. The method of claim 1 , wherein the first marking component is a first fluorescent dye emitting at an emitting wavelength which excites the second marking component which is a second fluorescent dye.
4. The method of claim 1 , wherein the nuclear receptor is Famesoid X Receptor ligand binding domain.
5. The method of claim 1 wherein the nuclear receptor coactivator peptide is SEQ ID NO.:1 , SEQ ID NO.:2, SEQ ID NO.:3, SEQ ID NO.:4, SEQ ID
NO.:5.
6. The method of claim 2, wherein the interaction of the markers is determined by scintillation proximity.
7. The method of claim 3, wherein the interaction of the markers is determined by homogenous time-resolved fluorimetry.
8. The method of claim 1 , wherein the interaction of the marking components is measured by comparing a signal produced by a combination of the nuclear receptor coactivator peptide, the isolated nuclear receptor binding domain and the component to be tested with a signal produced by a combination of the nuclear receptor coactivator peptide and the isolated nuclear receptor ligand binding domain in the absence of the component to be tested.
9. A method of identifying compounds for the treatment of diseases or disorders modulated by FXR, comprising the step of determining whether the compound interacts directly with FXR, wherein a compound that interacts directly with FXR is a compound for the treatment.
10. A method for the rapid determination of a ligand for a nuclear receptor which comprises contacting a component to be tested with an isolated nuclear receptor ligand binding domain which is associated with a first marking component, and a heterodimeric partner for the nuclear receptor ligand binding domain associated with a second marking component, and measuring the interaction between the marking components to determine whether the component to be tested modifies heterodimerization.
11. The method of claim 10, wherein the first marking component is a radioactive marker and the second marking component is a SPA bead.
12. The method of claim 10, wherein the first marking component is a first fluorescent dye emitting at an emitting wavelength which excites the second marking component which is a second fluorescent dye.
13. The method of claim 10 wherein the heterodimeric partner is an RXR, a PPAR, LXRα, LXRβ, ER α, ERβ, CAR α, an HNF4 , an NGFIB , PXR, PHR, EAR-1 , EAR-2, TR, RAR, ERRs or RAR.
14. The method of claim 11 , wherein the interaction of the markers is determined by scintillation proximity.
15. The method of claim 12, wherein the interaction of the markers is determined by homogenous time-resolved fluorimetry.
16. The method of claim 10, wherein the interaction of the marking components is measured by comparing a signal produced by a combination of the heterodimeric partner, the isolated nuclear receptor binding domain and the component to be tested with a signal produced by a combination of the heterodimeric partner and the isolated nuclear receptor ligand binding domain in the absence of the component to be tested.
PCT/US1999/024956 1998-10-23 1999-10-22 Assays for ligands for nuclear receptors WO2000025134A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
AU12290/00A AU1229000A (en) 1998-10-23 1999-10-22 Assays for ligands for nuclear receptors
JP2000578657A JP2002528721A (en) 1998-10-23 1999-10-22 Assays for nuclear receptor ligands
EP99971090A EP1137940A4 (en) 1998-10-23 1999-10-22 Assays for ligands for nuclear receptors

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US10539098P 1998-10-23 1998-10-23
US60/105,390 1998-10-23
US13509798P 1998-12-23 1998-12-23
US60/135,097 1998-12-23
US13483699P 1999-05-19 1999-05-19
US60/134,836 1999-05-19

Publications (1)

Publication Number Publication Date
WO2000025134A1 true WO2000025134A1 (en) 2000-05-04

Family

ID=27379915

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/024956 WO2000025134A1 (en) 1998-10-23 1999-10-22 Assays for ligands for nuclear receptors

Country Status (4)

Country Link
EP (1) EP1137940A4 (en)
JP (1) JP2002528721A (en)
AU (1) AU1229000A (en)
WO (1) WO2000025134A1 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002077229A2 (en) * 2001-03-01 2002-10-03 Lion Bioscience Ag Cofactors of the liver x receptor alpha and methods of use
US6639078B1 (en) 1998-12-23 2003-10-28 Smithkline Beecham Corporation Assays for ligands for nuclear receptors
US6987121B2 (en) 2002-04-25 2006-01-17 Smithkline Beecham Corporation Compositions and methods for hepatoprotection and treatment of cholestasis
US7115640B2 (en) 2001-12-21 2006-10-03 X-Ceptor Therapeutics, Inc. Heterocyclic modulators of nuclear receptors
US7138390B2 (en) 2001-03-12 2006-11-21 Intercept Pharmaceuticals Steroids as agonists for FXR
WO2010004052A2 (en) * 2008-07-11 2010-01-14 Medizinische Universität Innsbruck Agonists of nr2f6 for immunosuppression
US7994352B2 (en) 2005-05-19 2011-08-09 Intercept Pharmaceuticals, Inc. Process for preparing 3a(β)-7a(β)-dihydroxy-6a(β)-alkyl-5β-cholanic acid
US7998986B2 (en) 2001-12-21 2011-08-16 Exelixis Patent Company Llc Modulators of LXR
US8013001B2 (en) 2001-12-21 2011-09-06 Exelixis, Inc. Modulators of LXR
US8114862B2 (en) 2008-11-19 2012-02-14 Intercept Pharmaceuticals, Inc. TGR5 modulators and methods of use thereof
US8410083B2 (en) 2007-01-19 2013-04-02 Intercept Pharmaceuticals, Inc. 23-substituted bile acids as TGR5 modulators and methods of use thereof
US8568997B2 (en) 2006-11-10 2013-10-29 Dimerix Bioscience Pty Ltd. Detection system and uses therefor
WO2014028749A2 (en) 2012-08-15 2014-02-20 Boston Medical Center Corporation Production of red blood cells and platelets from stem cells
US8796249B2 (en) 2008-07-30 2014-08-05 Intercept Pharmaceuticals, Inc. TGR5 modulators and methods of use thereof
US9238673B2 (en) 2012-06-19 2016-01-19 Intercept Pharmaceuticals, Inc. Preparation and uses of obeticholic acid
US9314450B2 (en) 2011-01-11 2016-04-19 Dimerix Bioscience Pty Ltd. Combination therapy
US9498484B2 (en) 2004-03-12 2016-11-22 Intercept Pharmaceuticals, Inc. Treatment of fibrosis using FXR ligands
US9982008B2 (en) 2012-06-19 2018-05-29 Intercept Pharmaceuticals, Inc. Preparation and uses of obeticholic acid
US10987362B2 (en) 2004-03-12 2021-04-27 Intercept Pharmaceuticals, Inc. Treatment of fibrosis using FXR ligands

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4803976B2 (en) * 2003-07-09 2011-10-26 独立行政法人科学技術振興機構 Molecular sensor for intracellular IP3 measurement

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4568649A (en) * 1983-02-22 1986-02-04 Immunex Corporation Immediate ligand detection assay
US5670360A (en) * 1992-03-25 1997-09-23 Novo Nordisk A/S Mammalian receptors for glucagon-like-peptide-1 (GLP-1), corresponding DNA and recombinant expression systems, and screening assays for GLP-1 agonists and enhancers
US5776699A (en) * 1995-09-01 1998-07-07 Allergan, Inc. Method of identifying negative hormone and/or antagonist activities

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6005086A (en) * 1995-01-13 1999-12-21 The Salk Institute For Biological Studies Farnesoid activated receptor polypeptides, and nucleic acid encoding the same
CN1248916A (en) * 1997-01-24 2000-03-29 加利福尼亚大学董事会 Use of FXR, PPAR 'alpha' and LXR 'alpha' activators to restore barrier function, promote epidermal differentiation and inhibit proliferation
CA2305711A1 (en) * 1997-10-07 1999-04-15 Merck & Co., Inc. Assays for nuclear receptor ligands using fret
US6410245B1 (en) * 1998-04-01 2002-06-25 Affymax, Inc. Compositions and methods for detecting ligand-dependent nuclear receptor and coactivator interactions

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4568649A (en) * 1983-02-22 1986-02-04 Immunex Corporation Immediate ligand detection assay
US5670360A (en) * 1992-03-25 1997-09-23 Novo Nordisk A/S Mammalian receptors for glucagon-like-peptide-1 (GLP-1), corresponding DNA and recombinant expression systems, and screening assays for GLP-1 agonists and enhancers
US5776699A (en) * 1995-09-01 1998-07-07 Allergan, Inc. Method of identifying negative hormone and/or antagonist activities

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
MANGELSDORF D.J.: "The RXR Heterodimers and Orphan Receptors", CELL, vol. 83, 15 December 1995 (1995-12-15), pages 841 - 850, XP002922898 *
MUKHERJEE R.: "Sensitization of Diabetic and Obese Mice to Insulin by Retinoid X Receptor Agaonists", NATURE, vol. 386, 27 March 1997 (1997-03-27), pages 407 - 410, XP002922899 *
See also references of EP1137940A4 *

Cited By (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6639078B1 (en) 1998-12-23 2003-10-28 Smithkline Beecham Corporation Assays for ligands for nuclear receptors
US6984650B2 (en) 1998-12-23 2006-01-10 Smithkline Beecham Corporation Use of FXR ligands
WO2002077229A3 (en) * 2001-03-01 2003-12-04 Lion Bioscience Ag Cofactors of the liver x receptor alpha and methods of use
WO2002077229A2 (en) * 2001-03-01 2002-10-03 Lion Bioscience Ag Cofactors of the liver x receptor alpha and methods of use
US8058267B2 (en) 2001-03-12 2011-11-15 Intercept Pharmaceuticals, Inc. Steroids as agonists for FXR
US9732117B2 (en) 2001-03-12 2017-08-15 Intercept Pharmaceuticals, Inc. Steroids as agonists for FXR
US8969330B2 (en) 2001-03-12 2015-03-03 Intercept Pharmaceuticals, Inc. Steroids as agonists for FXR
US7138390B2 (en) 2001-03-12 2006-11-21 Intercept Pharmaceuticals Steroids as agonists for FXR
US10421772B2 (en) 2001-03-12 2019-09-24 Intercept Pharmaceuticals, Inc. Steroids as agonists for FXR
US7786102B2 (en) 2001-03-12 2010-08-31 Intercept Pharmaceuticals, Inc. Steroids as agonists for FXR
USRE48286E1 (en) 2001-03-12 2020-10-27 Intercept Pharmaceuticals, Inc. Steroids as agonists for FXR
US8377916B2 (en) 2001-03-12 2013-02-19 Intercept Pharmaceuticals, Inc. Steroids as agonists for FXR
US8013001B2 (en) 2001-12-21 2011-09-06 Exelixis, Inc. Modulators of LXR
US7998986B2 (en) 2001-12-21 2011-08-16 Exelixis Patent Company Llc Modulators of LXR
US7115640B2 (en) 2001-12-21 2006-10-03 X-Ceptor Therapeutics, Inc. Heterocyclic modulators of nuclear receptors
US6987121B2 (en) 2002-04-25 2006-01-17 Smithkline Beecham Corporation Compositions and methods for hepatoprotection and treatment of cholestasis
US9498484B2 (en) 2004-03-12 2016-11-22 Intercept Pharmaceuticals, Inc. Treatment of fibrosis using FXR ligands
US10987362B2 (en) 2004-03-12 2021-04-27 Intercept Pharmaceuticals, Inc. Treatment of fibrosis using FXR ligands
US10258633B2 (en) 2004-03-12 2019-04-16 Intercept Pharmaceuticals, Inc. Treatment of fibrosis using FXR ligands
US7994352B2 (en) 2005-05-19 2011-08-09 Intercept Pharmaceuticals, Inc. Process for preparing 3a(β)-7a(β)-dihydroxy-6a(β)-alkyl-5β-cholanic acid
US8568997B2 (en) 2006-11-10 2013-10-29 Dimerix Bioscience Pty Ltd. Detection system and uses therefor
US8410083B2 (en) 2007-01-19 2013-04-02 Intercept Pharmaceuticals, Inc. 23-substituted bile acids as TGR5 modulators and methods of use thereof
US9243027B2 (en) 2007-01-19 2016-01-26 Intercept Pharmaceuticals, Inc. TGR5 modulators and methods of use thereof
WO2010004052A3 (en) * 2008-07-11 2010-04-15 Medizinische Universität Innsbruck Agonists of nr2f6 for immunosuppression
WO2010004052A2 (en) * 2008-07-11 2010-01-14 Medizinische Universität Innsbruck Agonists of nr2f6 for immunosuppression
US8796249B2 (en) 2008-07-30 2014-08-05 Intercept Pharmaceuticals, Inc. TGR5 modulators and methods of use thereof
US9540414B2 (en) 2008-07-30 2017-01-10 Intercept Pharmaceuticals, Inc. TGR5 modulators and methods of use thereof
US9650409B2 (en) 2008-11-19 2017-05-16 Intercept Pharmaceuticals, Inc. TGR5 modulators and methods of use thereof
US8114862B2 (en) 2008-11-19 2012-02-14 Intercept Pharmaceuticals, Inc. TGR5 modulators and methods of use thereof
US8445472B2 (en) 2008-11-19 2013-05-21 Intercept Pharmaceuticals, Inc. TGR5 modulators and methods of use thereof
US10525038B2 (en) 2011-01-11 2020-01-07 Dimerix Bioscience Pty Ltd. Combination therapy
US9314450B2 (en) 2011-01-11 2016-04-19 Dimerix Bioscience Pty Ltd. Combination therapy
US11382896B2 (en) 2011-01-11 2022-07-12 Dimerix Bioscience Pty Ltd. Method for treating inflammatory disorders
US10058555B2 (en) 2011-01-11 2018-08-28 Dimerix Bioscience Pty Ltd. Combination therapy
US9982008B2 (en) 2012-06-19 2018-05-29 Intercept Pharmaceuticals, Inc. Preparation and uses of obeticholic acid
US9238673B2 (en) 2012-06-19 2016-01-19 Intercept Pharmaceuticals, Inc. Preparation and uses of obeticholic acid
US10174073B2 (en) 2012-06-19 2019-01-08 Intercept Pharmaceuticals, Inc. Preparation and uses of obeticholic acid
US10155787B2 (en) 2012-06-19 2018-12-18 Intercept Pharmaceuticals, Inc. Preparation and uses of obeticholic acid
US10047117B2 (en) 2012-06-19 2018-08-14 Intercept Pharmaceuticals, Inc. Preparation and uses of obeticholic acid
US9732116B2 (en) 2012-06-19 2017-08-15 Intercept Pharmaceuticals, Inc. Preparation and uses of obeticholic acid
WO2014028749A2 (en) 2012-08-15 2014-02-20 Boston Medical Center Corporation Production of red blood cells and platelets from stem cells
EP3789483A1 (en) 2012-08-15 2021-03-10 Boston Medical Center Corporation Production of red blood cells and platelets from stem cells

Also Published As

Publication number Publication date
JP2002528721A (en) 2002-09-03
AU1229000A (en) 2000-05-15
EP1137940A4 (en) 2004-06-02
EP1137940A1 (en) 2001-10-04

Similar Documents

Publication Publication Date Title
EP1137940A1 (en) Assays for ligands for nuclear receptors
EP1140079B1 (en) Assays for ligands for nuclear receptors
Tamrazi et al. Estrogen receptor dimerization: ligand binding regulates dimer affinity and DimerDissociation rate
Thomsen et al. Functional assays for screening GPCR targets
Chang et al. Evaluation of ligand-dependent changes in AR structure using peptide probes
Tamrazi et al. Molecular sensors of estrogen receptor conformations and dynamics
Liu et al. A homogeneous in vitro functional assay for estrogen receptors: coactivator recruitment
Legros et al. Characterization of the various functional pathways elicited by synthetic agonists or antagonists at the melatonin MT1 and MT2 receptors
Ozers et al. The androgen receptor T877A mutant recruits LXXLL and FXXLF peptides differently than wild-type androgen receptor in a time-resolved fluorescence resonance energy transfer assay
Weatherman et al. Ligand-selective interactions of ER detected in living cells by fluorescence resonance energy transfer
Stanley et al. Subtype specific effects of peroxisome proliferator-activated receptor ligands on corepressor affinity
EP1021462A1 (en) Assays for nuclear receptor ligands using fret
Berger et al. A PPARγ mutant serves as a dominant negative inhibitor of PPAR signaling and is localized in the nucleus
Kanayama et al. Basis of a high-throughput method for nuclear receptor ligands
US6689574B1 (en) Assays for nuclear receptor agonists and antagonists using fluorescence resonance energy transfer
Allikalt et al. Implementation of fluorescence anisotropy-based assay for the characterization of ligand binding to dopamine D1 receptors
Zhou et al. Use of homogeneous time-resolved fluorescence energy transfer in the measurement of nuclear receptor activation
Fujino et al. In vitro farnesoid X receptor ligand sensor assay using surface plasmon resonance and based on ligand-induced coactivator association
Gowda et al. Development of a coactivator displacement assay for the orphan receptor estrogen-related receptor-γ using time-resolved fluorescence resonance energy transfer
Cecon et al. Functional Investigation of Melatonin Receptor Activation by Homogenous cAMP Assay
Berbaum et al. Comparison of full-length versus ligand binding domain constructs in cell-free and cell-based peroxisome proliferator-activated receptor alpha assays
Murata et al. Piezo Electric Sensor for Endocrine-Disrupting Chemicals Using Receptor–co-factor Interaction
EP1876446B1 (en) Method for detection of substance bound to nuclear receptor
Awais et al. A fluorescent indicator to visualize ligand-induced receptor/coactivator interactions for screening of peroxisome proliferator-activated receptor γ ligands in living cells
Lee et al. Interactions of RXR with coactivators are differentially mediated by helix 11 of the receptor's ligand binding domain

Legal Events

Date Code Title Description
ENP Entry into the national phase

Ref country code: AU

Ref document number: 2000 12290

Kind code of ref document: A

Format of ref document f/p: F

AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US US US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 09807908

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 1999971090

Country of ref document: EP

ENP Entry into the national phase

Ref country code: JP

Ref document number: 2000 578657

Kind code of ref document: A

Format of ref document f/p: F

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1999971090

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1999971090

Country of ref document: EP