CA2305810C - Methods of assaying receptor activity and constructs useful in such methods - Google Patents

Methods of assaying receptor activity and constructs useful in such methods Download PDF

Info

Publication number
CA2305810C
CA2305810C CA002305810A CA2305810A CA2305810C CA 2305810 C CA2305810 C CA 2305810C CA 002305810 A CA002305810 A CA 002305810A CA 2305810 A CA2305810 A CA 2305810A CA 2305810 C CA2305810 C CA 2305810C
Authority
CA
Canada
Prior art keywords
cell
gpcr
conjugate
test
protein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
CA002305810A
Other languages
French (fr)
Other versions
CA2305810A1 (en
Inventor
Lawrence S. Barak
Marc G. Caron
Stephen S. Ferguson
Jie Zhang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Duke University
Original Assignee
Duke University
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 Duke University filed Critical Duke University
Publication of CA2305810A1 publication Critical patent/CA2305810A1/en
Application granted granted Critical
Publication of CA2305810C publication Critical patent/CA2305810C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

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
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/60Fusion polypeptide containing spectroscopic/fluorescent detection, e.g. green fluorescent protein [GFP]
    • 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
    • G01N2333/726G protein coupled receptor, e.g. TSHR-thyrotropin-receptor, LH/hCG receptor, FSH
    • 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
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/91Transferases (2.)

Abstract

Described are methods of detecting G-protein coupled receptor (GPCR) activit y in vivo and in vitro; methods of assaying GPCR activity; and methods of screening for GPCR ligands, G protein-coupled receptor kinase (GRK) activity, and compounds that interact with components of the GPCR regulatory process. Constructs useful in such methods are described.

Description

METHODS OF ASSAYING RECEPTOR ACTIVITY AND
CONSTRUCTS USEFUL IN SUCH METHODS
FEDERALLY SPONSORED RESEARCH

This invention was made with support of the United States Government under National Institutes of Health Grant No. HL03422-02 and NS 19576. The United States Government has certain rights to this invention.

FIELD OF THE INVENTION

This invention relates to methods of detecting G-protein coupled receptor (GPCR) activity in vivo and in vitro, and provides methods of assaying GPCR
activity, and methods of screening for GPCR ligands, G-protein coupled receptor kinase (GRK) activity, and compounds that interact with components of the GPCR regulatory process.
This invention also provides constructs useful in such methods.

BACKGROUND OF THE INVENTION

The actions of many extracellular signals are mediated by the interaction of G-protein coupled receptors (GPCRs) and guanine nucleotide-binding. regulatory proteins (G proteins). G
protein-mediated signaling systems have been identified in many divergent organisms, such as mammals and yeast. GPCRs respond to, among other extracellular signals, neurotransmitters, hormones, odorants and light. GPCRs are similar and possess a number of highly -, -~-conservecl amino aeicls; tlle GPCRs are thought to represent a 1ar-ge 'superfamily' of proteins. lndividual GPCR types activate a particular signal transcltrction pathway; at least ten differ-ent signal transduction pathways are known to be activated via GPCRs. For example, the beta 2-adt=energic receptor (JiAR) is a pr-ototype manlmalian GPCR. In response to agonist binding, (iAR receptors activate a G protein (G) wllich in turn stinurlates adenylate cyclase and cvclic adenosine monophosphate production in the cell.

It has been postulated that niernbers of tlle GPCR superfamily clesensitize via a cclmnlon mechanism involving G protein-coupled receptor kinase (GRK) phosphorylation followed by arrestin tiinding. Gur-evicll et al., .1.
13io1.

Chenr. 270:720 (1995); Ferguson et al., Cari. .1 Plz),siol. Phorrnacol.
74:1095 (19%). i-Iowever, the localization and tlle snurce of the pool cif arrestin molecules targeted to r-eceptor-s in response to agonist activation was unknown.
Moreover, except for a Iiinitecl number of receptors, a conlmon role foT- ~-arrestin in GPCR descnsiti7atirnl had not been established. The rele of ~-arrestins in GPCR signal transductinn was postulated primarily due to the biochemical observatirnls.

Many available theralleutic drugs in use today target C;I'CRs, as thev mediate vital physiological re~sponses, incluciing vasodilation, heart rate, hronciloclilation, endocrine secretion, anci gut peristalsis. See, eg.., T
efkcnvitz et al., Arrn. IZei,,. 13roncen7. 52:159 (1983). GPCRs inclirde the adrenergic receptors (alpha and beta); (igands to beta ARs are used in the treatment of anaphylaxis, shoek, hvpcrtension, hypotension, asthma and clther conditions.
Aclclitionally, sprnltancous activation of Gl'CRs occurs, where a GPCR cellular response is generated in the absence of a ligand. tncreased spontaneous activity can be decreased by antagonists of tlle GPCR (a process k-iown as inverse agonism);
sucll nlethocis are therapeutically llllp(lrtant wllere diseases cause an increase in spontaneous (iPCR activity.

rfforts such as the lluman Genome Project are identifying new GPCRs ('orphan' receptors) whose pllysiological roles and ligands are unknown.
It is estimated that several thousand GPCRs exist in tiie human genonle. With , -~-Mnly about 10'Y,> of Ilie human genome sequenced. 250 (;PC'RS have heen iclcnti(ied; fewer than 150 have been associated with ligands.
SUMl\iIARY OF TIiE INVENTION

A(irst aspect of the present inventirnl is a conjugate of a(i-arrestin protein and a cletectahle mnlecttle. The detectable niolecule may he an optically detectable molecule, such as Green Pluorescent Protein.

A furtlier aspect of the present invention is a nucleic acid construct comprising an expression cassette. The construct includes, in the 5' to 3' c{irection, a promoter and a nucleic acid segment operatively associated willi tlie promotet-, and the tiucleic acicl segment encocles a~-arrestin protein and detectahle molecule. The detectable molecule mav he an optically detectable molccule such as Green 1-luo--esce.nt ['rotcin.

A further aspect of the present invention is a host cell containing a nucleic acid molecule which includes, a promoter operable in the host cell anc!
a nucleic acid sequence encoding aP-arrestin protein and a detectable niolect.tle.

The detectahle molecule may he an optically detectable molecule such as Green f luorescent I't-otein. Tlie cell may be a matnnlalian, hacterial, yeast, fUngal, plant or aninial cell, and may he deposited on a suhstrate.

A furtlier aspect of the present invention is a niethod of assessing G protein couplecl receptor (GPCR) pathway activity under test conditions, by providing a test cell that expresses a GPCR and that contains a conjugate of a(3-arrestin protein and a visually detectable molecule; exposing the test cell to a knowti GPCR agonist uncler test conditions; and then detecting translocation of the detectable tiiolecule frotn the cytosol of the test cell to the menihrane ecige of the test cell. Tt-anslocation of the detectable molecuie in the test cell inclicates activation of the G('CR pathway. Exemplary test conciitions include the presence in the test cell of a test kinase and/or a test G-protein, or exposnre of the test cell to a test ligancl, or co-expression in the test cell of a second receptor.

A furthet- aspect of the present itivention is a metiiod for screening a(3-at-restin protein (or fragment of a(3-arrestin protein) for the ability tci bind to a phosphorylatecl GPCR. A cell is provicied that expresses a GPCR ancl conlains a conjugate of a test P-arrestin protein and a visually detectable nlolccule.
T'he cell is exposed to a known GPCR agonist ancl then transiocation of the detectable nlolectile from the cell cytosol to tlle cell ecige is cletected.
Translocation of the detectable nlolecule indicates that the (3-arrestin molectile can bind to phosphorylated GPCR in the test cell.
A further aspect of the present inventirni is a nletllod to screen a test comliound for G protein cottpled i-eceptor (GPCR) agonist activity. A
test cell is provided that expresses a GPCR and contains a conjugate of aP-arrestin protein and a visually detectable molecuie. The cell is exposed to a test compounci, and translocation of the detectable molecule froni the cell cytosol to the membrane ecige is cletected. Movenlent of the detectable moiecule to the memb--ane edge after exposure of the cell to the test conlpotllld indicates GPCR
agonisf activitv of the test compound. The test cell nlay express a known GPCR
ot- a variety of known GPCRs, or express an unknoNvn GPCR or a variety of unknown GPCRs. 'rhe GPCR may be, for exanlple, an odorant GPCR or a(3-adrenergic GI'CR. The test cell may be a mammalian, bacterial, yeast, fungal, plant ot' anlnlal cell.

A ftirtller aspect of the present inventicln is a method of screening a sanlple soluticln for the presence of an agonist to a G protein coupled receptor (GPCR). A test cell is provided that expresses a GPCR and contains a conjugate of aP-ari-estin protein and a visualfy detectable nlolecttle. The test cell is exposed to a sample solution, and translocatiotl of the detectable nlolecule fi-om the cell cytosol to the membrane edge is assessed. lviovement of tlle detectable molecufe to the menlbrane edge after expostire to the sanlple solttticln indicates the sample solution cotltafns an agonist fot- a GPCR expressed in tlle cell.
A furthe-- aspect of the present invention is a metliod of screening a test compound for G protein coupled receptor (GPCR) antagonist activity. A
cell is provided that expresses a GPCR and contains a cenjugate of aP-arrestin protein and a visually detectable nlolecuie. The cell is exposed to a test conlpound and to a GPCR agonist, and translocation of the detectable tnolecule fronl the cell evtosol to the nlenlbrane edge is detected. Wllen exposui-e to the agonist occurs at the sanle time as or sttlisequent to exposure to tlle test ~-crnnpouncl, movement of the detectable molecule frrnn tllc cvtosol to the membrane edge after exposure to the test compound indicates that the test compound is not a GPCR antagonist.

A furtller aspect of the present invention is a method of screening a test compound for G protein coupleci receptor (GPCR) antagonist activity. A
lest cell is provideci that expresses a GPCR and contains a conjuglte of ap-arrestin protein and a visually detectable moiecule. 'Tlie cell is exposed to a Gl'CR agonist so that translocation of tlie detectable molecr.rle from the cytosol of the cell to the membrane edge of the cell occurs, and the cell is then exposed to a test comrcnrnd. Wliere exposure to the agonist occurs prior to exposure to the tesl compound, movement of the detectable molecule froni the membrane edge of the cell to the cytosol after expostn-e of the cell to ilie lest comporrnd indicates that the test compouncl has GPCR antagonist activity.

A furtlier aspect of the pr-esent invention is a metlzod of screening a cell for- the rresence of a G protein conpled receptor (GPCR). A test cell is provicled that contains a conjugate of aP-arrestin protein and a visnaliv detectable molecule. Tlie test cell is exposeci to a solution containing a GPCR agonist.
nny translocation of the detectable molecule from the cvtosol to the memhrane eclge is detected; movement of the detectable molecule from tlhe cytosol to tlie memhr-ane ecige after exposure of tlie test cell to CTPCR agrniist indicates that the test cell contains a GPCR.
A further aspect of the present invention is a nietlioci of screening a plurality of cells for those cells which contain a G protein coupled receptor (GPCR). A plurality of test cells containing a conjugate of aP-arrestin prolein and a visually detectable niolecule are provided, and the test cells are exposed to a known GPCR agonist. Cells in wliich the detectable molecule is translocated fi-om the cvtosol to the memhrane edge are identified or cle.tected. Movernent of the detectable molecule to the membrane edge after exposure to a GPCR agonist indicates that the cell contains a GPCR responsive to that GPCR agonist. Tile plurality of test cells may be contained in a tissue, an organ, or an intact animal.
A furtlier aspect of the present invention is a suhstrate having clepositecl tiiereon a plurality of cells that express a GPCR and that contain a -G-conjugate of a[3-arrestin protein and a cietectable molecule. Such substrates may he made of glass, plastic. ceramic, semicondttctor, silica, fiber optic, diamond, hiocompatihle nirnlomer, or hiocoinpatible polymer rnaterials.

A fttrther aspect of the present invention is an apparatus for determining GPCR activity in a test cell. The apparatus inclucles means for measuring indicia of the intracellular distribution of a detectable molecule, and a computer rrogram product that includes a computer readable storage meditmi having computer-readable program code means embodied in the meclium. The computer-readahle program cocle means includes computer-readable program code means for detet-mining whetlier the indicia of the distribtttion of the detectable molecule in a test cell indicates concetltration of the detectable molecule at the cell membrane, haseci oti comparison to the measured indicia of the intracellular distrihtttion of a detectable molecule in a control cell. The indicia of the intraeellirlar- clistrihution of the detectabie molecule may be optical inclicia. and the mensuring means may he means for measuring fluorescent in(ensity. The mnlecule to be detected may be one that is fluorescently detectable, and the step of ineasuring the indicia of tlle intracellular distribtrtion of the detectable molecttle may include measurement of fluorescence signals fi'on'1 test and conti'(lI
cells.

A furtlier aspect of the present invention is an apparatus for deterniining c;=PCR activity in a test cell. The apparatus inciudes means for measuring indicia of the intracellular distribution of a detectable molecule in at least one test cell at multilile time points, and a computer program procluct.
The computer program product incindes a computer readable storage inediurli having computer-readahle program cocle means embodied in said medium. The computer-readable program code rneans includes computer-readable Program code means for cletei-mining wltetller the indicia of the distribution of the cfc(ectable molecule in the test cell at mtrltiPle time points inclicates translocation of the detectahie molecule to the cell menibrane.
A further aspect of the present invention is an apparatus for determining GPCR activity in a test cell, wbich includes means for measuring indicia of tlie intr-acellular- clistribution of a detectable molecuie in at least one test WO 98/55635 PCT/US98/1 ] 628 cell, and a computer program product. TSie compttter program product inclucles a cnmpttter re<tcfahle storage mediuln having coniputer-reaclable program code means embodied tllerein and iticluding computer-readable firogram code mcans for cletermining wliethcr the indicia of the distrihution of the cletectahle niolecufe in tiie test cell indicates concetitration of the detectable molecule at tiie cell membrane, hased on comparison to pre-established criteria.

13RIrF i)ESCRiPTION OF THC, DRAWINGS

Figure I is a linear model of the (i-arrestin2/S65T-Green Flunresccnt I'rolcin (6IT) cnnjttgate.
-() Figur=e 2A provictes the restilts of a Western Blot of homogenates nf I11;K-2()3 cclfs expressing thc (larr2-GPP conjugatc as well as cnclogenntts (3-arrestin2. (3arr2 inclicates endogenous cellular P-arrestin2; Parr2-GI--P
indicates (3arrestin2-GFP conjugate; approximate molecular weights are indicated to tlte right of the gel. Lane I was treated with anti-~arrestin antibody; Lane 2 witli anti-GFP antibody.
Figtire 2R shows the sequestratinn of P2AR in COS cells witli and without overexpressed (3-arrestin2 (left two bars) and Nvith atld withottt overexpressed (larr2-GFP (right two bars). Wild type P-arrestin2 and (lctrr2-enhanced (12l1R sequestration equally well above control levels, produeing a 2.5 and 2.4 fold increase, respectively.
Figttre 3A: Confocal microscopy phntoniicrographs show (3art-2-GrP translocation fi-om cytosol (panel I at left) to membrane (panel 2 at riglit) in HEK-293 cells containing the (32AR, due to the addition of the (iAR2 agonist isoproterenol. Bar = 10 microns.
Figure 313: Confocal microscopy photomicrographs sliow (iarr2-GrP tt-anslocation from cytosol (panel I at left) to niemhrane (panel 2 at right) in COS ceils containing the (i2nR, and due to additioti of the (iAR2 agonist isoproterenol. Bar = 10 micrrnis.
Figure 4 depicts a IIGK-293 cell containing 12CA5(11/\) tagged (32AR (confocal microscopic photographs). Row A sliows a cell ifter reorganization of (QAR into plasnia tnembrane clusters. Row B provicles three pictures of tlle sanie cell at 0, 3, and 10 minutes (left to rigllt) after tiie adclition of agonist. Redistribution of ~art2-GFP to the cell membrane is sliown hti' the enliancement of membrane fluorescence wltll a conconiita'nt loss of cvtosolic (luorescence. Arrows indicate areas of eo-localization; har= 10 niicrons.

Fihure 5 shows the influence of overexpressed GRK on the t-edistribution of (3ar--2-GFP in I-1CK-293 cells expressing the Y326/1 phosplhnrvl,,tition-impaircd P2/1R. Cefls without (Row A) and with (Rrnv B) overexpressed GRKs wer-e exposed to agonist, and the real-tinie redistrihuticn of Par,-2-GFP xvas observed. (iarr2-GFP translocation in cells containing overexpressed GRK (Row B) was niore robust, indicating an increased affinity of ~art-2-GFP for receptor. i3ar = 10 niicrons.
Figui-c 6A depicts the agonist-inciuced titne clependent translocation of (lart-2-GFP to heta2 adrenergic receptors in a representational T-1T;K-293 cell.
Figure 6i3 graplis the titne course of agonist-induced translocation of ~arr2-GFP to beta2 adrenergic receptors in HEK-293 cells; this graph is quantitative anci is haseci on the responses of a plurality of cells.

Figure 6C is depicts the agonist-induced translocation of Parr2-GFP to beta2 adrenergic receptors in representational i-lEK-293 cells, at varying doses of agonist.
Figure 6I) graphs the dose clependent agonist-induced translocation of (iarr2-GFP to beta2 adrenergic receptors in ITL;K-293 cells; this graph is quantitative and is haseci on the responses of a plurality of cells.

Figure 6E evaluates the translocation of (iarr2-GFP fi-om the cell cytosol to the cell memhrane, in response to exposure to receptor agonist (middle panel) and suhsequent exposure to receptor antagonist (right panel).

DETAILED DESCRIPTION OF TIii; INVENTION
The present inventors have determined that (3-arrestin redistrihution fi-om the cytosol to the plasma menzhrane occurs in response to aeonist activation of GPCRs. Tile present invcntors demonstrated a conimon role for (i-arrestin in agonist-niediated signal transduction termination following agonist activation of receptors. The present inventors have devised convenient niethods of assaying agonist stimulation of GPCRs in vivo and in vitro in real time. Although the pharmacology of members of the GPCR superfamily differs, the methods of the present invention utilize (3-arrestin translocation to provide a single-step, real-time assessment of GPCR function for multiple, distinct members of the GPCR superfamily. The present methods may additionally be utilized in studying and understanding the mechanisms of actions of various therapeutic agents. The present inventors have determined that a protein conjugate or chimera comprising an arrestin molecule and a detectable molecule (such as Green Fluorescent Protein) is useful in such methods of assaying in vivo GPCR
activity.

Due to the therapeutic importance of GPCRs, methods for the rapid screening of compounds for GPCR ligand activity are desirable. Additionally, methods of screening orphan GPCRs for interactions with known and putative GPCR ligands assist in characterizing such receptors. Optical methods are available for studying labelled protein dynamics in intact cells, including video microscopy, fluorescence recovery after photobleaching, and resonance energy transfer. However, such methods are of limited usefulness in labeling GPCRs for study, due to the relatively low level of GPCR expression and the alterations in receptor function that can occur after tagging or labeling of the receptor protein. Radiolabeling or fluorescent labeling of test ligands has also been utilized in screening for GPCR ligands. See, e.g. Atlas et al., Proc. Natl. Acad. Sci.
USA 74:5490 (1977); U.S. Pat. No. 5,576,436 to McCabe et al. The introduction of foreign epitopes into receptor cDNA to produce hybrid GPCRs is now a standard technique, and enhances detection of GPCRs by monoclonal antibody technology. However, such techniques are limited in their applicability to living cells. U.S. Pat. No. 5,284,746 to Sledziewski describes yeast-mammalian hybrid GPCRs and methods of screening for GPCR
ligands using such hybrid receptors. U.S. Patent No. 5,482,835 to King et al.
describes methods of testing in yeast cells for ligands of mammalian GPCRs. However, application of these techniques to the study or identification of orphan GPCRs requires prior knowledge of ligands or signal transduction events and are therefor not generally applicable or universal.

Phosphorylation Of GPCRs is a meclianisni leading to desensitization of (lie receptors; receptors that have been contintrotrsly or repeatedly stimttlated lose responsiveness, wliereas the ' responses of other receptors remain intact. See 1Iarden, I'hrn=nincol. Rev. 35:5 (1993); f3enovic et al., Amn. Rer. Cell. 13io1. 4:405(1988). In a variety of cells, specific kinascs liave evolved for specific GPCRs. Desensitization occurs via the following pathway: agonist occupancy of lhe receptor transforms the receptor in(o an appropriate suhstt-ate for an associated kinase; (i-arrestin binds to the kinase phosphoryla(ed receptor ancl pt-events st-bsequent interaction with tlle appropriate G-protein, as well as initiating hotli internalization and resensitizatirnl processes.
Ferguson et af, Science, 271:363 (1996); Lohse et al., Science 248:1547 (1990).
~-arrestin dependent ciesensitization is induced only when the GPCR is activated hy ligand binding, and is an example of homologous desensitization (i.e., the f igand (iesensitizes only its target receptors). T.,ohse et al. (1990) ancl nttramadal et al., .l. 13io1. (.'hem. 267:17982 (1992) provide cDNA and amino acid seduences of P-arrestin. Various isofornis of (3-arrestin are known; as used herein, (i-arrestin refers to all such isofornis of P-arrestin, proteitis having suhstantial sequence similarity thereto wlllch are ftrnctional (t-arrestins, and functional fragments thereof. Tunctional fragments of p-arrestin, its isofornis and analogs, may Eie deterniined using techniqties as known in the art.

N/lolecules detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical and optical means are known. Optically cietectahle molecules include fluorescent labels, sticli as comniercially available flttorescein ancl Texas Rect. Detectable molecttles ttseft-l in the present invention include any biologically compatible molecule which may he conjugatecl to a(3-arrestin protein Nvitliout cnmpr-omising the ability of P-arrestin to interact with the (;PCR
system, and without compromising tlie ability of the detectable niolecule to he cletected.
Conjugatecl molecules (or conjugates) of 0-arrestin and cletectalzle molecules (which also may be tet-med 'detectably labelled (3-arrestins') are tlius useful in the present invention. Preferred are detectable molecules capable of being synthesized in the cell to he studied (e.g., wliere the cell can be transformed witli heterologous DNA so that the (3arrestin-detectable molecule chimera is produced -il-within tlie cell). Particularly preferred are tiiose detectable molecules which are inherently (luorescent in Wi~o. Suitahle detectable molecules niust he able to be cletecteci with suf[icicnt resolution within a cell that translocation of P-arrestin fi-om the cytosol to the cell tnemhrane in response to agonist binding to GPCR
can be qualitatively or quantitatively assessecl. Molecules detectable by optical means are presently preferred.

Fusion proteins with coding sequences for heta-galactosiclase, fire(ly luciferase, anci bacterial luciferase liave been used in methnds of detecting gene expression and protein intet=actions in cells. Ilowever, these niethocis require exogenoEtsly-added substrates oi- cofactors. ln the methods of the present ]nvention, an inlierentiy fluorescent marker niolecule is preferred, sucli as GPp, since detection of such a marker ititracellularly requires only tiie radiation by the appropriate wavelengtii of light and is not suhstrate limitecl.

Gt-een riuorescent P--otein (GFP) was first isolated from the jelly fisli Aeqrioren vicloricr, and lias an inilerent green hioluminescence that can be excited optically by blue light or nonradiative enet-gy transfer. Sequences of GFP-encoding cDNA and GFP proteins are known; see, e.g., Praslier et al., Gene, 1 l 1:229 (1992). Tlie crystalline strticture of GFP is descrihed in Ormo et al., S'cience 273:1392 (1996). Purifiecl native Gri' absorbs blue light (maximally at 395 nm with a niinot- peak at 470 m) and emits green light (peak emission at nni) (Morise et al, 13iochemi,clrY, 13:2656 (1974); Ward et al., Pholnchem.
1'hnlohivl., 3 1 :61 1(19RO)). 1t lias heen shown that GFi' expressed in prokaryotic and eukaryotic cells produces a strong green fluorescence when excited by near (JV or blue liclit (see US Patent No. 5,491,084 to Chalfie and 1'raslter); as this fluorescence requires no additional gene products froni A. ilicforia, chrotnophore formation is not species specific and occurs eitl-ier tlirougli the uses of uhiquitous cellular coniponents or by autocatalysis. Expression of GPP in I;,c=cheric'I1i(1 co/i results in an easily detected green fluorescence that is not seen in crnitrol bacteria.
See Chaifie et al., .Science 263:$02 (1994); US Patent No. 5,491,084. Cells expressing the green-fluorescent proteins may he conve.niently separated from those whicil do not express tiie protein by a fluorescence-activateci cell sorter.

As used herein, Green Fiuorescent Protein refers to the various naturally occut-ring forms of GFP which can be isolated from nattn-al sottrces, as well as artificialfy mocii(ied GFPs which retain the iiuorescent ahilities of nalive GFP. As discussed in Ornio et at., Science 273:1392 (1996), various mutants of GFI' have hecn created with altered excitation and emission maxima. Two cliaracteristics of wild-type GFP which affect its useftiiness in mammalian cell lines are the neecl to excite it at UV waveiengths to obtain a maximal fluorescent signal, anci ciecreased (luorescence at teniperatures over 23 C. Iloweve.r, the S65T/GFP mutant overcotnes these liniitations. Heim et al., 1'roc. Nnll.
Acncl.

Sci. USA91:12501 (1994). Aclditional alterations in the GFP protein sequence which provide inherently fluorescent, biologically compatible molecules will be apparent to those in the art; sequence alterations may be made to alter the solubility characteristics of the protein, its excitation wavelengtli, or other charactet-istics, while retaining useful fluorescent properties. See, e.g. US
I'atent 5,625,048 to 'fsien and I-leini; WO 9711091 (Rjorn, Poulsen, Thastrup and Tullin); WO 9627675 (Haseloff, I-lodge, Prasher an(I Siemering); WO 9627027 (Ward); WO 9623898 (13jorn et al.); WO 9623810 (1-Ieim and Tsieti); WO
9521 191 (Clial fie an(i Ward).
Cells useful in the methods of the present invention inciude eukaryotic and p--okat-yotic cells, includitig hut not fimitecl to bacterial cells, yeast cells, fungal cells, insect cells, nematode cells, plant or animal cells.
Suitahle animal cells include, but are not limited to HEK cells. i1eLa cells, COS
cells, anci various primary maminallan cells. Cells contained in intact aninials, incluciing httt not limiteci to neniatodes, zebrafish (and other transparent or semi-transparent animals) and fi-uitflies, may also be ttsed in the methods of the present invention.
An animal mcxle.l expressing a(iarrestin-dctectahlc molecule fusion protein iht-cntghottt its tissues, or within a particttlar organ or tissue type, xvill be useful in studying cellular targets of known or unlcncnvn GPCR iigands.
Cells useful in the present methods include those which express a known GPCR or a variety of known GPCRs, or wliich express an unknown GPCR or a variety of unknowti GPC.Rs. As used lierein, a cell which expresses a GPCR is otie which contains that GPCR as a functional receptor in its memhrane; the cells may naturally express the GPCR(s) of intei-est, or mav he genetically cngineered to express the GPCR(s) of interest. As used herein, an 'unknown' or 'orphan' I-eceptor is one whose function is tmknown, and/nr whose ligands are unknown.

The present experiments Green fluorescent protein (GFP) has been used to sttidy pi-otein-protein interactions in living cells. See Kaether & Gerdes, rr;BS
Le1l.
369:267 (1995); Olsrni et al., J. Cell. Biol. 130:639 (1995). Green fluorescent protein (GFP) is useful as a reporter molecuie for ftlsion proteins due to its inllerent fluorescence and its folding, which apparently isolates it from its conjugated partner. Prasher et al., Gerre 1 1 1:229 (1992); Ormo et al., Screl7ce 273:1392 (1996). Tol- example, a seven transmembrane protein as complex as tiie (j2AlZ, w111C11 is three times larger than GFP, exhibits normal hiochemistry after GFP conjugation to its C-terminus. Barak et al., UnI. 1'hrnMtacr~l. 5 1:177 (1997).

The present inventors established that a fusion protein c(lnsist3ng of a(1-arrestin molecule (P-arrestin2) conjugated to a GFP at its C-termintls ((3arr2-G17P, Fil;ui-e 1) is expressed in cells and is biologically active.
The (iarr2-GFP ftlsion protein is approximately 50% larger tlian P-arrestin2, and this size increase is reflected by its slower migration on SDS-Page (Figure 2A).
The left lane of Fit;tn=e 2A, exposed to an antibocly against (j-arrestin, shows that (3arr2-GFP rllnS more slowly than endogenous (3-arrestin2 (highiighted middle band). The right lane of Figure 2A, treated witli a monoclonal anti-GFP
antibody, clemonstrates that the slower band does indeed contain GFP.

(i2AR normally seqtieSters poorly in COS cells, and this has becn cori-elated to the relatively poor expression of endogenous (i-arrestins in COS
cells. Menird et al., Ho1. 1'harmacol. 51:800 (1997); Zllang et al., .1. Rio1.
C..'hem.
271:18302 (1996). Ovei-expression of exogenous (3-arrestin enhances ~211R
sequesti-ation in these cells; similarly, as shown herein, Parr2-GFP
overexpression in COS cells augmented (i2AR internalization (Figure 213), demonstrating that Pa1-r2-GFr is biologically active and equivalent to native (1-arrestin.

Biochemical eviclence indicates that P-ai-restins are pi-edominantly cytosolic proteins. Ferguson et al., Can. J. PlY.ciol. 1'harmac.ol. 74:1095 (1996).
The present inventors, using confocal microscopy of Parr2-CTFT' in 11EK-293 cells (rigtii-e 3A, left panel), confirmecl that Parr2-GFP is distributeci tlii-otigliout the cytosol and exclucleci froni the nucleus. Tlie present data also establisli for the fii-st time that (3-arrestin is tiot predoittinantly compartmentalized at the plasma membrane in the absence of agonist but that, tipon addition of saturating concentrations of an agonist to the cell medium, (1-arrestin is translocaled from cell cytosol to cell membrane. Wltere P-arrestin is conjufiated to an optically cletectabie molectile such as GFP, as shown lierein, a rapid and readily observable optical eniiancement of the membrane and a concomitant loss of cytosolic optical signais occurs (see Figtires 3A and 3i3, where membrane fluoi-escence is enhanced and cytosol (luorescence is decrease(i dtie tn translocation of the (iarrestin-GI'P chimera).

To investigate xvhetlier the intraceiluiar translocation of (i-arrestin targeted hinding sites in the plasma menibrane otlier than tiie (i2AR, the hresent inventors (irst crosslinked the receptors ttsing monocional antibodies. As reported herein anci shmvn in rigure 4, the geometrv of the agonist-induceci time dependent translocation of (3-arrestin to the plasma membrane mimicked tiie distribution of rre-aggregated (t2ARs, indicating that the targetect sitc of ~-arrestin is incleed P2AR or an associated component.

It has Eieen postulated that phosphorylation ofGPCRs by GIZKs facilitates ciesensitization by increasing their affinity for (i-arrestins. Gurevich et al, .I. I3iol.
C77em. 268:16879 ( l 993); Gurevicil et al., .1. 13io1. Chem. 268:1 1628 (1993).

Wlien e.xpressed in i-IEK-293 cells and exposed to agonist, mutant Y326A-P2ARs ai-e not signi(icantiy pliosphorylateci by enclogenous GRKs (Ferguson et al., .I.
I3rol. C'hem., 270:24782 (1995). Tlierefore, the present inventors utilized tizis mutant receptor to investigate the above qttestion of P-arrestin aflinity in vivo.
Y326A-(32AR was cotransfected with Parr2-CiFP into F-1p;K cells in tlle absence and presence of co-transfected GRK. If the above hypothesis were true, reversal of pliospliorylation impairment by overexpressed GRKs woulcl result in a noticeable diffei-ence in (3arr2-GFP translocation. As reported herein, without added GRK, Parr2-GPP translocation in response to agnnist rroceeded poorly;
with the addition of GRK, Nrr2-GFP translocation to the plasma membrane was n1nCl7 more rohtist (rigurc 5), indicating the importance of phosphorylation to P-arreStln activity.

Tlie present inventors determined that translocation of (i-arrestin from tltie cell cytosol to the cell membrane is an indicator of agonist stimulation of GI'CR activity, and that a cliimerie protein coniprising P-arrestin and the detectable molecule GFP was capable of detectably displaying the real-time translocation of (i-arrestin in response to agonist activation of GPCRs.

lt) Thc results presented lierein establish that P-arrestin targets GPCRs or an associated molectile following agonist binding and receptor phosphorylation. These data clemonstrate a biological behavior for ~-arrestin that has only been postulated froni biochemical studies, and characterize for the first time how (1-arrestin compartmentalization changes after initiation of receptor signal transduction. Agonist activation of a GPCR ultimateiy culminates in the association of P-ari-estins witlt GPCRs, thc3s the visualiz,ation of the agonist mediated P-ai-restin translocatinn process provides a tiniversal indicator of GPCR
activation.
The pi-esent invetitors have denionstrated that GPCR signal transduction induces a rapicl, subslantial increase in the relative and ahsolute amount of plasma menibrane botuid (i-arrestin. The agonist-mediated i-edistribtition of (3-arrestin coupled to a detectable molecule provides an optical amplification of tlze extracellular signals transduced by GPCRs, and this occurs simultaneous witli, or within the same time frame as, the chemic.al aniplification normally provided by second messenger cascades. Chimeras of (3-arrestin and a detectable molectile are tiseful for the study of P-arrestin kinetics and GPCR
related behavior such as endocytosis. /ldditionally, such chimeras are usefttl as biosensrn-s for signaling when GPCRs become activated, and provide methods of screening compounds for GPCR activity, and screening orplian GPCRs for ligand responsiveness. In addition, the ability of co-transfected GRKs to enliance botli the i-ate and extent of (j-arrestin translocation indicate that the present metltiods and constrticts can also be tised to tnonitor GRK activity, as well as monitor druOs, proteins and compounds for activation or inhibition of the GRK/(~-arrestin pr-ocess.

The present invention provides a metiiod for sCreening comPounds for GPCR agonist activity, comprising: a) proviciing a cell expressing a known or unknown GPCR and containing a chimeric protein comprising a(3-arrestiil protein and a visually detectable protein; b) exposing the cell to a test compound;
and c) detecting translocation of the detectable molecule from the cytosol of the cell to the membrane edge of the cell; wliere translocation of the cletectahle rnoiecule from the cvtosol to the membrane edge of the cell indicates activation of the GPCR and, accordingly, the GPCR activating effect of the test compouncl.
Translocation of the cillmertc proteln is evidenced by an lncrelse in the intensity of detectable signal located at the membrane edge (and/or a (lecrease in the cytosol), vvhere the cliange occurs after exposure to the test compouncl.
Translocation may thus he detected by eomparing changes in the detectable signal in the same cell over time (i.e., pre an(i post test compound exposure).
nlternatively, a test cell may be compared to a crnitrol cell (no expostire to test compound), or a test cell may be compared to a pre-established standard. 1 f a known agnnist is available the present methods can he nsed to screen for and stucly GPCR antagonists. Additionally, the memhrane association of (i-arrestin shoulci he increased by expression of an excess of receptor or by a constittitively active GPCR that undergoes phosphorylation by GRKs even in the absence of agonist. Therefore, the present nletliods can be tised to nionitor for inverse agonists oF GPCRs.

Methods of detecting the intracellular translocation of the cliimeric protein will depend on the particular detectable protein utiiized; one skilled in the art will be able to readily devise detection metliods suitahle for particular detectable molecules, given the teachings of the present specification ancl knowleclge in the art. In a prefei-red embodirnent, the visually detectable protein is a green-fluoresc.ent protein (GFP) as discussed below.
The niethocis of the present invention provide easily detectable results. 'fhe translocation of (3-an-estin coupied to a detectable molectile such as GFP, in t'esponse to GPCR activation, restilts in a relative enliancement of the detectable sigrial at the cell edge (i.e., at the cell membrane). ln addition, the concomitant clecrease in detectable signal from the cell cytosol means that 'background noise' (detectable signals whicii do not change in response to GPCR
activa(ion) is minimizecl. In certain cells, activatioti of GPCRs will result in essential clearing of cietectable signal from the cytosol, and a 100-folci increase (or more) in the detectable signal at the cell membrane. Tn the present nietliods, it is preferred that the detectable signal at the membrane edge increase, after GPCR activation, at least two-fold, more preferably at least 3-fold, and more pr-eferably at least 5-fold or at least ten-fold.

As trsed herein, the introduction of a chimer-ic protein into a cell may be acconlhlished by introducing into the cell (or the cell's ancestor) a nucleic acid (e.g., DNA or RNA) seqtrence or constrtrct encoding tile cliinieric protein, and culturing the cell in an environment wliich allows expression of the chirneric protein. lntroduction of nticleic acids encociing the chimeric protein, or introduction of the protein itself, into a cell may be car-ried out by any of the rnany Stritable methods vvhich are known in the art, incluciing transfection, electroporation, rnicroinjection, anci liposome delivery.

Tlle present invention rrovides a DNA constT-uct compr-iGing a promoter, DNA encoding a(3-arrestin protein operatively associated therewith, ancl DNA encoding a visually detectable niarker protein operatively associated therewitli. The promoter is operativeiy associated witli the encoding DNn; DNA
encoding P-arrestin may be 5' from DNA encoding the visually detectable marker, or vice versa. In a pr-e.fer--ed enibodiment, the DNA encociing a vistrally detectable marker encodes a green-fltroreseent protein (GFP). Vectors comprising such DNA constructs are a furtlier aspect of the present invention.

The present invention furtlier provides conjugates (such as chimeric pr-oteins or fusion proteins) wliicli comprise a(l-arrestin protein and a visually detectable protein. In a prefet-red embodiment, the visually detectable protein is a green-fltrorescent pr-otein (GFP).

The present invention furtlier provides a cell cC}nlpt'Ising a DNA
molectrle, which DNA molecule comprises, in the 5' to 3' direction, a pr-omoter, DNA encoding a(3-arrestin protein operatively associated therewitli, and DNA

-1g-encociing a visually detectable marker protein operatively associated therewith.
In a preferrec] emhodinient, the DNA encoding a visually detectable market-encodes a green-fluorescent protein (GFP).
The cells of the present invention nlay be used to cletect the presence of speci(ic molecttles in various kinds of samples such as, e.g., aquecnis samples, biological samples (for example blood, urine or saliva), environmental samples, C1r 1ndlEstrlal samples. In such ttses. the cells contain a GPCR
whose agonists are known. Activation of the GPCR and the coneoniitant translocation of the detectable signal froin the cytosol to the niembrane ecige indicates the presence of the agonist for the GPCR. A cell used in such a tnetliod may contain only a single type of known GPCR, or a variety of known GPCRs. Such cletection will be useful for niedical and veterinary diagnostic purposes;
industrial purposes; and screening foi- drugs or chemicals of ahuse or biological toxins that affect GPCR-mediated signal transdtiction.

Tlte cells of tlie present invention mav be clepositecl on, affixed to, supported by, or immobilized on a sttbstrate. The substrate may he of any suitable material wliicli is not harmful or detrimentat to the living cells deposited thereon, i.e., which is bio-cotnpatible witli the living material deposited tiiereon.
The suhsti-ate may he rigid, seini-rigid or flexible; and may be opaque, transnarent, or semi-transparent. The size, geometry and otlter physical characteristics of the substrate will be dictated by the intended use, as will be apparent to one skilled in tiie art. Suitable substrates incltide, but are not limiteci to, plastics, glass, ceramics, silica, hiocompatible monomer and polymer compositions, semiconductor materials, fiber optic materials, polystyrene, membranes. sephadex, and hio-organic materiaEs. Examples of bioconipatible materials are provide(] in US Patent Nos. 5,578,079; 5,575,997 and 5,592,834 to i.,eung and Clark; and 5,522,896 to Prescott.

The present invention furtlier provides methods for screening for the pi-esence of a GPCR agonist in a soltition which comprises: a) providing a cell expressing a knowrt or ttnknown GPCR and containing a chimeric protein comprising a(~-arrestin protein and a visually detectable protein; b) exposing the cell to a test solution; an(i c) detecting translocation of the detectahle molecttle fron1 tlte cytosol of'the ccll to (lic membrane edge of the cell; where translocation of the detectable molectrle from the cytosol to the membrane edge of the cell indicates activation of the GPCR and, accordingly, the GPCR agonist effect of (lie test solution. Translocation of the ciiimeric protein is evidenced as discussed ahove.

The present invention ftn-ther provides metllods for screening for the presence of a GPCR antagonist in a solution whicll comprises: a) provi(iing a cell expressing a GPCR and containing a chinieric protein comprising aP-arrestin protein and n visuallv (letectahle protein; h) exposing the ccll to a test compound; then c) exposing the cell to a known agonist to the GI'CR expresse(i in the cell; and d) cletecting translocation of the detectable molecule from the cytosol of the cell to the membrane edge of the cell. lf the test cornpound contains an 'mntagonist, translocatloil of the detectahle molecule will be delaved for a period of time corresponding to dtrration of antagonist action on tlie receptor (wliicli time perioci will vary depending on the antagonist and/or (lie receptor).
Tr-anslocation of the detectable molecule frorn the cytosol to the membrane ecige of the cell inclicates activation of the GPCR hv the agonist. Accorciingly, wllen translocation does not occur or is delayed (compared to that which wotrl(l occur in the absence of test compotrnd), the test compound contains an antagonist to tiie GPCR. Ahsence or delay of translocation mav he assessed hv comparison to a contr-ol cell (not exposed to test cornpound) or to a predetermined standard.
Translocation of the chimeric protein is evidenced as discusseci above. Fxposw-e to the test compound and the known agnnist may occtrr at essentially the same time, or exposure to the agonist may occur strhsequent to exposure to the test compound. As used herein, subsequent exposure refers to exposure within the time period during which a potential antagonist woulcl be expecte(I to be interacting witli the GPCR (i.e., binding to or hound to the GPCR).

Tlte present invention ftrrther provides metliods for screening a cell for the presence of a GPCR, comprising: a) providing a test cell; b) introducing into the test cell a ehinier-ic protein coniprising aP-arrestin protein an(i a visually (ietectahle protein; and tlien c) exposing tile cell to a test solution containing a known agonist to a GPCR; and d) detecting translocation of the detectable molecule from the cytosol of the cell to the membrane edge of the cell;
~,vhere translocation of the detectable molecule from the cytosol to tlle tnemhrane edge of tiie cell indicates activation of a GPCR and, accordingly, (liat the test cell contains such a GPCR. Translocation of the chimeric pt-otein is evidence(i as discussed above.

The rresent invention further provides metliocis for screening a cell population for the presence of cells containing GPCRs, compt=ising: a) providing a population of test cells, said test cells containing cliimet-ic proteins comprising aP-arrestin protein and a visually detectable protein; and then b) exposing the cell population to a test solution containing an agonist to a GPCR; and (1) cletecting those cells in whicli translocation of the detectable niolecule frcrin tlie cytosol of the cell to the membrane edge of tiie cell occurs; where translocation of the detectable molecule fi-om the cytosol to the memhrane edge of a cell indicates activation of a GPCR and, accordingly, that the cell in question contains a GPCR. Translocalion of the ciiimeric protein is evidenced as discussed above.
Populations of cells to be screened inciude a collection of InCliv3citial cells, a tissue comprising a plurality of similar cells, an organ comprising a plurality of t-elatecl cells, or an org8ntsin eoniprising a plurality of tissues anci ot=gans.

As usecl lierein, 'exposing' a cell to a test compound or soltition means bringing tihe cell extet-ior in contact with the test compound or solution.
Wliere tlle test conipound or soltttion is being screened fot- GPCR ligand activity, expostn-e is carried out tuider conditions that would pertnit binding of a GCPR
ltgaild to a receptor expressed in that cell. As used herein, 'translocation' of P-arrestin refers to movement of tlie 0-arrestin molecule from one area of the cell to another.

The present metliods niay further be used to assess or stucly lhe effects of any molecule in the GPCR pathway Nvllicli exerts its effect upstream of (3-arrestin binciing (i.e., prior to P-arrestin binding to the phosphorylated GPCR). Thus the present invention provides methods for assessing GPCR
pathway ftinctions in general. As ttsecl liet-ein, the GPCR pathway refers to the series of events which starts witli agonist acttvation of a GPCR followed by uesensitization of tlle receptor via G protein-coupled receptor kinase (C'TRk) pliospllorylation ancl (i-arrestin binding.

In a hr-oad sense the present invention tllus provides a method of screening test compounds and test conditions for the ability to affect (activate or inhibit, enhance or depress) a GPCR patllway, and provides niethods of assessing GPCR patllway function in a cell in general. ln the present metllods, the extent of translocation of P-arrestin is indicated by the degree of detectable changes in the cell; the extent of (3-arrestin translocation is an indicator of the extent of GPCR pathway conipletion. The relative extent of translocation under varied test conditions nlay be compared, or a test condition may be compared to a control condition or to a predetermined standard.

For exampie, the specificity and effects of various kinases (including lllose known to interact with GPCR pathways and those not previously known to interact witll GPCRs) for a specific GPCR or a group of GI'CRs may be assessed by pi-oviding a test kinase to a test c.ell expressing a GPCR and containing a detectable P-arrestin molecule, exposing the cell to a GPCR
agonist, and assessing the tt'anslocatlon of detectable (3-arrestin from the cell cvtosol to the cell membrane (see Example 7 herein). Translocation of the P-arrestin to the cell nlenlbr-arie indicates that the test kinase, in t-esponse to agonist occupancy of the receptor, is able to bind to and pliosphorylate the receptor, so that P-arrestin will then bincl to the kinase pllosphorylated receptor and prevent subsequent interaction with the appropriate G-protein. In similar ways, the functl(ln of altered, reconlbinant or mutant kinases may he assessed; compotmds may be screened for tlle ability to activate or inllibit the GPCR pathway, G prcltein-coupled receptor kinases, or (3-arrestin binding; and the funetion of G-proteins nlay he assessed. For example, the following test conditions may be assessecl tising nletllocls as described herein: the effects of G-proteins (including natural, lleterologous, or artificially altered G-proteins) within the test cell;
exposure of tlle test cell to known or putative GPCR ligands; and co-expression of a second receptor in the test cell expr-essing a GPCR.

Still further, the present methods allow the screening of (3-arr-estins (naturallv occurring, artifically introduced, or altered, mutant or recombinant) for the ability to bind to a phosphorylated GPCR. In such nietliocls, the test (~-arrestin is conjLrgated to a detectable niolecule such as GPP, and is placed within a cell containing a GPCR. The cell is exposed to a known agonist of flie GPCR, and translocation of lhe detectahle inolecule frrnii the cvtosol of the cell to the membrane edge of the cell is detected. The translocation of the detec.tahle molecule indicates that the test (3-arrestin protein is able to hind to the hhosphorylated GPCR. As in other methods of the present invention, the translocation may be compared to a control cell contalning a known P-arrestin, or to a predetermined standard.

G Protein Coupled Receptors GPCRs sLiitable for use in the present metliuds are those in Nvhich agrnlist hinding induces Ci protein-coupled receptor kinase (GRK) phosphorylation; translocation of arrestin frrnii t1ie cytosol of tlie cell to the cell membrane suhsequently occurs. As it is believed that virtually all memhe.rs of the GPCR. superfamily clesensitize via this coininon mechanism, examples of suitable types of GPCRs inciucle bnt are not limited to beta and alpha adrenergic receptors; GPCRs hinding neurotransmitters (such as dopamine); GPCRs binding liormones; the class of odorant receptors (taste, sniell and cheniotactic i-eceptors as found in nasal mucosa and the tongue, and on sperm, egg, immune system cells an(i blood cells); the class of type 11 GPCRs including secr-etin, glucagon, and other- cligestive tract receptors; light-activated GPCRs (such as rhoclopsin);
and members of tlle type Ill family of GPCRs which include hut are iiot limited to metahotopic glutamate receptors and GnBA,, receptors. In addition to naturally OCCLIrrIng CiPCRS, C;PCRs rnay he specifically engineered or created hy random mutagenesis. Such non-naturally occurring GPCRs may also he utilized in and screenecl by the pi-esent methods. The present nietliods may he utilized with any membrane receptor protein in which agonist hincling results in the transiocation of (i-arrestin. Such receptors include grcnvtii factors that signal thrrugh G preteins.

Atrtomatecl Screening Nletilods The nietliocls of the present invention may he automated to provide convenienl, rcal tinic, liigli vcilume niethocis of scrcenirng compouncls 1'6r Gl'CR
ligand activity, or- screening for the presence of a GPCR ligancl in a test sample.
Autoniatecl rnethods are designed to detect the change in concentration of labelled (3-arrestin at tile cell membrane and/or in the cytosol after expostrre to C'rl'CR
agonist. The alteration of {3-arrestin distribution can be detecteci over time (i.e., compar-ing the same cell bef'ore and after expostrre to a test sample), or by comparison to a cnnt--ol cell which is not exposed to the test sample, or by comparison to pr=e-estahlished indicia. Botlz qualitative assessments (positive/negative) ancl qtrantitative assessments (comparative degree of translocation) may be provided by the present automated methods, as Nvill he apparent to those skilleci in the art.

It is tlitrs a further object of the present invention to provide methocls and apparatus for automated screening of GPCR activity, by cletecting tlie translocation of delectahly labeled j3-arrestin from cell cytosol to cell membrane in response to agonist activation of GPCRs. The translocation may be inclicatecl by an alteration in the distrihution of a detectahle signal within a celi over tiine, hc:tween a test cell and a control cell, or hv comparison to previously estahfisheci parameters. ln particular, according to one embodiment of the present invention, a plurality of cells expressing GPCRs and containing ciiimeric proteins compr-ising a detectable molecule and a P-arrestin molecule are provided.
Indicia of the distr-ihution of the detectable nlolecules are then measured using conventional teciiniques. In various embociiments, (a) measurement of optical inclicia occurs befor-e and after the addition of a test sample to a cell, and the tinie point measurenients are compared; (b) optical indicia are measurecl in a test cell exposed to a test sample and in a non-exposed control cell, anci tizese measurements are compared; and (c) rneastrrement of a test cell after .acldition of a test sample is compared to pr-eestablished parameters. The optical indicia being measured may be lluorescence signals (e.g., (luorescence ititensities) if the detectable molecule of the chimeric P-arrestin protein is a fltrorescent inclicator WO 98/55635 PCTIllS98/11628 such as GfP. Other optical indicia that at-e suitahie for real-time nieasurement may also be used, as will be apparent to those skillecl in the art.

nn embodiment of the present invention includes an apparatus for determining GPCR response to a test sample. This apparatus comprises means, such as a fluorescence measurement tool, for meastiring inclicia of the intracellular distribution of detectable (i-arrestin proteins in at least one test cell, ancl optionally also in a conti-ol or calibration cell. Measurement points may be over time, orn- among test and control cells. A coinputer pr-ogrim prodtrct controls operation of tiic measuring nteans and perfornis numeric.al operations rclating lo tlie ahove-ciescrihed steps. The preferred computer program prodtict comprises a computer readable storage illedit7i11 having coniputer-readahle hrogrstm cocle means eriil,iodiecl in the medium. FIaT-clware suitahle for t-se in such autoniateci apparatus Nvill he apparent to those of skill in flie art, and may include cornptiter controllers, automated sample handlers, (ltroresence rneasurement tools, printers and optical displays. The measurement tool may contnin one or more pImtodetectors for measuring the fluorescence signals from samples where fluorescently detectahle molecules are utilized in the detectahle P-lrrestin const--uct. The measurement tool inay also contain a coniptrter-controlled stepper motor so that each control and/or test sample can be arranged as an array of samples and automatically and repealedly positioned opposite a photodetector dtnring the step of ineasuring fluorescence intensity.

The meastirenient tool is preferably operatively coupled to a general purpose or aprlicatirni specific coniputer controller. The controller preferahly comprises a computer program product for controlling operation of the measurement tool and performing numerical operations relating to tlie above-described steps. I'he conti-oller may accept set-up and other related data via a file, disk input or data bus. A display and printer may also be provided to visuallv display the operations performed by the controller. It will be understooci by those having skill in the art that the functions performed by the contr-oIler may be realized in whole or in part as software modules running on a general purpose comptrter system. illternatively, a dedicated stand-alone system with application -'S-specific intet?rated circuits for perforniing tiie above described functions and operations may he provided.

As provided above, the inclicia of ~-arrestin clislribution may take the foi-m of tluoi-escent signals, altliough those skillect in the ai-t will appreciate that othei- indicia are known and may be used in tite practice of the present invention. such as may he provided by labels that produce signais detectable by flurn-escence, radioactivity, coloriinetry, X-ray diffraction or ahsorption or magnetism. Such labels inciude, for example, fluoropliores, chrotnonhores, radioactive isotopes (e.g., 32P or 1251) and electron-dense reagents.

DeCnitions As used herein, exogenous or heterologous DNA (or RNA) refers to DNA (oi- RNA) wliich has heen introcluced into a cell (or the cell's ancestor) ilirougli the effoi-ts of humans. Such lieterologous DNA may be a copy of a sequence which is naturally fotmd in the cell being transformed, or a sequence which is not naturally founcl in the cell heing transformed, or fragments tliereof.
As used herein, the term 'gene' refers to a DNA sequence that incorporates (1) upsti-eam (5') regulatory signals inciuding a promoter, (2) a coding region specifying the product, protein or RNA of the gene, (3) downstream (3') regions including transci-iption termination anci polyadenylation signals ancl (4) associated sequences required for efficient and specific expression.
Use of the phrase "suhstantial sequence similarity" in the present specification refers to DNA, RNA or amino acid sequences wliich have sliglit and non-consequential sequence variations from a seqtience of interest, and ar-e considei-ed to be equivalent to the sequence of interest. In this regard, "sliglit anci non-consequential sequence variations" mean that "similar" sequences (i.e., sequences that have suhstantial sequence simiiarity) will be functionaliy equivalent. Functionally equivalent sequences will function in suhstantially the same manner to produce stibstantially the same compositions.
As used herein, a "native DNA sequenee" or "natural DNA
sequence" means a DNA sequence whicli caii he isolated from non-transgenic cells or tissue. Native DNA sequences are those which liave not heen artificially altered, suclti as by site-directed mutagenesis. Once native DNA sequences are identified, DNA molecules having native DNA sequences mav be cliemically syntliesized or produced using recombinant DNA procedures'as are known in the art.

As used lierein, "a regulatory element" from a gene is the DNA
seqLtence which is necessary for the expression of the gene, sLicii as a promoter.
In this invention, the term "operatively linked" to -neans that following such a link a regulatory element can direct the expression of a linkeci DNA
seqttence.

The tet=m 'promoter' refers to a region of a DNA sequence that incorporates the necessary signals for the eff icient expt-ession of a coding seqttence. This may include sequences to which an RNA polynierase binds but is iiot limited to sucli sequences and may include regions to which other regulatory proteins hind together xuitli regions involved in the control of protein translation anc] may include cociing seqttences. Suitahle protnoters will he apparent to those skilled in the art, and will vary depending upon the cell in whicli the DNA is to he expressed. A suitable promoter for use in DNA
constt-ucts encoding a(i-arrestin/detectable niolecitle construct may he a pt-omoter naturally found in the cell in which expression is desired; optionally, the pt-omoter of the P-arrestin witliin the cotistruct may be utilized. fioth inducible and crnistitutive promoters are contemplated for use in the present invention.
DNA Constructs DNA constructs, or "expression cassettes," of the present invention inclucle, 5' to 3' in the dit-ection of transcription, a pt-onioter, a DN/1.
sequence operatively associated with the promoter, and, optionally, a termination sequence including stop signal for RNA polymerase and a polyadenylation signai for polyadenylase. All of tiiese r-egulatory regions should he capahle of operating in the cell to be transformed. Suitable termination signals for a given DNA
construct xviil be apparent to those of skill in tiie art.

The term "operatively associated," as used herein, t-efers to DNA
sequences on a singie DNA molecule which are associated so that the function of one is affectcd by the otlier. Thns, a pronioter is opet-atively associated witli a DNA when it is capable of affecting the transcription of that DNA (i.e., the DNA is uncler the transcriptional control of the promoter). The prrnnoter is said to be "upstream" from the DNA, wliich is in turn said to be "downstreani" from the p--omoter.

The expression or transcription cassette may he provided in a DNA
construct which also has at least one replication system. For convenience, it is co-nmon to have a replication system funetional in Lscherichia co,li, such as ColT:1, pSC l O l, pACYC 1 R4, or the like. In this nianner, at each stage after each manipulation, the rest-lting constrttct niay be cloned, sequenced, and the correctness of the manipulation determined. In addition, or in place of the E.
coli replication systeni, a broad host range replication systeni may he emploved, such as tlle replication svstems of the P-1 incompatibility pllsmids, e.g., pRK290.
In addition to the replication system, there will frecluenlly he at least one m1i-ker present, which may be useful in one or more hosts, or ciiffe--ent mat-kers foi-l5 individt-al hosts. That is, one marker may he employed for selection in a prokaryotic host, while another marker may he employed for selecfion in a cukaryotic host. 'I'Itc markers may he protcction against a hi-icide, st-ch as antibiotics, toxins, lieavy metals, or the like; may provide complenientation, by imparting prototrophy to an auxotropliic host; or may p--ovide a visible plietiotype thrcn-gh the production of a novel compound in the plant.
Tlte various fragments comprising the various constrncts, expression cassettes, niarkers, and the like may be introduced consecutively hy restriction enzyme cieavage of an appropriate replication system, and insertion of the particular construct or fi-agment into the available site. After ligation and cloning the DNA constrttct may be isolated for fttrther nianipt-lation. All of these techniques are amply exemplified in the literatt-re as exempliFied by J.
Samhrook et al., Molecular Cloning, A Laboratory Manual (2d Ed. 1989)(Col(I Spring Harbor Laboratory).
The examples which follow are set forth to illustrate the present invention, and are not to be constt-tted as limiting thereof. As usecl herein, (3arr2-GFP =(I-arrestin2 green fluorescent protein; GFP = green iluorescent protein;
GPCR = G protein-coupled receptor; (3ARK = beta adrenergic receptor kinase;

GRK = G prcltein-coupled receptor kinase; (32nR = beta 2 aclrenergic receptor;
i-1EK-293 = human embryonic kidtley cells; DMEM = Dulbecco's modiCied Eagle mediunl; and MEM = Mininial Essential Medium.

Materials and Methods Materials: Isoproterenol was obtaineci fi-oni Sigma RBI. Anti-nlouse antibody was obtained from Signla Chemicals or Molecular Probes.
Mouse monoclonal atltibody against tlle 12CA5 epitope was obtained fronl 13oehringet- Mannfleinl. Cell c.ttlture nieclia was obtained from Mediatech and fetal bovine serunl froni /ltlanta Biologicals. Physiological bnffers were fi-om Gihco-I,ife Tecllnologies itlc. Restriction enzymes were obtained it-clnl Pronlega or New England 13iolabs, T4 ligase was frotll Pronlega, and I-lot Tttb DNA
polymerase from nnlersllam. Commercially available plasmids containing variants of Green Flttorescent Protein were obtained from Clontecil.

Cell C'ultur-e and Transfection: II1;K-293 and ('OS cells werc maintained anci transfected as described by Barak et al., Uol. Phm-177. 51:177 (1997). Cells containing botll beta2 adrenergic receptor and P-arrestin constructs were transfecteci witb between 5-10}tg of receptor cDNA in pcDNA1/i1N/1f' and 0.5-1 g of 1:3arr2-Gr1' cDNA per 100 mnl disll. GRKs were expressed using 5 g of transfected cDNA in pcDNA 1/AMP per dish.

Confocal Microscopy: f1RK-293 cells transfected as described above wet-e plated onto 35 mm dislles containing a centered, 1 cnl well fornled frotll a hole in tlie plastic sealed by a glass coverslip. Primary and secondary antibody labeling of live cells were perfortlleci at 37 C for 30 mintttes in media without serum in a 5% CO2 incubator. Cells were was}leci tbree times between applications. Cells plated as above in MEM ot' DMFLM littffered with 20 n1M
I-iepes wet-e viewed on a Zeiss laser scanning confocal microscope.
Sequestration: Flow cytometry analysis was perfornled using techniques known in the art, as described in Barak et al., .I. Biol. C'l1em.
269:2790 (1994).

Construction of 3-arrestin2-GrP Plasniid (1-arrestin2 c[)NA in the hlasniid pCMV5 wag used as a template.
Oligonucle0tide primers stn-i-ounding a distal Xhol restriction site and the C-terminal stop codon of P-arrestin2 were cised to replace the stop c.odrni with an in frame Baml-TI restriction site by directed nlutagenesis (Valette et al.
Nucleic Acitls Res. 17:723 (1989); Attramadal et al., .1. Biol. Chem. 267:17942 (1992);
Lolise et al., Scieiice 248: l 547 (1990)). The Xhol, BamT 11 segment xvas isolated.
This segment was ligated to the N-terminal portion of (i-arrestin cDNA (ctit from pCMV5 by Sact and Xho[) in the polylinker of a plasmid that had Tieen prev3otisly cligested with SacT and Bam1-1[ and that contained S65"I'-Green Fluorescent Protein distal and in frame to the site of (3-arrestin cDNf1 insertion.
Lollse et al., Science 248: T 547 (1990). The resulting 0-ai-restin-GFP
construct was isolated following insertion and growth in E. coli. Constructs were verified by sequencing.
A linear niodel of the P-arrestin2/S65T-GFP conjugate is prcvided in PiT;ure 1.

Characterization of Qarr2-GFP Expressed bv IIEK-293 Cells llomogenates of IIEK-293 cells transfornied with the plasmid of Gxample 2 wei-e studied using known Western Blot techniques. The restllts showed that ITFK-293 cells expressed botli endogencnis (i-arrestin and the (iarr2-GFP conjugate.
Western Blots of liomogenates of 1-TCK-293 cells transfected witli the plasmici of Example 2 and expressing (3arr2-GFP were performed. /1n equal amount of homogenate material was loaded into eaclh of two lanes (Figure 2A).
Tlie left lane was exposed to anti-~arrestin antibody (Menard et al., Mnl.
1'harnt.
51 :800 (1997)), wliereas the right lane was exposed to a motise monoclonal --ntihody against C~FP. The Parr2-C,FP fusion protein is approximately 50%

larger tllilll JT-arrestin2, and wocild tiius be expected to migrate more slowly than P-arrestin on SDS-Page.

-~0-Txpostre to anti-Parrestin antibody revealecl multiple bars (left lane); exposure to anti-Gf P monoclonal antibody revealed a single bar (right lane). The position of endogenous cellular P-arrestin2 is indicatecl by tfle intermediate bar in the left lane ((iarr2). The heavy hand just below 71,000 on the left lane (Part-2-GFP) is tnirrored by a similar bancl in the right lane.
In contrast, no bancl corresponcling to endogenotas cellular (i-arrestin 2 is observed witli anti-GTP antibody exposure. The treatmetit of the riglit lane with anti-CiFP
antibody demnnstrateci that the slower band labelled by anti-parrestin aniihocly contained GFP.

Biological Activity of 3arrestin-GFP Conju ate P-arrestin activity cati indirectly be assessed by measuring its effect on receptor sequestration (rce Menard et al., R1nl. I'hnrm. 51:800 (1997);
Terguson et al., S'cience 271:363 (1996)). The (i2AR nornially sequesters poorly in COS cells, and this lias heen correlated to the relatively poor expression of endogenous P-arrestins (see Menard et al. Mol. T'harrnincol. 5 t:800 (1997);
Tet-guson et al. Science 271:363 (1996)). nverexpression ofexngencnus P-arrestin enhances (32AR sequestration in these cells. To demonstrate tbat the Parr2-Grt, conjugate is a biologically active P-arrestin, COS cells overexpressing Parr2-('YFP

were examined for augmentation of (32/1R internalization, comparecl to the augmentation of (3AR2 seeti witli the overexpression of (i-arrestin2. Results are sllown in Figure 2B.

Using epitope taggeci ~nR2 receptors, sequestration of PnR2 was strtdied in COS cells overexpressing either (1) exogenous P-arrestin2 or (2) the ~<1rr2-GTP conjugate. Figure 2B sliows the sequestration of P2AR in COS cells witli and witlicntt overexpressed P-arrestin2 (left two b(irs) and with and without overexpressed (iat-t-2-GFP (right two hars). Agonist mediated P211R
sequestration increased from 15 7% to 39 5% in the presence of overexpressed (3-arre.stin2;
overexpression of (3ari-2-CtrP similarly increased agonist mediated ~2AR

sequesti-ation from 25 4% to 58 1%. Wild type (3-arrestin2 anci (iarr2-GrP

enhanced (12nR sequestration equally well above control levels, producing a 2.5 and 2.4 fold increase in ~2nR sequestration, respectively.

'l'lic above resriits indicatcd that the (~arr2-Cilll' conjugatc acts as a biologically active arrestin.

Agoiiist Nlediated Translocation of Qarr2-GrP

Agonist mediated translocation of the Parr2-GFI' chimera from cell cvtosoi to membrane was studied tising HEK-293 and COS cells transfected witli plasmids containing cDNA for the P2AR receptor ancl for the Parr2-GFP
conjugate.
i ll/K-293 and COS cells were transfected with plasmids containing 101ig of cDNA for P2/1R and 0.5-1.0 }ig for (1arr2-CiFP. Cells were assessed using confocal microscopy to detect the inherent intracellular (7uorescence of GFP.
"rransfected I-IEK-293 cells are shown in Figtire 3A, where panel 1 depicts cells prior to the addition of PnR2 agonist, and panel 2 depicts cells following tlie addition of agonist. Transfected COS cells are shown in rigur-e :iR, wliei-e panel I depicts cells just prior to the addition of PAR2 agonist, and hanel 2 depicts ceils ten ininutes after the addition of agonist.

As shown in rigure 3A, Parr2-GFP distrihution in IIEK-239 cells was initially cytosolic (panel 1). No significant nuclear or rnembrane enhancement was apparent. Following the addition of the PAR2 agonist isoproterenol to tiie ceil medium, the real-time agonist-mecliated redistribution of (3arr2-GFP was viewed using confocal niicroscopy. Ten minutes after isoproterenol addition (saturating concentrations), enliancement of membrane (ltrorescence 'was seen with a concomitant loss of cytosolic fluorescence, indicating that the ~arr2-GFP distribution had sliifted to the membrane (panel 2).
These results establish that in I-IEK-293 cells containing the (12AR, Parr2-GFP
expressed by the cell is translocated from cytosol to membrane foilowing the addition of a(i11R2 agonist. Txposure of the test cells to GPCR agonist enhanced membrane bound fiuorescence ten-fold over that seen prior to agonist exposurc.

As shown in T'igure 3R, Parr2-GFP distribution in COS cells was initially cytosolic (panel I). No significant nuclear or niembrane enhancement was apparent. Following the acldition of the PnR2 agonist isoproterenol to tlte cell meditmi, the real-time agonist-mediated redistribution of Parr2-GFP was viewed using confocal nlicroscopy. Ten minutes after isoproterenol addition (saturating concentrations), enliancemetit of inembrane fluorescence was seen witli a concomitant loss of cytosolic fluorescence, indicating that the Parr2-GFP

distribution had shifteci to the niembrane (panel 2). These results establish that in COS cells containing the P2AR, Parr2-GFP expressed by the cell is translocated from cytosol to membrane following the addition of aP/1R2 agonist.

Comparing Figures 3A and 3I3 shows that the (luorescent signal is reduced in COS cells as compared to FiEK cells, reflecting the lower efficiency of sequestr-ation of the P2AR in COS cells. T-Towever, even in COS cells the sbift of (3art2-CTFP in COS cells frotn cytosol to membrane following the addition of ~nR2 agonist is clearly discernible due to tlie fluorescence of the Gr=1l moiety.
The above experiments with COS and 1;-1EK-239 cells were reproduced except that the PAR2 antagonist propranolol was added to the cell mediLnn. Using confocal microscopy to visually track Parr2-GFP in the cell in real time, as above, indicated that no shift in Parr2-GFP from cytosol to membrane occurred in response to aPnR2 antagonist. As shown in rigure 6E, addition of an agonist (micldle panei) resttlted in translocation of Parr2-GFP
from cytosol to membrane; subseqttent additioti of an antagonist (right panel) t-eversed tlie translocation (compare to control, left panel).

Biochemical evidence indicates that (i-arrestins are predominantly cytosolic pi-oteitis. Ferguson et al, Carz. .I. Phvsiol. 1'harmacol. 74:1095 (1996)..
The present resuits confirm that (iart-2-GFP is distril?uted tliroughout the cytosol and excluded from the nucleus. These data also establish that Parr2-GFP is not pt-edominantly compat-tnientalized at the plasnia membrane in the absence of agonist, but that upon exposure to an agonist the cellttlar Parr2-GFP shifts to the membrane. The present results further indicate that the slzift of the Parr2-GFP

WO 98/55635 PCT/US98/1 ] 628 con.jugate in response to tiie addition of a G protein cotipled receptor agonist can be cletectecl optically as an enhancement of membrane fluorescence and/or a concomitant loss of cytosolic fluorescence, and that this response is rapidly observed.

Intraceilular Qarr2-GFP Targets Membrane Receptors Figure 4 shows the time course of ~arr2-GFP redistribution to plasnia membrane 12CA5(IIn) tagged 02AR in HEK-293 cells, as shown by confocal rnicroscopy.

The present exaniple demonstrates that ~2ARs are the target of intracelluiar (3arr2-GrP conjugate proteins. I-IrK-239 cells containing 12Cn5(HA) tagged P2nR receptors were studied. The receptors in the ITEK-293 cells were reoi-ganizeci into plasma membrane clusters (Row A) by crosslinking Nvitli a mnuse monoclonal antibody clir-ectecl against an N-terminal epitope, followed by Texas Red conjugated goat anti-nlouse antibody. In Figure 4, the tlii-ee panels of Row A show the same iTrK-293 cell with (iAR2 receptors reorganized iiito plasma membrane clusters.

I1[;K-239 cells were then exposed to agonist (isoproterenol added to cell iiieclium, as above): the three panels of Row T3 in Figure 4 were taken consecutively after agonist addition (left to riglit, at 0, 3 and 10 minutes post agonist addition). The real-time redistribution of (3arr2-GTP to the receptors over a ten minute time perioci is thus demonstrated by comparing the panels of Row A anci Row B of Figure 4. In Figure 4, arrows inclicate areas of colocalization and the bar=10 microns.

Figure 4 demonstrates that the geometry of the agonist-induced time dependent translocation of (iarr2-GFP to the plasma membrane miniickeci the clistribution of pre-aggregated (I2ARs. This indicates that the primary site targeted by (i-arrestin is the (T2/1R or a closely associated component.

i?XAMPLr 7 Intracellular j3arr2-GrP Targets Membrane Receptors It has been postulated that phosphorylation of GPCRs by GRKs facilitates desensitization by iticreasing their affinity for (i-arrestins.
Gurevich et al., .1. Binl. Chem. 268:16879 (1993); Gurevich et al, .1. 13io1. C.'henr.
268:11628-11638 (1993); Fergusoti et al., Can. .1. I'hysiol. Phnr=ntacr,l.
74:1095 (1996). Wlien expressed in I-IEK-293 cells and exposeci to agonist, mutant Y326A-(i2nRs are not signiticantly pliosphorylated by endogenous GRKs. Barak et al., ]3iochem. 34:15407 (1995); Ferguson et al., .1. Biol. Cheln. 270:24782 (1995). This phosphorylation impairment in Y326A-PAR2s is reversed by overexpression of GRKs in the same cell. Menard et al., 13iochen,. 35:4155 (1996). The Y326A mutant receptor was used to investigate (i-at-restin affinity in Wi,o; the effect of overexpressed GRK on the Y326n-B2AR interactirnl with Parr2-GFP was sliown.

Y326n-p2AR and (iarr2-GFP were co-transfected into IIEK-239 cells, in the absence and presence of co-transfected CiRK. 1f phosphorylation of GPCRs by C:~RKs facilitates desensitization by increasing their affinity for (1-arrestins, tlien overexpression of GRK would result in a noticeable cliffet-ence in Parr2-GrP translocation.

Figure 5 shows the in(luence of ovet-expressed GRK on the reclistributiOn of (3arr2-GTP in I1l;K-293 cells expt-essing the Y326A
phosphorylation impaired P2/1R. Cells witiiout (Row A) and with (RoNv B) overexpressed C;RKs were exposed to agonist, and the real-time redistribution of ~art-2-GFP was observed. Witliottt added GRK. Parr2-GT'P translocation in response to agonist proceeded poorly, as shown in Row A of rigtn=e ~. (iarr2-GFP translocation in cells containing overexpressed GRK (Row B) was nmre robust, indicating an increased affinity of (iart-2-GFP for receptor and the relationship of phosplhorylation and P-arrestin activity.

rXAMPLI; 8 Testiniz of Additional Receptors in the 32AR/rhodopsin Subfamily Twelve different members of the P2AR/rhodopsin subfamily of GPCRs liave been sttidied. Cells expressing a pai-ticular GPCR, and containing S ~arrestin-GFP chinieric proteins were exposed to fcnown agonists for the GI'CR
being studied. In eacii case, an observable transloeation of the (3arrestin-GPP
chimeric proteins from the cell cytosol to tlie cell niembrane was produced witfiin miilutes following addition of the GPCR agonist (data not sliown).

Claims (46)

The embodiments of the invention in which an exclusive property or privilege is claimed are defined as follows:
1. A conjugate comprising a .beta.-arrestin protein and a detectable molecule, wherein said conjugate is capable of indicating intracellular translocation and/or intracellular distribution of the .beta.-arrestin protein.
2. The conjugate according to claim 1, which is a fusion protein.
3. The conjugate of claim 1 or 2, wherein the detectable molecule is an optically detectable molecule.
4. The conjugate of claim 3, wherein the detectable molecule is Green Fluorescent Protein.
5. A nucleic acid construct comprising, in the 5' to 3' direction, a promoter and a nucleic acid segment operatively associated therewith, the nucleic acid segment comprising a sequence encoding a conjugate comprising a .beta.-arrestin protein and a detectable molecule, wherein said conjugate is capable of indicating intracellular translocation and/or intracellular distribution of the .beta.-arrestin protein.
6. The nucleic acid construct according to claim 5, wherein the nucleic acid sequence encoding the detectable molecule encodes an optically detectable molecule.
7. The nucleic acid construct according to claim 6, wherein the nucleic acid sequence encoding the detectable molecule encodes Green Fluorescent Protein.
8. The nucleic acid construct according to any one of claims 5 to 7, including any one or more of the following features:
(i) the construct further comprises a plasmid;
(ii) the construct is a DNA or RNA construct; and (iii) the promoter is .beta.-arrestin promoter.
9. A host cell containing a nucleic acid construct as defined in any one of claims 5 to 8.
10. A host cell comprising a nucleic acid molecule comprising, in the 5' to 3' direction, a promoter operable in the host cell, and a nucleic acid sequence encoding a conjugate comprising a .beta.-arrestin protein and a detectable molecule, the encoding nucleic acid sequence being operably linked to the promoter, and wherein the nucleic acid molecule expresses said conjugate, and wherein said conjugate is capable of indicating intracellular translocation and/or intracellular distribution of the .beta.-arrestin protein.
11. The host cell according to claim 9 or 10, wherein the cell is a bacterial cell, a yeast cell, a fungal cell, a plant cell or an animal cell.
12. The host cell according to claim 11, wherein the animal cell is a mammalian cell.
13. The host cell according to claim 12, wherein the mammalian cell is a HEK-cell or a COS cell.
14. A substrate having deposited thereon a host cell as defined in any one of claims 9 to 13.
15. A method of assessing G protein coupled receptor (GPCR) pathway activity under test conditions, the test conditions optionally being the presence in a test cell of a test kinase or a test G-protein, or exposure of the test cell to a test ligand, or co-expression in the test cell of a second receptor, the method comprising:
(a) providing a test cell that expresses a GPCR, and contains a conjugate comprising a .beta.-arrestin protein and an optically detectable molecule;
(b) exposing the test cell to a known GPCR agonist under test conditions; and then (c) detecting translocation of said conjugate from the cytosol of the test cell to the membrane edge of the test cell;
wherein the translocation of said conjugate in the test cell indicates activation of the GPCR pathway.
16. A method for screening a .beta.-arrestin protein or fragment thereof for the ability to bind to a phosphorylated GPCR, the method comprising:
(a) providing a cell that:
(i) expresses a GPCR; and (ii) contains a conjugate comprising a test .beta.-arrestin protein and an optically detectable molecule;
(b) exposing the cell to a known GPCR agonist; and then (c) detecting translocation of said conjugate from the cytosol of the cell to the membrane edge of the cell;
wherein translocation of said conjugate in the test cell indicates 0-arrestin protein binding to the phosphorylated GPCR.
17. A method for screening a test compound for G protein coupled receptor (GPCR) agonist activity, the method comprising:
(a) providing a cell expressing a GPCR and containing a conjugate comprising a (i-arrestin protein and an optically detectable molecule;
(b) exposing the cell to the test compound, the test compound optionally being in aqueous solution; and then (c) detecting translocation of said conjugate from the cytosol of the cell to the membrane edge of the cell, the cells optionally being deposited on' a substrate;
wherein movement of said conjugate from the cytosol to the membrane edge of the cell after exposure of the cell to the test compound indicates GPCR agonist activity of the test compound.
18. The method according to claim 17, wherein the cell expresses any one or more of the following:
(i) a known GPCR;
(ii) an GPCR with an unknown function;
(iii) an odorant GPCR;
(iv) a .beta.-adrenergic GPCR.
19. The method according to claim 17 or 18, wherein the detectable molecule is a Green Fluorescent Protein.
20. The method according to any one of claims 17 to 19, wherein the cell is a host cell as defined in any one of claims 11 to 13.
21. The method according to any one of claims 17, 18 or 20, wherein the cell normally expresses a GPCR or optionally the cell has been transformed to express a GPCR not normally expressed by such a cell.
22. A method of screening a sample solution for the presence of an agonist to a G protein coupled receptor (GPCR), the method comprising:
(a) providing a cell expressing a GPCR and containing a conjugate, the conjugate comprising a .beta.-arrestin protein and an optically detectable molecule;
(b) exposing the cell to a sample solution; and then (c) detecting translocation of said conjugate from the cytosol of the cell to the membrane edge of the cell;
wherein movement of said conjugate from the cytosol to the membrane edge of the cell after exposure of the cell to the sample solution indicates the sample solution contains an agonist for a GPCR expressed in the cell.
23. The method according to claim 22, wherein the cell expresses any one or more of the following:
(i) a known GPCR;
(ii) an GPCR with an unknown function;
(iii) an odorant GPCR;
(iv) a .beta.-adrenergic GPCR.
24. The method according to claim 22 or 23, wherein the detectable molecule is a Green Fluorescent Protein.
25. The method according to any one of claims 22 to 24, wherein the cell is a host cell as defined in any one of claims 11 to 13.
26. A method of screening a cell for the presence of a G protein coupled receptor (GPCR), the method comprising:

(a) providing a test cell, said test cell containing a conjugate comprising a .beta.-arrestin protein and an optically detectable molecule;
(b) exposing the test cell to a solution containing a GPCR agonist; and (c) detecting translocation of said conjugate from the cytosol of the cell to the membrane edge of the cell;
wherein movement of said conjugate from the cytosol to the membrane edge of the test cell after exposure of the test cell to the GPCR agonist indicates that the test cell contains a GPCR.
27. The method according to claim 21, wherein the detectable molecule is a Green Fluorescent Protein.
28. A method of screening a plurality of cells for those cells which contain a G protein coupled receptor (GPCR), the method comprising:
(a) providing a plurality of test cells, said test cells containing a conjugate comprising a P-arrestin protein and an optically detectable molecule;
(b) exposing the test cells to one, or a plurality of, known GPCR agonist(s);
and (c) detecting those cells in which said conjugate is translocated from the cytosol of the cell to the membrane edge of the cell;
wherein movement of said conjugate from the cytosol to the membrane edge of a cell after exposure to the GPCR agonist indicates that the cell contains a GPCR for that known GPCR agonist.
29. The method according to claim 28, wherein the plurality of cells are contained in a tissue.
30. The method according to claim 28, wherein the plurality of cells are contained in an organ.
31. The method according to any one of claims 28 to 30, wherein the detectable molecule is a Green Fluorescent Protein.
32. Use of a GPCR-translocatable .beta.-arrestin protein coupled to a detectable molecule to screen a test compound for GPCR agonist function, by detecting for intracellular translocation from the cell cytosol to the membrane edge of the cell of the beta-arrestin protein responsive to exposure of the cell to the test compound.
33. A method of screening a test compound for G protein coupled receptor (GPCR) antagonist activity, the method comprising:
(a) providing a cell expressing a GPCR, and containing a conjugate, the conjugate comprising a .beta.-arrestin protein and an optically detectable molecule;
(b) exposing the cell to a test compound;
(c) exposing the cell to a GPCR agonist; and (d) detecting translocation of said conjugate from the cytosol of the cell to the membrane edge of the cell;
the cell optionally being deposited on a substrate;
wherein exposure to the agonist occurs at the same time as, or subsequent to, exposure to the test compound, and wherein movement of said conjugate from the cytosol to the membrane edge of the cell after exposure of the cell to the test compound indicates that the test compound is not a GPCR antagonist.
34. The method according to claim 33, wherein the detectable molecule is a Green Fluorescent Protein.
35. The method according to claims 33 or 34, wherein the cell is a host cell as defined in any one of claims 11 to 13.
36. The method according to any one of claims 33 to 35, wherein the cell expresses any one or more of the following:
(i) a known GPCR;
(ii) an GPCR with an unknown function;
(iii) an odorant GPCR;
(iv) a .beta.-adrenergic GPCR.
37. A method of screening a test compound for G protein coupled receptor (GPCR) antagonist activity, the test compound optionally being in aqueous solution, the method comprising:
(a) providing a cell expressing a GPCR and containing a conjugate, the conjugate comprising a .beta.-arrestin protein and an optically detectable molecule;
(b) exposing the cell to a GPCR agonist so that translocation of said conjugate from the cytosol of the cell to the membrane edge of the cell occurs;
(c) exposing the cell to a test compound, the cells optionally being deposited on a substrate;

wherein exposure to the agonist occurs prior to exposure to the test compound, and wherein movement of said conjugate molecule from the membrane edge of the cell to the cytosol after exposure of the cell to the test compound indicates that the test compound has a GPCR antagonist activity.
38. The method according to claim 37, wherein the detectable molecule is a Green Fluorescent Protein.
39. The method according to claim 37 or 38, wherein the cell is a host cell as defined in any one of claims 11 to 13.
40. The use of a GPCR-translocatable beta-arrestin protein coupled to a detectable molecule, to screen a test compound for GPCR antagonist function, by detecting translocation from the membrane edge of the cell to the cell cytosol of the .beta.-arrestin protein responsive to exposure of the cell to the test compound.
41. A substrate having deposited thereon a plurality of cells, said cells expressing a GPCR and a labelled .beta.-arrestin protein, wherein the label is capable of indicating intracellular translocation and/or distribution intracellular of the .beta.-arrestin.
42. The substrate according to claim 41, wherein the label is an optically detectable molecule.
43. The substrate according to claim 42, wherein the detectable molecule is Green Fluorescent Protein.
44. The substrate according to any one of claims 41 to 43, wherein said cells are host cells as defined in any one of claims 11 to 13.
45. The substrate according to any one of claims 41 to 44, wherein the cells express any one or more of the following:
(i) a known GPCR;
(ii) an GPCR with an unknown function;
(iii) an odorant GPCR;
(iv) a .beta.-adrenergic GPCR.
46. The substrate according to any one of claims 14 and 41 to 45, wherein the substrate is made of a material selected from glass, plastic, ceramic, semiconductor, silica, fiber optic, diamond, biocompatible monomer, and biocompatible polymer materials.
CA002305810A 1997-06-05 1998-06-04 Methods of assaying receptor activity and constructs useful in such methods Expired - Lifetime CA2305810C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/869,568 US5891646A (en) 1997-06-05 1997-06-05 Methods of assaying receptor activity and constructs useful in such methods
US08/869,568 1997-06-05
PCT/US1998/011628 WO1998055635A1 (en) 1997-06-05 1998-06-04 Methods of assaying receptor activity and constructs useful in such methods

Publications (2)

Publication Number Publication Date
CA2305810A1 CA2305810A1 (en) 1998-12-10
CA2305810C true CA2305810C (en) 2007-11-20

Family

ID=25353824

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002305810A Expired - Lifetime CA2305810C (en) 1997-06-05 1998-06-04 Methods of assaying receptor activity and constructs useful in such methods

Country Status (10)

Country Link
US (5) US5891646A (en)
EP (2) EP1441032B1 (en)
AT (2) ATE482276T1 (en)
AU (1) AU759347B2 (en)
CA (1) CA2305810C (en)
DE (3) DE69841908D1 (en)
DK (1) DK1015608T3 (en)
ES (1) ES2182736T3 (en)
PT (1) PT1015608E (en)
WO (1) WO1998055635A1 (en)

Families Citing this family (85)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5989835A (en) 1997-02-27 1999-11-23 Cellomics, Inc. System for cell-based screening
US7853411B2 (en) * 1997-02-27 2010-12-14 Cellomics, Inc. System for cell-based screening
US6759206B1 (en) 1997-02-27 2004-07-06 Cellomics, Inc. System for cell-based screening
US6528271B1 (en) * 1997-06-05 2003-03-04 Duke University Inhibition of βarrestin mediated effects prolongs and potentiates opioid receptor-mediated analgesia
US7541151B2 (en) * 1997-06-05 2009-06-02 Duke University Single-cell biosensor for the measurement of GPCR ligands in a test sample
US20050221280A1 (en) 1998-02-02 2005-10-06 Odyssey Thera, Inc. Protein-protein interactions for pharmacological profiling
WO1999049019A2 (en) 1998-03-27 1999-09-30 Prolume, Ltd. Luciferases, fluorescent proteins, nucleic acids encoding the luciferases and fluorescent proteins and the use thereof in diagnostics
US6692696B1 (en) 1998-06-18 2004-02-17 ARETé ASSOCIATES Biosensor
US6448005B1 (en) * 1998-06-30 2002-09-10 Millennium Pharmaceuticals, Inc. 14723 Receptor, a novel G-protein coupled receptor
US7534579B2 (en) * 1998-06-30 2009-05-19 Millennium Pharmaceuticals, Inc. 14273 receptor, a novel G-protein coupled receptor
WO2000000611A2 (en) * 1998-06-30 2000-01-06 Millennium Pharmaceuticals, Inc. 14273 receptor, a g-protein coupled receptor
EP1109931A4 (en) * 1998-09-03 2004-12-15 Univ Loma Linda METHOD FOR STUDYING PROTEIN INTERACTIONS $i(IN VIVO)
US20030017528A1 (en) * 1998-11-20 2003-01-23 Ruoping Chen Human orphan G protein-coupled receptors
USRE42190E1 (en) 1998-11-20 2011-03-01 Arena Pharmaceuticals, Inc. Method of identifying a compound for inhibiting or stimulating human G protein-coupled receptors
US7816492B2 (en) * 1998-11-20 2010-10-19 Arena Pharmaceuticals, Inc. Human G protein-coupled receptors
US20020038013A1 (en) * 2000-02-04 2002-03-28 Gregory Donoho Novel human membrane proteins and polynucleotides encoding the same
US6673554B1 (en) * 1999-06-14 2004-01-06 Trellie Bioinformatics, Inc. Protein localization assays for toxicity and antidotes thereto
CN1301726A (en) * 1999-12-27 2001-07-04 上海博德基因开发有限公司 New polypeptide-rhodopsin arrestin family 11 and polynucleotide coding such polypeptide
US6893827B1 (en) * 2000-02-07 2005-05-17 Applera Corporation Receptor function assay for G-protein coupled receptors and orphan receptors by reporter enzyme mutant complementation
JP2004500089A (en) * 2000-02-07 2004-01-08 トロピックス・インコーポレーテッド Enzyme assay for G protein-coupled receptor
US7109315B2 (en) * 2000-03-15 2006-09-19 Bruce J. Bryan Renilla reniformis fluorescent proteins, nucleic acids encoding the fluorescent proteins and the use thereof in diagnostics, high throughput screening and novelty items
WO2002018590A2 (en) * 2000-08-30 2002-03-07 John Hopkins University School Of Medicine Identification of activated receptors and ion channels
WO2002035234A1 (en) * 2000-10-26 2002-05-02 The University Of British Columbia A functional assay for g-protein-coupled receptors based on insect cells
US7163800B2 (en) * 2000-11-03 2007-01-16 Molecular Devices Corporation Methods of screening compositions for G protein-coupled receptor desensitization inhibitory activity
US20050136431A1 (en) * 2000-11-03 2005-06-23 Oakley Robert H. Methods of screening compositions for G protein-coupled receptor desensitization inhibitory activity
US7018812B2 (en) * 2000-11-03 2006-03-28 Duke University Modified G-protein coupled receptors
US20020123071A1 (en) * 2000-12-04 2002-09-05 Knudsen Sanne Moller Method of identifying compounds capable of acting as agonists or antagonists of G-protein coupled receptors
AU2002220526A (en) * 2000-12-04 2002-06-18 Novo Nordisk As Method of identifying compounds capable of acting as agonists or antagonists of g-protein coupled receptors
US7279324B2 (en) * 2001-01-23 2007-10-09 Duke University Nucleic acid encoding G-protein coupled receptor with modified DRY motif
US7166699B2 (en) * 2001-01-26 2007-01-23 Vanderbilt University Mosquito arrestin 1 polypeptides
CA2438212A1 (en) * 2001-02-08 2002-09-06 Temple University - Of The Commonwealth System Of Higher Education Biosensor for detecting chemical agents
US20030013137A1 (en) * 2001-03-13 2003-01-16 Barak Larry S. Automated methods of detecting receptor activity
JP2004537297A (en) * 2001-06-07 2004-12-16 ワイス G-protein coupled receptor and uses thereof
US20030119024A1 (en) * 2001-07-20 2003-06-26 Linsley Peter S. Genes and proteins associated with T cell activation
WO2003061594A2 (en) * 2002-01-23 2003-07-31 The Regents Of The University Of California Methods and compositions for reducing the development of drug tolerance and/or physical dependence
US20030182669A1 (en) * 2002-03-19 2003-09-25 Rockman Howard A. Phosphoinositide 3-kinase mediated inhibition of GPCRs
CA2521946C (en) 2002-04-12 2013-10-15 Brian F. O'dowd Method of identifying transmembrane protein-interacting compounds
EP1504030B1 (en) * 2002-05-13 2009-01-07 Molecular Devices Corporation Constitutively translocating cell line
US7297503B2 (en) * 2002-10-25 2007-11-20 Molecular Devices Corporation Methods of identifying reduced internalization transmembrane receptor agonists
FR2851379B1 (en) * 2003-02-18 2008-02-01 Cit Alcatel CONVERTER IN DIRECT TRANSFER OF ENERGY
US7290215B2 (en) * 2002-06-03 2007-10-30 Microsoft Corporation Dynamic wizard interface system and method
WO2004014296A2 (en) * 2002-08-13 2004-02-19 U.S. Department Of Veterans Affairs Method of detecting and preventing alzheimer’s disease, particularly at prodromal and early stages
EP1554581A2 (en) * 2002-10-11 2005-07-20 7TM Pharma A/S Method for enhancing and prolonging the bioluminescence resonance energy transfer (bret) signal in a bret assay and a substrate solution for use in a bret assay
CA2514179A1 (en) * 2003-01-24 2004-08-12 Duke University Modified trafficking patterns for arrestin and g-protein-coupled receptors via arrestin-ubiquitin chimera
CN1894581B (en) 2003-07-09 2012-02-01 生命技术公司 Method for assaying protein-protein interaction
US20070224615A1 (en) * 2003-07-09 2007-09-27 Invitrogen Corporation Methods for assaying protein-protein interactions
US20070196865A1 (en) * 2003-09-16 2007-08-23 Molecular Devices Corporation Methods Of Screening Compositions For G Protein-Coupled Receptors Aganist Agonists
US7488583B2 (en) * 2003-09-25 2009-02-10 Odyssey Thera, Inc. Fragment complementation assays for G-protein-coupled receptors and their signaling pathways
WO2005038455A1 (en) * 2003-10-17 2005-04-28 National Centre For Biological Sciences A novel assay for screening antipsychotic drugs
US20060292559A1 (en) * 2005-06-23 2006-12-28 Beckman Coulter, Inc. Cell-based microarrays, and methods for their preparation and use
ES2580108T3 (en) * 2005-07-11 2016-08-19 Aerie Pharmaceuticals, Inc Isoquinoline compounds
US20070135499A1 (en) * 2005-07-11 2007-06-14 Aerie Pharmaceuticals, Inc. Hydrazide compounds
US7763437B1 (en) * 2005-07-15 2010-07-27 Duke University Methods for identifying compounds that regulate β-Arrestin signaling complexes
US20070218456A1 (en) * 2006-02-08 2007-09-20 Invitrogen Corporation Cellular assays for signaling receptors
WO2007127538A1 (en) 2006-03-16 2007-11-08 Invitrogen Corporation Methods for assaying protein-protein interaction
CA2664335C (en) 2006-09-20 2014-12-02 Boehringer Ingelheim International Gmbh Rho kinase inhibitors
CA2669088C (en) * 2006-11-10 2016-04-05 Dimerix Bioscience Pty Ltd Detection system and uses therefor
AU2007336242B2 (en) 2006-12-19 2012-08-30 Ablynx N.V. Amino acid sequences directed against GPCRs and polypeptides comprising the same for the treatment of GPCR-related diseases and disorders
US8455513B2 (en) 2007-01-10 2013-06-04 Aerie Pharmaceuticals, Inc. 6-aminoisoquinoline compounds
JP2010529127A (en) 2007-06-05 2010-08-26 イエール ユニバーシティ Inhibitor of receptor tyrosine kinase and method of using the same
WO2009005815A1 (en) * 2007-07-05 2009-01-08 Enhanced Pharmaceuticals, Inc. Method for determining drug-molecular combinations that modulate and enhance the therapeutic safety and efficacy of biological or pharmaceutical drugs
BRPI0816289B1 (en) * 2007-09-04 2019-01-15 Sanofi Aventis method for identifying a compound that modulates a protein-protein interaction between a first protein and a second protein and assay system
US8455514B2 (en) * 2008-01-17 2013-06-04 Aerie Pharmaceuticals, Inc. 6-and 7-amino isoquinoline compounds and methods for making and using the same
JP5259207B2 (en) * 2008-02-05 2013-08-07 オリンパス株式会社 Cell image analysis apparatus and method and software thereof
US8450344B2 (en) 2008-07-25 2013-05-28 Aerie Pharmaceuticals, Inc. Beta- and gamma-amino-isoquinoline amide compounds and substituted benzamide compounds
FR2934684B1 (en) 2008-07-31 2012-11-16 Cis Bio Int METHOD OF DETECTING INTERNALIZATION OF MEMBRANE PROTEINS.
GB0817861D0 (en) * 2008-09-30 2008-11-05 Ge Healthcare Uk Ltd Methods and compounds for testing binding of a ligand to a g protein-coupled receptor
US8753604B2 (en) * 2008-12-23 2014-06-17 Sanford-Burnham Medical Research Institute Methods and compositions for synaphically-targeted treatment for cancer
GB0905419D0 (en) 2009-03-30 2009-05-13 Ge Healthcare Uk Ltd Methods for testing ligand binding to G protein-coupled receptors
ES2834451T3 (en) 2009-05-01 2021-06-17 Aerie Pharmaceuticals Inc Dual Mechanism Inhibitors for Treating Diseases
EP2507383B1 (en) * 2009-12-02 2013-10-16 Unilever NV Improved method for screening a potential modulator compound of a taste receptor
EP4215194A1 (en) 2011-01-11 2023-07-26 Dimerix Bioscience Pty Ltd Combination therapy
US10853819B2 (en) 2011-04-14 2020-12-01 Elwha Llc Cost-effective resource apportionment technologies suitable for facilitating therapies
US10445846B2 (en) 2011-04-14 2019-10-15 Elwha Llc Cost-effective resource apportionment technologies suitable for facilitating therapies
US9128079B2 (en) 2011-08-08 2015-09-08 The Coca-Cola Company Methods of using lung or bronchial epithelial cells to identify bitter taste modulators
US8514067B2 (en) 2011-08-16 2013-08-20 Elwha Llc Systematic distillation of status data relating to regimen compliance
JP5857168B2 (en) 2012-11-08 2016-02-10 ファイザー・インク Heteroaromatic compounds as dopamine D1 ligands and their use
PT3811943T (en) 2013-03-15 2023-03-15 Aerie Pharmaceuticals Inc Compound for use in the treatment of ocular disorders
RU2016102922A (en) 2013-06-29 2017-08-03 Фирмениш Са METHODS FOR IDENTIFICATION, ISOLATION AND APPLICATION OF RECEPTORS OF FRAGRANT SUBSTANCES AND AROMAS
WO2015050959A1 (en) 2013-10-01 2015-04-09 Yale University Anti-kit antibodies and methods of use thereof
US10550087B2 (en) 2015-11-17 2020-02-04 Aerie Pharmaceuticals, Inc. Process for the preparation of kinase inhibitors and intermediates thereof
US9643927B1 (en) 2015-11-17 2017-05-09 Aerie Pharmaceuticals, Inc. Process for the preparation of kinase inhibitors and intermediates thereof
CN109640966A (en) 2016-08-31 2019-04-16 爱瑞制药公司 Ophthalmic composition
AU2018243687C1 (en) 2017-03-31 2020-12-24 Aerie Pharmaceuticals, Inc. Aryl cyclopropyl-amino-isoquinolinyl amide compounds
EP3849555A4 (en) 2018-09-14 2022-05-04 Aerie Pharmaceuticals, Inc. Aryl cyclopropyl-amino-isoquinolinyl amide compounds

Family Cites Families (109)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US36547A (en) * 1862-09-23 Improvement in locks
US31006A (en) * 1861-01-01 Feeding mechanism for spoke-machines
USRE31006E (en) * 1968-09-24 1982-08-03 Akzona Incorporated Process for the demonstration and determination of reaction components having specific binding affinity for each other
US3654090A (en) * 1968-09-24 1972-04-04 Organon Method for the determination of antigens and antibodies
NL154598B (en) * 1970-11-10 1977-09-15 Organon Nv PROCEDURE FOR DETERMINING AND DETERMINING LOW MOLECULAR COMPOUNDS AND PROTEINS THAT CAN SPECIFICALLY BIND THESE COMPOUNDS AND TEST PACKAGING.
US4016043A (en) * 1975-09-04 1977-04-05 Akzona Incorporated Enzymatic immunological method for the determination of antigens and antibodies
US4219335A (en) * 1978-09-18 1980-08-26 E. I. Du Pont De Nemours And Company Immunochemical testing using tagged reagents
US4491632A (en) * 1979-10-22 1985-01-01 The Massachusetts General Hospital Process for producing antibodies to hepatitis virus and cell lines therefor
US4444887A (en) * 1979-12-10 1984-04-24 Sloan-Kettering Institute Process for making human antibody producing B-lymphocytes
US4342566A (en) * 1980-02-22 1982-08-03 Scripps Clinic & Research Foundation Solid phase anti-C3 assay for detection of immune complexes
EP0043718B1 (en) * 1980-07-07 1984-11-28 National Research Development Corporation Improvements in or relating to cell lines
US4341761A (en) * 1980-07-25 1982-07-27 E. I. Du Pont De Nemours And Company Antibodies to immunogenic peptides and their use to purify human fibroblast interferon
US4324633A (en) * 1980-10-20 1982-04-13 Lovejoy Curtis N Electrolytic apparatus for treating continuous strip material
US4466917A (en) * 1981-02-12 1984-08-21 New York University Malaria vaccine
US4493890A (en) 1981-03-23 1985-01-15 Miles Laboratories, Inc. Activated apoglucose oxidase and its use in specific binding assays
US4451570A (en) * 1981-03-26 1984-05-29 The Regents Of The University Of California Immunoglobulin-secreting human hybridomas from a cultured human lymphoblastoid cell line
US4534899A (en) * 1981-07-20 1985-08-13 Lipid Specialties, Inc. Synthetic phospholipid compounds
US4426330A (en) * 1981-07-20 1984-01-17 Lipid Specialties, Inc. Synthetic phospholipid compounds
US4399121A (en) * 1981-11-04 1983-08-16 Miles Laboratories, Inc. Iodothyronine immunogens and antibodies
US4427783A (en) * 1981-12-14 1984-01-24 Hoffmann-La Roche Inc. Immunoassay of thymosin α1
GB8308235D0 (en) * 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5972629A (en) * 1984-08-17 1999-10-26 The Scripps Research Institute Method for characterizing antigenic reactivity of biological sample
US4493795A (en) * 1983-10-17 1985-01-15 Syntex (U.S.A.) Inc. Synthetic peptide sequences useful in biological and pharmaceutical applications and methods of manufacture
US5034506A (en) * 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US5583024A (en) * 1985-12-02 1996-12-10 The Regents Of The University Of California Recombinant expression of Coleoptera luciferase
US4908773A (en) * 1987-04-06 1990-03-13 Genex Corporation Computer designed stabilized proteins and method for producing same
US4981784A (en) * 1987-12-02 1991-01-01 The Salk Institute For Biological Studies Retinoic acid receptor method
AU5154390A (en) * 1989-02-15 1990-09-05 Microtek Medical, Inc. Biocompatible material and prosthesis
US5080924A (en) * 1989-04-24 1992-01-14 Drexel University Method of making biocompatible, surface modified materials
US5744101A (en) * 1989-06-07 1998-04-28 Affymax Technologies N.V. Photolabile nucleoside protecting groups
US5284746A (en) * 1990-02-08 1994-02-08 Zymogenetics, Inc. Methods of producing hybrid G protein-coupled receptors
US5665710A (en) * 1990-04-30 1997-09-09 Georgetown University Method of making liposomal oligodeoxynucleotide compositions
US5462856A (en) * 1990-07-19 1995-10-31 Bunsen Rush Laboratories, Inc. Methods for identifying chemicals that act as agonists or antagonists for receptors and other proteins involved in signal transduction via pathways that utilize G-proteins
US5602240A (en) * 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
ATE199391T1 (en) * 1990-09-13 2001-03-15 Univ Duke EXPRESSION OF RECEPTORS COUPLED TO G-PROTEIN IN YEAST
US5569824A (en) * 1991-01-04 1996-10-29 Baylor College Of Medicine Transgenic mice containing a disrupted p53 gene
JP3220180B2 (en) * 1991-05-23 2001-10-22 三菱化学株式会社 Drug-containing protein-bound liposomes
US5539082A (en) * 1993-04-26 1996-07-23 Nielsen; Peter E. Peptide nucleic acids
US5714331A (en) * 1991-05-24 1998-02-03 Buchardt, Deceased; Ole Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
US5719262A (en) * 1993-11-22 1998-02-17 Buchardt, Deceased; Ole Peptide nucleic acids having amino acid side chains
WO1993003382A2 (en) * 1991-08-01 1993-02-18 Pharmaceutical Discovery Corporation Characterization of specific drug receptors with fluorescent ligands
US5576436A (en) * 1991-08-01 1996-11-19 Pharmaceutical Discovery Corporation Fluorescent ligands
EP0668930B1 (en) * 1991-10-01 2002-12-18 THE GOVERNMENT OF THE UNITED STATES OF AMERICA as represented by the SECRETARY OF THE DEPARTMENT OF HEALTH AND HUMAN SERVICES A method of identifying ligands and antagonists of ligands
US5352660A (en) * 1991-10-31 1994-10-04 Mount Sinai Hospital Corporation Method for assaying for a substance that affects a SH2-phosphorylated ligand regulatory system
US5324633A (en) * 1991-11-22 1994-06-28 Affymax Technologies N.V. Method and apparatus for measuring binding affinity
DE4138621A1 (en) * 1991-11-25 1993-06-17 Boehringer Ingelheim Int METHOD FOR SCREENING SUBSTANCES WITH MODULATING EFFECT ON A RECEPTACLE-RELATED CELLULAR SIGNAL TRANSMISSION PATH
US5670113A (en) * 1991-12-20 1997-09-23 Sibia Neurosciences, Inc. Automated analysis equipment and assay method for detecting cell surface protein and/or cytoplasmic receptor function using same
US6096756A (en) * 1992-09-21 2000-08-01 Albert Einstein College Of Medicine Of Yeshiva University Method of simultaneously enhancing analgesic potency and attenuating dependence liability caused by morphine and other bimodally-acting opioid agonists
USRE36547E (en) * 1992-09-21 2000-02-01 Albert Einstein College Of Medicine Of Yeshiva University Method of simultaneously enhancing analgesic potency and attenuating dependence liability caused by exogenous and endogenous opioid agonists
US5597699A (en) * 1992-09-30 1997-01-28 Lanzara; Richard G. Method for determining drug compositions to prevent desensitization of cellular receptors
US5366889A (en) * 1992-11-30 1994-11-22 The General Hospital Corporation DNA encoding a protein-coupled receptor kinase
US5360728A (en) * 1992-12-01 1994-11-01 Woods Hole Oceanographic Institution (W.H.O.I.) Modified apoaequorin having increased bioluminescent activity
WO1994016684A1 (en) * 1993-01-26 1994-08-04 The Johns Hopkins University INHIBITION OF βARK AND β-ARRESTIN
US6103492A (en) * 1993-03-08 2000-08-15 Indiana University Polynucleotide encoding mu opioid receptor
US6100042A (en) * 1993-03-31 2000-08-08 Cadus Pharmaceutical Corporation Yeast cells engineered to produce pheromone system protein surrogates, and uses therefor
US5328687A (en) * 1993-03-31 1994-07-12 Tri-Point Medical L.P. Biocompatible monomer and polymer compositions
US6255059B1 (en) * 1993-03-31 2001-07-03 Cadus Pharmaceutical Corporation Methods for identifying G protein coupled receptor effectors
US5912122A (en) * 1993-06-04 1999-06-15 Sibia Neurosciences, Inc. Nucleic acids encoding and method for detecting nucleic acid encoding human metabotropic glutamate receptor subtype mGluR6
US6007986A (en) * 1993-06-23 1999-12-28 The Regents Of The University Of California Methods for anti-addictive narcotic analgesic activity screening
US5882944A (en) 1993-06-23 1999-03-16 The Regents Of The University Of California Methods for G protein coupled receptor activity screening
US5491084A (en) * 1993-09-10 1996-02-13 The Trustees Of Columbia University In The City Of New York Uses of green-fluorescent protein
US5532151A (en) * 1993-09-17 1996-07-02 Icos Corporation G protein-coupled receptor kinase GRK6
US5658783A (en) * 1993-11-08 1997-08-19 State Of Oregon, Acting By And Through The Oregon State Board Of Higher Education On Behalf Of The Oregon Health Sciences University, A Non-Profit Organization Mammalian methadone-specific opioid receptor gene and uses
CA2177388A1 (en) * 1993-11-26 1995-06-01 Lawrence B. Hendry Design of drugs involving receptor-ligand-dna interactions
WO1995021191A1 (en) 1994-02-04 1995-08-10 William Ward Bioluminescent indicator based upon the expression of a gene for a modified green-fluorescent protein
GB9403600D0 (en) * 1994-02-24 1994-04-13 Univ Glasgow Three dimensional hormone structure
WO1995023231A1 (en) * 1994-02-28 1995-08-31 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Cell lines for the identification of substances affecting insulin receptor mediated signal transduction
US5627039A (en) * 1994-03-18 1997-05-06 Baylor College Of Medicine Mortalin and methods for determining complementation group assignment of cancer cells
US5569827A (en) * 1994-06-06 1996-10-29 Universite De Montreal Transgenic mouse for the neuronal expression of HIV gp160
US5661184A (en) * 1994-08-12 1997-08-26 Eli Lilly And Company Psychiatric agents
US5463564A (en) * 1994-09-16 1995-10-31 3-Dimensional Pharmaceuticals, Inc. System and method of automatically generating chemical compounds with desired properties
US5777079A (en) * 1994-11-10 1998-07-07 The Regents Of The University Of California Modified green fluorescent proteins
US5625048A (en) 1994-11-10 1997-04-29 The Regents Of The University Of California Modified green fluorescent proteins
US5744313A (en) * 1994-12-09 1998-04-28 The Regents Of The University Of California Assay employing novel protein domain which binds tyrosine phosphorylated proteins
US5958713A (en) * 1995-01-31 1999-09-28 Novo Nordisk A/S Method of detecting biologically active substances by using green fluorescent protein
ES2170220T3 (en) 1995-01-31 2002-08-01 Bioimage As PROCEDURE TO DETECT BIOLOGICALLY ACTIVE SUBSTANCES.
WO1996027027A1 (en) 1995-03-02 1996-09-06 Rutgers The State University Of New Jersey Improved method for purifying green fluorescent protein
GB9504446D0 (en) 1995-03-06 1995-04-26 Medical Res Council Improvements in or relating to gene expression
US6025129A (en) * 1995-04-25 2000-02-15 Irori Remotely programmable matrices with memories and uses thereof
US6017496A (en) * 1995-06-07 2000-01-25 Irori Matrices with memories and uses thereof
US6100026A (en) * 1995-04-25 2000-08-08 Irori Matrices with memories and uses thereof
US5545531A (en) * 1995-06-07 1996-08-13 Affymax Technologies N.V. Methods for making a device for concurrently processing multiple biological chip assays
US5767337A (en) * 1995-07-31 1998-06-16 Duke University Creation of human apolipoprotein E isoform specific transgenic mice in apolipoprotein deficient "knockout" mice
JPH11171896A (en) 1995-09-19 1999-06-29 Kirin Brewery Co Ltd New peptide compound and its pharmaceutical composition
US5770176A (en) * 1995-12-08 1998-06-23 Chiron Diagnostics Corporation Assays for functional nuclear receptors
US5874304A (en) * 1996-01-18 1999-02-23 University Of Florida Research Foundation, Inc. Humanized green fluorescent protein genes and methods
US6027890A (en) * 1996-01-23 2000-02-22 Rapigene, Inc. Methods and compositions for enhancing sensitivity in the analysis of biological-based assays
US5804387A (en) * 1996-02-01 1998-09-08 The Board Of Trustees Of The Leland Stanford Junior University FACS-optimized mutants of the green fluorescent protein (GFP)
US5989835A (en) * 1997-02-27 1999-11-23 Cellomics, Inc. System for cell-based screening
US5912137A (en) * 1996-07-16 1999-06-15 The Regents Of The University Of California Assays for protein kinases using fluorescent
US5912138A (en) * 1996-07-25 1999-06-15 Cold Spring Harbor Laboratory Substrate trapping protein tyrosine phosphatases
US5804436A (en) * 1996-08-02 1998-09-08 Axiom Biotechnologies, Inc. Apparatus and method for real-time measurement of cellular response
US5874268A (en) * 1996-09-23 1999-02-23 Duke University Method of introducing exogenous compounds into cells by electroporation and apparatus for same
US6087115A (en) * 1997-01-22 2000-07-11 Cornell Research Foundation, Inc. Methods of identifying negative antagonists for G protein coupled receptors
US5998204A (en) * 1997-03-14 1999-12-07 The Regents Of The University Of California Fluorescent protein sensors for detection of analytes
US5882067A (en) * 1997-06-09 1999-03-16 Sunrise Medical Hhg Inc. Rigid seat for folding invalid walker
US5972639A (en) * 1997-07-24 1999-10-26 Irori Fluorescence-based assays for measuring cell proliferation
US5987390A (en) * 1997-10-28 1999-11-16 Smithkline Beecham Corporation Methods and systems for identification of protein classes
WO1999052927A1 (en) * 1998-04-14 1999-10-21 Arena Pharmaceuticals, Inc. Non-endogenous, constitutively activated human serotonin receptors and small molecule modulators thereof
US6140509A (en) * 1998-06-26 2000-10-31 Arena Pharmaceuticals, Inc. Non-endogenous, constitutively activated human serotonin receptors and small molecule modulators thereof
US6438235B2 (en) * 1998-08-05 2002-08-20 Hewlett-Packard Company Media content protection utilizing public key cryptography
US6150393A (en) * 1998-12-18 2000-11-21 Arena Pharmaceuticals, Inc. Small molecule modulators of non-endogenous, constitutively activated human serotonin receptors
US6463534B1 (en) * 1999-03-26 2002-10-08 Motorola, Inc. Secure wireless electronic-commerce system with wireless network domain
US7073063B2 (en) * 1999-03-27 2006-07-04 Microsoft Corporation Binding a digital license to a portable device or the like in a digital rights management (DRM) system and checking out/checking in the digital license to/from the portable device or the like
US6697944B1 (en) * 1999-10-01 2004-02-24 Microsoft Corporation Digital content distribution, transmission and protection system and method, and portable device for use therewith
US7012503B2 (en) * 1999-11-30 2006-03-14 Bording Data A/S Electronic key device a system and a method of managing electronic key information
US6915425B2 (en) * 2000-12-13 2005-07-05 Aladdin Knowledge Systems, Ltd. System for permitting off-line playback of digital content, and for managing content rights
US6975202B1 (en) * 2000-11-21 2005-12-13 International Business Machines Corporation Electronic key system, apparatus and method

Also Published As

Publication number Publication date
WO1998055635A9 (en) 2003-05-08
ATE482276T1 (en) 2010-10-15
EP1441032A1 (en) 2004-07-28
ATE262040T1 (en) 2004-04-15
AU759347B2 (en) 2003-04-10
DE69822484D1 (en) 2004-04-22
US6110693A (en) 2000-08-29
DE69822484T2 (en) 2005-01-13
WO1998055635A1 (en) 1998-12-10
DK1015608T3 (en) 2004-06-01
EP1015608B1 (en) 2004-03-17
US20060188944A1 (en) 2006-08-24
US7138240B2 (en) 2006-11-21
US20080261256A1 (en) 2008-10-23
US20040101887A1 (en) 2004-05-27
AU7725598A (en) 1998-12-21
EP1441032B1 (en) 2010-09-22
DE69841908D1 (en) 2010-11-04
ES2182736T1 (en) 2003-03-16
US5891646A (en) 1999-04-06
ES2182736T3 (en) 2004-09-01
CA2305810A1 (en) 1998-12-10
PT1015608E (en) 2004-07-30
DE1015608T1 (en) 2002-11-14
EP1015608A1 (en) 2000-07-05

Similar Documents

Publication Publication Date Title
CA2305810C (en) Methods of assaying receptor activity and constructs useful in such methods
Al-Ani et al. Proteinase-activated receptor 2 (PAR2): development of a ligand-binding assay correlating with activation of PAR2 by PAR1-and PAR2-derived peptide ligands
CN113501881B (en) Fusion proteins
JPWO2019035476A1 (en) Method of quantifying odor, cells used for it and method of manufacturing the cells
US6770449B2 (en) Methods of assaying receptor activity and constructs useful in such methods
JP2005507650A (en) Assays for novel fusion proteins and molecular binding
JP2001507792A (en) Method for detecting IgE
US20120142015A1 (en) Gfp fusion proteins and their use
JP2009153399A (en) Single molecule-format real-time bioluminescence imaging probe
CA2627022A1 (en) Detection of intracellular enzyme complex
US20090307791A1 (en) Calcium-activated chloride channel and methods of use thereof
WO2013124482A2 (en) Identification of modulators of binding properties of antibodies reactive with a member of the insulin receptor family
US20200400567A1 (en) Fusion polypeptide
Nibbs et al. Structure–function dissection of D6, an atypical scavenger receptor
AU2003212054A1 (en) Methods of assaying receptor activity and constructs useful in such methods
US7150971B2 (en) Membrane-resident steroid receptors and methods of use thereof
JP2003035714A (en) Method for detection of existence of binding capacity of specimen to protein and marker protein generated in solution using expression vector used in the same as well as method of preparing the same
Dhesi Development of methods for studying the subcelluar trafficking of insulin-regulated glucose transporters
Chang Functional Analysis of Yeast Pheromone Receptors in ER Exit, Ligand-Induced Endocytosis and Oligomerization: A Dissertation
McVey An investigation of homo and heterodimerization of the human delta opioid receptor

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry

Effective date: 20180604