EP1137776A2 - Non-endogenous, constitutively activated human g protein-coupled receptors - Google Patents

Non-endogenous, constitutively activated human g protein-coupled receptors

Info

Publication number
EP1137776A2
EP1137776A2 EP99950301A EP99950301A EP1137776A2 EP 1137776 A2 EP1137776 A2 EP 1137776A2 EP 99950301 A EP99950301 A EP 99950301A EP 99950301 A EP99950301 A EP 99950301A EP 1137776 A2 EP1137776 A2 EP 1137776A2
Authority
EP
European Patent Office
Prior art keywords
endogenous
protein
human
cdna
plasmid
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.)
Withdrawn
Application number
EP99950301A
Other languages
German (de)
French (fr)
Inventor
Dominic P. Behan
Karin Lehmann-Bruinsma
Derek T. Chalmers
Ruoping Chen
Huong T. Dang
Martin Gore
Chen W. Liaw
I-Lin Lin
Kevin Lowitz
Carol White
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.)
Arena Pharmaceuticals Inc
Original Assignee
Arena Pharmaceuticals Inc
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
Priority claimed from US09/170,496 external-priority patent/US6555339B1/en
Application filed by Arena Pharmaceuticals Inc filed Critical Arena Pharmaceuticals Inc
Priority to EP05009877A priority Critical patent/EP1676861A3/en
Priority to EP10007972A priority patent/EP2264068A1/en
Priority claimed from PCT/US1999/024065 external-priority patent/WO2000022131A2/en
Publication of EP1137776A2 publication Critical patent/EP1137776A2/en
Withdrawn legal-status Critical Current

Links

Classifications

    • 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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/72Receptors; Cell surface antigens; Cell surface determinants for hormones
    • C07K14/723G protein coupled receptor, e.g. TSHR-thyrotropin-receptor, LH/hCG receptor, FSH receptor

Definitions

  • receptors and more particularly to human G protein-coupled receptors, and specifically to GPCRs that have been altered to establish or enhance constitutive activity of the receptor.
  • the altered GPCRs are used for the direct identification of candidate compounds
  • receptor agonists as receptor agonists, inverse agonists or partial agonists having potential applicability as receptor agonists, inverse agonists or partial agonists having potential applicability as
  • GPCR G protein-coupled receptor
  • GPCRs GPCRs
  • GPCRs for which the endogenous ligand has been identified are referred to as
  • GPCRs represent an important area for the development
  • each span is identified by number, i. e., transmembrane- 1 (TM-
  • transmebrane-2 (TM-2), etc.).
  • the transmembrane helices are joined by strands of amino acids
  • transmembrane-2 and transmembrane-3 acids between transmembrane-2 and transmembrane-3 , transmembrane-4 and transmembrane-
  • extracellular regions 1 , 2 and 3 (EC- 1 , EC-2 and EC-
  • transmembrane helices are also joined by strands of amino acids
  • activation of the receptor
  • Gq, Gs, Gi, Gz and Go are G proteins
  • signal transduction begins a signaling cascade process (referred to as “signal transduction"). Under normal
  • GPCRs exist in the cell membrane in
  • a receptor in an inactive state is unable to link to the intracellular signaling transduction
  • a receptor may be stabilized in an active state by an endogenous ligand or a compound such as a drug.
  • Figure 1 is a representation of 8XCRE-Luc reporter plasmid (see, Example
  • Figures 2A and 2B are graphic representations of the results of ATP and ADP
  • Figure 3 is a graphic representation of the comparative signaling results of
  • AGONISTS shall mean materials (e.g., ligands, candidate compounds) that activate the intracellular response when they bind to the receptor, or enhance GTP binding to
  • PARTIAL AGONISTS shall mean materials (e.g., ligands, candidate compounds)
  • ANTAGONIST shall mean materials (e.g., ligands, candidate compounds) that
  • the intracellular response initiated by the active form of the receptor can thereby inhibit
  • CANDIDATE COMPOUND shall mean a molecule (for example, and not limitation, a chemical compound) that is amenable to a screening technique.
  • a chemical compound for example, and not limitation, a chemical compound that is amenable to a screening technique.
  • COMPOSITION means a material comprising at least one component
  • composition is an example of a composition.
  • COMPOUND EFFICACY shall mean a measurement of the ability of a compound
  • CODON shall mean a grouping of three nucleotides (or equivalents to nucleotides)
  • nucleoside adenosine (A), guanosine (G), cytidine (C), uridine
  • CONSTITUTIVELY ACTIVATED RECEPTOR shall mean a receptor subject to
  • a constitutive receptor activation can be endogenous or non-
  • CONSTITUTIVE RECEPTOR ACTIVATION shall mean stabilization of a
  • receptor in the active state by means other than binding of the receptor with its endogenous ligand or a chemical equivalent thereof.
  • CONTACT or CONTACTING shall mean bringing at least two moieties together
  • constitutively activated receptor preferably a constitutively activated o ⁇ han receptor
  • ENDOGENOUS shall mean a material that a mammal naturally produces.
  • NON-ENDOGENOUS in this context shall mean
  • a mammal for example, and not limitation, a human
  • a receptor which is not constitutively active in its
  • non-endogenous, constitutively activated receptor referred to herein as a “non-endogenous, constitutively activated receptor.” Both terms can
  • the endogenous or non-endogenous receptor may be in reference to
  • a mammal has been manipulated to include a non-endogenous constitutively activated
  • PROTEIN in the context of the invention disclosed herein, each mean a non-endogenous
  • o ⁇ han GPCR For example, and not limitation, in an endogenous state, if the G protein
  • Gs ⁇ is the predominate G protein that couples with the GPCR, a GPCR Fusion Protein
  • Gs ⁇ fused to Gs ⁇ ; in some circumstances, as will be set forth below, a non-predominant G protein
  • the G protein can be fused directly to the c-terminus of the
  • GPCR constitutively active GPCR or there may be spacers between the two.
  • HOST CELL shall mean a cell capable of having a Plasmid and/or Vector
  • Plasmid In the case of a prokaryotic Host Cell, a Plasmid is typically replicated
  • Plasmid as a autonomous molecule as the Host Cell replicates (generally, the Plasmid is thereafter
  • Plasmid is integrated into the cellular DNA of the Host Cell such that when the eukaryotic
  • the Host Cell is eukaryotic, more preferably, mammalian, and most
  • INVERSE AGONISTS shall mean materials (e.g., ligand, candidate compound)
  • the baseline intracellular response is inhibited in the presence of the inverse agonist by at least
  • KNOWN RECEPTOR shall mean an endogenous receptor for which the endogenous
  • LIGAND shall mean an endogenous, naturally occurring molecule specific for an
  • a subsequent mutated form of a human receptor is considered to be equivalent to a first mutation of the human receptor if (a) the level of constitutive activation of the
  • the of the receptor is at least about 80%, more preferably at least about 90% and most preferably
  • for achieving constitutive activation includes a single amino acid and/or codon change
  • homology should be at least 98%.
  • NON-ORPHAN RECEPTOR shall mean an endogenous naturally occurring
  • ligand to a receptor activates an intracellular signaling pathway.
  • ORPHAN RECEPTOR shall mean an endogenous receptor for which the
  • PHARMACEUTICAL COMPOSITION shall mean a composition comprising at
  • composition is amenable to investigation for a
  • PLASMID shall mean the combination of a Vector and cDNA.
  • a Plasmid shall mean the combination of a Vector and cDNA.
  • VECTOR in reference to cDNA shall mean a circular DNA capable of inco ⁇ orating
  • At least one cDNA and capable of inco ⁇ oration into a Host Cell at least one cDNA and capable of inco ⁇ oration into a Host Cell.
  • any search for therapeutic compounds should start by screening compounds against
  • GPCRs GPCR (Accession No.) hARE-3 AL033379 1,260 bp 52.3% LPA-R U92642 hARE-4 AC006087 1,119 bp 36% P2Y5 AF000546 hARE-5 AC006255 1,104 bp 32% Oryzias D43633 latipes hGPR27 AA775870 1,128 bp hARE-1 AI090920 999 bp 43% D13626 KIAA0001 hARE-2 AA359504 1,122 bp 53% GPR27 hPPRl H67224 1,053 bp 39% EBI1 L31581 hG2A AA754702 1,113 bp 31% GPR4 L36148 hRTJP3 AL035423 1,005 bp 30% 2133653
  • Receptor homology is useful in terms of gaining an appreciation of a role of the
  • human GPCRs disclosed herein is expressed and/or over-expressed, it is possible to determine
  • human GPCR disclosed herein is based upon the distance from the proline residue at which
  • non-endogenous, constitutively activated GPCR can be identified by the methodologies of this
  • compositions a search for diseases and disorders associated with the GPCR
  • Tissue scans can be used to identify an endogenous ligand to the specific GPCR.
  • tissue scans provide a broad range of healthy and diseased tissues. Such tissue scans provide
  • a preferred first step in associating a specific receptor with a disease and/or disorder See, for example, co-pending application (docket number ARE-0050) for exemplary dot-blot and RT-
  • the DNA sequence of the human GPCR is used to make a probe for (a)
  • compared to a normal tissue can be preferably utilized to identify a correlation with a
  • treatment regimen including but not limited to, a disease associated with that disease.
  • Receptors can equally well be localized to regions of organs by this technique. Based on
  • a G protein receptor When a G protein receptor becomes constitutively active, it binds to a G protein (e.g.,
  • Gq Gs, Gi, Gz, Go
  • this assay system is for initial screening of candidate compounds because the system is generically applicable to all G protein-coupled receptors regardless of the particular
  • G protein that interacts with the intracellular domain of the receptor.
  • receptor assay (/. e., an assay to select compounds that are agonists, partial agonists, or inverse
  • a compound identified by the "generic" assay may not bind to the
  • the receptor may instead merely "uncouple" the G protein from the intracellular domain.
  • Gs stimulates the enzyme adenylyl cyclase. Gi (and Gz and Go), on the other hand,
  • Adenylyl cyclase catalyzes the conversion of ATP to cAMP; thus,
  • constitutively activated GPCRs that couple the Gs protein are associated with increased
  • a candidate compound is, e.g., an inverse agonist to the receptor
  • Cyclic AMP drives gene
  • cAMP cAMP -responsive DNA binding protein or transcription factor
  • Reporter systems can be constructed
  • Gs-linked receptor e.g., ⁇ -galactosidase or luciferase.
  • the reporter protein such as ⁇ -galactosidase or luciferase can then be detected using standard
  • Gq and Go are associated with activation of the enzyme phospholipase C, which in
  • DAG diacycloglycerol
  • IP 3 inistol 1,4,5-triphoisphate
  • a candidate compound is, e.g., an inverse agonist to a Gq- or Go-
  • phospholipase C causes activation of genes containing API elements; thus, activated Gq-
  • a preferred approach is the use of a GPCR Fusion Protein.
  • Coupling of the G protein to the GPCR provides a signaling pathway that can be assessed.
  • the GPCR Fusion Protein is intended to enhance the efficacy of G protein coupling with the non-endogenous GPCR.
  • the GPCR Fusion Protein is preferred for screening with
  • expression vectors and systems offer a variety of approaches that can fit the particular needs
  • endogenous GPCR sequence and the G protein sequence both be in-frame (preferably,
  • sequence for the endogenous GPCR is upstream of the G protein sequence
  • the G protein can also be expressed.
  • the GPCR can be linked directly to the G
  • the G protein is not used to, effectively, upon expression, become a spacer.
  • the G protein is not used to, effectively, upon expression, become a spacer.
  • a construct comprising the sequence of the G protein (i.e., a
  • G protein in an effort to establish a viable cyclase-based assay.
  • a Gz a Gz
  • a GPCR Fusion Protein can be established that utilizes a Gs
  • Candidate compounds selected for further development can be formulated into
  • human GPCRs can also be utilized in research settings. For example, in vitro and in vivo
  • cassettes from one sequence to another e.g. from rat receptor to human receptor or from
  • human receptor A to human receptor B is generally predicated upon sequence alignment
  • GPCRs (Base Pairs) hGPCR27 Mouse AA775870 1,125 bp 17 18 GPCR27 hARE-1 TDAG 1689643 999 bp 19 20 AI090920 hARE-2 GPCR27 68530 1,122 bp 21 22 AA359504 hPPRl Bovine 238667 1 ,053 bp 23 24 PPR1 H67224 hG2A Mouse See Example 2(a), 1,113 bp 25 26 1 179426 below hCHN3 N.A. EST 36581 1.113 bp 27 28 (full length) hCHN4 TDAG 1184934 1 ,077 bp 29 30 AA804531 hCHN6 N.A.
  • Mouse EST clone 1179426 was used to obtain a human genomic clone containing all but three amino acid G2A coding sequences. The 5 'of this coding sequence was obtained by
  • the disclosed human G2A was amplified by PCR using the G2A cDNA specific
  • PCR fragment was purified from agarose gel, digested with Hind III and Xba I and cloned into
  • the 5' primer sequence utilized was as follows:
  • the cycle condition was 30 cycles of 94°C for 1 min, 65°C for lmin
  • RUP4 The full length RUP4 was cloned by RT-PCR with human brain cDNA (Clontech) as
  • PCR was performed using TaqPlus PrecisionTM polymerase (Stratagene; manufacturing
  • PCR products were separated on a 1% agarose gel and a 500 bp PCR fragment
  • the 3' RACE product contained a poly(A) tail and a completed open reading frame ending
  • the 5' RACE product contained an incomplete 5' end; i.e., the ATG
  • oligo 3 Based on the new 5' sequence, oligo 3 and the following primer:
  • 5'-GCAATGCAGGTCATAGTGAGC -3' (SEQ.ID.NO.: 52; oligo 5) were used for the second round of 5 ' race PCR and the PCR products were analyzed as above.
  • 5'-GTGATGAGCAGGTCACTGAGCGCCAAG-3' (SEQ.ID.NO.: 54: oligo7).
  • the sequence of the 5' RACE PCR products revealed the presence of the initiation codon ATG, and further round of 5' race PCR did not generate any more 5' sequence.
  • the full length RUP5 was cloned by RT-PCR using a sense primer upstream from
  • step 2 through step 4 repeated 30 times: 94°C for 30 sec; 94° for 15 sec; 69° for 40 sec;
  • Cycles 2 through 4 were repeated 30 times.
  • step 2 to step 4 repeated 30 times: 94 °C for 2 minutes; 94 °C for 15 seconds; 60°C
  • ATI human angiotensin II type 1 receptor
  • each primer 0.25 ⁇ M of each primer, and 0.2 mM of each 4 nucleotides.
  • the cycle condition was 30 cycles of 94°C for 1 min, 55°C for lmin and 72 °C for 1.5 min.
  • the 5' PCR primer contains a Hindlll site with the sequence: 5'-CCCAAGCTTCCCCAGGTGTATTTGAT-3' (SEQ.ID.NO.: 63) and the 3' primer contains a BamHl site with the following sequence:
  • Nucleic acid SEQ.ID.NO.: 65
  • amino acid SEQ.ID.NO.: 66
  • PCR was performed by combining two PCR fragments, using
  • the cycle condition for each PCR reaction was 30 cycles of 94 °C for 1 min, 62 °C for lmin
  • the first fragment was amplified with the 5' PCR primer that contained an end site
  • the second PCR fragment was amplified with a 5' primer having the following sequence: 5'-GTCCGCGTCCTGCTGGTGGTGGTTCTGGCATTTATAATT-3' (SEQ.ID.NO.: 69) and a 3' primer that contained a BamHl site and having the following sequence:
  • the two fragments were used as templates to amplify GPR38, using SEQ.ID.NO.: 67 and
  • SEQ.ID.NO.: 70 as primers (using the above-noted cycle conditions).
  • the resulting 1.44kb PCR fragment was digested with BamHl and cloned into Blunt-BamHI site of pCMV
  • PCR was performed using human genomic cDNA as template and
  • the 5' PCR contained an EcoRI site with the sequence:
  • the 1.0 kb PCR fragment was digest with EcoRI and BamHl and cloned into EcoRI -BamHl
  • PCR was performed using human stomach cDNA as template and
  • the 5' PCR contained a Hindlll site with the sequence:
  • the resulting 1.44 kb PCR fragment was digest with Hindlll and EcoRI and cloned into Hindlll-EcoRI site of pCMV expression vector. Nucleic acid (SEQ.ID.NO.: 77) and amino
  • PCR was performed using genomic DNA as template and rTth
  • each primer and 0.2 mM of each 4 nucleotides.
  • the cycle condition was 30 cycles of 94°C
  • the 5' PCR primer contained a
  • the resulting 1.1 kb PCR fragment was digested with Hindlll and BamHl and cloned into
  • each primer and 0.2 mM of each 4 nucleotides.
  • the cycle condition was 30 cycles of 94°C
  • the 5' PCR primer contained a Hindlll site
  • the resulting 1.9 kb PCR fragment was digested with Hindlll and BamHl and cloned into
  • Hindlll-BamHI site of pCMV expression vector. H9 contained three potential polymo ⁇ hisms
  • TM6 region of the GPCR, near the TM6/IC3 interface is mutated, most preferably to a
  • Two mutagenesis primers are utilized, most preferably a lysine
  • mutagenesis oligonucleotide that creates the lysine mutation and a selection marker
  • the two mutagenesis primers were used, a lysine mutagenesis oligonucleotide
  • the 5' PCR sense primer used had the following sequence: 5'-CCCAAGCTTCCCCAGGTGTATTTGAT-3' (SEQ.ID.NO.: 95) and the antisense primer had the following sequence: 5'-CCTGCAGGCGAAACTGACTCTGGCTGAAG-3' (SEQ.ID.NO.: 96).
  • the resulting 400 bp PCR fragment was digested with Hindlll site and subcloned into
  • Nl 11 A construct The cycle condition was 25 cycles of 94°C for 1 min, 60°C for lmin
  • 3' untranslated region was generated by using the following sequence: 5'-AGATCTTAAGAAGATAATTATGGCAATTGTGCT-3' (SEQ.ID.NO.: 102) as sense primer and SEQ.ID.NO.: 64 as antisense primer.
  • the PCR condition was 30
  • Fragment A (720 bp) was digested with Hindlll and
  • Fragment B was digested with BamHl and subcloned into pCMV
  • Fragment C was inserted in front of Fragment B through EcoRI and Aflll site.
  • PCR sense primer had the following sequence:
  • the 3 ' PCR sense primer utilized had the following sequence:
  • the PCR condition was the
  • the first PCR fragment (lkb) was amplified by using SEQ.ID.NO.: 75 and an
  • antisense primer comprising a V322K mutation
  • the second PCR fragment (0.44kb) was amplified by using a sense primer comprising the
  • V322K mutation 5'-AGAAGCGCGTGAAGCGCATGCTGCTGGTGATCGTT-3' (SEQ.ID.NO: 114) and SEQ.ID.NO.:
  • Preparation of non-endogenous human GPCRs can also be accomplished by using
  • Endogenous GPCR is preferably used as a template and two mutagenesis
  • primers utilized utilized, as well as, most preferably, a lysine mutagenesis oligonucleotide and a
  • selection marker oligonucleotide (included in kit).
  • codon mutation For convenience, the codon mutation
  • yeast cells upon practicalities, i.e., utilization of, e.g., yeast cells for the expression of a GPCR, while
  • yeast does not) include the receptor-coupling, genetic-mechanism and secretary
  • mammalian cells while of potential use, are not as preferred as that obtained from mammalian
  • COS-7, 293 and 293T cells are particularly preferred, although
  • reaction tubes were prepared (the proportions to follow for each tube are per plate): tube A
  • DNA e.g., pCMV vector; pCMV vector with receptor
  • tube B was prepared by mixing 120 ⁇ l lipofectamine (Gibco BRL) in 1.2ml serum free DMEM. Tubes
  • a and B were admixed by inversions (several times), followed by incubation at room
  • the admixture is referred to as the "transfection mixture”.
  • the receptor couples to a G protein and stimulates the
  • the assay utilizes the ability of G protein coupled receptors to stimulate [ 35 S]GTP ⁇ S
  • the assay can, therefore, be used in
  • the assay is generic and has application
  • the [ 35 S]GTP ⁇ S assay can be incubated in 20 mM HEPES and between 1 and about
  • ⁇ g membrane protein e.g, COS-7 cells expressing the receptor; this amount can be adjusted
  • Flash platesTM and WallacTM scintistrips may be utilized
  • the assay can be utilized for known GPCRs to simultaneously monitor tritiated ligand binding
  • This assay may also be used to detect other types of membrane
  • the assay may be used to determine whether receptor activation events resulting in receptor activation.
  • the assay may be used to determine whether receptor activation events resulting in receptor activation.
  • the assay may be used to determine whether receptor activation events resulting in receptor activation.
  • the scintistrip label comes into proximity with the radiolabeled ligand resulting
  • Flash Plate wells contain a scintillant coating which also contains a specific antibody-
  • the cAMP generated in the wells was quantitated by a direct
  • Transfected cells are harvested approximately three days after transfection.
  • Membranes were prepared by homogenization of suspended cells in buffer containing 20mM
  • HEPES, pH 7.4 and lOmM MgCl 2 Homogenization is performed on ice using a Brinkman
  • Assay Buffer is prepared fresh for screening and contained
  • Assay Buffer can be stored on ice
  • the assay is initiated by addition of 50ul of assay buffer followed by addition
  • a method to detect Gs stimulation depends on the known property of the transcription
  • CREB CREB factor CREB
  • a PathDetectTM CREB trans- Reporting System (Stratagene, Catalogue # 219010) can utilized to assay for Gs coupled
  • Gal4 recognition sequences 40 ng pFA2-CREB (Gal4-CREB fusion protein containing the
  • Gal4 DNA-binding domain 80 ng pCMV -receptor expression plasmid (comprising the
  • CMV-SEAP secreted alkaline phosphatase expression plasmid; alkaline
  • phosphatase activity is measured in the media of transfected cells to control for variations in
  • a method to detect Gq stimulation depends on the known property of Gq-dependent
  • a PathdetectTM AP-1 cis-Reporting System (Stratagene, Catalogue # 219073) can be utilized
  • the components of the calcium phosphate precipitate were 410 ng pAP 1 -Luc. 80 ng pCMV-
  • 293 and 293T cells are plated-out on 96 well plates at a density of 2 x 10 4 cells per well and were transfected using Lipofectamine Reagent (BRL) the following day according
  • pCMV comprising endogenous receptor or non-endogenous receptor or pCMV alone
  • reporter plasmid was prepared as follows: vector SRIF- ⁇ -gal was obtained by cloning the rat
  • somatostatin promoter (-71/+51) at BglV-Hindlll site in the p ⁇ gal-Basic Vector (Clontech).
  • AdpCF126CCRE8 see, 1 Human Gene Therapy 1883 (1996)
  • 8xCRE-Luc reporter plasmid was generated by replacing the beta-galactosidase gene in the
  • DNA/lipid mixture was diluted with 400 ⁇ l of DMEM and lOO ⁇ l of the diluted mixture was
  • a PathdetectTM SRF-Luc-Reporting System (Stratagene) can be utilized to assay
  • Gq coupled activity in, e.g., COS7 cells.
  • Cells are transfected with the plasmid
  • alkaline phosphatase activity is measured in the media of transfected cells to control for
  • cells comprising the receptors can be endogenous and/or non-endogenous.
  • the cells are then incubated for 3-4 hrs at 37°C/5%CO 2 and then the
  • transfection media is removed and replaced with 1 ml/well of regular growth media.
  • the cells are then incubated for 30 min at 37 °C.
  • the cells are then incubated for 30 min at 37 °C.
  • the lysate is then transferred into 1.5 ml eppendorf tubes and 1 ml of
  • the transfected cells were grown in media containing serum for an assay
  • the serum-free media was comprised solely
  • DME Dulbecco's Modified Eagle's
  • the media with serum contained the following: 10%
  • a 96-well Adenylyl Cyclase Activation FlashplateTM was used (NEN: #SMP004A).
  • CMV or TDAG8 were harvested 24 (assay detection in serum media) or 48 hours post-

Abstract

The invention disclosed in this patent document relates to transmembrane receptors, more particularly to a human G protein-coupled receptor for which the endogenous ligand is unknown ('orphan GPCR receptors'), and most particularly to mutated (non-endogenous) versions of the human GPCRs for evidence of constitutive activity.

Description

NON-ENDOGENOUS, CONSTITUTIVELY ACTIVATED HUMAN G PROTEIN-COUPLED RECEPTORS
This patent application is a continuation-in-part of, and claims priority from, U.S.
Serial Number 09/170,496, filed with the United States Patent and Trademark Office on
October 13, 1998. This application also claims the benefit of priority from the following
provisional applications, all filed via U.S. Express Mail with the United States Patent and
Trademark Office on the indicated dates: U.S. Provisional Number 60/110,060, filed
November 27, 1998; U.S. Provisional Number 60/120,416, filed February 16, 1999; U.S.
Provisional Number 60/121,852, filed February 26, 1999 claiming benefit of U.S.
Provisional Number 60/109,213, filed November 20, 1998; U.S. Provisional Number
60/123,944, filed March 12, 1999; U.S. Provisional Number 60/123,945, filed March 12,
1999; U.S. Provisional Number 60/123,948, filed March 12, 1999; U.S. Provisional
Number 60/123,951, filed March 12, 1999; U.S. Provisional Number 60/123,946, filed
March 12, 1999; U.S. Provisional Number 60/123,949, filed March 12, 1999; U.S.
Provisional Number 60/152,524, filed September 3, 1999, claiming benefit of U.S.
Provisional Number 60/151,114, filed August 27, 1999 and U.S. Provisional Number
60/108,029, filed November 12, 1998; U.S. Provisional Number 60/136,436, filed May 28,
1999; U.S. Provisional Number 60/136,439, filed May 28, 1999; U.S. Provisional Number
60/136,567, filed May 28, 1999; U.S. Provisional Number 60/137,127, filed May 28,
1999; U.S. Provisional Number 60/137,131, filed May 28, 1999; U.S. Provisional Number 60/141,448, filed June 29, 1999 claiming benefit of U.S. Provisional Number 60/136,437,
filed May 28, 1999; U.S. Provisional Number 60/156,633, filed September 29, 1999; U.S.
Provisional Number 60/156,555, filed September 29, 1999; U.S. Provisional Number
60/156,634, filed September 29, 1999;U.S. Provisional Number (Arena
Pharmaceuticals, Inc. docket number: CHNlO-l), filed September 29, 1999; U.S.
Provisional Number (Arena Pharmaceuticals, Inc. docket number: RUP6-1), filed
October 1, 1999; U.S. Provisional Number (Arena Pharmaceuticals, Inc. docket
number: RUP7-1), filed October 1, 1999; U.S. Provisional Number (Arena
Pharmaceuticals, Inc. docket number: CHN6-1), filed October 1, 1999; U.S. Provisional
Number (Arena Pharmaceuticals, Inc. docket number: RUP5-1), filed October 1, 1999;
and U.S. Provisional Number (Arena Pharmaceuticals, Inc. docket number: CHN9-1),
filed October 1, 1999. This application is also related to co-pending U.S. Serial Number
(Woodcock, Washburn, Kurtz, Makiewicz & Norris, LLP docket number AREN-
0050), filed on October 12, 1999 (via U.S. Express Mail) and U.S. Serial Number
09/364,425, filed on July 30, 1999, both incoφorated herein by reference. This
application also claims priority to U.S. Serial Number (Woodcock, Washburn,
Kurtz, Makiewicz & Norris, LLP docket number AREN-0054), filed on October 12, 1999
(via U.S. Express Mail), incorporated by reference herein in its entirety. Each of the
foregoing applications are incoφorated by reference herein in their entirety.
FIELD OF THE INVENTION
The invention disclosed in this patent document relates to transmembrane
receptors, and more particularly to human G protein-coupled receptors, and specifically to GPCRs that have been altered to establish or enhance constitutive activity of the receptor.
Preferably, the altered GPCRs are used for the direct identification of candidate compounds
as receptor agonists, inverse agonists or partial agonists having potential applicability as
therapeutic agents.
BACKGROUND OF THE INVENTION
Although a number of receptor classes exist in humans, by far the most
abundant and therapeutically relevant is represented by the G protein-coupled receptor (GPCR
or GPCRs) class. It is estimated that there are some 100,000 genes within the human genome,
and of these, approximately 2%, or 2,000 genes, are estimated to code for GPCRs. Receptors,
including GPCRs, for which the endogenous ligand has been identified are referred to as
"known" receptors, while receptors for which the endogenous ligand has not been identified
are referred to as "oφhan" receptors. GPCRs represent an important area for the development
of pharmaceutical products: from approximately 20 of the 100 known GPCRs, 60% of all
prescription pharmaceuticals have been developed.
GPCRs share a common structural motif. All these receptors have seven
sequences of between 22 to 24 hydrophobic amino acids that form seven alpha helices, each
of which spans the membrane (each span is identified by number, i. e., transmembrane- 1 (TM-
1), transmebrane-2 (TM-2), etc.). The transmembrane helices are joined by strands of amino
acids between transmembrane-2 and transmembrane-3 , transmembrane-4 and transmembrane-
5, and transmembrane-6 and transmembrane-7 on the exterior, or "extracellular" side, of the
cell membrane (these are referred to as "extracellular" regions 1 , 2 and 3 (EC- 1 , EC-2 and EC-
3), respectively). The transmembrane helices are also joined by strands of amino acids
between transmembrane- 1 and transmembrane-2, transmembrane-3 andtransmembrane-4, and transmembrane-5 and transmembrane-6 on the interior, or "intracellular" side, of the cell
membrane (these are referred to as "intracellular" regions 1, 2 and 3 (IC-1, IC-2 and IC-3),
respectively). The "carboxy" ("C") terminus of the receptor lies in the intracellular space
within the cell, and the "amino" ("N") terminus of the receptor lies in the extracellular space
outside of the cell.
Generally, when an endogenous ligand binds with the receptor (often referred
to as "activation" of the receptor), there is a change in the conformation of the intracellular
region that allows for coupling between the intracellular region and an intracellular "G-
protein." It has been reported that GPCRs are "promiscuous" with respect to G proteins, i.e.,
that a GPCR can interact with more than one G protein. See, Kenakin, T., 43 Life Sciences
1095 (1988). Although other G proteins exist, currently, Gq, Gs, Gi, Gz and Go are G proteins
that have been identified. Endogenous ligand-activated GPCR coupling with the G-protein
begins a signaling cascade process (referred to as "signal transduction"). Under normal
conditions, signal transduction ultimately results in cellular activation or cellular inhibition.
It is thought that the IC-3 loop as well as the carboxy terminus of the receptor interact with
the G protein.
Under physiological conditions, GPCRs exist in the cell membrane in
equilibrium between two different conformations: an "inactive" state and an "active" state.
A receptor in an inactive state is unable to link to the intracellular signaling transduction
pathway to produce a biological response. Changing the receptor conformation to the active
state allows linkage to the transduction pathway (via the G-protein) and produces a biological
response.
A receptor may be stabilized in an active state by an endogenous ligand or a compound such as a drug. Recent discoveries, including but not exclusively limited to
modifications to the amino acid sequence of the receptor, provide means other than
endogenous ligands or drugs to promote and stabilize the receptor in the active state
conformation. These means effectively stabilize the receptor in an active state by
simulating the effect of an endogenous ligand binding to the receptor. Stabilization by
such ligand-independent means is termed "constitutive receptor activation."
SUMMARY OF THE INVENTION
Disclosed herein are non-endogenous versions of endogenous, human GPCRs and
uses thereof.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 is a representation of 8XCRE-Luc reporter plasmid (see, Example
4(c)3.)
Figures 2A and 2B are graphic representations of the results of ATP and ADP
binding to endogenous TDAG8 (2A) and comparisons in serum and serum free media (2B).
Figure 3 is a graphic representation of the comparative signaling results of
CMV versus the GPCR Fusion Protein H9(F236K):Gsα.
DETAILED DESCRIPTION
The scientific literature that has evolved around receptors has adopted a
number of terms to refer to ligands having various effects on receptors. For clarity and
consistency, the following definitions will be used throughout this patent document. To the
extent that these definitions conflict with other definitions for these terms, the following
definitions shall control:
AGONISTS shall mean materials (e.g., ligands, candidate compounds) that activate the intracellular response when they bind to the receptor, or enhance GTP binding to
membranes.
AMINO ACID ABBREVIATIONS used herein are set out in Table A:
TABLE A
ALANINE ALA A
ARGININE ARG R
ASPARAGINE ASN N
ASPARTIC ACID ASP D
CYSTEINE CYS C GLUTAMIC ACID GLU E
GLUTAMINE GLN Q
GLYCINE GLY G
HISTIDINE HIS H
ISOLEUCINE ILE I LEUCINE LEU L
LYSINE LYS K
METHIONINE MET M
PHENYLALANINE PHE F
PROLINE PRO P SERINE SER S
THREONINE THR T
TRYPTOPHAN TRP W
TYROSINE TYR Y
VALINE VAL V
PARTIAL AGONISTS shall mean materials (e.g., ligands, candidate compounds)
that activate the intracellular response when they bind to the receptor to a lesser degree/extent
than do agonists, or enhance GTP binding to membranes to a lesser degree/extent than do
agonists.
ANTAGONIST shall mean materials (e.g., ligands, candidate compounds) that
competitively bind to the receptor at the same site as the agonists but which do not activate
the intracellular response initiated by the active form of the receptor, and can thereby inhibit
the intracellular responses by agonists or partial agonists. ANTAGONISTS do not diminish
the baseline intracellular response in the absence of an agonist or partial agonist.
CANDIDATE COMPOUND shall mean a molecule (for example, and not limitation, a chemical compound) that is amenable to a screening technique. Preferably, the phrase
"candidate compound" does not include compounds which were publicly known to be
compounds selected from the group consisting of inverse agonist, agonist or antagonist to a
receptor, as previously determined by an indirect identification process ("indirectly identified
compound"); more preferably, not including an indirectly identified compound which has
previously been determined to have therapeutic efficacy in at least one mammal; and, most
preferably, not including an indirectly identified compound which has previously been
determined to have therapeutic utility in humans.
COMPOSITION means a material comprising at least one component; a
"pharmaceutical composition" is an example of a composition.
COMPOUND EFFICACY shall mean a measurement of the ability of a compound
to inhibit or stimulate receptor functionality, as opposed to receptor binding affinity.
Exemplary means of detecting compound efficacy are disclosed in the Example section of this
patent document.
CODON shall mean a grouping of three nucleotides (or equivalents to nucleotides)
which generally comprise a nucleoside (adenosine (A), guanosine (G), cytidine (C), uridine
(U) and thymidine (T)) coupled to a phosphate group and which, when translated, encodes an
amino acid.
CONSTITUTIVELY ACTIVATED RECEPTOR shall mean a receptor subject to
constitutive receptor activation. A constitutively activated receptor can be endogenous or non-
endogenous.
CONSTITUTIVE RECEPTOR ACTIVATION shall mean stabilization of a
receptor in the active state by means other than binding of the receptor with its endogenous ligand or a chemical equivalent thereof.
CONTACT or CONTACTING shall mean bringing at least two moieties together,
whether in an in vitro system or an in vivo system.
DIRECTLY IDENTIFYING or DIRECTLY IDENTIFIED, in relationship to the
phrase "candidate compound", shall mean the screening of a candidate compound against a
constitutively activated receptor, preferably a constitutively activated oφhan receptor, and
most preferably against a constitutively activated G protein-coupled cell surface oφhan
receptor, and assessing the compound efficacy of such compound. This phrase is, under no
circumstances, to be inteφreted or understood to be encompassed by or to encompass the
phrase "indirectly identifying" or "indirectly identified."
ENDOGENOUS shall mean a material that a mammal naturally produces.
ENDOGENOUS in reference to, for example and not limitation, the term "receptor," shall
mean that which is naturally produced by a mammal (for example, and not limitation, a
human) or a virus. By contrast, the term NON-ENDOGENOUS in this context shall mean
that which is not naturally produced by a mammal (for example, and not limitation, a human)
or a virus. For example, and not limitation, a receptor which is not constitutively active in its
endogenous form, but when manipulated becomes constitutively active, is most preferably
referred to herein as a "non-endogenous, constitutively activated receptor." Both terms can
be utilized to describe both "in vivo" and "in vitro" systems. For example, and not limitation,
in a screening approach, the endogenous or non-endogenous receptor may be in reference to
an in vitro screening system. As a further example and not limitation, where the genome of
a mammal has been manipulated to include a non-endogenous constitutively activated
receptor, screening of a candidate compound by means of an in vivo system is viable. G PROTEIN COUPLED RECEPTOR FUSION PROTEIN and GPCR FUSION
PROTEIN, in the context of the invention disclosed herein, each mean a non-endogenous
protein comprising an endogenous, constitutively activate GPCR or a non-endogenous,
constitutively activated GPCR fused to at least one G protein, most preferably the alpha (α)
subunit of such G protein (this being the subunit that binds GTP), with the G protein
preferably being of the same type as the G protein that naturally couples with endogenous
oφhan GPCR. For example, and not limitation, in an endogenous state, if the G protein
"Gsα" is the predominate G protein that couples with the GPCR, a GPCR Fusion Protein
based upon the specific GPCR would be a non-endogenous protein comprising the GPCR
fused to Gsα; in some circumstances, as will be set forth below, a non-predominant G protein
can be fused to the GPCR. The G protein can be fused directly to the c-terminus of the
constitutively active GPCR or there may be spacers between the two.
HOST CELL shall mean a cell capable of having a Plasmid and/or Vector
incoφorated therein. In the case of a prokaryotic Host Cell, a Plasmid is typically replicated
as a autonomous molecule as the Host Cell replicates (generally, the Plasmid is thereafter
isolated for introduction into a eukaryotic Host Cell); in the case of a eukaryotic Host Cell,
a Plasmid is integrated into the cellular DNA of the Host Cell such that when the eukaryotic
Host Cell replicates, the Plasmid replicates. Preferably, for the puφoses of the invention
disclosed herein, the Host Cell is eukaryotic, more preferably, mammalian, and most
preferably selected from the group consisting of 293, 293T and COS-7 cells.
INDIRECTLY IDENTIFYING or INDIRECTLY IDENTIFIED means the
traditional approach to the drug discovery process involving identification of an endogenous
ligand specific for an endogenous receptor, screening of candidate compounds against the receptor for determination of those which interfere and/or compete with the ligand-receptor
interaction, and assessing the efficacy of the compound for affecting at least one second
messenger pathway associated with the activated receptor.
INHIBIT or INHIBITING, in relationship to the term "response" shall mean that a
response is decreased or prevented in the presence of a compound as opposed to in the
absence of the compound.
INVERSE AGONISTS shall mean materials (e.g., ligand, candidate compound)
which bind to either the endogenous form of the receptor or to the constitutively activated
form of the receptor, and which inhibit the baseline intracellular response initiated by the
active form of the receptor below the normal base level of activity which is observed in the
absence of agonists or partial agonists, or decrease GTP binding to membranes. Preferably,
the baseline intracellular response is inhibited in the presence of the inverse agonist by at least
30%, more preferably by at least 50%, and most preferably by at least 75%, as compared with
the baseline response in the absence of the inverse agonist.
KNOWN RECEPTOR shall mean an endogenous receptor for which the endogenous
ligand specific for that receptor has been identified.
LIGAND shall mean an endogenous, naturally occurring molecule specific for an
endogenous, naturally occurring receptor.
MUTANT or MUTATION in reference to an endogenous receptor's nucleic acid
and/or amino acid sequence shall mean a specified change or changes to such endogenous
sequences such that a mutated form of an endogenous, non-constitutively activated receptor
evidences constitutive activation of the receptor. In terms of equivalents to specific
sequences, a subsequent mutated form of a human receptor is considered to be equivalent to a first mutation of the human receptor if (a) the level of constitutive activation of the
subsequent mutated form of a human receptor is substantially the same as that evidenced by
the first mutation of the receptor; and (b) the percent sequence (amino acid and/or nucleic
acid) homology between the subsequent mutated form of the receptor and the first mutation
of the receptor is at least about 80%, more preferably at least about 90% and most preferably
at least 95%. Ideally, and owing to the fact that the most preferred cassettes disclosed herein
for achieving constitutive activation includes a single amino acid and/or codon change
between the endogenous and the non-endogenous forms of the GPCR, the percent sequence
homology should be at least 98%.
NON-ORPHAN RECEPTOR shall mean an endogenous naturally occurring
molecule specific for an endogenous naturally occurring ligand wherein the binding of a
ligand to a receptor activates an intracellular signaling pathway.
ORPHAN RECEPTOR shall mean an endogenous receptor for which the
endogenous ligand specific for that receptor has not been identified or is not known.
PHARMACEUTICAL COMPOSITION shall mean a composition comprising at
least one active ingredient, whereby the composition is amenable to investigation for a
specified, efficacious outcome in a mammal (for example, and not limitation, a human). Those
of ordinary skill in the art will understand and appreciate the techniques appropriate for
determining whether an active ingredient has a desired efficacious outcome based upon the
needs of the artisan.
PLASMID shall mean the combination of a Vector and cDNA. Generally, a Plasmid
is introduced into a Host Cell for the puφoses of replication and/or expression of the cDNA
as a protein. STIMULATE or STIMULATING, in relationship to the term "response" shall mean
that a response is increased in the presence of a compound as opposed to in the absence of the
compound.
VECTOR in reference to cDNA shall mean a circular DNA capable of incoφorating
at least one cDNA and capable of incoφoration into a Host Cell.
The order of the following sections is set forth for presentational efficiency and is not
intended, nor should be construed, as a limitation on the disclosure or the claims to follow.
A. Introduction
The traditional study of receptors has always proceeded from the a priori assumption
(historically based) that the endogenous ligand must first be identified before discovery could
proceed to find antagonists and other molecules that could affect the receptor. Even in cases
where an antagonist might have been known first, the search immediately extended to looking
for the endogenous ligand. This mode of thinking has persisted in receptor research even after
the discovery of constitutively activated receptors. What has not been heretofore recognized
is that it is the active state of the receptor that is most useful for discovering agonists, partial
agonists, and inverse agonists of the receptor. For those diseases which result from an overly
active receptor or an under-active receptor, what is desired in a therapeutic drug is a
compound which acts to diminish the active state of a receptor or enhance the activity of the
receptor, respectively, not necessarily a drug which is an antagonist to the endogenous ligand.
This is because a compound that reduces or enhances the activity of the active receptor state
need not bind at the same site as the endogenous ligand. Thus, as taught by a method of this
invention, any search for therapeutic compounds should start by screening compounds against
the ligand-independent active state. B. Identification of Human GPCRs
The efforts of the Human Genome project has led to the identification of a plethora of
information regarding nucleic acid sequences located within the human genome; it has been
the case in this endeavor that genetic sequence information has been made available without
an understanding or recognition as to whether or not any particular genomic sequence does
or may contain open-reading frame information that translate human proteins. Several
methods of identifying nucleic acid sequences within the human genome are within the
purview of those having ordinary skill in the art. For example, and not limitation, a variety
of human GPCRs, disclosed herein, were discovered by reviewing the GenBank™ database,
while other GPCRs were discovered by utilizing a nucleic acid sequence of a GPCR,
previously sequenced, to conduct a BLAST™ search of the EST database. Table B, below,
lists several endogenous GPCRs that we have discovered, along with a GPCR's respective
homologous receptor.
TABLE B
Disclosed Accession Open Reading Per Cent Reference To
Human Number Frame Homology Homologous
Orphan Identified (Base Pairs) To Designated GPCR
GPCRs GPCR (Accession No.) hARE-3 AL033379 1,260 bp 52.3% LPA-R U92642 hARE-4 AC006087 1,119 bp 36% P2Y5 AF000546 hARE-5 AC006255 1,104 bp 32% Oryzias D43633 latipes hGPR27 AA775870 1,128 bp hARE-1 AI090920 999 bp 43% D13626 KIAA0001 hARE-2 AA359504 1,122 bp 53% GPR27 hPPRl H67224 1,053 bp 39% EBI1 L31581 hG2A AA754702 1,113 bp 31% GPR4 L36148 hRTJP3 AL035423 1,005 bp 30% 2133653
Drosophila melanogaster hRUP4 AI307658 1,296 bp 32% pNPGPR NP 004876 28% and 29 % AAC41276 Zebra fish Ya and and Yb, AAB94616 respectively hRUP5 AC005849 1,413 bp 25% DEZ Q99788 23% FMLPR P21462 hRUP6 AC005871 1,245 bp 48% GPR66 NP_006047 hRUP7 AC007922 1,173 bp 43% H3R AF 140538 hCHN3 EST 36581 1,113 bp 53% GPR27 hCHN4 AA804531 1,077 bp 32% thrombin 4503637 hCHN6 EST 2134670 1,503 bp 36% edg-l NP_001391 hCHN8 EST 764455 1,029 bp 47% D13626 KIAA0001 hCHN9 EST 1541536 1,077 bp 41% LTB4R NM 000752 hCHNIO EST 1365839 1,055 bp 35% P2Y NM_002563
Receptor homology is useful in terms of gaining an appreciation of a role of the
receptors within the human body. As the patent document progresses, we will disclose
techniques for mutating these receptors to establish non-endogenous, constitutively activated
versions of these receptors.
The techniques disclosed herein have also been applied to other human, oφhan
GPCRs known to the art, as will be apparent as the patent document progresses.
C. Receptor Screening
Screening candidate compounds against a non-endogenous, constitutively activated
version of the human GPCRs disclosed herein allows for the direct identification of candidate
compounds which act at this cell surface receptor, without requiring use of the receptor's
endogenous ligand. By determining areas within the body where the endogenous version of
human GPCRs disclosed herein is expressed and/or over-expressed, it is possible to determine
related disease/disorder states which are associated with the expression and/or over-expression of the receptor; such an approach is disclosed in this patent document.
With respect to creation of a mutation that may evidence constitutive activation of the
human GPCR disclosed herein is based upon the distance from the proline residue at which
is presumed to be located within TM6 of the GPCR; this algorithmic technique is disclosed
in co-pending and commonly assigned patent document U.S. Serial Number 09/170,496,
incoφorated herein by reference. The algorithmic technique is not predicated upon traditional
sequence "alignment" but rather a specified distance from the aforementioned TM6 proline
residue. By mutating the amino acid residue located 16 amino acid residues from this residue
(presumably located in the IC3 region of the receptor) to, most preferably, a lysine residue,
such activation may be obtained. Other amino acid residues may be useful in the mutation
at this position to achieve this objective.
D. Disease/Disorder Identification and/or Selection
As will be set forth in greater detail below, most preferably inverse agonists to the
non-endogenous, constitutively activated GPCR can be identified by the methodologies of this
invention. Such inverse agonists are ideal candidates as lead compounds in drug discovery
programs for treating diseases related to this receptor. Because of the ability to directly
identify inverse agonists to the GPCR, thereby allowing for the development of
pharmaceutical compositions, a search for diseases and disorders associated with the GPCR
is relevant. For example, scanning both diseased and normal tissue samples for the presence
of the GPCR now becomes more than an academic exercise or one which might be pursued
along the path of identifying an endogenous ligand to the specific GPCR. Tissue scans can
be conducted across a broad range of healthy and diseased tissues. Such tissue scans provide
a preferred first step in associating a specific receptor with a disease and/or disorder. See, for example, co-pending application (docket number ARE-0050) for exemplary dot-blot and RT-
PCR results of several of the GPCRs disclosed herein.
Preferably, the DNA sequence of the human GPCR is used to make a probe for (a)
dot-blot analysis against tissue-mRNA, and/or (b) RT-PCR identification of the expression
of the receptor in tissue samples. The presence of a receptor in a tissue source, or a
diseased tissue, or the presence of the receptor at elevated concentrations in diseased tissue
compared to a normal tissue, can be preferably utilized to identify a correlation with a
treatment regimen, including but not limited to, a disease associated with that disease.
Receptors can equally well be localized to regions of organs by this technique. Based on
the known functions of the specific tissues to which the receptor is localized, the putative
functional role of the receptor can be deduced.
E. Screening of Candidate Compounds
1. Generic GPCR screening assay techniques
When a G protein receptor becomes constitutively active, it binds to a G protein (e.g.,
Gq, Gs, Gi, Gz, Go) and stimulates the binding of GTP to the G protein. The G protein then
acts as a GTPase and slowly hydrolyzes the GTP to GDP, whereby the receptor, under normal
conditions, becomes deactivated. However, constitutively activated receptors continue to
exchange GDP to GTP. A non-hydrolyzable analog of GTP, [35S]GTPγS, can be used to
monitor enhanced binding to membranes which express constitutively activated receptors.
It is reported that [35S] GTPγS can be used to monitor G protein coupling to membranes in the
absence and presence of ligand. An example of this monitoring, among other examples well-
known and available to those in the art, was reported by Traynor and Nahorski in 1995. The
preferred use of this assay system is for initial screening of candidate compounds because the system is generically applicable to all G protein-coupled receptors regardless of the particular
G protein that interacts with the intracellular domain of the receptor.
2. Specific GPCR screening assay techniques
Once candidate compounds are identified using the "generic" G protein-coupled
receptor assay (/. e., an assay to select compounds that are agonists, partial agonists, or inverse
agonists), further screening to confirm that the compounds have interacted at the receptor site
is preferred. For example, a compound identified by the "generic" assay may not bind to the
receptor, but may instead merely "uncouple" the G protein from the intracellular domain.
a. Gs, Gz and Gi.
Gs stimulates the enzyme adenylyl cyclase. Gi (and Gz and Go), on the other hand,
inhibit this enzyme. Adenylyl cyclase catalyzes the conversion of ATP to cAMP; thus,
constitutively activated GPCRs that couple the Gs protein are associated with increased
cellular levels of cAMP. On the other hand, constitutively activated GPCRs that couple Gi
(or Gz, Go) protein are associated with decreased cellular levels of cAMP. See, generally,
"Indirect Mechanisms of Synaptic Transmission," Chpt. 8, From Neuron To Brain (3rd Ed.)
Nichols, J.G. et al eds. Sinauer Associates, Inc. (1992). Thus, assays that detect cAMP can
be utilized to determine if a candidate compound is, e.g., an inverse agonist to the receptor
(i.e., such a compound would decrease the levels of c AMP). A variety of approaches known
in the art for measuring cAMP can be utilized; a most preferred approach relies upon the use
of anti-cAMP antibodies in an ELISA-based format. Another type of assay that can be
utilized is a whole cell second messenger reporter system assay. Promoters on genes drive
the expression of the proteins that a particular gene encodes. Cyclic AMP drives gene
expression by promoting the binding of a cAMP -responsive DNA binding protein or transcription factor (CREB) that then binds to the promoter at specific sites called cAMP
response elements and drives the expression of the gene. Reporter systems can be constructed
which have a promoter containing multiple cAMP response elements before the reporter gene,
e.g., β-galactosidase or luciferase. Thus, a constitutively activated Gs-linked receptor causes
the accumulation of cAMP that then activates the gene and expression of the reporter protein.
The reporter protein such as β-galactosidase or luciferase can then be detected using standard
biochemical assays (Chen et al. 1995).
b. Go and Gq.
Gq and Go are associated with activation of the enzyme phospholipase C, which in
turn hydrolyzes the phospholipid PIP2, releasing two intracellular messengers:
diacycloglycerol (DAG) and inistol 1,4,5-triphoisphate (IP3). Increased accumulation of IP3
is associated with activation of Gq- and Go-associated receptors. See, generally, "Indirect
Mechanisms of Synaptic Transmission," Chpt. 8, From Neuron To Brain (3rd Ed.) Nichols,
J.G. et al eds. Sinauer Associates, Inc. (1992). Assays that detect IP3 accumulation can be
utilized to determine if a candidate compound is, e.g., an inverse agonist to a Gq- or Go-
associated receptor (i.e., such a compound would decrease the levels of IP3). Gq-associated
receptors can also been examined using an API reporter assay in that Gq-dependent
phospholipase C causes activation of genes containing API elements; thus, activated Gq-
associated receptors will evidence an increase in the expression of such genes, whereby
inverse agonists thereto will evidence a decrease in such expression, and agonists will
evidence an increase in such expression. Commercially available assays for such detection
are available. 3. GPCR Fusion Protein
The use of an endogenous, constitutively activate oφhan GPCR or anon-endogenous,
constitutively activated oφhan GPCR, for use in screening of candidate compounds for the
direct identification of inverse agonists, agonists and partial agonists provide an interesting
screening challenge in that, by definition, the receptor is active even in the absence of an
endogenous ligand bound thereto. Thus, in order to differentiate between, e.g., the non-
endogenous receptor in the presence of a candidate compound and the non-endogenous
receptor in the absence of that compound, with an aim of such a differentiation to allow for
an understanding as to whether such compound may be an inverse agonist, agonist, partial
agonist or have no affect on such a receptor, it is preferred that an approach be utilized that
can enhance such differentiation. A preferred approach is the use of a GPCR Fusion Protein.
Generally, once it is determined that a non-endogenous oφhan GPCR has been
constitutively activated using the assay techniques set forth above (as well as others), it is
possible to determine the predominant G protein that couples with the endogenous GPCR.
Coupling of the G protein to the GPCR provides a signaling pathway that can be assessed.
Because it is most preferred that screening take place by use of a mammalian expression
system, such a system will be expected to have endogenous G protein therein. Thus, by
definition, in such a system, the non-endogenous, constitutively activated oφhan GPCR will
continuously signal. In this regard, it is preferred that this signal be enhanced such that in the
presence of, e.g., an inverse agonist to the receptor, it is more likely that it will be able to more
readily differentiate, particularly in the context of screening, between the receptor when it is
contacted with the inverse agonist.
The GPCR Fusion Protein is intended to enhance the efficacy of G protein coupling with the non-endogenous GPCR. The GPCR Fusion Protein is preferred for screening with
a non-endogenous, constitutively activated GPCR because such an approach increases the
signal that is most preferably utilized in such screening techniques. This is important in
facilitating a significant "signal to noise" ratio; such a significant ratio is import preferred for
the screening of candidate compounds as disclosed herein.
The construction of a construct useful for expression of a GPCR Fusion Protein is
within the purview of those having ordinary skill in the art. Commercially available
expression vectors and systems offer a variety of approaches that can fit the particular needs
of an investigator. The criteria of importance for such a GPCR Fusion Protein construct is
that the endogenous GPCR sequence and the G protein sequence both be in-frame (preferably,
the sequence for the endogenous GPCR is upstream of the G protein sequence) and that the
"stop" codon of the GPCR must be deleted or replaced such that upon expression of the
GPCR, the G protein can also be expressed. The GPCR can be linked directly to the G
protein, or there can be spacer residues between the two (preferably, no more than about 12.
although this number can be readily ascertained by one of ordinary skill in the art). We have
a preference (based upon convenience) of use of a spacer in that some restriction sites that are
not used will, effectively, upon expression, become a spacer. Most preferably, the G protein
that couples to the non-endogenous GPCR will have been identified prior to the creation of
the GPCR Fusion Protein construct. Because there are only a few G proteins that have been
identified, it is preferred that a construct comprising the sequence of the G protein (i.e., a
universal G protein construct) be available for insertion of an endogenous GPCR sequence
therein; this provides for efficiency in the context of large-scale screening of a variety of
different endogenous GPCRs having different sequences. As noted above, constitutively activated GPCRs that couple to Gi, Gz and Go are
expected to inhibit the formation of c AMP making assays based upon these types of GPCRs
challenging (i.e., the cAMP signal decreases upon activation thus making the direct
identification of, e.g. inverse agonists (which would further decrease this signal), interesting).
As will be disclosed herein, we have ascertained that for these types of receptors, it is possible
to create a GPCR Fusion Protein that is not based upon the endogenous GPCR's endogenous
G protein, in an effort to establish a viable cyclase-based assay. Thus, for example, a Gz
coupled receptor such as H9, a GPCR Fusion Protein can be established that utilizes a Gs
fusion protein - we believe that such a fusion construct, upon expression, "drives" or "forces"
the non-endogenous GPCR to couple with, e.g., Gs rather than the "natural" Gz protein, such
that a cyclase-based assay can be established. Thus, for Gi, Gz and Go coupled receptors, we
prefer that that when a GPCR Fusion Protein is used and the assay is based upon detection of
adenyl cyclase activity, that the fusion construct be established with Gs (or an equivalent G
protein that stimulates the formation of the enzyme adenylyl cyclase).
F. Medicinal Chemistry
Generally, but not always, direct identification of candidate compounds is preferably
conducted in conjunction with compounds generated via combinatorial chemistry techniques,
whereby thousands of compounds are randomly prepared for such analysis. Generally, the
results of such screening will be compounds having unique core structures; thereafter, these
compounds are preferably subjected to additional chemical modification around a preferred
core structure(s) to further enhance the medicinal properties thereof. Such techniques are
known to those in the art and will not be addressed in detail in this patent document. G. Pharmaceutical compositions
Candidate compounds selected for further development can be formulated into
pharmaceutical compositions using techniques well known to those in the art. Suitable
pharmaceutically-acceptable carriers are available to those in the art; for example, see
Remington's Pharmaceutical Sciences, 16th Edition, 1980, Mack Publishing Co., (Oslo et al.,
eds.)
H. Other Utility
Although a preferred use of the non-endogenous versions the human GPCRs disclosed
herein may be for the direct identification of candidate compounds as inverse agonists,
agonists or partial agonists (preferably for use as pharmaceutical agents), these versions of
human GPCRs can also be utilized in research settings. For example, in vitro and in vivo
systems incoφorating GPCRs can be utilized to further elucidate and understand the roles
these receptors play in the human condition, both normal and diseased, as well as
understanding the role of constitutive activation as it applies to understanding the signaling
cascade. The value in non-endogenous human GPCRs is that their utility as a research tool
is enhanced in that, because of their unique features, non-endogenous human GPCRs can be
used to understand the role of these receptors in the human body before the endogenous
ligand therefor is identified. Other uses of the disclosed receptors will become apparent to
those in the art based upon, inter alia, a review of this patent document.
EXAMPLES
The following examples are presented for puφoses of elucidation, and not limitation, of
the present invention. While specific nucleic acid and amino acid sequences are disclosed
herein, those of ordinary skill in the art are credited with the ability to make minor modifications to these sequences while achieving the same or substantially similar results
reported below. The traditional approach to application or understanding of sequence
cassettes from one sequence to another (e.g. from rat receptor to human receptor or from
human receptor A to human receptor B) is generally predicated upon sequence alignment
techniques whereby the sequences are aligned in an effort to determine areas of commonality .
The mutational approach disclosed herein does not rely upon this approach but is instead
based upon an algorithmic approach and a positional distance from a conserved proline
residue located within the TM6 region of human GPCRs. Once this approach is secured,
those in the art are credited with the ability to make minor modifications thereto to achieve
substantially the same results (i.e., constitutive activation) disclosed herein. Such modified
approaches are considered within the purview of this disclosure
Example 1
ENDOGENOUS HUMAN GPCRS
1. Identification of Human GPCRs
Certain of the disclosed endogenous human GPCRs were identified based upon a
review of the GenBank™ database information. While searching the database, the following
cDNA clones were identified as evidenced below (Table C).
TABLE C
Disclosed Accession Complete DNA Open Reading Nucleic Amino
Human Number Sequence Frame Acid Acid
Orphan (Base Pairs) (Base Pairs) SEQ.ID. SEQ.ID.
GPCRs NO. NO. hARE-3 AL033379 111,389 bp 1,260 bp 1 2 hARE-4 AC006087 226,925 bp 1,119 bp 4 hARE-5 AC006255 127,605 bp 1,104 bp 5 6 hRUP3 AL035423 140,094 bp 1,005 bp 7 8 hRUP5 AC005849 169,144 bp 1,413 bp 9 10 hRUP6 AC005871 218,807 bp 1 ,245 bp 11 12 hRUP7 AC007922 158,858 bp 1,173 bp 13 14
Other disclosed endogenous human GPCRs were identified by conducting a BLAST™
search of EST database (dbest) using the following EST clones as query sequences. The
following EST clones identified were then used as a probe to screen a human genomic library
(Table D).
TABLE D
Disclosed Query EST Clone/ Open Nucleic Acid Amino Acid
Human (Sequence) Accession No. Reading SEQ.ID.NO. SEQ.ID.NO.
Orphan Identified Frame
GPCRs (Base Pairs) hGPCR27 Mouse AA775870 1,125 bp 17 18 GPCR27 hARE-1 TDAG 1689643 999 bp 19 20 AI090920 hARE-2 GPCR27 68530 1,122 bp 21 22 AA359504 hPPRl Bovine 238667 1 ,053 bp 23 24 PPR1 H67224 hG2A Mouse See Example 2(a), 1,113 bp 25 26 1 179426 below hCHN3 N.A. EST 36581 1.113 bp 27 28 (full length) hCHN4 TDAG 1184934 1 ,077 bp 29 30 AA804531 hCHN6 N.A. EST 2134670 1 ,503 bp 31 32 (full length) hCHN8 KIAA0001 EST 764455 1,029 bp 33 34 hCHN 9 1365839 EST 1541536 1 ,077 bp 35 36 hCHNIO Mouse EST Human 1365839 1,005 bp 37 38 1365839 hRUP4 N.A. AI307658 1,296 bp 39 40
N.A. = "not applicable".
2. Full Length Cloning
a. Human G2A
Mouse EST clone 1179426 was used to obtain a human genomic clone containing all but three amino acid G2A coding sequences. The 5 'of this coding sequence was obtained by
using 5 'RACE, and the template for PCR was Clontech' s Human Spleen Marathon-Ready™
cDNA. The disclosed human G2A was amplified by PCR using the G2A cDNA specific
primers for the first and second round PCR as shown in SEQ.ID.NO.: 41 and SEQ.ID.NO. :42
as follows:
5'-CTGTGTACAGCAGTTCGCAGAGTG-3' (SEQ.ID.NO.: 41 ; 1st round PCR)
5'-GAGTGCCAGGCAGAGCAGGTAGAC-3' (SEQ.ID.NO.: 42; second round PCR).
PCR was performed using Advantage GC Polymerase Kit (Clontech; manufacturing
instructions will be followed), at 94°C for 30 sec followed by 5 cycles of 94 °C for 5 sec and
72°C for 4 min; and 30 cycles of 94° for 5 sec and 70° for 4 min. An approximate 1.3 Kb
PCR fragment was purified from agarose gel, digested with Hind III and Xba I and cloned into
the expression vector pRC/CMV2 (Invitrogen). The cloned-insert was sequenced using the
T7 Sequenase™ kit (USB Amersham; manufacturer instructions followed) and the sequence
was compared with the presented sequence. Expression of the human G2A was detected by
probing an RNA dot blot (Clontech; manufacturer instructions followed) with the P32-labeled
fragment.
b. CHN9
Sequencing of the EST clone 1541536 showed CHN9 to be a partial cDNA clone
having only an initiation codon; i.e., the termination codon was missing. When CHN9
was used to blast against data base (nr), the 3' sequence of CHN9 was 100% homologous
to the 5' untranslated region of the leukotriene B4 receptor cDNA, which contained a
termination codon in the frame with CHN9 coding sequence. To determine whether the 5'
untranslated region of LTB4R cDNA was the 3' sequence of CHN9, PCR was performed
using primers based upon the 5' sequence flanking the initiation codon found in CHN9 and the 3' sequence around the termination codon found in the LTB4R 5' untranslated region.
The 5' primer sequence utilized was as follows:
5'-CCCGAATTCCTGCTTGCTCCCAGCTTGGCCC-3' (SEQ.ID.NO.: 43; sense) and 5'-TGTGGATCCTGCTGTCAAAGGTCCCATTCCGG-3' (SEQ.ID.NO.: 44; antisense). PCR was performed using thymus cDNA as a template and rTth polymerase (Perkin Elmer)
with the buffer system provided by the manufacturer, 0.25 uM of each primer, and 0.2 mM
of each 4 nucleotides. The cycle condition was 30 cycles of 94°C for 1 min, 65°C for lmin
and 72 °C for 1 min and 10 sec. A 1.1 kb fragment consistent with the predicted size was
obtained from PCR. This PCR fragment was subcloned into pCMV (see below) and
sequenced (see, SEQ.ID.NO.: 35).
c. RUP 4
The full length RUP4 was cloned by RT-PCR with human brain cDNA (Clontech) as
templates:
5'-TCACAATGCTAGGTGTGGTC-3' (SEQ.ID.NO.: 45; sense) and
5'-TGCATAGACAATGGGATTACAG-3' (SEQ.ID.NO.: 46; antisense).
PCR was performed using TaqPlus Precision™ polymerase (Stratagene; manufacturing
instructions followed) by the following cycles: 94°C for 2 min; 94°C 30 sec; 55 °C for 30 sec,
72°C for 45 sec, and 72°C for 10 min. Cycles 2 through 4 were repeated 30 times.
The PCR products were separated on a 1% agarose gel and a 500 bp PCR fragment
was isolated and cloned into the pCRII-TOPO™ vector (Invitrogen) and sequenced using the
T7 DNA Sequenase™ kit (Amsham) and the SP6/T7 primers (Stratagene). Sequence analysis
revealed that the PCR fragment was indeed an alternatively spliced form of AI307658 having
a continuous open reading frame with similarity to other GPCRs. The completed sequence
of this PCR fragment was as follows: 5'-TCACAATGCTAGGTGTGGTCTGGCTGGTGGCAGTCATCGTAGGATCACCCATGTGGCAC GTGCAACAACTTGAGATCAAATATGACTTCCTATATGAAAAGGAACACATCTGCTGCTTAAGA GTGGACCAGCCCTGTGCACCAGAAGATCTACACCACCTTCATCCTTGTCATCCTCTTCCTCCTGC CTCTTATGGTGATGCTTATTCTGTACGTAAAATTGGTTATGAACTTTGGATAAAGAAAAGAGTT GGGGATGGTTCAGTGCTTCGAACTATTCATGGAAAAGAAATGTCCAAAATAGCCAGGAAGAAG AAACGAGCTGTCATTATGATGGTGACAGTGGTGGCTCTCTTTGCTGTGTGCTGGGCACCATTCC ATGTTGTCCATATGATGATTGAATACAGTAATTTTGAAAAGGAATATGATGATGTCACAATCAA GATGATTTTTGCTATCGTGCAAATTATTGGATTTTCCAACTCCATCTGTAATCCCATTGTCTATGCA- 3' (SEQ.ID.NO.: 47)
Based on the above sequence, two sense oligonucleotide primer sets:
5'-CTGCTTAGAAGAGTGGACCAG-3' (SEQ.ID.NO.: 48; oligo 1),
5'-CTGTGCACCAGAAGATCTACAC-3' (SEQ.IDNO.: 49; oligo 2) and
two antisense oligonucleotide primer sets:
5'-CAAGGATGAAGGTGGTGTAGA-3' (SEQ.ID.NO.: 50; oligo 3) 5'-GTGTAGATCTTCTGGTGCACAGG-3' (SEQ.ID.NO.: 51; oligo 4) were used for 3'- and 5 '-RACE PCR with a human brain Marathon-Ready™ cDNA
(Clontech, Cat# 7400-1) as template, according to manufacture's instructions. DNA
fragments generated by the RACE PCR were cloned into the pCRII-TOPO™ vector
(Invitrogen) and sequenced using the SP6/T7 primers (Stratagene) and some internal primers.
The 3' RACE product contained a poly(A) tail and a completed open reading frame ending
at a TAA stop codon. The 5' RACE product contained an incomplete 5' end; i.e., the ATG
initiation codon was not present.
Based on the new 5' sequence, oligo 3 and the following primer:
5'-GCAATGCAGGTCATAGTGAGC -3' (SEQ.ID.NO.: 52; oligo 5) were used for the second round of 5 ' race PCR and the PCR products were analyzed as above.
A third round of 5' race PCR was carried out utilizing antisense primers:
5'-TGGAGCATGGTGACGGGAATGCAGAAG-3' (SEQ.ID.NO.: 53: oligo 6) and
5'-GTGATGAGCAGGTCACTGAGCGCCAAG-3' (SEQ.ID.NO.: 54: oligo7). The sequence of the 5' RACE PCR products revealed the presence of the initiation codon ATG, and further round of 5' race PCR did not generate any more 5' sequence. The
completed 5' sequence was confirmed by RT-PCR using sense primer 5'-GCAATGCAGGCGCTTAACATTAC-3' (SEQ.ID.NO.: 55; oligo 8) and oligo 4 as primers and sequence analysis of the 650 bp PCR product generated from
human brain and heart cDNA templates (Clontech, Cat# 7404- 1 ) . The completed 3 ' sequence
was confirmed by RT-PCR using oligo 2 and the following antisense primer: 5'-TTGGGTTACAATCTGAAGGGCA-3' (SEQ.ID.NO.:56; oligo 9) and sequence analysis of the 670 bp PCR product generated from human brain and heart
cDNA templates. (Clontech, Cat# 7404-1).
d. RUP5
The full length RUP5 was cloned by RT-PCR using a sense primer upstream from
ATG, the initiation codon (SEQ.ID.NO.:57), and an antisense primer containing TCA as the
stop codon (SEQ.ID.NO. :58), which had the following sequences:
5'-ACTCCGTGTCCAGCAGGACTCTG-3' (SEQ.ID.NO.: 57) 5'-TGCGTGTTCCTGGACCCTCACGTG-3' (SEQ.ID.NO.: 58) and human peripheral leukocyte cDNA (Clontech) as a template. Advantage™ cDNA
polymerase (Clontech) was used for the amplification in a 50ul reaction by the following cycle
with step 2 through step 4 repeated 30 times: 94°C for 30 sec; 94° for 15 sec; 69° for 40 sec;
72°C for 3 min; and 72°C fro 6 min. A 1.4kb PCR fragment was isolated and cloned with
the pCRII-TOPO™ vector (Invitrogen) and completely sequenced using the T7 DNA
Sequenase™ kit (Amsham). See, SEQ.ID.NO.: 9.
e. RUP6
The full length RUP6 was cloned by RT-PCR using primers:
5'-CAGGCCTTGGATTTTAATGTCAGGGATGG-3' (SEQ.ID.NO.: 59) and 5'-GGAGAGTCAGCTCTGAAAGAATTCAGG-3' (SEQ.ID.NO.: 60); and human thymus Marathon-Ready™ cDNA (Clontech) as a template. Advantage cDNA
polymerase (Clontech, according to manufacturer's instructions) was used for the
amplification in a 50ul reaction by the following cycle: 94°C for 30sec; 94°C for 5 sec; 66°C
for 40sec; 72 °C for 2.5 sec and 72 °C for 7 min. Cycles 2 through 4 were repeated 30 times.
A 1.3 Kb PCR fragment was isolated and cloned into the pCRII-TOPO™ vector (Invitrogen)
and completely sequenced (see, SEQ.ID.NO.: 11) using the ABI Big Dye Terminator™ kit
(P.E. Biosystem).
f. RUP7
The full length RUP7 was cloned by RT-PCR using primers:
5'-TGATGTGATGCCAGATACTAATAGCAC-3' (SEQ.ID.NO.: 61 ; sense) and
5'-CCTGATTCATTTAGGTGAGATTGAGAC-3' (SEQ.ID.NO.: 62; antisense) and human peripheral leukocyte cDNA (Clontech) as a template. Advantage™ cDNA
polymerase (Clontech) was used for the amplification in a 50 ul reaction by the following
cycle with step 2 to step 4 repeated 30 times: 94 °C for 2 minutes; 94 °C for 15 seconds; 60°C
for 20 seconds; 72 °C for 2 minutes; 72 °C for 10 minutes. A 1.25 Kb PCR fragment was
isolated and cloned into the pCRII-TOPO™ vector (Invitrogen) and completely sequenced
using the ABI Big Dye Terminator™ kit (P.E. Biosystem). See, SEQ.ID.NO.: 13.
3. Angiotensin II Type 1 Receptor ("ATI")
The endogenous human angiotensin II type 1 receptor ("ATI ") was obtained by PCR
using genomic DNA as template and rTth polymerase (Perkin Elmer) with the buffer system
provided by the manufacturer, 0.25 μM of each primer, and 0.2 mM of each 4 nucleotides.
The cycle condition was 30 cycles of 94°C for 1 min, 55°C for lmin and 72 °C for 1.5 min.
The 5' PCR primer contains a Hindlll site with the sequence: 5'-CCCAAGCTTCCCCAGGTGTATTTGAT-3' (SEQ.ID.NO.: 63) and the 3' primer contains a BamHl site with the following sequence:
5'-GTTGGATCCACATAATGCATTTTCTC-3' (SEQ.ID.NO.: 64).
The resulting 1.3 kb PCR fragment was digested with Hindlll and BamHl and cloned into
Hindlll-BamHI site of pCMV expression vector. The cDNA clone was fully sequenced.
Nucleic acid (SEQ.ID.NO.: 65) and amino acid (SEQ.ID.NO.: 66) sequences for human ATI
were thereafter determined and verified.
4. GPR38
To obtain GPR38, PCR was performed by combining two PCR fragments, using
human genomic cDNA as template and rTth poymerase (Perkin Elmer) with the buffer system
provided by the manufacturer, 0.25uM of each primer, and 0.2 mM of each 4 nucleotides.
The cycle condition for each PCR reaction was 30 cycles of 94 °C for 1 min, 62 °C for lmin
and 72 °C for 2 min.
The first fragment was amplified with the 5' PCR primer that contained an end site
with the following sequence:
5'-ACCATGGGCAGCCCCTGGAACGGCAGC-3' (SEQ.ID.NO.:67) and a 3' primer having the following sequence:
5'-AGAACCACCACCAGCAGGACGCGGACGGTCTGCCGGTGG-3' (SEQ.ID.NO.:68).
The second PCR fragment was amplified with a 5' primer having the following sequence: 5'-GTCCGCGTCCTGCTGGTGGTGGTTCTGGCATTTATAATT-3' (SEQ.ID.NO.: 69) and a 3' primer that contained a BamHl site and having the following sequence:
5'-CCTGGATCCTTATCCCATCGTCTTCACGTTAGC-3' (SEQ.ID.NO.: 70).
The two fragments were used as templates to amplify GPR38, using SEQ.ID.NO.: 67 and
SEQ.ID.NO.: 70 as primers (using the above-noted cycle conditions). The resulting 1.44kb PCR fragment was digested with BamHl and cloned into Blunt-BamHI site of pCMV
expression vector.
5. MC4
To obtain MC4, PCR was performed using human genomic cDNA as template and
rTth poymerase (Perkin Elmer) with the buffer system provided by the manufacturer, 0.25uM
of each primer, and 0.2 mM of each 4 nucleotides. The cycle condition for each PCR reaction
was 30 cycles of 94°C for 1 min, 54°C for lmin and 72°C for 1.5 min.
The 5' PCR contained an EcoRI site with the sequence:
5'-CTGGAATTCTCCTGCCAGCATGGTGA-3' (SEQ.ID.NO.: 71) and the 3' primer contained a BamHl site with the sequence:
5'-GCAGGATCCTATATTGCGTGCTCTGTCCCC'-3 (SEQ.ID.NO.: 72).
The 1.0 kb PCR fragment was digest with EcoRI and BamHl and cloned into EcoRI -BamHl
site of pCMV expression vector. Nucleic acid (SEQ.ID.NO.: 73) and amino acid
(SEQ.ID.NO.: 74) sequences for human MC4 were thereafter determined.
6. CCKB
To obtain CCKB, PCR was performed using human stomach cDNA as template and
rTth poymerase (Perkin Elmer) with the buffer system provided by the manufacturer, 0.25uM
of each primer, and 0.2 mM of each 4 nucleotides. The cycle condition for each PCR reaction
was 30 cycles of 94°C for 1 min, 65°C for lmin and 72°C for 1 min and 30 sec.
The 5' PCR contained a Hindlll site with the sequence:
5'-CCGAAGCTTCGAGCTGAGTAAGGCGGCGGGCT-3' (SEQ.ID.NO.: 75) and the 3' primer contained an EcoRI site with the sequence:
5'-GTGGAATTCATTTGCCCTGCCTCAACCCCCA-3 (SEQ.ID.NO.: 76).
The resulting 1.44 kb PCR fragment was digest with Hindlll and EcoRI and cloned into Hindlll-EcoRI site of pCMV expression vector. Nucleic acid (SEQ.ID.NO.: 77) and amino
acid (SEQ.ID.NO.: 78) sequences for human CCKB were thereafter determined.
7. TDAG8
To obtain TDAG8, PCR was performed using genomic DNA as template and rTth
polymerase (Perkin Elmer) with the buffer system provided by the manufacturer, 0.25 μM of
each primer, and 0.2 mM of each 4 nucleotides. The cycle condition was 30 cycles of 94°C
for 1 min, 56°C for lmin and 72 °C for 1 min and 20 sec. The 5' PCR primer contained a
Hindlll site with the following sequence:
5'-TGCAAGCTTAAAAAGGAAAAAATGAACAGC-3' (SEQ.ID.NO.: 79) and the 3' primer contained a BamHl site with the following sequence:
5'-TAAGGATCCCTTCCCTTCAAAACATCCTTG -3' (SEQ.ID.NO.: 80).
The resulting 1.1 kb PCR fragment was digested with Hindlll and BamHl and cloned into
Hindlll -BamHl site of pCMV expression vector. Three resulting clones sequenced contained
three potential polymoφhisms involving changes of amino acid 43 from Pro to Ala, amino
acid 97 from Lys to Asn and amino acid 130 from He to Phe. Nucleic acid (SEQ.ID.NO.: 81)
and amino acid (SEQ.ID.NO.: 82) sequences for human TDAG8 were thereafter determined.
8. H9
To obtain H9, PCR was performed using pituitary cDNA as template and rTth
polymerase (Perkin Elmer) with the buffer system provided by the manufacturer, 0.25 μM of
each primer, and 0.2 mM of each 4 nucleotides. The cycle condition was 30 cycles of 94°C
for 1 min, 62°C for 1 min and 72°C for 2 min. The 5' PCR primer contained a Hindlll site
with the following sequence:
5'-GGAAAGCTTAACGATCCCCAGGAGCAACAT-3' (SEQ.ID.NO.: 15)
and the 3' primer contained a BamHl site with the following sequence: 5'-CTGGGATCCTACGAGAGCATTTTTCACACAG-3' (SEQ.ID.NO.:16).
The resulting 1.9 kb PCR fragment was digested with Hindlll and BamHl and cloned into
Hindlll-BamHI site of pCMV expression vector. H9 contained three potential polymoφhisms
involving changes of amino acid P320S, S493N and amino acid G448A. Nucleic acid
(SEQ.ID.NO.: 139) and amino acid (SEQ.ID.NO.: 140) sequences for human H9 were
thereafter determined and verified.
Example 2
PREPARATION OF NON-ENDOGENOUS, CONSTITUTIVELY ACTIVATED GPCRS
Those skilled in the art are credited with the ability to select techniques for
mutation of a nucleic acid sequence. Presented below are approaches utilized to create
non-endogenous versions of several of the human GPCRs disclosed above. The mutations
disclosed below are based upon an algorithmic approach whereby the 16th amino acid
(located in the IC3 region of the GPCR) from a conserved proline residue (located in the
TM6 region of the GPCR, near the TM6/IC3 interface) is mutated, most preferably to a
lysine amino acid residue.
1. Tranformer Site-Directed ™ Mutagenesis
Preparation of non-endogenous human GPCRs may be accomplished on human
GPCRs using Transformer Site-Directed™ Mutagenesis Kit (Clontech) according to the
manufacturer instructions. Two mutagenesis primers are utilized, most preferably a lysine
mutagenesis oligonucleotide that creates the lysine mutation, and a selection marker
oligonucleotide. For convenience, the codon mutation to be incoφorated into the human
GPCR is also noted, in standard form (Table E): TABLE E
Receptor Identifier Codon Mutation hARE-3 F313K hARE-4 V233K hARE-5 A240K hGPCR14 L257K hGPCR27 C283K hARE-1 E232K hARE-2 G285K hPPRl L239K hG2A K232A hRUP3 L224K hRUP5 A236K hRUPό N267K hRUP7 A302K hCHN4 V236K hMC4 A244K hCHN3 S284K hCHN6 L352K hCHN8 N235K hCHN9 G223K hCHNlO L231K hH9 F236K
The following GPCRs were mutated according with the above method using the
designated sequence primers (Table F).
TABLE F
Receptor Codon Lysine Mutagenesis Selection Marker
Identifier Mutation (SEQ.ID.NO.) (SEQ.ID.NO.)
5'-3' orientation, mutation 5'-3' orientation sequence underlined hRUP4 V272K CAGGAAGAAGAAACGAGC CACTGTCACCATCATAATG
TGTCATTATGATGGTGACA ACAGCTCGTTTCTTCTTCC
GTG (83) TG (84) hATl see below alternative approach; see below alternative approach; see below hGPR38 V297K GGCCACCGGCAGACCAAAC CTCCTTCGGTCCTCCTATC
GCGTCCTGCTG (85) GTTGTCAGAAGT (86) hCCKB V332K alternative approach; see below alternative approach; see below hTDAG8 I225K GGAAAAGAAGAGAATCAA CTCCTTCGGTCCTCCTATC
AAAACTACTTGTCAGCATC GTTGTCAGAAGT (88)
(87) hH9 F236K GCTGAGGTTCGCAATAAAC CTCCTTCGGTCCTCCTATC
TAACCATGTTTGTG (143) GTTGTCAGAAGT (144) hMC4 A244K GCCAATATGAAGGGAAAA CTCCTTCGGTCCTCCTATC
ATTACCTTGACCATC (137) GTTGTCAGAAGT (138)
The non-endogenous human GPCRs were then sequenced and the derived and
verified nucleic acid and amino acid sequences are listed in the accompanying "Sequence
Listing" appendix to this patent document, as summarized in Table G below:
TABLE G
Non Endogenous Human Nucleic Acid Sequence Listing Amino Acid Sequence GPCR Listing hRUP4 SEQ.ID.NO.: 127 SEQ.ID.NO.: 128
(V272K) hATl (see alternative approaches (see alternative approaches,
(see alternative approaches below) below) below) hGPR38 SEQ.ID.NO.: 129 SEQ.ID.NO.: 130
(V297K) hCCKB SEQ.ID.NO.: 131 SEQ.ID.NO.: 132
(V332K)
HTDAG8 SEQ.ID.NO.: 133 SEQ.ID.NO.: 134
(I225K) hH9 SEQ.ID.NO.: 141 SEQ.ID.NO.: 142
(F236K) hMC4 SEQ.ID.NO.: 135 SEQ.ID.NO.: 136
(A244K) 2. Alternative Approaches For Creation of Non-Endogenous Human GPCRs
a. ATI
1. F239K Mutation
Preparation of a non-endogenous, constitutively activated human ATI receptor was
accomplished by creating an F239K mutation (see, SEQ.ID.NO. : 89 for nucleic acid sequence,
and SEQ.ID.NO.: 90 for amino acid sequence). Mutagenesis was performed using
Transformer Site-Directed Mutagenesis™ Kit (Clontech) according to the to manufacturer's
instructions. The two mutagenesis primers were used, a lysine mutagenesis oligonucleotide
(SEQ.ID.NO.: 91) and a selection marker oligonucleotide (SEQ.ID.NO.: 92), which had the
following sequences:
5'-CCAAGAAATGATGATATTAAAAAGATAATTATGGC-3' (SEQ.ID.NO.: 91) 5'-CTCCTTCGGTCCTCCTATCGTTGTCAGAAGT-3' (SEQ.ID.NO.: 92),
respectively.
2. N111A Mutation
Preparation of a non-endogenous human ATI receptor was also accomplished by
creating an Nl 11 A mutation (see, SEQ.ID.NO. :93 for nucleic acid sequence, and
SEQ.ID.NO.: 94 for amino acid sequence). Two PCR reactions were performed using pfu
polymerase (Stratagene) with the buffer system provided by the manufacturer,
supplemented with 10% DMSO, 0.25 μM of each primer, and 0.5 mM of each 4
nucleotides. The 5' PCR sense primer used had the following sequence: 5'-CCCAAGCTTCCCCAGGTGTATTTGAT-3' (SEQ.ID.NO.: 95) and the antisense primer had the following sequence: 5'-CCTGCAGGCGAAACTGACTCTGGCTGAAG-3' (SEQ.ID.NO.: 96). The resulting 400 bp PCR fragment was digested with Hindlll site and subcloned into
Hindlll-Smal site of pCMV vector (5' construct). The 3' PCR sense primer used had the
following sequence: 5'-CTGTACGCTAGTGTGTTTCTACTCACGTGTCTCAGCATTGAT-3' (SEQ.ID.NO: 97) and the antisense primer had the following sequence:. 5'-GTTGGATCCACATAATGCATTTTCTC-3' (SEQ.ID.NO: 98) The resulting 880 bp PCR fragment was digested with BamHl and inserted into Pst
(blunted by T4 polymerase) and BamHl site of 5" construct to generated the full length
Nl 11 A construct. The cycle condition was 25 cycles of 94°C for 1 min, 60°C for lmin
and 72 °C for 1 min (5' PCR) or 1.5 min (3' PCR).
3. AT2K255IC3 Mutation
Preparation of a non-endogenous, constitutively activated human ATI was
accomplished by creating an AT2K255IC3 "domain swap" mutation (see, SEQ.ID.NO.:99
for nucleic acid sequence, and SEQ.ID.NO.: 100 for amino acid sequence). Restriction
sites flanking IC3 of ATI were generated to facilitate replacement of the IC3 with
corresponding IC3 from angiotensin II type 2 receptor (AT2). This was accomplished by
performing two PCR reactions. A 5' PCR fragment (Fragment A) encoded from the 5'
untranslated region to the beginning of IC3 was generated by utilizing SEQ.ID.NO.: 63 as
sense primer and the following sequence:
5'-TCCGAATTCCAAAATAACTTGTAAGAATGATCAGAAA-3' (SEQ.ID.NO.: 101) as antisense primer. A 3' PCR fragment (Fragment B) encoding from the end of IC3 to the
3' untranslated region was generated by using the following sequence: 5'-AGATCTTAAGAAGATAATTATGGCAATTGTGCT-3' (SEQ.ID.NO.: 102) as sense primer and SEQ.ID.NO.: 64 as antisense primer. The PCR condition was 30
cycles of 94°C for 1 min, 55°C for lmin and 72 °C for 1.5 min using endogenous ATI
cDNA clone as template and pfu polymerase (Stratagene), with the buffer systems
provided by the manufacturer, supplemented with 10% DMSO, 0.25 μM of each primer,
and 0.5 mM of each 4 nucleotides. Fragment A (720 bp) was digested with Hindlll and
EcoRI and subcloned. Fragment B was digested with BamHl and subcloned into pCMV
vector with an EcoRI site 5' to the cloned PCR fragment.
The DNA fragment (Fragment C) encoding IC3 of AT2 with a L255K mutation
and containing an EcoRI cohesive end at 5' and a AfHI cohesive end at 3". was generated
by annealing 2 synthetic oligonucleotides having the following sequences:
5'AATTCGAAAACACTTACTGAAGACGAATAGCTATGGGAAGAACAGGATAACCCGTGACCAA G-3' (sense; SEQ.ID.NO: 103)
5'TTAACTTGGTCACGGGTTATCCTGTTCTTCCCATAGCTATTCGTCTTCAGT AAGTGTTTTCG-3' (antisense; SEQ.ID.NO.: 104).
Fragment C was inserted in front of Fragment B through EcoRI and Aflll site. The
resulting clone was then ligated with the Fragment A through the EcoRI site to generate ATI
with AT2K255IC3.
4. A243+ Mutation
Preparation of a non-endogenous human ATI receptor was also accomplished by
creating an A243+ mutation (see, SEQ.ID.NO.: 105 for nucleic acid sequence, and
SEQ.ID.NO.: 106 for amino acid sequence). An A243+ mutation was constructed using the
following PCR based strategy: Two PCR reactions was performed using pfu polymerase
(Stratagene) with the buffer system provided by the manufacturer supplemented with 10%
DMSO, 0.25 μM of each primer, and 0.5 mM of each 4 nucleotides. The 5" PCR sense primer utilized had the following sequence:
5'-CCCAAGCTTCCCCAGGTGTATTTGAT-3' (SEQ.ID.NO.: 107) and the antisense primer had the following sequence:
5'-AAGCACAATTGCTGCATAATTATCTTAAAAATATCATC-3' (SEQ.ID.NO.: 108).
The 3 ' PCR sense primer utilized had the following sequence:
5'-AAGATAATTATGGCAGCAATTGTGCTTTTCTTTTTCTTT-3' (SEQ.ID.NO: 109)
containing the Ala insertion and antisense primer: 5'-GTTGGATCCACATAATGCATTTTCTC-3'(SEQ.ID.NO.: 110). The cycle condition was 25 cycles of 94°C for 1 min, 54°C for lmin and 72 °C for 1.5 min.
An aliquot of the 5' and 3' PCR were then used as co-template to perform secondary PCR
using the 5' PCR sense primer and 3' PCR antisense primer. The PCR condition was the
same as primary PCR except the extention time was 2.5 min. The resulting PCR fragment
was digested with Hindlll and BamHl and subcloned into pCMV vector. (See,
SEQ.ID.NO.: 105)
4. CCKB
Preparation of the non-endogenous, constitutively activated human CCKB receptor
was accomplished by creating a V322K mutation (see, SEQ.ID.NO.: I l l for nucleic acid
sequence and SEQ.ID.NO.: 112 for amino acid sequence). Mutagenesis was performed by
PCR via amplification using the wildtype CCKB from Example 1.
The first PCR fragment (lkb) was amplified by using SEQ.ID.NO.: 75 and an
antisense primer comprising a V322K mutation:
5'-CAGCAGCATGCGCTTCACGCGCTTCTTAGCCCAG-3' (SEQ.ID.NO.: 113). The second PCR fragment (0.44kb) was amplified by using a sense primer comprising the
V322K mutation: 5'-AGAAGCGCGTGAAGCGCATGCTGCTGGTGATCGTT-3' (SEQ.ID.NO: 114) and SEQ.ID.NO.:
76.
The two resulting PCR fragments were then used as template for amplifying CCKB
comprising V332K, using SEQ.ID.NO.: 75 and SEQ.ID.NO.: 76 and the above-noted
system and conditions. The resulting 1.44kb PCR fragment containing the V332K
mutation was digested with Hindlll and EcoRI and cloned into Hindlll-EcoRI site of
pCMV expression vector. (See, SEQ.ID.NO.: 111).
3. QuikChange™ Site-Directed™ Mutagenesis
Preparation of non-endogenous human GPCRs can also be accomplished by using
QuikChange™ Site-Directed™ Mutagenesis Kit (Stratagene, according to manufacturer's
instructions). Endogenous GPCR is preferably used as a template and two mutagenesis
primers utilized, as well as, most preferably, a lysine mutagenesis oligonucleotide and a
selection marker oligonucleotide (included in kit). For convenience, the codon mutation
incoφorated into the human GPCR and the respective oligonucleotides are noted, in standard
form (Table H):
TABLE H
Receptor Codon Lysine Mutagenesis Selection Marker
Identifier Mutation (SEQ.ID.NO.) (SEQ.ID.NO.) 5'-3' orientation, mutation 5'-3' orientation underlined hCHN3 S284K ATGGAGAAAAGAATCAAAAGAA TATATAGAACATTCTTTT TGTTCTATATA (115) GATTCTTTTCTCCAT
(1 16) hCHN6 L352K CGCTCTCTGGCCTTGAAGCGCAC GCTGAGCGTGCGCTTCA GCTCAGC (117) AGGCCAGAGAGCG (118) hCHN8 N235K CCCAGGAAAAAGGTGAAAGTCA GAAAACTTTGACTTTCAC AAGTTTTC (119) CTTTTTCCTGGG (120) hCHN9 G223K GGGGCGCGGGTGAAACGGCTGG GCTCACCAGCCGTTTCA TGAGC (121) CCCGCGCCCC (122) hCHNlO L231K CCCCTTGAAAAGCCTAAGAACTT GATGACCAAGTTCTTAG GGTCATC (123) GCTTTTCAAGGGG (124)
Example 3
RECEPTOR EXPRESSION
Although a variety of cells are available to the art for the expression of proteins, it is
most preferred that mammalian cells be utilized. The primary reason for this is predicated
upon practicalities, i.e., utilization of, e.g., yeast cells for the expression of a GPCR, while
possible, introduces into the protocol a non-mammalian cell which may not (indeed, in the
case of yeast, does not) include the receptor-coupling, genetic-mechanism and secretary
pathways that have evolved for mammalian systems - thus, results obtained in non-
mammalian cells, while of potential use, are not as preferred as that obtained from mammalian
cells. Of the mammalian cells, COS-7, 293 and 293T cells are particularly preferred, although
the specific mammalian cell utilized can be predicated upon the particular needs of the artisan.
On day one, 1X107 293T cells per 150mm plate were plated out. On day two, two
reaction tubes were prepared (the proportions to follow for each tube are per plate): tube A
was prepared by mixing 20μg DNA (e.g., pCMV vector; pCMV vector with receptor
cDNA, etc.) in 1.2ml serum free DMEM (Irvine Scientific. Irvine, CA); tube B was prepared by mixing 120μl lipofectamine (Gibco BRL) in 1.2ml serum free DMEM. Tubes
A and B were admixed by inversions (several times), followed by incubation at room
temperature for 30-45min. The admixture is referred to as the "transfection mixture".
Plated 293T cells were washed with 1XPBS, followed by addition of 10ml serum free
5 DMEM. 2.4ml of the transfection mixture were added to the cells, followed by incubation
for 4hrs at 37°C/5% CO,. The transfection mixture was removed by aspiration, followed
by the addition of 25ml of DMEM/10% Fetal Bovine Serum. Cells were incubated at
37°C/5% CO2. After 72hr incubation, cells were harvested and utilized for analysis.
Example 4 l o ASSAYS FOR DETERMINATION OF CONSTITUTIVE ACTIVITY OF NON-ENDOGENOUS GPCRs
A variety of approaches are available for assessment of constitutive activity of the
non-endogenous human GPCRs. The following are illustrative; those of ordinary skill in
the art are credited with the ability to determine those techniques that are preferentially
15 beneficial for the needs of the artisan.
1. Membrane Binding Assays. [35S] GTPγS Assay
When a G protein-coupled receptor is in its active state, either as a result of ligand
binding or constitutive activation, the receptor couples to a G protein and stimulates the
release of GDP and subsequent binding of GTP to the G protein. The alpha subunit of the G
20 protein-receptor complex acts as a GTPase and slowly hydrolyzes the GTP to GDP, at which
point the receptor normally is deactivated. Constitutively activated receptors continue to
exchange GDP for GTP. The non-hydrolyzable GTP analog, [35S]GTPγS. can be utilized to
demonstrate enhanced binding of [35S] GTPγS to membranes expressing constitutively
activated receptors. The advantage of using [35S] GTPγS binding to measure constitutive activation is that: (a) it is generically applicable to all G protein-coupled receptors; (b) it is
proximal at the membrane surface making it less likely to pick-up molecules which affect the
intracellular cascade.
The assay utilizes the ability of G protein coupled receptors to stimulate [35S]GTPγS
binding to membranes expressing the relevant receptors. The assay can, therefore, be used in
the direct identification method to screen candidate compounds to known, oφhan and
constitutively activated G protein-coupled receptors. The assay is generic and has application
to drug discovery at all G protein-coupled receptors.
The [35S]GTPγS assay can be incubated in 20 mM HEPES and between 1 and about
20mM MgCl2 (this amount can be adjusted for optimization of results, although 20mM is
preferred) pH 7.4, binding buffer with between about 0.3 and about 1.2 nM [35S]GTPγS (this
amount can be adjusted for optimization of results, although 1.2 is preferred ) and 12.5 to 75
μg membrane protein (e.g, COS-7 cells expressing the receptor; this amount can be adjusted
for optimization, although 75 μg is preferred) and 1 μM GDP (this amount can be changed for
optimization) for 1 hour. Wheatgerm agglutinin beads (25 μl; Amersham) should then be
added and the mixture incubated for another 30 minutes at room temperature. The tubes are
then centrifuged at 1500 x g for 5 minutes at room temperature and then counted in a
scintillation counter.
A less costly but equally applicable alternative has been identified which also meets
the needs of large scale screening. Flash plates™ and Wallac™ scintistrips may be utilized
to format a high throughput [35S]GTPγS binding assay. Furthermore, using this technique,
the assay can be utilized for known GPCRs to simultaneously monitor tritiated ligand binding
to the receptor at the same time as monitoring the efficacy via [35S]GTPγS binding. This is possible because the Wallac beta counter can switch energy windows to look at both tritium
and 35S-labeled probes. This assay may also be used to detect other types of membrane
activation events resulting in receptor activation. For example, the assay may be used to
monitor 32P phosphorylation of a variety of receptors (both G protein coupled and tyrosine
kinase receptors). When the membranes are centrifuged to the bottom of the well, the bound
[35S]GTPγS or the 32P-phosphorylated receptor will activate the scintillant which is coated of
the wells. Scinti® strips (Wallac) have been used to demonstrate this principle. In addition, the
assay also has utility for measuring ligand binding to receptors using radioactively labeled
ligands. In a similar manner, when the radiolabeled bound ligand is centrifuged to the bottom
of the well, the scintistrip label comes into proximity with the radiolabeled ligand resulting
in activation and detection.
2. Adenylyl Cyclase
A Flash Plate™ Adenylyl Cyclase kit (New England Nuclear; Cat. No. SMP004A)
designed for cell-based assays can be modified for use with crude plasma membranes. The
Flash Plate wells contain a scintillant coating which also contains a specific antibody-
recognizing cAMP. The cAMP generated in the wells was quantitated by a direct
competition for binding of radioactive cAMP tracer to the cAMP antibody. The following
serves as a brief protocol for the measurement of changes in cAMP levels in membranes that
express the receptors.
Transfected cells are harvested approximately three days after transfection.
Membranes were prepared by homogenization of suspended cells in buffer containing 20mM
HEPES, pH 7.4 and lOmM MgCl2. Homogenization is performed on ice using a Brinkman
Polytron™ for approximately 10 seconds. The resulting homogenate is centrifuged at 49.000 X g for 15 minutes at 4°C. The resulting pellet is then resuspended in buffer containing
20mM HEPES, pH 7.4 and 0.1 mM EDTA, homogenized for 10 seconds, followed by
centrifugation at 49,000 X g for 15 minutes at 4°C. The resulting pellet can be stored at -
80 °C until utilized. On the day of measurement, the membrane pellet is slowly thawed at
room temperature, resuspended in buffer containing 20mM HEPES, pH 7.4 and lOmM
MgCL2 (these amounts can be optimized, although the values listed herein are preferred), to
yield a final protein concentration of 0.60mg/ml (the resuspended membranes were placed
on ice until use).
cAMP standards and Detection Buffer (comprising 2 μCi of tracer [125I cAMP (100
l] to 11 ml Detection Buffer) are prepared and maintained in accordance with the
manufacturer's instructions. Assay Buffer is prepared fresh for screening and contained
20mM HEPES, pH 7.4, lOmM MgCl2, 20mM (Sigma), 0.1 units/ml creatine phosphokinase
(Sigma), 50 μM GTP (Sigma), and 0.2 mM ATP (Sigma); Assay Buffer can be stored on ice
until utilized. The assay is initiated by addition of 50ul of assay buffer followed by addition
of 50ul of membrane suspension to the NEN Flash Plate. The resultant assay mixture is
incubated for 60 minutes at room temperature followed by addition of 1 OOul of detection
buffer. Plates are then incubated an additional 2-4 hours followed by counting in a Wallac
MicroBeta™ scintillation counter. Values of cAMP/well are extrapolated from a standard
cAMP curve that is contained within each assay plate.
C. Reporter-Based Assays
1. CREB Reporter Assay (Gs-associated receptors)
A method to detect Gs stimulation depends on the known property of the transcription
factor CREB, which is activated in a cAMP-dependent manner. A PathDetect™ CREB trans- Reporting System (Stratagene, Catalogue # 219010) can utilized to assay for Gs coupled
activity in 293 or 293T cells. Cells are transfected with the plasmids components of this
above system and the indicated expression plasmid encoding endogenous or mutant receptor
using a Mammalian Transfection Kit (Stratagene, Catalogue #200285) according to the
manufacturer' s instructions. Briefly, 400 ng pFR-Luc (luciferase reporter plasmid containing
Gal4 recognition sequences), 40 ng pFA2-CREB (Gal4-CREB fusion protein containing the
Gal4 DNA-binding domain), 80 ng pCMV -receptor expression plasmid (comprising the
receptor) and 20 ng CMV-SEAP (secreted alkaline phosphatase expression plasmid; alkaline
phosphatase activity is measured in the media of transfected cells to control for variations in
transfection efficiency between samples) are combined in a calcium phosphate precipitate as
per the Kit's instructions. Half of the precipitate is equally distributed over 3 wells in a 96-
well plate, kept on the cells overnight, and replaced with fresh medium the following morning.
Forty-eight (48) hr after the start of the transfection, cells are treated and assayed for, e.g.,
luciferase activity
2. API reporter assay (Gq-associated receptors)
A method to detect Gq stimulation depends on the known property of Gq-dependent
phospholipase C to cause the activation of genes containing API elements in their promoter.
A Pathdetect™ AP-1 cis-Reporting System (Stratagene, Catalogue # 219073) can be utilized
following the protocol set forth above with respect to the CREB reporter assay, except that
the components of the calcium phosphate precipitate were 410 ng pAP 1 -Luc. 80 ng pCMV-
receptor expression plasmid, and 20 ng CMV-SEAP.
3. CRE-LUC Reporter Assay
293 and 293T cells are plated-out on 96 well plates at a density of 2 x 104 cells per well and were transfected using Lipofectamine Reagent (BRL) the following day according
to manufacturer instructions. A DNA/lipid mixture is prepared for each 6-well transfection
as follows: 260ng of plasmid DNA in lOOμl of DMEM were gently mixed with 2μl of lipid
in lOOμl of DMEM (the 260ng of plasmid DNA consisted of 200ng of a 8xCRE-Luc reporter
plasmid (.see below and Figure 1 for a representation of a portion of the plasmid), 50ng of
pCMV comprising endogenous receptor or non-endogenous receptor or pCMV alone, and
lOng of a GPRS expression plasmid (GPRS in pcDNA3 (Invitrogen)). The 8XCRE-Luc
reporter plasmid was prepared as follows: vector SRIF-β-gal was obtained by cloning the rat
somatostatin promoter (-71/+51) at BglV-Hindlll site in the pβgal-Basic Vector (Clontech).
Eight (8) copies of cAMP response element were obtained by PCR from an adenovirus
template AdpCF126CCRE8 (see, 1 Human Gene Therapy 1883 (1996)) and cloned into the
SRIF-β-gal vector at the Kpn-BglV site, resulting in the 8xCRE-β-gal reporter vector. The
8xCRE-Luc reporter plasmid was generated by replacing the beta-galactosidase gene in the
8xCRE-β-gal reporter vector with the luciferase gene obtained from the pGL3 -basic vector
(Promega) at the Hindlll-BamHI site. Following 30 min. incubation at room temperature, the
DNA/lipid mixture was diluted with 400 μl of DMEM and lOOμl of the diluted mixture was
added to each well. 100 μl of DMEM with 10% FCS were added to each well after a 4hr
incubation in a cell culture incubator. The following day the transfected cells were changed
with 200 μl/well of DMEM with 10% FCS. Eight (8) hours later, the wells were changed to
100 μl /well of DMEM without phenol red, after one wash with PBS. Luciferase activity were
measured the next day using the LucLite™ reporter gene assay kit (Packard) following
manufacturer instructions and read on a 1450 MicroBeta™ scintillation and luminescence
counter (Wallac). 4. SRF-LUC Reporter Assay
One method to detect Gq stimulation depends on the known property of Gq-dependent
phospholipase C to cause the activation of genes containing serum response factors in their
promoter. A Pathdetect™ SRF-Luc-Reporting System (Stratagene) can be utilized to assay
for Gq coupled activity in, e.g., COS7 cells. Cells are transfected with the plasmid
components of the system and the indicated expression plasmid encoding endogenous or non-
endogenous GPCR using a Mammalian Transfection™ Kit (Stratagene, Catalogue #200285)
according to the manufacturer' s instructions. Briefly, 410 ng SRF-Luc.80 ng pCMV -receptor
expression plasmid and 20 ng CMV-SEAP (secreted alkaline phosphatase expression plasmid;
alkaline phosphatase activity is measured in the media of transfected cells to control for
variations in transfection efficiency between samples) are combined in a calcium phosphate
precipitate as per the manufacturer' s instructions. Half of the precipitate is equally distributed
over 3 wells in a 96-well plate, kept on the cells in a serum free media for 24 hours. The last
5 hours the cells are incubated with 1 μM Angiotensin, where indicated. Cells are then lysed
and assayed for luciferase activity using a Luclite™ Kit (Packard, Cat. # 601691 1) and "Trilux
1450 Microbeta" liquid scintillation and luminescence counter (Wallac) as per the
manufacturer's instructions. The data can be analyzed using GraphPad Prism™ 2.0a
(GraphPad Software Inc.).
5. Intracellular IP3 Accumulation Assay
On day 1 , cells comprising the receptors (endogenous and/or non-endogenous) can
be plated onto 24 well plates, usually IxlO5 cells/well (although his umber can be
optimized. On day 2 cells can be transfected by firstly mixing 0.25ug DNA in 50 ul serum
free DMEM/well and 2 ul lipofectamine in 50 μl serumfree DMEM/well. The solutions are gently mixed and incubated for 15-30 min at room temperature. Cells are washed with
0.5 ml PBS and 400 μl of serum free media is mixed with the transfection media and
added to the cells. The cells are then incubated for 3-4 hrs at 37°C/5%CO2 and then the
transfection media is removed and replaced with 1 ml/well of regular growth media. On
day 3 the cells are labeled with 3H-myo-inositol. Briefly, the media is removed and the
cells are washed with 0.5 ml PBS. Then 0.5 ml inositol-free/serum free media (GIBCO
BRL) is added/well with 0.25 μCi of 3H-myo-inositol / well and the cells are incubated for
16-18 hrs o/n at 37°C/5%CO2. On Day 4 the cells are washed with 0.5 ml PBS and 0.45
ml of assay medium is added containing inositol-free/serum free media 10 μM pargyline
10 mM lithium chloride or 0.4 ml of assay medium and 50 ul of lOx ketanserin (ket) to
final concentration of lOμM. The cells are then incubated for 30 min at 37 °C. The cells
are then washed with 0.5 ml PBSand 200 ul of fresh/icecold stop solution (1M KOH; 18
mM Na-borate; 3.8 mM EDTA) is added/well. The solution is kept on ice for 5-10 min or
until cells were lysed and then neutralized by 200 μl of fresh/ice cold neutralization sol.
(7.5 % HCL). The lysate is then transferred into 1.5 ml eppendorf tubes and 1 ml of
chloroform/methanol (1 :2) is added/tube. The solution is vortexed for 15 sec and the
upper phase is applied to a Biorad AG1-X8™ anion exchange resin (100-200 mesh).
Firstly, the resin is washed with water at 1 : 1.25 W/V and 0.9 ml of upper phase is loaded
onto the column. The column is washed with 10 mis of 5 mM myo-inositol and 10 ml of 5
mM Na-borate/60mM Na-formate. The inositol tris phosphates are eluted into scintillation
vials containing 10 ml of scintillation cocktail with 2 ml of 0.1 M formic acid/ 1 M
ammonium formate. The columns are regenerated by washing with 10 ml of 0.1 M formic
acid/3M ammonium formate and rinsed twice with dd H,O and stored at 4°C in water. Exemplary results are presented below in Table I:
TABLE I
Receptor Mutation Assay Signal Signal Percent
Utilized Generated: Generated: Difference
Endogenous Non-
Version Endogenous
(Relative Version
Light Units) (Relative Light Units) hATl F239K SRF-LUC 34 137 75% t
AT2K255IC3 SRF-LUC 34 127 73% 1
hTDAG8 I225K CRE-LUC 2,715 14,440 81%1 (293 cells)
I225K CRE-LUC 65,681 185,636 65% t (293 T cells) hH9 F236K CRE-LUC 1,887 6,096 69% 1 hCCKB V332K CRE-LUC 785 3,223 76% t
C. CELL-BASED DETECTION ASSAY (EXAMPLE -TDAG8)
293 cells were plated-out on 150mm plates at a density of 1.3 x 107 cells per plate, and
were transfected using 12ug of the respective DNA and 60ul of Lipofectamine Reagent
(BRL) per plate. The transfected cells were grown in media containing serum for an assay
performed 24 hours post-transfection. For detection assay performed 48 hours post-
transfection (assay comparing serum and serum-free media; see Figure 3). the initial media
was changed to either serum or serum-free media. The serum-free media was comprised solely
of Dulbecco's Modified Eagle's (DME) High Glucose Medium (Irvine Scientific #9024). In
addition to the above DME Medium, the media with serum contained the following: 10%
Fetal Bovine Serum (Hyclone #SH30071.03), 1% of lOOmM Sodium Pyruvate (Irvine
Scientific #9334). 1 % of 20mM L-Glutamine (Irvine Scientific #9317), and 1 % of Penicillin- Streptomycin solution (Irvine Scientific #9366).
A 96-well Adenylyl Cyclase Activation Flashplate™ was used (NEN: #SMP004A).
First, 50ul of the standards for the assay were added to the plate, in duplicate, ranging from
concentrations of 50pmol to zero pmol cAMP per well. The standard cAMP (NEN:
#SMP004A) was reconstituted in water, and serial dilutions were made using lxPBS (Irvine
Scientific: #9240). Next, 50ul of the stimulation buffer (NEN: #SMP004A) was added to all
wells. In the case of using compounds to measure activation or inactivation of cAMP, lOul
of each compound, diluted in water, was added to its respective well, in triplicate. Various
final concentrations used range from luM up to ImM. Adenosine 5'-triphosphate, ATP,
(Research Biochemicals International: #A- 141) and Adenosine 5'-diphosphate, ADP, (Sigma:
#A2754) were used in the assay. Next, the 293 cells transfected with the respective cDNA
(CMV or TDAG8) were harvested 24 (assay detection in serum media) or 48 hours post-
transfection (assay detection comparing serum and serum-free media). The media was
aspirated and the cells washed once with lxPBS. Then 5ml of lxPBS was added to the cells
along with 3ml of cell dissociation buffer (Sigma: #C-1544). The detached cells were
transferred to a centrifuge tube and centrifuged at room temperature for five minutes. The
supernatant was removed and the cell pellet was resuspended in an appropriate amount of
lxPBS to obtain a final concentration of 2x106 cells per milliliter. To the wells containing the
compound, 50ul of the cells in lxPBS (lxl 05 cells/well) were added. The plate was incubated
on a shaker for 15 minutes at room temperature. The detection buffer containing the tracer
cAMP was prepared. In 11ml of detection buffer (NEN: #SMP004A). 50ul (equal to luCi)
of [125I]cAMP (NEN: #SMP004A) was added. Following incubation, 50ul of this detection
buffer containing tracer cAMP was added to each well. The plate was placed on a shaker and incubated at room temperature for two hours. Finally, the solution from the wells of the plate
were aspirated and the flashplate was counted using the Wallac MicroBeta™ scintillation
counter.
In Figure 2A, ATP and ADP bind to endogenous TDAG8 resulting in an increase
of c AMP of about 59% and about 55% respectively. Figure 2B evidences ATP and ADP
binding to endogenous TDAG8 where endogenous TDAG8 was transfected and grown in
serum and serum-free medium. ATP binding to endogenous TDAG8 grown in serum
media evidences an increase in cAMP of about 65%, compared to the endogenous TDAG8
with no compounds; in serum-free media there was an increase of about 68%. ADP
binding to endogenous TDAG8 in serum evidences about a 61% increase, while in serum-
free ADP binding evidences an increase of about 62% increase. ATP and ADP bind to
endogenous TDAG8 with an EC50 value of 139.8uM and 120.5uM, respectively (data not
shown).
Although the results presented in Figure 2B indicate substantially the same results
when serum and serum-free media were compared, our choice is to use a serum based
media, although a serum-free media can also be utilized.
Example 6
GPCR FUSION PROTEIN PREPARATION
The design of the constitutively activated GPCR-G protein fusion construct was
accomplished as follows: both the 5' and 3' ends of the rat G protein Gsα (long form; Itoh.
H. et al., 83 PNAS 3776 (1986)) were engineered to include a Hindlll (5"-AAGCTT-3')
sequence thereon. Following confirmation of the correct sequence (including the flanking
Hindlll sequences), the entire sequence was shuttled into pcDNA3.1(-) (Invitrogen, cat. no.
V795-20) by subcloning using the Hindlll restriction site of that vector. The correct orientation for the Gsα sequence was determined after subcloning into pcDNA3.1(-). The
modified pcDNA3.1 (-) containing the rat Gsα gene at Hindlll sequence was then verified; this
vector was now available as a "universal" Gsα protein vector. The pcDNA3.1(-) vector
contains a variety of well-known restriction sites upstream of the Hindlll site, thus
beneficially providing the ability to insert, upstream of the Gs protein, the coding sequence
of an endogenous, constitutively active GPCR. This same approach can be utilized to create
other "universal" G protein vectors, and, of course, other commercially available or
proprietary vectors known to the artisan can be utilized - the important criteria is that the
sequence for the GPCR be upstream and in-frame with that of the G protein.
TDAG8 couples via Gs, while H9 couples via Gz. For the following exemplary GPCR
Fusion Proteins, fusion to Gsα was accomplished.
A TDAG8(I225K)-Gsα Fusion Protein construct was made as follows: primers were
designed as follows:
5'-gatcTCTAGAATGAACAGCACATGTATTGAAG-3' (SEQ.ID.NO.: 125; sense) 5'-ctagGGTACCCGCTCAAGGACCTCTAATTCCATAG-3' (SEQ.ID.NO.: 126; antisense).
Nucleotides in lower caps are included as spacers in the restriction sites between the
G protein and TDAG8. The sense and anti-sense primers included the restriction sites for
Xbal and Kpnl, respectively.
PCR was then utilized to secure the respective receptor sequences for fusion within
the Gsα universal vector disclosed above, using the following protocol for each: 1 OOng cDNA
for TDAG8 was added to separate tubes containing 2ul of each primer (sense and anti-sense),
3uL of lOmM dNTPs, lOuL of 1 OXTaqPlus™ Precision buffer, luL of TaqPlus™ Precision
polymerase (Stratagene: #600211), and 80uL of water. Reaction temperatures and cycle times
for TDAG8 were as follows: the initial denaturing step was done it 94 °C for five minutes, and a cycle of 94°C for 30 seconds; 55 °C for 30 seconds; 72°C for two minutes. A final
extension time was done at 72 ° C for ten minutes. PCR product for was run on a 1 % agarose
gel and then purified (data not shown). The purified product was digested with Xbal and
Kpnl (New England Biolabs) and the desired inserts purified and ligated into the Gs universal
vector at the respective restriction site. The positive clones was isolated following
transformation and determined by restriction enzyme digest; expression using 293 cells was
accomplished following the protocol set forth infra. Each positive clone for TDAG8:Gs -
Fusion Protein was sequenced to verify correctness.
GPCR Fusion Proteins comprising non-endogenous, constitutively activated
TDAG8(I225K) were analyzed as above and verified for constitutive activation.
An H9(F236K)-Gsα Fusion Protein construct was made as follows: primers were
designed as follows:
5'-TTAgatatcGGGGCCCACCCTAGCGGT-3' (SEQ.ID.NO.: 145; sense) 5'-ggtaccCCCACAGCCATTTCATCAGGATC-3' (SEQ.ID.NO.: 146; antisense). Nucleotides in lower caps are included as spacers in the restriction sites between the
G protein and H9. The sense and anti-sense primers included the restriction sites for EcoRV
and Kpnl, respectively such that spacers (attributed to the restriction sites) exists between the
G protein and H9.
PCR was then utilized to secure the respective receptor sequences for fusion within
the Gsα universal vector disclosed above, using the following protocol for each: 80ng cDNA
for H9 was added to separate tubes containing lOOng of each primer (sense and anti-sense).
and 45uL of PCR Supermix™ (Gibco-Brl, LifeTech) (50ul total reaction volume). Reaction
temperatures and cycle times for H9 were as follows: the initial denaturing step was done it
94°C for one, and a cycle of 94°C for 30 seconds: 55 °C for 30 seconds: 72°C for two minutes. A final extension time was done at 72 °C for seven minutes. PCR product for was
run on a 1 % agarose gel and then purified (data not shown). The purified product was cloned
into pCRII-TOPO™ System followed by identification of positive clones. Positive clones
were isolated, digested with EcoRV and Kpnl (New England Biolabs) and the desired inserts
were isolated, purified and ligated into the Gs universal vector at the respective restriction site .
The positive clones was isolated following transformation and determined by restriction
enzyme digest; expression using 293 cells was accomplished following the protocol set forth
infra. Each positive clone for H9(F236K):Gs - Fusion Protein was sequenced to verify
correctness. Membranes were frozen (-80 °C) until utilized.
To ascertain the ability of measuring a cAMP response mediated by the Gs protein
(even though H9 couples with Gz), the following cAMP membrane assay was utilized, based
upon an NEN Adenyl Cyclase Activation Flahplate™ Assay kit (96 well format). "Binding
Buffer" consisted of 1 OmM HEPES, 1 OOmM NaCl and 1 OmM MgCl (ph 7.4). "Regeneration
Buffer" was prepared in Binding Buffer and consisted of 20mM phosphocreatine, 20U
creatine phosphokinase, 20uM GTP, 0.2mM ATP, and 0.6mM IBMX. "cAMP Standards"
were prepared in Binding Buffer as follows: cAMP Stock Added to Final Assay Concentration
(5,000 pmol ml in 2ml H20) indicted amount of Binding (50ul into lOOul) in ul Buffer to achieve indicated pmol/well
A 250 1ml 50
B 500 of A 500ul 25
C 500 of B 500ul 12.5
D 500 of C 750ul 5.0
E 500 of D 500ul 2.5
F 500 of E 500ul 1.25
G 500 of F 750ul 0.5
Frozen membranes (both pCMV as control and the non-endogenous H(-Gs Fusion
Protein) were thawed (on ice at room temperature until in solution). Membranes were homogenized with a polytron until in suspension (2 x 15 seconds). Membrane protein
concentration was determined using the Bradford Assay Protocol (see infra). Membrane
concentration was diluted to 0.5mg/ml in Regeneration Buffer (final assay concentration -
25ug/well). Thereafter, 50ul of Binding Buffer was added to each well. For control, 50ul/well
of cAMP standard was added to wells 11 and 12 A-G, with Binding Buffer alone to 12H (on
the 96-well format). Thereafter, 50ul/well of protein was added to the wells and incubated at
room temperature (on shaker) for 60min. 100ul[125I]cAMP in Detection Buffer (see infra) was
added to each well (final - 50ul[125I]cAMP into 11ml Detection Buffer). These were
incubated for 2hrs at room temperature. Plates were aspirated with an 8 channel manifold and
sealed with plate covers. Results (pmoles cAMP bound) were read in a Wallac™ 1450 on
"prot #15). Results are presented in Figure 3.
The results presented in Figure 3 indicate that the Gs coupled fusion was able to
"drive" the cyclase reaction such that measurement of the consitutive activation of H9(F236K)
was viable. Based upon these results, the direct identification of candidate compounds that
are inverse agonists, agonists and partial agonists is possible using a cyclase-based assay.
Example 6
Protocol: Direct Identification of Inverse Agonists and Agonists Using [35S]GTPγS
Although we have utilized endogenous, constitutively active GPCRs for the direct
identification of candidate compounds as, e.g., inverse agonists, for reasons that are not
altogether understood, intra-assay variation can become exacerbated. Preferably, then, a
GPCR Fusion Protein, as disclosed above, is also utilized with a non-endogenous,
constitutively activated GPCR. We have determined that when such a protein is used, intra-
assay variation appears to be substantially stabilized, whereby an effective signal-to-noise
ratio is obtained. This has the beneficial result of allowing for a more robust identification of candidate compounds. Thus, it is preferred that for direct identification, a GPCR Fusion
Protein be used and that when utilized, the following assay protocols be utilized.
Membrane Preparation
Membranes comprising the non-endogenous, constitutively active oφhan GPCR
Fusion Protein of interest and for use in the direct identification of candidate compounds as
inverse agonists, agonists or partial agonists are preferably prepared as follows:
a. Materials
"Membrane Scrape Buffer" is comprised of 20mM HEPES and 1 OmM EDTA, pH 7.4;
"Membrane Wash Buffer" is comprised of 20 mM HEPES and 0.1 mM EDTA, pH 7.4;
"Binding Buffer" is comprised of 20mM HEPES, 100 mM NaCl, and 10 mM MgCl2, pH 7.4
b. Procedure
All materials are kept on ice throughout the procedure. Firstly, the media is aspirated
from a confluent monolayer of cells, followed by rinse with 10ml cold PBS, followed by
aspiration. Thereafter, 5ml of Membrane Scrape Buffer is added to scrape cells; this is
followed by transfer of cellular extract into 50ml centrifuge tubes (centrifuged at 20,000 φm
for 17 minutes at 4°C). Thereafter, the supernatant is aspirated and the pellet is resuspended
in 30ml Membrane Wash Buffer followed by centrifuge at 20,000 φm for 17 minutes at 4 ° C .
The supernatant is then aspirated and the pellet resuspended in Binding Buffer. This is then
homogenized using a Brinkman polytron™ homogenizer (15-20 second bursts until the all
material is in suspension). This is referred to herein as "Membrane Protein".
Bradford Protein Assay
Following the homogenization, protein concentration of the membranes is determined
using the Bradford Protein Assay (protein can be diluted to about 1.5mg/ml, aliquoted and frozen (-80°C) for later use; when frozen, protocol for use is as follows: on the day of the
assay, frozen Membrane Protein is thawed at room temperature, followed by vortex and then
homogenized with a polytron at about 12 x 1 ,000 φm for about 5-10 seconds; it is noted that
for multiple preparations, the homogenizor should be thoroughly cleaned between
homoginezation of different preparations).
a. Materials
Binding Buffer (as per above); Bradford Dye Reagent; Bradford Protein Standard are
utilized, following manufacturer instructions (Biorad. cat. no. 500-0006).
b. Procedure
Duplicate tubes are prepared, one including the membrane, and one as a control
"blank". Each contained 800ul Binding Buffer. Thereafter, lOul of Bradford Protein Standard
(lmg/ml) is added to each tube, and lOul of membrane Protein is then added to just one tube
(not the blank). Thereafter, 200ul of Bradford Dye Reagent is added to each tube, followed
by vortex of each. After five (5) minutes, the tubes were re-vortexed and the material therein
is transferred to cuvettes. The cuvettes are then read using a CECIL 3041 spectrophotometer,
at wavelength 595.
Direct Identification Assay
a. Materials
GDP Buffer consists of 37.5 ml Binding Buffer and 2mg GDP (Sigma, cat. no. G-
7127), followed by a series of dilutions in Binding Buffer to obtain 0.2 uM GDP (final
concentration of GDP in each well was 0.1 uM GDP); each well comprising a candidate
compound, has a final volume of 200ul consisting of lOOul GDP Buffer (final concentration,
O.luM GDP), 50ul Membrane Protein in Binding Buffer, and 50ul [35S]GTPγS (0.6 nM) in Binding Buffer (2.5 ul [35S]GTPγS per 10ml Binding Buffer).
b. Procedure
Candidate compounds are preferably screened using a 96-well plate format (these can
be frozen at -80 °C). Membrane Protein (or membranes with expression vector excluding the
GPCR Fusion Protein, as control), are homogenized briefly until in suspension. Protein
concentration is then determined using the Bradford Protein Assay set forth above. Membrane
Protein (and control) is then diluted to 0.25mg/ml in Binding Buffer (final assay
concentration, 12.5ug/well). Thereafter, 100 ul GDP Buffer is added to each well of a Wallac
Scintistrip™ (Wallac). A 5ul pin-tool is then used to transfer 5 ul of a candidate compound
into such well (i.e., 5ul in total assay volume of 200 ul is a 1 :40 ratio such that the final
screening concentration of the candidate compound is 1 OuM). Again, to avoid contamination,
after each transfer step the pin tool should be rinsed in three reservoirs comprising water (IX),
ethanol (IX) and water (2X) - excess liquid should be shaken from the tool after each rinse
and dried with paper and kimwipes. Thereafter, 50 ul of Membrane Protein is added to each
well (a control well comprising membranes without the GPCR Fusion Protein is also utilized),
and re-incubatedfor 5-10 minutes at room temperature. Thereafter, 50 ul of [35S]GTPγS (0.6
nM) in Binding Buffer is added to each well, followed by incubation on a shaker for 60
minutes at room temperature (again, in this example, plates were covered with foil). The
assay is then stopped by spinning of the plates at 4000 RPM for 15 minutes at 22 °C. The
plates are then aspirated with an 8 channel manifold and sealed with plate covers. The plates
are then read on a Wallace 1450 using setting "Prof #37" (as per manufacturer instructions).
Example 7
Protocol: Confirmation Assay
Using an independent assay approach to provide confirmation of a directly identified candidate compound as set forth above, it is preferred that a confirmation assay then be
utilized. In this case, the preferred confirmation assay is a cyclase-based assay.
A modified Flash Plate™ Adenylyl Cyclase kit (New England Nuclear; Cat. No.
SMP004A) is preferably utilized for confirmation of candidate compounds directly identified
as inverse agonists and agonists to non-endogenous, constitutively activated oφhan GPCRs
in accordance with the following protocol.
Transfected cells are harvested approximately three days after transfection.
Membranes are prepared by homogenization of suspended cells in buffer containing 20mM
HEPES, pH 7.4 and lOmM MgCl2. Homogenization is performed on ice using a Brinkman
Polytron™ for approximately 10 seconds. The resulting homogenate is centrifuged at 49,000
X g for 15 minutes at 4°C. The resulting pellet is then resuspended in buffer containing
20mM HEPES, pH 7.4 and 0.1 mM EDTA, homogenized for 10 seconds, followed by
centrifugation at 49,000 X g for 15 minutes at 4°C. The resulting pellet can be stored at -
80 °C until utilized. On the day of direct identification screening, the membrane pellet is
slowly thawed at room temperature, resuspended in buffer containing 20mM HEPES, pH 7.4
and lOmM MgCL2, to yield a final protein concentration of 0.60mg/ml (the resuspended
membranes are placed on ice until use).
cAMP standards and Detection Buffer (comprising 2 μCi of tracer [125I cAMP (100
μl] to 11 ml Detection Buffer) are prepared and maintained in accordance with the
manufacturer's instructions. Assay Buffer is prepared fresh for screening and contained
20mM HEPES, pH 7.4, lOmM MgCl2, 20mM phospocreatine (Sigma), 0.1 units/ml creatine
phosphokinase (Sigma), 50 μM GTP (Sigma), and 0.2 mM ATP (Sigma); Assay Buffer can
be stored on ice until utilized. Candidate compounds identified as per above (if frozen, thawed at room temperature)
are added, preferably, to 96-well plate wells (3μl/well; 12μM final assay concentration),
together with 40 μl Membrane Protein (30μg/well) and 50μl of Assay Buffer. This admixture
is then incubated for 30 minutes at room temperature, with gentle shaking.
Following the incubation, lOOμl of Detection Buffer is added to each well, followed
by incubation for 2-24 hours. Plates are then counted in a Wallac MicroBeta™ plate reader
using "Prot. #31 " (as per manufacturer instructions).
It is intended that each of the patents, applications, and printed publications mentioned
in this patent document be hereby incoφorated by reference in their entirety.
As those skilled in the art will appreciate, numerous changes and modifications may
be made to the preferred embodiments of the invention without departing from the spirit of
the invention. It is intended that all such variations fall within the scope of the invention.
Although a variety of expression vectors are available to those in the art, for
puφoses of utilization for both the endogenous and non-endogenous human GPCRs, it is
most preferred that the vector utilized be pCMV. This vector was deposited with the
American Type Culture Collection (ATCC) on October 13, 1998 (10801 University Blvd.,
Manassas, VA 20110-2209 USA) under the provisions of the Budapest Treaty for the
International Recognition of the Deposit of Microorganisms for the Puφose of Patent
Procedure. The DNA was tested by the ATCC and determined to be. The ATCC has
assigned the following deposit number to pCMV: ATCC #203351.

Claims

What is claimed is:
I . A cDNA encoding a non-endogenous, constitutively activated version of a human
G protein-coupled receptor comprising hARE-3(F313K).
2. A non-endogenous version of a human G protein-coupled receptor encoded by the
cDNA of claim 1.
3. A Plasmid comprising a Vector and the cDNA of claim 1.
4. A Host Cell comprising the Plasmid of claim 3.
5. A cDNA encoding a non-endogenous, constitutively activated version of a human
G protein-coupled receptor comprising hARE-4(V233K)
6. A non-endogenous version of a human G protein-coupled receptor encoded by the
cDNA of claim 5.
7. A Plasmid comprising a Vector and the cDNA of claim 5.
8. A Host Cell comprising the Plasmid of claim 7.
9. A cDNA encoding a non-endogenous, constitutively activated version of a human
G protein-coupled receptor comprising hARE-5(A240K).
10. A non-endogenous version of a human G protein-coupled receptor encoded by the
cDNA of claim 9.
I I. A Plasmid comprising a Vector and the cDNA of claim 5.
12. A Host Cell comprising the Plasmid of claim 11.
13. A cDNA encoding a non-endogenous, constitutively activated version of a human
G protein-coupled receptor comprising hGPCR14(L257K).
14. A non-endogenous version of a human G protein-coupled receptor encoded by the
cDNA of claim 13.
15. A Plasmid comprising a Vector and the cDNA of claim 13.
16. A Host Cell comprising the Plasmid of claim 15.
17. A cDNA encoding a non-endogenous, constitutively activated version of a human
G protein-coupled receptor comprising hGPCR27(C283K).
18. A non-endogenous version of a human G protein-coupled receptor encoded by the
cDNA of claim 17.
19. A Plasmid comprising a Vector and the cDNA of claim 17.
20. A Host Cell comprising the Plasmid of claim 19.
21. A cDNA encoding a non-endogenous, constitutively activated version of a human
G protein-coupled receptor comprising hARE-l(E232K).
22. A non-endogenous version of a human G protein-coupled receptor encoded by the
cDNA of claim 21.
23. A Plasmid comprising a Vector and the cDNA of claim 21.
24. A Host Cell comprising the Plasmid of claim 23.
25. A cDNA encoding a non-endogenous, constitutively activated version of a human
G protein-coupled receptor comprising hARE-2(G285K).
26. A non-endogenous version of a human G protein-coupled receptor encoded by the
cDNA of claim 25.
27. A Plasmid comprising a Vector and the cDNA of claim 25.
28. A Host Cell comprising the Plasmid of claim 27.
29. A cDNA encoding a non-endogenous, constitutively activated version of a human
G protein-coupled receptor comprising hPPRl(L239K).
30. A non-endogenous version of a human G protein-coupled receptor encoded by the
cDNA of claim 29.
31. A Plasmid comprising a Vector and the cDNA of claim 29.
32. A Host Cell comprising the Plasmid of claim 31.
33. A cDNA encoding a non-endogenous, constitutively activated version of a human
G protein-coupled receptor comprising hG2A(K232A).
34. A non-endogenous version of a human G protein-coupled receptor encoded by the
cDNA of claim 33.
35. A Plasmid comprising a Vector and the cDNA of claim 33.
36. A Host Cell comprising the Plasmid of claim 35.
37. A cDNA encoding a non-endogenous, constitutively activated version of a human
G protein-coupled receptor comprising hRUP3(L224K).
38. A non-endogenous version of a human G protein-coupled receptor encoded by the
cDNA of claim 37.
39. A Plasmid comprising a Vector and the cDNA of claim 37.
40. A Host Cell comprising the Plasmid of claim 39.
41. A cDNA encoding a non-endogenous, constitutively activated version of a human
G protein-coupled receptor comprising hRUP5(A236K).
42. A non-endogenous version of a human G protein-coupled receptor encoded by the
cDNA of claim 41.
43. A Plasmid comprising a Vector and the cDNA of claim 41.
44. A Host Cell comprising the Plasmid of claim 42.
45. A cDNA encoding a non-endogenous, constitutively activated version of a human
G protein-coupled receptor comprising hRUP6(N267K)
46. A non-endogenous version of a human G protein-coupled receptor encoded by the
cDNA of claim 45.
47. A Plasmid comprising a Vector and the cDNA of claim 45.
48. A Host Cell comprising the Plasmid of claim 47.
49. A cDNA encoding a non-endogenous, constitutively activated version of a human
G protein-coupled receptor comprising hRUP7(A302K).
50. A non-endogenous version of a human G protein-coupled receptor encoded by the
cDNA of claim 49.
51. A Plasmid comprising a Vector and the cDNA of claim 49.
52. A Host Cell comprising the Plasmid of claim 51.
53. A cDNA encoding a non-endogenous, constitutively activated version of a human
G protein-coupled receptor comprising hCHN4(V236K).
54. A non-endogenous version of a human G protein-coupled receptor encoded by the
cDNA of claim 53.
55. A Plasmid comprising a Vector and the cDNA of claim 53.
56. A Host Cell comprising the Plasmid of claim 55.
57. A cDNA encoding a non-endogenous, constitutively activated version of a human
G protein-coupled receptor comprising hMC4(A244K).
58. A non-endogenous version of a human G protein-coupled receptor encoded by the
cDNA of claim 57.
59. A Plasmid comprising a Vector and the cDNA of claim 57.
60. A Host Cell comprising the Plasmid of claim 60.
61. A cDNA encoding a non-endogenous, constitutively activated version of a human
G protein-coupled receptor comprising hCHN3(S284K).
62. A non-endogenous version of a human G protein-coupled receptor encoded by the
cDNA of claim 61.
63. A Plasmid comprising a Vector and the cDNA of claim 61.
64. A Host Cell comprising the Plasmid of claim 63.
65. A cDNA encoding a non-endogenous, constitutively activated version of a human
G protein-coupled receptor comprising hCHN6(L352K) .
66. A non-endogenous version of a human G protein-coupled receptor encoded by the
cDNA of claim 65.
67. A Plasmid comprising a Vector and the cDNA of claim 65.
68. A Host Cell comprising the Plasmid of claim 67.
69. A cDNA encoding a non-endogenous, constitutively activated version of a human
G protein-coupled receptor comprising hCHN8(N235K).
70. A non-endogenous version of a human G protein-coupled receptor encoded by the
cDNA of claim 69.
71. A Plasmid comprising a Vector and the cDNA of claim 69.
72. A Host Cell comprising the Plasmid of claim 71.
73. A cDNA encoding a non-endogenous, constitutively activated version of a human
G protein-coupled receptor comprising hH9(F236K).
74. A non-endogenous version of a human G protein-coupled receptor encoded by the cDNA of claim 73.
75. A Plasmid comprising a Vector and the cDNA of claim 73.
76. A Host Cell comprising the Plasmid of claim 74.
77. A cDNA encoding a non-endogenous, constitutively activated version of a human
G protein-coupled ATI receptor selected from the group consisting of:
hATl(F239K); hATl(Nl l lA); hATl(AT2K255IC3); and hATl (A243+).
78. A non-endogenous version of a human G protein-coupled receptor encoded by a
cDNA of claim 77.
79. A Plasmid comprising a Vector and the cDNA of claim 77.
80. A Host Cell comprising the Plasmid of claim 79.
- *A**********A**A*A*A**A*ιkA
EP99950301A 1998-10-13 1999-10-13 Non-endogenous, constitutively activated human g protein-coupled receptors Withdrawn EP1137776A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP05009877A EP1676861A3 (en) 1998-10-13 1999-10-13 Non-endogenous, constitutively activated human G protein-coupled receptors
EP10007972A EP2264068A1 (en) 1998-10-13 1999-10-13 Non-endogenous, constitutively activated human G protein-coupled receptors

Applications Claiming Priority (65)

Application Number Priority Date Filing Date Title
US09/170,496 US6555339B1 (en) 1997-04-14 1998-10-13 Non-endogenous, constitutively activated human protein-coupled receptors
US170496 1998-10-13
US10802998P 1998-11-12 1998-11-12
US108029P 1998-11-12
US10921398P 1998-11-20 1998-11-20
US109213P 1998-11-20
US11006098P 1998-11-27 1998-11-27
US110060P 1998-11-27
US12041699P 1999-02-16 1999-02-16
US120416P 1999-02-16
US12185299P 1999-02-26 1999-02-26
US121852P 1999-02-26
US12395199P 1999-03-12 1999-03-12
US12394699P 1999-03-12 1999-03-12
US12394499P 1999-03-12 1999-03-12
US12394599P 1999-03-12 1999-03-12
US12394999P 1999-03-12 1999-03-12
US12394899P 1999-03-12 1999-03-12
US123951P 1999-03-12
US123946P 1999-03-12
US123948P 1999-03-12
US123949P 1999-03-12
US123944P 1999-03-12
US123945P 1999-03-12
US13643699P 1999-05-28 1999-05-28
US13756799P 1999-05-28 1999-05-28
US13643799P 1999-05-28 1999-05-28
US13713199P 1999-05-28 1999-05-28
US13643999P 1999-05-28 1999-05-28
US13712799P 1999-05-28 1999-05-28
US136437P 1999-05-28
US137127P 1999-05-28
US136436P 1999-05-28
US136439P 1999-05-28
US137567P 1999-05-28
US137131P 1999-05-28
US14144899P 1999-06-29 1999-06-29
US141448P 1999-06-29
US15111499P 1999-08-27 1999-08-27
US151114P 1999-08-27
US15252499P 1999-09-03 1999-09-03
US152524P 1999-09-03
US15655599P 1999-09-29 1999-09-29
US15663499P 1999-09-29 1999-09-29
US15663399P 1999-09-29 1999-09-29
US15665399P 1999-09-29 1999-09-29
US156634P 1999-09-29
US156555P 1999-09-29
US156633P 1999-09-29
US15729399P 1999-10-01 1999-10-01
US15728199P 1999-10-01 1999-10-01
US15728299P 1999-10-01 1999-10-01
US15729499P 1999-10-01 1999-10-01
US15728099P 1999-10-01 1999-10-01
US157294P 1999-10-01
US157293P 1999-10-01
US157282P 1999-10-01
US157281P 1999-10-01
US157280P 1999-10-01
US41704499A 1999-10-12 1999-10-12
US41676099A 1999-10-12 1999-10-12
US417044 1999-10-12
US416760 1999-10-12
PCT/US1999/024065 WO2000022131A2 (en) 1998-10-13 1999-10-13 Non-endogenous, constitutively activated human g protein-coupled receptors
2000-02-23

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP05009877A Division EP1676861A3 (en) 1998-10-13 1999-10-13 Non-endogenous, constitutively activated human G protein-coupled receptors

Publications (1)

Publication Number Publication Date
EP1137776A2 true EP1137776A2 (en) 2001-10-04

Family

ID=27586909

Family Applications (1)

Application Number Title Priority Date Filing Date
EP99950301A Withdrawn EP1137776A2 (en) 1998-10-13 1999-10-13 Non-endogenous, constitutively activated human g protein-coupled receptors

Country Status (2)

Country Link
EP (1) EP1137776A2 (en)
HK (1) HK1042511A1 (en)

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0022131A3 *

Also Published As

Publication number Publication date
HK1042511A1 (en) 2002-08-16

Similar Documents

Publication Publication Date Title
WO2000022131A2 (en) Non-endogenous, constitutively activated human g protein-coupled receptors
AU2004203102B2 (en) Human G Protein-Coupled Receptors
AU2005244540B2 (en) Endogenous and non-endogenous versions of human G protein-coupled receptors
AU2001249885A1 (en) Non-endogenous, constitutively activated known g protein-coupled receptors
WO2000031258A2 (en) Human orphan g protein-coupled receptors
US20130165633A1 (en) Endogenous and Non-Endogenous Versions of Human G Protein-Coupled Receptors
US20130309766A1 (en) Endogenous and Non-Endogenous Versions of Human G Protein-Coupled Receptors
AU2002219890B2 (en) Endogenous and non-endogenous versions of human G protein-coupled receptors
AU2002219890A1 (en) Endogenous and non-endogenous versions of human G protein-coupled receptors
US8440391B2 (en) Constitutively activated human G protein coupled receptors
AU6299199A (en) Non-endogenous, constitutively activated human g protein-coupled receptors
EP2264068A1 (en) Non-endogenous, constitutively activated human G protein-coupled receptors
EP1137776A2 (en) Non-endogenous, constitutively activated human g protein-coupled receptors
US20070072248A1 (en) Constitutively activated human G protein coupled receptors
US20150153327A1 (en) Endogenous and Non-Endogenous Versions of Human G Protein-Coupled Receptors
JP2003525018A6 (en) Non-endogenous constitutively activated human G protein-coupled receptor
CA2732120A1 (en) Endogenous and non-endogenous versions of human g protein-coupled receptors
AU2007216752A1 (en) Endogenous and non-endogenous versions of human G protein-coupled receptor hRUP35
JP2011172577A (en) Non-endogenous, constitutively activated human g protein-coupled receptor

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20010508

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

AX Request for extension of the european patent

Free format text: AL;LT;LV;MK;RO;SI

17Q First examination report despatched

Effective date: 20030718

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20051109

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1042511

Country of ref document: HK