WO2001048189A1 - Novel guanosine triphosphate-binding protein-coupled receptors, genes thereof and production and use of the same - Google Patents
Novel guanosine triphosphate-binding protein-coupled receptors, genes thereof and production and use of the same Download PDFInfo
- Publication number
- WO2001048189A1 WO2001048189A1 PCT/JP2000/009409 JP0009409W WO0148189A1 WO 2001048189 A1 WO2001048189 A1 WO 2001048189A1 JP 0009409 W JP0009409 W JP 0009409W WO 0148189 A1 WO0148189 A1 WO 0148189A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- protein
- seq
- dna
- amino acid
- acid sequence
- 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.)
- Ceased
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70571—Receptors; Cell surface antigens; Cell surface determinants for neuromediators, e.g. serotonin receptor, dopamine receptor
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/72—Receptors; Cell surface antigens; Cell surface determinants for hormones
- C07K14/723—G protein coupled receptor, e.g. TSHR-thyrotropin-receptor, LH/hCG receptor, FSH receptor
Definitions
- the present invention relates to novel G protein-coupled receptors and their genes, and their production and use.
- G protein-coupled receptors are a general term for a group of cell membrane receptors that transmit signals into cells through the activation of trimeric GTP-binding proteins. G protein-coupled receptors are also called “seven-transmembrane receptors” because of their structural characteristics of having seven transmembrane regions in the molecule. G protein-coupled receptors transmit information on various physiologically active substances from the cell membrane into cells through the activation of trimeric GTP-binding proteins and the resulting changes in intracellular second messengers.
- Intracellular second messengers which are controlled by trimeric GTP-binding proteins cAMP which via adenylyl two cyclase, although such Ca 2 + which via a phospholipase C is well known, via trimeric GTP-binding protein
- cAMP which via adenylyl two cyclase
- Ca 2 + which via a phospholipase C is well known
- trimeric GTP-binding protein via trimeric GTP-binding protein
- Substrates (ligands) for G protein-coupled receptors are very diverse, including proteinaceous hormones, chemokines, peptides, amines, lipid-derived substances, and proteases such as thrombin.
- the number of G protein-coupled receptors whose genes have been identified is less than 300 in humans excluding sensory organ receptors, but the number of G protein-coupled receptors whose ligands have been identified is about There are only 140 types and the ligand is unknown. —There are more than 100 types of “fan G protein-coupled receptors”. However, it is assumed that there are at least 400, and sometimes as many as 1000, G-protein coupled receptors in the actual human genome (Trends Pharmacol. Sci. (97) 18: 430). This means that the number of orphan G protein-coupled receptors of unknown function will explode with the rapid progress of genome analysis in the future.
- the orphan G protein-coupled receptor identified by genetic manipulation is used to screen physiological agonism by function screening using changes in intracellular second messenger cAMP and Ca 2+ as indices. This is to discover strikes and perform in vivo functional analysis. At this time, by using a compound library to increase the throughput of the screening, it was possible to discover specific surrogate agonists for orphan G protein-coupled receptors and angel gonists, and eventually to identify specific Theoretically, the development of therapeutic drugs for diseases is possible. Disclosure of the invention
- the present invention has been made in view of the current situation surrounding such a G protein-coupled receptor, and an object thereof is to provide a novel G protein-coupled receptor and a gene thereof, as well as methods for producing and using the same. It is in.
- the aim is to provide these molecules as targets for drug development research.
- the present inventors have conducted intensive studies in order to solve the above-mentioned problems, and as a result, by carrying out the polymerase chain reaction using human tissue cDNA as a ⁇ type, the characteristic of G protein-coupled receptor
- the present invention relates to novel G protein-coupled receptors and their genes, and their production and use, and more specifically,
- SEQ ID NO: 9 to 16; 35 to 36; 46 to 50 SEQ ID NO: consisting of an amino acid sequence in which one or more amino acids are substituted, deleted, added and / or inserted in the amino acid sequence described in any one of 1 to 8, 33 to 34, 41 to 45 DNA encoding proteins,
- step (b) selecting a compound that reduces the binding activity detected in step (a) as compared to the binding activity in the absence of the test sample; (9) A method for screening for a compound that inhibits or promotes the activity of the protein according to (1) or (2),
- step (c) selecting a compound that suppresses or enhances the change in the cells detected in step (b) as compared with the change in the cells in the absence of the test sample;
- a pharmaceutical composition comprising the compound according to (12) as an active ingredient, and
- the “G protein-coupled receptor” refers to a cell membrane receptor that transmits a signal into a cell through activation of a GTP-binding protein.
- ligand refers to a physiological substance that binds to a G protein-coupled receptor and transmits a signal into a cell.
- physiological substance refers to a compound that binds to a G protein-coupled receptor in a living body.
- agonist refers to a compound capable of transmitting a signal into cells by binding to a G protein-coupled receptor, and includes physiological substances, artificially synthesized compounds, and naturally occurring compounds. Including.
- angigonist refers to a compound that inhibits the binding of a ligand to a G protein-coupled receptor or the transmission of a signal into a cell, and is a physiological substance or an artificially synthesized substance. Compounds, including naturally occurring compounds.
- the present invention provides a novel G protein-coupled receptor and a DNA encoding the protein. Included in the present invention, 15 human-derived cDNA clones isolated by the present inventors, ⁇ GPRv4 '', ⁇ GPRvll '', ⁇ GPRvl3 '', ⁇ GPRvl4 '', ⁇ GPRvl5 '', ⁇ GPRvl9 '', Named "GPRv20", “GPRv31", “GPRv38”, “GPRv39”, “GPRv68", “GPRv77”, “GPRv78”, “GPRv79", “GPRv81” (If necessary, collect these clones together. GPRv ”).
- nucleotide sequences of these cDNAs are shown in SEQ ID NOs: 9 to 16, 35 to 36, and 46 to 50, and the amino acid sequences of the proteins encoded by the cDNAs are shown in SEQ ID NOs: 1 to 8, 33 to 3 4, 4 1 to 45 are shown.
- GPRv 4 '' has 31% homology to ORYLA PROBABLE G PROTEIN-COUPLED RECEPTOR (Q91178, 428aa), and ⁇ GPRvll '' has HUMAN NEUROPEPTIDE Y RECEPTOR TYPE 2 (P49146,
- GPRv protein the protein encoded by the GPRv cDNA isolated by the present inventors (hereinafter sometimes referred to as “GPRv protein”) has seven transmembrane domains characteristic of a G protein-coupled receptor. It retained possible hydrophobic regions. From these facts, it is considered that all GPRv cDNAs encode proteins belonging to the G protein-coupled receptor family. G protein-coupled receptors have the activity of transmitting signals into cells through the activation of G proteins by the action of their ligands, and as described above, include genetic diseases, brain nervous system, circulatory It has been implicated in numerous areas of disease, including system, digestive, immune, motor, and genitourinary. Therefore, the GPRv protein can be used for screening of agonists and angiogonists that regulate the function of the GPRv protein, and is an important target for the development of drugs for the above diseases.
- the present invention also provides a protein functionally equivalent to the GPRv protein.
- “Functionally equivalent” means that the protein of interest has biological properties equivalent to the GPRv protein.
- the biological properties of GPRv proteins include the activity of transducing signals into cells through the activation of trimeric GTP-binding proteins. Trimeric GTP-binding proteins fall into one of three categories, depending on the type of intracellular signaling system that activates, Gq type that increases Ca 2+ , Gs type that increases cAMP, and Gi type that suppresses cAMP. (Trends Pharmacol. Sci. (99) 20: 118). Therefore, whether the target protein has the same biological properties as the GPRv protein This can be evaluated, for example, by detecting a change in intracellular cAMP concentration or calcium concentration due to the activation.
- One embodiment of a method for preparing a protein functionally equivalent to the GPRv protein includes a method of introducing a mutation into an amino acid sequence in a protein.
- Such methods include, for example, site-directed mutagenesis (Current Protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons Section 8.1-8.5)). Amino acid mutations in proteins may also occur in nature.
- the amino acid sequence of the GPRv protein (SEQ ID NO: 1 from 8, 33 to 34, 41 to 45) irrespective of whether it is artificial or naturally occurring is 1 or It includes proteins in which a plurality of amino acids are mutated by substitution, deletion, insertion, Z or addition, and the like, and includes proteins functionally equivalent to the GPRv protein.
- the number and location of amino acid mutations in these proteins are not limited as long as the function of the GPRv protein is maintained.
- the number of mutations will typically be within 10% of all amino acids, preferably within 5% of all amino acids, and more preferably within 1% of all amino acids.
- Another embodiment of a method for preparing a protein functionally equivalent to the GPRv protein includes a method utilizing a hybridization technique or a gene amplification technique. That is, those skilled in the art can use a hybridization technique (Current Protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons Section 6.3-6.4) to make a DNA sequence encoding a GPRv protein. (SEQ ID NOS: 9 to 16, 35 to 36, 46 to 50) or a portion thereof, from which a highly homologous DNA is isolated from a DNA sample derived from a homologous or heterologous organism. Obtaining a protein functionally equivalent to the GPRv protein from the DNA can be usually performed.
- a protein encoded by a DNA that hybridizes with a DNA encoding a GPRv protein and that is functionally equivalent to the GPRv protein is also included in the protein of the present invention.
- organisms for isolating such proteins include, but are not limited to, rats, mice, egrets, chicks, birds, and sea lions, in addition to humans.
- Stringent hybridization conditions for isolating DNA that encodes a protein functionally equivalent to the GPRv protein are usually of the order of ⁇ lxSSC, 0.1% SDS, 37 ° C ''.
- the more strict conditions are about 0.5xSSC, 0.13 ⁇ 4SDS, 42 ° C, and the more severe conditions are about 0.2xSSC, 0.1% SDS, 65 ° C. Is the condition.
- the isolation of DNA having higher homology to the probe sequence can be expected as the hybridization conditions become more severe.
- the combination of the above SSC, SDS and temperature conditions is merely an example, and those skilled in the art will recognize the above or other factors (eg, probe concentration, probe length, etc.) that determine the stringency of a high predication solution. , Hybridization reaction time, etc.) as appropriate, it is possible to realize the same stringency as described above.
- the protein encoded by DNA isolated using such a hybridization technique usually has high homology in amino acid sequence with the GPRv protein.
- High homology refers to sequence homology of at least 40% or more, preferably 60% or more, more preferably 80% or more (eg, 90% or more and 95% or more).
- PCR gene amplification technique
- the present invention also includes a partial peptide of the protein of the present invention.
- This partial peptide includes a peptide that binds to a ligand but does not transmit a signal.
- An affinity column prepared based on such a peptide can be suitably used for screening of a ligand.
- the partial peptide of the protein of the present invention can also be used for preparing an antibody.
- the partial peptide of the present invention can be produced, for example, by a genetic technique, a known peptide synthesis method, or by cleaving the protein of the present invention with an appropriate peptidase.
- the partial peptide of the present invention usually has 8 amino acid residues or more, preferably 12 amino acid residues or more (for example, 15 amino acid residues or more).
- the protein of the present invention can be prepared as a recombinant protein or as a natural protein.
- the recombinant protein can be prepared, for example, by introducing a vector into which a DNA encoding the protein of the present invention is inserted into an appropriate host cell as described below, and purifying the protein expressed in the transformant.
- a natural protein can be prepared using, for example, an affinity column to which an antibody against the protein of the present invention described below is bound (Current Protocols in Molecular Biology edit. Ausubel et al. (1987)). Publish. John Wiley & Sons Section 16. 16.16).
- the antibody used for affinity purification may be a polyclonal antibody or a monoclonal antibody.
- the present invention also provides a DNA encoding the protein of the present invention.
- the form of the DNA of the present invention is not particularly limited as long as it can encode the protein of the present invention, and includes genomic DNA, chemically synthesized DNA, and the like in addition to cDNA.
- DNAs having any base sequence based on the degeneracy of the genetic code are included as long as they can encode the protein of the present invention.
- the DNA of the present invention comprises a DNA sequence encoding a GPRv protein (SEQ ID NOS: 9 to 16, 35 to 36, 46 to 50) or a part thereof as a probe. It can be isolated by a conventional method such as the PCR method using a primer synthesized based on these DNA sequences.
- the present invention also provides a vector into which the DNA of the present invention has been inserted.
- the vector of the present invention is not particularly limited as long as it stably retains the inserted MA.
- pBluescript vector-1 (Stratagene Inc.) may be used as a vector for cloning. Is preferred.
- an expression vector is particularly useful.
- the expression vector is not particularly limited as long as it is a vector that expresses a protein in a test tube, in E. coli, in cultured cells, or in an individual organism.
- pBEST vector Promega for expression in a test tube PET vector (manufactured by Invitrogen) for Escherichia coli, PME18S-FL3 vector (GenBank Accession No. AB009864) for cultured cells, pME18S vector (Mol Cell Biol. 8: 466-472 (Mol Cell Biol. 8: 466-472) for living organisms. 1988)).
- Insertion of the DNA of the present invention into a vector can be performed by a conventional method, for example, by a ligase reaction using a restriction enzyme site (Current protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons. Section 11.4-11. 11).
- the present invention also provides a transformant carrying the DNA of the present invention or the vector of the present invention.
- the host cell into which the vector of the present invention is introduced is not particularly limited, and various host cells can be used depending on the purpose. Examples of eukaryotic cells for highly expressing a protein include COS cells and CH0 cells.
- Vector introduction into host cells includes, for example, calcium phosphate precipitation, electropulse perforation (Current protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons.Section 9.1-9.9), It can be performed by a known method such as a lipofectamine method (manufactured by GIBCO-BRL) or a microinjection method.
- the present invention also relates to a DNA encoding the protein of the present invention (a DNA comprising the nucleotide sequence of any one of SEQ ID NOs: 9 to 16, 35 to 36, and 46 to 50, or a complementary strand thereof).
- a DNA comprising the nucleotide sequence of any one of SEQ ID NOs: 9 to 16, 35 to 36, and 46 to 50, or a complementary strand thereof.
- the “complementary strand” refers to one strand of a double-stranded nucleic acid consisting of A: T (U for RNA) and G: C base pairs, with respect to the other strand.
- complementary is not limited to a sequence completely complementary to at least 15 contiguous nucleotide regions, but is at least 70%, preferably at least 80%, more preferably 90%, and still more preferably 95%. What is necessary is that they have homology on the base sequence of at least%.
- the algorithm described in this specification may be used as an algorithm for determining homology.
- Such nucleotides can be used as a probe for detecting and isolating the MA of the present invention, and as a primer for amplifying the DNA of the present invention. When used as a primer, it usually has a chain length of 15 bp to 100 bp, preferably 15 bp to 35 bp.
- nucleotide having a chain length of at least 15 bp containing at least a part or the entire sequence of the MA of the present invention is used.
- Such nucleotides preferably specifically hybridize to DNA encoding the protein of the present invention.
- “Specifically hybridizes” means under normal hybridization conditions, preferably under stringency.
- the DNA that encodes the protein of the present invention (SEQ ID NOs: 9 to 16, 35 to 36, 46 to 50) hybridizes with the DNA encoding the protein of the present invention under a simple condition, and encodes another protein. NA means do not hybridize.
- nucleotides can be used for testing and diagnosing abnormalities of the protein of the present invention.
- abnormal expression of the DNA encoding the protein of the present invention can be examined by Northern hybridization or RT-PCR using these nucleotides as probes or primers.
- the DNA encoding the protein of the present invention and its expression control region are amplified by polymerase chain reaction (PCR) using these nucleotides as primers, and the DNA is amplified by methods such as RFLP analysis, SSCP, and sequencing. Inspection and diagnosis of sequence abnormalities.
- PCR polymerase chain reaction
- these nucleotides include antisense DNA for suppressing the expression of the protein of the present invention.
- the antisense MA has a chain length of at least 15 bp or more, preferably 100 bp, more preferably 500 bp or more, and usually has a chain length of 3000 bp or less, preferably 2000 bp or less in order to cause an antisense effect.
- Such antisense MA may also be applied to gene therapy for diseases caused by abnormalities (functional abnormalities or current abnormalities) of the protein of the present invention.
- the antisense DNA is, for example, a phosphorothionate based on the sequence information of a DNA encoding the protein of the present invention (eg, SEQ ID NOS: 9 to 16, 35 to 36, 46 to 50).
- Nucleic Acids Res 16, 3209-21 (1988)), etc. can be prepared by the method (Stein, 1988 Physicochemical properties oi phosphorotnioate oligodeoxynucle otides. Nucleic Acids Res 16, 3209-21 (1988)).
- the nucleotides of the present invention can be used, for example, in ex vivo by using a viral vector such as a retrovirus vector, an adenovirus vector, an adeno-associated virus vector, or a non-viral vector such as a ribosome. o It may be possible to administer to patients by the method or in vivo method.
- the present invention also provides an antibody that binds to the protein of the present invention.
- the form of the antibody of the present invention is not particularly limited, and includes a polyclonal antibody, a monoclonal antibody, and a part thereof having antigen-binding properties. In addition, antibodies of all classes are included.
- the antibodies of the present invention also include special antibodies such as humanized antibodies.
- the antibody of the present invention can be obtained by synthesizing an oligonucleotide corresponding to the amino acid sequence of the protein of the present invention according to a conventional method, and immunizing a rabbit (Current protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons. Section 11.12-11.13).
- a monoclonal antibody a mouse is immunized with a protein expressed and purified in Escherichia coli according to a conventional method, and a hybridoma cell obtained by fusing the spleen cell and myeloma cell thereof is prepared. It can be obtained from dorma cells (Current protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons. Section 11.4- 11.11).
- Antibodies that bind to the protein of the present invention may be used, for example, for the examination and diagnosis of abnormal expression or structural abnormality of the protein of the present invention, in addition to purification of the protein of the present invention.
- proteins are extracted from tissues, blood, or cells, and abnormalities in expression and structure are detected through detection of the proteins of the present invention by methods such as Western blotting, immunoprecipitation, and ELISA. Inspection / presence can be diagnosed.
- an antibody that binds to the protein of the present invention for the purpose of treating a disease associated with the protein of the present invention.
- the antibody of the present invention can act as an agonist of the protein of the present invention.
- a human antibody or a humanized antibody is preferred because of its low immunogenicity.
- Human antibodies include mice in which the immune system has been replaced with that of a human (eg, "" Functional transpant of megabase human immunoglobulin loci recapitulates human antibody responses in mice, Mendez, MJ et al. (1997) Nat. Genet.15 : 146-156 ").
- humanized antibodies are It can be prepared by genetic recombination using the hypervariable region of a null antibody (Methods in Enzymology 203, 99-121 (1991)).
- the present invention also provides a method for screening for a ligand that binds to the protein of the present invention, using the protein of the present invention.
- This screening method includes (a) a step of bringing a test sample into contact with a protein of the present invention or a partial peptide thereof, and (b) a step of selecting a compound that binds to the protein or a partial peptide thereof.
- test sample is not particularly limited.
- known compounds or peptides whose ligand activities of various G protein-coupled receptors are unknown for example, those registered in a chemical file
- phage A random 'peptide group created by applying a display method J. Mol. Biol. (1991) 222, 301-310)
- culture supernatants of microorganisms and natural components derived from plants and marine organisms are also targets for screening.
- Other examples include, but are not limited to, brain and other biological tissue extracts, cell extracts, and expression products of gene libraries.
- the protein of the present invention used for screening may be, for example, a form expressed on a cell surface, a form as a cell membrane fraction of the cell, or a form bound to an affinity column.
- Specific screening techniques include, for example, a method of contacting a test sample with an affinity column for the protein of the present invention to purify a compound that binds to the protein of the present invention, and a number of known methods such as a West Western plotting method. Methods are available. When these methods are used, the test sample is appropriately labeled, and the binding to the protein of the present invention can be detected using the label.
- a cell membrane that expresses the protein of the present invention is prepared and immobilized on a chip, and the dissociation of the trimeric GTP-binding protein upon ligand binding is determined by surface plasmon resonance (surface plasmon resonance).
- the binding activity between the test sample and the protein of the present invention can be detected by using, as an index, a change in cells caused by binding of the test sample to the protein of the present invention expressed on the cell surface.
- changes include, but are not limited to, changes in intracellular Ca 2+ levels and changes in cAMP levels.
- agonist activity for G protein-coupled receptors can be measured by the GTP-S binding method.
- 20 mM HEPES cell membranes was expressed G protein-coupled receptors (pH7.4), lOOmM NaCl, lOmM gCl 2, 50 in M GDP solution, labeled with 35 S GTP After mixing with 400 pM of ⁇ S and incubating in the presence and absence of the test sample, filtration is performed, and a method of comparing the radioactivity of the bound GTP ⁇ S can be used.
- G protein-coupled receptors share a system that transmits signals into cells via activation of trimeric GTP-binding proteins.
- Trimeric GTP-binding proteins are classified into three types, depending on the type of intracellular signaling system that activates, Gq type that increases Ca 2+ , Gs type that increases cAMP, and Gi type that suppresses cAMP. This chimerized was Sabuyuni' Bok and non Gq protein shed Sabuyunitto and other G proteins by applying the positive signals upon ligand subscription-learning intracellular pathway of Gq, thereby resulting in Ca 2 + increase It is possible.
- the elevated Ca 2+ level can be detected using the repo overnight gene system having TRE (TP A responsive element) upstream, a staining indicator such as Fluor-3, and a change in the fluorescent protein aequorin as an index.
- TRE TP A responsive element
- Gs protein subunits are chimerized with other G protein subunits, and a positive signal is consequently increased to cAMP, a Gs intracellular transduction pathway, and CRE (cAMP-responsive element) is upstream. It is also possible to use as an index the change in the repo overnight gene system in (Trends Pharmacol. Sci. (99) 20: 118).
- the host cell that expresses the protein of the present invention in this screening system there are no particular restrictions on the host cell that expresses the protein of the present invention in this screening system, and various host cells may be used depending on the purpose.
- Cells CHO cells, HEK293 cells, and the like.
- the vector for expressing the protein of the present invention in vertebrate cells include a promoter located upstream of a gene encoding the protein of the present invention, an RNA splice site, a polyadenylation site, a transcription termination sequence, and an origin of replication. Those having the above can be preferably used.
- pSV2dhfr ol.Cell.Biol. (1981) 1, 854-864
- pEF-BOS Nucleic Acids Res.
- pCDM8 (Nature (1987) ) 329, 840-842), pCEP4 (Invitrogen) and the like are useful vectors for expressing G protein-coupled receptors.
- the DNA of the present invention can be inserted into a vector by a ligase reaction using a restriction enzyme site according to a conventional method (Current protocol in Molecular Biology eait. Ausubel et al. (1987) Publish. John Wiley). & Sons. Section 11.4 ⁇ ; 11.11).
- the introduction of a vector into a host cell can be performed, for example, by the calcium phosphate precipitation method or the electric pulse perforation method (Current protocols in Molecular Biology edit.
- the present invention also provides a method for screening a compound having an activity of inhibiting the binding of the protein of the present invention to its ligand.
- This screening method comprises the steps of: (a) contacting a ligand of the protein or its partial peptide in the presence of a test sample with a ligand, and detecting the binding activity between the protein or its partial peptide and the ligand; b) selecting a compound that reduces the binding activity detected in step (a) as compared to the binding activity in the absence of the test sample.
- test sample is not particularly limited.
- a compound group obtained by combinatorial chemistry—technology (Tetrahedron (1995) 51, 8135-8137), or a phage display method (J. Mol. Biol. (1991) 222, 301-310) can be used. Screening also includes culture supernatants of microorganisms and natural components derived from plants and marine organisms. Other examples include, but are not limited to, brain and other biological tissue extracts, cell extracts, expression products of gene libraries, synthetic low molecular compounds, synthetic peptides, natural compounds, and the like.
- the protein of the present invention used in the screening may be, for example, in a form expressed on the cell surface, as a cell membrane fraction of the cell, or in a form bound to an affinity column.
- a specific screening method for example, a method in which a ligand is labeled with a radioisotope or the like, and the ligand is contacted with the protein of the present invention in the presence of a test sample, and then compared with the case where detection is performed in the absence of a test sample Then, a method of detecting a compound that reduces the binding activity between the protein and the ligand of the present invention based on the label attached to the ligand can be used.
- a compound expressing the protein of the present invention is brought into contact with a ligand in the presence of a test sample, and a compound that reduces a change in the cell compared to the case where the ligand is detected in the absence of the test sample is selected.
- a compound that inhibits the binding between the protein of the present invention and a ligand Cells expressing the protein of the present invention can be prepared in the same manner as in the above-described screening for a ligand that binds to the protein of the present invention.
- the compound isolated by this screening is a candidate for the agonist of the protein of the present invention.
- the present invention also provides a method for screening a compound that inhibits or promotes the activity of the protein of the present invention.
- This screening method is based on (a) the existence of a test sample. Contacting a cell that expresses the protein of the present invention with a ligand of the protein in the presence of the protein, (b) detecting a change in the cell due to binding of the ligand to the protein of the present invention, (c) absence of a test sample Selecting a compound that suppresses or enhances the change in the cell detected in step (b) as compared to the change in the cell below.
- a compound group obtained by a combinatorial chemistry technique, a phage display method, etc. are applied in the same manner as the above-mentioned screening method of a compound that inhibits the binding between the protein and the ligand of the present invention.
- a compound isolated by screening a compound that inhibits the binding between the protein of the present invention and a ligand can be used as a test sample.
- Cells expressing the protein of the present invention can be prepared in the same manner as in the above-described screening for a ligand that binds to the protein of the present invention. Changes in the cells after contact with the test sample can be detected by using changes in intracellular Ca 2+ levels and cAMP levels as indices, as in the above-described screening method. When detecting intracellular signal transduction, it is also possible to detect using a measurement system such as a repo overnight system using luciferase or the like as a reporter gene.
- a measurement system such as a repo overnight system using luciferase or the like as a reporter gene.
- the sample is determined to be a compound that inhibits the activity of the protein of the present invention.
- the test sample enhances the change in the cells, the compound is determined to be a compound that promotes the activity of the protein of the present invention.
- “promoting or inhibiting the activity of the protein of the present invention” means whether the action is direct or indirect on the protein of the present invention. It means that the activity of the protein of the present invention is promoted or inhibited.
- compounds isolated by this screening include compounds that act on the protein or ligand of the present invention to inhibit or promote their binding and thereby inhibit or promote the activity of the protein of the present invention.
- compounds that do not inhibit or promote these bindings per se but result in inhibiting or promoting the activity of the protein of the present invention are also included.
- Such compounds include, for example, compounds that do not inhibit the binding of the protein of the present invention to the ligand, but inhibit or promote intracellular signaling pathways.
- a compound isolated by the screening method of the present invention is used as a pharmaceutical
- the isolated compound itself is administered directly to a patient or administered as a pharmaceutical composition formulated by a known pharmaceutical method. It is also possible to do.
- a pharmaceutical composition formulated by a known pharmaceutical method.
- it is conceivable to administer the composition by appropriately combining it with a pharmacologically acceptable carrier or vehicle, specifically, sterile water, physiological saline, vegetable oil, emulsifier, suspension, and the like.
- Administration to a patient can be generally performed by methods known to those skilled in the art, such as, for example, intraarterial injection, intravenous injection, and subcutaneous injection. The dose varies depending on the weight and age of the patient, the administration method, and the like, but those skilled in the art can appropriately select an appropriate dose.
- the compound can be encoded by DNA
- the DNA may be incorporated into a gene therapy vector to perform gene therapy.
- FIG. 1 is a diagram showing the result of performing a BLAST search on the entire sequence of SWISS-PR0T using the amino acid sequence of “GPRv4” as rQueryj. It showed 31% homology to ORYLA PROBABLE G PR0TEIN-C0UPL ED RECEPTOR.
- FIG. 2 is a diagram showing the results of performing a BLAST search on the entire sequence of SWISS-PR0T using the rcPRvllj amino acid sequence as Queryj. It showed 31% homology to HUMAN NEUROPEPTIDE Y RECEPTOR TYPE 2.
- FIG. 3 is a diagram showing the results of performing a BLAST search on all SWISS-PROT sequences using the “GPRvl3” amino acid sequence as rQueryj. PONPY C5A showed 39% homology to ANAPHYLATOXIN CHEM0T ACTIC RECEPTOR.
- FIG. 4 is a diagram showing the results of performing a BLAST search on the entire SWISS-PR0T sequence using the rGPRvHj amino acid sequence as rQueryj. It showed 40% homology to CHICK P2Y PURIN0CEPT0R5.
- FIG. 5 is a diagram showing the results of performing a BLAST search on the entire SWISS-PROT sequence using the amino acid sequence of “GPRvl5” as Queryj. It showed 26% homology to HUMAN 5-HYDR0XYTRYPTAMINE IE.
- FIG. 6 is a diagram showing the results of performing a BLAST search on the entire sequence of SWISS-PROT using the amino acid sequence of “GPRvl9” as rQueryj. It showed 25% homology to APIME OPSIN and BLUE-SENSITIVE.
- FIG. 7 is a diagram showing the results of performing a BLAST search on all SWISS-PROT sequences using the amino acid sequence of “GPRv20” as rQueryj. It showed 38% homology to RAT MAS PR0T0-ONCOGENE.
- Figure 8 shows the results of a BLAST search of all SWISS-PROT sequences using the amino acid sequence of “GPRv31” as “Queryj.” 29% homology to SHEEP THYROTROPIN- RELEASING HORMONE RECEPTOR showed that.
- FIG. 9 is a diagram showing the result of performing a BLAST search on the entire SWISS-PROT sequence with the amino acid sequence of “GPRv38” set to rQuer j. It showed 46% homology to P2Y PURIN0CEPT0R 7 (Q15722).
- FIG. 10 is a diagram showing the result of performing a BLAST search on the entire sequence of SWISS-PR0T using the amino acid sequence of “GPRv39” as rQueryj. It showed 35% homology to NEUROTENSIN RECEPTOR TYPE 1 (P20789).
- FIG. 11 is a diagram showing the results of performing a BLAST search on the entire SWISS-PROT sequence using the amino acid sequence of “GPRv68” as rQueryj. 39% showed the highest homology to TYPE-IB ANGIOTENSIN II RECEPTOR (Q13725).
- FIG. 12 is a diagram showing the result of performing a BLAST search on the entire SWISS-PR0T sequence using the amino acid sequence of “GPRv77” as rQueryj. It showed the highest homology at 29 ° to HUMAN PUTATIVE G PROTEIN-COU PLED RECEPTOR GPR17 (R12) (Q13304).
- FIG. 13 is a diagram showing the results of performing a BLAST search on the entire SWISS-PROT sequence using the amino acid sequence of “GPRv78” as rQueryj. 39% showed the highest homology with HUMAN GALANIN RECEPTOR TYPE 2 (043603).
- FIG. 14 is a diagram showing the results of performing a BLAST search on the entire SWISS-PROT sequence using the amino acid sequence of “GPRv79” as rQueryj. 39% showed the highest homology to RAT MAS PR0T0-0NC0GENE (P125 26).
- FIG. 15 is a diagram showing the results of performing a BLAST search on the entire sequence of SWISS-PR0T using the amino acid sequence of “GPRv81” as rQueryj. 25% showed the highest homology to HUMAN 5-HYDR0XYTRYPTAMINE IB RECEPTOR (P28222).
- the full length cDNA encoding the novel G protein-coupled receptor of the present invention (GPRv4, GPRvll, GPRvl3, GPRvH, GPRvl 5, GPRvl9, GPRv20, GPRv31, GPRv38, GPRv39, GPRv68, GPRv77, GPRv78, GPRv7 9, GPRv81) Obtained by PCR.
- Marathon Ready cDNA derived from human fetal brain (Clontech) is used as type II cDNA, and as a forward primer, 5,-ATGGCCA ACTCCACAGGGCTGAACGCCT-3 '(SEQ ID NO: 17) 5, -TCAGGAGAGAACTCTCAGGTGGCCCC-3 '(SEQ ID NO: 18) was used as a reverse primer.
- PCR was carried out using Pyrobes tDNA polymerase (Takara Shuzo) in the presence of 5% formamide, after 94 ° C (2 minutes), 98 ° C (30 seconds) / 65 ° C (30 seconds) / 75 ° C (2 Min) cycle was repeated 30 times.
- This sequence has an open reading frame of 1107 bases (from 1st to 1107th of SEQ ID NO: 9).
- the amino acid sequence (368 amino acids) predicted from the open reading frame is shown in SEQ ID NO: 1.
- the predicted amino acid sequence has a hydrophobic region that is thought to be seven transmembrane domains that are characteristic of G protein-coupled receptors, indicating that this gene encodes a G protein-coupled receptor. Revealed.
- Marathon Ready cDNA (Clontech) derived from human fetus is used as type II cDNA, and as a forward primer, 5,-ATGCAGGC GCTTAACATTACCCCGGAGC-3 '(SEQ ID NO: 19), reverse primer 5, -T TAATGCCCACTGTCTAAAGGAGAATTC-3 '(SEQ ID NO: 20) was used.
- PCR was performed using Pyrobest DNA polymerase (Takara Shuzo) in the presence of 5% formamide, followed by five cycles of 94 ° C (2.5 seconds), followed by 94 ° C (5 seconds) / 72 ° C (2 minutes).
- This sequence has an open reading frame of 1296 bases (1st to 1296th of SEQ ID NO: 10).
- the amino acid sequence (431 amino acids) predicted from the open reading frame is shown in SEQ ID NO: 2.
- the predicted amino acid sequence has a hydrophobic region that seems to be seven transmembrane domains characteristic of G protein-coupled receptors, indicating that this gene encodes a G protein-coupled receptor did.
- the sequence has an open reading frame of 1014 bases (1st to 1014th of SEQ ID NO: 11).
- the amino acid sequence (337 amino acids) predicted from the open reading frame is shown in SEQ ID NO: 3.
- the predicted amino acid sequence has a hydrophobic region that seems to be seven transmembrane domains characteristic of G protein-coupled receptors, indicating that this gene encodes a G protein-coupled receptor did.
- This sequence has an open reading frame of 1119 bases (1st to 1119th of SEQ ID NO: 12).
- the amino acid sequence (372 amino acids) predicted from the open reading frame is shown in SEQ ID NO: 4.
- the predicted amino acid sequence has a hydrophobic region that seems to be seven transmembrane domains characteristic of G protein-coupled receptors, indicating that this gene encodes a G protein-coupled receptor did.
- the amino acid sequence (609 amino acid) predicted from the open reading frame is shown in SEQ ID NO: 5.
- the predicted amino acid sequence has a hydrophobic region that is thought to be the seven transmembrane domains characteristic of a G protein-coupled receptor, indicating that this gene encodes a G protein-coupled receptor did.
- This sequence has an open reading frame of 951 bases (SEQ ID NO: 14).
- the amino acid sequence (316 amino acid) predicted from the open reading frame is shown in SEQ ID NO: 6. Since the predicted amino acid sequence has seven transmembrane domains that are likely to be characteristic of G protein-coupled receptors, the gene may encode a G protein-coupled receptor. found.
- Marathon Ready cDNA (Clontech) derived from human fetus is used as type I cDNA, and as a forward primer, 5,-ATGGATCC AACCATCTCAACCTTGGACAC-3 '(SEQ ID NO: 29), reverse 5, -TCAGGTTAGATAAACATCTATTTGAAGAC-3 '(SEQ ID NO: 30) was used as a primer.
- Marathon Ready cDNA (Clontech) derived from human fetus is used as type I cDNA, 5, -ATGGTTGG AGACACATTAAAACTTCTG-3 '(SEQ ID NO: 31) as the forward primer, reverse primer 5, -TC ATGGCATGACAACCAGATTAGGAAAG-3 '(SEQ ID NO: 32) was used.
- PCR was performed using Pyrobest DNA polymerase (Takara Shuzo), followed by a cycle of 94 ° C (30 seconds) / 50 ° C (30 seconds) / 72 ° C (2 minutes) after 94 ° C (2 minutes). Repeated times.
- This sequence has an open reading frame of 1062 bases (SEQ ID NO: 16).
- the amino acid sequence (353 amino acid) predicted from the open reading frame is shown in SEQ ID NO: 8.
- the predicted amino acid sequence is the G protein-coupled receptor The presence of the characteristic seven transmembrane hydrophobic regions suggests that this gene encodes a G protein-coupled receptor.
- a human brain-derived Marathon Ready cDNA (Clontech) was used as type I cMA, and as a forward primer, 5, -ATGTCGGTCT GCTACCGTCCCCCAGGGA-3 '(SEQ ID NO: 37) and reverse 5, -TCA AAGGTCCCATTCCGGACCGTCCTTC-3 ′ (SEQ ID NO: 38) was used as a primer.
- PCR was performed using Pyrobest DNA polymerase (Takara Shuzo) in the presence of 5% formamide, followed by 5 cycles of 98 ° C (2.5 minutes) followed by 98 ° C (5 seconds) / 72 ° C (4 minutes).
- amino acid sequence (358 amino acids) predicted from the open reading frame is shown in SEQ ID NO: 33. Since the predicted amino acid sequence has a hydrophobic region that seems to be seven transmembrane domains that are characteristic of G protein-coupled receptors, it was determined that this gene encodes a G protein-coupled receptor did.
- This sequence has an open reading frame of 1248 bases (SEQ ID NO: 36).
- the amino acid sequence (415 amino acid) predicted from the open reading frame is shown in SEQ ID NO: 34. Since the predicted amino acid sequence has a hydrophobic region that seems to be seven transmembrane domains that are characteristic of G protein-coupled receptors, it was determined that this gene encodes a G protein-coupled receptor did.
- human genomic DNA (Clontech) was converted to type I cDNA, 5, -ATGCAGATGGCCGATGCAGCCACGATA G-3 '(SEQ ID NO: 51) as a forward primer, and 5, -TCAGTAGGCAGAGCTGCTGG as a reverse primer.
- GCAGCAGG-3 ′ (SEQ ID NO: 52) was used.
- PCR use Pyrobest DNA polymerase (Takara Shuzo), and cycle 98 ° C (30 seconds) / 55 ° C (30 seconds) / 72 ° C (4 minutes) after 98 ° C (2.5 minutes). Repeated 35 times.
- This sequence has an open reading frame of 1410 bases (SEQ ID NO: 46).
- the amino acid sequence (469 amino acid) predicted from the open reading frame is shown in SEQ ID NO: 41. Since the predicted amino acid sequence has a hydrophobic region that seems to be seven transmembrane domains that are characteristic of G protein-coupled receptors, it was determined that this gene encodes a G protein-coupled receptor did.
- PCR was performed using Pyrobe st DNA polymerase (Takara Shuzo), followed by 30 cycles of 94 ° C (30 seconds) / 55 ° C (30 seconds) / 72 ° C (3 minutes) after 94 ° C (2.5 minutes). Repeated times. As a result, a DNA fragment of about 1.0 kbp was amplified. This fragment was cloned using pCR2.1 plasmid (Invitrogen). The nucleotide sequence of the obtained clone was analyzed using the ABI377 DNA Sequencer (Applied Biosystems) by the dideoxy-mine-one-time method. The elucidated sequence is shown in SEQ ID NO: 47.
- This sequence has an open reading frame of 1011 bases (SEQ ID NO: 47).
- the amino acid sequence (336 amino acid) predicted from the open reading frame is shown in SEQ ID NO: 42. Since the predicted amino acid sequence has a hydrophobic region that seems to be seven transmembrane domains that are characteristic of G protein-coupled receptors, it was determined that this gene encodes a G protein-coupled receptor did.
- This sequence has an open reading frame of 1197 bases (SEQ ID NO: 48). Amino acid sequence predicted from open reading frame (398 Amino acid) is shown in SEQ ID NO: 43. Since the predicted amino acid sequence has a hydrophobic region that seems to be seven transmembrane domains that are characteristic of G protein-coupled receptors, it was determined that this gene encodes a G protein-coupled receptor did.
- human genomic DNA (Clontech) was converted to type I cDNA, 5'-atggatccaaccaccccggcctgggga a-3 (SEQ ID NO: 57) as the primary primer, and 5,5 as the reverse primer.
- Pyrobest DNA polymerase (Takara Shuzo) was used, and after 94 ° C (2 minutes), a cycle of 94 ° C (30 seconds) / 55 ° C (30 seconds) / 72 ° C (2.5 minutes) was performed. Repeated 30 times.
- the sequence has an open reading frame of 993 bases (SEQ ID NO: 49).
- the amino acid sequence (330 amino acids) predicted from the open reading frame is shown in SEQ ID NO: 44.
- the predicted amino acid sequence has a hydrophobic region that is thought to be the seven transmembrane domains characteristic of a G protein-coupled receptor, indicating that this gene encodes a G protein-coupled receptor did.
- human genomic DNA (Clontech) is converted to type II cDNA, 5'-ATGGGGGATGAGCTGGCACCTTGCCCT G-3, (SEQ ID NO: 59) as a forward primer, and 5 CTAGGAAATGGTAAAGATGG CCTGGTGC- as a reverse primer. 3 ′ (SEQ ID NO: 60) was used.
- Pyrobest DNA polymerase (Takara Shuzo) was used, and after a cycle of 94 ° C (2 minutes), a cycle of 94 ° C (30 seconds) / 55 ° C (30 seconds) / 72 ° C (2.5 minutes) was performed. Repeated 30 times.
- This sequence has an open reading frame (SEQ ID NO: 50) of 1044 bases.
- the amino acid sequence (347 amino acids) predicted from the open reading frame is shown in SEQ ID NO: 45. Since the predicted amino acid sequence has a hydrophobic region that seems to be seven transmembrane domains that are characteristic of G protein-coupled receptors, it was determined that this gene encodes a G protein-coupled receptor did.
- Example 2 BLAST search for SWISS-PR0T using the amino acid sequence of a novel G protein-coupled receptor
- GPRvll The BLAST search results for SWISS-PR0T with the amino acid sequence of “GPRvll” are shown in FIG. “GPRvll” showed the highest homology at 31 ° to HUMAN NEUROPEPTIDE Y RECEPTOR TYPE 2 (P49146, 381aa) among known G protein-coupled receptors. This proved that "GPRvll” is a novel G protein-coupled receptor.
- GPRvl3 The BLAST search results for SWISS-PR0T with the amino acid sequence of “GPRvl3” are shown in FIG. “GPRvl3” showed the highest homology at 39% to P0NPY C5A A APHYLAT OXIN CHEMOTACTIC RECEPTOR (P79234, 340aa) among known G protein-coupled receptors. This proved that “GPRvl3” is a novel G protein-coupled receptor.
- GPRvl4 I is a known G protein-coupled receptor that is CHICK P2Y PURIN0CEP 40% showed the highest homology to TOR 5 (P32250, 308aa). This proved that "GPRvl4" was a novel G protein-coupled receptor.
- GPRvl5 BLAST search results for SWISS-PR0T with the amino acid sequence of “GPRvl5” are shown in FIG. “GPRvl5” showed the highest homology at 26% to HUMAN 5-HYDR0XYTRYP TAMINE IE RECEPTOR (P28566, 365aa) among known G protein-coupled receptors. This proved that “GPRvl5” is a novel G protein-coupled receptor.
- GPRvl9 The results of a BLAST search for SWISS-PR0T with the amino acid sequence of “GPRvl9” are shown in FIG. "GPRvl9” showed the highest homology at 25% among APIME OPSIN and BLUE-S ENSITIVE (P90680, 377aa) among known G protein-coupled receptors. This indicated that "GPRvl9” was a novel G protein-coupled receptor.
- GPRv20 showed the highest homology of 38% to RAT MAS PR0T0-0NC0G ENE (P12526, 324aa) among known G protein-coupled receptors. This indicated that “GPRv20” was a novel G protein-coupled receptor.
- GPRv31 The results of a BLAST search for SWISS-PR0T with the amino acid sequence of “GPRv31” are shown in FIG. “GPRv31” showed the highest homology at 29% to SHEEP THYR0TR0PIN-R ELEASING HORMONE RECEPTOR (Q28596, 398aa) among known G protein-coupled receptors. This indicated that “GPRv31” is a novel G protein-coupled receptor.
- FIG. 9 shows the results of a BLAST search for SWISS-PR0T using the amino acid sequence of “GPRv38”.
- GPRv38 was not the same among known G protein-coupled receptors and showed the highest homology at 46% to P2Y PURINOCEPTOR 7 (Q15722, 352aa). This indicated that “GPRv38” is a novel G protein-coupled receptor.
- FIG. 10 shows the results of a BLAST search for SWISS-PR0T using the amino acid sequence of “GPRv39”.
- GPRv39 is the same among known G protein-coupled receptors, and is the highest at 35% of RAT NEUROTENSIN RECEPTOR TYPE 1 (P20789, 424aa) Showed high homology. This indicated that “GPRv39” is a novel G protein-coupled receptor.
- GPRv68 was not the same among known G protein-coupled receptors, and showed the highest homology at 39 ° to TYPE-IB ANGIOTENSIN II RECEPTOR (Q13725, 359aa). This indicated that “GPRv68” is a novel G protein-coupled receptor.
- GPRv77 is a novel G protein-coupled receptor.
- GPRv78 was not the same among known G protein-coupled receptors, and showed the highest homology at 39% with HUMAN GAL IN IN RECEPTOR TYPE 2 (043603, 387aa). This proved that "GPRv78” is a novel G protein-coupled receptor.
- GPRv79 was not the same as any known G protein-coupled receptor, and showed the highest homology at 39% to RAT MAS PROTO-ONCOGENE (P12526, 324aa). From this, it was determined that “GPRv79” is a novel G protein-coupled receptor.
- GPRv81 is a novel G protein-coupled receptor.
- Industrial applicability is not the same among known G protein-coupled receptors, and is 25% of HUMAN 5-HYDROXYTRYPTAMINE IB RECEPTOR (P28222, 390aa). It showed the highest homology. This proved that “GPRv81” is a novel G protein-coupled receptor.
- a novel G protein-coupled receptor (GPRv4, GPRvll, GPRvl3, GPRv, GPRvl5, GPRvl9, GPRv20, GPRv31, GPRv38, GPRv39, GPRv68, GPRv77, GPRv78, GPRv79, GPRv81)
- a vector containing the gene, a host cell containing the vector, and a method for producing the protein are provided. Furthermore, a method for screening a compound that modifies the activity of the protein was provided.
- the protein of the present invention, its gene, or a compound that modulates the activity of the protein of the present invention is expected to be used for the development of new preventive or therapeutic agents for diseases involving the G protein-coupled receptor protein of the present invention. Is done.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Cell Biology (AREA)
- General Health & Medical Sciences (AREA)
- Immunology (AREA)
- Toxicology (AREA)
- Zoology (AREA)
- Gastroenterology & Hepatology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Biochemistry (AREA)
- Biophysics (AREA)
- Molecular Biology (AREA)
- Genetics & Genomics (AREA)
- Medicinal Chemistry (AREA)
- Neurology (AREA)
- Biomedical Technology (AREA)
- Engineering & Computer Science (AREA)
- Endocrinology (AREA)
- Peptides Or Proteins (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Description
明細書 新規なグァノシン三リン酸結合蛋白質共役型の受容体およびそれらの遺伝子、 並びにそれらの製造および用途 技術分野 Description Novel guanosine triphosphate-binding protein-coupled receptor, their genes, and their production and use
本発明は、 新規な G蛋白質共役型受容体およびそれらの遺伝子、 並びにそれら の製造および用途に関する。 背景技術 The present invention relates to novel G protein-coupled receptors and their genes, and their production and use. Background art
G蛋白質共役型受容体(G protein-coupled receptors )は、 三量体型 GTP結合 蛋白質の活性化を介して細胞内にシグナルを伝達する細胞膜受容体群の総称であ る。 G蛋白質共役型受容体は、 分子内に細胞膜貫通領域を 7回有する構造上の特 性から、 「7回膜貫通型受容体」 とも呼ばれる。 G蛋白質共役型受容体は様々な 生理活性物質の情報を、 三量体型 GTP結合蛋白質の活性化、 それにより引き起こ される細胞内セカンドメッセンジャーの変動を介して細胞膜から細胞内へと伝達 する。 三量体型 GTP結合蛋白質により制御される細胞内セカンドメッセンジャー は、 アデ二レートシクラーゼを介する cAMP、 フォスフォリパーゼ Cを介する Ca2 +などがよく知られているが、 三量体型 GTP結合蛋白質を介したチヤネルの制御、 リン酸化酵素の活性化など多くの細胞蛋白がその標的となっていることが最近明 らかとなつてきた (Annu. Rev. Neurosci , ( 97) 20 : 399) 。 G蛋白質共役型受容体 に対する基質 (リガンド) は、 大変多岐に渡っており、 タンパク性ホルモン、 ケ モカイン、 ペプチド、 ァミン、 脂質由来物質、 さらにはトロンビンの様なプロテ ァーゼもその一例となる。 現在、 遺伝子が同定された G蛋白質共役型受容体の数 は感覚器受容体を除くと、 ヒトで 300個弱存在するが、 リガンドが同定された G 蛋白質共役型受容体の数は、 そのうち約 140種類に過ぎず、 リガンド未知な 「ォ —ファン G蛋白質共役型受容体」 が 100種類以上存在している。 しかしながら実 際のヒトゲノム中には、 少なくとも 400種類、 場合によっては 1000種類もの G 蛋白質共役型受容体が存在する、 とも想定されている (Trends Pharmacol. Sci . ( 97) 18:430) 。 この事は、 今後のゲノム解析の飛躍的進展に伴って、 機能未知 なォーファン G蛋白質共役型受容体の数も爆発的に増加する事を意味している。 これまでに世界の製薬企業により創られてきた薬剤は、 その 9割以上が細胞外 空間での相互作用を標的としており、 その中でも G蛋白質共役型受容体に関連す る低分子薬は大部分を占めている。 その根拠としては、 G蛋白質共役型受容体が 関連する疾患が、 遺伝的疾患を始めとして、 脳神経系、 循環器系、 消化器系、 免 疫系、 運動器系、 泌尿器生殖器系など、 非常に多くの領域に関連することにある そのため、 最近では多くの製薬企業がゲノム解析で明らかとなったォ一ファン G 蛋白質共役型受容体を所有し、 リガンド探索と生理機能の解明に鎬を削っている こうした状況を背景として、 最近では新規 G蛋白質共役型受容体の生理的リガン ド探索の成功例も報告され始めている。 例えば、 calcitonin gene-related pept ide受容体 (J. Biol .Chem. (96 ) 271 : 11325)、 orexin (Cell (98) 92 : 573)そして prolactin- releasing peptide (Nature ( 98) 393 :272)などの事例は、 生命科学 分野での基礎研究としても大きな衝撃を持つ事例であった。 G protein-coupled receptors are a general term for a group of cell membrane receptors that transmit signals into cells through the activation of trimeric GTP-binding proteins. G protein-coupled receptors are also called “seven-transmembrane receptors” because of their structural characteristics of having seven transmembrane regions in the molecule. G protein-coupled receptors transmit information on various physiologically active substances from the cell membrane into cells through the activation of trimeric GTP-binding proteins and the resulting changes in intracellular second messengers. Intracellular second messengers which are controlled by trimeric GTP-binding proteins, cAMP which via adenylyl two cyclase, although such Ca 2 + which via a phospholipase C is well known, via trimeric GTP-binding protein Recently, it has been revealed that many cellular proteins are targeted, such as the control of channels and the activation of phosphorylase (Annu. Rev. Neurosci, (97) 20: 399). Substrates (ligands) for G protein-coupled receptors are very diverse, including proteinaceous hormones, chemokines, peptides, amines, lipid-derived substances, and proteases such as thrombin. At present, the number of G protein-coupled receptors whose genes have been identified is less than 300 in humans excluding sensory organ receptors, but the number of G protein-coupled receptors whose ligands have been identified is about There are only 140 types and the ligand is unknown. —There are more than 100 types of “fan G protein-coupled receptors”. However, it is assumed that there are at least 400, and sometimes as many as 1000, G-protein coupled receptors in the actual human genome (Trends Pharmacol. Sci. (97) 18: 430). This means that the number of orphan G protein-coupled receptors of unknown function will explode with the rapid progress of genome analysis in the future. Over 90% of drugs created by global pharmaceutical companies so far target interactions in the extracellular space, of which small molecule drugs related to G protein-coupled receptors are mostly Occupy. The basis for this is that diseases associated with G protein-coupled receptors are very rare, including genetic diseases, cerebral nervous system, circulatory system, digestive system, immune system, exercise system, urogenital system, etc. Therefore, many pharmaceutical companies have recently possessed an orphan G protein-coupled receptor that was revealed by genomic analysis, and have spent much of their time searching for ligands and elucidating physiological functions. Against this background, there have recently been reports of successful searches for physiological ligands for novel G protein-coupled receptors. For example, calcitonin gene-related peptide receptor (J. Biol. Chem. (96) 271: 11325), orexin (Cell (98) 92: 573) and prolactin-releasing peptide (Nature (98) 393: 272) This case had a great impact on basic research in the life science field.
特に、 才ーファン G蛋白質共役型受容体は新たな薬剤開発に繋がる可能性の高 い標的として、 多大な注目を集めている。 一般的にォーファン' G蛋白質共役型受 容体には特異的なリガンドが存在しないため、 そのァゴニスト、 アン夕ゴニスト を開発することは困難であった。 しかし、 近年、 充実された化合物ライブラリー とハイスループットスクリーニングと組み合わせることで、 ォ一ファン G蛋白質 共役型受容体を標的とした薬剤の創製が提唱されている (Trends Pharmacol . Sc i . ( 97) 18:430, Br. J.Pharm. ( 98) 125 : 1387) 。 すなわち、 遺伝子操作によつ て同定されたォーファン G蛋白質共役型受容体を、 細胞内セカンドメッセンジャ 一である cAMP, Ca2+の変化を指標とした機能スクリーニングにより生理的ァゴニ ストを発見し、 生体内機能解析を行うというものである。 この際、 化合物ライブ ラリーを利用して、 スクリーニングをハイスループット化することにより、 ォー ファン G蛋白質共役型受容体に対する特異的な代替 (surrogate) ァゴニスト及 びアン夕ゴニス卜の発見、 ひいては特定の疾患治療薬の開発も理論的には可能と なる。 発明の開示 In particular, human-fan G protein-coupled receptors have received a great deal of attention as targets that are likely to lead to new drug development. In general, since there is no specific ligand for orphan'G protein-coupled receptor, it has been difficult to develop its agonist and angyu gonist. However, in recent years, it has been proposed to create a drug targeting an orphan G protein-coupled receptor by combining an extensive compound library with high-throughput screening (Trends Pharmacol. Sc i. (97) 18: 430, Br. J. Pharm. (98) 125: 1387). In other words, the orphan G protein-coupled receptor identified by genetic manipulation is used to screen physiological agonism by function screening using changes in intracellular second messenger cAMP and Ca 2+ as indices. This is to discover strikes and perform in vivo functional analysis. At this time, by using a compound library to increase the throughput of the screening, it was possible to discover specific surrogate agonists for orphan G protein-coupled receptors and angel gonists, and eventually to identify specific Theoretically, the development of therapeutic drugs for diseases is possible. Disclosure of the invention
本発明は、 このような G蛋白質共役型受容体を取り巻く現状に鑑みてなされた ものであり、 その目的は新規な G蛋白質共役型受容体およびその遺伝子、 並びに それらの製造方法及び用途を提供することにある。 さらにこれら分子を薬剤開発 研究の標的として提供することを目的とする。 The present invention has been made in view of the current situation surrounding such a G protein-coupled receptor, and an object thereof is to provide a novel G protein-coupled receptor and a gene thereof, as well as methods for producing and using the same. It is in. The aim is to provide these molecules as targets for drug development research.
本発明者らは、 上記課題を解決するために鋭意研究を重ねた結果、 ヒト組織 c DNAを鎵型にしたポリメラーゼ連鎖反応を実施することにより、 G蛋白質共役型 受容体の特徴である 7個の膜貫通ドメインと考えられる疎水性領域を保持する 1 5種類の新規遺伝子を単離することに成功した。 これら遺伝子やその翻訳産物で ある蛋白質は、 リガンドのスクリーニングや医薬品として有用なァゴニストゃァ ン夕ゴニス卜のスクリーニングに利用し得る。 The present inventors have conducted intensive studies in order to solve the above-mentioned problems, and as a result, by carrying out the polymerase chain reaction using human tissue cDNA as a 鎵 type, the characteristic of G protein-coupled receptor We have succeeded in isolating 15 novel genes that have a hydrophobic region that is considered to be a transmembrane domain. These genes and the proteins that are their translation products can be used for screening for ligands and for screening agonists that are useful as pharmaceuticals.
即ち、 本発明は、 新規な G蛋白質共役型受容体およびそれらの遺伝子、 並びに それらの製造および用途に関し、 より具体的には、 That is, the present invention relates to novel G protein-coupled receptors and their genes, and their production and use, and more specifically,
( 1 ) グアノシン三リン酸結合蛋白質共役型の受容体をコ一ドする下記 ( a ) から (d ) のいずれかに記載の DNA、 (1) the DNA according to any one of (a) to (d) below, which encodes a guanosine triphosphate binding protein-coupled receptor;
( a ) 配列番号: 1から 8、 3 3から 3 4、 4 1から 4 5のいずれかに記載のァ ミノ酸配列からなる蛋白質をコードする DNA、 (a) SEQ ID NO: 1 to 8, 33 to 34, or 41 to 45, a DNA encoding a protein comprising the amino acid sequence described in any one of:
( b ) 配列番号: 9から 1 6、 3 5から 3 6、 4 6から 5 0のいずれかに記載の 塩基配列のコード領域を含む DNA、 (c) 配列番号: 1から 8、 33から 34、 41から 45のいずれかに記載のァ ミノ酸配列において 1もしくは複数のアミノ酸が置換、 欠失、 付加および/また は挿入したアミノ酸配列からなる蛋白質をコードする DNA、 (b) SEQ ID NO: 9 to 16; 35 to 36; 46 to 50 (c) SEQ ID NO: consisting of an amino acid sequence in which one or more amino acids are substituted, deleted, added and / or inserted in the amino acid sequence described in any one of 1 to 8, 33 to 34, 41 to 45 DNA encoding proteins,
( d ) 配列番号: 9から 16、 35から 36、 46から 50のいずれかに記載の 塩基配列からなる DNAにストリンジェントな条件下でハイブリダィズする DNA、 (d) a DNA that hybridizes under stringent conditions to a DNA consisting of the nucleotide sequence of any one of SEQ ID NOs: 9 to 16, 35 to 36, and 46 to 50,
(2) 配列番号: 1から 8、 33から 34、 41から 45のいずれかに記載 のアミノ酸配列からなる蛋白質の部分べプチドをコ一ドする DNA、 (2) a DNA encoding a partial peptide of a protein consisting of the amino acid sequence of any one of SEQ ID NOs: 1 to 8, 33 to 34, and 41 to 45;
(3) ( 1) または (2) に記載の DNAを含有するベクター、 (3) a vector containing the DNA of (1) or (2),
(4) ( 1) または (2) に記載の DNAまたは (3) に記載のベクターを保 持する形質転換体、 (4) a transformant carrying the DNA of (1) or (2) or the vector of (3),
(5) ( 1) または (2) に記載の DNAによりコードされる蛋白質またはべ プチド、 (5) a protein or peptide encoded by the DNA of (1) or (2),
(6) (4) に記載の形質転換体を培養し、 該形質転換体またはその培養上 清から発現させた蛋白質またはペプチドを回収する工程を含む、 (5) に記載の 蛋白質またはべプチドの製造方法、 (6) the step of culturing the transformant according to (4), and recovering the protein or peptide expressed from the transformant or the culture supernatant thereof; Production method,
(7) (5) に記載の蛋白質に結合するリガンドのスクリーニング方法であ つて、 (7) A method for screening a ligand that binds to a protein according to (5),
(a) (5) に記載の蛋白質またはペプチドに被検試料を接触させる工程、 (a) contacting a test sample with the protein or peptide according to (5),
(b) 該蛋白質またはペプチドに結合する化合物を選択する工程、 を含む方法、(b) selecting a compound that binds to the protein or peptide;
(8) ( 1) または (2) に記載の蛋白質とそのリガンドとの結合を阻害す る活性を有する化合物のスクリーニング方法であって、 (8) A method for screening a compound having an activity of inhibiting the binding of the protein according to (1) or (2) to a ligand thereof,
(a) 被検試料の存在下で (1) または (2) に記載の蛋白質またはその部分べ プチドにリガンドを接触させ、 該蛋白質またはその部分べプチドとリガンドとの 結合活性を検出する工程、 (a) contacting a ligand with the protein or the partial peptide thereof according to (1) or (2) in the presence of the test sample, and detecting the binding activity between the protein or the partial peptide and the ligand;
(b) 被検試料非存在下での結合活性と比較して、 工程 (a) で検出された結合 活性を低下させる化合物を選択する工程、 を含む方法、 (9) ( 1) または (2) に記載の蛋白質の活性を阻害または促進する化合 物をスクリーニングする方法であって、 (b) selecting a compound that reduces the binding activity detected in step (a) as compared to the binding activity in the absence of the test sample; (9) A method for screening for a compound that inhibits or promotes the activity of the protein according to (1) or (2),
(a) 被検試料の存在下で該蛋白質を発現する細胞に該蛋白質のリガンドを接触 させる工程、 (a) contacting a cell expressing the protein with a ligand for the protein in the presence of a test sample;
(b) 該リガンドの該蛋白質への結合による細胞における変化を検出する工程、 (b) detecting a change in a cell due to binding of the ligand to the protein,
( c) 被検試料非存在下での細胞における変化と比較して、 工程 (b) で検出さ れた細胞における変化を抑制または増強させる化合物を選択する工程、 を含む方 法、 (c) selecting a compound that suppresses or enhances the change in the cells detected in step (b) as compared with the change in the cells in the absence of the test sample;
( 10) 細胞における変化が、 cAMP濃度の変化またはカルシウム濃度の変 化である、 (8) または (9) に記載の方法、 (10) The method according to (8) or (9), wherein the change in the cell is a change in cAMP concentration or a change in calcium concentration.
( 1 1) ( 1) または (2) に記載の蛋白質に結合する抗体、 (1 1) an antibody that binds to the protein of (1) or (2),
( 12) (7) から ( 10) のいずれかに記載のスクリーニングにより単離 される化合物、 および (12) a compound isolated by the screening according to any of (7) to (10), and
( 13) (12) に記載の化合物を有効成分とする医薬組成物、 および (13) A pharmaceutical composition comprising the compound according to (12) as an active ingredient, and
( 14) 配列番号: 9から 16、 35から 36、 46から 50のいずれかに 記載の塩基配列からなる DNAまたはその相補鎖に相補的な、 少なくとも 15ヌク レオチドの鎖長を有するヌクレオチド、 を提供するものである。 (14) SEQ ID NO: 9 to 16, 35 to 36, 46 to 50 or a nucleotide having a nucleotide length of at least 15 nucleotides, which is complementary to a complementary strand thereof. Is what you do.
なお、 本発明において 「G蛋白質共役型受容体」 とは、 GTP結合蛋白質の活性 化を介して細胞内にシグナルを伝達する細胞膜受容体を指す。 In the present invention, the “G protein-coupled receptor” refers to a cell membrane receptor that transmits a signal into a cell through activation of a GTP-binding protein.
本発明において 「リガンド」 とは、 G蛋白質共役型受容体に結合し、 細胞内に シグナルを伝達する生理的物質を指す。 ここで 「生理的物質」 とは、 生体内で G 蛋白質共役型受容体に結合している化合物を指す。 In the present invention, “ligand” refers to a physiological substance that binds to a G protein-coupled receptor and transmits a signal into a cell. Here, “physiological substance” refers to a compound that binds to a G protein-coupled receptor in a living body.
本発明において 「ァゴ二スト」 とは、 G蛋白質共役型受容体に結合し、 細胞内 にシグナルを伝達しうる化合物を指し、 生理的物質、 人工的に合成した化合物、 天然由来の化合物を含む。 本発明において 「アン夕ゴニスト」 とは、 リガンドが G蛋白質共役型受容体に 結合すること、 もしくは細胞内にシグナルを伝達することを阻害する化合物を指 し、 生理的物質、 人工的に合成した化合物、 天然由来の化合物を含む。 In the present invention, “agonist” refers to a compound capable of transmitting a signal into cells by binding to a G protein-coupled receptor, and includes physiological substances, artificially synthesized compounds, and naturally occurring compounds. Including. In the present invention, the term “angigonist” refers to a compound that inhibits the binding of a ligand to a G protein-coupled receptor or the transmission of a signal into a cell, and is a physiological substance or an artificially synthesized substance. Compounds, including naturally occurring compounds.
本発明は、 新規な G蛋白質共役型受容体および該蛋白質をコードする DNAを提 供する。 本発明に含まれる、 本発明者等により単離された 1 5のヒト由来の cDN Aクローンを、 「GPRv4」、 「GPRvll」、 「GPRvl3」 、 「GPRvl4」、 「GPRvl5」 、 「GPRvl9」、 「GPRv20」、 「GPRv31」 、 「GPRv38」 、 「GPRv39」、 「GPRv68」 、 「GPRv77」 、 「GPRv78」、 「GPRv79」 、 「GPRv81」 と命名した (必要に応じてこ れらクローンをまとめて 「GPRv」 と称する) 。 これら cDNAの塩基配列を配列番 号: 9から 1 6、 3 5から 3 6、 4 6から 5 0に、 該 cDNAによりコードされる 蛋白質のアミノ酸配列を配列番号: 1から 8、 3 3から 3 4、 4 1から 4 5に示 す。 The present invention provides a novel G protein-coupled receptor and a DNA encoding the protein. Included in the present invention, 15 human-derived cDNA clones isolated by the present inventors, `` GPRv4 '', `` GPRvll '', `` GPRvl3 '', `` GPRvl4 '', `` GPRvl5 '', `` GPRvl9 '', Named "GPRv20", "GPRv31", "GPRv38", "GPRv39", "GPRv68", "GPRv77", "GPRv78", "GPRv79", "GPRv81" (If necessary, collect these clones together. GPRv ”). The nucleotide sequences of these cDNAs are shown in SEQ ID NOs: 9 to 16, 35 to 36, and 46 to 50, and the amino acid sequences of the proteins encoded by the cDNAs are shown in SEQ ID NOs: 1 to 8, 33 to 3 4, 4 1 to 45 are shown.
BLAST検索の結果、 GPRv cDNAがコードする蛋白質は、 いずれも既知の G蛋白 質共役型受容体と有意なアミノ酸配列上の相同性を示した。 具体的には、 「GPRv 4」 は ORYLA PROBABLE G PROTEIN-COUPLED RECEPTOR (Q91178, 428aa)に対して 3 1%の相同性を、 「GPRvll」 は HUMAN NEUROPEPTIDE Y RECEPTOR TYPE 2 (P49146, As a result of BLAST search, all proteins encoded by the GPRv cDNA showed significant amino acid sequence homology with known G protein-coupled receptors. Specifically, `` GPRv 4 '' has 31% homology to ORYLA PROBABLE G PROTEIN-COUPLED RECEPTOR (Q91178, 428aa), and `` GPRvll '' has HUMAN NEUROPEPTIDE Y RECEPTOR TYPE 2 (P49146,
381aa)に対して 31%の相同性を、 「GPRvl3」 は PONPY C5A ANAPHYLATOXIN CHEM0 TACTIC RECEPTOR (P79234, 340aa)に対して 39%の相同性を、 「GPRvl4」 は CHICK381aa) to 31% homology, `` GPRvl3 '' to 39% homology to PONPY C5A ANAPHYLATOXIN CHEM0 TACTIC RECEPTOR (P79234, 340aa), `` GPRvl4 '' to CHICK
P2Y PURIN0CEPT0R 5 (P32250, 308aa)に対して 40%の相同性を、 「GPRvl5」 は H UMAN 5-HYDR0XYTRYPTAMINE IE RECEPTOR (P28566, 365aa) に対して 26%の相同 性を、 「GPRvl9」 は APIME OPSIN, BLUE-SENSITIVE (P90680, 377aa)に対して 2 5%の相同性を、 「GPRv20」 は RAT MAS P画- ONCOGENE (P12526, 324aa)に対し て 38%の相同性を、 「GPRv31」 は SHEEP THYROTROPIN- RELEASING HORMONE RECEPT OR (Q28596, 398aa)に対して 29%の相同性を、 「GPRv38」 は P2Y PURIN0CEPT0R 740% homology to P2Y PURIN0CEPT0R 5 (P32250, 308aa), `` GPRvl5 '' 26% homology to H UMAN 5-HYDR0XYTRYPTAMINE IE RECEPTOR (P28566, 365aa), `` GPRvl9 '' to APIME OPSIN , BLUE-SENSITIVE (P90680, 377aa) has 25% homology, `` GPRv20 '' has 38% homology to RAT MAS P-ONCOGENE (P12526, 324aa), and `` GPRv31 '' has SHEEP 29% homology to THYROTROPIN- RELEASING HORMONE RECEPT OR (Q28596, 398aa), `` GPRv38 '' is P2Y PURIN0CEPT0R 7
(Q15722, 352aa)に対して 46%の相同性を、 「GPRv39」 は RAT NEUROTENSIN RECE PTOR TYPE 1 (P20789, 424aa)に対して 35%の相同性を、 厂 GPRv68」 は TYPE- IB A NGIOTENSIN I I RECEPTOR (Q13725, 359aa)に対して 39%の相同性を、 「GPRv77」 は HUMAN PUTATIVE G PROTEIN-COUPLED RECEPTOR GPR17 (R12) (Q13304, 339aa) に対して 29%の相同性を、 「GPRv78」 は HUMAN GALA IN RECEPTOR TYPE 2 (04360 3, 387aa)に対して 39%の相同性を、 「GPRv79」 は RAT MAS PR0T0- ONCOGENE (P12 526, 324aa)に対して 39%の相同性を、 「GPRv81」 は HUMAN 5-HYDR0XYTRYPTAMINE(Q15722, 352aa) has 46% homology, `` GPRv39 '' has 35% homology to RAT NEUROTENSIN RECE PTOR TYPE 1 (P20789, 424aa), and `` Factory GPRv68 '' has TYPE-IB A 39% homology to NGIOTENSIN II RECEPTOR (Q13725, 359aa), `` GPRv77 '' shows 29% homology to HUMAN PUTATIVE G PROTEIN-COUPLED RECEPTOR GPR17 (R12) (Q13304, 339aa), `` GPRv78 "Has 39% homology to HUMAN GALA IN RECEPTOR TYPE 2 (04360 3, 387aa)," GPRv79 "has 39% homology to RAT MAS PR0T0-ONCOGENE (P12 526, 324aa)," GPRv81 ”is HUMAN 5-HYDR0XYTRYPTAMINE
IB RECEPTOR (P28222, 390aa)に対して 25%の相同性をそれそれ示した。 Each showed 25% homology to IB RECEPTOR (P28222, 390aa).
また、 本発明者等が単離した GPRv cDNAがコードする蛋白質 (以下、 「GPRv 蛋白質」 と称することがある) は、 いずれも G蛋白質共役型受容体の特徴である 7個の膜貫通ドメインと考えられる疎水性領域を保持していた。 これら事実から、 GPRv cDNAは、 いずれも G蛋白質共役型受容体フアミリーに属する蛋白質をコ一 ドしていると考えられる。 G蛋白質共役型受容体は、 そのリガンドの作用により G蛋白質の活性化を通じて細胞内へシグナル伝達を行なう活性を有しており、 上 記したように遺伝的疾患を始めとして、 脳神経系、 循環器系、 消化器系、 免疫系、 運動器系、 泌尿器生殖器系などの非常に多くの領域の疾患に関連している。 従つ て、 GPRv蛋白質は、 GPRv蛋白質の機能を調節するァゴニストやアン夕ゴニスト などのスクリーニングに利用することができ、 上記疾患に対する医薬品の開発の 重要な標的となる。 In addition, the protein encoded by the GPRv cDNA isolated by the present inventors (hereinafter sometimes referred to as “GPRv protein”) has seven transmembrane domains characteristic of a G protein-coupled receptor. It retained possible hydrophobic regions. From these facts, it is considered that all GPRv cDNAs encode proteins belonging to the G protein-coupled receptor family. G protein-coupled receptors have the activity of transmitting signals into cells through the activation of G proteins by the action of their ligands, and as described above, include genetic diseases, brain nervous system, circulatory It has been implicated in numerous areas of disease, including system, digestive, immune, motor, and genitourinary. Therefore, the GPRv protein can be used for screening of agonists and angiogonists that regulate the function of the GPRv protein, and is an important target for the development of drugs for the above diseases.
本発明は、 また、 GPRv蛋白質と機能的に同等な蛋白質を提供する。 ここで The present invention also provides a protein functionally equivalent to the GPRv protein. here
「機能的に同等」 とは、 対象となる蛋白質が GPRv蛋白質と同等の生物学的特性 を有していることを意味する。 GPRv蛋白質が持つ生物学的特性としては、 三量 体型 GTP結合蛋白質の活性化を介して細胞内へシグナル伝達を行なう活性が挙げ られる。 三量体型 GTP結合蛋白質は、 活性化する細胞内伝達系の種類によって、 Ca2+を上昇させる Gq型、 cAMPを上昇させる Gs型、 そして cAMPを抑制する Gi型 の 3種類のカテゴリ一に分類される (Trends Pharmacol . Sci . ( 99) 20: 118)。 従 つて、 対象となる蛋白質が GPRv蛋白質と同等の生物学的特性を有しているか否 かは、 例えば、 その活性化による細胞内の cAMP濃度もしくはカルシウム濃度の 変化を検出することにより評価することが可能である。 “Functionally equivalent” means that the protein of interest has biological properties equivalent to the GPRv protein. The biological properties of GPRv proteins include the activity of transducing signals into cells through the activation of trimeric GTP-binding proteins. Trimeric GTP-binding proteins fall into one of three categories, depending on the type of intracellular signaling system that activates, Gq type that increases Ca 2+ , Gs type that increases cAMP, and Gi type that suppresses cAMP. (Trends Pharmacol. Sci. (99) 20: 118). Therefore, whether the target protein has the same biological properties as the GPRv protein This can be evaluated, for example, by detecting a change in intracellular cAMP concentration or calcium concentration due to the activation.
GPRv蛋白質と機能的に同等な蛋白質を調製するための方法の 1つの態様とし ては、 蛋白質中のアミノ酸配列に変異を導入する方法が挙げられる。 このような 方法には、 例えば、 部位特異的変異誘発法(Current Protocols in Molecular Bi ology edit. Ausubel et al . ( 1987) Publish. John Wiley & Sons Section 8.1 -8.5 )) が含まれる。 また、 蛋白質中のアミノ酸の変異は、 自然界において生じ ることもある。 本発明には、 このように人工的か自然に生じたものかを問わず、 GPRv蛋白質のアミノ酸配列 (配列番号: 1力ら 8、 3 3から 3 4、 4 1から 4 5 ) において 1もしくは複数のアミノ酸が置換、 欠失、 挿入および Zもしくは付 加などにより変異した蛋白質であって、 GPRv蛋白質と機能的に同等な蛋白質が 含まれる。 これら蛋白質におけるアミノ酸の変異数や変異部位は、 GPRv蛋白質 の機能が保持される限り制限はない。 変異数は、 典型的には、 全アミノ酸の 1 0%以内であり、 好ましくは全アミノ酸の 5%以内であり、 さらに好ましくは全 アミノ酸の 1%以内であると考えられる。 One embodiment of a method for preparing a protein functionally equivalent to the GPRv protein includes a method of introducing a mutation into an amino acid sequence in a protein. Such methods include, for example, site-directed mutagenesis (Current Protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons Section 8.1-8.5)). Amino acid mutations in proteins may also occur in nature. In the present invention, the amino acid sequence of the GPRv protein (SEQ ID NO: 1 from 8, 33 to 34, 41 to 45) irrespective of whether it is artificial or naturally occurring is 1 or It includes proteins in which a plurality of amino acids are mutated by substitution, deletion, insertion, Z or addition, and the like, and includes proteins functionally equivalent to the GPRv protein. The number and location of amino acid mutations in these proteins are not limited as long as the function of the GPRv protein is maintained. The number of mutations will typically be within 10% of all amino acids, preferably within 5% of all amino acids, and more preferably within 1% of all amino acids.
GPRv蛋白質と機能的に同等な蛋白質を調製するための方法の他の態様として は、 ハイプリダイゼーシヨン技術あるいは遺伝子増幅技術を利用する方法が挙げ られる。 即ち、 当業者であれば、 ハイブリダィゼ一シヨン技術 (Current Protoc ols in Molecular Biology edit. Ausubel et al . ( 1987) Publish. John Wiley & Sons Section 6.3- 6.4)を利用して GPRv蛋白質をコードする DNA配列 (配列 番号: 9から 1 6、 3 5から 3 6、 4 6から 5 0 ) またはその一部をもとに同種 または異種生物由来の DNA試料から、 これと相同性の高い DNAを単離して、 該 D NAから GPRv蛋白質と機能的に同等な蛋白質を得ることは、 通常行いうることで ある。 このように GPRv蛋白質をコ一ドする DNAとハイブリダィズする DNAによ りコードされる蛋白質であって、 GPRv蛋白質と機能的に同等な蛋白質もまた本 発明の蛋白質に含まれる。 このような蛋白質を単離するための生物としては、 ヒト以外に、 例えば、 ラッ ト、 マウス、 ゥサギ、 ニヮトリ、 ブ夕、 ゥシ等が挙げられるが、 これらに制限さ れない。 Another embodiment of a method for preparing a protein functionally equivalent to the GPRv protein includes a method utilizing a hybridization technique or a gene amplification technique. That is, those skilled in the art can use a hybridization technique (Current Protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons Section 6.3-6.4) to make a DNA sequence encoding a GPRv protein. (SEQ ID NOS: 9 to 16, 35 to 36, 46 to 50) or a portion thereof, from which a highly homologous DNA is isolated from a DNA sample derived from a homologous or heterologous organism. Obtaining a protein functionally equivalent to the GPRv protein from the DNA can be usually performed. Thus, a protein encoded by a DNA that hybridizes with a DNA encoding a GPRv protein and that is functionally equivalent to the GPRv protein is also included in the protein of the present invention. Examples of organisms for isolating such proteins include, but are not limited to, rats, mice, egrets, chicks, birds, and sea lions, in addition to humans.
GPRv蛋白質と機能的に同等な蛋白質をコ一ドする DNAを単離するためのスト リンジェン卜なハイブリダィゼーシヨン条件としては、 通常 「lxSSC、 0. 1% SDS、 37°C」 程度の条件であり、 より厳しい条件としては 「0.5xSSC、 0. 1¾ SDS、 4 2°C」 程度の条件であり、 さらに厳しい条件としては 「0.2xSSC、 0. 1% SDS、 6 5°C」 程度の条件である。 このようにハイブリダィゼーシヨンの条件が厳しくな るほどプローブ配列と高い相同性を有する DNAの単離を期待しうる。 但し、 上記 SSC、 SDSおよび温度の条件の組み合わせは例示であり、 当業者であれば、 ハイ プリダイゼ一シヨンのストリンジエンシーを決定する上記若しくは他の要素 (例 えば、 プローブ濃度、 プローブの長さ、 ハイブリダィゼ一シヨン反応時間など) を適宜組み合わせることにより、 上記と同様のストリンジエンシーを実現するこ とが可能である。 Stringent hybridization conditions for isolating DNA that encodes a protein functionally equivalent to the GPRv protein are usually of the order of `` lxSSC, 0.1% SDS, 37 ° C ''. The more strict conditions are about 0.5xSSC, 0.1¾SDS, 42 ° C, and the more severe conditions are about 0.2xSSC, 0.1% SDS, 65 ° C. Is the condition. Thus, the isolation of DNA having higher homology to the probe sequence can be expected as the hybridization conditions become more severe. However, the combination of the above SSC, SDS and temperature conditions is merely an example, and those skilled in the art will recognize the above or other factors (eg, probe concentration, probe length, etc.) that determine the stringency of a high predication solution. , Hybridization reaction time, etc.) as appropriate, it is possible to realize the same stringency as described above.
このようなハイブリダィゼ一シヨン技術を利用して単離される DN Aがコードす る蛋白質は、 通常、 GPRv蛋白質とアミノ酸配列において高い相同性を有する。 高い相同性とは、 少なくとも 40%以上、 好ましくは 60%以上、 さらに好ましく は 80%以上 (例えば、 90%以上や 95%以上) の配列の相同性を指す。 The protein encoded by DNA isolated using such a hybridization technique usually has high homology in amino acid sequence with the GPRv protein. High homology refers to sequence homology of at least 40% or more, preferably 60% or more, more preferably 80% or more (eg, 90% or more and 95% or more).
アミノ酸配列や塩基配列の同一性は、 Karl in and Altschulによるァルゴリズ ム BLAST(Proc . Natl . Acad. Sei . USA 90 : 5873-5877, 1993 )によって決定する ことができる。 このアルゴリズムに基づいて、 BLASTNや BLASTXと呼ばれるプロ グラムが開発されている(Altschul et al . J. ol . Biol .215 :403-410, 1990 )。 BLASTに基づいて BLASTNによって塩基配列を解析する場合には、 パラメ一夕一 はたとえば score = 100、 word length = 12とする。 また、 BLASTに基づいて BLA STXによってァミノ酸配列を解析する場合には、 パラメ一夕一はたとえば score = 50、 word length = 3とする。 BLASTと Gapped BLASTプログラムを用いる場合 には、 各プログラムのデフォルトパラメ一夕一を用いる。 これらの解析方法の具 体的な手法は公知である(http:〃 www. ncbi .nlm.nih.gov. )。 The identity of the amino acid sequence or nucleotide sequence can be determined by the algorithm BLAST by Karl in and Altschul (Proc. Natl. Acad. Sei. USA 90: 5873-5877, 1993). Based on this algorithm, programs called BLASTN and BLASTX have been developed (Altschul et al. J. ol. Biol. 215: 403-410, 1990). When analyzing a nucleotide sequence by BLASTN based on BLAST, the parameters are, for example, score = 100 and word length = 12. Also, when analyzing an amino acid sequence by BLA STX based on BLAST, the parameters are, for example, score = 50 and word length = 3. When using BLAST and Gapped BLAST programs Use the default parameters of each program. The specific methods of these analysis methods are known (http: @ www.ncbi.nlm.nih.gov.).
また、 遺伝子増幅技術 (PCR) (Current protocols in Molecular Biology ed it. Ausubel et al . ( 1987) Publish. John Wiley & Sons Section 6.1-6.4) を 用いて GPRv蛋白質をコードする DNA配列 (配列番号: 9から 1 6、 3 5から 3 6、 4 6から 5 0 ) の一部を基にプライマ一を設計し、 GPRv蛋白質をコードす る DNA配列と相同性の高い DNA断片を単離し、 該 DNAを基に GPRv蛋白質と機能 的に同等な蛋白質を得ることも可能である。 Also, using a gene amplification technique (PCR) (Current protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons Section 6.1-6.4), a DNA sequence encoding a GPRv protein (SEQ ID NO: 9). From 16 to 35, 36 to 36, and 46 to 50), a primer was designed, a DNA fragment having high homology to the DNA sequence encoding the GPRv protein was isolated, and the DNA was isolated. Based on this, it is also possible to obtain a protein functionally equivalent to the GPRv protein.
本発明は、 また、 本発明の蛋白質の部分ペプチドを含む。 この部分ペプチドに は、 リガンドに結合するがシグナル伝達を行なわないペプチドが含まれる。 この ようなぺプチドを基に作製したァフィ二ティ一カラムは、 リガンドのスクリ一二 ングに好適に用いることができる。 また、 本発明の蛋白質の部分ペプチドは、 抗 体作製に用いることも可能である。 本発明の部分ペプチドは、 例えば、 遺伝子ェ 学的手法、 公知のペプチド合成法、 あるいは本発明の蛋白質を適当なぺプチダ一 ゼで切断することによって製造することができる。 本発明の部分ペプチドは、 通 常、 8アミノ酸残基以上、 好ましくは 12アミノ酸残基以上 (例えば、 15ァミノ 酸残基以上) である。 The present invention also includes a partial peptide of the protein of the present invention. This partial peptide includes a peptide that binds to a ligand but does not transmit a signal. An affinity column prepared based on such a peptide can be suitably used for screening of a ligand. Further, the partial peptide of the protein of the present invention can also be used for preparing an antibody. The partial peptide of the present invention can be produced, for example, by a genetic technique, a known peptide synthesis method, or by cleaving the protein of the present invention with an appropriate peptidase. The partial peptide of the present invention usually has 8 amino acid residues or more, preferably 12 amino acid residues or more (for example, 15 amino acid residues or more).
本発明の蛋白質は、 組み換え蛋白質として、 また天然の蛋白質として調製する ことが可能である。 組み換え蛋白質は、 例えば、 後述するように本発明の蛋白質 をコードする DNAを挿入したベクターを適当な宿主細胞に導入し、 形質転換体内 で発現した蛋白質を精製することにより調製することが可能である。 一方、 天然 の蛋白質は、 例えば、 後述する本発明の蛋白質に対する抗体を結合したァフィ二 ティ一カラムを利用して調製することができる (Current Protocols in Molecul ar Biology edit. Ausubel et al . ( 1987) Publish. John Wiley & Sons Sectio n 16.卜 16.19)。 ァフィ二ティー精製に用いる抗体は、 ポリクローナル抗体であ つてもモノクローナル抗体であってもよい。 また、 インビトロトランスレ一ショ ン (例えば、 「0n the fidelity of mRNA translation in the nuclease - treate d rabbit reticulocyte lysate system. Dasso,M.C. , Jackson, R.J. ( 1989) NAR 1 7:3129-3144」 参照) などにより本発明の蛋白質を調製することも可能である。 また、 本発明は、 上記本発明の蛋白質をコードする DNAを提供する。 本発明の DNAとしては、 本発明の蛋白質をコードしうるものであれば、 その形態に特に制 限はなく、 cDNAの他、 ゲノム DNA、 化学合成 DNAなども含まれる。 また、 本発明 の蛋白質をコードしうる限り、 遺伝暗号の縮重に基づく任意の塩基配列を有する DNAが含まれる。 本発明の DNAは、 上記のように、 GPRv蛋白質をコードする DNA 配列 (配列番号: 9から 1 6、 3 5から 3 6、 4 6から 5 0 ) あるいはその一部 をプローブとしたハイプリダイゼ一シヨン法やこれら DNA配列をもとに合成した プライマーを用いた PCR法等の常法により単離することが可能である。 The protein of the present invention can be prepared as a recombinant protein or as a natural protein. The recombinant protein can be prepared, for example, by introducing a vector into which a DNA encoding the protein of the present invention is inserted into an appropriate host cell as described below, and purifying the protein expressed in the transformant. . On the other hand, a natural protein can be prepared using, for example, an affinity column to which an antibody against the protein of the present invention described below is bound (Current Protocols in Molecular Biology edit. Ausubel et al. (1987)). Publish. John Wiley & Sons Section 16. 16.16). The antibody used for affinity purification may be a polyclonal antibody or a monoclonal antibody. In addition, in vitro translation (For example, see “0n the fidelity of mRNA translation in the nuclease-treate d rabbit reticulocyte lysate system. Dasso, MC, Jackson, RJ (1989) NAR 17: 3129-3144”) and the like. It is also possible. The present invention also provides a DNA encoding the protein of the present invention. The form of the DNA of the present invention is not particularly limited as long as it can encode the protein of the present invention, and includes genomic DNA, chemically synthesized DNA, and the like in addition to cDNA. In addition, DNAs having any base sequence based on the degeneracy of the genetic code are included as long as they can encode the protein of the present invention. As described above, the DNA of the present invention comprises a DNA sequence encoding a GPRv protein (SEQ ID NOS: 9 to 16, 35 to 36, 46 to 50) or a part thereof as a probe. It can be isolated by a conventional method such as the PCR method using a primer synthesized based on these DNA sequences.
また、 本発明は、 本発明の DNAが挿入されたベクターを提供する。 本発明のベ クタ一としては、 挿入した MAを安定に保持するものであれば特に制限されず、 例えば宿主に大腸菌を用いるのであれば、 クロ一ニング用ベクターとしては pBl uescriptベクタ一(Stratagene社製) などが好ましい。 本発明の蛋白質を生産す る目的においてべクタ一を用いる場合には、 特に発現べクタ一が有用である。 発 現べクタ一としては、 試験管内、 大腸菌内、 培養細胞内、 生物個体内で蛋白質を 発現するベクターであれば特に制限されないが、 例えば、 試験管内発現であれば pBESTベクター (プロメガ社製) 、 大腸菌であれば pETベクター (Invitrogen社 製) 、 培養細胞であれば PME18S-FL3ベクタ一 (GenBank Accession No. AB00986 4) 、 生物個体であれば pME18Sベクター (Mol Cell Biol . 8:466-472( 1988)) な どが好ましい。 ベクターへの本発明の DNAの挿入は、 常法により、 例えば、 制限 酵素サイ トを用いたリガーゼ反応により行うことができる (Current protocols in Molecular Biology edit. Ausubel et al . ( 1987) Publish. John Wiley & S ons. Section 11.4-11. 11) 。 また、 本発明は、 本発明の DNAまたは本発明のベクタ一を保持する形質転換体 を提供する。 本発明のベクターが導入される宿主細胞としては特に制限はなく、 目的に応じて種々の宿主細胞が用いられる。 蛋白質を高発現させるための真核細 胞としては、 例えば、 COS細胞、 CH0細胞などを例示することができる。 宿主細 胞へのベクター導入は、 例えば、 リン酸カルシウム沈殿法、 電気パルス穿孔法 (Current protocols in Molecular Biology edit. Ausubel et al . ( 1987) Pub lish. John Wiley & Sons. Section 9. 1-9.9) 、 リポフエクタミン法 (GIBC0-BR L社製) 、 マイクロインジェクション法などの公知の方法で行うことが可能であ る。 The present invention also provides a vector into which the DNA of the present invention has been inserted. The vector of the present invention is not particularly limited as long as it stably retains the inserted MA. For example, if Escherichia coli is used as a host, pBluescript vector-1 (Stratagene Inc.) may be used as a vector for cloning. Is preferred. When a vector is used for the purpose of producing the protein of the present invention, an expression vector is particularly useful. The expression vector is not particularly limited as long as it is a vector that expresses a protein in a test tube, in E. coli, in cultured cells, or in an individual organism. For example, pBEST vector (Promega) for expression in a test tube PET vector (manufactured by Invitrogen) for Escherichia coli, PME18S-FL3 vector (GenBank Accession No. AB009864) for cultured cells, pME18S vector (Mol Cell Biol. 8: 466-472 (Mol Cell Biol. 8: 466-472) for living organisms. 1988)). Insertion of the DNA of the present invention into a vector can be performed by a conventional method, for example, by a ligase reaction using a restriction enzyme site (Current protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons. Section 11.4-11. 11). The present invention also provides a transformant carrying the DNA of the present invention or the vector of the present invention. The host cell into which the vector of the present invention is introduced is not particularly limited, and various host cells can be used depending on the purpose. Examples of eukaryotic cells for highly expressing a protein include COS cells and CH0 cells. Vector introduction into host cells includes, for example, calcium phosphate precipitation, electropulse perforation (Current protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons.Section 9.1-9.9), It can be performed by a known method such as a lipofectamine method (manufactured by GIBCO-BRL) or a microinjection method.
また、 本発明は、 本発明の蛋白質をコードする DNA (配列番号: 9から 1 6、 3 5から 3 6、 4 6から 5 0のいずれかに記載の塩基配列からなる DNAまたはそ の相補鎖) に相補的な、 少なくとも 15ヌクレオチドの鎖長を有するヌクレオチ ドを提供する。 ここで 「相補鎖」 とは、 A:T (ただし RNAの場合は U) 、 G:Cの 塩基対からなる 2本鎖核酸の一方の鎖に対する他方の鎖を指す。 また、 「相補 的」 とは、 少なくとも 15個の連続したヌクレオチド領域で完全に相補配列であ る場合に限られず、 少なくとも 70%、 好ましくは少なくとも 80%、 より好ましく は 90%、 さらに好ましくは 95%以上の塩基配列上の相同性を有すればよい。 相同 性を決定するためのアルゴリズムは本明細書に記載したものを使用すればよい。 このようなヌクレオチドは、 本発明の MAを検出、 単離するためのプローブとし て、 また、 本発明の DNAを増幅するためのプライマーとして利用することが可能 である。 プライマーとして用いる場合には、 通常、 15bp〜100bp、 好ましくは 15 bp〜35bpの鎖長を有する。 また、 プローブとして用いる場合には、 本発明の MA の少なくとも一部若しくは全部の配列を含む少なくとも 15bpの鎖長のヌクレオ チドが用いられる。 このようなヌクレオチドは、 好ましくは本発明の蛋白質をコ ―ドする DNAに特異的にハイプリダイズするものである。 「特異的にハイプリダ ィズする」 とは、 通常のハイプリダイゼーション条件下、 好ましくはストリンジ ェン卜な条件下で、 本発明の蛋白質をコ一ドする DNA (配列番号: 9から 1 6、 3 5から 3 6、 4 6から 5 0 ) とハイブリダィズし、 他の蛋白質をコードする D NAとはハイブリダィズしないことを意味する。 The present invention also relates to a DNA encoding the protein of the present invention (a DNA comprising the nucleotide sequence of any one of SEQ ID NOs: 9 to 16, 35 to 36, and 46 to 50, or a complementary strand thereof). A) a nucleotide having a chain length of at least 15 nucleotides that is complementary to Here, the “complementary strand” refers to one strand of a double-stranded nucleic acid consisting of A: T (U for RNA) and G: C base pairs, with respect to the other strand. The term "complementary" is not limited to a sequence completely complementary to at least 15 contiguous nucleotide regions, but is at least 70%, preferably at least 80%, more preferably 90%, and still more preferably 95%. What is necessary is that they have homology on the base sequence of at least%. The algorithm described in this specification may be used as an algorithm for determining homology. Such nucleotides can be used as a probe for detecting and isolating the MA of the present invention, and as a primer for amplifying the DNA of the present invention. When used as a primer, it usually has a chain length of 15 bp to 100 bp, preferably 15 bp to 35 bp. When used as a probe, a nucleotide having a chain length of at least 15 bp containing at least a part or the entire sequence of the MA of the present invention is used. Such nucleotides preferably specifically hybridize to DNA encoding the protein of the present invention. "Specifically hybridizes" means under normal hybridization conditions, preferably under stringency. The DNA that encodes the protein of the present invention (SEQ ID NOs: 9 to 16, 35 to 36, 46 to 50) hybridizes with the DNA encoding the protein of the present invention under a simple condition, and encodes another protein. NA means do not hybridize.
これらヌクレオチドは、 本発明の蛋白質の異常を検査 ·診断するために利用で きる。 例えば、 これらヌクレオチドをプローブやプライマーとして用いたノーザ ンハイブリダィゼ一シヨンや RT- PCRにより、 本発明の蛋白質をコードする DNA の発現異常を検査することができる。 また、 これらヌクレオチドをプライマーと して用いたポリメラーゼ連鎖反応 (PCR)により本発明の蛋白質をコードする DNA やその発現制御領域を増幅し、 RFLP解析、 SSCP、 シークェンシング等の方法に より、 DNA配列の異常を検査 ·診断することができる。 These nucleotides can be used for testing and diagnosing abnormalities of the protein of the present invention. For example, abnormal expression of the DNA encoding the protein of the present invention can be examined by Northern hybridization or RT-PCR using these nucleotides as probes or primers. In addition, the DNA encoding the protein of the present invention and its expression control region are amplified by polymerase chain reaction (PCR) using these nucleotides as primers, and the DNA is amplified by methods such as RFLP analysis, SSCP, and sequencing. Inspection and diagnosis of sequence abnormalities.
また、 これらヌクレオチドには、 本発明の蛋白質の発現を抑制するためのアン チセンス DNAが含まれる。 アンチセンス MAは、 アンチセンス効果を引き起こす ために、 少なくとも 15bp以上、 好ましくは 100bp、 さらに好ましくは 500bp以 上の鎖長を有し、 通常、 3000bp以内、 好ましくは 2000bp以内の鎖長を有する。 このようなアンチセンス MAには、 本発明の蛋白質の異常 (機能異常や ¾現異 常) などに起因した疾患の遺伝子治療への応用も考えられる。 該アンチセンス D NAは、 例えば、 本発明の蛋白質をコードする DNA (例えば、 配列番号: 9から 1 6、 3 5から 3 6、 4 6から 5 0 ) の配列情報を基にホスホロチォネート法 (St ein, 1988 Physicochemical properties oi phosphorotnioate oligodeoxynucle otides. Nucleic Acids Res 16, 3209-21 ( 1988)) などにより調製することが可 能である。 In addition, these nucleotides include antisense DNA for suppressing the expression of the protein of the present invention. The antisense MA has a chain length of at least 15 bp or more, preferably 100 bp, more preferably 500 bp or more, and usually has a chain length of 3000 bp or less, preferably 2000 bp or less in order to cause an antisense effect. Such antisense MA may also be applied to gene therapy for diseases caused by abnormalities (functional abnormalities or current abnormalities) of the protein of the present invention. The antisense DNA is, for example, a phosphorothionate based on the sequence information of a DNA encoding the protein of the present invention (eg, SEQ ID NOS: 9 to 16, 35 to 36, 46 to 50). Nucleic Acids Res 16, 3209-21 (1988)), etc. can be prepared by the method (Stein, 1988 Physicochemical properties oi phosphorotnioate oligodeoxynucle otides. Nucleic Acids Res 16, 3209-21 (1988)).
本発明のヌクレオチドは、 遺伝子治療に用いる場合には、 例えば、 レトロウイ ルスべクタ一、 アデノウイルスベクター、 アデノ随伴ウィルスベクターなどのゥ ィルスベクターやリボソームなどの非ウィルスベクターなどを利用して、 ex viv o法や in vivo法などにより患者へ投与を行うことが考えられる。 また、 本発明は、 本発明の蛋白質に結合する抗体を提供する。 本発明の抗体の 形態には特に制限はなく、 ポリクロ一ナル抗体やモノクローナル抗体または抗原 結合性を有するそれらの一部も含まれる。 また、 全てのクラスの抗体が含まれる さらに、 本発明の抗体には、 ヒト化抗体などの特殊抗体も含まれる。 When used for gene therapy, the nucleotides of the present invention can be used, for example, in ex vivo by using a viral vector such as a retrovirus vector, an adenovirus vector, an adeno-associated virus vector, or a non-viral vector such as a ribosome. o It may be possible to administer to patients by the method or in vivo method. The present invention also provides an antibody that binds to the protein of the present invention. The form of the antibody of the present invention is not particularly limited, and includes a polyclonal antibody, a monoclonal antibody, and a part thereof having antigen-binding properties. In addition, antibodies of all classes are included. Furthermore, the antibodies of the present invention also include special antibodies such as humanized antibodies.
本発明の抗体は、 ポリクローナル抗体の場合には、 常法に従い本発明の蛋白質 のアミノ酸配列に相当するオリゴぺプチドを合成し、 家兎に免疫することにより 得ることが可會 έで る (Current protocols in Molecular Biology edit. Ausub el et al . ( 1987) Publish. John Wiley & Sons. Section 11.12-11.13) 。 モノ クロ一ナル抗体の場合には、 常法に従い大腸菌で発現し精製した蛋白質を用いて マウスを免疫し、 その脾臓細胞と骨髄腫細胞を細胞融合させたハイプリ ドーマ細 胞を調製し、 該ハイブリ ドーマ細胞から得ることができる (Current protocols in Molecular Biology edit. Ausubel et al . ( 1987) Publish. John Wiley & S ons. Section 11.4- 11.11) 。 In the case of a polyclonal antibody, the antibody of the present invention can be obtained by synthesizing an oligonucleotide corresponding to the amino acid sequence of the protein of the present invention according to a conventional method, and immunizing a rabbit (Current protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons. Section 11.12-11.13). In the case of a monoclonal antibody, a mouse is immunized with a protein expressed and purified in Escherichia coli according to a conventional method, and a hybridoma cell obtained by fusing the spleen cell and myeloma cell thereof is prepared. It can be obtained from dorma cells (Current protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons. Section 11.4- 11.11).
本発明の蛋白質に結合する抗体は、 本発明の蛋白質の精製に加え、 例えば、 本 発明の蛋白質の発現異常や構造異常の検査 ·診断に利用することも考えられる。 具体的には、 例えば組織、 血液、 または細胞などから蛋白質を抽出し、 ウェス夕 ンブロッテイング、 免疫沈降、 ELISA等の方法による本発明の蛋白質の検出を通 して、 発現や構造の異常の有無を検査 ·診断することができる。 Antibodies that bind to the protein of the present invention may be used, for example, for the examination and diagnosis of abnormal expression or structural abnormality of the protein of the present invention, in addition to purification of the protein of the present invention. Specifically, for example, proteins are extracted from tissues, blood, or cells, and abnormalities in expression and structure are detected through detection of the proteins of the present invention by methods such as Western blotting, immunoprecipitation, and ELISA. Inspection / presence can be diagnosed.
また、 本発明の蛋白質に結合する抗体を、 本発明の蛋白質に関連した疾患の治 療などの目的に利用することも考えられる。 本発明の抗体は、 本発明の蛋白質の ァゴニストゃアン夕ゴニストとして作用し得る。 抗体を患者の治療目的で用いる 場合には、 ヒト抗体またはヒト化抗体が免疫原性の少ない点で好ましい。 ヒト抗 体は、 免疫系をヒトのものと入れ換えたマウス (例えば、 ""Functional transpl ant of megabase human immunoglobulin loci recapitulates human antibody r esponse in mice, Mendez, M.J. et al . ( 1997) Nat. Genet.15 : 146-156」 参照) に免疫することにより調製することができる。 また、 ヒト化抗体は、 モノクロ一 ナル抗体の超可変領域を用いた遺伝子組み換えによつて調製することができる(M ethods in Enzymology 203, 99-121( 1991 ) )。 It is also conceivable to use an antibody that binds to the protein of the present invention for the purpose of treating a disease associated with the protein of the present invention. The antibody of the present invention can act as an agonist of the protein of the present invention. When the antibody is used for the purpose of treating a patient, a human antibody or a humanized antibody is preferred because of its low immunogenicity. Human antibodies include mice in which the immune system has been replaced with that of a human (eg, "" Functional transpant of megabase human immunoglobulin loci recapitulates human antibody responses in mice, Mendez, MJ et al. (1997) Nat. Genet.15 : 146-156 "). In addition, humanized antibodies are It can be prepared by genetic recombination using the hypervariable region of a null antibody (Methods in Enzymology 203, 99-121 (1991)).
また、 本発明は、 本発明の蛋白質を利用した、 本発明の蛋白質に結合するリガ ンドのスクリーニング方法を提供する。 このスクリーニング方法は、 (a ) 本発 明の蛋白質またはその部分ペプチドに被検試料を接触させる工程、 (b ) 該蛋白 質またはその部分べプチドに結合する化合物を選択する工程を含む。 The present invention also provides a method for screening for a ligand that binds to the protein of the present invention, using the protein of the present invention. This screening method includes (a) a step of bringing a test sample into contact with a protein of the present invention or a partial peptide thereof, and (b) a step of selecting a compound that binds to the protein or a partial peptide thereof.
被検試料としては、 特に制限はなく、 例えば、 種々の G蛋白質共役型受容体の リガンド活性については不明の公知化合物やペプチド (例えば、 ケミカルフアイ ルに登録されているもの) あるいはファ一ジ 'ディスプレイ法 (J.Mol . Biol. ( 1 991 ) 222, 301-310) などを応用して作成されたランダム 'ペプチド群を用いる ことができる。 また、 微生物の培養上清や、 植物、 海洋生物由来の天然成分など もスクリーニングの対象となる。 その他、 脳をはじめとする生体組織抽出物、 細 胞抽出液、 遺伝子ライブラリーの発現産物などが挙げられるが、 これらに制限さ れない。 The test sample is not particularly limited. For example, known compounds or peptides whose ligand activities of various G protein-coupled receptors are unknown (for example, those registered in a chemical file) or phage A random 'peptide group created by applying a display method (J. Mol. Biol. (1991) 222, 301-310) can be used. In addition, culture supernatants of microorganisms and natural components derived from plants and marine organisms are also targets for screening. Other examples include, but are not limited to, brain and other biological tissue extracts, cell extracts, and expression products of gene libraries.
スクリーニングに用いる本発明の蛋白質は、 例えば、 細胞表面に発現した形態、 該細胞の細胞膜画分としての形態、 ァフィ二ティーカラムに結合した形態であつ てもよい。 The protein of the present invention used for screening may be, for example, a form expressed on a cell surface, a form as a cell membrane fraction of the cell, or a form bound to an affinity column.
具体的なスクリーニングの手法としては、 例えば、 本発明の蛋白質のァフィ二 ティーカラムに被検試料を接触させ本発明の蛋白質に結合する化合物を精製する 方法、 ウェストウエスタンプロッティング法など多くの公知の方法を利用するこ とができる。 これら方法を利用する場合には、 被検試料は適宜標識し、 この標識 を利用して本発明の蛋白質との結合を検出することができる。 また、 これら方法 の他に、 本発明の蛋白質を発現する細胞膜を調製して、 これをチップ上に固定し、 リガンド結合時に三量体型 GTP結合蛋白質が乖離する事を、 表面プラズモン共鳴 (surface plasmon resonance) の変ィ匕で検出する方法 (Nature Biotechnology ( 99) 17: 1105) を用いることも可能である。 また、 被検試料と本発明の蛋白質との結合活性は、 被検試料が細胞表面に発現 させた本発明の蛋白質へ結合することにより生じる細胞における変化を指標に検 出することもできる。 このような変化としては、 例えば、 細胞内の Ca2+レベルの 変化や cAMPレベルの変化が挙げられるが、 これらに制限されない。 具体的には、 G蛋白質共役型受容体に対するァゴニスト活性は GTPァ S結合法により測定でき る。 Specific screening techniques include, for example, a method of contacting a test sample with an affinity column for the protein of the present invention to purify a compound that binds to the protein of the present invention, and a number of known methods such as a West Western plotting method. Methods are available. When these methods are used, the test sample is appropriately labeled, and the binding to the protein of the present invention can be detected using the label. In addition to these methods, a cell membrane that expresses the protein of the present invention is prepared and immobilized on a chip, and the dissociation of the trimeric GTP-binding protein upon ligand binding is determined by surface plasmon resonance (surface plasmon resonance). It is also possible to use a method of detecting by resonance (Nature Biotechnology (99) 17: 1105). Further, the binding activity between the test sample and the protein of the present invention can be detected by using, as an index, a change in cells caused by binding of the test sample to the protein of the present invention expressed on the cell surface. Such changes include, but are not limited to, changes in intracellular Ca 2+ levels and changes in cAMP levels. Specifically, agonist activity for G protein-coupled receptors can be measured by the GTP-S binding method.
この方法の 1つの実施例として、 G蛋白質共役型受容体を発現させた細胞膜を 20mM HEPES (pH7.4), lOOmM NaCl , lOmM gCl2, 50 M GDP溶液中で、 35Sで標識 された GTPァ S 400pMと混合させ、 被検試料存在下と非存在下でィンキュベーシ ヨン後、 濾過 (filtration) を行い、 結合した GTPァ Sの放射活性を比較する手 法を用いることができる。 As one example of this method, 20 mM HEPES cell membranes was expressed G protein-coupled receptors (pH7.4), lOOmM NaCl, lOmM gCl 2, 50 in M GDP solution, labeled with 35 S GTP After mixing with 400 pM of αS and incubating in the presence and absence of the test sample, filtration is performed, and a method of comparing the radioactivity of the bound GTPαS can be used.
また G蛋白質共役型受容体は、 三量体型 GTP結合蛋白質の活性化を介して細胞 内にシグナルを伝達するシステムを共有している。 三量体型 GTP結合蛋白質は、 活性化する細胞内伝達系の種類によって、 Ca2+を上昇させる Gq型、 cAMPを上昇 させる Gs型、 そして cAMPを抑制する Gi型の 3種類に分類される。 このことを 応用して Gq蛋白ひサブュニットと他の G蛋白ひサブュニッ卜とをキメラ化し、 リガンドスクリ一二ングの際の陽性シグナルを Gqの細胞内伝達経路である、 Ca2 +上昇に帰結させることが可能である。 上昇した Ca2+レベルは、 TRE (TP A respon sive element) を上流に有するレポ一夕一遺伝子系、 Fluor- 3などの染色指示薬 そして蛍光蛋白 aequorinなどの変化を指標として検出ができる。 同様に、 Gs蛋 白ひサブュニッ卜と他の G蛋白ひサブュニッ卜とをキメラ化し、 陽性シグナルを Gsの細胞内伝達経路である、 cAMP上昇に帰結させ、 CRE(cAMP- responsive eleme nt)を上流に有するレポ一夕一遺伝子系での変化を指標とすることも可能である (Trends Pharmacol . Sci . (99) 20 : 118) 。 In addition, G protein-coupled receptors share a system that transmits signals into cells via activation of trimeric GTP-binding proteins. Trimeric GTP-binding proteins are classified into three types, depending on the type of intracellular signaling system that activates, Gq type that increases Ca 2+ , Gs type that increases cAMP, and Gi type that suppresses cAMP. This chimerized was Sabuyuni' Bok and non Gq protein shed Sabuyunitto and other G proteins by applying the positive signals upon ligand subscription-learning intracellular pathway of Gq, thereby resulting in Ca 2 + increase It is possible. The elevated Ca 2+ level can be detected using the repo overnight gene system having TRE (TP A responsive element) upstream, a staining indicator such as Fluor-3, and a change in the fluorescent protein aequorin as an index. Similarly, Gs protein subunits are chimerized with other G protein subunits, and a positive signal is consequently increased to cAMP, a Gs intracellular transduction pathway, and CRE (cAMP-responsive element) is upstream. It is also possible to use as an index the change in the repo overnight gene system in (Trends Pharmacol. Sci. (99) 20: 118).
このスクリーニング系において本発明の蛋白質を発現させる宿主細胞としては 特に制限はなく、 目的に応じて種々の宿主細胞が用いられるが、 例えば、 COS細 胞、 CHO細胞、 HEK293細胞などを例示することができる。 本発明の蛋白質を脊椎 動物細胞で発現させるためのベクターとしては、 本発明の蛋白質をコードする遺 伝子の上流に位置するプロモーター、 RNAのスプライス部位、 ポリアデニル化部 位および転写終結配列や複製起点等を有するものを好適に用いることができる。 例えば、 SV40の初期プロモーターを有する pSV2dhfr ( ol . Cell . Biol . ( 1981 )1, 8 54-864) や、 pEF-BOS (Nucleic Acids Res. ( 1990)18, 5322) 、 pCDM8 (Nature( 19 87)329, 840-842) 、 pCEP4 ( Invitrogen社) などは、 G蛋白質共役型受容体を発 現させるのに有用なベクターである。 ベクタ一への本発明の DNAの挿入は常法に より制限酵素サイ トを用いたリガーゼ反応により行うことができる (Current pr otocols in Molecular Biology eait. Ausubel et al . ( 1987) Publish. John W iley & Sons. Section 11.4〜; 11.11) 。 また、 宿主細胞へのベクタ一導入は、 例 えば、 リン酸カルシウム沈殿法、 電気パルス穿孔法 (Current protocols in Mol ecular Biology edit. Ausubel et al . ( 1987) Publish. John Wiley & Sons. S ection 9.1-9.9) 、 リボフェク夕ミン法 (GIBC0-BRL社製) 、 FuGENE6試薬 (ベ —リンガーマンハイム社) 、 マイクロインジェクション法などの公知の方法で行 うことが可能である。 There are no particular restrictions on the host cell that expresses the protein of the present invention in this screening system, and various host cells may be used depending on the purpose. Cells, CHO cells, HEK293 cells, and the like. Examples of the vector for expressing the protein of the present invention in vertebrate cells include a promoter located upstream of a gene encoding the protein of the present invention, an RNA splice site, a polyadenylation site, a transcription termination sequence, and an origin of replication. Those having the above can be preferably used. For example, pSV2dhfr (ol.Cell.Biol. (1981) 1, 854-864) having the early promoter of SV40, pEF-BOS (Nucleic Acids Res. (1990) 18, 5322), pCDM8 (Nature (1987) ) 329, 840-842), pCEP4 (Invitrogen) and the like are useful vectors for expressing G protein-coupled receptors. The DNA of the present invention can be inserted into a vector by a ligase reaction using a restriction enzyme site according to a conventional method (Current protocol in Molecular Biology eait. Ausubel et al. (1987) Publish. John Wiley). & Sons. Section 11.4 ~; 11.11). In addition, the introduction of a vector into a host cell can be performed, for example, by the calcium phosphate precipitation method or the electric pulse perforation method (Current protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons. Section 9.1-9.9). ), The ribofectamine method (GIBC0-BRL), the FuGENE6 reagent (Beringermannheim), the microinjection method, and the like.
上記の本発明の蛋白質に結合するリガンドのスクリーニング方法により、 リガ ンドが単離されれば、 本発明の蛋白質とリガンドの相互作用を阻害する化合物の スクリーニングが可能となる。 従って、 本発明は、 また、 本発明の蛋白質とその リガンドとの結合を阻害する活性を有する化合物のスクリーニング方法を提供す る。 このスクリーニング方法は、 (a ) 被検試料の存在下で本発明の蛋白質また はその部分べプチドにリガンドを接触させ、 該蛋白質またはその部分べプチドと リガンドとの結合活性を検出する工程、 (b ) 被検試料非存在下での結合活性と 比較して、 工程 (a ) で検出された結合活性を低下させる化合物を選択する工程、 を含む。 被検試料としては、 特に制限はなく、 例えば、 コンビナトリアル ·ケミストリ —技術 (Tetrahedron ( 1995 ) 51, 8135-8137) によって得られた化合物群、 ある いはファ一ジ 'ディスプレイ法 (J.Mol . Biol . ( 1991 ) 222, 301-310) などを応 用して作成されたランダム ·ペプチド群を用いることができる。 また、 微生物の 培養上清や、 植物、 海洋生物由来の天然成分などもスクリーニングの対象となる。 その他、 脳をはじめとする生体組織抽出物、 細胞抽出液、 遺伝子ライブラリーの 発現産物、 合成低分子化合物、 合成ペプチド、 天然化合物などが挙げられるが、 これらに制限されない。 If the ligand is isolated by the above-described method for screening a ligand that binds to the protein of the present invention, a compound that inhibits the interaction between the protein and the ligand of the present invention can be screened. Therefore, the present invention also provides a method for screening a compound having an activity of inhibiting the binding of the protein of the present invention to its ligand. This screening method comprises the steps of: (a) contacting a ligand of the protein or its partial peptide in the presence of a test sample with a ligand, and detecting the binding activity between the protein or its partial peptide and the ligand; b) selecting a compound that reduces the binding activity detected in step (a) as compared to the binding activity in the absence of the test sample. The test sample is not particularly limited. For example, a compound group obtained by combinatorial chemistry—technology (Tetrahedron (1995) 51, 8135-8137), or a phage display method (J. Mol. Biol. (1991) 222, 301-310) can be used. Screening also includes culture supernatants of microorganisms and natural components derived from plants and marine organisms. Other examples include, but are not limited to, brain and other biological tissue extracts, cell extracts, expression products of gene libraries, synthetic low molecular compounds, synthetic peptides, natural compounds, and the like.
スクリーニングに用いる本発明の蛋白質は、 例えば、 細胞表面に発現した形態、 該細胞の細胞膜画分としての形態、 あるいはァフィ二ティーカラムに結合した形 態であってもよい。 The protein of the present invention used in the screening may be, for example, in a form expressed on the cell surface, as a cell membrane fraction of the cell, or in a form bound to an affinity column.
具体的なスクリーニングの手法としては、 例えば、 リガンドを放射性同位元素 などで標識して、 被検試料の存在下において本発明の蛋白質と接触させ、 被検試 料非存在下で検出した場合と比較して、 本発明の蛋白質とリガンドとの結合活性 を低下させる化合物を、 該リガンドに付された標識を基に検出する方法を用いる ことができる。 また、 上記の本発明の蛋白質に結合するリガンドのスクリーニン グの場合と同様に、 細胞内の変化を指標にスクリーニングすることも可能である。 即ち、 本発明の蛋白質を発現する細胞に被検試料の存在下でリガンドを接触させ、 被検試料非存在下で検出した場合と比較して、 該細胞における変化を減少させる 化合物を選択することにより、 本発明の蛋白質とリガンドとの結合を阻害する化 合物をスクリーニングすることが可能である。 本発明の蛋白質を発現する細胞は、 上記した本発明の蛋白質に結合するリガンドのスクリーニングの場合と同様に調 製することができる。 このスクリーニングにより単離される化合物は、 本発明の 蛋白質のァゴニストゃアン夕ゴニス卜の候補となる。 As a specific screening method, for example, a method in which a ligand is labeled with a radioisotope or the like, and the ligand is contacted with the protein of the present invention in the presence of a test sample, and then compared with the case where detection is performed in the absence of a test sample Then, a method of detecting a compound that reduces the binding activity between the protein and the ligand of the present invention based on the label attached to the ligand can be used. In addition, as in the case of the screening of the ligand binding to the protein of the present invention, it is also possible to perform screening using changes in cells as an index. That is, a compound expressing the protein of the present invention is brought into contact with a ligand in the presence of a test sample, and a compound that reduces a change in the cell compared to the case where the ligand is detected in the absence of the test sample is selected. Thus, it is possible to screen for a compound that inhibits the binding between the protein of the present invention and a ligand. Cells expressing the protein of the present invention can be prepared in the same manner as in the above-described screening for a ligand that binds to the protein of the present invention. The compound isolated by this screening is a candidate for the agonist of the protein of the present invention.
また、 本発明は、 本発明の蛋白質の活性を阻害または促進する化合物をスクリ 一二ングする方法を提供する。 このスクリーニング方法は、 (a ) 被検試料の存 在下で本発明の蛋白質を発現する細胞に該蛋白質のリガンドを接触させる工程、 ( b ) 該リガンドの本発明の蛋白質への結合による細胞における変化を検出する 工程、 (c ) 被検試料非存在下での細胞における変化と比較して、 工程 (b ) で 検出された細胞における変化を抑制または増強させる化合物を選択する工程、 を 含む。 The present invention also provides a method for screening a compound that inhibits or promotes the activity of the protein of the present invention. This screening method is based on (a) the existence of a test sample. Contacting a cell that expresses the protein of the present invention with a ligand of the protein in the presence of the protein, (b) detecting a change in the cell due to binding of the ligand to the protein of the present invention, (c) absence of a test sample Selecting a compound that suppresses or enhances the change in the cell detected in step (b) as compared to the change in the cell below.
被検試料としては、 上記の本発明の蛋白質とリガンドとの結合を阻害する化合 物のスリ一ニング方法と同様に、 コンビナトリアル 'ケミストリー技術によって 得られた化合物群、 ファージ ·ディスプレイ法などを応用して作成されたランダ ム ·ベプチド群、 微生物の培養上清や、 植物、 海洋生物由来の天然成分、 生体組 織抽出物、 細胞抽出液、 遺伝子ライプラリーの発現産物、 合成低分子化合物、 合 成ペプチド、 天然化合物などを用いることができる。 また、 上記の本発明の蛋白 質とリガンドとの結合を阻害する化合物のスリーニングにより単離された化合物 を被検試料として用いることも可能である。 本発明の蛋白質を発現する細胞は、 上記した本発明の蛋白質に結合するリガンドのスクリーニングの場合と同様に調 製することができる。 被検試料接触後の細胞における変化は、 上記のスクリ一二 ング方法と同様に、 細胞内の Ca2+レベルや cAMPレベルの変化を指標に検出する ことができる。 また、 細胞内のシグナル伝達を検出する場合には、 ルシフェラ一 ゼなどをレポーター遺伝子とするレポ一夕一アツセィ系等の測定系を利用して検 出することも可能である。 As a test sample, a compound group obtained by a combinatorial chemistry technique, a phage display method, etc. are applied in the same manner as the above-mentioned screening method of a compound that inhibits the binding between the protein and the ligand of the present invention. Random peptides, culture supernatants of microorganisms, natural components derived from plants and marine organisms, biological tissue extracts, cell extracts, expression products of gene libraries, synthetic low molecular compounds, synthetic peptides And natural compounds. In addition, a compound isolated by screening a compound that inhibits the binding between the protein of the present invention and a ligand can be used as a test sample. Cells expressing the protein of the present invention can be prepared in the same manner as in the above-described screening for a ligand that binds to the protein of the present invention. Changes in the cells after contact with the test sample can be detected by using changes in intracellular Ca 2+ levels and cAMP levels as indices, as in the above-described screening method. When detecting intracellular signal transduction, it is also possible to detect using a measurement system such as a repo overnight system using luciferase or the like as a reporter gene.
この検出の結果、 被検試料非存在下においてリガンドを接触させた場合の細胞 における変化と比較して、 被検試料を接触させた場合における細胞における変化 が抑制されていれば、 用いた被検試料は、 本発明の蛋白質の活性を阻害する化合 物であると判定される。 逆に、 被検試料が該細胞における変化を増強させれば、 該化合物は、 本発明の蛋白質の活性を促進する化合物であると判定される。 なお、 ここでいう 「本発明の蛋白質の活性の促進または阻害する」 とは、 本発明の蛋白 質に対する直接的な作用であると、 間接的な作用であるとを問わず、 結果として 本発明の蛋白質の活性が促進または阻害されることを指す。 従って、 このスクリ —ニングにより単離される化合物には、 本発明の蛋白質またはリガンドに作用し てこれらの結合を阻害または促進することにより本発明の蛋白質の活性を阻害ま たは促進する化合物の他、 これらの結合自体を阻害または促進しないが、 結果と して本発明の蛋白質の活性を阻害または促進する化合物も含まれる。 このような 化合物には、 例えば、 本発明の蛋白質とリガンドとの結合を阻害しないが、 細胞 内のシグナル伝達経路を阻害若しくは促進する化合物が含まれる。 As a result of this detection, if the change in cells when the test sample is contacted is suppressed compared to the change in cells when the ligand is contacted in the absence of the test sample, The sample is determined to be a compound that inhibits the activity of the protein of the present invention. Conversely, if the test sample enhances the change in the cells, the compound is determined to be a compound that promotes the activity of the protein of the present invention. Here, “promoting or inhibiting the activity of the protein of the present invention” means whether the action is direct or indirect on the protein of the present invention. It means that the activity of the protein of the present invention is promoted or inhibited. Accordingly, compounds isolated by this screening include compounds that act on the protein or ligand of the present invention to inhibit or promote their binding and thereby inhibit or promote the activity of the protein of the present invention. However, compounds that do not inhibit or promote these bindings per se but result in inhibiting or promoting the activity of the protein of the present invention are also included. Such compounds include, for example, compounds that do not inhibit the binding of the protein of the present invention to the ligand, but inhibit or promote intracellular signaling pathways.
本発明のスクリーニング方法により単離される化合物を医薬品として用いる場 合には、 単離された化合物自体を直接患者に投与する以外に、 公知の製剤学的方 法により製剤化した医薬組成物として投与を行うことも可能である。 例えば、 薬 理学上許容される担体もしくは媒体、 具体的には、 滅菌水や生理食塩水、 植物油、 乳化剤、 懸濁剤などと適宜組み合わせて製剤化して投与することが考えられる。 患者への投与は、 一般的には、 例えば、 動脈内注射、 静脈内注射、 皮下注射など 当業者に公知の方法により行いうる。 投与量は、 患者の体重や年齢、 投与方法な どにより変動するが、 当業者であれば適当な投与量を適宜選択することが可能で ある。 また、 該化合物が DNAによりコードされうるものであれば、 該 DNAを遺伝 子治療用ベクターに組込み、 遺伝子治療を行うことも考えられる。 図面の簡単な説明 When a compound isolated by the screening method of the present invention is used as a pharmaceutical, the isolated compound itself is administered directly to a patient or administered as a pharmaceutical composition formulated by a known pharmaceutical method. It is also possible to do. For example, it is conceivable to administer the composition by appropriately combining it with a pharmacologically acceptable carrier or vehicle, specifically, sterile water, physiological saline, vegetable oil, emulsifier, suspension, and the like. Administration to a patient can be generally performed by methods known to those skilled in the art, such as, for example, intraarterial injection, intravenous injection, and subcutaneous injection. The dose varies depending on the weight and age of the patient, the administration method, and the like, but those skilled in the art can appropriately select an appropriate dose. In addition, if the compound can be encoded by DNA, the DNA may be incorporated into a gene therapy vector to perform gene therapy. BRIEF DESCRIPTION OF THE FIGURES
図 1は、 「GPRv4」 アミノ酸配列を rQueryj にして、 SWISS- PR0T全配列に対 する BLAST検索を行った結果を示す図である。 ORYLA PROBABLE G PR0TEIN-C0UPL ED RECEPTORに対し 31%の相同性を示した。 FIG. 1 is a diagram showing the result of performing a BLAST search on the entire sequence of SWISS-PR0T using the amino acid sequence of “GPRv4” as rQueryj. It showed 31% homology to ORYLA PROBABLE G PR0TEIN-C0UPL ED RECEPTOR.
図 2は、 rcPRvllj アミノ酸配列を Queryj にして、 SWISS- PR0T全配列に対 する BLAST検索を行った結果を示す図である。 HUMAN NEUROPEPTIDE Y RECEPTOR TYPE 2に対し 31%の相同性を示した。 図 3は、 「GPRvl3」 ァミノ酸配列を rQueryj にして、 SWISS-PROT全配列に対 する BLAST検索を行った結果を示す図である。 PONPY C5A ANAPHYLATOXIN CHEM0T ACTIC RECEPTORに対し 39%の相同性を示した。 FIG. 2 is a diagram showing the results of performing a BLAST search on the entire sequence of SWISS-PR0T using the rcPRvllj amino acid sequence as Queryj. It showed 31% homology to HUMAN NEUROPEPTIDE Y RECEPTOR TYPE 2. FIG. 3 is a diagram showing the results of performing a BLAST search on all SWISS-PROT sequences using the “GPRvl3” amino acid sequence as rQueryj. PONPY C5A showed 39% homology to ANAPHYLATOXIN CHEM0T ACTIC RECEPTOR.
図 4は、 rGPRvHj アミノ酸配列を rQueryj にして、 SWISS- PR0T全配列に対 する BLAST検索を行った結果を示す図である。 CHICK P2Y PURIN0CEPT0R 5に対 し 40%の相同性を示した。 FIG. 4 is a diagram showing the results of performing a BLAST search on the entire SWISS-PR0T sequence using the rGPRvHj amino acid sequence as rQueryj. It showed 40% homology to CHICK P2Y PURIN0CEPT0R5.
図 5は、 「GPRvl5」 のアミノ酸配列を Queryj にして、 SWISS-PROT全配列に 対する BLAST検索を行った結果を示す図である。 HUMAN 5-HYDR0XYTRYPTAMINE IE に対して 26%の相同性を示した。 FIG. 5 is a diagram showing the results of performing a BLAST search on the entire SWISS-PROT sequence using the amino acid sequence of “GPRvl5” as Queryj. It showed 26% homology to HUMAN 5-HYDR0XYTRYPTAMINE IE.
図 6は、 「GPRvl9」 のァミノ酸配列を rQueryj にして、 SWISS-PROT全配列に 対する BLAST検索を行った結果を示す図である。 APIME OPSIN, BLUE-SENSITIVE に対して 25%の相同性を示した。 FIG. 6 is a diagram showing the results of performing a BLAST search on the entire sequence of SWISS-PROT using the amino acid sequence of “GPRvl9” as rQueryj. It showed 25% homology to APIME OPSIN and BLUE-SENSITIVE.
図 7は、 「GPRv20」 のァミノ酸配列を rQueryj にして、 SWISS-PROT全配列に 対する BLAST検索を行った結果を示す図である。 RAT MAS PR0T0- ONCOGENEに対 して 38%の相同性を示した。 FIG. 7 is a diagram showing the results of performing a BLAST search on all SWISS-PROT sequences using the amino acid sequence of “GPRv20” as rQueryj. It showed 38% homology to RAT MAS PR0T0-ONCOGENE.
図 8は、 「GPRv31」 のァミノ酸配列を 「 Queryj にして、 SWISS-PROT全配列に 対する BLAST検索を行った結果を示す図である。 SHEEP THYROTROPIN- RELEASING HORMONE RECEPTORに対して 29%の相同性を示した。 Figure 8 shows the results of a BLAST search of all SWISS-PROT sequences using the amino acid sequence of “GPRv31” as “Queryj.” 29% homology to SHEEP THYROTROPIN- RELEASING HORMONE RECEPTOR showed that.
図 9は、 「GPRv38」 アミノ酸配列を rQuer j にして、 SWISS-PROT全配列に対 する BLAST検索を行った結果を示す図である。 P2Y PURIN0CEPT0R 7 (Q15722)に 対して 46%の相同性を示した。 FIG. 9 is a diagram showing the result of performing a BLAST search on the entire SWISS-PROT sequence with the amino acid sequence of “GPRv38” set to rQuer j. It showed 46% homology to P2Y PURIN0CEPT0R 7 (Q15722).
図 1 0は、 「GPRv39」 アミノ酸配列を rQueryj にして、 SWISS- PR0T全配列に 対する BLAST検索を行った結果を示す図である。 NEUROTENSIN RECEPTOR TYPE 1 (P20789)に対して 35%の相同性を示した。 図 1 1は、 「GPRv68」 アミノ酸配列を rQueryj にして、 SWISS-PROT全配列に 対する BLAST検索を行った結果を示す図である。 TYPE- IB ANGIOTENSIN I I RECEP TOR (Q13725 )に対して、 39%で最も高い相同性を示した。 FIG. 10 is a diagram showing the result of performing a BLAST search on the entire sequence of SWISS-PR0T using the amino acid sequence of “GPRv39” as rQueryj. It showed 35% homology to NEUROTENSIN RECEPTOR TYPE 1 (P20789). FIG. 11 is a diagram showing the results of performing a BLAST search on the entire SWISS-PROT sequence using the amino acid sequence of “GPRv68” as rQueryj. 39% showed the highest homology to TYPE-IB ANGIOTENSIN II RECEPTOR (Q13725).
図 1 2は、 「GPRv77」 アミノ酸配列を rQueryj にして、 SWISS- PR0T全配列に 対する BLAST検索を行った結果を示す図である。 HUMAN PUTATIVE G PROTEIN-COU PLED RECEPTOR GPR17 (R12) (Q13304)に対して、 29°で最も高い相同性を示した。 図 1 3は、 「GPRv78」 アミノ酸配列を rQueryj にして、 SWISS-PROT全配列に 対する BLAST検索を行った結果を示す図である。 HUMAN GALANIN RECEPTOR TYPE 2 (043603)に対して、 39%で最も高い相同性を示した。 FIG. 12 is a diagram showing the result of performing a BLAST search on the entire SWISS-PR0T sequence using the amino acid sequence of “GPRv77” as rQueryj. It showed the highest homology at 29 ° to HUMAN PUTATIVE G PROTEIN-COU PLED RECEPTOR GPR17 (R12) (Q13304). FIG. 13 is a diagram showing the results of performing a BLAST search on the entire SWISS-PROT sequence using the amino acid sequence of “GPRv78” as rQueryj. 39% showed the highest homology with HUMAN GALANIN RECEPTOR TYPE 2 (043603).
図 1 4は、 「GPRv79」 アミノ酸配列を rQueryj にして、 SWISS-PROT全配列に 対する BLAST検索を行った結果を示す図である。 RAT MAS PR0T0-0NC0GENE (P125 26 )に対して、 39%で最も高い相同性を示した。 FIG. 14 is a diagram showing the results of performing a BLAST search on the entire SWISS-PROT sequence using the amino acid sequence of “GPRv79” as rQueryj. 39% showed the highest homology to RAT MAS PR0T0-0NC0GENE (P125 26).
図 1 5は、 「GPRv81」 アミノ酸配列を rQueryj にして、 SWISS- PR0T全配列に 対する BLAST検索を行った結果を示す図である。 HUMAN 5-HYDR0XYTRYPTAMINE IB RECEPTOR (P28222 )に対して、 25%で最も高い相同性を示した。 発明を実施するための最良の形態 FIG. 15 is a diagram showing the results of performing a BLAST search on the entire sequence of SWISS-PR0T using the amino acid sequence of “GPRv81” as rQueryj. 25% showed the highest homology to HUMAN 5-HYDR0XYTRYPTAMINE IB RECEPTOR (P28222). BEST MODE FOR CARRYING OUT THE INVENTION
次に、 本発明を実施例によりさらに具体的に説明するが、 本発明は下記実施例 に限定されるものではない。 なお、 特に断りがない場合は、 公知の方法 (Maniat is, T. at al . ( 1982 ) :" Molecular Cloning - A Laboratory Manual" Cold Spr ing Harbor Laboratory, NY) に従って実施可能である。 Next, the present invention will be described more specifically with reference to examples, but the present invention is not limited to the following examples. Unless otherwise specified, it can be performed according to a known method (Maniat is, T. at al. (1982): "Molecular Cloning-A Laboratory Manual" Cold Spring Harbor Laboratory, NY).
[実施例 1 ] 新規 G蛋白質共役型受容体をコードする遺伝子の単離 [Example 1] Isolation of a gene encoding a novel G protein-coupled receptor
本発明の新規 G蛋白質共役型受容体 (GPRv4, GPRvll , GPRvl3, GPRvH, GPRvl 5, GPRvl9, GPRv20, GPRv31 , GPRv38, GPRv39, GPRv68, GPRv77, GPRv78, GPRv7 9, GPRv81) をコードする全長 cDNAは、 PCRにより取得した。 新規 G蛋白質共役型受容体 GPRv4の増幅にはヒト胎児脳由来の Marathon Read y cDNA (Clontech社) を鎵型 cDNAに、 フォワードプライマ一として 5, - ATGGCCA ACTCCACAGGGCTGAACGCCT-3' (配列番号: 1 7 ) 、 リバースプライマーとして 5, - TCAGGAGAGAGAACTCTCAGGTGGCCCC-3' (配列番号: 1 8 ) を用いた。 PCRは Pyrobes t DNA polymerase (宝酒造) を用い 5% ホルムアミ ド存在下で、 94°C (2分) の 後、 98°C (30秒) /65°C (30秒) /75°C (2分) のサイクルを 30回繰り返した。 その結果、 約 1.1 kbpの DNA断片が増幅された。 この断片を pCR2.1 plasmid ( I nvitrogen社) を用いてクローニングした。 得られたクローンの塩基配列はジデ ォキシ夕一ミネ一夕一法により ABI377 DNA Sequencer (Applied Biosystems 社) を用いて解析した。 明らかになった配列を配列番号: 9に示す。 The full length cDNA encoding the novel G protein-coupled receptor of the present invention (GPRv4, GPRvll, GPRvl3, GPRvH, GPRvl 5, GPRvl9, GPRv20, GPRv31, GPRv38, GPRv39, GPRv68, GPRv77, GPRv78, GPRv7 9, GPRv81) Obtained by PCR. To amplify the novel G protein-coupled receptor GPRv4, Marathon Ready cDNA derived from human fetal brain (Clontech) is used as type II cDNA, and as a forward primer, 5,-ATGGCCA ACTCCACAGGGCTGAACGCCT-3 '(SEQ ID NO: 17) 5, -TCAGGAGAGAGAACTCTCAGGTGGCCCC-3 '(SEQ ID NO: 18) was used as a reverse primer. PCR was carried out using Pyrobes tDNA polymerase (Takara Shuzo) in the presence of 5% formamide, after 94 ° C (2 minutes), 98 ° C (30 seconds) / 65 ° C (30 seconds) / 75 ° C (2 Min) cycle was repeated 30 times. As a result, a DNA fragment of about 1.1 kbp was amplified. This fragment was cloned using pCR2.1 plasmid (Invitrogen). The nucleotide sequence of the obtained clone was analyzed using the ABI377 DNA Sequencer (Applied Biosystems) according to the didoxy-one-mine method. The elucidated sequence is shown in SEQ ID NO: 9.
同配列は 1107塩基のオープンリーディングフレーム (配列番号: 9の第 1番 目から第 1107番目) を持っている。 オープンリーディングフレームから予測さ れるアミノ酸配列 (368アミノ酸) を配列番号: 1に示す。 予想アミノ酸配列は、 G蛋白質共役型受容体の特徴である 7個の膜貫通ドメインと思われる疎水性領域 を有していることから、 本遺伝子が G蛋白質共役型受容体をコードすることが判 明した。 This sequence has an open reading frame of 1107 bases (from 1st to 1107th of SEQ ID NO: 9). The amino acid sequence (368 amino acids) predicted from the open reading frame is shown in SEQ ID NO: 1. The predicted amino acid sequence has a hydrophobic region that is thought to be seven transmembrane domains that are characteristic of G protein-coupled receptors, indicating that this gene encodes a G protein-coupled receptor. Revealed.
新規 G蛋白質共役型受容体 GPRvllの増幅にはヒト胎児由来の Marathon Ready cDNA (Clontech社) を錄型 cDNAに、 フォワードプライマーとして 5, - ATGCAGGC GCTTAACATTACCCCGGAGC-3' (配列番号: 1 9 ) 、 リバースプライマーとして 5, -T TAATGCCCACTGTCTAAAGGAGAATTC-3' (配列番号: 2 0 ) を用いた。 PCRは Pyrobest DNA polymerase (宝酒造社) を用い 5% ホルムアミ ド存在下で、 94°C (2.5分) の後、 94°C (5秒) /72°C (2分) のサイクルを 5回、 94°C (5秒) /70°C (2 分) のサイクルを 5回、 94°C (5秒) /68°C (2分) のサイクルを 25回繰り返し た。 その結果、 約 1.2 kbpの DNA断片が増幅された。 この断片を pCR2.1 plasmi d ( Invitrogen社) を用いてクローニングした。 得られたクローンの塩基配列は dideoxy terminator法により ABI377 DNA Sequencer (Appl ied Biosystems社) を用いて解析した。 明らかになった配列を配列番号: 1 0に示す。 To amplify the novel G protein-coupled receptor GPRvll, Marathon Ready cDNA (Clontech) derived from human fetus is used as type II cDNA, and as a forward primer, 5,-ATGCAGGC GCTTAACATTACCCCGGAGC-3 '(SEQ ID NO: 19), reverse primer 5, -T TAATGCCCACTGTCTAAAGGAGAATTC-3 '(SEQ ID NO: 20) was used. PCR was performed using Pyrobest DNA polymerase (Takara Shuzo) in the presence of 5% formamide, followed by five cycles of 94 ° C (2.5 seconds), followed by 94 ° C (5 seconds) / 72 ° C (2 minutes). The cycle of 94 ° C (5 seconds) / 70 ° C (2 minutes) was repeated 5 times, and the cycle of 94 ° C (5 seconds) / 68 ° C (2 minutes) was repeated 25 times. As a result, a DNA fragment of about 1.2 kbp was amplified. This fragment was cloned using pCR2.1 plasmid (Invitrogen). The nucleotide sequence of the clone obtained is Analysis was performed using the ABI377 DNA Sequencer (Applied Biosystems) by the dideoxy terminator method. The elucidated sequence is shown in SEQ ID NO: 10.
同配列は 1296塩基のオープンリ一ディングフレーム (配列番号: 1 0の第 1 番目から第 1296番目) を持っている。 オープンリーディングフレームから予測 されるアミノ酸配列 (431アミノ酸) を配列番号: 2に示す。 予想アミノ酸配列 は、 G蛋白質共役型受容体の特徴である 7個の膜貫通ドメインと思われる疎水性 領域を有していることから、 本遺伝子が G蛋白質共役型受容体をコードすること が判明した。 This sequence has an open reading frame of 1296 bases (1st to 1296th of SEQ ID NO: 10). The amino acid sequence (431 amino acids) predicted from the open reading frame is shown in SEQ ID NO: 2. The predicted amino acid sequence has a hydrophobic region that seems to be seven transmembrane domains characteristic of G protein-coupled receptors, indicating that this gene encodes a G protein-coupled receptor did.
新規 G蛋白質共役型受容体 GPRvl3の増幅にはヒト胎盤由来の Marathon Ready cDNA (Clontech社) を錶型 cDNAに、 フォワードプライマ一として 5, - ATGGGGM CGATTCTGTCAGCTACGAGT-3' (配列番号: 2 1 ) 、 リバースプライマーとして 5, -C TACACCTCCATCTCCGAGACCAGGTCA-3' (配列番号: 2 2 ) を用いた。 PCRは Pyrobest DNA polymerase (宝酒造社) を用い 5 ホルムアミ ド存在下で、 94°C (2.5分) の後、 94°C (5秒) /72°C (2分) のサイクルを 5回、 94°C (5秒) /70°C (2 分) のサイクルを 5回、 94°C (5秒) /68°C (2分) のサイクルを 25回繰り返し た。 その結果、 約 1. 1 kbpの DNA断片が増幅された。 この断片を pCR2. 1 plasmi d ( Invitrogen社) を用いてクローニングした。 得られたクローンの塩基配列は ジデォキシ夕一ミネ一夕一法により ABI377 DNA Sequencer (Appl ied Biosystem s社) を用いて解析した。 明らかになった配列を配列番号: 1 1に示す。 To amplify the novel G protein-coupled receptor GPRvl3, Marathon Ready cDNA (Clontech) derived from human placenta was used as type I cDNA, and as a forward primer, 5,-ATGGGGM CGATTCTGTCAGCTACGAGT-3 '(SEQ ID NO: 21), reverse 5, -C TACACCTCCATCTCCGAGACCAGGTCA-3 '(SEQ ID NO: 22) was used as a primer. PCR was performed using Pyrobest DNA polymerase (Takara Shuzo Co., Ltd.) in the presence of 5 formamide, followed by five cycles of 94 ° C (2.5 minutes) followed by 94 ° C (5 seconds) / 72 ° C (2 minutes). The cycle of 5 ° C (5 seconds) / 70 ° C (2 minutes) was repeated 5 times, and the cycle of 94 ° C (5 seconds) / 68 ° C (2 minutes) was repeated 25 times. As a result, a DNA fragment of about 1.1 kbp was amplified. This fragment was cloned using pCR2.1 plasmid (Invitrogen). The nucleotide sequence of the obtained clone was analyzed using the ABI377 DNA Sequencer (Applied Biosystems) by the dideoxy-mine-one-one method. The elucidated sequence is shown in SEQ ID NO: 11.
同配列は 1014塩基のオープンリーディングフレーム (配列番号: 1 1の第 1 番目から第 1014番目) を持っている。 オープンリーディングフレームから予測 されるアミノ酸配列 (337アミノ酸) を配列番号: 3に示す。 予想アミノ酸配列 は、 G蛋白質共役型受容体の特徴である 7個の膜貫通ドメインと思われる疎水性 領域を有していることから、 本遺伝子が G蛋白質共役型受容体をコードすること が判明した。 新規 G蛋白質共役型受容体 GPRvl4の増幅にはヒト胎児脳由来の Marathon Rea dy cDNA (Clontech社) を銪型 cDNAに、 フォワードブライマ一として 5, -ATGTTA GCCAACAGCTCCTCAACCAACA-3' (配列番号: 2 3 ) 、 リバースプライマーとして 5, -TCAGAGGGCGGAATCCTGGGGACACTGT-3' (配列番号: 2 4 ) を用いた。 PCRは Pyrobe st DNA polymerase (宝酒造社) を用い 5%ホルムアミ ド存在下で 94°C (2.5分) の後、 94°C (5秒) /72°C (2分) のサイクルを 5回、 94°C (5秒) /70°C (2 分) のサイクルを 5回、 94°C (5秒) /68°C (2分) のサイクルを 25回繰り返し た。 その結果、 約 1.1 kbpの DNA断片が増幅された。 この断片を pCR2.1 plasmi d ( Invitrogen社) を用いてクローニングした。 得られたクローンの塩基配列は ジデォキシ夕一ミネ一夕一法により ABI377 DNA Sequencer (Applied Biosystem s社) を用いて解析した。 明らかになった配列を配列番号: 1 2に示す。 The sequence has an open reading frame of 1014 bases (1st to 1014th of SEQ ID NO: 11). The amino acid sequence (337 amino acids) predicted from the open reading frame is shown in SEQ ID NO: 3. The predicted amino acid sequence has a hydrophobic region that seems to be seven transmembrane domains characteristic of G protein-coupled receptors, indicating that this gene encodes a G protein-coupled receptor did. To amplify the novel G protein-coupled receptor GPRvl4, Marathon Ready cDNA (Clontech) derived from human fetal brain was used as type I cDNA, and as a forward primer 5, -ATGTTA GCCAACAGCTCCTCAACCAACA-3 '(SEQ ID NO: 23) ), 5, -TCAGAGGGCGGAATCCTGGGGACACTGT-3 ′ (SEQ ID NO: 24) was used as the reverse primer. PCR was performed using Pyrobe st DNA polymerase (Takara Shuzo) in the presence of 5% formamide at 94 ° C (2.5 minutes), followed by 5 cycles of 94 ° C (5 seconds) / 72 ° C (2 minutes). The cycle of 94 ° C (5 seconds) / 70 ° C (2 minutes) was repeated 5 times, and the cycle of 94 ° C (5 seconds) / 68 ° C (2 minutes) was repeated 25 times. As a result, a DNA fragment of about 1.1 kbp was amplified. This fragment was cloned using pCR2.1 plasmid (Invitrogen). The nucleotide sequence of the obtained clone was analyzed using the ABI377 DNA Sequencer (Applied Biosystems) by the dideoxy-mine-one-one method. The elucidated sequence is shown in SEQ ID NO: 12.
同配列は 1119塩基のオープンリーディングフレーム (配列番号: 1 2の第 1 番目から第 1119番目) を持っている。 オープンリーディングフレームから予測 されるアミノ酸配列 (372アミノ酸) を配列番号: 4に示す。 予想アミノ酸配列 は、 G蛋白質共役型受容体の特徴である 7個の膜貫通ドメインと思われる疎水性 領域を有していることから、 本遺伝子が G蛋白質共役型受容体をコードすること が判明した。 This sequence has an open reading frame of 1119 bases (1st to 1119th of SEQ ID NO: 12). The amino acid sequence (372 amino acids) predicted from the open reading frame is shown in SEQ ID NO: 4. The predicted amino acid sequence has a hydrophobic region that seems to be seven transmembrane domains characteristic of G protein-coupled receptors, indicating that this gene encodes a G protein-coupled receptor did.
新規 G蛋白質共役型受容体 GPRvl5の増幅にはヒト胎児脳由来の Marathon Rea dy cDNA (Clontech社) を錡型 cDNAに、 フォワードブライマ一として 5, -ATGAGT GATGAGCGGCGGCTGCCTGGCAG-3' (配列番号: 2 5 ) 、 リバースプライマ一として 5 ' -CTAGGACGCGGAGCCCAGCGAGTCCGAG-3' (配列番号 : 2 6 ) を用いた。 PCRは Pyrob est DNA polymerase (宝酒造) を用い 5% ホルムアミ ド存在下で、 94°C (2.5 分) の後、 98°C (5秒) /72°C (4分) のサイクルを 5回、 98°C (5秒) /70°C (4分) のサイクルを 5回、 98°C (5秒) /68°C (4分) のサイクルを 25回繰り 返した。 その結果、 約 1.8 kbpの DNA断片が増幅された。 この断片を pCR2. 1 pi asmid ( Invitrogen社) を用いてクロ一ニングした。 得られたクローンの塩基配 列はジデォキシ夕一ミネ一夕一法により ABI377 DNA Sequencer (Applied Biosy stems社) を用いて解析した。 明らかになった配列を配列番号: 1 3に示す。 同配列は 1830塩基のオープンリーディングフレーム (配列番号: 1 3 ) を持 つている。 オープンリーディングフレームから予測されるアミノ酸配列 (609ァ ミノ酸) を配列番号: 5に示す。 予想アミノ酸配列は、 G蛋白質共役型受容体の 特徴である 7個の膜貫通ドメインと思われる疎水性領域を有していることから、 本遺伝子が G蛋白質共役型受容体をコードすることが判明した。 To amplify the novel G protein-coupled receptor GPRvl5, Marathon Ready cDNA (Clontech) derived from human fetal brain was used as type I cDNA, and as a forward primer 5, -ATGAGT GATGAGCGGCGGCTGCCTGGCAG-3 '(SEQ ID NO: 25) ), 5′-CTAGGACGCGGAGCCCAGCGAGTCCGAG-3 ′ (SEQ ID NO: 26) was used as the reverse primer. PCR was performed using Pyrobest DNA polymerase (Takara Shuzo) in the presence of 5% formamide, followed by five cycles of 98 ° C (5 seconds) / 72 ° C (4 minutes) after 94 ° C (2.5 minutes). The cycle of 98 ° C (5 seconds) / 70 ° C (4 minutes) was repeated 5 times, and the cycle of 98 ° C (5 seconds) / 68 ° C (4 minutes) was repeated 25 times. As a result, a DNA fragment of about 1.8 kbp was amplified. This fragment was cloned using pCR2.1 pi asmid (Invitrogen). Base sequence of the clone obtained The columns were analyzed using the ABI377 DNA Sequencer (Applied Biosystems) according to the dideoxy and mineral method. The elucidated sequence is shown in SEQ ID NO: 13. This sequence has an open reading frame of 1830 bases (SEQ ID NO: 13). The amino acid sequence (609 amino acid) predicted from the open reading frame is shown in SEQ ID NO: 5. The predicted amino acid sequence has a hydrophobic region that is thought to be the seven transmembrane domains characteristic of a G protein-coupled receptor, indicating that this gene encodes a G protein-coupled receptor did.
新規 G蛋白質共役型受容体 GPRvl 9の増幅にはヒト胎盤由来の Marathon Ready cDNA (Clontech社) を铸型 cDMに、 フォワードプライマ一として 5, - ATGATGGG ACTCACCGAGGGGGTGTTCC-3' (配列番号: 2 7 ) 、 リバースプライマ一として 5, - C TAAGAGAAAATGGGTCCCTTGGATCCAG-3' (配列番号: 2 8 ) を用いた。 PCRは Pyrobes t DNA polymerase (宝酒造) を用い、 94°C (2分) の後、 94°C (30秒) /55°C To amplify the novel G protein-coupled receptor GPRvl9, Marathon Ready cDNA (Clontech) derived from human placenta was used for type III cDM, and as a forward primer, 5,-ATGATGGG ACTCACCGAGGGGGTGTTCC-3 '(SEQ ID NO: 27), 5, -C TAAGAGAAAATGGGTCCCTTGGATCCAG-3 '(SEQ ID NO: 28) was used as the reverse primer. PCR using Pyrobes t DNA polymerase (Takara Shuzo), 94 ° C (2 minutes), 94 ° C (30 seconds) / 55 ° C
(30秒) /72°C (2分) のサイクルを 30回繰り返した。 その結果、 約 1.0 kbp の DNA断片が増幅された。 この断片を pCR2. 1 plasmid ( Invitrogen社) を用い てクロ一ニングした。 得られたクロ一ンの塩基配列はジデォキシ夕一ミネ一夕一 法により ABI377 DNA Sequencer (Appl ied Biosystems社) を用いて解析した。 明らかになつた配列を配列番号: 1 4に示す。 (30 seconds) / 72 ° C (2 minutes) cycle was repeated 30 times. As a result, a DNA fragment of about 1.0 kbp was amplified. This fragment was cloned using pCR2.1 plasmid (Invitrogen). The nucleotide sequence of the resulting clone was analyzed using the ABI377 DNA Sequencer (Applied Biosystems) according to the dideoxy and mineral method. The sequence identified is shown in SEQ ID NO: 14.
同配列は 951塩基のオープンリ一ディングフレーム (配列番号: 1 4 ) を持つ ている。 オープンリーディングフレームから予測されるアミノ酸配列 (316アミ ノ酸) を配列番号: 6に示す。 予想アミノ酸配列は、 G蛋白質共役型受容体の特 徴である 7個の膜貫通ドメインと思われる疎水性領域を有していることから、 本 遺伝子が G蛋白質共役型受容体をコードすることが判明した。 This sequence has an open reading frame of 951 bases (SEQ ID NO: 14). The amino acid sequence (316 amino acid) predicted from the open reading frame is shown in SEQ ID NO: 6. Since the predicted amino acid sequence has seven transmembrane domains that are likely to be characteristic of G protein-coupled receptors, the gene may encode a G protein-coupled receptor. found.
新規 G蛋白質共役型受容体 GPRv20の増幅にはヒト胎児由来の Marathon Ready cDNA (Clontech社) を鎵型 cDNAに、 フォワードプライマ一として 5, - ATGGATCC AACCATCTCAACCTTGGACAC-3' (配列番号: 2 9 ) 、 リバースプライマーとして 5, - TCAGGTTAGATAAACATCTATTTGAAGAC-3' (配列番号: 3 0 ) を用いた。 PCRは Pyrobe st DNA polymerase (宝酒造) を用い、 5% ホルムアミ ド存在下で、 94°C (2.5 分) の後、 94°C (5秒) /72°C (4分) のサイクルを 5回、 94°C (5秒) /70°CTo amplify the novel G protein-coupled receptor GPRv20, Marathon Ready cDNA (Clontech) derived from human fetus is used as type I cDNA, and as a forward primer, 5,-ATGGATCC AACCATCTCAACCTTGGACAC-3 '(SEQ ID NO: 29), reverse 5, -TCAGGTTAGATAAACATCTATTTGAAGAC-3 '(SEQ ID NO: 30) was used as a primer. PCR Pyrobe Using st DNA polymerase (Takara Shuzo), 5 cycles of 94 ° C (5 seconds) / 72 ° C (4 minutes) after 94 ° C (2.5 minutes) in the presence of 5% formamide, 94 ° C C (5 seconds) / 70 ° C
(4分) のサイクルを 5回、 94°C (5秒) /68°C (4分) のサイクルを 25回繰り 返した。 その結果、 約 1. 1 kbpの MA断片が増幅された。 この断片を pCR2.1 pi asmid ( Invitrogen社) を用いてクローニングした。 得られたクローンの塩基配 列はジデォキシ夕一ミネ一夕一法により ABI377 DNA Sequencer (Applied Biosy stems社) を用いて解析した。 明らかになった配列を配列番号: 1 5に示す。 同配列は 1116塩基のオープンリーディングフレーム (配列番号: 1 5 ) を持 つている。 オープンリーディングフレームから予測されるアミノ酸配列 (322ァ ミノ酸) を配列番号: 7に示す。 予想アミノ酸配列は、 G蛋白質共役型受容体の 特徴である 7個の膜貫通ドメインと思われる疎水性領域を有していることから、 本遺伝子が G蛋白質共役型受容体をコードすることが判明した。 (4 min) cycle was repeated 5 times, and 94 ° C (5 sec) / 68 ° C (4 min) cycle was repeated 25 times. As a result, a MA fragment of about 1.1 kbp was amplified. This fragment was cloned using pCR2.1 pi asmid (Invitrogen). The nucleotide sequence of the obtained clone was analyzed using the ABI377 DNA Sequencer (Applied Biosystems) by the dideoxy-one-mine method. The elucidated sequence is shown in SEQ ID NO: 15. This sequence has an open reading frame of 1116 bases (SEQ ID NO: 15). The amino acid sequence (322 amino acids) predicted from the open reading frame is shown in SEQ ID NO: 7. The predicted amino acid sequence has a hydrophobic region that is thought to be the seven transmembrane domains characteristic of a G protein-coupled receptor, indicating that this gene encodes a G protein-coupled receptor did.
新規 G蛋白質共役型受容体 GPRv31の増幅にはヒト胎児由来の Marathon Ready cDNA (Clontech社) を錶型 cDNAに、 フォワードプライマーとして 5, -ATGGTTGG AGACACATTAAAACTTCTG-3' (配列番号: 3 1 ) 、 リバースプライマーとして 5, -TC ATGGCATGACAACCAGATTAGGAAAG-3' (配列番号: 3 2 ) を用いた。 PCRは Pyrobest DNA polymerase (宝酒造) を用い、 94°C (2.分) の後、 94°C (30秒) /50°C (3 0秒) /72°C (2分) のサイクルを 30回繰り返した。 その結果、 約 1.1 kbpの D NA断片が増幅された。 この断片を pCR2.1 plasmid ( Invitrogen社) を用いてク ローニングした。 得られたクローンの塩基配列はジデォキシ夕一ミネ一夕一法に より ABI377 DNA Sequencer (Applied Biosystems社) を用いて解析した。 明ら かになつた配列を配列番号: 1 6に示す。 To amplify the novel G protein-coupled receptor GPRv31, Marathon Ready cDNA (Clontech) derived from human fetus is used as type I cDNA, 5, -ATGGTTGG AGACACATTAAAACTTCTG-3 '(SEQ ID NO: 31) as the forward primer, reverse primer 5, -TC ATGGCATGACAACCAGATTAGGAAAG-3 '(SEQ ID NO: 32) was used. PCR was performed using Pyrobest DNA polymerase (Takara Shuzo), followed by a cycle of 94 ° C (30 seconds) / 50 ° C (30 seconds) / 72 ° C (2 minutes) after 94 ° C (2 minutes). Repeated times. As a result, a DNA fragment of about 1.1 kbp was amplified. This fragment was cloned using pCR2.1 plasmid (Invitrogen). The nucleotide sequence of the obtained clone was analyzed using the ABI377 DNA Sequencer (Applied Biosystems) by the dideoxy-one-mine-one-one method. The clearly identified sequence is shown in SEQ ID NO: 16.
同配列は 1062塩基のオープンリーディングフレーム (配列番号: 1 6 ) を持 つている。 オープンリーディングフレームから予測されるアミノ酸配列 (353ァ ミノ酸) を配列番号: 8に示す。 予想アミノ酸配列は、 G蛋白質共役型受容体の 特徴である 7個の膜貫通ドメインと思われる疎水性領域を有していることから、 本遺伝子が G蛋白質共役型受容体をコードすることが判明した。 This sequence has an open reading frame of 1062 bases (SEQ ID NO: 16). The amino acid sequence (353 amino acid) predicted from the open reading frame is shown in SEQ ID NO: 8. The predicted amino acid sequence is the G protein-coupled receptor The presence of the characteristic seven transmembrane hydrophobic regions suggests that this gene encodes a G protein-coupled receptor.
新規 G蛋白質共役型受容体 GPRv38の増幅にはヒト脳由来の Marathon Ready c DNA (Clontech社) を鎵型 cMAに、 フォワードプライマーとして 5, - ATGTCGGTCT GCTACCGTCCCCCAGGGA-3' (配列番号: 3 7 ) 、 リバースプライマーとして 5, -TCA AAGGTCCCATTCCGGACCGTCCTTC-3' (配列番号 : 3 8 ) を用いた。 PCRは Pyrobest D NA polymerase (宝酒造) を用い、 5% ホルムアミ ド存在下で、 98° C (2.5分) の後、 98° C (5秒) /72° C (4分) のサイクルを 5回、 98° C (5秒) /70° C (4分) のサイクルを 5回、 98° C (5秒) /68° C (4分) のサイクルを 25回繰 り返した。 その結果、 約 1. 1 kbpの DNA断片が増幅された。 この断片を pCR2. 1 plasmid ( Invitrogen社) を用いてクロ一ニングした。 得られたクローンの塩基 配列はジデォキシ夕一ミネ一夕一法により ABI377 DNA Sequencer (Appl ied Bio systems社) を用いて解析した。 明らかになった配列を配列番号: 3 5に示す。 同配列は 1077塩基のオープンリーディングフレーム (配列番号: 3 5 ) を持 つている。 オープンリーディングフレームから予測されるアミノ酸配列 (358ァ ミノ酸) を配列番号: 3 3に示す。 予想アミノ酸配列は、 G蛋白質共役型受容体 の特徴である 7個の膜貫通ドメインと思われる疎水性領域を有していることから、 本遺伝子が G蛋白質共役型受容体をコードすることが判明した。 To amplify the novel G protein-coupled receptor GPRv38, a human brain-derived Marathon Ready cDNA (Clontech) was used as type I cMA, and as a forward primer, 5, -ATGTCGGTCT GCTACCGTCCCCCAGGGA-3 '(SEQ ID NO: 37) and reverse 5, -TCA AAGGTCCCATTCCGGACCGTCCTTC-3 ′ (SEQ ID NO: 38) was used as a primer. PCR was performed using Pyrobest DNA polymerase (Takara Shuzo) in the presence of 5% formamide, followed by 5 cycles of 98 ° C (2.5 minutes) followed by 98 ° C (5 seconds) / 72 ° C (4 minutes). The cycle of 98 ° C (5 seconds) / 70 ° C (4 minutes) was repeated 5 times, and the cycle of 98 ° C (5 seconds) / 68 ° C (4 minutes) was repeated 25 times. As a result, a DNA fragment of about 1.1 kbp was amplified. This fragment was cloned using pCR2.1 plasmid (Invitrogen). The nucleotide sequence of the obtained clone was analyzed using the ABI377 DNA Sequencer (Applied Biosystems) according to the dideoxy-one-one-one method. The elucidated sequence is shown in SEQ ID NO: 35. The sequence has an open reading frame of 1077 bases (SEQ ID NO: 35). The amino acid sequence (358 amino acids) predicted from the open reading frame is shown in SEQ ID NO: 33. Since the predicted amino acid sequence has a hydrophobic region that seems to be seven transmembrane domains that are characteristic of G protein-coupled receptors, it was determined that this gene encodes a G protein-coupled receptor did.
新規 G蛋白質共役型受容体 GPRv39の増幅にはヒト胎児脳由来の Marathon Rea dy cDNA (Clontech社) を錡型 cDNAに、 フォワードプライマ一として 5,- ATGTCA GGGATGGAAAAACTTCAGAATG-3' (配列番号: 3 9 ) 、 リバースプライマーとして 5, -TCAGGTTTTGTTAAAGTGGAAGCTTTGATAG-3' (配列番号: 4 0 ) を用いた。 PCRは Pyr obest DNA polymerase (宝酒造) を用い、 5% ホルムアミ ド存在下で、 94° C (2 分) の後、 94° C (30秒) /50° C (30秒) /72° C ( 1.5分) のサイクルを 35 回繰り返した。 その結果、 約 1.2 kbpの DNA断片が増幅された。 この断片を pCR 2. 1 lasmid ( Invitrogen社) を用いてクロ一ニングした。 得られたクローンの 塩基配列はジデォキシ夕一ミネ一夕一法により ABI377 DNA Sequencer (Appl ied Biosystems社) を用いて解析した。 明らかになった配列を配列番号: 3 6に示 す。 To amplify the novel G protein-coupled receptor GPRv39, Marathon Ready cDNA (Clontech) derived from human fetal brain was used as type II cDNA, and 5, -ATGTCA GGGATGGAAAAACTTCAGAATG-3 'as the forward primer (SEQ ID NO: 39) 5, -TCAGGTTTTGTTAAAGTGGAAGCTTTGATAG-3 '(SEQ ID NO: 40) was used as a reverse primer. PCR was performed using Pyr obest DNA polymerase (Takara Shuzo) in the presence of 5% formamide, after 94 ° C (2 minutes), 94 ° C (30 seconds) / 50 ° C (30 seconds) / 72 ° C ( (1.5 minutes) was repeated 35 times. As a result, a DNA fragment of about 1.2 kbp was amplified. This fragment was cloned using pCR2.1 lasmid (Invitrogen). Of the resulting clone The nucleotide sequence was analyzed using the ABI377 DNA Sequencer (Applied Biosystems) by the dideoxy-mine-one-one method. The elucidated sequence is shown in SEQ ID NO: 36.
同配列は 1248塩基のオープンリ一ディングフレーム (配列番号: 3 6 ) を持 つている。 オープンリーディングフレームから予測されるアミノ酸配列 (415ァ ミノ酸) を配列番号: 3 4に示す。 予想アミノ酸配列は、 G蛋白質共役型受容体 の特徴である 7個の膜貫通ドメインと思われる疎水性領域を有していることから、 本遺伝子が G蛋白質共役型受容体をコードすることが判明した。 This sequence has an open reading frame of 1248 bases (SEQ ID NO: 36). The amino acid sequence (415 amino acid) predicted from the open reading frame is shown in SEQ ID NO: 34. Since the predicted amino acid sequence has a hydrophobic region that seems to be seven transmembrane domains that are characteristic of G protein-coupled receptors, it was determined that this gene encodes a G protein-coupled receptor did.
新規 G蛋白質共役型受容体 GPRv68の増幅にはヒトゲノム DNA (Clontech社) を錡型 cDNAに、 フォワードプライマーとして 5, -ATGCAGATGGCCGATGCAGCCACGATA G-3' (配列番号: 5 1 ) 、 リバースプライマーとして 5,- TCAGTAGGCAGAGCTGCTGG GCAGCAGG-3' (配列番号 : 5 2 ) を用いた。 PCRは Pyrobest DNA polymerase (宝 酒造) を用い、 98° C (2. 5分) の後、 98° C (30秒) /55° C (30秒) /72° C (4分) のサイクルを 35回繰り返した。 その結果、 約 1.4 kbpの DNA断片が増 幅された。 この断片を pCR2. 1 plasmid ( Invitrogen社) を用いてクロ一ニング した。 得られたクローンの塩基配列はジデォキシ夕一ミネ一夕一法により ABI37 7 DNA Sequencer (Appl ied Biosystems社) を用いて解析した。 明らかになった 配列を配列番号: 4 6に示す。 To amplify the novel G protein-coupled receptor GPRv68, human genomic DNA (Clontech) was converted to type I cDNA, 5, -ATGCAGATGGCCGATGCAGCCACGATA G-3 '(SEQ ID NO: 51) as a forward primer, and 5, -TCAGTAGGCAGAGCTGCTGG as a reverse primer. GCAGCAGG-3 ′ (SEQ ID NO: 52) was used. For PCR, use Pyrobest DNA polymerase (Takara Shuzo), and cycle 98 ° C (30 seconds) / 55 ° C (30 seconds) / 72 ° C (4 minutes) after 98 ° C (2.5 minutes). Repeated 35 times. As a result, a DNA fragment of about 1.4 kbp was amplified. This fragment was cloned using pCR2.1 plasmid (Invitrogen). The nucleotide sequence of the obtained clone was analyzed using the ABI377 DNA Sequencer (Applied Biosystems) by the dideoxy-one-mine-one-one method. The determined sequence is shown in SEQ ID NO: 46.
同配列は 1410塩基のオープンリ一ディングフレーム (配列番号: 4 6 ) を持 つている。 オープンリーディングフレームから予測されるアミノ酸配列 (469ァ ミノ酸) を配列番号: 4 1に示す。 予想アミノ酸配列は、 G蛋白質共役型受容体 の特徴である 7個の膜貫通ドメインと思われる疎水性領域を有していることから、 本遺伝子が G蛋白質共役型受容体をコードすることが判明した。 This sequence has an open reading frame of 1410 bases (SEQ ID NO: 46). The amino acid sequence (469 amino acid) predicted from the open reading frame is shown in SEQ ID NO: 41. Since the predicted amino acid sequence has a hydrophobic region that seems to be seven transmembrane domains that are characteristic of G protein-coupled receptors, it was determined that this gene encodes a G protein-coupled receptor did.
新規 G蛋白質共役型受容体 GPRv77の増幅にはヒ卜胎児脳由来の Marathon Rea dy cDNA (Clontech社) を銃型 cDNAに、 フォワードプラィマ一として 5, -atgaac aacaatacaacatgtattcaac-3' (西己列番号: 5 3 ) 、 リノ、一スフ。ライマーとして 5, -tcaaccatatgattgcatatgtgctgaa-3' (配列番号: 5 4 ) を用いた。 PCRは Pyrobe st DNA polymerase (宝酒造) を用い、 94° C (2.5分) の後、 94° C (30秒) /5 5° C (30秒) /72° C (3分) のサイクルを 30回繰り返した。 その結果、 約 1 , 0 kbpの DNA断片が増幅された。 この断片を pCR2. 1 plasmid ( Invitrogen社) を 用いてクローニングした。 得られたクローンの塩基配列はジデォキシ夕一ミネ一 夕一法により ABI377 DNA Sequencer (Appl ied Biosystems社) を用いて解析し た。 明らかになった配列を配列番号: 4 7に示す。 To amplify the novel G protein-coupled receptor GPRv77, Marathon Ready cDNA (Clontech) derived from human fetal brain was converted into a gun-shaped cDNA, and the forward primer was 5, -atgaac aacaatacaacatgtattcaac-3 '(Nishiki Numbers: 5 3), Reno, 1 Sufu. 5, as a limer -tcaaccatatgattgcatatgtgctgaa-3 ′ (SEQ ID NO: 54) was used. PCR was performed using Pyrobe st DNA polymerase (Takara Shuzo), followed by 30 cycles of 94 ° C (30 seconds) / 55 ° C (30 seconds) / 72 ° C (3 minutes) after 94 ° C (2.5 minutes). Repeated times. As a result, a DNA fragment of about 1.0 kbp was amplified. This fragment was cloned using pCR2.1 plasmid (Invitrogen). The nucleotide sequence of the obtained clone was analyzed using the ABI377 DNA Sequencer (Applied Biosystems) by the dideoxy-mine-one-time method. The elucidated sequence is shown in SEQ ID NO: 47.
同配列は 1011塩基のオープンリーディングフレーム (配列番号: 4 7 ) を持 つている。 オープンリーディングフレームから予測されるアミノ酸配列 (336ァ ミノ酸) を配列番号: 4 2に示す。 予想アミノ酸配列は、 G蛋白質共役型受容体 の特徴である 7個の膜貫通ドメインと思われる疎水性領域を有していることから、 本遺伝子が G蛋白質共役型受容体をコードすることが判明した。 This sequence has an open reading frame of 1011 bases (SEQ ID NO: 47). The amino acid sequence (336 amino acid) predicted from the open reading frame is shown in SEQ ID NO: 42. Since the predicted amino acid sequence has a hydrophobic region that seems to be seven transmembrane domains that are characteristic of G protein-coupled receptors, it was determined that this gene encodes a G protein-coupled receptor did.
新規 G蛋白質共役型受容体 GPRv78の増幅にはヒト胎児脳由来の Marathon Rea dy cDNA (Clontech社) を錡型 cDNAに、 フォワードプライマ一として 5, -ATGCAC ACCGTGGCTACGTCCGGACCCA-3' (配列番号: 5 5 ) 、 リバースプライマーとして 5' -TCAGAGAGGGGCGTTGTCCTCCCCCAGG-3' (配列番号: 5 6 ) を用いた。 PCRは Pyrobe st DNA polymerase (宝酒造) を用い、 5% ホルムアミ ド存在下で、 98° C (2.5 分) の後、 98° C (5秒) /72° C (4分) のサイクルを 5回、 98° C (5秒) /7 0° C (4分) のサイクルを 5回、 98° C (5秒) /68° C (4分) のサイクルを 25 回繰り返した。 その結果、 約 1.2 kbpの DNA断片が増幅された。 この断片を pCR 2. 1 plasmid ( Invitrogen社) を用いてクローニングした。 得られたクローンの 塩基配列はジデォキシ夕一ミネ一夕一法により ABI377 DNA Sequencer (Appl ied For amplification of the novel G protein-coupled receptor GPRv78, Marathon Ready cDNA (Clontech) derived from human fetal brain was used as type I cDNA, and as a forward primer, 5, -ATGCAC ACCGTGGCTACGTCCGGACCCA-3 '(SEQ ID NO: 55) As a reverse primer, 5'-TCAGAGAGGGGCGTTGTCCTCCCCCAGG-3 '(SEQ ID NO: 56) was used. PCR was performed using Pyrobe st DNA polymerase (Takara Shuzo), 5 cycles of 98 ° C (5 seconds) / 98 ° C (5 seconds) / 72 ° C (4 minutes) in the presence of 5% formamide. The cycle of 98 ° C. (5 seconds) / 70 ° C. (4 minutes) was repeated 5 times, and the cycle of 98 ° C. (5 seconds) / 68 ° C. (4 minutes) was repeated 25 times. As a result, a DNA fragment of about 1.2 kbp was amplified. This fragment was cloned using pCR2.1 plasmid (Invitrogen). The nucleotide sequence of the clone obtained was determined by the ABI377 DNA Sequencer (Applied
Biosystems社) を用いて解析した。 明らかになった配列を配列番号: 4 8に示 す。 (Biosystems). The elucidated sequence is shown in SEQ ID NO: 48.
同配列は 1197塩基のオープンリーディングフレーム (配列番号: 4 8 ) を持 つている。 オープンリーディングフレームから予測されるアミノ酸配列 (398ァ ミノ酸) を配列番号: 4 3に示す。 予想アミノ酸配列は、 G蛋白質共役型受容体 の特徴である 7個の膜貫通ドメインと思われる疎水性領域を有していることから、 本遺伝子が G蛋白質共役型受容体をコードすることが判明した。 This sequence has an open reading frame of 1197 bases (SEQ ID NO: 48). Amino acid sequence predicted from open reading frame (398 Amino acid) is shown in SEQ ID NO: 43. Since the predicted amino acid sequence has a hydrophobic region that seems to be seven transmembrane domains that are characteristic of G protein-coupled receptors, it was determined that this gene encodes a G protein-coupled receptor did.
新規 G蛋白質共役型受容体 GPRv79の増幅にはヒトゲノム DNA (Clontech社) を錡型 cDNAに、 フォヮ一ドプラィマ一として 5' -atggatccaaccaccccggcctgggga a- 3, (配列番号: 5 7 ) 、 リバースプライマーとして 5, -ctacaccagactgcttctcg acatctcc- 3, (配列番号 : 5 8 ) を用いた。 PCRは Pyrobest DNA polymerase (宝 酒造) を用い、 94° C (2分) の後、 94° C (30秒) /55° C (30秒) /72° C (2. 5分) のサイクルを 30回繰り返した。 その結果、 約 1.0 kbpの DNA断片が増幅 された。 この断片を pCR2. 1 plasmid ( Invitrogen社) を用いてクロ一ニングし た。 得られたクローンの塩基配列はジデォキシターミネータ一法により ABI377 DNA Sequencer (Appl ied Biosystems社) を用いて解析した。 明らかになった配 列を配列番号: 4 9に示す。 To amplify the novel G protein-coupled receptor GPRv79, human genomic DNA (Clontech) was converted to type I cDNA, 5'-atggatccaaccaccccggcctgggga a-3 (SEQ ID NO: 57) as the primary primer, and 5,5 as the reverse primer. -ctacaccagactgcttctcg acatctcc-3, (SEQ ID NO: 58) was used. For PCR, Pyrobest DNA polymerase (Takara Shuzo) was used, and after 94 ° C (2 minutes), a cycle of 94 ° C (30 seconds) / 55 ° C (30 seconds) / 72 ° C (2.5 minutes) was performed. Repeated 30 times. As a result, a DNA fragment of about 1.0 kbp was amplified. This fragment was cloned using pCR2.1 plasmid (Invitrogen). The nucleotide sequence of the resulting clone was analyzed by the dideoxy terminator method using an ABI377 DNA Sequencer (Applied Biosystems). The determined sequence is shown in SEQ ID NO: 49.
同配列は 993塩基のオープンリーディングフレーム (配列番号: 4 9 ) を持つ ている。 オープンリーディングフレームから予測されるアミノ酸配列 (330アミ ノ酸) を配列番号: 4 4に示す。 予想アミノ酸配列は、 G蛋白質共役型受容体の 特徴である 7個の膜貫通ドメインと思われる疎水性領域を有していることから、 本遺伝子が G蛋白質共役型受容体をコードすることが判明した。 The sequence has an open reading frame of 993 bases (SEQ ID NO: 49). The amino acid sequence (330 amino acids) predicted from the open reading frame is shown in SEQ ID NO: 44. The predicted amino acid sequence has a hydrophobic region that is thought to be the seven transmembrane domains characteristic of a G protein-coupled receptor, indicating that this gene encodes a G protein-coupled receptor did.
新規 G蛋白質共役型受容体 GPRv81の増幅にはヒトゲノム DNA (Clontech社) を銪型 cDNAに、 フォワードプライマーとして 5' -ATGGGGGATGAGCTGGCACCTTGCCCT G- 3, (配列番号: 5 9 ) 、 リバースプライマーとして 5 CTAGGAAATGGTAAAGATGG CCTGGTGC-3' (配列番号 : 6 0 ) を用いた。 PCRは Pyrobest DNA polymerase (宝 酒造) を用い、 94° C (2分) の後、 94° C (30秒) /55° C (30秒) /72° C (2. 5分) のサイクルを 30回繰り返した。 その結果、 約 1.0 kbpの DNA断片が増幅 された。 この断片を pCR2. 1 plasmid ( Invitrogen社) を用いてクローニングし た。 得られたクローンの塩基配列はジデォキシ夕一ミネ一夕一法により ABI377 DNA Sequencer (Appl ied Biosystems社) を用いて解析した。 明らかになった配 列を配列番号: 5 0に示す。 To amplify the novel G protein-coupled receptor GPRv81, human genomic DNA (Clontech) is converted to type II cDNA, 5'-ATGGGGGATGAGCTGGCACCTTGCCCT G-3, (SEQ ID NO: 59) as a forward primer, and 5 CTAGGAAATGGTAAAGATGG CCTGGTGC- as a reverse primer. 3 ′ (SEQ ID NO: 60) was used. For PCR, Pyrobest DNA polymerase (Takara Shuzo) was used, and after a cycle of 94 ° C (2 minutes), a cycle of 94 ° C (30 seconds) / 55 ° C (30 seconds) / 72 ° C (2.5 minutes) was performed. Repeated 30 times. As a result, a DNA fragment of about 1.0 kbp was amplified. This fragment was cloned using pCR2.1 plasmid (Invitrogen). The nucleotide sequence of the obtained clone was determined by ABI377 Analysis was performed using a DNA Sequencer (Applied Biosystems). The elucidated sequence is shown in SEQ ID NO: 50.
同配列は 1044塩基のオープンリ一ディングフレーム (配列番号: 5 0 ) を持 つている。 オープンリーディングフレームから予測されるアミノ酸配列 (347ァ ミノ酸) を配列番号: 4 5に示す。 予想アミノ酸配列は、 G蛋白質共役型受容体 の特徴である 7個の膜貫通ドメインと思われる疎水性領域を有していることから、 本遺伝子が G蛋白質共役型受容体をコードすることが判明した。 This sequence has an open reading frame (SEQ ID NO: 50) of 1044 bases. The amino acid sequence (347 amino acids) predicted from the open reading frame is shown in SEQ ID NO: 45. Since the predicted amino acid sequence has a hydrophobic region that seems to be seven transmembrane domains that are characteristic of G protein-coupled receptors, it was determined that this gene encodes a G protein-coupled receptor did.
[実施例 2 ] 新規 G蛋白質共役型受容体のアミノ酸配列での SWISS- PR0Tに対 する BLAST検索 [Example 2] BLAST search for SWISS-PR0T using the amino acid sequence of a novel G protein-coupled receptor
「GPRv4」 のアミノ酸配列での SWISS- PR0Tに対する BLAST (Basic local al ig nment search tool) [S. F. Altschul et al . , J.Mol . Biol . , 215 : 403-410 ( 19 90) ]検索結果を図 1に示した。 「GPRv4」 は既知 G蛋白質共役型受容体の中では ORYLA PROBABLE G PROTEIN-COUPLED RECEPTOR (Q91178, 428aa)に対して 31°で 最も高い相同性を示した。 このことから 「GPRv4」 が新規 G蛋白質共役型受容体 であることが判明した。 BLAST (Basic local alignment search tool) for SWISS-PR0T in the amino acid sequence of “GPRv4” [SF Altschul et al., J. Mol. Biol., 215: 403-410 (1990)] Shown in 1. “GPRv4” showed the highest homology at 31 ° to ORYLA PROBABLE G PROTEIN-COUPLED RECEPTOR (Q91178, 428aa) among known G protein-coupled receptors. This proved that "GPRv4" was a novel G protein-coupled receptor.
「GPRvll」 のァミノ酸配列での SWISS-PR0Tに対する BLAST検索結果を図 2に 示した。 「GPRvll」 は既知 G蛋白質共役型受容体の中では HUMAN NEUROPEPTIDE Y RECEPTOR TYPE 2 (P49146, 381aa)に対して、 31°で最も高い相同性を示した。 このことから 「GPRvll」 が新規 G蛋白質共役型受容体であることが判明した。 The BLAST search results for SWISS-PR0T with the amino acid sequence of “GPRvll” are shown in FIG. “GPRvll” showed the highest homology at 31 ° to HUMAN NEUROPEPTIDE Y RECEPTOR TYPE 2 (P49146, 381aa) among known G protein-coupled receptors. This proved that "GPRvll" is a novel G protein-coupled receptor.
「GPRvl3」 のアミノ酸配列での SWISS-PR0Tに対する BLAST検索結果を図 3に 示した。 「GPRvl3」 は既知 G蛋白質共役型受容体の中では P0NPY C5A A APHYLAT OXIN CHEMOTACTIC RECEPTOR (P79234, 340aa)に対して、 39%で最も高い相同性を 示した。 このことから 「GPRvl3」 が新規 G蛋白質共役型受容体であることが判明 した。 The BLAST search results for SWISS-PR0T with the amino acid sequence of “GPRvl3” are shown in FIG. “GPRvl3” showed the highest homology at 39% to P0NPY C5A A APHYLAT OXIN CHEMOTACTIC RECEPTOR (P79234, 340aa) among known G protein-coupled receptors. This proved that “GPRvl3” is a novel G protein-coupled receptor.
「GPRvl4」 のァミノ酸配列での SWISS-PR0Tに対する BLAST検索結果を図 4に 示した。 「GPRvl4 I は既知 G蛋白質共役型受容体の中では CHICK P2Y PURIN0CEP TOR 5 (P32250, 308aa)に対して、 40%で最も高い相同性を示した。 このことから 「 GPRvl4」 が新規 G蛋白質共役型受容体であることが判明した。 BLAST search results for SWISS-PR0T with the amino acid sequence of “GPRvl4” are shown in FIG. "GPRvl4 I is a known G protein-coupled receptor that is CHICK P2Y PURIN0CEP 40% showed the highest homology to TOR 5 (P32250, 308aa). This proved that "GPRvl4" was a novel G protein-coupled receptor.
「GPRvl5」 のアミノ酸配列での SWISS- PR0Tに対する BLAST検索結果を図 5に 示した。 「GPRvl5」 は既知 G蛋白質共役型受容体の中では HUMAN 5-HYDR0XYTRYP TAMINE IE RECEPTOR (P28566, 365aa)に対して 26%で最も高い相同性を示した。 このことから 「GPRvl5」 が新規 G蛋白質共役型受容体であることが判明した。 BLAST search results for SWISS-PR0T with the amino acid sequence of “GPRvl5” are shown in FIG. “GPRvl5” showed the highest homology at 26% to HUMAN 5-HYDR0XYTRYP TAMINE IE RECEPTOR (P28566, 365aa) among known G protein-coupled receptors. This proved that “GPRvl5” is a novel G protein-coupled receptor.
「GPRvl9」 のァミノ酸配列での SWISS- PR0Tに対する BLAST検索結果を図 6に 示した。 「GPRvl9」 は既知 G蛋白質共役型受容体の中では APIME OPSIN, BLUE - S ENSITIVE (P90680, 377aa)に対して 25%で最も高い相同性を示した。 このことか ら 「GPRvl9」 が新規 G蛋白質共役型受容体であることが判明した。 The results of a BLAST search for SWISS-PR0T with the amino acid sequence of “GPRvl9” are shown in FIG. "GPRvl9" showed the highest homology at 25% among APIME OPSIN and BLUE-S ENSITIVE (P90680, 377aa) among known G protein-coupled receptors. This indicated that "GPRvl9" was a novel G protein-coupled receptor.
「GPRv20」 のァミノ酸配列での SWISS- PR0Tに対する BLAST検索結果を図 7に 示した。 「GPRv20」 は既知 G蛋白質共役型受容体の中では RAT MAS PR0T0-0NC0G ENE (P12526, 324aa)に対して 38%で最も高い相同性を示した。 このことから 「G PRv20」 が新規 G蛋白質共役型受容体であることが判明した。 BLAST search results for SWISS-PR0T with the amino acid sequence of “GPRv20” are shown in FIG. “GPRv20” showed the highest homology of 38% to RAT MAS PR0T0-0NC0G ENE (P12526, 324aa) among known G protein-coupled receptors. This indicated that “GPRv20” was a novel G protein-coupled receptor.
「GPRv31」 のァミノ酸配列での SWISS- PR0Tに対する BLAST検索結果を図 8に 示した。 「GPRv31」 は既知 G蛋白質共役型受容体の中では SHEEP THYR0TR0PIN-R ELEASING HORMONE RECEPTOR (Q28596, 398aa)に対して 29%で最も高い相同性を 示した。 このことから 「GPRv31」 が新規 G蛋白質共役型受容体であることが判明 した。 The results of a BLAST search for SWISS-PR0T with the amino acid sequence of “GPRv31” are shown in FIG. “GPRv31” showed the highest homology at 29% to SHEEP THYR0TR0PIN-R ELEASING HORMONE RECEPTOR (Q28596, 398aa) among known G protein-coupled receptors. This indicated that “GPRv31” is a novel G protein-coupled receptor.
「GPRv38」 のアミノ酸配列での SWISS- PR0Tに対する BLAST検索結果を図 9に 示した。 「GPRv38」 は既知 G蛋白質共役型受容体の中では同一なものは存在せず、 P2Y PURINOCEPTOR 7 (Q15722, 352aa)に対して 46%で最も高い相同性を示した。 このことから 「GPRv38」 が新規 G蛋白質共役型受容体であることが判明した。 FIG. 9 shows the results of a BLAST search for SWISS-PR0T using the amino acid sequence of “GPRv38”. “GPRv38” was not the same among known G protein-coupled receptors and showed the highest homology at 46% to P2Y PURINOCEPTOR 7 (Q15722, 352aa). This indicated that “GPRv38” is a novel G protein-coupled receptor.
「GPRv39」 のアミノ酸配列での SWISS- PR0Tに対する BLAST検索結果を図 1 0 に示した。 「GPRv39」 は既知 G蛋白質共役型受容体の中では同一なものは存在せ ず、 RAT NEUROTENSIN RECEPTOR TYPE 1 (P20789, 424aa)に対して 35%で最も高 い相同性を示した。 このことから 「GPRv39」 が新規 G蛋白質共役型受容体である ことが判明した。 FIG. 10 shows the results of a BLAST search for SWISS-PR0T using the amino acid sequence of “GPRv39”. “GPRv39” is the same among known G protein-coupled receptors, and is the highest at 35% of RAT NEUROTENSIN RECEPTOR TYPE 1 (P20789, 424aa) Showed high homology. This indicated that “GPRv39” is a novel G protein-coupled receptor.
「GPRv68」 のアミノ酸配列での SWISS-PROTに対する BLAST検索結果を図 1 1 に示した。 「GPRv68」 は既知 G蛋白質共役型受容体の中では同一なものは存在せ ず、 TYPE-IB ANGIOTENSIN I I RECEPTOR (Q13725, 359aa)に対して、 39°で最も高 い相同性を示した。 このことから 「GPRv68」 が新規 G蛋白質共役型受容体である ことが判明した。 BLAST search results for SWISS-PROT with the amino acid sequence of “GPRv68” are shown in FIG. “GPRv68” was not the same among known G protein-coupled receptors, and showed the highest homology at 39 ° to TYPE-IB ANGIOTENSIN II RECEPTOR (Q13725, 359aa). This indicated that “GPRv68” is a novel G protein-coupled receptor.
「GPRv77」 のアミノ酸配列での SWISS- PR0Tに対する BLAST検索結果を図 1 2 に示した。 「GPRv77」 は既知 G蛋白質共役型受容体の中では同一なものは存在せ ず、 HUMAN PUTATIVE G PROTEIN-COUPLED RECEPTOR GPR17 (R12) (Q13304, 339a a)に対して、 29%で最も高い相同性を示した。 このことから 「GPRv77」 が新規 G 蛋白質共役型受容体であることが判明した。 BLAST search results for SWISS-PR0T with the amino acid sequence of “GPRv77” are shown in FIG. `` GPRv77 '' has no known G protein-coupled receptor, and has the highest homology at 29% with HUMAN PUTATIVE G PROTEIN-COUPLED RECEPTOR GPR17 (R12) (Q13304, 339a a) showed that. This proved that “GPRv77” is a novel G protein-coupled receptor.
「GPRv78」 のアミノ酸配列での SWISS-PROTに対する BLAST検索結果を図 1 3 に示した。 「GPRv78」 は既知 G蛋白質共役型受容体の中では同一なものは存在せ ず、 HUMAN GAL AN IN RECEPTOR TYPE 2 (043603, 387aa)に対して、 39%で最も高い 相同性を示した。 このことから 「GPRv78」 が新規 G蛋白質共役型受容体であるこ とが判明した。 BLAST search results for SWISS-PROT with the amino acid sequence of “GPRv78” are shown in FIG. “GPRv78” was not the same among known G protein-coupled receptors, and showed the highest homology at 39% with HUMAN GAL IN IN RECEPTOR TYPE 2 (043603, 387aa). This proved that "GPRv78" is a novel G protein-coupled receptor.
「GPRv79」 のアミノ酸配列での SWISS- PR0Tに対する BLAST検索結果を図 1 4 に示した。 「GPRv79」 は既知 G蛋白質共役型受容体の中では同一なものは存在せ ず、 RAT MAS PROTO- ONCOGENE (P12526, 324aa)に対して、 39%で最も高い相同性 を示した。 このことから 「GPRv79」 が新規 G蛋白質共役型受容体であることが判 明した。 The BLAST search result for SWISS-PR0T with the amino acid sequence of “GPRv79” is shown in FIG. “GPRv79” was not the same as any known G protein-coupled receptor, and showed the highest homology at 39% to RAT MAS PROTO-ONCOGENE (P12526, 324aa). From this, it was determined that “GPRv79” is a novel G protein-coupled receptor.
「GPRv81」 のアミノ酸配列での SWISS- PR0Tに対する BLAST検索結果を図 1 5 に示した。 「GPRv81」 は既知 G蛋白質共役型受容体の中では同一なものは存在せ ず、 HUMAN 5-HYDROXYTRYPTAMINE IB RECEPTOR (P28222, 390aa)に対して、 25%で 最も高い相同性を示した。 このことから 「GPRv81」 が新規 G蛋白質共役型受容体 であることが判明した。 産業上の利用の可能性 The BLAST search results for SWISS-PR0T for the amino acid sequence of “GPRv81” are shown in FIG. `` GPRv81 '' is not the same among known G protein-coupled receptors, and is 25% of HUMAN 5-HYDROXYTRYPTAMINE IB RECEPTOR (P28222, 390aa). It showed the highest homology. This proved that “GPRv81” is a novel G protein-coupled receptor. Industrial applicability
本発明により、 新規 G蛋白質共役型受容体 (GPRv4, GPRvll, GPRvl3, GPRv , GPRvl5, GPRvl9, GPRv20, GPRv31, GPRv38, GPRv39, GPRv68, GPRv77, GPRv78, GPRv79, GPRv81) 、 該蛋白質をコードする遺伝子、 該遺伝子を含むベクター、 該ベクターを含む宿主細胞、 該蛋白質の製造方法が提供された。 さらに、 該蛋白 質の活性を修飾する化合物のスクリーニング方法が提供された。 本発明の蛋白質 やその遺伝子、 または本発明の蛋白質の活性を修飾する化合物は、 本発明の G蛋 白質共役型受容体蛋白質が関与する疾患の新しい予防薬や治療薬の開発への利用 が期待される。 According to the present invention, a novel G protein-coupled receptor (GPRv4, GPRvll, GPRvl3, GPRv, GPRvl5, GPRvl9, GPRv20, GPRv31, GPRv38, GPRv39, GPRv68, GPRv77, GPRv78, GPRv79, GPRv81) A vector containing the gene, a host cell containing the vector, and a method for producing the protein are provided. Furthermore, a method for screening a compound that modifies the activity of the protein was provided. The protein of the present invention, its gene, or a compound that modulates the activity of the protein of the present invention is expected to be used for the development of new preventive or therapeutic agents for diseases involving the G protein-coupled receptor protein of the present invention. Is done.
Claims
Priority Applications (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| AU22304/01A AU2230401A (en) | 1999-12-28 | 2000-12-28 | Novel guanosine triphosphate-binding protein-coupled receptors, genes thereof and production and use of the same |
Applications Claiming Priority (6)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP11/375152 | 1999-12-28 | ||
| JP37515299 | 1999-12-28 | ||
| JP2000101339 | 2000-03-31 | ||
| JP2000/101339 | 2000-03-31 | ||
| JP2000/155978 | 2000-05-23 | ||
| JP2000155978 | 2000-05-23 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2001048189A1 true WO2001048189A1 (en) | 2001-07-05 |
Family
ID=27341859
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/JP2000/009409 Ceased WO2001048189A1 (en) | 1999-12-28 | 2000-12-28 | Novel guanosine triphosphate-binding protein-coupled receptors, genes thereof and production and use of the same |
Country Status (2)
| Country | Link |
|---|---|
| AU (1) | AU2230401A (en) |
| WO (1) | WO2001048189A1 (en) |
Cited By (24)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| GB2363793A (en) * | 2000-04-10 | 2002-01-09 | Smithkline Beecham Corp | Molecular cloning of a 7TM receptor (AXOR34) |
| GB2365870A (en) * | 2000-06-01 | 2002-02-27 | Smithkline Beecham Corp | Polynucleotides and polypeptides of the G-protein coupled receptor AXOR83 |
| GB2367297A (en) * | 2000-07-07 | 2002-04-03 | Smithkline Beecham Corp | AXOR95 polypeptides and polynucleotides |
| GB2367823A (en) * | 2000-07-24 | 2002-04-17 | Smithkline Beecham Corp | AXOR28 polypeptides and polynucleotides |
| WO2002031145A1 (en) * | 2000-10-13 | 2002-04-18 | Takeda Chemical Industries, Ltd. | Novel g protein-coupled receptor protein and dna thereof |
| WO2001098330A3 (en) * | 2000-06-20 | 2002-05-02 | Euroscreen Sa | A RECOMBINANT CELL LINE EXPRESSING GPCRx11 AS A FUNCTIONAL RECEPTOR VALIDATED BY ANGIOPEPTIN AND USEFUL FOR SCREENING OF AGONISTS AND ANTAGONISTS |
| WO2002034914A1 (en) * | 2000-10-25 | 2002-05-02 | Pe Corporation (Ny) | Isolated human g-protein coupled receptors, nucleic acid molecules encoding human gpcr proteins, and uses thereof |
| WO2002000001A3 (en) * | 2000-06-30 | 2002-08-22 | Ingenium Pharmaceuticals Ag | HUMAN G PROTEIN-COUPLED RECEPTOR IGPcR20, AND USES THEREOF |
| WO2001087930A3 (en) * | 2000-05-18 | 2002-08-29 | Bayer Ag | Human galanin receptor-like g protein coupled receptor |
| WO2002072829A1 (en) * | 2001-01-31 | 2002-09-19 | Takeda Chemical Industries, Ltd. | Novel g protein-coupled receptor protein and dna thereof |
| WO2001098323A3 (en) * | 2000-06-16 | 2002-10-17 | Incyte Genomics Inc | G-protein coupled receptors |
| FR2823762A1 (en) * | 2001-04-24 | 2002-10-25 | Servier Lab | New nucleic acid encoding G protein-coupled receptor, useful for identifying specific modulators, potentially useful for treating e.g. diabetes |
| WO2002004518A3 (en) * | 2000-07-06 | 2003-01-09 | Bayer Ag | Human neuropeptide y-like g protein-coupled receptor |
| WO2003025179A1 (en) * | 2001-09-14 | 2003-03-27 | Takeda Chemical Industries, Ltd. | Novel polypeptide, dna thereof and use of the same |
| WO2003057731A3 (en) * | 2001-12-27 | 2003-10-09 | Pherin Pharm Inc | Human pheromone receptors |
| WO2003087134A2 (en) | 2002-04-12 | 2003-10-23 | Schering Corporation | G-protein coupled receptor ligands and methods |
| WO2004022086A1 (en) * | 2002-09-02 | 2004-03-18 | Takeda Pharmaceutical Company Limited | Adrenocortical hormone secretion controller |
| EP1157038A4 (en) * | 1999-02-26 | 2005-01-19 | Smithkline Beecham Corp | Cloning of a p2y-like 7tm receptor (axor17) |
| WO2005075641A1 (en) | 2004-02-09 | 2005-08-18 | Eisai R & D Management Co., Ltd. | Screening method |
| US7056685B1 (en) | 2002-11-05 | 2006-06-06 | Amgen Inc. | Receptor ligands and methods of modulating receptors |
| US7510845B2 (en) | 2000-05-04 | 2009-03-31 | California Institute Of Technology | Assay employing G protein-coupled receptor expressed in dorsal root ganglia |
| US7524638B1 (en) | 2003-06-27 | 2009-04-28 | Osi Pharmaceuticals, Inc. | Methods for identification of modulators of OSGPR114 or OSGPR78 activity, and their use in the treatment of disease |
| EP2143796A3 (en) * | 2000-02-29 | 2010-03-17 | Millennium Pharmaceuticals, Inc. | 1983, 52881, 2398, 45449, 50289, and 52872, G protein-coupled receptors and uses therefor |
| US8916355B2 (en) | 2000-05-04 | 2014-12-23 | California Institute Of Technology | Pain signaling molecules |
Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| EP0711831A2 (en) * | 1994-11-14 | 1996-05-15 | Takeda Chemical Industries, Ltd. | Rabbit G protein coupled receptor |
| WO1999061463A1 (en) * | 1998-05-29 | 1999-12-02 | Millennium Pharmaceuticals, Inc. | Novel secreted and membrane-associated proteins and uses therefor |
| WO2000031258A2 (en) * | 1998-11-20 | 2000-06-02 | Arena Pharmaceuticals, Inc. | Human orphan g protein-coupled receptors |
-
2000
- 2000-12-28 WO PCT/JP2000/009409 patent/WO2001048189A1/en not_active Ceased
- 2000-12-28 AU AU22304/01A patent/AU2230401A/en not_active Abandoned
Patent Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| EP0711831A2 (en) * | 1994-11-14 | 1996-05-15 | Takeda Chemical Industries, Ltd. | Rabbit G protein coupled receptor |
| WO1999061463A1 (en) * | 1998-05-29 | 1999-12-02 | Millennium Pharmaceuticals, Inc. | Novel secreted and membrane-associated proteins and uses therefor |
| WO2000031258A2 (en) * | 1998-11-20 | 2000-06-02 | Arena Pharmaceuticals, Inc. | Human orphan g protein-coupled receptors |
Cited By (33)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| EP1157038A4 (en) * | 1999-02-26 | 2005-01-19 | Smithkline Beecham Corp | Cloning of a p2y-like 7tm receptor (axor17) |
| US6461836B1 (en) | 1999-11-05 | 2002-10-08 | Smithkline Beecham Corporation | Molecular cloning of a 7TM receptor (AxOR34) and screening methods thereof |
| EP2143796A3 (en) * | 2000-02-29 | 2010-03-17 | Millennium Pharmaceuticals, Inc. | 1983, 52881, 2398, 45449, 50289, and 52872, G protein-coupled receptors and uses therefor |
| GB2363793A (en) * | 2000-04-10 | 2002-01-09 | Smithkline Beecham Corp | Molecular cloning of a 7TM receptor (AXOR34) |
| GB2363793B (en) * | 2000-04-10 | 2003-03-12 | Smithkline Beecham Corp | Molecular cloning of a 7TM receptor (AXOR34) and screening methods thereof |
| US8916355B2 (en) | 2000-05-04 | 2014-12-23 | California Institute Of Technology | Pain signaling molecules |
| US7510845B2 (en) | 2000-05-04 | 2009-03-31 | California Institute Of Technology | Assay employing G protein-coupled receptor expressed in dorsal root ganglia |
| WO2001087930A3 (en) * | 2000-05-18 | 2002-08-29 | Bayer Ag | Human galanin receptor-like g protein coupled receptor |
| GB2365870A (en) * | 2000-06-01 | 2002-02-27 | Smithkline Beecham Corp | Polynucleotides and polypeptides of the G-protein coupled receptor AXOR83 |
| WO2001098323A3 (en) * | 2000-06-16 | 2002-10-17 | Incyte Genomics Inc | G-protein coupled receptors |
| JP2004513614A (en) * | 2000-06-16 | 2004-05-13 | インサイト・ゲノミックス・インコーポレイテッド | G protein-coupled receptor |
| WO2001098330A3 (en) * | 2000-06-20 | 2002-05-02 | Euroscreen Sa | A RECOMBINANT CELL LINE EXPRESSING GPCRx11 AS A FUNCTIONAL RECEPTOR VALIDATED BY ANGIOPEPTIN AND USEFUL FOR SCREENING OF AGONISTS AND ANTAGONISTS |
| WO2002000001A3 (en) * | 2000-06-30 | 2002-08-22 | Ingenium Pharmaceuticals Ag | HUMAN G PROTEIN-COUPLED RECEPTOR IGPcR20, AND USES THEREOF |
| WO2002004518A3 (en) * | 2000-07-06 | 2003-01-09 | Bayer Ag | Human neuropeptide y-like g protein-coupled receptor |
| GB2367297A (en) * | 2000-07-07 | 2002-04-03 | Smithkline Beecham Corp | AXOR95 polypeptides and polynucleotides |
| GB2367823A (en) * | 2000-07-24 | 2002-04-17 | Smithkline Beecham Corp | AXOR28 polypeptides and polynucleotides |
| US7250272B2 (en) | 2000-10-13 | 2007-07-31 | Takeda Pharmaceutical Company Limited | G protein-coupled receptor protein and DNA thereof |
| WO2002031145A1 (en) * | 2000-10-13 | 2002-04-18 | Takeda Chemical Industries, Ltd. | Novel g protein-coupled receptor protein and dna thereof |
| WO2002034914A1 (en) * | 2000-10-25 | 2002-05-02 | Pe Corporation (Ny) | Isolated human g-protein coupled receptors, nucleic acid molecules encoding human gpcr proteins, and uses thereof |
| WO2002072829A1 (en) * | 2001-01-31 | 2002-09-19 | Takeda Chemical Industries, Ltd. | Novel g protein-coupled receptor protein and dna thereof |
| FR2823762A1 (en) * | 2001-04-24 | 2002-10-25 | Servier Lab | New nucleic acid encoding G protein-coupled receptor, useful for identifying specific modulators, potentially useful for treating e.g. diabetes |
| US7323541B2 (en) | 2001-09-14 | 2008-01-29 | Takeda Pharmaceutical Company Limited | Polypeptide DNA thereof and use of the same |
| WO2003025179A1 (en) * | 2001-09-14 | 2003-03-27 | Takeda Chemical Industries, Ltd. | Novel polypeptide, dna thereof and use of the same |
| WO2003057731A3 (en) * | 2001-12-27 | 2003-10-09 | Pherin Pharm Inc | Human pheromone receptors |
| US7338772B2 (en) | 2002-04-12 | 2008-03-04 | Schering Corporation | G-protein coupled receptor ligands and methods |
| WO2003087134A2 (en) | 2002-04-12 | 2003-10-23 | Schering Corporation | G-protein coupled receptor ligands and methods |
| US7807390B2 (en) | 2002-04-12 | 2010-10-05 | Schering Corporation | G-protein coupled receptor ligands and methods |
| WO2004022086A1 (en) * | 2002-09-02 | 2004-03-18 | Takeda Pharmaceutical Company Limited | Adrenocortical hormone secretion controller |
| US7056685B1 (en) | 2002-11-05 | 2006-06-06 | Amgen Inc. | Receptor ligands and methods of modulating receptors |
| US7524638B1 (en) | 2003-06-27 | 2009-04-28 | Osi Pharmaceuticals, Inc. | Methods for identification of modulators of OSGPR114 or OSGPR78 activity, and their use in the treatment of disease |
| US7879566B2 (en) | 2003-06-27 | 2011-02-01 | Osi Pharmaceuticals, Inc. | Methods for the identification of modulators of OSGPR114 or OSGPR78 activity, and their use in the treatment of disease |
| WO2005075641A1 (en) | 2004-02-09 | 2005-08-18 | Eisai R & D Management Co., Ltd. | Screening method |
| US7638490B2 (en) | 2004-02-09 | 2009-12-29 | Eisai R&D Managment Co., Ltd. | Methods of affecting feeding and weight in mammals by administration of relaxin-3 |
Also Published As
| Publication number | Publication date |
|---|---|
| AU2230401A (en) | 2001-07-09 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| WO2001048189A1 (en) | Novel guanosine triphosphate-binding protein-coupled receptors, genes thereof and production and use of the same | |
| EP1243648A1 (en) | Novel guanosine triphosphate-binding protein-coupled receptors, genes thereof and production and uses thereof | |
| JPWO2002103005A1 (en) | Guanosine triphosphate binding protein-coupled receptor | |
| US20110129477A1 (en) | NOGO Receptor Homologs | |
| WO1999046378A1 (en) | Novel g protein-coupled receptor proteins | |
| US20050221420A1 (en) | Nogo receptor homologues and their use | |
| JP2002512779A (en) | Novel G-protein coupled receptor | |
| WO2001019986A1 (en) | Peptide leukotriene receptor | |
| WO2001004299A1 (en) | AMYLOID β PROTEIN AGGLUTINATION CONTROLLING FACTOR | |
| JP2005229804A (en) | A novel melanin concentrating hormone receptor | |
| US7374895B2 (en) | Guanosine triphosphate (GTP) binding protein-coupled receptor proteins that bind histamine and are expressed in the brain | |
| JP2003284573A (en) | Guanosine triphosphate binding protein coupled receptor | |
| WO2001009323A1 (en) | Guanosine triphosphate binding protein-coupled receptors, genes thereof and production and use of the same | |
| JPWO2001048189A1 (en) | Novel guanosine triphosphate-binding protein-coupled receptors and their genes, as well as their production and use | |
| US20020037548A1 (en) | Isolated human transporter proteins, nucleic acid molecules incoding hum an transporter proteins, and uses thereof | |
| MXPA01008944A (en) | Identification of a capacitative calcium channel in antigen presenting cells and uses thereof. | |
| WO2004055186A1 (en) | Guanosine triphosphate-binding protein-coupled receptor | |
| WO2001073023A1 (en) | Novel guanosine triphosphate-binding protein-coupled receptor gprv53, gene thereof and production and use of the same | |
| JP2003125780A (en) | Novel GPCRs, genes encoding them and their uses | |
| JPWO2001009323A1 (en) | Guanosine triphosphate-binding protein-coupled receptors and their genes, as well as their production and use | |
| JPWO2003027142A1 (en) | Novel G protein coupled receptor | |
| JPWO2001048188A1 (en) | Novel guanosine triphosphate-binding protein-coupled receptors and their genes, as well as their production and use | |
| JP2009060787A (en) | Method for screening and identification of substance that suppresses degranulation reaction of mast cell mediated by Rec168, and therapeutic agent for inflammatory disease involving mast cell, comprising Rec168 antagonist | |
| WO2005100566A1 (en) | Novel monkey gpr103 and monkey qrfp and method of evaluating compound by using gpr103 | |
| JPWO2001009322A1 (en) | Guanosine triphosphate-binding protein-coupled receptors and their genes, as well as their production and use |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| AK | Designated states |
Kind code of ref document: A1 Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW |
|
| AL | Designated countries for regional patents |
Kind code of ref document: A1 Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG |
|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application | ||
| DFPE | Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101) | ||
| ENP | Entry into the national phase |
Ref country code: JP Ref document number: 2001 548702 Kind code of ref document: A Format of ref document f/p: F |
|
| REG | Reference to national code |
Ref country code: DE Ref legal event code: 8642 |
|
| 122 | Ep: pct application non-entry in european phase |