[go: up one dir, main page]

WO2003089643A1 - Novel proteins and dnas encoding the same - Google Patents

Novel proteins and dnas encoding the same Download PDF

Info

Publication number
WO2003089643A1
WO2003089643A1 PCT/JP2003/004982 JP0304982W WO03089643A1 WO 2003089643 A1 WO2003089643 A1 WO 2003089643A1 JP 0304982 W JP0304982 W JP 0304982W WO 03089643 A1 WO03089643 A1 WO 03089643A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
dna
seq
amino 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
Application number
PCT/JP2003/004982
Other languages
French (fr)
Japanese (ja)
Inventor
Yoshihide Hayashizaki
Mamoru Kamiya
Hideo Kubodera
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Mitsubishi Chemical Corp
Dnaform KK
RIKEN
Original Assignee
Mitsubishi Chemical Corp
Dnaform KK
RIKEN
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Mitsubishi Chemical Corp, Dnaform KK, RIKEN filed Critical Mitsubishi Chemical Corp
Priority to AU2003227427A priority Critical patent/AU2003227427A1/en
Publication of WO2003089643A1 publication Critical patent/WO2003089643A1/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants

Definitions

  • the present invention has introduced a novel protein, a DNA encoding the protein, a full-length cDNA encoding the protein, a recombinant vector having the DNA, an oligonucleotide comprising a partial sequence of the DNA, and the DNA.
  • the present invention relates to a transgenic cell, an antibody specifically binding to the protein, and the like. Background art
  • cDNA and analyzing its base sequence are essential for analyzing the physiological activity of a protein expressed in a living body and developing a method of using the protein based on the activity.
  • creating a library that catalogs full-length cDNAs for all genotypes is one of the important tasks of the Human Genome Project.
  • the cataloged library means that there is no duplication in the cDNAs contained in the library, and is a library containing one kind of each cDNA.
  • the full-length cDNA cloning method is described in JP-A-9-1248187 and JP-A-10-127291. In this method, a tag molecule is bound to a diol structure present at the 5 ′ cap site of the mRNA.
  • RNA-DNA complex is formed by reverse transcription using oligo dT as a primer.
  • a method comprising preparing a body, and separating a complex having a DNA corresponding to the full-length mRNA from the complex using the function of a tag molecule.
  • the full-length cDNA library produced by such a technique does not include all the elements that are different evenly as individual elements of the library. Some clones do not exist. Since a clone present in only a trace amount is likely to be novel, a subtraction method and a normalization method for enriching such clones have also been developed (Japanese Patent Application Laid-Open No. 2000-325080; Carninci, P. et al., Genomics, 37, 327-336 (1996)).
  • the present invention analyzes the nucleotide sequence of a cDNA clone contained in a cataloged full-length cDNA library, and among those having a novel sequence, identifies the physiological activity of the protein encoded by the nucleotide sequence.
  • the purpose of the present invention is to propose a method of using a protein based on activity and a DNA encoding the protein.
  • the present inventors analyzed the nucleotide sequence of a cDNA clone in a mouse full-length cDNA library and searched a database based on the homology of the sequence, and found that a protein having a specific function was found in the sequence. Was found. The expression levels of these cDNAs in each tissue were analyzed. The present invention has been achieved based on these findings.
  • a protein comprising an amino acid sequence in which several amino acids have been deleted, substituted and / or added, and having G protein-coupled receptor activity.
  • nucleotide sequence in the nucleotide sequence of any one of SEQ ID NOs: 1 to 16, the nucleotide sequence has one or several nucleotides deleted, substituted and Z or added, and has G protein-coupled receptor activity.
  • a G protein-coupled receptor having a nucleotide sequence capable of hybridizing under stringent conditions to DNA having the nucleotide sequence of any of SEQ ID NOs: 1 to 16 or a sequence complementary thereto, and DNA encoding an active protein.
  • a sense oligonucleotide having the same sequence as 5 to 100 consecutive nucleotides in the base sequence of the DNA according to any of (2) to (4) above, and having a sequence complementary to the sense oligonucleotide.
  • An oligonucleotide selected from the group consisting of an antisense oligonucleotide and an oligonucleotide derivative of the sense or antisense oligonucleotide.
  • the monoclonal / antibody is a protein according to (1) or (7) above.
  • a computer-readable recording medium storing at least one or more selected base sequence information.
  • the DNA of the present invention may be a protein or an amino acid sequence comprising the amino acid sequence of SEQ ID NOs: 17 to 32, or one or several (the number is not particularly limited; And preferably 15 or less, more preferably 10 or less, and even more preferably 5 or less) amino acid residues containing substitutions, deletions, insertions, additions, or inversions, and G Any protein can be used as long as it can encode a protein having protein-coupled receptor activity. Specifically, it may be only the translation region encoding the amino acid sequence, or may include the full length of its cDNA.
  • the DNA containing the full-length cDNA includes, for example, SEQ ID NOs: 1 to 1 And DNA comprising the nucleotide sequence according to any one of the above (6). Also, as the translation area,
  • the DNA of the present invention may be obtained by any method as long as it can be obtained. Specifically, it can be obtained by the following method, for example.
  • niRNA is prepared from a suitable animal, preferably a mammalian tissue or the like, by a method known per se and generally used.
  • the temperature is increased by using a thermostable reverse transcriptase in the presence of trehalose or the like. It is preferable to use a method of performing reverse transcription below (JP-A-10-84961).
  • high temperature means 40 to 80 ° C.
  • the cDNA thus obtained is inserted into an appropriate cloning vector for cloning.
  • the vector used here has a recombinase recognition sequence at both ends of the cloning site that can uniformly clone DNA of various chain lengths, and is a linear vector that can be inserted into the host by a method other than infection.
  • the vector JP-A-119-1273
  • JP-A-119-1273 is preferably used.
  • not all clones exist uniformly hereinafter, this may be referred to as “cataloged”.
  • a clone that exists only in a plant has a high probability of being new.
  • the nucleotide sequence of the cataloged cDNA library is analyzed by a commonly used method known per se.
  • the base sequence obtained from the sequence based on the terminal 100 is obtained by using BLAST (http://www.ncbi.nlm.nih.gov/BLAST/; National Center). of Biotechnology Information) and searched databases such as NCBI Genbank, EMBL, DDB J, and PDB.
  • the novel nucleotide sequence obtained in this way is subjected to homology search (homology search) by BLAST (Basic local alignment search tool; Altschul, SF, et al., J. Mol. Biol., 215, 403-410 (1990)). ) And HMME R (sequence analysis method using hidden Markov model; Eddy, SR, Bioinformatics 14, 755-763 (1998)). : // pfam. Wustl. Edu), etc., to estimate the function of the protein encoded by the nucleotide sequence.
  • a sufficiently significant hit sequence means that the identity between the catalytic domain portion of the registered sequence and the corresponding portion of the DNA of the present invention is less than 10 -4 as e-value, or 30 % Or more.
  • HMMPFAM analysis is performed by a method for identifying whether or not a base sequence to be analyzed has the characteristics of the base sequence of an entry in a database in which a protein profile called Pfam is accumulated. Profiles are extracted from a series of proteins with the same characteristics, and cannot be clarified by comparing one-to-one sequences over their entire length. And function prediction.
  • amino acid sequence encoded by the nucleotide sequence of SEQ ID NO: 1 is a BLAST search. Has a 99% homology with G protein coupled receptor affecting testicular descent (Great) and e-value: 0.0, 579 amino acid residues.
  • the protein encoded by the nucleotide sequence of SEQ ID NO: 1 has G protein-coupled receptor activity including Leucine Rich Repeat, and testis development based on the function of homologous proteins. It is inferred that it has the function of maintaining the function.
  • More PROBABLE G PROTEIN-COUPLED RECEPTOR GPR72 PRECURSOR amino acid sequence encoded by the nucleotide sequence set forth in SEQ ID NO: 2 in BLAST searches (GLUCOCORTICOID - INDUCED RECEPTOR) and e- value is 5 X 1 0- 136, 2 4 0 Amino acid residue has a 1 0 0% homology with groups, also the orphan G- protein coupled receptor GPR72, e- va 1 ue: in 1 X 1 0- 84, with 4 5% degree of coincidence over the 301 amino acid residues a further glucocorticoid- induced receptor, e- value: in 3 X 1 0 one 84, has a 4 4% homology over 3 0 1 ⁇ amino acid residues.
  • the protein encoded by the nucleotide sequence shown in SEQ ID NO: 2 is a G protein-coupled receptor that may be induced by dalcocorticoid. Highly homologous proteins are upregulated when antipsychotics are stopped.
  • the amino acid sequence encoded by the nucleotide sequence of SEQ ID NO: 3 was More PROBABLE G- PROTEIN - COUPLED RECEPTOR GPR72 PRECURSOR (GLUCOCORTICOID- INDUCED RECEPTOR and by 3 ⁇ 4 100% homology with 5 X 10- 96 of e- value, 1 73 Amino acid residues, Si was orphan G - protein coupled receptor and GPR72, e- va 1 ue: with 2 X 10- 84, it has a 45% homology over 301 amino acid residues, even more, glucocorticoid - and induced receptor, e- va 1 ue: 4X 10 -84 And has 4'4% homology over 301 amino acid residues.
  • a protein characteristic search using HMMP FAM was performed on the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 3.
  • a sequence showing the characteristics of the G protein-coupled receptor (base sequence that is entered as 7tm_lJ in P f am) It can be inferred that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 3 has a G-protein-coupled receptor activity that can be induced by dalcocotide. Higher proteins are upregulated when antipsychotics are stopped.
  • Amino acid sequence nucleotide sequence is co one de set forth in SEQ ID NO: 6 in B LAS T Search TGR21 G- protein coupled receptor (GPCR) and e-value power 5 X 10 one 176,
  • HMMP FAM protein characteristic search by HMMP FAM, which is similar to 442 amino acid residues at 73%, shows a characteristic of G protein-coupled receptor (base sequence entered as “7tm 1” in P f am) ) Is found.
  • the protein encoded by the nucleotide sequence of SEQ ID NO: 6 has G protein-coupled receptor activity.
  • the amino acid sequence encoded by the nucleotide sequence described in SEQ ID NO: ⁇ is 6% with G protein-coupled receptor (GPR103): 0.0, 432. With homology.
  • GPR103 G protein-coupled receptor
  • HMMP FAM a sequence (a base sequence that is entered as “7tm-1” in P f am) is found that shows the characteristics of a G protein-coupled receptor.
  • the protein encoded by the nucleotide sequence shown in SEQ ID NO: 7 has G protein-coupled receptor activity.
  • proteins with high homology have high homology between the neuropeptide FF2, neuropeptide Y2, and galanin GalRl receptor and the B transmembrane region. .
  • the protein encoded by the nucleotide sequence shown in SEQ ID NO: 8 is a G protein-coupled receptor. Highly homologous proteins have high homology between the neuropeptide FF2, neuropeptide Y2, and the galanin GalRl receptor and the transmembrane domain.
  • Amino acid sequences are G protein-coupled receptor (GPR103) and e- value force 5X 10 one 160 according to the B LAS T search to nucleotide sequence encoding a SEQ ID NO: 9,
  • the protein encoded by the nucleotide sequence shown in SEQ ID NO: 9 is a G protein-coupled receptor. Highly homologous proteins have high homology between the neuropeptide FF2, the neuropeptide Y2, and the galanin GalRl receptor and the transmembrane domain.
  • the protein encoded by the nucleotide sequence shown in SEQ ID NO: 10 is a G protein-coupled receptor. Highly homologous proteins have high homology between the neuropeptide FF2, neuropeptide Y2, and the galanin GalRl receptor and the transmembrane domain.
  • amino acid sequence encoded by the nucleotide sequence set forth in SEQ ID NO: 1 1 to BLAST search a novel human g protein coupled receptor and e- va 1 ue: 5 X 1 0 179, 348 87% with an amino acid residue Shows homology. Also shown in SEQ ID NO: 11 When the amino acid sequence encoded by the nucleotide sequence was searched for protein characteristics using HMMPFAM, a sequence (a nucleotide sequence entered as “7tm-1” in P f am) showing characteristics of a G protein-coupled receptor was found.
  • the protein encoded by the nucleotide sequence shown in SEQ ID NO: 11 is a G protein-coupled receptor.
  • the amino acid sequence encoded by the nucleotide sequence of SEQ ID NO: 12 is gpcrxll as a functional receptor validated by angiopeptin and useful for screening of agonists and antagonists and e-value: 0.0, 507 amino acids remaining
  • Characteristic search by HMMPFAM with a homology of 72% in the amino acid group revealed a sequence (base sequence that is entered as “7tm-1J” in P f am) indicating the characteristics of a G protein-coupled receptor .
  • the protein encoded by the nucleotide sequence shown in SEQ ID NO: 12 is a G protein-coupled receptor.
  • highly homologous proteins are activated by the somatostatin analog angiopeptin, which suppresses atherosclerosis.
  • the amino acid sequence encoded by the nucleotide sequence of SEQ ID NO: 13 and G protein-coupled receptor C5L2 GPR77 are e-value: 5 X 10 1 114 , 61% homology at 331 amino acid residues, 61% homology In addition, it shows 44% homology with Oryctolagus cuniculus anaphylatoxin C5a receptor gene with e-value: 5 X 10 _60 and 329 amino acid residues.
  • HMMPFAM was used to perform a protein feature search on the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 13, and the sequence showing the characteristics of the G protein-coupled receptor (“7tm-1” in P f am) Base sequence to be entered).
  • the protein encoded by the nucleotide sequence shown in SEQ ID NO: 13 is a G protein-coupled receptor. Highly homologous proteins are involved in C5L2 expressed in granulocytes and undifferentiated dendritic cells and chemotaxis.
  • the nucleotide sequence of SEQ ID NO: 14 is Bos taurus latrophilin 3 splice variant abbf and e- value: 3 X 10 shows one 51, 302 ⁇ amino acids 37% homology at residues indicates the character of the G protein-coupled receptor proteins, wherein at the retrieval was performed by HMMPFAM The sequence (base sequence entered as “7tm 2” in P f am) is found.
  • the protein encoded by the nucleotide sequence shown in SEQ ID NO: 14 is a G protein-coupled receptor.
  • a highly homologous protein is brain-specific Latrophilin, a splicing variant of the exogenous toxin alpha-latrotoxin receptor Latrophilin.
  • SEQ ID NO: 1 5 nucleotide sequence according to the serpentine receptor (Cyt28) according to BLAST search e- value: 5 X 10 one 56, 271 38% ho th same or live was not in Amino acid residue, also , G protein-coupled receptor 56 and e_v alue: 8 X 10- 51, 281 36% of homology with Amino acid residue, further G-protein - is coupled receptor (GPR56), e- va 1 ue: 1 X At 10-50, it shows 36% homology over 281 amino acid residues.
  • G- protein coupled receptor GPR34 G- protein coupled receptor GPR34
  • Gpr34 G protein-coupled receptor Gpr34
  • a program tmHMM S. Moller, MDR that predicts a transmembrane helix for the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 16 Croning, R. Apweier ⁇ er.
  • the protein encoded by the nucleotide sequence shown in SEQ ID NO: 16 is a G protein-coupled receptor.
  • the gene encoding a similar protein is on the X chromosome, is abundantly expressed in various organs, and has weak homology to the PAF receptor.
  • the DNA of the present invention may be obtained in a state in which a base sequence has been deleted or inserted in the translated sequence.
  • the DNA If a deletion or insertion in the nucleotide sequence is suspected, use a method generally used by those skilled in the art, such as library screening, PCR cloning, etc., to delete the full length without deletion or insertion of bases.
  • cDNA can be obtained.
  • the protein of the present invention is expressed using the full-length cDNA thus obtained, and can be used for functional analysis.
  • the DNA of the present invention thus obtained, whose nucleotide sequence is determined, and whose function is presumed, has the nucleotide sequence shown in SEQ ID NOS: 1 to 16 or the nucleotide sequence shown above as its translation region.
  • these base sequences one or several (the number is not particularly limited, but is, for example, 60 or less, preferably 30 or less, more preferably 20 or less, and still more preferably Means 10 or less, particularly preferably 5 or less).
  • one or several amino acid sequences in the amino acid sequences described in SEQ ID NOs: 17 to 32 are deleted, substituted and / or substituted. Includes those encoding a protein consisting of an added amino acid sequence and having G protein-coupled receptor activity.
  • DNA that hybridizes under stringent conditions refers to a base sequence represented by SEQ ID NOS: 1 to 16 and a BLAST analysis of 80% or more, preferably 90% or more, and more preferably 95% or more. DNAs containing a base sequence having homology are exemplified.
  • Hybridization under stringent conditions refers to a reaction in a normal hybridization buffer at a temperature of 40 to 70 ° C, preferably 60 to 65 ° C, and the like. Washing can be performed according to a method of washing in a washing solution having a salt concentration of 15 mM to 30 O mM, preferably 15 mM to 6 O mM.
  • DNA of the present invention may be obtained by the above-described method or may be synthesized.
  • the DNA base sequence can be easily replaced with a sales kit such as a site-directed mutagenesis kit (Takara Shuzo) or a quick change site-directed mutagenesis kit (Stratagene). it can.
  • the nucleotide sequence of SEQ ID NOs: 1 to 16 is derived from a mouse.1S
  • a human cDNA library was prepared according to the above-described method for preparing a cDNA library.
  • a human homolog protein of the protein encoded by the nucleotide sequence of SEQ ID NOs: 1 to 16 (Hereinafter, this may be referred to as "human homolog DNA”).
  • human homolog DNA Such a DNA homologous to DNA that hybridizes under stringent conditions to DNA having the nucleotide sequence of SEQ ID NOS: 1 to 16 or a sequence complementary thereto according to the present invention is also included.
  • homologues of the target protein using informatics include, for example, (i) a cDNA database of a human or the like (a cDNA database predicted by informatics) using a query as a base sequence of a target cDNA; And (ii) homology search using BLAST etc. against EST databases such as humans using the base sequence of the target cDNA as a query. Then, a method is used in which the sequence of the hit EST is linked with reference to the base sequence of the target cDNA, and (iii) the base sequence of the target cDNA is used as a query against the genome database of humans or the like.
  • a homology search is performed using LAST, etc., to identify the location on the genome where the cDNA gene of interest is located, and Genscan (http: // genes, mit.edu/ GENSC A. html), Sim4 (Genome Res., 8: 976-74 (1998)), and the like, and a method of predicting the nucleotide sequence of the gene portion in the genome.
  • any of the above methods can be used, but any of the cDNAs having the nucleotide sequences of SEQ ID NOs: 1 to 16 of the present invention is novel. Since it is considered that the method (i) cannot obtain the nucleotide sequence of the human homologous DNA, the method described in (ii) or (iii) is preferably used.
  • a DNA encoding the human homolog protein of the protein encoded by the nucleotide sequence of SEQ ID NOS: 1 to 16 was obtained from the above human cDNA library. You can also.
  • the above-mentioned human cDNA library is prepared by using a primer having a nucleotide sequence complementary to the nucleotide sequence at the 5, 5, and 3 'end of the predicted human homolog DNA. Examples include a method of performing PCR as a template, and a method of performing hybridization on the human cDNA library using a partial sequence of the predicted human homolog DNA as a probe.
  • a similar gene having a nucleotide sequence having a high homology to the nucleotide sequence of the target gene is called a “homolog”, and the above-mentioned method also requires the acquisition of a human homolog. Although it is intended, it is important in gene function analysis not only to confirm that the nucleotide sequences are similar, but also to confirm that the gene obtained as a homolog is a family member of the target gene. Genes acquired as “homologs” between two species are identical genes that evolved from a common ancestral gene
  • the function of the protein encoded by the human-derived DNA must be In order to estimate and verify the function of the protein of the present invention in mice, it is preferable to confirm that the human homolog is an ortholog of a closely related species of the mouse gene of the present invention.
  • the following method is used as a method for confirming the ortholog.
  • (2) homology is analyzed for the nucleotide sequence of the obtained human homolog DNA and the corresponding nucleotide sequence of the cDNA of the present invention.
  • homology search was performed on the international nucleotide sequence database such as DDB J, EMBL, GenBank, and the mouse nucleotide sequence contained in the patent database. Confirm that the degree of matching between the cDNA of the invention and the base sequence of the query is higher than the degree of matching between the base sequence obtained from the database and the base sequence of the query.
  • the obtained human homolog can be identified as a human ortholog corresponding to the cDNA of the present invention.
  • the homology analysis described in (1) and (2) above is based on the amino acid sequence ratio. Comparison may be used, and a molecular evolutionary phylogenetic tree may be drawn and examined. In addition, it is preferable that the degree of matching by the homology analysis described in (1) and (2) above be analyzed as the degree of matching over the entire length of the query.
  • the DNA that hybridizes under stringent conditions with the DNA having the nucleotide sequence of SEQ ID NOS: 1 to 16 or the complementary sequence thereof includes DNA encoding such a human homolog or ortholog protein. It is.
  • the translation region of the protein encoded by the DNA of the present invention is, for example, the nucleotide sequence of the DNA, which is converted into amino acids by three reading frames, and the range encoding the longest polypeptide is translated by the present invention.
  • the amino acid sequence of the region can be determined. Such amino acid sequences include, for example, those described in SEQ ID NOs: 17 to 32.
  • the protein of the present invention is not limited to the above-described amino acid sequence, but comprises an amino acid sequence in which one or several amino acids have been substituted, deleted, and / or added in the amino acid sequence. And those having G protein-coupled receptor activity.
  • the method of transcription / translation of the DNA of the present invention described in the above (1) by an appropriate method is preferably used.
  • a recombinant vector inserted into a suitable expression vector or a suitable vector together with a suitable promoter is prepared, and this recombinant vector is used to transform a suitable host microorganism, or to a suitable cultured cell.
  • the expression can be obtained by introducing the DNA into a plasmid, and it can be obtained by purifying the protein.
  • Such salts of the protein of the present invention are also included in the protein of the present invention.
  • the protein produced by the above transformant may be modified before or after purification with an appropriate protein modifying enzyme to optionally modify the protein or partially remove the polypeptide to modify the protein. Quality.
  • modified proteins are also included in the scope of the present invention as long as they have G protein-coupled receptor activity.
  • the vector used for the production of the recombinant vector containing the DNA of the present invention is not particularly limited as long as the DNA is expressed in the transformant.
  • phage vectors usually, a commercially available protein expression vector into which an expression control region DNA such as a promoter suitable for a host into which the DNA is introduced has already been inserted is used.
  • Specific examples of such a protein expression vector include pET3 and pET11 (manufactured by Stratagene) and GEX (manufactured by Amersham Armacia Biotech) when the host is Escherichia coli.
  • p ESP-I expression vector manufactured by Stratagene
  • Bac PAK6 manufactured by Clontech
  • examples include ZAP Express (Stratagene), SVK3 (Amersham Pharmacia Biotech) and the like.
  • the promoter used herein is not limited to a promoter that can be used by a host microorganism or a promoter possessed by a cultured cell. Specifically, for example, when the host is Escherichia coli, T3, T7, tac , 1 ac promoter and the like, and in the case of yeast, nmt1 promoter, Ga11 promoter and the like can be used. When the host is an animal cell, SV40 promoter, CMV promoter and the like are preferably used.
  • a promoter specific to the gene of the present invention can also be used. Insertion of the DNA of the present invention into these vectors may be performed by linking the DNA or a DNA fragment containing the DNA to the amino acid sequence of the protein encoded by the gene DNA downstream of the open motor in the vector.
  • the recombinant vector thus prepared can be transformed into a host described below by a method known per se to prepare a DNA-introduced body.
  • a method for introducing the vector into a host specifically, a heat shock method (J. Mol. Biol., 53, 154, (1970)) and a calcium phosphate method (Science, 221, 551, (1983)) ), DEAE dextran method (Science, 215, 166, (1982)), in vitro packaging method (Proc. Natl. Acad. Sci. USA, 72, 581, (1975)), virus vector method (Cell, 37, 1053, (1984)), and the electric pulse method (Chu. Et al., Nuc. Acids Res., 15, 1331 (1987)).
  • the host for preparing the DNA transfectant is not particularly limited as long as the DNA of the present invention is expressed in the body.
  • the host include Escherichia coli, yeast, baculovirus (arthropod polyhedrovirus), insect cells, Alternatively, animal cells and the like can be mentioned.
  • BL21 and XL-2B1ue manufactured by Stratagene
  • SP—Q01 manufactured by Stratagene
  • AcNPV AcNPV
  • mice fibroblast C 127 J. Viol., 26, 291, (1978)
  • expression level ⁇ ⁇ cos-7 derived from African green monkey kidney is preferred because of simple screening.
  • a homologous recombination technique for directly inserting a promoter-ligated DNA fragment of the present invention into the chromosome of a host microorganism (AA Vertes et al., Biosci. Biotechnol. Biochem., 57, 2036, (1993))
  • a DNA transfectant can be prepared using a transposon, an insertion sequence (AA Vertes et al., Molecular Microbiol., 11, 739, (1994)) and the like.
  • cells or cells are collected by a method such as centrifugation, suspended in a suitable buffer, and then collected by a suitable method known per se such as ultrasonication, lysozyme, and / or freeze-thawing.
  • a suitable method known per se such as ultrasonication, lysozyme, and / or freeze-thawing.
  • a crude protein solution is obtained by centrifugation, filtration, or the like, and further purified by a combination of appropriate purification methods.
  • protein of the present invention is obtained.
  • protein expression is induced by subjecting the DNA of the present invention obtained in the above (1) to a cell-free transcription / translation system, and the protein of the present invention Can be obtained.
  • the cell-free transcription / translation system used in the present invention is a system containing all elements necessary for transcription from DNA to mRNA and translation of mRNA to protein, and by adding DNA thereto. It refers to any system by which the protein encoded by the DNA is synthesized.
  • Specific examples of the cell-free transcription / translation system include a transcription / translation system prepared based on an eukaryotic cell, a bacterial cell, or an extract from a part thereof, and a particularly preferred example is Egret A transcription / translation system prepared based on extracts from reticulocytes, wheat germ, and Escherichia coli (Escherichia coli S30 extract) may be mentioned.
  • Separation and purification of the protein of the present invention from the obtained transcription-translation product of the cell-free transcription / translation system can be carried out by a commonly used method known per se. Specifically, for example, a DNA region encoding an epitope peptide, a polyhistidine peptide, a glutathione-S-transferase (GST), a maltose binding protein, and the like is introduced into the DNA to be transcribed and translated, and And purified using the affinity of the protein with a substance having affinity.
  • GST glutathione-S-transferase
  • the expression of the target protein is separated by SDS-polyacrylamide gel electrophoresis and stained with Coomassie brilliant blue (manufactured by Sigma), or an antibody that specifically binds to the protein of the present invention described below. Detection method, etc. Can be confirmed. It is generally known that the expressed protein is cleaved (processed) by a proteolytic enzyme present in the living body.
  • the protein of the present invention is naturally included in the protein of the present invention, even if it is a fragment of the amino acid sequence that has been cleaved, as long as it has G protein-coupled receptor activity.
  • the proteins obtained by the effort can be used to find out the multifaceted functions in vivo.
  • a conventional method known per se can be used. Specifically, for example, yeast two-hybrid method, fluorescence depolarization method, surface plasmon method, phage display method, liposome method
  • yeast two-hybrid method fluorescence depolarization method
  • surface plasmon method phage display method
  • liposome method a conventional method known per se.
  • yeast two-hybrid method fluorescence depolarization method
  • surface plasmon method surface plasmon method
  • phage display method phage display method
  • liposome method a conventional method known per se.
  • the multiple display method is the multiple display method.
  • Oligonucleotides such as oligonucleotides can be prepared.
  • the oligonucleotide examples include a DNA having the same sequence as the continuous 5 to 100 bases in the base sequence of the DNA or a DNA having a sequence complementary to the DNA.
  • DNA having the same sequence as 5 to 100 consecutive nucleotides in the base sequence represented by any of SEQ ID NOS: 1 to 16 or DNA having a sequence complementary to the DNA Can be mentioned.
  • the above-mentioned oligonucleotides whose melting temperature (Tm) and number of bases do not extremely change are preferable.
  • the length of the sequence is generally 5 to: L00 bases, preferably 10 to 60 bases, and more preferably 15 to 50 bases.
  • oligonucleotide derivatives of these oligonucleotides can also be used as the oligonucleotide of the present invention.
  • examples of the oligonucleotide derivative include an oligonucleotide in which a phosphodiester bond in an oligonucleotide is converted to a phosphorothioate bond.
  • Nucleotide derivative an oligonucleotide derivative in which the phosphodiester bond in the oligonucleotide is converted to an N3,1 P5'phosphoamidate bond, and a report and a phosphodiester bond in the oligonucleotide that are converted to a peptide nucleic acid bond
  • Oligonucleotide derivatives Oligonucleotide derivatives in which peracyl in oligonucleotides are substituted with C-5 propynylperacyl, Oligonucleotide derivatives in which peracyl in oligonucleotides are substituted with C-15 thiazole peracyl, Oligonucleotides Oligonucleotide derivatives in which cytosine in nucleotides has been substituted with C-15-propynylcytosine; cytosine in oligonucleotides has been substituted with phenoxazine-modified cytosine; oligonucle
  • RNAi method RNA interference method
  • RNAi method RNA interference method
  • the method described in (Elbashir, S., et al., Nature, 411, 494-498 (2001)) can be used.
  • not all of the above-mentioned double-stranded RNAs need to be RNAs.
  • those described in WO 02/13734 can be used.
  • the target gene may be any as long as it is the DNA of the present invention.
  • a double-stranded RNA consisting of a sequence substantially identical to at least a part of the base sequence of these DNAs (hereinafter sometimes referred to as “double-stranded polynucleotide”) is referred to as a base sequence of a target gene. It is composed of a sequence substantially identical to a sequence of 15 bp or more, which may be any part of the sequence.
  • substantially identical means that the sequence has 80% or more homology with the sequence of the target gene.
  • the length of nucleotides ranges from 15 bp to the full length of the open reading frame (ORF) of the target gene.
  • RNA any length may be used, but a length of about 15 to 50 Obp is preferably used.
  • mammalian-derived cells have a signal transduction system that activates in response to long double-stranded RNA of 3 O bp or more. This is called an interfering reaction (Mareus, PI, et al., Interferon, 5, 115-180 (1983)), and when the double-stranded RNA enters the cell, PKR (dsRNA-responsive) protein kinase: The translation initiation of many genes is non-specifically inhibited via Bass, BL, Nature, 411, 428-429 (2001)), and at the same time, 2, 5, oligoadenylate synthetase
  • RNA is caused.
  • a double-stranded polynucleotide of 15 to 30 bp, preferably 19 to 22 bp, more preferably 21 bp It is preferable to use
  • the double-stranded polynucleotide does not need to be entirely double-stranded, and includes those with a partially protruding 5 'or 3' end, but preferably those having two or three bases protruding.
  • the double-stranded polynucleotide means a double-stranded polynucleotide having complementarity, but may be a self-annealed single-stranded polynucleotide having self-complementarity.
  • Examples of the single-stranded polynucleotide having self-complementarity include those having an inverted repeat sequence.
  • the method for preparing the double-stranded polynucleotide is not particularly limited, but it is preferable to use a chemical synthesis method known per se.
  • chemical synthesis a single-stranded polynucleotide having complementarity can be separately synthesized, and can be converted into a double-stranded strand by associating them by an appropriate method.
  • Specific examples of the method of association include a method in which the synthesized single-stranded polynucleotide is mixed, heated to a temperature at which the double-strand is dissociated, and then gradually cooled.
  • the associated double-stranded polynucleotide is confirmed using an agarose gel or the like, and the remaining single-stranded polynucleotide is removed by, for example, decomposing with a suitable enzyme.
  • the transfectant into which the double-stranded polynucleotide prepared in this way is introduced may be any as long as the target gene can be transcribed into RA or translated into protein in the cell. Specific examples include those belonging to plant, animal, protozoan, virus, bacterial, or fungal species.
  • the plant can be a monocotyledonous, dicotyledonous or gymnosperm, and the animal can be a vertebrate or invertebrate.
  • Preferred microorganisms are those used in agriculture or industry, and are pathogenic to plants or animals.
  • Fungi include organisms in both power and yeast forms.
  • vertebrates include mammals, including fish, sea lions, goats, pigs, sheep, hamsters, mice, rats, and humans, and invertebrates include nematodes and other insects. Includes creatures, Drosophila, and other insects.
  • the cells are vertebrate cells.
  • the transductant means a cell, tissue, or individual.
  • the cells may be from germ line or somatic, totipotent or pluripotent, divided or undivided, parenchymal or epithelial, immortalized or transformed, and the like.
  • the cells may be gametes or embryos, in the case of embryos, single cell embryos or constitutive cells, or cells from multicellular embryos, including fetal tissue. Further, it may be an undifferentiated cell such as a stem cell, or a differentiated cell such as a cell of an organ or tissue including fetal tissue, or any other cell present in an organism.
  • Differentiating cell types include fat cells, fibroblasts, muscle cells, cardiomyocytes, endothelial cells, neurons, glial, blood cells, megakaryocytes, lymphocytes, macrophages, neutrophils, and eosinophils Basophils, mast cells, leukocytes, granulocytes, keratinocytes, osteoblasts, osteoclasts, liver cells and cells of the endocrine or exocrine glands.
  • a method for introducing the double-stranded polynucleotide into the recipient when the recipient is a cell or tissue, calcium phosphate method, electroporation method, lipofection method, virus infection, two For example, immersion in a strand polynucleotide solution or a transformation method is used. Examples of the method for introduction into an embryo include microinjection, electoral poration, and virus infection. Can be When the recipient is a plant, a method of injecting or perfusing the plant into the body cavity or stromal cells, or spraying is used.
  • the double-stranded polynucleotide can be mixed directly with the food of the organism.
  • it can be administered, for example, by administration as an implanted long-term release preparation or the like, or by ingesting an introduced body into which a double-stranded polynucleotide has been introduced.
  • the amount of the double-stranded polynucleotide to be introduced can be appropriately selected depending on the introduced substance and the target gene, but it is preferable to introduce an amount sufficient to introduce at least one copy per cell. Specifically, for example, when the transfectant is a cultured human cell and the double-stranded polynucleotide is introduced by the calcium phosphate method, 0.1 to 100 OnM is preferred.
  • the function of the protein encoded by the gene of the present invention can be confirmed, or a new function can be analyzed.
  • an antibody that specifically binds to the protein of the present invention As a method for preparing an antibody that specifically binds to the protein of the present invention, a commonly used known method can be used. Even polypeptides used as antigens have high antigenicity according to known methods, A sequence suitable as (antigenic determinant) can be selected and used. As a method of selecting the epitope, for example, commercially available software such as Epitope Adviser (manufactured by Fujitsu Kyushu System Engineering Co., Ltd.) can be used.
  • polypeptide used as the above antigen a synthetic peptide synthesized according to a known method, or the protein itself of the present invention can be used.
  • a polypeptide serving as an antigen may be prepared in an appropriate solution or the like according to a known method, and immunization may be performed on a mammal, for example, a rabbit, a mouse, a rat, or the like.
  • immunization may be performed on a mammal, for example, a rabbit, a mouse, a rat, or the like.
  • the route of administration of the antigen upon immunization is not particularly limited, and any route such as subcutaneous, intraperitoneal, intravenous, or intramuscular route may be used. Specifically, for example, a method of inoculating a BALB / c mouse several times every several days to several weeks with an antigen polypeptide is used.
  • the antigen intake is preferably about 0.3 to 0.5 mg / time when the antigen is a polypeptide, but is appropriately adjusted depending on the type of the polypeptide and the animal species to be immunized.
  • test blood is collected as appropriate, and an increase in antibody titer is confirmed by enzyme-linked immunosorbent assay (hereinafter sometimes referred to as “ELISA”) or Western blotting.
  • ELISA enzyme-linked immunosorbent assay
  • Blood is collected from animals with elevated antibody titers.
  • a polyclonal antibody can be obtained by subjecting this to an appropriate treatment used for antibody preparation. Specifically, for example, a method of obtaining a purified antibody obtained by purifying an antibody component from serum according to a known method, and the like can be mentioned.
  • the antibody component can be purified by a method such as iontophoresis, ion exchange chromatography, and affinity chromatography.
  • a monoclonal antibody can be produced by using a hybridoma fused with spleen cells and myeloma cells of the animal according to a known method (Milstein, et al., Nature, 256, 495 (1975)). it can.
  • a monoclonal antibody can be obtained, for example, by the following method.
  • antibody-producing cells are obtained from an animal whose antibody titer has been raised by immunization with the above-mentioned antigen.
  • the antibody-producing cells are plasma cells and lymphocytes which are precursor cells thereof, which may be obtained from any of the individuals, but is preferably obtained from spleen, lymph nodes, peripheral blood and the like.
  • the myeloma to be fused with these cells is generally a cell line obtained from a mouse, for example, P3X63-Ag8.6653, an 8-azaguanine-resistant mouse (derived from BALB, etc.) myeloma cell line (ATCC: CRL -1580), P3-NSl / lAg4.1 (RIKEN cell bank: RCB0095) and the like are preferably used.
  • P3X63-Ag8.6653 an 8-azaguanine-resistant mouse (derived from BALB, etc.) myeloma cell line (ATCC: CRL -1580), P3-NSl / lAg4.1 (RIKEN cell bank: RCB0095) and the like are preferably used.
  • P3-NSl / lAg4.1 RIKEN cell bank: RCB0095
  • Iscov's Modified Dulbecco's Medium Certain, can be performed by using 50% polyethylene dalicol (PEG) dissolved in Dulbecco's Modified Eagle's Medium (DMEM) or the like. It can also be performed by the electrofusion method (U. Zimmermann. Et al., Naturwissenschaften, 68, 577 (1981)).
  • PEG polyethylene dalicol
  • DMEM Dulbecco's Modified Eagle's Medium
  • Hypridoma was prepared using 5% CO 2 in a normal medium (HAT medium) containing an appropriate amount of hypoxanthine 'aminopterin' thymidine (HAT) solution, utilizing the fact that the myeloma cell line used was an 8-azaguanine resistant strain. , At 37 ° C for an appropriate time.
  • This selection method can be appropriately selected and used depending on the myeoma cell line used.
  • the antibody titer of the antibody produced by the selected hybridoma is analyzed by the method described above, the hybridoma producing the antibody with a high antibody titer is separated by a limiting dilution method, etc., and the separated fused cells are cultured in an appropriate medium.
  • a monoclonal antibody can be obtained from the culture supernatant obtained by the purification by an appropriate method such as ammonium sulfate fractionation and affinity chromatography.
  • a commercially available monoclonal antibody purification kit can also be used for purification.
  • ascites containing a large amount of the monoclonal antibody of the present invention can also be obtained by growing the antibody-producing hybridoma obtained above in the abdominal cavity of an animal of the same strain as the immunized animal or a nude mouse. .
  • a human-derived protein When a human-derived protein is obtained as the protein of the present invention, the above-described polypeptide or a partial peptide thereof is used as an antigen in Severe combined immune deficiency (SCID) mice transplanted with human peripheral blood lymphocytes.
  • SCID Severe combined immune deficiency
  • Human antibodies can also be prepared by immunization in the same manner as described above and preparing a hybridoma between antibody-producing cells of the immunized animal and human myeloma cells (Mosier, DE, et al. Nature , 335, 256-259 (1988); Duchosal, MA, et al., Nature, 355, 258-262 (1992)).
  • RA is extracted from the obtained hybridoma producing the human antibody, the gene encoding the desired human antibody is cloned, this gene is inserted into an appropriate vector, and the gene is introduced into an appropriate host.
  • a humanized antibody can be prepared.
  • an antibody having a low binding property to an antigen can be obtained as an antibody having a higher binding property by using an evolutionary engineering technique known per se.
  • a partial fragment such as a monovalent antibody can be prepared by, for example, cleaving the Fab portion and the Fc portion using papain or the like, and collecting the Fab portion using an affinity column or the like.
  • the antibody that specifically binds to the protein of the present invention thus obtained can also be used as a neutralizing antibody that specifically binds to the protein of the present invention and thereby inhibits the activity of the protein.
  • the selection method is not particularly limited.
  • the function of the target protein in the introduced product is inhibited by bringing the antibody into contact with the DNA-introduced product prepared in (2) above. There is a method of analyzing whether or not this is the case.
  • Such a neutralizing antibody can be used alone for the clinical application, but can also be used as a pharmaceutical composition by mixing with a pharmaceutically acceptable carrier. At this time, the ratio of the active ingredient to the carrier can be varied between 1 and 90% by weight.
  • Such drugs can be administered in various forms, such as tablets, capsules, granules, powders, or syrups for oral administration, or injections, drops, ribosomes, Parenteral administration using suppositories and the like can be mentioned. The dose can be appropriately selected depending on the condition, age, body weight, and the like.
  • the protein of the present invention is prepared as a recombinant protein as described in the above (2), and the protein is analyzed by chromatography to confirm that it has the activity estimated in the above (1). . Furthermore, analysis can also be performed by combination with an antibody or the like prepared as described in (4) above.
  • Ligand sources for activity evaluation include transmitters, honolemon, cytokines, and G protein-coupled receptors. It contains routine agonists, biologically active peptides, cell culture supernatants, tissue extracts, and compounds not found in nature but that activate G protein-coupled receptors.
  • a functional assay or a binding test is performed using a ligand, a reporter gene, cAMP, a microphysiometer, or the like as described below, and a ligand for a G protein-coupled receptor is performed. Can be identified and the activity can be evaluated.
  • G protein-coupled receptors are linked to specific intracellular second messengers in response to G proteins coupled to them.
  • Gq-type G protein-coupled receptors are functionally linked to the increase in intracellular calcium concentration through activation of PLC, and Gs-type Gi-type G protein-coupled receptors are It has been shown to increase or decrease cAMP levels through activation or inhibition of rate cyclase.
  • HEK293 cells, CHO cells, cos cells, etc. expressing the recombinant protein of the present invention are loaded with calcium fluorescent dye fura2 or F1uo3, etc. at l-5 mM, and then the above-mentioned ligand source is added to the medium. After culturing, the transient increase in calcium concentration induced by agonists is evaluated using an intracellular calcium concentration measurement device (for example, CAF-100: Nihon Kohden or FD SS 6000: Hamamatsu Photonics, etc.). can do.
  • an intracellular calcium concentration measurement device for example, CAF-100: Nihon Kohden or FD SS 6000: Hamamatsu Photonics, etc.
  • cAMP assays include a reporter gene assay system using a cAMP response element (CRE) or a standard cAMP assay.
  • CRE cAMP response element
  • a standard cAMP assay for example, after the above-mentioned ligand source is added and cultured in a medium such as HEK293 cells, CHO cells, and cos cells expressing the recombinant protein of the present invention, stimulation or inhibition of cAMP production is performed. This can be done by evaluating
  • transient increase in calcium concentration or change in cAMP concentration Agonists that are active are tested in mottainfection cells in which only the vector has been introduced into similar cells to determine if the response is specific to the transformed cell expressing the receptor. Can be.
  • the activation of a wide range of second messenger systems secretes hydrogen ions out of cells.
  • This change in the extracellular hydrogen ion concentration can be detected by a site sensor, such as a microphysiometer (CYTO SENSOR micropy s i ome t er) (Molec cu l ar de vic e s L t d s, Men l o P ark, C soil). Therefore, the site sensor can detect receptor activation linked to an intracellular signaling pathway that uses energy, such as the G protein-coupled receptor of the present invention.
  • the binding of the above-mentioned ligand to the protein of the present invention can be examined by a normal binding experiment using a labeled ligand or a test using surface plasmon resonance.
  • the above functional assay system can also be used to evaluate agonists and antagonists of proteins having G protein-coupled receptor activity of the present invention. Note that confirmation of the activity of the protein of the present invention is not limited to these methods. Further, these functional assay systems can also be used for screening for a function activator or a function inhibitor of the protein of the present invention and a screening for a protein expression regulator of the present invention, which will be described later.
  • a method for analyzing the function of the protein of the present invention generally, for example, (i) a method for comparing and analyzing the expression state in each tissue, disease or developmental stage, and (ii) a method for analyzing the expression of other proteins and DNA (Iii) a method for introducing into appropriate cells or individuals and analyzing changes in phenotype; (iv) a method for inhibiting the expression of the protein in appropriate cells or individuals, and And a method of analyzing the change in Further, according to such a method, the activity specific to the target protein can be analyzed from many aspects.
  • the expression of the protein of the present invention is determined at the mRNA level or Can be analyzed at the protein level.
  • the expression level is analyzed at the mRNA level, for example, the in situ hybridization method (In situ hybridization: Application to Developmental Biology & Medicine., nd. by Harris, N. and Wilkinson, DG, Cambridge University) Press (1990)), a hybridization method using a DNA chip, a quantitative PCR method, and the like.
  • a tissue staining method using an antibody that specifically binds to the protein of the present invention described later, an ELISA, a Western plotting method, and the like can be mentioned.
  • a known variant exists in the protein to be analyzed, use a probe that exists only in the cDNA encoding the protein to be analyzed and does not hybridize with the cDNA that encodes the known variant. Is preferred.
  • the method is performed by selecting primers that can generate amplified fragments of different lengths between the target cDNA and the known variant (Wong, Y., Neuroscience Let., 320: 141-145 (2002)). And the like.
  • the function of the protein of the present invention can be analyzed by examining the presence or absence of interaction between the protein of the present invention and a known protein.
  • a method of analyzing interaction the ability to use a conventional method known per se, specifically, for example, yeast two-hybrid method, fluorescence depolarization method, surface plasmon method, phage display method, ribosome A multiple display method and the like can be mentioned.
  • the known variant if a known variant exists in the protein to be analyzed, the known variant also analyzes the interacting substance in the same manner and identifies a substance that interacts specifically with the target protein. Is preferred.
  • the cell into which the cDNA of the present invention is introduced is not particularly limited, but human cultured cells and the like are particularly preferably used. Methods for introducing DNA into cells include those described in (2) above.
  • the phenotype of the transfected cells includes cell viability, cell growth rate, cell differentiation, neurite outgrowth if the cells are neurons, This includes those that can be observed with a microscope, such as the localization and translocation of intracellular proteins, and those that can be analyzed by biochemical experiments, such as changes in the expression of specific proteins in cells.
  • these phenotypes can be similarly introduced into cells, and the phenotype associated with the target protein can be identified by comparative analysis. .
  • the protein of the present invention has a G protein-coupled receptor activity, it is also possible to analyze the protein by focusing on the phenotype and the like found in diseases associated with the G protein-coupled receptor. preferable.
  • the method can be efficiently performed by a method using an oligonucleotide described later or an RNA interference method.
  • a known variant is present in the target protein to be analyzed, the same analysis is performed for the known variant and other variants, and the target protein-specific protein is obtained by comparative analysis. Function can be identified.
  • This method of screening for a regulatory substance may be any method as long as it can obtain a substance that specifically binds to the protein of the present invention and has an activity of inhibiting, antagonizing, or enhancing the activity of the protein. .
  • a method is used in which a protein of the present invention is first brought into contact with a test substance, selection is performed using the binding property of the protein as an index, and then a change in the activity of the protein of the present invention is used as an index. be able to.
  • the test substance may be any substance as long as it interacts with the protein of the present invention and may affect the activity possessed by the protein. , Peptide, protein, non-peptide compound, small molecule compound Products, synthetic compounds, fermentation products, cell extracts, animal tissue extracts, and the like. These substances may be novel substances or known substances.
  • a method for analyzing the interaction between the test substance and the protein of the present invention a conventional method known per se can be used. Specifically, for example, the yeast two-hybrid method, the fluorescence depolarization method, the surface The plasmon method, the phage display method, the ribosomal display method, or the competition analysis method with the antibody described in the above (4) can be used.
  • the substance found to bind to the protein of the present invention is then analyzed by analyzing how the activity of the protein of the present invention is affected in the presence of the substance. Whether it is used as a modulator or not is identified.
  • the substance screened by the above method may be selected as a drug trap by screening in vivo.
  • a protein serving as a substrate is introduced into the DNA transfectant described in (2) by the same method. I do.
  • the dephosphorylation of the protein serving as a substrate in the presence / absence of the substance selected for the transductant is analyzed by a commonly used method known per se. Specifically, it can be performed using the method described in (5) above. If the calcium concentration, c AMP concentration, etc., are increased as compared to the absence of the substance, the substance may function as a G protein-coupled receptor activator, or may decrease, or When inhibited, the substance can be identified as possibly functioning as a G protein-coupled receptor inhibitor.
  • the above-mentioned human homologous protein or orthologous protein as the DNA or recombinant protein of the present invention to be used.
  • substances screened by the above method can be used as drug candidates by screening in vivo. You may make a selection.
  • the G protein-coupled receptor having the G protein-coupled receptor activity of the protein of the present invention is a superfamily of essential membrane proteins that transmit extracellular signals.
  • GPCRs include receptors for living amines such as dopamine, epinephrine, histamine, glutamic acid (a metabolic effect), acetylcholine (a musculinic effect), and serotonin, as well as prostaglandins and platelet activation.
  • receptors for sensory signaling media such as retinal photopigments and olfactory stimulants.
  • compounds that can be identified by this screening method include hypertension, angina, myocardial infarction, atherosclerosis, ulcer, asthma, allergy, psychosis, Alhaima's disease, chorea, schizophrenia, depression, and migraine , Ischemic brain disease, anxiety, vomiting, benign prostatic hypertrophy, Parkinson's disease, acute heart failure, diabetes, diabetic retinopathy, ophthalmic disease, endocrine disease, immune disease, inflammatory disease, respiratory disease, hypotension, urine It can be used as a remedy for closure, infertility, contraception, liver disease, and osteoporosis.
  • modulators can be used alone as the above active ingredients when applied to clinical applications, but can also be used as pharmaceutical compositions by blending with a pharmaceutically acceptable carrier.
  • the ratio of the active ingredient to the carrier can be varied between 1 and 90% by weight.
  • Such drugs can be administered in various forms, such as tablets, capsules, granules, powders, or syrups for oral administration, or injections, drops, ribosomes, Parenteral administration with suppositories and the like can be mentioned.
  • the dose can be appropriately selected depending on symptoms, age, weight, and the like.
  • Screening methods include the use of the protein of the present invention in the presence of a test substance. Or a method of analyzing the expression level of mRNA encoding the same. Specifically, for example, cells expressing the protein of the present invention described in (2) above are cultured in an appropriate medium containing a test substance, and the amount of the protein of the present invention expressed in the cells is determined by ELISA. Or by analyzing the amount of mRNA encoding the protein of the present invention in the cells by quantitative reverse transcription PCR, Northern blotting, or the like. Can be.
  • test substance those described in the above (6) can be used.
  • the substance promotes the expression of the DNA of the present invention.
  • the substance may function as a substance, and conversely decreases, it can be determined that the substance can be used as the DNA expression inhibitor of the present invention.
  • the above-mentioned active ingredient can be used alone for clinical application, but it can also be used as a pharmaceutical composition by blending it with a pharmaceutically acceptable carrier. At this time, the ratio of the active ingredient to the carrier can be varied between 1 and 90% by weight.
  • the drug can be administered in various forms. Examples of the dosage form include tablet, capsule, granule, powder, syrup, and the like, oral administration, injection, drip, ribosome And parenteral administration with suppositories and the like. In addition, the dose can be appropriately selected depending on symptoms, age, weight, and the like.
  • the transfected DNA containing the DNA of the present invention described in the above (1) is constructed, introduced into a fertilized egg of a mammal other than a human, and transplanted into a female individual uterus to generate the present DNA.
  • a non-human mammal into which the DNA of the present invention has been introduced can be produced. More specifically, for example, a female individual is superovulated by hormone administration, then mated with a male, a fertilized egg is excised from the oviduct on the first day after mating, and the DNA is introduced into the fertilized egg with a micro DNA. It is introduced by a method such as injection.
  • Non-human mammals include, for example, mice, rats, guinea pigs, hamsters, rabbits, goats, pigs, dogs, cats, and the like.
  • the DNA-introduced animal of the present invention thus obtained is bred with this individual, and its offspring are bred in a normal breeding environment while confirming that the introduced DNA is stably maintained. Obtainable. Also, by repeating in vitro fertilization, the offspring can be obtained and the strain can be maintained.
  • the non-human mammal into which the DNA of the present invention has been introduced can be used as an analysis of the function of the DNA of the present invention in a living body, or as a screening system for a substance regulating the function.
  • the protein of the present invention can be used as a carrier having it bound on a substrate.
  • a nucleotide sequence encoding the protein of the present invention for example, a DNA having the nucleotide sequence of any one of SEQ ID NOs: 1 to 16 and a partial fragment thereof can be used as a carrier obtained by binding them to a substrate.
  • these may be referred to as “protein chips”, “DNA chips” or “DNA arrays” (DNA microarrays and DNA macroarrays).
  • These protein chips or DNA chips or arrays may contain other proteins ⁇ DNA in addition to the protein and DNA of the present invention.
  • a resin substrate such as a nylon film or a polypropylene film, a nitrocellulose film, a glass plate, a silicon plate, or the like is used as a substrate for binding proteins and DNA, but the detection of hybridization is not performed by non-RI.
  • a glass plate or a silicon plate containing no fluorescent substance is preferably used.
  • the binding of the protein or DNA to the substrate can be easily carried out by a commonly used method known per se.
  • the amino acid sequence of the protein of the present invention and the nucleotide sequence of DNA can also be used as sequence information.
  • the nucleotide sequence of the DNA of the present invention includes the nucleotide sequence of the corresponding RNA. That is, a database of amino acid sequences and nucleotide sequences can be constructed by storing the obtained amino acid sequences and nucleotide sequences in an appropriate recording medium in a computer-readable predetermined format. This database may contain the nucleotide sequences of other types of proteins and the DNA encoding them. Further, in the present invention, the database also means a computer system that writes the above-mentioned sequence on an appropriate recording medium and performs a search according to a predetermined program.
  • Suitable recording media include, for example, magnetic media such as flexible disks, hard disks, and magnetic tapes; optical disks such as CD-ROM, MO, CD-R, CD-RW, DVD-R, and DVD-RW; and semiconductors. Examples include memory.
  • magnetic media such as flexible disks, hard disks, and magnetic tapes
  • optical disks such as CD-ROM, MO, CD-R, CD-RW, DVD-R, and DVD-RW
  • semiconductors such as CD-ROM, MO, CD-R, CD-RW, DVD-R, and DVD-RW
  • Examples include memory.
  • mRNA-prepared mouse (C57BL / 6) 0.5 to lg of each organ or tissue is homogenized with a 10 ml suspension, and the same amount of fuminol / clonal form as lm 1 of 2M sodium acetate at pH 4.0 (Volume ratio 5: 1) The mixture was added and extracted. When the same amount of isopropanol was added to the aqueous layer after the extraction, RNA separated and precipitated from the aqueous phase.
  • RNA was then centrifuged at room temperature at 4,000 rpm for 15 minutes to dissolve the RNA in 4 ml of 7 M guanidine-C1. After adding twice the volume of ethanol, the mixture was incubated on ice for 1 hour, centrifuged at 4,000 rpm for 15 minutes, and the resulting precipitate was washed with 70% ethanol to recover RNA. And the RNA purity was determined by reading the OD ratios 260Z280 (> 1.8) and 230Z260 «0.45).
  • Oligonucleotide containing recognition sequence of restriction enzyme Xho I (SEQ ID NO: 33) (wherein, V indicates A, G, or C, and N indicates A, G, C, or T) 12.6 ⁇ 1 was used as a primer.
  • RNAse-free water (hereinafter referred to as (Sometimes referred to as "RNase-free water").
  • Biotinylation of RN ⁇ diol Two steps to bind biotin to the diol site of RN ((present at both the 5 'end with the Cap structure and the 3' end report with the poly A chain) The reaction was performed. These are the oxidation of the diol group followed by the coupling reaction of biotin hydrazide with the oxidized RNA. First, 15 ⁇ g of the RNA-first strand cDNA complex obtained by the reverse transcription reaction was purified using 6.6 mM sodium acetate buffer (PH4.5) and sodium periodate as an oxidizing agent. ⁇ ⁇
  • reaction 1 This oxidation reaction was performed on ice for 45 minutes under light-shielded conditions. Then add 11 ⁇ l of 5 ⁇ sodium chloride, 0.5 ⁇ l of 10% SDS and 0.5 ⁇ l of the same amount of isopropanol, leave on ice for 60 minutes, and then at 4 ° C for 15 minutes.
  • the precipitate was obtained by centrifugation at 15,000 rpm. The precipitate was washed with 70% ethanol and redissolved in 50 ⁇ l of RNase-free water. To the sample, add 5 ⁇ l of sodium acetate ( ⁇ 6.1), 5 ⁇ l of 10% SDS, 10 ⁇ m of biotin hydrazide (Sigma) 150 ⁇ 1, and add it to room temperature (22 to 26 ° C ) Overnight. Finally, add 5 ⁇ l of 5 M NaCl, 1 M sodium acetate (pH 6.1), 751, and 2.5 volumes of ethanol, cool on ice for 1 hour, and centrifuge at 4 ° C for 15 minutes And biotinylated. After the reaction, the reaction solution was centrifuged for 15 minutes to precipitate the RNA-DNA complex again. The precipitate was washed once with 70% ethanol and once with 80% ethanol, and dissolved in RNase free water 701.
  • the beads were suspended in 500 ⁇ l of a solution of 50 mM EDTA and 2 M NaCl, and the RNase I-treated cDNA obtained in (4) was added. By stirring at room temperature for 30 minutes, the magnetic beads and the full-length cDNA were bound. The beads that captured the full-length cDNA were washed four times with a solution of 5 OmM EDTA and 2 M NaC1, 0.4% SDS, once with 50 ⁇ g ZiU1 yeast tRNA, and OmM NaC1, 0.
  • the single-stranded full-length cDNA recovered in this manner was extracted with phenol / chloroform, and the volume of the solution was reduced to 100 ⁇ l or less using a speed bag, and then subjected to G25ZGIOOS ephadex chromatography. Fractions having RI activity were collected in a silicon-treated microtube, 2 ⁇ g of dalycogen was added, and the precipitate obtained by ethanol precipitation was dissolved in 30 ⁇ l of ultrapure water. (6) Add oligo dG to single-stranded cDNA
  • the full-length double-stranded cDNA obtained by the above method was inserted into a ⁇ II vector and recovered as a library.
  • the ⁇ ZAP III vector is obtained by modifying SEQ ID NO: 35, which is a partial sequence of the multiple cloning site of ⁇ ZAP II (manufactured by STRATAGENE) vector, to SEQ ID NO: 36, and newly introducing two Sfi I sites. .
  • LPS (RIKEN) vector was prepared and cDNA was inserted.
  • RI KEN (named ⁇ -FLC-1 (FLC stands for FULL-LENGTH cDNA)) is a modified version of the LPS vector from Mo Bitec (Germany) for cDNA. is there. That is, BamHI and Sa1I, which are convenient for cDNA insertion, were introduced into the cloning sites on both sides of lO kbpstuffer, and cDNA from 0.5 kb to about 13 kb was cloned. A 6 kb DNA fragment inserted into the Xba I site
  • RNA driver The mRNA prepared in Example 1 (1)
  • RNA driver The RNA prepared by the in vitro transcription reaction
  • the latter RNA is further divided into two types (hereinafter referred to as “(b) RNA driver” and “(c) RNA driver”).
  • cDNA was recovered from RNA-cDNA that had been removed by normalization and cloned into a phage vector.
  • One starting material after infection with E. coli One thousand (2,000) plaques are mixed together to form one library (mini-library), which is converted to plasmid DNA by a conventional method.
  • mini-library One library (mini-library), which is converted to plasmid DNA by a conventional method.
  • the phage is again infected with Escherichia coli together with a helper phage, converted into a phagemid, and further infected again with a plasmid.) Get DNA).
  • RNA (b) RNA driver was obtained.
  • mini-libraries are created from nine types of tissues (pancreas, liver, lung, kidney, brain, spleen, testes, small intestine, stomach), and mixed with nine types of mini-libraries. To obtain RNA.
  • RNA is cultured from a library (about 20,000 clones) that is already stored as a non-overlapping clone, and the resulting DNA is subjected to in vitro transcription reaction in the same manner as the RNA driver (c). RN A driver.
  • RNA was labeled with biotin using the Label_ITB iotin labeling kit (manufactured by Mirus Corporation), and then added to tester cDNA at a ratio of 1: 1: 1. Reaction at t 10
  • One representative clone was selected from each cluster. Representative clones were selected using Q-bot (manufactured by G ENET IXLI MITED) and arrayed on a 384-well plate. At that time, E. coli was cultured in 50 ⁇ l of LB medium at 30 ° C for 18 to 24 hours. At this time, if the cDNA library has been introduced into the PS vector and Escherichia coli DH10B has been transformed, add 10 Omg / ml of ampicillin and 5 Omg / ml of kanamycin and introduce it into the Zap vector. Led to SOLR system If so, 10 Omg / ml of ampicillin and 25 m / ml of streptavidin were added.
  • Each clone cultured in (1) above is further cultured in 1.3 ml HT solution containing 10 OmgZm1 ampicillin, and after collecting cells by centrifugation, Q
  • Plasmid DNA was recovered and purified using I-Aperp 96 Turbo (manufactured by QIAGEN). In order to check the chain length of the cDNA inserted in the obtained plasmid, 1Z30 of the plasmid DNA obtained above was
  • Plasmids were divided into two categories: those with insert sequences shorter than 2.5 kb and those with longer insert sequences. Of these clones, the clone having an insertion sequence shorter than 2.5 kb was analyzed for the nucleotide sequence from both ends. At this time, the plasmid was prepared using the primers described in SEQ ID NO: 37 (sense strand) when the vector was PS, and the primers described in SEQ ID NO: 39 (sense strand) when the vector was Zap.
  • Gaps that could not be analyzed by the above nucleotide sequence analysis were determined by the primer walking method.
  • AB IPris m377 and Z or AB IPri sm3700 manufactured by Applied Biosystems Inc.
  • Big Dy ete rm inatorkit and Cyclic Sequencing FS ready Reacti on Kit (Ap plied B iosysterns Inc.).
  • sequencing of clones with inserted cDNAs longer than 2.5 kb was performed by the shotgun method.
  • Shimadzu RISA384 and DYEnam1cEtterminatorcyclesequencingkit manufactured by Amersham Pharma Pharma Pharmacia Biotech
  • To generate a shotgun library 48 DNA fragments grown in PCR from 48 independent representative clones were used. The ends of the amplified DNA fragments were blunt-ended with T4 DNA polymerase.
  • This DNA fragment was inserted into a pUC18 vector, and Escherichia coli DH10B was transformed with the recombinant vector.
  • a plasmid was prepared from this E. coli in the same manner as in (2) above.
  • nucleotide sequence was determined by nucleotide sequence analysis from both ends, and the nucleotide sequences were ligated on a computer, followed by Double Stroke S hearing Device (Fiore Inc. Was performed. Nucleotide sequencing by the shotgun method was performed with duplication of 12 to 15 clones. The gap whose sequence could not be determined by the nucleotide sequence determination was determined by primer walking in the same manner as described above.
  • dn a fo rm34147 is composed of 2386 bases, as shown in SEQ ID NO: 1, of which base numbers 107 to 1858 have an open reading frame.
  • the amino acid sequence predicted from the open reading frame consists of 583 amino acid residues (SEQ ID NO: 17).
  • SEQ ID NO: 17 The amino acid sequence predicted from the open reading frame consists of 583 amino acid residues.
  • the SPTR protein database SWI SS-PROT protein sequence database and the TrEMBL nucleic acid translation database were integrated.
  • I Database registration symbol Mus musc 1 us AF 346501, G roteinco up ledreceptoraffectingt esticula rdescent (Great) force e— value: 0.0 hit with 579 amino acid residue with 99% coincidence.
  • amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 1 was searched for protein characteristics using HMM PFAM.
  • amino acid sequence encoded by nucleotide numbers 182—301 in SEQ ID NO: 1 was encoded by amino acid sequence [J ow—density 1 ipoproteinreceptordoma inclass A (J (base sequence sequenced as “1 d1—recept—a” in P f am)) was identified.
  • J ow density 1 ipoproteinreceptordoma inclass A
  • base sequence entered as “LR R” in P fam was found.
  • dnaform37308 is composed of 2558 bases, of which base numbers 507 to 1874 are open reading frames.
  • the amino acid sequence predicted from the open reading frame consists of 455 amino acid residues (SEQ ID NO: 18).
  • SEQ ID NO: 18 the SPTR protein database (integrated SWI SS-PROT protein sequence database and TrEMBL nucleic acid translation database)
  • I Database registration code AK018327, proteins im ilarto PRO BABLE G PROTE IN—COUP LED RECEPTOR GPR 72 PRECURSOR (GLUCOCORT I CO I D- I NDUCED RECEPTOR) force e- va 1 ue: 5 in X 1 CT 136, with 100% degree of coincidence over the 240 amino acid residues, also have (ii) a database registration mark Q 9NYM4, orphan G- proteincouledreceptor R 72 , e- va 1 ue: 1 with X 1 CT 84, 45% one ⁇ over 301 amino acid residues, yet (iii) a database registration mark AY 02
  • amino acid sequence encoding the nucleotide sequence shown in SEQ ID NO: 2 was subjected to protein characteristic search using HMM PFAM. As a result, it was found that the amino acid sequence encoded by nucleotide numbers 840 to 1613 of SEQ ID NO: 2 had a G protein A sequence (a nucleotide sequence entered as “7 tm-1” in P f am) showing the characteristics of the conjugated receptor was found.
  • the above-mentioned GPR72 is found to be an orphan GPCR similar to the Y-receptor based on the literature information in the database (Biochim Biophys Acta 2000 Apr 25; 1491 (1-3): 369-75). 7
  • the glucocorticoid of intestinal ci ci-in du cedrecepto is based on the literature information in the database (J Neurosci 2001 Nov 15; 21 (22): 9027-35). Therefore, it is shown that the expression is enhanced upon withdrawal after chronic amphetamine administration.
  • the protein encoded by the nucleotide sequence shown in SEQ ID NO: 2 may be induced by darcocorticoid. It is a G protein-coupled receptor, and its ligand is peptide, and it was speculated that it is involved in neuroadaptation to stress and psychostimulants.
  • dnafo rm37953 was composed of 3015 bases, of which base numbers 9 to 1169 were open reading frames (including a stop codon).
  • the amino acid sequence predicted from the open reading frame consists of 386 amino acid residues (SEQ ID NO: 19).
  • the S PTR protein database (a combination of the SWI SS—P ROT protein sequence database and the Tr EMB L nucleic acid translation database) contained ( i) database registration symbol AK018327, PROBABLE G-pROTE I N -COUPL ED RECEPTOR GPR 72 PRECURSOR (GLUCOCOR TI CO I D- I NDUCED RECEPTOR is, e- value: at 5 X l 0-96, i 7 3 amino acid residues 10 0% degree of coincidence over the group, also (ii) data base over scan registration mark Q 9 NYM4, orhan G- proteincoupledrecepto r GPR 72 force e- value: with 2 X 10- 84, 30 1 amino acid residue in 45 ° / 0 degree of coincidence over, further (iii) a database registration mark AiT0290 ⁇ '1 attusnorveg ⁇ cusglucocor
  • a protein characteristic search using the HMM PFAM for the amino acid sequence encoding the nucleotide sequence shown in SEQ ID NO: 3 revealed that the G protein was conjugated to the amino acid sequence encoded by nucleotide numbers 135-908 of SEQ ID NO: 3.
  • a sequence (a nucleotide sequence entered as “7 tm-1” in P i am) showing the characteristics of the type receptor was found.
  • the above-mentioned GPR72 was found to be an orphan GPCR similar to the Y-receptor from literature information in the database (Biochim Biophys Acta 2000 Apr 25; 1491 (1-3): 369-75).
  • the glucocorticoid-induced receptor in ⁇ d above is expressed in the brain based on literature information in the database (J Neurosci 2001 Nov 15; 21 (22): 9027-35), and that the ligand is a peptide GPCR. However, it has been shown that the expression is enhanced at the time of drug withdrawal after chronic amphetamine administration. From these facts, the protein encoded by the nucleotide sequence shown in SEQ ID NO: 3 is an orphan G protein-coupled receptor that may be induced by glucocorticoid, the ligand is peptide, and the stress or psychostimulant It was speculated that it might be involved in the neural adaptation to the disease. (4) dn afo rm42851 (SEQ ID NOs: 4, 20)
  • dnafor m42851 consists of 3138 bases as shown in SEQ ID NO: 4, of which base numbers 474 to 1841 have an open reading frame.
  • the amino acid sequence predicted from the open reading frame consists of 455 amino acid residues (SEQ ID NO: 20).
  • SEQ ID NO: 20 The amino acid sequence predicted from the open reading frame consists of 455 amino acid residues (SEQ ID NO: 20).
  • the SPTR protein database integrated SWI SS-PROT protein sequence database and TrEMBL nucleic acid translation database
  • Database registration code AK018327 ptoteins im ilarto PRO BABLE G—PROTE IN—COUP LED RECEPTOR GPR 72 PRECURSOR (GLUCOCORT I CO I D- I N DUCED RECEPTOR) I e-va 1 ue: 5X in 10 one 136, also at 240 Amino acid residues 100% degree of coincidence over the group, also (ii) a database registration mark Q 9 NYM4, Homo sapiensorpnan Ur- proteincouledreceptor GPR 72 (GPR 72) power e- value: 2 X l 0 one 84, 45% degree of coincidence over
  • amino acid sequences encoded by the nucleotide sequence shown in 4 sequences showing characteristics of G protein-coupled receptor protein, characterized retrieval by HMM PF AM to the amino acid sequence nucleotide numbers 807- 1 580 of SEQ ID NO: 4 where having conducted encoded
  • the above-mentioned GPR72 is found to be an orphan GPCR similar to the Y-receptor based on the literature information in the database (Biochim Biophys Acta 2000 Apr 25; 1491 (1-3): 369-75).
  • the glucocorticoid-in du cedreceptor described above is based on literature information in the database (J Neurosci 2001 Nov 15; 21 (22): 9027-35), which indicates that it is expressed in the brain, that the ligand is a peptide GPCR, and that expression is enhanced upon discontinuation of the drug after chronic amphetamine administration.
  • the protein encoded by the nucleotide sequence shown in SEQ ID NO: 4 is an orphan G-protein-coupled receptor that may be induced by glucocorticoids, its ligand is peptide, and it has a neuronal response to stress and neurostimulants. It was presumed to be involved in adaptation.
  • dnafo rm27636 was composed of 2499 bases, of which nucleotides 281 to 1267 were an open reading frame (including a stop codon).
  • the amino acid sequence predicted from the open reading frame consists of 328 amino acid residues (SEQ ID NO: 21).
  • a homology search was performed on the amino acid sequence encoded by SEQ ID NO: 5 using BLAST, and a SPTR protein database (integrated SWI SS-PROT protein sequence database and TrEML nucleic acid translation database) Among them, (i) database registration code A F41 11 11 1, Homo saiens G protein-couledreceptor (GPR82) genej, e—va 1 ue: 5 X 10 " 159 , and 82% over 328 amino acid residues The amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 5 was subjected to a protein feature search using HMM PFAM, and the amino acid sequence encoded by nucleotides 374-1210 in SEQ ID NO: 5 was determined.
  • a sequence exhibiting characteristics of a G protein-coupled receptor (a nucleotide sequence entered as “7 tm-1” at P f am) was found. From the literature information (Gene2001 275 (1): 83-91) in the database for the human ortholog of GPR82, it is shown that ESTs are obtained from human colon tissue. From these facts, it was inferred that the protein encoded by the nucleotide sequence of SEQ ID NO: 5 is a novel mouse orphan G protein-coupled receptor and is expressed in the colon.
  • dnafo rm47734 (SEQ ID NOs: 6, 22) As shown in SEQ ID NO: 6, dnafo rm47734 is composed of 2146 bases, of which base numbers 173 to 1456 are open reading frames.
  • the amino acid sequence predicted from the open reading frame consists of 427 amino acid residues (SEQ ID NO: 22).
  • SEQ ID NO: 22 amino acid sequence encoded by SEQ ID NO: 6 using BLAST, the SPTR protein database (SWI SS—PROT protein sequence database integrated with the TrEMBL nucleic acid translation database) ), (I) Database registration code AX 37657 5, human TGR 21 G—protein coupled receptor (GPCR) force e—value: 5 ⁇ 10 1 176 , and 442 amino acid residues over 73 Hit with% match.
  • amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 6 was subjected to protein characteristic search using HMM PFAM, and the amino acid sequence encoded by nucleotide numbers 368-1 207 in SEQ ID NO: 6 was found to have a G protein-coupled receptor.
  • dnafo rm29894 was composed of 1575 bases, of which nucleotides 53 to 1354 had an open reading frame (including a stop codon).
  • the amino acid sequence predicted from the open reading frame consists of 433 amino acid residues (SEQ ID NO: 23).
  • a homology search was performed using BLAST on the amino acid sequence encoded by SEQ ID NO: 7, and it was found in the SPTR protein database (integrated SWI SS—PROT protein sequence database and TrEMBL nucleic acid translation database).
  • the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 7 was subjected to a protein feature search using HMM PFAM. As a result, it was found that the amino acid sequence encoded by nucleotide numbers 236-1048 of SEQ ID NO: 7 had a G protein-conjugated A sequence (a nucleotide sequence entered as “7 tm-1” in P f am) showing the characteristics of the receptor was found.
  • the above-mentioned human GPR103 is based on the literature information in the database (Gene 2001 Sep 5; 275 (1): 83-91) based on the information of neuropeptide FF2, neuropeptide Y2 and galanin GalRl receptor and transmembrane region.
  • dnafo rm was composed of 2864 bases, of which base numbers 64 to 1161 were open reading frames (including a stop codon).
  • the amino acid sequence predicted from the open reading frame consists of 365 amino acid residues (SEQ ID NO: 24).
  • a homology search was performed for the amino acid sequence encoded by SEQ ID NO: 8 using BLAST, and it was found in the SPTR protein database (integrated SWI SS-PROT protein sequence database and TrEMBL nucleic acid translation database). , (i) No.
  • amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 8 was searched for protein characteristics using HMM PFAM.
  • the above human GPR103 is based on the literature information in the database (Gene 2001 Sep 5; 275 (1): 83-91), and indicates the degree of coincidence between neuropeptide FF2, neuropeptide Y2 and galanin GalRl receptors and the transmembrane domain. Is 34-38%, and Northern Hidden Reduction experiments show that mRNA is expressed in the cerebral cortex, pituitary gland, thalamus, hypothalamus, basal forebrain, midbrain, and pons . From these facts, it was conjectured that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 8 was expressed in the brain, and that the ligand was a peptide G protein-coupled receptor and had GPCR activity. .
  • dnafo rm53728 was composed of 1642 bases, of which base numbers 88 to 1185 were an open reading frame (including a stop codon).
  • the amino acid sequence predicted from the open reading frame consists of 365 amino acid residues (SEQ ID NO: 25).
  • a homology search was performed using BLAST for the amino acid sequence encoded by SEQ ID NO: 9, and it was found in the SPTR protein database (integrated SWI S_PROT protein sequence database and TrEMBL nucleic acid translation database). to, (i) a database registration No.
  • amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 9 was searched for protein characteristics using HMMP FAM. As a result, it was found that the amino acid sequence encoded by SEQ ID NO: 9 nucleotide number 115-927 showed the characteristics of G protein-coupled receptor. The sequence shown (base sequence entered as “7 tm-1” in P f am) was found.
  • the above-mentioned human GPR103 is based on the literature information in the database (Gene 2001 Sep 5; 275 (1): 8391), such as force, et al., Neuropeptide FF2, neuropeptide Y2 and galanin GalRl receptor and 34-38% match, plus Northern Yong experiments show that niRNA is expressed in the cerebral cortex, pituitary gland, thalamus, hypothalamus, basal forebrain, midbrain, and pons. From these facts, it was speculated that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 9 was expressed in the brain, and that the ligand was a peptide-type G protein-coupled receptor and had GPCR activity. .
  • dnafo rm62766 was composed of 1698 bases, of which base numbers 160 to 1236 were open reading frames (including a stop codon).
  • the amino acid sequence predicted from the open reading frame consists of 358 amino acid residues (SEQ ID NO: 26).
  • a homology search was performed using BLAST for the amino acid sequence encoded by SEQ ID NO: 10, and it was found in the SPTR protein database (integrated SWI SS-PROT protein sequence database and TrEMBL nucleic acid translation database).
  • the above-mentioned human GPR103 is based on the literature information in the database (Gene 2001 Sep 5; 275 (1): 83-91), which indicates that neuropeptide FF2, neuropeptide Y2 and galanin GalRl receptor and transmembrane region The agreement is 34-38%, and the Northern Hidden Reduction experiments show that mRNA is expressed in the cerebral cortex, pituitary gland, thalamus, hypothalamus, basal forebrain, midbrain, and pons. ing. Therefore, the protein encoded by the nucleotide sequence of SEQ ID NO: 10 is expressed in the brain, and the ligand is a peptide G protein-coupled receptor and has GPCR activity. That was speculated.
  • dn afo rni325 & 7 consisted of 3792 bases, of which base numbers 92 to 1207 were open reading frames (including a stop codon).
  • the amino acid sequence predicted from the open reading frame consists of 371 amino acid residues (SEQ ID NO: 27).
  • a homology search was performed using BLAST on the amino acid sequence encoded by SEQ ID NO: 11, and it was found in the SPTR protein database (integrated SWI SS-PROT protein sequence database and TrEMBL nucleic acid translation database).
  • SPTR protein database integrated SWI SS-PROT protein sequence database and TrEMBL nucleic acid translation database.
  • the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 11 was subjected to a protein feature search using HMMP FAM. As a result, it was found that the amino acid sequence encoded by nucleotide numbers 287 to 1081 in SEQ ID NO: 11 had G protein-coupled A sequence (base sequence entered as “7 tm-1” in P f am) showing body characteristics was found. From these facts, it was inferred that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 11 has G protein-coupled receptor activity.
  • dn afo rm33729 consists of 3336 bases as shown in SEQ ID NO: 12, of which base numbers 661 to 2196 are open reading frames (including a stop codon).
  • the amino acid sequence predicted from the open reading frame consists of 511 amino acid residues (SEQ ID NO: 28).
  • amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 12 was subjected to a protein feature search using HMMP FAM, and the G protein was conjugated to the amino acid sequence encoded by nucleotide numbers 796 to 1548 in SEQ ID NO: 12.
  • a sequence (a nucleotide sequence entered as “7 tm-1” in P f am) showing the characteristics of the type receptor was found.
  • AX3755332 described above is a human GPCR, which is a patent information (W0198330-A927-DEC-2001) in the database, and has been shown to be activated by angiopeptin.
  • Angiopeptin is an analog of somatostatin and is shown to inhibit atherosclerosis in the coronary artery of the egret, based on literature information (Atherosclerosis 1989 Aug; 78 (2-3): 229-36) in the database. . From these facts, it is inferred that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 12 is a G protein-coupled receptor for somatostatin-like ligand, has GPCR activity, and is associated with arterial sclerosis and cancer. Was.
  • dnaform48477 was composed of 3379 bases, of which base numbers from 108 to 1184 were open reading frames (including a stop codon).
  • the amino acid sequence predicted from the open reading frame consists of 358 amino acid residues (SEQ ID NO: 29).
  • a homology search was performed on the amino acid sequence encoded by SEQ ID NO: 13 using BLAST, and it was found in the SPTR protein database (integrated SWI SS-PROT protein sequence database and TrEMBL nucleic acid translation database).
  • amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 13 was subjected to a protein feature search using HMMP FAM. (A nucleotide sequence entered as "7 tm-1" in P f am) was found.
  • C5L2 is expressed in granulocytes and undivided dendritic cells from literature information in the database (Mol Immunol 2000 Jun; 37 (8): 407-12), and C3a And homology with C5a chemoattractant receptor have been shown to be high. From these facts, it was presumed that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 13 had a G protein-coupled receptor activity related to chemotaxis.
  • dnaform54572 consists of 3005 bases, of which base numbers 218 to 970 have an open reading frame.
  • the amino acid sequence predicted from the open reading frame consists of 250 amino acid residues (SEQ ID NO: 30).
  • a homology search was performed on the amino acid sequence encoded by SEQ ID NO: 14 using BLAST, and a SPTR protein database (integrated SWI SS-PROT protein sequence database and TrEMBL nucleic acid translation database) during, (i) data base registration mark AF 1 1 1088, B ostaurus 1 atrophi 1 in 3 splicevariantabbf, e- value: hit with 3 X 10- 51, also 302 Amino acids 37% degree of coincidence over the residues .
  • the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 14 was searched for protein characteristics using HM MPF AM. Array indicating (Base sequence entered as “7 tm—2” in P f am). From the literature information in the database (FEBS Lett 1999 Jan 29; 443 (3): 348-52), latrophilin 3 splice variant abbf indicates that Latrophilin is a receptor for alpha-latrotoxin, a potent prosynaptic toxin. Latrophilin-3 has been shown to be specifically expressed in the brain. From these facts, it was inferred that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 14 is an orphan G protein-coupled receptor and has activity in the brain and the like.
  • dnafo rm29069 consisted of 3034 bases, of which base numbers 1557 to 2489 were open reading frames (including a stop codon).
  • the amino acid sequence predicted from the open reading frame consists of 310 amino acid residues (SEQ ID NO: 31).
  • the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 15 was subjected to a protein feature search using HMMP FAM. Show body characteristics Sequence (base sequence entered as “7 tm—2” in P f am), and the amino acid sequence encoded by base Nos. 1647–1790 contains Latrophi 1 in / C L-1-like GPS doma in (Base sequence that is entered as “GP S” in P fa).
  • dnafo rm45616 was composed of 2774 bases, of which base numbers 311 to 1210 were open reading frames (including a stop codon).
  • the amino acid sequence predicted from the open reading frame consists of 299 amino acid residues (SEQ ID NO: 32).
  • GPR 34 is analogous to the platelet-activating factor (PAF) receptor from literature information in the database (BiochimBiophys Acta 1999 Jul 7; 1446 (1-2): 57-70). It has been shown to be abundantly expressed in various human and mouse organs. Also, from the literature information in the database (um Genet 2000 Jul; 107 (1): 89-91), GPR34 is located near the causal locus of congenital night blindness (CSNB) on the X chromosome and is expressed in the eyes, It has been shown not to be the causative gene of CSNB. From these results, it was presumed that the protein consisting of the amino acid sequence shown in SEQ ID NO: 32 had a novel G protein-coupled receptor activity.
  • PAF platelet-activating factor
  • Tissue expression analysis was performed as described in Miki, R., et al., Proc. Natl. Acad. Sci. USA, 98, 2199-2204 (2001).
  • the detection sensitivity of this DNA microarray was 1 to 3 copies of mRNA per cell.
  • the signal intensity of clones with approximately 80% match with the target sequence was one-tenth that of clones with perfect sequence match.
  • the signal intensity of clones with less than 80% match with the target sequence was reduced at the background level.
  • mice 49 tissues of fetal, neonatal and adult C57BL / 6J mice (kidney, brain, spleen, lung, liver, testis, kidney, stomach, small intestine, colon, cecum, placenta, heart, tongue, thymus, breast Glands (1st day of pregnancy), cerebellum, medulla oblongata, olfactory brain, epididymis, eyes, cortex, follicles, uterus, ovaries and ovary (1st day of pregnancy), bones, muscles, mammary glands (10th day of lactation) ), Whole 10-day-old fetus, 11-day-old fetal whole body, 13-day-old fetal whole body, 11-day-old fetal head, 12-day-old fetal head, 13-day-old fetal head, 15-day-old fetal head Part, 16 days old fetal head, 17 days old fetal head Part, 16-day-old fetal lung, 13-
  • a CyDye-labeled cDNA probe was prepared by mixing a blocking solution consisting of 0 ⁇ l mouse C ⁇ t1 DNA, 5.1 ⁇ l of 20 XSSC, and 0.91 of 10% SDS. .
  • the DNA microarray was washed with 2 ⁇ SSC and 0.1% SDS, and then rinsed with 1 ⁇ SSC for 2 minutes and with 0.1 ⁇ SSC for 2 minutes.
  • the microarray was scanned using a ScanA 500 000 confocal laser scanner and the images were analyzed on an I MAGENE (BioDiversive).
  • the mRNA level (Cy3 label) in each tissue is expressed as the ratio (Cy3 / Cy5) to the reference 17.5-day-old fetal whole-body mRNA level (Cy5 label) as a logarithm (1 og). 2 ) in displayed. That is, if the expression level of mRNA corresponding to the full-length cDNA of each mouse to be analyzed is larger in each tissue than in the reference tissue, a positive value is used, if it is smaller, a negative value, and if it is equal, Indicated by 0. Experiments were performed twice independently to increase the accuracy of the data, and reproducible results were used. The results are shown in Table 1.
  • the results of expression analysis using a DNA microarray are considered to be experimental errors if the value is increased or decreased by a factor of about two. It was interpreted that the amount of mRNA to be analyzed in the tissue was more than twice as large as the mRNA amount of the whole fetal body at 17.5 days of age, and that the amount was significantly increased. Conversely, if the value of the expression analysis result in a certain tissue is less than or equal to -1, the amount of the mRNA to be analyzed in the tissue is one-half of that of the control 17.5-day-old fetus whole body. The following were interpreted as significantly reduced.
  • the difference between the values in each tissue when the difference between the values in each tissue is 1, the mRNA level is 2 times, and when the difference is 2, the mRNA level is 4 times. If the difference between the values is -1, the mRNA amount is 1Z 2 times, and if it is _2, the mRNA amount is 1/4 times.
  • Mouse cDNA clones belonging to the same cluster as the DNA spotted on the microarray and having a region having a nucleotide sequence identity of at least 80% with at least 200 bases are also shown in Table 1 And the numerical value of the measurement result of the DNA spotted on the microarray was used instead.
  • dnaform29069 FANTOM NO3830420G05 -0.494198 -0.204861 As can be seen from Table 1, dnaform 3 4 1 4 7 has increased fluorescence in the spleen and uterus compared to the control (17.5 day fetal whole body) The expression was similar to that of the control in testis and kidney, but the expression level was reduced as a whole as compared to the control. The expression of dnaform 42851 increased in muscle and spleen, increased in cerebellum, colon, and placenta compared to controls, and was equivalent in brain. dnaform 2 9 8 9 4 is more strongly expressed in the spleen compared to controls Increased, and also increased in the brain.
  • dnafo rm32567 increased strongly in spleen and brain, increased in uterus and testis, and tended to increase in liver and colon.
  • dn afo rm54572 is up-regulated in spleen, adipocytes, and 10-day-old neonatal cerebellum as compared to control, and is expressed to the same extent as control in brain, but is up-regulated overall as compared to control There was a tendency.
  • dnaio rm29069 increased strongly in bone and placenta and tended to increase in spleen and muscle as compared to control.
  • PCR was performed according to a standard method (Higuchi R, et al., Biotechnology, 11: 1026-30 (1993)). Was used to perform tissue expression analysis.
  • Total RA was extracted from 19 tissues of the following mice (Kazuo Moriwaki, 1st Edition, Molecular Medicine Separate Volume, Vol. 36 “Model Animals with Spontaneous Diseases”, Nakayama Shoten, 1999), and oligo dT was used as a primer. CDNA synthesis was performed using reverse transcriptase. (a) Tissue of normal mice and tissue of diabetic model mice
  • the quantification results were corrected using Glyceraldehyde 3-phosphate dehydrogenase (GAP DH) as the internal standard [5 standards]. That is, the expression level (copy number / ⁇ 1) of the target gene in each tissue is divided by the GAPDH expression level (copy number / 1), and the result is raised to a constant (1 ⁇ 10 6 ) (Note: 10 to the sixth power). And displayed.
  • GAP DH Glyceraldehyde 3-phosphate dehydrogenase
  • dnafo rm33729 As is evident from Table 2, expression of dnafo rm33729 was high in eyes, expression was observed in lung, fat and knee, and decreased in colon cancer.
  • the expression of dnaf 0 rm456 16 was high in the lung, and was observed in the viscera, fat, and colon, but decreased in diabetic knee and colon cancer.
  • dnafo rm47734 was highly expressed in brain tissues, especially in the frontal cortex and hippocampus, suggesting a relationship with memory learning, and was also observed in knees, fat, and bone marrow.
  • dnafo rm48477 was highly expressed in bone marrow and lung, and increased in diabetic adipose tissue.
  • the cDNA of the above clone and the protein encoded by the cDNA can be applied to the treatment and diagnosis of diabetes and cancer. Further, the protein encoded by the cDNA may be involved in a disease relating to a tissue in which the mRNA expression is varied or a tissue having a high mRNA expression level as described above.
  • Example 7 Function prediction (1) dn afo rm34 147
  • Example 4 (1) From the results of Example 4 (1), it is known that the proteins encoded by the present cDNA are similar proteins involved in testis development, function maintenance, and fertility.
  • the expression of the protein encoded by the cDNA tends to increase in the viscera and uterus as compared to the control (17.5-day-old fetal whole body), The expression was similar to that of the control in such as kidney.
  • the protein encoded by this cDNA has functions related to infertility, contraception, and diabetes, because it is involved in testis development and maintenance, function in fertility, and is expressed in the uterus. It was considered useful for the analysis of these therapeutic agents.
  • Example 4 (2) From the results of Example 4 (2), Examples 5 and 6, the GPCR that the protein encoded by this cDNA is expressed in the brain, induced to be expressed by dalcocorticoid, and enhanced when the psychotropic drug is stopped And homology was higher.
  • this protein was considered to be useful for developing drugs for withdrawing from drug dependence, schizophrenia, depression, anxiety, Alzheimer's disease, and Parkinson's disease.
  • Example 4 (3) From the results of Example 4 (3), Examples 5 and 6, the protein encoded by this cDNA is expressed in the brain, is induced to be expressed by dalcocorticoid, and has a GPCR that enhances the expression when the psychotropic drug is stopped. The homology was high. For these reasons, it was predicted that the protein encoded by this cDNA would be involved in neural adaptation to stress and psychostimulants. Therefore, this protein was considered to be useful for the development of therapeutic drugs for withdrawal from drug dependence, schizophrenia, depression, anxiety, Alzheimer's disease, Parkinson's disease, and the like.
  • Example 5 the protein encoded by the cDNA The protein was expressed in the brain, induced to be expressed by dalcocorticoids, and had a high homology with GPCRs, whose expression was enhanced when psychotropic drugs were stopped. In addition, the expression was increased in muscle and spleen compared to control, and increased in cerebellum, colon and placenta, and was similar in brain and the like.
  • this protein was considered to be useful for the development of a drug for withdrawal from drug dependence, schizophrenia, depression, anxiety, Alzheimer's disease, Parkinson's disease, and diabetes.
  • Example 4 From the results of Example 4 (5), and Examples 5 and 6, the protein encoded by the present cDNA was similar to GPR82 expressed in the colon. Based on these findings, it was considered that this protein has functions related to Crohn's disease, irritable colitis, gastrointestinal tract function regulation, cancer, etc., and is useful for the development of these therapeutic agents.
  • Example 4 (6) and Examples 5 and 6 the protein encoded by the present cDNA is similar to human TGR21, and is also found in brain tissue, especially in the frontal cortex and hippocampus. High expression was observed in spleen and fat.
  • the protein has functions related to Alzheimer's disease, Parkinson's disease, chorea, ischemic brain disease, schizophrenia, depression, anxiety, and diabetes. It was considered useful for development.
  • Example 4 (7) and Examples 5 and 6 the protein encoded by this cDNA is a human GPR103 with high homology between neuropeptide FF2, neuropeptide Y2 and galanin GalRl receptor and the transmembrane domain.
  • FF2 neuropeptide FF2
  • Y2 neuropeptide Y2
  • galanin GalRl receptor galanin GalRl receptor
  • this protein is useful for schizophrenia, depression, anxiety, parkin It has functions related to Sohn's disease, Alzheimer's disease, ischemic brain disease, diabetes, and endocrine diseases, and was considered to be useful as a therapeutic drug for these.
  • Example 4 From the results of Example 4 (8) and Examples 5 and 6, the protein encoded by this cDNA was converted to human GPR103, which has high homology between neuropeptide FF2, neuropeptide Y2 and the galanin GalRl receptor and the transmembrane region. Was similar. It was also expressed in the cerebral cortex, pituitary gland, thalamus, hypothalamus, basal forebrain, midbrain, and lung. From these facts, this protein has functions related to schizophrenia, depression, anxiety, Parkinson's disease, Alzheimer's disease, ischemic brain disease, diabetes, and endocrine disease, and is useful as a therapeutic drug for these. It was thought that there was.
  • Example 4 (9) and Examples 5 and 6 the protein encoded by this cDNA is a human GPR103 with high homology between neuropeptide FF2, neuropeptide Y2 and galanin GalRl receptor and transmembrane domain.
  • This protein has functions related to schizophrenia, depression, anxiety, Parkinson's disease, Alzheimer's disease, ischemic brain disease, diabetes, and endocrine disease, and is useful as a therapeutic drug for these. It was thought that there was.
  • Example 4 (10) and Examples 5 and 6 the protein encoded by the present cDNA is a human GPR having high homology with the neuropeptide FF2, neuropeptide Y2 and galanin GalRl receptor and the transmembrane region. It was similar to 103. It was also expressed in the cerebral cortex, pituitary gland, thalamus, hypothalamus, basal forebrain, midbrain, and pons. From these facts, this protein has functions related to schizophrenia, depression, anxiety, Parkinson's disease, Alzheimer's disease, ischemic brain disease, diabetes, and endocrine disease, and is useful as a therapeutic drug for these. It was thought that there was.
  • Example 4 (1 1) dnafo rm32567 From the results of Example 4 (1 1) and Examples 5 and 6, the protein encoded by the present cDNA increased strongly in the kidney and brain, increased in the uterus and testis, and increased in the liver compared to the control. And the colon tended to increase.
  • this protein has functions related to diabetes, schizophrenia, depression, anxiety, Parkinson's disease, Alzheimer's disease, ischemic brain disease, infertility, contraception, and liver disease. It was considered as useful.
  • Example 4 From the results of Example 4 (1 2), Examples 5 and 6, the protein encoded by the present cDNA is similar to human AX 375432, and AX 375432 is activated by the somatostatin analog angiopeptin, Atherosclerosis was suppressed. In addition, the expression of this protein was high in the eyes, and was observed in lung, fat, and spleen, and decreased in colon cancer.
  • the present protein has functions related to atherosclerosis, cancer, diabetes, glycemic retinopathy, and ophthalmic diseases, and is useful for the development of these therapeutic agents.
  • Example 4 From the results of Example 4 (13) and Examples 5 and 6, the protein encoded by this cDNA was found to be resistant to C5L2 and chemotaxis expressed in granulocytes and undifferentiated dendritic cells. Similar to the GP CR involved. The expression of this protein was high in bone marrow and lung, and increased in diabetic adipose tissue.
  • this protein has functions related to immune diseases, inflammatory diseases, allergic diseases, respiratory diseases such as lungs and trachea, and diabetes, and is useful for the development of these therapeutic agents. It was considered.
  • Example 4 From the results of Example 4 (14), Examples 5 and 6, the protein encoded by this cDNA is similar to brain-specific latrophilin 3, which is a splicing variant of the exogenous toxin alpha-latrotoxin receptor Latrophilin. I was The protein Was increased in the knee, adipocytes, and 10-day-old neonatal cerebellum as compared to the control, and was expressed to the same extent in the brain and the like.
  • this protein has functions related to endocrine diseases such as schizophrenia, depression, anxiety, Parkinson's disease, Alzheimer's disease, chorea and diabetes, and is useful for the development of these therapeutics. It was thought that there was.
  • Example 4 the protein encoded by the present cDNA was similar to Cyt 28 cloned from mouse hematopoietic stem cells.
  • GPR56 a similar protein, is strongly expressed in the thyroid gland, heart, and brain, is particularly strongly expressed in the hippocampus and thalamus in the brain, and has an extracellular mucin-like structure.
  • the present protein increased strongly in bone and placenta, and tended to increase in knee and muscle compared to control.
  • this protein has functions related to endocrine diseases such as osteoporosis and diabetes, schizophrenia, depression, anxiety, Parkinson's disease, Alzheimer's disease, immune diseases, inflammatory diseases, and allergic diseases. However, it was considered useful for the development of these therapeutic agents.
  • Example 4 From the results of Example 4 (16) and Examples 5 and 6, the GPR34 gene, which is similar in the protein encoded by this cDNA, is on the X chromosome, and is abundantly expressed in various organs. It was found that there was weak homology with the body.
  • the present protein has functions related to respiratory diseases (such as asthma), diabetes, cancer, and the like, and is useful for the development of these therapeutic agents.
  • Industrial applicability Since the protein of the present invention and the DNA encoding the same have the above-described G protein-coupled receptor activity, a substance that regulates the activity can be screened using the protein or the DNA encoding the protein. It can also be used for the development of a medicament capable of acting on diseases associated with the protein.
  • G-protein coupled receptors are membrane protein receptors that transmit signals from various extracellular ligands via G proteins.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cell Biology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The base sequences of cDNA clones involved in a catalogued full-length cDNA library are analyzed. Concerning cDNA clones having novel sequences from among those analyzed above, the physiological activities (functions) of proteins encoded thereby are specified. Based on these physiological activities (functions), a method of utilizing the proteins and DNAs encoding the same is proposed. Namely, the following proteins (a) and (b), DNAs encoding the same and a method of utilizing the same are provided: (a) a protein comprising an amino acid sequence represented by any of SEQ ID NOS:17 to 32; and (b) a protein comprising an amino acid sequence derived from an amino sequence represented by any of SEQ ID NOS: 17 to 32 by deletion, substitution and/or addition of one to several amino acids and having a G protein-coupled receptor activity.

Description

明細書 新規タンパク質及ぴそれをコードする DNA 技術分野  Description Novel proteins and their technical fields

本発明は、 新規なタンパク質、 該タンパク質をコードする DNA、 該タンパク 質をコードする完全長 c DNA、 該 DNAを有する組換えベクター、 該 DNAの 部分配列から成るオリゴヌクレオチド、 該 DN Aを導入した遺伝子導入細胞、 及 ぴ該タンパク質に特異的に結合する抗体等に関する。 背景技術  The present invention has introduced a novel protein, a DNA encoding the protein, a full-length cDNA encoding the protein, a recombinant vector having the DNA, an oligonucleotide comprising a partial sequence of the DNA, and the DNA. The present invention relates to a transgenic cell, an antibody specifically binding to the protein, and the like. Background art

c DNAの取得及びその塩基配列解析は、 生体内に発現するタンパク質の生理 活性を解析し、 その活性に基づくタンパク質の利用方法を開発するうえで不可欠 である。 さらに、 全遺伝子種に対応する完全長 c DN Aをカタログ化したライブ ラリーの作製は、 ヒトゲノムプロジェクトの重要な課題の一つである。 カタログ 化したライブラリーとは、 ライブラリーに含まれる c DNAに重複がないという 意味であり、 各 c DN Aが 1種類ずつ含まれているライブラリーのことである。 完全長 c DNAクローニング法については、 特開平 9一 248 187号公報及 ぴ特開平 10— 127291号公報に記載されている。 この方法は、 mRNAの 5'キャップサイトに存在するジオール構造にタグになる分子を結合させる工程、 前記タグ分子を結合させた mRNAを鍀型とし、 o l i g o dTをプライマー として逆転写により RNA— DNA複合体を作製し、 この複合体の内、 mRNA の完全長に対応する DNAを有するものをタグ分子の機能を利用して分離するェ 程を含むことを特徴とする方法である。  Obtaining cDNA and analyzing its base sequence are essential for analyzing the physiological activity of a protein expressed in a living body and developing a method of using the protein based on the activity. In addition, creating a library that catalogs full-length cDNAs for all genotypes is one of the important tasks of the Human Genome Project. The cataloged library means that there is no duplication in the cDNAs contained in the library, and is a library containing one kind of each cDNA. The full-length cDNA cloning method is described in JP-A-9-1248187 and JP-A-10-127291. In this method, a tag molecule is bound to a diol structure present at the 5 ′ cap site of the mRNA. The mRNA bound to the tag molecule is type II, and RNA-DNA complex is formed by reverse transcription using oligo dT as a primer. A method comprising preparing a body, and separating a complex having a DNA corresponding to the full-length mRNA from the complex using the function of a tag molecule.

また効率のよい逆転写法として、 铸型が高次構造を形成しないような高温で行 うための方法も開発されている (特開平 10— 84961号公報)。 さらに、合成 された完全長 c DNAライブラリーに含まれる DNA断片についてその鎖長に関 わらず一律にクローニングすることができるクローニングベクターも開発されて いる (特開平 1 1一 9273号公報)。 Further, as an efficient reverse transfer method, a method for performing the method at a high temperature such that the type III does not form a higher-order structure has been developed (Japanese Patent Application Laid-Open No. H10-84961). Furthermore, regarding the DNA fragments contained in the synthesized full-length cDNA library, Regardless, a cloning vector that can be cloned uniformly has been developed (Japanese Patent Application Laid-Open No. 11-9273).

このような技術により作製された完全長 cDN Aライブラリ一は、 ライブラリ 一の個々の要素として全て均等に異なるものが含まれている訳ではなく、 存在割 合の高いクローンや逆に極微量にしか存在しないクローンもある。 この極微量に しか存在しないクローンは新規である可能性が高いため、 このようなクローンを 濃縮するためのサブトラクション法ゃノーマライゼーション法も開発されている (特開 2000— 3 2508 0号公報; Carninci, P. et al. , Genomics, 37, 327-336(1996))。  The full-length cDNA library produced by such a technique does not include all the elements that are different evenly as individual elements of the library. Some clones do not exist. Since a clone present in only a trace amount is likely to be novel, a subtraction method and a normalization method for enriching such clones have also been developed (Japanese Patent Application Laid-Open No. 2000-325080; Carninci, P. et al., Genomics, 37, 327-336 (1996)).

かくして得られるカタログ化された完全長 c DNAライブラリーの各クローン について、 公知の方法により塩基配列の解析を行えば、 その塩基配列は同定され るが、 該 c DNAがコードするタンパク質の生理活性は依然不明のままである。 発明の開示  When the nucleotide sequence of each clone of the cataloged full-length cDNA library thus obtained is analyzed by a known method, the nucleotide sequence is identified, but the physiological activity of the protein encoded by the cDNA is It remains unknown. Disclosure of the invention

本発明は、 カタログ化された完全長 cDNAライブラリーに含まれる cDNA クローンの塩基配列を解析し、 このうち配列が新規なものについては、 これがコ ードするタンパク質の生理活性を特定し、 該生理活性に基づくタンパク質および それをコードする DNAの利用方法を提案することを目的とする。  The present invention analyzes the nucleotide sequence of a cDNA clone contained in a cataloged full-length cDNA library, and among those having a novel sequence, identifies the physiological activity of the protein encoded by the nucleotide sequence. The purpose of the present invention is to propose a method of using a protein based on activity and a DNA encoding the protein.

本発明者らは、 マウス完全長 c DNAライブラリ一中の c DNAクローンが有 する塩基配列を解析し、該配列の相同性に基づきデータベースを検索したところ、 該配列中に特定の機能を有するタンパク質に特異的な配列を見出した。 また、 こ れらの c DNAの各組織における発現量を解析した。 本発明は、 これらの知見に 基づいて成し遂げられたものである。  The present inventors analyzed the nucleotide sequence of a cDNA clone in a mouse full-length cDNA library and searched a database based on the homology of the sequence, and found that a protein having a specific function was found in the sequence. Was found. The expression levels of these cDNAs in each tissue were analyzed. The present invention has been achieved based on these findings.

すなわち本発明によれば、以下の(1) 〜(15) に記載の発明が提供される。  That is, according to the present invention, the following inventions (1) to (15) are provided.

(1) 以下の (a) または (b) のタンパク質。  (1) The following protein (a) or (b):

(&)配列番号17〜32のいずれかに記載のアミノ酸配列からなるタンパク質。 (&) A protein comprising the amino acid sequence of any one of SEQ ID NOs: 17 to 32.

(b) 配列番号 17〜32のいずれかに記載のアミノ酸配列において 1若しくは 数個のアミノ酸が欠失、 置換及び/または付加されたアミノ酸配列からなり、 か つ Gタンパク質共役型受容体活性を有するタンパク質。 (b) in the amino acid sequence of any one of SEQ ID NOs: 17 to 32, A protein comprising an amino acid sequence in which several amino acids have been deleted, substituted and / or added, and having G protein-coupled receptor activity.

(2) 上記 (1) に記載のタンパク質をコードする DNA。  (2) DNA encoding the protein of (1) above.

(3) 上記 (1) に記載のタンパク質をコードする完全長 cDNA。  (3) A full-length cDNA encoding the protein described in (1) above.

(4) 以下の (a) 、 (b)又は (c) のいずれかの DNA。  (4) Any one of the following DNAs (a), (b) or (c):

(a) 配列番号 1〜1 6のいずれかに記載の塩基配列を有する DNA。  (a) DNA having the nucleotide sequence of any one of SEQ ID NOs: 1 to 16.

(b) 配列番号 1〜16のいずれかに記載の塩基配列において、 1若しくは数個 の塩基が欠失、 置換及び Zまたは付加された塩基配列を有し、 かつ Gタンパク質 共役型受容体活性を有するタンパク質をコードする DNA。  (b) in the nucleotide sequence of any one of SEQ ID NOs: 1 to 16, the nucleotide sequence has one or several nucleotides deleted, substituted and Z or added, and has G protein-coupled receptor activity. DNA encoding a protein having

(c) 配列番号 1〜16のいずれかに記載の塩基配列あるいはその相補配列を有 する DNAとストリンジェントな条件下でハイブリダイズすることができる塩基 配列を有し、 かつ Gタンパク質共役型受容体活性を有するタンパク質をコードす る DNA。  (c) a G protein-coupled receptor having a nucleotide sequence capable of hybridizing under stringent conditions to DNA having the nucleotide sequence of any of SEQ ID NOs: 1 to 16 or a sequence complementary thereto, and DNA encoding an active protein.

(5) 上記(2)〜(4) のいずれかに記載の DN Aを含む組換えベクター。 (5) A recombinant vector containing the DNA according to any one of (2) to (4).

(6) 上記 (2) 〜 (4) のいずれかに記載の DN Aまたは上記 (5) に記 載の組み換えべクタ一を導入した遺伝子導入細胞または該細胞からなる個体。 (6) A transgenic cell into which the DNA according to any of (2) to (4) or the recombinant vector according to (5) is introduced, or an individual comprising the cell.

(7) 上記 (6) に記載の細胞により産生される、 上記 (1) に記載のタン パク質。  (7) The protein according to (1), which is produced by the cell according to (6).

(8) 上記 (2) 〜 (4) のいずれかに記載の DNAの塩基配列中の連続し た 5〜100塩基と同じ配列を有するセンスオリゴヌクレオチド、 当該センスォ リゴヌクレオチドと相補的な配列を有するアンチセンスオリゴヌクレオチド、 及 び、 当該センス又はアンチセンスオリゴヌクレオチドのオリゴヌクレオチド誘導 体から成る群から選ばれるオリゴヌクレオチド。  (8) A sense oligonucleotide having the same sequence as 5 to 100 consecutive nucleotides in the base sequence of the DNA according to any of (2) to (4) above, and having a sequence complementary to the sense oligonucleotide. An oligonucleotide selected from the group consisting of an antisense oligonucleotide and an oligonucleotide derivative of the sense or antisense oligonucleotide.

(9) 上記 (1) または (7) に記載のタンパク質に特異的に結合する抗体 あるいはその部分フラグメント。  (9) An antibody or a partial fragment thereof that specifically binds to the protein of (1) or (7).

(10) 抗体がモノクローナル抗体である上記 (9) に記載の抗体。  (10) The antibody according to the above (9), wherein the antibody is a monoclonal antibody.

(1 1) モノクローナ /レ抗体が上記 ( 1 ) または ( 7 ) に記載のタンパク質 の Gタンパク質共役型受容体活性を中和する作用を有することを特徴とする上記 (10) に記載の抗体。 (1 1) The monoclonal / antibody is a protein according to (1) or (7) above. The antibody according to the above (10), which has an action of neutralizing the G protein-coupled receptor activity of the above.

(12) 上記(1) または(7) に記載のタンパク質と被検物質を接触させ、 該被検物質による該タンパク質が有する活性の変化を測定することを特徴とする、 該タンパク質の活"生調節物質のスクリーユング方法。  (12) contacting the protein according to (1) or (7) with a test substance, and measuring a change in the activity of the protein caused by the test substance; Screening method for modulators.

(13) 上記 (6) に記載の遺伝子導入細胞と被検物質を接触させ、 該細胞 に導入されている DNAの発現レベルの変化を検出することを特徴とする、 該 D N Aの発現調節物質のスクリーニング方法。  (13) contacting the gene-transfected cell according to (6) with a test substance, and detecting a change in the expression level of the DNA introduced into the cell; Screening method.

(14) 上記 (1) に記載のタンパク質のアミノ酸配列から選択される少な くとも 1以上のァミノ配列情報、 および Zまたは上記 (2) 〜 (4) のいずれか に記載の DNAの塩基配列から選択される少なくとも 1以上の塩基配列情報を保存 したコンピュータ読み取り可能記録媒体。  (14) At least one or more amino acid sequence information selected from the amino acid sequence of the protein described in (1) above, and Z or the DNA base sequence described in any of (2) to (4) above. A computer-readable recording medium storing at least one or more selected base sequence information.

(15) 上記 (1) に記載のタンパク質、 および/または上記 (2) 〜 (4) のいずれかに記載の D N Aを結合させた担体。 発明を実施するための最良の形態  (15) A carrier to which the protein according to (1) and / or the DNA according to any of (2) to (4) are bound. BEST MODE FOR CARRYING OUT THE INVENTION

以下、 本発明をさらに詳細に説明する。  Hereinafter, the present invention will be described in more detail.

( 1 ) 完全長 c DN Aの取得及び塩基配列の解析  (1) Acquisition of full-length cDNA and analysis of nucleotide sequence

本発明の DNAは、 配列番号 17〜32に記載のアミノ酸配列からなるタンパ ク質、 またはアミノ酸配列において、 1若しくは数個 (ここで言う数個の数は特 には限定されないが、 例えば 20個以下、 好ましく 15個以下、 より好ましくは 10個以下、 さらに好ましくは 5個以下を意味する) のアミノ酸残基の置換、 欠 失、 挿入、 付加、 若しくは逆位を含むアミノ酸配列からなり、 かつ Gタンパク質 共役型受容体活性を有するタンパク質をコードし得るものであれば如何なるもの であってもよい。 具体的には、 該アミノ酸配列をコードする翻訳領域のみでも、 あるいはその c DNAの全長を含むものでもよい。  The DNA of the present invention may be a protein or an amino acid sequence comprising the amino acid sequence of SEQ ID NOs: 17 to 32, or one or several (the number is not particularly limited; And preferably 15 or less, more preferably 10 or less, and even more preferably 5 or less) amino acid residues containing substitutions, deletions, insertions, additions, or inversions, and G Any protein can be used as long as it can encode a protein having protein-coupled receptor activity. Specifically, it may be only the translation region encoding the amino acid sequence, or may include the full length of its cDNA.

具体的には、 c DNAの全長を含む DNAとしては、 例えば、 配列番号 1〜1 6のいずれかに記載の塩基配列からなる DNA等が挙げられる。 また、 その翻訳 領域としては、 Specifically, the DNA containing the full-length cDNA includes, for example, SEQ ID NOs: 1 to 1 And DNA comprising the nucleotide sequence according to any one of the above (6). Also, as the translation area,

配列番号 1の塩基番号 107〜 1858に示される配列、 A sequence represented by base numbers 107 to 1858 of SEQ ID NO: 1,

配列番号 2の塩基番号 507〜 1874に示される配列、 A sequence represented by base numbers 507 to 1874 of SEQ ID NO: 2,

配列番号 3の塩基番号 9〜 1169に示される配列、 A sequence represented by base numbers 9 to 1169 of SEQ ID NO: 3,

配列番号 4の塩基番号 474〜 1841に示される配列、 A sequence represented by base numbers 474 to 1841 of SEQ ID NO: 4,

配列番号 5の塩基番号 281〜 1267に示される配列、 A sequence represented by base numbers 281 to 1267 of SEQ ID NO: 5,

配列番号 6の塩基番号 173〜 1456に示される配列、 A sequence represented by base numbers 173 to 1456 of SEQ ID NO: 6,

配列番号 7の塩基番号 53〜 1354に示される配列、 A sequence represented by base numbers 53 to 1354 of SEQ ID NO: 7,

配列番号 8の塩基番号 64〜 1 161に示される配列、 A sequence represented by base numbers 64 to 1161 of SEQ ID NO: 8,

配列番号 9の塩基番号 88〜1 185に示される配列、 A sequence represented by base numbers 88 to 1185 of SEQ ID NO: 9,

配列番号 10の塩基番号 160〜 1236に示される配列、 A sequence represented by base numbers 160 to 1236 of SEQ ID NO: 10,

配列番号 11の塩基番号 92〜 1207に示される配列、 A sequence represented by base numbers 92 to 1207 of SEQ ID NO: 11,

配列番号 12の塩基番号 661〜 2196に示される配列、 A sequence represented by base numbers 661 to 2196 of SEQ ID NO: 12,

配列番号 13の塩基番号 108〜 1 184に示される配列、 A sequence represented by base numbers 108 to 1184 of SEQ ID NO: 13,

配列番号 14の塩基番号 218〜 970に示される配列、 A sequence represented by base numbers 218 to 970 of SEQ ID NO: 14,

配列番号 15の塩基番号 1557〜 2489に示される配列、 A sequence represented by base numbers 1557 to 2489 of SEQ ID NO: 15,

配列番号 16の塩基番号 311〜 1210に示される配列、 A sequence represented by base numbers 311-1210 of SEQ ID NO: 16,

を挙げることができる。 Can be mentioned.

さらに上記の cDNAの全長でなくても、 上記翻訳領域とその 3, 及ぴ Zまた は 5' 端に隣接する、 翻訳領域の発現に最低限必要な部分を含むもの等も本発明 の DNAに含まれる。  Furthermore, even if not the full-length of the above-mentioned cDNA, those containing the above-mentioned translation region and a part adjacent to the 3, and Z or 5 'end thereof, which contains the minimum necessary for expression of the translation region, etc., are also included in the DNA of the present invention. included.

本発明の DN Aは、 これを取得できる方法であれば如何なる方法により取得し たものでもよいが、 具体的には、 例えば次の方法により取得することができる。 まず、 適当な動物、 好ましくは哺乳動物の組織等からそれ自体既知の通常用いら れる方法により niRNAを調製する。 次に、 この mRNAを铸型として c DNA を合成するが、 このとき完全長の c DNAを合成するために 5, キャップ (7MeG PPPN) サイトに特異的なジオール構造にタグになる分子を化学結合させ、 この m RNAを铸型として o l i g o d Tをプライマーとして逆転写した後に、 タグ 分子の機能を利用して完全長の cDNAのみを分離する方法 (特開平 9一 248 1 87号公報;特開平 10_ 1 27291号公報) を用いることが好ましい。 ま た、 逆転写の際には、 铸型が高次構造を形成して逆転写の効率が低下することを 阻止するために、 トレハロース等の存在下で、 耐熱性逆転写酵素を用いて高温下 で逆転写を行う方法 (特開平 10— 84961号公報) を用いるのが好ましい。 ここで、 高温下とは 40〜80°Cを意味する。 The DNA of the present invention may be obtained by any method as long as it can be obtained. Specifically, it can be obtained by the following method, for example. First, niRNA is prepared from a suitable animal, preferably a mammalian tissue or the like, by a method known per se and generally used. Next, using this mRNA as type I cDNA Although synthesize, 5 to synthesize c DNA of the full-length this time, a molecule consisting the tag-specific diol structures in the cap (7Me G PPP N) site is chemically bonded, the m RNA as铸型After reverse transcription using oligod T as a primer, it is possible to use a method of separating only full-length cDNA using the function of a tag molecule (JP-A-9-1248187; JP-A-10_127291). preferable. In addition, in the case of reverse transcription, in order to prevent the type III from forming a higher-order structure and reducing the efficiency of reverse transcription, the temperature is increased by using a thermostable reverse transcriptase in the presence of trehalose or the like. It is preferable to use a method of performing reverse transcription below (JP-A-10-84961). Here, high temperature means 40 to 80 ° C.

このようにして取得された c DNAは、 これを適当なクローニングベクターに 挿入してクローニングを行う。 ここで用いられるベクターとしては、 様々な鎖長 の DNAを一律にクローエングすることが可能な、 クローニングサイトの両末端 にリコンビナーゼ認識配列を有し、 感染以外の方法で宿主に挿入される直鎖状の ベクター (特開平 1 1一 9273号公報) が好ましく用いられる。 かくして得ら れる c DNAライブラリ一は、 全てのクローンが均一に存在している (以下、 こ れを 「カタログ化されている」 と称することがある) 訳ではなく、 このライブラ リー中に極微量にしか存在しないクローンこそ新規である確率が高い。 そこで、 このようなクロ ^"ンを濃縮するためのサブトラクション法ゃノーマライゼーショ ン法 (特開 2000— 325080号公報; Carninci, P. et al. , Genomics, 37, 327-336(1996)) を用いることが好ましい。  The cDNA thus obtained is inserted into an appropriate cloning vector for cloning. The vector used here has a recombinase recognition sequence at both ends of the cloning site that can uniformly clone DNA of various chain lengths, and is a linear vector that can be inserted into the host by a method other than infection. The vector (JP-A-119-1273) is preferably used. In the thus obtained cDNA library, not all clones exist uniformly (hereinafter, this may be referred to as “cataloged”). A clone that exists only in a plant has a high probability of being new. Therefore, a subtraction method for concentrating such clones and a normalization method (Japanese Unexamined Patent Publication No. 2000-325080; Carninci, P. et al., Genomics, 37, 327-336 (1996)) have been proposed. Preferably, it is used.

カタログ化された c DNAライブラリ一は、 それ自体既知の通常用いられる方 法により塩基配列の解析を行う。 本発明の DNAは、 cDNA全長の場合にはそ の末端 1 0 0ベースの配列について得られた塩基配列を、 B L A S T (http://www.ncbi.nlm.nih.gov/BLAST/ ; National Center of Biotechnology Information) を用いて、 NC B Iの G e n b a n k、 EMBL、 DDB J、 PD B等のデータベースについて検索し、 最も高い相同性を示す配列でも相同性が 3 The nucleotide sequence of the cataloged cDNA library is analyzed by a commonly used method known per se. In the case of the DNA of the present invention, in the case of full-length cDNA, the base sequence obtained from the sequence based on the terminal 100 is obtained by using BLAST (http://www.ncbi.nlm.nih.gov/BLAST/; National Center). of Biotechnology Information) and searched databases such as NCBI Genbank, EMBL, DDB J, and PDB.

0%以下であり、 かつ該 DNAの翻訳領域の全長について最も高い相同性を示す 配列でもその相同性が 40%以下であるものを新規として以下の解析に供するこ ととした。 このような完全長 cDNAの塩基配列を有する DNA、 その翻訳領域 としては、 例えば、 上記したものが挙げられる。 0% or less and shows the highest homology with respect to the entire length of the translated region of the DNA Sequences with a homology of 40% or less were newly submitted to the following analysis. Examples of a DNA having such a full-length cDNA base sequence and its translation region include those described above.

かくして取得された新規な塩基配列を、 BLAST (Basic local alignment search tool; Altschul, S. F. , et al. , J. Mol. Biol., 215, 403-410(1990)) に よる相同性検索 (homology search)や、 HMME R (隠れ Markovモデルによる配 列解析手法; Eddy, S. R. , Bio informatics 14, 755-763 (1998)) の機能群のひ とつである HMMPF AMによるタンパク質特徴検索 (profile search: http: //pfam. wustl. edu)等を行うことにより、 該塩基配列がコードするタンパ ク質の機能を推定することができる。  The novel nucleotide sequence obtained in this way is subjected to homology search (homology search) by BLAST (Basic local alignment search tool; Altschul, SF, et al., J. Mol. Biol., 215, 403-410 (1990)). ) And HMME R (sequence analysis method using hidden Markov model; Eddy, SR, Bioinformatics 14, 755-763 (1998)). : // pfam. Wustl. Edu), etc., to estimate the function of the protein encoded by the nucleotide sequence.

B LASTによる相同性検索においては、 検索の結果得られた相同性が十分有 意なヒット配列に付随する種々のァノテーション情報から、 解析対象としている クローンの機能を推定することができる。 ここで、 十分有意なヒット配列とは、 登録されている配列の触媒ドメイン部分と本発明の DN Aのこれに対応する部分 との i d e n t i t yが e— v a l u eとして 10_4以下のものか、 あるいは 3 0%以上のものを示す。 In the homology search by BLAST, the function of the clone to be analyzed can be estimated from various annotation information associated with the hit sequence in which the homology obtained as a result of the search is sufficiently significant. Here, a sufficiently significant hit sequence means that the identity between the catalytic domain portion of the registered sequence and the corresponding portion of the DNA of the present invention is less than 10 -4 as e-value, or 30 % Or more.

例えば、 上位にヒッ トした触媒ドメイン配列の多くが Gタンパク質共役型受容 体としての機能を確認されているものであるならば、 それと配列上類似である解 析対象クローンもまた同じ機能を持つであろうという予測が成り立つ。  For example, if many of the catalytic domain sequences hit at the top are confirmed to function as G protein-coupled receptors, clones to be analyzed that are sequence-similar to those will also have the same function. The expectation holds.

HMMPFAMでは、 P f a mというタンパク質プロファイルを集積したデー タベース中にあるェントリーが有する塩基配列の特徴を、 解析対象である塩基配 列が有するかどうかを洗い出す方法による解析が行われる。 プロファイルは一連 の同一特徴を持つタンパク質群から抽出されており、 一配列対一配列の全長に亘 る比較では明確化できなレ、機能でも、 配列中にその特徴領域があればこれを見出 し、 機能予測ができる。  In HMMPFAM, analysis is performed by a method for identifying whether or not a base sequence to be analyzed has the characteristics of the base sequence of an entry in a database in which a protein profile called Pfam is accumulated. Profiles are extracted from a series of proteins with the same characteristics, and cannot be clarified by comparing one-to-one sequences over their entire length. And function prediction.

かくして行われるタンパク質の機能予測の具体的な例として以下に説明する。 配列番号 1に記載の塩基配列がコードするアミノ酸配列は、 B LAS Tサーチ により G protein coupled receptor affecting testicular descent (Great)と e — v a l u e : 0 . 0、 5 7 9ァミノ酸残基で 9 9 %の相同性を有する。 A specific example of the prediction of the function of a protein thus performed will be described below. The amino acid sequence encoded by the nucleotide sequence of SEQ ID NO: 1 is a BLAST search. Has a 99% homology with G protein coupled receptor affecting testicular descent (Great) and e-value: 0.0, 579 amino acid residues.

また、 配列番号 1に示す塩基配列がコードするアミノ酸配列について、 HMM P F AMによるタンパク質特徴検索を行うと Low - density lipoprotein receptor domain class Aの特敷を示す酉己列 ( P f a mに 「ldl— recept— a」 としてエントリ 一される塩基配列) が見出され、 塩基番号 467 - 1186がコードするアミノ酸配列に 1 0力所にわたって Leucine Rich Repeatの特徴を示す配列 (P f a に 「LRR」 としてエントリーされる塩基配列) が見い出される。  In addition, a protein feature search using the HMM PFAM for the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 1 showed that the low-density lipoprotein receptor domain class A had a special feature (P fam: “ldl-recept — A sequence that shows the characteristics of Leucine Rich Repeat over 10 amino acids in the amino acid sequence encoded by nucleotides 467-1186 (entry as “LRR” in Pfa) Base sequence).

これらのことから配列番号 1に示す塩基配列がコードするタンパク質は Leucine Rich Repeatを含む Gタンパク質共役型受容体活性を有することが推測で き、 また、 相同性を有するタンパク質の機能から、 精巣の発達と機能を維持する 機能を有すると推測できる。  From these facts, it can be inferred that the protein encoded by the nucleotide sequence of SEQ ID NO: 1 has G protein-coupled receptor activity including Leucine Rich Repeat, and testis development based on the function of homologous proteins. It is inferred that it has the function of maintaining the function.

配列番号 2に記載の塩基配列がコードするアミノ酸配列は B L A S Tサーチに より PROBABLE G PROTEIN-COUPLED RECEPTOR GPR72 PRECURSOR (GLUCOCORTICOID - INDUCED RECEPTOR)と e— v a l u eが 5 X 1 0— 136、 2 4 0ァミノ酸残基で 1 0 0 %の相同性を有し、 また orphan G— protein coupled receptor GPR72と、 e— v a 1 u e : 1 X 1 0— 84で、 301アミノ酸残基に亘り 4 5 %の一致度で、 さらに glucocorticoid— induced receptorと、 e— v a l u e : 3 X 1 0一84で、 3 0 1ァ ミノ酸残基に亘り 4 4 %の相同性を有する。 More PROBABLE G PROTEIN-COUPLED RECEPTOR GPR72 PRECURSOR amino acid sequence encoded by the nucleotide sequence set forth in SEQ ID NO: 2 in BLAST searches (GLUCOCORTICOID - INDUCED RECEPTOR) and e- value is 5 X 1 0- 136, 2 4 0 Amino acid residue has a 1 0 0% homology with groups, also the orphan G- protein coupled receptor GPR72, e- va 1 ue: in 1 X 1 0- 84, with 4 5% degree of coincidence over the 301 amino acid residues a further glucocorticoid- induced receptor, e- value: in 3 X 1 0 one 84, has a 4 4% homology over 3 0 1 § amino acid residues.

また、 配列番号 2に示す塩基配列がコードするァミノ酸配列について、 HMM P F AMによるタンパク質特徴検索によれば Gタンパク質共役型受容体の特徴を 示す配列(P f a mに「7tm_ljとしてェントリーされる塩基配列)が見出される。 これらのこと力ゝら、 配列番号 2に示す塩基配列がコードするタンパク質はダル ココルチコィドで誘導される可能性のある Gタンパク質共役型受容体であること が推測される。 また、 相同性の高いタンパク質は、 抗精神薬の断薬時に発現が増 強する。  In addition, according to the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 2, a sequence showing the characteristics of the G protein-coupled receptor according to the protein feature search by HMM PFAM (the nucleotide sequence "7tm_lj From these results, it is presumed that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 2 is a G protein-coupled receptor that may be induced by dalcocorticoid. Highly homologous proteins are upregulated when antipsychotics are stopped.

配列番号 3に記載の塩基配列がコードするアミノ酸配列は B L A S Tサーチに より PROBABLE G- PROTEIN - COUPLED RECEPTOR GPR72 PRECURSOR (GLUCOCORTICOID- INDUCED RECEPTORと 5 X 10— 96の e— v a l u e、 1 73ァミノ酸残基で 100 % の相同性を ¾し、 Siた orphan G - protein coupled receptor GPR72と、 e— v a 1 u e : 2 X 10— 84で、 301アミノ酸残基に亘り 45%の相同性を有し、 さらに は、 glucocorticoid - induced receptorと、 e— v a 1 u e : 4X 10-84で、 3 01アミノ酸残基に亘り 4 '4%の相同性を有する。 The amino acid sequence encoded by the nucleotide sequence of SEQ ID NO: 3 was More PROBABLE G- PROTEIN - COUPLED RECEPTOR GPR72 PRECURSOR (GLUCOCORTICOID- INDUCED RECEPTOR and by ¾ 100% homology with 5 X 10- 96 of e- value, 1 73 Amino acid residues, Si was orphan G - protein coupled receptor and GPR72, e- va 1 ue: with 2 X 10- 84, it has a 45% homology over 301 amino acid residues, even more, glucocorticoid - and induced receptor, e- va 1 ue: 4X 10 -84 And has 4'4% homology over 301 amino acid residues.

配列番号 3に示す塩基配列がコードするアミノ酸配列について、 HMMP FA Mによるタンパク質特徴検索を行つたところ Gタンパク質共役型受容体の特徴を 示す配列(P f amに「7tm_lJとしてェントリーされる塩基配列)が見出される。 これらのことカゝら配列番号 3に示す塩基配列がコードするタンパク質はダルココ ルチコィドで誘導される可能性のある Gタンパク質共役型受容体活性を有すると 推測できる。 また、 相同性の高いタンパク質は、 抗精神薬の断薬時に発現が増強 する。  A protein characteristic search using HMMP FAM was performed on the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 3. A sequence showing the characteristics of the G protein-coupled receptor (base sequence that is entered as 7tm_lJ in P f am) It can be inferred that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 3 has a G-protein-coupled receptor activity that can be induced by dalcocotide. Higher proteins are upregulated when antipsychotics are stopped.

配列番号 4に記載の塩基配列がコードするアミノ酸配列は B LAS Tサーチに より PROBABLE G PROTEIN-COUPLED RECEPTOR GPR72 PRECURSOR (GLUCOCORTICOID- INDUCED RECEPTOR)と e— v a l u eが 5 X 10— 136、 240ァミノ酸残基で 10 0 %の相同性を有し、 また orphan G- protein coupled receptor GPR72と、 e - v a 1 u e : 1 X 10一84で、 301ァミノ酸残基に!:り 45 %の一致度で、 さらに glucocorticoid - induced receptorと、 e— v a l u e : 3 X 10— 84で、 301ァ ミノ酸残基に亘り 44%の相同性を有する。 Sequence amino acid sequence encoded by the nucleotide sequence set forth in ID NO: 4 B LAS T More search PROBABLE G PROTEIN-COUPLED RECEPTOR GPR72 PRECURSOR (GLUCOCORTICOID- INDUCED RECEPTOR) and e- value is 5 X 10- 136, 240 Amino acid residues in a 10 0% homology, also the orphan G- protein coupled receptor GPR72, e - va 1 ue: at 1 X 10 one 84, 301 Amino acid residue! : 45% degree of coincidence is, further glucocorticoid - and induced receptor, e- value: with 3 X 10- 84, has a 44% homology over 301 § amino acid residues.

また、 配列番号 4に示す塩基配列がコードするアミノ酸配列について、 HMM P F AMによるタンパク質特徴検索によれば G タンパク質共役型受容体の特徴を 示す配列(P f amに「7tm— 1」としてエントリーされる塩基配列)が見出される。 これらのことから配列番号 4に示す塩基配列がコードするタンパク質はダルコ コルチコィドで誘導される可能性のある Gタンパク質共役型受容体活性を有する ことが推測される。 また、 相同性の高いタンパク質は、 抗精神薬の断薬時に発現 が増強する。 配列番号 5に記載の塩基配列がコ一ドするァミノ酸配列は BLASTサーチに より G protein-coupled receptor (GPR82) geneと e— v a 1 u e力 5X 10— 159In addition, according to the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 4, a sequence showing the characteristics of a G protein-coupled receptor according to HMM PFAM protein search (“7tm-1” was entered in P f am Base sequence). From these facts, it is presumed that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 4 has a G protein-coupled receptor activity that may be induced by dalcocorticoid. In addition, the expression of highly homologous proteins increases when antipsychotic drugs are stopped. SEQ ID NO: G protein-coupled receptor (GPR82) More Amino acid sequence BLAST search base sequence is co one de described in 5 gene and e- va 1 ue force 5X 10- 159,

328ァミノ酸残基で 82 %で類似し、 HMMP F AMによるタンパク質特徴検 索によれば Gタンパク質共役型受容体の特徴を示す配列 (P f amに 「7tm_l」 と してエントリーされる塩基配列) が見出される。 A sequence that is similar to 82% at 328 amino acid residues and shows characteristics of a G protein-coupled receptor according to a protein feature search by HMMP FAM (base sequence entered as “7tm_l” in P f am) ) Is found.

これらのことから配列番号 5に示す塩基配列がコードするタンパク質は Gタン パク質共役型受容体活性を有することが推測される。 また、 相同性の高いタンパ ク質は、 結腸に多く発現している。  From these facts, it is presumed that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 5 has G protein-coupled receptor activity. In addition, proteins with high homology are frequently expressed in the colon.

配列番号 6に記載の塩基配列がコ一ドするァミノ酸配列は B LAS Tサーチに よれば TGR21 G— protein coupled receptor (GPCR)と e—v a l u e力 5 X 10一176According the Amino acid sequence nucleotide sequence is co one de set forth in SEQ ID NO: 6 in B LAS T Search TGR21 G- protein coupled receptor (GPCR) and e-value power 5 X 10 one 176,

442ァミノ酸残基で 73 %で類似し、 HMMP F AMによるタンパク質特徴検 索を行ったところ Gタンパク質共役型受容体の特徴を示す配列(P f amに「7tm 1」 としてエントリーされる塩基配列) が見出される。 A protein characteristic search by HMMP FAM, which is similar to 442 amino acid residues at 73%, shows a characteristic of G protein-coupled receptor (base sequence entered as “7tm 1” in P f am) ) Is found.

これらのことから配列番号 6に示す塩基配列がコードするタンパク質は Gタン パク質共役型受容体活性を有することが推測できる。  From these facts, it can be inferred that the protein encoded by the nucleotide sequence of SEQ ID NO: 6 has G protein-coupled receptor activity.

配列番号 Ίに記載の塩基配列がコードするァミノ酸配列は BLASTサーチに よれば G protein-coupled receptor (GPR103)と 6— & 1''11 6 : 0. 0、 43 2アミノ酸残基で 74%の相同性を有する。 また、 HMMP F AMによるタンパ ク質特徴検索によれば Gタンパク質共役型受容体の特徴を示す配列 (P f amに 「7tm— 1」 としてエントリーされる塩基配列) が見出される。  According to the BLAST search, the amino acid sequence encoded by the nucleotide sequence described in SEQ ID NO: と is 6% with G protein-coupled receptor (GPR103): 0.0, 432. With homology. According to a protein feature search using HMMP FAM, a sequence (a base sequence that is entered as “7tm-1” in P f am) is found that shows the characteristics of a G protein-coupled receptor.

これらのことから配列番号 7に示す塩基配列がコードするタンパク質は Gタン パク質共役型受容体活性を有すると推測できる。 また、 相同性の高いタンパク質 は neuropeptide FF2、 neuropeptide Y2、 および galanin GalRl受容体と B莫貫通 領域の相同性が高い。 .  From these facts, it can be inferred that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 7 has G protein-coupled receptor activity. In addition, proteins with high homology have high homology between the neuropeptide FF2, neuropeptide Y2, and galanin GalRl receptor and the B transmembrane region. .

配列番号 8に記載の塩基配列がコードするァミノ酸配列は B LASTサーチに よ ば G protein— coupled receptor (GPR103)と e— v a l u e力 5 X 10-157、 377ァミノ酸残基で 73 %で類似し、 HMM P F AMによるタンパク質特徴検 索を行ったところ Gタンパク質共役型受容体の特徴を示す配列(P f amに「7tm 1」 としてエントリーされる塩基配列) が見出される。 If the Amino acid sequence encoded by the nucleotide sequence set forth in SEQ ID NO: 8 good in B LAST search G protein- coupled receptor (GPR103) and e- value force 5 X 10 -157, similar 73% 377 Amino acid residues And HMM PF AM for protein characterization When the cable was searched, a sequence (a base sequence entered as “7tm1” in P f am) showing characteristics of the G protein-coupled receptor was found.

これらのこと力 ら配列番号 8に示す塩基配列がコードするタンパク質は Gタン パク質共役型受容体であると推測できる。 また、 相同性の高いタンパク質は neuropeptide FF2、 neuropeptide Y2、 および galanin GalRl受容体と膜貫通領域 の相同性が高い。  From these results, it can be inferred that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 8 is a G protein-coupled receptor. Highly homologous proteins have high homology between the neuropeptide FF2, neuropeptide Y2, and the galanin GalRl receptor and the transmembrane domain.

配列番号 9に記載の塩基配列がコードするアミノ酸配列は B LAS Tサーチに よれば G protein-coupled receptor (GPR103)と e— v a l u e力 5X 10一160Amino acid sequences are G protein-coupled receptor (GPR103) and e- value force 5X 10 one 160 according to the B LAS T search to nucleotide sequence encoding a SEQ ID NO: 9,

386アミノ酸残基で 73%の相同性を示す。 また、 HMMPFAMによるタン パク質特徴検索によれば Gタンパク質共役型受容体の特徴を示す配列 (P f am に 「7tm— 1」 としてエントリーされる塩基配列) が見出される。 It shows 73% homology at 386 amino acid residues. According to a protein feature search using HMMPFAM, a sequence (a base sequence that is entered as “7tm-1” in P f am) showing characteristics of a G protein-coupled receptor is found.

これらのこと力 ら配列番号 9に示す塩基配列がコードするタンパク質は Gタン パク質共役型受容体であると推測できる。 また、 相同性の高いタンパク質は neuropeptide FF2、 neuropeptide Y2、 およぴ galanin GalRl受容体と膜貫通領域 の相同性が高い。  From these facts, it can be inferred that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 9 is a G protein-coupled receptor. Highly homologous proteins have high homology between the neuropeptide FF2, the neuropeptide Y2, and the galanin GalRl receptor and the transmembrane domain.

配列番号 10に記載の塩基配列がコードするァミノ酸配列は BLASTサーチ によれば G protein - coupled receptor (GPR103)と e— v a l u e : 5X 10— 172According the Amino acid sequence encoded by the nucleotide sequence set forth in SEQ ID NO: 10 in the BLAST search G protein - coupled receptor and (GPR103) e- value: 5X 10- 172,

409アミノ酸残基で 73%の相同性を示し、 HMMPFAMによるタンパク質 特徴検索を行ったところ Gタンパク質共役型受容体の特徴を示す配列 (P f am に 「7tm— 1」 としてエントリーされる塩基配列) が見出される。 A protein characteristic search using HMMPFAM with 73% homology at 409 amino acid residues showed a characteristic of G protein-coupled receptor (base sequence entered as “7tm-1” in P f am) Is found.

これらのことから配列番号 10に示す塩基配列がコードするタンパク質は Gタ ンパク質共役型受容体であると推測できる。 また、 相同性の高いタンパク質は neuropeptide FF2、 neuropeptide Y2、 および galanin GalRl受容体と膜貫通領域 の相同性が高い。  From these facts, it can be assumed that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 10 is a G protein-coupled receptor. Highly homologous proteins have high homology between the neuropeptide FF2, neuropeptide Y2, and the galanin GalRl receptor and the transmembrane domain.

配列番号 1 1に記載の塩基配列がコードするァミノ酸配列は BLASTサーチ によれば、 a novel human g protein coupled receptorと e— v a 1 u e : 5 X 1 0 179、 348アミノ酸残基で 87%の相同性を示す。 また、 配列番号 1 1に示す 塩基配列がコードするアミノ酸配列について、 HMMPFAMによるタンパク質 特徴検索を行ったところ Gタンパク質共役型受容体の特徴を示す配列 (P f am に 「7tm— 1」 としてエントリーされる塩基配列) が見出される。 According the Amino acid sequence encoded by the nucleotide sequence set forth in SEQ ID NO: 1 1 to BLAST search, a novel human g protein coupled receptor and e- va 1 ue: 5 X 1 0 179, 348 87% with an amino acid residue Shows homology. Also shown in SEQ ID NO: 11 When the amino acid sequence encoded by the nucleotide sequence was searched for protein characteristics using HMMPFAM, a sequence (a nucleotide sequence entered as “7tm-1” in P f am) showing characteristics of a G protein-coupled receptor was found.

これらのことから配列番号 1 1に示す塩基配列がコードするタンパク質は Gタ ンパク質共役型受容体であると推測できる。  From these facts, it can be assumed that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 11 is a G protein-coupled receptor.

配列番号 1 2に記載の塩基配列がコードするァミノ酸配列は BLASTサーチ によれば gpcrxll as a functional receptor validated by angiopeptin and useful for screening of agonists and antagonists と e— v a l u e : 0. 0、 50 7アミノ酸残基で 72%の相同性を有し、 HMMPFAMによるタンパク質特徴 検索を行ったところ Gタンパク質共役型受容体の特徴を示す配列 (P f amに 「7tm— 1J としてエントリーされる塩基配列) が見出される。  According to the BLAST search, the amino acid sequence encoded by the nucleotide sequence of SEQ ID NO: 12 is gpcrxll as a functional receptor validated by angiopeptin and useful for screening of agonists and antagonists and e-value: 0.0, 507 amino acids remaining Characteristic search by HMMPFAM with a homology of 72% in the amino acid group revealed a sequence (base sequence that is entered as “7tm-1J” in P f am) indicating the characteristics of a G protein-coupled receptor .

これらのことから配列番号 1 2に示す塩基配列がコードするタンパク質は Gタ ンパク質共役型受容体であると推測できる。 また、 相同性の高いタンパク質はソ マトスタチンアナログの angiopeptinで活性化され、 ァテローム性動脈硬化を抑 制する。  From these facts, it can be assumed that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 12 is a G protein-coupled receptor. In addition, highly homologous proteins are activated by the somatostatin analog angiopeptin, which suppresses atherosclerosis.

配列番号 1 3に記載の塩基配列がコードするァミノ酸配列は BLASTサーチ によれば G protein-coupled receptor C5L2 GPR77と e— v a l ue : 5 X 10 一114、 331アミノ酸残基で 61%の相同性を示し、 また、 Oryctolagus cuniculus anaphylatoxin C5a receptor gene と、 e— v a l u e : 5 X 10_60、 329ァ ミノ酸残基で 44%の相同性を示す。 また、 配列番号 13に示す塩基配列がコー ドするアミノ酸配列について、 HMMPFAMによるタンパク質特徴検索を行つ たところ、 Gタンパク質共役型受容体の特徴を示す配列 (P f amに 「7tm— 1」 と してエントリーされる塩基配列) が見出される。 According to the BLAST search, the amino acid sequence encoded by the nucleotide sequence of SEQ ID NO: 13 and G protein-coupled receptor C5L2 GPR77 are e-value: 5 X 10 1 114 , 61% homology at 331 amino acid residues, 61% homology In addition, it shows 44% homology with Oryctolagus cuniculus anaphylatoxin C5a receptor gene with e-value: 5 X 10 _60 and 329 amino acid residues. In addition, HMMPFAM was used to perform a protein feature search on the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 13, and the sequence showing the characteristics of the G protein-coupled receptor (“7tm-1” in P f am) Base sequence to be entered).

これらのことから配列番号 1 3に示す塩基配列がコードするタンパク質は Gタ ンパク質共役型受容体であると推測できる。 また、 相同性の高いタンパク質は顆 粒球や未分化な榭状細胞で発現する C 5 L 2や走化性に係わる。  From these facts, it can be inferred that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 13 is a G protein-coupled receptor. Highly homologous proteins are involved in C5L2 expressed in granulocytes and undifferentiated dendritic cells and chemotaxis.

配列番号 1 4に記載の塩基配列は B L A S Tサーチによれば Bos taurus latrophilin 3 splice variant abbf と e— v a l u e : 3 X 10一51、 302ァ ミノ酸残基で 37%の相同性を示し、 HMMPFAMによるタンパク質特徴検索 を行ったところ Gタンパク質共役型受容体の特徴を示す配列 (P f amに 「7tm 2」 としてエントリーされる塩基配列) が見出される。 According to the BLAST search, the nucleotide sequence of SEQ ID NO: 14 is Bos taurus latrophilin 3 splice variant abbf and e- value: 3 X 10 shows one 51, 302 § amino acids 37% homology at residues indicates the character of the G protein-coupled receptor proteins, wherein at the retrieval was performed by HMMPFAM The sequence (base sequence entered as “7tm 2” in P f am) is found.

これらのことから配列番号 14に示す塩基配列がコードするタンパク質は Gタ ンパク質共役型受容体であると推測できる。 また、 相同性の高いタンパク質は外 来性毒 alpha- latrotoxinの受容体 Latrophilinのスプライシングバリアントであ る脳特異的 Latrophilinである。  From these facts, it can be inferred that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 14 is a G protein-coupled receptor. A highly homologous protein is brain-specific Latrophilin, a splicing variant of the exogenous toxin alpha-latrotoxin receptor Latrophilin.

配列番号 1 5に記載の塩基配列は B L A S Tサーチによれば serpentine receptor (Cyt28)と e— v a l u e : 5 X 10一56、 271ァミノ酸残基で 38 % のホ目同や生を不し、 また、 G protein-coupled receptor 56 と e_v a l u e : 8 X 10— 51、 281ァミノ酸残基で 36 %の相同性を、 さらに G- protein - coupled receptor (GPR56)が、 e— v a 1 u e : 1 X 10— 50で、 28 1ァミノ酸残基に亘 り 36%の相同性を示す。 また、 配列番号 1 5に示す塩基配列がコードするアミ ノ酸配列について、 HMMPFAMによるタンパク質特徴検索を行ったところ G タンパク質共役型受容体の特徴を示す配列 (P f a mに 「7tm— 2」 としてェント リーされる塩基配列)、及ぴ Latrophilin/CL- 1- likeGPSdomain(P f amに「GPS」 としてエントリーされる塩基配列) が見出される。 SEQ ID NO: 1 5 nucleotide sequence according to the serpentine receptor (Cyt28) according to BLAST search e- value: 5 X 10 one 56, 271 38% ho th same or live was not in Amino acid residue, also , G protein-coupled receptor 56 and e_v alue: 8 X 10- 51, 281 36% of homology with Amino acid residue, further G-protein - is coupled receptor (GPR56), e- va 1 ue: 1 X At 10-50, it shows 36% homology over 281 amino acid residues. When the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 15 was searched for protein characteristics using HMMPFAM, a sequence exhibiting the characteristics of a G protein-coupled receptor (“7tm-2” in P f am) Entry, and Latrophilin / CL-1-likeGPSdomain (base sequence entered as “GPS” in P fam).

これらのことから配列番号 1 5に示す塩基配列がコードするタンパク質は Gタ ンパク質共役型受容体であると推測できる。 また、 類似タンパク質をコードする c DNAは、 マウス造血幹細胞の mRNAからクローニングされた。  From these facts, it can be assumed that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 15 is a G protein-coupled receptor. CDNA encoding a similar protein was cloned from mouse hematopoietic stem cell mRNA.

配列番号 16に記載の塩基配列は B LAS Tサ '·ーチによれば、 G- protein coupled receptor GPR34 (GPR34)と e— v a l u e : 1 X 10— 19、 294ァミノ 酸残基で 27 %の相同性を、 さらに G protein-coupled receptor Gpr34 (Gpr34) geneと、 e— v a l u e : 5X 10— 18で、 294ァミノ酸残基に亘り 38 %の相 同性を有する。 また、 配列番号 16に示す塩基配列がコードするアミノ酸配列に ついて、 膜貫通へリックスを予測するプログラム tmHMM (S. Moller, M. D. R. Croning, R. Apwei丄 er. Evaluation of methods for the prediction of membrane spanning regions. Bioinformatics, 17(7) :646- 653, 2001) を用いて膜貫通部 位を予測したところ、 7つの膜貫通部位 (配列番号 32のァミノ酸番号で、 30 〜52、 65〜87、 107〜129、 141〜1 63、 197〜219、 23 9〜261、 276〜 298) に膜貫通部位が予測された。 According the nucleotide sequence of SEQ ID NO: 16 in the B LAS T Sa '- over switch, G- protein coupled receptor GPR34 (GPR34 ) and e- value: 1 X 10- 19, 294 Amino acid residues in the 27% homology, further G protein-coupled receptor Gpr34 (Gpr34 ) gene and, e- value: in 5X 10- 18, has a 38% homology over 294 Amino acid residues. A program tmHMM (S. Moller, MDR) that predicts a transmembrane helix for the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 16 Croning, R. Apweier 丄 er. Evaluation of methods for the prediction of membrane spanning regions.Bioinformatics, 17 (7): 646-653, 2001). With the amino acid number of SEQ ID NO: 32, transmembrane sites were predicted at 30 to 52, 65 to 87, 107 to 129, 141 to 163, 197 to 219, 239 to 261 and 276 to 298).

これらのことから配列番号 1 6に示す塩基配列がコードするタンパク質は Gタ ンパク質共役型受容体であると推測できる。 また、 類似のタンパク質をコードす る遺伝子は、 X染色体上にあり、 各種臓器で豊富に発現し、 PAF受容体と弱い相 同性を有する。  From these facts, it can be inferred that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 16 is a G protein-coupled receptor. The gene encoding a similar protein is on the X chromosome, is abundantly expressed in various organs, and has weak homology to the PAF receptor.

本発明の DN Aは、 翻訳配列中に塩基の欠失もしくは挿入を有した状態で取得 されることがあるが、 上記のような相同性検索やタンパク質特徴検索を行つた結 果、 該 DNAの塩基配列中の欠失もしくは揷入が推測された場合には、 当業者に おいて通常用いられているライブラリースクリーニングゃ PC Rクローニング等 の方法を用いて塩基の欠失もしくは挿入の無い完全長 c DNAを取得することが できる。 かくして得られる完全長 cDNAを用いて本発明のタンパク質を発現さ せ、 これを機能解析に用いることができる。  The DNA of the present invention may be obtained in a state in which a base sequence has been deleted or inserted in the translated sequence. As a result of the above-described homology search and protein feature search, the DNA If a deletion or insertion in the nucleotide sequence is suspected, use a method generally used by those skilled in the art, such as library screening, PCR cloning, etc., to delete the full length without deletion or insertion of bases. cDNA can be obtained. The protein of the present invention is expressed using the full-length cDNA thus obtained, and can be used for functional analysis.

かくして取得され、 塩基配列が決定され、 また機能が推定される本発明の DN Aは上記の配列番号 1〜 16に記載の塩基配列、 あるいはその翻訳領域として上 記に示した塩基配列を有するものだけでなく、 これらの塩基配列において、 1若 しくは数個 (ここで言う数個の数は特には限定されないが、 例えば 60個以下、 好ましく 30個以下、より好ましくは 20個以下、さらに好ましくは 10個以下、 特に好ましくは 5個以下を意味する。)の塩基が欠失、置換及ぴ /または付加され た塩基配列を有し、 かつ Gタンパク質共役型受容体活性を有するタンパク質をコ ードする DNA、 並びに、 これらとストリンジェントな条件下でハイブリダィズ し、 かつ Gタンパク質共役型受容体活性を有するタンパク質をコードする D N A 等も含まれる。 これら DNAには上記したとおり、 配列番号 1 7〜32に記載の ァミノ酸配列において 1若しくは数個のァミノ酸配列が欠失、 置換及ぴ Zまたは 付加されたァミノ酸配列からなり、 かつ Gタンパク質共役型受容体活性を有する タンパク質をコードするものが含まれる。 The DNA of the present invention thus obtained, whose nucleotide sequence is determined, and whose function is presumed, has the nucleotide sequence shown in SEQ ID NOS: 1 to 16 or the nucleotide sequence shown above as its translation region. In addition, in these base sequences, one or several (the number is not particularly limited, but is, for example, 60 or less, preferably 30 or less, more preferably 20 or less, and still more preferably Means 10 or less, particularly preferably 5 or less). A protein having a base sequence in which bases are deleted, substituted and / or added, and having G protein-coupled receptor activity. And a DNA that hybridizes with these under stringent conditions and encodes a protein having G protein-coupled receptor activity. As described above, in these DNAs, one or several amino acid sequences in the amino acid sequences described in SEQ ID NOs: 17 to 32 are deleted, substituted and / or substituted. Includes those encoding a protein consisting of an added amino acid sequence and having G protein-coupled receptor activity.

ここで、 ス トリンジェントな条件でハイブリダィズする D N Aとは、 配列番号 1〜 1 6に示される塩基配列と B L A S T解析で 8 0 %以上、 好ましくは 9 0 % 以上、 さらに好ましくは 9 5 %以上の相同性を有する塩基配列を含む D N A等が 挙げられる。 また、 ストリンジェントな条件下のハイブリダィゼーシヨンとは、 通常のハイブリダイゼーション緩衝液中で、 温度が 4 0〜 7 0 °C、 好ましくは 6 0〜6 5 °C等で反応を行い、 塩濃度が 1 5 mM〜3 0 O mM、 好ましくは 1 5 m M〜6 O mM等の洗浄液中で洗浄を行う方法に従って行うことができる。  Here, DNA that hybridizes under stringent conditions refers to a base sequence represented by SEQ ID NOS: 1 to 16 and a BLAST analysis of 80% or more, preferably 90% or more, and more preferably 95% or more. DNAs containing a base sequence having homology are exemplified. Hybridization under stringent conditions refers to a reaction in a normal hybridization buffer at a temperature of 40 to 70 ° C, preferably 60 to 65 ° C, and the like. Washing can be performed according to a method of washing in a washing solution having a salt concentration of 15 mM to 30 O mM, preferably 15 mM to 6 O mM.

さらに、 本発明の D NAは、 上述の方法により取得されたものでも、 また合成 されたものでもよい。 D NAの塩基配列の置換は、 例えばサイトダイレクテッド ミュータジエネシスキット (宝酒造社製) や、 クイックチェンジサイトダイレク テッドミュータジエネシスキット (ストラタジーン社製) 等の巿販キットで容易 に行うことができる。  Further, the DNA of the present invention may be obtained by the above-described method or may be synthesized. The DNA base sequence can be easily replaced with a sales kit such as a site-directed mutagenesis kit (Takara Shuzo) or a quick change site-directed mutagenesis kit (Stratagene). it can.

また、 配列番号 1〜1 6に記載の塩基配列は、 マウスを由来とするものである 1S 上記した c D NAライブラリーの作製法に従ってヒ トの c D NAライブラリ 一を作製し、 該ライブラリーに対して配列番号 1〜1 6の塩基配列を有する D N A断片をプローブとしたハイブリダィゼーシヨンを行うことにより、 配列番号 1 〜1 6に記載の塩基配列がコードするタンパク質のヒトのホモログタンパク質を コードする D NA (以下、 これを 「ヒ トホモログ D NA」 と称することがある) を取得することもできる。 本発明の配列番号 1〜 1 6に記載の塩基配列またはそ の相補配列を有する D N Aとストリンジェントな条件でハイブリダイズする D N Aには、 このようなヒトホモログ D NAも含まれる。  The nucleotide sequence of SEQ ID NOs: 1 to 16 is derived from a mouse.1S A human cDNA library was prepared according to the above-described method for preparing a cDNA library. By performing hybridization using a DNA fragment having the nucleotide sequence of SEQ ID NOS: 1 to 16 as a probe, a human homolog protein of the protein encoded by the nucleotide sequence of SEQ ID NOs: 1 to 16 (Hereinafter, this may be referred to as "human homolog DNA"). Such a DNA homologous to DNA that hybridizes under stringent conditions to DNA having the nucleotide sequence of SEQ ID NOS: 1 to 16 or a sequence complementary thereto according to the present invention is also included.

また、 インフォマティックスを利用して、 ヒ トホモログ D N Aが有する塩基酉 S 列を予測し、 該塩基配列を基に上記のヒ ト c D NAライブラリーなどからヒ トホ モログ D N Aを取得することもできる。  Further, it is also possible to predict the base sequence S of the human homolog DNA by using informatics, and obtain the human homolog DNA from the above human cDNA library or the like based on the nucleotide sequence. .

一般的に、 インフォマティックスを利用して目的とするタンパク質のホモログ タンパク質をコードする塩基配列を予測する方法としては、例えば、 (i) 目的と する c DNAの塩基配列をクエリーとして、 ヒ ト等の c DNAデータベース (ィ ンフォマティックスにより予測される c DNAデータベースを含む) に対し B L ASTなどを用いて相同性検索を行う方法や、 (i i )目的とする c DNAの塩基 配列をクエリーとして、 ヒト等の ESTデータベースに対し BLASTなどを用 いて相同性検索を行い、 ヒットした E STが有する配列を目的とする c DNAの 塩基配列を参照して連結する方法、 さらに (i i i) 目的とする c DNAの塩基 配列をクエリーとして、 ヒトなどのゲノムデータベースに対し B LAS Tなどを 用いて相同性検索を行い、 目的とする c DNAの遺伝子が存在するゲノム上の位 置を特定し、 そのゲノム領域に対して G e n s c a n (http:// genes, mit.edu/ GENSCA . html) や S i m4 (Genome Res. , 8: 976-74 (1998)) 等を用いて、 該ゲノム中の遺伝子部分の塩基配列を予測する方法等が挙げられる。 Generally, homologues of the target protein using informatics Methods for predicting the nucleotide sequence encoding a protein include, for example, (i) a cDNA database of a human or the like (a cDNA database predicted by informatics) using a query as a base sequence of a target cDNA; And (ii) homology search using BLAST etc. against EST databases such as humans using the base sequence of the target cDNA as a query. Then, a method is used in which the sequence of the hit EST is linked with reference to the base sequence of the target cDNA, and (iii) the base sequence of the target cDNA is used as a query against the genome database of humans or the like. A homology search is performed using LAST, etc., to identify the location on the genome where the cDNA gene of interest is located, and Genscan (http: // genes, mit.edu/ GENSC A. html), Sim4 (Genome Res., 8: 976-74 (1998)), and the like, and a method of predicting the nucleotide sequence of the gene portion in the genome.

マウス由来の cDNAのヒトホモログ DN Aの塩基配列を予測する場合、 上記 の方法のいずれも用いることができるが、 本発明の配列番号 1〜16に記載の塩 基配列を有する c DNAはいずれも新規であり、 上記 (i) の方法では、 ヒトホモ ログ DNAの塩基配列を取得できないと考えられるため、 (ii) あるいは (iii) に記載の方法などが好ましく用いられる。  When predicting the nucleotide sequence of human homolog DNA of mouse-derived cDNA, any of the above methods can be used, but any of the cDNAs having the nucleotide sequences of SEQ ID NOs: 1 to 16 of the present invention is novel. Since it is considered that the method (i) cannot obtain the nucleotide sequence of the human homologous DNA, the method described in (ii) or (iii) is preferably used.

かくして予測されたヒトホモログ DNAの塩基配列を基に、 上記のヒ ト cDN Aライブラリ一から、 配列番号 1〜 16に記載の塩基配列がコードするタンパク 質のヒ トのホモログタンパク質をコードする DNAを取得することもできる。 具 体的な取得方法としては、 例えば、 予測されたヒトホモログ DNAの 5, 端、 お よび 3' 端の塩基配列に相補的な塩基配列を有するプライマーを用いて、 上記ヒ ト cDNAライブラリーを鎳型として PCRを行う方法や、 予測されたヒトホモ ログ DNAの一部の配列をプローブとして、 上記ヒト c DNAライブラリーに対 してハイブリダィゼーションを行う方法等が挙げられる。  Based on the predicted nucleotide sequence of the human homolog DNA, a DNA encoding the human homolog protein of the protein encoded by the nucleotide sequence of SEQ ID NOS: 1 to 16 was obtained from the above human cDNA library. You can also. As a specific acquisition method, for example, the above-mentioned human cDNA library is prepared by using a primer having a nucleotide sequence complementary to the nucleotide sequence at the 5, 5, and 3 'end of the predicted human homolog DNA. Examples include a method of performing PCR as a template, and a method of performing hybridization on the human cDNA library using a partial sequence of the predicted human homolog DNA as a probe.

一般的に、 目的遺伝子が有する塩基配列とホモロジ一の高い塩基配列を有する 類似遺伝子を 「ホモログ」 と呼ぴ、 上記の方法においてもヒトホモログの取得を 目的としているが、 遺伝子の機能解析においては、 塩基配列が類似していること だけではなく、 ホモログとして取得された遺伝子が、 目的遺伝子のフアミリーメ ンバーであることを確認することが重要である。 2種類の生物間で 「ホモログ」 として取得された遺伝子は、 共通の祖先遺伝子から進化した同一の遺伝子であるIn general, a similar gene having a nucleotide sequence having a high homology to the nucleotide sequence of the target gene is called a “homolog”, and the above-mentioned method also requires the acquisition of a human homolog. Although it is intended, it is important in gene function analysis not only to confirm that the nucleotide sequences are similar, but also to confirm that the gene obtained as a homolog is a family member of the target gene. Genes acquired as “homologs” between two species are identical genes that evolved from a common ancestral gene

「オルソログ」 である可能性と、 また、 共通の祖先遺伝子からの重複によって生 じた異なる遺伝子である 「パラログ」 である可能性がある。 It may be an "ortholog" or a "paralog" which is a different gene created by duplication from a common ancestral gene.

つまり、 上記でホモロ として取得されたヒト由来の DNAは、 これを、 本発 明のタンパク質と同一の機能を有すると解するには、 また、 該ヒト由来の DNA がコードするタンパク質の機能を、 本発明のタンパク質のマウスにおける機能と して推定検証するには、 上記ヒトホモログが本発明のマウス遺伝子の近縁種のォ ルソログであることを確認することが好ましい。  In other words, in order for the human-derived DNA obtained as a homologue described above to have the same function as the protein of the present invention, the function of the protein encoded by the human-derived DNA must be In order to estimate and verify the function of the protein of the present invention in mice, it is preferable to confirm that the human homolog is an ortholog of a closely related species of the mouse gene of the present invention.

オルソログであることの確認方法は、 例えば、 以下の方法などが用いられる。  For example, the following method is used as a method for confirming the ortholog.

(1) まず、 本発明の c DN Aの塩基配列と、 取得されたヒトホモログ DNAの 塩基配列について相同性を解析する。 次に、 本発明の cDNAの塩基配列をタエ リーとして、 DDB J、 EMBL、 G e n B a n kなどの国際塩基配列データべ ースや、 特許データベースに含まれるヒト塩基配列について相同性検索を行い、 取得されたヒ トホモログ DNAとクエリーの塩基配列の一致度が、 データベース から得られた塩基配列とクエリ一の塩基配列の一致度より高いことを確認する。 さらに、 (2)取得されたヒトホモログ DNAの塩基配列と、対応する本発明の c DN Aの塩基配列について相同性を解析する。 次に、 取得されたヒトホモログ D NAの塩基配列をクエリーとして、 DDB J、 EMBL、 Ge nB a n kなどの 国際塩基配列データベースや、 特許データベースに含まれるマウス塩基配列につ いて相同性検索を行い、 本発明の c DN Aとクエリーの塩基配列の一致度が、 デ —タベースから得られた塩基配列とクエリーの塩基配列との一致度より高いこと を確認する。 上記 (1) および (2) を確認することにより、 取得されたヒトホ モログが、 本発明の c DNAに対応するヒトオルソログであると同定することが できる。 上記 (1) および (2) に記載した相同性の解析はァミノ酸配列の比 較を用いても良く、 また、 分子進化系統樹を描いて検討することもで きる。 また、 上記 ( 1 ) および ( 2 ) に記載した相同性解析による一 致度は、 クエリ一の全長にわたる一致度と して解析することが好まし レヽ o (1) First, the homology between the nucleotide sequence of the cDNA of the present invention and the nucleotide sequence of the obtained human homolog DNA is analyzed. Next, using the nucleotide sequence of the cDNA of the present invention as a query, homology search was performed on international nucleotide sequence databases such as DDB J, EMBL, Gen Bank, and human nucleotide sequences contained in patent databases. Confirm that the degree of matching between the obtained human homolog DNA and the base sequence of the query is higher than the degree of matching between the base sequence obtained from the database and the base sequence of the query. Further, (2) homology is analyzed for the nucleotide sequence of the obtained human homolog DNA and the corresponding nucleotide sequence of the cDNA of the present invention. Next, using the obtained nucleotide sequence of the human homolog DNA as a query, homology search was performed on the international nucleotide sequence database such as DDB J, EMBL, GenBank, and the mouse nucleotide sequence contained in the patent database. Confirm that the degree of matching between the cDNA of the invention and the base sequence of the query is higher than the degree of matching between the base sequence obtained from the database and the base sequence of the query. By confirming the above (1) and (2), the obtained human homolog can be identified as a human ortholog corresponding to the cDNA of the present invention. The homology analysis described in (1) and (2) above is based on the amino acid sequence ratio. Comparison may be used, and a molecular evolutionary phylogenetic tree may be drawn and examined. In addition, it is preferable that the degree of matching by the homology analysis described in (1) and (2) above be analyzed as the degree of matching over the entire length of the query.

かくして取得されたヒトホモログ DNA、 あるいはオルソログ DNAの塩基配 列を、 BLASTによる相同性検索や HMMPF AMによる蛋白質特徴検索等を 行うことにより、 該塩基配列がコードするタンパク質の機能を推定および確認す ることができる。  Estimating and confirming the function of the protein encoded by the base sequence of the human homolog DNA or ortholog DNA thus obtained by performing homology search by BLAST or protein characteristic search by HMMPFAM, etc. Can be.

本発明の配列番号 1〜 1 6に記載の塩基配列またはその相補配列を有する DN Aとストリンジェントな条件でハイブリダィズする DN Aには、 このようなヒト ホモログ、 あるいはオルソログタンパク質をコードする DNAも含まれる。 The DNA that hybridizes under stringent conditions with the DNA having the nucleotide sequence of SEQ ID NOS: 1 to 16 or the complementary sequence thereof includes DNA encoding such a human homolog or ortholog protein. It is.

(2) 新規 c DN Aがコードするタンパク質 (2) Novel protein encoded by cDNA

本発明の DNAがコードするタンパク質の翻訳領域は、 例えば、 該 DNAが有 する塩基配列について 3種類の読み枠によりアミノ酸に変換していき、 最も長い ポリぺプチドをコードする範囲を本発明の翻訳領域としてそのアミノ酸配列を決 定すること等ができる。 このようなアミノ酸配列として、 例えば、 配列番号 1 7 〜32に記載のもの等が挙げられる。 また、 本発明のタンパク質は、 上記のアミ ノ酸配列に限られるものではなく、 該アミノ酸配列において 1若しくは数個のァ ミノ酸が置換、 欠失、 及び または付カ卩されたアミノ酸配列からなり、 かつ Gタ ンパク質共役型受容体活性を有するものも含まれる。  The translation region of the protein encoded by the DNA of the present invention is, for example, the nucleotide sequence of the DNA, which is converted into amino acids by three reading frames, and the range encoding the longest polypeptide is translated by the present invention. The amino acid sequence of the region can be determined. Such amino acid sequences include, for example, those described in SEQ ID NOs: 17 to 32. Further, the protein of the present invention is not limited to the above-described amino acid sequence, but comprises an amino acid sequence in which one or several amino acids have been substituted, deleted, and / or added in the amino acid sequence. And those having G protein-coupled receptor activity.

本発明のタンパク質の取得方法としては、 上記 (1) に記載の本発明の DNA を適当な方法により転写/翻訳する方法が好ましく用いられる。 具体的には、 適 当な発現用ベクター若しくは適当なベクターに、 適当なプロモーターとともに挿 入した組換えベクターを作製し、 この組換えベクターで適当な宿主微生物を形質 転換したり、 適当な培養細胞に導入することにより発現させ、 これを精製するこ とにより取得することができる。  As a method for obtaining the protein of the present invention, the method of transcription / translation of the DNA of the present invention described in the above (1) by an appropriate method is preferably used. Specifically, a recombinant vector inserted into a suitable expression vector or a suitable vector together with a suitable promoter is prepared, and this recombinant vector is used to transform a suitable host microorganism, or to a suitable cultured cell. The expression can be obtained by introducing the DNA into a plasmid, and it can be obtained by purifying the protein.

かくして得られるタンパク質が遊離体で得られた場合には、 公知の方法あるい はそれに準じる方法によつて塩に変換することができ、 逆に塩で得られた場合に は遊離体、 又は他の塩に変換することができる。 この様な本発明のタンパク質の 塩も本発明のタンパク質に含まれる。 また、 上記形質転換体が産生するタンパク 質を、 精製前、 又は後に適当なタンパク質修飾酵素を作用させることにより、 任 意に修飾を加えたり、 ポリぺプチドを部分的に除去することにより修飾タンパク 質とすることができる。 これらの修飾タンパク質も Gタンパク質共役型受容体活 性を有するものであれば本発明の範囲に含まれる。 When the protein thus obtained is obtained in a free form, a known method or Can be converted into a salt by a method analogous thereto, and conversely, when obtained as a salt, it can be converted into a free form or another salt. Such salts of the protein of the present invention are also included in the protein of the present invention. In addition, the protein produced by the above transformant may be modified before or after purification with an appropriate protein modifying enzyme to optionally modify the protein or partially remove the polypeptide to modify the protein. Quality. These modified proteins are also included in the scope of the present invention as long as they have G protein-coupled receptor activity.

本発明のタンパク質の産生を行う際、 本発明の DN Aを含む組換えベクターの 作製に用いるベクターとしては、 形質転換体内で該 DNAが発現されるものであ れば特に制限はなく、プラスミドベクター、ファージべクターのいずれでもよい。 これらのうち通常は、 該 DNAが導入される宿主に適したプロモーター等の発現 制御領域 DN Aが既に挿入されている市販のタンパク質発現用ベクターを用いる。 このようなタンパク質発現用ベクターとして、 具体的には例えば、 宿主が大腸菌 の場合では、 pET3、 p ET 1 1 (ストラタジーン社製) GEX (アマシャ ムフアルマシアバイオテク社製) 等が挙げられ、 酵母の場合では p E S P— Iェ クスプレツシヨンベクター (ストラタジーン社製) 等が挙げられ、 さらに昆虫細 胞の場合では B a c PAK6 (クロンテック社製) 等が用いられる。 また宿主が 動物細胞の場合では、 ZAP Ex p r e s s (ストラタジーン社製)、 S VK 3 (アマシャムフアルマシアバイオテク社製) 等が挙げられる。  When producing the protein of the present invention, the vector used for the production of the recombinant vector containing the DNA of the present invention is not particularly limited as long as the DNA is expressed in the transformant. And phage vectors. Of these, usually, a commercially available protein expression vector into which an expression control region DNA such as a promoter suitable for a host into which the DNA is introduced has already been inserted is used. Specific examples of such a protein expression vector include pET3 and pET11 (manufactured by Stratagene) and GEX (manufactured by Amersham Armacia Biotech) when the host is Escherichia coli. In the case of (1), p ESP-I expression vector (manufactured by Stratagene) and the like can be mentioned. In the case of insect cells, Bac PAK6 (manufactured by Clontech) can be used. When the host is an animal cell, examples include ZAP Express (Stratagene), SVK3 (Amersham Pharmacia Biotech) and the like.

発現制御領域が揷入されていないベクターを用いる場合には、 発現制御領域と して少なくともプロモーターを挿入する必要がある。 ここでプロモーターとして は、 宿主微生物、 または培養細胞が保有するプロモーターを用いることができる 力、 これに限られるものではなく、 具体的には、 例えば、 宿主が大腸菌の場合に は T3、 T7、 t a c、 1 a cプロモーター等を用いることができ、 酵母の場合 には nmt 1プロモーター、 Ga 1 1プロモーター等を用いることができる。 ま た宿主が動物細胞の場合には SV 40プロモーター、 CMVプロモーター等が好 ましく用いられる。 また哺乳動物由来のプロモーターが機能可能な宿主を用いる場合には、 本発明 の遺伝子に固有のプロモーターを用いることもできる。 これらのベクターへの本 発明の DNAの挿入は、 該 DNAまたはこれを含む DNA断片をベクター中のプ 口モーターの下流に該遺伝子 DNAがコードするタンパク質のアミノ酸配列を連 結して行えばよい。 When a vector without an expression control region is used, it is necessary to insert at least a promoter as the expression control region. The promoter used herein is not limited to a promoter that can be used by a host microorganism or a promoter possessed by a cultured cell. Specifically, for example, when the host is Escherichia coli, T3, T7, tac , 1 ac promoter and the like, and in the case of yeast, nmt1 promoter, Ga11 promoter and the like can be used. When the host is an animal cell, SV40 promoter, CMV promoter and the like are preferably used. When a host capable of functioning as a mammalian promoter is used, a promoter specific to the gene of the present invention can also be used. Insertion of the DNA of the present invention into these vectors may be performed by linking the DNA or a DNA fragment containing the DNA to the amino acid sequence of the protein encoded by the gene DNA downstream of the open motor in the vector.

このようにして作製した組換えベクターは、 それ自体既知の方法により後述す る宿主を形質転換して、 DN A導入体を作製することができる。 宿主への該べク ターの導入方法として、 具体的には、 ヒートショ ック法 (J. Mol.Biol. ,53, 154, (1970))、 リン酸カルシウム法 (Science, 221, 551, (1983))、 DEAEデキストラ ン法 (Science, 215, 166, (1982))、インビトロパッケージング法 (Proc. Natl. Acad. Sci.USA, 72,581, (1975))、 ウィルスベクター法 (Cell, 37, 1053, (1984) )、 および 電気パルス法 (Chu. et al. , Nuc. Acids Res. , 15, 1331(1987)) 等が挙げられる。  The recombinant vector thus prepared can be transformed into a host described below by a method known per se to prepare a DNA-introduced body. As a method for introducing the vector into a host, specifically, a heat shock method (J. Mol. Biol., 53, 154, (1970)) and a calcium phosphate method (Science, 221, 551, (1983)) ), DEAE dextran method (Science, 215, 166, (1982)), in vitro packaging method (Proc. Natl. Acad. Sci. USA, 72, 581, (1975)), virus vector method (Cell, 37, 1053, (1984)), and the electric pulse method (Chu. Et al., Nuc. Acids Res., 15, 1331 (1987)).

DNA導入体を作製するための宿主としては、 本発明の DNAが体内で発現す るものであれば特に限定されないが、例えば大腸菌、酵母、バキュロウィルス(節 足動物多角体ウィルス) 一昆虫細胞、 あるいは動物細胞等が挙げられる。 具体的 には、 大腸菌では BL 21、 XL- 2 B 1 u e (ストラタジーン社製) 等、 酵母 では SP— Q0 1 (ス トラタジーン社製) 等、 バキュロウィルスでは A c NPV The host for preparing the DNA transfectant is not particularly limited as long as the DNA of the present invention is expressed in the body. Examples of the host include Escherichia coli, yeast, baculovirus (arthropod polyhedrovirus), insect cells, Alternatively, animal cells and the like can be mentioned. Specifically, BL21 and XL-2B1ue (manufactured by Stratagene) for E. coli, SP—Q01 (manufactured by Stratagene) for yeast, and AcNPV for baculovirus.

(J. Biol. Chem. , 263, 7406, (1988))とその宿主である S f — 9 (J. Biol. C em. , 263, 7406, (1988)) 等が挙げられる。 また動物細胞としてはマウス繊維芽細胞 C 12 7 (J. Viol., 26, 291, (1978)) やチャイニーズハムスター卵巣細胞 CHO細胞(J. Biol. Chem., 263, 7406, (1988)) and its host, Sf-9 (J. Biol. Cem., 263, 7406, (1988)). Examples of animal cells include mouse fibroblast C 127 (J. Viol., 26, 291, (1978)) and Chinese hamster ovary cell CHO cells

(Proc. Natl. Acad. Sci. USA, 77, 4216, (1980)) 等が挙げられるが、 発現量ゃス クリ一二ングの簡便さから好ましくはアフリカミ ドリザル腎臓由来 c o S- 7(Proc. Natl. Acad. Sci. USA, 77, 4216, (1980)), and the like. Preferably, expression level か ら cos-7 derived from African green monkey kidney is preferred because of simple screening.

(ATCC CRL1651:アメリカン タイプ 力ルチヤ一 コレクション保存細胞) 用いられる。 (ATCC CRL1651: American type Karyuchi collection collection preserved cells) Used.

上記したようなタンパク質発現用ベクターを用いる発現方法の他に、 プロモー ターを連結した本発明の DN A断片を宿主微生物の染色体中に直接挿入する相同 組換え技術 (A. A. Vertes et al. , Biosci. Biotechnol. Biochem. , 57, 2036, (1993) )、 あるいはトランスポゾンや挿入配列 (A. A. Vertes et al. , Molecular Microbiol. , 11, 739, (1994) ) 等を用いて D N A導入体を作製することもできる。 得られた培養物は、 細胞あるいは菌体を遠心分離等の方法により収集し、 これ を適当な緩衝液に懸濁し、 超音波、 リゾチーム、 および/または凍結融解等のそ れ自体既知の適当な方法により破壊した後、 遠心分離や濾過等によりタンパク質 粗精製液を得、 さらに適当な精製方法を組み合わせることにより精製することが できる。 かくして、 本発明のタンパク質が取得される。 上記したタンパク質発現 組換えベクターを用いる発現方法の他に、 上記 (1 ) で取得された本発明の D N Aを無細胞転写翻訳系に供することによりタンパク質発現を誘導し、 本発明のタ ンパク質を取得することができる。 本発明で用いられる無細胞転写翻訳系とは、 D N Aから m R NAへの転写、 および m R N Aからタンパク質への翻訳に必要な 全ての要素を含む系であり、 そこに D NAを加えることによってその D NAがコ ードしているタンパク質が合成されるようなあらゆる系を指す。 無細胞転写翻訳 系の具体例としては、 真核細胞、 およびバクテリア細胞、 又はそれらの一部から の抽出液に基づいて調製された転写翻訳系が挙げられ、 特に好ましい具体例とし ては、 ゥサギ網状赤血球、小麦胚芽、 大腸菌からの抽出液 (大腸菌 S 3 0抽出液) に基づいて調製された転写翻訳系が挙げられる。 In addition to the expression method using a protein expression vector as described above, a homologous recombination technique for directly inserting a promoter-ligated DNA fragment of the present invention into the chromosome of a host microorganism (AA Vertes et al., Biosci. Biotechnol. Biochem., 57, 2036, (1993)), Alternatively, a DNA transfectant can be prepared using a transposon, an insertion sequence (AA Vertes et al., Molecular Microbiol., 11, 739, (1994)) and the like. In the obtained culture, cells or cells are collected by a method such as centrifugation, suspended in a suitable buffer, and then collected by a suitable method known per se such as ultrasonication, lysozyme, and / or freeze-thawing. After disruption by the method, a crude protein solution is obtained by centrifugation, filtration, or the like, and further purified by a combination of appropriate purification methods. Thus, the protein of the present invention is obtained. In addition to the above-described expression method using a protein expression recombinant vector, protein expression is induced by subjecting the DNA of the present invention obtained in the above (1) to a cell-free transcription / translation system, and the protein of the present invention Can be obtained. The cell-free transcription / translation system used in the present invention is a system containing all elements necessary for transcription from DNA to mRNA and translation of mRNA to protein, and by adding DNA thereto. It refers to any system by which the protein encoded by the DNA is synthesized. Specific examples of the cell-free transcription / translation system include a transcription / translation system prepared based on an eukaryotic cell, a bacterial cell, or an extract from a part thereof, and a particularly preferred example is Egret A transcription / translation system prepared based on extracts from reticulocytes, wheat germ, and Escherichia coli (Escherichia coli S30 extract) may be mentioned.

得られた無細胞転写翻訳系の転写翻訳産物からの、本発明のタンパク質の分離、 および精製は、 それ自体既知の通常用いられる方法で行うことができる。 具体的 には、 例えばェピトープぺプチド、 ポリヒスチジンぺプチド、 グルタチオン一 S 一トランスフェラーゼ(G S T)、マルトース結合タンパク質等をコードする D N A領域を、 前記した転写翻訳されるべき D NAに導入し、 前記の通り発現させ、 該タンパク質と親和性を有する物質とのァフィ二ティーを利用して精製すること ができる。  Separation and purification of the protein of the present invention from the obtained transcription-translation product of the cell-free transcription / translation system can be carried out by a commonly used method known per se. Specifically, for example, a DNA region encoding an epitope peptide, a polyhistidine peptide, a glutathione-S-transferase (GST), a maltose binding protein, and the like is introduced into the DNA to be transcribed and translated, and And purified using the affinity of the protein with a substance having affinity.

目的とするタンパク質の発現は、 S D S—ポリアクリルアミ ドゲル電気泳動等 で分離し、 クマシ一ブリリアントブルー (シグマ社製) 等で染色するか、 または 後述する本発明のタンパク質に特異的に結合する抗体により検出する方法等によ つて確認できる。 また一般的に、 発現されたタンパク質は生体内に存在するタン パク質分解酵素により切断されること (プロセッシング) が知られている。 本発 明のタンパク質も当然のことながら切断されたアミノ酸配列の部分断片であって も、 Gタンパク質共役型受容体活性を有するものであれば、 本発明のタンパク質 に含まれる。 The expression of the target protein is separated by SDS-polyacrylamide gel electrophoresis and stained with Coomassie brilliant blue (manufactured by Sigma), or an antibody that specifically binds to the protein of the present invention described below. Detection method, etc. Can be confirmed. It is generally known that the expressed protein is cleaved (processed) by a proteolytic enzyme present in the living body. The protein of the present invention is naturally included in the protein of the present invention, even if it is a fragment of the amino acid sequence that has been cleaved, as long as it has G protein-coupled receptor activity.

力べして得られたタンパク質は、 他のタンパク質や核酸との相互作用等を解析 することにより、 生体内における多面的な機能を知ることができる。 上記相互作 用の解析法としては、それ自体既知の常法を用いることができるが、具体的には、 例えば、 酵母ツーハイブリッド法、 蛍光偏光解消法、 表面プラズモン法、 ファー ジディスプレイ法、 リポソ一マルディスプレイ法等が挙げられる。  By analyzing the interaction of other proteins and nucleic acids, etc., the proteins obtained by the effort can be used to find out the multifaceted functions in vivo. As a method for analyzing the interaction, a conventional method known per se can be used. Specifically, for example, yeast two-hybrid method, fluorescence depolarization method, surface plasmon method, phage display method, liposome method One example is the multiple display method.

( 3 ) オリゴヌクレオチドの調製  (3) Preparation of oligonucleotide

上記 (1 ) に記載の方法で取得した本発明の D N Aまたはその断片を用いて、 D N A合成機などを用いる常法により、 本発明の D N Aの一部の配列を有するァ ンチセンス ·オリゴヌクレオチド、 センス .オリゴヌクレオチド等のォリゴヌク レオチドを調製することができる。  Using the DNA of the present invention or a fragment thereof obtained by the method described in (1) above, antisense oligonucleotides having a partial sequence of the DNA of the present invention, Oligonucleotides such as oligonucleotides can be prepared.

該オリゴヌクレオチドとしては、 上記 D N Aの有する塩基配列中の連続した 5 〜1 0 0塩基と同じ配列を有する D NAまたは該 D N Aと相補的な配列を有する D NAを挙げることができる。 具体例としては、 配列番号 1〜1 6のいずれかで 表される塩基配列中の連続した 5〜1 0 0塩基と同じ配列を有する D NAまたは 該 D NAと相補的な配列を有する D NAを挙げることができる。 センスプライマ 一おょぴアンチセンスプライマーとして用いる場合には、両者の融解温度(T m) および塩基数が極端に変わることのない上記のオリゴヌクレオチドが好ましい。 また、 配列の長さは、 一般的には 5〜: L 0 0塩基であり、 好ましくは 1 0〜 6 0 塩基であり、 より好ましくは 1 5〜5 0塩基である。  Examples of the oligonucleotide include a DNA having the same sequence as the continuous 5 to 100 bases in the base sequence of the DNA or a DNA having a sequence complementary to the DNA. As a specific example, DNA having the same sequence as 5 to 100 consecutive nucleotides in the base sequence represented by any of SEQ ID NOS: 1 to 16 or DNA having a sequence complementary to the DNA Can be mentioned. When used as a sense primer or antisense primer, the above-mentioned oligonucleotides whose melting temperature (Tm) and number of bases do not extremely change are preferable. The length of the sequence is generally 5 to: L00 bases, preferably 10 to 60 bases, and more preferably 15 to 50 bases.

また、 これらオリゴヌクレオチドの誘導体も本発明のオリゴヌクレオチドとし て利用することができる。 該オリゴヌクレオチド誘導体としては、 オリゴヌタレ ォチド中のリン酸ジエステル結合がホスホロチォエート結合に変換されたオリゴ ヌクレオチド誘導体、 オリゴヌクレオチド中のリン酸ジエステル結合が N 3, 一 P 5 ' ホスフォアミデート結合に変換されたオリゴヌクレオチド誘導体、 オリゴ ヌクレオチド中のリポースとリン酸ジエステル結合がぺプチド核酸結合に変換さ れたォリゴヌクレオチド誘導体、 オリゴヌクレオチド中のゥラシルが C— 5プロ ピニルゥラシルで置換されたォリゴヌクレオチド誘導体、 ォリゴヌクレオチド中 のゥラシルが C一 5チアゾールゥラシルで置換されたォリゴヌクレオチド誘導体、 オリゴヌクレオチド中のシトシンが C一 5プロピニルシトシンで置換されたオリ ゴヌクレオチド誘導体、 オリゴヌクレオチド中のシトシンがフエノキサジン修飾 シトシン (phenoxazine - modified cytosine) で置換さ; たオリゴヌクレオチド誘 導体、 オリゴヌクレオチド中のリポースが 2 ' —O—プロピルリボースで置換さ れたォリゴヌクレオチド誘導体、あるいはォリゴヌクレオチド中のリボースが 2, —メ トキシエトキシリボースで置換されたオリゴヌクレオチド誘導体等を挙げる ことができる。 In addition, derivatives of these oligonucleotides can also be used as the oligonucleotide of the present invention. Examples of the oligonucleotide derivative include an oligonucleotide in which a phosphodiester bond in an oligonucleotide is converted to a phosphorothioate bond. Nucleotide derivative, an oligonucleotide derivative in which the phosphodiester bond in the oligonucleotide is converted to an N3,1 P5'phosphoamidate bond, and a report and a phosphodiester bond in the oligonucleotide that are converted to a peptide nucleic acid bond Oligonucleotide derivatives, Oligonucleotide derivatives in which peracyl in oligonucleotides are substituted with C-5 propynylperacyl, Oligonucleotide derivatives in which peracyl in oligonucleotides are substituted with C-15 thiazole peracyl, Oligonucleotides Oligonucleotide derivatives in which cytosine in nucleotides has been substituted with C-15-propynylcytosine; cytosine in oligonucleotides has been substituted with phenoxazine-modified cytosine; oligonucleotide derivatives, oligonucleotides Oligonucleotide derivatives in which the report in the oligonucleotide is substituted with 2'-O-propylribose, or oligonucleotide derivatives in which the ribose in the oligonucleotide is substituted with 2, -methoxyethoxyribose, etc. .

また、 本発明のオリゴヌクレオチドは、 これを 2本鎖 R NAとして調製し、 被 導入体へ導入し、標的遺伝子の発現を阻害する R NAインターフェアレンス法(以 下、 これを 「RNAi法」 と称することがある) に用いることができる。 R NAイン ターフェアレンス法については、 例えば、 (Elbashir, S. , et al. , Nature, 411, 494-498 (2001) ) に記載の方法等を用いることができる。 また、上記 2本鎖 R N A は必ずしも全てが R N Aである必要はなく、 例えば、 WO O 2 / 1 0 3 7 4号公 報に記載のもの等も用いることができる。  In addition, the oligonucleotide of the present invention is prepared as a double-stranded RNA, introduced into a transfectant, and inhibits the expression of a target gene by an RNA interference method (hereinafter referred to as “RNAi method”). ) May be used. As the RNA interference method, for example, the method described in (Elbashir, S., et al., Nature, 411, 494-498 (2001)) can be used. Also, not all of the above-mentioned double-stranded RNAs need to be RNAs. For example, those described in WO 02/13734 can be used.

ここで、 標的遺伝子としては、 本発明の DNAであれば、 如何なるものであって もよレ、。 これら D NAの少なくとも一部の塩基配列と実質的に同一な配列からな る 2本鎖 R NA (以下、 これを 「2本鎖ポリヌクレオチド」 と称することがある) とは、標的遺伝子の塩基配列のうち、 いずれの部分でもよい 1 5 bp以上の配列と 実質的に同一な配列からなるものである。 ここで、 実質的に同一とは、 標的遺伝 子の配列と 8 0 %以上の相同性を有することを意味する。 ヌクレオチドの鎮長は 1 5 bpから標的遺伝子のオープンリーディングフレーム (O R F ) の全長までの 如何なる長さでもよいが、 1 5〜5 0 O bp程度のものが好ましく用いられる。 た だし、哺乳類動物由来の細胞おいては、 3 O bp以上の長い 2本鎖 R N Aに反応し て活性化するシグナル伝達系の存在が知られている。 これはィンターフェ口ン反 応と呼ばれており (Mareus, P. I. , et al. , Interferon, 5, 115-180 (1983) )、 該 2本鎖 R N Aが細胞内に侵入すると、 P K R (dsRNA-responsive protein kinase: Bass, B. L. , Nature, 411, 428-429 (2001) ) を介して多くの遺伝子の翻 訳開始が非特異的に阻害され、それと同時に 2,、 5, oligoadenylate synthetaseHere, the target gene may be any as long as it is the DNA of the present invention. A double-stranded RNA consisting of a sequence substantially identical to at least a part of the base sequence of these DNAs (hereinafter sometimes referred to as “double-stranded polynucleotide”) is referred to as a base sequence of a target gene. It is composed of a sequence substantially identical to a sequence of 15 bp or more, which may be any part of the sequence. Here, “substantially identical” means that the sequence has 80% or more homology with the sequence of the target gene. The length of nucleotides ranges from 15 bp to the full length of the open reading frame (ORF) of the target gene. Any length may be used, but a length of about 15 to 50 Obp is preferably used. However, it is known that mammalian-derived cells have a signal transduction system that activates in response to long double-stranded RNA of 3 O bp or more. This is called an interfering reaction (Mareus, PI, et al., Interferon, 5, 115-180 (1983)), and when the double-stranded RNA enters the cell, PKR (dsRNA-responsive) protein kinase: The translation initiation of many genes is non-specifically inhibited via Bass, BL, Nature, 411, 428-429 (2001)), and at the same time, 2, 5, oligoadenylate synthetase

(Bass, B丄, Nature, 411, 428- 429 (2001) )を介して RNaseLの活性化が起こり、 細胞内の; R N Aの非特異的な分解が惹起される。 これらの非特異的な反応のため に、 標的遺伝子の特異的反応が隠蔽されてしまう。 従って哺乳類動物、 または該 動物由来の細胞、 あるいは組織を被導入体として用いる場合には 1 5〜3 O bp, 好ましくは 1 9〜 2 2 bp、 さらに好ましくは 2 1 bpの 2本鎖ポリヌクレオチドを 用いることが好ましい。 2本鎖ポリヌクレオチドはその全体が 2本鎮である必要 はなく、 5 ' または 3 ' 末端が一部突出したものも含むが、 3, 末端が 2塩基突 出したものを用いることが好ましい。 (Bass, B 丄, Nature, 411, 428-429 (2001)), activation of RNaseL occurs, and nonspecific degradation of intracellular; RNA is caused. These non-specific reactions mask the specific response of the target gene. Therefore, when a mammal, or a cell or tissue derived from the animal is used as a transfectant, a double-stranded polynucleotide of 15 to 30 bp, preferably 19 to 22 bp, more preferably 21 bp It is preferable to use The double-stranded polynucleotide does not need to be entirely double-stranded, and includes those with a partially protruding 5 'or 3' end, but preferably those having two or three bases protruding.

2本鎖ポリヌクレオチドは相補性を有する 2本鎖のポリヌクレオチドを意味す るが、 自己相補性を有する 1本鎖ポリヌクレオチドが自己アニーリングしたもの でもよい。 自己相補性を有する 1本鎖ポリヌクレオチドとしては、 例えば、 逆方 向反復配列を有するもの等が挙げられる。  The double-stranded polynucleotide means a double-stranded polynucleotide having complementarity, but may be a self-annealed single-stranded polynucleotide having self-complementarity. Examples of the single-stranded polynucleotide having self-complementarity include those having an inverted repeat sequence.

2本鎖ポリヌクレオチドの調製方法としては、 特に制限はないが、 それ自体既 知の化学合成方法を用いることが好ましレ、。 化学合成は、 相補性を有する 1本鎖 ポリヌクレオチドを別個に合成し、 これを適当な方法で会合させることにより 2 本鎖とすることができる。 会合の方法として具体的には、 例えば、 合成した 1本 鎖ポリヌクレオチドを混合し、 2本鎖が解離する温度にまで加熱し、 その後徐々 に冷却する方法等が挙げられる。 会合した 2本鎖ポリヌクレオチドは、 ァガロー スゲル等を用いて確認し、 残存する 1本鎖ポリヌクレオチドを適当な酵素により 分解する等して除去する。 このようにして調製した 2本鎖ポリヌクレオチドを導入する被導入体としては、 標的遺伝子がその細胞内で R Aに転写、 またはタンパク質に翻訳を受け得るもの であれば如何なるものであってもよいが、 具体的には、 植物、 動物、 原生動物、 ウィルス、 バクテリア、 または真菌種に属するものが挙げられる。 植物は単子葉 植物、 双子葉植物または裸子植物であってよく、 動物は、 脊椎動物または無脊椎 動物であってよレ、。好ましい微生物は、農業または工業で使用されるものであり、 そして植物または動物に対して病原性のものである。 真菌には、 力ビ及ぴ酵母形 態両方での生物体が含まれる。 脊椎動物の例には、 魚類、 ゥシ、 ャギ、 ブタ、 ヒ ッジ、 ハムスター、 マウス、 ラット及びヒトを含む哺乳動物が含まれ、 無脊椎動 物には、線虫類及び他の虫類、 キイ口ショウジョゥバエ (Drosophila)、及び他の 昆虫が含まれる。 好ましくは、 細胞は脊椎動物細胞である。 The method for preparing the double-stranded polynucleotide is not particularly limited, but it is preferable to use a chemical synthesis method known per se. In chemical synthesis, a single-stranded polynucleotide having complementarity can be separately synthesized, and can be converted into a double-stranded strand by associating them by an appropriate method. Specific examples of the method of association include a method in which the synthesized single-stranded polynucleotide is mixed, heated to a temperature at which the double-strand is dissociated, and then gradually cooled. The associated double-stranded polynucleotide is confirmed using an agarose gel or the like, and the remaining single-stranded polynucleotide is removed by, for example, decomposing with a suitable enzyme. The transfectant into which the double-stranded polynucleotide prepared in this way is introduced may be any as long as the target gene can be transcribed into RA or translated into protein in the cell. Specific examples include those belonging to plant, animal, protozoan, virus, bacterial, or fungal species. The plant can be a monocotyledonous, dicotyledonous or gymnosperm, and the animal can be a vertebrate or invertebrate. Preferred microorganisms are those used in agriculture or industry, and are pathogenic to plants or animals. Fungi include organisms in both power and yeast forms. Examples of vertebrates include mammals, including fish, sea lions, goats, pigs, sheep, hamsters, mice, rats, and humans, and invertebrates include nematodes and other insects. Includes creatures, Drosophila, and other insects. Preferably, the cells are vertebrate cells.

被導入体は、 細胞、 組織、 あるいは個体を意味する。 ここで細胞とは、 生殖系 列または体性、 分化全能、 または多分化能、 分割または非分割、 実質組織または 上皮、 不滅ィ匕したものまたは形質転換したもの等からであってよい。 細胞は、 配 偶子または胚であってよく、 胚の場合、 単一細胞胚または構成性細胞、 または多 重細胞胚からの細胞であり、 胎児組織を含む。 さらには、. 幹細胞のような未分化 細胞、 または胎児組織を含む器官または組織の細胞からのような分化細胞、 また は生物内に存在する任意の他の細胞であってよい。 分化している細胞型には、 脂 肪細胞、 繊維芽細胞、 筋細胞、 心筋細胞、 内皮細胞、 神経細胞、 グリア、 血液細 胞、 巨核球、 リンパ球、 マクロファージ、 好中球、 好酸球、 好塩基球、 マスト細 胞、 白血球、 顆粒球、 ケラチン生成細胞、 軟骨細胞、 骨芽細胞、 破骨細胞、 肝細 胞及び内分泌腺または外分泌腺の細胞が含まれる。  The transductant means a cell, tissue, or individual. Here, the cells may be from germ line or somatic, totipotent or pluripotent, divided or undivided, parenchymal or epithelial, immortalized or transformed, and the like. The cells may be gametes or embryos, in the case of embryos, single cell embryos or constitutive cells, or cells from multicellular embryos, including fetal tissue. Further, it may be an undifferentiated cell such as a stem cell, or a differentiated cell such as a cell of an organ or tissue including fetal tissue, or any other cell present in an organism. Differentiating cell types include fat cells, fibroblasts, muscle cells, cardiomyocytes, endothelial cells, neurons, glial, blood cells, megakaryocytes, lymphocytes, macrophages, neutrophils, and eosinophils Basophils, mast cells, leukocytes, granulocytes, keratinocytes, chondrocytes, osteoblasts, osteoclasts, liver cells and cells of the endocrine or exocrine glands.

被導入体への 2本鎖ポリヌクレオチドの導入法としては、 被導入体が細胞、 あ るいは組織の場合は、カルシウムフォスフエ一ト法、エレクトロポレーション法、 リポフエクシヨン法、 ウィルス感染、 2本鎖ポリヌクレオチド溶液への浸漬、 あ るいは形質転換法等が用いられる。 また、 胚に導入する方法としては、 マイクロ インジェクション、 エレクト口ポレーシヨン法、 あるいはウイスル感染等が挙げ られる。 被導入体が植物の場合には、 植物体の体腔または間質細胞等への注入ま たは灌流、 あるいは噴霧による方法が用いられる。 また、 動物個体の場合には、 経口、局所、非経口 (皮下、筋肉内及び静脈内投与を含む)、経膣、経直腸、経鼻、 経眼、 腹膜内投与等によって全身的に導入する方法、 あるいはエレクト口ポレー シヨン法やウィルス感染等が用いられる。 経口導入のための方法には、 2本鎖ポ リヌクレオチドを生物の食物と直接混合することができる。 さらに、 個体に導入 する場合には、 例えば埋め込み長期放出製剤等として投与することや、 2本鎖ポ リヌクレオチドを導入した導入体を摂取させることにより行うこともできる。 導入する 2本鎖ポリヌクレオチドの量は、 導入体や、 標的遺伝子によって適宜 選択することができるが、 細胞あたり少なくとも 1コピー導入されるに充分量を 導入することが好ましい。 具体的には、 例えば、 被導入体がヒ ト培養細胞で、 力 ルシゥムフォスフェート法により 2本鎖ポリヌクレオチドを導入する場合、 0 . 1〜1 0 0 O nMが好ましレヽ。 As a method for introducing the double-stranded polynucleotide into the recipient, when the recipient is a cell or tissue, calcium phosphate method, electroporation method, lipofection method, virus infection, two For example, immersion in a strand polynucleotide solution or a transformation method is used. Examples of the method for introduction into an embryo include microinjection, electoral poration, and virus infection. Can be When the recipient is a plant, a method of injecting or perfusing the plant into the body cavity or stromal cells, or spraying is used. In the case of an individual animal, it is introduced systemically by oral, topical, parenteral (including subcutaneous, intramuscular, and intravenous administration), vaginal, rectal, nasal, ophthalmic, and intraperitoneal administration Method, electoral poration method, virus infection, etc. are used. For methods for oral introduction, the double-stranded polynucleotide can be mixed directly with the food of the organism. Furthermore, when introduced into an individual, it can be administered, for example, by administration as an implanted long-term release preparation or the like, or by ingesting an introduced body into which a double-stranded polynucleotide has been introduced. The amount of the double-stranded polynucleotide to be introduced can be appropriately selected depending on the introduced substance and the target gene, but it is preferable to introduce an amount sufficient to introduce at least one copy per cell. Specifically, for example, when the transfectant is a cultured human cell and the double-stranded polynucleotide is introduced by the calcium phosphate method, 0.1 to 100 OnM is preferred.

R NAインターフェアレンスによる本発明の遺伝子の導入体内での発現抑制に より、 本発明の遺伝子がコードするタンパク質の機能の確認、 あるいは新たな機 能の解析等を行うことができる。  By suppressing the expression of the gene of the present invention in the transfectant by RNA interference, the function of the protein encoded by the gene of the present invention can be confirmed, or a new function can be analyzed.

( 4 ) 本発明のタンパク質に特異的に結合する抗体  (4) an antibody that specifically binds to the protein of the present invention

本発明のタンパク質と特異的に結合する抗体の調製方法としては、 通常用いら れる公知の方法を用いることができ、 抗原として用いられるポリぺプチドについ ても、 公知の方法に従って抗原性が高くェピトープ (抗原決定基) として適した 配列を選択して用いることができる。 ェピトープの選択方法としては、 例えば Epitope Adviser (富士通九州システムエンジニアリング社製) 等の市販のソフト ウェアを用いることができる。  As a method for preparing an antibody that specifically binds to the protein of the present invention, a commonly used known method can be used. Even polypeptides used as antigens have high antigenicity according to known methods, A sequence suitable as (antigenic determinant) can be selected and used. As a method of selecting the epitope, for example, commercially available software such as Epitope Adviser (manufactured by Fujitsu Kyushu System Engineering Co., Ltd.) can be used.

上記の抗原として用いるポリペプチドは、 公知の方法に従って合成した合成べ プチドでも、 また本発明のタンパク質そのものを用いることもできる。 抗原とな るポリペプチドは、 公知の方法に従って適当な溶液等に調製して、 哺乳動物、 例 えば、 ゥサギ、 マウス、 ラット等に免疫を行えばよいが、 安定的な免疫を行った り抗体価を高めるために抗原べプチドを適当なキャリアタンパク質とのコンジュ ゲートにして用いたり、 アジュバント等を加えて免疫を行うのが好ましい。 As the polypeptide used as the above antigen, a synthetic peptide synthesized according to a known method, or the protein itself of the present invention can be used. A polypeptide serving as an antigen may be prepared in an appropriate solution or the like according to a known method, and immunization may be performed on a mammal, for example, a rabbit, a mouse, a rat, or the like. In order to increase the antibody titer, it is preferable to use the antigen peptide as a conjugate with an appropriate carrier protein, or to carry out immunization by adding an adjuvant or the like.

免疫に際しての抗原の投与経路は特に限定されず、 例えば皮下、 腹腔内、 静脈 内、あるいは筋肉内等のいずれの経路を用いてもよい。具体的には、例えば BALB/c マウスに抗原ポリペプチドを数日〜数週間おきに数回接種する方法等が用いられ る。 また、 抗原の摂取量としては、 抗原がポリペプチドの場合 0 . 3〜0 . 5 m g / 1回程度が好ましいが、 ポリペプチドの種類、 また免疫する動物種によっては 適宜調節される。  The route of administration of the antigen upon immunization is not particularly limited, and any route such as subcutaneous, intraperitoneal, intravenous, or intramuscular route may be used. Specifically, for example, a method of inoculating a BALB / c mouse several times every several days to several weeks with an antigen polypeptide is used. The antigen intake is preferably about 0.3 to 0.5 mg / time when the antigen is a polypeptide, but is appropriately adjusted depending on the type of the polypeptide and the animal species to be immunized.

免疫後、適宜試験的に採血を行って固相酵素免疫検定法 (以下、 これを 「ELISA 法」 と称することがある) やウェスタンブロッテイング等の方法で抗体価の上昇 を確認し、 十分に抗体価の上昇した動物から採血を行う。 これに抗体の調製に用 いられる適当な処理を行えばポリクローナル抗体を得ることができる。 具体的に は、 例えば、 公知の方法に従い血清から抗体成分を精製した精製抗体を取得する 方法等が挙げられる。抗体成分の精製は、遠析、イオン交換ク口マトグラフィー、 ァフイエティークロマトグラフィ一等の方法を用いることができる。  After immunization, test blood is collected as appropriate, and an increase in antibody titer is confirmed by enzyme-linked immunosorbent assay (hereinafter sometimes referred to as “ELISA”) or Western blotting. Blood is collected from animals with elevated antibody titers. A polyclonal antibody can be obtained by subjecting this to an appropriate treatment used for antibody preparation. Specifically, for example, a method of obtaining a purified antibody obtained by purifying an antibody component from serum according to a known method, and the like can be mentioned. The antibody component can be purified by a method such as iontophoresis, ion exchange chromatography, and affinity chromatography.

また、 該動物の脾臓細胞とミエローマ細胞とを用いて公知の方法に従って融合 させたハイプリ ドーマを用いる (Milstein, et al. , Nature, 256, 495 (1975) ) こと によりモノクローナル抗体を作製することもできる。 モノクローナル抗体は、 例 えば以下の方法により取得することができる。  Alternatively, a monoclonal antibody can be produced by using a hybridoma fused with spleen cells and myeloma cells of the animal according to a known method (Milstein, et al., Nature, 256, 495 (1975)). it can. A monoclonal antibody can be obtained, for example, by the following method.

まず、 上記した抗原の免疫により抗体価の高まった動物から抗体産生細胞を取 得する。 抗体産生細胞は、 形質細胞、 及びその前駆細胞であるリンパ球であり、 これは個体の何れから取得してもよいが、 好ましくは脾臓、 リンパ節、 末梢血等 から取得する。 これらの細胞と融合させるミエローマとしては、 一般的にはマウ スから得られた株化細胞、 例えば 8—ァザグァニン耐性マウス (BALBん由来等) ミエローマ細胞株である P3X63- Ag8. 653 (ATCC: CRL - 1580)、 P3 - NSl/lAg4. 1 (理研 セルバンク : RCB0095) 等が好ましく用いられる。 細胞の融合は、抗体産生細胞と ミエローマ細胞を適当な割合で混合し、 適当な細胞融合培地、 例えば RPMI 1640 やイスコフ改変ダルベッコ培地 (IMDM)、 あるレ、はダルベッコ改変イーグル培地 (DMEM) 等に、 5 0 %ポリエチレンダリコール(PEG) を溶解したもの等を用いる ことにより行うことができる。 また電気融合法 (U. Zimmer- mann. et al. , Naturwissenschaften, 68, 577 (1981) ) によっても行うことができる。 First, antibody-producing cells are obtained from an animal whose antibody titer has been raised by immunization with the above-mentioned antigen. The antibody-producing cells are plasma cells and lymphocytes which are precursor cells thereof, which may be obtained from any of the individuals, but is preferably obtained from spleen, lymph nodes, peripheral blood and the like. The myeloma to be fused with these cells is generally a cell line obtained from a mouse, for example, P3X63-Ag8.6653, an 8-azaguanine-resistant mouse (derived from BALB, etc.) myeloma cell line (ATCC: CRL -1580), P3-NSl / lAg4.1 (RIKEN cell bank: RCB0095) and the like are preferably used. For cell fusion, antibody-producing cells and myeloma cells are mixed at an appropriate ratio, and an appropriate cell fusion medium such as RPMI 1640 is used. And Iscov's Modified Dulbecco's Medium (IMDM), Certain, can be performed by using 50% polyethylene dalicol (PEG) dissolved in Dulbecco's Modified Eagle's Medium (DMEM) or the like. It can also be performed by the electrofusion method (U. Zimmermann. Et al., Naturwissenschaften, 68, 577 (1981)).

ハイプリ ドーマは、 用いたミエローマ細胞株が 8—ァザグァニン耐性株である ことを利用して適量のヒポキサンチン 'アミノプテリン'チミジン(HAT)液を含 む正常培地 (HAT培地) 中で 5 % C O2、 3 7 °Cで適当時間培養することにより選 択することができる。 この選択方法は用いるミエ口一マ細胞株によつて適宜選択 して用いることができる。 選択されたハイプリ ドーマが産生する抗体の抗体価を 上記した方法により解析し、 抗体価の高い抗体を産生するハイプリ ドーマを限界 希釈法等により分離し、 分離した融合細胞を適当な培地で培養して得られる培養 上清から硫安分画、 ァフィ二テイクロマトググラフィ一等の適当な方法により精 製してモノクローナル抗体を得ることができる。 また精製には市販のモノクロー ナル抗体精製キットを用いることもできる。 さらには、 免疫した動物と同系統の 動物、 またはヌードマウス等の腹腔内で上記で得られた抗体産生ハイプリ ドーマ を増殖させることにより、 本発明のモノクローナル抗体を大量に含む腹水を得る こともできる。 Hypridoma was prepared using 5% CO 2 in a normal medium (HAT medium) containing an appropriate amount of hypoxanthine 'aminopterin' thymidine (HAT) solution, utilizing the fact that the myeloma cell line used was an 8-azaguanine resistant strain. , At 37 ° C for an appropriate time. This selection method can be appropriately selected and used depending on the myeoma cell line used. The antibody titer of the antibody produced by the selected hybridoma is analyzed by the method described above, the hybridoma producing the antibody with a high antibody titer is separated by a limiting dilution method, etc., and the separated fused cells are cultured in an appropriate medium. A monoclonal antibody can be obtained from the culture supernatant obtained by the purification by an appropriate method such as ammonium sulfate fractionation and affinity chromatography. A commercially available monoclonal antibody purification kit can also be used for purification. Furthermore, ascites containing a large amount of the monoclonal antibody of the present invention can also be obtained by growing the antibody-producing hybridoma obtained above in the abdominal cavity of an animal of the same strain as the immunized animal or a nude mouse. .

また、 本発明のタンパク質としてヒト由来のものを取得した場合には、 かかる ポリペプチド、 あるいはその部分ペプチドを抗原として、 ヒ ト末梢血リンパ球を 移植した Severe combined immune deficiency (SCID) マウスに上記した方法と 同様にして免疫し、 該免疫動物の抗体産生細胞とヒトのミエローマ細胞とのハイ プリ ドーマを作製することによってもヒ ト型抗体を作製することができる (Mosier, D. E. , et al. Nature, 335, 256-259 (1988); Duchosal, M. A. , et al. , Nature, 355, 258-262 (1992) )。  When a human-derived protein is obtained as the protein of the present invention, the above-described polypeptide or a partial peptide thereof is used as an antigen in Severe combined immune deficiency (SCID) mice transplanted with human peripheral blood lymphocytes. Human antibodies can also be prepared by immunization in the same manner as described above and preparing a hybridoma between antibody-producing cells of the immunized animal and human myeloma cells (Mosier, DE, et al. Nature , 335, 256-259 (1988); Duchosal, MA, et al., Nature, 355, 258-262 (1992)).

また、 取得したヒト型抗体を産生するハイプリ ドーマから R Aを抽出し、 目的 のヒト型抗体をコードする遺伝子をクローニングして、 この遺伝子を適当なベタ ターに挿入し、 これを適当な宿主に導入して発現させることにより、 さらに大量 にヒト型抗体を作製することができる。 ここで、 抗原との結合性の低い抗体は、 それ自体既知の進化工学的手法を用いることによりさらに結合性の高い抗体とし て取得することもできる。 一価性抗体等の部分フラグメントは、 例えばパパイン 等を用いて Fab部分と Fc部分を切断し、ァフィ二ティカラム等を用いて Fab部分 を回収することによって作製することができる。 In addition, RA is extracted from the obtained hybridoma producing the human antibody, the gene encoding the desired human antibody is cloned, this gene is inserted into an appropriate vector, and the gene is introduced into an appropriate host. To express even more First, a humanized antibody can be prepared. Here, an antibody having a low binding property to an antigen can be obtained as an antibody having a higher binding property by using an evolutionary engineering technique known per se. A partial fragment such as a monovalent antibody can be prepared by, for example, cleaving the Fab portion and the Fc portion using papain or the like, and collecting the Fab portion using an affinity column or the like.

かくして得られる本発明のタンパク質と特異的に結合する抗体は、 本発明のタ ンパク質に特異的に結合することによって該タンパク質が有する活性を阻害する 中和抗体として用いることもできる。 タンパク質が有する活性を阻害するものの. 選択方法としては特に制限はないが、 例えば、 上記 (2 ) で作製した D NA導入 体に抗体を接触させ、 導入体中の目的タンパク質の機能が阻害されるか否かを解 析する方法等が挙げられる。  The antibody that specifically binds to the protein of the present invention thus obtained can also be used as a neutralizing antibody that specifically binds to the protein of the present invention and thereby inhibits the activity of the protein. Although the activity of the protein is inhibited, the selection method is not particularly limited. For example, the function of the target protein in the introduced product is inhibited by bringing the antibody into contact with the DNA-introduced product prepared in (2) above. There is a method of analyzing whether or not this is the case.

かかる中和抗体は、 臨床へ応用するに際し、 上記有効成分を単独で用いること も可能であるが、 薬学的に許容され得る担体と配合して医薬品組成物として用い ることもできる。 この時の有効成分の担体に対する割合は、 1〜 9 0重量%の間 で変動され得る。 また、 かかる薬剤は種々の形態で投与することができ、 それら の投与形態としては、 錠剤、 カプセル剤、 顆粒剤、 散剤、 あるいはシロップ剤等 による経口投与、 または注射剤、 点滴剤、 リボソーム剤、 坐薬剤等による非経口 投与を挙げることができる。 また、 その投与量は、 症状、 年齢、 体重等によって 適宜選択することができる。  Such a neutralizing antibody can be used alone for the clinical application, but can also be used as a pharmaceutical composition by mixing with a pharmaceutically acceptable carrier. At this time, the ratio of the active ingredient to the carrier can be varied between 1 and 90% by weight. Such drugs can be administered in various forms, such as tablets, capsules, granules, powders, or syrups for oral administration, or injections, drops, ribosomes, Parenteral administration using suppositories and the like can be mentioned. The dose can be appropriately selected depending on the condition, age, body weight, and the like.

( 5 ) 本発明のタンパク質が有する活性の確認および解析  (5) Confirmation and analysis of the activity of the protein of the present invention

本発明のタンパク質は、 これを上記 (2 ) に記載のとおり組み換えタンパク質 として作製し、 これを角?析することにより上記 (1 ) で推測した活性を有してい ることを確認することができる。 さらに上記 (4 ) のとおりに作製した抗体等と の組み合わせにより解析することもできる。  The protein of the present invention is prepared as a recombinant protein as described in the above (2), and the protein is analyzed by chromatography to confirm that it has the activity estimated in the above (1). . Furthermore, analysis can also be performed by combination with an antibody or the like prepared as described in (4) above.

本発明のタンパク質が、 Gタンパク質共役型受容体活性を有することは、 例え ば、 以下の様にして検定することができる。 活性評価のためのリガンドソースに は、 伝達物質、 ホノレモン、 サイトカイン、 Gタンパク質共役型受容体に対する推 定上のァゴニスト、 生物学的な活性を有するペプチド、 細胞培養上清、 組織抽出 物、 ならびに自然界には見いだされないが Gタンパク質共役型受容体を活性化す る化合物を含んでいる。 このリガンドソースを用いて、 下述する、 すなわち、 力 ルシゥム、 レポータージーン、 cAMP、 およびマイクロフイジオメ一ター等を 用いた機能アツセィ、 または結合試験を実施し、 Gタンパク質共役型受容体に対 するリガンドの同定並びに活性の評価を行うことができる。 Whether the protein of the present invention has G protein-coupled receptor activity can be assayed, for example, as follows. Ligand sources for activity evaluation include transmitters, honolemon, cytokines, and G protein-coupled receptors. It contains routine agonists, biologically active peptides, cell culture supernatants, tissue extracts, and compounds not found in nature but that activate G protein-coupled receptors. Using this ligand source, a functional assay or a binding test is performed using a ligand, a reporter gene, cAMP, a microphysiometer, or the like as described below, and a ligand for a G protein-coupled receptor is performed. Can be identified and the activity can be evaluated.

機能アツセィの方法として、 Gタンパク質共役型受容体の以下の機能を利用す ることができる。 例えば、 Gタンパク質共役型受容体は、 これに共役する Gタン パク質に対応して特定の細胞内セカンドメッセンジャーに連動している。 Gq型 の Gタンパク質共役型受容体は PLCの活性化を通して細胞内カルシウム濃度の 増加に機能的に連動しており、 G s型おょぴ G i型 Gタンパク質共役型受容体は、 それぞれアデ二レートシクラーゼの活性化または阻害を通して c AMP濃度を上 昇または減少させることが示されている。  The following functions of the G protein-coupled receptor can be used as a method of functional access. For example, G protein-coupled receptors are linked to specific intracellular second messengers in response to G proteins coupled to them. Gq-type G protein-coupled receptors are functionally linked to the increase in intracellular calcium concentration through activation of PLC, and Gs-type Gi-type G protein-coupled receptors are It has been shown to increase or decrease cAMP levels through activation or inhibition of rate cyclase.

このような機能アツセィの例として、 カルシウムアツセィに関して以下に説明 する。 本発明の組換えタンパク質を発現する HEK 293細胞、 CHO細胞、 c o s細胞等にカルシウム蛍光色素 f u r a 2または F 1 u o 3等を l〜5mM負 荷後、 上記リガンドソースを培地中に添カ卩して培養した後に、 ァゴニストによつ て誘導される一過性カルシウム濃度の上昇を細胞内カルシウム濃度測定装置 (例 えば、 CAF— 100 : 日本光電または FD S S 6000 :浜松ホトニクス等) を用いて評価することができる。  As an example of such a functional assay, a calcium assay will be described below. HEK293 cells, CHO cells, cos cells, etc. expressing the recombinant protein of the present invention are loaded with calcium fluorescent dye fura2 or F1uo3, etc. at l-5 mM, and then the above-mentioned ligand source is added to the medium. After culturing, the transient increase in calcium concentration induced by agonists is evaluated using an intracellular calcium concentration measurement device (for example, CAF-100: Nihon Kohden or FD SS 6000: Hamamatsu Photonics, etc.). can do.

さらに c AMPアツセィとしては、 cAMP応答エレメント (CRE) を用い たレポータージーンアツセィ系または標準的な cAMP定量アツセィが挙げられ る。 具体的には、 例えば本発明の組換えタンパク質を発現している HEK293 細胞、 CHO細胞、 c o s細胞等の培地中に、 上記リガンドソースを添加して培 養した後に、 c AMP産生の刺激または阻害について評価することにより行うこ とができる。  Further, cAMP assays include a reporter gene assay system using a cAMP response element (CRE) or a standard cAMP assay. Specifically, for example, after the above-mentioned ligand source is added and cultured in a medium such as HEK293 cells, CHO cells, and cos cells expressing the recombinant protein of the present invention, stimulation or inhibition of cAMP production is performed. This can be done by evaluating

上記アツセィによって、 カルシウム濃度の一過性上昇または c AMP濃度の変 動を示すァゴニストを、 同様の細胞にベクターのみを導入したモッタインフエク ション細胞において試験して、 応答が受容体を発現している形質転換細胞に特異 的なものであるかどうかを決定することができる。 Due to the above-mentioned atssay, transient increase in calcium concentration or change in cAMP concentration Agonists that are active are tested in mottainfection cells in which only the vector has been introduced into similar cells to determine if the response is specific to the transformed cell expressing the receptor. Can be.

他方、 広範囲の 2次メッセンジャー系の活性化により、 水素イオンが細胞外に 分泌される。 この細胞外水素イオン濃度変化は、 サイトセンサー 'マイクロフィ ジォメーター(CYTO S ENSOR m i c r o p y s i ome t e r) (M o l e c u l a r De v i c e s L t d . , M e n 1 o P a r k , C 土製) 等により検出可能である。 よって、 サイトセンサーは、 本発明の Gタンパク質共 役型受容体のごときエネルギーを使用する細胞内シグナリング経路に連動した受 容体の活性化を検出することができる。  On the other hand, the activation of a wide range of second messenger systems secretes hydrogen ions out of cells. This change in the extracellular hydrogen ion concentration can be detected by a site sensor, such as a microphysiometer (CYTO SENSOR micropy s i ome t er) (Molec cu l ar de vic e s L t d s, Men l o P ark, C soil). Therefore, the site sensor can detect receptor activation linked to an intracellular signaling pathway that uses energy, such as the G protein-coupled receptor of the present invention.

また、 上記のリガンドが本発明タンパク質に結合することは、 標識したリガン ドを用いた通常の結合実験や表面プラズモン共鳴を用いた試験で検討することが できる。  The binding of the above-mentioned ligand to the protein of the present invention can be examined by a normal binding experiment using a labeled ligand or a test using surface plasmon resonance.

以上の機能ァッセィ系は本発明 Gタンパク質共役型受容体活性を有するタンパ ク質のァゴニストやアンタゴニストの評価にも用いることができる。 なお、 本発 明のタンパク質が有する活性の確認はこれらの方法に限定されない。 また、 これ らの機能ァッセィ系は、 後述する本発明のタンパク質の機能賦活物資や機能阻害 物質のスクリーニングや本発明のタンパク質の発現調節物質のスクリーニングに も用いることができる。  The above functional assay system can also be used to evaluate agonists and antagonists of proteins having G protein-coupled receptor activity of the present invention. Note that confirmation of the activity of the protein of the present invention is not limited to these methods. Further, these functional assay systems can also be used for screening for a function activator or a function inhibitor of the protein of the present invention and a screening for a protein expression regulator of the present invention, which will be described later.

本発明のタンパク質の機能解析の方法として一般的には、例えば、 (i)各組織、 疾患、あるいは発生段階における発現状態を比較解析する方法、 (ii)他のタンパ ク質、 DN Aとの相互作用を解析する方法、 (iii) 適当な細胞あるいは個体へ導 入し、表現型の変化を解析する方法、 (iv)適当な細胞あるいは個体において該タ ンパク質の発現を阻害して表現型の変化を解析する方法などが挙げられる。また、 このような方法によれば、 対象タンパク質に特異的な活性を多面的に解析するこ とができる。  As a method for analyzing the function of the protein of the present invention, generally, for example, (i) a method for comparing and analyzing the expression state in each tissue, disease or developmental stage, and (ii) a method for analyzing the expression of other proteins and DNA (Iii) a method for introducing into appropriate cells or individuals and analyzing changes in phenotype; (iv) a method for inhibiting the expression of the protein in appropriate cells or individuals, and And a method of analyzing the change in Further, according to such a method, the activity specific to the target protein can be analyzed from many aspects.

(i) の方法においては、本発明のタンパク質の発現を、 mRNAレベルあるい はタンパク質レベルで解析することができる。 mR NAレベルで発現量を解析す る場合は、 例えば、 in situハイブリダィゼーシヨン法 (In situ hybridization: Application to Developmental Biology & Medicine. , nd. by Harris, N. and Wilkinson, D. G., Cambridge University Press (1990) )、 D NAチップを利用 したハイブリダィゼーシヨン法、 定量 P C R法等が用いられる。 また、 タンパク 質レベルで解析する場合には、 後述する本発明のタンパク質に特異的に結合する 抗体を用いた組織染色法、 E L I S A、 ウェスタンプロッティング法などが挙げ られる。ここで、解析の対象タンパク質に公知のバリアントが存在する場合には、 解析対象タンパク質をコードする c D NAにのみ存在し、 公知のバリアントをコ 一ドする c D NAとはハイブリダィズしないプローブを用いることが好ましい。 定量 P C R法の場合には、 対象 c D N Aと公知バリアント間で異なる長さの増幅 断片ができるプライマーを選択して行う方法(Wong, Y. , Neuroscience Let. , 320: 141-145 (2002) )等が挙げられる。また、タンパク質レベルで解析する場合にも、 対象タンパク質にのみ反応し、 公知のバリアントには反応しない抗体を用いるこ とが好ましい。 In the method (i), the expression of the protein of the present invention is determined at the mRNA level or Can be analyzed at the protein level. When the expression level is analyzed at the mRNA level, for example, the in situ hybridization method (In situ hybridization: Application to Developmental Biology & Medicine., nd. by Harris, N. and Wilkinson, DG, Cambridge University) Press (1990)), a hybridization method using a DNA chip, a quantitative PCR method, and the like. In the case of analyzing at the protein level, a tissue staining method using an antibody that specifically binds to the protein of the present invention described later, an ELISA, a Western plotting method, and the like can be mentioned. If a known variant exists in the protein to be analyzed, use a probe that exists only in the cDNA encoding the protein to be analyzed and does not hybridize with the cDNA that encodes the known variant. Is preferred. In the case of the quantitative PCR method, the method is performed by selecting primers that can generate amplified fragments of different lengths between the target cDNA and the known variant (Wong, Y., Neuroscience Let., 320: 141-145 (2002)). And the like. Also, when analyzing at the protein level, it is preferable to use an antibody that reacts only with the target protein and does not react with a known variant.

(ii) の方法においては、 本発明のタンパク質と既知のタンパク質との相互作 用の有無を調べて、 本発明のタンパク質の機能を解析することができる。 相互作 用の解析法としては、それ自体既知の常法を用いることができる力、具体的には、 例えば、 酵母ツーハイブリッド法、 蛍光偏光解消法、 表面プラズモン法、 ファー ジディスプレイ法、 リボソ一マルディスプレイ法等が挙げられる。 該方法におい ても、 解析対象タンパク質に公知のバリアントが存在する場合には、 公知のバリ アントも同様にして相互作用する物質を解析し、 対象タンパク質特異的に相互作 用する物質を同定することが好ましい。  In the method (ii), the function of the protein of the present invention can be analyzed by examining the presence or absence of interaction between the protein of the present invention and a known protein. As a method of analyzing interaction, the ability to use a conventional method known per se, specifically, for example, yeast two-hybrid method, fluorescence depolarization method, surface plasmon method, phage display method, ribosome A multiple display method and the like can be mentioned. In this method, if a known variant exists in the protein to be analyzed, the known variant also analyzes the interacting substance in the same manner and identifies a substance that interacts specifically with the target protein. Is preferred.

(iii) の方法では、本発明の c D NAを導入する細胞は特に制限はないが、 ヒ ト培養細胞等が特に好ましく用いられる。 D NAの細胞への導入法としては、 上 記 (2 ) に記載のものが挙げられる。 さらに導入細胞の表現型としては、 細胞の 生死、細胞の増殖速度、細胞分化、細胞が神経細胞の場合には神経突起の伸長度、 細胞内タンパク質の局在や移行など顕微鏡等で観察可能なものや、 細胞内の特定 タンパク質の発現変化など生化学的実験により解析可能なものも含む。 これらの 表現型は、 対象タンパク質に公知のバリアントが存在する場合には、 公知のもの も同様に細胞へ導入し、 比較解析することにより、 対象タンパク質が関連する表 現型を同定することができる。 また、 本発明のタンパク質は Gタンパク質共役型 受容体活性を有するものであることがわかっているので、 Gタンパク質共役型受 容体が関連する疾患に見られる表現型等に注目して解析することも好ましい。 In the method (iii), the cell into which the cDNA of the present invention is introduced is not particularly limited, but human cultured cells and the like are particularly preferably used. Methods for introducing DNA into cells include those described in (2) above. In addition, the phenotype of the transfected cells includes cell viability, cell growth rate, cell differentiation, neurite outgrowth if the cells are neurons, This includes those that can be observed with a microscope, such as the localization and translocation of intracellular proteins, and those that can be analyzed by biochemical experiments, such as changes in the expression of specific proteins in cells. When a known variant of the target protein exists, these phenotypes can be similarly introduced into cells, and the phenotype associated with the target protein can be identified by comparative analysis. . Further, since it is known that the protein of the present invention has a G protein-coupled receptor activity, it is also possible to analyze the protein by focusing on the phenotype and the like found in diseases associated with the G protein-coupled receptor. preferable.

( iv) の方法では、 後述するオリゴヌクレオチドを用いた方法や、 R NAイン ターフェアレンス法により効率的に行うことができる。 この方法においても、 解 析する対象タンパク質に、 公知のバリアントが存在する場合には、 公知のバリア ントやその他のバリアントについても同様の解析を行い、 比較解析することによ り対象タンパク質特異的な機能を同定することができる。  In the method (iv), the method can be efficiently performed by a method using an oligonucleotide described later or an RNA interference method. In this method, if a known variant is present in the target protein to be analyzed, the same analysis is performed for the known variant and other variants, and the target protein-specific protein is obtained by comparative analysis. Function can be identified.

( 6 ) 本発明のタンパク質が有する活性を調節する分子のスクリーニング 本発明のタンパク質に特異的に結合し、かつ本発明のタンパク質の機能(活性) を阻害、 拮抗または増強する作用を有する物質をスクリーニングすることにより 本発明のタンパク質の機能調節物質 (以下、 これを 「調節物質」 と称することが ある) を得ることができる。 (6) Screening for a molecule that regulates the activity of the protein of the present invention Screening for a substance that specifically binds to the protein of the present invention and has an effect of inhibiting, antagonizing or enhancing the function (activity) of the protein of the present invention By doing so, a function modulator of the protein of the present invention (hereinafter, this may be referred to as “modulator”) can be obtained.

この調節物質のスクリーニング方法は、本発明のタンパク質に特異的に結合し、 且つ該タンパク質の活性を阻害、 拮抗または増強する作用を有する物質が得られ る方法であれば如何なるものであってもよい。 例えば、 まず本発明のタンパク質 と被検物質とを接触させ、 該タンパク質との結合性を指標として選抜した後に、 本発明のタンパク質が有する活性の変化を指標として被検物質を選抜する方法を 用いることができる。  This method of screening for a regulatory substance may be any method as long as it can obtain a substance that specifically binds to the protein of the present invention and has an activity of inhibiting, antagonizing, or enhancing the activity of the protein. . For example, a method is used in which a protein of the present invention is first brought into contact with a test substance, selection is performed using the binding property of the protein as an index, and then a change in the activity of the protein of the present invention is used as an index. be able to.

被検物質としては、 本発明のタンパク質と相互作用して、 該タンパク質が有す る活性に影響を及ぼす可能性のある物質であれば如何なるものであってもよいが、 具体的には、 例えば、 ペプチド、 タンパク質、 非ペプチド性化合物、 低分子化合 物、 合成化合物、 発酵生産物、 細胞抽出液、 動物組織抽出液等が挙げられる。 こ れらの物質は新規な物質であってもよいし、 公知の物質であってもよい。 被検物 質と本発明のタンパク質の相互作用の解析法としては、 それ自体既知の常法を用 いることができるが、 具体的には、 例えば、 酵母ツーハイブリッド法、 蛍光偏光 解消法、 表面プラズモン法、 ファージディスプレイ法、 リボソ一マルディスプレ ィ法、 あるいは上記 (4 ) に記載した抗体との競合解析法等が挙げられる。 この ような方法により、 本発明のタンパク質に結合する活性を見いだされた物質は、 次に該物質の存在下で本発明のタンパク質が有する活性がどのような影響を受け るかを解析することによって、 調節物質として用いられるか否かが同定される。 ここで、 医薬活性成分のスクリーニングを目的とするため、 用いる本発明の D NA、 あるいは組み換えタンパク質については、 上記したヒ トのホモログタンパ ク質またはオルソログタンパク質を用いることが好ましい。 さらに上記方法によ つてスクリーユングされた物質は、 これらの生体内でのスクリーユングによって 医薬候捕としての選択を行ってもよい。 The test substance may be any substance as long as it interacts with the protein of the present invention and may affect the activity possessed by the protein. , Peptide, protein, non-peptide compound, small molecule compound Products, synthetic compounds, fermentation products, cell extracts, animal tissue extracts, and the like. These substances may be novel substances or known substances. As a method for analyzing the interaction between the test substance and the protein of the present invention, a conventional method known per se can be used. Specifically, for example, the yeast two-hybrid method, the fluorescence depolarization method, the surface The plasmon method, the phage display method, the ribosomal display method, or the competition analysis method with the antibody described in the above (4) can be used. By such a method, the substance found to bind to the protein of the present invention is then analyzed by analyzing how the activity of the protein of the present invention is affected in the presence of the substance. Whether it is used as a modulator or not is identified. Here, for the purpose of screening for a pharmaceutically active ingredient, it is preferable to use the above-described human homologous protein or orthologous protein for the DNA or recombinant protein of the present invention to be used. Further, the substance screened by the above method may be selected as a drug trap by screening in vivo.

具体的な解析方法としては、 例えば、 Gタンパク質共役型受容体活性を調節す る物質を解析する場合には、 (2 )に記載した D N A導入体に基質となるタンパク 質も同様の方法で導入する。 この導入体について選択された物質の存在下/また は非存在下で基質となるタンパク質の脱リン酸化をそれ自体既知の通常用いられ る方法により解析する。 具体的には、 上記 (5 ) に記載の方法等を用いて行うこ とができる。 カルシウム濃度、 c AM P濃度等が、 物質の非存在下の場合と比べ て増加した場合には、 該物質は Gタンパク質共役型受容体活性化物質として機能 する可能性があり、 また低下、 または阻害された場合には物質は Gタンパク質共 役型受容体阻害物質として機能する可能性があると同定できる。 ここで、 医薬活 性成分のスクリーニングを目的とするため、 用いる本発明の D NA、 あるいは組 み換えタンパク質については、 上記したヒ トのホモログタンパク質またはオルソ ログタンパク質を用いることが好ましい。 さらに上記方法によってスクリーニン グされた物質は、 これらの生体内でのスクリーニングによって医薬候補としての 選択を行ってもよい。 As a specific analysis method, for example, when analyzing a substance that regulates G protein-coupled receptor activity, a protein serving as a substrate is introduced into the DNA transfectant described in (2) by the same method. I do. The dephosphorylation of the protein serving as a substrate in the presence / absence of the substance selected for the transductant is analyzed by a commonly used method known per se. Specifically, it can be performed using the method described in (5) above. If the calcium concentration, c AMP concentration, etc., are increased as compared to the absence of the substance, the substance may function as a G protein-coupled receptor activator, or may decrease, or When inhibited, the substance can be identified as possibly functioning as a G protein-coupled receptor inhibitor. Here, for the purpose of screening for a pharmaceutically active ingredient, it is preferable to use the above-mentioned human homologous protein or orthologous protein as the DNA or recombinant protein of the present invention to be used. Furthermore, substances screened by the above method can be used as drug candidates by screening in vivo. You may make a selection.

本発明のタンパク質が有する Gタンパク質共役型受容体活性を有する Gタンパ ク質共役型受容体は、 細胞外のシグナルを伝達する必須の膜タンパク質のスーパ 一ファミ リーである。 G P C Rには、 例えばドーパミン、 ェピネフリン、 ヒスタ ミン、グルタミン酸(向代謝性の効果)、ァセチルコリン(ムス力リン性の効果)、 セロトニン等の生体ァミンに対する受容体や、 プロスタグランジン、 血小板活†生 化因子、 ロイコトリエンのような炎症の脂質媒介物等に対する受容体や、 カルシ トニン、 C5a ァナフイラトキシン、 卵胞刺激ホルモン、 ゴナドトロピン放出ホル モン、 ニューロキニン、 ォキシトシン、 トロンビン等のようなペプチドホ/レモン に対する受容体や、 網膜感光色素、 嗅覚刺激性分子のような感覚性のシグナル媒 介物に対する受容体が含まれる。  The G protein-coupled receptor having the G protein-coupled receptor activity of the protein of the present invention is a superfamily of essential membrane proteins that transmit extracellular signals. GPCRs include receptors for living amines such as dopamine, epinephrine, histamine, glutamic acid (a metabolic effect), acetylcholine (a musculinic effect), and serotonin, as well as prostaglandins and platelet activation. Receptors for factors, lipid mediators of inflammation such as leukotriene, and receptors for calcitonin, C5a anaphylatoxin, follicle-stimulating hormone, gonadotropin-releasing hormone, neurokinin, oxytocin, thrombin, etc. And receptors for sensory signaling media such as retinal photopigments and olfactory stimulants.

そこで、 本スクリーニング方法により同定できる化合物は、 高血圧、 狭心症、 心筋梗塞、 ァテローム性動脈硬化、 潰瘍、 喘息、 アレルギー、 精神病、 アルッハ イマ一病、舞踏病、統合失調症、 うつ病、片頭痛、虚血性脳疾患、不安症、嘔吐、 良性前立腺肥大、 パーキンソン病、 急性心不全、 糖尿病、 糖尿病性網膜症、 眼科 疾患、 内分泌疾患、免疫疾患、炎症性疾患、 呼吸器系疾患、低血圧、尿閉、不妊、 避妊、 肝疾患、 および骨粗霜症などの治療剤として用いられ得るものである。 かかる調節物質は、 臨床へ応用するに際し、 上記有効成分を単独で用いること も可能であるが、 薬学的に許容され得る担体と配合して医薬品組成物として用い ることもできる。 この時の有効成分の担体に対する割合は、 1〜9 0重量%の間 で変動され得る。 また、 かかる薬剤は種々の形態で投与することができ、 それら の投与形態としては、 錠剤、 カプセル剤、 顆粒剤、 散剤、 あるいはシロップ剤等 による経口投与、 または注射剤、 点滴剤、 リボソーム剤、 坐薬剤等による非経口 投与を挙げることができる。 また、 その投与量は、 症状、 年齢、 体重等によって 適宜選択することができる。  Therefore, compounds that can be identified by this screening method include hypertension, angina, myocardial infarction, atherosclerosis, ulcer, asthma, allergy, psychosis, Alhaima's disease, chorea, schizophrenia, depression, and migraine , Ischemic brain disease, anxiety, vomiting, benign prostatic hypertrophy, Parkinson's disease, acute heart failure, diabetes, diabetic retinopathy, ophthalmic disease, endocrine disease, immune disease, inflammatory disease, respiratory disease, hypotension, urine It can be used as a remedy for closure, infertility, contraception, liver disease, and osteoporosis. Such modulators can be used alone as the above active ingredients when applied to clinical applications, but can also be used as pharmaceutical compositions by blending with a pharmaceutically acceptable carrier. At this time, the ratio of the active ingredient to the carrier can be varied between 1 and 90% by weight. Such drugs can be administered in various forms, such as tablets, capsules, granules, powders, or syrups for oral administration, or injections, drops, ribosomes, Parenteral administration with suppositories and the like can be mentioned. In addition, the dose can be appropriately selected depending on symptoms, age, weight, and the like.

( 7 ) 本発明の D NAの発現調節物質のスクリーニング  (7) Screening of the DNA expression regulator of the present invention

スクリーユングの方法としては、 被検物質の存在下で本発明のタンパク質、 あ るいはそれをコードする mR N Aの発現量を解析する方法等が挙げられる。 具体 的には、 例えば、 上記 (2 ) に記載した本発明のタンパク質を発現する細胞を被 検物質を含む適当な培地で培養し、 該細胞内に発現している本発明のタンパク質 量を E L I S A等の常法を用いて解析するか、 あるいは該細胞内の本発明のタン パク質をコードする m R NA量を、 定量的逆転写 PCR法や、 ノーザンブロット法 等により解析することにより行うことができる。 Screening methods include the use of the protein of the present invention in the presence of a test substance. Or a method of analyzing the expression level of mRNA encoding the same. Specifically, for example, cells expressing the protein of the present invention described in (2) above are cultured in an appropriate medium containing a test substance, and the amount of the protein of the present invention expressed in the cells is determined by ELISA. Or by analyzing the amount of mRNA encoding the protein of the present invention in the cells by quantitative reverse transcription PCR, Northern blotting, or the like. Can be.

被検物質としては、 上記 (6 ) に記載のものを用いることができる。 この角爭析 により、 被検物質の非存在下で培養された当該細胞内で発現されたタンパク質、 あるいは m R NA量と比べてその量が増加すれば、 物質は本発明の D N Aの発現 促進物質として機能する可能性があり、 逆に減少した場合には、 物質は本発明の D NAの発現阻害物質として用いられ得ると判断することができる。  As the test substance, those described in the above (6) can be used. As a result of this angular dissolution, if the amount of the protein or mRNA expressed in the cells cultured in the absence of the test substance is increased as compared with the amount of the test substance, the substance promotes the expression of the DNA of the present invention. When the substance may function as a substance, and conversely decreases, it can be determined that the substance can be used as the DNA expression inhibitor of the present invention.

かかる発現調節物質は、 臨床へ応用するに際し、 上記有効成分を単独で用いる ことも可能であるが、 薬学的に許容され得る担体と配合して医薬品組成物として 用いることもできる。 この時の有効成分の担体に対する割合は、 1〜9 0重量% の間で変動され得る。 また、 かかる薬剤は種々の形態で投与することができ、 そ れらの投与形態としては、 錠剤、 カプセル剤、 顆粒剤、 散剤、 あるいはシロップ 剤等による経口投与、 または注射剤、 点滴剤、 リボソーム剤、 坐薬剤等による非 経口投与を挙げることができる。 また、 その投与量は、 症状、 年齢、 体重等によ つて適宜選択することができる。  The above-mentioned active ingredient can be used alone for clinical application, but it can also be used as a pharmaceutical composition by blending it with a pharmaceutically acceptable carrier. At this time, the ratio of the active ingredient to the carrier can be varied between 1 and 90% by weight. The drug can be administered in various forms. Examples of the dosage form include tablet, capsule, granule, powder, syrup, and the like, oral administration, injection, drip, ribosome And parenteral administration with suppositories and the like. In addition, the dose can be appropriately selected depending on symptoms, age, weight, and the like.

( 8 ) 本発明の D N A導入動物  (8) The DNA-introduced animal of the present invention

上記 (1 ) に記載の、 本発明の D N Aを含む導入 D N Aを構築し、 ヒト以外の 哺乳動物の受精卵に導入して、 これを雌個体子宮に移植して発生させることによ り、本発明の D NAが導入された非ヒト哺乳動物を作製することができる。より、 具体的には、 例えば、 雌個体をホルモン投与により過剰排卵させた後、 雄と交配 し、 交配後 1日目の卵管から受精卵を摘出し、 該受精卵に導入 D NAをマイクロ インジェクション等の方法により導入する。 この後、 適当な方法で培養した後、 生存している受精卵を、 偽妊娠させた雌個体 (仮親) の子宮に移植して出産させ る。 新生仔に目的の D NAが導入されているか否かは、 該個体の細胞から抽出し た D N Aのサザンブロット解析を行うことにより同定することができる。 ヒト以 外の哺乳動物としては、 例えばマウス、 ラット、 モルモッ ト、 ハムスター、 ゥサ ギ、 ャギ、 ブタ、 ィヌ、 ネコ等が挙げられる。 The transfected DNA containing the DNA of the present invention described in the above (1) is constructed, introduced into a fertilized egg of a mammal other than a human, and transplanted into a female individual uterus to generate the present DNA. A non-human mammal into which the DNA of the present invention has been introduced can be produced. More specifically, for example, a female individual is superovulated by hormone administration, then mated with a male, a fertilized egg is excised from the oviduct on the first day after mating, and the DNA is introduced into the fertilized egg with a micro DNA. It is introduced by a method such as injection. After culturing by an appropriate method, the surviving fertilized eggs are transplanted into the uterus of a pseudopregnant female individual (foster parent) and allowed to give birth. You. Whether or not the desired DNA has been introduced into the newborn can be identified by Southern blot analysis of DNA extracted from cells of the individual. Non-human mammals include, for example, mice, rats, guinea pigs, hamsters, rabbits, goats, pigs, dogs, cats, and the like.

かくして得られた本発明の DNA導入動物は、 この個体を交配し、 導入された D N Aが安定的に保持されていることを確認しながら通常の飼育環境で継代飼育す ることによりその子孫を得ることができる。 また、 体外受精を繰り返すことによ りその子孫を得て、 系統を維持することもできる。  The DNA-introduced animal of the present invention thus obtained is bred with this individual, and its offspring are bred in a normal breeding environment while confirming that the introduced DNA is stably maintained. Obtainable. Also, by repeating in vitro fertilization, the offspring can be obtained and the strain can be maintained.

本発明の D N Aが導入された非ヒト哺乳動物は、 本発明の D NAの生体内にお ける機能の解析や、 またこれを調節する物質のスクリーニング系等として用いる ことができる。  The non-human mammal into which the DNA of the present invention has been introduced can be used as an analysis of the function of the DNA of the present invention in a living body, or as a screening system for a substance regulating the function.

( 9 ) 本発明のタンパク質及びそれをコードする塩基配列を含む D NAの他の利 用  (9) Other uses of the protein of the present invention and the DNA containing the nucleotide sequence encoding the same

本発明のタンパク質は、 それを基盤上に結合させた担体として利用することが できる。 また、 本発明のタンパク質をコードする塩基配列、 例えば、 配列番号 1 〜1 6のいずれかに記載の塩基配列を有する D N A及びその部分断片は、 それら を基板上に結合させた担体としてもちいられ得る。 これらを、以下、 「プロテイン チップ」、 「D NAチップ」または「D N Aアレイ」 (D NAマイクロアレイ及び D NAマクロアレイ) と称することがある。 これらのプロテインチップ、 又は D N Aチップもしくはアレイには、 本発明のタンパク質や D N A以外に、 他のタンパ ク質ゃ D N Aが含まれていてもよい。  The protein of the present invention can be used as a carrier having it bound on a substrate. In addition, a nucleotide sequence encoding the protein of the present invention, for example, a DNA having the nucleotide sequence of any one of SEQ ID NOs: 1 to 16 and a partial fragment thereof can be used as a carrier obtained by binding them to a substrate. . Hereinafter, these may be referred to as “protein chips”, “DNA chips” or “DNA arrays” (DNA microarrays and DNA macroarrays). These protein chips or DNA chips or arrays may contain other proteins ゃ DNA in addition to the protein and DNA of the present invention.

ここで、 タンパク質や D N Aを結合させる基盤としては、 ナイロン膜、 ポリプ ロピレン膜等の樹脂基板、 ニトロセルロース膜、 ガラスプレート、 シリコンプレ ート等が用いられるが、 ハイプリダイゼーシヨンの検出を非 R I的に、 例えば、 蛍光物質等を用いて行う場合には、 蛍光物質を含まないガラスプレート、 シリコ ンプレート等が好適に用いられる。 また該基盤へのタンパク質、 あるいは D NA の結合は、 それ自体公知の通常用いられる方法により容易に行うことができる。 これらのプロテインチップ、 DNAチップ、 あるいは DNAアレイも、 本発明の 範囲に含まれる。 Here, a resin substrate such as a nylon film or a polypropylene film, a nitrocellulose film, a glass plate, a silicon plate, or the like is used as a substrate for binding proteins and DNA, but the detection of hybridization is not performed by non-RI. Specifically, for example, in the case of using a fluorescent substance or the like, a glass plate or a silicon plate containing no fluorescent substance is preferably used. The binding of the protein or DNA to the substrate can be easily carried out by a commonly used method known per se. These protein chips, DNA chips, or DNA arrays are also included in the scope of the present invention.

また、 本発明のタンパク質のアミノ酸配列及ぴ DNAの塩基配列は、 配列情報 としても用いることができる。 ここで、 本発明の DN Aの塩基配列には、 対応す る RNAの塩基配列も含まれる。 すなわち、 得られたアミノ酸配列や塩基配列を コンピューターが読みとり可能な所定の形式で適当な記録媒体に格納することに より、 ァミノ酸配列や塩基配列のデータベースが構築できる。 このデータベース には、 他の種類のタンパク質やそれをコードする DN Aの塩基配列が含まれてい てもよい。 また、 本発明においてデータベースとは、 上記配列を適当な記録媒体 に書き込み、 所定のプログラムに従って検索を行うコンピューターシステムをも 意味する。 ここで適当な記録媒体としては、 例えば、 フレキシブルディスク、 ハ ードディスク、 磁気テープ等の磁気媒体、 CD— ROM、 MO、 CD-R, CD — RW、 DVD-R, DVD— RW等の光ディスク、 半導体メモリ等を挙げるこ とができる。 実施例  Further, the amino acid sequence of the protein of the present invention and the nucleotide sequence of DNA can also be used as sequence information. Here, the nucleotide sequence of the DNA of the present invention includes the nucleotide sequence of the corresponding RNA. That is, a database of amino acid sequences and nucleotide sequences can be constructed by storing the obtained amino acid sequences and nucleotide sequences in an appropriate recording medium in a computer-readable predetermined format. This database may contain the nucleotide sequences of other types of proteins and the DNA encoding them. Further, in the present invention, the database also means a computer system that writes the above-mentioned sequence on an appropriate recording medium and performs a search according to a predetermined program. Suitable recording media include, for example, magnetic media such as flexible disks, hard disks, and magnetic tapes; optical disks such as CD-ROM, MO, CD-R, CD-RW, DVD-R, and DVD-RW; and semiconductors. Examples include memory. Example

以下、 実施例を挙げて本発明を詳細に説明するが、 本発明の範囲はこれらの実 施例により限定されるものではない。  Hereinafter, the present invention will be described in detail with reference to Examples, but the scope of the present invention is not limited by these Examples.

実施例 1 cDNAライブラリーの調製 Example 1 Preparation of cDNA Library

(1) mRNAの調製  (1) Preparation of mRNA

mRNA調製マウス (C 57 B L/6) 各器官または組織 0. 5〜l gを 10 m 1の懸濁液でホモジェナイズし、 pH4. 0の 2M酢酸ナトリウム lm 1 と、 同量のフユノール/クロ口ホルム(体積比 5: 1)混液を加え抽出した。 抽出後水 層に同量のィソプロパノールを加えると、 RNAが水相から分離沈澱した。  mRNA-prepared mouse (C57BL / 6) 0.5 to lg of each organ or tissue is homogenized with a 10 ml suspension, and the same amount of fuminol / clonal form as lm 1 of 2M sodium acetate at pH 4.0 (Volume ratio 5: 1) The mixture was added and extracted. When the same amount of isopropanol was added to the aqueous layer after the extraction, RNA separated and precipitated from the aqueous phase.

この試料を氷の上で 1時間インキュベーションした後、 1 5分間、 4, 000 r p mで冷却遠心機にかけ、 沈澱物を回収した。 この検体を 70 %エタノ一ルで洗 い、 8 m l の水に溶解後、 2 m l の 5 M N a C 1 , 1 % C TAB (cetyltrimethy- 1 ammonium bromide)、 4M尿素、 50 mM T r i sを含む p H 7. 0の水溶液 1 6 m 1を加えることで RNAを沈澱させ、 ポリサッカライド を除いた(CTAB沈澱)。 After incubating the sample on ice for 1 hour, the precipitate was collected in a refrigerated centrifuge at 4,000 rpm for 15 minutes. Wash this sample with 70% ethanol, dissolve in 8 ml of water, and add 2 ml of 5 M Na C 1, 1% C TAB. RNA was precipitated by adding 16 ml of an aqueous solution of pH 7.0 containing (cetyltrimethy-1 ammonium bromide), 4 M urea, and 50 mM Tris to remove polysaccharide (CTAB precipitation).

続いて室温で 4, 00 0 r pm、 1 5分間遠心機にかけ、 R N Aを 4 m 1の 7 Mグァニジン一 C 1に溶解した。 そして 2倍量のエタノールを加えた後、 氷上で 1時間インキュベーションし、 4, 000 r pm、 1 5分間遠心機にかけ、 生じ た沈澱物を 70%エタノールで洗い RNAを回収した、 これを再度水に溶解し、 RNAの純度を OD比 2 60Z280 (> 1. 8) と 230Z260 « 0. 4 5) を読むことによって計測した。  The RNA was then centrifuged at room temperature at 4,000 rpm for 15 minutes to dissolve the RNA in 4 ml of 7 M guanidine-C1. After adding twice the volume of ethanol, the mixture was incubated on ice for 1 hour, centrifuged at 4,000 rpm for 15 minutes, and the resulting precipitate was washed with 70% ethanol to recover RNA. And the RNA purity was determined by reading the OD ratios 260Z280 (> 1.8) and 230Z260 «0.45).

(2) 第 1鎖 c DNAの調製  (2) Preparation of first strand cDNA

上記 (1) で調製した mRNA 1 5 μ gを使って逆転写酵素 3, 000 u n i tにより、 最終容量 1 65 μ 1の反応液中で、 5—メチルー d CTP、 d AT P、 dTTP、 dGTPを各々 0. 54mM、 0. 6Mトレハロース、 50 mM T r i s—HC l ( Η 8. 3)、 7 5 mM KC 1、 3 mM Mg C 1 2、 1 0 mM DTT, 5 2 n g/μ 1 B SA、 RNa s eインヒビター 5 u n i t の条件下で逆転写反応を行った。 制限酵素 Xh o Iの認識配列を含むオリゴヌク レオチド (配列番号 3 3) (配列中、 Vは A,G,又は Cを示し、 Nは A, G, C,又は T を示す) 1 2. 6 μ 1をプライマーとして用いた。  Using 5-μg of the mRNA prepared in (1) above, 5-methyl-dCTP, dATP, dTTP, and dGTP were reacted with 3,000 units of reverse transcriptase in a final reaction volume of 165 μl. 0.54 mM, 0.6 M trehalose, 50 mM Tris-HCl (l8.3), 75 mM KC1, 3 mM MgC12, 10 mM DTT, 52 ng / μ1 BSA respectively The reverse transcription reaction was performed under the condition of 5 units of RNase inhibitor. Oligonucleotide containing recognition sequence of restriction enzyme Xho I (SEQ ID NO: 33) (wherein, V indicates A, G, or C, and N indicates A, G, C, or T) 12.6 μ1 was used as a primer.

この反応を始める際、 反応液の 1Z4を採取し、 それに 1. 5 z lの [ひ一32 P] 一 dGTP (300 0 C i /mm o 1、 1 0 μ C i 1 ; Am e r s a m社製) を加えるこことにより、 第 1鎖 cDNAの合成効率を測定した。 R I標 識した反応液の 0. 5 μ 1を DE— 8 1ペーパー上にスポットし、 0. 5Μリン 酸ナトリウム緩衝液 (ρΗ7. 0) で 3回洗った前後の R I活性を測定し、 計算 した。 その後、 R I標識した反応液と非標識の反応液を混合し、 0. 5Μ ED ΤΑ 8 ^ 1 , 1 0 % S D S 2 z l、プロティナーゼ (P r o t e i n a s e) K 20 / gを加え、 4 5 °Cで 1 5分間加熱した。 フエノール Zクロ口ホルムに よる抽出、エタノール沈澱後、沈澱を RN a s eフリーに処理してある水(以下、 「RNa s eフリー水」 と称することがある) 47 μ 1に溶解した。 At the beginning of this reaction, the 1Z4 of the reaction solution was taken and thereto 1. 5 zl in the shed one 32 P] one dGTP (300 0 C i / mm o 1, 1 0 μ C i 1; manufactured Am Ersam Co.) Thus, the efficiency of the synthesis of the first strand cDNA was measured. 0.5 μl of the RI-labeled reaction solution was spotted on DE-81 paper, and the RI activity before and after washing three times with 0.5Μ sodium phosphate buffer (ρΗ7.0) was measured and calculated. did. Then, mix the RI-labeled reaction solution with the unlabeled reaction solution, add 0.5Μ ED ^ 8 ^ 1, 10% SDS 2 zl, and proteinase (Proteinase) K 20 / g, and add the mixture at 45 ° C. Heated for 15 minutes. After extraction with phenol Z-form and ethanol precipitation, the precipitate was treated with RNAse-free water (hereinafter referred to as (Sometimes referred to as "RNase-free water").

(3) 5' キャップ構造及ぴ 3, 末端へのピオチン付カロ  (3) 5 'cap structure and 3.

RN Αジオールのビォチン化 RN Αのジオール部位 (C a p構造のある 5, 末 端と、 ポリ A鎖のある 3, 末端のリポースの双方に存在) にビォチンを結合させ るために、 2段階の反応を行った。 それらは、 ジオール基の酸化とそれに続くビ ォチンヒドラジドと酸化 RNA体のカップリング反応である。 まず、 逆転写反応 で得られた RNA—第 1鎖 c DNA複合体 15 μ gを、 6. 6 mM酢酸ナトリウ ム緩衝液 (PH4. 5) と、 酸化剤として過ヨウ素酸ナトリウムを用いて 5 Ο μ Biotinylation of RN Α diol Two steps to bind biotin to the diol site of RN ((present at both the 5 'end with the Cap structure and the 3' end report with the poly A chain) The reaction was performed. These are the oxidation of the diol group followed by the coupling reaction of biotin hydrazide with the oxidized RNA. First, 15 μg of the RNA-first strand cDNA complex obtained by the reverse transcription reaction was purified using 6.6 mM sodium acetate buffer (PH4.5) and sodium periodate as an oxidizing agent. Ο μ

1の反応液中で処理した。この酸化反応は遮光条件の下、氷上で 45分間行った。 続いて、 5Μ塩化ナトリウム 1 1 μ 1、 10% SD S 0. 5 μ 1、 そし て同量のイソプロパノールを加え、 60分間氷上に放置した後、 4 °Cで 1 5分間、Treated in reaction 1 This oxidation reaction was performed on ice for 45 minutes under light-shielded conditions. Then add 11 μl of 5Μ sodium chloride, 0.5 μl of 10% SDS and 0.5 μl of the same amount of isopropanol, leave on ice for 60 minutes, and then at 4 ° C for 15 minutes.

15, 000 r pm遠心し沈澱を取得した。 沈澱物は 70 %エタノールで洗い、 RNa s eフリー水 50 μ 1に再溶解させた。その試料に 1 Μ酢酸ナトリゥム(ρ Η6. 1) 5 μ 1、 10% SDS 5 μ 1、 10 mMビォチンヒドラジド ( S i gma社製) 150 ^ 1を加え、 室温 ( 22〜 26 °C) で終夜反応させた。 最 後に、 5 μ 1の 5M Na C l、 1 M酢酸ナトリゥム (pH6. 1) 75 1、 及び 2. 5倍量のエタノールを加え、 1時間の氷上冷却後、 4°Cにおいて 15分 間遠心し、 ピオチン化した。 反応後、 反応液を 15分間遠心し、 再度 RNA— D N A複合体を沈澱させた。 沈澱物は 70 %エタノールで 1回、 更に 80 %ェタノ ールで 1回洗い、 R N a s eフリ一水 70 1に溶角军した。 The precipitate was obtained by centrifugation at 15,000 rpm. The precipitate was washed with 70% ethanol and redissolved in 50 μl of RNase-free water. To the sample, add 5 μl of sodium acetate (ρ 6.1), 5 μl of 10% SDS, 10 μm of biotin hydrazide (Sigma) 150 ^ 1, and add it to room temperature (22 to 26 ° C ) Overnight. Finally, add 5 μl of 5 M NaCl, 1 M sodium acetate (pH 6.1), 751, and 2.5 volumes of ethanol, cool on ice for 1 hour, and centrifuge at 4 ° C for 15 minutes And biotinylated. After the reaction, the reaction solution was centrifuged for 15 minutes to precipitate the RNA-DNA complex again. The precipitate was washed once with 70% ethanol and once with 80% ethanol, and dissolved in RNase free water 701.

(4) RNa s e Iによる完全長 c D N Aの選択  (4) Selection of full-length c DN A by RNa s e I

上記 (3) で取得したビォチン化 RNA— DN A複合体について、 1本鎖 RN Aを消化する RNa s e Iで処理することにより、 逆転写反応時に完全な c D NAの伸長が得られなかった mRNA、 および mRNAの 3, 末端に標識された ビォチン残基を取り除いた。 具体的には、 上記 (3) で得られた試料 70 μ 1に 10 X RN a s e Iバッファー(l O OmM T r i s— H C 1 ( p H 7.5 )、 5 OmM EDTA、 2M N a OA c) 10 μ 1、 RN a s e I (RNa s e On e™ ; P r ome g a社製) 200 u n i tを加えて、 37 °Cで 1 5分 間 1本鎖 R N Aを消化した。 When the biotinylated RNA-DNA complex obtained in (3) above was treated with RNase I that digests single-stranded RNA, complete cDNA extension was not obtained during the reverse transcription reaction. mRNA and the biotin residue labeled at the 3 'end of the mRNA were removed. Specifically, 10 X RNase I buffer (10 OmM Tris-HC1 (pH 7.5), 5 OmM EDTA, 2 M NaOAc) was added to 70 μl of the sample obtained in (3) above. μ1, RNase I (RNa s eOne ™; manufactured by Promega) was added, and the single-stranded RNA was digested at 37 ° C for 15 minutes.

(5) 完全長 cDNAの採取  (5) Collection of full-length cDNA

ストレプトアビジンコートしたマグネティックビーズに c DN Aが非特異的吸 着するのを防止するため、 100 μ gの酵母 t RNA (DNa s e I処理した もの) を 5mg (500 μ 1 ) のマグネティックビーズ (ma g n e t i c p o r o u s g l a s s (MP G) p a r t i c l e s c o a t e d w i t h s t r e p t a v i d i n (CPG, NJ)) に加え、 1時間氷上に放置した 後、 50mM EDTA、 2M N a C 1の溶液にて洗った。  To prevent nonspecific adsorption of cDNA to streptavidin-coated magnetic beads, 100 μg of yeast tRNA (treated with DNase I) was added to 5 mg (500 μl) of magnetic beads (matrix). In addition to gneticporousglass (MPG) particles coated withst reptavidin (CPG, NJ)), the mixture was left on ice for 1 hour, and then washed with a solution of 50 mM EDTA and 2 M NaC1.

このビーズを 50mM EDTA、 2M N a C 1の溶液 500 μ 1中に懸濁 し、 (4)で取得した RNa s e I処理を施された c D N Aを加えた。室温にて 30分間撹拌することで、マグネテイツクビーズと完全長 c D N Aを結合させた。 完全長 c DNAを捕獲したビーズを 5 OmM EDTA、 2M N a C 1の溶液 で 4回、 0. 4%SDS、 50 μ gZiU 1酵母 t RNAで 1回、 l OmM N a C 1、 0. 2mM EDTA、 1 OmMT r i s— HC 1 (pH7. 5)、 20% グリセ n—ルで 1回、 50 μ g/μ 1酵母 t RNA水溶液で 1回、 RNa s e H バッファー (20 mMT r i s _HC l (p H 7. 5)、 10 mM Mg C 12、 2 OmM KC 1、 0. 1 mM EDTA、 0. 1 mM ジチオスレイト一ノレ(D TT) で 1回洗浄した後、 RNa s e H用バッファー 100 μ 1に懸濁し、 R Na s e H 3 un i tを加え、 37°C下 30分間加温した。 その後、 10% SDS 1 μ 1、 0. 5Μ EDTA 2 μ 1を加えて、 10分間、 65°Cに曝 し、 その上清を回収した。 The beads were suspended in 500 μl of a solution of 50 mM EDTA and 2 M NaCl, and the RNase I-treated cDNA obtained in (4) was added. By stirring at room temperature for 30 minutes, the magnetic beads and the full-length cDNA were bound. The beads that captured the full-length cDNA were washed four times with a solution of 5 OmM EDTA and 2 M NaC1, 0.4% SDS, once with 50 μg ZiU1 yeast tRNA, and OmM NaC1, 0. 2 mM EDTA, 1 OmMT ris—HC 1 (pH 7.5), once with 20% glycerol, once with 50 μg / μ1 yeast tRNA aqueous solution, RNase H buffer (20 mM Tris_HCl ( p H 7. 5), 10 mM Mg C 1 2, 2 OmM KC 1, 0. 1 mM EDTA, washed once with 0. 1 mM dithiothreitol one Honoré (D TT), RNa se H buffer 100 mu Suspend in 1 and add RNase H 3 unit and heat for 30 minutes at 37 ° C. Then, add 10 μl of 10% SDS and 2 μl of 0.5Μ EDTA, and add 10 μl of 65 ° C for 10 minutes. C and the supernatant was collected.

このようにして回収された 1本鎖完全長 c DN Aはフエノール/クロロホルム で抽出され、 スピードバッグにて液量を 100 μ 1以下に減じてから G 25ZG l O O S e p h a d e xクロマトグラフィ一に付した。 R I活性を持った分画は シリコン処理したマイクロチューブに収集するとともに、 ダリコーゲン 2 μ gを 加え、 エタノール沈澱にて得られた沈澱物を 30 μ 1の超純水に溶解した。 (6) 1本鎖 c DNAへのオリゴ dG付加 The single-stranded full-length cDNA recovered in this manner was extracted with phenol / chloroform, and the volume of the solution was reduced to 100 μl or less using a speed bag, and then subjected to G25ZGIOOS ephadex chromatography. Fractions having RI activity were collected in a silicon-treated microtube, 2 μg of dalycogen was added, and the precipitate obtained by ethanol precipitation was dissolved in 30 μl of ultrapure water. (6) Add oligo dG to single-stranded cDNA

上記 (5) で回収された 1本鎖 c DNA3 0 μ 1は、 最終容量 50 μ 1の反応 液中で、 2 0 OmM力コジル酸ナトリウム (ρΗ6· 9)、 1 mM Mg C l 2、 ImM C o C l 2、 1 mM 2—メルカプトエタノール、 l O O ^uM dGTP の条件のもと、 ターミナルデォキシヌクレオチジルトランスフェラーゼ (T a K a R a社製) 3 2 u n i tを用いて 3 7 °Cで 30分間のオリゴ d G付加反応に付 した。 反応終了時に EDTAを 5 OmMとなるように加え、 一連のフエノール/ クロ口ホルムによる抽出、エタノール沈澱を経て、 3 1 μ 1の超純水に溶解した。30 μl of the single-stranded cDNA recovered in the above (5) was used in a reaction solution having a final volume of 50 μl in a reaction solution of 20 OmM sodium codylate (ρΗ6.9), 1 mM MgCl 2 , ImM C o C l 2, 1 mM 2- mercaptoethanol, l OO ^ original uM dGTP conditions, terminal de o carboxymethyl nucleotidyl transferase (T a K a R a, Inc.) 3 3 7 ° with 2 Unit C. The mixture was subjected to an oligo dG addition reaction for 30 minutes. At the end of the reaction, EDTA was added to 5 OmM, and it was dissolved in 31 μl of ultrapure water through a series of extraction with phenol / chloroform and ethanol precipitation.

(7) 第 2鎖 c DNA合成 (7) Second strand cDNA synthesis

第 1鎖 c DN Aを铸型にした第 2鎖 cDN Aの合成は以下のように行った。 最 終容量 60 μ 1の反応系で、 第 2鎖低バッファー (20 OmM T r i s一 HC 1 (pH8. 75)、 1 0 OmM KC 1、 1 00 mM (NH4) 2 S O4、 2 OmM Mg SO4、 1% T r i t o n X— 1 00、 l mg/ 1 B SA) 3 / 1、 第 2鎖高バッファー(20 OmM T r i s -HC l (pH9. 2) 60 OmM K C l、 2 OmM M g C 12) 3 1、 d CTP、 d ATP, d TTP、 d GTP 各々 0. 2 5mM、 β -NADH 6 μ 1、 オリゴ d G付カ卩された第 1鎖 c D N A3 1 μ 1、第 2鎖プライマ一一アダプター(配列番号 34) 600 n gを加え、 E x T a q D N Aポリメラーゼ (T a K a R a E x T a q ; T a K a a社製) 1 5 u n i t、 而熱性 DNAリガーゼ (Amp 1 i g a s e ; E p i c e n t r e社製) 1 50 u n i t、耐熱性 R Na s e H (Hy b r i d a s e ; E p i c e n t r e社製) 3 u n i tによって第 2鎖 c D N Aを合成した。 The synthesis of the second-chain cDNA in which the first-chain cDNA was changed into a 铸 -type was performed as follows. In a reaction system with a final volume of 60 μl, use a second strand low buffer (20 OmM Tris-HC1 (pH 8.75), 10 OmM KC1, 100 mM (NH 4 ) 2 SO 4 , 2 OmM Mg SO 4 , 1% Triton X—100, lmg / 1 BSA) 3/1, 2nd strand high buffer (20 OmM Tris-HCl (pH9.2) 60 OmM KCl, 2 OmM Mg C 1 2 ) 3 1, d CTP, d ATP, d TTP, d GTP each 0.25 mM, β-NADH 6 μ1, first strand cDNA A3 with oligo dG, 1 μ1, Add 600 ng of a two-strand primer-one adapter (SEQ ID NO: 34), add 15 units of ExTaq DNA polymerase (TaKaRaExTaq; manufactured by TaKaa), 15 units, and thermophilic DNA ligase ( Second strand cDNA was synthesized using 150 units of Amp 1 igase (E picentre) and 3 units of heat-resistant R Nase H (Hybridase; E picentre).

0. 5M EDTAを 1 μ 1加えることで反応を停止させ、 更にタンパク成分 を溶解するために、 1 0%SD S 1 μ 1、 プロティナーゼ (P r o t e i n a s e) K 1 0 μ gの存在下に 45°Cで 1 5分間加熱し、 最終的にフエノールノ クロ口ホルムによる抽出、 ェタノール沈澱にて精製した 2本鎖完全長 c D N Aを 得た。  Stop the reaction by adding 1 μl of 0.5 M EDTA, and further dissolve the protein components by 45 ° in the presence of 1 μl of 10% SDS and 10 μg of proteinase K. The mixture was heated at C for 15 minutes, and finally, a double-stranded full-length cDNA was purified by extraction with phenol-noroform form and ethanol precipitation.

(8) ライブラリーの調製 以上の方法により得られた二本鎖完全長 c DN Aは、 ΖΑΡ Ι I Iベクター に挿入し、 ライブラリ一として回収した。 λ ZAP I I Iベクターは λ ZAP I I (STRATAGENE社製) ベクターのマルチクローニングサイトの一部の 配列である配列番号 35を配列番号 36に改変し、 二つの S f i Iサイトを新た に導入したものである。 (8) Library preparation The full-length double-stranded cDNA obtained by the above method was inserted into a ΙII vector and recovered as a library. The λ ZAP III vector is obtained by modifying SEQ ID NO: 35, which is a partial sequence of the multiple cloning site of λ ZAP II (manufactured by STRATAGENE) vector, to SEQ ID NO: 36, and newly introducing two Sfi I sites. .

さらに; L PS (R I KEN) ベクターを作製し、 cDNAを挿入した。 PS Further, LPS (RIKEN) vector was prepared and cDNA was inserted. PS

(R I KEN) (λ— FLC— 1と命名(FLCとは FULL— LENGTH c DNAを意味する)) とは、 Mo B i T e c社(ドイツ) の L P Sベクターを c D N A用に改変したものである。 即ち l O k b p s t u f f e rの両側に存在す るクローニングサイトに c DNA挿入に便利な B a mH Iならびに S a 1 Iを 各々導入するとともに、 0. 5 k bから 1 3 k b程度までの c DNAがクロー二 ングできるように Xb a Iサイトに 6 k bの D N A断片を挿入したものである(RI KEN) (named λ-FLC-1 (FLC stands for FULL-LENGTH cDNA)) is a modified version of the LPS vector from Mo Bitec (Germany) for cDNA. is there. That is, BamHI and Sa1I, which are convenient for cDNA insertion, were introduced into the cloning sites on both sides of lO kbpstuffer, and cDNA from 0.5 kb to about 13 kb was cloned. A 6 kb DNA fragment inserted into the Xba I site

(特開 2000— 325080号公報)。 この λ— FLC— 1を用いると、例えば 肺臓 c DNAライブラリーの場合には、 インサートの平均鎖長は 2. 57 k bと なり、 実際に 0. 5 k bから 12 k bまでのインサートをクロー-ングすること が出来た。 従来法の; L ZAPの場合には、 インサートの平均鎖長は 0. 97 k b であったことから、 L— FLC— 1を用いることによって、 サイズの大きな cD NAも L ZAPに比べて効率よくクローニングできることがわかる。 (Japanese Patent Laid-Open No. 2000-325080). Using this λ-FLC-1, for example, in the case of a lung cDNA library, the average insert length is 2.57 kb, and actually inserts from 0.5 kb to 12 kb are cloned. I was able to do it. In the case of the conventional method; in the case of LZAP, the average length of the insert was 0.97 kb, so the use of L-FLC-1 enables large-sized cDNA to be more efficient than LZAP. It turns out that it can be cloned.

実施例 2 完全長 c DN Aライブラリーのノーマライゼーションノサブトラクシ ョン Example 2 Normalization of the full-length cDNA library No subtraction

(1) ドライバーの調製  (1) Preparation of driver

実施例 1 (1) で作製した mRNA (以下、 これを 「(a) RNAドライバー」 と称することがある)、及び i n v i t r o転写反応で作成した RNAをドライ バーとして用いた。 後者の RNAはさらに 2種類 (以下、 これを 「(b) RNAド ライバー、 及ぴ 「(c) RNAドライバー」 と称する) に分けられる。 1つはノー マライゼーションにより除かれた RNA— c DNAから c DNAを回収し、 ファ ージベクターにクローユングしたものである。 大腸菌に感染後 1つの出発材料あ たり 1000から 2000プラークを混ぜ合わせて 1つのライブラリー (ミニラ イブラリー) とし、 常法によりプラスミド DNAに変換する (ファージをヘルパ 一ファージとともに再度大腸菌に感染させ、 ファージミドとし、 さらにもう一度 感染させてプラスミ ド DNAを得る)。 The mRNA prepared in Example 1 (1) (hereinafter, this may be referred to as “(a) RNA driver”) and the RNA prepared by the in vitro transcription reaction were used as drivers. The latter RNA is further divided into two types (hereinafter referred to as “(b) RNA driver” and “(c) RNA driver”). In the first, cDNA was recovered from RNA-cDNA that had been removed by normalization and cloned into a phage vector. One starting material after infection with E. coli One thousand (2,000) plaques are mixed together to form one library (mini-library), which is converted to plasmid DNA by a conventional method. (The phage is again infected with Escherichia coli together with a helper phage, converted into a phagemid, and further infected again with a plasmid.) Get DNA).

得られた DNAについて i n v i t r o転写反応 (T3RNAポリメラーゼ または T 7RNAポリメラーゼを用いる) を行い、 DNa s e I (RQ1— R Na s e f r e e ; P r ome g a社製)、 P r o t e i n a s e K処理後、 フエノール/クロ口ホルム抽出をして RNA (b) RNAドライバーを得た。 こ の際、 通常出発材料としては 9種類(すい臓、肝臓、肺、 腎臓、脳、脾臓、睾丸、 小腸、 胃) の組織からそれぞれミニライブラリーを作成して、 9種類のミニライ ブラリ一を混合して RNAを得る。 もう一つの RNAはすでに重複のないクロー ンとして保存されているライプラリー (クローン数約 2万個) を培養し、 得られ た DNAについて (b) RNAドライバーと同様に i n v i t r o転写反応を 行い (c) RN Aドライバ一とした。  The obtained DNA was subjected to an in vitro transcription reaction (using T3 RNA polymerase or T7 RNA polymerase), and treated with DNase I (RQ1-R Nasefree; manufactured by Promega) and Proteinase K, followed by phenol / clonal form. After extraction, an RNA (b) RNA driver was obtained. In this case, as a starting material, mini-libraries are created from nine types of tissues (pancreas, liver, lung, kidney, brain, spleen, testes, small intestine, stomach), and mixed with nine types of mini-libraries. To obtain RNA. Another RNA is cultured from a library (about 20,000 clones) that is already stored as a non-overlapping clone, and the resulting DNA is subjected to in vitro transcription reaction in the same manner as the RNA driver (c). RN A driver.

これら 3種の RNAは、 L a b e l _ I T B i o t i n L a b e l i n g K i t (M i r u s C o r p o r a t i o n製) を用いてビォチン化標識を行 つたあと、 1 : 1 : 1の割合でテスター c DNAに添加し、 Ro t 10での反応 These three types of RNA were labeled with biotin using the Label_ITB iotin labeling kit (manufactured by Mirus Corporation), and then added to tester cDNA at a ratio of 1: 1: 1. Reaction at t 10

(42 ) を行い、 ストレプトアビジンビーズ (CPG) 処理を行って回収した 上清について、 第 2鎖の合成を行った。 (42) was performed, and the second strand was synthesized from the supernatant collected by streptavidin bead (CPG) treatment.

実施例 3 完全長 c D N Aクローンの塩基配列決定 Example 3 Nucleotide sequencing of full-length cDNA clone

、1) クロ1―ンの r e a r r a y , 1) rear ray of black 1

各クラスタからひとつの代表クローンを選んだ。代表クローンは Q—b o t (G ENET I X L I MI TED製) で選択し、 384穴プレートに a r r a y化 した。 その際、 大腸菌は 30°Cで 18〜24時間、 50 μ 1の LB培地で培養し た。 このとき、 cDNAライブラリーが PSベクターに導入され大腸菌 DH10 Bを形質転換している場合には 10 Omg/m 1のアンピシリン及ぴ 5 Omg/ m 1のカナマイシンを添加し、 Z a pベクターに導入し、 SOLRシステムに導 入している場合には 10 Omg /m 1のアンピシリン及ぴ 25m /m 1のスト レプトァビジンを添加して行つた。 One representative clone was selected from each cluster. Representative clones were selected using Q-bot (manufactured by G ENET IXLI MITED) and arrayed on a 384-well plate. At that time, E. coli was cultured in 50 µl of LB medium at 30 ° C for 18 to 24 hours. At this time, if the cDNA library has been introduced into the PS vector and Escherichia coli DH10B has been transformed, add 10 Omg / ml of ampicillin and 5 Omg / ml of kanamycin and introduce it into the Zap vector. Led to SOLR system If so, 10 Omg / ml of ampicillin and 25 m / ml of streptavidin were added.

(2) プラスミドの抽出と I n s S i z i n g  (2) Extraction of plasmid and InsSizinng

上記 (1) で培養した各クローンは、 さらに 10 OmgZm 1のアンピシリン を含む 1. 3m 1の HT液中で培養され、 遠心分離により菌体を回収した後、 Q Each clone cultured in (1) above is further cultured in 1.3 ml HT solution containing 10 OmgZm1 ampicillin, and after collecting cells by centrifugation, Q

I Ap r e p 96 Tu r b o (Q I AG E N社製) を用いてプラスミド D N Aを回収、 精製した。 取得されたプラスミ ド中に挿入されている cDNAの鎖長 を調べるために、 上記で取得したプラスミ ド DNAの 1Z30を制限酵素 P V uPlasmid DNA was recovered and purified using I-Aperp 96 Turbo (manufactured by QIAGEN). In order to check the chain length of the cDNA inserted in the obtained plasmid, 1Z30 of the plasmid DNA obtained above was

I Iで消化し、 1%の a g a r o s eゲル電気泳動を行った。 After digestion with II, 1% agarose gel electrophoresis was performed.

(3) 配列決定  (3) Sequence determination

かくして取得されたプラスミド中に挿入された完全長 c DNAの全長の塩基配 列解析には、 3種類のシークェンサを用いた。 また、 プラスミドは挿入配列の長 さが 2.5 k bより短いものと長いものの 2つのカテゴリに分けた。このうち 2. 5 k bより短い挿入配列を有するクローンについては両端から塩基配列を解析し た。 その際、 プラスミドはベクターが PSの場合には配列番号 37 (センス鎖)、 及ぴ 38 (アンチセンス鎖) に記載のプライマーを用いて、 またベクターが Z a pの場合には配列番号 39 (センス鎖)、及ぴ 40 (アンチセンス鎖) に記載のプ フ マーを用レヽて Ί h e rmo s e q u e n a s e P r i me r Cy c l e S e q u e n c i n g K i t 、Am e r s h am Ph a rma c i a B i o t e c h社製) で反応し、 L i c o r DNA 4200 ( l o n g r e a d s e q u e n c e r) を用いて解析した。  Three types of sequencers were used for full-length nucleotide sequence analysis of the full-length cDNA inserted into the plasmid thus obtained. Plasmids were divided into two categories: those with insert sequences shorter than 2.5 kb and those with longer insert sequences. Of these clones, the clone having an insertion sequence shorter than 2.5 kb was analyzed for the nucleotide sequence from both ends. At this time, the plasmid was prepared using the primers described in SEQ ID NO: 37 (sense strand) when the vector was PS, and the primers described in SEQ ID NO: 39 (sense strand) when the vector was Zap. Using the primers described in Sections 40 and 40 (antisense strand) (Hermo Sequenase Primer Cycling Sequencing Kit, manufactured by Amersham Pharmacia Biotech). And analyzed using Licor DNA 4200 (longreadsequencer).

上記塩基配列解析により解析ができなかったギャップは、 プライマウォーキン グ法により決定した。その際、 AB I P r i s m377及ぴ Zまたは AB I P r i sm3700 (A p l i e d B i o s y s t erns I n c. 製) と B i gDy e t e rm i n a t o r k i tと Cy c l e S e qu e n c i n g F S r e a d y Re a c t i on K i t (Ap p l i e d B i o s y s t erns I n c. 製) を用レヽた。 また、 挿入されている cDNAが 2. 5 k bより長いクローンの配列決定は、 ショットガン法によった。 その際、 Sh i ma d z u R I SA 384と DY E n a m 1 c E T t e rm i n a t o r c y c l e s e qu e n c i n g k i t (Am e r s h am Ph a rma c i a B i o t e c h社製) を 用いた。 ショットガンライブラリを作製するために、 48の独立な代表クローン から PC Rで増殖した 48の DN Aフラグメントを用いた。 増幅された DN A断 片の末端を T 4 DN Aポリメラーゼによって平滑化した。 Gaps that could not be analyzed by the above nucleotide sequence analysis were determined by the primer walking method. At that time, AB IPris m377 and Z or AB IPri sm3700 (manufactured by Applied Biosystems Inc.), Big Dy ete rm inatorkit, and Cyclic Sequencing FS ready Reacti on Kit (Ap plied B iosysterns Inc.). In addition, sequencing of clones with inserted cDNAs longer than 2.5 kb was performed by the shotgun method. At that time, Shimadzu RISA384 and DYEnam1cEtterminatorcyclesequencingkit (manufactured by Amersham Pharma Pharmacia Biotech) were used. To generate a shotgun library, 48 DNA fragments grown in PCR from 48 independent representative clones were used. The ends of the amplified DNA fragments were blunt-ended with T4 DNA polymerase.

この DNA断片を、 pUC 18ベクターへ挿入し、 更に該組み換えベクターに より大腸菌 DH10Bを形質転換した。 この大腸菌から上記 (2) と同様にして プラスミドを調製した。  This DNA fragment was inserted into a pUC18 vector, and Escherichia coli DH10B was transformed with the recombinant vector. A plasmid was prepared from this E. coli in the same manner as in (2) above.

それらの代表クローンについては、 両末端からの塩基配列解析によつて塩基配 列を決定し、 該塩基配列をコンピューター上で連結した後、 Do u b l e S t r o k e S h e a r i n g D e v i c e (F i o r e I n c. 製) による s h e a r i n gを行った。 ショットガン法による塩基配列決定は、 12〜1 5 クローンの重複をもって行った。 この塩基配列決定により配列が決定できなかつ たギャップは、 上記と同様にプライマウォーキングによって決定した。  For those representative clones, the nucleotide sequence was determined by nucleotide sequence analysis from both ends, and the nucleotide sequences were ligated on a computer, followed by Double Stroke S hearing Device (Fiore Inc. Was performed. Nucleotide sequencing by the shotgun method was performed with duplication of 12 to 15 clones. The gap whose sequence could not be determined by the nucleotide sequence determination was determined by primer walking in the same manner as described above.

実施例 4 塩基配列の解析 Example 4 Analysis of base sequence

(1) d n a f o rm34147 (配列番号 1、 17)  (1) d n a f o rm34147 (SEQ ID NO: 1, 17)

dn a f o rm34147は、 配列番号 1に示すように、 2386塩基から成 り、 そのうち塩基番号 107から 1858までがオープンリーディングフレーム dn a fo rm34147 is composed of 2386 bases, as shown in SEQ ID NO: 1, of which base numbers 107 to 1858 have an open reading frame.

(終止コドンを含む) になっていた。 オープンリーディングフレームから予測さ れるァミノ酸配列は、 583ァミノ酸残基から成る (配列番号 17)。配列番号 1 がコードするアミノ酸配列について B LAS Tを用いて相同性検索を行ったとこ ろ、 S PTR蛋白質データベース (SWI S S— PROT蛋白質配列データべ一 スと T r EMB L核酸翻訳データベースを統合したもの)中に、 ( i)データべ一 ス登録記号 Mu s mu s c 1 u s AF 346501, G r o t e i n c o up l e d r e c e p t o r a f f e c t i n g t e s t i c u l a r d e s c e n t (Gr e a t) 力 e— v a l u e : 0. 0で 579ァミノ 酸残基に苴り 99%の一致度でヒットした。 (Including the stop codon). The amino acid sequence predicted from the open reading frame consists of 583 amino acid residues (SEQ ID NO: 17). When a homology search was performed using the BLAST for the amino acid sequence encoded by SEQ ID NO: 1, the SPTR protein database (SWI SS-PROT protein sequence database and the TrEMBL nucleic acid translation database were integrated). (I) Database registration symbol Mus musc 1 us AF 346501, G roteinco up ledreceptoraffectingt esticula rdescent (Great) force e— value: 0.0 hit with 579 amino acid residue with 99% coincidence.

また、 配列番号 1に示す塩基配列がコードするアミノ酸配列について、 HMM PF AMによる蛋白質特徴検索を行ったところ配列番号 1の塩基番号 1 82— 3 01がコードするァミノ酸酉己歹 [Jに L ow— d e n s i t y 1 i p o p r o t e i n r e c e p t o r d oma i n c l a s s Aの特徴を示す酉己歹 (J (P f amに 「 1 d 1— r e c e p t— a」 としてェントリーされる塩基配列) を見出 し、 塩基番号 467— 1 18 6がコードするアミノ酸配列に 10力所にわたって L e u c i n e R i c h Re e a tの特徴を示す配列 (P f a mに 「LR R」 としてエントリーされる塩基配列) を見い出した。  In addition, the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 1 was searched for protein characteristics using HMM PFAM. As a result, the amino acid sequence encoded by nucleotide numbers 182—301 in SEQ ID NO: 1 was encoded by amino acid sequence [J ow—density 1 ipoproteinreceptordoma inclass A (J (base sequence sequenced as “1 d1—recept—a” in P f am)) was identified. In 10 amino acid sequences encoded by, a sequence exhibiting the characteristics of Leucine Rich Reeat (base sequence entered as “LR R” in P fam) was found.

上記の G r e a t は、 データベース中の文献情報 (Genesis 2001 May;30(1) :26-35) 力 ら、精巣の腹腔から陰嚢への下降に関与し、 この遺伝子に異 常があると停留睾丸になることが判明している。 これらのことから配列番号 1に 示す塩基配列がコードするタンパク質は L e u c i n e R i c h R e p e a tを含む Gタンパク質共役型受容体であることが推測され、 また、 精巣の発達や 生殖機能を制御する機能を有することが推測された。  The above-mentioned Great is involved in the descent of the testis from the abdominal cavity to the scrotum based on the literature information in the database (Genesis 2001 May; 30 (1): 26-35), and stops when this gene is abnormal. It has been found to be testicular. From these facts, it is inferred that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 1 is a G protein-coupled receptor including Leucine Rich Repeat, and also has a function to control testis development and reproductive function. Was presumed to have.

(2) d n a f o rm37308 (配列番号 2、 18)  (2) d n a f o rm37308 (SEQ ID NOs: 2, 18)

d n a f o rm37308は、 配列番号 2に示すように、 2558塩基から成 り、 そのうち塩基番号 507から 1 874までがオープンリーディングフレーム As shown in SEQ ID NO: 2, dnaform37308 is composed of 2558 bases, of which base numbers 507 to 1874 are open reading frames.

(終止コドンを含む) になっていた。 オープンリーディングフレームから予測さ れるァミノ酸配列は、 455ァミノ酸残基から成る (配列番号 18)。配列番号 2 がコードするアミノ酸配列について BLASTを用いて相同性検索を行ったとこ ろ、 S PTR蛋白質データベース (SWI S S—PROT蛋白質配列データべ一 スと T r EMB L核酸翻訳データベースを統合したもの)中に、 ( i)データべ一 ス登録記号 AK018327、 p r o t e i n s im i l a r t o PRO BABLE G PROTE I N— COUP LED RECEPTOR GPR 72 PRECURSOR (GLUCOCORT I CO I D- I NDUCED RECEPTOR) 力 e— v a 1 u e : 5 X 1 CT136で、 240アミノ酸残基に 亘り 100%の一致度で、 また (i i) データベース登録記号 Q 9NYM4、 o r p h a n G— p r o t e i n c o u l e d r e c e p t o r R 72が、 e— v a 1 u e : 1 X 1 CT84で、 301アミノ酸残基に亘り 45 %の一 致度で、 さらに ( i i i ) データベース登録記号 AY 029071、 R a t t u s n o r v e g i c u s g l u c o c o r t i c o i d— i n d u c e d r e c e p t o r力 e— v a l u e : 3 X 1 (T84で、 301アミノ酸残基に亘 り 44 %の一致度でヒットした。 (Including the stop codon). The amino acid sequence predicted from the open reading frame consists of 455 amino acid residues (SEQ ID NO: 18). When a homology search was performed using BLAST for the amino acid sequence encoded by SEQ ID NO: 2, the SPTR protein database (integrated SWI SS-PROT protein sequence database and TrEMBL nucleic acid translation database) (I) Database registration code AK018327, proteins im ilarto PRO BABLE G PROTE IN—COUP LED RECEPTOR GPR 72 PRECURSOR (GLUCOCORT I CO I D- I NDUCED RECEPTOR) force e- va 1 ue: 5 in X 1 CT 136, with 100% degree of coincidence over the 240 amino acid residues, also have (ii) a database registration mark Q 9NYM4, orphan G- proteincouledreceptor R 72 , e- va 1 ue: 1 with X 1 CT 84, 45% one致度over 301 amino acid residues, yet (iii) a database registration mark AY 029071, R attusnorvegicusglucoc orticoid- inducedreceptor force e- value: 3 X 1 (T 84 The hit was 44% over 301 amino acid residues.

また、 配列番号 2に示す塩基配列がコ一ドするァミノ酸配列について、 HMM PF AMによる蛋白質特徴検索を行ったところ配列番号 2の塩基番号 840— 1 6 1 3がコードするアミノ酸配列に Gタンパク質共役型受容体の特徴を示す配列 (P f amに 「7 tm— 1」 としてエントリーされる塩基配列) を見出した。  In addition, the amino acid sequence encoding the nucleotide sequence shown in SEQ ID NO: 2 was subjected to protein characteristic search using HMM PFAM. As a result, it was found that the amino acid sequence encoded by nucleotide numbers 840 to 1613 of SEQ ID NO: 2 had a G protein A sequence (a nucleotide sequence entered as “7 tm-1” in P f am) showing the characteristics of the conjugated receptor was found.

上記の GPR72は、 データベース中の文献情報 (Biochim Biophys Acta 2000 Apr 25 ;1491 (1-3) :369 - 75) から、 Y -受容体類似のォーファン GPCRであることが判明 して ヽる。 7こ、 上目 ciの g l u c o c o r t i c o i d— i n du c e d r e c e p t o で は、 データベース中の文献情報 (J Neurosci 2001 Nov 15;21 (22) :9027-35) から、 脳に発現すること、 リガンドがペプチド性の GPCRで あること、 慢性アンフエタミン投与後の断薬時に発現が亢進することが示される ため、 これらのことから、 配列番号 2に示す塩基配列がコードするタンパク質は ダルココルチコィドで誘導される可能性のある Gタンパク質共役型受容体であり、 リガンドはぺプチド性であり、 ス トレスや精神刺激剤に対する神経適応に関与す ることが推測された。  The above-mentioned GPR72 is found to be an orphan GPCR similar to the Y-receptor based on the literature information in the database (Biochim Biophys Acta 2000 Apr 25; 1491 (1-3): 369-75). 7 The glucocorticoid of intestinal ci ci-in du cedrecepto is based on the literature information in the database (J Neurosci 2001 Nov 15; 21 (22): 9027-35). Therefore, it is shown that the expression is enhanced upon withdrawal after chronic amphetamine administration.Therefore, the protein encoded by the nucleotide sequence shown in SEQ ID NO: 2 may be induced by darcocorticoid. It is a G protein-coupled receptor, and its ligand is peptide, and it was speculated that it is involved in neuroadaptation to stress and psychostimulants.

(3) d n a f o rm37953 (配列番号 3、 1 9)  (3) d n a f o rm37953 (SEQ ID NO: 3, 19)

d n a f o rm37953は、 配列番号 3に示すように、 3015塩基から成 り、 そのうち塩基番号 9から 1 169までがオープンリーディングフレーム (終 止コドンを含む) になっていた。 オープンリーディングフレームから予測される アミノ酸配列は、 386アミノ酸残基から成る (配列番号 19)。配列番号 3がコ 一ドするアミノ酸配列について B LASTを用いて相同性検索を行ったところ、 S PTR蛋白質データベース (SWI S S— P ROT蛋白質配列データベースと T r EMB L核酸翻訳データベースを銃合したもの)中に、 ( i )データベース登 録記号 AK018327、 PROBABLE G-PROTE I N-COUPL ED RECEPTOR GPR 72 PRECURSOR (GLUCOCOR T I CO I D- I NDUCED RECEPTORが、 e— v a l u e : 5 X l 0-96で、 i 73アミノ酸残基に亘り 100%の一致度で、 また (i i) データべ ース登録記号 Q 9 NYM4、 o r h a n G— p r o t e i n c o u p l e d r e c e p t o r GPR 72力 e— v a l u e : 2 X 10— 84で、 30 1 アミノ酸残基に亘り 45 °/0の一致度で、 さらに (i i i) データベース登録記号 AiT0290 ί' 1 a t t u s n o r v e g ι c u s g l u c o c o r t i c o i d— i n du c e d r e c e p t o r力 s、 e— v a l u e: 4 X 10 一84で、 301ァミノ酸残基に亘り 44 %の一致度でヒッ トした。 As shown in SEQ ID NO: 3, dnafo rm37953 was composed of 3015 bases, of which base numbers 9 to 1169 were open reading frames (including a stop codon). The amino acid sequence predicted from the open reading frame consists of 386 amino acid residues (SEQ ID NO: 19). SEQ ID NO: 3 When a homology search was performed using BLAST for the amino acid sequence to be read, the S PTR protein database (a combination of the SWI SS—P ROT protein sequence database and the Tr EMB L nucleic acid translation database) contained ( i) database registration symbol AK018327, PROBABLE G-pROTE I N -COUPL ED RECEPTOR GPR 72 PRECURSOR (GLUCOCOR TI CO I D- I NDUCED RECEPTOR is, e- value: at 5 X l 0-96, i 7 3 amino acid residues 10 0% degree of coincidence over the group, also (ii) data base over scan registration mark Q 9 NYM4, orhan G- proteincoupledrecepto r GPR 72 force e- value: with 2 X 10- 84, 30 1 amino acid residue in 45 ° / 0 degree of coincidence over, further (iii) a database registration mark AiT0290 ί '1 attusnorveg ι cusglucocorticoid- in du cedreceptor force s, e- value: with 4 X 10 one 84, over a 301 Amino acid residue 44 Hits with% match.

また、 配列番号 3に示す塩基配列がコ一ドするァミノ酸配列について、 HMM P F AMによる蛋白質特徴検索を行つたところ配列番号 3の塩基番号 1 35— 9 08がコードするアミノ酸配列に Gタンパク質共役型受容体の特徴を示す配列 (P i amに 「7 tm— 1」 としてエントリーされる塩基配列) を見出した。 上記の GPR72は、 データベース中の文献情報 (Biochim Biophys Acta 2000 Apr 25 ;1491(1 - 3) :369- 75) から、 Y -受容体類似のォーファン GPCRであることが判明 してレヽる。 7こ、 上 §dの g l u c o c o r t i c o i d— i n d u c e d r e c e p t o r は、 データベース中の文献情報 (J Neurosci 2001 Nov 15;21 (22) :9027-35) から、 脳に発現すること、 リガンドがペプチド性の GPCRで あること、 慢性アンフエタミン投与後の断薬時に発現が亢進することが示されて いる。 これらのことから配列番号 3に示す塩基配列がコードするタンパク質はグ ルココルチコィドで誘導される可能性のあるォーファン Gタンパク質共役型受容 体であり、 リガンドはぺプチド性であり、 ス トレスや精神刺激剤に対する神経適 応に関与することが推測された。 (4) dn a f o rm42851 (配列番号 4、 20) In addition, a protein characteristic search using the HMM PFAM for the amino acid sequence encoding the nucleotide sequence shown in SEQ ID NO: 3 revealed that the G protein was conjugated to the amino acid sequence encoded by nucleotide numbers 135-908 of SEQ ID NO: 3. A sequence (a nucleotide sequence entered as “7 tm-1” in P i am) showing the characteristics of the type receptor was found. The above-mentioned GPR72 was found to be an orphan GPCR similar to the Y-receptor from literature information in the database (Biochim Biophys Acta 2000 Apr 25; 1491 (1-3): 369-75). 7 The glucocorticoid-induced receptor in §d above is expressed in the brain based on literature information in the database (J Neurosci 2001 Nov 15; 21 (22): 9027-35), and that the ligand is a peptide GPCR. However, it has been shown that the expression is enhanced at the time of drug withdrawal after chronic amphetamine administration. From these facts, the protein encoded by the nucleotide sequence shown in SEQ ID NO: 3 is an orphan G protein-coupled receptor that may be induced by glucocorticoid, the ligand is peptide, and the stress or psychostimulant It was speculated that it might be involved in the neural adaptation to the disease. (4) dn afo rm42851 (SEQ ID NOs: 4, 20)

d n a f o r m42851は、 配列番号 4に示すように、 3138塩基から成 り、 そのうち塩基番号 474から 1841までがオープンリーディングフレーム dnafor m42851 consists of 3138 bases as shown in SEQ ID NO: 4, of which base numbers 474 to 1841 have an open reading frame.

(終止コドンを含む) になっていた。 オープンリーディングフレームから予測さ れるアミノ酸配列は、 455アミノ酸残基から成る (配列番号 20)。配列番号 4 がコードするアミノ酸配列について B LASTを用いて相同性検索を行ったとこ ろ、 S PTR蛋白質データベース (SWI S S— PROT蛋白質配列データべ一 スと T r EMBL核酸翻訳データベースを統合したもの)中に、 ( i)データべ一 ス登録記号 AK018327, p t o t e i n s im i l a r t o PRO BABLE G— PROTE I N— COUP LED RECEPTOR GPR 72 PRECURSOR (GLUCOCORT I CO I D- I NDUCED RECEPTOR) I e- v a 1 u e : 5 X 10一136で、 また 240ァミノ酸残 基に亘り 100%の一致度で、また( i i )データベース登録記号 Q 9 NYM4、 Homo s a p i e n s o r p n a n Ur— p r o t e i n c o u l e d r e c e p t o r GPR 72 (GPR 72) 力 e— v a l u e : 2 X l 0一84で、 301アミノ酸残基に亘り 45%の一致度で、 さらに (i i i) データ ベース登録記号 AY 029071, Ra t t u s n o r v e g i c u s g 1 u c o c o r t i c o i d— i n d u c e d r e c e p t o r力、 e— v a 1 u e : 4 X 1 (Γ84で、 301アミノ酸残基に亘り 44%の一致度でヒットした。 また、 配列番号 4に示す塩基配列がコードするアミノ酸配列について、 HMM P F AMによる蛋白質特徴検索を行つたところ配列番号 4の塩基番号 807— 1 580がコードするアミノ酸配列に Gタンパク質共役型受容体の特徴を示す配列(Including the stop codon). The amino acid sequence predicted from the open reading frame consists of 455 amino acid residues (SEQ ID NO: 20). When a homology search was performed using BLAST for the amino acid sequence encoded by SEQ ID NO: 4, the SPTR protein database (integrated SWI SS-PROT protein sequence database and TrEMBL nucleic acid translation database) Inside, (i) Database registration code AK018327, ptoteins im ilarto PRO BABLE G—PROTE IN—COUP LED RECEPTOR GPR 72 PRECURSOR (GLUCOCORT I CO I D- I N DUCED RECEPTOR) I e-va 1 ue: 5X in 10 one 136, also at 240 Amino acid residues 100% degree of coincidence over the group, also (ii) a database registration mark Q 9 NYM4, Homo sapiensorpnan Ur- proteincouledreceptor GPR 72 (GPR 72) power e- value: 2 X l 0 one 84, 45% degree of coincidence over the 301 amino acid residues, yet (iii) database registration mark AY 029071, Ra ttusnorvegicusg 1 ucocorticoid- inducedreceptor force, e- va 1 ue: in 4 X 1 (gamma 84 And a hit of 44% over 301 amino acid residues. For amino acid sequences encoded by the nucleotide sequence shown in 4, sequences showing characteristics of G protein-coupled receptor protein, characterized retrieval by HMM PF AM to the amino acid sequence nucleotide numbers 807- 1 580 of SEQ ID NO: 4 where having conducted encoded

(P f amに 「7 tm_l」 としてエントリーされる塩基配列) を見出した。 上記の GPR72は、 データベース中の文献情報 (Biochim Biophys Acta 2000 Apr 25;1491 (1-3) :369-75) から、 Y-受容体類似のォーファン GPCRであることが判明 してレヽる。 また、 上記の g l u c o c o r t i c o i d— i n du c e d r e c e p t o r は、 データベース中の文献情報 (J Neurosci 2001 Nov 15;21 (22) :9027-35) から、 脳に発現すること、 リガンドがペプチド性の GPCRで あること、 慢性アンフエタミン投与後の断薬時に発現が亢進することが示される ため、 これらのことから配列番号 4に示す塩基配列がコードするタンパク質はグ ルココルチコイドで誘導される可能性のあるォーファン Gタンパク質共役型受容 体であり、 リガンドはぺプチド性であり、 ス トレスや神経刺激剤に対する神経適 応に関わることが推測された。 (Base sequence entered as “7 tm_l” in P f am). The above-mentioned GPR72 is found to be an orphan GPCR similar to the Y-receptor based on the literature information in the database (Biochim Biophys Acta 2000 Apr 25; 1491 (1-3): 369-75). In addition, the glucocorticoid-in du cedreceptor described above is based on literature information in the database (J Neurosci 2001 Nov 15; 21 (22): 9027-35), which indicates that it is expressed in the brain, that the ligand is a peptide GPCR, and that expression is enhanced upon discontinuation of the drug after chronic amphetamine administration. The protein encoded by the nucleotide sequence shown in SEQ ID NO: 4 is an orphan G-protein-coupled receptor that may be induced by glucocorticoids, its ligand is peptide, and it has a neuronal response to stress and neurostimulants. It was presumed to be involved in adaptation.

(5) dn a f o rm27636 (配列番号 5、 21)  (5) dn a f o rm27636 (SEQ ID NOS: 5, 21)

d n a f o rm27636は、 配列番号 5に示すように、 2499塩基から成 り、 そのうち塩基番号 281から 1267までがオープンリーディングフレーム (終止コドンを含む) になっていた。 オープンリーディングフレームから予測さ れるアミノ酸配列は、 328アミノ酸残基から成る (配列番号 21)。配列番号 5 がコードするアミノ酸配列について B LASTを用いて相同性検索を行ったとこ ろ、 S PTR蛋白質データベース (SWI S S— PROT蛋白質配列データべ一 スと T r EML核酸翻訳データベースを統合したもの)中に、 (i)データベース 登録記号 A F41 11 1 1, Homo s a i e n s G p r o t e i n- c o u l e d r e c e p t o r (GPR82) g e n e j、 e— v a 1 u e : 5 X 10"159で、 また 328ァミノ酸残基に亘り 82 %の一致度でヒットした。 また、 配列番号 5に示す塩基配列がコードするァミノ酸配列について、 HMM PF AMによる蛋白質特徴検索を行ったところ配列番号 5の塩基番号 374-1 210がコードするアミノ酸配列に Gタンパク質共役型受容体の特徴を示す配列 (P f amに 「7 tm— 1」 としてエントリーされる塩基配列) を見出した。 上記の、 GPR82のヒトオルソログはデータベース中の文献情報(Gene2001 275(1):83-91) から、 E S Tがヒト結腸組織から得られていることが示されてい る。 これらのことから配列番号 5に示す塩基配列がコードするタンパク質は m o u s eの新規ォーファン Gタンパク質共役型受容体であり、 結腸に発現している ことが推測された。 As shown in SEQ ID NO: 5, dnafo rm27636 was composed of 2499 bases, of which nucleotides 281 to 1267 were an open reading frame (including a stop codon). The amino acid sequence predicted from the open reading frame consists of 328 amino acid residues (SEQ ID NO: 21). A homology search was performed on the amino acid sequence encoded by SEQ ID NO: 5 using BLAST, and a SPTR protein database (integrated SWI SS-PROT protein sequence database and TrEML nucleic acid translation database) Among them, (i) database registration code A F41 11 11 1, Homo saiens G protein-couledreceptor (GPR82) genej, e—va 1 ue: 5 X 10 " 159 , and 82% over 328 amino acid residues The amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 5 was subjected to a protein feature search using HMM PFAM, and the amino acid sequence encoded by nucleotides 374-1210 in SEQ ID NO: 5 was determined. A sequence exhibiting characteristics of a G protein-coupled receptor (a nucleotide sequence entered as “7 tm-1” at P f am) was found. From the literature information (Gene2001 275 (1): 83-91) in the database for the human ortholog of GPR82, it is shown that ESTs are obtained from human colon tissue. From these facts, it was inferred that the protein encoded by the nucleotide sequence of SEQ ID NO: 5 is a novel mouse orphan G protein-coupled receptor and is expressed in the colon.

(6) d n a f o rm47734 (配列番号 6、 22) d n a f o rm47734は、 配列番号 6に示すように、 2146塩基から成 り、 そのうち塩基番号 173から 1456までがオープンリーディングフレーム(6) dnafo rm47734 (SEQ ID NOs: 6, 22) As shown in SEQ ID NO: 6, dnafo rm47734 is composed of 2146 bases, of which base numbers 173 to 1456 are open reading frames.

(終止コドンを含む) になっていた。 オープンリーディングフレームから予測さ れるアミノ酸配列は、 427アミノ酸残基から成る (配列番号 22)。配列番号 6 がコードするァミノ酸配列について BLASTを用いて相同性検索を行つたとこ ろ、 S PTR蛋白質データベース (SWI S S— PROT蛋白質配列データべ一 スと T r EMB L核酸翻訳データベースを統合したもの)中に、 ( i)データべ一 ス登録記号 AX 37657 5、 h uma n TGR 21 G— p r o t e i n c o u p l e d r e c e p t o r (GPCR) 力 e— v a l u e : 5 X 10 一176で、 また 442ァミノ酸残基に亘り 73 %の一致度でヒットした。 (Including the stop codon). The amino acid sequence predicted from the open reading frame consists of 427 amino acid residues (SEQ ID NO: 22). When a homology search was performed on the amino acid sequence encoded by SEQ ID NO: 6 using BLAST, the SPTR protein database (SWI SS—PROT protein sequence database integrated with the TrEMBL nucleic acid translation database) ), (I) Database registration code AX 37657 5, human TGR 21 G—protein coupled receptor (GPCR) force e—value: 5 × 10 1 176 , and 442 amino acid residues over 73 Hit with% match.

また、 配列番号 6に示す塩基配列がコードするアミノ酸配列について、 HMM PF AMによる蛋白質特徴検索を行ったところ配列番号 6の塩基番号 368- 1 207がコードするアミノ酸配列に Gタンパク質共役型受容体の特徴を示す配列 In addition, the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 6 was subjected to protein characteristic search using HMM PFAM, and the amino acid sequence encoded by nucleotide numbers 368-1 207 in SEQ ID NO: 6 was found to have a G protein-coupled receptor. An array indicating the features

(P f amに 「7 tm— 1」 としてエントリーされる塩基配列) を見出した。 これらのことから配列番号 6に示す塩基配列がコードするタンパク質はヒト T GR21と類似の Gタンパク質共役型受容体活性を有するであることが推測され た。 (Base sequence entered as “7 tm—1” in P f am). From these facts, it was presumed that the protein encoded by the nucleotide sequence of SEQ ID NO: 6 had a G protein-coupled receptor activity similar to that of human TGR21.

(7) d n a f o rm29894 (配列番号 7、 23)  (7) d n a f o rm29894 (SEQ ID NOs: 7, 23)

d n a f o rm29894は、 配列番号 7に示すように、 1 575塩基から成 り、そのうち塩基番号 53から 1354までがオープンリーディングフレーム(終 止コドンを含む) になっていた。 オープンリーディングフレームから予測される アミノ酸配列は、 433アミノ酸残基から成る (配列番号 23)。配列番号 7がコ ードするアミノ酸配列について B L A S Tを用いて相同性検索を行ったところ、 S PTR蛋白質データベース (SWI S S— P ROT蛋白質配列データベースと T r EMB L核酸翻訳データベースを統合したもの)中に、 ( i )データベース登 録 BG"^ A F41 11 l 7、 Homo s a i e n s G p r o t e i n— c o u p 1 e d r e c e p t o r (GPR 103) 力 S、 e— v a l u e : 0. 0 で、 また 432ァミノ酸残基に亘り 74 %の一致度でヒットした。 As shown in SEQ ID NO: 7, dnafo rm29894 was composed of 1575 bases, of which nucleotides 53 to 1354 had an open reading frame (including a stop codon). The amino acid sequence predicted from the open reading frame consists of 433 amino acid residues (SEQ ID NO: 23). A homology search was performed using BLAST on the amino acid sequence encoded by SEQ ID NO: 7, and it was found in the SPTR protein database (integrated SWI SS—PROT protein sequence database and TrEMBL nucleic acid translation database). In addition, (i) database registration BG "^ A F41 11 l7, Homo saiens G protein—coup 1 edreceptor (GPR 103) power S, e—value: 0.0 And a hit of 74% over 432 amino acid residues.

また、 配列番号 7に示す塩基配列がコ一ドするァミノ酸配列について、 HMM PF AMによる蛋白質特徴検索を行ったところ配列番号 7の塩基番号 236- 1 048がコードするアミノ酸配列に Gタンパク質共役型受容体の特徴を示す配列 (P f amに 「7 tm— 1」 としてエントリーされる塩基配列) を見出した。 上記のヒ ト G P R 1 0 3はデータベース中の文献情報 (Gene 2001 Sep 5 ;275(1) :83- 91)力 ら、 neuropeptide FF 2, neuropeptide Y2およぴ galanin GalRl 受容体と膜貫通領域の一致度が 34〜 38%であること、 さらにノーザンハイブ リダィゼンシヨン実験から mRNAが大脳皮質、 脳下垂体、 視床、 視床下部、 前 脳基底核、 中脳、 および橋に発現していることが示されている。 これらのことか ら配列番号 7に示す塩基配列がコードするタンパク質は脳で発現しており、 リガ ンドがぺプチド性の Gタンパク質共役型受容体であり、 G P C R活性を有するこ とが推測された。  The amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 7 was subjected to a protein feature search using HMM PFAM. As a result, it was found that the amino acid sequence encoded by nucleotide numbers 236-1048 of SEQ ID NO: 7 had a G protein-conjugated A sequence (a nucleotide sequence entered as “7 tm-1” in P f am) showing the characteristics of the receptor was found. The above-mentioned human GPR103 is based on the literature information in the database (Gene 2001 Sep 5; 275 (1): 83-91) based on the information of neuropeptide FF2, neuropeptide Y2 and galanin GalRl receptor and transmembrane region. The agreement is 34-38%, and Northern Hidden Reduction experiments show that mRNA is expressed in the cerebral cortex, pituitary gland, thalamus, hypothalamus, basal forebrain, midbrain, and pons. ing. From these facts, it was conjectured that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 7 was expressed in the brain, and that the ligand was a peptide G protein-coupled receptor and had GPCR activity. .

(8) dn a f o rm53455 (配列番号 8、 24)  (8) dn a f o rm53455 (SEQ ID NOS: 8, 24)

d n a f o rmは、 53455配列番号 8に示すように、 2864塩基から成 り、そのうち塩基番号 64から 1 161までがオープンリーディングフレーム(終 止コドンを含む) になっていた。 オープンリーディングフレームから予測される ァミノ酸配列は、 365ァミノ酸残基から成る (配列番号 24)。配列番号 8がコ 一ドするアミノ酸配列について BLASTを用いて相同性検索を行ったところ、 S PTR蛋白質データベース (SWI S S— PROT蛋白質配列データベースと T r EMB L核酸翻訳データベースを統合したもの)中に、 ( i )データベース登 d号 A F41 1 1 17、 Homo s a p i e n s G : r o t e i n— c o u 1 e d r e c e p t o r (GPR 103) 力 e— v a l u e : 5 X 1 0 -157また 3 77ァミノ酸残基に亘り 73 %の一致度でヒッ トした。 As shown in SEQ ID NO: 8 of 53455, dnafo rm was composed of 2864 bases, of which base numbers 64 to 1161 were open reading frames (including a stop codon). The amino acid sequence predicted from the open reading frame consists of 365 amino acid residues (SEQ ID NO: 24). A homology search was performed for the amino acid sequence encoded by SEQ ID NO: 8 using BLAST, and it was found in the SPTR protein database (integrated SWI SS-PROT protein sequence database and TrEMBL nucleic acid translation database). , (i) No. database Noboru d a F41 1 1 17, Homo sapiens G: rotein- cou 1 edreceptor (GPR 103) power e- value: 5 X 1 0 - 157 the 3 77 Amino acid residues 73% of over Hits were made for the degree of coincidence.

また、 配列番号 8に示す塩基配列がコードするアミノ酸配列について、 HMM PF AMによる蛋白質特徴検索を行ったところ配列番号 8の塩基番号 91— 90 3がコードするアミノ酸配列に Gタンパク質共役型受容体の特徴を示す配列 (P f amに 「7 tm— 1」 としてエントリーされる塩基配列) を見出した。 In addition, the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 8 was searched for protein characteristics using HMM PFAM. As a result, the amino acid sequence encoded by nucleotide numbers 91 to 903 in SEQ ID NO: 8 Characteristic array (P The nucleotide sequence entered as “7 tm—1” at f am.

上記のヒ ト G P R 1 0 3はデータベース中の文献情報 (Gene 2001 Sep 5 ;275(1): 83-91)力 ら、 neuropeptide FF 2, neuropeptide Y2および galanin GalRl 受容体と膜貫通領域の一致度が 34〜 38%であること、 さらにノーザンハイブ リダィゼンシヨン実験から mRNAが大脳皮質、 脳下垂体、 視床、 視床下部、 前 脳基底核、 中脳、 および橋に発現していることが示されている。 これらのことか ら配列番号 8に示す塩基配列がコードするタンパク質は脳で発現しており、 リガ ンドがぺプチド性の Gタンパク質共役型受容体であり、 G P C R活性を有するこ とが推測された。  The above human GPR103 is based on the literature information in the database (Gene 2001 Sep 5; 275 (1): 83-91), and indicates the degree of coincidence between neuropeptide FF2, neuropeptide Y2 and galanin GalRl receptors and the transmembrane domain. Is 34-38%, and Northern Hidden Reduction experiments show that mRNA is expressed in the cerebral cortex, pituitary gland, thalamus, hypothalamus, basal forebrain, midbrain, and pons . From these facts, it was conjectured that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 8 was expressed in the brain, and that the ligand was a peptide G protein-coupled receptor and had GPCR activity. .

(9) d n a f o rm53728 (配列番号 9、 25)  (9) d n a f o rm53728 (SEQ ID NOS: 9, 25)

dn a f o rm53728は、 配列番号 9に示すように、 1642塩基から成 り、そのうち塩基番号 88から 1 185までがオープンリーディングフレーム(終 止コドンを含む) になっていた。 オープンリーディングフレームから予測される ァミノ酸配列は、 365ァミノ酸残基から成る (配列番号 25)。配列番号 9がコ 一ドするアミノ酸配列について BLASTを用いて相同性検索を行ったところ、 S PTR蛋白質データベース (SWI S S_ PRO T蛋白質配列データベースと T r EMB L核酸翻訳データベースを統合したもの)中に、 ( i )データベース登 録 ΰ号 A F41 1 1 1 7、 Homo s a i e n s G p r o t e i n— c o u 1 e d r e c e p t o r (GPR103) 力 e— v a l u e : 5 X 1 0 -16°で、 また 386ァミノ酸残基に亘り 73 %の一致度でヒッ トした。 As shown in SEQ ID NO: 9, dnafo rm53728 was composed of 1642 bases, of which base numbers 88 to 1185 were an open reading frame (including a stop codon). The amino acid sequence predicted from the open reading frame consists of 365 amino acid residues (SEQ ID NO: 25). A homology search was performed using BLAST for the amino acid sequence encoded by SEQ ID NO: 9, and it was found in the SPTR protein database (integrated SWI S_PROT protein sequence database and TrEMBL nucleic acid translation database). to, (i) a database registration No. ΰ a F41 1 1 1 7, Homo saiens G protein- cou 1 edreceptor (GPR103) power e- value: 5 X 1 0 - over at 16 °, also 386 Amino acid residues Hits with 73% match.

また、 配列番号 9示す塩基配列がコードするアミノ酸配列について、 HMMP F AMによる蛋白質特徴検索を行ったところ配列番号 9塩基番号 1 15— 927 がコードするアミノ酸配列に Gタンパク質共役型受容体の特徴を示す配列 (P f amに 「7 tm— 1」 としてエントリーされる塩基配列) を見出した。  The amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 9 was searched for protein characteristics using HMMP FAM. As a result, it was found that the amino acid sequence encoded by SEQ ID NO: 9 nucleotide number 115-927 showed the characteristics of G protein-coupled receptor. The sequence shown (base sequence entered as “7 tm-1” in P f am) was found.

上記のヒ ト G P R 1 0 3はデータベース中の文献情報 (Gene 2001 Sep 5 ;275(1) :83 91)力、ら、 neuropeptide FF 2, neuropeptide Y2およぴ galanin GalRl 受容体と膜貫通領域の一致度が 34〜 38%であること、 さらにノーザン ヨン実験から niRNAが大脳皮質、 脳下垂体、 視床、 視床下部、 前 脳基底核、 中脳、 および橋に発現していることが示されている。 これらのことか ら配列番号 9に示す塩基配列がコードするタンパク質は脳で発現しており、 リガ ンドがぺプチド性の Gタンパク質共役型受容体であり、 G P C R活性を有するこ とが推測された。 The above-mentioned human GPR103 is based on the literature information in the database (Gene 2001 Sep 5; 275 (1): 8391), such as force, et al., Neuropeptide FF2, neuropeptide Y2 and galanin GalRl receptor and 34-38% match, plus Northern Yong experiments show that niRNA is expressed in the cerebral cortex, pituitary gland, thalamus, hypothalamus, basal forebrain, midbrain, and pons. From these facts, it was speculated that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 9 was expressed in the brain, and that the ligand was a peptide-type G protein-coupled receptor and had GPCR activity. .

(10) d n a f o rm62766 (配列番号 10、 26)  (10) d n a f o rm62766 (SEQ ID NO: 10, 26)

dn a f o rm62766は、 配列番号 10に示すように、 1698塩基から 成り、 そのうち塩基番号 160から 1236までがオープンリーディングフレー ム (終止コドンを含む) になっていた。 オープンリーディングフレームから予測 されるァミノ酸配列は、 358ァミノ酸残基から成る (配列番号 26)。配列番号 10がコードするアミノ酸配列について BLASTを用いて相同性検索を行った ところ、 S PTR蛋白質データベース (SWI S S— PROT蛋白質配列データ ベースと T r EMB L核酸翻訳データベースを統合したもの)中に、 ( i )データ ベース登録記号 A F 41 1 1 17、 Homo s a i e n s G p r o t e i n— c o u 1 e d r e c e p t o r (GPR 103)力 S、 e— v a 1 u e : 5 X 10"172で、 また 409ァミノ酸残基に亘り 73 %の一致度でヒットした。 また、 配列番号 10に示す塩基配列がコードするアミノ酸配列について、 HM MP F AMによる蛋白質特徴検索を行ったところ配列番号 10の塩基番号 1 18 一 930がコードするアミノ酸配列に Gタンパク質共役型受容体の特徴を示す配 列 (P f amに 「7 tm— 1」 としてエントリーされる塩基配列) を見出した。 上記のヒ ト G P R 1 0 3はデータベース中の文献情報 (Gene 2001 Sep 5 ;275(1) :83 - 91)力 ら、 neuropeptide FF 2, neuropeptide Y2およぴ galanin GalRl 受容体と膜貫通領域の一致度が 34〜38%であること、 さらにノーザンハイブ リダィゼンシヨン実験から mRNAが大脳皮質、 脳下垂体、 視床、 視床下部、 前 脳基底核、 中脳、 および橋に発現していることが示されている。 これらのことか ら配列番号 1 0に示す塩基配列がコードするタンパク質は脳で発現しており、 リ ガンドがぺプチド性の Gタンパク質共役型受容体であり、 G P C R活性を有する ことが推測された。 As shown in SEQ ID NO: 10, dnafo rm62766 was composed of 1698 bases, of which base numbers 160 to 1236 were open reading frames (including a stop codon). The amino acid sequence predicted from the open reading frame consists of 358 amino acid residues (SEQ ID NO: 26). A homology search was performed using BLAST for the amino acid sequence encoded by SEQ ID NO: 10, and it was found in the SPTR protein database (integrated SWI SS-PROT protein sequence database and TrEMBL nucleic acid translation database). (I) Database registration code AF 41 1 117, Homo saiens G protein—cou 1 edreceptor (GPR 103) force S, e—va 1 ue: 5 × 10 ” 172 and over 409 amino acid residues 73 The amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 10 was subjected to a protein feature search using HMMP FAM, and as a result, the amino acid sequence encoded by nucleotide numbers 118 to 930 in SEQ ID NO: 10 was found. A sequence (a nucleotide sequence entered as “7 tm-1” in P f am) was found that exhibits the characteristics of a G protein-coupled receptor in the sequence. The above-mentioned human GPR103 is based on the literature information in the database (Gene 2001 Sep 5; 275 (1): 83-91), which indicates that neuropeptide FF2, neuropeptide Y2 and galanin GalRl receptor and transmembrane region The agreement is 34-38%, and the Northern Hidden Reduction experiments show that mRNA is expressed in the cerebral cortex, pituitary gland, thalamus, hypothalamus, basal forebrain, midbrain, and pons. ing. Therefore, the protein encoded by the nucleotide sequence of SEQ ID NO: 10 is expressed in the brain, and the ligand is a peptide G protein-coupled receptor and has GPCR activity. That was speculated.

(11) d n a f o rm32567 (配列番号 1 1、 27)  (11) d n a f o rm32567 (SEQ ID NOS: 11, 27)

dn a f o rni325 & 7は、 配列番号 1 1に示すように、 3792塩基から 成り、 そのうち塩基番号 92から 1 207までがオープンリーディングフレーム (終止コドンを含む) になっていた。 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は、 371アミノ酸残基から成る (配列番号 27)。配列番号 1 1がコードするアミノ酸配列について BLASTを用いて相同性検索を行った ところ、 S PTR蛋白質データベース (SWI S S— PROT蛋白質配列デー タベースと T r EMB L核酸翻訳データベースを統合したもの) 中に、 ( i)デー タベース登録記号 AX 339740、 a n o v e l huma n g r o t e i n c o u l e d r e c e p t o r力 S、 e— v a l u e : 5 X 10 ~179 で、 また 348ァミノ酸残基に亘り 87 %の一致度でヒッ トした。 As shown in SEQ ID NO: 11, dn afo rni325 & 7 consisted of 3792 bases, of which base numbers 92 to 1207 were open reading frames (including a stop codon). The amino acid sequence predicted from the open reading frame consists of 371 amino acid residues (SEQ ID NO: 27). A homology search was performed using BLAST on the amino acid sequence encoded by SEQ ID NO: 11, and it was found in the SPTR protein database (integrated SWI SS-PROT protein sequence database and TrEMBL nucleic acid translation database). (I) The database registration symbol AX 339740, anovel huma ngroteincouled receptor force S, e—value: 5 X 10 to 179 , and 87% of the hits over 348 amino acid residues were hit.

また、 配列番号 1 1に示す塩基配列がコードするアミノ酸配列について、 HM MP F AMによる蛋白質特徴検索を行ったところ配列番号 11の塩基番号 287 — 108 1がコードするアミノ酸配列に Gタンパク質共役型受容体の特徴を示す 配列 (P f amに 「7 tm— 1」 としてエントリーされる塩基配列) を見出した。 これらのことから配列番号 1 1に示す塩基配列がコードするタンパク質は Gタ ンパク質共役型受容体活性を有することが推測された。  The amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 11 was subjected to a protein feature search using HMMP FAM. As a result, it was found that the amino acid sequence encoded by nucleotide numbers 287 to 1081 in SEQ ID NO: 11 had G protein-coupled A sequence (base sequence entered as “7 tm-1” in P f am) showing body characteristics was found. From these facts, it was inferred that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 11 has G protein-coupled receptor activity.

(12) d n a f o rm33729 (配列番号 1 2、 28)  (12) d n a f o rm33729 (SEQ ID NOS: 12, 28)

dn a f o rm33729は、 配列番号 1 2【こ示すように、 3336塩基から 成り、 そのうち塩基番号 66 1から 2196までがオープンリーディングフレー ム (終止コドンを含む) になっていた。 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は、 51 1アミノ酸残基から成る (配列番号 28)。配列番号 1 2がコードするアミノ酸配列について B LASTを用いて相同性検索を行った ところ、 S PTR蛋白質データベース (SWI S S— PROT蛋白質配列データ ベースと Tr EMBL核酸翻訳データベースを統合したもの) 中に、 (i)データ ベース登録記号 AX 375432 g p c r x l l a s a f u n c t i o n a 1 r e c e p t o r v a l i d a t e d b y a n g i o p e p t i n a n d u s e f u l f o r s c r e e n i n g o f a g o n i s t s a n d a n t a g o n i s t s力 s、 e— v a l u e : 0. 0で、 'また 5 0 7ァミノ酸残基に亘り 7 2 %の一致度でヒットした。 dn afo rm33729 consists of 3336 bases as shown in SEQ ID NO: 12, of which base numbers 661 to 2196 are open reading frames (including a stop codon). The amino acid sequence predicted from the open reading frame consists of 511 amino acid residues (SEQ ID NO: 28). When a homology search was performed using BLAST for the amino acid sequence encoded by SEQ ID NO: 12, the SPTR protein database (integrating the SWI SS-PROT protein sequence database and TrEMBL nucleic acid translation database) (I) Database registration code AX 375432 gpcrxllasafunctio na 1 receptorvalidatedbyan giopeptinandusefulfor screeningofagonistsand dantagonists power s , e-value: 0.0, 'and also hit 507 amino acid residues with a 72% match.

また、 配列番号 1 2に示す塩基配列がコードするアミノ酸配列について、 HM MP F AMによる蛋白質特徴検索を行ったところ配列番号 1 2の塩基番号 7 96 - 1 548がコードするアミノ酸配列に Gタンパク質共役型受容体の特徴を示す 配列 (P f amに 「7 tm— 1」 としてエントリーされる塩基配列) を見出した。 上記の、 AX 3 7 5 4 3 2はデータベース中の特許情報 (W0 0198330- A 9 27 - DEC - 2001) 力、ら、 ヒト GPCRであり、 angiopeptinで活性化されることが示 されている。 また、 angiopeptinはデータベース中の文献情報 (Atherosclerosis 1989 Aug ;78 (2-3) :229 - 36) から、 ソマトスタチンのアナログであり、 ゥサギ冠状 動脈のァテローム性動脈硬化を抑制することが示されている。 これらのことから 配列番号 1 2に示す塩基配列がコードするタンパク質はソマトスタチン類似のリ ガンドに対する Gタンパク質共役型受容体であり、 GPCR活性を有し、 動脈硬 化や癌に関連することが推測された。  In addition, the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 12 was subjected to a protein feature search using HMMP FAM, and the G protein was conjugated to the amino acid sequence encoded by nucleotide numbers 796 to 1548 in SEQ ID NO: 12. A sequence (a nucleotide sequence entered as “7 tm-1” in P f am) showing the characteristics of the type receptor was found. AX3755332 described above is a human GPCR, which is a patent information (W0198330-A927-DEC-2001) in the database, and has been shown to be activated by angiopeptin. Angiopeptin is an analog of somatostatin and is shown to inhibit atherosclerosis in the coronary artery of the egret, based on literature information (Atherosclerosis 1989 Aug; 78 (2-3): 229-36) in the database. . From these facts, it is inferred that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 12 is a G protein-coupled receptor for somatostatin-like ligand, has GPCR activity, and is associated with arterial sclerosis and cancer. Was.

(1 3) d n a f o r m4847 7 (配列番号 1 3、 2 9)  (1 3) d n a f o r m 4847 7 (SEQ ID NOS: 13, 29)

d n a f o r m4847 7は、 配列番号 1 3に示すように、 3 3 79塩基から 成り、 そのうち塩基番号 1 08から 1 1 84までがオープンリーディングフレー ム (終止コドンを含む) になっていた。 オープンリーディングフレームから予測 されるァミノ酸配列は、 3 5 8ァミノ酸残基から成る (配列番号 29)。配列番号 1 3がコードするアミノ酸配列について BLASTを用いて相同性検索を行った ところ、 S PTR蛋白質データベース (SWI S S—PROT蛋白質配列データ ベースと T r EMB L核酸翻訳データベースを統合したもの) 中に、 ( i )データ ベ—ス登録記号 AB 0 38 2 3 7、 Homo s a i e n s mRNA f o r G p r o t e i n— c o u p l e d r e c e p t o r C 5 L 2 ^ P R 7 7力 e— v a 1 u e : 5 X 1 0一114で、また 3 3 1ァミノ酸残基に亘り 6 1 % の一致度で、 (i i) AF 068680、 O r y c t o l a g u s c u n i c u 1 u s a n a p hy l a t o x i n C 5 a r e c e p t o r g e n e力 s、 e— v a l u e : 5 X 10—6°で、 また 329アミノ酸残基に亘り 44%の一致度 でヒッ トした。 As shown in SEQ ID NO: 13, dnaform48477 was composed of 3379 bases, of which base numbers from 108 to 1184 were open reading frames (including a stop codon). The amino acid sequence predicted from the open reading frame consists of 358 amino acid residues (SEQ ID NO: 29). A homology search was performed on the amino acid sequence encoded by SEQ ID NO: 13 using BLAST, and it was found in the SPTR protein database (integrated SWI SS-PROT protein sequence database and TrEMBL nucleic acid translation database). , (I) Database registration code AB 0 382 3 37, Homo saiens mRNA for G protein— coupled receptor C 5 L 2 ^ PR 77, e— va 1 ue: 5 × 10 1 114 , and 3 3 1 1% over 1 amino acid residue In the matching degree, (ii) AF 068680, O ryctolaguscunicu 1 usanap hy latoxin C 5 areceptorgene force s, e- value: hit at 5 X 10- 6 °, and in 44% of the degree of coincidence over the 329 amino acid residues did.

また、 配列番号 13に示す塩基配列がコードするァミノ酸配列について、 HM MP F AMによる蛋白質特徴検索を行ったところ配列番号 13の塩基番号 321 一 1040がコードするアミノ酸配列に Gタンパク質共役型受容体の特徴を示す 配列 (P f amに 「7 tm— 1」 としてエントリーされる塩基配列) を見出した。  In addition, the amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 13 was subjected to a protein feature search using HMMP FAM. (A nucleotide sequence entered as "7 tm-1" in P f am) was found.

上記の C 5 L 2はデータベース中の文献情幸 (Mol Immunol 2000 Jun; 37 (8):407-12) から、 顆粒球や未分ィヒな樹状細胞で発現しており、 C 3 aや C 5 a走化性因子受容体とホモロジ一が高いことが示されている。 これらのこと から配列番号 13に示す塩基配列がコードするタンパク質は走化性にかかわる G タンパク質共役型受容体活性を有することが推測された。  The above-mentioned C5L2 is expressed in granulocytes and undivided dendritic cells from literature information in the database (Mol Immunol 2000 Jun; 37 (8): 407-12), and C3a And homology with C5a chemoattractant receptor have been shown to be high. From these facts, it was presumed that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 13 had a G protein-coupled receptor activity related to chemotaxis.

(14) d n a f o rm54572 (配列番号 14、 30)  (14) d n a f o rm54572 (SEQ ID NOs: 14, 30)

d n a f o rm54572は、 配列番号 14に示すように、 3005塩基から 成り、 そのうち塩基番号 21 8から 970までがオープンリ一ディングフレーム As shown in SEQ ID NO: 14, dnaform54572 consists of 3005 bases, of which base numbers 218 to 970 have an open reading frame.

(終止コドンを含む) になっていた。 オープンリーディングフレームから予測さ れるアミノ酸配列は、 250アミノ酸残基から成る (配列番号 30)。配列番号 1 4がコードするアミノ酸配列について B LASTを用いて相同性検索を行ったと ころ、 S PTR蛋白質データベース (SWI S S—PROT蛋白質配列データべ ースと T r EMBL核酸翻訳データベースを統合したもの) 中に、 ( i )データ ベース登録記号 A F 1 1 1088、 B o s t a u r u s 1 a t r o p h i 1 i n 3 s p l i c e v a r i a n t a b b f 、 e— v a l u e : 3 X 10— 51で、 また 302ァミノ酸残基に亘り 37 %の一致度でヒッ .トした。 (Including the stop codon). The amino acid sequence predicted from the open reading frame consists of 250 amino acid residues (SEQ ID NO: 30). A homology search was performed on the amino acid sequence encoded by SEQ ID NO: 14 using BLAST, and a SPTR protein database (integrated SWI SS-PROT protein sequence database and TrEMBL nucleic acid translation database) during, (i) data base registration mark AF 1 1 1088, B ostaurus 1 atrophi 1 in 3 splicevariantabbf, e- value: hit with 3 X 10- 51, also 302 Amino acids 37% degree of coincidence over the residues .

また、 配列番号 14に示す塩基配列がコードするアミノ酸配列について、 HM MPF AMによる蛋白質特徴検索を行つたところ配列番号 14の塩基番号 32— 784がコードするアミノ酸配列に Gタンパク質共役型受容体の特徴を示す配列 (P f amに 「7 tm— 2」 としてエントリーされる塩基配列) を見出した。 上記の latrophilin 3 splice variant abbf はデータベース中の文献情報 (FEBS Lett 1999 Jan 29 ;443(3): 348-52) から、 Latrophilin は強力な前シナプス毒で ある alpha- latrotoxinの受容体であり、多数のスプライシングバリアントを有す るが、 Latrophilin- 3 は脳特異的に発現することが示されている。 これらのこと から配列番号 14に示す塩基配列がコードするタンパク質はォーファン Gタンパ ク質共役型受容体であり、 脳などで活性を有することが推測された。 The amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 14 was searched for protein characteristics using HM MPF AM. Array indicating (Base sequence entered as “7 tm—2” in P f am). From the literature information in the database (FEBS Lett 1999 Jan 29; 443 (3): 348-52), latrophilin 3 splice variant abbf indicates that Latrophilin is a receptor for alpha-latrotoxin, a potent prosynaptic toxin. Latrophilin-3 has been shown to be specifically expressed in the brain. From these facts, it was inferred that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 14 is an orphan G protein-coupled receptor and has activity in the brain and the like.

(15) d n a f o rm29069 (配列番号 15、 31)  (15) d n a f o rm29069 (SEQ ID NOs: 15, 31)

d n a f o rm29069は、 配列番号 1 5に示すように、 3034塩基から 成り、 そのうち塩基番号 1557から 2489までがオープンリーディングフレ ーム (終止コドンを含む) になっていた。 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は、 310アミノ酸残基から成る (配列番号 31)。配列番 号 1 5がコードするアミノ酸配列について B LASTを用いて相同性検索を行つ たところ、 S PTR蛋白質データベース (SWI S S— PROT蛋白質配列デー タベースと T r EMB L核酸翻訳データベースを統合したもの) 中に、 ( i )デー タベース登録記号 A F 1 66382、 Mu s mu s c u 1 u s s e r e n t i n e r e c e p t o r (Cy t 28)力 e - v a l u e : 5X 10一56で、 また 271アミノ酸残基に亘り 38%の一致度で、 また (i i) データベース登 録記号 B C 008770N Homo s a i e n s, G p r o t e i n— c o u 1 e d r e c e p t o r 56力 e— v a l u e : 8 X 10一51で、 2 81アミノ酸残基に亘り 36%の一致度で、 さらに ( i i i) データベース登録 §己号 A F 106858 Homo s a i e n s G— p r o t e i n— c o u 1 e d r e c e p t o r (GPR56) 力 e_v a l u e : l X 10一50 で、 28 1ァミノ酸残基に亘り 36 %の一致度でヒッ トした。 As shown in SEQ ID NO: 15, dnafo rm29069 consisted of 3034 bases, of which base numbers 1557 to 2489 were open reading frames (including a stop codon). The amino acid sequence predicted from the open reading frame consists of 310 amino acid residues (SEQ ID NO: 31). When a homology search was performed using BLAST for the amino acid sequence encoded by SEQ ID NO: 15, the S PTR protein database (SWI SS-PROT protein sequence database and Tr EMB L nucleic acid translation database were integrated). ) in, (i) database registration mark AF 1 66382, Mu s mu scu 1 usserentinereceptor (Cy t 28) force e - value: in 5X 10 one 56, and in 38% of the degree of coincidence over the 271 amino acid residues and (ii) a database registration symbol BC 008770 N Homo saiens, G protein- cou 1 edreceptor 56 force e- value: at 8 X 10 one 51, 2 81 36% degree of coincidence over the amino acid residues, and ( iii) database registration § himself No. AF 106858 Homo saiens G- protein- cou 1 edreceptor (GPR56) force e_v alue: at l X 10 one 50 was hit with 36% degree of coincidence over 28 1 Amino acid residues.

また、 配列番号 1 5に示す塩基配列がコードするアミノ酸配列について、 HM MP F AMによる蛋白質特徴検索を行ったところ配列番号 15の塩基番号 180 0- 2596がコードするアミノ酸配列に Gタンパク質共役型受容体の特徴を示 す配列 (P f amに 「7 tm— 2」 としてエントリーされる塩基配列) を見出し、 塩基番号 1647—1 790がコードするアミノ酸配列に L a t r o p h i 1 i n/C L- 1 - l i k e GPS d oma i n (P f a に 「GP S」 として エントリーされる塩基配列) を見出した。 The amino acid sequence encoded by the nucleotide sequence shown in SEQ ID NO: 15 was subjected to a protein feature search using HMMP FAM. Show body characteristics Sequence (base sequence entered as “7 tm—2” in P f am), and the amino acid sequence encoded by base Nos. 1647–1790 contains Latrophi 1 in / C L-1-like GPS doma in (Base sequence that is entered as “GP S” in P fa).

上記の C y t 28はデータベース登録記号 AF 166382中の記載によると、 マウス造血幹細胞からクローニングされたことが示されている。 上記の、 GPR 56はデータベース中の文献情報 (Genomics 1999 Feb 1;55(3) :296-305) から、 甲状腺、 心臓、 脳に強く発現し、 脳内では海馬、 視床に特に強く発現し、 細胞外 にムチン様の構造を有することから細胞間相互作用に関与することが示唆されて いる。 これらのことから配列番号 1.5に示す塩基配列がコードするタンパク質は Gタンパク質共役型受容体であり、 細胞間相互作用に関連する活性を有すること が推測された。 +  According to the description in the database entry number AF 166382, the above Cyt28 was shown to be cloned from mouse hematopoietic stem cells. From the literature information in the database (Genomics 1999 Feb 1; 55 (3): 296-305), GPR 56 is strongly expressed in the thyroid gland, heart and brain, and is particularly strongly expressed in the hippocampus and thalamus in the brain, Its extracellular mucin-like structure has been suggested to be involved in cell-cell interactions. From these facts, it was presumed that the protein encoded by the nucleotide sequence shown in SEQ ID NO: 1.5 was a G protein-coupled receptor and had an activity related to cell-cell interaction. +

(16) d n a f o rm45616 (配列番号 16、 32)  (16) d n a f o rm45616 (SEQ ID NO: 16, 32)

d n a f o rm45616は、 配列番号 1 6に示すように、 2774塩基から 成り、 そのうち塩基番号 31 1から 1210までがオープンリーディングフレー ム (終止コドンを含む) になっていた。 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は、 299アミノ酸残基から成る (配列番号 32)。配列番号 16がコードするアミノ酸配列について B LASTを用いて相同性検索を行った ところ、 S PTR蛋白質データベース (SWI S S—PROT蛋白質配列データ ベースと T r EMBL核酸翻訳データベースを統合したもの) 中に、 ( i )データ ベース登録記号 A F 039686、 Homo s a i e n s G— p r o t e i n c o u l e d r e c e p t o r GPR 34 (GPR3 ) 力 e— v a 1 u e : 1 X 10— 19で、 また 294アミノ酸残基に亘り 27%の一致度で、 また(1 ¾0データべース登録記号<391 11:6、1^ 1 3 mu s c u l u s G p r o t e i n— c o up l e d r e c e p t o r Gp r ^ 4(G r 34) g e n e力 e— v a l u e : 5 X 10一18で、 294ァミノ酸残基に亘り 38 % の一致度でヒットした。 また、 配列番号 16に示す塩基配列がコードするアミノ 酸配列について、 膜貫通へリックスを予測するプログラム t m匪 (S. Moller, M. D. R. Croning, R. Apweiler. Evaluation of methods for tne prediction of membrane spanning regions. Bioinformatics, 17 (7) :646-653, 2001) を用いて 膜貫通部位を予測したところ、 7つの膜貫通部位 (配列番号 32のァミノ酸番号 で、 30〜52、 65〜87、 107〜 1 29、 141〜 163、 197〜 21 9、 239〜 261、 276〜 298) に膜貫通部位が予測された。 As shown in SEQ ID NO: 16, dnafo rm45616 was composed of 2774 bases, of which base numbers 311 to 1210 were open reading frames (including a stop codon). The amino acid sequence predicted from the open reading frame consists of 299 amino acid residues (SEQ ID NO: 32). When a homology search was performed using BLAST for the amino acid sequence encoded by SEQ ID NO: 16, the SPTR protein database (the SWI SS-PROT protein sequence database and the TrEMBL nucleic acid translation database were integrated) (i) the data base registration mark AF 039686, Homo saiens G- proteincouledreceptor GPR 34 (GPR3) force e- va 1 ue: at 1 X 10- 19, and in 27% of the degree of coincidence over the 294 amino acid residues, and ( 1 ¾0 data base over to register symbol <391 11: 6,1 ^ 1 3 mu sculus G protein- co up ledreceptor Gp r ^ 4 (G r 34) gene force e- value: at 5 X 10 one 18, 294 Amino The hit was achieved with 38% identity over the acid residues, and the amino acid encoded by the nucleotide sequence shown in SEQ ID NO: 16 A program that predicts transmembrane helices for acid sequences tm marauder (S. Moller, MDR Croning, R. Apweiler. Evaluation of methods for tne prediction of membrane spanning regions. Bioinformatics, 17 (7): 646-653, 2001) When the transmembrane site was predicted using, the seven transmembrane sites (30-52, 65-87, 107-129, 141-163, 197-219, 239- 261, 276-298).

上記の G P R 34はデータベース中の文献情報(BiochimBiophys Acta 1999 Jul 7; 1446 (1-2): 57-70)から、platelet- activating factor (PAF)受容体類似であり、 胎児脳のライブラリーからクローユングされ、 ヒトおよびマウスの各種臓器で豊 富に発現していることが示されている。 また、 データベース中の文献情報 (um Genet 2000 Jul; 107 (1): 89-91) から、 GPR34は X染色体上の先天性夜盲症 (CSNB) の原因遺伝子座近傍に位置し眼に発現するが、 CSNBの原因遺伝子ではないことが 示されている。 これらの結果より配列番号 32に示したアミノ酸配列からなるタ ンパク質は新規な Gタンパク質共役型受容体活性を有することが推測された。 これらの結果より配列番号 32に示したアミノ酸配列からなるタンパク質は G タンパク質共役型受容体活性を有することが推測された。 また、 類似のタンパク 質をコードする遺伝子は、 X染色体上にあり、 各種臓器で豊富に発現し、 PAF受 容体と弱い相同性を有する。  The above GPR 34 is analogous to the platelet-activating factor (PAF) receptor from literature information in the database (BiochimBiophys Acta 1999 Jul 7; 1446 (1-2): 57-70). It has been shown to be abundantly expressed in various human and mouse organs. Also, from the literature information in the database (um Genet 2000 Jul; 107 (1): 89-91), GPR34 is located near the causal locus of congenital night blindness (CSNB) on the X chromosome and is expressed in the eyes, It has been shown not to be the causative gene of CSNB. From these results, it was presumed that the protein consisting of the amino acid sequence shown in SEQ ID NO: 32 had a novel G protein-coupled receptor activity. From these results, it was presumed that the protein consisting of the amino acid sequence shown in SEQ ID NO: 32 had G protein-coupled receptor activity. In addition, genes encoding similar proteins are on the X chromosome, are abundantly expressed in various organs, and have weak homology to PAF receptors.

実施例 5 組織発現解析 Example 5 Tissue expression analysis

組織発現解析は、 Miki, R. , et al. , Proc. Natl. Acad. Sci. USA, 98, 2199-2204 (2001)の記載に従って行つた。  Tissue expression analysis was performed as described in Miki, R., et al., Proc. Natl. Acad. Sci. USA, 98, 2199-2204 (2001).

(1) DN Aマイクロアレーの作成  (1) Creation of DN A microarray

配列番号 1に記載の塩基配列を含むマウス cDNAクローン: d n a f o rm 34147、 配列番号 4に記載の塩基配列を含むマウス cDNAクローン: d n a f o rm4285 1、 配列番号 7に記載の塩基配列を含むマウス c DNAク口 ーン: dn a f o rm29894、 配列番号 1 1に記載の塩基配列を含むマウス cDNAクローン: dn a f o rm32567、 配列番号 14に記載の塩基配列 を含むマウス c DNAクローン: d n a f o rm54572と同じクラスタに属 する c DNAクローン: FANTOM 9330 161N09 Mouse cDNA clone containing the nucleotide sequence of SEQ ID NO: 1: dnafo rm 34147, mouse cDNA clone containing the nucleotide sequence of SEQ ID NO: 4: dnafo rm42851, mouse cDNA clone containing the nucleotide sequence of SEQ ID NO: 7 Mouth: dn afo rm29894, mouse cDNA clone containing the nucleotide sequence of SEQ ID NO: 11: dn afo rm32567, nucleotide sequence of SEQ ID NO: 14 Mouse cDNA clone containing: cDNA clone belonging to the same cluster as dnafo rm54572: FANTOM 9330 161N09

(http: //fantom. gsc. riken. go. jp/)、およぴ配列番号 15に記載の塩基配列を含 むマウス c DNAクローン: d n a f o rm29069と同じクラスタに属する c DNAクロ一ン: FAN TOM 3830420G05  (http: // fantom. gsc. riken. go. jp /), and a mouse cDNA clone containing the nucleotide sequence of SEQ ID NO: 15: cDNA clone belonging to the same cluster as dnafo rm29069: FAN TOM 3830420G05

(http: //fantom. gsc. riken. go. jp/) 中のマウス c D N A部分の塩基配列を含む DNAを、 Ml 3フォワードおよびリバースプライマーを用いて増幅後、 この P CR産物をイソプロパノールにて沈澱させ 15 β 1の 3 X S SC液に溶解した。 得られた DN Α溶液をポリ Lリジンコートしたガラススライドに、 16チップ(S MP d、 丄、 e l eし! i em I n t e r n a t l o n a Sunny v a l e、 CA) の DNAアレイヤーを用いてスポットし、 DNAマイクロアレーを作成し 7こ 、万法の 糸田 fihttp://cmgm. stanford.edu/pbrown/ mguide/index. htm こ目 ι3載 されている)。マウス i3ァクチンとダリセルアルデヒ ド- 3 -フォスフエ一トデヒ ドロゲナーゼの c DNAをポジティブコントロールとし、 シロイヌナズナの c D NAをネガティブコントロールとして用いた。  (http: //fantom.gsc.riken.go.jp/) After amplifying the DNA containing the nucleotide sequence of the mouse cDNA portion using Ml3 forward and reverse primers, the PCR product was amplified with isopropanol. Precipitated and dissolved in 3XS SC solution of 15β1. The obtained DNΑ solution was spotted on a glass slide coated with poly-L-lysine using a DNA arrayer of 16 chips (S MP d, 丄, ele! I Internatlona Sunny vale, CA), and a DNA microarray was prepared. It has been created and is listed in Manpo's Itoda fihttp: // cmgm. Stanford.edu/pbrown/mguide/index.htm. Mouse i3 actin and daricell aldehyde-3-phosphate dehydrogenase cDNA were used as a positive control, and Arabidopsis thaliana cDNA was used as a negative control.

この DNAマイクロアレーの検出感度は、 1細胞当たり mRNA 1ないし 3コ ピーであった。 ターゲット配列との一致度がおよそ 80%のクローンのシグナル 強度は、 完全に配列が一致するクローンの 10分の 1であった。 ターゲット配列 との一致度が 80 %未満のクローンのシグナル強度は、 バックグランドレベルで めった。  The detection sensitivity of this DNA microarray was 1 to 3 copies of mRNA per cell. The signal intensity of clones with approximately 80% match with the target sequence was one-tenth that of clones with perfect sequence match. The signal intensity of clones with less than 80% match with the target sequence was reduced at the background level.

(2) プローブの調製  (2) Preparation of probe

C 57BL/6 Jマウスの胎児、 新生仔、 アダルトの 49組織 (腎臓、 脳、 脾 臓、 肺、 肝臓、 精巣、 脖臓、 胃、 小腸、 結腸、 盲腸、 胎盤、 心臓、 舌、 胸腺、 胸 腺 (妊娠 1日目)、 小脳、 延髄、 嗅脳、 副精巣、 眼球、 皮質、 小胞腺、 子宮、 卵巣 およぴ子宫 (妊娠 1 1日目)、 骨、 筋肉、 乳腺 (授乳 10日目)、 10日齢胎児全 身、 1 1日齢胎児全身、 1 3日齢胎児全身、 1 1 日齢胎児頭部、 12日齢胎児頭 部、 13日齢胎児頭部、 15日齢胎児頭部、 16日齢胎児頭部、 17日齢胎児頭 部、 1 6日齢胎児肺、 1 3日齢胎児肝臓、 1 4日齢胎児肝臓、 0日齢新生児全頭 部、 6日齢新生児全頭部、 1 0日齢新生児全頭部、 1 0日齢新生児腸、 0日齢新 生児肺、 1 0日齢新生児小脳、 0日齢新生児皮膚、 1 0日齢新生児皮膚、 SV4 0感染) から抽出した mRNA l gを定法に従いランダムプライム逆転写反応 を行い蛍光色素 C y 3 (Ame r s h a m P h a r ma c i a社) を取りこま せた。 他方、 1 7. 5日齢の胎児全身から抽出した mRNA l μ gをランダムプ ライム逆転写反応を行い、 蛍光色素 C y 5を取りこませ発現解析のリファレンス とした。 Cy D y e標識 c DNAプローブは、 C y S c r i b e GFX P u r i i i c a t i o n K i t (Am e r s h a m P h a r ma c i aネ ι 用レ、 て精製し、 滅菌水 1 7 1にてカラムから溶出した。 これに 3 μ 1の 1 0 μ gノ μ 1 o l i g o (dA)、 3 μ 1の酵母 t RNA 2 0 μ g/ μ 1 , Ι μ ΐの 249 tissues of fetal, neonatal and adult C57BL / 6J mice (kidney, brain, spleen, lung, liver, testis, kidney, stomach, small intestine, colon, cecum, placenta, heart, tongue, thymus, breast Glands (1st day of pregnancy), cerebellum, medulla oblongata, olfactory brain, epididymis, eyes, cortex, follicles, uterus, ovaries and ovary (1st day of pregnancy), bones, muscles, mammary glands (10th day of lactation) ), Whole 10-day-old fetus, 11-day-old fetal whole body, 13-day-old fetal whole body, 11-day-old fetal head, 12-day-old fetal head, 13-day-old fetal head, 15-day-old fetal head Part, 16 days old fetal head, 17 days old fetal head Part, 16-day-old fetal lung, 13-day-old fetal liver, 14-day-old fetal liver, 0-day-old newborn whole head, 6-day-old newborn whole head, 10-day-old newborn whole head, 10 Random prime reverse transcription reaction of mRNA lg extracted from day-old neonatal intestine, 0-day-old neonatal lung, 10-day-old neonatal cerebellum, 0-day-old neonatal skin, 10-day-old neonatal skin, SV40 infection Then, the fluorescent dye Cy 3 (Amersham Pharmacia) was incorporated. On the other hand, 1 μg of mRNA extracted from the whole body of a 17.5-day-old fetus was subjected to a random prime reverse transcription reaction, and the fluorescent dye Cy5 was taken in as a reference for expression analysis. The CyDye-labeled cDNA probe was purified using CyScribe GFX Purification Kit (Amersham Pharmacia), and eluted from the column with sterile water 171, and 3 μl was added thereto. 1 of 10 μg no μ 1 oligo (dA), 3 μ 1 of yeast tRNA 20 μg / μ 1, Ι μ 2 2

0 μ 1マウス C ο t 1 DNA, 5. 1 μ 1の 20 X S S C、 およぴ 0. 9 1の 1 0%SD Sからなるブロッキング溶液を混和して C yD y e標識 c D N Aプローブを調製した。 A CyDye-labeled cDNA probe was prepared by mixing a blocking solution consisting of 0 μl mouse Cοt1 DNA, 5.1 μl of 20 XSSC, and 0.91 of 10% SDS. .

(3) DNAマイクロアレーのハイブリダィゼイシヨン  (3) DNA microarray hybridization

発現解析対象組織由来 c DN Aプローブ(C y 3標識)とリファレンスの 1 7. 5日齢胎児由来 c D N Aプローブ(C y 5標識)を混和した溶液 3 0 μ 1を 9 5 °C にて 1分間熱処理を行い室温にて冷却した。 DNAマイクロアレーに上記プロ一 ブ溶液を添加し力バースリップを被せ、 Hy b r i c a s e t t e (A r r a y A 30 μl solution of the cDNA analysis target tissue-derived cDNA probe (Cy3 label) and the reference 17.5-day-old fetal cDNA probe (Cy5 label) was mixed at 95 ° C. Heat treatment was carried out for 1 minute and cooled at room temperature. Add the above probe solution to the DNA microarray and cover with a force bar slip.Hybricacsette (Arraya

1 t社) 中にて 6 5 °C—晚ハイブリダィズさせた。 次に、 DNAマイクロアレー を 2 X S S C、 0. 1 %S D Sを用いて洗浄し、 続いて 1 X S S Cにて 2分間、 0. 1 X S S Cにて 2分間リンスした。 マイクロアレーは S c a nA r r a y 5 0 0 0共焦点レーザースキャナーを用いてスキャンし、画像を I MAGENE (B i o D i s c o v e r y社) で解析した。 It was hybridized at 65 ° C- 晚 in 1 t company). Next, the DNA microarray was washed with 2 × SSC and 0.1% SDS, and then rinsed with 1 × SSC for 2 minutes and with 0.1 × SSC for 2 minutes. The microarray was scanned using a ScanA 500 000 confocal laser scanner and the images were analyzed on an I MAGENE (BioDiversive).

(4) データ解析  (4) Data analysis

各組織中の mRNA量 (C y 3標識) は、 リファレンスの 1 7. 5日齢の胎児 全身 mRNA量 (C y 5標識) との比 (C y 3/C y 5) を対数 ( 1 o g 2) で 表示した。 すなわち、 解析対象とする各マウス全長 c DNAに対応する mRNA 発現量が、 リファレンス組織中よりも各組織中の方が多い場合は正の数値で、 少 ない場合は負の数値で、 等しい場合は 0で示される。 データの正確性を増すため に実験は独立に 2回行い、再現性の有る結果を採用した。その結果を表 1に示す。 一般的に、 DNAマイクロアレーを使用した発現解析結果は、 その数値の 2倍程 度の増減は実験誤差とみなすため、 ある組織における発現解析結果の数値が 1以 上の場合には、 対照である 1 7. 5日齢の胎児全身の mRNA量に対して該組織 中の解析対象 mRNAの量が 2倍以上であり、 有意に増加していると解釈した。 逆に、 ある組織における発現解析結果の数値が— 1以下の場合は、 対照である 1 7. 5日齢の胎児全身の mRNA量に対して該組織中の解析対象 mRNA量が 2 分の 1以下であり、 有意に減少していると解釈した。 The mRNA level (Cy3 label) in each tissue is expressed as the ratio (Cy3 / Cy5) to the reference 17.5-day-old fetal whole-body mRNA level (Cy5 label) as a logarithm (1 og). 2 ) in displayed. That is, if the expression level of mRNA corresponding to the full-length cDNA of each mouse to be analyzed is larger in each tissue than in the reference tissue, a positive value is used, if it is smaller, a negative value, and if it is equal, Indicated by 0. Experiments were performed twice independently to increase the accuracy of the data, and reproducible results were used. The results are shown in Table 1. In general, the results of expression analysis using a DNA microarray are considered to be experimental errors if the value is increased or decreased by a factor of about two. It was interpreted that the amount of mRNA to be analyzed in the tissue was more than twice as large as the mRNA amount of the whole fetal body at 17.5 days of age, and that the amount was significantly increased. Conversely, if the value of the expression analysis result in a certain tissue is less than or equal to -1, the amount of the mRNA to be analyzed in the tissue is one-half of that of the control 17.5-day-old fetus whole body. The following were interpreted as significantly reduced.

また、 組織間の mRNA発現量を比較検討する際は、 各組織における数値の差 が 1であれば mRNA量は 2倍、 2であれば mR N A量は 4倍であり、 逆に、 組 織間の数値の差が— 1であれば mRNA量は 1Z 2倍、 _ 2であれば mRNA量 は 1/4倍となる。  When comparing the mRNA expression levels between tissues, when the difference between the values in each tissue is 1, the mRNA level is 2 times, and when the difference is 2, the mRNA level is 4 times. If the difference between the values is -1, the mRNA amount is 1Z 2 times, and if it is _2, the mRNA amount is 1/4 times.

なお、 マイクロアレイにスポットした DNAと同じクラスタに属し、 該 DNA と少なくとも 200塩基に亘り 80%以上の塩基配列の一致度を有する領域を有 するマウス c DNAクローンについても、表 1に解析対象 c DNAとして記載し、 マイクロアレイにスポットした該 DN Aの測定結果の数値を代用して記載した。 Mouse cDNA clones belonging to the same cluster as the DNA spotted on the microarray and having a region having a nucleotide sequence identity of at least 80% with at least 200 bases are also shown in Table 1 And the numerical value of the measurement result of the DNA spotted on the microarray was used instead.

表 1 : table 1 :

解析対象 cDN マイクロアレ一にスポットし 腎臓 脳 脾臓 肺 肝臓 Analysis target Spot on cDN microarray Kidney Brain Spleen Lung Liver

A _ _ fcD A A _ _ fcD A

dnaform34147 dnaform34147 0 一 0.604644 -0.918401 -0.394848 0.003062 dnaform42851 dnaform42851 -0.028749 0 -0.035681 0.263115 0.512224 dnaform29894 dnaform29894 0.211066 1.21558 0.525265 -0.218343 0.712455 dnaform32567 dnaform32567 0 1.52082 0.375472 0 0.868992 dnaform54572 FANTOM NO:9330161 09 0 0 0.701116 -0.136448 0 dnaform29069 FANTOM NO3830420G05 -0.297707 -0.817491 —0,694666 -0.009974 0.177833 解析対象 cDN マイクロアレーにスポットし 精巣 膝臓 小腸 結腸 dnaform34147 dnaform34147 0--1 0.604644 -0.918401 -0.394848 0.003062 dnaform42851 dnaform42851 -0.028749 0 -0.035681 0.263115 0.512224 dnaform29894 dnaform29894 0.211066 1.21558 0.525265 -0.218343 0.712455 dnaform32567 dnaform32567 0 1.52081 0.375na0 0.792 ANT017201F 072 0.297707 -0.817491 —0,694666 -0.009974 0.177833 Test spots spotted on the cDN microarray to be analyzed testis knee small intestine colon

A た DNA A was DNA

dnaform34147 dnaform34l / 0.002757 0.389407 0.04 38 一 0.699333 -0.333782 dnaform42851 dnaform42851 -0.49458 1.16233 0.217832 -0.070995 0.658057 dnaform29894 dnaform29894 0.007426 1.79989 0.483078 0.310433 -0.213772 dnaform32567 dnaform32567 1.01259 1.55638 0 0.292057 0.690966 dnaform54572 FANTOM NO:9330161 09 0 1.39164 0.265034 0.494954 0.401273 dnaform29069 FANTOM NO3830420G05 -0.992702 0.549824 一 0.305854 -0.074891 -0.001997 解析対象 cDN マイクロアレーにスポットし 胎盤 心臓 胸腺 dnaform34147 dnaform34l / 0.002757 0.389407 0.04 38 i 0.699333 -0.333782 dnaform42851 dnaform42851 -0.49458 1.16233 0.217832 -0.070995 0.658057 dnaform29894 dnaform29894 0.007426 1.79989 0.483078 0.310433 -0.213772 dnaform32567 dnaform32567 1.01920 1.596 0. 0.992702 0.549824 1 0.305854 -0.074891 -0.001997 Analyzed target spot on cDN microarray Placental heart Thymus

A た DNA A was DNA

dnaform34l 47 dnaform3414/ 一 0.283769 —0.372257 - 0.659996 -1.14156 0.361602 dnaform42851 dnaform42851 0.567144 0.188979 -0.661836 0.669685 0.139335 dnaform29894 dnaform29894 0.148378 0.049242 0.171353 -0.440989 -0.091025 dnafortn32567 dnaform32567 0 0.594181 0.620701 -0.132696 1.12992 dnaform54572 FANTOM NO.9330161 09 0.113867 0.344982 0.169048 -0.168131 0 dnaform29069 FANTOM NO3830420G05 1.80195 -0.294741 -0.140147 -0.943278 -0.16034 解析対象 cDN マイクロアレーにスポットし 骨 筋肉 背側腎臓由 副精巣由来 内臓脂肪 A fcDNA 来脂肪細胞 脂肪細胞 dnaform34l 47 dnaform3414 / ichi 0.283769 -0.372257-0.659996 -1.14156 0.361602 dnaform42851 dnaform42851 0.567144 0.188979 -0.661836 0.669685 0.139335 dnaform29894 dnaform29894 0.148378 0.049242 0.171353 -0.440989 -0.091025 dnafortn320.1 0.194930.10940.18 dnaform29069 FANTOM NO3830420G05 1.80195 -0.294741 -0.140147 -0.943278 -0.16034 Analysis target Spot on cDN microarray Bone muscle Muscle Dorsal kidney Derived from epididymis Visceral fat A fcDNA Original fat cell Adipocyte

dnaTorm34l47 dnaform34147 -0.3937 0.012674 - 0.332067 -0.358397 - 1.09285 dnaform42851 dnaform42851 0.200827 0.4848 0.241472 0 dnaform29894 dnaform29894 -0.100222 0.633098 0.54399 0.505093 0.086003 dnaform32567 dnaform32567 -0.91889 0.54258 0.635356 0.431214 0.150435 dnaform54572 FANTOM NO:9330161 N09 0.320531 0.74831 1.17013 1.30375 0.912332 dnaform29069 FANTOM NO3830420G05 1.85781 0.161977 -0.247306 -0.361052 -0.086625 解析対象 cDN マイクロアレーにスポットし 10曰齢新 10曰齢新生 dnaTorm34l47 dnaform34147 -0.3937 0.012674 - 0.332067 -0.358397 - 1.09285 dnaform42851 dnaform42851 0.200827 0.4848 0.241472 0 dnaform29894 dnaform29894 -0.100222 0.633098 0.54399 0.505093 0.086003 dnaform32567 dnaform32567 -0.91889 0.54258 0.635356 0.431214 0.150435 dnaform54572 FANTOM NO: 9330161 N09 0.320531 0.74831 1.17013 1.30375 0.912332 dnaform29069 FANTOM NO3830420G05 1.85781 0.161977 - 0.247306 -0.361052 -0.086625 Spot on the analysis target cDN microarray.

A た DNA 生児小脳 児皮膚 ― A DNA Liver cerebellum Child skin ―

dnaform34147 dnaform34147 -0.579103 -0.935412 dnaform34147 dnaform34147 -0.579103 -0.935412

dnaform42851 dnaform42851 0.246094 0.517231 dnaform42851 dnaform42851 0.246094 0.517231

dnaform29894 dnaform29894 0 0.55205 dnaform29894 dnaform29894 0 0.55205

dnaform32567 dnaform32567 -0.1335 —0.124381 dnaform32567 dnaform32567 -0.1335 —0.124381

dnaform54572 FANTOM NO:9330161 N09 1.12068 0.643064 dnaform54572 FANTOM NO: 9330161 N09 1.12068 0.643064

dnaform29069 FANTOM NO3830420G05 -0.494198 -0.204861 表 1から明らかなように、 d n a f o r m 3 4 1 4 7は、 対照 (1 7 . 5日胎 児全身) と比較して、 脾臓おょぴ子宮で発現が増加する蛍光があり、 精巣および 腎臓などでは対照と同程度の発現であつたが、 全体として対照と比較して発現量 が減少した。 d n a f o r m 4 2 8 5 1は対照と比較して筋肉および脾臓で発現 が増加し、 小脳、 結腸、 および胎盤で発現が増加する傾向にあり、 脳では同等の 発現であった。 d n a f o r m 2 9 8 9 4は、 対照と比較して脾臓で発現が強く 増加し、 脳でも増加した。 dn a f o rm32567は対照と比較して脾臓およ ぴ脳で強く増加し、 子宮および精巣でも増加し、 肝臓や結腸などで増加する傾向 があった。 dn a f o rm54572は、 対照と比較して脾臓、 脂肪細胞、 10 日齢新生児小脳で発現が増加し、 脳では対照と同程度発現していたが、 全体的に 対照と比較して発現が増加する傾向があった。 d n a i o rm29069は、 対 照と比較して骨およぴ胎盤で強く増カ卩し、 脾臓および筋肉でも増加する傾向があ つた。 dnaform29069 FANTOM NO3830420G05 -0.494198 -0.204861 As can be seen from Table 1, dnaform 3 4 1 4 7 has increased fluorescence in the spleen and uterus compared to the control (17.5 day fetal whole body) The expression was similar to that of the control in testis and kidney, but the expression level was reduced as a whole as compared to the control. The expression of dnaform 42851 increased in muscle and spleen, increased in cerebellum, colon, and placenta compared to controls, and was equivalent in brain. dnaform 2 9 8 9 4 is more strongly expressed in the spleen compared to controls Increased, and also increased in the brain. Compared to controls, dnafo rm32567 increased strongly in spleen and brain, increased in uterus and testis, and tended to increase in liver and colon. dn afo rm54572 is up-regulated in spleen, adipocytes, and 10-day-old neonatal cerebellum as compared to control, and is expressed to the same extent as control in brain, but is up-regulated overall as compared to control There was a tendency. dnaio rm29069 increased strongly in bone and placenta and tended to increase in spleen and muscle as compared to control.

実施例 6 P CR法を用いた組織発現解析 Example 6 Tissue expression analysis using PCR method

本発明のタンパク質をコードする niRN Aの正常マウスおよび疾患マウスでの 組織発現変動を検討するために、 定法 (Higuchi R, et al. , Biotechnology, 11: 1026-30 (1993)) に従い、 P C R法を用いた組織発現解析を行った。  In order to examine the changes in tissue expression in normal and diseased mice of niRNA encoding the protein of the present invention, PCR was performed according to a standard method (Higuchi R, et al., Biotechnology, 11: 1026-30 (1993)). Was used to perform tissue expression analysis.

(1) cDNA合成  (1) cDNA synthesis

以下のマウス (森脇和郎、 外 1名編、 Molecular Medicine別冊、 Vol. 36 「自 然発症疾患モデル動物 」、 中山書店、 1999年) の 1 9組織からトータル R Aを抽 出し、オリゴ dTをプライマーとして逆転写酵素を用いて cDNA合成を行った。 ( a ) 正常マゥスの組織およぴ糖尿病モデルマゥスの組織  Total RA was extracted from 19 tissues of the following mice (Kazuo Moriwaki, 1st Edition, Molecular Medicine Separate Volume, Vol. 36 “Model Animals with Spontaneous Diseases”, Nakayama Shoten, 1999), and oligo dT was used as a primer. CDNA synthesis was performed using reverse transcriptase. (a) Tissue of normal mice and tissue of diabetic model mice

①対照マウス C57BL/KsJ - +m/+m Jcl (メス、 8週齢) の全脳、 視床、 肺、 腎 臓、 骨髄、 膝臓、 脂肪細胞、 肝臓、 眼  ① Control mouse C57BL / KsJ-+ m / + m Jcl (female, 8 weeks old) whole brain, thalamus, lung, kidney, bone marrow, knee, adipocyte, liver, eye

②糖尿病モデルマウス CWBL/KsJ- db/db Jcl (メス、 8週齢) の脾臓、 脂肪 細胞、 肝臓、 眼  ② Diabetes model mouse CWBL / KsJ-db / db Jcl (female, 8 weeks old) spleen, fat cells, liver, eyes

(b) 老化促進マウスの組織  (b) Aging-promoting mouse tissue

①正常老化マウス SAM Rl/TA Sic (ォス、 13週齢) の海馬、 前頭葉皮質 (1) Hippocampus and frontal cortex of normal aging mouse SAM Rl / TA Sic (13 weeks old)

②老化促進マウス SAM P8/Ta Sic (ォス、 15週齢) の海馬、 前頭葉皮質② Senescence-accelerated mouse SAM P8 / Ta Sic (Oss, 15 weeks old) hippocampus, frontal cortex

(c) 癌転移モデルマウスの組織 (c) Tissue of cancer metastasis model mouse

①対照マウス Balb/c (メス、 5週齢) の正常結腸  ① Normal colon of control mouse Balb / c (female, 5 weeks old)

②癌転移モデルマウス Balb/c (メス、 6週齢) の結腸癌 (マウス腹腔に結腸 癌細胞 Colon26を移植し、 2週間後に結腸癌を摘出) (2) PCR法による定量 ② Colon metastasis of cancer metastasis model mouse Balb / c (female, 6 weeks old) (Colony 26 colon cancer cells are transplanted into the abdominal cavity of the mouse and colon cancer is removed 2 weeks later) (2) Quantification by PCR method

下記の 4個の、 本発明のタンパク質をコードしている mRNAの発現は、 ライ トサイクラー定量 PCR 装置 (ロシュ · ダイァグノスティ タス社) と LightCycler-FastStart DNAマスター SYBR Green I試薬を用いて、 製品に添付 されているプロトコールに従い定量した。 定量 PCRに用いた合成 DNA配列を以 下に示す。  Expression of the following four mRNAs encoding the protein of the present invention was attached to the product using a light cycler quantitative PCR device (Roche Diagnostics) and LightCycler-FastStart DNA Master SYBR Green I reagent. Quantification was performed according to the protocol provided. The synthetic DNA sequences used for quantitative PCR are shown below.

(a) dn a f o rm33729  (a) dn a f o rm33729

5, 側プライマー: (AGCAAAGGGTCTCCATTGTC) (配列番号 41 )  5. Side primer: (AGCAAAGGGTCTCCATTGTC) (SEQ ID NO: 41)

3, 側プライマー: (AAACCCAAGCTGAGGTAGCA) (配列番号 42)  3. Side primer: (AAACCCAAGCTGAGGTAGCA) (SEQ ID NO: 42)

( b ) d n a i o rm456 16  (b) d n a i o rm456 16

5, 側プライマー: (TGATCATTGAACTTTGGGTCA) (配列番号 43)  5, side primer: (TGATCATTGAACTTTGGGTCA) (SEQ ID NO: 43)

3, 側プライマー: (CCCTGAAGTATGGGAGCAAA) (配列番号 44)  3. Side primer: (CCCTGAAGTATGGGAGCAAA) (SEQ ID NO: 44)

( c) d n a f o r m47734  (c) d n a f o r m47734

5, 側プライマー: (CAAACAACTAAGCGCAAATCC) (配列番号 45)  5, side primer: (CAAACAACTAAGCGCAAATCC) (SEQ ID NO: 45)

3, 側プライマー: (ACCGCTTAAAAGGCCAAAGT) (配列番号 46)  3. Side primer: (ACCGCTTAAAAGGCCAAAGT) (SEQ ID NO: 46)

(d) d n a f o r m48477  (d) d n a f o r m48477

5, 側プライマー: (CTGGGGAACTATTCCAAGCA) (配列番号 47)  5, side primer: (CTGGGGAACTATTCCAAGCA) (SEQ ID NO: 47)

3, 側プライマー: (TCTGGGCATTTGTGTTGAAG) (配列番号 48)  3. Side primer: (TCTGGGCATTTGTGTTGAAG) (SEQ ID NO: 48)

定量結果は Glyceraldehyde 3 - phosphate dehydrogenase (GAP DH) を内咅 [5 標準として、 補正した。 即ち、 各組織での対象遺伝子の発現量 (コピー数/ μ 1 ) を GAPDH の発現量 (コピー数/ 1 ) で除し、 定数 ( 1 X 106) (注: 10の 6 乗) を乗して表示した。 その結果を表 2に示す。 表 2 : The quantification results were corrected using Glyceraldehyde 3-phosphate dehydrogenase (GAP DH) as the internal standard [5 standards]. That is, the expression level (copy number / μ1) of the target gene in each tissue is divided by the GAPDH expression level (copy number / 1), and the result is raised to a constant (1 × 10 6 ) (Note: 10 to the sixth power). And displayed. The results are shown in Table 2. Table 2:

Figure imgf000069_0001
表 2から明らかなように、 dn a f o rm33729は眼での発現が高く、肺、 脂肪、 膝臓で発現が観察され、 結腸癌で発現が減少した。 d n a f 0 rm456 16は肺での発現が高く、 勝臓、 脂肪、 結腸で発現が観察されたが、 糖尿病の膝 臓おょぴ結腸癌では発現が減少した。 d n a f o rm47734は脳組織特に前 頭葉皮質および海馬での発現が高く、記憶学習との関連が示唆され、膝臓や脂肪、 および骨髄でも発現が観察された。 d n a f o rm48477は骨髄、 肺で発現 が高く、 糖尿病の脂肪組織で発現が増加した。 上記クローンの cDNAおよぴ該 cDNAによってコードされるタンパク質は、 糖尿病や癌などの治療や診断に応 用できる。 また該 cDNAによってコードされるタンパク質は、 上記のような m RNA発現の変動が見られる組織あるいは mRNA発現量の多い組織に関わる疾 患に関与している可能性がある。 実施例 7 機能予測 (1) dn a f o rm34 147
Figure imgf000069_0001
As is evident from Table 2, expression of dnafo rm33729 was high in eyes, expression was observed in lung, fat and knee, and decreased in colon cancer. The expression of dnaf 0 rm456 16 was high in the lung, and was observed in the viscera, fat, and colon, but decreased in diabetic knee and colon cancer. dnafo rm47734 was highly expressed in brain tissues, especially in the frontal cortex and hippocampus, suggesting a relationship with memory learning, and was also observed in knees, fat, and bone marrow. dnafo rm48477 was highly expressed in bone marrow and lung, and increased in diabetic adipose tissue. The cDNA of the above clone and the protein encoded by the cDNA can be applied to the treatment and diagnosis of diabetes and cancer. Further, the protein encoded by the cDNA may be involved in a disease relating to a tissue in which the mRNA expression is varied or a tissue having a high mRNA expression level as described above. Example 7 Function prediction (1) dn afo rm34 147

実施例 4 (1) の結果より、 本 cDNAがコードするタンパク質は、 その類似 のタンパク質が精巣の発達と機能維持、 生殖能に関与することがわかっている。 また、 実施例 5および 6の結果より、 本 c DNAがコードするタンパク質は、 対 照 (1 7. 5日齢胎児全身) と比較して勝臓および子宮で発現が増加する傾向が あり、 精巣おょぴ腎臓などでは対照と同程度の発現であった。  From the results of Example 4 (1), it is known that the proteins encoded by the present cDNA are similar proteins involved in testis development, function maintenance, and fertility. In addition, from the results of Examples 5 and 6, the expression of the protein encoded by the cDNA tends to increase in the viscera and uterus as compared to the control (17.5-day-old fetal whole body), The expression was similar to that of the control in such as kidney.

これらのことから、 本 cDNAがコードするタンパク質は、 精巣の発達と機能 維持、 生殖能に関与することと子宮に発現していることから、 不妊 ·避妊、 糖尿 病に関連する機能を有し、 これらの治療薬の解析に有用であると考えられた。  Based on these facts, the protein encoded by this cDNA has functions related to infertility, contraception, and diabetes, because it is involved in testis development and maintenance, function in fertility, and is expressed in the uterus. It was considered useful for the analysis of these therapeutic agents.

(2) d n a f o rm37308  (2) d n a f o rm37308

実施例 4 (2)、実施例 5および 6の結果より、本 c DNAがコードするタンパ ク質は、 脳に発現し、 ダルココルチコイドで発現誘導され、 向精神薬の断薬時に 発現増強する GPCRとホモロジ一が高かった。  From the results of Example 4 (2), Examples 5 and 6, the GPCR that the protein encoded by this cDNA is expressed in the brain, induced to be expressed by dalcocorticoid, and enhanced when the psychotropic drug is stopped And homology was higher.

これらのことから、 本 c DNAがコードするタンパク質は、 ス トレスや精神刺 激剤に対する神経適応に関与すると予測された。 従って、 本タンパク質は、 薬剤 依存症からの離脱、 統合失調症、 うつ病、 不安症、 アルツハイマー病、 パーキン ソン病の治療薬の開発に有用であると考えられた。  From these facts, it was predicted that the protein encoded by this cDNA is involved in neuroadaptation to stress and psychostimulants. Therefore, this protein was considered to be useful for developing drugs for withdrawing from drug dependence, schizophrenia, depression, anxiety, Alzheimer's disease, and Parkinson's disease.

(3) dn a f o rm37953  (3) dn a f o rm37953

実施例 4 (3)、実施例 5および 6の結果から、本 cDNAがコードするタンパ ク質は、 脳に発現し、 ダルココルチコイドで発現誘導され、 向精神薬の断薬時に 発現増強する GPCRとホモロジ一が高かった。これらのこと力、ら、本 c DNAがコ 一ドするタンパク質はストレスや精神刺激剤に対する神経適応に関与すると予測 された。従って、本タンパク質は、薬剤依存症からの離脱、統合失調症、 うつ病、 不安症、 アルツハイマー病、 パーキンソン病、 等の治療薬の開発に有用であると 考えられた。  From the results of Example 4 (3), Examples 5 and 6, the protein encoded by this cDNA is expressed in the brain, is induced to be expressed by dalcocorticoid, and has a GPCR that enhances the expression when the psychotropic drug is stopped. The homology was high. For these reasons, it was predicted that the protein encoded by this cDNA would be involved in neural adaptation to stress and psychostimulants. Therefore, this protein was considered to be useful for the development of therapeutic drugs for withdrawal from drug dependence, schizophrenia, depression, anxiety, Alzheimer's disease, Parkinson's disease, and the like.

(4) d n a f o rm42851  (4) d n a f o rm42851

実施例 4 (4)、実施例 5および 6の結果から、本 c DNAがコードするタンパ ク質は、 脳に発現し、 ダルココルチコイドで発現誘導され、 向精神薬の断薬時に 発現増強する GPCRとホモロジ一が高かった。また、対照と比較して筋肉おょぴ脖 臓で発現が増加し、 小脳、 結腸、 および胎盤で発現が増加する傾向があり、 脳な どでは同程度の発現であった。 From the results of Example 4 (4), Examples 5 and 6, the protein encoded by the cDNA The protein was expressed in the brain, induced to be expressed by dalcocorticoids, and had a high homology with GPCRs, whose expression was enhanced when psychotropic drugs were stopped. In addition, the expression was increased in muscle and spleen compared to control, and increased in cerebellum, colon and placenta, and was similar in brain and the like.

これらのことから、 本 c DNAがコードするタンパク質はスストレスや精神刺 激剤に対する神経適応に関与すると思われた。 従って、 本タンパク質は、 薬剤依 存症からの離脱、 統合失調症、 うつ病、 不安症、 アルツハイマー病、 パーキンソ ン病、 糖尿病の治療薬の開発に有用であると考えられた。  These results suggest that the protein encoded by this cDNA is involved in neuroadaptation to stress and psychostimulants. Therefore, this protein was considered to be useful for the development of a drug for withdrawal from drug dependence, schizophrenia, depression, anxiety, Alzheimer's disease, Parkinson's disease, and diabetes.

(5) d n a f o rm27636  (5) d n a f o rm27636

実施例 4 (5)、実施例 5および 6の結果から、本 c DNAがコードするタンパ ク質は、 結腸に発現している GPR82と類似していた。 これらのことから、 本タン パク質は、 クローン病、 過敏性大腸炎、 消化管機能調整、 癌等に関連する機能を 有し、 これらの治療薬の開発に有用であると考えられた。  From the results of Example 4 (5), and Examples 5 and 6, the protein encoded by the present cDNA was similar to GPR82 expressed in the colon. Based on these findings, it was considered that this protein has functions related to Crohn's disease, irritable colitis, gastrointestinal tract function regulation, cancer, etc., and is useful for the development of these therapeutic agents.

(6) dn a f o rm.47734  (6) dn a f o rm. 47734

実施例 4 (6)、実施例 5および 6の結果から、本 c DNAがコードするタンパ ク質は、 ヒト TGR2 1と類似しており、 また、 脳組織特に前頭葉皮質おょぴ海 馬での発現が高く、 脾臓や脂肪でも発現が観察された。  From the results of Example 4 (6) and Examples 5 and 6, the protein encoded by the present cDNA is similar to human TGR21, and is also found in brain tissue, especially in the frontal cortex and hippocampus. High expression was observed in spleen and fat.

これらのこと力、ら、 本タンパク質は、 アルツハイマー病、 パーキンソン病、 舞 踏病、 虚血性脳疾患、 統合失調症、 うつ病、 不安症、 糖尿病に関連する機能を有 し、 これらの治療薬の開発に有用であると考えられた。  The protein has functions related to Alzheimer's disease, Parkinson's disease, chorea, ischemic brain disease, schizophrenia, depression, anxiety, and diabetes. It was considered useful for development.

(7) d n a f o rm29894  (7) d n a f o rm29894

実施例 4 (7)、実施例 5および 6の結果から、本 c DNAがコードするタンパ ク質は、 neuropeptide FF 2, neuropeptide Y2 および galanin GalRl 受容体と 膜貫通領域の相同性が高いヒト GPR 103に類似していた。 また、 大脳皮質、 脳下垂体、 視床、 視床下部、 前脳基底核、 中脳、 およぴ肺に発現しており、 膝臓 で発現が強く増加し、 脳でも増加した。  From the results of Example 4 (7) and Examples 5 and 6, the protein encoded by this cDNA is a human GPR103 with high homology between neuropeptide FF2, neuropeptide Y2 and galanin GalRl receptor and the transmembrane domain. Was similar to It was also expressed in the cerebral cortex, pituitary gland, thalamus, hypothalamus, basal forebrain, midbrain, and lungs. Expression was strongly increased in the knee and increased in the brain.

これらのこと力 ら、 本タンパク質は、 統合失調症、 うつ病、 不安症、 パーキン ソン病、 アルツハイマー病、 虚血性脳疾患、 糖尿病、 内分泌疾患に関連する機能 を有し、 これらの治療薬として有用であると考えられた。 Based on these strengths, this protein is useful for schizophrenia, depression, anxiety, parkin It has functions related to Sohn's disease, Alzheimer's disease, ischemic brain disease, diabetes, and endocrine diseases, and was considered to be useful as a therapeutic drug for these.

(8) dn a f o rm53455  (8) dn a f o rm53455

実施例 4 (8)、実施例 5および 6の結果から、本 c DNAがコードするタンパ ク質は、 neuropeptide FF 2, neuropeptide Y2 および galanin GalRl 受容体と 膜貫通領域の相同性が高いヒト GPR103に類似していた。 また、 大脳皮質、 脳下垂体、 視床、 視床下部、 前脳基底核、 中脳、 および肺に発現していた。 これらのことから、 本タンパク質は、 統合失調症、 うつ病、 不安症、 パーキン ソン病、 アルツハイマー病、 虚血性脳疾患、 糖尿病、 内分泌疾患に関連する機能 を有し、 これらの治療薬として有用であると考えられた。  From the results of Example 4 (8) and Examples 5 and 6, the protein encoded by this cDNA was converted to human GPR103, which has high homology between neuropeptide FF2, neuropeptide Y2 and the galanin GalRl receptor and the transmembrane region. Was similar. It was also expressed in the cerebral cortex, pituitary gland, thalamus, hypothalamus, basal forebrain, midbrain, and lung. From these facts, this protein has functions related to schizophrenia, depression, anxiety, Parkinson's disease, Alzheimer's disease, ischemic brain disease, diabetes, and endocrine disease, and is useful as a therapeutic drug for these. It was thought that there was.

(9) dn a f o rm53728  (9) dn a f o rm53728

実施例 4 (9)、実施例 5および 6の結果から、本 c DNAがコードするタンパ ク質は、 neuropeptide FF 2, neuropeptide Y2 および galanin GalRl 受容体と 膜貫通領域の相同性が高いヒト GPR 103に類似していた。 また、 大脳皮質、 脳下垂体、 視床、 視床下部、 前脳基底核、 中脳、 および肺に発現していた。 これらのことから、 本タンパク質は、 統合失調症、 うつ病、 不安症、 パーキン ソン病、 アルツハイマー病、 虚血性脳疾患、 糖尿病、 内分泌疾患に関連する機能 を有し、 これらの治療薬として有用であると考えられた。  From the results of Example 4 (9) and Examples 5 and 6, the protein encoded by this cDNA is a human GPR103 with high homology between neuropeptide FF2, neuropeptide Y2 and galanin GalRl receptor and transmembrane domain. Was similar to It was also expressed in the cerebral cortex, pituitary gland, thalamus, hypothalamus, basal forebrain, midbrain, and lung. From these facts, this protein has functions related to schizophrenia, depression, anxiety, Parkinson's disease, Alzheimer's disease, ischemic brain disease, diabetes, and endocrine disease, and is useful as a therapeutic drug for these. It was thought that there was.

(10) dn a f o rm62766  (10) dn a f o rm62766

実施例 4 (10)、実施例 5および 6の結果から、本 cDNAがコードするタン パク質は、 neuropeptide FF 2, neuropeptide Y2およぴ galanin GalRl 受容体 と膜貫通領域の相同性が高いヒト GPR 103に類似していた。また、大脳皮質、 脳下垂体、 視床、 視床下部、 前脳基底核、 中脳、 および橋に発現していた。 これらのことから、 本タンパク質は、 統合失調症、 うつ病、 不安症、 パーキン ソン病、 アルツハイマー病、 虚血性脳疾患、 糖尿病、 内分泌疾患に関連する機能 を有し、 これらの治療薬として有用であると考えられた。  From the results of Example 4 (10) and Examples 5 and 6, the protein encoded by the present cDNA is a human GPR having high homology with the neuropeptide FF2, neuropeptide Y2 and galanin GalRl receptor and the transmembrane region. It was similar to 103. It was also expressed in the cerebral cortex, pituitary gland, thalamus, hypothalamus, basal forebrain, midbrain, and pons. From these facts, this protein has functions related to schizophrenia, depression, anxiety, Parkinson's disease, Alzheimer's disease, ischemic brain disease, diabetes, and endocrine disease, and is useful as a therapeutic drug for these. It was thought that there was.

(1 1) d n a f o rm32567 実施例 4 (1 1)、実施例 5および 6の結果から、本 c DNAがコードするタン パク質は、 対照と比較して脖臓および脳で強く増加し、 子宮および精巣でも増加 し、 肝臓や結腸などで増加する傾向があった。 (1 1) dnafo rm32567 From the results of Example 4 (1 1) and Examples 5 and 6, the protein encoded by the present cDNA increased strongly in the kidney and brain, increased in the uterus and testis, and increased in the liver compared to the control. And the colon tended to increase.

これらのことから、 本タンパク質は、 糖尿病、 統合失調症、 うつ病、 不安症、 パーキンソン病、 アルツハイマー病、 虚血性脳疾患、 不妊.避妊、 肝疾患に関連 する機能を有し、 これらの治療薬として有用であると考えられた。  Based on these findings, this protein has functions related to diabetes, schizophrenia, depression, anxiety, Parkinson's disease, Alzheimer's disease, ischemic brain disease, infertility, contraception, and liver disease. It was considered as useful.

(12) d n a f o rm33729  (12) d n a f o rm33729

実施例 4 (1 2)、実施例 5および 6の結果から、本 c DNAがコードするタン パク質は、 ヒト AX 375432と類似しており、 AX 375432はソマトス タチンアナログの angiopeptinで活性化され、ァテローム性動脈硬化を抑制した。 また、 本タンパク質は、 眼での発現が高く、 肺、 脂肪、 脾臓で発現が観察され、 結腸癌で発現が減少した。  From the results of Example 4 (1 2), Examples 5 and 6, the protein encoded by the present cDNA is similar to human AX 375432, and AX 375432 is activated by the somatostatin analog angiopeptin, Atherosclerosis was suppressed. In addition, the expression of this protein was high in the eyes, and was observed in lung, fat, and spleen, and decreased in colon cancer.

これらのことから、 本タンパク質は、 ァテローム性動脈硬化、 癌、 糖尿病、 糖 尿病性網膜症、 眼科疾患に関連する機能を有し、 これらの治療薬の開発に有用で あると考えられた。  Based on these facts, it was considered that the present protein has functions related to atherosclerosis, cancer, diabetes, glycemic retinopathy, and ophthalmic diseases, and is useful for the development of these therapeutic agents.

(13) d n a f o r m48477  (13) d n a f o r m48477

実施例 4 (1 3)、実施例 5および 6の結果から、本 c DNAがコードするタン パク質は、 顆粒球や未分化な樹状細胞で発現する C 5 L 2や走ィヒ性に関わる GP CRと類似していた。 また、 本タンパク質は、 骨髄、 肺で発現が高く、 糖尿病の 脂肪組織で発現が増加した。  From the results of Example 4 (13) and Examples 5 and 6, the protein encoded by this cDNA was found to be resistant to C5L2 and chemotaxis expressed in granulocytes and undifferentiated dendritic cells. Similar to the GP CR involved. The expression of this protein was high in bone marrow and lung, and increased in diabetic adipose tissue.

これらのことから、本タンパク質は、免疫疾患、炎症性疾患、ァレルギ一疾患、 肺や気管等の呼吸器系疾患、 糖尿病に関連する機能を有し、 これらの治療薬の開 発に有用であると考えられた。  From these facts, this protein has functions related to immune diseases, inflammatory diseases, allergic diseases, respiratory diseases such as lungs and trachea, and diabetes, and is useful for the development of these therapeutic agents. It was considered.

(14) d n a f o rm54572  (14) d n a f o rm54572

実施例 4 (14)、実施例 5および 6の結果から、本 c DNAがコードするタン パク質は、 外来性毒 alpha - latrotoxinの受容体 Latrophilin のスプライシング バリアントである脳特異的 latrophilin 3に類似していた。 また、 本タンパク質 は、 対照と比較して膝臓、 脂肪細胞、 10日齢新生児小脳で発現が増加し、 脳な. どでは対照と同程度発現していた。 From the results of Example 4 (14), Examples 5 and 6, the protein encoded by this cDNA is similar to brain-specific latrophilin 3, which is a splicing variant of the exogenous toxin alpha-latrotoxin receptor Latrophilin. I was The protein Was increased in the knee, adipocytes, and 10-day-old neonatal cerebellum as compared to the control, and was expressed to the same extent in the brain and the like.

これらのことから、 本タンパク質は、 統合失調症、 うつ病、 不安症、 パーキン ソン病、 アルツハイマー病、 舞踏病、 糖尿病などの内分泌疾患に関連する機能を 有し、 これら治療薬の開発に有用であると考えられた。  Based on these facts, this protein has functions related to endocrine diseases such as schizophrenia, depression, anxiety, Parkinson's disease, Alzheimer's disease, chorea and diabetes, and is useful for the development of these therapeutics. It was thought that there was.

(15) d n a f o rm29069  (15) d n a f o rm29069

実施例 4 (15)、実施例 5および 6の結果から、本 c DNAがコードするタン パク質は、マウス造血幹細胞からクローニングされた Cy t 28に類似していた。 また、 類似のタンパク質である GPR56は甲状腺、 心臓、 脳に強く発現し、 脳 内では海馬、 視床に特に強く発現し、 細胞外にムチン様の構造を有することから 細胞間相互作用に関与することが示唆されている。 また、 本タンパク質は、 対照 と比較して骨およぴ胎盤で強く増加し、 膝臓およぴ筋肉などでも増加する傾向が めった。  From the results of Example 4 (15) and Examples 5 and 6, the protein encoded by the present cDNA was similar to Cyt 28 cloned from mouse hematopoietic stem cells. GPR56, a similar protein, is strongly expressed in the thyroid gland, heart, and brain, is particularly strongly expressed in the hippocampus and thalamus in the brain, and has an extracellular mucin-like structure. Has been suggested. In addition, the present protein increased strongly in bone and placenta, and tended to increase in knee and muscle compared to control.

これらのことから、 本タンパク質は、 骨粗鬆症、 糖尿病などの内分泌疾患、 統 合失調症、 うつ病、 不安症、 パーキンソン病、 アルツハイマー病、 免疫疾患、 炎 症性疾患、 アレルギー疾患に関連する機能を有し、 これらの治療薬の開発に有用 であると考えられた。  Based on these findings, this protein has functions related to endocrine diseases such as osteoporosis and diabetes, schizophrenia, depression, anxiety, Parkinson's disease, Alzheimer's disease, immune diseases, inflammatory diseases, and allergic diseases. However, it was considered useful for the development of these therapeutic agents.

(16) d n a f o rm456 16  (16) d n a f o rm456 16

実施例 4 (16)、実施例 5および 6の結果から、本 c DNAがコードするタン パク質が類似している GPR34遺伝子は X染色体上にあり、 各種臓器で豊富に 発現すること、 PAF受容体などと弱い相同性があることがわかった。  From the results of Example 4 (16) and Examples 5 and 6, the GPR34 gene, which is similar in the protein encoded by this cDNA, is on the X chromosome, and is abundantly expressed in various organs. It was found that there was weak homology with the body.

また、 本タンパク質は、 肺での発現が高く、 脖臓、 脂肪、 結腸で発現が観察され たが、 糖尿病の膝臓および結腸癌では発現が減少した。 In addition, expression of this protein was high in lung and was observed in kidney, fat and colon, but decreased in diabetic knee and colon cancer.

これらのこと力ゝら、 本タンパク質は、 呼吸器疾患 (喘息など)、 糖尿病、 癌、 等 に関連する機能を有し、 これらの治療薬の開発に有用であると考えられた。 産業上の利用可能性 本発明のタンパク質およびそれをコードする D N Aは、 前記した Gタンパク質 共役型受容体活性を有することから、 該タンパク質あるいはそれをコードする D NAを用いて該活性を調節する物質をスクリーニングすることができ、 また該タ ンパク質が関連する疾患等に作用し得る医薬の開発に用いることができる。 特に Gタンパク質共役型受容体 (G- Protein Coupled Receptor) は、 それぞれ、 細胞 外の様々なリガンドからのシグナルを、 Gタンパク質を介して伝達する膜タンパ ク質受容体である。 細胞の様々なシグナル伝達に関与しており、 医薬品開発のタ 一ゲットとして、 重要であって、 Gタンパク質共役型受容が関与するシグナル伝 達の阻害剤、活性化剤等が、多く医薬品として上巿され利用されている。従って、 新規の Gタンパク質共役型受容を見いだすことは、 医薬開発産業上、 非常に有益 である。 本出願は、 2 0 0 2年 4月 1 9日付けの日本特許出願(特願 2 0 0 2 - 1 1 7 8 1 5 )、 2 0 0 2年 1 2月 4日付けの日本特許出願(特願 2 0 0 2 - 3 5 2 2 5 2 ) に基づくものであり、その内容はここに参照として取り込まれる。 また、本明細書 にて引用した文献の内容もここに参照として取り込まれる。 From these facts, it is considered that the present protein has functions related to respiratory diseases (such as asthma), diabetes, cancer, and the like, and is useful for the development of these therapeutic agents. Industrial applicability Since the protein of the present invention and the DNA encoding the same have the above-described G protein-coupled receptor activity, a substance that regulates the activity can be screened using the protein or the DNA encoding the protein. It can also be used for the development of a medicament capable of acting on diseases associated with the protein. In particular, G-protein coupled receptors (G-protein coupled receptors) are membrane protein receptors that transmit signals from various extracellular ligands via G proteins. It is involved in various signal transduction of cells and is important as a target for drug development, and many inhibitors and activators of G protein-coupled receptor-related signal transduction are listed as drugs.巿 has been used. Therefore, finding a novel G protein-coupled receptor is very useful in the pharmaceutical development industry. This application is for a Japanese patent application filed on April 19, 2012 (Japanese Patent Application No. 2002-111), and for a Japanese patent application filed on February 1, 2002 (Japanese Patent Application No. 2002-3502252), the contents of which are incorporated herein by reference. The contents of the documents cited in this specification are also incorporated herein by reference.

Claims

匚 =f求の範囲 匚 = range of f 1. 以下の (a) または (b) のタンパク質。  1. The following protein (a) or (b): (a)配列番号 17〜32のいずれかに記載のアミノ酸配列からなるタンパク質。 ( b ) 配列番号 1 7〜 32のいずれかに記載のァミノ酸配列において 1若しくは 数個のアミノ酸が欠失、 置換及び Zまたは付加されたアミノ酸配列からなり、 か つ Gタンパク質共役型受容体活性を有するタンパク質。  (a) a protein comprising the amino acid sequence of any one of SEQ ID NOs: 17 to 32; (b) one or more amino acids in the amino acid sequence of any one of SEQ ID NOs: 17 to 32 comprising deletion, substitution, Z or addition of an amino acid sequence, and G protein-coupled receptor activity A protein having 2. 請求項 1に記載のタンパク質をコードする DNA。  2. A DNA encoding the protein of claim 1. 3. 請求項 1に記載のタンパク質をコードする完全長 c DNA。  3. A full-length cDNA encoding the protein of claim 1. 4. 以下の (a) 、 (b)又は (c) の何れかの DNA。  4. DNA of any of the following (a), (b) or (c): (a) 配列番号 1〜16のいずれかに記載の塩基配列を有する DNA。  (a) a DNA having the nucleotide sequence of any one of SEQ ID NOs: 1 to 16; (b) 配列番号 1〜16のいずれかに記載の塩基配列において、 1若しくは数個 の塩基が欠失、 置換及び/または付加された塩基配列を有し、 かつ Gタンパク質 共役型受容体活性を有するタンパク質をコードする DNA。  (b) in the nucleotide sequence of any one of SEQ ID NOs: 1 to 16, having one or several nucleotides deleted, substituted and / or added, and having a G protein-coupled receptor activity DNA encoding a protein having (c) 配列番号 1〜16のいずれかに記載の塩基配列を有する DN Aあるいはそ の相補配列とス トリンジェントな条件下でハイブリダイズすることができる塩基 配列を有し、 かつ Gタンパク質共役型受容体活性を有するタンパク質をコードす る DNA。  (c) having a nucleotide sequence capable of hybridizing under stringent conditions to DNA having the nucleotide sequence of any one of SEQ ID NOs: 1 to 16 or a complementary sequence thereof, and a G protein-coupled type DNA encoding a protein having receptor activity. 5. 請求項 2〜 4のいずれかに記載の D N Aを含む組換えべクター。  5. A recombinant vector containing the DNA according to any one of claims 2 to 4. 6. 請求項 2〜 4のいずれかに記載の D N Aまたは請求項 5に記載の組み換 えベクターを導入した遺伝子導入細胞または該細胞からなる個体。  6. A gene-transfected cell into which the DNA according to any one of claims 2 to 4 or the recombinant vector according to claim 5 has been introduced, or an individual comprising the cell. 7. 請求項 6に記載の細胞により産生される、請求項 1に記載のタンパク質。 7. The protein of claim 1, which is produced by the cell of claim 6. 8. 請求項 2〜4のいずれかに記載の DN Aの塩基配列中の連続した 5〜 1 00塩基と同じ配列を有するセンスオリゴヌクレオチド、 当該センスオリゴヌク レオチドと相補的な配列を有するアンチセンスオリゴヌクレオチド、 及び、 当該 センス又はアンチセンスオリゴヌクレオチドのオリゴヌクレオチド誘導体から成 る群から選ばれるオリゴヌクレオチド。 8. A sense oligonucleotide having the same sequence as 5 to 100 consecutive nucleotides in the nucleotide sequence of DNA according to any one of claims 2 to 4, an antisense having a sequence complementary to the sense oligonucleotide. An oligonucleotide selected from the group consisting of an oligonucleotide and an oligonucleotide derivative of the sense or antisense oligonucleotide. 9 . 請求項 1または 7に記載のタンパク質に特異的に結合する抗体あるいは その部分フラグメント。 9. An antibody or a partial fragment thereof that specifically binds to the protein of claim 1 or 7. 1 0 . 抗体がモノクローナノレ抗体である請求項 9に記載の抗体。  10. The antibody according to claim 9, wherein the antibody is a monoclonal antibody. 1 1 . モノクローナル抗体が請求項 1または 7に記載のタンパク質の Gタン パク質共役型受容体活性を中和する作用を有することを特徴とする請求項 1 0に 記載の 体。  11. The body according to claim 10, wherein the monoclonal antibody has an action of neutralizing the G protein-coupled receptor activity of the protein according to claim 1 or 7. 1 2 . 請求項 1または 7に記載のタンパク質と被検物質を接触させ、 該被検 物質による該タンパク質が有する活性の変化を測定することを特徴とする、 該タ ンパク質の活性調節物質のスクリーニング方法。  12. The method according to claim 1, wherein the protein according to claim 1 or 7 is brought into contact with a test substance, and a change in the activity of the protein caused by the test substance is measured. Screening method. 1 3 . 請求項 6に記載の遺伝子導入細胞と被検物質を接触させ、 該細胞に導 入されている D NAの発現レベルの変化を検出することを特徴とする、 該 D NA の発現調節物質のスクリーニング方法。  13. A method for regulating the expression of a DNA, comprising bringing the gene-introduced cell according to claim 6 into contact with a test substance and detecting a change in the expression level of the DNA introduced into the cell. A method for screening substances. 1 4 . 請求項 1に記載のタンパク質のアミノ酸配列から選択される少なくと も 1以上のァミノ配列情報おょぴ Zまたは請求項 2 〜 4のいずれかに記載の D N Aの塩基配列から選択される少なくとも 1以上の塩基配列情報を保存したコンビ ユータ読み取り可能記録媒体。  14. At least one or more amino acid sequence information selected from the amino acid sequence of the protein according to claim 1 or selected from the DNA base sequence according to any one of claims 2 to 4. A computer-readable recording medium that stores at least one or more base sequence information. 1 5 . 請求項 1に記載のタンパク質および/または請求項 2〜 4のいずれか に記載の D N Aを結合させた担体。  15. A carrier to which the protein according to claim 1 and / or the DNA according to any one of claims 2 to 4 are bound.
PCT/JP2003/004982 2002-04-19 2003-04-18 Novel proteins and dnas encoding the same Ceased WO2003089643A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2003227427A AU2003227427A1 (en) 2002-04-19 2003-04-18 Novel proteins and dnas encoding the same

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
JP2002117815 2002-04-19
JP2002-117815 2002-04-19
JP2002-352252 2002-12-04
JP2002352252 2002-12-04

Publications (1)

Publication Number Publication Date
WO2003089643A1 true WO2003089643A1 (en) 2003-10-30

Family

ID=29253602

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2003/004982 Ceased WO2003089643A1 (en) 2002-04-19 2003-04-18 Novel proteins and dnas encoding the same

Country Status (2)

Country Link
AU (1) AU2003227427A1 (en)
WO (1) WO2003089643A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002014489A2 (en) * 2000-08-10 2002-02-21 Amgen, Inc. Leucine-rich repeat-containing g-protein coupled receptor-8 molecules and uses thereof
WO2002026824A2 (en) * 2000-09-27 2002-04-04 Bristol-Myers Squibb Company A novel human g-protein coupled receptor, hgprbmy5, expressed highly in brain and ovarian tissues

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002014489A2 (en) * 2000-08-10 2002-02-21 Amgen, Inc. Leucine-rich repeat-containing g-protein coupled receptor-8 molecules and uses thereof
WO2002026824A2 (en) * 2000-09-27 2002-04-04 Bristol-Myers Squibb Company A novel human g-protein coupled receptor, hgprbmy5, expressed highly in brain and ovarian tissues

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
OVERBEEK P.A. ET AL.: "A transgenic insertion causing cryptor-chidism in mice", GENESIS, vol. 30, no. 1, 2001, pages 26 - 35, XP002196450 *
THE RIKEN GENOME EXPLORATION RESEARCH GROUP PHASE II TEAM AND THE FANTOM CONSORTIUM: "Functional annotation of a full-length mouse cDNA collection", NATURE, vol. 409, 2001, pages 685 - 690, XP001009930 *

Also Published As

Publication number Publication date
AU2003227427A8 (en) 2003-11-03
AU2003227427A1 (en) 2003-11-03

Similar Documents

Publication Publication Date Title
US9568482B2 (en) Antibodies specific for ZNT8
WO2000075314A1 (en) Novel hemopoietin receptor protein nr10
JP2001211885A (en) Novel polypeptide
WO2003089643A1 (en) Novel proteins and dnas encoding the same
JP2004505648A (en) Inflammation-related G protein-coupled receptor
WO2003091435A1 (en) Novel proteins and dnas encoding the same
JP2004229641A (en) Novel protein and DNA encoding it
US6803184B1 (en) MPR-related ABC transporter encoding nucleic acids and methods of use thereof
TW200532019A (en) Nucleic acid encoding a novel DP receptor protein and methods of use thereof
JP2004229653A (en) Novel protein and DNA encoding it
WO2003089646A1 (en) Novel proteins and dnas encoding the same
JP2004229648A (en) Novel protein and DNA encoding it
Wright et al. Cloning strategies for peptide hormone receptors
JP2004041181A (en) Novel protein and DNA encoding it
JP2004229651A (en) Novel protein and DNA encoding it
JP2004229644A (en) Novel protein and DNA encoding it
JP2004229646A (en) Novel protein and DNA encoding it
WO2003089466A1 (en) Novel proteins and dnas encoding the same
US20020155473A1 (en) Methods for identifying G-protein coupled receptors associated with diseases
JP2004041178A (en) Novel protein and DNA encoding it
WO2003091428A1 (en) Novel proteins and dnas encoding the same
JP2004229650A (en) Novel protein and DNA encoding it
JP2004229649A (en) Novel protein and DNA encoding it
JP2004041180A (en) Novel protein and DNA encoding it
JP2004229645A (en) Novel protein and DNA encoding it

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 CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS KE KG KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PH PL PT RO RU SC SD SE SG SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

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

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