WO2001066720A1 - Mouse adipocyte-origin genes - Google Patents

Mouse adipocyte-origin genes Download PDF

Info

Publication number
WO2001066720A1
WO2001066720A1 PCT/JP2001/001863 JP0101863W WO0166720A1 WO 2001066720 A1 WO2001066720 A1 WO 2001066720A1 JP 0101863 W JP0101863 W JP 0101863W WO 0166720 A1 WO0166720 A1 WO 0166720A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
dna
present
seq
amino acid
Prior art date
Application number
PCT/JP2001/001863
Other languages
French (fr)
Japanese (ja)
Inventor
Toshio Kitamura
Hiromichi Tsuruga
Original Assignee
Toshio Kitamura
Hiromichi Tsuruga
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 Toshio Kitamura, Hiromichi Tsuruga filed Critical Toshio Kitamura
Priority to AU2001241080A priority Critical patent/AU2001241080A1/en
Publication of WO2001066720A1 publication Critical patent/WO2001066720A1/en

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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals

Definitions

  • the present invention relates to a novel protein derived from an adipocyte and having a signal sequence, a gene thereof, and their production and use.
  • Adipose tissue is simply fat-free until it is known that the mutation causes hereditary obesity in mice (Zhang, Y. et al. (1994) Nature 372, 425-432). It was thought to be just a storage. At present, adipose tissue is recognized as the largest secretory organ.
  • TNF-chicken plasminogen activator inhibitor PAI-1
  • TNF-H affects the metabolism of lipids and darcos and is overexpressed in adipose tissue of genetically obese rodents (Hotamisligil, GS et al. (1993) Science 259, 87-91 ), Leads to insulin resistance (Hotamisligil 3 GS et al. (1995) J. Clin. Invest. 95, 240 9-2415)
  • PAI-1 is highly expressed in visceral adipocytes and It has been implicated in the onset of the disease (Shimomura J. et al. (1996) Nature medicine 2, 800-803).
  • the present invention provides novel proteins derived from adipocytes and having a signal sequence, their genes, molecules functionally equivalent thereto, and their production and use.
  • 3T3-L1 cell line (Meuth 5 M. and Green, H. (1974) Cell 3, 367-A) which is a well-characterized model for adipocyte differentiation. 374; Green 5 H. and Kehinde 3 0. (1975) Cell 5, a cDNA library prepared from 19-27), retrovirus-mediated expression cloning system (. Kojima, T and K itamura 3 T. (1999) Nature Biotechnol.17, 487-490), and has a signal peptide (von Heijne, (1985) J. Mol. Biol. 184, 99-105) using an efficient signal sequence trapping method. Molecular screening was performed.
  • a cDNA encoding a protein capable of expressing MPL on the cell surface is searched for by screening a library that expresses a fusion protein with the constitutively active cytokine receptor MPL.
  • the target clone can be easily selected because the expression of the autonomic proliferation ability to an IL-3-dependent cell line by expression of MPL on the cell surface is used as an index.
  • genes showed expression associated with adipocyte differentiation.
  • the expressed tissues were different, they showed tissue-specific expression. Therefore, although these genes have different functional sites, they are thought to be molecules that play a role related to adipocyte differentiation in vivo.
  • the present invention relates to a novel protein derived from an adipocyte and having a signal sequence, a gene thereof, a molecule functionally equivalent thereto, and production and use thereof, and more specifically,
  • a DNA according to any of the following (a) to (f), (a) a DNA encoding a protein consisting of the amino acid sequence of any one of SEQ ID NOs: 8 to 11;
  • a host cell carrying the DNA of (1) or (2) or the vector of (4) (6)
  • the method for producing the protein or peptide according to (3) comprising culturing the host cell according to (5) and recovering the expressed protein from the host cell or a culture supernatant thereof.
  • a polynucleotide comprising at least 15 nucleotides complementary to a DNA consisting of the nucleotide sequence of any one of SEQ ID NOs: 1 to 7 or a complementary strand thereof, and
  • the present invention provides a gene encoding a novel protein derived from an adipocyte and having a signal sequence.
  • the present inventors searched for fat cell-derived cDNAs encoding secretory and membrane proteins by the recently established novel signal sequence trapping method, SST-REX method, and found 63 known cDNAs and 8 novel cDNAs. Got a piece.
  • novel cDNAs isolated by the present inventors using the SST-REX method seven cDNAs (these clones were named “# 101” to “# 107”, respectively. Are collectively referred to as "# 10X.")
  • the nucleotide sequences of The amino acid sequence of the protein encoded by the # 103 to # 106 cDNA is shown in SEQ ID NOs: 8-11.
  • Collagen sph-1 (IV) and sph-2 (IV) synthesis and secretion are negligible in fibroblasts, but markedly increased in adipocytes. Enhancement of endocin secretion was also observed (Aratani, Y. and Kitagawa 3 Y. (1988) J. Biol. Chem. 263, 16163-16169). The results shown by the present inventors are consistent with these results, and suggest that the synthesis of extracellular matrix proteins is important for adipose tissue morphogenesis.
  • LPL lipoprotein lipase
  • the level of clone # 106 mRNA increased during differentiation.
  • this may be related to adipocyte differentiation.
  • the expression of clone # 105 is interesting. Two transcripts were detected by Northern blot analysis. The two transcripts can be from different genes.
  • this gene may be involved in insulin-mediated signal transduction, which has important implications for the treatment and prevention of diabetes and other diseases. There may be.
  • the nucleotide sequence of the human homolog of the # 104 gene has been registered in the gene bank, and according to the description, the expression is enhanced by canceration in the liver system. Expression of # 104 was observed in various cancer cell lines, and their expression patterns were different. Depending on the type of cancer, the expression of the # 104 gene may or may not be seen, and there may be some causal relationship between the # 104 gene and cancer.
  • # 101, key, and # 107 are not full-length sequences.
  • those skilled in the art will Techniques for isolating full-length genes have become common technical knowledge. For example, those skilled in the art can isolate the full-length gene by performing colony hybridization using these fragments as probes. Therefore, the present invention includes full-length genes corresponding to these fragments.
  • the present invention also provides a functionally equivalent to # 10X protein (a protein consisting of the amino acid sequence of SEQ ID NOs: 8 to 11; a protein encoded by full-length MA corresponding to SEQ ID NOs: 1, 2, and 7).
  • # 10X protein a protein consisting of the amino acid sequence of SEQ ID NOs: 8 to 11; a protein encoded by full-length MA corresponding to SEQ ID NOs: 1, 2, and 7.
  • proteins include, for example, mutants of these proteins, homologs of organisms other than mice, and the like.
  • “functionally equivalent” means that the target protein has a function as a protein having a signal sequence, like the # 10X protein.
  • a function for example, a function described in International Publication No. WO 00/00610 or a function as a secretory protein can be considered. Whether or not the target protein is a secretory protein can be determined by the following method.
  • a fusion gene in which a gene encoding a commercially available peptide (eg, His-tag or FLAG) is ligated to the 3rd end of the # 10X gene is prepared.
  • the fusion gene is introduced into animal cells (eg, COS cells, etc.) using an expression vector for animal cells (eg, pcDNA3 or pCOS-1), and the # 10X protein is converted as a fusion protein with the peptide. To be expressed. Whether or not this fusion protein is secreted into the culture supernatant is evaluated by ELISA, Western blotting or immunoprecipitation using an antibody against the peptide.
  • Secretory protein S has various industrial advantages. For example, when it is desired to obtain a certain recombinant protein, if the protein is expressed in a cell as a fusion protein with a secretory protein or a partial peptide capable of secreting the same, the fusion protein is secreted out of the cell. However, there is an advantage that the purification of the recombinant protein is facilitated. In addition, since many secreted proteins are useful drugs, they can be applied as drugs themselves.
  • the protein of the present invention was derived from adipocytes and showed expression associated with adipocyte differentiation. Accordingly, the gene or protein of the present invention, or a compound that regulates the expression or activity of the protein, is not limited thereto. For example, for the treatment or prevention of diseases such as obesity, hyperlipidemia, diabetes, and arteriosclerosis. The application of is considered.
  • a method of introducing a mutation into a protein is known.
  • those skilled in the art can use a site-directed mutagenesis method (Hashimoto-Gotoh 5 T. et al. (1995) Gene 152, 271-275, Zoller, MJ, and Smith, M. (1983) Methods Enzymol. 100).
  • 468-500 Kramer, W. et al. (1984) Nucleic Acids Res. 12, 944 9456, Kramer W, and Fritz HJ (1987) Methods.Enzymol. 154, 350-367, Kunkel, TA (1985) ) Proc Natl Acad Sci USA.
  • a protein functionally equivalent to the protein can be prepared by appropriately mutating amino acids of the protein (a protein encoded by full-length DNA corresponding to SEQ ID NOs: 1, 2, and 7). Amino acid mutations can also occur in nature.
  • a protein having an amino acid sequence in which one or more amino acids are mutated in the amino acid sequence of the # 10X protein and functionally equivalent to the protein is also included in the protein of the present invention.
  • the number of amino acids to be mutated in such a mutant is usually within 50 amino acids, preferably within 30 amino acids, and more preferably within 10 amino acids (eg, within 5 amino acids).
  • the amino acid residue to be mutated is mutated to another amino acid that preserves the properties of the amino acid side chain.
  • the properties of amino acid side chains include hydrophobic amino acids (A, I, L, M, F, P, W, Y, V) and hydrophilic amino acids (R, D, N, C, E, QG, H, K, S, T), an amino acid having an aliphatic side chain (A, V, L, I, P), an amino acid having a hydroxyl group-containing side chain (S, ⁇ , ⁇ ), an amino acid having a sulfur atom-containing side chain Acids ( ⁇ ), carboxylic acids and amino acids with amide-containing side chains (D, N, E, Q ), Amino acids having base-containing side chains (R, K, ⁇ ), and amino acids having aromatic-containing side chains (H, F, Y,). Character mark).
  • the fusion protein is a fusion of these proteins with another peptide or protein, and is included in the present invention.
  • the fusion protein is prepared by encoding the # 10X protein (a protein consisting of the amino acid sequence of SEQ ID NOs: 8 to 11; a protein encoded by the full-length DNA corresponding to SEQ ID NOs: 1, 2, and 7).
  • DNA and DNA encoding another peptide or protein may be ligated in frame so that they are introduced into an expression vector and expressed in a host, and a method known to those skilled in the art can be used.
  • Other peptides or proteins to be fused with the protein of the present invention are not particularly limited.
  • peptides to be fused to the protein of the present invention include, for example, FLAG (H ⁇ , ⁇ . ⁇ . et al., BioTechnology (1988) 6, 1204-1210) and 6 His (histidine).
  • Residues consisting of 6 xHis,! O xHiS influenza agglutinin (HA), human cmyc fragment, VSV-GP fragment, pl8HIV fragment, T7-tag, HSV-tag, E-tag, SV40T antigen
  • peptides such as fragments, lck tags, a-tubulin fragments, B-tags, and Protein C fragments can be used.
  • proteins to be fused with the protein of the present invention include, for example, GST (Gluythione-S-transferase), H A (influenza agglutinin), immunoglobulin constant region, ⁇ -galactosidase, MBP (maltose binding protein) and the like.
  • a fusion protein can be prepared by fusing commercially available MA encoding the peptide or the protein with DNA encoding the protein of the present invention, and expressing the fusion DNA prepared thereby. .
  • proteins in which a plurality of amino acid residues are deleted from the amino acid sequence of the # 10X protein include proteins in which signal sequences have been removed from these proteins.
  • the present invention includes proteins encoded by DNA that hybridizes to MA encoding the protein and functionally equivalent to the # 10X protein.
  • proteins include, for example, homologs of mouse and other mammals (eg, proteins encoded by humans, rodents, rabbits, magpies, etc.).
  • Hybridization conditions for isolating MA encoding a protein functionally equivalent to the # 10X protein can be appropriately selected by those skilled in the art.
  • the conditions for the hybridization include, for example, low stringent conditions.
  • the low stringent conditions are, for example, conditions of 42 ° C and 0.1% SDS of O.l SSC in washing after hybridization, preferably 50 ° C, 0.1 x SSC and 0.1% SDS. Is the condition.
  • More preferable hybridization conditions include high stringent conditions. Highly stringent conditions are, for example, conditions of 65 ° C., 5 ⁇ SSC and 0.1% SDS. In these conditions Thus, it can be expected that DNA with higher homology can be obtained more efficiently as the temperature is increased. However, a plurality of factors such as temperature and salt concentration can be considered as factors affecting the stringency of the hybridization, and those skilled in the art can realize similar stringency by appropriately selecting these factors. It is possible to
  • a gene amplification method using primers synthesized based on the sequence information of DNA (SEQ ID NOS: 1 to 7) encoding the # 10X protein instead of the hybridization method, for example, polymerase chain reaction (PCR) method It can also be used for isolation.
  • PCR polymerase chain reaction
  • Proteins functionally equivalent to the # 10X protein encoded by DNA isolated by these hybridization techniques or gene amplification techniques are usually the # 10X protein (the amino acid sequence described in SEQ ID NOs: 8 to 11).
  • the proteins of the present invention also include proteins that are functionally equivalent to the # 10X protein and that have high homology to the amino acid sequence of the protein. High homology usually means at least 50% identity, preferably 75% identity, more preferably 85% identity, and even more preferably 95% identity at the amino acid level. .
  • the identity of amino acid sequences and nucleotide sequences can be determined by the algorithm BLAST (Proc.
  • the protein of the present invention may differ in amino acid sequence, molecular weight, isoelectric point, presence / absence and form of sugar chains, etc., depending on the cell, host, or purification method for producing the protein described below. However, as long as the obtained protein has a function equivalent to that of the # 10X protein, it is included in the present invention.
  • a prokaryotic cell for example, Escherichia coli
  • a methionine residue is added to the N-terminal of the amino acid sequence of the original protein.
  • the proteins of the present invention also include such proteins.
  • the protein of the present invention can be prepared as a recombinant protein or a natural protein by methods known to those skilled in the art. If it is a recombinant protein, a DNA encoding the protein of the present invention (for example, a DNA having the nucleotide sequence of SEQ ID NOS: 1 to 7) is inserted into an appropriate expression vector and introduced into an appropriate host cell. The resulting transformant is collected and an extract is obtained, which is then subjected to chromatography such as ion exchange, reverse phase, gel filtration, or affinity chromatography in which an antibody against the protein of the present invention is immobilized on a column.
  • chromatography such as ion exchange, reverse phase, gel filtration, or affinity chromatography in which an antibody against the protein of the present invention is immobilized on a column.
  • the protein of the present invention can be used as a fusion protein with glutathione S-transferase protein or histidine.
  • host cells eg, animal cells, E. coli, etc.
  • the set seen recombinant protein expressed can and child purified using Guru evening Chionkaramu or nickel column. After purification of the fusion protein, if necessary, regions of the fusion protein other than the target protein can be cleaved with thrombin or Factor Xa and removed.
  • the protein is a natural protein, a method known to those skilled in the art, for example, an affinity in which an antibody that binds to the protein of the present invention described below is bound to a tissue or cell extract expressing the protein of the present invention, as described below. Isolation can be achieved by purifying using one column. it can.
  • the antibody may be a polyclonal antibody or a monoclonal antibody.
  • the present invention also includes partial peptides of the protein of the present invention.
  • the partial peptide of the present invention has an amino acid sequence of at least 7 amino acids or more, preferably 8 amino acids or more, and more preferably 9 amino acids or more.
  • the partial peptide can be used, for example, for preparing an antibody against the protein of the present invention, for screening a compound binding to the protein of the present invention, and for screening for a promoter or inhibitor of the protein of the present invention. Further, it can be an antagonist of the protein of the present invention or a competitive inhibitor.
  • the partial peptide of the present invention can be produced by a genetic engineering technique, a known peptide synthesis method, or by cleaving the protein of the present invention with an appropriate peptidase.
  • the peptide may be synthesized by, for example, either a solid phase synthesis method or a liquid phase synthesis method.
  • the DNA encoding the protein of the present invention is used for in vivo and in vitro production of the protein of the present invention as described above, and also includes, for example, diseases caused by abnormalities in the gene encoding the protein of the present invention. And gene therapy for diseases treatable by the protein of the present invention.
  • the DNA of the present invention may be in any form as long as it can encode the protein of the present invention. That is, it does not matter whether it is cDNA synthesized from mRNA, genomic DNA, or chemically synthesized DNA. Also, as long as it can encode the protein of the present invention, MA having an arbitrary nucleotide sequence based on the degeneracy of the genetic code is included.
  • the DNA of the present invention can be prepared by a method known to those skilled in the art. For example, it is prepared by preparing a cDNA library from cells expressing the protein of the present invention and performing hybridization using a part of the sequence of the DNA of the present invention (for example, SEQ ID NOS: 1 to 7) as a probe. it can.
  • the cDNA library may be prepared, for example, by the method described in the literature (Sambrook, J. et al., Molecular Clonings Cold Spring Harbor Laboratory Press (1989)), or using a commercially available DNA library. Good.
  • oligo DNA is synthesized based on the sequence of the DNA of the present invention (for example, SEQ ID NOS: 1 to 7), and the PCR reaction is performed using this as a primer. It can also be prepared by amplifying a cDNA encoding the protein of the present invention.
  • Genomic DNA can be isolated by screening a genomic DNA library using the obtained cDNA as a probe.
  • mRNA is isolated from a cell, tissue, or organ that expresses the protein of the present invention (eg, an adipocyte or a tissue in which expression has been observed by Northern blotting in this example).
  • mRNA can be isolated by known methods, for example, guanidine ultracentrifugation (Chirgwin, JM et al., Biochemistry (1979) 18, 5294-5299), and AGPC method (Chomczynski, P. and Sacchi, N. 3 Anal.
  • Total RNA is prepared by Biochem. (1987) 162, 156-159) and the like, and mRNA is purified from the total RNA using the mRNA Purification Kit (Pharmacia).
  • mRNA can be directly prepared by using the QuickPrep mRNA Purification Kit (Pharmacia).
  • CDNA is synthesized from the obtained mRNA using reverse transcriptase. cDNA synthesis, AMV
  • Reverse Transcriptase First-strand cDNA Synthesis Kit (Seikagaku Corporation) can also be used.
  • the 5, -RACE method (Frohman, MA) using the 5, -Ampli FINDER RACE Kit (manufactured by Clontech) and the polymerase chain reaction (polymerase chain reaction; PCR).
  • cDNA can be synthesized and amplified.
  • a target DNA fragment is prepared from the obtained PCR product, and ligated to a vector DNA. Furthermore, a recombinant vector is prepared from this, introduced into E. coli, etc., and colonies are selected. To prepare the desired recombinant vector.
  • the base sequence of the target DNA can be confirmed by a known method, for example, the dideoxynucleotide chain termination method.
  • a nucleotide sequence with higher expression efficiency can be designed in consideration of the codon usage of the host used for expression (Grantham, R. et al. 3 Nucelic Acids Research ( 1981) 9, r43-74).
  • the MA of the present invention can be modified by a commercially available kit or a known method. Modifications include, for example, digestion with restriction enzymes, insertion of synthetic oligonucleotides or appropriate DNA fragments, addition of a linker, insertion of an initiation codon (ATG) and / or stop codon (TAA, TGA, or TAG). And the like.
  • the DNA of the present invention is a DNA consisting of a base sequence from base c at position 1 to base g at position 165 in the base sequence of SEQ ID NO: 1, or a DNA containing the base sequence, A DNA consisting of a base sequence from the 1st base to the 438th base t in the base sequence or a DNA containing the base sequence, and a base sequence from the 83rd base a to the 1408th base a in the base sequence of SEQ ID NO: 3 DNA consisting of a base sequence or DNA containing said base sequence, DNA consisting of a base sequence from base a at position 214 to base g at position 1662 in base sequence of SEQ ID NO: 4, or DNA containing said base sequence, SEQ ID NO: A DNA consisting of the base sequence from the base a at position 289 to the base c at position 1407 in the base sequence of 5, or a DNA containing the base sequence, and the base a from position 86 to position 3244 of base 86 at position 86 in the base
  • DNA consisting of a base sequence up to base a or Nucleotide sequence including MA encompasses DNA containing the DNA or nucleotide sequence consisting of the nucleotide sequence of the 7 from nucleotide position 1 of the nucleotide c to 522-positional base c.
  • the DNA of the present invention also includes a DNA that hybridizes with the MA having the nucleotide sequence shown in SEQ ID NOs: 1 to 7, and that encodes a protein functionally equivalent to the protein of the present invention.
  • the conditions for the hybridization can be appropriately selected by those skilled in the art. Specifically, the conditions described above can be used. You. Under these conditions, DNA with higher homology can be obtained as the temperature is increased.
  • the hybridizing DNA is preferably a naturally occurring DNA, for example, cDNA or chromosomal DNA.
  • the present invention also provides a vector into which the MA of the present invention has been inserted.
  • the vector of the present invention is useful for retaining the DNA of the present invention in a host cell or expressing the protein of the present invention.
  • the vector when E. coli is used as a host, the vector
  • E.g., DH9, DH5, Ver.1, XLlBlue for large-scale amplification and preparation of large amounts of E. coli for ⁇ ori '' amplification and selection of transformed E. coli
  • a gene for example, a drug resistance gene that can be identified by any drug (ampicillin, tetracycline, kanamycin, chloramphenicol)
  • vectors include M13-based vectors, pUC-based vectors, pBR322. PBluescript, pCR-Script, and the like.
  • examples include pGEM-T, pDIRECT, pT7, and the like.
  • an expression vector is particularly useful.
  • the expression vector may have the above-mentioned characteristics such that the vector is amplified in E. coli, and may be used in a host such as JM109, DH5a, HB101, or XU-Blue.
  • a promoter that can be efficiently expressed in Escherichia coli for example, lacZ promoter, Yuichi (Ward et al., Nature (1989) 341,
  • the vector may also include a signal sequence for polypeptide secretion.
  • the pelB signal sequence Lei, SP et al J. Bacteriol. (1987) 169, 4379
  • the introduction of the vector into the host cell can be performed using, for example, a calcium chloride method or an electroporation method.
  • a mammalian expression vector for example, pcDNA3 (manufactured by Invitrogen) or pEGF-BOS (Nucleic Acids. Res.
  • insect cells eg, “Bac-to-BAC baculovairus expression systemj (manufactured by Gibco BRL), pBacPAK8), plant-derived expression vector ( For example, ⁇ 1, pMH2), an expression vector derived from an animal virus (eg, pHSV, pMV, pAdexLcw), an expression vector derived from a retrovirus (eg, pZIPneo), an expression vector derived from a yeast (eg, rpichia Expression Kitj (Invitrogen) ), PNVU, SP-Q01) and Bacillus subtilis-derived expression vectors (eg, pPL608, pKTH50).
  • insect cells eg, “Bac-to-BAC baculovairus expression systemj (manufactured by Gibco BRL), pBacPAK8), plant-derived expression vector ( For example, ⁇ 1, pMH2), an expression vector derived from an animal virus (eg, pH
  • promoters necessary for expression in cells such as the SV40 promoter (Muligan et al., Nature (1979) 277, 108) ), MMLV-LTR promoter, EF1 promoter Yuichi (Mizushima et al., Nucleic Acids Res. (1990) 18, 5322), CMV promoter — It is essential to have a promoter and transform cells. It is more preferable to have a gene (for example, a drug resistance gene that can be discriminated by a drug (neomycin, G418, etc.)) for selecting Cp. Examples of vectors with such characteristics include, p negation, pDR2, pBK-RSV, can be mentioned up such pBK-CMV ⁇ p0PRSV s p0P13.
  • a vector having a DHFR gene complementing the nucleic acid synthesis pathway-deficient CH0 cell is used.
  • pCHOI pCHOI
  • methotrexe MTX
  • replication of SV40 is performed using COS cells having a gene expressing the SV40 T antigen on the chromosome. Transformation can be performed using a single vector (eg, pcD) having an origin.
  • pcD a single vector having an origin.
  • the replication origin those derived from poliovirus, adenovirus, sipapirovirus (BPV) and the like can also be used.
  • the expression vector is used as a selection marker for amplification of gene copy number in the host cell system, such as aminoglycoside transferase (APH) gene, thymidine kinase (TK) gene, and Escherichia coli xanthinguanine phosphoribosyltransferase (Ecogp t). Gene, dihydrofolate reductase (dhfr) gene and the like.
  • the DNA of the present invention is incorporated into an appropriate vector, for example, by a retrovirus method, a ribosome method, a cationic ribosome method, an adenovirus method, or the like.
  • a retrovirus method for introduction into a living body.
  • the vector used include, but are not limited to, an adenovirus vector (eg, pAdexlcw) and a retrovirus vector (eg, pZIPneo).
  • General genetic manipulations such as insertion of the DNA of the present invention into a vector can be performed according to a conventional method (Molecular Cloning, 5.61-5.63).
  • Administration into a living body may be an ex vivo method or an in vivo method.
  • the present invention also provides a host cell into which the vector of the present invention has been introduced.
  • the host cell into which the vector of the present invention is introduced is not particularly limited, and for example, Escherichia coli and various animal cells can be used.
  • the host cell of the present invention can be used, for example, as a production system for producing or expressing the protein of the present invention.
  • Production systems for protein production include in vitro and in vivo production systems. Examples of in vitro production systems include production systems using eukaryotic cells and production systems using prokaryotic cells.
  • eukaryotic cells for example, animal cells, plant cells, and fungal cells can be used as hosts.
  • mammalian cells for example, CHO (J. Exp. Med.
  • CH0 cells in particular, dhfr-CHO (Proc. Natl. Acad. Scad. USA (1980) 77, 4216-4220) and CHO K-1 (Proc. Natl. Acad. Sci. USA (1968) 60, 1275) can be suitably used.
  • CH0 cells are particularly preferred.
  • Vector vectors can be introduced into host cells by, for example, the calcium phosphate method, the DEAE dextran method, the method using Cationic Ribosome D0TAP (manufactured by Behringer Mannheim), the electoral poration method, or the Lipofexion method. Is possible.
  • Nicotiana tabacum As plant cells, for example, cells derived from Nicotiana tabacum (Nicotiana tabacum) are known as protein production systems, which may be callus cultured.
  • Fungal cells include yeast, for example, the genus Saccharomyces, for example, Saccharomyces cerevisiae, filamentous fungi, for example, the genus Aspergillus, for example, Aspergillus niger s. are known.
  • bacterial cells When prokaryotic cells are used, there are production systems using bacterial cells.
  • bacterial cells include Escherichia coli (E. coli) such as JM109, DH5 and HB101, and Bacillus subtilis.
  • the protein is obtained by transforming these cells with the target DNA and culturing the transformed cells in vitro.
  • the culturing can be performed according to a known method.
  • a culture solution of animal cells for example, DMEM, MEM, RPMI1640, and IMDM can be used.
  • serum replacement fluid such as fetal calf serum (FCS) They may be used in combination, or may be serum-free culture.
  • FCS fetal calf serum
  • the pH during culturing is preferably about 6-8. Culture is usually performed at about 30 to 40 ° C for about 15 to 200 hours, and the medium is replaced, aerated, and agitated as necessary.
  • examples of a system for producing a protein in vivo include a production system using animals and a production system using plants.
  • the target DNA is introduced into these animals or plants, and proteins are produced and recovered in the animals or plants.
  • the “host” in the present invention includes these animals and plants.
  • mice When using animals, there are production systems using mammals and insects. As mammals, goats, bushes, ovines, mice, and mice can be used (Vicki Glaser, SPECTRUM Biotechnology Applications, 1993). When a mammal is used, a transgenic animal can be used.
  • the target DNA is prepared as a fusion gene with a gene encoding a protein that is specifically produced in milk, such as goat casein.
  • the DNA fragment containing the fusion gene is injected into a goat embryo, and the embryo is transplanted into a female goat.
  • the target protein can be obtained from milk produced by the transgenic goat born from the goat that has received the embryo or its progeny. Hormones may be used in transgenic goats as appropriate to increase the amount of milk containing proteins produced by transgenic goats (Ebert J. M. et al., Bio / Technology (1994) 12, 699-702).
  • silkworms can be used as insects, for example.
  • the target protein can be obtained from the body fluid of the silkworm by infecting the silkworm with a baculovirus into which DNA encoding the target protein has been inserted (Sus picture, M. et al. ., Nature (1985) 315, 592-594).
  • tobacco when using a plant, for example, tobacco can be used.
  • the DNA encoding the target protein is inserted into a plant expression vector, for example, pMON530, and this vector is inserted into Agrobacterium 'mmfemaciens. (Agrobacterium tumefaciens).
  • the bacterium is infected with tobacco, for example, Nicotiana tabacum, and the desired polypeptide can be obtained from the leaves of this tobacco (Julian K. -C. Ma et al., Eur. J. Immunol. (1994) 24, 131-138).
  • the protein of the present invention thus obtained can be isolated from inside or outside the host cell (such as a medium) and purified as a substantially pure and homogeneous protein.
  • the separation and purification of the protein may be carried out by using the separation and purification methods used in ordinary protein purification, and is not limited at all. For example, chromatography columns, filters, ultrafiltration, salting out, solvent precipitation, solvent extraction, distillation, immunoprecipitation, SDS-polyacrylamide gel electrophoresis, isoelectric focusing, dialysis, recrystallization, etc. When combined, proteins can be separated and purified.
  • chromatography examples include affinity chromatography, ion-exchange chromatography, hydrophobic chromatography, gel filtration, reversed-phase chromatography, and adsorption chromatography (Strategies for Protein Purification). and Cnaracterization: A Laboratory Course Manual. Ed Daniel R. Marshak et al., Cold Spring Harbor Laboratory Press, 1996).
  • chromatographys can be performed using liquid phase chromatography, for example, liquid phase chromatography such as HPLC and FPLC.
  • the present invention also includes highly purified proteins using these purification methods.
  • the protein can be arbitrarily modified or partially removed by reacting the protein with an appropriate protein modifying enzyme before or after purification.
  • an appropriate protein modifying enzyme for example, trypsin, chymotrypsin, lysylendopidase, protein kinase, glucosidase and the like are used.
  • the present invention also provides an antibody that binds to the protein of the present invention.
  • the form of the antibody of the present invention is not particularly limited, and includes a monoclonal antibody as well as a polyclonal antibody.
  • antiserum obtained by immunizing an immunized animal such as a heron with the protein of the present invention; This includes all classes of polyclonal and monoclonal antibodies, as well as human and recombinant humanized antibodies.
  • the protein of the present invention used as a sensitizing antigen for obtaining an antibody is not limited to the animal species from which it is derived, but is preferably a protein derived from a mammal, for example, a human, a mouse or a rat, and particularly preferably a protein derived from a human.
  • a human-derived protein can be obtained using the gene sequence or amino acid sequence disclosed herein.
  • the protein used as the sensitizing antigen may be a complete protein or a partial peptide of the protein.
  • the partial peptide of the protein include an amino group (N) terminal fragment and a carboxy (C) terminal fragment of the protein.
  • antibody refers to an antibody that reacts with the full length or fragment of a protein.
  • a gene encoding the protein of the present invention or a fragment thereof is inserted into a known expression vector system, and the vector is used to transform the host cell described in the present specification. Fragments may be obtained by a known method, and these may be used as a sensitizing antigen. Further, a cell expressing the protein, a lysate thereof, or a chemically synthesized protein of the present invention may be used as the sensitizing antigen. It is preferable that the short peptide is appropriately bound to a carrier protein such as keyhole lysine hepatocyanin, pepsin albumin, ovalbumin, etc. to form an antigen.
  • a carrier protein such as keyhole lysine hepatocyanin, pepsin albumin, ovalbumin, etc.
  • the mammal to be immunized with the sensitizing antigen is not particularly limited, but is preferably selected in consideration of compatibility with the parent cell used for cell fusion. In general, rodents are used. Eyes, egrets, and primates are used.
  • mice for example, mice, rats, hamsters and the like are used.
  • a heronoid animal for example, a heron is used.
  • monkeys are used as primates.
  • monkeys of the lower nose old world monkeys
  • cynomolgus monkeys macaques, baboons, and chimpanzees are used.
  • Immunization of an animal with a sensitizing antigen is performed according to a known method.
  • a sensitizing antigen is injected intraperitoneally or subcutaneously into a mammal.
  • the sensitizing antigen is diluted and suspended in an appropriate amount with PBS (Phosphate-Buffered Saline) or physiological saline, and then mixed with an appropriate amount of a normal adjuvant, for example, Freund's complete adjuvant, if desired. After emulsification, it is administered to mammals. Thereafter, it is preferable to administer the sensitizing antigen mixed with an appropriate amount of incomplete Freund's adjuvant several times every 4 to 21 days.
  • a suitable carrier can be used at the time of immunization with the sensitizing antigen. Immunization is performed in this manner, and an increase in the desired antibody level in the serum is confirmed by a conventional method.
  • the blood of a mammal sensitized with the antigen is taken out after confirming that the desired antibody repel in serum has been increased.
  • the serum is separated from the blood by a known method.
  • a serum containing the polyclonal antibody may be used.
  • a fraction containing the polyclonal antibody may be further isolated from this serum and used. .
  • immunoglobulin G or M can be prepared.
  • the immune cells may be removed from the mammal and subjected to cell fusion.
  • preferred immune cells used for cell fusion include splenocytes in particular.
  • the other parent cell to be fused with the immunocyte is preferably a mammalian myeloma cell, more preferably a myeloma cell that has acquired the properties for fusion cell selection by a drug.
  • Cell fusion between the immune cells and myeloma cells is basically performed according to a known method, for example, the method of Milstein et al. (Galfre, G. and Milstein, C, Methods Enzymol. (1981) 73, 3-46). It can be carried out.
  • the hybridoma obtained by cell fusion is selected by culturing it in a normal selective culture medium, for example, a HAT culture medium (a culture medium containing hypoxanthine, aminopterin and thymidine). Culturing in the HAT culture solution is continued for a period of time sufficient to kill cells other than the target hybridoma (non-fused cells), usually for several days to several weeks. Next, screening and cloning of hybridomas producing the desired antibody are performed by the usual limiting dilution method.
  • a HAT culture medium a culture medium containing hypoxanthine, aminopterin and thymidine
  • human lymphocytes for example, human lymphocytes infected with EB virus are sensitized in vitro with proteins, protein-expressing cells or lysates thereof. It is also possible to fuse a sensitized lymphocyte with a human-derived myeloma cell having permanent division ability, for example, U266, to obtain a hybridoma that produces a desired human antibody having a protein binding activity (Japanese Patent Application Laid-Open No. -17 688 publication).
  • the obtained hybridoma is transplanted into the peritoneal cavity of a mouse, ascites is collected from the mouse, and the obtained monoclonal antibody is subjected to, for example, ammonium sulfate precipitation, protein A, protein G column, DEAE ion exchange chromatography, and the present invention. It can be prepared by purifying the above protein using a coupled affinity column or the like.
  • the antibody of the present invention is used for purification and detection of the protein of the present invention, and is also a candidate for an agonist and an angoniist of the protein of the present invention. It is also conceivable to apply this antibody to antibody therapy for diseases involving the protein of the present invention.
  • a human antibody or a humanized antibody is preferable in order to reduce immunogenicity.
  • a transgenic animal having a human antibody gene repertoire is immunized with a protein serving as an antigen, protein-expressing cells or a lysate thereof to obtain antibody-producing cells.
  • a human antibody against the protein can be obtained using a hybridoma fused with myeloma cells (International Publication Nos. W092--03918, W093-2227, W094-02602, W094-25585, W096-33735 and W096). -See 34096).
  • cells in which immune cells such as sensitized lymphocytes that produce antibodies are immortalized by oncogenes may be used.
  • the thus obtained monoclonal antibody can also be obtained as a recombinant antibody produced using a genetic recombination technique (for example, Borrebaeck, AK and Larrick, JW, THERAPEUTIC MONOCLONAL ANTIBODIES, Published in the United Kingdom by MCMILLAN PUBLISHERS LTD, 1990).
  • Recombinant antibodies are produced by cloning the DNA encoding them from hybridomas or immunized cells such as sensitized lymphocytes that produce the antibody, inserting the DNA into an appropriate vector, and introducing it into a host.
  • the present invention includes this recombinant antibody.
  • the antibody of the present invention may be a modified antibody fragment thereof as long as it binds to the protein of the present invention.
  • Fab, F (ab ') 2, Fv, or a single chain Fv (scFv) in which an Fv of an H chain and an L chain are linked by an appropriate linker Huston, JS et al., Proc. Natl. Acad. Sci. USA (1988) 85, 5879-5883.
  • the antibody is treated with an enzyme, for example, papain or pepsin, to generate an antibody fragment, or a gene encoding these antibody fragments is constructed and introduced into an expression vector. Expressed in cells (for example, Co, MS et al., J.
  • modified antibody an antibody bound to various molecules such as polyethylene glycol (PEG) can be used.
  • PEG polyethylene glycol
  • the “antibody” of the present invention also includes these modified antibodies. Such a modified antibody can be obtained by subjecting the obtained antibody to chemical modification. These methods are already established in this field.
  • the antibody of the present invention can be prepared by using a chimeric antibody comprising a non-human antibody-derived variable region and a human antibody-derived constant region or a non-human antibody-derived CDR (complementarity determining region) and a human It can be obtained as a humanized antibody consisting of antibody-derived FR (framework region) and constant region.
  • the antibody obtained as described above can be purified to homogeneity.
  • the separation and purification of the antibody used in the present invention may be performed by the separation and purification methods used for ordinary proteins. For example, chromatography columns such as affinity chromatography, filtration, ultrafiltration, salting out, dialysis, SDS polyacrylamide gel electrophoresis, isoelectric focusing, etc. can be appropriately selected and combined.
  • Antibodies can be separated and purified (Antibodies: A Laboratory Manual; Ed Harlow and David Lane, Cold Spring Harbor Laboratory, 1988), but are not limited thereto.
  • the concentration of the antibody obtained above can be measured by measuring the absorbance or by using an enzyme-linked immunosorbent assay (ELISA) or the like.
  • Columns used for affinity chromatography include a protein A column and a protein G column.
  • columns using a protein A column include Hyper D, POROS, Sepharose F.F. (Pharmacia), and the like.
  • Examples of chromatography other than affinity chromatography include, for example, ion exchange chromatography, hydrophobic chromatography, gel filtration, reverse phase chromatography, and adsorption chromatography (Strategies for Protein Purification and Characterization: A Laboratory Course Manual.Ed Daniel R. Marshak et al., Cold Spring Harbor Laboratory Press, 1996). These chromatographys can be performed using liquid phase chromatography such as HPI and FPLC.
  • Methods for measuring the antigen-binding activity of the antibody of the present invention include, for example, measurement of absorbance, enzyme-linked immunosorbent assay (ELISA), EIA (enzyme-linked immunosorbent assay), Immunoassay) or a fluorescent antibody method can be used.
  • ELISA enzyme-linked immunosorbent assay
  • EIA enzyme-linked immunosorbent assay
  • Immunoassay a fluorescent antibody method
  • the protein of the present invention is added to a plate on which the antibody of the present invention is immobilized, and then a sample containing the target antibody, for example, a culture supernatant of antibody-producing cells or a purified antibody is added.
  • a secondary antibody that recognizes an enzyme for example, an antibody labeled with alkaline phosphatase, etc.
  • an enzyme substrate such as P-nitrophenylphosphoric acid and measure the absorbance.
  • Antigen binding activity can be evaluated.
  • a protein fragment for example, a fragment comprising the C-terminus thereof may be used.
  • BI Acore Pharmacia
  • the antibody of the present invention is brought into contact with the sample contained in the sample which is expected to contain the protein of the present invention, and an immune complex of the antibody and the protein is detected or measured.
  • the method for detecting or measuring a protein of the present invention can be carried out.
  • INDUSTRIAL APPLICABILITY The method for detecting or measuring a protein of the present invention can specifically detect or measure a protein, and thus is useful for various experiments and the like using proteins.
  • the present invention also provides a polynucleotide comprising at least 15 nucleotides complementary to the DNA encoding the # 10X protein (SEQ ID NOS: 1 to 7) or its complementary strand.
  • the “complementary strand” refers to one strand of a double-stranded nucleic acid consisting of A: T (U in the case of RNA) and G: C base pairs with respect to the other strand.
  • the term “complementary” is not limited to a case where the sequence is completely complementary to at least 15 contiguous nucleotide regions, but is at least 70 °, preferably at least 80%, more preferably 90%, and more preferably It is preferable that they have a homology of 95% or more on the base sequence.
  • the algorithm for determining homology may use the algorithm described in this specification.
  • nucleic acids include probes and primers used for detection and amplification of DNA encoding the protein of the present invention, probe primers for detecting the expression of the DNA, and expression of the protein of the present invention.
  • Nucleotides or nucleotide derivatives eg, antisense oligonucleotides, ribozymes, or DNA encoding them
  • Such a nucleic acid can also be used for producing a DNA chip.
  • the 3′-side region is complementary, and a restriction enzyme recognition sequence, a tag, or the like can be added to the 5′-side.
  • Antisense oligonucleotides include, for example, antisense oligonucleotides that hybridize at any position in the base sequence of SEQ ID NOs: 1 to 7.
  • This antisense oligonucleotide is preferably an antisense oligonucleotide for at least 15 or more consecutive nucleotides in the nucleotide sequence of SEQ ID NOs: 1 to 7. More preferably, it is an antisense oligonucleotide in which at least 15 or more consecutive nucleotides contain a translation initiation codon.
  • the antisense oligonucleotide derivatives and modifications thereof can be used.
  • the modified product include a modified lower alkyl phosphonate such as a methylphosphonate type or an ethylphosphonate type, a phosphorothioate modified product, a phosphoroamidate modified product, and the like.
  • Antisense oligonucleotides include not only those whose nucleotides corresponding to nucleotides constituting a predetermined region of DNA or mRNA are all complementary sequences, but also DNAs or mRNAs and oligo nucleotides shown in SEQ ID NOs: 1 to 7. As long as it can specifically hybridize to the base sequence to be used, one including one or more nucleotide mismatches is also included.
  • the antisense oligonucleotide derivative of the present invention acts on cells producing the protein of the present invention to bind to DNA or mRNA encoding the protein, thereby inhibiting the transcription or translation of the protein or producing mA. By suppressing the expression of the protein of the present invention, for example, by promoting the degradation, the effect of suppressing the action of the protein of the present invention is obtained.
  • the antisense oligonucleotide derivative of the present invention can be mixed with a suitable base material which is inactive against the derivative to prepare an external preparation such as a liniment or a poultice.
  • excipients may be added to tablets, splinters, granules, capsules, ribosome capsules, It can be a lyophilized agent such as a propellant, a liquid, a nasal drop and the like. These can be prepared according to a conventional method.
  • the antisense oligonucleotide derivative of the present invention is applied directly to the affected area of the patient, or is applied to the patient so as to be able to reach the affected area as a result of intravenous administration or the like.
  • an antisense-encapsulated material that enhances durability and membrane permeability can be used. Examples include liposomes, poly-L-lysine, lipids, cholesterol, ribofectin or derivatives thereof.
  • the dosage of the antisense oligonucleotide derivative of the present invention can be appropriately adjusted according to the condition of the patient, and a preferred amount can be used. For example, it can be administered in the range of 0.1 to 100 mg / kg, preferably 0.1 to 50 mg / kg.
  • the antisense oligonucleotide of the present invention inhibits the expression of the protein of the present invention and is therefore useful in suppressing the biological activity of the protein of the present invention. Further, the expression inhibitor containing the antisense oligonucleotide of the present invention is useful in that it can suppress the biological activity of the protein of the present invention.
  • the protein of the present invention is useful for screening for a compound that binds to the protein. That is, the protein of the present invention is brought into contact with a test sample expected to contain a compound binding to the protein, and a compound having an activity of binding to the protein of the present invention is selected. And a method of screening for a compound that binds to the protein of the present invention.
  • the protein of the present invention used for the screening may be a recombinant protein or a protein of natural origin. It may be a partial peptide. It may also be a form expressed on the cell surface or a form as a membrane fraction.
  • the test sample is not particularly limited and includes, for example, a cell extract, a cell culture supernatant, a fermentation microorganism product, a marine organism extract, a plant extract, a purified or crude protein, a peptide, a non-peptidic compound, and a synthesis. Low molecular weight compounds and natural compounds.
  • the protein of the present invention which is brought into contact with a test sample, may be, for example, a purified protein, a soluble protein, a form bound to a carrier, a fusion protein with another protein, or a form expressed on a cell membrane.
  • the sample can be brought into contact with a test sample as a membrane fraction.
  • a method for screening a protein that binds to the protein using the protein of the present invention for example, many methods known to those skilled in the art can be used. Such screening can be performed, for example, by immunoprecipitation. Specifically, it can be performed as follows.
  • the gene encoding the protein of the present invention is introduced into a vector for expression of a foreign gene such as pSV2neo, pcDNA I, or pCD8, so that the gene is expressed in animal cells or the like.
  • Promoters used for expression include SV40 early promoter (Rigoy In Williamson ea.), Genetic Engineering, Vol. 3. Academic Press, London, p. 83-141 (1982)), EF-1 promoter (Kim et al.
  • CAG promoter Niwa et al. Gene 108, p.193-200 (1991)
  • SR a promoter Takebe et al. Mol. Cell. Biol. 8, p. 66 (1988)
  • SV40 late promoter Gheysen and Fiers J. Mol. A ppl. Genet. II, p. 385-394 (1982)
  • Adenovirus late promoter Kaufman et a 1. Mol. Cell. Biol. 9, p. 946 (1989)
  • HSV TK promoter any other commonly used promoters can be used.
  • the protein of the present invention is expressed as a fusion protein having a monoclonal antibody recognition site by introducing a recognition site (epitope) of the monoclonal antibody of which specificity is known into the N-terminal or C-terminal of the protein of the present invention. be able to .
  • a commercially available epitope-antibody system can be used (Experimental Medicine 1 ⁇ , 85-90 (1995)).
  • Vectors that can express a fusion protein with ⁇ -galactosidase, maltose binding protein, glutathione S-transferase, green fluorescent protein (GFP), and the like via a multicloning site are commercially available.
  • an immune complex is formed by adding these antibodies to a cell lysate prepared using an appropriate surfactant.
  • This immune complex comprises the protein of the present invention, a protein capable of binding thereto, and an antibody.
  • immunoprecipitation can also be performed using antibodies against the protein of the present invention.
  • Antibodies against the protein of the present invention include, for example, a gene encoding the protein of the present invention introduced into an appropriate E. coli expression vector, expressed in E. coli, and the expressed protein is purified. It can be prepared by immunizing goats and chickens. Alternatively, it can be prepared by immunizing the above animal with the synthesized partial peptide of the protein of the present invention.
  • the immune complex can be precipitated using Protein A Sepharose or Protein G Sepharose.
  • an epitope such as GST
  • a substance specifically binding to these epitopes such as glutathione-Sepharose 4B is used to prepare the protein of the present invention.
  • An immune complex can be formed in the same manner as in the case where an antibody of the above protein is used.
  • SDS-PAGE is generally used for analysis of immunoprecipitated proteins.
  • the bound proteins can be analyzed by the molecular weight of the protein.
  • the protein of interest can be purified directly from the SDS-polyacrylamide gel and its sequence determined.
  • Examples of the method for isolating a protein that binds to the protein using the protein of the present invention include, for example, the West Western blotting method (Skolnik, EY et al., Cell (1991) 65, 83-90). ). That is, cells, tissues, and organs that are expected to express the protein that binds to the protein of the present invention (for example, fat cells and tissues in which expression has been observed by Northern blotting in the Examples) are phage-derived.
  • a cDNA library using a vector human gtll, ZAP, etc.
  • the protein expressed on a filter was fixed, purified and labeled with the protein of the present invention.
  • the plaque expressing the protein bound to the protein of the present invention may be detected by labeling by reacting the above-mentioned filter with the protein.
  • the method for labeling the protein of the present invention include a method utilizing the binding property of biotin and avidin, a method of specifically binding to the protein of the present invention or a peptide or polypeptide fused to the protein of the present invention (for example, GST).
  • the method include a method using an antibody, a method using a radioisotope, and a method using fluorescence.
  • a 2-hybrid system using cells Yields, S., and Sternglanz, R., Trends. Genet. (1994) 10, 286-292, Dalton S, and Treisman R (1992) Characterization of SAP-1, a protein recruited by serum response factor to the c-fos serum response element.Cell 68, 597-612, ⁇ MATCHMARKER Two-Hybrid Systemj, '' Mamm alian MATCHMAKER Two-Hybrid Assay Kit ”,“ MATCHMAKEE One-Hybrid System ”(all manufactured by Clontech), and“ HybriZAP Two-Hybrid Vector Systemj ”(Strata Gene) are used.
  • the protein of the present invention or a partial peptide thereof is fused with an SRF DNA binding region or GAL4 DNA binding region and expressed in yeast cells, and the protein of the present invention and From the cells that are expected to express the protein to be bound, a cDNA library was prepared that could be expressed in a form fused with the VP16 or GAL4 transcription activation region, and introduced into the yeast cells described above for detection.
  • the library-derived cDNA is isolated from the obtained positive clones (when the protein that binds to the protein of the present invention is expressed in yeast cells, the binding of the two activates the reporter gene, and a positive clone can be confirmed).
  • the protein encoded by the cDNA can be obtained by introducing the isolated cDNA into Escherichia coli for expression. As a result, it is possible to prepare a protein that binds to the protein of the present invention or a gene thereof.
  • the repo overnight gene used in the 2-hybrid system includes, for example, the HIS3 gene, Ade2 gene, LacZ gene, CAT gene, luciferase gene, PAI-1 (Plasminogen activator inhibitor typel) gene, etc. But not limited to these. Screening by the two-hybrid method can be performed using mammalian cells in addition to yeast cells.
  • Screening for a compound that binds to the protein of the present invention can also be performed by affinity mouth chromatography.
  • the protein of the present invention is immobilized on an affinity carrier, and a test sample which is expected to express a protein that binds to the protein of the present invention is applied thereto.
  • the test sample in this case includes, for example, a cell extract, a cell lysate, and the like. After applying the test sample, the column is washed to prepare a protein bound to the protein of the present invention.
  • the obtained protein is analyzed for its amino acid sequence, an oligo DNA is synthesized based on the amino acid sequence, and a DNA encoding the protein can be obtained by screening a cDNA library using the DNA as a probe. .
  • a biosensor utilizing the surface plasmon resonance phenomenon can be used as a means for detecting or measuring the bound compound.
  • a biosensor utilizing the surface plasmon resonance phenomenon can be used to detect the interaction between the protein of the present invention and a test compound using a trace amount of protein and without labeling the surface plasmon resonance signal. It can be observed in real time as a null (for example, BIAcore, manufactured by Pharmacia). Therefore, it is possible to evaluate the binding between the protein of the present invention and a test compound by using a biosensor such as BIAcore.
  • Methods for isolating not only proteins but also compounds that bind to the protein of the present invention include, for example, immobilized protein of the present invention, synthetic compounds, and natural compounds.
  • a method for screening molecules that bind to the protein of the present invention using a product bank or a random phage peptide display library, and a screening method using high throughput by combinatorial chemistry technology (Wrighton NC; Farrell FX; Chang R Kashyap AK; Barbone FP; Mulcahy LS Johnson DL; Barrett RW; Jolliffe LK; Dower WJ.
  • the compound that can be isolated by the screening of the present invention is a candidate for the agent for regulating the activity of the protein of the present invention, and is a disease caused by abnormal expression or dysfunction of the protein of the present invention or the activity of the protein of the present invention. It can be applied to the treatment of diseases that can be treated by controlling the disease. Substances that can be converted by addition, deletion, and / or substitution of a part of the structure of a compound that can be isolated using the screening method of the present invention are also included in the compounds that bind to the protein of the present invention.
  • the protein of the present invention or a compound that can be isolated by the screening of the present invention, is used in a human mammal, such as a mouse, a rat, a guinea pig, a heron, a chicken, a cat, a dog, a sheep, a bush, a monkey, a monkey, a baboon,
  • a human mammal such as a mouse, a rat, a guinea pig, a heron, a chicken, a cat, a dog, a sheep, a bush, a monkey, a monkey, a baboon
  • the protein or isolated compound itself can be administered directly to a patient, or can be formulated and administered by a known pharmaceutical method.
  • pharmacologically acceptable carriers or vehicles specifically, sterile water or saline, vegetable oils, emulsifiers, suspending agents, surfactants, stabilizers, flavoring agents, excipients, vehicles, It is possible to formulate the drug product by combining it with a preservative, binder and the like as appropriate and mixing it in the unit dosage form required for accepted pharmaceutical practice.
  • the effective components of these preparations are intended to provide a suitable volume in the specified range.
  • Additives that can be incorporated into tablets and capsules include, for example, binders such as gelatin, corn starch, tragacanth gum, acacia, excipients such as crystalline cellulose, corn starch, gelatin, and alginic acid. Suitable leavening agents, lubricants such as magnesium stearate, sweeteners such as sucrose, lactose or saccharin, and flavoring agents such as peppermint, cocoa oil or cherry are used.
  • the unit dosage form is a capsule, the above-mentioned materials may further contain a liquid carrier such as an oil or fat.
  • a sterile composition for injection can be formulated using a potable vehicle such as distilled water for injection according to normal pharmaceutical practice.
  • Aqueous injection solutions include, for example, physiological saline, isotonic solutions containing pudose and other adjuvants, such as D-sorbitol, D-mannose, D-mannitol, and sodium chloride. It may be used in combination with an agent, for example, an alcohol, specifically ethanol, a polyalcohol, for example, propylene glycol, polyethylene glycol, a nonionic surfactant, for example, polysorbate 80 (TM) or HC0-50.
  • an agent for example, an alcohol, specifically ethanol, a polyalcohol, for example, propylene glycol, polyethylene glycol, a nonionic surfactant, for example, polysorbate 80 (TM) or HC0-50.
  • the oily liquid includes sesame oil and soybean oil, and may be used in combination with benzyl benzoate or benzyl alcohol as a solubilizer.
  • buffers such as phosphate buffers, sodium acetate buffers, soothing agents such as proforce hydrochloride, stabilizers, such as It may be blended with benzyl alcohol, phenol and antioxidants.
  • the prepared injection solution is usually filled into an appropriate ampoule.
  • Administration to patients can be performed, for example, by intraarterial injection, intravenous injection, subcutaneous injection, etc., or intranasally, transbronchially, intramuscularly, transdermally, or orally by methods known to those skilled in the art. It can do better.
  • the dose varies depending on the weight and age of the patient, the administration method, and the like, but those skilled in the art can appropriately select an appropriate dose.
  • the compound can be encoded by DNA
  • the DNA may be incorporated into a gene therapy vector to perform gene therapy.
  • the dose and administration method vary depending on the patient's body weight, age, symptoms, etc., but those skilled in the art can appropriately select the dose of the protein of the present invention.
  • target organ, symptoms and method of administration for example, in the form of an injection, it is generally considered to be about 100 to 20 mg per day for an adult (with a body weight of 60 kg).
  • the dose of the compound that binds to the protein of the present invention or the compound that modulates the activity of the protein of the present invention varies depending on the symptoms. However, in the case of oral administration, in general, in adults (with a body weight of 60 kg), 1 It is believed to be about 0.1 to 100 mg per day, preferably about 1.0 to 50 mg, more preferably about 1.0 to 20 mg.
  • FIG. 1 is an electrophoretic photograph showing the result of detecting the expression of the gene of the present invention in 3T3-LI cells by Northern blot analysis. Lane 1 shows proliferating 3T3-L1 cells, lane 2 shows confluent 3T3-L1 cells, and lane 3 shows 3T3-L1 cells induced to differentiate into adipocytes.
  • FIG. 2 is an electrophoretic photograph showing the results of detecting the expression of the gene of the present invention in various tissues by Northern blot analysis.
  • the lanes are as follows: 1: heart, 2: brain, 3: spleen, 4: lung, 5: liver, 6: skeletal muscle, :: kidney, 8: testis.
  • Figure 3 shows the expression of # 103 to # 106 and GAPDH in 3T3-L1 in log phase, 3T3-Ll in confluent state, and 3T3-L1 1 to 9 days after induction of differentiation into fat cells.
  • It is an electrophoresis photograph which shows the result detected by Northern blot analysis.
  • FIG. 4 is an electrophoretic photograph showing the results of Northern blot analysis of the expression of # 105 and GAPDH in adipocytes to which OnM (control: MOCK), 25 nM, and 250 nM insulin was added.
  • FIG. 5 is an electrophoretic photograph showing the results of detecting the expression of # 104 in various cancer cell lines, adipocytes and 3T3-L1 by Northern blot analysis.
  • a cDNA library was constructed and expressed using the retroviral vector pMX-SST (Kojima. And Kitamura 5 T. (1999) Nature Biotechnol. 17, 487-490).
  • Differentiated ST3-L1 cells were extracted using Fast Track 2.0 mRNA isolation kit (Invitrogen, Lisbad, Calif.) According to the manufacturer's protocol.
  • Complementary DNA (cDNA) was synthesized from poly (A) + RNA using random hexamers using the Superscript 'Thiois' system (Gibco-BRL, Rockville, Maryland, USA).
  • the BstXI adapter was used to insert into the BstXI site of the pMX-SST vector (Invitrogen, Lisbad, CA).
  • the ligated DNA was amplified into DH10B cells (Electromax, Gibco-BRL) and the Qiagen Plasmid kit (Qiagen-Inc, Valencia, CA) was used. To prepare library DNA.
  • a cell line containing a high-potency retrovirus presenting the SST-REX library was packaged (Pear, W. S. et al. (1993) Proc. Natl. Acad. Sci. U. S.A. 90, 839
  • genomic DNA was extracted from factor-independent Ba / F3 clones, subjected to genomic PCR, and the integrated cDNA was recovered using vector primers (GGGG GTGGACCATCCTCTA / SEQ ID NO: 12, and CGCGCAGCTGTAAACGGTAG / sequence). Number: 1 3).
  • GeneAmp PCR System 2400 Perkin Elmer, Norwalk, Conn.
  • LA Taq Polymerase Yukara, Kyoto, Japan
  • the obtained PCR fragments were sequenced using the Taq Dye Mineral One-Day Cycle Sequencing Kit (Applied Biosystems, Inc., Foss Yuichi City, Calif-Ornier) to obtain an automated sequencer (310 Analyzed by Gene Analyzer, Applied'Biosystems, Inc.). We screened 9 ⁇ 10 5 clones. Cells growing in a total of 71 cells out of 1152 cells generated a single PCR band, which was subjected to further analysis. The differentiated 3T3-LI cells used in the experiment were prepared as follows.
  • the 3T 3-L1 cells were cultured in 10% FCS, 50 units / ml penicillin, and 50 ⁇ G / ml Sutorebutoma containing leucine DMEM (DMEM-FCS) 5% C0 2 at 37 ° C in. Cells were grown to confluence and maintained for 2 days. To induce differentiation, the medium was changed to DMEM-FCS containing 0.5 IDM trimethyl-3-isobutylxanthine, 0.25 / M dexamethasone, and 10 ⁇ g / ml insulin. Two days later, the medium was changed to DMEM-; FCS to differentiate the cells.
  • Ba / F3, a mouse IL-3 dependent pre-B cell line was cultured in RPMI 1640 medium containing 10% FCS and 2 ng / ml mouse IL-3 (MD Systems).
  • 63 out of 71 integrants contained the 5 'sequence of the cDNA for 28 known proteins, all of which contained the signal sequence (Table 1).
  • FKBP23 FK506-binding protein
  • Amyloid precursor-like protein 2 (APLP2) 4
  • RNA 1 ⁇ g of poly (A) + RNA is electrophoresed on a 1.0% agarose gel containing 2% formaldehyde, and then transferred to Hybond-N-nylon membrane (Amersham Pharmacia-Biotech). did.
  • Membrane is incubated at 42 ° C in hybridization buffer (50% formamide, 10X Denhardt's reagent, 5x SSC, 0.1% SDS, 200 ⁇ g / ml denatured salmon sperm DNA) at 42 ° C. the 3 2 P- labeled DNA fragments derived from examined as a probe.
  • hybridization buffer 50% formamide, 10X Denhardt's reagent, 5x SSC, 0.1% SDS, 200 ⁇ g / ml denatured salmon sperm DNA
  • One of the membranes was used as a loading control to evaluate the amount of RNA in each lane.
  • the cDNA for hydrogenase (GAPDH) was probed. After hybridization, the filter was washed in
  • Clone # 105 showed two bands.
  • low molecular mass mA was significantly expressed.
  • clone # 103 the only increase in mRNA was sufficient for 3T3-L1 cells to grow to confluence.
  • FIG. 2 The expression profile of the novel adipocyte-derived gene in multiple mouse tissues was examined by Northern plot analysis (FIG. 2).
  • Mouse multi-tissue Northern plots were purchased from Clontech (Palo Alto, CA).
  • Example 5 Daily change of mRNA Northern blot analysis was performed by the usual method (samples in each lane were 3T3-Ll in log phase, 3T3-Ll in confluent state, and 1 to 9 days after induction of adipocyte differentiation) This is 3T3-L1 of the eye, and # 103 to # 106, GAPDH was used as the probe.
  • Northern plot analysis was performed by the usual method.
  • the samples in each lane are control adipocytes, adipocytes supplemented with 25 nM and 250 nM insulin, respectively.
  • # 105, GAPDH was used as a probe.
  • cancer cell lines showed low molecular weight bands and some did not.
  • a gene encoding a novel protein derived from an adipocyte and having a signal sequence is provided. These genes may be important molecules involved in adipocyte differentiation due to their expression characteristics.
  • the protein of the present invention is a target for drug development
  • the compound that regulates the function of the protein of the present invention is expected to be applied as a pharmaceutical.

Landscapes

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

Abstract

By screening molecules having signal peptides with the use of an originally developed efficient signal sequence trapping method, 8 novel genes likely encoding membranes or soluble proteins are successfully identified. These genes show expression in association with the differentiation of adipocytes.

Description

マウス脂肪細胞由来遺伝子 技術分野  Mouse adipocyte-derived gene
本発明は、 脂肪細胞に由来し、 シグナル配列を有する新規な蛋白質およびその 遺伝子、 並びにそれらの製造および用途に関する。 背景技術  The present invention relates to a novel protein derived from an adipocyte and having a signal sequence, a gene thereof, and their production and use. Background art
脂肪組織は、 その変異がマウスにおける遺伝性の肥満を引き起こす (Zhang,Y .et al.(1994) Nature 372, 425-432) レブチンと呼ばれるサイ トカインを産生 することが知られるまで、 単なる脂肪の貯蔵庫に過ぎないと考えられてきた。 現 在では、 脂肪組織は最大の分泌臓器であると認識されている。  Adipose tissue is simply fat-free until it is known that the mutation causes hereditary obesity in mice (Zhang, Y. et al. (1994) Nature 372, 425-432). It was thought to be just a storage. At present, adipose tissue is recognized as the largest secretory organ.
脂肪細胞から産生されるその他の分泌型蛋白質としては、 TNF-ひやプラスミ ノーゲン活性化因子阻害剤 (PAI-1) が挙げられる。 TNF-ひは、 脂質やダルコ一 スの代謝に影響を及ぼし、 遺伝的に肥満の齧歯類の脂肪組織において過剰発現し ており (Hotamisligil,G.S. et al.(1993) Science 259, 87-91) 、 インスリン 抵抗性につながる (Hotamisligil3G.S. et al.(1995) J.Clin. Invest. 95, 240 9-2415) o PAI- 1は、 内蔵の脂肪細胞において高度に発現し、 内臓肥満における 血管疾患の発症に関係している (ShimomuraJ. et al.(1996) Nature medicine 2, 800-803) 。 Other secreted proteins produced by adipocytes include TNF-chicken plasminogen activator inhibitor (PAI-1). TNF-H affects the metabolism of lipids and darcos and is overexpressed in adipose tissue of genetically obese rodents (Hotamisligil, GS et al. (1993) Science 259, 87-91 ), Leads to insulin resistance (Hotamisligil 3 GS et al. (1995) J. Clin. Invest. 95, 240 9-2415) o PAI-1 is highly expressed in visceral adipocytes and It has been implicated in the onset of the disease (Shimomura J. et al. (1996) Nature medicine 2, 800-803).
現在まで、 脂肪細胞から同定されているサイ トカインはごくわずかであるが、 脂肪組織においては、 これら以外にも、 脂質やグルコースの代謝を調節する可溶 性因子が分泌されていると考えられる。 発明の開示 本発明は、 脂肪細胞に由来し、 シグナル配列を有する新規な蛋白質、 それらの 遺伝子、 およびそれらと機能的に同等な分子、 並びにそれらの製造および用途を 提供する。 To date, very few cytokines have been identified in fat cells, but it is thought that adipose tissue secretes other soluble factors that regulate lipid and glucose metabolism. Disclosure of the invention The present invention provides novel proteins derived from adipocytes and having a signal sequence, their genes, molecules functionally equivalent thereto, and their production and use.
本発明者等は、 上記課題を解決するために、 脂肪細胞分化に関してよく特徴付 けされたモデルである 3T3-L1細胞株 (Meuth5M. and Green,H. ( 1974) Cell 3, 367-374; Green5H. and Kehinde30. ( 1975 ) Cell 5, 19-27) から cDNAライブラ リを調製し、 レトロウイルス媒介発現クローニングシステム (Kojima,T. and K itamura3 T. ( 1999) Nature Biotechnol .17, 487-490) を用いて独自に閧発した 効率的なシグナル配列トラヅプ法を用い、 シグナルペプチド (von Heijne、 ( 19 85 ) J. Mol . Biol .184, 99-105) を有する分子のスクリーニングを行なった。 SST- REX法では恒常的活性型サイ トカインレセプ夕ー MPLとの融合蛋白質を発現する ライブラリーをスクリーニングすることによって、 MPLを細胞表面に発現させる ことができる蛋白質をコードする cDNAを探索する。 この方法では、 MPLの細胞 表面への発現による、 IL- 3依存性の細胞株への自律増殖能の賦与を指標とする ため、 簡便に目的のクローンを選別できる。 In order to solve the above-mentioned problems, the present inventors have developed a 3T3-L1 cell line (Meuth 5 M. and Green, H. (1974) Cell 3, 367-A) which is a well-characterized model for adipocyte differentiation. 374; Green 5 H. and Kehinde 3 0. (1975) Cell 5, a cDNA library prepared from 19-27), retrovirus-mediated expression cloning system (. Kojima, T and K itamura 3 T. (1999) Nature Biotechnol.17, 487-490), and has a signal peptide (von Heijne, (1985) J. Mol. Biol. 184, 99-105) using an efficient signal sequence trapping method. Molecular screening was performed. In the SST-REX method, a cDNA encoding a protein capable of expressing MPL on the cell surface is searched for by screening a library that expresses a fusion protein with the constitutively active cytokine receptor MPL. In this method, the target clone can be easily selected because the expression of the autonomic proliferation ability to an IL-3-dependent cell line by expression of MPL on the cell surface is used as an index.
本発明者等は、 9 X 105個のクローンをスクリーニングした結果、 膜または可 溶型蛋白質をコードする可能性がある新規遺伝子を 8個を同定することに成功 した。 そのうち、 7個をそれそれ 「#101」 から 「#107」 と命名した。 As a result of screening 9 × 10 5 clones, the present inventors succeeded in identifying eight novel genes that may encode a membrane or a soluble protein. Seven of them were named "# 101" to "# 107", respectively.
これら遺伝子は、 脂肪細胞の分化に関連した発現を示した。 また、 発現する組 織は異なるものの、 組織特異的な発現を示した。 従って、 これら遺伝子は、 機能 部位は異なるが、 それそれが生体内において脂肪細胞の分化に関連した役割を担 う分子であると考えられる。  These genes showed expression associated with adipocyte differentiation. In addition, although the expressed tissues were different, they showed tissue-specific expression. Therefore, although these genes have different functional sites, they are thought to be molecules that play a role related to adipocyte differentiation in vivo.
本発明は、 脂肪細胞に由来し、 シグナル配列を有する新規な蛋白質、 その遺伝 子、 およびそれらと機能的に同等な分子、 並びにそれらの製造および用途に関し 、 より具体的には、  The present invention relates to a novel protein derived from an adipocyte and having a signal sequence, a gene thereof, a molecule functionally equivalent thereto, and production and use thereof, and more specifically,
( 1 ) 下記 (a ) から (f ) のいずれかに記載の DNA、 (a) 配列番号: 8〜1 1のいずれかに記載のアミノ酸配列からなる蛋白質をコ ードする DNA。 (1) a DNA according to any of the following (a) to (f), (a) a DNA encoding a protein consisting of the amino acid sequence of any one of SEQ ID NOs: 8 to 11;
(b) 配列番号: 1〜7のいずれかに記載の塩基配列のコード領域を含む DNA。  (b) DNA comprising the coding region of the nucleotide sequence of any one of SEQ ID NOs: 1 to 7.
(c) 配列番号: 8〜1 1のいずれかに記載のアミノ酸配列において 1若しくは 複数のアミノ酸が置換、 欠失、 挿入、 および/または付加したアミノ酸配列を有 し、 配列番号: 8〜1 1のいずれかに記載のアミノ酸配列からなる蛋白質と機能 的に同等な蛋白質をコードする DNA。  (c) one or more amino acids in the amino acid sequence of any of SEQ ID NOs: 8 to 11 having substitution, deletion, insertion, and / or addition of an amino acid sequence, and SEQ ID NOs: 8 to 11 A DNA encoding a protein functionally equivalent to the protein comprising the amino acid sequence of any one of the above.
(d) 配列番号: 1、 2、 7のいずれかに記載の塩基配列からなる MAに対応 する全長 DNAによりコードされる蛋白質のアミノ酸配列において 1若しくは複 数のアミノ酸が置換、 欠失、 挿入、 および/または付加したアミノ酸配列を有し 、 該全長 DNAによりコードされる蛋白質と機能的に同等な蛋白質をコードする D  (d) one or more amino acids in the amino acid sequence of the protein encoded by the full-length DNA corresponding to MA consisting of the nucleotide sequence of any of SEQ ID NOs: 1, 2, and 7; substitution, deletion, insertion, And / or has an added amino acid sequence and encodes a protein functionally equivalent to the protein encoded by the full-length DNA.
( e ) 配列番号: 3〜 6のいずれかに記載の塩基配列からなる DNAとストリン ジェントな条件下でハイブリダイズし、 配列番号: 8〜 1 1に記載のアミノ酸配 列からなる蛋白質と機能的に同等な蛋白質をコードする DNA。 (e) hybridizes with a DNA comprising the nucleotide sequence of any one of SEQ ID NOs: 3 to 6 under stringent conditions, and functions as a protein comprising the amino acid sequence of SEQ ID NOs: 8 to 11; DNA that encodes a protein equivalent to
(f) 配列番号: 1、 2、 7のいずれかに記載の塩基配列からなる DNAとスト リンジヱントな条件下でハイブリダイズし、 これら MAに対応する全長 MAに よりコ一ドされる蛋白質と機能的に同等な蛋白質をコ一ドする DNA。  (f) a protein that hybridizes under stringent conditions with DNA consisting of the nucleotide sequence of any of SEQ ID NOs: 1, 2, and 7, and functions as a protein encoded by the full-length MA corresponding to these MAs DNA that encodes proteins that are equivalent to each other.
(2) 配列番号: 8〜1 1のいずれかに記載のアミノ酸配列からなる蛋白質 の部分べプチドをコードする DNA、  (2) a DNA encoding a partial peptide of a protein consisting of the amino acid sequence of any of SEQ ID NOs: 8 to 11,
(3) ( 1) または (2) に記載の DNAによりコードされる蛋白質または ぺプチド、  (3) a protein or peptide encoded by the DNA of (1) or (2),
(4) (1) または (2) に記載の DNAが挿入されたベクター、  (4) a vector into which the DNA of (1) or (2) has been inserted,
(5) (1) または (2) に記載の DNAまたは (4) に記載のベクターを 保持する宿主細胞、 (6) (5) に記載の宿主細胞を培養し、 該宿主細胞またはその培養上清か ら発現させた蛋白質を回収する工程を含む、 (3) に記載の蛋白質またはべプチ ドの製造方法、 (5) a host cell carrying the DNA of (1) or (2) or the vector of (4), (6) The method for producing the protein or peptide according to (3), comprising culturing the host cell according to (5) and recovering the expressed protein from the host cell or a culture supernatant thereof. ,
(7) (3) に記載の蛋白質に結合する抗体、  (7) an antibody that binds to the protein according to (3),
( 8 ) 配列番号: 1から 7のいずれかに記載の塩基配列からなる DNAまた はその相補鎖に相補的な少なくとも 15ヌクレオチドを含むポリヌクレオチド、 および  (8) a polynucleotide comprising at least 15 nucleotides complementary to a DNA consisting of the nucleotide sequence of any one of SEQ ID NOs: 1 to 7 or a complementary strand thereof, and
(9) (3) に記載の蛋白質に結合する化合物のスクリーニング方法であつ て、  (9) A method for screening a compound that binds to the protein according to (3),
(a) 該蛋白質またはその部分べプチドに被検試料を接触させる工程、  (a) contacting a test sample with the protein or a partial peptide thereof,
(b) 該蛋白質またはその部分べプチドと被検試料との結合活性を検出する工程  (b) a step of detecting a binding activity between the protein or a partial peptide thereof and a test sample
(c) 該蛋白質またはその部分べプチドに結合する活性を有する化合物を選択す る工程、 を含む方法、 を提供するものである。 and (c) selecting a compound having an activity of binding to the protein or a partial peptide thereof.
本発明は、 脂肪細胞に由来し、 シグナル配列を有する新規な蛋白質をコードす る遺伝子を提供する。 本発明者等は、 分泌型および膜蛋白質をコードする脂肪細 胞由来 cDNAを、 最近確立された新規シグナル配列トラップ法、 SST- REX法によ つて検索し、 既知の cDNA 63個および新規 cDNA 8個を得た。 本発明者等により SST-REX法を利用して単離された新規 cDNAのうち、 7つの cDNA (これらクロ一 ンをそれそれ 「#101」 〜 「#107」 と命名した。 また、 これらクローンをまとめ て 「#10X」 と称する。 ) の塩基配列をそれそれ配列番号: 1〜7に示した。 ま た、 #103から #106 cDNAによりコードされる蛋白質のアミノ酸配列を配列番号 : 8-1 1に示した。  The present invention provides a gene encoding a novel protein derived from an adipocyte and having a signal sequence. The present inventors searched for fat cell-derived cDNAs encoding secretory and membrane proteins by the recently established novel signal sequence trapping method, SST-REX method, and found 63 known cDNAs and 8 novel cDNAs. Got a piece. Among the novel cDNAs isolated by the present inventors using the SST-REX method, seven cDNAs (these clones were named “# 101” to “# 107”, respectively. Are collectively referred to as "# 10X.") The nucleotide sequences of The amino acid sequence of the protein encoded by the # 103 to # 106 cDNA is shown in SEQ ID NOs: 8-11.
SST-REX法により単離された 63個の cDNA全てが既知の蛋白質 28個の cMAの 5'配列を含むことが判明し、 それらの全てが膜または分泌型蛋白質であった。 クローン 63個 (79%) 中、 50個は分泌型蛋白質であった。 これは脂肪細胞が分 泌臓器を構築する細胞としての集団を有することを示している。 特にクローン 6 3個中 28個 (44%) は、 コラーゲンのような細胞外マトリクスまたは関連蛋白 質であった。 繊維芽細胞の細胞外マトリックス蛋白質分泌能は、 脂肪細胞への分 化の際に増強されることが示された。 コラーゲンひ- 1 ( IV) およびひ- 2 ( IV) の合成および分泌は、 繊維芽細胞では無視できる程度であるが、 脂肪細胞では顕 著に増加している。 ェン夕クチンの分泌増強もまた認められた (Aratani,Y. an d Kitagawa3 Y. ( 1988) J. Biol . Chem. 263, 16163-16169) 。 本発明者等により示 された結果は、 これらの結果と一致しており、 細胞外マトリックス蛋白質の合成 が脂肪組織の形態形成にとつて重要であることを示唆している。 All 63 cDNAs isolated by the SST-REX method were found to contain 28 cMA 5 'sequences of 28 known proteins, all of which were membrane or secreted proteins. Of 63 clones (79%), 50 were secreted proteins. This is a fat cell It shows that it has a population as cells that make up a urinary organ. In particular, 28 out of 63 clones (44%) were extracellular matrices such as collagen or related proteins. It has been shown that the ability of fibroblasts to secrete extracellular matrix proteins is enhanced upon differentiation into adipocytes. Collagen sph-1 (IV) and sph-2 (IV) synthesis and secretion are negligible in fibroblasts, but markedly increased in adipocytes. Enhancement of endocin secretion was also observed (Aratani, Y. and Kitagawa 3 Y. (1988) J. Biol. Chem. 263, 16163-16169). The results shown by the present inventors are consistent with these results, and suggest that the synthesis of extracellular matrix proteins is important for adipose tissue morphogenesis.
また、 10個のクローンは、 コラーゲン ' リピート (Scherer,P. E. et al . ( 199 5 ) J. Biol . Chem. 270, 26746-26749) を有する 30kDa (Acrp30) の脂肪細胞補 体関連蛋白質であった。 Acrp30 mRNAは脂肪細胞分化の際に 100倍以上誘導され る。 従って、 分泌型蛋白質 50個中 10個 (20% ) が Acrp30であった。  Also, 10 clones were 30 kDa (Acrp30) adipocyte complement-related proteins with collagen 'repeats (Scherer, PE et al. (1995) J. Biol. Chem. 270, 26746-26749). . Acrp30 mRNA is induced more than 100-fold during adipocyte differentiation. Therefore, 10 out of 50 (20%) secreted proteins were Acrp30.
また、 リポ蛋白質リパーゼ (LPL) クローンが 2個存在した。 LPLは、 脂肪の 蓄積に関係する脂肪細胞特異的遺伝子であり、 その発現は脂肪細胞では PPARァ によって増強される。 SST-REX法による脂肪細胞 cDNAライブラリのスクリー二 ングによって LPL遺伝子が得られたことは妥当である。 一方、 シンデカン- 1お よびライソゾ一ム膜糖蛋白質タイプ Aのような蛋白質は、 ハウスキーピング遺 伝子である可能性がある。  There were also two lipoprotein lipase (LPL) clones. LPL is an adipocyte-specific gene involved in fat accumulation, and its expression is enhanced in adipocytes by PPARa. It is appropriate that the LPL gene was obtained by screening the adipocyte cDNA library by the SST-REX method. On the other hand, proteins such as syndecan-1 and lysosomal membrane glycoprotein type A may be housekeeping genes.
ノザンプロット分析によって、 脂肪細胞 cDNAライブラリから得られたほとん どの SSTクローンは、 脂肪細胞特異的発現パターンを示した。 前脂肪細胞から 脂肪細胞への分化は、 遺伝子発現パターンを劇的に変化させるかもしれない。 ク ローン #103は、 3T3-L1前脂肪細胞ではほとんど発現されなかったが、 コンフル ェント細胞および脂肪細胞では認められた。 一つの可能性は、 それがリポ蛋白質 リパーゼ、 コラーゲンおよび FAAR (脂肪酸活性化受容体) のような脂肪細胞分 化の初期マーカ一であることかも知れない (MacDougald, 0.A. and Lane,M. D. ( l 995 ) Annu. Rev. Biochem. 64, 345-375) 。 初期マーカ一遺伝子の発現はその後 の前脂肪細胞分化にとって必須であるため、 本質的で重要である。 特にクローン #106 mRNAのレベルは分化の際に増加した。 従って、 これは脂肪細胞分化に関連 する可能性がある。 クローン #105の発現は興味深い。 2つの転写物がノザンブ ロット分析によって検出された。 2つの転写物は異なる遺伝子に由来する可能性 もある。 By Northern blot analysis, most SST clones obtained from adipocyte cDNA libraries showed an adipocyte-specific expression pattern. Differentiation of preadipocytes into adipocytes may dramatically alter gene expression patterns. Clone # 103 was hardly expressed in 3T3-L1 preadipocytes, but was found in confluent and adipocytes. One possibility may be that it is one of the early markers of adipocyte differentiation such as lipoprotein lipase, collagen and FAAR (fatty acid activated receptor) (MacDougald, 0.A. and Lane, MD (l 995) Annu. Rev. Biochem. 64, 345-375). Expression of the early marker gene is essential and important for subsequent preadipocyte differentiation. In particular, the level of clone # 106 mRNA increased during differentiation. Thus, this may be related to adipocyte differentiation. The expression of clone # 105 is interesting. Two transcripts were detected by Northern blot analysis. The two transcripts can be from different genes.
さらに、 ノザンプロット分析により、 #103では分化誘導後初期の段階で一時 的にほとんど発現が消失した。 3T3- L1を脂肪細胞に分化誘導すると、 まず一部 の細胞が分裂 ·増殖し、 それが脂肪細胞になるという説がある。 また活発に増殖 している対数期の 3T3-L1細胞でも発現量が少なく、 細胞の増殖が止まったコン フルェントや分化した脂肪細胞では発現量が亢進していることから、 この #103 遺伝子は脂肪細胞の分化において重要な役割を演じているのみならず、 一般的に 細胞の増殖 ·休止に関与している、 またはマ一カー遺伝子である可能性がある。 #105では分化誘導後 2日目までは高分子量のバンドが見られるが、 3日目から は低分子量のバンドのみが見られるなど、 脂肪細胞の分化を考える上で重要な意 味がある可能性がある。  Furthermore, Northern blot analysis showed that expression of # 103 was almost temporarily lost in the early stage after induction of differentiation. It is theorized that when 3T3-L1 is induced to differentiate into adipocytes, some cells will first divide and proliferate and become adipocytes. In addition, the expression level is low in actively growing logarithmic 3T3-L1 cells, and the expression level is increased in confluent cells in which cell growth has stopped or in differentiated adipocytes. It may not only play an important role in cell differentiation, but may also be involved in cell growth and arrest in general, or may be a marker gene. In # 105, high-molecular-weight bands are observed until the second day after differentiation induction, but only low-molecular-weight bands are seen from the third day, indicating that there is an important meaning in considering adipocyte differentiation. There is.
また、 インスリン添加により #105遺伝子の発現が抑制されたことから、 イン スリンを介したシグナル伝達にこの遺伝子が関与している可能性があり、 糖尿病 等の治療や予防にとつて重要な意味がある可能性がある。  In addition, since the addition of insulin suppressed the expression of the # 105 gene, this gene may be involved in insulin-mediated signal transduction, which has important implications for the treatment and prevention of diabetes and other diseases. There may be.
また、 #104遺伝子のヒトホモログの塩基配列がジーンバンクに登録され、 そ の説明によると肝臓の系で癌化によって発現が亢進するということである。 様々 な癌の細胞株で #104の発現が見られ、 その発現パターンが異なっていた。 癌の 種類によってこの #104遺伝子の発現が見られるものとそうでないものがあり、 この #104遺伝子と癌との間に何らかの因果関係がある可能性がある。  In addition, the nucleotide sequence of the human homolog of the # 104 gene has been registered in the gene bank, and according to the description, the expression is enhanced by canceration in the liver system. Expression of # 104 was observed in various cancer cell lines, and their expression patterns were different. Depending on the type of cancer, the expression of the # 104 gene may or may not be seen, and there may be some causal relationship between the # 104 gene and cancer.
本発明者等により単離された #10Xクローンのうち、 #101、 鍵、 および #107 は全長配列ではない。 しかしながら、 当業者においては、 これら断片に対応する 全長遺伝子を単離する手法は技術常識となっている。 例えば、 当業者であれば、 これら断片をプローブとしてコロニーハイブリダィゼーシヨンを行なうことによ り全長遺伝子を単離することができる。 従って、 本発明には、 これら断片に対応 する全長遺伝子が含まれる。 Of the # 10X clones isolated by the inventors, # 101, key, and # 107 are not full-length sequences. However, those skilled in the art will Techniques for isolating full-length genes have become common technical knowledge. For example, those skilled in the art can isolate the full-length gene by performing colony hybridization using these fragments as probes. Therefore, the present invention includes full-length genes corresponding to these fragments.
本発明は、 また、 #10X蛋白質 (配列番号: 8〜1 1に記載のアミノ酸配列か らなる蛋白質、 配列番号: 1, 2 , 7に対応する全長 MAがコードする蛋白質 ) と機能的に同等な蛋白質を包含する。 このような蛋白質には、 例えば、 これら 蛋白質の変異体、 マウス以外の生物のホモログ等が含まれる。 ここで 「機能的に 同等」 とは、 対象となる蛋白質が #10X蛋白質と同様に、 シグナル配列を有する 蛋白質としての機能を有することを指す。 このような機能としては、 例えば、 国 際公開番号 W0 00/00610に記載されている機能や分泌蛋白質としての機能が考 えられる。 目的の蛋白質が分泌蛋白質であるか否かは、 以下の手法により判定す ることができる。  The present invention also provides a functionally equivalent to # 10X protein (a protein consisting of the amino acid sequence of SEQ ID NOs: 8 to 11; a protein encoded by full-length MA corresponding to SEQ ID NOs: 1, 2, and 7). Various proteins. Such proteins include, for example, mutants of these proteins, homologs of organisms other than mice, and the like. Here, “functionally equivalent” means that the target protein has a function as a protein having a signal sequence, like the # 10X protein. As such a function, for example, a function described in International Publication No. WO 00/00610 or a function as a secretory protein can be considered. Whether or not the target protein is a secretory protein can be determined by the following method.
まず、 #10X遺伝子の 3,末端に市販のペプチド (例えば、 His-tagあるいは FL AG等) をコードする遺伝子を連結させた融合遗伝子を作製する。 次に、 その融 合遺伝子を動物細胞用発現ベクター (例えば、 pcDNA3あるいは pCOS- 1等) を用 いて動物細胞 (例えば、 COS細胞等) に導入し、 #10X蛋白質をペプチドとの融 合蛋白質として発現させる。 この融合蛋白質が培養上清中に分泌するか否かを、 該ペプチドに対する抗体を用いて ELISA、 ウエスタンブロッテイング、 あるいは 免疫沈降によって評価する。  First, a fusion gene in which a gene encoding a commercially available peptide (eg, His-tag or FLAG) is ligated to the 3rd end of the # 10X gene is prepared. Next, the fusion gene is introduced into animal cells (eg, COS cells, etc.) using an expression vector for animal cells (eg, pcDNA3 or pCOS-1), and the # 10X protein is converted as a fusion protein with the peptide. To be expressed. Whether or not this fusion protein is secreted into the culture supernatant is evaluated by ELISA, Western blotting or immunoprecipitation using an antibody against the peptide.
分泌蛋 S質には、 種々の産業上の利点がある。 例えば、 ある組換え蛋白質を取 得することを望む場合に、 該蛋白質を分泌蛋白質またはその分泌能を有する部分 ペプチドとの融合蛋白質として細胞内で発現させれば、 融合蛋白質が細胞外へ分 泌され、 組換え蛋白質の精製が容易になるという利点がある。 また、 多くの分泌 蛋白質が有用な医薬品となっていることから、 それ自体に医薬品としての応用も 考えられる。 本発明の蛋白質は、 脂肪細胞に由来すると共に、 脂肪細胞の分化に関連した発 現を示した。 従って、 本発明の遺伝子や蛋白質、 あるいは該蛋白質の発現や活性 を調節する化合物は、 これらに制限されないが、 例えば、 肥満、 高脂血症、 糖尿 病、 動脈硬化などの疾患の治療や予防への応用が考えられる。 Secretory protein S has various industrial advantages. For example, when it is desired to obtain a certain recombinant protein, if the protein is expressed in a cell as a fusion protein with a secretory protein or a partial peptide capable of secreting the same, the fusion protein is secreted out of the cell. However, there is an advantage that the purification of the recombinant protein is facilitated. In addition, since many secreted proteins are useful drugs, they can be applied as drugs themselves. The protein of the present invention was derived from adipocytes and showed expression associated with adipocyte differentiation. Accordingly, the gene or protein of the present invention, or a compound that regulates the expression or activity of the protein, is not limited thereto. For example, for the treatment or prevention of diseases such as obesity, hyperlipidemia, diabetes, and arteriosclerosis. The application of is considered.
ある蛋白質と機能的に同等な蛋白質を調製するための、 当業者によく知られた 方法としては、 蛋白質に変異を導入する方法が知られている。 例えば、 当業者で あれば、 部位特異的変異誘発法 (Hashimoto-Gotoh5 T. et al . ( 1995 ) Gene 152, 271-275, Zoller,MJ, and Smith,M. ( 1983) Methods Enzymol . 100, 468-500 、 Kramer, W. et al. ( 1984) Nucleic Acids Res . 12, 944卜 9456、 Kramer W, a nd Fritz HJ( 1987) Methods. Enzymol . 154, 350-367, Kunkel, TA( 1985 ) Proc Natl Acad Sci USA. 82, 488-492、 Kunkel ( 1988) Methods Enzymol . 85, 276 3-2766) などを用いて、 #10X蛋白質 (配列番号: 8〜1 1に記載のアミノ酸配 列からなる蛋白質、 配列番号: 1 , 2, 7に対応する全長 DNAがコードする蛋 白質) のアミノ酸に適宜変異を導入することにより、 該蛋白質と機能的に同等な 蛋白質を調製することができる。 また、 アミノ酸の変異は自然界においても生じ うる。 このように、 #10X蛋白質のアミノ酸配列において 1もしくは複数のアミ ノ酸が変異したアミノ酸配列を有し、 該蛋白質と機能的に同等な蛋白質もまた本 発明の蛋白質に含まれる。 このような変異体における、 変異するアミノ酸数は、 通常、 50アミノ酸以内であり、 好ましくは 30アミノ酸以内であり、 さらに好ま しくは 10アミノ酸以内 (例えば、 5アミノ酸以内) であると考えられる。 As a method well known to those skilled in the art for preparing a protein functionally equivalent to a certain protein, a method of introducing a mutation into a protein is known. For example, those skilled in the art can use a site-directed mutagenesis method (Hashimoto-Gotoh 5 T. et al. (1995) Gene 152, 271-275, Zoller, MJ, and Smith, M. (1983) Methods Enzymol. 100). , 468-500, Kramer, W. et al. (1984) Nucleic Acids Res. 12, 944 9456, Kramer W, and Fritz HJ (1987) Methods.Enzymol. 154, 350-367, Kunkel, TA (1985) ) Proc Natl Acad Sci USA. 82, 488-492, Kunkel (1988) Methods Enzymol. 85, 276 3-2766) and the like, using the # 10X protein (from the amino acid sequence described in SEQ ID NOs: 8 to 11). A protein functionally equivalent to the protein can be prepared by appropriately mutating amino acids of the protein (a protein encoded by full-length DNA corresponding to SEQ ID NOs: 1, 2, and 7). Amino acid mutations can also occur in nature. As described above, a protein having an amino acid sequence in which one or more amino acids are mutated in the amino acid sequence of the # 10X protein and functionally equivalent to the protein is also included in the protein of the present invention. The number of amino acids to be mutated in such a mutant is usually within 50 amino acids, preferably within 30 amino acids, and more preferably within 10 amino acids (eg, within 5 amino acids).
変異するァミノ酸残基においては、 ァミノ酸側鎖の性質が保存されている別の アミノ酸に変異されることが望ましい。 例えばアミノ酸側鎖の性質としては、 疎 水性アミノ酸 (A、 I、 L、 M、 F、 P、 W、 Y、 V) 、 親水性アミノ酸 (R、 D、 N、 C、 E、 Q G、 H、 K、 S、 T) 、 脂肪族側鎖を有するアミノ酸 ( A、 V、 L、 I、 P) 、 水酸基含有側鎖を有するアミノ酸 (S、 Τ、 Υ) 、 硫黄原子含有側鎖を有するアミ ノ酸 ( Μ) 、 カルボン酸及びアミ ド含有側鎖を有するアミノ酸 (D、 N、 E、 Q ) 、 塩基含有側鎖を有するアミノ離 (R、 K、 Η) 、 芳香族含有側鎖を有するアミ ノ酸 (H、 F、 Y、 ) を挙げることができる (括弧内はいずれもアミノ酸の一文 字標記を表す) 。 It is desirable that the amino acid residue to be mutated is mutated to another amino acid that preserves the properties of the amino acid side chain. For example, the properties of amino acid side chains include hydrophobic amino acids (A, I, L, M, F, P, W, Y, V) and hydrophilic amino acids (R, D, N, C, E, QG, H, K, S, T), an amino acid having an aliphatic side chain (A, V, L, I, P), an amino acid having a hydroxyl group-containing side chain (S, Τ, Υ), an amino acid having a sulfur atom-containing side chain Acids (Μ), carboxylic acids and amino acids with amide-containing side chains (D, N, E, Q ), Amino acids having base-containing side chains (R, K, Η), and amino acids having aromatic-containing side chains (H, F, Y,). Character mark).
あるアミノ酸配列に対する 1又は複数個のアミノ酸残基の欠失、 付加及び/又 は他のアミノ酸による置換により修飾されたアミノ酸配列を有する蛋白質がその 生物学的活性を維持することはすでに知られている (Mark,D.F. et al . , Proc. Nat 1. Acad. Sci . USA ( 1984) 81, 5662-5666、 Zoller3M.J. & Smith,M. Nucleic Acids Research (1982) 10, 6487-6500、 Wang, A. et al. , Science 224, 1431 -1433、 Dalbadie-McFarland, G. et al. , Proc. Natl . Acad. Sci . USA ( 1982) 79, 6409-6413)。 It is already known that a protein having an amino acid sequence modified by deletion, addition and / or substitution of one or more amino acid residues relative to an amino acid sequence maintains its biological activity. Nat. Acad. Sci. USA (1984) 81, 5662-5666, Zoller 3 MJ & Smith, M. Nucleic Acids Research (1982) 10, 6487-6500, Wang, A. et al., Science 224, 1431-1433, Dalbadie-McFarland, G. et al., Proc. Natl. Acad. Sci. USA (1982) 79, 6409-6413).
#1 OX蛋白質のアミノ酸配列に複数個のアミノ酸残基が付加された蛋白質には # 1 For proteins with multiple amino acid residues added to the amino acid sequence of OX protein
、 これら蛋白質を含む融合蛋白質が含まれる。 融合蛋白質は、 これら蛋白質と他 のペプチド又は蛋白質とが融合したものであり、 本発明に含まれる。 融合蛋白質 を作製する方法は、 #10X蛋白質 (配列番号: 8〜1 1に記載のアミノ酸配列か らなる蛋白質、 配列番号: 1 , 2 , 7に対応する全長 DNAがコードする蛋白質 ) をコードする DNAと他のぺプチド又は蛋白質をコードする DNAをフレームが 一致するように連結してこれを発現ベクターに導入し、 宿主で発現させればよく 、 当業者に公知の手法を用いることができる。 本発明の蛋白質との融合に付され る他のぺプチド又は蛋白質としては、 特に限定されない。 And fusion proteins containing these proteins. The fusion protein is a fusion of these proteins with another peptide or protein, and is included in the present invention. The fusion protein is prepared by encoding the # 10X protein (a protein consisting of the amino acid sequence of SEQ ID NOs: 8 to 11; a protein encoded by the full-length DNA corresponding to SEQ ID NOs: 1, 2, and 7). DNA and DNA encoding another peptide or protein may be ligated in frame so that they are introduced into an expression vector and expressed in a host, and a method known to those skilled in the art can be used. Other peptides or proteins to be fused with the protein of the present invention are not particularly limited.
本発明の蛋白質との融合に付される他のペプチドとしては、 例えば、 FLAG (H ορρ,Τ.Ρ. et al . , BioTechnology ( 1988) 6, 1204-1210)、 6個の His (ヒスチ ジン) 残基からなる 6 xHis、 !O xHiS インフルエンザ凝集素 (HA) 、 ヒト c-m ycの断片、 VSV- GPの断片、 pl8HIVの断片、 T7- tag、 HSV- tag、 E- tag、 SV40T抗 原の断片、 lck tag、 a- tubulinの断片、 B-tag、 Protein Cの断片等の公知の ペプチドを使用することができる。 また、 本発明の蛋白質との融合に付される他 の蛋白質としては、 例えば、 GST (グル夕チオン一S—トランスフェラーゼ) 、 H A (インフルエンザ凝集素) 、 ィムノグロブリン定常領域、 ?一ガラクトシダー ゼ、 MBP (マルト一ス結合蛋白質) 等が挙げられる。 市販されているこれらぺプ チドまたは蛋白質をコードする MAを本発明の蛋白質をコ一ドする DNAと融合 させ、 これにより調製された融合 DNAを発現させることにより、 融合蛋白質を 調製することができる。 Other peptides to be fused to the protein of the present invention include, for example, FLAG (Høρρ, Τ.Ρ. et al., BioTechnology (1988) 6, 1204-1210) and 6 His (histidine). ) Residues consisting of 6 xHis,! O xHiS influenza agglutinin (HA), human cmyc fragment, VSV-GP fragment, pl8HIV fragment, T7-tag, HSV-tag, E-tag, SV40T antigen Known peptides such as fragments, lck tags, a-tubulin fragments, B-tags, and Protein C fragments can be used. Other proteins to be fused with the protein of the present invention include, for example, GST (Gluythione-S-transferase), H A (influenza agglutinin), immunoglobulin constant region, 領域 -galactosidase, MBP (maltose binding protein) and the like. A fusion protein can be prepared by fusing commercially available MA encoding the peptide or the protein with DNA encoding the protein of the present invention, and expressing the fusion DNA prepared thereby. .
また、 #10X蛋白質のァミノ酸配列から複数個のァミノ酸残墓が欠失した蛋白 質には、 これら蛋白質からシグナル配列が除去された蛋白質が含まれる。  In addition, proteins in which a plurality of amino acid residues are deleted from the amino acid sequence of the # 10X protein include proteins in which signal sequences have been removed from these proteins.
ある蛋白質と機能的に同等な蛋白質を調製する当業者によく知られた他の方法 としては、 ハイブリダィゼ一シヨン技術 (Sambrook,J et al. , Molecular Clon ing 2nd ed. , 9.47-9.58, Cold Spring Harbor Lab. press, 1989) を利用する 方法が挙げられる。 即ち、 当業者であれば、 #10X蛋白質をコードする DNA配列 (配列番号: 1〜7 ) もしくはその一部を基に、 これと相同性の高い DNAを単 離して、 該 DNAから #10X蛋白質と機能的に同等な蛋白質を単離することも通常 行いうることである。  Other methods well known to those skilled in the art for preparing proteins functionally equivalent to a protein include the hybridization technique (Sambrook, J et al., Molecular Cloning 2nd ed., 9.47-9.58, Cold Spring Harbor Lab. Press, 1989). That is, those skilled in the art can isolate a DNA having high homology with the DNA sequence (SEQ ID NOS: 1 to 7) encoding the # 10X protein or a part thereof, and extract the # 10X protein from the DNA. Isolation of a protein that is functionally equivalent to this can also usually be performed.
本発明には、 讓蛋白質をコードする MAとハイブリダィズする DNAがコー ドし、 #10X蛋白質と機能的に同等な蛋白質が含まれる。 このような蛋白質とし ては、 例えば、 マウスおよび他の哺乳動物のホモログ (例えば、 ヒト、 ラヅト、 ゥサギ、 ゥシなどがコードする蛋白質) が挙げられる。  The present invention includes proteins encoded by DNA that hybridizes to MA encoding the protein and functionally equivalent to the # 10X protein. Such proteins include, for example, homologs of mouse and other mammals (eg, proteins encoded by humans, rodents, rabbits, magpies, etc.).
#10X蛋白質と機能的に同等な蛋白質をコードする MAを単離するためのハイ ブリダィゼーシヨンの条件は、 当業者であれば適宜選択することができる。 ハイ ブリダィゼ一シヨンの条件としては、 例えば、 低ストリンジヱントな条件が挙げ られる。 低ストリンジェントな条件とは、 ハイブリダィゼーシヨン後の洗浄にお いて、 例えば 42°C、 O. l SSC 0.1 %SDSの条件であり、 好ましくは 50°C、 0.1 x SSC、 0.1%SDSの条件である。 より好ましいハイブリダィゼ一シヨンの条件 としては、 高ストリンジヱントな条件が挙げられる。 高ストリンジヱントな条件 とは、 例えば 65°C、 5 X SSC及び 0.1 %SDSの条件である。 これらの条件におい て、 温度を上げる程に高い相同性を有する DNAが効率的に得られることが期待 できる。 但し、 ハイブリダィゼ一シヨンのストリンジエンシーに影響する要素と しては温度や塩濃度など複数の要素が考えられ、 当業者であればこれら要素を適 宜選択することで同様のストリンジエンシーを実現することが可能である。 Hybridization conditions for isolating MA encoding a protein functionally equivalent to the # 10X protein can be appropriately selected by those skilled in the art. The conditions for the hybridization include, for example, low stringent conditions. The low stringent conditions are, for example, conditions of 42 ° C and 0.1% SDS of O.l SSC in washing after hybridization, preferably 50 ° C, 0.1 x SSC and 0.1% SDS. Is the condition. More preferable hybridization conditions include high stringent conditions. Highly stringent conditions are, for example, conditions of 65 ° C., 5 × SSC and 0.1% SDS. In these conditions Thus, it can be expected that DNA with higher homology can be obtained more efficiently as the temperature is increased. However, a plurality of factors such as temperature and salt concentration can be considered as factors affecting the stringency of the hybridization, and those skilled in the art can realize similar stringency by appropriately selecting these factors. It is possible to
また、 ハイプリダイゼ一シヨンにかえて、 #10X蛋白質をコードする DNA (配 列番号: 1から 7 ) の配列情報を基に合成したプライマーを用いる遺伝子増幅法 、 例えば、 ポリメラーゼ連鎖反応 (PCR) 法を利用して単離することも可能であ る。  In addition, a gene amplification method using primers synthesized based on the sequence information of DNA (SEQ ID NOS: 1 to 7) encoding the # 10X protein instead of the hybridization method, for example, polymerase chain reaction (PCR) method It can also be used for isolation.
これらハイプリダイゼーシヨン技術や遺伝子増幅技術により単離される DNA がコードする、 #10X蛋白質と機能的に同等な蛋白質は、 通常、 #10X蛋白質 (配 列番号: 8〜 1 1に記載のアミノ酸配列からなる蛋白質、 配列番号: 1, 2 , 7 に対応する全長 DNAがコードする蛋白質) とアミノ酸配列において高い相同性 を有する。 本発明の蛋白質には、 #10X蛋白質と機能的に同等であり、 かつ該蛋 白質のアミノ酸配列と高い相同性を有する蛋白質も含まれる。 高い相同性とは、 アミノ酸レベルにおいて、 通常、 少なくとも 50%以上の同一性、 好ましくは 75 %以上の同一性、 さらに好ましくは 85%以上の同一性、 さらに好ましくは 95% 以上の同一性を指す。 アミノ酸配列や塩基配列の同一性は、 Karl in and Altsch ulによるアルゴリズム BLAST(Proc. Natl. Acad. Sei. USA 90: 5873-5877, 199 3)によって決定することができる。 このアルゴリズムに基づいて、 BLASTNや BL ASTXと呼ばれるプログラムが開発されている(Altschul et al. J. Mol . Biol . 215 :403-410, 1990)。 BLASTNによって塩基配列を解析する場合には、 パラメ一 夕一はたとえば score = 100、 wordlength = 12とする。 また、 BLASTXによつ てアミノ酸配列を解析する場合には、 パラメ一夕一はたとえば score = 50、 wor dlength = 3とする。 BLASTと Gapped BLASTプログラムを用いる場合には、 各 プログラムのデフォルトパラメ一夕一を用いる。 これらの解析方法の具体的な手 法は公知である (https://www.ncbi .nlm.nih.gov. )0 本発明の蛋白質は、 後述するそれを産生する細胞や宿主あるいは精製方法によ り、 アミノ酸配列、 分子量、 等電点又は糖鎖の有無や形態などが異なり得る。 し かしながら、 得られた蛋白質が、 #10X蛋白質と同等の機能を有している限り、 本発明に含まれる。 例えば、 本発明の蛋白質を原核細胞、 例えば大腸菌で発現さ せた場合、 本来の蛋白質のアミノ酸配列の N末端にメチォニン残基が付加され る。 本発明の蛋白質はこのような蛋白質も包含する。 Proteins functionally equivalent to the # 10X protein encoded by DNA isolated by these hybridization techniques or gene amplification techniques are usually the # 10X protein (the amino acid sequence described in SEQ ID NOs: 8 to 11). , A protein encoded by the full-length DNA corresponding to SEQ ID NOs: 1, 2, and 7) and has a high homology in the amino acid sequence. The proteins of the present invention also include proteins that are functionally equivalent to the # 10X protein and that have high homology to the amino acid sequence of the protein. High homology usually means at least 50% identity, preferably 75% identity, more preferably 85% identity, and even more preferably 95% identity at the amino acid level. . The identity of amino acid sequences and nucleotide sequences can be determined by the algorithm BLAST (Proc. Natl. Acad. Sei. USA 90: 5873-5877, 1993) by Karl in and Altschul. Based on this algorithm, programs called BLASTN and BL ASTX have been developed (Altschul et al. J. Mol. Biol. 215: 403-410, 1990). When analyzing the nucleotide sequence by BLASTN, the parameters are, for example, score = 100 and wordlength = 12. When analyzing amino acid sequences by BLASTX, the parameters are, for example, score = 50 and wordlength = 3. When using BLAST and Gapped BLAST programs, use the default parameters of each program. Specific methods of these analysis methods are known (https://www.ncbi.nlm.nih.gov.) 0 The protein of the present invention may differ in amino acid sequence, molecular weight, isoelectric point, presence / absence and form of sugar chains, etc., depending on the cell, host, or purification method for producing the protein described below. However, as long as the obtained protein has a function equivalent to that of the # 10X protein, it is included in the present invention. For example, when the protein of the present invention is expressed in a prokaryotic cell, for example, Escherichia coli, a methionine residue is added to the N-terminal of the amino acid sequence of the original protein. The proteins of the present invention also include such proteins.
本発明の蛋白質は、 当業者に公知の方法により、 組み換え蛋白質として、 また 天然の蛋白質として調製することが可能である。 組み換え蛋白質であれば、 本発 明の蛋白質をコードする DNA (例えば、 配列番号: 1から 7に記載の塩基配列を 有する DNA)を、 適当な発現ベクターに組み込み、 これを適当な宿主細胞に導入 して得た形質転換体を回収し、 抽出物を得た後、 イオン交換、 逆相、 ゲル濾過な どのクロマトグラフィー、 あるいは本発明の蛋白質に対する抗体をカラムに固定 したァフィ二ティ一クロマトグラフィーにかけることにより、 または、 さらにこ れらのカラムを複数組み合わせることにより精製し、 調製することが可能である また、 本発明の蛋白質をグル夕チオン S-トランスフェラーゼ蛋白質との融合 蛋白質として、 あるいはヒスチジンを複数付加させた組み換え蛋白質として宿主 細胞 (例えば、 動物細胞や大腸菌など) 内で発現させた場合には、 発現させた組 み換え蛋白質はグル夕チオンカラムあるいはニッケルカラムを用いて精製するこ とができる。 融合蛋白質の精製後、 必要に応じて融合蛋白質のうち、 目的の蛋白 質以外の領域を、 トロンビンまたはファクター Xaなどにより切断し、 除去する ことも可能である。  The protein of the present invention can be prepared as a recombinant protein or a natural protein by methods known to those skilled in the art. If it is a recombinant protein, a DNA encoding the protein of the present invention (for example, a DNA having the nucleotide sequence of SEQ ID NOS: 1 to 7) is inserted into an appropriate expression vector and introduced into an appropriate host cell. The resulting transformant is collected and an extract is obtained, which is then subjected to chromatography such as ion exchange, reverse phase, gel filtration, or affinity chromatography in which an antibody against the protein of the present invention is immobilized on a column. The protein of the present invention can be used as a fusion protein with glutathione S-transferase protein or histidine. When expressed in host cells (eg, animal cells, E. coli, etc.) as multiple added recombinant proteins , The set seen recombinant protein expressed can and child purified using Guru evening Chionkaramu or nickel column. After purification of the fusion protein, if necessary, regions of the fusion protein other than the target protein can be cleaved with thrombin or Factor Xa and removed.
天然の蛋白質であれば、 当業者に周知の方法、 例えば、 本発明の蛋白質を発現 している組織や細胞の抽出物に対し、 後述する本発明の蛋白質に結合する抗体が 結合したァフィ二ティ一カラムを作用させて精製することにより単離することが できる。 抗体はポリク口一ナル抗体であってもモノクローナル抗体であってもよ い。 If the protein is a natural protein, a method known to those skilled in the art, for example, an affinity in which an antibody that binds to the protein of the present invention described below is bound to a tissue or cell extract expressing the protein of the present invention, as described below. Isolation can be achieved by purifying using one column. it can. The antibody may be a polyclonal antibody or a monoclonal antibody.
本発明は、 また、 本発明の蛋白質の部分ペプチドを包含する。 本発明の部分べ プチドは、 少なくとも 7アミノ酸以上、 好ましくは 8アミノ酸以上、 さらに好ま しくは 9アミノ酸以上のアミノ酸配列からなる。 該部分ペプチドは、 例えば、 本 発明の蛋白質に対する抗体の作製、 本発明の蛋白質に結合する化合物のスクリー ニングゃ、 本発明の蛋白質の促進剤や阻害剤のスクリーニングに利用し得る。 ま た、 本発明の蛋白質のアン夕ゴニストや競合阻害剤になり得る。 本発明の部分べ プチドは、 遺伝子工学的手法、 公知のペプチド合成法、 あるいは本発明の蛋白質 を適切なぺプチダーゼで切断することによって製造することができる。 ぺプチド の合成は、 例えば、 固相合成法、 液相合成法のいずれによってもよい。  The present invention also includes partial peptides of the protein of the present invention. The partial peptide of the present invention has an amino acid sequence of at least 7 amino acids or more, preferably 8 amino acids or more, and more preferably 9 amino acids or more. The partial peptide can be used, for example, for preparing an antibody against the protein of the present invention, for screening a compound binding to the protein of the present invention, and for screening for a promoter or inhibitor of the protein of the present invention. Further, it can be an antagonist of the protein of the present invention or a competitive inhibitor. The partial peptide of the present invention can be produced by a genetic engineering technique, a known peptide synthesis method, or by cleaving the protein of the present invention with an appropriate peptidase. The peptide may be synthesized by, for example, either a solid phase synthesis method or a liquid phase synthesis method.
本発明の蛋白質をコードする DNAは、 上述したような本発明の蛋白質の in vi voや in v roにおける生産に利用される他、 例えば、 本発明の蛋白質をコード する遺伝子の異常に起因する疾患や本発明の蛋白質により治療可能な疾患の遺伝 子治療などへの応用も考えられる。 本発明の DNAは、 本発明の蛋白質をコード しうるものであればいかなる形態でもよい。 即ち、 mRNAから合成された cDNAで あるか、 ゲノム DNAであるか、 化学合成 DNAであるかなどを問わない。 また、 本発明の蛋白質をコードしうる限り、 遺伝暗号の縮重に基づく任意の塩基配列を 有する MAが含まれる。  The DNA encoding the protein of the present invention is used for in vivo and in vitro production of the protein of the present invention as described above, and also includes, for example, diseases caused by abnormalities in the gene encoding the protein of the present invention. And gene therapy for diseases treatable by the protein of the present invention. The DNA of the present invention may be in any form as long as it can encode the protein of the present invention. That is, it does not matter whether it is cDNA synthesized from mRNA, genomic DNA, or chemically synthesized DNA. Also, as long as it can encode the protein of the present invention, MA having an arbitrary nucleotide sequence based on the degeneracy of the genetic code is included.
本発明の DNAは、 当業者に公知の方法により調製することができる。 例えば 、 本発明の蛋白質を発現している細胞より cDNAライブラリーを作製し、 本発明 の DNAの配列 (例えば、 配列番号: 1から 7 ) の一部をプローブにしてハイブ リダィゼーシヨンを行うことにより調製できる。 cDNAライブラリ一は、 例えば 、 文献 (Sambrook, J. et al . , Molecular Clonings Cold Spring Harbor Labo ratory Press (1989)) に記載の方法により調製してもよいし、 市販の DNAライ ブラリーを用いてもよい。 また、 本発明の蛋白質を発現している細胞より RNA を調製し、 逆転写酵素により cDNAを合成した後、 本発明の DNAの配列 (例えば 、 配列番号: 1から 7 ) に基づいてオリゴ DNAを合成し、 これをプライマーと して用いて PCR反応を行い、 本発明の蛋白質をコ一ドする cDNAを増幅させるこ とにより調製することも可能である。 The DNA of the present invention can be prepared by a method known to those skilled in the art. For example, it is prepared by preparing a cDNA library from cells expressing the protein of the present invention and performing hybridization using a part of the sequence of the DNA of the present invention (for example, SEQ ID NOS: 1 to 7) as a probe. it can. The cDNA library may be prepared, for example, by the method described in the literature (Sambrook, J. et al., Molecular Clonings Cold Spring Harbor Laboratory Press (1989)), or using a commercially available DNA library. Good. In addition, cells expressing the protein of the present invention After synthesizing cDNA using reverse transcriptase, oligo DNA is synthesized based on the sequence of the DNA of the present invention (for example, SEQ ID NOS: 1 to 7), and the PCR reaction is performed using this as a primer. It can also be prepared by amplifying a cDNA encoding the protein of the present invention.
また、 得られた cDNAの塩基配列を決定することにより、 それがコードする翻 訳領域を決定でき、 本発明の蛋白質のアミノ酸配列を得ることができる。 また、 得られた cDNAをプローブとしてゲノム DNAライブラリーをスクリーニングする ことにより、 ゲノム DNAを単離することができる。  Also, by determining the nucleotide sequence of the obtained cDNA, the translation region encoded by the cDNA can be determined, and the amino acid sequence of the protein of the present invention can be obtained. Genomic DNA can be isolated by screening a genomic DNA library using the obtained cDNA as a probe.
具体的には、 次のようにすればよい。 まず、 本発明の蛋白質を発現する細胞、 組織、 臓器 (例えば、 脂肪細胞や本実施例におけるノザンブロッテイングにより 発現が認められた組織) から、 mRNAを単離する。 mRNAの単離は、 公知の方法、 例えば、 グァニジン超遠心法(Chirgwin,J.M. et al ., Biochemistry ( 1979 ) 18 , 5294-5299), AGPC法(Chomczynski,P. and Sacchi,N. 3 Anal. Biochem. ( 1987 ) 162, 156- 159)等により全 RNAを調製し、 mRNA Purification Kit(Pharmacia) 等を使用して全 RNAから mRNAを精製する。 また、 QuickPrep mRNA Purificatio n Kit(Pharmacia)を用いることにより mRNAを直接調製することもできる。 Specifically, the following may be performed. First, mRNA is isolated from a cell, tissue, or organ that expresses the protein of the present invention (eg, an adipocyte or a tissue in which expression has been observed by Northern blotting in this example). mRNA can be isolated by known methods, for example, guanidine ultracentrifugation (Chirgwin, JM et al., Biochemistry (1979) 18, 5294-5299), and AGPC method (Chomczynski, P. and Sacchi, N. 3 Anal. Total RNA is prepared by Biochem. (1987) 162, 156-159) and the like, and mRNA is purified from the total RNA using the mRNA Purification Kit (Pharmacia). Alternatively, mRNA can be directly prepared by using the QuickPrep mRNA Purification Kit (Pharmacia).
得られた mRNAから逆転写酵素を用いて cDNAを合成する。 cDNAの合成は、 AMV CDNA is synthesized from the obtained mRNA using reverse transcriptase. cDNA synthesis, AMV
Reverse Transcriptase First-strand cDNA Synthesis Kit (生化学工業)等を 用いて行うこともできる。 また、 本明細書に記載されたプライマー等を用いて、 5,- Ampli FINDER RACE Kit(Clontech製)およびポリメラーゼ連鎖反応 (polymer ase chain reaction; PCR) を用いた 5, - RACE法 (Frohman,M.A. et al . , Proc .N atl.Acad. Sci . U. S.A. ( 1988) 85, 8998-9002; Belyavsky,A. et al . , NucleicReverse Transcriptase First-strand cDNA Synthesis Kit (Seikagaku Corporation) can also be used. In addition, using the primers and the like described in the present specification, the 5, -RACE method (Frohman, MA) using the 5, -Ampli FINDER RACE Kit (manufactured by Clontech) and the polymerase chain reaction (polymerase chain reaction; PCR). Acad. Sci. USA (1988) 85, 8998-9002; Belyavsky, A. et al., Nucleic.
Acids Res . ( 1989) 17, 2919- 2932)にしたがい、 cDNAの合成および増幅を行 うことができる。 According to Acids Res. (1989) 17, 2919-2932), cDNA can be synthesized and amplified.
得られた PCR産物から目的とする DNA断片を調製し、 ベクター DNAと連結する 。 さらに、 これより組換えベクターを作製し、 大腸菌等に導入してコロニーを選 択して所望の組換えべクタ一を調製する。 目的とする DNAの塩基配列は、 公知 の方法、 例えば、 ジデォキシヌクレオチドチヱインターミネ一シヨン法により確 認することができる。 A target DNA fragment is prepared from the obtained PCR product, and ligated to a vector DNA. Furthermore, a recombinant vector is prepared from this, introduced into E. coli, etc., and colonies are selected. To prepare the desired recombinant vector. The base sequence of the target DNA can be confirmed by a known method, for example, the dideoxynucleotide chain termination method.
また、 本発明の DNAにおいては、 発現に使用する宿主のコドン使用頻度を考 慮して、 より発現効率の高い塩基配列を設計することができる (Grantham,R. e t al . 3 Nucelic Acids Research ( 1981 ) 9, r43-74) 。 また、 本発明の MAは 、 市販のキットや公知の方法によって改変することができる。 改変としては、 例 えば、 制限酵素による消化、 合成オリゴヌクレオチドや適当な DNAフラグメン 卜の挿入、 リンカ一の付加、 開始コドン (ATG) 及び/又は終止コドン (TAA、 T GA、 又は TAG) の挿入等が挙げられる。 In the DNA of the present invention, a nucleotide sequence with higher expression efficiency can be designed in consideration of the codon usage of the host used for expression (Grantham, R. et al. 3 Nucelic Acids Research ( 1981) 9, r43-74). The MA of the present invention can be modified by a commercially available kit or a known method. Modifications include, for example, digestion with restriction enzymes, insertion of synthetic oligonucleotides or appropriate DNA fragments, addition of a linker, insertion of an initiation codon (ATG) and / or stop codon (TAA, TGA, or TAG). And the like.
本発明の DNAは、 具体的には、 配列番号: 1の塩基配列において 1位の塩基 c から 165位の塩基 gまでの塩基配列からなる DNAまたは該塩基配列を含む DNA、 配列番号: 2の塩基配列において 1位の塩基 から 438位の塩基 tまでの塩基配 列からなる DNAまたは該塩基配列を含む DNA、 配列番号: 3の塩基配列において 83位の塩基 aから 1408位の塩基 aまでの塩基配列からなる DNAまたは該塩基配 列を含む DNA、 配列番号: 4の塩基配列において 214位の塩基 aから 1662位の 塩基 gまでの塩基配列からなる DNAまたは該塩基配列を含む DNA、 配列番号: 5 の塩基配列において 289位の塩基 aから 1407位の塩基 cまでの塩基配列からな る DNAまたは該塩基配列を含む DNA、 配列番号: 6の塩基配列において 86位の 塩基 aから 3244位の塩基 aまでの塩基配列からなる DNAまたは該塩基配列を含 む MA、 および配列番号: 7の塩基配列において 1位の塩基 cから 522位の塩基 cまでの塩基配列からなる DNAまたは該塩基配列を含む DNAを包含する。  Specifically, the DNA of the present invention is a DNA consisting of a base sequence from base c at position 1 to base g at position 165 in the base sequence of SEQ ID NO: 1, or a DNA containing the base sequence, A DNA consisting of a base sequence from the 1st base to the 438th base t in the base sequence or a DNA containing the base sequence, and a base sequence from the 83rd base a to the 1408th base a in the base sequence of SEQ ID NO: 3 DNA consisting of a base sequence or DNA containing said base sequence, DNA consisting of a base sequence from base a at position 214 to base g at position 1662 in base sequence of SEQ ID NO: 4, or DNA containing said base sequence, SEQ ID NO: A DNA consisting of the base sequence from the base a at position 289 to the base c at position 1407 in the base sequence of 5, or a DNA containing the base sequence, and the base a from position 86 to position 3244 of base 86 at position 86 in the base sequence of SEQ ID NO: 6. DNA consisting of a base sequence up to base a or Nucleotide sequence including MA, and SEQ ID NO: encompasses DNA containing the DNA or nucleotide sequence consisting of the nucleotide sequence of the 7 from nucleotide position 1 of the nucleotide c to 522-positional base c.
本発明の DNAはまた、 配列番号: 1から 7に示す塩基配列からなる MAとハイ ブリダィズする MAであり、 且つ上記本発明の蛋白質と機能的に同等な蛋白質 をコードする DNAを含む。 ハイプリダイゼ一シヨンにおける条件は当業者であ れば適宜選択することができるが、 具体的には上記した条件を用レ、ることができ る。 これらの条件において、 温度を上げる程に高い相同性を有する DNAを得る ことができる。 上記のハイブリダィズする DNAは、 好ましくは天然由来の DNA、 例えば cDNA又は染色体 DNAである。 The DNA of the present invention also includes a DNA that hybridizes with the MA having the nucleotide sequence shown in SEQ ID NOs: 1 to 7, and that encodes a protein functionally equivalent to the protein of the present invention. The conditions for the hybridization can be appropriately selected by those skilled in the art. Specifically, the conditions described above can be used. You. Under these conditions, DNA with higher homology can be obtained as the temperature is increased. The hybridizing DNA is preferably a naturally occurring DNA, for example, cDNA or chromosomal DNA.
本発明は、 また、 本発明の MAが挿入されたベクターを提供する。 本発明の ベクターとしては、 宿主細胞内において本発明の DNAを保持したり、 本発明の 蛋白質を発現させるために有用である。  The present invention also provides a vector into which the MA of the present invention has been inserted. The vector of the present invention is useful for retaining the DNA of the present invention in a host cell or expressing the protein of the present invention.
ベクタ一としては、 例えば、 大腸菌を宿主とする場合には、 ベクターを大腸菌 For example, when E. coli is used as a host, the vector
(例えば、 扁 9、 DH5ひ、 翻 1、 XLlBlue) などで大量に増幅させ大量調製す るために、 大腸菌で増幅されるための 「ori」 をもち、 さらに形質転換された大 腸菌の選抜遺伝子 (例えば、 なんらかの薬剤 (アンピシリンやテトラサイクリン 、 カナマイシン、 クロラムフエ二コール) により判別できるような薬剤耐性遺伝 子) を有すれば特に制限はない。 ベクターの例としては、 M13系ベクター、 pUC 系ベクター、 pBR322. pBluescript, pCR-Scriptなどが挙げられる。 また、 cDN Aのサブクローニング、 切り出しを目的とした場合、 上記ベクターの他に、 例え ば、 pGEM- T、 pDIRECT、 pT7などが挙げられる。 本発明の蛋白質を生産する目的 においてベクタ一を使用する場合には、 特に、 発現ベクターが有用である。 発現 ベクターとしては、 例えば、 大腸菌での発現を目的とした場合は、 ベクターが大 腸菌で増幅されるような上記特徴を持つほかに、 宿主を JM109、 DH5 a、 HB101, XU- Blueなどの大腸菌とした場合においては、 大腸菌で効率よく発現できるよ うなプロモ一夕一、 例えば、 lacZプロモー夕一 (Wardら, Nature ( 1989) 341,(E.g., DH9, DH5, Ver.1, XLlBlue) for large-scale amplification and preparation of large amounts of E. coli for `` ori '' amplification and selection of transformed E. coli There is no particular limitation as long as it has a gene (for example, a drug resistance gene that can be identified by any drug (ampicillin, tetracycline, kanamycin, chloramphenicol)). Examples of vectors include M13-based vectors, pUC-based vectors, pBR322. PBluescript, pCR-Script, and the like. In addition, for the purpose of subcloning and excision of cDNA, in addition to the above vectors, examples include pGEM-T, pDIRECT, pT7, and the like. When a vector is used for the purpose of producing the protein of the present invention, an expression vector is particularly useful. For example, when the expression vector is intended for expression in E. coli, the expression vector may have the above-mentioned characteristics such that the vector is amplified in E. coli, and may be used in a host such as JM109, DH5a, HB101, or XU-Blue. In the case of Escherichia coli, a promoter that can be efficiently expressed in Escherichia coli, for example, lacZ promoter, Yuichi (Ward et al., Nature (1989) 341,
544-546; FASEB J. ( 1992) 6, 242Z-2427) 、 araBプロモーター (Betterら, Science ( 1988) 240, 1041-1043) 、 または T7プロモーターなどを持っている ことが不可欠である。 このようなベクターとしては、 上記ベクターの他に pGEX - 5X-1 (フアルマシア社製) 、 「QIAexpress systemj (キアゲン社製) 、 pEGFP 、 または pET (この場合、 宿主は T7 RNAポリメラーゼを発現している BL21が好 ましい)などが挙げられる。 また、 ベクターには、 ポリペプチド分泌のためのシグナル配列が含まれていて もよい。 蛋白質分泌のためのシグナル配列としては、 大腸菌のペリブラズムに産 生させる場合、 pelBシグナル配列 (Lei,S.P. et al J. Bacteriol . ( 1987) 169 , 4379) を使用すればよい。 宿主細胞へのベクターの導入は、 例えば塩化カル シゥム法、 エレクトロポレ一シヨン法を用いて行うことができる。 544-546; it is essential to have FASEB J. (1992) 6, 242Z-2427), araB promoter (Better et al., Science (1988) 240, 1041-1043), or T7 promoter. Such vectors include pGEX-5X-1 (Pharmacia), QIAexpress systemj (Qiagen), pEGFP, or pET (in this case, the host expresses T7 RNA polymerase in addition to the above vectors). BL21 is preferred). The vector may also include a signal sequence for polypeptide secretion. As a signal sequence for protein secretion, the pelB signal sequence (Lei, SP et al J. Bacteriol. (1987) 169, 4379) may be used for production in the periplasm of Escherichia coli. The introduction of the vector into the host cell can be performed using, for example, a calcium chloride method or an electroporation method.
大腸菌以外にも、 例えば、 本発明の蛋白質を製造するためのベクターとしては 、 哺乳動物由来の発現ベクター (例えば、 pcDNA3(インビトロゲン社製) や、 pE GF-BOS(Nucleic Acids. Res.1990, 18( 17) , p5322)、 pEF、 pCDM8) 、 昆虫細胞由 来の発現べク夕一 (例えば 「Bac - to - BAC baculovairus expression systemj ( ギブコ BRL社製) 、 pBacPAK8) 、 植物由来の発現ベクター (例えば ρΜΗ1、 pMH2 ) 、 動物ウィルス由来の発現べクタ一 (例えば、 pHSV、 pMV、 pAdexLcw) 、 レト ロウィルス由来の発現ベクター (例えば、 pZIPneo) 、 酵母由来の発現ベクター (例えば、 rpichia Expression Kitj (インビトロゲン社製) 、 pNVU、 SP-Q0 1) 、 枯草菌由来の発現ベクター (例えば、 pPL608、 pKTH50) が挙げられる。  In addition to Escherichia coli, for example, as a vector for producing the protein of the present invention, a mammalian expression vector (for example, pcDNA3 (manufactured by Invitrogen) or pEGF-BOS (Nucleic Acids. Res. 1990, 18 (17), p5322), pEF, pCDM8), expression vector derived from insect cells (eg, “Bac-to-BAC baculovairus expression systemj (manufactured by Gibco BRL), pBacPAK8), plant-derived expression vector ( For example, ρΜΗ1, pMH2), an expression vector derived from an animal virus (eg, pHSV, pMV, pAdexLcw), an expression vector derived from a retrovirus (eg, pZIPneo), an expression vector derived from a yeast (eg, rpichia Expression Kitj (Invitrogen) ), PNVU, SP-Q01) and Bacillus subtilis-derived expression vectors (eg, pPL608, pKTH50).
CH0細胞、 COS細胞、 NIH3T3細胞等の動物細胞での発現を目的とした場合には 、 細胞内で発現させるために必要なプロモー夕一、 例えば SV40プロモーター ( Mulliganら, Nature ( 1979 ) 277, 108)、 MMLV-LTRプロモーター、 EF1ひプロ モー夕一 (Mizushimaら, Nucleic Acids Res. ( 1990) 18, 5322)、 CMVプロモ —夕一などを持っていることが不可欠であり、 細胞への形質転換を選抜するため の遺伝子 (例えば、 薬剤 (ネオマイシン、 G418など) により判別できるような 薬剤耐性遺伝子) を有すればさらに好ましい。 このような特性を有するベクター としては、 例えば、 p匪、 pDR2、 pBK-RSV, pBK-CMVヽ p0PRSVs p0P13などが挙 げられる。 In the case of expression in animal cells such as CH0 cells, COS cells, and NIH3T3 cells, promoters necessary for expression in cells, such as the SV40 promoter (Muligan et al., Nature (1979) 277, 108) ), MMLV-LTR promoter, EF1 promoter Yuichi (Mizushima et al., Nucleic Acids Res. (1990) 18, 5322), CMV promoter — It is essential to have a promoter and transform cells. It is more preferable to have a gene (for example, a drug resistance gene that can be discriminated by a drug (neomycin, G418, etc.)) for selecting Cp. Examples of vectors with such characteristics include, p negation, pDR2, pBK-RSV, can be mentioned up such pBK-CMVヽp0PRSV s p0P13.
さらに、 遺伝子を安定的に発現させ、 かつ、 細胞内での遺伝子のコピー数の増 幅を目的とする場合には、 核酸合成経路を欠損した CH0細胞にそれを相補する D HFR遺伝子を有するベクタ一 (例えば、 pCHOIなど) を導入し、 メ ト トレキセ一 ト (MTX) により増幅させる方法が挙げられ、 また、 遺伝子の一過性の発現を目 的とする場合には、 SV40 T抗原を発現する遺伝子を染色体上に持つ COS細胞を 用いて SV40の複製起点を持つベクタ一 (pcDなど) で形質転換する方法が挙げ られる。 複製開始点としては、 また、 ポリオ一マウィルス、 アデノウイルス、 ゥ シパピロ一マウィルス (BPV) 等の由来のものを用いることもできる。 さらに、 宿主細胞系で遺伝子コピー数増幅のため、 発現べクタ一は選択マーカーとして、 アミノグリコシドトランスフェラ一ゼ (APH) 遺伝子、 チミジンキナーゼ (TK) 遺伝子、 大腸菌キサンチングァニンホスホリボシルトランスフェラーゼ (Ecogp t) 遺伝子、 ジヒドロ葉酸還元酵素 (dhfr) 遺伝子等を含むことができる。 Furthermore, when the gene is to be stably expressed and the number of copies of the gene is to be increased in a cell, a vector having a DHFR gene complementing the nucleic acid synthesis pathway-deficient CH0 cell is used. (For example, pCHOI) and methotrexe (MTX). In the case of transient expression of a gene, replication of SV40 is performed using COS cells having a gene expressing the SV40 T antigen on the chromosome. Transformation can be performed using a single vector (eg, pcD) having an origin. As the replication origin, those derived from poliovirus, adenovirus, sipapirovirus (BPV) and the like can also be used. In addition, the expression vector is used as a selection marker for amplification of gene copy number in the host cell system, such as aminoglycoside transferase (APH) gene, thymidine kinase (TK) gene, and Escherichia coli xanthinguanine phosphoribosyltransferase (Ecogp t). Gene, dihydrofolate reductase (dhfr) gene and the like.
一方、 動物の生体内で本発明の DNAを発現させる方法としては、 本発明の DNA を適当なベクタ一に組み込み、 例えば、 レトロウイルス法、 リボソーム法、 カチ ォニックリボソーム法、 アデノウイルス法などにより生体内に導入する方法など が挙げられる。 これにより、 本発明の #10X遺伝子の変異に起因する疾患に対す る遺伝子治療を行うことが可能である。 用いられるベクタ一としては、 例えば、 アデノウイルスベクター (例えば pAdexlcw) やレトロウイルスベクタ一(例えば pZIPneo) などが挙げられるが、 これらに制限されない。 ベクタ一への本発明の DNAの挿入などの一般的な遺伝子操作は、 常法に従って行うことが可能である ( Molecular Cloning , 5.61-5.63) 。 生体内への投与は、 ex vivo法であっても 、 in vivo法であってもよい。  On the other hand, as a method for expressing the DNA of the present invention in an animal body, the DNA of the present invention is incorporated into an appropriate vector, for example, by a retrovirus method, a ribosome method, a cationic ribosome method, an adenovirus method, or the like. There is a method for introduction into a living body. This makes it possible to carry out gene therapy for a disease caused by a mutation in the # 10X gene of the present invention. Examples of the vector used include, but are not limited to, an adenovirus vector (eg, pAdexlcw) and a retrovirus vector (eg, pZIPneo). General genetic manipulations such as insertion of the DNA of the present invention into a vector can be performed according to a conventional method (Molecular Cloning, 5.61-5.63). Administration into a living body may be an ex vivo method or an in vivo method.
また、 本発明は、 本発明のベクターが導入された宿主細胞を提供する。 本発明 のベクターが導入される宿主細胞としては特に制限はなく、 例えば、 大腸菌や種 々の動物細胞などを用いることが可能である。 本発明の宿主細胞は、 例えば、 本 発明の蛋白質の製造や発現のための産生系として使用することができる。 蛋白質 製造のための産生系は、 in vitroおよび in vivoの産生系がある。 in vitroの 産生系としては、 真核細胞を使用する産生系や原核細胞を使用する産生系が挙げ られる。 真核細胞を使用する場合、 例えば、 動物細胞、 植物細胞、 真菌細胞を宿主に用 いることができる。 動物細胞としては、 哺乳類細胞、 例えば、 CHO (J . Exp. Med.The present invention also provides a host cell into which the vector of the present invention has been introduced. The host cell into which the vector of the present invention is introduced is not particularly limited, and for example, Escherichia coli and various animal cells can be used. The host cell of the present invention can be used, for example, as a production system for producing or expressing the protein of the present invention. Production systems for protein production include in vitro and in vivo production systems. Examples of in vitro production systems include production systems using eukaryotic cells and production systems using prokaryotic cells. When eukaryotic cells are used, for example, animal cells, plant cells, and fungal cells can be used as hosts. As animal cells, mammalian cells, for example, CHO (J. Exp. Med.
( 1995 ) 108, 945) 、 COSヽ 3T3、 ミエローマ、 BHK (baby hamster kidney) 、 H eLa、 Vero、 両生類細胞、 例えばアフリカヅメガエル卵母細胞 (Valle, et al .,(1995) 108, 945), COS-3T3, myeloma, BHK (baby hamster kidney), HeLa, Vero, amphibian cells such as African Megafrog oocytes (Valle, et al.,
Nature ( 1981 ) 291, 358-340) 、 あるいは昆虫細胞、 例えば、 Sf9、 Sf21、 Tn5 が知られている。 CH0細胞としては、 特に、 DHFR遺伝子を欠損した CH0細胞で ある dhfr- CHO (Proc .Natl .Acad. Sci .USA ( 1980 ) 77, 4216-4220) や CHO K-1 ( Proc .Natl .Acad. Sci .USA ( 1968) 60, 1275) を好適に使用することができる。 動物細胞において、 大量発現を目的とする場合には特に CH0細胞が好ましい。 宿主細胞へのベクタ一の導入は、 例えば、 リン酸カルシウム法、 DEAEデキスト ラン法、 カチォニックリボソーム D0TAP (ベ一リンガーマンハイム社製) を用い た方法、 エレクト口ポーレーシヨン法、 リポフエクシヨンなどの方法で行うこと が可能である。 Nature (1981) 291, 358-340) or insect cells such as Sf9, Sf21, and Tn5 are known. As the CH0 cells, in particular, dhfr-CHO (Proc. Natl. Acad. Scad. USA (1980) 77, 4216-4220) and CHO K-1 (Proc. Natl. Acad. Sci. USA (1968) 60, 1275) can be suitably used. In animal cells, when large-scale expression is intended, CH0 cells are particularly preferred. Vector vectors can be introduced into host cells by, for example, the calcium phosphate method, the DEAE dextran method, the method using Cationic Ribosome D0TAP (manufactured by Behringer Mannheim), the electoral poration method, or the Lipofexion method. Is possible.
植物細胞としては、 例えば、 ニコチアナ '夕バカム (Nicotiana tabacum) 由 来の細胞が蛋白質生産系として知られており、 これをカルス培養すればよい。 真 菌細胞としては、 酵母、 例えば、 サヅカロミセス (Saccharomyces) 属、 例えば 、 サヅカロミセス -セレビシェ (Saccharomyces cerevisiae) 、 糸状菌、 例え ば、 ァスペルギルス (Aspergillus) 属、 例えば、 ァスペルギルス ·ニガ一 (As pergillus niger) が知られている。  As plant cells, for example, cells derived from Nicotiana tabacum (Nicotiana tabacum) are known as protein production systems, which may be callus cultured. Fungal cells include yeast, for example, the genus Saccharomyces, for example, Saccharomyces cerevisiae, filamentous fungi, for example, the genus Aspergillus, for example, Aspergillus niger s. Are known.
原核細胞を使用する場合、 細菌細胞を用いる産生系がある。 細菌細胞としては 、 大腸菌 (E. coli) 、 例えば、 JM109、 DH5ひ、 HB101等が挙げられ、 その他、 枯草菌が知られている。  When prokaryotic cells are used, there are production systems using bacterial cells. Examples of bacterial cells include Escherichia coli (E. coli) such as JM109, DH5 and HB101, and Bacillus subtilis.
これらの細胞を目的とする DNAにより形質転換し、 形質転換された細胞を in vitroで培養することにより蛋白質が得られる。 培養は、 公知の方法に従い行う ことができる。 例えば、 動物細胞の培養液として、 例えば、 DMEM、 MEM, RPMI16 40、 IMDMを使用することができる。 その際、 牛胎児血清 (FCS) 等の血清補液を 併用することもできるし、 無血清培養してもよい。 培養時の pHは、 約 6〜8で あるのが好ましい。 培養は、 通常、 約 30〜40°Cで約 15〜200時間行い、 必要に 応じて培地の交換、 通気、 攪拌を加える。 The protein is obtained by transforming these cells with the target DNA and culturing the transformed cells in vitro. The culturing can be performed according to a known method. For example, as a culture solution of animal cells, for example, DMEM, MEM, RPMI1640, and IMDM can be used. At this time, serum replacement fluid such as fetal calf serum (FCS) They may be used in combination, or may be serum-free culture. The pH during culturing is preferably about 6-8. Culture is usually performed at about 30 to 40 ° C for about 15 to 200 hours, and the medium is replaced, aerated, and agitated as necessary.
—方、 in vivoで蛋白質を産生させる系としては、 例えば、 動物.を使用する産 生系や植物を使用する産生系が挙げられる。 これらの動物又は植物に目的とする DNAを導入し、 動物又は植物の体内で蛋白質を産生させ、 回収する。 本発明にお ける 「宿主」 とは、 これらの動物、 植物を包含する。  On the other hand, examples of a system for producing a protein in vivo include a production system using animals and a production system using plants. The target DNA is introduced into these animals or plants, and proteins are produced and recovered in the animals or plants. The “host” in the present invention includes these animals and plants.
動物を使用する場合、 哺乳類動物、 昆虫を用いる産生系がある。 哺乳類動物と しては、 ャギ、 ブ夕、 ヒヅジ、 マウス、 ゥシを用いることができる (Vicki Gla ser, SPECTRUM Biotechnology Applications, 1993) 。 また、 哺乳類動物を用 いる場合、 トランスジエニック動物を用いることができる。  When using animals, there are production systems using mammals and insects. As mammals, goats, bushes, ovines, mice, and mice can be used (Vicki Glaser, SPECTRUM Biotechnology Applications, 1993). When a mammal is used, a transgenic animal can be used.
例えば、 目的とする DNAを、 ャギ ?カゼインのような乳汁中に固有に産生さ れる蛋白質をコードする遺伝子との融合遺伝子として調製する。 次いで、 この融 合遺伝子を含む DNA断片をャギの胚へ注入し、 この胚を雌のャギへ移植する。 胚を受容したャギから生まれるトランスジエニックャギ又はその子孫が産生する 乳汁から、 目的の蛋白質を得ることができる。 トランスジヱニックャギから産生 される蛋白質を含む乳汁量を増加させるために、 適宜ホルモンをトランスジェニ ヅクャギに使用してもよい (EbertJ.M. et al ., Bio/Technology (1994) 12, 699-702) 。  For example, the target DNA is prepared as a fusion gene with a gene encoding a protein that is specifically produced in milk, such as goat casein. Next, the DNA fragment containing the fusion gene is injected into a goat embryo, and the embryo is transplanted into a female goat. The target protein can be obtained from milk produced by the transgenic goat born from the goat that has received the embryo or its progeny. Hormones may be used in transgenic goats as appropriate to increase the amount of milk containing proteins produced by transgenic goats (Ebert J. M. et al., Bio / Technology (1994) 12, 699-702).
また、 昆虫としては、 例えばカイコを用いることができる。 カイコを用いる場 合、 目的の蛋白質をコ一ドする DNAを挿入したバキュロウィルスをカイコに感 染させることにより、 このカイコの体液から目的の蛋白質を得ることができる ( Sus画 ,M. et al., Nature ( 1985) 315, 592-594) 。  In addition, silkworms can be used as insects, for example. When a silkworm is used, the target protein can be obtained from the body fluid of the silkworm by infecting the silkworm with a baculovirus into which DNA encoding the target protein has been inserted (Sus picture, M. et al. ., Nature (1985) 315, 592-594).
さらに、 植物を使用する場合、 例えばタバコを用いることができる。 タバコを 用いる場合、 目的とする蛋白質をコードする DNAを植物発現用ベクター、 例え ば pMON 530に挿入し、 このベクターをァグロパクテリゥム 'ッメファシエンス (Agrobacterium tumefaciens) のようなバクテリアに導入する。 このパクテリ ァをタバコ、 例えば、 ニコチアナ · タパカム (Nicotiana tabacum) に感染させ 、 本タバコの葉より所望のポリペプチドを得ることができる (Julian K. -C. Ma et al ., Eur. J. Immunol . ( 1994) 24, 131-138) 。 Furthermore, when using a plant, for example, tobacco can be used. When tobacco is used, the DNA encoding the target protein is inserted into a plant expression vector, for example, pMON530, and this vector is inserted into Agrobacterium 'mmfemaciens. (Agrobacterium tumefaciens). The bacterium is infected with tobacco, for example, Nicotiana tabacum, and the desired polypeptide can be obtained from the leaves of this tobacco (Julian K. -C. Ma et al., Eur. J. Immunol. (1994) 24, 131-138).
これにより得られた本発明の蛋白質は、 宿主細胞内または細胞外 (培地など) から単離し、 実質的に純粋で均一な蛋白質として精製することができる。 蛋白質 の分離、 精製は、 通常の蛋白質の精製で使用されている分離、 精製方法を使用す ればよく、 何ら限定されるものではない。 例えば、 クロマトグラフィーカラム、 フィルター、 限外濾過、 塩析、 溶媒沈殿、 溶媒抽出、 蒸留、 免疫沈降、 SDS-ポ リアクリルアミ ドゲル電気泳動、 等電点電気泳動法、 透析、 再結晶等を適宜選択 、 組み合わせれば蛋白質を分離、 精製することができる。  The protein of the present invention thus obtained can be isolated from inside or outside the host cell (such as a medium) and purified as a substantially pure and homogeneous protein. The separation and purification of the protein may be carried out by using the separation and purification methods used in ordinary protein purification, and is not limited at all. For example, chromatography columns, filters, ultrafiltration, salting out, solvent precipitation, solvent extraction, distillation, immunoprecipitation, SDS-polyacrylamide gel electrophoresis, isoelectric focusing, dialysis, recrystallization, etc. When combined, proteins can be separated and purified.
クロマトグラフィーとしては、 例えばァフィ二ティークロマトグラフィー、 ィ オン交換ク口マトグラフィ一、 疎水性ク口マトグラフィ一、 ゲル濾過、 逆相ク口 マトグラフィ一、 吸着クロマトグラフィー等が挙げられる (Strategies for Pr otein Purification and Cnaracterization: A Laboratory Course Manual . E d Daniel R. Marshak et al . , Cold Spring Harbor Laboratory Press, 1996 ) 。 これらのクロマトグラフィーは、 液相クロマトグラフィー、 例えば HPLC、 F PLC等の液相クロマトグラフィーを用いて行うことができる。 本発明は、 これら の精製方法を用い、 高度に精製された蛋白質も包含する。  Examples of the chromatography include affinity chromatography, ion-exchange chromatography, hydrophobic chromatography, gel filtration, reversed-phase chromatography, and adsorption chromatography (Strategies for Protein Purification). and Cnaracterization: A Laboratory Course Manual. Ed Daniel R. Marshak et al., Cold Spring Harbor Laboratory Press, 1996). These chromatographys can be performed using liquid phase chromatography, for example, liquid phase chromatography such as HPLC and FPLC. The present invention also includes highly purified proteins using these purification methods.
なお、 蛋白質を精製前又は精製後に適当な蛋白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部分的にペプチドを除去することもできる。 蛋白質 修飾酵素としては、 例えば、 トリプシン、 キモトリプシン、 リシルェンドぺプチ ダーゼ、 プロテインキナーゼ、 グルコシダ一ゼなどが用いられる。  The protein can be arbitrarily modified or partially removed by reacting the protein with an appropriate protein modifying enzyme before or after purification. As the protein modifying enzyme, for example, trypsin, chymotrypsin, lysylendopidase, protein kinase, glucosidase and the like are used.
本発明は、 また、 本発明の蛋白質と結合する抗体を提供する。 本発明の抗体の 形態には、 特に制限はなく、 ポリクローナル抗体の他、 モノクローナル抗体も含 まれる。 また、 ゥサギなどの免疫動物に本発明の蛋白質を免疫して得た抗血清、 すべてのクラスのポリクロ一ナル抗体およびモノクローナル抗体、 さらにヒト抗 体や遺伝子組み換えによるヒト型化抗体も含まれる。 The present invention also provides an antibody that binds to the protein of the present invention. The form of the antibody of the present invention is not particularly limited, and includes a monoclonal antibody as well as a polyclonal antibody. Further, antiserum obtained by immunizing an immunized animal such as a heron with the protein of the present invention; This includes all classes of polyclonal and monoclonal antibodies, as well as human and recombinant humanized antibodies.
抗体取得の感作抗原として使用される本発明の蛋白質は、 その由来となる動物 種に制限されないが哺乳動物、 例えばヒト、 マウス又はラット由来の蛋白質が好 ましく、 特にヒト由来の蛋白質が好ましい。 ヒト由来の蛋白質は、 本明細書に開 示される遺伝子配列又はアミノ酸配列を用いて得ることができる。  The protein of the present invention used as a sensitizing antigen for obtaining an antibody is not limited to the animal species from which it is derived, but is preferably a protein derived from a mammal, for example, a human, a mouse or a rat, and particularly preferably a protein derived from a human. . A human-derived protein can be obtained using the gene sequence or amino acid sequence disclosed herein.
本発明において、 感作抗原として使用される蛋白質は、 完全な蛋白質であって もよいし、 また、 蛋白質の部分ペプチドであってもよい。 蛋白質の部分ペプチド としては、 例えば、 蛋白質のアミノ基 (N) 末端断片やカルボキシ (C) 末端断 片が挙げられる。 本明細書で述べる 「抗体」 とは蛋白質の全長又は断片に反応す る抗体を意味する。  In the present invention, the protein used as the sensitizing antigen may be a complete protein or a partial peptide of the protein. Examples of the partial peptide of the protein include an amino group (N) terminal fragment and a carboxy (C) terminal fragment of the protein. As used herein, “antibody” refers to an antibody that reacts with the full length or fragment of a protein.
本発明の蛋白質又はその断片をコードする遺伝子を公知の発現べクタ一系に挿 入し、 該ベクターで本明細書で述べた宿主細胞を形質転換させ、 該宿主細胞内外 から目的の蛋白質又はその断片を公知の方法で得て、 これらを感作抗原として用 いればよい。 また、 蛋白質を発現する細胞又はその溶解物あるいは化学的に合成 した本発明の蛋白質を感作抗原として使用してもよい。 短いペプチドは、 キ一ホ —ルリンペッ トへモシァニン、 ゥシ血清アルブミン、 卵白アルブミンなどのキ リア蛋白質と適宜結合させて抗原とすることが好ましい。  A gene encoding the protein of the present invention or a fragment thereof is inserted into a known expression vector system, and the vector is used to transform the host cell described in the present specification. Fragments may be obtained by a known method, and these may be used as a sensitizing antigen. Further, a cell expressing the protein, a lysate thereof, or a chemically synthesized protein of the present invention may be used as the sensitizing antigen. It is preferable that the short peptide is appropriately bound to a carrier protein such as keyhole lysine hepatocyanin, pepsin albumin, ovalbumin, etc. to form an antigen.
感作抗原で免疫ざれる哺乳動物としては、 特に限定されるものではないが、 細 胞融合に使用する親細胞との適合性を考慮して選択するのが好ましく、 一般的に は、 げっ歯目、 ゥサギ目、 霊長目の動物が使用される。  The mammal to be immunized with the sensitizing antigen is not particularly limited, but is preferably selected in consideration of compatibility with the parent cell used for cell fusion. In general, rodents are used. Eyes, egrets, and primates are used.
げっ歯目の動物としては、 例えば、 マウス、 ラット、 ハムスター等が使用され る。 ゥサギ目の動物としては、 例えば、 ゥサギが使用される。 霊長目の動物とし ては、 例えば、 サルが使用される。 サルとしては、 狭鼻下目のサル (旧世界ザル ) 、 例えば、 力二クイザル、 ァカゲザル、 マントヒヒ、 チンパンジー等が使用さ れる。 感作抗原を動物に免疫するには、 公知の方法にしたがって行われる。 一般的方 法としては、 感作抗原を哺乳動物の腹腔内又は皮下に注射する。 具体的には、 感 作抗原を PBS( Phosphate- Buffered Saline)や生理食塩水等で適当量に希釈、 懸 濁したものに対し、 所望により通常のアジュバント、 例えば、 フロイント完全ァ ジュバントを適量混合し、 乳化後、 哺乳動物に投与する。 さらに、 その後、 フロ ィント不完全アジュバントに適量混合した感作抗原を、 4〜21日毎に数回投与す ることが好ましい。 また、 感作抗原免疫時に適当な担体を使用することができる 。 このように免疫し、 血清中に所望の抗体レベルが上昇するのを常法により確認 する。 As rodent animals, for example, mice, rats, hamsters and the like are used.動物 As an heronoid animal, for example, a heron is used. For example, monkeys are used as primates. As the monkeys, monkeys of the lower nose (old world monkeys), for example, cynomolgus monkeys, macaques, baboons, and chimpanzees are used. Immunization of an animal with a sensitizing antigen is performed according to a known method. As a general method, a sensitizing antigen is injected intraperitoneally or subcutaneously into a mammal. Specifically, the sensitizing antigen is diluted and suspended in an appropriate amount with PBS (Phosphate-Buffered Saline) or physiological saline, and then mixed with an appropriate amount of a normal adjuvant, for example, Freund's complete adjuvant, if desired. After emulsification, it is administered to mammals. Thereafter, it is preferable to administer the sensitizing antigen mixed with an appropriate amount of incomplete Freund's adjuvant several times every 4 to 21 days. In addition, a suitable carrier can be used at the time of immunization with the sensitizing antigen. Immunization is performed in this manner, and an increase in the desired antibody level in the serum is confirmed by a conventional method.
ここで、 本発明の蛋白質に対するポリクローナル抗体を得るには、 血清中の所 望の抗体レペルが上昇したことを確認した後、 抗原を感作した哺乳動物の血液を 取り出す。 この血液から公知の方法により血清を分離する。 ポリクロ一ナル抗体 としては、 ポリクロ一ナル抗体を含む血清を使用してもよいし、 必要に応じこの 血清からポリクロ一ナル抗体を含む画分をさらに単離して、 これを使用してもよ い。 例えば、 本発明の蛋白質をカップリングさせたァフィ二ティ一カラムを用い て、 本発明の蛋白質のみを認識する画分を得て、 さらにこの画分をプロテイン A あるいはプロティン Gカラムを利用して精製することにより、 免疫グロプリン G あるいは Mを調製することができる。  Here, in order to obtain a polyclonal antibody against the protein of the present invention, the blood of a mammal sensitized with the antigen is taken out after confirming that the desired antibody repel in serum has been increased. The serum is separated from the blood by a known method. As the polyclonal antibody, a serum containing the polyclonal antibody may be used.If necessary, a fraction containing the polyclonal antibody may be further isolated from this serum and used. . For example, using an affinity column to which the protein of the present invention is coupled, a fraction that recognizes only the protein of the present invention is obtained, and this fraction is further purified using a protein A or protein G column. Thus, immunoglobulin G or M can be prepared.
モノクローナル抗体を得るには、 上記抗原を感作した哺乳動物の血清中に所望 の抗体レベルが上昇するのを確認した後に、 哺乳動物から免疫細胞を取り出し、 細胞融合に付せばよい。 この際、 細胞融合に使用される好ましい免疫細胞として 、 特に脾細胞が挙げられる。 前記免疫細胞と融合される他方の親細胞としては、 好ましくは哺乳動物のミエローマ細胞、 より好ましくは、 薬剤による融合細胞選 別のための特性を獲得したミエローマ細胞が挙げられる。 前記免疫細胞とミエローマ細胞の細胞融合は基本的には公知の方法、 例えば、 ミルスティンらの方法(Galfre, G. and Milstein,C , Methods Enzymol . ( 1981 ) 73, 3- 46)等に準じて行うことができる。 To obtain a monoclonal antibody, after confirming that the desired antibody level is increased in the serum of the mammal sensitized with the antigen, the immune cells may be removed from the mammal and subjected to cell fusion. At this time, preferred immune cells used for cell fusion include splenocytes in particular. The other parent cell to be fused with the immunocyte is preferably a mammalian myeloma cell, more preferably a myeloma cell that has acquired the properties for fusion cell selection by a drug. Cell fusion between the immune cells and myeloma cells is basically performed according to a known method, for example, the method of Milstein et al. (Galfre, G. and Milstein, C, Methods Enzymol. (1981) 73, 3-46). It can be carried out.
細胞融合により得られたハイプリ ドーマは、 通常の選択培養液、 例えば、 HAT 培養液 (ヒポキサンチン、 アミノプテリンおよびチミジンを含む培養液) で培養 することにより選択される。 当該 HAT培養液での培養は、 目的とするハイプリ ドーマ以外の細胞 (非融合細胞) が死滅するのに十分な時間、 通常、 数日〜数週 間継続して行う。 次いで、 通常の限界希釈法を実施し、 目的とする抗体を産生す るハイブリ ドーマのスクリ一ニングおよびクローニングを行う。  The hybridoma obtained by cell fusion is selected by culturing it in a normal selective culture medium, for example, a HAT culture medium (a culture medium containing hypoxanthine, aminopterin and thymidine). Culturing in the HAT culture solution is continued for a period of time sufficient to kill cells other than the target hybridoma (non-fused cells), usually for several days to several weeks. Next, screening and cloning of hybridomas producing the desired antibody are performed by the usual limiting dilution method.
また、 ヒト以外の動物に抗原を免疫して上記ハイプリ ドーマを得る他に、 ヒト リンパ球、 例えば EBウィルスに感染したヒトリンパ球を in vitroで蛋白質、 蛋白質発現細胞又はその溶解物で感作し、 感作リンパ球をヒト由来の永久分裂能 を有するミエローマ細胞、 例えば U266と融合させ、 蛋白質への結合活性を有す る所望のヒト抗体を産生するハイプリ ドーマを得ることもできる (特開昭 63-17 688号公報) 。  In addition, in addition to obtaining the above-mentioned hybridoma by immunizing non-human animals with an antigen, human lymphocytes, for example, human lymphocytes infected with EB virus are sensitized in vitro with proteins, protein-expressing cells or lysates thereof, It is also possible to fuse a sensitized lymphocyte with a human-derived myeloma cell having permanent division ability, for example, U266, to obtain a hybridoma that produces a desired human antibody having a protein binding activity (Japanese Patent Application Laid-Open No. -17 688 publication).
次いで、 得られたハイプリ ドーマをマウス腹腔内に移植し、 同マウスより腹水 を回収し、 得られたモノクローナル抗体を、 例えば、 硫安沈殿、 プロテイン A、 プロテイン Gカラム、 DEAEイオン交換クロマトグラフィー、 本発明の蛋白質を カップリングしたァフィ二ティーカラムなどにより精製することで調製すること が可能である。 本発明の抗体は、 本発明の蛋白質の精製、 検出に用いられる他、 本発明の蛋白質のァゴニストやアン夕ゴニストの候補になる。 また、 この抗体を 本発明の蛋白質が関与する疾患の抗体治療へ応用することも考えられる。 得られ た抗体を人体に投与する目的 (抗体治療) で使用する場合には、 免疫原性を低下 させるため、 ヒト抗体やヒト型抗体が好ましい。  Next, the obtained hybridoma is transplanted into the peritoneal cavity of a mouse, ascites is collected from the mouse, and the obtained monoclonal antibody is subjected to, for example, ammonium sulfate precipitation, protein A, protein G column, DEAE ion exchange chromatography, and the present invention. It can be prepared by purifying the above protein using a coupled affinity column or the like. The antibody of the present invention is used for purification and detection of the protein of the present invention, and is also a candidate for an agonist and an angoniist of the protein of the present invention. It is also conceivable to apply this antibody to antibody therapy for diseases involving the protein of the present invention. When the obtained antibody is used for administration to the human body (antibody therapy), a human antibody or a humanized antibody is preferable in order to reduce immunogenicity.
例えば、 ヒト抗体遺伝子のレパートリーを有するトランスジエニック動物に抗 原となる蛋白質、 蛋白質発現細胞又はその溶解物を免疫して抗体産生細胞を取得 し、 これをミエローマ細胞と融合させたハイプリ ドーマを用いて蛋白質に対する ヒト抗体を取得することができる (国際公開番号 W092- 03918、 W093-2227, W094 - 02602、 W094- 25585、 W096- 33735および W096- 34096参照) 。 For example, a transgenic animal having a human antibody gene repertoire is immunized with a protein serving as an antigen, protein-expressing cells or a lysate thereof to obtain antibody-producing cells. A human antibody against the protein can be obtained using a hybridoma fused with myeloma cells (International Publication Nos. W092--03918, W093-2227, W094-02602, W094-25585, W096-33735 and W096). -See 34096).
ハイプリ ドーマを用いて抗体を産生する以外に、 抗体を産生する感作リンパ球 等の免疫細胞を癌遺伝子 (oncogene)により不死化させた細胞を用いてもよい。 このように得られたモノクローナル抗体はまた、 遺伝子組換え技術を用いて産 生させた組換え型抗体として得ることができる (例えば、 Borrebaeck, A.K. a nd Larrick,J.W., THERAPEUTIC MONOCLONAL ANTIBODIES, Published in the U nited Kingdom by MCMILLAN PUBLISHERS LTD, 1990参照)。 組換え型抗体は、 それをコ一ドする DNAをハイプリ ド一マ又は抗体を産生する感作リンパ球等の 免疫細胞からクローニングし、 適当なベクターに組み込んで、 これを宿主に導入 し産生させる。 本発明は、 この組換え型抗体を包含する。  In addition to producing antibodies using hybridomas, cells in which immune cells such as sensitized lymphocytes that produce antibodies are immortalized by oncogenes may be used. The thus obtained monoclonal antibody can also be obtained as a recombinant antibody produced using a genetic recombination technique (for example, Borrebaeck, AK and Larrick, JW, THERAPEUTIC MONOCLONAL ANTIBODIES, Published in the United Kingdom by MCMILLAN PUBLISHERS LTD, 1990). Recombinant antibodies are produced by cloning the DNA encoding them from hybridomas or immunized cells such as sensitized lymphocytes that produce the antibody, inserting the DNA into an appropriate vector, and introducing it into a host. . The present invention includes this recombinant antibody.
さらに、 本発明の抗体は、 本発明の蛋白質に結合する限り、 その抗体断片ゃ抗 体修飾物であってよい。 例えば、 抗体断片としては、 Fab、 F(ab')2、 Fv又は H 鎖と L鎖の Fvを適当なリンカーで連結させたシングルチエイン Fv(scFv)(Husto n,J.S. et al., Proc. Natl. Acad. Sci. U.S.A. (1988) 85, 5879-5883)が挙げ られる。 具体的には、 抗体を酵素、 例えば、 パパイン、 ペプシンで処理し抗体断 片を生成させるか、 又は、 これら抗体断片をコードする遺伝子を構築し、 これを 発現ベクターに導入した後、 適当な宿主細胞で発現させる (例えば、 Co,M.S. e t al., J. Immunol. (1994) 152, 2968-2976; Better5M. and Horwitz,A.H. , M ethods Enzymol. (1989) 178, 476-496; Pluckthun5A. and Skerra,A., Metho ds Enzymol. (1989) 178, 497-515; Lamoyi,E.5 Methods Enzymol. (1986) 12 1, 652-663; Rousseaux,J. et al., Methods Enzymol. (1986) 121, 663-669; Bird,R.E. and Walker, B.W., Trends Biotechnol. (1991) 9, 132- 137参照) 抗体修飾物として、 ポリエチレングリコール (PEG) 等の各種分子と結合した 抗体を使用することもできる。 本発明の 「抗体」 にはこれらの抗体修飾物も包含 される。 このような抗体修飾物を得るには、 得られた抗体に化学的な修飾を施す ことによって得ることができる。 これらの方法はこの分野において既に確立され ている。 Further, the antibody of the present invention may be a modified antibody fragment thereof as long as it binds to the protein of the present invention. For example, as an antibody fragment, Fab, F (ab ') 2, Fv, or a single chain Fv (scFv) in which an Fv of an H chain and an L chain are linked by an appropriate linker (Huston, JS et al., Proc. Natl. Acad. Sci. USA (1988) 85, 5879-5883). Specifically, the antibody is treated with an enzyme, for example, papain or pepsin, to generate an antibody fragment, or a gene encoding these antibody fragments is constructed and introduced into an expression vector. Expressed in cells (for example, Co, MS et al., J. Immunol. (1994) 152, 2968-2976; Better 5 M. and Horwitz, AH, Methods Enzymol. (1989) 178, 476-496; Pluckthun 5 A. and Skerra, A., Methods Enzymol. (1989) 178, 497-515; Lamoyi, E. 5 Methods Enzymol. (1986) 121, 652-663; Rousseaux, J. et al., Methods Enzymol. (1986) 121, 663-669; Bird, RE and Walker, BW, Trends Biotechnol. (1991) 9, 132-137.) As the modified antibody, an antibody bound to various molecules such as polyethylene glycol (PEG) can be used. The “antibody” of the present invention also includes these modified antibodies. Such a modified antibody can be obtained by subjecting the obtained antibody to chemical modification. These methods are already established in this field.
また、 本発明の抗体は、 公知の技術を使用して非ヒト抗体由来の可変領域とヒ ト抗体由来の定常領域からなるキメラ抗体又は非ヒト抗体由来の CDR (相補性決 定領域) とヒト抗体由来の FR (フレームワーク領域) 及び定常領域からなるヒ ト型化抗体として得ることができる。  In addition, the antibody of the present invention can be prepared by using a chimeric antibody comprising a non-human antibody-derived variable region and a human antibody-derived constant region or a non-human antibody-derived CDR (complementarity determining region) and a human It can be obtained as a humanized antibody consisting of antibody-derived FR (framework region) and constant region.
前記のように得られた抗体は、 均一にまで精製することができる。 本発明で使 用される抗体の分離、 精製は通常の蛋白質で使用されている分離、 精製方法を使 用すればよい。 例えば、 ァフィ二ティ一クロマトグラフィー等のクロマトグラフ ィ一カラム、 フィル夕一、 限外濾過、 塩析、 透析、 SDSポリアクリルアミ ドゲル 電気泳動、 等電点電気泳動等を適宜選択、 組み合わせれば、 抗体を分離、 精製す ることができる (Antibodies : A Laboratory Manual . Ed Harlow and David Lan e, Cold Spring Harbor Laboratory, 1988)が、 これらに限定されるものではな い。 上記で得られた抗体の濃度測定は吸光度の測定又は酵素結合免疫吸着検定法 (Enzyme- linked immunosorbent assay; ELISA)等により行うこと力できる。 ァフィ二ティ一クロマトグラフィ一に用いるカラムとしては、 プロティン A力 ラム、 プロテイン Gカラムが挙げられる。 例えば、 プロテイン Aカラムを用いた カラムとして、 Hyper D, POROS, Sepharose F .F . (Pharmacia)等が挙げられる。 ァフィ二ティ一クロマトグラフィ一以外のクロマトグラフィ一としては、 例え ば、 イオン交換クロマトグラフィー、 疎水性クロマトグラフィー、 ゲル濾過、 逆 相クロマトグラフィー、 吸着クロマトグラフィー等が挙げられる(Strategies f or Protein Purification and Characterization : A Laboratory Course Man ual. Ed Daniel R. Marshak et al . , Cold Spring Harbor Laboratory Press, 1996)。 これらのクロマトグラフィーは HPI 、 FPLC等の液相クロマトグラフィ 一を用いて行うことができる。 The antibody obtained as described above can be purified to homogeneity. The separation and purification of the antibody used in the present invention may be performed by the separation and purification methods used for ordinary proteins. For example, chromatography columns such as affinity chromatography, filtration, ultrafiltration, salting out, dialysis, SDS polyacrylamide gel electrophoresis, isoelectric focusing, etc. can be appropriately selected and combined. Antibodies can be separated and purified (Antibodies: A Laboratory Manual; Ed Harlow and David Lane, Cold Spring Harbor Laboratory, 1988), but are not limited thereto. The concentration of the antibody obtained above can be measured by measuring the absorbance or by using an enzyme-linked immunosorbent assay (ELISA) or the like. Columns used for affinity chromatography include a protein A column and a protein G column. For example, columns using a protein A column include Hyper D, POROS, Sepharose F.F. (Pharmacia), and the like. Examples of chromatography other than affinity chromatography include, for example, ion exchange chromatography, hydrophobic chromatography, gel filtration, reverse phase chromatography, and adsorption chromatography (Strategies for Protein Purification and Characterization: A Laboratory Course Manual.Ed Daniel R. Marshak et al., Cold Spring Harbor Laboratory Press, 1996). These chromatographys can be performed using liquid phase chromatography such as HPI and FPLC.
また、 本発明の抗体の抗原結合活性を測定する方法として、 例えば、 吸光度の 測定、 酵素結合免疫吸着検定法(Enzyme- linked immunosorbent assay; EL ISA) 、 EIA (酵素免疫測定法) 、 RIA (放射免疫測定法) あるいは蛍光抗体法を用い ることができる。 ELISAを用いる場合、 本発明の抗体を固相化したプレートに本 発明の蛋白質を添加し、 次いで目的の抗体を含む試料、 例えば、 抗体産生細胞の 培養上清や精製抗体を加える。 酵素、 例えば、 アルカリフォスファターゼ等で標 識した抗体を認識する二次抗体を添加し、 プレートをインキュベーションし、 次 いで洗浄した後、 P-ニトロフヱニル燐酸などの酵素基質を加えて吸光度を測定 することで抗原結合活性を評価することができる。 蛋白質として蛋白質の断片、 例えばその C末端からなる断片を使用してもよい。 本発明の抗体の活性評価に は、 B I Acore ( Pharmacia製)を使用することができる。  Methods for measuring the antigen-binding activity of the antibody of the present invention include, for example, measurement of absorbance, enzyme-linked immunosorbent assay (ELISA), EIA (enzyme-linked immunosorbent assay), Immunoassay) or a fluorescent antibody method can be used. When ELISA is used, the protein of the present invention is added to a plate on which the antibody of the present invention is immobilized, and then a sample containing the target antibody, for example, a culture supernatant of antibody-producing cells or a purified antibody is added. Add a secondary antibody that recognizes an enzyme, for example, an antibody labeled with alkaline phosphatase, etc., incubate the plate, wash the plate, and then add an enzyme substrate such as P-nitrophenylphosphoric acid and measure the absorbance. Antigen binding activity can be evaluated. As the protein, a protein fragment, for example, a fragment comprising the C-terminus thereof may be used. For evaluation of the activity of the antibody of the present invention, BI Acore (Pharmacia) can be used.
これらの手法を用いることにより、 本発明の抗体と試料中に含まれる本発明の 蛋白質が含まれると予想される試料とを接触せしめ、 該抗体と該蛋白質との免疫 複合体を検出又は測定することからなる、 本発明の蛋白質の検出又は測定方法を 実施することができる。 本発明の蛋白質の検出又は測定方法は、 蛋白質を特異的 に検出又は測定することができるため、 蛋白質を用いた種々の実験等に有用であ る。  By using these techniques, the antibody of the present invention is brought into contact with the sample contained in the sample which is expected to contain the protein of the present invention, and an immune complex of the antibody and the protein is detected or measured. Thus, the method for detecting or measuring a protein of the present invention can be carried out. INDUSTRIAL APPLICABILITY The method for detecting or measuring a protein of the present invention can specifically detect or measure a protein, and thus is useful for various experiments and the like using proteins.
本発明はまた、 #10X蛋白質をコードする DNA (配列番号: 1から 7 ) または その相補鎖に相補的な少なくとも 15ヌクレオチドを含むポリヌクレオチドを提 供する。  The present invention also provides a polynucleotide comprising at least 15 nucleotides complementary to the DNA encoding the # 10X protein (SEQ ID NOS: 1 to 7) or its complementary strand.
ここで 「相補鎖」 とは、 A:T (ただし RNAの場合は U) 、 G:Cの塩基対からな る 2本鎖核酸の一方の鎖に対する他方の鎖を指す。 また、 「相補的」 とは、 少 なくとも 15個の連続したヌクレオチド領域で完全に相補配列である場合に限ら れず、 少なくとも 70°ん 好ましくは少なくとも 80%、 より好ましくは 90%、 さら に好ましくは 95%以上の塩基配列上の相同性を有すればよい。 相同性を決定する ためのァルゴリズムは本明細書に記載したものを使用すればよい。 Here, the “complementary strand” refers to one strand of a double-stranded nucleic acid consisting of A: T (U in the case of RNA) and G: C base pairs with respect to the other strand. The term “complementary” is not limited to a case where the sequence is completely complementary to at least 15 contiguous nucleotide regions, but is at least 70 °, preferably at least 80%, more preferably 90%, and more preferably It is preferable that they have a homology of 95% or more on the base sequence. The algorithm for determining homology may use the algorithm described in this specification.
このような核酸には、 本発明の蛋白質をコードする DNAの検出や増幅に用い るプローブやプライマ一、 該 DNAの発現を検出するためのプロ一ブゃプラィマ 一、 本発明の蛋白質の発現を制御するためのヌクレオチド又はヌクレオチド誘導 体 (例えば、 アンチセンスオリゴヌクレオチドやリボザィム、 またはこれらをコ ードする DNA等) が含まれる。 また、 このような核酸は、 DNAチップの作製に利 用することもできる。  Such nucleic acids include probes and primers used for detection and amplification of DNA encoding the protein of the present invention, probe primers for detecting the expression of the DNA, and expression of the protein of the present invention. Nucleotides or nucleotide derivatives (eg, antisense oligonucleotides, ribozymes, or DNA encoding them) for control are included. Such a nucleic acid can also be used for producing a DNA chip.
プライマーとして用いる場合、 3'側の領域は相補的とし、 5'側には制限酵素 認識配列やタグなどを付加することができる。  When used as a primer, the 3′-side region is complementary, and a restriction enzyme recognition sequence, a tag, or the like can be added to the 5′-side.
アンチセンスオリゴヌクレオチドとしては、 例えば、 配列番号: 1から 7の塩 基配列中のいずれかの箇所にハイブリダイズするアンチセンスォリゴヌクレオチ ドが含まれる。 このアンチセンスオリゴヌクレオチドは、 好ましくは配列番号: 1から 7の塩基配列中の連続する少なくとも 15個以上のヌクレオチドに対する アンチセンスオリゴヌクレオチドである。 さらに好ましくは、 連続する少なくと も 15個以上のヌクレオチドが翻訳開始コドンを含むアンチセンスオリゴヌクレ ォチドである。  Antisense oligonucleotides include, for example, antisense oligonucleotides that hybridize at any position in the base sequence of SEQ ID NOs: 1 to 7. This antisense oligonucleotide is preferably an antisense oligonucleotide for at least 15 or more consecutive nucleotides in the nucleotide sequence of SEQ ID NOs: 1 to 7. More preferably, it is an antisense oligonucleotide in which at least 15 or more consecutive nucleotides contain a translation initiation codon.
アンチセンスオリゴヌクレオチドとしては、 それらの誘導体や修飾体を使用す ることができる。 修飾体として、 例えばメチルホスホネート型又はェチルホスホ ネート型のような低級アルキルホスホネート修飾体、 ホスホロチォエート修飾体 又はホスホロアミデート修飾体等が挙げられる。  As the antisense oligonucleotide, derivatives and modifications thereof can be used. Examples of the modified product include a modified lower alkyl phosphonate such as a methylphosphonate type or an ethylphosphonate type, a phosphorothioate modified product, a phosphoroamidate modified product, and the like.
アンチセンスオリゴヌクレオチドは、 DNA又は mRNAの所定の領域を構成する ヌクレオチドに対応するヌクレオチドが全て相補配列であるもののみならず、 D NAまたは mRNAとォリゴヌクレオチドとが配列番号: 1から 7に示される塩基配 列に特異的にハイブリダイズできる限り、 1又は複数個のヌクレオチドのミスマ ツチが存在しているものも含まれる。 本発明のアンチセンスオリゴヌクレオチド誘導体は、 本発明の蛋白質の産生細 胞に作用して、 該蛋白質をコードする DNA又は mRNAに結合することにより、 そ の転写又は翻訳を阻害したり、 m Aの分解を促進したりして、 本発明の蛋白質 の発現を抑制することにより、 結果的に本発明の蛋白質の作用を抑制する効果を 有" 5 ·る。 Antisense oligonucleotides include not only those whose nucleotides corresponding to nucleotides constituting a predetermined region of DNA or mRNA are all complementary sequences, but also DNAs or mRNAs and oligo nucleotides shown in SEQ ID NOs: 1 to 7. As long as it can specifically hybridize to the base sequence to be used, one including one or more nucleotide mismatches is also included. The antisense oligonucleotide derivative of the present invention acts on cells producing the protein of the present invention to bind to DNA or mRNA encoding the protein, thereby inhibiting the transcription or translation of the protein or producing mA. By suppressing the expression of the protein of the present invention, for example, by promoting the degradation, the effect of suppressing the action of the protein of the present invention is obtained.
本発明のアンチセンスオリゴヌクレオチド誘導体は、 それらに対して不活性な 適当な基剤と混和して塗布剤、 パップ剤等の外用剤とすることができる。  The antisense oligonucleotide derivative of the present invention can be mixed with a suitable base material which is inactive against the derivative to prepare an external preparation such as a liniment or a poultice.
また、 必要に応じて、 賦形剤、 等張化剤、 溶解補助剤、 安定化剤、 防腐剤、 無 痛化剤等を加えて錠剤、 散財、 顆粒剤、 カプセル剤、 リボソームカプセル剤、 注 射剤、 液剤、 点鼻剤など、 さらに凍結乾燥剤とすることができる。 これらは常法 にしたがって調製することができる。  If necessary, excipients, isotonic agents, solubilizing agents, stabilizers, preservatives, soothing agents, etc. may be added to tablets, splinters, granules, capsules, ribosome capsules, It can be a lyophilized agent such as a propellant, a liquid, a nasal drop and the like. These can be prepared according to a conventional method.
本発明のアンチセンスオリゴヌクレオチド誘導体は患者の患部に直接適用する か、 又は血管内に投与するなどして結果的に患部に到達し得るように患者に適用 する。 さらには、 持続性、 膜透過性を高めるアンチセンス封入素材を用いること もできる。 例えば、 リポソ一ム、 ポリ- L-リジン、 リピヅ ド、 コレステロール、 リボフヱクチン又はこれらの誘導体が挙げられる。  The antisense oligonucleotide derivative of the present invention is applied directly to the affected area of the patient, or is applied to the patient so as to be able to reach the affected area as a result of intravenous administration or the like. Furthermore, an antisense-encapsulated material that enhances durability and membrane permeability can be used. Examples include liposomes, poly-L-lysine, lipids, cholesterol, ribofectin or derivatives thereof.
本発明のアンチセンスオリゴヌクレオチド誘導体の投与量は、 患者の状態に応 じて適宜調整し、 好ましい量を用いることができる。 例えば、 0. 1〜100mg/kg、 好ましくは 0. l〜50mg/kgの範囲で投与することができる。  The dosage of the antisense oligonucleotide derivative of the present invention can be appropriately adjusted according to the condition of the patient, and a preferred amount can be used. For example, it can be administered in the range of 0.1 to 100 mg / kg, preferably 0.1 to 50 mg / kg.
本発明のアンチセンスオリゴヌクレオチドは本発明の蛋白質の発現を阻害し、 従って本発明の蛋白質の生物学的活性を抑制することにおいて有用である。 また 、 本発明のアンチセンスオリゴヌクレオチドを含有する発現阻害剤は、 本発明の 蛋白質の生物学的活性を抑制することが可能である点で有用である。  The antisense oligonucleotide of the present invention inhibits the expression of the protein of the present invention and is therefore useful in suppressing the biological activity of the protein of the present invention. Further, the expression inhibitor containing the antisense oligonucleotide of the present invention is useful in that it can suppress the biological activity of the protein of the present invention.
本発明の蛋白質は、 これに結合する化合物のスクリーニングに有用である。 す なわち、 本発明の蛋白質と、 該蛋白質に結合する化合物を含むと予想される被検 試料とを接触せしめ、 そして本発明の蛋白質に結合する活性を有する化合物を選 択する、 ことからなる本発明の蛋白質に結合する化合物をスクリーニングする方 法において使用される。 The protein of the present invention is useful for screening for a compound that binds to the protein. That is, the protein of the present invention is brought into contact with a test sample expected to contain a compound binding to the protein, and a compound having an activity of binding to the protein of the present invention is selected. And a method of screening for a compound that binds to the protein of the present invention.
スクリーニングに用いられる本発明の蛋白質は組換え蛋白質であっても、 天然 由来の蛋白質であってもよい。 また部分ペプチドであってもよい。 また細胞表面 に発現させた形態、 または膜画分としての形態であってもよい。 被検試料として は特に制限はなく、 例えば、 細胞抽出物、 細胞培養上清、 発酵微生物産生物、 海 洋生物抽出物、 植物抽出物、 精製若しくは粗精製蛋白質、 ペプチド、 非ペプチド 性化合物、 合成低分子化合物、 天然化合物が挙げられる。 被検試料を接触させる 本発明の蛋白質は、 例えば、 精製した蛋白質として、 可溶型蛋白質として、 担体 に結合させた形態として、 他の蛋白質との融合蛋白質として、 細胞膜上に発現さ せた形態として、 膜画分として被検試料に接触させることができる。  The protein of the present invention used for the screening may be a recombinant protein or a protein of natural origin. It may be a partial peptide. It may also be a form expressed on the cell surface or a form as a membrane fraction. The test sample is not particularly limited and includes, for example, a cell extract, a cell culture supernatant, a fermentation microorganism product, a marine organism extract, a plant extract, a purified or crude protein, a peptide, a non-peptidic compound, and a synthesis. Low molecular weight compounds and natural compounds. The protein of the present invention, which is brought into contact with a test sample, may be, for example, a purified protein, a soluble protein, a form bound to a carrier, a fusion protein with another protein, or a form expressed on a cell membrane. The sample can be brought into contact with a test sample as a membrane fraction.
本発明の蛋白質を用いて、 例えば該蛋白質に結合する蛋白質をスクリーニング する方法としては、 当業者に公知の多くの方法を用いることが可能である。 この ようなスクリーニングは、 例えば、 免疫沈降法により行うことができる。 具体的 には、 以下のように行うことができる。 本発明の蛋白質をコードする遺伝子を、 pSV2neo, pcDNA I, pCD8 などの外来遺伝子発現用のベクターに揷入することで 動物細胞などで当該遺伝子を発現させる。 発現に用いるプロモータ一としては S V40 early promoter (Rigoy In Williamson ea. ), Genetic Engineering, Vol .3. Academic Press, London, p.83-141(1982)), EF-1 promoter(Kimら Ge ne , P.217-223 (1990)), CAG promoter(Niwa et al. Gene 108, p.193-200 (1991)), RSV LTR promoter^ Cull en Methods in Enzymology 152, p.684-704 (1987), SR a promoter (Takebe et al. Mol. Cell. Biol. 8, p. 66 (1988)), CMV immediate early promoter^ Seed and Aruffo Proc. Natl. Acad. Sci. USA 84, p.3365-3369 (1987)), SV40 late promoter(Gheysen and Fiers J. Mol. A ppl. Genet. 丄, p.385-394 (1982)), Adenovirus late promoter (Kaufman et a 1. Mol. Cell. Biol. 9, p. 946 (1989)), HSV TK promoter等の一般的に使用 できるプロモ一夕一であれば何を用いてもよい。 As a method for screening a protein that binds to the protein using the protein of the present invention, for example, many methods known to those skilled in the art can be used. Such screening can be performed, for example, by immunoprecipitation. Specifically, it can be performed as follows. The gene encoding the protein of the present invention is introduced into a vector for expression of a foreign gene such as pSV2neo, pcDNA I, or pCD8, so that the gene is expressed in animal cells or the like. Promoters used for expression include SV40 early promoter (Rigoy In Williamson ea.), Genetic Engineering, Vol. 3. Academic Press, London, p. 83-141 (1982)), EF-1 promoter (Kim et al. , P.217-223 (1990)), CAG promoter (Niwa et al. Gene 108, p.193-200 (1991)), RSV LTR promoter ^ Cull en Methods in Enzymology 152, p.684-704 (1987) , SR a promoter (Takebe et al. Mol. Cell. Biol. 8, p. 66 (1988)), CMV immediate early promoter ^ Seed and Aruffo Proc. Natl. Acad. Sci. USA 84, p. 3365-3369 ( 1987)), SV40 late promoter (Gheysen and Fiers J. Mol. A ppl. Genet. II, p. 385-394 (1982)), Adenovirus late promoter (Kaufman et a 1. Mol. Cell. Biol. 9, p. 946 (1989)), HSV TK promoter, and any other commonly used promoters can be used.
動物細胞に遺伝子を導入することで外来遺伝子を発現させるためには、 エレク トロポレーシヨン法(Chu,G. et al. Nuc 1. Ac id Res. 15, 1311-1326 (1987))、 リン酸カルシウム法(Chen,C and Okayama,H. Mol. Cell. Biol. 7, 2745-2752 (1 987))、 DEAEデキストラン法 (Lopata,M.A. et al. Nuc 1. Ac ids Res. 12, 5707- 5717 (1984); Sussman,D.J. and Milman3G. Mol. Cell. Biol. , 1642-1643 (1 985))、 リポフエクチン法(Derijard,B. Cell 7, 1025-1037 (1994); Lamb,B.T. et al. Nature Genetics 5, 22 - 30 (1993); Rabindran, S.K. et al. Science 259, 230-234 ( 1993) )等の方法があるが、 いずれの方法によってもよい。 In order to express a foreign gene by introducing a gene into an animal cell, the electroporation method (Chu, G. et al. Nuc 1. Acid Res. 15, 1311-1326 (1987)) and the calcium phosphate method (Chen , C and Okayama, H. Mol. Cell. Biol. 7, 2745-2752 (1 987)), DEAE dextran method (Lopata, MA et al. Nuc 1. Acids Res. 12, 5707-5717 (1984); Sussman, DJ and Milman 3 G. Mol. Cell. Biol., 1642-1643 (1 985)), lipofectin method (Derijard, B. Cell 7, 1025-1037 (1994)); Lamb, BT et al. Nature Genetics 5 , 22-30 (1993); Rabindran, SK et al. Science 259, 230-234 (1993)) and the like.
特異性の明らかとなっているモノクローナル抗体の認識部位 (ェピトープ) を 本発明の蛋白質の N末または C末に導入することにより、 モノクローナル抗体 の認識部位を有する融合蛋白質として本発明の蛋白質を発現させることができる 。 用いるェピトープ一抗体系としては市販されているものを利用することができ る (実験医学 1^, 85-90 (1995))。 マルチクローニングサイ トを介して、 β— ガラクトシダーゼ、 マルトース結合蛋白質、 グル夕チオン S-トランスフェラー ゼ、 緑色蛍光蛋白質 (GFP) などとの融合蛋白質を発現することができるベクタ —が市販されている。  The protein of the present invention is expressed as a fusion protein having a monoclonal antibody recognition site by introducing a recognition site (epitope) of the monoclonal antibody of which specificity is known into the N-terminal or C-terminal of the protein of the present invention. be able to . A commercially available epitope-antibody system can be used (Experimental Medicine 1 ^, 85-90 (1995)). Vectors that can express a fusion protein with β-galactosidase, maltose binding protein, glutathione S-transferase, green fluorescent protein (GFP), and the like via a multicloning site are commercially available.
融合蛋白質にすることにより本発明の蛋白質の性質をできるだけ変化させない ようにするために数個から十数個のアミノ酸からなる小さなェピトープ部分のみ を導入して、 融合蛋白質を調製する方法も報告されている。 例えば、 ポリヒスチ ジン (His-tag) 、 インフルエンザ凝集素 HA、 ヒト c- myc、 FLAG, Vesicular st omatitisウィルス糖蛋白質 (VSV- GP)、 T7 genel0蛋白質 (T7-tag) 、 ヒト単 純へルぺスウィルス糖蛋白質 (HSV- tag)、 E-tag (モノクローナルファージ上 のェピトープ) などのェピトープとそれを認識するモノクローナル抗体を、 本発 明の蛋白質に結合する蛋白質のスクリーニングのためのェピトーブー抗体系とし て利用できる (実験医学 , 85-90 ( 1995 ) )。 In order to minimize the property of the protein of the present invention by changing it into a fusion protein, a method for preparing a fusion protein by introducing only a small epitope portion consisting of several to several tens of amino acids has been reported. I have. For example, polyhistidine (His-tag), influenza agglutinin HA, human c-myc, FLAG, Vesicular stomatitis virus glycoprotein (VSV-GP), T7 genel0 protein (T7-tag), human pure virus We developed epitopes such as viral glycoproteins (HSV-tags) and E-tags (epitopes on monoclonal phages) and monoclonal antibodies that recognize them. It can be used as an Epitoboo antibody system for screening proteins that bind to the Myoprotein (Experimental Medicine, 85-90 (1995)).
免疫沈降においては、 これらの抗体を、 適当な界面活性剤を利用して調製した 細胞溶解液に添加することにより免疫複合体を形成させる。 この免疫複合体は本 発明の蛋白質、 それと結合能を有する蛋白質、 および抗体からなる。 上記ェピト ープに対する抗体を用いる以外に、 本発明の蛋白質に対する抗体を利用して免疫 沈降を行うことも可能である。 本発明の蛋白質に対する抗体は、 例えば、 本発明 の蛋白質をコードする遺伝子を適当な大腸菌発現ベクターに導入して大腸菌内で 発現させ、 発現させた蛋白質を精製し、 これをゥサギやマウス、 ラット、 ャギ、 ニヮトリなどに免疫することで調製することができる。 また、 合成した本発明の 蛋白質の部分ペプチドを上記の動物に免疫することによって調製することもでき る。  In immunoprecipitation, an immune complex is formed by adding these antibodies to a cell lysate prepared using an appropriate surfactant. This immune complex comprises the protein of the present invention, a protein capable of binding thereto, and an antibody. In addition to using antibodies against the above-mentioned epitopes, immunoprecipitation can also be performed using antibodies against the protein of the present invention. Antibodies against the protein of the present invention include, for example, a gene encoding the protein of the present invention introduced into an appropriate E. coli expression vector, expressed in E. coli, and the expressed protein is purified. It can be prepared by immunizing goats and chickens. Alternatively, it can be prepared by immunizing the above animal with the synthesized partial peptide of the protein of the present invention.
免疫複合体は、 例えば、 抗体がマウス IgG抗体であれば、 Protein A Sepharo seや Protein G Sepharoseを用いて沈降させることができる。 また、 本発明の 蛋白質を、 例えば、 GSTなどのェピトープとの融合蛋白質として調製した場合に は、 グル夕チオン- Sepharose 4Bなどのこれらェピトープに特異的に結合する物 質を利用して、 本発明の蛋白質の抗体を利用した場合と同様に、 免疫複合体を形 成させることができる。  If the antibody is a mouse IgG antibody, for example, the immune complex can be precipitated using Protein A Sepharose or Protein G Sepharose. Further, when the protein of the present invention is prepared as a fusion protein with an epitope such as GST, for example, a substance specifically binding to these epitopes such as glutathione-Sepharose 4B is used to prepare the protein of the present invention. An immune complex can be formed in the same manner as in the case where an antibody of the above protein is used.
免疫沈降の一般的な方法については、 例えば、 文献 (Harlow, E. and Lane, D .: Antibodies, pp.511-552, Cold Spring Harbor Laboratory publications, New York ( 1988)) 記載の方法に従って、 または準じて行えばよい。  For general methods of immunoprecipitation, for example, according to the method described in the literature (Harlow, E. and Lane, D .: Antibodies, pp. 511-552, Cold Spring Harbor Laboratory publications, New York (1988)), or It may be performed according to.
免疫沈降された蛋白質の解析には SDS- PAGEが一般的であり、 適当な濃度のゲ ルを用いることで蛋白質の分子量により結合していた蛋白質を解析することがで きる。 また、 この際、 一般的には本発明の蛋白質に結合した蛋白質は、 クマシ一 染色や銀染色といった蛋白質の通常の染色法では検出することは困難であるので 、 放射性同位元素である 35S-メチォニンや35 S-システィンを含んだ培養液で細胞 を培養し、 該細胞内の蛋白質を標識して、 これを検出することで検出感度を向上 させることができる。 蛋白質の分子量が判明すれば直接 SDS-ポリアクリルアミ ドゲルから目的の蛋白質を精製し、 その配列を決定することもできる。 SDS-PAGE is generally used for analysis of immunoprecipitated proteins. By using an appropriate concentration of gel, the bound proteins can be analyzed by the molecular weight of the protein. In addition, at this time, it is generally difficult to detect the protein bound to the protein of the present invention by ordinary staining methods such as Coomassie staining and silver staining, so that 35 S- Cells in culture containing methionine and 35 S-cysteine Is cultured, the protein in the cell is labeled, and the detection is performed, whereby the detection sensitivity can be improved. Once the molecular weight of the protein is known, the protein of interest can be purified directly from the SDS-polyacrylamide gel and its sequence determined.
また、 本発明の蛋白質を用いて、 該蛋白質に結合する蛋白質を単離する方法と しては、 例えば、 ウェストウエスタンブロッテイング法 (Skolnik, E.Y. et al . , Cell ( 1991 ) 65, 83-90) を用いて行うことができる。 すなわち、 本発明の蛋 白質と結合する蛋白質を発現していることが予想される細胞、 組織、 臓器 (例え ば、 脂肪細胞や実施例におけるノザンブロッテイングにより発現が認められた組 織) よりファージベクター (人 gtll, ZAPなど) を用いた cDNAライブラリ一を 作製し、 これを LB-ァガロース上で発現させフィル夕一に発現させた蛋白質を固 定し、 精製して標識した本発明の蛋白質と上記フィル夕一とを反応させ、 本発明 の蛋白質と結合した蛋白質を発現するプラークを標識により検出すればよい。 本 発明の蛋白質を標識する方法としては、 ピオチンとアビジンの結合性を利用する 方法、 本発明の蛋白質又は本発明の蛋白質に融合したぺプチド又はポリぺプチド (例えば GSTなど) に特異的に結合する抗体を利用する方法、 ラジオアイソト ープを利用する方法又は蛍光を利用する方法等が挙げられる。  Examples of the method for isolating a protein that binds to the protein using the protein of the present invention include, for example, the West Western blotting method (Skolnik, EY et al., Cell (1991) 65, 83-90). ). That is, cells, tissues, and organs that are expected to express the protein that binds to the protein of the present invention (for example, fat cells and tissues in which expression has been observed by Northern blotting in the Examples) are phage-derived. A cDNA library using a vector (human gtll, ZAP, etc.) was prepared, and expressed on LB-agarose. The protein expressed on a filter was fixed, purified and labeled with the protein of the present invention. The plaque expressing the protein bound to the protein of the present invention may be detected by labeling by reacting the above-mentioned filter with the protein. Examples of the method for labeling the protein of the present invention include a method utilizing the binding property of biotin and avidin, a method of specifically binding to the protein of the present invention or a peptide or polypeptide fused to the protein of the present invention (for example, GST). Examples of the method include a method using an antibody, a method using a radioisotope, and a method using fluorescence.
また、 本発明のスクリーニング方法の他の態様としては、 細胞を用いた 2-ハ イブリツ ドシステム (Fields, S. , and Sternglanz, R. , Trends . Genet . ( 1994) 1 0, 286-292、 Dalton S, and Treisman R ( 1992 )Characterization of SAP- 1, a protein recruited by serum response factor to the c-fos serum respon se element. Cell 68, 597-612、 「MATCHMARKER Two-Hybrid Systemj , 「Mamm alian MATCHMAKER Two-Hybrid Assay Kit」, 「MATCHMAKEE One-Hybrid System 」 (いずれもクロンテック社製)、 「HybriZAP Two-Hybrid Vector Systemj (ス トラ夕ジーン社製)) を用いて行う方法が挙げられる。 2 -ハイブリッドシステム においては、 本発明の蛋白質またはその部分ペプチドを SRF DNA結合領域また は GAL4 DNA結合領域と融合させて酵母細胞の中で発現させ、 本発明の蛋白質と 結合する蛋白質を発現していることが予想される細胞より、 VP16または GAL4転 写活性化領域と融合する形で発現するような cDNAライプラリ一を作製し、 これ を上記酵母細胞に導入し、 検出された陽性クローンからライブラリ一由来 cDNA を単離する (酵母細胞内で本発明の蛋白質と結合する蛋白質が発現すると、 両者 の結合によりレポーター遺伝子が活性化され、 陽性のクローンが確認できる) 。 単離した cDNAを大腸菌に導入して発現させることにより、 該 cDNAがコードす る蛋白質を得ることができる。 これにより本発明の蛋白質に結合する蛋白質また はその遺伝子を調製することが可能である。 2-ハイプリッドシステムにおいて 用いられるレポ一夕一遺伝子としては、 例えば、 HIS3遺伝子の他、 Ade2遺伝子 、 LacZ遺伝子、 CAT遺伝子、 ルシフヱラ一ゼ遺伝子、 PAI-1 (Plasminogen acti vator inhibitor typel) 遺伝子等が挙げられるが、 これらに制限されない。 2 ハイブリッド法によるスクリーニングは、 酵母の他、 哺乳動物細胞などを使って 行うこともできる。 Further, as another embodiment of the screening method of the present invention, a 2-hybrid system using cells (Fields, S., and Sternglanz, R., Trends. Genet. (1994) 10, 286-292, Dalton S, and Treisman R (1992) Characterization of SAP-1, a protein recruited by serum response factor to the c-fos serum response element.Cell 68, 597-612, `` MATCHMARKER Two-Hybrid Systemj, '' Mamm alian MATCHMAKER Two-Hybrid Assay Kit ”,“ MATCHMAKEE One-Hybrid System ”(all manufactured by Clontech), and“ HybriZAP Two-Hybrid Vector Systemj ”(Strata Gene) are used. In the system, the protein of the present invention or a partial peptide thereof is fused with an SRF DNA binding region or GAL4 DNA binding region and expressed in yeast cells, and the protein of the present invention and From the cells that are expected to express the protein to be bound, a cDNA library was prepared that could be expressed in a form fused with the VP16 or GAL4 transcription activation region, and introduced into the yeast cells described above for detection. The library-derived cDNA is isolated from the obtained positive clones (when the protein that binds to the protein of the present invention is expressed in yeast cells, the binding of the two activates the reporter gene, and a positive clone can be confirmed). The protein encoded by the cDNA can be obtained by introducing the isolated cDNA into Escherichia coli for expression. As a result, it is possible to prepare a protein that binds to the protein of the present invention or a gene thereof. The repo overnight gene used in the 2-hybrid system includes, for example, the HIS3 gene, Ade2 gene, LacZ gene, CAT gene, luciferase gene, PAI-1 (Plasminogen activator inhibitor typel) gene, etc. But not limited to these. Screening by the two-hybrid method can be performed using mammalian cells in addition to yeast cells.
本発明の蛋白質と結合する化合物のスクリーニングは、 ァフィ二テイク口マト グラフィ一を用いて行うこともできる。 例えば、 本発明の蛋白質をァフィ二ティ 一力ラムの担体に固定し、 ここに本発明の蛋白質と結合する蛋白質を発現してい ることが予想される被検試料を適用する。 この場合の被検試料としては、 例えば 細胞抽出物、 細胞溶解物等が挙げられる。 被検試料を適用した後、 カラムを洗浄 し、 本発明の蛋白質に結合した蛋白質を調製することができる。  Screening for a compound that binds to the protein of the present invention can also be performed by affinity mouth chromatography. For example, the protein of the present invention is immobilized on an affinity carrier, and a test sample which is expected to express a protein that binds to the protein of the present invention is applied thereto. The test sample in this case includes, for example, a cell extract, a cell lysate, and the like. After applying the test sample, the column is washed to prepare a protein bound to the protein of the present invention.
得られた蛋白質は、 そのアミノ酸配列を分析し、 それを基にオリゴ DNAを合 成し、 該 DNAをプローブとして cDNAライブラリ一をスクリーニングすることに より、 該蛋白質をコードする DNAを得ることができる。  The obtained protein is analyzed for its amino acid sequence, an oligo DNA is synthesized based on the amino acid sequence, and a DNA encoding the protein can be obtained by screening a cDNA library using the DNA as a probe. .
本発明において、 結合した化合物を検出又は測定する手段として表面ブラズモ ン共鳴現象を利用したバイオセンサーを使用することもできる。 表面プラズモン 共鳴現象を利用したバイオセンサーは、 本発明の蛋白質と被検化合物との間の相 互作用を微量の蛋白質を用いてかつ標識することなく、 表面プラズモン共鳴シグ ナルとしてリアルタイムに観察することが可能である (例えば BIAcore、 Pharma cia製) 。 したがって、 BIAcore等のバイオセンサ一を用いることにより本発明 の蛋白質と被検化合物との結合を評価することが可能である。 In the present invention, a biosensor utilizing the surface plasmon resonance phenomenon can be used as a means for detecting or measuring the bound compound. A biosensor utilizing the surface plasmon resonance phenomenon can be used to detect the interaction between the protein of the present invention and a test compound using a trace amount of protein and without labeling the surface plasmon resonance signal. It can be observed in real time as a null (for example, BIAcore, manufactured by Pharmacia). Therefore, it is possible to evaluate the binding between the protein of the present invention and a test compound by using a biosensor such as BIAcore.
また、 蛋白質に限らず、 本発明の蛋白質に結合する化合物 (ァゴ二ストおよび アン夕ゴニストを含む) を単離する方法としては、 例えば、 固定した本発明の蛋 白質に、 合成化合物、 天然物バンク、 もしくはランダムファージペプチドデイス プレイライブラリーを作用させ、 本発明の蛋白質に結合する分子をスクリーニン グする方法や、 コンビナトリアルケミストリー技術によるハイスループットを用 いたスクリーニング方法 (Wrighton NC; Farrell FX; Chang R Kashyap AK; B arbone FP; Mulcahy LS Johnson DL; Barrett RW; Jolliffe LK; Dower WJ. 5 Small peptides as potent mimetics of the protein hormone erythropoieti n, Science (環 ITED STATES) Jul 26 1996, 273 p458- 64、 Verdine GL. , The combinatorial chemistry of nature . Nature (ENGLAND) Nov 7 1996, 384 pi 1-13、 Hogan JC Jr. , Directed combinatorial chemistry. Nature ( ENGLAND ) Nov 7 1996, 384 pl7-9) が当業者に公知である。 Methods for isolating not only proteins but also compounds that bind to the protein of the present invention (including agonist and angelic gonist) include, for example, immobilized protein of the present invention, synthetic compounds, and natural compounds. A method for screening molecules that bind to the protein of the present invention using a product bank or a random phage peptide display library, and a screening method using high throughput by combinatorial chemistry technology (Wrighton NC; Farrell FX; Chang R Kashyap AK; Barbone FP; Mulcahy LS Johnson DL; Barrett RW; Jolliffe LK; Dower WJ. 5 Small peptides as potent mimetics of the protein hormone erythropoietin, Science (ring ITED STATES) Jul 26 1996, 273 p458-64, Verdine GL., The combinatorial chemistry of nature.Nature (ENGLAND) Nov 7 1996, 384 pi 1-13, Hogan JC Jr., Directed combinatorial chemistry.Nature (ENG LAND) Nov 7 1996, 384 pl7-9) are known to those skilled in the art.
本発明のスクリーニングにより単離しうる化合物は、 本発明の蛋白質の活性を 調節するための桀剤の候補となり、 本発明の蛋白質の発現異常や機能異常などに 起因する疾患や本発明の蛋白質の活性を制御することにより治療可能な疾患の治 療への応用が考えられる。 本発明のスクリーニング方法を用いて単離しうる化合 物の構造の一部を、 付加、 欠失及び/又は置換により変換される物質も、 本発明 の蛋白質に結合する化合物に含まれる。  The compound that can be isolated by the screening of the present invention is a candidate for the agent for regulating the activity of the protein of the present invention, and is a disease caused by abnormal expression or dysfunction of the protein of the present invention or the activity of the protein of the present invention. It can be applied to the treatment of diseases that can be treated by controlling the disease. Substances that can be converted by addition, deletion, and / or substitution of a part of the structure of a compound that can be isolated using the screening method of the present invention are also included in the compounds that bind to the protein of the present invention.
本発明の蛋白質、 または本発明のスクリーニングにより単離しうる化合物をヒ トゃ哺乳動物、 例えばマウス、 ラット、 モルモット、 ゥサギ、 ニヮトリ、 ネコ、 ィヌ、 ヒヅジ、 ブ夕、 ゥシ、 サル、 マントヒヒ、 チンパンジーの医薬として使用 する場合には、 蛋白質や単離された化合物自体を直接患者に投与する以外に、 公 知の製剤学的方法により製剤化して投与を行うことも可能である。 例えば、 必要 に応じて糖衣を施した錠剤、 カプセル剤、 エリキシル剤、 マイクロカプセル剤と して経口的に、 あるいは水もしくはそれ以外の薬学的に許容し得る液との無菌性 溶液、 又は懸濁液剤の注射剤の形で非経口的に使用できる。 例えば、 薬理学上許 容される担体もしくは媒体、 具体的には、 滅菌水や生理食塩水、 植物油、 乳化剤 、 懸濁剤、 界面活性剤、 安定剤、 香味剤、 賦形剤、 べヒクル、 防腐剤、 結合剤な どと適宜組み合わせて、 一般に認められた製薬実施に要求される単位用量形態で 混和することによって製剤化することが考えられる。 これら製剤における有効成 分量は指示された範囲の適当な容量が得られるようにするものである。 The protein of the present invention, or a compound that can be isolated by the screening of the present invention, is used in a human mammal, such as a mouse, a rat, a guinea pig, a heron, a chicken, a cat, a dog, a sheep, a bush, a monkey, a monkey, a baboon, When used as a chimpanzee medicament, the protein or isolated compound itself can be administered directly to a patient, or can be formulated and administered by a known pharmaceutical method. For example, need Injection of tablets, capsules, elixirs, microcapsules orally, or sterile solutions or suspensions with water or other pharmaceutically acceptable liquids, as appropriate for sugar coating It can be used parenterally in dosage form. For example, pharmacologically acceptable carriers or vehicles, specifically, sterile water or saline, vegetable oils, emulsifiers, suspending agents, surfactants, stabilizers, flavoring agents, excipients, vehicles, It is possible to formulate the drug product by combining it with a preservative, binder and the like as appropriate and mixing it in the unit dosage form required for accepted pharmaceutical practice. The effective components of these preparations are intended to provide a suitable volume in the specified range.
錠剤、 カプセル剤に混和することができる添加剤としては、 例えばゼラチン、 コーンスターチ、 トラガントガム、 アラビアゴムのような結合剤、 結晶性セル口 ースのような賦形剤、 コーンスターチ、 ゼラチン、 アルギン酸のような膨化剤、 ステアリン酸マグネシウムのような潤滑剤、 ショ糖、 乳糖又はサヅカリンのよう な甘味剤、 ペパーミント、 ァカモノ油又はチェリ一のような香味剤が用いられる 。 調剤単位形態がカプセルである場合には、 上記の材料にさらに油脂のような液 状担体を含有することができる。 注射のための無菌組成物は注射用蒸留水のよう なべヒクルを用いて通常の製剤実施に従って処方することができる。  Additives that can be incorporated into tablets and capsules include, for example, binders such as gelatin, corn starch, tragacanth gum, acacia, excipients such as crystalline cellulose, corn starch, gelatin, and alginic acid. Suitable leavening agents, lubricants such as magnesium stearate, sweeteners such as sucrose, lactose or saccharin, and flavoring agents such as peppermint, cocoa oil or cherry are used. When the unit dosage form is a capsule, the above-mentioned materials may further contain a liquid carrier such as an oil or fat. A sterile composition for injection can be formulated using a potable vehicle such as distilled water for injection according to normal pharmaceutical practice.
注射用の水溶液としては、 例えば生理食塩水、 プドウ糖やその他の補助薬を含 む等張液、 例えば D-ソルビトール、 D-マンノース、 D-マンニトール、 塩化ナト リウムが挙げられ、 適当な溶解補助剤、 例えばアルコール、 具体的にはエタノー ル、 ポリアルコール、 例えばプロピレングリコール、 ポリエチレングリコ一ル、 非イオン性界面活性剤、 例えばポリソルベート 80 (TM) 、 HC0- 50と併用しても よい。  Aqueous injection solutions include, for example, physiological saline, isotonic solutions containing pudose and other adjuvants, such as D-sorbitol, D-mannose, D-mannitol, and sodium chloride. It may be used in combination with an agent, for example, an alcohol, specifically ethanol, a polyalcohol, for example, propylene glycol, polyethylene glycol, a nonionic surfactant, for example, polysorbate 80 (TM) or HC0-50.
油性液としてはゴマ油、 大豆油があげられ、 溶解補助剤として安息香酸ベンジ ル、 ベンジルアルコールと併用してもよい。 また、 緩衝剤、 例えばリン酸塩緩衝 液、 酢酸ナトリウム緩衝液、 無痛化剤、 例えば、 塩酸プロ力イン、 安定剤、 例え ばべンジルアルコール、 フヱノール、 酸化防止剤と配合してもよい。 調製された 注射液は通常、 適当なアンプルに充填させる。 The oily liquid includes sesame oil and soybean oil, and may be used in combination with benzyl benzoate or benzyl alcohol as a solubilizer. Also, buffers such as phosphate buffers, sodium acetate buffers, soothing agents such as proforce hydrochloride, stabilizers, such as It may be blended with benzyl alcohol, phenol and antioxidants. The prepared injection solution is usually filled into an appropriate ampoule.
患者への投与は、 例えば、 動脈内注射、 静脈内注射、 皮下注射などのほか、 鼻 腔内的、 経気管支的、 筋内的、 経皮的、 または経口的に当業者に公知の方法によ り行いうる。 投与量は、 患者の体重や年齢、 投与方法などにより変動するが、 当 業者であれば適当な投与量を適宜選択することが可能である。 また、 該化合物が DNAによりコ一ドされうるものであれば、 該 DNAを遺伝子治療用ベクタ一に組込 み、 遺伝子治療を行うことも考えられる。 投与量、 投与方法は、 患者の体重や年 齢、 症状などにより変動するが、 当業者であれば適宜選択することが可能である 本発明の蛋白質の投与量は、 その 1回投与量は投与対象、 対象臓器、 症状、 投与方法によっても異なるが、 例えば注射剤の形では通常成人 (体重 60kgとし て) においては、 1日あたり約 100〃gから 20mgであると考えられる。  Administration to patients can be performed, for example, by intraarterial injection, intravenous injection, subcutaneous injection, etc., or intranasally, transbronchially, intramuscularly, transdermally, or orally by methods known to those skilled in the art. It can do better. The dose varies depending on the weight and age of the patient, the administration method, and the like, but those skilled in the art can appropriately select an appropriate dose. Further, if the compound can be encoded by DNA, the DNA may be incorporated into a gene therapy vector to perform gene therapy. The dose and administration method vary depending on the patient's body weight, age, symptoms, etc., but those skilled in the art can appropriately select the dose of the protein of the present invention. Depending on the subject, target organ, symptoms and method of administration, for example, in the form of an injection, it is generally considered to be about 100 to 20 mg per day for an adult (with a body weight of 60 kg).
本発明の蛋白質と結合する化合物や本発明の蛋白質の活性を調節する化合物の 投与量は、 症状により差異はあるが、 経口投与の場合、 一般的に成人 (体重 60k gとして) においては、 1日あたり約 0. 1から 100mg、 好ましくは約 1. 0から 50 mg、 より好ましくは約 1. 0から 20mgであると考えられる。  The dose of the compound that binds to the protein of the present invention or the compound that modulates the activity of the protein of the present invention varies depending on the symptoms. However, in the case of oral administration, in general, in adults (with a body weight of 60 kg), 1 It is believed to be about 0.1 to 100 mg per day, preferably about 1.0 to 50 mg, more preferably about 1.0 to 20 mg.
非経口的に投与する場合は、 その 1回投与量は投与対象、 対象臓器、 症状、 投 与方法によっても異なるが、 例えば注射剤の形では通常成人 (体重 60kgとして ) においては、 通常、 1日当り約 0. 01から 30mg、 好ましくは約 0. 1から 20mg、 より好ましくは約 0. 1から 10mg程度を静脈注射により投与するのが好都合であ ると考えられる。 他の動物の場合も、 体重 60kg当たりに換算した量、 あるいは 体表面積あたりに換算した量を投与することができる。 図面の簡単な説明 図 1は、 3T3- LI細胞における本発明の遺伝子の発現をノザンプロット解析に より検出した結果を示す電気泳動写真である。 レーン 1は増殖しつつある 3T3-L 1細胞、 レーン 2はコンフルェント 3T3-L1細胞、 レーン 3は脂肪細胞に分化誘 導した 3T3- L1細胞である。 In the case of parenteral administration, the single dose varies depending on the subject of administration, target organ, symptoms, and administration method. For example, in the case of injection, it is usually 1 dose for adults (with a body weight of 60 kg). It may be convenient to administer about 0.01 to 30 mg, preferably about 0.1 to 20 mg, more preferably about 0.1 to 10 mg per day by intravenous injection. In the case of other animals, the dose can be administered in terms of the amount converted per 60 kg body weight or the amount converted per body surface area. BRIEF DESCRIPTION OF THE FIGURES FIG. 1 is an electrophoretic photograph showing the result of detecting the expression of the gene of the present invention in 3T3-LI cells by Northern blot analysis. Lane 1 shows proliferating 3T3-L1 cells, lane 2 shows confluent 3T3-L1 cells, and lane 3 shows 3T3-L1 cells induced to differentiate into adipocytes.
図 2は、 種々の組織における本発明の遺伝子の発現をノザンプロット解析によ り検出した結果を示す電気泳動写真である。 レーンはそれそれ、 1 :心臓、 2 : 脳、 3 :脾臓、 4 :肺、 5 :肝臓、 6 :骨格筋、 Ί :腎臓、 8 :精巣、 である。 図 3は、 対数期 (log phase) の 3T3-L1、 コンフルェント状態の 3T3- Ll、 脂 肪細胞に分化誘導後 1から 9日目の 3T3- L1における #103から #106および GAPDH の発現を、 ノザンブロット解析により検出した結果を示す電気泳動写真である。 図 4は、 インスリンを OnM (対照: MOCK)、 25nM、 250nM加えた脂肪細胞にお ける #105および GAPDHの発現を、 ノザンプロット解析により検出した結果を示 す電気泳動写真である。  FIG. 2 is an electrophoretic photograph showing the results of detecting the expression of the gene of the present invention in various tissues by Northern blot analysis. The lanes are as follows: 1: heart, 2: brain, 3: spleen, 4: lung, 5: liver, 6: skeletal muscle, :: kidney, 8: testis. Figure 3 shows the expression of # 103 to # 106 and GAPDH in 3T3-L1 in log phase, 3T3-Ll in confluent state, and 3T3-L1 1 to 9 days after induction of differentiation into fat cells. It is an electrophoresis photograph which shows the result detected by Northern blot analysis. FIG. 4 is an electrophoretic photograph showing the results of Northern blot analysis of the expression of # 105 and GAPDH in adipocytes to which OnM (control: MOCK), 25 nM, and 250 nM insulin was added.
図 5は、 様々な癌細胞株、 脂肪細胞および 3T3-L1における #104の発現を、 ノ ザンブロット解析により検出した結果を示す電気泳動写真である。 発明を実施するための最良の形態  FIG. 5 is an electrophoretic photograph showing the results of detecting the expression of # 104 in various cancer cell lines, adipocytes and 3T3-L1 by Northern blot analysis. BEST MODE FOR CARRYING OUT THE INVENTION
以下に実施例により本発明をさらに詳細に説明するが、 本発明はこれら実施例 に制限されるものではない。  Hereinafter, the present invention will be described in more detail with reference to Examples, but the present invention is not limited to these Examples.
[実施例 1 ] cDNAライブラリの構築およびスクリ一ニング  [Example 1] Construction and screening of cDNA library
レトロウイルスベクター pMX-SST (Kojima . and Kitamura5 T. ( 1999 ) Nature Biotechnol . l7, 487-490) を用いて、 cDNAライブラリ構築および発現を行なつ た。 A cDNA library was constructed and expressed using the retroviral vector pMX-SST (Kojima. And Kitamura 5 T. (1999) Nature Biotechnol. 17, 487-490).
ファスト · トラック 2. 0 mRNA単離キット (インビトロゲン社、 カリフオル二 ァ州力一ルスバッ ド) を用いて、 製造元のプロトコ一ルに従って、 分化した ST3 -L1細胞から抽出した。 スーパ一スクリプト 'チヨイス 'システム (ギブコ- BRL社、 ロックビル、 メ リーランド州、 アメリカ) を用いて、 相補的 DNA (cDNA) をランダム 6量体によ つてポリ(A)+RNAから合成し、 BstXIアダプタ一を用いて pMX- SSTベクタ一の B stXI部位に揷入した (インビトロゲン社、 力一ルスバッド、 カリフォルニア州 ) 。 SST-REXライブラリを構築するために、 ライゲ一シヨンした DNAを DH10B細 胞に増幅して (エレクトロマックス、 ギブコ -BRL社) 、 キアゲン ·プラスミ ド キット (キアゲン -インク、 バレンシア、 カリフォルニア州) を用いてライブラ リ DNAを調製した。 Differentiated ST3-L1 cells were extracted using Fast Track 2.0 mRNA isolation kit (Invitrogen, Lisbad, Calif.) According to the manufacturer's protocol. Complementary DNA (cDNA) was synthesized from poly (A) + RNA using random hexamers using the Superscript 'Thiois' system (Gibco-BRL, Rockville, Maryland, USA). The BstXI adapter was used to insert into the BstXI site of the pMX-SST vector (Invitrogen, Lisbad, CA). To construct the SST-REX library, the ligated DNA was amplified into DH10B cells (Electromax, Gibco-BRL) and the Qiagen Plasmid kit (Qiagen-Inc, Valencia, CA) was used. To prepare library DNA.
SST- REXライブラリを提示する高カ価レトロウィルスを、 パッケージングした 細胞株爾 23 (Pear,W. S. et al . (1993) Proc. Natl . Acad. Sci . U. S.A. 90, 839 A cell line containing a high-potency retrovirus presenting the SST-REX library was packaged (Pear, W. S. et al. (1993) Proc. Natl. Acad. Sci. U. S.A. 90, 839
2-8396) を用いて産生し、 記述のように Ba/F3細胞に感染させた。 1日感染さ せた後細胞を 3回洗浄し、 96ウェルマルチ夕イタ一プレート (103個/ゥエル) を用いて IL- 3の非存在下でクローンを選択した。 2-8396) and infected Ba / F3 cells as described. Washing the cells three times after infection day, and clones were selected in the absence of IL- 3 using 96 well multi-evening Ita first plate (10 3 / Ueru).
12日後、 ゲノム DNAを因子非依存的 Ba/F3クローンから抽出し、 ゲノム PCR に供し、 ベクタ一プライマーを用いて、 組み入れられた cDNAを回収した (GGGG GTGGACCATCCTCTA/配列番号: 12、 および CGCGCAGCTGTAAACGGTAG/配列番号: 1 3) 。 ジーンアンプ PCRシステム 2400 (パーキン 'エルマ一、 ノーウォーク、 コ ネチカヅト州) および LA Taqポリメラ一ゼ (夕カラ、 京都、 日本) を用いて、 PCRを 30サイクル (98°Cで変性 20秒、 68°Cでァ二一リングおよび伸長 2分) 行 つた。 得られた PCR断片を、 Taqダイターミネ一夕一 ·サイクルシークェンシン グキット (アプライ ド ·バイオシステムズ ·インク、 フォス夕一シティ、 カリフ オルニァ州) を用いてシ一クェンシングして、 自動シークェンサ一 (310遺伝子 アナライザー、 アプライ ド 'バイオシステムズ ·インク) によって分析した。 本発明者等は、 クローン 9 X 105個をスクリーニングした。 全体でゥヱル 1152 個中 71ゥエルにおいて増殖しつつある細胞は単一の PCRバンドを生じ、 これを さらなる分析に供した。 なお、 実験に用いた分化した 3T3- LI細胞は、 以下のようにして調製した。 3T 3-L1細胞を、 10 %FCS、 50単位/ mlペニシリン、 および 50〃g/mlストレブトマ イシンを含む DMEM (DMEM-FCS) 中で 5 %C02において 37°Cで培養した。 細胞を コンフルェントになるまで増殖させ、 2日間維持した。 分化を誘導するために、 培地を 0. 5IDM 卜メチル -3-イソプチルキサンチン、 0 . 25 /Mデキサメタゾン、 お よび 10〃g/mlインスリンを含む DMEM- FCSに交換した。 2日後、 培地を DMEM -; FC Sに交換して、 細胞を分化させた。 マウス IL- 3依存的前 B細胞株である Ba/F3 を 10 %FCSおよび 2 ng/mlマウス IL- 3 (MDシステムズ) を含む RPMI 1640培地 で培養した。 Twelve days later, genomic DNA was extracted from factor-independent Ba / F3 clones, subjected to genomic PCR, and the integrated cDNA was recovered using vector primers (GGGG GTGGACCATCCTCTA / SEQ ID NO: 12, and CGCGCAGCTGTAAACGGTAG / sequence). Number: 1 3). Using the GeneAmp PCR System 2400 (Perkin Elmer, Norwalk, Conn.) And LA Taq Polymerase (Yukara, Kyoto, Japan), perform 30 cycles of PCR (denaturation at 98 ° C for 20 seconds, 68 (Ringing and elongation at 2 ° C for 2 minutes). The obtained PCR fragments were sequenced using the Taq Dye Mineral One-Day Cycle Sequencing Kit (Applied Biosystems, Inc., Foss Yuichi City, Calif-Ornier) to obtain an automated sequencer (310 Analyzed by Gene Analyzer, Applied'Biosystems, Inc.). We screened 9 × 10 5 clones. Cells growing in a total of 71 cells out of 1152 cells generated a single PCR band, which was subjected to further analysis. The differentiated 3T3-LI cells used in the experiment were prepared as follows. The 3T 3-L1 cells were cultured in 10% FCS, 50 units / ml penicillin, and 50〃G / ml Sutorebutoma containing leucine DMEM (DMEM-FCS) 5% C0 2 at 37 ° C in. Cells were grown to confluence and maintained for 2 days. To induce differentiation, the medium was changed to DMEM-FCS containing 0.5 IDM trimethyl-3-isobutylxanthine, 0.25 / M dexamethasone, and 10 μg / ml insulin. Two days later, the medium was changed to DMEM-; FCS to differentiate the cells. Ba / F3, a mouse IL-3 dependent pre-B cell line, was cultured in RPMI 1640 medium containing 10% FCS and 2 ng / ml mouse IL-3 (MD Systems).
[実施例 2 ] 単離された cDNAクロ一ンの分析  [Example 2] Analysis of isolated cDNA clone
組み込み体 71個中 63個は、 既知の蛋白質 28個に関する cDNAの 5'配列を含 み、 それらの全てはシグナル配列を含んでいた (表 1 ) 。 63 out of 71 integrants contained the 5 'sequence of the cDNA for 28 known proteins, all of which contained the signal sequence (Table 1).
4 表 1 4 Table 1
No. 同一性 クローン数 分泌タンパク質 (全) 50 No. Identity Number of clones Secreted protein (all) 50
細胞外マトリックスおよび関連タンパク質 (全) 28  Extracellular matrix and related proteins (all) 28
1 Procollagen alpha-2(I) 5  1 Procollagen alpha-2 (I) 5
2 Collagen alpha- 1 (ΙΠ) 2  2 Collagen alpha- 1 (ΙΠ) 2
3 Collagen alpha- 1 (VI) 2  3 Collagen alpha-1 (VI) 2
4 Collagen alpha-2 (IV) 2  4 Collagen alpha-2 (IV) 2
5 Collagen alpha- 1 (IV) 1  5 Collagen alpha- 1 (IV) 1
6 Collagen alpha- 1 (XV) 1  6 Collagen alpha- 1 (XV) 1
7 Procollagen C-proteinase enhancer protein 3  7 Procollagen C-proteinase enhancer protein 3
8 Cysteine- rich glycoprotein SPARC 4  8 Cysteine-rich glycoprotein SPARC 4
9 Extracellular matrix associated protein (Sc l) 3  9 Extracellular matrix associated protein (Sc l) 3
10 Entactin (ENT) 2  10 Entactin (ENT) 2
11 Fibulin-2 1  11 Fibulin-2 1
12 Lysyl oxidase 1  12 Lysyl oxidase 1
13 Dystroglycan (DAG1) 1 その他の分泌タンパク質 (全) 22  13 Dystroglycan (DAG1) 1 Other secreted proteins (all) 22
14 Adipocyte complement-related protein of 30kDa (Acrp30) 10  14 Adipocyte complement-related protein of 30kDa (Acrp30) 10
15 Sulfated glycoprotein (Sgpl) 4  15 Sulfated glycoprotein (Sgpl) 4
16 Lipoprotein lipase 2  16 Lipoprotein lipase 2
17 Cy statin C 2  17 Cy statin C 2
. 18 FK506-binding protein (FKBP23) 1  . 18 FK506-binding protein (FKBP23) 1
19 Epithelin 1  19 Epithelin 1
20 Disulfide isomerase- related protein (ERp72) 1  20 Disulfide isomerase- related protein (ERp72) 1
21 Interferon receptor soluble isoform (EFNAR2) 1 膜タンパク質 (全) 13  21 Interferon receptor soluble isoform (EFNAR2) 1 Membrane protein (all) 13
22 Amyloid precursor-like protein 2 (APLP2) 4  22 Amyloid precursor-like protein 2 (APLP2) 4
23 Amyloid beta protein precursor 1  23 Amyloid beta protein precursor 1
24 Syndecan-1 3  24 Syndecan-1 3
25 Lysosomal membrane glycoprotein-typeA 2  25 Lysosomal membrane glycoprotein-typeA 2
26 Rat ribophorin I homologue 1  26 Rat ribophorin I homologue 1
2 / Tissue factor Cf-3 1  2 / Tissue factor Cf-3 1
28 Putatative transmembrane receptor (frizzled 7) 1  28 Putatative transmembrane receptor (frizzled 7) 1
(全) 63 同定された蛋白質 63個中、 50個 (79% ) が分泌型蛋白質であり、 分泌型蛋白 50個中 28個が細胞外マトリクス (ECM) 蛋白質または関連蛋白質であった。 13個は膜蛋白質であった。 残りの 8クローンはシグナルぺプチドを有する独立 した新規蛋白質 7個を表した (表 2 ) 。 (Total) 63 Of the 63 proteins identified, 50 (79%) were secreted proteins, and 28 of 50 secreted proteins were extracellular matrix (ECM) proteins or related proteins. Thirteen were membrane proteins. The remaining eight clones represented seven independent novel proteins with signal peptides (Table 2).
表 2 クローン 類似性 クローン数 Table 2 Clones Similarity Number of clones
101 No similarity to database sequences 101 No similarity to database sequences
102 No similarity to database sequences  102 No similarity to database sequences
103 No similarity to database sequences  103 No similarity to database sequences
104 No similarity to database sequences  104 No similarity to database sequences
105 Cell surface protein MCAR  105 Cell surface protein MCAR
106 Collagen alpha 1(XI) 2 107 No similarity to database sequences 1 蛋白質 5個は既知の蛋白質との類似性を示さなかった。 1つは細胞表面蛋白質 MCAR (マウスコクサツキ一ウィルスおよびアデノウイルス受容体) (Bergelson ,J.M. et al . ( 1998) J. Virol . 72, 415-419) と類似であり、 もう一つはコラー ゲン類と類似性があった。  106 Collagen alpha 1 (XI) 2 107 No similarity to database sequences 1 Five proteins did not show similarity to known proteins. One is similar to the cell surface protein MCAR (mouse oxpecker virus and adenovirus receptor) (Bergelson, JM et al. (1998) J. Virol. 72, 415-419) and the other is collagen And similarities.
[実施例 3 ] 分化の際の新規脂肪細胞遺伝子の発現  [Example 3] Expression of a novel adipocyte gene during differentiation
ノザンプロット分析によって、 分化の際のクローン #101- 107を含む新規遺伝 子 7個の発現パターンを調べた (図 1 ) 。 ポリ(A)+MAは、 実施例 1と同様の手 法で、 増殖しつつある 3T3- L1細胞、 コンフルェント 3T3- L1細胞、 および脂肪 細胞に分化誘導した 3T3- L1細胞から調製した。  Northern blot analysis examined the expression patterns of seven novel genes, including clone # 101-107, during differentiation (Figure 1). Poly (A) + MA was prepared in the same manner as in Example 1 from proliferating 3T3-L1 cells, confluent 3T3-L1 cells, and 3T3-L1 cells induced to differentiate into adipocytes.
ポリ(A)+RNA 1〃gを、 2 %ホルムアルデヒドを含む 1.0%ァガロース ·ゲル 上で電気泳動して、 その後ハイボンド- N-ナイロンメンプレン (アマシャム · フ アルマシア -バイオテック (株) ) に移した。 メンブレンをハイブリダィゼ一シ ヨン緩衝液 (50%ホルムアミ ド、 10 Xデンハート試薬、 5 x SSC、 0. 1%SDS、 20 0〃g/ml変性サケ精子 DNA) 中、 42°Cで因子依存的クロー に由来する3 2 P-標識 DNA断片をプローブとして調べた。 各レーンにおける RNA量を評価するために、 ローデイング対照としてメンブレンの 1つをマウスリン酸グルタルアルデヒドデ ヒドロゲナーゼ (GAPDH) に対する cDNAをプロ一ブとして調べた。 ハイブリダ ィゼ一シヨン後、 フィル夕一を 0. 1 x SSC、 0. 1 %SDS中で 42°Cで洗浄して、 ォ一 トラジオグラフィーを行った。 1 μg of poly (A) + RNA is electrophoresed on a 1.0% agarose gel containing 2% formaldehyde, and then transferred to Hybond-N-nylon membrane (Amersham Pharmacia-Biotech). did. Membrane is incubated at 42 ° C in hybridization buffer (50% formamide, 10X Denhardt's reagent, 5x SSC, 0.1% SDS, 200 μg / ml denatured salmon sperm DNA) at 42 ° C. the 3 2 P- labeled DNA fragments derived from examined as a probe. One of the membranes was used as a loading control to evaluate the amount of RNA in each lane. The cDNA for hydrogenase (GAPDH) was probed. After hybridization, the filter was washed in 0.1 × SSC, 0.1% SDS at 42 ° C. and subjected to autoradiography.
その結果、 mRNAの発現は、 クローン #102を除く全てのクローンでは分化と共 に変化した。 クローン #101、 #104、 #106、 および #107の mRNAは分化後増加した 。 クローン #105は 2つのバンドを示した。 細胞が脂肪細胞に分化すると、 低分 子量 m Aは著しく発現された。 クローン #103の場合、 mRNAの増加に関して十 分であつたのは、 3T3-L1細胞がコンフルエンスになるまで増殖することのみで あった。  As a result, mRNA expression changed with differentiation in all clones except clone # 102. The mRNA of clones # 101, # 104, # 106, and # 107 increased after differentiation. Clone # 105 showed two bands. When the cells differentiated into adipocytes, low molecular mass mA was significantly expressed. For clone # 103, the only increase in mRNA was sufficient for 3T3-L1 cells to grow to confluence.
[実施例 4 ] 多マウス組織のノザンブロヅト分析  [Example 4] Northern blot analysis of multiple mouse tissues
新規脂肪細胞由来遺伝子の多マウス組織での発現プロフィ一ルをノザンプロッ ト分析により調べた (図 2 ) 。 マウス多組織ノザンプロットをクロンテヅク社 ( パロアルト、 カリフォルニア州) から購入した。 実施例 3において、 発現が脂肪 細胞において有意に増加していることが示された、 クローン #102を除く遺伝子 6個を選択し、 検討した。  The expression profile of the novel adipocyte-derived gene in multiple mouse tissues was examined by Northern plot analysis (FIG. 2). Mouse multi-tissue Northern plots were purchased from Clontech (Palo Alto, CA). In Example 3, six genes excluding clone # 102, whose expression was shown to be significantly increased in adipocytes, were selected and examined.
その結果、 クローン #101の発現は精巣において極めて高いが、 mMAは心臓、 脾臓、 および肝臓での検出は弱かった。 クローン #103は、 心臓および肺におい て特異的に発現されたが、 精巣では低分子量のバンドの検出は弱かった。 クロー ン #104の発現は心臓および肝臓において極めて高いが、 脳、 肺および腎臓にお ける mMAの検出は弱かった。 クローン #105は心臓および脳に限って転写された 。 脳では高分子量バンドのみが検出されたことは注目に値する。 クローン #106 の発現は心臓のみに検出された。 心臓、 肝臓、 腎臓および精巣におけるクローン 顏 mRNAレベルはその他より高かった。 このように、 新規脂肪細胞由来遺伝子 の組織での発現には顕著な差が存在することが判明した。  As a result, expression of clone # 101 was extremely high in testis, but mMA was weakly detected in heart, spleen, and liver. Clone # 103 was specifically expressed in heart and lung, but low molecular weight bands were weakly detected in testis. Clone # 104 expression was extremely high in heart and liver, but weak detection of mMA in brain, lung and kidney. Clone # 105 was transcribed only in the heart and brain. It is noteworthy that only high molecular weight bands were detected in the brain. Expression of clone # 106 was detected only in the heart. Clone face mRNA levels in heart, liver, kidney and testis were higher than others. Thus, it was found that there is a remarkable difference in the expression of the novel adipocyte-derived gene in tissues.
[実施例 5 ] mRNAの経日的変化 通常の方法により、 ノザンプロット分析 (を行った。 各レーンのサンプルはそ れそれ、 対数期 (log phase) の 3T3- Ll、 コンフルェント状態の 3T3- Ll、 脂肪 細胞に分化誘導後 1から 9日目の 3T3-L1である。 #103から #106、 GAPDHをプロ ーブとした。 [Example 5] Daily change of mRNA Northern blot analysis was performed by the usual method (samples in each lane were 3T3-Ll in log phase, 3T3-Ll in confluent state, and 1 to 9 days after induction of adipocyte differentiation) This is 3T3-L1 of the eye, and # 103 to # 106, GAPDH was used as the probe.
その結果、 図 3に示すように、 #103では興味深いことに、 コンフルェントで 発現量が上昇するが、 分化誘導開始後初期の 1から 3日目までは発現量が減少し 、 ほとんど見られないが、 4日目以降再び発現量が上昇した。 #105では、 分化 誘導後 2日目までは高分子量のバンドが見られるが、 3日目からは低分子量のバ ンドのみが見られた。 #104と #106では分化誘導後 6日目までは強い発現が認め られるが、 7日目以降は発現量が減少した。  As a result, as shown in Fig. 3, interestingly, in # 103, the expression level increased in the confluent, but the expression level decreased during the first 1 to 3 days after the initiation of differentiation induction, but was hardly observed. On the fourth day, the expression level increased again. In # 105, a high molecular weight band was observed until the second day after the differentiation induction, but only a low molecular weight band was observed from the third day. In # 104 and # 106, strong expression was observed up to 6 days after induction of differentiation, but the expression level decreased after 7 days.
[実施例 6 ] インスリン (insulin) 添加による効果  [Example 6] Effect of adding insulin
通常の方法により、 ノザンプロッ ト分析を行った。 各レーンのサンプルはそれ それ、 対照の脂肪細胞、 25nM、 250nMのインスリンを加えた脂肪細胞である。 #1 05、 GAPDHをプローブとした。  Northern plot analysis was performed by the usual method. The samples in each lane are control adipocytes, adipocytes supplemented with 25 nM and 250 nM insulin, respectively. # 105, GAPDH was used as a probe.
その結果、 図 4に示すように、 250nMのインスリンを加えた脂肪細胞では #105 の発現は抑制された。  As a result, as shown in FIG. 4, expression of # 105 was suppressed in adipocytes to which 250 nM insulin was added.
[実施例 7 ] 癌細胞株における #104の発現  [Example 7] Expression of # 104 in cancer cell line
通常の方法により、 ノザンプロット分析を行った。 各レーンのサンプルは、 そ れそれ図 5に示した細胞株を使用した。 #104をプローブとした。  Northern plot analysis was performed by the usual method. The samples in each lane used the cell lines shown in FIG. 5, respectively. # 104 was used as a probe.
その結果、 図 5の通り癌細胞株 (cancer cell l ine) では種類によって低分 子量のバンドの発現が見られるものと見られないものがあった。 産業上の利用の可能性  As a result, as shown in Fig. 5, some cancer cell lines (cancer cell lines) showed low molecular weight bands and some did not. Industrial applicability
本発明により脂肪細胞に由来し、 シグナル配列を有する新規な蛋白質をコード する遺伝子が提供された。 これら遺伝子は、 その発現特性から、 脂肪細胞の分化 に関わる重要な分子である可能性がある。 本発明の蛋白質は、 医薬品開発の標的 として有用であり、 また、 本発明の蛋白質の機能を調節する化合物は、 医薬品と しての応用が期待される。 According to the present invention, a gene encoding a novel protein derived from an adipocyte and having a signal sequence is provided. These genes may be important molecules involved in adipocyte differentiation due to their expression characteristics. The protein of the present invention is a target for drug development The compound that regulates the function of the protein of the present invention is expected to be applied as a pharmaceutical.

Claims

請求の範囲 The scope of the claims
1 . 下記 (a ) から (f ) のいずれかに記載の DNA。 1. DNA according to any one of (a) to (f) below.
( a ) 配列番号: 8〜1 1のいずれかに記載のアミノ酸配列からなる蛋白質をコ ードする DNAo  (a) DNA encoding a protein consisting of the amino acid sequence of any one of SEQ ID NOs: 8 to 11
( b ) 配列番号: 1〜7のいずれかに記載の塩基配列のコード領域を含む DNA。 (b) DNA containing the coding region of the nucleotide sequence of any one of SEQ ID NOs: 1 to 7.
( c ) 配列番号: 8〜1 1のいずれかに記載のアミノ酸配列において 1若しくは 複数のアミノ酸が置換、 欠失、 挿入、 および Zまたは付加したアミノ酸配列を有 し、 配列番号: 8〜1 1のいずれかに記載のアミノ酸配列からなる蛋白質と機能 的に同等な蛋白質をコ一ドする DNA。 (c) one or more amino acids in the amino acid sequence of any one of SEQ ID NOs: 8 to 11 having substitution, deletion, insertion, and Z or added amino acid sequences, and SEQ ID NOs: 8 to 11 A DNA encoding a protein functionally equivalent to the protein comprising the amino acid sequence of any one of the above.
( d ) 配列番号: 1、 2、 7のいずれかに記載の塩基配列からなる DNAに対応 する全長 DNAによりコードされる蛋白質のアミノ酸配列において 1若しくは複 数のアミノ酸が置換、 欠失、 挿入、 および/または付加したアミノ酸配列を有し 、 該全長 DNAによりコードされる蛋白質と機能的に同等な蛋白質をコ一ドする D NA。  (d) one or more amino acids are substituted, deleted, inserted, or substituted in the amino acid sequence of the protein encoded by the full-length DNA corresponding to the DNA comprising the nucleotide sequence of any of SEQ ID NOs: 1, 2, and 7; And / or a DNA having an added amino acid sequence and encoding a protein functionally equivalent to the protein encoded by the full-length DNA.
( e ) 配列番号: 3〜 6のいずれかに記載の塩基配列からなる DNAとストリン ジェントな条件下でハイプリダイズし、 配列番号: 8〜: L 1に記載のアミノ酸配 列からなる蛋白質と機能的に同等な蛋白質をコ一ドする MA。  (e) hybridizes with a DNA consisting of the nucleotide sequence of any of SEQ ID NOs: 3 to 6 under stringent conditions, and functions as a protein comprising the amino acid sequence of SEQ ID NOs: 8 to L1. MA that encodes proteins that are equivalent to each other.
( f ) 配列番号: 1、 2、 7のいずれかに記載の塩基配列からなる DNAとスト リンジヱントな条件下でハイブリダイズし、 これら DNAに対応する全長 DNAに よりコードされる蛋白質と機能的に同等な蛋白質をコ一ドする DNA。  (f) hybridizes with a DNA consisting of the nucleotide sequence of any one of SEQ ID NOs: 1, 2, and 7 under stringent conditions, and functionally interacts with a protein encoded by a full-length DNA corresponding to the DNA. DNA that codes for an equivalent protein.
2 . 配列番号: 8〜1 1のいずれかに記載のアミノ酸配列からなる蛋白質の部 分ぺプチドをコードする DNA。  2. A DNA encoding a partial peptide of a protein consisting of the amino acid sequence of any one of SEQ ID NOs: 8 to 11.
3 . 請求項 1または 2に記載の DNAによりコードされる蛋白質またはぺプチ  3. A protein or peptide encoded by the DNA according to claim 1 or 2.
4 . 請求項 1または 2に記載の DNAが挿入されたべクタ一。 4. A vector into which the DNA according to claim 1 or 2 has been inserted.
5 . 請求項 1または 2に記載の DNAまたは請求項 4に記載のベクターを保持 する宿主細胞。 5. A host cell carrying the DNA according to claim 1 or 2 or the vector according to claim 4.
6 . 請求項 5に記載の宿主細胞を培養し、 該宿主細胞またはその培養上清から 発現させた蛋白質を回収する工程を含む、 請求項 3に記載の蛋白質またはべプチ ドの製造方法。  6. The method for producing a protein or peptide according to claim 3, comprising a step of culturing the host cell according to claim 5, and recovering the expressed protein from the host cell or a culture supernatant thereof.
7 . 請求項 3に記載の蛋白質に結合する抗体。  7. An antibody that binds to the protein of claim 3.
8 . 配列番号: 1〜7のいずれかに記載の塩基配列からなる DNAまたはその 相補鎖に相補的な少なくとも 15ヌクレオチドを含むポリヌクレオチド。  8. SEQ ID NO: A polynucleotide comprising at least 15 nucleotides complementary to a DNA consisting of the nucleotide sequence of any one of 1 to 7 or a complementary strand thereof.
9 . 請求項 3に記載の蛋白質に結合する化合物のスクリーニング方法であって  9. A method for screening a compound that binds to the protein according to claim 3,
( a ) 該蛋白質またはその部分べプチドに被検試料を接触させる工程、 (a) contacting a test sample with the protein or a partial peptide thereof,
( b ) 該蛋白質またはその部分べプチドと被検試料との結合活性を検出する工程  (b) a step of detecting the binding activity between the protein or its partial peptide and a test sample
( c ) 該蛋白質またはその部分べプチドに結合する活性を有する化合物を選択す る工程、 を含む方法。 (c) selecting a compound having an activity of binding to the protein or a partial peptide thereof.
PCT/JP2001/001863 2000-03-10 2001-03-09 Mouse adipocyte-origin genes WO2001066720A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2001241080A AU2001241080A1 (en) 2000-03-10 2001-03-09 Mouse adipocyte-origin genes

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2000-72502 2000-03-10
JP2000072502 2000-03-10

Publications (1)

Publication Number Publication Date
WO2001066720A1 true WO2001066720A1 (en) 2001-09-13

Family

ID=18590896

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2001/001863 WO2001066720A1 (en) 2000-03-10 2001-03-09 Mouse adipocyte-origin genes

Country Status (2)

Country Link
AU (1) AU2001241080A1 (en)
WO (1) WO2001066720A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003018805A1 (en) * 2001-08-23 2003-03-06 Japan Science And Technology Agency Hematopoietic stem cell proliferation regulators and polynucleotides encoding the same
EP1703917A2 (en) * 2003-12-22 2006-09-27 Amgen, Inc. Heh4 molecules and uses thereof
US7632935B2 (en) 2002-10-30 2009-12-15 Chungai Seiyaku Kabushiki Kaisha DNA encoding a mast cell-derived membrane protein

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000012708A2 (en) * 1998-09-01 2000-03-09 Genentech, Inc. Further pro polypeptides and sequences thereof
WO2000021986A2 (en) * 1998-10-09 2000-04-20 Incyte Pharmaceuticals, Inc. Matrix-remodeling genes
WO2000053750A1 (en) * 1999-03-08 2000-09-14 Genentech, Inc. Compositions and methods for the treatment of tumors
WO2000063438A2 (en) * 1999-04-20 2000-10-26 Curagen Corporation Method of classifying a thyroid carcinoma using differential gene expression

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000012708A2 (en) * 1998-09-01 2000-03-09 Genentech, Inc. Further pro polypeptides and sequences thereof
WO2000021986A2 (en) * 1998-10-09 2000-04-20 Incyte Pharmaceuticals, Inc. Matrix-remodeling genes
WO2000053750A1 (en) * 1999-03-08 2000-09-14 Genentech, Inc. Compositions and methods for the treatment of tumors
WO2000063438A2 (en) * 1999-04-20 2000-10-26 Curagen Corporation Method of classifying a thyroid carcinoma using differential gene expression

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003018805A1 (en) * 2001-08-23 2003-03-06 Japan Science And Technology Agency Hematopoietic stem cell proliferation regulators and polynucleotides encoding the same
US7262026B2 (en) 2001-08-23 2007-08-28 Japan Science And Technology Agency Hematopoietic stem cell proliferation regulators and polynucleotides encoding the same
US7632935B2 (en) 2002-10-30 2009-12-15 Chungai Seiyaku Kabushiki Kaisha DNA encoding a mast cell-derived membrane protein
EP1703917A2 (en) * 2003-12-22 2006-09-27 Amgen, Inc. Heh4 molecules and uses thereof
AU2004308908B2 (en) * 2003-12-22 2009-02-26 Amgen Inc. HEH4 molecules and uses thereof

Also Published As

Publication number Publication date
AU2001241080A1 (en) 2001-09-17

Similar Documents

Publication Publication Date Title
JP2020036596A (en) Novel hemopoietin receptor protein, NR10
DE60025241T2 (en) HEMOPOIETIN RECEPTOR PROTEIN, NR12
US7829083B2 (en) Antibodies to brain specific membrane protein
WO2002077230A1 (en) Nr10 splicing variants
WO2001009315A1 (en) Novel gene participating in the maintenance of smooth muscle cell differentiation
JP4838961B2 (en) Transporter genes OATP-B, C, D, and E
US20090197317A1 (en) TSG-Like Gene
US20090275082A1 (en) Mast Cell-Derived Membrane Proteins
WO2001066720A1 (en) Mouse adipocyte-origin genes
US20070105143A1 (en) Novel bHLH type transcription factor genes
EP1120426A1 (en) Novel g protein-coupled receptors
JP4632953B2 (en) Protein expressed in NK cells
JP4590107B2 (en) New fetal gene
JP2008099690A (en) Novel hemopoietin receptor protein, nr12
US7794973B2 (en) YS68 gene involved in primitive hematopoiesis
WO2001012808A1 (en) Novel frizzled family genes 584
JP2002000279A (en) Ys 68 gene participating in primary hemopoiesis
WO2001083738A1 (en) Sex determinative differentiation regulatory factor
EP1260584A1 (en) Novel protein having signal sequence
JP2004267003A (en) New gene present in human leukocyte antigen region

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

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
ENP Entry into the national phase

Ref country code: JP

Ref document number: 2001 565876

Kind code of ref document: A

Format of ref document f/p: F

122 Ep: pct application non-entry in european phase