WO2006098304A1 - Novel secretory protein derived from pituitary cell and use of the same - Google Patents

Novel secretory protein derived from pituitary cell and use of the same Download PDF

Info

Publication number
WO2006098304A1
WO2006098304A1 PCT/JP2006/304969 JP2006304969W WO2006098304A1 WO 2006098304 A1 WO2006098304 A1 WO 2006098304A1 JP 2006304969 W JP2006304969 W JP 2006304969W WO 2006098304 A1 WO2006098304 A1 WO 2006098304A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
polypeptide
amino acid
polynucleotide
acid sequence
Prior art date
Application number
PCT/JP2006/304969
Other languages
French (fr)
Japanese (ja)
Inventor
Kinji Inoue
Original Assignee
Saitama University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Saitama University filed Critical Saitama University
Priority to JP2007508144A priority Critical patent/JPWO2006098304A1/en
Publication of WO2006098304A1 publication Critical patent/WO2006098304A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/711Natural deoxyribonucleic acids, i.e. containing only 2'-deoxyriboses attached to adenine, guanine, cytosine or thymine and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/06Drugs for disorders of the endocrine system of the anterior pituitary hormones, e.g. TSH, ACTH, FSH, LH, PRL, GH
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/02Non-specific cardiovascular stimulants, e.g. drugs for syncope, antihypotensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy

Definitions

  • the present invention relates to a composition for promoting cell growth, maintaining survival, or suppressing tumor growth. More specifically, the present invention relates to a composition comprising a polypeptide involved in promoting cell growth and maintaining survival, a nucleotide encoding the polypeptide, an antagonist to the polypeptide, or the like.
  • Control of cell proliferation and maintenance of survival are considered to be extremely important mechanisms for maintaining biological order. For example, if cell growth control is not fully functioning and disordered cell growth can occur, it can cause serious diseases such as cancer, while cells are properly grown or maintained. If not, the onset of a disease related to the developmental disorder of the in vivo organ or the living body itself is expected.
  • cancer remains the leading cause of death due to disease, and the development of drugs and treatment methods for the effective treatment of cancer is the most important in the medical and biological fields. It is a research subject.
  • the pituitary gland is one of the organs deeply involved in functions such as growth, reproduction, and immunity.
  • the pituitary gland is an organ that secretes hormones. Factors expressed and functioning in this organ are expected to play an important role in the control of biological growth. In view of the function of the pituitary gland at the cellular level, the pituitary gland seems to play some regulatory role in the growth, proliferation, and maintenance of cells that form various biological organs.
  • Patent Document 3 it has been reported that the gene expression rate in gastric cancer is high (Patent Document 3), and more recently, the cgrl l gene has been identified as a gene suggested to be related to pain (Patent Document). 4) There is no unified view of the actual function of cgrl l, and the function has not yet been elucidated. In addition, it depends on what form the protein encoded by cgrl l functions, ie, the ability to function as it is translated into protein, the ability to function as a secreted protein, or the sugar chain, etc. There is a lack of information that is important for understanding functional mechanisms in vivo, such as whether post-translational modifications are performed.
  • Non-Patent Document 1 Madden et al. Cancer Res. 56; 5384_5390 1996
  • Patent Document 1 International Publication WO99 / 01581
  • Patent Document 2 International Publication WO 97/45542
  • Patent Document 3 International Publication WO2005 / 010213
  • Patent Document 4 International Publication WO03 / 016475
  • an object of the present invention is to provide a composition for promoting cell growth or maintaining survival, and a method for treating a related disease.
  • an object of the present invention is to provide a composition for inhibiting cell growth and a method for treating a related disease.
  • the present invention also relates to a diagnostic composition for determining the presence or absence of onset of cancer, hypertension, etc.
  • the purpose is to provide products and diagnostic methods.
  • Another object of the present invention is to provide a cglrl-encoded protein that functions in an actual living body.
  • the present invention relates to a cgrl gene product as a factor that functions in the pituitary gland that plays an important role in controlling the growth of a living body, and is considered to be deeply involved in cell growth control. This is based on the finding. Specifically, focusing on differences in protein expression between pituitary cells with different hormone-producing ability, we searched for factors that are only expressed in hormone-producing cells, and identified cgrl l as one of them. did. The inventor clarified a result completely different from the conventional finding that the cgrl l gene product is involved in cell suppression downstream of p53, and focused on the novel activity of the cgrl l protein to solve this problem. It came to do.
  • the present invention relates to the following (1) to (34).
  • a first aspect of the present invention is a cell proliferation promoting or comprising the polypeptide shown in the following (a) or (b), or an agonist thereof, and a pharmaceutically acceptable carrier: A composition for maintaining survival.
  • the second aspect of the present invention is “promoting cell growth, comprising a recombinant vector comprising a polynucleotide shown in the following (a) or (b) and a pharmaceutically acceptable carrier: Or a composition for maintaining survival.
  • SEQ ID NO: 1 SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11
  • the third aspect of the present invention is “the composition according to (1) or (2) above, wherein the cells are vascular endothelial cells”.
  • the fourth aspect of the present invention is “the composition according to (1) or (2) above, wherein the cell is a cell forming a pituitary gland”.
  • the fifth aspect of the present invention is “the composition according to (1) or (2) above, wherein the cell is a neuronal cell or a chromaffin cell”.
  • the sixth aspect of the present invention is the above-mentioned (used in the treatment or prevention of a blood flow disorder-related disease (
  • the seventh aspect of the present invention is the above (1) or (1) used for the treatment or prevention of hypotension.
  • the eighth aspect of the present invention is the above-mentioned (used for the treatment or prevention of a developmental disorder-related disease (
  • a ninth aspect of the present invention is the above (1) or (9)
  • the tenth aspect of the present invention is “cell growth inhibition or survival comprising an antagonist to the polypeptide shown in the following (a) or (b) and a pharmaceutically acceptable carrier”: Composition for suppression.
  • the twelfth aspect of the present invention is “the composition according to (11) above, wherein the antibody specifically binds to a polypeptide consisting of the amino acid sequence represented by SEQ ID NO: 13 or 14”. It is.
  • the thirteenth aspect of the present invention includes "a recombinant vector containing a nucleic acid that inhibits the function of the polynucleotide shown in the following (a) or (b)” and a pharmaceutically acceptable carrier.
  • polynucleotide J which is a nucleotide and the polypeptide encoded by the polynucleotide has a cell growth promoting activity or a survival maintaining activity.
  • a fourteenth aspect of the present invention is “the composition according to (13) above, wherein the nucleic acid that inhibits the function is an antisense DNA or RNA against the cgrl l gene”.
  • the fifteenth aspect of the present invention is “the composition according to (10) or (13) above, wherein the cell is a vascular endothelial cell”.
  • a sixteenth aspect of the present invention is "the composition according to (10) or (13) above, wherein the cell is a cell forming a pituitary gland".
  • the seventeenth aspect of the present invention is the above (10) or (13
  • the eighteenth aspect of the present invention is “the pharmaceutical composition according to the above (10) or (13), which is used for the treatment or prevention of a developmental disorder-related disease”.
  • the nineteenth aspect of the present invention is “the pharmaceutical composition according to the above (10) or (13), which is used for the treatment or prevention of hypertension”.
  • a blood flow disorder-related disease, hypotension, developmental disorder-related disease, cancer comprising an antibody against the polypeptide shown in the following (a) or (b)”: High blood pressure Composition for diagnosis of presence or absence, prognosis, or grade of malignancy or neurological disease.
  • the twenty-first aspect of the present invention is the diagnostic composition according to the above (20), wherein the antibody specifically binds to a polypeptide consisting of the amino acid sequence represented by SEQ ID NO: 13 or 14. Things ".
  • the twenty-second aspect of the present invention is “an antagonist against the polypeptide shown in the following (a) or (b).
  • a twenty-third aspect of the present invention is “an antibody against a polypeptide shown in the following (a) or (b):
  • the 24th aspect of the present invention is “the antibody according to the above (23), which specifically binds to a polypeptide consisting of the amino acid sequence represented by SEQ ID NO: 13 or 14.”
  • a twenty-fifth aspect of the present invention is a method for treating a blood of a mammal comprising administering a therapeutically effective amount of a polypeptide shown in the following (a) or (b), or an agonist thereof: A method for the treatment or prevention of flow disorders-related diseases, hypotension, developmental disorder-related diseases or neurological diseases.
  • the twenty-sixth aspect of the present invention is a cancer, developmental disorder in mammals comprising administering a therapeutically effective amount of an antagonist to the polypeptide shown in the following (a) or (b): A method for treating or preventing harm-related diseases or hypertension.
  • a twenty-seventh aspect of the present invention is “the method according to (26) above, wherein the antagonist is an antibody”.
  • the twenty-eighth aspect of the present invention includes "administering a therapeutically effective amount of a recombinant vector comprising a nucleic acid that inhibits the function of the polynucleotide shown in the following (a) or (b)”
  • a method for treating or preventing cancer, developmental disorder-related disease or hypertension in mammals includes "administering a therapeutically effective amount of a recombinant vector comprising a nucleic acid that inhibits the function of the polynucleotide shown in the following (a) or (b)"
  • a twenty-ninth aspect of the present invention is “the method according to (28) above, wherein the nucleic acid that inhibits the function is an antisense DNA or RNA against the cgrl l gene”.
  • the thirtieth aspect of the present invention includes "quantitatively detecting the amount of the polypeptide shown in the following (a) or (b) in tissue cells or blood obtained from mammals" Obviously
  • Blood flow disorder-related diseases hypotension, developmental disorder-related diseases, cancer, hypertension, heart disease or neurological disease diagnostic method.
  • a thirty-first aspect of the present invention is “a method for producing an immortalized cell by introducing a recombinant vector containing a polynucleotide shown in the following (a) or (b) into a cell:
  • polynucleotide that hybridizes with a complementary strand of a polynucleotide comprising the nucleotide sequence represented by SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11 under highly stringent conditions
  • SEQ ID NO: 1 SEQ ID NO: 3
  • SEQ ID NO: 5 SEQ ID NO: 7
  • SEQ ID NO: 9 SEQ ID NO: 11 under highly stringent conditions
  • a polynucleotide in which the polypeptide encoded by the polynucleotide has a cell growth-promoting activity or survival-sustaining activity.
  • a thirty-second aspect of the present invention is “the cell obtained by the method according to (31) above”.
  • a thirty-third aspect of the present invention is “a polypeptide shown in the following (a) or (b).
  • polypeptide comprising the amino acid sequence represented by SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12
  • polypeptide in the amino acid sequence represented by SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12, consisting of an amino acid sequence having one or several amino acid substitutions, deletions or insertions, and cell growth promoting activity or A polypeptide having a life-sustaining activity ".
  • a thirty-fourth aspect of the present invention is “the polynucleotide shown in the following (a) or (b).
  • polynucleotide that hybridizes with a complementary strand of a polynucleotide consisting of the nucleotide sequence represented by SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11 under highly stringent conditions, and encoded by the polynucleotide
  • SEQ ID NO: 7 the nucleotide sequence represented by SEQ ID NO: 7
  • SEQ ID NO: 9 the nucleotide sequence represented by SEQ ID NO: 9 or SEQ ID NO: 11 under highly stringent conditions
  • composition according to the present invention it is possible to develop a pharmaceutical effective for the prevention and / or treatment of cancer.
  • composition according to the present invention By using the composition according to the present invention, it becomes possible to develop a pharmaceutical effective for the prevention and / or treatment of various diseases related to blood flow disorders.
  • composition according to the present invention By using the composition according to the present invention, it becomes possible to develop a pharmaceutical effective for the prevention and Z or treatment of hypertension, hypotension and the like.
  • composition according to the present invention By using the composition according to the present invention, it becomes possible to develop a pharmaceutical effective for prevention and Z or treatment of developmental disorders.
  • composition according to the present invention By using the composition according to the present invention, it becomes possible to develop a pharmaceutical effective for prevention and Z or treatment of neurological diseases and the like.
  • composition according to the present invention By using the composition according to the present invention, it is possible to contribute to the development of products used for industrial applications such as maintaining the survival of cultured cells, or to development in agricultural applications.
  • the composition according to the present invention includes means for detecting the cgrl gene or its product, which is strongly involved in the secretory function of various cells. For example, by measuring the concentration in the blood, it becomes possible to develop a product for examining the degree of aging and fatigue of a living body.
  • FIG. 1 Two-dimensional electrophoresis using a pituitary-derived cell line produced by hormone-producing cells (Mt T / S) but not by hormone-producing cells (MtTZEB-3) The result of identifying the quality (spot2) is shown.
  • FIG. 2 shows a mass spectrometric pattern of a tryptic peptide of Spot2. This spot was found to correspond to the cgrl 1 gene product.
  • FIG. 3 shows alignment of amino acid sequences of rat and human derived cgrl 1 proteins.
  • FIG.5 Alignment of amino acid sequence of cgrl 1 protein from foot (Rattus norvegicus), mouse (Mus musculus), human (Homo sapien s), and N-terminal amino acid analysis of secreted cgrl 1 protein Shows the actual signal sequence (underlined) as revealed by R2 and H2 represent rat and human antigenic sites, respectively.
  • FIG. 6 shows the position of a hypoxic response element found in the cgrl 1 promoter region.
  • A shows the HIF-1 binding consensus sequence
  • B shows the position of the hypoxia response element present in the promoter region of the cgrl 1 gene.
  • FIG. 7 Quantitative PCR was performed on pituitary-derived hormone-producing cells (MtTZS cells) and non-hormone-producing cells (MtT / Se cells, MtTZSM cells, MtT / E_B3 cells). The result of having measured is shown.
  • FIG. 8 shows the results of immunohistochemical staining using an antibody against cglrl protein.
  • A shows staining of MtTZS cells using anti-cgrl 1 antibody and B using anti-growth hormone antibody.
  • FIG. 9 Results of immunohistochemical staining using anti-cgrl 1 antibody on the pituitary gland (A), enlarged view of the anterior pituitary gland (B), large intestine (C), and outer follicle membrane of mature follicle (D) Indicates.
  • PN, PI, and PD represent the posterior pituitary, the middle pituitary, and the anterior pituitary, respectively.
  • FIG. 10 shows a vector construct for cgrl l gene transfer.
  • FIG. ll shows the result of immunostaining using the anti-egrl protein antibody after introducing the cgrl gene into MtT / E-B3 cells (A) and MtT / Se (C). In both cases, a positive reaction was observed in the Golgi apparatus. In contrast, no positive reaction was observed in MtT / E_B3 cells (B) and MtT / Se cells (D) into which only the vector was introduced.
  • Fig. 12 Shows the results of Western blotting using anti-cgrl protein antibodies on cell extracts, culture supernatants, and cell extracts of MtT / S cells forcibly expressing cgrl 1 protein.
  • FIG. 13 Shows the results of stamp stamping using anti-cgrl 1 protein antibody using cgrl l protein non-expressing cells (lane 1) and cgrl l protein expressing cells (lane 2) as samples. . M: Marker
  • FIG. 3 is a diagram showing that a blood vessel-rich tumor was formed at the transplantation site.
  • FIG. 16 shows a graph comparing the growth rates of cells in which the cglrl expression vector was forcibly expressed (MtT / E-B3-So) and cells into which only the vector was introduced (MtT / E-B3-Ve).
  • cgrl l forced expression PC12 cells (clone C to G) and cgrl l not forcibly expressed PC12 cells (control) were cultured in serum-free medium, and the number of viable cells on day 8 was compared. The graph is shown.
  • FIG. 19 Viable cells cultured in MtT / Se cells (cglrl non-expressing, estrogen-dependent growth) with medium containing CGR11 (serum-free), medium containing serum, and medium (serum-free) Is a graph of the number of.
  • FIG. 20 shows changes in blood pressure after injecting purified secreted cgrl into the abdominal cavity of rats. The arrow indicates the time when cgrl l protein was injected.
  • the blood pressure is shown as an average value of dilated blood pressure and systolic blood pressure.
  • the cgrl polynucleotide which is one of the active ingredients of the composition of the present invention includes not only DNA consisting of the base sequence represented by SEQ ID NO: 1, 3, 5, 7, 9 or 11, but also SEQ ID NO: 1, It hybridizes under high stringent conditions with DNA consisting of a base sequence complementary to DNA consisting of the base sequence represented by 3, 5, 7, 9 or 11 and has cell growth promoting activity or survival maintenance activity. Also included are those consisting of DNA encoding a polypeptide.
  • the “cell” may be any cell as long as the cgrl polypeptide can act, for example, animal cell, preferably human, rat, mouse, urushi, horse, hidge. , Cells from chickens, birds, cats, etc.
  • the polynucleotide of the present invention also includes the “gene” of the present invention.
  • the gene of the present invention includes cDNA and genomic DNA, and includes not only exons and introns of the gene, but also transcriptional regulatory regions such as a promoter and an enzyme.
  • the polynucleotide of the present invention includes DNA and RNA, and may be double-stranded or single-stranded. In the case of double-stranded, double-stranded DNA, double-stranded RNA or DNA and It may be a hybrid with RNA.
  • DNA that can hybridize with the polynucleotide containing the nucleotide sequence represented by SEQ ID NO: 1, 3, 5, 7, 9, or 11 under stringent conditions includes SEQ ID NO: 1, 3, 5, 7, 9, or Preferably about 70% or more, more preferably about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90% , 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, most preferably about 99% of polynucleotides that contain a homologous nucleotide sequence. It is done.
  • the stringent condition is a hybridization condition that is easily determined by those skilled in the art, and is generally an empirical calculation that depends on the probe length, the washing temperature, and the salt concentration. In general, the longer the probe, the higher the temperature for proper annealing. The temperature decreases as the temperature increases and the probe becomes shorter. However, hybrid formation generally depends on the ability of the denatured DNA to reanneal when the complementary strand is present in an environment near its melting point but below it.
  • the washing of the post-hybridization of the final letter should be performed at a temperature of 37 ° C to 42 ° C, 0.1 X
  • a temperature of 37 ° C to 42 ° C 0.1 X
  • washing in SSC 0.1% SDS solution.
  • highly stringent conditions include washing in 65 ° C, 5 X SSC and 0.1% SDS in the washing step.
  • the cgrl polynucleotide can be obtained from, for example, a cell derived from the pituitary gland and producing hormone. For example, extract all proteins present in hormone-producing cells and non-hormone-producing cells as controls, and perform two-dimensional electrophoresis, etc., and compare their migration profiles, and only in hormone-producing cells. Amino acid sequence information is obtained from confirmed protein spots using mass spectrometry. Next, based on the obtained amino acid sequence information, DNA having a cgrl polynucleotide sequence can be isolated from an appropriate cDNA library or the like.
  • sequence information of a known cgrl l gene from a database or the like and obtain a cgrl l gene derived from a desired tissue or animal species based on the sequence information.
  • Cloning of the cgrl gene can be carried out based on ordinary knowledge in the art.
  • the gene can be obtained by preparing a cDNA library from cells expressing the gene and using a conventional screening method.
  • RNA is prepared from cells expressing the gene
  • cDNA is synthesized by reverse transcriptase
  • PCR primers are prepared based on the gene sequence to amplify the cDNA.
  • the cDNA may be obtained.
  • the cell that can be used for obtaining the cgrl polynucleotide in the present invention is any cell that can express any mRNA that expresses cgrl polypeptide mRNA.
  • Pituitary hormone-producing cells are preferred.
  • the movement from which the cells are derived The product may be any animal (for example, human, rat, etc.) as long as it has the gene of the present invention.
  • cgrl l To obtain mRNA for obtaining polynucleotides and cDNA, use commercially available kits such as mRNA Purification Kit (Pnarmasia) and AMV Reverse Transcriptase First-strand cDNA Synthesis Kit (Seikagaku Corporation). Use it.
  • the cgrl polypeptide in the present invention is a polypeptide comprising the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 2, 4, 6, 8, 10 or 12.
  • the “polypeptide containing substantially the same amino acid sequence” means about 60% or more, preferably about 70% or more, of the amino acid sequence represented by SEQ ID NO: 2, 4, 6, 8, 10, or 12. More preferably, about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94% 95%, 96%, 97%, 98%, most preferably a polypeptide having an amino acid sequence having about 99% amino acid identity and having cell growth promotion or activity.
  • the “cell” may be any cell as long as it can act on cgrl polypeptide, for example, animal cell, preferably human, rat, mouse, urushi, horse, It is a cell derived from Hedge, chicken, dog, cat, etc.
  • polypeptide and “protein” are used in the same meaning unless otherwise specified.
  • polypeptide comprising an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 2, 4, 6, 8, 10, or 12, SEQ ID NO: 2, 4, 6, 8, 10 Or one or several (preferably:! To about 30, more preferably about 1 to 10, more preferably :! to 5) amino acids in the amino acid sequence represented by 12 are deleted, It is a polypeptide having an amino acid sequence substituted or added and having cell growth promotion or activity.
  • the “cell” may be any cell as long as the cglrl polypeptide can act, for example, an animal cell, preferably a human, rat, mouse, urushi, horse, hidge. It is a cell derived from nitrite, nu, cat, etc.
  • amino acid deletions, additions and substitutions are present in isolated natural polypeptides.
  • the substitution of a specific amino acid residue is a known method such as Guppedduplex method or Kunkel method using a commercially available kit (for example, MutanTM-G (TAKARA), MutanTM-K (TAKARA)).
  • TAKARA MutanTM-G
  • TAKARA MutanTM-K
  • the C-terminus of the cgrl polypeptide in the present invention is usually a carboxyl group (_C0OH) or a carboxylate (_C00_), and the carboxyl group is an amide (one CONH2). Or may be chemically modified with ester (_C0OR).
  • C1-6 alkyl group for example, methinole, ethyl, n-propyl, isopropyl, n-butyl
  • C3-8 cycloalkyl group for example, cyclopentyl, cyclohexylene
  • C1-6 aryl group for example, phenylolene, mononaphthyl
  • phenyl-1-C1-2anolyl group for example, benzyl, phenethyl
  • ⁇ -naphthyl-C1-2 alkyl group for example, a naphthylmethyl
  • the polypeptide of the present invention has a carboxyl group in the polypeptide chain other than at the C-terminus, the polypeptide of the present invention includes those in which the carboxyl group is amidated or esterified. Examples of the ester in this case include the above-mentioned esters.
  • the N-terminus of the polypeptide of the present invention is usually an amino group (one NH2), but the amino group may be chemically modified with a C16 acyl group such as a formyl group or a acetyl group. .
  • the daltamyl group generated by cleaving the N-terminal side in vivo is pyrologretamine oxidized, or a substituent on the side chain of an amino acid in the molecule (for example, OH, —SH, amino group, imidazole group, indole group, Polypeptides of the present invention include those in which a guanidino group or the like) is chemically modified with an appropriate functional group (for example, formyl group, acetyl), or a sugar chain.
  • a peptide containing a partial amino acid sequence in the cglrl polypeptide of the present invention may be included in the composition of the present invention.
  • the partial peptide can also be an antigen for preparing the antibody of the present invention. That is, the partial peptide of the present invention is the same or substantially the same as the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4 or SEQ ID NO: 6. As long as it contains a part of the amino acid sequence of the same amino acid sequence.
  • the number of amino acids constituting the partial peptide of the present invention is at least 10 or more, preferably 30 or more, more preferably 80 or more.
  • the partial peptide of the present invention may be mechanically modified such that the C-terminus is a carboxyl group (_COH) and the N-terminus is an amino group (_NH2).
  • the cgrl l polypeptide or a salt thereof in the present invention is obtained from a cultured cell or tissue derived from a human animal (eg, rat, mouse, etc.) expressing the cgrl l polypeptide by a conventional technique in the art. Extraction and isolation from the culture can be performed by culturing a transformant that can be extracted or isolated, or can be expressed in a state in which the DNA encoding the polypeptide of the present invention can be expressed as described later. Can also be prepared. When preparing from human tissues or cells, homogenize human tissues or cells and extract with acid, etc., and use the resulting extract for hydrophobic chromatography, reverse phase chromatography, ion exchange chromatography, etc. Isolation and purification can be achieved by combining these various chromatographies.
  • the partial peptide or its salt in the present invention is produced by a known peptide synthesis method or cgrrl polypeptide cleaved with an appropriate peptidase (eg, trypsin, chymotrypsin, arginyl endopeptidase). be able to.
  • an appropriate peptidase eg, trypsin, chymotrypsin, arginyl endopeptidase.
  • the peptide synthesis method for example, either a solid phase synthesis method or a liquid phase synthesis method may be used. That is, the desired peptide is produced by condensing the partial peptide or amino acid that can constitute the polypeptide of the present invention and the remaining part, and removing the protecting group when the product has a protecting group.
  • the partial peptide of the present invention can be isolated and purified by combining ordinary purification methods such as solvent extraction, distillation, column chromatography, high performance liquid chromatography, recrystallization and the like.
  • cgrl polypeptide obtained by the above method or a partial peptide thereof is a free form
  • it can be converted to an appropriate salt by a known method.
  • the free form is obtained by a known method.
  • a recombinant vector incorporating the cgrl 1 polynucleotide or a part thereof can be obtained by ligating the cgrl polynucleotide or a part thereof to an appropriate vector.
  • the recombinant vector is not particularly limited as long as it can be replicated in a host when it is used for cloning.
  • a vector for expressing cgrl 1 polypeptide or a part thereof a vector capable of replicating in a host and having a promoter capable of expressing a DNA fragment encoding the polypeptide, etc. Can be used.
  • Plasmid DNA includes plasmids derived from E. coli (eg, pBR322, pBR325, pUC118, pUC119, pUC18, pUC19, pCBD-C, etc.), plasmids derived from Bacillus subtilis (eg, pUB110, pTP5, pC194, etc.), and yeast-derived plasmids (eg, ⁇ 13, ⁇ 24, YCp50, YIp30, etc.), and phage DNA includes ⁇ phage.
  • animal viruses such as retrovirus and vaccinia virus, and insect virus vectors such as baculovirus and toga virus can also be used.
  • the promoter used in the present invention is not particularly limited as long as it is an appropriate promoter corresponding to the host used for gene expression.
  • SRa promoter when an animal cell is used as a host, SRa promoter, CMV promoter, SV40 promoter, LTR promoter, HSV-TK promoter, EF-1 ⁇ promoter and the like can be mentioned.
  • the host is Escherichia coli, tac promoter, ⁇ ⁇ ⁇ ⁇ promoter, lac promoter, recA promoter, e.g. PL promoter, lpp promoter, etc. If the host is Bacillus subtilis, SPOl promoter, SP02 promoter, penP promoter Etc.
  • PH05 promoter When the host is yeast, PH05 promoter, PGK promoter, GAP promoter, ADH promoter and the like can be mentioned.
  • polyhedrin promoter When the host is an insect cell, polyhedrin promoter, P10 promoter, etc. are preferable.
  • the recombinant vector of the present invention includes a cgrl 1 polypeptide or a portion of a coding cage 1J,
  • a selection marker, terminator, enhancer, splicing signal, poly A addition signal, ribosome binding sequence (SD sequence), SV40 origin of replication (SV40ori), etc. can be linked.
  • Selection markers include, but are not limited to, hygromycin resistance marker (Hygr), dihydrofolate reductase gene (dMr), ampicillin resistance gene (Ampr), kanamycin resistance gene (Kanr), neomycin resistance gene (Neor, G418) Etc. are available.
  • an appropriate signal sequence may be added to the N-terminal side of the polypeptide of the present invention.
  • Inserting a cgrl polynucleotide or a part thereof into the above-described vector means that the cloned DNA is digested with a restriction enzyme as it is or as desired, and a linker is added to the vector DNA. It can be performed by inserting into a restriction enzyme site or a multicloning site.
  • the DNA to be ligated may have ATG as a translation initiation codon on the 5 ′ end side and TAA, TGA or TAG as a translation termination codon on the 3 ′ end side. These translation initiation codons and translation termination codons can also be added using an appropriate synthetic DNA adapter.
  • the DNA to be ligated needs to be incorporated into a vector so that the polypeptide of the present invention encoded in the DNA is expressed in the host cell.
  • a transformant of cgrl can be obtained by introducing the recombinant expression vector of the present invention into a host so that the cgrl l gene can be expressed.
  • the host is not particularly limited as long as it can express the cgrl 1 gene.
  • Escherichia coli and other genus Escherichia, Bacillus subtilis and other Bacillus genus, Pseudomonas putida and other shyudomonas genus, Rhizobium meliloti and other lysovivum genus Bacteria belonging to, Saccharomyces cerevisiae, Szozoaccharomyces pombe, Pichia pastoris, monkey cells COS _ 7, Vero, Chinese hamster ovary cells (CHO cells) Or insect cells such as Sf 9 and Sf 21.
  • a method for introducing a recombinant vector into Escherichia coli a method using calcium ions (Cohen et al., 1972), an electo mouth position method (Shigekawa and Dower, 1988) and the like can be used.
  • a method for introducing a recombinant vector into yeast the Elect Mouth Position method (B ecker et al., 1990), the Suguchi Higuchi plast method (Hinnen et al., 1978), the lithium acetate method (Itoh et al., 1983), etc. can be used. .
  • Methods for introducing recombinant vectors into animal cells or animal cells include the DEAE dextran method (Lopata et al., 1984), the electopore position method, the phosphate phosphate method (Chen and Okayama, 1988), and the method using cationic lipids. (Elroy-Stein and Moss, 1990).
  • the medium can be any medium that is suitable for the desired cells to normally survive, for example, about 5 to 20%. MEM medium, DMEM medium, RPMI-1640 medium, etc. containing fetal bovine serum are used.
  • conditions suitable for the desired cells can be set for pH and culture temperature, and these conditions can be easily set by those skilled in the art. As described above, if the cgrl polypeptide is expressed in a cell in which immortalization or survival is desired, Cell immortalization or viability maintenance is achieved.
  • the cgrl l polypeptide or a partial peptide thereof contained in the composition of the present invention is obtained by culturing a transformant introduced with an expression vector of the cgrl l gene or a part thereof, and cgrl 1 polypeptide or a part thereof. Can be produced by isolating the polypeptide or a part thereof from the culture. “Culture” means any of culture supernatant, cultured cells or cultured cells, or disrupted cells or cells.
  • the medium contains a carbon source, a nitrogen source, inorganic salts and the like that can be assimilated by the microorganism. Either natural or synthetic media can be used as long as the culture can be carried out efficiently.
  • a carbon source carbohydrates such as glucose, fructose, sucrose and starch, organic acids such as acetic acid and propionic acid, and alcohols such as ethanol and propanol are used.
  • nitrogen sources include ammonia, ammonium chloride, ammonium sulfate, ammonium acetate, ammonium salts of organic acids such as ammonium salts, or other nitrogen-containing compounds, peptone, meat extract, corn steep liquor, etc. Used.
  • inorganic salts include monopotassium phosphate, dipotassium phosphate, magnesium phosphate, magnesium sulfate, sodium chloride, ferrous sulfate, manganese sulfate, copper sulfate, and calcium carbonate.
  • the culture is performed under conditions suitable for the host cell.
  • a medium for culturing Escherichia coli LB medium, M9 medium and the like are preferable. Agents such as isopropyl-1-thio ⁇ D galactoside, 3-indolylacrylic acid can be added to make the promoter work efficiently if desired.
  • the culture is usually carried out at about 15 to 37 ° C for about 3 to 24 hours, and if necessary, aeration and agitation can be controlled.
  • cultivation is usually performed at about 30-40 ° C for about 6-24 hours, and if necessary, aeration or agitation is added.
  • Examples of the medium for culturing yeast include SD medium and YPD medium. It is preferable to adjust the pH of the medium to about 5-8. Incubate at about 20 to 35 ° C for about 24 to 72 hours, and if necessary, add aeration and agitation. Transformation where the host is an insect cell or insect When culturing the body, examples of the medium include Grace insect medium containing urine serum. The pH of the medium is preferably adjusted to about 6.2 to 6.4. Incubate at about 27 ° C for about 3 to 5 days, and add aeration or agitation as necessary.
  • a transformant whose host is an animal cell is cultured, for example, a MEM medium, DMEM medium, RPMI-1640 medium or the like containing about 5 to 20% urine fetal serum is used. .
  • the pH is preferably about 6-8.
  • the cells or cells are collected by a known method after culturing and suspended in an appropriate buffer solution.
  • a method of obtaining a crude extract of cgrl polypeptide or a partial peptide thereof by centrifugation or filtration after disrupting cells or cells by ultrasonic waves, lysozyme, Z, or freeze-thawing is appropriately used.
  • the buffer may contain a protein denaturant such as urea or guanidine hydrochloride, or a surfactant such as Triton X-100.
  • the cells or cells and the supernatant are separated by a method known per se, and the supernatant is collected.
  • Purification of the polypeptide of the present invention contained in the thus obtained culture supernatant or extract can be performed by appropriately combining known separation and purification methods.
  • known separation and purification methods methods using solubility such as salting-out solvent precipitation method, dialysis method, ultrafiltration method, gel filtration method, and mainly using differences in molecular weight such as SDS-PAGE.
  • Methods using charge differences such as ion exchange chromatography, methods using specific affinity such as affinity mouthmatography, and differences in hydrophobicity such as reversed-phase high-performance liquid chromatography
  • a method using a difference in isoelectric point such as a method or an isoelectric focusing method may be used.
  • Nucleic acids that inhibit the function of the cgrl l gene may inhibit the growth or survival of cells that function as the cgrl l gene is expressed by losing the function of the cgrl l gene. It becomes possible. As a result, for example, it is possible to suppress the growth of cells that have become cancerous or tumorous due to, for example, disordered overexpression of cglrl, or cells that may become cancerous or tumorous.
  • Examples of nucleic acids that inhibit gene functions include single-stranded nucleic acids such as antisense RNA or DNA and derivatives thereof, short-length or double-stranded RNA having a sequence complementary to a part of the gene region, etc. Can be mentioned.
  • Antisense acts as an effective inhibitor for the expression of the cgrl gene, which can be RNA or DNA or derivatives thereof.
  • Antisense RNAs are designed, for example, to hybridize with mRNA in vivo and inhibit translation from mRNA to cgrl polypeptide (Okano et al., 1991).
  • the DNA oligonucleotide is designed to be complementary to the transcription initiation region of the cglrl gene, for example, and as a result, inhibits the expression of the gene (Cohen, 1989).
  • antisense RNAs or DNAs can be introduced into cells so that they can function in vivo to inhibit the expression of cglrl genes or polypeptides.
  • antisense DNA for example, an oligonucleotide that binds to a position between about 10 and +10 of the target gene sequence is desirable.
  • RNAi is a phenomenon in which the target mRNA (for example, mRNA of the cglrl gene) is degraded by double-stranded RNA having a sequence complementary to the target mRNA.
  • the target mRNA for example, mRNA of the cglrl gene
  • double-stranded RNA having a sequence complementary to the target mRNA By artificially introducing double-stranded RNA using this phenomenon, the expression of the target gene can be suppressed. Therefore, it is considered to be based on a principle different from the above-described antisense method.
  • An antibody against the cgrl 1 polypeptide is capable of inhibiting the growth or survival of cells in which the cgrl 1 polypeptide is expressed and functioning by losing the activity of the cgrl 1 polynucleotide. As a result, it is possible to suppress the proliferation of cells that have become cancerous or tumorous due to, for example, disordered overexpression of cglrl, or cells that may become cancerous or tumorous.
  • the presence or absence of cancer due to overexpression of cgrl l polypeptide, or prognosis can be made using an antibody against the cgrl polypeptide.
  • the presence or absence of canceration or the prognosis of cancer can be determined by measuring the concentration of cgrl polypeptide present in blood obtained from a specimen using the antibody.
  • Antibodies against cgrl l include antibodies that specifically bind to cgrl l polypeptide, and antibody fragments such as Fab or F (ab ') thereof.
  • the “antibody” in the present invention includes a monoepitope specific antibody against the cgrl polypeptide, Polyepotope specific antibodies, single chain antibodies, and fragments thereof are included. These antibodies include, for example, monoclonal antibodies, polyclonal antibodies, humanized antibodies and the like.
  • Polyclonal antibodies can be prepared, for example, by incubating a mixture of immunogen and adjuvant against a mammalian host animal. Usually, the immunogen and / or adjuvant is injected multiple times subcutaneously or intraperitoneally into the host animal.
  • Immunogens include polypeptides of the invention and fusions with heterologous polypeptides or fragments thereof. For example, a peptide consisting of the amino acid sequence represented by SEQ ID NO: 13 or 14 is a very good immunogen.
  • adjuvants include complete Freud and monophosphoryl lipid A synthesis trehalose dicorynomycolate (MPL-TDM).
  • MPL-TDM monophosphoryl lipid A synthesis trehalose dicorynomycolate
  • KLH keyhole limpet hemocyanin
  • serum albumin serum albumin
  • thythyroglobulin thythyroglobulin
  • soybean trypsin inhibitor are used to enhance the immune response. After binding the immunogen such as a protein having immunogenicity, it may be digitized.
  • Monoclonal antibodies can be prepared using the Hypridoma method (Milstein et al. Cuello, 1983).
  • This method includes the following four steps: (i) immunizing the host animal or lymphocytes derived from the host animal, (ii) a monoclonal antibody secreting (or potentially secreting) linker. Collect spheres, (iii) fuse lymphocytes to immortalized cells, (iv) select cells that secrete the desired monoclonal antibody.
  • Immunogens also include cgrl polypeptide and fusions with heterologous polypeptides or fragments thereof.
  • an immunogen for example, a peptide consisting of the amino acid sequence represented by SEQ ID NO: 13 or 14 is a very excellent immunogen.
  • lymphocytes obtained from the host animal are fused with an immortalized cell line using a fusion agent such as polyethylene glycol to establish hyperpridoma cells (Goding, 1
  • a cartilage or mouse myeloma cell line in which rodents, rabbits, or human myeloma cells immortalized by transformation are used is used.
  • the cells are grown in a suitable medium containing one or more substrates that inhibit the growth or survival of unfused lymphocytes and immortalized cell lines.
  • Conventional techniques use parental cells that lack the enzyme hypoxanthine guanine phosphoribosyltransferase (HGPRT or HPRT). In this case, hypoxanthine, aminopterin and thymidine are added to a medium (HAT medium) that inhibits the growth of HGPRT-deficient cells and allows the growth of hypridoma.
  • HGPRT hypoxanthine guanine phosphoribosyltransferase
  • a preferred immortal cell line is the mouse myeloma line, which is available from the American Type Culture Collection (Manassas, VA). See Kozbor et al., 1984; Schook, 1987 for the production of human monoclonal antibodies by human myeloma and mouse human heteromyeloma cell lines.
  • Monoclonal antibody-secreting hyperpridoma cells can be isolated as single clones by limiting dilution and subculture (Goding, 1996). Suitable media include Dulbecco's Modified Eagle Medium, RPMI-1640, and in some cases, protein-free or serum-free medium. Hypridoma cells may also be grown in the ascites of suitable host animals.
  • Monoclonal antibodies can be obtained from medium or ascites, et al. Protein A sepharose, hydoxyxpatite chromatography, gel electrophoresis, dialysis, ammonium sulfate precipitation or affinity chromatography (Harlow and Lane, 1988; Harlow and Lane, 1999). It is isolated and purified by methods well known to those skilled in the art.
  • Monoclonal antibodies can also be produced by gene recombination techniques (US Pat. No. 4166452, 1979).
  • To identify the gene encoding the desired monoclonal antibody polypeptide from the hyperidoma cell line secreting the antibody of interest for example, using oligonucleotide probes that specifically bind to the mouse heavy and light chain antibody genes. May be.
  • the target antibody gene can be identified by determining the distribution 1J of the genes.
  • the isolated DNA fragment is transferred to an appropriate expression vector and the antibody gene is not produced by any other immunoglobulin protein, simian COS-7 cells, Chinese hamster ovary Transfect into (CHO) cells or host cells such as myeloma cells.
  • Isolated DNA fragments can be obtained, for example, by replacing coding sequences for human heavy and light chain constant domains with homologous mouse sequences (US Pat. No. 4,816,567; 1989; Morrison et al., 1987) or non- Modifications can be made by fusing the immunoglobulin coding sequence with all or part of the sequence encoding the immunoglobulin polypeptide.
  • non-immunoglobulin polypeptides can be substituted for the constant domain of an antibody or can be substituted for the constant domain of a single antigen binding site to prepare a chimeric bivalent antibody.
  • 7-3 Humanized and human antibodies
  • cgrl polypeptide antibodies include human rabbits or human antibodies.
  • Humanized forms of non-human antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (Fv, Fab, Fab ′, F (ab ′)) or other antibody antigen-binding regions that contain minimal sequences derived from non-human immunoglobulin.
  • Etc
  • human rabbit antibodies have one or more amino acid residues introduced from non-human immunoglobulin. These non-human amino acid residues are often selected from variable domains.
  • Humanized antibodies can be produced, for example, by replacing mouse CDRs or CDR (complementarity determining region) sequences with corresponding human antibody sequences (Jones et al., 1986; Riechmann et al., 1988; Verhoeyen et al. , 1988).
  • a humanized antibody is a human antibody that is substituted with a residue in a specific human-derived CDR and a residue in a CDR of a non-human species such as mouse, rat, or rabbit corresponding to the residue. That is.
  • nonhuman-derived residues may replace Fv framework residues of human immunoglobulin (Jones et al., 1986; Presta, 1992; Riechmann et al., 1988).
  • the “agonist” in the present invention is a substance that specifically binds to cgrl polypeptide, its receptor, or interacting factor (binding partner) to promote the biological activity of the polypeptide of the present invention. And a part of the above-mentioned antibody is also included in the “agonist”. Examples of non-antibody antibodies include, but are not limited to, polypeptides or fragments thereof, nucleic acids, and other low molecular weight compounds.
  • the “antagonist” in the present invention means a substance that specifically binds to cglrl polypeptide, its receptor, or an interaction factor (binding partner) and suppresses the biological activity of the polypeptide of the present invention. However, some of the above-mentioned antibodies are also included in “antagonists”.
  • Abutama is a nucleic acid molecule or peptide capable of specifically binding to a target protein and can inhibit the activity of the target protein. In addition, even if the activity cannot be inhibited, it can be used to examine the presence or absence of the protein because it can bind to the target protein. Therefore, aptamers to cgrl l polypeptide can inhibit the growth or survival of functioning cells by expressing cgrl l polypeptide by losing the activity of cgrl l polypeptide. .
  • a disease caused by overexpression of cgrl l polypeptide for example, the presence or absence of onset of cancer, or prognosis can be determined using an aptamer to cgrl l polypeptide. For example, the presence or absence of canceration or the prognosis of cancer can be determined by measuring the concentration of cgrl polypeptide present in blood obtained from a specimen using the aptamer.
  • RNA or DNA specifically binds to the cglrl polypeptide.
  • it is DNA.
  • Aptamers also include those that have been modified in the phosphate skeleton to increase in vivo stability, and are not particularly limited, but include phosphorothioate bonds, phosphorodithioate bonds, and phosphoramidothioates. A phosphate skeleton containing a bond, a phosphoramidate bond, a phosphordiamidate bond, or a methylphosphonate bond is preferred.
  • the factor that interacts with the cgrl l polypeptide is expected to have a promoting or suppressing effect on the cell growth promotion or survival maintenance activity of the cgrl l polypeptide. Therefore, by further adding the factor to the composition of the present invention, it is possible to achieve the effect according to the purpose.
  • “interacting factors” include those that can be assumed by those skilled in the art, such as proteins, peptides, and small compounds.
  • proteins include receptors for eg rl l polypeptides.
  • the cgrl l polypeptide or a part thereof and a candidate factor eg, a receptor
  • a candidate factor eg, a receptor
  • a cell that presents a candidate factor on its surface eg, a candidate
  • a cell that expresses a receptor protein on its surface, etc. the binding amount of cgrl l polypeptide or a part thereof with a candidate factor, or the cell, etc.
  • the presence or absence of interaction can be determined by detecting biological changes in the body.
  • the cgrl l polypeptide or a part thereof, or a candidate factor is labeled (for example, Fluorescent labeling, radiolabeling), contacting the two, and using the intensity of the signal obtained from the label (for example, fluorescence, radioactivity, etc.) as an index, detect the amount of binding of both based on conventional methods and common technical knowledge As a result, if it is possible to evaluate that both are strongly bound, the candidate factor can be determined to be an interacting factor.
  • labeled for example, Fluorescent labeling, radiolabeling
  • a cell expressing a candidate receptor is contacted with a cgrl polypeptide or a part thereof, and a cell-specific intracellular signal via the receptor (for example, cell When changes in intracellular Ca 2+ release, intracellular cAMP production, cell membrane potential fluctuation, intracellular protein phosphorylation, etc.) are detected, it is predicted that the candidate receptor actually functions as a receptor It becomes possible.
  • a composition comprising a cgrl polynucleotide, a polypeptide, an agonist for the polypeptide, etc. can be used for the purpose of promoting cell growth or maintaining the survival of the polynucleotide.
  • a composition comprising an antibody against a peptide, an antagonist, etc. can be used for the purpose of suppressing abnormal cell growth (eg, tumorigenesis).
  • the composition can also be used in vitro for use in cultured cells and the like.
  • the present composition includes substances for maintaining isotonicity of cells, substances for promoting the transfer of the active ingredients into cells, and cuffers, etc. Auxiliary substances are included. Any of these auxiliary substances can be used if they are available to those skilled in the art.
  • a composition comprising a cgrl polynucleotide, a polypeptide, an agonist for the polypeptide, etc. can be used for the purpose of promoting cell growth or maintaining the survival of the polynucleotide.
  • a composition comprising an antibody against a peptide, an antagonist, etc. can be used as a pharmaceutical composition because it can be used for the purpose of suppressing abnormal cell growth (eg, tumorigenesis).
  • the pharmaceutical composition can be expected to be effective in treating a blood flow disorder-related disease if, for example, vascular endothelial cells are used as a target. Also, if the pituitary-forming cells are used as a target, the pharmaceutical composition is considered to be a growth disorder. It can be expected to be effective in treating related diseases, while tumors or cancerous diseases If cells that show normal growth are used as targets, an effect can be expected in the treatment or prevention of cancer.
  • composition comprising the cglrl polynucleotide according to the present invention, a polypeptide, an agonist for the polypeptide, etc. has an effect of increasing blood pressure, and also inhibits the activity of the polynucleotide.
  • a composition comprising an nucleic acid or an antibody against the polypeptide, an antagonist or the like has an effect of lowering blood pressure, and thus can be used as a pharmaceutical composition.
  • the pharmaceutical composition is expected to be effective as, for example, an antihypertensive agent, a hypotensive agent, a hypotensive therapeutic agent, or a hypertensive therapeutic agent.
  • blood flow disorder-related disease refers to a disease caused as a result of hindering blood circulation in the whole or a part of the body or a disease causing hindrance to blood circulation.
  • arteriosclerosis inflammatory vasculitis, aneurysm, thrombophlebitis, and kidney disease are all diseases within the scope of common technical knowledge of those skilled in the art.
  • hypertension and “hypotension” include related diseases caused by abnormal blood pressure, such as heart diseases (for example, myocardial infarction, aortic stenosis, cardiac hypertrophy, hypertrophic cardiomyopathy and heart failure). included.
  • heart diseases for example, myocardial infarction, aortic stenosis, cardiac hypertrophy, hypertrophic cardiomyopathy and heart failure.
  • Cancer refers to a disease associated with abnormal growth or proliferation of cells, and is within the scope of common technical knowledge of those skilled in the art, such as adenocarcinoma, lymphoma, blastoma, melanoma, sarcoma, and leukemia. It refers to all diseases.
  • Neurological disease is to be understood in a general sense and includes all diseases caused by the degeneration or damage of nerve cells or para-neurons, eg Parkinson's disease, Alzheimer's disease, Huntington All diseases within the scope of common technical knowledge of those skilled in the art, such as illness, amyotrophic lateral sclerosis, spinocerebellar degeneration
  • “Developmental disorder” is a condition in which some abnormality is observed in physical growth. Related diseases include, but are not limited to, dwarfism, giantism, etc. .
  • the above cgrl polynucleotide can be used as a therapeutic agent in the form of a pharmaceutical composition that does not adversely affect the living body.
  • a pharmaceutical composition that does not adversely affect the living body.
  • such compositions contain a nucleic acid component.
  • a pharmaceutically acceptable carrier is included.
  • “Pharmaceutically acceptable carrier” includes solvents, dispersion media, coatings, antibacterial and antifungal agents, agents that act isotonically to delay adsorption and the like and are suitable for pharmaceutical administration. Yes (Gennaro, 2000).
  • the carrier and preferred for diluting the carrier include, but are not limited to, water, saline, finger solution, dextrose solution, human serum albumin, and the like.
  • Non-water soluble media such as ribosomes and non-volatile oils are also used.
  • certain compounds that protect or promote activity such as the above cgrl polynucleotides may be included in the composition.
  • the pharmaceutical composition according to the present invention includes intravenous, intradermal, subcutaneous, oral (for example, including inhalation), transdermal, and transmucosal administration, and is suitable for a therapeutically appropriate route of administration. It is formulated as follows. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application include, but are not limited to, sterile diluents such as water for injection, saline solution, non-volatile oil, polyethylene.
  • Preservatives such as glycol, glycerin, propylene glycol, or other synthetic solvents, benzenolic alcohol or other methylparabens, antioxidants such as ascorbic acid or sodium hydrogen sulfite, painless such as salt benzalkonium, prohydroin hydrochloride It may contain a chemical, a chelating agent such as ethylenediaminetetraacetic acid (EDTA), a buffer such as acetate, citrate, or phosphate, and an agent for osmotic pressure adjustment such as sodium chloride or dextrose.
  • EDTA ethylenediaminetetraacetic acid
  • buffer such as acetate, citrate, or phosphate
  • agent for osmotic pressure adjustment such as sodium chloride or dextrose.
  • the pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • acids or bases such as hydrochloric acid or sodium hydroxide.
  • Parenteral preparations are contained in ampoules, disposable glass or plastic syringes or multiple dose vials.
  • compositions suitable for injection include sterile aqueous solutions (dispersible) or dispersion media and sterile powders to be prepared at the time of use.
  • suitable carriers include physiological saline, bacteriostatic water, CREMOP HOR EL TM (BASF, Parsippany, NJ), or phosphate buffered saline (PBS).
  • the composition When used as an injection, the composition must be sterile and must have sufficient fluidity to be administered using a syringe.
  • the composition The product must be stable to chemical changes and corrosion during preparation and storage, and should prevent contamination from microorganisms such as bacteria and fungi.
  • the carrier can be made using a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol), and suitable mixtures.
  • a coating agent such as lectin
  • the required particle size is maintained in a dispersion medium, and an appropriate fluidity is maintained by using a surfactant.
  • Various antibacterial and antifungal agents such as parabens, chlorobutanol, phenol, ascorbic acid, and thimerosal, can be used to prevent microbial contamination.
  • compositions such as sugar, mannitol, sorbitol, and agents that maintain isotonicity such as sodium chloride may be included in the composition.
  • Compositions that can delay adsorption include agents such as aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by sterilizing the required ingredient alone or in combination with the other ingredients and then adding the required amount of the active compound in a suitable solvent.
  • dispersion media are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the other necessary ingredients discussed above.
  • Sterile powder preparation methods for the preparation of sterile injectable solutions include vacuum drying and lyophilization to prepare a powder containing the active ingredient and any desired ingredients derived from the sterile solution. It is.
  • Oral compositions usually include an inert diluent or a carrier that does not cause harm when taken into the body.
  • Oral compositions are, for example, contained in gelatin capsules or compressed into tablets.
  • the active compound is incorporated with excipients and used in the form of tablets, troches, or capsules.
  • the oral composition can also be prepared using a fluid carrier, and the composition in the fluid carrier is applied orally.
  • pharmaceutically compatible binding agents, and Z or adjuvant substances may be included.
  • Tablets, pills, capsules, troches and the like have the following ingredients or similar properties
  • excipients such as microcrystalline cellulose, binders such as gum arabic, tragacanth or gelatin
  • bulking agents such as alginic acid, PRIM GEL, or corn starch, such as starch or ratatose Lubricants such as magnesium stearate or STRROTES; lubricants such as colloidal silicon dioxide; sweeteners such as sucrose or saccharin; or flavoring agents
  • the cgrl 1 polynucleotide and the like can be prepared as a sustained-release preparation such as a delivery system encapsulated in implantable tablets and microcapsules using a carrier that can prevent immediate removal from the body.
  • a sustained-release preparation such as a delivery system encapsulated in implantable tablets and microcapsules using a carrier that can prevent immediate removal from the body.
  • Biodegradable, biocompatible polymers can be used, such as ethylene butyrate, polyanhydrides, polydaricol acid, collagen, polyorthoesters, and polylactic acid. Such materials are available from ALZA Corporation (Mountain View, CA) and NOVA Pharmaceuticals, Inc. (Lake Elsinore, CA), and can also be readily prepared by those skilled in the art.
  • Ribosome suspensions can also be used as pharmaceutically acceptable carriers.
  • Useful ribosomes are prepared as a lipid composition containing, but not limited to, phosphatidylcholine, cholesterol and PEG-derived phosphatidylethanol (PEG-PE) through a filter of appropriate pore size so that it is sized for use, Purified by reverse phase evaporation.
  • PEG-PE PEG-derived phosphatidylethanol
  • Fab ′ fragments of antibodies can be bound to ribosomes via a disulfide exchange reaction (Martin and Papahadjopoulos, 1982).
  • the appropriate dosage level depends on the condition of the patient to be administered, the administration method, etc. If so, it can be easily optimized. In the case of injection administration, for example, it is generally preferred to administer from about 0.1 ⁇ g Zkg to about 500 mg Zkg per day of the patient's body weight, and would generally be administered in one or more divided doses. Preferably, the dosage level is from about 0.1 ⁇ g / kg to about 250 mgZkg per day, more preferably from about 0.5 to about 10 mg / kg.
  • the composition is preferably provided in the form of a tablet IJ containing preferably 1.0 to 10 mg of the active ingredient, and preferably has a productive force of .0, 5. 0, 10. 0, 15 0, 20. 0, 25. 0, 50. 0, 75. 0, 100. 0, 150. 0, 200. 0, 250. 0, 300. 0, 400. 0, 500. 0, 600. 0 , 750.0, 800.0, 900.0 and 1000.
  • the compound is administered on a regimen of! To 4 times a day, preferably once or twice a day.
  • a pharmaceutical composition or formulation should consist of uniform unit doses that ensure a certain dose.
  • a unit dose is a unit formulated with a pharmaceutically acceptable carrier, including a single dose effective for treating a patient.
  • the unit dosage of the present invention it is influenced by the physical and chemical characteristics of the compound to be formulated, the expected therapeutic effect, and the formulation considerations specific to the compound. Receive.
  • the method for introducing a nucleic acid molecule disclosed in the present invention for example, a cgrl polynucleotide, a vector having the polynucleotide inserted therein, and an antisense nucleic acid against the cgrl polynucleotide
  • a nucleic acid molecule disclosed in the present invention for example, a cgrl polynucleotide, a vector having the polynucleotide inserted therein, and an antisense nucleic acid against the cgrl polynucleotide
  • a nucleic acid molecule disclosed in the present invention for example, a cgrl polynucleotide, a vector having the polynucleotide inserted therein, and an antisense nucleic acid against the cgrl polynucleotide
  • in vivo delivery it is injected directly into the site of the patient in need of treatment.
  • ex vivo treatment cells at
  • the technique available for introducing a nucleic acid molecule into a living cell is selected depending on the force introduced in vitro into cultured cells or the like or whether it is introduced into a patient in vivo. Suitable techniques for introducing nucleic acid molecules into mammalian cells in vitro include ribosomes, electopore position, microinjection, transfusion, cell fusion, the DEAE-dextran method, and the calcium phosphate precipitation method. Transfection involves the binding of a recombinant viral (preferably retroviral) particle to a cellular receptor, followed by introduction of the nucleic acid molecule contained in the particle into the cell.
  • a commonly used vector for ex vivo delivery of genes is a retrovirus.
  • nucleic acid transfer techniques are viral or non-viral vectors (adenoviruses, lentiviruses, herpes simplex virus I, or adeno-associated viruses (AAV)), and cationic lipid-based Lipids (useful for lipid-mediated transfer of genes are eg D0TMA, DOPE, and DC_Chol; see, for example, Tonkinson et al., Cancer Investigation, 14 (1): 54_65 (1996))
  • the system is included.
  • the most preferred vector for use in gene therapy is a virus, among which is most preferably an adenovirus, AAV, lentivirus or retrovirus.
  • Viral vectors include at least one transcription promoter / enhancer or positioning factor.
  • a viral vector such as a retroviral vector, for example, a nucleic acid that functions as a cis element, that is, a translation initiation sequence, enables translation of the encoded gene when it is transcribed in a state containing a narcolepsy-related gene.
  • a viral vector such as a retroviral vector, for example, a nucleic acid that functions as a cis element, that is, a translation initiation sequence, enables translation of the encoded gene when it is transcribed in a state containing a narcolepsy-related gene.
  • Such vector constructs contain a packaging signal, a terminal repeat (LTR) or part thereof suitable for the virus used.
  • these vectors usually contain a signal sequence that causes the expressed polypeptide to be secreted from the host cell containing the vector.
  • the signal sequence for this purpose is a mammalian signal
  • the vector construct also includes a polyadenylation as well as a translation termination marker IJ.
  • a polyadenylation as well as a translation termination marker IJ.
  • it includes a 5 ′ LTR, a tRNA binding site, a packaging signal, an initiation point for DNA synthesis, and a 3 ′ LTR or a portion thereof.
  • Other non-viral vectors can use, for example, cationic lipids, polylysine, and dendrimers.
  • nucleic acid used for treatment may be provided with a reagent that targets the cell of interest, such as an antibody specific for a cell surface membrane protein or a receptor ligand on the target cell.
  • a reagent that targets the cell of interest such as an antibody specific for a cell surface membrane protein or a receptor ligand on the target cell.
  • the pharmaceutical composition can be included in the kit, container, pack along with instructions for administration.
  • the pharmaceutical composition according to the present invention is supplied as a kit, different components of the pharmaceutical composition are packaged in separate containers and mixed immediately before use. In this way the components They are packaged separately to allow long-term storage without losing the function of the active component.
  • Reagents contained in the kit are supplied in any type of container in which the components remain active for an extended period of time, are not adsorbed by the container material, and are not subject to alteration.
  • a sealed glass ampoule contains a buffer packaged under a neutral, non-reactive gas such as nitrogen gas.
  • Ampoules are composed of organic polymers such as glass, polycarbonate, polystyrene, ceramics, metals, or any other suitable material commonly used to hold reagents.
  • Examples of other suitable containers include simple bottles made from similar materials such as ampoules, and packaging materials that are internally lined with foil such as aluminum or alloys.
  • Other containers include test tubes, nozzles, flasks, bottles, syringes, or the like.
  • the container has a sterile access port such as a bot- nore with a stopper that can be penetrated by a hypodermic needle.
  • the kit also includes instructions for use. Instructions for using the kit comprising the pharmaceutical composition are printed on paper or other material and / or floppy disk, CD-ROM, DVD-ROM, Zip disk, video tape, audio tape, etc. It may be supplied as an electrically or electromagnetically readable medium. Detailed instructions for use may actually be included in the kit or may be posted on a website designated by the kit manufacturer or distributor or notified by e-mail, etc. .
  • the cgrl l gene (including not only the etason region but also the intron region and the promoter region) or cgrl l polypeptide variation (eg, point mutation, deletion, etc.), mutation in the expression control region of the gene
  • cgrl l gene including not only the etason region but also the intron region and the promoter region
  • cgrl l polypeptide variation eg, point mutation, deletion, etc.
  • a test sample is obtained from a patient suspected of having an abnormality in the proliferation and survival of specific cells, the antibody is brought into contact with the test sample, and the test sample and the antibody By detecting the presence or absence of binding, it is possible to examine the onset or possibility of onset of abnormalities in the proliferation and survival of the cells.
  • the presence or absence of binding between the test sample and the antibody can be confirmed by immunoprecipitation using antibodies, Western blotting, immunohistochemistry, ELI SA, and the like.
  • the polypeptide of the present invention is not detected at all or it is determined that the polypeptide is present in an extremely small amount in the living body, the onset of the abnormality is suspected.
  • primers, probes, etc. that can anneal with the cDNA sequence
  • genomic DNA sequence including transcriptional control region, intron region, etc.
  • Primers that can be used are generally 15 bp to: OO bp, and preferably ⁇ has a length of 17 bp to 30 bp.
  • the probe that can be used generally has a length of 15 bp or more, and hybridizes with at least a partial region of the coding region and non-coding region (including transcription control region, intron region, etc.) of the gene of the present invention. Anything that can be used can be used. Further, in order to confirm that the probe hybridizes to the target DNA region, the probe can be used in a state where it can be detected by a fluorescent dye, a radiolabel or the like.
  • Other methods for confirming functional abnormality of the cgrl 1 gene include, for example, obtaining a test sample from a subject and preparing the mRNA of the cgrl 1 gene prepared from the test sample, or preparing the mRNA force thereof.
  • a step of amplifying the partial region is included.
  • abnormalities were found in the expression level of the gene of the present invention compared to samples derived from healthy subjects. If possible, the onset or risk of dysfunction of the cgrl l gene can be predicted.
  • “mammal” means any animal classified as a mammal, and is not particularly limited. For example, in addition to humans, pet animals such as dogs and cats, ushi, pigs, hidges, horses, etc. It is a domestic animal. Particularly preferred “mammals” are humans.
  • the present invention includes a method for preventing or treating a diseased mammal suffering from or at risk of suffering from the disease.
  • treatment means to prevent or alleviate the progression of the pathological condition of a disease in a mammal that is likely to suffer from or suffers from the disease. It is used as a broad meaning including.
  • disease includes all pathological conditions caused by the presence or quantitative fluctuation of cgrl l gene or polypeptide, such as blood flow disorder, developmental disorder-related disease, neurological disease, cancer, hypotension. , Hypertension, and heart disease.
  • “mammal” means any animal classified as a mammal, and is not particularly limited.
  • pet animals such as dogs and cats, ushi, pigs, hidges, horses, etc. It is a domestic animal.
  • Particularly preferred “mammals” are humans.
  • the gene encoding the protein corresponding to spot2 in Fig. 1 is cgr 11 and has an EF-hand motif, and it has been reported that p53 suppresses cell proliferation.
  • this protein (cglrl polypeptide) has a secretion signal and is cleaved at the position of the secretion signal, and the measured molecular weight and the estimated molecular force of the protein are larger than expected. Therefore, it was guessed that it was modified with sugar. This indicates that this protein is a new secreted protein compared to previous reports.
  • Figure 3 shows the alignment of the amino acid sequences of rat and human derived cglrl polypeptides.
  • rat cgrl l was forcibly expressed in the cells (using the construct shown in FIG. 10), and the N-terminal amino acid sequence of the cgrl l protein purified from the culture supernatant was analyzed according to a conventional method. It was revealed that the signal peptide region is composed of 29 amino acid sequences (Fig. 5). This finding is different from the conventionally known cleavage rules for signal peptide sequences. Therefore, this result reveals the in vivo functional region of cgrl l protein for the first time and is an important finding.
  • HRE hypoxia response element
  • An antibody was prepared by synthesizing a peptide against the hydrophilic site of the amino acid sequence deduced from the gene sequence.
  • the site (sequence site shown as an antigenic site) present in FIGS. 3 and 5 was effective as an antigen, and a specific antibody was successfully produced. This part can be applied to measure the abundance of cgr 11 in the future.
  • PCR showed that cgrl 1 was specifically expressed in pituitary hormone-producing cells (MtTZS) (Fig. 7). This suggests that cgrl l protein is strongly related to the secretory function of cells. It was also suggested that it can be used to measure the secretory ability of hormones in vivo by using the abundance of cgrl l protein as an index.
  • cgrl l protein was specifically present in the Golgi apparatus (Fig. 8), supporting the above view that cgrl l polynucleotide was a secreted protein. Furthermore, the distribution was consistent with the distribution of growth hormone in hormone-producing cells (Fig. 8). Immunocytochemistry using an antibody specific for cgrl 1 protein revealed that cells with cgrl 1 protein were the anterior lobe after the pituitary gland and were not expressed at all in the pituitary midlobe ( Figure 9A). ).
  • cgrl l protein is abundantly expressed in sites with abundant blood vessels (posterior pituitary, anterior lobe), while it is not expressed at all in the region without any blood vessels (pituitary midlobe). It was strongly suggested that there was not.
  • the expression of cgrl 1 protein was also confirmed in the large follicle (Fig. 9C) and the outer follicular membrane of mature ovarian follicles (Fig. 9D). All of these sites are rich in blood vessels, suggesting that the cgrl l protein plays an important role in the formation and / or maintenance of the vasculature.
  • FIG. 10 shows the expression vector construct.
  • the cgrl 1 gene was inserted into the EcoRI and Xbal sites of the vector pCl_neo Vector (Promega) used for expression, and subcloning was performed according to a conventional method.
  • the resulting expression vector uses Lipofectamine 2000 It was introduced into pituitary-derived cell lines, MtT / E—B3 cells (MtT / E cells registered in RIKEN BioResource Center: subclone cells of RCB1278) and MtT / Se cells (registered RIKEN Bioresource Center: RCB0529).
  • cgrl l forced expression cells the cgrl l protein was localized in the gonoredi apparatus (FIGS. 11A and C), suggesting that the cgrl l protein also functions as a secreted protein in the forced expression cells. Furthermore, the presence of cgrl l protein was confirmed in the culture medium, strongly suggesting that cgrl l protein is a secreted protein (Figs. 12 and 13).
  • cgrl 1 gene product was forcibly expressed in MtTZE- B3 cells, and the obtained cells were transplanted subcutaneously into the groin of rats (Fischer344: female, lOOg) (2xl0 6 cells) Z200 ⁇ 1 / rat). On the 20th day after transplantation, the animals were laparotomized, the tumor was removed, and its weight was measured.
  • PC12 cells which are well known as neural progenitor cells in adrenal pheochromocytoma
  • PC12 cells are known to be unable to survive in serum-free media. Since the expression of cgrl 1 protein is low in this cell, it was introduced into PC12 cells into which the cgrl 1 gene was introduced, and a forced expression system was constructed. Inoculate 48 well plates with a forced expression clone (Fig. 17; clones C to G) and a non-forced clone (Fig. 17; control) with 5.00E + 4Z (200 ⁇ ), 2 The serum-free medium was replaced after a day. The number of viable cells was counted on day 8 of serum-free culture. As a result, it was revealed that the survival of PC12 cells in which cgrl l protein was forcibly expressed was significantly promoted even in serum-free medium (Fig. 17).
  • cgrl l protein is highly expressed in sites with blood vessels rich in the posterior and anterior pituitary glands, while it is not expressed at all in sites without blood vessels (pituitary middle lobe) and the colon (Fig. 9c) and the outer follicular membrane of mature ovarian follicles (Fig. 9D) have also been confirmed to express cgrl 1 protein, and cgrl l protein plays an important role in the formation and / or maintenance of vasculature. It has been suggested.
  • the left thigh was incised and the left femoral artery was detached. Hemostasis was achieved by fixing the left femoral artery with tweezers. A suture was passed through the proximal and distal sides of the left femoral artery. A hole was made in the femoral artery with a Weeckenor's scissors, and the PE10 side of a blood pressure measuring force neule filled with 500 U of Parin (Mochida Pharmaceutical) Z saline was inserted. The force needle and the blood vessel were fixed with the suture thread that had been passed through earlier, and it was confirmed that blood entered the tube while pulsating.
  • Parin Parin
  • the force neulet was delivered to the back of the head through the subcutaneous site. At this time, I was careful not to break the power. Once again, it was confirmed that blood was rising while pulsating, and stoppered with a stopper (length 2 cm, diameter 0.66 mm) (Silver Scale No. 16, TRAY). Subcutaneous chew The tube that touched the skin on the back of the head was squeezed with gum tape to fix the hub, the gum tape and the skin were sewn, and the tube exiting from the back of the head was cut to about 4 cm. One day after the operation, the blood pressure was measured by the following method under non-anesthesia after the animal had recovered.
  • the rat was placed in a cage, a blood pressure measuring force neuron and an extension tube were connected, and connected to a transducer (Bioresearch Corporation).
  • the tube was filled with 10 U of Parin Z physiological saline, and care was taken to prevent air from entering.
  • Stainless steel pipes were used for the connection between the force neulet and the extension tube.
  • PowerLab Bioresearch Corporation was used as blood pressure measurement and analysis software.
  • the cgrl1 protein (0.1 ⁇ g / 500 ⁇ 1) purified with an antibody from the culture medium of cgrl1 protein-secreting cells was injected into the abdominal cavity of the rat, and the time course of mean blood pressure was measured. As a result, an increase in blood pressure was observed after the purified cgrl l protein was injected intraperitoneally (Fig. 20). On the other hand, no increase in blood pressure was observed when a control sample was injected using a culture solution containing no purified cgrl protein.

Abstract

Disclosed is a composition or method for the promotion of cell growth, the life support or the inhibition of tumor growth. A new activity of a cgr11 protein has been found which is quite different from the physiological activities thereof which have ever been reported. Based on this finding, there are provided a composition which can promote the cell growth and can be involved in the life support, comprising a cgr11 polypeptide, a nucleotide encoding the polypeptide, an antagonist of the polypeptide or the like; and a method for the treatment, prevention or diagnosis of a disease.

Description

明 細 書  Specification
下垂体細胞由来の新規分泌タンパク質とその用途  Novel secreted proteins derived from pituitary cells and their uses
技術分野  Technical field
[0001] 本発明は、細胞の増殖促進、生存維持、または腫瘍の増殖抑制のための組成物に 関する。より詳細には、細胞の増殖促進、生存維持に関与するポリペプチド、該ポリ ペプチドをコードするヌクレオチド、または該ポリペプチドに対するアンタゴニストなど を包含せしめた組成物に関する。  [0001] The present invention relates to a composition for promoting cell growth, maintaining survival, or suppressing tumor growth. More specifically, the present invention relates to a composition comprising a polypeptide involved in promoting cell growth and maintaining survival, a nucleotide encoding the polypeptide, an antagonist to the polypeptide, or the like.
背景技術  Background art
[0002] 細胞の増殖の制御および生存維持は、生体秩序を維持する上で極めて重要な機 構であると考えられる。例えば、細胞の増殖の制御が十分に機能せず、無秩序な細 胞の増殖が行われると、癌などの深刻な疾患を引き起こす可能性があり、一方、細胞 の増殖または維持が適切に行われない場合には、生体内器官または生体自体の発 達障害に関連する疾患の発症が予期される。  Control of cell proliferation and maintenance of survival are considered to be extremely important mechanisms for maintaining biological order. For example, if cell growth control is not fully functioning and disordered cell growth can occur, it can cause serious diseases such as cancer, while cells are properly grown or maintained. If not, the onset of a disease related to the developmental disorder of the in vivo organ or the living body itself is expected.
[0003] なかでも、癌は、依然として疾患による死因のトップを占めており、癌を効果的に治 療するための薬剤および治療方法を開発することは、医学 ·生物学領域において、 最も重要な研究課題となっている。  [0003] Among them, cancer remains the leading cause of death due to disease, and the development of drugs and treatment methods for the effective treatment of cancer is the most important in the medical and biological fields. It is a research subject.
また、発達障害についても、生活の質(Quality of life)の向上を行う上でも解決すベ き重要な課題となっている。  Developmental disabilities are also important issues that need to be resolved in order to improve the quality of life.
生体の成長、生殖、免疫などの機能に深く関わる器官の一つに下垂体を挙げること 力 Sできる。下垂体はホルモンなどを分泌する器官で、この器官において発現、機能し ている因子は、生体の成長の制御等に重要な役割を果たしている可能性が予想され る。そして、下垂体の細胞レベルにおける機能について鑑みるに、下垂体は各種生 体臓器を形成する細胞の成長 ·増殖、維持等にも何らかの制御的役割を果たしてい ると思われる。  The pituitary gland is one of the organs deeply involved in functions such as growth, reproduction, and immunity. The pituitary gland is an organ that secretes hormones. Factors expressed and functioning in this organ are expected to play an important role in the control of biological growth. In view of the function of the pituitary gland at the cellular level, the pituitary gland seems to play some regulatory role in the growth, proliferation, and maintenance of cells that form various biological organs.
このように、秩序立った正常な生体機能を発揮する上での下垂体の重要性が明ら かになつてきた反面、下垂体における未知なる機能因子の探索はなかなか進展しな い状況が続いていた。 [0004] Madden等により、 p53により発現が制御される遺伝子として cgrl 1遺伝子が同定 された (非特許文献 1)。また、 cgrl l遺伝子は機能的 p53遺伝子発現に伴い、発現 上昇が認められる(特許文献 1及び 2)。 Madden等によると、 cgrl lは p53経路の下 流において細胞増殖の抑制的制御に関与しているとのことである力 その詳細につ レ、ては明らかにされていない。その他、胃ガンにおける遺伝子発現率が高いこと(特 許文献 3)、さらに最近では、 cgrl l遺伝子が痛みに関連することが示唆される遺伝 子として同定されたことが報告されており(特許文献 4)、 cgrl lの実際の機能につい ては統一的見解が得られておらず、未だその機能を解明するには至っていなレ、。さ らには、 cgrl lによってコードされるタンパク質がどのような形態、すなわち、タンパク 質に翻訳されたままで機能するの力、、分泌タンパク質して機能しているの力 \或いは 、糖鎖などによる翻訳後修飾などが行われているかどうかなど、生体内における機能 メカニズムを理解する上で重要な情報が不足している。 In this way, the importance of the pituitary gland for demonstrating orderly and normal biological functions has become clear, but the search for unknown functional factors in the pituitary gland continues to be difficult. It was. [0004] Madden et al. Identified the cgrl 1 gene as a gene whose expression is regulated by p53 (Non-patent Document 1). In addition, the cgrl l gene is upregulated with functional p53 gene expression (Patent Documents 1 and 2). According to Madden et al., Cgrl l is involved in the inhibitory control of cell proliferation in the downstream of the p53 pathway. In addition, it has been reported that the gene expression rate in gastric cancer is high (Patent Document 3), and more recently, the cgrl l gene has been identified as a gene suggested to be related to pain (Patent Document). 4) There is no unified view of the actual function of cgrl l, and the function has not yet been elucidated. In addition, it depends on what form the protein encoded by cgrl l functions, ie, the ability to function as it is translated into protein, the ability to function as a secreted protein, or the sugar chain, etc. There is a lack of information that is important for understanding functional mechanisms in vivo, such as whether post-translational modifications are performed.
[0005] 非特許文献 1 : Madden等 Cancer Res. 56 ; 5384_5390 1996  [0005] Non-Patent Document 1: Madden et al. Cancer Res. 56; 5384_5390 1996
特許文献 1:国際公開公報 WO99/01581  Patent Document 1: International Publication WO99 / 01581
特許文献 2:国際公開公報 WO 97/45542  Patent Document 2: International Publication WO 97/45542
特許文献 3 :国際公開公報 WO2005/010213  Patent Document 3: International Publication WO2005 / 010213
特許文献 4 :国際公開公報 WO03/016475  Patent Document 4: International Publication WO03 / 016475
発明の開示  Disclosure of the invention
発明が解決しょうとする課題  Problems to be solved by the invention
[0006] 本発明者らは、上記事情に鑑み、細胞の増殖制御等に関与する新規下垂体因子 の存否について鋭意研究を行った結果、下垂体由来の遺伝子産物が腫瘍形成の促 進 ·細胞生存の維持などに関与していること、分泌タンパク質として機能する可能性 などを明らかにした。 [0006] In view of the above circumstances, the present inventors have conducted extensive research on the existence of a novel pituitary factor involved in cell growth control and the like. As a result, the gene product derived from the pituitary gland promotes tumor formation. It has been clarified that it is involved in maintaining survival and functions as a secreted protein.
よって、本発明は、細胞の増殖促進又は生存維持のための組成物、及び関連疾患 の治療方法の提供を目的とする。  Therefore, an object of the present invention is to provide a composition for promoting cell growth or maintaining survival, and a method for treating a related disease.
さらに、本発明は細胞増殖抑制のための組成物、及び関連疾患の治療方法の提 供を目的とする。  Furthermore, an object of the present invention is to provide a composition for inhibiting cell growth and a method for treating a related disease.
また、本発明は、癌、高血圧症などの発症の有無等を判定するための診断用組成 物及び診断方法の提供を目的とする。 The present invention also relates to a diagnostic composition for determining the presence or absence of onset of cancer, hypertension, etc. The purpose is to provide products and diagnostic methods.
また、本発明は、実際の生体内で機能する cgrl lコードタンパク質を提供することを 目的とする。  Another object of the present invention is to provide a cglrl-encoded protein that functions in an actual living body.
課題を解決するための手段  Means for solving the problem
[0007] 本発明は、生体の成長等をコントロールする上で重要な役割を担っている下垂体 で機能する因子において、特に細胞の増殖制御等に深く関与すると考えられる因子 として cgrl l遺伝子産物を見出したことに基づくものである。具体的には、ホルモン産 生能の異なる下垂体細胞間におけるタンパク質発現の差に着目し、ホルモン産生細 胞においてのみ発現が認められる因子の探索を行レ、、その 1つとして cgrl lを同定し た。発明者は、 cgrl l遺伝子産物が p53の下流において細胞抑制に関与していると の従来の知見とは全く異なる結果を明らかにし、 cgrl lタンパク質の新規な活性に着 目し、本課題を解決するに至った。  [0007] The present invention relates to a cgrl gene product as a factor that functions in the pituitary gland that plays an important role in controlling the growth of a living body, and is considered to be deeply involved in cell growth control. This is based on the finding. Specifically, focusing on differences in protein expression between pituitary cells with different hormone-producing ability, we searched for factors that are only expressed in hormone-producing cells, and identified cgrl l as one of them. did. The inventor clarified a result completely different from the conventional finding that the cgrl l gene product is involved in cell suppression downstream of p53, and focused on the novel activity of the cgrl l protein to solve this problem. It came to do.
[0008] すなわち、本発明は以下の(1)〜(34)に関する。  That is, the present invention relates to the following (1) to (34).
(1)本発明の第 1の態様は、「以下の(a)又は (b)に示されるポリペプチド、又はその ァゴニスト、及び薬剤的に許容される担体を含んでなる、細胞の増殖促進又は生存 維持のための組成物。  (1) A first aspect of the present invention is a cell proliferation promoting or comprising the polypeptide shown in the following (a) or (b), or an agonist thereof, and a pharmaceutically acceptable carrier: A composition for maintaining survival.
(a)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列からなるポリペプチド  (a) a polypeptide comprising the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12
(b)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列において、 1若しくは数個のアミノ酸の置換、欠失若しくは揷 入をもつアミノ酸配列からなり、かつ、細胞の増殖促進活性または生存維持活性を有 するポリペプチド」である。  (b) In the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12, substitution of one or several amino acids, deletion or insertion A polypeptide having an amino acid sequence and having a cell growth-promoting activity or survival-sustaining activity ".
(2)本発明の第 2の態様は、「以下の(a)又は (b)に示されるポリヌクレオチドを含む 組換えベクター、及び薬剤的に許容される担体を含んでなる、細胞の増殖促進又は 生存維持のための組成物。  (2) The second aspect of the present invention is “promoting cell growth, comprising a recombinant vector comprising a polynucleotide shown in the following (a) or (b) and a pharmaceutically acceptable carrier: Or a composition for maintaining survival.
(a)配列番号 1、配列番号 3、配列番号 5、配列番号 7、配列番号 9又は配列番号 11 で表されるヌクレオチド配列からなるポリヌクレオチド  (a) a polynucleotide comprising the nucleotide sequence represented by SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11
(b)配列番号 1、配列番号 3、配列番号 5、配列番号 7、配列番号 9又は配列番号 11 で表されるヌクレオチド配列からなるポリヌクレオチドの相補鎖と高ストリンジェントな 条件下でハイブリダィズするポリヌクレオチドであって、該ポリヌクレオチドがコードす るポリペプチドが細胞の増殖促進活性または生存維持活性を有するポリヌクレオチド 」である。 (b) SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11 A polynucleotide that hybridizes under high stringency conditions with a complementary strand of a polynucleotide comprising the nucleotide sequence represented by the above, wherein the polypeptide encoded by the polynucleotide has a cell growth-promoting activity or a life-sustaining activity. Polynucleotide ".
(3)本発明の第 3の態様は、「前記細胞が血管内皮細胞である上記(1)又は(2)に 記載の組成物」である。  (3) The third aspect of the present invention is “the composition according to (1) or (2) above, wherein the cells are vascular endothelial cells”.
(4)本発明の第 4の態様は、「前記細胞が下垂体を形成する細胞である上記(1)又 は(2)に記載の組成物」である。  (4) The fourth aspect of the present invention is “the composition according to (1) or (2) above, wherein the cell is a cell forming a pituitary gland”.
(5)本発明の第 5の態様は、「前記細胞が神経細胞又はクロム親和細胞である上記( 1)又は(2)に記載の組成物」である。  (5) The fifth aspect of the present invention is “the composition according to (1) or (2) above, wherein the cell is a neuronal cell or a chromaffin cell”.
(6)本発明の第 6の態様は、「血流障害関連疾患の治療、又は予防に供される上記( (6) The sixth aspect of the present invention is the above-mentioned (used in the treatment or prevention of a blood flow disorder-related disease (
1)又は(2)に記載の医薬組成物」である。 “The pharmaceutical composition according to 1) or (2)”.
(7)本発明の第 7の態様は、「低血圧症の治療、又は予防に供される上記(1)又は(( 7 ) The seventh aspect of the present invention is the above (1) or (1) used for the treatment or prevention of hypotension.
2)に記載の医薬組成物」である。 The pharmaceutical composition according to 2).
(8)本発明の第 8の態様は、「発育障害関連疾患の治療、又は予防に供される上記( (8) The eighth aspect of the present invention is the above-mentioned (used for the treatment or prevention of a developmental disorder-related disease (
1)又は(2)に記載の医薬組成物」である。 “The pharmaceutical composition according to 1) or (2)”.
(9)本発明の第 9の態様は、「神経疾患の治療、又は予防に供される上記(1)又は( (9) A ninth aspect of the present invention is the above (1) or (
2)に記載の医薬組成物」である。 The pharmaceutical composition according to 2).
(10)本発明の第 10の態様は、「以下の(a)又は (b)に示されるポリペプチドに対する アンタゴニスト、及び薬剤的に許容される担体を含んでなる、細胞の増殖抑制又は生 存抑制のための組成物。  (10) The tenth aspect of the present invention is “cell growth inhibition or survival comprising an antagonist to the polypeptide shown in the following (a) or (b) and a pharmaceutically acceptable carrier”: Composition for suppression.
(a)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列からなるポリペプチド  (a) a polypeptide comprising the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12
(b)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列において、 1若しくは数個のアミノ酸の置換、欠失若しくは揷 入をもつアミノ酸配列からなり、かつ、細胞の増殖促進活性または生存維持活性を有 するポリペプチド」である。  (b) In the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12, substitution of one or several amino acids, deletion or insertion A polypeptide having an amino acid sequence and having a cell growth-promoting activity or survival-sustaining activity ".
(11)本発明の第 11の態様は、「前記アンタゴニストが抗体である上記(10)に記載の 組成物」である。 (11) The eleventh aspect of the present invention is as described in (10) above, wherein the antagonist is an antibody. A "composition".
(12)本発明の第 12の態様は、「前記抗体が配列番号 13又は 14で表されるアミノ酸 配列からなるポリペプチドと特異的に結合するものである上記(11)に記載の組成物」 である。  (12) The twelfth aspect of the present invention is “the composition according to (11) above, wherein the antibody specifically binds to a polypeptide consisting of the amino acid sequence represented by SEQ ID NO: 13 or 14”. It is.
(13)本発明の第 13の態様は、「以下の(a)又は (b)に示されるポリヌクレオチドの機 能を阻害する核酸を含む組換えベクター、及び薬剤的に許容される担体を含んでな る、細胞の増殖抑制又は生存抑制のための組成物。  (13) The thirteenth aspect of the present invention includes "a recombinant vector containing a nucleic acid that inhibits the function of the polynucleotide shown in the following (a) or (b)" and a pharmaceutically acceptable carrier. A composition for inhibiting cell growth or survival.
(a)配列番号 1、配列番号 3、配列番号 5、配列番号 7、配列番号 9又は配列番号 11 で表されるヌクレオチド配列からなるポリヌクレオチド  (a) a polynucleotide comprising the nucleotide sequence represented by SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11
(b)配列番号 1、配列番号 3、配列番号 5、配列番号 7、配列番号 9又は配列番号 11 で表されるヌクレオチド配列からなるポリヌクレオチドの相補鎖と高ストリンジェントな 条件下でハイブリダィズするポリヌクレオチドであって、該ポリヌクレオチドがコードす るポリペプチドが細胞の増殖促進活性または生存維持活性を有するポリヌクレオチド Jである。  (b) a polynucleotide that hybridizes with a complementary strand of a polynucleotide comprising the nucleotide sequence represented by SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11 under highly stringent conditions Polynucleotide J, which is a nucleotide and the polypeptide encoded by the polynucleotide has a cell growth promoting activity or a survival maintaining activity.
(14)本発明の第 14の態様は、「前記機能を阻害する核酸が cgrl l遺伝子に対する アンチセンス DNA又は RNAである上記(13)に記載の組成物」である。  (14) A fourteenth aspect of the present invention is “the composition according to (13) above, wherein the nucleic acid that inhibits the function is an antisense DNA or RNA against the cgrl l gene”.
(15)本発明の第 15の態様は、「前記細胞が血管内皮細胞である上記(10)又は(1 3)に記載の組成物」である。  (15) The fifteenth aspect of the present invention is “the composition according to (10) or (13) above, wherein the cell is a vascular endothelial cell”.
(16)本発明の第 16の態様は、「前記細胞が下垂体を形成する細胞である上記(10 )又は(13)に記載の組成物」である。  (16) A sixteenth aspect of the present invention is "the composition according to (10) or (13) above, wherein the cell is a cell forming a pituitary gland".
(17)本発明の第 17の態様は、「癌の治療、又は予防に供される上記(10)又は(13 (17) The seventeenth aspect of the present invention is the above (10) or (13
)に記載の医薬組成物」である。 ).
(18)本発明の第 18態様は、「発育障害関連疾患の治療、又は予防に供される上記 (10)又は(13)に記載の医薬組成物」である。  (18) The eighteenth aspect of the present invention is “the pharmaceutical composition according to the above (10) or (13), which is used for the treatment or prevention of a developmental disorder-related disease”.
(19)本発明の第 19態様は、「高血圧症の治療、又は予防に供される上記(10)又は (13)に記載の医薬組成物」である。  (19) The nineteenth aspect of the present invention is “the pharmaceutical composition according to the above (10) or (13), which is used for the treatment or prevention of hypertension”.
(20)本発明の第 20の態様は、「以下の(a)又は (b)に示されるポリペプチドに対する 抗体を含んでなる、血流障害関連疾患、低血圧症、発育障害関連疾患、癌、高血圧 症又は神経疾患の発症の有無、予後、又は悪性度の診断用組成物。 (20) According to a twentieth aspect of the present invention, “a blood flow disorder-related disease, hypotension, developmental disorder-related disease, cancer, comprising an antibody against the polypeptide shown in the following (a) or (b)”: High blood pressure Composition for diagnosis of presence or absence, prognosis, or grade of malignancy or neurological disease.
(a)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列からなるポリペプチド  (a) a polypeptide comprising the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12
(b)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列において、 1若しくは数個のアミノ酸の置換、欠失若しくは揷 入をもつアミノ酸配列からなり、かつ、細胞の増殖促進活性または生存維持活性を有 するポリペプチド」である。  (b) In the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12, substitution of one or several amino acids, deletion or insertion A polypeptide having an amino acid sequence and having a cell growth-promoting activity or survival-sustaining activity ".
(21)本発明の第 21の態様は、「前記抗体が配列番号 13又は 14で表されるアミノ酸 配列からなるポリペプチドと特異的に結合するものである上記(20)に記載の診断用 組成物」である。  (21) The twenty-first aspect of the present invention is the diagnostic composition according to the above (20), wherein the antibody specifically binds to a polypeptide consisting of the amino acid sequence represented by SEQ ID NO: 13 or 14. Things ".
(22)本発明の第 22の態様は、「以下の(a)又は (b)に示されるポリペプチドに対する アンタゴニスト。  (22) The twenty-second aspect of the present invention is “an antagonist against the polypeptide shown in the following (a) or (b).
(a)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列からなるポリペプチド  (a) a polypeptide comprising the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12
(b)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列において、 1若しくは数個のアミノ酸の置換、欠失若しくは挿 入をもつアミノ酸配列からなり、かつ、細胞の増殖促進活性または生存維持活性を有 するポリペプチド」である。  (b) In the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12, substitution of one or several amino acids, deletion or insertion A polypeptide having an amino acid sequence and having a cell growth-promoting activity or survival-sustaining activity ".
(23)本発明の第 23の態様は、「以下の(a)又は (b)に示されるポリペプチドに対する 抗体。  (23) A twenty-third aspect of the present invention is “an antibody against a polypeptide shown in the following (a) or (b):
(a)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列からなるポリペプチド  (a) a polypeptide comprising the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12
(b)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列において、 1若しくは数個のアミノ酸の置換、欠失若しくは揷 入をもつアミノ酸配列からなり、かつ、細胞の増殖促進活性または生存維持活性を有 するポリペプチド」である。  (b) In the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12, substitution of one or several amino acids, deletion or insertion A polypeptide having an amino acid sequence and having a cell growth-promoting activity or survival-sustaining activity ".
(24)本発明の第 24の態様は、「配列番号 13又は 14で表されるアミノ酸配列からな るポリペプチドと特異的に結合する上記(23)に記載の抗体」である。 (25)本発明の第 25の態様は、「以下の(a)又は (b)に示されるポリペプチド、又はそ のァゴニストの治療的有効量を投与することを含んでなる、哺乳動物の血流障害関 連疾患、低血圧症、発育障害関連疾患又は神経疾患の治療又は予防方法。 (24) The 24th aspect of the present invention is “the antibody according to the above (23), which specifically binds to a polypeptide consisting of the amino acid sequence represented by SEQ ID NO: 13 or 14.” (25) A twenty-fifth aspect of the present invention is a method for treating a blood of a mammal comprising administering a therapeutically effective amount of a polypeptide shown in the following (a) or (b), or an agonist thereof: A method for the treatment or prevention of flow disorders-related diseases, hypotension, developmental disorder-related diseases or neurological diseases.
(a)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列からなるポリペプチド  (a) a polypeptide comprising the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12
(b)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列において、 1若しくは数個のアミノ酸の置換、欠失若しくは揷 入をもつアミノ酸配列からなり、かつ、細胞の増殖促進活性または生存維持活性を有 するポリペプチド」である。  (b) In the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12, substitution of one or several amino acids, deletion or insertion A polypeptide having an amino acid sequence and having a cell growth-promoting activity or survival-sustaining activity ".
(26)本発明の第 26の態様は、「以下の(a)又は (b)に示されるポリペプチドに対する アンタゴニストの治療的有効量を投与することを含んでなる、哺乳動物の癌、発育障 害関連疾患又は高血圧症の治療又は予防方法。  (26) The twenty-sixth aspect of the present invention is a cancer, developmental disorder in mammals comprising administering a therapeutically effective amount of an antagonist to the polypeptide shown in the following (a) or (b): A method for treating or preventing harm-related diseases or hypertension.
(a)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列からなるポリペプチド  (a) a polypeptide comprising the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12
(b)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列において、 1若しくは数個のアミノ酸の置換、欠失若しくは挿 入をもつアミノ酸配列からなり、かつ、細胞の増殖促進活性または生存維持活性を有 するポリペプチド」である。  (b) In the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12, substitution of one or several amino acids, deletion or insertion A polypeptide having an amino acid sequence and having a cell growth-promoting activity or survival-sustaining activity ".
(27)本発明の第 27の態様は、「前記アンタゴニストが抗体である上記(26)に記載の 方法」である。  (27) A twenty-seventh aspect of the present invention is “the method according to (26) above, wherein the antagonist is an antibody”.
(28)本発明の第 28の態様は、「以下の(a)又は (b)に示されるポリヌクレオチドの機 能を阻害する核酸を含む組換えベクターの治療的有効量を投与することを含んでな る、哺乳動物の癌、発育障害関連疾患又は高血圧症の治療又は予防方法。  (28) The twenty-eighth aspect of the present invention includes "administering a therapeutically effective amount of a recombinant vector comprising a nucleic acid that inhibits the function of the polynucleotide shown in the following (a) or (b)" A method for treating or preventing cancer, developmental disorder-related disease or hypertension in mammals.
(a)配列番号 1、配列番号 3、配列番号 5、配列番号 7、配列番号 9又は配列番号 11 で表されるヌクレオチド配列からなるポリヌクレオチド  (a) a polynucleotide comprising the nucleotide sequence represented by SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11
(b)配列番号 1、配列番号 3、配列番号 5、配列番号 7、配列番号 9又は配列番号 11 で表されるヌクレオチド配列からなるポリヌクレオチドの相補鎖と高ストリンジェントな 条件下でハイブリダィズするポリヌクレオチドであって、該ポリヌクレオチドがコードす るポリペプチドが細胞の増殖促進活性または生存維持活性を有するポリヌクレオチド Jである。 (b) a polynucleotide that hybridizes with a complementary strand of a polynucleotide comprising the nucleotide sequence represented by SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11 under highly stringent conditions A nucleotide, the polynucleotide encoding Is a polynucleotide J having a cell growth promoting activity or a survival maintaining activity.
(29)本発明の第 29の態様は、「前記機能を阻害する核酸が cgrl l遺伝子に対する アンチセンス DNA又は RNAである上記(28)に記載の方法」である。  (29) A twenty-ninth aspect of the present invention is “the method according to (28) above, wherein the nucleic acid that inhibits the function is an antisense DNA or RNA against the cgrl l gene”.
(30)本発明の第 30の態様は、「哺乳動物から得た組織細胞又は血液中における、 以下の(a)又は (b)に示されるポリペプチドの量を定量的に検出する事を含んでなる (30) The thirtieth aspect of the present invention includes "quantitatively detecting the amount of the polypeptide shown in the following (a) or (b) in tissue cells or blood obtained from mammals" Become
、血流障害関連疾患、低血圧症、発育障害関連疾患、癌、高血圧症、心臓疾患又は 神経疾患の診断方法。 , Blood flow disorder-related diseases, hypotension, developmental disorder-related diseases, cancer, hypertension, heart disease or neurological disease diagnostic method.
(a)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列からなるポリペプチド  (a) a polypeptide comprising the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12
(b)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列において、 1若しくは数個のアミノ酸の置換、欠失若しくは揷 入をもつアミノ酸配列からなり、かつ、細胞の増殖促進活性または生存維持活性を有 するポリペプチド」である。  (b) In the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12, substitution of one or several amino acids, deletion or insertion A polypeptide having an amino acid sequence and having a cell growth-promoting activity or survival-sustaining activity ".
(31)本発明の第 31の態様は、「以下の(a)又は (b)に示されるポリヌクレオチドを含 む組換えベクターを細胞に導入し、不死化した細胞を製造する方法。  (31) A thirty-first aspect of the present invention is “a method for producing an immortalized cell by introducing a recombinant vector containing a polynucleotide shown in the following (a) or (b) into a cell:
(a)配列番号 1、配列番号 3、配列番号 5、配列番号 7、配列番号 9又は配列番号 11 で表されるヌクレオチド配列からなるポリヌクレオチド  (a) a polynucleotide comprising the nucleotide sequence represented by SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11
(b)配列番号 1、配列番号 3、配列番号 5、配列番号 7、配列番号 9又は配列番号 11 で表されるヌクレオチド配列からなるポリヌクレオチドの相補鎖と高ストリンジェントな 条件下でハイブリダィズするポリヌクレオチドであって、該ポリヌクレオチドがコードす るポリペプチドが細胞の増殖促進活性または生存維持活性を有するポリヌクレオチド 」である。  (b) a polynucleotide that hybridizes with a complementary strand of a polynucleotide comprising the nucleotide sequence represented by SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11 under highly stringent conditions A polynucleotide in which the polypeptide encoded by the polynucleotide has a cell growth-promoting activity or survival-sustaining activity.
(32)本発明の第 32の態様は、「上記(31)に記載の方法により得られた細胞」である  (32) A thirty-second aspect of the present invention is “the cell obtained by the method according to (31) above”.
(33)本発明の第 33の態様は、「以下の(a)又は (b)に示されるポリペプチド。 (33) A thirty-third aspect of the present invention is “a polypeptide shown in the following (a) or (b).
(a)配列番号 8、配列番号 10又は配列番号 12で表されるアミノ酸配列からなるポリべ プチド (b)配列番号 8、配列番号 10又は配列番号 12で表されるアミノ酸配列において、 1 若しくは数個のアミノ酸の置換、欠失若しくは挿入をもつアミノ酸配列からなり、かつ、 細胞の増殖促進活性または生存維持活性を有するポリペプチド」である。 (a) a polypeptide comprising the amino acid sequence represented by SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12 (b) in the amino acid sequence represented by SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12, consisting of an amino acid sequence having one or several amino acid substitutions, deletions or insertions, and cell growth promoting activity or A polypeptide having a life-sustaining activity ".
(34)本発明の第 34の態様は、「以下の(a)又は (b)に示されるポリヌクレオチド。 (34) A thirty-fourth aspect of the present invention is “the polynucleotide shown in the following (a) or (b).
(a)配列番号 7、配列番号 9又は配列番号 11で表されるヌクレオチド配列からなるポ リヌタレ才チド (a) Polytale talent comprising the nucleotide sequence represented by SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11
(b)配列番号 7、配列番号 9又は配列番号 1 1で表されるヌクレオチド配列からなるポ リヌクレオチドの相補鎖と高ストリンジヱントな条件下でハイブリダィズするポリヌクレオ チドであって、該ポリヌクレオチドがコードするポリペプチドが細胞の増殖促進活性ま たは生存維持活性を有するポリヌクレオチド」である。  (b) a polynucleotide that hybridizes with a complementary strand of a polynucleotide consisting of the nucleotide sequence represented by SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11 under highly stringent conditions, and encoded by the polynucleotide A polynucleotide in which the polypeptide has a cell growth-promoting activity or survival-sustaining activity.
発明の効果  The invention's effect
[0009] 本発明により、 cgrl lタンパク質の成熟形態のアミノ酸配列が明らかとなったことか ら、各種疾患の治療又は診断等に有効な組成物又は方法の提供が可能となる。  [0009] According to the present invention, since the amino acid sequence of the mature form of the cgrl l protein has been clarified, it is possible to provide a composition or method effective for the treatment or diagnosis of various diseases.
[0010] 本発明に係る組成物を用いることにより、癌の予防及び/又は治療に有効な医薬 の開発が可能となる。 [0010] By using the composition according to the present invention, it is possible to develop a pharmaceutical effective for the prevention and / or treatment of cancer.
[0011] 本発明に係る組成物を用いることにより、血流障害に関わる種々の疾患の予防及 び/又は治療に有効な医薬の開発が可能となる。  [0011] By using the composition according to the present invention, it becomes possible to develop a pharmaceutical effective for the prevention and / or treatment of various diseases related to blood flow disorders.
[0012] 本発明に係る組成物を用いることにより、高血圧症、低血圧症などの予防及び Z又 は治療に有効な医薬の開発が可能となる。  [0012] By using the composition according to the present invention, it becomes possible to develop a pharmaceutical effective for the prevention and Z or treatment of hypertension, hypotension and the like.
[0013] 本発明に係る組成物を用いることにより、発育障害などの予防及び Z又は治療に 有効な医薬の開発が可能となる。  [0013] By using the composition according to the present invention, it becomes possible to develop a pharmaceutical effective for prevention and Z or treatment of developmental disorders.
[0014] 本発明に係る組成物を用いることにより、神経疾患などの予防及び Z又は治療に 有効な医薬の開発が可能となる。  [0014] By using the composition according to the present invention, it becomes possible to develop a pharmaceutical effective for prevention and Z or treatment of neurological diseases and the like.
[0015] 本発明に係る組成物を用いることにより、培養細胞の生存維持などの工業的応用 に供される製品の開発、又は農学的な応用分野における発展に寄与することが可能 となる。  [0015] By using the composition according to the present invention, it is possible to contribute to the development of products used for industrial applications such as maintaining the survival of cultured cells, or to development in agricultural applications.
[0016] 本発明に係る組成物は、種々の細胞の分泌機能と強く関与する cgrl l遺伝子また はその産物の検出手段を包含していることから、該遺伝子またはその産物の生体内 、例えば、血中における存在濃度などを測定することにより、生体の老化、疲労など の度合いを検査するための製品などの開発が可能となる。 [0016] The composition according to the present invention includes means for detecting the cgrl gene or its product, which is strongly involved in the secretory function of various cells. For example, by measuring the concentration in the blood, it becomes possible to develop a product for examining the degree of aging and fatigue of a living body.
図面の簡単な説明 Brief Description of Drawings
[図 1]下垂体由来細胞株を使用して二次元電気泳動を行い、ホルモン産生細胞(Mt T/S)で産生され、非ホルモン産生細胞(MtTZEB— 3)では産生されなレ、タンパ ク質 (spot2)を同定した結果を示す。 [Fig. 1] Two-dimensional electrophoresis using a pituitary-derived cell line produced by hormone-producing cells (Mt T / S) but not by hormone-producing cells (MtTZEB-3) The result of identifying the quality (spot2) is shown.
[図 2]Spot2のトリプシン分解ペプチドの質量分析パターンを示す。このスポットが cgrl 1遺伝子産物に相当することが明らかとなった。  FIG. 2 shows a mass spectrometric pattern of a tryptic peptide of Spot2. This spot was found to correspond to the cgrl 1 gene product.
[図 3]ラットおよびヒト由来の cgrl 1タンパク質のアミノ酸配列のァライメントを示す。  FIG. 3 shows alignment of amino acid sequences of rat and human derived cgrl 1 proteins.
[図 4]シグナルペプチド予測を行う SOSUIsignal (http: //sosui. proteome. bio . tuat. ac. jp/ sosuisignal/ sosuisignal一 submit, ntml)によって予測 れたフ ット cgrl 1タンパク質のシグナルペプチド配列を示す。 [Figure 4] The signal peptide sequence of the foot cgrl 1 protein predicted by SOSUIsignal (http://sosui.proteome.bio.tuat.ac.jp/sosuisignal/sosuisignal1 submit, ntml) for signal peptide prediction Show.
[図 5]フット (Rattus norvegicus)、マウス (Mus musculus)、ヒト (Homo sapien s)由来の cgrl 1タンパク質のアミノ酸配列のァライメントを行レ、、分泌された cgrl 1タ ンパク質の N末端アミノ酸解析から明らかとなった実際のシグナル配列(下線部)を示 す。 R2及び H2は、各々、ラットとヒトの antigenic siteを示す。  [Fig.5] Alignment of amino acid sequence of cgrl 1 protein from foot (Rattus norvegicus), mouse (Mus musculus), human (Homo sapien s), and N-terminal amino acid analysis of secreted cgrl 1 protein Shows the actual signal sequence (underlined) as revealed by R2 and H2 represent rat and human antigenic sites, respectively.
[図 6]cgrl 1プロモーター領域に見出された低酸素反応エレメントの位置を示す。 (A )は、 HIF—1ひ結合コンセンサス配列を、(B)は、 cgrl 1遺伝子プロモーター領域に 存在する低酸素反応エレメントの位置を示す。 FIG. 6 shows the position of a hypoxic response element found in the cgrl 1 promoter region. (A) shows the HIF-1 binding consensus sequence, and (B) shows the position of the hypoxia response element present in the promoter region of the cgrl 1 gene.
[図 7]下垂体由来のホルモン産生細胞(MtTZS細胞)とホルモン非産生細胞(MtT /Se細胞、 MtTZSM細胞、 MtT/E_ B3細胞)において、定量 PCRを行い cgrl 1の細胞内 mRNAの存在量を測定した結果を示す。  [Fig. 7] Quantitative PCR was performed on pituitary-derived hormone-producing cells (MtTZS cells) and non-hormone-producing cells (MtT / Se cells, MtTZSM cells, MtT / E_B3 cells). The result of having measured is shown.
[図 8]cgrl lタンパク質に対する抗体を用いて免疫組織化学染色を行った結果を示 す。 Aは抗 cgrl 1抗体を用いて、 Bは抗成長ホルモン抗体を用いて MtTZS細胞を 染色した図である。  FIG. 8 shows the results of immunohistochemical staining using an antibody against cglrl protein. A shows staining of MtTZS cells using anti-cgrl 1 antibody and B using anti-growth hormone antibody.
[図 9]下垂体 (A)、下垂体前葉拡大図(B)、大腸 (C)、成熟卵胞の外卵胞膜 (D)に 対し、抗 cgrl 1抗体を用いて免疫組織染色を行った結果を示す。 (A)の、 PN, PI, PDは、それぞれ、下垂体後葉、下垂体中葉、下垂体前葉を示す。 [図 10]cgrl l遺伝子導入のためのベクターのコンストラクトを示す。 [Fig. 9] Results of immunohistochemical staining using anti-cgrl 1 antibody on the pituitary gland (A), enlarged view of the anterior pituitary gland (B), large intestine (C), and outer follicle membrane of mature follicle (D) Indicates. In (A), PN, PI, and PD represent the posterior pituitary, the middle pituitary, and the anterior pituitary, respectively. FIG. 10 shows a vector construct for cgrl l gene transfer.
[図 ll]cgrl l遺伝子を MtT/E— B3細胞(A)および MtT/Se (C)に導入し、抗 eg rl lタンパク質抗体を用いて免疫染色した結果を示す。いずれもゴルジ装置に陽性 反応が認められた。これに対しベクターのみを導入した MtT/E_B3細胞(B)およ び MtT/Se細胞(D)では陽性反応は認められない。 [Figure ll] shows the result of immunostaining using the anti-egrl protein antibody after introducing the cgrl gene into MtT / E-B3 cells (A) and MtT / Se (C). In both cases, a positive reaction was observed in the Golgi apparatus. In contrast, no positive reaction was observed in MtT / E_B3 cells (B) and MtT / Se cells (D) into which only the vector was introduced.
園 12]cgrl 1タンパク質を強制発現した細胞の細胞抽出物、培養上清及び MtT/S 細胞の細胞抽出物に対して抗 cgrl lタンパク質抗体を用いてウェスタンブロッテイン グを行った結果を示す。 Fig. 12] Shows the results of Western blotting using anti-cgrl protein antibodies on cell extracts, culture supernatants, and cell extracts of MtT / S cells forcibly expressing cgrl 1 protein.
[図 13]cgrl lタンパク質非発現細胞(レーン 1)と cgrl lタンパク質発現細胞(レーン 2 )の培養液をサンプルとして抗 cgrl 1タンパク質抗体を用いてゥヱスタンプロッテイン グを行った結果を示す。 M :マーカー  [Fig. 13] Shows the results of stamp stamping using anti-cgrl 1 protein antibody using cgrl l protein non-expressing cells (lane 1) and cgrl l protein expressing cells (lane 2) as samples. . M: Marker
[図 14]cgrl lを MtT/E_B3細胞に強制発現させ、得られた細胞をラット(Fischer メス 100g)の鼠径部皮下に注射(2xl06細胞/ 200 / l/ラット)し、 20日後に腫瘍 を採取しその重量を測定した。移植部位に血管の豊富な腫瘍が形成されたことを示 す図である。 [Fig.14] cgrl l is forcibly expressed in MtT / E_B3 cells, and the obtained cells are injected subcutaneously into the groin of a rat (Fischer female 100g) (2xl0 6 cells / 200 / l / rat), and 20 days later, the tumor Was collected and its weight was measured. FIG. 3 is a diagram showing that a blood vessel-rich tumor was formed at the transplantation site.
園 15]図 14に示した腫瘍を採取しその重量を測定し、腫瘍形成率(%)と腫瘍重量を 定量した結果を示す。 [15] The tumors shown in Fig. 14 were collected and weighed, and the tumor formation rate (%) and tumor weight were quantified.
[図 16]cgrl l発現ベクターを強制発現させた細胞(MtT/E— B3— So)とベクター のみを導入した細胞(MtT/E— B3— Ve)の増殖率を比較した図を示す。  FIG. 16 shows a graph comparing the growth rates of cells in which the cglrl expression vector was forcibly expressed (MtT / E-B3-So) and cells into which only the vector was introduced (MtT / E-B3-Ve).
[図 17]cgrl l強制発現 PC12細胞(クローン C〜G)と cgrl lを強制発現させていない PC12細胞(コントロール)を、無血清培地中で培養し、 8日目の生存細胞数を比較し たグラフを示す。 [Fig.17] cgrl l forced expression PC12 cells (clone C to G) and cgrl l not forcibly expressed PC12 cells (control) were cultured in serum-free medium, and the number of viable cells on day 8 was compared. The graph is shown.
園 18]cgrl l遺伝子強制発現細胞と cgrl l遺伝子を発現させていない対照細胞を 培養した培養液を 0%、 1 %、 5%、 10%加えて 5日後に細胞数をカウントした結果を 示す。 18] Shows the results of counting the number of cells after 5 days after adding 0%, 1%, 5%, 10% of the culture medium in which cgrl l gene forced expression cells and control cells not expressing cgrl l gene were cultured .
[図 19]MtT/Se細胞(cgrl l非発現、エストロゲン依存性増殖を示す)に CGR11を 含む培養液 (無血清)と含まなレ、培養液 (無血清)を加えて培養し、生存細胞の数を グラフ化したものである。 [図 20]精製した分泌型 cgrl lをラット腹腔内に注入した後、血圧の変化を示す図であ る。矢印は cgrl lタンパク質を注入した時点を示す。ここでの、血圧は、拡張血圧と 収縮血圧の平均値として示してある。 [Fig. 19] Viable cells cultured in MtT / Se cells (cglrl non-expressing, estrogen-dependent growth) with medium containing CGR11 (serum-free), medium containing serum, and medium (serum-free) Is a graph of the number of. FIG. 20 shows changes in blood pressure after injecting purified secreted cgrl into the abdominal cavity of rats. The arrow indicates the time when cgrl l protein was injected. Here, the blood pressure is shown as an average value of dilated blood pressure and systolic blood pressure.
発明を実施するための最良の形態  BEST MODE FOR CARRYING OUT THE INVENTION
[0018] 1. cgrl lポリヌクレオチド [0018] 1. cgrl l polynucleotide
本発明の組成物の有効成分の 1つである cgrl lポリヌクレオチドには、配列番号 1、 3、 5、 7、 9又は 11で表される塩基配列からなる DNAのみならず、配列番号 1、 3、 5 、 7、 9又は 11で表される塩基配列からなる DNAと相補的な塩基配列からなる DNA と高ストリンジヱント条件下でハイブリダィズし、かつ、細胞の増殖促進活性または生 存維持活性を有するポリペプチドをコードする DNAからなるものも含まれる。ここで、 「細胞」とは、 cgrl lポリペプチドが作用し得る細胞であればいかなる細胞であっても よぐ例えば、動物細胞、好ましくは、ヒト、ラット、マウス、ゥシ、ゥマ、ヒッジ、ニヮトリ、 ィヌ、ネコなど由来の細胞である。  The cgrl polynucleotide which is one of the active ingredients of the composition of the present invention includes not only DNA consisting of the base sequence represented by SEQ ID NO: 1, 3, 5, 7, 9 or 11, but also SEQ ID NO: 1, It hybridizes under high stringent conditions with DNA consisting of a base sequence complementary to DNA consisting of the base sequence represented by 3, 5, 7, 9 or 11 and has cell growth promoting activity or survival maintenance activity. Also included are those consisting of DNA encoding a polypeptide. Here, the “cell” may be any cell as long as the cgrl polypeptide can act, for example, animal cell, preferably human, rat, mouse, urushi, horse, hidge. , Cells from chickens, birds, cats, etc.
また、本発明のポリヌクレオチドには、本発明の「遺伝子」も含まれる。本発明の遺 伝子とは、 cDNAおよびゲノム DNAも含まれ、該遺伝子のェクソン、イントロンのみな らず、プロモーター、ェンノヽンサ一などの転写調節領域も含まれる。本発明のポリヌク レオチドとしては、 DNA、 RNAが含まれ、二本鎖であっても一本鎖であってもよい、 二本鎖の場合は、二本鎖 DNA、二本鎖 RNA又は DNAと RNAとのハイブリッドでも よい。  The polynucleotide of the present invention also includes the “gene” of the present invention. The gene of the present invention includes cDNA and genomic DNA, and includes not only exons and introns of the gene, but also transcriptional regulatory regions such as a promoter and an enzyme. The polynucleotide of the present invention includes DNA and RNA, and may be double-stranded or single-stranded. In the case of double-stranded, double-stranded DNA, double-stranded RNA or DNA and It may be a hybrid with RNA.
[0019] 配列番号 1、 3、 5、 7、 9又は 11で表わされる塩基配列を含有するポリヌクレオチドと ストリンジヱントな条件下でハイブリダィズできる DNAとしては、配列番号 1、 3、 5、 7、 9又は 11で表わされる塩基配列と好ましくは約 70%以上、より好ましくは約 80%, 81 %, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93 %, 94%, 95%, 96%, 97%, 98%,最も好ましくは約 99%のポリヌクレオチド酉己歹 相同性を有する塩基配列を含有する DNA等が挙げられる。  [0019] DNA that can hybridize with the polynucleotide containing the nucleotide sequence represented by SEQ ID NO: 1, 3, 5, 7, 9, or 11 under stringent conditions includes SEQ ID NO: 1, 3, 5, 7, 9, or Preferably about 70% or more, more preferably about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90% , 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, most preferably about 99% of polynucleotides that contain a homologous nucleotide sequence. It is done.
[0020] ここで、ストリンジェントな条件とは、当業者によって容易に決定されるハイブリダィ ゼーシヨン条件のことで、一般的にプローブ長、洗浄温度、及び塩濃度に依存する 経験的な計算である。一般に、プローブが長くなると適切なアニーリングのための温 度が高くなり、プローブが短くなると温度は低くなる。ノ、イブリツド形成は、一般的に、 相補的鎖がその融点に近いがそれより低い環境に存在する場合における変性 DNA の再ァニールする能力に依存する。 Here, the stringent condition is a hybridization condition that is easily determined by those skilled in the art, and is generally an empirical calculation that depends on the probe length, the washing temperature, and the salt concentration. In general, the longer the probe, the higher the temperature for proper annealing. The temperature decreases as the temperature increases and the probe becomes shorter. However, hybrid formation generally depends on the ability of the denatured DNA to reanneal when the complementary strand is present in an environment near its melting point but below it.
具体的には、例えば、低ストリンジヱントな条件として、ハイブリダィゼーシヨン後のフ イノレターの洗净段皆 ίこおレ、て、 37°C〜42°Cの温度条件下、 0. 1 X SSC, 0. 1%S DS溶液中で洗浄することなどが上げられる。また、高ストリンジェントな条件として、例 えば、洗浄段階において、 65°C、 5 X SSCおよび 0. 1 %SDS中で洗浄することなど が挙げられる。ストリンジェントな条件をより高くすることにより、相同性の高いポリヌク レ才チドを得ること力 Sできる。  Specifically, for example, as a low-stringent condition, the washing of the post-hybridization of the final letter should be performed at a temperature of 37 ° C to 42 ° C, 0.1 X For example, washing in SSC, 0.1% SDS solution. In addition, examples of highly stringent conditions include washing in 65 ° C, 5 X SSC and 0.1% SDS in the washing step. By making stringent conditions higher, the ability to obtain highly homologous polynucleotide tides can be achieved.
[0021] cgrl lポリヌクレオチドは、例えば、下垂体由来の細胞であってホルモン産生を行 つている細胞から取得することができる。例えば、ホルモンを産生する細胞およびそ の対照としてホルモンを産生しない細胞中に存在する全タンパク質を抽出し、各々を 二次元電気泳動などを行って、その泳動プロフィールを比較し、ホルモン産生細胞 においてのみ確認されるタンパク質のスポットから、質量分析法などを用いてアミノ酸 配列情報を取得する。次に、得られたアミノ酸配列情報に基づいて、適当な cDNAラ イブラリーなどから、 cgrl lポリヌクレオチド配列を有する DNAを単離することができ る。 [0021] The cgrl polynucleotide can be obtained from, for example, a cell derived from the pituitary gland and producing hormone. For example, extract all proteins present in hormone-producing cells and non-hormone-producing cells as controls, and perform two-dimensional electrophoresis, etc., and compare their migration profiles, and only in hormone-producing cells. Amino acid sequence information is obtained from confirmed protein spots using mass spectrometry. Next, based on the obtained amino acid sequence information, DNA having a cgrl polynucleotide sequence can be isolated from an appropriate cDNA library or the like.
[0022] また、すでに公知となっている cgrl l遺伝子の配列情報をデータベースなどから取 得し、その配列情報に基づいて、所望の組織または動物種由来の cgrl l遺伝子を 得ることも可能である。 cgrl l遺伝子のクローニングは当該技術分野における通常の 知識に基づいて行うことができる。該遺伝子は、該遺伝子を発現している細胞より cD NAライブラリーを調製し、定法のスクリーニング方法により取得することができる。あ るいは、該遺伝子が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、該遺伝子配列に基づレ、て PCR用プライマーを調製して cDNAを増幅 させることにより該 cDNAを取得してもよい。  [0022] It is also possible to obtain sequence information of a known cgrl l gene from a database or the like and obtain a cgrl l gene derived from a desired tissue or animal species based on the sequence information. . Cloning of the cgrl gene can be carried out based on ordinary knowledge in the art. The gene can be obtained by preparing a cDNA library from cells expressing the gene and using a conventional screening method. Alternatively, RNA is prepared from cells expressing the gene, cDNA is synthesized by reverse transcriptase, and then PCR primers are prepared based on the gene sequence to amplify the cDNA. The cDNA may be obtained.
[0023] 本発明における cgrl lポリヌクレオチドの取得に使用可能な細胞は、 cgrl lポリべ プチドの mRNAが発現してレ、る細胞であれば如何なる細胞も使用可能である力 例 えば、脳下垂体のホルモン産生細胞などが好ましい。また、前記細胞が由来する動 物は、本発明の遺伝子を有するものであれば如何なる動物(例えば、ヒト、ラットなど) であってもよい。 [0023] The cell that can be used for obtaining the cgrl polynucleotide in the present invention is any cell that can express any mRNA that expresses cgrl polypeptide mRNA. Pituitary hormone-producing cells are preferred. In addition, the movement from which the cells are derived The product may be any animal (for example, human, rat, etc.) as long as it has the gene of the present invention.
cgrl lポリヌクレオチドを取得するための mRNAの調製、 cDNAの調製には、 mR NA Purification Kit (Pnarmasia社)、 AMV Reverse Transcriptase First - strand cDNA Synthesis Kit (生化学工業社)、などの市販のキットを用いて あよい。  cgrl l To obtain mRNA for obtaining polynucleotides and cDNA, use commercially available kits such as mRNA Purification Kit (Pnarmasia) and AMV Reverse Transcriptase First-strand cDNA Synthesis Kit (Seikagaku Corporation). Use it.
[0024] 2. cgrl lポリペプチド及びその部分ペプチド  [0024] 2. cgrl l polypeptide and partial peptide thereof
本発明における cgrl lポリペプチドは、配列番号 2、 4、 6、 8、 10又は 12で表される アミノ酸配列と同一又は実質的に同一のアミノ酸配列を含むポリペプチドである。ここ で、「実質的に同一のアミノ酸配列を含むポリペプチド」とは、配列番号 2、 4、 6、 8、 1 0又は 12で表わされるアミノ酸配列と約 60%以上、好ましくは約 70%以上、より好ま しくは約 80%, 81 %, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90% , 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%,最も好ましくは約 99%の アミノ酸同一性を有するアミノ酸配列を含み、かつ細胞の増殖促進又は活性を有す るポリペプチドである。ここで、「細胞」とは、 cgrl lポリペプチドが作用し得る細胞であ ればいかなる細胞であってもよぐ例えば、動物細胞、好ましくは、ヒト、ラット、マウス 、ゥシ、ゥマ、ヒッジ、ニヮトリ、ィヌ、ネコなど由来の細胞である。  The cgrl polypeptide in the present invention is a polypeptide comprising the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 2, 4, 6, 8, 10 or 12. Here, the “polypeptide containing substantially the same amino acid sequence” means about 60% or more, preferably about 70% or more, of the amino acid sequence represented by SEQ ID NO: 2, 4, 6, 8, 10, or 12. More preferably, about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94% 95%, 96%, 97%, 98%, most preferably a polypeptide having an amino acid sequence having about 99% amino acid identity and having cell growth promotion or activity. Here, the “cell” may be any cell as long as it can act on cgrl polypeptide, for example, animal cell, preferably human, rat, mouse, urushi, horse, It is a cell derived from Hedge, chicken, dog, cat, etc.
なお、本明細書中において、特に断らない限り、「ポリペプチド」と「タンパク質」は同 じ意味で用レ、るものとする。  In the present specification, “polypeptide” and “protein” are used in the same meaning unless otherwise specified.
[0025] あるいは、配列番号 2、 4、 6、 8、 10又は 12で表わされるアミノ酸配列と実質的に同 一のアミノ酸配列を含むポリペプチドとしては、配列番号 2、 4、 6、 8、 10又は 12で表 わされるアミノ酸配列中の 1又は数個(好ましくは、:!〜 30個程度、より好ましくは 1〜 10個程度、さらに好ましくは:!〜 5個)のアミノ酸が欠失、置換若しくは付加されたアミ ノ酸配列からなり、かつ細胞の増殖促進又は活性を有するポリペプチドである。ここ で、「細胞」とは、 cgrl lポリペプチドが作用し得る細胞であればいかなる細胞であつ てもよく、例えば、動物細胞、好ましくは、ヒト、ラット、マウス、ゥシ、ゥマ、ヒッジ、ニヮト リ、ィヌ、ネコなど由来の細胞である。  [0025] Alternatively, as a polypeptide comprising an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 2, 4, 6, 8, 10, or 12, SEQ ID NO: 2, 4, 6, 8, 10 Or one or several (preferably:! To about 30, more preferably about 1 to 10, more preferably :! to 5) amino acids in the amino acid sequence represented by 12 are deleted, It is a polypeptide having an amino acid sequence substituted or added and having cell growth promotion or activity. Here, the “cell” may be any cell as long as the cglrl polypeptide can act, for example, an animal cell, preferably a human, rat, mouse, urushi, horse, hidge. It is a cell derived from nitrite, nu, cat, etc.
上記アミノ酸の欠失、付加及び置換は、単離した天然ポリペプチドに存在していて もよぐまた、本発明のポリペプチドをコードする遺伝子を、当該技術分野で公知の手 法によって改変することによって新たに導入したものでもよい。例えば、特定のァミノ 酸残基の置換は、市販のキット(例えば、 MutanTM— G (TAKARA社)、 MutanTM— K (TAKARA社))等を使用し、 Guppedduplex法や Kunkel法等の公知の方法あるレ、 はそれらに準じる方法により塩基の置換を行なうことによって達成することができる。 The above amino acid deletions, additions and substitutions are present in isolated natural polypeptides. Alternatively, a gene newly introduced by modifying a gene encoding the polypeptide of the present invention by a method known in the art. For example, the substitution of a specific amino acid residue is a known method such as Guppedduplex method or Kunkel method using a commercially available kit (for example, MutanTM-G (TAKARA), MutanTM-K (TAKARA)). Can be achieved by substituting the base by a method analogous thereto.
[0026] また、本発明における cgrl lポリペプチドの C末端は、通常カルボキシル基(_ C〇 OH)又はカルボキシレート(_ C〇0 _ )であるが、当該カルボキシル基は、アミド(一 CONH2)やエステル(_ C〇OR)等に化学修飾されていてもよレ、。ここで、エステル 中の Rとしては、 C1— 6アルキル基(例えば、メチノレ、ェチル、 n—プロピル、イソプロ ピル、 n—ブチル)、 C3— 8シクロアルキル基(例えば、シクロペンチル、シクロへキシ ノレ)、 C1— 6ァリール基(例えば、フエ二ノレ、 ひ一ナフチル)、フエニル一C1— 2ァノレ キル基(例えば、ベンジル、フエネチル)、 α—ナフチル— C1— 2アルキル基(例えば 、 a ナフチルメチル)等が挙げられる。その他、経口用エステルとして汎用されてい るビバロイルォキシメチルエステルとすることも可能である。本発明のポリペプチドが C 末端以外にもそのポリペプチド鎖中にカルボキシル基を有する場合には、当該カル ボキシル基がアミド化又はエステルイ匕されているものも本発明のポリペプチドに含ま れる。この場合のエステルとしては上記の各エステルが挙げられる。同様に、本発明 のポリペプチドの N末端は、通常アミノ基(一NH2)であるが、当該アミノ基は、ホルミ ル基、ァセチル基等の C1 6ァシル基等で化学修飾されていてもよい。その他、 N 端側が生体内で切断され生成したダルタミル基がピログノレタミン酸化したものや、分 子内のアミノ酸の側鎖上の置換基(例えば、 OH、— SH、アミノ基、イミダゾール基 、インドール基、グァニジノ基など)が適当な官能基 (例えば、ホルミル基、ァセチル 等)で化学修飾されているものや糖鎖の結合しているものも本発明のポリペプチドに 含まれる。 [0026] The C-terminus of the cgrl polypeptide in the present invention is usually a carboxyl group (_C0OH) or a carboxylate (_C00_), and the carboxyl group is an amide (one CONH2). Or may be chemically modified with ester (_C0OR). Here, as R in the ester, C1-6 alkyl group (for example, methinole, ethyl, n-propyl, isopropyl, n-butyl), C3-8 cycloalkyl group (for example, cyclopentyl, cyclohexylene) , C1-6 aryl group (for example, phenylolene, mononaphthyl), phenyl-1-C1-2anolyl group (for example, benzyl, phenethyl), α-naphthyl-C1-2 alkyl group (for example, a naphthylmethyl) Etc. In addition, it is possible to use a bivalyloxymethyl ester which is widely used as an oral ester. When the polypeptide of the present invention has a carboxyl group in the polypeptide chain other than at the C-terminus, the polypeptide of the present invention includes those in which the carboxyl group is amidated or esterified. Examples of the ester in this case include the above-mentioned esters. Similarly, the N-terminus of the polypeptide of the present invention is usually an amino group (one NH2), but the amino group may be chemically modified with a C16 acyl group such as a formyl group or a acetyl group. . In addition, the daltamyl group generated by cleaving the N-terminal side in vivo is pyrologretamine oxidized, or a substituent on the side chain of an amino acid in the molecule (for example, OH, —SH, amino group, imidazole group, indole group, Polypeptides of the present invention include those in which a guanidino group or the like) is chemically modified with an appropriate functional group (for example, formyl group, acetyl), or a sugar chain.
[0027] 本発明における cgrl lポリペプチド中の部分アミノ酸配列を含むペプチド(部分ぺ プチドともいう)も本発明の組成物に含まれてもよい。また、部分ペプチドは本発明に おける抗体を調製するための抗原にもなり得る。すなわち、本発明の部分ペプチドは 、配列番号 2、配列番号 4又は配列番号 6で表されるアミノ酸配列と同一又は実質的 に同一のアミノ酸配列の一部のアミノ酸配列を含むものである限り、如何なるものであ つてもよい。 [0027] A peptide containing a partial amino acid sequence in the cglrl polypeptide of the present invention (also referred to as a partial peptide) may be included in the composition of the present invention. The partial peptide can also be an antigen for preparing the antibody of the present invention. That is, the partial peptide of the present invention is the same or substantially the same as the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4 or SEQ ID NO: 6. As long as it contains a part of the amino acid sequence of the same amino acid sequence.
本発明の部分ペプチドを構成するアミノ酸数は、少なくとも 10個以上、好ましくは 3 0個以上、より好ましくは 80個以上である。通常、本発明の部分ペプチドの C末端は カルボキシル基(_C〇OH)、 N末端はァミノ基(_NH2)である力 化学修飾されて いてもよい。  The number of amino acids constituting the partial peptide of the present invention is at least 10 or more, preferably 30 or more, more preferably 80 or more. Usually, the partial peptide of the present invention may be mechanically modified such that the C-terminus is a carboxyl group (_COH) and the N-terminus is an amino group (_NH2).
[0028] 本発明における cgrl lポリペプチド又はその塩は、 cgrl lポリペプチドを発現して いるヒトゃ動物(例えば、ラット、マウスなど)由来の培養細胞又は組織から、当該分野 における通常の技術により抽出 ·分離することができ、あるいは後述のように本発明の ポリペプチドをコードする DNAを発現可能な状態で含む形質転換体を培養すること により、該培養物から抽出'分離することによつても調製することができる。ヒトゃ動物 の組織又は細胞から調製する場合、ヒトゃ動物の組織または細胞をホモジナイズ後、 酸等で抽出を行ない、得られた抽出液を疎水クロマトグラフィー、逆相クロマトグラフィ 一、イオン交換クロマトグラフィー等の各種クロマトグラフィーを組み合わせることによ り単離精製することができる。  [0028] The cgrl l polypeptide or a salt thereof in the present invention is obtained from a cultured cell or tissue derived from a human animal (eg, rat, mouse, etc.) expressing the cgrl l polypeptide by a conventional technique in the art. Extraction and isolation from the culture can be performed by culturing a transformant that can be extracted or isolated, or can be expressed in a state in which the DNA encoding the polypeptide of the present invention can be expressed as described later. Can also be prepared. When preparing from human tissues or cells, homogenize human tissues or cells and extract with acid, etc., and use the resulting extract for hydrophobic chromatography, reverse phase chromatography, ion exchange chromatography, etc. Isolation and purification can be achieved by combining these various chromatographies.
[0029] また、本発明における部分ペプチドまたはその塩は、公知のペプチド合成法又は c grl lポリペプチドを適当なぺプチダーゼ(例えば、トリプシン、キモトリブシン、アルギ ニルエンドべプチダーゼ)で切断することによって製造することができる。ペプチド合 成法としては、例えば、固相合成法、液相合成法のいずれによってもよい。すなわち 、本発明のポリペプチドを構成し得る部分ペプチドもしくはアミノ酸と残余部分とを縮 合させ、生成物が保護基を有する場合は保護基を脱離することにより目的のぺプチ ドを製造することができる(例えば、 Bondanszky等, 1996; Schroeder等, 1965を参照の こと)。合成反応後は通常の精製法、例えば、溶媒抽出、蒸留、カラムクロマトグラフィ 一、高速液体クロマトグラフィー、再結晶などを組み合わせて本発明の部分ペプチド を単離精製することができる。  [0029] Further, the partial peptide or its salt in the present invention is produced by a known peptide synthesis method or cgrrl polypeptide cleaved with an appropriate peptidase (eg, trypsin, chymotrypsin, arginyl endopeptidase). be able to. As the peptide synthesis method, for example, either a solid phase synthesis method or a liquid phase synthesis method may be used. That is, the desired peptide is produced by condensing the partial peptide or amino acid that can constitute the polypeptide of the present invention and the remaining part, and removing the protecting group when the product has a protecting group. (See, for example, Bondanszky et al., 1996; Schroeder et al., 1965). After the synthesis reaction, the partial peptide of the present invention can be isolated and purified by combining ordinary purification methods such as solvent extraction, distillation, column chromatography, high performance liquid chromatography, recrystallization and the like.
上記の方法で得られる cgrl lポリペプチド又はその部分ペプチドが遊離体である 場合は、公知の方法によって適当な塩に変換することができる、塩で得られた場合は 、公知の方法によって遊離体に変換することができる。 [0030] 3. cgrl l糸且換えベクター When the cgrl polypeptide obtained by the above method or a partial peptide thereof is a free form, it can be converted to an appropriate salt by a known method. When obtained by a salt, the free form is obtained by a known method. Can be converted to [0030] 3. cgrl l thread changeable vector
cgrl 1ポリヌクレオチド又はその一部を組み込んだ組換えベクターは、適切なベタ ターに cgrl lポリヌクレオチド又はその一部を連結することにより得ることができる。該 組換えベクターは、クローニング用に供される場合には、宿主中で複製可能なもので あれば特に限定されない。また、 cgrl 1ポリペプチド又はその一部を発現するための ベクターとしては、宿主中で複製可能なものであって、該ポリペプチドをコードする D NA断片を発現させることができるプロモーターなどを有するものが使用可能である。  A recombinant vector incorporating the cgrl 1 polynucleotide or a part thereof can be obtained by ligating the cgrl polynucleotide or a part thereof to an appropriate vector. The recombinant vector is not particularly limited as long as it can be replicated in a host when it is used for cloning. Moreover, as a vector for expressing cgrl 1 polypeptide or a part thereof, a vector capable of replicating in a host and having a promoter capable of expressing a DNA fragment encoding the polypeptide, etc. Can be used.
[0031] 使用可能なベクターとしては、例えば、プラスミド DNA、ファージ DNA等が挙げら れる。プラスミド DNAとしては、大腸菌由来のプラスミド(例えば pBR322、 pBR325 、 pUC118、 pUC119、 pUC18、 pUC19、 pCBD— C等)、枯草菌由来のプラスミド (例えば pUB110、 pTP5、 pC194等)、酵母由来のプラスミド(例えば ΥΕρ13、 ΥΕρ 24、 YCp50、 YIp30等)などが挙げられ、ファージ DNAとしては λファージ等が挙 げられる。さらに、レトロウイルス、ワクシニアウィルスなどの動物ウィルス、バキュロウィ ルス、トガウィルスなどの昆虫ウィルスベクターを用いることもできる。  [0031] Examples of vectors that can be used include plasmid DNA and phage DNA. Plasmid DNA includes plasmids derived from E. coli (eg, pBR322, pBR325, pUC118, pUC119, pUC18, pUC19, pCBD-C, etc.), plasmids derived from Bacillus subtilis (eg, pUB110, pTP5, pC194, etc.), and yeast-derived plasmids (eg, ΥΕρ13, ΥΕρ24, YCp50, YIp30, etc.), and phage DNA includes λ phage. Furthermore, animal viruses such as retrovirus and vaccinia virus, and insect virus vectors such as baculovirus and toga virus can also be used.
[0032] 本発明で用いられるプロモーターとしては、遺伝子の発現に用いる宿主に対応して 適切なプロモーターであれば特に限定されなレ、。  [0032] The promoter used in the present invention is not particularly limited as long as it is an appropriate promoter corresponding to the host used for gene expression.
例えば、動物細胞を宿主として用いる場合は、 SR aプロモーター、 CMVプロモー ター、 SV40プロモーター、 LTRプロモーター、 HSV— TKプロモーター、 EF— 1 α プロモーター等が挙げられる。  For example, when an animal cell is used as a host, SRa promoter, CMV promoter, SV40 promoter, LTR promoter, HSV-TK promoter, EF-1α promoter and the like can be mentioned.
宿主が大腸菌である場合には、 tacプロモーター、 ΐι·ρプロモーター、 lacプロモータ 一、 recAプロモーター、 え PLプロモーター、 lppプロモーター等力 宿主が枯草菌で ある場合には、 SPOlプロモーター、 SP02プロモーター、 penPプロモーター等が挙 げられる。  If the host is Escherichia coli, tac promoter, · ι · ρ promoter, lac promoter, recA promoter, e.g. PL promoter, lpp promoter, etc. If the host is Bacillus subtilis, SPOl promoter, SP02 promoter, penP promoter Etc.
宿主が酵母である場合には、 PH05プロモーター、 PGKプロモーター、 GAPプロ モーター、 ADHプロモーター等が挙げられる。  When the host is yeast, PH05 promoter, PGK promoter, GAP promoter, ADH promoter and the like can be mentioned.
宿主が昆虫細胞である場合は、ポリヘドリンプロモーター、 P10プロモーターなどが 好ましい。  When the host is an insect cell, polyhedrin promoter, P10 promoter, etc. are preferable.
[0033] 本発明の組換えベクターには cgrl 1ポリペプチド又はその一部のコード化配歹 1J、プ 口モーター配列以外にも、選択マーカー、ターミネータ一、ェンハンサー、スプライシ ングシグナル、ポリ A付加シグナル、リボソーム結合配列(SD配列)、 SV40複製起点 (SV40ori)などを連結することができる。 [0033] The recombinant vector of the present invention includes a cgrl 1 polypeptide or a portion of a coding cage 1J, In addition to the oral motor sequence, a selection marker, terminator, enhancer, splicing signal, poly A addition signal, ribosome binding sequence (SD sequence), SV40 origin of replication (SV40ori), etc. can be linked.
選択マーカーとしては、限定はしないが、ハイグロマイシン耐性マーカー(Hygr)、 ジヒドロ葉酸還元酵素遺伝子(dMr)、アンピシリン耐性遺伝子 (Ampr)、カナマイシ ン耐性遺伝子 (Kanr)、ネオマイシン耐性遺伝子 (Neor, G418)などが利用可能 である。  Selection markers include, but are not limited to, hygromycin resistance marker (Hygr), dihydrofolate reductase gene (dMr), ampicillin resistance gene (Ampr), kanamycin resistance gene (Kanr), neomycin resistance gene (Neor, G418) Etc. are available.
また、組換えタンパク質の単離 ·精製を容易にするなどの目的で、本発明のポリぺ プチドの N末端側に適当なシグナル配列を付加してもよい。  For the purpose of facilitating the isolation and purification of the recombinant protein, an appropriate signal sequence may be added to the N-terminal side of the polypeptide of the present invention.
宿主が大腸菌である場合にはアルカリホスファターゼシグナル、〇mPAシグナルな どが利用可能であり、宿主が枯草菌である場合にはひ—アミラーゼシグナル配歹 IJ、ズ プチリスシグナル配列などが利用可能であり、宿主が酵母である場合には、 ひ因子シ ダナル配歹 lj、インベルターゼシグナル配列などが利用可能であり、宿主が動物細胞 である場合には、例えば、インシュリンシグナル配歹 IJ、 a—インターフェロンシグナノレ 配歹 I」、抗体分子シグナル配列などが利用可能である。 Host alkaline phosphatase signal when an E. coli, etc. 〇_M P A signal is available and Wahi When the host is Bacillus subtilis - amylase signal Hai歹IJ, etc.'s Petit squirrels signal sequences available In the case where the host is yeast, the factor factorial lj, the invertase signal sequence, etc. can be used. In the case where the host is an animal cell, for example, the insulin signal distribution IJ, a —Interferon Signale I ", antibody molecule signal sequences, etc. are available.
[0034] 上述のベクターに対して cgrl lポリヌクレオチド又はその一部を挿入することは、ク ローニングされた DNAをそのまま、又は所望により制限酵素で消化して、リンカ一を 付加し、ベクター DNAの制限酵素部位又はマルチクローニングサイトに挿入すること により行うことができる。連結する DNAはその 5 '末端側に翻訳開始コドンとしての AT Gを有し、また 3 '末端側には翻訳終止コドンとしての TAA、 TGA又は TAGを有して いてもよい。これらの翻訳開始コドンや翻訳終止コドンは、適当な合成 DNAアダプタ 一を用いて付加することもできる。連結する DNAは、当該 DNA中にコードされてい る本発明のポリペプチドが宿主細胞中で発現されるようにベクターに組み込まれるこ とが必要である。 [0034] Inserting a cgrl polynucleotide or a part thereof into the above-described vector means that the cloned DNA is digested with a restriction enzyme as it is or as desired, and a linker is added to the vector DNA. It can be performed by inserting into a restriction enzyme site or a multicloning site. The DNA to be ligated may have ATG as a translation initiation codon on the 5 ′ end side and TAA, TGA or TAG as a translation termination codon on the 3 ′ end side. These translation initiation codons and translation termination codons can also be added using an appropriate synthetic DNA adapter. The DNA to be ligated needs to be incorporated into a vector so that the polypeptide of the present invention encoded in the DNA is expressed in the host cell.
以上の方法により、 cgrl lポリペプチドをコードする DNA配列を含むベクターを構 築すること力 Sできる。  By the above method, it is possible to construct a vector containing a DNA sequence encoding a cglrl polypeptide.
[0035] 4.形質転換体 [0035] 4. Transformant
4- 1. cgrl lポリペプチド発現のための形質転換体 cgrl lの形質転換体は、本発明の組換え発現ベクターを、 cgrl l遺伝子が発現し 得るように宿主中に導入することにより得ること力 Sできる。ここで、宿主としては、 cgrl 1遺伝子を発現できるものであれば特に限定されるものではない。例えば、大腸菌(E scherichia coli)等のェシヱリシァ属、枯草菌(Bacillus subtilis)等のバチルス属、シュ ードモナス'プチダ(Pseudomonas putida)等のシユードモナス属、リゾビゥム 'メリロテ ィ(Rhizobium meliloti)等のリゾビゥム属に属する細菌、サッカロミセス'セレピシェ(Sa ccharomyces cerevisiae)、ンゾサッカロ:セス -ホンへ (schizosaccharomyces pombe) 、ピキア'パストリス(Pichia pastoris)等の酵母、サル細胞 COS _ 7、 Vero、チヤィニ ーズハムスター卵巣細胞(CHO細胞)、あるいは Sf 9、 Sf 21等の昆虫細胞が挙げら れる。 4- 1. Transformants for cgrl l polypeptide expression A transformant of cgrl can be obtained by introducing the recombinant expression vector of the present invention into a host so that the cgrl l gene can be expressed. Here, the host is not particularly limited as long as it can express the cgrl 1 gene. For example, Escherichia coli and other genus Escherichia, Bacillus subtilis and other Bacillus genus, Pseudomonas putida and other shyudomonas genus, Rhizobium meliloti and other lysovivum genus Bacteria belonging to, Saccharomyces cerevisiae, Szozoaccharomyces pombe, Pichia pastoris, monkey cells COS _ 7, Vero, Chinese hamster ovary cells (CHO cells) Or insect cells such as Sf 9 and Sf 21.
[0036] 大腸菌への組換えベクターの導入方法としては、カルシウムイオンを用いる方法(C ohen等, 1972)、エレクト口ポレーシヨン法(Shigekawa及び Dower, 1988)等が利用可 能である。酵母への組換えベクターの導入方法としては、エレクト口ポレーシヨン法(B ecker等, 1990)、スフヱ口プラスト法(Hinnen等, 1978)、酢酸リチウム法(Itoh等, 1983 )等が利用可能である。動物細胞又は動物細胞への組換えベクターの導入方法とし ては、 DEAEデキストラン法(Lopata等, 1984)、エレクト口ポレーシヨン法、リン酸カル シゥム法(Chen及び Okayama, 1988)、カチオン性脂質による方法(Elroy-Stein及び Moss, 1990)等が挙げられる。  [0036] As a method for introducing a recombinant vector into Escherichia coli, a method using calcium ions (Cohen et al., 1972), an electo mouth position method (Shigekawa and Dower, 1988) and the like can be used. As a method for introducing a recombinant vector into yeast, the Elect Mouth Position method (B ecker et al., 1990), the Suguchi Higuchi plast method (Hinnen et al., 1978), the lithium acetate method (Itoh et al., 1983), etc. can be used. . Methods for introducing recombinant vectors into animal cells or animal cells include the DEAE dextran method (Lopata et al., 1984), the electopore position method, the phosphate phosphate method (Chen and Okayama, 1988), and the method using cationic lipids. (Elroy-Stein and Moss, 1990).
[0037] 4- 2.不死化又は生存維持を達成するための形質転換体  [0037] 4- 2. Transformants for achieving immortalization or survival maintenance
本発明において、 cgrl lが細胞、特に動物細胞の増殖促進活性又は生存維持活 性を有することが初めて開示された。従って、 cgrl l遺伝子を不死化または生存維持 を望む細胞に発現可能に導入することで該細胞の不死化又は生存維持を達成する こと力 Sできる。不死化又は生存維持が望まれる形質転換体を培養する際、培地として は所望の細胞が通常生存するのに適した培地であればレ、かなるものでもよぐ例え ば、約 5〜20%のゥシ胎児血清を含む MEM培地、 DMEM培地、 RPMI—1640培 地等が用いられる。また、 pHや培養の温度も所望の細胞に適した条件を設定するこ とができ、これらの条件は当業者であれば容易に設定可能である。以上のようにして 、 cgrl lポリペプチドが不死化又は生存維持が望まれる細胞中で発現されれば、該 細胞の不死化又は生存維持が達成される。 In the present invention, it has been disclosed for the first time that cgrl has cell growth promotion activity or survival maintenance activity. Therefore, by introducing the cglrl gene into cells that are desired to be immortalized or to maintain survival, it is possible to achieve immortalization or maintenance of survival of the cells. When cultivating transformants that are desired to be immortalized or remain viable, the medium can be any medium that is suitable for the desired cells to normally survive, for example, about 5 to 20%. MEM medium, DMEM medium, RPMI-1640 medium, etc. containing fetal bovine serum are used. In addition, conditions suitable for the desired cells can be set for pH and culture temperature, and these conditions can be easily set by those skilled in the art. As described above, if the cgrl polypeptide is expressed in a cell in which immortalization or survival is desired, Cell immortalization or viability maintenance is achieved.
[0038] 5. cgrl lポリペプチド及びその部分ペプチドの製造  [0038] 5. Production of cgrl l polypeptide and partial peptide thereof
本発明の組成物に含まれる cgrl lポリペプチド又はその部分ペプチドは、 cgrl l遺 伝子又はその一部の発現ベクターを導入した形質転換体を培養し、 cgrl 1ポリぺプ チド又はその一部を発現させ、培養物から該ポリペプチド又はその一部を単離するこ とにより製造すること力 Sできる。 「培養物」とは、培養上清、あるいは培養細胞若しくは 培養菌体又は細胞若しくは菌体の破砕物のいずれをも意味するものである。  The cgrl l polypeptide or a partial peptide thereof contained in the composition of the present invention is obtained by culturing a transformant introduced with an expression vector of the cgrl l gene or a part thereof, and cgrl 1 polypeptide or a part thereof. Can be produced by isolating the polypeptide or a part thereof from the culture. “Culture” means any of culture supernatant, cultured cells or cultured cells, or disrupted cells or cells.
[0039] 大腸菌や酵母菌等の微生物を宿主として得られた形質転換体を培養する培地とし ては、微生物が資化し得る炭素源、窒素源、無機塩類等を含有し、形質転換体の培 養を効率的に行うことができる培地であれば、天然培地、合成培地のいずれを用い てもよレ、。炭素源としては、グルコース、フルクトース、スクロース、デンプン等の炭水 化物、酢酸、プロピオン酸等の有機酸、エタノール、プロパノール等のアルコール類 が用いられる。窒素源としては、アンモニア、塩化アンモニゥム、硫酸アンモニゥム、 酢酸アンモニゥム、リン酸アンモニゥム等の無機酸若しくは有機酸のアンモニゥム塩 又はその他の含窒素化合物のほ力、ペプトン、肉エキス、コーンスティープリカ一等 が用いられる。無機塩類としては、リン酸第一カリウム、リン酸第二カリウム、リン酸マ グネシゥム、硫酸マグネシウム、塩化ナトリウム、硫酸第一鉄、硫酸マンガン、硫酸銅 、炭酸カルシウム等が用いられる。  [0039] As a medium for culturing a transformant obtained using a microorganism such as E. coli or yeast as a host, the medium contains a carbon source, a nitrogen source, inorganic salts and the like that can be assimilated by the microorganism. Either natural or synthetic media can be used as long as the culture can be carried out efficiently. As the carbon source, carbohydrates such as glucose, fructose, sucrose and starch, organic acids such as acetic acid and propionic acid, and alcohols such as ethanol and propanol are used. Examples of nitrogen sources include ammonia, ammonium chloride, ammonium sulfate, ammonium acetate, ammonium salts of organic acids such as ammonium salts, or other nitrogen-containing compounds, peptone, meat extract, corn steep liquor, etc. Used. Examples of inorganic salts include monopotassium phosphate, dipotassium phosphate, magnesium phosphate, magnesium sulfate, sodium chloride, ferrous sulfate, manganese sulfate, copper sulfate, and calcium carbonate.
[0040] 培養は、宿主細胞に適した条件下で行う。例えば、大腸菌を培養する際の培地とし ては、 LB培地、 M9培地等が好ましい。所望によりプロモーターを効率よく働かせる ために、イソプロピルー1 チォー β D ガラクトシド、 3 一インドリルアクリル酸の ような薬剤を加えることができる。大腸菌の場合、培養は通常約 15〜37°Cで約 3〜2 4時間行い、必要により、通気や撹拌をカ卩えることもできる。宿主が枯草菌の場合、培 養は通常約 30〜40°Cで約 6〜24時間行レ、、必要に応じて通気や撹拌を加えること あでさる。  [0040] The culture is performed under conditions suitable for the host cell. For example, as a medium for culturing Escherichia coli, LB medium, M9 medium and the like are preferable. Agents such as isopropyl-1-thio βD galactoside, 3-indolylacrylic acid can be added to make the promoter work efficiently if desired. In the case of Escherichia coli, the culture is usually carried out at about 15 to 37 ° C for about 3 to 24 hours, and if necessary, aeration and agitation can be controlled. When the host is Bacillus subtilis, cultivation is usually performed at about 30-40 ° C for about 6-24 hours, and if necessary, aeration or agitation is added.
[0041] 酵母を培養するための培地としては、 SD培地、 YPD培地があげられる。培地の p Hは約 5〜8に調整するのが好ましレ、。培養は通常約 20〜35°Cで約 24〜72時間行 レ、、必要に応じて通気や撹拌を加える。宿主が昆虫細胞または昆虫である形質転換 体を培養する際、培地としては、ゥシ血清を含むグレース昆虫培地等が挙げられる。 培地の pHは約 6. 2〜6. 4に調整するのが好ましい。培養は通常約 27°Cで約 3〜5 日間行い、必要に応じて通気や撹拌を加える。 [0041] Examples of the medium for culturing yeast include SD medium and YPD medium. It is preferable to adjust the pH of the medium to about 5-8. Incubate at about 20 to 35 ° C for about 24 to 72 hours, and if necessary, add aeration and agitation. Transformation where the host is an insect cell or insect When culturing the body, examples of the medium include Grace insect medium containing urine serum. The pH of the medium is preferably adjusted to about 6.2 to 6.4. Incubate at about 27 ° C for about 3 to 5 days, and add aeration or agitation as necessary.
[0042] 宿主が動物細胞である形質転換体を培養する際、培地としては、例えば、約 5〜2 0%のゥシ胎児血清を含む MEM培地、 DMEM培地、 RPMI— 1640培地等が用い られる。 pHは約 6〜8であるのが好ましレ、。培養は通常約 30〜40°Cで約 15〜60時 間行い、必要に応じて通気や撹拌を加える。上記培養物から cgrl lポリペプチド又 はその部分ペプチドを分離精製するには、例えば、下記の方法により行うことができ る。 [0042] When a transformant whose host is an animal cell is cultured, for example, a MEM medium, DMEM medium, RPMI-1640 medium or the like containing about 5 to 20% urine fetal serum is used. . The pH is preferably about 6-8. Incubate at about 30–40 ° C for about 15–60 hours with aeration and agitation as necessary. Separation and purification of cgrl polypeptide or partial peptide thereof from the above culture can be performed, for example, by the following method.
[0043] cgrl lポリペプチド又はその部分ペプチドを培養菌体あるいは細胞から抽出するに 際しては、培養後、公知の方法で菌体あるいは細胞を集め、これを適当な緩衝液に 懸濁し、超音波、リゾチーム及び Z又は凍結融解などによって菌体あるいは細胞を 破壊したのち、遠心分離や濾過により cgrl lポリペプチド又はその部分ペプチドの粗 抽出液を得る方法などが適宜用いられる。緩衝液の中に尿素や塩酸グァニジン等の タンパク質変性剤や、トリトン X— 100などの界面活性剤が含まれていてもよい。培養 液中に cgrl lポリペプチド又はその部分ペプチドが分泌される場合には、培養終了 後、それ自体公知の方法で菌体あるいは細胞と上清とを分離し、上清を集める。この ようにして得られた培養上清又は抽出液中に含まれる本発明のポリペプチドの精製 は、公知の分離 ·精製法を適切に組み合わせて行うことができる。これらの公知の分 離、精製法としては、塩析ゃ溶媒沈澱法などの溶解度を利用する方法、透析法、限 外ろ過法、ゲルろ過法、及び SDS— PAGE等の主として分子量の差を利用する方 法、イオン交換クロマトグラフィーなどの電荷の差を利用する方法、ァフィ二ティーク口 マトグラフィーなどの特異的親和性を利用する方法、逆相高速液体クロマトグラフィー などの疎水性の差を利用する方法、等電点電気泳動法などの等電点の差を利用す る方法などが用いられる。  [0043] When the cgrl polypeptide or its partial peptide is extracted from cultured cells or cells, the cells or cells are collected by a known method after culturing and suspended in an appropriate buffer solution. For example, a method of obtaining a crude extract of cgrl polypeptide or a partial peptide thereof by centrifugation or filtration after disrupting cells or cells by ultrasonic waves, lysozyme, Z, or freeze-thawing is appropriately used. The buffer may contain a protein denaturant such as urea or guanidine hydrochloride, or a surfactant such as Triton X-100. When the cglrl polypeptide or a partial peptide thereof is secreted into the culture solution, after completion of the culture, the cells or cells and the supernatant are separated by a method known per se, and the supernatant is collected. Purification of the polypeptide of the present invention contained in the thus obtained culture supernatant or extract can be performed by appropriately combining known separation and purification methods. As these known separation and purification methods, methods using solubility such as salting-out solvent precipitation method, dialysis method, ultrafiltration method, gel filtration method, and mainly using differences in molecular weight such as SDS-PAGE. , Methods using charge differences such as ion exchange chromatography, methods using specific affinity such as affinity mouthmatography, and differences in hydrophobicity such as reversed-phase high-performance liquid chromatography For example, a method using a difference in isoelectric point such as a method or an isoelectric focusing method may be used.
[0044] 6. cgrl lポリヌクレオチド(遺伝子を含む)の機能を阻害する核酸  [0044] 6. Nucleic acids that inhibit the function of cgrl l polynucleotides (including genes)
cgrl l遺伝子の機能を阻害する核酸は、 cgrl l遺伝子の機能を喪失させることによ り、 cgrl l遺伝子が発現し、機能している細胞の増殖又は生存維持を阻害することが 可能となる。その結果、例えば、 cgrl lの無秩序な過剰発現などが原因で癌化、腫 瘍化した細胞、又は癌化、腫瘍化する可能性のある細胞の増殖を抑えることが可能 となる。遺伝子の機能を阻害する核酸としては、例えば、アンチセンス RNA又は DN Aなどの一本鎖核酸およびその誘導体、該遺伝子領域の一部と相補的配列を有す る短レ、二本鎖 RNAなどを挙げることができる。 Nucleic acids that inhibit the function of the cgrl l gene may inhibit the growth or survival of cells that function as the cgrl l gene is expressed by losing the function of the cgrl l gene. It becomes possible. As a result, for example, it is possible to suppress the growth of cells that have become cancerous or tumorous due to, for example, disordered overexpression of cglrl, or cells that may become cancerous or tumorous. Examples of nucleic acids that inhibit gene functions include single-stranded nucleic acids such as antisense RNA or DNA and derivatives thereof, short-length or double-stranded RNA having a sequence complementary to a part of the gene region, etc. Can be mentioned.
アンチセンスは、 RNA又は DNAまたはそれらの誘導体であってよぐ cgrl l遺伝 子の発現に関する有効な阻害因子として作用する。アンチセンス RNAは、例えば、 インビボにおいて mRNAとハイブリダィズし、 mRNAから cgrl lポリペプチドへの翻 訳を阻害するようにデザインされる(Okano等, 1991)。また、 DNAオリゴヌクレオチド は、例えば、 cgrl l遺伝子の転写開始領域に対して相補的となるようにデザインされ 、その結果、該遺伝子の発現を阻害する(Cohen, 1989)。  Antisense acts as an effective inhibitor for the expression of the cgrl gene, which can be RNA or DNA or derivatives thereof. Antisense RNAs are designed, for example, to hybridize with mRNA in vivo and inhibit translation from mRNA to cgrl polypeptide (Okano et al., 1991). In addition, the DNA oligonucleotide is designed to be complementary to the transcription initiation region of the cglrl gene, for example, and as a result, inhibits the expression of the gene (Cohen, 1989).
これらのアンチセンス RNA又は DNAが cgrl l遺伝子又はポリペプチドの発現を阻 害するようにインビボにおレ、て機能し得るように細胞へ導入することができる。アンチ センス DNAが用いられる場合には、例えば、標的遺伝子配列の約 10と + 10の間 の位置に結合するオリゴヌクレオチドであることが望ましい。  These antisense RNAs or DNAs can be introduced into cells so that they can function in vivo to inhibit the expression of cglrl genes or polypeptides. When antisense DNA is used, for example, an oligonucleotide that binds to a position between about 10 and +10 of the target gene sequence is desirable.
また、 cgrl l遺伝子と相補的な配列を有する二本鎖 RNAは RNAi (RNA干渉)に 用レヽることもできる。 RNAiは、 目的の mRNA (例えば、 cgrl l遺伝子の mRNA)が、 目的の mRNAと相補的な配列を持つ二本鎖 RNAにより分解される現象のことである 。この現象を利用して人工的に二本鎖 RNAを導入することにより、 目的の遺伝子の 発現を抑制することができる。従って、上述のアンチセンス法とは異なる原理に基づく と考えられる。  In addition, double-stranded RNA having a sequence complementary to the cglrl gene can be used for RNAi (RNA interference). RNAi is a phenomenon in which the target mRNA (for example, mRNA of the cglrl gene) is degraded by double-stranded RNA having a sequence complementary to the target mRNA. By artificially introducing double-stranded RNA using this phenomenon, the expression of the target gene can be suppressed. Therefore, it is considered to be based on a principle different from the above-described antisense method.
7. cgrl lポリペプチドに対する抗体 7. Antibodies against cgrl l polypeptide
cgrl 1ポリペプチドに対する抗体は、 cgrl 1ポリヌクレオチドの活性を喪失させるこ とにより、 cgrl lポリペプチドが発現し、機能している細胞の増殖又は生存維持を阻 害すること力 S可能となる。その結果、例えば、 cgrl lの無秩序な過剰発現などが原因 で癌化、腫瘍化した細胞、又は癌化、腫瘍化する可能性のある細胞の増殖を抑える ことが可能となる。  An antibody against the cgrl 1 polypeptide is capable of inhibiting the growth or survival of cells in which the cgrl 1 polypeptide is expressed and functioning by losing the activity of the cgrl 1 polynucleotide. As a result, it is possible to suppress the proliferation of cells that have become cancerous or tumorous due to, for example, disordered overexpression of cglrl, or cells that may become cancerous or tumorous.
さらに、例えば、 cgrl lポリペプチドの過剰発現による癌の発症の有無、又は予後 の判断を cgrl lポリペプチドに対する抗体を用いて行うことができる。例えば、検体か ら取得した血中に存在する cgrl lポリペプチドの濃度を、該抗体を用いて測定するこ とにより、癌化の有無又は癌の予後の判断を行うことができる。 Further, for example, the presence or absence of cancer due to overexpression of cgrl l polypeptide, or prognosis This determination can be made using an antibody against the cgrl polypeptide. For example, the presence or absence of canceration or the prognosis of cancer can be determined by measuring the concentration of cgrl polypeptide present in blood obtained from a specimen using the antibody.
[0046] cgrl lに対する抗体には、 cgrl lポリペプチドと特異的に結合する抗体、及びその Fab又は F(ab ' )などの抗体断片が含まれる。 [0046] Antibodies against cgrl l include antibodies that specifically bind to cgrl l polypeptide, and antibody fragments such as Fab or F (ab ') thereof.
2  2
本発明における「抗体」(本発明のポリペプチドの活性を促進する抗体、本発明の ポリペプチドの活性を阻害する抗体の両方を含む)には、 cgrl lポリペプチドに対す るモノエピトープ特異抗体、ポリェピトープ特異抗体、単一鎖抗体、及びこれらの断 片が含まれる。これらの抗体には、例えば、モノクローナル抗体、ポリクローナル抗体 、ヒト化抗体などが含まれる。  The “antibody” in the present invention (including both an antibody that promotes the activity of the polypeptide of the present invention and an antibody that inhibits the activity of the polypeptide of the present invention) includes a monoepitope specific antibody against the cgrl polypeptide, Polyepotope specific antibodies, single chain antibodies, and fragments thereof are included. These antibodies include, for example, monoclonal antibodies, polyclonal antibodies, humanized antibodies and the like.
7- 1.ポリクローナル抗体  7- 1. Polyclonal antibodies
ポリクローナル抗体は、例えば、哺乳類宿主動物に対して、免疫原及びアジュバン トの混合物をインジヱタトすることにより調製することができる。通常は、免疫原及び/ 又はアジュバントを宿主動物の皮下又は腹腔内へ複数回インジェクトする。免疫原に は本発明のポリペプチド及びその異種ポリペプチドとの融合体又はこれらの断片が 含まれる。例えば、配列番号 13又は 14で表されるアミノ酸配列からなるペプチドは、 非常に優れた免疫原となる。  Polyclonal antibodies can be prepared, for example, by incubating a mixture of immunogen and adjuvant against a mammalian host animal. Usually, the immunogen and / or adjuvant is injected multiple times subcutaneously or intraperitoneally into the host animal. Immunogens include polypeptides of the invention and fusions with heterologous polypeptides or fragments thereof. For example, a peptide consisting of the amino acid sequence represented by SEQ ID NO: 13 or 14 is a very good immunogen.
アジュバントの例には、完全フロイト及びモノホスホリル脂質 A合成 トレハロースジ コリノミコレート(MPL—TDM)が含まれる。特に、本発明のポリペプチドの部分ぺプ チドを免疫原とする場合は、免疫応答を増強するために、キーホールリンペットへモ シァニン(KLH)、血清アルブミン、ゥシサイログロブリン及び大豆トリプシンインヒビタ 一などの免疫原性を有するタンパク質と該免疫原とを結合させたのち、インジヱタトし てもよい。  Examples of adjuvants include complete Freud and monophosphoryl lipid A synthesis trehalose dicorynomycolate (MPL-TDM). In particular, when the partial peptide of the polypeptide of the present invention is used as an immunogen, keyhole limpet hemocyanin (KLH), serum albumin, thythyroglobulin, and soybean trypsin inhibitor are used to enhance the immune response. After binding the immunogen such as a protein having immunogenicity, it may be digitized.
あるいは、 IgY分子を産生するニヮトリを用いて調製してもよレ、(Schade等, 1996)。 抗体産生方法の詳細については、例えば、 Ausubel等, 1987又は Harlow及び Lane, 1988を参照されたい。  Alternatively, it can be prepared using a chicken that produces IgY molecules (Schade et al., 1996). See, for example, Ausubel et al., 1987 or Harlow and Lane, 1988 for details on antibody production methods.
[0047] 7 - 2.モノクローナル抗体 [0047] 7-2. Monoclonal antibodies
モノクローナル抗体は、ハイプリドーマ法を用いて調製することができる(Milstein及 び Cuello, 1983)。 Monoclonal antibodies can be prepared using the Hypridoma method (Milstein et al. Cuello, 1983).
この方法には以下に示す 4つの工程が含まれる:(i)宿主動物または、宿主動物由 来のリンパ球を免疫する、 (ii)モノクローナル抗体分泌性 (又は潜在的に分泌性)のリ ンパ球を回収する、(iii)リンパ球を不死化細胞に融合させる、(iv)所望のモノクロ一 ナル抗体を分泌する細胞を選択する。  This method includes the following four steps: (i) immunizing the host animal or lymphocytes derived from the host animal, (ii) a monoclonal antibody secreting (or potentially secreting) linker. Collect spheres, (iii) fuse lymphocytes to immortalized cells, (iv) select cells that secrete the desired monoclonal antibody.
マウス、ラット、モルモット、ハムスター、又は他の適当な宿主動物力 免疫動物とし て選択され免疫原がインジ工クトされる。或いは、免疫動物から取得したリンパ球をィ ンビトロで免疫化してもよレ、。ヒト細胞が望ましい場合には、末梢血リンパ球(PBLs) が一般に使用される。し力 ながら、他の哺乳類由来の脾臓細胞又はリンパ球がより 一般的で好ましい。免疫原には、 cgrl lポリペプチド及びその異種ポリペプチドとの 融合体又はこれらの断片も含まれる。免疫原としては、例えば、配列番号 13又は 14 で表されるアミノ酸配列からなるペプチドは、非常に優れた免疫原となる。  Mouse, rat, guinea pig, hamster, or other suitable host animal immunity is selected as an immunized animal and the immunogen is injected. Alternatively, lymphocytes obtained from immunized animals can be immunized in vitro. If human cells are desired, peripheral blood lymphocytes (PBLs) are commonly used. However, spleen cells or lymphocytes from other mammals are more common and preferred. Immunogens also include cgrl polypeptide and fusions with heterologous polypeptides or fragments thereof. As an immunogen, for example, a peptide consisting of the amino acid sequence represented by SEQ ID NO: 13 or 14 is a very excellent immunogen.
[0048] 免疫後、宿主動物から得られたリンパ球はハイプリドーマ細胞を樹立するために、 ポリエチレングリコールなどの融合剤を用いて不死化細胞株と融合される(Goding, 1[0048] After immunization, lymphocytes obtained from the host animal are fused with an immortalized cell line using a fusion agent such as polyethylene glycol to establish hyperpridoma cells (Goding, 1
996)。融合細胞としては、トランスフォーメーションによって不死化されたげつ歯類、ゥ シ、又はヒトのミエローマ細胞が使用されるカ ラットもしくはマウスのミエローマ細胞 株が使用される。細胞融合を行った後、融合しなかったリンパ球及び不死化細胞株 の成長又は生存を阻害する一又は複数の基質を含む適切な培地中で細胞を生育さ せる。通常の技術では、酵素のヒポ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(HGPRT又は HPRT)を欠く親細胞を使用する。この場合、ヒポキサンチン、ァ ミノプテリン及びチミジンが HGPRT欠損細胞の成長を阻害し、ハイプリドーマの成長 を許容する培地 (HAT培地)に添加される。 996). As the fusion cell, a cartilage or mouse myeloma cell line in which rodents, rabbits, or human myeloma cells immortalized by transformation are used is used. After cell fusion, the cells are grown in a suitable medium containing one or more substrates that inhibit the growth or survival of unfused lymphocytes and immortalized cell lines. Conventional techniques use parental cells that lack the enzyme hypoxanthine guanine phosphoribosyltransferase (HGPRT or HPRT). In this case, hypoxanthine, aminopterin and thymidine are added to a medium (HAT medium) that inhibits the growth of HGPRT-deficient cells and allows the growth of hypridoma.
[0049] モノクローナル抗体の調製にあたり、好ましい不死化細胞株はマウスミエローマ株 で、アメリカンタイプカルチャーコレクション(Manassas, VA)より入手可能である。ヒトミ エローマ及びマウス一ヒトヘテロミエローマ細胞株による、ヒトモノクローナル抗体産生 に関しては、 Kozbor等, 1984 ; Schook, 1987を参照のこと。 [0049] For the preparation of monoclonal antibodies, a preferred immortal cell line is the mouse myeloma line, which is available from the American Type Culture Collection (Manassas, VA). See Kozbor et al., 1984; Schook, 1987 for the production of human monoclonal antibodies by human myeloma and mouse human heteromyeloma cell lines.
ハイプリドーマ細胞は細胞外に抗体を分泌するため、 目的のモノクローナル抗体の 産生の有無を培養液を用いて確認することができる。産生されたモノクローナル抗体 の結合特異性は、ラジオィムノアッセィ (RIA)又は酵素結合免疫吸着検定法 (ELIS A)などの免疫沈降又はインビトロでの結合アツセィにより評価することができる(Harlo w及び Lane, 1988; Harlow及び Lane, 1999)。 Since hyperpridoma cells secrete antibodies extracellularly, the presence or absence of production of the desired monoclonal antibody can be confirmed using a culture solution. Produced monoclonal antibody The binding specificity of can be assessed by immunoprecipitation such as radioimmunoassay (RIA) or enzyme-linked immunosorbent assay (ELIS A) or in vitro binding assays (Harlow and Lane, 1988; Harlow And Lane, 1999).
モノクローナル抗体分泌性ハイプリドーマ細胞は、限界希釈法及びサブカルチャー により単一クローンとして単離することができる(Goding, 1996)。適切な培地にはダル べッコ改変イーグル培地、 RPMI— 1640、場合によっては、タンパク質を含まない培 地若しくは無血清培地などが含まれる。また、ハイプリドーマ細胞は、適切な宿主動 物の腹水中で増殖させてもよい。  Monoclonal antibody-secreting hyperpridoma cells can be isolated as single clones by limiting dilution and subculture (Goding, 1996). Suitable media include Dulbecco's Modified Eagle Medium, RPMI-1640, and in some cases, protein-free or serum-free medium. Hypridoma cells may also be grown in the ascites of suitable host animals.
モノクローナル抗体は、培地又は腹水力、らプロテイン Aセファロース、ハイド口キシァ パタイトクロマトグラフィー、ゲル電気泳動、透析、硫安沈殿又はァフィ二ティークロマ トグラフィー(Harlow及び Lane, 1988 ; Harlow及び Lane, 1999)などの当業者にとって 周知の方法によって単離、精製される。  Monoclonal antibodies can be obtained from medium or ascites, et al. Protein A sepharose, hydoxyxpatite chromatography, gel electrophoresis, dialysis, ammonium sulfate precipitation or affinity chromatography (Harlow and Lane, 1988; Harlow and Lane, 1999). It is isolated and purified by methods well known to those skilled in the art.
また、モノクローナル抗体は遺伝子組換え技術によっても作製することができる(米 国特許第 4166452号, 1979)。 目的の抗体を分泌するハイプリドーマ細胞株から目的 のモノクローナル抗体ポリペプチドをコードする遺伝子を同定するのに、例えば、マウ スの重鎖及び軽鎖抗体遺伝子と特異的に結合するオリゴヌクレオチドプローブを用 いてもよい。その結果、抗体重鎖及び軽鎖遺伝子が取得された場合は、その遺伝子 の配歹 1Jを決定することにより目的の抗体遺伝子を同定することができる。単離された DNA断片は、モノクローナル抗体を発現させるために、適当な発現ベクターに抗体 遺伝子を導入し、該ベクターを他の免疫グロブリンタンパク質を生産しなレ、simian COS— 7細胞、チャイニーズハムスター卵巣(CHO)細胞、又はミエローマ細胞など のホスト細胞中へトランスフヱタトする。単離された DNA断片は、例えば、ヒト重鎖及 び軽鎖定常ドメインに対するコード化配列を相同なマウス配列と置換することにより( 米国特許第 4816567号, 1989 ; Morrison等, 1987)、又は非免疫グロブリンポリべプチ ドをコードする配列の全て又は一部と免疫グロブリンコード化配列を融合することによ り、修飾することができる。そのような非免疫グロブリンポリペプチドは、キメラ二価抗 体を調製するために、抗体の定常ドメインと置換することが可能であり、又は一抗原 結合部位の定常ドメインと置換することができる。 [0051] 7—3.ヒト化及びヒト抗体 Monoclonal antibodies can also be produced by gene recombination techniques (US Pat. No. 4166452, 1979). To identify the gene encoding the desired monoclonal antibody polypeptide from the hyperidoma cell line secreting the antibody of interest, for example, using oligonucleotide probes that specifically bind to the mouse heavy and light chain antibody genes. May be. As a result, when antibody heavy chain and light chain genes are obtained, the target antibody gene can be identified by determining the distribution 1J of the genes. In order to express monoclonal antibodies, the isolated DNA fragment is transferred to an appropriate expression vector and the antibody gene is not produced by any other immunoglobulin protein, simian COS-7 cells, Chinese hamster ovary Transfect into (CHO) cells or host cells such as myeloma cells. Isolated DNA fragments can be obtained, for example, by replacing coding sequences for human heavy and light chain constant domains with homologous mouse sequences (US Pat. No. 4,816,567; 1989; Morrison et al., 1987) or non- Modifications can be made by fusing the immunoglobulin coding sequence with all or part of the sequence encoding the immunoglobulin polypeptide. Such non-immunoglobulin polypeptides can be substituted for the constant domain of an antibody or can be substituted for the constant domain of a single antigen binding site to prepare a chimeric bivalent antibody. [0051] 7-3. Humanized and human antibodies
cgrl lポリペプチド抗体には、ヒトイ匕又はヒト抗体が含まれる。非ヒト抗体のヒト化型 は、非ヒト免疫グロブリン由来の最小配列を含むキメラ免疫グロブリン、免疫グロブリン 鎖又はその断片(Fv, Fab, Fab ' , F (ab ' ) 又は他の抗体の抗原結合領域など)で  cgrl polypeptide antibodies include human rabbits or human antibodies. Humanized forms of non-human antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (Fv, Fab, Fab ′, F (ab ′)) or other antibody antigen-binding regions that contain minimal sequences derived from non-human immunoglobulin. Etc
2  2
ある。  is there.
一般に、ヒトイ匕抗体は非ヒト由来の免疫グロブリンから導入された一又は複数のアミ ノ酸残基を持つ。これらの非ヒトアミノ酸残基は、多くの場合、可変ドメインから選ばれ る。ヒト化抗体は、例えばマウスの CDRs又は CDR (相補性決定領域)配列と対応す るヒト抗体配列とを置換することにより作製することができる(Jones等, 1986 ; Riechman n等, 1988 ;Verhoeyen等, 1988)。つまり、ヒト化抗体とは、ヒト由来の特定の CDR中の ある残基と、該残基に相当するマウス、ラット又はゥサギなどの非ヒト種の CDR中の残 基と置換されているヒト抗体のことである。また、非ヒト由来の残基によって、ヒト免疫グ ロブリンの Fvフレームワーク残基が置換される場合もある(Jones等, 1986 ; Presta, 19 92 ; Riechmann等, 1988)。  In general, human rabbit antibodies have one or more amino acid residues introduced from non-human immunoglobulin. These non-human amino acid residues are often selected from variable domains. Humanized antibodies can be produced, for example, by replacing mouse CDRs or CDR (complementarity determining region) sequences with corresponding human antibody sequences (Jones et al., 1986; Riechmann et al., 1988; Verhoeyen et al. , 1988). In other words, a humanized antibody is a human antibody that is substituted with a residue in a specific human-derived CDR and a residue in a CDR of a non-human species such as mouse, rat, or rabbit corresponding to the residue. That is. In addition, nonhuman-derived residues may replace Fv framework residues of human immunoglobulin (Jones et al., 1986; Presta, 1992; Riechmann et al., 1988).
[0052] 8. cgrl lポリペプチドに対するァゴニストおよびアンタゴニスト [0052] 8. Agents and antagonists to cgrl l polypeptide
本発明における「ァゴ二スト」とは、 cgrl lポリペプチド、そのレセプター、又は相互 作用因子 (結合パートナー)に特異的に結合して、本発明のポリペプチドの生物学的 活性を促進するものを意味し、上述の抗体の一部も「ァゴ二スト」に含まれる。抗体以 外のァゴストとしては、限定はしないが、ポリペプチドまたはその断片、核酸、その他 低分子化合物などを挙げることができる。また、本発明における「アンタゴニスト」とは 、 cgrl lポリペプチドまたはそのレセプター、相互作用因子(結合パートナー)に特異 的に結合して、本発明のポリペプチドの生物学的活性を抑制するものを意味し、上 述の抗体の一部も「アンタゴニスト」に含まれる。  The “agonist” in the present invention is a substance that specifically binds to cgrl polypeptide, its receptor, or interacting factor (binding partner) to promote the biological activity of the polypeptide of the present invention. And a part of the above-mentioned antibody is also included in the “agonist”. Examples of non-antibody antibodies include, but are not limited to, polypeptides or fragments thereof, nucleic acids, and other low molecular weight compounds. The “antagonist” in the present invention means a substance that specifically binds to cglrl polypeptide, its receptor, or an interaction factor (binding partner) and suppresses the biological activity of the polypeptide of the present invention. However, some of the above-mentioned antibodies are also included in “antagonists”.
[0053] 9. cgrl lポリペプチドに対するァプタマ一  [0053] 9. Aptamer for cgrl l polypeptide
アブタマ一は標的タンパク質と特異的に結合する能力を持った核酸分子又はぺプ チドで、標的タンパク質の活性を阻害することができる。また、活性阻害ができないも のでも、標的タンパク質と結合することができるため、該タンパク質の有無を調べるた めに使用することができる。 従って、 cgrl lポリペプチドに対するァプタマ一は、 cgrl lポリペプチドの活性を喪 失させることにより、 cgrl lポリペプチドが発現し、機能している細胞の増殖又は生存 維持を阻害することが可能となる。あるいは、 cgrl lポリペプチドの過剰発現による疾 患、例えば、癌の発症の有無、又は予後の判断を cgrl lポリペプチドに対するァプタ マーを用いて行うことができる。例えば、検体から取得した血中に存在する cgrl lポリ ペプチドの濃度を、該ァプタマ一を用いて測定することにより、例えば、癌化の有無 又は癌の予後の判断を行うことができる。 Abutama is a nucleic acid molecule or peptide capable of specifically binding to a target protein and can inhibit the activity of the target protein. In addition, even if the activity cannot be inhibited, it can be used to examine the presence or absence of the protein because it can bind to the target protein. Therefore, aptamers to cgrl l polypeptide can inhibit the growth or survival of functioning cells by expressing cgrl l polypeptide by losing the activity of cgrl l polypeptide. . Alternatively, a disease caused by overexpression of cgrl l polypeptide, for example, the presence or absence of onset of cancer, or prognosis can be determined using an aptamer to cgrl l polypeptide. For example, the presence or absence of canceration or the prognosis of cancer can be determined by measuring the concentration of cgrl polypeptide present in blood obtained from a specimen using the aptamer.
[0054] 「ァプタマ一」が核酸の場合、 RNAであっても DNAであってもよぐこれらの RNA または DNAが cgrl lポリペプチドに特異的に結合するものである限り特に限定はし ないが、好ましくは DNAである。また、「ァプタマ一」には、リン酸骨格において修飾 を加え、生体内における安定性を増大させたものも含み、特に限定はしないが、ホス ホロチォエート結合、ホスホロジチォエート結合、ホスホラミドチォエート結合、ホスホ ラミデート結合、ホスホルジアミデート結合、メチルホスホネート結合を含むリン酸骨格 が好ましい。 [0054] When the "aptamer" is a nucleic acid, it is not particularly limited as long as the RNA or DNA, whether it is RNA or DNA, specifically binds to the cglrl polypeptide. Preferably, it is DNA. “Aptamers” also include those that have been modified in the phosphate skeleton to increase in vivo stability, and are not particularly limited, but include phosphorothioate bonds, phosphorodithioate bonds, and phosphoramidothioates. A phosphate skeleton containing a bond, a phosphoramidate bond, a phosphordiamidate bond, or a methylphosphonate bond is preferred.
[0055] 10. cgrl lポリペプチドと相互作用する因子の決定方法  [0055] 10. Method for Determining Factors That Interact with cgrl l Polypeptide
cgrl lポリペプチドと相互作用する因子は、 cgrl lポリペプチドによる細胞の増殖 促進又は生存維持活性に対し促進又は抑制効果をもたらすことが期待される。従つ て、該因子を本発明の組成物にさらに含有せしめることで、 目的に応じた効果を実現 すること力 S可能となる。ここで「相互作用する因子」には、タンパク質、ペプチド、小分 子化合物など、当業者の想定し得るものが含まれる。例えば、タンパク質としては、 eg rl lポリペプチドに対するレセプターが含まれる。 cgrl lポリペプチドと相互作用する 因子を決定する場合、限定はしないが、 cgrl lポリペプチドもしくはその一部と候補 因子 (例えば、レセプターなど)又は候補因子をその表面に提示する細胞(例えば、 候補レセプタータンパク質をその表面に発現させた細胞など)とを接触させた場合に おける、 cgrl lポリペプチドもしくはその一部と候補因子との結合量、あるいは、細胞 などを使用した場合には、該細胞内における生物学的な変化を検出することにより相 互作用の有無を決定することができる。  The factor that interacts with the cgrl l polypeptide is expected to have a promoting or suppressing effect on the cell growth promotion or survival maintenance activity of the cgrl l polypeptide. Therefore, by further adding the factor to the composition of the present invention, it is possible to achieve the effect according to the purpose. Here, “interacting factors” include those that can be assumed by those skilled in the art, such as proteins, peptides, and small compounds. For example, proteins include receptors for eg rl l polypeptides. When determining a factor that interacts with a cgrl l polypeptide, the cgrl l polypeptide or a part thereof and a candidate factor (eg, a receptor) or a cell that presents a candidate factor on its surface (eg, a candidate) A cell that expresses a receptor protein on its surface, etc.), the binding amount of cgrl l polypeptide or a part thereof with a candidate factor, or the cell, etc. The presence or absence of interaction can be determined by detecting biological changes in the body.
具体的には、 cgrl lポリペプチドもしくはその一部、又は候補因子を標識 (例えば、 蛍光標識、放射標識)し、両者を接触させた後、標識から得られるシグナル (例えば、 蛍光、放射活性など)の強度を指標にし、定法及び技術常識に基づいて、両者の結 合量を検出し、その結果、両者が強く結合しているとの評価が可能である場合には、 候補因子を相互作用因子であると決定することができる。あるいは、レセプターを決 定する場合には、候補レセプターを発現させている細胞と、 cgrl lポリペプチドもしく はその一部とを接触させ、レセプターを介した細胞特有の細胞内シグナル (例えば、 細胞内 Ca2+遊離、細胞内 cAMP生成、細胞膜電位変動、細胞内タンパク質のリン 酸化など)における変動が検出されたときは、候補レセプターが実際にレセプターと して機能しているとの予測をすることが可能となる。 Specifically, the cgrl l polypeptide or a part thereof, or a candidate factor is labeled (for example, Fluorescent labeling, radiolabeling), contacting the two, and using the intensity of the signal obtained from the label (for example, fluorescence, radioactivity, etc.) as an index, detect the amount of binding of both based on conventional methods and common technical knowledge As a result, if it is possible to evaluate that both are strongly bound, the candidate factor can be determined to be an interacting factor. Alternatively, when determining a receptor, a cell expressing a candidate receptor is contacted with a cgrl polypeptide or a part thereof, and a cell-specific intracellular signal via the receptor (for example, cell When changes in intracellular Ca 2+ release, intracellular cAMP production, cell membrane potential fluctuation, intracellular protein phosphorylation, etc.) are detected, it is predicted that the candidate receptor actually functions as a receptor It becomes possible.
[0056] 11.組成物 [0056] 11. Composition
cgrl lポリヌクレオチド、ポリペプチド、該ポリペプチドに対するァゴニストなどを含ん でなる組成物は、細胞の増殖促進若しくは生存維持を目的として使用することができ 、また、該ポリヌクレオチドの活性阻害核酸又は該ポリペプチドに対する抗体、アンタ ゴニストなどを含んでなる組成物は、細胞の異常増殖 (腫瘍化など)を抑制することを 目的として使用することができる。本組成物は、培養細胞などへの使用のためにイン ビトロにおいても使用することができる。本組成物には、 cgrl lポリヌクレオチドなどの 有効成分の他に、細胞の等張性を保っための物質、あるいは該有効成分の細胞内 への移行を促進するための物質、ノくッファーなどの補助物質が含まれる。これらの補 助物質は当業者が利用可能なものであれば全て利用することができる。  A composition comprising a cgrl polynucleotide, a polypeptide, an agonist for the polypeptide, etc. can be used for the purpose of promoting cell growth or maintaining the survival of the polynucleotide. A composition comprising an antibody against a peptide, an antagonist, etc. can be used for the purpose of suppressing abnormal cell growth (eg, tumorigenesis). The composition can also be used in vitro for use in cultured cells and the like. In addition to active ingredients such as cgrl polynucleotides, the present composition includes substances for maintaining isotonicity of cells, substances for promoting the transfer of the active ingredients into cells, and cuffers, etc. Auxiliary substances are included. Any of these auxiliary substances can be used if they are available to those skilled in the art.
[0057] 12.医薬組成物 [0057] 12. Pharmaceutical composition
cgrl lポリヌクレオチド、ポリペプチド、該ポリペプチドに対するァゴニストなどを含ん でなる組成物は、細胞の増殖促進若しくは生存維持を目的として使用することができ 、また、該ポリヌクレオチドの活性阻害核酸又は該ポリペプチドに対する抗体、アンタ ゴニストなどを含んでなる組成物は、細胞の異常増殖 (腫瘍化など)を抑制することを 目的として使用することができることから医薬組成物としても利用可能である。該医薬 組成物は、例えば、血管内皮細胞を標的として使用すれば、血流障害関連疾患の 治療にその効果が期待でき、また、下垂体を形成する細胞等を標的として使用すれ ば、発育障害関連疾患の治療にその効果が期待でき、一方、腫瘍又は癌化した異 常増殖を示す細胞を標的として使用すれば、癌の治療又は予防などにおいて効果 が期待できる。 A composition comprising a cgrl polynucleotide, a polypeptide, an agonist for the polypeptide, etc. can be used for the purpose of promoting cell growth or maintaining the survival of the polynucleotide. A composition comprising an antibody against a peptide, an antagonist, etc. can be used as a pharmaceutical composition because it can be used for the purpose of suppressing abnormal cell growth (eg, tumorigenesis). The pharmaceutical composition can be expected to be effective in treating a blood flow disorder-related disease if, for example, vascular endothelial cells are used as a target. Also, if the pituitary-forming cells are used as a target, the pharmaceutical composition is considered to be a growth disorder. It can be expected to be effective in treating related diseases, while tumors or cancerous diseases If cells that show normal growth are used as targets, an effect can be expected in the treatment or prevention of cancer.
[0058] さらに、本発明に係る cgrl lポリヌクレオチド、ポリペプチド、該ポリペプチドに対す るァゴニストなどを含んでなる組成物は、血圧を上昇させる効果を有し、また、該ポリ ヌクレオチドの活性阻害核酸又は該ポリペプチドに対する抗体、アンタゴニストなどを 含んでなる組成物は、血圧を降下させる効果を有することから、医薬組成物としても 利用可能である。該医薬組成物は、例えば、昇圧剤、血圧降下剤、低血圧症の治療 剤、高血圧症の治療剤としての効果が期待される。  [0058] Furthermore, the composition comprising the cglrl polynucleotide according to the present invention, a polypeptide, an agonist for the polypeptide, etc. has an effect of increasing blood pressure, and also inhibits the activity of the polynucleotide. A composition comprising an nucleic acid or an antibody against the polypeptide, an antagonist or the like has an effect of lowering blood pressure, and thus can be used as a pharmaceutical composition. The pharmaceutical composition is expected to be effective as, for example, an antihypertensive agent, a hypotensive agent, a hypotensive therapeutic agent, or a hypertensive therapeutic agent.
[0059] ここで、「血流障害関連疾患」とは、体の全体又は一部において血液の循環に支障 を来す結果引き起こされる疾患又は血液の循環に支障を来す原因となる疾患のこと で、例えば、動脈硬化症、炎症性脈管炎、動脈瘤、血栓静脈炎、さらに、腎臓病など も含まれ、当業者の技術常識の範囲に含まれる全ての疾患のことである。  [0059] Here, "blood flow disorder-related disease" refers to a disease caused as a result of hindering blood circulation in the whole or a part of the body or a disease causing hindrance to blood circulation. In addition, for example, arteriosclerosis, inflammatory vasculitis, aneurysm, thrombophlebitis, and kidney disease are all diseases within the scope of common technical knowledge of those skilled in the art.
また、「高血圧症」、「低血圧症」には血圧の異常によって引き起こされる関連疾患、 例えば、心臓疾患 (例えば、心筋梗塞、大動脈狭窄、心臓肥大症、肥大性心筋症及 び心不全など)も含まれる。  In addition, “hypertension” and “hypotension” include related diseases caused by abnormal blood pressure, such as heart diseases (for example, myocardial infarction, aortic stenosis, cardiac hypertrophy, hypertrophic cardiomyopathy and heart failure). included.
「癌」とは、細胞の異常な成長又は増殖と伴う疾患のことで、例えば、腺癌、リンパ腫、 芽細胞腫、黒色腫、肉腫、及び白血病など、当業者の技術常識の範囲に含まれる全 ての疾患のことである。  “Cancer” refers to a disease associated with abnormal growth or proliferation of cells, and is within the scope of common technical knowledge of those skilled in the art, such as adenocarcinoma, lymphoma, blastoma, melanoma, sarcoma, and leukemia. It refers to all diseases.
「神経疾患」とは、一般的な用いられる意味で理解されるべきもので、神経細胞又は パラニューロンの変性もしくは損傷によって引き起こされる全ての疾患がふくまれ、例 えば、パーキンソン病、アルツハイマー病、ハンチントン病、筋萎縮性側索硬化症、 脊髄小脳変性症など、当業者の技術常識の範囲に含まれる全ての疾患のことである  “Neurological disease” is to be understood in a general sense and includes all diseases caused by the degeneration or damage of nerve cells or para-neurons, eg Parkinson's disease, Alzheimer's disease, Huntington All diseases within the scope of common technical knowledge of those skilled in the art, such as illness, amyotrophic lateral sclerosis, spinocerebellar degeneration
「発育障害」とは、主として身体的な発育に何らかの異常が認められること状態のこと で、その関連疾患としては、限定はしないが、例えば、小人症、巨人症などを挙げる こと力 Sできる。 “Developmental disorder” is a condition in which some abnormality is observed in physical growth. Related diseases include, but are not limited to, dwarfism, giantism, etc. .
[0060] 上記 cgrl lポリヌクレオチドなどは、生体に対して悪影響を及ぼさない医薬組成物 の形態で治療薬として使用することができる。通常、そのような組成物には、核酸分 子、タンパク質又は抗体などの他、薬剤的に受容可能な担体が含まれる。 [0060] The above cgrl polynucleotide can be used as a therapeutic agent in the form of a pharmaceutical composition that does not adversely affect the living body. Usually, such compositions contain a nucleic acid component. In addition to a child, protein or antibody, a pharmaceutically acceptable carrier is included.
「薬剤的に受容可能な担体」は、溶媒、分散媒、コーティング剤、抗菌及び抗真菌 剤、アイソトニックに作用して吸着を遅らせる薬剤及びその類似物を含み、薬剤的投 与に適するもののことである(Gennaro, 2000)。該担体及び該担体を希釈するために 好ましいものの例には、限定はしないが、水、生理食塩水、フィンガー溶液、デキスト ロース溶液、及びヒト血清アルブミンなどが含まれる。また、リボソーム及び不揮発性 油などの非水溶性媒体も用いられる。さらに、上記 cgrl lポリヌクレオチドなどの活性 を保護又は促進するような特定の化合物が、該組成物中に包含されてレ、てもよレ、。  “Pharmaceutically acceptable carrier” includes solvents, dispersion media, coatings, antibacterial and antifungal agents, agents that act isotonically to delay adsorption and the like and are suitable for pharmaceutical administration. Yes (Gennaro, 2000). Examples of the carrier and preferred for diluting the carrier include, but are not limited to, water, saline, finger solution, dextrose solution, human serum albumin, and the like. Non-water soluble media such as ribosomes and non-volatile oils are also used. Furthermore, certain compounds that protect or promote activity such as the above cgrl polynucleotides may be included in the composition.
[0061] 本発明に係る医薬組成物は、静脈内、皮内、皮下、経口(例えば、吸入なども含む )、経皮及び経粘膜への投与を含み、治療上適切な投与経路に適合するように製剤 化される。非経口、皮内、又は皮下への適用に使用される溶液又は懸濁液には、限 定はしないが、注射用の水などの滅菌的希釈液、生理食塩水溶液、不揮発性油、ポ リエチレングリコール、グリセリン、プロピレングリコール、又は他の合成溶媒、ベンジ ノレアルコール又は他のメチルパラベンなどの保存剤、ァスコルビン酸又は亜硫酸水 素ナトリウムなどの抗酸化剤、塩ィ匕ベンザルコニゥム、塩酸プロ力インなどの無痛化 剤、エチレンジアミンテトラ酢酸 (EDTA)などのキレート剤、酢酸塩、クェン酸塩、又 はリン酸塩などの緩衝剤、塩化ナトリウム又はデキストロースなど浸透圧調製のため の薬剤を含んでもよい。  [0061] The pharmaceutical composition according to the present invention includes intravenous, intradermal, subcutaneous, oral (for example, including inhalation), transdermal, and transmucosal administration, and is suitable for a therapeutically appropriate route of administration. It is formulated as follows. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application include, but are not limited to, sterile diluents such as water for injection, saline solution, non-volatile oil, polyethylene. Preservatives such as glycol, glycerin, propylene glycol, or other synthetic solvents, benzenolic alcohol or other methylparabens, antioxidants such as ascorbic acid or sodium hydrogen sulfite, painless such as salt benzalkonium, prohydroin hydrochloride It may contain a chemical, a chelating agent such as ethylenediaminetetraacetic acid (EDTA), a buffer such as acetate, citrate, or phosphate, and an agent for osmotic pressure adjustment such as sodium chloride or dextrose.
pHは塩酸又は水酸化ナトリウムなどの酸又は塩基で調製することができる。非経口 的標品はアンプル、ガラスもしくはプラスチック製の使い捨てシリンジ又は複数回投 与用バイアル中に収納される。  The pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. Parenteral preparations are contained in ampoules, disposable glass or plastic syringes or multiple dose vials.
[0062] 12- 1.注射可能な製剤  [0062] 12- 1. Injectable preparation
注射に適する医薬組成物には、滅菌された注射可能な溶液又は分散媒を、使用 時に調製するための滅菌水溶液 (水溶性の)又は分散媒及び滅菌されたパウダーが 含まれる。静脈内の投与に関し、適切な担体には生理食塩水、静菌水、 CREMOP HOR ELTM (BASF, Parsippany, N.J.)、又はリン酸緩衝化生理食塩水(PBS)が 含まれる。注射剤として使用する場合、組成物は滅菌的でなくてはならず、また、シリ ンジを用いて投与されるために十分な流動性を保持していなくてはならなレ、。該組成 物は、調剤及び保存の間、化学変化及び腐食等に対して安定でなくてはならず、細 菌及び真菌などの微生物由来のコンタミネーシヨンを防止する必要がある。担体は、 例えば、水、エタノール、ポリオール(グリセロール、プロピレングリコール、及び液体 ポリエチレングリコールなど)、及び適切な混合物を含む溶媒又は分散媒培地を使用 すること力 Sできる。例えば、レクチンなどのコーティング剤を用レ、、分散媒においては 必要とされる粒子サイズを維持し、界面活性剤を用いることにより適度な流動性が維 持される。種々の抗菌剤及び抗真菌剤、例えば、パラベン、クロロブタノール、フヱノ ール、ァスコルビン酸、及びチメロサールなどは、微生物のコンタミネーシヨンの防止 に対して使用可能である。また、糖、マンニトール、ソルビトールなどのポリアルコール 及び塩化ナトリウムのような等張性を保つ薬剤が組成物中に含まれてもよい。吸着を 遅らせることができる組成物には、モノステアリン酸アルミニウム及びゼラチンなどの 薬剤が含まれる。 Pharmaceutical compositions suitable for injection include sterile aqueous solutions (dispersible) or dispersion media and sterile powders to be prepared at the time of use. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, CREMOP HOR EL ™ (BASF, Parsippany, NJ), or phosphate buffered saline (PBS). When used as an injection, the composition must be sterile and must have sufficient fluidity to be administered using a syringe. The composition The product must be stable to chemical changes and corrosion during preparation and storage, and should prevent contamination from microorganisms such as bacteria and fungi. The carrier can be made using a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol), and suitable mixtures. For example, when a coating agent such as lectin is used, the required particle size is maintained in a dispersion medium, and an appropriate fluidity is maintained by using a surfactant. Various antibacterial and antifungal agents, such as parabens, chlorobutanol, phenol, ascorbic acid, and thimerosal, can be used to prevent microbial contamination. In addition, polyalcohols such as sugar, mannitol, sorbitol, and agents that maintain isotonicity such as sodium chloride may be included in the composition. Compositions that can delay adsorption include agents such as aluminum monostearate and gelatin.
[0063] 滅菌的な注射可能溶液は、必要な成分を単独で、又は他の成分と組み合わせた 後に、適切な溶媒中に必要量の活性化合物を加え、滅菌することで調製される。一 般に、分散媒は、基本的な分散培地及び上述したその他の必要成分を含む滅菌的 媒体中に活性化合物を取り込むことにより調製される。滅菌的な注射可能な溶液の 調製のための滅菌的なパウダーの調製方法には、活性な成分及び滅菌溶液に由来 する何れかの所望な成分を含むパウダーを調製する真空乾燥及び凍結乾燥が含ま れる。  [0063] Sterile injectable solutions are prepared by sterilizing the required ingredient alone or in combination with the other ingredients and then adding the required amount of the active compound in a suitable solvent. In general, dispersion media are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the other necessary ingredients discussed above. Sterile powder preparation methods for the preparation of sterile injectable solutions include vacuum drying and lyophilization to prepare a powder containing the active ingredient and any desired ingredients derived from the sterile solution. It is.
[0064] 12— 2.経口組成物  [0064] 12— 2. Oral composition
通常、経口組成物には、不活性な希釈剤又は体内に取り込んでも害を及ぼさない 担体が含まれる。経口組成物には、例えば、ゼラチンのカプセル剤に包含されるか、 加圧されて錠剤化される。経口的治療のためには、活性化合物は賦形剤と共に取り 込まれ、錠剤、トローチ又はカプセル剤の形態で使用される。また、経口組成物は、 流動性担体を用いて調製することも可能であり、流動性担体中の該組成物は経口的 に適用される。さらに、薬剤的に適合する結合剤、及び Z又はアジュバント物質など が包含されてもよい。  Oral compositions usually include an inert diluent or a carrier that does not cause harm when taken into the body. Oral compositions are, for example, contained in gelatin capsules or compressed into tablets. For oral treatment, the active compound is incorporated with excipients and used in the form of tablets, troches, or capsules. The oral composition can also be prepared using a fluid carrier, and the composition in the fluid carrier is applied orally. In addition, pharmaceutically compatible binding agents, and Z or adjuvant substances may be included.
錠剤、丸薬、カプセル剤、トローチ及びその類似物は以下の成分又は類似の性質 を持つ化合物の何れかを含み得る:微結晶性セルロースのような賦形剤、アラビアゴ ム、トラガント又はゼラチンなどの結合剤;スターチ又はラタトースなどの、アルギン酸 、 PRIM〇GEL、又はコーンスターチなどの膨化剤;ステアリン酸マグネシウム又は S TRROTESなどの潤滑剤;コロイド性シリコン二酸化物などの滑剤;スクロース又はサ ッカリンなどの甘味剤;又はペパーミント、メチルサリチル酸又はオレンジフレイバーな どの香料添加剤。 Tablets, pills, capsules, troches and the like have the following ingredients or similar properties Can include any of the following compounds: excipients such as microcrystalline cellulose, binders such as gum arabic, tragacanth or gelatin; bulking agents such as alginic acid, PRIM GEL, or corn starch, such as starch or ratatose Lubricants such as magnesium stearate or STRROTES; lubricants such as colloidal silicon dioxide; sweeteners such as sucrose or saccharin; or flavoring agents such as peppermint, methyl salicylic acid or orange flavor.
[0065] 12- 3.担体  [0065] 12- 3. Carrier
上記 cgrl 1ポリヌクレオチドなどは、植込錠及びマイクロカプセルに封入された送達 システムなどの徐放性製剤として、体内から即時に除去されることを防ぎ得る担体を 用いて調製することができる。エチレンビュル酢酸塩、ポリ酸無水物、ポリダリコール 酸、コラーゲン、ポリオルトエステル、及びポリ乳酸などの、生物分解性、生物適合性 ポリマーを用いることができる。このような材料は、 ALZA Corporation (Mountain View , CA)及び NOVA Pharmaceuticals, Inc. (Lake Elsinore, CA)などから入手することが 可能で、また、当業者によって容易に調製することもできる。また、リボソームの懸濁 液も薬剤的に受容可能な坦体として使用することができる。有用なリボソームは、限 定はしないが、ホスファチジルコリン、コレステロール及び PEG誘導ホスファチジルェ タノール (PEG— PE)を含む脂質組成物として、使用に適するサイズになるように、 適当なポアサイズのフィルターを通して調製され、逆相蒸発法によって精製される。 例えば、抗体の Fab'断片などは、ジスルフイド交換反応を介して、リボソームに結合 させてもょレ、(Martin及び Papahadjopoulos, 1982)。  The cgrl 1 polynucleotide and the like can be prepared as a sustained-release preparation such as a delivery system encapsulated in implantable tablets and microcapsules using a carrier that can prevent immediate removal from the body. Biodegradable, biocompatible polymers can be used, such as ethylene butyrate, polyanhydrides, polydaricol acid, collagen, polyorthoesters, and polylactic acid. Such materials are available from ALZA Corporation (Mountain View, CA) and NOVA Pharmaceuticals, Inc. (Lake Elsinore, CA), and can also be readily prepared by those skilled in the art. Ribosome suspensions can also be used as pharmaceutically acceptable carriers. Useful ribosomes are prepared as a lipid composition containing, but not limited to, phosphatidylcholine, cholesterol and PEG-derived phosphatidylethanol (PEG-PE) through a filter of appropriate pore size so that it is sized for use, Purified by reverse phase evaporation. For example, Fab ′ fragments of antibodies can be bound to ribosomes via a disulfide exchange reaction (Martin and Papahadjopoulos, 1982).
[0066] 12-4.投与量  [0066] 12-4. Dosage
上記 cgrl 1ポリペプチドの又は該ポリペプチドをコードする遺伝子等による特定の 疾患の治療又は予防において、適切な投与量レベルは、投与される患者の状態、投 与方法等に依存するが、当業者であれば、容易に最適化することが可能である。 注射投与の場合は、例えば、一日に患者の体重あたり約 0. 1 μ gZkgから約 500 mgZkgを投与するのが好ましぐ一般に一回又は複数回に分けて投与され得るで あろう。好ましくは、投与量レベルは、一日に約 0. 1 μ g/kgから約 250mgZkgであ り、より好ましくは一曰に約 0. 5〜約 lOOmgZkgである。 経口投与の場合は、組成物は、好ましくは 1. 0から lOOOmgの活性成分を含む錠 斉 IJの形態で提供され、好ましくは活十生成分力 . 0, 5. 0, 10. 0, 15. 0, 20. 0, 25 . 0, 50. 0, 75. 0, 100. 0, 150. 0, 200. 0, 250. 0, 300. 0, 400. 0, 500. 0 , 600. 0, 750. 0, 800. 0, 900. 0及び 1000. Omgである。ィ匕合物は一日に:!〜 4 回の投与計画で、好ましくは一日に一回又は二回投与される。 In the treatment or prevention of a specific disease using the cgrl 1 polypeptide or a gene encoding the polypeptide, the appropriate dosage level depends on the condition of the patient to be administered, the administration method, etc. If so, it can be easily optimized. In the case of injection administration, for example, it is generally preferred to administer from about 0.1 μg Zkg to about 500 mg Zkg per day of the patient's body weight, and would generally be administered in one or more divided doses. Preferably, the dosage level is from about 0.1 μg / kg to about 250 mgZkg per day, more preferably from about 0.5 to about 10 mg / kg. For oral administration, the composition is preferably provided in the form of a tablet IJ containing preferably 1.0 to 10 mg of the active ingredient, and preferably has a productive force of .0, 5. 0, 10. 0, 15 0, 20. 0, 25. 0, 50. 0, 75. 0, 100. 0, 150. 0, 200. 0, 250. 0, 300. 0, 400. 0, 500. 0, 600. 0 , 750.0, 800.0, 900.0 and 1000. Omg. The compound is administered on a regimen of! To 4 times a day, preferably once or twice a day.
[0067] 12- 5.単位投与量 [0067] 12-5. Unit dosage
医薬組成物又は製剤は、一定の投与量を保障すベぐ均一単位投与量により構成 されなくてはならない。単位投与量は、患者の治療に有効な一回の投与量を含み、 薬剤的に受容可能な担体と共に製剤化された一単位のことである。本発明の単位投 与量を決定する場合には、製剤化される化合物の物理的、化学的特徴、期待される 治療上の効果、及び該化合物に特有な製剤化における留意事項等により影響を受 ける。  A pharmaceutical composition or formulation should consist of uniform unit doses that ensure a certain dose. A unit dose is a unit formulated with a pharmaceutically acceptable carrier, including a single dose effective for treating a patient. When determining the unit dosage of the present invention, it is influenced by the physical and chemical characteristics of the compound to be formulated, the expected therapeutic effect, and the formulation considerations specific to the compound. Receive.
[0068] 12- 6.遺伝子治療組成物  [0068] 12- 6. Gene Therapy Composition
本発明において開示される核酸分子(例えば、 cgrl lポリヌクレオチド、該ポリヌクレ ォチドが挿入されたベクター、 cgrl lポリヌクレオチドに対するアンチセンス核酸も含 む)を患者の細胞に導入する方法には、主としてインビボ又はェキソビボの 2つの方 法がある。インビボ送達においては、治療が必要とされる患者の部位に直接注入され る。ェキソビボ処理では、患者の治療が意図される部位の細胞を単離し、単離された 細胞に製剤化した核酸分子を導入し、導入された細胞を患者に直接又は、例えば、 患者に埋め込まれる多孔性膜にカプセル化して投与することができる(米国特許第 4, The method for introducing a nucleic acid molecule disclosed in the present invention (for example, a cgrl polynucleotide, a vector having the polynucleotide inserted therein, and an antisense nucleic acid against the cgrl polynucleotide) into a patient's cells is mainly used in vivo. Or there are two ways of ex vivo. For in vivo delivery, it is injected directly into the site of the patient in need of treatment. In ex vivo treatment, cells at a site intended for treatment of a patient are isolated, the formulated nucleic acid molecule is introduced into the isolated cells, and the introduced cells are porous directly into the patient or, for example, implanted in the patient. Can be administered in an encapsulated membrane (US Pat. No. 4,
892,538号及び第 5,283, 187号参照)。核酸分子を生細胞に導入するために利用可 能な技術は、培養細胞等にインビトロで導入される力 又は患者にインビボで導入す るかに依存して選択される。哺乳動物細胞にインビトロで核酸分子を導入するのに適 した技術としては、リボソーム、エレクト口ポレーシヨン、マイクロインジェクション、トラン スフヱクシヨン、細胞融合、 DEAE—デキストラン法、リン酸カルシウム沈降法などが 挙げられる。トランスフエクシヨンには、組換えウィルス(好ましくはレトロウイルス)粒子 の細胞レセプターとの結合、次いで粒子に含まれる核酸分子の細胞への導入が含ま れる。遺伝子のェキソビボ送達に通常用いられるベクターはレトロウイルスである。 [0069] 現在、インビボ核酸移入技術で好ましいのは、ウィルス又は非ウィルスベクター(ァ デノウィルス、レンチウィルス、単純へルぺス Iウィルス、又はアデノ関連ウィルス(AA V) )、及びカチオン性脂質ベースの系 (遺伝子の脂質媒介移入に有用な脂質は、例 えば、 D〇TMA、 DOPE,及び DC_Cholである;例えば、 Tonkinson等, Cancer In vestigation, 14(1):54_65 (1996)参照)を利用した系が含まれる。遺伝子治療で使用 するために最も好ましいベクターはウィルスでありその中でも、最も好ましくはアデノウ ィルス、 AAV、レンチウィルス又はレトロウイルスである。レトロウイルスベクター等の ウィルスベクターには、少なくとも 1つの転写プロモーター/ェンハンサー又は位置 決定因子などが含まれる。さらに、レトロウイルスベクター等のウィルスベクターは、例 えば、ナルコレプシ一関連遺伝子を含んだ状態で転写される場合、該コード化遺伝 子の翻訳を可能とするシスエレメント、即ち翻訳開始配列として機能する核酸配列を 含む。このようなベクター構築物は、用いるウィルスに適したパッケージングシグナル 、末端反復配列(LTR)又はその一部を含む。さらに、これらのベクターには、通常、 該ベクターを含む宿主細胞から発現ポリペプチドを分泌させるシグナル配列が含ま れる。好ましくは、この目的のためのシグナル配列は哺乳動物シグナル配列である。 場合によっては、ベクター構築物は、ポリアデニルイ匕並びに翻訳終結配歹 IJも含む。 例えば、 5' LTR、 tRNA結合部位、パッケージングシグナル、 DNA合成の開始点、 及び 3' LTR又はその一部を含む。非ウィルス性の他のベクターは、例えばカチオン 性脂質、ポリリジン、及びデンドリマーを用いることもできる。 892,538 and 5,283,187). The technique available for introducing a nucleic acid molecule into a living cell is selected depending on the force introduced in vitro into cultured cells or the like or whether it is introduced into a patient in vivo. Suitable techniques for introducing nucleic acid molecules into mammalian cells in vitro include ribosomes, electopore position, microinjection, transfusion, cell fusion, the DEAE-dextran method, and the calcium phosphate precipitation method. Transfection involves the binding of a recombinant viral (preferably retroviral) particle to a cellular receptor, followed by introduction of the nucleic acid molecule contained in the particle into the cell. A commonly used vector for ex vivo delivery of genes is a retrovirus. [0069] Currently preferred in vivo nucleic acid transfer techniques are viral or non-viral vectors (adenoviruses, lentiviruses, herpes simplex virus I, or adeno-associated viruses (AAV)), and cationic lipid-based Lipids (useful for lipid-mediated transfer of genes are eg D0TMA, DOPE, and DC_Chol; see, for example, Tonkinson et al., Cancer Investigation, 14 (1): 54_65 (1996)) The system is included. The most preferred vector for use in gene therapy is a virus, among which is most preferably an adenovirus, AAV, lentivirus or retrovirus. Viral vectors, such as retroviral vectors, include at least one transcription promoter / enhancer or positioning factor. In addition, a viral vector such as a retroviral vector, for example, a nucleic acid that functions as a cis element, that is, a translation initiation sequence, enables translation of the encoded gene when it is transcribed in a state containing a narcolepsy-related gene. Contains an array. Such vector constructs contain a packaging signal, a terminal repeat (LTR) or part thereof suitable for the virus used. In addition, these vectors usually contain a signal sequence that causes the expressed polypeptide to be secreted from the host cell containing the vector. Preferably, the signal sequence for this purpose is a mammalian signal sequence. In some cases, the vector construct also includes a polyadenylation as well as a translation termination marker IJ. For example, it includes a 5 ′ LTR, a tRNA binding site, a packaging signal, an initiation point for DNA synthesis, and a 3 ′ LTR or a portion thereof. Other non-viral vectors can use, for example, cationic lipids, polylysine, and dendrimers.
場合によっては、治療に用いる核酸を目的の細胞にターゲテイングする試薬、例え ば、細胞表面膜タンパク質に特異的な抗体、又は標的細胞上のレセプターのリガン ドなどと共に提供するのが望ましい。現在知られている遺伝子標識化及び遺伝子治 療プロトコールの概説については、 Anderson等, Science, 256:808-813 (1992)などを 参照のこと。  In some cases, it may be desirable to provide the nucleic acid used for treatment with a reagent that targets the cell of interest, such as an antibody specific for a cell surface membrane protein or a receptor ligand on the target cell. For a review of currently known gene labeling and gene therapy protocols, see Anderson et al., Science, 256: 808-813 (1992).
[0070] 13.医薬組成物に関するキット  [0070] 13. Kit for pharmaceutical composition
医薬組成物はキット、容器、パック中に投与の説明書と共に含めることができる。本 発明に係る医薬組成物がキットとして供給される場合、該医薬組成物のうち異なる構 成成分が別々の容器中に包装され、使用直前に混合される。このように構成成分を 別々に包装するのは、活性構成成分の機能を失うことなく長期間の貯蔵を可能にす るためである。 The pharmaceutical composition can be included in the kit, container, pack along with instructions for administration. When the pharmaceutical composition according to the present invention is supplied as a kit, different components of the pharmaceutical composition are packaged in separate containers and mixed immediately before use. In this way the components They are packaged separately to allow long-term storage without losing the function of the active component.
[0071] 13— 1.容器  [0071] 13— 1. Container
キット中に含まれる試薬は、構成成分が活性を長期間有効に持続し、容器の材質 によって吸着されず、変質を受けないような何れかの種類の容器中に供給される。例 えば、封着されたガラスアンプルは、窒素ガスのような中性で不反応性ガスの下にお いて包装されたバッファーを含む。アンプルは、ガラス、ポリカーボネート、ポリスチレ ンなどの有機ポリマー、セラミック、金属、又は試薬を保持するために通常用いられる 他の何れかの適切な材料などから構成される。他の適切な容器の例には、アンプル などの類似物質から作られる簡単なボトル、及び内部がアルミニウム又は合金などの ホイルで裏打ちされた包装材が含まれる。他の容器には、試験管、ノィアル、フラス コ、ボトル、シリンジ、又はその類似物が含まれる。容器は、皮下用注射針で貫通可 能なストッパーを有するボトノレなどの無菌のアクセスポートを有する。  Reagents contained in the kit are supplied in any type of container in which the components remain active for an extended period of time, are not adsorbed by the container material, and are not subject to alteration. For example, a sealed glass ampoule contains a buffer packaged under a neutral, non-reactive gas such as nitrogen gas. Ampoules are composed of organic polymers such as glass, polycarbonate, polystyrene, ceramics, metals, or any other suitable material commonly used to hold reagents. Examples of other suitable containers include simple bottles made from similar materials such as ampoules, and packaging materials that are internally lined with foil such as aluminum or alloys. Other containers include test tubes, nozzles, flasks, bottles, syringes, or the like. The container has a sterile access port such as a bot- nore with a stopper that can be penetrated by a hypodermic needle.
[0072] 13- 2.使用説明書  [0072] 13- 2. Instructions for use
また、キットには使用説明書も添付される。当該医薬組成物からな成るキットの使用 説明は、紙又は他の材質上に印刷され、及び/又はフロッピー(登録商標)ディスク 、 CD-ROM, DVD-ROM, Zipディスク、ビデオテープ、オーディオテープなどの 電気的又は電磁的に読み取り可能な媒体として供給されてもよい。詳細な使用説明 は、キット内に実際に添付されていてもよぐあるいは、キットの製造者又は分配者に よって指定され又は電子メール等で通知されるウェブサイトに掲載されていてもよレ、。  The kit also includes instructions for use. Instructions for using the kit comprising the pharmaceutical composition are printed on paper or other material and / or floppy disk, CD-ROM, DVD-ROM, Zip disk, video tape, audio tape, etc. It may be supplied as an electrically or electromagnetically readable medium. Detailed instructions for use may actually be included in the kit or may be posted on a website designated by the kit manufacturer or distributor or notified by e-mail, etc. .
[0073] 14.診断  [0073] 14. Diagnosis
ある細胞内において、 cgrl lポリヌクレオチド又はポリペプチドの発現、及び/又は 活性に異常が認められる場合には、該細胞の増殖、生存維持に異常を来している危 険性が高いものと判断することができる。この場合、 cgrl l遺伝子(エタソン領域のみ ならずイントロン領域、およびプロモーター領域も含む)又は cgrl lポリペプチドの変 異 (例えば、点突然変異、欠失など)、該遺伝子の発現制御領域の変異の有無を調 ベることによって、上記異常の発症およびその発症の危険性を予測することができる [0074] cgrl lポリペプチドの発現量を調べるには、該ポリペプチドに特異的な抗体を利用 すること力 Sできる。例えば、特定の細胞の増殖、生存維持に異常を来していることが 疑われる患者から被検試料を取得し、該被検試料に前記抗体を接触させ、該被検試 料と該抗体との結合の有無を検出することで、該細胞の増殖、生存維持における異 常の発症または発症の可能性を調べることができる。被検試料と抗体との結合の有 無は、抗体を用いた免疫沈降法、ウェスタンブロッテイング法、免役組織化学法、 ELI SA法などにより確認することができる。確認の結果、本発明のポリペプチドが全く検 出されないか、または著しく少ない生体内存在量であると判断される場合には、上記 異常の発症が疑われる。 If there is an abnormality in the expression and / or activity of cgrl l polynucleotide or polypeptide in a cell, it is judged that there is a high risk of an abnormality in the proliferation and survival of the cell. can do. In this case, the cgrl l gene (including not only the etason region but also the intron region and the promoter region) or cgrl l polypeptide variation (eg, point mutation, deletion, etc.), mutation in the expression control region of the gene By investigating the presence or absence, the onset of the above abnormalities and the risk of the onset can be predicted [0074] In order to examine the expression level of a cglrl polypeptide, it is possible to use an antibody specific for the polypeptide. For example, a test sample is obtained from a patient suspected of having an abnormality in the proliferation and survival of specific cells, the antibody is brought into contact with the test sample, and the test sample and the antibody By detecting the presence or absence of binding, it is possible to examine the onset or possibility of onset of abnormalities in the proliferation and survival of the cells. The presence or absence of binding between the test sample and the antibody can be confirmed by immunoprecipitation using antibodies, Western blotting, immunohistochemistry, ELI SA, and the like. As a result of the confirmation, if the polypeptide of the present invention is not detected at all or it is determined that the polypeptide is present in an extremely small amount in the living body, the onset of the abnormality is suspected.
[0075] cgrl 1遺伝子の異常を確認するには、該遺伝子の cDNA配列、ゲノム DNA配列( 転写制御領域、イントロン領域なども含む)またはこれらの相補鎖とァニールすること ができるプライマー、プローブなどを利用することができる。利用可能なプライマーと しては、一般に、 15bp〜: !OObpであり、好まし <は、 17bp〜30bpの長さを有し、本 発明の遺伝子のコード領域、非コード領域 (転写制御領域、イントロン領域などを含 む)の少なくとも部分領域を増幅することができるものであれば如何なるものであって も利用可能である。  [0075] In order to confirm abnormalities in the cgrl 1 gene, primers, probes, etc. that can anneal with the cDNA sequence, genomic DNA sequence (including transcriptional control region, intron region, etc.) of these genes or their complementary strands are used. Can be used. Primers that can be used are generally 15 bp to: OO bp, and preferably <has a length of 17 bp to 30 bp. The coding region of the gene of the present invention, the non-coding region (transcriptional control region, Any material can be used as long as it can amplify at least a partial region (including an intron region).
[0076] 利用可能なプローブとしては、一般に、 15bp以上の長さを有し、本発明の遺伝子 のコード領域、非コード領域 (転写制御領域、イントロン領域などを含む)の少なくとも 部分領域とハイブリダィズすることができるものであれば如何なるものであっても利用 可能である。また、プローブが目的の DNA領域にハイブリダィズすることを確認する ために、該プローブは蛍光色素、放射標識などにより検出可能な状態で利用するこ とができる。  [0076] The probe that can be used generally has a length of 15 bp or more, and hybridizes with at least a partial region of the coding region and non-coding region (including transcription control region, intron region, etc.) of the gene of the present invention. Anything that can be used can be used. Further, in order to confirm that the probe hybridizes to the target DNA region, the probe can be used in a state where it can be detected by a fluorescent dye, a radiolabel or the like.
[0077] cgrl 1遺伝子の機能異常を確認する他の方法には、例えば、被検者から被検試料 を取得し、該被検試料から調製した cgrl l遺伝子の mRNA、または該 mRNA力 調 製した cDNAなどに前記プローブ、プライマーなどを接触させ、本発明の遺伝子のコ ード領域、非コード領域 (転写制御領域、イントロン領域などを含む)の少なくとも部 分領域との結合を検出する工程、または該部分領域を増幅する工程が含まれる。検 出の結果、本発明の遺伝子の発現量等を健常者由来の試料と比較して異常が認め られる場合には、 cgrl l遺伝子の機能異常の発症又はその危険性を予測することが できる。 [0077] Other methods for confirming functional abnormality of the cgrl 1 gene include, for example, obtaining a test sample from a subject and preparing the mRNA of the cgrl 1 gene prepared from the test sample, or preparing the mRNA force thereof. A step of detecting the binding of at least a partial region of the coding region and non-coding region (including transcription control region, intron region, etc.) of the gene of the present invention by contacting the probe, primer, etc. Alternatively, a step of amplifying the partial region is included. As a result of the detection, abnormalities were found in the expression level of the gene of the present invention compared to samples derived from healthy subjects. If possible, the onset or risk of dysfunction of the cgrl l gene can be predicted.
ここで、診断の対象となるのはヒトのみならず「哺乳動物」全般である。「哺乳動物」と は、哺乳類に分類される任意の動物を意味し、特に限定はしないが、例えば、ヒトの 他、ィヌ、ネコなどのペット動物、ゥシ、ブタ、ヒッジ、ゥマなどの家畜動物などのことで ある。特に好ましい「哺乳動物」は、ヒトである。  Here, not only humans but also “mammals” in general are targeted for diagnosis. “Mammal” means any animal classified as a mammal, and is not particularly limited. For example, in addition to humans, pet animals such as dogs and cats, ushi, pigs, hidges, horses, etc. It is a domestic animal. Particularly preferred “mammals” are humans.
[0078] 15.治療 [0078] 15. Treatment
本発明には、疾患に罹患した、又は罹患する危険性のある哺乳動物の該疾患に関 する予防又は治療方法が含まれる。  The present invention includes a method for preventing or treating a diseased mammal suffering from or at risk of suffering from the disease.
ここで、「治療」とは、疾患に罹患するおそれがあるか又は罹患した哺乳動物におい て、該疾患の病態の進行を阻止又は緩和することを意味し、治療的処置のみならず 予防的処置をも含む広い意味として使用される。また、「疾患」には、 cgrl l遺伝子又 はポリペプチドの存否又は量的変動によって引き起こされる病態の全てがふくまれ、 例えば、血流障害、発育障害関連疾患、神経疾患、癌、低血圧症、高血圧症、心臓 疾患などが含まれる。  Here, “treatment” means to prevent or alleviate the progression of the pathological condition of a disease in a mammal that is likely to suffer from or suffers from the disease. It is used as a broad meaning including. In addition, “disease” includes all pathological conditions caused by the presence or quantitative fluctuation of cgrl l gene or polypeptide, such as blood flow disorder, developmental disorder-related disease, neurological disease, cancer, hypotension. , Hypertension, and heart disease.
ここで、治療の対象となるのはヒトのみならず「哺乳動物」全般である。「哺乳動物」と は、哺乳類に分類される任意の動物を意味し、特に限定はしないが、例えば、ヒトの 他、ィヌ、ネコなどのペット動物、ゥシ、ブタ、ヒッジ、ゥマなどの家畜動物などのことで ある。特に好ましい「哺乳動物」は、ヒトである。  Here, not only humans but also “mammals” in general are targeted for treatment. “Mammal” means any animal classified as a mammal, and is not particularly limited. For example, in addition to humans, pet animals such as dogs and cats, ushi, pigs, hidges, horses, etc. It is a domestic animal. Particularly preferred “mammals” are humans.
[0079] 以下に実施例を示すが、本発明はこれに限定されるものではない。 [0079] Examples are shown below, but the present invention is not limited thereto.
実施例  Example
[0080] 1. cgrl lポリペプチドの化学的性質:  [0080] 1. Chemical properties of cgrl l polypeptide:
ホルモン産生能の異なる下垂体細胞間の異なるタンパク質のプロテオーム解析か ら特定のタンパク質を同定した(図 1 , 2)。具体的には、下垂体腫瘍 (ラット由来)から 分離されたクローン細胞のうち、ホルモンを産生しない MtT/E— B3細胞とホルモン を産生する MtT/S細胞の細胞抽出液の 2次元電気泳動を行った。この結果図 1に 示すようにホルモン産生の MtT/S細胞で特異的に発現するスポット 2を同定した。 このスポットのトリプシン分解ペプチドの質量分析から図 2のパターンを得た。このべ プチドパターン相当するアミノ酸配列をコードする遺伝子として cgrl lが同定された。 図 1の spot2に対応するタンパク質をコードする遺伝子は cgr 11で EF-hand motif を有し、 p53により細胞増殖を抑制することが報告されていたものである。本発明に おいて、このタンパク質(cgrl lポリペプチド)が分泌シグナルを持ち、分泌シグナル の位置で切られること、分子量の実測値と遺伝子力 推定されるタンパク質の分子量 が予想される値よりも大きいことから糖による修飾を受けていることが推察された。この ことは従来の報告になぐ本タンパク質が新規の分泌タンパク質であることを示してい る。図 3には、ラットとヒト由来の cgrl lポリペプチドのアミノ酸配列のァライメントを示 す。 Specific proteins were identified from proteomic analysis of different proteins between pituitary cells with different hormone production capacities (Figs. 1 and 2). Specifically, of the clonal cells isolated from pituitary tumors (derived from rats), two-dimensional electrophoresis of cell extracts of MtT / E-B3 cells that do not produce hormone and MtT / S cells that produce hormone is performed. went. As a result, spot 2 specifically expressed in hormone-producing MtT / S cells was identified as shown in FIG. The pattern of FIG. 2 was obtained from the mass analysis of the tryptic peptide at this spot. This Cgrl l was identified as a gene encoding the amino acid sequence corresponding to the peptide pattern. The gene encoding the protein corresponding to spot2 in Fig. 1 is cgr 11 and has an EF-hand motif, and it has been reported that p53 suppresses cell proliferation. In the present invention, this protein (cglrl polypeptide) has a secretion signal and is cleaved at the position of the secretion signal, and the measured molecular weight and the estimated molecular force of the protein are larger than expected. Therefore, it was guessed that it was modified with sugar. This indicates that this protein is a new secreted protein compared to previous reports. Figure 3 shows the alignment of the amino acid sequences of rat and human derived cglrl polypeptides.
[0081] 上述したように cgrl lポリペプチドのアミノ酸配列には分泌シグナル配列の特徴が みとめられるため、シグナルペプチド予測プログラム SOSUIsignal (http: //sosui . proteome. bio. tuat. ac. jp/ sosmsignal/ sosuisignal― submit, html)を 用いてシグナル領域の予測を行ったところ、 21残基からなるアミノ酸配列が予測され た(図 4)。この結果は他の予測プログラムである SignalP (http : //www. cbs. dt u. dk/ services/ SignalP/)でも同し あった。  [0081] As described above, since the amino acid sequence of cgrl l polypeptide has a characteristic of the secretory signal sequence, the signal peptide prediction program SOSUIsignal (http: //sosui.proteome.bio.tuat.ac.jp/sosmsignal/ When the signal region was predicted using sosuisignal-submit, html), an amino acid sequence consisting of 21 residues was predicted (Fig. 4). This result was the same for SignalP (http://www.cbs.dtu.dk/services/SignalP/), which is another prediction program.
し力 ながら、ラット cgrl lを細胞内に強制発現させ(図 10に示すコンストラクトを使 用)、培養上清からァフィ二ティー精製した cgrl lタンパク質の N末端アミノ酸配列を 、定法に従って解析したところ、シグナルペプチド領域 29アミノ酸配列から構成され ることが明らかになった(図 5)。この知見は、従来から知られているシグナルペプチド 配列の切断ルールとは異なるものである。従って、この結果は、 cgrl lタンパク質の i n vivoにおける機能領域を初めて明らかにしたもので、重要な知見である。  However, rat cgrl l was forcibly expressed in the cells (using the construct shown in FIG. 10), and the N-terminal amino acid sequence of the cgrl l protein purified from the culture supernatant was analyzed according to a conventional method. It was revealed that the signal peptide region is composed of 29 amino acid sequences (Fig. 5). This finding is different from the conventionally known cleavage rules for signal peptide sequences. Therefore, this result reveals the in vivo functional region of cgrl l protein for the first time and is an important finding.
[0082] さらに、発明者は、 cgrl l遺伝子のプロモーター領域の解析を行ったところ、低酸 素反応エレメント(HRE ; hypoxia response element)の存在を明らかにした(図 6)。この DNAエレメントには共通のコンセンサス配列が存在しており、低酸素状態に 応答し、 HIF—1 (hypoxia inducible factor- 1)などの転写因子がこのエレメント に作用し、遺伝子の転写制御を行っていることが明らかになつている。低酸素状態で 誘導される因子として、血管内皮細胞増殖因子 (VEGF)など、血管新生、癌化に関 与している因子などが報告されている。このことにより、プロモーター領域に低酸素反 応エレメントを持つ cgr 11遺伝子は、低酸素に反応し血管新生、血管透過性、の促 進により腫瘍形成、組織の維持にかかわることが示唆される。 [0082] Furthermore, the inventor analyzed the promoter region of the cglrl gene and revealed the presence of a hypoxia response element (HRE) (Fig. 6). This DNA element has a common consensus sequence. In response to hypoxia, transcription factors such as HIF-1 (hypoxia inducible factor-1) act on this element to regulate gene transcription. It is clear that Factors involved in angiogenesis and canceration, such as vascular endothelial growth factor (VEGF), have been reported as factors induced in hypoxia. This allows the hypoxic reaction in the promoter region. It is suggested that the cgr11 gene having a response element is involved in tumor formation and tissue maintenance by promoting angiogenesis and vascular permeability in response to hypoxia.
[0083] 2.抗体の作成:  [0083] 2. Antibody production:
遺伝子配列上から推定されるアミノ酸配列の親水性部位に対するペプチドを合成 し抗体を作成した。この結果、図 3及び図 5に存在する部位(antigenic siteとして 示す配列部位)が抗原として有効であり、特異的な抗体の作成に成功した。この部位 は今後 cgr 11の存在量の測定などに応用できる。  An antibody was prepared by synthesizing a peptide against the hydrophilic site of the amino acid sequence deduced from the gene sequence. As a result, the site (sequence site shown as an antigenic site) present in FIGS. 3 and 5 was effective as an antigen, and a specific antibody was successfully produced. This part can be applied to measure the abundance of cgr 11 in the future.
[0084] 3. cgrl lの産生細胞と組織内分布:  [0084] 3. cgrl l producing cells and tissue distribution:
PCR法により cgrl 1が下垂体のホルモン産生細胞(MtTZS)に特異的に発現す ることを示した(図 7)。このことから、 cgrl lタンパク質が細胞の分泌機能と関わりが強 レ、ことが示唆された。そして、 cgrl lタンパク質の存在量などを指標にすることにより、 生体内のホルモン等分泌能を測定するために使用され得ることも示唆された。  PCR showed that cgrl 1 was specifically expressed in pituitary hormone-producing cells (MtTZS) (Fig. 7). This suggests that cgrl l protein is strongly related to the secretory function of cells. It was also suggested that it can be used to measure the secretory ability of hormones in vivo by using the abundance of cgrl l protein as an index.
また、細胞レベルでは、 cgrl lタンパク質がゴルジ装置に特異的に存在することが 確認され(図 8)、 cgrl lポリヌクレオチドが分泌タンパク質であるとの上記見解が支持 された。さらに、その分布はホルモン産生細胞内の成長ホルモンの分布と一致してい た(図 8)。 cgrl 1タンパク質に特異的な抗体を使用した免疫細胞化学では cgrl 1タ ンパク質を持つ細胞は下垂体の後、前葉であり、下垂体中葉にはまったく発現されて いないことが分かった(図 9A)。このような発現の様式から、 cgrl lタンパク質が豊富 な血管を持つ部位 (下垂体後葉、前葉)に多く発現し、一方、血管を全く持たない部 位(下垂体中葉)には全く発現していないことが強く示唆された。このほか大腸(図 9C )および卵巣の成熟卵胞の外卵胞膜(図 9D)などにも cgrl 1タンパク質の発現が確 認された。これらの部位は、いずれも血管の豊富な部位であることから、 cgrl lタンパ ク質が血管系の形成及び/又は維持の重要な役割を担っていることが示唆される。  At the cellular level, it was confirmed that cgrl l protein was specifically present in the Golgi apparatus (Fig. 8), supporting the above view that cgrl l polynucleotide was a secreted protein. Furthermore, the distribution was consistent with the distribution of growth hormone in hormone-producing cells (Fig. 8). Immunocytochemistry using an antibody specific for cgrl 1 protein revealed that cells with cgrl 1 protein were the anterior lobe after the pituitary gland and were not expressed at all in the pituitary midlobe (Figure 9A). ). Because of this mode of expression, cgrl l protein is abundantly expressed in sites with abundant blood vessels (posterior pituitary, anterior lobe), while it is not expressed at all in the region without any blood vessels (pituitary midlobe). It was strongly suggested that there was not. In addition, the expression of cgrl 1 protein was also confirmed in the large follicle (Fig. 9C) and the outer follicular membrane of mature ovarian follicles (Fig. 9D). All of these sites are rich in blood vessels, suggesting that the cgrl l protein plays an important role in the formation and / or maintenance of the vasculature.
[0085] 4. cgr 11強制発現系: [0085] 4. cgr 11 forced expression system:
cgrl lタンパク質の機能を検討するために、細胞内強制発現系の確立を行った。 図 10には、発現ベクターのコンストラクトを示す。発現に用いたベクター pCl_neo Vector (Promega社)の EcoRI、 Xbalサイトに cgrl 1遺伝子を揷入し、定法に従って サブクローニングを行った。得られた発現ベクターは、 Lipofectamine 2000を用 いて下垂体由来細胞株、 MtT/E— B3細胞(理研バイオリソースセンター登録 MtT /E細胞: RCB1278のサブクローン細胞)および MtT/Se細胞(理研バイオリソー スセンター登録: RCB0529)に導入した。 In order to investigate the function of the cgrl l protein, an intracellular forced expression system was established. FIG. 10 shows the expression vector construct. The cgrl 1 gene was inserted into the EcoRI and Xbal sites of the vector pCl_neo Vector (Promega) used for expression, and subcloning was performed according to a conventional method. The resulting expression vector uses Lipofectamine 2000 It was introduced into pituitary-derived cell lines, MtT / E—B3 cells (MtT / E cells registered in RIKEN BioResource Center: subclone cells of RCB1278) and MtT / Se cells (registered RIKEN Bioresource Center: RCB0529).
cgrl l強制発現細胞において、 cgrl lタンパク質はゴノレジ装置に局在し(図 11 A, C)、強制発現細胞においても cgrl lタンパク質が分泌タンパク質として機能している ことが示唆された。さらに、培養液中に、 cgrl lタンパク質の存在が確認されたことか ら、 cgrl lタンパク質が分泌タンパク質であることが強く示唆された(図 12、 13)。  In cgrl l forced expression cells, the cgrl l protein was localized in the gonoredi apparatus (FIGS. 11A and C), suggesting that the cgrl l protein also functions as a secreted protein in the forced expression cells. Furthermore, the presence of cgrl l protein was confirmed in the culture medium, strongly suggesting that cgrl l protein is a secreted protein (Figs. 12 and 13).
[0086] 5. cgrl lの細胞増殖、生存維持に対する影響:  [0086] 5. Effect of cgrl l on cell proliferation and survival:
cgrl 1遺伝子産物の細胞増殖に対する影響を調べるために、 MtTZE— B3細胞 に cgrl lタンパク質を強制発現させ、得られた細胞をラット(Fischer344 :メス、 lOOg )の鼠径部皮下に移植(2xl06細胞 Z200 μ 1/ラット)した。移植後、 20日目に動物 を開腹し腫瘍を取り出し、その重量を測定した。 In order to examine the effect of cgrl 1 gene product on cell proliferation, cgrl l protein was forcibly expressed in MtTZE- B3 cells, and the obtained cells were transplanted subcutaneously into the groin of rats (Fischer344: female, lOOg) (2xl0 6 cells) Z200 μ 1 / rat). On the 20th day after transplantation, the animals were laparotomized, the tumor was removed, and its weight was measured.
その結果、強制発現系では腫瘍の形成が早く(図 14、 15)、血管の豊富な腫瘍を 形成した(図 14)。また、強制発現細胞はバッチ培養法による細胞の死滅期が遅延し た(図 16)。なお、ベクターのみを導入した細胞の分裂時間(DT)が 25. 7時間であ つたのに対し、 cgrl l強制発現細胞の分裂時間(DT)が 27. 7時間であり、細胞の増 殖速度には有意な差は認められなかった。  As a result, tumor formation was rapid in the forced expression system (Figs. 14 and 15) and tumors rich in blood vessels were formed (Fig. 14). In addition, forced death cells were delayed in the cell death phase by batch culture (Fig. 16). Note that the cell division rate (DT) of the cells into which only the vector was introduced was 25.7 hours, whereas the cell division time (DT) of cgrl l forced expression cells was 27.7 hours. There was no significant difference.
[0087] また、副腎の褐色細胞腫で神経前駆細胞として有名な PC12細胞の増殖及び生存 維持に対する cgrl lタンパク質の影響を検討した。 PC12細胞は、無血清培地下で は生存できなレ、ことが知られてレ、る。この細胞では cgrl 1タンパク質の発現が少なレヽ ため、 cgrl l遺伝子を導入した PC12細胞に導入し、強制発現系を構築した。 48ゥェ ルプレートに強制発現クローン(図 17 ;クローン C〜G)と強制発現させていないクロ ーン(図 17 ;コントロール)を 5. 00E + 4Zゥヱル(200 μ ΐΖゥエル)でまきこみ、 2日 後に無血清培地に交換した。無血清培養 8日目に生存細胞数をカウントした。その 結果、 cgrl lタンパク質が強制発現される PC12細胞は、無血清培地下においても その生存が顕著に促進されることが明らかになった(図 17)。  [0087] In addition, the effect of cgrl protein on the proliferation and maintenance of PC12 cells, which are well known as neural progenitor cells in adrenal pheochromocytoma, was examined. PC12 cells are known to be unable to survive in serum-free media. Since the expression of cgrl 1 protein is low in this cell, it was introduced into PC12 cells into which the cgrl 1 gene was introduced, and a forced expression system was constructed. Inoculate 48 well plates with a forced expression clone (Fig. 17; clones C to G) and a non-forced clone (Fig. 17; control) with 5.00E + 4Z (200 μΐΖ), 2 The serum-free medium was replaced after a day. The number of viable cells was counted on day 8 of serum-free culture. As a result, it was revealed that the survival of PC12 cells in which cgrl l protein was forcibly expressed was significantly promoted even in serum-free medium (Fig. 17).
[0088] さらに、無血清培地で培養した cgrl lタンパク質産生細胞もしくは強制発現系の細 胞の培養液を無血清培地で培養した PC12細胞に添加した場合の影響にっレ、て検 ff寸した(図 18)。 48ウエノレプレート ίこ 4. 00Ε + 3/ウエノレ(200 /i l/ウエノレ)で PC12 細胞をまきこみ、 2日後に無血清培地に交換した。交換時に、 cgrl l強制発現細胞( MtT/B3— So)及び強制発現させていない対照細胞(MtT/E— B3vector)を培 養した培養液を 0%、 1 %、 5%、 10%濃度になるように無血清培地にカ卩えた。 5日間 培養した後、細胞数をカウントした。その結果、濃度依存的に細胞数が増加し、その 生存が濃度依存的に促進されることが明らかとなった(図 18)。 [0088] Furthermore, the effect of adding a culture solution of cglrl protein-producing cells or forced expression cells cultured in serum-free medium to PC12 cells cultured in serum-free medium was examined. f It was sized (Fig. 18). PC12 cells were seeded with 48 uenore plate ίko 4.00 + 3 / uenore (200 / il / uenore) and replaced with serum-free medium after 2 days. At the time of exchange, the culture solution in which cgrl l forced expression cells (MtT / B3—So) and control cells not forcedly expressed (MtT / E—B3vector) were cultivated to 0%, 1%, 5%, 10% concentration It was placed in a serum-free medium. After culturing for 5 days, the number of cells was counted. As a result, it became clear that the number of cells increased in a concentration-dependent manner and that survival was promoted in a concentration-dependent manner (Fig. 18).
[0089] またさらに、ラット下垂体前葉由来の細胞を用いて同様の実験を行った(図 19)。ェ ストロゲン依存性下垂体前葉細胞(MtTZSe)を 1. 00E + 5/ウエノレ(200 μ lZゥヱ ノレ)でまきこみ、無血清 D/F培地に cgrl 1タンパク質を含む培養液(図 19; CGR11 を含む培養液)と含まなレ、培養液(図 19; CGR11を含まなレ、培養液)を添加して培 養を行った。その結果、エストロゲンの有無によらず、無血清培地下での細胞の生存 を顕著に促進した。 [0089] Furthermore, a similar experiment was performed using cells derived from the rat anterior pituitary gland (Fig. 19). Estrogen-dependent anterior pituitary cells (MtTZSe) were seeded with 1.00E + 5 / wenore (200 μlZ ヱ nore), and a medium containing cgrl 1 protein in serum-free D / F medium (Fig. 19; CGR11) The culture broth containing), the ladle contained, and the broth (FIG. 19; the pan containing CGR11, the broth) were added for cultivation. As a result, the survival of cells under serum-free medium was significantly promoted regardless of the presence or absence of estrogen.
[0090] 6. cgrl lの血管系に対する影響 [0090] 6. Effects of cgrl l on vascular system
cgrl lタンパク質が豊富な血管を持つ部位(下垂体後葉、前葉)に多く発現し、一 方、血管を全く持たない部位(下垂体中葉)には全く発現していないこと、また、大腸 (図 9C)及び卵巣の成熟卵胞の外卵胞膜(図 9D)などにも cgrl 1タンパク質の発現 が確認されたことから、 cgrl lタンパク質が血管系の形成及び/又は維持の重要な 役割を担っていることが示唆された。  cgrl l protein is highly expressed in sites with blood vessels rich in the posterior and anterior pituitary glands, while it is not expressed at all in sites without blood vessels (pituitary middle lobe) and the colon (Fig. 9c) and the outer follicular membrane of mature ovarian follicles (Fig. 9D) have also been confirmed to express cgrl 1 protein, and cgrl l protein plays an important role in the formation and / or maintenance of vasculature. It has been suggested.
そこで、 cgrl lの血圧に対する影響を調べた。  Therefore, the effect of cgrl l on blood pressure was examined.
まず、血圧測定用力ニューレの留置を行った。  First, a force neulet for blood pressure measurement was placed.
ラットをエーテルで麻酔した後、左大腿部を切開し、左大腿動脈を剥離した。ピンセ ットで左大腿動脈を固定することで止血した。左大腿動脈の近位側と遠位側に縫合 糸を通した。ゥエッケノレ剪刀で大腿動脈に穴をあけ、 500Uへパリン (持田製薬) Z生 理食塩水を満たした血圧測定用力ニューレの PE10側を揷入した。先に通しておい た縫合糸で力ニューレと血管を固定し、血液がチューブ内へ拍動しながら入るかを確 認した。力ニューレは切開した部位から皮下を通して後頭部に出した。このとき、力二 ユーレが折れないよう注意した。再度、血液が拍動しながら上がってくるか確認し、ス トッパー(長さ 2cm、直径 0. 66mm) (銀鱗 16号、 TRAY)で栓をした。皮下のチュー ブを固定するために後頭部の皮膚に接するチューブをガムテープで卷き、ガムテー プと皮膚を縫い、後頭部から外に出るチューブを 4cm程度に切断した。手術後 1日 後に動物の回復を待って非麻酔下で以下の方法で血圧を測定した。 After anesthetizing the rat with ether, the left thigh was incised and the left femoral artery was detached. Hemostasis was achieved by fixing the left femoral artery with tweezers. A suture was passed through the proximal and distal sides of the left femoral artery. A hole was made in the femoral artery with a Weeckenor's scissors, and the PE10 side of a blood pressure measuring force neule filled with 500 U of Parin (Mochida Pharmaceutical) Z saline was inserted. The force needle and the blood vessel were fixed with the suture thread that had been passed through earlier, and it was confirmed that blood entered the tube while pulsating. The force neulet was delivered to the back of the head through the subcutaneous site. At this time, I was careful not to break the power. Once again, it was confirmed that blood was rising while pulsating, and stoppered with a stopper (length 2 cm, diameter 0.66 mm) (Silver Scale No. 16, TRAY). Subcutaneous chew The tube that touched the skin on the back of the head was squeezed with gum tape to fix the hub, the gum tape and the skin were sewn, and the tube exiting from the back of the head was cut to about 4 cm. One day after the operation, the blood pressure was measured by the following method under non-anesthesia after the animal had recovered.
[0091] ラットをケージに入れ、血圧測定用力ニューレと延長チューブを接続し、トランスデュ ーサ(バイオリサーチ株式会社)につないだ。チューブ内を 10Uへパリン Z生理食塩 水で満たし、空気が入らないように気をつけた。また、力ニューレと延長チューブの接 続は、ステンレスパイプを用いた。また、血圧の測定および解析ソフトは、 PowerLab (バイオリサーチ株式会社)を用いた。  [0091] The rat was placed in a cage, a blood pressure measuring force neuron and an extension tube were connected, and connected to a transducer (Bioresearch Corporation). The tube was filled with 10 U of Parin Z physiological saline, and care was taken to prevent air from entering. Stainless steel pipes were used for the connection between the force neulet and the extension tube. In addition, PowerLab (Bioresearch Corporation) was used as blood pressure measurement and analysis software.
ラット腹腔内に cgrl 1タンパク質分泌細胞の培養液から抗体でァフィ二ティー精製 した cgrl lタンパク質(0. 1 μ g/500 μ 1)を注入し平均血圧の経時変化を測定した 。その結果、精製 cgrl lタンパク質を腹腔内に注入した後、血圧の上昇が認められ た(図 20)。これに対し、精製 cgrl lタンパク質を含まない培養液を用いたコントロー ルサンプノレを注入した場合には血圧の上昇が認められなかった。  The cgrl1 protein (0.1 μg / 500 μ1) purified with an antibody from the culture medium of cgrl1 protein-secreting cells was injected into the abdominal cavity of the rat, and the time course of mean blood pressure was measured. As a result, an increase in blood pressure was observed after the purified cgrl l protein was injected intraperitoneally (Fig. 20). On the other hand, no increase in blood pressure was observed when a control sample was injected using a culture solution containing no purified cgrl protein.
この結果から、 cgrl lタンパク質は血圧上昇効果を有することが明らかとなった。  From this result, it was revealed that cgrl l protein has a blood pressure increasing effect.
[0092] 参考文献  [0092] References
Ausuoel等,し urrent protocols in molecular Diology. John Wiley & Sons, New York. 1 987  Ausuoel et al., Urrent protocols in molecular Diology. John Wiley & Sons, New York. 1 987
Anderson等, Science, 256:808—813, 1992  Anderson et al., Science, 256: 808-813, 1992
Becker等, Methods. Enzymol., 194 : 182, 1990  Becker et al., Methods. Enzymol., 194: 182, 1990
Bondanszky等, Peptide Synthesis, Interscience Publishers, New York 199b  Bondanszky et al., Peptide Synthesis, Interscience Publishers, New York 199b
Chen及び Okayama, BioTechniques. 6:632-638, 1988  Chen and Okayama, BioTechniques. 6: 632-638, 1988
Cohen等, Pro Natl.Acad.Sci. USA 69 : 2110, 1972  Cohen et al., Pro Natl. Acad. Sci. USA 69: 2110, 1972
Elroy - Stein及び Moss, Pro Natl. Acad. Sci. USA 87:6743-6747, 1990  Elroy-Stein and Moss, Pro Natl. Acad. Sci. USA 87: 6743-6747, 1990
Lrennaro等: The science and practice of pharmacy. Lippincott, Williams & Wilkins, P hiladelphia, PA. 2000  Lrennaro et al: The science and practice of pharmacy. Lippincott, Williams & Wilkins, P hiladelphia, PA. 2000
Eroding等, Academic Press, San Diego. 492 pp. 1996  Eroding et al., Academic Press, San Diego. 492 pp. 1996
Harlow及び Lane, Antibodies: A laboratory manual. Cold Spring Harbor Laboratory Press, Cold Spring Harbor. 726 pp, 1988 Harlow及び Lane, Using antibodies: A laboratory manual. Cold Spring Harbor Labor atory Press, Cold Spring Harbor, New York. 1999 Harlow and Lane, Antibodies: A laboratory manual. Cold Spring Harbor Laboratory Press, Cold Spring Harbor. 726 pp, 1988 Harlow and Lane, Using antibodies: A laboratory manual. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York. 1999
Hinnen等, Proc. Natl. Acad. Sci. USA. 75: 1929, 1978  Hinnen et al., Proc. Natl. Acad. Sci. USA. 75: 1929, 1978
Jones等, Nature. 321 :522-5, 1986  Jones et al., Nature. 321: 522-5, 1986
Kozbor等, J Immunol. 133:300ト 5, 1984  Kozbor et al., J Immunol. 133: 300 g 5, 1984
Lopata等, Nucleic Acids Research. 12:5707. 1984  Lopata et al., Nucleic Acids Research. 12: 5707. 1984
Madden等, Cancer Res, 56:5384-5390, 1996  Madden et al., Cancer Res, 56: 5384-5390, 1996
Martin及び Papahadjopoulos, J Biol Chem. 257:286-288, 1982  Martin and Papahadjopoulos, J Biol Chem. 257: 286-288, 1982
Milstein及び Cuello, Nature 305:537-40, 1983  Milstein and Cuello, Nature 305: 537-40, 1983
Morrison等, enetically engineered antibody molecules and their application. Ann N Morrison et al., Enetically engineered antibody molecules and their application.Ann N
Y Acad Sci. 507: 187-98, 1987 Y Acad Sci. 507: 187-98, 1987
Okano等, J Neurochem. 56:560-7, 1991  Okano et al., J Neurochem. 56: 560-7, 1991
Presta等, Curr Opin Biotechnol. 3:394-8, 1992  Presta et al., Curr Opin Biotechnol. 3: 394-8, 1992
Riechmann等, Nature 332:323-7, 1988  Riechmann et al., Nature 332: 323-7, 1988
Sambrook, J. Molecular cloning: a laboratory manual. Cold Spring Harbor Laborator y, Cold Spring Harbor. 1989  Sambrook, J. Molecular cloning: a laboratory manual. Cold Spring Harbor Laborator y, Cold Spring Harbor. 1989
Schroeder等, The peptide, Academic Press, New York 1965  Schroeder et al., The peptide, Academic Press, New York 1965
Shigekawa及び Dower, BioTechniques. 6:742-751, 1988 Shigekawa and Dower, BioTechniques. 6: 742-751, 1988
Tonkinson等, Cancer Investigation, 14(1): 54-65, 1996 Tonkinson et al., Cancer Investigation, 14 (1): 54-65, 1996
Verhoeyen等, Science. 239: 1534-6, 1988 Verhoeyen et al., Science. 239: 1534-6, 1988

Claims

請求の範囲 The scope of the claims
[1] 以下の(a)又は (b)に示されるポリペプチド、又はそのァゴニスト、及び薬剤的に許 容される担体を含んでなる、細胞の増殖促進又は生存維持のための組成物。  [1] A composition for promoting cell growth or maintaining survival, comprising a polypeptide shown in the following (a) or (b), or an agonist thereof, and a pharmaceutically acceptable carrier.
(a)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列からなるポリペプチド  (a) a polypeptide comprising the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12
(b)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列において、 1若しくは数個のアミノ酸の置換、欠失若しくは挿 入をもつアミノ酸配列からなり、かつ、細胞の増殖促進活性または生存維持活性を有 するポリペプチド  (b) In the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12, substitution of one or several amino acids, deletion or insertion A polypeptide comprising an amino acid sequence having a cell growth promoting activity or survival maintaining activity
[2] 以下の(a)又は (b)に示されるポリヌクレオチドを含む組換えベクター、及び薬剤的 に許容される担体を含んでなる、細胞の増殖促進又は生存維持のための組成物。 [2] A composition for promoting cell growth or maintaining survival, comprising a recombinant vector comprising the polynucleotide shown in (a) or (b) below, and a pharmaceutically acceptable carrier.
(a)配列番号 1、配列番号 3、配列番号 5、配列番号 7、配列番号 9又は配列番号 11 で表されるヌクレオチド配列からなるポリヌクレオチド (a) a polynucleotide comprising the nucleotide sequence represented by SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11
(b)配列番号 1、配列番号 3、配列番号 5、配列番号 7、配列番号 9又は配列番号 11 で表されるヌクレオチド配列からなるポリヌクレオチドの相補鎖と高ストリンジェントな 条件下でハイブリダィズするポリヌクレオチドであって、該ポリヌクレオチドがコードす るポリペプチドが細胞の増殖促進活性または生存維持活性を有するポリヌクレオチド (b) a polynucleotide that hybridizes with a complementary strand of a polynucleotide comprising the nucleotide sequence represented by SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11 under highly stringent conditions A polynucleotide, wherein the polypeptide encoded by the polynucleotide has cell growth promoting activity or survival maintaining activity
[3] 前記細胞が血管内皮細胞である請求項 1又は 2に記載の組成物。 3. The composition according to claim 1 or 2, wherein the cell is a vascular endothelial cell.
[4] 前記細胞が下垂体を形成する細胞である請求項 1又は 2に記載の組成物。 4. The composition according to claim 1 or 2, wherein the cell is a cell forming a pituitary gland.
[5] 前記細胞が神経細胞又はクロム親和細胞である請求項 1又は 2に記載の組成物。 5. The composition according to claim 1 or 2, wherein the cell is a nerve cell or a chromaffin cell.
[6] 血流障害関連疾患の治療、又は予防に供される請求項 1又は 2に記載の医薬組成 物。 [6] The pharmaceutical composition according to claim 1 or 2, which is used for treatment or prevention of diseases related to blood flow disorders.
[7] 低血圧症の治療、又は予防に供される請求項 1又は 2に記載の医薬組成物。  [7] The pharmaceutical composition according to claim 1 or 2, which is used for treatment or prevention of hypotension.
[8] 発育障害関連疾患の治療、又は予防に供される請求項 1又は 2に記載の医薬組成 物。  [8] The pharmaceutical composition according to claim 1 or 2, which is used for treatment or prevention of a developmental disorder-related disease.
[9] 神経疾患の治療、又は予防に供される請求項 1又は 2に記載の医薬組成物。  [9] The pharmaceutical composition according to claim 1 or 2, which is used for treatment or prevention of a neurological disease.
[10] 以下の(a)又は (b)に示されるポリペプチドに対するアンタゴニスト、及び薬剤的に 許容される担体を含んでなる、細胞の増殖抑制又は生存抑制のための組成物。 (a)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列からなるポリペプチド [10] A composition for inhibiting cell growth or survival, comprising an antagonist to the polypeptide shown in (a) or (b) below and a pharmaceutically acceptable carrier. (a) a polypeptide comprising the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12
(b)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列において、 1若しくは数個のアミノ酸の置換、欠失若しくは揷 入をもつアミノ酸配列からなり、かつ、細胞の増殖促進活性または生存維持活性を有 するポリペプチド  (b) In the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12, substitution of one or several amino acids, deletion or insertion A polypeptide comprising an amino acid sequence having a cell growth promoting activity or survival maintaining activity
[11] 前記アンタゴニストが抗体である請求項 10に記載の組成物。  11. The composition according to claim 10, wherein the antagonist is an antibody.
[12] 前記抗体が配列番号 13又は 14で表されるアミノ酸配列からなるポリペプチドと特 異的に結合するものである請求項 11に記載の組成物。  [12] The composition according to claim 11, wherein the antibody specifically binds to a polypeptide consisting of the amino acid sequence represented by SEQ ID NO: 13 or 14.
[13] 以下の(a)又は (b)に示されるポリヌクレオチドの機能を阻害する核酸を含む組換 えベクター、及び薬剤的に許容される担体を含んでなる、細胞の増殖抑制又は生存 抑制のための組成物。 [13] Inhibition of cell growth or survival, comprising a recombinant vector containing a nucleic acid that inhibits the function of a polynucleotide shown in (a) or (b) below, and a pharmaceutically acceptable carrier: For the composition.
(a)配列番号 1、配列番号 3、配列番号 5、配列番号 7、配列番号 9又は配列番号 11 で表されるヌクレオチド配列からなるポリヌクレオチド  (a) a polynucleotide comprising the nucleotide sequence represented by SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11
(b)配列番号 1、配列番号 3、配列番号 5、配列番号 7、配列番号 9又は配列番号 11 で表されるヌクレオチド配列からなるポリヌクレオチドの相補鎖と高ストリンジェントな 条件下でハイブリダィズするポリヌクレオチドであって、該ポリヌクレオチドがコードす るポリペプチドが細胞の増殖促進活性または生存維持活性を有するポリヌクレオチド  (b) a polynucleotide that hybridizes with a complementary strand of a polynucleotide comprising the nucleotide sequence represented by SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11 under highly stringent conditions A polynucleotide, wherein the polypeptide encoded by the polynucleotide has cell growth promoting activity or survival maintaining activity
[14] 前記機能を阻害する核酸が cgrl l遺伝子に対するアンチセンス DNA又は RNAで ある請求項 13に記載の組成物。  [14] The composition according to [13], wherein the nucleic acid that inhibits the function is antisense DNA or RNA to the cglrl gene.
[15] 前記細胞が血管内皮細胞である請求項 10又は 13に記載の組成物。 15. The composition according to claim 10 or 13, wherein the cell is a vascular endothelial cell.
[16] 前記細胞が下垂体を形成する細胞である請求項 10又は 13に記載の組成物。 16. The composition according to claim 10 or 13, wherein the cell is a cell forming a pituitary gland.
[17] 癌の治療、又は予防に供される請求項 10又は 13に記載の医薬組成物。 [17] The pharmaceutical composition according to claim 10 or 13, which is used for treatment or prevention of cancer.
[18] 発育障害関連疾患の治療、又は予防に供される請求項 10又は 13に記載の医薬 組成物。 [18] The pharmaceutical composition according to claim 10 or 13, which is used for treatment or prevention of a developmental disorder-related disease.
[19] 高血圧症の治療、又は予防に供される請求項 10又は 13に記載の医薬組成物。  [19] The pharmaceutical composition according to claim 10 or 13, which is used for treatment or prevention of hypertension.
[20] 以下の(a)又は (b)に示されるポリペプチドに対する抗体を含んでなる、血流障害 関連疾患、低血圧症、発育障害関連疾患、癌、高血圧症又は神経疾患の発症の有 無、予後、又は悪性度の診断用組成物。 [20] The presence of the onset of a blood flow disorder-related disease, hypotension, developmental disorder-related disease, cancer, hypertension or neurological disease comprising an antibody against the polypeptide shown in (a) or (b) below: A diagnostic composition for absence, prognosis, or malignancy.
(a)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列からなるポリペプチド  (a) a polypeptide comprising the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12
(b)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列において、 1若しくは数個のアミノ酸の置換、欠失若しくは揷 入をもつアミノ酸配列からなり、かつ、細胞の増殖促進活性または生存維持活性を有 するポリペプチド  (b) In the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12, substitution of one or several amino acids, deletion or insertion A polypeptide comprising an amino acid sequence having a cell growth promoting activity or survival maintaining activity
[21] 前記抗体が配列番号 13又は 14で表されるアミノ酸配列からなるポリペプチドと特 異的に結合するものである請求項 20に記載の診断用組成物。  21. The diagnostic composition according to claim 20, wherein the antibody specifically binds to a polypeptide consisting of the amino acid sequence represented by SEQ ID NO: 13 or 14.
[22] 以下の(a)又は (b)に示されるポリペプチドに対するアンタゴニスト。 [22] An antagonist to the polypeptide shown in the following (a) or (b).
(a)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列からなるポリペプチド  (a) a polypeptide comprising the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12
(b)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列において、 1若しくは数個のアミノ酸の置換、欠失若しくは挿 入をもつアミノ酸配列からなり、かつ、細胞の増殖促進活性または生存維持活性を有 するポリペプチド  (b) In the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12, substitution of one or several amino acids, deletion or insertion A polypeptide comprising an amino acid sequence having a cell growth promoting activity or survival maintaining activity
[23] 以下の(a)又は (b)に示されるポリペプチドに対する抗体。  [23] An antibody against the polypeptide shown in the following (a) or (b).
(a)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列からなるポリペプチド  (a) a polypeptide comprising the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12
(b)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列において、 1若しくは数個のアミノ酸の置換、欠失若しくは挿 入をもつアミノ酸配列からなり、かつ、細胞の増殖促進活性または生存維持活性を有 するポリペプチド  (b) In the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12, substitution of one or several amino acids, deletion or insertion A polypeptide comprising an amino acid sequence having a cell growth promoting activity or survival maintaining activity
[24] 配列番号 13又は 14で表されるアミノ酸配列からなるポリペプチドと特異的に結合 する請求項 23に記載の抗体。  [24] The antibody according to claim 23, which specifically binds to a polypeptide consisting of the amino acid sequence represented by SEQ ID NO: 13 or 14.
[25] 以下の(a)又は (b)に示されるポリペプチド、又はそのァゴニストの治療的有効量を 投与することを含んでなる、哺乳動物の血流障害関連疾患、低血圧症、発育障害関 連疾患又は神経疾患の治療又は予防方法。 (a)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列からなるポリペプチド [25] Blood flow disorder-related disease, hypotension, developmental disorder in mammals comprising administering a therapeutically effective amount of the polypeptide shown in the following (a) or (b), or an agonist thereof: A method for treating or preventing a related disease or a neurological disease. (a) a polypeptide comprising the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12
(b)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列において、 1若しくは数個のアミノ酸の置換、欠失若しくは揷 入をもつアミノ酸配列からなり、かつ、細胞の増殖促進活性または生存維持活性を有 するポリペプチド  (b) In the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12, substitution of one or several amino acids, deletion or insertion A polypeptide comprising an amino acid sequence having a cell growth promoting activity or survival maintaining activity
[26] 以下の(a)又は (b)に示されるポリペプチドに対するアンタゴニストの治療的有効量 を投与することを含んでなる、哺乳動物の癌、発育障害関連疾患又は高血圧症の治 療又は予防方法。  [26] Treatment or prevention of mammalian cancer, developmental disorder-related disease or hypertension, comprising administering a therapeutically effective amount of an antagonist to the polypeptide shown in (a) or (b) below: Method.
(a)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列からなるポリペプチド  (a) a polypeptide comprising the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12
(b)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列において、 1若しくは数個のアミノ酸の置換、欠失若しくは挿 入をもつアミノ酸配列からなり、かつ、細胞の増殖促進活性または生存維持活性を有 するポリペプチド  (b) In the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12, substitution of one or several amino acids, deletion or insertion A polypeptide comprising an amino acid sequence having a cell growth promoting activity or survival maintaining activity
[27] 前記アンタゴニストが抗体である請求項 26に記載の方法。  [27] The method of claim 26, wherein the antagonist is an antibody.
[28] 以下の(a)又は (b)に示されるポリヌクレオチドの機能を阻害する核酸を含む組換 えベクターの治療的有効量を投与することを含んでなる、哺乳動物の癌、発育障害 関連疾患又は高血圧症の治療又は予防方法。  [28] A mammalian cancer, developmental disorder comprising administering a therapeutically effective amount of a recombinant vector comprising a nucleic acid that inhibits the function of the polynucleotide shown in the following (a) or (b): A method of treating or preventing a related disease or hypertension.
(a)配列番号 1、配列番号 3、配列番号 5、配列番号 7、配列番号 9又は配列番号 11 で表されるヌクレオチド配列からなるポリヌクレオチド  (a) a polynucleotide comprising the nucleotide sequence represented by SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11
(b)配列番号 1、配列番号 3、配列番号 5、配列番号 7、配列番号 9又は配列番号 11 で表されるヌクレオチド配列からなるポリヌクレオチドの相補鎖と高ストリンジェントな 条件下でハイブリダィズするポリヌクレオチドであって、該ポリヌクレオチドがコードす るポリペプチドが細胞の増殖促進活性または生存維持活性を有するポリヌクレオチド (b) a polynucleotide that hybridizes with a complementary strand of a polynucleotide comprising the nucleotide sequence represented by SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11 under highly stringent conditions A polynucleotide, wherein the polypeptide encoded by the polynucleotide has cell growth promoting activity or survival maintaining activity
[29] 前記機能を阻害する核酸が cgrl l遺伝子に対するアンチセンス DNA又は RNAで ある請求項 28に記載の方法。 [29] The method according to claim 28, wherein the nucleic acid that inhibits the function is antisense DNA or RNA to the cglrl gene.
[30] 哺乳動物から得た組織細胞又は血液中における、以下の(a)又は (b)に示される ポリペプチドの量を定量的に検出する事を含んでなる、血流障害関連疾患、低血圧 症、発育障害関連疾患、癌、高血圧症、心臓疾患又は神経疾患の診断方法。 [30] As shown in the following (a) or (b) in tissue cells or blood obtained from mammals A method for diagnosing a blood flow disorder-related disease, hypotension, developmental disorder-related disease, cancer, hypertension, heart disease or neurological disease, comprising quantitatively detecting the amount of the polypeptide.
(a)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列からなるポリペプチド  (a) a polypeptide comprising the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12
(b)配列番号 2、配列番号 4、配列番号 6、配列番号 8、配列番号 10又は配列番号 1 2で表されるアミノ酸配列において、 1若しくは数個のアミノ酸の置換、欠失若しくは揷 入をもつアミノ酸配列からなり、かつ、細胞の増殖促進活性または生存維持活性を有 するポリペプチド  (b) In the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12, substitution of one or several amino acids, deletion or insertion A polypeptide comprising an amino acid sequence having a cell growth promoting activity or survival maintaining activity
[31] 以下の(a)又は (b)に示されるポリヌクレオチドを含む組換えベクターを細胞に導入 し、不死化した細胞を製造する方法。  [31] A method for producing an immortalized cell by introducing a recombinant vector containing a polynucleotide shown in the following (a) or (b) into a cell.
(a)配列番号 1、配列番号 3、配列番号 5、配列番号 7、配列番号 9又は配列番号 11 で表されるヌクレオチド配列からなるポリヌクレオチド  (a) a polynucleotide comprising the nucleotide sequence represented by SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11
(b)配列番号 1、配列番号 3、配列番号 5、配列番号 7、配列番号 9又は配列番号 11 で表されるヌクレオチド配列からなるポリヌクレオチドの相補鎖と高ストリンジェントな 条件下でハイブリダィズするポリヌクレオチドであって、該ポリヌクレオチドがコードす るポリペプチドが細胞の増殖促進活性または生存維持活性を有するポリヌクレオチド (b) a polynucleotide that hybridizes with a complementary strand of a polynucleotide comprising the nucleotide sequence represented by SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11 under highly stringent conditions A polynucleotide, wherein the polypeptide encoded by the polynucleotide has cell growth promoting activity or survival maintaining activity
[32] 請求項 31に記載の方法により得られた細胞。 [32] A cell obtained by the method according to claim 31.
[33] 以下の(a)又は (b)に示されるポリペプチド。 [33] A polypeptide represented by the following (a) or (b):
(a)配列番号 8、配列番号 10又は配列番号 12で表されるアミノ酸配列からなるポリべ プチド  (a) a polypeptide comprising the amino acid sequence represented by SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12
(b)配列番号 8、配列番号 10又は配列番号 12で表されるアミノ酸配列において、 1 若しくは数個のアミノ酸の置換、欠失若しくは揷入をもつアミノ酸配列からなり、かつ、 細胞の増殖促進活性または生存維持活性を有するポリペプチド  (b) in the amino acid sequence represented by SEQ ID NO: 8, SEQ ID NO: 10 or SEQ ID NO: 12, consisting of an amino acid sequence having one or several amino acid substitutions, deletions or insertions, and cell growth promoting activity Or polypeptide having life-sustaining activity
[34] 以下の(a)又は(b)に示されるポリヌクレオチド。  [34] A polynucleotide shown in the following (a) or (b):
(a)配列番号 7、配列番号 9又は配列番号 11で表されるヌクレオチド配列からなるポ リヌタレ才チド  (a) Polytale talent comprising the nucleotide sequence represented by SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11
(b)配列番号 7、配列番号 9又は配列番号 1 1で表されるヌクレオチド配列からなるポ リヌクレオチドの相補鎖と高ストリンジヱントな条件下でハイブリダィズするポリヌクレオ チドであって、該ポリヌクレオチドがコードするポリペプチドが細胞の増殖促進活性ま たは生存維持活性を有するポリヌクレオチド (b) a polynucleotide that hybridizes with a complementary strand of a polynucleotide comprising the nucleotide sequence represented by SEQ ID NO: 7, SEQ ID NO: 9 or SEQ ID NO: 11 under highly stringent conditions. A polynucleotide wherein the polypeptide encoded by the polynucleotide has a cell growth-promoting activity or survival-sustaining activity.
PCT/JP2006/304969 2005-03-15 2006-03-14 Novel secretory protein derived from pituitary cell and use of the same WO2006098304A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2007508144A JPWO2006098304A1 (en) 2005-03-15 2006-03-14 Novel secreted proteins derived from pituitary cells and their uses

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2005073506 2005-03-15
JP2005-073506 2005-03-15

Publications (1)

Publication Number Publication Date
WO2006098304A1 true WO2006098304A1 (en) 2006-09-21

Family

ID=36991647

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2006/304969 WO2006098304A1 (en) 2005-03-15 2006-03-14 Novel secretory protein derived from pituitary cell and use of the same

Country Status (2)

Country Link
JP (1) JPWO2006098304A1 (en)
WO (1) WO2006098304A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108220443A (en) * 2018-01-31 2018-06-29 北京泱深生物信息技术有限公司 Applications of the CGREF1 as marker in clear cell carcinoma of kidney diagnosis and treatment

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2000512494A (en) * 1996-05-29 2000-09-26 ジェンザイム・コーポレーション Cell growth regulatory gene

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2000512494A (en) * 1996-05-29 2000-09-26 ジェンザイム・コーポレーション Cell growth regulatory gene

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MADDEN S.L. ET AL.: "Induction of cell growth regulating genes by P53", CANCER RES., vol. 56, no. 23, 1996, pages 5384 - 5390, XP002046025 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108220443A (en) * 2018-01-31 2018-06-29 北京泱深生物信息技术有限公司 Applications of the CGREF1 as marker in clear cell carcinoma of kidney diagnosis and treatment

Also Published As

Publication number Publication date
JPWO2006098304A1 (en) 2008-08-21

Similar Documents

Publication Publication Date Title
JP3695642B2 (en) Promotion or inhibition of angiogenesis and cardiovascularization
JP4145314B2 (en) Compositions and methods for tumor cell growth inhibition
KR20010084882A (en) Methods and Compositions for Inhibiting Neoplastic Cell Growth
JP2003116587A (en) Polypeptide and nucleic acid encoding the same
KR100543857B1 (en) Promotion or Inhibition of Angiogenesis and Cardiovascularization
EP1225224A1 (en) Shear stress-response dna
JP2002526099A (en) NLK1-interacting protein
US9109043B2 (en) Screening method for antidiabetic agent using newly identified insulin secretion regulation factor
JP4841054B2 (en) Novel insulin / IGF / relaxin family polypeptide and DNA thereof
EA011261B1 (en) Cell surface glycoprotein
JP2006526762A (en) Methods and compositions for modulating the interaction between adiponectin and its receptor
JP2003530082A (en) Compositions and methods for treating immune related diseases
WO2006098304A1 (en) Novel secretory protein derived from pituitary cell and use of the same
JP2002517998A (en) Interaction of p27 (KIP1) with FKBP-12
WO2001046415A1 (en) Novel tachykinin-like polypeptides and use thereof
EP1902729A1 (en) Int6 PROTEIN INVOLVED IN HYPOXIA STRESS INDUCTION AND USE THEREOF
WO2006085648A1 (en) Novel narcolepsy-related gene
WO2005033266A2 (en) Regulation of acheron expression
KR20010085915A (en) Methods and Compositions for Inhibiting Neoplastic Cell Growth
KR100678523B1 (en) A method for identifying an agonist or an inhibitor of a pro840 polypeptide
JP2003511076A (en) Aortic carboxypeptidase-like protein and nucleic acid encoding the same
JP2008063244A (en) Treatment or prevention of saccharometabolism abnormality
WO2007128545A9 (en) Mac30 gene
WO2005071098A1 (en) Method of screening substance improving lipid metabolism
Emr et al. National Academy

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2007508144

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

NENP Non-entry into the national phase

Ref country code: RU

122 Ep: pct application non-entry in european phase

Ref document number: 06729022

Country of ref document: EP

Kind code of ref document: A1