WO2003072780A1 - Novel proteins, dnas thereof and use of the same - Google Patents

Novel proteins, dnas thereof and use of the same Download PDF

Info

Publication number
WO2003072780A1
WO2003072780A1 PCT/JP2003/002133 JP0302133W WO03072780A1 WO 2003072780 A1 WO2003072780 A1 WO 2003072780A1 JP 0302133 W JP0302133 W JP 0302133W WO 03072780 A1 WO03072780 A1 WO 03072780A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
present
peptide
salt
seq
Prior art date
Application number
PCT/JP2003/002133
Other languages
French (fr)
Japanese (ja)
Inventor
Tsukasa Sugo
Masaaki Mori
Original Assignee
Takeda Chemical Industries, Ltd.
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 Takeda Chemical Industries, Ltd. filed Critical Takeda Chemical Industries, Ltd.
Priority to AU2003211722A priority Critical patent/AU2003211722A1/en
Publication of WO2003072780A1 publication Critical patent/WO2003072780A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/72Receptors; Cell surface antigens; Cell surface determinants for hormones
    • C07K14/723G protein coupled receptor, e.g. TSHR-thyrotropin-receptor, LH/hCG receptor, FSH receptor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/04Drugs for genital or sexual disorders; Contraceptives for inducing labour or abortion; Uterotonics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • 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
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to a novel G protein-coupled receptor protein, a DNA encoding the same, and uses thereof.
  • anorexia anemia associated with anorexia
  • prevention and treatment of hypoproteinemia weak labor, laxative bleeding, uterine remodeling failure or milk stasis, or obesity
  • hyperphagia affective disorder, sex Dysfunction, labour, tonic contractions, preterm birth, Prader-Willi syndrome, diabetes and its complications, hypertension, hyperlipidemia, coronary atherosclerosis, gout, respiratory disease, fatty liver, infertility or
  • the present invention relates to screening for compounds having a prophylactic and therapeutic effect on osteoarthritis. Background art
  • Obesity is known to be a risk factor for the development of many lifestyle-related diseases such as diabetes and heart disease. With changes in eating habits and lifestyles, fertility is increasing, especially in developed countries, and it has become a very socially significant problem from the health economic perspective. Obesity is known to be caused by various factors.Recently, several peptides located in the hypothalamus, the feeding center, are involved in promoting or suppressing feeding behavior in mammals. Thus, the contribution of these peptides to obesity has received particular attention. This is because drugs that inhibit or activate the action of these peptides are considered to be potential central antiobesity drugs.
  • One such hypothalamic peptide, melanin-concentrating hormone (Melanin) melanin-concentrating hormone
  • MCH Concentrating Hormone
  • MCH is localized in the feeding center, the hypothalamic lateral area and the irregular zone. And were more likely to be involved in eating and drinking behavior, but Qu et al. First showed that MCH was involved in eating behavior as follows (Qu, D. et al., Nature, vol. 380). , 243-247, 1996). Expression of altered mRNA levels in the hypothalamus of ob / ob mice and wild-type mice, which are genetically obese models, was searched for by the di ferent al al di sp lay method. Was found to be significantly increased. Increased MCH gene expression was observed in both wild-type and ob / ob mice after a 24-hour fast.
  • MCH functioned as a food-stimulating peptide, but in fact, administration of MCH directly to the lateral ventricle of rats significantly increased food intake. In addition, it was reported that MCH knockout mice lost significant weight due to reduced food intake and increased metabolism.
  • mice deficient in the MCH gene have not been reported in animals knocked down by these peptides, and MCH functions as a particularly important regulator in eating behavior. it seems to do.
  • MCH gene-deficient mice did not show any prominent phenotype other than weight loss, it was suggested that suppressing the effect of MCH could lead to the development of anti-obesity drugs with few side effects.
  • SLC-1 (MCHK MCHR1) was previously known as a functional receptor for MCH (Nature, 400, 26-1265, 1999, Nature, 400, 265-269, 1999, Bi ochei. Bi ophys. Res. Commun., 261, 622-626, 1999, Nature Cell Biol., 1st, 267-271, 1999, FEBS Lett. 524, 1999, WO 00/40725, etc.), and recently, SLT (MCH2, CHR2) has been cloned as the second receptor subtype of MCH (J. Biol. Chem. , Volume 276, 20125-20129 Co., 2001, Biochem. Biophys. Res.Co, Vol. 283, 1013- 1018, 2001, Proc. Natl.
  • SLC-1 receptor is particularly strongly expressed in the hypothalamus region involved in feeding (arcus nucleus, ventral nucleus, dorsolateral nucleus, paraventricular nucleus) and in the nucleus accumbens which is thought to be involved in the reward system
  • SLT is expressed in the hippocampus and tonsils and is low in the hypothalamus, so SLC-1 is mainly involved in the inducing action of MCH, and SLT is It has been suggested that they may be involved in memory and emotion's effects.
  • SLT is distributed in the hypothalamus in areas different from SLC-1 (pronucleus, lateral nucleus, etc.), and is involved in feeding because it is located near the causative gene of childhood obesity.
  • the present inventors have conducted intensive studies in order to solve the above-mentioned problems, and as a result, have found that genes encoding monkey, canine, and feline SLTs showing homology to human SLT can be obtained from monkey, canine, and feline brain cDNA. More successfully found and cloned. In addition, these receptors bind to MCH. As a result of further study based on these findings, the present invention has been completed.
  • the present invention (1) a protein or a salt thereof containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 4, SEQ ID NO: 14 or SEQ ID NO: 24;
  • a diagnostic agent comprising the polynucleotide according to (6),
  • a medical comprising the antisense polynucleotide according to (28).
  • a method for quantifying the mRNA of the protein according to (1) which comprises using the polynucleotide or a part thereof according to (6);
  • a method for screening a compound or a salt thereof that alters the expression level of the protein described in (1) above which comprises using the quantification method described in (30) above.
  • a medicament comprising the compound according to (35) or (36) or a salt thereof,
  • An obesity, hyperphagia, affective disorder, sexual dysfunction, or hyperactivity comprising administering to a mammal an effective amount of the compound according to (25) or a salt thereof.
  • Obesity, hyperphagia, affective disorders, sexual dysfunction, overwork labor, ankylosing uterine contractions, preterm birth, Prader-Willi syndrome, diabetes and its complications, hypertension, hyperlipidemia, coronary Provide the use of the compound described in (25) or a salt thereof for the manufacture of a prophylactic or therapeutic agent for arteriosclerosis, gout, respiratory disease, fatty liver, infertility or osteoarthritis. I do. BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 is a graph showing the intracellular Ca ion elevating activity of various concentrations of MCH on cat SLT-expressing CH0 cells measured using FLIPR.
  • FIG. 2 is a graph of the expression level of the cat SLT gene in various parts of the cat brain.
  • the expression level was expressed as 1000 times the value obtained by dividing the expression level of the cat SLT gene for the same type II RNA level by the expression level of the cat GAPDH gene.
  • the protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 4, SEQ ID NO: 14 or SEQ ID NO: 24 (hereinafter, may be referred to as the protein of the present invention) is It is a G protein-coupled receptor protein and is used in human warm-blooded animals (eg, guinea pig, rat, mouse, chicken, Cells of egrets, bushes, sheep, sheep, monkeys, monkeys, dogs, cats, etc.
  • human warm-blooded animals eg, guinea pig, rat, mouse, chicken, Cells of egrets, bushes, sheep, sheep, monkeys, monkeys, dogs, cats, etc.
  • amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 4 includes amino acids having 98% or more, preferably about 99% or more homology with the amino acid sequence represented by SEQ ID NO: 4. And the like.
  • Examples of the protein containing an amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 4 include, for example, a protein containing an amino acid sequence substantially identical to the amino acid sequence represented by the aforementioned SEQ ID NO: 4 However, a protein having substantially the same activity as the protein having the amino acid sequence represented by SEQ ID NO: 4 is preferred.
  • amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 14 includes 92% or more, preferably about 95% or more, more preferably about 95% or more of the amino acid sequence represented by SEQ ID NO: 14. Amino acid sequences having a homology of 97% or more, more preferably about 99% or more, may be mentioned.
  • proteins containing an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 14 include, for example, amino acids substantially the same as the amino acid sequence represented by the aforementioned SEQ ID NO: 14
  • a protein containing a sequence and having substantially the same activity as a protein having the amino acid sequence represented by SEQ ID NO: 14 is preferred.
  • the amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 24 includes 92% or more, preferably about 95% or more, more preferably about 95% or more of the amino acid sequence represented by SEQ ID NO: 24. Amino acid sequences having a homology of 97% or more, more preferably about 99% or more, and the like can be mentioned.
  • proteins having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 24 include, for example, amino acids substantially the same as the amino acid sequence represented by SEQ ID NO: 24 above.
  • a protein containing a sequence and having substantially the same activity as a protein having the amino acid sequence represented by SEQ ID NO: 24 is preferred.
  • Substantially the same activity includes, for example, a binding activity to MCH, a signal information transduction effect, and the like. Substantially the same means that their activities are the same in nature. Therefore, it is preferable that the activities such as the binding activity to MCH and the activity of signal transduction are equivalent (eg, about 0.5 to 2 times), but the quantitative factors such as the degree of these activities and the molecular weight of the protein are It may be different.
  • the activity such as the binding activity and the signal transduction action can be measured according to a method known per se or a method analogous thereto.
  • Examples of the protein of the present invention include (1) (i) one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 4 (eg, about 1 to 8, preferably about 1 to 5, (Ii) preferably one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 4 (for example, about 1 to 200, preferably 1 to 1); About 50, preferably about 1 to 100, preferably about;!
  • amino acids in the amino acid sequence represented by SEQ ID NO: 14 for example, about 1 to 200, preferably about 1 to 150, and preferably 1 to 1 About 100, preferably about 1 to 50, preferably about 1 to 30, preferably about 1 to 10, and more preferably the number (1 to 5) of amino acids is other amino acids.
  • a so-called mutin such as a protein containing a substituted amino acid sequence or (V) a protein containing an amino acid sequence combining them; 1 or 2 or more (for example, about 1 to 25, preferably about 1 to 10, and more preferably about 1 to 5) amino acids in the amino acid sequence represented by SEQ ID NO: 24; (Ii) one or two or more amino acid sequences represented by SEQ ID NO: 24 (for example, about 1 to 200, preferably about 1 to 150, and preferably About 100, preferably about 1 to 50, preferably about 1 to 30, preferably about 1 to 10, and more preferably about (1 to 5) amino acids (Iii) one or two or more amino acid sequences represented by SEQ ID NO: 24 (for example, about 1 to 200, preferably about 1 to 150, and preferably 1 to 1 About 100 pieces, preferably about 1 to 50 pieces, preferably about 1 to 30 pieces, preferably about 1 to 10 pieces, and more preferably Or (iv) one or two or more amino acids in the amino acid sequence represented by
  • a so-called mutin such as a protein containing an amino acid sequence in which a number (1 to 5) of amino acids are substituted with another amino acid, or (V) an amino acid sequence obtained by combining them is also included.
  • the position of the insertion, deletion or substitution is not particularly limited.
  • protein of the present invention examples include, for example, a protein containing the amino acid sequence represented by SEQ ID NO: 4, a protein containing the amino acid sequence represented by SEQ ID NO: 14, SEQ ID NO: 24 And proteins containing the amino acid sequence represented by
  • the partial peptide of the protein of the present invention (hereinafter sometimes abbreviated as the partial peptide of the present invention) is a partial peptide of the protein of the present invention described above, and is preferably the same as the above-described protein of the present invention. Any material having properties may be used.
  • the partial peptide of the present invention lacks one or more (preferably about 1 to 10, more preferably, about 1 to 5) amino acids in its amino acid sequence, Alternatively, one or more (preferably, about 1 to 20, more preferably, about 1 to 10, and more preferably, about 1 to 5) amino acids are added to the amino acid sequence. Or 1 or 2 or more amino acids in the amino acid sequence thereof (preferably, about 1 to 20; more preferably, about 1 to 10;
  • amino acids or one or more (preferably about 1 to 10, more preferably several, and more preferably 1 to (About 5 amino acids) may be substituted with another amino acid.
  • the partial peptide of the present invention for example, in the amino acid sequence represented by SEQ ID NO: 4, for example, the 1st to 41st amino acids, the 31st to 34th amino acids, SEQ ID NO: 1
  • the amino acid sequence represented by the amino acid sequence represented by SEQ ID NO: 24 the 1st to 41st amino acid sequence, the 31st amino acid sequence to the 34th amino acid sequence, SEQ ID NO: 24
  • the amino acid sequences of the 1st to 41st and the 31st to 330th amino acids are preferred.
  • a peptide containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 25 is a human Cells of warm-blooded animals (eg, guinea pigs, rats, mice, chicks, egrets, pigs, higgies, lions, monkeys, dogs, cats, etc.) (eg, hepatocytes, spleen cells, nerve cells, glial cells) 3 cells, bone marrow cells, mesangial cells, Langer's cells, epidermal cells, epithelial cells, goblet cells, endothelial cells, smooth muscle cells, fibroblasts, fiber cells, muscle cells, adipocytes, immune cells (Eg, macrophages, T cells, B cells, natural killer cells, mast cells, neutrophils, basophils, eosinophils, monocytes), megakaryocyte
  • Examples of the peptide having an amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 25 include, for example, an amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 25 Peptides having substantially the same activity as the peptide having the amino acid sequence represented by SEQ ID NO: 25 are preferred.
  • Substantially equivalent activities include, for example, SLT binding activity, etc. It is. Substantially the same indicates that the activity is the same in nature. Therefore, it is preferable that the binding activity to SLT is equivalent (eg, about 0.5 to 2 times), but quantitative factors such as the degree of this activity and the molecular weight of the protein may be different. No.
  • the binding activity can be measured according to a method known per se.
  • a peptide containing the amino acid sequence represented by SEQ ID NO: 25 is preferably used.
  • Examples of the peptide of the present invention include: (i) one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 25 (for example, about 1 to 30, preferably about 1 to 10; More preferably, an amino acid sequence in which a number (1 to 5) of amino acids are deleted, and (ii) one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 25 (for example, about 1 to 30 amino acids, Preferably about 1 to 10 amino acids, more preferably about 1 to 5 amino acids, and (iii) one or more amino acid sequences in the amino acid sequence represented by SEQ ID NO: 25. (For example, about 1 to 30 amino acids, preferably about 1 to 10 amino acids, and more preferably about 1 to 5 amino acids).
  • SEQ ID NO: 25 1 or 2 or more in the amino acid sequence for example, about 1 to 30, preferably about 1 to 10, more preferably
  • a peptide containing an amino acid sequence in which a number (1 to 5) of amino acids are substituted with another amino acid, or (V) an amino acid sequence obtained by combining them is also included.
  • the position of the insertion, deletion or substitution is not particularly limited.
  • Specific examples include a peptide containing the 5th to 19th partial sequences from the N-terminal of the amino acid sequence represented by SEQ ID NO: 25. More specifically, it has the amino acid sequence represented by SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30 or SEQ ID NO: 31. Peptides are examples. Preferably, a peptide having the amino acid sequence represented by SEQ ID NO: 28 is used.
  • Substantially identical substitutions of amino acids in the amino acid sequence may be selected, for example, from other amino acids of the class to which the amino acid belongs.
  • Non-polar (hydrophobic) amino acids include alanine, leucine, isoleucine, valin, proline, phenylalanine, tributofan, methionine, and the like.
  • Polar (neutral) amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine.
  • Positively charged (basic) amino acids include arginine, lysine and histidine.
  • Negatively charged (acidic) amino acids include aspartic acid and glutamic acid. '
  • the position at which the above-mentioned other amino acid is deleted or substituted is preferably a position other than Cys in the amino acids constituting the peptide of the present invention.
  • the peptide of the present invention may be labeled.
  • Specific examples include peptides labeled with an isotope by a method known per se, fluorescently labeled peptides (for example, fluorescently labeled with fluorescein), biotinylated peptides, enzyme-labeled peptides, and the like. .
  • Porton A labeled product of the peptide of the present invention prepared by a known method using one Hunter reagent may be used.
  • the left end of the protein and the peptide is the N-terminus (amino end) and the right end is the C-terminus (potassium terminal) according to the convention of peptide labeling.
  • This onset Ming proteins and peptides, C-terminal, carboxyl group (-C00H), carbo Kishireto (-C00-), amide (- C0M 2) or ester (- C00R) any derconnection may be of.
  • R in the ester e.g., methyl, Echiru, n- propyl, isopropyl, n - C, such as butyl, _ 6 alkyl groups, such as cyclopentyl, such as hexyl cyclo ⁇ 3 - 8 cycloalkyl group, for example, phenyl, alpha-naphthyl of which C 6 _ 12 7 aryl group, e.g., benzyl, phenyl, such as phenethyl - C, C 7 _ such as alpha-Nafuchiru alkyl groups such as _ 2 Al kill group or carry one naphthylmethyl
  • a 14 aralkyl group, a bivaloyloxymethyl group and the like are used.
  • the protein and peptide of the present invention have a lipoxyl group (or lipoxylate) other than at the C-terminus
  • those in which the carboxyl group is amidated or esterified are also included in the proteins and peptides of the present invention.
  • the ester in this case, for example, the above-mentioned C-terminal ester and the like are used.
  • amino acid residues at the N-terminus eg, Mechionin residues
  • Amino group protecting groups e.g., formyl group, etc. C M Ashiru group such Arukanoiru such Asechiru group
  • Substituent on the side chain of amino acid in the molecule for example, -OH, -SH, amino group, imidazo Ichiru group, indole group, etc. Guanijino group
  • a suitable protecting group e.g., formyl group, etc.
  • c, _ 6 Ashiru groups such Arukanoiru groups such as ⁇ Se butyl group), or Also included are complex proteins such as so-called glycoproteins to which sugar chains are bound.
  • the force Rupokishiru group (-C00H)
  • the force Lupo - Kishireto (-C00-)
  • amide (- C0Nh 2) or may be filed in any of ester (-C00R).
  • the partial peptide of the present invention includes, as in the case of the protein of the present invention, those having a carbonyl group (or carboxylate) other than the C-terminus, N-terminal amino acid residues (eg, methionine). Residue is protected with a protecting group, the glutamine residue formed by cleavage of the N-terminal in vivo is a pyroglutamate, and the substituent on the side chain of the amino acid in the molecule is appropriate. Also included are those protected with a protecting group or complex peptides such as so-called glycopeptides to which sugar chains are bonded.
  • Salts of the protein or partial peptide of the present invention and salts of the peptide of the present invention include salts with physiologically acceptable acids (eg, inorganic acids, organic acids) and bases (eg, alkali metal salts). Preference is given to the use of, especially the physiologically acceptable acid addition salts.
  • physiologically acceptable acids eg, inorganic acids, organic acids
  • bases eg, alkali metal salts
  • Such salts include, for example, salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid), and organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid) Salts with formic acid, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid) are used.
  • inorganic acids eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid
  • organic acids eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid
  • Salts with formic acid, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid are used.
  • the protein of the present invention or a partial peptide thereof, the peptide of the present invention or a salt thereof can be produced from the above-mentioned human or warm-blooded animal cell or tissue by a known protein purification method. Can also be produced by culturing a transformant containing DNA encoding It can also be produced according to the peptide synthesis method described below.
  • the human or mammalian tissues or cells are homogenized and then extracted with an acid or the like, and the resulting extract is subjected to reverse phase chromatography, ion exchange chromatography, etc. Purification and isolation can be achieved by combining the above chromatography.
  • the protein of the present invention for the synthesis of the peptide of the present invention, its partial peptide, their amides or their salts, commercially available resins for protein / peptide synthesis can be used.
  • a resin for example, Methyl resin, hydroxymethyl resin, benzhydrylamine resin, aminomethyl resin, 4-benzyloxybenzyl alcohol resin, 4-methylbenzhydrylamine resin, PAM resin, 4-hydroxymethylmethylphenylacetamide Methyl resin, polyacrylamide resin, 4- (2 ', 4'-dimethoxyphenylhydroxymethyl) phenoxy resin, 41- (2', 4'-dimethoxyphenyl Fmocaminoethyl) phenoxy resin, etc. Can be.
  • an amino acid appropriately protected with an ⁇ -amino group and a side chain functional group is condensed on the resin in accordance with the sequence of the desired protein / peptide according to various known condensation methods. Let it. At the end of the reaction, proteins and peptides are cleaved from the resin, and at the same time, various protecting groups are removed. Further, an intramolecular disulfide bond formation reaction is performed in a highly diluted solution to obtain the desired protein, peptide or amide thereof.
  • the protected amino acid may be added directly to the resin along with a racemization inhibitor additive (eg, HOBt, HOOBt) or pre-protected as a symmetrical acid anhydride or HOBt ester or HOOBt ester. It can be added to the resin after activation of the amino acid.
  • a racemization inhibitor additive eg, HOBt, HOOBt
  • pre-protected as a symmetrical acid anhydride or HOBt ester or HOOBt ester it can be added to the resin after activation of the amino acid.
  • the solvent used for activating the protected amino acid or for condensing with the resin may be appropriately selected from solvents known to be usable for the protein-peptide condensation reaction.
  • acid amides such as N, N-dimethylformamide, N, N-dimethylacetamide, N-methylpyrrolidone, halogenated hydrocarbons such as methylene chloride and chloroform, alcohols such as trifluoroethanol , Sulfoxides such as dimethyl sulfoxide, ethers such as pyridine, dioxane, and tetrahydrofuran; nitriles such as acetonitrile and propionitrile; esters such as methyl acetate and ethyl acetate; or an appropriate mixture thereof.
  • the reaction temperature is appropriately selected from a range known to be usable for a protein bond formation reaction, and is usually appropriately selected from a range of about 120 ° (: to 5 Ot :).
  • the sensitized amino acid derivative is usually used in a 1.5 to 4-fold excess.
  • Examples of the protecting group for the amino group of the starting material include Z, Boc, Yuichi Sharipentyloxycarbonyl, Isoporonyloxycarbonyl, 4-methoxybenzyloxycarbonyl, C11Z, Br-Z , Adamantyloxycarbonyl, trifluoroacetyl, phthaloyl, formyl, 2-ditrophenylsulfenyl, diphenylphosphinothioyl, Fmoc and the like are used.
  • Lepoxyl groups can be, for example, alkyl esterified (e.g., methyl, ethyl, propyl, butyl, tert-butyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, 2-adamantyl, etc.).
  • alkyl esterified e.g., methyl, ethyl, propyl, butyl, tert-butyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, 2-adamantyl, etc.
  • aralkyl esterification e.g., benzyl ester, 412 trobenzyl ester, 4-methoxybenzyl ester, 4-methyl benzyl ester, benzhydryl esterification
  • phenacyl esterification Benzyloxycarbonyl hydrazide, tert-butoxycarbonyl hydrazide, trityl hydrazide and the like.
  • the hydroxyl group of serine can be protected, for example, by esterification or etherification.
  • a group suitable for this esterification for example, a lower alkanol group such as an acetyl group, an aroyl group such as a benzoyl group, a group derived from carbonic acid such as a benzyloxycarbonyl group, an ethoxycarponyl group, and the like are used.
  • Examples of a group suitable for etherification include a benzyl group, a tetrahydroviranyl group, and a t-butyl group.
  • the protecting group of the phenolic hydroxyl group of tyrosine for example, B zl, C 1 2 -
  • Bzl 2-nitrobenzyl, Br-Z, Yuichi Sharybutyl and the like are used.
  • protecting group for imidazole of histidine for example, Tos, 4-methoxy-12,3,6-trimethylbenzenesulfonyl, DNP, benzyloxymethyl, Bum, Boc, Trt, Fmoc and the like are used.
  • activated carboxylic acid groups of the raw material include, for example, corresponding acid anhydrides, azides, and activated esters [alcohols (for example, pentachlorophenol, 24,5-trichloromouth phenol, 2,4-dinitrophenol) , Cyanomethyl alcohol, paranitrophenol, HONB, N-hydroxysuccinimide, N-hydroxyphthalimide, and esters with HOBt).
  • activated amino group of the raw material for example, a corresponding phosphoric acid amide is used.
  • Methods for removing (eliminating) the protecting group include, for example, catalytic reduction in a hydrogen stream in the presence of a catalyst such as Pd-black or Pd-carbon, or hydrogen fluoride anhydride or methanesulfonic acid.
  • a catalyst such as Pd-black or Pd-carbon, or hydrogen fluoride anhydride or methanesulfonic acid.
  • the elimination reaction by the above acid treatment is generally carried out at a temperature of about 120 ° C.
  • the protection of the functional group which should not be involved in the reaction of the raw materials, the protective group, the elimination of the protective group, the activation of the functional group involved in the reaction, and the like can be appropriately selected from known groups or known means.
  • an amide of a protein or peptide for example, first, after amidating and protecting the ⁇ -hydroxyl group of the amino acid at the terminal end of the amino acid, a peptide chain is extended to a desired chain length on the amino side. After that, the protein / peptide obtained by removing only the protecting group for the ⁇ -amino group at the ⁇ -terminal of the peptide chain and the C-terminal lipoxyl group are removed. A protein and a peptide from which only the protecting group has been removed are produced, and the peptide and the protein are condensed in a mixed solvent as described above. Details of the condensation reaction are the same as described above.
  • the crude peptide / crude protein is purified by various known purification means, and the main fraction is freeze-dried to obtain an amide of the desired protein / peptide.
  • an ester of protein / peptide for example, after condensing the ⁇ -hydroxyloxyl group of a carboxy-terminal amino acid with a desired alcohol to form an amino acid ester, the same procedure as in the amide of protein / peptide is carried out. It is possible to obtain an ester of a protein and a peptide.
  • the peptide of the present invention and the protein of the present invention can be produced according to a peptide synthesis method known per se. Further, the partial peptide of the protein of the present invention or a salt thereof can be produced according to a peptide synthesis method known per se, or by cleaving the protein of the present invention with an appropriate peptidase.
  • any of a solid phase synthesis method and a liquid phase synthesis method may be used. That is, the peptide of the present invention is condensed with the peptide of the present invention or the partial peptide or amino acid capable of constituting the protein of the present invention and the remaining portion, and when the product has a protecting group, the protecting group is eliminated to remove the target peptide. Can be manufactured. Examples of the known condensation method and elimination of the protecting group include the methods described in the following (i) to (V).
  • the peptide of the present invention or the partial peptide of the present invention can be purified and isolated by a combination of ordinary purification methods, for example, solvent extraction, distillation, laffification, liquid chromatography, and recrystallization. .
  • solvent extraction for example, solvent extraction, distillation, laffification, liquid chromatography, and recrystallization.
  • the peptide or partial peptide obtained by the above method is in a free form, it can be converted to an appropriate salt by a known method.
  • the peptide or partial peptide is obtained in a salt form, it can be converted to a free form by a known method Can be converted.
  • the polynucleotide encoding the protein of the present invention may be any polynucleotide containing the above-described nucleotide sequence encoding the protein of the present invention.
  • it is DNA.
  • the DNA may be any of genomic DNA, genomic DNA library, cDNA derived from the cells and tissues described above, cDNA library derived from the cells and tissues described above, and synthetic DNA.
  • the vector used for the library may be any of bacteriophage, plasmid, cosmid, phagemid and the like.
  • a reverse RNA was directly prepared using the total RNA or mRNA fraction prepared from the cells and tissues described above.
  • RT-PCR method Transcriptase Polymerase Chain Reaction
  • the DI ⁇ A encoding the protein of the present invention includes, for example, (i) a DNA containing the nucleotide sequence represented by SEQ ID NO: 3 or a nucleotide sequence represented by SEQ ID NO: 3 under highly stringent conditions.
  • Examples of the DNA that can hybridize with the nucleotide sequence represented by SEQ ID NO: 3 under high stringency conditions include, for example, 98% or more, preferably about 99% or more of the nucleotide sequence represented by SEQ ID NO: 3 DNA containing a nucleotide sequence having homology of
  • Examples of the DNA that can hybridize with the nucleotide sequence represented by SEQ ID NO: 13 under high stringency conditions include, for example, a nucleotide sequence represented by SEQ ID NO: 13 and 92% or more, preferably about 9% or less.
  • a DNA containing a nucleotide sequence having a homology of 5% or more, more preferably about 97% or more, more preferably about 99% or more is used.
  • Examples of the DNA that can hybridize with the nucleotide sequence represented by SEQ ID NO: 23 under high stringent conditions include, for example, the nucleotide sequence represented by SEQ ID NO: 23 and 92% or more, preferably about 9% or less.
  • DNA containing a base sequence having a homology of 5% or more, more preferably about 97% or more, more preferably about 99% or more is used.
  • Hybridization is performed by a known method or a method equivalent thereto, such as the method described in Molecular Cloning (Molecular Cloning) 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). It can be done according to. If a commercially available library is still used, it can be performed according to the method described in the attached instruction manual. More preferably, it can be performed under high stringency conditions.
  • High stringency conditions include, for example, a sodium concentration of about 19 to 40 mM, preferably about 19 to 20 mM, and a temperature of about 50 to 70, preferably about 60 to 65.
  • the conditions at ° C are shown. In particular, the case where the sodium concentration is about 19 mM and the temperature is about 65 is most preferable.
  • DNAs encoding the protein containing the amino acid sequence represented by SEQ ID NO: 4 include DNA containing the base sequence represented by SEQ ID NO: 3, and SEQ ID NO: 38.
  • the DNA encoding the protein having the amino acid sequence represented by SEQ ID NO: 14 has the nucleotide sequence represented by SEQ ID NO: 13 DNA, represented by SEQ ID NO: 39
  • Examples of the DNA encoding a protein having the amino acid sequence represented by SEQ ID NO: 24, such as a DNA having a nucleotide sequence represented by SEQ ID NO: 24, include a DNA having a base sequence represented by SEQ ID NO: 23, DNA containing the base sequence represented by 40 or the like is used.
  • the polynucleotide encoding the partial peptide of the present invention may be any polynucleotide as long as it contains a base sequence encoding the above-described partial peptide of the present invention.
  • DNA is preferred.
  • the DNA may be any of genomic DNA, genomic DNA library, cDNA derived from the above-described cells and tissues, cDNA library derived from the above-described cells and tissues, and synthetic DNA. '
  • Examples of the DNA encoding the partial peptide of the present invention include, for example, DNA having a part of the DNA having the base sequence represented by SEQ ID NO: 3, SEQ ID NO: 13 or SEQ ID NO: 23, or SEQ ID NO: 3 A DN having a nucleotide sequence that hybridizes under high stringent conditions to the nucleotide sequence represented by SEQ ID NO: 1.3 or SEQ ID NO: 23, and encoding a protein having substantially the same activity as the protein of the present invention. DNA containing a part of A is used.
  • the DNA that can be hybridized with the base sequence represented by SEQ ID NO: 3, SEQ ID NO: 13 or SEQ ID NO: 23 has the same significance as described above.
  • the DNA or DNA encoding the protein or partial peptide of the present invention (hereinafter, these may be simply referred to as the protein of the present invention in the description of the cloning and expression of the DNA encoding the same).
  • the DNA of the present invention is amplified by PCR using a synthetic DNA primer having a part of the nucleotide sequence encoding the protein of the present invention, or the DNA of the present invention is incorporated into an appropriate vector. Selection can be performed by hybridization with a DNA fragment encoding a part or all of the region or labeled with a synthetic DNA. The hybridization can be performed according to, for example, the method described in Molecular Cloning 2nd (J. Sabrook et al., Cold Spring Harbor Lab. Press, 1989). Ma When a commercially available library is used, it can be performed according to the method described in the attached instruction manual.
  • the DNA base sequence can be converted by PCR or a known kit such as Mutan TM -Super Express Km (Takara Shuzo) or Mutan TM -K (Takara Shuzo) using the 0DA-LA PCR method or the like. It can be performed according to a known method such as the gapped duplex method or the Kunkel method, or a method analogous thereto.
  • the DNA encoding the cloned protein can be used as it is depending on the purpose, or can be used after digestion with a restriction enzyme or addition of a linker, if desired.
  • the DNA may have ATG as a translation initiation codon on the 5, terminal side, and TAA, TGA or TAG as a translation termination codon on the 3, terminal side. These translation initiation codon and translation termination codon can also be added using a suitable synthetic DNA adapter.
  • the expression vector of the protein of the present invention can be prepared, for example, by (a) cutting out a DNA fragment of interest from DNA encoding the protein of the present invention, and (mouth) converting the DNA fragment into a promoter in an appropriate expression vector. It can be manufactured by connecting downstream of
  • the vector examples include a plasmid derived from E. coli (eg, pBR322, PBR32 5, pUC 12, pUC 13), a plasmid derived from Bacillus subtilis (eg, pUB 110, pTP 5, pC 194), a yeast-derived plasmid (eg, pSH19, pSH15)
  • pacteriophage such as ⁇ phage
  • animal viruses such as retrovirus, vaccinia virus, and baculovirus, pAl-11, pXTl, pR cZCMV, pRc / RSV, pc DNA I ZNe o and the like are used.
  • the promoter used in the present invention may be any suitable promoter corresponding to the host used for gene expression.
  • SRa promoter SV40 promoter, LTR promoter, CMV promoter, HSV-TK promoter, etc.
  • CMV (cytomegalovirus) promoter It is preferable to use SRo!
  • the host is Escherichia, the trp promoter, lac promoter, recA promoter, ⁇ PL promoter If the host is a Bacillus genus, SP01 promoter, SP02 promoter, penP promoter, etc.If the host is yeast, the host is yeast.
  • the expression vector may contain, in addition to the above, an enhancer, a splicing signal, a poly-A addition signal, a selection marker, and an SV40 replication origin (hereinafter sometimes abbreviated as SV400 ri), if desired. Anything can be used.
  • the selection marker one, for example, dihydrofolate reductase (hereinafter sometimes abbreviated as dh fr) gene [Mesotorekise Ichito (MTX) resistance], ampicillin phosphorus resistant gene (hereinafter sometimes abbreviated as Amp r ), Neomai, Shin-resistant gene (hereinafter sometimes abbreviated as Ne o r, include G418 resistance) and the like.
  • dh fr dihydrofolate reductase
  • Amp r ampicillin phosphorus resistant gene
  • Ne o r include G418 resistance
  • the target gene can be selected using a thymidine-free medium.
  • a signal sequence suitable for the host is added to the N-terminal side of the protein of the present invention. If the host is Escherichia, the Pho A signal sequence, Omp A signal sequence, etc., if the host is Bacillus, the amylase signal sequence, subtilisin signal sequence, etc. In the case of yeast, MFa signal sequence and SUC2 signal sequence can be used. When the host is an animal cell, insulin signal sequence, high interferon signal sequence, antibody molecule and signal sequence can be used, respectively. .
  • a transformant can be produced.
  • Examples of the host include Escherichia, Bacillus, yeast, insect cells, insects, animal cells, and the like.
  • Escherichia include, for example, Escherichia coli.
  • Bacillus bacteria examples include, for example, Bacillus subtilis (Bacillus).
  • subtilis MI114 (Gene, 24, 255 (1983)), 207-21 (Journal of Biochemistry, 95, 87 (1984)) and the like are used.
  • yeast include, for example, Saccharomyces cerevisiae AH22, AH22R—, ⁇ 87-11A, DKD-5D, 20B-12, Schizosaccharomyces porabe, NC YC 1913, NCYC 2036 Pastoris (Pichia pastoris) K # 71 or the like is used.
  • insect cells for example, when the virus is Ac NPV, a cell line derived from a larva of night roth moth (Spodoptera frugiperda cell; Sf cell), MG1 cell derived from the midgut of Trichoplusia ni, and egg derived from Trichoplusia ni egg High Five TM cells, cells derived from Mamestra brassicae, cells derived from Estigmena acrea, and the like are used.
  • Sf cells include Sf9 cells (ATCC
  • insects for example, silkworm larvae are used [Maeda et al., Nature, Vol. 315, 592 (1985)].
  • animal cells examples include monkey cell COS-7, Vero, Chinese hamster cell CHO (hereinafter abbreviated as CHO cell), dh fr gene-deficient chinini—zhamster cell CH ⁇ (hereinafter CHO (dh fr)). Cells), mouse L cells, mouse AtT-20, mouse myeoma cells, rat GH3, human FL cells, and the like.
  • Transformation of the genus Escherichia can be performed, for example, according to the method described in Proc. Natl; Acad. Sci. USA, 69, 2110 (1972) or Gene, 17, 107 (1982). it can. Transformation of Bacillus spp. Can be performed, for example, according to the method described in Molecular & General Genetics, Volume 168, 111 (1979).
  • the yeast is transformed according to the method described in, for example, Methods in Enzymology, Vol. 194, 182-187 (1991), Proc. Natl. Acad. Sci. USA, Vol. 75, 1929 (1978). be able to.
  • Insect cells or insects can be transformed, for example, according to the method described in Bio / Technology, 6, 47-55 (1988).
  • a liquid medium is suitable as a medium used for the cultivation, and a carbon source necessary for the growth of the transformant is contained therein.
  • Nitrogen sources inorganic substances and others.
  • carbon sources include glucose, dextrin, soluble starch, and sucrose.
  • nitrogen sources include ammonium salts, nitrates, corn chip lica, peptone, zein, meat extract, soybean meal, and potato extract.
  • the inorganic or organic substance such as a liquid and the inorganic substance include calcium chloride, sodium dihydrogen phosphate, and magnesium chloride.
  • yeast extract, vitamins, growth promoting factors and the like may be added.
  • the pH of the medium is preferably about 5-8.
  • Examples of a medium for culturing Escherichia bacteria include, for example, an M9 medium containing glucose and casamino acids [Miller, Journal “Obexperiment”, “Molecular”, Journal of Experiments in
  • a drug such as 3; 6-indolylacrylic acid can be added to make the promoter work efficiently.
  • the host is a bacterium belonging to the genus Escherichia
  • the cultivation is usually carried out at about 15 to 43 ° C for about 3 to 24 hours, and if necessary, aeration and stirring can be applied.
  • cultivation is usually performed at about 30 to 40 ° C for about 6 to 24 hours.
  • the medium used is Grace's Insect Medium (Grace, TC, Nature, 195, 788 (1962)). Those to which additives such as serum are appropriately added are used.
  • the ⁇ of the culture medium is adjusted to about 6.2 to 6.4. Culture is usually performed at about 27 ° C for about 3 to 5 days, and aeration and agitation are added as necessary.
  • examples of the medium include a MEM medium containing about 5 to 20% fetal bovine serum (Science, 122, 501 (1952)), a DMEM medium [Virology, 8, 396 (1959)], RPMI 1640 medium [The Journal of the American Medical Association 199, 519 (1967)], 199 medium [Proceeding of the Society for the Biological Medicine, 73] , 1 (1950)].
  • the pH is about 6-8.
  • Cultivation is usually performed at about 30 ° C (: up to 40 ° C for about 15 to 60 hours, and aeration and agitation are added as necessary.
  • the protein of the present invention can be produced in the cells, in the cell membrane, or outside the cells of the transformant.
  • the protein of the present invention can be separated and purified from the culture by, for example, the following method.
  • the cells or cells are collected by a known method, suspended in an appropriate buffer, After lysing cells or cells by lysozyme and Z or freeze-thawing, a method of obtaining a crude protein extract by centrifugation or filtration is used as appropriate.
  • the buffer may contain a protein denaturant such as urea or guanidine hydrochloride, or a surfactant such as Triton X-1000 TM .
  • Purification of the protein contained in the culture supernatant or extract obtained in this manner can be performed by appropriately combining known separation and purification methods.
  • These known separation and purification methods mainly include methods using solubility such as salting out and solvent precipitation, dialysis, ultrafiltration, gel filtration, and SDS-polyacrylamide gel electrophoresis, mainly molecular weight.
  • Methods that utilize differences in charge methods that use differences in charge, such as ion-exchange chromatography, methods that use specific affinities, such as affinity chromatography, and hydrophobicity, such as reverse-phase high-performance liquid chromatography.
  • a method using the difference between the isoelectric points, such as an isoelectric focusing method, and the like, may be used.
  • the protein thus obtained when obtained in a free form, it can be converted to a salt by a known method or a method analogous thereto, and conversely, when the protein is obtained in the form of a salt, a known method or a method analogous thereto Can be converted into a free form or another salt.
  • the protein produced by the recombinant can be arbitrarily modified or the polypeptide can be partially removed by the action of an appropriate protein modifying enzyme before or after purification.
  • an appropriate protein modifying enzyme for example, trypsin, chymotrypsin, arginyl endopeptidase, protein kinase, glycosidase and the like are used.
  • the presence of the protein of the present invention thus produced can be measured by, for example, enzymatic immunoassay western blotting using a specific antibody.
  • the antibody against the protein or partial peptide of the present invention or a salt thereof may be any of a polyclonal antibody and a monoclonal antibody as long as it can recognize the protein or partial peptide of the present invention or a salt thereof.
  • An antibody against the protein or partial peptide of the present invention or a salt thereof (hereinafter, these may be simply abbreviated to the protein of the present invention in the description of the antibody) is obtained by using the protein of the present invention as an antigen.
  • the antibody or antiserum can be produced according to the following method.
  • the protein of the present invention is administered to a warm-blooded animal itself or together with a carrier or diluent at a site capable of producing an antibody upon administration.
  • Complete Freund's adjuvant / incomplete Freund's adjuvant may be administered in order to enhance antibody production upon administration. Administration is usually performed once every 2 to 6 weeks, for a total of 2 to 10 times.
  • Examples of the warm-blooded animal to be used include monkeys, egrets, dogs, guinea pigs, mice, rats, sheep, goats, and chickens, and mice and rats are preferably used.
  • a warm-blooded animal immunized with an antigen for example, an individual with an antibody titer is selected from a mouse, and the spleen or lymph node is collected 2 to 5 days after the final immunization and contained in them.
  • an individual with an antibody titer is selected from a mouse, and the spleen or lymph node is collected 2 to 5 days after the final immunization and contained in them.
  • a monoclonal antibody-producing hybridoma can be prepared.
  • the antibody titer in the antiserum can be measured, for example, by reacting the labeled protein described below with the antiserum, and then measuring the activity of the labeling agent bound to the antibody.
  • the fusion operation can be performed according to a known method, for example, the method of Koehler and Milstein Nature, 256, 495 (1975)].
  • 'Fusion promoters include, for example, polyethylene glycol (PEG), Sendai virus and the like, and preferably PEG is used.
  • myeloma cells examples include myeloma cells of warm-blooded animals such as NS-1, P3U1, SP 2/0, and AP-1, but P3U1 is preferably used.
  • the preferred ratio between the number of antibody-producing cells (spleen cells) and the number of myeloma cells used is about 1: 1 to 20: 1, and PEG (preferably PEG1000 to PEG6000) is used at a concentration of about 10 to 80%.
  • Cell fusion can be carried out efficiently by adding the mixture and incubating at 20 to 40 ° C, preferably 30 to 37 ° C, for 1 to 10 minutes.
  • a hybridoma culture supernatant is added to a solid phase (eg, a microplate) on which a protein antigen is adsorbed directly or together with a carrier.
  • a solid phase eg, a microplate
  • an anti-immunoglobulin antibody (anti-mouse immunoglobulin antibody is used if the cell used for cell fusion is a mouse) or protein A labeled with a radioactive substance or an enzyme, and a monoclonal antibody bound to the solid phase Antibody detection method, Add the hybridoma culture supernatant to a solid phase to which anti-immunoglobulin antibody or protein A is adsorbed, add proteins labeled with radioactive substances, enzymes, etc. Examples include a method for detecting a single antibody.
  • the selection of the monoclonal antibody can be performed according to a known method or a method analogous thereto. Usually, it can be performed in an animal cell culture medium supplemented with HAT (hypoxanthine, aminopterin, thymidine).
  • HAT hyperxanthine, aminopterin, thymidine
  • any medium can be used as long as it can grow a hybridoma.
  • RPMI 1640 medium containing 1 to 20%, preferably 10 to 20% fetal bovine serum, and GIT medium containing 1 to 10% fetal bovine serum (Wako Pure Chemical Industries, Ltd. )
  • serum-free medium for hybridoma culture SFM-101, Nissui Pharmaceutical Co., Ltd.
  • the culture temperature is usually 20 to 40, preferably about 37 ° C.
  • the culture time is generally 5 days to 3 weeks, preferably 1 week to 2 weeks.
  • the culture can be usually performed under 5% carbon dioxide gas.
  • the antibody titer of the hybridoma culture supernatant can be measured in the same manner as the measurement of the antibody titer in the antiserum described above.
  • Monoclonal antibodies can be separated and purified by known methods, for example, immunoglobulin separation and purification methods (eg, salting out method, alcohol precipitation method, isoelectric point precipitation method, electrophoresis method, ion exchanger (eg, DEAE)).
  • immunoglobulin separation and purification methods eg, salting out method, alcohol precipitation method, isoelectric point precipitation method, electrophoresis method, ion exchanger (eg, DEAE)
  • an active adsorbent such as protein A or protein G and the bond is dissociated to obtain the antibody.
  • the polyclonal antibody of the present invention is produced according to a known method or a method analogous thereto. can do.
  • a immunizing antigen protein antigen
  • a complex thereof with a carrier protein is formed, and immunization is performed on a warm-blooded animal in the same manner as in the above-described method for producing a monoclonal antibody. It can be produced by collecting the contents and separating and purifying the antibody.
  • the type of the carrier protein and the mixing ratio between the carrier and the hapten are different from those of the hapten immunized by cross-linking the carrier.
  • Any antibody may be cross-linked at any ratio as long as it can be efficiently produced.
  • serum albumin, thyroglobulin, hemocyanin, etc. in a weight ratio of about 0.1 to hapten per hapten, may be used. -20, preferably about 1-5.
  • Various condensing agents can be used for force coupling between the hapten and the carrier, but glutaraldehyde, carbodiimide, a maleimide active ester, an active ester reagent containing a thiol group or a dithioviridyl group, or the like is used. .
  • the condensation product is administered to a warm-blooded animal itself or together with a carrier or diluent at a site where antibody production is possible.
  • Complete Freund's adjuvant / incomplete Freund's adjuvant may be administered in order to enhance the antibody-producing ability upon administration.
  • the administration is usually made once every about 2 to 6 weeks, for a total of about 3 to 10 times.
  • the polyclonal antibody can be collected from the blood, ascites, etc., preferably from the blood of a warm-blooded animal immunized by the above method.
  • the measurement of the polyclonal antibody titer in the antiserum can be performed in the same manner as the measurement of the antibody titer in the antiserum described above. Separation and purification of the polyclonal antibody can be performed according to the same method for separation and purification of immunoglobulin as in the above-described separation and purification of the monoclonal antibody.
  • an antisense polynucleotide having a substantially complementary base sequence or a part thereof includes a base sequence complementary to or substantially complementary to the base sequence of the DNA of the present invention or a part thereof. Any antisense having an action capable of suppressing the expression of the DNA. Although it may be a polynucleotide, antisense DNA is preferred.
  • the nucleotide sequence substantially complementary to the DNA of the present invention is, for example, the entire nucleotide sequence or a partial nucleotide sequence of the nucleotide sequence complementary to the DNA of the present invention (that is, the complementary strand of the DNA of the present invention). And about 70% or more, preferably about 80% or more, more preferably about 90% or more, and most preferably about 95% or more.
  • the complementary strand of the nucleotide sequence of the portion encoding the N-terminal portion of the protein of the present invention is about 70%. %, Preferably at least about 80%, more preferably at least about 90%, most preferably at least about 95%.
  • nucleotide sequence complementary or substantially complementary to the nucleotide sequence of DNA having the nucleotide sequence represented by SEQ ID NO: 3, SEQ ID NO: 13 or SEQ ID NO: 23 Or a part thereof, preferably, for example, a nucleotide sequence complementary to the nucleotide sequence of DNA having the nucleotide sequence represented by SEQ ID NO: 3, SEQ ID NO: 13 or SEQ ID NO: 23, or Antisense polynucleotides having a part thereof are exemplified.
  • An antisense polynucleotide is usually composed of about 10 to 40 bases, preferably about 15 to 30 bases.
  • the phosphate residues (phosphates) of each nucleotide constituting the antisense DNA are, for example, chemically modified phosphate residues such as phosphorothioate, methylphosphonate, and phosphorodithionate. May be substituted.
  • These antisense polynucleotides can be produced using a known DNA synthesizer or the like.
  • an antisense polynucleotide (nucleic acid) corresponding to the protein gene of the present invention, which can inhibit the replication or expression of the gene, is cloned or the determined protein is encoded. It can be designed and synthesized based on DNA base sequence information.
  • Such an antisense polynucleotide can hybridize to RNA of the protein gene of the present invention, inhibit the synthesis or function of the RNA, or bind to the protein-related RNA of the present invention.
  • Polynucleotides complementary to the selected sequence of the protein-related RNA of the present invention, and polynucleotides capable of specifically hybridizing with the protein-related RNA of the present invention include the protein of the present invention in vivo and in vitro. It is useful for regulating and controlling gene expression, and is also useful for treating or diagnosing diseases.
  • the term "corresponding" means having homology or being complementary to a specific sequence of nucleotides, base sequences or nucleic acids including genes.
  • nucleotide, nucleotide sequence or nucleic acid usually refers to the amino acids of the (directed) protein derived from the nucleotide (nucleic acid) sequence or its complement.
  • the 5 'end hairpin loop of the protein gene usually refers to the amino acids of the (directed) protein derived from the nucleotide (nucleic acid) sequence or its complement.
  • any region in a protein gene can be selected as a target.
  • a hairpin loop or the like can be selected as a preferred target region, any region in a protein gene can be selected as a target.
  • the nucleic acid of interest can hybridize to the target region, the nucleic acid of interest can be said to be "antisense" to the polynucleotide of the target region.
  • Antisense polynucleotides can be polydexoxy liponucleotides containing 2-deoxy D-reports, polyliponucleotides containing D-reports, N-glycosides of purine or pyrimidine bases and other types of polynucleotides.
  • Other polymers having nucleotide or non-nucleotide backbones eg, commercially available protein nucleic acids and synthetic sequence-specific nucleic acid polymers
  • other polymers containing special bonds provided that such polymers are found in DNA or RNA
  • nucleases nucleases, nuclease inhibitors, toxins, antibodies, signal peptides, poly-L-lysine, etc.
  • sugars eg, monosaccharides
  • containing acridine, psoralen, etc. containing chelating compounds (eg, metals, radioactive metals, boron, oxidizing metals, etc.), containing alkylating agents, modified bonds (Eg, a anomeric nucleic acid).
  • chelating compounds eg, metals, radioactive metals, boron, oxidizing metals, etc.
  • alkylating agents eg, a anomeric nucleic acid
  • nucleoside may include not only those containing purine and pyrimidine bases but also those having other modified heterocyclic bases.
  • Modified nucleotides and modified nucleotides may also be modified at the sugar moiety, e.g., where one or more hydroxyl groups have been replaced with a halogen, an aliphatic group, or the like, or a functional group, such as an ether or an amine. Conversion.
  • the antisense polynucleotide of the present invention is RNA, DNA or a modified nucleic acid (RNA, DNA).
  • modified nucleic acid include a sulfur derivative of a nucleic acid, a thiophosphate derivative, a polynucleoside amide / a nucleic acid that is resistant to degradation of oligonucleoside amide, and the like.
  • the antisense polynucleotide of the present invention can be designed, for example, as follows.
  • the antisense polynucleotide is more stable in the cell, the cell permeability of the antisense polynucleotide is increased, the affinity for the target sense strand is increased, and If so, reduce the toxicity of the antisense polynucleotide.
  • the antisense polynucleotides of the present invention may contain altered or modified sugars, bases, or bonds, may be provided in special forms such as ribosomes, microspheres, or may be applied by genetic therapy. Can be given in an added form.
  • the addition forms include polycations such as polylysine, which acts to neutralize the charge on the phosphate backbone, and lipids, which enhance the interaction with cell membranes and increase the uptake of nucleic acids ( For example, phosphoric acid, hydrophobic, cholesterol, etc.).
  • Preferred lipids for addition include cholesterol and its derivatives (eg, cholesteryl chromate formate, cholic acid, etc.). These can be attached to the 3 'or 5' end of the nucleic acid and can be attached via a base, sugar, or intramolecular nucleoside linkage.
  • Other groups are cap groups specifically located at the 3 'or 5' end of nucleic acids that prevent degradation by nucleases such as exonuclease and RNase. No. Examples of such a capping group include, but are not limited to, hydroxyl-protecting groups known in the art, such as diols such as polyethylene glycol and tetraethylene glycol.
  • the inhibitory activity of the antisense polynucleotide can be examined using the transformant of the present invention, the in vivo or in vitro gene expression system of the present invention, or the in vivo or in vitro translation system of the protein of the present invention. Can be.
  • the protein or partial peptide of the present invention or a salt thereof hereinafter sometimes abbreviated as the protein of the present invention
  • the DNA encoding the protein or partial peptide of the present invention hereinafter referred to as the D NA
  • an antibody against the protein or partial peptide of the present invention or a salt thereof hereinafter may be abbreviated as the antibody of the present invention
  • an antisense polynucleotide of the DNA of the present invention hereinafter may be referred to as an antibody.
  • an antibody which may be abbreviated as the antisense polynucleotide of the present invention.
  • the protein of the present invention is a G protein-coupled receptor protein, MC By binding to H, it plays an important role in promoting appetite (feeding) and promoting oxytocin secretion.
  • anorexia eg, nervous appetite
  • Anemia or hypoproteinia due to anorexia, weak labor, laxative bleeding, uterine remodeling failure, milk stasis, etc. develop various diseases.
  • the protein of the present invention and the DNA of the present invention include, for example, an appetite (feeding) enhancer, anorexia (eg, anorexia nervosa), anemia or hypoproteinemia associated with anorexia, weak labor pain It can be used as a medicament such as a preventive and remedy for flaccid hemorrhage, uterine remodeling failure, and milk stasis.
  • the protein of the present invention is reduced or deficient in the living body, and there is a patient in which the appetite (feeding) enhancement or oxytocin secretion promoting activity of MCH is not sufficiently or normally exerted
  • the DNA of the present invention When the DNA of the present invention is used as the above-described prophylactic or therapeutic agent, the DNA is used alone or in an appropriate vector such as a retrovirus vector, an adenovirus vector, or an adenovirus associated virus vector. Thereafter, it can be administered to humans or warm-blooded animals according to conventional means.
  • the DNA of the present invention can be administered as it is or in the form of a formulation together with a physiologically acceptable carrier such as an auxiliary agent for promoting uptake, and can be administered using a gene gun or a catheter such as a hide mouth gel catheter.
  • the protein of the present invention When the protein of the present invention is used as the prophylactic / therapeutic agent, the protein purified to at least 90%, preferably 95% or more, more preferably 98% or more, and still more preferably 99% or more. It is preferred to use.
  • the protein of the present invention can be used, for example, in the form of tablets, capsules, elixirs, microcapsules, and the like, which are sugar-coated as necessary, orally, or aseptically with water or other pharmaceutically acceptable liquids. It can be used parenterally in the form of injections, such as aqueous solutions or suspensions.
  • the protein of the present invention may be used together with physiologically acceptable carriers, flavoring agents, excipients, vehicles, preservatives, stabilizers, binders, and the like in a unit dosage form required for generally accepted pharmaceutical practice. It can be manufactured by mixing. The amount of the active ingredient in these preparations is such that a suitable dosage in the specified range can be obtained. '' Additives that can be incorporated into tablets, capsules, etc.
  • binders such as gelatin, corn starch, tragacanth, gum arabic, excipients such as crystalline cellulose, corn starch, gelatin, Swelling agents such as alginic acid, lubricants such as magnesium stearate, sweeteners such as sucrose, lactose or saccharine, and flavoring agents such as peppermint, cocoa oil or cellulose.
  • Swelling agents such as alginic acid, lubricants such as magnesium stearate, sweeteners such as sucrose, lactose or saccharine, and flavoring agents such as peppermint, cocoa oil or cellulose.
  • sweeteners such as sucrose, lactose or saccharine
  • flavoring agents such as peppermint, cocoa oil or cellulose.
  • a liquid carrier such as oil and fat can be further contained in the above-mentioned type of material.
  • Sterile compositions for injection can be formulated according to standard pharmaceutical practice, such as dissolving or suspending the active substance in vehicles such as
  • Aqueous liquids for injection include, for example, physiological saline, isotonic solutions containing glucose and other adjuvants (eg, D-sorbitol, D-mannitol, sodium chloride, etc.).
  • Agents for example, alcohols (eg, alcohol), polyalcohols (eg, propylene glycol, polyethylene glycol, etc.), non-ionic surfactants (eg, Polysorbate 80 TM , HC ⁇ —50 etc.).
  • examples of the oily liquid include sesame oil, soybean, and oil, and may be used in combination with a solubilizing agent such as benzyl benzoate or benzyl alcohol.
  • buffers eg, phosphate buffer, sodium acetate, buffer, etc.
  • soothing agents eg, benzalkonium chloride, proforce hydrochloride, etc.
  • the prepared injection is usually filled in an appropriate ampoule.
  • Stabilizers eg, human serum albumin, polyethylene glycol, etc.
  • preservatives eg, benzyl alcohol, phenol, etc.
  • antioxidants etc. ' May be combined.
  • the prepared injection is usually filled in an appropriate ampoule.
  • the vector into which the DNA of the present invention has been inserted is also formulated in the same manner as described above, and is usually used parenterally.
  • the preparations obtained in this way are safe and have low toxicity, for example, in warm-blooded animals (e.g., humans, rats, mice, guinea pigs, egrets, birds, higgies, bush, pests, pests, cats, Dogs, monkeys, chimpanzees, etc.).
  • warm-blooded animals e.g., humans, rats, mice, guinea pigs, egrets, birds, higgies, bush, pests, pests, cats, Dogs, monkeys, chimpanzees, etc.
  • the dosage of the protein of the present invention varies depending on the target disease, the administration subject, the administration route, and the like.
  • the adult as 60 kg
  • the single dose varies depending on the administration subject, the target disease, and the like.
  • the protein of the present invention when administered to an adult (with a body weight of 60 kg) in the form of an injection for the purpose of treating anorexia nervosa, It is convenient to administer the protein by injecting about 0.01 to 30 mg, preferably about 0.1 to 20 mg, more preferably about 0.1 to 10 mg of the protein per day into the affected area. 60 kg for other animals Amount converted to have enough can be administered.
  • the protein of the present invention is useful as a reagent for screening a compound or a salt thereof that changes the binding between the peptide of the present invention (eg, MCH) and the protein of the present invention.
  • a compound or a salt thereof that changes the binding between the peptide of the present invention (eg, MCH) and the protein of the present invention.
  • a method for screening a salt thereof, or a kit for screening a compound or a salt thereof, which changes the binding property between the peptide of the present invention and the protein of the present invention, which comprises using the peptide of the present invention and the protein of the present invention (
  • the screening method of the present invention and the kit for screening of the present invention are abbreviated in detail below.
  • a compound that alters the binding between the peptide of the present invention and the protein of the present invention a compound that alters the binding between the peptide of the present invention and the protein of the present invention (for example, peptide, protein, non-peptide) Compounds, synthetic compounds, fermentation products, etc.) or salts thereof.
  • Such compounds cell stimulating activity via protein of the present invention (e.g., Arakidon acid release, acetylcholine release, intracellular C a 2 + release, intracellular c AM P producing formation, intracellular c AM P production inhibition , Intracellular cGMP production, Inositol phosphate production, Cell membrane potential fluctuation, Intracellular protein phosphorylation, c-fos activation, pH decrease, cAMP-dependent protein kinase activation, cGMP Activation of protein-dependent protein kinase, activation of phospholipid-dependent protein kinase, activity of promoting or inhibiting the activation of microtubule-associated protein kinase (MAP kinase), etc.
  • MAP kinase microtubule-associated protein kinase
  • the present invention relates to (i) the case where the protein of the present invention is brought into contact with the peptide of the present invention, and (ii) the case where the above-mentioned protein of the present invention is brought into contact with the peptide of the present invention and a test compound. And a method for screening a compound or a salt thereof, which changes the binding property between the peptide of the present invention and the protein of the present invention, characterized in that the comparison is performed.
  • the screening method of the present invention (i) the case where the above-mentioned protein of the present invention is brought into contact with the peptide of the present invention and (ii) the above-mentioned protein of the present invention which comes in contact with the peptide of the present invention and a test compound.
  • the amount of the peptide of the present invention bound to the protein of the present invention, the cell stimulating activity and the like are measured and compared.
  • screening method of the present invention include, for example, (a) the peptide of the present invention relative to the protein of the present invention when the peptide of the present invention is brought into contact with the protein of the present invention, and when the peptide and the test compound of the present invention are brought into contact with the protein of the present invention; A method for screening a compound or a salt thereof that changes the binding property between the peptide of the present invention and the protein of the present invention, which comprises measuring and comparing the amount of binding;
  • the peptide of the present invention and the peptide of the present invention are characterized by measuring and comparing the amount of binding of the peptide of the present invention to the cell or the membrane fraction when brought into contact with the membrane fraction of the cell.
  • the peptide of the present invention is a labeled peptide, such as the screening method according to any one of (a) to (c) above,
  • the protein of the present invention used in the screening method of the present invention may be any protein containing the above-described protein of the present invention.
  • the protein of the present invention which is expressed in a large amount using a recombinant is suitable.
  • the aforementioned methods and the like are used.
  • the preparation method described later when cells containing the protein of the present invention or the cell membrane fraction are used, the preparation method described later may be followed.
  • the cell When a cell containing the protein of the present invention is used, the cell may be immobilized with datalaldehyde, formalin, or the like.
  • the immobilization method can be performed according to a known method.
  • the cell containing the protein of the present invention refers to a host cell expressing the protein of the present invention. Examples of the host cell include the aforementioned Escherichia coli, Bacillus subtilis, yeast, insect cells, animal cells and the like. .
  • the membrane fraction refers to a fraction containing a large amount of cell membrane obtained by a known method after cell disruption.
  • the cells can be crushed by crushing the cells with a Potter-Elvehj em homogenizer, crushing with a Warlinda blender-Polytron (Kinematica), crushing with ultrasonic waves, or pressing with a French press. Crushing by ejecting cells from thin nozzles.
  • centrifugal fractionation methods such as differential centrifugation and density gradient centrifugation are mainly used.
  • the cell lysate is centrifuged at a low speed (500-3000 rpm) for a short time (usually about 1-10 minutes), and the supernatant is further centrifuged at a high speed (15000-30,000 rpm) for 30 minutes to 2 hours to obtain a precipitate.
  • a low speed 500-3000 rpm
  • a high speed 15000-30,000 rpm
  • the membrane fraction is rich in the expressed protein of the present invention and membrane components such as cell-derived phospholipids and membrane proteins.
  • the amount of the protein of the present invention in the cells or membrane fraction containing the protein of the present invention is preferably 10 3 to 10 8 molecules per cell, more preferably 10 5 to 10 7 molecules per cell. is there.
  • the protein fraction of the present invention and the peptide of the present invention are used.
  • the protein fraction of the present invention is preferably a natural protein fraction of the present invention or a recombinant protein fraction of the present invention having an activity equivalent thereto.
  • the equivalent activity means equivalent ligand binding activity and the like.
  • the labeled peptide such as a radioactive isotope (e.g., [125 1], [131 1], [3 ⁇ 4], [ "C], [32 P], [33 P], etc.
  • Fluorescent substances eg, cyanine fluorescent dyes (eg, Cy2, Cy3, Cy5, Cy5.5, Cy7 (manufactured by Amersham Bioscience), etc.), fluorescamine, fluorescein sothiosinate, etc.
  • enzymes eg, —galactosidase, j6—labeled with darcosidase, alkaline phosphatase, oxidase, malate dehydrogenase, etc.
  • luminescent substances eg, luminol, luminol derivatives, J-reciferin, lucigenin, etc.
  • piotin lanthanide, etc.
  • the peptide of the present invention prepared by a known method using a Porton-Hunter reagent can be used. It is also possible to use the body.
  • a compound that alters the binding between the peptide of the present invention and the protein of the present invention first, cells containing the protein of the present invention or the membrane fraction of the cells are screened.
  • a buffer suitable for tanning Any buffer may be used as long as it does not inhibit the binding between the ligand and the receptor, such as a phosphate buffer having a pH of 4 to 10 (preferably, a pH of 6 to 8), such as Tris-HCl buffer.
  • a surfactant such as CHAPS, Tween-80 TM (Kao-Atlas), digitonin, and dexcholate can be added to the buffer.
  • a protease inhibitor such as PMSF, leptin, E-64 (manufactured by Peptide Research Laboratories), and peptide sutin is added to suppress the degradation of the protein of the present invention and the peptide of the present invention by protease. You can also.
  • a certain amount 5,000 to 500,000 cpm
  • NBS non-specific binding
  • the reaction is carried out at 0 to 50 ° C, preferably 4 to 37 ° C, for 20 minutes to 24 hours, preferably 30 minutes to 3 hours.
  • the reaction solution is filtered through a glass fiber filter or the like, washed with an appropriate amount of the same buffer, and the radioactivity remaining on the glass fiber filter is measured using a liquid scintillation counter or a counter.
  • the specific binding amount (B-NSB) is For example, a test compound having 50% or less can be selected as a candidate substance having a competitive inhibitory ability.
  • BIAcore manufactured by Amersham Pharmacia Biotech
  • the peptide of the present invention is immobilized on a sensor chip by an amino coupling method according to the protocol attached to the device, and contains a cell containing the protein of the present invention or a DNA encoding the protein of the present invention.
  • the protein of the present invention is immobilized on a sensor chip, and a buffer solution such as a phosphate buffer or a Tris buffer containing the peptide of the present invention or the peptide of the present invention and a test compound is passed over the sensor chip.
  • a buffer solution such as a phosphate buffer or a Tris buffer containing the peptide of the present invention or the peptide of the present invention and a test compound is passed over the sensor chip.
  • Test compounds include those similar to the above.
  • Atsusi system mediated cell stimulating activities proteins of the present invention e.g., Arakidon acid release, Asechirukori emissions release, intracellular C a 2 + release, intracellular c AM P generated , Inhibition of intracellular cAMP production, Intracellular cGMP production, Wild boar!
  • ⁇ -Monophosphate production Cell membrane potential fluctuation, Intracellular protein Phosphorylation, activation of c-fos, decrease of pH, activation of cAMP-dependent protein kinase, activation of cGMP-dependent protein kinase, activation of phospholipid-dependent protein kinase,
  • the activity of promoting or suppressing the activation of microtubule-associated protein kinase (MAP kinase), etc.) can be measured using a known method or a commercially available measurement kit. Specifically, first, cells containing the protein of the present invention are cultured on a multiwell plate or the like.
  • cells expressing an appropriate protein of the present invention are used.
  • the cells expressing the protein of the present invention the above-mentioned recombinant cell lines expressing the protein of the present invention and the like are desirable.
  • the transformant expressing the protein of the present invention may be a stable expression strain or a transient expression strain.
  • the same kind of animal cells as described above are used.
  • Test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and the like.
  • GTPaS When a receptor-expressing cell is stimulated by a receptor agonist, an intracellular G protein is activated and GTP is bound. This phenomenon is also observed in the membrane fraction of receptor-expressing cells. Normally, GTP is hydrolyzed to GDP, but if GTPaS is added to the reaction solution at this time, GTP7S binds to G protein like GTP, but is hydrolyzed. Without G cells The state of being bonded to the membrane is maintained. If labeled GTPaS is used, the activity of stimulating receptor agonist receptor-expressing cells can be measured by measuring the labeled GTPaS remaining on the cell membrane.
  • the stimulatory activity of the peptide of the present invention on the protein-expressing cell of the present invention is measured, whereby a compound that changes the binding property between the peptide of the present invention and the protein of the present invention is obtained. You can screen.
  • This method is performed using a membrane fraction containing the protein of the present invention.
  • the substance exhibiting the activity of promoting the GTP-A binding to the protein membrane fraction of the present invention is agonist.
  • the case where the peptide of the present invention is brought into contact with the protein cell membrane fraction of the present invention in the presence of labeled GTP TS and the case where the peptide and test compound of the present invention are brought into contact with the protein cell membrane fraction of the present invention.
  • a compound that alters the binding property between the peptide of the present invention and the protein of the present invention is determined. Screen.
  • the peptide GTP T S binding promoting activity test compound showing activity to suppress to proteins the cell membrane fraction of the present invention according to the present invention can be selected as Ah Ru candidates competitive inhibition.
  • screening of agonist is carried out by bringing only the test compound into contact with the protein cell membrane fraction of the present invention and measuring the GTP-AS binding promoting activity to the protein cell membrane fraction of the present invention. You can also.
  • the cell membrane fraction containing the protein of the present invention prepared according to known methods, membrane dilution buffer (50 mM T ris, 5 mM M g C 1 2, 150 mM N a C 1, 1 M GDP, 0. Dilute with 1% BSA; pH 7.4).
  • the dilution ratio depends on the expression level of the receptor. This was dispensed by 0. 2 ml min F a 1 c 0 n 2053, a peptide or peptide and a test compound of the present invention of the present invention was added, further to a final concentration of 200 pM added [35 S] GTPrS.
  • wash ice-cold buffer 50 mM Tr is, 5mM MgC 1 2, 150 mM NaCl, 0.1% BSA, 0.05% CHAP S; pH 7.4
  • Add 15 ml and filter through glass fiber filter paper GFZF. After incubating for 65 and 30 minutes and drying, it was bound to the membrane fraction remaining on the filter paper with a liquid scintillation counter.
  • the radioactivity of [ 35 S] GTPrS is measured.
  • the radioactivity of the experimental group to which only the peptide of the present invention was added was 100%, and the radioactivity of the experimental group to which the peptide of the present invention was not added was 100%.
  • test compound having a GTPaS binding promoting activity of, for example, 50% or less can be selected as a candidate substance having competitive inhibitory ability.
  • the production of intracellular cAMP is suppressed by stimulation of the peptide of the present invention.
  • the binding between the peptide of the present invention and the protein of the present invention is changed.
  • the compound can be screened.
  • a compound that alters the binding between the peptide of the present invention and the protein of the present invention is determined by measuring the activity of inhibiting the production of intracellular cAMP in the cell when the cell is brought into contact with the cell and comparing the results. I do.
  • a substance that increases the amount of intracellular cAMP for example, forskolin, calcitonin and the like are used.
  • the amount of cAMP produced in the protein-expressing cells of the present invention is determined by comparing the amount of anti-cAMP antibody obtained by immunizing mice, rats, rabbits, goats, rabbits, etc. with [ 125 1] -labeled cAMP (both commercially available products). Can be measured using an RIA system by using EIA or an EIA system combining an anti-cAMP antibody and labeled cAMP. Moreover, the anti-cAMP antibody, using a protein A or beads containing scintillant which is secured using such antibodies to etc.
  • c A of the protein-expressing cell of the present invention by the peptide of the present invention A test compound exhibiting an activity of inhibiting MP production inhibitory activity can be selected as a candidate substance having a competitive inhibition ability.
  • a compound showing an agonist activity can be screened by contacting only the test compound with the protein-expressing cell of the present invention and examining the cAMP production inhibitory activity.
  • Cells expressing the protein of the present invention are seeded on a 24-well plate at 5 ⁇ 10 4 ce 1 LZwe 11 and cultured for 48 hours. Wash the cells with Hank's buffer (pH 7.4) containing 0.2 ⁇ 3-isobutyl-methylxanthine, 0.05% BSA and 2 OmM HEPES (hereinafter abbreviated as reaction buffer 1). Then add 0.5 ml of reaction buffer and incubate in the incubator for 30 minutes.
  • Hank's buffer pH 7.4
  • reaction buffer 1 containing 0.2 ⁇ 3-isobutyl-methylxanthine, 0.05% BSA and 2 OmM HEPES
  • reaction buffer Excluding the reaction buffer, add 0.25 ml of the reaction buffer to the cells, and then add 1 M of the peptide of the present invention or 1 M of the peptide of the present invention and 2 M forskolin to which the test compound is added. Add 0.25 ml of reaction buffer to the cells and incubate at 37 ° C for 24 minutes. Stop the reaction by adding 100 1 of 20% perchloric acid, and then leave it on ice for 1 hour to extract intracellular cAMP. The amount of cAMP in the extract is measured using a cAMP EIA kit (Amersham Pharmacia Biotech).
  • the amount of cAMP produced by the stimulation of forskolin was defined as 100%, and the amount of cAMP suppressed by the addition of 1 / M of the peptide of the present invention was defined as 0%.
  • the effect of the test compound on is calculated.
  • a test compound that inhibits the activity of the peptide of the present invention and has a cAMP production activity of, for example, 50% or more can be selected as a candidate substance capable of competitive inhibition.
  • the peptide of the present invention when using the protein-expressing cell of the present invention, which exhibits the property of increasing the amount of intracellular CAMP by stimulation of the peptide of the present invention, the peptide of the present invention was brought into contact with the protein-expressing cell of the present invention. And a test compound obtained by contacting the peptide of the present invention and the test compound with the protein-expressing cell of the present invention, by measuring and comparing the intracellular cAMP production promoting activity of the cell. Compounds that alter the binding to the proteins of the invention can be screened. In this method, a test compound showing an activity of inhibiting the cAMP production promoting activity of the protein-expressing cells of the present invention by the peptide of the present invention can be selected as a candidate substance having an antagonistic inhibitory ability. ''
  • a compound exhibiting agonist activity can be screened by contacting the test compound alone with the protein-expressing cell of the present invention and examining cAMP production promoting activity.
  • the AMP production promoting activity can be measured by adding the peptide of the present invention or the peptide of the present invention and the test compound to the protein-expressing cells of the present invention (eg, animal cells such as CHO cells) without adding forskolin in the above screening method.
  • the amount of cAMP produced by the addition of is determined and measured by the method described above.
  • the peptide of the present invention and the protein of the present invention can be measured.
  • Compounds that alter binding can be screened.
  • DNA containing CRE (cAMP response element) is inserted into the vector upstream of the reporter gene to obtain the CRE—repo overnight gene vector.
  • CRE cAMP response element
  • stimulation accompanied by an increase in cAMP is accompanied by CRE-mediated expression of the reporter gene, and subsequent stimulation of the gene product (protein) of the repo overnight gene. Induce production.
  • the reporter gene protein by measuring the enzymatic activity of the reporter gene protein, it is possible to detect a change in the amount of CAMP in cells into which the CRE-repo overnight gene vector has been introduced.
  • the peptide of the present invention when the peptide of the present invention is brought into contact with the protein-expressing cell of the present invention into which the peptide of the present invention has been introduced in the presence of a substance that increases the amount of intracellular cAMP, the peptide of the present invention And the test compound was brought into contact with the protein-expressing cell of the present invention when the CRE-repo overnight gene vector was introduced. The enzyme activity of the reporter gene protein was measured and compared. Screening for a compound that alters the binding between the peptide of the present invention and the protein of the present invention.
  • a substance that increases the amount of intracellular CAMP for example, forskolin, calcitonin and the like are used.
  • the vector for example, Pitka Gene Basic Vector, Pitka Gene Enhancer Vector (Toyo Ink Manufacturing Co., Ltd.) and the like are used.
  • the CRE-containing DNA is inserted into the above-mentioned vector in the repo overnight gene, for example, the multicloning site upstream of the luciferase gene, to obtain the CRE-reporter gene vector.
  • a test compound that restores the inhibition of the enzyme activity of the reporter gene protein by the peptide of the present invention can be selected as a candidate substance capable of competitive inhibition.
  • agonists can be screened by bringing only the test compound into contact with the protein-expressing cells of the present invention and measuring the same inhibition of the amount of luminescence increased by forskolin stimulation as with the peptides of the present invention.
  • Cells expressing the protein of the present invention into which the CRE-repo overnight gene (luciferase) has been introduced are seeded on a 24-well plate at 5 ⁇ 10 3 ce 1 lZwe 11 and cultured for 48 hours.
  • the cells are washed with Hank's buffer (pH 7.4) containing 0.2 mM 3_isobutyl-methylxanthine, 0.05% BSA and 20 mM HE PES (hereinafter abbreviated as reaction buffer). Then add 0.5 ml of application buffer and incubate for 30 minutes in the incubator.
  • the peptides of the present invention provide for luciferase-induced luminescence upon forskolin stimulation. Suppress increase in volume. A compound that restores the inhibition can be selected as a candidate substance having competitive inhibitory ability.
  • reporter genes for example, alkaline phosphatase, chloramphene
  • Acetyl trans f erase or a gene such as iS-galactosidase may be used.
  • the enzyme activity of these reporter gene proteins is measured according to a known method or using a commercially available measurement kit.
  • Alkaline phosphatase activity can be measured using, for example, Lumi-Phos 530 manufactured by Wako Pure Chemical, and chloramphenicol acetyltransferase can be measured using, for example, FAST CAT chrol amphenicol manufactured by Wako Pure Chemical.
  • the i3-galactosidase activity is measured using, for example, Acetyl trans erase erase Assay KiT using, for example, Aurora Ga1-XE manufactured by Wako Pure Chemical Industries.
  • the protein-expressing cells of the present invention release arachidonic acid metabolites extracellularly by stimulation of the peptides of the present invention.
  • This reaction to measure the stimulating activity of the peptide of the present invention on the cells expressing the protein of the present invention, it is possible to screen for a compound that alters the binding property between the peptide of the present invention and the protein of the present invention. .
  • the arachidonic acid metabolite-releasing activity can be measured by measuring the labeled arachidonic acid metabolite released outside the cell by incorporating the labeled arachidonic acid into the cells expressing the protein of the present invention in advance. can do. Specifically, when the peptide of the present invention is brought into contact with a cell expressing the protein of the present invention containing labeled arachidonic acid, the peptide of the present invention and the test compound are brought into contact with the cell of the present invention containing labeled arachidonic acid. By measuring and comparing the arachidonic acid metabolite release activity when brought into contact with a protein-expressing cell, a compound that changes the binding property between the peptide of the present invention and the protein of the present invention is screened. .
  • a test compound that inhibits the arachidonic acid metabolite release activity of the peptide of the present invention can be selected as a candidate substance having a competitive inhibition ability.
  • the test compound alone is brought into contact with the protein-expressing cell of the present invention, and the arachidonic acid metabolite releasing activity of the protein-expressing cell of the present invention is examined by a known method.
  • a compound exhibiting agonist activity can also be screened.
  • the protein-expressing cells of the present invention are seeded on a 24-well plate at 5 ⁇ 10 4 ce 11 / we 11, and cultured for 24 hours, and [ 3 H] arachidonic acid is added to 0.25 Ci / well. After 16 hours, wash the cells with Hanks buffer (pH 7.4) containing 0.05% BSA and 20 mM HE PES (hereinafter abbreviated as reaction buffer 1). A reaction buffer 500 ⁇ 1 containing a final concentration of 10 M of the peptide of the present invention or a final concentration of 10 / M of the peptide of the present invention and a test compound is added to each we11. After incubating at 37 ° C for 60 minutes, the reaction solution 4001 is added to the scintillator overnight, and the amount of [ 3 H] arachidonic acid metabolite + released in the reaction solution is measured by scintillation counting.
  • reaction buffer containing peptide free [3 H] Arakidon the amount of acid metabolites as 100%, free [3 H] Arakidon acid when the test compound added 'to addition of (test compound non-added additive) Calculate the amount of metabolite.
  • test compound having an arachidonic acid metabolite releasing activity of, for example, 50% or less can be selected as a candidate substance having an antagonistic ability.
  • the intracellular Ca concentration increases by stimulation with the peptide of the present invention.
  • the stimulating activity of the peptide of the present invention on the protein-expressing cell of the present invention is measured to screen for a compound that changes the binding property between the peptide of the present invention and the protein of the present invention. be able to. '
  • the intracellular calcium concentration in the case where the peptide of the present invention is brought into contact with the protein-expressing cell of the present invention and in the case where the peptide of the present invention and the test compound are brought into contact with the protein-expressing cell of the present invention By measuring and comparing the increasing activity, a compound that changes the binding property between the peptide of the present invention and the protein of the present invention is screened. The measurement is performed according to a known method.
  • an increase in intracellular calcium concentration by the peptide of the present invention is suppressed.
  • the test compound to be tested can be selected as a candidate substance capable of competitive inhibition.
  • an agonist can be screened by measuring an increase in fluorescence intensity due to the addition of the test compound alone.
  • the protein-expressing cells of the present invention are seeded on a sterilized cover glass for a microscope. Two days later, the culture solution is replaced with HBSS suspended in 4 mM Fura-2AM (Dojindo Laboratories), and incubated at room temperature. Leave for 30 minutes. After washing with HBSS, place a coverslip on the cuvette, add the peptide of the present invention or the peptide of the present invention and a test compound, and emit fluorescence at 505 nm at excitation wavelengths of 300 nm and 380 nm. Measure the increase in intensity ratio with a fluorimeter and compare.
  • FLIPR manufactured by Molecular Devices
  • F1uo-3AM manufactured by Dojindo Laboratories
  • F1uo-3AM manufactured by Dojindo Laboratories
  • a protein gene (eg, aequorin, etc.) that emits light due to an increase in intracellular Ca ion is co-expressed in the protein-expressing cell of the present invention, and the intracellular Ca ion concentration is increased.
  • the gene protein eg, aequorin or the like
  • a gene for a protein that emits light when the intracellular Ca ion increases is co-expressed, and the protein-expressing cell of the present invention is seeded on a 96-well plate, and the peptide of the present invention or the Add the peptide and test compound, measure the increase in the ratio of fluorescence intensity with a fluorimeter, and compare.
  • a test compound that suppresses an increase in fluorescence intensity due to the peptide of the present invention can be selected as a candidate substance having a competitive inhibition ability.
  • clone 24 of ETA (endothelin A receptor) -expressing CH0 cell Abbreviated as ETA24 cells. Journal of Pharmacology and Experimental
  • Therapeutics Vol. 279, pp. 675-685, 1996) was used to measure the activity of increasing the intracellular Ca ion concentration of the protein-expressing cells of the present invention and ETA24 cells using FLIPR (manufactured by Molecular Devices). This is performed using The cells expressing the protein of the present invention and ETA24 cells should be subcultured in DMEM supplemented with 10% dialyzed fetal calf serum (hereinafter referred to as dFBS). Protein expressing cells of the present invention, suspended in medium (10% d FBS-DMEM) such that ETA24 cells each 15xl0 4 cells / ml, in 96 well plate for FLIPR (Black plate clear bottom.
  • medium 10% d FBS-DMEM
  • H / HBSS Nisy Hanks 2 (Nissui Pharmaceutical Co., Ltd.) 9.8 g, sodium hydrogen carbonate 0.35 g, HEPES 4.77 g, adjusted to pH 7.4 with sodium hydroxide solution, filter sterilized) 20 ml, 250 mM Probenecid 200 ⁇ Mix 200 fetal serum (FBS).
  • Fluo 3-AM Diojindo Research Laboratories 2 piles (50 xg) was converted to dimethyl sulfoxide 401, 20% Pluronic acid
  • H / HBSS-Probenecid-FBS Molecular Probes 40, and added to the above H / HBSS-Probenecid-FBS. After mixing, pipette 100 l of each well into the cell plate from which the culture solution has been removed using an 8-tube pipette. % C0 2 incubator base 1 hour at 37 ° C for at one coater in incubate (dye the loading). H / HBSS 1501 containing 2.5 mM Probenecid and 0.1% CHAPS is added to the sample for each assay (each fraction), diluted, and transferred to a 96-well plate for FLIPR (V-Bottom plate, Coster) (hereafter, sample plate) ).
  • FLIPR V-Bottom plate, Coster
  • a peptide of the present invention was brought into contact with a protein-expressing cell of the present invention in the presence of labeled inositol, and a peptide and a test compound of the present invention were brought into contact with a protein-expressing cell of the present invention.
  • the inositol triphosphate-producing activity is measured and compared to screen for a compound that alters the binding property between the peptide of the present invention and the protein of the present invention. The measurement is performed according to a known method. .
  • a test compound that suppresses inositol triphosphate-producing activity can be selected as a candidate substance capable of competitive inhibition.
  • agonists can be screened by bringing only the test compound into contact with the protein-expressing cells of the present invention and measuring the increase in inositol triphosphate production.
  • the cells expressing the protein of the present invention are seeded on a 24-well plate and cultured for one day. Thereafter, the cells are cultured for 1 day in a medium supplemented with myo- [2-3 ⁇ 4] inositol (2.5 Ci / well), and the cells are thoroughly washed with a medium without radioactive inositol. After adding the peptide of the present invention or the peptide and the test compound of the present invention, 10% perchloric acid is added to stop the reaction. 1. neutralized with 5M potassium hydroxide and 60 mM HEPES solution and then passed through a column packed with AGlx8 tree fat (Bi o-Rad) in 0.
  • the radioactivity eluted with 1 M ammonium formate and 0.1 M formic acid is measured with a liquid scintillation counter.
  • the radioactivity when the peptide of the present invention is not added is 0%, and the radioactivity when the peptide of the present invention is added is 100%.
  • test compound having an inositol triphosphate-producing activity of, for example, 50% or less can be selected as a candidate substance having an antagonistic ability.
  • DNA containing a TRE is inserted upstream of the reporter gene of the vector to obtain a TRE-repo overnight gene vector.
  • TPA response element DNA containing a TRE (TPA response element) is inserted upstream of the reporter gene of the vector to obtain a TRE-repo overnight gene vector.
  • stimulation accompanied by an increase in the intracellular calcium concentration is caused by the TRE-mediated repo overnight gene expression and the subsequent reporter Induces the production of the gene product (protein) of the gene.
  • the TRE-reporter gene vector by measuring the enzymatic activity of the reporter gene protein, it is possible to detect fluctuations in the amount of calcium in the cells into which the TRE-reporter gene vector has been introduced.
  • the peptide of the present invention when the peptide of the present invention is brought into contact with a TRE-reporter gene vector-introduced protein-expressing cell of the present invention, the peptide of the present invention and the test compound are expressed in the TRE-reporter gene.
  • the vector examples include Pitska Gene Basic Vector and Pitska Gene Enhancer Vector (Toyo Ink Mfg. Co., Ltd.).
  • the TRE-containing DNA is inserted into a reporter gene of the above vector, for example, a multi-cloning site upstream of the luciferase gene, to obtain a TRE-reporter gene vector.
  • a test compound that suppresses the enzymatic activity of the repo overnight gene protein by the peptide of the present invention can be selected as a candidate substance having a competitive inhibition ability.
  • agonist screening is performed by contacting only the test compound with the TRE-repo allele-introduced gene-introduced protein-expressing cells of the present invention and measuring the increase in luminescence in the same manner as the peptide of the present invention. You can do it.
  • luciferase As a reporter gene.
  • Cells expressing the protein of the present invention into which the TRE-reporter gene (luciferase) has been introduced are seeded on a 24-well plate at 510 3 6 1 176 11 and cultured for 48 hours. After washing the cells with Hanks buffer (pH 7.4) containing 0.05% BSA and 20 mM HEPES, 10 nM of the peptide of the present invention or 10 nM of the peptide of the present invention and a test compound are added thereto, and the mixture is added at 37 ° C. And react for 60 minutes.
  • Hanks buffer pH 7.4
  • BSA Hanks buffer
  • 10 nM of the peptide of the present invention or 10 nM of the peptide of the present invention and a test compound are added thereto, and the mixture is added at 37 ° C. And react for 60 minutes.
  • Lyse the cells with a cell lysing agent for Pitka Gene (Toyo Ink Mfg. Co., Ltd.) and add a luminescent substrate (Toyo Ink Mfg. Co., Ltd.) to the lysate.
  • Luminescence from luciferase is measured using a luminometer, liquid scintillation counter or top counter. The amount of luminescence by luciferase when the peptide of the present invention is added and when ⁇ ⁇ the peptide of the present invention and a test compound are added are measured and compared.
  • the increase in intracellular calcium by the peptide of the present invention increases the amount of luminescence by luciferase.
  • a compound that suppresses this increase can be selected as a candidate substance capable of competitive inhibition.
  • Reporter genes include, for example, alkaline phosphatase, chloramphenic acid, acetylfurantransferrase ⁇ chloramphenicol
  • Genes such as acetyltransferase) and _galactosidase may be used.
  • the enzyme activities of these reporter gene proteins are measured according to a known method or using a commercially available measurement kit.
  • '' Alkali phosphatase activity was measured using, for example, Lumi-Phos 530 manufactured by Wako Pure Chemical, and chloramphenicol acetyltransferase was evaluated using, for example, FAST CAT chrola immediately manufactured by Wako Pure Chemical.
  • S-galactosidase activity is measured using, for example, Aurora Gat XE manufactured by Wako Pure Chemical Industries.
  • MAP kinase is activated and proliferates by stimulation of the peptide of the present invention.
  • this reaction to measure the stimulating activity of the peptide of the present invention on the cells expressing the protein of the present invention, it is possible to screen for a compound that alters the binding property between the peptide of the present invention and the protein of the present invention. .
  • the peptide of the present invention was brought into contact with a cell expressing the protein of the present invention.
  • the binding between the peptide of the present invention and the protein of the present invention can be determined. Screen for compounds that alter the.
  • Proliferation of the protein-expressing cells of the present invention may be measured, for example, by measuring MAP kinase activity, thymidine uptake activity, cell number, and the like.
  • MAP kinase activity after adding the peptide of the present invention or the peptide of the present invention and a test compound to the cells expressing the protein of the present invention, an anti-VIAP kinase antibody was used from a cell lysate. After obtaining the MAP kinase fraction by immunoprecipitation, the MAP kinase activity is measured using a known method, for example, MAP Kinase Assay II- [ 32 P] -ATP manufactured by Wako Pure Chemical Industries, and compared.
  • the protein-expressing cells of the present invention were seeded on a 24-well plate, cultured, added with the peptide of the present invention or the peptide of the present invention and a test compound, and then labeled with radioactivity.
  • thymidine e.g., [methy Bok 3 H] - thymidine down, etc.
  • the cells were lysed and the radioactivity of the thymidine incorporated into the cells, by counting in a liquid scintillator one Chillon counter, thymidylate ' Measure gin uptake activity and compare.
  • the protein-expressing cells of the present invention were seeded on a 24-well plate, cultured, and after adding the peptide of the present invention or the peptide of the present invention and a test compound, MTT (3- ( Add 4,5-dimethyl-2-thiazolyl) -2,5-diphenyl-2H-tetrazolium bromide).
  • MTT (3- ( Add 4,5-dimethyl-2-thiazolyl) -2,5-diphenyl-2H-tetrazolium bromide).
  • MTT formazan in which MTT has been changed by being taken into cells is dissolved in an aqueous solution of isopropanol acidified with hydrochloric acid, and then measured by absorption at 570 nm for comparison.
  • a test compound that suppresses the growth of the protein-expressing cell of the present invention can be selected as a candidate substance having a competitive inhibition ability.
  • agonists can be screened by bringing only the test compound into contact with the protein-expressing cell of the present invention and measuring the cell proliferation activity similar to that of the peptide of the present invention.
  • the evening protein-expressing cells of the present invention are seeded at 5,000 cells / well in a 24-well plate and cultured for one day. The cells are then starved in a serum-free medium for 2 days.
  • the peptide or peptide and a test compound of the present invention of the present invention after incubation was added to the cells 24 hours, [methy Bok 3 H] - thymidine was added Ueru per 0. 015MBq, incubation 6 hours. After washing the cells with PBS, add methanol and let stand for 10 minutes. Next, add 5% trichloroacetic acid and let stand for 15 minutes. Wash the fixed cells four times with distilled water. Lyse the cells with 0.3N sodium hydroxide solution and measure the radioactivity in the lysate with a liquid scintillation counter.
  • a test compound that suppresses an increase in radioactivity when the peptide of the present invention is added can be selected as a candidate substance capable of competitive inhibition.
  • the stimulation of the peptide of the present invention activates the potassium channel, and intracellular K ions flow out of the cell.
  • the stimulating activity of the peptide of the present invention on cells expressing the protein of the present invention is measured to screen for compounds that alter the binding between the peptide of the present invention and the protein of the present invention. be able to.
  • Rb ions rubberidium ions
  • Rb ions which are homologous to K ions, flow out of the cell through potassium channels without distinction from K ions.
  • the protein-expressing cells of the present invention a radioactive isotope R b After allowed incorporation of ([8 6 R b]) , the 8 6 R b flowing out by stimulation of the peptides of the present invention
  • the stimulating activity of the peptide of the present invention on the cells expressing the protein of the present invention is measured.
  • the peptide of the present invention when the peptide of the present invention is brought into contact with the protein-expressing cell of the present invention in the presence of 86 Rb, the peptide of the present invention and the test compound are brought into contact with the protein-expressing cell of the present invention. in the case of it is to measure the efflux activity of 8 6 R b, by comparing, for screening the compound that changes the binding property between the protein of the peptide and the invention of the present invention.
  • the 8 6 R b test compound to suppress an increase in efflux activity of by peptide stimulation of the present invention can be selected as a candidate substance capable of competitive inhibition.
  • only the test compound is brought into contact with the protein-expressing cell of the present invention, It is also possible to perform the disk re-learning of Agonisu Bok by measuring the increase in peptide similar 8 6 R b of efflux activity.
  • the protein-expressing cells of the present invention are seeded on a 24-well plate and cultured for 2 days. Thereafter, the cells are incubated for 2 hours in a medium containing 1 mCiZm1 and 86 RbC1. Wash cells thoroughly with medium to completely remove 86 RbC1 in the external solution.
  • the peptide of the present invention or the peptide of the present invention and a test compound are added to the cells, and after 30 minutes, the external solution is collected, and the radioactivity is measured and compared at a local time.
  • a test compound that suppresses an increase in the 86 Rb efflux activity due to peptide stimulation of the present invention can be selected as a candidate substance having a competitive inhibitory ability.
  • the extracellular pH change is measured using, for example, a Cytosensor device (Molecular Devices). ,
  • a test compound that suppresses the extracellular pH change caused by the peptide of the present invention can be selected as a candidate substance capable of competitive inhibition.
  • agonist screening can be performed by bringing only the test compound into contact with the protein-expressing cell of the present invention and measuring the extracellular pH change similar to that of the peptide of the present invention.
  • the protein-expressing cells of the present invention are cultured overnight in a capsule for a Cytosensor device, set in the chamber of the device, and set to 0.1% for about 2 hours until the extracellular pH is stabilized.
  • Perfuse 1 to 1640 medium (Molecular Devices) containing 83-8. After the pH is stabilized, a medium containing the peptide of the present invention or the peptide of the present invention and the test compound is perfused on the cells. Measure and compare the pH change of the medium caused by perfusion.
  • a compound that suppresses the extracellular pH change caused by the peptide of the present invention can be selected as a candidate substance having a competitive inhibition ability.
  • the sex pheromone receptor, ST e2, of the haploida-mating type (MAT) of yeast (Sacc aromyces cerevisiae) is conjugated to the G protein, Gpa1, and responds to the sex pheromone mating factor.
  • MAT haploida-mating type
  • Gpa1 G protein
  • Stel 2 induces the expression of various proteins (eg, FUS 1 involved in conjugation).
  • the control factor S st 2 functions in the above process in a suppressive manner.
  • a yeast into which a receptor gene has been introduced is produced, a signal transduction system in the yeast cell is activated by stimulation of the receptor gene, and the resulting proliferation is used as an index.
  • Attempts have been made to measure the reaction between a protein and a receptor (Trends in Biotechnology, 15, 487-494, 1997).
  • a compound capable of changing the binding property between the peptide of the present invention and the protein of the present invention can be screened.
  • the genes encoding Ste2 and Gpa1 of the MAT ⁇ yeast are removed, and instead, the protein gene of the present invention and a gene encoding the Gpa1-Gai2 fusion protein are introduced.
  • the gene encoding Far is removed to prevent ceU-cycle arrest from occurring, and the gene encoding Sst is removed to increase the sensitivity of the response to the peptide of the present invention.
  • the FUS1—HIS3 gene in which the histidine biosynthesis gene HIS3 is linked to FUS1 is introduced. This genetic recombination operation can be performed, for example, by replacing the somatostin-tin receptor-type-1 (SSTR2) gene with the protein of the present invention in the method described in Molecular and Cellular Biology, vol.
  • SSTR2 somatostin-tin receptor-type-1
  • the transformed yeast thus constructed reacts with high sensitivity to the peptide of the present invention, resulting in the activation of MAP kinase, the synthesis of histidine biosynthesis enzyme, and the histidine-deficient medium. It is possible to grow in.
  • the above-described protein-expressing yeast of the present invention (the Ste2 gene and the Gpa1 gene were removed, the protein gene of the present invention and the Gpa1-Gai2 fusion protein-encoding gene were introduced, and the Far gene was introduced.
  • S ⁇ yeast with the FUS1-1HIS3 gene removed and the Sst gene removed is cultured in a histidine-deficient medium, and the peptide of the present invention or the peptide of the present invention and a test compound are contacted with each other; By measuring and comparing the growth of the yeast, it is possible to screen for compounds that alter the binding between the peptide of the present invention and the protein of the present invention. In this method, a test compound that suppresses the growth of the yeast can be selected as a candidate substance having a competitive inhibition ability.
  • agonist screening can also be performed by bringing only the test compound into contact with the above-described yeast expressing the protein of the present invention and measuring the growth of yeast similar to the peptide of the present invention.
  • the above-described protein-expressing yeast of the present invention is cultured overnight in a liquid medium of a complete synthetic medium, and then added to a dissolved agar medium from which histidine has been removed to a concentration of 2 ⁇ 10 4 ce 11 / m 1. Then sow in a 9 x 9 cm square petri dish. After the agar solidifies, a sterile filter paper impregnated with the peptide of the present invention or the peptide of the present invention and the test compound is placed on the agar surface, and cultured at 30 ° C for 3 days. The effect of the test compound is to compare the growth of yeast around the filter paper with the use of sterile filter paper impregnated with only the peptide of the present invention. In addition, the peptide of the present invention is added to the agar medium from which histidine has been removed in advance, and the test compound is soaked in sterile filter paper to culture the yeast. You may observe that they are affected.
  • a compound that suppresses the growth of yeast can be selected as a candidate substance having a competitive inhibition ability.
  • RNA-introduced African Xenopus oocytes RNA-introduced African Xenopus oocytes
  • test compound of the present invention By measuring and comparing changes in cell membrane potential when brought into contact with the introduced Xenopus laevis oocytes, a compound that changes the binding property between the peptide of the present invention and the protein of the present invention is screened. I do.
  • a test compound that suppresses a change in cell membrane potential can be selected as a candidate substance having a competitive inhibition ability.
  • agonists can be screened by bringing only test compounds into contact with the protein gene of the present invention, RNA-introduced African oocytes, and measuring changes in cell membrane potential similar to those of the peptides of the present invention.
  • the protein gene mRN ⁇ of the present invention may be prepared from a tissue or a cell, or may be transcribed from a plasmid in vitro.
  • the protein gene mRNA of the present invention was cultured in MBS solution at 20 ° C for 3 days, and this was cultured in a voltage clamp equipped with Ringer solution. Insert the potential fixing glass microelectrode and the potential measurement glass microelectrode into the cell, and place the electrode outside the cell. When the potential is stabilized, a Ringer solution containing the peptide of the present invention or the peptide of the present invention and a test compound is passed, and the change in potential is recorded.
  • the effect of the test compound can be measured by comparing the change in the cell membrane potential of the oocytes of the present invention into which the protein gene of the present invention has been transfected with RNA, compared to the case where a Ringer solution containing only the peptide of the present invention is applied. .
  • a compound that suppresses cell membrane electrical changes can be selected as a candidate substance having a competitive inhibition ability.
  • the increase in the amount of change in the potential facilitates the measurement, and thus, the pO1A-added RNA of each G protein gene may be introduced.
  • polyA-added RNA of a gene of a protein eg, aequorin, etc.
  • a screening kit for a compound or a salt thereof that alters the binding property between the peptide of the present invention and the protein of the present invention includes a protein of the present invention, a cell containing the protein of the present invention, or a cell containing the protein of the present invention.
  • a membrane fraction, and a peptide of the present invention includes a protein of the present invention, a cell containing the protein of the present invention, or a cell containing the protein of the present invention.
  • screening kit of the present invention examples include the following. 1. Screening reagent
  • CH0 cells expressing the protein of the present invention were subcultured in 12-well plates at 5 ⁇ 10 5 Z-wells and cultured at 37 ° C., 5% CO 2 , 95% air for 2 days. .
  • the peptide of the present invention is dissolved to a concentration of lmM in PBS containing 0.1% ⁇ serum albumin (Sigma) and stored at -20 ° C.
  • test compound solution (ii) 10- 3 ⁇ 1 ( ) M 5 1 added, the peptide of the present invention labeled 5 1 was added to react at room temperature for one hour.
  • 5/1 of the peptide of the present invention is added instead of the test compound.
  • NSB Non-specific Binding ''
  • the compound or a salt thereof obtained by using the screening method or the screening kit of the present invention is a compound that changes the binding (inhibits or promotes the binding) between the peptide of the present invention and the protein of the present invention.
  • a compound having a cell stimulating activity via the protein of the present invention or a salt thereof a so-called agonist of the protein of the present invention
  • a compound not having the stimulating activity a so-called antagonist of the protein of the present invention.
  • Such compounds include peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, and the like. It may be a novel compound or a known compound.
  • the specific method for evaluating whether the above-mentioned protein of the present invention is an agonist or an antagonist may be according to the following ( ⁇ ) or ( ⁇ ).
  • the binding assay shown in the above screening method is performed to obtain a compound that changes the binding property (particularly, inhibits the binding) between the peptide of the present invention and the protein of the present invention. Is determined whether it has the above-described cell stimulating activity via the protein of the present invention.
  • a compound having a cell stimulating activity or a salt thereof is an agonist of the protein of the present invention, and a compound or a salt thereof having no such activity is an agonist of the protein of the present invention.
  • test compound is brought into contact with a cell containing the protein of the present invention, and the cell stimulating activity mediated by the protein of the present invention is measured.
  • the compound having a cell stimulating activity or a salt thereof is an agonist of the protein of the present invention.
  • a compound that activates the protein of the present invention for example, a peptide of the present invention or an agonist of the protein of the present invention
  • a cell containing the protein of the present invention The cell stimulating activity mediated by the protein of the present invention when the compound to be converted and the test compound are brought into contact with the cells containing the protein of the present invention is measured and compared.
  • the compound capable of decreasing the cell stimulating activity of the compound activating the protein of the present invention or a salt thereof is an antagonist of the protein of the present invention.
  • the agonist of the protein of the present invention has the same activity as the physiological activity of the peptide of the present invention on the protein of the present invention, it is useful as a safe and low-toxic drug like the peptide of the present invention. It is.
  • the protein antagonist of the present invention can suppress the physiological activity of the peptide of the present invention on the protein of the present invention, and is therefore useful as a safe and low-toxic drug for suppressing the receptor activity. is there.
  • the protein of the present invention is involved in an appetite (feeding) promoting action and an oxytocin secretion promoting action. Therefore, among the compounds obtained using the above-mentioned screening method or screening kit, agonists of the protein of the present invention include, for example, an appetite (feeding) enhancer, anorexia (eg, anorexia nervosa), Appetite It can be used as a medicament for the prevention and treatment of anemia or hypoproteinemia due to tremor, weak labor, laxative bleeding, uterine remodeling failure, and milk stasis.
  • the protein antagonists of the present invention include, for example, obesity (eg, malignant mastocytosis), exogenous obesity, hyperinsulin inar obesity, hyperplasma Hyperplasmic obesity ⁇ hypophyseal adiposity, hypoplasmic obesity> hypothyroid obesity, hypothalamic obesity, hypothalamic obesity, symptoms Obesity (sy immediate tomatic obesity), childhood obesity (infantile obesity), upper body obesity, alimentary obesity, hypogonadal obesity, systemic mastocytosis
  • central obesity central obesity
  • hyperphagia affective disorders, sexual dysfunction, overwork, tonic contractions, fetal asphyxia, uterine rupture, tubal lacerations, preterm birth, Prader-Willi syndrome, glucoseuria and Its complications (eg, diabetic nephropathy, diabetic retinopathy, diabetic neuropathy, etc.), hypertension, hyperlipidemia, coronary atherosclerosis, gout, respiratory disease (Pickwick syndrome, sleep apnea syndrome) It can be used as a medicine for prophylactic and therapeutic agents such as fatty liver, infertility, osteoarthritis, etc. (especially antiobesity agents, appetite (feeding) regulators, etc.).
  • a pharmaceutically acceptable salt or the like is used as a salt of the compound obtained by using the above-described screening method or screening kit.
  • a pharmaceutically acceptable salt or the like examples thereof include salts with inorganic bases, salts with organic bases, salts with inorganic acids, salts with organic acids, and salts with basic or acidic amino acids.
  • the salt with an inorganic base include an alkali metal salt such as a sodium salt and a potassium salt, an alkaline earth metal salt such as a calcium salt and a magnesium salt, and an aluminum salt and an ammonium salt.
  • Preferred examples of the salt with an organic base include, for example, trimethylamine, triethylamine, pyridine, picoline, 2,6-alutidine, ethanolamine, genoaluminamine, triethanolamine, cyclohexylamine, dicyclohexane.
  • Examples include salts with xylamine, N, N, and dibenzylethylenediamine.
  • Preferred examples of the salt with an inorganic acid include salts with hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, and the like.
  • Suitable examples of salts with organic acids include, for example, formic acid, acetic acid, propionic acid, fumaric acid, oxalic acid, tartaric acid, maleic acid, citric acid, succinic acid, malic acid, methanesulfonic acid, benzenesulfonic acid, benzoic acid And the like.
  • salts with a basic amino acid include salts with, for example, arginine, lysine, orcinus
  • salts with an acidic amino acid include, for example, salts with aspartic acid, glutamic acid, etc. Is raised.
  • a compound or a salt thereof obtained by using the screening method or the screening kit of the present invention can be carried out according to a conventional method.
  • It can be used parenterally in the form of injections, such as aqueous solutions or suspensions.
  • a physiologically acceptable carrier, flavoring agent, excipient, vehicle, preservative, stabilizer, binder and the like in a generally accepted unit dosage form.
  • the amount of the active ingredient in these preparations is such that a suitable dosage in the specified range can be obtained.
  • Additives that can be incorporated into tablets, capsules, etc. include, for example, binders such as gelatin, corn starch, tragacanth gum, gum arabic, excipients such as crystalline cellulose, corn starch, gelatin, alginic acid, etc.
  • binders such as gelatin, corn starch, tragacanth gum, gum arabic
  • excipients such as crystalline cellulose, corn starch, gelatin, alginic acid, etc.
  • a leavening agent such as magnesium stearate
  • a sweetening agent such as sucrose, lactose or saccharin
  • a flavoring agent such as peppermint, cocoa oil or cellulose
  • a liquid carrier such as oils and fats can be further contained in the above-mentioned type of material.
  • Sterile compositions for injection include active substances in vehicles such as water for injection, natural substances such as sesame oil, coconut oil and the like. It can be formulated according to the usual formulation practice such as dissolv
  • Aqueous liquids for injection include, for example, physiological saline, isotonic solutions containing glucose and other adjuvants (eg, D-sorbitol, D-mannitol, sodium chloride, etc.).
  • Agents such as alcohols (eg, ethanol), polyalcohols (eg, propylene glycol, polyethylene glycol), nonionic surfactants (eg, Polysorbate 80 TM, HCO-50) and the like may be used in combination.
  • examples of the oily liquid include sesame oil and soybean oil, which may be used in combination with a dissolution aid such as benzyl benzoate or benzyl alcohol.
  • buffers eg, phosphate buffer, sodium acetate buffer
  • the prepared injection solution is usually filled in a suitable ampoule.
  • the preparations obtained in this way are safe and low toxic, and can be used, for example, in mammals (eg, humans, mice, rats, guinea pigs, egrets, sheep, sheep, bushus, dogs, cats, dogs, monkeys, chimpanzees, etc. ) Can be administered.
  • mammals eg, humans, mice, rats, guinea pigs, egrets, sheep, sheep, bushus, dogs, cats, dogs, monkeys, chimpanzees, etc.
  • the dose of a compound or a salt thereof obtained by using the screening method or the screening kit of the present invention varies depending on symptoms and the like.
  • the single dose varies depending on the administration subject, target organ, symptoms, administration method, etc.
  • injection into adult obese patients with a body weight of 60 kg
  • the dose can be administered in terms of 60 kg.
  • the antibody of the present invention can specifically recognize the protein of the present invention, It can be used for quantification of the protein of the present invention in a test solution, particularly for quantification by sandwich immunoassay.
  • the present invention provides a method for quantifying the protein of the present invention in a test solution, characterized in that in the quantification method of the above (ii), one of the antibodies is an antibody that recognizes the N-terminal of the protein of the present invention, and the other is It is desirable that the above-mentioned antibody reacts with the C-terminal of the protein of the present invention.
  • the protein of the present invention can be quantified using a monoclonal antibody against the protein of the present invention (hereinafter sometimes referred to as the monoclonal antibody of the present invention), and can also be detected by tissue staining or the like.
  • the anti-body molecule itself may be used, or the F (ab,) 2 , Fab ′ or Fab fraction of the antibody molecule may be used.
  • the method for quantifying the protein of the present invention using the antibody of the present invention is not particularly limited, and may be an antibody, an antigen, or an antibody-antigen complex corresponding to the amount of antigen (eg, the amount of protein) in the test solution. Any measurement method may be used as long as the amount of the body is detected by chemical or physical means, and this is calculated from a standard curve prepared using a standard solution containing a known amount of antigen. . For example, nephrometry, competition method, immunometric method and sandwich method are preferably used, but it is particularly preferable to use the sandwich method described later in terms of sensitivity and specificity.
  • radioactive isotopes elemental eg, [125 1], [131 1], [3 ⁇ 4], [ '4 C], [32 P], [33 P], [ 35s ], etc.
  • fluorescent substances eg, cyanine fluorescent dyes (eg, Cy2, Cy3, Cy5, Cy5.5, Cy7 (manufactured by Amersham Biosciences), etc.), fluorescamine, fluorescenisochi Etc.
  • enzymes eg, -galactosidase,) 3-darcosidase, Lucariphosphatase, passoxidase, malate dehydrogenase, etc.
  • luminescent substances eg, luminol, luminol derivatives, luciferin, lucigenin, etc.
  • a biotin-avidin system can be used for binding the antibody or antigen to the labeling agent.
  • the carrier include insoluble polysaccharides such as agarose, dextran, and cellulose; synthetic resins such as polystyrene, polyacrylamide, and silicon; and glass.
  • a test solution is reacted with the insolubilized monoclonal antibody of the present invention (primary reaction), and further reacted with another labeled monoclonal antibody of the present invention (secondary reaction).
  • primary reaction the insolubilized monoclonal antibody of the present invention
  • secondary reaction another labeled monoclonal antibody of the present invention
  • the primary reaction and the secondary reaction may be performed in the reverse order, may be performed simultaneously, or may be performed at staggered times.
  • the labeling agent and the method of insolubilization can be in accordance with those described above.
  • the antibody used for the solid phase antibody or the labeling antibody does not necessarily need to be one kind, and a mixture of two or more kinds of antibodies is used for the purpose of improving measurement sensitivity and the like. May be used.
  • the monoclonal antibody of the present invention used in the primary reaction and the secondary reaction is preferably an antibody having a different site to which the protein of the present invention binds.
  • the antibody used in the primary reaction and the secondary reaction is, for example, when the antibody used in the secondary reaction recognizes the C-terminal of the protein of the present invention, the antibody used in the primary reaction is Preferably, an antibody that recognizes other than the C-terminal, for example, the N-terminal, is used.
  • the monoclonal antibody of the present invention can be used in a measurement system other than the sandwich method, for example, a competition method, an immunometric method or a nephelometry.
  • a competition method the antigen in the test solution and the labeled antigen are used for the antibody.
  • the unreacted labeled antigen (F) and the labeled antigen (B) bound to the antibody are separated (BZF separation), and the amount of labeling of either B or F is measured. Quantify the amount of antigen in the test solution You.
  • a soluble antibody was used as the antibody
  • B / F separation was performed using polyethylene glycol
  • a liquid phase method using a second antibody against the antibody was used as the first antibody.
  • a solid phase antibody was used as the first antibody.
  • an immobilization method using a soluble first antibody and an immobilized antibody as the second antibody is used.
  • the antigen in the test solution and the immobilized antigen are subjected to a competitive reaction with a certain amount of labeled antibody, and then the solid phase and the liquid phase are separated.
  • the antigen is allowed to react with an excess amount of the labeled antibody, then the immobilized antigen is added, and the unreacted labeled antibody is bound to the solid phase, and then the solid phase and the liquid phase are separated.
  • the amount of label in either phase is measured to determine the amount of antigen in the test solution.
  • nephrometry the amount of insoluble sediment resulting from an antigen-antibody reaction in a gel or in a solution is measured. Even when the amount of antigen in the test solution is small and only a small amount of sediment is obtained, laser-nephrometry using laser-dispersion is preferably used.
  • the protein measuring system of the present invention may be constructed by adding ordinary technical considerations of those skilled in the art to ordinary conditions and operation methods in each method. For details of these general technical means, reference can be made to reviews, documents, etc.
  • the protein of the present invention can be quantified with high sensitivity by using the antibody of the present invention.
  • anorexia eg, anorexia nervosa, etc.
  • Anemia or hypoproteinemia due to anorexia, weak labor, lax hemorrhage, uterine remodeling failure, milk stasis, etc. can be diagnosed as having a high possibility of developing.
  • an increase in the concentration of the protein of the present invention is detected, for example, obesity (eg, vein mastocytosis, exogenous obesity, hyperinsulinic obesity, hyperplasmic obesity, pituitary) Obesity, reduced plasma obesity, hypothyroid obesity, hypothalamic obesity, symptomatic obesity, pediatric obesity, upper body obesity, dietary obesity, hypogonadism, systemic mastocytosis, simple obesity , Central obesity, etc.), hyperphagia, affective disorders, sexual dysfunction, excessive labor, tonic contractions, fetal asphyxia, uterine rupture, facial lacerations, preterm birth, Prader-Willi syndrome, diabetes and its complications Disease (eg, diabetic nephropathy, diabetic retinopathy, diabetic neuropathy, etc.), hypertension, hyperlipidemia, coronary atherosclerosis, gout, respiratory disease (Pickck syndrome, sleep apnea syndrome), Fatty liver, infertility It can be diagnosed that the possibility of osteoarthritis and the
  • the antibody of the present invention can be used for detecting the protein of the present invention present in a subject such as a body fluid or a tissue.
  • a subject such as a body fluid or a tissue.
  • the detection of the protein of the present invention in each fraction during purification, and the analysis of the behavior of the protein of the present invention in test cells, etc. Can be used.
  • the DNA of the present invention can be used, for example, in humans or warm-blooded animals (e.g., rats, mice, guinea pigs, egrets, birds, higgies, bushes, horses, cats, cats, dogs, Abnormalities (genetic abnormalities) in the DNA or mRNA encoding the protein of the present invention or a partial peptide thereof in monkeys, chimpanzees, etc.). It is useful as a gene diagnostic agent for mutation or decrease in expression, and increase or excessive expression of the DNA or mRNA.
  • warm-blooded animals e.g., rats, mice, guinea pigs, egrets, birds, higgies, bushes, horses, cats, cats, dogs, Abnormalities (genetic abnormalities) in the DNA or mRNA encoding the protein of the present invention or a partial peptide thereof in monkeys, chimpanzees, etc.
  • the above-described genetic diagnosis using the DNA of the present invention can be performed, for example, by the well-known Northern Hybridization ⁇ PCR-SSCP method (Genomics, Vol. 5, pp. 874-879 (1989)) Proceedings of the National Academy of Sciences of The United States of America, Vol. 86, pp. 2766-2770 (1989)).
  • obesity eg, malignant mastocytosis, exogenous obesity, hyperinsulinic obesity, hyperplasmic obesity, pituitary obesity, hypoplasma
  • Obesity hypothyroid obesity, hypothalamic obesity, symptomatic obesity, childhood obesity, upper body obesity, dietary obesity, hypogonadism obesity, systemic mastocytosis, simple obesity, centrality Obesity, etc.
  • hyperphagia affective disorders, sexual dysfunction, excessive labor, tonic contractions, fetal asphyxia, uterine rupture, tubal lacerations, preterm birth, Prader-Willi syndrome, diabetes and its complications ( Eg, diabetic nephropathy, diabetic retinopathy, diabetic neuropathy), hypertension, hyperlipidemia, coronary atherosclerosis, gout, respiratory disease (Pickwick syndrome, sleep apnea syndrome), fat Liver, It can be diagnosed that the possibility of infertility or osteoarthritis is high.
  • anorexia eg, anorexia nervosa
  • anorexia associated with anorexia It can be diagnosed as likely to be low protein, weak labor, lax hemorrhage, uterine remodeling failure, and milk stasis.
  • the antisense polynucleotide of the present invention which complementarily binds to the DNA of the present invention and can suppress the expression of the DNA, has low toxicity, and functions of the protein of the present invention or the DNA of the present invention in vivo (eg, Since it can suppress eating (appetite) promoting activity, for example, obesity (eg, malignant mastocytosis, exogenous obesity, hyperinsulinic obesity, hyperplasmic obesity, pituitary obesity) , Hypoplasmic obesity, hypothyroid obesity, hypothalamic obesity, symptomatic obesity, childhood obesity, upper body obesity, dietary obesity, hypogonadism, systemic mastocytosis, simple obesity , Central obesity), Hyperphagia, affective disorders, sexual dysfunction, overwork labor, tonic contractions, fetal asphyxia, ruptured pupils, facial tears, premature birth, Prader-Willi syndrome, diabetes and its complications (eg, diabetic Nephropathy, diabetic retinopathy, diabetic neuropathy, etc.), hypertension,
  • the above-mentioned antisense polynucleotide When used as the above-mentioned prophylactic / therapeutic agent, it can be formulated and administered according to a known method.
  • the antisense polynucleotide when used, the antisense polynucleotide is inserted alone or into a suitable vector such as a retrovirus vector, an adenovirus vector, an adenovirus associated virus vector, and the like. It can be administered orally or parenterally to humans or mammals (eg, rats, puppies, sheep, pigs, puppies, cats, dogs, monkeys, etc.).
  • the antisense polynucleotide can be administered as it is or in the form of a formulation together with a physiologically acceptable carrier such as an auxiliary agent for promoting uptake, and can be administered by a gene gun or a catheter such as a hydrogel catheter.
  • the dosage of the antisense polynucleotide varies depending on the target disease, the administration subject, the administration route, and the like.For example, when the antisense polynucleotide of the present invention is intravenously administered for the purpose of treating obesity, Generally, in an adult (body weight 60 kg), about 0.1 to 100 mg of the antisense polynucleotide is administered per day.
  • antisense polynucleotide can also be used as a diagnostic oligonucleotide probe for examining the presence or expression of the DNA of the present invention in tissues or cells.
  • a medicament comprising the lipozyme
  • an expression vector or the like containing the gene (DNA) encoding the lipozyme is provided.
  • double-stranded RNA, lipozyme and the like can disrupt RNA transcribed from the DNA of the present invention or suppress the function thereof, and the protein or the same of the present invention in vivo.
  • Obstructions for example, obesity (eg, malignant mastocytosis, exogenous obesity, hyperinsulinic obesity, hyperplasmic obesity, pituitary obesity, Plasma obesity, hypothyroid obesity, hypothalamic obesity, symptomatic obesity, pediatric obesity, upper body obesity, dietary obesity, hypogonadism, systemic mastocytosis, simple obesity, centrality Obesity, etc.), hyperphagia, affective disorders, sexual dysfunction, overwork labor, tonic contractions, fetal asphyxia, ruptured uterus, tubal lacerations, preterm birth, Prader-Willi syndrome, diabetes and its complications (Eg, diabetic nephropathy, diabetic retinopathy, diabetic neuropathy, etc.), hypertension, hyperlipidemia, coronary arteriosclerosis, gout, respiratory disease (Pickwick syndrome, sleep apnea syndrome group) ), Fatty liver, infertility, osteoarthritis, etc. (especially anti-obesity agents, appetite (especially anti-obe
  • the double-stranded RNA can be designed and produced based on the sequence of the polynucleotide of the present invention according to a known method (eg, Nature, 411, 494, 2001).
  • the lipozyme can be designed and manufactured based on the sequence of the polynucleotide of the present invention according to a known method (eg, TRENDS in Molecular Medicine, Vol. 7, pp. 221, 2001). For example, it can be produced by substituting a part of a known lipozyme sequence with a part of RNA encoding the protein of the present invention.
  • a consensus sequence NU X (where N represents all bases, X represents a base other than G) which can be cleaved by a known lipozyme, And the like.
  • RNA or lipozyme When the above-mentioned double-stranded RNA or lipozyme is used as the above-mentioned prophylactic / therapeutic agent, it can be formulated and administered in the same manner as the antisense polynucleotide.
  • the expression vector (V) is used in the same manner as known gene therapy methods and the like, and is used as the above-mentioned prophylactic / therapeutic agent.
  • the antibody of the present invention having the action of neutralizing the activity of the protein of the present invention can suppress the function of the protein of the present invention (eg, inactivate signal transduction), for example, obesity (eg, obesity) Malignant mastocytosis, exogenous obesity, hyperinsulinic obesity, hyperplasmic obesity, pituitary obesity, hypoplasmic obesity, hypothyroid obesity, hypothalamic obesity, symptomatic obesity, childhood obesity , Upper body obesity, dietary obesity, hypogonadism, obesity, systemic mastocytosis, simple obesity, central obesity, etc.), hyperphagia, affective disorder, sexual dysfunction, labour, tonicity Uterine contractions, fetal asphyxia, uterine rupture, ruptured tubules, preterm birth, Prader-Willi syndrome, diabetes and its complications (eg, diabetic nephropathy, diabetic retinopathy, diabetic neuropathy, etc.), hypertension, Hyperlipidemia, coronary Pulse sclerosis, gout, respiratory disease (Pickwick syndrome
  • the antibody of the present invention having an activity of activating the activity of the protein of the present invention can promote the function of the protein of the present invention (eg, activate signal transduction), for example, increase appetite (feeding) Drugs, anorexia (eg, anorexia nervosa), anemia or hypoproteinemia associated with anorexia, weak labor, lax bleeding, uterine remodeling failure, milk stasis, etc. it can.
  • anorexia eg, anorexia nervosa
  • anemia or hypoproteinemia associated with anorexia weak labor, lax bleeding, uterine remodeling failure, milk stasis, etc. it can.
  • the antibodies of the present invention can be administered by themselves or as a suitable pharmaceutical composition.
  • the pharmaceutical composition used for the administration contains the antibody or a salt thereof and a pharmacologically acceptable carrier, diluent or excipient.
  • Such compositions are provided in dosage forms suitable for oral or parenteral administration.
  • compositions for oral administration include solid or liquid dosage forms, specifically tablets (including sugar-coated tablets and film-coated tablets), pills, granules, powders, capsules ( Soft capsules), syrups, emulsions, suspensions and the like.
  • Such a composition is produced by a known method and contains a carrier, diluent or excipient usually used in the pharmaceutical field.
  • a carrier for example, lactose, starch, sucrose, magnesium stearate and the like are used as carriers and excipients for tablets.
  • compositions for parenteral administration for example, injections, suppositories, etc. are used.
  • Injections are in the form of intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections, infusions, etc. Is included. Such injections are prepared according to known methods, for example, by dissolving, suspending or emulsifying the antibody or a salt thereof in a sterile aqueous or oily liquid commonly used for injections.
  • Aqueous liquids for injection include, for example, saline, isotonic solutions containing glucose and other adjuvants, and suitable solubilizing agents, such as alcohol (eg, ethanol) and polyalcohol (eg, , Propylene glycol, polyethylene glycol), nonionic surfactants (eg, polysorbate 80, HCO-50 (polyoxythylene (50 mol) adduc tof hydrogenated cas tor oil)), etc. Good.
  • alcohol eg, ethanol
  • polyalcohol eg, Propylene glycol, polyethylene glycol
  • nonionic surfactants eg, polysorbate 80, HCO-50 (polyoxythylene (50 mol) adduc tof hydrogenated cas tor oil)
  • oily liquid for example, sesame oil, soybean oil, and the like are used, and benzyl benzoate, benzyl alcohol, and the like may be used as a solubilizing agent.
  • the prepared injection is usually filled
  • each dosage unit dosage form is 5 to 500 mg, especially for injections.
  • 5 to 100 mg, and other dosage forms contain 10 to 25 Omg of the antibody.
  • compositions may contain other active ingredients as long as no undesirable interaction occurs due to the combination with the above antibody.
  • the prophylactic / therapeutic agent for the above-mentioned diseases containing the antibody of the present invention has low toxicity and can be used as it is as a liquid or as a pharmaceutical composition of an appropriate dosage form, in humans or mammals (eg, rat, egret, sheep, etc.). Can be administered orally or parenterally (eg, intravenously) to mice, dogs, cats, cats, dogs, monkeys, etc.). The dosage varies depending on the administration subject, target disease, symptoms, administration route, and the like.
  • 0.01 to 20 mg / kg body weight preferably 0.1 to 10 mg / kg body weight, more preferably 0.1 to 5 mg / kg body weight, about 1 to 5 times a day, preferably 1 to 5 times a day About 3 times as injection It is convenient to give. In the case of other parenteral administration and oral administration, an equivalent amount can be administered. If the symptoms are particularly severe, the dose may be increased accordingly.
  • the antibody of the present invention is also useful, for example, as a diagnostic agent for the above-mentioned diseases and the like.
  • the present invention has a DNA encoding the exogenous protein of the present invention (hereinafter abbreviated as the exogenous DNA of the present invention) or a mutant DNA thereof (sometimes abbreviated as the exogenous mutant DNA of the present invention).
  • the exogenous DNA of the present invention or a mutant DNA thereof (sometimes abbreviated as the exogenous mutant DNA of the present invention).
  • a non-human mammal is provided.
  • Non-human mammals having the exogenous DNA of the present invention or the mutant DNA thereof include unfertilized eggs, fertilized eggs, germ cells including spermatozoa and their progenitor cells, and the like.
  • the calcium phosphate method, the electric pulse method, the Lipofection It can be produced by introducing the target DNA by a method such as a coagulation method, a coagulation method, a microinjection method, a particle gun method, or a DEAE-dextran method.
  • the exogenous DNA of the present invention intended for somatic cells, organs of living organisms, tissue cells, and the like can be introduced and used for cell culture, tissue culture, and the like.
  • the DNA-introduced animal of the present invention can also be produced by fusing the cells with the above-mentioned germ cells by a known cell fusion method.
  • mice for example, pure strains such as C57B LZ6 strain and DBA2 strain, and hybrid strains such as BSC SFi strain, BDFi strain, B6D2 Ft strain, BALBZc strain, ICR strain, etc.
  • light eg, Wistar, SD, etc.
  • Examples of the “mammal” in the recombinant vector that can be expressed in mammals include humans and the like in addition to the above-mentioned non-human mammals.
  • exogenous DNA of the present invention does not mean the DNA of the present invention originally possessed by a non-human mammal but refers to the DNA of the present invention once isolated and extracted from the mammal.
  • mutant DNA of the present invention DNA having a mutation (for example, mutation) in the base sequence of the original DNA of the present invention, specifically, addition, deletion, or substitution of another base to another base DNA that has been used is used, and also includes abnormal DNA.
  • mutation for example, mutation
  • the abnormal DNA means a DNA that expresses an abnormal protein of the present invention.
  • a DNA that expresses a protein that suppresses the function of the normal protein of the present invention is used.
  • the exogenous DNA of the present invention may be derived from a mammal of the same species or a different species as the target animal.
  • a promoter capable of being expressed in animal cells e.g, egret, dog, cat, guinea pig, hamster, rat, mouse, etc.
  • the DNA construct of the present invention eg, a vector
  • a target mammal eg, a mouse fertilized egg
  • various promoters capable of expressing the DNA of the present invention downstream of various promoters capable of expressing the DNA of the present invention.
  • a DNA-introduced mammal that highly expresses the DNA of the present invention can be produced.
  • Examples of the expression vector of the protein of the present invention include a plasmid derived from Escherichia coli, a plasmid derived from Bacillus subtilis, a plasmid derived from yeast, a bacteriophage such as ⁇ phage, a retrovirus such as Moroni leukemia virus, a vaccinia virus and the like.
  • animal viruses such as baculovirus are used.
  • plasmids derived from Escherichia coli, plasmids derived from Bacillus subtilis or plasmids derived from yeast are preferred. It is used well. ',
  • promoters that regulate the DNA expression include, for example, (i) DNA promoters derived from viruses (eg, Simian virus, cytomegalovirus, Moroni leukemia virus, JC virus, breast cancer virus, poliovirus, etc.) Yuichi, (ii) Promoters derived from various mammals (humans, egrets, dogs, cats, guinea pigs, mussels, rats, mice, etc.), such as albumin, insulin II, ⁇ roplakin II, Erasu evening Protease, erythropoietin, endothelin, muscle creatine kinase, glial fibrillary acidic protein, dalyuthione S-transferase, platelet-derived growth factor 13, keratin Kl, 10 and 14, collagen I And II, cyclic AMP-dependent protein kinase / 3I subunit, dystrophy Int, tartrate-resistant alkaline phosphatase, atrial sodium diuretic factor, end
  • the vector preferably has a sequence that terminates the transcription of the target mRNA in the DNA-transfected mammal (generally referred to as "Yuichi Mineta-1").
  • Yuichi Mineta-1 the DNA-transfected mammal
  • the sequence of each DNA can be used.
  • Simian virus SV40 or the like is used.
  • the splicing of each DNA for the purpose of further expressing the desired foreign DNA It is also possible to link a licensing signal, an enhancer region, a part of an intron of a eukaryotic DNA, etc., at 5, upstream of the promoter region, between the promoter region and the translation region, or 3 'downstream of the translation region.
  • the normal translation region of the protein of the present invention is DNA derived from liver, kidney, thyroid cells, fibroblasts derived from humans or various mammals (eg, egrets, dogs, cats, guinea pigs, hamsters, rats, mice, etc.). And all or part of genomic DNA from various commercially available genomic DNA libraries, or complementary DNA prepared by known methods from liver, kidney, thyroid cells, and fibroblast-derived RNA as raw materials I can do it.
  • an exogenous abnormal DNA can produce a translation region obtained by mutating a normal polypeptide translation region obtained from the above cells or tissues by a point mutagenesis method.
  • the translation region is a DNA construct that can be expressed in an introduced animal, which is usually ligated to the downstream of the promoter and optionally the upstream of a transcription termination site.
  • the exogenous DNA of the present invention at the fertilized egg cell stage is ensured to be present in all germ cells and somatic cells of the target mammal.
  • the presence of the exogenous DNA of the present invention in the germinal cells of the transgenic animal after the DNA transfer means that all the progeny of the transgenic animal retain the exogenous DNA of the present invention in all of the germ cells and somatic cells Means that.
  • the offspring of this type of animal that has inherited the exogenous DNA of the present invention have the exogenous DNA of the present invention in all of its germ cells and somatic cells.
  • a non-human mammal into which the exogenous normal DNA of the present invention has been introduced can be subcultured in a normal breeding environment as an animal having the DNA after confirming that the exogenous DNA is stably retained by the crossing. .
  • exogenous DNA of the present invention is provided to be present in excess in all germ cells and somatic cells of the target mammal.
  • Excessive presence of the exogenous DNA of the present invention in the germinal cells of the produced animal after the introduction of the DNA indicates that all the offspring of the produced animal carry the exogenous DNA of the present invention in all of its germ cells and somatic cells. It means every time you have it.
  • the progeny of this type of animal that has inherited the exogenous DNA of the present invention has an excess of the exogenous DNA of the present invention in all of its germinal and somatic cells.
  • the non-human mammal having the normal DNA of the present invention expresses the normal DNA of the present invention at a high level, and finally promotes the function of the endogenous normal DNA, thereby finally obtaining the protein of the present invention. May develop functional hyperplasia, and can be used as a disease model animal. For example, using the normal DNA-introduced animal of the present invention to elucidate the pathological mechanism of hyperactivity of the protein of the present invention and diseases associated with the protein of the present invention, and to examine a method for treating these diseases. Is possible.
  • a non-human mammal having the exogenous abnormal DNA of the present invention can be subcultured in a normal breeding environment as an animal having the DNA after confirming that the exogenous DNA is stably maintained by mating. I can do it.
  • the desired exogenous DNA can be incorporated into the above-mentioned plasmid and used as a raw material.
  • the DNA construct with the promoter can be prepared by ordinary DNA engineering techniques. Introduction of the abnormal DNA of the present invention at the fertilized egg cell stage is ensured to be present in all germ cells and somatic cells of the target mammal.
  • the presence of the abnormal DNA of the present invention in the germinal cells of the animal produced after the transfer of DNA means that all the offspring of the animal produced have the abnormal DNA of the present invention in all of the germ cells and somatic cells.
  • the progeny of this type of animal that has inherited the exogenous DNA of the present invention has the abnormal DNA of the present invention in all of its germ cells and somatic cells. 'A homozygous animal having the introduced DNA on both homologous chromosomes is obtained, and by crossing these male and female animals, it is possible to propagate the offspring so that all offspring have the DNA.
  • the non-human mammal having the abnormal DNA of the present invention expresses the abnormal DNA of the present invention at a high level, and finally inhibits the function of endogenous normal DNA, thereby ultimately obtaining the protein of the present invention. In some cases, it becomes a functional inactive refractory disease and can be used as a model animal for the disease. For example, using the abnormal DNA-introduced animal of the present invention, It is possible to elucidate the pathological mechanism of the function-inactive refractory of the protein of the present invention and to examine a method for treating this disease.
  • the abnormal DNA-highly expressing animal of the present invention can be used to inhibit the function of a normal protein by the abnormal protein of the present invention in function-inactive refractory disease of the protein of the present invention (dominant negatase). ive effect).
  • the mammal into which the foreign abnormal DNA of the present invention has been introduced has an increased symptom of the released protein of the present invention, it can be used in a therapeutic drug screening test for the protein of the present invention or its functionally inactive refractory disease. Is also available.
  • cells of tissues having DNA are cultured by standard tissue culture techniques, and these are used to study the function of cells from generally difficult tissues,
  • a therapeutic agent for a disease associated with the protein of the present invention including a functionally inactive refractory type of the protein of the present invention
  • using the DNA-transfected animal of the present invention Using a quantitative method or the like, it is possible to provide an effective and rapid screening method for the therapeutic agent for the disease.
  • using the animal into which the DNA of the present invention has been introduced or the exogenous DNA expression vector of the present invention it is possible to study and develop a method for treating DNA associated with the protein of the present invention.
  • bases, amino acids, and the like are represented by abbreviations based on the respective abbreviations by the I UPAC- IUB Commission on Biochemical Nomenclature or commonly used abbreviations in the art, and examples thereof are described below.
  • the L-form is indicated unless otherwise specified.
  • a 1 a Alanine
  • Th r Threonine
  • 1 shows the nucleotide sequence of cDNA encoding the cynomolgus SLT receptor.
  • Fig. 3 shows the amino acid sequence of the cynomolgus monkey SLT receptor.
  • [SEQ ID NO: 5] 2 shows the nucleotide sequence of primer 11 used in the PCR reaction in Example 2 below.
  • FIG. 3 shows the nucleotide sequence of cDNA encoding the C-terminal region of the feline SLT receptor obtained in Example 2 below.
  • Example 3 shows the nucleotide sequence of cDNA encoding the N-terminal region of the feline SLT receptor obtained in Example 3 below.
  • 1 shows the nucleotide sequence of a cDNA encoding a cat SLT receptor.
  • Example 5 shows the nucleotide sequence of cDNA encoding the C-terminal region of canine SLT receptor obtained in Example 5 below.
  • [SEQ ID NO: 18] 7 shows the nucleotide sequence of primer 11 used in the PCR reaction in Example 6 below. [SEQ ID NO: 19]
  • Example 7 shows the nucleotide sequence of cDNA encoding the N-terminal region of canine SLT receptor obtained in Example 6 below.
  • 1 shows the nucleotide sequence of cDNA encoding canine SLT receptor.
  • 1 shows the amino acid sequence of MCH. .
  • Example 1 shows the nucleotide sequence of cDNA which encodes the entire length of the cynomolgus monkey SLT receptor obtained in Example 1 below.
  • Example 7 shows the nucleotide sequence of cDNA encoding the full length of cat SLT receptor obtained in Example 4 below.
  • Example 7 shows the nucleotide sequence of cDNA encoding the entire length of the canine SLT receptor obtained in Example 7 below.
  • Escherichia col i T0P10 / pCR4_monSLT obtained in Example 1 described below is an independent administrative corporation of Chuo No. 6 (Zip code 305-8566) 1-1 Toto, Tsukuba-shi, Ibaraki, Japan from June 4, 2002. Accession No. FERM BP-8066 to National Institute of Advanced Industrial Science and Technology Patent Organism Depositary Has been deposited as
  • Escherichia coli TOPlO / pcDNA-catSLT obtained in Example 4 described below has been administered by Independent Administration of 1-1 Chukoku No. 6 Tsukuba Totoro, Ibaraki, Japan (Postal Code 305-8566) since April 25, 2002.
  • the Patent Organism Depositary the deposit number FERM BP-8030, from April 16, 2002, 2-17-85, Jusanhoncho, Yodogawa-ku, Osaka-shi, Osaka (postal code 532-8686) They have been deposited with the Fermentation Research Institute (IF0) under the accession number IF0 16789.
  • Escherichia coli TOPlO / pAKKO-dogSLT obtained in Example 11 described below has been established on April 25, 2002, as an independent company of 1-1 Chukoku No. 1-1, Tsukuba Totoro, Ibaraki, Japan (zip code 305-8566). Deposited number at Patent Organism Depositary, National Institute of Advanced Industrial Science and Technology
  • RNA was prepared from the hypothalamus of the cynomolgus monkey using Isogen (Futtsubon Gene), a poly (A) + RNA fraction was prepared using Oligotex (dT) 30 (Takara Shuzo). ⁇ oly (A) T RNA 2. Power, et al. Superscript reversetranscr iptase
  • a cynomolgus monkey hypothalamic cDNA A PCR reaction was performed using the ⁇ type. The reaction was carried out using a cDNA equivalent to 20 ng of mRNA as a type II in a volume of 201.
  • the composition of the reaction solution was a primer concentration of 0.5 M, a dNTP mixture solution of 0.2 mM, Z-Taq (Takara Shuzo).
  • the cycle for amplification is After keeping it at 94 ° C for 120 seconds, a cycle of 98 ° C for 1 second and 72 ° C for 30 seconds was repeated 40 times.
  • the resulting reaction solution 21 was subcloned into a plasmid vector pcr4 using a 1 "0-0 TA cloning kit (Invitrogen), and then introduced into E. coli T0P10.
  • a poly (A) + RNA fraction was prepared using Oligotex (dT) 30 (Takara Shuzo).
  • Type III for 3'-RACE was prepared based on feline whole brain poly (A) + RNA using SMART RACE cDNA Amplification kit.
  • the primer set used for the RACE PCR reaction was as follows: the first PCR reaction included the Universal Primer Mix and primer 1 (SEQ ID NO: 5) attached to the kit; the second PCR reaction included the Nested Universal Primer and primer 2 (sequence number) attached to the kit. : 6) was used. The reaction was carried out in the form of ⁇ with a volume of 201 liquid, ie, 11 * 6 equivalents of the reversely transcribed 00 ⁇ .
  • the composition of the reaction solution was a primer concentration of 0.5 zM, a dNTP mixed solution of 0.2 mM, Advantage? Polymerase Mix (Clontech) 1/50 volume ⁇ 10-fold concentrated Buffer 1/10 volume.
  • the cycle for amplification is 94X ⁇ 5 seconds for both PCRs,
  • Type I for 5'-RACE was prepared from feline whole brain poly (A) + RNA using Marathon ready cDNA Amplification'kit.
  • the set of primers used for the RACE PCR reaction included adapter primer 1 for the Marathon ready cDNA Amplification kit in the first PCR reaction.
  • Primer 2 (SEQ ID NO: 9) and primer 2 (SEQ ID NO: 9) for Marathon ready cDNA Amplification kit were used in the second PCR reaction. The reaction was carried out in the amount of 20 ng of the reverse transcribed cDNA corresponding to 2 ng of mRNA at a volume of 20 / i1.
  • the composition of the reaction solution was primer concentration 0.5 M, dNTP mixture 0.2 mM, Advantage 2 Polymerase Mix (Clontech) 1/50 volume, and 10-fold concentrated Buffer 1/10 volume.
  • the cycle for amplification is 10 cycles of 94 ° C for 5 seconds, 66 ° C for 120 seconds for both PCRs, 94 ° C for 5 seconds, 60 ° C for 20 seconds, 72 ° C for After repeating the cycle of 120 seconds 25 times, it was kept at 72 ° C for 10 minutes.
  • the reaction product was electrophoresed on a 1.2% Seakem GTG Agarose (Takara Shuzo), and a band around 500 bp, which was visible when stained with ethidium amide, was extracted with the GeneClean Spin kit (Bio 101). After subcloning into plasmid vector pcr4-T0P0 using E. coli, Escherichia coli E0 was introduced. Plasmid DNA was purified from the resulting transformant using QIA prep8 mini prep (Qiagen). The reaction for base sequence determination was performed using BigDye Terminator Cycle Sequence Ready Reaction Kit (Perkin Elmer). As a result of decoding using a fluorescent automatic sequencer, a base sequence represented by SEQ ID NO: 10 including the N-terminal region of cat SLT was obtained.
  • the cycle for amplification is as follows: after holding at 94 ° C for 15 seconds, 98t: 1 second, 64 ° C, 15 seconds, 72 ° C Keep warm for a minute.
  • the reaction product was electrophoresed on 1.2% Seakein GTG Agarose (Takara Shuzo), and a band around 1200 bp, which was visible when stained with ethidium bromide, was extracted using the GeneClean Spin kit (Bio 101) and the Eukaryotic T0P0 TA cloning kit (Invitrogen) After subcloning into plasmid vector pcDNA3.1 / V5 / His-T0P0 using Escherichia coli, E. coli TOP10 was introduced.
  • Plasmid DM was purified from the resulting transformant using QIA prep8 mini prep (Qiagen). The reaction for base sequence determination was performed using a BigDye Terminator Cycle Sequence Ready Reaction Kit (Pakkin Elma). As a result of decoding using a fluorescent automatic sequencer, the nucleotide sequence represented by SEQ ID NO: 39 was obtained. Since this sequence contained the nucleotide sequence (SEQ ID NO: 13) encoding the entire amino acid sequence of cat SLT (SEQ ID NO: 14), Escherichia coli TOP10 was transformed with this plasmid to transform Escherichia coli TOP10. / pcDNA-catSLT was obtained.
  • SEQ ID NO: 13 the nucleotide sequence represented by SEQ ID NO: 13
  • SEQ ID NO: 14 the nucleotide sequence represented by SEQ ID NO: 14
  • a poly (A) + RNA fraction was prepared using Oligotex (dT) 30 (Takara Shuzo).
  • a type for 3'_RACE was prepared using SMART RACE cDNA Amplification kit based on canine frontal lobe poly (A) + RNA.
  • the primer set used for the RACE PCR reaction was the Universal Primer Mix and primer 1 (SEQ ID NO: 15) attached to the kit in the fourth PCR reaction, and the Nested Universal Primer and primer 1 in the kit for the second PCR reaction. (SEQ ID NO: 16) was used. The reaction was performed by converting 20 ng of reverse-transcribed cDNA into Performed with 1 liquid volume.
  • the composition of the reaction solution was primer concentration 0.5 / M, dNTP mixture 0.2 mM, Advantage2 Polymerase Mix (Clontech) 1/50 volume ⁇ 10-fold concentrated Buffer 1/10 volume.
  • the cycles for amplification were as follows: 94 ° C for 5 seconds, 72 ° C for 90 seconds, three cycles for each PCR, 94 ° C for 5 seconds, 70 ° C for 90 seconds, three cycles. After repeating the cycle of ° C for 5 seconds and 68 ° C for 90 seconds 40 times, it was kept at 72 ° C for 10 minutes.
  • reaction product was electrophoresed on 1.2% Seakem GTG Agarose (Takara Shuzo), and a band around 900 bp, which was visible when stained with ethidium bromide, was extracted with the GeneClean Spin kit (Bio 101). ) was used for subcloning into a plasmid vector pc-TOPO, and then introduced into E. coli T0P10. Plasmid DNA was purified from the resulting transformant using QIA prep8 mini prep (Qiagen). The reaction for base sequence determination is BigDye Terminator Cycle Sequence Ready
  • -Type II for RACE was prepared from canine frontal lobe poly (A) + RNA using Marathon ready cDNA Amplification kit.
  • the primer set used for the RACE PCR reaction was the adapter primer 1 and primer 1 (SEQ ID NO: 18) for the Marathon ready cDNA Amplification kit in the first PCR reaction, and the Marathon ready cDNA Amplification kit in the second PCR reaction.
  • the adaptor primer 2 and the primer 1 (SEQ ID NO: 19) were used.
  • the reaction was carried out in the form of type II with a volume of 201, corresponding to 2 ng of mRNA of the reverse-transcribed cDNA.
  • the composition of the reaction solution was primer concentration 0.5 M, dNTP mixture 0.2 mM,
  • a cycle of 66 ° C for 120 seconds was repeated 10 times, a cycle of 94 for 5 seconds, a cycle of 60 ° C for 20 seconds, and a cycle of 72 for 120 seconds was repeated 25 times, followed by incubation at 72 ° C for 10 minutes.
  • Reactants to 1.2% Seakem GTG 1.2% Seakem GTG
  • primer 1 SEQ ID NO: 21
  • 3 'PCR was performed by setting Primer 2 (SEQ ID NO: 22) to the untranslated region.
  • the reaction was carried out in the form of type II with a volume of 201, corresponding to 2 ng of mRNA of the reverse-transcribed cDNA.
  • the composition of the reaction solution was a primer concentration of 0.5 M, a dNTP mixed solution of 0.2 mL, a Z-Taq (TaKaRa) 1/50 volume, and a 10-fold concentrated Buffer 1/10 volume.
  • the cycle for amplification is as follows: after holding at 94 ° C for 15 seconds, repeat the cycle of 98 ° C for 1 second, 64 ° C for 15 seconds, 72 ° C for 20 seconds 40 times, and then for 72 ° C. For 2 minutes.
  • the reaction product was electrophoresed on a 1.2% Seakem GTG Agarose (Takara Shuzo), and a band around 1200 bp, which was visible when stained with ethidium umide, was extracted with the GeneClean Spin kit (Bio 101).
  • Eukaryotic T0P0 TA cloning kit (Invitrogen) After subcloning into the plasmid vector PCDNA3.1 / V5 / His-TOP0 using Escherichia coli, Escherichia coli T0P10 was introduced. Plasmid DNA was purified from the resulting transformant using QIA prep8 mini prep (Qiagen). The reaction for base sequence determination was carried out using BigDye Terminator Cycle Sequence Ready Reaction Kit (Pakkin Elma). As a result of decoding using a fluorescent automatic sequencer, the base sequence represented by SEQ ID NO: 40 was obtained.
  • This sequence contained the nucleotide sequence (SEQ ID NO: 23) encoding the entire amino acid sequence of canine SLT (SEQ ID NO: 24).
  • Bacterium TOP10 was transformed to obtain Escherichia coli TOPlO / pcDNA-dogSLT.
  • Plasmids were prepared from the cloned Escherichia coli using the Plasmid Midi Kit (Qiagen) and cut with restriction enzymes Sal I and Spe I to cut out the insert. After the electrophoresis, the insert DNA was cut out from an agarose gel with a force razor, and then recovered by performing fragmentation, phenol extraction, phenol-chloroform extraction, and ethanol precipitation.
  • Plasmid Midi Kit Plasmid Midi Kit (Qiagen). This was added to the CHO dhfr cell 500,000 cells / 150 1 O tiMEM (Gibco) cell suspension at 50 g / ml, and 240 V 960 F pulse was applied to the cells by Genepulsa (Bio-Rad).
  • CHO / catSLT cells are suspended in DMEM containing 10% dialyzed fetal serum to a concentration of 15 x 10 4 cells / ml and dispensed into a 96-well plate for FLIPR (Black plate clear bottom, Costar). 200 1 by implantation into the Ueru using (3.0X10 4 cells / 200 l / ⁇ El), 5% C0 2 was 37 ° C De ⁇ cultured at incubator base one coater in was used to Atsusi (hereinafter This plate is called a cell plate).
  • HANKS '/ HBSS Nesy Hanks 2
  • a sample plate was prepared by adding MCH at a concentration of 1. The color of the cell plate After finishing the washing, wash the cell plate 4 times using a plate washer (Molecular Devices) with HANKS '/ HBSS and 2.5 mM Probenecid using a washing buffer. The cell plate and the sample plate were set on the FLIPR and assembled (the sample from the sample plate was transferred to the cell plate by the FLIPR).
  • MCH increased the intracellular Ca ion concentration of feline SLT-expressing CH0 cells in a concentration-dependent manner (Fig. 1).
  • the increase in intracellular Ca ion concentration is indicated by the increase in fluorescence of the dye loaded on the cell caused by Ca.
  • RNA fraction was prepared using ISOGEN (Futtsubon Gene). The obtained RNA was digested with RNase by digestion with proteinase K (Invitrogen), DNase I digested genomic DNA using the Message Clean Kit (GenHunter), and total RNA lxg at each site was extracted.
  • Reverse transcription was carried out using Superscript II reversetranscriptase (Invitrogen) as a template and random primers according to the attached manual to produce cDNA.
  • the standard cDNA was prepared by PCR using the primers 1 and 2 (SEQ ID NO: 32 and SEQ ID NO: 33) in the form of 200 xl using primers 1 and 2 (SEQ ID NO: 32 and SEQ ID NO: 33) with 50 pg of the plasmid pcDNA-catSLT obtained in Example 4. Done and created.
  • the composition was performed using primers 1 and 2, 0.5 M, 2.5 mM MgCl 2 , dNTP 0.2 niM, AmliTaq Gold (Pakinkin Elmer) 1/100 volume, and 10-fold concentrated Ampl iTaq Gold Buffer 1/10 volume.
  • the reaction was incubated at 95 ° C for 10 minutes, and then repeated at 95 ° C for 15 seconds, 60 at 15 seconds, and 72 ° C for 10 seconds 40 times.
  • the amplification product is purified from the reaction mixture using the PCR purification kit (Qiagen), the absorbance at 260 nm is measured, the concentration is calculated, the exact copy number is calculated, and the copy is diluted with distilled water.
  • a standard cDNA solution of 7 copies of lxlO was prepared. Also,
  • TaqMan PCR probes and primers were designed using Primer Express (Version I.O) (PE Biosystems).
  • the expression level was calculated using ABI PRISM 7700 SDS software. The number of cycles at the moment when the fluorescence intensity of the reporter reaches the set value is plotted on the vertical axis. A standard curve was created using the logarithmic value of the initial concentration as the horizontal axis. The initial concentration of each reverse transcription product was calculated from the standard curve, and the expression amount of the cat SLT gene per 25 ng of total RNA at each site was determined. Further, the expression level of the feline GAP DH gene in each sample was determined using the primers # 3 (SEQ ID NO: 35), primer 4 (SEQ ID NO: 36) and the probe (Fam-ccaggagcgagatcccgcca-Tamura) shown in SEQ ID NO: 37. The expression of the feline SLT gene was corrected by the TaqMan: PCR method in the same manner as in the analysis of the SLT gene expression level.
  • the expression level of the feline SLT gene relative to the feline GAPDH gene was 0.084% in the frontal lobe, 0.32% in the temporal lobe, 0.046% in the occipital lobe, 0.056% in the parietal lobe, 0.086% in the pons, and 0.012% in the cerebellum.
  • a plasmid into which a gene encoding the full-length amino acid sequence of canine SLT to which the sequence was confirmed in Example 7 and a Sal I recognition sequence was added on the 5 'side and a Spe I recognition sequence was added on the 3' side was added.
  • E. coli was transformed with pcDNA-dogSLT. Plasmids were prepared from the E. coli clones using Plasmid Midi Kit (Qiagen) and cut with restriction enzymes Sal I and Spe I to cut out the insert.
  • proteins, polynucleotides, antisense polynucleotides and antibodies of the present invention include, for example, anorexia (eg, anorexia nervosa), anemia or hypoproteinemia associated with anorexia, weak labor, lax hemorrhage, uterus It is useful as a marker for diagnosis of retro failure, milk stasis, etc.
  • anorexia eg, anorexia nervosa
  • anemia or hypoproteinemia associated with anorexia weak labor
  • lax hemorrhage uterus It is useful as a marker for diagnosis of retro failure, milk stasis, etc.
  • An agonist of the protein obtained by a screening method using the protein and the peptide of the present invention includes an appetite (feeding) enhancer, anorexia (eg, anorexia nervosa), anemia associated with anorexia, As a prophylactic / therapeutic agent for hypoproteinemia, weak labor, laxative bleeding, uterine remodeling failure, milk stasis, etc.
  • anorexia eg, anorexia nervosa
  • anemia associated with anorexia As a prophylactic / therapeutic agent for hypoproteinemia, weak labor, laxative bleeding, uterine remodeling failure, milk stasis, etc.
  • Obesity hyperplasmic obesity, pituitary obesity, hypoplasmic obesity, hypothyroid obesity, hypothalamic obesity, symptomatic obesity, childhood obesity, upper body obesity, dietary obesity, hypogonadism Obesity, systemic mastocytosis, simple obesity, central obesity, etc.), hyperphagia, affective disorders, sexual dysfunction, excessive labor, tonic contractions, fetal distress, uterine rupture , Vascular laceration, premature birth, Prader-Willi syndrome, diabetes and its complications (eg, diabetic nephropathy, diabetic retinopathy, diabetic neuropathy, etc.), hypertension, hyperlipidemia, coronary atherosclerosis, Prevention and treatment of gout, respiratory diseases (Pickwick syndrome, sleep apnea syndrome), fatty liver, infertility, osteoarthritis (especially antiobesity agents, appetite (feeding) regulators, etc.) It can be used as an agent.
  • respiratory diseases Pickwick syndrome, sleep apnea syndrome

Abstract

Novel proteins of ape-, feline- and canine-origins or salts thereof, polynucleotides encoding these proteins, antibodies against these proteins, etc. are useful as, for example, diagnostic markers for inappetence, uterine inertia and so on. Compounds obtained by screening with the use of these proteins and MCH are usable as appetite enhancers and preventives/remedies for inappetence, etc.

Description

明細 新規タンパク質、 その D N Aおよび用途 技術分野  Description New protein, its DNA and application Technical field
本発明は、 新規な Gタンパク質共役型レセプタ一タンパク質、 それをコードす る D NA、 それらの用途などに関する。 詳しくは、 食欲不振、.食欲不振に伴う貧 血ならびに低蛋白症、 微弱陣痛、 弛緩出血、 子宮復古不全もしくは乳汁うっ滞の 予防 ·治療作用、 または肥満症、 摂食亢進症、 情動障害、 性機能障害、 過強陣痛、 強直性子宮収縮、 早産、 Prader-Wi l l i症候群、 糖尿病およびその合併症、 高血圧、 高脂血症、 冠状動脈硬化症、 痛風、 呼吸器疾患、 脂肪肝、 不妊症もしくは変形性 骨関節症の予防 ·治療作用を有する化合物のスクリーニングなどに関する。 背景技術  The present invention relates to a novel G protein-coupled receptor protein, a DNA encoding the same, and uses thereof. For more information, anorexia, anemia associated with anorexia, prevention and treatment of hypoproteinemia, weak labor, laxative bleeding, uterine remodeling failure or milk stasis, or obesity, hyperphagia, affective disorder, sex Dysfunction, labour, tonic contractions, preterm birth, Prader-Willi syndrome, diabetes and its complications, hypertension, hyperlipidemia, coronary atherosclerosis, gout, respiratory disease, fatty liver, infertility or The present invention relates to screening for compounds having a prophylactic and therapeutic effect on osteoarthritis. Background art
肥満は、 糖尿病や心疾患など数多くの生活習慣病発症のリスクファクターとな ることが知られている。 食生活や生活様式の変化に伴い、 特に先進国において肥 満は増大しており、 医療経済学的側面から社会的にも非常に大きな問題となって いる。 肥満は種々の要因に起因することが知られているが、 最近、 摂食中枢であ る視床下部に存在するいくつかのぺプチドが哺乳動物において摂食行動の促進あ るいは抑制に関与することが明らかとなり、 これらのペプチドの肥満に対する寄 与が特に注目を集めている。 これらのペプチドの作用に対して阻害または活性化 を示す薬剤は中枢性抗肥満薬としての可能性が考えられるからである。 このよう な視床下部ペプチドの一つであるメラニン凝集ホルモン (Melanin  Obesity is known to be a risk factor for the development of many lifestyle-related diseases such as diabetes and heart disease. With changes in eating habits and lifestyles, fertility is increasing, especially in developed countries, and it has become a very socially significant problem from the health economic perspective. Obesity is known to be caused by various factors.Recently, several peptides located in the hypothalamus, the feeding center, are involved in promoting or suppressing feeding behavior in mammals. Thus, the contribution of these peptides to obesity has received particular attention. This is because drugs that inhibit or activate the action of these peptides are considered to be potential central antiobesity drugs. One such hypothalamic peptide, melanin-concentrating hormone (Melanin
Concentrat ing Hormone) (以下、 MCHと記載することがある) は、 もともと魚類 の黒色素胞の色素顆粒を凝集する体色変化ホルモンとして見出されたペプチドで あるが、 その後、 ヒト、 ラットおよびマウスのような哺乳類にも存在することが 明らかとなった。 最近、 MCHが哺乳類における摂食行動およびエネルギー代謝に おいて重要な役割を果たすことを示す事実がいくつか報告されている。 Concentrating Hormone) (hereinafter sometimes referred to as MCH) is a peptide originally found as a color changing hormone that aggregates pigment granules of fish melanophores. It is clear that it exists in mammals such as. Recently, several facts have been reported indicating that MCH plays an important role in feeding behavior and energy metabolism in mammals.
哺乳類では M C Hは摂食中枢である視床下部外側野および不定帯に局在するこ とより摂食および飲水行動への関与が予想されていれたが、 Quらによって MCHの 摂食行動への関与が最初に以下のように示された (Qu, D.ら、 Nature, 380巻, 243- 247頁、 1996年) 。 遺伝的肥満モデルである ob/obマウスと野生型マウスの視 床下部において発現量が変化している mRNAを di f ferent i al di sp l ay法で探索した ところ、 MCHの前駆体遺伝子の発現が顕著に増加していることが見出された。 ま た、 MCH遺伝子発現の増加は、 24時間の絶食によって野生型および ob/obマウスの いずれにおいても認められた。 これらの結果は、 MCHが摂食刺激ペプチドとして 機能していることを示唆したが、 事実、 MCHをラットの側脳室に直接投与するこ とにより有意に摂食量が増加した。 さらに、 MCH遺伝子ノックアウトマウスが摂 食量の減少および代謝の増加により顕著に体重が減少することが報告されたIn mammals, MCH is localized in the feeding center, the hypothalamic lateral area and the irregular zone. And were more likely to be involved in eating and drinking behavior, but Qu et al. First showed that MCH was involved in eating behavior as follows (Qu, D. et al., Nature, vol. 380). , 243-247, 1996). Expression of altered mRNA levels in the hypothalamus of ob / ob mice and wild-type mice, which are genetically obese models, was searched for by the di ferent al al di sp lay method. Was found to be significantly increased. Increased MCH gene expression was observed in both wild-type and ob / ob mice after a 24-hour fast. These results suggested that MCH functioned as a food-stimulating peptide, but in fact, administration of MCH directly to the lateral ventricle of rats significantly increased food intake. In addition, it was reported that MCH knockout mice lost significant weight due to reduced food intake and increased metabolism.
(Nature, 396巻, 670- 674頁、 1998年) 。 注目すべきことに、 ごのマウスでは体 重減少以外の行動あるいはその他の生化学パラメータ一は、 野生型に比べて何の 変化も認められなかった。 最近になつて MCHを過剰産生する遺伝子改変マウスが 肥満の表現型を示し、 インスリン抵抗性を呈することも報告されている (J . Cl in. Inves t . , 107巻, 3793- 86頁、 2001年) 。 以上の事実は、 MCHが摂食行動お よびエネルギー代謝において中心的な役割を担つていることを意味している。 視床下部には MCH以外にも、 ニューロペプチド Y、 ガラニンまたはォレキシンに 代表される種々の摂食促進ペプチドが存在している。 しかし、 MCH遺伝子欠損マ ウスに認められた摂食行動の抑制や体重減少は、 これらのペプチドの遺伝子ノッ クァゥト動物では報告されておらず、 MCHが摂食行動において特に重要な調節因 子として機能していると考えられる。 また、 MCH遺伝子欠損マウスが体重減少以 外に目立つた表現型を示さないことから、 MCHの作用を抑制することによつて副 作用の少ない抗肥満薬が開発される可能性が示唆された。 (Nature, 396, 670-674, 1998). Remarkably, no change in behavior or other biochemical parameters other than weight loss was observed in the mice compared to the wild type. It has also recently been reported that genetically modified mice that overproduce MCH exhibit an obese phenotype and exhibit insulin resistance (J. Clin. Invest., 107, 3793-86, 2001). Year) . These facts imply that MCH plays a central role in feeding behavior and energy metabolism. In the hypothalamus, in addition to MCH, various feeding-promoting peptides represented by neuropeptide Y, galanin or orexin are present. However, suppression of eating behavior and weight loss observed in mice deficient in the MCH gene have not been reported in animals knocked down by these peptides, and MCH functions as a particularly important regulator in eating behavior. it seems to do. In addition, since the MCH gene-deficient mice did not show any prominent phenotype other than weight loss, it was suggested that suppressing the effect of MCH could lead to the development of anti-obesity drugs with few side effects.
MCHの機能的受容体としてはこれまでに SLC- 1 (MCHK MCHR1) が知られていた (Nature, 400巻、 26-1265頁、 1999年、 Nature、 400巻、 265-269頁、 1999年、 Bi ochei. Bi ophys. Res. Commun.、 261巻、 622- 626頁、 1999年、 Nature Cel l Bi o l . , 1卷、 267- 271頁、 1999年、 FEBS Let t . , 457巻、 522- 524頁、 1999年、 W0 00/40725号公報等) が、 最近、 さらに、 MCHの第二の受容体サブタイプとして SLT (MCH2、 CHR2) がクロ一ニングされた (J . Bio l . Chem.、 276巻、 20125-20129 頁、 2001年、 Biochem. Biophys. Res. Co腿亂、 283卷、 1013- 1018頁、 2001年、 Proc. Nat l. Acad. Sci. USA、 98巻、 7564- 7569頁、 2001年、 Pro at l . Acad. Sc i . .USA、 98巻、 7576-7581頁、 2001年、 J. Biol . Chem.、 276巻、 34664-34670 頁、 2001年、 Mol. Pharmacol . , 60巻、 632- 639頁、 2001年、 W0 02/03070号公報, W0 00/49046号公報等) 。 これら二つの MCH受容体についていずれの受容体が MCH の生理作用、 特に摂食に対する作用に関与しているかは現時点において明らかで はない。 SLC- 1受容体は、 特に、 摂食に関与する視床下部の領域 (弓状核、 腹内 側核、 背内側核、 室傍核) や報酬系に関わるとされる側坐核に強く発現している のに対し、 SLTは海馬や扁桃に発現レていて視床下部での発現が低いことから、 MCHの摂食宂進作用には主として SLC- 1が関与し、 SLTはそれ以外の MCHの作用であ ' る記憶や情動に関与している可能性が示唆されている。 一方、 SLTは視床下部に おいて SLC— 1とは異なる領域 (前核、 外側核など) に分布し、.また、 小児肥満の 原因遺伝子の近傍に配座していることから摂食に関与する可能性があるとする報 告もある (Proc. Nat l . Acad. Sci. USA, 98巻、 7564- 7569頁、 2001年) 。 しか し、 SLC- 1がヒト以外にラットなどにもその遺伝子が存在し、 組織分布解析など が報告されているのに対し、 SLTは現在までにヒト以外の適当な実験動物にその 存在が報告されておらず、 MCHの摂食亢進などの生理作用への関与の機構につい て必ずしも十分には明らかにされていない。 SLC-1 (MCHK MCHR1) was previously known as a functional receptor for MCH (Nature, 400, 26-1265, 1999, Nature, 400, 265-269, 1999, Bi ochei. Bi ophys. Res. Commun., 261, 622-626, 1999, Nature Cell Biol., 1st, 267-271, 1999, FEBS Lett. 524, 1999, WO 00/40725, etc.), and recently, SLT (MCH2, CHR2) has been cloned as the second receptor subtype of MCH (J. Biol. Chem. , Volume 276, 20125-20129 Co., 2001, Biochem. Biophys. Res.Co, Vol. 283, 1013- 1018, 2001, Proc. Natl. Acad. Sci. USA, 98, 7576-7581, 2001, J. Biol. Chem., 276, 34664-34670, 2001, Mol. Pharmacol., 60, 632-639 2001, W002 / 03070, W00 / 49046, etc.). It is not clear at this time which of these two MCH receptors is involved in the physiological effects of MCH, particularly on feeding. SLC-1 receptor is particularly strongly expressed in the hypothalamus region involved in feeding (arcus nucleus, ventral nucleus, dorsolateral nucleus, paraventricular nucleus) and in the nucleus accumbens which is thought to be involved in the reward system On the other hand, SLT is expressed in the hippocampus and tonsils and is low in the hypothalamus, so SLC-1 is mainly involved in the inducing action of MCH, and SLT is It has been suggested that they may be involved in memory and emotion's effects. On the other hand, SLT is distributed in the hypothalamus in areas different from SLC-1 (pronucleus, lateral nucleus, etc.), and is involved in feeding because it is located near the causative gene of childhood obesity. Some reports have reported that this may be done (Proc. Natl. Acad. Sci. USA, 98, 75664-7569, 2001). However, while SLC-1 has its gene in rats as well as humans and its tissue distribution analysis has been reported, SLT has been reported in appropriate non-human experimental animals to date. The mechanism of MCH's involvement in physiological effects such as increased food intake has not always been fully elucidated.
M C Hの摂食亢進などの生理作用に対する M C H受容体である S L Tの関与を 明らかにし、 医薬のスクリーニング系を利用することにより、 全く新規な作用機 序を有する抗肥満薬などの医薬の開発が望まれていた。 発明の開示  By elucidating the involvement of SLT, which is an MCH receptor, in the physiological effects of MCH, such as increased food intake, and using a drug screening system, it is hoped that new drugs, such as anti-obesity drugs, having a novel mechanism of action will be developed. Was rare. Disclosure of the invention
本発明者らは上記の課題を解決するために鋭意研究を重ねた結果、 ヒト SLTに 相同性を示すサル、 ィヌおよびネコ SLTをコードする遺伝子を、 サル、 ィヌおよ びネコ脳 cDNAより,見出してクローニングすることに成功した。 さらに、 これらの 受容体は MCHと結合する。 これらの知見に基づいて、 さらに検討を重ねた結果、 本発明を完成するに至った。  The present inventors have conducted intensive studies in order to solve the above-mentioned problems, and as a result, have found that genes encoding monkey, canine, and feline SLTs showing homology to human SLT can be obtained from monkey, canine, and feline brain cDNA. More successfully found and cloned. In addition, these receptors bind to MCH. As a result of further study based on these findings, the present invention has been completed.
すなわち、 本発明は、 ( 1 ) 配列番号: 4、 配列番号: 14または配列番号: 24で表わされるアミ ノ酸配列と同一もしくは実質的に同一のァミノ酸配列を含有するタンパク質また はその塩、 That is, the present invention (1) a protein or a salt thereof containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 4, SEQ ID NO: 14 or SEQ ID NO: 24;
(2) 配列番号: 4で表わされるアミノ酸配列からなるタンパク質またはその 塩、  (2) a protein consisting of the amino acid sequence represented by SEQ ID NO: 4 or a salt thereof;
(3) 配列番号: 14で表わされるアミノ酸配列からなるタンパク質またはそ の塩、  (3) a protein comprising the amino acid sequence represented by SEQ ID NO: 14 or a salt thereof;
(4) 配列番号: 24で表わされるアミノ酸配列からなるタンパク質またはそ の塩、  (4) a protein consisting of the amino acid sequence represented by SEQ ID NO: 24 or a salt thereof;
( (55)) 上記 (1) 記載のタンパク質の部分ペプチドまたはその塩、  ((55)) a partial peptide of the protein according to (1) or a salt thereof,
(6) 上記 (1) 記載のタンパク質または上記 (5) 記載の部分ペプチドをコ ―ドするポリヌクレオチドを含有するポリヌクレオチド、  (6) a polynucleotide containing a polynucleotide encoding the protein of (1) or the partial peptide of (5),
(7) DNAである上記 (6) 記載のポリヌクレオチド、  (7) the polynucleotide according to (6), which is a DNA,
( 8 ) 配列番号: 3、 配列番号: 13または配列番号: 23で表わされる塩基 配列を含有する上記 (7) 記載のポリヌクレオチド、  (8) The polynucleotide according to the above (7), comprising the nucleotide sequence represented by SEQ ID NO: 3, SEQ ID NO: 13 or SEQ ID NO: 23,
(9) 上記 (8) 記載のポリヌクレオチドを含有する組換えベクター、' (9) a recombinant vector containing the polynucleotide according to (8),
(10) 上記 (9) 記載の組換えべクタ一で形質転換された形質転換体、(10) a transformant transformed with the recombinant vector according to the above (9),
(11) 上記 (10) 記載の形質転換体を培養し、 上言己 (1) 記載のタンパク 質または上記 (5) 記載の部分ペプチドを生成、 蓄積せしめ、 これを採取するこ とを特徴とする上記 (1) 記載のタンパク質もしくは上記 (5) 記載の部分ぺプ チドまたはその塩の製造法、 (11) culturing the transformant described in (10) above, producing and accumulating the protein described in (1) or the partial peptide described in (5) above, and collecting this. A method for producing the protein according to the above (1) or the partial peptide or the salt thereof according to the above (5),
(12) 上記 (1) 記載のタンパク質もしくは上記 (5) 記載の部分ペプチド またはその塩を含有してなる医薬、  (12) a medicine comprising the protein of (1) or the partial peptide of (5) or a salt thereof,
(13) 上記 (6) 記載のポリヌクレオチドを含有してなる医薬、  (13) a medicament comprising the polynucleotide according to the above (6),
(14) 上記 (6) 記載のポリヌクレオチドを含有してなる診断薬、  (14) a diagnostic agent comprising the polynucleotide according to (6),
(15) 上記 (1) 記載のタンパク質もしくは上記 (5) 記載の部分ペプチド またはその塩に対する抗体、  (15) an antibody against the protein according to (1) or the partial peptide according to (5) or a salt thereof,
(16) 上記 (1) 記載のタンパク質のシグナル伝達を不活性化する中和抗体 である上記 (15) 記載の抗体、 (17) 上記 (1) 記載のタンパク質のシグナル伝達を活性化する抗体である 上記 (15) 記載の抗体、 (16) the antibody according to (15), which is a neutralizing antibody that inactivates the signal transduction of the protein according to (1); (17) The antibody according to (15), which is an antibody that activates signal transduction of the protein according to (1).
(18) 上記 (15) 記載の抗体を含有してなる診断薬、  (18) a diagnostic agent comprising the antibody according to the above (15),
(19) 上記 (16) 記載の抗体を含有してなる医薬、  (19) a pharmaceutical comprising the antibody according to the above (16),
(20) 上記 (17) 記載の抗体を含有してなる医薬、  (20) a medicine comprising the antibody according to the above (17),
(21) 上記 (1) 記載のタンパク質または上記 (5) 記載の部分ペプチドま たはその塩および配列番号: 25で表されるアミノ酸配列と同一もしくは実質的 に同一のアミノ酸配列を含有するペプチドまたはその塩を用いることを特徴とす る、 配列番号: 25で表されるアミノ酸配列と同一もしくは実質的に同一のアミ ノ酸配列を含有するペプチドと上記 (1) 記載のタンパク貪またはその塩との結 合性を変化させる化合物またはその塩のスクリーニング方法、 一  (21) The protein described in the above (1) or the partial peptide or a salt thereof described in the above (5) and a peptide containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 25, or A peptide comprising the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 25, characterized by using a salt thereof, and a peptide according to the above (1) or a salt thereof. Screening method for a compound or a salt thereof that alters the binding of
(22) 上記 (1) 記載のタンパク質または上記 (5) 記載の部分ペプチドま たはその塩および配列番号: 25で表されるアミノ酸配列と同一もしくは実質的 に同一のアミノ酸配列を含有するペプチドまたはその塩を含有することを特徴と する、 配列番号: 25で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するペプチドと上記 (1) 記載のタンパク質またはその塩との 結合性を変化させる化合物またはその塩のスクリーニング用キット、  (22) The protein described in the above (1) or the partial peptide or a salt thereof described in the above (5) and a peptide containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 25, or A peptide comprising an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 25, characterized by containing a salt thereof, and a protein according to the above (1) or a salt thereof. A kit for screening a compound that changes binding property or a salt thereof,
(23) 上記 (21) 記載のスクリーニング方法または上記 (22) 記載のス クリーニング用キットを用いて得られうる、 配列番号: 25で表されるアミノ酸 配列と同一もしくは実質的に同一のァミノ酸配列を含有するぺプチドまたはその 塩と上記 (1) 記載のタンパク質またはその塩との結合性を変化させる化合物ま たはその塩、  (23) An amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 25, which can be obtained using the screening method described in (21) or the screening kit described in (22). A compound or a salt thereof that alters the binding property between the peptide or a salt thereof and the protein or the salt thereof according to (1) above,
(24) ァゴニストである上記 (23) 記載め化合物またはその塩、  (24) The compound described in (23) above, which is an agonist, or a salt thereof,
(25) アンタゴニストである上記 (23) 記載の化合物またはその塩、 (26) 上記 (24) 記載の化合物またはその塩を含有してなる医薬、 (27) 上記 (25) 記載の化合物またはその塩を含有してなる医薬、 (28) 上記 (6) 記載のポリヌクレオチドと相補的な塩基配列またはその一 部を含有してなるアンチセンスポリヌクレオチド、  (25) The compound according to the above (23) or a salt thereof, which is an antagonist; (26) a medicine comprising the compound or the salt thereof according to the above (24); (27) the compound according to the above (25) or a salt thereof (28) an antisense polynucleotide comprising a nucleotide sequence complementary to the polynucleotide of (6) or a part thereof,
(29) 上記 (28) 記載のアンチセンスポリヌクレオチドを含有してなる医 (30) 上記 (6) 記載のポリヌクレオチドまたはその一部を用いることを特 徵とする上記 (1) 記載のタンパク質の mRNAの定量方法、 (29) A medical comprising the antisense polynucleotide according to (28). (30) a method for quantifying the mRNA of the protein according to (1), which comprises using the polynucleotide or a part thereof according to (6);
(31) 上記 (15) 記載の抗体を用いることを特徴とする上記 (1) 記載の タンパク質の定量方法、  (31) The method for quantifying a protein according to (1), wherein the antibody according to (15) is used.
(32) 上記 (30) または (31) 記載の定量方法を用いることを特徴とす る上記 (1) 記載のタンパク質の機能が関連する疾患の診断方法、  (32) A method for diagnosing a disease associated with the function of the protein according to (1), which comprises using the quantification method according to (30) or (31).
(33) 上記 (30) 記載の定量方法を用いることを特徴とする上記 (1) 記 ' 載のタンパク質の発現量を変化させる化合物またはその塩のスクリーニング方法、 (34) 上記 (30) 記載の定量方法を用いることを特徴とする細胞膜におけ る上記 (1) 記載のタンパク質量を変化させる化合物またはその塩のスクリー二 ング方法、  (33) A method for screening a compound or a salt thereof that alters the expression level of the protein described in (1) above, which comprises using the quantification method described in (30) above. A method for screening a compound or a salt thereof, which changes the amount of a protein in a cell membrane according to the above (1), characterized by using a quantification method;
(35) 上記 (33) 記載のスクリーニング方法を用いて得られうる上記 ( 1 ) 記載のタンパク質の発現量を変化させる化合物またはその塩、  (35) A compound or a salt thereof that alters the expression level of the protein according to (1), which can be obtained by using the screening method according to (33).
(36) 上記 (34) 記載のスクリーニング方法を用いて得られうる細胞膜に おける上記 (1) 記載のタンパク質量を変化させる化合物またはその塩、  (36) a compound or a salt thereof, which alters the amount of the protein according to (1) above in a cell membrane obtainable by using the screening method according to (34);
(37) 上記 (35) または (36) 記載の化合物またはその塩を含有してな る医薬、  (37) A medicament comprising the compound according to (35) or (36) or a salt thereof,
(38) 上記 (6) 記載のポリヌクレオチドとハイストリンジェントな条件下 でハイブリダイズするポリヌクレオチド、  (38) a polynucleotide that hybridizes to the polynucleotide according to (6) under conditions of high stringency,
(39) タ来性の、 上記 (1) 記載のタンパク質をコードする DNAまたはそ の変異 DNAを有する非ヒトトランスジヱニック動物、  (39) a non-human transgenic animal having a DNA encoding the protein of (1) or a mutant DNA thereof,
(40) 食欲不振、 食欲不振に伴う貧血ならびに低蛋白症、 微弱陣痛、,弛緩出 血、 子宮復古不全または乳汁うっ滞の予防 ·治療剤である上記 (12) 、 (1 3) 、 (20) または (26) 記載の医薬、  (40) The above (12), (13), (20) which is an agent for the prevention and treatment of anorexia, anemia associated with anorexia, hypoproteinemia, weak labor, bleeding blood, uterine remodeling failure or milk stasis ) Or the medicament according to (26),
(41) 巴満症、 摂食宂進症、 情動障害、 性機能障害、 過強陣痛、 強直性子宮 収縮、 早産、 Prader-Willi症候群、 糖尿病およびその合併症、 高血圧、 高脂血症、 冠状動脈硬化症 痛風、 呼吸器疾患、 脂肪肝、 不妊症または変形性骨関節症の予 防,治療剤である、 上記 (19) 、 (27) または (29) 記載の医薬、 ( 4 2 ) 哺乳動物に対して、 上記 (2 4 ) 記載の化合物またはその塩の有効量 を投与することを特徴とする、 食欲不振、 食欲不振に伴う貧血ならびに低蛋白症、 微弱陣痛、 弛緩出血、 子宮復古不全または乳汁うっ滞の予防 ·治療方法、 (41) Tomomiasis, eating disorders, affective disorders, sexual dysfunction, overwork labor, ankylosing contractions, preterm birth, Prader-Willi syndrome, diabetes and its complications, hypertension, hyperlipidemia, coronary The medicament according to (19), (27) or (29), which is a prophylactic or therapeutic agent for arteriosclerosis gout, respiratory disease, fatty liver, infertility or osteoarthritis; (42) Anorexia, anemia associated with anorexia, hypoproteinemia, weak labor, and relaxation characterized by administering to a mammal an effective amount of the compound or a salt thereof according to (24). Prevention and treatment of bleeding, uterine repair failure or milk stasis
( 4 3 ) 哺乳動物に対して、 上記 (2 5 ) 記載の化合物またはその塩の有効量 を投与することを特徴とする、 肥満症、 摂食亢進症、 情動障害、 性機能障害、 過 強陣痛、 強直性子宮収縮、 早産、 Prader-Wi l l i症候群、 糖尿病およびその合併症、 高血圧、 高脂血症、 冠状動脈硬化症、 痛風、 呼吸器疾患、 脂肪肝、 不妊症または 変形性骨関節症の予防 ·治療方法、 . ,  (43) An obesity, hyperphagia, affective disorder, sexual dysfunction, or hyperactivity, comprising administering to a mammal an effective amount of the compound according to (25) or a salt thereof. Labor, ankylosing uterine contractions, preterm birth, Prader-Willi syndrome, diabetes and its complications, hypertension, hyperlipidemia, coronary atherosclerosis, gout, respiratory disease, fatty liver, infertility or osteoarthritis Prevention and treatment of
( 4 4 ) 食欲不振、 食欲不振に伴う貧血ならびに低蛋白症、 微弱陣痛、 弛緩出 血、 子宮復古不全または乳汁うっ滞の予防 ·治療剤を製造するための、 上記 (2 4 ) 記載の化合物またはその塩の使用、 '  (44) The compound according to (24) for the manufacture of a prophylactic or therapeutic agent for anorexia, anemia associated with anorexia, hypoproteinemia, weak labor, lax blood, uterine remodeling failure or milk stasis. Or the use of its salts, '
( 4 5 ) 肥満症、 摂食亢進症、 情動障害、 性機能障害、 過強陣痛、 強直性子宮 収縮、 早産、 Prader-Wi l l i症候群、 糖尿病およびその合併症、 高血圧、 高脂血症、 冠状動脈硬化症、 痛風、 呼吸器疾患、 脂肪肝、 不妊症または変形性骨関節症の予 防 ·治療剤を製造するための、 上記 (2 5 ) 記載の化合物またはその塩の使用な どを提供する。 図面の簡単な説明  (45) Obesity, hyperphagia, affective disorders, sexual dysfunction, overwork labor, ankylosing uterine contractions, preterm birth, Prader-Willi syndrome, diabetes and its complications, hypertension, hyperlipidemia, coronary Provide the use of the compound described in (25) or a salt thereof for the manufacture of a prophylactic or therapeutic agent for arteriosclerosis, gout, respiratory disease, fatty liver, infertility or osteoarthritis. I do. BRIEF DESCRIPTION OF THE FIGURES
図 1は、 FLIPRを用いて測定した種々の濃度の MCHのネコ SLT発現 CH0細胞に対す る細胞内 Caイオン上昇活性を示す図である。  FIG. 1 is a graph showing the intracellular Ca ion elevating activity of various concentrations of MCH on cat SLT-expressing CH0 cells measured using FLIPR.
図 2は、 ネコ SLT遺伝子のネコ脳各部位における発現量のグラフである。 発現 量は、 同じ铸型 RNA量に対するネコ SLT遺伝子の発現量をネコ GAPDH遺伝子の発現 量で除した値を 1000倍にして示した。 発明を実施するための最良の形態 _  FIG. 2 is a graph of the expression level of the cat SLT gene in various parts of the cat brain. The expression level was expressed as 1000 times the value obtained by dividing the expression level of the cat SLT gene for the same type II RNA level by the expression level of the cat GAPDH gene. BEST MODE FOR CARRYING OUT THE INVENTION _
配列番号: 4、 配列番号: 1 4または配列番号: 2 4で表わされるアミノ酸配 列と同一もしくは実質的に同一のアミノ酸配列を含有するタンパク質 (以下、 本 発明のタンパク質と称することもある) は Gタンパク質共役型レセプタータンパ ク質であり、 ヒトゃ温血動物 (例えば、 モルモット、 ラット、 マウス、 ニヮトリ、 ゥサギ、 ブ夕、 ヒッジ、 ゥシ、 サル、 ィヌ、 ネコなど) の細胞 (例えば、 肝細胞、 脾細胞、 神経細胞、 グリア細胞、 塍臓 i3細胞、 骨髄細胞、 メサンギゥム細胞、 ラ ンゲルハンス細胞、 表皮細胞、 上皮細胞、 杯細胞、 内皮細胞、 平滑筋細胞、 繊維 芽細胞、 繊維細胞、 筋細胞、 脂肪細胞、 免疫細胞 (例、 マクロファージ、 T細胞、 B細胞、 ナチュラルキラ一細胞、 肥満細胞、 好中球、 好塩基球、 好酸球、 単球) 、 巨核球、 滑膜細胞、 軟骨細胞、 骨細胞、 骨芽細胞、 破骨細胞、 乳腺細胞、 肝細胞 もしくは間質細胞、 またはこれら細胞の前駆細胞、 幹細胞もしくはガン細胞な ど) もしくはそれらの細胞が存在するあらゆる組織、 例えば、 脳、 脳の各部位 (例、 嗅球、 扁桃核、 大脳基底球、 海馬、 視床、 視床下部、 大脳皮質、 延髄、 小 脳) 、 脊髄、 下垂体、 胃、 滕臓、 腎臓、 肝臓、 生殖腺、 甲状腺、 胆のう、 骨髄、 副腎、 皮膚、 筋肉、 肺、 消化管 (例、 大腸、 小腸) 、 血管、 心臓、 胸腺、 脾臓、 顎下腺、 末梢血、 前立腺、 睾丸、 卵巣、 胎盤、 子宮、 骨、 関節、 骨格筋などに由 来するタンパク質であってもよく、 合成タンパク質であってもよい。 The protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 4, SEQ ID NO: 14 or SEQ ID NO: 24 (hereinafter, may be referred to as the protein of the present invention) is It is a G protein-coupled receptor protein and is used in human warm-blooded animals (eg, guinea pig, rat, mouse, chicken, Cells of egrets, bushes, sheep, sheep, monkeys, monkeys, dogs, cats, etc. Epidermal cells, epithelial cells, goblet cells, endothelial cells, smooth muscle cells, fibroblasts, fiber cells, muscle cells, fat cells, immune cells (eg, macrophages, T cells, B cells, natural killer cells, mast cells, Neutrophils, basophils, eosinophils, monocytes), megakaryocytes, synovial cells, chondrocytes, osteocytes, osteoblasts, osteoclasts, breast cells, hepatocytes or stromal cells, or these cells Progenitor cells, stem cells or cancer cells, etc.) or any tissue in which those cells are present, such as the brain, each part of the brain (eg, olfactory bulb, amygdala, basal sphere, hippocampus, thalamus, hypothalamus) , Cerebral cortex, medulla oblongata, cerebellum), spinal cord, pituitary gland, stomach, ligament, kidney, liver, gonad, thyroid, gall bladder, bone marrow, adrenal gland, skin, muscle, lung, digestive tract (eg, large intestine, small intestine), It may be a protein derived from blood vessels, heart, thymus, spleen, submandibular gland, peripheral blood, prostate, testicle, ovary, placenta, uterus, bone, joint, skeletal muscle, etc., or may be a synthetic protein. .
配列番号: 4で表されるアミノ酸配列と実質的に同一のアミノ酸配列としては、 配列番号: 4で表わされるアミノ酸配列と 9 8 %以上、 好ましくは約 9 9 %以上 の相同性を有するァミノ酸配列などが挙げられる。  The amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 4 includes amino acids having 98% or more, preferably about 99% or more homology with the amino acid sequence represented by SEQ ID NO: 4. And the like.
配列番号: 4で表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有す るタンパク質としては、 例えば、 前記の配列番号: 4で表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有し、 配列番号: 4で表されるアミノ酸配列を 有するタンパク質と実質的に同質の活性を有するタンパク質などが好ましい。  Examples of the protein containing an amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 4 include, for example, a protein containing an amino acid sequence substantially identical to the amino acid sequence represented by the aforementioned SEQ ID NO: 4 However, a protein having substantially the same activity as the protein having the amino acid sequence represented by SEQ ID NO: 4 is preferred.
配列番号: 1 4で表されるアミノ酸配列と実質的に同一のアミノ酸配列として は、 配列番号: 1 4で表わされるアミノ酸配列と 9 2 %以上、 好ましくは約 9 5 %以上、 さらに好ましくは約 9 7 %以上、 より好ましくは約 9 9 %以上の相同 性を有するァミノ酸配列などが挙げられる。  The amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 14 includes 92% or more, preferably about 95% or more, more preferably about 95% or more of the amino acid sequence represented by SEQ ID NO: 14. Amino acid sequences having a homology of 97% or more, more preferably about 99% or more, may be mentioned.
配列番号: 1 4で表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有 - するタンパク質としては、 例えば、 前記の配列番号: 1 4で表されるアミノ酸配 列と実質的に同一のアミノ酸配列を含有し、 配列番号: 1 4で表されるアミノ酸 配列を有するタンパク質と実質的に同質の活性を有するタンパク質などが好まし い。 03 Examples of proteins containing an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 14 include, for example, amino acids substantially the same as the amino acid sequence represented by the aforementioned SEQ ID NO: 14 A protein containing a sequence and having substantially the same activity as a protein having the amino acid sequence represented by SEQ ID NO: 14 is preferred. 03
9  9
配列番号: 2 4で表されるアミノ酸配列と実質的に同一のアミノ酸配列として は、 配列番号: 2 4で表わされるアミノ酸配列と 9 2 %以上、 好ましくは約 9 5 %以上、 さらに好ましくは約 9 7 %以上、 より好ましくは約 9 9 %以上の相同 性を有するアミノ酸配列などが挙げられる。  The amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 24 includes 92% or more, preferably about 95% or more, more preferably about 95% or more of the amino acid sequence represented by SEQ ID NO: 24. Amino acid sequences having a homology of 97% or more, more preferably about 99% or more, and the like can be mentioned.
配列番号': 2 4で表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有 するタンパク質としては、 例えば、 前記の配列番号: 2 4で表されるアミノ酸配 列と実質的に同一のアミノ酸配列を含有し、 配列番号: 2 4で表されるアミノ酸 配列を有するタンパク質と実質的に同質の活性を有するタンパク質などが好まし い。  Examples of proteins having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 24 include, for example, amino acids substantially the same as the amino acid sequence represented by SEQ ID NO: 24 above. A protein containing a sequence and having substantially the same activity as a protein having the amino acid sequence represented by SEQ ID NO: 24 is preferred.
実質的に同質の活性としては、 例えば、 M C Hに対する結合活性、 シグナル情 報伝達作用などが挙げられる。 実質的に同質とは、 それらの活性が性質的に同質 であることを示す。 したがって、 M C Hに対する結合活性やシグナル情報伝達作 用などの活性が同等 (例、 約 0 . 5〜2倍) であることが好ましいが、 これらの 活性の程度やタンパク質の分子量などの量的要素は異なっていてもよい。  Substantially the same activity includes, for example, a binding activity to MCH, a signal information transduction effect, and the like. Substantially the same means that their activities are the same in nature. Therefore, it is preferable that the activities such as the binding activity to MCH and the activity of signal transduction are equivalent (eg, about 0.5 to 2 times), but the quantitative factors such as the degree of these activities and the molecular weight of the protein are It may be different.
結合活性やシグナル情報伝達作用などの活性の測定は、 自体公知の方法または それに準じた方法に従つて行なうことができる。  The activity such as the binding activity and the signal transduction action can be measured according to a method known per se or a method analogous thereto.
また、 本発明のタンパク質としては、 例えば、 (1 ) (i) 配列番号: 4で表 されるアミノ酸配列中の 1または 2個以上 (例えば 1〜8個程度、 好ましくは 1 〜5個程度、 好ましくは 1〜2個) のアミノ酸が欠失したアミノ酸配列、 (i i) 配列番号: 4で表されるアミノ酸配列に 1または 2個以上 (例えば 1〜2 0 0個 程度、 好ましくは 1〜1 5 0個程度、 好ましくは 1〜1 0 0個程度、 好ましくは ;!〜 5 0個程度、 好ましくは 1〜3 0個程度、 好ましくは 1〜1 0個程度、 さら に好ましくは数 (1〜5 ) 偭) のアミノ酸が付加したアミノ酸配列、 (i i i) 配 列番号: 4で表されるアミノ酸配列に 1または 2個以上 (例えば 1〜2 0 0個程 度、 好ましくは 1〜1 5 0個程度、 好ましくは 1〜1 0 0個程度、 好ましくは 1 〜5 0個程度、 好ましくは 1〜3 0個程度、 好ましくは 1〜1 0個程度、 さらに 好ましくは数 (1〜5 ) 個) のアミノ酸が挿入されたアミノ酸配列、 (iv) 配列 番号: 4で表されるアミノ酸配列中の 1または 2個以上 (例えば 1〜2 0 0個程 度、 好ましくは 1〜1 5 0個程度、 好ましくは 1〜1 0 0個程度、 好ましくは 1 〜5 0個程度、 好ましくは 1〜3 0個程度、 好ましくは 1〜1 0個程度、 さらに 好ましくは数 (1〜5 ) 個) のアミノ酸が他のアミノ酸で置換されたアミノ酸配 列、 または (V) それらを組み合わせたアミノ酸配列を含有するタンパク質など のいわゆるムティン、 (2 ) (i) 配列番号: 1 4で表されるアミノ酸配列中の 1または 2個以上 (例えば 1〜2 5個程度、 好ましくは 1〜1 0個程度、 さらに 好ましくは数 (1〜5 ) 個) のアミノ酸が欠失したアミノ酸配列、 (i i) 配列番 号: 1 4で表されるアミノ酸配列に 1または 2個以上 (例えば 1〜2 0 0個程度、 好ましくは 1〜1 5 0個程度、 好ましくは 1〜1 0 0個程度、 好ましくは 1〜 5 0個程度、 好ましくは 1〜3 0個程度、 好ましくは 1〜1 0個程度、 さらに好ま しくは数 (1〜5 ) 個) のアミノ酸が付加したアミノ酸配列、 (i i i) 配列番 号: 1 4で表されるアミノ酸配列に 1または 2個以上 (例えば 1〜2 0 0個程度、 好ましくは 1 ~ 1 5 0個程度、 好ましくは 1〜1 0 0個程度、 好ましくは 1〜5, 0個程度、 好ましくは 1〜3 0個程度、 好ましくは 1〜1 0個程度、 さらに好ま しくは数 (1〜5 ) 個) のアミノ酸が挿入されたアミノ酸配列、 (iv) 配列番 号: 1 4で表されるアミノ酸配列中の 1または 2個以上 (例えば 1〜2 0 0個程 度、 好ましくは 1〜 1 5 0個程度、 好ましくは 1〜 1 0 0個程度、 好ましくは 1 〜5 0個程度、 好ましくは 1〜3 0個程度、 好ましくは 1〜1 0個程度、 さらに 好ましくは数 (1〜5 ) 個) のアミノ酸が他のアミノ酸で置換されたアミノ酸配' 列、 または (V) それらを組み合わせたアミノ酸配列を含有するタンパク質など のいわゆるムティン、 (3 ) (i) 配列番号: 2 4で表されるアミノ酸配列中の 1または 2個以上 (例えば 1〜2 5個程度、 好ましくは 1〜1 0個程度、 さらに 好ましくは数 (1〜5 ) 個) のアミノ酸が欠失したアミノ酸配列、 (i i) 配列番 号: 2 4で表されるアミノ酸配列に 1または 2個以上 (例えば 1〜2 0 0個程度、 好ましくは 1〜1 5 0個程度、 好ましくは 1〜1 0 0個程度、 好ましくは 1〜5 0個程度、 好ましくは 1〜3 0個程度、 好ましくは 1〜1 0個程度、 さらに好ま しくは数 (1〜5 ) 個) のアミノ酸が付加したアミノ酸配列、 (i i i) 配列番 号: 2 4で表されるアミノ酸配列に 1または 2個以上 (例えば 1〜2 0 0個程度、 好ましくは 1〜1 5 0個程度、 好ましくは 1〜1 0 0個程度、 好ましくは 1〜 5 0個程度、 好ましくは 1〜3 0個程度、 好ましくは 1〜1 0個程度、 さらに好ま しくは数 (1〜5 ) 個) のアミノ酸が挿入されたアミノ酸配列、 (iv) 配列番 号: 2 4で表されるアミノ酸配列中の 1または 2個以上 (例えば 1〜2 0 0個程 度、 好ましくは 1〜1 5 0個程度、 好ましくは 1〜1 0 0個程度、 好ましくは 1 〜5 0個程度、 好ましくは 1〜3 0個程度、 好ましくは 1〜1 0個程度、 さらに 好ましくは数 (1〜5 ) 個) のアミノ酸が他のアミノ酸で置換されたアミノ酸配 列、 または (V) それらを組み合わせたアミノ酸配列を含有するタンパク質など のいわゆるムティンも含まれる。 Examples of the protein of the present invention include (1) (i) one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 4 (eg, about 1 to 8, preferably about 1 to 5, (Ii) preferably one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 4 (for example, about 1 to 200, preferably 1 to 1); About 50, preferably about 1 to 100, preferably about;! To about 50, preferably about 1 to 30, preferably about 1 to 10, more preferably a number (1 (5) an amino acid sequence to which the amino acid of (iii) is added; (iii) one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 4 (eg, about 1 to 200, preferably 1 to 15) About 0, preferably about 1 to 100, preferably about 1 to 50, preferably about 1 to 30, preferably 1 to 10 An amino acid sequence into which about, more preferably a number (1 to 5) amino acids have been inserted; (iv) one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 4 (eg, 1 to 200) About 1, preferably about 1 to 150, preferably about 1 to 100, preferably 1 An amino acid sequence in which up to about 50, preferably about 1 to 30, preferably about 1 to 10, and more preferably a number (1 to 5) of amino acids have been substituted with another amino acid; or (V) a so-called mutin such as a protein containing an amino acid sequence obtained by combining them; (2) (i) one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 14 (for example, about 1 to 25 An amino acid sequence in which about 1 to about 10 amino acids have been deleted, more preferably about 1 to 5 amino acids, and (ii) one or two amino acids in the amino acid sequence represented by SEQ ID NO: 14. (For example, about 1 to 200 pieces, preferably about 1 to 150 pieces, preferably about 1 to 100 pieces, preferably about 1 to 50 pieces, preferably about 1 to 30 pieces, preferably Is about 1 to 10 amino acids, more preferably about 1 to 5 amino acids Sequence, (iii) SEQ ID NO: 1 or 2 or more (for example, about 1 to 200, preferably about 1 to 150, preferably 1 to 100) About 1 amino acid, preferably about 1 to 5.0 amino acids, preferably about 1 to 30 amino acids, more preferably about 1 to 10 amino acids, and even more preferably (1 to 5) amino acids. Sequence, (iv) one or more amino acids in the amino acid sequence represented by SEQ ID NO: 14 (for example, about 1 to 200, preferably about 1 to 150, and preferably 1 to 1 About 100, preferably about 1 to 50, preferably about 1 to 30, preferably about 1 to 10, and more preferably the number (1 to 5) of amino acids is other amino acids. (3) (i) a so-called mutin such as a protein containing a substituted amino acid sequence or (V) a protein containing an amino acid sequence combining them; 1 or 2 or more (for example, about 1 to 25, preferably about 1 to 10, and more preferably about 1 to 5) amino acids in the amino acid sequence represented by SEQ ID NO: 24; (Ii) one or two or more amino acid sequences represented by SEQ ID NO: 24 (for example, about 1 to 200, preferably about 1 to 150, and preferably About 100, preferably about 1 to 50, preferably about 1 to 30, preferably about 1 to 10, and more preferably about (1 to 5) amino acids (Iii) one or two or more amino acid sequences represented by SEQ ID NO: 24 (for example, about 1 to 200, preferably about 1 to 150, and preferably 1 to 1 About 100 pieces, preferably about 1 to 50 pieces, preferably about 1 to 30 pieces, preferably about 1 to 10 pieces, and more preferably Or (iv) one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 24 (for example, about 1 to 200 amino acids). Degree, preferably about 1 to 150 pieces, preferably about 1 to 100 pieces, preferably about 1 to 50 pieces, preferably about 1 to 30 pieces, and preferably about 1 to 10 pieces. Preferably, a so-called mutin such as a protein containing an amino acid sequence in which a number (1 to 5) of amino acids are substituted with another amino acid, or (V) an amino acid sequence obtained by combining them is also included.
上記のようにアミノ酸配列が挿入、 欠失または置換されている場合、 その挿入、 欠失または置換の位置は、 とくに限定されない。  When the amino acid sequence is inserted, deleted or substituted as described above, the position of the insertion, deletion or substitution is not particularly limited.
本発明のタンパク質の具体例としては、 例えば、 配列番号: 4で表されるアミ ノ酸配列を含有するタンパク質、 配列番号: 1 4で表されるアミノ酸配列を含有 するタンパク質、 配列番号: 2 4で表されるアミノ酸配列を含有するタンパク質 などがあげられる。  Specific examples of the protein of the present invention include, for example, a protein containing the amino acid sequence represented by SEQ ID NO: 4, a protein containing the amino acid sequence represented by SEQ ID NO: 14, SEQ ID NO: 24 And proteins containing the amino acid sequence represented by
本発明のタンパク質の部分ペプチド (以下、 本発明の部分ペプチドと略記する 場合がある) としては、 前記した本発明のタンパク質の部分ペプチドであって、 好ましくは、 前記した本発明のタンパク質と同様の性質を有するものであればい ずれのものでもよい。  The partial peptide of the protein of the present invention (hereinafter sometimes abbreviated as the partial peptide of the present invention) is a partial peptide of the protein of the present invention described above, and is preferably the same as the above-described protein of the present invention. Any material having properties may be used.
例えば、 本発明のタンパク質の構成アミノ酸配列のうち少なくとも 2 0個以上、 好ましくは 5 0個以上、 さらに好ましくは 7 0個以上、 より好ましくは 1 0 0個 以上、 最も好ましくは 2 0 0個以上のアミノ酸配列を有するペプチドなどが用い られる。  For example, at least 20 or more, preferably 50 or more, more preferably 70 or more, more preferably 100 or more, most preferably 200 or more of the constituent amino acid sequences of the protein of the present invention. For example, a peptide having the amino acid sequence of
また、 本発明の部分ペプチドほ、 そのアミノ酸配列中の 1または 2個以上 (好 ましくは、 1〜1 0個程度、 さらに好ましくは数 (1〜5 ) 個) のアミノ酸が欠 失し、 または、 そのアミノ酸配列に 1または 2個以上 (好ましくは、 1〜2 0個 程度、 より好ましくは 1〜1 0個程度、 さらに好ましくは数 (1〜5 ) 個) のァ ミノ酸が付加し、 または、 そのアミノ酸配列に 1または 2個以上 (好ましくは、 1〜2 0個程度、 より好ましくは 1〜1 0個程度、 さらに好ましくは数 (1〜  In addition, the partial peptide of the present invention lacks one or more (preferably about 1 to 10, more preferably, about 1 to 5) amino acids in its amino acid sequence, Alternatively, one or more (preferably, about 1 to 20, more preferably, about 1 to 10, and more preferably, about 1 to 5) amino acids are added to the amino acid sequence. Or 1 or 2 or more amino acids in the amino acid sequence thereof (preferably, about 1 to 20; more preferably, about 1 to 10;
5 ) 個) のアミノ酸が揷入され、 または、 そのアミノ酸配列中の 1または 2個以 上 (好ましくは、 1〜1 0個程度、 より好ましくは数個、 さらに好ましくは 1〜 5個程度) のァミノ酸が他のアミノ酸で置換されていてもよい。 5) amino acids, or one or more (preferably about 1 to 10, more preferably several, and more preferably 1 to (About 5 amino acids) may be substituted with another amino acid.
本発明の部分ペプチドとしては、 例えば、 配列番号: 4で表されるアミノ酸配 列において例えば、 第 1番目〜 4 1番目、 第 3 1 1番目〜 3 4 0番目のアミノ酸 配列、 配列番号: 1 4で表されるアミノ酸配列において例えば、 第 1番目〜 4 1 番目、 第 3 1 1番目〜 3 4 0番目のアミノ酸配列、 配列番号: 2 4で表されるァ ミノ酸配列において例えば、 第 1番目〜 4 1番目、 第 3 1 1番目〜 3 3 0番目の ァミノ酸配列などが好ましい。  As the partial peptide of the present invention, for example, in the amino acid sequence represented by SEQ ID NO: 4, for example, the 1st to 41st amino acids, the 31st to 34th amino acids, SEQ ID NO: 1 For example, in the amino acid sequence represented by the amino acid sequence represented by SEQ ID NO: 24, the 1st to 41st amino acid sequence, the 31st amino acid sequence to the 34th amino acid sequence, SEQ ID NO: 24 The amino acid sequences of the 1st to 41st and the 31st to 330th amino acids are preferred.
配列番号: 2 5で表わされるアミノ酸配列と同一もしくは実質的に同一のアミ ノ酸配列を含有するペプチド (以下、 本発明で用いられるペプチドまたは本発明 のペプチドと称することもある) は、 ヒトゃ温血動物 (例えば、 モルモット、 ラ ット、 マウス、 ニヮトリ、 ゥサギ、 ブタ、 ヒッジ、 ゥシ、 サル、 ィヌ、 ネコな ど) の細胞 (例えば、 肝細胞、 脾細胞、 神経細胞、 グリア細胞、 腠臓 ]3細胞、 骨 髄細胞、 メサンギゥム細胞、 ランゲル八ンス細胞、 表皮細胞、 上皮細胞、 杯細胞、 内皮細胞、 平滑筋細胞、 繊維芽細胞、 繊維細胞、 筋細胞、 脂肪細胞、 免疫細胞 (例、 マクロファージ、 T細胞、 B細胞、 ナチュラルキラー細胞、 肥満細胞、 好 中球、 好塩基球、 好酸球、 単球) 、 巨核球、 滑膜細胞、 軟骨細胞、 骨細胞、 骨芽 細胞、 破骨細胞、 乳腺細胞、 肝細胞もしくは間質細胞、 またはこれら細胞の前駆 細胞、 幹細胞もしくはガン細胞など) もしくはそれらの細胞が存在するあらゆる 組織、 例えば、 脳、 脳の各部位 (例、 嗅球、 扁桃核、 大脳基底球、 海馬、 視床、 視床下部、 大脳皮質、 延髄、 小脳) 、 脊髄、 下垂体、 胃、 塍臓、 腎臓、 肝臓、 生 殖腺、 甲状腺、 胆のう、 骨髄、 副腎、 皮膚、 筋肉、 肺、 消化管 (例、 大腸、 小 腸) 、 血管、 心臓、 胸腺、 脾臓、 顎下腺、 末梢血、 前立腺、 睾丸、 卵巣、 胎盤、 子宮、 骨、 関節、 骨格筋などに由来するタンパク質であってもよく、 合成タンパ ク質であってもよい。  A peptide containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 25 (hereinafter sometimes referred to as the peptide used in the present invention or the peptide of the present invention) is a human Cells of warm-blooded animals (eg, guinea pigs, rats, mice, chicks, egrets, pigs, higgies, lions, monkeys, dogs, cats, etc.) (eg, hepatocytes, spleen cells, nerve cells, glial cells) 3 cells, bone marrow cells, mesangial cells, Langer's cells, epidermal cells, epithelial cells, goblet cells, endothelial cells, smooth muscle cells, fibroblasts, fiber cells, muscle cells, adipocytes, immune cells (Eg, macrophages, T cells, B cells, natural killer cells, mast cells, neutrophils, basophils, eosinophils, monocytes), megakaryocytes, synovial cells, chondrocytes, bone Cells, osteoblasts, osteoclasts, mammary cells, hepatocytes or stromal cells, or precursors of these cells, stem cells or cancer cells) or any tissue in which these cells are present, for example, brain, brain Anatomy (eg, olfactory bulb, amygdala, basal sphere, hippocampus, thalamus, hypothalamus, cerebral cortex, medulla, cerebellum), spinal cord, pituitary, stomach, kidney, kidney, liver, gonad, thyroid, gall bladder, Bone marrow, adrenal gland, skin, muscle, lung, digestive tract (eg, large intestine, small intestine), blood vessels, heart, thymus, spleen, submandibular gland, peripheral blood, prostate, testicle, ovary, placenta, uterus, bone, joint, It may be a protein derived from skeletal muscle or the like, or may be a synthetic protein.
配列番号: 2 5で表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有 するペプチドとしては、 例えば、 配列番号: 2 5で表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有し、 配列番号: 2 5で表されるアミノ酸配列を有 するペプチドと実質的に同質の活性を有するペプチドなどが好ましい。  Examples of the peptide having an amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 25 include, for example, an amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 25 Peptides having substantially the same activity as the peptide having the amino acid sequence represented by SEQ ID NO: 25 are preferred.
'実質的に同質の活性としては、 例えば、 S L Tに対する結合活性などが挙げら れる。 実質的に同質とは、 その活性が性質的に同質であることを示す。 したがつ て、 S L Tに対する結合活性が同等 (例、 約 0 . 5〜2倍) であることが好まし いが、 この活性の程度やタンパク質の分子量などの量的要素は異なっていてもよ い。 'Substantially equivalent activities include, for example, SLT binding activity, etc. It is. Substantially the same indicates that the activity is the same in nature. Therefore, it is preferable that the binding activity to SLT is equivalent (eg, about 0.5 to 2 times), but quantitative factors such as the degree of this activity and the molecular weight of the protein may be different. No.
結合活性の測定は、 自体公知の方法に準じて行なうことができる。  The binding activity can be measured according to a method known per se.
本発明のペプチドとしては、 配列番号: 2 5で表されるアミノ酸配列を含有す るペプチドなどが好ましく用いられる。  As the peptide of the present invention, a peptide containing the amino acid sequence represented by SEQ ID NO: 25 is preferably used.
また、 本発明のペプチドとしては、 例えば、 (i) 配列番号: 2 5で表される アミノ酸配列中の 1または 2個以上 (例えば 1〜3 0個程度、 好ましくは 1〜1 0個程度、 さらに好ましくは数 (1〜5 ) 個) のアミノ酸が欠失したアミノ酸配 列、 (i i) 配列番号: 2 5で表されるアミノ酸配列に 1または 2個以上 (例えば 1〜3 0個程度、 好ましくは 1〜1 0個程度、 さらに好ましくは数 (1〜5 ) 個) のアミノ酸が付加したアミノ酸配列、 (i i i) 配列番号: 2 5で表されるァ ミノ酸配列に 1または 2個以上 (例えば 1〜3 0個程度、 好ましくは 1〜1 0個 程度、 さらに好ましくは数 (1〜5 ) 個) のアミノ酸が揷入されたアミノ酸配列、 (iv) 配列番号: 2 5で表されるアミノ酸配列中の 1または 2個以上 (例えば 1 〜3 0個程度、 好ましくは 1 ~ 1 0個程度、'さらに好ましくは数 (1〜5 ) 個) のアミノ酸が他のアミノ酸で置換されたアミノ酸配列、 または (V) それらを組 み合わせたアミノ酸配列を含有するべプチドも含まれる。  Examples of the peptide of the present invention include: (i) one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 25 (for example, about 1 to 30, preferably about 1 to 10; More preferably, an amino acid sequence in which a number (1 to 5) of amino acids are deleted, and (ii) one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 25 (for example, about 1 to 30 amino acids, Preferably about 1 to 10 amino acids, more preferably about 1 to 5 amino acids, and (iii) one or more amino acid sequences in the amino acid sequence represented by SEQ ID NO: 25. (For example, about 1 to 30 amino acids, preferably about 1 to 10 amino acids, and more preferably about 1 to 5 amino acids). (Iv) SEQ ID NO: 25 1 or 2 or more in the amino acid sequence (for example, about 1 to 30, preferably about 1 to 10, more preferably Preferably, a peptide containing an amino acid sequence in which a number (1 to 5) of amino acids are substituted with another amino acid, or (V) an amino acid sequence obtained by combining them is also included.
上記のように'アミノ酸配列が挿入、 欠失または置換されている場合、 その挿入、 欠失または置換の位置は、 とくに限定されない。  When the amino acid sequence is inserted, deleted or substituted as described above, the position of the insertion, deletion or substitution is not particularly limited.
具体的には、 配列番号: 2 5で表されるアミノ酸配列の N末端から第 5番目〜 第 1 9番目の部分配列を含有するペプチドなどがあげられる。 より具体的には、 配列番号: 2 6、 配列番号: 2 7、 配列番号: 2 8、 配列番号: 2 9、 配列番 号: 3 0または配列番号: 3 1で表わされるアミノ酸配列を有するぺプチドなど があげられる。 好ましくは配列番号: 2 8で表されるアミノ酸配列を有するぺプ チドなどが用いられる。  Specific examples include a peptide containing the 5th to 19th partial sequences from the N-terminal of the amino acid sequence represented by SEQ ID NO: 25. More specifically, it has the amino acid sequence represented by SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30 or SEQ ID NO: 31. Peptides are examples. Preferably, a peptide having the amino acid sequence represented by SEQ ID NO: 28 is used.
該アミノ酸配列中のアミノ酸の実質的に同一な置換物としては、 たとえばその アミノ酸が属するところのクラスのうち他のアミノ酸類から選ぶことができうる。 非極性 (疎水性) アミノ酸としては、 ァラニン、 ロイシン、 イソロイシン、 バリ ン、 プロリン、 フエ二ルァラニン、 トリブトファン、 メチォニンなどがあげられ る。 極性 (中性) アミノ酸としてはグリシン、 セリン、 スレオニン、 システィン、 チロシン、 ァスパラギン、 グルタミンなどがあげられる。,陽電荷をもつ (塩基 性) ァ.ミノ酸としてはアルギニン、 リジン、 ヒスチジンなどがあげられる。 負電 荷をもつ (酸性) アミノ酸としては、 ァスパラギン酸、 グルタミン酸などがあげ られる。 ' Substantially identical substitutions of amino acids in the amino acid sequence may be selected, for example, from other amino acids of the class to which the amino acid belongs. Non-polar (hydrophobic) amino acids include alanine, leucine, isoleucine, valin, proline, phenylalanine, tributofan, methionine, and the like. Polar (neutral) amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine. , Positively charged (basic) amino acids include arginine, lysine and histidine. Negatively charged (acidic) amino acids include aspartic acid and glutamic acid. '
上記する他のアミノ酸が欠失、 置換する位置としては、 本発明のペプチドの構 成アミノ酸中、 Cy s以外の位置であることが好ましい。  The position at which the above-mentioned other amino acid is deleted or substituted is preferably a position other than Cys in the amino acids constituting the peptide of the present invention.
さらに、 本発明のペプチドは標識化されていてもよい。 具体例としては、 自体 公知の方法でァイソトーブラベル化されたペプチド、 蛍光標識されたペプチド (例えば、 フルォレセインなどによる蛍光標識) 、 ピオチン化されたペプチド、 酵素標識されたぺプチドなどがあげられる。  Further, the peptide of the present invention may be labeled. Specific examples include peptides labeled with an isotope by a method known per se, fluorescently labeled peptides (for example, fluorescently labeled with fluorescein), biotinylated peptides, enzyme-labeled peptides, and the like. .
具体的には、 例えば公知の方法によって、 〔¾〕 、 (1251] 、 〔l4C〕 、 〔35S〕 な どで標識された本発明のペプチドなどを利用することができる。 また、 ポルトン 一ハンター試薬を用いて公知の方法で調製した本発明のペプチドの標識体を用い てもよい。 Specifically, for example, by known methods, [¾], (125 1], it can be utilized, such as a peptide of [l4 C], the present invention labeled with etc. [35 S]. Moreover, Porton A labeled product of the peptide of the present invention prepared by a known method using one Hunter reagent may be used.
標識化された本発明のぺプチドの具体例としては、 例えば、  Specific examples of the labeled peptide of the present invention include, for example,
(1) [1251 • [N- (3- (4-ヒドロキシ - 3_ョ ドフエニル)プ口ピオニル) -Asp1] . MCH、 (1) [ 125 1 • [N- (3- (4-hydroxy-3_dodophenyl) -pulpionyl) -Asp 1 ] .MCH,
(2) [I25I [N -(3- (4-ヒドロキシ -3 -ョ一ドフエニル)プロピオ二ル)- Phe2]' MCH(2- 19)、 (2) [I25 I [N - (3- (4- hydroxy-3 - ® one Dofueniru) propionic sulfonyl) - Phe 2] 'MCH ( 2- 19),
(3) [125I - [N- (3- (4 -ヒドロキシ- 3-ョードフエニル)プロピオ二ル)- Asp3]' MCH(3- 19)、 (3) [ 125 I- [N- (3- (4-hydroxy-3-iodophenyl) propionyl) -Asp 3 ] ′ MCH (3-19),
(4) [125I —[N -(3 - (4 -ヒドロキシ -3-ョ一ドフエニル)プロピオニル) -Met4]' MCH(4- 19)、 (4) [ 125 I— [N- (3- (4-hydroxy-3-iodophenyl) propionyl) -Met 4 ] ′ MCH (4-19),
(5) [125I — [N-(3 -(4-ヒドロキシ- 3-ョードフエニル)プロピオニル) -Leu5]. MCH(5- 19)、 (5) [ 125 I — [N- (3- (4-hydroxy-3-iodophenyl) propionyl) -Leu 5 ]. MCH (5-19),
(6) [125I - [N- (3- (4-ヒドロキシ- 3-ョードフエニル)プ口ピォニル) -Arg6]■ MCH (6-19)、 (6) [ 125 I-[N- (3- (4-hydroxy-3-iodophenyl) -pulpionyl) -Arg 6 ] ■ MCH (6-19),
( 7 ) [I25I] 一 [N_ (3— (4—ヒドロキシ— 3 -ョードフエニル)プロピオニル) -Cys7] - MCH (7-19) などがあげられる。 ' (7) [I25 I] one [N_ (3- (4- hydroxy - 3 - Yodofueniru) propionyl) -Cys 7] - such as MCH (7-19) can be mentioned. '
なかでも、 [|251] - [N- (3- (4-ヒドロキシ- 3-ョードフエニル)プロピオニル) - Met4] -MCH (4 - 19)が好ましく用いられる。 Of these, [ | 251]-[N- (3- (4-hydroxy-3-iodophenyl) propionyl) -Met 4 ] -MCH (4-19) is preferably used.
本明細書におけるタンパク質およびペプチドは、 ペプチド標記の慣例に従って 左端が N末端 (ァミノ末端) 、 右端が C末端 (力ルポキシル末端) である。 本発 明のタンパク質およびペプチドは、 C末端がカルボキシル基 (-C00H) 、 カルボ キシレート (-C00—) 、 アミド (- C0M2) またはエステル (- C00R) のいずれであ つてもよい。 In the present specification, the left end of the protein and the peptide is the N-terminus (amino end) and the right end is the C-terminus (potassium terminal) according to the convention of peptide labeling. This onset Ming proteins and peptides, C-terminal, carboxyl group (-C00H), carbo Kishireto (-C00-), amide (- C0M 2) or ester (- C00R) any der connexion may be of.
ここでエステルにおける Rとしては、 例えば、 メチル、 ェチル、 n—プロピル、 イソプロピル、 n -ブチルなどの C ,_6アルキル基、 例えば、 シクロペンチル、 シ クロへキシルなどの〇38シクロアルキル基、 例えば、 フエニル、 α—ナフチルな どの C 6_127リール基、 例えば、 ベンジル、 フエネチルなどのフエニル— C ,_2アル キル基もしくはひ一ナフチルメチルなどの α—ナフチルー アルキル基などの C7_14ァラルキル基、 ビバロイルォキシメチル基などが用いられる。 Here, as R in the ester, e.g., methyl, Echiru, n- propyl, isopropyl, n - C, such as butyl, _ 6 alkyl groups, such as cyclopentyl, such as hexyl cyclo 〇 3 - 8 cycloalkyl group, for example, phenyl, alpha-naphthyl of which C 6 _ 12 7 aryl group, e.g., benzyl, phenyl, such as phenethyl - C, C 7 _ such as alpha-Nafuchiru alkyl groups such as _ 2 Al kill group or carry one naphthylmethyl A 14 aralkyl group, a bivaloyloxymethyl group and the like are used.
本発明のタンパク質およびペプチドが C末端以外に力ルポキシル基 (または力 ルポキシレート) を有している場合、 カルボキシル基がアミド化またはエステル 化されているものも本発明のタンパク質およびペプチドに含まれる。 この場合の エステルとしては、 例えば上記した C末端のエステルなどが用いられる。  When the protein and peptide of the present invention have a lipoxyl group (or lipoxylate) other than at the C-terminus, those in which the carboxyl group is amidated or esterified are also included in the proteins and peptides of the present invention. As the ester in this case, for example, the above-mentioned C-terminal ester and the like are used.
さらに、 本発明のタンパク質およびペプチドには、 N末端のアミノ酸残基 (例、 メチォニン残基) のァミノ基が保護基 (例えば、 ホルミル基、 ァセチル基などの アルカノィルなどの C Mァシル基など) で保護されているもの、 生体内で切 断されて生成する N末端のグルタミン残基がピログルタミン酸化したもの、 分子 内のアミノ酸の側鎖上の置換基 (例えば- 0H、 -SH、 アミノ基、 イミダゾ一ル基、 インドール基、 グァニジノ基など) が適当な保護基 (例えば、 ホルミル基、 ァセ チル基などの アルカノィル基などの c ,_6ァシル基など) で保護されているも の、 あるいは糖鎖が結合したいわゆる糖タンパク質などの複合タンパク質なども 含まれる。 また、 本発明の部分ペプチドは、 C末端が力ルポキシル基 (-C00H) 、 力ルポ - キシレート (-C00—) 、 アミド (- C0NH2) またはエステル (-C00R) の何れであつ てもよい。 Furthermore, in the proteins and peptides of the present invention, the amino acid residues at the N-terminus (eg, Mechionin residues) Amino group protecting groups (e.g., formyl group, etc. C M Ashiru group such Arukanoiru such Asechiru group) Protected, N-terminal glutamine residue generated by cleavage in vivo, pyroglutamine oxidation, Substituent on the side chain of amino acid in the molecule (for example, -OH, -SH, amino group, imidazo Ichiru group, indole group, etc. Guanijino group) is also protected with a suitable protecting group (e.g., formyl group, etc. c, _ 6 Ashiru groups such Arukanoiru groups such as § Se butyl group), or Also included are complex proteins such as so-called glycoproteins to which sugar chains are bound. In the partial peptide of the present invention, C-terminal, the force Rupokishiru group (-C00H), the force Lupo - Kishireto (-C00-), amide (- C0Nh 2) or may be filed in any of ester (-C00R).
さらに、 本発明の部分ペプチドには、 前記した本発明のタンパク質と同様に、 C末端以外に力ルポキシル基 (またはカルポキシレート) を有しているもの、 N 末端のアミノ酸残基 (例、 メチォニン残基) のァミノ基が保護基で保護されてい るもの、 N端側が生体内で切断され生成したグルタミン残基がピログルタミン酸 化したもの、 分子内のアミノ酸の側鎖上の置換基が適当な保護基で保護されてい るもの、 あるいは糖鎖が結合したいわゆる糖ペプチドなどの複合べプチドなども 含まれる。  Furthermore, the partial peptide of the present invention includes, as in the case of the protein of the present invention, those having a carbonyl group (or carboxylate) other than the C-terminus, N-terminal amino acid residues (eg, methionine). Residue is protected with a protecting group, the glutamine residue formed by cleavage of the N-terminal in vivo is a pyroglutamate, and the substituent on the side chain of the amino acid in the molecule is appropriate. Also included are those protected with a protecting group or complex peptides such as so-called glycopeptides to which sugar chains are bonded.
本発明のタンパク質または部分ペプチド、 本発明のペプチドの塩としては、 生 理学的 ίこ許容される酸 (例、 無機酸、 有機酸) や塩基 (例、 アルカリ金属塩) な どとの塩が用いられ、 とりわけ生理学的に許容される酸付加塩が好ましい。 この 様な塩としては、 例えば、 無機酸 (例えば、 塩酸、 リン酸、 臭化水素酸、 硫酸) との塩、 あ'るいは有機酸 (例えば、 酢酸、 ギ酸、 プロピオン酸、 フマル酸、 マレ イン酸、 コハク酸、 酒石酸、 クェン酸、 リンゴ酸、 蓚酸、 安息香酸、 メタンスル ホン酸、 ベンゼンスルホン酸) との塩などが用いられる。  Salts of the protein or partial peptide of the present invention and salts of the peptide of the present invention include salts with physiologically acceptable acids (eg, inorganic acids, organic acids) and bases (eg, alkali metal salts). Preference is given to the use of, especially the physiologically acceptable acid addition salts. Such salts include, for example, salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid), and organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid) Salts with formic acid, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid) are used.
本発明のタンパク質もしくはその部分べプチド、 本発明のぺプチドまたはそれ らの塩は、 前述したヒトゃ温血動物の細胞または組織から公知のタンパク質の精 製方法によって製造することもできるし、 タンパク質をコードする D N Aを含有 する形質転換体を培養することによつても製造することができる。 また、 後述の ぺプチド合成法に準じて製造することもできる。  The protein of the present invention or a partial peptide thereof, the peptide of the present invention or a salt thereof can be produced from the above-mentioned human or warm-blooded animal cell or tissue by a known protein purification method. Can also be produced by culturing a transformant containing DNA encoding It can also be produced according to the peptide synthesis method described below.
ヒトゃ哺乳動物の組織または細胞から製造する場合、 ヒトゃ哺乳動物の組織ま たは細胞をホモジナイズした後、 酸などで抽出を行ない、 該抽出液を逆相クロマ トグラフィ一、 イオン交換クロマトグラフィーなどのクロマトグラフィーを組み ' 合わせることにより精製単離することができる。  When producing from human or mammalian tissues or cells, the human or mammalian tissues or cells are homogenized and then extracted with an acid or the like, and the resulting extract is subjected to reverse phase chromatography, ion exchange chromatography, etc. Purification and isolation can be achieved by combining the above chromatography.
本発明のぺプチドまたは本発明のタンパク質もしくはその部分べプチドまたは それらのアミド体またはそれらの塩の合成には、 通常市販のタンパク質 ·ぺプチ ド合成用樹脂を用いることができる。 そのような樹脂としては、 例えば、 クロ口 メチル樹脂、 ヒドロキシメチル樹脂、 ベンズヒドリルァミン樹脂、 アミノメチル 樹脂、 4一べンジルォキシベンジルアルコール樹脂、 4一メチルベンズヒドリル ァミン樹脂、 P AM樹脂、 4—ヒドロキシメチルメチルフエニルァセトアミドメ チル樹脂、 ポリアクリルアミド樹脂、 4— ( 2 ' , 4 '—ジメトキシフエ二ルーヒ ドロキシメチル) フエノキシ樹脂、 4一 (2 ' , 4 '—ジメトキシフエ二ルー Fm o cアミノエチル) フエノキシ樹脂などを挙げることができる。 このような樹脂 を用い、 α—ァミノ基と側鎖官能基を適当に保護したアミノ酸を、 目的とする夕 ンパク質 ·ペプチドの配列通りに、 自体公知の各種縮合方法に従い、 樹脂上で縮 合させる。 反応の最後に樹脂からタンパク質 ·ペプチドを切り出すと同時に各種 保護基を除去し、 さらに高希釈溶液中で分子内ジスルフイド結合形成反応を実施 し、 目的のタンパク質 ·ペプチドまたはそれらのアミド体を取得する。 For the synthesis of the peptide of the present invention, the protein of the present invention, its partial peptide, their amides or their salts, commercially available resins for protein / peptide synthesis can be used. As such a resin, for example, Methyl resin, hydroxymethyl resin, benzhydrylamine resin, aminomethyl resin, 4-benzyloxybenzyl alcohol resin, 4-methylbenzhydrylamine resin, PAM resin, 4-hydroxymethylmethylphenylacetamide Methyl resin, polyacrylamide resin, 4- (2 ', 4'-dimethoxyphenylhydroxymethyl) phenoxy resin, 41- (2', 4'-dimethoxyphenyl Fmocaminoethyl) phenoxy resin, etc. Can be. Using such a resin, an amino acid appropriately protected with an α-amino group and a side chain functional group is condensed on the resin in accordance with the sequence of the desired protein / peptide according to various known condensation methods. Let it. At the end of the reaction, proteins and peptides are cleaved from the resin, and at the same time, various protecting groups are removed. Further, an intramolecular disulfide bond formation reaction is performed in a highly diluted solution to obtain the desired protein, peptide or amide thereof.
上記した保護アミノ酸の縮合に関しては、 タンパク質 ·ペプチド合成に使用で きる各種活性化試薬を用いることができるが、 特に、 カルポジイミド類がよい。 ' カルポジイミド類としては、 D C C、 N, N '—ジイソプロピルカルポジイミド. N—ェチル— N'— (3—ジメチルァミノプロリル) カルポジイミドなどが用い られる。 これらによる活性化にはラセミ化抑制添加剤 (例えば、 HO B t , H O O B t ) とともに保護アミノ酸を直接樹脂に添加するかまたは、 対称酸無水物ま たは H O B tエステルあるいは H O O B tエステルとしてあらかじめ保護アミノ 酸の活性化を行なった後に樹脂に添加することができる。  Regarding the condensation of the protected amino acids described above, various activating reagents that can be used for protein / peptide synthesis can be used, and carbodiimides are particularly preferable. As the carbodiimides, DCC, N, N'-diisopropyl carbopimide. N-ethyl-N '-(3-dimethylaminoprolyl) carbopimide is used. For these activations, the protected amino acid may be added directly to the resin along with a racemization inhibitor additive (eg, HOBt, HOOBt) or pre-protected as a symmetrical acid anhydride or HOBt ester or HOOBt ester. It can be added to the resin after activation of the amino acid.
保護アミノ酸の活性化や樹脂との縮合に用いられる溶媒としては、 タンパク 質 ·ペプチド縮合反応に使用しうることが知られている溶媒から適宜選択されう る。 例えば、 N, N—ジメチルホルムアミド, N, N—ジメチルァセトアミド, N—メチルピロリドンなどの酸アミド類、 塩化メチレン, クロ口ホルムなどのハ ロゲン化炭化水素類、 トリフルォロエタノールなどのアルコール類、 ジメチルス ルホキシドなどのスルホキシド類、 ピリジン, ジォキサン, テトラヒドロフラン などのェ一テル類、 ァセトニトリル, プロピオ二トリルなどの二トリル類、 酢酸 メチル, 酢酸ェチルなどのエステル類あるいはこれらの適宜の混合物などが用い られる。 反応温度はタンパク質結合形成反応に使用され得ることが知られている 範囲から適宜選択され、 通常約一 2 0 ° (:〜 5 O t:の範囲から適宜選択される。 活 性化されたアミノ酸誘導体は通常 1 . 5〜4倍過剰で用いられる。 ニンヒドリン 反応を用いたテストの結果、 縮合が不十分な場合には保護基の脱離を行うことな く縮合反応を繰り返すことにより十分な縮合を行なうことができる。 反応を繰り 返しても十分な縮合が得られないときには、 無水酢酸またはァセチルイミダゾ一 ルを用いて未反応アミノ酸をァセチル化することができる。 The solvent used for activating the protected amino acid or for condensing with the resin may be appropriately selected from solvents known to be usable for the protein-peptide condensation reaction. For example, acid amides such as N, N-dimethylformamide, N, N-dimethylacetamide, N-methylpyrrolidone, halogenated hydrocarbons such as methylene chloride and chloroform, alcohols such as trifluoroethanol , Sulfoxides such as dimethyl sulfoxide, ethers such as pyridine, dioxane, and tetrahydrofuran; nitriles such as acetonitrile and propionitrile; esters such as methyl acetate and ethyl acetate; or an appropriate mixture thereof. Can be The reaction temperature is appropriately selected from a range known to be usable for a protein bond formation reaction, and is usually appropriately selected from a range of about 120 ° (: to 5 Ot :). The sensitized amino acid derivative is usually used in a 1.5 to 4-fold excess. As a result of the test using the ninhydrin reaction, when the condensation is insufficient, sufficient condensation can be performed by repeating the condensation reaction without removing the protecting group. When a sufficient condensation cannot be obtained by repeating the reaction, the unreacted amino acid can be acetylated using acetic anhydride or acetylimidazole.
原料のァミノ基の保護基としては、 例えば、 Z、 B o c、 夕一シャリーペンチ ルォキシカルポニル、 ィソポルニルォキシカルポニル、 4ーメトキシベンジルォ キシカルボニル、 C 1 一 Z、 B r— Z、 ァダマンチルォキシカルポニル、 トリフ ルォロアセチル、 フタロイル、 ホルミル、 2—二トロフエニルスルフエニル、 ジ フエニルホスフイノチオイル、 Fm o c よどが用いられる。  Examples of the protecting group for the amino group of the starting material include Z, Boc, Yuichi Sharipentyloxycarbonyl, Isoporonyloxycarbonyl, 4-methoxybenzyloxycarbonyl, C11Z, Br-Z , Adamantyloxycarbonyl, trifluoroacetyl, phthaloyl, formyl, 2-ditrophenylsulfenyl, diphenylphosphinothioyl, Fmoc and the like are used.
力ルポキシル基は、 例えば、 アルキルエステル化 (例えば、 メチル、 ェチル、 プロピル、 プチル、 ターシャリーブチル、 シクロペンチル、 シクロへキシル、 シ クロへプチル、 シクロォクチル、 2ーァダマンチルなどの直鎖状、 分枝状もしく は環状アルキルエステル化) 、 ァラルキルエステル化 (例えば、 ベンジルエステ ル、 4一二トロべンジルエステル、 4ーメトキシベンジルエステル、 4一クロ口 ベンジルエステル、 ベンズヒドリルエステル化) 、 フエナシルエステル化、 ベン ジルォキシカルポニルヒドラジド化、 ターシャリ一ブトキシカルポニルヒドラジ ド化、 トリチルヒドラジド化などによって保護することができる。  Lepoxyl groups can be, for example, alkyl esterified (e.g., methyl, ethyl, propyl, butyl, tert-butyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, 2-adamantyl, etc.). Or cyclic alkyl esterification), aralkyl esterification (e.g., benzyl ester, 412 trobenzyl ester, 4-methoxybenzyl ester, 4-methyl benzyl ester, benzhydryl esterification), phenacyl esterification , Benzyloxycarbonyl hydrazide, tert-butoxycarbonyl hydrazide, trityl hydrazide and the like.
セリンの水酸基は、 例えば、 エステル化またはエーテル化によって保護するこ とができる。 このエステル化に適する基としては、 例えば、 ァセチル基などの低 級アルカノィル基、 ベンゾィル基などのァロイル基、 ベンジルォキシカルポニル 基、 エトキシカルポニル基などの炭酸から誘導される基などが用いられる。 また、 エーテル化に適する基としては、 例えば、 ベンジル基、 テトラヒドロビラニル基、 t -ブチル基などである。  The hydroxyl group of serine can be protected, for example, by esterification or etherification. As a group suitable for this esterification, for example, a lower alkanol group such as an acetyl group, an aroyl group such as a benzoyl group, a group derived from carbonic acid such as a benzyloxycarbonyl group, an ethoxycarponyl group, and the like are used. Examples of a group suitable for etherification include a benzyl group, a tetrahydroviranyl group, and a t-butyl group.
チロシンのフエノール性水酸基の保護基としては、 例えば、 B z l、 C 1 2 -The protecting group of the phenolic hydroxyl group of tyrosine, for example, B zl, C 1 2 -
B z l、 2—二トロベンジル、 B r— Z、 夕一シャリーブチルなどが用いられる。 ヒスチジンのイミダゾールの保護基としては、 例えば、 T o s、 4ーメトキシ 一 2 , 3, 6—トリメチルベンゼンスルホニル、 D N P、 ベンジルォキシメチル、 B u m, B o c、 T r t、 F m o cなどが用いられる。 原料の力ルポキシル基の活性化されたものとしては、 例えば、 対応する酸無水 物、 アジド、 活性エステル 〔アルコール (例えば、 ペンタクロロフエノール、 2 4 , 5—トリクロ口フエノール、 2 , 4—ジニトロフエノール、 シァノメチルァ ルコール、 パラニトロフエノール、 HO N B、 N—ヒドロキシスクシミド、 N— ヒドロキシフタルイミド、 H O B t ). とのエステル〕 などが用いられる。 原料の ァミノ基の活性化されたものとしては、 例えば、 対応するリ.ン酸アミドが用いら れる。 Bzl, 2-nitrobenzyl, Br-Z, Yuichi Sharybutyl and the like are used. As the protecting group for imidazole of histidine, for example, Tos, 4-methoxy-12,3,6-trimethylbenzenesulfonyl, DNP, benzyloxymethyl, Bum, Boc, Trt, Fmoc and the like are used. Examples of activated carboxylic acid groups of the raw material include, for example, corresponding acid anhydrides, azides, and activated esters [alcohols (for example, pentachlorophenol, 24,5-trichloromouth phenol, 2,4-dinitrophenol) , Cyanomethyl alcohol, paranitrophenol, HONB, N-hydroxysuccinimide, N-hydroxyphthalimide, and esters with HOBt). As the activated amino group of the raw material, for example, a corresponding phosphoric acid amide is used.
保護基の除去 (脱離) 方法としては、 例えば、 P d—黒あるいは P d—炭素な どの触媒の存在下での水素気流中での接触還元や、 また、 無水フッ化水素、 メタ ンスルホン酸、 トリフルォロメタンスルホン酸、 トリフルォロ酢酸あるいはこれ らの混合液などによる酸処理や、 ジィソプロピルェチルァミン、 トリェチルァミ ン、 ピぺリジン、 ピぺラジンなどによる塩基処理、 また液体アンモニア中ナトリ ゥムによる還元なども用いられる。 上記酸処理による脱離反応は、 一般に約一 2 0 ° (:〜 4 0 °Cの温度で行なわれるが、 酸処理においては、 例えば、 ァニソール、 フエノール、 チオアニソ一ル、 メタクレゾール、 パラクレゾ一ル、 ジメチルスル フイド、 1 , 4 -ブタンジチオール、 1 , 2—エタンジチオールなどのようなカチォ ン捕捉剤の添加が有効である。 また、 ヒスチジンのイミダゾール保護基として用 いられる 2 , 4—ジニトロフエニル基はチォフエノール処理により除去され、 ト リブトファンのィンドール保護基として用いられるホルミル基は上記の 1 , 2— エタンジチオール、 1, 4—ブタンジチオールなどの存在下の酸処理による脱保 護以外に、 希水酸化ナトリウム溶液、 希アンモニアなどによるアルカリ処理によ つても除去される。  Methods for removing (eliminating) the protecting group include, for example, catalytic reduction in a hydrogen stream in the presence of a catalyst such as Pd-black or Pd-carbon, or hydrogen fluoride anhydride or methanesulfonic acid. Acid treatment with trifluoromethanesulfonic acid, trifluoroacetic acid, or a mixture thereof; base treatment with diisopropylethylamine, triethylamine, piperidine, piperazine, etc .; Reduction by a system is also used. The elimination reaction by the above acid treatment is generally carried out at a temperature of about 120 ° C. (: up to 40 ° C.) In the acid treatment, for example, anisol, phenol, thioanisole, methacresol, paracresol It is effective to add a cation scavenger such as dimethyl sulfide, 1,4-butanedithiol, 1,2-ethanedithiol, etc. The 2,4-dinitrophenyl group used as an imidazole protecting group for histidine is effective. The formyl group which is removed by thiophenol treatment and used as an indole protecting group for tributofan is not only protected by acid treatment in the presence of 1,2-ethanedithiol and 1,4-butanedithiol as described above, but also dilute hydroxylated. Also removed by alkali treatment with sodium solution, dilute ammonia, etc.
原料の反応に関与すべきでない官能基の保護ならびに保護基、 およびその保護 基の脱離、 反応に関与する官能基の活性ィ匕などは公知の基または公知の手段から 適宜選択しうる。  The protection of the functional group which should not be involved in the reaction of the raw materials, the protective group, the elimination of the protective group, the activation of the functional group involved in the reaction, and the like can be appropriately selected from known groups or known means.
タンパク質 ·ペプチドのアミド体を得る別の方法としては、 例えば、 まず、 力 ルポキシ末端アミノ酸の α—力ルポキシル基をアミド化して保護した後、 ァミノ 基側にぺプチド鎖を所望の鎖長まで延ばした後、 該ぺプチド鎖の Ν末端の α—ァ ミノ基の保護基のみを除いたタンパク質 ·ペプチドと C末端の力ルポキシル基の 保護基のみを除去したタンパク質 ·ペプチドとを製造し、 この両ペプチド ·両夕 ンパク質を上記したような混合溶媒中で縮合させる。 縮合反応の詳細については 上記と同様である。 縮合により得られた保護ペプチド ·保護タンパク質を精製し た後、 上記方法によりすベての保護基を除去し、 所望の粗ペプチド ·粗タンパク 質を得ることができる。 この粗ペプチド ·粗タンパク質は既知の各種精製手段を 駆使して精製し、 主要画分を凍結乾燥することで所望のタンパク質 ·ペプチドの アミド体を得ることができる。 As another method for obtaining an amide of a protein or peptide, for example, first, after amidating and protecting the α-hydroxyl group of the amino acid at the terminal end of the amino acid, a peptide chain is extended to a desired chain length on the amino side. After that, the protein / peptide obtained by removing only the protecting group for the α-amino group at the Ν-terminal of the peptide chain and the C-terminal lipoxyl group are removed. A protein and a peptide from which only the protecting group has been removed are produced, and the peptide and the protein are condensed in a mixed solvent as described above. Details of the condensation reaction are the same as described above. After purifying the protected peptide / protected protein obtained by the condensation, all the protecting groups are removed by the above-mentioned method, and a desired crude peptide / crude protein can be obtained. The crude peptide / crude protein is purified by various known purification means, and the main fraction is freeze-dried to obtain an amide of the desired protein / peptide.
タンパク質 ·ペプチドのエステル体を得るには、 例えば、 カルポキシ末端アミ ノ酸の α—力ルポキシル基を所望のアルコール類と縮合しアミノ酸エステルとし た後、 タンパク質 ·ペプチドのアミド体と同様にして、 所望のタンパク質 ·ぺプ チドのエステル体を得ることができる。  In order to obtain an ester of protein / peptide, for example, after condensing the α-hydroxyloxyl group of a carboxy-terminal amino acid with a desired alcohol to form an amino acid ester, the same procedure as in the amide of protein / peptide is carried out. It is possible to obtain an ester of a protein and a peptide.
本発明のペプチドおよび本発明のタンパク質は、 自体公知のペプチドの合成法 に従って製造することができる。 また、 本発明のタンパク質の部分ペプチドまた はその塩は、 自体公知のペプチドの合成法に従って、 あるいは本発明のタンパク 質を適当なぺプチダーゼで切断することによって製造することができる。  The peptide of the present invention and the protein of the present invention can be produced according to a peptide synthesis method known per se. Further, the partial peptide of the protein of the present invention or a salt thereof can be produced according to a peptide synthesis method known per se, or by cleaving the protein of the present invention with an appropriate peptidase.
ペプチドの合成法としては、 例えば、 固相合成法、 液相合成法のいずれによつ ても良い。 すなわち、 本発明のペプチドもしくは本発明のタンパク質を構成し得 る部分ペプチドまた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ペプチドを製造することができる。 公知の縮合方法や保護基の脱離としては、 例えば、 以下の (i ) 〜 (V) に記載さ れた方法が挙げられる。  As a method for synthesizing a peptide, for example, any of a solid phase synthesis method and a liquid phase synthesis method may be used. That is, the peptide of the present invention is condensed with the peptide of the present invention or the partial peptide or amino acid capable of constituting the protein of the present invention and the remaining portion, and when the product has a protecting group, the protecting group is eliminated to remove the target peptide. Can be manufactured. Examples of the known condensation method and elimination of the protecting group include the methods described in the following (i) to (V).
(i) M. Bodanszkyおよび M. A. Ondet t i , ペプチド シンセシス (Pept ide Synthes i s) , Intersc ience Publ i shers, New York (1966年) ' (i) M. Bodanszky and M.A. Ondet t i, Peptide Synthes is, Interscience Publ i shers, New York (1966) ''
(i i) Schroederおよび Luebke、 ザペプチド (The Pept ide) , Academic Press, New York (1965年) (ii) Schroeder and Luebke, The Peptide, Academic Press, New York (1965)
(i i i) 泉屋信夫他、 ペプチド合成の基礎と実験、 丸善 (株) (1975年)  (iii) Nobuo Izumiya et al., Fundamentals and experiments of peptide synthesis, Maruzen Co., Ltd. (1975)
(iv) 矢島治明 および榊原俊平、 生化学実験講座 1、 蛋白質の化学 IV、 205、 (1977年)  (iv) Haruaki Yajima and Shunpei Sakakibara, Laboratory of Biochemistry 1, Protein Chemistry IV, 205, (1977)
(V) 矢島治明監修、 続医薬品の開発第 14巻 ペプチド合成広川書店 また、 反応後は通常の精製法、 たとえば、 溶媒抽出,蒸留 ラフィ一 ·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ぺプチ ドまたは本発明の部分べプチドを精製単離することができる。 上記方法で得られ るペプチドまたは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し、 逆に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。 (V) Supervised by Haruaki Yajima, Development of Pharmaceuticals Volume 14 Peptide Synthesis Hirokawa Shoten After the reaction, the peptide of the present invention or the partial peptide of the present invention can be purified and isolated by a combination of ordinary purification methods, for example, solvent extraction, distillation, laffification, liquid chromatography, and recrystallization. . When the peptide or partial peptide obtained by the above method is in a free form, it can be converted to an appropriate salt by a known method. Conversely, when the peptide or partial peptide is obtained in a salt form, it can be converted to a free form by a known method Can be converted.
本発明の夕ンパク質をコードするポリヌクレオチドとしては、 前述した本発明 のタンパク質をコードする塩基配列を含有するものであればいかなるものであつ てもよい。 好ましくは DNAである。 DNAとしては、 ゲノム DNA、 ゲノム D NAライブラリー、 前記した細胞 ·組織由来の cDNA、 前記した細胞 ·組織由 来の c DNAライブラリー、 合成 DNAのいずれでもよい。  The polynucleotide encoding the protein of the present invention may be any polynucleotide containing the above-described nucleotide sequence encoding the protein of the present invention. Preferably it is DNA. The DNA may be any of genomic DNA, genomic DNA library, cDNA derived from the cells and tissues described above, cDNA library derived from the cells and tissues described above, and synthetic DNA.
ライブラリ一に使用するベクターは、 バクテリオファージ、 プラスミド、 コス ミド、 ファージミドなどいずれであってもよい。 また、 前記した細胞 ·組織より total RN Aまたは mRN A画分を調製したものを用いて直接 Reverse  The vector used for the library may be any of bacteriophage, plasmid, cosmid, phagemid and the like. In addition, a reverse RNA was directly prepared using the total RNA or mRNA fraction prepared from the cells and tissues described above.
Transcriptase Polymerase Chain Reaction (以下、 R T- P C R法と略称する) によって増幅することもできる。 It can also be amplified by Transcriptase Polymerase Chain Reaction (hereinafter abbreviated as RT-PCR method).
本発明のタンパク質をコードする DI^Aとしては、 例えば (i) 配列番号: 3 で表される塩基配列を含有する DNA、 または配列番号: 3で表される塩基配列 とハイストリンジェントな条件下でハイブリダィズする塩基配列を有し、 配列番 号: 4で表されるアミノ酸配列を含有するタンパク質と実質的に同質の性質を有 するタンパク質をコードする DNA、 (ii) 配列番号: 13で表される塩基配列 を含有する DNA、 または配列番号: 13で表される塩基配列とハイストリンジ ェントな条件下でハイブリダィズする塩基配列を有し、 配列番号: 14で表され るアミノ酸配列を含有するタンパク質と実質的に同質の性質を有するタンパク質 をコードする DNA、 (iii) 配列番号: 23で表される塩基配列を含有する D NA、 または配列番号: 23で表される塩基配列とハイストリンジェン卜な条件 下でハイブリダィズする塩基配列を有し、 配列番号: 24で表されるアミノ酸配 列を含有するタンパク質と実質的に同質の性質を有するタンパク質をコードする DN Aであれば何れのものでもよい。 配列番号: 3で表される塩基配列とハイストリンジェントな条件下でハイプリ ダイズできる D N Aとしては、 例えば、 配列番号: 3で表される塩基配列と 9 8 %以上、 好ましくは約 9 9 %以上の相同性を有する塩基配列を含有する D NA などが用いられる。 The DI ^ A encoding the protein of the present invention includes, for example, (i) a DNA containing the nucleotide sequence represented by SEQ ID NO: 3 or a nucleotide sequence represented by SEQ ID NO: 3 under highly stringent conditions. A DNA encoding a protein having substantially the same properties as a protein having an amino acid sequence represented by SEQ ID NO: 4, having a base sequence hybridizing with: (ii) SEQ ID NO: 13 And a protein having a nucleotide sequence that hybridizes under high stringent conditions with the nucleotide sequence represented by SEQ ID NO: 13 or a protein comprising the amino acid sequence represented by SEQ ID NO: 14 (Iii) a DNA containing the nucleotide sequence represented by SEQ ID NO: 23, or a DNA encoding a protein having substantially the same properties. Any DNA encoding a protein having a nucleotide sequence that hybridizes under stringent conditions and having substantially the same properties as the protein containing the amino acid sequence represented by SEQ ID NO: 24 It may be. Examples of the DNA that can hybridize with the nucleotide sequence represented by SEQ ID NO: 3 under high stringency conditions include, for example, 98% or more, preferably about 99% or more of the nucleotide sequence represented by SEQ ID NO: 3 DNA containing a nucleotide sequence having homology of
配列番号: 1 3で表される塩基配列とハイストリンジエンドな条件下でハイブ リダィズできる D NAとしては、 例えば、 配列番号: 1 3で表される塩基配列と 9 2 %以上、 好ましくは約 9 5 %以上、 さらに好ましくは約 9 7 %以上、 より好 ましくは約 9 9 %以上の相同性を有する塩基配列を含有する D NAなどが用いら れる。  Examples of the DNA that can hybridize with the nucleotide sequence represented by SEQ ID NO: 13 under high stringency conditions include, for example, a nucleotide sequence represented by SEQ ID NO: 13 and 92% or more, preferably about 9% or less. A DNA containing a nucleotide sequence having a homology of 5% or more, more preferably about 97% or more, more preferably about 99% or more is used.
配列番号: 2 3で表される塩基配列とハイストリンジェントな条件下でハイブ リダィズできる D NAとしては、 例えば、 配列番号: 2 3で表される塩基配列と 9 2 %以上、 好ましくは約 9 5 %以上、 さらに好ましくは約 9 7 %以上、 より好 ましくは約 9 9 %以上の相同性を有する塩基配列を含有する D N Aなどが用いら れる。  Examples of the DNA that can hybridize with the nucleotide sequence represented by SEQ ID NO: 23 under high stringent conditions include, for example, the nucleotide sequence represented by SEQ ID NO: 23 and 92% or more, preferably about 9% or less. DNA containing a base sequence having a homology of 5% or more, more preferably about 97% or more, more preferably about 99% or more is used.
ハイブリダィゼーシヨンは、 公知の方法あるいはそれに準じる方法、 例えば、 モレキュラー ·クローニング (Molecul ar Cl oning) 2 nd (J. Sambrook et al. , Cold Spring Harbor Lab. Press, 1989) に記載の方法などに従って行なうこと ができる。 まだ、 市販のライブラリーを使用する場合、 添付の使用説明書に記載 の方法に従って行なうことができる。 より好ましく'は、 ハイストリンジェントな 条件に従って行なうことができる。  Hybridization is performed by a known method or a method equivalent thereto, such as the method described in Molecular Cloning (Molecular Cloning) 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). It can be done according to. If a commercially available library is still used, it can be performed according to the method described in the attached instruction manual. More preferably, it can be performed under high stringency conditions.
ハイストリンジェントな条件とは、 例えば、 ナトリゥム濃度が約 1 9〜 4 0 m M、 好ましくは約 1 9〜 2 0 mMで、 温度が約 5 0〜 7 0 、 好ましくは約 6 0 〜 6 5 °Cの条件を示す。 特に、 ナトリゥム濃度が約 1 9 mMで温度が約 6 5 の 場合が最も好ましい。  High stringency conditions include, for example, a sodium concentration of about 19 to 40 mM, preferably about 19 to 20 mM, and a temperature of about 50 to 70, preferably about 60 to 65. The conditions at ° C are shown. In particular, the case where the sodium concentration is about 19 mM and the temperature is about 65 is most preferable.
より具体的には、 配列番号: 4で表されるアミノ酸配列を含有するタンパク質 をコ一ドする D NAとしては、 配列番号: 3で表される塩基配列を含有する D N A、 配列番号: 3 8で表される塩基配列を含有する D NAなどが、 配列番号: 1 4で表されるアミノ酸配列を含有するタンパク質をコードする D N Aとしては、 配列番号: 1 3で表される塩基配列を含有する D NA、 配列番号: 3 9で表され る塩基配列を含有する DNAなどが、 配列番号: 24で表されるアミノ酸配列を 含有するタンパク質をコードする DNAとしては、 配列番号: 23で表される塩 基配列を含有する DNA、 配列番号: 40で表される塩基配列を含有する DN A などが用いられる。 More specifically, DNAs encoding the protein containing the amino acid sequence represented by SEQ ID NO: 4 include DNA containing the base sequence represented by SEQ ID NO: 3, and SEQ ID NO: 38. The DNA encoding the protein having the amino acid sequence represented by SEQ ID NO: 14 has the nucleotide sequence represented by SEQ ID NO: 13 DNA, represented by SEQ ID NO: 39 Examples of the DNA encoding a protein having the amino acid sequence represented by SEQ ID NO: 24, such as a DNA having a nucleotide sequence represented by SEQ ID NO: 24, include a DNA having a base sequence represented by SEQ ID NO: 23, DNA containing the base sequence represented by 40 or the like is used.
本発明の部分ペプチドをコードするポリヌクレオチドとしては、 前述した本発 明の部分べプチドをコ一ドする塩基配列を含有するものであればいかなる'もので あってもよい。 DNAが好ましい。 DNAとしては、 ゲノム DNA、 ゲノム DN Aライブラリ一、 前記した細胞 ·組織由来の cDNA、 前記した細胞 ·組織由来 の cDNAライブラリー、 合成 DNAのいずれでもよい。 '  The polynucleotide encoding the partial peptide of the present invention may be any polynucleotide as long as it contains a base sequence encoding the above-described partial peptide of the present invention. DNA is preferred. The DNA may be any of genomic DNA, genomic DNA library, cDNA derived from the above-described cells and tissues, cDNA library derived from the above-described cells and tissues, and synthetic DNA. '
本発明の部分ペプチドをコードする DNAとしては、 例えば、 配列番号: 3、 配列番号: 13または配列番号: 23で表される塩基配列を有する DNAの一部 分を有する DNA、 または配列番号: 3、 配列番号: 1.3または配列番号: 23 で表される塩基配列とハイストリンジェントな条件下でハイブリダィズする塩基 配列を含有し、 本発明のタンパク質と実質的に同質の活性を有するタンパク質を コードする DN Aの一部分を含有する DN Aなどが用いられる。  Examples of the DNA encoding the partial peptide of the present invention include, for example, DNA having a part of the DNA having the base sequence represented by SEQ ID NO: 3, SEQ ID NO: 13 or SEQ ID NO: 23, or SEQ ID NO: 3 A DN having a nucleotide sequence that hybridizes under high stringent conditions to the nucleotide sequence represented by SEQ ID NO: 1.3 or SEQ ID NO: 23, and encoding a protein having substantially the same activity as the protein of the present invention. DNA containing a part of A is used.
配列番号: 3、 配列番号: 13または配列番号: 23で表される塩基配列と八 イブリダィズできる DNAは、 前記と同意義を示す。  The DNA that can be hybridized with the base sequence represented by SEQ ID NO: 3, SEQ ID NO: 13 or SEQ ID NO: 23 has the same significance as described above.
ハイブリダイゼーションの方法およびハイストリンジェン卜な条件は前記と同 様のものが用いられる。  The same hybridization method and high stringency conditions as described above are used.
本発明のタン ク質、 部分ペプチド (以下、 これらをコードする DNAのクロ 一二ングおよび発現の説明においては、 これらを単に本発明のタンパク質と略記 する場合がある) を完全にコードする DNAのクローニングの手段としては、 本 発明のタンパク質をコードする塩基配列の一部分を有する合成 DN Aプライマー を用いて PCR法によつて増幅するか、 または適当なベクタ一に組み込んだ D N Aを本発明のタンパク質の一部あるいは全領域をコードする DNA断片もしくは 合成 DN Aを用いて標識したものとのハイブリダィゼーシヨンによって選別する ことができる。 ハイブリダィゼーシヨンの方法は、 例えば、 モレキュラー ·クロ —ニング (Molecular Cloning) 2nd (J. Sa brook et al., Cold Spring Harbor Lab. Press, 1989) に記載の方法などに従って行なうことができる。 ま た、 市販のライブラリ一を使用する場合、 添付の使用説明書に記載の方法に従つ て行なうことができる。 The DNA or DNA encoding the protein or partial peptide of the present invention (hereinafter, these may be simply referred to as the protein of the present invention in the description of the cloning and expression of the DNA encoding the same). As a means of cloning, the DNA of the present invention is amplified by PCR using a synthetic DNA primer having a part of the nucleotide sequence encoding the protein of the present invention, or the DNA of the present invention is incorporated into an appropriate vector. Selection can be performed by hybridization with a DNA fragment encoding a part or all of the region or labeled with a synthetic DNA. The hybridization can be performed according to, for example, the method described in Molecular Cloning 2nd (J. Sabrook et al., Cold Spring Harbor Lab. Press, 1989). Ma When a commercially available library is used, it can be performed according to the method described in the attached instruction manual.
DNAの塩基配列の変換は、 PCRや公知のキット、 例えば、 Mutan™- super Express Km (宝酒造 (株) ) 、 Mutan™- K (宝酒造 (株) ) 等を用いて、 0DA-LA PCR法や Gapped duplex法や Kunkel法等の公知の方法あるいはそれらに準じる方法 に従つて行なうことができる。  The DNA base sequence can be converted by PCR or a known kit such as Mutan ™ -Super Express Km (Takara Shuzo) or Mutan ™ -K (Takara Shuzo) using the 0DA-LA PCR method or the like. It can be performed according to a known method such as the gapped duplex method or the Kunkel method, or a method analogous thereto.
クローン化されたタンパク質をコードする DNAは目的によりそのまま、 また は所望により制限酵素で消化したり、 リンカ一を付加したりして使用することが できる。 該 DNAはその 5,末端側に翻訳開始コドンとしての ATGを有し、 また 3,末端側には翻訳終止コドンとしての TAA、 TGAまたは 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ァダプ タ一を用いて付加することもできる。  The DNA encoding the cloned protein can be used as it is depending on the purpose, or can be used after digestion with a restriction enzyme or addition of a linker, if desired. The DNA may have ATG as a translation initiation codon on the 5, terminal side, and TAA, TGA or TAG as a translation termination codon on the 3, terminal side. These translation initiation codon and translation termination codon can also be added using a suitable synthetic DNA adapter.
本発明のタンパク質の発現べクタ一は、 例えば、 (ィ) 本発明のタンパク質を コードする DNAから目的とする DNA断片を切り出し、 (口) 該 DNA断片を 適当な発現べクタ一中のプロモータ一の下流に連結することにより製造すること ができる。  The expression vector of the protein of the present invention can be prepared, for example, by (a) cutting out a DNA fragment of interest from DNA encoding the protein of the present invention, and (mouth) converting the DNA fragment into a promoter in an appropriate expression vector. It can be manufactured by connecting downstream of
ベクタ一としては、 大腸菌由来のプラスミド (例、 pBR322, PBR32 5, pUC 12, pUC 13) 、 枯草菌由来のプラスミド (例、 pUB 1 10, pTP 5, p C 194) 、 酵母由来プラスミド (例、 p SH 19, p SH 15) λファージなどのパクテリオファージ、 レトロウイルス, ワクシニアウィルス, バキュロウィルスなどの動物ウィルスなどの他、 pAl— 1 1、 pXTl、 pR cZCMV、 pRc/RSV、 p c DNA I ZNe oなどが用いられる。  Examples of the vector include a plasmid derived from E. coli (eg, pBR322, PBR32 5, pUC 12, pUC 13), a plasmid derived from Bacillus subtilis (eg, pUB 110, pTP 5, pC 194), a yeast-derived plasmid (eg, pSH19, pSH15) In addition to pacteriophage such as λ phage, animal viruses such as retrovirus, vaccinia virus, and baculovirus, pAl-11, pXTl, pR cZCMV, pRc / RSV, pc DNA I ZNe o and the like are used.
本発明で用いられるプロモー夕一としては、 遺伝子の発現に用いる宿主に対応 して適切なプロモー夕一であればいかなるものでもよい。 例えば、 動物細胞を宿 主として用いる場合は、 SRaプロモーター、 SV40プロモータ一、 LTRプ ロモ—夕—、 CMVプロモータ一、 HS V-TKプロモーターなどが挙げられる < これらのうち、 CMV (サイトメガロウィルス) プロモーター、 SRo!プロモ 一夕一などを用いるのが好ましい。 宿主がェシエリヒア属菌である場合は、 t r pプロモーター、 l a cプロモーター、 r e cAプロモーター、 λ PLプロモ一 ター、 l pp:/°口モーター、 T 7プロモータ一などが、 宿主がバチルス属菌であ る場合は、 SP01プロモー夕一、 SP02プロモータ一、 p enPプロモータ 一など、 宿主が酵母である場合は、 PHO 5プロモーター、 PGKプロモーター GAPプロモータ一、 ADHプロモーターなどが好ましい。 宿主が昆虫細胞であ る場合は、 ポリヘドリンプロモータ 、 P 10プロモーターなどが好ましい。 発現ベクターには、 以上の他に、 所望によりェンハンサ一、 スプライシングシ ダナル、 ポリ A付加シグナル、 選択マーカ一、 SV40複製オリジン (以下、 S V400 r iと略称する場合がある) などを含有しているものを用いることがで きる。 選択マーカ一としては、 例えば、 ジヒドロ葉酸還元酵素 (以下、 dh f r と略称する場合がある) 遺伝子 〔メソトレキセ一ト (MTX) 耐性〕 、 アンピシ リン耐性遺伝子 (以下、 Amp rと略称する場合がある) 、 ネオマイ,シン耐性遺 伝子 (以下、 Ne orと略称する場合がある、 G418耐性) 等が挙げられる。 特に、 dh ί r遺伝子欠損チャイニーズハムスター細胞を用いて dh f r遺伝子 を選択マーカ一として使用する場合、 目的遺伝子をチミジンを含まない培地によ つても選択できる。 The promoter used in the present invention may be any suitable promoter corresponding to the host used for gene expression. For example, when animal cells are used as hosts, SRa promoter, SV40 promoter, LTR promoter, CMV promoter, HSV-TK promoter, etc. may be mentioned. Of these, CMV (cytomegalovirus) promoter It is preferable to use SRo! When the host is Escherichia, the trp promoter, lac promoter, recA promoter, λPL promoter If the host is a Bacillus genus, SP01 promoter, SP02 promoter, penP promoter, etc.If the host is yeast, the host is yeast. , PHO5 promoter, PGK promoter, GAP promoter, ADH promoter and the like are preferable. When the host is an insect cell, a polyhedrin promoter, a P10 promoter and the like are preferable. The expression vector may contain, in addition to the above, an enhancer, a splicing signal, a poly-A addition signal, a selection marker, and an SV40 replication origin (hereinafter sometimes abbreviated as SV400 ri), if desired. Anything can be used. The selection marker one, for example, dihydrofolate reductase (hereinafter sometimes abbreviated as dh fr) gene [Mesotorekise Ichito (MTX) resistance], ampicillin phosphorus resistant gene (hereinafter sometimes abbreviated as Amp r ), Neomai, Shin-resistant gene (hereinafter sometimes abbreviated as Ne o r, include G418 resistance) and the like. In particular, when the dhfr gene is used as a selection marker using Chinese hamster cells deficient in the dh 遺 伝 子 r gene, the target gene can be selected using a thymidine-free medium.
また、 必要に応じて、 宿主に合ったシグナル配列を、 本発明のタンパク質の N 端末側に付加する。 宿主がェシェリヒア属菌である場合は、 P h o A ·シグナル 配列、 Omp A ·シグナル配列などが、 宿主がバチルス属菌である場合は、 一 アミラーゼ ·シグナル配列、 サブチリシン ·シグナル配列などが、 宿主が酵母で ある場合は、 MFa ·シグナル配列、 SUC2 ·シグナル配列など、 宿主が動物 細胞である場合には、 インシュリン ·シグナル配列、 ひ—インターフェロン ·シ グナル配列、 抗体分子 ·シグナル配列などがそれぞれ利用できる。  If necessary, a signal sequence suitable for the host is added to the N-terminal side of the protein of the present invention. If the host is Escherichia, the Pho A signal sequence, Omp A signal sequence, etc., if the host is Bacillus, the amylase signal sequence, subtilisin signal sequence, etc. In the case of yeast, MFa signal sequence and SUC2 signal sequence can be used. When the host is an animal cell, insulin signal sequence, high interferon signal sequence, antibody molecule and signal sequence can be used, respectively. .
このようにして構築された本発明のタンパク質をコードする DN Aを含有する ベクターを用いて、 形質転換体を製造することができる。  Using the vector containing the DNA encoding the protein of the present invention thus constructed, a transformant can be produced.
宿主としては、 例えば、 ェシエリヒア属菌、 バチルス属菌、'酵母、 昆虫細胞、 昆虫、 動物細胞などが用いられる。  Examples of the host include Escherichia, Bacillus, yeast, insect cells, insects, animal cells, and the like.
ェシエリヒア属菌の具体例としては、 例えば、 ェシエリヒア 'コリ  Specific examples of the genus Escherichia include, for example, Escherichia coli.
(Escherichia coli) K 12 - DH 1 (Proc. Natl. Acad. Sci. USA, 60巻, 160(1968)) , JM103 (Nucleic Acids Research, 9巻, 309 (1981)) , J A 2 21 (Journal of Molecular Biology, 120巻, 517 (1978)) , HB 101 〔ジャ ーナル ·ォブ 'モレキュラー 'バイオロジー, 41巻, 459 (1969)〕 , C 600 (Genetics, 39卷, 440(1954))などが用いられる。 (Escherichia coli) K12-DH1 (Proc. Natl. Acad. Sci. USA, 60, 160 (1968)), JM103 (Nucleic Acids Research, 9, 309 (1981)), JA2 21 (Journal of Molecular Biology, 120, 517 (1978)), HB101 [Journal of 'Molecular' Biology, 41, 459 (1969)], C600 (Genetics, 39, 440 (1954)) )) Is used.
バチルス属菌としては、 例えば、 バチルス ·サブチルス (Bacillus  Examples of Bacillus bacteria include, for example, Bacillus subtilis (Bacillus
subtilis) MI 114 (Gene, 24巻, 255 (1983) ) , 207 - 21 (Journal of Biochemistry, 95巻, 87(1984)) などが用いられる。 ' 酵母としては、 例えば、 サッカロマイセス セレピシェ (Saccharomyces cerevisiae) AH22, AH22R— , ΝΑ87 - 11 A, DKD- 5D, 20 B— 12、 シゾサッカロマイセス ボンべ, (Schizosaccharomyces porabe) NC YC 1913, NCYC 2036、 ピキア パストリス (Pichia pastoris) K Μ71などが用いられる。 subtilis) MI114 (Gene, 24, 255 (1983)), 207-21 (Journal of Biochemistry, 95, 87 (1984)) and the like are used. '' Examples of yeast include, for example, Saccharomyces cerevisiae AH22, AH22R—, ΝΑ87-11A, DKD-5D, 20B-12, Schizosaccharomyces porabe, NC YC 1913, NCYC 2036 Pastoris (Pichia pastoris) K # 71 or the like is used.
昆虫細胞としては、 例えば、 ウィルスが Ac NPVの場合は、 夜盗蛾の幼虫由 来株化細胞 (Spodoptera frugiperda cell; S f細胞) 、 Trichoplusia niの中 腸由来の MG1細胞、 Trichoplusia niの卵由来の High FiveTM細胞、 Mamestra brassicae由来の細胞または Estigmena acrea由来の細胞などが用いられる。 ウイ ルスが BmNP Vの場合は、 蚕由来株化細胞 (Bombyx mori N細胞; BmN細 胞) などが用いられる。 該 S f細胞としては、 例えば、 S f 9細胞 (ATCC  As insect cells, for example, when the virus is Ac NPV, a cell line derived from a larva of night roth moth (Spodoptera frugiperda cell; Sf cell), MG1 cell derived from the midgut of Trichoplusia ni, and egg derived from Trichoplusia ni egg High Five ™ cells, cells derived from Mamestra brassicae, cells derived from Estigmena acrea, and the like are used. When the virus is BmNPV, a cell line derived from a silkworm (Bombyx mori N cell; BmN cell) is used. Examples of the Sf cells include Sf9 cells (ATCC
CRL1711) 、 S f 21細胞 (以上、 Vaughn, J.L.ら、 イン .ヴィポ (In CRL1711), Sf21 cells (Vaughn, J.L. et al., In.
Vivo) ,13, 213-217, (1977)) などが用いられる。 Vivo), 13, 213-217, (1977)).
昆虫としては、 例えば、 カイコの幼虫などが用いられる 〔前田ら、 ネイチヤー (Nature) , 315卷, 592 (1985)〕 。  As insects, for example, silkworm larvae are used [Maeda et al., Nature, Vol. 315, 592 (1985)].
動物細胞としては、 例えば、 サル細胞 COS— 7, Ve r o, チャイニーズハ ムスター細胞 CHO (以下、 CHO細胞と略記) , dh f r遺伝子欠損チヤィニ —ズハムスター細胞 CH〇 (以下、 CHO (dh f r") 細胞と略記) , マウス L 細胞, マウス At T— 20, マウスミエ口一マ細胞, ラット GH3, ヒト FL細 胞などが用いられる。  Examples of animal cells include monkey cell COS-7, Vero, Chinese hamster cell CHO (hereinafter abbreviated as CHO cell), dh fr gene-deficient chinini—zhamster cell CH〇 (hereinafter CHO (dh fr)). Cells), mouse L cells, mouse AtT-20, mouse myeoma cells, rat GH3, human FL cells, and the like.
ェシエリヒア属菌を形質転換するには、 例えば、 Proc. Natl; Acad. Sci. USA, 69巻, 2110 (1972)や Gene, 17巻, 107 (1982)などに記載の方法に従つて行なうこと ができる。 バチルス属菌を形質転換するには、 例えば、 モレキュラー ·アンド ·ジエネラ ル'ジェネティックス (Molecular & General Genetics) , 168巻, 1 11 (1979)などに記載の方法に従って行なうことができる。 Transformation of the genus Escherichia can be performed, for example, according to the method described in Proc. Natl; Acad. Sci. USA, 69, 2110 (1972) or Gene, 17, 107 (1982). it can. Transformation of Bacillus spp. Can be performed, for example, according to the method described in Molecular & General Genetics, Volume 168, 111 (1979).
' 酵母を形質転換するには、 例えば、 Methods in Enzymology, 194卷, 182 - 187(1991), Proc. Natl. Acad. Sci. USA, 75巻, 1929 (1978)などに記載の方法に 従って行なうことができる。 '' The yeast is transformed according to the method described in, for example, Methods in Enzymology, Vol. 194, 182-187 (1991), Proc. Natl. Acad. Sci. USA, Vol. 75, 1929 (1978). be able to.
昆虫細胞または昆虫を形質転換するには、 例えば、 Bio/Technology, 6, 47- 55 (1988)などに記載の方法に従って行なうことができる。  Insect cells or insects can be transformed, for example, according to the method described in Bio / Technology, 6, 47-55 (1988).
動物細胞を形質転換するには、 例えば、 細胞工学別冊 8 新細胞工学実験プロ トコール. 263— 267 (1995) (秀潤社発行) 、 Virology, 52  To transform animal cells, for example, see Cell Engineering Annex 8 New Cell Engineering Experimental Protocol. 263—267 (1995) (published by Shujunsha), Virology, 52
巻, 456 (1973)に記載の方法に従って行なうことができる。 Volume, 456 (1973).
このようにして、 夕ンパク質をコ ドする D N Aを含有する発現ベクターで形 質転換された形質転換体を得ることができる。  Thus, a transformant transformed with the expression vector containing DNA encoding the protein can be obtained.
宿主がェシエリヒア属菌、 バチルス属菌である形質転換体を培養する際、 培養 に使用される培地としては液体培地が適当であり、 その中には該形質転換体の生 育に必要な炭素源、 窒素源、 無機物その他が含有せしめられる。 炭素源としては、 例えば、 グルコース、 デキストリン、 可溶性澱粉、 ショ糖など、 窒素源としては、 例えば、 アンモニゥム塩類、 硝酸塩類、 コーンスチープ ·リカ一、 ペプトン、 力 ゼイン、 肉エキス、 大豆粕、 バレイショ抽出液などの無機または有機物質、 無機 物としては、 例えば、 塩化カルシウム、 リン酸二水素ナトリウム、 塩化マグネシ ゥムなどが挙げられる。 また、 酵母エキス、 ビタミン類、 生長促進因子などを添 加してもよい。 培地の pHは約 5〜8が望ましい。  When culturing a transformant whose host is a bacterium belonging to the genus Escherichia or Bacillus, a liquid medium is suitable as a medium used for the cultivation, and a carbon source necessary for the growth of the transformant is contained therein. , Nitrogen sources, inorganic substances and others. Examples of carbon sources include glucose, dextrin, soluble starch, and sucrose. Examples of nitrogen sources include ammonium salts, nitrates, corn chip lica, peptone, zein, meat extract, soybean meal, and potato extract. Examples of the inorganic or organic substance such as a liquid and the inorganic substance include calcium chloride, sodium dihydrogen phosphate, and magnesium chloride. In addition, yeast extract, vitamins, growth promoting factors and the like may be added. The pH of the medium is preferably about 5-8.
ェシエリヒア属菌を培養する際の培地としては、 例えば、 グルコース、 カザミ ノ酸を含む M 9培地 〔ミラー (Miller) , ジャーナル'ォブ ·ェクスペリメン ッ 'イン 'モレキュラー 'ジェ不ティックス (Journal of Experiments in  Examples of a medium for culturing Escherichia bacteria include, for example, an M9 medium containing glucose and casamino acids [Miller, Journal “Obexperiment”, “Molecular”, Journal of Experiments in
Molecular Genetics) , 431—433, Cold Spring Harbor Laboratory, New York 1972〕 が好ましい。 ここに必要によりプロモーターを効率よく働かせ るために、 例えば、 3 ;6—インドリルアクリル酸のような薬剤を加えることがで さる。 宿主がェシェリヒァ属菌の場合、 培養は通常約 1 5〜 43 °Cで約 3〜 24時間 行ない、 必要により、 通気や撹拌を加えることもできる。 Molecular Genetics), 431-433, Cold Spring Harbor Laboratory, New York 1972]. Here, if necessary, a drug such as 3; 6-indolylacrylic acid can be added to make the promoter work efficiently. When the host is a bacterium belonging to the genus Escherichia, the cultivation is usually carried out at about 15 to 43 ° C for about 3 to 24 hours, and if necessary, aeration and stirring can be applied.
宿主がバチルス属菌の場合、 培養は通常約 30〜 40 °Cで約 6〜 24時間行な レ、 -必要により通気や撹拌を加えることもできる。  When the host is a bacterium belonging to the genus Bacillus, cultivation is usually performed at about 30 to 40 ° C for about 6 to 24 hours.
宿主が酵母である形質転換体を培養する際、 培地としては、 例えば、 バークホ 一ルダー (Burkholder) 最小培地 [Bostian, K. L. ら、 Proc. Natl. Acad. Sci. USA, 77巻, 4505 (1980)〕 や 0.5 %カザミノ酸を含有する S D培地 〔Bitter, G. A. ら、 Proc. Natl. Acad. Sci. USA, 81巻, 5330 (1984)〕 が挙げられる。 培地の pHは約 5〜 8に調整するのが好ましい。 培養は通常約 20°C〜35°Cで約 24 〜72時間行ない、 必要に応じて通気や撹拌を加える。  When culturing a transformant in which the host is yeast, for example, Burkholder's minimal medium [Bostian, KL et al., Proc. Natl. Acad. Sci. USA, 77, 4505 (1980) And a SD medium containing 0.5% casamino acid [Bitter, GA et al., Proc. Natl. Acad. Sci. USA, 81, 5330 (1984)]. The pH of the medium is preferably adjusted to about 5-8. The cultivation is usually performed at about 20 ° C to 35 ° C for about 24 to 72 hours, and aeration and stirring are added as necessary.
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、 Grace's Insect Medium (Grace, T. C.,ネィチヤ一 (Nature) , 195, 788(1962)) に非動化した 10%ゥシ血清等の添加物を適宜加えたものなどが用いられる。 培 地の ρΗ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日間行ない、 必要に応じて通気や撹拌を加える。  When culturing an insect cell or a transformant whose host is an insect, the medium used is Grace's Insect Medium (Grace, TC, Nature, 195, 788 (1962)). Those to which additives such as serum are appropriately added are used. Preferably, the ρΗ of the culture medium is adjusted to about 6.2 to 6.4. Culture is usually performed at about 27 ° C for about 3 to 5 days, and aeration and agitation are added as necessary.
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 〜20 %の胎児牛血清を含む MEM培地 〔サイエンス (Science) , 122巻, 501 (1952)) , DMEM培地 〔ヴイロロジ一 (Virology) , 8巻, 396 (1959)〕 , RPMI 1640培地 〔The Journal of the American Medical Association 199巻, 519 (1967)〕 , 199培地 [Proceeding of the Society for the Biological Medicine, 73巻, 1 (1950)〕 などが用いられる。 pHは約 6〜8 であるのが好ましい。 培養は通常約 30° (:〜 40°Cで約 15〜60時間行ない、 必要に応じて通気や撹拌を加える。  When culturing a transformant in which the host is an animal cell, examples of the medium include a MEM medium containing about 5 to 20% fetal bovine serum (Science, 122, 501 (1952)), a DMEM medium [Virology, 8, 396 (1959)], RPMI 1640 medium [The Journal of the American Medical Association 199, 519 (1967)], 199 medium [Proceeding of the Society for the Biological Medicine, 73] , 1 (1950)]. Preferably, the pH is about 6-8. Cultivation is usually performed at about 30 ° C (: up to 40 ° C for about 15 to 60 hours, and aeration and agitation are added as necessary.
以上のようにして、 形質転換体の細胞内、 細胞膜または細胞外に本発明のタン パク質を生成せしめることができる。  As described above, the protein of the present invention can be produced in the cells, in the cell membrane, or outside the cells of the transformant.
上記培養物から本発明のタンパク質を分離精製するには、 例えば、 下記の方法 により行なうことができる。  The protein of the present invention can be separated and purified from the culture by, for example, the following method.
本発明のタンパク質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、 リゾチームおよび Zまたは凍結融解などによって菌体あるいは細胞を破壌したの ち、 遠心分離やろ過によりタンパク質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などのタンパク質変性剤や、 トリトン X - 1 0 0 TMなどの界面活性剤が含まれていてもよい。 培養液中にタンパク質が分 泌される場合には、 培養終了後、 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。 When extracting the protein of the present invention from cultured cells or cells, after the culture, the cells or cells are collected by a known method, suspended in an appropriate buffer, After lysing cells or cells by lysozyme and Z or freeze-thawing, a method of obtaining a crude protein extract by centrifugation or filtration is used as appropriate. The buffer may contain a protein denaturant such as urea or guanidine hydrochloride, or a surfactant such as Triton X-1000 . When the protein is secreted into the culture solution, after completion of the culture, the supernatant is separated from the cells or cells by a known method, and the supernatant is collected.
このようにして得られた培養上清、 あるいは抽出液中に含まれるタンパク質の 精製は、 公知の分離 ·精製法を適切に組み合わせて行なうことができる。 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、 および S D S—ポリアクリルアミドゲル電気 泳動法など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 一などの荷電の差を利用する方法、 ァフィ二ティークロマトグラフィーなどの特 異的親和性を利用する方法、 逆相高速液体ク口マトグラフィ一などの疎水性の差 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。  Purification of the protein contained in the culture supernatant or extract obtained in this manner can be performed by appropriately combining known separation and purification methods. These known separation and purification methods mainly include methods using solubility such as salting out and solvent precipitation, dialysis, ultrafiltration, gel filtration, and SDS-polyacrylamide gel electrophoresis, mainly molecular weight. Methods that utilize differences in charge, methods that use differences in charge, such as ion-exchange chromatography, methods that use specific affinities, such as affinity chromatography, and hydrophobicity, such as reverse-phase high-performance liquid chromatography. A method using the difference between the isoelectric points, such as an isoelectric focusing method, and the like, may be used.
かくして得られるタンパク質が遊離体で得られた場合には、 公知の方法あるい はそれに準じる方法によって塩に変換することができ、 逆に塩で得られた場合に は公知の方法あるいはそれに準じる方法により、 遊離体または他の塩に変換する ことができる。  When the protein thus obtained is obtained in a free form, it can be converted to a salt by a known method or a method analogous thereto, and conversely, when the protein is obtained in the form of a salt, a known method or a method analogous thereto Can be converted into a free form or another salt.
なお、 組換え体が産生するタンパク質を、 精製前または精製後に適当な蛋白修 飾酵素を作用させることにより、 任意に修飾を加えたり、 ポリペプチドを部分的 に除去することもできる。 蛋白修飾酵素としては、 例えば、 トリプシン、 キモト リプシン、 アルギニルエンドべプチダ一ゼ、 プロテインキナーゼ、 グリコシダ一 ゼなどが用いられる。  The protein produced by the recombinant can be arbitrarily modified or the polypeptide can be partially removed by the action of an appropriate protein modifying enzyme before or after purification. As the protein modifying enzyme, for example, trypsin, chymotrypsin, arginyl endopeptidase, protein kinase, glycosidase and the like are used.
かくして生成する本発明のタンパク質の存在は、 特異抗体を用いたェンザィム ィムノアツセィゃウエスタンプロッティングなどにより測定することができる。 本発明のタンパク質もしくは部分ペプチドまたはその塩に対する抗体は、 本発 明の夕ンパク質もしくは部分べプチドまたはその塩を認識し得る抗体であれば、 ポリクローナル抗体、 モノクローナル抗体の何れであってもよい。 本発明のタンパク質もしくは部分ペプチドまたはその塩 (以下、 抗体の説明に おいては、 これらを単に本発明のタンパク質と略記する場合がある) に対する抗 体は、 本発明のタンパク質を抗原として用い、 公知の抗体または抗血清の製造法 に従つて製造することができる。 The presence of the protein of the present invention thus produced can be measured by, for example, enzymatic immunoassay western blotting using a specific antibody. The antibody against the protein or partial peptide of the present invention or a salt thereof may be any of a polyclonal antibody and a monoclonal antibody as long as it can recognize the protein or partial peptide of the present invention or a salt thereof. An antibody against the protein or partial peptide of the present invention or a salt thereof (hereinafter, these may be simply abbreviated to the protein of the present invention in the description of the antibody) is obtained by using the protein of the present invention as an antigen. The antibody or antiserum can be produced according to the following method.
〔モノクローナル抗体の作製〕  [Preparation of monoclonal antibody]
(a) モノクローナル抗体産生細胞の作製  (a) Preparation of monoclonal antibody-producing cells
本発明のタンパク質は、 温血動物に対して投与により抗体産生が可能な部位に それ自体あるいは担体、 希釈剤とともに投与される。 投与に際して抗体産生能を 高めるため、 完全フロイントアジュバントゃ不完全フロイントアジュバントを投 与してもよい。 投与は通常 2〜 6週毎に 1回ずつ、 計 2〜10回程度行われる。 用いられる温血動物としては、 例えば、 サル、 ゥサギ、 ィヌ、 モルモット、 マウ ス、 ラット、 ヒッジ、 ャギ、 ニヮトリが挙げられるが、 マウスおよびラットが好 ましく用いられる。  The protein of the present invention is administered to a warm-blooded animal itself or together with a carrier or diluent at a site capable of producing an antibody upon administration. Complete Freund's adjuvant / incomplete Freund's adjuvant may be administered in order to enhance antibody production upon administration. Administration is usually performed once every 2 to 6 weeks, for a total of 2 to 10 times. Examples of the warm-blooded animal to be used include monkeys, egrets, dogs, guinea pigs, mice, rats, sheep, goats, and chickens, and mice and rats are preferably used.
モノクローナル抗体産生細胞の作製に際しては、 抗原で免疫された温血動物、 例えばマウスから抗体価の認められた個体を選択し最終免疫の 2〜 5日後に脾臓 またはリンパ節を採取し、 それらに含まれる抗体産生細胞を同種または異種動物 の骨髄腫細胞と融合させることにより、 モノクローナル抗体産生ハイプリドーマ を調製することができる。 抗血清中の抗体価の測定は、 例えば、 後記の標識化夕 ンパク質と抗血清とを反応させたのち、 抗体に結合した標識剤の活性を測定する ことにより行なうことができる。 融合操作は既知の方法、 例えば、 ケーラーとミ ルスタインの方法 Nature, 256、 495 (1975)〕 に従い実施することができる。 '融合促進剤としては、 例えば、 ポリエチレングリコール (PEG) やセンダイゥ ィルスなどが挙げられるが、 好ましくは P E Gが用いられる。  When preparing monoclonal antibody-producing cells, a warm-blooded animal immunized with an antigen, for example, an individual with an antibody titer is selected from a mouse, and the spleen or lymph node is collected 2 to 5 days after the final immunization and contained in them. By fusing the antibody-producing cells obtained with myeloma cells of the same or different species, a monoclonal antibody-producing hybridoma can be prepared. The antibody titer in the antiserum can be measured, for example, by reacting the labeled protein described below with the antiserum, and then measuring the activity of the labeling agent bound to the antibody. The fusion operation can be performed according to a known method, for example, the method of Koehler and Milstein Nature, 256, 495 (1975)]. 'Fusion promoters include, for example, polyethylene glycol (PEG), Sendai virus and the like, and preferably PEG is used.
骨髄腫細胞としては、 例えば、 NS— 1、 P 3U1、 S P 2/0、 AP— 1な どの温血動物の骨髄腫細胞が挙げられるが、 P 3 U 1が好ましく用いられる。 用 いられる抗体産生細胞 (脾臓細胞) 数と骨髄腫細胞数との好ましい比率は 1 : 1 〜20 : 1程度であり、 PEG (好ましくは PEG 1000〜PEG6000) が 10〜 80 %程度の濃度で添加され、 20〜 40 °C、 好ましくは 30〜 37 °C で 1〜10分間インキュベートすることにより効率よく細胞融合を実施できる。 モノクローナル抗体産生ハイプリドーマのスクリ一ニングには種々の方法が使 用できるが、 例えば、 タンパク質抗原を直接あるいは担体とともに吸着させた固 相 (例、 マイクロプレート) にハイブリド一マ培養上清を添加し、 次に放射性物 質や酵素などで標識した抗免疫グロプリン抗体 (細胞融合に用いられる細胞がマ ウスの場合、 抗マウス免疫グロブリン抗体が用いられる) またはプロテイン Aを 加え、 固相に結合したモノクローナル抗体を検出する方法、 抗免疫グロブリン抗 体またはプロテイン Aを吸着させた固相にハイプリドーマ培養上清を添加し、 放 射性物質や酵素などで標識したタンパク質を加え、 固相に結合したモノクロ一ナ ル抗体を検出する方法などが挙げられる。 Examples of myeloma cells include myeloma cells of warm-blooded animals such as NS-1, P3U1, SP 2/0, and AP-1, but P3U1 is preferably used. The preferred ratio between the number of antibody-producing cells (spleen cells) and the number of myeloma cells used is about 1: 1 to 20: 1, and PEG (preferably PEG1000 to PEG6000) is used at a concentration of about 10 to 80%. Cell fusion can be carried out efficiently by adding the mixture and incubating at 20 to 40 ° C, preferably 30 to 37 ° C, for 1 to 10 minutes. Various methods can be used to screen monoclonal antibody-producing hybridomas.For example, a hybridoma culture supernatant is added to a solid phase (eg, a microplate) on which a protein antigen is adsorbed directly or together with a carrier. Then, an anti-immunoglobulin antibody (anti-mouse immunoglobulin antibody is used if the cell used for cell fusion is a mouse) or protein A labeled with a radioactive substance or an enzyme, and a monoclonal antibody bound to the solid phase Antibody detection method, Add the hybridoma culture supernatant to a solid phase to which anti-immunoglobulin antibody or protein A is adsorbed, add proteins labeled with radioactive substances, enzymes, etc. Examples include a method for detecting a single antibody.
モノクローナル抗体の選別は、 公知あるいはそれに準じる方法に従って行なう ことができる。 通常 HAT (ヒポキサンチン、 アミノプテリン、 チミジン) を添 加した動物細胞用培地で行なうことができる。 選別および育種用培地としては、 ハイプリドーマが生育できるものならばどのような培地を用いても良い。 例えば、 1〜2 0 %、 好ましくは 1 0〜2 0 %の牛胎児血清を含む R P M I 1 6 4 0培 地、 1〜 1 0 %の牛胎児血清を含む G I T培地 (和光純薬工業 (株) ) あるいは ハイプリドーマ培養用無血清培地 (S F M— 1 0 1、 日水製薬 (株) ) などを用 いることができる。 培養温度は、 通常 2 0〜4 0 、 好ましくは約 3 7 °Cである。 培養時間は、 通常 5日〜 3週間、 好ましくは 1週間〜 2週間である。 培養は、 通 常 5 %炭酸ガス下で行なうことができる。 ハイプリドーマ培養上清の抗体価は、 上記の抗血清中の抗体価の測定と同様にして測定できる。  The selection of the monoclonal antibody can be performed according to a known method or a method analogous thereto. Usually, it can be performed in an animal cell culture medium supplemented with HAT (hypoxanthine, aminopterin, thymidine). As a selection and breeding medium, any medium can be used as long as it can grow a hybridoma. For example, RPMI 1640 medium containing 1 to 20%, preferably 10 to 20% fetal bovine serum, and GIT medium containing 1 to 10% fetal bovine serum (Wako Pure Chemical Industries, Ltd. )) Alternatively, serum-free medium for hybridoma culture (SFM-101, Nissui Pharmaceutical Co., Ltd.) can be used. The culture temperature is usually 20 to 40, preferably about 37 ° C. The culture time is generally 5 days to 3 weeks, preferably 1 week to 2 weeks. The culture can be usually performed under 5% carbon dioxide gas. The antibody titer of the hybridoma culture supernatant can be measured in the same manner as the measurement of the antibody titer in the antiserum described above.
( b ) モノクローナル抗体の精製  (b) Purification of monoclonal antibody
モノクローナル抗体の分離精製は、 公知の方法、 例えば、 免疫グロブリンの分 離精製法 〔例、 塩析法、 アルコール沈殿法、 等電点沈殿法、 電気泳動法、 イオン 交換体 (例、 D E A E ) による吸脱着法、 超遠心法、 ゲルろ過法、 抗原結合固相 あるいはプロテイン Aあるいはプロテイン Gなどの活性吸着剤により抗体のみを 採取し、 結合を解離させて抗体を得る特異的精製法〕 に従って行なうことができ る。  Monoclonal antibodies can be separated and purified by known methods, for example, immunoglobulin separation and purification methods (eg, salting out method, alcohol precipitation method, isoelectric point precipitation method, electrophoresis method, ion exchanger (eg, DEAE)). Adsorption-desorption method, ultracentrifugation method, gel filtration method, antigen-binding solid phase or specific purification method in which the antibody is collected using an active adsorbent such as protein A or protein G and the bond is dissociated to obtain the antibody). Can be done.
〔ポリクロ一ナル抗体の作製〕  (Preparation of polyclonal antibody)
本発明のポリクローナル抗体は、 公知あるいはそれに準じる方法に従って製造 することができる。 例えば、 免疫抗原 (タンパク質抗原) 自体、 あるいはそれと キャリアー蛋白質との複合体をつくり、 上記のモノクローナル抗体の製造法と同 様に温血動物に免疫を行ない、 該免疫動物から本発明のタンパク質に対する抗体 含有物を採取して、 抗体の分離精製を行なうことにより製造することができる。 温血動物を免疫するために用いられる免疫抗原とキャリア一蛋白質との複合体 に関し、 キャリアー蛋白質の種類およびキャリア一とハプテンとの混合比は、 キ ャリァ一に架橋させて免疫したハプテンに対して抗体が効率良くできれば、 どの 様なものをどの様な比率で架橋させてもよいが、 例えば、 ゥシ血清アルブミンや ゥシサイログロブリン、 へモシァニン等を重量比でハプテン 1に対し、 約 0 . 1 〜2 0、 好ましくは約 1〜5の割合でカプルさせる方法が用いられる。 The polyclonal antibody of the present invention is produced according to a known method or a method analogous thereto. can do. For example, a immunizing antigen (protein antigen) itself or a complex thereof with a carrier protein is formed, and immunization is performed on a warm-blooded animal in the same manner as in the above-described method for producing a monoclonal antibody. It can be produced by collecting the contents and separating and purifying the antibody. Regarding a complex of an immunizing antigen and a carrier-protein used for immunizing a warm-blooded animal, the type of the carrier protein and the mixing ratio between the carrier and the hapten are different from those of the hapten immunized by cross-linking the carrier. Any antibody may be cross-linked at any ratio as long as it can be efficiently produced.For example, serum albumin, thyroglobulin, hemocyanin, etc., in a weight ratio of about 0.1 to hapten per hapten, may be used. -20, preferably about 1-5.
また、 ハプテンとキャリア一の力プリングには、 種々の縮合剤を用いることが できるが、 グルタルアルデヒドやカルポジイミド、 マレイミド活性エステル、 チ オール基、 ジチオビリジル基を含有する活性エステル試薬等が用いら lる。  Various condensing agents can be used for force coupling between the hapten and the carrier, but glutaraldehyde, carbodiimide, a maleimide active ester, an active ester reagent containing a thiol group or a dithioviridyl group, or the like is used. .
縮合生成物は、 温血動物に対して、 抗体産生が可能な部位にそれ自体あるいは 担体、 希釈剤とともに投与される。 投与に際して抗体産生能を高めるため、 完全 フロイントアジュバントゃ不完全フロイントアジュバントを投与してもよい。 投 与は、 通常約 2〜 6週毎に 1回ずつ、 計約 3〜1 0回程度行なわれる。  The condensation product is administered to a warm-blooded animal itself or together with a carrier or diluent at a site where antibody production is possible. Complete Freund's adjuvant / incomplete Freund's adjuvant may be administered in order to enhance the antibody-producing ability upon administration. The administration is usually made once every about 2 to 6 weeks, for a total of about 3 to 10 times.
ポリクロ一ナル抗体は、 上記の方法で免疫された温血動物の血液、 腹水など、 好ましくは血液から採取することができる。  The polyclonal antibody can be collected from the blood, ascites, etc., preferably from the blood of a warm-blooded animal immunized by the above method.
抗血清中のポリクローナル抗体価の測定は、 上記の抗血清中の抗体価の測定と 同様にして測定できる。 ポリクロ一ナル抗体の分離精製は、 上記のモノクロ一ナ ル抗体の分離精製と同様の免疫グロプリンの分離精製法に従って行なうことがで さる。  The measurement of the polyclonal antibody titer in the antiserum can be performed in the same manner as the measurement of the antibody titer in the antiserum described above. Separation and purification of the polyclonal antibody can be performed according to the same method for separation and purification of immunoglobulin as in the above-described separation and purification of the monoclonal antibody.
本発明のタンパク質または部分ペプチドをコードする D N A (以下、 アンチセ ンスポリヌクレオチドの説明においては、 これらの D NAを本発明の D NAと略 記する場合がある) の塩基配列に相補的な、 または実質的に相補的な塩基配列ま たはその一部を有するアンチセンスポリヌクレオチドとしては、 本発明の D NA の塩基配列に相補的な、 または実質的に相補的な塩基配列またはその一部を有し、 該 D N Aの発現を抑制し得る作用を有するものであれば、 いずれのアンチセンス ポリヌクレオチドであってもよいが、 アンチセンス D N Aが好ましい。 Complementary to the nucleotide sequence of DNA encoding the protein or partial peptide of the present invention (hereinafter, these DNAs may be abbreviated as the DNA of the present invention in the description of antisense polynucleotide), or An antisense polynucleotide having a substantially complementary base sequence or a part thereof includes a base sequence complementary to or substantially complementary to the base sequence of the DNA of the present invention or a part thereof. Any antisense having an action capable of suppressing the expression of the DNA. Although it may be a polynucleotide, antisense DNA is preferred.
本発明の D NAに実質的に相補的な塩基配列とは、 例えば、 本発明の D NAに 相補的な塩基配列 (すなわち、 本発明の D NAの相補鎖) の全塩基配列あるいは 部分塩基配列と約 7, 0 %以上、 好ましくは約 8 0, %以上、 より好ましくは約 9 0 %以上、 最も好ましくは約 9 5 %以上の相同性を有する塩基配列などが挙げら れる。 特に、 本発明の D NAの相補鎖の全塩基配列うち、 本発明のタンパク質の N末端部位をコードする部分の塩基配列 (例えば、 開始コドン付近の塩基配列な ど) の相補鎖と約 7 0 %以上、 好ましくは約 8 0 %以上、 より好ましくは約 9 0 %以上、 最も好ましくは約 9 5 %以上の相同性を有するアンチセンスポリヌク レオチドが好適である。  The nucleotide sequence substantially complementary to the DNA of the present invention is, for example, the entire nucleotide sequence or a partial nucleotide sequence of the nucleotide sequence complementary to the DNA of the present invention (that is, the complementary strand of the DNA of the present invention). And about 70% or more, preferably about 80% or more, more preferably about 90% or more, and most preferably about 95% or more. In particular, of the total nucleotide sequence of the complementary strand of the DNA of the present invention, the complementary strand of the nucleotide sequence of the portion encoding the N-terminal portion of the protein of the present invention (for example, the nucleotide sequence near the start codon) is about 70%. %, Preferably at least about 80%, more preferably at least about 90%, most preferably at least about 95%.
具体的には、 配列番号: 3、 配列番号: 1 3または配列番号: 2 3で表わされ る塩基配列を有する D NAの塩基配列に相補的な、 もしくは実質的に相補的な塩 基配列、 またはその一部分を有するアンチセンスポリヌクレオチド、 好ましくは 例えば、 配列番号: 3、 配列番号: 1 3または配列番号: 2 3で表わされる塩基 配列を有する D NAの塩基配列に相補な塩基配列、 またはその一部分を有するァ ンチセンスポリヌクレオチドなどが挙げられる。  Specifically, a nucleotide sequence complementary or substantially complementary to the nucleotide sequence of DNA having the nucleotide sequence represented by SEQ ID NO: 3, SEQ ID NO: 13 or SEQ ID NO: 23 Or a part thereof, preferably, for example, a nucleotide sequence complementary to the nucleotide sequence of DNA having the nucleotide sequence represented by SEQ ID NO: 3, SEQ ID NO: 13 or SEQ ID NO: 23, or Antisense polynucleotides having a part thereof are exemplified.
アンチセンスポリヌクレオチドは通常、 1 0〜4 0個程度、 好ましくは 1 5〜 3 0個程度の塩基から構成される。  An antisense polynucleotide is usually composed of about 10 to 40 bases, preferably about 15 to 30 bases.
ヌクレアーゼなどの加水分解酵素による分解を防ぐために、 アンチセンス D N Aを構成する各ヌクレオチドのりん酸残基 (ホスフェート) は、 例えば、 ホスホ ロチォエート、 メチルホスホネート、 ホスホロジチォネートなどの化学修飾りん 酸残基に置換されていてもよい。 これらのアンチセンスポリヌクレオチドは、 公 知の D NA合成装置などを用いて製造することができる。  To prevent degradation by hydrolases such as nucleases, the phosphate residues (phosphates) of each nucleotide constituting the antisense DNA are, for example, chemically modified phosphate residues such as phosphorothioate, methylphosphonate, and phosphorodithionate. May be substituted. These antisense polynucleotides can be produced using a known DNA synthesizer or the like.
本発明に従えば、 本発明のタンパク質遺伝子の複製または発現を阻害すること のできる該遺伝子に対応するアンチセンスポリヌクレオチド (核酸) を、 クロー ンィ匕した、 あるいは決定されたタンパク質をコ一ドする D N Aの塩基配列情報に 基づき設計し、 合成しうる。 かかるアンチセンスポリヌクレオチドは、 本発明の タンパク質遺伝子の R N Aとハイブリダィズすることができ、 該 R N Aの合成ま たは機能を阻害することができるか、 あるいは本発明のタンパク質関連 R NAと の相互作用を介して本発明のタンパク質遺伝子の発現を調節 ·制御することがで きる。 本発明のタンパク質関連 R N Aの選択された配列に相補的なポリヌクレオ チド、.および本発明のタンパク質関連 R N Aと特異的にハイプリダイズすること ができるポリヌクレオチドは、 生体内および生体外で本発明のタンパク質遺伝子 の発現を調節 ·制御するのに有用であり、 また病気などの治療または診断に有用 である。 用語 「対応する」 とは、 遺伝子を含めたヌクレオチド、 塩基配列または 核酸の特定の配列に相同性を有するあるいは相補的であることを意味する。 ヌク レオチド、 塩基配列または核酸とタンパク質との間で 「対応する」 とは、 ヌクレ ォチド (核酸) の配列またはその相補体から誘導される (指令にある) タンパク 質のアミノ酸を通常指している。 タンパク質遺伝子の 5 ' 端ヘアピンループ、According to the present invention, an antisense polynucleotide (nucleic acid) corresponding to the protein gene of the present invention, which can inhibit the replication or expression of the gene, is cloned or the determined protein is encoded. It can be designed and synthesized based on DNA base sequence information. Such an antisense polynucleotide can hybridize to RNA of the protein gene of the present invention, inhibit the synthesis or function of the RNA, or bind to the protein-related RNA of the present invention. Can regulate and control the expression of the protein gene of the present invention through the interaction of Polynucleotides complementary to the selected sequence of the protein-related RNA of the present invention, and polynucleotides capable of specifically hybridizing with the protein-related RNA of the present invention, include the protein of the present invention in vivo and in vitro. It is useful for regulating and controlling gene expression, and is also useful for treating or diagnosing diseases. The term "corresponding" means having homology or being complementary to a specific sequence of nucleotides, base sequences or nucleic acids including genes. The "correspondence" between a nucleotide, nucleotide sequence or nucleic acid and a protein usually refers to the amino acids of the (directed) protein derived from the nucleotide (nucleic acid) sequence or its complement. The 5 'end hairpin loop of the protein gene,
5 ' 端 6—ベースペア ' リピート、 5 ' 端非翻訳領域、 ポリペプチド翻訳開始コ ドン、 タンパク質コード領域、 O R F翻訳終止コドン、 3 ' 端非翻訳領域、 3 ' 端パリンドローム領域または 3 ' 端ヘアピンループなどは、 好ましい対象領域と して選択しうるが、 タンパク質遺伝子内の如何なる領域も対象として選択しうる 目的核酸と、 対象領域の少なくとも一部に相補的なポリヌクレオチドとの関係 については、 目的核酸が対象領域とハイブリダィズすることができる場合は、 そ の目的核酸は、 当該対象領域のポリヌクレオチドに対して 「アンチセンス」 であ るということができる。 ァンチセンスポリヌクレオチドは、 2ーデォキシー D— リポースを含有しているポリデォキシリポヌクレオチド、 D—リポースを含有し ているポリリポヌクレオチド、 プリンまたはピリミジン塩基の N—グリコシドで あるその他のタイプのポリヌクレオチド、 非ヌクレオチド骨格を有するその他の ポリマー (例えば、 市販のタンパク質核酸および合成配列特異的な核酸ポリマ 一) または特殊な結合を含有するその他のポリマー (但し、 該ポリマーは D N A や R N A中に見出されるような塩基のペアリングや塩基の付着を許容する配置を もつヌクレオチドを含有する) などが挙げられる。 それらは、 2本鎖 D NA、 1 本鎖 D N A、 2本鎖 R NA、 1本鎖 R N A、 D NA: R NAハイブリッドであつ てもよく、 さらに非修飾ポリヌクレオチド (または非修飾オリゴヌクレオチド) 、 公知の修飾の付加されたもの、 例えば当該分野で知られた標識のあるもの、 キヤ ップの付いたもの、 メチル化されたもの、 1個以上の天然のヌクレオチドを類縁 物で置換したもの、 分子内ヌクレオチド修飾のされたもの、 例えば非荷電結合5 'end 6—base pair' repeat, 5 'end untranslated region, polypeptide translation initiation codon, protein coding region, ORF translation stop codon, 3' end untranslated region, 3 'end palindrome region or 3' end Although a hairpin loop or the like can be selected as a preferred target region, any region in a protein gene can be selected as a target. Regarding the relationship between a target nucleic acid and a polynucleotide complementary to at least a part of the target region, If the nucleic acid of interest can hybridize to the target region, the nucleic acid of interest can be said to be "antisense" to the polynucleotide of the target region. Antisense polynucleotides can be polydexoxy liponucleotides containing 2-deoxy D-reports, polyliponucleotides containing D-reports, N-glycosides of purine or pyrimidine bases and other types of polynucleotides. Other polymers having nucleotide or non-nucleotide backbones (eg, commercially available protein nucleic acids and synthetic sequence-specific nucleic acid polymers) or other polymers containing special bonds (provided that such polymers are found in DNA or RNA) Base pairing and nucleotides having a configuration that allows base attachment). They may be double-stranded DNA, single-stranded DNA, double-stranded RNA, single-stranded RNA, DNA: RNA hybrids, as well as unmodified polynucleotides (or unmodified oligonucleotides), Related modifications with known modifications, e.g., those with labels, caps, methylated, one or more natural nucleotides known in the art Substituted, Intramolecular nucleotide modification, eg uncharged binding
(例えば、 メチルホスホネート、 ホスホトリエステル、 ホスホルアミデート、 力 ルバメートなど) を持つもの、 電荷を有する結合または硫黄含有結合 (例、 ホス ホロチォエート、 ホスホロジチォェ一トなど) を持つもの、 例えばタンパク質 (Eg, having a methylphosphonate, phosphotriester, phosphoramidate, potassium salt, etc.), having a charged bond or a sulfur-containing bond (eg, phosphorothioate, phosphorodithioate, etc.), eg, protein
(例、 ヌクレアーゼ、 ヌクレア一ゼ ·インヒビター、 トキシン、 抗体、 シグナル ペプチド、 ポリ一 L _リジンなど) や糖 (例、 モノサッカライドなど) などの側 鎖基を有しているもの、 インターカレント化合物 (例、 ァクリジン、 ソラレンな ど) を持つもの、 キレート化合物 (例えば、 金属、 放射活性をもつ金属、 ホウ素、 酸化性の金属など) を含有するもの、 アルキル化剤を含有するもの、 修飾された 結合を持つもの (例えば、 ひァノマー型の核酸など) であってもよい。 ここで 「ヌクレオシド」 、 「ヌクレオチド」 および 「核酸」 とは、 プリンおよびピリミ ジン塩基を含有するのみでなく、 修飾されたその他の複素環型塩基をもつような ものを含んでいて良い。 このような修飾物は、 メチル化されたプリンおよびピリ ミジン、 ァシル化されたプリンおよびピリミジン、 あるいはその他の複素環を含 むものであってよい。 修飾されたヌクレオチドおよび修飾されたヌクレオチドは また糖部分が修飾されていてよく、 例えば、 1個以上の水酸基がハロゲンとか、 脂肪族基などで置換されていたり、 またはエーテル、 ァミンなどの官能基に変換 . されていてよい。  (Eg, nucleases, nuclease inhibitors, toxins, antibodies, signal peptides, poly-L-lysine, etc.) and sugars (eg, monosaccharides), etc. Eg, containing acridine, psoralen, etc., containing chelating compounds (eg, metals, radioactive metals, boron, oxidizing metals, etc.), containing alkylating agents, modified bonds (Eg, a anomeric nucleic acid). Here, "nucleoside", "nucleotide", and "nucleic acid" may include not only those containing purine and pyrimidine bases but also those having other modified heterocyclic bases. Such modifications may include methylated purines and pyrimidines, acylated purines and pyrimidines, or other heterocycles. Modified nucleotides and modified nucleotides may also be modified at the sugar moiety, e.g., where one or more hydroxyl groups have been replaced with a halogen, an aliphatic group, or the like, or a functional group, such as an ether or an amine. Conversion.
本発明のアンチセンスポリヌクレオチドは、 R N A、 D NAまたは修飾された 核酸 (R N A、 D N A) である。 修飾された核酸の具体例としては、 核酸の硫黄 誘導体、 チォホスフェート誘導体、 ポリヌクレオシドアミドゃオリゴヌクレオシ ドアミドの分解に抵抗性のものなどが挙げられる。 本発明のアンチセンスポリヌ クレオチドは、 例えば、 以下のように設計されうる。 すなわち、 細胞内でのアン チセンスポリヌクレオチドをより安定なものにする、 アンチセンスポリヌクレオ チドの細胞透過性をより高める、 目標とするセンス鎖に対する親和性をより大き なものにする、 また、 もし毒性があるような場合はアンチセンスポリヌクレオチ ドの毒性をより小さなものにする。 このような修飾は、 例えば Pharm Tech Japan, 8巻, 247頁または 395頁, 1992年、 Ant i sense Research and Appl i cat i ons, CRC Press, 1993年などで数多く報告されている。 本発明のアンチセンスポリヌクレオチドは、 変化せしめられたり、 修飾された 糖、 塩基、 結合を含有していて良く、 リボゾーム、 ミクロスフエアのような特殊 な形態で供与されたり、 遺 子治療により適用されたり、 付加された形態で与え られることができうる。 こうして付加形態で用いられるものとしては、 リン酸基 骨格の電荷を中和するように働くポリリジンのようなポリカチオン体、 細胞膜と の相互作用を高めたり、 核酸の取込みを増大せしめるような脂質 (例、 ホスホリ .ピド、. コレステロールなど) などの疎水性のものが挙げられる。 付加するに好ま しい脂質としては、 コレステロールやその誘導体 (例、 コレステリルクロ口ホル メート、 コール酸など) が挙げられる。 こうしたものは、 核酸の 3 ' 端または 5 ' 端に付着させることができ、 塩基、 糖、 分子内ヌクレオシド結合を介して付 着させることができうる。 その他の基としては、 核酸の 3 ' 端または 5 ' 端に特 異的に配置されたキャップ用の基で、 ェキソヌクレア一ゼ、 R N a s eなどのヌ クレア一ゼによる分解を阻止するためのものが挙げられる。 こうしたキャップ用 の基としては、 ポリエチレングリコール、 テトラエチレングリコールなどのダリ コ一ルをはじめとした当該分野で知られた水酸基の保護基が挙げられるが、 それ に限定されるものではない。 The antisense polynucleotide of the present invention is RNA, DNA or a modified nucleic acid (RNA, DNA). Specific examples of the modified nucleic acid include a sulfur derivative of a nucleic acid, a thiophosphate derivative, a polynucleoside amide / a nucleic acid that is resistant to degradation of oligonucleoside amide, and the like. The antisense polynucleotide of the present invention can be designed, for example, as follows. In other words, the antisense polynucleotide is more stable in the cell, the cell permeability of the antisense polynucleotide is increased, the affinity for the target sense strand is increased, and If so, reduce the toxicity of the antisense polynucleotide. Many such modifications are reported, for example, in Pharm Tech Japan, 8, 247 or 395, 1992, Antisense Research and Appli cations, CRC Press, 1993. The antisense polynucleotides of the present invention may contain altered or modified sugars, bases, or bonds, may be provided in special forms such as ribosomes, microspheres, or may be applied by genetic therapy. Can be given in an added form. Thus, the addition forms include polycations such as polylysine, which acts to neutralize the charge on the phosphate backbone, and lipids, which enhance the interaction with cell membranes and increase the uptake of nucleic acids ( For example, phosphoric acid, hydrophobic, cholesterol, etc.). Preferred lipids for addition include cholesterol and its derivatives (eg, cholesteryl chromate formate, cholic acid, etc.). These can be attached to the 3 'or 5' end of the nucleic acid and can be attached via a base, sugar, or intramolecular nucleoside linkage. Other groups are cap groups specifically located at the 3 'or 5' end of nucleic acids that prevent degradation by nucleases such as exonuclease and RNase. No. Examples of such a capping group include, but are not limited to, hydroxyl-protecting groups known in the art, such as diols such as polyethylene glycol and tetraethylene glycol.
アンチセンスポリヌクレオチドの阻害活性は、 本発明の形質転換体、 本発明の 生体内や生体外の遺伝子発現系、 または本発明の夕ンパク質の生体内や生体外の 翻訳系を用いて調べることができる。 以下に、 本発明のタンパク質もしくは部分ペプチドまたはその塩 (以下、 本発 明のタンパク質と略記する場合がある) 、 本発明のタンパク質または部分べプチ ドをコードする D NA (以下、 本発明の D NAと略記する場合がある) 、 本発明 のタンパク質もしくは部分ペプチドまたはその塩に対する抗体 (以下、 本発明の 抗体と略記する場合がある) 、 および本発明の D NAのアンチセンスポリヌクレ ォチド (以下、 本発明のアンチセンスポリヌクレオチドと略記する場合がある) の用途などを説明する。  The inhibitory activity of the antisense polynucleotide can be examined using the transformant of the present invention, the in vivo or in vitro gene expression system of the present invention, or the in vivo or in vitro translation system of the protein of the present invention. Can be. Hereinafter, the protein or partial peptide of the present invention or a salt thereof (hereinafter sometimes abbreviated as the protein of the present invention), the DNA encoding the protein or partial peptide of the present invention (hereinafter referred to as the D NA), an antibody against the protein or partial peptide of the present invention or a salt thereof (hereinafter may be abbreviated as the antibody of the present invention), and an antisense polynucleotide of the DNA of the present invention (hereinafter may be referred to as an antibody). , Which may be abbreviated as the antisense polynucleotide of the present invention).
〔1〕 本発明のタンパク質が関与する各種疾病の予防 ·治療剤  [1] Agent for preventing and treating various diseases related to the protein of the present invention
本発明の夕ンパク質は G夕ンパク質共役型レセプ夕一タンパク質であり、 M C Hと結合するため、 食欲 (摂食) 増進作用、 ォキシトシン分泌促進作用などの重 要な役割を果たしている。 The protein of the present invention is a G protein-coupled receptor protein, MC By binding to H, it plays an important role in promoting appetite (feeding) and promoting oxytocin secretion.
したがって、 本発明のタンパク質をコードする D NAに異常があったり、 欠損 している場合あるいは本発明のタンパク質の発現量が減少している場合には、 例 えば、 食欲不振 (例、 神経性食欲不振症など) 、 食欲不振に伴う貧血または低蛋 白症、 微弱陣痛、 弛緩出血、 子宮復古不全、 乳汁うっ滞などの種々の疾患が発症 する。  Therefore, when the DNA encoding the protein of the present invention is abnormal or defective, or when the expression level of the protein of the present invention is reduced, for example, anorexia (eg, nervous appetite) Anemia or hypoproteinia due to anorexia, weak labor, laxative bleeding, uterine remodeling failure, milk stasis, etc. develop various diseases.
したがって、 本発明のタンパク質および本発明の D NAは、 例えば、 食欲 (摂 食) 増進剤、 食欲不振 (例、 神経性食欲不振症など) 、 食欲不振に伴う貧血また は低蛋白症、 微弱陣痛、 弛緩出血、 子宮復古不全、 乳汁うっ滞などの予防 ·治療 剤などの医薬として使用することができる。  Therefore, the protein of the present invention and the DNA of the present invention include, for example, an appetite (feeding) enhancer, anorexia (eg, anorexia nervosa), anemia or hypoproteinemia associated with anorexia, weak labor pain It can be used as a medicament such as a preventive and remedy for flaccid hemorrhage, uterine remodeling failure, and milk stasis.
例えば、 生体内において本発明のタンパク質が減少あるいは欠損しているため に、 M C Hの食欲 (摂食) 増進またはォキシトシン分泌促進活性が十分に、 ある いは正常に発揮されない患者がいる場合に、 (ィ) 本発明の D NAを該患者に投 与し、 生体内で本発明のタンパク質を発現させることによって、 (口) 細胞に本 発明の D N Aを挿入し、 本発明のタンパク質を発現させた後に、 該細胞を患者に 移植することによって、 または (八) 本発明のタンパク質を該患者に投与するこ となどによって、 該患者における本発明のタンパク質の役割を十分に、 あるいは 正常に発揮させることができる。  For example, if the protein of the present invention is reduced or deficient in the living body, and there is a patient in which the appetite (feeding) enhancement or oxytocin secretion promoting activity of MCH is not sufficiently or normally exerted, B) by administering the DNA of the present invention to the patient and expressing the protein of the present invention in vivo, by inserting the DNA of the present invention into cells and expressing the protein of the present invention. By transplanting the cells into a patient, or (8) administering the protein of the present invention to the patient, etc., to fully or normally exert the role of the protein of the present invention in the patient. it can.
本発明の D NAを上記の予防 ·治療剤として使用する場合は、 該 D NAを単独 あるいはレトロウイルスベクター、 アデノウイルスベクター、 アデノウイルスァ ソシエーテツドウィルスベクタ一などの適当なベクターに揷入した後、 常套手段 に従って、 ヒトまたは温血動物に投与することができる。 本発明の D N Aは、 そ のままで、 あるいは摂取促進のための補助剤などの生理学的に認められる担体と ともに製剤化し、 遺伝子銃やハイド口ゲルカテーテルのようなカテーテルによつ て投与できる。  When the DNA of the present invention is used as the above-described prophylactic or therapeutic agent, the DNA is used alone or in an appropriate vector such as a retrovirus vector, an adenovirus vector, or an adenovirus associated virus vector. Thereafter, it can be administered to humans or warm-blooded animals according to conventional means. The DNA of the present invention can be administered as it is or in the form of a formulation together with a physiologically acceptable carrier such as an auxiliary agent for promoting uptake, and can be administered using a gene gun or a catheter such as a hide mouth gel catheter.
本発明のタンパク質を上記の予防 ·治療剤として使用する場合は、 少なくとも 9 0 %、 好ましくは 9 5 %以上、 より好ましくは 9 8 %以上、 さらに好ましくは 9 9 %以上に精製されたものを使用するのが好ましい。 本発明のタンパク質は、 例えば、 必要に応じて糖衣を施した錠剤、 カプセル剤、 エリキシル剤、 マイクロカプセル剤などとして経口的に、 あるいは水もしくはそ れ以外の薬学的に許容し得る液との無菌性溶液、 または懸濁液剤などの注射剤の 形で非経口的に使用できる。 例えば、 本発明のタンパク質を生理学的に認められ る担体、 香味剤、 賦形剤、 べヒクル、 防腐剤、 安定剤、 結合剤などとともに一般 に認められた製剤実施に要求される単位用量形態で混和することによって製造す ることができる。 これら製剤における有効成分量は指示された範囲の適当な用量 が得られるようにするものである。 ' ' 錠剤、 カプセル剤などに混和することができる添加剤としては、 例えば、 ゼラ チン、 コーンスターチ、 トラガント、 アラビアゴムのような結合剤、 結晶性セル ロースのような賦形剤、 コーンスターチ、 ゼラチン、 アルギン酸などのような膨 化剤、 ステアリン酸マグネシウムのような潤滑剤、 ショ糖、 乳糖またはサッカリ ンのような甘味剤、 ペパーミント、 ァカモノ油またはチェリ一のような香味剤な どが用いられる。 調剤単位形態がカプセルである場合には、 前記タイプの材料に さらに油脂のような液状担体を含有することができる。 注射のための無菌組成物 は注射用水のようなべヒクル中の活性物質、 胡麻油、 椰子油などのような天然産 出植物油などを溶解または懸濁させるなどの通常の製剤実施に従って処方するこ とができる。 When the protein of the present invention is used as the prophylactic / therapeutic agent, the protein purified to at least 90%, preferably 95% or more, more preferably 98% or more, and still more preferably 99% or more. It is preferred to use. The protein of the present invention can be used, for example, in the form of tablets, capsules, elixirs, microcapsules, and the like, which are sugar-coated as necessary, orally, or aseptically with water or other pharmaceutically acceptable liquids. It can be used parenterally in the form of injections, such as aqueous solutions or suspensions. For example, the protein of the present invention may be used together with physiologically acceptable carriers, flavoring agents, excipients, vehicles, preservatives, stabilizers, binders, and the like in a unit dosage form required for generally accepted pharmaceutical practice. It can be manufactured by mixing. The amount of the active ingredient in these preparations is such that a suitable dosage in the specified range can be obtained. '' Additives that can be incorporated into tablets, capsules, etc. include, for example, binders such as gelatin, corn starch, tragacanth, gum arabic, excipients such as crystalline cellulose, corn starch, gelatin, Swelling agents such as alginic acid, lubricants such as magnesium stearate, sweeteners such as sucrose, lactose or saccharine, and flavoring agents such as peppermint, cocoa oil or cellulose. When the preparation unit form is a capsule, a liquid carrier such as oil and fat can be further contained in the above-mentioned type of material. Sterile compositions for injection can be formulated according to standard pharmaceutical practice, such as dissolving or suspending the active substance in vehicles such as water for injection, and naturally occurring vegetable oils such as sesame oil and coconut oil. it can.
注射用の水性液としては、 例えば、 生理食塩水、 ブドウ糖やその他の補助薬を 含む等張液 (例えば、 D—ソルビトール、 D—マンニトール、 塩化ナトリウムな ど) などが挙げられ、 適当な溶解補助剤、 例えば、 アルコール (例えば、 ェ夕ノ ールなど) 、 ポリアルコール (例えば、 プロピレンダリコール、 ポリエチレング .リコールなど) 、 非イオン性界面活性剤 (例えば、 ポリソルべ一卜 8 0 TM、 H C Ο—5 0など) などと併用してもよい。 油性液としては、 例えば、 ゴマ油、 大豆 , 油などが挙げられ、 溶解補助剤として安息香酸ベンジル、 ベンジルアルコールな どと併用してもよい。 また、 緩衝剤 (例えば、 リン酸塩緩衝液、 酢酸ナトリウム , 緩衝液など) 、 無痛化剤 (例えば、 塩化ベンザルコニゥム、 塩酸プロ力インな Aqueous liquids for injection include, for example, physiological saline, isotonic solutions containing glucose and other adjuvants (eg, D-sorbitol, D-mannitol, sodium chloride, etc.). Agents, for example, alcohols (eg, alcohol), polyalcohols (eg, propylene glycol, polyethylene glycol, etc.), non-ionic surfactants (eg, Polysorbate 80 , HC Ο—50 etc.). Examples of the oily liquid include sesame oil, soybean, and oil, and may be used in combination with a solubilizing agent such as benzyl benzoate or benzyl alcohol. In addition, buffers (eg, phosphate buffer, sodium acetate, buffer, etc.), soothing agents (eg, benzalkonium chloride, proforce hydrochloride, etc.)
ど) 、 安定剤 (例えば、 ヒ卜血清アルブミン、 ポリエチレングリコールなど) 、 保存剤 (例えば、 ベンジルアルコール、 フエノールなど) 、 酸化防止剤などと配 ' 合してもよい。 調製された注射液は、 通常、 適当なアンプルに充填される。 本発明の D NAが挿入されたベクターも上記と同様に製剤化され、 通常、 非経 口的に使用される。 ), Stabilizers (eg, human serum albumin, polyethylene glycol, etc.), preservatives (eg, benzyl alcohol, phenol, etc.), antioxidants, etc. 'May be combined. The prepared injection is usually filled in an appropriate ampoule. The vector into which the DNA of the present invention has been inserted is also formulated in the same manner as described above, and is usually used parenterally.
このようにして得られる製剤は、 安全で低毒性であるので、 例えば、 温血動物 (例えば、 ヒト、 ラット、 マウス、 モルモット、 ゥサギ、 トリ、 ヒッジ、 ブ夕、 ゥシ、 ゥマ、 ネコ、 ィヌ、 サル、 チンパンジーなど) に対して投与することがで きる。  The preparations obtained in this way are safe and have low toxicity, for example, in warm-blooded animals (e.g., humans, rats, mice, guinea pigs, egrets, birds, higgies, bush, pests, pests, cats, Dogs, monkeys, chimpanzees, etc.).
本発明のタンパク質の投 量は、 対象疾患、 投与対象、 投与ルートなどにより 差異はあるが、 例えば、 腎不全の治療目的で本発明のタンパク質を経口投与する 場合、 一般的に成人 (60kgとして) においては、 一日につき該タンパク質を約 0. 1〜100mg、 好ましくは約 1. (!〜 50mg、 より好ましくは約 1. 0〜20mg投与する。 非 経口的に投与する場合は、 該タンパク質の 1回投与量は投与対象、 対象疾患など によっても異なるが、 例えば、 神経性食欲不振症の治療目的で本発明のタンパク 質を注射剤の形で成人 (体重 60kgとして) に投与する場合、 一日につき該タンパ ク質を約 0. 01〜30mg程度、 好ましくは約 0. l〜20mg程度、 より好ましくは約 0. 1〜 l Omg程度を患部に注射することにより投与するのが好都合である。 他の動物の場 合も、 60kg当たりに換算した量を投与することができる。  The dosage of the protein of the present invention varies depending on the target disease, the administration subject, the administration route, and the like. For example, when the protein of the present invention is orally administered for the purpose of treating renal failure, generally, the adult (as 60 kg) is used. Is administered in an amount of about 0.1 to 100 mg, preferably about 1. (! To 50 mg, more preferably about 1.0 to 20 mg. Per day. The single dose varies depending on the administration subject, the target disease, and the like. For example, when the protein of the present invention is administered to an adult (with a body weight of 60 kg) in the form of an injection for the purpose of treating anorexia nervosa, It is convenient to administer the protein by injecting about 0.01 to 30 mg, preferably about 0.1 to 20 mg, more preferably about 0.1 to 10 mg of the protein per day into the affected area. 60 kg for other animals Amount converted to have enough can be administered.
〔2〕 疾病に対する医薬候補化合物のスクリーニング [2] Screening of drug candidate compounds for disease
' 本発明のタンパク質は、 本発明のペプチド (例、 M C H) と本発明のタンパク 質との結合を変化させる化合物またはその塩のスクリーニングのための試薬とし て有用である。  'The protein of the present invention is useful as a reagent for screening a compound or a salt thereof that changes the binding between the peptide of the present invention (eg, MCH) and the protein of the present invention.
本発明のぺプチドおよび本発明のタンパク質 (本発明のタンパク質の部分ぺプ チドも含む) を用いることを特徴とする、 本発明のペプチドと本発明のタンパク 質との結合性を変化させる化合物またはその塩のスクリーニング方法、 または本 発明のペプチドおよび本発明のタンパク質を用いることを特徴とする本発明のぺ プチドと本発明のタンパク質との結合性を変化させる化合物またはその塩のスク リーニング用キット (以下、 本発明のスクリーニング方法、 本発明のスクリー二 ング用キッ小と略記する) について以下に詳述する。 本発明のタンパク質を用いるか、 または組換え型本発明の夕ンパク質の発現系 を構築し、 該発現系を用いた本発明のペプチドとの結合アツセィ系 (リガンド - レセプターアツセィ系) を用いることによって、 本発明のペプチドと本発明の夕 ンパク質との結合性を変化させる化合物、 本発明のペプチドと本発明のタンパク 質との結合性を変化させる化合物 (例えば、 ペプチド、 タンパク質、 非ペプチド 性化合物、 合成化合物、 発酵生産物など) またはその塩をスクリーニングするこ とができる。 A compound or a compound that alters the binding between the peptide of the present invention and the protein of the present invention, which comprises using the peptide of the present invention and the protein of the present invention (including a partial peptide of the protein of the present invention). A method for screening a salt thereof, or a kit for screening a compound or a salt thereof, which changes the binding property between the peptide of the present invention and the protein of the present invention, which comprises using the peptide of the present invention and the protein of the present invention ( Hereinafter, the screening method of the present invention and the kit for screening of the present invention are abbreviated in detail below. Using the protein of the present invention, or constructing a recombinant expression system of the protein of the present invention, and using a binding assay system (ligand-receptor assay system) with the peptide of the present invention using the expression system. Thus, a compound that alters the binding between the peptide of the present invention and the protein of the present invention, a compound that alters the binding between the peptide of the present invention and the protein of the present invention (for example, peptide, protein, non-peptide) Compounds, synthetic compounds, fermentation products, etc.) or salts thereof.
このような化合物には、 本発明のタンパク質を介して細胞刺激活性 (例えば、 ァラキドン酸遊離、 アセチルコリン遊離、 細胞内 C a 2 +遊離、 細胞内 c AM P生 成、 細胞内 c AM P産生抑制、 細胞内 c GM P生成、 ィノシトールリン酸産生、 細胞膜電位変動、 細胞内タンパク質のリン酸化、 c一 f o sの活性化、 p Hの低 下、 c AM P依存性プロティンキナーゼの活性化、 c GM P依存性プロティンキ ナ一ゼの活性化、 リン脂質依存性プロテインキナーゼの活性化、 微小管結合蛋白 質リン酸化酵素 (MA Pキナーゼ) の活性化などを促進する活性または抑制する 活性など) を有する化合物 (ァゴ二スト) と該細胞刺激活性を有しない化合物 (アンタゴニスト) などが含まれる。 「本発明のペプチドと本発明のタンパク質 との結合性を変化させる」 とは、 本発明のペプチドと本発明のタンパク質との結 合を阻害する場合と促進する場合の両方を包含するものである。 Such compounds, cell stimulating activity via protein of the present invention (e.g., Arakidon acid release, acetylcholine release, intracellular C a 2 + release, intracellular c AM P producing formation, intracellular c AM P production inhibition , Intracellular cGMP production, Inositol phosphate production, Cell membrane potential fluctuation, Intracellular protein phosphorylation, c-fos activation, pH decrease, cAMP-dependent protein kinase activation, cGMP Activation of protein-dependent protein kinase, activation of phospholipid-dependent protein kinase, activity of promoting or inhibiting the activation of microtubule-associated protein kinase (MAP kinase), etc. Compounds (agonist) and compounds (antagonists) having no such cell stimulating activity. "Altering the binding between the peptide of the present invention and the protein of the present invention" includes both cases of inhibiting and promoting the binding of the peptide of the present invention to the protein of the present invention. .
すなわち、 本発明は、 (i) 本発明のタンパク質に、 本発明のペプチドを接触 させた場合と (i i) 上記した本発明のタンパク質に、 本発明のペプチドおよび試 験化合物を接触させた場合との比較を行なうことを特徴とする本発明のぺプチド と本発明のタンパク質との結合性を変化させる化合物またはその塩のスクリ一二 ング方法を提供する。  That is, the present invention relates to (i) the case where the protein of the present invention is brought into contact with the peptide of the present invention, and (ii) the case where the above-mentioned protein of the present invention is brought into contact with the peptide of the present invention and a test compound. And a method for screening a compound or a salt thereof, which changes the binding property between the peptide of the present invention and the protein of the present invention, characterized in that the comparison is performed.
本発明のスクリーニング方法においては、 (i) 上記した本発明のタンパク質 に本発明のペプチドを接触させた場合と (i i) 上記した本発明のタンパク質に本 発明のぺプチドおよび試験化合物を接触させた場合における、 例えば該本発明の 夕ンパク質に対する本発明のぺプチドの結合量、 細胞刺激活性などを測定して比 較する。  In the screening method of the present invention, (i) the case where the above-mentioned protein of the present invention is brought into contact with the peptide of the present invention and (ii) the above-mentioned protein of the present invention which comes in contact with the peptide of the present invention and a test compound. In such a case, for example, the amount of the peptide of the present invention bound to the protein of the present invention, the cell stimulating activity and the like are measured and compared.
本発明のスクリーニング方法としての具体例としては、 例えば、 (a) 本発明のペプチドを本発明のタンパク質に接触させた場合と、 本発明のぺ プチドおよび試験化合物を本発明のタンパク質に接触させた場合における、 本発 明のペプチドの本発明のタンパク質に対する結合量を測定し、 比較することを特 徵とする、 本発明のペプチドと本発明のタンパク質との結合性を変化させる化合 物またはその塩のスクリーニング方法、 Specific examples of the screening method of the present invention include, for example, (a) the peptide of the present invention relative to the protein of the present invention when the peptide of the present invention is brought into contact with the protein of the present invention, and when the peptide and the test compound of the present invention are brought into contact with the protein of the present invention; A method for screening a compound or a salt thereof that changes the binding property between the peptide of the present invention and the protein of the present invention, which comprises measuring and comparing the amount of binding;
(b) 本発明のペプチドを、 本発明のタンパク質を含有する細胞または該細胞の 膜画分に接触させた場合と、 本発明のぺプチドおよび試験化合物を本発明のタン パク質を含有する細胞または該細胞の膜画分に接触させた場合における、 本発明 のべプチドの該細胞または該膜画分に対する結合量を測定し、 比較することを特 徴とする、 本発明のペプチドと本発明のタンパク質との結合性を変化させる化合 物またはその塩のスクリーニング方法、 および  (b) when the peptide of the present invention is brought into contact with a cell containing the protein of the present invention or a membrane fraction of the cell, and a cell containing the peptide and the test compound of the present invention and the protein of the present invention. Alternatively, the peptide of the present invention and the peptide of the present invention are characterized by measuring and comparing the amount of binding of the peptide of the present invention to the cell or the membrane fraction when brought into contact with the membrane fraction of the cell. A method for screening a compound or a salt thereof that changes the binding property to a protein, and
(c) 本発明のタンパク質が、 本発明のタンパク質をコードする D NAを含有す る形質転換体を培養することによって細胞膜上に発現した本発明のタンパク質で ある上記 (b) 記載のスクリーニング方法、  (c) the screening method according to the above (b), wherein the protein of the present invention is a protein of the present invention expressed on a cell membrane by culturing a transformant containing a DNA encoding the protein of the present invention;
(d) 本発明のペプチドが、 標識したペプチドである上記 (a) 〜 (c) のスクリ —ニング方法などのレセプ夕一結合アツセィ系、  (d) the peptide of the present invention is a labeled peptide, such as the screening method according to any one of (a) to (c) above,
(e) 本発明のペプチドを本発明のタンパク質に接触させた場合と、 本発明のぺ プチドおよび試験化合物を本発明のタンパク質に接触させた場合における、 本発 明のタンパク質を介した細胞刺激活性を測定し、 比較することを特徴とする、 本 発明のペプチドと本発明のタンパク質との結合性を変化させる化合物またはその 塩のスクリ一ニング方法、  (e) Cell stimulating activity mediated by the protein of the present invention when the peptide of the present invention is brought into contact with the protein of the present invention and when the peptide of the present invention and a test compound are brought into contact with the protein of the present invention A method of screening a compound or a salt thereof, which changes the binding property between the peptide of the present invention and the protein of the present invention, which comprises measuring and comparing
(f) 本発明のペプチドを本発明のタンパク質を含有する細胞または該細胞の膜 画分に接触させた場合と、 本発明のペプチドおよび試験化合物を本発明のタンパ ク質を含有する細胞または該細胞の膜画分に接触させた場合における、 本発明の タンパク質を介した細胞刺激活性を測定し、 比較することを特徴とする、 本発明 のペプチドと本発明のタンパク質との結合性を変化させる化合物またはその塩の スクリーニング方法、 および  (f) when the peptide of the present invention is brought into contact with a cell containing the protein of the present invention or a membrane fraction of the cell, and the peptide or test compound of the present invention or the cell containing the protein of the present invention or It changes the binding property between the peptide of the present invention and the protein of the present invention, which is characterized by measuring and comparing the cell stimulating activity mediated by the protein of the present invention when brought into contact with the membrane fraction of the cell. A method for screening a compound or a salt thereof, and
(g) 本発明のタンパク質が、 本発明のタンパク質をコードする D N Aを含有す る形質転換体を培養することによって細胞膜上に発現した本発明のタンパク質で ある上記 (f) のスクリーニング方法などの細胞刺激アツセィ系などが挙げられ る。 (g) a protein of the present invention expressed on a cell membrane by culturing a transformant containing a DNA encoding the protein of the present invention; A cell stimulating Atsay system such as the screening method of the above (f) and the like can be mentioned.
本発明のスクリーニング方法の具体的な説明を以下にする。  The specific description of the screening method of the present invention is as follows.
まず、 本発明のスクリーニング方法に用いる本発明のタンパク質としては、 上 記の本発明のタンパク質を含有するものであれば何れのものであってもよい。 例 えば、 組換え体を用いて大量発現させた本発明のタンパク質などが適している。 本発明のタンパク質の製造には、 前述の方法などが用いられる。  First, the protein of the present invention used in the screening method of the present invention may be any protein containing the above-described protein of the present invention. For example, the protein of the present invention which is expressed in a large amount using a recombinant is suitable. For the production of the protein of the present invention, the aforementioned methods and the like are used.
本発明のスクリーニング方法において、 本発明のタンパク質を含有する細胞あ るいは該細胞膜画分などを用いる場合、 後述の調製法に従えばよい。  In the screening method of the present invention, when cells containing the protein of the present invention or the cell membrane fraction are used, the preparation method described later may be followed.
本発明のタンパク質を含有する細胞を用いる場合、 該細胞をダルタルアルデヒ ド、 ホルマリンなどで固定化してもよい。 固定化方法は公知の方法に従って行う ことができる。 ' 本発明のタンパク質を含有する細胞としては、 本発明のタンパク質を発現した 宿主細胞をいうが、 該宿主細胞としては、 前述の大腸菌、 枯草菌、 酵母、 昆虫細 胞、 動物細胞などがあげられる。  When a cell containing the protein of the present invention is used, the cell may be immobilized with datalaldehyde, formalin, or the like. The immobilization method can be performed according to a known method. '' The cell containing the protein of the present invention refers to a host cell expressing the protein of the present invention. Examples of the host cell include the aforementioned Escherichia coli, Bacillus subtilis, yeast, insect cells, animal cells and the like. .
膜画分としては、 細胞を破碎した後、 公知の方法で得られる細胞膜が多く含ま れる画分のことをいう。 細胞の破砕方法としては、 Pot ter—Elvehj em型ホモジナ ィザ一で細 を押し潰す方法、 ワーリンダブレンダーゃポリトロン (Kinemat i ca 社製) による破砕、 超音波による破砕、 フレンチプレスなどで加圧しながら細胞 を細いノズルから噴出させることによる破砕などがあげられる。 細胞膜の分画に は、 分画遠心分離法や密度勾配遠心分離法などの遠心力による分画法が主として 用いられる。 例えば、 細胞破碎液を低速 (500〜3000rpm) で短時間 (通常、 約 1 〜10分) 遠心し、上清をさらに高速 (15000〜30000rpm) で通常 30分〜 2時間遠心 し、 得られる沈澱を膜画分とする。 該膜画分中には、 発現した本発明のタンパク 質と細胞由来のリン脂質や膜タンパク質などの膜成分が多く含まれる。  The membrane fraction refers to a fraction containing a large amount of cell membrane obtained by a known method after cell disruption. The cells can be crushed by crushing the cells with a Potter-Elvehj em homogenizer, crushing with a Warlinda blender-Polytron (Kinematica), crushing with ultrasonic waves, or pressing with a French press. Crushing by ejecting cells from thin nozzles. For cell membrane fractionation, centrifugal fractionation methods such as differential centrifugation and density gradient centrifugation are mainly used. For example, the cell lysate is centrifuged at a low speed (500-3000 rpm) for a short time (usually about 1-10 minutes), and the supernatant is further centrifuged at a high speed (15000-30,000 rpm) for 30 minutes to 2 hours to obtain a precipitate. Is the membrane fraction. The membrane fraction is rich in the expressed protein of the present invention and membrane components such as cell-derived phospholipids and membrane proteins.
該本発明のタンパク質を含有する細胞や膜画分中の本発明のタンパク質の量は、 1細胞当たり 103〜 108分子であるのが好ましく、 105〜 107分子であるのが好適であ る。 なお、 発現量が多いほど膜画分当たりのリガンド結合活性 (比活性) が高く なり、 高感度なスクリーニング系の構築が可能になるばかりでなく、 同一ロット で大量の試料を測定できるようになる。 The amount of the protein of the present invention in the cells or membrane fraction containing the protein of the present invention is preferably 10 3 to 10 8 molecules per cell, more preferably 10 5 to 10 7 molecules per cell. is there. The higher the expression level, the higher the ligand binding activity (specific activity) per membrane fraction, which not only enables the construction of a highly sensitive screening system, but also allows the same lot Can measure a large amount of samples.
前記のレセプター結合アツセィ系や細胞刺激アツセィ系などのスクリーニング 方法を実施するためには、 例えば、 本発明のタンパク質画分と、 本発明のぺプチ ド (例、 標識した本発明のペプチド) などが用いられる。 本発明のタンパク質画 分としては、 天然型の本発明のタンパク質画分か、 またはそれと同等の活性を有 する組換え型本発明のタンパク質画分などが望ましい。 ここで、 同等の活性とは、 同等のリガンド結合活性などを示す。 標識したペプチドとしては、 例えば放射性 同位元素 (例、 〔1251〕 、 〔1311〕 、 〔¾〕 、 〔"C〕 、 〔32P〕 、 〔33P〕 、 〔35s〕 な ど) 、 蛍光物質 〔例、 シァニン蛍光色素 (例、 Cy2、 Cy3、 Cy5、 Cy5. 5、 Cy7 (ァ マシャムバイオサイエンス社製) など) 、 フルォレスカミン、 フルォレツセンィ ソチオシァネートなど〕 、 酵素 (例、 —ガラクトシダ一ゼ、 j6—ダルコシダ一 ゼ、 アルカリフォスファタ一ゼ、 パ一ォキシダーゼ、 リンゴ酸脱水素酵素など) 、 発光物質 (例、 ルミノール、 ルミノール誘導体、 Jレシフェリン、 ルシゲニンな ど) 、 ピオチン、 ランタニド元素などで標識された本発明のペプチドなどを用い ることができる。 特に、 ポルトン—ハンター試薬を用いて公知の方法で調製した 本発明のペプチドの標識体を利用することもできる。 In order to carry out the above-described screening method for the receptor binding assay system or the cell stimulation assay system, for example, the protein fraction of the present invention and the peptide of the present invention (eg, the labeled peptide of the present invention) and the like are used. Used. The protein fraction of the present invention is preferably a natural protein fraction of the present invention or a recombinant protein fraction of the present invention having an activity equivalent thereto. Here, the equivalent activity means equivalent ligand binding activity and the like. The labeled peptide, such as a radioactive isotope (e.g., [125 1], [131 1], [¾], [ "C], [32 P], [33 P], etc. [35 s]), Fluorescent substances (eg, cyanine fluorescent dyes (eg, Cy2, Cy3, Cy5, Cy5.5, Cy7 (manufactured by Amersham Bioscience), etc.), fluorescamine, fluorescein sothiosinate, etc.), enzymes (eg, —galactosidase, j6—labeled with darcosidase, alkaline phosphatase, oxidase, malate dehydrogenase, etc., luminescent substances (eg, luminol, luminol derivatives, J-reciferin, lucigenin, etc.), piotin, lanthanide, etc. In particular, the peptide of the present invention prepared by a known method using a Porton-Hunter reagent can be used. It is also possible to use the body.
具体的には、 本発明のペプチドと本発明のタンパク質との結合性を変化させる 化合物のスクリーニングを行うには、 まず本発明のタンパク質を含有する細胞ま たは細胞の膜画分を、 スクリ一ニングに適したバッファーに懸濁することにより レセプター標品を調製する。 バッファ一には、 p H 4〜1 0 (望ましくは p H 6 〜8 ) のリン酸バッファー、 トリス—塩酸バッファ一などのリガンドとレセプタ —との結合を阻害しないバッファーであればいずれでもよい。 また、 非特異的結' 合を低減させる目的で、 CHAPS、 Tween-80™ (花王—アトラス社) 、 ジギトニン、 デォキシコレートなどの界面活性剤をバッファーに加えることもできる。 さらに、 プロテアーゼによる本発明のタンパク質や本発明のペプチドの分解を抑える目的 で P M S F、 ロイぺプチン、 E- 64 (ペプチド研究所製) 、 ぺプス夕チンなどのプ 口テア一ゼ阻害剤を添加することもできる。 0. 01〜10mlの該レセプ夕一溶液に、 一定量 (5000〜500000 cpm) の標識した本発明のペプチドを添加し、 同時に 10— 4 ~10— Mの試験化合物を共存させる。 非特異的結合量 (N S B ) を知るために大 過剰の未標識の本発明のぺプチドを加えた反応チューブも用意する。 反応は 0〜 50°C、 望ましくは 4〜37°Cで 20分〜 24時間、 望ましくは 30分〜 3時間行う。 反応後、 ガラス繊維濾紙等で濾過し、 適量の同バッファーで洗浄した後、 ガラス繊維濾紙 に残存する放射活性を液体シンチレ一ションカウンターまたはァ—カウンターで 計測する。 拮抗する物質がない場合のカウント (B。) から非特異的結合量 (N S B ) を引いたカウント (B。一 N S B ) を 1 0 0 %とした時、 特異的結合量 (B - N S B) が例えば 5 0 %以下になる試験化合物を拮抗阻害能力のある候補物質 として選択することができる。 Specifically, to screen for a compound that alters the binding between the peptide of the present invention and the protein of the present invention, first, cells containing the protein of the present invention or the membrane fraction of the cells are screened. Prepare the receptor standard by suspending it in a buffer suitable for tanning. Any buffer may be used as long as it does not inhibit the binding between the ligand and the receptor, such as a phosphate buffer having a pH of 4 to 10 (preferably, a pH of 6 to 8), such as Tris-HCl buffer. Also, for the purpose of reducing non-specific binding, a surfactant such as CHAPS, Tween-80 ™ (Kao-Atlas), digitonin, and dexcholate can be added to the buffer. In addition, a protease inhibitor such as PMSF, leptin, E-64 (manufactured by Peptide Research Laboratories), and peptide sutin is added to suppress the degradation of the protein of the present invention and the peptide of the present invention by protease. You can also. To the receptions evening first solution of 0. 01~10ml, added labeled peptide of the present invention a certain amount (5,000 to 500,000 cpm), coexist simultaneously 10-4 to 10-test compound M. Large to know the amount of non-specific binding (NSB) A reaction tube containing an excess of the unlabeled peptide of the present invention is also prepared. The reaction is carried out at 0 to 50 ° C, preferably 4 to 37 ° C, for 20 minutes to 24 hours, preferably 30 minutes to 3 hours. After the reaction, the reaction solution is filtered through a glass fiber filter or the like, washed with an appropriate amount of the same buffer, and the radioactivity remaining on the glass fiber filter is measured using a liquid scintillation counter or a counter. When the count (B. One NSB) obtained by subtracting the non-specific binding amount (NSB) from the count (B.) when there is no antagonistic substance is 100%, the specific binding amount (B-NSB) is For example, a test compound having 50% or less can be selected as a candidate substance having a competitive inhibitory ability.
また、 本発明のタンパク質と本発明のペプチドとの結合を測定する方法として、 BIAcore (アマシャムフアルマシアバイオテク社製) を用いることもできる。 こ の方法では、 本発明のペプチドを装置に添付のプロトコールに従ったァミノカツ プリング法によってセンサーチップに固定し、 本発明のタンパク質を含有する細 胞または本発明のタンパク質をコードする D NAを含有する形質変換体から精製 した本発明のタンパク質または本発明のタンパク質を含む膜画分、 あるいは精製 した本発明のタンパク質または本発明のタンパク質を含む膜画分および試験化合 物を含むリン酸バッファーまたはトリスバッファ一などの緩衝液をセンサーチッ プ上を毎分 2〜 20 ^ Iの流量で通過させる。 センサーチップ上の本発明のぺプチ ドと本発明のタンパク質とが結合することによって生じる表面プラズモン共鳴の 変化を共存する試験化合物が変化させることを観察することによって本発明のタ ンパク質と本発明のペプチドとの結合を変化させる化合物のスクリーニングを行 なうことができる。 この方法は、 本発明のタンパク質をセンサーチップに固定し、 本発明のぺプチドまたは本発明のぺプチドおよび試験化合物を含むリン酸バッフ ァ一またはトリスバッファ一などの緩衝液をセンサーチップ上を通過させる方法 を用いても同様に測定することができる。 試験化合物としては、 上記と同様のも のなどがあげられる。  Further, as a method for measuring the binding between the protein of the present invention and the peptide of the present invention, BIAcore (manufactured by Amersham Pharmacia Biotech) can be used. In this method, the peptide of the present invention is immobilized on a sensor chip by an amino coupling method according to the protocol attached to the device, and contains a cell containing the protein of the present invention or a DNA encoding the protein of the present invention. Phosphate buffer or Tris buffer containing the protein of the present invention purified from the transformant or the membrane fraction containing the protein of the present invention, or the purified protein of the present invention or the membrane fraction containing the protein of the present invention, and the test compound Pass one or more buffers over the sensor chip at a rate of 2-20 ^ I per minute. By observing that the coexisting test compound changes the surface plasmon resonance caused by the binding of the peptide of the present invention and the protein of the present invention on the sensor chip, the protein of the present invention and the present invention are changed. A compound that changes the binding to a peptide can be screened. In this method, the protein of the present invention is immobilized on a sensor chip, and a buffer solution such as a phosphate buffer or a Tris buffer containing the peptide of the present invention or the peptide of the present invention and a test compound is passed over the sensor chip. The same method can be used for measurement. Test compounds include those similar to the above.
前記の細胞刺激アツセィ系のスクリーニング方法を実施するためには、 本発明 のタンパク質を介する細胞刺激活性 (例えば、 ァラキドン酸遊離、 ァセチルコリ ン遊離、 細胞内 C a 2 +遊離、 細胞内 c AM P生成、 細胞内 c AM P産生抑制、 細 胞内 c GM P生成、 イノシ! ^一ルリン酸産生、 細胞膜電位変動、 細胞内タンパク 質のリン酸化、 c一 f o sの活性化、 p Hの低下、 c AM P依存性プロティンキ ナーゼの活性化、 c GM P依存性プロテインキナーゼの活性化、 リン脂質依存性 プロティンキナーゼの活性化、 微小管結合蛋白質リン酸化酵素 (MA Pキナー ゼ) の活性化などを促進する活性または抑制する活性など) を公知の方法または 市販の測定用キットを用いて測定することができる。 具体的には、 まず、 本発明 のタンパク質を含有する細胞をマルチウエルプレート等に培養する。 スクリー二 ングを行うにあたっては前もつて新鮮な培地あるいは細胞に毒性を示さない適当 なバッファ一に交換し、 試験化合物などを添加して一定時間インキュベートした 後、 細胞を抽出あるいは上清液を回収して、 生成した産物をそれぞれの方法に従 つて定量する。 細胞刺激活性の指標とする物質 (例えば、 ァラキドン酸など) の 生成が、 細胞が含有する分解酵素によって検定困難な場合は、 該分解酵素に対す る阻害剤を添加してアツセィを行なってもよい。 また、 c AM P産生抑制などの 活性については、 フオルスコリンなどで細胞の基礎的産生量を増大させておいた 細胞に対する産生抑制作用として検出することができる。 To perform the screening methods of the cell stimulating Atsusi system mediated cell stimulating activities proteins of the present invention (e.g., Arakidon acid release, Asechirukori emissions release, intracellular C a 2 + release, intracellular c AM P generated , Inhibition of intracellular cAMP production, Intracellular cGMP production, Wild boar! ^ -Monophosphate production, Cell membrane potential fluctuation, Intracellular protein Phosphorylation, activation of c-fos, decrease of pH, activation of cAMP-dependent protein kinase, activation of cGMP-dependent protein kinase, activation of phospholipid-dependent protein kinase, The activity of promoting or suppressing the activation of microtubule-associated protein kinase (MAP kinase), etc.) can be measured using a known method or a commercially available measurement kit. Specifically, first, cells containing the protein of the present invention are cultured on a multiwell plate or the like. Before performing screening, replace the medium with a fresh medium or an appropriate buffer that is not toxic to cells, add test compounds, etc., incubate for a certain period of time, and then extract cells or collect supernatant. Then, the generated product is quantified according to each method. When it is difficult to produce a substance (for example, arachidonic acid) as an indicator of cell stimulating activity using a degrading enzyme contained in a cell, an inhibitor for the degrading enzyme may be added to perform the assay. . In addition, activities such as inhibition of cAMP production can be detected as an activity of inhibiting production of cells whose basic production has been increased by forskolin or the like.
細胞刺激活性を測定してスクリーニングを行なうには、 適当な本発明のタンパ ク質を発現した細胞が用いられる。 本発明のタンパク質を発現した細胞としては、 前述の組換え型本発明のタンパク質発現細胞株などが望ましい。 形質転換体であ る本発明のタンパク質発現細胞は安定発現株でも一過性発現株でも構わない。 ま た、 動物細胞の種類は上記と同様のものが用いられる。  For screening by measuring the cell stimulating activity, cells expressing an appropriate protein of the present invention are used. As the cells expressing the protein of the present invention, the above-mentioned recombinant cell lines expressing the protein of the present invention and the like are desirable. The transformant expressing the protein of the present invention may be a stable expression strain or a transient expression strain. In addition, the same kind of animal cells as described above are used.
試験化合物としては、 例えばペプチド、 タンパク、 非ペプチド性化合物、 合成 化合物、 発酵生産物、 細胞抽出液、 植物抽出液、 動物組織抽出液などがあげられ る。  Test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and the like.
上記細胞刺激アツセィ系のスクリーニング方法について、 さらに具体的に以下 ( 1 ) 〜 (1 2 ) に記載する。  The above-mentioned screening method for the cell-stimulating assay system is described more specifically in the following (1) to (12).
( 1 ) レセプター発現細胞がレセプターァゴニストによって刺激されると細胞 内の Gタンパク質が活性化されて G T Pが結合する。 この現象はレセプ夕一発現 細胞の膜画分においても観察される。 通常、 G T Pは加水分解されて GD Pへと 変化するが、 このとき反応液中に G T Pァ Sを添加しておくと、 G T P 7 Sは G T Pと同様に Gタンパクに結合するが、 加水分解されずに G夕ンパクを含む細胞 膜に結合した状態が維持される。 標識した GTPァ Sを用いると細胞膜に残存し た標識された GTPァ Sを測定することにより、 レセプターァゴニストのレセプ ター発現細胞刺激活性を測定することができる。 (1) When a receptor-expressing cell is stimulated by a receptor agonist, an intracellular G protein is activated and GTP is bound. This phenomenon is also observed in the membrane fraction of receptor-expressing cells. Normally, GTP is hydrolyzed to GDP, but if GTPaS is added to the reaction solution at this time, GTP7S binds to G protein like GTP, but is hydrolyzed. Without G cells The state of being bonded to the membrane is maintained. If labeled GTPaS is used, the activity of stimulating receptor agonist receptor-expressing cells can be measured by measuring the labeled GTPaS remaining on the cell membrane.
この反応を利用して、 本発明のペプチドの本発明のタンパク質発現細胞に対す る刺激活性を測定することにより、 本発明のぺプチドと本発明の夕ンパク質との 結合性を変化させる化合物をスクリ一二ングすることができる。  By utilizing this reaction, the stimulatory activity of the peptide of the present invention on the protein-expressing cell of the present invention is measured, whereby a compound that changes the binding property between the peptide of the present invention and the protein of the present invention is obtained. You can screen.
この方法は、 本発明のタンパク質を含む膜画分を用いて行う。 本測定法におい て本発明のタンパク質膜画分への GTPァ S結合促進活性を示す物質はァゴニス トである。  This method is performed using a membrane fraction containing the protein of the present invention. In this assay method, the substance exhibiting the activity of promoting the GTP-A binding to the protein membrane fraction of the present invention is agonist.
具体的には、 標識した GTP T Sの存在下、 本発明のペプチドを本発明のタン パク質細胞膜画分に接触させた場合と、 本発明のぺプチドおよび試験化合物を本 発明のタンパク質細胞膜画分に接触させた場合における、 本発明のタンパク質細 胞膜画分への GTP T S結合促進活性を測定し、 比較することにより、 本発明の ペプチドと本発明のタンパク質との結合性を変化させる化合物をスクリーニング する。  Specifically, the case where the peptide of the present invention is brought into contact with the protein cell membrane fraction of the present invention in the presence of labeled GTP TS, and the case where the peptide and test compound of the present invention are brought into contact with the protein cell membrane fraction of the present invention. By measuring and comparing the activity of promoting GTPTS binding to the protein cell membrane fraction of the present invention when it is brought into contact with, a compound that alters the binding property between the peptide of the present invention and the protein of the present invention is determined. Screen.
本方法において、 本発明のペプチドによる本発明のタンパク質細胞膜画分への GTP T S結合促進活性を抑制する活性を示す試験化合物を、 拮抗阻害能力のあ る候補物質として選択することができる。 In this method, the peptide GTP T S binding promoting activity test compound showing activity to suppress to proteins the cell membrane fraction of the present invention according to the present invention can be selected as Ah Ru candidates competitive inhibition.
一方、 試験化合物のみを本発明のタンパク質細胞膜画分に接触させ、 本発明の 夕ンパク質細胞膜画分への G T Pァ S結合促進活性を測定することにより、 ァゴ 二ストのスクリ一ニングを行なうこともできる。  On the other hand, screening of agonist is carried out by bringing only the test compound into contact with the protein cell membrane fraction of the present invention and measuring the GTP-AS binding promoting activity to the protein cell membrane fraction of the present invention. You can also.
スクリーニング法の一例についてより具体的に以下に述べる。  One example of the screening method is described below more specifically.
公知の方法に準じて調製した本発明のタンパク質を含む細胞膜画分を、 膜希釈 緩衝液 ( 50 mM T r i s、 5 mM M g C 12、 150 mM N a C 1、 1 M GDP、 0. 1 % B S A ; pH7. 4) で希釈する。 希釈率は、 レセプターの 発現量により異なる。 これを F a 1 c 0 n 2053に 0. 2mlずつ分注し、 本 発明のペプチドまたは本発明のペプチドおよび試験化合物を加え、 さらに終濃度 200 pMとなるように [35S] GTPrSを加える。 25 °Cで 1時間保温した 後、 氷冷した洗浄用緩衝液 (50 mM Tr i s, 5mM MgC 12, 150 mM NaC l , 0. 1 % B S A, 0. 05% CHAP S ; pH7. 4) 1 5mlを加えて、 ガラス繊維ろ紙 GFZFでろ過する。 65 、 30分保温して 乾燥後、 液体シンチレ一ションカウンターでろ紙上に残った膜画分に結合した The cell membrane fraction containing the protein of the present invention prepared according to known methods, membrane dilution buffer (50 mM T ris, 5 mM M g C 1 2, 150 mM N a C 1, 1 M GDP, 0. Dilute with 1% BSA; pH 7.4). The dilution ratio depends on the expression level of the receptor. This was dispensed by 0. 2 ml min F a 1 c 0 n 2053, a peptide or peptide and a test compound of the present invention of the present invention was added, further to a final concentration of 200 pM added [35 S] GTPrS. After incubation for 1 hour at 25 ° C, wash ice-cold buffer (50 mM Tr is, 5mM MgC 1 2, 150 mM NaCl, 0.1% BSA, 0.05% CHAP S; pH 7.4) Add 15 ml, and filter through glass fiber filter paper GFZF. After incubating for 65 and 30 minutes and drying, it was bound to the membrane fraction remaining on the filter paper with a liquid scintillation counter.
[35S] GTPrSの放射活性を測定する。 本発明のペプチドのみを加えた実験 区の放射活性を 100%、 本発明のぺプチドを加えなかった実験区の放射活性をThe radioactivity of [ 35 S] GTPrS is measured. The radioactivity of the experimental group to which only the peptide of the present invention was added was 100%, and the radioactivity of the experimental group to which the peptide of the present invention was not added was 100%.
0%とし、 本発明のペプチドによる GTPrs結合促進活性に対する試験化合物 の影響を算出する。 GTPァ S結合促進活性が例えば 50 %以下になる試験化合 物を拮抗阻害能力のある候補物質として選択することができる。 Assuming 0%, the effect of the test compound on the GTP rs binding promoting activity of the peptide of the present invention is calculated. A test compound having a GTPaS binding promoting activity of, for example, 50% or less can be selected as a candidate substance having competitive inhibitory ability.
(2) 本発明のタンパク質発現細胞は、 本発明のペプチドの刺激により、 細胞 内 c AMPの産生が抑制される。 この反応を利用して、 本発明のペプチドの本発 明の夕ンパク質発現細胞に対する刺激活性を測定することにより、 本発明のぺプ チドと本発明の夕ンパク質との結合性を変化させる化合物をスクリ一二ングする ことができる。  (2) In the cell expressing the protein of the present invention, the production of intracellular cAMP is suppressed by stimulation of the peptide of the present invention. By utilizing this reaction to measure the stimulating activity of the peptide of the present invention on the protein-expressing cells of the present invention, the binding between the peptide of the present invention and the protein of the present invention is changed. The compound can be screened.
具体的には、 細胞内 cAM P量を増加させる物質の存在下、 本発明のペプチド を本発明のタンパク質発現細胞に接触させた場合と、 本発明のペプチドおよび試 験化合物を本発明のタンパク質発現細胞に接触させた場合における、 該細胞の細 胞内 c AM Pの産生抑制活性を測定し、 比較することにより、 本発明のぺプチ と本発明のタンパク質との結合性を変化させる化合物をスクリーニングする。  Specifically, the case where the peptide of the present invention is brought into contact with the protein-expressing cell of the present invention in the presence of a substance that increases the amount of intracellular cAMP, the case where the peptide of the present invention and the test compound express the protein of the present invention, A compound that alters the binding between the peptide of the present invention and the protein of the present invention is determined by measuring the activity of inhibiting the production of intracellular cAMP in the cell when the cell is brought into contact with the cell and comparing the results. I do.
細胞内 c AMP量を増加させる物質としては、 例えば、 フオルスコリン、 カル シトニンなどが用いられる。  As a substance that increases the amount of intracellular cAMP, for example, forskolin, calcitonin and the like are used.
本発明のタンパク質発現細胞内の c AMP産生量は、 マウス、 ラット、 ゥサギ、 ャギ、 ゥシなどを免疫して得られた抗 c AMP抗体と 〔1251〕 標識 c AMP (ともに市販品) を使用することによる R I A系、 または抗 cAMP抗体と標識 c AMPとを組み合わせた E I A系で測定することができる。 また、 抗 cAMP 抗体を、 p r o t e i n Aまたは抗 c AMP抗体産生に用いた動物の I gGな どに対する抗体などを使用して固定したシンチラントを含むビーズと 〔1251〕 標 識 c AMPとを使用する S PA (Scintillation Proximity Assay) 法による定 量も可能である (アマシャムフアルマシアバイオテク社製のキットを使用する) 。 本方法において、 本発明のペプチドによる本発明のタンパク質発現細胞の c A MP産生抑制活性を阻害する活性を示す試験化合物を、 拮抗阻害能力のある候補 物質として選択することができる。 The amount of cAMP produced in the protein-expressing cells of the present invention is determined by comparing the amount of anti-cAMP antibody obtained by immunizing mice, rats, rabbits, goats, rabbits, etc. with [ 125 1] -labeled cAMP (both commercially available products). Can be measured using an RIA system by using EIA or an EIA system combining an anti-cAMP antibody and labeled cAMP. Moreover, the anti-cAMP antibody, using a protein A or beads containing scintillant which is secured using such antibodies to etc. I gG of animals used in the anti-c AMP antibody production [125 1]-labeled c AMP Quantification by the SPA (Scintillation Proximity Assay) method is also possible (using a kit manufactured by Amersham Pharmacia Biotech). In the method, c A of the protein-expressing cell of the present invention by the peptide of the present invention A test compound exhibiting an activity of inhibiting MP production inhibitory activity can be selected as a candidate substance having a competitive inhibition ability.
一方、 試験化合物のみを本発明のタンパク質発現細胞に接触させて、 cAMP 産生抑制活性を調べることによりァゴニスト活性を示す化合物のスクリーニング を行なうことができる。  On the other hand, a compound showing an agonist activity can be screened by contacting only the test compound with the protein-expressing cell of the present invention and examining the cAMP production inhibitory activity.
スクリーニング法の一具体例を以下に述べる。  One specific example of the screening method is described below.
本発明のタンパク質発現細胞 (例、 CHO細胞などの動物細胞) .を 24穴プレ 一卜に 5 X 104c e 1 lZwe 1 1で播種し、 48時間培養する。 細胞を 0. 2 ηιΜ 3一イソブチルーメチルキサンチン、 0. 05% B S Aおよび 2 OmM HEP ESを含むハンクスバッファ一 (pH7. 4) で洗浄する (以下、 反応用 バッファ一と略記する) 。 その後、 0. 5m 1の反応用バッファーを加えて 30 分間培養器で保温する。 反応用バッファーを除き、 新たに 0. 25mlの反応用 バッファーを細胞に加えた後、 1 Mの本発明のペプチドまたは 1 Mの本発明 のべプチドおよび試験化合物を添加した 2 M フオルスコリンを含む 0. 25 m 1の反応用バッファーを、 細胞に加え、 37 °Cで 24分間反応させる。 100 1の 20%過塩素酸を加えて反応を停止させ、 その後氷上で 1時間置くことに より細胞内 c AMPを抽出する。 抽出液中の c AMP量を、 cAMP E I Aキ ット (アマシャムフアルマシアバイォテク) を用いて測定する。 フオルスコリン の刺激によって産生された CAM P量を 100%とし、 1 / Mの本発明のぺプチ ドの添加によって抑制された c AMP量を 0 %として、 本発明のペプチドによる c AMP産生抑制活性に対する試験化合物の影響を算出する。 本発明のぺプチド の活性を阻害して、 c AMP産生活性が例えば 50 %以上になる試験化合物を、 拮抗阻害能力のある候補物質として選択することができる。 Cells expressing the protein of the present invention (eg, animal cells such as CHO cells) are seeded on a 24-well plate at 5 × 10 4 ce 1 LZwe 11 and cultured for 48 hours. Wash the cells with Hank's buffer (pH 7.4) containing 0.2 ηιΜ3-isobutyl-methylxanthine, 0.05% BSA and 2 OmM HEPES (hereinafter abbreviated as reaction buffer 1). Then add 0.5 ml of reaction buffer and incubate in the incubator for 30 minutes. Excluding the reaction buffer, add 0.25 ml of the reaction buffer to the cells, and then add 1 M of the peptide of the present invention or 1 M of the peptide of the present invention and 2 M forskolin to which the test compound is added. Add 0.25 ml of reaction buffer to the cells and incubate at 37 ° C for 24 minutes. Stop the reaction by adding 100 1 of 20% perchloric acid, and then leave it on ice for 1 hour to extract intracellular cAMP. The amount of cAMP in the extract is measured using a cAMP EIA kit (Amersham Pharmacia Biotech). The amount of cAMP produced by the stimulation of forskolin was defined as 100%, and the amount of cAMP suppressed by the addition of 1 / M of the peptide of the present invention was defined as 0%. The effect of the test compound on is calculated. A test compound that inhibits the activity of the peptide of the present invention and has a cAMP production activity of, for example, 50% or more can be selected as a candidate substance capable of competitive inhibition.
また、 本発明のペプチドの刺激により、 細胞内 CAM P量が増加する性質を示 す本発明のタンパク質発現細胞を使用する場合、 本発明のペプチドを本発明の夕 ンパク質発現細胞に接触させた場合と、 本発明のぺプチドおよび試験化合物を本 発明のタンパク質発現細胞に接触させた場合における、 該細胞の細胞内 cAMP の産生促進活性を測定し、 比較することにより、 本発明のペプチドと本発明の夕 ンパク質との結合性を変化させる化合物をスクリーニングすることができる。 本方法において、 本発明のぺプチドによる本発明の夕ンパク質発現細胞の c A M P産生促進活性を阻害する活性を示す試験化合物を、 拮抗阻害能力のある候補 物質として選択することができる。 ' ' In addition, when using the protein-expressing cell of the present invention, which exhibits the property of increasing the amount of intracellular CAMP by stimulation of the peptide of the present invention, the peptide of the present invention was brought into contact with the protein-expressing cell of the present invention. And a test compound obtained by contacting the peptide of the present invention and the test compound with the protein-expressing cell of the present invention, by measuring and comparing the intracellular cAMP production promoting activity of the cell. Compounds that alter the binding to the proteins of the invention can be screened. In this method, a test compound showing an activity of inhibiting the cAMP production promoting activity of the protein-expressing cells of the present invention by the peptide of the present invention can be selected as a candidate substance having an antagonistic inhibitory ability. ''
一方、 試験化合物のみを本発明のタンパク質発現細胞に接触させて c AMP産 生促進活性を調べることによりァゴニスト活性を示す化合物のスクリ一ニングを 行なうことができる。  On the other hand, a compound exhibiting agonist activity can be screened by contacting the test compound alone with the protein-expressing cell of the present invention and examining cAMP production promoting activity.
c AMP産生促進活性は、 上記のスクリ一ニング法においてフォルスコリンを 添加せずに本発明のタンパク質発現細胞 (例、 CHO細胞などの動物細胞) に本 発明のペプチドまたは本発明のペプチドおよび試験化合物を添加して産生された c AMPを上記の方法で定量して測定する。  c The AMP production promoting activity can be measured by adding the peptide of the present invention or the peptide of the present invention and the test compound to the protein-expressing cells of the present invention (eg, animal cells such as CHO cells) without adding forskolin in the above screening method. The amount of cAMP produced by the addition of is determined and measured by the method described above.
(3) CRE—レポ一ター遺伝子ベクターを用いて、 本発明のペプチドの本発 明のタンパク質発現細胞に対する刺激活性を測定することにより、 本発明のぺプ チドと本発明の夕ンパク質との結合性を変化させる化合物をスクリ一ニングする ことができる。  (3) By measuring the stimulating activity of the peptide of the present invention on cells expressing the protein of the present invention using the CRE-reporter gene vector, the peptide of the present invention and the protein of the present invention can be measured. Compounds that alter binding can be screened.
CRE (cAMP response element) を含む DNAを、 ベクタ一のレポーター遺 伝子上流に挿入し、 CRE—レポ一夕一遺伝子べクタ一を得る。 CRE—レポ一 ター遺伝子ベクターを導入した本発明のタンパク質発現細胞において、 c AMP の上昇を伴う刺激は、 CREを介したレポーター遺伝子発現と、 それに引き続く レポ一夕一遺伝子の遺伝子産物 (タンパク質) の産生を誘導する。 つまり、 レポ —タ一遺伝子タンパク質の酵素活性を測定することにより、 CRE—レポ一夕一 遺伝子ベクター導入細胞内の CAM P量の変動を検出することができる。  DNA containing CRE (cAMP response element) is inserted into the vector upstream of the reporter gene to obtain the CRE—repo overnight gene vector. In the protein-expressing cells of the present invention into which the CRE-reporter gene vector has been introduced, stimulation accompanied by an increase in cAMP is accompanied by CRE-mediated expression of the reporter gene, and subsequent stimulation of the gene product (protein) of the repo overnight gene. Induce production. In other words, by measuring the enzymatic activity of the reporter gene protein, it is possible to detect a change in the amount of CAMP in cells into which the CRE-repo overnight gene vector has been introduced.
具体的には、 細胞内 c AMP量を増加させる物質の存在下、 本発明のペプチド を、 C R E—レポ一夕一遺伝子ベクター導入本発明のタンパク質発現細胞に接触 させた場合と、 本発明のペプチドおよび試験化合物を、 CRE—レポ一夕一遺伝 子べクタ一導入本発明のタンパク ¾発現細胞に接触させた場合における、 レポ一 ター遺伝子タンパク質の酵素活性を測定し、 比較することにより、 本発明のぺプ チドと本発明のタンパク質との結合性を変化させる化合物をスクリーニングする- 細胞内 CAM P量を増加させる物質としては、 例えば、 フオルスコリン、 カル シトニンなどが用いられる。 ベクターとしては、 例えば、 ピツカジーン べィシックベクター、 ピツカジー ン ェンハンサーベクター (東洋インキ製造 (株) ) などが用いられる。 CRE を含む DNAを、 上記べクタ一のレポ一夕一遺伝子、 例えばルシフェラ一ゼ遺伝 子上流のマルチクローニングサイトに挿入し、 CRE—レポーター遺伝子べクタ 一とする。 Specifically, when the peptide of the present invention is brought into contact with the protein-expressing cell of the present invention into which the peptide of the present invention has been introduced in the presence of a substance that increases the amount of intracellular cAMP, the peptide of the present invention And the test compound was brought into contact with the protein-expressing cell of the present invention when the CRE-repo overnight gene vector was introduced. The enzyme activity of the reporter gene protein was measured and compared. Screening for a compound that alters the binding between the peptide of the present invention and the protein of the present invention. As a substance that increases the amount of intracellular CAMP, for example, forskolin, calcitonin and the like are used. As the vector, for example, Pitka Gene Basic Vector, Pitka Gene Enhancer Vector (Toyo Ink Manufacturing Co., Ltd.) and the like are used. The CRE-containing DNA is inserted into the above-mentioned vector in the repo overnight gene, for example, the multicloning site upstream of the luciferase gene, to obtain the CRE-reporter gene vector.
本方法において、 本発明のぺプチドによるレポ一ター遺伝子タンパク質の酵素 活性抑制を回復させる試験化合物を、 拮抗阻害能力のある候補物質として選択す ることができる。  In this method, a test compound that restores the inhibition of the enzyme activity of the reporter gene protein by the peptide of the present invention can be selected as a candidate substance capable of competitive inhibition.
一方、 試験化合物のみを本発明のタンパク質発現細胞に接触させて、 フォルス コリン刺激によって上昇した発光量の本発明のぺプチドと同様な抑制を測定する ことによりァゴニストのスクリーニングを行なうこともできる。  On the other hand, agonists can be screened by bringing only the test compound into contact with the protein-expressing cells of the present invention and measuring the same inhibition of the amount of luminescence increased by forskolin stimulation as with the peptides of the present invention.
レポ一ター遺伝子として、 ルシフェラーゼを利用する例を用いて、 このスクリ 一二ング方法の具体例を以下に述べる。  A specific example of this screening method will be described below using an example using luciferase as a reporter gene.
CRE—レポ一夕一遺伝子 (ルシフェラ一ゼ) を導入した本発明のタンパク質 発現細胞を、 24穴プレートに 5 x 103c e 1 lZwe 1 1で播種し、 48時 間培養する。 細胞を 0. 2mM 3 _イソプチルーメチルキサンチン、 0. 0 5 % BSAおよび 20mM HE P E Sを含むハンクスバッファー ( p H 7. 4) で洗浄する (以下、 反応用バッファーと略記する) 。 その後 0. 5m lの反 応用バッファーを加えて 30分間培養器で保温する。 反応用バッファーを除き、 新たに 0. 25m 1の反応用バッファ一を細胞に加えた後、 ΙμΜの本発明のぺ プチドまたは 1 μΜの本発明のぺプチドおよび試験化合物を添加した 2 μΜ フォ ルスコリンを含む 0. 25m 1の反応用バッファーを、 細胞に加え、 37口で 2 4分間反応させる。 細胞をピツカジーン用細胞溶解剤 (東洋インキ製造 (株) ) で溶かし、 溶解液に発光基質 (東洋インキ製造 (株) ) を添加する。 ルシフェラ ーゼによる発光は、 ルミノメーター、 液体シンチレーシヨンカウンターまたはト ップカウン夕一により測定する。 本発明のペプチド単独を添加した場合と、 1 Mの本発明のぺプチドおよび試験化合物を添加した場合のルシフェラーゼによる 発光量を測定して、 比較する。 Cells expressing the protein of the present invention into which the CRE-repo overnight gene (luciferase) has been introduced are seeded on a 24-well plate at 5 × 10 3 ce 1 lZwe 11 and cultured for 48 hours. The cells are washed with Hank's buffer (pH 7.4) containing 0.2 mM 3_isobutyl-methylxanthine, 0.05% BSA and 20 mM HE PES (hereinafter abbreviated as reaction buffer). Then add 0.5 ml of application buffer and incubate for 30 minutes in the incubator. After removing the reaction buffer and adding 0.25 ml of a new reaction buffer to the cells, add 2 μl of forskolin containing 1 μl of the peptide of the present invention or 1 μl of the peptide of the present invention and a test compound. Add 0.25 ml of reaction buffer containing to the cells and incubate for 24 minutes with 37 tubes. Lyse the cells with a cell lysing agent for Pitka Gene (Toyo Ink Mfg. Co., Ltd.) and add a luminescent substrate (Toyo Ink Mfg. Co., Ltd.) to the lysate. Luminescence from luciferase is measured using a luminometer, liquid scintillation counter, or top counter. The amount of luminescence by luciferase when the peptide of the present invention alone is added and when 1 M of the peptide of the present invention and a test compound are added are measured and compared.
本発明のペプチドは、 フオルスコリン刺激に基づくルシフェラ一ゼによる発光 量の増加を抑制する。 該抑制を回復させる化合物を拮抗阻害能力のある候補物質 として選択することができる。 The peptides of the present invention provide for luciferase-induced luminescence upon forskolin stimulation. Suppress increase in volume. A compound that restores the inhibition can be selected as a candidate substance having competitive inhibitory ability.
レポーター遺伝子として、 例えば、 アルカリフォスファタ一ゼ、 クロラムフエ ニコ一レ ·ァセチリレトランスフェラ一セ ch l orampheni co l  As reporter genes, for example, alkaline phosphatase, chloramphene
acetyl t rans f erase) 、 iS—ガラクトシダ一ゼなどの遺伝子を用いてもよい。 こ れらのレポ一ター遺伝子タンパク質の酵素活性は、 公知の方法に従い、 または市 販の測定キットを用いて測定する。 アルカリフォスファタ一ゼ活性は、 例えば和 光純薬製 Lumi-Phos 530を用いて、 クロラムフエ二コール 'ァセチルトランスフ エラ一ゼ活性は、 例えば和光純薬製 FAST CAT chrol amphenicol Acetyl trans f erase) or a gene such as iS-galactosidase may be used. The enzyme activity of these reporter gene proteins is measured according to a known method or using a commercially available measurement kit. Alkaline phosphatase activity can be measured using, for example, Lumi-Phos 530 manufactured by Wako Pure Chemical, and chloramphenicol acetyltransferase can be measured using, for example, FAST CAT chrol amphenicol manufactured by Wako Pure Chemical.
Ace tyl t rans f erase Assay KiTを用いて、 i3 _ガラクトシダーゼ活性は、 例えば 和光純薬製 Au rora Ga 1 - XEを用いて測定する。 The i3-galactosidase activity is measured using, for example, Acetyl trans erase erase Assay KiT using, for example, Aurora Ga1-XE manufactured by Wako Pure Chemical Industries.
( 4 ) 本発明のタンパク質発現細胞は、 本発明のペプチドの刺激により、 ァラ キドン酸代謝物を細胞外に放出する。 この反応を利用して、 本発明のペプチドの 本発明のタンパク質発現細胞に対する刺激活性を測定することにより、 本発明の ペプチドと本発明のタンパク質との結合性を変化させる化合物をスクリーニング することができる。  (4) The protein-expressing cells of the present invention release arachidonic acid metabolites extracellularly by stimulation of the peptides of the present invention. By utilizing this reaction to measure the stimulating activity of the peptide of the present invention on the cells expressing the protein of the present invention, it is possible to screen for a compound that alters the binding property between the peptide of the present invention and the protein of the present invention. .
あらかじめ、 標識したァラキドン酸を、 本発明のタンパク質発現細胞に取り込 ませておくことによって、 ァラキドン酸代謝物放出活性を、 細胞外に放出された 標識されたァラキドン酸代謝物を測定することによって測定することができる。 具体的には、 本発明のペプチドを、 標識したァラキドン酸を含有する本発明の タンパク質発現細胞に接触させた場合と、 本発明のペプチドおよび試験化合物を、 標識したァラキドン酸を含有する本発明のタンパク質発現細胞に接触させた場合 における、 ァラキドン酸代謝物の放出活性を測定し、 比較することにより、 本発 明のペプチドと本発明のタンパク質との結合性を変化させる化合物をスクリ一二 ングする。  The arachidonic acid metabolite-releasing activity can be measured by measuring the labeled arachidonic acid metabolite released outside the cell by incorporating the labeled arachidonic acid into the cells expressing the protein of the present invention in advance. can do. Specifically, when the peptide of the present invention is brought into contact with a cell expressing the protein of the present invention containing labeled arachidonic acid, the peptide of the present invention and the test compound are brought into contact with the cell of the present invention containing labeled arachidonic acid. By measuring and comparing the arachidonic acid metabolite release activity when brought into contact with a protein-expressing cell, a compound that changes the binding property between the peptide of the present invention and the protein of the present invention is screened. .
本方法において、 本発明のペプチドによるァラキドン酸代謝物放出活性を阻害 する試験化合物を、 拮抗阻害能力のある候補物質として選択することができる。 また、 試験化合物のみを本発明のタンパク質発現細胞に接触させ、 本発明の夕 ンパク質発現細胞のァラキドン酸代謝物放出活性を公知の方法で調べることによ りァゴニスト活性を示す化合物のスクリーニングを行なうこともできる。 In this method, a test compound that inhibits the arachidonic acid metabolite release activity of the peptide of the present invention can be selected as a candidate substance having a competitive inhibition ability. Alternatively, the test compound alone is brought into contact with the protein-expressing cell of the present invention, and the arachidonic acid metabolite releasing activity of the protein-expressing cell of the present invention is examined by a known method. A compound exhibiting agonist activity can also be screened.
スクリーニング法の一具体例を以下に述べる。  One specific example of the screening method is described below.
本発明のタンパク質発現細胞を 24穴プレートに 5 X 104c e 1 1 /we 1 1 で播種し、 24時間培養後、 [3H] ァラキドン酸を 0. 25 C i/we l lと なるよう添加し、 16時間後、 細胞を 0. 05% BSAぉょび20mM HE PESを含むハンクスバッファ一 (pH7. 4) (以下、 反応用バッファ一と略 記する) で洗浄する。 終濃度 10 Mの本発明のペプチドまたは終濃度 10 /M の丰発明のぺプチドおよび試験化合物を含む反応用バッファー 500 ^ 1を、 各 we 1 1に添加する。 37°Cで 60分間インキュベートした後、 反応液 400 1をシンチレ一夕一に加え、 反応液中に遊離した [3H] ァラキドン酸代謝物 + の量をシンチレーションカウン夕一により測定する。 The protein-expressing cells of the present invention are seeded on a 24-well plate at 5 × 10 4 ce 11 / we 11, and cultured for 24 hours, and [ 3 H] arachidonic acid is added to 0.25 Ci / well. After 16 hours, wash the cells with Hanks buffer (pH 7.4) containing 0.05% BSA and 20 mM HE PES (hereinafter abbreviated as reaction buffer 1). A reaction buffer 500 ^ 1 containing a final concentration of 10 M of the peptide of the present invention or a final concentration of 10 / M of the peptide of the present invention and a test compound is added to each we11. After incubating at 37 ° C for 60 minutes, the reaction solution 4001 is added to the scintillator overnight, and the amount of [ 3 H] arachidonic acid metabolite + released in the reaction solution is measured by scintillation counting.
反応用バッファー 500 ^ 1のみを添加した場合 (本発明のペプチド非添 加 ·試験化合物非添加) の遊離 [3H] ァラキドン酸代謝物の量を 0%、 1 O n Mの本発明のぺプチドを含む反応用バッファーを添加した場合 (試験化合物非添 加) の遊離 [3H] ァラキドン酸代謝物の量を 100%として、 試験化合物を添 '加した場合の遊離 [3H] ァラキドン酸代謝物の量を算出する。 When only 500 ^ 1 of the reaction buffer was added (without adding the peptide of the present invention and without adding the test compound), the amount of free [ 3 H] arachidonic acid metabolite was reduced to 0%, and 1 OnM of the present invention was added. If the addition of reaction buffer containing peptide free [3 H] Arakidon the amount of acid metabolites as 100%, free [3 H] Arakidon acid when the test compound added 'to addition of (test compound non-added additive) Calculate the amount of metabolite.
ァラキドン酸代謝物放出活性が、 例えば 50%以下になる試験化合物を拮抗阻 害能力のある候補物質として選択することができる。  A test compound having an arachidonic acid metabolite releasing activity of, for example, 50% or less can be selected as a candidate substance having an antagonistic ability.
(5) 本発明のタンパク質発現細胞は、 本発明のペプチドの刺激により、 細胞 内の C a濃度が上昇する。 この反応を利用して、 本発明のペプチドの本発明の夕 ンパク質発現細胞に対する刺激活性を測定することにより、 本発明のぺプチドと 本発明のタンパク質との結合性を変化させる化合物をスクリーニングすることが できる。 '  (5) In the cell expressing the protein of the present invention, the intracellular Ca concentration increases by stimulation with the peptide of the present invention. By utilizing this reaction, the stimulating activity of the peptide of the present invention on the protein-expressing cell of the present invention is measured to screen for a compound that changes the binding property between the peptide of the present invention and the protein of the present invention. be able to. '
具体的には、 本発明のペプチドを、 本発明のタンパク質発現細胞に接触させた 場合と、 本発明のペプチドおよび試験化合物を、 本発明のタンパク質発現細胞に 接触させた場合における、 細胞内カルシウム濃度上昇活性を測定し、 比較するこ とにより、 本発明のペプチドと本発明のタンパク質との結合性を変化させる化合 物をスクリーニングする。 測定は公知の方法に従つて行う。  Specifically, the intracellular calcium concentration in the case where the peptide of the present invention is brought into contact with the protein-expressing cell of the present invention and in the case where the peptide of the present invention and the test compound are brought into contact with the protein-expressing cell of the present invention By measuring and comparing the increasing activity, a compound that changes the binding property between the peptide of the present invention and the protein of the present invention is screened. The measurement is performed according to a known method.
本方法において、 本発明のペプチドによる細胞内カルシウム濃度の上昇を抑制 する試験化合物を、 拮抗阻害能力のある候補物質として選択することができる。 一方、 試験化合物のみの添加による蛍光強度の上昇を測定することによってァ ゴニス卜のスクリーニングを行なうこともできる。 In the method, an increase in intracellular calcium concentration by the peptide of the present invention is suppressed. The test compound to be tested can be selected as a candidate substance capable of competitive inhibition. On the other hand, an agonist can be screened by measuring an increase in fluorescence intensity due to the addition of the test compound alone.
'スクリーニング法の一具体例を以下に述べる。  'A specific example of the screening method is described below.
本発明のタンパク質発現細胞を、 滅菌した顕微鏡用カバーグラス上に播き、 2 日後、 培養液を、 4 mM F u r a— 2 AM (同仁化学研究所) を縣濁した H B S Sに置換し、 室温で 2時間 3 0分おく。 H B S Sで洗浄した後、 キュベットに カバーグラスをセットし、 本発明のペプチドまたは本発明のペプチドおよび試験 化合物を添加し、 励起波長 3 4 0 n mおよび 3 8 0 n mでの、 5 0 5 nmの蛍光 強度の比の上昇を蛍光測定器で測定し、 比較する。  The protein-expressing cells of the present invention are seeded on a sterilized cover glass for a microscope. Two days later, the culture solution is replaced with HBSS suspended in 4 mM Fura-2AM (Dojindo Laboratories), and incubated at room temperature. Leave for 30 minutes. After washing with HBSS, place a coverslip on the cuvette, add the peptide of the present invention or the peptide of the present invention and a test compound, and emit fluorescence at 505 nm at excitation wavelengths of 300 nm and 380 nm. Measure the increase in intensity ratio with a fluorimeter and compare.
また、 F L I P R (モレキュラーデバイス社製) を使って行ってもよい。 本発 明のタンパク質発現細胞縣濁液に F 1 u o— 3 AM (同仁化学研究所製) を添 加し、 細胞に取り込ませた後、 上清を遠心により数度洗浄後、 9 6穴プレートに 細胞を播く。 ' F L I P R装置にセットし、 F u r a _ 2の場合と同様に、 本発明 のペプチドまたは本発明のペプチドおよび試験化合物を添加し、 蛍光強度の比の 上昇を蛍光測定器で測定し、 比較する。  Alternatively, FLIPR (manufactured by Molecular Devices) may be used. Add F1uo-3AM (manufactured by Dojindo Laboratories) to the protein-expressing cell suspension of the present invention, allow the cells to take up, and wash the supernatant several times by centrifugation. Seed the cells. 'Set in a FLIPR apparatus, add the peptide of the present invention or the peptide of the present invention and a test compound in the same manner as in the case of Fura_2, and measure the increase in the ratio of the fluorescence intensities using a fluorimeter and compare.
さらに、 本発明のタンパク質発現細胞に、 細胞内 C aイオンの上昇によって発 光するようなタンパク質の遺伝子 (例、 aequorinなど) を共発現させておき、 細 胞内 C aイオン濃度の上昇によって、 該遺伝子タンパク質 (例、 aequor inなど) が C a結合型となり発光することを利用して、 本発明のペプチドと本発明のタン パク質との結合性を変化させる化合物をスクリーニングすることもできる。 細胞内 C aイオンの上昇によつて発光するようなタンパク質の遺伝子を共発現 させ 本発明のタンパク質発現細胞を、 9 6穴プレートに播き、 上記と同様に、 本発明のぺプチドまたは本発明のぺプチドおよび試験化合物を添加し、 蛍光強度 の比の上昇を蛍光測定器で測定し、 比較する。  Furthermore, a protein gene (eg, aequorin, etc.) that emits light due to an increase in intracellular Ca ion is co-expressed in the protein-expressing cell of the present invention, and the intracellular Ca ion concentration is increased. By utilizing the fact that the gene protein (eg, aequorin or the like) becomes Ca-bound and emits light, it is also possible to screen for a compound that changes the binding property between the peptide of the present invention and the protein of the present invention. A gene for a protein that emits light when the intracellular Ca ion increases is co-expressed, and the protein-expressing cell of the present invention is seeded on a 96-well plate, and the peptide of the present invention or the Add the peptide and test compound, measure the increase in the ratio of fluorescence intensity with a fluorimeter, and compare.
本発明のぺプチドによる蛍光強度の上昇を、 抑制する試験化合物を拮抗阻害能 力のある候補物質として選択することができる。  A test compound that suppresses an increase in fluorescence intensity due to the peptide of the present invention can be selected as a candidate substance having a competitive inhibition ability.
スクリ一ニング法の一具体例を以下に述べる。  One specific example of the screening method is described below.
対照として ETA (エンドセリン Aレセプター) 発現 CH0細胞 24番クローン (以後 ETA24細胞と略称する。 Journal of Pharmacology and Experimental As a control, clone 24 of ETA (endothelin A receptor) -expressing CH0 cell Abbreviated as ETA24 cells. Journal of Pharmacology and Experimental
Therapeutics, 279卷、 675- 685頁、 1996年参照) を用い、 アツセィ用サンプルに ついて、 本発明のタンパク質発現細胞および ETA24細胞における細胞内 Caイオン 濃度上昇活性の測定を FLIPR (モレキュラーデバイス社製) を用いて行う。 本発 明のタンパク質発現細胞細胞、 ETA24細胞共に 10%透析処理済ゥシ胎児血清 (以 後 dFBSとする) を加えた DMEMで継代培養しているものを用いる。 本発明のタンパ ク質発現細胞、 ETA24細胞をそれぞれ 15xl04cells/mlとなるように培地 (10% d FBS-DMEM) に懸濁し、 FLIPR用 96穴プレート(Black plate clear bottom. Coster 社)に分注器を用いて各ゥエルに 200 1ずつ植え込み (3.0xl04cells/200 /l/ゥェ ル) 、 5% C02インキュベータ一中にて 37Cで一晩培養した後用いる (以後、 細胞 プレー卜とする) 。 H/HBSS (二ッスィハンクス 2 (日水製薬株式会社) 9.8g、 炭酸水素ナトリウム 0.35g、 HEPES 4.77 g、 水酸化ナトリウム溶液で pH7.4に 合わせた後、 フィルター滅菌処理) 20 ml、 250 mM Probenecid 200 ゥシ胎 児血清 (FBS) 200 を混合する。 また、 Fluo 3-AM (同仁化学研究所) 2パイァ ル(50 xg)をジメチルスルフオキサイド 40 1、 20% Pluronic acid Therapeutics, Vol. 279, pp. 675-685, 1996) was used to measure the activity of increasing the intracellular Ca ion concentration of the protein-expressing cells of the present invention and ETA24 cells using FLIPR (manufactured by Molecular Devices). This is performed using The cells expressing the protein of the present invention and ETA24 cells should be subcultured in DMEM supplemented with 10% dialyzed fetal calf serum (hereinafter referred to as dFBS). Protein expressing cells of the present invention, suspended in medium (10% d FBS-DMEM) such that ETA24 cells each 15xl0 4 cells / ml, in 96 well plate for FLIPR (Black plate clear bottom. Coster, Inc.) dispenser by 200 1 each Ueru with implantation (3.0xl0 4 cells / 200 / l / © E Le), used after overnight incubation at 37C in 5% C0 2 incubator in one (hereinafter, cells play ). H / HBSS (Nissy Hanks 2 (Nissui Pharmaceutical Co., Ltd.) 9.8 g, sodium hydrogen carbonate 0.35 g, HEPES 4.77 g, adjusted to pH 7.4 with sodium hydroxide solution, filter sterilized) 20 ml, 250 mM Probenecid 200 ゥ Mix 200 fetal serum (FBS). In addition, Fluo 3-AM (Dojindo Research Laboratories) 2 piles (50 xg) was converted to dimethyl sulfoxide 401, 20% Pluronic acid
(Molecular Probes社) 40 に溶解し、 これを上記 H/HBSS—Probenecid— FBS に加え、 混和後、 8連ピペットを用いて培養液を除いた細胞プレートに各ゥエル 100 lずつ分注し、 5% C02インキュべ一ター中にて 37 °Cで 1時間インキュベー トする (色素ローデイング) 。 アツセィ用サンプル (各フラクション) に、 2.5 mM Probenecid, 0.1 % CHAPSを含む H/HBSS 150 1を加えて希釈し、 FLIPR用 96穴 プレート (V- Bottomプレート、 Coster社) へ移す (以後、 サンプルプレートとす る) 。 細胞プレートの色素ローデイング終了後、 ¾ 33に2.5 mM Probenecidを 加えた洗浄バッファーでプレートウォッシャー(Molecular Devices社)を用いて 細胞プレートを 4回洗浄し、 洗浄後 100 /1の洗浄バッファーを残す。 この細胞プ レートとサンプルプレートを FLIPRにセットしアツセィを行う (FLIPRにより、 サ ンプルプレ一トから のサンプルが細胞プレートへと移される) 。 (Molecular Probes) 40, and added to the above H / HBSS-Probenecid-FBS. After mixing, pipette 100 l of each well into the cell plate from which the culture solution has been removed using an 8-tube pipette. % C0 2 incubator base 1 hour at 37 ° C for at one coater in incubate (dye the loading). H / HBSS 1501 containing 2.5 mM Probenecid and 0.1% CHAPS is added to the sample for each assay (each fraction), diluted, and transferred to a 96-well plate for FLIPR (V-Bottom plate, Coster) (hereafter, sample plate) ). After dye loading of the cell plate is completed, wash the cell plate four times using a plate washer (Molecular Devices) with a washing buffer containing 2.5 mM Probenecid in ¾33, and leave 100/1 washing buffer after washing. Set the cell plate and sample plate in the FLIPR and perform the assay (the sample from the sample plate is transferred to the cell plate by the FLIPR).
(6) レセプターを発現する細胞に、 レセプ夕一ァゴニストを添加すると、 細 胞内イノシトール三リン酸濃度は上昇する。 本発明のペプチドの、 本発明のタン パク質発現細胞における細胞内ィノシト一ル三リン酸産生活性を利用することに より、 本発明のペプチドと本発明のタンパク質との結合性を変化させる化合物を スクリーニングすることができる。 (6) Addition of receptor agonist to cells expressing the receptor increases intracellular inositol triphosphate concentration. By utilizing the intracellular inositol triphosphate-producing activity of the peptide of the present invention in the protein-expressing cell of the present invention. Thus, a compound that changes the binding property between the peptide of the present invention and the protein of the present invention can be screened.
具体的には、 標識したイノシトールの存在下、 本発明のペプチドを、 本発明の タンパク質発現細胞に接触させた場合と、 本発明のペプチドおよび試験化合物を、 本発明のタンパク質発現細胞に接触させた場合における、 イノシトール三リン酸 産生活性を測定し、 比較することにより、 本発明のペプチドと本発明のタンパク 質との結合性を変化させる化合物をスクリーニングする。 測定は公知の方法に従 つて行う。 .  Specifically, a peptide of the present invention was brought into contact with a protein-expressing cell of the present invention in the presence of labeled inositol, and a peptide and a test compound of the present invention were brought into contact with a protein-expressing cell of the present invention. In this case, the inositol triphosphate-producing activity is measured and compared to screen for a compound that alters the binding property between the peptide of the present invention and the protein of the present invention. The measurement is performed according to a known method. .
本方法において、 イノシトール三リン酸産生活性を抑制する試験化合物を、 拮 抗阻害能力のある候補物質として選択することができる。  In this method, a test compound that suppresses inositol triphosphate-producing activity can be selected as a candidate substance capable of competitive inhibition.
一方、 試験化合物のみを本発明のタンパク質発現細胞に接触させ、 イノシト一 ル三リン酸産生上昇を測定することによってァゴニストのスクリーニングを行な うこともできる。  On the other hand, agonists can be screened by bringing only the test compound into contact with the protein-expressing cells of the present invention and measuring the increase in inositol triphosphate production.
スクリーニング法の一具体例を以下に述べる。  One specific example of the screening method is described below.
本発明のタンパク質発現細胞を 24穴プレートに播き、 1日間培養する。 その後、 myo- [2-¾] inos i to l (2. 5 Ci/we l l) を添加した培地で 1日間培養し、 細胞を放射 活性を有するィノシトールを無添加の培地でよく洗浄する。 本発明のぺプチドま たは本発明のペプチドおよび試験化合物を添加後、 10%過塩素酸を加え、 反応を 止める。 1. 5M水酸化カリウムおよび 60mM HEPES溶液で中和し、 0. 5mlの AGlx8樹 脂 (Bi o- Rad) を詰めたカラムに通し、 5mM 四ホウ酸ナトリウム (Na2B407) および 60raMギ酸アンモニゥムで洗浄した後、 1M ギ酸アンモニゥムおよび 0. 1Mギ酸で 溶出した放射活性を、 液体シンチレ一シヨンカウンタ一で測定する。 本発明のぺ プチドを添加しない場合の放射活性を 0%、 本発明のペプチドを添加した場合の 放射活性を 100%とし、 試験化合物の、 本発明のペプチドと本発明のタンパク質 の結合に対する影響を算出する。 The cells expressing the protein of the present invention are seeded on a 24-well plate and cultured for one day. Thereafter, the cells are cultured for 1 day in a medium supplemented with myo- [2-¾] inositol (2.5 Ci / well), and the cells are thoroughly washed with a medium without radioactive inositol. After adding the peptide of the present invention or the peptide and the test compound of the present invention, 10% perchloric acid is added to stop the reaction. 1. neutralized with 5M potassium hydroxide and 60 mM HEPES solution and then passed through a column packed with AGlx8 tree fat (Bi o-Rad) in 0. 5 ml, 5 mM sodium tetraborate (Na 2 B 4 0 7) and 60raM After washing with ammonium formate, the radioactivity eluted with 1 M ammonium formate and 0.1 M formic acid is measured with a liquid scintillation counter. The radioactivity when the peptide of the present invention is not added is 0%, and the radioactivity when the peptide of the present invention is added is 100%. calculate.
イノシトール三リン酸産生活性が、 例えば 50 %以下になる試験ィヒ合物を拮抗阻 害能力のある候補物質として選択することができる。  A test compound having an inositol triphosphate-producing activity of, for example, 50% or less can be selected as a candidate substance having an antagonistic ability.
( 7 ) T R E—レポ一夕一遺伝子ベクターを用いて、 本発明のペプチドの本発 明のタンパク質発現細胞に対する刺激活性を測定することにより、 本発明のぺプ チドと本発明のタンパク質との結合性を変化させる化合物をスクリーニングする ことができる。 (7) Using the TRE-repo overnight gene vector to measure the stimulatory activity of the peptide of the present invention on cells expressing the protein of the present invention, Compounds that alter the binding between the peptide and the protein of the present invention can be screened.
TRE (TPA response element) を含む DNAを、 ベクターのレポーター遺伝 子上流に挿入し、 TRE—レポ一夕一遺伝子ベクターを得る。 TRE—レポ一夕 一遺伝子ベクターを導入した本発明のタンパク質発現細胞において、 細胞内カル シゥム濃度の上昇を伴う刺激は、 TREを介したレポ一夕一遺伝子発現と、 それ に引き続くレポ一タ一遺伝子の遺伝子産物 (タンパク質) の産生を誘導する。 つ まり、 レポーター遺伝子タンパク質の酵素活性を測定することにより、 TRE— レポ一ター遺伝子べクタ一導入細胞内のカルシウム量の変動を検出することがで きる。  DNA containing a TRE (TPA response element) is inserted upstream of the reporter gene of the vector to obtain a TRE-repo overnight gene vector. In the protein-expressing cells of the present invention into which the TRE-repo overnight gene vector has been introduced, stimulation accompanied by an increase in the intracellular calcium concentration is caused by the TRE-mediated repo overnight gene expression and the subsequent reporter Induces the production of the gene product (protein) of the gene. In other words, by measuring the enzymatic activity of the reporter gene protein, it is possible to detect fluctuations in the amount of calcium in the cells into which the TRE-reporter gene vector has been introduced.
具体的には、 本発明のペプチドを、 TRE—レポ一ター遺伝子ベクター導入本 発明のタンパク質発現細胞に接触させた場合と、 本発明のペプチドおよび試験化 合物を、 TRE—レポ一夕一遺伝子ベクター導入本発明のタンパク質発現細胞に 接触させた場合における、 レポ一ター遺伝子タンパク質の酵素活性を測定し、 比 較することにより、 本発明のペプチドと本発明のタンパク質との結合性を変化さ せる化合物をスクリーニングする。  Specifically, when the peptide of the present invention is brought into contact with a TRE-reporter gene vector-introduced protein-expressing cell of the present invention, the peptide of the present invention and the test compound are expressed in the TRE-reporter gene. Vector-introduced Changes in the binding activity between the peptide of the present invention and the protein of the present invention by measuring and comparing the enzymatic activities of the reporter gene protein when brought into contact with the cells expressing the protein of the present invention. Screen compounds.
ベクタ一としては、 例えば、 ピツカジーン べィシックべクタ一、 ピツカジ一 ン ェンハンサ一ベクター (東洋インキ製造 (株) ) などが用いられる。 TRE を含む DNAを、 上記べクタ一のレポーター遺伝子、 例えばルシフェラ一ゼ遺伝 子上流のマルチクロ一ニングサイトに揷入し、 TRE—レポーター遺伝子べクタ 一とする。  Examples of the vector include Pitska Gene Basic Vector and Pitska Gene Enhancer Vector (Toyo Ink Mfg. Co., Ltd.). The TRE-containing DNA is inserted into a reporter gene of the above vector, for example, a multi-cloning site upstream of the luciferase gene, to obtain a TRE-reporter gene vector.
本方法において、 本発明のペプチドによるレポ一夕一遺伝子タンパク質の酵素 活性を抑制する試験化合物を、 拮抗阻害能力のある候補物質として選択すること ができる。  In this method, a test compound that suppresses the enzymatic activity of the repo overnight gene protein by the peptide of the present invention can be selected as a candidate substance having a competitive inhibition ability.
—方、 試験化合物のみを TRE—レポ一夕一遺伝子べクタ一導入本発明のタン パク質発現細胞に接触させ、 本発明のペプチドと同様な発光量の増加を測定する ことによりァゴニストのスクリーニングを行なうこともできる。  On the other hand, agonist screening is performed by contacting only the test compound with the TRE-repo allele-introduced gene-introduced protein-expressing cells of the present invention and measuring the increase in luminescence in the same manner as the peptide of the present invention. You can do it.
レポーター遺伝子として、 ルシフェラーゼを利用する例を用いて、 このスクリ —エング方法の具体例を以下に述べる。 T RE—レポ一ター遺伝子 (ルシフェラーゼ) を導入した本発明のタンパク質 発現細胞を、 24穴プレ一トに5 103じ 6 1 17 6 1 1で播種し、 48時間 培養する。 細胞を 0. 05% BSAおよび 20mM HEPESを含むハンク スバッファー (pH7. 4) で洗浄した後、 10 nMの本発明のペプチドまたは 10 nMの本発明のペプチドおよび試験化合物を添加し、 37°Cで 60分間反応 させる。 細胞をピツカジーン用細胞溶解剤 (東洋インキ製造 (株) ) で溶かし、 溶解液に発光基質 (東洋インキ製造 (株) ) を添加する。 ルシフェラーゼによる 発光は、 ルミノメ一ター、 液体シンチレ一シヨンカウン夕一またはトップカウン 夕一により測定する。 本発明のペプチドを添加した場合と、 Ι ΟηΜの本発明の ペプチドおよび試験化合物を添加した場合のルシフェラーゼによる発光量を測定 して、 比較する。 A specific example of this screening method will be described below using an example using luciferase as a reporter gene. Cells expressing the protein of the present invention into which the TRE-reporter gene (luciferase) has been introduced are seeded on a 24-well plate at 510 3 6 1 176 11 and cultured for 48 hours. After washing the cells with Hanks buffer (pH 7.4) containing 0.05% BSA and 20 mM HEPES, 10 nM of the peptide of the present invention or 10 nM of the peptide of the present invention and a test compound are added thereto, and the mixture is added at 37 ° C. And react for 60 minutes. Lyse the cells with a cell lysing agent for Pitka Gene (Toyo Ink Mfg. Co., Ltd.) and add a luminescent substrate (Toyo Ink Mfg. Co., Ltd.) to the lysate. Luminescence from luciferase is measured using a luminometer, liquid scintillation counter or top counter. The amount of luminescence by luciferase when the peptide of the present invention is added and when {と η} the peptide of the present invention and a test compound are added are measured and compared.
本発明のペプチドによる細胞内カルシウムの上昇によって、 ルシフェラ一ゼに よる発光量が増加する。 この増加を抑制する化合物を拮抗阻害能力のある候補物 質として選択することができる。  The increase in intracellular calcium by the peptide of the present invention increases the amount of luminescence by luciferase. A compound that suppresses this increase can be selected as a candidate substance capable of competitive inhibition.
レポーター遺伝子として、 例えば、 アルカリフォスファタ一ゼ、 クロラムフエ ニコ一レ ·ァセチリレ卜ランスフェラーセ ^chloramphenicol  Reporter genes include, for example, alkaline phosphatase, chloramphenic acid, acetylfurantransferrase ^ chloramphenicol
acetyltransferase) 、 _ガラクトシダ一ゼなどの遺伝子を用いてもよい。 こ れらのレポーター遺伝子タンパク質の酵素活性は、 公知の方法に従い、 または市 販の測定キットを用いて測定する。 'アル力リフォスファタ一ゼ活性は、 例えば和 光純薬製 Lumi-Phos 530を用いて、 クロラムフエニコ一ル ·ァセチルトランスフ エラーゼ活性は、 例えば和光純薬製 FAST CAT chrola即 henicol Genes such as acetyltransferase) and _galactosidase may be used. The enzyme activities of these reporter gene proteins are measured according to a known method or using a commercially available measurement kit. '' Alkali phosphatase activity was measured using, for example, Lumi-Phos 530 manufactured by Wako Pure Chemical, and chloramphenicol acetyltransferase was evaluated using, for example, FAST CAT chrola immediately manufactured by Wako Pure Chemical.
Acetyltransferase Assay KiTを用いて、 |S—ガラクトシダーゼ活性は、 例えば 和光純薬製 Aurora Ga卜 XEを用いて測定する。 Using Acetyltransferase Assay KiT, | S-galactosidase activity is measured using, for example, Aurora Gat XE manufactured by Wako Pure Chemical Industries.
(8) 本発明のタンパク質発現細胞は、 本発明のペプチドの刺激により、 MA Pキナーゼが活性化され、 増殖する。 この反応を利用して、 本発明のペプチドの 本発明のタンパク質発現細胞に対する刺激活性を測定することにより、 本発明の ペプチドと本発明のタンパク質との結合性を変化させる化合物をスクリーニング することができる。  (8) In the protein-expressing cell of the present invention, MAP kinase is activated and proliferates by stimulation of the peptide of the present invention. By utilizing this reaction to measure the stimulating activity of the peptide of the present invention on the cells expressing the protein of the present invention, it is possible to screen for a compound that alters the binding property between the peptide of the present invention and the protein of the present invention. .
具体的には、 本発明のペプチドを、 本発明のタンパク質発現細胞に接触させた 場合と、 本発明のペプチドおよび試験化合物を、 本発明のタンパク質発現細胞に 接触させた場合における、 細胞増殖を測定し、 比較することにより、 本発明のぺ プチドと本発明のタンパク質との結合性を変化させる化合物をスクリーニングす る。 Specifically, the peptide of the present invention was brought into contact with a cell expressing the protein of the present invention. By measuring and comparing the cell proliferation when the peptide of the present invention and the test compound are brought into contact with the protein-expressing cells of the present invention, the binding between the peptide of the present invention and the protein of the present invention can be determined. Screen for compounds that alter the.
本発明のタンパク質発現細胞の増殖は、 例えば、 MA Pキナーゼ活性、 チミジ ン取り込み活性、 細胞数などを測定すればよい。  Proliferation of the protein-expressing cells of the present invention may be measured, for example, by measuring MAP kinase activity, thymidine uptake activity, cell number, and the like.
具体例としては、 MA Pキナーゼ活性については、 本発明のペプチドまたは本 発明のペプチドおよび試験化合物を、 本発明のタンパク質発現細胞に添加した後、 細胞溶解液から抗] VI A Pキナーゼ抗体を用いた免疫沈降により M A Pキナーゼ分 画を得た後、 公知の方法、 例えば和光純薬製 MAP Kinase Assay〖 ぉょびァ- [32P] -ATPを使用して M A Pキナーゼ活性を測定し、 比較する。 As a specific example, for the MAP kinase activity, after adding the peptide of the present invention or the peptide of the present invention and a test compound to the cells expressing the protein of the present invention, an anti-VIAP kinase antibody was used from a cell lysate. After obtaining the MAP kinase fraction by immunoprecipitation, the MAP kinase activity is measured using a known method, for example, MAP Kinase Assay II- [ 32 P] -ATP manufactured by Wako Pure Chemical Industries, and compared.
チミジン取り込み活性については、 本発明のタンパク質発現細胞を 2 4穴プレ —トに播種し、 培養し、 本発明のペプチドまたは本発明のペプチドおよび試験化 合物を添加した後、 放射活性により標識したチミジン (例、 [methy卜3 H] -チミジ ンなど) を加え、 その後、 細胞を溶解し、 細胞内に取り込まれたチミジンの放射 活性を、 液体シンチレ一シヨンカウンターで計数することにより、 チミ'ジン取り 込み活性を測定し、 比較する。 Regarding the thymidine incorporation activity, the protein-expressing cells of the present invention were seeded on a 24-well plate, cultured, added with the peptide of the present invention or the peptide of the present invention and a test compound, and then labeled with radioactivity. thymidine (e.g., [methy Bok 3 H] - thymidine down, etc.) is added, then the cells were lysed and the radioactivity of the thymidine incorporated into the cells, by counting in a liquid scintillator one Chillon counter, thymidylate ' Measure gin uptake activity and compare.
細胞数の測定については、 本発明のタンパク質発現細胞を 2 4穴プレートに播 種し、 培養し、 本発明のペプチドまたは本発明のペプチドおよび試験化合物を添 加した後、 MT T (3- (4, 5-dimethyl-2-thiazolyl) -2, 5-diphenyl-2H- tetrazo l ium bromide) を添加する。 細胞内に取り込まれて MT Tが変化した M T Tホルマザンを、 塩酸にて酸性としたイソプロパノール水溶液で細胞を溶解し た後、 5 7 0 n mの吸収によって測定し、 比較する。  For the measurement of the cell number, the protein-expressing cells of the present invention were seeded on a 24-well plate, cultured, and after adding the peptide of the present invention or the peptide of the present invention and a test compound, MTT (3- ( Add 4,5-dimethyl-2-thiazolyl) -2,5-diphenyl-2H-tetrazolium bromide). MTT formazan in which MTT has been changed by being taken into cells is dissolved in an aqueous solution of isopropanol acidified with hydrochloric acid, and then measured by absorption at 570 nm for comparison.
本方法において、 本発明のタンパク質発現細胞の増殖を抑制する試験化合物を、 拮抗阻害能力のある候補物質として選択することができる。  In this method, a test compound that suppresses the growth of the protein-expressing cell of the present invention can be selected as a candidate substance having a competitive inhibition ability.
一方、 試験化合物のみを本発明のタンパク質発現細胞に接触させ、 本発明のぺ プチドと同様な細胞増殖活性を測定することによりァゴニス卜のスクリ一ニング を行なうこともできる。  On the other hand, agonists can be screened by bringing only the test compound into contact with the protein-expressing cell of the present invention and measuring the cell proliferation activity similar to that of the peptide of the present invention.
チミジン取り込み活性を利用するスクリーニング法の一具体例を以下に述べる。 本発明の夕ンパク質発現細胞を 24穴プレートに 5000個/ゥエル播き、 1日間培養 する。 次に血清を含まない培地で 2日間培養し、 細胞を飢餓状態にする。 本発明 のペプチドまたは本発明のペプチドおよび試験化合物を、 細胞に添加して 24時間 培養した後、 [methy卜3 H] -チミジンをゥエル当たり 0. 015MBq添加し、 6時間培養す る。 細胞を PBSで洗った後、 メタノールを添加して 10分間放置する。 次に 5 %トリ クロ口酢酸を添加して 15分間放置後、 固定された細胞を蒸留水で 4回洗う。 0. 3N 水酸化ナトリゥム溶液で細胞を溶解し、 溶解液中の放射活性を液体シンチレーシ ョンカウンタ一で測定する。 One specific example of the screening method using the thymidine uptake activity is described below. The evening protein-expressing cells of the present invention are seeded at 5,000 cells / well in a 24-well plate and cultured for one day. The cells are then starved in a serum-free medium for 2 days. The peptide or peptide and a test compound of the present invention of the present invention, after incubation was added to the cells 24 hours, [methy Bok 3 H] - thymidine was added Ueru per 0. 015MBq, incubation 6 hours. After washing the cells with PBS, add methanol and let stand for 10 minutes. Next, add 5% trichloroacetic acid and let stand for 15 minutes. Wash the fixed cells four times with distilled water. Lyse the cells with 0.3N sodium hydroxide solution and measure the radioactivity in the lysate with a liquid scintillation counter.
本発明のペプチドを添加した場合の放射活性の増加を抑制する試験化合物を、 拮抗阻害能力のある候補物質として選択することができる。  A test compound that suppresses an increase in radioactivity when the peptide of the present invention is added can be selected as a candidate substance capable of competitive inhibition.
( 9 ) 本発明のタンパク質発現細胞は、 本発明のペプチドの刺激により、 カリ ゥムチャネルが活性化し、 細胞内にある Kイオンが、 細胞外に流出する。 この反 応を利用して、 本発明のペプチドの本発明のタンパク質発現細胞に対する刺激活 性を測定することにより、 本発明のぺプチドと本発明のタンパク質との結合性を 変化させる化合物をスクリーニングすることができる。  (9) In the protein-expressing cell of the present invention, the stimulation of the peptide of the present invention activates the potassium channel, and intracellular K ions flow out of the cell. By utilizing this reaction, the stimulating activity of the peptide of the present invention on cells expressing the protein of the present invention is measured to screen for compounds that alter the binding between the peptide of the present invention and the protein of the present invention. be able to.
Kイオンと同族元素である R bイオン (ルビジウムイオン) は、 Kイオンと区 別無く、 カリウムチャネルを通って細胞外に流出する。 よって、 本発明のタンパ ク質発現細胞に、 放射活性同位体である R b ( [ 8 6 R b ] ) を取り込ませておい た後、 本発明のペプチドの刺激によって流出する8 6 R bの流れ (流出活性) を測 定することにより、 本発明のペプチドの本発明のタンパク質発現細胞に対する刺 激活性を測定する。 Rb ions (rubidium ions), which are homologous to K ions, flow out of the cell through potassium channels without distinction from K ions. Thus, the protein-expressing cells of the present invention, a radioactive isotope R b After allowed incorporation of ([8 6 R b]) , the 8 6 R b flowing out by stimulation of the peptides of the present invention By measuring the flow (efflux activity), the stimulating activity of the peptide of the present invention on the cells expressing the protein of the present invention is measured.
具体的には、 86R bの存在下、 本発明のペプチドを、 本発明のタンパク質発現 細胞に接触させた場合と、 本発明のペプチドおよび試験化合物を、 本発明のタン パク質発現細胞に接触させた場合における、 8 6 R bの流出活性を測定し、 比較す ることにより、 本発明のペプチドと本発明のタンパク質との結合性を変化させる 化合物をスクリーニングする。 Specifically, when the peptide of the present invention is brought into contact with the protein-expressing cell of the present invention in the presence of 86 Rb, the peptide of the present invention and the test compound are brought into contact with the protein-expressing cell of the present invention. in the case of it is to measure the efflux activity of 8 6 R b, by comparing, for screening the compound that changes the binding property between the protein of the peptide and the invention of the present invention.
本方法において、 本発明のペプチド刺激による8 6 R bの流出活性の上昇を抑制 する試験化合物を、 拮抗阻害能力のある候補物質として選択することができる。 一方、 試験化合物のみを本発明のタンパク質発現細胞に接触させ、 本発明のぺ プチドと同様な8 6 R bの流出活性の上昇を測定することによりァゴニス卜のスク リ一ニングを行なうこともできる。 In this method, the 8 6 R b test compound to suppress an increase in efflux activity of by peptide stimulation of the present invention, it can be selected as a candidate substance capable of competitive inhibition. On the other hand, only the test compound is brought into contact with the protein-expressing cell of the present invention, It is also possible to perform the disk re-learning of Agonisu Bok by measuring the increase in peptide similar 8 6 R b of efflux activity.
スクリ一ニング法の一具体例を以下に述べる。  One specific example of the screening method is described below.
本発明のタンパク質発現細胞を 2 4穴プレートに播き、 2日間培養する。 その 後、 1 m C i Zm 1の86 R b C 1を含む培地中で 2時間保温する。 細胞を培地でよ く洗浄し、 外液中の86 R b C 1を完全に除く。 本発明のペプチドまたは本発明の ペプチドおよび試験化合物を細胞に添加し、 3 0分後外液を回収し、 ァカウン夕 一で放射活性を測定し、 比較する。 The protein-expressing cells of the present invention are seeded on a 24-well plate and cultured for 2 days. Thereafter, the cells are incubated for 2 hours in a medium containing 1 mCiZm1 and 86 RbC1. Wash cells thoroughly with medium to completely remove 86 RbC1 in the external solution. The peptide of the present invention or the peptide of the present invention and a test compound are added to the cells, and after 30 minutes, the external solution is collected, and the radioactivity is measured and compared at a local time.
本発明のペプチド刺激による86 R bの流出活性の上昇を抑制する試験化合物を, 拮抗阻害能力のある候補物質として選択することができる。 A test compound that suppresses an increase in the 86 Rb efflux activity due to peptide stimulation of the present invention can be selected as a candidate substance having a competitive inhibitory ability.
( 1 0 ) 本発明のタンパク質発現細胞が本発明のペプチドに反応し、 細胞外の p Hが変化する。 この反応を利用して、 本発明のペプチドの本発明のタンパク質 発現細胞に対する刺激活性を測定することにより、 本発明のぺプチドと本発明の タンパク質との結合性を変化させる化合物をスクリーニングすることができる。 具体的には、 本発明のペプチドを、 本発明のタンパク質発現細胞に接触させた 場合と、 本発明のペプチドおよび試験化合物を、 本発明のタンパク質発現細胞に 接触させた場合における、 細胞外の p H変化を測定し、 比較することにより、 本 発明のぺ: °チドと本発明のタンパク質との結合性を変化させる化合物をスクリー ニングする。  (10) Extracellular pH changes when cells expressing the protein of the present invention react with the peptide of the present invention. By utilizing this reaction to measure the stimulating activity of the peptide of the present invention on cells expressing the protein of the present invention, it is possible to screen for a compound that alters the binding between the peptide of the present invention and the protein of the present invention. it can. Specifically, the extracellular p in the case where the peptide of the present invention is brought into contact with the protein-expressing cell of the present invention and in the case where the peptide of the present invention and the test compound are brought into contact with the protein-expressing cell of the present invention. By measuring and comparing the change in H, a compound that changes the binding property between the peptide of the present invention and the protein of the present invention is screened.
細胞外 p H変化は、 例えば、 Cytosensor装置 (モレキュラーデバイス社) を使 用して測定する。 ,  The extracellular pH change is measured using, for example, a Cytosensor device (Molecular Devices). ,
本方法において、 本発明のペプチドによる細胞外 p H変化を抑制する試験化合 物を、 拮抗阻害能力のある候補物質として選択することができる。  In this method, a test compound that suppresses the extracellular pH change caused by the peptide of the present invention can be selected as a candidate substance capable of competitive inhibition.
一方、 試験化合物のみを本発明のタンパク質発現細胞に接触させ、 本発明のぺ プチドと同様な細胞外 p H変化を測定することによりァゴニス卜のスクリーニン グを行なうこともできる。  On the other hand, agonist screening can be performed by bringing only the test compound into contact with the protein-expressing cell of the present invention and measuring the extracellular pH change similar to that of the peptide of the present invention.
スクリーニング法の一具体例を以下に述べる。  One specific example of the screening method is described below.
本発明のタンパク質発現細胞を Cytosensor装置用のカプセル内で終夜培養し、 装置のチャンバ一にセットして細胞外 p Hが安定するまで約 2時間、 0 . 1 % 83八を含む1 ? 1 1640培地 (モレキュラーデバイス社製) を灌流させる。 PHが安定した後、 本発明のペプチドまたは本発明のペプチドおょぴ試験化合物 を含む培地を細胞上に灌流させる。 灌流によって生じた培地の pH変化を測定し、 '比較する。 The protein-expressing cells of the present invention are cultured overnight in a capsule for a Cytosensor device, set in the chamber of the device, and set to 0.1% for about 2 hours until the extracellular pH is stabilized. Perfuse 1 to 1640 medium (Molecular Devices) containing 83-8. After the pH is stabilized, a medium containing the peptide of the present invention or the peptide of the present invention and the test compound is perfused on the cells. Measure and compare the pH change of the medium caused by perfusion.
本発明のペプチドによる細胞外 pH変化を抑制する化合物を拮抗阻害能力のあ る候補物質として選択することができる。 '  A compound that suppresses the extracellular pH change caused by the peptide of the present invention can be selected as a candidate substance having a competitive inhibition ability. '
丄 1) 酵母 (Sacc aromyces cerevisiae) の haploida - mating Type (MAT ひ) の性フェロモンレセプ夕一 ST e 2は、 Gタンパク質 Gp a 1と共役してお り、 性フェロモンひ—mating factorに応答して MAPキナーゼを活性化し、 こ れに引き続き、 F a r 1 (cell-cycle arrest) および転写活性化因子 S t e 1 2が活性化される。 S t e l 2は、 種々のタンパク質 (例えば、 接合に関与する FUS 1) の発現を誘導する。 一方、 制御因子 S s t 2は上記の過程に抑制的に 機能する。 この系において、 レセプ夕一遺伝子を導入し'た酵母を作製し、 レセプ 夕一ァゴニストの刺激により酵母細胞内のシグナル伝達系を活性化し、 その結果 生じる増殖などを指標として用いる、 レセプ夕一ァゴニストとレセプターとの反 応の測定系の試みが行なわれている (Trends in Biotechnology, 15巻, 487-494 頁, 1997年) 。 上記のレセプター遺伝子導入酵母の系を利用して、 本発明のぺプ チドと本発明のタンパク質との結合性を変化させる化合物をスクリーニングする ことができる。 '  丄 1) The sex pheromone receptor, ST e2, of the haploida-mating type (MAT) of yeast (Sacc aromyces cerevisiae) is conjugated to the G protein, Gpa1, and responds to the sex pheromone mating factor. To activate MAP kinase, followed by activation of Far 1 (cell-cycle arrest) and the transcriptional activator Ste12. Stel 2 induces the expression of various proteins (eg, FUS 1 involved in conjugation). On the other hand, the control factor S st 2 functions in the above process in a suppressive manner. In this system, a yeast into which a receptor gene has been introduced is produced, a signal transduction system in the yeast cell is activated by stimulation of the receptor gene, and the resulting proliferation is used as an index. Attempts have been made to measure the reaction between a protein and a receptor (Trends in Biotechnology, 15, 487-494, 1997). Using the above-described receptor gene-introduced yeast system, a compound capable of changing the binding property between the peptide of the present invention and the protein of the present invention can be screened. '
具体例を以下に示す。  Specific examples are shown below.
MAT α酵母の S t e 2および Gp a 1をコードする遺伝子を除去し、 代わり に、 本発明のタンパク質遺伝子および Gp a 1-Ga i 2融合タンパク質をコー ドする遺伝子を導入する。 F a rをコードする遺伝子を除去して ceU- cycle arrestが生じないようにし、 また、 S s tをコードする遺伝子を除去して本発明 のペプチドに対する応答の感度を向上させておく。 さらに、 FUS 1にヒスチジ ン生合成遺伝子 H I S 3を結合した FUS 1— H I S 3遺伝子を導入する。 この 遺伝子組換え操作は、 例えば、 Molecular and Cellular Biology, 15巻, 6188- 6195頁, 1995年に記載の方法において、 ソマトス夕チンレセプ夕一タイプ 2 (SSTR2) 遺伝子を、 本発明のタンパク質に置き換えて実施することができる。 このように構築された形質変換酵母は、 本発明のぺプチドに高感度で反応し、 その結果、 MA Pキナーゼの活性化が起き、 ヒスチジン生合成酵素が合成される ようになり、 ヒスチジン欠乏培地で生育可能になる。 The genes encoding Ste2 and Gpa1 of the MATα yeast are removed, and instead, the protein gene of the present invention and a gene encoding the Gpa1-Gai2 fusion protein are introduced. The gene encoding Far is removed to prevent ceU-cycle arrest from occurring, and the gene encoding Sst is removed to increase the sensitivity of the response to the peptide of the present invention. Furthermore, the FUS1—HIS3 gene in which the histidine biosynthesis gene HIS3 is linked to FUS1 is introduced. This genetic recombination operation can be performed, for example, by replacing the somatostin-tin receptor-type-1 (SSTR2) gene with the protein of the present invention in the method described in Molecular and Cellular Biology, vol. 15, p. 6188-6195, 1995. Can be implemented. The transformed yeast thus constructed reacts with high sensitivity to the peptide of the present invention, resulting in the activation of MAP kinase, the synthesis of histidine biosynthesis enzyme, and the histidine-deficient medium. It is possible to grow in.
従って、 上記の本発明のタンパク質発現酵母 (S t e 2遺伝子および G p a 1 遺伝子が除去され、 本発明のタンパク質遺伝子および G p a 1 - G a i 2融合夕 ンパク質コード遺伝子が導入され、 F a r遺伝子および S s t遺伝子が除去され、 F U S 1一 H I S 3遺伝子が導入された ΜΑ Τα酵母) を、 ヒスチジン欠乏培地 で培養し、 本発明のぺプチドまたは本発明のぺプチドおよび試験化合物を接触さ せ、 該酵母の生育を測定し、 比較することにより、 本発明のペプチドと本発明の タンパク質との結合性を変化させる化合物をスクリーニングすることができる。 本方法において、 該酵母の生育を抑制する試験化合物を、 拮抗阻害能力のある 候補物質として選択することができる。  Therefore, the above-described protein-expressing yeast of the present invention (the Ste2 gene and the Gpa1 gene were removed, the protein gene of the present invention and the Gpa1-Gai2 fusion protein-encoding gene were introduced, and the Far gene was introduced. Sα yeast with the FUS1-1HIS3 gene removed and the Sst gene removed) is cultured in a histidine-deficient medium, and the peptide of the present invention or the peptide of the present invention and a test compound are contacted with each other; By measuring and comparing the growth of the yeast, it is possible to screen for compounds that alter the binding between the peptide of the present invention and the protein of the present invention. In this method, a test compound that suppresses the growth of the yeast can be selected as a candidate substance having a competitive inhibition ability.
一方、 試験化合物のみを上記の本発明のタンパク質発現酵母に接触させ、 本発 明のぺプチドと同様な酵母の生育を測定することによりァゴニストのスクリ一二 ングを行なうこともできる。  On the other hand, agonist screening can also be performed by bringing only the test compound into contact with the above-described yeast expressing the protein of the present invention and measuring the growth of yeast similar to the peptide of the present invention.
スクリーニング法の一具体例を以下に述べる。  One specific example of the screening method is described below.
上記の本発明のタンパク質発現酵母を完全合成培地の液体培地で終夜培養し、 その後、 ヒスチジンを除去した溶解寒天培地に、 2 X 1 04 c e 1 1 /m 1の濃度 になるように加える。 ついで、 9 X 9 c mの角形シャーレに播く。 寒天が固化し た後、 本発明のペプチドまたは本発明のペプチドおよび試験化合物をしみこませ た滅菌濾紙を寒天表面におき、 3 0 °Cで 3日間培養する。 試験化合物の影響は、 濾紙の周囲の酵母の生育を、 本発明のぺプチドのみをしみこませた滅菌濾紙を用 いた場合と比較する。 また、 あらかじめ、 ヒスチジンを除去した寒天培地に本発 明のぺプチドを添加しておき、 滅菌濾紙に試験化合物のみをしみこませて酵母を 培養し、 シャーレ全面での酵母の生育が濾紙の周囲で影響を受けることを観察し てもよい。 The above-described protein-expressing yeast of the present invention is cultured overnight in a liquid medium of a complete synthetic medium, and then added to a dissolved agar medium from which histidine has been removed to a concentration of 2 × 10 4 ce 11 / m 1. Then sow in a 9 x 9 cm square petri dish. After the agar solidifies, a sterile filter paper impregnated with the peptide of the present invention or the peptide of the present invention and the test compound is placed on the agar surface, and cultured at 30 ° C for 3 days. The effect of the test compound is to compare the growth of yeast around the filter paper with the use of sterile filter paper impregnated with only the peptide of the present invention. In addition, the peptide of the present invention is added to the agar medium from which histidine has been removed in advance, and the test compound is soaked in sterile filter paper to culture the yeast. You may observe that they are affected.
酵母の生育を抑制する化合物を拮抗阻害能力のある候補物質として選択するこ とができる。  A compound that suppresses the growth of yeast can be selected as a candidate substance having a competitive inhibition ability.
( 1 2 ) 本発明のタンパク質遺伝子 R N Aをアフリカッメガエル卵母細胞に注 入し、 本発明のペプチドによって刺激すると細胞カルシウム濃度が上昇して、 calcium-activated chloride currentが生じる。 これは、 膜電位の変化としてと らえることができる (Kイオン濃度勾配に変化がある場合も同様) 。 本発明のぺ プチドによって生じる本発明のタンパク質導入アフリカッメガエル卵母細胞にお ける上記反応を利用して、 本発明のペプチドの本発明のタンパク質発現細胞に対 する刺激活性を測定することにより、 本発明のペプチドと本発明のタンパク質と の結合性を変化させる化合物をスクリーニングすることができる。 (12) Inject the protein gene RNA of the present invention into Xenopus oocytes When stimulated with the peptide of the present invention, the concentration of cellular calcium increases, resulting in calcium-activated chloride current. This can be considered as a change in the membrane potential (even when there is a change in the K ion concentration gradient). By utilizing the above reaction in the Xenopus laevis oocytes transfected with the protein of the present invention caused by the peptide of the present invention, the stimulating activity of the peptide of the present invention on the protein-expressing cell of the present invention is measured. Compounds that alter the binding between the peptide of the present invention and the protein of the present invention can be screened.
具体的には、 本発明のペプチドを、 本発明のタンパク質遺伝子 RN A導入ァフ リカツメガエル卵母細胞に接触させた場合と、 本発明のぺプチドおよび試験化合 物を、 本発明のタンパク質遺伝子 RNA導入アフリカッメガエル卵母細胞に接触 させた場合における、 細胞膜電位の変化を測定し、 比較することにより、 本発明 のペプチドと本発明のタンパク質との結合性を変化させる化合物をスクリ一ニン グする。  Specifically, the case where the peptide of the present invention is brought into contact with the protein gene of the present invention, RNA-introduced African Xenopus oocytes, and the case where the peptide and the test compound of the present invention are brought into contact with the protein gene of the present invention By measuring and comparing changes in cell membrane potential when brought into contact with the introduced Xenopus laevis oocytes, a compound that changes the binding property between the peptide of the present invention and the protein of the present invention is screened. I do.
本方法において、 細胞膜電位変化を抑制する試験化合物を、 拮抗阻害能力のあ る候補物質として選択することができる。  In this method, a test compound that suppresses a change in cell membrane potential can be selected as a candidate substance having a competitive inhibition ability.
一方、 試験化合物のみを本発明のタンパク質遺伝子 RN A導入アフリカッメガ エル卵母細胞に接触させ、 本発明のぺプチドと同様な細胞膜電位変化を測定する ことによりァゴニストのスクリーニングを行なうこともできる。  On the other hand, agonists can be screened by bringing only test compounds into contact with the protein gene of the present invention, RNA-introduced African oocytes, and measuring changes in cell membrane potential similar to those of the peptides of the present invention.
スクリーニング法の一具体例を以下に述べる。  One specific example of the screening method is described below.
氷冷して動けなくなった雌のアフリカッメガエルから取り出した、 卵母細胞塊 を、 MBS液 (88mM NaCl, lniM KC1, 0.41mM CaCl2, 0.33mM Ca(N03) 2, 0.82mM MgS04, 2.4mM NaHC03, lOmM HEPES; pH7.4) に溶かしたコラーゲナーゼ (0. 5 mg/m 1 ) で卵塊がほぐれるまで 19 °C、 1〜 6時間、 150 r p mで処理す る。 外液を MB S液に置換することで 3度洗浄し、 マイクロマニピュレーターで 本発明のタンパク質遺伝子 p o 1 y A付加 cRN A (50n g/50 η 1 ) を 卵母細胞にマイクロインジェクションする。 In ice water was removed from female African Tsu mega El became stuck, the oocytes mass, MBS solution (88mM NaCl, lniM KC1, 0.41mM CaCl 2, 0.33mM Ca (N0 3) 2, 0.82mM MgS0 4 , 2.4mM NaHC0 3, lOmM HEPES; pH7.4) in melted collagenase (0. 5 mg / m 1) 19 ° until the oocytes are loosened by C,. 1 to 6 hours, that processes at 0.99 rpm. The external solution is washed three times by replacing the MBS solution, and the oocyte is microinjected with the protein gene po1yA-added cRNA (50 ng / 50η1) of the present invention using a micromanipulator.
本発明のタンパク質遺伝子 mRN Αは、 組織や細胞から調製してもよく、 ブラ スミドから in vitroで転写してもよい。 本発明のタンパク質遺伝子 mRNAを M B S液中で 20°Cで 3日培養し、 これを Ringer液を流している voltage clamp装 置のくぼみに置き、 電位固定用ガラス微小電極および電位測定用ガラス微小電極 を細胞内に刺入し、 (一) 極は細胞外に置く。 電位が安定したら、 本発明のぺプ チドまたは本発明のペプチドおよび試験化合物を含む Ringer液を流して電位変化 を記録する。 試験化合物の影響は、 本発明のタンパク質遺伝子 RNA導入ァフリ カツメガエル卵母細胞の細胞膜電位変化を、 本発明のぺプチドのみ含む Ringer液 を流した場合と比較するこ によつて測定することができる。 The protein gene mRNΑ of the present invention may be prepared from a tissue or a cell, or may be transcribed from a plasmid in vitro. The protein gene mRNA of the present invention was cultured in MBS solution at 20 ° C for 3 days, and this was cultured in a voltage clamp equipped with Ringer solution. Insert the potential fixing glass microelectrode and the potential measurement glass microelectrode into the cell, and place the electrode outside the cell. When the potential is stabilized, a Ringer solution containing the peptide of the present invention or the peptide of the present invention and a test compound is passed, and the change in potential is recorded. The effect of the test compound can be measured by comparing the change in the cell membrane potential of the oocytes of the present invention into which the protein gene of the present invention has been transfected with RNA, compared to the case where a Ringer solution containing only the peptide of the present invention is applied. .
細胞膜電 変化を抑制する化合物を拮抗阻害能力のある候補物質として選択す ることができる。  A compound that suppresses cell membrane electrical changes can be selected as a candidate substance having a competitive inhibition ability.
上記の系において、 電位の変化量を増大させると、 測定しやすくなるため、 各 種の Gタンパク質遺伝子の p o 1 y A付加 R NAを導入してもよい。 また、 力 ルシゥム存在下で発光を生じるようなタンパク質 (例、 aequorinなど) の遺伝子 の p o l y A付加 R NAを共インジェクションすることにより'、 膜電位変化で はなく発光量を測定することもできる。  In the above system, the increase in the amount of change in the potential facilitates the measurement, and thus, the pO1A-added RNA of each G protein gene may be introduced. In addition, by co-injecting polyA-added RNA of a gene of a protein (eg, aequorin, etc.) that produces luminescence in the presence of potassium, it is possible to measure luminescence instead of membrane potential change.
本発明のペプチドと本発明のタンパク質との結合性を変化させる化合物または その塩のスクリーニング用キットは、 本発明のタンパク質、 本発明のタンパク質 を含有する細胞、 あるいは本発明のタンパク質を含有する細胞の膜画分、 および 本発明のペプチドを含有するものである。  A screening kit for a compound or a salt thereof that alters the binding property between the peptide of the present invention and the protein of the present invention includes a protein of the present invention, a cell containing the protein of the present invention, or a cell containing the protein of the present invention. A membrane fraction, and a peptide of the present invention.
本発明のスクリーニング用キッ卜の例としては、 次のものがあげられる。 1 . スクリーニング用試薬  Examples of the screening kit of the present invention include the following. 1. Screening reagent
(i) 測定用緩衝液および洗浄用緩衝液  (i) Measurement buffer and washing buffer
Hanks' Bal anced Sal t Solut ion (ギブコ社製) に、 0. 05%のゥシ血清アルブ ミン (シグマ社製) を加えたもの。  Hanks' Balanced Salt Solution (manufactured by Gibco) plus 0.05% serum albumin (manufactured by Sigma).
孔径 0. 45 mのフィルターで濾過滅菌し、 4°Cで保存するか、 あるいは用時調製 してもよい。  Sterilize by filtration with a 0.45 m pore size filter and store at 4 ° C, or prepare freshly before use.
(i i) 本発明のタンパク質の標品  (ii) Sample of the protein of the present invention
本発明の夕ンパク質を発現させた CH0細胞を、 12穴プレー卜に 5 X 105個 Z穴で継 代し、 37°C、 5% C02、 95 % ai rで 2日間培養したもの。 CH0 cells expressing the protein of the present invention were subcultured in 12-well plates at 5 × 10 5 Z-wells and cultured at 37 ° C., 5% CO 2 , 95% air for 2 days. .
(i i i) 標識リガンド  (i i i) Labeled ligand
〔¾〕 、 (1251] 、 〔14C〕 、 〔35S〕 などで標識した本発明のペプチド。 適当な溶媒または緩衝液に溶解したものを 4であるいは- 20 °Cにて保存し、 用時 に測定用緩衝液にて 1 /Mに希釈する。 [¾], (125 1], [14 C], the peptides of the present invention labeled with a [35 S]. Dissolve in a suitable solvent or buffer at 4 or at -20 ° C, and dilute to 1 / M with the measuring buffer before use.
(iv) リガンド標準液  (iv) Ligand standard solution
本発明のペプチドを 0.1%ゥシ血清アルブミン (シグマ社製) を含む PBSで lmM となるように溶解し、 _20°Cで保存する。  The peptide of the present invention is dissolved to a concentration of lmM in PBS containing 0.1% ゥ serum albumin (Sigma) and stored at -20 ° C.
2. 測定法 - 2. Measurement method-
(i) 12穴組織培養用プレ一トにて培養した本発明のタンパク質を発現させた細 胞を、 測定用緩衝液 lmlで 2回洗浄した後、 490/ lの測定用緩衝液を各穴に加える ( (i) The cells expressing the protein of the present invention cultured on a 12-well tissue culture plate were washed twice with 1 ml of the measurement buffer, and 490 / l of the measurement buffer was added to each well. Add to (
(ii) 10— 31()Mの試験化合物溶液を 5 1加えた後、 標識した本発明のペプチド を 5 1加え、 室温にて 1時間反応させる。 非特異的結合量を知るためには試験化 合物のかわりに 10— ¾の本発明のペプチドを 5 / 1加えておく。 After the test compound solution (ii) 10- 3 ~ 1 ( ) M 5 1 added, the peptide of the present invention labeled 5 1 was added to react at room temperature for one hour. In order to know the amount of non-specific binding, 5/1 of the peptide of the present invention is added instead of the test compound.
(iii) 反応液を除去し、 1mlの洗浄用緩衝液で 3回洗浄する。 細胞に結合した標 識した本発明のペプチドを 0.2N NaOH-1 % SDSで溶解し、 4mlの液体シンチレ一夕 — A (和光純薬製) と混合する。 ,  (iii) Remove the reaction solution and wash three times with 1 ml of washing buffer. The labeled peptide of the present invention bound to the cells is dissolved in 0.2N NaOH-1% SDS, and mixed with 4 ml of liquid scintillator overnight-A (manufactured by Wako Pure Chemical). ,
(iv) 液体シンチレ一シヨンカウンター (ベックマン社製) を用いて放射活性を 測定し、 Percent Maximum Binding (PMB) を次の式 〔数 1〕 で求める。  (iv) The radioactivity is measured using a liquid scintillation counter (manufactured by Beckman), and the Percent Maximum Binding (PMB) is determined by the following equation (Equation 1).
〔数 1〕  (Equation 1)
PMB= [ (B - NSB) / (B0-NSB) ] xl O O PMB = [(B-NSB) / (B 0 -NSB)] xl OO
PMB : Percent Ma imum Binding  PMB: Percent Ma imum Binding
B :検体を加えた時の値  B: Value when the sample is added
NSB : Non-specific Binding (非特異的結合量) '  NSB: Non-specific Binding ''
B。 :最大結合量  B. : Maximum binding amount
本発明のスクリーニング方法またはスクリーニング用キットを用いて得られる 化合物またはその塩は、 本発明のペプチドと本発明のタンパク質との結合を変化 させる (結合を阻害または促進する) 化合物であり、 具体的には本発明のタンパ ク質を介して細胞刺激活性を有する化合物またはその塩 (いわゆる本発明のタン パク質のァゴニスト) 、 または該刺激活性を有しない化合物 (いわゆる本発明の タンパク質のアンタゴニスト) である。 該化合物としては、 ペプチド、 タンパク、 非ペプチド性化合物、 合成化合物、 発酵生産物などがあげられ、 これら化合物は 新規な化合物であってもよいし、 公知の化合物であってもよい。 The compound or a salt thereof obtained by using the screening method or the screening kit of the present invention is a compound that changes the binding (inhibits or promotes the binding) between the peptide of the present invention and the protein of the present invention. Is a compound having a cell stimulating activity via the protein of the present invention or a salt thereof (a so-called agonist of the protein of the present invention), or a compound not having the stimulating activity (a so-called antagonist of the protein of the present invention). . Such compounds include peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, and the like. It may be a novel compound or a known compound.
上記本発明のタンパク質のァゴ: ϋス卜であるかアンタゴニストであるかの具体 的な評価方法は以下の (Α) または (Β ) に従えばよい。  The specific method for evaluating whether the above-mentioned protein of the present invention is an agonist or an antagonist may be according to the following (Α) or (Β).
(Α) 前記のスクリ一ニング方法で示されるバインディング ·アツセィを行い、 本発明のペプチドと本発明のタンパク質との結合性を変化させる (特に、 結合を 阻害する) 化合物を得た後、 該化合物が上記した本発明のタンパク質を介する細 胞刺激活性を有しているか否かを測定する。 細胞刺激活性を有する化合物または その塩は本発明のタンパク質のァゴニストであり、 該活性を有しない化合物また はその塩は本発明のタンパク質のアン夕ゴニストである。  (Ii) The binding assay shown in the above screening method is performed to obtain a compound that changes the binding property (particularly, inhibits the binding) between the peptide of the present invention and the protein of the present invention. Is determined whether it has the above-described cell stimulating activity via the protein of the present invention. A compound having a cell stimulating activity or a salt thereof is an agonist of the protein of the present invention, and a compound or a salt thereof having no such activity is an agonist of the protein of the present invention.
(B ) (a)試験化合物を本発明のタンパク質を含有する細胞に接触させ、 上記本 発明のタンパク質を介した細胞刺激活性を測定する。 細胞刺激活性を有する化合 物またはその塩は本発明のタンパク質のァゴニストである。  (B) (a) A test compound is brought into contact with a cell containing the protein of the present invention, and the cell stimulating activity mediated by the protein of the present invention is measured. The compound having a cell stimulating activity or a salt thereof is an agonist of the protein of the present invention.
(b) 本発明のタンパク質を活性化する化合物 (例えば、 本発明のペプチドまたは 本発明のタンパク質のァゴニストなど) を本発明のタンパク質を含有する細胞に 接触させた場合と、 本発明のタンパク質を活性化する化合物および試験化合物を 本発明のタンパク質を含有する細胞に接触させた場合における、 本発明のタンパ ク質を介した細胞刺激活性を測定し、 比較する。 本発明のタンパク質を活性化す る化合物による細胞刺激活性を減少させ得る化合物またはその塩は本発明のタン パク質のアン夕ゴニストである。 (b) when a compound that activates the protein of the present invention (for example, a peptide of the present invention or an agonist of the protein of the present invention) is brought into contact with a cell containing the protein of the present invention; The cell stimulating activity mediated by the protein of the present invention when the compound to be converted and the test compound are brought into contact with the cells containing the protein of the present invention is measured and compared. The compound capable of decreasing the cell stimulating activity of the compound activating the protein of the present invention or a salt thereof is an antagonist of the protein of the present invention.
該本発明のタンパク質のァゴニストは、 本発明のタンパク質に対する本発明の ぺプチドが有する生理活性と同様の作用を有しているので、 本発明のぺプチドと 同様に安全で低毒性な医薬として有用である。  Since the agonist of the protein of the present invention has the same activity as the physiological activity of the peptide of the present invention on the protein of the present invention, it is useful as a safe and low-toxic drug like the peptide of the present invention. It is.
逆に、 本発明のタンパク質アン夕ゴニストは、 本発明のタンパク質に対する 本発明のぺプチドが有する生理活性を抑制することができるので、 該レセプター 活性を抑制する安全で低毒性な医薬とじて有用である。  Conversely, the protein antagonist of the present invention can suppress the physiological activity of the peptide of the present invention on the protein of the present invention, and is therefore useful as a safe and low-toxic drug for suppressing the receptor activity. is there.
本発明のタンパク質は食欲 (摂食) 増進作用およびォキシトシン分泌促進作用 などに関与している。 よって、 上記のスクリーニング方法またはスクリーニング 用キットを用いて得られる化合物のうち、 本発明のタンパク質のァゴニストは、 例えば、 食欲 (摂食) 増進剤、 食欲不振 (例、 神経性食欲不振症など) 、 食欲不 振に伴う貧血または低蛋白症、 微弱陣痛、 弛緩出血、 子宮復古不全、 乳汁うっ滞 などの予防 ·治療剤などの医薬として用いることができる。 また、 本発明のタン パク質のアンタゴニストは、 例えば、 肥満症 〔例、 悪性肥満細胞症 (malignant mastocytosis), 外因性肥満 (exogenous obesity), 過インシュリン性肥満症 (hyper insul inar obesity)、 過血漿性肥滴 (hyperplasmic obesity)^ 下垂体性 肥満 (hypophyseal adiposity), 減血漿性肥満症 (hypoplasmic obesity)> 甲状 腺機能低下肥満症 (hypothyroid obesity), 視床卞部性肥満 (hypothalamic . obesity)、 症候性肥満症 (sy即 tomatic obesity), 小児肥満 (infantile obesity), 上半身肥満 (upper body obesity), 食事性肥満症 (alimentary obesity), 性機能低下性肥満 (hypogonadal obesity), 全身性肥満細胞症 The protein of the present invention is involved in an appetite (feeding) promoting action and an oxytocin secretion promoting action. Therefore, among the compounds obtained using the above-mentioned screening method or screening kit, agonists of the protein of the present invention include, for example, an appetite (feeding) enhancer, anorexia (eg, anorexia nervosa), Appetite It can be used as a medicament for the prevention and treatment of anemia or hypoproteinemia due to tremor, weak labor, laxative bleeding, uterine remodeling failure, and milk stasis. The protein antagonists of the present invention include, for example, obesity (eg, malignant mastocytosis), exogenous obesity, hyperinsulin inar obesity, hyperplasma Hyperplasmic obesity ^ hypophyseal adiposity, hypoplasmic obesity> hypothyroid obesity, hypothalamic obesity, hypothalamic obesity, symptoms Obesity (sy immediate tomatic obesity), childhood obesity (infantile obesity), upper body obesity, alimentary obesity, hypogonadal obesity, systemic mastocytosis
(systemic mastocytosis)、 単純性肥満 (simple obesity), 中心性肥満  (systemic mastocytosis), simple obesity, central obesity
(central obesity)など〕 、 摂食亢進症、 情動障害、 性機能障害、 過強陣痛、 強 直性子宮収縮、 胎児仮死、 子宮破裂、 類管裂傷、 早産、 Prader-Willi症候群、 糖 尿病およびその合併症 (例、 糖尿病性腎症、 糖尿病性網膜症、 糖尿病性神経障害 など) 、 高血圧、 高脂血症、 冠状動脈硬化症、 痛風、 呼吸器疾患 (Pickwick症候 群、 睡眠時無呼吸症候群) 、 脂肪肝、 不妊症、 変形性骨関節症など (特に抗肥満 剤、 食欲 (摂食) 調節剤など) などの予防 ·治療剤などの医薬として用いること ができる。  (central obesity)), hyperphagia, affective disorders, sexual dysfunction, overwork, tonic contractions, fetal asphyxia, uterine rupture, tubal lacerations, preterm birth, Prader-Willi syndrome, glucoseuria and Its complications (eg, diabetic nephropathy, diabetic retinopathy, diabetic neuropathy, etc.), hypertension, hyperlipidemia, coronary atherosclerosis, gout, respiratory disease (Pickwick syndrome, sleep apnea syndrome) It can be used as a medicine for prophylactic and therapeutic agents such as fatty liver, infertility, osteoarthritis, etc. (especially antiobesity agents, appetite (feeding) regulators, etc.).
上記のスクリーニング方法またはスクリーニング用キットを用いて得られる化 合物の塩としては、 例えば、 薬学的に許容可能な塩などが用いられる。 例えば、 無機塩基との塩、 有機塩基との塩、 無機酸との塩、 有機酸との塩、 塩基性または 酸性ァミノ酸との塩などがあげられる。  As a salt of the compound obtained by using the above-described screening method or screening kit, for example, a pharmaceutically acceptable salt or the like is used. Examples thereof include salts with inorganic bases, salts with organic bases, salts with inorganic acids, salts with organic acids, and salts with basic or acidic amino acids.
無機塩基との塩の好適な例としては、 例えばナトリウム塩、 カリウム塩などの アルカリ金属塩、 カルシウム塩、 マグネシウム塩などのアルカリ土類金属塩、 な らびにアルミニウム塩、 アンモニゥム塩などがあげられる。  Preferable examples of the salt with an inorganic base include an alkali metal salt such as a sodium salt and a potassium salt, an alkaline earth metal salt such as a calcium salt and a magnesium salt, and an aluminum salt and an ammonium salt.
有機塩基との塩の好適な例としては、 例えばトリメチルァミン、 トリェチルァ ミン、 ピリジン、 ピコリン、 2, 6ールチジン、 エタノールァミン、 ジェ夕ノー ルァミン、 卜リエタノールァミン、 シクロへキシルァミン、 ジシクロへキシルァ ミン、 N, N, 一ジべンジルェチレンジァミンなどとの塩などがあげられる。 無機酸との塩の好適な例としては、 例えば塩酸、 臭化水素酸、 硫酸、 リン酸な どとの塩があげられる。 Preferred examples of the salt with an organic base include, for example, trimethylamine, triethylamine, pyridine, picoline, 2,6-alutidine, ethanolamine, genoaluminamine, triethanolamine, cyclohexylamine, dicyclohexane. Examples include salts with xylamine, N, N, and dibenzylethylenediamine. Preferred examples of the salt with an inorganic acid include salts with hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, and the like.
有機酸との塩の好適な例としては、 例えばギ酸、 酢酸、 プロピオン酸、 フマル 酸、 シユウ酸、 酒石酸、 マレイン酸、 クェン酸、 コハク酸、 リンゴ酸、 メタンス ルホン酸、 ベンゼンスルホン酸、 安息香酸などとの塩があげられる。  Suitable examples of salts with organic acids include, for example, formic acid, acetic acid, propionic acid, fumaric acid, oxalic acid, tartaric acid, maleic acid, citric acid, succinic acid, malic acid, methanesulfonic acid, benzenesulfonic acid, benzoic acid And the like.
塩基性アミノ酸との塩の好適な例としては、 例えばアルギニン、 リジン、 オル チニシなどとの塩があげられ、'酸性アミノ酸との好適な例としては、 例えばァス パラギン酸、 グルタミン酸などとの塩があげられる。  Preferable examples of the salt with a basic amino acid include salts with, for example, arginine, lysine, orcinus, and preferable examples of the salt with an acidic amino acid include, for example, salts with aspartic acid, glutamic acid, etc. Is raised.
本発明のスクリーニング方法またはスクリーニング用キットを用いて得られる 化合物またはその塩を上述の医薬として使用する場合、 上記の本発明のペプチド を医薬として実施する場合と同様にして実施することができる。  When a compound or a salt thereof obtained by using the screening method or the screening kit of the present invention is used as the above-mentioned medicament, it can be carried out in the same manner as when the above-mentioned peptide of the present invention is used as a medicament.
本発明のスクリーニング方法またはスクリーニング用キットを用いて得られる 化合物またはその塩を上述の医薬として使用する場合、 常套手段に従って実施す ることができる。 例えば、 必要に応じて糖衣や腸溶性被膜を施した錠剤、 カプセ ル剤、 エリキシル剤、 マイクロカプセル剤などとして経口的に、 あるいは水もし くはそれ以外の薬学的に許容し得る液との無菌性溶液、 または 濁液剤などの注 射剤の形で非経口的に使用できる。 例えば、 該化合物またはその塩を生理学的に 認められる担体、 香味剤、 賦形剤、 べヒクル、 防腐剤、 安定剤、 結合剤などとと もに一般に認められた単位用量形態で混和することによって製造することができ る。 これら製剤における有効成分量は指示された範囲の適当な用量が得られるよ うにするものである。  When a compound or a salt thereof obtained by using the screening method or the screening kit of the present invention is used as the above-mentioned medicine, it can be carried out according to a conventional method. For example, sterile orally with tablets or capsules, capsules, elixirs, microcapsules, etc., if necessary, coated with sugar or enteric coating, or with water or other pharmaceutically acceptable liquids It can be used parenterally in the form of injections, such as aqueous solutions or suspensions. For example, by mixing the compound or a salt thereof with a physiologically acceptable carrier, flavoring agent, excipient, vehicle, preservative, stabilizer, binder and the like in a generally accepted unit dosage form. Can be manufactured. The amount of the active ingredient in these preparations is such that a suitable dosage in the specified range can be obtained.
錠剤、 カプセル剤などに混和することができる添加剤としては、 例えばゼラチ ン、 コーンスターチ、 トラガントガム、 アラビアゴムのような結合剤、 結晶性セ ルロースのような賦形剤、 コーンスターチ、 ゼラチン、 アルギン酸などのような 膨化剤、 ステアリン酸マグネシウムのような潤滑剤、 ショ糖、 乳糖またはサッカ リンのような甘味剤、 ぺパ一ミント、 ァカモノ油またはチェリ一のような香味剤 などが用いられる。 調剤単位形態がカプセルである場合には、 前記タイプの材料 にさらに油脂のような液状担体を含有することができる。 注射のための無菌組成 物は注射用水のようなべヒクル中の活性物質、 胡麻油、 椰子油などのような天然 産出植物油などを溶解または懸濁させるなどの通常の製剤実施にしたがって処方 することができる。 Additives that can be incorporated into tablets, capsules, etc. include, for example, binders such as gelatin, corn starch, tragacanth gum, gum arabic, excipients such as crystalline cellulose, corn starch, gelatin, alginic acid, etc. Such a leavening agent, a lubricant such as magnesium stearate, a sweetening agent such as sucrose, lactose or saccharin, and a flavoring agent such as peppermint, cocoa oil or cellulose are used. When the preparation unit form is a capsule, a liquid carrier such as oils and fats can be further contained in the above-mentioned type of material. Sterile compositions for injection include active substances in vehicles such as water for injection, natural substances such as sesame oil, coconut oil and the like. It can be formulated according to the usual formulation practice such as dissolving or suspending the produced vegetable oil.
注射用の水性液としては、 例えば、 生理食塩水、 ブドウ糖やその他の補助薬を 含む等張液 (例えば、 D—ソルビトール、 D—マンニトール、 塩化ナトリウムな ど) などがあげられ、 適当な溶解補助剤、 たとえばアルコール (たとえばェタノ ール) 、 ポリアルコール (たとえばプロピレングリコール、 ポリエチレングリコ ール) 、 非ィォン性界面活性剤 (たとえばポリソルベート 8 0™、 H C O - 5 0 ) などと併用してもよい。 油性液としてはゴマ油、 大豆油などがあげられ、 溶 解補助剤として安息香酸ベンジル、 ベンジルアルコ一ルなどと併用してもよい。 また、 緩衝剤 (例えば、 リン酸塩緩衝液、 酢酸ナトリウム緩衝液) 、 無痛化剤 Aqueous liquids for injection include, for example, physiological saline, isotonic solutions containing glucose and other adjuvants (eg, D-sorbitol, D-mannitol, sodium chloride, etc.). Agents such as alcohols (eg, ethanol), polyalcohols (eg, propylene glycol, polyethylene glycol), nonionic surfactants (eg, Polysorbate 80 ™, HCO-50) and the like may be used in combination. Examples of the oily liquid include sesame oil and soybean oil, which may be used in combination with a dissolution aid such as benzyl benzoate or benzyl alcohol. Also, buffers (eg, phosphate buffer, sodium acetate buffer), soothing agents
(例えば、 塩ィ匕ベンザルコニゥム、 塩酸プロ力インなど) 、 安定剤 (例えば、 ヒ ト血清アルブミン、 ポリエチレングリコールなど) 、 保存剤 (例えば.、 ベンジル アルコール、 フエノ一ルなど) 、 酸化防止剤などと配合してもよい。 調製された 注射液は通常、 適当なアンプルに充填される。 (Eg, Shirazidani Benzalkonium, Proforce hydrochloride, etc.), stabilizers (eg, human serum albumin, polyethylene glycol, etc.), preservatives (eg, benzyl alcohol, phenol, etc.), antioxidants, etc. You may mix. The prepared injection solution is usually filled in a suitable ampoule.
このようにして得られる製剤は安全で低毒性であるので、 例えば哺乳動物 (例 えば、 ヒト、 マウス、 ラット、 モルモット、 ゥサギ、 ヒッジ、 ブ夕、 ゥシ、 ネコ、 ィヌ、 サル、 チンパンジーなど) に対して投与することができる。  The preparations obtained in this way are safe and low toxic, and can be used, for example, in mammals (eg, humans, mice, rats, guinea pigs, egrets, sheep, sheep, bushus, dogs, cats, dogs, monkeys, chimpanzees, etc. ) Can be administered.
本発明のスクリーニング方法またはスクリーニング用キットを用いて得られる 化合物またはその塩の投与量は、 症状などにより差異はあるが、 経口投与の場合、 一般的に成人 (体重 60kgとして) においては、 一日につき約 0. l〜1000mg、 好ま しくは約 1. 0〜300mg、 より好ましくは約 3. 0〜50mgである。 非経口的に投与する 場合は、 その 1回投与量は投与対象、 対象臓器、 症状、 投与方法などによっても 異なるが、 たとえば注射剤の形では成人の肥満症患者 (体重 60kgとして) への投 与においては、 アンタゴニストを一日につき約 0. 01〜30mg、 好ましくは約 0. 1〜 20mg、 より好ましくは約 0. 1〜10mgを静脈注射により投与するのが好都合である。 他の動物の場合も、 60kg当たりに換算した量を投与することができる。  The dose of a compound or a salt thereof obtained by using the screening method or the screening kit of the present invention varies depending on symptoms and the like. About 0.1 to 1000 mg, preferably about 1.0 to 300 mg, more preferably about 3.0 to 50 mg. In the case of parenteral administration, the single dose varies depending on the administration subject, target organ, symptoms, administration method, etc. For example, in the case of injection, injection into adult obese patients (with a body weight of 60 kg) is required. In administration, it may be convenient to administer the antagonist by intravenous injection at about 0.01 to 30 mg, preferably about 0.1 to 20 mg, more preferably about 0.1 to 10 mg per day. In the case of other animals, the dose can be administered in terms of 60 kg.
〔3〕 本発明のタンパク質、 その部分ペプチドまたはその塩の定量 [3] Quantification of the protein of the present invention, its partial peptide or its salt
本発明の抗体は、 本発明のタンパク質を特異的に認識することができるので、 被検液中の本発明のタンパク質の定量、 特にサンドイッチ免疫測定法による定量 などに使用することができる。 Since the antibody of the present invention can specifically recognize the protein of the present invention, It can be used for quantification of the protein of the present invention in a test solution, particularly for quantification by sandwich immunoassay.
すなわち、 本発明は、  That is, the present invention
' (i) 本発明の抗体と、 被検液および標識化された本発明のタンパク質とを競合 的に反応させ、 該抗体に結合した標識化された本発明のタンパク質の割合を測定 することを特徴とする被検液中の本発明のタンパク質の定量法、 および (i) reacting the antibody of the present invention with a test solution and the labeled protein of the present invention competitively, and measuring the ratio of the labeled protein of the present invention bound to the antibody. A method for quantifying the protein of the present invention in a test solution, and
( i i ) 被検液と担体上に不溶化した本発明の抗体および標識化された本発明の別 の抗体とを同時あるいは連続的に反応させたのち、 不溶化担体上の標識剤の活性 を測定することを特徴とする被検液中の本発明のタンパク質の定量法を提供する, 上記 (i i) の定量法においては、 一方の抗体が本発明のタンパク質の N端部を 認識する抗体で、 他方の抗体が本発明のタンパク質の C端部に反応する抗体であ ることが望ましい。  (ii) After reacting the test liquid with the antibody of the present invention insolubilized on the carrier and another labeled antibody of the present invention simultaneously or continuously, the activity of the labeling agent on the insolubilized carrier is measured. The present invention provides a method for quantifying the protein of the present invention in a test solution, characterized in that in the quantification method of the above (ii), one of the antibodies is an antibody that recognizes the N-terminal of the protein of the present invention, and the other is It is desirable that the above-mentioned antibody reacts with the C-terminal of the protein of the present invention.
また、 本発明のタンパク質に対するモノクローナル抗体 (以下、 本発明のモノ クローナル抗体と称する場合がある) を用いて本発明のタンパク質の定量を行な えるほか、 組織染色等による検出を行なうこともできる。 これらの目的には、 抗, 体分子そのものを用いてもよく、 また、 抗体分子の F (ab,)2 、 Fab'または Fab画分 を用いてもよい。 In addition, the protein of the present invention can be quantified using a monoclonal antibody against the protein of the present invention (hereinafter sometimes referred to as the monoclonal antibody of the present invention), and can also be detected by tissue staining or the like. For these purposes, the anti-body molecule itself may be used, or the F (ab,) 2 , Fab ′ or Fab fraction of the antibody molecule may be used.
本発明の抗体を用いる本発明のタンパク質の定量法は、 特に制限されるべきも のではなく、 被測定液中の抗原量 (例えば、 タンパク質量) に対応した抗体、 抗 原もしくは抗体一抗原複合体の量を化学的または物理的手段により検出し、 これ を既知量の抗原を含む標準液を用いて作製した標準曲線より算出する測定法であ れば、 いずれの測定法を用いてもよい。 例えば、 ネフロメトリ一、 競合法、 ィム ノメトリック法およびサンドイッチ法が好適に用いられるが、 感度、 特異性の点 で、 後述するサンドィツチ法を用いるのが特に好ましい。  The method for quantifying the protein of the present invention using the antibody of the present invention is not particularly limited, and may be an antibody, an antigen, or an antibody-antigen complex corresponding to the amount of antigen (eg, the amount of protein) in the test solution. Any measurement method may be used as long as the amount of the body is detected by chemical or physical means, and this is calculated from a standard curve prepared using a standard solution containing a known amount of antigen. . For example, nephrometry, competition method, immunometric method and sandwich method are preferably used, but it is particularly preferable to use the sandwich method described later in terms of sensitivity and specificity.
標識物質を用いる測定法に用いられる標識剤としては、 例えば、 放射性同位元 素 (例、 〔1251〕 、 〔1311〕 、 〔¾〕 、 〔'4C〕 、 〔32P〕 、 〔33P〕 、 〔35s〕 など) 、 蛍光物質 〔例、 シァニン蛍光色素 (例、 Cy2、 Cy3、 Cy5、 Cy5. 5、 Cy7 (アマシャ ムバイオサイエンス社製) など) 、 フルォレスカミン、 フルォレツセンイソチォ シァネートなど〕 、 酵素 (例、 —ガラクトシダ一ゼ、 )3—ダルコシダ一ゼ、 ァ ルカリフォスファタ一ゼ、 パ一ォキシダ一ゼ、 リンゴ酸脱水素酵素など) 、 発光 物質 (例、 ルミノール、 ルミノール誘導体、 ルシフェリン、 ルシゲニンなど) 、 ピオチン、 ランタニド元素などが用いられる。 さらに、 抗体あるいは抗原と標識 剤との結合にピオチン一アビジン系を用いることもできる。 Examples of the labeling agent used in the assay method using the labeling substance, for example, radioactive isotopes elemental (eg, [125 1], [131 1], [¾], [ '4 C], [32 P], [33 P], [ 35s ], etc.), fluorescent substances (eg, cyanine fluorescent dyes (eg, Cy2, Cy3, Cy5, Cy5.5, Cy7 (manufactured by Amersham Biosciences), etc.), fluorescamine, fluorescenisochi Etc.), enzymes (eg, -galactosidase,) 3-darcosidase, Lucariphosphatase, passoxidase, malate dehydrogenase, etc., luminescent substances (eg, luminol, luminol derivatives, luciferin, lucigenin, etc.), piotin, lanthanide elements, etc. are used. Further, a biotin-avidin system can be used for binding the antibody or antigen to the labeling agent.
抗原あるいは抗体の不溶化に当っては、 物理吸着を用いてもよく、 また通常夕 ンパク質あるいは酵素等を不溶化、 固定化するのに用いられる化学結合を用いる 方法でもよい。 担体としては、 ァガロース、 デキストラン、 セルロースなどの不 溶性多糖類、 ポリスチレン、 ポリアクリルアミド、 シリコン等の合成樹脂、 ある いはガラス等が挙げられる。  For the insolubilization of the antigen or antibody, physical adsorption may be used, or a method using a chemical bond usually used for insolubilizing and immobilizing proteins or enzymes may be used. Examples of the carrier include insoluble polysaccharides such as agarose, dextran, and cellulose; synthetic resins such as polystyrene, polyacrylamide, and silicon; and glass.
サンドィツチ法においては不溶化した本発明のモノクローナル抗体に被検液を 反応させ (1次反応) 、 さらに標識化した別の本発明のモノクローナル抗体を反 応させ (2次反応) たのち、 不溶化担体上の標識剤の活性を測定することにより 被検液中の本発明のタンパク質量を定量することができる。 1次反応と 2次反応 は逆の順序に行っても、 また、 同時に行なってもよいし時間をずらして行なって もよい。 標識化剤および不溶化の方法は前記のそれらに準じることができる。 ま た、 サンドイッチ法による免疫測定法において、 固相用抗体あるいは標識用抗体 に用いられる抗体は必ずしも 1種類である必要はなく、 測定感度を向上させる等 の目的で 2種類以上の抗体の混合物を用いてもよい。  In the sandwich method, a test solution is reacted with the insolubilized monoclonal antibody of the present invention (primary reaction), and further reacted with another labeled monoclonal antibody of the present invention (secondary reaction). By measuring the activity of the labeling agent, the amount of the protein of the present invention in the test solution can be determined. The primary reaction and the secondary reaction may be performed in the reverse order, may be performed simultaneously, or may be performed at staggered times. The labeling agent and the method of insolubilization can be in accordance with those described above. Also, in the immunoassay by the sandwich method, the antibody used for the solid phase antibody or the labeling antibody does not necessarily need to be one kind, and a mixture of two or more kinds of antibodies is used for the purpose of improving measurement sensitivity and the like. May be used.
本発明のサンドィツチ法による本発明の夕ンパク質の測定法においては、 1次 反応と 2次反応に用いられる本発明のモノクローナル抗体は、 本発明のタンパク 質の結合する部位が相異なる抗体が好ましく用いられる。 すなわち、 1次反応お よび 2次反応に用いられる抗体は、 例えば、 2次反応で用いられる抗体が、 本発 明のタンパク質の C端部を認識する場合、 1次反応で用いられる抗体は、 好まし くは C端部以外、 例えば N端部を認識する抗体が用いられる。  In the method for measuring the protein of the present invention by the sandwich method of the present invention, the monoclonal antibody of the present invention used in the primary reaction and the secondary reaction is preferably an antibody having a different site to which the protein of the present invention binds. Used. That is, the antibody used in the primary reaction and the secondary reaction is, for example, when the antibody used in the secondary reaction recognizes the C-terminal of the protein of the present invention, the antibody used in the primary reaction is Preferably, an antibody that recognizes other than the C-terminal, for example, the N-terminal, is used.
本発明のモノクローナル抗体をサンドイッチ法以外の測定システム、 例えば、 競合法、 ィムノメトリック法あるいはネフロメトリーなどに用いることができる ( 競合法では、 被検液中の抗原と標識抗原とを抗体に対して競合的に反応させた のち、 未反応の標識抗原(F) と、 抗体と結合した標識抗原 (B) とを分離し (BZF分離) 、 B, Fいずれかの標識量を測定し、 被検液中の抗原量を定量す る。 本反応法には、 抗体として可溶性抗体を用い、 B /F分離をポリエチレング リコール、 前記抗体に対する第 2抗体などを用いる液相法、 および、 第 1抗体と して固相化抗体を用いるか、 あるいは、 第 1抗体は可溶性のものを用い第 2抗体 として固相化抗体を用いる固相化法とが用いられる。 The monoclonal antibody of the present invention can be used in a measurement system other than the sandwich method, for example, a competition method, an immunometric method or a nephelometry. (In the competition method, the antigen in the test solution and the labeled antigen are used for the antibody. After reacting competitively, the unreacted labeled antigen (F) and the labeled antigen (B) bound to the antibody are separated (BZF separation), and the amount of labeling of either B or F is measured. Quantify the amount of antigen in the test solution You. In this reaction method, a soluble antibody was used as the antibody, B / F separation was performed using polyethylene glycol, a liquid phase method using a second antibody against the antibody, and a solid phase antibody was used as the first antibody. Alternatively, an immobilization method using a soluble first antibody and an immobilized antibody as the second antibody is used.
ィムノメトリック法では、 被検液中の抗原と固相化抗原とを一定量の標識化抗体 に対して競合反応させた後固相と液相を分離するか、 あるいは、 被検液中の抗原 と過剰量の標識化抗体とを反応させ、 次に固相化抗原を加え未反応の標識化抗体 を固相に結合させたのち、 固相と液相を分離する。 次に、 いずれかの相の標識量 を測定し被検液中の抗原量を定量する。 In the immunometric method, the antigen in the test solution and the immobilized antigen are subjected to a competitive reaction with a certain amount of labeled antibody, and then the solid phase and the liquid phase are separated. The antigen is allowed to react with an excess amount of the labeled antibody, then the immobilized antigen is added, and the unreacted labeled antibody is bound to the solid phase, and then the solid phase and the liquid phase are separated. Next, the amount of label in either phase is measured to determine the amount of antigen in the test solution.
また、 ネフロメトリ一では、 ゲル内あるいは溶液中で抗原抗体反応の結果生じ た不溶性の沈降物の量を測定する。 被検液中の抗原量が僅かであり、 少量の沈降 物しか得られない場合にもレーザ一の散 ¾^を利用するレーザ一ネフロメトリーな どが好適に用いられる。  In nephrometry, the amount of insoluble sediment resulting from an antigen-antibody reaction in a gel or in a solution is measured. Even when the amount of antigen in the test solution is small and only a small amount of sediment is obtained, laser-nephrometry using laser-dispersion is preferably used.
これら個々の免疫学的測定法を本発明の定量方法に適用するにあたっては、 特 別の条件、 操作等の設定は必要とされない。 それぞれの方法における通常の条件、 操作法に当業者の通常の技術的配慮を加えて本発明のタンパク質の測定系を構築 すればよい。 これらの一般的な技術手段の詳細については、 総説、 成書などを参 照することができる。  In applying these individual immunological measurement methods to the quantification method of the present invention, no special conditions, operations, and the like need to be set. The protein measuring system of the present invention may be constructed by adding ordinary technical considerations of those skilled in the art to ordinary conditions and operation methods in each method. For details of these general technical means, reference can be made to reviews, documents, etc.
例えば、 入江 寛編 「ラジオィムノアツセィ」 (講談社、 昭和 4 9年発行) 、 入江 寛編 「続ラジオィムノアツセィ」 (講談社、 昭和 5 4年発行) 、 石川栄治 ら編 「酵素免疫測定法」 (医学書院、 昭和 5 3年発行) 、 石川栄治ら編 「酵素免 疫測定法」 (第 2版) (医学書院、 昭和 5 7年発行) 、 石川栄治ら編 「酵素免疫 測定法」 (第 3版) (医学書院、 昭和 6 2年発行) 、 「Methods in  For example, edited by Hiro Irie, "Radio Nonotsusei" (Kodansha, published in Showa 49), edited by Hiroshi Irie, "Continued Radio Immnoatsusy" (Kodansha, published in 1954), Eiji Ishikawa et al. "Measurement Method" (Medical Shoin, published in 1958), Eiishi Ishikawa et al., "Enzyme Immunoassay" (Second Edition) (Medical Publishing, published in 1977), Eiji Ishikawa, et al., "Enzyme Immunoassay" (3rd edition) (Medical Shoin, published in 1962), "Methods in
ENZYMOLOGYJ Vol . 70 (Immunochemi cal Techniques (Par t A) )、 同書 Vol . ENZYMOLOGYJ Vol. 70 (Immunochemi cal Techniques (Part A)), Ibid.
73 (Immunochemical Techniques (Par t B) )、 同書 Vol . 7 (Immunochemical Techniques (Part C) )、 同書 Vol . 84 (Immunochemical Techniques (Part 73 (Immunochemical Techniques (Part B)), ibid.Vol. 7 (Immunochemical Techniques (Part C)), ibid.Vol. 84 (Immunochemical Techniques (Part B)
D: Sel ec ted Immunoassays) )、 同書 Vo l . 92 (Immunochemical Techniques (Part E :Monoc lonal Ant ibodies and General Immunoassay Methods)) , 同書 Vol . D: Selected Immunoassays)), ibid.Vol. 92 (Immunochemical Techniques (Part E: Monoclonal Ant ibodies and General Immunoassay Methods)), ibid.
121 (Immunochemical Techniques (Part I: Hybridoma Technology and Monoclonal An t i b o d i e s ) ) (以上、 アカデミックプレス社発行)などを参照することができる。 以上のようにして、 本発明の抗体を用いることによって、 本発明のタンパク質 を感度良く定量することができる。 121 (Immunochemical Techniques (Part I: Hybridoma Technology and Monoclonal Antibodies))) (published by Academic Press). As described above, the protein of the present invention can be quantified with high sensitivity by using the antibody of the present invention.
さらには、 本発明の抗体を用いて本発明のタンパク質の濃度を定量することに よって、 本発明のタンパク質の濃度の減少が検出された場合、 例えば、 食欲不振 (例、 神経性食欲不振症など) 、 食欲不振に伴う貧血または低蛋白症、 微弱陣痛、 弛緩出血、 子宮復古不全、 乳汁うっ滞などが発症している可能性が高いと診断す ることができる。 反対に、 例えば、 本発明のタンパク質の濃度の上昇が検出され た場合、 例えば、 肥満症 (例、 惠性肥満細胞症、 外因性肥満、 過インシュリン性 肥満症、 過血漿性肥満、 下垂体性肥満、 減血漿性肥満症、 甲状腺機能低下肥満症、 視床下部性肥満、 症候性肥満症、 小児肥満、 上半身肥満、 食事性肥満症、 性機能 低下性肥満、 全身性肥満細胞症、 単純性肥満、 中心性肥満など) 、 摂食亢進症、 情動障害、 性機能障害、 過強陣痛、 強直性子宮収縮、 胎児仮死、 子宮破裂、 顏管 裂傷、 早産、 Prader-Wi l l i症候群、 糖尿病およびその合併症 (例、 糖尿病性腎症、 糖尿病性網膜症、 糖尿病性神経障害など) 、 高血圧、 高脂血症、 冠状動脈硬化症、 痛風、 呼吸器疾患 (Pi ckwick症候群、 睡眠時無呼吸症候群) 、 脂肪肝、 不妊症、 変形性骨関節症などが発症している可能性が高いと診断することが出来る。  Furthermore, when a decrease in the concentration of the protein of the present invention is detected by quantifying the concentration of the protein of the present invention using the antibody of the present invention, for example, anorexia (eg, anorexia nervosa, etc.) Anemia or hypoproteinemia due to anorexia, weak labor, lax hemorrhage, uterine remodeling failure, milk stasis, etc. can be diagnosed as having a high possibility of developing. Conversely, if an increase in the concentration of the protein of the present invention is detected, for example, obesity (eg, vein mastocytosis, exogenous obesity, hyperinsulinic obesity, hyperplasmic obesity, pituitary) Obesity, reduced plasma obesity, hypothyroid obesity, hypothalamic obesity, symptomatic obesity, pediatric obesity, upper body obesity, dietary obesity, hypogonadism, systemic mastocytosis, simple obesity , Central obesity, etc.), hyperphagia, affective disorders, sexual dysfunction, excessive labor, tonic contractions, fetal asphyxia, uterine rupture, facial lacerations, preterm birth, Prader-Willi syndrome, diabetes and its complications Disease (eg, diabetic nephropathy, diabetic retinopathy, diabetic neuropathy, etc.), hypertension, hyperlipidemia, coronary atherosclerosis, gout, respiratory disease (Pickck syndrome, sleep apnea syndrome), Fatty liver, infertility It can be diagnosed that the possibility of osteoarthritis and the like is high.
また、 本発明の抗体は,、 体液や組織などの被検体中に存在する本発明のタンパ ク質を検出するために使用することができる。 また、 本発明のタンパク質を精製 するために使用する抗体カラムの作製、 精製時の各分画中の本発明のタンパク質 の検出、 被検細胞内における本発明のタンパク質の挙動の分析などのために使用 することができる。  Further, the antibody of the present invention can be used for detecting the protein of the present invention present in a subject such as a body fluid or a tissue. In addition, for the preparation of an antibody column used for purifying the protein of the present invention, the detection of the protein of the present invention in each fraction during purification, and the analysis of the behavior of the protein of the present invention in test cells, etc. Can be used.
〔4〕 遺伝子診断剤 ' (4) Gene diagnostics ''
本 明の D NAは、 例えば、 プローブとして使用することにより、 ヒトまたは 温血動物 (例えば、 ラット、 マウス、 モルモット、 ゥサギ、 トリ、 ヒッジ、 ブ夕、 ゥシ、 ゥマ、 ネコ、 ィヌ、 サル、 チンパンジーなど) における本発明のタンパク 質またはその部分べプチドをコードする D N Aまたは m R N Aの異常 (遺伝子異 常) を検出することができるので、 例えば、 該 D NAまたは mR NAの損傷、 突 然変異あるいは発現低下や、 該 D N Aまたは m R N Aの増加あるいは発現過多な どの遺伝子診断剤として有用である。 The DNA of the present invention can be used, for example, in humans or warm-blooded animals (e.g., rats, mice, guinea pigs, egrets, birds, higgies, bushes, horses, cats, cats, dogs, Abnormalities (genetic abnormalities) in the DNA or mRNA encoding the protein of the present invention or a partial peptide thereof in monkeys, chimpanzees, etc.). It is useful as a gene diagnostic agent for mutation or decrease in expression, and increase or excessive expression of the DNA or mRNA.
本発明の D N Aを用いる上記の遺伝子診断は、 例えば、 公知のノーザンハイブ リダィゼーションゃ P C R— S S C P法 (Genomics, 第 5卷, 874〜879頁(1989年) Proceedings of the Nat ional Academy of Sciences of the Uni ted States of America, 第 86巻, 2766〜2770頁(1989年)) などにより実施することができる。 例えば、 ノーザンハイプリダイゼーシヨンにより発現増加が検出された場合、 例えば、 肥満症 (例、 悪性肥満細胞症、 外因性肥満、 過インシュリン性肥満症、 過血漿性肥満、 下垂体性肥満、 減血漿性肥満症、 甲状腺機能低下肥満症、 視床下 部性肥満、 症候性肥満症、 小児肥満、 上半身肥満、 食事性肥満症、 性機能低下性 肥満、 全身性肥満細胞症、 単純性肥満、 中心性肥満など) 、 摂食亢進症、 情動障 害、 性機能障害、 過強陣痛、 強直性子宮収縮、 胎児仮死、 子宮破裂、 類管裂傷、 早産、 Prader- Wi l l i症候群、 糖尿病およびその合併症 (例、 糖尿病性腎症、 糖尿 病性網膜症、 糖尿病性神経障害など) 、 高血圧、 高脂血症、 冠状動脈硬化症、 痛 風、 呼吸器疾患 (Pickwick症候群、 睡眠時無呼吸症候群) 、 脂肪肝、 不妊症、 変 形性骨関節症などである可能性が高いと診断することが出来る。 反対に、 発現低 下が検出された場合や P C R— S S C P法により D NAの突然変異が検出された 場合は、 例えば、 食欲不振 (例、 神経性食欲不振症など) 、 食欲不振に伴う貧血 または低蛋白症、 微弱陣痛、 弛緩出血、 子宮復古不全、 乳汁うっ滞などである可 能性が高いと診断することができる。  The above-described genetic diagnosis using the DNA of the present invention can be performed, for example, by the well-known Northern Hybridization ゃ PCR-SSCP method (Genomics, Vol. 5, pp. 874-879 (1989)) Proceedings of the National Academy of Sciences of The United States of America, Vol. 86, pp. 2766-2770 (1989)). For example, when an increase in expression is detected by Northern hybridization, for example, obesity (eg, malignant mastocytosis, exogenous obesity, hyperinsulinic obesity, hyperplasmic obesity, pituitary obesity, hypoplasma) Obesity, hypothyroid obesity, hypothalamic obesity, symptomatic obesity, childhood obesity, upper body obesity, dietary obesity, hypogonadism obesity, systemic mastocytosis, simple obesity, centrality Obesity, etc.), hyperphagia, affective disorders, sexual dysfunction, excessive labor, tonic contractions, fetal asphyxia, uterine rupture, tubal lacerations, preterm birth, Prader-Willi syndrome, diabetes and its complications ( Eg, diabetic nephropathy, diabetic retinopathy, diabetic neuropathy), hypertension, hyperlipidemia, coronary atherosclerosis, gout, respiratory disease (Pickwick syndrome, sleep apnea syndrome), fat Liver, It can be diagnosed that the possibility of infertility or osteoarthritis is high. Conversely, if down-regulation is detected or if a DNA mutation is detected by PCR-SSCP, for example, anorexia (eg, anorexia nervosa), anorexia associated with anorexia or It can be diagnosed as likely to be low protein, weak labor, lax hemorrhage, uterine remodeling failure, and milk stasis.
〔5〕 アンチセンスポリヌクレオチドを含有する医薬 [5] a drug containing an antisense polynucleotide
本発明の D N Aに相補的に結合し、 該 D N Aの発現を抑制することができる本 発明のアンチセンスポリヌクレオチドは低毒性であり、 生体内における本発明の タンパク質または本発明の D N Aの機能 (例、 摂食 (食欲) 増進活性) を抑制す ることができるので、 例えば、 肥満症 (例、 悪性肥満細胞症、 外因性肥満、 過ィ ンシュリン性肥満症、 過血漿性肥満、 下垂体性肥満、 減血漿性肥満症、 甲状腺機 能低下肥満症、 視床下部性肥満、 症候性肥満症、 小児肥満、 上半身肥満、 食事性 肥満症、 性機能低下性肥満、 全身性肥満細胞症、 単純性肥満、 中心性肥満など) 、 摂食亢進症、 情動障害、 性機能障害、 過強陣痛、 強直性子宮収縮、 胎児仮死、 子 宮破裂、 顏管裂傷、 早産、 Prader-Wi l l i症候群、 糖尿病およびその合併症 (例、 糖尿病性腎症、 糖尿病性網膜症、 糖尿病性神経障害など) 、 高血圧、 高脂血症、 冠状動脈硬化症、 痛風、 呼吸器疾患 (Pickwick症候群; 睡眠時無呼吸症候群) 、 脂肪肝、 不妊症、 変形性骨関節症など (特に抗肥満剤、 食欲 (摂食) 調節剤な ど) などの予防 ·治療剤などとして使用することができる。 The antisense polynucleotide of the present invention, which complementarily binds to the DNA of the present invention and can suppress the expression of the DNA, has low toxicity, and functions of the protein of the present invention or the DNA of the present invention in vivo (eg, Since it can suppress eating (appetite) promoting activity, for example, obesity (eg, malignant mastocytosis, exogenous obesity, hyperinsulinic obesity, hyperplasmic obesity, pituitary obesity) , Hypoplasmic obesity, hypothyroid obesity, hypothalamic obesity, symptomatic obesity, childhood obesity, upper body obesity, dietary obesity, hypogonadism, systemic mastocytosis, simple obesity , Central obesity), Hyperphagia, affective disorders, sexual dysfunction, overwork labor, tonic contractions, fetal asphyxia, ruptured pupils, facial tears, premature birth, Prader-Willi syndrome, diabetes and its complications (eg, diabetic Nephropathy, diabetic retinopathy, diabetic neuropathy, etc.), hypertension, hyperlipidemia, coronary atherosclerosis, gout, respiratory disease (Pickwick syndrome; sleep apnea syndrome), fatty liver, infertility, deformity It can be used as a prophylactic / therapeutic agent for osteoarthritis (especially antiobesity agents, appetite (feeding) regulators, etc.).
上記アンチセンスポリヌクレオチドを上記の予防 ·治療剤として使用する場合、 公知の方法に従つて製剤化し、 投与することができる。  When the above-mentioned antisense polynucleotide is used as the above-mentioned prophylactic / therapeutic agent, it can be formulated and administered according to a known method.
例えば、 該ァンチセンスボリヌクレオチドを用いる場合、 該アンチセンスポリ ヌクレオチドを単独あるいはレトロウイルスベクター、 アデノウイルスベクタ一、 アデノウイルスァソシエーテッドウィルスベクターなどの適当なベクターに挿入 した後、 常套手段に従って、 ヒトまたは哺乳動物 (例、 ラット、 ゥサギ、 ヒッジ、 ブタ、 ゥシ、 ネコ、 ィヌ、 サルなど) に対して経口的または非経口的に投与する ことができる。 該アンチセンスポリヌクレオチドは、 そのままで、 あるいは摂取 促進のために補助剤などの生理学的に認められる担体とともに製剤化し、 遺伝子 銃やハイドロゲルカテーテルのようなカテーテルによって投与できる。  For example, when the antisense polynucleotide is used, the antisense polynucleotide is inserted alone or into a suitable vector such as a retrovirus vector, an adenovirus vector, an adenovirus associated virus vector, and the like. It can be administered orally or parenterally to humans or mammals (eg, rats, puppies, sheep, pigs, puppies, cats, dogs, monkeys, etc.). The antisense polynucleotide can be administered as it is or in the form of a formulation together with a physiologically acceptable carrier such as an auxiliary agent for promoting uptake, and can be administered by a gene gun or a catheter such as a hydrogel catheter.
該アンチセンスポリヌクレオチドの投与量は、 対象疾患、 投与対象、 投与ル一 トなどにより差異はあるが、 例えば、 肥満症の治療の目的で本発明のアンチセン スポリヌクレオチドを経静脈投与する場合、 一般的に成人 (体重 60kg) において は、 一日につき該アンチセンスポリヌクレオチドを約 0. l〜100mg投与する。  The dosage of the antisense polynucleotide varies depending on the target disease, the administration subject, the administration route, and the like.For example, when the antisense polynucleotide of the present invention is intravenously administered for the purpose of treating obesity, Generally, in an adult (body weight 60 kg), about 0.1 to 100 mg of the antisense polynucleotide is administered per day.
さらに、 該アンチセンスポリヌクレオチドは、 組織や細胞における本発明の D N Aの存在やその発現状況を調べるための診断用オリゴヌクレオチドプローブと して使用することもできる。  Further, the antisense polynucleotide can also be used as a diagnostic oligonucleotide probe for examining the presence or expression of the DNA of the present invention in tissues or cells.
さらに、 本発明は、  Further, the present invention provides
(i) 本発明のタンパク質をコードする R NAの一部とそれに相補的な R NAを 含有する二重鎖 R NA、  (i) a double-stranded RNA containing a part of the RNA encoding the protein of the present invention and an RNA complementary thereto,
(i i) 前記二重鎖 R NAを含有してなる医薬、  (i i) a medicine comprising the double-stranded RNA,
(i i i) 本発明のタンパク質をコードする R NAの一部を含有するリポザィム、 (iii) a lipozyme containing a part of the RNA encoding the protein of the present invention,
(iv) 前記リポザィムを含有してなる医薬、 (v) 前記リポザィムをコードする遺伝子 (D NA) を含有する発現ベクターな ども提供する。 (iv) a medicament comprising the lipozyme, (v) An expression vector or the like containing the gene (DNA) encoding the lipozyme is provided.
上記アンチセンスポリヌクレオチドと同様に、 二重鎖 R NA、 リポザィムなど も、 本発明の D N Aから転写される R N Aを破壌またはその機能を抑制すること ができ、 生体内における本発明のタンパク質またはそれをコードする D NAの機 能を抑制することができるので、 例えば、 肥満症 (例、 悪性肥満細胞症、 外因性 肥満、 過インシュリン性肥満症、 過血漿性肥満、 下垂体性肥満、' 減血漿性肥満症、 甲状腺機能低下肥満症、 視床下部性肥満、 症候性肥満症、 小児肥満、 上半身肥満、 食事性肥満症、 性機能低下性肥満、 全身性肥満細胞症、 単純性肥満、 中心性肥満 など) 、 摂食亢進症、 情動障害、 性機能障害、 過強陣痛、 強直性子宮収縮、 胎児 仮死、 子宮破裂、 類管裂傷、 早産、 Prader-Wi l l i症候群、 糖尿病およびその合併 症 (例、 糖尿病性腎症、 糖尿病性網膜症、 糖尿病性神経障害など) 、 高血圧、 高 脂血症、 冠状動脈硬化症、 痛風、 呼吸器疾患 (Pi ckwick症候群、 睡眠時無呼吸症 候群) 、 脂肪肝、 不妊症、 変形性骨関節症など (特に抗肥満剤、 食欲 (摂食) 調 節剤など) などの予防 ·治療剤などとして使用することができる。  Like the above-mentioned antisense polynucleotide, double-stranded RNA, lipozyme and the like can disrupt RNA transcribed from the DNA of the present invention or suppress the function thereof, and the protein or the same of the present invention in vivo. Can inhibit the function of DNA encoding, for example, obesity (eg, malignant mastocytosis, exogenous obesity, hyperinsulinic obesity, hyperplasmic obesity, pituitary obesity, Plasma obesity, hypothyroid obesity, hypothalamic obesity, symptomatic obesity, pediatric obesity, upper body obesity, dietary obesity, hypogonadism, systemic mastocytosis, simple obesity, centrality Obesity, etc.), hyperphagia, affective disorders, sexual dysfunction, overwork labor, tonic contractions, fetal asphyxia, ruptured uterus, tubal lacerations, preterm birth, Prader-Willi syndrome, diabetes and its complications (Eg, diabetic nephropathy, diabetic retinopathy, diabetic neuropathy, etc.), hypertension, hyperlipidemia, coronary arteriosclerosis, gout, respiratory disease (Pickwick syndrome, sleep apnea syndrome group) ), Fatty liver, infertility, osteoarthritis, etc. (especially anti-obesity agents, appetite (feeding) regulating agents, etc.) can be used as a preventive and therapeutic agent.
二重鎖 R N Aは、 公知の方法 (例、 Nature, 411巻, 494頁, 2001年) に準じて、 本発明のポリヌクレオチドの配列を基に設計して製造することができる。  The double-stranded RNA can be designed and produced based on the sequence of the polynucleotide of the present invention according to a known method (eg, Nature, 411, 494, 2001).
リポザィムは、 公知の方法 (例、 TRENDS in Molecular Medic ine, 7巻, 221頁, 2001年) に準じて、 本発明のポリヌクレオチドの配列を基に設計して製造するこ とができる。 例えば、 公知のリポザィムの配列の一部を本発明のタンパク質をコ ードする R N Aの一部に置換することによって製造することができる。 本発明の タンパク質をコードする R NAの一部としては、 公知のリポザィムによって切断 され得るコンセンサス配列 NU X (式中、 Nはすべての塩基を、 Xは G以外の塩 基を示す) の近傍の配列などが挙げられる。 , 上記の二重鎖 R NAまたはリポザィムを上記予防 ·治療剤として使用する場合、 ァンチセンスポリヌクレオチドと同様にして製剤化し、 投与することができる。 また、 前記 (V) の発現ベクターは、 公知の遺伝子治療法などと同様に用い、 上 記予防 ·治療剤として使用する。 〔6〕 本発明の抗体を含有する医薬 The lipozyme can be designed and manufactured based on the sequence of the polynucleotide of the present invention according to a known method (eg, TRENDS in Molecular Medicine, Vol. 7, pp. 221, 2001). For example, it can be produced by substituting a part of a known lipozyme sequence with a part of RNA encoding the protein of the present invention. As a part of the RNA encoding the protein of the present invention, a consensus sequence NU X (where N represents all bases, X represents a base other than G) which can be cleaved by a known lipozyme, And the like. When the above-mentioned double-stranded RNA or lipozyme is used as the above-mentioned prophylactic / therapeutic agent, it can be formulated and administered in the same manner as the antisense polynucleotide. The expression vector (V) is used in the same manner as known gene therapy methods and the like, and is used as the above-mentioned prophylactic / therapeutic agent. [6] A drug containing the antibody of the present invention
本発明のタンパク質の活性を中和する作用を有する本発明の抗体は、 本発明の タンパク質の機能を抑制 (例、 シグナル伝達を不活性化) することができるので、 例えば、 肥満症 (例、 悪性肥満細胞症、 外因性肥満、 過インシュリン性肥満症、 過血漿性肥満、 下垂体性肥満、 減血漿性肥満症、 甲状腺機能低下肥満症、 視床下 部性肥満、 症候性肥満症、 小児肥満、 上半身肥満、 食事性肥満症、 性機能低下性 · 肥満、 全身性肥満細胞症、 単純性肥満、 中心性肥満など) 、 摂食亢進症、 情動障 害、 性機能障害、 過強陣痛、 強直性子宮収縮、 胎児仮死、 子宮破裂、 類管裂傷、 早産、 Prader-Wi l l i症候群、 糖尿病およびその合併症 (例、 糖尿病性腎症、 糖尿 病性網膜症、 糖尿病性神経障害など) 、 高血圧、 高脂血症、 冠状動脈硬化症、 痛 風、 呼吸器疾患 (Pi ckwick症候群、 睡眠時無呼吸症候群) 、 脂肪肝、 不妊症、 変 形性骨関節症など (特に抗肥満剤、 食欲 (摂食) 調節剤など) などの予防 ·治療 剤などとして使用することができる。  Since the antibody of the present invention having the action of neutralizing the activity of the protein of the present invention can suppress the function of the protein of the present invention (eg, inactivate signal transduction), for example, obesity (eg, obesity) Malignant mastocytosis, exogenous obesity, hyperinsulinic obesity, hyperplasmic obesity, pituitary obesity, hypoplasmic obesity, hypothyroid obesity, hypothalamic obesity, symptomatic obesity, childhood obesity , Upper body obesity, dietary obesity, hypogonadism, obesity, systemic mastocytosis, simple obesity, central obesity, etc.), hyperphagia, affective disorder, sexual dysfunction, labour, tonicity Uterine contractions, fetal asphyxia, uterine rupture, ruptured tubules, preterm birth, Prader-Willi syndrome, diabetes and its complications (eg, diabetic nephropathy, diabetic retinopathy, diabetic neuropathy, etc.), hypertension, Hyperlipidemia, coronary Pulse sclerosis, gout, respiratory disease (Pickwick syndrome, sleep apnea syndrome), fatty liver, infertility, osteoarthritis, etc. (especially antiobesity agents, appetite (feeding) regulators, etc.) It can be used as a prophylactic and therapeutic agent.
本発明のタンパク質の活性を活性化する作用を有する本発明の抗体は、 本発明 のタンパク質の機能を促進 (例、 シグナル伝達を活性化) することができるので、 例えば、 食欲 (摂食) 増進剤、 食欲不振 (例、 神経性食欲不振症など) 、 食欲不 振に伴う貧血または低蛋白症、 微弱陣痛、 弛緩出血、 子宮復古不全、 乳汁うっ滞 などの予防 ·治療剤などとして用いることができる。  Since the antibody of the present invention having an activity of activating the activity of the protein of the present invention can promote the function of the protein of the present invention (eg, activate signal transduction), for example, increase appetite (feeding) Drugs, anorexia (eg, anorexia nervosa), anemia or hypoproteinemia associated with anorexia, weak labor, lax bleeding, uterine remodeling failure, milk stasis, etc. it can.
本発明の抗体は、 それ自体または適当な医薬組成物として投与することができ る。 上記投与に用いられる医薬組成物は、 上記抗体またはその塩と薬理学的に許 容され得る担体、 希釈剤もしくは賦形剤とを含むものである。 かかる組成物は、 経口または非経口投与に適する剤形として提供される。  The antibodies of the present invention can be administered by themselves or as a suitable pharmaceutical composition. The pharmaceutical composition used for the administration contains the antibody or a salt thereof and a pharmacologically acceptable carrier, diluent or excipient. Such compositions are provided in dosage forms suitable for oral or parenteral administration.
すなわち、 例えば、 経口投与のための組成物としては、 固体または液体の剤形、 具体的には錠剤 (糖衣錠、 フィルムコ一ティング錠を含む) 、 丸剤、 顆粒剤、 散 剤、 カプセル剤 (ソフトカプセル剤を含む) 、 シロップ剤、 乳剤、 懸濁剤などが あげられる。 かかる組成物は公知の方法によって製造され、 製剤分野において通 常用いられる担体、 希釈剤もしくは賦形剤を含有するものである。 例えば、 錠剤 用の担体、 賦形剤としては、 乳糖、 でんぷん、 蔗糖、 ステアリン酸マグネシウム などが用いられる。 非経口投与のための組成物としては、 例えば、 注射剤、 坐剤などが用いられ、 注射剤は静脈注射剤、 皮下注射剤、 皮内注射剤、 筋肉注射剤、 点滴注射剤などの 剤形を包含する。 かかる注射剤は、 公知の方法に従って、 例えば、, 上記抗体また はその塩を通常注射剤に用いられる無菌の水性もしくは油性液に溶解、 懸濁また は乳化することによって調製する。. 注射用の水性液としては、 例えば、 生理食塩 水、 ブドウ糖やその他の補助薬を含む等張液などが用いられ、 適当な溶解補助剤、 例えば、 アルコール (例、 エタノール) 、 ポリアルコール (例、 プロピレンダリ コール、 ポリエチレングリコ一ル) 、 非イオン界面活性剤 〔例、 ポリソルべ一ト 80、 HCO-50 (po lyoxye thylene (50mo l) adduc t o f hydrogenated cas tor o i l) 〕 などと併用してもよい。 油性液としては、 例えば、 ゴマ油、 大豆油など が用いられ、 溶解補助剤として安息香酸ベンジル、 ベンジルアルコールなどを併 用してもよい。 調製された注射液は、 通常、 適当なアンプルに充填される。 直腸 投与に用いられる坐剤は、 上記抗体またはその塩を通常の坐薬用基剤に混合する ことによって調製される。 That is, for example, compositions for oral administration include solid or liquid dosage forms, specifically tablets (including sugar-coated tablets and film-coated tablets), pills, granules, powders, capsules ( Soft capsules), syrups, emulsions, suspensions and the like. Such a composition is produced by a known method and contains a carrier, diluent or excipient usually used in the pharmaceutical field. For example, lactose, starch, sucrose, magnesium stearate and the like are used as carriers and excipients for tablets. As compositions for parenteral administration, for example, injections, suppositories, etc. are used. Injections are in the form of intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections, infusions, etc. Is included. Such injections are prepared according to known methods, for example, by dissolving, suspending or emulsifying the antibody or a salt thereof in a sterile aqueous or oily liquid commonly used for injections. Aqueous liquids for injection include, for example, saline, isotonic solutions containing glucose and other adjuvants, and suitable solubilizing agents, such as alcohol (eg, ethanol) and polyalcohol (eg, , Propylene glycol, polyethylene glycol), nonionic surfactants (eg, polysorbate 80, HCO-50 (polyoxythylene (50 mol) adduc tof hydrogenated cas tor oil)), etc. Good. As the oily liquid, for example, sesame oil, soybean oil, and the like are used, and benzyl benzoate, benzyl alcohol, and the like may be used as a solubilizing agent. The prepared injection is usually filled in an appropriate ampoule. A suppository for rectal administration is prepared by mixing the above antibody or a salt thereof with a conventional suppository base.
上記の経口用または非経口用医薬組成物は、 活性成分の投与量に適合するよう な投薬単位の剤形に調製されることが好都合である。 かかる投薬単位の剤形とし ては、 錠剤、 丸剤、 カプセル剤、 注射剤 (アンプル) 、 坐剤などが例示され、 そ れぞれの投薬単位剤形当たり通常 5〜500mg、 とりわけ注射剤では 5〜100mg、 その 他の剤形では 10〜 25 Omgの上記抗体が含有されていることが好ましい。  The above-mentioned oral or parenteral pharmaceutical composition is conveniently prepared in a unit dosage form adapted to the dose of the active ingredient. Examples of such dosage unit dosage forms include tablets, pills, capsules, injections (ampoules), suppositories and the like. Usually, each dosage unit dosage form is 5 to 500 mg, especially for injections. Preferably, 5 to 100 mg, and other dosage forms, contain 10 to 25 Omg of the antibody.
なお前 した各組成物は、 上記抗体との配合により好ましくない相互作用を生 じない限り他の活性成分を含有してもよい。  Each of the above-mentioned compositions may contain other active ingredients as long as no undesirable interaction occurs due to the combination with the above antibody.
本発明の抗体を含有する上記疾患の予防 ·治療剤は低毒性であり、 そのまま液 剤として、 または適当な剤型の医薬組成物として、 ヒトまたは哺乳動物 (例、 ラ ット、 ゥサギ、 ヒッジ、 ブ夕、 ゥシ、 ネコ、 ィヌ、 サルなど) に対して経口的ま たは非経口的 (例、 静脈投与) に投与することができる。 投与量は、 投与対象、 対象疾患、 症状、 投与ルートなどによっても異なるが、 例えば、 成人の肥満症の 治療のために使用する場合には、 本発明の中和抗体を 1回量として、 通常 0. 01〜 20mg/kg体重程度、 好ましくは 0. l〜10mg/kg体重程度、 さらに好ましくは 0. 1〜 5mg/kg体重程度を、 1日 1〜5回程度、 好ましくは 1日 1〜3回程度、 注射剤として投 与するのが好都合である。 他の非経口投与および経口投与の場合もこれに準ずる 量を投与することができる。 症状が特に重い場合には、 その症状に応じて増量し てもよい。 The prophylactic / therapeutic agent for the above-mentioned diseases containing the antibody of the present invention has low toxicity and can be used as it is as a liquid or as a pharmaceutical composition of an appropriate dosage form, in humans or mammals (eg, rat, egret, sheep, etc.). Can be administered orally or parenterally (eg, intravenously) to mice, dogs, cats, cats, dogs, monkeys, etc.). The dosage varies depending on the administration subject, target disease, symptoms, administration route, and the like. 0.01 to 20 mg / kg body weight, preferably 0.1 to 10 mg / kg body weight, more preferably 0.1 to 5 mg / kg body weight, about 1 to 5 times a day, preferably 1 to 5 times a day About 3 times as injection It is convenient to give. In the case of other parenteral administration and oral administration, an equivalent amount can be administered. If the symptoms are particularly severe, the dose may be increased accordingly.
また、 本発明の抗体は、'例えば、 上記疾患などの診断薬としても有用である。  Further, the antibody of the present invention is also useful, for example, as a diagnostic agent for the above-mentioned diseases and the like.
〔7〕 本発明の DNAを有する動物の作出 [7] Creation of an animal having the DNA of the present invention
本発明は、 外来性の本発明のタンパク質をコードする DNA (以下、 本発明の 外来性 DNAと略記する) またはその変異 DNA (本発明の外来性変異 DN Aと 略記する場合がある) を有する非ヒト哺乳動物を提供する。  The present invention has a DNA encoding the exogenous protein of the present invention (hereinafter abbreviated as the exogenous DNA of the present invention) or a mutant DNA thereof (sometimes abbreviated as the exogenous mutant DNA of the present invention). A non-human mammal is provided.
すなわち、 本発明は、  That is, the present invention
(1) 本発明の外来性 D N Aまたはその変異 D N Aを有する非ヒト哺乳動物、 (1) a non-human mammal having an exogenous DNA of the present invention or a mutant DNA thereof,
(2) 非ヒト哺乳動物がゲッ歯動物である上記 (1) 記載の動物、 (2) the animal according to (1), wherein the non-human mammal is a rodent;
(3) ゲッ歯動物がマウスまたはラットである上記 (2) 記載の動物、 および (3) The animal according to (2), wherein the rodent is a mouse or a rat, and
(4) 本発明の外来性 DNAまたはその変異 DNAを含有し、 哺乳動物において 発現しうる組換えベクターなどを提供する。 (4) To provide a recombinant vector or the like containing the exogenous DNA of the present invention or its mutant DNA, which can be expressed in mammals.
本発明の外来性 DNAまたはその変異 DNAを有する非ヒ卜哺乳動物 (以下、 本発明の DNA導入動物と略記する) は、 未受精卵、 受精卵、 精子およびその始 原細胞を含む胚芽細胞などに対して、 好ましくは、 非ヒト哺乳動物の発生におけ る胚発生の段階 (さらに好ましくは、 単細胞または受精卵細胞の段階でかつ一般 fo 8細胞期以前) に、 リン酸カルシウム法、 電気パルス法、 リポフエクシヨン法、 凝集法、 マイクロインジェクション法、 パーティクルガン法、 DEAE—デキス トラン法などにより目的とする DN Aを導入することによって作出することがで きる。 また、 該 DNA導入方法により、 体細胞、 生体の臓器、 組織細胞などに目 的とする本発明の外来性 DN Aを導入し、 細胞培養、 組織培養などに利用するこ ともでき、 さらに、 これら細胞を上述の胚芽細胞と公知の細胞融合法により融合 させることにより本発明の DN A導入動物を作出することもできる。  Non-human mammals having the exogenous DNA of the present invention or the mutant DNA thereof (hereinafter abbreviated as the DNA-transfected animal of the present invention) include unfertilized eggs, fertilized eggs, germ cells including spermatozoa and their progenitor cells, and the like. Preferably, at the stage of embryonic development in non-human mammal development (more preferably, at the stage of a single cell or a fertilized egg and before the general fo 8 cell stage), the calcium phosphate method, the electric pulse method, the Lipofection It can be produced by introducing the target DNA by a method such as a coagulation method, a coagulation method, a microinjection method, a particle gun method, or a DEAE-dextran method. Further, by the DNA introduction method, the exogenous DNA of the present invention intended for somatic cells, organs of living organisms, tissue cells, and the like can be introduced and used for cell culture, tissue culture, and the like. The DNA-introduced animal of the present invention can also be produced by fusing the cells with the above-mentioned germ cells by a known cell fusion method.
非ヒト哺乳動物としては、 例えば、 ゥシ、 ブタ、 ヒッジ、 ャギ、 ゥサギ、 ィヌ、 ネコ、 モルモット、 八ムスター、 マウス、 ラットなどが用いられる。 なかでも、 病体動物モデル系の作成の面から個体発生および生物サイクルが比較的短く、 ま た、 繁殖が容易なゲッ歯動物、 とりわけマウス (例えば、 純系として、 C 57B LZ6系統, DBA2系統など、 交雑系として、 B S C SFi系統, BDFi系統, B 6D2 Ft系統, BALBZc系統, I CR系統など) またはラヅト (例えば、 W i s t a r , SDなど) などが好ましい。 ' As the non-human mammal, for example, porcupine, pig, hidge, goat, magpie, dog, cat, guinea pig, eight-star, mouse, rat and the like are used. Above all, the ontogeny and biological cycle are relatively short in terms of the creation of disease animal model systems. In addition, rodents that are easy to breed, especially mice (for example, pure strains such as C57B LZ6 strain and DBA2 strain, and hybrid strains such as BSC SFi strain, BDFi strain, B6D2 Ft strain, BALBZc strain, ICR strain, etc. ) Or light (eg, Wistar, SD, etc.) is preferred. '
哺乳動物において発現しうる組換えべクタ一における 「哺乳動物」 としては、 上記の非ヒト哺乳動物の他にヒトなどがあげられる。  Examples of the “mammal” in the recombinant vector that can be expressed in mammals include humans and the like in addition to the above-mentioned non-human mammals.
本発明の外来性 DN Aとは、'非ヒト哺乳動物が本来有している本発明の DN A ではなく、 いったん哺乳動物から単離 ·抽出された本発明の DNAをいう。  The exogenous DNA of the present invention does not mean the DNA of the present invention originally possessed by a non-human mammal but refers to the DNA of the present invention once isolated and extracted from the mammal.
本発明の変異 DNAとしては、 元の本発明の DN Aの塩基配列に変異 (例えば、 突然変異など) が生じたもの、 具体的には、 塩基の付加、 欠損、 他の塩基への置 換などが生じた DNAなどが用いられ、 また、 異常 DNAも含まれる。  As the mutant DNA of the present invention, DNA having a mutation (for example, mutation) in the base sequence of the original DNA of the present invention, specifically, addition, deletion, or substitution of another base to another base DNA that has been used is used, and also includes abnormal DNA.
該異常 DN Aとしては、 異常な本発明のタンパク質を発現させる DNAを意味 し、 例えば、 正常な本発明のタンパク質の機能を抑制するタンパク質を発現させ 'る DNAなどが用いられる。  The abnormal DNA means a DNA that expresses an abnormal protein of the present invention. For example, a DNA that expresses a protein that suppresses the function of the normal protein of the present invention is used.
本発明の外来性 DNAは、 対象とする動物と同種あるいは異種のどちらの哺乳 動物由来のものであってもよい。 本発明の DN Aを対象動物に導入するにあたつ ては、 該 DNAを動物細胞で発現させうるプロモータ一の下流に結合した DNA コンストラクトとして用いるのが一般に有利である。 例えば、 本発明のヒト DN Aを導入する場合、 これと相同性が高い本発明の DN Aを有する各種哺乳動物 (例えば、 ゥサギ、 ィヌ、 ネコ、 モルモット、 ハムスター、 ラット、 マウスな ど) 由来の DNAを発現させうる各種プロモーターの下流に、 本発明のヒト DN Aを結合した DNAコンストラクト (例、 ベクタ一など) を対象哺乳動物の受精 卵、 例えば、 マウス受精卵へマイクロインジェクションすることによって本発明 の DN Aを高発現する DN A導入哺乳動物を作出することができる。  The exogenous DNA of the present invention may be derived from a mammal of the same species or a different species as the target animal. In introducing the DNA of the present invention into a subject animal, it is generally advantageous to use the DNA as a DNA construct linked downstream of a promoter capable of being expressed in animal cells. For example, when the human DNA of the present invention is introduced, it is derived from various mammals having the DNA of the present invention having a high homology with the DNA (eg, egret, dog, cat, guinea pig, hamster, rat, mouse, etc.). By injecting the DNA construct of the present invention (eg, a vector) into a fertilized egg of a target mammal, eg, a mouse fertilized egg, downstream of various promoters capable of expressing the DNA of the present invention. A DNA-introduced mammal that highly expresses the DNA of the present invention can be produced.
本発明のタンパク質の発現べクタ一としては、 大腸菌由来のプラスミド、 枯草 菌由来のプラスミド、 酵母由来のプラスミド、 λファ一ジなどのパクテリオファ ージ、 モロニ一白血病ウィルスなどのレトロウイルス、 ワクシニアウィルスまた はバキュロウィルスなどの動物ウィルスなどが用いられる。 なかでも、 大腸菌由 来のプラスミド、 枯草菌由来のプラスミドまたは酵母由来のプラスミドなどが好 ましく用いられる。 ' , Examples of the expression vector of the protein of the present invention include a plasmid derived from Escherichia coli, a plasmid derived from Bacillus subtilis, a plasmid derived from yeast, a bacteriophage such as λ phage, a retrovirus such as Moroni leukemia virus, a vaccinia virus and the like. For example, animal viruses such as baculovirus are used. Among them, plasmids derived from Escherichia coli, plasmids derived from Bacillus subtilis or plasmids derived from yeast are preferred. It is used well. ',
上記の DNA発現調節を行なうプロモーターとしては、 例えば、 (i) ウィル ス (例、 シミアンウィルス、 サイトメガロウィルス、 モロニ一白血病ウィルス、 J Cウィルス、 乳癌ウィルス、 ポリオウイルスなど) に由来する DNAのプロモ —夕一、 (ii) 各種哺乳動物 (ヒト、 ゥサギ、 ィヌ、 ネコ、 モルモット、 八ムス 夕一、 ラット、 マウスなど) 由来のプロモー夕一、 例えば、 アルブミン、 インス リン I I、 ゥロプラキン I I、 エラス夕ーゼ、 エリスロポエチン、 エンドセリン、 筋クレアチンキナーゼ、 グリア線維性酸性タンパク質、 ダル夕チオン S—トラン スフエラ一ゼ、 血小板由来成長因子 13、 ケラチン K l, 1 0ぉょび 14、 コ ラ一ゲン I型および I I型、 サイクリック AMP依存タンパク質キナーゼ /3 Iサ ブュニット、 ジストロフィン、 酒石酸抵抗性アル力リフォスファターゼ、 心房ナ トリウム利尿性因子、 内皮レセプターチ口シンキナーゼ (一般に T i e 2と略さ れる) 、 ナトリウムカリウムアデノシン 3リン酸化酵素 (Na, K一 ATP a s e) 、 ニューロフィラメント軽鎖、 メタ口チォネイン Iおよび I I A、 メタロブ ロティナーゼ 1組織インヒビ夕一、 MHCクラス I抗原 (H— 2 L) 、 H— r a s、 レニン、 ド一パミン) 3—水酸化酵素、 甲状腺ペルォキシダ一ゼ (TPO) 、 ' ポリペプチド鎖延長因子 1 a (EF- 1 ) 、 βァクチン、 αおよび )3ミオシン 重鎖、 ミオシン軽鎖 1および 2、 ミエリン基礎タンパク質、 チログロブリン、 Τ hy_ l、 免疫グロブリン、 H鎖可変部 (VNP) 、 血清アミロイド Pコンポ一 ネント、 ミオグロビン、 トロポニン (:、 平滑筋ひァクチン、 プレプロエンケファ リン A、 バソプレシンなどのプロモーターなどが用いられる。 なかでも、 全身で 高発現することが可能なサイトメガロウィルスプロモーター、 ヒトポリペプチド 鎖延長因子 1 ά (EF- 1 α) のプロ'モ一ター、 ヒトおよびニヮトリ)3ァクチン プロモー夕一などが好適である。  Examples of promoters that regulate the DNA expression include, for example, (i) DNA promoters derived from viruses (eg, Simian virus, cytomegalovirus, Moroni leukemia virus, JC virus, breast cancer virus, poliovirus, etc.) Yuichi, (ii) Promoters derived from various mammals (humans, egrets, dogs, cats, guinea pigs, mussels, rats, mice, etc.), such as albumin, insulin II, ゥ roplakin II, Erasu evening Protease, erythropoietin, endothelin, muscle creatine kinase, glial fibrillary acidic protein, dalyuthione S-transferase, platelet-derived growth factor 13, keratin Kl, 10 and 14, collagen I And II, cyclic AMP-dependent protein kinase / 3I subunit, dystrophy Int, tartrate-resistant alkaline phosphatase, atrial sodium diuretic factor, endothelial receptor thymic synthase (generally abbreviated as Tie 2), sodium potassium adenosine 3 kinase (Na, K-ATPase), Neurofilament light chain, meta-oral thionine I and IIA, metallobrotinase 1 tissue inhibitor, MHC class I antigen (H-2L), H-ras, renin, dopamine) 3-hydroxylase, thyroid gland peroxida-1 (TPO), '' polypeptide chain elongation factor 1a (EF-1), β-actin, α and) 3 myosin heavy chain, myosin light chain 1 and 2, myelin basic protein, thyroglobulin, Τhy_l, immunoglobulin , H chain variable region (VNP), serum amyloid P component, myoglobin, troponin (:, smooth muscle actin, preproenke Promoters such as farin A and vasopressin are used, among them, cytomegalovirus promoter, which can be highly expressed in the whole body, and human polypeptide chain elongation factor 1ά (EF-1α) promoter. , Human and chicken) 3 actin promoter.
上記べクタ一は、 DNA導入哺乳動物において目的とする mRNAの転写を終 結する配列 (一般に夕一ミネタ一と呼ばれる) を有していることが好ましく、 例 えば、 ウィルス由来および各種哺乳動物由来の各 DNAの配列を用いることがで き、 好ましくは、 シミアンウィルスの SV40夕一ミネタ一などが用いられる。 その他、 目的とする外来性 DNAをさらに高発現させる目的で各 DNAのスプ ライシングシグナル、 ェンハンサ一領域、 真核 D NAのイントロンの一部などを プロモーター領域の 5, 上流、 プロモーター領域と翻訳領域間あるいは翻訳領域 の 3 ' 下流 に連結することも目的により可能である。 The vector preferably has a sequence that terminates the transcription of the target mRNA in the DNA-transfected mammal (generally referred to as "Yuichi Mineta-1"). For example, it is derived from viruses and various mammals. The sequence of each DNA can be used. Preferably, Simian virus SV40 or the like is used. In addition, the splicing of each DNA for the purpose of further expressing the desired foreign DNA It is also possible to link a licensing signal, an enhancer region, a part of an intron of a eukaryotic DNA, etc., at 5, upstream of the promoter region, between the promoter region and the translation region, or 3 'downstream of the translation region.
正常な本発明のタンパク質の翻訳領域は、 ヒトまたは各種哺乳動物 (例えば、 ゥサギ、 ィヌ、 ネコ、 モルモット、 ハムスター、 ラット、 マウスなど) 由来の肝 臓、 腎臓、 甲状腺細胞、 線維芽細胞由来 D N Aおよび市販の各種ゲノム D N Aラ イブラリーよりゲノム D NAの全てあるいは一部として、 または肝臓、 腎臓、 甲 状腺細胞、 線維芽細胞由来 R NAより公知の方法により調製された相補 D NAを 原料として取得することが出来る。 また、 外来性の異常 D N Aは、 上記の細胞ま たは組織より得られた正常なポリぺプチドの翻訳領域を点突然変異誘発法により 変異した翻訳領域を作製することができる。  The normal translation region of the protein of the present invention is DNA derived from liver, kidney, thyroid cells, fibroblasts derived from humans or various mammals (eg, egrets, dogs, cats, guinea pigs, hamsters, rats, mice, etc.). And all or part of genomic DNA from various commercially available genomic DNA libraries, or complementary DNA prepared by known methods from liver, kidney, thyroid cells, and fibroblast-derived RNA as raw materials I can do it. In addition, an exogenous abnormal DNA can produce a translation region obtained by mutating a normal polypeptide translation region obtained from the above cells or tissues by a point mutagenesis method.
該翻訳領域は導入動物において発現しうる D N Aコンストラクトとして、 前記 のプロモーターの下流および所望により転写終結部位の上流に連結させる通常の  The translation region is a DNA construct that can be expressed in an introduced animal, which is usually ligated to the downstream of the promoter and optionally the upstream of a transcription termination site.
D N A工学的手法により作製することができる。 It can be produced by a DNA engineering technique.
受精卵細胞段階における本発明の外来性 D N Aの導入は、 対象哺乳動物の胚芽 細胞および体細胞のすべてに存在するように確保される。 D N A導入後の作出動 物の胚芽細胞において、 本発明の外来性 D N Aが存在することは、 作出動物の後 代がすべて、 その胚芽細胞および体細胞のすべてに本発明の外来性 D N Aを保持 することを意味する。 本発明の外来性 D N Aを受け継いだこの種の動物の子孫は その胚芽細胞および体細胞のすべてに本発明の外籴性 D N Aを有する。  Introduction of the exogenous DNA of the present invention at the fertilized egg cell stage is ensured to be present in all germ cells and somatic cells of the target mammal. The presence of the exogenous DNA of the present invention in the germinal cells of the transgenic animal after the DNA transfer means that all the progeny of the transgenic animal retain the exogenous DNA of the present invention in all of the germ cells and somatic cells Means that. The offspring of this type of animal that has inherited the exogenous DNA of the present invention have the exogenous DNA of the present invention in all of its germ cells and somatic cells.
本発明の外来性正常 D NAを導入した非ヒト哺乳動物は、 交配により外来性 D N Aを安定に保持することを確認して、 該 D N A保有動物として通常の飼育環境 で継代飼育することが出来る。  A non-human mammal into which the exogenous normal DNA of the present invention has been introduced can be subcultured in a normal breeding environment as an animal having the DNA after confirming that the exogenous DNA is stably retained by the crossing. .
受精卵細胞段階における本発明の外来性 D N Aの導入は、 対象哺乳動物の胚芽 細胞および体細胞の全てに過剰に存在するように確保される。 D NA導入後の作 出動物の胚芽細胞において本発明の外来性 D NAが過剰に存在することは、 作出 動物の子孫が全てその胚芽細胞および体細胞の全てに本発明の外来性 D N Aを過 剰に有するごとを意味する。 本発明の外来性 D NAを受け継いだこの種の動物の 子孫はその胚芽細胞および体細胞の全てに本発明の外来性 D N Aを過剰に有する < 導入 D NAを相同染色体の両方に持つホモザィゴ一ト動物を取得し、 この雌雄 の動物を交配することによりすべての子孫が該 D N Aを過剰に有するように繁殖 継代することがで る。 Introduction of the exogenous DNA of the present invention at the fertilized egg cell stage is ensured to be present in excess in all germ cells and somatic cells of the target mammal. Excessive presence of the exogenous DNA of the present invention in the germinal cells of the produced animal after the introduction of the DNA indicates that all the offspring of the produced animal carry the exogenous DNA of the present invention in all of its germ cells and somatic cells. It means every time you have it. The progeny of this type of animal that has inherited the exogenous DNA of the present invention has an excess of the exogenous DNA of the present invention in all of its germinal and somatic cells. By obtaining a homozygous animal having the introduced DNA on both homologous chromosomes and mating the male and female animals, it is possible to breed and passage so that all offspring have the DNA in excess.
本発明の正常 D NAを有する非ヒ卜哺乳動物は、 本発明の正常 D NAが高発現 させられており、 内在性の正常 D NAの機能を促進することにより最終的に本発 明のタンパク質の機能宂進症を発症することがあり、 その病態モデル動物として 利用することができる。 例えば、 本発明の正常 D NA導入動物を用いて、 本発明 のタンパク質の機能亢進症や、 本発明のタンパク質が関連する疾患の病態機序の 解明およびこれらの疾患の治療方法の検討を行なう とが可能である。 ' また、 本発明の外来性正常 D NAを導入した哺乳動物は、 遊離した本発明の夕 ンパク質の増加症状を有することから、 本発明のタンパク質に関連する疾患に対 する治療薬のスクリーニング試験にも利用可能である。  The non-human mammal having the normal DNA of the present invention expresses the normal DNA of the present invention at a high level, and finally promotes the function of the endogenous normal DNA, thereby finally obtaining the protein of the present invention. May develop functional hyperplasia, and can be used as a disease model animal. For example, using the normal DNA-introduced animal of the present invention to elucidate the pathological mechanism of hyperactivity of the protein of the present invention and diseases associated with the protein of the present invention, and to examine a method for treating these diseases. Is possible. '' In addition, since a mammal into which the exogenous normal DNA of the present invention has been introduced has an increased symptom of the released protein of the present invention, a screening test for a therapeutic agent for a disease associated with the protein of the present invention is performed. Is also available.
一方、 本発明の外来性異常 D NAを有する非ヒト哺乳動物は、 交配により外来 性 D N Aを安定に保持することを確認して該 D N A保有動物として通常の飼育環 境で継代飼育することが出来る。 さらに、 目的とする外来 D NAを前述のプラス ミドに組み込んで原科として用いることができる。 プロモーターとの D N Aコン ストラク卜は、 通常の D NA工学的手法によって作製することができる。 受精卵 細胞段階における本発明の異常 D N Aの導入は、 対象哺乳動物の胚芽細胞および 体細胞の全てに存在するように確保される。 D NA転移後の作出動物の胚芽細胞 において本発明の異常 D N Aが存在することは、 作出動物の子孫が全てその胚芽 細胞および体細胞の全てに本発明の異常 D N Aを有することを意味する。 本発明 の外来性 D N Aを受け継いだこの種の動物の子孫は、 その胚芽細胞および体細胞 の全てに本発明の異常 D N Aを有する。'導入 D N Aを相同染色体の両方に持つホ モザィゴート動物を取得し、 この雌雄の動物を交配することによりすべての子孫 が該 D NAを有するように繁飨継代することができる。  On the other hand, a non-human mammal having the exogenous abnormal DNA of the present invention can be subcultured in a normal breeding environment as an animal having the DNA after confirming that the exogenous DNA is stably maintained by mating. I can do it. Furthermore, the desired exogenous DNA can be incorporated into the above-mentioned plasmid and used as a raw material. The DNA construct with the promoter can be prepared by ordinary DNA engineering techniques. Introduction of the abnormal DNA of the present invention at the fertilized egg cell stage is ensured to be present in all germ cells and somatic cells of the target mammal. The presence of the abnormal DNA of the present invention in the germinal cells of the animal produced after the transfer of DNA means that all the offspring of the animal produced have the abnormal DNA of the present invention in all of the germ cells and somatic cells. The progeny of this type of animal that has inherited the exogenous DNA of the present invention has the abnormal DNA of the present invention in all of its germ cells and somatic cells. 'A homozygous animal having the introduced DNA on both homologous chromosomes is obtained, and by crossing these male and female animals, it is possible to propagate the offspring so that all offspring have the DNA.
本発明の異常 D NAを有する非ヒ卜哺乳動物は、 本発明の異常 D NAが高発現 させられており、 内在性の正常 D N Aの機能を阻害することにより最終的に本発 明の夕ンパク質の機能不活性型不応症となることがあり、 その病態モデル動物と して利用することができる。 例えば、 本発明の異常 D NA導入動物を用いて、 本 発明のタンパク質の機能不活性型不応症の病態機序の解明およびこの疾患を治療 方法の検討を行なうことが可能である。 The non-human mammal having the abnormal DNA of the present invention expresses the abnormal DNA of the present invention at a high level, and finally inhibits the function of endogenous normal DNA, thereby ultimately obtaining the protein of the present invention. In some cases, it becomes a functional inactive refractory disease and can be used as a model animal for the disease. For example, using the abnormal DNA-introduced animal of the present invention, It is possible to elucidate the pathological mechanism of the function-inactive refractory of the protein of the present invention and to examine a method for treating this disease.
また、 具体的な利用可能性としては、 本発明の異常 D NA高発現動物は、 本発 明のタンパク質の機能不活性型不応症における本発明の異常タンパク質による正 常タンパク質の機能阻害 (dominant negat ive作用) を解明するモデルとなる。 また、 本発明の外来異常 D NAを導入した哺乳動物は、 遊離した本発明のタン パク質の増加症状を有することから、 本発明のタンパク質またはその機能不活性 型不応症に対する治療薬スクリーニング試験にも利用可能である。  Further, as a specific possibility, the abnormal DNA-highly expressing animal of the present invention can be used to inhibit the function of a normal protein by the abnormal protein of the present invention in function-inactive refractory disease of the protein of the present invention (dominant negatase). ive effect). In addition, since the mammal into which the foreign abnormal DNA of the present invention has been introduced has an increased symptom of the released protein of the present invention, it can be used in a therapeutic drug screening test for the protein of the present invention or its functionally inactive refractory disease. Is also available.
また、 上記 2種類の本発明の D N A導入動物のその他の利用可能性として、 例 えば、  Further, as other possible uses of the above two kinds of DNA-introduced animals of the present invention, for example,
( 組織培養のための細胞源としての使用、  (Use as a cell source for tissue culture,
(i i) 本発明の D N A導入動物の組織中の D NAもしくは R N Aを直接分析する . または D N Aにより発現されたポリペプチド組織を分析することによる、 本発明 のタンパク質により特異的に発現あるいは活性化するタンパク質との関連性につ いての解析、  (ii) Direct analysis of DNA or RNA in the tissue of the DNA-introduced animal of the present invention or specific expression or activation by the protein of the present invention by analyzing a polypeptide tissue expressed by DNA Analysis of the relationship with proteins,
(i i i) D NAを有する組織の細胞を標準組織培養技術により培養し、 これらを 使用して、 一般に培養困難な組織からの細胞の機能の研究、  (iii) cells of tissues having DNA are cultured by standard tissue culture techniques, and these are used to study the function of cells from generally difficult tissues,
(iv) 上記 (i i i) 記載の細胞を用いることによる細胞の機能を高めるような薬 剤のスクリーニング、 および  (iv) screening for a drug that enhances cell function by using the cell described in (iii) above, and
(v) 本発明の変異タンパク質を単離精製およびその抗体作製などが考えられる, さらに、 本発明の D NA導入動物を用いて、 本発明のタンパク質の機能不活性 型不応症などを含む、 本発明のタンパク質に関連する疾患の臨床症状を調べるこ とができ、 また、 本発明のタンパク質に関連する疾患モデルの各臓器におけるよ り詳細な病理学的所見が得られ、 新しい治療方法の開発、 さらには、 該疾患によ る二次的疾患の研究および治療に貢献することができる。  (v) It is conceivable to isolate and purify the mutant protein of the present invention and to prepare an antibody thereof. Furthermore, using the DNA-introduced animal of the present invention, The clinical symptoms of the disease related to the protein of the present invention can be examined, and more detailed pathological findings in each organ of the disease model related to the protein of the present invention can be obtained. Furthermore, it can contribute to research and treatment of secondary diseases caused by the disease.
また、 本発明の D NA導入動物から各臓器を取り出し、 細切後、 トリプシンな どのタンパク質分解酵素により、 遊離した D NA導入細胞の取得、 その培養また はその培養細胞の系統化を行なうことが可能である。 さらに、 本発明のタンパク 質産生細胞の特定化、 アポトーシス、 分化あるいは増殖との関連性、 またはそれ らにおけるシグナル伝達機構を調べ、 それらの異常を調べることなどができ、 本 発明のタンパク質およびその作用解明のための有効な研究材料となる。 In addition, it is possible to remove each organ from the DNA-introduced animal of the present invention, cut it into small pieces, and then use a protease such as trypsin to obtain the released DNA-introduced cells, culture them, or systematize the cultured cells. It is possible. Further, the specification of the protein-producing cell of the present invention, its relationship with apoptosis, differentiation or proliferation, or They can examine the signal transduction mechanism of them and investigate their abnormalities, etc., and are useful research materials for elucidating the protein of the present invention and its action.
さらに、 本発明の DNA導入動物を用いて、 本発明のタンパク質の機能不活性 型不応症を含む、 本発明のタンパク質に関連する疾患の治療薬の開発を行なうた めに、 上述の検査法および定量法などを用いて、 有効で迅速な該疾患治療薬のス クリーニング法を提供することが可能となる。 また、 本発明の DN Α導入動物ま たは本発明の外来性 DN A発現ベクターを用いて、 本発明のタンパク質が関連す る疾患の DN A治療法を検討、 開発することが可能である。 本明細書および図面において、 塩基やアミノ酸などを略号で表示する場合、 I UPAC— IUB Commission on Biochemical Nomenclature による田各号ある いは当該分野における慣用略号に基づくものであり、 その例を下記する。 またァ ミノ酸に関し光学異性体があり得る場合は、 特に明示しなければ L体を示すもの とする。 ,  Further, in order to develop a therapeutic agent for a disease associated with the protein of the present invention, including a functionally inactive refractory type of the protein of the present invention, using the DNA-transfected animal of the present invention, Using a quantitative method or the like, it is possible to provide an effective and rapid screening method for the therapeutic agent for the disease. In addition, using the animal into which the DNA of the present invention has been introduced or the exogenous DNA expression vector of the present invention, it is possible to study and develop a method for treating DNA associated with the protein of the present invention. In the present specification and drawings, bases, amino acids, and the like are represented by abbreviations based on the respective abbreviations by the I UPAC- IUB Commission on Biochemical Nomenclature or commonly used abbreviations in the art, and examples thereof are described below. When there is an optical isomer with respect to the amino acid, the L-form is indicated unless otherwise specified. ,
DNA デォキシリポ核酸  DNA deoxylipo nucleic acid
c DNA 相補的デォキシリポ核酸  c DNA complementary deoxylipo nucleic acid
A アデニン  A adenine
T チミン  T thymine
G グァニン  G Guanin
C C
RNA リポ核酸 RNA liponucleic acid
mRNA メッセンジャ一リポ核酸  mRNA messenger liponucleic acid
d ATP デォキシアデノシン三リン酸  d ATP Deoxyadenosine triphosphate
dTTP デォキシチミジン三リン酸  dTTP Deoxythymidine triphosphate
dGTP デォキシグァノシン三リン酸  dGTP Deoxyguanosine triphosphate
d CTP '三リン酸  d CTP 'triphosphate
ATP '三リン酸  ATP 'triphosphate
EDTA エチレンジァミン四酢酸  EDTA ethylenediaminetetraacetic acid
SDS ドデシル硫酸: G 1 y :グリシン SDS dodecyl sulfate: G 1 y: glycine
A 1 a :ァラニン  A 1 a: Alanine
Va 1 :バリン  Va 1: Valine
Le u : ロイシン  Le u: Leucine
I 1 e :イソロイシン  I 1 e: Isoleucine
S e r :セリン  S e r: Serine
Th r :スレオニン  Th r: Threonine
C y s :システィン  Cys: Sistine
Me t :メチォニン  Me t: Methionin
G 1 u :グルタミン酸  G 1 u: Glutamic acid
As p :ァスパラギン酸  As p: Aspartic acid
L y s : リジン  L y s: lysine
A r g :アルギニン  A r g: Arginine
H i s : ヒスチジン  H i s: Histidine
P h e : フエ二ルァラニン  P he: feniralanin
Ty r :チロシン  Ty r: Tyrosine
T r p : 卜リブ卜ファン  T r p: Tribute fan
P r o :プロリン  Pro: Proline
A s n :ァスパラギン  A s n: Asparagine
G 1 n :ダル夕ミン  G 1 n: Dal Yu Min
p G 1 u : ピログルタミン酸  pG1u: pyroglutamic acid
また、 本明細書中で繁用される置換基、 保護基および試薬を下記の記号で表記 する。  The substituents, protecting groups and reagents frequently used in the present specification are represented by the following symbols.
Me :メチル基  Me: methyl group
E t :ェチル基  E t: ethyl group
B u :プチル基  B u: butyl group
P h :フエニル基  P h: phenyl group
TC :チアゾリジン— 4 (R) 一力ルポキサミド基  TC: Thiazolidine-4 (R) one-pot lipoxamide group
To s : p—トルエンスルフォニル CHO ホルミル To s: p-toluenesulfonyl CHO Holmill
B z 1  B z 1
Cl2-Bzl 2, 6—ジクロ口べンジル Cl 2 -Bzl 2,6-dichlorobenzene
Bom ベンジルォキシメチル  Bom benzyloxymethyl
Z ベンジルォキシカルポニル  Z benzyloxycarponyl
C 1一 Z  C 1 Z
B r -Z 2一ブロモベンジルォキシカルポニル  B r -Z 2 bromobenzyloxycarponyl
B o c t一ブトキシカルポニル  B oc t-butoxycarponyl
DNP ジニトロフエニル · T r t 卜 Uチル  DNP dinitrophenyl
Bum t—ブトキシメチル  Bum t—butoxymethyl
Fmo c N— 9 _フルォレニルメトキシカルポニル  Fmo c N— 9 _fluorenylmethoxycarbonyl
HOB t 1ーヒドロキシべンズトリアゾール  HOB t 1-hydroxybenztriazole
HOOB t 3, 4—ジヒドロー 3 _ヒドロキシ一 4一ォキソ一  HOOB t 3, 4-dihydro-3-hydroxy-1 4-oxo-1
1, 2, 3—ベンゾトリアジン  1, 2, 3-benzotriazine
HONB トヒドロキシ -5 -ノルポルネン -2, 3-ジカルボキシイミド HONB Tohydroxy-5-norpolene-2,3-dicarboximide
DCCDCC
GAPDH ダリセルアルデヒドリン酸デヒドロゲナーゼ 本願明細書の配列表の配列番号は、 以下の配列を示す。 GAPDH dariceraldehyde phosphate dehydrogenase The sequence numbers in the sequence listing in the present specification show the following sequences.
〔配列番号: 1〕  [SEQ ID NO: 1]
以下の実施例 1における P C R反応で使用したプライマ一 1の塩基配列を示す。 〔配列番号: 2〕 1 shows the nucleotide sequence of primer 11 used in the PCR reaction in Example 1 below. [SEQ ID NO: 2]
以下の実施例 1における P C R反応で使用したプライマ一 2の塩基配列を示す。 〔配列番号: 3〕 1 shows the nucleotide sequence of Primer 12 used in the PCR reaction in Example 1 below. [SEQ ID NO: 3]
力二クイザル S LT受容体をコ一ドする c DNAの塩基配列を示す。 1 shows the nucleotide sequence of cDNA encoding the cynomolgus SLT receptor.
〔配列番号: 4〕  [SEQ ID NO: 4]
力二クイザル S L T受容体のァミノ酸配列を示す。 Fig. 3 shows the amino acid sequence of the cynomolgus monkey SLT receptor.
〔配列番号: 5〕 以下の実施例 2における P C R反応で使用したプライマ一 1の塩基配列を示す。 〔配列番号: 6〕 [SEQ ID NO: 5] 2 shows the nucleotide sequence of primer 11 used in the PCR reaction in Example 2 below. [SEQ ID NO: 6]
以下の実施 112における P C R反応で使用したプライマー 2の塩基配列を示す。 〔配列番号: 7〕 The base sequence of primer 2 used in the PCR reaction in Example 112 below is shown. [SEQ ID NO: 7]
以下の実施例 2で得られたネコ SLT受容体の C末端領域をコードする cDN A の塩基配列を示す。 FIG. 3 shows the nucleotide sequence of cDNA encoding the C-terminal region of the feline SLT receptor obtained in Example 2 below.
〔配列番号: 8〕  [SEQ ID NO: 8]
以下の実施例 3における P CR反応で使用したプライマー 1の塩基配列を示す。 〔配列番号: 9〕 The base sequence of primer 1 used in the PCR reaction in Example 3 below is shown. [SEQ ID NO: 9]
以下の実施例 3における P C R反応で使用したプライマー 2の塩基配列を示す。 〔配列番号: 10〕 3 shows the base sequence of primer 2 used in the PCR reaction in Example 3 below. [SEQ ID NO: 10]
以下の実施例 3で得られたネコ S L T受容体の N末端領域をコードする c DNA の塩基配列を示す。 3 shows the nucleotide sequence of cDNA encoding the N-terminal region of the feline SLT receptor obtained in Example 3 below.
〔配列番号: 1 1〕  [SEQ ID NO: 11]
以下の実施例 4における P C R反応で使用したプライマー 1の塩基配列を示す。 〔配列番号: 12〕 The base sequence of primer 1 used in the PCR reaction in Example 4 below is shown. [SEQ ID NO: 12]
以下の実施例 4における P C R反応で使用したプライマー 2の塩基配列を示す。The base sequence of primer 2 used in the PCR reaction in Example 4 below is shown.
〔配列番号: 1 3〕 [SEQ ID NO: 13]
ネコ S L T受容体をコードする cDNAの塩基配列を示す。 1 shows the nucleotide sequence of a cDNA encoding a cat SLT receptor.
〔配列番号: 14〕  [SEQ ID NO: 14]
ネコ S L T受容体のアミノ酸配列を示す。 2 shows the amino acid sequence of the feline SLT receptor.
〔配列番号: 1 5〕  [SEQ ID NO: 15]
以下の実施例 5における P C R反応で使用したプライマー 1の塩基配列を示す。 〔配列番号: 16〕 7 shows the base sequence of primer 1 used in the PCR reaction in Example 5 below. [SEQ ID NO: 16]
以下の実施例 5における P C R反応で使用したプライマー 2の塩基配列を示す。 〔配列番号: 1 7〕 7 shows the base sequence of primer 2 used in the PCR reaction in Example 5 below. [SEQ ID NO: 17]
以下の実施例 5で得られたィヌ S L T受容体の C末端領域をコードする cDNA の塩基配列を示す。 5 shows the nucleotide sequence of cDNA encoding the C-terminal region of canine SLT receptor obtained in Example 5 below.
〔配列番号: 18〕 以下の実施例 6における P C R反応で使用したプライマ一 1の塩基配列を示す。 〔配列番号: 19〕 [SEQ ID NO: 18] 7 shows the nucleotide sequence of primer 11 used in the PCR reaction in Example 6 below. [SEQ ID NO: 19]
以下の実施例 6における P C R反応で使用したプライマー 2の塩基配列を示す。 〔配列番号: 20〕 7 shows the base sequence of primer 2 used in the PCR reaction in Example 6 below. [SEQ ID NO: 20]
以下の実施例 6で得られたィヌ S L T受容体の N末端領域をコードする c DNA の塩基配列を示す。 7 shows the nucleotide sequence of cDNA encoding the N-terminal region of canine SLT receptor obtained in Example 6 below.
〔配列番号: 21〕  [SEQ ID NO: 21]
以下の実施例 7における P C R反応で使用したプライマー 1の塩基配列を示す。 〔配列番号: 22〕 7 shows the base sequence of primer 1 used in the PCR reaction in Example 7 below. [SEQ ID NO: 22]
以下の実施例 7における P C R反応で使用したプライマー 2の塩基配列を示す。 〔配列番号: 23〕 · 7 shows the nucleotide sequence of primer 2 used in the PCR reaction in Example 7 below. [SEQ ID NO: 23]
ィヌ S LT受容体をコードする c DNAの塩基配列を示す。 1 shows the nucleotide sequence of cDNA encoding canine SLT receptor.
〔配列番号: 24〕  [SEQ ID NO: 24]
ィヌ S L T受容体のアミノ酸配列を示す。 2 shows the amino acid sequence of the canine SLT receptor.
〔配列番号: 25〕  [SEQ ID NO: 25]
MCHのアミノ酸配列を示す。 .  1 shows the amino acid sequence of MCH. .
〔配列番号: 26〕  [SEQ ID NO: 26]
Des-Asp'-MCH (MCH (2-19))のアミノ酸配列を示す。 2 shows the amino acid sequence of Des-Asp′-MCH (MCH (2-19)).
〔配列番号: 27〕  [SEQ ID NO: 27]
Des- [Asp1, Phe2]-MCH (MCH (3- 19))のアミノ酸配列を示す。 2 shows the amino acid sequence of Des- [Asp 1 , Phe 2 ] -MCH (MCH (3-19)).
〔配列番号: 28〕  [SEQ ID NO: 28]
Des- [Asp1, Phe2, Asp3] -MCH (MCH (4 - 19))のアミノ酸配列を示す。 2 shows the amino acid sequence of Des- [Asp 1 , Phe 2 , Asp 3 ] -MCH (MCH (4-19)).
〔配列番号: 29〕  [SEQ ID NO: 29]
Des- [Asp1, Phe2, Asp3, Met4] -MCH (MCH (5- 19))のアミノ酸配列を示す。 2 shows the amino acid sequence of Des- [Asp 1 , Phe 2 , Asp 3 , Met 4 ] -MCH (MCH (5-19)).
〔配列番号: 30〕  [SEQ ID NO: 30]
Des- [Asp1, Phe2, Asp3, Met4, Leu5] -MCH (MCH (6-19))のアミノ酸配列を示す。 〔配列番号: 31〕 2 shows the amino acid sequence of Des- [Asp 1 , Phe 2 , Asp 3 , Met 4 , Leu 5 ] -MCH (MCH (6-19)). [SEQ ID NO: 31]
Des- [Asp1, Phe2, Asp3, Met4, Leu5, Arg6]- MCH (MCH (7-19))のアミノ酸配列を示 す。 〔配列番号: 32〕 ' 以下の実施例 10で使用したプライマー 1の塩基配列を示す。 Des- [Asp 1, Phe 2, Asp 3, Met 4, Leu 5, Arg 6] - shows the amino acid sequence of the MCH (MCH (7-19)). [SEQ ID NO: 32] 'This shows the base sequence of primer 1 used in EXAMPLE 10 below.
〔配列番号: 33〕  [SEQ ID NO: 33]
以下の実施例 10で使用したプライマー 2の塩基配列を示す。 The base sequence of primer 2 used in Example 10 below is shown.
〔配列番号: 34〕  [SEQ ID NO: 34]
以下の実施例 10で使用したプローブの塩基配列を示す 〔5'末端にリポーター色 素として FAM (6-carboxy- fluorescein) を、 3,末端にはクェンチヤ一として TAMRA (6-carboxy-ietrameihyl-rhodamine) を標識した〕 。 The base sequence of the probe used in Example 10 below is shown (FAM (6-carboxy-fluorescein) as the reporter dye at the 5 'end, TAMRA (6-carboxy-ietrameihyl-rhodamine as the quencher at the 3' end). )]].
〔配列番号: 35〕 ,  [SEQ ID NO: 35],
以下の実施例 10で使用したプライマ一 3の塩基配列を示す。 The base sequence of primer 13 used in Example 10 below is shown.
〔配列番号: 36〕  [SEQ ID NO: 36]
以下の実施例 10で使用したプライマ一 4の塩基配列を示す。 The base sequence of primer 14 used in Example 10 below is shown.
〔配列番号: 37〕  [SEQ ID NO: 37]
以下の実施例 10で使用したプローブの塩基配列を示す 〔5'末端にリポーター色 素として FAM (6-carboxy- fluorescein) を、 3,末端にはクェンチヤ一として TAMRA (6-carboxy-tetramethyl-rhodamine) を標識した〕 。 The base sequence of the probe used in Example 10 below is shown [FAM (6-carboxy-fluorescein) as a reporter dye at the 5 'end, TAMRA (6-carboxy-tetramethyl-rhodamine) as a quencher at the 3 end. )]].
〔配列番号: 38〕  [SEQ ID NO: 38]
以下の実施例 1で得られた力二クイザル S L T受容体の全長をコ一ドする cDN Aの塩基配列を示す。 1 shows the nucleotide sequence of cDNA which encodes the entire length of the cynomolgus monkey SLT receptor obtained in Example 1 below.
〔配列番号: 39〕  [SEQ ID NO: 39]
以下の実施例 4で得られたネコ S LT受容体の全長をコードする c DNAの塩基 配列を示す。 7 shows the nucleotide sequence of cDNA encoding the full length of cat SLT receptor obtained in Example 4 below.
〔配列番号: 40〕  [SEQ ID NO: 40]
以下の実施例 7で得られたィヌ S LT受容体の全長をコードする c DNAの塩基 配列を示す。 後述の実施例 1で取得された Escherichia col i T0P10/pCR4_monSLTは、 2002年 6月 4日から、 日本国茨城県つくば市東卜 1-1 中央第 6 (郵便番号 305- 8566) の独 立行政法人産業技術総合研究所特許生物寄託センターに受託番号 FERM BP- 8066 として寄託されている。 7 shows the nucleotide sequence of cDNA encoding the entire length of the canine SLT receptor obtained in Example 7 below. Escherichia col i T0P10 / pCR4_monSLT obtained in Example 1 described below is an independent administrative corporation of Chuo No. 6 (Zip code 305-8566) 1-1 Toto, Tsukuba-shi, Ibaraki, Japan from June 4, 2002. Accession No. FERM BP-8066 to National Institute of Advanced Industrial Science and Technology Patent Organism Depositary Has been deposited as
後述の実施例 4で取得された Escherichia coli TOPlO/pcDNA- catSLTは、 2002 年 4月 25日から、 日本国茨城県つくば巿東卜 1-1 中央第 6 (郵便番号 305- 8566) の 独立行政法人産業技術総合研究所特許生物寄託センターに寄託番号 FERM BP - 8030として、 2002年 4月 16日から、 大阪府大阪市淀川区十三本町 2-17-85 (郵便番 号 532-8686) の財団法人発酵研究所 (IF0) に受託番号 IF0 16789としてそれぞ れ寄託されている。  Escherichia coli TOPlO / pcDNA-catSLT obtained in Example 4 described below has been administered by Independent Administration of 1-1 Chukoku No. 6 Tsukuba Totoro, Ibaraki, Japan (Postal Code 305-8566) since April 25, 2002. At the National Institute of Advanced Industrial Science and Technology, the Patent Organism Depositary, the deposit number FERM BP-8030, from April 16, 2002, 2-17-85, Jusanhoncho, Yodogawa-ku, Osaka-shi, Osaka (postal code 532-8686) They have been deposited with the Fermentation Research Institute (IF0) under the accession number IF0 16789.
後述の実施例 1 1で取得された Escherichia coli TOPlO/pAKKO-dogSLTは、 2002年 4月 25日から、 日本国茨城県つくば巿東卜 1-1 中央第 6 (郵便番号 305 - 8566) の独立行政法人産業技術総合研究所特許生物寄託センターに寄託番号 Escherichia coli TOPlO / pAKKO-dogSLT obtained in Example 11 described below has been established on April 25, 2002, as an independent company of 1-1 Chukoku No. 1-1, Tsukuba Totoro, Ibaraki, Japan (zip code 305-8566). Deposited number at Patent Organism Depositary, National Institute of Advanced Industrial Science and Technology
FERM BP-8031として、 2002年 4月 16日から、 大阪府大阪市淀川区十三本町 2-17-85From April 16, 2002, FERM BP-8031, 2-17-85 Jusanhoncho, Yodogawa-ku, Osaka, Osaka
(郵便番号 532- 8686) の財団法人発酵研究所 (IF0) に受託番号 IF0 16790とし てそれぞれ寄託されている。 以下に実施例を示して、 本発明をより詳細に説明するが、 これらは本発明の範 囲を限定するものではない。 なお、 大腸菌を用いての遺伝子操作法は、 モレキュ ラー .クロ一ニング(Molecular cloning)に記載されている方法に従った。 They have been deposited with the Fermentation Research Institute (IF0) of Japan (zip code 532-8686) under the accession number IF0 16790. Hereinafter, the present invention will be described in more detail with reference to Examples, but these do not limit the scope of the present invention. In addition, the gene manipulation method using Escherichia coli followed the method described in Molecular cloning.
実施例 1 Example 1
力二クイザル S LT遺伝子の取得  Acquisition of RLT cynomolgus SLT gene
力二クイザル視床下部より Isogen (二ツボンジーン社) を用いて total RNAを 調製後、 Oligotex (dT)30 (宝酒造) を用いて poly (A)+ RNA画分を調製した。.力二 クイサソレ視床下咅! ¾oly (A)T RNA 2. 力、ら Superscript reversetranscr iptaseAfter total RNA was prepared from the hypothalamus of the cynomolgus monkey using Isogen (Futtsubon Gene), a poly (A) + RNA fraction was prepared using Oligotex (dT) 30 (Takara Shuzo).力 oly (A) T RNA 2. Power, et al. Superscript reversetranscr iptase
(ギブコ BRL社) を用い、 マニュアルにしたがってランダムプライマーを用いて 逆転写を行なって cDNAを作成した。 ヒト S LTの 5'非翻訳領域を参考に作製した プライマー 1 (配列番号: 1 ) およびヒト 3'非翻訳領域の配列をもつプライマー 2 (配列番号: 2) を用いて力二クイザル視床下部 cDNA を錶型に PCR反応を行 なった。 反応は、 mRNA 20 ng相当分の cDNAを铸型に 20 1の液量で行なった。 反 応液の組成は、 プライマー濃度 0.5 M、 dNTP混合液 0.2mM、 Z-Taq (宝酒造). 1/100 volume, 10倍濃縮 Buffer 1/10 volumeとした。 増幅のためのサイクルは、 94°Cで 120秒保温した後、 98°C · 1秒、 72°C · 30秒のサイクルを 40回繰り返した。 得られた反応液 2 1を1"0?0 TA cloning kit (インビトロジェン社) を用いてプ ラスミドベクタ一 pcr4へサブクロ一ニングした後、 大腸菌 T0P10へ導入した。 生 じた形質転換体から QIA prep8 mini prep (キアゲン社) を用いてプラスミド DNA を精製した。 塩基配列決定のための反応は、 BigDye Terminator Cycle Sequence Ready Reaction Kit (パーキンエルマ一社) を用いて行なった。 蛍光式自動シー ケンサ一を用いて解読した結果、 配列番号: 38に示す塩基配列が得られた。 こ の配列には、 力二クイザル SLTの全アミノ酸配列 (配列番号: 4) をコードす る塩基配列 (配列番号: 3) が含まれていたので、 このプラスミドで大腸菌 TOP10を形質転換して Escherichia coli TOP10/pCR4- monSLTを得た。 実施例 2 (Gibco BRL) and reverse transcription was performed using random primers according to the manual to prepare cDNA. Using the primer 1 (SEQ ID NO: 1) prepared with reference to the 5 'untranslated region of human SLT and the primer 2 (SEQ ID NO: 2) having the sequence of the human 3' untranslated region, a cynomolgus monkey hypothalamic cDNA A PCR reaction was performed using the 錶 type. The reaction was carried out using a cDNA equivalent to 20 ng of mRNA as a type II in a volume of 201. The composition of the reaction solution was a primer concentration of 0.5 M, a dNTP mixture solution of 0.2 mM, Z-Taq (Takara Shuzo). 1/100 volume, 10-fold concentrated Buffer 1/10 volume. The cycle for amplification is After keeping it at 94 ° C for 120 seconds, a cycle of 98 ° C for 1 second and 72 ° C for 30 seconds was repeated 40 times. The resulting reaction solution 21 was subcloned into a plasmid vector pcr4 using a 1 "0-0 TA cloning kit (Invitrogen), and then introduced into E. coli T0P10. From the resulting transformant, QIA prep8 mini Plasmid DNA was purified using prep (Qiagen) The reaction for nucleotide sequence determination was performed using the BigDye Terminator Cycle Sequence Ready Reaction Kit (PerkinElmer Inc.) Using a fluorescent automatic sequencer As a result, the nucleotide sequence shown in SEQ ID NO: 38 was obtained, and this sequence contained the nucleotide sequence (SEQ ID NO: 3) encoding the entire amino acid sequence of Cynomolgus SLT (SEQ ID NO: 4) Therefore, Escherichia coli TOP10 / pCR4-monSLT was obtained by transforming Escherichia coli TOP10 with this plasmid.
3' —RACE法によるネコ SLT遺伝子の 3' 側配列の取得  3 '— Acquisition of 3' sequence of cat SLT gene by RACE method
ネコ全脳より Isogen (二ツボンジーン社) を用いて total RNAを調製後、  After preparing total RNA from the whole cat brain using Isogen (Futtsubon Gene),
Oligotex (dT) 30 (宝酒造) を用いて poly(A)+ RNA画分を調製した。 3'- RACE用の 錶型はネコ全脳 poly (A)+ RNAをもとに SMART RACE cDNA Amplification kitを用い て作製した。 RACE PCR反応に用いたプライマーセットは、 1回目の PCR反応では kit添付の Universal Primer Mixとプライマー 1 (配列番号: 5) 、 2回目の PCR 反応では kit添付の Nested Universal Primerとプライマー 2 (配列番号: 6) を 用いた。 反応は、 逆転写した00^のぅち11* 卜6 ½相当分を铸型に20 1の液量 で行なった。 反応液の組成は、 プライマー濃度 0.5 zM、 dNTP混合液 0.2 mM、 Advantage? Polymerase Mix (クロンテック社) 1/50 volumeゝ 10倍濃縮 Buffer 1/10 volumeとした。 増幅のためのサイクルは、 どちらの PCRも 94X · 5秒、 A poly (A) + RNA fraction was prepared using Oligotex (dT) 30 (Takara Shuzo). Type III for 3'-RACE was prepared based on feline whole brain poly (A) + RNA using SMART RACE cDNA Amplification kit. The primer set used for the RACE PCR reaction was as follows: the first PCR reaction included the Universal Primer Mix and primer 1 (SEQ ID NO: 5) attached to the kit; the second PCR reaction included the Nested Universal Primer and primer 2 (sequence number) attached to the kit. : 6) was used. The reaction was carried out in the form of 铸 with a volume of 201 liquid, ie, 11 * 6 equivalents of the reversely transcribed 00 ^. The composition of the reaction solution was a primer concentration of 0.5 zM, a dNTP mixed solution of 0.2 mM, Advantage? Polymerase Mix (Clontech) 1/50 volume ゝ 10-fold concentrated Buffer 1/10 volume. The cycle for amplification is 94X · 5 seconds for both PCRs,
60°C · 25秒、 72°C · 120秒のサイクルを 40回繰り返した後、 72°Cで 10分保温した c 反応物を 1.2% Seakein GTG Agarose (宝酒造) で電気泳動し、 ェチジゥムブロマ ィドで染色した時に見える 2000 bp付近のバンドを GeneClean Spin kit (バイオ 101) で抽出し、 T0P0 TA cloning kit (インビトロジェン社) を用いてプラスミ ドベクタ一 pcr4- T0P0へサブクローニングした後、 大腸菌 T0P10へ導入した。 生じ た形質転換体から QIA prep8 mini prep (キアゲン社) を用いてプラスミド DNAを 精製した。 '塩基配列決定のための反応は、 BigDye Terminator Cycle Sequence Ready Reaction Kit (パーキンエルマ一社) を用いて行なった。 蛍光式自動シー ケンサ一を用いて解読した結果、 ネコ SLTの C末端領域を含む配列番号: 7で 表される塩基配列が得られた。 実施例 3 After repeating the cycle of 60 ° C for 25 seconds and 72 ° C for 120 seconds for 40 times, the c- reactant incubated at 72 ° C for 10 minutes was electrophoresed on 1.2% Seakein GTG Agarose (Takara Shuzo), and then purified with ethidium bromide. A band around 2000 bp, which was visible when stained with, was extracted with a GeneClean Spin kit (Bio 101), subcloned into a plasmid vector pcr4-T0P0 using a T0P0 TA cloning kit (Invitrogen), and then introduced into E. coli T0P10. Plasmid DNA was isolated from the resulting transformant using QIA prep8 mini prep (Qiagen). Purified. 'The reaction for base sequence determination was performed using the BigDye Terminator Cycle Sequence Ready Reaction Kit (PerkinElmer). As a result of decoding using a fluorescent automatic sequencer, a base sequence represented by SEQ ID NO: 7 including the C-terminal region of cat SLT was obtained. Example 3
5 ' 一 RACE法によるネコ SLT遺伝子の 5' 側配列の取得  Acquisition of 5 'side sequence of cat SLT gene by 5'-RACE method
5'- RACE用の铸型は Marathon ready cDNA Amplif ication'kitを用いてネコ全脳 poly(A)+ RNAから作製した。 RACE PCR反応に用いたプライマ一セットは、 1回目 の PCR反応では Marathon ready cDNA Amplification kit用 adaptor primer 1とフ。 ライマー 1 (配列番号: 8) 、 2回目の PCR反応では Marathon ready cDNA Amplification kit用 adaptor primer 2とプライマー 2 (配列番号: 9) を用い た。 反応は、 逆転写した cDNAのうち mRNA 2 ng相当分を铸型に 20 /i 1の液量で行な つた。 反応液の組成は、 プライマー濃度 0.5 M、 dNTP混合液 0.2 mM、 Advantage 2 Polymerase Mix (クロンテック社) 1/50 volume, 10倍濃縮 Buffer 1/10 volume とした。 増幅のためのサイクルは、 どちらの PCRも 94°C · 5秒、 66°C · 120秒の'サ イクルを 10回、 94°C · 5秒、 60°C · 20秒、 72°C · 120秒のサイクルを 25回繰り返し た後、 72°Cで 10分保温した。 反応物を 1.2% Seakem GTG Agarose (宝酒造) で、 電気泳動し、 ェチジゥムブ口マイドで染色した時に見える 500. bp付近のバンドを GeneClean Spin kit (バイオ 101) で抽出し、 T0P0 TA cloning kit (インビトロ ジェン社) を用いてプラスミドベクタ一 pcr4-T0P0へサブクロ一ニングした後、 大腸菌 ΤΟΠ0へ導入した。 生じた形質転換体から QIA prep8 mini prep (キアゲン 社) を用いてプラスミド DNAを精製した。 塩基配列決定のための反応は、 BigDye Terminator Cycle Sequence Ready Reaction Kit (パーキンエルマ一社) を用い て行なった。 蛍光式自動シーケンサ一を用いて解読した結果、 ネコ SLTの N末 端領域を含む配列番号: 10で表される塩基配列が得られた。 実施例 4 Type I for 5'-RACE was prepared from feline whole brain poly (A) + RNA using Marathon ready cDNA Amplification'kit. The set of primers used for the RACE PCR reaction included adapter primer 1 for the Marathon ready cDNA Amplification kit in the first PCR reaction. Primer 2 (SEQ ID NO: 9) and primer 2 (SEQ ID NO: 9) for Marathon ready cDNA Amplification kit were used in the second PCR reaction. The reaction was carried out in the amount of 20 ng of the reverse transcribed cDNA corresponding to 2 ng of mRNA at a volume of 20 / i1. The composition of the reaction solution was primer concentration 0.5 M, dNTP mixture 0.2 mM, Advantage 2 Polymerase Mix (Clontech) 1/50 volume, and 10-fold concentrated Buffer 1/10 volume. The cycle for amplification is 10 cycles of 94 ° C for 5 seconds, 66 ° C for 120 seconds for both PCRs, 94 ° C for 5 seconds, 60 ° C for 20 seconds, 72 ° C for After repeating the cycle of 120 seconds 25 times, it was kept at 72 ° C for 10 minutes. The reaction product was electrophoresed on a 1.2% Seakem GTG Agarose (Takara Shuzo), and a band around 500 bp, which was visible when stained with ethidium amide, was extracted with the GeneClean Spin kit (Bio 101). After subcloning into plasmid vector pcr4-T0P0 using E. coli, Escherichia coli E0 was introduced. Plasmid DNA was purified from the resulting transformant using QIA prep8 mini prep (Qiagen). The reaction for base sequence determination was performed using BigDye Terminator Cycle Sequence Ready Reaction Kit (Perkin Elmer). As a result of decoding using a fluorescent automatic sequencer, a base sequence represented by SEQ ID NO: 10 including the N-terminal region of cat SLT was obtained. Example 4
ネコ SLT全長配列の取得 5' -および 3'- RACE法の結果から予想されるネコ S LTの全長を含む配列を得るた め forward primerとして 5,非翻訳領域にプライマー 1 (配列番号: 1 1) 、 およ び reverse primerとして 3,非翻訳領域にプライマー 2 (配列番号: 12) を設定 して PCR反応を行なった。 反応は、 逆転写した cDNAのうち mRNA 2 ng相当分を錶型 に 20^1の液量で行なった。 反応液の組成は、 プライマー濃度 0.5 M、 dNTP混合 液 0.2 mM、 Z-Taq (TaKaRa) 1/50 volume, 10倍濃縮 Buffer 1/10 volumeとした。 増幅のためのサイクルは、 94°Cで 15秒保温した後、 98t: · 1秒、 64°C · 15秒、 72°C · 20秒のサイクルを 40回繰り返した後、 72°Cで 2分保温した。 反応物を 1.2% Seakein GTG Agarose (宝酒造) で電気泳動し、 ェチジゥムプロマイドで染色した 時に見える 1200 bp付近のバンドを GeneClean Spin kit (バイオ 101) で抽出し、 Eukaryotic T0P0 TA cloning kit (インビトロジェン社) を用いてプラスミドべ クタ一 pcDNA3.1/V5/His- T0P0へサブクローニングした後、 大腸菌 TOP10へ導入し た。 生じた形質転換体から QIA prep8 mini prep (キアゲン社) を用いてプラス ミド DMを精製した。 塩基配列決定のための反応は、 BigDye Terminator Cycle Sequence Ready Reaction Kit (パ一キンエルマ一社) を用いて行なった。 蛍光 式自動シーケンサーを用いて解読した結果、 配列番号: 39に示す塩基配列が得 られた。 この配列にはネコ SLTの全アミノ酸配列 (配列番号: 14) をコード する塩基配列 (配列番号: 1 3) が含まれていたので、 このプラスミドを用いて 大腸菌 TOP10を形質転換し、 Escherichia coli TOPlO/pcDNA-catSLTを得た。 実施例 5 Acquisition of full-length cat SLT sequence In order to obtain a sequence containing the full length of cat SLT predicted from the results of the 5'- and 3'-RACE methods, 5 was used as the forward primer, primer 1 (SEQ ID NO: 11) was added to the untranslated region, and reverse PCR was performed with 3 as a primer and primer 2 (SEQ ID NO: 12) in the untranslated region. The reaction was performed in the form of type II with a volume of 20 ^ 1 corresponding to 2 ng of mRNA of the reverse transcribed cDNA. The composition of the reaction solution was primer concentration 0.5 M, dNTP mixture 0.2 mM, Z-Taq (TaKaRa) 1/50 volume, and 10-fold concentrated Buffer 1/10 volume. The cycle for amplification is as follows: after holding at 94 ° C for 15 seconds, 98t: 1 second, 64 ° C, 15 seconds, 72 ° C Keep warm for a minute. The reaction product was electrophoresed on 1.2% Seakein GTG Agarose (Takara Shuzo), and a band around 1200 bp, which was visible when stained with ethidium bromide, was extracted using the GeneClean Spin kit (Bio 101) and the Eukaryotic T0P0 TA cloning kit (Invitrogen) After subcloning into plasmid vector pcDNA3.1 / V5 / His-T0P0 using Escherichia coli, E. coli TOP10 was introduced. Plasmid DM was purified from the resulting transformant using QIA prep8 mini prep (Qiagen). The reaction for base sequence determination was performed using a BigDye Terminator Cycle Sequence Ready Reaction Kit (Pakkin Elma). As a result of decoding using a fluorescent automatic sequencer, the nucleotide sequence represented by SEQ ID NO: 39 was obtained. Since this sequence contained the nucleotide sequence (SEQ ID NO: 13) encoding the entire amino acid sequence of cat SLT (SEQ ID NO: 14), Escherichia coli TOP10 was transformed with this plasmid to transform Escherichia coli TOP10. / pcDNA-catSLT was obtained. Example 5
3 ' —RACE法によるィヌ SLT遺伝子の 3' 側配列の取得  3 '— RACE method to obtain 3' sequence of canine SLT gene
ィヌ前頭葉より Isogen (二ツボンジーン社) により total RNAを調製後、  After preparing total RNA from the canine frontal lobe with Isogen (Futtsubon Gene),
Oligotex (dT)30 (宝酒造) を用いて poly(A)+ RNA画分を調製した。 3'_RACE用の铸 型はィヌ前頭葉 poly (A)+ RNAをもとに SMART RACE cDNA Amplification kitを用い て作製した。 RACE PCR反応に用いたプライマーセットは、 4回目の PCR反応では、 kit添付の Universal Primer Mix とプライマー 1 (配列番号: 15) 、 2回目の PCR反応では、 kit添付の Nested Universal Primerとプライマ一 2 (配列番号: 16) を用いた。 反応は、 逆転写した cDNAのうち mRNA 20 ng相当分を錶型に 20 1の液量で行なった。 反応液の組成は、 プライマー濃度 0.5 /M、 dNTP混合液 0.2 mM、 Advantage2 Polymerase Mix (クロンテック社) 1/50 volume^ 10倍濃縮 Buffer 1/10 volumeとした。 増幅のためのサイクルは、 どちらの PCRも 94°C .5秒、 72°C · 90秒のサイクルを 3回、 94°C · 5秒、 70°C · 90秒のサイクルを 3回、 94°C · 5 秒、 68°C · 90秒のサイクルを 40回繰り返した後、 72°Cで 10分保温した。 '反応物を 1.2% Seakem GTG Agarose (宝酒造) で電気泳動し、 ェチジゥムブ口マイドで染 色した時に見える 900 bp付近のバンドを GeneClean Spin kit (バイオ 101)で抽出 し、 T0P0 TA cloning kit (インビトロジェン社) を用いてプラスミドベクタ一 pc -TOPOへサブクローニングした後、 大腸菌 T0P10へ導入した。 生じた形質転換 体から QIA prep8 mini prep (キアゲン社) を用いてプラスミド DNAを精製した。 塩基配列決定のための反応は、 BigDye Terminator Cycle Sequence Ready A poly (A) + RNA fraction was prepared using Oligotex (dT) 30 (Takara Shuzo). A type for 3'_RACE was prepared using SMART RACE cDNA Amplification kit based on canine frontal lobe poly (A) + RNA. The primer set used for the RACE PCR reaction was the Universal Primer Mix and primer 1 (SEQ ID NO: 15) attached to the kit in the fourth PCR reaction, and the Nested Universal Primer and primer 1 in the kit for the second PCR reaction. (SEQ ID NO: 16) was used. The reaction was performed by converting 20 ng of reverse-transcribed cDNA into Performed with 1 liquid volume. The composition of the reaction solution was primer concentration 0.5 / M, dNTP mixture 0.2 mM, Advantage2 Polymerase Mix (Clontech) 1/50 volume ^ 10-fold concentrated Buffer 1/10 volume. The cycles for amplification were as follows: 94 ° C for 5 seconds, 72 ° C for 90 seconds, three cycles for each PCR, 94 ° C for 5 seconds, 70 ° C for 90 seconds, three cycles. After repeating the cycle of ° C for 5 seconds and 68 ° C for 90 seconds 40 times, it was kept at 72 ° C for 10 minutes. 'The reaction product was electrophoresed on 1.2% Seakem GTG Agarose (Takara Shuzo), and a band around 900 bp, which was visible when stained with ethidium bromide, was extracted with the GeneClean Spin kit (Bio 101). ) Was used for subcloning into a plasmid vector pc-TOPO, and then introduced into E. coli T0P10. Plasmid DNA was purified from the resulting transformant using QIA prep8 mini prep (Qiagen). The reaction for base sequence determination is BigDye Terminator Cycle Sequence Ready
Reaction Kit (パ一キンエルマ一社) を用いて行なった。 蛍光式自動シーケンサ 一を用いて解読した結果、 ィヌ SLTの C末端領域を含む配列番号: 17で表さ れる塩基配列が得られた。 実施例 6 The reaction was performed using Reaction Kit (Pakkin Elma). As a result of decoding using a fluorescent automatic sequencer, a base sequence represented by SEQ ID NO: 17 including the C-terminal region of canine SLT was obtained. Example 6
5 ' 一 RACE法によるィヌ SLT遺伝子の 5 ' 側配列の取得  Acquisition of 5 'sequence of canine SLT gene by 5'-RACE method
5,- RACE用の铸型は Marathon ready cDNA Amplification kitを用いてィヌ前頭 葉 poly (A)+RNAから作製した。 RACE PCR反応に用いたプライマーセットは、 1回 目の PCR反応では、 Marathon ready cDNA Amplification kit用 adaptor primer 1 とプライマー 1 (配列番号: 18) 、 2回目の PCR反応では、 Marathon ready cDNA Amplification kit用 adaptor primer 2とプライマ一 2 (配列番号: 1 9) を用いた。 反応は、 逆転写した cDNAのうち mRNA 2 ng相当分を铸型に 20 1の液量 で行なった。 反応液の組成は、 プライマー濃度 0.5 M、 dNTP混合液 0.2 mM、 5, -Type II for RACE was prepared from canine frontal lobe poly (A) + RNA using Marathon ready cDNA Amplification kit. The primer set used for the RACE PCR reaction was the adapter primer 1 and primer 1 (SEQ ID NO: 18) for the Marathon ready cDNA Amplification kit in the first PCR reaction, and the Marathon ready cDNA Amplification kit in the second PCR reaction. The adaptor primer 2 and the primer 1 (SEQ ID NO: 19) were used. The reaction was carried out in the form of type II with a volume of 201, corresponding to 2 ng of mRNA of the reverse-transcribed cDNA. The composition of the reaction solution was primer concentration 0.5 M, dNTP mixture 0.2 mM,
Advantage2 Polymerase Mix (クロンテック社) 1/50 volume^ 10倍濃縮 Buffer 1/10 volumeとした。 増幅のためのサイクルは、 どちらの PCRも 94°C · 5秒、 Advantage2 Polymerase Mix (Clontech) 1/50 volume ^ 10 times concentrated Buffer 1/10 volume. The cycle for amplification was 94 ° C for 5 seconds for both PCRs.
66°C · 120秒のサイクルを 10回、 94で · 5秒、 60°C · 20秒、 72で · 120秒のサイク ルを 25回繰り返した後、 72°Cで 10分保温した。 反応物を 1.2% Seakem GTG A cycle of 66 ° C for 120 seconds was repeated 10 times, a cycle of 94 for 5 seconds, a cycle of 60 ° C for 20 seconds, and a cycle of 72 for 120 seconds was repeated 25 times, followed by incubation at 72 ° C for 10 minutes. Reactants to 1.2% Seakem GTG
Agarose (宝酒造) で電気泳動し、 ェチジゥムブロマイドで染色した時に見える 400 bp付近のバンドを GeneClean Spin kit (バイオ 101) で抽出し、 TOPO TA cloning kit (インビトロジェン社) を用いてプラスミドベクター pcr4-T0P0へサ ブクローニングした後、 大腸菌 TOP10へ導入した。 生じた形質転換体から QIA prep8 mini prep (キアゲン社) を用いてプラスミド DNAを精製した。 塩基配列決 疋のための反 j¾、は、 BigDye Terminator Cycle Sequence Ready Reaction KitAppears when electrophoresed on Agarose (Takara Shuzo) and stained with ethidium bromide A band near 400 bp was extracted with a GeneClean Spin kit (Bio 101), subcloned into a plasmid vector pcr4-T0P0 using a TOPO TA cloning kit (Invitrogen), and then introduced into E. coli TOP10. Plasmid DNA was purified from the resulting transformant using QIA prep8 mini prep (Qiagen). The anti-sequence for nucleotide sequencing is the BigDye Terminator Cycle Sequence Ready Reaction Kit
(パーキンエルマ一社) を用いて行なった。 蛍光式自動シーケンサーを用いて解 読した結果、 ィヌ SLTの N末端領域を含む配列番号: 20で表される塩基配列 が得られた。 ' 実施例 7 (PerkinElmer). As a result of decoding using a fluorescent automatic sequencer, a nucleotide sequence represented by SEQ ID NO: 20 including the N-terminal region of canine SLT was obtained. '' Example 7
ィヌ SLT全長配列の取得1 Dog SLT full length sequence acquisition 1
5,-および 3'- RACE法の結果から予想されるィヌ SLTの全長を含む配列を得るため forward primerとして 5'非翻訳領域にプライマー 1 (配列番号: 2 1) : および reverse primerとして 3'非翻訳領域にプライマ一 2 (配列番号: 22) を設定し て PCR反応を行なった。 反応は、 逆転写した cDNAのうち mRNA 2 ng相当分を铸型に 20 1の液量で行なった。 反応液の組成は、 プライマー濃度 0.5 M、 dNTP混合液 0.2禮、 Z-Taq (TaKaRa) 1/50 volume, 10倍濃縮 Buffer 1/10 volumeとした。 増 幅のためのサイクルは、 94°Cで 15秒保温した後、 98°C · 1秒、 64°C · 15秒、 72°C · 20秒のサイクルを 40回繰り返した後、 72°Cで 2分保温した。 反応物を 1.2% Seakem GTG Agarose (宝酒造) で電気泳動し、 ェチジゥムブ口マイドで染色した 時に見える 1200 bp付近のバンドを GeneClean Spin kit (バイオ 101) で抽出し、 Eukaryotic T0P0 TA cloning kit (インビトロジェン社) を用いてプラスミドべ クタ一 PCDNA3.1/V5/His- T0P0へサブクロ一ニングした後、 大腸菌 T0P10へ導入し た。 生じた形質転換体から QIA prep8 mini prep (キアゲン社) を用いてプラス ミド DNAを精製した。 塩基配列決定のための反応は、 BigDye Terminator Cycle Sequence Ready Reaction Kit (パ一キンエルマ一社) を用いて行なった。 蛍光 式自動シーケンサ一を用いて解読した結果、 配列番号: 40 に示す塩基配列が 得られた。 この配列にはィヌ SLTの全アミノ酸配列 (配列番号: 24) をコ一 ドする塩基配列 (配列番号: 23) が含まれていたので、 このプラスミドで大腸 菌 TOP10を形質転換して Escherichia coli TOPlO/pcDNA- dogSLTを得た。 実施例 8 In order to obtain a sequence containing the full length of canine SLT predicted from the results of the 5,-and 3'-RACE methods, primer 1 (SEQ ID NO: 21) was added to the 5 'untranslated region as a forward primer, and 3 'PCR was performed by setting Primer 2 (SEQ ID NO: 22) to the untranslated region. The reaction was carried out in the form of type II with a volume of 201, corresponding to 2 ng of mRNA of the reverse-transcribed cDNA. The composition of the reaction solution was a primer concentration of 0.5 M, a dNTP mixed solution of 0.2 mL, a Z-Taq (TaKaRa) 1/50 volume, and a 10-fold concentrated Buffer 1/10 volume. The cycle for amplification is as follows: after holding at 94 ° C for 15 seconds, repeat the cycle of 98 ° C for 1 second, 64 ° C for 15 seconds, 72 ° C for 20 seconds 40 times, and then for 72 ° C. For 2 minutes. The reaction product was electrophoresed on a 1.2% Seakem GTG Agarose (Takara Shuzo), and a band around 1200 bp, which was visible when stained with ethidium umide, was extracted with the GeneClean Spin kit (Bio 101). Eukaryotic T0P0 TA cloning kit (Invitrogen) After subcloning into the plasmid vector PCDNA3.1 / V5 / His-TOP0 using Escherichia coli, Escherichia coli T0P10 was introduced. Plasmid DNA was purified from the resulting transformant using QIA prep8 mini prep (Qiagen). The reaction for base sequence determination was carried out using BigDye Terminator Cycle Sequence Ready Reaction Kit (Pakkin Elma). As a result of decoding using a fluorescent automatic sequencer, the base sequence represented by SEQ ID NO: 40 was obtained. This sequence contained the nucleotide sequence (SEQ ID NO: 23) encoding the entire amino acid sequence of canine SLT (SEQ ID NO: 24). Bacterium TOP10 was transformed to obtain Escherichia coli TOPlO / pcDNA-dogSLT. Example 8
ネコ S L T発現 C H O細胞の作製  Preparation of feline SLT expressing CHO cells
実施例 4で作製したネコ SLTの全長アミノ酸配列をコードし、 5'側に Sal I認識 配列が付加し、 また 3'側に Spe I認識配列を付加した遺伝子が導入されたプラスミ ドによって形質転換された大腸菌のクローンより Plasmid Midi Kit (キアゲン) を用いてプラスミドを調製し、 制限酵素 Sal Iおよび Spe Iで切断してインサート 部分を切り出した。 インサート DNAは電気泳動後、 ァガ口一スゲルから力ミソリ で切り出し、 次に細片化、 フエノール抽出、 フエノール 'クロ口ホルム抽出、 ェ 夕ノール沈殿を行なって回収した。 このインサート DNAを Sal Iおよび Spe Iで切 断した動物細胞発現用ベクタープラスミ FpAKKO-lllH (Hinuma, S.ら、 Biochim. Biophys. Acta、 1219卷、 251-259頁、 1994年、 記載の pAKKOl.11Hと同一のベクタ —プラスミド) に加え、 T4ライゲ一ス (宝酒造) を用いてライゲーシヨンを行な い、 タンパク発現用プラスミド pAKKO-catSLTを構築した。  Transformation with a plasmid encoding the full-length amino acid sequence of the feline SLT prepared in Example 4 with a SalI recognition sequence added to the 5 'side and a SpeI recognition sequence added to the 3' side. Plasmids were prepared from the cloned Escherichia coli using the Plasmid Midi Kit (Qiagen) and cut with restriction enzymes Sal I and Spe I to cut out the insert. After the electrophoresis, the insert DNA was cut out from an agarose gel with a force razor, and then recovered by performing fragmentation, phenol extraction, phenol-chloroform extraction, and ethanol precipitation. This insert DNA was cut with Sal I and Spe I for animal cell expression vector Plasmid FpAKKO-llH (Hinuma, S. et al., Biochim. Biophys. Acta, Vol. 1219, pp. 251-259, 1994, pAKKOl. In addition to ligation using T4 ligase (Takara Shuzo) in addition to the same vector—plasmid as 11H, a plasmid pAKKO-catSLT for protein expression was constructed.
pAKKO-catSLTで形質転換した Escherichia coli TOP 10 co即 etent cell (イン ビトロジェン社) を培養後、 Plasmid Midi Kit (キアゲン) を用いて pAKKO - catSLTのプラスミド DNAを調製した。 これを CHO dhfr細胞 5000000 cells/ 150 1 O tiMEM (ギブコ社) 細胞懸濁液に 50 g/mlになるように加え、 ジーンパルサ一 (バイオラッド社) により 240V 960 Fのパルスをかけることで細胞に導入した t 10%ゥシ胎児血清を含む ΜΕΜα培地で 1日間培養した後、 継代し、 選択培地である 10 %透析ゥシ胎児血清を含む核酸不含 MEM a培地で培養した。 選択培地中で増殖 してくるネコ SLT発現 CH0細胞である形質転換細胞を選択した。 実施例 9 After culturing Escherichia coli TOP10 co-immediate etent cells (Invitrogen) transformed with pAKKO-catSLT, plasmid DNA of pAKKO-catSLT was prepared using Plasmid Midi Kit (Qiagen). This was added to the CHO dhfr cell 500,000 cells / 150 1 O tiMEM (Gibco) cell suspension at 50 g / ml, and 240 V 960 F pulse was applied to the cells by Genepulsa (Bio-Rad). after culturing for 1 day ΜΕΜα medium containing the introduced t 10% © shea calf serum and passaged and cultured in a nucleic acid-free MEM a medium containing 10% dialyzed © shea calf serum selective medium. Transformed cells that were feline SLT-expressing CH0 cells growing in the selection medium were selected. Example 9
FL I PRを用いた MCHによるネコ S LT発現 CHO細胞の細胞内 C aィォ ン濃度上昇活性の測定  Measurement of intracellular Ca ion concentration increasing activity of cat SLT expressing CHO cells by MCH using FLIPR
実施例 8で得られたネコ SLT発現 CH0細胞の、 MCH (配列番号: 2 5、 (株)ぺプ チド研究所) による細胞内 Caイオン濃度上昇活性の測定を、 FLIPR (モレキュラー デバイス社)を用いて行なった。 Measurement of the intracellular Ca ion concentration increasing activity of the feline SLT-expressing CH0 cells obtained in Example 8 by MCH (SEQ ID NO: 25, Peptide Research Laboratories) was performed using FLIPR (Molecular Device Company).
CHO/catSLT細胞を 15 x 104 cells/mlとなるように 10%透析ゥシ胎児血清を含む DMEMに懸濁し、 FLIPR用 96穴プレート(Black plate clear bottom, コ一スター 社)に分注器を用いて各ゥエルに 200 1ずつ植え込み (3.0X104 cells/200 l/ゥ エル) 、 5% C02インキュべ一ター中にて 37°Cでー晚培養した後、 アツセィに用い た(以後このプレートを細胞プレートと言う)。 HANKS'/HBSS (二ッスィハンクス 2CHO / catSLT cells are suspended in DMEM containing 10% dialyzed fetal serum to a concentration of 15 x 10 4 cells / ml and dispensed into a 96-well plate for FLIPR (Black plate clear bottom, Costar). 200 1 by implantation into the Ueru using (3.0X10 4 cells / 200 l / © El), 5% C0 2 was 37 ° C De晚cultured at incubator base one coater in was used to Atsusi (hereinafter This plate is called a cell plate). HANKS '/ HBSS (Nissy Hanks 2
(日水製薬株式会社) 9.8 g、 炭酸水素ナトリウム 0.35 g、 HEPES 4.77 g、 6 M水酸化ナトリウム溶液で PH7.4に合わせた後、 フィルタ" 1滅菌処理) 20 ml、 250 mM Probenecid 200/zK ゥシ胎児血清(FBS) 200 1を混合したものに、 Fluo 3-A (同仁化学研究所) 2バイアル(50 )をジメチルスルフォキサイド 40 lおよび 20% Pluronic acid (モレキュラープロ一ブ社) 40 1に溶解して加え て混和後、 8連ピペットを用いて培養液を除いた細胞プレートに各ゥエル 100 ずつ分注した後、 5% C02インキュベータ一中にて 37°Cで 1時間インキュベートし、 細胞に色素をロードした。 FLIPR用 96穴プレ一ト(V- Bottomプレート、 コースター 社)の各ゥエルに 2.5 mM Probenecid, 0.05% BSAを含む HAMS,/HBSS 150 lを入 れ、 さらに種々の濃度の MCHを添加してサンプルプレートを調製した。 細胞プレ ートの色素口一.ディング終了後、 HANKS'/HBSSに 2.5 mM Probenecidを加えた洗浄 バッファ一でプレートウォッシャー (モレキュラーデバイス社) を用いて細胞プ レートを 4回洗浄し、 洗浄後 100 1の洗浄バッファ一を残した。 この細胞プレー トとサンプルプレー卜を FLIPRにセットしアツセィを行なった (FLIPRにより、 サ ンプルプレー卜から のサンプルが細胞プレートへと移される) 。 (Nissui Pharmaceutical Co., Ltd.) 9.8 g, sodium hydrogencarbonate 0.35 g, HEPES 4.77 g, adjusted to PH7.4 with 6 M sodium hydroxide solution, filter " 1 sterilized" 20 ml, 250 mM Probenecid 200 / zKゥ Two vials (50) of Fluo 3-A (Dojindo Laboratories) are mixed with fetal serum (FBS) 200 1 and 40 l of dimethyl sulfoxide and 20% Pluronic acid (Molecular Probes) 40 after mixing added dissolved in 1, after dispensed at each Ueru 100 the cell plate except the culture solution using an 8 channel pipette, incubated for 1 hour at 37 ° C for at 5% C0 2 incubator in one Each well of a 96-well plate for FLIPR (V-Bottom plate, Coaster) was loaded with 150 l of 2.5 mM Probenecid, HAMS / HBSS containing 0.05% BSA, and various other components. A sample plate was prepared by adding MCH at a concentration of 1. The color of the cell plate After finishing the washing, wash the cell plate 4 times using a plate washer (Molecular Devices) with HANKS '/ HBSS and 2.5 mM Probenecid using a washing buffer. The cell plate and the sample plate were set on the FLIPR and assembled (the sample from the sample plate was transferred to the cell plate by the FLIPR).
その結果、 MCHは濃度依存的にネコ SLT発現 CH0細胞の細胞内 Caイオン濃度を上 昇させることが示された (図 1) 。 なお、 細胞内 Caイオン濃度の上昇は、 Caによ つて生じる細胞にロードされた色素の蛍光の上昇によって示される。  The results showed that MCH increased the intracellular Ca ion concentration of feline SLT-expressing CH0 cells in a concentration-dependent manner (Fig. 1). The increase in intracellular Ca ion concentration is indicated by the increase in fluorescence of the dye loaded on the cell caused by Ca.
, 実施例 1 0  , Example 10
ネコ脳各部位でのネコ S L T遺伝子の発現分布および T a qMa n PCR法 を用いた発現量の定量  Distribution of Feline SLT Gene Expression in Feline Brain and Quantification of Its Expression Using TaqMan PCR
ネコ全脳から、 前頭葉、 側頭葉、 後頭葉、 頭頂葉、 橋、 小脳、 延髄、 海馬、 扁 桃体、 視床、 脳梁、 大脳脚、 脊髄、 黒質、 尾状核、 視床下部を取り出し、 ISOGEN (二ツボンジーン社) を用いて total RNA画分を調製した。 得られた RNAは、 プロ ティナ一ゼ K (インビトロジェン社)消化により RNaseを分解後、 Message Clean Kit (GenHunter社) を用いてゲノム DNAを DNase I消化した後、 各部位の total RNA l xgを铸型に、 Superscript II reversetranscr iptase (インビ卜ロジェン 社) を用い、 添付のマニュアルにしたがってランダムプライマーを用いて逆転写 を行ない、 cDNAを作製した。 得られた total RNA 25 ng相当の逆転写産物または 後に述べるようにして作製した標準 cDNA、 1 X Universsal PCR Master Mix (P Eバイオシステムズ社) 、 配列番号: 3 2で表されるプライマ一 1および配列番 号: 33で表されるプライマ一 2、 各 100 nM、 および配列番号: 34 (Fam - acaggtact tggctctcgtccaaccat t'tc-Tamra;目 ΰ列中、 Famは 6-carboxy - fluorescein を、 Tamraは 6 - carboxy-tetramethyト rhodamine をそれぞれ示す)で表される TaqManプローブ 100 nMを含む反応混合液 25^ 1について ABI PRISM 7700 Sequence Detector (P Eバイオシステムズ社) を用いて P C Rを行なった。 PCRは、 50°C · 2分、 95°C · 10分で処理後、 95で · 15秒、 60°C · 60秒のサイクルを 40回繰 り返すことにより行なった。 From cat whole brain, frontal lobe, temporal lobe, occipital lobe, parietal lobe, pons, cerebellum, medulla oblongata, hippocampus, flat The amygdala, thalamus, corpus callosum, cerebral peduncle, spinal cord, substantia nigra, caudate nucleus, and hypothalamus were removed, and a total RNA fraction was prepared using ISOGEN (Futtsubon Gene). The obtained RNA was digested with RNase by digestion with proteinase K (Invitrogen), DNase I digested genomic DNA using the Message Clean Kit (GenHunter), and total RNA lxg at each site was extracted. Reverse transcription was carried out using Superscript II reversetranscriptase (Invitrogen) as a template and random primers according to the attached manual to produce cDNA. A reverse transcript equivalent to 25 ng of the obtained total RNA or a standard cDNA prepared as described later, 1 × Universsal PCR Master Mix (PE Biosystems), the primer 1 represented by SEQ ID NO: 32 and the sequence Primer 1 represented by No. 33, 100 nM each, and SEQ ID NO: 34 (Fam-accaggtact tggctctcgtccaaccat t'tc-Tamra; in the column, Fam is 6-carboxy-fluorescein, Tamra is 6- PCR was carried out using ABI PRISM 7700 Sequence Detector (PE Biosystems) for a reaction mixture 25 ^ 1 containing 100 nM of a TaqMan probe represented by carboxy-tetramethy and rhodamine. PCR was performed by treating the cells at 50 ° C for 2 minutes and 95 ° C for 10 minutes, and then repeating the cycle of 95 for 15 seconds and 60 ° C for 60 seconds 40 times.
標準 cDNAは、 実施例 4で得られたプラスミド pcDNA-catSLT 50 pgを铸型に、 プ ライマー 1および 2 (配列番号: 32および配列番号: 33) を用いて 200 xlの 液量で PCR反応を行ない、 作成した。 組成は、 プライマー 1および 2、 0.5 M、 2.5 mM MgCl2、 dNTP 0.2 niM, Am liTaq Gold (パ一キンエルマ一社) 1/100 volume 、 10倍濃縮 Ampl iTaq Gold Buffer 1/10 volumeで行った。 反応は、 95°C で 10分保温した後、 95°C · 15秒、 60で · 15秒、 72°C · 10秒のサイクルを 40回繰り 返した。 反応液を PCR purification kit (キアゲン社) により、 増幅産物を精製 し、 260 nmの吸光度を測定して濃度を算出して正確なコピー数を算出した後、 蒸 留水で希釈し、 1コピーから lxlO7コピーの標準 cDNA溶液を調製した。 また、The standard cDNA was prepared by PCR using the primers 1 and 2 (SEQ ID NO: 32 and SEQ ID NO: 33) in the form of 200 xl using primers 1 and 2 (SEQ ID NO: 32 and SEQ ID NO: 33) with 50 pg of the plasmid pcDNA-catSLT obtained in Example 4. Done and created. The composition was performed using primers 1 and 2, 0.5 M, 2.5 mM MgCl 2 , dNTP 0.2 niM, AmliTaq Gold (Pakinkin Elmer) 1/100 volume, and 10-fold concentrated Ampl iTaq Gold Buffer 1/10 volume. The reaction was incubated at 95 ° C for 10 minutes, and then repeated at 95 ° C for 15 seconds, 60 at 15 seconds, and 72 ° C for 10 seconds 40 times. The amplification product is purified from the reaction mixture using the PCR purification kit (Qiagen), the absorbance at 260 nm is measured, the concentration is calculated, the exact copy number is calculated, and the copy is diluted with distilled water. A standard cDNA solution of 7 copies of lxlO was prepared. Also,
TaqMan PCR用プローブおよびプライマ一は Primer Express (Versionl.O) (P E バイオシステムズ社) により設計した。 TaqMan PCR probes and primers were designed using Primer Express (Version I.O) (PE Biosystems).
発現量は ABI PRISM 7700 SDSソフトウェアによって算出した。 リポーターの蛍 光強度が設定された値に達した瞬間のサイクル数を縦軸にとり、 また標準 cDNAの 初期濃度の対数値を横軸にとって標準曲線を作成した。 標準曲線より各逆転写産 物の初期濃度を算出し、 各部位の total RNA 25 ng当たりのネコ S L T遺伝子発 現量を求めた。 さらに各々のサンプルのネコ GAP DH遺伝子発現量をプライマ —3 (配列番号: 35) およびプライマー 4 (配列番号: 36) および配列番 号: 37に示すプローブ (Fam-ccaggagcgagatcccgcca-Tamura) を用いてネコ S LT遺伝子発現量の解析と同様にして TaqMan: PCR法により求め、 ネコ SLT遺伝 子発現量の補正を行なった。 The expression level was calculated using ABI PRISM 7700 SDS software. The number of cycles at the moment when the fluorescence intensity of the reporter reaches the set value is plotted on the vertical axis. A standard curve was created using the logarithmic value of the initial concentration as the horizontal axis. The initial concentration of each reverse transcription product was calculated from the standard curve, and the expression amount of the cat SLT gene per 25 ng of total RNA at each site was determined. Further, the expression level of the feline GAP DH gene in each sample was determined using the primers # 3 (SEQ ID NO: 35), primer 4 (SEQ ID NO: 36) and the probe (Fam-ccaggagcgagatcccgcca-Tamura) shown in SEQ ID NO: 37. The expression of the feline SLT gene was corrected by the TaqMan: PCR method in the same manner as in the analysis of the SLT gene expression level.
その結果、 ネコ GAPDH遺伝子に対するネコ SLT遺伝子の発現量は、 前頭 葉では 0.084%、 側頭葉では 0.32%、 後頭葉では 0.046%、 頭頂葉では 0.056%、 橋では 0.086%、 小脳では 0.012%、 延髄では 0.0075%、 海馬では 0.092%、 扁桃 体では 0.15%、 視床では 0.020%、 脳梁では 0.035%、 大脳脚では 0.016%、 脊髄 では 0.014%、 黒質では 0.018%、 尾状核では 0.039%、 視床下部では 0.012%であ つた。 以上より、 ネコ SLT遺伝子は、 ネコ脳において前頭葉、 側頭葉、 橋、 海 馬、 扁桃体において特に多く発現していることがわかった。  As a result, the expression level of the feline SLT gene relative to the feline GAPDH gene was 0.084% in the frontal lobe, 0.32% in the temporal lobe, 0.046% in the occipital lobe, 0.056% in the parietal lobe, 0.086% in the pons, and 0.012% in the cerebellum. 0.0075% in medulla oblongata, 0.092% in hippocampus, 0.15% in amygdala, 0.020% in thalamus, 0.035% in corpus callosum, 0.016% in cerebral peduncle, 0.014% in spinal cord, 0.018% in substantia nigra, 0.039% in caudate nucleus In the hypothalamus, it was 0.012%. From the above, it was found that the feline SLT gene was particularly highly expressed in the frontal lobe, temporal lobe, pons, hippocampus, and amygdala in the cat brain.
ネコ SLTのネコ脳各部位における発現量のグラフを 〔図 2〕 に示す。 実施例 1 1  A graph of the expression level of cat SLT in each part of the cat brain is shown in FIG. Example 1 1
ィヌ S L T動物細胞発現用ベクターの作製  Preparation of canine SLT animal cell expression vector
実施例 7で配列が確認された、 5' 側に Sal I認識配列が、 また 3' 側に Spe I認 識配列がそれぞれ付加されたィヌ SLTの全長アミノ酸配列をコードする遺伝子を 導入したプラスミド pcDNA- dogSLTによって大腸菌を形質転換した。 この大腸菌の クローンより Plasmid Midi Kit (キアゲン) を用いてプラスミドを調製し、 制限 酵素 Sal Iおよび Spe Iで切断してインサ一ト部分を切り出した。 インサート DNA は、 電気泳動後、 ァガロースゲルから力ミソリで切り出し、 次に細片化、 フエノ —ル抽出、 フエノール 'クロ口ホルム抽出、 エタノール沈殿を行なって回収しだ c このインサート DNAを Sal Iおよび Spe Iで切断した動物細胞発現用べクタ一ブラ スミド pAKKO - 111H (Biochim. Biophys. Acta, 1219卷、 251-259 頁、 1994年記載 の PAKK01.11Hと同一のベクタープラスミド) に加え、 T4ライゲ一ス (宝酒造) を 用いてライゲ一シヨンを行ない、 タンパク質発現用プラスミド pAKKO-dogSLTを構 築した。 この 1¾0-(1(½3 で大腸菌1"01)10 competent cel l (インビトロジェン 社) を形質転換し、 Escherichi a col i TOPlO/pAKKO-dogSLTを得た。 産業上の利用可能性 A plasmid into which a gene encoding the full-length amino acid sequence of canine SLT to which the sequence was confirmed in Example 7 and a Sal I recognition sequence was added on the 5 'side and a Spe I recognition sequence was added on the 3' side was added. E. coli was transformed with pcDNA-dogSLT. Plasmids were prepared from the E. coli clones using Plasmid Midi Kit (Qiagen) and cut with restriction enzymes Sal I and Spe I to cut out the insert. Insert DNA after electrophoresis, excised with a force Misori from Agarosugeru, then morselized, Fueno - Le extraction, phenol 'black hole Holm extraction, it is recovered by performing an ethanol precipitation c Sal This insert DNA I and Spe Vector plasmid for animal cell expression pAKKO-111H (Biochim. Biophys. Acta, Vol. 1219, pp. 251-259, same vector plasmid as described in PAKK01.11H described in 1994, 1994) cut with T4 Using Takara Shuzo to construct the protein expression plasmid pAKKO-dogSLT. I built it. This 1¾0- (1 (½3 Escherichia coli 1 "01 ) 10 competent cell (Invitrogen) was transformed to obtain Escherichia coli TOPlO / pAKKO-dogSLT.
本発明のタンパク質、 ポリヌクレオチド、 アンチセンスポリヌクレオチドおよ び抗体などは、 例えば食欲不振 (例、 神経性食欲不振症など) 、 食欲不振に伴う 貧血または低蛋白症、 微弱陣痛、 弛緩出血、 子宮復古不全、 乳汁うっ滞などの診 断マーカー等として有用である。 該タンパク質および本発明のペプチドなどを用 いるスクリーニング法により得られる該タンパク質のァゴニストは、 食欲 (摂 食) 増進剤、 食欲不振 (例、 神経性食欲不振症など) 、 食欲不振に伴う貧血また は低蛋白症、 微弱陣痛、 弛緩出血、 子宮復古不全、 乳汁うっ滞などの予防 ·治療 剤などとして、 アン夕ゴニストは、 例えば、 肥満症 (例、 悪性肥満細胞症、 外因 性肥満、 過インシュリン性肥満症、 過血漿性肥満、 下垂体性肥満、 減血漿性肥満 症、 甲状腺機能低下肥満症、 視床下部性肥満、 症候性肥満症、 小児肥満、 上半身 肥満、 食事性肥満症、 性機能低下性肥満、 全身性肥満細胞症、 単純性肥満、 中心 性肥満など) 、 摂食宂進症、 情動障害、 性機能障害、 過強陣痛、 強直性子宮収縮、 胎児仮死、 子宮破裂、 類管裂傷、 早産、 Prader-Wi l l i症候群、 糖尿病およびその 合併症 (例、 糖尿病性腎症、 糖尿病性網膜症、 糖尿病性神経障害など) 、 高血圧、 高脂血症、 冠状動脈硬化症、 痛風、. 呼吸器疾患 (Pickwi ck症候群、 睡眠時無呼吸 症候群) 、 脂肪肝、 不妊症、 変形性骨関節症など (特に抗肥満剤、 食欲 (摂食) 調節剤など) などの予防 ·治療剤などとして使用することができる。  The proteins, polynucleotides, antisense polynucleotides and antibodies of the present invention include, for example, anorexia (eg, anorexia nervosa), anemia or hypoproteinemia associated with anorexia, weak labor, lax hemorrhage, uterus It is useful as a marker for diagnosis of retro failure, milk stasis, etc. An agonist of the protein obtained by a screening method using the protein and the peptide of the present invention includes an appetite (feeding) enhancer, anorexia (eg, anorexia nervosa), anemia associated with anorexia, As a prophylactic / therapeutic agent for hypoproteinemia, weak labor, laxative bleeding, uterine remodeling failure, milk stasis, etc. Obesity, hyperplasmic obesity, pituitary obesity, hypoplasmic obesity, hypothyroid obesity, hypothalamic obesity, symptomatic obesity, childhood obesity, upper body obesity, dietary obesity, hypogonadism Obesity, systemic mastocytosis, simple obesity, central obesity, etc.), hyperphagia, affective disorders, sexual dysfunction, excessive labor, tonic contractions, fetal distress, uterine rupture , Vascular laceration, premature birth, Prader-Willi syndrome, diabetes and its complications (eg, diabetic nephropathy, diabetic retinopathy, diabetic neuropathy, etc.), hypertension, hyperlipidemia, coronary atherosclerosis, Prevention and treatment of gout, respiratory diseases (Pickwick syndrome, sleep apnea syndrome), fatty liver, infertility, osteoarthritis (especially antiobesity agents, appetite (feeding) regulators, etc.) It can be used as an agent.

Claims

請求 の 範 囲 The scope of the claims
1 . 配列 φ号: 4、 ,配列番号: 1 4または配列番号: 2 4で表わされるァミノ 酸配列と同一もしくは実質的に同一のァミノ酸配列を含有するタンパク質または その塩。 1. A protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 4,, SEQ ID NO: 14 or SEQ ID NO: 24, or a salt thereof.
2 . 配列番号: 4で表わされるアミノ酸配列からなるタンパク質またはその塩。 2. A protein comprising an amino acid sequence represented by SEQ ID NO: 4, or a salt thereof.
3 . 配列番号: 1 4で表わされるアミノ酸配列からなるタンパク質またはその 塩。 3. A protein comprising an amino acid sequence represented by SEQ ID NO: 14 or a salt thereof.
4. 配列番号: 2 4で表わされるァミノ酸配列からなるタンパク質またはその 4. A protein comprising an amino acid sequence represented by SEQ ID NO: 24 or a protein thereof
■ΙίΤΤΤ ■ ΙίΤΤΤ
5 . 請求項 1記載のタンパク質の部分べプチドまたはその塩。  5. A partial peptide of the protein according to claim 1, or a salt thereof.
6 . 請求項 1記載の夕ンパク質または請求項 5記載の部分べプチドをコードす るポリヌクレオチドを含有するポリヌクレオチド。 6. A polynucleotide comprising the polynucleotide encoding the protein according to claim 1 or the partial peptide according to claim 5.
7 . D NAである請求項 6記載のポリヌクレオチド。 7. The polynucleotide according to claim 6, which is DNA.
8 . 配列番号: 3、 配列番号: 1 3または配列番号: 2 3で表わされる塩基配 列を含有する請求項 7記載のポリヌクレオチド。 8. The polynucleotide according to claim 7, which comprises the base sequence represented by SEQ ID NO: 3, SEQ ID NO: 13 or SEQ ID NO: 23.
9 . 請求項 8記載のポリヌクレオチドを含有する組換えベクター。  9. A recombinant vector containing the polynucleotide according to claim 8.
1 0 . 請求項 9記載の組換えべクタ一で形質転換された形質転換体。  10. A transformant transformed with the recombinant vector according to claim 9.
1 1 . 請求項 1 0記載の形質転換体を培養し、 請求項 1記載のタンパク質また は請求項 5記載の部分ペプチドを生成、 蓄積せしめ、 これを採取することを特徴 とする請求項 1記載のタンパク質もしくは請求項 5記載の部分べプチドまたはそ の塩の製造法。  11. The transformant according to claim 10 is cultured to produce and accumulate the protein according to claim 1 or the partial peptide according to claim 5, and to collect the protein. A method for producing the protein of claim 1 or the partial peptide of claim 5 or a salt thereof.
1 2 . 請求項 1記載のタンパク質もしくは請求項 5記載の部分ペプチドまたは その塩を含有してなる医薬。  12. A medicament comprising the protein according to claim 1 or the partial peptide according to claim 5 or a salt thereof.
1 3 . 請求項 6記載のポリヌクレオチドを含有してなる医薬。 13. A medicine comprising the polynucleotide according to claim 6.
1 4 . 請求項 6記載のポリヌクレオチドを含有してなる診断薬。  14. A diagnostic agent comprising the polynucleotide according to claim 6.
1 5 . 請求項 1記載のタンパク質もしくは請求項 5記載の部分ペプチドまたは その塩に対する抗体。  15. An antibody against the protein according to claim 1, the partial peptide according to claim 5, or a salt thereof.
1 6 . 請求項 1記載のタンパク質のシグナル伝達を不活性化する中和抗体であ る請求項 1 5記載の抗体。 16. A neutralizing antibody that inactivates the signal transduction of the protein according to claim 1. 16. The antibody according to claim 15, wherein
1 7 . 請求項 1記載のタンパク質のシグナル伝達を活性化する抗体である請求 項 1 5記載の抗体。  17. The antibody according to claim 15, which is an antibody that activates signal transduction of the protein according to claim 1.
1 8 . 請求項 1 5記載の抗体を含有してなる診断薬。  18. A diagnostic agent comprising the antibody according to claim 15.
1 9 . 請求項 1 6記載の抗体を含有してなる医薬。  19. A pharmaceutical comprising the antibody according to claim 16.
2 0 . 請求項 1 7記載の抗体を含有してなる医薬。  20. A medicament comprising the antibody according to claim 17.
2 1 . 請求項 1記載のタンパク質または請求項 5記載の部分ペプチドまたはそ の塩および配列番号: 2 5で表されるアミノ酸配列と同一もしくは実質的に同一 のアミノ酸配列を含有するペプチドまたはその塩を用いることを特徴とする、 配 列番号: 2 5で表されるアミノ酸配列と同一もしくは実質的に同一のアミノ酸配 列を含有するべプチドと請求項 1記載の夕ンパク質またはその塩との結合性を変 化させる化合物またはその塩のスクリーニング方法。  21. The protein according to claim 1 or the partial peptide according to claim 5, or a salt thereof, and a peptide containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 25, or a salt thereof Wherein the peptide containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 25 and the protein or the salt thereof according to claim 1 are used. A method for screening a compound or a salt thereof that changes the binding property.
2 2 . 請求項 1記載のタンパク質または請求項 5記載の部分ペプチドまたはそ の塩および配列番号: 2 5で表されるアミノ酸配列と同一もしくは実質的に同一 のアミノ酸配列を含有するぺプチドまたはその塩を含有することを '特徴とする、 配列番号: 2 5で表されるアミノ酸配列と同一もしくは実質的に同一のアミノ酸 配列を含有するぺプチドと請求項 1記載のタンパク質またはその塩との結合性を 変化させる化合物またはその塩のスクリーニング用キット。  22. The protein according to claim 1 or the partial peptide according to claim 5, or a salt thereof, and a peptide containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 25, or a peptide thereof. 2. A bond between the protein according to claim 1 and a peptide having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 25, characterized by containing a salt. A screening kit for a compound or a salt thereof that changes the sex.
2 3 . 請求項 2 1記載のスクリ一ニング方法または請求項 2 2記載のスクリ一 ニング用キットを用いて得られうる、 配列番号: 2 5で表されるアミノ酸配列と 同一もしくは実質的に同一のアミノ酸配列を含有するペプチドまたはその塩と請 求項 1記載のタンパク質またはその塩との結合性を変化させる化合物またはその 塩。  23. The same or substantially the same as the amino acid sequence represented by SEQ ID NO: 25, which can be obtained by using the screening method according to claim 21 or the screening kit according to claim 22. A compound or a salt thereof, which alters the binding between a peptide or a salt thereof containing the amino acid sequence of claim 1 and the protein or a salt thereof according to claim 1.
2 4 . ァゴニストである請求項 2 3記載の化合物またはその塩。  24. The compound or a salt thereof according to claim 23, which is an agonist.
2 5 . アン夕ゴニストである請求項 2 3記載の化合物またはその塩。 25. The compound or a salt thereof according to claim 23, which is an angonist.
2 6 . 請求項 2 4記載の化合物またはその塩を含有してなる医薬。 26. A medicament comprising the compound according to claim 24 or a salt thereof.
2 7 . 請求項 2 5記載の化合物またはその塩を含有してなる医薬。 27. A medicament comprising the compound according to claim 25 or a salt thereof.
2 8 . 請求項 6記載のポリヌクレオチドと相補的な塩基配列またはその一部を 含有してなるアンチセンスポリヌクレオチド。 28. An antisense polynucleotide comprising a nucleotide sequence complementary to the polynucleotide according to claim 6, or a part thereof.
2 9 . 請求項 2 8記載のアンチセンスポリヌクレオチドを含有してなる医薬。 29. A medicament comprising the antisense polynucleotide according to claim 28.
3 0 . 請求項 6記載のポリヌクレオチドまたはその一部を用いることを特徴と する請求項 1記載の夕ンパク質の mR N Aの定量方法。 30. The method for quantifying mRNA of evening protein according to claim 1, wherein the polynucleotide according to claim 6 or a part thereof is used.
3 1 . 請求項 1 5記載の抗体を用いることを特徴とする請求項 1記載のタンパ ク質の定量方法。  31. The method for quantifying a protein according to claim 1, wherein the antibody according to claim 15 is used.
3 2 . 請求項 3 0または請求項 3 1記載の定量方法を用いることを特徴とする 請求項 1記載のタンパク質の機能が 連する疾患の診断方法。  32. A method for diagnosing a disease associated with the function of the protein according to claim 1, wherein the quantification method according to claim 30 or 31 is used.
3 3 . 請求項 3 0記載の定量方法を用いることを特徴とする請求項 1記載の夕 ンパク質の発現量を変化させる化合物またはその塩のスクリーニング方法。  33. The method for screening a compound or a salt thereof that changes the expression level of a protein according to claim 1, wherein the method for quantification according to claim 30 is used.
3 4. 請求項 3 0記載の定量方法を用いることを特徴とする細胞膜における請 求項 1記載の夕ンパク質量を変化させる化合物またはその塩のスクリーニング方 法。  3 4. The method for screening a compound or a salt thereof that changes the mass of protein according to claim 1, wherein the compound or the salt thereof is used in a cell membrane, characterized by using the quantification method according to claim 30.
3 5 . 請求項 3 3記載のスクリーニング方法を用いて得られうる請求項 1記載 のタンパク質の発現量を変化させる化合物またはその塩。  35. A compound or a salt thereof that changes the expression level of the protein according to claim 1, which can be obtained by using the screening method according to claim 33.
3 6 . 請求項 3 4記載のスクリーニング方法を用いて得られうる細胞膜におけ る請求項 1記載のタンパク質量を変化させる化合物またはその塩。  36. A compound or a salt thereof that alters the amount of the protein according to claim 1 in a cell membrane obtainable by using the screening method according to claim 34.
3 7 . 請求項 3 5または請求項 3 6記載の化合物またはその塩を含有してなる  37. It comprises the compound according to claim 35 or claim 36 or a salt thereof.
3 8 . 請求項 6記載のポリ クレオチドとハイ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。 38. A polynucleotide that hybridizes with the polynucleotide of claim 6 under high stringency conditions.
3 9 . 外来性の、 請求項 1記載のタンパク質をコードする D NAまたはその変 異 D N Aを有する非ヒトトランスジエニック動物。  39. A non-human transgenic animal having exogenous DNA encoding the protein of claim 1 or a mutant DNA thereof.
4 0 . 食欲不振、 食欲不振に伴う貧血ならびに低蛋白症、 微弱陣痛、 弛緩出血、 子宮復古不全または乳汁うっ滞の予防 ·治療剤である請求項 1 2、 請求項 1 3、 請求項 2 0または請求項 2 6記載の医薬。  40. Claim 12, Claim 13, Claim 20 which is a preventive / therapeutic agent for anorexia, anemia associated with anorexia, hypoproteinemia, faint labor, lax hemorrhage, uterine remodeling failure or milk stasis. Or the medicament according to claim 26.
4 1 . 肥満症、 摂食亢進症、 情動障害、 性機能障害、 過強陣痛、 強直性子宮収 縮、 早産、 Prader-Wi l l i症候群、 糖尿病およびその合併症、 高血圧、 高脂血症、 冠状動脈硬化症、 痛風、 呼吸器疾患、 脂肪肝、 不妊症または変形性骨関節症の予 防 ·治療剤である、 請求項 1 9、 請求項 2 7または請求項 2 9記載の医薬。 4 1. Obesity, hyperphagia, affective disorders, sexual dysfunction, overwork labor, ankylosing uterine atrophy, preterm birth, Prader-Willi syndrome, diabetes and its complications, hypertension, hyperlipidemia, coronary The medicament according to claim 19, claim 27, or claim 29, which is a prophylactic and / or therapeutic agent for arteriosclerosis, gout, respiratory disease, fatty liver, infertility or osteoarthritis.
4 2 . 哺乳動物に対して、 請求項 2 4記載の化合物またはその塩の有効量を投 与することを特徴とする、 食欲不振、 食欲不振に伴う貧血ならびに低蛋白症、 微 弱陣痛、 弛緩出血、 子宮復古不全または乳汁うっ滞の予防 ·治療方法。 42. Anorexia, anemia associated with anorexia, hypoproteinemia, weak labor, and relaxation characterized by administering an effective amount of the compound according to claim 24 or a salt thereof to a mammal. Prevention and treatment of bleeding, uterine reconstruction failure or milk stasis.
4 3 . 哺乳動物に対して、 請求項 2 5記載の化合物またはその塩の有効量を投 与することを特徴とする、 肥満症、 摂食亢進症、 情動障害、 性機能障害、 過強陣 痛、 強直性子宮収縮、 早産、 Prader-Wi l l i症候群、 糖尿病およびその合併症、 高 血圧、 高脂血症、 冠状動脈硬化症、 痛風、 呼吸器疾患、 脂肪肝、 不妊症または変 形性骨関節症の予防 ·治療方法。 43. An obesity, anorexia nervosa, affective disorder, sexual dysfunction, overwork, characterized by administering an effective amount of the compound according to claim 25 or a salt thereof to a mammal. Pain, ankylosing uterine contractions, preterm birth, Prader-Willi syndrome, diabetes and its complications, hypertension, hyperlipidemia, coronary atherosclerosis, gout, respiratory disease, fatty liver, infertility or deformed bone How to prevent and treat arthropathy.
4 4 . 食欲不振、 食欲不振に伴う貧血ならびに低蛋白症、 微弱陣痛、 弛緩出血、 子宮復古不全または乳汁うっ滞の予防 ·治療剤を製造するための、 請求項 2 4記 載の化合物またはその塩の使用。  44. The compound according to claim 24 or a compound thereof for producing a prophylactic or therapeutic agent for anorexia, anemia associated with anorexia, hypoproteinemia, weak labor, laxative bleeding, uterine remodeling failure or milk stasis. Use of salt.
4 5 . 肥満症、 摂食亢進症、 情動障害、 性機能障害、 過強陣痛、 強直性子宮収 縮、 早産、 Prader-Wi l l i症候群、 糖尿病およびその合併症、 高血圧、 高脂血症、 冠状動脈硬化症、 痛風、 呼吸器疾患、 脂肪肝、 不妊症または変形性骨関節症の予 防 ·治療剤を製造するための、 請求項 2 5記載の化合物またはその塩の使用。  4 5. Obesity, hyperphagia, affective disorders, sexual dysfunction, overwork labor, ankylosing uterine atrophy, preterm birth, Prader-Willi syndrome, diabetes and its complications, hypertension, hyperlipidemia, coronary Use of the compound according to claim 25 or a salt thereof for the manufacture of a prophylactic or therapeutic agent for arteriosclerosis, gout, respiratory disease, fatty liver, infertility or osteoarthritis.
PCT/JP2003/002133 2002-02-27 2003-02-26 Novel proteins, dnas thereof and use of the same WO2003072780A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2003211722A AU2003211722A1 (en) 2002-02-27 2003-02-26 Novel proteins, dnas thereof and use of the same

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
JP2002/51946 2002-02-27
JP2002051946 2002-02-27
JP2002178740 2002-06-19
JP2002/178740 2002-06-19

Publications (1)

Publication Number Publication Date
WO2003072780A1 true WO2003072780A1 (en) 2003-09-04

Family

ID=27767192

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2003/002133 WO2003072780A1 (en) 2002-02-27 2003-02-26 Novel proteins, dnas thereof and use of the same

Country Status (2)

Country Link
AU (1) AU2003211722A1 (en)
WO (1) WO2003072780A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004023870A1 (en) * 2002-09-10 2004-03-25 Takeda Pharmaceutical Company Limited Human slt transgenic animal

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001070975A1 (en) * 2000-03-24 2001-09-27 Yamanouchi Pharmaceutical Co., Ltd. Novel melanin concentrating hormone receptor

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001070975A1 (en) * 2000-03-24 2001-09-27 Yamanouchi Pharmaceutical Co., Ltd. Novel melanin concentrating hormone receptor

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
HILL J. ET AL.: "Molecular cloning and functional characterization of MCH2, a novel human MCH receptor", J. BIOL. CHEM., vol. 276, no. 23, June 2001 (2001-06-01), pages 20125 - 20129, XP002947038 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004023870A1 (en) * 2002-09-10 2004-03-25 Takeda Pharmaceutical Company Limited Human slt transgenic animal

Also Published As

Publication number Publication date
AU2003211722A1 (en) 2003-09-09

Similar Documents

Publication Publication Date Title
WO2002006483A1 (en) Novel physiologically active peptide and use thereof
US7671174B2 (en) Ligand to GPR8
JP2002112772A (en) New polypeptide and its dna
WO2001077325A1 (en) Novel g protein-coupled receptor protein and dna thereof
KR20010081109A (en) Novel g protein-coupled receptor protein, its dna and ligand thereof
JP5317318B2 (en) Novel polypeptides and uses thereof
US7250272B2 (en) G protein-coupled receptor protein and DNA thereof
US7268212B2 (en) Ligand of G protein-coupled receptor protein and DNA thereof
JP2002345468A (en) New insulin/igf/relaxin family polypeptide and its dna
US7235531B2 (en) Tachykinin-like polypeptides and use thereof
US7375074B2 (en) Body weight gain inhibitor
WO2003072780A1 (en) Novel proteins, dnas thereof and use of the same
JP4128030B2 (en) Novel ligand and its DNA
US20090227501A1 (en) Agents for preventing and/or treating upper digestive tract disorders
JP4959880B2 (en) Ligand for GPR8 and DNA thereof
JP4535670B2 (en) Novel polypeptides, their DNA and their uses
WO2004022086A1 (en) Adrenocortical hormone secretion controller
JP2004073179A (en) New protein, and dna and use of the same
JP4965776B2 (en) Novel bioactive peptides and uses thereof
JP2004051622A (en) New ligand and its dna
JP2003116582A (en) New physiologically active peptide and use thereof
JP2001149072A (en) New g protein conjugation type receptor protein, its dna and its ligand
WO2004084945A1 (en) Eating controllers
JP2001299362A (en) New polypeptide and dna encoding the same
JP2003174888A (en) New physiologically active peptide and use thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

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

Ref country code: JP