WO2004002515A1 - Diagnostics/preventives/remedies for respiratory diseases - Google Patents

Diagnostics/preventives/remedies for respiratory diseases Download PDF

Info

Publication number
WO2004002515A1
WO2004002515A1 PCT/JP2003/008167 JP0308167W WO2004002515A1 WO 2004002515 A1 WO2004002515 A1 WO 2004002515A1 JP 0308167 W JP0308167 W JP 0308167W WO 2004002515 A1 WO2004002515 A1 WO 2004002515A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
present
amino acid
dna
acid sequence
Prior art date
Application number
PCT/JP2003/008167
Other languages
French (fr)
Japanese (ja)
Inventor
Atsushi Nakanishi
Hiroki Iwashita
Shigeru Morita
Tatsumi Matsumoto
Masashi Yamasaki
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 AU2003246085A priority Critical patent/AU2003246085A1/en
Publication of WO2004002515A1 publication Critical patent/WO2004002515A1/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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • 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
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/34Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase
    • C12Q1/37Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase involving peptidase or proteinase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6884Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids from lung

Definitions

  • the present invention relates to an agent for preventing or treating respiratory diseases, a diagnostic agent, and the like.
  • Chronic obstructive pulmonary disease chronic bronchitis, emphysema, diffuse panbronchiolitis, endogenous asthma, etc. It is thought to be.
  • smoking can be a definite etiology of chronic obstructive pulmonary disease.
  • Smoking causes obstructive disorders and depends on the number of cigarettes. The younger the age at which smoking starts, the easier it is to progress.
  • a dose correlation between smoking and bronchial gland hyperplasia has been confirmed.
  • inhalation of tobacco can cause emphysema.
  • C0PD chronic obstructive pulmonary disease
  • Central airway involvement involves changes in secretory tissue morphology, such as goblet cell hyperplasia, hyperplasia of cells in the submucosal gland, and hypertrophy.
  • goblet cell hyperplasia For inflammatory cells, an increase in macrophage-activated T lymphocytes has been shown in the airway mucosa.
  • Lesions in the bronchiole region include mucus embolism in the airway lumen, goblet cell dysplasia in the airway epithelium, inflammatory cell infiltration in the airway wall, smooth muscle hypertrophy, and fibrosis.
  • PC0LCE2 has been cloned as one of the isoforms of PC0LCE1 (Genomics, 66, 264, 2000). Year) . It has been reported that PC0LCE1 has a high homology at the C-terminus to the N-terminus of tissue inhibitor of metaro protease (TIMP) (Protein Sci 8, 1636, In 1999, it has been reported that this C-terminal portion is actually cleaved after translation and acts as a meta-oral protease inhibitor (L Biol. Diem.) 275 , 1384, 2000). Disclosure of the invention
  • the present inventors have conducted intensive studies in order to solve the above-mentioned problems, and as a result, have found a gene whose expression is remarkably reduced in lung tissue having emphysema lesions, and based on this finding, conducted further studies based on this finding As a result, the present invention has been completed.
  • (1) contains a compound or a salt thereof which promotes the activity of a protein or a partial peptide thereof or a salt thereof containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1
  • the medicament according to the above (6) which is an agent for preventing or treating respiratory diseases.
  • a diagnostic agent for a respiratory disease comprising a polynucleotide encoding a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof,
  • a prophylactic / therapeutic agent for chronic obstructive pulmonary disease or bronchial asthma comprising a compound having a procollagen C-terminal truncation promoting activity or a salt thereof, '(12) SEQ ID NO: 1
  • a method for screening a remedy for the prevention and treatment of respiratory diseases which comprises using a protein having the same or substantially the same amino acid sequence as the amino acid sequence to be obtained or a partial peptide thereof or a salt thereof;
  • the pharmaceutical compound is a compound for preventing or treating respiratory disease, a compound used for preventing or treating respiratory disease and / or a compound having a preventive or treating effect for respiratory disease. (12a) The screening method according to the above,
  • the pharmaceutical compound is a compound for preventing or treating respiratory diseases, a compound used for preventing or treating respiratory diseases and / or a compound having a preventive or treating effect for respiratory diseases.
  • the compound or a salt thereof according to the above (15a) is a compound for preventing or treating respiratory diseases, a compound used for preventing or treating respiratory diseases and / or a compound having a preventive or treating effect for respiratory diseases.
  • a prophylactic / therapeutic agent for respiratory diseases comprising the compound or salt thereof according to (15b) above,
  • a respiratory tract characterized by using a polynucleotide encoding a protein or a partial peptide thereof containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1.
  • the pharmaceutical compound is a compound for preventing or treating respiratory disease, a compound used for preventing or treating respiratory disease and / or a compound having a preventive or treating effect for respiratory disease. (17a) the screening method described above,
  • a prophylactic or therapeutic agent for respiratory disease which can be obtained by using the screening method according to (17) or the screening kit according to (19).
  • the pharmaceutical compound is a compound for preventing or treating respiratory disease, a compound used for preventing or treating respiratory disease and / or a compound having a preventive or treating effect for respiratory disease. (20a) the compound or a salt thereof,
  • (22) a compound having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a compound that promotes the activity of a partial peptide or a salt thereof in mammals, or a compound thereof;
  • a method for preventing or treating respiratory diseases which comprises administering an effective amount of a salt, or a compound or a salt thereof that promotes the expression of a gene of the protein,
  • FIG. 1 is a diagram showing a pressure-volume curve of a mouse extirpated lung obtained in Example 1.
  • FIG. 1 is a diagram showing a pressure-volume curve of a mouse extirpated lung obtained in Example 1. In the figure,
  • FIG. 2 is a diagram showing the results of the expression level of PC0LCE2 gene obtained in Example 2.
  • ⁇ on the horizontal axis represents the lungs of a mouse exposed to cigarette smoke for 1 month
  • B represents the lungs of the control mouse
  • C represents the lungs of a mouse exposed to cigarette smoke for 3 months
  • D represents the lungs of the control mouse
  • E represents the lungs of 6 months.
  • F indicates the control mouse lung.
  • FIG. 3 is a diagram showing the results of mouse PCOLCE2 gene expression distribution obtained in Example 3.
  • FIG. 4 is a diagram showing a pressure-volume curve of a mouse extirpated lung obtained in Example 4.
  • one garden one was elastase administration.
  • FIG. 5 is a graph showing the time-dependent change in the compliance value of the mouse extirpated lung after administration of Erasinase obtained in Example 4.
  • the horizontal axis shows the number of days after elastase administration
  • the vertical axis shows the compliance value.
  • FIG. 6 is a graph showing the time course of the expression level of PC PLCE2 gene in mouse lung tissue obtained after administration of elastase obtained in Example 4.
  • the horizontal axis shows the number of days after Erasase administration
  • the vertical axis shows the amount of PCOLCE2 gene expression.
  • a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 used in the present invention (hereinafter referred to as the protein of the present invention or 2003/008167
  • the protein used in the present invention may be referred to as a human warm-blooded animal (eg, guinea pig, rat, mouse, chicken, egret, pig, sheep, hidge, horse, monkey, etc.) cells (eg, hepatocyte, spleen, etc.).
  • a human warm-blooded animal eg, guinea pig, rat, mouse, chicken, egret, pig, sheep, hidge, horse, monkey, etc.
  • cells eg, hepatocyte, spleen, etc.
  • Cells neurons, glial cells, splenic jS cells, bone marrow cells, mesangial cells, Langerhans 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 , Osteocytes, osteoblasts, osteoclasts, mammary cells, hepatocytes or stromal cells, or precursors, stem cells or cancer cells of these cells) or their cells Any tissue that does, such as the brain, parts of the brain (eg, olfactory bulb, amygdala, basal sphere, hippocampus, thalamus, hypothalamus, cerebral cortex, medulla, cerebell
  • amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 about 50% or more, preferably about 60% or more, more preferably about 50% or more of the amino acid sequence represented by SEQ ID NO: 1
  • Examples of the protein having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 include, for example, a protein having the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1.
  • a protein having substantially the same activity as the protein containing the amino acid sequence represented by SEQ ID NO: 1 is preferred.
  • SEQ ID NO: protein containing an amino acid sequence substantially identical to the amino acid sequence represented by 1 Examples of the quality include a protein containing the amino acid sequence represented by SEQ ID NO: 3.
  • Substantially equivalent activities include, for example, protease inhibitory activity, procollagen
  • the protease inhibitory activity, procollagen C-terminal cleavage promoting activity and the like are equivalent (eg, about 0.01 to 100 times, preferably about 0.1 to 10 times, more preferably 0.5 to 10 times).
  • the quantitative factors such as the degree of these activities and the molecular weight of the protein may be different.
  • proteases inhibitory activity and procollagen C-terminal cleavage promoting activity is performed by a method known per se, for example, J. Biol. Chem. 275 (2), 1384-90, 2000, Eur. J. Biochem. 186 Vol., U5-121, 1989, etc. or a method analogous thereto.
  • the protease inhibitory activity is measured by reacting the protein of the present invention, a labeled (eg, fluorescent label) peptide substrate and a protease, and measuring the mass of the peptide group cleaved by protease activity. This reaction is performed in an appropriate buffer.
  • the labeling agent is, for example, a fluorescent substance
  • the peptide group mass may be measured by measuring the fluorescence intensity. The measurement of the fluorescence intensity may be performed according to a known method using a fluorescence measurement device or the like.
  • procollagen C-terminal cleavage promoting activity is carried out by reacting the protein of the present invention, labeled (eg, radiolabeled) procollagen, and procollagen C-terminal region cleavage proteinase (eg, BMP-1). Then, the mass of the C-terminal protein of the open collagen cleaved by the protease activity is measured. This reaction is performed in an appropriate buffer.
  • labeling agent is, for example, a radioisotope
  • the amount of the C-terminal protein may be measured according to a known method using high performance liquid chromatography.
  • Examples of the protein used in the present invention include: (1) 1 or 2 or more (for example, about 1 to 100, preferably 1 to 100) in the amino acid sequence represented by SEQ ID NO: 1; An amino acid sequence in which about 30 amino acids have been deleted, preferably about 1 to 10 amino acids, and more preferably about 1 to 5 amino acids, and 2) the amino acid sequence represented by SEQ ID NO: 1.
  • One or two or more amino acids for example, about 1 to 100, preferably about 1 to 30, preferably about 1 to 10, and more preferably about 1 to 5)
  • the amino acid sequence represented by SEQ ID NO: 1 or 2 or more for example, about 1 to 100, preferably 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 inserted.
  • amino acid sequence represented by SEQ ID NO: 1 An amino acid sequence in which 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 an amino acid obtained by combining them.
  • So-called mutin such as a protein containing a sequence, represented by (2) SEQ ID NO: 3 1 or 2 or more in the amino acid sequence (eg, about 1 to 100, preferably about 1 to 30, preferably about 1 to 10, more preferably number (1 to 5))
  • amino acid sequence for example, about 1 to 100, preferably about 1 to 30, and preferably 1 to So-called muteins such as proteins containing an amino acid sequence in which about 10 amino acids are substituted, and more preferably a number (1 to 5) amino acids, or an amino acid sequence combining them are also included. included.
  • the position of the insertion, deletion or substitution is not particularly limited.
  • the left end is the N-terminus (amino terminus) and the right end is the C-terminus (capilloxy terminus) according to the convention of peptide labeling.
  • the proteins used in the present invention include carboxyl group (-C00H), carboxylate (-C00 "), amide (- 2 ) Or an ester (-C00R).
  • R in the ester e.g., methyl, Echiru, n- propyl, isopropyl
  • alkyl groups such as n- butyl, for example, C 3 _ 8 cycloalkyl group such as cyclohexyl cyclopentyl
  • sik port for example, phenyl, one naphth> C, such as les 6 - 12
  • Ariru group e.g., benzyl, C 7 one i 4 Ararukiru groups such as ⁇ - Nafuchiru C Bok 2 alkyl group such as phenylene Lou alkyl or a- naphthylmethyl such phenethyl, A bivaloyloxymethyl group or the like is used.
  • the protein used in the present invention has a lipoxyl group (or carboxylate) other than the C-terminus
  • a protein in which the carboxyl group is amidated or esterified is also included in the protein used in the present invention.
  • the ester J for example, the above-mentioned C-terminal ester or the like is used.
  • the amino group at the N-terminal amino acid residue (eg, methionine residue) is protected by a protecting group (eg, a CM acyl group such as an alkanol such as a formyl group or an acetyl group).
  • a protecting group eg, a CM acyl group such as an alkanol such as a formyl group or an acetyl group.
  • Substituent on the side chain of amino acid in the molecule for example, -0H, -SH, amino group , Imidazole, indole, guanidino, etc.
  • an appropriate protecting group for example, CHW alkyl such as CW alkanoyl such as formyl or acetyl
  • complex proteins such as so-called glycoproteins are also included.
  • protein used in the present invention include, for example, a protein containing the amino acid sequence represented by SEQ ID NO: 1, a protein containing the amino acid sequence represented by SEQ ID NO: 3, and the like.
  • the partial peptide of the protein used in the present invention is the partial peptide of the protein used in the present invention described above, and is preferably any peptide having the same properties as the protein used in the present invention described above. It may be.
  • the amino acid sequence at positions 380-390 in the amino acid sequence shown Examples include peptides having a sequence. For example, at least 20 or more, preferably 50 or more, more preferably 70 or more, more preferably 100 or more, most preferably 2 or more of the constituent amino acid sequences of the protein used in the present invention. Peptides having 100 or more amino acid sequences are used.
  • one or more (preferably about 1 to 10, more preferably a number (1 to 5)) amino acids in the amino acid sequence are deleted.
  • Or 1 or 2 or more (preferably, about 1 to 20; more preferably, about 1 to 10; more preferably, about 1 to 5) amino acids are added to the amino acid sequence;
  • Or 1 or 2 or more (preferably, about 1 to 20; more preferably, about 1 to 10; more preferably, about 1 to 5) amino acids are inserted into the amino acid sequence; or
  • one or more (preferably about 1 to 10, more preferably several, and still more preferably about 1 to 5) amino acids are substituted with another amino acid. Is also good.
  • the partial peptide used in the present invention may be either.
  • the partial peptides used in the present invention include those having a carbonyl group (or carboxylate) in addition to the C-terminus and the amino acid residues at the N-terminus, similarly to the proteins used in the present invention described above.
  • the amino group of a group eg, methionine residue
  • the glutamine residue generated by cleavage of the N-terminal side in vivo is pyroglutamine-oxidized
  • Also included are those in which the substituent is protected by an appropriate protecting group, and complex peptides such as so-called glycopeptides to which a sugar chain is bound.
  • the partial peptide used in the present invention can also be used as an antigen for producing an antibody.
  • a salt with a physiologically acceptable acid eg, an inorganic acid, an organic acid
  • a base eg, an alkali metal salt
  • Physiologically acceptable acid addition salts are preferred.
  • Such salts include: For example, salts with inorganic acids (eg, hydrochloric, phosphoric, hydrobromic, sulfuric) or organic acids (eg, acetic, formic, propionic, fumaric, maleic, succinic, tartaric, Salts with citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, and benzenesulfonic acid are used.
  • the protein or its partial peptide or a salt thereof used in the present invention can be produced from the above-mentioned human or warm-blooded animal cell or tissue by a known method for purifying a protein, or a DNA encoding the protein can be produced. It can also be produced by culturing the containing transformant. Also, it can be produced according to the peptide synthesis method described later.
  • the human or mammalian tissues or cells are homogenized, and then extracted with an acid or the like, and the extract is subjected to reverse phase chromatography, ion exchange chromatography, or the like. Purification and isolation can be achieved by combining chromatography.
  • a commercially available resin for protein synthesis can be usually used.
  • resins include chloromethyl resin, hydroxymethyl resin, benzylhydramine resin, aminomethyl resin, 4-benzyloxybenzyl alcohol resin, 4-methylbenzhydrylamine resin, PAM resin, and the like.
  • 4-Hydroxymethylmethylphenylacetamidomethyl resin, polyacrylamide resin, 4_ (2 ', 4, dimethoxyphenylhydroxymethyl) phenoxy resin, 4- (2,, 4, dimethoxyphenylaminoethyl) Phenoxy resins and the like can be mentioned.
  • an amino acid in which the amino group and the side chain functional group are appropriately protected is condensed on the resin according to the sequence of the target protein according to various known condensation methods.
  • protein or partial peptide is excised from the resin, and at the same time, various protecting groups are removed.
  • an intramolecular disulfide bond formation reaction is carried out in a highly diluted solution, and the target protein or partial peptide or their peptide Obtain the amide form.
  • Activating reagents can be used, and carbodiimides are particularly preferable.
  • carbopimides include DCC, N, ⁇ '-diisopropyl carbopimide, and ⁇ -ethyl-N '-(3-dimethylaminoprolyl) carbopimide.
  • Activation by these involves adding the protected amino acid directly to the resin along with a racemization inhibitor additive (eg, HOBt, HOOBt), or symmetrical acid anhydrides or HOBT esters or HOOBt.
  • the ester can be added to the resin after activation of the protected amino acid in advance.
  • the solvent used for activating the protected amino acid or condensing with the resin can be appropriately selected from solvents known to be usable for the protein condensation reaction.
  • acid amides such as N, N-dimethylformamide, N, N-dimethylacetamide, N-methylpyrrolidone, halogenated hydrocarbons such as methylene chloride, chloroform, and alcohols such as trifluoroethanol.
  • Sulfoxides such as dimethyl sulfoxide, ethers such as pyridine, dioxane, and tetrahydrofuran
  • nitriles such as aceto nitrile and propionitrile
  • esters such as methyl acetate and ethyl acetate; or an appropriate mixture thereof.
  • the reaction temperature is appropriately selected from the range known to be usable for the protein bond formation reaction, and is usually selected appropriately from the range of about 120 ° C. (: to 50 ° C ..
  • the activated amino acid derivative Is usually used in an excess of 1.5 to 4 times.As a result of a test using the ninhydrin reaction, if the condensation is insufficient, sufficient condensation can be carried out by repeating the condensation reaction without removing the protecting group. When sufficient condensation cannot be obtained even after repeating the reaction, acetylation of the unreacted amino acid with acetic anhydride or acetylimidazole is carried out so that the subsequent reaction is not affected. can do.
  • Examples of the protecting group for the amino group of the starting material include Z, Boc, t-pentyloxycarbonyl, isobornyloxycarbonyl, 4-methoxybenzyloxycarbonyl, C1-1Z, Br-Z, Damantyloxycarbonyl, trifluoroacetyl, phthaloyl, formyl, 2212 phenylsulfenyl, diphenylphosphinothioyl, Fmoc and the like are used.
  • Carbonyl groups are, for example, alkyl esterified (eg, methyl, ethyl, Linear, branched or cyclic alkylesters such as pill, butyl, t-butyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, 2-adamantyl, etc., aralkyl esterification (for example, benzyl ester) , 4-nitrobenzyl ester, 4-methoxybenzyl ester, 4-chlorobenzyl ester, benzylhydryl esterification), phenacyl esterification, benzyloxycarbonyl hydrazide, t-butoxycarbonyl hydrazide, trityl hydrazide Can be protected.
  • alkyl esterified eg, methyl, ethyl, Linear, branched or cyclic alkylesters such as pill, butyl, t
  • the hydroxyl group of serine can be protected, for example, by esterification or etherification.
  • groups appropriately used for the esterification include a lower (C! _ 6) Arukanoiru groups such Asechiru group, Aroiru group such Benzoiru group, benzyl O carboxymethyl Cal Poni group, and a group derived from carbonic acid such as ethoxycarbonyl group Is used.
  • groups of a group suitable for etherification include a benzyl group, a tetrahydropyranyl group, and a t-butyl group.
  • the protecting group of the phenolic hydroxyl group of tyrosine for example, B z 1, C 1 2 -B zl, 2- nitrobenzyl, B R_Z, such as t- butyl are used.
  • Examples of the protecting group for histidine imidazole include Tos, 4-methoxy-2,3,6-trimethylbenzenesulfonyl, DNP, benzyloxymethyl, Bum, Boc, Trt, and Fmoc. Can be
  • Examples of the activated carbonyl group of the raw material include, for example, corresponding acid anhydrides, azides, and active esters [alcohols (eg, phenol, 2,4,5-trichlorophenol, 2,4 —Dinitrophenol, cyanomethyl alcohol, paranitrophenol, HONB, N-hydroxysuccinimide, N-hydroxyfurimide, ester with HOB t)].
  • alcohols eg, phenol, 2,4,5-trichlorophenol, 2,4 —Dinitrophenol, cyanomethyl alcohol, paranitrophenol, HONB, N-hydroxysuccinimide, N-hydroxyfurimide, ester with HOB t
  • As the activated amino group of the raw material for example, a corresponding phosphoric amide is used.
  • Methods for removing (eliminating) protecting groups include, for example, catalytic reduction in the presence of a catalyst such as Pd-black or Pd-carbon in a stream of hydrogen, or hydrogen fluoride anhydride, methanesulfonic acid, trifluoromethane, or the like.
  • Acid treatment with sulfonic acid, trifluoroacetic acid or a mixture thereof, diisopropylethylamine, triethylamine, Base treatment with lysine, piperazine, etc., and reduction with sodium in liquid ammonia are also used.
  • the elimination reaction by the above-mentioned acid treatment is generally performed at a temperature of about 120 ° (: to 40 ° C).
  • the acid treatment for example, anisol, phenol, thioanisole, methacrylol, paracresol, It is effective to add a cation trapping agent such as dimethinoresulfide, 1,4-butanedithiol, 1,2-ethanedithiol, etc.
  • a cation trapping agent such as dimethinoresulfide, 1,4-butanedithiol, 1,2-ethanedithiol, etc.
  • 2,4-dinititol used as an imidazole protecting group for histidine
  • the mouth phenyl group is removed by thiophenol treatment, and the formyl group used as an indole protecting group for tributanone is deprotected by acid treatment in the presence of 1,2-ethanedithiol, 1,4-butanedithiol and the like.
  • it is also removed by alkali treatment with dilute sodium hydroxide solution, dilute ammonia and the like.
  • 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.
  • a peptide (protein) chain is desired on the amino group side.
  • a protein or partial peptide from which only the protecting group for the ⁇ -amino group at the ⁇ -terminal of the peptide chain has been removed, and a protein or partial peptide from which only the protecting group for the C-terminal lipoxyl group has been removed.
  • an ester of a protein or peptide for example, after condensing the ⁇ -hydroxyl group of the amino acid at the carboxy terminal with a desired alcohol to form an amino acid ester, And an ester form of the protein or peptide.
  • the partial peptide or a salt thereof used in the present invention can be produced according to a peptide synthesis method known per se, or by cleaving the protein used in the present invention with an appropriate peptidase. .
  • a method for synthesizing a peptide for example, any of a solid phase synthesis method and a liquid phase synthesis method may be used.
  • the objective peptide is produced by condensing a partial peptide or amino acid that can constitute the partial peptide used in the present invention with the remaining portion, and if the product has a protecting group, removing the protecting group to produce the desired peptide.
  • a protecting group removing the protecting group to produce the desired peptide.
  • Known condensation methods and elimination of protecting groups include, for example,
  • a general purification method such as solvent extraction, distillation, column chromatography, liquid chromatography, recrystallization, etc. may be combined to purify and isolate the partial peptide used in the present invention.
  • a general purification method such as solvent extraction, distillation, column chromatography, liquid chromatography, recrystallization, etc. may be combined to purify and isolate the partial peptide used in the present invention.
  • the partial peptide obtained by the above method is a free form, it can be converted into an appropriate salt by a known method or a method analogous thereto. It can be converted to a free form or other salts by a method analogous thereto.
  • the polynucleotide encoding the protein used in the present invention may be any polynucleotide containing the above-described nucleotide sequence encoding the protein used in the present invention.
  • it is DNA.
  • the DNA may be any of a genomic DNA, a genomic DNA library, the above-described cell-tissue-derived cDNA, the above-described cell-tissue-derived cDNA library, and a synthetic DNA.
  • the vectors used for the library include bacteriophages, plasmids, And phagemid. In addition, it can be directly amplified by reverse transcriptase polymerase chain reaction (hereinafter abbreviated as RT-PCR) using a total RNA or mRNA fraction prepared from the cells and tissues described above.
  • RT-PCR reverse transcriptase polymerase chain reaction
  • Examples of the DNA encoding the protein used in the present invention include a DNA containing the nucleotide sequence represented by SEQ ID NO: 2, or a DNA that is highly stringent with the nucleotide sequence represented by SEQ ID NO: 2. Any DNA that contains a base sequence that hybridizes under the conditions and encodes a protein having substantially the same properties as the protein containing the amino acid sequence represented by SEQ ID NO: 1 described above. May be. Examples of the DNA that can be hybridized with the nucleotide sequence represented by SEQ ID NO: 2 under eight stringent conditions include, for example, about 50% or more, preferably 50% or more, of the nucleotide sequence represented by SEQ ID NO: 2.
  • DNA or the like contains a nucleotide sequence having a homology of about 60% or more, more preferably about 70% or more, more preferably about 80% or more, particularly preferably about 90% or more, and most preferably about 95% or more.
  • DNA or the like is used.
  • a DNA containing the base sequence represented by SEQ ID NO: 4 and the like can be mentioned.
  • Hybridization is performed according to a method known per se or a method analogous thereto, for example, the method described in Molecular Cloning 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). Can be done. When a commercially available library is used, it can be performed according to the method described in the attached instruction manual. More preferably, the reaction can be performed under high stringent conditions.
  • Highly stringent 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 X :, preferably about 60 to 70 X.
  • the conditions at 65 ° C are shown. In particular, when the sodium concentration is about 19 mM and the temperature is about 65 ° C, Most preferred.
  • the DNA encoding the protein containing the amino acid sequence represented by SEQ ID NO: 1 includes a DNA containing the base sequence represented by SEQ ID NO: 2, and the like.
  • DNA containing the base sequence represented by SEQ ID NO: 4 or the like is used.
  • the polynucleotide (eg, DNA) encoding the partial peptide used in the present invention may be any polynucleotide containing the above-described nucleotide sequence encoding the partial peptide used in the present invention. Further, it 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.
  • DNA encoding the partial peptide used in the present invention for example, a DNA containing a part of the DNA containing the base sequence represented by SEQ ID NO: 2, or the base sequence represented by SEQ ID NO: 2
  • DNA containing a part of DNA encoding a protein having a nucleotide sequence that hybridizes under high stringent conditions and encoding a protein having substantially the same activity as the protein of the present invention may be used.
  • DNA that can hybridize with the nucleotide sequence represented by SEQ ID NO: 2 has the same meaning as described above.
  • the cloning of the DNA completely encoding DNA is amplified by the PCR method using a synthetic DNA primer having a part of the nucleotide sequence encoding the protein of the present invention, or the DNA incorporated into an appropriate vector is a part of the protein of the present invention.
  • selection can be performed by hybridization with a DNA fragment encoding the entire region or a DNA fragment labeled with a synthetic DNA.
  • the method of hybridization is, for example, molecular claw. This can be performed according to the method described in Molecular Cloning 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). 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 using PCR, a known kit, for example, Mutan TM -Super Express Km (Takara Shuzo Co., Ltd.), Mutan TM -K (Takara Shuzo Co., Ltd.), ODA-LAPCR method, Gapped
  • the method can be carried out according to a method known per se, such as the duplex method and 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 digesting with a restriction enzyme or adding a linker, if desired.
  • the DNA may have ATG as a translation initiation codon at its 5 'end and TAA, TGA or TAG as a translation termination codon at its 3' end. Codons and translation termination codons can also be added using a suitable synthetic DNA adapter.
  • the expression vector of the protein of the present invention includes, for example, (a) cutting out a target DNA fragment from DNA encoding the protein of the present invention, and (mouth) converting the DNA fragment into a promoter of an appropriate expression vector. It can be manufactured by connecting downstream.
  • the vector examples include a plasmid derived from E. coli (eg, PBR322, pBR325, pUC12, pUC13), a plasmid derived from Bacillus subtilis (eg, pUB110, TP5, pC194), a plasmid derived from yeast (eg, pSHl9) , PSHI 5), bacteriophage such as ⁇ phage, animal viruses such as retrovirus, vaccinia virus, baculovirus, etc., A1-11, pXT1, pRc / CMV, pRcZRSV, pcDNA I ZNeo or the like is used.
  • E. coli eg, PBR322, pBR325, pUC12, pUC13
  • Bacillus subtilis eg, pUB110, TP5, pC194
  • yeast eg, pSHl9
  • PSHI 5 bacteriophage
  • bacteriophage
  • the promoter used in the present invention may be any promoter as long as it is appropriate for the host used for gene expression.
  • SRa promoter when animal cells are used as a host, SRa promoter, SV40 promoter, LTR promoter, CMV promoter, HSV-TK promoter and the like can be mentioned.
  • CMV cytomegalovirus
  • the host is a bacterium, SP01 promoter, SP02 promoter, pen P promoter, etc., and when the host is yeast, PHO5 promoter, PGK promoter, GAP promoter, ADH promoter, etc. are preferable.
  • 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 SV40 ori), if desired.
  • an enhancer e.g., a splicing signal
  • a poly-A addition signal e.g., a poly-A addition signal
  • a selection marker e.g., SV40 replication origin
  • SV40 ori SV40 replication origin
  • the selection marker include a dihydrofolate reductase (hereinafter sometimes abbreviated as dh fr) gene (methotrexate (MTX) resistance) and an ampicillin resistance gene (hereinafter abbreviated as Amp) ), the neomycin resistance gene (hereinafter sometimes abbreviated as Ne o r, include G41 8-resistant) and the like.
  • dh fr dihydrofolate reductase
  • Amp ampicillin
  • the target gene when used as a selection marker using Chinese hamster cells deficient in the dh fr gene, the target gene can be selected using a thymidine-free medium. If necessary, a signal sequence suitable for the host is added to the N-terminal of the protein of the present invention.
  • a host is a bacterium belonging to the genus Escherichia, a PhoA signal sequence, an OmpA signal sequence, etc.
  • a bacterium belonging to the genus Bacillus a single amylase-signal sequence, subtilisin, a signal sequence, etc., and when the host is a yeast
  • MFa signal sequence, SUC2 signal sequence, etc. and when the host is an animal cell, insulin signal sequence, Hi-interferon signal sequence, antibody molecule, signal sequence, etc. can be used. .
  • a transformant can be produced using the thus-constructed vector containing DNA encoding the protein of the present invention.
  • Escherichia bacteria for example, Escherichia bacteria, Bacillus bacteria, yeast, insect cells, insects, animal cells, and the like are used.
  • Specific examples of the genus Escherichia include, for example, Escherichia coli Kl 2 -DH1 CProc. Natl. Acad. Sci. USA, 60, 160 (1968)], JM103 [Nucleic Acids Research, 9] 309 (1981)), JA 221 (Journal of Molecular Biology, 120, 517 (1978)), HB 101 [Journal of Molecular Biology, 41, 459 (1969)], C600 [Genetics, 39, 440 (1954)].
  • Bacillus bacterium for example, Bacillus subtilis MI114 [Gene, 24, 255 (1983)], 207-21 [Journal of Biochemistry, 95, 87 (1984)] and the like are used.
  • yeast examples include, for example, Saccharomyces cerevisiae AH22, AH22R—, NA87-11A, DKD-5D, 2OB-12, Schizosaccharomyces pombe NCYC 1913, NCYC 2036, Pichia pastoris Pichia pastoris) KM 71 and the like are used.
  • Insect cells include, for example, when the virus is Ac NPV, a cell line derived from a larva of night roth moth (Spodoptera frugiperda cell; S f cell), MG1 cell derived from the midgut of Trichoplusia ni, and egg derived from Tridioplusiani egg HigliFive TM cells, cells derived from Mamestra brassicae or cells derived from Estigmena acrea are used.
  • the virus is Bm NPV
  • a silkworm-derived cell line (Bombyx mori N cell; BmN cell) or the like is used.
  • Sf9 cells ATCCCRL1711
  • Sf21 cells Vaughn, J ⁇ et al., In Vivo, 13, 213-217, (1977)
  • 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 Chinese hamster cell CHO (hereinafter abbreviated as CHO (dh fr—) cell. ), Mouse L cells, mouse AtT-20, mouse myeloma cells, mouse ATDC5 cells, rat GH3, human FL cells, etc. are used.
  • Transformation of Bacillus spp. can be performed, for example, according to the method described in Molecular & General Genetics, vol. 168, 111 (1979).
  • Transformation of yeast can be performed, for example, according to the method described in Methods in Enzymology, Vol. 194, 182-187 (1991), Pro Natl. Acad. Sc. USA, Vol. 75, 1929 (1978). Can be.
  • Transformation of animal cells can be performed, for example, by the method described in Cell Engineering Separate Volume 8 New Cell Engineering Experimental Protocol. 263-267 (1995) (published by Shujunsha), Virology, 52, 456 (1973). So you can do it.
  • a liquid medium is suitable as a medium used for culturing, and a carbon source necessary for growth of the transformant may be used therein.
  • Nitrogen sources, inorganic substances, etc. are included.
  • Carbon sources include, for example, glucose, dextrin, soluble starch, sucrose, etc.
  • Nonrogen sources include, for example, ammonium salts, nitrates, copper steep liquor, peptone, casein, meat extract, soybean meal, potato inorganic or organic substances such as extract, etc.
  • the inorganic materials are calcium chloride, sodium dihydrogen phosphate, magnesium chloride and levator only be 5.
  • yeast extract, vitamins, growth promoting factors and the like may be added.
  • the pH of the medium is preferably about 5 to 8.
  • an M9 medium containing glucose and casamino acid As a medium for cultivating a bacterium belonging to the genus Escherichia, for example, an M9 medium containing glucose and casamino acid [Miller, Journal of Experiments in Molecular
  • an agent such as 3] 3-indolylacrylic acid can be added in order to make the promoter work efficiently.
  • the host is a bacterium belonging to the genus Escherichia
  • the cultivation is usually performed at about 15 to 43 ° C for about 3 to 24 hours, and if necessary, aeration and stirring can be applied.
  • the cultivation is usually performed at about 30 to 40 for about 6 to 24 hours, and if necessary, aeration and stirring can be applied.
  • the culture medium When culturing a transformant in which the host is yeast, the culture medium may be, for example, Burkholder's minimal medium (CBostian, KL et al., Proc. Natl. Acad. Sci. USA, 77, 4505 (1980)). And SD medium containing 0.5% casamino acid [Bitter, GA et al., Pro Natl. Acad. Sci. USA, 81, 5330 (1984)]. The pH of the medium is preferably adjusted to about 5-8. Culture is usually performed at about 20 ° C to 35 ⁇ for about 24 to 72 hours, and aeration and agitation are added as necessary.
  • examples of the medium include MEM medium containing about 5 to 20% fetal bovine serum [Science, 122, 501 (1952)], DMEM medium [Virology , 8, 396 (1959)], RPM I 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)] Are used.
  • the pH is about 6-8.
  • Culture is usually performed at about 30 to 401: about 15 to 60 hours, and aeration and stirring 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, and then subjected to ultrasound, A method of obtaining a crude protein extract by centrifugation or filtration after disrupting the cells or cells by zozyme and z or freeze-thawing or the like is appropriately used.
  • the buffer may contain a protein denaturant such as urea or guanidine hydrochloride, or a surfactant S such as Triton X-100 TM .
  • Purification of the protein contained in the culture supernatant or extract thus obtained can be carried out by appropriately combining known separation and purification methods.
  • Methods for separating and purifying these losses include methods using solubility such as salting out and solvent precipitation, dialysis, ultrafiltration, gel filtration, and SDS-polyacrylamide gel electrophoresis.
  • Methods that mainly use differences in molecular weight of DNA methods that use differences in charges such as ion exchange chromatography, methods that use specific affinity such as affinity chromatography, and reversed-phase high-performance liquid chromatography.
  • a method using a difference in hydrophobicity such as isoelectric point electrophoresis and a method using a difference in isoelectric point are used.
  • 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 enzymatic immunoassay western blotting using a specific antibody.
  • the antibody against the protein or partial peptide or a salt thereof used in the present invention may be any of a polyclonal antibody and a monoclonal antibody as long as it can recognize the protein or partial peptide or a salt thereof used in the present invention.
  • Antibodies against the protein or partial peptide used in the present invention or a salt thereof are prepared by using the protein of the present invention as an antigen, It can be produced according to a method for producing an antibody or antiserum known per se.
  • the protein of the present invention is administered to a warm-blooded animal itself or together with a carrier or a diluent at a site capable of producing an antibody upon administration.
  • Complete Freund's adjuvant or incomplete Freund's adjuvant may be administered in order to enhance the antibody-producing ability upon administration. Administration is usually performed once every 2 to 6 weeks, for a total of about 2 to 10 times.
  • warm-blooded animals to be used include monkeys, rabbits, rabbits, 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, a mouse with an antibody titer was selected from a mouse, and the spleen or lymph node was collected 2 to 5 days after the final immunization.
  • an antigen for example, a mouse with an antibody titer was selected from a mouse, and the spleen or lymph node was collected 2 to 5 days after the final immunization.
  • 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 a labeled protein described below with the antiserum, and then measuring the activity of a labeling agent bound to the antibody.
  • the fusion operation can be performed according to a known method, for example, the method of Köhler and Milstein [Nature, 256, 495 (1975)].
  • the fusion promoter include polyethylene glycol (PEG) and Sendai virus, but PEG is preferably used.
  • myeloma cells examples include warm-blooded animal myeloma cells 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 PEG 1000 to PEG 6000) is 10 to 8%.
  • Cell fusion can be carried out efficiently by adding at a concentration of about 0% 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 directly or adsorbed together with a carrier, and then Monoclonal antibody bound to solid phase using anti-immunoglobulin antibody labeled with a radioactive substance or enzyme (anti-mouse immunoglobulin antibody is used if the cells used for cell fusion are mice) or protein A
  • a radioactive substance or enzyme anti-mouse immunoglobulin antibody is used if the cells used for cell fusion are mice
  • Monoclonal antibody bound to the solid phase by adding the hybridoma culture supernatant to a solid phase to which anti-immunoglobulin antibody or protein A has been adsorbed, adding a protein labeled with a radioactive enzyme, etc. And the like.
  • the selection of the monoclonal antibody can be performed according to a method known per se 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 may be used as long as it can grow viable hybridomas.
  • RPMI 1640 medium containing 1 to 20%, preferably 10 to 20% fetal bovine serum, GIT medium containing 1 to 10% fetal bovine serum (Wako Pure Chemical Industries, Ltd.)
  • a serum-free culture 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 antibody titer of the supernatant of the hybridoma culture medium can be measured in the same manner as in the measurement of the antibody titer in the antiserum described above.
  • Monoclonal antibodies can be separated and purified by methods known per se, for example, immunoglobulin separation and purification methods (eg, salting out method, alcohol precipitation method, isoelectric point precipitation method, electrophoresis method, ion exchanger (eg, DEAE)) Absorption / desorption method, ultracentrifugation method, gel filtration method, specific purification method in which only the antibody is collected using an antigen-binding solid phase or an active adsorbent such as protein A or protein G and the bond is dissociated to obtain the antibody.) Can be performed according to (Preparation of polyclonal antibody)
  • immunoglobulin separation and purification methods eg, salting out method, alcohol precipitation method, isoelectric point precipitation method, electrophoresis method, ion exchanger (eg, DEAE)
  • Absorption / desorption method eg, ultracentrifugation method, gel filtration method, specific purification method in which only the antibody is collected using an antigen-binding solid phase
  • the polyclonal antibody of the present invention can be produced according to a method known per se or a method analogous thereto.
  • 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 monoglonal antibody.
  • the antibody can be produced by collecting an antibody-containing substance against the antibody and separating and purifying the antibody.
  • the type of carrier protein and the mixing ratio of the carrier and the hapten are determined by the antibody against the hapten immunized by cross-linking the carrier.
  • Any material may be cross-linked at any ratio as long as it can be efficiently used.
  • serum albumin, thyroglobulin, hemocyanin, etc. in a weight ratio of about 0.1 to 2 per hapten per hapten.
  • a method of coupling at a rate of 0, preferably about 1 to 5 is used.
  • Various condensing agents can be used for force coupling between the hapten and the carrier.
  • an active ester reagent containing a daltaraldehyde ⁇ ⁇ carpoimide, a maleimide active ester, a thiol group, or a dithioviridyl group is used.
  • the condensation product is administered to a warm-blooded animal itself or together with a carrier and a diluent at a site where antibody production is possible.
  • Complete Freund's adjuvant or incomplete Freund's adjuvant may be administered in order to enhance the antibody-producing ability upon administration.
  • the administration is usually performed once every about 2 to 6 weeks, for a total of about 3 to 10 times.
  • the polyclonal antibody can be collected from blood, ascites, etc., preferably from 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.
  • the polyclonal antibody can be separated and purified according to the same method for separating and purifying immunoglobulin as in the above-described method for separating and purifying a monoclonal antibody.
  • Polynucleotide encoding the protein or partial peptide used in the present invention eg, DNA (hereinafter, these DNAs may be abbreviated as the DNA of the present invention in the description of antisense polynucleotides)
  • Antisense polynucleotide having a nucleotide sequence complementary to or substantially complementary to a nucleotide sequence of As a peptide a nucleotide having a nucleotide sequence complementary to or substantially complementary to the nucleotide sequence of the DNA of the present invention or a part thereof and having an action capable of suppressing the expression of the DNA is provided. Any antisense polynucleotide may be used, but antisense DNA is preferred.
  • the nucleotide sequence substantially complementary to the DNA of the present invention refers to, for example, the entire nucleotide sequence or 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 nucleotide sequence of the portion encoding the N-terminal portion of the protein of the present invention for example, an antisense polynucleotide having a homology of about 70% or more, preferably about 80% or more, more preferably about 90% or more, and most preferably about 95% or more with the complementary strand of Mouth
  • an antisense polynucleotide which directs RNA degradation by RNaseH it is about 70% or more, preferably about 80% or more, complementary to the entire nucleotide sequence of the DNA of the present invention including introns. More preferably, antisense polynucleotides each having a homology of about 90% or more, most preferably about 95% or more are suitable.
  • an antisense polynucleotide containing a base sequence complementary to or substantially complementary to the base sequence of DNA containing the base sequence represented by SEQ ID NO: 2, or a part thereof An antisense polynucleotide containing a nucleotide, preferably, for example, a nucleotide sequence complementary to the nucleotide sequence of DNA containing the nucleotide sequence represented by SEQ ID NO: 2, or a part thereof (more preferably, a nucleotide sequence represented by SEQ ID NO: 2; A base sequence complementary to the base sequence of the DNA containing the base sequence represented, or an antisense polynucleotide containing a part thereof), (2) DNA containing the base sequence represented by SEQ ID NO: 4 An antisense polynucleotide containing a nucleotide sequence complementary to or substantially complementary to the nucleotide sequence of SEQ ID NO: 1, or a portion thereof, preferably, for example, represented by SEQ ID NO: 4 A base
  • An antisense polynucleotide is usually composed of about 10 to 40 bases, preferably about 15 to 30 bases.
  • the phosphate residue (phosphate) of each nucleotide constituting the antisense DNA may be, for example, a chemically modified phosphate residue such as phosphorothioate, methylphosphonate, or phosphorodithionate. It may be substituted by a group.
  • the sugar (doxylipose) of each nucleotide may be substituted with a chemically modified sugar structure such as 2, -O-methylation, and the base (pyrimidine, purine) may also be chemically modified. Any one may be used as long as it hybridizes to the DNA containing the base sequence represented by SEQ ID NO: 2.
  • These antisense polynucleotides can be produced using a known DNA synthesizer or the like.
  • an antisense polynucleotide capable of inhibiting the replication or expression of the protein gene of the present invention is designed based on the nucleotide sequence information of the cloned or determined DNA encoding the protein. And can be synthesized.
  • a nucleotide nucleic acid
  • Such a nucleotide can hybridize with the RNA of the protein gene of the present invention, inhibit the synthesis or function of the RNA, or inhibit the interaction with the protein-related RNA of the present invention.
  • the expression of the protein gene of the present invention can be regulated.
  • Polynucleotides that are complementary to the selected sequence of the protein-related RNA of the present invention, and that can specifically hybridize with the protein-related RNA of the present invention, can be used 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 “responds” means having homology or being complementary to a nucleotide, base sequence or a specific sequence of a nucleic acid including a gene.
  • “Corresponding” between a nucleotide, nucleotide sequence or nucleic acid and a peptide (protein) usually refers to the amino acid of the peptide (protein) specified by the sequence derived from the nucleotide (nucleic acid) sequence or its complement.
  • the relationship between the target nucleic acid and a polynucleotide complementary to at least a part of the target region can be said to be "antisense” if the relationship between the target nucleic acid and the polynucleotide that can hybridize with the target is.
  • Antisense polynucleotides are polynucleotides that contain 2-deoxy D-reports, polynucleotides that contain D-reports, and other types of polynucleotides that are N-glycosides of purine or pyrimidine bases.
  • nucleotide backbone eg, commercially available protein nucleic acids and synthetic sequence-specific nucleic acid polymers
  • other polymers containing special bonds provided that the polymer is not contained in DNA or RNA.
  • Pairing of bases (contains nucleotides having a configuration that allows base attachment) as described in (1). They can be double-stranded DNA, single-stranded DNA, double-stranded RNA, single-stranded RNA, or even a DNA: RNA hybrid, and can be unmodified polynucleotides (or unmodified polynucleotides).
  • Oligonucleotides and those with known modifications, e.g., those with labels, capped, methylated, and one or more natural nucleotides that are known in the art.
  • Substituted with an intramolecular nucleotide for example, having an uncharged bond (eg, methylphosphonate, phosphotriester, phosphoramidate, olebamate, etc.), charged bond or sulfur-containing Those having a bond (for example, phosphorothioate, phosphorodithioate, etc.), for example, protein (nuclease, nuclease inhibitor, toxin) Antibodies, signal peptides, poly-L-lysine, etc.) or sugars (for example, monosaccharides), etc., which have side-chain groups, or those with interactive compounds (for example, acridine, psoralen, etc.), (Eg, metals, radioactive metals, boron, oxidizable metal
  • nucleoside may include not only those containing purine and pyrimidine bases but also those having other modified heterocyclic bases.
  • Such decorations 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 halogens, aliphatic groups, etc., or ethers, amines, etc. It may have been converted to a functional group.
  • the antisense polynucleotide of the present invention is RNA, DNA, or a modified nucleic acid (RNA, DNA).
  • modified nucleic acid include, but are not limited to, a sulfur derivative of a nucleic acid, a thiophosphoate derivative, and a polynucleoside amide, which is resistant to degradation of an oligonucleoside amide.
  • the antisense nucleic acid of the present invention can be preferably designed according to the following policy. That is, to make the antisense nucleic acid more stable in the cell, to make the antisense nucleic acid more cell-permeable, to have a greater affinity for the target sense strand, and if toxic Minimize the toxicity of antisense nucleic acids.
  • the antisense nucleic acids of the present invention may contain altered or modified sugars, bases, or bonds, and may be provided in special forms such as ribosomes or microspheres, applied by gene therapy, or added. Can be given in a prescribed form.
  • additional forms include polythiones 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. (Eg, phospholipids, cholesterol, etc.).
  • Preferred lipids for addition include cholesterol and its derivatives (eg, cholesteryl chromate formate, Etc.).
  • Such a substance can be attached to the 3 'end or 5' end of a nucleic acid, and can be attached via a base, sugar, or intramolecular nucleoside bond.
  • Other groups include capping groups specifically located at the 3 'or 5' end of nucleic acids that prevent degradation by nucleases such as exonuclease and RNase. . Examples of such capping groups include, but are not limited to, hydroxyl-protecting groups known in the art, such as glycols such as polyethylene glycol and tetraethylene dalicol.
  • the inhibitory activity of the antisense nucleic acid 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.
  • the nucleic acid can be applied to cells by various methods known per se.
  • the protein or partial peptide of the present invention or a salt thereof (hereinafter, sometimes abbreviated as the protein of the present invention), a polynucleotide encoding the protein or partial peptide of the present invention (eg, DNA (hereinafter, referred to as the present invention) May be abbreviated as DNA))), 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 abbreviated as the antisense polynucleotide of the present invention).
  • the expression of the protein of the present invention is reduced in the lung having emphysema lesions, it can be used as a disease marker. That is, it is useful as a marker for early diagnosis in the lung, judgment of the severity of symptoms, and prediction of disease progression. Therefore, a drug containing the antisense polynucleotide of the gene encoding the protein of the present invention, a compound that inhibits the activity of the protein of the present invention or a salt thereof, or an antibody against the protein of the present invention is, for example, a respiratory tract.
  • rhinitis eg, allergic rhinitis
  • Hay fever acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry rhinitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc.
  • immune diseases eg, myasthenia gravis, glomeruli
  • Nephritis multiple sclerosis, siedalen syndrome, insulin resistant diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopy Dermatitis, leukocyte abnormalities, spleen insufficiency or immunodeficiency associated with thymic abnormal
  • a preventive agent can be used as a preventive agent.
  • it is a prophylactic / therapeutic agent for a respiratory disease or the like, more preferably a prophylactic / therapeutic agent for a chronic obstructive pulmonary disease or the like, and further preferably a prophylactic / therapeutic agent for a pulmonary emphysema or the like.
  • the compound or its salt that regulates (preferably enhances) the activity of the protein of the present invention may be used, for example, for respiratory diseases [eg, chronic obstructive Lung disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.), rhinitis (eg, allergic rhinitis, pollinosis, acute rhinitis, Chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry rhinosinusitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc., immune diseases (eg, myasthenia gravis, glomerulonephritis, multiple sclerosis) , Siedaren syndrome, insulin resistance diabetes, r
  • it is a prophylactic / therapeutic agent for a respiratory disease or the like, more preferably a prophylactic / therapeutic agent for a chronic obstructive pulmonary disease or the like, and further preferably a prophylactic / therapeutic agent for an emphysema or the like.
  • the protein of the present invention is useful as a reagent for screening a compound or a salt thereof that regulates (preferably promotes) the activity of the protein of the present invention.
  • the present invention regulates (promotes or inhibits) the activity of the protein of the present invention (eg, a protease inhibitory activity, a procollagen C-terminal cleavage promoting activity, etc.) characterized by using the protein of the present invention. And a method for screening a compound or a salt thereof.
  • the protein of the present invention eg, a protease inhibitory activity, a procollagen C-terminal cleavage promoting activity, etc.
  • protease inhibitory activity of the protein of the present invention or the C-terminal cleavage promoting activity of procollagen and (ii) the protease inhibitory activity or procollagen c-terminal of the mixture of the protein of the present invention and a test compound.
  • Compounds that regulate (promote or inhibit) the activity of the protein of the present invention which are characterized by comparison with the cleavage promoting activity.
  • a screening method for a salt thereof is used.
  • the protease inhibitory activity or procollagen C-terminal cleavage promoting activity can be determined by a method known per se, for example, J. Biol. Chera. 275 (2 ), Pp. 1384-90, 2000, Eur. J. Biochem. 186, pp. 115-121, 1989, or the like, or a method analogous thereto, and compared.
  • the labeled peptide substrate and protease may be reacted with the protein of the present invention after being mixed with the test compound, or after contacting the peptide substrate and the protease with the protein of the present invention, Test compounds may be added. Specifically, (i) a reaction of the protein of the present invention with a labeled (eg, radiolabeled) procollagen and a procollagen C-terminal region cleaving protease (eg, BMP-1 etc.) ii) Reaction of the protein of the present invention, labeled (eg, radiolabeled) procollagen, and procollagen C-terminal region cleavage proteinase (eg, BMP-1) in the presence of a test compound Then, the mass of the C-terminal protein of procollagen cleaved by the protease activity is measured, and a compound or a salt thereof that regulates (promotes or inhibits) the activity of the protein of the present invention is screened.
  • This reaction is performed in an appropriate buffer.
  • the amount of the c-terminal protein of the cleaved procollagen is measured by a known method.
  • the labeling agent is, for example, a radioisotope
  • the amount of the C-terminal protein can be measured by a known method using high performance liquid chromatography or the like. It may be performed according to.
  • the labeled procollagen and procollagen C-terminal region cleaving protease may be mixed with a test compound and then reacted with the protein of the present invention. After contacting the domain-cleaving protease, the test compound may be added.
  • the above-mentioned protein of the present invention those produced by culturing cells having the ability to produce the protein of the present invention and the like are used. Furthermore, a culture solution of the cells, a supernatant thereof, a cell crushed product, or the like may be used.
  • a host transformed with a vector containing the above-described DNA encoding the protein of the present invention
  • a host for example, animal cells such as COS 7 cells, CHO cells, and HEK293 cells are preferably used.
  • a transformant which secretes the protein of the present invention extracellularly or expresses it intracellularly by culturing by the above-mentioned method is preferably used.
  • the method for culturing cells capable of expressing the protein of the present invention is the same as the above-described method for culturing the transformant of the present invention.
  • Test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and the like.
  • the protease inhibitory activity or procollagen C-terminal cleavage promoting activity in the case of the above (ii) is about 20% or more, preferably 30% or more, more preferably about 50% or more, as compared with the case of the above (i).
  • test compound which increases the activity of the protein of the present invention by at least 10% is considered to be a compound that promotes the activity of the protein of the present invention, in which the protease inhibitory activity or procollagen C-terminal cleavage promoting activity in the above (ii) is about
  • a test compound that reduces by 20% or more, preferably 30% or more, more preferably about 50% or more can be selected as a compound that inhibits the activity of the protein of the present invention.
  • the compound having the activity of promoting the activity of the protein of the present invention may be a safe and low-toxic drug for enhancing the action of the protein of the present invention, for example, respiratory diseases [eg, chronic obstructive ⁇ J disease ( 1 man bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cyst Dysfibrosis, irritable pneumonia, pulmonary fibrosis, etc.), rhinitis (eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrene Rhinitis, sinusitis, etc., immune diseases (eg, myasthenia gravis, glomerulonephritis, multiple sclerosis, Scheigren's syndrome, insulin resistance diabetes, rheumatoid arthritis, systemic lupus
  • the compound having the activity of inhibiting the activity of the protein of the present invention is a safe and low-toxic drug for suppressing the physiological activity of the protein of the present invention, for example, respiratory diseases (eg, bronchial asthma, cystic Fibrosis, pulmonary fibrosis, idiopathic interstitial pneumonia, etc.), liver disease (eg, chronic active hepatitis, cirrhosis, hepatic fibrosis, etc.), arteriosclerosis, renal sclerosis, myelofibrosis, diabetic vascular disorder, etc. It is useful as a preventive and therapeutic agent.
  • respiratory diseases eg, bronchial asthma, cystic Fibrosis, pulmonary fibrosis, idiopathic interstitial pneumonia, etc.
  • liver disease eg, chronic active hepatitis, cirrhosis, hepatic fibrosis, etc.
  • arteriosclerosis eg, chronic active hepatitis, cirrhosis, hepatic
  • Compounds or salts thereof obtained using the screening method or the screening kit of the present invention include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts It is a compound selected from liquids, plasma, etc.
  • the salt of the compound those similar to the aforementioned salts of the peptide of the present invention are used.
  • the expression of the gene encoding the protein of the present invention also decreases in lung tissue having emphysema lesions, a compound or a salt thereof that regulates (preferably promotes) the expression of the gene encoding the protein of the present invention may be used.
  • respiratory diseases eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.
  • Rhinitis eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrenous rhinitis, sinusitis, etc.
  • immunological diseases example) , Myasthenia gravis, glomerulonephritis, multiple sclerosis, Siedalen syndrome, insulin resistance diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, white Spheres abnormalities, such as spleen dysfunction or thymus abnormally
  • a prophylactic or therapeutic agent for respiratory diseases etc.
  • it is a prophylactic / therapeutic agent for chronic obstructive pulmonary disease and the like, and further preferably, a prophylactic / therapeutic agent for emphysema and the like.
  • the polynucleotide (eg, DNA) of the present invention is useful as a reagent for screening a compound or a salt thereof that regulates the expression of a gene encoding the protein of the present invention.
  • the screening methods include (iii) culturing cells having the ability to produce the protein of the present invention, and (iv) cells having the ability to produce the protein used in the present invention in the presence of the test compound.
  • a screening method characterized by performing a comparison with the case of culturing is used.
  • the expression level of the gene (specifically, the amount of the protein of the present invention or the amount of mRNA encoding the protein) in the cases (ii) and (iv) is measured and compared.
  • test compound and cells having the ability to produce the protein of the present invention include those similar to those described above.
  • the amount of the protein is measured by a known method, for example, using an antibody that recognizes the protein of the present invention, and analyzing the protein present in a cell extract or the like according to a method such as Western analysis, ELISA, or a method analogous thereto. Can be measured.
  • the amount of mRNA can be measured by a known method, for example, Northern hybridization using a nucleic acid containing SEQ ID NO: 2 or a part thereof as a probe, or a nucleic acid containing SEQ ID NO: 2 or a part thereof as a primer Northern hybridization using a nucleic acid containing SEQ ID NO: 4 or a part thereof, or PCR using a nucleic acid containing SEQ ID NO: 4 or a part thereof as a primer or a method analogous thereto can be used. it can.
  • a test compound that inhibits about 20% or more, preferably 30% or more, more preferably about 50% or more of the protein of the present invention Compounds that suppress the expression of encoded genes Can be selected as an object.
  • the screening kit of the present invention contains the protein or partial peptide used in the present invention or a salt thereof, or a cell capable of producing the protein or partial peptide used in the present invention.
  • the compound or a salt thereof obtained by using the screening method or the screening kit of the present invention may be a test compound described above, for example, a peptide, a protein, a non-peptidic compound, a synthetic compound, a fermentation product, a cell extract, or a plant extract.
  • salt of the compound those similar to the aforementioned salts of the protein of the present invention are used.
  • a compound or a salt thereof that regulates (preferably promotes) the activity of the protein of the present invention and a compound or a salt thereof that regulates (preferably promotes) the expression of a gene encoding the protein of the present invention are, for example, respiratory diseases, respectively.
  • rhinitis eg, allergic rhinitis, Hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry rhinitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc.
  • immune diseases eg, myasthenia gravis, glomerulonephritis
  • Siedalen syndrome insulin-resistant diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, spleen dysfunction Or the like thymic abnormally accompanying immunodeficiency
  • inflammatory diseases eg, myasthenia gravis, glomerulonephritis
  • it is a prophylactic / therapeutic agent for a respiratory disease or the like, more preferably a prophylactic / therapeutic agent for a chronic obstructive pulmonary disease or the like, and further preferably a prophylactic / therapeutic agent for a pulmonary emphysema or the like.
  • compositions for oral administration include solid or liquid dosage forms such as tablets (including sugar-coated tablets and film-coated tablets), pills, granules, powders, capsules (soft capsules and the like). ), Syrups, emulsions, suspensions and the like.
  • Such a composition is produced by a method known per se and contains a carrier, diluent or excipient usually used in the field of pharmaceuticals.
  • compositions for parenteral administration include, for example, injections, suppositories, injections, intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections, intravenous injections, intraarticular injections Includes dosage forms such as injections.
  • Such injections are prepared according to a method known per se, 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 liquid for injection for example, physiological saline, isotonic solution containing glucose and other auxiliary agents and the like are used, and suitable dissolution and trapping !!, for example, alcohol (eg, ethanol), polyalcohol (Eg, propylene glycol, polyethylene glycol), nonionic surfactants (eg, polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated catalyst)), etc. Good.
  • alcohol eg, ethanol
  • polyalcohol Eg, propylene glycol, polyethylene glycol
  • nonionic surfactants eg, polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated catalyst)
  • oily liquid for example, sesame oil, soybean oil, and the like are used, and benzyl benzoate, benzyl alcohol, and the like may be used in combination as a solubilizing agent.
  • the prepared injection is usually filled in an appropriate ampoule.
  • the oral or parenteral pharmaceutical composition of the above 3 is conveniently prepared in a dosage unit form so as to be compatible with the dosage of the active ingredient.
  • dosage unit dosage forms include tablets, pills, capsules, injections (ampoules), suppositories, etc., and usually 5 to 500 mg, especially 5 for injections. 110 O mg, and other dosage forms preferably contain 10-25 O mg of the compound.
  • Each of the above-mentioned compositions may contain another active ingredient as long as the composition does not cause an undesirable interaction with the compound.
  • the preparations obtained in this way are safe and of low toxicity, for example, It may be orally or parenterally administered to blood animals (eg, mice, rats, egrets, higgins, bush, pests, pests, birds, cats, dogs, monkeys, chimpanzees, etc.). it can.
  • blood animals eg, mice, rats, egrets, higgins, bush, pests, pests, birds, cats, dogs, monkeys, chimpanzees, etc.
  • the dose of the compound or a salt thereof varies depending on its action, target disease, subject to be administered, administration route, and the like.
  • a compound or a salt thereof that regulates the activity of the protein of the present invention for the purpose of treating emphysema When the compound is orally administered, generally, in an adult (with a body weight of 60 kg), the compound or a salt thereof is used in an amount of about 0.1 to 100 mg, preferably about 1.0 to 5 Omg, more preferably about 1 to 10 mg per day. Administer 0-20 mg.
  • the single dose of the compound or a salt thereof varies depending on the administration subject, the target disease, and the like.
  • a compound that regulates the activity of the protein of the present invention for the purpose of treating emphysema when administered to an adult (with a body weight of 60 kg) usually in the form of an injection, the compound or its salt is administered in an amount of about 0.01 to 30 mg, preferably about 0.1 to 2 Omg per day. It is convenient to administer preferably about 0.1 to 10 mg by intravenous injection. In the case of other animals, the dose can be administered in terms of weight per 60 kg.
  • an antibody against the protein of the present invention (hereinafter sometimes abbreviated as the antibody of the present invention) can specifically recognize the protein of the present invention. It can be used for quantification by San Deutsch immunoassay.
  • the monoclonal antibody of the present invention In addition to quantification of the protein of the present invention using a monoclonal antibody against the protein of the present invention (hereinafter sometimes referred to as the monoclonal antibody of the present invention), detection by tissue staining or the like can also be performed.
  • the antibody molecule itself may be used, or 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. Any measurement method may be used as long as the amount of the body is detected by chemical or physical means and calculated from a standard curve prepared using a standard solution containing a known amount of antigen. .
  • nephelometry, a competition method, an immunometric method and a sandwich method are preferably used, but it is particularly preferable to use a sandwich method described later in terms of sensitivity and specificity.
  • a labeling agent used in a measurement method using a labeling substance for example, a radioisotope, an enzyme, a fluorescent substance, a luminescent substance and the like are used.
  • radioisotopes for example,
  • the above-mentioned enzyme those which are stable and have high specific activity are preferable, and for example, ⁇ -galactosidase, ⁇ -dalcosidase, alkaline phosphatase, peroxidase, malate dehydrogenase and the like are used.
  • the fluorescent substance for example, fluorescamine, fluorescein isothiocyanate or the like is used.
  • the luminescent substance for example, luminol, luminol derivative, reluciferin, lucigenin and the like are used.
  • a biotin-avidin system can be used for binding the antibody or antigen to the labeling agent.
  • 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.
  • the carrier include insoluble polysaccharides such as agarose, dextran, and cellulose; synthetic resins such as polystyrene, polyacrylamide, and silicon; and glass.
  • the 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 liquid 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.
  • the antibody used for the solid phase antibody or the labeling antibody is not necessarily 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 competitive method, an immunometric method, or a nephrometry.
  • a competitive method the antigen in the test solution and the labeled antigen are allowed to react competitively with the antibody, and then the unreacted labeled antigen (F) is separated from the labeled antigen (B) bound to the antibody. Then (BZF separation), the labeling amount of either B or F is measured, and the amount of antigen in the test solution is quantified.
  • a soluble antibody is used as the antibody
  • BZF separation is performed using polyethylene glycol
  • a liquid phase method using a second antibody to the antibody a solid phase antibody is used as the first antibody
  • a solid-phase method using a soluble first antibody and a solid-phased 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 the 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, and then the immobilized antigen is added to bind the unreacted labeled antibody to the solid phase, and then the solid phase and the liquid phase are separated.
  • measure the amount of label in either phase is quantified.
  • the amount of insoluble sediment resulting from the 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 can be obtained, a laser-Nef orifice utilizing laser scattering is preferably used.
  • the protein measurement 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 and written documents.
  • the protein of the present invention can be quantified with high sensitivity by using the antibody of the present invention.
  • respiratory diseases eg, chronic obstructive Pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.
  • rhinitis eg, allergy Rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc.
  • immune diseases eg, myasthenia gravis, Glomerulonephritis, multiple sclerosis, siedalen syndrome, insulin resistance diabetes, rheumatoid arthritis, systemic lupus erythemat
  • 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.
  • detecting the protein of the present invention in each fraction during purification, and analyzing 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, pigs, pigs, dogs, cats, dogs, monkeys, Abnormalities (gene abnormalities) in the DNA or mRNA encoding the protein of the present invention or a partial peptide thereof in a chimpanzee or the like can be detected. Or, it is useful as a diagnostic agent for genes such as decreased expression, increased DNA or mRNA, or overexpression.
  • warm-blooded animals e.g., rats, mice, guinea pigs, egrets, birds, higgies, pigs, pigs, dogs, cats, dogs, monkeys
  • Abnormalities (gene abnormalities) in the DNA or mRNA encoding the protein of the present invention or a partial peptide thereof in a chimpanzee or the like can be detected.
  • it is useful
  • the above-described genetic diagnosis using the DNA of the present invention can be performed, for example, by the well-known Northern high predication or 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)).
  • respiratory diseases eg, chronic obstructive pulmonary disease ('
  • rhinitis eg, allergic rhinitis, pollinosis, acute rhinitis, chronic
  • Rhinitis eg, allergic rhinitis, pollinosis, acute rhinitis, chronic
  • Rhinitis eg, allergic rhinitis, pollinosis, acute rhinitis, chronic
  • Rhinitis eg, allergic rhinitis, pollinosis, acute rhinitis, chronic
  • Rhinitis eg, hypertrophic rhinitis, atrophic rhinitis, before drying Rhinitis, vasomotor rhinitis, gangrenous rhinitis, sinusitis,
  • 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 has the protein of the present invention or the present invention in vivo.
  • DNA function eg, protease inhibitory activity, procollagen C-terminal truncation promoting activity, etc.
  • DNA function eg, protease inhibitory activity, procollagen C-terminal truncation promoting activity, etc.
  • Prophylactic and therapeutic agents for liver disease eg, chronic active hepatitis, cirrhosis, hepatic fibrosis, etc.
  • arteriosclerosis eg, chronic active hepatitis, cirrhosis, hepatic fibrosis, etc.
  • arteriosclerosis eg, chronic active hepatitis, cirrhosis, hepatic fibrosis, etc.
  • arteriosclerosis eg, chronic active hepatit
  • the antisense polynucleotide When used as the above-mentioned prophylactic / therapeutic agent, it can be formulated and administered according to a method known per se.
  • a human or mammal can be treated in a conventional manner. It can be given orally or parenterally to animals (eg, rats, puppies, sheep, sheep, bush, 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. Alternatively, they can be aerosolized and administered topically into the trachea as an inhalant.
  • the antisense polynucleotide is formulated alone or together with a carrier such as liposome (injection), and is intravenously, subcutaneously, It may be administered to the respiratory tract or lung lesions.
  • a carrier such as liposome (injection)
  • Dosage depends on target disease, subject, route of administration
  • the antisense polynucleotide of the present invention is administered for the treatment of emphysema, generally, in an adult (body weight 60 kg), the antisense polynucleotide is administered daily. About 0.1 to 10 O mg is administered.
  • the antisense polynucleotide can also be used as a diagnostic oligonucleotide probe for examining the presence of DNA of the present invention in tissues or cells and the state of its expression.
  • RNA containing a part of the RNA encoding the protein of the present invention a lipozyme containing a part of the RNA encoding the protein of the present invention, etc.
  • Gene expression, and the function of the protein used in the present invention or the DNA used in the present invention in vivo can be suppressed.
  • respiratory diseases eg, bronchial asthma, cysts
  • Cystic fibrosis pulmonary fibrosis, idiopathic interstitial pneumonia
  • liver disease eg, chronic active hepatitis, cirrhosis, hepatic fibrosis, etc.
  • arteriosclerosis renal sclerosis, myelofibrosis, diabetic It can be used as a prophylactic and therapeutic agent for vascular disorders.
  • 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 linking a known lipozyme to a part of the RNA encoding the protein of the present invention.
  • a part of the RNA encoding the protein of the present invention includes a portion (RNA fragment) adjacent to the cleavage site on the RNA of the present invention which can be cleaved by a known lipozyme.
  • RNA or lipozyme When the above-described 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 an antisense polynucleotide. (5) a drug containing the antibody of the present invention
  • the antibody of the present invention may be used, for example, for respiratory diseases [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis Etc.), rhinitis (eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic nose Inflammation, atrophic rhinitis, dry rhinitis, vasomotor rhinitis, gangrenous rhinitis, sinusitis, etc., immune disorders (eg, myasthenia gravis, glomerulonephritis, multiple sclerosis, Scheddaren syndrome, Insulin-resistant diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, immunodeficiency associated with
  • Or respiratory disease eg, bronchial asthma, cystic fibrosis, pulmonary fibrosis, idiopathic interstitial pneumonia, etc.
  • liver disease eg, chronic active hepatitis, cirrhosis, liver fibrosis, etc.
  • arteriosclerosis It can also be used as a prophylactic and therapeutic agent for renal sclerosis, myelofibrosis, and diabetic vascular disorders.
  • the antibodies of the present invention can be administered as such or as a suitable pharmaceutical composition.
  • the pharmaceutical composition used for the above administration contains the above 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, forcepsels (soft Syrups, emulsions, suspensions and the like.
  • Such a composition is produced by a known method and contains a carrier, diluent or excipient commonly used in the field of pharmaceuticals.
  • a carrier for example, lactose, starch, sucrose, magnesium stearate and the like are used as carriers and excipients for tablets.
  • compositions for parenteral administration include, for example, injections, suppositories, etc., and injections include intravenous, subcutaneous, intradermal, intramuscular and intravenous injections. Shape. 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 examples include physiological saline, isotonic solutions containing glucose and other adjuvants, and suitable solubilizing agents such as alcohol (eg, ethanol) and polyalcohol ( For example, propylene glycol, polyethylene glycol), nonionic surfactant [for example, polysorbate 80, HCO- 50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)].
  • alcohol eg, ethanol
  • polyalcohol for example, propylene glycol, polyethylene glycol
  • nonionic surfactant for example, polysorbate 80, HCO- 50 (polyoxyethylene (50 mol) adduct of hydrogenated castor 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 in combination as a solubilizer.
  • the prepared injection is usually filled in an appropriate ampoule.
  • Suppositories used for rectal administration are prepared by mixing the above antibody or a salt thereof with a usual base for suppositories.
  • the above-mentioned oral or parenteral pharmaceutical composition is conveniently prepared in a unit dosage form so as to be compatible with the dosage of the active ingredient.
  • dosage unit dosage forms include tablets, pills, capsules, injections (ampoules), suppositories and the like.
  • each dosage unit dosage form is 5 to 500 mg, particularly 5 to 100 mg for injection.
  • Other dosage forms preferably contain 10-25 Omg of the antibody.
  • Each of the above-mentioned compositions may contain another active ingredient as long as the composition does not cause an undesirable interaction 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, porcupine, sheep, pig, etc.). It can be given orally or parenterally (eg, intravenously) to the evening, cats, cats, dogs, monkeys, etc.).
  • the dosage varies depending on the administration subject, target disease, symptoms, administration route, and the like.For example, when used for the treatment or prevention of pulmonary emphysema in adults, the antibody of the present invention is usually administered in a single dose.
  • 0.1 1 to 20 mg / kg body weight preferably 0.1 to 1 OmgZ kg body weight, more preferably 0.1 to 5 mg / kg body weight, about 1 to 5 times a day, preferably 1 to 3 times a day It is convenient to administer the compound as an injection at a time. 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 may be used, for example, for respiratory diseases [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, lung Fibrosis, etc.), rhinitis (e.g., allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry proctitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc.
  • respiratory diseases eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, lung Fibrosis, etc.
  • rhinitis e.g., allergic rhinit
  • Immune diseases eg, myasthenia gravis, glomerulonephritis, multiple sclerosis, siedarenosis
  • Diagnostic group insulin resistant diabetes mellitus, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, immunodeficiency associated with spleen dysfunction or thymic abnormalities), inflammatory bowel disease, allergic conjunctivitis, etc. It is also useful.
  • procollagen C-terminal cleavage promoting activity or a salt thereof The “prophylactic / therapeutic agent for chronic obstructive pulmonary disease or bronchial asthma comprising a compound having a function of regulating procollagen C-terminal cleavage promoting activity or a salt thereof” of the present invention
  • the “compound having an effect of regulating the activity of promoting procollagen C-terminal cleavage” may be any compound having an activity of regulating the activity of promoting procollagen C-terminal cleavage.
  • chronic obstructive pulmonary disease eg, Chronic bronchitis, emphysema
  • diffuse panbronchiolitis bronchial asthma
  • cystic fibrosis irritable pneumonia
  • immune disease eg, myasthenia gravis, glomerulonephritis, multiple sclerosis, siedalen syndrome, insulin resistance
  • inflammatory diabetes rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, immunodeficiency associated with spleen dysfunction or thymic abnormalities
  • inflammatory bowel disease allergic conjunctivitis, etc.
  • the prophylactic / therapeutic agent is produced in the same manner as described above.
  • the present invention relates to 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 having the formula:
  • Non-human mammals having the exogenous DNA of the present invention or the mutant DNA thereof include unfertilized eggs, fertilized eggs, germ cells containing spermatozoa and their primordial cells, and the like.
  • embryo development in non-human mammal development During the raw stage (more preferably, at the single cell or fertilized egg cell stage and generally before the 8-cell stage), calcium phosphate, electric pulse, lipofection, coagulation, microinjection, particle gun, DEAE-dextran, etc. It can be produced by transferring the target DNA.
  • the exogenous DNA of the present invention can be transferred to somatic cells, organs of living organisms, tissue cells, and the like, and used for cell culture, tissue culture, and the like.
  • the DNA-transferred animal of the present invention can also be produced by fusing the above-mentioned germ cells with a cell fusion method known per se.
  • mice for example, porcupines, pigs, higgins, goats, magpies, dogs, cats, guinea pigs, hamsters, mice, rats and the like are used.
  • mammals in a recombinant vector that can be expressed in mammals include humans and the like in addition to the above-mentioned non-human mammals.
  • the exogenous DNA of the present invention refers not to the DNA of the present invention originally possessed by a non-human mammal but to the DNA of the present invention once isolated and extracted from a 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, substitution of another base, etc. DNA that has been used is used, and abnormal DNA is also included.
  • the abnormal DNA refers to a DNA that expresses an abnormal protein of the present invention.
  • a DNA that expresses a protein that suppresses the function of a normal protein of the present invention is used.
  • the exogenous DNA of the present invention may be derived from a mammal that is the same or different from the animal of interest.
  • a DNA construct linked to a downstream of a promoter capable of expressing the DNA in animal cells is used. It is generally advantageous to use it as a struct.
  • DNs derived from various mammals eg, egrets, dogs, cats, guinea pigs, hamsters, rats, mice, etc.
  • a DNA-transferred mammal that highly expresses the DNA of the present invention can be created.
  • 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 or a baculovirus. Animal viruses and the like are used. Among them, a plasmid derived from Escherichia coli, a plasmid derived from Bacillus subtilis or a plasmid derived from yeast are preferably used.
  • Examples of the promoter which regulates the expression of D ⁇ are, for example, a promoter of DNA derived from 1 virus (eg, Simian virus, cytomegalovirus, Moroni leukemia virus, JC virus, breast cancer virus, poliovirus, etc.).
  • a promoter of DNA derived from 1 virus eg, Simian virus, cytomegalovirus, Moroni leukemia virus, JC virus, breast cancer virus, poliovirus, etc.
  • Promoters derived from various mammals such as albumin, insulin II, peropkin II, eras ichie, erythropoietin, endothelin, Muscle creatine kinase, glial fibrillary acidic protein, daltathione S-transferase, platelet-derived growth factor i3, keratin Kl, 10 and 1 ⁇ 14, collagen type I and II, cyclic AMP Dependent protein kinase / 3 I subunit, dystrophy Protein, tartrate-resistant alkaline phosphatase, atrial natriuretic factor, endothelial receptor oral synthase (generally abbreviated as Tie 2), sodium potassium adenosine 3 kinase (Na, K-ATPase), neurofilament Light chain, meta-oral thionein I and IIA
  • cytomegalovirus promoter capable of high expression throughout the whole body, a human peptide chain elongation factor 1a; (EF-1a) promoter, a human and a chicken j8 actin promoter, and the like are preferable.
  • the vector preferably has a sequence that terminates the transcription of the messenger RNA of interest in the DNA-transferred mammal (generally called terminator).
  • terminator For example, DNs derived from viruses and various mammals can be used.
  • the sequence of A can be used, and preferably, SV40 terminator of simian virus or the like is used.
  • the splicing signal of each DNA, the enhancer region, a part of the intron of eukaryotic DNA, etc. are added 5 'upstream of the promoter region, between the promoter region and the translation region, in order to further express the target exogenous DNA.
  • it can be linked to the 3 'downstream of the translation region depending on the purpose.
  • the normal translation region of the protein of the present invention is DNA derived from liver, kidney, thyroid cells, and fibroblasts derived from humans or various mammals (eg, rabbit, dog, cat, guinea pig, hamster, rat, mouse, etc.). And all or part of the genomic DNA from commercially available genomic DNA libraries, or the complementary DNA prepared by known methods from liver, kidney, thyroid cells, and fibroblast-derived RNA. Can be done.
  • an exogenous abnormal DNA can produce a translation region obtained by mutating the translation region of a normal protein obtained from the above cells or tissues by point mutagenesis.
  • the translation region can be prepared as a DNA construct that can be expressed in a transgenic animal by a conventional DNA engineering technique in which the translation region is ligated downstream of the promoter and, if desired, upstream of the transcription termination site.
  • the transfer of the exogenous DNA of the present invention at the fertilized egg cell stage is dependent on the germ cell of the target mammal. Vesicles and somatic cells.
  • the presence of the exogenous DNA of the present invention in the germinal cells of the animal after transfer of the DNA indicates that the exogenous DNA of the present invention is retained in all of the germ cells and somatic cells of the offspring of the animal. Means to do.
  • the offspring of such animals that have inherited the foreign DNA of the present invention have the foreign DNA of the present invention in all of their germ cells and somatic cells.
  • the non-human mammal to which the exogenous normal DNA of the present invention has been transferred is confirmed to stably retain exogenous DNA by mating, and can be subcultured as a DNA-bearing animal in a normal breeding environment. I can do it.
  • Transfer 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 transgenic animal after the DNA transfer indicates that all the offspring of the produced animal will have excessively exogenous DNA of the present invention in all of the 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 eventually promotes the function of the endogenous normal DNA. It may develop hyperfunction and can be used as a model animal for the disease. For example, using the normal DNA-transferred animal of the present invention, elucidation of the pathological mechanism of the protein hyperactivity of the present invention and diseases associated with the protein of the present invention, and examination of treatment methods for these diseases. It is possible.
  • the mammal into which the exogenous normal DNA of the present invention has been transferred has an increased symptom of the released protein of the present invention, it is a prophylactic / therapeutic agent for a disease associated with the protein of the present invention.
  • Respiratory disease eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.
  • rhinitis eg, Allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, Atrophic rhinitis, dry rhinitis, vasomotor rhinitis, gangrenous rhinitis, sinusitis, etc.
  • immune disorders eg, myasthenia gravis, glomerulonephritis, multiple sclerosis, siedalen syndrome, insulin resistance
  • the 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. . Further, the desired foreign 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 a usual DNA engineering technique. The transfer 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 produced animal after the transfer of the DNA means that all the offspring of the produced animal have the abnormal DNA of the present invention in all of the germinal 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 germinal and somatic cells.
  • a homozygous animal having the introduced DNA on both homologous chromosomes is obtained, and by crossing the male and female animals, it is possible to breed 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 the endogenous normal DNA by inhibiting the function of the endogenous normal DNA. It may be functionally inactive refractory and can be used as a model animal for the disease. For example, using the abnormal DNA-transferred animal of the present invention, it is possible to elucidate the pathological mechanism of the protein inactive refractory of the protein of the present invention and to consider a method for treating this disease.
  • the abnormal DNA-highly expressing animal of the present invention is useful for inhibiting the function of a normal protein by the abnormal protein of the present invention (dominant negative effect) in the inactive refractory disease of the protein of the present invention. ) Is a model for elucidating. Further, since the mammal to which the foreign abnormal DNA of the present invention has been transferred has an increased symptom of the released protein of the present invention, the agent for preventing or treating the protein of the present invention or the functionally inactive refractory disease is used.
  • respiratory diseases eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.
  • rhinitis eg, Allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry anterior rhinitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc.
  • immune diseases eg, myasthenia gravis
  • Asthenia glomerulonephritis, multiple sclerosis, Sheddalen syndrome, insulin resistance diabetes, 'rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, spleen dysfunction or Thymus
  • immune diseases
  • the protein of the present invention can be specifically analyzed by directly analyzing the DNA or RNA in the tissue of the DNA-transferred animal of the present invention or by analyzing the peptide tissue expressed by DNA. Analysis of the relationship with expressed or activating peptides, 3 culturing cells of DNA-containing tissue by standard tissue culture techniques, and using these to study the function of cells from tissues that are generally difficult to culture,
  • the DNA-transferred animal of the present invention it is possible to examine the clinical symptoms of a disease associated with the protein of the present invention, including a functionally inactive refractory disease of the protein of the present invention. More detailed pathological findings in each organ of the protein-related disease model can be obtained, which can contribute to the development of new treatment methods and the research and treatment of secondary diseases caused by the disease.
  • each organ is removed from the DNA-transferred animal of the present invention, and after minced.
  • the proteolytic enzyme of the present invention it is possible to obtain free DNA transfer cells, culture them, or systematize the cultured cells.
  • a therapeutic agent for a disease associated with the protein of the present invention including a functionally inactive refractory disease of the protein of the present invention
  • using the DNA-transferred animal of the present invention It is possible to provide an effective and rapid screening method for the therapeutic agent for the disease by using a method or the like.
  • using the transgenic animal of the present invention or the exogenous DNA expression vector of the present invention it is possible to examine and develop a method for treating DNA associated with the protein of the present invention.
  • the present invention provides a non-human mammalian embryonic stem cell in which the DNA of the present invention has been inactivated, and a non-human mammal deficient in expression of the DNA of the present invention.
  • the DNA is inactivated by introducing a repo overnight gene (eg, a j8-galactosidase gene derived from Escherichia coli), and the repo overnight gene controls the promoter for the DNA of the present invention.
  • a repo overnight gene eg, a j8-galactosidase gene derived from Escherichia coli
  • the non-human mammal according to item (6) which can be expressed under
  • test compound is administered to the animal described in (7), and the reporter gene expression And a method for screening a compound or a salt thereof that promotes or inhibits the promoter activity of the DNA of the present invention, characterized by detecting the following.
  • a non-human mammalian embryonic stem cell in which the DNA of the present invention has been inactivated is an artificially mutated DNA of the present invention possessed by the non-human mammal, which suppresses the expression ability of the DNA.
  • the DNA has substantially no ability to express the protein of the present invention (hereinafter referred to as the knockout DNA of the present invention).
  • This may be referred to as a non-human mammalian embryonic stem cell (hereinafter abbreviated as ES cell).
  • non-human mammal the same one as described above is used.
  • the DNA of the present invention can be artificially mutated by, for example, deleting part or all of the DNA sequence, or inserting or substituting another DNA sequence by a genetic engineering method. With these mutations, the knockout DNA of the present invention may be prepared, for example, by shifting the codon reading frame or disrupting the function of the promoter or exon.
  • Non-human mammalian embryonic stem cells in which the DNA of the present invention has been inactivated include:
  • the DNA of the present invention possessed by the target non-human mammal is isolated, and its exon portion is a drug resistance gene typified by a neomycin resistance gene, a hygromycin resistance gene, or 1acZ (i3-galactosidase gene), cat (Chloramphenicol acetyltransferase gene) Insertion of a repo-specific gene, such as the representative gene, disrupts the function of exons or terminates gene transcription in the intron between exons.
  • a DNA chain having a DNA sequence constructed so as to disrupt the gene (hereinafter abbreviated as a targeting vector) is introduced into the chromosome of the animal by, for example, a homologous recombination method.
  • Southern hybridization analysis using a DNA sequence on or near the DNA of the present invention as a probe, or preparation of DNA sequence on targeting vector and evening targeting vector The DNA sequence can be obtained by analyzing the DNA sequence of a neighboring region other than the DNA of the present invention used as a primer by PCR and selecting the knockout ES cells of the present invention.
  • ES cells from which the DNA of the present invention is inactivated by the homologous recombination method or the like for example, those already established as described above may be used, or according to the known Evans and Kaufma method. It may be a newly established one.
  • 129 ES cells are generally used, but since the immunological background is not clear, it is an alternative pure immunological and genetic background.
  • BDFi mice C57BL / 6 and DBAZ2 It can be used satisfactorily with those established using F.
  • BD Fi mice have the advantage of high number of eggs collected and the robustness of eggs, and also have the background of C57BL / 6 mice.
  • ES cells obtained by using C57BL / 6 mice are advantageous in that when they create a disease model mouse, their genetic background can be replaced by C57BL / 6 mice by backcrossing with C57BL / 6 mice.
  • ES cells When ES cells are established, generally 3.5 days after fertilization Although blastocysts of the eye are used, a large number of early embryos can be obtained efficiently by collecting and culturing the 8-cell stage embryos up to the blastocysts.
  • male ES cells are generally more convenient for producing germline chimeras. It is also desirable to discriminate between males and females as soon as possible in order to reduce the complexity of culturing.
  • An example of a method for determining the sex of ES cells is a method of amplifying and detecting a gene in the sex-determining region on the Y chromosome by PCR.
  • the number of ES cells in one colony (about 50) can be reduced, compared to about 10 6 cells for karyotype analysis.
  • the primary selection of ES cells in the early stage of culture can be performed by discriminating between males and females, and the early stages of culture can be greatly reduced by enabling the selection of male cells at an early stage.
  • Examples of the second selection include chromosomes by the G-banding method. This can be done by checking the number.
  • Embryonic stem cell lines obtained in this way usually have very good proliferative properties, but must be subcultured carefully since they tend to lose their ontogenetic potential. For example,
  • a CO2 incubator preferably 5% CO2, 95% air or 5% oxygen, 5%
  • a suitable feeder cell such as STO fibroblasts in the presence of LIF (1 to 10,000 U / ml) Culture at about 37 ° C in carbon dioxide gas, 90% air, etc.
  • trypsin ZE DTA solution usually 0.001-0.5% trypsin / 0.1 l
  • mM EDTA preferably, about 0.1% trypsin / ImM EDTA
  • Such subculture is usually performed every 113 days. At this time, it is desirable to observe the cells and, if any morphologically abnormal cells are found, discard the cultured cells.
  • ES cells are differentiated into various types of cells, such as parietal, visceral, and cardiac muscle, by monolayer culture up to high density or suspension culture until cell clumps are formed under appropriate conditions.
  • monolayer culture up to high density or suspension culture until cell clumps are formed under appropriate conditions.
  • DNA-deficient cell of the present invention obtained by differentiating the ES cell of the present invention is a cell biology of the protein of the present invention in the mouth. Is useful in technical studies.
  • the non-human mammal deficient in DNA expression of the present invention can be distinguished from a normal animal by measuring the mRNA level of the animal using a known method and indirectly comparing the expression level. .
  • non-human mammal the same one as described above is used.
  • the non-human mammal deficient in DNA expression of the present invention can be prepared, for example, by introducing the targeting vector prepared as described above into mouse embryonic stem cells or mouse egg cells, A homologous set in which a DNA sequence in which the DNA of the present invention is inactivated, which is one of the targeting vectors, replaces the DNA of the present invention on the chromosome of mouse embryonic stem cells or mouse egg cells by gene homologous recombination. By performing the replacement, the DNA of the present invention can be knocked out.
  • Cells in which the DNA of the present invention has been knocked out are subjected to Southern hybridization analysis using DNA sequences on or near the DNA of the present invention as a probe or DNA sequences on a targeting vector and targeting DNA.
  • the determination can be made by PCR analysis using a mouse as a primer and a DNA sequence in the vicinity region other than the DNA of the present invention derived from the mouse used in the vector.
  • the DNA of the present invention is inactivated by the homologous recombination to clone a follicular H follicle strain, and the cell is cultured at an appropriate time, for example, at the 8-cell stage.
  • the chimeric embryo is injected into a human mammalian embryo or blastocyst, and the resulting chimeric embryo is implanted into the uterus of the pseudopregnant non-human mammal.
  • the produced animal is a chimeric animal composed of both a normal cell having the DNA locus of the present invention and an artificially mutated cell having the DNA locus of the present invention.
  • all tissues are artificially mutated from a population obtained by crossing such a chimeric individual with a normal individual. It can be obtained by selecting individuals composed of cells having the added DNA locus of the present invention by, for example, determining the coat color.
  • the individual thus obtained is usually an individual having a heterozygous expression of the protein of the present invention, which is crossed with an individual having a heterozygous expression of the protein of the present invention. Can be obtained.
  • a transgenic non-human mammal having a targeting vector introduced into a chromosome can be obtained by injecting a DNA solution into the egg cell nucleus by a microinjection method. Compared to a genic non-human mammal, it can be obtained by selecting those having a mutation at the DNA locus of the present invention by homologous recombination of the gene.
  • the animal individual obtained by mating is usually bred with the nuclear recognition that the DNA has been knocked out. Breeding can be carried out in the environment.
  • the germ line can be obtained and maintained according to a standard method. That is, by crossing male and female animals having the inactivated DNA, a homozygote having the inactivated DNA in both homologous chromosomes can be obtained. Animals can be obtained. The obtained homozygous animal can be efficiently obtained by rearing it in a mother animal in such a manner that there are 1 normal individual and a plurality of homozygotes. By crossing male and female heterozygous animals, homozygous and heterozygous animals having the inactivated DNA are bred and passaged.
  • the non-human mammalian embryonic stem cells in which the DNA of the present invention has been inactivated are extremely useful for producing the non-human mammal deficient in expressing the DNA of the present invention.
  • the non-human mammal deficient in expression of the DNA of the present invention lacks various biological activities that can be induced by the protein of the present invention, it causes inactivation of the biological activity of the protein of the present invention. Since it can be a model of disease, it is useful for investigating the causes of these diseases and examining treatment methods.
  • the non-human mammal deficient in DNA expression of the present invention can be used for screening for a compound having a therapeutic / preventive effect against diseases caused by deficiency or damage of the DNA of the present invention.
  • the present invention comprises administering a test compound to a non-human mammal deficient in expressing the DNA of the present invention, and observing and measuring changes in the animal.
  • Diseases caused by, for example, respiratory diseases eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.
  • Rhinitis eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrenous rhinitis, sinusitis, etc.
  • immune diseases examples, myasthenia gravis, glomerulonephritis, multiple sclerosis, siedalen syndrome, insulin resistance diabetes, rheumatoid arthritis, system
  • Examples of the non-human mammal deficient in DNA expression of the present invention used in the screening method include those described above.
  • Test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and plasma, and these compounds are novel compounds. Or a known compound.
  • a non-human mammal deficient in expression of the DNA of the present invention is treated with a test compound, compared with a non-treated control animal, and changes in organs, tissues, disease symptoms, etc. of the animal are used as indices. Therapeutic and prophylactic effects of test compounds can be tested.
  • test compound for example, oral administration, intravenous injection and the like are used, and it can be appropriately selected according to the symptoms of the test animal, the properties of the test compound, and the like.
  • the dose of the test compound can be appropriately selected according to the administration method, the properties of the test compound, and the like.
  • respiratory diseases eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.
  • rhinitis eg, allergy
  • Rhinitis hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dryness proprietary rhinitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc.
  • immune diseases eg, myasthenia gravis, Glomerulonephritis, multiple sclerosis, siedalen syndrome, insulin resistance diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, immunodeficiency associated with splenic dysfunction or thymic abnormalities
  • test compound when the test compound is administered to a test animal, the disease symptom of the test animal is improved by about 10% or more, preferably about 30% or more, more preferably about 50% or more.
  • Test compound has therapeutic and preventive effects on the above diseases Can be selected as the compound.
  • the compound obtained by using the screening method is a compound selected from the above-described test compounds, and has a therapeutic / preventive effect on a disease caused by a deficiency or damage of the protein of the present invention.
  • compounds derived from the compounds obtained by the above screening can be used in the same manner.
  • the compound obtained by the screening method may form a salt.
  • the salt of the compound include physiologically acceptable acids (eg, inorganic acids, organic acids, etc.) and bases (eg, alkali metal And the like, and especially preferred are physiologically acceptable acid addition salts.
  • physiologically acceptable acids eg, inorganic acids, organic acids, etc.
  • bases eg, alkali metal And the like, and especially preferred are physiologically acceptable acid addition salts.
  • physiologically acceptable acids eg, inorganic acids, organic acids, etc.
  • bases eg, alkali metal And the like, and especially preferred are physiologically acceptable acid addition salts.
  • such salts include salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.), and organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, Salts with succinic acid, tartaric acid, citric acid, malic acid, ox
  • a drug containing the compound or a salt thereof obtained by the screening method can be produced in the same manner as the above-mentioned drug containing the protein of the present invention.
  • the preparations obtained in this way are safe and low toxic and can be used, for example, in humans or mammals (eg, rats, mice, guinea pigs, egrets, sheep, pigs, pigs, dogs, cats, dogs, monkeys). Etc.).
  • the dose of the compound or a salt thereof varies depending on the target disease, the subject of administration, the administration route, and the like.
  • the compound when administered, it is generally used in adult (as a body weight of 60 kg) emphysema patients.
  • the single dose of the compound when administered parenterally, the single dose of the compound varies depending on the administration subject, target disease, and the like.
  • the compound is usually administered in the form of an injection to an adult (with a body weight of 6 O kg).
  • the present invention provides a compound which promotes or inhibits the activity of a promoter for DNA of the present invention, which comprises administering a test compound to a non-human mammal deficient in expressing the DNA of the present invention and detecting the expression of a reporter gene. Or a method for screening a salt thereof.
  • the non-human mammal deficient in expression of the DNA of the present invention may be such that the DNA of the present invention is inactivated by introducing a reporter gene among the non-human mammals deficient in expressing the DNA of the present invention.
  • a gene whose reporter gene can be expressed under the control of a promoter for the DNA of the present invention is used.
  • Examples of the test compound include the same compounds as described above.
  • reporter gene As the reporter gene, the same ones as described above are used, and a / 3-galactosidase gene (1 ac Z), a soluble alkaline phosphatase gene or a luciferase gene is preferable.
  • the reporter gene is under the control of the promoter for the DNA of the present invention, so that the substance encoded by the reporter gene may By tracing the expression, the activity of the promoter can be detected.
  • the tissue of the present invention that originally expresses the protein of the present invention J3_galactosidase is expressed instead of this protein. Therefore, for example, the present invention can be easily carried out by staining with a reagent which is a substrate of 3-galactosidase, such as 5-bromo-4-chloro-3-indolyl / 3 / 3-galactopyranoside (X-ga1). It is possible to observe the state of expression of this protein in animal organisms.
  • a protein-deficient mouse of the present invention or a tissue section thereof is fixed with dartalaldehyde or the like, washed with a phosphate buffered saline (PBS), and then stained with Xga1 at room temperature or at 37 ° C. After reacting for about 30 minutes to 1 hour at around ° C, wash the tissue specimen with ImM EDTA / PBS solution. Therefore, it is sufficient to stop the] S-galactosidase reaction and observe the coloration. Further, mRNA encoding 1 ac Z may be detected according to a conventional method.
  • PBS phosphate buffered saline
  • the compound or a salt thereof obtained by using the above-mentioned screening method is a compound selected from the above-mentioned test compounds, and is a compound that promotes or inhibits the promoter activity for DNA of the present invention.
  • the compound obtained by the screening method may form a salt.
  • the salt of the compound include physiologically acceptable acids (eg, inorganic acids) and bases (eg, alkali metals). And the like, and especially preferred are physiologically acceptable acid addition salts.
  • physiologically acceptable acids eg, inorganic acids
  • bases eg, alkali metals
  • physiologically acceptable acid addition salts include inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.).
  • a salt with an organic acid eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc.
  • organic acid eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, succinic acid, tarta
  • the compound or a salt thereof that promotes or inhibits the promoter activity of the DNA of the present invention or the salt thereof can regulate the expression of the protein of the present invention and regulate the function of the protein.
  • Diseases chronic bronchitis, emphysema
  • diffuse panbronchiolitis bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.
  • rhinitis eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis
  • immune diseases eg, myasthenia gravis, glomerulonephritis, multiple sclerosis, siedalen syndrome
  • Insulin resistance diabetes rheumatoid arthritis, systemic lupus erythematosus
  • a drug containing a compound or a salt thereof obtained by the screening method can be produced in the same manner as the above-mentioned drug containing a protein of the present invention or a salt thereof.
  • the preparations obtained in this way are safe and of low toxicity, for example human or It can be administered to mammals (for example, rats, mice, guinea pigs, egrets, sheep, sheep, bush, horses, horses, cats, dogs, monkeys, etc.).
  • mammals for example, rats, mice, guinea pigs, egrets, sheep, sheep, bush, horses, horses, cats, dogs, monkeys, etc.
  • the dose of the compound or a salt thereof varies depending on the disease to be treated, the subject of administration, the administration route, and the like.
  • the compound is administered at about 0.1 to 100 mg, preferably about 1.0 to 50 mg, more preferably about 1.0 to 20 mg per day.
  • the single dose of the compound varies depending on the administration subject, target disease, and the like.
  • a compound that inhibits the promoter activity against DNA of the present invention may be in the form of an injection.
  • the compound when administered to an adult (assuming a body weight of 60 kg) emphysema patients, the compound is administered daily at about 0.01-3 Omg, preferably about 0.1-2 Omg, more preferably about 0.1-: L per day. It is convenient to administer 0 mg by intravenous injection. In the case of other animals, the dose can be administered in terms of weight per 60 kg.
  • the non-human mammal deficient in expression of the DNA of the present invention is extremely useful for screening a compound or a salt thereof which promotes or inhibits the activity of the promoter of the DNA of the present invention overnight. Yes, it can greatly contribute to the investigation of the causes of various diseases caused by DNA expression deficiency of the present invention or the development of preventive and therapeutic agents.
  • transgenic animal using a DNA containing the promoter region of the protein of the present invention, genes encoding various proteins are ligated downstream thereof and injected into egg cells of an animal to produce a so-called transgenic animal (transgenic animal). ) Makes it possible to specifically synthesize the protein and examine its action in living organisms. Furthermore, by linking an appropriate reporter gene to the above promoter portion and establishing a cell line that expresses the reporter gene, a low-molecular compound having an action of specifically promoting or suppressing the production ability of the protein itself of the present invention in the body. Can be used as a search system. (9) a medicine containing the protein of the present invention
  • the protein of the present invention is defective or defective in the DNA encoding the protein of the present invention, or the protein of the present invention is defective. If the expression level of is reduced, for example, Respiratory disease (eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.), rhinitis (eg, a Rarginous rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pre-rhinitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc., immune diseases (eg, myasthenia gravis) Disease, glomerulonep
  • the protein of the present invention and the DNA of the present invention include, for example, respiratory diseases [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, Hypersensitivity pneumonia, pulmonary fibrosis, etc.), rhinitis (eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pro-rhinitis, vasomotor rhinitis, gangrene rhinitis, Sinusitis, etc., immune diseases (eg, myasthenia gravis, glomerulonephritis, multiple sclerosis, siedalen syndrome, insulin resistant diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, It can be used
  • a prophylactic / therapeutic agent for a respiratory disease or the like is a prophylactic / therapeutic agent for a respiratory disease or the like, more preferably a prophylactic / therapeutic agent for a chronic obstructive pulmonary disease or the like, and further preferably a prophylactic / therapeutic agent for an emphysema or the like.
  • the protein of the present invention when there is a patient in whom the protein of the present invention is reduced or deficient in a living body, (a) by administering the DNA of the present invention to the patient and expressing the protein of the present invention in the living body, (Mouth) inserting the DNA of the present invention into cells and expressing the protein of the present invention, and then transplanting the cells into a patient; or (8) administering the protein of the present invention to the patient.
  • the role of the protein of the present invention in the patient can be sufficiently or normally exerted.
  • the DNA of the present invention When the DNA of the present invention is used as the above-mentioned prophylactic or therapeutic agent, the DNA may be used alone or after being inserted into an appropriate vector such as a retrovirus vector, an adenovirus vector, an adenovirus associated virus vector, or the like. , According to conventional means And can be administered to humans or warm-blooded animals.
  • 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 adjuvant for promoting uptake, and administered with a gene gun or a catheter such as a hydrogel catheter.
  • the protein of the present invention When the protein of the present invention is used as the above-mentioned 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, or the like, which are sugar-coated as required, orally, or aseptic with water or other pharmaceutically acceptable liquids. It can be used parenterally in the form of injections, such as solutions or suspensions.
  • the protein of the present invention is mixed with physiologically acceptable carriers, flavors, excipients, vehicles, preservatives, stabilizers, binders, etc. in a unit dosage form generally required for the practice of preparations. It can be manufactured by 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, alginic acid, etc. Swelling agents such as magnesium stearate, sweeteners such as sucrose, lactose or saccharin, and flavoring agents such as peppermint, cocoa oil or cherry.
  • the unit dosage form is forcepsel, the above type of material may further contain a liquid carrier such as oil and fat.
  • Sterile compositions for injection can be formulated according to standard pharmaceutical practice, such as by dissolving or suspending the active substance in a vehicle such as water for injection, or naturally occurring vegetable oils such as sesame oil or coconut oil. it can.
  • Aqueous solutions for injection include, for example, physiological saline, isotonic solutions containing glucose and other adjuvants (eg, D-sorbyl), D-manni! And suitable solubilizers, for example, alcohols (eg, ethanol), polyalcohols (eg, propylene glycol, polyethylene glycol, etc.), nonionic surfactants (polysorbate 80 TM, HCO — 5 0 Etc.).
  • suitable solubilizers for example, alcohols (eg, ethanol), polyalcohols (eg, propylene glycol, polyethylene glycol, etc.), nonionic surfactants (polysorbate 80 TM, HCO — 5 0 Etc.).
  • suitable solubilizers for example, alcohols (eg, ethanol), polyalcohols (eg, propylene glycol, polyethylene glycol, etc.), nonionic surfactants (polysorbate 80 TM, HCO — 5 0 Etc.).
  • buffers eg, phosphate buffer, sodium acetate buffer, etc.
  • soothing agents eg, benzalkonium chloride, proforce hydrochloride, etc.
  • stabilizers eg, human serum albumin, polyethylene glycol, etc.
  • preservatives eg, benzyl alcohol, phenol, etc.
  • 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 (eg, humans, rats, mice, guinea pigs, egrets, birds, birds, higgies, bushus, dogs, dogs, 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 protein of the present invention is orally administered for the purpose of treating infertility, it is generally required for an adult (body weight 6).
  • about 0.1 to 10 Omg preferably about 1.0 to 5 Omg, more preferably about 1.0 to 20 mg of the protein is administered per day.
  • a single dose of the protein may vary depending on the administration subject, target disease, and the like.
  • the protein per day is about 0.01 to 3 O mg, preferably about 0.1 to 2 O mg, more preferably about 0.1 to 1 O mg per day.
  • the dose can be administered in terms of the body weight of 60 kg.
  • bases, amino acids, and the like are represented by abbreviations based on the abbreviations by IUPAC-IUB Communication on Biochemical Nomenclature or commonly used abbreviations in the art, and examples thereof are described below.
  • amino acids can have optical isomers, the L-form is indicated unless otherwise specified.
  • DNA Deoxyribonucleic acid
  • RNA Messenger liponucleic acid dATP Deoxyadenosine triphosphate dTTP Deoxythymidine triphosphate dGTP Deoxyguanosine triphosphate d CTP Deoxycytidine triphosphate
  • HONB trihydroxy-5-norpolene-2,3-dicarpoxyimide
  • the amino acid sequence of human procollagen C-protease enhancer protein 2 is shown.
  • mouse procollagen C-protease enhancer protein 2 The amino acid sequence of mouse procollagen C-protease enhancer protein 2 is shown.
  • C57BL / 6N mice (6-week-old, Japan Charls River) were inhaled with mainstream smoke generated from Kentucky Reference Cigarette 1R1 for 1 to 4 hours / day, 5 days / week for a total of 6 months.
  • the Kentucky Reference Cigarette 1R1 was attached to an evening smoke generator (SG-200, Shibata Kagaku Co., Ltd.), and mainstream smoke was collected under the conditions of 35 ml / puff, 10 puff / min, and 25 puif / cigarette.
  • the tracheal force Nyure ventilator connecting the (Harvard Co.), under the diaphragm resection was facilities for 10 minutes ventilator by 99.995% 0 2.
  • the ventilation was adjusted so that the change in airway pressure at that time was 10 cm cm0.
  • the trachea was closed with an arterial clip and degassing was performed in the lungs, and the lungs were removed.
  • a formalin buffer solution was sequentially injected into the isolated lung at a pressure of 0 to 25 cm3 ⁇ 40, and the volume of the lung at every 5 cm3 ⁇ 40 was measured using a prethysmograph (OJgobasil) to obtain a pressure-volume curve of the removed lung.
  • OJgobasil prethysmograph
  • cDNA was synthesized by reverse transcription in a reaction mixture of 501 using TaQMan Gold RT-PCR Kit (manufactured by Applied Biosystems). After diluting the reaction solution 5 times with distilled water, use it for real-time quantitative analysis using ABI PRISM 7900 Sequence Detector (Applied Biosystems) and Quant iTect SYBR Green PCR Kit (QIAGEN). The mouse PC0LCE2 gene copy number was measured by PCR.
  • the primers [Primer 1 (SEQ ID NO: 5) and Primer 2 (SEQ ID NO: 6)] used for the detection of the gene amount were obtained from the Primer Expression program based on the nucleotide sequence of the mouse PC0LCE2 gene [GenBank Accession Number: AF352788]. Designed using As a standard sample for calculating the copy number, total RNA extracted from mouse lung tissue was subjected to RT-PCR by using primers 3 (SEQ ID NO: 7) and primer 14 (SEQ ID NO: 8) for type I. The concentration of the amplified DNA fragment consisting of 515 base pairs was measured with a Spectrophotometer and prepared by serial dilution. Similarly, copy of GAPDH gene as housekeeping gene The number was measured.
  • Mouse cDNA (Mouse MTC panel I and Mouse MTC panel II) for each mouse tissue (bone marrow, lymph node, prostate, thymus, stomach, uterus, heart, brain, spleen, lung, liver, kidney, testis, day 11 embryo): ABI PRISM 7900 Sequence Detector (manufactured by Applied Biosystems) and QuantiTect (Clontech) and cigarette smoke-exposed model mouse lung and control mouse lung cDNA (mixed group treated for 1 month, 3 months and 6 months) Mouse PC0LCE gene expression distribution was examined by real-time quantitative PCR using SYBR Green PC Kit (QIAGEN).
  • the primers [Primer 1 (SEQ ID NO: 5) and Primer 2 (SEQ ID NO: 6)] used for the detection of the gene amount were designed using the Primer Express program from the mouse PC0LCE2 gene base sequence CGenBank Accession Number: AF352788].
  • As a standard sample for calculating the copy number total RNA extracted from mouse lung tissue was subjected to RT-PCR using primers (SEQ ID NO: 7) and primer 4 (SEQ ID NO: 8) with type III.
  • the concentration of the DNA fragment consisting of 503 base pairs amplified by the above was measured using a Spectrophotometer and prepared by serial dilution.
  • the copy number of the GAPDH gene as a housekeeping gene was measured.
  • samples without reverse transcriptase are treated in the same manner, and the expression level is determined by calculating the number of gene copies per total RNA from the following formula. I asked.
  • mice In the C0PD model, C57BL / 6N mice (8-week-old, Nippon-Charles River) were administered nasal administration of bushu knee elastase solution (Wako Pure Chemical Industries) ⁇ imits O xL / mouse under halothane anesthesia. Produced. In addition, a physiological saline solution nasal mouse was used as a control group. Mouse lung function was evaluated using the pressure-volume curve of the isolated lung. That is, 1, 3, and 7 days after administration of elastase, the mice were anesthetized with pentobarbital (70 mg / kg intraperitoneal injection) 14 days, 21 days and 35 days later, and then the neck was incised and the tracheal force was applied to the trachea.
  • pentobarbital 70 mg / kg intraperitoneal injection
  • a self-flow indwelling needle, 18G was introduced.
  • the tracheal force Nyure the ventilator (Harvard Co., Ltd.) consolidated and subjected to 10 minutes ventilator with 99.995% 0 2 under diaphragmatic resection.
  • the ventilation was adjusted so that the airway load pressure at that time was 10 cm0.
  • the trachea was closed with an arterial clip to perform degassing in the lung, and the lung was removed.
  • formalin buffer was sequentially injected under a load pressure of 0 to 25 cm3 ⁇ 40, and the volume of the lung at intervals of 5 cm3 ⁇ 40 was measured using a lethysmograph (Ugobasil).
  • a lung pressure-volume curve was determined.
  • the compliance value was calculated as an index of the ease of lung expansion according to the following equation.
  • mice were sacrificed by ventalpital anesthesia 1 day, 3 days, 7 days, 14 days, 21 days and 35 days after administration of Elas evening, and the lungs were removed.
  • the removed lung was frozen in liquid nitrogen, crushed with a frozen tissue crusher, and then immersed in IS0GEN (Wako Pure Chemical Industries), which is equivalent to 10 times its wet weight.
  • RNA was prepared using IS0GEN according to the attached manual.
  • RNA Ig prepared from mouse lung tissue as a starting material
  • M-MLV Reverse Transcriptase manufactured by Invitrogen
  • the amount of mouse PC0LCE2 gene was measured by a real-time quantitative PCR method using ABI PRISM 7700 Sequence Detector I (manufactured by Applied Biosystems).
  • the primers [Primer 1 (SEQ ID NO: 5) and Primer 2 (SEQ ID NO: 6)] used for the detection of the gene amount were obtained from the mouse PC0LCE2 gene base sequence CGenBank Accession Number: AE 352788] using the Primer Express program. Designed.
  • the amount of 18S liposomal RNA gene as a housekeeping gene was measured.
  • the PC0LCE2 gene expression amount per 18S liposomal RNA gene expression amount was determined from the following equation, and the expression of each of the lungs of the C0PD model mouse and the control mouse lung induced by Erasase was compared.
  • the proteins and polynucleotides of the present invention include, for example, respiratory diseases [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, sac Alveolar fibrosis, irritable pneumonia, pulmonary fibrosis, etc.), rhinitis (eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrene Rhinitis, sinusitis, etc., immune diseases (eg, myasthenia gravis, glomerulonephritis, multiple sclerosis, siedalen syndrome, insulin resistance diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic skin Inflammation, leukocyte abnormalities, immunodefic
  • Modulators obtained by screening using the antibody, antibodies against the protein, and the like include, for example, respiratory diseases (eg, Chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.), rhinitis (eg, allergic rhinitis, pollinosis, acute) Rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry proprietary rhinitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc., immune diseases (eg, myasthenia gravis, glomerulonephritis, multiple occurrences) Sclerosis, siedalen syndrome, insulin resistance diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leuk
  • the antisense polynucleotide of the present invention can suppress the expression of the protein of the present invention.
  • respiratory diseases eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse Panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.
  • rhinitis eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dryness Pronasalitis, vasomotor rhinitis, gangrenous rhinitis, sinusitis, etc.
  • immune disorders eg, myasthenia gravis, glomerulonephritis, multiple sclerosis, Siedaren syndrome, insulin resistant diabetes, rheumatoid arthritis, Systemic lupus erythematosus, a
  • various polynucleotides of the present invention include, for example, respiratory diseases (eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, Pulmonary fibrosis etc.), rhinitis (eg, allergy Rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc.), immune diseases (eg, myasthenia gravis, Glomerulonephritis, multiple sclerosis, siedalen syndrome, insulin resistance diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, immunodeficiency associated

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Public Health (AREA)
  • Immunology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pulmonology (AREA)
  • Biochemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Zoology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Urology & Nephrology (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • Food Science & Technology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Epidemiology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

It is intended to provide diagnostics/preventives/remedies, etc. for respiratory diseases. Namely, a compound or its salt promoting the activity of a protein having an amino acid sequence which is the same or substantially the same as the amino acid sequence represented by SEQ ID NO:1, a compound or its salt promoting the expression of a gene of the above protein, an antisense nucleotide having a base sequence or a part thereof which is complementary or substantially complementary to the base sequence of a DNA encoding the above protein or its peptide fragment, an antibody against the above protein or its peptide fragment, etc. are usable as diagnostics/preventives/remedies, etc. for respiratory diseases.

Description

明細書  Specification
呼吸器疾患の診断 ·予防 .治療剤 技術分野  Diagnosis and prevention of respiratory diseases
本発明は、 呼吸器疾患の予防 ·治療剤および診断薬などに関する。 背景技術  The present invention relates to an agent for preventing or treating respiratory diseases, a diagnostic agent, and the like. Background art
慢性閉塞性肺疾患、 慢性気管支炎、 肺気腫、 びまん性汎細気管支炎、 内因性喘息 などは、 喫煙世代の高齢化、 平均寿命の延長等にともなって、 今後、 呼吸器疾患の 中心的な病気になると考えられている。  Chronic obstructive pulmonary disease, chronic bronchitis, emphysema, diffuse panbronchiolitis, endogenous asthma, etc. It is thought to be.
喫煙は慢性閉塞性肺疾患の明確な病因になりえることが示されている。喫煙によ り閉塞性障害が進行し、 タバコ本数に依存する。喫煙開始年齢が若年であるほど進 行しやすい。 また、 喫煙と気管支腺の過形成との用量相関が確かめられている。 動物実験においても、タバコを吸入させることによつて気腫性変化を起こし得る ことについては多くの報告がある。  It has been shown that smoking can be a definite etiology of chronic obstructive pulmonary disease. Smoking causes obstructive disorders and depends on the number of cigarettes. The younger the age at which smoking starts, the easier it is to progress. In addition, a dose correlation between smoking and bronchial gland hyperplasia has been confirmed. In animal studies, there are many reports that inhalation of tobacco can cause emphysema.
慢性閉塞性肺疾患 (以下、 C0PDと略称することもある) の病理学的変化は、 中枢 気道、 末梢気道、 肺実質の 3領域で特有の異常が観察される。 中枢気道病変は、 杯 細胞の過形成や粘液下腺における細胞の増生、肥大といった分泌組織の形態変化が みられる。炎症細胞に関しては、 気道粘膜にマクロファージゃ活性化 Tリンパ球の 増加が示されている。細気管支領域の病変は、気道内腔の粘液塞栓や気道上皮での 杯細胞異形成、 気道壁における炎症細胞浸潤、 平滑筋肥厚、 線維化が観察される。 いずれの変化も気道狭窄をもたらす。肺胞実質では、肺胞の破壊消失と気腔の拡大 で定義された肺気腫病変が観察される。 これらには、 プロテアーゼ Zアンチプロテ ァーゼのバランスの不均衡が関与すると考えられている。  Pathological changes in chronic obstructive pulmonary disease (hereinafter sometimes abbreviated as C0PD) include specific abnormalities in the central airway, peripheral airway, and lung parenchyma. Central airway involvement involves changes in secretory tissue morphology, such as goblet cell hyperplasia, hyperplasia of cells in the submucosal gland, and hypertrophy. For inflammatory cells, an increase in macrophage-activated T lymphocytes has been shown in the airway mucosa. Lesions in the bronchiole region include mucus embolism in the airway lumen, goblet cell dysplasia in the airway epithelium, inflammatory cell infiltration in the airway wall, smooth muscle hypertrophy, and fibrosis. Either change results in airway narrowing. In the alveolar parenchyma, emphysema lesions defined by destruction and disappearance of the alveoli and enlargement of the air space are observed. These are thought to involve an imbalance in the protease Z antiprotease balance.
一方、遺伝子発現を網羅的に解析するために、 cDNAまたはオリゴヌクレオチドを 固定化したマイクロアレイ法が開発され、疾患特異的な遺伝子発現の変化を見出す 技術が普及し、 その有用性が確認されている。 例えば、 Aifynietrix社の GeneCMp システムはがんなどの疾患の診断や創薬標的遺伝子の発見に多用されつつある。し 2003/008167 On the other hand, a microarray method in which cDNA or oligonucleotides are immobilized has been developed to comprehensively analyze gene expression, and the technology for finding disease-specific changes in gene expression has become widespread and its usefulness has been confirmed. . For example, Aifynietrix's GeneCMp system is increasingly being used to diagnose diseases such as cancer and to discover drug discovery target genes. I 2003/008167
2  Two
かしながら、 この技術を利用して肺気腫の発症や進展に関与する遺伝子を見出した 報告はない。 However, there have been no reports using this technique to find genes involved in the development or progression of emphysema.
プロコラーゲン C—プロテア一ゼ ェンハンサ一プロテイン 2 (procol lagen C- protease enhancer protein 2, PC0LCE2) は、 PC0LCE1のァイソフォームの一つ としてクローニングされている (ゲノミックス (Genomics) 66巻、 264頁、 2000 年) 。 PC0LCE1は、 C末端側が組織メタ口プロテア一ゼインヒビター (t issue inhibi tor of metaro protease, TIMP) の N末端側と相同性が高いことが報告され (プロテイン サイエン (Protein Sci) 8巻、 1636頁、 1999年) 、 また、実際に、 この C末端部分が翻訳後に切断され、メタ口プロテア一ゼィンヒビターとして作用 することが報告されている(ジャーナル ォブ バイオロジカル ケミストリ一(L Biol. diem. ) 275巻、 1384頁、 2000年) 。 発明の開示  Procollagen C-protease enhancer protein 2, PC0LCE2 has been cloned as one of the isoforms of PC0LCE1 (Genomics, 66, 264, 2000). Year) . It has been reported that PC0LCE1 has a high homology at the C-terminus to the N-terminus of tissue inhibitor of metaro protease (TIMP) (Protein Sci 8, 1636, In 1999, it has been reported that this C-terminal portion is actually cleaved after translation and acts as a meta-oral protease inhibitor (L Biol. Diem.) 275 , 1384, 2000). Disclosure of the invention
副作用の少ない優れた呼吸器疾患 (例、 慢性閉塞性肺疾患など) の予防 ·治療剤 および診断剤の開発が望まれている。  There is a need for the development of prophylactic and therapeutic agents and diagnostic agents for superior respiratory diseases with few side effects (eg, chronic obstructive pulmonary disease, etc.).
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、肺気腫病変 を有する肺組織に発現が顕著に低下する遺伝子を見出し、 この知見に基づいて、 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 a gene whose expression is remarkably reduced in lung tissue having emphysema lesions, and based on this finding, conducted further studies based on this finding As a result, the present invention has been completed.
すなわち、 本発明は、  That is, the present invention
( 1 ) 配列番号: 1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夕ンパク質もしくはその部分べプチドまたはその塩の活性を 促進する化合物またはその塩を含有してなる呼吸器疾患の予防 ·治療剤、  (1) contains a compound or a salt thereof which promotes the activity of a protein or a partial peptide thereof or a salt thereof containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 Prevention and treatment of respiratory diseases
( 2 ) 配列番号: 1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夕ンパク質もしくはその部分べプチドまたはその塩の遺伝子 の発現を促進する化合物またはその塩を含有してなる呼吸器疾患の予防 ·治療剤、 (2) A compound or a salt thereof that promotes the expression of a protein or a partial peptide or a salt thereof containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 Prevention and treatment of respiratory diseases comprising
( 3 ) 配列番号: 1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タンパク質またはその部分べプチドをコードするポリヌクレ ォチドの塩基配列に相補的もしくは実質的に相補的な塩基配列またはその一部分 を含有するァンチセンスポリヌクレオチド、 (3) Complementary or substantially complementary to the nucleotide sequence of a polynucleotide encoding a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof Base sequence or part of it Antisense polynucleotide containing,
(4) 上記 (3) 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、 (4) a medicine comprising the antisense polynucleotide according to the above (3),
(5) 配列番号: 1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タンパク質またはその部分ペプチドまたはその塩に対する抗 体、 (5) an antibody against a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof;
(6) 上記 (5) 記載の抗体を含有してなる医薬、  (6) a medicament comprising the antibody according to the above (5),
( 7 ) 呼吸器疾患の予防 ·治療剤である上記 ( 6 ) 記載の医薬、  (7) The medicament according to the above (6), which is an agent for preventing or treating respiratory diseases.
(8) 上記 (5) 記載の抗体を含有してなる診断薬、  (8) a diagnostic agent comprising the antibody according to (5) above,
(9) 呼吸器疾患の診断薬である上記 (8) 記載の診断薬、  (9) The diagnostic agent according to the above (8), which is a diagnostic agent for respiratory disease,
(10) 配列番号: 1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タンパク質またはその部分ペプチドをコードするポリヌク レオチドを含有してなる呼吸器疾患の診断薬、  (10) a diagnostic agent for a respiratory disease comprising a polynucleotide encoding a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof,
(11) プロコラーゲン C末端切断促進活性を促進する作用を有する化合物また はその塩を含有してなる慢性閉塞性肺疾患または気管支喘息の予防 ·治療剤、 ' (12) 配列番号: 1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タンパク質もしくはその部分ペプチドまたはその塩を用い ることを特徴とする呼吸器疾患の予防 ·治療剤のスクリ一二ング方法、  (11) A prophylactic / therapeutic agent for chronic obstructive pulmonary disease or bronchial asthma, comprising a compound having a procollagen C-terminal truncation promoting activity or a salt thereof, '(12) SEQ ID NO: 1 A method for screening a remedy for the prevention and treatment of respiratory diseases, which comprises using a protein having the same or substantially the same amino acid sequence as the amino acid sequence to be obtained or a partial peptide thereof or a salt thereof;
(12 a) 配列番号: 1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タンパク質もしくはその部分べプチドまたはその塩を用 いることを特徴とする医薬用化合物のスクリ一エング方法、  (12a) A screen for a pharmaceutical compound characterized by using a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof. Eng method,
(12 b) 医薬用化合物が、 呼吸器疾患の予防 ·治療用の化合物、 呼吸器疾患の 予防 ·治療に用いられる化合物および/または呼吸器疾患の予防 ·治療効果を有す る化合物である上記 (12 a) 記載のスクリーニング方法、  (12b) The pharmaceutical compound is a compound for preventing or treating respiratory disease, a compound used for preventing or treating respiratory disease and / or a compound having a preventive or treating effect for respiratory disease. (12a) The screening method according to the above,
(13) タバコ煙曝露慢性閉塞性肺疾患モデルマウスまたはエラス夕一ゼ誘発慢 性閉塞性肺疾患モデルマウスを用いる上記 (12) 記載のスクリーニング方法、 (13) The screening method according to the above (12), wherein the tobacco smoke-exposed chronic obstructive pulmonary disease model mouse or the elastase-induced chronic obstructive pulmonary disease model mouse is used,
(14) 配列番号: 1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タンパク質もしくはその部分べプチドまたはその塩を含有 することを特徴とする呼吸器疾患の予防 ·治療剤のスクリーニング用キット、 (1 5) 上記 (12) 記載のスクリーニング方法または上記 (14) 記載のスク リーニング用キットを用いて得られうる呼吸器疾患の予防 ·治療剤、 (14) Prevention of a respiratory disease characterized by containing a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof · Screening kits for therapeutic agents, (15) A prophylactic / therapeutic agent for a respiratory disease obtainable by using the screening method according to (12) or the screening kit according to (14).
(1 5 a) 上記 (1 2 a) 記載のスクリーニング方法を用いて得られうる医薬用 化合物、  (15a) a pharmaceutical compound obtainable by using the screening method described in (12a) above,
(1 5 b) 医薬用化合物が、 呼吸器疾患の予防 ·治療用の化合物、 呼吸器疾患の 予防 ·治療に用いられる化合物および/または呼吸器疾患の予防 ·治療効果を有す る化合物である上記 (1 5 a) 記載の化合物またはその塩、  (15b) The pharmaceutical compound is a compound for preventing or treating respiratory diseases, a compound used for preventing or treating respiratory diseases and / or a compound having a preventive or treating effect for respiratory diseases. The compound or a salt thereof according to the above (15a),
(1 5 c) 上記 (1 5 b) 記載の化合物またはその塩を含有してなる呼吸器疾患 の予防 ·治療剤、  (15c) a prophylactic / therapeutic agent for respiratory diseases comprising the compound or salt thereof according to (15b) above,
(16) 呼吸器疾患が肺気腫である上記 (15) 記載の予防 ·治療剤、  (16) The prophylactic or therapeutic agent according to (15) above, wherein the respiratory disease is emphysema.
(17) 配列番号: 1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夕ンパク質またはその部分べプチドをコードするポリヌク レオチドを用いることを特徴とする呼吸器疾患の予防 ·治療剤のスクリーニング方 法、  (17) A respiratory tract, characterized by using a polynucleotide encoding a protein or a partial peptide thereof containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1. Disease prevention and treatment screening methods,
(17 a) 配列番号: 1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タンパク質またはその部分ペプチドをコードするポリヌ クレオチドを用いることを特徴とする医薬用化合物のスクリ一二ング方法、 . (17a) A script of a pharmaceutical compound characterized by using a polynucleotide encoding a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof. One way,.
(17 b) 医薬用化合物が、 呼吸器疾患の予防 ·治療用の化合物、 呼吸器疾患の 予防 ·治療に用いられる化合物および/または呼吸器疾患の予防 ·治療効果を有す る化合物である上記 (17 a) 記載のスクリーニング方法、 (17b) The pharmaceutical compound is a compound for preventing or treating respiratory disease, a compound used for preventing or treating respiratory disease and / or a compound having a preventive or treating effect for respiratory disease. (17a) the screening method described above,
(1 8) タバコ煙曝露慢性閉塞性肺疾患モデルマウスまたはエラスターゼ誘発慢 性閉塞性肺疾患モデルマウスを用いる上記 (17) 記載のスクリーニング方法、 (18) The screening method according to the above (17), wherein the tobacco smoke-exposed chronic obstructive pulmonary disease model mouse or elastase-induced chronic obstructive pulmonary disease model mouse is used,
(19) 配列番号: 1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タンパク質またはそ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ことを特徴とする呼吸器疾患の予防 ·治療剤のスクリ一ニング 用キット、 (19) a respiratory disease characterized by containing a protein having an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 or a polynucleotide encoding a partial peptide thereof Kits for screening of preventive and therapeutic agents
(20) 上記 (17) 記載のスクリーニング方法または上記 (1 9) 記載のスク リーニング用キットを用いて得られうる呼吸器疾患の予防 ·治療剤、 (20 a) 上記 (17 a) 記載のスクリーニング方法を用いて得られうる医薬用 化合物、 (20) A prophylactic or therapeutic agent for respiratory disease, which can be obtained by using the screening method according to (17) or the screening kit according to (19). (20a) a pharmaceutical compound obtainable by using the screening method according to (17a),
(2 Ob) 医薬用化合物が、 呼吸器疾患の予防 ·治療用の化合物、 呼吸器疾患の 予防 ·治療に用いられる化合物および/または呼吸器疾患の予防 ·治療効果を有す る化合物である上記 (20 a) 記載の化合物またはその塩、  (2 Ob) The pharmaceutical compound is a compound for preventing or treating respiratory disease, a compound used for preventing or treating respiratory disease and / or a compound having a preventive or treating effect for respiratory disease. (20a) the compound or a salt thereof,
(20 c) 上記 (20 b) 記載の化合物またはその塩を含有してなる呼吸器疾患 の予防 ·治療剤、  (20c) a prophylactic / therapeutic agent for respiratory diseases comprising the compound or salt thereof according to (20b) above,
(21) 呼吸器疾患が肺気腫である上記 (20) 記載の予防 ·治療剤、  (21) The prophylactic or therapeutic agent according to (20), wherein the respiratory disease is emphysema.
(22) 哺乳動物に対し、 配列番号: 1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タンパク質もしくはその部分べプチドま たはその塩の活性を促進する化合物またはその塩、または該タンパク質の遺伝子の 発現を促進する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呼吸器 疾患の予防 ·治療方法、  (22) a compound having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a compound that promotes the activity of a partial peptide or a salt thereof in mammals, or a compound thereof; A method for preventing or treating respiratory diseases, which comprises administering an effective amount of a salt, or a compound or a salt thereof that promotes the expression of a gene of the protein,
(23) 配列番号: 1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タンパク質もしくはその部分べプチドまたはその塩の活性 を促進する、または該タンパク質の遺伝子の発現を促進することを特徴とする呼吸 器疾患の予防 ·治療方法、  (23) Promotes the activity of a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof, or expresses a gene of the protein Prevention and treatment of respiratory diseases characterized by promoting
(24) 呼吸器疾患の予防 ·治療剤を製造するための、 配列番号: 1で表される 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夕ンパク質 もしくはその部分ペプチドまたはその塩の活性を促進する化合物またはその塩、ま たは該タンパク質の遺伝子の発現を促進する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  (24) A protein or a partial peptide thereof containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 for producing a prophylactic or therapeutic agent for respiratory disease. It is intended to provide a compound or a salt thereof which promotes the activity of the salt, or a compound or a salt thereof which promotes the expression of a gene of the protein. BRIEF DESCRIPTION OF THE FIGURES
図 1は、実施例 1で得られたマウス摘出肺の圧-容量曲線を表す図である。図中、 FIG. 1 is a diagram showing a pressure-volume curve of a mouse extirpated lung obtained in Example 1. In the figure,
(a) の— ·一は 1ヶ月間のタバコ煙曝露マウス群 (n = 8) 、 一〇一はコント口 ール群 (n = 7) を、 (b) のー秦一は 3ヶ月間のタバコ煙曝露マウス群 (n= l 0) 、 一〇一はコントロール群 (n = 9) を、 (c) の—秦一は 6ヶ月間のタバコ 煙曝露マウス群 (n=10) 、 —〇一はコントロール群 (n = 9) を示す。 * *は p≤0. 01を、 *は ρ≤0. 05を示す。 In (a), one group was exposed to cigarette smoke for one month (n = 8), one group was in control group (n = 7), and (b) one group was for three months. Group of mice exposed to cigarette smoke (n = l0), 1101 the control group (n = 9), (c) —Hinichi a 6 month cigarette The group of mice exposed to smoke (n = 10) and the group of 〇 indicate the control group (n = 9). ** indicates p≤0.01, and * indicates ρ≤0.05.
図 2は、実施例 2で得られた PC0LCE 2遺伝子発現量の結果を表す図である。 図中、横軸の Αは 1ヶ月間のタバコ煙曝露マウス肺、 Bはそのコン卜ロールマウス 肺、 Cは 3ヶ月間のタバコ煙曝露マウス肺、 Dはそのコントロールマウス肺、 Eは 6ヶ月間のタバコ煙曝露マウス肺、 Fはそのコントロールマウス肺を示す。  FIG. 2 is a diagram showing the results of the expression level of PC0LCE2 gene obtained in Example 2. In the figure, Α on the horizontal axis represents the lungs of a mouse exposed to cigarette smoke for 1 month, B represents the lungs of the control mouse, C represents the lungs of a mouse exposed to cigarette smoke for 3 months, D represents the lungs of the control mouse, and E represents the lungs of 6 months. The tobacco smoke-exposed mouse lung, F indicates the control mouse lung.
図 3は、実施例 3で得られたマウス P C O L C E 2遺伝子発現分布の結果を表す 図である。  FIG. 3 is a diagram showing the results of mouse PCOLCE2 gene expression distribution obtained in Example 3.
図 4は、実施例 4で得られたマウス摘出肺の圧—容量曲線を表す図である。 図中 、 (a) の一睡一はエラスターゼ投与 1日後のマウス群 (n = 6) 、 一口一はコン トロール群 (n = 6) を、 (b) の一園一はエラス夕一ゼ投与 7日後のモデルマウ ス群 (n = 5) 、 一口一はコントロール群 (n=6) を、 (c) の一議一はエラス ターゼ投与 21日後のモデルマウス群 (n = 6) 、 一口一はコントロール群 (n = 6) を示す。 *は p≤0. 05、 * *は p≤0. 01を示す。  FIG. 4 is a diagram showing a pressure-volume curve of a mouse extirpated lung obtained in Example 4. In the figure, (a) one sleep group was a group of mice (n = 6) one day after elastase administration, one bite was a control group (n = 6), and (b) one garden one was elastase administration. Model mouse group (n = 5) after 1 day, control group (n = 6), control group (n = 6), model mouse group (n = 6) 21 days after elastase administration, control Groups (n = 6) are shown. * Indicates p≤0.05, ** indicates p≤0.01.
図 5は、実施例 4で得られたエラス夕一ゼ投与後のマウス摘出肺のコンプライア ンス値の経時的変化を表す図である。 図中、 横軸はエラスターゼ投与後の日数、 縦 軸はコンプライアンス値を示す。 ー麵一はエラスターゼ投与マウス群 (n = 6) 、 一口一はコントロール群 (n = 6) を示す。 *は p≤0. 05、 は p≤0. 0 1を示す。  FIG. 5 is a graph showing the time-dependent change in the compliance value of the mouse extirpated lung after administration of Erasinase obtained in Example 4. In the figure, the horizontal axis shows the number of days after elastase administration, and the vertical axis shows the compliance value.麵 indicates the elastase-administered mouse group (n = 6), and 一 indicates the control group (n = 6). * Indicates p≤0.05 and indicates p≤0.01.
図 6は、実施例 4で得られたェラスターゼ投与後のマウス肺組織での P C〇 L C E 2遺伝子発現量の経時的変化を表す図である。 図中、横軸はエラス夕ーゼ投与後 の日数、縦軸は PCOLCE 2遺伝子発現量を示す。 —騸—はエラス夕ーゼ投与マ ウス群 (n = 6) 、 一口一はコントロール群 (n = 6) を示す。 *は p≤0. 05 を示す。 発明を実施するための最良の形態  FIG. 6 is a graph showing the time course of the expression level of PC PLCE2 gene in mouse lung tissue obtained after administration of elastase obtained in Example 4. In the figure, the horizontal axis shows the number of days after Erasase administration, and the vertical axis shows the amount of PCOLCE2 gene expression. — 騸 — indicates the mouse group (n = 6) to which Erasase was administered, and each bit indicates the control group (n = 6). * Indicates p≤0.05. BEST MODE FOR CARRYING OUT THE INVENTION
本発明で用いられる配列番号: 1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タンパク質(以下、本発明のタンパク質または本 2003/008167 A protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 used in the present invention (hereinafter referred to as the protein of the present invention or 2003/008167
7  7
発明で用いられるタンパク質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 jS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 ァ一ジ、 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、 膝臓、 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 used in the present invention may be referred to as a human warm-blooded animal (eg, guinea pig, rat, mouse, chicken, egret, pig, sheep, hidge, horse, monkey, etc.) cells (eg, hepatocyte, spleen, etc.). Cells, neurons, glial cells, splenic jS cells, bone marrow cells, mesangial cells, Langerhans 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 , Osteocytes, osteoblasts, osteoclasts, mammary cells, hepatocytes or stromal cells, or precursors, stem cells or cancer cells of these cells) or their cells Any tissue that does, such as the brain, parts of the brain (eg, olfactory bulb, amygdala, basal sphere, hippocampus, thalamus, hypothalamus, cerebral cortex, medulla, cerebellum), spinal cord, pituitary, stomach, knee, kidney , Liver, gonad, thyroid, gall bladder, bone marrow, adrenal gland, skin, muscle, lung, gastrointestinal tract (eg, large intestine, small intestine), blood vessels, heart, thymus, spleen, submandibular gland, peripheral blood, prostate, testicle, ovary, It may be a protein derived from placenta, uterus, bone, joint, skeletal muscle, or the like, or may be a synthetic protein.
配列番号: 1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で表されるアミノ酸配列と約 5 0 %以上、 好ましくは約 6 0 %以上、 さらに好ましくは約 7 0 %以上、 より好ましくは約 8 0 %以上、特に好ましくは約 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ミノ酸配列などが挙 げられる。  As the amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1, about 50% or more, preferably about 60% or more, more preferably about 50% or more of the amino acid sequence represented by SEQ ID NO: 1 Amino acid sequences having a homology of 70% or more, more preferably about 80% or more, particularly preferably about 90% or more, and most preferably about 95% or more.
アミノ酸配列の相同性は、相同性計算アルゴリズム NCBI BLAST (Nat ional Center for Biotechnology Informat ion Bas ic Local Al ignment Search Tool) を用レ 以下の条件 (期待値 = 10;ギヤップを許す;マトリクス ==BLOSUM62;フィルタリン グ =0FF) にて計算することができる。  The homology of amino acid sequences was determined using the homology calculation algorithm NCBI BLAST (National Center for Biotechnology Information Basic Local Search Tool). The following conditions (expected value = 10; gap allowed; matrix = BLOSUM62; Filtering = 0FF).
配列番号: 1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活性を有するタンパク質などが好ましい。 配列番 号: 1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、 配列番号: 3で表されるアミノ酸配列を含有するタンパク質 などが挙げられる。 Examples of the protein having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 include, for example, a protein having the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1. A protein having substantially the same activity as the protein containing the amino acid sequence represented by SEQ ID NO: 1 is preferred. SEQ ID NO: protein containing an amino acid sequence substantially identical to the amino acid sequence represented by 1 Examples of the quality include a protein containing the amino acid sequence represented by SEQ ID NO: 3.
実質的に同質の活性としては、 例えば、 プロテアーゼ阻害活性、 プロコラーゲン Substantially equivalent activities include, for example, protease inhibitory activity, procollagen
C末端切断促進活性などが挙げられる。実質的に同質とは、それらの性質が性質的 に (例、 生理学的に、 または薬理学的に) 同質であることを示す。 したがって、 プ 口テアーゼ阻害活性、 プロコラーゲン C末端切断促進活性などが同等 (例、 約 0 . 0 1〜1 0 0倍、 好ましくは約 0 . 1〜1 0倍、 より好ましくは 0 . 5〜2倍) で あることが好ましいが、 これらの活性の程度、 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。 C-terminal cleavage promoting activity and the like. Substantially identical indicates that the properties are qualitatively (eg, physiologically or pharmacologically) identical. Therefore, the protease inhibitory activity, procollagen C-terminal cleavage promoting activity and the like are equivalent (eg, about 0.01 to 100 times, preferably about 0.1 to 10 times, more preferably 0.5 to 10 times). However, the quantitative factors such as the degree of these activities and the molecular weight of the protein may be different.
プロテア一ゼ阻害活性、 プロコラーゲン C末端切断促進活性の測定は、 自体公知 の方法、 例えば J. Biol. Chem. 275 (2)巻、 1384-90頁、 2000年、 Eur. J. Biochem. 186巻、 U5- 121頁、 1989年などに記載の方法またはそれに準じる方法に従って測定 することができる。  The measurement of proteases inhibitory activity and procollagen C-terminal cleavage promoting activity is performed by a method known per se, for example, J. Biol. Chem. 275 (2), 1384-90, 2000, Eur. J. Biochem. 186 Vol., U5-121, 1989, etc. or a method analogous thereto.
具体的には、 プロテアーゼ阻害活性の測定は、 本発明のタンパク質、 標識された (例、 蛍光ラベル) ペプチド基質およびプロテアーゼを反応させ、 プロテア一ゼ活 性により切断されたべプチド基質量を測定する。本反応は、適当な緩衝液中で行う。 '標識剤が例えば蛍光物質の場合、 ペプチド基質量は蛍光強度を測定すればよい。蛍 光強度の測定は、 蛍光測定装置などを使用する公知の方法に準じて行えばよい。 プロコラーゲン C末端切断促進活性の測定は、 本発明のタンパク質、標識された (例、 放射ラベル) プロコラーゲン、 およびプロコラーゲン C末端領域切断プロテ ァ一ゼ (例、 B M P— 1など) を反応させ、 プロテアーゼ活性により切断されたプ 口コラーゲンの C末端夕ンパク質量を測定する。本反応は、適当な緩衝液中で行う。 標識剤が例えば放射性同位元素の場合、 C末端タンパク質量の測定は、 高速液体ク 口マトグラフィーなどを使用する公知の方法に準じて行えばよい。  Specifically, the protease inhibitory activity is measured by reacting the protein of the present invention, a labeled (eg, fluorescent label) peptide substrate and a protease, and measuring the mass of the peptide group cleaved by protease activity. This reaction is performed in an appropriate buffer. 'When the labeling agent is, for example, a fluorescent substance, the peptide group mass may be measured by measuring the fluorescence intensity. The measurement of the fluorescence intensity may be performed according to a known method using a fluorescence measurement device or the like. The measurement of procollagen C-terminal cleavage promoting activity is carried out by reacting the protein of the present invention, labeled (eg, radiolabeled) procollagen, and procollagen C-terminal region cleavage proteinase (eg, BMP-1). Then, the mass of the C-terminal protein of the open collagen cleaved by the protease activity is measured. This reaction is performed in an appropriate buffer. When the labeling agent is, for example, a radioisotope, the amount of the C-terminal protein may be measured according to a known method using high performance liquid chromatography.
また、 本発明で用いられるタンパク質としては、 例えば、 (1 ) ①配列番号: 1 で表されるアミノ酸配列中の 1または 2個以上(例えば 1〜1 0 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) のアミノ酸が欠失したアミノ酸配列、 ②配列番号: 1で表されるアミノ酸配列 に 1または 2個以上(例えば 1〜1 0 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、 ③配列番号: 1で表されるアミノ酸配列に 1または 2個以上 (例え ば 1〜1 0 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、 ④配列番 号: 1で表されるアミノ酸配列中の 1または 2個以上 (例えば 1〜1 0 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⑤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 )①配列番号: 3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〜 1 0 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) のアミノ酸が欠失したアミノ酸配列、 ②配列番号: 3で 表されるアミノ酸配列に 1または 2個以上 (例えば 1〜1 0 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) のァミノ酸が付加したアミノ酸配列、③配列番号: 3で表されるァミノ酸配列に 1 または 2個以上(例えば 1〜1 0 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ァ ミノ酸配列、 ④配列番号: 3で表されるアミノ酸配列中の 1または 2個以上 (例え ば 1〜1 0 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⑤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 わゆるムテインも含まれる。 Examples of the protein used in the present invention include: (1) 1 or 2 or more (for example, about 1 to 100, preferably 1 to 100) in the amino acid sequence represented by SEQ ID NO: 1; An amino acid sequence in which about 30 amino acids have been deleted, preferably about 1 to 10 amino acids, and more preferably about 1 to 5 amino acids, and 2) the amino acid sequence represented by SEQ ID NO: 1. One or two or more amino acids (for example, about 1 to 100, preferably about 1 to 30, preferably about 1 to 10, and more preferably about 1 to 5) The amino acid sequence represented by SEQ ID NO: 1 or 2 or more (for example, about 1 to 100, preferably 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 inserted. ④ 1 or 2 or more (eg, about 1 to 100) in the amino acid sequence represented by SEQ ID NO: 1 An amino acid sequence in which 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 an amino acid obtained by combining them. So-called mutin, such as a protein containing a sequence, represented by (2) SEQ ID NO: 3 1 or 2 or more in the amino acid sequence (eg, about 1 to 100, preferably about 1 to 30, preferably about 1 to 10, more preferably number (1 to 5)) The amino acid sequence represented by SEQ ID NO: 3; 1 or 2 or more amino acids (eg, about 1 to 100, preferably about 1 to 30, preferably 1 to 1 An amino acid sequence to which about 0, more preferably a number (1 to 5) amino acids have been added; ③ one or more amino acid sequences (for example, 1 to 100) in the amino acid sequence represented by SEQ ID NO: 3 Amino acid sequence into which about, preferably about 1 to 30 amino acids, preferably about 1 to 10 amino acids, and more preferably about 1 to 5 amino acids have been inserted, represented by SEQ ID NO: 3. 1 or 2 or more in the amino acid sequence (for example, about 1 to 100, preferably about 1 to 30, and preferably 1 to So-called muteins such as proteins containing an amino acid sequence in which about 10 amino acids are substituted, and more preferably a number (1 to 5) amino acids, or an amino acid sequence combining them are also included. 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としては、 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.
本明細書におけるタンパク質は、 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。 配列番号: 1で表される アミノ酸配列を含有するタンパク質をはじめとする、本発明で用いられるタンパク 質は、 C末端がカルボキシル基(- C00H)、カルボキシレート (-C00")、アミド (-画 2) またはエステル (- C00R) の何れであってもよい。 In the protein in the present specification, the left end is the N-terminus (amino terminus) and the right end is the C-terminus (capilloxy terminus) according to the convention of peptide labeling. The proteins used in the present invention, including the protein containing the amino acid sequence represented by SEQ ID NO: 1, include carboxyl group (-C00H), carboxylate (-C00 "), amide (- 2 ) Or an ester (-C00R).
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 アルキル基、 例えば、 シクロペンチル、 シク 口へキシルなどの C 3_8シクロアルキル基、 例えば、 フエニル、 一ナフチ>レなどの C 612ァリール基、 例えば、 ベンジル、 フエネチルなどのフエ二ルー アルキル基 もしくは a—ナフチルメチルなどの α—ナフチルー C卜2アルキル基などの C 7一 i 4 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, alkyl groups such as n- butyl, for example, C 3 _ 8 cycloalkyl group such as cyclohexyl cyclopentyl, to sik port, for example, phenyl, one naphth> C, such as les 6 - 12 Ariru group, e.g., benzyl, C 7 one i 4 Ararukiru groups such as α- Nafuchiru C Bok 2 alkyl group such as phenylene Lou alkyl or a- naphthylmethyl such phenethyl, A bivaloyloxymethyl group or the like is used.
本発明で用いられるタンパク質が 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 いるものも本発明で用いられるタンパク質に含まれる。この場合のエステ Jレとして 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 used in the present invention has a lipoxyl group (or carboxylate) other than the C-terminus, a protein in which the carboxyl group is amidated or esterified is also included in the protein used in the present invention. In this case, as the ester J, for example, the above-mentioned C-terminal ester or the like is used.
さらに、 本発明で用いられるタンパク質には、 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、 イミダゾール基、 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 C Wアルカノィル基などの C HJ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。 Further, in the protein used in the present invention, the amino group at the N-terminal amino acid residue (eg, methionine residue) is protected by a protecting group (eg, a CM acyl group such as an alkanol such as a formyl group or an acetyl group). Protected, N-terminal glutamine residue generated by cleavage in vivo, pyroglutamine oxidized, Substituent on the side chain of amino acid in the molecule (for example, -0H, -SH, amino group , Imidazole, indole, guanidino, etc.) are protected with an appropriate protecting group (for example, CHW alkyl such as CW alkanoyl such as formyl or acetyl), or a sugar chain is bonded. Complex proteins such as so-called glycoproteins are also included.
本発明で用いられるタンパク質の具体例としては、 例えば、 配列番号: 1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: 3で表されるアミノ酸配列を 含有する夕ンパク質などがあげられる。  Specific examples of the protein used in the present invention include, for example, a protein containing the amino acid sequence represented by SEQ ID NO: 1, a protein containing the amino acid sequence represented by SEQ ID NO: 3, and the like.
本発明で用いられるタンパク質の部分ペプチドとしては、前記した本発明で用い られるタンパク質の部分ペプチドであって、好ましくは、前記した本発明で用いら れる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 used in the present invention is the partial peptide of the protein used in the present invention described above, and is preferably any peptide having the same properties as the protein used in the present invention described above. It may be.
具体的には、 後述する本発明の抗体を調製する目的には、 配列番号: 1で表され るアミノ酸配列において第 3 8 1〜3 9 1番目のアミノ酸配列を有するペプチド、 配列番号: 3で表されるアミノ酸配列において第 3 8 0〜3 9 0番目のアミノ酸配 列を有するペプチドなどがあげられる。例えば、本発明で用いられるタンパク質の 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 のァミノ酸配列を有するぺプチドなどが用いられる。 Specifically, for the purpose of preparing the antibody of the present invention described below, a peptide having the amino acid sequence of the 381st to 391st amino acids in the amino acid sequence represented by SEQ ID NO: 1; The amino acid sequence at positions 380-390 in the amino acid sequence shown Examples include peptides having a sequence. For example, at least 20 or more, preferably 50 or more, more preferably 70 or more, more preferably 100 or more, most preferably 2 or more of the constituent amino acid sequences of the protein used in the present invention. Peptides having 100 or more amino acid sequences are used.
また、本発明で用いられる部分ペプチドは、 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、 さらに好ましくは数 (1〜5 ) 個) のァミノ 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 アミノ酸が付加し、 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 (1〜5 ) 個) のアミノ酸が挿入され、 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) のアミノ酸が他のアミノ酸で置換されていてもよい。  In the partial peptide used in the present invention, one or more (preferably about 1 to 10, more preferably a number (1 to 5)) amino acids in the amino acid sequence are deleted. Or 1 or 2 or more (preferably, about 1 to 20; more preferably, about 1 to 10; more preferably, about 1 to 5) amino acids are added to the amino acid sequence; Or 1 or 2 or more (preferably, about 1 to 20; more preferably, about 1 to 10; more preferably, about 1 to 5) amino acids are inserted into the amino acid sequence; or In the amino acid sequence, one or more (preferably about 1 to 10, more preferably several, and still more preferably about 1 to 5) amino acids are substituted with another amino acid. Is also good.
また、 本発明で用いられる部分ペプチドは C末端が力ルポキシル基 (- C00H) 、 カルポキシレート (- C00— ) 、 アミド (- C0NH2) またはエステル (- C00R) の何れで あってもよい。 The partial peptide used in the present invention, the C-terminus force Rupokishiru group (- C00H), Karupokishireto (- C00-), amide (- C0NH 2) or ester (- C00R) may be either.
さらに、本発明で用いられる部分ペプチドには、 前記した本発明で用いられる夕 ンパク質と同様に、 C末端以外に力ルポキシル基(またはカルボキシレート) 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 peptides used in the present invention include those having a carbonyl group (or carboxylate) in addition to the C-terminus and the amino acid residues at the N-terminus, similarly to the proteins used in the present invention described above. The amino group of a group (eg, methionine residue) is protected by a protecting group, the glutamine residue generated by cleavage of the N-terminal side in vivo is pyroglutamine-oxidized, Also included are those in which the substituent is protected by an appropriate protecting group, and complex peptides such as so-called glycopeptides to which a sugar chain is bound.
本発明で用いられる部分ペプチドは抗体作成のための抗原としても用いること ができる。  The partial peptide used in the present invention can also be used as an antigen for producing an antibody.
本発明で用いられるタンパク質または部分ペプチドの塩としては、生理学的に許 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。 As the salt of the protein or partial peptide used in the present invention, a salt with a physiologically acceptable acid (eg, an inorganic acid, an organic acid) or a base (eg, an alkali metal salt) is used. Physiologically acceptable acid addition salts are preferred. Such salts include: For example, salts with inorganic acids (eg, hydrochloric, phosphoric, hydrobromic, sulfuric) or organic acids (eg, acetic, formic, propionic, fumaric, maleic, succinic, tartaric, Salts with citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, and benzenesulfonic acid are used.
本発明で用いられるタンパク質もしくはその部分ペプチドまたはその塩は、前述 したヒトゃ温血動物の細胞または組織から自体公知のタンパク質の精製方法によ つ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、後述のペプチド合成法 に準じて製造することもできる。  The protein or its partial peptide or a salt thereof used in the present invention can be produced from the above-mentioned human or warm-blooded animal cell or tissue by a known method for purifying a protein, or a DNA encoding the protein can be produced. It can also be produced by culturing the containing transformant. Also, it can be produced according to the peptide synthesis method described later.
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 extract is subjected to reverse phase chromatography, ion exchange chromatography, or the like. Purification and isolation can be achieved by combining chromatography.
本発明で用いられるタンパク質もしくは部分べプチドまたはその塩、またはその アミド体の合成には、 通常市販のタンパク質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、 クロロメチル樹脂、 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—メチルベンズヒドリルァミン樹脂、 P AM樹脂、 4ーヒドロキシメチ 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榭脂、 4 _ ( 2 ' , 4, ー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— ( 2, , 4, ージメトキシフエ二ルー Fm o cアミノエチル)フェ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、 ひーァミノ基と側鎖官能基を適当に保護したァ ミノ酸を、 目的とするタンパク質の配列通りに、 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夕ンパク質または部分べプチドを切り 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ィド結合 形成反応を実施し、 目的のタンパク質もしくは部分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 protein or partial peptide or its salt or its amide used in the present invention, a commercially available resin for protein synthesis can be usually used. Examples of such resins include chloromethyl resin, hydroxymethyl resin, benzylhydramine resin, aminomethyl resin, 4-benzyloxybenzyl alcohol resin, 4-methylbenzhydrylamine resin, PAM resin, and the like. 4-Hydroxymethylmethylphenylacetamidomethyl resin, polyacrylamide resin, 4_ (2 ', 4, dimethoxyphenylhydroxymethyl) phenoxy resin, 4- (2,, 4, dimethoxyphenylaminoethyl) Phenoxy resins and the like can be mentioned. Using such a resin, an amino acid in which the amino group and the side chain functional group are appropriately protected is condensed on the resin according to the sequence of the target protein according to various known condensation methods. At the end of the reaction, protein or partial peptide is excised from the resin, and at the same time, various protecting groups are removed.In addition, an intramolecular disulfide bond formation reaction is carried out in a highly diluted solution, and the target protein or partial peptide or their peptide Obtain the amide form.
上記した保護アミノ酸の縮合に関しては、タンパク質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, Ν'—ジイソプロピルカルポジイミド、 Ν—ェチル— N' 一 (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, HO 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 HO 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つた後 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 activities that can be used for protein synthesis Activating reagents can be used, and carbodiimides are particularly preferable. Examples of the carbopimides include DCC, N, Ν'-diisopropyl carbopimide, and Ν-ethyl-N '-(3-dimethylaminoprolyl) carbopimide. Activation by these involves adding the protected amino acid directly to the resin along with a racemization inhibitor additive (eg, HOBt, HOOBt), or symmetrical acid anhydrides or HOBT esters or HOOBt. The ester can be added to the resin after activation of the protected amino acid in advance.
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エーテル類、 ァセト 二トリル, プロピオ二トリルなどの二トリル類、 酢酸メチル, 酢酸ェチルなどのェ 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 0 °C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な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ールを用いて未反応アミノ酸をァセチル化す ることによって、 後の反応に影響を与えないようにすることができる。  The solvent used for activating the protected amino acid or condensing with the resin can be appropriately selected from solvents known to be usable for the protein condensation reaction. For example, acid amides such as N, N-dimethylformamide, N, N-dimethylacetamide, N-methylpyrrolidone, halogenated hydrocarbons such as methylene chloride, chloroform, and alcohols such as trifluoroethanol. , Sulfoxides such as dimethyl sulfoxide, ethers such as pyridine, dioxane, and tetrahydrofuran; nitriles such as aceto nitrile and propionitrile; esters such as methyl acetate and ethyl acetate; or an appropriate mixture thereof. Used. The reaction temperature is appropriately selected from the range known to be usable for the protein bond formation reaction, and is usually selected appropriately from the range of about 120 ° C. (: to 50 ° C .. The activated amino acid derivative Is usually used in an excess of 1.5 to 4 times.As a result of a test using the ninhydrin reaction, if the condensation is insufficient, sufficient condensation can be carried out by repeating the condensation reaction without removing the protecting group. When sufficient condensation cannot be obtained even after repeating the reaction, acetylation of the unreacted amino acid with acetic anhydride or acetylimidazole is carried out so that the subsequent reaction is not affected. can do.
原料のァミノ基の保護基としては、 例えば、 Z、 B o c、 t一ペンチルォキシカ ルポニル、イソボルニルォキシカルポニル、 4—メトキシベンジルォキシカルボ二 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、 2一二卜口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などが用いられる。  Examples of the protecting group for the amino group of the starting material include Z, Boc, t-pentyloxycarbonyl, isobornyloxycarbonyl, 4-methoxybenzyloxycarbonyl, C1-1Z, Br-Z, Damantyloxycarbonyl, trifluoroacetyl, phthaloyl, formyl, 2212 phenylsulfenyl, 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 口ピル、ブチル、 t一ブチル、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—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ェ 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—ニトロベン ジルエステル、 4ーメトキシベンジルエステル、 4一クロ口べンジルエステル、 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、 t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 よつて保護することができる。 Carbonyl groups are, for example, alkyl esterified (eg, methyl, ethyl, Linear, branched or cyclic alkylesters such as pill, butyl, t-butyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, 2-adamantyl, etc., aralkyl esterification (for example, benzyl ester) , 4-nitrobenzyl ester, 4-methoxybenzyl ester, 4-chlorobenzyl ester, benzylhydryl esterification), phenacyl esterification, benzyloxycarbonyl hydrazide, t-butoxycarbonyl hydrazide, trityl hydrazide Can be protected.
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(C !_6) 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、 エトキシカルボ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、 テトラヒドロピラニル基、 t一ブチル基などである。 The hydroxyl group of serine can be protected, for example, by esterification or etherification. Examples of groups appropriately used for the esterification include a lower (C! _ 6) Arukanoiru groups such Asechiru group, Aroiru group such Benzoiru group, benzyl O carboxymethyl Cal Poni group, and a group derived from carbonic acid such as ethoxycarbonyl group Is used. Examples of a group suitable for etherification include a benzyl group, a tetrahydropyranyl 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 1、 C 12-B z l、 2—ニトロベンジル、 B r_Z、 t—ブチルなどが用いられる。 The protecting group of the phenolic hydroxyl group of tyrosine, for example, B z 1, C 1 2 -B zl, 2- nitrobenzyl, B R_Z, such as t- butyl are used.
ヒスチジンのイミダゾールの保護基としては、 例えば、 To s、 4—メトキシー 2, 3, 6—卜リメチルベンゼンスルホニル、 DNP、 ベンジルォキシメチル、 B um、 Bo c、 Tr t、 Fmo cなどが用いられる。  Examples of the protecting group for histidine imidazole include Tos, 4-methoxy-2,3,6-trimethylbenzenesulfonyl, DNP, benzyloxymethyl, Bum, Boc, Trt, and Fmoc. Can be
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、 ペン夕クロ口フエノール、 2, 4, 5—トリクロ口フエノール、 2, 4—ジニトロフエノール、 シァノメチルアルコー ル、 パラニトロフエノール、 HONB、 N—ヒドロキシスクシミド、 N—ヒドロキ シフ夕ルイミド、 HOB 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。  Examples of the activated carbonyl group of the raw material include, for example, corresponding acid anhydrides, azides, and active esters [alcohols (eg, phenol, 2,4,5-trichlorophenol, 2,4 —Dinitrophenol, cyanomethyl alcohol, paranitrophenol, HONB, N-hydroxysuccinimide, N-hydroxyfurimide, ester with HOB t)]. As the activated amino group of the raw material, for example, a corresponding phosphoric amide is used.
保護基の除去 (脱離) 方法としては、 例えば、 Pd—黒あるいは Pd—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、 トリフルォロメ夕ンスルホン酸、 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、 ジイ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ウムによる 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 オアニソール、 メタクレゾ一ル、 パラクレゾール、 ジメチノレスルフイド、 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一ル保護基として用いられる 2 , 4— ジニト口フエニル基はチォフエノ一ル処理により除去され、 卜リブトフアンのイン 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, 4一ブタンジチオールなどの存在下の酸処理による脱保護 外に、希水酸化ナトリ ゥム溶液、 希アンモニアなどによるアルカリ処理によっても除去される。 Methods for removing (eliminating) protecting groups include, for example, catalytic reduction in the presence of a catalyst such as Pd-black or Pd-carbon in a stream of hydrogen, or hydrogen fluoride anhydride, methanesulfonic acid, trifluoromethane, or the like. Acid treatment with sulfonic acid, trifluoroacetic acid or a mixture thereof, diisopropylethylamine, triethylamine, Base treatment with lysine, piperazine, etc., and reduction with sodium in liquid ammonia are also used. The elimination reaction by the above-mentioned acid treatment is generally performed at a temperature of about 120 ° (: to 40 ° C). In the acid treatment, for example, anisol, phenol, thioanisole, methacrylol, paracresol, It is effective to add a cation trapping agent such as dimethinoresulfide, 1,4-butanedithiol, 1,2-ethanedithiol, etc. Also, 2,4-dinititol used as an imidazole protecting group for histidine The mouth phenyl group is removed by thiophenol treatment, and the formyl group used as an indole protecting group for tributanone is deprotected by acid treatment in the presence of 1,2-ethanedithiol, 1,4-butanedithiol and the like. In addition, it is also removed by alkali treatment with dilute sodium hydroxide solution, dilute ammonia and the like.
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化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.
タンパク質または部分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、 カ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 ミノ基側にペプチド (タンパク質) 鎖を所望の鎖長まで延 した後、 該ペプチド鎖 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または部分べプチドと 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または部分ペプチド とを製造し、 これらのタンパク質またはペプチド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 タンパク質またはべプチドを精製した後、上記方法によりすべての保護基を除去し、 所望の粗タンパク質またはペプチドを得ることができる。この粗タンパク質または ぺプチド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 所望のタンパク質または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 form of a protein or a partial peptide, for example, after amidating and protecting the α-hydroxyl group of the amino acid at the carboxy terminal, a peptide (protein) chain is desired on the amino group side. And a protein or partial peptide from which only the protecting group for the α-amino group at the Ν-terminal of the peptide chain has been removed, and a protein or partial peptide from which only the protecting group for the C-terminal lipoxyl group has been removed. Is produced, and these proteins or peptides are condensed in a mixed solvent as described above. Details of the condensation reaction are the same as described above. After purifying the protected protein or peptide obtained by the condensation, all the protecting groups are removed by the above-mentioned method to obtain a desired crude protein or peptide. The crude protein or peptide is purified by using various known purification means, and the main fraction is freeze-dried to obtain an amide of the desired protein or peptide.
タンパク質またはペプチドのエステル体を得るには、例えば、 カルポキシ末端ァ 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、 タンパク質またはペプチドのアミド体と同様にして、 所望のタンパク質ま たはぺプチドのエステル体を得ることができる。 本発明で用いられる部分ペプチドまたはそれらの塩は、自体公知のペプチドの合 成法に従つて、あるいは本発明で用いられる夕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。 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っても良い。 すなわち、 本発明で用いられる 部分ペプチドを構成し得る部分ペプチドもしくはアミノ酸と残余部分とを縮合さ せ、生成物が保護基を有する場合は保護基を脱離することにより目的のペプチドを 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To obtain an ester of a protein or peptide, for example, after condensing the α-hydroxyl group of the amino acid at the carboxy terminal with a desired alcohol to form an amino acid ester, And an ester form of the protein or peptide. The partial peptide or a salt thereof used in the present invention can be produced according to a peptide synthesis method known per se, or by cleaving the protein used in the present invention with an appropriate peptidase. . 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 objective peptide is produced by condensing a partial peptide or amino acid that can constitute the partial peptide used in the present invention with the remaining portion, and if the product has a protecting group, removing the protecting group to produce the desired peptide. Can be. Known condensation methods and elimination of protecting groups include, for example,
①〜⑤に記載された方法が挙げられる。 The methods described in ① to ⑤ are mentioned.
ΦΜ. Bodanszkyおよび M. A. Ondet t i、ペプチド ·シンセシス (Pept ide Synthes is) , Interscience Publ ishers, New York (1966年)  ΦΜ. Bodanszky and M.A. Ondet ti, Peptide Synthes is, Interscience Publ ishers, New York (1966)
② Schroederおよび Luebke、 ザ ·ペプチド(The Pept ide), Academic Press, New York (1965年)  ② Schroeder and Luebke, The Peptide, Academic Press, New York (1965)
③泉屋信夫他、 ペプチド合成の基礎と実験、 丸善 (株) (1975年)  (3) Nobuo Izumiya et al., Basics and experiments on peptide synthesis, Maruzen Co., Ltd. (1975)
④矢島治明 および榊原俊平、生化学実験講座 1、 タンパク質の化学 IV、 205、 (1977 年)  治 Haruaki Yajima and Shunpei Sakakibara, Laboratory for Biochemical Experiments 1, Protein Chemistry IV, 205, (1977)
⑤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  治 Supervised by Haruaki Yajima, Development of Continuing Drugs, Volume 14, Peptide Synthesis, Hirokawa Shoten
また、 反応後は通常の精製法、 例えば、 溶媒抽出 ·蒸留 ·カラムクロマトグラフ ィ一 ·液体ク口マトグラフィー ·再結晶などを組み合わせて本発明で用いられる部 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部分べプチドが遊離 体である場合は、公知の方法あるいはそれに準じる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、公知の方法あるいはそれに準じる方 法によって遊離体または他の塩に変換することができる。  After the reaction, a general purification method such as solvent extraction, distillation, column chromatography, liquid chromatography, recrystallization, etc. may be combined to purify and isolate the partial peptide used in the present invention. Can be. When the partial peptide obtained by the above method is a free form, it can be converted into an appropriate salt by a known method or a method analogous thereto. It can be converted to a free form or other salts by a method analogous thereto.
本発明で用いられるタ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 た本発明で用いられる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 いかなるものであってもよい。 好ましくは D NAである。 D NAとしては、 ゲノム D NA、 ゲノム D NAライブラリー、 前記した細胞'組織由来の c D NA、 前記し た細胞 ·組織由来の c D NAライブラリー、 合成 D N Aのいずれでもよい。  The polynucleotide encoding the protein used in the present invention may be any polynucleotide containing the above-described nucleotide sequence encoding the protein used in the present invention. Preferably it is DNA. The DNA may be any of a genomic DNA, a genomic DNA library, the above-described cell-tissue-derived cDNA, the above-described cell-tissue-derived cDNA library, and a synthetic DNA.
ライブラリー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 RNAまたは mR NA画分を調製したものを用いて直接 Reverse Transcriptase Polymerase Chain React ion (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。 The vectors used for the library include bacteriophages, plasmids, And phagemid. In addition, it can be directly amplified by reverse transcriptase polymerase chain reaction (hereinafter abbreviated as RT-PCR) using a total RNA or mRNA fraction prepared from the cells and tissues described above.
本発明で用いられるタンパク質をコードする D NAとしては、 例えば、 配列番 号: 2で表される塩基配列を含有する D NA、 または配列番号: 2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する塩基配列を含有し、前 記した配列番号: 1で表されるァ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 D N Aであれば何れのものでもよい。 配列番号: 2で表される塩基配列と八ィストリンジェン卜な条件下でハイプリダ ィズできる D NAとしては、 例えば、 配列番号: 2で表される塩基配列と約 5 0 % 以上、 好ましくは約 6 0 %以上、 さらに好ましくは約 7 0 %以上、 より好ましくは 約 8 0 %以上、 特に好ましくは約 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を含有する D NAなどが用いられる。 例えば、 配列番号: 4で 表される塩基配列を含有する D NAなどが挙げられる。  Examples of the DNA encoding the protein used in the present invention include a DNA containing the nucleotide sequence represented by SEQ ID NO: 2, or a DNA that is highly stringent with the nucleotide sequence represented by SEQ ID NO: 2. Any DNA that contains a base sequence that hybridizes under the conditions and encodes a protein having substantially the same properties as the protein containing the amino acid sequence represented by SEQ ID NO: 1 described above. May be. Examples of the DNA that can be hybridized with the nucleotide sequence represented by SEQ ID NO: 2 under eight stringent conditions include, for example, about 50% or more, preferably 50% or more, of the nucleotide sequence represented by SEQ ID NO: 2. It contains a nucleotide sequence having a homology of about 60% or more, more preferably about 70% or more, more preferably about 80% or more, particularly preferably about 90% or more, and most preferably about 95% or more. DNA or the like is used. For example, a DNA containing the base sequence represented by SEQ ID NO: 4 and the like can be mentioned.
塩基配列の相同性は、 相同性計算アルゴリズム NCBI BLAST (Nat ional Center for Biotechnology Informat ion Basic Local Al ignment Search Tool) を用レ、 以下 の条件 (期待値 = 10;ギャップを許す;フィルタリング =0N;マッチスコア = 1; ミスマッチスコア =-3) にて計算することができる。  The homology of the nucleotide sequences was determined using the homology calculation algorithm NCBI BLAST (National Center for Biotechnology Information Basic Local Search Tool) under the following conditions (expected value = 10; gap allowed; filtering = 0N; match). Score = 1; mismatch score = -3).
ハイプリダイゼーションは、自体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 (Molecular Cloning) 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。 また、 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 according to a method known per se or a method analogous thereto, for example, the method described in Molecular Cloning 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). Can be done. When a commercially available library is used, it can be performed according to the method described in the attached instruction manual. More preferably, the reaction can be performed under high stringent conditions.
ハイストリンジェン卜な条件とは、例えば、ナトリウム濃度が約 1 9〜4 0 mM、 好ましくは約 1 9〜 2 0 mMで、 温度が約 5 0〜 7 0 X:、 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 °Cの場合が 最も好ましい。 Highly stringent 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 X :, preferably about 60 to 70 X. The conditions at 65 ° C are shown. In particular, when the sodium concentration is about 19 mM and the temperature is about 65 ° C, Most preferred.
より具体的には、 配列番号: 1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: 2で表される塩基配列を含有する DNAな どが、配列番号: 3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 NAとしては、配列番号: 4で表される塩基配列を含有する DNAなどが用いられ る。  More specifically, the DNA encoding the protein containing the amino acid sequence represented by SEQ ID NO: 1 includes a DNA containing the base sequence represented by SEQ ID NO: 2, and the like. As the DNA encoding the protein containing the amino acid sequence represented by the above, DNA containing the base sequence represented by SEQ ID NO: 4 or the like is used.
本発明で用いられる部分ペプチドをコードするポリヌクレオチド (例、 DNA) としては、前述した本発明で用いられる部分ペプチド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ってもよい。 また、 ゲノム DNA、 ゲノム DN Aライブラリー、 前記した細胞 ·組織由来の c DNA、 前記した細胞 ·組織由来の cDNAライブラリ一、 合成 DNAのいずれでもよい。  The polynucleotide (eg, DNA) encoding the partial peptide used in the present invention may be any polynucleotide containing the above-described nucleotide sequence encoding the partial peptide used in the present invention. Further, it 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.
本発明で用いられる部分ペプチドをコードする DNAとしては、例えば、配列番 号: 2で表される塩基配列を含有する DNAの一部分を含有する DNA、 または配 列番号: 2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 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。  As the DNA encoding the partial peptide used in the present invention, for example, a DNA containing a part of the DNA containing the base sequence represented by SEQ ID NO: 2, or the base sequence represented by SEQ ID NO: 2 For example, DNA containing a part of DNA encoding a protein having a nucleotide sequence that hybridizes under high stringent conditions and encoding a protein having substantially the same activity as the protein of the present invention may be used.
配列番号: 2で表される塩基配列とハイブリダィズできる DNAは、 前記と同意 義を示す。  DNA that can hybridize with the nucleotide sequence represented by SEQ ID NO: 2 has the same meaning 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トな条件は前記と同 様のものが用いられる。  The same hybridization method and high stringency conditions as described above are used.
本発明で用いられるタンパク質、 部分ペプチド (以下、 これらをコードする DN Aのクローニ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 一を用いて PC R法 よって増幅するか、または適当なベクターに組み込んだ DN 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 ·クロー ニング (Molecular Cloning) 2nd (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。 また、 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 さる。 In the description of the cloning and expression of the DNAs and partial peptides used in the present invention (hereinafter, these may be simply referred to as the proteins of the present invention in the description of the cloning and expression of the DNAs encoding them), the cloning of the DNA completely encoding As a means, DNA is amplified by the PCR method using a synthetic DNA primer having a part of the nucleotide sequence encoding the protein of the present invention, or the DNA incorporated into an appropriate vector is a part of the protein of the present invention. Alternatively, selection can be performed by hybridization with a DNA fragment encoding the entire region or a DNA fragment labeled with a synthetic DNA. The method of hybridization is, for example, molecular claw. This can be performed according to the method described in Molecular Cloning 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). 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、 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、 ODA- LAPCR 法、 Gapped duplex法、 Kunkel法等の自体公知の方法あるいはそれらに準じる方法 に従って行なうことができる。  The DNA base sequence can be converted using PCR, a known kit, for example, Mutan ™ -Super Express Km (Takara Shuzo Co., Ltd.), Mutan ™ -K (Takara Shuzo Co., Ltd.), ODA-LAPCR method, Gapped The method can be carried out according to a method known per se, such as the duplex method and 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、 TG Aまたは TAGを有していてもよ レ^ これらの翻訳開始コドンや翻訳終止コドンは、 適当な合成 DNAアダプタ一を 用いて付加することもできる。  The DNA encoding the cloned protein can be used as it is depending on the purpose, or can be used after digesting with a restriction enzyme or adding a linker, if desired. The DNA may have ATG as a translation initiation codon at its 5 'end and TAA, TGA or TAG as a translation termination codon at its 3' end. Codons and translation termination codons can also be added using a suitable synthetic DNA adapter.
本発明のタンパク質の発現ベクターは、 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ーの下流に連結することにより製造することがで さる。  The expression vector of the protein of the present invention includes, for example, (a) cutting out a target DNA fragment from DNA encoding the protein of the present invention, and (mouth) converting the DNA fragment into a promoter of an appropriate expression vector. It can be manufactured by connecting downstream.
ベクタ一としては、大腸菌由来のプラスミド(例、 PBR 322, pBR 325, pUC 12, pUC13) 、 枯草菌由来のプラスミド (例、 pUB 110, TP 5, pC 194) 、 酵母由来プラスミド (例、 pSHl 9, p SHI 5) 、 λファ ージなどのバクテリオファージ、 レトロウイルス, ワクシニアウィルス, バキュ口 ゥィルスなどの動物ゥィルスなどの他、 A 1 - 11、 pXT 1、 pRc/CMV、 pRcZRS V、 p cDNA I ZN e oなどが用いられる。  Examples of the vector include a plasmid derived from E. coli (eg, PBR322, pBR325, pUC12, pUC13), a plasmid derived from Bacillus subtilis (eg, pUB110, TP5, pC194), a plasmid derived from yeast (eg, pSHl9) , PSHI 5), bacteriophage such as λ phage, animal viruses such as retrovirus, vaccinia virus, baculovirus, etc., A1-11, pXT1, pRc / CMV, pRcZRSV, pcDNA I ZNeo or the like is 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 て適切なプロモーター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プロモ一夕一、 LTRプロモー 夕一、 CMVプロモーター、 HSV-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) プロモータ一、 SRaプロモー ター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 口モー夕—、 プロモ一夕一、 r e cAプロモ一夕一、 APLプロモーター、 1 ppプロモー夕一、 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プロモータ一、 SP02プロモ一夕一、 p e n P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 一夕一、 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ー、 P 10プロモータ一などが好ましい。 The promoter used in the present invention may be any promoter as long as it is appropriate for the host used for gene expression. For example, when animal cells are used as a host, SRa promoter, SV40 promoter, LTR promoter, CMV promoter, HSV-TK promoter and the like can be mentioned. Among them, it is preferable to use CMV (cytomegalovirus) promoter, SRa promoter and the like. When the host is Eshierihia genus bacterium, trp flop port mode evening -, Promo Isseki one, re cA promoter Isseki one, AP L promoter, 1 pp promoter evening one, such as T 7 promoter one is, host Bacillus When the host is a bacterium, SP01 promoter, SP02 promoter, pen P promoter, etc., and when the host is yeast, PHO5 promoter, PGK promoter, GAP promoter, ADH promoter, etc. are preferable. When the host is an insect cell, a polyhedrin promoter, a P10 promoter and the like are preferable.
発現べクタ一には、 以上の他に、 所望によりェンハンサ一、 スプライシングシグ ナル、 ポリ A付加シグナル、 選択マーカー、 SV40複製オリジン (以下、 SV4 0 o 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。 選択マ一カーとしては、 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 〔メソトレキセート (MTX) 耐性〕 、 アンピシリン耐性遺 伝子 (以下、 Amp 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、 G41 8耐性) 等が挙げられる。 特に、 dh f 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ー 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っても選択できる。 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。 宿主がェシエリヒア属菌である場合は、 PhoA ·シグナル配列、 OmpA ·シグナル配列などが、宿主がバチルス属菌である場合は、 ひ一アミラーゼ- 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、 M F a ·シグナル配列、 SUC 2 ·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、 ひ一インターフェロン ·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。 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 SV40 ori), if desired. Can be used. Examples of the selection marker include a dihydrofolate reductase (hereinafter sometimes abbreviated as dh fr) gene (methotrexate (MTX) resistance) and an ampicillin resistance gene (hereinafter abbreviated as Amp) ), the neomycin resistance gene (hereinafter sometimes abbreviated as Ne o r, include G41 8-resistant) and the like. In particular, when the dh fr gene is used as a selection marker using Chinese hamster cells deficient in the dh fr gene, the target gene can be selected using a thymidine-free medium. If necessary, a signal sequence suitable for the host is added to the N-terminal of the protein of the present invention. When the host is a bacterium belonging to the genus Escherichia, a PhoA signal sequence, an OmpA signal sequence, etc., and when the host is a bacterium belonging to the genus Bacillus, a single amylase-signal sequence, subtilisin, a signal sequence, etc., and when the host is a yeast, In some cases, MFa signal sequence, SUC2 signal sequence, etc., and when the host is an animal cell, insulin signal sequence, Hi-interferon signal sequence, antibody molecule, signal sequence, etc. can be used. .
このようにして構築された本発明のタンパク質をコ一ドする 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。  A transformant can be produced using the thus-constructed vector containing DNA encoding the protein of the present invention.
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。 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 (Escherichia coli) Kl 2 - DH1 CProc. Natl. Acad. Sci. USA, 60巻, 160 (1968)〕 , JM1 03 [Nucleic Acids Research, 9巻, 309 (1981)〕, J A 221 (Journal of Molecular Biology, 120巻, 517 (1978)〕 , HB 101 [Journal of Molecular Biology, 41 巻, 459 (1969)〕 , C 600 [Genetics, 39巻, 440 (1954)〕 などが用いられる。 As the host, for example, Escherichia bacteria, Bacillus bacteria, yeast, insect cells, insects, animal cells, and the like are used. Specific examples of the genus Escherichia include, for example, Escherichia coli Kl 2 -DH1 CProc. Natl. Acad. Sci. USA, 60, 160 (1968)], JM103 [Nucleic Acids Research, 9] 309 (1981)), JA 221 (Journal of Molecular Biology, 120, 517 (1978)), HB 101 [Journal of Molecular Biology, 41, 459 (1969)], C600 [Genetics, 39, 440 (1954)].
バチルス属菌としては、 例えば、 バチルス ·サブチルス (Bacillus subtilis) M I 114 [Gene, 24巻, 255 (1983)〕 , 207 - 21 [Journal of Biochemistry, 95 巻, 87 (1984)〕 などが用いられる。  As the Bacillus bacterium, for example, Bacillus subtilis MI114 [Gene, 24, 255 (1983)], 207-21 [Journal of Biochemistry, 95, 87 (1984)] and the like are used.
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—, NA 87 - 11 A, DKD- 5D, 2 OB 一 12、 シゾサッカロマイセス ボンべ (Schizosaccharomyces pombe) NCYC 1913, NCYC 2036, ピキア パストリス (Pichia pastoris) KM 71 などが用いられる。  Examples of yeast include, for example, Saccharomyces cerevisiae AH22, AH22R—, NA87-11A, DKD-5D, 2OB-12, Schizosaccharomyces pombe NCYC 1913, NCYC 2036, Pichia pastoris Pichia pastoris) KM 71 and the like are 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 MG 1細胞、 Tridioplusianiの卵由来の HigliFive™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。 ウィルスが Bm NPV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 (ATCCCRL1711) 、 S f 21細 胞 (以上、 Vaughn, J丄ら、 In Vivo, 13, 213-217, (1977)) などが用いられる。 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、 Nature, 315 巻, 592(1985)〕 。  Insect cells include, for example, when the virus is Ac NPV, a cell line derived from a larva of night roth moth (Spodoptera frugiperda cell; S f cell), MG1 cell derived from the midgut of Trichoplusia ni, and egg derived from Tridioplusiani egg HigliFive ™ cells, cells derived from Mamestra brassicae or cells derived from Estigmena acrea are used. When the virus is Bm NPV, a silkworm-derived cell line (Bombyx mori N cell; BmN cell) or the like is used. As the Sf cells, for example, Sf9 cells (ATCCCRL1711), Sf21 cells (Vaughn, J 丄 et al., In Vivo, 13, 213-217, (1977)) and the like are used. 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 ro, 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遺伝子欠損チャイニーズ ハムスター細胞 CHO (以下、 CHO (dh f r— )細胞と略記) , マウス L細胞, マウス At T— 20, マウスミエローマ細胞, マウス ATDC5細胞, ラット GH 3, 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 Chinese hamster cell CHO (hereinafter abbreviated as CHO (dh fr—) cell. ), Mouse L cells, mouse AtT-20, mouse myeloma cells, mouse ATDC5 cells, rat GH3, human FL cells, etc. are used.
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) Gene, 17巻, 107 (1982)などに記載の方法に従って行なうことができ る。 For transformation of Escherichia sp., For example, Proc. Natl. Acad. Sci. USA, 69 Vol., 2110 (1972) Gene, Vol. 17, 107 (1982), and the like.
バチルス属菌を形質転換するには、 例えば、 Molecular & General Genet ics, 168 巻, 111 (1979)などに記載の方法に従って行なうことができる。  Transformation of Bacillus spp. Can be performed, for example, according to the method described in Molecular & General Genetics, vol. 168, 111 (1979).
酵母を形質転換するには、例えば、 Methods in Enzymology, 194巻, 182-187 (1991)、 Pro Nat l. Acad. Sc i. 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。  Transformation of yeast can be performed, for example, according to the method described in Methods in Enzymology, Vol. 194, 182-187 (1991), Pro Natl. Acad. Sc. USA, Vol. 75, 1929 (1978). Can be.
昆虫細胞または昆虫を形質転換するには、 例えば、 Bio/Technology, 6,  To transform insect cells or insects, see, for example, 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。 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卜 コール. 263-267 (1995) (秀潤社発行) 、 Virology, 52巻, 456 (1973)に記載の方法に 従って行なうことができる。  Transformation of animal cells can be performed, for example, by the method described in Cell Engineering Separate Volume 8 New Cell Engineering Experimental Protocol. 263-267 (1995) (published by Shujunsha), Virology, 52, 456 (1973). So you can do it.
このようにして、タンパク質をコードする 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 the DNA encoding the protein can be obtained.
宿主がェシェリヒア属菌、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、 コ一ンスチープ ·リカー、ペプトン、カゼイン、 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、 塩化マグネシウムなどが挙け5 られる。 また、 酵母エキス、 ビタミン類、 成長促進因子などを添加してもよい。 培 地の p H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 culturing, and a carbon source necessary for growth of the transformant may be used therein. Nitrogen sources, inorganic substances, etc. are included. Carbon sources include, for example, glucose, dextrin, soluble starch, sucrose, etc.Nitrogen sources include, for example, ammonium salts, nitrates, copper steep liquor, peptone, casein, meat extract, soybean meal, potato inorganic or organic substances such as extract, etc. Examples of the inorganic materials are calcium chloride, sodium dihydrogen phosphate, magnesium chloride and levator only be 5. In addition, yeast extract, vitamins, growth promoting factors and the like may be added. The pH of the medium is preferably about 5 to 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 [Mi l ler, Journal of Experiments in Molecular  As a medium for cultivating a bacterium belonging to the genus Escherichia, for example, an M9 medium containing glucose and casamino acid [Miller, Journal of Experiments in Molecular
Genet ics, 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 ]3—インド リルアクリル酸のような薬剤を加えることができる。 宿主がェシェリヒァ属菌の場合、培養は通常約 15〜 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。 Genetics, 431-433, Cold Spring Harbor Laboratory, New York 1972]. If necessary, an agent such as 3] 3-indolylacrylic acid can be added in order to make the promoter work efficiently. When the host is a bacterium belonging to the genus Escherichia, the cultivation is usually performed 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 で約 6〜24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。  When the host is a bacterium belonging to the genus Bacillus, the cultivation is usually performed at about 30 to 40 for about 6 to 24 hours, and if necessary, aeration and stirring can be applied.
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、 バークホー ルダ一 (Burkholder)最小培地 CBostian, 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、 Pro 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 ^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, the culture medium may be, for example, Burkholder's minimal medium (CBostian, KL et al., Proc. Natl. Acad. Sci. USA, 77, 4505 (1980)). And SD medium containing 0.5% casamino acid [Bitter, GA et al., Pro Natl. Acad. Sci. USA, 81, 5330 (1984)]. The pH of the medium is preferably adjusted to about 5-8. Culture is usually performed at about 20 ° C to 35 ^ for about 24 to 72 hours, and aeration and agitation 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、 When culturing a transformant whose host is insect cells or insects,
Grace's Insect Medium (Grace, T. C. C. , 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 pHは約 6. 2〜6. 4に調整するのが好ましい。 培養は通常約 27 °Cで約 3〜 5日間行ない、 必要に応じて通気や撹拌を加える。 Grace's Insect Medium (Grace, T.C.C., Nature), 195, 788 (1962)) to which an additive such as immobilized 10% pepsin serum or the like is appropriately added is used. The pH of the medium is preferably 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 [Science, 122巻, 501 (1952)〕 , DMEM培 地 [Virology, 8 , 396(1959)] , RPM I 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。 p Hは約 6〜 8であるのが好ましい。培養は通常約 30〜401: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 MEM medium containing about 5 to 20% fetal bovine serum [Science, 122, 501 (1952)], DMEM medium [Virology , 8, 396 (1959)], RPM I 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)] Are used. Preferably, the pH is about 6-8. Culture is usually performed at about 30 to 401: about 15 to 60 hours, and aeration and stirring 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壊したのち 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蛋白質変性剤や、 トリトン X— 1 0 0 TM などの界面活性剤力 S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、 それ自体公知の方法で菌体あるいは細胞と上清とを分離し、 上 清を集める。 When extracting the protein of the present invention from the cultured cells or cells, after the culture, the cells or cells are collected by a known method, suspended in an appropriate buffer, and then subjected to ultrasound, A method of obtaining a crude protein extract by centrifugation or filtration after disrupting the cells or cells by zozyme and z or freeze-thawing or the like is appropriately used. The buffer may contain a protein denaturant such as urea or guanidine hydrochloride, or a surfactant S such as Triton X-100 . 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 method known per se, 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夕ンパク質の精 製は、 自体公知の分離 ·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 の公失 Π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、ィォン交換ク口マトグラフィ一な どの荷電の差を利用する方法、ァフィ二ティ一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、逆相高速液体クロマトグラフィーなどの疎水性の差を利用す る方法、 等電点電気泳動法などの等電点の差を利用する方法などが用いられる。 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、 自体公知の方法ある 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 は自体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。  Purification of the protein contained in the culture supernatant or extract thus obtained can be carried out by appropriately combining known separation and purification methods. Methods for separating and purifying these losses include methods using solubility such as salting out and solvent precipitation, dialysis, ultrafiltration, gel filtration, and SDS-polyacrylamide gel electrophoresis. Methods that mainly use differences in molecular weight of DNA, methods that use differences in charges such as ion exchange chromatography, methods that use specific affinity such as affinity chromatography, and reversed-phase high-performance liquid chromatography. For example, a method using a difference in hydrophobicity such as isoelectric point electrophoresis and a method using a difference in isoelectric point are used. When the protein thus obtained is obtained in a free form, it can be converted to a salt by a method known per se or a method analogous thereto. The compound can be converted into a free form or another salt by an analogous method.
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、 アルギニルェンドぺプチダーゼ、 プロテインキナーゼ、 グリコシダーゼなどが 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 ムノアツセィゃウェスタンブロッテイングなどにより測定することができる。 本発明で用いられるタンパク質もしくは部分べプチドまたはその塩に対する抗 体は、本発明で用いられるタンパク質もしくは部分ペ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つてもよ い。 The presence of the protein of the present invention thus produced can be measured by enzymatic immunoassay western blotting using a specific antibody. The antibody against the protein or partial peptide or a salt thereof used in the present invention may be any of a polyclonal antibody and a monoclonal antibody as long as it can recognize the protein or partial peptide or a salt thereof used in the present invention. Yo No.
本発明で用いられる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、 自体公知の抗体または抗血清 の製造法に従って製造することができる。  Antibodies against the protein or partial peptide used in the present invention or a salt thereof (hereinafter sometimes simply referred to as the protein of the present invention in the description of the antibody) are prepared by using the protein of the present invention as an antigen, It can be produced according to a method for producing an antibody or antiserum known per se.
〔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卜や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、 ラッ卜、 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 a diluent at a site capable of producing an antibody upon administration. Complete Freund's adjuvant or incomplete Freund's adjuvant may be administered in order to enhance the antibody-producing ability upon administration. Administration is usually performed once every 2 to 6 weeks, for a total of about 2 to 10 times. Examples of warm-blooded animals to be used include monkeys, rabbits, rabbits, 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、 後記の標識化タンパク質 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、 ケ一ラーとミルスタインの 方法 〔ネイチヤー (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) やセンダイウィル 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, a mouse with an antibody titer was selected from a mouse, and the spleen or lymph node was collected 2 to 5 days after the final immunization. By fusing the antibody-producing cells contained therein 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 a labeled protein described below with the antiserum, and then measuring the activity of a labeling agent bound to the antibody. The fusion operation can be performed according to a known method, for example, the method of Köhler and Milstein [Nature, 256, 495 (1975)]. Examples of the fusion promoter include polyethylene glycol (PEG) and Sendai virus, but PEG is preferably used.
骨髄腫細胞としては、 例えば、 NS— 1、 P 3U1、 SP 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 3U1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 10〜8 0 %程度の濃度で添加され、 2 0〜4 0 °C、好ましくは 3 0〜3 7 °Cで 1〜1 0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。 Examples of the myeloma cells include warm-blooded animal myeloma cells 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 PEG 1000 to PEG 6000) is 10 to 8%. Cell fusion can be carried out efficiently by adding at a concentration of about 0% and incubating at 20 to 40 ° C, preferably 30 to 37 ° C for 1 to 10 minutes.
モノクローナル抗体産生ハイプリドーマのスクリー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) にハイプリドー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を力 Πえ、 固 相に結合したモノクローナル抗体を検出する方法、抗免疫グロプリン抗体またはプ 口ティン 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、放射性物質ゃ酵 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ーナル抗体を検出す る方法などが挙げられる。  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 directly or adsorbed together with a carrier, and then Monoclonal antibody bound to solid phase using anti-immunoglobulin antibody labeled with a radioactive substance or enzyme (anti-mouse immunoglobulin antibody is used if the cells used for cell fusion are mice) or protein A Monoclonal antibody bound to the solid phase by adding the hybridoma culture supernatant to a solid phase to which anti-immunoglobulin antibody or protein A has been adsorbed, adding a protein labeled with a radioactive enzyme, etc. And the like.
モノクローナル抗体の選別は、 自体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、 八 イブ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 ハイブ 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。ハイプリドーマ培饕上清の抗体価は、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 method known per se or a method analogous thereto. Usually, it can be performed in an animal cell culture medium supplemented with HAT (hypoxanthine, aminopterin, thymidine). As a medium for selection and breeding, any medium may be used as long as it can grow viable hybridomas. For example, RPMI 1640 medium containing 1 to 20%, preferably 10 to 20% fetal bovine serum, GIT medium containing 1 to 10% fetal bovine serum (Wako Pure Chemical Industries, Ltd.) Alternatively, a serum-free culture 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 antibody titer of the supernatant of the hybridoma culture medium can be measured in the same manner as in 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 antibodies
モノクローナル抗体の分離精製は、 自体公知の方法、 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。 〔ポリクロ一ナル抗体の作製〕 Monoclonal antibodies can be separated and purified by methods known per se, for example, immunoglobulin separation and purification methods (eg, salting out method, alcohol precipitation method, isoelectric point precipitation method, electrophoresis method, ion exchanger (eg, DEAE)) Absorption / desorption method, ultracentrifugation method, gel filtration method, specific purification method in which only the antibody is collected using an antigen-binding solid phase or an active adsorbent such as protein A or protein G and the bond is dissociated to obtain the antibody.) Can be performed according to (Preparation of polyclonal antibody)
本発明のポリクローナル抗体は、それ自体公知あるいはそれに準じる方法に従つ 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 れとキャリア一蛋白質との複合体をつくり、上記のモノグ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ー蛋白質との複合体 に関し、 キャリアー蛋白質の種類およびキャリアーとハプテンとの混合比は、 キヤ リア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 can be produced according to a method known per se or a method analogous thereto. 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 monoglonal antibody. The antibody can be produced by collecting an antibody-containing substance against the antibody and separating and purifying the antibody. Regarding the complex of an immunizing antigen and a carrier protein used for immunizing a warm-blooded animal, the type of carrier protein and the mixing ratio of the carrier and the hapten are determined by the antibody against the hapten immunized by cross-linking the carrier. Any material may be cross-linked at any ratio as long as it can be efficiently used.For example, serum albumin, thyroglobulin, hemocyanin, etc., in a weight ratio of about 0.1 to 2 per hapten per hapten. A method of coupling at a rate of 0, preferably about 1 to 5 is used.
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、 ダルタルアルデヒドゃカルポジィミド、 マレイミド活性エステル、 チォ一 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れる。  Various condensing agents can be used for force coupling between the hapten and the carrier.For example, an active ester reagent containing a daltaraldehyde タ ル carpoimide, a maleimide active ester, a thiol group, or a dithioviridyl group is used. Can be
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や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 and a diluent at a site where antibody production is possible. Complete Freund's adjuvant or incomplete Freund's adjuvant may be administered in order to enhance the antibody-producing ability upon administration. The administration is usually performed once every about 2 to 6 weeks, for a total of about 3 to 10 times.
ポリクローナル抗体は、 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 blood, ascites, etc., preferably from 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。ポリクローナル抗体の分離精製は、上記のモノクローナル抗 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きる。 本発明で用いられるタンパク質または部分ペプチドをコードするポリヌクレオ チド (例、 D NA (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 ォチドとしては、本発明の DNA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 DN 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 DN Aが好ましい。 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. The polyclonal antibody can be separated and purified according to the same method for separating and purifying immunoglobulin as in the above-described method for separating and purifying a monoclonal antibody. Polynucleotide encoding the protein or partial peptide used in the present invention (eg, DNA (hereinafter, these DNAs may be abbreviated as the DNA of the present invention in the description of antisense polynucleotides)) Antisense polynucleotide having a nucleotide sequence complementary to or substantially complementary to a nucleotide sequence of As a peptide, a nucleotide having a nucleotide sequence complementary to or substantially complementary to the nucleotide sequence of the DNA of the present invention or a part thereof and having an action capable of suppressing the expression of the DNA is provided. Any antisense polynucleotide may be used, but antisense DNA is preferred.
本発明の DN Aに実質的に相補的な塩基配列とは、例えば、本発明の DN Aに相 補的な塩基配列 (すなわち、 本発明の DNAの相補鎖) の全塩基配列あるいは部分 塩基配列と約 70%以上、好ましくは約 80%以上、より好ましくは約 90%以上、 最も好ましくは約 95%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 DNAの相補鎖の全塩基配列うち、 (ィ)翻訳阻害を指向したアンチセン スポリヌクレオチドの場合は、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 70%以上、 好ましくは約 80 %以上、 より好ましくは約 90 %以上、 最も好ましくは約 95 % 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、 (口) RNa s eHによ る R N A分解を指向するァンチセンスポリヌクレオチドの場合は、イントロンを含 む本発明の DN Aの全塩基配列の相補鎖と約 70%以上、好ましくは約 80%以上、 より好ましくは約 90 %以上、最も好ましくは約 95%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それぞれ好適である。  The nucleotide sequence substantially complementary to the DNA of the present invention refers to, for example, the entire nucleotide sequence or 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, among the total nucleotide sequences of the complementary strand of the DNA of the present invention, (a) in the case of an antisense polynucleotide directed to translation inhibition, the nucleotide sequence of the portion encoding the N-terminal portion of the protein of the present invention (for example, An antisense polynucleotide having a homology of about 70% or more, preferably about 80% or more, more preferably about 90% or more, and most preferably about 95% or more with the complementary strand of Mouth) In the case of an antisense polynucleotide which directs RNA degradation by RNaseH, it is about 70% or more, preferably about 80% or more, complementary to the entire nucleotide sequence of the DNA of the present invention including introns. More preferably, antisense polynucleotides each having a homology of about 90% or more, most preferably about 95% or more are suitable.
具体的には、 (1) 配列番号: 2で表される塩基配列を含有する DNA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含有する 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: 2で表される塩基 配列を含有する DN Aの塩基配列に相補な塩基配列、またはその一部分を含有する アンチセンスポリヌクレオチド (より好ましくは、 配列番号: 2で表される塩基配 列を含有する DNAの塩基配列に相補な塩基配列、またはその一部分を含有するァ ンチセンスポリヌクレオチド) 、 (2) 配列番号: 4で表される塩基配列を含有す る DNA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 一部分を含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: 4で表される塩基配列を含有する DN Aの塩基配列に相補な塩基配列、またはその 一部分を含有するアンチセンスポリヌクレオチド (より好ましくは、 配列番号: 4 で表される塩基配列を含有する D NAの塩基配列に相補な塩基配列、またはその一 部分を含有するアンチセンスポリヌクレオチド) などが挙げられる。 Specifically, (1) an antisense polynucleotide containing a base sequence complementary to or substantially complementary to the base sequence of DNA containing the base sequence represented by SEQ ID NO: 2, or a part thereof An antisense polynucleotide containing a nucleotide, preferably, for example, a nucleotide sequence complementary to the nucleotide sequence of DNA containing the nucleotide sequence represented by SEQ ID NO: 2, or a part thereof (more preferably, a nucleotide sequence represented by SEQ ID NO: 2; A base sequence complementary to the base sequence of the DNA containing the base sequence represented, or an antisense polynucleotide containing a part thereof), (2) DNA containing the base sequence represented by SEQ ID NO: 4 An antisense polynucleotide containing a nucleotide sequence complementary to or substantially complementary to the nucleotide sequence of SEQ ID NO: 1, or a portion thereof, preferably, for example, represented by SEQ ID NO: 4 A base sequence complementary to the base sequence of DNA containing the base sequence, or an antisense polynucleotide containing a part thereof (more preferably, SEQ ID NO: 4 And a base sequence complementary to the base sequence of the DNA containing the base sequence represented by the following formula, or an antisense polynucleotide containing a part thereof.
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 NA 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。 また、 各ヌクレオチドの糖(デォキシリポース) は、 2, —O—メチル化などの化学修飾糖構造に置換されていてもよいし、塩基部分(ピリ ミジン、 プリン) も化学修飾を受けたものであってもよく、 配列番号: 2で表され る塩基配列を含有する D N Aにハイブリダィズするものであればいずれのもので 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 residue (phosphate) of each nucleotide constituting the antisense DNA may be, for example, a chemically modified phosphate residue such as phosphorothioate, methylphosphonate, or phosphorodithionate. It may be substituted by a group. In addition, the sugar (doxylipose) of each nucleotide may be substituted with a chemically modified sugar structure such as 2, -O-methylation, and the base (pyrimidine, purine) may also be chemically modified. Any one may be used as long as it hybridizes to the DNA containing the base sequence represented by SEQ ID NO: 2. 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 できるアンチセンスポリヌクレオチドを、 クローン化した、 あるいは決定された夕 ンパク質をコードする D NAの塩基配列情報に基づき設計し、 合成しうる。かかる ヌクレオチド (核酸) は、 本発明のタンパク質遺伝子の R NAとハイブリダィズす ることができ、該 R NA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 N A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 NA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 「对応する」 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ペプチド(蛋白質) との間で「対 応する」 とは、 ヌクレオチド (核酸) の配列またはその相補体から誘導される指令 にあるペプチド (蛋白質) 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、 5 ' 端 6—ベ一スペア ·リピート、 5 ' 端非翻訳領域、 ポリべ プチド翻訳開始コドン、 蛋白質コード領域、 O R F翻訳終止コドン、 3 '端 翻訳 領域、 3 ' 端パリンドローム領域、 および 3 ' 端ヘアピンループは好ましい対象領 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。 According to the present invention, an antisense polynucleotide capable of inhibiting the replication or expression of the protein gene of the present invention is designed based on the nucleotide sequence information of the cloned or determined DNA encoding the protein. And can be synthesized. Such a nucleotide (nucleic acid) can hybridize with the RNA of the protein gene of the present invention, inhibit the synthesis or function of the RNA, or inhibit the interaction with the protein-related RNA of the present invention. Thus, the expression of the protein gene of the present invention can be regulated. Polynucleotides that are complementary to the selected sequence of the protein-related RNA of the present invention, and that can specifically hybridize with the protein-related RNA of the present invention, can be used 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 “responds” means having homology or being complementary to a nucleotide, base sequence or a specific sequence of a nucleic acid including a gene. “Corresponding” between a nucleotide, nucleotide sequence or nucleic acid and a peptide (protein) usually refers to the amino acid of the peptide (protein) specified by the sequence derived from the nucleotide (nucleic acid) sequence or its complement. I have. 5 'of protein gene End hairpin loop, 5 'end 6-base spare repeat, 5' end untranslated region, polypeptide translation initiation codon, protein coding region, ORF translation stop codon, 3 'end translation region, 3' end palindrome region , And the 3 'end hairpin loop may be selected as preferred regions of interest, but any region within the protein gene may be selected.
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は、 対象物とハイプリダイズすることができるポリヌクレオチドとの関係は、 「アンチ センス」 であるということができる。 7ンチセンスポリヌクレオチドは、 2—デォ キシー D—リポースを含有しているポリヌクレオチド、 D—リポースを含有してい るポリヌクレオチド、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、あるいは非ヌクレオチド骨格を有するその他のポリ マー (例えば、 市販の蛋白質核酸および合成配列特異的な核酸ポリマ—) 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 RN A中に見 出されるような塩基のペアリングゃ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 NA、 2本 鎖 R NA、 1本鎖 R NA、 さらに D NA: R NAハイブリッドであること力 Sでき、 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) 、 さらには公 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、キャップ 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 (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 (例えば、 ホスホロチォェ ート、 ホスホロジチォエートなど) を持つもの、 例えば蛋白質 (ヌクレアーゼ、 ヌ クレア—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ーリジ ンなど) や糖 (例えば、 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 (例えば、 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、ァ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。こうした修 飾物は、 メチル化されたプリンぉよびピリミジン、 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、 あるいはェ 一テル、 ァミンなどの官能基に変換されていてよい。 The relationship between the target nucleic acid and a polynucleotide complementary to at least a part of the target region can be said to be "antisense" if the relationship between the target nucleic acid and the polynucleotide that can hybridize with the target is. Antisense polynucleotides are polynucleotides that contain 2-deoxy D-reports, polynucleotides that contain D-reports, and other types of polynucleotides that are N-glycosides of purine or pyrimidine bases. Or other polymers having a non-nucleotide backbone (eg, commercially available protein nucleic acids and synthetic sequence-specific nucleic acid polymers) or other polymers containing special bonds (provided that the polymer is not contained in DNA or RNA). Pairing of bases (contains nucleotides having a configuration that allows base attachment) as described in (1). They can be double-stranded DNA, single-stranded DNA, double-stranded RNA, single-stranded RNA, or even a DNA: RNA hybrid, and can be unmodified polynucleotides (or unmodified polynucleotides). Oligonucleotides) and those with known modifications, e.g., those with labels, capped, methylated, and one or more natural nucleotides that are known in the art. , Substituted with an intramolecular nucleotide, for example, having an uncharged bond (eg, methylphosphonate, phosphotriester, phosphoramidate, olebamate, etc.), charged bond or sulfur-containing Those having a bond (for example, phosphorothioate, phosphorodithioate, etc.), for example, protein (nuclease, nuclease inhibitor, toxin) Antibodies, signal peptides, poly-L-lysine, etc.) or sugars (for example, monosaccharides), etc., which have side-chain groups, or those with interactive compounds (for example, acridine, psoralen, etc.), (Eg, metals, radioactive metals, boron, oxidizable metals, etc.), those containing alkylating agents, those with modified bonds (eg, Anomeric nucleic acids). 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 decorations 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 halogens, aliphatic groups, etc., or ethers, amines, etc. It may have been converted to a functional group.
本発明のアンチセンスポリヌクレオチドは、 R NA、 D NA、 あるいは修飾され た核酸 (R NA、 D NA) である。 修飾された核酸の具体例としては核酸の硫黄誘 導体ゃチォホスフエ一ト誘導体、そしてポリ.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が挙げられるが、それに限定されるものではない。 本発明のアンチセンス核酸は次のような方針で好ましく設計されうる。 すなわち、 細胞内でのアンチセンス核酸をより安定なものにする、アンチセンス核酸の細胞透 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、 そしてもし毒性があるならァンチセンス核酸の毒性をより小さなものにする。  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, but are not limited to, a sulfur derivative of a nucleic acid, a thiophosphoate derivative, and a polynucleoside amide, which is resistant to degradation of an oligonucleoside amide. The antisense nucleic acid of the present invention can be preferably designed according to the following policy. That is, to make the antisense nucleic acid more stable in the cell, to make the antisense nucleic acid more cell-permeable, to have a greater affinity for the target sense strand, and if toxic Minimize the toxicity of antisense nucleic acids.
こうして修飾は当該分野で数多く知られており、 例えば J. Kawakami e t al. , Pharm Tech Japan, Vol. 8, pp. 247, 1992 ; Vol. 8, pp. 395, 1992 ; S. T. Crooke et al. ed. , Ant isense Research and App l icat ions, CRC Press, 1993 などに開 示がある。  Thus, many modifications are known in the art, for example, J. Kawakami et al., Pharm Tech Japan, Vol. 8, pp. 247, 1992; Vol. 8, pp. 395, 1992; ST Crooke et al. Ed. , Ant isense Research and Applicat ions, CRC Press, 1993.
本発明のアンチセンス核酸は、 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、遺伝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力チォン体、 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 (例えば、 ホスホリピド、 コレステロール など) といった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 (例えば、 コレステリルクロ口ホルメート、 コ一ル酸な ど) が挙げられる。 こうしたものは、 核酸の 3 ' 端あるいは 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 '端あるいは 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、テトラエチレンダリコールなどのグリコー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 nucleic acids of the present invention may contain altered or modified sugars, bases, or bonds, and may be provided in special forms such as ribosomes or microspheres, applied by gene therapy, or added. Can be given in a prescribed form. Such additional forms include polythiones 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. (Eg, phospholipids, cholesterol, etc.). Preferred lipids for addition include cholesterol and its derivatives (eg, cholesteryl chromate formate, Etc.). Such a substance can be attached to the 3 'end or 5' end of a nucleic acid, and can be attached via a base, sugar, or intramolecular nucleoside bond. Other groups include capping groups specifically located at the 3 'or 5' end of nucleic acids that prevent degradation by nucleases such as exonuclease and RNase. . Examples of such capping groups include, but are not limited to, hydroxyl-protecting groups known in the art, such as glycols such as polyethylene glycol and tetraethylene dalicol.
アンチセンス核酸の阻害活性は、 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、あるいは本発明のタ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。 該核酸それ自体公知の各種の方法で細胞に適用できる。 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ペプチドを コードするポリヌクレオチド (例、 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 ンスポリヌクレオチド (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) の用途を説明する。  The inhibitory activity of the antisense nucleic acid 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. The nucleic acid can be applied to cells by various methods known per se. Hereinafter, the protein or partial peptide of the present invention or a salt thereof (hereinafter, sometimes abbreviated as the protein of the present invention), a polynucleotide encoding the protein or partial peptide of the present invention (eg, DNA (hereinafter, referred to as the present invention) May be abbreviated as DNA))), 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 abbreviated as the antisense polynucleotide of the present invention).
本発明のタンパク質は、 肺気腫病変を有する肺で発現が低下するので、 疾患マー カーとして利用することが出来る。 すなわち、 肺における早期診断、 症状の重症度 の判定、 疾患進行の予測のためのマーカーとして有用である。 よって、 本発明の夕 ンパク質をコードする遺伝子のァンチセンスポリヌクレオチド、本発明のタンパク 質の活性を阻害する化合物もしくはその塩または本発明のタンパク質に対する抗 体を含有する医薬は、例えば、呼吸器疾患〔例、慢性閉塞性肺疾患(慢性気管支炎、 肺気腫) 、 びまん性汎細気管支炎、 気管支喘息、 嚢胞性線維症、 過敏性肺炎、 肺線 維症など〕 、 鼻炎 (例、 アレルギー性鼻炎、 花粉症、 急性鼻炎、 慢性鼻炎、 肥厚性 鼻炎、萎縮性鼻炎、乾燥性前鼻炎、血管運動性鼻炎、壊疽性鼻炎、副鼻腔炎など)、 免疫疾患 (例、 重症筋無力症、 糸球体腎炎、 多発性硬化症、 シエーダレン症候群、 インスリン抵抗性糖尿病、 慢性関節リウマチ、 全身性エリテマトーデス、 アトピー 性皮膚炎、 白血球異常、 脾機能不全または胸腺異常にともなう免疫不全など) 、 炎 症性腸疾患、アレルギー性結膜炎などの予防'治療剤として使用することができる。 好ましくは、 呼吸器疾患などの予防 ·治療剤、 さらに好ましくは慢性閉塞性肺疾患 などの予防 ·治療剤、 さらに好ましくは肺気腫などの予防 ·治療剤である。 Since the expression of the protein of the present invention is reduced in the lung having emphysema lesions, it can be used as a disease marker. That is, it is useful as a marker for early diagnosis in the lung, judgment of the severity of symptoms, and prediction of disease progression. Therefore, a drug containing the antisense polynucleotide of the gene encoding the protein of the present invention, a compound that inhibits the activity of the protein of the present invention or a salt thereof, or an antibody against the protein of the present invention is, for example, a respiratory tract. Diseases (eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.), rhinitis (eg, allergic rhinitis) , Hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry rhinitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc.), immune diseases (eg, myasthenia gravis, glomeruli) Nephritis, multiple sclerosis, siedalen syndrome, insulin resistant diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopy Dermatitis, leukocyte abnormalities, spleen insufficiency or immunodeficiency associated with thymic abnormalities), inflammatory bowel disease, allergic conjunctivitis, etc. can be used as a preventive agent. Preferably, it is a prophylactic / therapeutic agent for a respiratory disease or the like, more preferably a prophylactic / therapeutic agent for a chronic obstructive pulmonary disease or the like, and further preferably a prophylactic / therapeutic agent for a pulmonary emphysema or the like.
( 1 ) 疾病に対する医薬候補化合物のスクリーニング  (1) Screening of drug candidate compounds for diseases
本発明のタンパク質は肺気腫病変を有する肺で発現が低下するので、本発明の夕 ンパク質の活性を調節 (好ましくは促進) する化合物またはその塩は、 例えば、 呼 吸器疾患 〔例、 慢性閉塞性肺疾患 (慢性気管支炎、 肺気腫) 、 びまん性汎細気管支 炎、 気管支喘息、 嚢胞性線維症、 過敏性肺炎、 肺線維症など〕 、 鼻炎 (例、 アレル ギー性鼻炎、 花粉症、 急性鼻炎、 慢性鼻炎、 肥厚性鼻炎、 萎縮性鼻炎、 乾燥性前鼻 炎、血管運動性鼻炎、壊疽性鼻炎、副鼻腔炎など)、免疫疾患(例、重症筋無力症、 糸球体腎炎、 多発性硬化症、 シエーダレン症候群、 インスリン抵抗性糖尿病、 慢性 関節リウマチ、 全身性エリテマトーデス、 アト'ピー性皮膚炎、 白血球異常、 脾機能 不全または胸腺異常にともなう免疫不全など) 、 炎症性腸疾患、 アレルギー性結膜 炎などの予防 ·治療剤として使用することができる。 好ましくは、 呼吸器疾患など の予防 ·治療剤、 さらに好ましくは慢性閉塞性肺疾患などの予防 ·治療剤、 さらに 好ましくは肺気腫などの予防 ·治療剤である。  Since the expression of the protein of the present invention is reduced in lungs having emphysema lesions, the compound or its salt that regulates (preferably enhances) the activity of the protein of the present invention may be used, for example, for respiratory diseases [eg, chronic obstructive Lung disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.), rhinitis (eg, allergic rhinitis, pollinosis, acute rhinitis, Chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry rhinosinusitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc., immune diseases (eg, myasthenia gravis, glomerulonephritis, multiple sclerosis) , Siedaren syndrome, insulin resistance diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, immunodeficiency associated with splenic dysfunction or thymic abnormalities), inflammation It can be used as a prophylactic and therapeutic agent for inflammatory bowel disease, allergic conjunctivitis, etc. Preferably, it is a prophylactic / therapeutic agent for a respiratory disease or the like, more preferably a prophylactic / therapeutic agent for a chronic obstructive pulmonary disease or the like, and further preferably a prophylactic / therapeutic agent for an emphysema or the like.
したがって、 本発明のタンパク質は、 本発明のタンパク質の活性を調節 (好まし くは促進)す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。  Therefore, the protein of the present invention is useful as a reagent for screening a compound or a salt thereof that regulates (preferably promotes) the activity of the protein of the present invention.
すなわち、 本発明は、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夕 ンパク質の活性 (例えば、 プロテア一ゼ阻害活性、 プロコラーゲン C末端切断促進 活性など) を調節 (促進または阻害) する化合物またはその塩のスクリーニング方 法を提供する。  That is, the present invention regulates (promotes or inhibits) the activity of the protein of the present invention (eg, a protease inhibitory activity, a procollagen C-terminal cleavage promoting activity, etc.) characterized by using the protein of the present invention. And a method for screening a compound or a salt thereof.
具体的には、 (i) 本発明のタンパク質のプロテアーゼ阻害活性またはプロコラ 一ゲン C末端切断促進活性と、 (i i)本発明のタンパク質と試験化合物の混合物の プロテア一ゼ阻害活性またはプロコラーゲン c末端切断促進活性との比較をする ことを特徴する本発明のタンパク質の活性を調節(促進または阻害)する化合物ま たはその塩のスクリーニング方法が用いられる。 Specifically, (i) the protease inhibitory activity of the protein of the present invention or the C-terminal cleavage promoting activity of procollagen; and (ii) the protease inhibitory activity or procollagen c-terminal of the mixture of the protein of the present invention and a test compound. Compounds that regulate (promote or inhibit) the activity of the protein of the present invention, which are characterized by comparison with the cleavage promoting activity. Alternatively, a screening method for a salt thereof is used.
上記スクリーニング方法においては、 例えば (i) と (i i) の場合において、 プ 口テア一ゼ阻害活性またはプロコラーゲン C末端切断促進活性を自体公知の方法、 例えば、 J. Biol. Chera. 275 (2)巻、 1384- 90頁、 2000年、 Eur. J. Biochem. 186 巻、 115- 121頁、 1989年などに記載の方法またはそれに準じる方法に従って測定し、 比較する。  In the above screening method, for example, in the cases of (i) and (ii), the protease inhibitory activity or procollagen C-terminal cleavage promoting activity can be determined by a method known per se, for example, J. Biol. Chera. 275 (2 ), Pp. 1384-90, 2000, Eur. J. Biochem. 186, pp. 115-121, 1989, or the like, or a method analogous thereto, and compared.
具体的には、 (i)本発明のタンパク質、 標識された (例、 蛍光ラベル) ペプチド 基質およびプロテアーゼを反応させた場合と、 (i i)試験化合物の存在下、 本発明の タンパク質、 標識された (例、 蛍光ラベル) ペプチド基質およびプロテア一ゼを反 応させた場合の、プロテアーゼ活性により切断されたべプチ 'ド基質量をそれぞれ測 定し、 本発明のタンパク質の活性を調節 (促進または阻害) する化合物またはその 塩をスクリーニングする。 本反応は、 適当な緩衝液中で行う。 切断されたペプチド 基質量は公知の方法によって測定する。標識剤が例えば蛍光物質の場合、 ペプチド 基質量は蛍光強度を測定すればよい。 蛍光強度の測定は、 蛍光測定装置などを使用 する公知の方法に準じて行えばよい。  Specifically, (i) the case where the protein of the present invention is reacted with a labeled (eg, fluorescent label) peptide substrate and a protease; and (ii) the case where the protein of the present invention is labeled in the presence of a test compound. (Example: fluorescent label) When the peptide substrate and the protease are reacted, the mass of the peptide group cleaved by the protease activity is measured, respectively, to regulate the activity of the protein of the present invention (promoting or inhibiting) To screen for compounds or salts thereof. This reaction is performed in an appropriate buffer. The mass of the cleaved peptide group is measured by a known method. When the labeling agent is, for example, a fluorescent substance, the fluorescence intensity may be measured for the peptide group mass. The measurement of the fluorescence intensity may be performed according to a known method using a fluorescence measurement device or the like.
標識されたペプチド基質およびプロテア一ゼは、試験化合物と混合した後に、本 発明のタンパク質と反応させてもよく、 また、本発明のタンパク質に標識されたぺ プチド基質およびプロテアーゼを接触させた後、 試験化合物を添加してもよい。 具体的には、 (i)本発明のタンパク質、 標識された (例、 放射ラベル) プロコラ 一ゲン、およびプロコラーゲン C末端領域切断プロテアーゼ(例、 B M P— 1など) を反応させた場合と、 (i i)試験化合物の存在下、 本発明のタンパク質、 標識された (例、 放射ラベル) プロコラーゲン、 およびプロコラーゲン C末端領域切断プロテ ァ一ゼ (例、 BM P— 1など) を反応させた場合の、 プロテアーゼ活性により切断 されたプロコラーゲンの C末端夕ンパク質量をそれぞれ測定し、本発明の夕ンパク 質の活性を調節(促進または阻害)する化合物またはその塩をスクリーニングする。 本反応は、適当な緩衝液中で行う。切断されたプロコラーゲンの C末端タンパク質 量は公知の方法によって測定する。標識剤が例えば放射性同位元素の場合、 C末端 タンパク質量の測定は、高速液体クロマトグラフィーなどを使用する公知の方法に 準じて行えばよい。 The labeled peptide substrate and protease may be reacted with the protein of the present invention after being mixed with the test compound, or after contacting the peptide substrate and the protease with the protein of the present invention, Test compounds may be added. Specifically, (i) a reaction of the protein of the present invention with a labeled (eg, radiolabeled) procollagen and a procollagen C-terminal region cleaving protease (eg, BMP-1 etc.) ii) Reaction of the protein of the present invention, labeled (eg, radiolabeled) procollagen, and procollagen C-terminal region cleavage proteinase (eg, BMP-1) in the presence of a test compound Then, the mass of the C-terminal protein of procollagen cleaved by the protease activity is measured, and a compound or a salt thereof that regulates (promotes or inhibits) the activity of the protein of the present invention is screened. This reaction is performed in an appropriate buffer. The amount of the c-terminal protein of the cleaved procollagen is measured by a known method. When the labeling agent is, for example, a radioisotope, the amount of the C-terminal protein can be measured by a known method using high performance liquid chromatography or the like. It may be performed according to.
標識されたプロコラーゲンおよびプロコラーゲン C末端領域切断プロテアーゼ は、 試験化合物と混合した後に、 本発明のタンパク質と反応させてもよく、 また、 本発明のタンパク質に標識されたプロコラーゲンおよびプロコラーゲン c末端領 域切断プロテアーゼを接触させた後、 試験化合物を添加してもよい。  The labeled procollagen and procollagen C-terminal region cleaving protease may be mixed with a test compound and then reacted with the protein of the present invention. After contacting the domain-cleaving protease, the test compound may be added.
上記の本発明のタンパク質は、本発明のタンパク質を産生する能力を有する細胞 を培養することによって製造されたものなどが用いられる。 さらには、 前記細胞の 培養液、 その上清、 細胞破砕物などを用いてもよい。  As the above-mentioned protein of the present invention, those produced by culturing cells having the ability to produce the protein of the present invention and the like are used. Furthermore, a culture solution of the cells, a supernatant thereof, a cell crushed product, or the like may be used.
本発明のタンパク質を産生する能力を有する細胞としては、例えば、前述した本 発明のタンパク質をコードする D NAを含有するべクタ一で形質転換された宿主 (形質転換体) が用いられる。宿主としては、例えば、 C O S 7細胞、 C HO細胞、 H E K 2 9 3細胞などの動物細胞が好ましく用いられる。 該スクリーニングには、 例えば、 前述の方法で培養することによって、本発明のタンパク質を細胞外に分泌 させる、 または細胞内に発現させる形質転換体が好ましく用いられる。本発明の夕 ンパク質を発現し得る細胞の培養方法は、前記した本発明の形質変換体の培養法と 同様である。  As a cell having the ability to produce the protein of the present invention, for example, a host (transformant) transformed with a vector containing the above-described DNA encoding the protein of the present invention is used. As a host, for example, animal cells such as COS 7 cells, CHO cells, and HEK293 cells are preferably used. For the screening, for example, a transformant which secretes the protein of the present invention extracellularly or expresses it intracellularly by culturing by the above-mentioned method is preferably used. The method for culturing cells capable of expressing the protein of the present invention is the same as the above-described method for culturing the transformant of the present invention.
試験化合物としては、 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、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。 例えば、上記 (i i) の場合におけるプロテアーゼ阻害活性またはプロコラーゲン C末端切断促進活性が上記 (i) の場合に比べて、約 2 0 %以上、好ましくは 3 0 % 以上、 より好ましくは約 5 0 %以上上昇させる試験化合物を、 本発明のタンパク質 の活性を促進する化合物として、 上記 (i i) の場合におけるプロテアーゼ阻害活性 またはプロコラーゲン C末端切断促進活性が上記 (i) の場合に比べて、 約 2 0 % 以上、 好ましくは 3 0 %以上、 より好ましくは約 5 0 %以上減少させる試験化合物 を本発明のタンパク質の活性を阻害する化合物として選択することができる。 本発明のタンパク質の活性を促進する活性を有する化合物は、本発明のタンパク 質の作用を増強するための安全で低毒性な医薬、 例えば、 呼吸器疾患 〔例、 慢性閉 塞性^ J疾患 (1曼性気管支炎、 肺気腫) 、 びまん性汎細気管支炎、 気管支喘息、 嚢胞 性線維症、 過敏性肺炎、 肺線維症など〕 、 鼻炎 (例、 アレルギー性鼻炎、 花粉症、 急性鼻炎、 慢性鼻炎、 肥厚性鼻炎、 萎縮性鼻炎、 乾燥性前鼻炎、 血管運動性鼻炎、 壊疽性鼻炎、 副鼻腔炎など) 、 免疫疾患 (例、 重症筋無力症、 糸球体腎炎、 多発性 硬化症、 シェ一グレン症候群、 インスリン抵抗性糖尿病、 慢性関節リウマチ、 全身 性エリテマトーデス、 アトピー性皮膚炎、 白血球異常、 脾機能不全または胸腺異常 にともなう免疫不全など) 、 炎症性腸疾患、 アレルギー性結膜炎などの予防,治療 剤として有用である。 Test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and the like. For example, the protease inhibitory activity or procollagen C-terminal cleavage promoting activity in the case of the above (ii) is about 20% or more, preferably 30% or more, more preferably about 50% or more, as compared with the case of the above (i). The test compound which increases the activity of the protein of the present invention by at least 10% is considered to be a compound that promotes the activity of the protein of the present invention, in which the protease inhibitory activity or procollagen C-terminal cleavage promoting activity in the above (ii) is about A test compound that reduces by 20% or more, preferably 30% or more, more preferably about 50% or more can be selected as a compound that inhibits the activity of the protein of the present invention. The compound having the activity of promoting the activity of the protein of the present invention may be a safe and low-toxic drug for enhancing the action of the protein of the present invention, for example, respiratory diseases [eg, chronic obstructive ^ J disease ( 1 man bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cyst Dysfibrosis, irritable pneumonia, pulmonary fibrosis, etc.), rhinitis (eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrene Rhinitis, sinusitis, etc., immune diseases (eg, myasthenia gravis, glomerulonephritis, multiple sclerosis, Scheigren's syndrome, insulin resistance diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis It is useful as a prophylactic and therapeutic agent for inflammatory bowel disease, allergic conjunctivitis, etc.)
本発明のタンパク質の活性を阻害する活性を有する化合物は、本発明のタンパク 質の生理活性を抑制するための安全で低毒性な医薬、 例えば、 呼吸器疾患 (例、 気 管支喘息、 嚢胞性線維症、 肺線維症、 特発性間質性肺炎など) 、 肝疾患 〔例、 慢性 活動性肝炎、 肝硬変、 肝線維化など〕 、 動脈硬化、 腎硬化症、 骨髄線維症、 糖尿病 性血管障害などの予防 ·治療剤として有用である。  The compound having the activity of inhibiting the activity of the protein of the present invention is a safe and low-toxic drug for suppressing the physiological activity of the protein of the present invention, for example, respiratory diseases (eg, bronchial asthma, cystic Fibrosis, pulmonary fibrosis, idiopathic interstitial pneumonia, etc.), liver disease (eg, chronic active hepatitis, cirrhosis, hepatic fibrosis, etc.), arteriosclerosis, renal sclerosis, myelofibrosis, diabetic vascular disorder, etc. It is useful as a preventive and therapeutic agent.
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、 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、 細胞抽出液、 植物抽出液、 動物組織抽出液、 血漿などから 選ばれた化合物である。該化合物の塩としては、 前記した本発明のペプチドの塩と 同様のものが用いられる。  Compounds or salts thereof obtained using the screening method or the screening kit of the present invention include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts It is a compound selected from liquids, plasma, etc. As the salt of the compound, those similar to the aforementioned salts of the peptide of the present invention are used.
さらに、本発明のタンパク質をコードする遺伝子も、 肺気腫病変を有する肺組織 で発現が低下するので、 本発明のタンパク質をコードする遺伝子の発現を調節(好 ましくは促進) する化合物またはその塩は、 例えば、 例えば、 呼吸器疾患 〔例、 慢 性閉塞性肺疾患 (慢性気管支炎、 肺気腫) 、 びまん性汎細気管支炎、 気管支喘息、 嚢胞性線維症、 過敏性肺炎、 肺線維症など〕 、 鼻炎 (例、 アレルギー性鼻炎、 花粉 症、 急性鼻炎、 慢性鼻炎、 肥厚性鼻炎、 萎縮性鼻炎、 乾燥性前鼻炎、 血管運動性鼻 炎、 壊疽性鼻炎、 副鼻腔炎など) 、 免疫疾患 (例、 重症筋無力症、 糸球体腎炎、 多 発性硬化症、 シエーダレン症候群、 インスリン抵抗性糖尿病、 慢性関節リウマチ、 全身性エリテマトーデス、 アトピー性皮膚炎、 白血球異常、 脾機能不全または胸腺 異常にともなう免疫不全など) 、 炎症性腸疾患、 アレルギー性結膜炎などの予防 - 治療剤として使用することができる。好ましくは、呼吸器疾患などの予防 ·治療剤、 さらに好ましくは慢性閉塞性肺疾患などの予防 ·治療剤、 さらに好ましくは肺気腫 などの予防 ·治療剤である。 Furthermore, since the expression of the gene encoding the protein of the present invention also decreases in lung tissue having emphysema lesions, a compound or a salt thereof that regulates (preferably promotes) the expression of the gene encoding the protein of the present invention may be used. For example, for example, respiratory diseases (eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.), Rhinitis (eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrenous rhinitis, sinusitis, etc.), immunological diseases (example) , Myasthenia gravis, glomerulonephritis, multiple sclerosis, Siedalen syndrome, insulin resistance diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, white Spheres abnormalities, such as spleen dysfunction or thymus abnormally accompanying immunodeficiency), inflammatory bowel disease, preventing such allergic conjunctivitis - can be used as a therapeutic agent. Preferably, a prophylactic or therapeutic agent for respiratory diseases, etc., More preferably, it is a prophylactic / therapeutic agent for chronic obstructive pulmonary disease and the like, and further preferably, a prophylactic / therapeutic agent for emphysema and the like.
したがって、 本発明のポリヌクレオチド (例、 D NA) は、 本発明のタンパク質 をコードする遺伝子の発現を調節す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。  Therefore, the polynucleotide (eg, DNA) of the present invention is useful as a reagent for screening a compound or a salt thereof that regulates the expression of a gene encoding the protein of the present invention.
スクリーニング方法としては、 (i i i) 本発明のタンパク質を産生する能力を有 する細胞を培養した場合と、 (iv) 試験化合物の存在下、 本発明で用いられるタン パク質を産生する能力を有する細胞を培養した場合との比較を行うことを特徴と するスクリ一ニング方法が挙げられる。  The screening methods include (iii) culturing cells having the ability to produce the protein of the present invention, and (iv) cells having the ability to produce the protein used in the present invention in the presence of the test compound. A screening method characterized by performing a comparison with the case of culturing is used.
上記方法において、 (i i i) と (iv) の場合における、 前記遺伝子の発現量 (具 体的には、 本発明のタンパク質量または前記タンパク質をコードする mR NA量) を測定して、 比較する。  In the above method, the expression level of the gene (specifically, the amount of the protein of the present invention or the amount of mRNA encoding the protein) in the cases (ii) and (iv) is measured and compared.
試験化合物および本発明のタンパク質を産生する能力を有する細胞としては、上 記と同様のものが挙げられる。  The test compound and cells having the ability to produce the protein of the present invention include those similar to those described above.
タンパク質量の測定は、 公知の方法、 例えば、 本発明のタンパク質を認識する抗 体を用いて、 細胞抽出液中などに存在する前記タンパク質を、 ウェスタン解析、 E L I S A法などの方法またはそれに準じる方法に従い測定することができる。 mR NA量の測定は、 公知の方法、 例えば、 プローブとして配列番号: 2または その一部分を含有する核酸を用いるノーザンハイプリダイゼ一シヨン、あるいはプ ライマーとして配列番号: 2またはその一部分を含有する核酸、 配列番号: 4また はその一部分を含有する核酸を用いるノーザンハイブリダィゼーシヨン、あるいは プライマ一として配列番号: 4またはその一部分を含有する核酸を用いる P C R法 またはそれに準じる方法に従い測定することができる。  The amount of the protein is measured by a known method, for example, using an antibody that recognizes the protein of the present invention, and analyzing the protein present in a cell extract or the like according to a method such as Western analysis, ELISA, or a method analogous thereto. Can be measured. The amount of mRNA can be measured by a known method, for example, Northern hybridization using a nucleic acid containing SEQ ID NO: 2 or a part thereof as a probe, or a nucleic acid containing SEQ ID NO: 2 or a part thereof as a primer Northern hybridization using a nucleic acid containing SEQ ID NO: 4 or a part thereof, or PCR using a nucleic acid containing SEQ ID NO: 4 or a part thereof as a primer or a method analogous thereto can be used. it can.
例えば、 上記 (iv) の場合における遺伝子発現量を、 上記 (i i i) の場合に比べ て、 約 2 0 %以上、 好ましくは 3 0 %以上、 より好ましくは約 5 0 %以上上昇させ る試験化合物を、本発明のタンパク質をコードする遺伝子の発現を促進する化合物 として、 約 2 0 %以上、 好ましくは 3 0 %以上、 より好ましくは約 5 0 %以上阻害 する試験化合物を、本発明のタンパク質をコードする遺伝子の発現を抑制する化合 物として選択することができる。 For example, a test compound that increases the gene expression level in the case of the above (iv) by about 20% or more, preferably 30% or more, more preferably about 50% or more as compared with the case of the above (iii) As a compound that promotes the expression of a gene encoding the protein of the present invention, a test compound that inhibits about 20% or more, preferably 30% or more, more preferably about 50% or more of the protein of the present invention. Compounds that suppress the expression of encoded genes Can be selected as an object.
本発明のスクリーニング用キットは、本発明で用いられるタンパク質もしくは部 分ペプチドまたはその塩、または本発明で用いられるタンパク質もしくは部分ぺプ チドを産生する能力を有する細胞を含有するものである。  The screening kit of the present invention contains the protein or partial peptide used in the present invention or a salt thereof, or a cell capable of producing the protein or partial peptide used in the present invention.
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、 上記した試験化合物、 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、 細胞抽出液、 植物抽出液、 動物組織 抽出液、 血漿などから選ばれた化合物またはその塩であり、 本発明のタンパク質の 活性 (例、 プロテアーゼ阻害活性またはプロコラーゲン C末端切断促進活性など) を調節する化合物またはその塩である。  The compound or a salt thereof obtained by using the screening method or the screening kit of the present invention may be a test compound described above, for example, a peptide, a protein, a non-peptidic compound, a synthetic compound, a fermentation product, a cell extract, or a plant extract. A compound selected from animal tissue extract, plasma or the like, or a salt thereof, which is a compound or a salt thereof that regulates the activity of the protein of the present invention (eg, protease inhibitory activity or procollagen C-terminal cleavage promoting activity). .
該化合物の塩としては、前記した本発明のタンパク質の塩と同様のものが用いら れる。  As the salt of the compound, those similar to the aforementioned salts of the protein of the present invention are used.
本発明のタンパク質の活性を調節 (好ましくは促進) する化合物またはその塩、 および本発明のタンパク質をコードする遺伝子の発現を調節(好ましくは促進)す る化合物またはその塩はそれぞれ、 例えば呼吸器疾患 〔例、 慢性閉塞性肺疾患 (慢 性気管支炎、 肺気腫) 、 びまん性汎細気管支炎、 気管支喘息、 嚢胞性線維症、 過敏 性肺炎、 肺線維症など〕 、 鼻炎 (例、 アレルギー性鼻炎、 花粉症、 急性鼻炎、 慢性 鼻炎、 肥厚性鼻炎、 萎縮性鼻炎、 乾燥性前鼻炎、 血管運動性鼻炎、 壊疽性鼻炎、 副 鼻腔炎など) 、 免疫疾患 (例、 重症筋無力症、 糸球体腎炎、 多発性硬化症、 シエー ダレン症候群、 ィンスリン抵抗性糖尿病、 慢性関節リゥマチ、 全身性エリテマトー デス、 アトピー性皮膚炎、 白血球異常、 脾機能不全または胸腺異常にともなう免疫 不全など) 、 炎症性腸疾患、 アレルギー性結膜炎などの予防,治療剤として使用す ることができる。 好ましくは、 呼吸器疾患などの予防 ·治療剤、 さらに好ましくは 慢性閉塞性肺疾患などの予防 ·治療剤、 さらに好ましくは肺気腫などの予防 ·治療 剤である。  A compound or a salt thereof that regulates (preferably promotes) the activity of the protein of the present invention and a compound or a salt thereof that regulates (preferably promotes) the expression of a gene encoding the protein of the present invention are, for example, respiratory diseases, respectively. [Eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.], rhinitis (eg, allergic rhinitis, Hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry rhinitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc., immune diseases (eg, myasthenia gravis, glomerulonephritis) , Multiple sclerosis, Siedalen syndrome, insulin-resistant diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, spleen dysfunction Or the like thymic abnormally accompanying immunodeficiency), inflammatory bowel disease, preventing such allergic conjunctivitis, it is that you use as therapeutic agents. Preferably, it is a prophylactic / therapeutic agent for a respiratory disease or the like, more preferably a prophylactic / therapeutic agent for a chronic obstructive pulmonary disease or the like, and further preferably a prophylactic / therapeutic agent for a pulmonary emphysema or the like.
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予防 ·治療剤として使用する場合、 常套手段に従って 製剤化することができる。 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 かる組成物は自体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 射剤ま静脈注射剤、 皮下注射剤、 皮内注射剤、 筋肉注射剤、 点滴注射剤、 関節内注 射剤などの剤形を包含する。 かかる注射剤は、 自体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 捕助斉 !!、 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 ングリコール、 ポリエチレングリコール) 、 非イオン界面活性剤 〔例、 ポリソルべ ート 8 0、 H C O— 5 0 (polyoxyethylene (50mol) adduct of hydrogenated cas tor oi 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 つて調製される。 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 prophylactic / therapeutic agent, it can be formulated into a preparation according to a conventional method. For example, compositions for oral administration include solid or liquid dosage forms such as tablets (including sugar-coated tablets and film-coated tablets), pills, granules, powders, capsules (soft capsules and the like). ), Syrups, emulsions, suspensions and the like. Such a composition is produced by a method known per se and contains a carrier, diluent or excipient usually used in the field of pharmaceuticals. For example, lactose, starch, sucrose, magnesium stearate and the like are used as carriers and excipients for tablets. Compositions for parenteral administration include, for example, injections, suppositories, injections, intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections, intravenous injections, intraarticular injections Includes dosage forms such as injections. Such injections are prepared according to a method known per se, for example, by dissolving, suspending or emulsifying the antibody or a salt thereof in a sterile aqueous or oily liquid commonly used for injections. As the aqueous liquid for injection, for example, physiological saline, isotonic solution containing glucose and other auxiliary agents and the like are used, and suitable dissolution and trapping !!, for example, alcohol (eg, ethanol), polyalcohol (Eg, propylene glycol, polyethylene glycol), nonionic surfactants (eg, polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated catalyst)), 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 in combination as a solubilizing agent. The prepared injection is usually filled in an appropriate ampoule. Suppositories to be used for rectal administration are prepared by mixing the antibody or a salt thereof with a conventional suppository base.
上言 3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〜 5 0 0 m g、 とりわけ注射剤では 5〜1 0 O m g、 そ の他の剤形では 1 0〜2 5 O m gの上記化合物が含有されていることが好ましい。 なお前記した各組成物は、上記化合物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。  The oral or parenteral pharmaceutical composition of the above 3 is conveniently prepared in a dosage unit form so as to be compatible with the dosage of the active ingredient. Examples of such dosage unit dosage forms include tablets, pills, capsules, injections (ampoules), suppositories, etc., and usually 5 to 500 mg, especially 5 for injections. 110 O mg, and other dosage forms preferably contain 10-25 O mg of the compound. Each of the above-mentioned compositions may contain another active ingredient as long as the composition does not cause an undesirable interaction with the compound.
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、 ヒ卜または温 血動物 (例えば、 マウス、 ラット、 ゥサギ、 ヒッジ、 ブ夕、 ゥシ、 ゥマ、 トリ、 ネ コ、 ィヌ、 サル、 チンパンジーなど) に対して経口的にまたは非経口的に投与する ことができる。 The preparations obtained in this way are safe and of low toxicity, for example, It may be orally or parenterally administered to blood animals (eg, mice, rats, egrets, higgins, bush, pests, pests, birds, cats, dogs, monkeys, chimpanzees, etc.). it can.
該化合物またはその塩の投与量は、 その作用、 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、 肺気腫の治療の目的で本発明のタンパク質の活 性を調節する化合物またはその塩を経口投与する場合、 一般的に成人(体重 60 k gとして)においては、一日につき該化合物またはその塩を約 0. 1〜100mg、 好ましくは約 1. 0〜5 Omg、 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、該化合物またはその塩の 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、 肺気腫の治療の目的で本発明のタンパク質の 活性を調節する化合物またはその塩を注射剤の形で通常成人 (体重 60 k gとし て) に投与する場合、 一日につき該化合物またはその塩を約 0. 01〜30mg、 好ましくは約 0. 1〜2 Omg、 より好ましくは約 0. l〜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、体重 60 kg当たりに換算し た量を投与することができる。  The dose of the compound or a salt thereof varies depending on its action, target disease, subject to be administered, administration route, and the like.For example, a compound or a salt thereof that regulates the activity of the protein of the present invention for the purpose of treating emphysema When the compound is orally administered, generally, in an adult (with a body weight of 60 kg), the compound or a salt thereof is used in an amount of about 0.1 to 100 mg, preferably about 1.0 to 5 Omg, more preferably about 1 to 10 mg per day. Administer 0-20 mg. When administered parenterally, the single dose of the compound or a salt thereof varies depending on the administration subject, the target disease, and the like.For example, a compound that regulates the activity of the protein of the present invention for the purpose of treating emphysema Or, when the salt or its salt is administered to an adult (with a body weight of 60 kg) usually in the form of an injection, the compound or its salt is administered in an amount of about 0.01 to 30 mg, preferably about 0.1 to 2 Omg per day. It is convenient to administer preferably about 0.1 to 10 mg by intravenous injection. In the case of other animals, the dose can be administered in terms of weight per 60 kg.
(2) 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  (2) Quantification of the protein of the present invention, its partial peptide or its salt
本発明のタンパク質に対する抗体 (以下、 本発明の抗体と略記する場合がある) 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、特にサンドイツチ免疫測定法による定量などに使用すること ができる。  An antibody against the protein of the present invention (hereinafter sometimes abbreviated as the antibody of the present invention) can specifically recognize the protein of the present invention. It can be used for quantification by San Deutsch 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夕ンパク質の定量法、 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
(ii)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 (ii) 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 solution with the antibody of the present invention insoluble on the carrier and another labeled antibody of the present invention simultaneously or continuously, measuring the activity of the labeling agent on the insoluble carrier And a method for quantifying the protein of the present invention in a test solution. In the quantification method (ii), one of the antibodies recognizes the N-terminal of the protein of the present invention. It is desirable that the other antibody be an antibody that 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 ( a b ' ) 2 、 F a b '、 あるいは F a b画分を用いてもよい。 In addition to quantification of the protein of the present invention using a monoclonal antibody against the protein of the present invention (hereinafter sometimes referred to as the monoclonal antibody of the present invention), detection by tissue staining or the like can also be performed. For these purposes, the antibody molecule itself may be used, or 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—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、 ネフロメトリー、 競合法、 ィムノメトリツ 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. Any measurement method may be used as long as the amount of the body is detected by chemical or physical means and calculated from a standard curve prepared using a standard solution containing a known amount of antigen. . For example, nephelometry, a competition method, an immunometric method and a sandwich method are preferably used, but it is particularly preferable to use a 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、 酵素、 蛍光物質、 発光物質などが用いられる。 放射性同位元素としては、 例えば、 As a labeling agent used in a measurement method using a labeling substance, for example, a radioisotope, an enzyme, a fluorescent substance, a luminescent substance and the like are used. As radioisotopes, for example,
1251〕 、 〔1311〕 、 〔¾〕 、 〔14c〕 などが用いられる。 上記酵素としては、 安定で比活 性の大きなものが好ましく、例えば、 β一ガラクトシダーゼ、 β一ダルコシダーゼ、 アルカリフォスファタ一ゼ、パーォキシダーゼ、 リンゴ酸脱水素酵素などが用いら れる。 蛍光物質としては、 例えば、 フルォレスカミン、 フルォレツセンイソチオシ ァネートなどが用いられる。 発光物質としては、 例えば、 ルミノール、 ルミノール 誘導体、 リレシフェリン、 ルシゲニンなどが用いられる。 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。 [125 1], [131 1], [¾] used and [14 c]. As the above-mentioned enzyme, those which are stable and have high specific activity are preferable, and for example, β-galactosidase, β-dalcosidase, alkaline phosphatase, peroxidase, malate dehydrogenase and the like are used. As the fluorescent substance, for example, fluorescamine, fluorescein isothiocyanate or the like is used. As the luminescent substance, for example, luminol, luminol derivative, reluciferin, lucigenin and the like 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、 また通常タン 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。 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。 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. In the sandwich method, the 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 liquid 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. In addition, in the immunoassay by the sandwich method, the antibody used for the solid phase antibody or the labeling antibody is not necessarily 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.
本発明のサンドイッチ法による本発明のタ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. . 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.
本発明のモノク口一ナル抗体をサンドィツ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一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 (B Z F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 Z 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 competitive method, an immunometric method, or a nephrometry. In the competitive method, the antigen in the test solution and the labeled antigen are allowed to react competitively with the antibody, and then the unreacted labeled antigen (F) is separated from the labeled antigen (B) bound to the antibody. Then (BZF separation), the labeling amount of either B or F is measured, and the amount of antigen in the test solution is quantified. In this reaction method, a soluble antibody is used as the antibody, BZF separation is performed using polyethylene glycol, a liquid phase method using a second antibody to the antibody, a solid phase antibody is used as the first antibody, or A solid-phase method using a soluble first antibody and a solid-phased 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 the 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, and then the immobilized antigen is added to bind the unreacted labeled antibody to the solid phase, and then the solid phase and the liquid phase are separated. Next, measure the amount of label in either phase Then, the amount of antigen in the test solution is quantified.
また、 ネフロメトリーでは、 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ーの散乱を利用するレーザ一ネフ口メトリ一などが 好適に用いられる。  In nephelometry, the amount of insoluble sediment resulting from the 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 can be obtained, a laser-Nef orifice utilizing laser scattering 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 measurement 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 and written documents.
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 ENZYMOLOGYj Vol. 70 (Immunochemical Tec ni ues (Part A) ) > 同書 Vol. 73 (Immunochemical Tec ni ues (Part B) )、 同書 Vol. 74 (Immunochemical Techniaues (Part C) )、 同書 Vol. 84 (Immunochemical Techniaues (Part D: Selected Immunoassays) ) , 同書 Vol. 92 (Immunochemical Techniaues (Part E: Monoclonal Ant ibodies and General Immunoassay Methods) ) , 同書 Vol. 121 (Immunochemical Techniaues (Part For example, edited by Hiro Irie "Radio Nono Atsui" (Kodansha, published in Showa 49), edited by Hiro Irie "Radio Imuno Atsue" (Kodansha, published in 1954), edited by Eiji Ishikawa et al. "Immunoassay method" (Medical Shoin, published in 1938), "Enzyme immunoassay" (ed. 2nd edition) (ed. Eiji Ishikawa et al.) (Medical Publishing, published in 1977), "Enzyme immunoassay method, edited by Eiji Ishikawa et al. (3rd edition) (Medical Shoin, published in 1962), "Methods in ENZYMOLOGYj Vol. 70 (Immunochemical Tec niues (Part A))> Ibid. Vol. 73 (Immunochemical Tec niues (Part B)), Ibid.Vol. 74 (Immunochemical Techniaues (Part C)), Ibid.Vol. 84 (Immunochemical Techniaues (Part D: Selected Immunoassays)), Ibid.Vol. 92 (Immunochemical Techniaues (Part E: Monoclonal Ant ibodies and General Immunoassay Methods)), Ibid. Vol. 121 (Immunochemical Techniaues (Part
I :Hybridoma Technology and Monoclonal Ant ibodies) ) (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。 I: Hybridoma Technology and Monoclonal Ant ibodies)) (published by Academic Press).
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。  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 つて、 本発明のタンパク質の濃度の増加または減少が検出された場合、例えば呼吸 器疾患〔例、慢性閉塞性肺疾患(慢性気管支炎、肺気腫)、びまん性汎細気管支炎、 気管支喘息、 嚢胞性線維症、 過敏性肺炎、 肺線維症など〕 、 鼻炎 (例、 アレルギー 性鼻炎、 花粉症、 急性鼻炎、 慢性鼻炎、 肥厚性鼻炎、 萎縮性鼻炎、 乾燥性前鼻炎、 血管運動性鼻炎、 壊疽性鼻炎、 副鼻腔炎など) 、 免疫疾患 (例、 重症筋無力症、 糸 球体腎炎、 多発性硬化症、 シエーダレン症候群、 インスリン抵抗性糖尿病、 慢性関 節リウマチ、 全身性エリテマトーデス、 アトピー性皮膚炎、 白血球異常、 脾機能不 全または胸腺異常にともなう免疫不全など) 、 炎症性腸疾患、 アレルギー性結膜炎 などである、 または将来罹患する可能性が高いと診断することができる。 Furthermore, when an increase or 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, respiratory diseases (eg, chronic obstructive Pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.), rhinitis (eg, allergy Rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc.), immune diseases (eg, myasthenia gravis, Glomerulonephritis, multiple sclerosis, siedalen syndrome, insulin resistance diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, immunodeficiency associated with impaired splenic function or thymus, etc.), inflammatory It can be diagnosed as having bowel disease, allergic conjunctivitis, etc., or is likely to be affected in the future.
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 preparing an antibody column used for purifying the protein of the present invention, detecting the protein of the present invention in each fraction during purification, and analyzing the behavior of the protein of the present invention in test cells, etc. Can be used.
( 3 ) 遺伝子診断薬  (3) Gene diagnostics
本発明の D N A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、 ブタ、 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 たはその部分ペプチドをコードする D NAまたは mR NA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 診断薬として有用である。  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, pigs, pigs, dogs, cats, dogs, monkeys, Abnormalities (gene abnormalities) in the DNA or mRNA encoding the protein of the present invention or a partial peptide thereof in a chimpanzee or the like can be detected. Or, it is useful as a diagnostic agent for genes such as decreased expression, increased DNA or mRNA, or overexpression.
本発明の D N Aを用いる上記の遺伝子診断は、例えば、 自体公知のノーザンハイ プリダイゼーシヨンや P C R— S S C P法(Genomics,第 5巻, 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。  The above-described genetic diagnosis using the DNA of the present invention can be performed, for example, by the well-known Northern high predication or 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)).
例えば、ノーザンハイブリダイゼーシヨンにより発現過多または減少が検出され た場合や P C R— S S C P法により D N Aの突然変異が検出された場合は、例えば 呼吸器疾患 〔例、 慢性閉塞性肺疾患 ('|¾性気管支炎、 肺気腫) 、 びまん性汎細気管 支炎、 気管支喘息、 嚢胞性線維症、 過敏性肺炎、 肺線維症など〕 、 鼻炎 (例、 ァレ ルギー性鼻炎、 花粉症、 急性鼻炎、 慢性鼻炎、 肥厚性鼻炎、 萎縮性鼻炎、 乾燥性前 鼻炎、 血管運動性鼻炎、 壊疽性鼻炎、 副鼻腔炎など) 、 免疫疾患 (例、 重症筋無力 症、 糸球体腎炎、 多発性硬化症、 シエーダレン症候群、 インスリン抵抗性糖尿病 慢性関節リウマチ、 全身性エリテマ卜一デス、 アトピー性皮膚炎、 白血球異常、 脾 機能不全または胸腺異常にともなう免疫不全など) 、 炎症性腸疾患、 アレルギー性 結膜炎などである可能性が高いと診断することができる。 For example, when overexpression or decrease is detected by Northern hybridization, or when DNA mutation is detected by PCR-SSCP method, for example, respiratory diseases [eg, chronic obstructive pulmonary disease ('| ¾ Respiratory bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.), rhinitis (eg, allergic rhinitis, pollinosis, acute rhinitis, chronic) Rhinitis, hypertrophic rhinitis, atrophic rhinitis, before drying Rhinitis, vasomotor rhinitis, gangrenous rhinitis, sinusitis, etc., immune disorders (eg, myasthenia gravis, glomerulonephritis, multiple sclerosis, siedaren syndrome, insulin resistant diabetes, rheumatoid arthritis, systemic lupus erythema It can be diagnosed as having a high probability of being a child with autism, atopic dermatitis, leukocyte abnormalities, immunodeficiency due to spleen dysfunction or thymic abnormalities), inflammatory bowel disease, allergic conjunctivitis, etc.
( 4 ) アンチセンスポリヌクレオチドを含有する医薬  (4) a drug containing an antisense polynucleotide
本発明の D NAに相補的に結合し、該 D NA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夕ン パク質または本発明の D N Aの機能 (例、 プロテアーゼ阻害活性、 プロコラーゲン C末端切断促進活性など)を抑制することができるので、例えば、 Ιί乎吸器疾患(例、 気管支喘息、 嚢胞性線維症、 肺線維症、 特発性間質性肺炎など) 、 肝疾 〔例、 慢 性活動性肝炎、 肝硬変、 肝線維化など〕 、 動脈硬化、 腎硬化症、 骨髄線維症、 糖尿 病性血管障害などの予防 ·治療剤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 has the protein of the present invention or the present invention in vivo. DNA function (eg, protease inhibitory activity, procollagen C-terminal truncation promoting activity, etc.) can be suppressed, so that, for example, genuine respiratory tract disease (eg, bronchial asthma, cystic fibrosis, pulmonary fibrosis, idiopathic) Prophylactic and therapeutic agents for liver disease (eg, chronic active hepatitis, cirrhosis, hepatic fibrosis, etc.), arteriosclerosis, renal sclerosis, myelofibrosis, diabetic vascular disorders, etc. Can be used.
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、 自体公知の方法に従って製剤化し、 投与することができる。  When the antisense polynucleotide is used as the above-mentioned prophylactic / therapeutic agent, it can be formulated and administered according to a method known per se.
また、 例えば、 前記の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ツドウィル 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。 あるいは、 エアロゾル化して吸入剤として気管内に局所投与 することもできる。  Further, for example, after the above-mentioned antisense polynucleotide is inserted alone or into an appropriate vector such as a retrovirus vector, an adenovirus vector, an adenovirus associated virus vector, or the like, a human or mammal can be treated in a conventional manner. It can be given orally or parenterally to animals (eg, rats, puppies, sheep, sheep, bush, 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. Alternatively, they can be aerosolized and administered topically into the trachea as an inhalant.
さらに、体内動態の改良、半減期の長期化、細胞内取り込み効率の改善を目的に、 前記のアンチセンスポリヌクレオチドを単独またはリポゾームなどの担体ととも に製剤 (注射剤) 化し、 静脈、 皮下、 気道、 肺病変部等に投与してもよい。  Furthermore, for the purpose of improving pharmacokinetics, prolonging the half-life, and improving the efficiency of intracellular uptake, the antisense polynucleotide is formulated alone or together with a carrier such as liposome (injection), and is intravenously, subcutaneously, It may be administered to the respiratory tract or lung lesions.
)投与量は、 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、 肺気腫の治療の目的で本発明のアンチセンスポ リヌクレオチドを投与する場合、 一般的に成人 (体重 6 0 k g ) においては、 一日 にっき該アンチセンスポリヌクレオチドを約 0 . 1〜1 0 O m g投与する。 ) Dosage depends on target disease, subject, route of administration For example, when the antisense polynucleotide of the present invention is administered for the treatment of emphysema, generally, in an adult (body weight 60 kg), the antisense polynucleotide is administered daily. About 0.1 to 10 O mg is administered.
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。  Furthermore, the antisense polynucleotide can also be used as a diagnostic oligonucleotide probe for examining the presence of DNA of the present invention in tissues or cells and the state of its expression.
上記アンチセンスポリヌクレオチドと同様に、本発明のタンパク質をコードする R NAの一部を含有する二重鎖 RNA、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なども、 本発明の遺伝子の発現を抑制することができ、 生体内における本発明で用いられるタンパク質または本発明で用いられる D NA の機能を抑制することができるので、 例えば、 呼吸器疾患 (例、 気管支喘息、 嚢胞 性線維症、 肺線維症、 特発性間質性肺炎など) 、 肝疾患 〔例、 慢性活動性肝炎、 肝 硬変、 肝線維化など〕 、 動脈硬化、 腎硬化症、 骨髄線維症、 糖尿病性血管障害など の予防 ·治療剤などとして使用することができる。  Similarly to the above-mentioned antisense polynucleotide, a double-stranded RNA containing a part of the RNA encoding the protein of the present invention, a lipozyme containing a part of the RNA encoding the protein of the present invention, etc. Gene expression, and the function of the protein used in the present invention or the DNA used in the present invention in vivo can be suppressed. For example, respiratory diseases (eg, bronchial asthma, cysts) Cystic fibrosis, pulmonary fibrosis, idiopathic interstitial pneumonia), liver disease (eg, chronic active hepatitis, cirrhosis, hepatic fibrosis, etc.), arteriosclerosis, renal sclerosis, myelofibrosis, diabetic It can be used as a prophylactic and therapeutic agent for vascular disorders.
二重鎖 R NA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 Medicine, 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、 本発明のタンパク質をコードする R NAの一部に公知のリポザ ィムを連結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 RN A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本発明の R N A上の切断部位に近接した部分 (R NA断片) が挙げられる。  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 linking a known lipozyme to a part of the RNA encoding the protein of the present invention. A part of the RNA encoding the protein of the present invention includes a portion (RNA fragment) adjacent to the cleavage site on the RNA of the present invention which can be cleaved by a known lipozyme.
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、 ア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。 ( 5 ) 本発明の抗体を含有する医薬  When the above-described 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 an antisense polynucleotide. (5) a drug containing the antibody of the present invention
本発明の抗体は、 例えば呼吸器疾患 〔例、 慢性閉塞性肺疾患 (慢性気管支炎、 肺 気腫) 、 びまん性汎細気管支炎、 気管支喘息、 嚢胞性線維症、 過敏性肺炎、 肺線維 症など〕 、 鼻炎 (例、 アレルギー性鼻炎、 花粉症、 急性鼻炎、 慢性鼻炎、 肥厚性鼻 炎、 萎縮性鼻炎、 乾燥性前鼻炎、 血管運動性鼻炎、 壊疽性鼻炎、 副鼻腔炎など) 、 免疫疾患 (例、 重症筋無力症、 糸球体腎炎、 多発性硬化症、 シェ一ダレン症候群、 インスリン抵抗性糖尿病、 慢性関節リウマチ、 全身性エリテマトーデス、 アトピー 性皮膚炎、 白血球異常、 脾機能不全または胸腺異常にともなう免疫不全など) 、 炎 症性腸疾患、アレルギー性結膜炎などの予防'治療剤として使用することができる。 あるいは、 呼吸器疾患 (例、 気管支喘息、 嚢胞性線維症、 肺線維症、 特発性間質性 肺炎など) 、 肝疾患 〔例、 慢性活動性肝炎、 肝硬変、 肝線維化など〕 、 動脈硬化、 腎硬化症、 骨髄線維症、 糖尿病性血管障害などの予防 ·治療剤としても使用するこ とができる。 The antibody of the present invention may be used, for example, for respiratory diseases [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis Etc.), rhinitis (eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic nose Inflammation, atrophic rhinitis, dry rhinitis, vasomotor rhinitis, gangrenous rhinitis, sinusitis, etc., immune disorders (eg, myasthenia gravis, glomerulonephritis, multiple sclerosis, Scheddaren syndrome, Insulin-resistant diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, immunodeficiency associated with splenic dysfunction or thymic abnormalities), inflammatory bowel disease, allergic conjunctivitis, etc. Can be used. Or respiratory disease (eg, bronchial asthma, cystic fibrosis, pulmonary fibrosis, idiopathic interstitial pneumonia, etc.), liver disease (eg, chronic active hepatitis, cirrhosis, liver fibrosis, etc.), arteriosclerosis, It can also be used as a prophylactic and therapeutic agent for renal sclerosis, myelofibrosis, and diabetic vascular disorders.
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 as such or as a suitable pharmaceutical composition. The pharmaceutical composition used for the above administration contains the above 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、 フィルムコーティング錠を含む)、丸剤、顆粒剤、散剤、 力プセル剤 (ソフト力プセル剤を含む) 、 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。  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, forcepsels (soft Syrups, emulsions, suspensions and the like. Such a composition is produced by a known method and contains a carrier, diluent or excipient commonly used in the field of pharmaceuticals. For example, lactose, starch, sucrose, magnesium stearate and the like are used as carriers and excipients for tablets.
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、 ァ ルコール (例、 エタノール) 、 ポリアルコール (例、 プロピレングリコール、 ポリ エチレングリコール) 、 非イオン界面活性剤 〔例、 ポリソルべ一ト 8 0、 H C O - 50 (polyoxyethylene (50mol) adduct of hydrogenated castor oil) 〕 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。 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。 なお前記した各組成物は、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。 Compositions for parenteral administration include, for example, injections, suppositories, etc., and injections include intravenous, subcutaneous, intradermal, intramuscular and intravenous injections. Shape. 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. Examples of aqueous liquids for injection include physiological saline, isotonic solutions containing glucose and other adjuvants, and suitable solubilizing agents such as alcohol (eg, ethanol) and polyalcohol ( For example, propylene glycol, polyethylene glycol), nonionic surfactant [for example, polysorbate 80, HCO- 50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)]. 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 in combination as a solubilizer. The prepared injection is usually filled in an appropriate ampoule. Suppositories used for rectal administration are prepared by mixing the above antibody or a salt thereof with a usual base for suppositories. The above-mentioned oral or parenteral pharmaceutical composition is conveniently prepared in a unit dosage form so as to be compatible with the dosage 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, particularly 5 to 100 mg for injection. Other dosage forms preferably contain 10-25 Omg of the antibody. Each of the above-mentioned compositions may contain another active ingredient as long as the composition does not cause an undesirable interaction 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、ラッ卜、 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、 成人の肺気腫の治療 ·予防の ために使用する場合には、 本発明の抗体を 1回量として、 通常 0. 0 1〜20mg /kg体重程度、 好ましくは 0. 1〜1 OmgZkg体重程度、 さらに好ましくは 0. l〜5mg/k g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, porcupine, sheep, pig, etc.). It can be given orally or parenterally (eg, intravenously) to the evening, cats, cats, dogs, monkeys, etc.). The dosage varies depending on the administration subject, target disease, symptoms, administration route, and the like.For example, when used for the treatment or prevention of pulmonary emphysema in adults, the antibody of the present invention is usually administered in a single dose. 0.1 1 to 20 mg / kg body weight, preferably 0.1 to 1 OmgZ kg body weight, more preferably 0.1 to 5 mg / kg body weight, about 1 to 5 times a day, preferably 1 to 3 times a day It is convenient to administer the compound as an injection at a time. 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、 呼吸器疾患 〔例、 慢性閉塞性肺疾患 (慢性気管 支炎、肺気腫)、びまん性汎細気管支炎、気管支喘息、嚢胞性線維症、過敏性肺炎、 肺線維症など〕 、 鼻炎 (例、 アレルギー性鼻炎、 花粉症、 急性鼻炎、 慢性鼻炎、 肥 厚性鼻炎、 萎縮性鼻炎、 乾燥性前鼻炎、 血管運動性鼻炎、 壊疽性鼻炎、 副鼻腔炎な ど) 、 免疫疾患 (例、 重症筋無力症、 糸球体腎炎、 多発性硬化症、 シエーダレン症 候群、 インスリン抵抗性糖尿病、 慢性関節リウマチ、 全身性エリテマトーデス、 ァ トビー性皮膚炎、白血球異常、脾機能不全または胸腺異常にともなう免疫不全など)、 炎症性腸疾患、 アレルギー性結膜炎などの診断薬としても有用である。 Further, the antibody of the present invention may be used, for example, for respiratory diseases [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, lung Fibrosis, etc.), rhinitis (e.g., allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry proctitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc. ), Immune diseases (eg, myasthenia gravis, glomerulonephritis, multiple sclerosis, siedarenosis) Diagnostic group, insulin resistant diabetes mellitus, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, immunodeficiency associated with spleen dysfunction or thymic abnormalities), inflammatory bowel disease, allergic conjunctivitis, etc. It is also useful.
(6)本発明の 「プロコラーゲン C末端切断促進活性を調節する作用を有する化合 物またはその塩を含有してなる慢性閉塞性肺疾患または気管支喘息の予防 ·治療 剤」 について  (6) The “prophylactic / therapeutic agent for chronic obstructive pulmonary disease or bronchial asthma comprising a compound having a function of regulating procollagen C-terminal cleavage promoting activity or a salt thereof” of the present invention
「プロコラーゲン C末端切断促進活性を調節する作用を有する化合物」 は、 プロ コラーゲン C末端切断促進活性を調節する作用を有する化合物であればいかなる ものでもよく、 例えば、 慢性閉塞性肺疾患 (例、 慢性気管支炎、 肺気腫) 、 びまん 性汎細気管支炎、 気管支喘息、 嚢胞性線維症、 過敏性肺炎、 免疫疾患 (例、 重症筋 無力症、 糸球体腎炎、 多発性硬化症、 シエーダレン症候群、 インスリン抵抗性糖尿 病、慢性関節リゥマチ、全身性エリテマトーデス、ァトピー性皮膚炎、白血球異常、 脾機能不全または胸腺異常にともなう免疫不全など) 、 炎症性腸疾患、 アレルギー 性結膜炎などの予防 ·治療剤などとして用いられる。  The “compound having an effect of regulating the activity of promoting procollagen C-terminal cleavage” may be any compound having an activity of regulating the activity of promoting procollagen C-terminal cleavage. For example, chronic obstructive pulmonary disease (eg, Chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, immune disease (eg, myasthenia gravis, glomerulonephritis, multiple sclerosis, siedalen syndrome, insulin resistance) For the prevention and treatment of inflammatory diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, immunodeficiency associated with spleen dysfunction or thymic abnormalities), inflammatory bowel disease, allergic conjunctivitis, etc. Can be
該予防 ·治療剤は、 上記と同様にして製造される。  The prophylactic / therapeutic agent is produced in the same manner as described above.
(7) DNA転移動物  (7) DNA transfer animal
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 DN Aと略記する) またはその変異 DN A (本発明の外来性変異 DNA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 relates to 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 having the formula:
すなわち、 本発明は、  That is, the present invention
(1) 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、 (1) a non-human mammal having the 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第 (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第 (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 DN Aまたはその変異 DN Aを含有し、 哺乳動物において発 現しうる組換えベクターを提供するものである。  (4) It is intended to provide a recombinant vector 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を有する非ヒト哺乳動物(以下、 本 発明の DNA転移動物と略記する) 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に 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 DNAを転移することによって作出することができる。 また、 該 DNA転移方法により、 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 D N Aを転移し、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自体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 DNA転移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-transferred animal of the present invention) include unfertilized eggs, fertilized eggs, germ cells containing spermatozoa and their primordial cells, and the like. In contrast, preferably, embryo development in non-human mammal development During the raw stage (more preferably, at the single cell or fertilized egg cell stage and generally before the 8-cell stage), calcium phosphate, electric pulse, lipofection, coagulation, microinjection, particle gun, DEAE-dextran, etc. It can be produced by transferring the target DNA. Further, by the DNA transfer method, the exogenous DNA of the present invention can be transferred to somatic cells, organs of living organisms, tissue cells, and the like, and used for cell culture, tissue culture, and the like. The DNA-transferred animal of the present invention can also be produced by fusing the above-mentioned germ cells with a cell fusion method known per se.
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、 ハムスター、 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、 C57BLZ6 系統, DBA 2系統など、 交雑系として、 Be CSFi系統, BDF 系統, B 6D 2 系統, BALB/c系統, I CR系統など) またはラット (例えば、 Wi s t a r, SDなど) などが好ましい。  As non-human mammals, for example, porcupines, pigs, higgins, goats, magpies, dogs, cats, guinea pigs, hamsters, mice, rats and the like are used. Above all, rodents with relatively short ontogeny and biological cycle in terms of the creation of disease animal model systems, and easy to breed rodents, especially mice (for example, pure strains such as C57BLZ6 and DBA 2 As the system, Be CSFi system, BDF system, B6D2 system, BALB / c system, ICR system, etc.) or rat (for example, Wistar, SD, etc.) are preferable.
哺乳動物において発現しうる組換えベクターにおける 「哺乳動物」 としては、 上 記の非ヒ卜哺乳動物の他にヒトなどがあげられる。  "Mammals" in a 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 DNAとは、非ヒト哺乳動物が本来有している本発明の DNA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 refers not to the DNA of the present invention originally possessed by a non-human mammal but to the DNA of the present invention once isolated and extracted from a 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, substitution of another base, etc. DNA that has been used is used, and abnormal DNA is also included.
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 DN A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 DN Aなどが用いられる。  The abnormal DNA refers to a DNA that expresses an abnormal protein of the present invention. For example, a DNA that expresses a protein that suppresses the function of a 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を対象動物に転移させるにあたって は、該 D N Aを動物細胞で発現させうるプロモーターの下流に結合した 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 DNAを転 移させる場合、これと相同性が高い本発明の DNA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 DN A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 DNAを結合した DN A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つて本発明の D N Aを高発現す る DNA転移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 that is the same or different from the animal of interest. When the DNA of the present invention is transferred to a target animal, a DNA construct linked to a downstream of a promoter capable of expressing the DNA in animal cells is used. It is generally advantageous to use it as a struct. For example, when the human DNA of the present invention is transferred, DNs derived from various mammals (eg, egrets, dogs, cats, guinea pigs, hamsters, rats, mice, etc.) having the DNA of the present invention having high homology to the human DNA are transferred. Microinjection of a DNA construct (eg, vector 1) with the human DNA of the present invention downstream of various promoters capable of expressing A into a fertilized egg of a target mammal, for example, a mouse fertilized egg. A DNA-transferred mammal that highly expresses the DNA of the present invention can be created.
本発明のタンパク質の発現ベクターとしては、 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ファ一ジなどのバクテリオファージ、 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イ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 or a baculovirus. Animal viruses and the like are used. Among them, a plasmid derived from Escherichia coli, a plasmid derived from Bacillus subtilis or a plasmid derived from yeast are preferably used.
上記の D Ν Α発現調節を行なうプロモーターとしては、例えば、①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、 乳がんウィルス、 ポリオウイルスなど) に由来する DN Aのプロモータ一、 ② 各種哺乳動物 (ヒト、 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) 由来のプロモーター、 例えば、 アルブミン、 インスリン I I、 ゥロプラ キン I I、 エラス夕一ゼ、 エリスロポエチン、 エンドセリン、 筋クレアチンキナー ゼ、 グリァ線維性酸性夕ンパク質、 ダルタチオン S—トランスフエラーゼ、 血小板 由来成長因子 i3、 ケラチン K l, 1 0ぉょび1^ 1 4、 コラーゲン 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サブユニット、 ジストロフィ ン、 酒石酸抵抗性アルカ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ァ デノシン 3リン酸化酵素(Na, K-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、 メタ口プロティナーゼ 1組織インヒビター、 M HCクラス I抗原(H— 2 L) 、 H— r a s、 レニン、 ドーパミン] 3—水酸化酵素、 甲状腺ペルォキシダーゼ (TPO) 、 ペプチド鎖延長因子 la (EF- 1 ) 、 β ァクチン、 ο: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、 Thy_ l、 免疫グロブリン、 H鎖可変部 (VNP) 、 血清アミロイド Pコンポーネント、 ミオグロビン、 トロポニン C、 平滑筋 αァクチ ン、 プレプロエンケフアリン 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、 ヒトペプチド鎖延長因子 1 a; (EF- 1 a) のプロモーター、 ヒ卜およびニヮトリ j8ァクチンプロモーターなどが好適である。 Examples of the promoter which regulates the expression of DΝ are, for example, a promoter of DNA derived from ① virus (eg, Simian virus, cytomegalovirus, Moroni leukemia virus, JC virus, breast cancer virus, poliovirus, etc.). 1, ② Promoters derived from various mammals (humans, egrets, dogs, cats, guinea pigs, hamsters, rats, mice, etc.), such as albumin, insulin II, peropkin II, eras ichie, erythropoietin, endothelin, Muscle creatine kinase, glial fibrillary acidic protein, daltathione S-transferase, platelet-derived growth factor i3, keratin Kl, 10 and 1 ^ 14, collagen type I and II, cyclic AMP Dependent protein kinase / 3 I subunit, dystrophy Protein, tartrate-resistant alkaline phosphatase, atrial natriuretic factor, endothelial receptor oral synthase (generally abbreviated as Tie 2), sodium potassium adenosine 3 kinase (Na, K-ATPase), neurofilament Light chain, meta-oral thionein I and IIA, meta-oral proteinase 1 tissue inhibitor, MHC class I antigen (H-2L), H-ras, renin, dopamine] 3-hydroxylase, thyroid peroxidase (TPO), peptide Chain elongation factor la (EF-1), β Actin, ο: and] 3 myosin heavy chain, myosin light chain 1 and 2, myelin basic protein, thyroglobulin, Thy_l, immunoglobulin, heavy chain variable region (VNP), serum amyloid P component, myoglobin, troponin C For example, promoters such as smooth muscle α-actin, preproenkephalin A, and vasopressin are used. Among them, a cytomegalovirus promoter capable of high expression throughout the whole body, a human peptide chain elongation factor 1a; (EF-1a) promoter, a human and a chicken j8 actin promoter, and the like are preferable.
上記ベクターは、 DNA転移哺乳動物において目的とするメッセンジャー RNA の転写を終結する配列(一般にターミネ夕一と呼ばれる) 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 DN A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ターミネタ一などが用いられ る。  The vector preferably has a sequence that terminates the transcription of the messenger RNA of interest in the DNA-transferred mammal (generally called terminator). For example, DNs derived from viruses and various mammals can be used. The sequence of A can be used, and preferably, SV40 terminator of simian virus or the like is used.
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、 ェンハンサー領域、 真核 DN A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。  In addition, the splicing signal of each DNA, the enhancer region, a part of the intron of eukaryotic DNA, etc., are added 5 'upstream of the promoter region, between the promoter region and the translation region, in order to further express the target exogenous DNA. Alternatively, it can be linked to the 3 'downstream of the translation region depending on the purpose.
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 DNAおよび市販の各種ゲノム DNAライブラ リーよりゲノム DN A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 RN Aより公知の方法により調製された相補 DN Aを原料として 取得することが出来る。 また、 外来性の異常 DN Aは、 上記の細胞または組織より 得られた正常なタンパク質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, and fibroblasts derived from humans or various mammals (eg, rabbit, dog, cat, guinea pig, hamster, rat, mouse, etc.). And all or part of the genomic DNA from commercially available genomic DNA libraries, or the complementary DNA prepared by known methods from liver, kidney, thyroid cells, and fibroblast-derived RNA. Can be done. In addition, an exogenous abnormal DNA can produce a translation region obtained by mutating the translation region of a normal protein obtained from the above cells or tissues by point mutagenesis.
該翻訳領域は転移動物において発現しうる DNA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 D N A工学的手法により作製することができる。  The translation region can be prepared as a DNA construct that can be expressed in a transgenic animal by a conventional DNA engineering technique in which the translation region is ligated downstream of the promoter and, if desired, upstream of the transcription termination site.
受精卵細胞段階における本発明の外来性 DN Aの転移は、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 NA転移後の作出動物の 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 ベて、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来性 D N Aを有する。 The transfer of the exogenous DNA of the present invention at the fertilized egg cell stage is dependent on the germ cell of the target mammal. Vesicles and somatic cells. The presence of the exogenous DNA of the present invention in the germinal cells of the animal after transfer of the DNA indicates that the exogenous DNA of the present invention is retained in all of the germ cells and somatic cells of the offspring of the animal. Means to do. The offspring of such animals that have inherited the foreign DNA of the present invention have the foreign DNA of the present invention in all of their germ cells and somatic cells.
本発明の外来性正常 D NAを転移させ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。  The non-human mammal to which the exogenous normal DNA of the present invention has been transferred is confirmed to stably retain exogenous DNA by mating, and can be subcultured as a DNA-bearing animal in a normal breeding environment. I can do it.
受精卵細胞段階における本発明の外来性 D N Aの転移は、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転移後の作出動 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 することを意味する。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。  Transfer 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 transgenic animal after the DNA transfer indicates that all the offspring of the produced animal will have excessively exogenous DNA of the present invention in all of the germinal and somatic cells. Means having. The progeny of such animals that have inherited the exogenous DNA of the present invention have an excess of the exogenous DNA of the present invention in all of their germinal and somatic cells.
導入 D NA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 N Aを過剰に有するように繁殖継 代することができる。  By obtaining a homozygous animal having the introduced DNA on both homologous chromosomes and mating the male and female animals, all offspring can be bred to have the DNA in excess.
本発明の正常 D NAを有する非ヒト哺乳動物は、本発明の正常 D N Aが高発現さ せられており、内在性の正常 D N Aの機能を促進することにより最終的に本発明の 夕ンパク質の機能亢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 N A転移動物を用いて、 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ことが可能である。  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 eventually promotes the function of the endogenous normal DNA. It may develop hyperfunction and can be used as a model animal for the disease. For example, using the normal DNA-transferred animal of the present invention, elucidation of the pathological mechanism of the protein hyperactivity of the present invention and diseases associated with the protein of the present invention, and examination of treatment methods for these diseases. It is possible.
また、 本発明の外来性正常 D N Aを転移させた哺乳動物は、 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 る予防 ·治療剤、 例えば呼吸器疾患 〔例、 慢性閉塞性肺疾患 (慢性気管支炎、 肺気 腫) 、 びまん性汎細気管支炎、 気管支喘息、 嚢胞性線維症、 過敏性肺炎、 肺線維症 など〕 、 鼻炎(例、 アレルギー性鼻炎、花粉症、急性鼻炎、慢性鼻炎、肥厚性鼻炎、 萎縮性鼻炎、 乾燥性前鼻炎、 血管運動性鼻炎、 壊疽性鼻炎、 副鼻腔炎など) 、 免疫 疾患 (例、 重症筋無力症、 糸球体腎炎、 多発性硬化症、 シエーダレン症候群、 イン スリン抵抗性糖尿病、 慢性関節リウマチ、 全身性エリテマトーデス、 アトピー性皮 膚炎、 白血球異常、 脾機能不全または胸腺異常にともなう免疫不全など) 、 炎症性 腸疾患、 アレルギー性結膜炎などの予防 ·治療剤のスクリーニング試験にも利用可 能である。 Further, since the mammal into which the exogenous normal DNA of the present invention has been transferred has an increased symptom of the released protein of the present invention, it is a prophylactic / therapeutic agent for a disease associated with the protein of the present invention. Respiratory disease (eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.), rhinitis (eg, Allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, Atrophic rhinitis, dry rhinitis, vasomotor rhinitis, gangrenous rhinitis, sinusitis, etc., immune disorders (eg, myasthenia gravis, glomerulonephritis, multiple sclerosis, siedalen syndrome, insulin resistance) Screening tests for preventive and therapeutic agents such as diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, immunodeficiency associated with spleen dysfunction or thymic abnormalities), inflammatory bowel disease, allergic conjunctivitis, etc. 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 に組み込んで原料として用いることができる。プロモーターとの D NA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の転移は、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 N A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 N Aを有するよう に繁殖継代することができる。  On the other hand, the 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. . Further, the desired foreign 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 a usual DNA engineering technique. The transfer 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 produced animal after the transfer of the DNA means that all the offspring of the produced animal have the abnormal DNA of the present invention in all of the germinal 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 germinal and somatic cells. A homozygous animal having the introduced DNA on both homologous chromosomes is obtained, and by crossing the male and female animals, it is possible to breed so that all offspring have the DNA.
本発明の異常 D NAを有する非ヒト哺乳動物は、本発明の異常 D N A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転移動物を用いて、 本発明の夕 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 the endogenous normal DNA by inhibiting the function of the endogenous normal DNA. It may be functionally inactive refractory and can be used as a model animal for the disease. For example, using the abnormal DNA-transferred animal of the present invention, it is possible to elucidate the pathological mechanism of the protein inactive refractory of the protein of the present invention and to consider 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を転移させた哺乳動物は、 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機能不活性型不応 症に対する予防 ·治療剤、 例えば呼吸器疾患 〔例、 慢性閉塞性肺疾患 (慢性気管支 炎、 肺気腫) 、 びまん性汎細気管支炎、 気管支喘息、 嚢胞性線維症、 過敏性肺炎、 肺線維症など〕 、 鼻炎 (例、 アレルギー性鼻炎、 花粉症、 急性鼻炎、 慢性鼻炎、 肥 厚性鼻炎、 萎縮性鼻炎、 乾燥性前鼻炎、 血管運動性鼻炎、 壊疽性鼻炎、 副鼻腔炎な ど) 、 免疫疾患 (例、 重症筋無力症、 糸球体腎炎、 多発性硬化症、 シェ一ダレン症 候群、 インスリン抵抗性糖尿病、 '隱性関節リウマチ、 全身性エリテマト一デス、 ァ 卜ピー性皮膚炎、白血球異常、脾機能不全または胸腺異常にともなう免疫不全など)、 炎症性腸疾患、 アレルギー性結膜炎などの予防 ·治療剤などの予防 ·治療剤のスク 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 is useful for inhibiting the function of a normal protein by the abnormal protein of the present invention (dominant negative effect) in the inactive refractory disease of the protein of the present invention. ) Is a model for elucidating. Further, since the mammal to which the foreign abnormal DNA of the present invention has been transferred has an increased symptom of the released protein of the present invention, the agent for preventing or treating the protein of the present invention or the functionally inactive refractory disease is used. , For example, respiratory diseases (eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.), rhinitis (eg, Allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry anterior rhinitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc., immune diseases (eg, myasthenia gravis) Asthenia, glomerulonephritis, multiple sclerosis, Sheddalen syndrome, insulin resistance diabetes, 'rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, spleen dysfunction or Thymus Always such accompanying immunodeficiency), it can also be utilized in the screening test prophylactic or therapeutic agent such as an agent for the prophylaxis or treatment of such inflammatory bowel diseases, allergic conjunctivitis.
また、上記 2種類の本発明の D NA転移動物のその他の利用可能性として、 ·例え ば、  In addition, other possible uses of the above two types of DNA-transferred animals of the present invention include, for example,
①組織培養のための細胞源としての使用、  ① Use as a cell source for tissue culture,
②本発明の D NA転移動物の組織中の D NAもしくは R NAを直接分析するか、ま たは D N Aにより発現されたぺ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ペプチドとの関連性についての解析、 ③ D N A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、 (2) The protein of the present invention can be specifically analyzed by directly analyzing the DNA or RNA in the tissue of the DNA-transferred animal of the present invention or by analyzing the peptide tissue expressed by DNA. Analysis of the relationship with expressed or activating peptides, ③ culturing cells of DNA-containing tissue by standard tissue culture techniques, and using these to study the function of cells from tissues that are generally difficult to culture,
④上記③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 —ニング、 および 薬 剤 Screening of drugs that enhance cell function by using the cells described in ③ above, and
⑤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。  単 離 Isolation and purification of the mutant protein of the present invention and production of its antibody are considered.
さらに、 本発明の D NA転移動物を用いて、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。  Furthermore, using the DNA-transferred animal of the present invention, it is possible to examine the clinical symptoms of a disease associated with the protein of the present invention, including a functionally inactive refractory disease of the protein of the present invention. More detailed pathological findings in each organ of the protein-related disease model can be obtained, which can contribute to the development of new treatment methods and the research and treatment of secondary diseases caused by the disease.
また、 本発明の D NA転移動物から各臓器を取り出し、 細切後、 ト のタンパク質分解酵素により、 遊離した DN A転移細胞の取得、 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、 本発明の夕ン 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, each organ is removed from the DNA-transferred animal of the present invention, and after minced, By using the proteolytic enzyme of the present invention, it is possible to obtain free DNA transfer cells, culture them, or systematize the cultured cells. Furthermore, it is possible to examine the specificity of the protein-producing cell of the present invention, its relationship with apoptosis, differentiation or proliferation, or its signal transduction mechanism, and its abnormalities. And effective research materials for elucidating its action.
さらに、本発明の DNA転移動物を用いて、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二 ング法を提供することが可能となる。 また、 本発明の DNA転移動物または本 明 の外来性 DNA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 D N A治療法を検討、 開発することが可能である。  Further, in order to develop a therapeutic agent for a disease associated with the protein of the present invention, including a functionally inactive refractory disease of the protein of the present invention, using the DNA-transferred animal of the present invention, It is possible to provide an effective and rapid screening method for the therapeutic agent for the disease by using a method or the like. Further, using the transgenic animal of the present invention or the exogenous DNA expression vector of the present invention, it is possible to examine and develop a method for treating DNA associated with the protein of the present invention.
(8) ノックアウト動物  (8) Knockout animal
本発明は、本発明の DN Aが不活性化された非ヒ卜哺乳動物胚幹細胞および本発 明の DNA発現不全非ヒト哺乳動物を提供する。  The present invention provides a non-human mammalian embryonic stem cell in which the DNA of the present invention has been inactivated, and a non-human mammal deficient in expression of the DNA of the present invention.
すなわち、 本発明は、  That is, the present invention
( 1 ) 本発明の DNAが不活性化された非ヒ卜哺乳動物胚幹細胞、  (1) a non-human mammalian embryonic stem cell in which the DNA of the present invention has been inactivated,
(2) 該 DNAがレポーター遺伝子 (例、 大腸菌由来の i3—ガラクトシダーゼ遺伝 子) を導入することにより不活性化された第 (1) 項記載の胚幹細胞、  (2) The embryonic stem cell according to (1), wherein the DNA is inactivated by introducing a reporter gene (eg, an i3-galactosidase gene derived from Escherichia coli).
(3) ネオマイシン耐性である第 (1) 項記載の胚幹細胞、  (3) The embryonic stem cell according to (1), which is neomycin-resistant,
(4) 非ヒト哺乳動物がゲッ歯動物である第 (1) 項記載の胚幹細胞、  (4) The embryonic stem cell according to (1), wherein the non-human mammal is a rodent,
(5) ゲッ歯動物がマウスである第 (4) 項記載の胚幹細胞、  (5) The embryonic stem cell according to (4), wherein the rodent is a mouse,
(6) 本発明の DNAが不活性化された該 DNA発現不全非ヒ卜哺乳動物、  (6) a DNA-deficient non-human mammal in which the DNA of the present invention has been inactivated,
(7) 該 DNAがレポ一夕一遺伝子(例、 大腸菌由来の j8—ガラクトシダーゼ遺伝 子) を導入することにより不活性化され、該レポ一夕一遺伝子が本発明の DN Aに 対するプロモーターの制御下で発現しうる第 (6) 項記載の非ヒト哺乳動物、  (7) The DNA is inactivated by introducing a repo overnight gene (eg, a j8-galactosidase gene derived from Escherichia coli), and the repo overnight gene controls the promoter for the DNA of the present invention. The non-human mammal according to item (6), which can be expressed under
(8) 非ヒト哺乳動物がゲッ歯動物である第 (6) 項記載の非ヒト哺乳動物、 (8) the non-human mammal according to (6), wherein the non-human mammal is a rodent;
(9) ゲッ歯動物がマウスである第 (8) 項記載の非ヒト哺乳動物、 および (9) the non-human mammal according to (8), wherein the rodent is a mouse; and
(10) 第 (7) 項記載の動物に、 試験化合物を投与し、 レポーター遺伝子の発現 を検出することを特徴とする本発明の DNAに対するプロモーター活性を促進ま たは阻害する化合物またはその塩のスクリーニング方法を提供する。 (10) The test compound is administered to the animal described in (7), and the reporter gene expression And a method for screening a compound or a salt thereof that promotes or inhibits the promoter activity of the DNA of the present invention, characterized by detecting the following.
本発明の DNAが不活性化された非ヒト哺乳動物胚幹細胞とは、該非ヒト哺乳動 物が有する本発明の DN Aに人為的に変異を加えることにより、 DN Aの発現能を 抑制するか、もしくは該 DNA力 Sコードしている本発明のタンパク質の活性を実質 的に喪失させることにより、 DN Aが実質的に本発明のタンパク質の発現能を有さ ない (以下、 本発明のノックアウト DNAと称することがある) 非ヒト哺乳動物の 胚幹細胞 (以下、 ES細胞と略記する) をいう。  A non-human mammalian embryonic stem cell in which the DNA of the present invention has been inactivated is an artificially mutated DNA of the present invention possessed by the non-human mammal, which suppresses the expression ability of the DNA. Alternatively, by substantially losing the activity of the S-encoding protein of the present invention, the DNA has substantially no ability to express the protein of the present invention (hereinafter referred to as the knockout DNA of the present invention). This may be referred to as a non-human mammalian embryonic stem cell (hereinafter abbreviated as ES cell).
非ヒト哺乳動物としては、 前記と同様のものが用いられる。  As the non-human mammal, the same one as described above is used.
本発明の DNAに人為的に変異を加える方法としては、 例えば、遺伝子工学的手 法により該 D N A配列の一部又は全部の削除、他 D N Aを揷入または置換させるこ とによって行なうことができる。 これらの変異により、 例えば、 コドンの読み取り 枠をずらしたり、プロモーターあるいはェキソンの機能を破壊することにより本発 明のノックアウト DN Aを作製すればよい。  The DNA of the present invention can be artificially mutated by, for example, deleting part or all of the DNA sequence, or inserting or substituting another DNA sequence by a genetic engineering method. With these mutations, the knockout DNA of the present invention may be prepared, for example, by shifting the codon reading frame or disrupting the function of the promoter or exon.
本発明の DNAが不活性化された非ヒト哺乳動物胚幹細胞(以下、 本発明の DN A不活性化 E S細胞または本発明のノックァゥト E S細胞と略記する)の具体例と しては、 例えば、 目的とする非ヒト哺乳動物が有する本発明の DNAを単離し、 そ のェキソン部分にネオマイシン耐性遺伝子、ハイグロマイシン耐性遺伝子を代表と する薬剤耐性遺伝子、 あるいは 1 a c Z (i3—ガラクトシダーゼ遺伝子) 、 c a t (クロラムフエニコールァセチフレトランスフェラーゼ遺伝子)を代表とするレポ一 夕一遺伝子等を挿入することによりェキソンの機能を破壌する力、、あるいはェキソ ン間のイントロン部分に遺伝子の転写を終結させる DNA配列(例えば、 po l y A付加シグナルなど) を挿入し、 完全なメッセンジャー RNAを合成できなくする ことによって、結果的に遺伝子を破壊するように構築した DN A配列を有する DN A鎖 (以下、 ターゲッティングベクターと略記する) を、 例えば相同組換え法によ り該動物の染色体に導入し、得られた ES細胞について本発明の DNA上あるいは その近傍の D N A配列をプローブとしたサザンハイプリダイゼーシヨン解析ある いはターゲッティングベクター上の DN A配列と夕ーゲッティングベクタ一作製 に使用した本発明の D N A以外の近傍領域の D N A配列をプライマ一とした PC R法により解析し、本発明のノックアウト ES細胞を選別することにより得ること ができる。 Specific examples of the non-human mammalian embryonic stem cells in which the DNA of the present invention has been inactivated (hereinafter abbreviated as the DNA-inactivated ES cells of the present invention or the knockout ES cells of the present invention) include: The DNA of the present invention possessed by the target non-human mammal is isolated, and its exon portion is a drug resistance gene typified by a neomycin resistance gene, a hygromycin resistance gene, or 1acZ (i3-galactosidase gene), cat (Chloramphenicol acetyltransferase gene) Insertion of a repo-specific gene, such as the representative gene, disrupts the function of exons or terminates gene transcription in the intron between exons. By inserting a DNA sequence (for example, a poly A addition signal) and preventing the synthesis of complete messenger RNA, A DNA chain having a DNA sequence constructed so as to disrupt the gene (hereinafter abbreviated as a targeting vector) is introduced into the chromosome of the animal by, for example, a homologous recombination method. Southern hybridization analysis using a DNA sequence on or near the DNA of the present invention as a probe, or preparation of DNA sequence on targeting vector and evening targeting vector The DNA sequence can be obtained by analyzing the DNA sequence of a neighboring region other than the DNA of the present invention used as a primer by PCR and selecting the knockout ES cells of the present invention.
また、相同組換え法等により本発明の DNAを不活化させる元の ES細胞として は、 例えば、 前述のような既に樹立されたものを用いてもよく、 また公知 Evans と Kaufmaの方法に準じて新しく樹立したものでもよい。例えば、 マウスの ES細胞 の場合、 現在、 一般的には 129系の ES細胞が使用されているが、 免疫学的背景 がはっきりしていないので、 これに代わる純系で免疫学的に遺伝的背景が明らかな E S細胞を取得するなどの目的で例えば、 C 57 BLZ6マウスや C 57 BL/6 の採卵数の少なさを DBAZ2との交雑により改善した BDFiマウス (C 57 B L/6と DBAZ2との F を用いて樹立したものなども良好に用いうる。 BD Fiマウスは、 採卵数が多く、 かつ、 卵が丈夫であるという利点に加えて、 C 57 B L / 6マウスを背景に持つので、これを用いて得られた E S細胞は病態モデルマ ウスを作出したとき、 C 57 BL/ 6マウスとバッククロスすることでその遺伝的 背景を C57 BL/6マウスに代えることが可能である点で有利に用い得る。 また、 ES細胞を樹立する場合、 一般には受精後 3.5日目の胚盤胞を使用する が、 これ以外に 8細胞期胚を採卵し胚盤胞まで培養して用いることにより効率よく 多数の初期胚を取得することができる。  As the ES cells from which the DNA of the present invention is inactivated by the homologous recombination method or the like, for example, those already established as described above may be used, or according to the known Evans and Kaufma method. It may be a newly established one. For example, in the case of mouse ES cells, currently, 129 ES cells are generally used, but since the immunological background is not clear, it is an alternative pure immunological and genetic background. For example, for the purpose of obtaining ES cells in which C57BLZ6 mice and C57BL / 6 mice have a low number of eggs collected by crossing with DBAZ2, BDFi mice (C57BL / 6 and DBAZ2 It can be used satisfactorily with those established using F. BD Fi mice have the advantage of high number of eggs collected and the robustness of eggs, and also have the background of C57BL / 6 mice. ES cells obtained by using C57BL / 6 mice are advantageous in that when they create a disease model mouse, their genetic background can be replaced by C57BL / 6 mice by backcrossing with C57BL / 6 mice. When ES cells are established, generally 3.5 days after fertilization Although blastocysts of the eye are used, a large number of early embryos can be obtained efficiently by collecting and culturing the 8-cell stage embryos up to the blastocysts.
また、雌雄いずれの ES細胞を用いてもよいが、通常雄の ES細胞の方が生殖系 列キメラを作出するのに都合が良い。 また、 煩雑な培養の手間を削減するためにも できるだけ早く雌雄の判別を行なうことが望ましい。  Although either male or female ES cells may be used, male ES cells are generally more convenient for producing germline chimeras. It is also desirable to discriminate between males and females as soon as possible in order to reduce the complexity of culturing.
E S細胞の雌雄の判定方法としては、例えば、 P C R法により Y染色体上の性決 定領域の遺伝子を増幅、 検出する方法が、 その 1例としてあげることができる。 こ の方法を使用すれば、 従来、 核型分析をするのに約 106個の細胞数を要していた のに対して、 1コロニー程度の ES細胞数 (約 50個) で済むので、 培養初期にお ける E S細胞の第一次セレクションを雌雄の判別で行なうことが可能であり、早期 に雄細胞の選定を可能にしたことにより培養初期の手間は大幅に削減できる。 また、 第二次セレクシヨンとしては、 例えば、 G—バンデイング法による染色体 数の確認等により行うことができる。得られる E S細胞の染色体数は正常数の 1 0 0 %が望ましいが、 樹立の際の物理的操作等の関係上困難な場合は、 E S細胞の遺 伝子をノックアウトした後、 正常細胞 (例えば、 マウスでは染色体数が 2 n = 4 0 である細胞) に再びクローニングすることが望ましい。 . An example of a method for determining the sex of ES cells is a method of amplifying and detecting a gene in the sex-determining region on the Y chromosome by PCR. By using this method, the number of ES cells in one colony (about 50) can be reduced, compared to about 10 6 cells for karyotype analysis. The primary selection of ES cells in the early stage of culture can be performed by discriminating between males and females, and the early stages of culture can be greatly reduced by enabling the selection of male cells at an early stage. Examples of the second selection include chromosomes by the G-banding method. This can be done by checking the number. The number of chromosomes in the obtained ES cells is preferably 100% of the normal number, but if it is difficult due to physical operations at the time of establishment, knock out the gene of the ES cells, and However, it is desirable to clone again into cells whose chromosome number is 2 n = 40 in mice). .
このようにして得られた胚幹細胞株は、 通常その増殖性は大変良いが、個体発生 できる能力を失いやすいので、 注意深く継代培養することが必要である。 例えば、 Embryonic stem cell lines obtained in this way usually have very good proliferative properties, but must be subcultured carefully since they tend to lose their ontogenetic potential. For example,
S T O繊維芽細胞のような適当なフィーダ一細胞上で L I F (l~10000U/ml)存在 下に炭酸ガス培養器内 (好ましくは、 5 %炭酸ガス、 9 5 %空気または 5 %酸素、 5 %炭酸ガス、 9 0 %空気) で約 3 7 °Cで培養するなどの方法で培養し、 継代時に は、例えば、 トリプシン ZE D T A溶液(通常 0. 001-0. 5%トリプシン/ 0. l-5mM EDTA, 好ましくは約 0. 1 %トリプシン/ ImM EDTA) 処理により単細胞化し、 新たに用意した フィーダ一細胞上に播種する方法などがとられる。 このような継代は、通常 1一 3 日毎に行なうが、 この際に細胞の観察を行い、 形態的に異常な細胞が見受けられた 場合はその培養細胞は放棄することが望まれる。 In a CO2 incubator (preferably 5% CO2, 95% air or 5% oxygen, 5%) on a suitable feeder cell such as STO fibroblasts in the presence of LIF (1 to 10,000 U / ml) Culture at about 37 ° C in carbon dioxide gas, 90% air, etc. At the time of subculture, for example, trypsin ZE DTA solution (usually 0.001-0.5% trypsin / 0.1 l) -5 mM EDTA (preferably, about 0.1% trypsin / ImM EDTA), and the cells are seeded on a freshly prepared feeder cell. Such subculture is usually performed every 113 days. At this time, it is desirable to observe the cells and, if any morphologically abnormal cells are found, discard the cultured cells.
E S細胞は、 適当な条件により、 高密度に至るまで単層培養するか、 または細胞 集塊を形成するまで浮遊培養することにより、 頭頂筋、 内臓筋、 心筋などの種々の タイプの細胞に分化させることが可能であり 〔M. J. Evans及び Μ· H. Kaufman, ES cells are differentiated into various types of cells, such as parietal, visceral, and cardiac muscle, by monolayer culture up to high density or suspension culture until cell clumps are formed under appropriate conditions. [MJ Evans and Μ · H. Kaufman,
Nature第 292巻、 154頁、 1981年; G. R. Mart in, Proc. Nat l. Acad. Sci. U. S. A. 第 78巻、 7634頁、 1981年; T. C. Doetschman ら、 ジャーナル ·ォブ ·ェンブリオ ロジー 'アンド ·ェクスペリメンタル 'モルフォロジ一、第 87巻、 27頁、 1985年〕、 本発明の E S細胞を分化させて得られる本発明の D NA発現不全細胞は、インビト 口における本発明のタンパク質の細胞生物学的検討において有用である。 Nature, 292, 154, 1981; GR Mart in, Proc. Natl. Acad. Sci. USA 78, 7634, 1981; TC Doetschman et al., Journal of Embryology 'Ande'. Experimental 'Morphology I, Vol. 87, p. 27, 1985], and the DNA-deficient cell of the present invention obtained by differentiating the ES cell of the present invention is a cell biology of the protein of the present invention in the mouth. Is useful in technical studies.
本発明の D NA発現不全非ヒト哺乳動物は、該動物の mR NA量を公知方法を用 いて測定して間接的にその発現量を比較することにより、正常動物と区別すること が可能である。  The non-human mammal deficient in DNA expression of the present invention can be distinguished from a normal animal by measuring the mRNA level of the animal using a known method and indirectly comparing the expression level. .
該非ヒト哺乳動物としては、 前記と同様のものが用いられる。  As the non-human mammal, the same one as described above is used.
本発明の D N A発現不全非ヒト哺乳動物は、例えば、 前述のようにして作製した ターゲッティングベクターをマウス胚幹細胞またはマウス卵細胞に導入し、導入に より夕ーゲッティングベクタ一の本発明の D NAが不活性化された D N A配列が 遺伝子相同組換えにより、マウス胚幹細胞またはマウス卵細包の染色体上の本発明 の D NAと入れ換わる相同組換えをさせることにより、本発明の D NAをノックァ ゥトさせることができる。 The non-human mammal deficient in DNA expression of the present invention can be prepared, for example, by introducing the targeting vector prepared as described above into mouse embryonic stem cells or mouse egg cells, A homologous set in which a DNA sequence in which the DNA of the present invention is inactivated, which is one of the targeting vectors, replaces the DNA of the present invention on the chromosome of mouse embryonic stem cells or mouse egg cells by gene homologous recombination. By performing the replacement, the DNA of the present invention can be knocked out.
本発明の D N Aがノックアウトされた細胞は、本発明の D N A上またはその近傍 の D N A配列をプロ一ブとしたサザンハイブリダイゼ一ション解析または夕一ゲ ッティングベクター上の D NA配列と、ターゲッティングべクターに使用したマウ ス由来の本発明の D NA以外の近傍領域の D N A配列とをプライマーとした P C R法による解析で判定することができる。非ヒト哺乳動物胚幹細胞を用いた場合は、 遺伝子相同組換えにより、本発明の D N Aが不活性化された糸 H胞株をクローニング し、 その細胞を適当な時期、 例えば、 8細胞期の非ヒト哺乳動物胚または胚盤胞に 注入し、作製したキメラ胚を偽妊娠させた該非ヒト哺乳動物の子宮に移植する。作 出された動物は正常な本発明の D N A座をもつ細胞と人為的に変異した本発明の D N A座をもつ細胞との両者から構成されるキメラ動物である。  Cells in which the DNA of the present invention has been knocked out are subjected to Southern hybridization analysis using DNA sequences on or near the DNA of the present invention as a probe or DNA sequences on a targeting vector and targeting DNA. The determination can be made by PCR analysis using a mouse as a primer and a DNA sequence in the vicinity region other than the DNA of the present invention derived from the mouse used in the vector. When a non-human mammalian embryonic stem cell is used, the DNA of the present invention is inactivated by the homologous recombination to clone a follicular H follicle strain, and the cell is cultured at an appropriate time, for example, at the 8-cell stage. The chimeric embryo is injected into a human mammalian embryo or blastocyst, and the resulting chimeric embryo is implanted into the uterus of the pseudopregnant non-human mammal. The produced animal is a chimeric animal composed of both a normal cell having the DNA locus of the present invention and an artificially mutated cell having the DNA locus of the present invention.
該キメラ動物の生殖細胞の一部が変異した本発明の D N A座をもつ場合、 このよ うなキメラ個体と正常個体を交配することにより得られた個体群より、全ての組織 が人為的に変異を加えた本発明の D NA座をもつ細胞で構成された個体を、例えば、 コートカラ一の判定等により選別することにより得られる。 このようにして得られ た個体は、 通常、 本発明のタンパク質のヘテロ発現不全個体であり、 本発明のタン パク質のへテロ発現不全個体同志を交配し、それらの産仔から本発明のタンパク質 のホモ発現不全個体を得ることができる。  When a part of the germ cells of the chimeric animal has the mutated DNA locus of the present invention, all tissues are artificially mutated from a population obtained by crossing such a chimeric individual with a normal individual. It can be obtained by selecting individuals composed of cells having the added DNA locus of the present invention by, for example, determining the coat color. The individual thus obtained is usually an individual having a heterozygous expression of the protein of the present invention, which is crossed with an individual having a heterozygous expression of the protein of the present invention. Can be obtained.
卵細胞を使用する場合は、例えば、卵細胞核内にマイクロインジェクション法で D N A溶液を注入することによりターゲッティングベクタ一を染色体内に導入し たトランスジエニック非ヒト哺乳動物を得ることができ、これらのトランスジェニ ック非ヒト哺乳動物に比べて、遺伝子相同組換えにより本発明の D NA座に変異の あるものを選択することにより得られる。  When an egg cell is used, for example, a transgenic non-human mammal having a targeting vector introduced into a chromosome can be obtained by injecting a DNA solution into the egg cell nucleus by a microinjection method. Compared to a genic non-human mammal, it can be obtained by selecting those having a mutation at the DNA locus of the present invention by homologous recombination of the gene.
このようにして本発明の D NAがノックアウトされている個体は、交配により得 られた動物個体も該 D N Aがノックアウトされていることを核認して通常の飼育 環境で飼育継代を行なうことができる。 In the individual knocked out of the DNA of the present invention in this manner, the animal individual obtained by mating is usually bred with the nuclear recognition that the DNA has been knocked out. Breeding can be carried out in the environment.
さらに、 生殖系列の取得および保持についても常法に従えばよレ^すなわち、 該 不活化 D NAの保有する雌雄の動物を交配することにより、該不活化 D NA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うる。得られたホモザィゴート動 物は、 母親動物に対して、 正常個体 1 , ホモザィゴート複数になるような状態で飼 育することにより効率的に得ることができる。ヘテロザィゴート動物の雌雄を交配 することにより、該不活化 D N Aを有するホモザィゴートおよびへテロザィゴート 動物を繁殖継代する。  Furthermore, the germ line can be obtained and maintained according to a standard method. That is, by crossing male and female animals having the inactivated DNA, a homozygote having the inactivated DNA in both homologous chromosomes can be obtained. Animals can be obtained. The obtained homozygous animal can be efficiently obtained by rearing it in a mother animal in such a manner that there are 1 normal individual and a plurality of homozygotes. By crossing male and female heterozygous animals, homozygous and heterozygous animals having the inactivated DNA are bred and passaged.
本発明の D NAが不活性化された非ヒト哺乳動物胚幹細胞は、本発明の D NA発 現不全非ヒト哺乳動物を作出する上で、 非常に有用である。  The non-human mammalian embryonic stem cells in which the DNA of the present invention has been inactivated are extremely useful for producing the non-human mammal deficient in expressing the DNA of the present invention.
また、本発明の D NA発現不全非ヒト哺乳動物は、本発明のタンパク質により誘 導され得る種々の生物活性を欠失するため、本発明のタンパク質の生物活性の不活 性化を原因とする疾病のモデルとなり得るので、これらの疾病の原因究明及び治療 法の検討に有用である。  In addition, since the non-human mammal deficient in expression of the DNA of the present invention lacks various biological activities that can be induced by the protein of the present invention, it causes inactivation of the biological activity of the protein of the present invention. Since it can be a model of disease, it is useful for investigating the causes of these diseases and examining treatment methods.
( 8 a )本発明の D NAの欠損や損傷などに起因する疾病に対して治療 ·予防効果 を有する化合物のスクリーニング方法  (8a) A method for screening a compound having a therapeutic / preventive effect against diseases caused by DNA deficiency or damage, etc. of the present invention
本発明の D NA発現不全非ヒト哺乳動物は、本発明の D NAの欠損や損傷などに 起因する疾病に対して治療 ·予防効果を有する化合物のスクリ一ニングに用いるこ とができる。  The non-human mammal deficient in DNA expression of the present invention can be used for screening for a compound having a therapeutic / preventive effect against diseases caused by deficiency or damage of the DNA of the present invention.
すなわち、 本発明は、 本発明の D NA発現不全非ヒト哺乳動物に試験化合物を投 与し、 該動物の変化を観察 ·測定することを特徴とする、 本発明の D NAの欠損や 損傷などに起因する疾病、 例えば呼吸器疾患 〔例、 慢性閉塞性肺疾患 (慢性気管支 炎、 肺気腫) 、 びまん性汎細気管支炎、 気管支喘息、 嚢胞性線維症、 過敏性肺炎、 肺線維症など〕 、 鼻炎 (例、 アレルギー性鼻炎、 花粉症、 急性鼻炎、 慢性鼻炎、 肥 厚性鼻炎、 萎縮性鼻炎、 乾燥性前鼻炎、 血管運動性鼻炎、 壊疽性鼻炎、 副鼻腔炎な ど) 、 免疫疾患 (例、 重症筋無力症、 糸球体腎炎、 多発性硬化症、 シエーダレン症 候群、 インスリン抵抗性糖尿病、 慢性関節リウマチ、 全身性エリテマトーデス、 ァ 卜ピー性皮膚炎、白血球異常、脾機能不全または胸腺異常にともなう免疫不全など)、 炎症性腸疾患、 アレルギー性結膜炎などに対して治療 ·予防効果を有する化合物ま たはその塩のスクリーニング方法を提供する。 That is, the present invention comprises administering a test compound to a non-human mammal deficient in expressing the DNA of the present invention, and observing and measuring changes in the animal. Diseases caused by, for example, respiratory diseases (eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.), Rhinitis (eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrenous rhinitis, sinusitis, etc.), immune diseases ( Examples, myasthenia gravis, glomerulonephritis, multiple sclerosis, siedalen syndrome, insulin resistance diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, splenic dysfunction or thymic dysthymus Such as an immune deficiency) associated with, Provided is a method for screening a compound or a salt thereof having a therapeutic / preventive effect on inflammatory bowel disease, allergic conjunctivitis, and the like.
該スクリーニング方法において用いられる本発明の D NA発現不全非ヒト哺乳 動物としては、 前記と同様のものがあげられる。  Examples of the non-human mammal deficient in DNA expression of the present invention used in the screening method include those described above.
試験化合物としては、 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、 細胞抽出液、 植物抽出液、 動物組織抽出液、 血漿などがあ 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。  Test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and plasma, and these compounds are novel compounds. Or a known compound.
具体的には、 本発明の D NA発現不全非ヒト哺乳動物を、 試験化合物で処理し、 無処理の対照動物と比較し、 該動物の各器官、 組織、 疾病の症状などの変化を指標 として試験化合物の治療 ·予防効果を試験することができる。  Specifically, a non-human mammal deficient in expression of the DNA of the present invention is treated with a test compound, compared with a non-treated control animal, and changes in organs, tissues, disease symptoms, etc. of the animal are used as indices. Therapeutic and prophylactic effects of test compounds can be tested.
試験動物を試験化合物で処理する方法としては、 例えば、 経口投与、 静脈注射な どが用いられ、 試験動物の症状、 試験化合物の性質などにあわせて適宜選択す ることができる。 また、 試験化合物の投与量は、 投与方法、 試験化合物の性質など にあわせて適宜選択することができる。  As a method for treating a test animal with a test compound, for example, oral administration, intravenous injection and the like are used, and it can be appropriately selected according to the symptoms of the test animal, the properties of the test compound, and the like. The dose of the test compound can be appropriately selected according to the administration method, the properties of the test compound, and the like.
例えば呼吸器疾患 〔例、 慢性閉塞性肺疾患 (慢性気管支炎、 肺気腫) 、 びまん性 汎細気管支炎、気管支喘息、嚢胞性線維症、過敏性肺炎、肺線維症など〕、鼻炎(例、 アレルギー性鼻炎、 花粉症、 急性鼻炎、 慢性鼻炎、 肥厚性鼻炎、 萎縮性鼻炎、 乾燥 性前鼻炎、 血管運動性鼻炎、 壊疽性鼻炎、 副鼻腔炎など) 、 免疫疾患 (例、 重症筋 無力症、 糸球体腎炎、 多発性硬化症、 シエーダレン症候群、 インスリン抵抗性糖尿 病、慢性関節リゥマチ、全身性エリテマトーデス、ァトピー性皮膚炎、白血球異常、 脾機能不全または胸腺異常にともなう免疫不全など) 、 炎症性腸疾患、 アレルギー 性結膜炎などのに対して治療 ·予防効果を有する化合物をスクリーニングする場合、 本発明の D NA発現不全非ヒト哺乳動物に試験化合物を投与し、試験化合物非投与 群と肺の気腫化の違い、 呼吸機能の違いなどを上記組織で経時的に観察する。  For example, respiratory diseases (eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.), rhinitis (eg, allergy) Rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dryness proprietary rhinitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc.), immune diseases (eg, myasthenia gravis, Glomerulonephritis, multiple sclerosis, siedalen syndrome, insulin resistance diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, immunodeficiency associated with splenic dysfunction or thymic abnormalities), inflammatory bowel When screening for a compound having a therapeutic or prophylactic effect against diseases, allergic conjunctivitis, etc., a test compound may be added to a non-human mammal deficient in DNA expression of the present invention. Administered, the difference of the test compound non-administration group and emphysema of the lungs, the differences in respiratory function is observed over time in the tissue.
該スクリーニング方法において、 試験動物に試験化合物を投与した場合、該試験 動物の上記疾患症状が約 1 0 %以上、好ましくは約 3 0 %以上、 より好ましくは約 5 0 %以上改善した場合、 該試験化合物を上記の疾患に対して治療 ·予防効果を有 する化合物として選択することができる。 In the screening method, when the test compound is administered to a test animal, the disease symptom of the test animal is improved by about 10% or more, preferably about 30% or more, more preferably about 50% or more. Test compound has therapeutic and preventive effects on the above diseases Can be selected as the compound.
該スクリーニング方法を用いて得られる化合物は、上記した試験化合物から選ば れた化合物であり、本発明のタンパク質の欠損や損傷などによって引き起こされる 疾患に対して治療 ·予防効果を有するので、 該疾患に対する安全で低毒性な予防 - 治療剤などの医薬として使用することができる。 さらに、 上記スクリーニングで得 られた化合物から誘導される化合物も同様に用いることができる。  The compound obtained by using the screening method is a compound selected from the above-described test compounds, and has a therapeutic / preventive effect on a disease caused by a deficiency or damage of the protein of the present invention. Safe and low toxic prophylaxis-can be used as a medicament such as a therapeutic agent. Furthermore, compounds derived from the compounds obtained by the above screening can be used in the same manner.
該スクリーニング方法で得られた化合物は塩を形成していてもよく、該化合物の 塩としては、 生理学的に許容される酸 (例、 無機酸、 有機酸など) や塩基 (例、 ァ ルカリ金属など) 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、硫酸など) 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など) との塩などが用いられる。  The compound obtained by the screening method may form a salt. Examples of the salt of the compound include physiologically acceptable acids (eg, inorganic acids, organic acids, etc.) and bases (eg, alkali metal And the like, and especially preferred are physiologically acceptable acid addition salts. Examples of such salts include salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.), and organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, Salts with succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc. are used.
該スクリーニング方法で得られた化合物またはその塩を含有する医薬は、前記し た本発明のタンパク質を含有する医薬と同様にして製造することができる。  A drug containing the compound or a salt thereof obtained by the screening method can be produced in the same manner as the above-mentioned drug containing the protein of the present invention.
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、 ヒトまたは 哺乳動物 (例えば、 ラット、 マウス、 モルモット、 ゥサギ、 ヒッジ、 ブタ、 ゥシ、 ゥマ、 ネコ、 ィヌ、 サルなど) 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 humans or mammals (eg, rats, mice, guinea pigs, egrets, sheep, pigs, pigs, dogs, cats, dogs, monkeys). Etc.).
該化合物またはその塩の投与量は、 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、 該化合物を経口投与する場合、 一般的に成人 (体重 6 0 k gとして)の肺気腫患者においては、一日につき該化合物を約 0. 1〜1 0 O m g、 好ましくは約 1 . 0〜5 O m g、 より好ましくは約 1 . 0〜2 0 m g投与する。 非 経口的に投与する場合は、該化合物の 1回投与量は投与対象、対象疾患などによつ ても異なるが、 例えば、 該化合物を注射剤の形で通常成人 (体重 6 O k gとして) の肺気腫患者に投与する場合、 一日につき該化合物を約 0 . 0 1〜3 O m g、 好ま しくは約 0 . 1〜2 O m g、 より好ましくは約 0 . 1〜1 O m g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、体重 6 0 k g当たりに換算した量 を投与することができる。 (8 b)本発明の DNAに対するプロモーターの活性を促進または阻害する化合物 をスクリーニング方法 The dose of the compound or a salt thereof varies depending on the target disease, the subject of administration, the administration route, and the like. For example, when the compound is orally administered, it is generally used in adult (as a body weight of 60 kg) emphysema patients. Administers about 0.1 to 10 Omg, preferably about 1.0 to 5 Omg, more preferably about 1.0 to 20 mg of the compound per day. When administered parenterally, the single dose of the compound varies depending on the administration subject, target disease, and the like. For example, the compound is usually administered in the form of an injection to an adult (with a body weight of 6 O kg). When administered to patients with pulmonary emphysema, intravenous injection of about 0.01 to 3 Omg, preferably about 0.1 to 2 Omg, more preferably about 0.1 to 1 Omg of the compound per day It is convenient to administer by In the case of other animals, the dose can be administered in terms of weight per 60 kg. (8b) Method for screening for a compound that promotes or inhibits the activity of a promoter for DNA of the present invention
本発明は、 本発明の DNA発現不全非ヒト哺乳動物に、 試験化合物を投与し、 レ ポーター遺伝子の発現を検出することを特徴とする本発明の DNAに対するプロ モーターの活性を促進または阻害する化合物またはその塩のスクリーニング方法 を提供する。  The present invention provides a compound which promotes or inhibits the activity of a promoter for DNA of the present invention, which comprises administering a test compound to a non-human mammal deficient in expressing the DNA of the present invention and detecting the expression of a reporter gene. Or a method for screening a salt thereof.
上記スクリーニング方法において、本発明の DNA発現不全非ヒト哺乳動物とし ては、前記した本発明の DNA発現不全非ヒト哺乳動物の中でも、本発明の DNA がレポーター遺伝子を導入することにより不活性化され、該レポーター遺伝子が本 発明の D N Aに対するプロモーターの制御下で発現しうるものが用いられる。 試験化合物としては、 前記と同様のものがあげられる。  In the above screening method, the non-human mammal deficient in expression of the DNA of the present invention may be such that the DNA of the present invention is inactivated by introducing a reporter gene among the non-human mammals deficient in expressing the DNA of the present invention. A gene whose reporter gene can be expressed under the control of a promoter for the DNA of the present invention is used. Examples of the test compound include the same compounds as described above.
レポーター遺伝子としては、 前記と同様のものが用いられ、 /3—ガラクトシダー ゼ遺伝子 (1 a c Z) 、 可溶性アルカリフォスファターゼ遺伝子またはルシフェラ ーゼ遺伝子などが好適である。  As the reporter gene, the same ones as described above are used, and a / 3-galactosidase gene (1 ac Z), a soluble alkaline phosphatase gene or a luciferase gene is preferable.
本発明の DN Aをレポーター遺伝子で置換された本発明の DN A発現不全非ヒ ト哺乳動物では、 レポーター遺伝子が本発明の D N Aに対するプロモーターの支配 下に存在するので、レポーター遺伝子がコードする物質の発現をトレースすること により、 プロモーターの活性を検出することができる。  In a non-human mammal deficient in expression of the DNA of the present invention in which the DNA of the present invention has been replaced with a reporter gene, the reporter gene is under the control of the promoter for the DNA of the present invention, so that the substance encoded by the reporter gene may By tracing the expression, the activity of the promoter can be detected.
例えば、本発明のタンパク質をコードする D N A領域の一部を大腸菌由来の i3 _ ガラクトシダーゼ遺伝子 ( 1 a c Z) で置換している場合、 本来、 本発明のタンパ ク質の発現する組織で、本発明のタンパク質の代わりに j3_ガラクトシダーゼが発 現する。 従って、 例えば、 5—ブロモ—4—クロロー 3—インドリル一 /3_ガラク トピラノシド (X— g a 1) のような ]3—ガラクトシダーゼの基質となる試薬を用 いて染色することにより、簡便に本発明のタンパク質の動物生体内における発現状 態を観察することができる。具体的には、本発明のタンパク質欠損マウスまたはそ の組織切片をダルタルアルデヒドなどで固定し、 リン酸緩衝生理食塩液 (PBS) で洗浄後、 X-g a 1を含む染色液で、 室温または 37°C付近で、 約 30分ないし 1時間反応させた後、 組織標本を ImM EDTA/PBS溶液で洗浄することに よって、 ]S—ガラク卜シダーゼ反応を停止させ、 呈色を観察すればよい。 また、 常 法に従い、 1 a c Zをコードする mR NAを検出してもよい。 For example, when a part of the DNA region encoding the protein of the present invention is replaced by an i3_galactosidase gene (1 ac Z) derived from Escherichia coli, the tissue of the present invention that originally expresses the protein of the present invention J3_galactosidase is expressed instead of this protein. Therefore, for example, the present invention can be easily carried out by staining with a reagent which is a substrate of 3-galactosidase, such as 5-bromo-4-chloro-3-indolyl / 3 / 3-galactopyranoside (X-ga1). It is possible to observe the state of expression of this protein in animal organisms. Specifically, a protein-deficient mouse of the present invention or a tissue section thereof is fixed with dartalaldehyde or the like, washed with a phosphate buffered saline (PBS), and then stained with Xga1 at room temperature or at 37 ° C. After reacting for about 30 minutes to 1 hour at around ° C, wash the tissue specimen with ImM EDTA / PBS solution. Therefore, it is sufficient to stop the] S-galactosidase reaction and observe the coloration. Further, mRNA encoding 1 ac Z may be detected according to a conventional method.
上記スクリーニング方法を用いて得られる化合物またはその塩は、上記した試験 化合物から選ばれた化合物であり、本発明の D NAに対するプロモーター活性を促 進または阻害する化合物である。  The compound or a salt thereof obtained by using the above-mentioned screening method is a compound selected from the above-mentioned test compounds, and is a compound that promotes or inhibits the promoter activity for DNA of the present invention.
該スクリ一二ング方法で得られた化合物は塩を形成していてもよく、該化合物の 塩としては、 生理学的に許容される酸 (例、 無機酸など) や塩基 (例、 アルカリ金 属など) 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 レ 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、 硫酸など) 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など) との塩などが用いられる。  The compound obtained by the screening method may form a salt. Examples of the salt of the compound include physiologically acceptable acids (eg, inorganic acids) and bases (eg, alkali metals). And the like, and especially preferred are physiologically acceptable acid addition salts. Examples of such salts include inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.). Or a salt with an organic acid (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc.) And the like.
本発明の D N Aに対するプロモーター活性を促進または阻害する化合物または その塩は、 本発明のタンパク質の発現の調節、 該タンパク質の機能を調節すること ができるので、例えば呼吸器疾患〔例、慢性閉塞性肺疾患(慢性気管支炎、肺気腫)、 びまん性汎細気管支炎、気管支喘息、嚢胞性線維症、過敏性肺炎、肺線維症など〕、 鼻炎 (例、 アレルギー性鼻炎、 花粉症、 急性鼻炎、 慢性鼻炎、 肥厚性鼻炎、 萎縮性 鼻炎、乾燥性前鼻炎、血管運動性鼻炎、壊疽性鼻炎、副鼻腔炎など)、免疫疾患(例、 重症筋無力症、 糸球体腎炎、 多発性硬化症、 シエーダレン症候群、 インスリン抵抗 性糖尿病、 慢性関節リウマチ、 全身性エリテマトーデス、 アトピー性皮膚炎、 白血 球異常、 脾機能不全または胸腺異常にともなう免疫不全など) 、 炎症性腸疾患、 ァ レルギ一性結膜炎などの予防 ·治療剤として有用である。  The compound or a salt thereof that promotes or inhibits the promoter activity of the DNA of the present invention or the salt thereof can regulate the expression of the protein of the present invention and regulate the function of the protein. Diseases (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.), rhinitis (eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis) , Hypertrophic rhinitis, atrophic rhinitis, dry rhinitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc., immune diseases (eg, myasthenia gravis, glomerulonephritis, multiple sclerosis, siedalen syndrome) , Insulin resistance diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, immunodeficiency associated with splenic dysfunction or thymic abnormalities ), Inflammatory bowel disease, is useful as a prophylactic or therapeutic agent such as § Rerugi one conjunctivitis.
さらに、上記スクリーニングで得られた化合物から誘導される化合物も同様に用 いることができる。  Further, compounds derived from the compounds obtained by the above screening can be used in the same manner.
該スクリーニング方法で得られた化合物またはその塩を含有する医薬は、前記し た本発明のタンパク質またはその塩を含有する医薬と同様にして製造することが できる。  A drug containing a compound or a salt thereof obtained by the screening method can be produced in the same manner as the above-mentioned drug containing a protein of the present invention or a salt thereof.
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、 ヒトまたは 哺乳動物 (例えば、 ラッ卜、 マウス、 モルモット、 ゥサギ、 ヒッジ、 ブ夕、 ゥシ、 ゥマ、 ネコ、 ィヌ、 サルなど) に対して投与することができる。 The preparations obtained in this way are safe and of low toxicity, for example human or It can be administered to mammals (for example, rats, mice, guinea pigs, egrets, sheep, sheep, bush, horses, horses, cats, dogs, monkeys, etc.).
該化合物またはその塩の投与量は、 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、本発明の DN Aに対するプロモーター活性を阻害する化合 物を経口投与する場合、 一般的に成人 (体重 60 kgとして) の肺気腫患者におい ては、 一日につき該化合物を約 0. 1〜100mg、 好ましくは約 1. 0〜50m g、 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 化合物の 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、 本発 明の DN Aに対するプロモータ一活性を阻害する化合物を注射剤の形で通常成人 (体重 60 k gとして)の肺気腫患者に投与する場合、一日にっき該化合物を約 0. 01〜3 Omg、 好ましくは約 0. 1〜2 Omg、 より好ましくは約 0. 1〜: L 0 mg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、体重 60 k g当たりに換算した量を投与することができる。  The dose of the compound or a salt thereof varies depending on the disease to be treated, the subject of administration, the administration route, and the like. In a patient with emphysema weighing 60 kg, the compound is administered at about 0.1 to 100 mg, preferably about 1.0 to 50 mg, more preferably about 1.0 to 20 mg per day. When administered parenterally, the single dose of the compound varies depending on the administration subject, target disease, and the like.For example, a compound that inhibits the promoter activity against DNA of the present invention may be in the form of an injection. Normally, when administered to an adult (assuming a body weight of 60 kg) emphysema patients, the compound is administered daily at about 0.01-3 Omg, preferably about 0.1-2 Omg, more preferably about 0.1-: L per day. It is convenient to administer 0 mg by intravenous injection. In the case of other animals, the dose can be administered in terms of weight per 60 kg.
このように、 本発明の DNA発現不全非ヒ卜哺乳動物は、 本発明の DNAに対す るプロモ一夕一の活性を促進または阻害する化合物またはその塩をスクリ一ニン グする上で極めて有用であり、本発明の D N A発現不全に起因する各種疾患の原因 究明または予防 ·治療剤の開発に大きく貢献することができる。  As described above, the non-human mammal deficient in expression of the DNA of the present invention is extremely useful for screening a compound or a salt thereof which promotes or inhibits the activity of the promoter of the DNA of the present invention overnight. Yes, it can greatly contribute to the investigation of the causes of various diseases caused by DNA expression deficiency of the present invention or the development of preventive and therapeutic agents.
また、 本発明のタンパク質のプロモーター領域を含有する DNAを使って、その 下流に種々のタンパクをコードする遺伝子を連結し、 これを動物の卵細胞に注入し ていわゆるトランスジヱニック動物 (遺伝子移入動物) を作成すれば、 特異的にそ のタンパク質を合成させ、その生体での作用を検討することも可能となる。 さらに 上記プロモーター部分に適当なレポーター遺伝子を結合させ、 これが発現するよう な細胞株を樹立すれば、本発明のタンパク質そのものの体内での産生能力を特異的 に促進もしくは抑制する作用を持つ低分子化合物の探索系として使用できる。 (9) 本発明のタンパク質を含有してなる医薬  Also, using a DNA containing the promoter region of the protein of the present invention, genes encoding various proteins are ligated downstream thereof and injected into egg cells of an animal to produce a so-called transgenic animal (transgenic animal). ) Makes it possible to specifically synthesize the protein and examine its action in living organisms. Furthermore, by linking an appropriate reporter gene to the above promoter portion and establishing a cell line that expresses the reporter gene, a low-molecular compound having an action of specifically promoting or suppressing the production ability of the protein itself of the present invention in the body. Can be used as a search system. (9) a medicine containing the protein of the present invention
本発明のタンパク質は、本発明のタンパク質は肺気腫病変を有する肺で発現が低 下するため、 本発明のタンパク質をコードする DNAに異常があったり、 欠損して いる場合あるいは本発明の夕ンパク質の発現量が減少している場合には、 例えば、 呼吸器疾患 〔例、 慢性閉塞性肺疾患 (慢性気管支炎、 肺気腫) 、 びまん性汎細気管 支炎、 気管支喘息、 嚢胞性線維症、 過敏性肺炎、 肺線維症など〕 、 鼻炎 (例、 ァレ ルギー性鼻炎、 花粉症、 急性鼻炎、 慢性鼻炎、 肥厚性鼻炎、 萎縮性鼻炎、 乾燥性前 鼻炎、 血管運動性鼻炎、 壊疽性鼻炎、 副鼻腔炎など) 、 免疫疾患 (例、 重症筋無力 症、 糸球体腎炎、 多発性硬化症、 シェ一ダレン症候群、 インスリン抵抗性糖尿病、 慢性関節リウマチ、 全身性エリテマトーデス、 アトピー性皮膚炎、 白血球異常、 脾 機能不全または胸腺異常にともなう免疫不全など) 、 炎症性腸疾患、 アレルギー性 結膜炎などの種々の疾患が発症する。 Since the expression of the protein of the present invention is reduced in the lung having emphysema lesions, the protein of the present invention is defective or defective in the DNA encoding the protein of the present invention, or the protein of the present invention is defective. If the expression level of is reduced, for example, Respiratory disease (eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.), rhinitis (eg, a Rarginous rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pre-rhinitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc., immune diseases (eg, myasthenia gravis) Disease, glomerulonephritis, multiple sclerosis, Sheddalen syndrome, insulin resistance diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, immunodeficiency associated with spleen dysfunction or thymic abnormalities), Various diseases such as inflammatory bowel disease and allergic conjunctivitis develop.
したがって、 本発明のタンパク質および本発明の D N Aは、 例えば、 呼吸器疾患 〔例、 慢性閉塞性肺疾患 (慢性気管支炎、 肺気腫) 、 びまん性汎細気管支炎、 気管 支喘息、 嚢胞性線維症、 過敏性肺炎、 肺線維症など〕 、 鼻炎 (例、 アレルギー性鼻 炎、 花粉症、 急性鼻炎、 慢性鼻炎、 肥厚性鼻炎、 萎縮性鼻炎、 乾燥性前鼻炎、 血管 運動性鼻炎、 壊疽性鼻炎、 副鼻腔炎など) 、 免疫疾患 (例、 重症筋無力症、 糸球体 腎炎、 多発性硬化症、 シエーダレン症候群、 インスリン抵抗性糖尿病、 慢性関節リ ゥマチ、 全身性エリテマトーデス、 アトピー性皮膚炎、 白血球異常、 脾機能不全ま たは胸腺異常にともなう免疫不全など) 、 炎症性腸疾患、 アレルギー性結膜炎など の予防 ·治療剤などの医薬として使用することができる。 好ましくは、 呼吸器疾患 などの予防 ·治療剤、 さらに好ましくは慢性閉塞性肺疾患などの予防 ·治療剤、 さ らに好ましくは肺気腫などの予防 ·治療剤である。  Therefore, the protein of the present invention and the DNA of the present invention include, for example, respiratory diseases [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, Hypersensitivity pneumonia, pulmonary fibrosis, etc.), rhinitis (eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pro-rhinitis, vasomotor rhinitis, gangrene rhinitis, Sinusitis, etc., immune diseases (eg, myasthenia gravis, glomerulonephritis, multiple sclerosis, siedalen syndrome, insulin resistant diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, It can be used as a medicine to prevent or treat spleen dysfunction or immunodeficiency due to thymic abnormalities), inflammatory bowel disease, allergic conjunctivitis, etc. Wear. Preferably, it is a prophylactic / therapeutic agent for a respiratory disease or the like, more preferably a prophylactic / therapeutic agent for a chronic obstructive pulmonary disease or the like, and further preferably a prophylactic / therapeutic agent for an emphysema or the like.
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 NAを挿入し、 本発明の夕 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, when there is a patient in whom the protein of the present invention is reduced or deficient in a living body, (a) by administering the DNA of the present invention to the patient and expressing the protein of the present invention in the living body, (Mouth) inserting the DNA of the present invention into cells and expressing the protein of the present invention, and then transplanting the cells into a patient; or (8) administering the protein of the present invention to the patient. By doing so, the role of the protein of the present invention in the patient can be sufficiently or normally exerted.
本発明の D N Aを上記の予防,治療剤として使用する場合は、該 D N Aを単独あ るいはレトロウイルスベクタ一、 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ーなどの適当なベクターに挿入した後、常套手段に従つ 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 NA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。 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。 When the DNA of the present invention is used as the above-mentioned prophylactic or therapeutic agent, the DNA may be used alone or after being inserted into an appropriate vector such as a retrovirus vector, an adenovirus vector, an adenovirus associated virus vector, or the like. , According to conventional means And can be administered to humans or warm-blooded animals. 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 adjuvant for promoting uptake, and administered with a gene gun or a catheter such as a hydrogel catheter. When the protein of the present invention is used as the above-mentioned 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.
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。  The protein of the present invention can be used, for example, in the form of tablets, capsules, elixirs, microcapsules, or the like, which are sugar-coated as required, orally, or aseptic with water or other pharmaceutically acceptable liquids. It can be used parenterally in the form of injections, such as solutions or suspensions. For example, the protein of the present invention is mixed with physiologically acceptable carriers, flavors, excipients, vehicles, preservatives, stabilizers, binders, etc. in a unit dosage form generally required for the practice of preparations. It can be manufactured by 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、 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ーのような香味剤などが用いら れる。調剤単位形態が力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。  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, alginic acid, etc. Swelling agents such as magnesium stearate, sweeteners such as sucrose, lactose or saccharin, and flavoring agents such as peppermint, cocoa oil or cherry. When the unit dosage form is forcepsel, the above type of material may further contain a liquid carrier such as oil and fat. Sterile compositions for injection can be formulated according to standard pharmaceutical practice, such as by dissolving or suspending the active substance in a vehicle such as water for injection, or naturally occurring vegetable oils such as sesame oil or coconut oil. it can.
注射用の水性液としては、 例え 、 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] ル、 D—マンニ! ^一ル、 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、 エタノールな ど) 、 ポリアルコール (例えば、 プロピレングリコール、 ポリエチレングリコール など) 、 非イオン性界面活性剤 (伊 [Jえば、 ポリソルベート 8 0™、 H C O— 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。 Aqueous solutions for injection include, for example, physiological saline, isotonic solutions containing glucose and other adjuvants (eg, D-sorbyl), D-manni! And suitable solubilizers, for example, alcohols (eg, ethanol), polyalcohols (eg, propylene glycol, polyethylene glycol, etc.), nonionic surfactants (polysorbate 80 ™, HCO — 5 0 Etc.). Examples of the oily liquid include sesame oil and soybean oil, and may be used in combination with benzyl benzoate, benzyl alcohol, or the like as a solubilizing agent. In addition, buffers (eg, phosphate buffer, sodium acetate buffer, etc.), soothing agents (eg, benzalkonium chloride, proforce hydrochloride, etc.), stabilizers (eg, human serum albumin, polyethylene glycol, etc.) It may be blended with preservatives (eg, benzyl alcohol, phenol, etc.), antioxidants and the like. The prepared injection liquid is usually filled in a suitable ampoule.
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。  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。 本発明のタンパク質の投与量は、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、不妊症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(体重 6 0 k gとして)においては、一日につき該タンパク質を約 0. 1〜1 0 O m g、 好ましくは約 1 . 0〜5 O m g、 より好ましくは約 1 . 0〜2 0 m g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、 肺気腫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 6 O k gとして) に投与する場合、 一日につき該夕ン パク質を約 0 . 0 1〜3 O m g、 好ましくは約 0 . 1〜2 O m g、 より好ましくは 約 0 . 1〜1 O m g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、 体重 6 0 k g当たりに換算した量を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 (eg, humans, rats, mice, guinea pigs, egrets, birds, birds, higgies, bushus, dogs, dogs, 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. For example, when the protein of the present invention is orally administered for the purpose of treating infertility, it is generally required for an adult (body weight 6). (As 0 kg), about 0.1 to 10 Omg, preferably about 1.0 to 5 Omg, more preferably about 1.0 to 20 mg of the protein is administered per day. When administered parenterally, a single dose of the protein may vary depending on the administration subject, target disease, and the like. (As O kg), the protein per day is about 0.01 to 3 O mg, preferably about 0.1 to 2 O mg, more preferably about 0.1 to 1 O mg per day. Is conveniently administered by injection into the affected area. In the case of other animals, the dose can be administered in terms of the body weight of 60 kg.
本明細書において、 塩基やアミノ酸などを略号で表示する場合、 IUPAC-IUB Commiss ion on Biochemical Nomenclature による略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。 またア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。  In the present specification, bases, amino acids, and the like are represented by abbreviations based on the abbreviations by IUPAC-IUB Communication on Biochemical Nomenclature or commonly used abbreviations in the art, and examples thereof are described below. When amino acids can have optical isomers, the L-form is indicated unless otherwise specified.
D NA :デォキシリボ核酸  DNA: Deoxyribonucleic acid
c D NA :相補的デォキシリポ核酸 A アデニン c DNA: complementary deoxylipo nucleic acid A adenine
T チミン  T thymine
G グァニン  G Guanin
C RNA リポ核酸  C RNA Liponucleic acid
mRNA メッセンジャ一リポ核酸 dATP デォキシアデノシン三リン酸 dTTP デォキシチミジン三リン酸 dGTP デォキシグアノシン三リン酸 d CTP デォキシシチジン三リン酸mRNA Messenger liponucleic acid dATP Deoxyadenosine triphosphate dTTP Deoxythymidine triphosphate dGTP Deoxyguanosine triphosphate d CTP Deoxycytidine triphosphate
ATP アデノシン三リン酸 ATP adenosine triphosphate
EDTA エチレンジアミン四酢酸 EDTA ethylenediaminetetraacetic acid
SD S ドデシル硫酸ナトリウムSD S Sodium dodecyl sulfate
G 1 y グリシン G 1 y Glycine
A l a ァラニン A la alanine
Va 1 バリン  Va 1 Valine
Leu ロイシン  Leu Leucine
I 1 e  I 1 e
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  C y s Sistine
Me t メチォニン  Me t Methionin
G 1 u ダル夕ミン酸  G 1 u Dalminic acid
As ァスパラギン酸  As aspartic acid
L y s リジン Lys lysine
Ar g アルギニン  Ar g Arginine
H i s ヒスチジン  H is histidine
Ph e フエ二ルァラニン Ty r チロシン Ph e feniralanin Ty r tyrosine
T r p 卜リブ卜ファン  T r p Tribute fan
P r o  P ro
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  p G 1 u pyroglutamic acid
S e c (selenocysteine)  S e c (selenocysteine)
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  Etethyl group
Bu ブチル基  Bu butyl group
P h フエニル基  P h phenyl group
TC チアゾリジン一 4 (R) 一カルボキサミド基  TC thiazolidine-1 (R) monocarboxamide group
T o s p -トルエンスルフォニル  Tosp-toluenesulfonyl
CHO ホルミル  CHO Holmill
B z 1  B z 1
Cl Bzl 2, 6—シ  Cl Bzl 2, 6—Si
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
Figure imgf000073_0001
Z benzyloxycarponyl
Figure imgf000073_0001
B o c t—ブトキシカルポニル  B o c t—butoxycarponyl
DNP ジニトロフエニル  DNP dinitrophenyl
T r t トリチル  T r t Trityl
B um t一ブトキシメチル  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—ヒドロキシベンズトリアゾール HO O B t : 3 , 4—ジヒドロ一 3—ヒドロキシー 4一ォキソ一 HOB t 1-hydroxybenztriazole HO OB t: 3, 4-dihydro-3-hydroxy-4-oxo-1
1 , 2 , 3—ベンゾ卜リアジン  1,2,3-Benzotriazine
HO N B :卜ヒドロキシ- 5-ノルポルネン- 2, 3-ジカルポキシイミド 本願明細書の配列表の配列番号は、 以下の配列を示す。  HONB: trihydroxy-5-norpolene-2,3-dicarpoxyimide The sequence numbers in the sequence listing in the present specification show the following sequences.
〔配列番号: 1〕  [SEQ ID NO: 1]
ヒトプロコラーゲン C—プロテアーゼ ェンハンサープロテイン 2のァミノ 酸配列を示す。  The amino acid sequence of human procollagen C-protease enhancer protein 2 is shown.
〔配列番号: 2〕  [SEQ ID NO: 2]
配列番号: 1で表されるアミノ酸配列を有するヒトプロコラーゲン C—プロテ ァ一ゼ ェンハンサーブロティン 2をコードする D N Aの塩基配列を示す。  This shows the base sequence of DNA encoding human procollagen C—protein-enhancer brotin 2 having the amino acid sequence represented by SEQ ID NO: 1.
〔配列番号: 3〕  [SEQ ID NO: 3]
マウスプロコラーゲン C一プロテアーゼ ェンハンサープロテイン 2のアミ ノ酸配列を示す。  The amino acid sequence of mouse procollagen C-protease enhancer protein 2 is shown.
〔配列番号: 4〕  [SEQ ID NO: 4]
配列番号: 3で表されるアミノ酸配列を有するマウスプロコラーゲン C—プロ テアーゼ ェンハンサーブロティン 2をコ一ドする D N Aの塩基配列を示す。 〔配列番号: 5〕  This shows the nucleotide sequence of DNA encoding mouse procollagen C-proteinase enhancer brotin 2 having the amino acid sequence represented by SEQ ID NO: 3. [SEQ ID NO: 5]
実施例 2および 3で用いられたプライマーの塩基配列を示す。  3 shows the nucleotide sequences of primers used in Examples 2 and 3.
〔配列番号: 6〕  [SEQ ID NO: 6]
実施例 2および 3で用いられたプライマーの塩基配列を示す。  3 shows the nucleotide sequences of primers used in Examples 2 and 3.
〔配列番号: 7〕  [SEQ ID NO: 7]
実施例 2および 3で用いられたプライマーの塩基配列を示す。  3 shows the nucleotide sequences of primers used in Examples 2 and 3.
〔配列番号: 8〕  [SEQ ID NO: 8]
実施例 2および 3で用いられたプライマ一の塩基配列を示す。  3 shows the nucleotide sequence of a primer used in Examples 2 and 3.
以下において、実施例により本発明をより具体的にするが、 この発明はこれらに 限定されるものではない。 実施例 Hereinafter, the present invention will be more specifically described with reference to examples, but the present invention is not limited thereto. Example
実施例 1 Example 1
(1) タバコ煙曝露 COP Dモデルマウスの作製  (1) Tobacco smoke exposure COP D model mouse production
C0PDモデルは、 C57BL/6Nマウス (6週齡、 日本チヤ一ルスリバ一) に Kentucky Reference Cigarette 1R1から発生する主流煙を、 1〜4時間/日、 5日/週ずつ計 6 ヶ月間吸入させて作製した。 すなわち、 Kentucky Reference Cigarette 1R1を夕バ コ煙発生装置 (SG-200, 柴田科学株式会社) に装着し、 35 ml/puff, 10puff/min, 25puif/cigaretteの条件で主流煙を採取した。 得られた主流煙を空気で 3% (V/V) に希釈した後に、 マウスを入れたアクリル製の曝露チャンバ一に送気し、 自発呼吸 下のマウスに所定の時間タバコ煙を吸入させた。なお、 コントロール群には正常マ ウスを用いた。マウスの肺機能は摘出肺の圧一容量曲線を用いて評価した。すなわ ち、 1ヶ月、 3ヶ月および 6ヶ月間のタバコ煙曝露が終了した翌日に、 マウスをペン トバルビタール (70mg/kg, i.p.) で麻酔した後、 頸部を切開し、 気管に力ニュー レ (サーフロー留置針、 18G) を挿入した。 この気管力ニューレに人工呼吸器 (Harvard社) を連結し、 横隔膜切除下で、 99.995% 02により 10分間人工呼吸を施 した。 なお、 その際の気道内圧変化が 10 cm ¾0となるように換気量を調節した。 人工呼吸終了後、 気管を動脈クリップで閉じて肺内の degassingを行った後、 肺を 摘出した。この摘出肺に、 0〜25 cm ¾0の圧力でホルマリン緩衝液を順次注入して、 5 cm ¾0ごとの肺の体積を prethysmograph OJgobasil社)を用いて測定し、 摘出肺 の圧一容量曲線を求めた。 結果を図 1に示す。 In the C0PD model, C57BL / 6N mice (6-week-old, Japan Charls River) were inhaled with mainstream smoke generated from Kentucky Reference Cigarette 1R1 for 1 to 4 hours / day, 5 days / week for a total of 6 months. Produced. That is, the Kentucky Reference Cigarette 1R1 was attached to an evening smoke generator (SG-200, Shibata Kagaku Co., Ltd.), and mainstream smoke was collected under the conditions of 35 ml / puff, 10 puff / min, and 25 puif / cigarette. After the mainstream smoke obtained was diluted to 3% (V / V) with air, it was sent to the acrylic exposure chamber containing the mouse, and the spontaneously breathing mouse was allowed to inhale cigarette smoke for a specified period of time. . A normal mouse was used for the control group. The lung function of the mouse was evaluated using the pressure-volume curve of the isolated lung. That is, the mice were anesthetized with pentobarbital (70 mg / kg, ip) on the day after exposure to cigarette smoke for 1 month, 3 months and 6 months, and the neck was incised and the trachea was forced into the air. ((Surflow indwelling needle, 18G) was inserted. The tracheal force Nyure ventilator connecting the (Harvard Co.), under the diaphragm resection was facilities for 10 minutes ventilator by 99.995% 0 2. The ventilation was adjusted so that the change in airway pressure at that time was 10 cm cm0. After artificial respiration, the trachea was closed with an arterial clip and degassing was performed in the lungs, and the lungs were removed. A formalin buffer solution was sequentially injected into the isolated lung at a pressure of 0 to 25 cm¾0, and the volume of the lung at every 5 cm¾0 was measured using a prethysmograph (OJgobasil) to obtain a pressure-volume curve of the removed lung. Was. The results are shown in Figure 1.
( 2 ) タバコ煙曝露 C 0 P Dモデルマウス肺で発現変動する遺伝子の探索 タバコ煙曝露 C0PDモデルマウス肺で特異的に発現変動している遺伝子を明らか にするため、最終曝露が終了した翌日にマウスをベントバルピタール麻酔により致 死させ、 気管支肺胞洗浄を施行した後に肺を摘出した。摘出肺は液体窒素中で凍結 させ、 凍結組織破砕装置で粉砕した後に、 その湿重量の 10倍量に相当する Isogen (和光純薬社製) に浸した。 1ヶ月タバコ煙曝露群 (n=10) 、 そのコントロー ル群 (n=6) 、 3ヶ月タバコ煙曝露群 (n=8) 、 そのコントロール群 (n=8) 、 6ヶ月 タバコ煙曝露群 (n=8) 、 そのコントロール群 (n=8) から ISOGENを用いて、 添付の マニュアルに従って抽出した total R Aを材料とし、 ol igonucleot ide microarray (Mouse Genome U94A, U94B, U94C ; Asymetrix社) を用いて遺伝子発現解析を行つ た。 (2) Search for genes whose expression fluctuates in the lungs of C0PD model mice exposed to cigarette smoke To identify genes whose expression is fluctuated specifically in the lungs of C0PD model mice exposed to cigarette smoke, Was killed by ventalpital anesthesia, and bronchoalveolar lavage was performed, and the lungs were removed. The isolated lung was frozen in liquid nitrogen, crushed with a frozen tissue crusher, and then immersed in Isogen (Wako Pure Chemical Industries, Ltd.) equivalent to 10 times its wet weight. 1 month tobacco smoke exposure group (n = 10), its control group (n = 6), 3 month tobacco smoke exposure group (n = 8), its control group (n = 8), 6 month tobacco smoke exposure group ( n = 8) and the control group (n = 8) Using total RA extracted according to the manual as a material, gene expression analysis was performed using an oligonucleotide microarray (Mouse Genome U94A, U94B, U94C; Asymetrix).
•実験方法は、 Af iymet rix社の実験手引き書 (Express ion analys is technical manual) に従った。 その結果、 タバコ煙曝露 C0PDモデルマウスで特異的に発現が低 下する遺伝子として af fymetrix No. 108315_atが検出された。この配列を基に BLAST 検索をしたところ、当該遺伝子はマウスプロコラーゲン C一プロテア一ゼ ェン ハンサ一プロテイン 2 (PC0LCE2) をコードしていた。 そのヒトカウンターパート は、 ヒトプロコラーゲン C一プロテアーゼ ェンハンサープロテイン 2  • The experiment method was in accordance with the experiment guide (Expression analys is technical manual) of Aiymetrix. As a result, af fymetrix No. 108315_at was detected as a gene whose expression was specifically reduced in C0PD model mice exposed to cigarette smoke. When a BLAST search was performed based on this sequence, the gene was found to encode mouse procollagen C-protein-Hansa-protein 2 (PC0LCE2). Its human counterpart is human procollagen C-protease enhancer protein 2
(hPC0LCE2) であることが判明した。 実施例 2  (hPC0LCE2). Example 2
定量的 R T— P C R法による発現変動の確認  Confirmation of expression fluctuation by quantitative RT-PCR method
PC0LCE2遺伝子の発現変動が個体レベルでも顕著に認められることを確認するた め、 定量的 RT- PCR法により、 個体毎の発現を調べた。  In order to confirm that the expression fluctuation of the PC0LCE2 gene was remarkable even at the individual level, the expression of each individual was examined by quantitative RT-PCR.
マウス肺組織から調製した total RNA 200ngを出発材料として TaQMan Gold RT- PCR Ki t (アプライドバイオシステムズ社製) を用いて 50 1の反応液中で逆転写反応に より c D NAを合成した。 反応液を蒸留水で 5倍に希釈した後、 そのうちの を用いて ABI PRISM 7900シークェンスディテクター (アプライドバイオシステムズ 社製) と Quant iTect SYBR Green PCR Ki t (QIAGEN社製) を用いたリアルタイム定 量的 PCR法により、 マウス PC0LCE2遺伝子コピー数を測定した。遺伝子量検出に用い たプライマ一〔プライマー 1 (配列番号: 5 )およびプライマー 2 (配列番号: 6 )〕 はマウス PC0LCE2遺伝子の塩基配列 〔GenBank Access ion Number : AF352788] から Pr ime r Expres sプログラムを用いて設計した。 コピー数算出のための標準サンプル としては、マウス肺組織から抽出した total RNAを錶型にプライマー 3 (配列番号: 7 ) およびプライマ一 4 (配列番号: 8 ) を用いて、 RT- PCR法により増幅した 515 塩基対からなる DNA断片の濃度を Spectrophotometerにより、測定し、段階希釈する ことにより調製した。 同様にハウスキーピング遺伝子として GAPDH遺伝子のコピー 数を測定した。 また、非特異的な増幅を除去するために逆転写酵素を含まないサン プルも同様に処理し、 次式より、 total RNAあたりの遺伝子コピー数を求め、 夕バ コ煙曝露 C0PDモデルマウス肺とコントロールマウス肺の個体毎の発現を比較した。 (逆転写酵素含有サンプル中の PC0LCE2遺伝子コピー数/ GAPDH遺伝子コピ一数) 一 (逆転写酵素非含有サンプル中の PC0LCE2遺伝子コピー数/ GAPDH遺伝子コピー数) = (PC0LCE2遺伝子発現量) Using 200 ng of total RNA prepared from mouse lung tissue as a starting material, cDNA was synthesized by reverse transcription in a reaction mixture of 501 using TaQMan Gold RT-PCR Kit (manufactured by Applied Biosystems). After diluting the reaction solution 5 times with distilled water, use it for real-time quantitative analysis using ABI PRISM 7900 Sequence Detector (Applied Biosystems) and Quant iTect SYBR Green PCR Kit (QIAGEN). The mouse PC0LCE2 gene copy number was measured by PCR. The primers [Primer 1 (SEQ ID NO: 5) and Primer 2 (SEQ ID NO: 6)] used for the detection of the gene amount were obtained from the Primer Expression program based on the nucleotide sequence of the mouse PC0LCE2 gene [GenBank Accession Number: AF352788]. Designed using As a standard sample for calculating the copy number, total RNA extracted from mouse lung tissue was subjected to RT-PCR by using primers 3 (SEQ ID NO: 7) and primer 14 (SEQ ID NO: 8) for type I. The concentration of the amplified DNA fragment consisting of 515 base pairs was measured with a Spectrophotometer and prepared by serial dilution. Similarly, copy of GAPDH gene as housekeeping gene The number was measured. In order to eliminate non-specific amplification, samples without reverse transcriptase were treated in the same manner, and the number of gene copies per total RNA was calculated from the following formula. The expression of each control mouse lung was compared. (PC0LCE2 gene copy number in reverse transcriptase-containing sample / one copy of GAPDH gene) i (PC0LCE2 gene copy number / GAPDH gene copy number in reverse transcriptase-free sample) = (PC0LCE2 gene expression level)
結果を図 2に示す。  The result is shown in figure 2.
これより、 マウス PC0LCE2遺伝子 (配列番号: 4) の発現が C0PDモデルマウス肺 で有意に低下している (* *p≤0. 01) ことがわかった。 実施例 3  From this, it was found that the expression of the mouse PC0LCE2 gene (SEQ ID NO: 4) was significantly reduced in the lungs of the C0PD model mouse (** p≤0.01). Example 3
マウス PCOLCE 2遺伝子産物の組織分布の解析  Analysis of tissue distribution of mouse PCOLCE 2 gene product
マウスの各組織 (骨髄、 リンパ節、 前立腺、 胸腺、 胃、 子宮、 心齓 脳、 脾臓、 肺、 肝臓、 腎臓、 精巣、 11日目胚) の cDNA (Mouse MTC panel Iおよび Mouse MTC panel II:クロンテック社製) およびタバコ煙曝露モデルマウス肺およびコント口 —ルマウス肺 cDNA (1ヶ月、 3ヶ月および 6ヶ月処理混合群)を鎢型として、 ABI PRISM 7900シークェンスディテクター (アプライドバイオシステムズ社製) と QuantiTect SYBR Green PC Kit (Q I AGEN社製) を用いたリアルタイム定量的 PCR法によ り、 マウス PC0LCE遺伝子発現分布を調べた。 遺伝子量検出に用いたプライマー 〔プ ライマー 1 (配列番号: 5)およびプライマー 2 (配列番号: 6)〕はマウス PC0LCE2 遺伝子の塩基配列 CGenBank Accession Number: AF352788] から Primer Express プログラムを用いて設計した。 コピー数算出のための標準サンプルとしては、 マウ ス肺組織から抽出した total RNAを铸型にプライマー 3 (配列番号: 7) およびプ ライマー 4 (配列番号: 8) を用いて、 RT-PCR法により増幅した 503塩基対からな る DNA断片の濃度を Spectrophotometerにより、 測定し、 段階希釈することにより調 製した。同様にハウスキーピング遺伝子として GAPDH遺伝子のコピー数を測定した。 また、非特異的な増幅を除去するために逆転写酵素を含まないサンプルも同様に処 理し、 次式より、 total RNAあたりの遺伝子コピー数を求めることにより発現量を 求めた。 Mouse cDNA (Mouse MTC panel I and Mouse MTC panel II) for each mouse tissue (bone marrow, lymph node, prostate, thymus, stomach, uterus, heart, brain, spleen, lung, liver, kidney, testis, day 11 embryo): ABI PRISM 7900 Sequence Detector (manufactured by Applied Biosystems) and QuantiTect (Clontech) and cigarette smoke-exposed model mouse lung and control mouse lung cDNA (mixed group treated for 1 month, 3 months and 6 months) Mouse PC0LCE gene expression distribution was examined by real-time quantitative PCR using SYBR Green PC Kit (QIAGEN). The primers [Primer 1 (SEQ ID NO: 5) and Primer 2 (SEQ ID NO: 6)] used for the detection of the gene amount were designed using the Primer Express program from the mouse PC0LCE2 gene base sequence CGenBank Accession Number: AF352788]. As a standard sample for calculating the copy number, total RNA extracted from mouse lung tissue was subjected to RT-PCR using primers (SEQ ID NO: 7) and primer 4 (SEQ ID NO: 8) with type III. The concentration of the DNA fragment consisting of 503 base pairs amplified by the above was measured using a Spectrophotometer and prepared by serial dilution. Similarly, the copy number of the GAPDH gene as a housekeeping gene was measured. In order to eliminate non-specific amplification, samples without reverse transcriptase are treated in the same manner, and the expression level is determined by calculating the number of gene copies per total RNA from the following formula. I asked.
(逆転写酵素含有サンプル中の PC0LCE2遺伝子コピ一数 /GAPDH遺伝子コピー数) 一 (逆転写酵素非含有サンプル中の PC0LCE2遺伝子コピー数/ GAPDH遺伝子コピー数) = (PC0LCE2遺伝子発現量)  (One copy of the PC0LCE2 gene in the sample containing reverse transcriptase / number of copies of the GAPDH gene) one (number of copies of the PC0LCE2 gene / number of copies of the GAPDH gene in the sample containing no reverse transcriptase) = (expression of the PC0LCE2 gene)
結果を図 3に示す。  The results are shown in Figure 3.
その結果、 マウス PC0LCE2遺伝子産物 (niRNA) は肺に特異的に発現していること が判明した。 実施例 4  As a result, it was revealed that the mouse PC0LCE2 gene product (niRNA) was specifically expressed in the lung. Example 4
( 1 ) エラスターゼ誘発 C O P Dモデルマウスの作製  (1) Preparation of elastase-induced COPD model mouse
C0PDモデルは、 C57BL/6Nマウス (8週齡、 日本チヤ一ルスリバ一) にブ夕膝臓ェ ラスターゼ溶液 (和光純薬) ^ imits O xL /マウス) をハロタン麻酔下にて点鼻 投与して作製した。 なお、 コントロール群には生理食塩液点鼻マウスを用いた。 マ ウスの肺機能は摘出肺の圧—容量曲線を用いて評価した。すなわち、 エラスターゼ 投与 1日、 3日、 7曰、 14日、 21日および 35日後にマウスをペントバルビタール (70 mg/kg腹腔内投与) で麻酔した後、 頸部を切開し気管に力ニューレ (サ一フロー留 置針、 18G) を揷入した。 この気管力ニューレに人工呼吸器 (Harvard社製) を連 結し、 横隔膜切除下で 99. 995% 02を用いて 10分間人工呼吸を施した。 なお、 その 際の気道内負荷圧が 10 cm 0となるように換気量を調節した。 人工呼吸終了後、 気管を動脈クリップで閉じて肺内の degassingを行った後、 肺を摘出した。 この摘 出肺に、 0〜25 cm ¾0の負荷圧下でホルマリン緩衝液を順次注入して、 5 cm ¾0ご との肺の体積を]? lethysmograph (Ugobasi l社製)を用いて測定し、 摘出肺の圧一容 量曲線を求めた。 また、 以下の式に従い、 肺の広がり易さの指標としてコンプライ アンス値を算出した。 In the C0PD model, C57BL / 6N mice (8-week-old, Nippon-Charles River) were administered nasal administration of bushu knee elastase solution (Wako Pure Chemical Industries) ^ imits O xL / mouse under halothane anesthesia. Produced. In addition, a physiological saline solution nasal mouse was used as a control group. Mouse lung function was evaluated using the pressure-volume curve of the isolated lung. That is, 1, 3, and 7 days after administration of elastase, the mice were anesthetized with pentobarbital (70 mg / kg intraperitoneal injection) 14 days, 21 days and 35 days later, and then the neck was incised and the tracheal force was applied to the trachea. A self-flow indwelling needle, 18G) was introduced. The tracheal force Nyure the ventilator (Harvard Co., Ltd.) consolidated and subjected to 10 minutes ventilator with 99.995% 0 2 under diaphragmatic resection. The ventilation was adjusted so that the airway load pressure at that time was 10 cm0. After the end of the artificial respiration, the trachea was closed with an arterial clip to perform degassing in the lung, and the lung was removed. To the isolated lung, formalin buffer was sequentially injected under a load pressure of 0 to 25 cm¾0, and the volume of the lung at intervals of 5 cm¾0 was measured using a lethysmograph (Ugobasil). A lung pressure-volume curve was determined. In addition, the compliance value was calculated as an index of the ease of lung expansion according to the following equation.
(25 cm ¾0の負荷圧下での肺容量増加量 (mL) ) /25 (cmH20) = コンプライアンス値 結果を図 4および図 5に示す。 (Lung volume increase under a load pressure of 25 cm¾0 (mL)) / 25 (cmH 20 ) = Compliance value The results are shown in FIGS. 4 and 5.
( 2 ) エラス夕ーゼ誘発 C〇 P Dモデルマゥス肺で発現変動する遺伝子の探索 エラス夕一ゼ誘発 C0PDモデルマウス肺で特異的に発現変動している遺伝子を明 らかにするため、 エラス夕ーゼ投与 1日、 3日、 7日、 14日、 21日および 35日後にマ ウスをベントバルピタール麻酔により致死させ肺を摘出した。摘出肺は液体窒素中 で凍結させ、 凍結組織破砕装置で粉砕した後に、 その湿重量の 10倍量に相当する IS0GEN (和光純薬) に浸した。 エラス夕一ゼ投与 1日後群、 エラス夕ーゼ投与 3日後 群、 エラス夕一ゼ投与 7日後群、 エラスターゼ投与 14日後群、 エラス夕一ゼ投与 21 日後群、 エラスターゼ投与 35日後群 (いずれも n=6) およびそれらコントロール群 (いずれも n=6) から IS0GENを用いて、 添付のマニュアルに従って total RNAを調製 した。 (2) Search for genes whose expression is fluctuated in the lungs of the C〇PD model induced by Erasasease. For clarification, mice were sacrificed by ventalpital anesthesia 1 day, 3 days, 7 days, 14 days, 21 days and 35 days after administration of Elas evening, and the lungs were removed. The removed lung was frozen in liquid nitrogen, crushed with a frozen tissue crusher, and then immersed in IS0GEN (Wako Pure Chemical Industries), which is equivalent to 10 times its wet weight. Group 1 day after administration of Elas Yuze, group 3 days after administration of Elas Yuze, group 7 days after administration of Elas Yuise, group 14 days after administration of elastase, group 21 days after administration of Elas Yuze, group 35 days after administration of elastase n = 6) and their control groups (n = 6 in each case), total RNA was prepared using IS0GEN according to the attached manual.
マウス肺組織から調製した total RNA l gを出発材料として M- MLV Reverse Transcriptase (インビトロジェン社製)を用いて 50 1の反応液中で逆転写反応に より cDNAを合成した。 その反応液を用いて ABI PRISM 7700シークェンスディテクタ 一(アプライドバイオシステムズ社製)を用いたリアルタイム定量的 PCR法により、 マウス PC0LCE2遺伝子量を測定した。 遺伝子量検出に用いたプライマー 〔プライマ 一 1 (配列番号: 5 ) およびプライマー 2 (配列番号: 6 ) 〕 はマウス PC0LCE2遺 伝子の塩基配列 CGenBank Access ion Number: A E 352788 〕 から Primer Express プログラムを用いて設計した。 同様にハウスキーピング遺伝子として 18Sリポゾ一 マル RNA遺伝子の遺伝子量を測定した。 次式より、 18Sリポゾ一マル RNA遺伝子発現 量あたりの PC0LCE2遺伝子発現量を求め、 エラス夕ーゼ誘発 C0PDモデルマウス肺と コントロールマウス肺の個体毎の発現を比較した。  Using total RNA Ig prepared from mouse lung tissue as a starting material, M-MLV Reverse Transcriptase (manufactured by Invitrogen) was used to synthesize cDNA by a reverse transcription reaction in 501 reaction solutions. Using the reaction solution, the amount of mouse PC0LCE2 gene was measured by a real-time quantitative PCR method using ABI PRISM 7700 Sequence Detector I (manufactured by Applied Biosystems). The primers [Primer 1 (SEQ ID NO: 5) and Primer 2 (SEQ ID NO: 6)] used for the detection of the gene amount were obtained from the mouse PC0LCE2 gene base sequence CGenBank Accession Number: AE 352788] using the Primer Express program. Designed. Similarly, the amount of 18S liposomal RNA gene as a housekeeping gene was measured. The PC0LCE2 gene expression amount per 18S liposomal RNA gene expression amount was determined from the following equation, and the expression of each of the lungs of the C0PD model mouse and the control mouse lung induced by Erasase was compared.
(PC0CE2遺伝子発現量 /18Sリポゾ一マル RNA遺伝子発現量) = (PC0LCE2遺伝子発現 結果を図 6に示す。  (PC0CE2 gene expression level / 18S liposome RNA gene expression level) = (PC0LCE2 gene expression results The results are shown in FIG.
これより、 PC0LCE2遺伝子 (配列番号: 4 ) の発現がエラス夕ーゼ誘発 C0PDモデ ルマウス肺で有意に減少している (*≤0 . 0 5 ) ことがわかった。 産業上の利用可能性  From this, it was found that the expression of the PC0LCE2 gene (SEQ ID NO: 4) was significantly reduced (* ≦ 0.05) in the lungs of Erasase-induced C0PD model mice. Industrial applicability
本発明のタンパク質およびポリヌクレオチドは、 例えば、 呼吸器疾患 〔例、 慢性 閉塞性肺疾患 (慢性気管支炎、 肺気腫) 、 びまん性汎細気管支炎、 気管支喘息、 嚢 胞性線維症、 過敏性肺炎、肺線維症など〕 、鼻炎(例、 アレルギー性鼻炎、花粉症、 急性鼻炎、 慢性鼻炎、 肥厚性鼻炎、 萎縮性鼻炎、 乾燥性前鼻炎、 血管運動性鼻炎、 壊疽性鼻炎、 副鼻腔炎など) 、 免疫疾患 (例、 重症筋無力症、 糸球体腎炎、 多発性 硬化症、 シエーダレン症候群、 インスリン抵抗性糖尿病、 慢性関節リウマチ、 全身 性エリテマ卜一デス、 アトピー性皮膚炎、 白血球異常、 脾機能不全または胸腺異常 にともなう免疫不全など) 、 炎症性腸疾患、 アレルギー性結膜炎などの診断マーカ 一等として有用であり、 該タンパク質、 ポリヌクレオチドまたは該タンパク質に対 する抗体などを用いるスクリーニングにより得られる調節剤、該タンパク質に対す る抗体などは、 例えば、 例えば、 呼吸器疾患 〔例、 慢性閉塞性肺疾患 (慢性気管支 炎、 肺気腫) 、 びまん性汎細気管支炎、 気管支喘息、 嚢胞性線維症、 過敏性肺炎、 肺線維症など〕 、 鼻炎 (例、 アレルギー性鼻炎、 花粉症、 急性鼻炎、 慢性鼻炎、 肥 厚性鼻炎、 萎縮性鼻炎、 乾燥性前鼻炎、 血管運動性鼻炎、 壊疽性鼻炎、 副鼻腔炎な ど) 、 免疫疾患 (例、 重症筋無力症、 糸球体腎炎、 多発性硬化症、 シエーダレン症 候群、 インスリン抵抗性糖尿病、 慢性関節リウマチ、 全身性エリテマトーデス、 ァ トピー性皮膚炎、白血球異常、脾機能不全または胸腺異常にともなう免疫不全など)、 炎症性腸疾患、 アレルギー性結膜炎などの予防 ·治療剤として使用することができ る。 また、 本発明のアンチセンスポリヌクレオチドは、 本発明のタンパク質の発現 を抑制することができ、 例えば、 例えば、 呼吸器疾患 〔例、 慢性閉塞性肺疾患 (慢 性気管支炎、 肺気腫) 、 びまん性汎細気管支炎、 気管支喘息、 囊胞性線維症、 過敏 性肺炎、 肺線維症など〕 、 鼻炎 (例、 アレルギー性鼻炎、 花粉症、 急性鼻炎、 慢性 鼻炎、 肥厚性鼻炎、 萎縮性鼻炎、 乾燥性前鼻炎、 血管運動性鼻炎、 壊疽性鼻炎、 副 鼻腔炎など) 、 免疫疾患 (例、 重症筋無力症、 糸球体腎炎、 多発性硬化症、 シエー ダレン症候群、 インスリン抵抗性糖尿病、 慢性関節リウマチ、 全身性エリテマトー デス、 アトピー性皮膚炎、 白血球異常、 脾機能不全または胸腺異常にともなう免疫 不全など) 、 炎症性腸疾患、 アレルギー性結膜炎などの疾病の予防 ·治療剤として 使用することができる。 さらに、 本発明の各種のポリヌクレオチドは例えば、 呼吸 器疾患〔例、慢性閉塞性肺疾患(慢性気管支炎、肺気腫)、 びまん性汎細気管支炎、 気管支喘息、 嚢胞性線維症、 過敏性肺炎、 肺線維症など〕 、 鼻炎 (例、 アレルギー 性鼻炎、 花粉症、 急性鼻炎、 慢性鼻炎、 肥厚性鼻炎、 萎縮性鼻炎、 乾燥性前鼻炎、 血管運動性鼻炎、 壊疽性鼻炎、 副鼻腔炎など) 、 免疫疾患 (例、 重症筋無力症、 糸 球体腎炎、 多発性硬化症、 シエーダレン症候群、 インスリン抵抗性糖尿病、 慢性関 節リウマチ、 全身性エリテマトーデス、 アトピー性皮膚炎、 白血球異常、 脾機能不 全または胸腺異常にともなう免疫不全など) 、 炎症性腸疾患、 アレルギー性結膜炎 など、 好ましくは、 呼吸器疾患など、 さらに好ましくは慢性閉塞性肺疾患など、 さ らに好ましくは肺気腫などの診断、 予防または治療に有用である。 The proteins and polynucleotides of the present invention include, for example, respiratory diseases [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, sac Alveolar fibrosis, irritable pneumonia, pulmonary fibrosis, etc.), rhinitis (eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrene Rhinitis, sinusitis, etc., immune diseases (eg, myasthenia gravis, glomerulonephritis, multiple sclerosis, siedalen syndrome, insulin resistance diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic skin Inflammation, leukocyte abnormalities, immunodeficiency associated with spleen dysfunction or thymic abnormalities), diagnostic markers for inflammatory bowel disease, allergic conjunctivitis, etc., such as proteins, polynucleotides or antibodies against the proteins. Modulators obtained by screening using the antibody, antibodies against the protein, and the like include, for example, respiratory diseases (eg, Chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.), rhinitis (eg, allergic rhinitis, pollinosis, acute) Rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry proprietary rhinitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc., immune diseases (eg, myasthenia gravis, glomerulonephritis, multiple occurrences) Sclerosis, siedalen syndrome, insulin resistance diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, immunodeficiency associated with splenic dysfunction or thymic abnormalities), inflammatory bowel disease, allergies It can be used as a prophylactic and therapeutic agent for conjunctivitis. Further, the antisense polynucleotide of the present invention can suppress the expression of the protein of the present invention. For example, for example, respiratory diseases [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse Panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, pulmonary fibrosis, etc.), rhinitis (eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dryness Pronasalitis, vasomotor rhinitis, gangrenous rhinitis, sinusitis, etc., immune disorders (eg, myasthenia gravis, glomerulonephritis, multiple sclerosis, Siedaren syndrome, insulin resistant diabetes, rheumatoid arthritis, Systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, immunodeficiency associated with splenic dysfunction or thymic abnormalities), inflammatory bowel disease, allergic conjunctivitis It can be used as a preventive and therapeutic agent for diseases such as. Furthermore, various polynucleotides of the present invention include, for example, respiratory diseases (eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, Pulmonary fibrosis etc.), rhinitis (eg, allergy Rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc.), immune diseases (eg, myasthenia gravis, Glomerulonephritis, multiple sclerosis, siedalen syndrome, insulin resistance diabetes, rheumatoid arthritis, systemic lupus erythematosus, atopic dermatitis, leukocyte abnormalities, immunodeficiency associated with impaired splenic function or thymus, etc.), inflammatory It is useful for diagnosis, prevention or treatment of intestinal diseases, allergic conjunctivitis, etc., preferably respiratory diseases, etc., more preferably chronic obstructive pulmonary diseases, etc., and more preferably emphysema.

Claims

請求の範囲 The scope of the claims
I . 配列番号: 1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 酸配列を含有する夕ンパク質もしくはその部分べプチドまたはその塩の活性を促 '進する化合物またはその塩を含有してなる呼吸器疾患の予防 ·治療剤。 I. Contains a compound or a salt thereof which promotes the activity of a protein or a partial peptide thereof or a salt thereof containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1. Prevention and treatment of respiratory diseases.
2 . 配列番号: 1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 酸配列を含有するタンパク質もしくはその部分べプチドまたはその塩の遺伝子の 発現を促進する化合物またはその塩を含有してなる呼吸器疾患の予防 ·治療剤。  2. It contains a compound or a salt thereof which promotes the expression of a gene or a partial peptide or a salt thereof which has the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1. Prevention and treatment of respiratory diseases.
3 . 配列番号: 1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 酸配列を含有するタンパク質またはその部分ペプチドをコードするポリヌクレオ チドの塩基配列に相補的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 含有するァンチセンスボリヌクレオチド。 3. A base complementary or substantially complementary to the base sequence of a polynucleotide encoding a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof Antisense polynucleotide containing the sequence or a part thereof.
4 . 請求項 3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。  4. A pharmaceutical comprising the antisense polynucleotide according to claim 3.
5 . 配列番号: 1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 酸配列を含有するタンパク質またはその部分ペプチドまたはその塩に対する抗体。 5. An antibody against a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1, a partial peptide thereof, or a salt thereof.
6 . 請求項 5記載の抗体を含有してなる医薬。 6. A pharmaceutical comprising the antibody according to claim 5.
7 . 呼吸器疾患の予防 ·治療剤である請求項 6記載の医薬。  7. The medicament according to claim 6, which is an agent for preventing or treating respiratory diseases.
8 . 請求項 5記載の抗体を含有してなる診断薬。  8. A diagnostic agent comprising the antibody according to claim 5.
9 . 呼吸器疾患の診断薬である請求項 8記載の診断薬。  9. The diagnostic agent according to claim 8, which is a diagnostic agent for a respiratory disease.
1 0 . 配列番号: 1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タンパク質またはその部分べプチドをコードするポリヌクレ ォチドを含有してなる呼吸器疾患の診断薬。  10. A diagnostic agent for a respiratory disease comprising a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a polynucleotide encoding a partial peptide thereof .
I I . プロコラーゲン c末端切断促進活性を促進する作用を有する化合物または その塩を含有してなる慢性閉塞性肺疾患または気管支喘息の予防 ·治療剤。  I I. A prophylactic / therapeutic agent for chronic obstructive pulmonary disease or bronchial asthma comprising a compound having a function of promoting c-terminal cleavage promoting activity of procollagen or a salt thereof.
1 2 . 配列番号: 1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タンパク質もしくはその部分ぺプチドまたはその塩を用いる ことを特徴とする呼吸器疾患の予防 ·治療剤のスクリーニング方法。  12. Prevention of respiratory diseases characterized by using a protein or a partial peptide or a salt thereof containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 · A method for screening a therapeutic agent.
1 3 . タバコ煙曝露慢性閉塞性肺疾患モデルマウスまたはエラス夕ーゼ誘発慢性 閉塞性肺疾患モデルマウスを用いる請求項 1 2記載のスクリーニング方法。 1 3. Tobacco smoke-exposed chronic obstructive pulmonary disease model mouse or elastose-induced chronic 13. The screening method according to claim 12, wherein an obstructive pulmonary disease model mouse is used.
1 4 . 配列番号: 1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タンパク質もしくはその部分ペプチドまたはその塩を含有す ることを特徴とする呼吸器疾患の予防 ·治療剤のスクリーニング用キット。 14. Prevention of respiratory disease characterized by containing a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof · Kit for screening therapeutic agents.
1 5 . 請求項 1 2記載のスクリーニング方法または請求項 1 4記載のスクリー二 ング用キットを用いて得られうる呼吸器疾患の予防 ·治療剤。 15. A prophylactic / therapeutic agent for a respiratory disease obtainable by using the screening method according to claim 12 or the screening kit according to claim 14.
1 6 . 呼吸器疾患が肺気腫である請求項 1 5記載の予防 ·治療剤。 16. The preventive or therapeutic agent according to claim 15, wherein the respiratory disease is emphysema.
1 7 . 配列番号: 1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タンパク質またはその部分ペプチドをコードするポリヌクレ ォチドを用いることを特徴とする呼吸器疾患の予防 ·治療剤のスクリ一二ング方法。 17. Prevention of a respiratory disease characterized by using a polynucleotide encoding a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof · How to screen therapeutic agents.
1 8 . タバコ煙曝露慢性閉塞性肺疾患モデルマウスまたはエラス夕ーゼ誘発慢性 閉塞性肺疾患モデルマウスを用いる請求項 1 7記載のスクリ一二ング方法。 18. The screening method according to claim 17, wherein a tobacco smoke-exposed chronic obstructive pulmonary disease model mouse or an elastase-induced chronic obstructive pulmonary disease model mouse is used.
1 9 . 配列番号: 1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タンパク質またはその部分べプチドをコ一ドするポリヌクレ ォチドを含有することを特徴とする呼吸器疾患の予防 ·治療剤のスクリ一ニング用 キッ卜。  1 9. A respiratory tract characterized by containing a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a polynucleotide encoding a partial peptide thereof. Kit for screening of therapeutic and therapeutic agents for organ diseases.
2 0 . 請求項 1 7記載のスクリーニング方法または請求項 1 9記載のスクリー二 ング用キッ卜を用いて得られうる呼吸器疾患の予防 ·治療剤。  20. A prophylactic / therapeutic agent for respiratory diseases obtainable by using the screening method according to claim 17 or the screening kit according to claim 19.
2 1 . 呼吸器疾患が肺気腫である請求項 2 0記載の予防 ·治療剤。  21. The preventive or therapeutic agent according to claim 20, wherein the respiratory disease is emphysema.
2 2 . 哺乳動物に対し、 配列番号: 1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タンパク質もしくはその部分べプチドまた はその塩の活性を促進する化合物またはその塩、または該タンパク質の遺伝子の発 現を促進する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呼吸器疾 患の予防 ·治療方法。 22. A compound that promotes the activity of a protein containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide or a salt thereof to a mammal; A method for preventing and treating respiratory diseases, which comprises administering an effective amount of a salt thereof, a compound that promotes the expression of a gene of the protein, or a salt thereof.
2 3 . 配列番号: 1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夕ンパク質もしくはその部分べプチドまたはその塩の活性を 促進する、または該タンパク質の遺伝子の発現を促進することを特徴とする呼吸器 疾患の予防 ·治療方法。 23. Promotes the activity of a protein or a partial peptide thereof or a salt thereof containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1, or a gene for the protein A method for preventing and treating respiratory diseases characterized by promoting the expression of respiratory diseases.
2 4. 呼吸器疾患の予防 ·治療剤を製造するための、 配列番号: 1で表されるァ 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も しくはその部分べプチドまたはその塩の活性を促進する化合物またはその塩、また は該タンパク質の遺伝子の発現を促進する化合物またはその塩の使用。 2 4. A protein or partial peptide containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 for producing an agent for preventing or treating respiratory diseases Or a compound or a salt thereof which promotes the activity of a salt thereof, or a compound or a salt thereof which promotes expression of a gene of the protein.
PCT/JP2003/008167 2002-06-28 2003-06-27 Diagnostics/preventives/remedies for respiratory diseases WO2004002515A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2003246085A AU2003246085A1 (en) 2002-06-28 2003-06-27 Diagnostics/preventives/remedies for respiratory diseases

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2002-190732 2002-06-28
JP2002190732 2002-06-28

Publications (1)

Publication Number Publication Date
WO2004002515A1 true WO2004002515A1 (en) 2004-01-08

Family

ID=29996898

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2003/008167 WO2004002515A1 (en) 2002-06-28 2003-06-27 Diagnostics/preventives/remedies for respiratory diseases

Country Status (2)

Country Link
AU (1) AU2003246085A1 (en)
WO (1) WO2004002515A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102007040395A1 (en) 2007-08-27 2009-03-05 Optotech Optikmaschinen Gmbh Single and multi-strength spectacle lenses producing method for patient, involves controlling spindle such that speed of translatory relative movement at individual surface elements of surface is high corresponding to material removal
US7695718B2 (en) 2006-12-20 2010-04-13 Xoma Technology Ltd. Methods for the treatment of IL-1β related diseases

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH1095797A (en) * 1996-09-19 1998-04-14 Terumo Corp Procollagen c-protease enhancer protein
WO2000032221A2 (en) * 1998-12-01 2000-06-08 Genentech, Inc. Promotion or inhibition of angiogenesis and cardiovascularization
WO2001004311A1 (en) * 1999-07-07 2001-01-18 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO2001012660A2 (en) * 1999-08-17 2001-02-22 Sagami Chemical Research Center HUMAN PROTEINS HAVING HYDROPHOBIC DOMAINS AND DNAs ENCODING THESE PROTEINS
JP2001516580A (en) * 1997-09-17 2001-10-02 ジェネンテック・インコーポレーテッド Secretory and transmembrane polypeptides and nucleic acids encoding them

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH1095797A (en) * 1996-09-19 1998-04-14 Terumo Corp Procollagen c-protease enhancer protein
JP2001516580A (en) * 1997-09-17 2001-10-02 ジェネンテック・インコーポレーテッド Secretory and transmembrane polypeptides and nucleic acids encoding them
WO2000032221A2 (en) * 1998-12-01 2000-06-08 Genentech, Inc. Promotion or inhibition of angiogenesis and cardiovascularization
WO2001004311A1 (en) * 1999-07-07 2001-01-18 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO2001012660A2 (en) * 1999-08-17 2001-02-22 Sagami Chemical Research Center HUMAN PROTEINS HAVING HYDROPHOBIC DOMAINS AND DNAs ENCODING THESE PROTEINS

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
MOTT J.D. ET AL.: "Post-transcriptional proteolytic processing of procollagen C-terminal proteinase enhancer releases a metalloproteinase inhibitor", J. BIOL. CHEM., vol. 275, no. 2, 2000, pages 1384 - 1390, XP002972173 *
XU H. ET AL.: "Identification and expression of a novel type I procollagen C-proteinase enhancer protein gene form the glaucoma candidate region on 3q21-q24", GENOMICS, vol. 66, 2000, pages 264 - 273, XP004439337 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7695718B2 (en) 2006-12-20 2010-04-13 Xoma Technology Ltd. Methods for the treatment of IL-1β related diseases
DE102007040395A1 (en) 2007-08-27 2009-03-05 Optotech Optikmaschinen Gmbh Single and multi-strength spectacle lenses producing method for patient, involves controlling spindle such that speed of translatory relative movement at individual surface elements of surface is high corresponding to material removal

Also Published As

Publication number Publication date
AU2003246085A1 (en) 2004-01-19

Similar Documents

Publication Publication Date Title
WO2007004692A1 (en) Prophylactic/therapeutic agent and diagnostic agent for non-small cell lung cancer
WO2002053738A1 (en) Novel proteins and dnas thereof
JP4829780B2 (en) Preventive and therapeutic agents for respiratory diseases
WO2004048565A1 (en) Apoptosis-associated protein and use thereof
WO2004002515A1 (en) Diagnostics/preventives/remedies for respiratory diseases
WO2004028558A1 (en) Preventives/remedies for neurodegenerative disease
WO2001048203A1 (en) Novel protein and dna thereof
JP4445291B2 (en) Novel protein and its DNA
JP4300008B2 (en) Novel protein and its DNA
WO2004002516A1 (en) Diagnostics/preventives/re medies for respiratory diseases
JP2001299364A (en) New polypeptide and dna encoding the same
JP2001228146A (en) Use of disease-associated gene
WO2004024760A1 (en) Novel protein and dna thereof
WO2004002517A1 (en) Diagnostics/preventives/re medies for respiratory diseases
JP2001299363A (en) New polypeptide and dna encoding the same
WO2003087155A1 (en) Novel protein and dna thereof
JP2004097207A (en) Prophylactic/remedial agent for respiratory disease
JP2004105173A (en) Prophylactic/remedy for respiratory disease
JP2003219880A (en) New protein and dna thereof
JP2004105172A (en) Prophylactic/remedy for respiratory disease
JP2004187680A (en) New protein and dna thereof
WO2004039974A1 (en) Novel protein and dna thereof
WO2003062426A1 (en) Novel protein, its dna and use thereof
JP2004121245A (en) New protein and its dna
JP2003189878A (en) New protein and its dna

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 NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

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

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

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP