WO2003062426A1 - Novel protein, its dna and use thereof - Google Patents

Novel protein, its dna and use thereof Download PDF

Info

Publication number
WO2003062426A1
WO2003062426A1 PCT/JP2003/000408 JP0300408W WO03062426A1 WO 2003062426 A1 WO2003062426 A1 WO 2003062426A1 JP 0300408 W JP0300408 W JP 0300408W WO 03062426 A1 WO03062426 A1 WO 03062426A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
present
salt
dna
seq
Prior art date
Application number
PCT/JP2003/000408
Other languages
French (fr)
Japanese (ja)
Inventor
Atsushi Nakanishi
Shigeru Morita
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.
Publication of WO2003062426A1 publication Critical patent/WO2003062426A1/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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • 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

Definitions

  • the present invention relates to a novel protein, DNA encoding the protein, a method for screening a compound that inhibits the activity of the protein, a compound obtained by the screening method, and the like. More specifically, the present invention relates to a novel protein useful as a prophylactic / therapeutic agent or diagnostic agent for respiratory diseases, digestive diseases, and the like. Background art
  • Chronic obstructive pulmonary disease (chronic bronchitis, emphysema) is considered to become the central disease of respiratory illness with the aging of the smoking generation and prolonged life expectancy.
  • Bronchial asthma is a chronic inflammatory disease of the respiratory tract, with airway narrowing and paroxysmal dyspnea, wheezing, and coughing. Many cells are involved in its development and progression, including airway epithelial cells, mast cells, eosinophils, and T lymphocytes.
  • airway hyperresponsiveness One of the most important features of bronchial asthma is that the airway is more responsive to stimuli (airway hyperresponsiveness). This airway hyperreactivity is caused by inflammation of the airway, mainly by detachment of the airway epithelium by chemical mediators secreted from cells infiltrating the airway such as eosinophils, but genetic factors and environmental factors also have complex effects Is believed to be.
  • adhesion molecules such as VCAM-K ICAM-1 are found on airway epithelial cells and capillary endothelial cells around the bronchi. (J. Allergy Cl in. I Thigh unol., 96, 941 (1995)), and the site force and chemical migrants Produced.
  • Patients with bronchial asthma have enhanced function of Th2-type helper T cells, and Th2-type cytokines such as IL-3, IL-4, IL_5, IL-13, and GM-CSF. Chemokine production increases. IL-4 and IL-13 induce IgE production, and IL-3 and IL-4 increase mast cells.
  • eosinophils are differentiated and proliferated by the action of IL-5, GM-CSF, etc., and infiltrate the respiratory tract by eotaxin and RANTES (Allergy Asthma Proc.), 20, 141 (1999)].
  • the nucleotide sequence of the human CLCA1 gene has been reported in Genomics, Vol. 54, p. 200 (1998), Biochemical and Biophysical Research Communications, Biochem Biophys Res Commun, Vol. This gene has been reported to be associated with bronchial asthma and chronic obstructive pulmonary disease (W001 / 38530).
  • the one that shows homology to the base sequence of the human CLCA1 gene is the base sequence of the mouse gob-5 gene (Biochemical and Biophysical Research Communication (Biochem.
  • the present inventors have conducted intensive studies to solve the above-mentioned problems, and as a result, have found a novel gene belonging to a calcium-dependent chloride channel family, and have completed the present invention.
  • polynucleotide according to the above (8) or (9), which is a DNA (10) the polynucleotide according to the above (8) or (9), which is a DNA; (11) a polynucleotide having a base sequence represented by SEQ ID NO: 2 or SEQ ID NO: 18,
  • a medicament comprising the protein of (1) or (4) or a partial peptide thereof or a salt thereof,
  • an antisense polynucleotide comprising a nucleotide sequence complementary or substantially complementary to the nucleotide sequence of the polynucleotide according to (8) or (9) or a part thereof,
  • (26) a method for screening a compound or a salt thereof that inhibits the expression of the protein gene according to (1) or (4), characterized by using the polynucleotide according to (8) or (9);
  • a medicine comprising the compound or salt thereof according to (32) above, (34)
  • the diagnostic agent according to (19) above which is a diagnostic agent for respiratory disease, rhinitis or gastrointestinal disease.
  • the protein is (a) the amino acid sequence represented by SEQ ID NO: 1 or SEQ ID NO: 17; (b) one or two amino acids in the amino acid sequence represented by SEQ ID NO: 1 or SEQ ID NO: 17; Or more (preferably about 1 to 30, preferably 1 to 10 An amino acid sequence in which about 1 amino acid has been deleted, more preferably a number (1 to 5) amino acids, and (c) 1 or 2 or more amino acids in the amino acid sequence represented by SEQ ID NO: 1 or 17 (Preferably about 1 to 30, preferably about 1 to 10, and more preferably a number (1 to 5) of amino acid sequences; (SEQ ID NO: 1 or SEQ ID NO: 1) One or more (preferably about 1 to 30, preferably about 1 to 10, and more preferably a number of (1 to 5)) amino acids were inserted into the amino acid sequence represented by 7 An amino acid sequence, (e) one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 1 or SEQ ID NO: 17 (preferably, about 1 to 30, preferably about
  • a diagnostic agent or the like containing the polynucleotide according to the above (8) or (9) is also provided.
  • Proteins containing substantially the same amino acid sequence are human warm non-human animals (eg, guinea pigs, rats, mice, chickens, egrets, bush, Cells (eg, hepatocytes, spleen cells, neurons, glial cells, kidney B cells, bone marrow cells, mesangial cells, Langerhans cells, epidermal cells, epithelial cells, goblet cells, etc.) Endothelial cells, smooth muscle cells, fibroblasts, fibrocytes, muscle cells, adipocytes, immune cells (eg, macrophages, ⁇ cells, ⁇ cells, natura Killer cells, mast cells, neutrophils, basophils, eosinophils, monocytes),
  • the amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 is about 70% or more, preferably about 80% or more, preferably about 90% or more as the amino acid sequence represented by SEQ ID NO: 1. % Or more, more preferably about 95% or more, and even more preferably about 97% or more.
  • 'A a protein having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1, for example, a protein having an amino acid sequence substantially the same as the amino acid sequence represented by the aforementioned SEQ ID NO: 1 And a protein having substantially the same activity as the protein having the amino acid sequence represented by SEQ ID NO: 1.
  • the amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 17 includes about 70% or more, preferably about 80% or more, preferably Amino acid sequences having about 90% or more, more preferably about 95% or more, more preferably about 97% or more homology, and the like.
  • Examples of the protein having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 17 include, for example, an amino acid sequence substantially the same as the amino acid sequence represented by the aforementioned SEQ ID NO: 17 And a protein having substantially the same activity as the protein having the amino acid sequence represented by SEQ ID NO: 17 is preferable.
  • the amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 19 includes about 70% or more, preferably about 80% or more, preferably Amino acid sequences having a homology of about 90% or more, more preferably about 95% or more, and even more preferably about 97% or more.
  • Examples of the protein having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 19 include, for example, an amino acid sequence substantially the same as the amino acid sequence represented by the aforementioned SEQ ID NO: 19 And a protein having substantially the same activity as the protein having the amino acid sequence represented by SEQ ID NO: 19.
  • substantially the same activity examples include chloride channel activity (eg, calcium-dependent chloride channel activity and the like).
  • Substantially homogenous indicates that the properties are homologous in nature (eg, physiologically or pharmacologically). Therefore, it is preferable that the chloride channel activities are equivalent (eg, about 0.01 to: L 0 times, preferably about 0,0 to 10 times, more preferably 0.5 to 2 times).
  • the quantitative factors such as the degree of these activities and the molecular weight of the protein may be different.
  • the activity such as chloride channel activity can be measured according to a known method.
  • the activity can be measured according to the method described in Genomics, Vol. 54, p. 200 (1998) or a method analogous thereto. it can.
  • Examples of the protein of the present invention include (1) (a) one or more (preferably about 1 to 30 and preferably 1 to 30) amino acids in the amino acid sequence represented by SEQ ID NO: 1. About 10 amino acids, more preferably an amino acid sequence in which the number of amino acids (1 to 5) has been deleted, and (b) one or two or more amino acids (preferably About 30 amino acids, preferably about 1-10 amino acids, and more preferably about (1-5) amino acids; (c) the amino acid sequence represented by SEQ ID NO: 1 An amino acid sequence into which one or more (preferably about 1 to 30, preferably about 1 to 10, and more preferably a number (1 to 5)) amino acids have been inserted; ) SEQ ID NO: 1 or 2 or more in the amino acid sequence represented by 1 (preferably about 1 to 30 About 1 to 10 amino acids, more preferably about 1 to 5 amino acids are substituted with another amino acid, or (e) a protein containing an amino acid sequence combining them. So-called mutein,
  • amino acid sequence represented by SEQ ID NO: 17 preferably Preferably, the amino acid sequence has about 1 to 30 amino acids, preferably about 1 to 10 amino acids, more preferably about 1 to 5 amino acids, and (b) SEQ ID NO: 17.
  • amino acid sequence in which one or more (preferably about 1 to 30, preferably about 1 to 10, and more preferably about 1 to 5) amino acids have been added to the amino acid sequence to be (C) one or more amino acid sequences represented by SEQ ID NO: 17 (preferably about 1 to 30, preferably about 1 to 10, more preferably about 1 to 5 ) Amino acid sequence; (d) one or more (preferably about 1 to 30; preferably 1 to 1) amino acids in the amino acid sequence represented by SEQ ID NO: 17
  • the protein has a N-terminus at the left end (amino terminus) and a C-terminus at the right end (capillary end) according to the convention of peptide labeling.
  • the protein of the present invention including the protein containing the amino acid sequence represented by SEQ ID NO: 1, has a C-terminus having a lipoxyl group (-C00H), a carboxylate (-C00-), and an amide (-C0NH). 2 ) or ester (-C00R).
  • R in the ester e.g., methyl, Echiru, n- propyl Le, isopropyl, CM alkyl group, such as n- butyl, for example, C 3.
  • cycloalkyl groups cyclopentyl Le, cyclohexane, etc. cyclohexyl, for example, phenyl, alpha-Na C 6 such Fuchiru - 12 ⁇ Li Ichiru group, e.g., benzyl, phenyl, such as phenethyl - C, _ 2 alkyl or flight one naphthylmethyl etc.
  • a- Nafuchiru C M such alkyl Le group C 7 _ 14
  • Ararukiru group such as pin bar opening Iruokishimechiru group is used.
  • the protein of the present invention When the protein of the present invention has a lipoxyl group (or carboxylate) other than the C-terminus, the protein of the present invention includes a lipoxyl group amidated or esterified.
  • the ester in this case, for example, the above-mentioned C-terminal ester and the like are used.
  • the amino group at the N-terminal amino acid residue (eg, methionine residue) is protected by a protecting group (eg, C Hi acetyl group such as alkanol such as formyl group and acetyl group).
  • a protecting group eg, C Hi acetyl group such as alkanol such as formyl group and acetyl group.
  • N-terminal glutamine residue generated by cleavage in vivo, which has been oxidized with lipamine Substituents on the side chains of amino acids in the molecule (eg, -0H, -SH, amino groups , imidazo Ichiru group, I Ndoru group is protected with Guanijino group, etc.) a suitable protecting group (e.g., formyl group, C WINCH 6 Ashiru groups such as C Bok 6 Al force Noiru group such as ⁇ Se methyl group) And complex proteins such as so-called glycoproteins to which sugar chains are bound.
  • a suitable protecting group e.g., formyl group, C WINCH 6 Ashiru groups such as C Bok 6 Al force Noiru group such as ⁇ Se methyl group
  • complex proteins such as so-called glycoproteins to which sugar chains are bound.
  • protein of the present invention examples include, for example, a protein containing an amino acid sequence represented by SEQ ID NO: 1, a protein containing an amino acid sequence represented by SEQ ID NO: 17, SEQ ID NO: 19 And a protein containing the amino acid sequence represented by
  • the partial peptide of the protein of the present invention is the partial peptide of the protein of the present invention described above, and preferably any peptide having the same properties as the protein of the present invention described above.
  • 20 or more, preferably 50 or more, more preferably 70 or more, more preferably 100 or more, and most preferably 200 or more Peptides having the above amino acid sequences are used.
  • one or more (preferably about 1 to 10, more preferably about 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 One or two or more (preferably about 1 to 20, more preferably about 1 to 10, and more preferably a number (1 to 5)) amino acids are inserted into the amino acid sequence, or Even if one or two or more (preferably about 1 to 10, more preferably several, and more preferably about 1 to 5) amino acids in the amino acid sequence are substituted with another amino acid, Good.
  • the C-terminus force Rupokishiru group (-C00H), Karupoki Shireto (-C00-), can be any of the amide (_C0NH 2) or ester (-C00R).
  • the partial peptide of the present invention includes, as in the case of the protein of the present invention described above, a peptide having a carbonyl group (or carboxylate) other than the C-terminal, an N-terminal amino acid residue (eg, methionine).
  • the amino group of the system is protected with a protecting group, the N-terminal is cleaved in vivo, the glutamine residue is oxidized by pyridalamine, and the substituent on the side chain of the amino acid in the molecule is Also included are those protected with an appropriate protecting group, and complex peptides such as so-called glycopeptides to which a sugar chain is bound.
  • the partial peptide of the present invention can also be used as an antigen for producing an antibody.
  • salts with physiologically acceptable acids eg, inorganic acids, organic acids
  • bases eg, alkali metal salts
  • Acceptable acid addition salts are preferred.
  • Such salts include, for example, salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid) or organic acids (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).
  • inorganic acids eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid
  • organic acids 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.
  • the protein of the present invention or a partial peptide thereof or a salt thereof can be prepared by the above-mentioned human
  • the protein can be produced from a warm-blooded animal cell or tissue by a known method for purifying a protein, or can be produced by culturing a transformant containing a DNA encoding the protein. It can also be produced according to the peptide synthesis method described below.
  • the tissues or cells of a human or non-human mammal are homogenized, extracted with an acid or the like, and the extracted liquid is subjected to reverse phase chromatography and ion exchange. Purification and isolation can be performed by combining chromatography such as chromatography.
  • a commercially available resin for protein synthesis can be generally used.
  • resins include chloromethyl resin, hydroxymethyl resin, benzylhydrylamine resin, aminomethyl resin, 4-benzyloxybenzyl alcohol resin, 4-methylbenzhydrylamine resin, and PAM resin.
  • an amino acid having an amino group and a side chain functional group appropriately protected is condensed on the resin in accordance with the sequence of the target protein according to various known condensation methods.
  • the protein or partial peptide is cleaved 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 to obtain the target protein or partial peptide or an amide thereof. get.
  • carbodiimides are particularly preferable.
  • the carbodiimides DCC, N, N'-diisopropylcarbodiimide, N-ethyl-N, 1- (3-dimethylaminoprolyl) carbodiimide and the like are used.
  • Activation by these involves adding the protected amino acid directly to the resin along with a racemization inhibitor additive (eg, HOBt, H ⁇ Bt), or by adding a symmetrical anhydride or HOBt ester or H ⁇ OB.
  • a racemization inhibitor additive eg, HOBt, H ⁇ Bt
  • Pre-protected amino as t-ester The acid can be added to the resin after activation.
  • the solvent used for the condensation of the protected amino acid with the activated 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-dimethylacetoamide, N-methylpyrrolidone, halogenated hydrocarbons such as methylene chloride, methylform, and trifluoromethyl Alcohols such as ethanol, sulfoxides such as dimethyl sulfoxide, ethers such as pyridine, dioxane, and tetrahydrofuran; nitriles such as acetonitrile and propionitrile; esters such as methyl acetate and ethyl acetate; or an appropriate mixture thereof.
  • Etc acid amides such as N, N-dimethylformamide, N, N-dimethylacetoamide, N-methylpyrrolidone, halogenated hydrocarbons such as methylene chloride, methylform, and trifluoromethyl Alcohol
  • the reaction temperature is appropriately selected from the range known to be usable for the protein bond formation reaction, and is usually selected from the range of about 120 ° C to 50 ° C.
  • the activated amino acid derivative is usually used in a 1.5 to 4-fold excess.
  • Test Bok results with two Nhidorin reaction condensation is also sufficient Repeat c reactions which can perform sufficient condensation by repeating the condensation reaction without removal of the protecting group to be insufficient When condensation cannot be obtained, acetylation of unreacted amino acid using acetic anhydride or acetylimidazole can prevent the subsequent reaction from being affected.
  • Examples of the protecting group for the amino group of the starting material include Z, Boc, t-pentyloxycarbonyl, isoboriloxycarbonyl, 4-methoxybenzyloxycarbonyl, C1-1Z, Br—Z, and adamantylo.
  • Xycarponyl, trifluoroacetyl, phthaloyl, formyl, 2-nitrophenylsulfenyl, diphenylphosphinothioyl, Fmoc and the like are used.
  • the lipoxyl group can be, for example, alkyl esterified (eg, methyl, ethyl, propyl, butyl, t-butyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, 2-adamantyl, etc.) Alkyl esterification), aralkyl esterification (e.g., benzyl ester, 412-methyl benzyl ester, 4-methoxybenzyl ester, 4-methyl benzyl ester, benzhydryl esterification), phenacyl esterification, benzene Ziroxycarponyl hydrazide, t-butoxycarbonyl hydrazide, It can be protected by trityl hydrazide or the like.
  • alkyl esterified eg, methyl, ethyl, propyl, butyl, t-butyl, cyclopentyl,
  • the hydroxyl group of serine can be protected, for example, by esterification or etherification.
  • Suitable groups for this esterification include, for example, lower (( ⁇ -6) alkanoyl groups such as acetyl group, aroyl groups such as benzoyl group, and groups derived from carbonic acid such as benzyloxycarbonyl and ethoxycarbonyl groups.
  • Examples 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, Bz 1, C 1 2 - B zl, 2- nitrobenzyl, B r- Z, such as t one-butyl is used.
  • Examples of the protecting group for histidine imidazole include Tos, 4-methoxy-1,2,3,6-trimethylbenzenesulfonyl, DNP, benzyloxymethyl, Bum, Boc, Trt, and Fmoc.
  • Examples of the activated form of the raw oxypoxy group include, for example, the corresponding acid anhydride, azide, and active ester [alcohol (for example, pen phenol, 2,4,5-trichloro phenol, 2, 4-dinitrophenol, cyanomethyl alcohol, paranitrophenol, HONB, N-hydroxysuccinimide, N-hydroxyphthalimide, and esters with HOB t)].
  • alcohol for example, pen phenol, 2,4,5-trichloro phenol, 2, 4-dinitrophenol, cyanomethyl alcohol, paranitrophenol, HONB, N-hydroxysuccinimide, N-hydroxyphthalimide, and esters with HOB t
  • As the activated amino group of the raw material for example, a corresponding phosphoramide is used.
  • Methods for removing (eliminating) the protecting group include, for example, catalytic reduction in a hydrogen stream in the presence of a catalyst such as Pd-black or Pd-carbon, or hydrogen fluoride anhydride, methanesulfonic acid, trifluoromethane, or the like.
  • the elimination reaction by the above-mentioned acid treatment is generally carried out at a temperature of about 20 ° C to 40 ° C.
  • the protection of the functional group which should not be involved in the reaction of the raw materials, the protection group, the elimination of the protective group, the activation of the functional group involved in the reaction, and the like can be appropriately selected from known groups or known means.
  • an amide form of a protein or partial peptide for example, first, amidation and protection of the ⁇ -hydroxyl group of the amino acid at the carboxy terminal, and then a peptide (protein) chain having a desired chain length on the amino group side Then, a protein or partial peptide from which only the protecting group of the ⁇ -amino group at the ⁇ -terminal of the peptide chain is removed and a protein or partial peptide from which only the protecting group of the C-terminal lipoxyl group is removed Then, these proteins or peptides are condensed in a mixed solvent as described above. Details of the condensation reaction are the same as described above.
  • an ester of a protein or peptide for example, after condensing the ⁇ -lipoxyl group of the terminal amino acid with a desired alcohol to form an amino acid ester, in the same manner as the amide of a protein or peptide, It is possible to obtain an ester of the desired protein or peptide.
  • the partial peptide of the present invention or a salt thereof can be produced according to a known peptide synthesis method or by cleaving the protein of the present invention with an appropriate beptidase.
  • 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 remaining peptide is condensed with the partial peptide or amino acid that can constitute the partial peptide of the present invention, and the product is protected.
  • the target peptide can be produced by removing the protecting group.
  • Known condensation methods and elimination of protecting groups include, for example, the methods described in the following (a) to (e).
  • the partial peptide of the present invention can be purified and isolated by combining these methods.
  • the partial peptide obtained by the above method is an isomer, it can be converted into an appropriate salt by a known method or a method analogous thereto, and conversely, when the partial peptide is obtained by a salt, the known method or Can be converted into a free form or another salt by a method according to the above.
  • the polynucleotide encoding the protein of the present invention may be any polynucleotide containing a nucleotide sequence (DNA or RNA, preferably DNA) encoding the protein of the present invention described above. Is also good.
  • the polynucleotide is RNA such as DNA or mRNA encoding the protein of the present invention, and may be double-stranded or single-stranded. In the case of double-stranded, it may be double-stranded DNA, double-stranded RNA or DNA: RNA hybrid. In the case of a single strand, it may be a sense strand (that is, a coding strand) or an antisense strand (that is, a non-coding strand).
  • the DNA encoding the protein of the present invention includes the aforementioned protein of the present invention. It may be any as long as it contains a nucleotide sequence encoding a protein. Also, genomic DNA, genomic DNA library, cDNA derived from the above-mentioned cells and tissues, and cells and tissues derived from the above-mentioned cells Any of a cDNA library or synthetic DNA may be used.
  • the vector used for the library may be any of bacteriophage, plasmid, cosmid, phagemid and the like. Alternatively, it can be directly amplified by Reverse Transcriptase Polymerase Chain Reaction (hereinafter abbreviated as RT-PCR method) using a preparation of the entire RNA or mRNA fraction from the above-mentioned cell'tissue.
  • RT-PCR method Reverse Transcriptase Polymerase Chain Reaction
  • Examples of the DNA encoding the protein of the present invention include: (1) a DNA containing the nucleotide sequence represented by SEQ ID NO: 2 or a highly stringent DNA with the nucleotide sequence represented by SEQ ID NO: 2 A DNA encoding a protein having a nucleotide sequence that hybridizes under the conditions and having substantially the same properties as a protein having the amino acid sequence represented by SEQ ID NO: 1, (2) SEQ ID NO: 18 A DNA containing the nucleotide sequence represented by SEQ ID NO: 17, or a nucleotide sequence hybridizing under high stringent conditions with the nucleotide sequence represented by SEQ ID NO: 18; A DNA encoding a protein having substantially the same properties as the contained protein, (3) a DNA containing the nucleotide sequence represented by SEQ ID NO: 20, or a nucleotide sequence represented by SEQ ID NO: 20; DNA containing a nucleotide sequence that hybridizes under high stringent conditions and encoding a protein having substantially the same properties
  • DNA that can be hybridized with the nucleotide sequence represented by SEQ ID NO: 2 under high stringency conditions include, for example, about 70% or more, preferably about 80% or more of the nucleotide sequence represented by SEQ ID NO: 2.
  • DNA containing a base sequence having a homology of about 90% or more, more preferably about 95% or more, and more preferably about 97% or more is used.
  • Examples of the DNA that can hybridize with the base sequence represented by SEQ ID NO: 18 under high stringent conditions include, for example, the base sequence represented by SEQ ID NO: 18 DNA containing a base sequence having about 70% or more, preferably about 80% or more, preferably about 90% or more, more preferably about 95% or more, more preferably about 97% or more with the sequence. Is used.
  • Examples of the DNA that can hybridize with the nucleotide sequence represented by SEQ ID NO: 20 under high stringency conditions include, for example, about 70% or more, preferably about 80% or more of the nucleotide sequence represented by SEQ ID NO: 20 DNA containing a base sequence having homology of preferably about 90% or more, more preferably about 95% or more, and more preferably about 97% or more is used.
  • Hybridization is performed according to a known method or a method analogous thereto, for example, a method described in Molecular Cloning 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). be able to.
  • a commercially available library it can be performed according to the method described in the attached instruction manual. More preferably, it can be performed under high stringency conditions.
  • the high stringent conditions include, for example, a sodium concentration of about 19 to 40 mM, preferably about 19 to 2 OmM, and a temperature of about 50 to 70 ° (:, preferably about 60 to 65 ° C.). In particular, a sodium concentration of about 19 mM and a temperature of about 65 ° C. are most preferable.
  • the DNA encoding the protein containing the amino acid sequence represented by SEQ ID NO: 1 is a DNA containing the base sequence represented by SEQ ID NO: 2, represented by SEQ ID NO: 17
  • the DNA encoding the protein containing the amino acid sequence represented by SEQ ID NO: 18 includes the DNA containing the base sequence represented by SEQ ID NO: 18, and the DNA encoding the protein containing the amino acid sequence represented by SEQ ID NO: 19
  • a DNA containing the base sequence represented by SEQ ID NO: 20 is used.
  • any DNA may be used as long as it contains the above-described nucleotide sequence encoding the partial peptide of the present invention.
  • DNA encoding the partial peptide of the present invention include a DNA having a part of a DNA having a base sequence represented by SEQ ID NO: 2, SEQ ID NO: 18 or SEQ ID NO: 20; SEQ ID NO: 2, SEQ ID NO: 18 or SEQ ID NO: 20 contains a nucleotide sequence that hybridizes under high stringent conditions with the nucleotide sequence represented by SEQ ID NO: 20; SEQ ID NO: 1, SEQ ID NO: 17 or A DNA containing a part of a DNA encoding a protein having substantially the same activity as the protein containing the amino acid sequence represented by SEQ ID NO: 19 is used.
  • the DNA hybridizable with the nucleotide sequence represented by SEQ ID NO: 2, SEQ ID NO: 18 or SEQ ID NO: 20 has the same significance as described above.
  • Means for cloning DNA that completely encodes the protein or partial peptide of the present invention (hereinafter, in the description of the cloning and expression of DNAs encoding them, these may be simply referred to as 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. Hybridization can be carried out, for example, 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 by PCR or a known kit such as Mutan TM _super Express Km (Takara Shuzo Co., Ltd.) or Mutan TM -K (Takara Shuzo Co., Ltd.) using the 0DA-LA PCR method. It can be performed according to a known method such as the gapped duplex method or the Kunkel method, or a method analogous thereto.
  • the DNA encoding the cloned protein may be used as is, or may be digested with restriction enzymes or added with a linker, if desired. Can be.
  • the DN ⁇ may have ATG as a translation initiation codon at its 5 ′ end, and may have TAA, TGA or TAG as a translation termination codon at its 3 ′ end. These translation initiation codon and translation termination codon can also be added using an appropriate synthetic DNA adapter.
  • the expression vector of the protein of the present invention may be prepared, for example, by (a) cutting out a DNA fragment of interest from DNA encoding the protein of the present invention, and (mouth) converting the DNA fragment into a promoter in an appropriate expression vector. It can be manufactured by connecting downstream of
  • the vector examples include a plasmid derived from Escherichia coli (eg, pBR322, pBR325, pUC12, pUC13), a plasmid derived from Bacillus subtilis (eg, pUB110, TP5, pC194), a plasmid derived from yeast ( For example, pSH19, pSH15), bacteriophages such as phage ⁇ , animal viruses such as retrovirus, vaccinia virus, baculovirus, etc., ⁇ 1-11, ⁇ Tl, pRc / CMV, pRc / RSV, pc DN AI ZNeo is used.
  • Escherichia coli eg, pBR322, pBR325, pUC12, pUC13
  • Bacillus subtilis eg, pUB110, TP5, pC194
  • yeast e.g, pSH19, pSH15
  • the promoter of the present invention may be any promoter as long as it is suitable for the host used for gene expression.
  • SRo! Promoter SV40 promoter, LTR promoter, CMV promoter, HSV-TK Promoter and the like can be mentioned.
  • the CMV (cytomegalovirus) promoter the SRa promoter, etc.
  • the host is Eshierihia genus
  • trp promoter one, lac promoter, rec A promoter, XP L pro motor-, l pp promoter, T 7 promoter
  • the host is Bacillus, SP_ ⁇ 1
  • yeast such as a promoter, SP02 promoter, penP promoter, etc., PH05 promoter, PGK promoter, GAP promoter, ADH promoter and the like are preferable.
  • a polyhedrin promoter, a P10 promoter and the like are preferable.
  • the expression vector may include an enhancer, A signal containing a signal, a poly-A addition signal, a selection marker, an SV40 replication origin (hereinafter sometimes abbreviated as SV400ri) and the like can be used.
  • Selection methods include, for example, dihydrofolate reductase (hereinafter sometimes abbreviated as dhfr) gene (methotrexate (MTX) resistance), ampicillin resistance gene (hereinafter sometimes abbreviated as Amp 1 ) And neomycin resistance gene (hereinafter sometimes abbreviated as Ne ⁇ 1 ⁇ G418 resistance), etc.
  • dh fr gene is used as a selection marker using Chinese hamster cells lacking the dh fr gene.
  • the target gene can be selected by using a medium containing no thymidine.
  • a signal sequence suitable for the host is added to the N-terminal side of the protein of the present invention.
  • the PhoA * signal sequence and OmpA signal sequence are used.
  • the host is a Bacillus genus
  • the human amylase signal sequence and subtilisin signal sequence are used.
  • the host is an animal cell, an insulin signal sequence, an ⁇ -interferon signal sequence, an antibody molecule, a signal sequence, etc. Available for each.
  • a transformant can be produced using the vector containing D ⁇ ⁇ ⁇ ⁇ which encodes the protein of the present invention thus constructed.
  • Escherichia bacteria for example, Escherichia bacteria, Bacillus bacteria, yeast, insect cells, insects, animal cells, and the like are used.
  • Escherichia include, for example, Escherichia coli
  • Bacillus bacteria examples include, for example, Bacillus subtilis MI 114 [Gene, 24, 255 (1983)], 207-21 [Journal of Biochemistry, 95] , 87 (1984)].
  • yeast examples include, for example, Saccharomyces cerevisiae AH22, AH22R-, NA87-11A, DKD-5D, 20B-12 Shizosaccharomyces' bomb (Schizosaccharomyces pombe) NC YC 1913, NCYC 2036 , Pichia pastoris K M71 and the like are used.
  • insect cells for example, when the virus is Ac NPV, a cell line derived from a larva of night moth (Spodoptera frugiperda cell; Sf cell), MGl cell derived from the midgut of Trichoplusia ni, High derived from egg of Trichoplusia ni Five TM cells, cells derived from Mamestra brassicae or cells derived from Estigmena acrea are used.
  • Sf cell a silkworm-derived cell line (Bombyx mori N cell; BmN cell) or the like is used.
  • Sf cells include Sf9 cells (ATCC CRL1711), Sf21 cells (Vaughn, J.L., et al., In Vipo).
  • insects for example, silkworm larvae are used [Maeda et al., Nature, 315, 592 (1985)].
  • animal cells examples include monkey cell COS-7, Vero, Chinese Hams Yuichi cell CHO '(hereinafter abbreviated as CHO cell), dh fr gene-deficient Chinese eight-muster cell CHO (hereinafter CHO (dh fr- ) Cells), mouse L cells, mouse AtT-20, mouse myeloma cells, rat GH3, and human FL cells.
  • Transformation of Bacillus can be performed, for example, according to the method described in Molecular & General Genetics, Vol. 168, 11 (1979).
  • a liquid medium is suitable as a medium used for culturing, and a carbon source necessary for the growth of the transformant is contained therein.
  • Nitrogen sources inorganic substances and others.
  • carbon sources include glucose, dextrin, soluble starch, and sucrose.
  • nitrogen sources include ammonium salts, nitrates, corncheap, lica-peptone, casein, meat extract, soybean meal, and potato extract.
  • the inorganic or organic substance such as a liquid and the inorganic substance include calcium chloride, sodium dihydrogen phosphate, and magnesium chloride.
  • yeast extract, vitamins, growth promoting factors and the like may be added.
  • the pH of the medium is preferably about 5 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 ed. Preferred is "in 'molecular” Genetics (Journal of Experiments in Molecular Genetics), 431-433, Cold Spring Harbor Laboratory, New York 1972). If necessary, a drug such as 3) 3-indolylacrylic acid can be added to make the promoter work efficiently if necessary.
  • culturing is usually performed at about 15 to 43 ° C for about 3 to 24 hours, and if necessary, aeration and stirring may be added.
  • the cultivation is usually performed at about 30 to 40 ° C for about 6 to 24 hours, and if necessary, aeration and stirring may be added.
  • the medium used is Grace's Insect Medium (Grace, T.C.,
  • an immobilized additive such as 10% pepsin serum or the like is used as needed.
  • the pH of the medium is adjusted to about 6, 2 to 6.4.
  • Culture is usually performed at about 27 ° C for about 3 to 5 days, and aeration and agitation are added as necessary.
  • the medium When culturing a transformant in which the host is an animal cell, the medium may be, for example, a MEM medium containing about 5 to 20% fetal bovine serum [Science, 122,
  • the pH is about 6-8.
  • Culture is usually about 30-40. Perform at C for about 15-60 hours, adding aeration and agitation as needed.
  • the protein of the present invention can be produced in cells, cell membranes or extracellular 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, lysozyme and Z or freezing. After the cells or cells are disrupted by thawing or the like, a method of obtaining a crude protein extract by centrifugation or filtration is appropriately used.
  • the buffer may contain a protein denaturant such as urea or guanidine hydrochloride, or a surfactant such as Triton X-100 TM . If the protein is secreted into the culture, after the culture is completed, the supernatant is separated from the cells or cells by a known method, and the supernatant is collected.
  • the protein contained in the culture supernatant or the extract obtained in this manner can be purified by appropriately combining known separation and purification methods.
  • known separation and purification methods include methods utilizing solubility such as salting out and solvent precipitation, dialysis, ultrafiltration, gel filtration, and SDS-polyacrylamide gel electrophoresis.
  • a method utilizing a difference in hydrophobicity, a method utilizing a difference in isoelectric point such as isoelectric focusing, and the like are used.
  • the salt can be converted into a salt by a method or a method analogous thereto, and when the salt is obtained, it can be converted into a free form or another salt by a known method or a method analogous thereto.
  • the protein produced by the recombinant can be arbitrarily modified or the polypeptide can be partially removed by applying an appropriate protein modifying enzyme before or after purification.
  • an appropriate protein modifying enzyme for example, trypsin, chymotrypsin, arginyl endopeptidase, proteinase, glycosidase and the like are used.
  • the presence of the protein of the present invention thus produced can be measured by enzyme immunoassay using a specific antibody, Western plotting, or the like.
  • An antibody against the protein or partial peptide of the present invention or a salt thereof may be any of a polyclonal antibody and a monoclonal mononal antibody as long as it can recognize the protein or partial peptide of the present invention or a salt thereof.
  • an antibody against the protein or partial peptide of the present invention or a salt thereof uses the protein of the present invention as an antigen, and is a known antibody. It can be produced according to a method for producing an antibody or antiserum.
  • the protein of the present invention is administered to a warm-blooded animal at a site capable of producing an antibody upon administration, itself or together with a carrier or diluent.
  • Complete Freund's adjuvant or incomplete Freund's adjuvant may be administered in order to enhance antibody production upon administration.
  • the administration is usually performed once every 2 to 6 weeks, for a total of about 2 to 10 times.
  • Examples of the warm-blooded animal used include monkeys, rabbits, rabbits, dogs, guinea pigs, mice, rats, sheep, goats, and chickens, and mice and rats are preferably used.
  • a monoclonal antibody-producing hybridoma can be prepared by collecting fl cells or lymph nodes and fusing the antibody-producing cells contained therein with myeloma cells of the same or different species.
  • 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 carried out according to known methods, for example, the method of Köhler and Milstein (Nature, 256, 495 (1975)).
  • the fusion promoter include polyethylene glycol (PEG) and Sendai virus, and PEG is preferably used.
  • myeloma cells examples include myeloma cells of warm-blooded animals such as NS-1, P3U1, SP 2/0, and AP-1. P3U1 is preferably used.
  • ⁇ Antibody-producing cells used The preferred ratio of the number of spleen cells to the number of myeloma cells is about 1 ::! To 20: 1, and PEG (preferably PEG 1000 to PEG6 000) is added at a concentration of about 10 to 80%. Incubation at 40, preferably 30 to 37 ° C for 1 to 10 minutes allows efficient cell fusion.
  • a hybridoma culture supernatant is added to a solid phase (eg, microplate) on which a protein antigen is adsorbed directly or together with a carrier, and then An 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 is added to the monoclonal antibody bound to the solid phase.
  • a radioactive substance or enzyme anti-mouse immunoglobulin antibody is used if the cells used for cell fusion are mice
  • protein A is added to the monoclonal antibody bound to the solid phase.
  • Anti-immunoglobulin antibody or protein A is adsorbed on a solid phase to which a hybridoma culture supernatant is added, and a protein labeled with a radioactive substance or an enzyme is added, and a monoclonal antibody bound to the solid phase is detected. And the like.
  • the selection of the monoclonal antibody can be performed according to a known method or a method analogous thereto. Usually, it can be performed in a medium for animal cells supplemented with HAT (hypoxanthine, aminopterin, thymidine).
  • HAT hyperxanthine, aminopterin, thymidine
  • any medium can be used as long as it can grow a hybridoma.
  • RPMI 1640 medium containing 1 to 20%, preferably 10 to 20% fetal bovine serum, and GIT medium containing 1 to 10% fetal bovine serum (Wako Pure Chemical Industries, Ltd.)
  • a serum-free culture medium for hybridoma culture SFM-101, Nissui Pharmaceutical Co., Ltd.
  • the cultivation temperature is usually from 20 to 40 ° (:, preferably, about 37 ° C.
  • the culturing time is usually from 5 days to 3 weeks, preferably from 1 week to 2 weeks.
  • the antibody titer of the hybridoma culture supernatant can be measured in the same manner as the measurement of the antibody titer in the antiserum described above.
  • Monoclonal antibodies can be separated and purified by known methods, for example, immunoglobulin separation and purification methods (eg, salting out method, alcohol precipitation method, isoelectric point precipitation method, electrophoresis method, ion exchanger (eg, DEAE)).
  • immunoglobulin separation and purification methods eg, salting out method, alcohol precipitation method, isoelectric point precipitation method, electrophoresis method, ion exchanger (eg, DEAE)
  • an active adsorbent such as protein A or protein G and the bond is dissociated to obtain the antibody
  • the polyclonal body of the present invention can be manufactured according to a known method or a method analogous thereto.
  • a immunizing antigen protein antigen
  • a warm-blooded animal is immunized in the same manner as in the above-described method for producing a monoclonal antibody. It can be produced by collecting an antibody-containing substance and separating and purifying the antibody.
  • the type of carrier protein and the mixing ratio between carrier-1 and hapten are determined by the antibody against hapten immunized by cross-linking with carrier. As long as it can be efficiently carried out, any kind may be cross-linked at any ratio.
  • serum serum albumin, psiloglopurine, hemocyanin, etc. are used in a weight ratio of about 0.1 to 1 for hapten.
  • a method of pulling at a rate of about 20 to 20 and preferably about 1 to 5 is used.
  • Glutaraldehyde, carbodiimide, maleimide active ester, an active ester reagent containing a thiol group or a dithioviridyl group, etc. are used.
  • the condensation product is administered to a warm-blooded animal at a site where antibody production is possible or together with a carrier or diluent.
  • Complete Freund's adjuvant or incomplete Freund's adjuvant may be administered to increase antibody production ability during administration.
  • administeristration 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 the blood, ascites, etc., preferably from the blood of a warm-blooded animal immunized by the above method.
  • the measurement of the polyclonal antibody titer in the antiserum can be performed in the same manner as the measurement of the antibody titer in the antiserum described above. Separation and purification of polyclonal antibodies
  • the antisense polynucleotide having a substantially complementary base sequence or a part thereof includes a base sequence complementary to or substantially complementary to the base sequence of the DNA of the present invention or a part thereof, Any antisense polynucleotide may be used as long as it has an action of suppressing the expression of the DNA, but antisense DNA is preferable.
  • the nucleotide sequence substantially complementary to the DNA of the present invention is, for example, the entire nucleotide sequence or a part of the nucleotide sequence complementary to the DNA of the present invention (that is, the complementary strand of the DNA of the present invention).
  • a base sequence having about 70% or more, preferably about 80% or more, more preferably about 90% or more, and most preferably about 95% or more homology with the base sequence is exemplified.
  • the complementary sequence of the nucleotide sequence of the portion encoding the N-terminal portion of the protein of the present invention is approximately 70%.
  • Antisense polynucleotides having a homology of at least about 80%, preferably at least about 80%, more preferably at least about 90%, and most preferably at least about 95% are suitable. Specifically, a nucleotide sequence complementary to or substantially complementary to the nucleotide sequence of DNA having the nucleotide sequence represented by SEQ ID NO: 2, SEQ ID NO: 18 or SEQ ID NO: 20, or a part thereof An antisense polynucleotide having, preferably, a nucleotide sequence complementary to the nucleotide sequence of DNA having the nucleotide sequence represented by SEQ ID NO: 2, SEQ ID NO: 18 or SEQ ID NO: 20, or a part thereof Antisense polynucleotide (more preferably, a nucleotide sequence complementary to the nucleotide sequence of DNA having the nucleotide sequence represented by SEQ ID NO: 2, SEQ ID NO: 18 or SEQ ID NO: 20, or a portion thereof. Antisense polynu
  • the number of antisense polynucleotides is usually about 10 to 40, preferably 15 to
  • the phosphate residues (phosphates) of each nucleotide constituting the antisense DNA should be chemically modified, for example, phosphorothioate, methylphosphonate, and phosphorodithionate. It may be substituted with a phosphoric acid residue.
  • phosphate residues phosphates
  • These antisense polynucleotides can be produced using a known DNA synthesizer or the like.
  • an antisense polynucleotide (nucleic acid) corresponding to the protein gene of the present invention, which can inhibit the replication or expression of the gene, has been cloned or encoded. It can be designed and synthesized based on DNA base sequence information. Such antisense polynucleotides can hybridize with the RNA of the protein gene of the present invention, inhibit the synthesis or function of the RNA, or interact with the protein-related RNA of the present invention. Thus, the expression of the protein gene of the present invention can be regulated and controlled.
  • Polynucleotides that are complementary to a 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, are those of the present invention in vivo and in vitro. It is useful for regulating and controlling the expression of protein genes, and is also useful for treating or diagnosing diseases and the like.
  • corresponding refers to a nucleotide, base sequence or specific sequence of a nucleic acid, including or homologous to a gene, Means that The "correspondence" between a nucleotide, nucleotide sequence or nucleic acid and a protein usually refers to the amino acids of the protein (as specified by the instructions) derived from the nucleotide (nucleic acid) sequence or its complement.
  • 5 'end hairpin loop of protein gene 5' end 6—base pair repeat, 5 'end untranslated region, polypeptide translation start codon, protein coding region, ORF translation stop codon, 3' end untranslated region, A 3'-end palindrome region or a 3'-end hairpin loop may be selected as a preferred region of interest, but any region within a protein gene may be selected as a target.
  • Antisense polynucleotides are polynucleotides containing 2-dexoxy-D-reports, polynucleotides containing D-ribose, or other types of polynucleotides that are N-glycosides of purine or pyrimidine bases.
  • polymers with non-nucleotide backbones eg, commercially available protein nucleic acids and synthetic sequence-specific nucleic acid polymers
  • polymers containing special bonds provided that the polymer is not contained in DNA or RNA
  • Pairing of bases as found contains nucleotides having a configuration that allows base attachment
  • They may be double-stranded DNA, single-stranded DNA, double-stranded RNA, single-stranded RNA, DNA: RNA hybrid, and may further comprise unmodified polynucleotides (or unmodified oligonucleotides).
  • nucleoside may include not only those containing purine and pyrimidine bases but also those having other modified heterocyclic bases. Such modifications may include methylated purines and pyrimidines, acylated purines and pyrimidines, or other heterocycles. Modified nucleotides and modified nucleotides may also be modified at the sugar moiety, e.g., where one or more hydroxyl groups have been replaced by 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 an RNA, a DNA or a modified nucleic acid (RNA, DNA).
  • modified nucleic acid include a sulfur derivative of a nucleic acid, a thiophosphate derivative, and a polynucleoside amide / polynucleoside amide that is resistant to degradation of oligonucleoside amide.
  • the antisense polynucleotide of the present invention can be designed, for example, as follows.
  • the antisense polynucleotide makes the antisense polynucleotide more stable in the cell, enhances the cell permeability of the antisense polynucleotide, increases the affinity for the target sense strand, and In some cases, the toxicity of the antisense polynucleotide is reduced. Many such modifications have been reported, for example, in Pharm Tech Japan, vol. 8, p. 247 or p. 395, 1992, Antisense Research and Appli cations, CRC Press, 1993.
  • the antisense polynucleotides of the present invention may contain altered or modified sugars, bases, or bonds, are provided in special forms such as ribosomes, microspheres, are applied by gene therapy, It can be provided in an added form.
  • additional forms include polycations such as polylysine, which acts to neutralize the charge on the phosphate backbone, and lipids, which enhance the interaction with cell membranes and increase the uptake of nucleic acids ( For example, Hydrophobic substances such as phospholipid and cholesterol).
  • Preferred lipids for addition include cholesterol and its derivatives (eg, cholesteryl chloroformate, cholic acid, etc.).
  • nucleic acids can be attached via bases, sugars, intramolecular nucleoside bonds.
  • Other groups include capping groups specifically located at the 3, 5 'or 5' end of nucleic acids to prevent degradation by nucleases such as exonucleases and RNases .
  • capping groups include, but are not limited to, hydroxyl-protecting groups known in the art, including glycols such as polyethylene glycol and tetraethylene glycol. .
  • the inhibitory activity of the antisense polynucleotide can be examined using the transformant of the present invention, the in vivo or in vitro gene expression system of the present invention, or the in vivo or in vitro translation system of the protein of the present invention. .
  • the protein or partial peptide of the present invention or a salt thereof hereinafter, sometimes abbreviated as the protein of the present invention
  • the DNA encoding the protein or partial peptide of the present invention hereinafter, referred to as the DNA of the present invention
  • an antibody against the protein or partial peptide of the present invention or a salt thereof hereinafter may be abbreviated as the antibody of the present invention
  • an antisense polynucleotide of the DNA of the present invention hereinafter referred to as the present invention. May be abbreviated as an antisense polynucleotide).
  • the protein of the present invention can be used as a disease marker because its expression is increased in a tissue-specific manner in chronic obstructive pulmonary disease and rhinitis. Therefore, the protein of the present invention may be used, for example, for respiratory diseases such as pulmonary thoracic disease accompanied by inflammation of the lungs and airways [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, Bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), allergic conjunctivitis, rhinitis
  • respiratory diseases such as pulmonary thoracic disease accompanied by inflammation of the lungs and airways
  • chronic obstructive pulmonary disease chronic bronchitis, emphysema
  • diffuse panbronchiolitis Bronchial asthma, cystic fibrosis, irritable pneumonia, etc.
  • allergic conjunctivitis rhinitis
  • gastrointestinal diseases eg, Irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease, reflux It is useful as a marker for early diagnosis of esophagitis, etc.), judgment of symptom severity, and prediction of disease progression.
  • Antisense polynucleotide of a gene encoding the protein of the present invention (protein gene of the present invention), a compound or a salt thereof that inhibits the activity of the protein of the present invention, a compound that inhibits the expression of the gene of the protein of the present invention, or Pharmaceuticals containing a salt thereof or an antibody against the protein of the present invention include, for example, respiratory diseases such as lungs, lungs accompanied by inflammation of the respiratory tract, and chest diseases [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema) , Diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), allergic conjunctivitis, rhinitis (eg, allergic rhinitis, pollinosis, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis , Dry rhinitis, vasomotor
  • the protein of the present invention and the gene of the present invention have an increased tissue-specific expression in chronic obstructive pulmonary disease and rhinitis, and have a mucus production promoting action and an alveolar wall destruction promoting action in the lung, airway or nasal mucosa.
  • the compound or its salt that inhibits the activity of the protein of the present invention and the compound or its salt that inhibits the expression of the protein gene of the present invention can be used, for example, for respiratory diseases such as lung-thoracic diseases accompanied by inflammation of the lungs and airways.
  • Respiratory tract disease eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.
  • allergic conjunctivitis rhinitis (eg, allergy) Rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc.)
  • gastrointestinal diseases eg, irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease, reflux esophagitis, etc.
  • Therapeutic agents preferably prevention of respiratory diseases, gastrointestinal diseases, etc.
  • the protein of the present invention inhibits the activity of the protein of the present invention.
  • the polynucleotide encoding the protein is useful as a reagent for screening a compound or a salt thereof, and the polynucleotide encoding the protein is useful as a reagent for screening a compound or a salt thereof that inhibits expression of the protein gene of the present invention.
  • the present invention provides (1) a compound that inhibits the activity of the protein of the present invention (eg, chloride channel activity and the like) characterized by using the protein of the present invention, or a salt thereof (hereinafter, abbreviated as an inhibitor).
  • a compound that inhibits expression of the protein gene of the present invention which comprises using a polynucleotide encoding the protein of the present invention, or a salt thereof.
  • an inhibitor a compound or a salt thereof that inhibits the expression of the protein of the present invention characterized by using the antibody of the present invention (hereinafter referred to as an inhibitor) (May be abbreviated as).
  • screening method examples include: (i) the case where cells having the ability to produce the protein of the present invention are activated with a calcium activator; and (ii) the case where cells having the ability to produce the protein of the present invention are tested.
  • a method for screening for an inhibitor which is characterized by comparing with a case where a mixture of compounds is activated with a calcium activator, is mentioned.
  • the chloride channel activity of the protein of the present invention in the cases (0 and (ii)) is measured and compared.
  • the calcium activator may be added to cells having the ability to produce the protein of the present invention after being mixed with the test compound, or the calcium activator may be added to cells having the ability to produce the protein of the present invention. After that, the test compound may be added.
  • the cells having the ability to produce the protein of the present invention and the test compound may be mixed before or after the cells having the ability to produce the protein of the present invention are activated with a calcium activator.
  • a mixture of a test compound and a calcium activator may be added to cells having the ability to produce the protein of the present invention.
  • a calcium activator ionomycin, A2187 (calcimycin) and the like are used.
  • the chloride channel activity of the protein of the present invention can be determined according to a known method, for example, the method described in Genomics, vol. 54, p. 200 (1998) or a method analogous thereto.
  • Test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, and animal tissue extracts, and these compounds are novel compounds. Or a known compound.
  • cells having the ability to produce the protein of the present invention are prepared by suspending them in a buffer suitable for screening.
  • the buffer may be any one of phosphate buffer and borate buffer having a pH of about 4 to 10 (preferably, a pH of about 6 to 8).
  • the screening method include (iii) culturing cells capable of producing the protein of the present invention and (iv) culturing a mixture of cells capable of producing the protein of the present invention and a test compound. Then, the expression level of the protein gene of the present invention (specifically, the amount of the protein of the present invention or the amount of mRNA encoding the protein) is measured and compared.
  • the amount of the protein of the present invention can be measured by a known method, for example, using an antibody recognizing the protein of the present invention to detect the protein present in a cell extract or the like. It can be measured in accordance with the corresponding method.
  • the expression level of the protein gene of the present invention can be determined by a known method, for example, Northern blotting, reverse transcription-polymerase chain reaction (RT-PCR), real-time PCR analysis system (manufactured by ABI, TaqMan polymerase chain).
  • RT-PCR reverse transcription-polymerase chain reaction
  • ABI TaqMan polymerase chain
  • Test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, and animal tissue extracts, and these compounds are novel compounds. Or a known compound.
  • the buffer may be a buffer that does not inhibit the chloride channel activity of the protein of the present invention, such as a phosphate buffer having a pH of about 4 to 10 (preferably, a pH of about 6 to 8) or a borate buffer. Either may be used.
  • the screening method include ( ⁇ ) ′ when a cell capable of producing the protein of the present invention is cultured and (vi) when a mixture of a cell capable of producing the protein of the present invention and a test compound is cultured. Make a comparison with the case.
  • the expression level of the protein of the present invention (specifically, the amount of the protein of the present invention) in the cases (V) and (vi) is measured using the antibody of the present invention. (Eg, detection of expression, quantification of expression level, etc.) and compare.
  • the amount of the protein of the present invention can be measured by a known method, for example, using an antibody recognizing the protein of the present invention to detect the protein present in a cell extract or the like. It can be measured in accordance with the corresponding method.
  • Test compounds include, for example, peptides, tanks, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, etc., and these compounds are new compounds. Or a known compound.
  • a host transformed with a vector containing a DNA encoding the protein of the present invention described above is used.
  • the host for example, animal cells such as CHO cells are preferably used.
  • a transformant in which the protein of the present invention is expressed on a cell membrane by culturing by the above-mentioned method is preferably used.
  • a test compound that inhibits chloride channel activity in the case of the above (ii) by about 20% or more, preferably 30% or more, and more preferably about 50% or more compared to the case of the above (i) can be selected as a compound that inhibits the activity of the protein of the present invention or a salt thereof.
  • the expression level of the protein gene of the present invention in the case (iv) is about 20% or more, preferably 30% or more, more preferably about 50%, as compared with the case (iii).
  • the test compound that inhibits the above can be selected as a compound that inhibits the expression of the protein gene of the present invention or a salt thereof.
  • test compound that inhibits about 20% or more, preferably 30% or more, more preferably about 50% or more of that in the case of (V) as a compound that inhibits expression of the protein of the present invention or a salt thereof. You can choose.
  • the screening kit of the present invention includes a protein or a partial peptide of the present invention or a salt thereof, a polypeptide encoding a protein or a partial peptide of the present invention, an antibody of the present invention, or a protein or partial base of the present invention. It contains cells that have the ability to produce peptides.
  • Compounds or salts thereof obtained by using the screening method or the screening kit of the present invention include the test compounds described above, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, and plant extracts. Liquid, animal tissue extract, plasma, or a compound thereof, or a salt thereof, wherein the activity of the protein of the present invention (eg, chloride channel activity, etc.), the expression of the protein gene of the present invention, or A compound or a salt thereof that inhibits expression of the protein of the invention.
  • the activity of the protein of the present invention eg, chloride channel activity, etc.
  • salt of the compound those similar to the aforementioned salts of the protein of the present invention are used. '
  • Compounds or salts thereof obtained using the screening method or screening kit of the present invention include, for example, respiratory diseases such as lungs, lungs with inflammation of the airways, and chest diseases [eg, chronic obstructive pulmonary diseases (chronic bronchial Inflammation, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), allergic conjunctivitis, rhinitis (eg, allergic rhinitis, pollinosis, acute rhinitis, chronic rhinitis, hypertrophic) Rhinitis, atrophic rhinitis, dry rhinitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc., gastrointestinal diseases (eg, irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease, Prophylactic and therapeutic agents for reflux esophagitis, etc., preferably respiratory disease,
  • the compound or a salt thereof obtained by using the screening method or the screening kit of the present invention can be safely administered by itself or as a suitable drug.
  • the medicament used for the above-mentioned administration contains the above-mentioned compound or a salt thereof and a pharmacologically acceptable carrier, diluent or vehicle, and is provided as a pharmaceutical composition suitable for oral or parenteral administration. Is done.
  • compositions for oral administration include solid or liquid dosage forms, specifically tablets (including sugar-coated tablets and film-coated tablets), pills, granules, powders, and capsules (including soft capsules). 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 pharmaceutical field. For example, lactose, starch, sucrose, magnesium stearate and the like are used as carriers and excipients for tablets.
  • compositions for parenteral administration for example, injections, suppositories, etc. are used.
  • Injections are in the form of intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections, infusions, etc. Is included.
  • Such injections are prepared according to a known method, for example, by dissolving, suspending or emulsifying the above antibody or a salt thereof in a sterile aqueous or oily liquid usually used for injections.
  • aqueous liquid for injection for example, physiological saline, isotonic solution containing glucose and other adjuvants, and the like, suitable solubilizing agents, for example, alcohol (eg, ethanol), polyalcohol (eg, Propylene glycol, polyethylene glycol), a nonionic surfactant [eg, polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated catalyst)], and the like.
  • alcohol eg, ethanol
  • polyalcohol eg, Propylene glycol, polyethylene glycol
  • a nonionic surfactant 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 into a suitable ampoule.
  • 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 form examples include tablets, pills, capsules, injections (ampoules), suppositories, etc., usually 5 to 500 mg per dosage unit, especially 5 to 100 mg for injections, and other dosage forms. Preferably, it contains 10-250 mg of the above antibody.
  • compositions for nasal administration include inhalants, nasal drops, aerosols and the like.
  • the preparation obtained by a conventional method is used as an inhaler, it is converted into a powder inhaler, a suspension for inhalation, a solution for inhalation or a capsule inhaler using a known method, and an appropriate inhaler for use.
  • powder inhalants are particularly preferably used.
  • the average particle size of the powder inhalant is not particularly limited, but is preferably about 0.1 to 20 im, and particularly preferably about 1 to 5 m.
  • the particle size of the powder inhaler is not particularly limited, but the amount of particles having a size of about 25 m or more is preferably about 5% or less, particularly preferably about 1% or less.
  • ventolin / rotacaps VENTOLIN
  • the preparations obtained in this way are safe and low toxic and can be used, for example, in humans or in warm-blooded animals (eg, mice, rats, puppies, higgs, bushus, puppies, puppies, birds, cats, dogs). , Monkeys, chimpanzees, etc.) orally or parenterally.
  • warm-blooded animals eg, mice, rats, puppies, higgs, bushus, puppies, puppies, birds, cats, dogs.
  • Monkeys, chimpanzees, etc. orally or parenterally.
  • the dose of the compound or its salt that inhibits the activity of the protein of the present invention may vary depending on its action, target disease, subject to be administered, administration route and the like.
  • administration route and the like for example, for the purpose of treating chronic obstructive pulmonary disease.
  • the compound or a salt thereof is administered orally, in general, for an adult (assuming a body weight of 60 kg), about 0.1 to 100 mg, preferably about 1.0 to 50 mg, of the compound or its salt per day is used. More preferably, about 1.0 to 20 mg is administered.
  • the single dose of the above compound or a salt thereof varies depending on the administration subject, target disease, etc., for example, the activity of the protein of the present invention for the purpose of treating chronic obstructive pulmonary disease.
  • the compound or a salt thereof When a compound or a salt thereof is administered to an adult (with a body weight of 60 kg) usually in the form of an injection, the compound or a salt thereof is about 0.01 to 30 mg, preferably about 0.1 to 20 mg per day. Degree, more preferably about
  • 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, and therefore, the quantification of the protein of the present invention in a test solution.
  • the antibody of the present invention can be used for quantification by sandwich immunoassay.
  • a method for quantifying the protein of the present invention in a test solution which comprises:
  • the protein of the present invention can be quantified using a monoclonal antibody against the protein of the present invention (hereinafter sometimes referred to as the monoclonal antibody of the present invention), and can also be detected by tissue staining or the like.
  • the antibody molecule itself may be used, or the FCab ') 2 , Fab ⁇ 5 or Fab fraction of the antibody molecule may be used.
  • the method for quantifying the protein of the present invention using the antibody of the present invention is not particularly limited, and may be an antibody, an antigen, or an antibody-antigen complex corresponding to the amount of antigen (eg, the amount of protein) in the test solution. Any measurement method may be used as long as the amount is detected by chemical or physical means and the amount is measured 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.
  • Examples of the labeling agent used in the assay method using the labeling substance for example, a radioactive isotope (e.g., [125 1], [131 1], [3 ⁇ 4], etc. [14 c]), fluorescent substances [e.g., Shianin 5 Fluorescent dyes (eg, Cy2, Cy3, Cy5, Cy5.5, Cy7 (manufactured by Amersham Biosciences), etc.), fluorescamine, fluorescein sotiocinet, etc.), enzymes (eg, ⁇ -galactosidase, ⁇ -darcosidase, alpholipophorase, peroxidase, malate dehydrogenase, etc., luminescent substances (eg, luminol, luminol derivatives, reciferin, lucigenin, etc.), piotin, lanthanide, etc.
  • a biotin-avidin system can be used for binding the antibody or antigen to the labeling agent.
  • the carrier include insoluble polysaccharides such as agarose, dextran, and cellulose; synthetic resins such as polystyrene, polyacrylamide, and silicon; and glass.
  • 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). Measuring the activity of the above labeling agent Thus, the mass of the protein of the present invention in the test solution can be determined.
  • the primary reaction and the secondary reaction may be performed in the reverse order, may be performed simultaneously, or may be performed at staggered times.
  • the labeling agent and the method of insolubilization can be the same as those described above.
  • the antibody used for the solid phase antibody or the labeling antibody is not necessarily required to be one type, and two or more types of antibodies are used for the purpose of improving measurement sensitivity and the like. Mixtures may be used.
  • the monoclonal antibody of the present invention used in the primary reaction and the secondary reaction differs in the binding site of the protein of the present invention.
  • Antibodies are preferably used. That is, when the antibody used in the primary reaction and the secondary reaction is, for example, 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 sample 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.
  • the competition method after the antigen in the test wave and the labeled antigen are allowed to react competitively with the antibody, the unreacted labeled antigen (F) is separated from the labeled antigen (B) bound to the antibody ( B / F separation), measure the amount of labeling for either B or F, and quantify the amount of antigen in the test solution.
  • a soluble antibody is used as an antibody
  • B / F separation is performed using polyethylene glycol
  • a liquid phase method using a second antibody against the antibody a solid phase antibody is used as the first antibody
  • An immobilization method using a soluble antibody as the first antibody and an immobilized antibody as the second antibody is used.
  • the antigen in the test wave 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 original and an excess amount of the labeled antibody are allowed to react, 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.
  • the amount of label in either phase is measured to quantify the amount of antigen in the test solution.
  • the amount of insoluble sediment generated as a result of an antigen-antibody reaction in a gel or in a solution is measured.
  • the amount of antigen in the test solution is small, Even when only a sediment is obtained, a laser nephrometry utilizing scattering by a laser 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 documents.
  • the protein of the present invention can be quantified with high sensitivity by using the antibody of the present invention.
  • lung / thoracic disease accompanied by inflammation of the lung airways
  • Respiratory diseases eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.
  • allergic conjunctivitis rhinitis (eg, Allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc.
  • gastrointestinal diseases eg, irritable bowel
  • flame Eg, inflammatory bowel disease, ulcerative colitis, Crohn's disease, reflux
  • 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.
  • preparation of an antibody column used for purifying the protein of the present invention, detection of the protein of the present invention in each fraction at the time of purification, analysis of the behavior of the protein of the present invention in test cells, etc. Can be used for
  • the DNA of the present invention can be used, for example, as a probe to produce human or warm-blooded animals (eg, rats, mice, guinea pigs, egrets, birds, higgies, pigs, pigs, dogs, cats, cats). (DNA, monkey, chimpanzee, etc.) can detect an abnormality (gene abnormality) in DNA or mRNA that encodes the protein of the present invention or a partial peptide thereof. Mutations or decreased expression or increases in the DNA or mRNA are useful as diagnostic agents for gene expression such as overexpression.
  • human or warm-blooded animals eg, rats, mice, guinea pigs, egrets, birds, higgies, pigs, pigs, dogs, cats, cats.
  • an abnormality gene abnormality
  • Mutations or decreased expression or increases in the DNA or mRNA are useful as diagnostic agents for gene expression such as overexpression.
  • the above-described genetic diagnosis using the DNA of the present invention includes, for example, the well-known Northern hybridization and PCR-SSCP method (Genomics, Vol. 5, pp. 874-879 (1989), Proc. Proceedings of the National Academy
  • Respiratory diseases such as chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, etc., allergic conjunctivitis, rhinitis (eg , Allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc., gastrointestinal diseases (eg, hypersensitivity) Bowel syndrome, It can be diagnosed as being highly likely to be a disease such as inflammatory bowel disease, ulcerative colitis, Crohn's disease, or reflux esoph
  • the antisense polynucleotide of the present invention which can complementarily bind to the DNA of the present invention and suppresses the expression of the DNA, has low toxicity and has the activity of the protein of the present invention or the DNA of the present invention in vivo.
  • ⁇ Functions eg, chloride channel activity, etc.
  • lungs ⁇ lungs with inflammation of the airways ⁇ respiratory diseases such as chest diseases [eg, chronic obstructive pulmonary disease '(chronic bronchitis, Pulmonary emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), alegiitis conjunctivitis, rhinitis (eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic) Rhinitis, atrophic rhinitis, dry rhinitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc., gastrointestinal diseases (eg, irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease) Reflux esophagitis, etc) agent for the prophy
  • chest diseases e
  • the antisense polynucleotide When used as the prophylactic or therapeutic agent, it can be formulated and administered according to a known method.
  • the antisense polynucleotide when used, the antisense polynucleotide may be used alone or in a suitable vector such as a retrovirus vector, an adenovirus vector, an adenovirus associated virus vector, or the like.
  • a suitable vector such as a retrovirus vector, an adenovirus vector, an adenovirus associated virus vector, or the like.
  • the antisense polynucleotide can be administered as it is, or can be formulated with a physiologically acceptable carrier such as an adjuvant to promote uptake, and administered with a gene gun or a catheter such as a hide mouth gel catheter. Alternatively, they can be aerosolized and administered topically into the trachea as an inhalant.
  • a physiologically acceptable carrier such as an adjuvant to promote uptake
  • a gene gun or a catheter such as a hide mouth gel catheter.
  • they can be aerosolized and administered topically into the trachea as an inhalant.
  • the dosage of the antisense polynucleotide may vary depending on the target disease, the target of administration, the route of administration, and the like.
  • the present invention relates to the treatment of chronic obstructive pulmonary disease.
  • the antisense polynucleotide is locally administered into the trachea as an inhalant, generally, in an adult (body weight 60 kg), about 0.1 to 100 mg of the antisense polynucleotide is administered per day.
  • the antisense polynucleotide can also be used as a diagnostic oligonucleotide probe for examining the presence or the expression status of the DNA of the present invention in tissues or cells.
  • the present invention further provides
  • RNAi double-stranded RNAs
  • lipozymes and the like can suppress the expression of the polynucleotide (eg, DNA) of the present invention in the same manner as the above-mentioned antisense polynucleotides.
  • Chest disease eg, chronic obstructive pulmonary disease (chro
  • the double-stranded RNA can be designed and manufactured 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 produced 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 RNA encoding the peptide of the present invention.
  • RNA fragment As a part of the RNA encoding the peptide of the present invention, a portion (RNA fragment) close to the cleavage site on the RNA of the present invention, which can be cleaved by a known lipozyme, can be mentioned.
  • RNA fragment RNA fragment close to the cleavage site on the RNA of the present invention, which can be cleaved by a known lipozyme.
  • the above-mentioned double-stranded RNA or lipozyme is used as the above-mentioned prophylactic / therapeutic agent, it can be formulated and administered in the same manner as for the antisense polynucleotide.
  • Antibodies of the present invention having an activity of neutralizing the activity of the protein of the present invention include, for example, respiratory diseases such as lung and chest diseases accompanied by inflammation of the lungs and airways [eg, chronic obstructive pulmonary disease (chronic bronchitis, Emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), allergic conjunctivitis, rhinitis (eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, Atrophic rhinitis, dry rhinitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc., gastrointestinal diseases (eg, irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease, reflux disease) It can be used as a medicine for diseases such as esophagitis.
  • respiratory diseases
  • the prophylactic / therapeutic agent for the above-mentioned diseases containing the antibody of the present invention has low toxicity, and can be used directly as a liquid or as a pharmaceutical composition of an appropriate dosage form in human or non-human mammals (eg, rat, egret, hidge) Or parenteral administration to mice, dogs, 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 of chronic obstructive pulmonary disease in adults, the antibody of the present invention is used as a single dose.
  • about 0.01 to 20 mg / kg body weight preferably about 0.1 to 10 mg / kg body weight, more preferably about 0.1 to 5 rag / kg body weight, about 1 to 5 times a day, preferably about 1 to 5 rag / kg body weight. It is convenient to administer by intravenous injection about once to three times a day. In the case of other parenteral administration and oral administration, an equivalent dose can be administered. If the symptoms are particularly severe, the dose may be increased accordingly.
  • the antibodies of the present invention can be administered by themselves or as a suitable pharmaceutical composition.
  • the pharmaceutical composition used for the administration contains the antibody or a salt thereof and a pharmacologically acceptable carrier, diluent or excipient.
  • Such compositions are provided in dosage forms suitable for oral or parenteral administration.
  • a solid or liquid agent examples include tablets (including sugar-coated tablets and film-coated tablets), pills, granules, powders, capsules (including soft capsules), syrups, emulsions, and suspensions.
  • Such a composition is produced by a known method and contains a carrier, diluent or excipient commonly used in the pharmaceutical field.
  • a carrier for example, lactose, starch, sucrose, magnesium stearate and the like are used as carriers and excipients for tablets.
  • compositions for parenteral administration for example, injections, suppositories, etc. are used.
  • Injections are in the form of intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections, infusions, etc. Is included.
  • Such injections are prepared according to a known method, for example, by dissolving, suspending or emulsifying the above antibody or a salt thereof in a sterile aqueous or oily liquid usually used for injections.
  • aqueous liquid for injection for example, physiological saline, isotonic solution containing glucose and other adjuvants, and the like, suitable solubilizing agents, for example, alcohol (eg, ethanol), polyalcohol (eg, It may be used in combination with propylene glycol, polyethylene glycol), a nonionic surfactant [eg, polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated catalyst)], and the like.
  • suitable solubilizing agents for example, alcohol (eg, ethanol), polyalcohol (eg, It may be used in combination with propylene glycol, polyethylene glycol), a nonionic surfactant [eg, polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated catalyst)], and the like.
  • oily liquid for example, sesame oil, soybean oil, and the like are used, and benzyl benzoate, benzyl alcohol, and the like may be
  • 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 forms include tablets, pills, capsules, injections (ampoules), and suppositories, and usually 5 to 500 mg per dosage unit form, and especially 5 to 500 mg for injections.
  • 100 mg and other dosage forms contain 10 to 250 mg of the above antibody.
  • compositions may contain other active ingredients as long as the composition does not cause an undesirable interaction with the above-mentioned antibody.
  • a drug containing the protein or DNA of the present invention may contain other active ingredients as long as the composition does not cause an undesirable interaction with the above-mentioned antibody.
  • the protein (preferably a partial peptide) of the present invention may be used, for example, for respiratory diseases such as lungs, lungs with respiratory tract inflammation, and chest diseases [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse pan- Bronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), allergic conjunctivitis, rhinitis (eg, allergic rhinitis, pollinosis, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dryness Pronasalitis, vasomotor rhinitis, gangrenous rhinitis, sinusitis, etc., gastrointestinal disorders (eg, irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease, reflux esophagitis, etc.) It is also useful as a prophylactic / therapeutic
  • the protein of the present invention can be used as a tablet, capsule, elixir, microcapsule, etc., if necessary, orally coated with water or other pharmaceutically acceptable liquids. It can be used parenterally in the form of injections, such as sterile solutions or suspensions.
  • the protein of the present invention may be used together with known physiologically acceptable carriers, flavoring agents, excipients, vehicles, preservatives, stabilizers, binders, etc., in a unit dosage form generally required for the practice of the formulation. It can be manufactured by mixing with. The amount of the active ingredient in these preparations is such that a suitable dosage in the specified range can be obtained.
  • the dose of the protein of the present invention varies depending on the administration subject, target organ, symptoms, administration method, and the like.
  • oral administration generally, for example, in a patient (as 60 kg), about 0. It is 1 to 100 mg, preferably about 1.0 to 50 mg, more preferably about 1.0 to 20 mg.
  • parenteral administration the single dose varies depending on the target of administration, target organ, symptoms, administration method, etc., for example, usually in the form of injection, for example, in patients (as 60 kg), It is convenient to administer about 0.01 to 30 mg, preferably about 0.1 to 20 mg, more preferably about 0.1 to 10 mg per day by intravenous injection. In the case of other animals, the dose can be administered in terms of 60 kg. [7] Creation of an animal having the DNA of the present invention
  • the present invention relates to a DNA encoding an 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).
  • Non-human mammals having the exogenous DNA of the present invention or the mutant DNA thereof can be used for non-fertilized eggs, fertilized eggs, spermatozoa and germ cells including their progenitor cells.
  • the DNA-transfected animal of the present invention can be used for non-fertilized eggs, fertilized eggs, spermatozoa and germ cells including their progenitor cells.
  • the calcium phosphate method, the electric pulse method, the ribofection method It can be created by introducing the desired DNA by agglutination, microinjection, particle gun, DEAE-dextran, etc.
  • exogenous DNA of the present invention can be introduced into somatic cells, organs of living organisms, tissue cells, and the like by the DNA introduction method, and used for cell culture, tissue culture, and the like. Can be fused with the above-mentioned germ cells by a known cell fusion method to produce the DNA-introduced animal of the present invention.
  • non-human mammal for example, porcupine, pig, sheep, goat, goat, egret, dog, cat, guinea pig, hamster, mouse, rat and the like are used.
  • rodents with relatively short ontogeny and biological cycle in terms of the creation of disease animal model systems and easy reproduction are particularly useful in mice (for example, pure strains such as C57BL / 6 strain and DBA2 strain).
  • crossing strain B6C3F, strain, BDF, strain, B6D2F, strain, BALB / c strain, ICR strain, etc.) or rat (eg, Wistar, SD, etc.) are preferable.
  • "Mammals" in a recombinant vector that can be expressed in mammals include humans 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 non-human mammals, but to the DNA of the present invention once isolated and extracted from the mammal.
  • the mutant DNA of the present invention is a DNA in which a mutation (for example, mutation) has occurred in the base sequence of the original DNA of the present invention, specifically, base addition, deletion, substitution with another base. DNA or the like that has occurred is used, and abnormal DNA is also included.
  • DNA that expresses an abnormal protein of the present invention is used.
  • DNA that expresses a protein that suppresses the function of the normal protein of the present invention is used.
  • the exogenous DNA of the present invention may be derived from a mammal that is the same or different from the animal of interest.
  • a promoter capable of being expressed in animal cells e.g., a promoter capable of being expressed in animal cells.
  • the human DNA of the present invention when introduced, it is derived from various mammals (eg, egrets, dogs, cats, guinea pigs, hamsters, rats, mice, etc.) having the DNA of the present invention that are highly homologous thereto.
  • a mammal into which the DNA of the present invention is highly expressed can be produced.
  • Examples of the expression vector of the protein of the present invention include a plasmid derived from Escherichia coli, a plasmid derived from Bacillus subtilis, a plasmid derived from yeast, a bacteriophage such as ⁇ phage, a retrovirus such as Moroni leukemia virus, a vaccinia virus or a baculovirus.
  • animal viruses such as E. coli are used.
  • a plasmid derived from E. coli, a plasmid derived from Bacillus subtilis or a plasmid derived from yeast are preferably used.
  • promoters that regulate DNA expression include, for example, viruses (eg, Simian virus, cytomegalovirus, Moroni leukemia virus, Promoters of DNA derived from JC virus, breast cancer virus, poliovirus, etc., promoters derived from various mammals (human, egret, dog, cat, guinea pig, hams, etc., rat, mouse, etc.), for example, albumin, Insulin II, peroplakin II, Erasuyose, erythropoietin, endothelin, muscle creatine kinase, glial fibrillary acidic protein, daltathione S_transferase, platelet-derived growth factor i3, keratin K 1,10 And 14, collagen type I and I 13 ⁇ 4, cyclic AMP-dependent protein kinase — 61 subunit, dystrophin, tartrate-resistant alkaline phosphatase, atrial natriuretic factor, endothelial
  • the vector preferably has a sequence that terminates the transcription of the messenger RNA of interest in the DNA-transfected mammal (generally called terminator).
  • terminator a sequence that terminates the transcription of the messenger RNA of interest in the DNA-transfected mammal.
  • terminator is derived from viruses and various mammals.
  • a sequence of each DNA can be used, and preferably, Simian virus SV40, Minne, etc. is used.
  • translation of splicing signal of each DNA, enhancer region, part of intron of eukaryotic DNA, etc. to 5 'upstream of promoter region, promoter region, etc. for the purpose of further expressing the target foreign DNA Inter-domain or translation It is also possible to connect 3 ′ downstream of the region depending on the purpose.
  • Complementary DNA prepared by known methods from cell-derived DNA and all or part of genomic DNA from one of various commercially available genomic DNA libraries, or from liver, kidney, thyroid cells, and fibroblast-derived RNA. It can be obtained as a raw material.
  • a foreign abnormal DNA can produce a translation region obtained by mutating a normal polypeptide translation region obtained from the above cells or tissues by a point mutagenesis method.
  • the translation region can be prepared as a DNA construct that can be expressed in a DNA-transduced 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.
  • exogenous DNA of the present invention at the fertilized egg cell stage is ensured to be present in all germ cells and somatic cells of the target mammal.
  • the presence of the exogenous DNA of the present invention in the germinal cells of the produced animal after the introduction of the DNA indicates that all the progeny of the produced animal retain the exogenous DNA of the present invention in all of the germ cells and somatic cells. Means to do. Progeny of such animals that inherit the foreign DNA of the present invention have the foreign DNA of the present invention in all of their germinal and somatic cells.
  • the non-human mammal to which the exogenous normal DNA of the present invention has been transferred is confirmed to stably maintain the exogenous DNA by mating, and is subcultured as an animal having the DNA in a normal breeding environment. Can be done.
  • the introduction of the exogenous DNA of the present invention at the fertilized egg cell stage is ensured to be present in excess in all germ cells and somatic cells of the target mammal.
  • Excessive presence of the exogenous DNA of the present invention in the germinal cells of the produced animal after the introduction of the DNA indicates that all the offspring of the produced animal have the exogenous DNA of the present invention in all of the germ cells and somatic cells.
  • the offspring 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 germ cells 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 endogenous normal DNA, thereby finally obtaining the protein of the present invention. It may develop quality hyperfunction and can be used as a model animal for the disease. For example, using the normal DNA-introduced animal of the present invention, elucidation of the pathological mechanism of hyperactivity of the protein of the present invention and diseases associated with the protein of the present invention, and examination of a method for treating these diseases. It is possible.
  • the mammal into which the exogenous normal DNA of the present invention has been introduced has an increased symptom of the released protein of the present invention
  • screening for a therapeutic agent for the disease associated with the protein of the present invention is performed. It can also be used for testing.
  • the non-human mammal having the foreign abnormal DNA of the present invention can be subcultured in a normal breeding environment as an animal having the DNA after confirming that the foreign DNA is stably maintained by the crossing. I can do it.
  • 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 ordinary DNA engineering techniques. Introduction of the abnormal DNA of the present invention at the fertilized egg cell stage is ensured to be present in all germinal and somatic cells of the target mammal.
  • the presence of the abnormal DNA of the present invention in the germ cells of the produced animal after the introduction of the DNA means that all the offspring of the produced animal have the abnormal DNA of the present invention in all of the germ cells and somatic cells.
  • the progeny of this type of animal that has inherited the exogenous DNA of the present invention has the abnormal DNA of the present invention in all of its germinal and somatic cells.
  • the abnormal DNA of the present invention is highly expressed, and the function of the protein of the present invention is ultimately reduced by inhibiting the function of endogenous normal DNA.
  • Inactive type refractory disease Can be used as an animal.
  • using the abnormal DNA-introduced animal of the present invention it is possible to elucidate the pathological mechanism of the function-inactive refractory of the protein of the present invention and to examine a method for treating this disease.
  • the abnormal DNA highly expressing animal of the present invention can be used to inhibit the function of a normal protein by the abnormal protein of the present invention in the function-inactive refractory disease of the protein of the present invention (dominant negative active protein). Action).
  • the mammal into which the foreign abnormal DNA of the present invention has been introduced since the mammal into which the foreign abnormal DNA of the present invention has been introduced has an increased symptom of the released protein of the present invention, it can be used in a therapeutic drug screening test for the protein of the present invention or its function-inactive refractory disease. Is also available.
  • each organ is removed from the DNA-introduced animal of the present invention, cut into small pieces, and the DNA-introduced cells that have been released with a protease such as trypsin are obtained and cultured.
  • a protease such as trypsin
  • the present inventors can identify the protein-producing cells of the present invention, examine their relationship to apoptosis, differentiation or proliferation, or examine their signal transduction mechanisms, and investigate their abnormalities. It is an effective research material for elucidating the protein and its action.
  • the use of the DNA-introduced animal of the present invention to develop a therapeutic agent for a disease associated with the protein of the present invention including refractory inactive type of the protein of the present invention, Using a test method and a quantitative method, it is possible to provide an effective and rapid screening method for a therapeutic agent for the disease. Also, using the DNA-introduced animal of the present invention or the exogenous DNA-expressing vector of the present invention, it is possible to examine and develop a method for treating a 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 reporter gene (eg, a ⁇ -galactosidase gene derived from Escherichia coli), and
  • a method for screening the salt is provided.
  • a non-human mammalian embryonic stem cell in which the DNA of the present invention has been inactivated is a DNA which is obtained by artificially mutating the DNA of the present invention possessed by the non-human mammal, thereby suppressing the DNA expression ability, Alternatively, the DNA substantially does not have the ability to express the protein of the present invention by substantially losing the activity of the protein of the present invention encoded by the DNA (hereinafter referred to as the knockout DNA of the present invention). In some cases, refers to embryonic stem cells of non-human mammals (hereinafter abbreviated as ES cells).
  • non-human mammal the same one as described above is used.
  • the method of artificially mutating the DNA of the present invention can be performed, for example, by deleting a part or all of the DNA sequence and inserting or substituting another DNA by a genetic engineering technique.
  • the knockout DNA of the present invention may be prepared by, for example, shifting the reading frame of a codon or disrupting the function of a promoter or exon by these mutations.
  • Non-human mammalian embryonic stem cells in which the DNA of the present invention has been inactivated include, for example, The DNA of the present invention possessed by a non-human mammal is isolated, and its exon portion is a drug resistance gene typified by a neomycin resistance gene, a hygromycin resistance gene, lacZ ( ⁇ -galactosidase gene), cat (clo DNA that disrupts exon function by inserting a reporter gene such as the ramphenicol acetyltransferase gene) or terminates gene transcription in the intron between exons.
  • a drug resistance gene typified by a neomycin resistance gene, a hygromycin resistance gene, lacZ ( ⁇ -galactosidase gene), cat (clo DNA that disrupts exon function by inserting a reporter gene such as the ramphenicol acetyltransferase gene) or terminates gene transcription in the intron between exons.
  • a DNA strand having a DNA sequence constructed so as to disrupt the gene (hereinafter abbreviated as “Yuichi Getting Vector”) was introduced into the chromosome of the animal by, for example, homologous recombination, and the obtained ES was obtained.
  • the DNA sequence on or near the DNA of the present invention is purified.
  • the DNA sequence on the Southern hybridization analysis or evening getter vector and the DNA sequence of the neighboring region other than the DNA of the present invention used in the evening getter vector were prepared. It can be obtained by analyzing by the PCR method using primers 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.
  • ⁇ ⁇ ⁇ It may be newly established according to the method of ma.
  • mouse ES cells currently, 129 ES cells are generally used, but since the immunological background is not clear, an alternative pure immunological and genetically
  • BDFi mice C57BL / 6 and C57BL / 6 mice
  • C57BL / 6 mice and C57BL / 6 have reduced the number of eggs collected by crossing with DBA / 2
  • DBA / 2 It is also possible to use a mouse established with DBAZ2, etc.
  • BDFi mice have the advantage of high number of eggs collected and robust eggs, and also have a background of C57BLZ6 mice.
  • ES cells obtained by using C57BLZ6 mice can be used to advantage in that their genetic background can be replaced with C57BLZ6 mice by backcrossing with C57B LZ6 mice when creating disease model mice. .
  • blastocysts 3.5 days after fertilization are generally used. Early embryos can be obtained.
  • Either male or female ES cells may be used, but male ES cells are generally more convenient for producing breeding line 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 (about 50 cells) per colony 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 male and female. Can be reduced to width.
  • the secondary selection can be performed, for example, by confirming the number of chromosomes by the G-banding method.
  • Embryonic stem cell lines obtained in this way usually have very good proliferative properties, but must be carefully subcultured because they tend to lose their ontogenetic potential.
  • a suitable feeder cell such as STO fibroblasts
  • a carbon dioxide incubator preferably 5% carbon dioxide, 95% air or 5% oxygen
  • Culture at about 37 ° C in 5% carbon dioxide gas and 90% air preferably 0.001 to 0.5% trypsin ZO.
  • L to 5 mM EDTA preferably A method is used in which cells are converted into single cells by treatment with about 0.1% trypsin / ImMEDTA and seeded on freshly prepared feeder cells. Such subculture is usually carried out every 1 to 3 days. At this time, it is desirable to observe the cells and, if morphologically abnormal cells are found, discard the cultured cells.
  • ES cells are differentiated into various types of cells, such as parietal, visceral, and cardiac muscles, by monolayer culture up to high density or suspension culture until cell clumps are formed under appropriate conditions.
  • MJ Evans and MH Kaufman Nature, 292, 154, 1981; GR Martin Proceedings of National Academy of Sciences, Science, Proc. Natl. Acad. Sci. USA) Vol. 78, p. 7634, 1981; TC Doetschman et al., Journal of Obemblilogi and Eximental Morphology, Vol. 87, p. 27, 1985.
  • the DNA-deficient cells of the present invention obtained by differentiating the ES cells of the present invention are useful in the cell biology of the protein of the present invention in the mouth of in vivo.
  • the non-human mammal deficient in DNA expression of the present invention is distinguished from a normal animal by measuring the mRNA level of the animal using a known method and indirectly comparing the expression level. It is possible to
  • non-human mammal those similar to the aforementioned can be used.
  • the non-human mammal deficient in expression of the DNA of the present invention can be obtained, for example, by introducing the evening getter vector prepared as described above into mouse embryonic stem cells or mouse egg cells.
  • the DNA of the present invention can be knocked out by homologous recombination in which the inactivated DNA sequence replaces the DNA of the present invention on the chromosome of mouse embryonic stem cells or mouse egg cells by gene homologous recombination. it can.
  • Cells in which the DNA of the present invention has been knocked out can be analyzed by Southern hybridization analysis using a DNA sequence on or near the DNA of the present invention as a probe, or a DNA sequence on a evening-getting vector, and evening-getting. The determination can be made by PCR analysis using, as a primer, the DNA sequence of a neighboring region other than the DNA of the present invention derived from the mouse used in the vector.
  • a non-human mammalian embryonic stem cell is used, a cell line in which the DNA of the present invention has been inactivated by gene homologous recombination is cloned, 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 produced chimeric embryo is transplanted 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 obtained from the population obtained by crossing such a chimeric individual with a normal individual. It can be obtained by selecting an individual composed of cells having the DNA locus of the present invention having a mutation, for example, by judging coat color.
  • the individual obtained in this manner 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. It is possible to obtain an individual with poor homo-expression.
  • a transgenic non-human mammal having a chromosome into which a gettering vector has been introduced can be obtained by injecting a DNA solution into the egg cell nucleus by a microinjection method. Tiger Compared to nicked non-human mammals, they can be obtained by selecting those having a mutation in the DNA locus of the present invention by gene homologous recombination.
  • the individual in which the DNA of the present invention has been knocked out may be subjected to rearing in an ordinary breeding environment after confirming that the DNA has been knocked out even in an animal obtained by mating. it can.
  • the germline can be obtained and maintained according to a standard method. That is, by crossing male and female animals having the inactivated DNA, a homozygous animal having the inactivated DNA on both homologous chromosomes can be obtained. The obtained homozygous animal can be efficiently obtained by rearing the mother animal in such a manner that one normal individual and one homozygote are obtained. By mating male and female heterozygous animals, homozygous and heterozygous animals having the inactivated DNA are bred and subcultured.
  • 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 expression of the DNA of the present invention.
  • the non-human mammal deficient in expressing the DNA of the present invention lacks various biological activities that can be induced by the protein of the present invention, it may be caused by the inactivity of the biological activity of the protein of the present invention. It is useful for investigating the causes of these diseases and examining treatment methods, because they can serve as a model for such diseases.
  • the non-human mammal deficient in DNA expression of the present invention can be used for screening for a compound having a preventive / therapeutic effect against diseases caused by DNA deficiency or damage of the present invention.
  • the present invention is characterized in that a test compound is administered to a non-human mammal deficient in DNA expression of the present invention, and changes in the animal are observed and measured.
  • the present invention provides a method for screening a compound or a salt thereof having a preventive / therapeutic effect on a disease to be caused.
  • test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and plasma.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 and compared with an untreated control animal, and changes in organs, tissues, disease symptoms, etc. of the animal are indicated as an index.
  • the preventive and therapeutic effects of the test compound can be tested.
  • test compound for example, oral administration, intravenous injection, or the like is used, and it can be appropriately selected according to the symptoms of the test animal, 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.
  • a non-human mammal deficient in expression of the DNA of the present invention is treated with coconut smoke or antigen exposure treatment, and tobacco
  • the test compound is administered before or after the smoke or antigen exposure treatment, and changes in the airway responsiveness of the animal are measured over time.
  • the compound obtained by using the screening method is a compound selected from the test compounds described above, and has a preventive / therapeutic effect against a disease caused by deficiency or damage of the protein of the present invention. It can be used as a medicament such as a safe and low toxic prophylactic / therapeutic agent for the disease.
  • a compound derived from the compound obtained by the above-mentioned 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 metals). And the like, and particularly preferred are physiologically acceptable acid addition salts.
  • physiologically acceptable acids eg, inorganic acids, organic acids, etc.
  • bases eg, alkali metals
  • physiologically acceptable acid addition 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) Succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc.).
  • inorganic acids eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric
  • 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, for example, in humans or non-human mammals (eg, rats, mice, guinea pigs, egrets, higgies, bushus, dogs, 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. For example, when the compound is orally administered, it is generally used for adults (with a body weight of 60 kg).
  • the single dose of the compound may vary depending on the administration subject, target disease, etc.
  • the compound is usually injectable in the form of an injection (typically 60 kg) in chronic obstructive pulmonary lungs of adults.
  • the dose can be administered in terms of 60 kg.
  • the present invention relates to a method for administering a test compound to a non-human mammal deficient in expression of a DNA of the present invention, and detecting the expression of a reporter gene.
  • a method for screening a compound or a salt thereof that inhibits the inhibition is provided.
  • the non-human mammal deficient in DNA expression of the present invention may be a non-human mammal deficient in expression of DNA of the present invention, wherein the DNA of the present invention is obtained by introducing a repo allele gene. Those inactivated and capable of expressing the repo overnight gene under the control of a promoter for the DNA of the present invention are used.
  • test compound examples include the same compounds as described above.
  • 3-galactosidase gene (1 ac Z), soluble alkaline phosphatase gene or The sifellanase gene and the like are preferred.
  • tissue of the present invention expressing the protein of the present invention originally ] -Galactosidase is expressed instead of protein.
  • a reagent that is a substrate for 3-galactosidase such as 5-promo-4-chloro-3-indolyl-1) 8-galactopyranoside (X-ga 1)
  • the protein-deficient mouse of the present invention or a tissue section thereof is fixed with dartalaldehyde and the like, washed with phosphate buffered saline (PBS), and then stained with X-ga1 at room temperature or at room temperature. After reacting for about 30 minutes to 1 hour at around 7 ° C, the / 3-galactosidase reaction can be stopped by washing the tissue sample with 1 mM EDTA / PBS solution, and the coloration can be observed. . Further, mRNA encoding 1acZ 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 of the 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, organic acids). And the like, and particularly preferred are physiologically acceptable acid addition salts.
  • physiologically acceptable acids eg, inorganic acids
  • bases eg, organic acids
  • physiologically acceptable acid addition salts examples 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) Succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc.).
  • inorganic acids eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.
  • organic acids eg, acetic acid,
  • the compound of the present invention which inhibits the promoter activity for DNA or the salt thereof can inhibit the expression of the protein of the present invention and inhibit the function of the protein.
  • Respiratory diseases such as chest diseases (eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), allergic conjunctivitis, Rhinitis
  • gastrointestinal diseases eg, As preventive and therapeutic agents for irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease, reflux esophagitis, etc., preferably as preventive and therapeutic agents for respiratory diseases, gastrointestinal diseases, etc. Useful.
  • a compound derived from the compound obtained by the above screening can also be used.
  • 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 low toxic and can be used, for example, in humans or non-human mammals (for example, rats, mice, guinea pigs, egrets, sheep, pigs, horses, cats, cats, Dogs, monkeys, etc.).
  • the dose of the compound or a salt thereof varies depending on the target disease, the administration subject, the administration route, and the like.
  • the compound of the present invention that promotes the promoter activity for DNA is orally administered, generally, the adult ( In patients with chronic obstructive pulmonary disease (with a body weight of 60 kg), about 0.1 to 100 mg, preferably about 1.0 to 50 mg, more preferably about 1.0 to 20 mg of the ehine compound is administered per day. I do.
  • the single dose of the compound varies depending on the administration subject, target disease, and the like.
  • the compound that promotes the promoter activity for DNA of the present invention is usually in the form of an injection.
  • an adult (as 60 kg) patient with chronic obstructive pulmonary disease about 0.01 to 30 mg, preferably about 0.1 to 20 mg, more preferably about 0.1 to 10 mg of the compound per day is administered. It is conveniently administered by intravenous injection. In the case of other animals, the dose can be administered in terms of 60 kg.
  • the compound of the present invention that inhibits the promoter activity against DNA when orally administered, generally, in an adult (assuming a body weight of 60 kg) patient with chronic obstructive pulmonary disease, the compound is reduced to about 0 per day. l-100 mg, preferably about 1.0-50 mg, more preferably about 1.0-20 mg.
  • the single dose of the compound varies depending on the administration subject, target disease, and the like.
  • a compound that inhibits promoter activity against DNA of the present invention is usually administered in the form of an injection. adult
  • 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 that 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 cause of various diseases caused by DNA expression deficiency or the development of preventive and therapeutic agents of the present invention.
  • transgenic animal In addition, 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 polypeptide and examine its action in living organisms. Further, by binding an appropriate repo overnight gene to a part of the promoter and establishing a cell line in which the gene is expressed, the action of specifically promoting or suppressing the in vivo production ability of the protein of the present invention itself is achieved. It can be used as a search system for low molecular weight compounds.
  • bases, amino acids, and the like are indicated by abbreviations based on the abbreviations by IUPAC-IUB Communications 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 Deoxylipo nucleic acid c DNA complementary deoxyribonucleic acid
  • R adenine (A) or guanine (G) Y cytosine (C) or thymine (T)
  • dATP Deoxyadenosine triphosphate
  • dTTP Deoxythymidine triphosphate
  • dGTP Deoxyguanosine triphosphate
  • dCTP Deoxycytidine triphosphate.
  • ATP Adenosine triphosphate
  • the nucleotide sequence of the mouse CLCA4 gene on the 3 'side is shown.
  • Example 1 shows the nucleotide sequence of T7promoteperprimer (promoter primer 1) used in Example 1.
  • Example 1 shows the nucleotide sequence of Ml 3 RV primer (primary) used in Example 1.
  • [SEQ ID NO: 22] 7 shows the nucleotide sequence of primer 12 used in Example 5.
  • Example 14 shows the base sequence of primer 14 used in Example 5.
  • Transformant Escherichia coli Escherichia coli (Escherichia coli) TOPlO / pCR-Bluntll-mCLCA4 obtained in Example 1 described below has been used since February 12, 2002, 1-1 1-1 Tsukuba-Higashi, Ibaraki Pref. Deposit No.FERM BP-7887 at the National Institute of Advanced Industrial Science and Technology (AIST) with a postal code of 305-8566) as a deposit number FERM BP-7887 from January 29, 2002 at 2-317 Jusanhoncho, Yodogawa-ku, Osaka, Osaka It has been deposited with the Fermentation Research Institute (IF0) of No. 85 (zip code 532-8686) under the accession number IF0 16751.
  • IF0 Fermentation Research Institute
  • Example 1 The transformant Escherichia coli TOPlO / pCR-Bluntll-mCLCA4A obtained in Example 3 described below has been used since March 4, 2002, 1-1 1-1 Tsukuba, Higashi, Ibaraki Pref. National Institute of Advanced Industrial Science and Technology (ZIP code 305-8566). Deposited at the Patent Organisms Depositary Center as deposit number FERM BP-7934. From February 19, 2002, 2-3-17 Jusanhoncho, Yodogawa-ku, Osaka, Osaka, Japan 85 No. (postal code 532-8686) and deposited with the Fermentation Research Institute (IF0) under the accession number IF0 16764.
  • IF0 Fermentation Research Institute
  • mRNA was prepared using mRNA purification kit (manufactured by Amersham Pharmacia Biotech). Using this mRNA 1 ig as a starting material, cDNA was synthesized by reverse transcription using a Marathon cDNA Amplification kit (manufactured by Clontech). After performing PCR using this cDNA as type I and a combination of Primer 1 and Primer 1 and Primer 3 and Primer 4, a DNA fragment of the expected size is amplified by agarose gel electrophoresis. Admitted.
  • the reaction was carried out using Pyrobest DNA polymerase (Takara Shuzo Co., Ltd.) on a Thermocycler Gene Amp PCR System 9700 (PerkinElmer) at 94 ° C for 30 seconds, then at 94 for 10 seconds.
  • the reaction cycle was repeated 35 times at 60 ° C for 30 seconds, 72 ° C for 2 minutes and 30 seconds, and finally the reaction was performed at 72 ° C for 10 minutes. It was cloned into the obtained DNA fr3 ⁇ 4pCR_BluntII-T0P0 (manufactured by Invitrogen).
  • T7 promoter primer SEQ ID NO: 15
  • M13RV primer SEQ ID NO: 16
  • six synthetic primers Primer 5 (SEQ ID NO: 9), Primer 6 (SEQ ID NO: 10), Primer 7 ( Cycle sequence reaction was performed using SEQ ID NO: 11), Primer 8 (SEQ ID NO: 12), Primer 9 (SEQ ID NO: 13), and Primer 10 (SEQ ID NO: 14)] to obtain a fluorescent DNA sequencer.
  • the base sequence of the reaction product obtained by I-377 (PerkinElmer) was determined. As a result, clone No.
  • mouse CLCA4 was found to be composed of 2772 nucleotide sequences (SEQ ID NO: 2). Based on the nucleotide sequence represented by SEQ ID NO: 2, 924 amino acid sequences were determined (SEQ ID NO: 1), and named as mouse CLCA4 protein. Subsequently, clone No.
  • mice CLCA4 7-3 was digested with Hind I II to cut out a DNA fragment containing the C-terminal region of mouse CLCA4 of 1.4 Kb. This was ligated to a vector sequence containing the N-terminal region of mouse CLCA4 of clone No. 3-1 also digested with Hindi II to construct a plasmid pCR-Bluntll-mCLCA4 containing a mouse CLCA4 full-length sequence. A transformant containing this plasmid was designated as Escherichia coli TOP10 / pCR-BluntII-mCLCA4. At the amino acid level, mouse CLCA4 had 68% homology with human CLCA4 and 51% homology with mouse gob-5.
  • Example 2 Example 2
  • mouse tissues bone marrow, eyes, lymph nodes, smooth muscle, ovary, thymus, stomach, bladder, heart
  • Brain spleen, lung, liver, skeletal muscle, kidney, testis, embryos on day 7, 11, 15, 17 and colon (Mouse MTC panel I and Mouse TC panel II: Clontech) PCR was performed.
  • the reaction was performed using Takara Ex Taq (manufactured by Takara Shuzo Co., Ltd.) on a Thermocycler GeneAmp PCR System 9600 (manufactured by Applied Biosystems) first at 94 ° C for 30 seconds, then at 94 ° C for 10 seconds and 60 ° C. The reaction was repeated for 35 cycles, with one reaction cycle consisting of 30 seconds at 72 ° C and 2 minutes at 72 ° C. The obtained reaction product was subjected to electrophoresis on a 2% agarose gel, and then stained with B. bromide to detect an amplified 800 bp band. As a result, the mouse CLCA4 gene product (mRNA) was strongly expressed in smooth muscle and large intestine, and was also expressed in skeletal muscle.
  • mRNA mouse CLCA4 gene product
  • mouse smooth muscle cDNA (Mouse MTC panel II: manufactured by Clontech) as type I
  • PCR was performed using a combination of primer 1 (SEQ ID NO: 5) and primer 11 (SEQ ID NO: 21), and agarose electrophoresis was performed. It was confirmed that a 2.7 Kb DNA fragment was amplified by electrophoresis.
  • the reaction was carried out using Pyrobest DNA polymerase (Takara Shuzo Co., Ltd.) on a Thermocycler Gene Amp PCR System 9700 (Kin-Elma Co., Ltd.) for 30 seconds at 94 ° C. The reaction was repeated for 35 cycles, each cycle consisting of 30 seconds at 60 seconds and 5 minutes at 72, and the reaction was continued at 72 ° C for 10 minutes. The obtained DNA fragment was pCR-B1 until -T0P0 (Manufactured by Invitrogen).
  • T7 promoter primer SEQ ID NO: 15
  • 13RV primer SEQ ID NO: 16
  • six synthetic primers [Primer 1 (SEQ ID NO: 9), Primer 6 (SEQ ID NO: 10), Primer 7 (SEQ ID NO: 11), PRIMER-1 (SEQ ID NO: 12), PRIMER-1 (SEQ ID NO: 13) and PRIMER-10 (SEQ ID NO: 14)
  • the nucleotide sequence of the reaction product obtained using DNA Sequencer-377 was determined.
  • a 1020 bp DNA fragment amplified by PCR using a combination of primer 2 (SEQ ID NO: 6) and primer 3 (SEQ ID NO: 7), which are the common sequence of mouse CLCA4 and mouse CLCA4A, is derived from mouse CLCA4 by the restriction enzyme PvuII (Takara Shuzo) Using the fact that mouse CLCA4A-derived DNA was digested to 680 bp and 340 bp, the tissue distribution of each was examined.
  • the PCR reaction was performed using Takara Ex Taq (Takara Shuzo) in a thermal cycler GeneAmp PCR System 9600 (Applied Biosystems) at 94 ° C for 30 seconds, then at 94 for 10 seconds and 6 (TC The reaction was carried out by repeating 35 cycles with 1 reaction cycle consisting of 30 seconds and 2 minutes at 72 ° C.
  • the obtained reaction product was converted into PvuII, subjected to electrophoresis on a 2% agarose gel, and then stained with CHBr.
  • mRNA was prepared using a purification kit (Amersham Pharmacia Biotech). Using this mRNA lpg as a starting material, cDNA was synthesized by a reverse transcription reaction using a Marathon cDNA Amplification kit (manufactured by Clontech). After performing PCR using this cDNA as type I and a combination of primers 12 and 13, it was confirmed that a DNA fragment of the expected size was amplified by agarose electrophoresis. The reaction was performed using Pyrobest DNA polymerase (Takara Shuzo Co., Ltd.) in a Thermocycler Gene Amp PCR System 9700 (PerkinElmer Co., Ltd.) first at 94 ° C for 30 seconds, and then at 94 ° C.
  • the reaction cycle was repeated 35 cycles, each consisting of 10 seconds, 30 seconds at 60 ° C, and 2 minutes at 72 ° C. Finally, the reaction was performed at 72 ° C for 10 minutes.
  • the obtained DNA fragment was cloned into pCR-Bluntll-TOP0 (Invitrogen).
  • T7 promoter primer SEQ ID NO: 15
  • M13RV primer SEQ ID NO: 16
  • three synthetic primers [Primer 12 (SEQ ID NO: 22), Primer 13 (SEQ ID NO: 23), Primer 14 (SEQ ID NO: 24)]
  • the nucleotide sequence of the reaction product obtained with a fluorescent DNA sequencer-377 was determined.
  • a clone No. a-20 (SEQ ID NO: 20) in which 1119 nucleotide sequences were inserted as a partial nucleotide sequence of rat CLCA4 gene was obtained.
  • SEQ ID NO: 20 Based on the nucleotide sequence represented by SEQ ID NO: 20, a partial amino acid sequence of 373 was determined (SEQ ID NO: 19).
  • the partial amino acid sequence had 66% homology with the corresponding sequence of human CLCA4 and 81% homology with the corresponding sequences of mouse CLCA4 and mouse CLCA4A. '' Industrial applicability
  • the proteins and polynucleotides of the present invention include, for example, lungs, lungs with inflammation of the airways, respiratory diseases such as chest diseases [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolosis Inflammation, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), allergic conjunctivitis, rhinitis (eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, before drying Rhinitis, vasomotor rhinitis, gangrenous rhinitis, sinusitis, etc., gastrointestinal diseases (eg, irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease, reflux esophagitis, etc.) It is useful as a diagnostic marker or the like, and is an inhibitor obtained
  • respiratory diseases such as lungs, lungs with inflammation of the respiratory tract, and chest disease
  • chronic obstructive pulmonary disease chronic bronchitis, emphysema
  • diffuse panbronchiolitis bronchial asthma
  • cystic fibrosis Disease irritable pneumonia
  • allergic conjunctivitis rhinitis (eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry prorhinitis, vascular hunting rhinitis, gangrene
  • Prophylactic / therapeutic agents such as rhinitis, sinusitis, etc.
  • gastrointestinal diseases eg, irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease, reflux esophagitis, etc.
  • respiration preferably respiration It can be used as a preventive and therapeutic agent for organ diseases and gastrointestinal diseases.
  • the antisense polynucleotide of the present invention can suppress the expression of the protein of the present invention.
  • respiratory diseases such as lungs, lungs accompanied by airway inflammation, and chest diseases (eg, chronic obstructive pulmonary disease).
  • rhinitis eg, allergic rhinitis, pollinosis, acute rhinitis, chronic Rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry proctitis, vasomotor rhinitis, 'gangrene rhinitis, sinusitis, etc.
  • gastrointestinal disorders eg, irritable bowel syndrome, inflammatory bowel disease, ulcerative colon
  • Prevention and treatment of diseases such as inflammation, Crohn's disease, reflux esophagitis, etc., preferably prevention of respiratory diseases, gastrointestinal diseases, etc.
  • various polynucleotides of the present invention may be used for respiratory diseases such as lungs, lungs with airway inflammation, and chest diseases [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiole Inflammation, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), allergic conjunctivitis, rhinitis (eg, rhinorrhea, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, before drying Rhinitis, vasomotor rhinitis, gangrenous rhinitis, sinusitis, etc., gastrointestinal disorders (eg, irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease, reflux esophagitis, etc.) Useful for diagnosis, prevention or treatment.
  • respiratory diseases such as lungs, lungs with airway

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pulmonology (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Peptides Or Proteins (AREA)

Abstract

A protein and a polynucleotide which are useful as diagnostic markers for respiratory diseases, nephritis, digestive diseases, etc. Inhibitors obtained by screening with the use of the above protein or polynucleotide or a neutralizing antibody inhibiting the activity of the protein are usable as, for example, preventives/remedies for the above-described diseases.

Description

明 細 書 新規タンパク質、 その D N Aおよびその用途 技術分野  Description New protein, its DNA and its application Technical field
本発明は、 新規タンパク質、 該タンパク質をコードする D NA、 該タンパク 質の活性を阻害する化合物のスクリーニング方法、 該スクリ一エング方法で得 られる化合物等に関する。 さらに詳しくは、 呼吸器疾患、 消化器疾患などの予 防 ·治療剤または診断薬として有用な新規タンパク質等に関する。 背景技術  The present invention relates to a novel protein, DNA encoding the protein, a method for screening a compound that inhibits the activity of the protein, a compound obtained by the screening method, and the like. More specifically, the present invention relates to a novel protein useful as a prophylactic / therapeutic agent or diagnostic agent for respiratory diseases, digestive diseases, and the like. Background art
慢性閉塞性肺疾患 (慢性気管支炎、 肺気腫) 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) is considered to become the central disease of respiratory illness with the aging of the smoking generation and prolonged life expectancy.
また、 気管支喘息は気道の慢性炎症性疾患であり、 気道狭窄を示し、 発作性 の呼吸困難、 喘鳴、 咳などの症状が見られる。 その発症と進展には気道上皮細 胞、 肥満細胞、 好酸球、 Tリンパ球などの多くの細胞が関与している。 気管支 喘息の最も重要な特徴の 1つは、 気道が刺激に対して反応しやすいこと (気道 過敏性) である。 この気道過敏性は、 好酸球など気道に浸潤した細胞から分泌 される化学伝達物質による気道上皮の剥離を中心とする気道の炎症に起因する が、 さらに、 遺伝因子や環境因子も複雑に影響していると考えられている。 外界からの刺激 (アレルゲン、 排気物) やウィルス感染により気道の炎症反 応の引き金が引かれると、 気道上皮細胞や気管支周辺の毛細血管内皮細胞上に VCAM-K ICAM-1などの接着分子が発現し 〔ジャーナル ·ォブ ·ァラジ一 'アン ド ·クリニカル.ィムノロジ一 (J. Al lergy Cl in. I腿 unol . ) 、 96巻、 941頁 (1995) 〕 、 サイト力インや化学遊走物質が産生される。 気管支喘息の患者は Th2型のへルパ一 T細胞の機能が亢進しており、 IL- 3、 IL- 4、 IL_5、 IL-13、 GM - CSFなどの Th2型のサイトカインゃ eotaxin、 RANTESなどのケモカインの産生が増 加する。 IL- 4や IL-13は IgEの産生誘導作用があり、 IL- 3や IL-4は肥満細胞の増 殖誘導作用がある。 さらに、 IL- 5、 GM- CSFなどの作用により好酸球が分化増殖 し、 eotaxin、 RANTESにより気道に浸潤してくる 〔ァラジー ·アンド 'ァズマ · プロシーデイング (Al l ergy As thma Proc. ) 、 20巻、 141頁 (1999) 〕 。 Bronchial asthma is a chronic inflammatory disease of the respiratory tract, with airway narrowing and paroxysmal dyspnea, wheezing, and coughing. Many cells are involved in its development and progression, including airway epithelial cells, mast cells, eosinophils, and T lymphocytes. One of the most important features of bronchial asthma is that the airway is more responsive to stimuli (airway hyperresponsiveness). This airway hyperreactivity is caused by inflammation of the airway, mainly by detachment of the airway epithelium by chemical mediators secreted from cells infiltrating the airway such as eosinophils, but genetic factors and environmental factors also have complex effects Is believed to be. When an inflammation reaction in the airways is triggered by external stimuli (allergens, exhausts) or viral infection, adhesion molecules such as VCAM-K ICAM-1 are found on airway epithelial cells and capillary endothelial cells around the bronchi. (J. Allergy Cl in. I Thigh unol., 96, 941 (1995)), and the site force and chemical migrants Produced. Patients with bronchial asthma have enhanced function of Th2-type helper T cells, and Th2-type cytokines such as IL-3, IL-4, IL_5, IL-13, and GM-CSF. Chemokine production increases. IL-4 and IL-13 induce IgE production, and IL-3 and IL-4 increase mast cells. There is a breeding induction effect. In addition, eosinophils are differentiated and proliferated by the action of IL-5, GM-CSF, etc., and infiltrate the respiratory tract by eotaxin and RANTES (Allergy Asthma Proc.), 20, 141 (1999)].
ヒト由来 CLCA (Cl-channel, Ca2+-act ivated) 4の DNAおよびタンパク質は、 力 ルシゥム依存性のクロライドチャネルタンパク質ファミリ一に属するものの 〔フエブス · レターズ (FEBS Let ters) 、 455卷、 295頁 (1999) 〕 、 慢性閉塞 性肺疾患 (C0PD; Chronic Obs truct ive Pulmonary disease) 、 気管支喘息との 関連は知られていない。 The DNA and protein of human-derived CLCA (Cl-channel, Ca 2+ -activated) 4 belong to the power channel-dependent chloride channel protein family, although they belong to FEBS Let ters, Vol. 455, p. 295. (1999)], No association with chronic obstructive pulmonary disease (C0PD; Chronic Obstructive Pulmonary disease) or bronchial asthma is known.
' 一方、 ヒト CLCA1遺伝子の塩基配列は、 ジエノミクス (Genomics) 、 54卷、 200頁 (1998) 、 バイオケミカル アンド バイオフィジカル リサーチ コミ ュニケーシヨンズ (Biochem Biophys Res Commun) 、 255巻、 347頁 (1999) な どに記載されており、 この遺伝子が気管支喘息、 慢性閉塞性肺疾患と関連して いることが報告されている (W0 01/38530号公報) 。 ヒト CLCA1遺伝子の塩基配 列と相同性を示すものとして、 マウス gob-5遺伝子の塩基配列 〔バイオケミカル アンド バイオフィジカル リサーチ コミュニケーション (Bi ochem. 'On the other hand, the nucleotide sequence of the human CLCA1 gene has been reported in Genomics, Vol. 54, p. 200 (1998), Biochemical and Biophysical Research Communications, Biochem Biophys Res Commun, Vol. This gene has been reported to be associated with bronchial asthma and chronic obstructive pulmonary disease (W001 / 38530). The one that shows homology to the base sequence of the human CLCA1 gene is the base sequence of the mouse gob-5 gene (Biochemical and Biophysical Research Communication (Biochem.
Biophys. Res. Commun. ) 、 255巻、 347頁 (1999) 〕 、 ブタ CLCA1遺伝子の塩基 配列 〔フイジォロジカル ゲノミックス (Phys iol . Genomics) 、 3巻、 101頁 (2000) 〕 などが知られている。  Biophys. Res. Commun.), 255, 347 (1999)], and the base sequence of porcine CLCA1 gene (Physiol. Genomics, 3, 101 (2000)).
副作用の少ない優れた慢性閉塞性肺疾患、 気管支喘息などの予防 ·治療剤お よび診断薬の開発が望まれている。 発明の開示  There is a need for the development of prophylactic and therapeutic agents and diagnostics for chronic obstructive pulmonary disease and bronchial asthma with few side effects. Disclosure of the invention
本発明者らは、 上記の課題を解決するために鋭意研究を重ねた結果、 カルシ ゥム依存性のクロライドチャネルファミリーに属する新規遺伝子を見出し、 本 発明を完成するに至った。  The present inventors have conducted intensive studies to solve the above-mentioned problems, and as a result, have found a novel gene belonging to a calcium-dependent chloride channel family, and have completed the present invention.
すなわち、 本発明は、  That is, the present invention
( 1 ) 配列番号: 1もしくは配列番号: 1 7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塩、 (1) a protein containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 or SEQ ID NO: 17 or a salt thereof,
( 2 ) 配列番号: 1で表されるアミノ酸配列からなるタンパク質またはその 塩、 (2) a protein consisting of the amino acid sequence represented by SEQ ID NO: 1 or a protein thereof; salt,
(3) 配列番号: 17で表されるアミノ酸配列からなるタンパク質またはそ の塩、  (3) a protein comprising an amino acid sequence represented by SEQ ID NO: 17 or a salt thereof;
(4) 配列番号: 19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塩、  (4) a protein or a salt thereof having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 19,
(5) 配列番号: 19で表されるアミノ酸配列を含有するタンパク質または その塩、  (5) a protein containing the amino acid sequence represented by SEQ ID NO: 19 or a salt thereof,
(6) 上記'(1) 記載のタンパク質の部分ペプチドまたはその塩、  (6) The partial peptide of the protein according to the above (1) or a salt thereof,
(7) 上記 (4) 記載のタンパク質の部分ペプチドまたはその塩、  (7) a partial peptide of the protein according to (4) or a salt thereof,
(8) 上記 (1) 記載の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するポリヌクレオチド、  (8) a polynucleotide containing a polynucleotide encoding the protein or a partial peptide thereof according to (1),
(9) 上記 (4) 記載の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するポリヌクレオチド、  (9) a polynucleotide containing a polynucleotide encoding the protein or a partial peptide thereof according to the above (4),
(10) DNAである上記 (8) または (9) 記載のポリヌクレオチド、 (11) 配列番号: 2または配列番号: 18で表される塩基配列からなるポ リヌクレオチド、  (10) the polynucleotide according to the above (8) or (9), which is a DNA; (11) a polynucleotide having a base sequence represented by SEQ ID NO: 2 or SEQ ID NO: 18,
(12) 配列番号: 20で表される塩基配列を含有する上記 (10) 記載の ポリヌクレオチド、  (12) the polynucleotide according to (10), which comprises the nucleotide sequence represented by SEQ ID NO: 20;
(13) 上記 (8) または (9) 記載のポリヌクレオチドを含有する組換え ベクタ一、  (13) a recombinant vector containing the polynucleotide according to (8) or (9) above,
(14) 上記 (13) 記載の組換えべクタ一で形質転換された形質転換体、 (14) a transformant transformed with the recombinant vector according to (13),
(15) 上記 (14) 記載の形質転換体を培養し、 上記 (1) もしくは (4) 記載のタンパク質またはその部分ペプチドまたはそれらの塩を生成、 蓄 積せしめ、 これを採取することを特徴とする上記 (1) もしくは (4) 記載の タンパク質またはその部分ペプチドまたはそれらの塩の製造法、 (15) culturing the transformant according to (14) above, producing and accumulating the protein or partial peptide thereof or a salt thereof according to (1) or (4), and collecting the resulting protein; A method for producing the protein or partial peptide thereof or a salt thereof according to the above (1) or (4),
(16) 上記 (1) もしくは (4) 記載のタンパク質またはその部分べプチ ドまたはそれらの塩を含有してなる医薬、  (16) a medicament comprising the protein of (1) or (4) or a partial peptide thereof or a salt thereof,
(17) 上記 (1) または (4) 記載のタンパク質またはその部分ペプチド またはそれらの塩に対する抗体、 (18) 上記 (17) 記載の抗体を含有してなる医薬、 (17) an antibody against the protein according to (1) or (4) or a partial peptide thereof or a salt thereof, (18) a medicament comprising the antibody according to (17),
(19) 上記 (17) 記載の抗体を含有してなる診断薬、  (19) a diagnostic agent comprising the antibody according to the above (17),
(20) 上記 (8) または (9) 記載の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を含有するアンチセンス ポリヌクレオチド、  (20) an antisense polynucleotide comprising a nucleotide sequence complementary or substantially complementary to the nucleotide sequence of the polynucleotide according to (8) or (9) or a part thereof,
(21) 上記 (20) 記載のアンチセンスポリヌクレオチドを含有してなる  (21) comprising the antisense polynucleotide according to (20) above.
(22) 上記 (1) もしくは (4) 記載のタンパク質またはその部分べプチ ドまたはそれらの塩を用いることを特徴とする、 上記 (1) もしくは (4) 記 載のタンパク質またはその部分ペプチドまたはそれらの塩の活性を阻害する化 合物またはその塩のスクリーニング方法、 (22) The protein according to (1) or (4) or a partial peptide thereof or a salt thereof, wherein the protein according to (1) or (4) or a partial peptide thereof or a salt thereof is used. A method for screening for a compound that inhibits the activity of a salt of
(23) 上記 (1) もしくは (4) 記載のタンパク質またはその部分べプチ ドまたはそれらの塩を含有することを特徴とする、 上記 (1) もしくは (4) 記載のタンパク質またはその部分べプチドまたはそれらの塩の活性を阻害する 化合物またはその塩のスクリーニング用キット、  (23) The protein according to (1) or (4) or a partial peptide or a partial peptide thereof, which contains the protein according to the above (1) or (4) or a partial peptide thereof or a salt thereof. Kits for screening compounds or salts thereof that inhibit the activity of those salts,
(24) 上記 (22) 記載のスクリーニング方法または上記 (23) 記載の スクリーニング用キットを用いて得られうる、 上記 (1) もしくは (4) 記載 のタンパク質またはその部分ペプチドまたはそれらの塩の活性を阻害する化合 物またはその塩、  (24) The activity of the protein of (1) or (4) or a partial peptide thereof or a salt thereof, which can be obtained using the screening method of (22) or the screening kit of (23). Inhibiting compounds or salts thereof,
(25) 上記 (24) 記載の化合物またはその塩を含有してなる医薬、 (25) a medicament comprising the compound according to (24) or a salt thereof,
(26) 上記 (8) または (9) 記載のポリヌクレオチドを用いることを特 徴,とする、 上記 (1) または (4) 記載のタンパク質遺伝子の発現を阻害する 化合物またはその塩のスクリーニング方法、 (26) a method for screening a compound or a salt thereof that inhibits the expression of the protein gene according to (1) or (4), characterized by using the polynucleotide according to (8) or (9);
(27) 上記 (8) または (9) 記載のポリヌクレオチドを含有することを 特徴とする、 上記 (1) または (4) 記載のタンパク質遺伝子の発現を阻害す る化合物またはその塩のスクリーニング用キット、  (27) A kit for screening a compound or a salt thereof that inhibits the expression of the protein gene according to (1) or (4), which comprises the polynucleotide according to (8) or (9). ,
(28) 上記 (26) 記載のスクリーニング方法または上記 (27) 記載の スクリーニング用キットを用いて得られうる、 上記 (1) または (4) 記載の タンパク質遺伝子の発現を阻害する化合物またはその塩、 (29) 上記 (28) 記載の化合物またはその塩を含有してなる医薬、(28) A compound or a salt thereof that inhibits the expression of the protein gene according to (1) or (4), which can be obtained using the screening method according to (26) or the screening kit according to (27). (29) a medicament comprising the compound according to (28) or a salt thereof,
(30) 上記 (17) 記載の抗体を用いることを特徴とする、 上記 (1) ま たは (4) 記載のタンパク質の発現を阻害する化合物またはその塩のスクリ一 ニング方法、 (30) A method for screening a compound or a salt thereof that inhibits the expression of a protein according to (1) or (4), which comprises using the antibody according to (17).
(31) 上記 (17) 記載の抗体を含有することを特徴とする、 上記 (1) または (4) 記載のタンパク質の発現を阻害する化合物またはその塩のスクリ 一二ング用キッ卜、  (31) A kit for screening a compound or a salt thereof, which inhibits the expression of the protein according to (1) or (4), which comprises the antibody according to (17).
(32) 上記 (30) 記載のスクリーニング方法または上記 (31) 記載の スクリーニング用キットを用いて得られうる、 上記 (1) または (4) 記載の タンパク質の発現を阻害する化合物またはその塩、  (32) A compound or a salt thereof that inhibits the expression of the protein according to (1) or (4), which can be obtained using the screening method according to (30) or the screening kit according to (31).
(33) 上記 (32) 記載の化合物またはその塩を含有してなる医薬、 (34) 呼吸器疾患、 鼻炎または消化器疾患の予防 ·治療剤である上記 ( 1 6) 、 (18) 、' (21) 、 (25) 、 (29) または (33) 記載の医薬、 (35) 慢性閉塞性肺疾患、 気管支喘息、 炎症性腸疾患または逆流性食道炎 の予防 ·治療剤である上記 (34) 記載の医薬、  (33) A medicine comprising the compound or salt thereof according to (32) above, (34) The above-mentioned (16), (18), which is an agent for preventing or treating respiratory diseases, rhinitis or digestive diseases. (21), (25), (29) or the drug according to (33), (35) the above-mentioned (34) which is an agent for preventing or treating chronic obstructive pulmonary disease, bronchial asthma, inflammatory bowel disease or reflux esophagitis. ) Described medicament,
(36) 呼吸器疾患、 鼻炎または消化器疾患の診断薬である上記 (19) 記 載の診断薬、  (36) The diagnostic agent according to (19) above, which is a diagnostic agent for respiratory disease, rhinitis or gastrointestinal disease.
(37) 慢性閉塞性肺疾患、 気管支喘息、 炎症性腸疾患または逆流性食道炎 の診断薬である上記 (36) 記載の診断薬、  (37) The diagnostic agent according to the above (36), which is a diagnostic agent for chronic obstructive pulmonary disease, bronchial asthma, inflammatory bowel disease or reflux esophagitis.
(38) 哺乳動物に対して、 上記 (24) 、 (28) または (32) 記載の 化合物またはその塩の有効量を投与することを特徴とする慢性閉塞性肺疾患、 気管支喘息、 炎症性腸疾患または逆流性食道炎の予防 ·治療方法、  (38) Chronic obstructive pulmonary disease, bronchial asthma, inflammatory bowel characterized by administering to a mammal an effective amount of the compound according to (24), (28) or (32) or a salt thereof. How to prevent or treat disease or reflux esophagitis,
(39) 慢性閉塞性肺疾患、 気管支喘息、 炎症性腸疾患または逆流性食道炎 の予防 ·治療剤を製造するための上記 (24) 、 (28) または (32) 記載 の化合物またはその塩の使用などを提供する。  (39) The compound or salt thereof according to the above (24), (28) or (32) for producing a prophylactic or therapeutic agent for chronic obstructive pulmonary disease, bronchial asthma, inflammatory bowel disease or reflux esophagitis. Provide use and so on.
さらには、 ,  Moreover, ,
(40) タンパク質が、 (a) 配列番号: 1または配列番号: 17で表される アミノ酸配列、 (b) 配列番号: 1または配列番号: 17で表されるアミノ酸配 列中の 1または 2個以上 (好ましくは、 1〜30個程度、 好ましくは 1〜10 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、 (c) 配列番号: 1または配列番号: 1 7で表されるアミノ酸配列に 1また は 2個以上 (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、 ( 配列 番号: 1または配列番号: 1 7で表されるアミノ酸配列に 1または 2個以上 (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、 (e) 配列番号: 1 または配列番号: 1 7で表されるアミノ酸配列中の 1または 2個以上 (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 (f) これら (b) 〜 (e) を組み合わせたアミノ酸配列を含有するタンパク質である 上記 (1 ) 記載のタンパク質、 (40) the protein is (a) the amino acid sequence represented by SEQ ID NO: 1 or SEQ ID NO: 17; (b) one or two amino acids in the amino acid sequence represented by SEQ ID NO: 1 or SEQ ID NO: 17; Or more (preferably about 1 to 30, preferably 1 to 10 An amino acid sequence in which about 1 amino acid has been deleted, more preferably a number (1 to 5) amino acids, and (c) 1 or 2 or more amino acids in the amino acid sequence represented by SEQ ID NO: 1 or 17 (Preferably about 1 to 30, preferably about 1 to 10, and more preferably a number (1 to 5) of amino acid sequences; (SEQ ID NO: 1 or SEQ ID NO: 1) One or more (preferably about 1 to 30, preferably about 1 to 10, and more preferably a number of (1 to 5)) amino acids were inserted into the amino acid sequence represented by 7 An amino acid sequence, (e) one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 1 or SEQ ID NO: 17 (preferably, about 1 to 30, preferably about 1 to 10) An amino acid sequence in which the number (1 to 5) of amino acids is more preferably replaced by another amino acid Or (f) thereof (b) ~ protein in the above (1) containing the amino acid sequence that is a combination of (e) wherein the protein,
( 4 1 ) 上記 (8 ) または (9 ) 記載のポリヌクレオチドとハイストリンジ ェントな条件下でハイプリダイズするポリヌクレオチド、  (41) a polynucleotide that hybridizes with the polynucleotide according to (8) or (9) above under high stringent conditions,
( 4 2 ) 上記 (8 ) または (9 ) 記載のポリヌクレオチドを含有してなる診 断薬なども提供する。 発明を実施するための最良の形態  (42) A diagnostic agent or the like containing the polynucleotide according to the above (8) or (9) is also provided. BEST MODE FOR CARRYING OUT THE INVENTION
本発明の配列番号: 1または配列番号: 1 7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、 および配列番 号: 1 9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と称することもある) は、 ヒトゃ非ヒト温血動物 (例えば、 モルモット、 ラット、 マウス、 ニヮトリ、 ゥ サギ、 ブ夕、 ヒッジ、 ゥシ、 サルなど) の細胞 (例えば、 肝細胞、 脾細胞、 神 経細胞、 グリア細胞、 塍臓 B細胞、 骨髄細胞、 メサンギゥム細胞、 ランゲルハン ス細胞、 表皮細胞、 上皮細胞、 杯細胞、 内皮細胞、 平滑筋細胞、 繊維芽細胞、 繊維細胞、 筋細胞、 脂肪細胞、 免疫細胞 (例、 マクロファージ、 Τ細胞、 Β細 胞、 ナチュラルキラー細胞、 肥満細胞、 好中球、 好塩基球、 好酸球、 単球) 、 巨核球、 滑膜細胞、 軟骨細胞、 骨細胞、 骨芽細胞、 破骨細胞、 乳腺細胞、 肝細 胞もしくは間質細胞、 またはこれら細胞の前駆細胞、 幹細胞もしくはガン細胞 など) もしくはそれらの細胞が存在するあらゆる組織、 例えば、 脳、 脳の各部 位 (例、 嗅球、 扁桃核、 大脳基底球、 海馬、 視床、 視床下部、 大脳皮質、 延髄、 小脳) 、 脊髄、 下垂体、 胃、 塍臓、 腎臓、 肝臓、 生殖腺、 甲状腺、 胆のう、 骨 髄、 副腎、 皮膚、 筋肉、 肺、 消化管 (例、 大腸、 小腸) 、 血管、 心臓、 胸腺、 脾臓、 顎下腺、 末梢血、 前立腺、 睾丸、卵巣、 胎盤、 子宮、 骨、 関節、 骨格筋な どに由来するタンパク質であってもよく、 合成タンパク質であってもよい。 A protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or 17 of the present invention, and the same or substantially the same as the amino acid sequence represented by SEQ ID NO: 19 Proteins containing substantially the same amino acid sequence (hereinafter sometimes referred to as the protein of the present invention) are human warm non-human animals (eg, guinea pigs, rats, mice, chickens, egrets, bush, Cells (eg, hepatocytes, spleen cells, neurons, glial cells, kidney B cells, bone marrow cells, mesangial cells, Langerhans cells, epidermal cells, epithelial cells, goblet cells, etc.) Endothelial cells, smooth muscle cells, fibroblasts, fibrocytes, muscle cells, adipocytes, immune cells (eg, macrophages, Τ cells, Β cells, natura Killer cells, mast cells, neutrophils, basophils, eosinophils, monocytes), megakaryocytes, synovial cells, chondrocytes, osteocytes, osteoblasts, osteoclasts, mammary gland cells, hepatocyte Vesicles or stromal cells, or their precursors, stem cells, or cancer cells) or any tissue in which these cells are present, such as the brain, various parts of the brain (eg, olfactory bulb, amygdala, basal sphere, hippocampus) , Thalamus, hypothalamus, cerebral cortex, medulla, cerebellum), spinal cord, pituitary, stomach, kidney, kidney, liver, gonad, thyroid, gall bladder, bone marrow, adrenal gland, skin, muscle, lung, digestive tract (eg, Large intestine, small intestine), blood vessels, heart, thymus, spleen, submandibular gland, peripheral blood, prostate, testicle, ovary, placenta, uterus, bone, joint, skeletal muscle, etc. It may be.
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で表わされるアミノ酸配列と約 7 0 %以上、 好ましくは約 8 0 %以上、 好ましくは約 9 0 %以上、 さらに好ましくは約 9 5 %以上、 より好 ましくは約 9 7 %以上の相同性を有するァミノ酸配列などが挙げられる。  The amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 is about 70% or more, preferably about 80% or more, preferably about 90% or more as the amino acid sequence represented by SEQ ID NO: 1. % Or more, more preferably about 95% or more, and even more preferably about 97% or more.
'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。  'As a protein having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1, for example, a protein having an amino acid sequence substantially the same as the amino acid sequence represented by the aforementioned SEQ ID NO: 1 And a protein having substantially the same activity as the protein having the amino acid sequence represented by SEQ ID NO: 1.
配列番号: 1 7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 7で表わされるアミノ酸配列と約 7 0 %以上、 好ましくは 約 8 0 %以上、 好ましくは約 9 0 %以上、 さらに好ましくは約 9 5 %以上、 よ り好ましくは約 9 7 %以上の相同性を有するアミノ酸配列などが挙げられる。 配列番号: 1 7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 7で表されるァミノ 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 7で表されるァ ミノ酸配列を有するタンパク質と実質的に同質の活性を有するタンパク質など が好ましい。  The amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 17 includes about 70% or more, preferably about 80% or more, preferably Amino acid sequences having about 90% or more, more preferably about 95% or more, more preferably about 97% or more homology, and the like. Examples of the protein having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 17 include, for example, an amino acid sequence substantially the same as the amino acid sequence represented by the aforementioned SEQ ID NO: 17 And a protein having substantially the same activity as the protein having the amino acid sequence represented by SEQ ID NO: 17 is preferable.
配列番号: 1 9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 9で表わされるアミノ酸配列と約 7 0 %以上、 好ましくは 約 8 0 %以上、 好ましくは約 9 0 %以上、 さらに好ましくは約 9 5 %以上、 よ り好ましくは約 9 7 %以上の相同性を有するァミノ酸配列などが挙げられる。 配列番号: 1 9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 9で表されるァミノ 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 9で表されるァ ミノ酸配列を有するタンパク質と実質的に同質の活性を有するタンパク質など が好ましい。 The amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 19 includes about 70% or more, preferably about 80% or more, preferably Amino acid sequences having a homology of about 90% or more, more preferably about 95% or more, and even more preferably about 97% or more. Examples of the protein having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 19 include, for example, an amino acid sequence substantially the same as the amino acid sequence represented by the aforementioned SEQ ID NO: 19 And a protein having substantially the same activity as the protein having the amino acid sequence represented by SEQ ID NO: 19.
実質的に同質の活性としては、 例えば、 クロライドチャネル活性 (例、 カル シゥム依存性クロライドチャネル活性など) 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、 クロライドチャネル活性が同等 (例、 約 0 . 0 1 〜: L 0 0倍、 好ましくは約 0 . ;!〜 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。  Examples of substantially the same activity include chloride channel activity (eg, calcium-dependent chloride channel activity and the like). Substantially homogenous indicates that the properties are homologous in nature (eg, physiologically or pharmacologically). Therefore, it is preferable that the chloride channel activities are equivalent (eg, about 0.01 to: L 0 times, preferably about 0,0 to 10 times, more preferably 0.5 to 2 times). However, the quantitative factors such as the degree of these activities and the molecular weight of the protein may be different.
クロライドチャネル活性などの活性の測定は、 公知の方法に準じて行うこと が出来、 例えば、 ジエノミクス (Genomics) 、 54巻、 200頁 (1998) 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 activity such as chloride channel activity can be measured according to a known method. For example, the activity can be measured according to the method described in Genomics, Vol. 54, p. 200 (1998) or a method analogous thereto. it can.
また、 本発明のタンパク質としては、 例えば、 (1 ) (a) 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 (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、 (b) 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 (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、 (c) 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 (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、 (d) 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 (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 (e) それらを組み合わせたアミノ酸配列を含有する夕 ンパク質などのいわゆるムテイン、  Examples of the protein of the present invention include (1) (a) one or more (preferably about 1 to 30 and preferably 1 to 30) amino acids in the amino acid sequence represented by SEQ ID NO: 1. About 10 amino acids, more preferably an amino acid sequence in which the number of amino acids (1 to 5) has been deleted, and (b) one or two or more amino acids (preferably About 30 amino acids, preferably about 1-10 amino acids, and more preferably about (1-5) amino acids; (c) the amino acid sequence represented by SEQ ID NO: 1 An amino acid sequence into which one or more (preferably about 1 to 30, preferably about 1 to 10, and more preferably a number (1 to 5)) amino acids have been inserted; ) SEQ ID NO: 1 or 2 or more in the amino acid sequence represented by 1 (preferably about 1 to 30 About 1 to 10 amino acids, more preferably about 1 to 5 amino acids are substituted with another amino acid, or (e) a protein containing an amino acid sequence combining them. So-called mutein,
( 2 ) (a) 配列番号: 1 7で表されるアミノ酸配列中の 1または 2個以上 (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、 (b) 配列番号: 1 7で表 されるアミノ酸配列に 1または 2個以上 (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、 (c) 配列番号: 1 7で表されるアミノ酸配列に 1または 2 個以上 (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、 (d) 配列番 号: 1 7で表されるアミノ酸配列中の 1または 2個以上 (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 (e) それらを組み 合わせたァ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) (a) one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 17 (preferably Preferably, the amino acid sequence has about 1 to 30 amino acids, preferably about 1 to 10 amino acids, more preferably about 1 to 5 amino acids, and (b) SEQ ID NO: 17. An amino acid sequence in which one or more (preferably about 1 to 30, preferably about 1 to 10, and more preferably about 1 to 5) amino acids have been added to the amino acid sequence to be (C) one or more amino acid sequences represented by SEQ ID NO: 17 (preferably about 1 to 30, preferably about 1 to 10, more preferably about 1 to 5 ) Amino acid sequence; (d) one or more (preferably about 1 to 30; preferably 1 to 1) amino acids in the amino acid sequence represented by SEQ ID NO: 17 An amino acid sequence in which about 0, more preferably a number (1 to 5) amino acids have been substituted with another amino acid, or (e So-called mucins, such as proteins containing an amino acid sequence,
( 3 ) (a) 配列番号: 1 9で表されるアミノ酸配列中の 1または 2個以上 (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、 (b) 配列番号: 1 9で表 されるアミノ酸配列に 1または 2個以上 (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、 (c) 配列番号: 1 9で表されるアミノ酸配列に 1または 2 個以上 (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、 (d) 配列番 号: 1 9で表されるアミノ酸配列中の 1または 2個以上 (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 (e) それらを組み 合わせたアミノ酸配列を含有する夕ンパク質などのいわゆるムテインなども含 まれる。  (3) (a) 1 or 2 or more in the amino acid sequence represented by SEQ ID NO: 19 (preferably, about 1 to 30, preferably about 1 to 10, more preferably (B) amino acid sequence represented by SEQ ID NO: 1 or 2 or more (preferably 1 to 30 amino acids, preferably 1 to 5 amino acids); An amino acid sequence having about 1 to 10 amino acids, more preferably (1 to 5) amino acids, and (c) one or two or more amino acids (preferably, an amino acid sequence represented by SEQ ID NO: 19) An amino acid sequence having about 1 to 30 amino acids, preferably about 1 to 10 amino acids, and more preferably a number (1 to 5) of amino acids inserted therein; (d) SEQ ID NO: 19 1 or 2 or more in the amino acid sequence (preferably about 1 to 30, preferably about 1 to 10, more preferably 1 to 5) (E) an amino acid sequence obtained by substituting an amino acid with another amino acid, or (e) so-called mutein such as protein containing an amino acid sequence obtained by combining them.
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 表わ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、 本発明のタン パク質は、 C末端が、 力ルポキシル基 (- C00H) 、 カルボキシレート (- C00— ) 、 アミド (- C0NH2) またはエステル (- C00R) の何れであってもよい。 ここでエステルにおける Rとしては、 例えば、 メチル、 ェチル、 n—プロピ ル、 イソプロピル、 n—ブチルなどの C Mアルキル基、 例えば、 シクロペンチ ル、 シクロへキシルなどの C 3.8シクロアルキル基、 例えば、 フエニル、 α—ナ フチルなどの C 612ァリ一ル基、 例えば、 ベンジル、 フエネチルなどのフエニル — C ,_2アルキル基もしくはひ一ナフチルメチルなどの a—ナフチルー C Mアルキ ル基などの C 7_14ァラルキル基、 ピバ口ィルォキシメチル基などが用いられる。 本発明のタンパク質が C末端以外に力ルポキシル基 (またはカルポキシレ一 ト) を有している場合、 力ルポキシル基がアミド化またはエステル化されてい るものも本発明のタンパク質に含まれる。 この場合のエステルとしては、 例え ば上記した C末端のエステルなどが用いられる。 In the present specification, the protein has a N-terminus at the left end (amino terminus) and a C-terminus at the right end (capillary end) according to the convention of peptide labeling. The protein of the present invention, including the protein containing the amino acid sequence represented by SEQ ID NO: 1, has a C-terminus having a lipoxyl group (-C00H), a carboxylate (-C00-), and an amide (-C0NH). 2 ) or ester (-C00R). Here, as R in the ester, e.g., methyl, Echiru, n- propyl Le, isopropyl, CM alkyl group, such as n- butyl, for example, C 3. 8 cycloalkyl groups cyclopentyl Le, cyclohexane, etc. cyclohexyl, for example, phenyl, alpha-Na C 6 such Fuchiru - 12 § Li Ichiru group, e.g., benzyl, phenyl, such as phenethyl - C, _ 2 alkyl or flight one naphthylmethyl etc. a- Nafuchiru C M such alkyl Le group C 7 _ 14 Ararukiru group, such as pin bar opening Iruokishimechiru group is used. When the protein of the present invention has a lipoxyl group (or carboxylate) other than the C-terminus, the protein of the present invention includes a lipoxyl group amidated or esterified. As the ester in this case, for example, the above-mentioned C-terminal ester and the like are used.
さらに、 本発明のタンパク質には、 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 Hi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口ダル夕ミン酸化したもの、 分子内の アミノ酸の側鎖上の置換基 (例えば - 0H、 -SH、 アミノ基、 イミダゾ一ル基、 ィ 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卜6アル力ノィル基などの C卜6ァシル基など) で保護されているも の、 あるいは糖鎖が結合したいわゆる糖タンパク質などの複合タンパク質など も含まれる。 Furthermore, in the protein of the present invention, the amino group at the N-terminal amino acid residue (eg, methionine residue) is protected by a protecting group (eg, C Hi acetyl group such as alkanol such as formyl group and acetyl group). N-terminal glutamine residue generated by cleavage in vivo, which has been oxidized with lipamine, Substituents on the side chains of amino acids in the molecule (eg, -0H, -SH, amino groups , imidazo Ichiru group, I Ndoru group is protected with Guanijino group, etc.) a suitable protecting group (e.g., formyl group, C WINCH 6 Ashiru groups such as C Bok 6 Al force Noiru group such as § Se methyl group) And complex proteins such as so-called glycoproteins to which sugar chains are bound.
本発明のタンパク質の具体例としては、 例えば、 配列番号: 1で表されるァ ミノ酸配列を含有するタンパク質、 配列番号: 1 7で表されるアミノ酸配列を 含有するタンパク質、 配列番号: 1 9で表されるアミノ酸配列を含有するタン パ 質などがあげられる。  Specific examples of the protein of the present invention include, for example, a protein containing an amino acid sequence represented by SEQ ID NO: 1, a protein containing an amino acid sequence represented by SEQ ID NO: 17, SEQ ID NO: 19 And a protein containing the amino acid sequence represented by
本発明のタンパク質の部分ペプチドとしては、 前記した本発明のタンパク質 の部分ペプチドであって、 好ましくは、 前記した本発明のタンパク質と同様の 性質を有するものであればいずれのものでもよい。 例えば、 本発明のタンパク 質の構成アミノ酸配列のうち 2 0個以上、 好ましくは 5 0個以上、 さらに好ま しくは 7 0個以上、 より好ましくは 1 0 0個以上、 最も好ましくは 2 0 0個以 上のアミノ酸配列を有するペプチドなどが用いられる。 また、 本発明の部分ペプチドは、 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。 The partial peptide of the protein of the present invention is the partial peptide of the protein of the present invention described above, and preferably any peptide having the same properties as the protein of the present invention described above. For example, among the constituent amino acid sequences of the protein of the present invention, 20 or more, preferably 50 or more, more preferably 70 or more, more preferably 100 or more, and most preferably 200 or more Peptides having the above amino acid sequences are used. Further, in the partial peptide of the present invention, one or more (preferably about 1 to 10, more preferably about 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 One or two or more (preferably about 1 to 20, more preferably about 1 to 10, and more preferably a number (1 to 5)) amino acids are inserted into the amino acid sequence, or Even if one or two or more (preferably about 1 to 10, more preferably several, and more preferably about 1 to 5) amino acids in the amino acid sequence are substituted with another amino acid, Good.
また、 本発明の部分ペプチドは C末端が力ルポキシル基 (-C00H) 、 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。 In the partial peptide of the present invention, the C-terminus force Rupokishiru group (-C00H), Karupoki Shireto (-C00-), can be any of the amide (_C0NH 2) or ester (-C00R).
さらに、 本発明の部分ペプチドには、 前記した本発明のタンパク質と同様に、 C末端以外に力ルポキシル基 (またはカルポキシレート) を有しているもの、 N末端のアミノ酸残基 (例、 メチォニン歹考基) 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ダル夕 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ぺプ チドなども含まれる。  Further, the partial peptide of the present invention includes, as in the case of the protein of the present invention described above, a peptide having a carbonyl group (or carboxylate) other than the C-terminal, an N-terminal amino acid residue (eg, methionine). The amino group of the system is protected with a protecting group, the N-terminal is cleaved in vivo, the glutamine residue is oxidized by pyridalamine, and the substituent on the side chain of the amino acid in the molecule is Also included are those protected with an appropriate protecting group, and complex peptides such as so-called glycopeptides to which a sugar chain is bound.
本発明の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。 本発明のタンパク質または部分べ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。  The partial peptide of the present invention can also be used as an antigen for producing an antibody. As the salt of the protein or partial peptide of the present invention, salts with physiologically acceptable acids (eg, inorganic acids, organic acids) and bases (eg, alkali metal salts) are used. Acceptable acid addition salts are preferred. Such salts include, for example, salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid) or organic acids (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).
本発明のタンパク質もしくはその部分べプチドまたはその塩は、 前述したヒ トゃ温血動物の細胞または組織から公知のタンパク質の精製方法によって製造 することもできるし、 タンパク質をコードする D NA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 of the present invention or a partial peptide thereof or a salt thereof can be prepared by the above-mentioned human The protein can be produced from a warm-blooded animal cell or tissue by a known method for purifying a protein, or can be produced by culturing a transformant containing a DNA encoding the protein. It can also be produced according to the peptide synthesis method described below.
ヒトゃ非ヒト哺乳動物の組織または細胞から製造する場合、 ヒトゃ非ヒト哺 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ー、 イオン交換クロマトグラフィーなどのクロマ トグラフィ一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 tissues or cells of a human or non-human mammal, the tissues or cells of a human or non-human mammal are homogenized, extracted with an acid or the like, and the extracted liquid is subjected to reverse phase chromatography and ion exchange. Purification and isolation can be performed by combining chromatography such as chromatography.
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、 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—ベンジルォキシベンジルァ ルコール樹脂、 4一メチルベンズヒドリルァミン樹脂、 P AM樹脂、 4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樹 fl旨、 4— ( 2 ', 4,—ジメトキシフエ二ル一ヒドロキシメチル) フエノキシ樹脂、 4— ( 2,, 4,ージメトキシフエ二ルー F m o cアミノエチル) フエノキシ樹脂 などを挙げることができる。 このような樹脂を用い、 《—ァミノ基と側鎖官能 基を適当に保護したアミノ酸を、 目的とするタンパク質の配列通りに、 公知の 各種縮合方法に従い、 樹脂上で縮合させる。 反応の最後に樹脂からタンパク質 または部分ペプチドを切り出すと同時に各種保護基を除去し、 さらに高希釈溶 液中で分子内ジスルフ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 of the present invention, or a salt thereof, or an amide thereof, a commercially available resin for protein synthesis can be generally used. Examples of such resins include chloromethyl resin, hydroxymethyl resin, benzylhydrylamine resin, aminomethyl resin, 4-benzyloxybenzyl alcohol resin, 4-methylbenzhydrylamine resin, and PAM resin. 4-Hydroxymethylmethylphenylacetamidomethyl resin, polyacrylamide resin, 4- (2 ', 4, -dimethoxyphenyl-hydroxymethyl) phenoxy resin, 4- (2,4, dimethoxyphenyl) Lu Fmoc aminoethyl) phenoxy resin. Using such a resin, an amino acid having an amino group and a side chain functional group appropriately protected is condensed on the resin in accordance with the sequence of the target protein according to various known condensation methods. At the end of the reaction, the protein or partial peptide is cleaved 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 to obtain the target protein or partial peptide or an amide thereof. get.
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。 カルボ ジイミド類としては、 D C C、 N, N'—ジイソプロピルカルポジイミド、 N— ェチルー 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、 H O B t、 H〇〇 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〇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 activating reagents that can be used for protein synthesis can be used, and carbodiimides are particularly preferable. As the carbodiimides, DCC, N, N'-diisopropylcarbodiimide, N-ethyl-N, 1- (3-dimethylaminoprolyl) carbodiimide and the like are used. Activation by these involves adding the protected amino acid directly to the resin along with a racemization inhibitor additive (eg, HOBt, H〇〇Bt), or by adding a symmetrical anhydride or HOBt ester or H〇OB. Pre-protected amino as t-ester The acid can be added to the resin after activation.
保護アミノ酸の活性化ゃ榭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、 塩ィ匕メチレン, 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、 ァセトニトリル, プロピオ二トリルなどの二トリル類、 酢 酸メチル, 酢酸ェチルなどのエステル類あるいはこれらの適宜の混合物などが 用いられる。 反応温度はタンパク質結合形成反応に使用され得ることが知られ ている範囲から適宜選択され、 通常約一 2 0 °C〜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。 二 ンヒドリン反応を用いたテス卜の結果、 縮合が不十分な場合には保護基の脱離 を行うことなく縮合反応を繰り返すことにより十分な縮合を行うことができる c 反応を繰り返しても十分な縮合が得られないときには、 無水酢酸またはァセチ ルイミダゾ一ルを用いて未反応ァ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 the condensation of the protected amino acid with the activated 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-dimethylacetoamide, N-methylpyrrolidone, halogenated hydrocarbons such as methylene chloride, methylform, and trifluoromethyl Alcohols such as ethanol, sulfoxides such as dimethyl sulfoxide, ethers such as pyridine, dioxane, and tetrahydrofuran; nitriles such as acetonitrile and propionitrile; esters such as methyl acetate and ethyl acetate; or an appropriate mixture thereof. Etc. are used. The reaction temperature is appropriately selected from the range known to be usable for the protein bond formation reaction, and is usually selected from the range of about 120 ° C to 50 ° C. The activated amino acid derivative is usually used in a 1.5 to 4-fold excess. Test Bok results with two Nhidorin reaction, condensation is also sufficient Repeat c reactions which can perform sufficient condensation by repeating the condensation reaction without removal of the protecting group to be insufficient When condensation cannot be obtained, acetylation of unreacted amino acid using acetic anhydride or acetylimidazole can prevent the subsequent reaction from being affected.
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、 ィソボル二ルォキシカルポニル、 4ーメトキシベンジルォキシ カルボニル、 C 1 一 Z、 B r— Z、 ァダマンチルォキシカルポニル、 トリフル ォロアセチル、 フタロイル、 ホルミル、 2—ニトロフエニルスルフエニル、 ジ フエニルホスフイノチオイル、 Fm o c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, isoboriloxycarbonyl, 4-methoxybenzyloxycarbonyl, C1-1Z, Br—Z, and adamantylo. Xycarponyl, trifluoroacetyl, phthaloyl, formyl, 2-nitrophenylsulfenyl, diphenylphosphinothioyl, Fmoc and the like are used.
力ルポキシル基は、 例えば、 アルキルエステル化 (例えば、 メチル、 ェチル、 プロピル、 ブチル、 tーブチル、 シクロペンチル、 シクロへキシル、 シクロへ プチル、 シクロォクチル、 2—ァダマンチルなどの直鎖状、 分枝状もしくは環 状アルキルエステル化) 、 ァラルキルエステル化 (例えば、 ベンジルエステル、 4一二ト口べンジルエステル、 4ーメトキシベンジルエステル、 4一クロ口べ 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一ブトキシカルボニルヒドラジド化、 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。 The lipoxyl group can be, for example, alkyl esterified (eg, methyl, ethyl, propyl, butyl, t-butyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, 2-adamantyl, etc.) Alkyl esterification), aralkyl esterification (e.g., benzyl ester, 412-methyl benzyl ester, 4-methoxybenzyl ester, 4-methyl benzyl ester, benzhydryl esterification), phenacyl esterification, benzene Ziroxycarponyl hydrazide, t-butoxycarbonyl hydrazide, It can be protected by trityl hydrazide or the like.
セリンの水酸基は、 例えば、 エステル化またはエーテル化によって保護する ことができる。 このエステル化に適する基としては、 例えば、 ァセチル基など の低級 ((^-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、 エトキシカルポニル基などの炭酸から誘導される基などが 用いられる。 また、 エーテル化に適する基としては、 例えば、 ベンジル基、 テ 卜ラヒドロピラニル基、 t—ブチル基などである。  The hydroxyl group of serine can be protected, for example, by esterification or etherification. Suitable groups for this esterification include, for example, lower ((^ -6) alkanoyl groups such as acetyl group, aroyl groups such as benzoyl group, and groups derived from carbonic acid such as benzyloxycarbonyl and ethoxycarbonyl groups. 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、 Bz 1、 C 12- B z l、 2—ニトロベンジル、 B r— Z、 t一ブチルなどが用いられる。 The protecting group of the phenolic hydroxyl group of tyrosine, for example, Bz 1, C 1 2 - B zl, 2- nitrobenzyl, B r- Z, such as t one-butyl is used.
ヒスチジンのイミダゾ一ルの保護基としては、 例えば、 To s、 4ーメトキ シ一 2, 3, 6—トリメチルベンゼンスルホニル、 DNP、 ベンジルォキシメ チル、 Bum、 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-1,2,3,6-trimethylbenzenesulfonyl, DNP, benzyloxymethyl, Bum, Boc, Trt, and Fmoc.
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, 5一トリクロ口フエノール、 2, 4ー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—ヒドロキシフタルイミド、 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、 対応するリン酸ァ ミドが用いられる。  Examples of the activated form of the raw oxypoxy group include, for example, the corresponding acid anhydride, azide, and active ester [alcohol (for example, pen phenol, 2,4,5-trichloro phenol, 2, 4-dinitrophenol, cyanomethyl alcohol, paranitrophenol, HONB, N-hydroxysuccinimide, N-hydroxyphthalimide, and esters with HOB t)]. As the activated amino group of the raw material, for example, a corresponding phosphoramide 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、 トリエ 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 ァ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 20°C〜40°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、 ジメチルスルフィド、 1, 4一ブタンジチオール、 1, 2一エタンジチオール などのようなカチオ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—ジニトロフエニル基はチォフエノール 処理により除去され、 トリブトファンのィンドール保護基として用いられるホ ルミル基は上記の 1, 2一エタンジチオール、 1 , 4—ブタンジチオールなど の存在下の酸処理による脱保護以外に、 希水酸化ナトリウム溶液、 希アンモニ ァ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) the protecting group include, for example, catalytic reduction in a hydrogen stream in the presence of a catalyst such as Pd-black or Pd-carbon, or hydrogen fluoride anhydride, methanesulfonic acid, trifluoromethane, or the like. Acid treatment with sulfonic acid, trifluoroacetic acid or a mixture thereof, base treatment with diisopropylethylamine, triethylamine, piperidine, piperazine, etc., and reduction with sodium in liquid ammonia, etc. Used. The elimination reaction by the above-mentioned acid treatment is generally carried out at a temperature of about 20 ° C to 40 ° C. In the acid treatment, for example, anisol, phenol, thioanisole, methacrylol, paracresol, dimethylsulfide, It is effective to add a cation scavenger such as 1,4-butanedithiol or 1,2-ethanedithiol. Also, histidine imida The 2,4-dinitrophenyl group used as the zole protecting group is removed by thiophenol treatment, and the formyl group used as the indole protecting group for tributofan is removed from the above 1,1,1-ethanedithiol and 1,4-butanedithiol. In addition to deprotection by acid treatment in the presence, it is also removed by alkali treatment with dilute sodium hydroxide solution, dilute ammonia, or 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 protection 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、 アミノ基側にペプチド (タンパク質) 鎖を所望の鎖長まで延ばした後、 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 分ペプチドと C末端の力ルポキシル基の保護基のみを除去したタンパク質また 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 よりすベ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べ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べプチドのァ ミド体を得ることができる。  As another method for obtaining an amide form of a protein or partial peptide, for example, first, amidation and protection of the α-hydroxyl group of the amino acid at the carboxy terminal, and then a peptide (protein) chain having a desired chain length on the amino group side Then, a protein or partial peptide from which only the protecting group of the α-amino group at the の -terminal of the peptide chain is removed and a protein or partial peptide from which only the protecting group of the C-terminal lipoxyl group is removed Then, 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 method, and a desired crude protein or peptide can be obtained. This crude protein or peptide is purified 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、 力ルポキシ末 端アミノ酸の α—力ルポキシル基を所望のアルコール類と縮合しアミノ酸エス テルとした後、 タンパク質またはペプチドのアミド体と同様にして、 所望の夕 ンパク質またはべプチドのエステル体を得ることができる。  In order to obtain an ester of a protein or peptide, for example, after condensing the α-lipoxyl group of the terminal amino acid with a desired alcohol to form an amino acid ester, in the same manner as the amide of a protein or peptide, It is possible to obtain an ester of the desired protein or peptide.
本発明の部分ペプチドまたはそれらの塩は、 公知のペプチドの合成法に従つ て、 あるいは本発明の夕ンパク質を適当なべプチダーゼで切断することによつ 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、 本発明の部分ペプチドを構成 し得る部分べ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 (a) 〜 (e) に記載された方法が挙げられる。 The partial peptide of the present invention or a salt thereof can be produced according to a known peptide synthesis method or by cleaving the protein of the present invention with an appropriate beptidase. 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 remaining peptide is condensed with the partial peptide or amino acid that can constitute the partial peptide of the present invention, and the product is protected. When the compound has a group, the target peptide can be produced by removing the protecting group. Known condensation methods and elimination of protecting groups include, for example, the methods described in the following (a) to (e).
(a) M. Bodanszkyおよび M.A. Ondetti, ペプチド ·シンセシス (Peptide Synthesis) , Interscience Publishers, New York (1966年)  (a) M. Bodanszky and M.A.Ondetti, Peptide Synthesis, Interscience Publishers, New York (1966)
(b) Schroederおよび Luebke、 ザ'ペプチド(The Peptide), Academic Press, New York (1965年)  (b) Schroeder and Luebke, The Peptide, Academic Press, New York (1965)
(c) 泉屋信夫他、 ペプチド合成の基礎と実験、 丸善 (株) (1975年)  (c) Nobuo Izumiya et al., Fundamentals and experiments of peptide synthesis, Maruzen Co., Ltd. (1975)
(d) 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)  (d) Haruaki Yajima and Shunpei Sakakibara, Laboratory of Biochemistry 1, Protein Chemistry IV, 205, (1977)
(e) 矢島治明監修、 続医薬品の開発、 第 14巻、 ペプチド合成、 広川書店 また、 反応後は通常の精製法、 例えば、 溶媒抽出 ·蒸留 ·カラムクロマトグ ラフィ一 ·液体クロマトグラフィー '再結晶などを組み合わせて本発明の部分 ペ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ペプチドが遊 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。  (e) Supervision of Haruaki Yajima, Development of Continuing Pharmaceuticals, Vol. 14, Peptide Synthesis, Hirokawa Shoten Also, after the reaction, conventional purification methods, such as solvent extraction, distillation, column chromatography, liquid chromatography, and liquid chromatography 'recrystallization' The partial peptide of the present invention can be purified and isolated by combining these methods. When the partial peptide obtained by the above method is an isomer, it can be converted into an appropriate salt by a known method or a method analogous thereto, and conversely, when the partial peptide is obtained by a salt, the known method or Can be converted into a free form or another salt by a method according to the above.
本発明の夕ンパク質をコ一ドするポリヌクレオチドとしては、 上記した本発 明のタンパク質をコードする塩基配列 (DNAまたは RNA、 好ましくは DN A) を含有するものであればいかなるものであってもよい。 該ポリヌクレオチ ドとしては、 本発明のタンパク質をコードする DNA、 mRNA等の RNAで あり、 二本鎖であっても、 一本鎖であってもよい。 二本鎖の場合は、 二本鎖 D NA、 二本鎖 RNAまたは DNA: RNAのハイブリッドでもよい。 一本鎖の 場合は、 センス鎖 (すなわち、 コード鎖) であっても、 アンチセンス鎖 (すな わち、 非コード鎖) であってもよい。  The polynucleotide encoding the protein of the present invention may be any polynucleotide containing a nucleotide sequence (DNA or RNA, preferably DNA) encoding the protein of the present invention described above. Is also good. The polynucleotide is RNA such as DNA or mRNA encoding the protein of the present invention, and may be double-stranded or single-stranded. In the case of double-stranded, it may be double-stranded DNA, double-stranded RNA or DNA: RNA hybrid. In the case of a single strand, it may be a sense strand (that is, a coding strand) or an antisense strand (that is, a non-coding strand).
本発明のタンパク質をコードするポリヌクレオチドを用いて、 例えば、 公知 の実験医学増刊 「新 PC Rとその応用」 15(7)、 1997記載の方法またはそれに準 じた方法により、 本発明のタンパク質の mRNAを定量することができる。 本発明のタンパク質をコードする DNAとしては、 前述した本発明のタンパ ク質をコードする塩基配列を含有するものであればいかなるものであってもよ レ^ 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 cDNA、 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。 Using the polynucleotide encoding the protein of the present invention, for example, the method described in the known experimental medicine special edition “New PCR and its Application” 15 (7), 1997, or a method analogous thereto, or mRNA can be quantified. The DNA encoding the protein of the present invention includes the aforementioned protein of the present invention. It may be any as long as it contains a nucleotide sequence encoding a protein. Also, genomic DNA, genomic DNA library, cDNA derived from the above-mentioned cells and tissues, and cells and tissues derived from the above-mentioned cells Any of a cDNA library or synthetic DNA may be used.
ライブラリ一に使用するべクタ一は、 バクテリオファージ、 プラスミド、 コ スミド、 ファ一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 より全 RN Aまたは mRN A画分を調製したものを用いて直接 Reverse Transcriptase Polymerase Chain Reaction (以下、 RT— PCR法と略称す る) によって増幅することもできる。  The vector used for the library may be any of bacteriophage, plasmid, cosmid, phagemid and the like. Alternatively, it can be directly amplified by Reverse Transcriptase Polymerase Chain Reaction (hereinafter abbreviated as RT-PCR method) using a preparation of the entire RNA or mRNA fraction from the above-mentioned cell'tissue.
本発明のタンパク質をコ一ドする DNAとしては、 例えば、 (1) 配列番 号: 2で表される塩基配列を含有する 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ブリダイズする塩基配列を含 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 同質の性質を有するタンパク質をコードする DNA、 (2) 配列番号: 18で 表される塩基配列を含有する DNA、 または配列番号: 18で表される塩基配 列とハイストリンジェントな条件下でハイブリダイズする塩基配列を含有し、 配列番号: 17で表されるアミノ酸配列を含有するタンパク質と実質的に同質 の性質を有するタンパク質をコードする DNA、 (3) 配列番号: 20で表さ れる塩基配列を含有する DNA、 または配列番号: 20で表される塩基配列と ハイストリンジェントな条件下でハイブリダィズする塩基配列を含有し、 配列 番号: 19で表されるアミノ酸配列を含有するタンパク質と実質的に同質の性 質を有するタンパク質をコ一ドする DN 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ハイストリンジェントな条件下で八イブ リダィズできる DNAとしては、 例えば、 配列番号: 2で表される塩基配列と 約 70 %以上、 好ましくは約 80 %以上、 好ましくは約 90 %以上、 さらに好 ましくは約 95%以上、 より好ましくは約 97%以上の相同性を有する塩基配 列を含有する D N Aなどが用いられる。  Examples of the DNA encoding the protein of the present invention include: (1) a DNA containing the nucleotide sequence represented by SEQ ID NO: 2 or a highly stringent DNA with the nucleotide sequence represented by SEQ ID NO: 2 A DNA encoding a protein having a nucleotide sequence that hybridizes under the conditions and having substantially the same properties as a protein having the amino acid sequence represented by SEQ ID NO: 1, (2) SEQ ID NO: 18 A DNA containing the nucleotide sequence represented by SEQ ID NO: 17, or a nucleotide sequence hybridizing under high stringent conditions with the nucleotide sequence represented by SEQ ID NO: 18; A DNA encoding a protein having substantially the same properties as the contained protein, (3) a DNA containing the nucleotide sequence represented by SEQ ID NO: 20, or a nucleotide sequence represented by SEQ ID NO: 20; DNA containing a nucleotide sequence that hybridizes under high stringent conditions and encoding a protein having substantially the same properties as a protein containing the amino acid sequence represented by SEQ ID NO: 19 Any one may be used. Examples of DNA that can be hybridized with the nucleotide sequence represented by SEQ ID NO: 2 under high stringency conditions include, for example, about 70% or more, preferably about 80% or more of the nucleotide sequence represented by SEQ ID NO: 2. For example, DNA containing a base sequence having a homology of about 90% or more, more preferably about 95% or more, and more preferably about 97% or more is used.
配列番号: 18で表される塩基配列とハイストリンジェン卜な条件下でハイ ブリダィズできる DNAとしては、 例えば、 配列番号: 18で表される塩基配 列と約 70%以上、 好ましくは約 80%以上、 好ましくは約 90%以上、 さら に好ましくは約 95%以上、 より好ましくは約 97 %以上の相同性を有する塩 基配列を含有する DNAなどが用いられる。 Examples of the DNA that can hybridize with the base sequence represented by SEQ ID NO: 18 under high stringent conditions include, for example, the base sequence represented by SEQ ID NO: 18 DNA containing a base sequence having about 70% or more, preferably about 80% or more, preferably about 90% or more, more preferably about 95% or more, more preferably about 97% or more with the sequence. Is used.
配列番号: 20で表される塩基配列とハイストリンジェントな条件下でハイ ブリダィズできる DNAとしては、 例えば、 配列番号: 20で表される塩基配 列と約 70%以上、 好ましくは約 80%以上、 好ましくは約 90%以上、 さら に好ましくは約 95%以上、 より好ましくは約 97 %以上の相同性を有する塩 基配列を含有する D N Aなどが用いられる。  Examples of the DNA that can hybridize with the nucleotide sequence represented by SEQ ID NO: 20 under high stringency conditions include, for example, about 70% or more, preferably about 80% or more of the nucleotide sequence represented by SEQ ID NO: 20 DNA containing a base sequence having homology of preferably about 90% or more, more preferably about 95% or more, and more preferably about 97% or more is used.
ハイブリダィゼ一シヨンは、 公知の方法あるいはそれに準じる方法、 例えば、 モレキュラー ·ク口一ニンク (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うことが できる。 また、 市販のライブラリ一を使用する場合、 添付の使用説明書に記載 の方法に従って行うことができる。 より好ましくは、 ハイストリンジェントな 条件に従って行うことができる。  Hybridization is performed according to a known method or a method analogous thereto, for example, a method described in Molecular Cloning 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). be able to. When a commercially available library is used, it can be performed according to the method described in the attached instruction manual. More preferably, it can be performed under high stringency conditions.
ハイストリンジェントな条件とは、 例えば、 ナトリゥム濃度が約 19〜 40 mM、 好ましくは約 19〜2 OmMで、 温度が約 50〜70° (:、 好ましくは約 60〜65°Cの条件を示す。 特に、 ナトリウム濃度が約 19 mMで温度が約 6 5 °Cの場合が最も好ましい。  The high stringent conditions include, for example, a sodium concentration of about 19 to 40 mM, preferably about 19 to 2 OmM, and a temperature of about 50 to 70 ° (:, preferably about 60 to 65 ° C.). In particular, a sodium concentration of about 19 mM and a temperature of about 65 ° C. are most preferable.
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、 配列番号: 17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: 18で表される塩基配列を含有する DNA、 配列番号: 19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: 20で表される塩基配列を含有する DNAなどが用 いられる。 本発明の部分ペプチドをコードする DNAとしては、 前述した本発 明の部分べプチドをコ一ドする塩基配列を含有するものであればいかなるもの であってもよい。 また、 ゲノム DNA、 ゲノム DNAライブラリー、 前記した 細胞'組織由来の cDNA、 前記した細胞 ·組織由来の cDNAライブラリ一、 合成 DN Aのいずれでもよい。 本発明の部分ペプチドをコードする D N Aとしては、 例えば、 配列番号: 2、 配列番号: 1 8または配列番号: 2 0で表される塩基配列を有する D NAの一 部分を有する D NA、 または配列番号: 2、 配列番号: 1 8または配列番号: 2 0で表される塩基配列とハイストリンジェントな条件下でハイブリダイズす る塩基配列を含有し、 配列番号: 1、 配列番号: 1 7または配列番号: 1 9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夕 ンパク質をコードする D N Aの一部分を含有する D N A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 is a DNA containing the base sequence represented by SEQ ID NO: 2, represented by SEQ ID NO: 17 The DNA encoding the protein containing the amino acid sequence represented by SEQ ID NO: 18 includes the DNA containing the base sequence represented by SEQ ID NO: 18, and the DNA encoding the protein containing the amino acid sequence represented by SEQ ID NO: 19 For example, a DNA containing the base sequence represented by SEQ ID NO: 20 is used. As the DNA encoding the partial peptide of the present invention, any DNA may be used as long as it contains the above-described nucleotide sequence encoding the partial peptide of the present invention. Further, it may be any of genomic DNA, genomic DNA library, cDNA derived from the above-mentioned cells and tissues, cDNA library derived from the above-described cells and tissues, and synthetic DNA. Examples of the DNA encoding the partial peptide of the present invention include a DNA having a part of a DNA having a base sequence represented by SEQ ID NO: 2, SEQ ID NO: 18 or SEQ ID NO: 20; SEQ ID NO: 2, SEQ ID NO: 18 or SEQ ID NO: 20 contains a nucleotide sequence that hybridizes under high stringent conditions with the nucleotide sequence represented by SEQ ID NO: 20; SEQ ID NO: 1, SEQ ID NO: 17 or A DNA containing a part of a DNA encoding a protein having substantially the same activity as the protein containing the amino acid sequence represented by SEQ ID NO: 19 is used.
配列番号: 2、 配列番号: 1 8または配列番号: 2 0で表される塩基配列と ハイブリダィズできる D NAは、 前記と同意義を示す。  The DNA hybridizable with the nucleotide sequence represented by SEQ ID NO: 2, SEQ ID NO: 18 or SEQ ID NO: 20 has the same significance as described above.
ハイブリダイゼ一ションの方法およびハイストリンジェン卜な条件は前記と 同様のものが用いられる。  The same hybridization method and high stringency conditions as described above are used.
本発明のタンパク質、 部分ペプチド (以下、 これらをコードする D NA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 D N A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 D NAプ ライマ一を用いて P C R法によって増幅するか、 または適当なベクタ一に組み 込んだ D N Aを本発明のタンパク質の一部あるいは全領域をコードする D N A 断片もしくは合成 D N Aを用いて標識したものとのハイプリダイゼーシヨンに 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 レキユラ一 'クローニング (Molecular Cloning) 2 nd (J. Sambrook et al . , Cold Spr ing Harbor Lab. Press, 1989) に記載の方法などに従って行うことが できる。 また、 市販のライブラリ一を使用する場合、 添付の使用説明書に記載 の方法に従って行うことができる。  Means for cloning DNA that completely encodes the protein or partial peptide of the present invention (hereinafter, in the description of the cloning and expression of DNAs encoding them, these may be simply referred to as the protein of the present invention). Amplification by PCR using a synthetic DNA primer having a part of the nucleotide sequence encoding the protein of the present invention, or DNA integration into an appropriate vector Alternatively, selection can be performed by hybridization with a DNA fragment encoding the entire region or a DNA fragment labeled with a synthetic DNA. Hybridization can be carried out, for example, 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.
D NAの塩基配列の変換は、 P C Rや公知のキット、 例えば、 MutanTM_super Express Km (宝酒造 (株) ) 、 Mutan™- K (宝酒造 (株) ) 等を用いて、 0DA - LA PCR法や Gapped duplex法や Kunkel法等の公知の方法あるいはそれらに準じる方 法に従って行うことができる。 The DNA base sequence can be converted by PCR or a known kit such as Mutan _super Express Km (Takara Shuzo Co., Ltd.) or Mutan ™ -K (Takara Shuzo Co., Ltd.) using the 0DA-LA PCR method. It can be performed according to a known method such as the gapped duplex method or the Kunkel method, or a method analogous thereto.
クローン化されたタンパク質をコードする D N A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、 リンカーを付加したりして使用するこ とができる。 該 DN Αはその 5'末端側に翻訳開始コドンとしての AT Gを有し, また 3'末端側には翻訳終止コドンとしての TAA、 TGAまたは TAGを有し ていてもよい。 これらの翻訳開始コドンや翻訳終止コドンは、 適当な合成 DN A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 may be used as is, or may be digested with restriction enzymes or added with a linker, if desired. Can be. The DNΑ may have ATG as a translation initiation codon at its 5 ′ end, and may have TAA, TGA or TAG as a translation termination codon at its 3 ′ end. These translation initiation codon and translation termination codon can also be added using an appropriate synthetic DNA adapter.
本発明のタンパク質の発現べクタ一は、 例えば、 (ィ) 本発明のタンパク質 をコードする DNAから目的とする DNA断片を切り出し、 (口) 該 DNA断 片を適当な発現べクタ一中のプロモーターの下流に連結することにより製造す ることができる。  The expression vector of the protein of the present invention may be prepared, for example, by (a) cutting out a DNA fragment of interest from DNA encoding the protein of the present invention, and (mouth) converting the DNA fragment into a promoter in an appropriate expression vector. It can be manufactured by connecting downstream of
ベクターとしては、 大腸菌由来のプラスミド (例、 pBR 322, pBR 3 25, pUC 12, pUC 13) 、 枯草菌由来のプラスミド (例、 pUB 1 1 0, TP 5, p C 194) 、 酵母由来プラスミド (例、 p SH 19, p SH 15) 、 λファージなどのバクテリオファージ、 レトロウイルス, ワクシニアゥ ィルス, バキュロウィルスなどの動物ウィルスなどの他、 ρΑ1— 11、 ρΧ Tl、 pRc/CMV、 pRc/RSV、 p c DN A I ZN e oなどが用いら れる。  Examples of the vector include a plasmid derived from Escherichia coli (eg, pBR322, pBR325, pUC12, pUC13), a plasmid derived from Bacillus subtilis (eg, pUB110, TP5, pC194), a plasmid derived from yeast ( For example, pSH19, pSH15), bacteriophages such as phage λ, animal viruses such as retrovirus, vaccinia virus, baculovirus, etc., ρΑ1-11, ρΧTl, pRc / CMV, pRc / RSV, pc DN AI ZNeo is used.
本発明のプロモーターとしては、 遺伝子の発現に用いる宿主に対応して適切 なプロモー夕一であればいかなるものでもよい。 例えば、 動物細胞を宿主とし て用いる場合は、 SRo!プロモ一夕一、 SV40プロモーター、 LTRプロモ 一ター、 CMVプロモーター、 HS V- TKプロモ一夕一などが挙げられる。  The promoter of the present invention may be any promoter as long as it is suitable for the host used for gene expression. For example, when an animal cell is used as a host, SRo! Promoter, SV40 promoter, LTR promoter, CMV promoter, HSV-TK Promoter and the like can be mentioned.
これらのうち、 CMV (サイトメガロウィルス) プロモーター、 SRaプロ モ一夕一などを用いるのが好ましい。 宿主がェシエリヒア属菌である場合は、 t r pプロモータ一、 l a cプロモーター、 r e c Aプロモーター、 XPLプロ モータ一、 l ppプロモーター、 T 7プロモーターなどが、 宿主がバチルス属 菌である場合は、 SP〇 1プロモータ一、 SP02プロモー夕一、 p enPプ 口モータ一など、 宿主が酵母である場合は、 PH05プロモータ一、 PGKプ 口モーター、 GAPプロモーター、 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一、 P 10プロモーターな どが好ましい。 Of these, it is preferable to use the CMV (cytomegalovirus) promoter, the SRa promoter, etc. When the host is Eshierihia genus, when trp promoter one, lac promoter, rec A promoter, XP L pro motor-, l pp promoter, T 7 promoter, the host is Bacillus, SP_〇 1 When the host is yeast, such as a promoter, SP02 promoter, penP promoter, etc., PH05 promoter, PGK promoter, GAP promoter, ADH promoter and the like are preferable. When the host is an insect cell, a polyhedrin promoter, a P10 promoter and the like are preferable.
発現べクタ一には、 以上の他に、 所望によりェンハンサー、 シグナル、 ポリ A付加シグナル、 選択マーカー、 S V40複製オリジン (以下、 SV400 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。 選択マ一力一としては、 例えば、 ジヒドロ葉酸還元酵素 (以下、 d h 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、 Amp1"と略称する場合がある) 、 ネオマイシ ン耐性遺伝子 (以下、 Ne ο1·と略称する場合がある、 G418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一として使用する場合、 目的遺伝子をチミジンが含ま れない培地によっても選択できる。 In addition to the above, the expression vector may include an enhancer, A signal containing a signal, a poly-A addition signal, a selection marker, an SV40 replication origin (hereinafter sometimes abbreviated as SV400ri) and the like can be used. Selection methods include, for example, dihydrofolate reductase (hereinafter sometimes abbreviated as dhfr) gene (methotrexate (MTX) resistance), ampicillin resistance gene (hereinafter sometimes abbreviated as Amp 1 ) And neomycin resistance gene (hereinafter sometimes abbreviated as Ne ο 1 · G418 resistance), etc. Particularly, when the dh fr gene is used as a selection marker using Chinese hamster cells lacking the dh fr gene. Alternatively, the target gene can be selected by using a medium containing no thymidine.
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、 MFa ·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、 α—インタ一フ 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。 このようにして構築された本発明のタンパク質をコードする D Ν Α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。  If necessary, a signal sequence suitable for the host is added to the N-terminal side of the protein of the present invention. When the host is a genus Escherichia, the PhoA * signal sequence and OmpA signal sequence are used.When the host is a Bacillus genus, the human amylase signal sequence and subtilisin signal sequence are used. If the host is an animal cell, an insulin signal sequence, an α-interferon signal sequence, an antibody molecule, a signal sequence, etc. Available for each. A transformant can be produced using the vector containing D ベ ク タ ー which encodes the protein of the present invention thus constructed.
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。  As the host, for example, Escherichia bacteria, Bacillus bacteria, yeast, insect cells, insects, animal cells, and the like are used.
ェシェリヒア属菌の具体例としては、 例えば、 ェシェリヒア ·コリ  Specific examples of the genus Escherichia include, for example, Escherichia coli
(Escherichia coli) K 12 · DH 1 〔プロシ一ジングズ ·ォブ ·ザ ·ナショ ナル ·アカデミー ·ォブ ·サイェンシィズ ·ォブ ·ザ ·ュ一エスエー (Proc. Natl. Acad. Sci. USA) , 60巻, 160 (1968)〕 , JM103 〔ヌクイ レック ·ァシッズ ' リサーチ (Nucleic Acids Research) , 9巻, 309 (19 81)〕 , J A221 〔ジャーナル ·ォブ ·モレキュラー ·バイオロジー  (Escherichia coli) K12 · DH 1 [Proc. Natl. Acad. Sci. USA], 60 Vol. 160 (1968)], JM103 [Nucleic Acids Research ', Vol. 9, 309 (1981)], JA221 [Journal of Molecular Biology]
(Journal of Molecular Biology) , 120卷, 517 (1978)〕 , ΗΒ 1 01 〔ジャーナル ·ォブ ·モレキュラー ·バイオロジー, 41卷, 459 (19 69)〕 , C 600 〔ジエネティックス (Genetics) , 39巻, 440 (195 4)〕 などが用いられる。 (Journal of Molecular Biology), 120 volumes, 517 (1978)], ΗΒ101 [Journal of Molecular Biology, 41 volumes, 459 (1969)], C 600 [Genetics, 39 volumes] , 440 (195 4)] is used.
バチルス属菌としては、 例えば、 バチルス ·サブチルス (Bacillus subtilis) M I 1 14 〔ジーン, 24巻, 255 (1983)〕 , 207— 21 〔ジャーナル 'ォブ 'バイオケミストリ— (Journal of Biochemistry) , 95 巻, 87 (1984)〕 などが用いられる。  Examples of Bacillus bacteria include, for example, Bacillus subtilis MI 114 [Gene, 24, 255 (1983)], 207-21 [Journal of Biochemistry, 95] , 87 (1984)].
酵母としては、 例えば、 サッカロマイセス ·セレビシェ (Saccharomyces cerevisiae) AH22, AH 22 R -, NA87- 11 A, DKD- 5 D, 20 B - 12 シゾサッカロマイセス 'ボンべ (Schizosaccharomyces pombe) NC YC 1913, NCYC 2036, ピキア -パストリス (Pichia pastoris) K M71などが用いられる。  Examples of yeast include, for example, Saccharomyces cerevisiae AH22, AH22R-, NA87-11A, DKD-5D, 20B-12 Shizosaccharomyces' bomb (Schizosaccharomyces pombe) NC YC 1913, NCYC 2036 , Pichia pastoris K M71 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 MGl細胞、 Trichoplusia niの卵由来の High Five™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。 ゥ ィルスが BmNPVの場合は、 蚕由来株化細胞 (Bombyx mori N細胞; BmN 細胞) などが用いられる。 該 S f細胞としては、 例えば、 S f 9細胞 (ATCC CRL1711) 、 S f 21細胞 (以上、 Vaughn, J.L.ら、 イン ·ヴィポ (In  As insect cells, for example, when the virus is Ac NPV, a cell line derived from a larva of night moth (Spodoptera frugiperda cell; Sf cell), MGl cell derived from the midgut of Trichoplusia ni, High derived from egg of Trichoplusia ni Five ™ cells, cells derived from Mamestra brassicae or cells derived from Estigmena acrea are used. When the virus is BmNPV, a silkworm-derived cell line (Bombyx mori N cell; BmN cell) or the like is used. Examples of the Sf cells include Sf9 cells (ATCC CRL1711), Sf21 cells (Vaughn, J.L., et al., In Vipo).
Vivo) ,13, 213-217, (1977)) などが用いられる。 Vivo), 13, 213-217, (1977)).
昆虫としては、 例えば、 カイコの幼虫などが用いられる 〔前田ら、 ネィチヤ 一 (Nature) , 315巻, 592 (1985)〕 。  As insects, for example, silkworm larvae are used [Maeda et al., Nature, 315, 592 (1985)].
動物細胞としては、 例えば、 サル細胞 COS— 7, Ve r o, チャイニーズ ハムス夕一細胞 CHO' (以下、 CHO細胞と略記) , dh f r遺伝子欠損チヤ ィニーズ八ムスター細胞 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, ラット GH3, ヒ ト F L細胞などが用いられる。  Examples of animal cells include monkey cell COS-7, Vero, Chinese Hams Yuichi cell CHO '(hereinafter abbreviated as CHO cell), dh fr gene-deficient Chinese eight-muster cell CHO (hereinafter CHO (dh fr- ) Cells), mouse L cells, mouse AtT-20, mouse myeloma cells, rat GH3, and human FL cells.
ェシエリヒア属菌を形質転換するには、 例えば、 プロシ一ジングズ'ォブ' ザ ·ナショナル ·アカデミー ·ォブ ·サイェンジィズ ·ォブ ·ザ ·ュ一エスェ 一 (Proc. Natl. Acad. Sci. USA) , 69巻, 21 10 (1972)やジーン (Gene) , 17巻, 107 (1982)などに記載の方法に従って行うことがで さる。 For example, to transform a microorganism belonging to the genus Escherichia, for example, Proc. Natl. Acad. Sci. USA, Proc. Natl. Acad. Sci. USA , 69, 2110 (1972) and Gene, 17, 107 (1982). Monkey
バチルス属菌を形質転換するには、 例えば、 モレキュラー ·アンド ·ジエネ ラル'ジェネティックス (Molecular & General Genetics) , 168巻, 1 1 1 (1979)などに記載の方法に従って行うことができる。  Transformation of Bacillus can be performed, for example, according to the method described in Molecular & General Genetics, Vol. 168, 11 (1979).
酵母を形質転換するには、 例えば、 メソッズ ·イン ·ェンザィモ口ジー To transform yeast, for example, the Method in Enzymo
(Methods in Enzymology) , 194巻, 182 - 187 (1991) 、 プロシ —ジングズ ·ォブ ·ザ ·ナショナル ·アカデミー ·ォブ ·サイェンシィズ ·ォ ブ 'ザ ·ュ一エスェ一 (Proc. Natl. Acad. Sci. USA) , 75巻, 1929 (1 978) などに記載の方法に従って行うことができる。 (Methods in Enzymology), Vol. 194, 182-187 (1991), Proc. Natl. Acad., "The Processing of the National Academy of the National Academy of Sciences" Sci. USA), Vol. 75, 1929 (1 978).
昆虫細胞または昆虫を形質転換するには、 例えば、 バイオ/テクノロジー To transform insect cells or insects, e.g., bio / technology
(Bio/Technology) ,6, 47-55 (1988) などに記載の方法に従って行うことがで さる。 (Bio / Technology), 6, 47-55 (1988).
動物細胞を形質転換するには、 例えば、 細胞工学別冊 8 新細胞工学実験プ 口トコ一ル. 263 - 267 (1995) (秀潤社発行) 、 ヴイロロジー (Virology) , 52巻, 456 (1973 )に記載の方法に従って行うことがで きる。  To transform animal cells, for example, see Cell Engineering Separate Volume 8 New Cell Engineering Experiment Protocol. 263-267 (1995) (published by Shujunsha), Virology, 52, 456 (1973). The method can be performed according to the method described in.
このようにして、 タンパク質をコードする DN Aを含有する発現べクタ一で 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.
宿主がェシエリヒア属菌、 バチルス属菌である形質転換体を培養する際、 培 養に使用される培地としては液体培地が適当であり、 その中には該形質転換体 の生育に必要な炭素源、 窒素源、 無機物その他が含有せしめられる。 炭素源と しては、 例えば、 グルコース、 デキストリン、 可溶性澱粉、 ショ糖など、 窒素 源としては、 例えば、 アンモニゥム塩類、 硝酸塩類、 コーンスチープ · リカ一 ペプトン、 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げられる。 また、 酵母エキス、 ビタミン類、 生 長促進因子などを添加してもよい。 培地の PHは約 5〜8が望ましい。  When culturing a transformant whose host is a bacterium belonging to the genus Escherichia or Bacillus, a liquid medium is suitable as a medium used for culturing, and a carbon source necessary for the growth of the transformant is contained therein. , Nitrogen sources, inorganic substances and others. Examples of carbon sources include glucose, dextrin, soluble starch, and sucrose.Examples of nitrogen sources include ammonium salts, nitrates, corncheap, lica-peptone, casein, meat extract, soybean meal, and potato extract. Examples of the inorganic or organic substance such as a liquid and the inorganic substance include calcium chloride, sodium dihydrogen phosphate, and magnesium chloride. In addition, yeast extract, vitamins, growth promoting factors and the like may be added. The pH of the medium is preferably about 5 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 〔ミラー (Miller) , ジャーナル ·ォブ ·ェクスぺリメ ンッ 'イン 'モレキュラー 'ジェネティックス (Journal of Experiments in Molecular Genetics) , 431— 433, Cold Spring Harbor Laboratory, New York 1972) が好ましい。 ここに必要によりプロモータ一を効率よく働 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。 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 ed. Preferred is "in 'molecular" Genetics (Journal of Experiments in Molecular Genetics), 431-433, Cold Spring Harbor Laboratory, New York 1972). If necessary, a drug such as 3) 3-indolylacrylic acid can be added to make the promoter work efficiently if necessary.
宿主がェシエリヒア属菌の場合、 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。  When the host is a bacterium belonging to the genus Escherichia, culturing is usually performed at about 15 to 43 ° C for about 3 to 24 hours, and if necessary, aeration and stirring may be added.
宿主がバチルス属菌の場合、 培養は通常約 30〜40°Cで約 6〜24時間行 ない、 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 ° C for about 6 to 24 hours, and if necessary, aeration and stirring may be added.
宿主が酵母である形質転換体を培養する際、 培地としては、 例えば、 バーク ホ一ルダー (Burkholder) 最小培地 〔Bostian, K. L. ら、 プロシ一ジングズ- ォブ ·ザ ·ナショナル ·アカデミー ·ォブ ·サイェンシィズ ·ォブ ·ザ ·ユー エスェ一 (Proc. Natl. Acad. Sci. USA) , 77巻, 4505 (1980)〕 や 0.5%カザミノ酸を含有する SD培地 (Bitter, G. A. ら、 プロ'シ一ジング ズ ·ォブ ·ザ ·ナショナル ·アカデミー■ォブ ·サイェンシィズ ·ォブ ·ザ · ユーエスエー (Proc. Natl. Acad. Sci. USA) , 81巻, 5330 (198 4) 〕 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〜 35 °Cで約 24〜 72時間行ない、 必要に応じて通気や撹拌を加 える。  When culturing a transformant whose host is yeast, for example, Burkholder's minimal medium [Bostian, KL et al., Processings-of-the-National Academy of Cultures] Proc. Natl. Acad. Sci. USA, 77, 4505 (1980)] and an SD medium containing 0.5% casamino acid (Bitter, GA et al., Proc. Zing's of the National Academy of Sciences, of the USA (Proc. Natl. Acad. Sci. USA), 81, 5330 (198 4)]. The pH is preferably adjusted to about 5 to 8. Culture is usually performed at about 20 to 35 ° C for about 24 to 72 hours, and aeration and stirring are added as necessary.
宿主が昆虫細胞または昆虫である形質転換体を培養する際、 培地としては、 Grace's Insect Medium (Grace, T. C.,ネィチヤ一  When culturing a transformant in which the host is an insect cell or an insect, the medium used is Grace's Insect Medium (Grace, T.C.,
(Nature) ,195, 788 (1962)) に非動化した 10 %ゥシ血清等の添加物を適宜カロ 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。  (Nature), 195, 788 (1962)), an immobilized additive such as 10% pepsin serum or the like is used as needed. Preferably, the pH of the medium is adjusted to about 6, 2 to 6.4. Culture is usually performed at about 27 ° C for about 3 to 5 days, and aeration and agitation are added as necessary.
宿主が動物細胞である形質転換体を培養する際、 培地としては、 例えば、 約 5〜20 %の胎児牛血清を含む MEM培地 〔サイエンス (Science) , 122巻, When culturing a transformant in which the host is an animal cell, the medium may be, for example, a MEM medium containing about 5 to 20% fetal bovine serum [Science, 122,
501 (1952)〕 , DMEM培地 〔ヴイロロジ一 (Virology) , 8巻, 39501 (1952)], DMEM medium [Virology, 8, 39
6 (1959)] , RPM I 1640培地 〔ジャーナル ·ォブ 'ザ.アメリカ ン 'メディカル'アソシエーション (The Journal of the American Medical Association) 199卷, 5 19 (1967)〕 , 1 99培地 〔プロシージング- ォブ 'ザ'ソサイエティ ·フォー ·ザ'バイオロジカル ·メディスン 6 (1959)], RPM I 1640 medium [Journal of the America. 'The Medical' Association (The Journal of the American Medical Association) 199, May 19 (1967)], 199 Medium [Proceding-The 'The Society for the Biological Medicine'
(Proceeding of the Society for the Biological Medicine) , 73巻, 1 (1950)〕 などが用いられる。 pHは約 6〜8であるのが好ましい。 培養は 通常約 30〜 40。Cで約 15〜 60時間行ない、 必要に応じて通気や撹拌を加 える。  (Proceeding of the Society for the Biological Medicine), Vol. 73, 1 (1950)]. Preferably, the pH is about 6-8. Culture is usually about 30-40. Perform at C for about 15-60 hours, adding aeration and agitation as needed.
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夕 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 cells, cell membranes or extracellular 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、 リゾチ一ムおよび 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タンパク質変性剤や、 トリトン X— 100TMなどの界面活性剤が含まれていてもよい。 培養液中に夕 ンパク質が分泌される場合には、 培養終了後、 公知の方法で菌体あるいは細胞 と上清とを分離し、 上清を集める。 When extracting the protein of the present invention from cultured cells or cells, after culturing, the cells or cells are collected by a known method, suspended in an appropriate buffer, and then subjected to ultrasound, lysozyme and Z or freezing. After the cells or cells are disrupted by thawing or the like, a method of obtaining a crude protein extract by centrifugation or filtration is appropriately used. The buffer may contain a protein denaturant such as urea or guanidine hydrochloride, or a surfactant such as Triton X-100 . If the protein is secreted into the culture, after the culture is completed, the supernatant is separated from the cells or cells by a known method, and the supernatant is collected.
このようにして得られた培養上清、 あるいは抽出液中に含まれるタンパク質 の精製は、 公知の分離 ·精製法を適切に組み合わせて行うことができる。 これ らの公知の分離、 精製法としては、 塩析ゃ溶媒沈澱法などの溶解度を利用する 方法、 透析法、 限外ろ過法、 ゲルろ過法、 および SDS—ポリアクリルアミド ゲル電気泳動法などの主として分子量の差を利用する方法、 イオン交換クロマ トグラフィー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 ィ一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。  The protein contained in the culture supernatant or the extract obtained in this manner can be purified by appropriately combining known separation and purification methods. These known separation and purification methods include methods utilizing solubility such as salting out and solvent precipitation, dialysis, ultrafiltration, gel filtration, and SDS-polyacrylamide gel electrophoresis. Methods using molecular weight differences, methods using charge differences such as ion-exchange chromatography, methods using specific affinity such as affinity chromatography, reverse-phase high-performance liquid chromatography, etc. A method utilizing a difference in hydrophobicity, a method utilizing a difference in isoelectric point such as isoelectric focusing, and the like are used.
このようにして得られるタンパク質が遊離体で得られた場合には、 公知の方 法あるいはそれに準じる方法によって塩に変換することができ、 逆に塩で得ら れた場合には公知の方法あるいはそれに準じる方法により、 遊離体または他の 塩に変換することができる。 If the protein thus obtained is obtained in a free form, The salt can be converted into a salt by a method or a method analogous thereto, and when the salt is obtained, it can be converted into a free form or another salt by a known method or a method analogous thereto.
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タン パク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。 タンパク修飾酵素としては、 例えば、 トリ プシン、 キモトリブシン、 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 applying an appropriate protein modifying enzyme before or after purification. As the protein modifying enzyme, for example, trypsin, chymotrypsin, arginyl endopeptidase, proteinase, glycosidase and the like are used.
このように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 ンザィムィムノアッセィやウェスタンプロッティングなどにより測定すること ができる。  The presence of the protein of the present invention thus produced can be measured by enzyme immunoassay using a specific antibody, Western plotting, or the like.
本発明のタンパク質もしくは部分ペプチドまたはその塩に対する抗体は、 本 発明の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、 モノ夕口一ナル抗体の何れであってもよい。  An antibody against the protein or partial peptide of the present invention or a salt thereof may be any of a polyclonal antibody and a monoclonal mononal antibody as long as it can recognize the protein or partial peptide of the present invention or a salt thereof.
本発明のタンパク質もしくは部分ペプチドまたはその塩 (以下、 抗体の説明 においては、 これらを単に本発明のタンパク質と略記する場合がある) に対す る抗体は、 本発明のタンパク質を抗原として用い、 公知の抗体または抗血清の 製造法に従つて製造することができる。  An antibody against the protein or partial peptide of the present invention or a salt thereof (hereinafter, these may be simply abbreviated as the protein of the present invention in the description of the antibody) uses the protein of the present invention as an antigen, and is a known antibody. It can be produced according to a method for producing an antibody or antiserum.
〔モノクローナル抗体の作製〕  [Preparation of monoclonal antibody]
( a ) モノクローナル抗体産生細胞の作製  (a) Preparation of monoclonal antibody-producing cells
本発明のタンパク質は、 温血動物に対して投与により抗体産生が可能な部位 にそれ自体あるいは担体、 希釈剤とともに投与される。 投与に際して抗体産生 能を高めるため、 完全フロイントアジュバントゃ不完全フロイントアジュバン トを投与してもよい。 投与は通常 2〜 6週毎に 1回ずつ、 計 2〜 1 0回程度行 われる。 用いられる温血動物としては、 例えば、 サル、 ゥサギ、 ィヌ、 モルモ ット、 マウス、 ラット、 ヒッジ、 ャギ、 ニヮトリが挙げられるが、 マウスおよ びラットが好ましく用いられる。  The protein of the present invention is administered to a warm-blooded animal at a site capable of producing an antibody upon administration, itself or together with a carrier or diluent. Complete Freund's adjuvant or incomplete Freund's adjuvant may be administered in order to enhance antibody production upon administration. The administration is usually performed once every 2 to 6 weeks, for a total of about 2 to 10 times. Examples of the warm-blooded animal 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 fl蔵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うことができる。 融合操作は既知の方法、 例えば、 ケ —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 —ル (PEG) やセンダイウィルスなどが挙げられるが、 好ましくほ PEGが 用いられる。 When producing monoclonal antibody-producing cells, select a warm-blooded animal immunized with the antigen, for example, an individual with an antibody titer from a mouse, and spleen 2 to 5 days after the final immunization. A monoclonal antibody-producing hybridoma can be prepared by collecting fl cells or lymph nodes and fusing the antibody-producing cells contained therein with myeloma cells of the same or different species. 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 carried out according to known methods, for example, the method of Köhler and Milstein (Nature, 256, 495 (1975)). Examples of the fusion promoter include polyethylene glycol (PEG) and Sendai virus, and 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 : :!〜 20 : 1程度であり、 PEG (好ましくは PEG 1000〜PEG6 000) が 10〜 80 %程度の濃度で添加され、 20〜40 、 好ましくは 3 0〜37°Cで 1〜10分間インキュベートすることにより効率よく細胞融合を 実施できる。  Examples of myeloma cells include myeloma cells of warm-blooded animals such as NS-1, P3U1, SP 2/0, and AP-1. P3U1 is preferably used. <Antibody-producing cells used ( The preferred ratio of the number of spleen cells to the number of myeloma cells is about 1 ::! To 20: 1, and PEG (preferably PEG 1000 to PEG6 000) is added at a concentration of about 10 to 80%. Incubation at 40, preferably 30 to 37 ° C for 1 to 10 minutes allows efficient cell fusion.
モノクローナル抗体産生ハイプリド一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ブリン抗体 (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 ティン Aを加え、 固相に結合したモノクローナル抗体を検出する方法、 抗免疫 グロプリン抗体またはプロテイン Aを吸着させた固相にハイプリドーマ培養上 清を添加し、 放射性物質や酵素などで標識したタンパク質を加え、 固相に結合 したモノクロ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, microplate) on which a protein antigen is adsorbed directly or together with a carrier, and then An 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 is added to the monoclonal antibody bound to the solid phase. Detection method, Anti-immunoglobulin antibody or protein A is adsorbed on a solid phase to which a hybridoma culture supernatant is added, and a protein labeled with a radioactive substance or an enzyme is added, and a monoclonal antibody bound to the solid phase is detected. And the like.
モノクローナル抗体の選別は、 公知あるいはそれに準じる方法に従って行う 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うことができる。 選別および育種用培地としては、 ハイプリドーマが生育できるものならばどのような培地を用いても良い。 例え ば、 1〜2 0 %、 好ましくは 1 0〜2 0 %の牛胎児血清を含む R P M I 1 6 4 0培地、 1〜1 0 %の牛胎児血清を含む G I T培地 (和光純薬工業 (株) ) あ るいはハイブリド一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 %炭酸ガス下で行うことができる。 ハイプリドーマ培養上 清の抗体価は、 上記の抗血清中の抗体価の測定と同様にして測定できる。 The selection of the monoclonal antibody can be performed according to a known method or a method analogous thereto. Usually, it can be performed in a medium for animal cells supplemented with HAT (hypoxanthine, aminopterin, thymidine). As a selection and breeding medium, any medium can be used as long as it can grow a hybridoma. example For example, RPMI 1640 medium containing 1 to 20%, preferably 10 to 20% fetal bovine serum, and GIT medium containing 1 to 10% fetal bovine serum (Wako Pure Chemical Industries, Ltd.) Alternatively, a serum-free culture medium for hybridoma culture (SFM-101, Nissui Pharmaceutical Co., Ltd.) can be used. The cultivation temperature is usually from 20 to 40 ° (:, preferably, about 37 ° C. The culturing time is usually from 5 days to 3 weeks, preferably from 1 week to 2 weeks. The antibody titer of the hybridoma culture supernatant can be measured in the same manner as the measurement of the antibody titer in the antiserum described above.
( b ) モノクローナル抗体の精製  (b) Purification of monoclonal antibody
モノクローナル抗体の分離精製は、 公知の方法、 例えば、 免疫グロブリンの 分離精製法 〔例、 塩析法、 アルコール沈殿法、 等電点沈殿法、 電気泳動法、 ィ 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あるいはプロティン 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〕 に従って行うこ とができる。  Monoclonal antibodies can be separated and purified by known methods, for example, immunoglobulin separation and purification methods (eg, salting out method, alcohol precipitation method, isoelectric point precipitation method, electrophoresis method, ion exchanger (eg, DEAE)). Adsorption / desorption method, ultracentrifugation method, gel filtration method, antigen-binding solid phase or specific purification method in which only the antibody is collected using an active adsorbent such as protein A or protein G and the bond is dissociated to obtain the antibody) It can be carried out.
〔ポリク口一ナル抗体の作製〕  (Preparation of polyclonal antibody)
本発明のポリクロ一ナル坊体は、 公知あるいはそれに準じる方法に従って製 造することができる。 例えば、 免疫抗原 (タンパク質抗原) 自体、 あるいはそ れとキャリアータンパク質との複合体をつくり、 上記のモノクローナル抗体の 製造法と同様に温血動物に免疫を行ない、 該免疫動物から本発明のタンパク質 に対する抗体含有物を採取して、 抗体の分離精製を行うことにより製造するこ とができる。  The polyclonal body of the present invention can be manufactured according to a known method or a method analogous thereto. For example, a immunizing antigen (protein antigen) itself or a complex thereof with a carrier protein is formed, and a warm-blooded animal is immunized in the same manner as in the above-described method for producing a monoclonal antibody. It can be produced by collecting an antibody-containing substance and separating and purifying the antibody.
温血動物を免疫するために用いられる免疫抗原とキャリア一タンパク質との 複合体に関し、 キャリアータンパク質の種類およびキャリア一とハプテンとの 混合比は、 キヤリァ一に架橋させて免疫したハプテンに対して抗体が効率良く できれば、 どの様なものをどの様な比率で架橋させてもよいが、 例えば、 ゥシ 血清アルブミンゃゥシサイログロプリン、 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の割合で力プルさせる方法が 用いられる。  Regarding the complex of immunizing antigen and carrier-1 protein used to immunize warm-blooded animals, the type of carrier protein and the mixing ratio between carrier-1 and hapten are determined by the antibody against hapten immunized by cross-linking with carrier. As long as it can be efficiently carried out, any kind may be cross-linked at any ratio. For example, serum serum albumin, psiloglopurine, hemocyanin, etc. are used in a weight ratio of about 0.1 to 1 for hapten. A method of pulling at a rate of about 20 to 20 and 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、 グルタルアルデヒドやカルポジイミド、 マレイミド活性エステル、 チオール基、 ジチオビリジル基を含有する活性エステル試薬等が用いられる。 縮合生成物は、 温血動物に対して、 抗体産生が可能な部位にそれ自体あるい は担体、 希釈剤とともに投与される。 投与に際して抗体産生能を高めるため、 完全フロイントアジュバントゃ不完全フロイントアジュバントを投与してもよ レ^ 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。 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。 Also, use various condensing agents for the force coupling between the hapten and the carrier. Glutaraldehyde, carbodiimide, maleimide active ester, an active ester reagent containing a thiol group or a dithioviridyl group, etc. are used. The condensation product is administered to a warm-blooded animal at a site where antibody production is possible or together with a carrier or diluent. Complete Freund's adjuvant or incomplete Freund's adjuvant may be administered to increase antibody production ability during administration.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 the blood, ascites, etc., preferably from the blood of a warm-blooded animal immunized by the above method.
抗血清中のポリクローナル抗体価の測定は、 上記の抗血清中の抗体価の測定 と同様にして測定できる。 ポリクローナル抗体の分離精製は、 上記のモノクロ The measurement of the polyclonal antibody titer in the antiserum can be performed in the same manner as the measurement of the antibody titer in the antiserum described above. Separation and purification of polyclonal antibodies
—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うこと ができる。 —It can be performed according to the same immunoglobulin separation and purification method as that of the null antibody.
本発明のタンパク質または部分ペプチドをコードする D 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オチドとしては、 本発明 の D N Aの塩基配列に相補的な、 または実質的に相補的な塩基配列またはその 一部を有し、 該 D N Aの発現を抑制し得る作用を有するものであれば、 いずれ のアンチセンスポリヌクレオチドであってもよいが、 アンチセンス D N Aが好 ましい。  Complementary to the nucleotide sequence of a DNA encoding the protein or partial peptide of the present invention (hereinafter, these DNAs may be abbreviated as the DNA of the present invention in the description of the antisense polynucleotide), or The antisense polynucleotide having a substantially complementary base sequence or a part thereof includes a base sequence complementary to or substantially complementary to the base sequence of the DNA of the present invention or a part thereof, Any antisense polynucleotide may be used as long as it has an action of suppressing the expression of the DNA, but antisense DNA is preferable.
本発明の D NAに実質的に相補的な塩基配列とは、 例えば、 本発明の D NA に相補的な塩基配列 (すなわち、 本発明の D NAの相補鎖) の全塩基配列ある いは部分塩基配列と約 7 0 %以上、 好ましくは約 8 0 %以上、 より好ましくは 約 9 0 %以上、 最も好ましくは約 9 5 %以上の相同性を有する塩基配列などが 挙げられる。 特に、 本発明の D N Aの相補鎖の全塩基配列うち、 本発明のタン パク質の N末端部位をコードする部分の塩基配列 (例えば、 開始コドン付近の 塩基配列など) の相補鎖と約 7 0 %以上、 好ましくは約 8 0 %以上、 より好ま しくは約 9 0 %以上、 最も好ましくは約 9 5 %以上の相同性を有するアンチセ ンスポリヌクレオチドが好適である。 具体的には、 配列番号: 2、 配列番号: 1 8または配列番号: 2 0で表わさ れる塩基配列を有する D N Aの塩基配列に相補的な、 もしくは実質的に相補的 な塩基配列、 またはその一部分を有するアンチセンスポリヌクレオチド、 好ま しくは例えば、 配列番号: 2、 配列番号: 1 8または配列番号: 2 0で表わさ れる塩基配列を有する D N Aの塩基配列に相補な塩基配列、 またはその一部分 を有する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、 配 列番号: 1 8または配列番号: 2 0で表わされる塩基配列を有する D NAの塩 基配列に相補な塩基配列、 またはその一部分を有するアンチセンスポリヌクレ ォチド) などが挙げられる。 The nucleotide sequence substantially complementary to the DNA of the present invention is, for example, the entire nucleotide sequence or a part of the nucleotide sequence complementary to the DNA of the present invention (that is, the complementary strand of the DNA of the present invention). A base sequence having about 70% or more, preferably about 80% or more, more preferably about 90% or more, and most preferably about 95% or more homology with the base sequence is exemplified. In particular, of the entire nucleotide sequence of the complementary strand of the DNA of the present invention, the complementary sequence of the nucleotide sequence of the portion encoding the N-terminal portion of the protein of the present invention (for example, the nucleotide sequence near the start codon) is approximately 70%. Antisense polynucleotides having a homology of at least about 80%, preferably at least about 80%, more preferably at least about 90%, and most preferably at least about 95% are suitable. Specifically, a nucleotide sequence complementary to or substantially complementary to the nucleotide sequence of DNA having the nucleotide sequence represented by SEQ ID NO: 2, SEQ ID NO: 18 or SEQ ID NO: 20, or a part thereof An antisense polynucleotide having, preferably, a nucleotide sequence complementary to the nucleotide sequence of DNA having the nucleotide sequence represented by SEQ ID NO: 2, SEQ ID NO: 18 or SEQ ID NO: 20, or a part thereof Antisense polynucleotide (more preferably, a nucleotide sequence complementary to the nucleotide sequence of DNA having the nucleotide sequence represented by SEQ ID NO: 2, SEQ ID NO: 18 or SEQ ID NO: 20, or a portion thereof. Antisense polynucleotide) and the like.
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 The number of antisense polynucleotides is usually about 10 to 40, preferably 15 to
〜3 0個程度の塩基から構成される。 It is composed of about 30 bases.
ヌクレア一ゼなどの加水分解酵素による分解を防ぐために、 アンチセンス D N Aを構成する各ヌクレオチドのりん酸残基 (ホスフェート) は、 例えば、 ホ スホロチォエート、 メチルホスホネ一ト、 ホスホロジチォネートなどの化学修 飾りん酸残基に置換されていてもよい。 これらのアンチセンスポリヌクレオチ ドは、 公知の D N A合成装置などを用いて製造することができる。  To prevent degradation by hydrolases such as nucleases, the phosphate residues (phosphates) of each nucleotide constituting the antisense DNA should be chemically modified, for example, phosphorothioate, methylphosphonate, and phosphorodithionate. It may be substituted with a phosphoric acid residue. These antisense polynucleotides can be produced using a known DNA synthesizer or the like.
本発明に従えば、 本発明のタンパク質遺伝子の複製または発現を阻害するこ とのできる該遺伝子に対応するアンチセンスポリヌクレオチド (核酸) を、 ク 口一ン化した、 あるいは決定されたタ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。 かかるアンチセンスポリヌクレオチドは、 本発明の夕ンパク質遺伝子の R N Aとハイプリダイズすることができ、 該 R N Aの合成または機能を阻害することができるか、 あるいは本発明のタンパク質 関連 R NAとの相互作用を介して本発明のタンパク質遺伝子の発現を調節 ·制 御することができる。 本発明のタンパク質関連 R N Aの選択された配列に相補 的なポリヌクレオチド、 および本発明のタンパク質関連 R 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 「対応する」 とは、 遺伝子を含めたヌク レオチド、 塩基配列または核酸の特定の配列に相同性を有するあるいは相補的 であることを意味する。 ヌクレオチド、 塩基配列または核酸とタンパク質との 間で 「対応する」 とは、 ヌクレオチド (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) タンパク質のアミノ酸を通常指している。 タンパク質 遺伝子の 5 '端ヘアピンループ、 5 '端 6—ベースペア · リピート、 5 '端非翻訳領 域、 ポリペプチド翻訳開始コドン、 タンパク質コード領域、 O R F翻訳終止コ ドン、 3 '端非翻訳領域、 3 '端パリンドローム領域または 3 '端ヘアピンループな どは、 好ましい対象領域として選択しうるが、 タンパク質遺伝子内の如何なる 領域も対象として選択しうる。 According to the present invention, an antisense polynucleotide (nucleic acid) corresponding to the protein gene of the present invention, which can inhibit the replication or expression of the gene, has been cloned or encoded. It can be designed and synthesized based on DNA base sequence information. Such antisense polynucleotides can hybridize with the RNA of the protein gene of the present invention, inhibit the synthesis or function of the RNA, or interact with the protein-related RNA of the present invention. Thus, the expression of the protein gene of the present invention can be regulated and controlled. Polynucleotides that are complementary to a 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, are those of the present invention in vivo and in vitro. It is useful for regulating and controlling the expression of protein genes, and is also useful for treating or diagnosing diseases and the like. The term "corresponding" refers to a nucleotide, base sequence or specific sequence of a nucleic acid, including or homologous to a gene, Means that The "correspondence" between a nucleotide, nucleotide sequence or nucleic acid and a protein usually refers to the amino acids of the protein (as specified by the instructions) derived from the nucleotide (nucleic acid) sequence or its complement. 5 'end hairpin loop of protein gene, 5' end 6—base pair repeat, 5 'end untranslated region, polypeptide translation start codon, protein coding region, ORF translation stop codon, 3' end untranslated region, A 3'-end palindrome region or a 3'-end hairpin loop may be selected as a preferred region of interest, but any region within a protein gene may be selected as a target.
目的核酸と、 対象領域の少なくとも一部に相補的なポリヌクレオチドとの関 係については、 目的核酸が対象領域とハイブリダィズすることができる塲合は、 その目的核酸は、 当該対象領域のポリヌクレオチドに対して 「アンチセンス」 であるということができる。 アンチセンスポリヌクレオチドは、 2—デォキシ 一 D—リポースを含有しているポリヌクレオチド、 D—リボースを含有してい 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、 非ヌクレオチド骨格を有するその他のポリ マ一 (例えば、 市販のタンパク質核酸および合成配列特異的な核酸ポリマー) または特殊な結合を含有するその他のポリマー (但し、 該ポリマ一は D NAや R N 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、 D NA: R NAハイブリッドで あってもよく、 さらに非修飾ポリヌクレオチド (または非修飾オリゴヌクレオ チド) 、 公知の修飾の付加されたもの、 例えば当該分野で知られた標識のある もの、 キ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 ォ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、 ホスホロチォエート、 ホスホロジチォェ一トなど) を持つもの、 例えばタンパク質 (例、 ヌクレア一ゼ、 ヌクレア一ゼ ·インヒビター、 トキシ ン、 抗体、 シグナルペプチド、 ポリ一 L一リジンなど) や糖 (例、 モノサッカ ライドなど) などの側鎖基を有しているもの、 インターカレント化合物 (例、 ァクリジン、 ソラレンなど) を持つもの、 キレート化合物 (例えば、 金属、 放 射活性をもつ金属、 ホウ素、 酸化性の金属など) を含有するもの、 アルキル化 剤を含有するもの、 修飾された結合を持つもの (例えば、 ひァノマ一型の核酸 など) であってもよい。 ここで 「ヌクレオシド」 、 「ヌクレオチド」 および 「核酸」 とは、 プリンおよびピリミジン塩基を含有するのみでなく、 修飾され たその他の複素環型塩基をもつようなものを含んでいて良い。 このような修飾 物は、 メチル化されたプリンおよびピリミジン、 ァシル化されたプリンおよび ピリミジン、 あるいはその他の複素環を含むものであってよい。 修飾されたヌ クレオチドおよび修飾されたヌクレオチドはまた糖部分が修飾されていてよく、 例えば、 1個以上の水酸基がハロゲンとか、 脂肪族基などで置換されていたり、 またはェ一テル、 ァミンなどの官能基に変換されていてよい。 Regarding the relationship between the target nucleic acid and a polynucleotide complementary to at least a part of the target region, if the target nucleic acid can hybridize with the target region, the target nucleic acid is included in the polynucleotide of the target region. On the other hand, it can be said that it is “antisense”. Antisense polynucleotides are polynucleotides containing 2-dexoxy-D-reports, polynucleotides containing D-ribose, or other types of polynucleotides that are N-glycosides of purine or pyrimidine bases. Other polymers with non-nucleotide backbones (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 as found (contains nucleotides having a configuration that allows base attachment)). They may be double-stranded DNA, single-stranded DNA, double-stranded RNA, single-stranded RNA, DNA: RNA hybrid, and may further comprise unmodified polynucleotides (or unmodified oligonucleotides). ), Those with known modifications, e.g., those with labels known in the art, capped, methylated, one or more natural nucleotides replaced with analogs Or modified with an intramolecular nucleotide, for example, having an uncharged bond (eg, methylphosphonate, phosphotriester, phosphoramidate, olebamate, etc.), a charged bond or a sulfur-containing bond (eg, Those with rotioate, phosphorodithioate, etc., such as proteins (eg, nucleases, nuclease inhibitors, toxins, antibodies) Signal peptides, poly-one L one lysine, etc.) or a sugar (e.g., Monosakka Compounds having side-chain groups such as amides, etc., compounds having an interactive compound (eg, acridine, psoralen, etc.), chelating compounds (eg, metals, radioactive metals, boron, oxidizable metals) ), Those containing an alkylating agent, and those having a modified bond (for example, a nucleic acid of the Hyanoma type 1). Here, "nucleoside", "nucleotide" and "nucleic acid" may include not only those containing purine and pyrimidine bases but also those having other modified heterocyclic bases. Such modifications may include methylated purines and pyrimidines, acylated purines and pyrimidines, or other heterocycles. Modified nucleotides and modified nucleotides may also be modified at the sugar moiety, e.g., where one or more hydroxyl groups have been replaced by 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または修飾され 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、 チォホスフェート誘導体、 ポリヌクレオシドアミドゃオリゴヌク レオシドアミドの分解に抵抗性のものなどが挙げられる。 本発明のアンチセン スポリヌグレオチドは、 例えば、 以下のように設計されうる。 すなわち、 細胞 内でのアンチセンスポリヌクレオチドをより安定なものにする、 アンチセンス ポリヌクレオチドの細胞透過性をより高める、 目標とするセンス鎖に対する親 和性をより大きなものにする、 また、 もし毒性があるような場合はアンチセン スポリヌクレオチドの毒性をより小さなものにする。 このような修飾は、 例え ば Pharm Tech Japan, 8卷, 247頁または 395頁, 1992年、 Ant i sense Research and Appl i cat i ons, CRC Press, 1993年などで数多く報告されている。  The antisense polynucleotide of the present invention is an RNA, a DNA or a modified nucleic acid (RNA, DNA). Specific examples of the modified nucleic acid include a sulfur derivative of a nucleic acid, a thiophosphate derivative, and a polynucleoside amide / polynucleoside amide that is resistant to degradation of oligonucleoside amide. The antisense polynucleotide of the present invention can be designed, for example, as follows. In other words, it makes the antisense polynucleotide more stable in the cell, enhances the cell permeability of the antisense polynucleotide, increases the affinity for the target sense strand, and In some cases, the toxicity of the antisense polynucleotide is reduced. Many such modifications have been reported, for example, in Pharm Tech Japan, vol. 8, p. 247 or p. 395, 1992, Antisense Research and Appli cations, CRC Press, 1993.
本発明のアンチセンスポリヌクレオチドは、 変化せしめられたり、 修飾され た糖、 塩基、 結合を含有していて良く、 リボゾーム、 ミクロスフエアのような 特殊な形態で供与されたり、 遺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カチオン体、 細胞膜との相互作用を高めたり、 核酸の取込みを増大せしめるような脂質 (例、 ホスホリピド、 コレステロールなど) などの疎水性のものが挙げられる。 付加 するに好ましい脂質としては、 コレステロールやその誘導体 (例、 コ'レステリ ルクロロホルメート、 コール酸など) が挙げられる。 こうしたものは、 核酸の 3 '端または 5 '端に付着させることができ、 塩基、 糖、 分子内ヌクレオシド結合 を介して付着させることができうる。 その他の基としては、 核酸の 3,端または 5 '端に特異的に配置されたキャップ用の基で、 ェキソヌクレア一ゼ、 R N a s eなどのヌクレア一ゼによる分解を阻止するためのものが挙げられる。 こうし たキャップ用の基としては、 ポリエチレングリコ一ル、 テトラエチレングリコ —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 polynucleotides of the present invention may contain altered or modified sugars, bases, or bonds, are provided in special forms such as ribosomes, microspheres, are applied by gene therapy, It can be provided in an added form. Such additional forms include polycations such as polylysine, which acts to neutralize the charge on the phosphate backbone, and lipids, which enhance the interaction with cell membranes and increase the uptake of nucleic acids ( For example, Hydrophobic substances such as phospholipid and cholesterol). Preferred lipids for addition include cholesterol and its derivatives (eg, cholesteryl chloroformate, cholic acid, etc.). These can be attached to the 3 'or 5' end of the nucleic acid and can be attached via bases, sugars, intramolecular nucleoside bonds. Other groups include capping groups specifically located at the 3, 5 'or 5' end of nucleic acids to prevent degradation by nucleases such as exonucleases and RNases . Such capping groups include, but are not limited to, hydroxyl-protecting groups known in the art, including glycols such as polyethylene glycol and tetraethylene glycol. .
アンチセンスポリヌクレオチドの阻害活性は、 本発明の形質転換体、 本発明 の生体内や生体外の遺伝子発現系、 または本発明の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。 以下に、 本発明のタンパク質もしくは部分ペプチドまたはその塩 (以下、 本 発明のタンパク質と略記する場合がある) 、 本発明のタンパク質または部分べ プチドをコードする D NA (以下、 本発明の D NAと略記する場合がある) 、 本発明のタンパク質もしくは部分べ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 N A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 polynucleotide can be examined using the transformant of the present invention, the in vivo or in vitro gene expression system of the present invention, or the in vivo or in vitro translation system of the protein of the present invention. . Hereinafter, the protein or partial peptide of the present invention or a salt thereof (hereinafter, sometimes abbreviated as the protein of the present invention), the DNA encoding the protein or partial peptide of the present invention (hereinafter, referred to as the DNA of the present invention) Abbreviated), 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 referred to as the present invention). May be abbreviated as an antisense polynucleotide).
本発明のタンパク質は、 慢性閉塞性肺疾患や鼻炎において組織特異的に発現 が増加するので、 疾患マーカーとして利用することができる。 よって、 本発明 のタンパク質は、 例えば、 肺 ·気道の炎症を伴う肺'胸部疾患などの呼吸器疾 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、 過敏性肺炎など〕 、 ァレルギ一性結膜炎、 鼻炎  The protein of the present invention can be used as a disease marker because its expression is increased in a tissue-specific manner in chronic obstructive pulmonary disease and rhinitis. Therefore, the protein of the present invention may be used, for example, for respiratory diseases such as pulmonary thoracic disease accompanied by inflammation of the lungs and airways [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, Bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), allergic conjunctivitis, rhinitis
(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) 、 消化管 疾患 (例、 過敏性腸症候群、 炎症性腸疾患、 潰瘍性大腸炎、 クローン病、 逆流 性食道炎など) などにおける早期診断、 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。 (Eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc.), gastrointestinal diseases (eg, Irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease, reflux It is useful as a marker for early diagnosis of esophagitis, etc.), judgment of symptom severity, and prediction of disease progression.
本発明のタンパク質をコードする遺伝子 (本発明のタンパク質遺伝子) 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、 本発明のタンパク質遺伝子の発現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、 肺 ·気道の炎症を伴う肺 ·胸部疾患などの呼吸器疾患 〔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、 過敏性肺炎など〕 、 アレルギー性結膜炎、 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) 、 消化管疾患 (例、 過敏性腸症候群、 炎症性腸疾患、'潰瘍性大腸炎、 クローン病、 逆流性食道炎など) 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、 消化管疾患など の予防 ·治療剤である。  Antisense polynucleotide of a gene encoding the protein of the present invention (protein gene of the present invention), a compound or a salt thereof that inhibits the activity of the protein of the present invention, a compound that inhibits the expression of the gene of the protein of the present invention, or Pharmaceuticals containing a salt thereof or an antibody against the protein of the present invention include, for example, respiratory diseases such as lungs, lungs accompanied by inflammation of the respiratory tract, and chest diseases [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema) , Diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), allergic conjunctivitis, rhinitis (eg, allergic rhinitis, pollinosis, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis , Dry rhinitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc., gastrointestinal disorders (eg, irritable bowel syndrome, inflammatory bowel disease) 'Ulcerative colitis, Crohn's disease, etc. reflux esophagitis) prevention of such - can be used as a therapeutic agent. Preferred are preventive and therapeutic agents for respiratory diseases, gastrointestinal diseases and 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 disease
本発明のタンパク質および本発明のタンパク質遺伝子は、 慢性閉塞性肺疾患 や鼻炎において組織特異的に発現が増加し、 肺、 気道あるいは鼻粘膜における 粘液産生促進作用および肺胞壁破壊促進作用を有するので、 本発明のタンパク 質の活性を阻害する化合物またはその塩、 および本発明のタンパク質遺伝子の 発現を阻害する化合物またはその塩は、 例えば、 肺 ·気道の炎症を伴う肺-胸 部疾患などの呼吸器疾患 〔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、 過敏性肺炎など〕 、 ァレ ルギー性結膜炎、 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) 、 消化管疾患 (例、 過敏性腸症候群、 炎症性腸疾患、 潰瘍性大腸炎、 クローン病、 逆流性食道炎など) などの予防 ·治療剤、 好ましくは呼吸器疾患、 消化管疾患などの予防 ·治療剤などの低毒性な医薬として安全に使用できる。 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阻害する 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り、 該夕ン パク質をコードするポリヌクレオチドは、 本発明のタンパク質遺伝子の発現を 阻害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。 すなわち、 本発明は、 (1 ) 本発明のタンパク質を用いることを特徴とする 本発明のタンパク質の活性 (例、 クロライドチャネル活性など) を阻害する化 合物またはその塩 (以下、 阻害剤と略記する場合がある) のスクリーニング方 法、 (2 ) 本発明のタンパク質をコードするポリヌクレオチドを用いることを 特徴とする本発明のタンパク質遺伝子の発現を阻害する化合物またはその塩 The protein of the present invention and the gene of the present invention have an increased tissue-specific expression in chronic obstructive pulmonary disease and rhinitis, and have a mucus production promoting action and an alveolar wall destruction promoting action in the lung, airway or nasal mucosa. The compound or its salt that inhibits the activity of the protein of the present invention and the compound or its salt that inhibits the expression of the protein gene of the present invention can be used, for example, for respiratory diseases such as lung-thoracic diseases accompanied by inflammation of the lungs and airways. Respiratory tract disease [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.], allergic conjunctivitis, rhinitis (eg, allergy) Rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc.), Prevention of gastrointestinal diseases (eg, irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease, reflux esophagitis, etc.) · Therapeutic agents, preferably prevention of respiratory diseases, gastrointestinal diseases, etc. · It can be used safely as a low-toxic drug such as a therapeutic agent. Therefore, the protein of the present invention inhibits the activity of the protein of the present invention. The polynucleotide encoding the protein is useful as a reagent for screening a compound or a salt thereof, and the polynucleotide encoding the protein is useful as a reagent for screening a compound or a salt thereof that inhibits expression of the protein gene of the present invention. is there. That is, the present invention provides (1) a compound that inhibits the activity of the protein of the present invention (eg, chloride channel activity and the like) characterized by using the protein of the present invention, or a salt thereof (hereinafter, abbreviated as an inhibitor). (2) a compound that inhibits expression of the protein gene of the present invention, which comprises using a polynucleotide encoding the protein of the present invention, or a salt thereof.
(以下、 阻害剤と略記する場合がある) のスクリーニング方法、 (3 ) 本発明 の抗体を用いることを特徴とする本発明の夕ンパク質の発現を阻害する化合物 またはその塩 (以下、 阻害剤と略記する場合がある) のスクリーニング方法な どを提供する。  (Hereinafter sometimes abbreviated as an inhibitor), (3) a compound or a salt thereof that inhibits the expression of the protein of the present invention characterized by using the antibody of the present invention (hereinafter referred to as an inhibitor) (May be abbreviated as).
スクリーニング方法の具体例としては、 (i) 本発明のタンパク質を産生する 能力を有する細胞をカルシウム賦活剤で活性化した場合と (i i) 本発明のタン パク質を産生する能力を有する細胞と試験化合物の混合物とをカルシウム賦活 剤で活性化した場合との比較を行うことを特徴とする阻害剤のスクリーニング 方法が挙げられる。  Specific examples of the screening method include: (i) the case where cells having the ability to produce the protein of the present invention are activated with a calcium activator; and (ii) the case where cells having the ability to produce the protein of the present invention are tested. A method for screening for an inhibitor, which is characterized by comparing with a case where a mixture of compounds is activated with a calcium activator, is mentioned.
上記スクリ一ニング方法においては、 例えば、 (0 と (i i) の場合における、 本発明のタンパク質のクロライドチャネル活性を測定して、 比較する。  In the above screening method, for example, the chloride channel activity of the protein of the present invention in the cases (0 and (ii)) is measured and compared.
カルシウム賦活剤は、 試験化合物と混合した後に、 本発明のタンパク質を産 生する能力を有する細胞に添加してもよく、 また、 本発明のタンパク質を産生 する能力を有する細胞にカルシウム賦活剤を添加した後、 試験化合物を添加し てもよい。  The calcium activator may be added to cells having the ability to produce the protein of the present invention after being mixed with the test compound, or the calcium activator may be added to cells having the ability to produce the protein of the present invention. After that, the test compound may be added.
本発明のタンパク質を産生する能力を有する細胞と試験化合物との混合は、 本発明のタンパク質を産生する能力を有する細胞をカルシウム賦活剤で活性化 する前または後の何れであってもよく、 また本発明のタンパク質を産生する能 力を有する細胞に試験化合物とカルシウム賦活剤の混合物を添加してもよい。 カルシウム賦活剤としてはィオノマイシン、 A 2 3 1 8 7 (カルシマイシ ン) などが用いられる。 本発明のタンパク質のクロライドチャネル活性は、 公知の方法、 例えば、 ジ エノミクス (Genomics) 、 54巻、 200頁 (1998) に記載の方法あるいはそれに準 じる方法に従って ^定することができる。 The cells having the ability to produce the protein of the present invention and the test compound may be mixed before or after the cells having the ability to produce the protein of the present invention are activated with a calcium activator. A mixture of a test compound and a calcium activator may be added to cells having the ability to produce the protein of the present invention. As a calcium activator, ionomycin, A2187 (calcimycin) and the like are used. The chloride channel activity of the protein of the present invention can be determined according to a known method, for example, the method described in Genomics, vol. 54, p. 200 (1998) or a method analogous thereto.
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挙 げられ、 これら化合物は新規な化合物であってもよいし、 公知の化合物であつ , てもよい。  Test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, and animal tissue extracts, and these compounds are novel compounds. Or a known compound.
上記のスクリーニング方法を実施するには、 本発明のタンパ 質を産生する 能力を有する細胞をスクリーニングに適したバッファーに浮遊して調製する。 バッファーには、 p H約 4〜 1 0 (望ましくは、 p H約 6〜8 ) のリン酸バッ ファー、 ほう酸バッファ一などであればいずれでもよい。  In order to carry out the above screening method, cells having the ability to produce the protein of the present invention are prepared by suspending them in a buffer suitable for screening. The buffer may be any one of phosphate buffer and borate buffer having a pH of about 4 to 10 (preferably, a pH of about 6 to 8).
スクリーニング方法の具体例としては、 (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) 本発明のタンパク質を産生する能 力を有する細胞と試験化合物の混合物を培養した場合における、 本発明のタン パク質遺伝子の発現量 (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 N A量) を測定して、 比較する。  Specific examples of the screening method include (iii) culturing cells capable of producing the protein of the present invention and (iv) culturing a mixture of cells capable of producing the protein of the present invention and a test compound. Then, the expression level of the protein gene of the present invention (specifically, the amount of the protein of the present invention or the amount of mRNA encoding the protein) is measured and compared.
本発明の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、 ゥ エスタン解析、 ELISA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。  The amount of the protein of the present invention can be measured by a known method, for example, using an antibody recognizing the protein of the present invention to detect the protein present in a cell extract or the like. It can be measured in accordance with the corresponding method.
本発明のタンパク質遺伝子発現量は、 公知の方法、 例えば、 ノーザンブロッ ティングや Reverse transcr ipt ion - polymerase chain react ion (RT-PCR) 、 リ アルタイム P C R解析システム (ABI社製、 TaqMan polymerase chain  The expression level of the protein gene of the present invention can be determined by a known method, for example, Northern blotting, reverse transcription-polymerase chain reaction (RT-PCR), real-time PCR analysis system (manufactured by ABI, TaqMan polymerase chain).
r eac U on) などの方法あるいはそれに準じる方法にしたがつて測定することが できる。  r eac Uon) or a method analogous thereto.
試験化合物としては、 例えば、 ペプチド、 タンパク、 非ペプチド性化合物、 合成化合物、 発酵生産物、 細胞抽出液、 植物抽出液、 動物組織抽出液などが挙 げられ、 これら化合物は新規な化合物であってもよいし、 公知の化合物であつ てもよい。 上記のスクリーニング方法を実施するには、 本発明のタンパク質を産生する 能力を有する細胞をスクリーニングに適したバッファ一に浮遊して調製する。 バッファーには、 p H約 4〜1 0 (望ましくは、 p H約 6〜8 ) のリン酸バッ ファー、 ほう酸バッファ一などの、 本発明のタンパク質のクロライドチャネル 活性を阻害しないバッファ一であればいずれでもよい。 Test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, and animal tissue extracts, and these compounds are novel compounds. Or a known compound. To carry out the above screening method, cells having the ability to produce the protein of the present invention are prepared by suspending them in a buffer suitable for screening. The buffer may be a buffer that does not inhibit the chloride channel activity of the protein of the present invention, such as a phosphate buffer having a pH of about 4 to 10 (preferably, a pH of about 6 to 8) or a borate buffer. Either may be used.
スクリーニング方法の具体例としては、 (ν)'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 (v i ) 本発明のタンパク質を産生する能力 を有する細胞と試験化合物の混合物を培養した場合との比較を行う。 上記スク リーニング方法においては、' 本発明の抗体を用いて (V) と (v i ) の場合におけ る、 本発明のタンパク質の発現量 (具体的には、 本発明のタンパク質量) を測 定 (例、 発現の検出、 発現量の定量など) して、 比較する。  Specific examples of the screening method include (ν) ′ when a cell capable of producing the protein of the present invention is cultured and (vi) when a mixture of a cell capable of producing the protein of the present invention and a test compound is cultured. Make a comparison with the case. In the above screening method, the expression level of the protein of the present invention (specifically, the amount of the protein of the present invention) in the cases (V) and (vi) is measured using the antibody of the present invention. (Eg, detection of expression, quantification of expression level, etc.) and compare.
本発明の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、 ゥ エスタン解析、 ELISA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。  The amount of the protein of the present invention can be measured by a known method, for example, using an antibody recognizing the protein of the present invention to detect the protein present in a cell extract or the like. It can be measured in accordance with the corresponding method.
試験化合物としては、 例えば、 ペプチド、 タン ク、 非ペプチド性化合物、 合成化合物、 発酵生産物、 細胞抽出液、 植物抽出液、 動物組織抽出液などが挙 げられ、 これら化合物は新癍な化合物であってもよいし、 公知の化合物であつ てもよい。  Test compounds include, for example, peptides, tanks, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, etc., and these compounds are new compounds. Or a known compound.
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 N Aを含有するベクターで形質転換され た宿主 (形質転換体) が用いられる。 宿主としては、 例えば、 C HO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膜上に発現させた形質 転換体が好ましく用いられる。  As a cell having the ability to produce the protein of the present invention, for example, a host (transformant) transformed with a vector containing a DNA encoding the protein of the present invention described above is used. As the host, for example, animal cells such as CHO cells are preferably used. For the screening, for example, a transformant in which the protein of the present invention is expressed on a cell membrane by culturing by the above-mentioned method is preferably used.
例えば、 上記 (i i ) の場合におけるクロライドチャネル活性を、 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 %以上阻害す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ま たはその塩として選択することができる。 例えば、 上記 (iv) め場合における本発明のタンパク質遺伝子の発現量を、 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 %以上阻害する試験化合物を本発明の夕ンパク質遺伝子の発現を 阻害する化合物またはその塩として選択することができる。 For example, a test compound that inhibits chloride channel activity in the case of the above (ii) by about 20% or more, preferably 30% or more, and more preferably about 50% or more compared to the case of the above (i) It can be selected as a compound that inhibits the activity of the protein of the present invention or a salt thereof. For example, the expression level of the protein gene of the present invention in the case (iv) is about 20% or more, preferably 30% or more, more preferably about 50%, as compared with the case (iii). The test compound that inhibits the above can be selected as a compound that inhibits the expression of the protein gene of the present invention or a salt thereof.
例えば、 上記 (vi) の場合における本発明のタンパク質の発現量を、 上記  For example, the expression level of the protein of the present invention in the case of the above (vi) is
(V) 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 %以上阻害する試験化合物を本発明のタンパク質の発現を阻害する化合 物またはその塩として選択することができる。  A test compound that inhibits about 20% or more, preferably 30% or more, more preferably about 50% or more of that in the case of (V) as a compound that inhibits expression of the protein of the present invention or a salt thereof. You can choose.
本発明のスクリーニング用キットは、 本発明のタンパク質もしくは部分ぺプ チドまたはその塩、 本発明のタンパク質もしくは部分ペプチドをコードするポ リぺプチド、 本発明の抗体または本発明の夕ンパク質もしくは部分べプチドを 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 includes a protein or a partial peptide of the present invention or a salt thereof, a polypeptide encoding a protein or a partial peptide of the present invention, an antibody of the present invention, or a protein or partial base of the present invention. It contains cells that have the ability to produce peptides.
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、 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、 本発明の夕 ンパク質の活性 (例、 クロライドチャネル活性など) 、 本発明のタンパク質遺 伝子の発現、 または本発明の蛋白質の発現を阻害する化合物またはその塩であ る。  Compounds or salts thereof obtained by using the screening method or the screening kit of the present invention include the test compounds described above, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, and plant extracts. Liquid, animal tissue extract, plasma, or a compound thereof, or a salt thereof, wherein the activity of the protein of the present invention (eg, chloride channel activity, etc.), the expression of the protein gene of the present invention, or A compound or a salt thereof that inhibits expression of the protein of the invention.
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、 肺 ·気道の炎症を伴う肺 ·胸部疾患などの 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、 過敏性肺炎など〕 、 アレルギー性結膜 炎、 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) 、 消化管疾患 (例、 過敏性腸症候群、 炎症性腸疾患、 潰瘍性大腸炎、 クローン病、 逆流性食道炎など) などに対する予防 ·治療剤、 好ましくは呼吸器疾患、 消化 管疾患などの予防 ·治療剤などの医薬として有用である。 Compounds or salts thereof obtained using the screening method or screening kit of the present invention include, for example, respiratory diseases such as lungs, lungs with inflammation of the airways, and chest diseases [eg, chronic obstructive pulmonary diseases (chronic bronchial Inflammation, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), allergic conjunctivitis, rhinitis (eg, allergic rhinitis, pollinosis, acute rhinitis, chronic rhinitis, hypertrophic) Rhinitis, atrophic rhinitis, dry rhinitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc., gastrointestinal diseases (eg, irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease, Prophylactic and therapeutic agents for reflux esophagitis, etc., preferably respiratory disease, digestion It is useful as a medicine for the prevention and treatment of tract diseases and the like.
本発明のスクリ一ニング方法またはスクリ一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、 それ自体または適当な医薬として、 安全に投与する ことができる。 上記投与に用いられる医薬は、 上記化合物またはその塩と薬理 学的に許容され得る担体、 希釈剤もしくは陚形剤とを含むものであり、 経口ま たは非経口投与に適する医薬組成物として提供される。  The compound or a salt thereof obtained by using the screening method or the screening kit of the present invention can be safely administered by itself or as a suitable drug. The medicament used for the above-mentioned administration contains the above-mentioned compound or a salt thereof and a pharmacologically acceptable carrier, diluent or vehicle, and is provided as a pharmaceutical composition suitable for oral or parenteral administration. Is done.
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、 カプセル剤 (ソフトカプセル剤を含む) 、 シロップ剤、 乳剤、 懸濁剤などがあ げられる。 かかる組成物は公知の方法によって製造され、 製剤分野において通 常用いられる担体、 希釈剤もしくは賦形剤を含有するものである。 例えば、 錠 剤用の担体、 賦形剤としては、 乳糖、 でんぷん、 蔗糖、 ステアリン酸マグネシ ゥムなどが用いられる。  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, and capsules (including soft capsules). 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 pharmaceutical field. 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、 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、 ポリソルベート 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って調製される。  As compositions for parenteral administration, for example, injections, suppositories, etc. are used. Injections are in the form of intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections, infusions, etc. Is included. Such injections are prepared according to a known method, for example, by dissolving, suspending or emulsifying the above antibody or a salt thereof in a sterile aqueous or oily liquid usually used for injections. As an aqueous liquid for injection, for example, physiological saline, isotonic solution containing glucose and other adjuvants, and the like, suitable solubilizing agents, for example, alcohol (eg, ethanol), polyalcohol (eg, Propylene glycol, polyethylene glycol), a nonionic surfactant [eg, polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated catalyst)], and the like. 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 into a suitable ampoule. Suppositories used for rectal administration are prepared by mixing the antibody or a salt thereof with a conventional suppository base.
上記の経口用または非経口用医薬組成物は、 活性成分の投与量に適合するよ うな投薬単位の剤形に調製されることが好都合である。 かかる投薬単位の剤形 としては、 錠剤、 丸剤、 カプセル剤、 注射剤 (アンプル) 、 坐剤などが例示さ れ、 それぞれの投薬単位剤形当たり通常 5〜500mg、 とりわけ注射剤では 5〜 100mg、 その他の剤形では 10〜250mgの上記抗体が含有されていることが好まし い。 The above-mentioned oral or parenteral pharmaceutical composition is conveniently prepared in a unit dosage form so as to be compatible with the dosage of the active ingredient. Such dosage unit dosage form Examples include tablets, pills, capsules, injections (ampoules), suppositories, etc., usually 5 to 500 mg per dosage unit, especially 5 to 100 mg for injections, and other dosage forms. Preferably, it contains 10-250 mg of the above antibody.
例えば、 経鼻投与のための組成物としては、 吸入剤、 点鼻剤、 エアゾール剤 などが用いられる。  For example, compositions for nasal administration include inhalants, nasal drops, aerosols and the like.
常套手段に従って得られた製剤を、 吸入剤として使用する場合、 公知の方法 を用いて、 粉末吸入剤、 吸入用懸濁剤、 吸入用溶液またはカプセル状吸入剤と し、 用時適当な吸入器を用いて適用することができ、 特に粉末吸入剤が好まし く用いられる。  When the preparation obtained by a conventional method is used as an inhaler, it is converted into a powder inhaler, a suspension for inhalation, a solution for inhalation or a capsule inhaler using a known method, and an appropriate inhaler for use. , And powder inhalants are particularly preferably used.
粉末吸入剤の平均粒子径としては、 特に限定されないが、 約 0. 1〜20 imが好 ましく、 特に約 1〜5 mが好ましい。  The average particle size of the powder inhalant is not particularly limited, but is preferably about 0.1 to 20 im, and particularly preferably about 1 to 5 m.
- また、 粉末吸入剤の粒度としては、 特に限定されないが、 約 25 ^m以上の粒子 の量が約 5%以下、 特に約 1 %以下であることが好ましい。  -The particle size of the powder inhaler is not particularly limited, but the amount of particles having a size of about 25 m or more is preferably about 5% or less, particularly preferably about 1% or less.
また、 吸入剤を使用する場合、 適用の際に使用する器具としては、 市販の吸 入器を用いても良く、 例えば、 ベントリン ·ロタキャップス (VENTOLIN  When using an inhalant, a commercially available inhaler may be used as a device for application. For example, ventolin / rotacaps (VENTOLIN)
R0TACAPS;ダラクソ社) 、 スピンへラー (登録商標、 藤沢薬品工業 (株) ) 、 インタール ·スピンキャップス (INTAL SPINCAPS; フィソンズ社) 、 アト口べ ント .アンド .ベロテック 'ィンハレツテン (ATROVENT AND BER0TEC R0TACAPS; Daraxo Co., Ltd., Spin Heller (registered trademark, Fujisawa Pharmaceutical Co., Ltd.), INTAL SPINCAPS (Fisons Co., Ltd.), Atovento and Verotech's ATROVENT AND BER0TEC
INHALETTEN;ベ一リンガー ·ィンゲルハイム社) 、 フオラディル (F0RADIL;チ バ社) 、 ベントディスク (BENTODISKS;ダラクソ社) 、 パブライザ一 (登録商 標、 帝人 (株) ) 、 プリカ二ル'ターブハラ一 (BRICANYL TURBUHALER;ァスト ラ社) 、 ミア卜 ·ィンスフアレイター (MI AT INSUFFLATOR) などが挙げられる。 投与経路としては、 通常、 吸入器具を用いて鼻、 口などへ直接吸入するが好 ましい。 INHALETTEN; BERINGER INGELHEIM, FORADIL (F0RADIL; Ciba), BENTODISKS (Daraxo), publisher (registered trademark, Teijin Ltd.), BRICANYL TURBUHALER ; Astra) and MI AT INSUFFLATOR. As a route of administration, it is usually preferable to inhale directly into the nose and mouth using an inhaler.
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。 本発明のタンパク質の活性を阻害する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、 慢 性閉塞性肺疾患治療の目的で、 上記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 60kgとして) においては、 一日につき該ィヒ合物またはその 塩を約 0. l〜100mg、 好ましくは約 1. 0〜50mg、 より好ましくは約 1. 0〜20mg投与 する。 非経口的に投与する場合は、 上記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、 慢性閉塞性肺疾患治療の 目的で、 本発明のタンパク質の活性を阻害する化合物またはその塩を注射剤の 形で通常成人 (体重 60kg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程度、 好ましくは約 0. l〜20mg程度、 より好ましくは約The preparations obtained in this way are safe and low toxic and can be used, for example, in humans or in warm-blooded animals (eg, mice, rats, puppies, higgs, bushus, puppies, puppies, birds, cats, dogs). , Monkeys, chimpanzees, etc.) orally or parenterally. The dose of the compound or its salt that inhibits the activity of the protein of the present invention may vary depending on its action, target disease, subject to be administered, administration route and the like.For example, for the purpose of treating chronic obstructive pulmonary disease, When the compound or a salt thereof is administered orally, in general, for an adult (assuming a body weight of 60 kg), about 0.1 to 100 mg, preferably about 1.0 to 50 mg, of the compound or its salt per day is used. More preferably, about 1.0 to 20 mg is administered. When administered parenterally, the single dose of the above compound or a salt thereof varies depending on the administration subject, target disease, etc., for example, the activity of the protein of the present invention for the purpose of treating chronic obstructive pulmonary disease. When a compound or a salt thereof is administered to an adult (with a body weight of 60 kg) usually in the form of an injection, the compound or a salt thereof is about 0.01 to 30 mg, preferably about 0.1 to 20 mg per day. Degree, more preferably about
0. 1〜10mg程度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 60kg当たりに換算した量を投与することができる。 It is convenient to administer 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 ¾明のタンパク質を特異的に認識することができるので、 被検液中 の本発明のタンパク質の定量、 特にサンドイッチ免疫測定法による定量などに 使用することができる。  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, and therefore, the quantification of the protein of the present invention in a test solution. In particular, it can be used for quantification by sandwich immunoassay.
すなわち、 本発明は、  That is, the present invention
( i ) 本発明の抗体と、 被検液および標識化された本発明のタンパク質とを競 合的に反応させ、 該抗体に結合した標識化された本発明の ンパク質の割合を 測定することを特徴とする被検液中の本発明のタンパク質の定量法、 および  (i) Competitively reacting the antibody of the present invention with a test solution and the labeled protein of the present invention, 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, which comprises:
(i i) 被検液と担体上に不溶化した本発明の抗体および標識化された本発明の 別の抗体とを同時あるいは連続的に反応させたのち、 不溶化担体上の標識剤の 活性を測定することを特徴とする被検液中の本発明のタンパク質の定量法を提 供する。 ' 上記 (i i) の定量法においては、 一方の抗体が本発明のタンパク質の N端部 を認識する抗体で、 他方の抗体が本発明のタンパク質の C端部に反応する抗体 であることが望ましい。 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 ノクロ一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 Ca b ' ) 2 、 F a b \ 5 あるいは F a b画分を用いてもよい。 (ii) After reacting the test solution with the antibody of the present invention insolubilized on the carrier and another labeled antibody of the present invention simultaneously or continuously, measuring the activity of the labeling agent on the insolubilized carrier A method for quantifying the protein of the present invention in a test solution is provided. '' In the quantification method (ii) above, it is desirable that one antibody is an antibody that recognizes the N-terminal of the protein of the present invention and the other antibody is an antibody that reacts with the C-terminal of the protein of the present invention. . In addition, the protein of the present invention can be quantified using a monoclonal antibody against the protein of the present invention (hereinafter sometimes referred to as the monoclonal antibody of the present invention), and can also be detected by tissue staining or the like. . For these purposes, the antibody molecule itself may be used, or the FCab ') 2 , Fab \ 5 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0 法であれば、 いずれの測定法を用いてもよい。 例えば、 ネフロメトリ一、 競合 法、 ィムノメトリック法およびサンドィツチ法が好適に用いられるが、 感度、 特異性の点で、 後述するサンドィツチ法を用いるのが特に好ましい。  The method for quantifying the protein of the present invention using the antibody of the present invention is not particularly limited, and may be an antibody, an antigen, or an antibody-antigen complex corresponding to the amount of antigen (eg, the amount of protein) in the test solution. Any measurement method may be used as long as the amount is detected by chemical or physical means and the amount is measured 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 (例、 〔1251〕 、 〔1311〕 、 〔¾〕 、 〔14c〕 など) 、 蛍光物質 〔例、 シァニン5 蛍光色素 (例、 Cy2、 Cy3、 Cy5、 Cy5. 5、 Cy7 (アマシャムバイオサイエンス社 製) など) 、 フルォレスカミン、 フルォレツセンィソチォシァネ一卜など〕 、 酵素 (例、 β一ガラクトシダーゼ、 β -ダルコシダ一ゼ、 アル力リフォスファ 夕一ゼ、 パーォキシダ一ゼ、 リンゴ酸脱水素酵素など) 、 発光物質 (例、 ルミ ノール、 ルミノール誘導体、 レシフェリン、 ルシゲニンなど) 、 ピオチン、 ラ ンタニド元素などが用いられる。 さらに、 抗体あるいは抗原と標識剤との結合 にピオチン一アビジン系を用いることもできる。 Examples of the labeling agent used in the assay method using the labeling substance, for example, a radioactive isotope (e.g., [125 1], [131 1], [¾], etc. [14 c]), fluorescent substances [e.g., Shianin 5 Fluorescent dyes (eg, Cy2, Cy3, Cy5, Cy5.5, Cy7 (manufactured by Amersham Biosciences), etc.), fluorescamine, fluorescein sotiocinet, etc.), enzymes (eg, β-galactosidase, β-darcosidase, alpholipophorase, peroxidase, malate dehydrogenase, etc., luminescent substances (eg, luminol, luminol derivatives, reciferin, lucigenin, etc.), piotin, lanthanide, etc. Can be Furthermore, 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、 デキス卜ラン、 セルロースな どの不溶性多糖類、 ポリスチレン、 ポリアクリルアミド、 シリコン等の合成樹 脂、 あるいはガラス等が挙げられる。 For the insolubilization of an antigen or an antibody, physical adsorption may be used, or a method using a chemical bond usually used for insolubilizing and immobilizing a protein or an enzyme may be used. Examples of the carrier include insoluble polysaccharides such as agarose, dextran, and cellulose; synthetic resins such as polystyrene, polyacrylamide, and silicon; and glass.
サンドィツチ法においては不溶化した本発明のモノクロ一ナル抗体に被検液 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夕ンパク質量を定量することができる。 1次反応と 2次反応は逆の順序に行っても、 また、 同時に行なってもよいし時間をずらし て行なってもよい。 標識化剤および不溶化の方法は前記のそれらに準じること ができる。 また、 サンドイッチ法による免疫測定法において、 固相用抗体ある いは標識用抗体に用いられる抗体は必ずしも 1種類である必要はなく、 測定感 度を向上させる等の目的で 2種類以上の抗体の混合物を用いてもよい。 In the sandwich method, 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). Measuring the activity of the above labeling agent Thus, the mass of the protein of the present invention in the test solution can be determined. The primary reaction and the secondary reaction may be performed in the reverse order, may be performed simultaneously, or may be performed at staggered times. The labeling agent and the method of insolubilization can be the same as those described above. Also, in the immunoassay by the sandwich method, the antibody used for the solid phase antibody or the labeling antibody is not necessarily required to be one type, and two or more types of antibodies are used for the purpose of improving measurement sensitivity and the like. Mixtures 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口一ナル抗体は、 本発明の夕ン 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 differs in the binding site of the protein of the present invention. Antibodies are preferably used. That is, when the antibody used in the primary reaction and the secondary reaction is, for example, the antibody used in the secondary reaction recognizes the C-terminal of the protein of the present invention, the antibody used in the primary reaction is preferably An antibody that recognizes other than the C-terminal, for example, the N-terminal, is used.
本発明のモノクローナル坊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。  The monoclonal sample 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.
競合法では、 被検波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 / 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 / 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。  In the competition method, after the antigen in the test wave and the labeled antigen are allowed to react competitively with the antibody, the unreacted labeled antigen (F) is separated from the labeled antigen (B) bound to the antibody ( B / F separation), measure the amount of labeling for either B or F, and quantify the amount of antigen in the test solution. In this reaction method, a soluble antibody is used as an antibody, B / F separation is performed using polyethylene glycol, a liquid phase method using a second antibody against the antibody, a solid phase antibody is used as the first antibody, or An immobilization method using a soluble antibody as the first antibody and an immobilized antibody as the second antibody is used.
ィムノメトリック法では、 被検波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 の钪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 wave 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 original and an excess amount of the labeled antibody are allowed to react, 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, the amount of label in either phase is measured to quantify the amount of antigen in the test solution.
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一の散乱を利用するレーザーネフロメト リーなどが好適に用いられる。 In nephelometry, the amount of insoluble sediment generated as a result of an antigen-antibody reaction in a gel or in a solution is measured. The amount of antigen in the test solution is small, Even when only a sediment is obtained, a laser nephrometry utilizing scattering by a laser 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 immunoassays to the quantification method of the present invention, no special conditions, procedures, 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 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 Techniques (Part A))、 同書 Vol . 73 (Immunochemical Techniques (Part B) )、 同書 Vol . 74 (Immunochemical Techniques (Part C))、 同書 Vol. 84 (Immunochemical Techniques (Part For example, edited by Hiro Irie, "Radio Imuno Atsushi" (Kodansha, published in Showa 49) Edited by Hiro Irie "Radio Imno Atssey" (Kodansha, published in Showa 54), edited by Eiji Ishikawa et al. "The Enzyme Immunoassay Method" (2nd edition) (Ed.Ishikawa et al., Ed. Ishikawa et al.), 2nd Edition (Ed.) (3rd Edition) (Medical Publishing, published in 1962), "Methods in ENZYMOLOGYJ Vol. 70 (Immunochemical Techniques (Part A)), Ibid. Vol. 73 (Immunochemical Techniques (Part B)), Ibid. Vol. 74 (Immunochemical Techniques (Part C)), ibid.Vol. 84 (Immunochemical Techniques (Part C)
D: Selec ted I匪翻 assays) )、 同書 Vol . 92 (Immunochemical Techniques (Part E :Monoc lonal Ant ibodies and General Immunoassay Methods) )、 同書 Vo l . 121 (Immunochemical Techniques (Part I: Hybr idoma Technology and Vol. 92 (Immunochemical Techniques (Part E: Monoclonal Ant ibodies and General Immunoassay Methods)), ibid.Vol. 121 (Immunochemical Techniques (Part I: Hybridoma Technology and
Monoc lonal 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。 Monoclonal Ant ibodies)) (above, 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 によって、 本発明のタンパク質の濃度の増加が検出された場合、 例えば、 肺' 気道の炎症を伴う肺 ·胸部疾患などの呼吸器疾患 〔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、 過敏性肺炎など〕 、 アレルギー性結膜炎、 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) 、 消化管疾患 (例、 過敏性腸症候群、 炎 症性腸疾患、 潰瘍性大腸炎、 クローン病、 逆流性食道炎など) などの疾病であ 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 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, lung / thoracic disease accompanied by inflammation of the lung airways Respiratory diseases (eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), allergic conjunctivitis, rhinitis (eg, Allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc., gastrointestinal diseases (eg, irritable bowel) Syndrome, flame (Eg, inflammatory bowel disease, ulcerative colitis, Crohn's disease, reflux esophagitis, etc.) or are 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, preparation of an antibody column used for purifying the protein of the present invention, detection of the protein of the present invention in each fraction at the time of purification, analysis of the behavior of the protein of the present invention in test cells, etc. Can be used for
〔3〕 遺伝子診断薬 [3] Gene diagnostics
本発明の DN Aは、 例えば、 プロ一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 DN Aまたは mRN Aの異常 (遺伝子異常) を検出することができるので、 例えば、 該 DN Aまたは mRN Aの損傷、 突然変異あるいは発現低下や、 該 DNAまたは mRNAの増加ある レ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, as a probe to produce human or warm-blooded animals (eg, rats, mice, guinea pigs, egrets, birds, higgies, pigs, pigs, dogs, cats, cats). (DNA, monkey, chimpanzee, etc.) can detect an abnormality (gene abnormality) in DNA or mRNA that encodes the protein of the present invention or a partial peptide thereof. Mutations or decreased expression or increases in the DNA or mRNA are useful as diagnostic agents for gene expression such as overexpression.
本発明の DNAを用いる上記の遺伝子診断は、 例えば、 公知のノーザンハイ ブリダィゼ一シヨンや P C R— S S C P法 (ゲノミックス (Genomics) , 第 5卷, 874〜879頁 (1989年) 、 プロシ一ジングズ ·ォブ ·ザ ·ナショナル ·ァカデミ ― ·ォブ ·サイェンシィズ ·ォブ ·ュ一エスエー (Proceedings of the  The above-described genetic diagnosis using the DNA of the present invention includes, for example, the well-known Northern hybridization and PCR-SSCP method (Genomics, Vol. 5, pp. 874-879 (1989), Proc. Proceedings of the National Academy
National Academy of Sciences of the United 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。  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ィゼ一ションにより発現過多が検出された場合 や PCR— S SC P法により DN Aの突然変異が検出された場合は、 例えば、 肺 ·気道の炎症を伴う肺 ·胸部疾患などの呼吸器疾患 〔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、 過敏性肺炎など〕 、 アレルギー性結膜炎、 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) 、 消化管疾患 (例、 過敏性腸症候群、 炎症性腸疾患、 潰瘍性大腸炎、 クローン病、 逆流性食道炎など) などの疾病で ある可能性が高いと診断することができる。 For example, when overexpression is detected by Northern hybridization or when a DNA mutation is detected by PCR-SSCP method, for example, lung and chest diseases with inflammation of the lungs and airways Respiratory diseases such as chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, etc., allergic conjunctivitis, rhinitis (eg , Allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc., gastrointestinal diseases (eg, hypersensitivity) Bowel syndrome, It can be diagnosed as being highly likely to be a disease such as inflammatory bowel disease, ulcerative colitis, Crohn's disease, or reflux esophagitis.
〔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 N Aに相補的に結合し、 該 D N Aの発現を抑制することができる 本発明のアンチセンスポリヌクレオチドは低毒性であり、 生体内における本発 明のタンパク質または本発明の D NAの活性 ·機能 (例、 クロライドチャネル 活性など) を抑制することができるので、 例えば、 肺 ·気道の炎症を伴う肺 · 胸部疾患などの呼吸器疾患 〔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、 過敏性肺炎など〕 、 ァレ ルギ一性結膜炎、 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) 、 消化管疾患 (例、 過敏性腸症候群、 炎症性腸疾患、 潰瘍性大腸炎、 クローン病、 逆流性食道炎など) などの予防 ·治療剤、 好ましくは呼吸器疾患、 消化管疾患などの予防 ·治療剤として使用することができる。  The antisense polynucleotide of the present invention, which can complementarily bind to the DNA of the present invention and suppresses the expression of the DNA, has low toxicity and has the activity of the protein of the present invention or the DNA of the present invention in vivo. · Functions (eg, chloride channel activity, etc.) can be suppressed, such as lungs · lungs with inflammation of the airways · respiratory diseases such as chest diseases [eg, chronic obstructive pulmonary disease '(chronic bronchitis, Pulmonary emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), alegiitis conjunctivitis, rhinitis (eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic) Rhinitis, atrophic rhinitis, dry rhinitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc., gastrointestinal diseases (eg, irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease) Reflux esophagitis, etc) agent for the prophylaxis or treatment of such can preferably be used as a prophylactic or therapeutic agent for respiratory diseases, gastrointestinal disorders.
上記アンチセンスポリヌクレオチドを上記の予防 ·治療剤として使用する場 合、 公知の方法に従って製剤化し、 投与することができる。  When the antisense polynucleotide is used as the prophylactic or therapeutic agent, it can be formulated and administered according to a known method.
例えば、 該アンチセンスポリヌクレオチドを用いる場合、 該アンチセンスポ リヌクレオチドを単独あるいはレトロウィルスベクタ一、 アデノウィルスべク ター、 アデノウイルスァソシェ一テッドウィルスベクターなどの適当なベクタ For example, when the antisense polynucleotide is used, the antisense polynucleotide may be used alone or in a suitable vector such as a retrovirus vector, an adenovirus vector, an adenovirus associated virus vector, or the like.
—に挿入した後、 常套手段に従って、 ヒトまたは非ヒト哺乳動物 (例、 ラット、 ゥサギ、 ヒッジ、 ブタ、 ゥシ、 ネコ、 ィヌ、 サルなど) に対して経口的または 非経口的に投与することができる。 該アンチセンスポリヌクレオチドは、 その ままで、 あるいは摂取促進のために補助剤などの生理学的に認めら る担体と ともに製剤化し、 遺伝子銃やハイド口ゲルカテーテルのようなカテーテルによ つて投与できる。 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。 And then orally or parenterally administered to humans or non-human mammals (eg, rats, puppies, higgins, pigs, puppies, cats, dogs, monkeys, etc.) according to standard procedures be able to. The antisense polynucleotide can be administered as it is, or can be formulated with a physiologically acceptable carrier such as an adjuvant to promote uptake, and administered with a gene gun or a catheter such as a hide mouth gel catheter. Alternatively, they can be aerosolized and administered topically into the trachea as an inhalant.
該アンチセンスポリヌクレオチドの投与量は、 対象疾患、 投与対象、 投与ル —トなどにより差異はあるが、 例えば、 慢性閉塞性肺疾患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吸入剤として気管内に局所投与する場合、 一般的に成人 (体重 60kg) においては、 一日につき該アンチセンスポリヌクレ ォチドを約 0. l〜100mg投与する。 The dosage of the antisense polynucleotide may vary depending on the target disease, the target of administration, the route of administration, and the like. For example, the present invention relates to the treatment of chronic obstructive pulmonary disease. When the antisense polynucleotide is locally administered into the trachea as an inhalant, generally, in an adult (body weight 60 kg), about 0.1 to 100 mg of the antisense polynucleotide is administered per day.
さらに、 該ァ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一 ブとして使用することもできる。  Further, the antisense polynucleotide can also be used as a diagnostic oligonucleotide probe for examining the presence or the expression status of the DNA of the present invention in tissues or cells.
本発明は、 さらに  The present invention further provides
(a) 本発明のタンパク質をコードする R N Aの一部を含有する二重鎖 R NA、 (a) a double-stranded RNA containing a part of the RNA encoding the protein of the present invention,
(b) 前記二重鎖 R N Aを含有してなる医薬、 (b) a medicament comprising the double-chain R NA,
(c) 本発明のタンパク質をコードする R N Aの一部を含有するリポザィム、 (d) 前記リポザィムを含有してなる医薬を提供する。  (c) a lipozyme containing a part of RNA encoding the protein of the present invention; and (d) a medicament containing the lipozyme.
これらの二重鎖 R NA (RNAi; RNA interference法) 、 リポザィムなどは、 上記アンチセンスポリヌクレオチドと同様に、 本発明のポリヌクレオチド (例、 D NA) の発現を抑制することができ、 生体内における本発明のペプチドまた は本発明のポリヌクレオチド (例、 D NA) の活性や機能 (例、 クロライドチ ャネル活性など) を阻害することができるので、 例えば、 肺 ·気道の炎症を伴 う肺,胸部疾患 〔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、 過敏性肺炎など〕 、 炎症性腸疾患、 アレルギー性結膜炎、 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) などの予防 ·治療剤として使用することができる。  These double-stranded RNAs (RNAi; RNA interference method), lipozymes, and the like can suppress the expression of the polynucleotide (eg, DNA) of the present invention in the same manner as the above-mentioned antisense polynucleotides. Can inhibit the activity or function (eg, chloride channel activity, etc.) of the peptide of the present invention or the polynucleotide (eg, DNA) of the present invention in, for example, lungs with lung and airway inflammation, Chest disease (eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), inflammatory bowel disease, allergic conjunctivitis, rhinitis (Eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrenous rhinitis, sinusitis, etc.) It can be used as anti-therapeutic agent.
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。 リポザィムは、 公知の方法 (例、 TRENDS in Molecular Medi c ine, 7巻, 221 頁, 2001年) に準じて、 本発明のポリヌクレオチドの配列を基に設計して製造 することができる。 例えば、 本発明のペプチドをコードする R N Aの一部に公 知のリポザィムを連結することによつて製造することができる。 本発明のぺプ チドをコードする R NAの一部としては、 公知のリポザィムによって切断され 得る本発明の R NA上の切断部位に近接した部分 (R N A断片) が挙げられる。 上記の二重鎖 R NAまたはリポザィムを上記予防 ·治療剤として使用する場 合、 7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 さる。 〔5〕 本発明の抗体を含有する医薬 The double-stranded RNA can be designed and manufactured 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 produced 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 RNA encoding the peptide of the present invention. As a part of the RNA encoding the peptide of the present invention, a portion (RNA fragment) close to the cleavage site on the RNA of the present invention, which can be cleaved by a known lipozyme, can be mentioned. When the above-mentioned double-stranded RNA or lipozyme is used as the above-mentioned prophylactic / therapeutic agent, it can be formulated and administered in the same manner as for the antisense polynucleotide. [5] A drug containing the antibody of the present invention
本発明のタンパク質の活性を中和する作用を有する本発明の抗体は、 例えば、 肺,気道の炎症を伴う肺 ·胸部疾患などの呼吸器疾患 〔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、 過敏性肺炎など〕 、 アレルギ一性結膜炎、 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) 、 消化管疾患 (例、 過敏性腸症候群、 炎症性腸疾患、 潰瘍性大腸炎、 クローン病、 逆流性食道炎など) などの疾患に 対する医薬として使用することができる。  Antibodies of the present invention having an activity of neutralizing the activity of the protein of the present invention include, for example, respiratory diseases such as lung and chest diseases accompanied by inflammation of the lungs and airways [eg, chronic obstructive pulmonary disease (chronic bronchitis, Emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), allergic conjunctivitis, rhinitis (eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, Atrophic rhinitis, dry rhinitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc., gastrointestinal diseases (eg, irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease, reflux disease) It can be used as a medicine for diseases such as esophagitis.
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、 ヒトまたは非ヒト哺乳動 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。 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、 成人の慢性閉塞 性肺疾患の治療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. 01〜20mg/kg体重程度、 好ましくは 0. l〜10mg/kg体重程度、 さらに好ましく は 0. 1〜5rag/kg体重程度を、 1日 1〜5回程度、 好ましくは 1日 1〜3回程度、 静脈注 射により投与するのが纾都合である。 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 directly as a liquid or as a pharmaceutical composition of an appropriate dosage form in human or non-human mammals (eg, rat, egret, hidge) Or parenteral administration to mice, dogs, 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 of chronic obstructive pulmonary disease in adults, the antibody of the present invention is used as a single dose. Usually, about 0.01 to 20 mg / kg body weight, preferably about 0.1 to 10 mg / kg body weight, more preferably about 0.1 to 5 rag / kg body weight, about 1 to 5 times a day, preferably about 1 to 5 rag / kg body weight. It is convenient to administer by intravenous injection about once to three times a day. In the case of other parenteral administration and oral administration, an equivalent dose can be administered. If the symptoms are particularly severe, the dose may be increased accordingly.
本発明の抗体は、 それ自体または適当な医薬組成物として投与することがで きる。 上記投与に用いられる医薬組成物は、 上記抗体またはその塩と薬理学的 に許容され得る担体、 希釈剤もしくは賦形剤とを含むものである。 かかる組成 物は、 経口または非経口投与に適する剤形として提供される。  The antibodies of the present invention can be administered by themselves or as a suitable pharmaceutical composition. The pharmaceutical composition used for the administration contains the antibody or a salt thereof and a pharmacologically acceptable carrier, diluent or excipient. Such compositions are provided in dosage forms suitable for oral or parenteral administration.
すなわち、 例えば、 経口投与のための組成物としては、 固体または液体の剤 形、 具体的には錠剤 (糖衣錠、 フィルムコーティング錠を含む) 、 丸剤、 顆粒 剤、 散剤、 カプセル剤 (ソフトカプセル剤を含む) 、 シロップ剤、 乳剤、 懸濁 剤などがあげられる。 かかる組成物は公知の方法によって製造され、 製剤分野 において通常用いられる担体、 希釈剤もしくは賦形剤を含有するものである。 例えば、 錠剤用の担体、 賦形剤としては、 乳糖、 でんぷん、 蔗糖、 ステアリン 酸マグネシウムなどが用いられる。 That is, for example, as a composition for oral administration, a solid or liquid agent Examples include tablets (including sugar-coated tablets and film-coated tablets), pills, granules, powders, capsules (including soft capsules), syrups, emulsions, and suspensions. Such a composition is produced by a known method and contains a carrier, diluent or excipient commonly used in the pharmaceutical field. 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、 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、 ポリソルべ一ト 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って調製される。  As compositions for parenteral administration, for example, injections, suppositories, etc. are used. Injections are in the form of intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections, infusions, etc. Is included. Such injections are prepared according to a known method, for example, by dissolving, suspending or emulsifying the above antibody or a salt thereof in a sterile aqueous or oily liquid usually used for injections. As an aqueous liquid for injection, for example, physiological saline, isotonic solution containing glucose and other adjuvants, and the like, suitable solubilizing agents, for example, alcohol (eg, ethanol), polyalcohol (eg, It may be used in combination with propylene glycol, polyethylene glycol), a nonionic surfactant [eg, polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated catalyst)], and the like. 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 a suitable ampoule. A suppository for rectal administration is prepared by mixing the above antibody or a salt thereof with a usual suppository base.
上記の経口用または非経口用医薬組成物は、 活性成分の投与量に適合するよ うな投薬単位の剤形に調製されることが好都合である。 かかる投薬単位の剤形 としては、 錠剤、 丸剤、 カプセル剤、 注射剤 (アンプル) 、 坐剤などが例示さ れ、 それぞれの投薬単位剤形当たり通常 5〜500mg、 とりわけ注射剤では 5〜  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 forms include tablets, pills, capsules, injections (ampoules), and suppositories, and usually 5 to 500 mg per dosage unit form, and especially 5 to 500 mg for injections.
100mg、 その他の剤形では 10〜250mgの上記抗体が含有されていることが好まし い。 It is preferred that 100 mg and other dosage forms contain 10 to 250 mg of the above antibody.
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。 〔6〕 本発明のタンパク質または D N Aを含有する医薬 Each of the above-mentioned compositions may contain other active ingredients as long as the composition does not cause an undesirable interaction with the above-mentioned antibody. [6] A drug containing the protein or DNA of the present invention
本発明のタンパク質 (好ましくは部分ペプチド) は、 例えば、 肺 ·気道の炎 症を伴う肺 ·胸部疾患などの呼吸器疾患 〔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 炎など〕 、 アレルギー性結膜炎、 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) 、 消化管疾患 (例、 過敏性腸症候群、 炎症性腸疾 患、 潰瘍性大腸炎、 クローン病、 逆流性食道炎など) などの予防 ·治療剤、 好 ましくは呼吸器疾患、 消化管疾患などの予防 ·治療剤としても有用である。 本 発明のタンパク質を上記予防 ·治療剤として使用する場合は、 常套手段に従つ て製剤化することができる。  The protein (preferably a partial peptide) of the present invention may be used, for example, for respiratory diseases such as lungs, lungs with respiratory tract inflammation, and chest diseases [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse pan- Bronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), allergic conjunctivitis, rhinitis (eg, allergic rhinitis, pollinosis, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dryness Pronasalitis, vasomotor rhinitis, gangrenous rhinitis, sinusitis, etc., gastrointestinal disorders (eg, irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease, reflux esophagitis, etc.) It is also useful as a prophylactic / therapeutic agent, preferably as a prophylactic / therapeutic agent for respiratory diseases and gastrointestinal diseases. When the protein of the present invention is used as the above-mentioned prophylactic / therapeutic agent, it can be formulated according to a conventional method.
例えば、 本発明のタンパク質は、 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 'に認められる公知の担体、 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。  For example, the protein of the present invention can be used as a tablet, capsule, elixir, microcapsule, etc., if necessary, orally coated with water or other pharmaceutically acceptable liquids. It can be used parenterally in the form of injections, such as sterile solutions or suspensions. For example, the protein of the present invention may be used together with known physiologically acceptable carriers, flavoring agents, excipients, vehicles, preservatives, stabilizers, binders, etc., in a unit dosage form generally required for the practice of the formulation. It can be manufactured by mixing with. The amount of the active ingredient in these preparations is such that a suitable dosage in the specified range can be obtained.
本発明のタンパク質の投与量は、 投与対象、 対象臓器、 症状、 投与方法など により差異はあるが、 経口投与の場合、 一般的に例えば、 患者 (60kgとして) においては、 一日につき約 0. 1〜100mg、 好ましくは約 1. 0〜50mg、 より好ましく は約 1. 0〜20m gである。 非経口的に投与する場合は、 その 1回投与量は投与対 象、 対象臓器、 症状、 投与方法などによっても異なるが、 例えば、 注射剤の形 では通常例えば、 患者 (60kgとして) においては、 一日につき約 0. 01〜30mg程 度、 好ましくは約 0. l〜20mg程度、 より好ましくは約 0. 1〜10mg程度を静脈注射 により投与するのが好都合である。 他の動物の場合も、 60kg当たりに換算した 量を投与することができる。 〔7〕 本発明の DNAを有する動物の作出 The dose of the protein of the present invention varies depending on the administration subject, target organ, symptoms, administration method, and the like. However, in the case of oral administration, generally, for example, in a patient (as 60 kg), about 0. It is 1 to 100 mg, preferably about 1.0 to 50 mg, more preferably about 1.0 to 20 mg. In the case of parenteral administration, the single dose varies depending on the target of administration, target organ, symptoms, administration method, etc., for example, usually in the form of injection, for example, in patients (as 60 kg), It is convenient to administer about 0.01 to 30 mg, preferably about 0.1 to 20 mg, more preferably about 0.1 to 10 mg per day by intravenous injection. In the case of other animals, the dose can be administered in terms of 60 kg. [7] Creation of an animal having the DNA of the present invention
本発明は、 外来性の本発明のタンパク質をコードする DNA (以下、 本発明 の外来性 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 DN Aと略記する場合がある) を有する非ヒト哺乳動物を提供する。  The present invention relates to a DNA encoding an 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 DN Aまたはその変異 DN Aを有する非ヒト哺乳動物、 (1) a non-human mammal having an exogenous DNA of the present invention or a mutant DNA thereof,
(2) 非ヒ卜哺乳動物がゲッ歯動物である上記 (1) 記載の動物、 (2) the animal according to (1), wherein the non-human mammal is a rodent;
(3) ゲッ歯動物がマウスまたはラットである上記 (2) 記載の動物、 および (3) The animal according to (2), wherein the rodent is a mouse or a rat, and
(4) 本発明の外来性 DNAまたはその変異 DNAを含有し、 哺乳動物におい て発現しうる組換えべクタ一などを提供する。 (4) A recombinant vector containing the exogenous DNA of the present invention or its mutant DNA, which can be expressed in mammals, and the like.
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導入動物と略記する) は、 未受精卵、 受精卵、 精子およびその 始原細胞を含む胚芽細胞などに対して、 好ましくは、 非ヒト哺乳動物の発生に おける胚発生の段階 (さらに好ましくは、 単細胞または受精卵細胞の段階でか つ一般に 8細胞期以前) に、 リン酸カルシウム法、 電気パルス法、 リボフェク 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、 DE AE—デキストラン法などにより目的とする DN Aを導入することによって作 出することができる。 また、 該 DNA導入方法により、 体細胞、 生体の臓器、 組織細胞などに目的とする本発明の外来性 DNAを導入し、 細胞培養、 組織培 養などに利用することもでき、 さらに、 これら細胞を上述の胚芽細胞と公知の 細胞融合法により融合させることにより本発明の DN A導入動物を作出するこ ともできる。  Non-human mammals having the exogenous DNA of the present invention or the mutant DNA thereof (hereinafter abbreviated as the DNA-transfected animal of the present invention) can be used for non-fertilized eggs, fertilized eggs, spermatozoa and germ cells including their progenitor cells. Preferably, during the stage of embryonic development in non-human mammal development (more preferably, at the stage of a single cell or fertilized egg and generally before the 8-cell stage), the calcium phosphate method, the electric pulse method, the ribofection method, It can be created by introducing the desired DNA by agglutination, microinjection, particle gun, DEAE-dextran, etc. Further, the exogenous DNA of the present invention can be introduced into somatic cells, organs of living organisms, tissue cells, and the like by the DNA introduction method, and used for cell culture, tissue culture, and the like. Can be fused with the above-mentioned germ cells by a known cell fusion method to produce the DNA-introduced animal of the present invention.
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、 C 57BL/6系統, DBA 2系統など、 交雑系として、 B 6 C3F,系統, B DF,系統, B 6D2F,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。 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。 As the non-human mammal, for example, porcupine, pig, sheep, goat, goat, egret, dog, cat, guinea pig, hamster, mouse, rat 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 reproduction are particularly useful in mice (for example, pure strains such as C57BL / 6 strain and DBA2 strain). As the crossing strain, B6C3F, strain, BDF, strain, B6D2F, strain, BALB / c strain, ICR strain, etc.) or rat (eg, Wistar, SD, etc.) are preferable. "Mammals" in a recombinant vector that can be expressed in mammals include humans in addition to the above-mentioned non-human mammals.
本発明の外来性 D N Aとは、 非ヒト哺乳動物が本来有している本発明の D N Aではなく、 いったん哺乳動物から単離 ·抽出された本発明の D NAをいう。 本発明の変異 D NAとしては、 元の本発明の D N 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 D NAなどが用いられ、 また、 異常 D N Aも含まれる。 該異常 D N Aとしては、 異常な本発明のタンパク質を発現させる D N 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 D NAなどが用いられる。  The exogenous DNA of the present invention refers not to the DNA of the present invention originally possessed by non-human mammals, but to the DNA of the present invention once isolated and extracted from the mammal. The mutant DNA of the present invention is a DNA in which a mutation (for example, mutation) has occurred in the base sequence of the original DNA of the present invention, specifically, base addition, deletion, substitution with another base. DNA or the like that has occurred is used, and abnormal DNA is also included. As the abnormal DNA, DNA that expresses an abnormal protein of the present invention is used. For example, DNA that expresses a protein that suppresses the function of the normal protein of the present invention is used.
本発明の外来性 D NA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 D N Aを対象動物に導入させるに 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 D NA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 D NAを導入させる場合、 これと相同性が高い本発明の D NA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 D NA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 D NAを結合した D NAコンストラクト (例、 ベクタ一など) を対 象哺乳動物の受精卵、 例えば、 マウス受精卵へマイクロインジェクションする ことによって本発明の D N Aを高発現する D N A導入哺乳動物を作出すること ができる。  The exogenous DNA of the present invention may be derived from a mammal that is the same or different from the animal of interest. In introducing the DNA of the present invention into a target animal, it is generally advantageous to use the DNA as a DNA construct linked downstream of a promoter capable of being expressed in animal cells. For example, when the human DNA of the present invention is introduced, it is derived from various mammals (eg, egrets, dogs, cats, guinea pigs, hamsters, rats, mice, etc.) having the DNA of the present invention that are highly homologous thereto. Microinjection of a DNA construct (eg, a vector, etc.) to which the human DNA of the present invention is bound downstream of various promoters capable of expressing the DNA of the present invention into a fertilized egg of a target mammal, for example, a mouse fertilized egg As a result, a mammal into which the DNA of the present invention is highly expressed can be produced.
本発明のタンパク質の発現べクタ一としては、 大腸菌由来のプラスミド、 枯 草菌由来のプラスミド、 酵母由来のプラスミド、 λファー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ィルスなどが用いられる。 なかでも、 大 腸菌由来のプラスミド、 枯草菌由来のプラスミドまたは酵母由来のプラスミド などが好ましく用いられる。  Examples of the expression vector of the protein of the present invention include a plasmid derived from Escherichia coli, a plasmid derived from Bacillus subtilis, a plasmid derived from yeast, a bacteriophage such as λ phage, a retrovirus such as Moroni leukemia virus, a vaccinia virus or a baculovirus. For example, animal viruses such as E. coli are used. Among them, a plasmid derived from E. coli, a plasmid derived from Bacillus subtilis or a plasmid derived from yeast are preferably used.
上記の D NA発現調節を行なうプロモー夕一としては、 例えば、 ウィルス (例、 シミアンウィルス、 サイトメガロウィルス、 モロニ一白血病ウィルス、 J Cウィルス、 乳癌ウィルス、 ポリオウイルスなど) に由来する DNAのプロ モーター、 各種哺乳動物 (ヒト、 ゥサギ、 ィヌ、 ネコ、 モルモット、 ハムス夕 ―、 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、 筋クレアチンキナ一ゼ、 グリア線維性酸性タンパク質、 ダルタチオン S_ トランスフェラ一ゼ、 血小板由来成長因子 i3、 ケラチン K 1, 1 0ぉょび 14、 コラーゲン I型および I 1¾、 サイクリック AMP依存タンパク質キナ —ゼ 61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一 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一ゼ (一般に Τ i e 2と略される) 、 ナトリウムカリウムアデノシン 3リン酸化酵素 (N a, 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組織インヒビ夕一、 MHCクラス I抗原 (H - 2 L) : H— r a s、 レニン、 ド一パミン —水酸化酵素、 甲状腺ペルォキ シダ一ゼ (TP〇) 、 ポリペプチド鎖延長因子 1 a (EF- 1 α) 、 β了ゥチ ン、 αおよび /3ミオシン重鎖、 ミオシン軽鎖 1および 2、 ミエリン基礎タンパ ク質、 チログロブリン、 Th y— 1、 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 (:、 平滑筋 αァ クチン、 プレプロエンケフアリン Α、 バソプレシンなどのプロモータ一などが 用いられる。 なかでも、 全身で高発現することが可能なサイトメガロウィルス プロモーター、 ヒトポリペプチド鎖延長因子 1 ひ (EF- 1 ) のプロモ一夕 一、 ヒトおよびニヮトリ i3ァクチンプロモ一ターなどが好適である。 Examples of promoters that regulate DNA expression include, for example, viruses (eg, Simian virus, cytomegalovirus, Moroni leukemia virus, Promoters of DNA derived from JC virus, breast cancer virus, poliovirus, etc., promoters derived from various mammals (human, egret, dog, cat, guinea pig, hams, etc., rat, mouse, etc.), for example, albumin, Insulin II, peroplakin II, Erasuyose, erythropoietin, endothelin, muscle creatine kinase, glial fibrillary acidic protein, daltathione S_transferase, platelet-derived growth factor i3, keratin K 1,10 And 14, collagen type I and I 1¾, cyclic AMP-dependent protein kinase — 61 subunit, dystrophin, tartrate-resistant alkaline phosphatase, atrial natriuretic factor, endothelial receptor chin mouth kinase (generally Τ ie Abbreviated as 2), sodium potassium Denosine 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-hydroxylase, thyroid peroxidase (TP〇), polypeptide chain elongation factor 1a (EF-1α), β-rhtin, α and / 3 myosin heavy chain, myosin Light chains 1 and 2, myelin-based protein, thyroglobulin, Thy-1, immunoglobulin, heavy-chain variable region (VNP) 'serum amyloid P component, myoglobin, troponin (:, smooth muscle α-actin, preproenke A promoter such as hualin リ ン or vasopressin is used, among which a cytomegalovirus promoter capable of high expression throughout the body, a human polypeptide chain extension, etc. Factor 1 flight (EF-1) promo Isseki one, such as humans and Niwatori i3 Akuchinpuromo one coater is preferred.
上記べクタ一は、 DNA導入哺乳動物において目的とするメッセンジャー R NAの転写を終結する配列 (一般にターミネタ一と呼ばれる) を有しているこ とが好ましく、 例えば、 ウィルス由来および各種哺乳動物由来の各 DNAの配 列を用いることができ、 好ましくは、 シミアンウィルスの SV40夕一ミネ夕 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-transfected mammal (generally called terminator). For example, it is derived from viruses and various mammals. A sequence of each DNA can be used, and preferably, Simian virus SV40, Minne, etc. 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、 ェンハンサ一領域、 真核 DN Aのイントロンの一部な どをプロモータ一領域の 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, translation of splicing signal of each DNA, enhancer region, part of intron of eukaryotic DNA, etc. to 5 'upstream of promoter region, promoter region, etc. for the purpose of further expressing the target foreign DNA Inter-domain or translation It is also possible to connect 3 ′ downstream of the 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、 モルモッ卜、 ハムスター、 ラット、 マウスなど) 由来の S=F臓、 腎臓、 甲状腺細胞、 線維芽細胞由来 D NAおよび市販の各種ゲノム D N Aライブラリ一よりゲノム D NAの全てあるいは一部として、 または肝臓、 腎 臓、 甲状腺細胞、 線維芽細胞由来 R NAより公知の方法により調製された相補 D NAを原料として取得することが出来る。 また、 外来性の異常 D NAは、 上 記の細胞または組織より得られた正常なポリぺプチドの翻訳領域を点突然変異 誘発法により変異した翻訳領域を作製することができる。  The normal translation region of the protein of the present invention may be S = F kidney, kidney, thyroid cell, fibroblast derived from human or various mammals (eg, egret, dog, cat, guinea pig, hamster, rat, mouse, etc.). Complementary DNA prepared by known methods from cell-derived DNA and all or part of genomic DNA from one of various commercially available genomic DNA libraries, or from liver, kidney, thyroid cells, and fibroblast-derived RNA. It can be obtained as a raw material. In addition, a foreign abnormal DNA can produce a translation region obtained by mutating a normal polypeptide translation region obtained from the above cells or tissues by a point mutagenesis method.
該翻訳領域は D N A導入動物において発現しうる D N Aコンストラクトとし て、 前記のプロモーターの下流および所望により'転写終結部位の上流に連結さ せる通常の 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 DNA-transduced 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 D N Aの導入は、 対象哺乳動物の胚 芽細胞および体細胞のすべてに存在するように確保される。 D NA導入後の作 出動物の胚芽細胞において、 本発明の外来性 D N Aが存在することは、 作出動 物の後代がすべて、 その胚芽細胞および体細胞のすべてに本発明の外来性 D N Aを保持することを意味する。 本発明の外来性 D 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すべてに本発明の外来性 D NAを有す る。  Introduction of the exogenous DNA of the present invention at the fertilized egg cell stage is ensured to be present in all germ cells and somatic cells of the target mammal. The presence of the exogenous DNA of the present invention in the germinal cells of the produced animal after the introduction of the DNA indicates that all the progeny of the produced animal retain the exogenous DNA of the present invention in all of the germ cells and somatic cells. Means to do. Progeny of such animals that inherit the foreign DNA of the present invention have the foreign DNA of the present invention in all of their germinal 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 maintain the exogenous DNA by mating, and is subcultured as an animal having the DNA in a normal breeding environment. Can be done.
受精卵細胞段階における本発明の外来性 D N Aの導入は、 対象哺乳動物の胚 芽細胞および体細胞の全てに過剰に存在するように確保される。 D NA導入後 の作出動物の胚芽細胞において本発明の外来性 D 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 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外来性 D N Aを過 剰に有する。 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を過剰に有するように 繁殖継代することができる。 The introduction of the exogenous DNA of the present invention at the fertilized egg cell stage is ensured to be present in excess in all germ cells and somatic cells of the target mammal. Excessive presence of the exogenous DNA of the present invention in the germinal cells of the produced animal after the introduction of the DNA indicates that all the offspring of the produced animal have the exogenous DNA of the present invention in all of the germ cells and somatic cells. Means that. The offspring 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 germ cells and somatic cells. By obtaining a homozygous animal having the introduced DNA on both homologous chromosomes and mating the male and female animals, it is possible to breed and passage so that all offspring have the DNA in excess.
本発明の正常 D NAを有する非ヒ卜哺乳動物は、 本発明の正常 D NAが高発 現させられており、 内在性の正常 D 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 NA導入動物を用いて、 本発明のタンパク質の機能亢進症や、 本発明のタンパク質が関連する疾患の病 態機序の解明およびこれらの疾患の治療方法の検討を行なうことが可能である。 また、 本発明の外来性正常 D NAを導入させた哺乳動物は、 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、 本発明の夕ンパク質に関連する疾 患に対する治療薬のスクリーニング試験にも利用可能である。  The non-human mammal having the normal DNA of the present invention expresses the normal DNA of the present invention at a high level, and eventually promotes the function of endogenous normal DNA, thereby finally obtaining the protein of the present invention. It may develop quality hyperfunction and can be used as a model animal for the disease. For example, using the normal DNA-introduced animal of the present invention, elucidation of the pathological mechanism of hyperactivity of the protein of the present invention and diseases associated with the protein of the present invention, and examination of a method for treating these diseases. It is possible. Further, since the mammal into which the exogenous normal DNA of the present invention has been introduced has an increased symptom of the released protein of the present invention, screening for a therapeutic agent for the disease associated with the protein of the present invention is performed. It can also be used for testing.
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 環境で継代飼育することが出来る。 さらに、 目的とする外来 D NAを前述の プラスミドに組み込んで原科として用いることができる。 プロモ一ターとの D N Aコンストラク卜は、 通常の D N A工学的手法によって作製することができ る。 受精卵細胞段階における本発明の異常 D N Aの導入は、 対象哺乳動物の胚 芽細胞および体細胞の全てに存在するように確保される。 D NA導入後の作出 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 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一ト動物を取得し、 この雌雄の動物を交配す ることによりすべての子孫が該 D N Aを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 foreign abnormal DNA of the present invention can be subcultured in a normal breeding environment as an animal having the DNA after confirming that the foreign DNA is stably maintained by the crossing. I can do it. Furthermore, 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 ordinary DNA engineering techniques. Introduction of the abnormal DNA of the present invention at the fertilized egg cell stage is ensured to be present in all germinal and somatic cells of the target mammal. The presence of the abnormal DNA of the present invention in the germ cells of the produced animal after the introduction of the DNA means that all the offspring of the produced animal have the abnormal DNA of the present invention in all of the germ cells and somatic cells. The progeny of this type of animal that has inherited the exogenous DNA of the present invention has the abnormal DNA of the present invention in all of its germinal and somatic cells. By obtaining a homozygous animal having the introduced DNA on both homologous chromosomes, and crossing the male and female animals, it is possible to breed subculture 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 NAが高発 現させられており、 内在性の正常 D N Aの機能を阻害することにより最終的に 本発明のタ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 N A導入動物を用 いて、 本発明のタンパク質の機能不活性型不応症の病態機序の解明およびこの 疾患を治療方法の検討を行なうことが可能である。 In the non-human mammal having the abnormal DNA of the present invention, the abnormal DNA of the present invention is highly expressed, and the function of the protein of the present invention is ultimately reduced by inhibiting the function of endogenous normal DNA. Inactive type refractory disease Can be used as an animal. For example, using the abnormal DNA-introduced animal of the present invention, it is possible to elucidate the pathological mechanism of the function-inactive refractory of the protein of the present invention and to examine a method for treating this disease.
また、 具体的な利用可能性としては、 本発明の異常 D N A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。  Further, as a specific possibility, the abnormal DNA highly expressing animal of the present invention can be used to inhibit the function of a normal protein by the abnormal protein of the present invention in the function-inactive refractory disease of the protein of the present invention (dominant negative active protein). Action).
また、 本発明の外来異常 D N Aを導入させた哺乳動物は、 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またはその機能 不活性型不応症に対する治療薬スクリーニング試験にも利用可能である。  In addition, since the mammal into which the foreign abnormal DNA of the present invention has been introduced has an increased symptom of the released protein of the present invention, it can be used in a therapeutic drug screening test for the protein of the present invention or its function-inactive refractory disease. Is also available.
また、 上記 2種類の本発明の D N A導入動物のその他の利用可能性として、 例えば、  Further, as other possible uses of the above two kinds of DNA-introduced animals of the present invention, for example,
(a) 組織培養のための細胞源としての使用、  (a) use as a cell source for tissue culture,
(b) 本発明の D NA導入動物の組織中の D NAもしくは R N Aを直接分析する、 または D NAにより発現されたポリペプチド組織を分析することによる、 本発 明のタンパク質により特異的に発現あるいは活性化するタンパク質との関連性 についての解析、  (b) direct analysis of DNA or RNA in the tissue of the DNA-introduced animal of the present invention, or analysis of a polypeptide tissue expressed by DNA, to specifically express or express the protein of the present invention. Analysis of the relationship with the activating protein,
(c) D NAを有する組織の細胞を標準組織培養技術により培養し、 これらを使 用して、 一般に培養困難な組織からの細胞の機能の研究、  (c) Cells of a tissue having DNA are cultured by standard tissue culture techniques, and these are used to study the function of cells from tissues that are generally difficult to culture,
(d) 上記 (c) 記載の細胞を用いることによる細胞の機能を高めるような薬剤 のスクリーニング、 および '  (d) screening for a drug that enhances cell function by using the cell described in (c) above; and
(e) 本発明の変異タンパク質を単離精製およびその抗体作製などが考えられる。 さらに、 本発明の D NA導入動物を用いて、 本発明のタンパク質の機能不活 性型不応症などを含む、 本発明のタンパク質に関連する疾患の臨床症状を調べ ることができ、 また、 本発明のタンパク質に関連する疾患モデルの各臓器にお けるより詳細な病理学的所見が得られ、 新しい治療方法の開発、 さらには、 該 疾患による二次的疾患の研究および治療に貢献することができる。  (e) Isolation and purification of the mutant protein of the present invention and production of its antibody can be considered. Further, using the DNA-introduced animal of the present invention, it is possible to examine clinical symptoms of diseases related to the protein of the present invention, including refractory inactivation of the protein of the present invention. It is possible to obtain more detailed pathological findings in each organ of the disease model related to the protein of the present invention, to develop new treatment methods, and to contribute to the research and treatment of secondary diseases caused by the disease. it can.
また、 本発明の D NA導入動物から各臓器を取り出し、 細切後、 トリプシン などのタンパク質分解酵素により、 遊離した D NA導入細胞の取得、 その培養 またはその培養細胞の系統化を行なうことが可能である。 さらに、 本発明の夕 ンパク質産生細胞の特定化、 アポ卜一シス、 分化あるいは増殖との関連性、 ま たはそれらにおけるシグナル伝達機構を調べ、 それらの異常を調べることなど ができ、 本発明のタンパク質およびその作用解明のための有効な研究材料とな る。 In addition, each organ is removed from the DNA-introduced animal of the present invention, cut into small pieces, and the DNA-introduced cells that have been released with a protease such as trypsin are obtained and cultured. Alternatively, it is possible to systematize the cultured cells. Furthermore, the present inventors can identify the protein-producing cells of the present invention, examine their relationship to apoptosis, differentiation or proliferation, or examine their signal transduction mechanisms, and investigate their abnormalities. It is an effective research material for elucidating the protein and its action.
さらに、 本発明の DN A導入動物を用いて、 本発明のタンパク質の機能不活 性型不応症を含む、 本発明のタンパク質に関連する疾患の治療薬の開発を行な うために、 上述の検査法および定量法などを用いて、 有効で迅速な該疾患治療 薬のスクリーニング法を提供することが可能となる。 また、 本発明の DN A導 入動物または本発明の外来性 DN A発現ベクターを用いて、 本発明のタンパク 質が関連する疾患の DN A治療法を検討、 開発することが可能である。  Further, the use of the DNA-introduced animal of the present invention to develop a therapeutic agent for a disease associated with the protein of the present invention, including refractory inactive type of the protein of the present invention, Using a test method and a quantitative method, it is possible to provide an effective and rapid screening method for a therapeutic agent for the disease. Also, using the DNA-introduced animal of the present invention or the exogenous DNA-expressing vector of the present invention, it is possible to examine and develop a method for treating a 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 DNAが不活性化された非ヒト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 DN Aが不活性化された非ヒト哺乳動物胚幹細胞、  (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 β—ガラクトシダ一ゼ遺 伝子) を導入することにより不活性化された上記 (1) 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, a β-galactosidase enzyme gene derived from E. coli).
(3) ネオマイシン耐性である上記 (1) 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上記 (1) 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上記 (4) 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発現不全非ヒト哺乳動物、 (6) a DNA-deficient non-human mammal in which the DNA of the present invention has been inactivated,
(7) 該 DNAがレポーター遺伝子 (例、 大腸菌由来の β—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、 ¾レポ一夕一遺伝子が本発明の D(7) The DNA is inactivated by introducing a reporter gene (eg, a β-galactosidase gene derived from Escherichia coli), and
ΝΑに対するプロモーターの制御下で発現しうる上記 (6) 記載の非ヒト哺乳 動物、 The non-human mammal according to the above (6), which can be expressed under the control of a promoter for ΝΑ,
(8) 非ヒト哺乳動物がゲッ歯動物である上記 (6) 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上記 (8) 記載の非ヒト哺乳動物、 および (10) 上記 (7) 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 DN Aに対するプロモーター活性を 促進または阻害 (好ましくは阻害) 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。 (9) The non-human mammal according to (8), wherein the rodent is a mouse; and (10) A compound that promotes or inhibits (preferably inhibits) the promoter activity of DNA of the present invention, which comprises administering a test compound to the animal according to (7) above and detecting the expression of a reporter gene. A method for screening the salt is provided.
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、 DNAの 発現能を抑制するか、 あるいは該 DNAが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、 D N 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 a DNA which is obtained by artificially mutating the DNA of the present invention possessed by the non-human mammal, thereby suppressing the DNA expression ability, Alternatively, the DNA substantially does not have the ability to express the protein of the present invention by substantially losing the activity of the protein of the present invention encoded by the DNA (hereinafter referred to as the knockout DNA of the present invention). In some cases, refers to embryonic stem cells of non-human mammals (hereinafter abbreviated as ES cells).
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 DN A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 DN A配列の一部又は全部の削除、 他 DN Aを挿入または置換 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、 プロモータ一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 DNAを作製すればよい。  The method of artificially mutating the DNA of the present invention can be performed, for example, by deleting a part or all of the DNA sequence and inserting or substituting another DNA by a genetic engineering technique. The knockout DNA of the present invention may be prepared by, for example, shifting the reading frame of a codon or disrupting the function of a promoter or exon by these mutations.
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 DNA不活性化 E S細胞または本発明のノックアウト 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 l a c Z (β—ガラクトシダ一 ゼ遺伝子) 、 c a t (クロラムフエニコールァセチルトランスフェラ一ゼ遣伝 子) を代表とするレポ一ター遺伝子等を挿入することによりェキソンの機能を 破壊するか、 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、 p o l yA付加シグナルなど) を挿入し、 完全なメッ センジャー RN A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 DNA配列を有する DNA鎖 (以下、 夕一ゲッティングべク 夕一と略記する) 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 DN A上あるいはその近傍の DN A配列をプ 口一ブとしたサザンハイブリダィゼ一シヨン解析あるいは夕一ゲッティングべ クタ一上の DN A配列と夕一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 DN A配列をプライマーとした PC R法により解析し、 本発明のノックアウト E S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, for example, The DNA of the present invention possessed by a non-human mammal is isolated, and its exon portion is a drug resistance gene typified by a neomycin resistance gene, a hygromycin resistance gene, lacZ (β-galactosidase gene), cat (clo DNA that disrupts exon function by inserting a reporter gene such as the ramphenicol acetyltransferase gene) or terminates gene transcription in the intron between exons. By inserting a sequence (eg, a polyA additional signal) and preventing the synthesis of the complete messenger RNA, A DNA strand having a DNA sequence constructed so as to disrupt the gene (hereinafter abbreviated as “Yuichi Getting Vector”) was introduced into the chromosome of the animal by, for example, homologous recombination, and the obtained ES was obtained. For a cell, the DNA sequence on or near the DNA of the present invention is purified. The DNA sequence on the Southern hybridization analysis or evening getter vector and the DNA sequence of the neighboring region other than the DNA of the present invention used in the evening getter vector were prepared. It can be obtained by analyzing by the PCR method using primers 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 な Ev an sと Kau ί m a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 DBA/ 2との交雑 により改善した BDFiマウス (C 57 BL/6と DBAZ2との を用い て樹立したものなども良好に用いうる。 BDFi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 BLZ 6マウスを背景に持つので、 これを用いて得られた ES細胞は病態モデルマウスを作出したとき、 C57B LZ 6マウスとバッククロスすることでその遺伝的背景を C 57 BLZ6マウ スに代えることが可能である点で有利に用い得る。  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.新 し く It may be newly established according to the method of ma. For example, in the case of mouse ES cells, currently, 129 ES cells are generally used, but since the immunological background is not clear, an alternative pure immunological and genetically For the purpose of obtaining ES cells with a clear background, for example, BDFi mice (C57BL / 6 and C57BL / 6 mice) whose C57BL / 6 mice and C57BL / 6 have reduced the number of eggs collected by crossing with DBA / 2 It is also possible to use a mouse established with DBAZ2, etc. BDFi mice have the advantage of high number of eggs collected and robust eggs, and also have a background of C57BLZ6 mice. ES cells obtained by using C57BLZ6 mice can be used to advantage in that their genetic background can be replaced with C57BLZ6 mice by backcrossing with C57B LZ6 mice when creating disease model mice. .
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。  In addition, when ES cells are established, blastocysts 3.5 days after fertilization are generally used. Early embryos can be obtained.
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。  Either male or female ES cells may be used, but male ES cells are generally more convenient for producing breeding line chimeras. It is also desirable to discriminate between males and females as soon as possible in order to reduce the complexity of culturing.
ES細胞の雌雄の判定方法としては、 例えば、 PCR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コロニ一程度の E S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。 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 (about 50 cells) per colony 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 male and female. Can be reduced to width.
また、 第二次セレクションとしては、 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 ES細胞の染色体数は正常 数の 100%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、 ES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 = 40である細胞) に再びクローニングすることが望ましい。  The secondary selection can be performed, for example, by confirming the number of chromosomes by the G-banding method. The number of chromosomes in the obtained ES cells is preferably 100% of the normal number. However, if it is difficult due to physical operations at the time of establishment, knock out the gene of the ES cells, and It is desirable to clone again into cells with a body number of 2 n = 40).
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、 STO繊維芽細胞のような適当なフィーダ一細胞上で L I F (1〜 ΙΟΟΟΟϋ/ml) 存在下に炭酸ガス培養器内 (好ましくは、 5 %炭酸ガス、 95 %空 気または 5%酸素、 5%炭酸ガス、 90%空気) で約 37 °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/ EDTA溶液 (通常 0.001〜 0.5%トリプシン ZO. 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 1〜3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。  Embryonic stem cell lines obtained in this way usually have very good proliferative properties, but must be carefully subcultured because they tend to lose their ontogenetic potential. For example, on a suitable feeder cell such as STO fibroblasts, in the presence of LIF (1-ΙΟΟΟΟϋ / ml) in a carbon dioxide incubator (preferably 5% carbon dioxide, 95% air or 5% oxygen, Culture at about 37 ° C in 5% carbon dioxide gas and 90% air) .At the time of subculture, for example, trypsin / EDTA solution (usually 0.001 to 0.5% trypsin ZO. L to 5 mM EDTA, preferably A method is used in which cells are converted into single cells by treatment with about 0.1% trypsin / ImMEDTA and seeded on freshly prepared feeder cells. Such subculture is usually carried out every 1 to 3 days. At this time, it is desirable to observe the cells and, if morphologically abnormal cells are found, discard the cultured cells.
ES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 M. H. Kaufman, ネイチヤー (Nature) 第 292巻、 154頁、 1981年; G. R. Martin プロ シーディングス ·ォブ ·ナショナル ·アカデミー ·ォブ ·サイエンス ·ユーェ スェ一 (Proc. Natl. Acad. Sci. U.S.A.) 第 78巻、 7634頁、 1981年; T. C. Doetschman ら、 ジャーナル ·ォブ ·ェンブリオロジ一 ·アンド ·ェクスぺリメ ンタル ·モルフォロジ一、 第 87巻、 27頁、 1985年〕 、 本発明の ES細胞を分化 させて得られる本発明の DN A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。  ES cells are differentiated into various types of cells, such as parietal, visceral, and cardiac muscles, by monolayer culture up to high density or suspension culture until cell clumps are formed under appropriate conditions. [MJ Evans and MH Kaufman, Nature, 292, 154, 1981; GR Martin Proceedings of National Academy of Sciences, Science, Proc. Natl. Acad. Sci. USA) Vol. 78, p. 7634, 1981; TC Doetschman et al., Journal of Obemblilogi and Eximental Morphology, Vol. 87, p. 27, 1985. The DNA-deficient cells of the present invention obtained by differentiating the ES cells of the present invention are useful in the cell biology of the protein of the present invention in the mouth of in vivo.
本発明の DNA発現不全非ヒト哺乳動物は、 該動物の mRNA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 is distinguished from a normal animal by measuring the mRNA level of the animal using a known method and indirectly comparing the expression level. It is possible to
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, those similar to the aforementioned can be 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 した夕一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- 導入により夕一ゲッティングベクターの本発明の D N A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胞の染色 体上の本発明の D N A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をノックアウトさせることができる。  The non-human mammal deficient in expression of the DNA of the present invention can be obtained, for example, by introducing the evening getter vector prepared as described above into mouse embryonic stem cells or mouse egg cells. The DNA of the present invention can be knocked out by homologous recombination in which the inactivated DNA sequence replaces the DNA of the present invention on the chromosome of mouse embryonic stem cells or mouse egg cells by gene homologous recombination. it can.
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配列をプローブとしたサザンハイブリダイゼ一シヨン解析または 夕ーゲッティングベクター上の D NA配列と、 夕ーゲッティングベクタ一に使 用したマウス由来の本発明の D N A以外の近傍領域の D N A配列とをプライマ —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 た細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 卜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 can be analyzed by Southern hybridization analysis using a DNA sequence on or near the DNA of the present invention as a probe, or a DNA sequence on a evening-getting vector, and evening-getting. The determination can be made by PCR analysis using, as a primer, the DNA sequence of a neighboring 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, a cell line in which the DNA of the present invention has been inactivated by gene homologous recombination is cloned, 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 produced chimeric embryo is transplanted 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 N A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ヘテロ発現不全個体同志を交配し、 それらの 産仔から本発明の夕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 obtained from the population obtained by crossing such a chimeric individual with a normal individual. It can be obtained by selecting an individual composed of cells having the DNA locus of the present invention having a mutation, for example, by judging coat color. The individual obtained in this manner 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. It is possible to obtain an individual with poor homo-expression.
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 NA溶液を注入することにより夕一ゲッティングベクターを染色体内に 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 .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 N A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 chromosome into which a gettering vector has been introduced can be obtained by injecting a DNA solution into the egg cell nucleus by a microinjection method. Tiger Compared to nicked non-human mammals, they can be obtained by selecting those having a mutation in the DNA locus of the present invention by gene homologous recombination.
このようにして本発明の D N A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確認して通常 の飼育環境で飼育継代を行なうことができる。  In this way, the individual in which the DNA of the present invention has been knocked out may be subjected to rearing in an ordinary breeding environment after confirming that the DNA has been knocked out even in an animal obtained by mating. it can.
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い。 すなわち、 該不活化 D N A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一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 , ホモザィゴ一ト複数になる 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一ト およびへテロザィゴート動物を繁殖継代する。  Furthermore, the germline can be obtained and maintained according to a standard method. That is, by crossing male and female animals having the inactivated DNA, a homozygous animal having the inactivated DNA on both homologous chromosomes can be obtained. The obtained homozygous animal can be efficiently obtained by rearing the mother animal in such a manner that one normal individual and one homozygote are obtained. By mating male and female heterozygous animals, homozygous and heterozygous animals having the inactivated DNA are bred and subcultured.
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 N A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 expression of the DNA of the present invention.
また、 本発明の D N A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 性の不活性ィヒ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 expressing the DNA of the present invention lacks various biological activities that can be induced by the protein of the present invention, it may be caused by the inactivity of the biological activity of the protein of the present invention. It is useful for investigating the causes of these diseases and examining treatment methods, because they can serve as a model for such diseases.
〔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予防 ·治療 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 preventive / therapeutic effect on a disease caused by DNA deficiency or damage according to 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 N Aの欠損や損傷な どに起因する疾病に対して予防 ·治療効果を有する化合物のスクリーニングに 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 preventive / therapeutic effect against diseases caused by DNA deficiency or damage of the present invention.
すなわち、 本発明は、 本発明の D N A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に対して予防 ·治療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 is characterized in that a test compound is administered to a non-human mammal deficient in DNA expression of the present invention, and changes in the animal are observed and measured. The present invention provides a method for screening a compound or a salt thereof having a preventive / therapeutic effect on a disease to be caused.
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。 試験化合物としては、 例えば、 ペプチド、 タンパク、 非ペプチド性化合物、 合成化合物、 発酵生産物、 細胞抽出液、 植物抽出液、 動物組織抽出液、 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。 Examples of the non-human mammal deficient in DNA expression of the present invention used in the screening method include the same ones as 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.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予防 ·治療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 and compared with an untreated control animal, and changes in organs, tissues, disease symptoms, etc. of the animal are indicated as an index. As a test, the preventive and therapeutic effects of the test compound 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 of treating a test animal with a test compound, for example, oral administration, intravenous injection, or the like is used, and it can be appropriately selected according to the symptoms of the test animal, 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、 慢性閉塞性肺疾患、 気管支喘息に対して予防 ·治療効果を有する化 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夕 バコ煙または抗原曝露処置を行ない、 タバコ煙または抗原曝露処置前または処 置後に試験化合物を投与し、 該動物の気道反応性変化などを経時的に測定する。 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予防 ·治療効果を有するので、 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 さる。  For example, when screening for a compound having a preventive / therapeutic effect on chronic obstructive pulmonary disease or bronchial asthma, a non-human mammal deficient in expression of the DNA of the present invention is treated with coconut smoke or antigen exposure treatment, and tobacco The test compound is administered before or after the smoke or antigen exposure treatment, and changes in the airway responsiveness of the animal are measured over time. The compound obtained by using the screening method is a compound selected from the test compounds described above, and has a preventive / therapeutic effect against a disease caused by deficiency or damage of the protein of the present invention. It can be used as a medicament such as a safe and low toxic prophylactic / therapeutic agent for the disease. Further, a compound derived from the compound obtained by the above-mentioned 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、 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。 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。 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 たは非ヒト哺乳動物 (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。 該ィ匕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 60kgとして) の慢性閉塞性肺疾患の患者においては、 一日につき該化合物を 約 0. l〜100mg、 好ましくは約 1. 0〜50mg、 より好ましくは約 1. 0〜20mg投与する。 非経口的に投与する場合は、 該化合物の 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、 該化合物を注射剤の形で通常成人 (60kgとし て) の慢性閉塞性肺疾患の患者に投与する場合、 一日につき該化合物を約 0. 01 〜30mg、 好ましくは約 0. l〜20mg、 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、 60 k g当たりに換算した量 を投与することができる。 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 metals). And the like, and particularly 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) Succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc.). 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, for example, in humans or non-human mammals (eg, rats, mice, guinea pigs, egrets, higgies, bushus, dogs, 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. For example, when the compound is orally administered, it is generally used for adults (with a body weight of 60 kg). In patients with chronic obstructive pulmonary disease, about 0.1 to 100 mg, preferably about 1.0 to 50 mg, more preferably about 1.0 to 20 mg of the compound is administered per day. In the case of parenteral administration, the single dose of the compound may vary depending on the administration subject, target disease, etc. For example, the compound is usually injectable in the form of an injection (typically 60 kg) in chronic obstructive pulmonary lungs of adults. When administered to a patient with a disease, it is convenient to administer about 0.01 to 30 mg, preferably about 0.1 to 20 mg, more preferably about 0.1 to 10 mg of the compound by intravenous injection per day. It is. In the case of other animals, the dose can be administered in terms of 60 kg.
[ 8 b ) 本発明の D NAに対するプロモーターの活性を促進または阻害する化 合物のスクリーニング方法  [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 D NA発現不全非ヒト哺乳動物に、 試験化合物を投与し、 レポ一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 D NAに対する プロモータ一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 relates to a method for administering a test compound to a non-human mammal deficient in expression of a DNA of the present invention, and detecting the expression of a reporter gene. Provided is a method for screening a compound or a salt thereof that inhibits the inhibition.
上記スクリーニング方法において、 本発明の D NA発現不全非ヒト哺乳動物 としては、 前記した本発明の D NA発現不全非ヒト哺乳動物の中でも、 本発明 の D N Aがレポ一夕一遺伝子を導入することにより不活性化され、 該レポ一夕 —遺伝子が本発明の D N Aに対するプロモ一ターの制御下で発現しうるものが 用いられる。  In the above-described screening method, the non-human mammal deficient in DNA expression of the present invention may be a non-human mammal deficient in expression of DNA of the present invention, wherein the DNA of the present invention is obtained by introducing a repo allele gene. Those inactivated and capable of expressing the repo overnight gene under the control of a promoter for the DNA of the present invention are used.
試験化合物としては、 前記と同様のものがあげられる。  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 シフエラ一ゼ遺伝子などが好適である。 As the reporter gene, the same one as described above can be used.) 3-galactosidase gene (1 ac Z), soluble alkaline phosphatase gene or The sifellanase gene and the like are preferred.
本発明の D N Aをレポ一夕一遺伝子で置換された本発明の D N A発現不全非 ヒト哺乳動物では、 レポ一夕一遺伝子が本発明の D NAに対するプロモ一夕一 の支配下に存在するので、 レポ一夕一遺伝子がコ一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 DNA expression of the present invention in which the DNA of the present invention is replaced with a repo overnight gene, since the repo overnight gene is under the control of the promoter for the DNA of the present invention, By tracing the expression of a substance encoded by the repo overnight gene, 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 NA領域の一部を大腸菌由来の β一ガラクトシダーゼ遺伝子 ( 1 a c Ζ ) 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 ]3—ガラクト シダ一ゼが発現する。 従って、 例えば、 5—プロモー 4一クロロー 3—インド リル一] 8 _ガラクトピラノシド (X - g a 1 ) のような) 3—ガラクトシダ一ゼ 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 (P B S ) で洗浄後、 X— g a 1を含む染色液で、 室温 または 3 7 °C付近で、 約 3 0分ないし 1時間反応させた後、 組織標本を 1 mM E D TA/ P B S溶液で洗浄することによって、 /3—ガラクトシダーゼ反応を 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 N A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 with a β-galactosidase gene (1 ac 由来) derived from Escherichia coli, the tissue of the present invention expressing the protein of the present invention originally ] -Galactosidase is expressed instead of protein. Thus, for example, by staining with a reagent that is a substrate for 3-galactosidase (such as 5-promo-4-chloro-3-indolyl-1) 8-galactopyranoside (X-ga 1), The state of expression of the protein of the present invention in an animal organism can be easily observed. Specifically, the protein-deficient mouse of the present invention or a tissue section thereof is fixed with dartalaldehyde and the like, washed with phosphate buffered saline (PBS), and then stained with X-ga1 at room temperature or at room temperature. After reacting for about 30 minutes to 1 hour at around 7 ° C, the / 3-galactosidase reaction can be stopped by washing the tissue sample with 1 mM EDTA / PBS solution, and the coloration can be observed. . Further, mRNA encoding 1acZ 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 of the DNA of the present invention.
該スクリ一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、 無機酸など) や塩基 (例、 有 機酸など) 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。 また、 本発明の D N Aに対するプロモーター活性を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を阻害し、 該タンパク質の機能を阻害する ことができるので、 例えば肺 ·気道の炎症を伴う肺 ·胸部疾患などの呼吸器疾 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、 過敏性肺炎など〕 、 アレルギー性結膜炎、 鼻炎 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, organic acids). And the like, and particularly 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) Succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc.). Further, the compound of the present invention which inhibits the promoter activity for DNA or the salt thereof can inhibit the expression of the protein of the present invention and inhibit the function of the protein. · Respiratory diseases such as chest diseases (eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), allergic conjunctivitis, Rhinitis
(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) 、 消化管 疾患 (例、 過敏性腸症候群、 炎症性腸疾患、 潰瘍性大腸炎、 クローン病、 逆流 性食道炎など) などの予防,治療剤、 好ましくは呼吸器疾患、 消化管疾患など の予防 ·治療剤などの医薬として有用である。  (Eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry pronasitis, vasomotor rhinitis, gangrene rhinitis, sinusitis, etc.), gastrointestinal diseases (eg, As preventive and therapeutic agents for irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease, reflux esophagitis, etc., preferably as preventive and therapeutic agents for respiratory diseases, gastrointestinal diseases, etc. Useful.
さらに、 上記スクリ一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, a compound derived from the compound obtained by the above screening can also be 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 たは非ヒト哺乳動物 (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。 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 D N Aに対するプロモーター活性を促進 する化合物を経口投与する場合、 一般的に成人 (体重 60kgとして) の慢性閉塞 性肺疾患の患者においては、 一日につき該ィヒ合物を約 0. l〜100mg、 好ましくは 約 1. 0〜50mg、 より好ましくは約 1. 0〜20mg投与する。 非経口的に投与する場合 は、 該化合物の 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 D NAに対するプロモーター活性を促進する化合物を注射剤の 形で通常成人 (60kgとして) の慢性閉塞性肺疾患の患者に投与する場合、 一日 にっき該化合物を約 0. 01〜30mg、 好ましくは約 0. l〜20mg、 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、 60kg当たりに換算した量を投与することができる。 一方、 例えば、 本発明の D NA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 60kgとして) の慢性閉塞性肺疾患の 患者においては、 一日につき該化合物を約 0. l〜100mg、 好ましくは約 1. 0〜50mg、 より好ましくは約 1. 0〜20mg投与する。 非経口的に投与する場合は、 該化合物の 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 D N Aに対するプロモー夕一活性を阻害する化合物を注射剤の形で通常成人The preparations obtained in this way are safe and low toxic and can be used, for example, in humans or non-human mammals (for example, rats, mice, guinea pigs, egrets, sheep, pigs, horses, cats, cats, Dogs, monkeys, etc.). The dose of the compound or a salt thereof varies depending on the target disease, the administration subject, the administration route, and the like. For example, when the compound of the present invention that promotes the promoter activity for DNA is orally administered, generally, the adult ( In patients with chronic obstructive pulmonary disease (with a body weight of 60 kg), about 0.1 to 100 mg, preferably about 1.0 to 50 mg, more preferably about 1.0 to 20 mg of the ehine compound is administered per day. I do. When administered parenterally, the single dose of the compound varies depending on the administration subject, target disease, and the like.For example, the compound that promotes the promoter activity for DNA of the present invention is usually in the form of an injection. When administered to an adult (as 60 kg) patient with chronic obstructive pulmonary disease, about 0.01 to 30 mg, preferably about 0.1 to 20 mg, more preferably about 0.1 to 10 mg of the compound per day is administered. It is conveniently administered by intravenous injection. In the case of other animals, the dose can be administered in terms of 60 kg. On the other hand, for example, when the compound of the present invention that inhibits the promoter activity against DNA is orally administered, generally, in an adult (assuming a body weight of 60 kg) patient with chronic obstructive pulmonary disease, the compound is reduced to about 0 per day. l-100 mg, preferably about 1.0-50 mg, more preferably about 1.0-20 mg. 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 promoter activity against DNA of the present invention is usually administered in the form of an injection. adult
(60kgとして) の慢性閉塞性肺疾患の患者に投与する場合、 一日につき該ィ匕合 物を約 0. 01〜30mg、 好ましくは約 0. l〜20mg、 より好ましくは約 0. l〜10mg度を 静脈注射により投与するのが好都合である。 他の動物の場合も、 60kg当たりに 換算した量を投与することができる。 (As 60 kg) to a patient with chronic obstructive pulmonary disease of about 0.01 to 30 mg, preferably about 0.1 to 20 mg, and more preferably about 0.1 to 1 mg of the compound per day. It is convenient to administer 10 mg by intravenous injection. In the case of other animals, the dose can be administered in terms of 60 kg.
このように、 本発明の D NA発現不全非ヒト哺乳動物は、 本発明の D NA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 一二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 ·治療薬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 that 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 cause of various diseases caused by DNA expression deficiency or the development of preventive and therapeutic agents of the present invention.
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 D NA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エニック動物 (遺伝子移入動物) を作成すれば、 特異的にそのポリぺプチドを合成させ、 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一部分に適当なレポ一夕一遺伝子を結合させ、 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。 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、 I U P A C 一 I U B Commi ss ion on Biochemical Nomenc lature 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 addition, 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 polypeptide and examine its action in living organisms. Further, by binding an appropriate repo overnight gene to a part of the promoter and establishing a cell line in which the gene is expressed, the action of specifically promoting or suppressing the in vivo production ability of the protein of the present invention itself is achieved. It can be used as a search system for low molecular weight compounds. In the present specification, bases, amino acids, and the like are indicated by abbreviations based on the abbreviations by IUPAC-IUB Communications 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 :デォキシリポ核酸 c DNA 相補デォキシリボ核酸 DNA: Deoxylipo nucleic acid c DNA complementary deoxyribonucleic acid
A アデニン  A adenine
T チミン  T thymine
G グァニン  G Guanin
C  C
R アデニン (A) またはグァニン (G) Y シトシン (C) またはチミン (T) R adenine (A) or guanine (G) Y cytosine (C) or thymine (T)
K , グァニン (G) またはチミン (T)K, guanine (G) or thymine (T)
M アデニン (A) またはシ卜シン (C)M adenine (A) or cytosine (C)
S グァニン (G) またはシトシン (C) w アデニン (A) またはチミン (T)S guanine (G) or cytosine (C) w adenine (A) or thymine (T)
RNA リポ核酸 RNA liponucleic acid
mRNA メッセンジャーリボ核酸 mRNA messenger ribonucleic acid
d ATP 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 d ATP Deoxyadenosine triphosphate dTTP Deoxythymidine triphosphate
dGTP デォキシグアノシン三リン酸 dCTP デォキシシチジン三リン酸 . ATP アデノシン三リン酸 dGTP Deoxyguanosine triphosphate dCTP Deoxycytidine triphosphate. ATP Adenosine triphosphate
EDTA エチレンジァミン四酢酸 EDTA ethylenediaminetetraacetic acid
SDS ドデシル硫酸: SDS dodecyl sulfate:
G 1 y グリシン G 1 y Glycine
A 1 a ァラニン A 1 a Alanin
Va 1 バリン Va 1 Valine
Le u ロイシン Le u leucine
I 1 e イソロイシン  I 1 e isoleucine
S e r セリン S e r serine
Th r スレオニン Th r threonine
Cy s Cy s
Me t メチォニン G 1 u グル夕ミン酸 Me t Methionin G 1 u glumic acid
As ァスパラギン酸  As aspartic acid
L y s リジン  Lys lysine
A r g アルギニン  A r g Arginine
H i s ヒスチジン  H is histidine
P h e フエ二ルァラニン  P h e feniralanin
Ty r チロシン  Ty r tyrosine
T r p  T r p
P r o プロリン  Pro proline
AA ss nn :ァスパラギン  AA ss nn: Asparagine
G 1 n グルタミン  G 1 n Glutamine
p G 1 u ピログルタミン酸  p G 1 u pyroglutamic acid
また、 本明細書中で繁用される置換基、 保護基および試薬を下記の記号で表 記する。  Further, substituents, protecting groups and reagents frequently used in the present specification are represented by the following symbols.
Me : メチル基  Me: methyl group
E t :ェチル基  E t: ethyl group
B u :ブチル基  B u: butyl group
Ph : フエニル基  Ph: phenyl group
TC :チアゾリジン— 4 (R) 一力ルポキサミド基  TC: Thiazolidine-4 (R) one-pot lipoxamide group
To s : p一トルエンスルフォニル  To s: p-toluenesulfonyl
CHO :ホルミル  CHO: Formyl
B z 1 :ベンジル  B z 1: benzyl
Cl2-Bzl : 2, 6ージクロ口べンジル Cl 2 -Bzl: 2,6
Bom :ベンジルォキシメチル  Bom: benzyloxymethyl
Z :ベンジルォキシカルポニル  Z: benzyloxycarponyl
C 1一 Z : 2  C 1 Z: 2
B r - Z : 2
Figure imgf000070_0001
Br-Z: 2
Figure imgf000070_0001
Bo c : t一ブトキシカルボニル  Bo c: t-butoxycarbonyl
DNP : ジニトロフエニル T r t 卜リチル DNP: dinitrophenyl T rt Trityl
Bum t一ブトキシメチル  Bum t-butoxymethyl
Fmo c N— 9一フルォレニルメトキシカルボニル  Fmo c N—9-Fluorenylmethoxycarbonyl
HOB t  HOB t
HOOB t 3, 4—ジヒドロ一 3—ヒドロキシ一 4ーォキソ一  HOOB t 3, 4-dihydro-1-hydroxy-1-4-oxo-1
1, 2, 3—ベンゾ卜リアジン  1,2,3-benzotriazine
HONB 1 -ヒドロキシ -5-ノルポルネン- 2,3-ジカルポキシィ DCC 本願明細書の配列表の配列番号は、 以下の配列を示す。  HONB 1-hydroxy-5-norporene-2,3-dicaroxy DCC The sequence numbers in the sequence listing in the present specification show the following sequences.
〔配列番号: 1〕  [SEQ ID NO: 1]
マウス CLCA4タンパク質のアミノ酸配列を示す。  2 shows the amino acid sequence of mouse CLCA4 protein.
〔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マウス CLCA4タンパク質 のァミノ酸配列をコードする DNAの塩基配列を示す。  This shows the base sequence of DNA encoding the amino acid sequence of mouse CLCA4 protein having the amino acid sequence represented by SEQ ID NO: 1.
〔配列番号: 3〕  [SEQ ID NO: 3]
マウス CLCA4遺伝子 5,側の塩基配列を示す。  The nucleotide sequence of mouse CLCA4 gene 5 is shown.
〔配列番号: 4〕  [SEQ ID NO: 4]
マウス CLCA4遺伝子 3 '側の塩基配列を示す。  The nucleotide sequence of the mouse CLCA4 gene on the 3 'side is shown.
〔配列番号: 5〕  [SEQ ID NO: 5]
実施例 1および実施例 3で用いられたプライマ一 1の塩基配列を示す。 〔配列番号: 6〕  3 shows the nucleotide sequence of primer 11 used in Examples 1 and 3. [SEQ ID NO: 6]
実施例 1および実施例 4で用いられたプライマー 2の塩基配列を示す。 〔配列番号: 7〕  2 shows the nucleotide sequence of primer 2 used in Examples 1 and 4. [SEQ ID NO: 7]
実施例 1および実施例 4で用いられたプライマー 3の塩基配列を示す。 3 shows the nucleotide sequence of primer 3 used in Examples 1 and 4.
〔配列番号: 8〕 [SEQ ID NO: 8]
実施例 1および実施例 2で用いられたプライマー 4の塩基配列を示す。 〔配列番号: 9〕  2 shows the nucleotide sequence of primer 4 used in Examples 1 and 2. [SEQ ID NO: 9]
実施例 1で用いられたプライマー 5の塩基配列を示す。 〔配列番号: 10〕 2 shows the base sequence of primer 5 used in Example 1. [SEQ ID NO: 10]
実施例 1および実施例 2で用いられたプライマー 6の塩基配列を示す。  3 shows the nucleotide sequence of primer 6 used in Examples 1 and 2.
〔配列番号: 11〕  [SEQ ID NO: 11]
実施例 1で用いられたプライマー 7の塩基配列を示す。  2 shows the base sequence of primer 7 used in Example 1.
〔配列番号: 12〕  [SEQ ID NO: 12]
実施例 1で用いられたプライマー 8の塩基配列を示す。  2 shows the nucleotide sequence of primer 8 used in Example 1.
〔配列番号: 13〕  [SEQ ID NO: 13]
実施例 1で用いられたプライマー 9の塩基配列を示す。  2 shows the nucleotide sequence of primer 9 used in Example 1.
〔配列番号: 14〕  [SEQ ID NO: 14]
実施例 1で用いられたプライマー 10の塩基配列を示す。  2 shows the nucleotide sequence of primer 10 used in Example 1.
〔配列番号: 15〕  [SEQ ID NO: 15]
実施例 1で用いられた T 7 p r omo t e r p r ime r (プロモーター プライマ一) の塩基配列を示す。  1 shows the nucleotide sequence of T7promoteperprimer (promoter primer 1) used in Example 1.
〔配列番号: 16〕  [SEQ ID NO: 16]
実施例 1で用いられた Ml 3 RV p r ime r (プライ 一) の塩基配列を 示す。  1 shows the nucleotide sequence of Ml 3 RV primer (primary) used in Example 1.
〔配列番号: 17〕  [SEQ ID NO: 17]
マウス CLCA4 Aタンパク質のアミノ酸配列を示す。  2 shows the amino acid sequence of mouse CLCA4 A protein.
〔配列番号: 18〕  [SEQ ID NO: 18]
配列番号: 17で表されるアミノ酸配列を有するマウス CLCA4 Aタンパ ク質のアミノ酸配列をコードする DN Aの塩基配列を示す。  The nucleotide sequence of DNA encoding the amino acid sequence of mouse CLCA4A protein having the amino acid sequence represented by SEQ ID NO: 17 is shown.
〔配列番号: 19〕  [SEQ ID NO: 19]
ラット CLCA4夕ンパク質の部分アミノ酸配列を示す。  2 shows a partial amino acid sequence of rat CLCA4 protein.
〔配列番号: 20〕  [SEQ ID NO: 20]
配列番号: 19で表される部分アミノ酸配列を有するラット CLCA4タン パク質の部分アミノ酸配列をコードする DNAの塩基配列を示す。  This shows the base sequence of DNA encoding the partial amino acid sequence of rat CLCA4 protein having the partial amino acid sequence represented by SEQ ID NO: 19.
〔配列番号: 21〕  [SEQ ID NO: 21]
実施例 3で用いられたプライマ一 11の塩基配列を示す。  3 shows the nucleotide sequence of Primer 11 used in Example 3.
〔配列番号: 22〕 実施例 5で用いられたプライマー 12の塩基配列を示す。 [SEQ ID NO: 22] 7 shows the nucleotide sequence of primer 12 used in Example 5.
〔配列番号: 23〕  [SEQ ID NO: 23]
実施例 5で用いられたプライマー 13の塩基配列を示す。  13 shows the base sequence of primer 13 used in Example 5.
〔配列番号: 24〕  [SEQ ID NO: 24]
実施例 5で用いられたプライマー 14の塩基配列を示す。 後述の実施例 1で得られた形質転換体ェシェリヒア 'コリ (Escherichia coli) TOPlO/pCR-Bluntll - mCLCA4は、 2002年 2月 12日から茨城県つくば巿東 1丁 目 1番地 1 中央第 6 (郵便番号 305-8566) の独立行政法人産業技術総合研究所 特許生物寄託セン夕一に寄託番号 FERM BP- 7887として、 2002年 1月 29日から大阪 府大阪市淀川区十三本町 2丁目 17番 85号 (郵便番号 532-8686) の財団法人発酵研 究所 (IF0) に受託番号 IF0 16751として寄託されている。  14 shows the base sequence of primer 14 used in Example 5. Transformant Escherichia coli Escherichia coli (Escherichia coli) TOPlO / pCR-Bluntll-mCLCA4 obtained in Example 1 described below has been used since February 12, 2002, 1-1 1-1 Tsukuba-Higashi, Ibaraki Pref. Deposit No.FERM BP-7887 at the National Institute of Advanced Industrial Science and Technology (AIST) with a postal code of 305-8566) as a deposit number FERM BP-7887 from January 29, 2002 at 2-317 Jusanhoncho, Yodogawa-ku, Osaka, Osaka It has been deposited with the Fermentation Research Institute (IF0) of No. 85 (zip code 532-8686) under the accession number IF0 16751.
後述の実施例 3で得られた形質転換体ェシエリヒア ·コリ (Escherichia coli) TOPlO/pCR- Bluntll- mCLCA4Aは、 2002年 3月 4日から茨城県つくば巿東 1丁 目 1番地 1 中央第 6 (郵便番号 305- 8566) の独立行政法人産業技術総合研究所 . 特許生物寄託センターに寄託番号 FERM BP- 7934として、 2002年 2月 19日から大阪 府大阪市淀川区十三本町 2丁目 17番 85号 (郵便番号 532- 8686) の財団法人発酵研 究所 (IF0) に受託番号 IF0 16764として寄託されている。 以下に、 実施例を挙げて本発明をさらに具体的に説明するが、 本発明はそれ に限定されるものではない。 実施例 1  The transformant Escherichia coli TOPlO / pCR-Bluntll-mCLCA4A obtained in Example 3 described below has been used since March 4, 2002, 1-1 1-1 Tsukuba, Higashi, Ibaraki Pref. National Institute of Advanced Industrial Science and Technology (ZIP code 305-8566). Deposited at the Patent Organisms Depositary Center as deposit number FERM BP-7934. From February 19, 2002, 2-3-17 Jusanhoncho, Yodogawa-ku, Osaka, Osaka, Japan 85 No. (postal code 532-8686) and deposited with the Fermentation Research Institute (IF0) under the accession number IF0 16764. Hereinafter, the present invention will be described more specifically with reference to Examples, but the present invention is not limited thereto. Example 1
マウス CLCA4遺伝子のクローニング  Cloning of mouse CLCA4 gene
ヒト CLCA4遺伝子の塩基配列 〔FEBS Letters, 455巻、 295頁 (1999) 〕 、 マウ ス gob- 5遺伝子の塩基配列 〔Biochem. Biophys. Res. Commun.、 255卷、 347頁 (1999) 〕 を参考にして 4種のプライマ一 〔プライマー 1 (配列番号: 5) 、 プライマー 2 (配列番号: 6) 、 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) 〕 を設計 ·合成した。 次に、 マウス大腸組織から ISOGEN (和光純薬社製) を用いて全 RNAを調製した。 さらに mRNA purification kit (アマシャムフアルマシアバイオテク社製) を用いて mRNAを調製した。 この mRNA 1 igを出発材料として Marathon cDNA Amplification kit (Clontech社 製) を用いて逆転写反応により cDNAを合成した。 この cDNAを铸型としてプライ マー 1とプライマ一 2、 プライマー 3とプライマー 4の組み合わせで PCRを行つ た後、 ァガ口一ス電気泳動により予想される大きさの DNA断片が増幅されること を認めた。 反応は pyrobest DNA polymerase (宝酒造社製) を用いてサ一マルサ イクラ一 Gene Amp PCR System 9700 (パーキンエルマ一社製) にて、 最初 94°C で 30秒間反応させた後、 94 で 10秒、 60°Cで 30秒、 72°Cで 2分 30秒を 1反応サイ クルとして 35サイクル繰り返し、 最後は 72°Cで 10分間反応させた。 得られた DNA fr¾pCR_BluntII-T0P0 (Invitrogen社製) にクローニングした。 さらに T7 promoter primer (配列番号: 15) 、 M13RV primer (配列番号: 16) および 6種の合成プライマ一 〔プライマー 5 (配列番号: 9) 、 プライマ一 6 (配列番 号: 10) 、 プライマー 7 (配列番号: 1 1) 、 プライマ一 8 (配列番号: 1 2) 、 プライマー 9 (配列番号: 1 3) およびプライマー 10 (配列番号: 1 4) 〕 を用いてサイクルシークェンス反応を行い、 蛍光 DNAシークェンサ一377 (パーキンエルマ一社製) で得られた反応物の塩基配列を決定した。 その結果、 マウス CLCA4遺伝子の部分塩基配列としてマウス N末端配列を含有する 2027個の 塩基配列を揷入したクローン No.3-1 (配列番号: 3) とマウス C末端配列を含有 する 1869個の塩基配列を挿入したクロ一ン No.7-3 (配列番号: 4) を取得した。 両者の重複配列を除き、 マウス CLCA4は 2772個の塩基配列からなることが判明し た (配列番号: 2) 。 配列番号: 2で表される塩基配列を基に、 924個のアミノ 酸配列を決定し (配列番号: 1) 、 マウス CLCA4タンパク質と命名した。 つづい てクローン No.7-3を Hi nd I IIで消化し、 1.4Kbのマウス CLCA4の C末端領域を含む DNA断片を切り出した。 これを同じく Hindi IIで消化したクローン No.3-1のマウ ス CLCA4の N末端領域を含むベクター配列に連結し、 マウス CLCA4完全長配列を含 むプラスミド pCR- Bluntll- mCLCA4を構築した。 このプラスミドを含む形質転換 体をェシエリヒア 'コリ (Escherichia coli) TOP10/pCR-BluntII- mCLCA4と命 名した。 マウス CLCA4は、 アミノ酸レベルで、 ヒト CLCA4と 68%の相同性を、 マウス gob- 5と 51%の相同性を有していた。 実施例 2 See the nucleotide sequence of the human CLCA4 gene (FEBS Letters, vol. 455, p. 295 (1999)) and the nucleotide sequence of the mouse gob-5 gene [Biochem. Biophys. Res. Commun., Vol. 255, p. 347 (1999)] And designed and synthesized four primers [Primer 1 (SEQ ID NO: 5), Primer 2 (SEQ ID NO: 6), Primer 3 (SEQ ID NO: 7) and Primer 4 (SEQ ID NO: 8)] . Next, from mouse colon tissue Total RNA was prepared using ISOGEN (manufactured by Wako Pure Chemical Industries, Ltd.). Furthermore, mRNA was prepared using mRNA purification kit (manufactured by Amersham Pharmacia Biotech). Using this mRNA 1 ig as a starting material, cDNA was synthesized by reverse transcription using a Marathon cDNA Amplification kit (manufactured by Clontech). After performing PCR using this cDNA as type I and a combination of Primer 1 and Primer 1 and Primer 3 and Primer 4, a DNA fragment of the expected size is amplified by agarose gel electrophoresis. Admitted. The reaction was carried out using Pyrobest DNA polymerase (Takara Shuzo Co., Ltd.) on a Thermocycler Gene Amp PCR System 9700 (PerkinElmer) at 94 ° C for 30 seconds, then at 94 for 10 seconds. The reaction cycle was repeated 35 times at 60 ° C for 30 seconds, 72 ° C for 2 minutes and 30 seconds, and finally the reaction was performed at 72 ° C for 10 minutes. It was cloned into the obtained DNA fr¾pCR_BluntII-T0P0 (manufactured by Invitrogen). Furthermore, T7 promoter primer (SEQ ID NO: 15), M13RV primer (SEQ ID NO: 16) and six synthetic primers [Primer 5 (SEQ ID NO: 9), Primer 6 (SEQ ID NO: 10), Primer 7 ( Cycle sequence reaction was performed using SEQ ID NO: 11), Primer 8 (SEQ ID NO: 12), Primer 9 (SEQ ID NO: 13), and Primer 10 (SEQ ID NO: 14)] to obtain a fluorescent DNA sequencer. The base sequence of the reaction product obtained by I-377 (PerkinElmer) was determined. As a result, clone No. 3-1 (SEQ ID NO: 3) containing a 2027 nucleotide sequence containing the mouse N-terminal sequence as a partial nucleotide sequence of the mouse CLCA4 gene and 1869 clones containing the mouse C-terminal sequence The clone No. 7-3 (SEQ ID NO: 4) into which the nucleotide sequence was inserted was obtained. Excluding the overlapping sequence of both, mouse CLCA4 was found to be composed of 2772 nucleotide sequences (SEQ ID NO: 2). Based on the nucleotide sequence represented by SEQ ID NO: 2, 924 amino acid sequences were determined (SEQ ID NO: 1), and named as mouse CLCA4 protein. Subsequently, clone No. 7-3 was digested with Hind I II to cut out a DNA fragment containing the C-terminal region of mouse CLCA4 of 1.4 Kb. This was ligated to a vector sequence containing the N-terminal region of mouse CLCA4 of clone No. 3-1 also digested with Hindi II to construct a plasmid pCR-Bluntll-mCLCA4 containing a mouse CLCA4 full-length sequence. A transformant containing this plasmid was designated as Escherichia coli TOP10 / pCR-BluntII-mCLCA4. At the amino acid level, mouse CLCA4 had 68% homology with human CLCA4 and 51% homology with mouse gob-5. Example 2
マウス C L C A 4遺伝子産物の組織分布の解析  Analysis of tissue distribution of mouse CLC A4 gene product
プライマ一 4 (配列番号: 8) およびプライマ一 6 (配列番号: 1 0) を用 いて、 マウスの各組織 (骨髄、 眼、 リンパ節、 平滑筋、 卵巣、 胸腺、 胃、 膀胱、 心齓 全脳、 脾臓、 肺、 肝臓、 骨格筋、 腎臓、 精巣、 7 · 11 · 15 · 17日目胚、 大 腸) の cDNA (Mouse MTC panel Iおよび Mouse TC panel II:クロンテック社 製) を铸型として PCRを行った。 反応は Takara Ex Taq (宝酒造社製) を用いて サ一マルサイクラ一 GeneAmp PCR System 9600 (アプライドバイオシステムズ社 製) にて、 最初 94°C30秒間おいた後で、 94°Cで 10秒、 60°Cで 30秒、 72 で 2分を 1反応サイクルとして 35サイクル繰り返し、 最後は 72°Cで 10分間反応させた。 得 られた反応産物を 2%ァガロースゲルにて電気泳動にかけた後、 臭化工チジゥム で染色して増幅された 800bpのバンドを検出した。 その結果、 マウス CLCA4遺伝 子産物 (mRNA) は平滑筋、 大腸で強い発現が見られ、 骨格筋でも発現が見られ た。 実施例 3  Using Primer 4 (SEQ ID NO: 8) and Primer 1 (SEQ ID NO: 10), mouse tissues (bone marrow, eyes, lymph nodes, smooth muscle, ovary, thymus, stomach, bladder, heart) Brain, spleen, lung, liver, skeletal muscle, kidney, testis, embryos on day 7, 11, 15, 17 and colon (Mouse MTC panel I and Mouse TC panel II: Clontech) PCR was performed. The reaction was performed using Takara Ex Taq (manufactured by Takara Shuzo Co., Ltd.) on a Thermocycler GeneAmp PCR System 9600 (manufactured by Applied Biosystems) first at 94 ° C for 30 seconds, then at 94 ° C for 10 seconds and 60 ° C. The reaction was repeated for 35 cycles, with one reaction cycle consisting of 30 seconds at 72 ° C and 2 minutes at 72 ° C. The obtained reaction product was subjected to electrophoresis on a 2% agarose gel, and then stained with B. bromide to detect an amplified 800 bp band. As a result, the mouse CLCA4 gene product (mRNA) was strongly expressed in smooth muscle and large intestine, and was also expressed in skeletal muscle. Example 3
マウス CL CA4 A遺伝子のクロ一ニング  Cloning of mouse CL CA4 A gene
マウス CLCA4遺伝子のクローニングの際、 マウス CLCA4とは異なる塩基配列を 有するクローンの存在が示唆された。 そこで、 マウス平滑筋 cDNA (Mouse MTC panel II :クロンテック社製) を錶型として、 プライマ一 1 (配列番号: 5) とプライマー 1 1 (配列番号: 2 1) の組み合わせで PCRを行い、 ァガロース電 気泳動により 2.7Kbの DNA断片が増幅されることを認めた。 反応は pyrobest DNA polymerase (宝酒造社製) を用いてサ一マルサイクラ一 Gene Amp PCR System 9700 ひ°—キンエルマ一社製) にて、 最初 94°Cで 30秒間反応させた後、 94°Cで 10秒、 60 で 30秒、 72 で 5分を 1反応サイクルとして 35サイクル繰り返し、 最 後は 72°Cで 10分間反応させた。 得られた DNA断片は pCR- B 1 until -T0P0 (Invitrogen社製) にクローニングした。 さらに T7 promoter primer (配列番 号: 1 5) 、 13RV primer (配列番号: 16) および 6種の合成プライマー 〔プ ライマ一 5 (配列番号: 9) 、 プライマー 6 (配列番号: 10) 、 プライマー 7 (配列番号: 1 1) 、 プライマ一 8 (配列番号: 12) 、 プライマ一 9 (配 列番号: 13) およびプライマ一 1 0 (配列番号: 14) 〕 を用いてサイクル シークェンス反応を行い、 蛍光 DNAシークェンサ一377 (パ一キンエルマ一社 製) で得られた反応物の塩基配列を決定した。 その結果、 マウス CLCA4とァミノ' 酸レベルで 94%、 ヒト CLCA4と 68%の相同性を示す 924個のアミノ酸からなるタ ンパク質 (配列番号: 17) がコードされていることが判明し (配列番号: 1 8) 、 マウス CLCA4Aタンパク質と命名した。 上記プラスミド pCR- Bluntll - mCLCA4を構築した。 このプラスミド pCR_BluntII- T0P0を含む形質転換体をェシ エリヒア 'コリ (Escherichia coli) TOP10/pCR- Bluntll- mCLCA4Aと命名した。 実施例 4 When cloning the mouse CLCA4 gene, it was suggested that a clone having a nucleotide sequence different from that of mouse CLCA4 was present. Therefore, using a mouse smooth muscle cDNA (Mouse MTC panel II: manufactured by Clontech) as type I, PCR was performed using a combination of primer 1 (SEQ ID NO: 5) and primer 11 (SEQ ID NO: 21), and agarose electrophoresis was performed. It was confirmed that a 2.7 Kb DNA fragment was amplified by electrophoresis. The reaction was carried out using Pyrobest DNA polymerase (Takara Shuzo Co., Ltd.) on a Thermocycler Gene Amp PCR System 9700 (Kin-Elma Co., Ltd.) for 30 seconds at 94 ° C. The reaction was repeated for 35 cycles, each cycle consisting of 30 seconds at 60 seconds and 5 minutes at 72, and the reaction was continued at 72 ° C for 10 minutes. The obtained DNA fragment was pCR-B1 until -T0P0 (Manufactured by Invitrogen). Furthermore, T7 promoter primer (SEQ ID NO: 15), 13RV primer (SEQ ID NO: 16) and six synthetic primers [Primer 1 (SEQ ID NO: 9), Primer 6 (SEQ ID NO: 10), Primer 7 (SEQ ID NO: 11), PRIMER-1 (SEQ ID NO: 12), PRIMER-1 (SEQ ID NO: 13) and PRIMER-10 (SEQ ID NO: 14) The nucleotide sequence of the reaction product obtained using DNA Sequencer-377 (manufactured by Pakinkin Elma) was determined. As a result, it was found that a protein consisting of 924 amino acids (SEQ ID NO: 17) showing homology of 94% at the amino acid level with mouse CLCA4 and 68% with human CLCA4 was encoded (SEQ ID NO: 17). No .: 18), designated as mouse CLCA4A protein. The above plasmid pCR-Bluntll-mCLCA4 was constructed. A transformant containing this plasmid pCR_BluntII-TOP0 was designated as Escherichia coli TOP10 / pCR-Bluntll-mCLCA4A. Example 4
マウス CLCA4 A遺伝子産物の組織分布の解析 ,  Analysis of tissue distribution of mouse CLCA4 A gene product,
マウス CLCA4とマウス CLCA4Aの共通配列であるプライマー 2 (配列番号: 6) とプライマー 3 (配列番号: 7) の組み合わせで PCRにより増幅した 1020bpの DNA断片が、 マウス CLCA4由来は制限酵素 PvuII (宝酒造社製) で消化されないの に対し、 マウス CLCA4A由来は 680bpと 340bpに消化されることを利用して、 それ ぞれの組織分布を調べた。 PCR反応は Takara Ex Taq (宝酒造社製) を用いて サーマルサイクラ一 GeneAmp PCR System 9600 (アプライドバイオシステムズ社 製) にて、 最初 94°C30秒間おいた後で、 94 で 10秒、 6(TCで 30秒、 72°Cで 2分を 1反応サイクルとして 35サイクル繰り返すことにより行つた。 得られた反応産物 を PvuIIで 化し、 2%ァガロースゲルにて電気泳動にかけた後、 臭化工チジゥ ムで染色してバンドを検出した。 その結果、 大腸由来の増幅した DNA断片は消化 されなかつ..たが、 平滑筋由来の増幅した DNA断片はそのうちの約半量が PvuIIに より消化され、 マウス CLCA4遺伝子産物 (mRNA) は平滑筋および大腸に発現して いるが、 マウス CLCA4A遺伝子産物 (mRNA) は平滑筋にのみ発現していることが 判明した。 実施例 5 A 1020 bp DNA fragment amplified by PCR using a combination of primer 2 (SEQ ID NO: 6) and primer 3 (SEQ ID NO: 7), which are the common sequence of mouse CLCA4 and mouse CLCA4A, is derived from mouse CLCA4 by the restriction enzyme PvuII (Takara Shuzo) Using the fact that mouse CLCA4A-derived DNA was digested to 680 bp and 340 bp, the tissue distribution of each was examined. The PCR reaction was performed using Takara Ex Taq (Takara Shuzo) in a thermal cycler GeneAmp PCR System 9600 (Applied Biosystems) at 94 ° C for 30 seconds, then at 94 for 10 seconds and 6 (TC The reaction was carried out by repeating 35 cycles with 1 reaction cycle consisting of 30 seconds and 2 minutes at 72 ° C. The obtained reaction product was converted into PvuII, subjected to electrophoresis on a 2% agarose gel, and then stained with CHBr. As a result, the amplified DNA fragment derived from the large intestine was not digested, but about half of the amplified DNA fragment derived from smooth muscle was digested with PvuII, and the mouse CLCA4 gene product ( mRNA) was expressed in smooth muscle and large intestine, whereas mouse CLCA4A gene product (mRNA) was found to be expressed only in smooth muscle. Example 5
ラット CLCA4遺伝子のクローニング  Cloning of rat CLCA4 gene
ヒト CLCA4遺伝子の塩基配列 (FEBS Letters, 455巻、 295頁、 1999年) 、 マウ ス CLCA4遺伝子の塩基配列 (配列番号: 2) およびマウス CLCA4A遺伝子の塩基配 列を参考にして 2種のプライマー 〔プライマ一 12 (配列番号: 22) およびプ ライマ一 13 (配列番号: 23) 〕 を設計 '合成した。 次に、 ラット腸組織か ら IS0GEN (和光純薬社製) を用いて全 RNAを調製した。 さらに mRNA  Two primers were used with reference to the nucleotide sequence of the human CLCA4 gene (FEBS Letters, vol. 455, p. 295, 1999), the nucleotide sequence of the mouse CLCA4 gene (SEQ ID NO: 2), and the nucleotide sequence of the mouse CLCA4A gene. Primer 12 (SEQ ID NO: 22) and Primer 13 (SEQ ID NO: 23)] were designed and synthesized. Next, total RNA was prepared from rat intestinal tissue using IS0GEN (manufactured by Wako Pure Chemical Industries, Ltd.). Further mRNA
purification kit (アマシャムフアルマシアバイオテク社製) を用いて mRNAを 調製した。 この mRNA lpgを出発材料として Marathon cDNA Amplification kit (Clontech社製) を用いて逆転写反応により cDNAを合成した。 この cDNAを铸型 としてプライマー 12とプライマー 13の組み合わせで PCRを行った後、 ァガロ ース電気泳動により予想される大きさの DNA断片が増幅されることを認めた。 反 応は pyrobest DNA polymerase (宝酒造社製) を用いてサ一マルサイクラ一 Gene Amp PCR System 9700 (パーキンエルマ一社製) にて、 最初 94°Cで 30秒間反応さ せた後、 94°Cで 10秒、 60°Cで 30秒、 72°Cで 2分を 1反応サイクルとして 35サイク ル繰り返し、 最後は 72°Cで 10分間反応させた。 得られた DNA断片は pCR-Bluntll - T0P0 (Invitrogen社製) にクロ一ニングした'。 さらに T7 promoter primer (配 列番号: 1 5) 、 M13RV primer (配列番号: 16) および 3種の合成プライマー 〔プライマ一 12 (配列番号: 22) 、 プライマ一 13 (配列番号: 23) 、 プライマー 14 (配列番号: 24) 〕 を用いてサイクルシークェンス反応を行 い、 蛍光 DNAシークェンサ一377 (パーキンエルマ一社製) で得られた反応物の 塩基配列を決定した。 その結果、 ラット CLCA4遺伝子の部分塩基配列として 1119 個の塩基配列を揷入したクローン No. a- 20 (配列番号: 20) を取得した。 配列 番号: 20で表される塩基配列を基に、 373個の部分アミノ酸配列を決定した (配列番号: 1 9) 。 該部分アミノ酸配列は、 ヒト CLCA4の対応する配列とは 66%、 マウス CLCA4およびマウス CLCA4Aの対応する配列とは 81%の相同性を有し ていた。 ' 産業上の利用可能性 mRNA was prepared using a purification kit (Amersham Pharmacia Biotech). Using this mRNA lpg as a starting material, cDNA was synthesized by a reverse transcription reaction using a Marathon cDNA Amplification kit (manufactured by Clontech). After performing PCR using this cDNA as type I and a combination of primers 12 and 13, it was confirmed that a DNA fragment of the expected size was amplified by agarose electrophoresis. The reaction was performed using Pyrobest DNA polymerase (Takara Shuzo Co., Ltd.) in a Thermocycler Gene Amp PCR System 9700 (PerkinElmer Co., Ltd.) first at 94 ° C for 30 seconds, and then at 94 ° C. The reaction cycle was repeated 35 cycles, each consisting of 10 seconds, 30 seconds at 60 ° C, and 2 minutes at 72 ° C. Finally, the reaction was performed at 72 ° C for 10 minutes. The obtained DNA fragment was cloned into pCR-Bluntll-TOP0 (Invitrogen). In addition, T7 promoter primer (SEQ ID NO: 15), M13RV primer (SEQ ID NO: 16) and three synthetic primers [Primer 12 (SEQ ID NO: 22), Primer 13 (SEQ ID NO: 23), Primer 14 (SEQ ID NO: 24)], and the nucleotide sequence of the reaction product obtained with a fluorescent DNA sequencer-377 (manufactured by PerkinElmer) was determined. As a result, a clone No. a-20 (SEQ ID NO: 20) in which 1119 nucleotide sequences were inserted as a partial nucleotide sequence of rat CLCA4 gene was obtained. Based on the nucleotide sequence represented by SEQ ID NO: 20, a partial amino acid sequence of 373 was determined (SEQ ID NO: 19). The partial amino acid sequence had 66% homology with the corresponding sequence of human CLCA4 and 81% homology with the corresponding sequences of mouse CLCA4 and mouse CLCA4A. '' Industrial applicability
本発明のタンパク質およびポリヌクレオチドは、 例えば肺 ·気道の炎症を伴 う肺 ·胸部疾患などの呼吸器疾患 〔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、 過敏性肺炎な ど〕 、 アレルギー性結膜炎、 鼻炎 (例、 アレルギ一性鼻炎、 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) 、 消化管疾患 (例、 過敏性腸症候群、 炎症性腸疾患、 潰 瘍性大腸炎、 クローン病、 逆流性食道炎など) 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 を用いるスクリーニングにより得られる阻害剤、 該タンパク質の活性を阻害す る中和抗体などは、 例えば、 肺 ·気道の炎症を伴う肺 ·胸部疾患などの呼吸器 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、 過敏性肺炎など〕 、 アレルギー性結膜炎、 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、 血管琿動性鼻炎、 壊疽性鼻炎、 副鼻腔炎など) 、 消化 管疾患 (例、 過敏性腸症候群、 炎症性腸疾患、 潰瘍性大腸炎、 クローン病、 逆 流性食道炎など) などの予防 ·治療剤、 好ましくは呼吸器疾患、 消化管疾患な どの予防 ·治療剤として使用することができる。  The proteins and polynucleotides of the present invention include, for example, lungs, lungs with inflammation of the airways, respiratory diseases such as chest diseases [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolosis Inflammation, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), allergic conjunctivitis, rhinitis (eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, before drying Rhinitis, vasomotor rhinitis, gangrenous rhinitis, sinusitis, etc., gastrointestinal diseases (eg, irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease, reflux esophagitis, etc.) It is useful as a diagnostic marker or the like, and is an inhibitor obtained by screening using the protein, polynucleotide or an antibody against the protein, and a neutralizing antibody that inhibits the activity of the protein. For example, respiratory diseases such as lungs, lungs with inflammation of the respiratory tract, and chest disease [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis Disease, irritable pneumonia, etc.), allergic conjunctivitis, rhinitis (eg, allergic rhinitis, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry prorhinitis, vascular hunting rhinitis, gangrene Prophylactic / therapeutic agents, such as rhinitis, sinusitis, etc., gastrointestinal diseases (eg, irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease, reflux esophagitis, etc.), preferably respiration It can be used as a preventive and therapeutic agent for organ diseases and gastrointestinal diseases.
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、 肺 ·気道の炎症を伴う肺 ·胸部疾患などの 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、 過敏性肺炎など〕 、 アレルギー性結膜 炎、 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) 、 消化管疾患 (例、 過敏性腸症候群、 炎症性腸疾患、 潰瘍性大腸炎、 クローン病、 逆流性食道炎など) などの疾病の予防 ·治療剤、 好ましくは呼吸器疾患、 消化 管疾患などの予防。治療剤など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肺 ·気道の炎症を伴う肺 ·胸部疾患などの呼吸器 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、 過敏性肺炎など〕 、 アレルギー性結膜炎、 鼻 炎 (例、 ア 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) 、 消化 管疾患 (例、 過敏性腸症候群、 炎症性腸疾患、 潰瘍性大腸炎、 クローン病、 逆 流性食道炎など) などの診断、 予防または治療に有用である。 In addition, the antisense polynucleotide of the present invention can suppress the expression of the protein of the present invention. Examples thereof include respiratory diseases such as lungs, lungs accompanied by airway inflammation, and chest diseases (eg, chronic obstructive pulmonary disease). Diseases (chronic bronchitis, emphysema), diffuse panbronchiolitis, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), allergic conjunctivitis, rhinitis (eg, allergic rhinitis, pollinosis, acute rhinitis, chronic) Rhinitis, hypertrophic rhinitis, atrophic rhinitis, dry proctitis, vasomotor rhinitis, 'gangrene rhinitis, sinusitis, etc.), gastrointestinal disorders (eg, irritable bowel syndrome, inflammatory bowel disease, ulcerative colon) Prevention and treatment of diseases such as inflammation, Crohn's disease, reflux esophagitis, etc., preferably prevention of respiratory diseases, gastrointestinal diseases, etc. It can be used as a therapeutic agent and the like. Furthermore, various polynucleotides of the present invention may be used for respiratory diseases such as lungs, lungs with airway inflammation, and chest diseases [eg, chronic obstructive pulmonary disease (chronic bronchitis, emphysema), diffuse panbronchiole Inflammation, bronchial asthma, cystic fibrosis, irritable pneumonia, etc.), allergic conjunctivitis, rhinitis (eg, rhinorrhea, hay fever, acute rhinitis, chronic rhinitis, hypertrophic rhinitis, atrophic rhinitis, before drying Rhinitis, vasomotor rhinitis, gangrenous rhinitis, sinusitis, etc., gastrointestinal disorders (eg, irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease, reflux esophagitis, etc.) Useful for diagnosis, prevention or treatment.

Claims

請求 の 範 囲 The scope of the claims
I . 配列番号: 1もしくは配列番号: 1 7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塩。 I. A protein containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 or SEQ ID NO: 17, or a salt thereof.
2 . 配列番号: 1で表されるアミノ酸配列からなるタンパク質またはその塩。 2. A protein comprising an amino acid sequence represented by SEQ ID NO: 1 or a salt thereof.
3 . 配列番号: 1 7で表されるアミノ酸配列からなるタンパク質またはその 塩。 3. A protein comprising an amino acid sequence represented by SEQ ID NO: 17 or a salt thereof.
4. 配列番号: 1 9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塩。  4. A protein having an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 19, or a salt thereof.
5 . 配列番号: 1 9で表されるアミノ酸配列を含有するタンパク質またはそ の塩。  5. A protein comprising an amino acid sequence represented by SEQ ID NO: 19 or a salt thereof.
6 . 請求項 1記載の夕ンパク質の部分べプチドまたはその塩。  6. The partial protein of the protein of claim 1 or a salt thereof.
7 . 請求項 4記載のタンパク質の部分べプチドまたはその塩。  7. A partial peptide of the protein according to claim 4, or a salt thereof.
8 . 請求項 1記載の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 クレオチドを含有するポリヌクレオチド。  8. A polynucleotide comprising a polynucleotide encoding the protein of claim 1 or a partial peptide thereof.
9 . 請求項 4記載の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ポリヌクレオチド。  9. A polynucleotide containing a polynucleotide encoding the protein according to claim 4 or a partial peptide thereof.
1 0 . D NAである請求項 8または請求項 9記載のポリヌクレオチド。  10. The polynucleotide according to claim 8 or 9, which is 10. DNA.
I I . 配列番号: 2または配列番号: 1 8で表される塩基配列からなるポリ ヌクレオチド。  II. A polynucleotide consisting of the nucleotide sequence of SEQ ID NO: 2 or SEQ ID NO: 18.
1 2 . 配列番号: 2 0で表される塩基配列を含有する請求項 1 0記載のポリ ヌクレオチド。  12. The polynucleotide according to claim 10, comprising the nucleotide sequence represented by SEQ ID NO: 20.
1 3 . 請求項 8または請求項 9記載のポリヌクレオチドを含有する組換えべ 1 4. 請求項 1 3記載の組換えべクタ一で形質転換された形質転換体。  13. A transformant containing the polynucleotide according to claim 8 or 9. 14. A transformant transformed with the recombinant vector according to claim 13.
1 5 . 請求項 1 4記載の形質転換体を培養し、 請求項 1もしくは請求項 4記 載のタンパク質またはその部分ペプチドまたはそれらの塩を生成、 蓄積せしめ、 これを採取することを特徴とする請求項 1もしくは請求項 4記載の夕ンパク質 またはその部分べプチドまたはそれらの塩の製造法。 15. The method according to claim 14, wherein the transformant according to claim 14 is cultured to produce and accumulate the protein according to claim 1 or claim 4, a partial peptide thereof, or a salt thereof, and collect this. A method for producing the protein or its partial peptide or a salt thereof according to claim 1 or 4.
1 6 . 請求項 1もしくは請求項 4記載のタンパク質またはその部分ペプチド またはそれらの塩を含有してなる医薬。 16. A medicament comprising the protein according to claim 1 or 4, or a partial peptide thereof, or a salt thereof.
1 7 . 請求項 1または請求項 4記載のタンパク質またはその部分ペプチドま たはそれらの塩に対する抗体。  17. An antibody against the protein according to claim 1 or 4, or a partial peptide thereof, or a salt thereof.
1 8 . 請求項 1 7記載の抗体を含有してなる医薬。  18. A pharmaceutical comprising the antibody according to claim 17.
1 9 . 請求項 1 7記載の抗体を含有してなる診断薬。  19. A diagnostic agent comprising the antibody according to claim 17.
2 0 . 請求項 8または請求項 9記載の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を含有するアンチセンスポ リヌクレオチド。  20. An antisense polynucleotide comprising a nucleotide sequence complementary to or substantially complementary to the nucleotide sequence of the polynucleotide according to claim 8 or 9, or a part thereof.
2 1 . 請求項 2 0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。 2 2 . 請求項 1もしくは請求項 4記載のタンパク質またはその部分ペプチド またはそれらの塩を用いることを特徴とする、 請求項 1もしくは請求項 4記載 のタンパク質またはその部分べプチドまたはそれらの塩の活性を阻害する化合 物またはその塩のスクリーニング方法。  21. A medicament comprising the antisense polynucleotide according to claim 20. 22. The activity of the protein according to claim 1 or claim 4, or a partial peptide thereof or a salt thereof, wherein the protein according to claim 1 or claim 4 or a partial peptide thereof or a salt thereof is used. A method for screening for a compound that inhibits or a salt thereof.
2 3 . 請求項 1もしくは請求項 4記載のタンパク質またはその部分ペプチド またはそれらの塩を含有することを特徴とする、 請求項 1もしくは請求項 4記 載のタンパク質またはその部分ペプチドまたはそれらの塩の活性を阻害する化 合物またはその塩のスクリーニング用キット。  23. The protein according to claim 1 or claim 4, or a partial peptide thereof, or a salt thereof, characterized by containing the protein according to claim 1 or claim 4, a partial peptide thereof, or a salt thereof. A kit for screening for a compound that inhibits the activity or a salt thereof.
2 4. 請求項 2 2記載のスクリーニング方法または請求項 2 3記載のスクリ —ニング用キットを用いて得られうる、 請求項 1もしくは請求項 4記載のタン パク質またはその部分べプチドまたはそれらの塩の活性を阻害する化合物また はその塩。  2 4. The protein according to claim 1 or 4, or a partial peptide thereof or a partial peptide thereof, which can be obtained by using the screening method according to claim 22 or the screening kit according to claim 23. A compound that inhibits the activity of a salt or a salt thereof.
2 5 . 請求項 2 4記載の化合物またはその塩を含有してなる医薬。  25. A medicament comprising the compound according to claim 24 or a salt thereof.
2 6 . 請求項 8または請求項 9記載のポリヌクレオチドを用いることを特徴 とする、 請求項 1または請求項 4記載のタンパク質遺伝子の発現を阻害する化 合物またはその塩のスクリーニング方法。  26. A method for screening a compound or a salt thereof that inhibits the expression of the protein gene according to claim 1 or 4, characterized by using the polynucleotide according to claim 8 or 9.
2 7 . 請求項 8または請求項 9記載のポリヌクレオチドを含有することを特 徴とする、 請求項 1または請求項 4記載のタンパク質遺伝子の発現を阻害する 化合物またはその塩のスクリーニング用キット。 27. A kit for screening a compound that inhibits the expression of the protein gene according to claim 1 or 4, or a salt thereof, which comprises the polynucleotide according to claim 8 or 9.
2 8 . 請求項 2 6記載のスクリーニング方法または請求項 2 7記載のスクリ 一二ング用キットを用いて得られうる、 請求項 1または請求項 4記載のタンパ ク質遺伝子の発現を阻害する化合物またはその塩。 28. A compound that inhibits expression of the protein gene according to claim 1 or 4, which can be obtained by using the screening method according to claim 26 or the screening kit according to claim 27. Or its salt.
2 9 . 請求項 2 8記載の化合物またはその塩を含有してなる医薬。  29. A medicament comprising the compound according to claim 28 or a salt thereof.
3 0 . 請求項 1 7記載の抗体を用いることを特徴とする、 請求項 1または請 求項 4記載のタンパク質の発現を阻害する化合物またはその塩のスクリーニン グ方法。  30. A method for screening a compound or a salt thereof that inhibits the expression of a protein according to claim 1 or claim 4, characterized by using the antibody according to claim 17.
3 1 . 請求項 1 7記載の抗体を含有することを特徴とする、 請求項 1または 請求項 4記載のタンパク質の発現を阻害する化合物またはその塩のスクリー二 ング用キッ卜。  31. A screening kit for a compound or a salt thereof that inhibits the expression of the protein according to claim 1 or 4, which comprises the antibody according to claim 17.
3 2 . 請求項 3 0記載のスクリーニング方法まだは請求項 3 1記載のスクリ 一二ング用キットを用いて得られうる、 請求項 1または請求項 4記載のタンパ ク質の発現を阻害する化合物またはその塩。  32. A compound that inhibits the expression of the protein according to claim 1 or 4, which can be obtained by using the screening method according to claim 30 or the screening kit according to claim 31. Or its salt.
3 3 . 請求項 3 2記載の化合物またはその塩を含有してなる医薬。  33. A medicament comprising the compound according to claim 32 or a salt thereof.
3 4. 呼吸器疾患、 鼻炎または消化器疾患の予防 ·治療剤である請求項 1 6、 3 4. Claim 16 which is an agent for preventing or treating respiratory diseases, rhinitis or digestive diseases.
1 8、 2 1、 2 5、 2 9または 3 3記載の医薬。 18. The medicament according to 18, 21, 25, 29 or 33.
3 5 . 慢性閉塞性肺疾患、 気管支喘息、 炎症性腸疾患または逆流性食道炎の 予防 ·治療剤である請求項 3 4記載の医薬。  35. The medicament according to claim 34, which is an agent for preventing or treating chronic obstructive pulmonary disease, bronchial asthma, inflammatory bowel disease or reflux esophagitis.
3 6 . 呼吸器疾患、 鼻炎または消化器疾患の診断薬である請求項 1 9記載の  36. The method according to claim 19, which is a diagnostic agent for respiratory disease, rhinitis or gastrointestinal disease.
3 7 . 慢性閉塞性肺疾患、 気管支喘息、 炎症性腸疾患または逆流性食道炎の 診断薬である請求項 3 6記載の診断薬。 37. The diagnostic agent according to claim 36, which is a diagnostic agent for chronic obstructive pulmonary disease, bronchial asthma, inflammatory bowel disease or reflux esophagitis.
3 8 . 哺乳動物に対して、 請求項 2 4、 請求項 2 8または請求項 3 2記載の 化合物またはその塩の有効量を投与することを特徴とする慢性閉塞性肺疾患、 気管支喘息、 炎症性腸疾患または逆流性食道炎の予防 ·治療方法。  38. Chronic obstructive pulmonary disease, bronchial asthma, inflammation characterized by administering to a mammal an effective amount of the compound according to claim 24, claim 28 or claim 32 or a salt thereof. How to prevent and treat genital bowel disease or reflux esophagitis.
3 9 . 慢性閉塞性肺疾患、 気管支喘息、 炎症性腸疾患または逆流性食道炎の 予防 ·治療剤を製造するための請求項 2 4、 請求項 2 8または請求項 3 2記載 の化合物またはその塩の使用。 39. The compound according to claim 24, claim 28 or claim 32 for producing a prophylactic or therapeutic agent for chronic obstructive pulmonary disease, bronchial asthma, inflammatory bowel disease or reflux esophagitis, or a compound thereof. Use of salt.
PCT/JP2003/000408 2002-01-21 2003-01-20 Novel protein, its dna and use thereof WO2003062426A1 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
JP2002-12180 2002-01-21
JP2002012180 2002-01-21
JP2002-44981 2002-02-21
JP2002044981 2002-02-21
JP2002-61668 2002-03-07
JP2002061668 2002-03-07

Publications (1)

Publication Number Publication Date
WO2003062426A1 true WO2003062426A1 (en) 2003-07-31

Family

ID=27617297

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2003/000408 WO2003062426A1 (en) 2002-01-21 2003-01-20 Novel protein, its dna and use thereof

Country Status (1)

Country Link
WO (1) WO2003062426A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999044620A1 (en) * 1998-03-03 1999-09-10 Magainin Pharmaceuticals, Inc. Asthma associated factors as targets for treating atopic allergies including asthma and related disorders
WO2002014366A2 (en) * 2000-08-16 2002-02-21 Universiteit Utrecht Genes involved in immune related responses observed with asthma

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999044620A1 (en) * 1998-03-03 1999-09-10 Magainin Pharmaceuticals, Inc. Asthma associated factors as targets for treating atopic allergies including asthma and related disorders
WO2002014366A2 (en) * 2000-08-16 2002-02-21 Universiteit Utrecht Genes involved in immune related responses observed with asthma

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
AGNEL M. ET AL.: "Identification of three novel members of the calcium-dependent chloride channel (CaCC) family predominantly expressed in the digestive tract and treachea", FEBS LETT., vol. 455, no. 3, 1999, pages 295 - 301, XP000986013 *
GRUBER A.D. ET AL.: "Genomic cloning, molecular characterization and functional analysis of human CLCA1, the first human member of the family of Ca2+-activated C1 channel proteins", GENOMICS, vol. 54, no. 2, 1998, pages 200 - 214, XP004448920 *
NAKANISHI A. ET AL.: "Role of gob-5 in mucus overproduction and airway hyperresponsiveness in asthma", PROC. NATL. ACAD. SCI. USA, vol. 98, no. 9, 2001, pages 5175 - 5180, XP002227786 *
NEWSHOLME P. ET AL.: "Identification of a novel complement-dependent serum-elicited inward current in the xenopus oocyte provoking Ca2+ influx and subsequent activation of C1 channels", BIOCHEM. PHARMACOL., vol. 57, no. 5, 1999, pages 491 - 501, XP002967656 *

Similar Documents

Publication Publication Date Title
WO2007004692A1 (en) Prophylactic/therapeutic agent and diagnostic agent for non-small cell lung cancer
WO2001036633A1 (en) Novel protein and dna thereof
WO2001000799A1 (en) Novel protein and dna thereof
WO2000071581A1 (en) Novel polypeptide
WO2003062426A1 (en) Novel protein, its dna and use thereof
WO2004028558A1 (en) Preventives/remedies for neurodegenerative disease
WO2004002515A1 (en) Diagnostics/preventives/remedies for respiratory diseases
WO2003054190A1 (en) Novel proteins and dnas thereof
WO2004024760A1 (en) Novel protein and dna thereof
JP2001029090A (en) Novel polypeptide
JP2001228146A (en) Use of disease-associated gene
WO2004002517A1 (en) Diagnostics/preventives/re medies for respiratory diseases
WO2004095023A1 (en) Novel method of screening
JP2003274982A (en) New protein, its dna and use thereof
WO2003087155A1 (en) Novel protein and dna thereof
WO2002033082A1 (en) Novel disease-associated gene and use thereof
JP2004075675A (en) Prophylactic/remedy for osteal/arthral disease
WO2004002516A1 (en) Diagnostics/preventives/re medies for respiratory diseases
WO2002103020A1 (en) Novel disease-associated gene and use thereof
JP2004121245A (en) New protein and its dna
JP2003325187A (en) New protein, dna encoding the same and application of the same
JP2003189878A (en) New protein and its dna
WO2004013174A1 (en) Use of disease-associated gene
WO2003097686A1 (en) Novel protein, its dna and use thereof
JP2003219880A (en) New protein and dna thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR IE IT LU MC NL PT SE 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