WO2002036772A1 - Novel disease-associated gene and use thereof - Google Patents

Novel disease-associated gene and use thereof Download PDF

Info

Publication number
WO2002036772A1
WO2002036772A1 PCT/JP2001/009477 JP0109477W WO0236772A1 WO 2002036772 A1 WO2002036772 A1 WO 2002036772A1 JP 0109477 W JP0109477 W JP 0109477W WO 0236772 A1 WO0236772 A1 WO 0236772A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
present
salt
dna
cells
Prior art date
Application number
PCT/JP2001/009477
Other languages
French (fr)
Japanese (ja)
Inventor
Nobuyuki Koyama
Seiichi Tanida
Toshifumi Watanabe
Original Assignee
Takeda Chemical Industries, Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Takeda Chemical Industries, Ltd. filed Critical Takeda Chemical Industries, Ltd.
Priority to AU2002210960A priority Critical patent/AU2002210960A1/en
Publication of WO2002036772A1 publication Critical patent/WO2002036772A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6887Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids from muscle, cartilage or connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value

Definitions

  • the present invention relates to uses of disease-related genes. More specifically, a method for screening a drug using the disease-related gene product, an antisense nucleotide against a disease-related gene useful as a marker for diagnosis of heart diseases such as cardiomyopathy, myocardial infarction, heart failure, and angina; It relates to an antibody against a related gene product and the like.
  • a method for screening a drug using the disease-related gene product an antisense nucleotide against a disease-related gene useful as a marker for diagnosis of heart diseases such as cardiomyopathy, myocardial infarction, heart failure, and angina; It relates to an antibody against a related gene product and the like.
  • Heart failure is considered to be inadequate contraction of the heart muscle.
  • the mechanism of the onset may be as follows. Myocardial damage, mechanical and functional abnormalities of the heart pump, pressure overload due to hypertension and pulmonary hypertension, volume overload such as anemia and acute nephritis can cause the heart to pump out blood volume according to the demands of the organism A state of disappearance occurs.
  • the sympathetic nervous system, the nervous system, the endocrine system and other compensatory mechanisms operate to maintain the homeostasis of the body.
  • the compensatory mechanisms of heart failure include: 1) the above-mentioned load increases and the contractility of the heart increases, and the heart expands as the length of the sarcomer increases.2) The contraction unit of the myocardium increases, resulting in Myocardial hypertrophy occurs as a result. 3) Nerve f nocturnal factors are activated to compensate for the inability to excrete the blood necessary for the whole body, and myocardial fibrosis progresses locally. Basically, the mechanism is to adapt to a given load.However, insufficient operation may promote heart failure, while excessive operation may cause myocardial injury and exacerbate heart failure There is also. As a result of activation of the compensatory mechanism, myocardial cells become enlarged and cardiac hypertrophy occurs.
  • cardiotonic drugs include: 1. cardiac glycosides such as digoxin, 2.
  • sympathomimetics such as dobuyumin, and 3. phosphogesterase inhibitors such as amrinone.
  • vasodilators hydralazine, calcium antagonists, angiotensin I converting enzyme inhibitors, angiotensin II receptor antagonists and the like have been used.
  • a / 3 blocker is used for treatment of other dilated cardiomyopathy.
  • the present inventors have conducted intensive studies in order to solve the above-described problems, and as a result, have found a gene whose expression increases when heart failure occurs in a myocardial infarction model rat due to coronary artery ligation.
  • this mRNA was significantly reduced one week after the operation, and decreased from 8 weeks after the operation to 20 weeks. Weeks showed an increasing trend, and at week 30 it was evident that the expression port file returned to the same level as the sham-operated group.
  • Nkx2.5 also known as Csx, Processings of the National Academy of Sciences'. Proc. Natl. Acad. Sci. USA, Vol. 90, pp. 8145-8149, 1993; Development, Vol. 119, pp. 419-431, 1993; History of Medicine 185, pp. 1-7, 1998) and cloned from a rat heart cDNA library. Functional analysis of Nkx2.5 as a transcription factor that plays an important role in the developmental process of the heart is ongoing, but its relationship to heart failure pathology has not been clarified. On the other hand, the Nkx2.5 similar gene cloned this time increased at the onset of heart failure. As a result of further studies based on these findings, the present invention was completed.
  • the present invention (1) a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by arrangement J number: 1, or a salt thereof;
  • the transformant according to (8) is cultured, and the protein or salt thereof according to (1) or the partial peptide or salt thereof according to (3) is produced, accumulated, and collected.
  • the protein or salt thereof according to (1) or the partial peptide or salt thereof according to (3) contains a DNA encoding the protein according to (1) or the partial peptide according to (3).
  • the protein or salt thereof according to (1) which comprises the protein or salt thereof according to (1) or the partial peptide or salt thereof according to (3).
  • a medicament comprising the compound according to (16) or a salt thereof, (18) a medicament according to (17), which is a prophylactic or therapeutic agent for heart disease.
  • (23) a method for preventing or treating heart disease, which comprises administering to a mammal an effective amount of the compound or a salt thereof according to (16);
  • a compound or a salt thereof which comprises administering a test compound to the animal according to (36) and detecting the expression of a reporter gene ⁇ , which promotes or inhibits the activity of a promoter against DNA of the present invention. Also provided are screening methods and the like.
  • FIG. 1 shows an alignment of DNA (NolOrCsx full) and AF006664 (AF006664Csx. SE) encoding the novel Nkx2.5 analogous protein of the present invention shown in Example 1 (continued from FIG. 2).
  • FIG. 2 shows an alignment of DNA (NolOrCsx full) and AF006664 (AF006664Csx.SE) encoding the novel Nkx2.5-like protein of the present invention shown in Example 1 (continued from FIG. 1 and FIG. Continue) .
  • FIG. 3 shows an alignment of DNA (NolOrCsx full) and AF006664 (AF006664Csx. SE) encoding the novel Nkx2.5-like protein of the present invention shown in Example 1 (continued from FIG. 2 and FIG. Continue) .
  • FIG. 4 shows the alignment of DNA (NolOrCsx full) and AF006664 (AF006664Csx.SE) encoding the novel Nkx2.5-like protein of the present invention shown in Example 1 (continued from FIG. 3, FIG. Continue) .
  • FIG. 5 shows the alignment of DNA (NolOrcsx full) and AF006664 (AF006664Csx. SE) encoding the novel Nkx2.5 analogous protein of the present invention shown in Example 1 (FIG. (Continued from Figure 4 and continued from Figure 6).
  • FIG. 6 shows an alignment between DNA (NolO rCsx ful) encoding the novel Nkx2.5 analogous protein of the present invention shown in Example 1 and AF006664 (AF006664Csx.SE) (continuation of FIG. 5 and FIG. 7). Continue) .
  • FIG. 7 shows an alignment between DNA (NolO rCsx ful) encoding the novel Nkx2.5 analogous protein of the present invention and ⁇ 6664 (AF006664Csx.SE) shown in Example 1 (continuation of FIG. 6 and FIG. Continue) .
  • FIG. 8 shows an alignment between DNA (NolO rCsx ful) encoding the novel Nkx2.5 analogous protein of the present invention and AF006664 (AF006664Csx. SE) shown in Example 1 (continuation of FIG. 7, FIG. followed by) .
  • FIG. 9 shows an alignment of DNA (NolO rCsx ful) encoding the novel Nkx2.5 analogous protein of the present invention shown in Example 1 and AF006664 (AF006664Csx. SE) (continuation of FIG. 8).
  • FIG. 10 shows the time course of DNA encoding the novel Nkx2.5-like protein of the present invention in a myocardial infarction model rat.
  • a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 of the present invention (hereinafter, also referred to as the protein of the present invention or the protein used in the present invention.
  • the protein of the present invention and the protein used in the present invention may be used in the sense that their amides and esters are also included.)
  • warm-blooded animals for example, humans, guinea pigs, rats, mice, chickens, Egret, bush, sheep, wedge, monkey, etc.
  • cells eg, hepatocytes, spleen cells, nerve cells, glial cells, knees
  • amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 is about 97% or more, preferably about 98% or more, more preferably the amino acid sequence represented by SEQ ID NO: 1.
  • amino acid sequences having about 99% or more homology include amino acid sequences having about 99% or more homology.
  • Examples of the protein having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 include, for example, a protein having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 described above. However, a protein having substantially the same activity as the protein having the amino acid sequence represented by SEQ ID NO: 1 is preferable.
  • Examples of the activity of substantially the same quality include, for example, activity for promoting cardiac function decline. Substantially identical indicates that the activity is qualitatively (eg, physiologically or pharmacologically) equivalent. Therefore, it is preferable that the activity to promote cardiac function decline is equivalent (eg, about 0.01 to 100 times, preferably about 0.1 to 10 times, more preferably 0.5 to 2 times). However, quantitative factors such as the degree of this activity and the molecular weight of the protein may be different.
  • Measurement of activity such as cardiac dysfunction promoting activity can be measured using an echocardiograph (Cell, Vol. 97, pp. 189-199, 1989) or cardiac function measurement using a cardiac catheter (circulation) (Research, Vol. 69, p. 370—p. 377, 1991).
  • RAS renin-angiotensin system
  • ACE angiotensin I converting enzyme
  • the activity can be measured by using, as an index, the activity of increasing catecholamine in blood (a full-automatic catecholamine analyzer manufactured by Tosoh Corporation).
  • Examples of the protein of the present invention include: (1) an amino acid represented by SEQ ID NO: 1; An amino acid sequence in which one or two or more (preferably about 1 to 10 and more preferably (1 to 5)) amino acids have been deleted from the amino acid sequence; 2) the amino acid represented by SEQ ID NO: 1 An amino acid sequence obtained by adding one or more (preferably about 1 to 10, more preferably a number of (1 to 5)) amino acids to the sequence; 3 the amino acid sequence represented by SEQ ID NO: 1 An amino acid sequence into which 1 or 2 or more (preferably about 1 to 10, more preferably a number (1 to 5)) amino acids have been inserted; 4 in the amino acid sequence represented by SEQ ID NO: 1 An amino acid sequence in which one or more (preferably preferably about 1 to 10 and more preferably 1 to 5) amino acids have been substituted with another amino acid, or an amino acid sequence combining them So-called muti, such as contained protein It is also included.
  • the left end is the N-terminus (amino terminus) and the right end is the C-terminus (carboxyl terminus) according to the convention of peptide labeling.
  • the proteins used in the present invention include C-terminal lipoxyl group (one COOH), carboxylate (one COO—), and amide It may be either (—CONH 2 ) or ester (one COOR).
  • R in the ester e.g., methyl, Echiru, n _ propyl, isopropyl, 6 alkyl group, such as n- heptyl, for example, C 3, such as cyclohexyl cyclopentyl, cyclohexylene - 8 cycloalkyl group, for example, phenyl, ⁇ - naphthyl, such as C 6 - 1 2 Ariru group, e.g., benzyl, C 7 _ 4 Ararukiru groups such as flying one Nafuchiru ⁇ alkyl group such as a phenyl primary alkyl group or flying one naphthylmethyl such phenethyl, Bibaroiruo A xymethyl group or the like is used.
  • 6 alkyl group such as n- heptyl, for example, C 3, such as cyclohexyl cyclopentyl, cyclohexylene - 8 cyclo
  • the protein used in the present invention has a carboxyl group (or propyloxylate) other than the C-terminal
  • the protein used in the present invention includes amidated or esterified lipoxyl group.
  • the ester in this case, for example, the above-mentioned C-terminal ester and the like are used.
  • the amino group of the N-terminal amino acid residue (eg, methionine residue) has a protecting group (eg, formyl group, acetyl group, etc.). It is protected with a protecting such Ashiru group such bets 6 Arukanoiru), which dull evening Min residues of N-terminal region is cleaved in vivo to form pin hole dull evening Min oxide, the side chains of amino acids in the molecular
  • the above substituents eg, 1 OH, 1 SH, amino group, imidazole group, indole group, guanidino group, etc.
  • suitable protecting groups eg, alkenyl groups such as alkanoyl groups such as formyl group, acetyl group, etc.
  • Protected proteins, or multiproteins such as so-called glycoproteins to which sugar chains are bound are also included.
  • protein of the present invention include, for example, the rat-derived protein represented by SEQ ID NO: 1.
  • Partial peptide of the protein of the present invention (hereinafter sometimes referred to as the partial peptide of the present invention. Further, the partial peptide of the present invention may be used in the sense of including its amide form and ester form.) Is a partial peptide of the above-mentioned protein of the present invention, and preferably any peptide having the same activity as the above-mentioned protein of the present invention.
  • one or more (preferably 1 to 5) amino acids in the amino acid sequence are deleted, or one or two or more (preferably, about 1 to 20 amino acids, more preferably about 1 to 10 amino acids, and even more preferably about 1 to 5 amino acids are added, or 1 or 2 or more (preferably , 1 to 5) amino acids may be inserted, or one or more (preferably 1 to 5) amino acids in the amino acid sequence may be substituted with another amino acid.
  • the partial peptide of the present invention in the amino acid sequence represented by SEQ ID NO: 1, (1) the 53rd (G1u) to 55th (Tyr) )) A partial amino acid sequence consisting of amino acid residues, 2 a partial amino acid sequence consisting of the 131st (Arg) to 134th (Ala) amino acid residues from the N-terminal, 3 And the partial amino acid sequence consisting of the 164th (Ala) to 165th (Pro) amino acid residues, and 4 the 266th (A1a) to 266th from the N-terminal.
  • the above-mentioned partial peptides of the protein of the present invention having any or a plurality (2 to 5) of the selected partial amino acid sequences are exemplified.
  • the C-terminus of the present invention the force Rupokishiru group (one CO OH), Cal Pokishireto (one COO-), amide (- CO NH 2) or an ester (one COOR) (R is as defined above ).
  • the partial peptide of the present invention has an amino terminal residue at the N-terminal (eg, methionine residue) in which the amino group is protected by a protecting group, and the N-terminal side has a native amino acid residue.
  • N-terminal eg, methionine residue
  • composite peptides are also included.
  • 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
  • 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 may be produced by a known protein purification method from human or warm-blooded animal cells or tissues as described above, or by culturing a transformant containing DNA encoding the protein. It can be manufactured by Further, it can be produced according to the peptide synthesis method described later.
  • the extract When manufactured from human or warm-blooded animal tissues or cells, human or warm-blooded animal After homogenizing the tissue or cells, the extract can be extracted with an acid or the like, and the extract can be purified and isolated by a combination of chromatography such as reverse phase chromatography and ion exchange chromatography.
  • a commercially available resin for protein synthesis can be usually used.
  • resins include, for example, chloromethyl resin, hydroxymethyl resin, benzhydrylamine resin, aminomethyl resin, 4-benzyloxybenzyl alcohol resin, 4-methylbenzhydrylamine resin, PAM resin, 4-hydroxymethyl resin Methyl phenylacetamidomethyl resin, polyacrylamide resin, 4- (2 ', 4, dimethoxyphenyl-hydroxymethyl) phenoxy resin, 4-(2', 4 'dimethoxyphenyl-Fmocaminoethyl) phenoxy resin And so on.
  • an amino acid having a suitably protected amino group and side chain functional group is condensed on the resin according to the sequence of the target protein according to various known condensation methods.
  • the protein 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 a target protein or partial peptide or an amide thereof.
  • the protected amino acid may be added directly to the resin along with a racemization inhibitor additive (eg, H ⁇ Bt, HOOBt) or as a symmetrical anhydride or HOBtester or HOOBtester. After the protected amino acid is activated in advance, it can be added to the resin.
  • a racemization inhibitor additive eg, H ⁇ Bt, HOOBt
  • HOBtester or HOOBtester After the protected amino acid is activated in advance, it can be added to the resin.
  • the solvent used for activating the protective amino acid or condensing with the resin can be appropriately selected from solvents known to be usable for the protein condensation reaction.
  • acid amides such as N, N-dimethylformamide, N, N-dimethylacetamide, N-methylpiperidone, and halogenated carbons such as methylene chloride and chloroform.
  • Hydrogens, alcohols such as trifluoroethanol, sulfoxides such as dimethyl sulfoxide, ethers such as pyridine, dioxane, and tetrahydrofuran; nitriles such as acetonitrile and propionitrile; methyl acetate and ethyl acetate; Esters or an appropriate mixture thereof are used.
  • the reaction temperature is appropriately selected from a range that is known to be usable for a protein bond formation reaction, and is usually appropriately selected from a range of about 120 ° C. to 50 ° C.
  • the activated amino acid derivative is usually used in a 1.5 to 4-fold excess.
  • Examples of the protecting group for the amino group of the starting material include Z, Boc, t-pentyloxycarbonyl, isopolnyoxycarbonyl, 4-methoxybenzyloxycarbonyl, C11Z, Br-Z, and adaman.
  • Tyloxycarbonyl, trifluoroacetyl, phthaloyl, formyl, 2-nitrophenylsulfenyl, diphenylphosphinothioyl, Fmoc and the like are used.
  • the carboxyl group may be, for example, alkyl-esterified (eg, methyl, ethyl, propyl, butyl, t-butyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, 2-adamantyl, etc.).
  • alkyl-esterified eg, methyl, ethyl, propyl, butyl, t-butyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, 2-adamantyl, etc.
  • Alkyl esterification Alkyl esterification
  • aralkyl esterification eg, benzyl ester, 4-nitrobenzyl ester, 4-methoxybenzyl ester, 4-chlorobenzyl ester, benzhydryl esterification
  • phenacyl esterification benzyloxycarbonyl It can be protected by hydrazide, t-butoxycarbonyl hydrazide, trityl hydrazide and the like.
  • the hydroxyl group of serine can be protected, for example, by esterification or etherification.
  • a group suitable for the esterification include a lower group such as an acetyl group ((: an aroyl group such as an alkanoyl group and a benzoyl group, a group derived from carbonic acid such as a benzyloxycarbonyl group and an ethoxycarbonyl group). It is.
  • groups 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 and C 1 2 - B z K 2_ nitrobenzyl, B r- Z, such as t one-heptyl is used.
  • protecting group for imidazole of histidine for example, Tos, 4-methoxy-2,3,6-trimethylbenzenesulfonyl, DNP, benzyloxymethyl, Bum, Boc, Trt, Fmoc and the like are used.
  • Examples of the activated form of the carboxylic group of the raw material include, for example, corresponding acid anhydrides, azides, and active esters [alcohols (eg, phenol, 2,4,5-trichlorophenol, 2,4 —Dinitrophenol, cyanomethyl alcohol, paranitrophenol, HONB, N-hydroxysuccinimide, N-hydroxyphthalimide, ester with HOB t)].
  • alcohols eg, phenol, 2,4,5-trichlorophenol, 2,4 —Dinitrophenol, cyanomethyl alcohol, paranitrophenol, HONB, N-hydroxysuccinimide, N-hydroxyphthalimide, ester with HOB t
  • As the activated amino group of the raw material for example, a corresponding phosphoric amide is used.
  • Methods for removing (eliminating) protecting groups include, for example, catalytic reduction in a stream of hydrogen in the presence of a catalyst such as Pd-black or Pd-carbon, or hydrogen fluoride anhydride, methanesulfonic acid, or trifluoromethane.
  • a catalyst such as Pd-black or Pd-carbon, or hydrogen fluoride anhydride, methanesulfonic acid, or trifluoromethane.
  • 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 Also used.
  • the elimination reaction by the above-mentioned acid treatment is generally performed at a temperature of about 120 ° C to 40 ° C.
  • anisol for example, anisol, phenol, thioanisole, methacresol, paracresol, dimethylsulfur
  • a force-thione scavenger such as FID, 1,4-butanedithiol, 1,2-ethanedithiol, etc.
  • the 2,4-dinitrophenyl group used as an imidazole protecting group of histidine is removed by thiophenol treatment
  • the formyl group used as an indole protecting group of tryptophan is the above 1,2-ethanedithiol, 1,4-1.
  • an amide form of a protein or partial peptide for example, first, after amidating and protecting a lipoxyl group of a carboxy terminal amino acid, a peptide (protein) chain having a desired chain length on the amino group side is obtained. And a protein or partial peptide from which only the protecting group at the N-terminal ⁇ -amino group of the peptide chain has been removed and a protein or partial peptide from which only the protecting group at the C-terminal carboxyl group has been removed. Are produced, and these proteins or peptides are condensed in a mixed solvent as described above. Details of the condensation reaction are the same as described above.
  • the crude protein or peptide is purified by using various known purification means, and the main fraction is lyophilized to obtain an amide of the desired protein or peptide.
  • an ester of a protein or peptide for example, after condensing the carboxyl group of the carboxy terminal amino acid with a desired alcohol to form an amino acid ester, the desired protein can be obtained in the same manner as the amide of a protein or peptide. Alternatively, an ester of a peptide can be obtained.
  • the protein or 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 peptidase.
  • a method for synthesizing a peptide for example, any of a solid phase synthesis method and a liquid phase synthesis method may be used. That is, the desired peptide can be produced by condensing a partial peptide or amino acid capable of constituting the partial peptide of the present invention with the remaining portion, and if the product has a protective group, removing the protective group to produce the desired peptide.
  • Known condensation methods and elimination of protecting groups include, for example, the methods described in the following 1 to 5.
  • the partial peptide used in the present invention can be purified and isolated by a combination of ordinary purification methods such as solvent extraction, distillation, column chromatography, liquid chromatography, and recrystallization.
  • the protein or partial peptide obtained by the above method is a free form, it can be converted to an appropriate salt by a known method or a method analogous thereto, and conversely, when the protein or the partial peptide is obtained by a salt, it is known. It can be converted to the free form or other salts by the method of or a method analogous thereto.
  • the polynucleotide encoding the protein of the present invention may be any polynucleotide containing a base sequence (DNA or RNA, preferably DNA) encoding the protein of the present invention.
  • 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. If single-stranded, it may be the sense strand (ie, the coding strand) or the antisense strand (ie, the non-coding strand).
  • RNA of the protein of the invention can be quantified.
  • the DNA encoding the protein of the present invention may be any DNA as long as it contains the above-described nucleotide sequence encoding the protein of the present invention.
  • any of genomic DNA, genomic DNA library, the above-mentioned cells, tissue-derived cDNA, the above-mentioned cell ⁇ tissue-derived cDNA library, and synthetic DNA May be.
  • the vector used for the library may be any of bacteriophage, plasmid, cosmid, phagemid and the like.
  • amplification can be directly performed by reverse transcription polymerase chain reaction (hereinafter abbreviated as RT-PCR method) using a total RNA or mRNA fraction prepared from the cells and tissues described above.
  • Examples of the DNA encoding the protein of the present invention include a DNA having the nucleotide sequence represented by SEQ ID NO: 2 or a DNA having the nucleotide sequence represented by SEQ ID NO: 2 and a high stringency. Any DNA may be used as long as it contains DNA that hybridizes under the conditions and encodes a protein having substantially the same properties as the protein of the present invention.
  • Examples of a DNA that can hybridize under high stringent conditions with a DNA containing the nucleotide sequence represented by SEQ ID NO: 2 include, for example, about 97% or more of the nucleotide sequence represented by SEQ ID NO: 2, Preferably, a DNA containing a base sequence having a homology of about 98% or more, more preferably about 99% or more is used.
  • Hybridization can be performed according to a known method or a method analogous thereto, for example, the method described in Molecular Cloning 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). Can be. 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 stringent conditions.
  • High stringent conditions include, for example, a sodium concentration of about 19 to 40 mM, preferably about 19 to 20 mM, and a temperature of about 50 to 70 ° (: The conditions are from 0 to 65. Particularly preferred is a sodium concentration of about 19 mM and a temperature of about 65 ° C.
  • DNA encoding the protein containing the amino acid sequence represented by SEQ ID NO: 1 a DNA containing the base sequence represented by SEQ ID NO: 2 or the like is used.
  • Examples of the DNA encoding the partial peptide of the present invention include the aforementioned partial peptide of the present invention. Any DNA containing a nucleotide sequence encoding the peptide may be used. Further, any of genomic DNA, genomic DNA library, cDNA derived from the above-described cells and tissues, cDNA library derived from the above-described cells and tissues, and synthetic DNA may be used.
  • Examples of the DNA encoding the partial peptide of the present invention include a DNA having a partial base sequence of DNA containing the base sequence represented by SEQ ID NO: 2, or a base sequence represented by SEQ ID NO: 2
  • a partial nucleotide sequence of a DNA encoding a protein having a DNA that hybridizes with DNA under high stringent conditions and having substantially the same activity as a protein containing the amino acid sequence represented by SEQ ID NO: 1 DNA or the like is used.
  • Examples of the DNA that can hybridize with the DNA containing the nucleotide sequence represented by SEQ ID NO: 2 under high stringent conditions include, for example, about 97% or more, preferably about 98%, of the nucleotide sequence represented by SEQ ID NO: 2 %, More preferably a DNA containing a base sequence having a homology of about 99% or more is used.
  • the DNA encodes the partial peptide of the present invention in the amino acid sequence represented by SEQ ID NO: 1, (1) 53rd (G1u) to 55th (Ty r) the base sequence encoding the partial amino acid sequence consisting of the amino acid residues (eg, in the base sequence represented by SEQ ID NO: 2, the partial base sequence from the 157th to the 165th from the 5 'end (sequence No .: 18)), 2 Nucleotide sequence encoding the partial amino acid sequence consisting of the 131st (Arg) to 134th (Ala) amino acid residues from the N-terminus (eg, SEQ ID NO: 2 391st to 402th partial nucleotide sequence from the 5 'end (SEQ ID NO: 19)), 3 164th (A1a) to 165th (Pro) from the N-terminal
  • a nucleotide sequence encoding an amino acid sequence eg, 5 ′ in the nucleotide sequence represented by
  • Hybridization methods and high stringency conditions are the same as described above.
  • Cloning means may be amplified by the PCR method using a synthetic DNA primer having a part of the nucleotide sequence encoding the protein of the present invention, or the DNA incorporated into an appropriate vector may be used as the DNA of the present invention. Selection can be performed by hybridization with a DNA fragment encoding a part or the whole region of the protein or labeled with a synthetic DNA.
  • Hybridization can be carried out, for example, according to the method described in Molecular-Cloning (Molecular Cloning) 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). When a commercially available library is used, it can be performed according to the method described in the attached instruction manual.
  • the DNA base sequence can be converted using PCR or a known kit, for example, Mutan TM -suer Expression Km (Takara Shuzo), Mutan TM -K (Takara Shuzo), etc.
  • the method can be carried out according to a known method such as the Gap ed dulex method or the Knuke 1 method or a method analogous thereto.
  • the DNA encoding the cloned protein can be used as it is depending on the purpose, or can be used by digesting with a restriction enzyme or adding a linker if desired.
  • the DNA may have ATG as a translation initiation codon on its 5, terminal side, and may have TAA, TGA or TAG as a translation termination codon on its 3, terminal side. These translation start codons and translation stop codons are It can also be added using one.
  • the expression vector for the protein of the present invention includes, for example, (a) cutting out a DNA fragment of interest from DNA encoding the protein of the present invention, and (mouth) ligating the DNA fragment downstream of a promoter in an appropriate expression vector. It can be manufactured by
  • 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 (eg, pSH 19, pSH15), bacteriophages such as ⁇ phage, animal viruses such as retrovirus, vaccinia virus, baculovirus, etc., pAl-11, XT1, ⁇ Rc / CMV, pRc / RSV, pcD NA I ZN eo or the like is used.
  • Escherichia coli eg, pBR322, pBR325, pUC12, pUC13
  • Bacillus subtilis eg, pUB110, TP5, pC194
  • yeast eg, pSH 19, pSH15
  • the promoter used in the present invention may be any promoter as long as it is appropriate for the host used for gene expression.
  • SRa promoter when animal cells are used as host, SRa promoter, SV40 promoter, LTR motor, CMV promoter, HSV-TK promoter, etc. are mentioned.
  • CMV (cytomegalovirus) promoter, SRa promoter It is preferable to use one or the like.
  • tr When the host is Eshierihia genus bacteria, tr [rho promoter, lac promoter, re cA promoter, AP L promoter, if 1 pp promoter, T 7 including promoter Isseki one is, host Ru der Bacillus, When the host is yeast, such as SPOL promoter, SPO2 promoter, penP promoter, etc., PH05 promoter, PGK promoter, GAP promoter, ADH promoter, etc. are preferable. When the host is an insect cell, a polyhedrin promoter, a P10 promoter and the like are preferable.
  • the expression vector may further include an enhancer, a splicing signal, a polyA addition signal, a selection marker, an SV40 replication origin (hereinafter, sometimes abbreviated as SV40 ori), and the like, if desired.
  • Selection markers include, for example, dihydrofolate reductase (hereinafter sometimes abbreviated as dh fr) gene (methotrexate (MTX) resistance), ampicillin Phosphorus resistant gene (hereinafter sometimes abbreviated as Amp 1 "), neomycin resistance gene (hereinafter sometimes abbreviated as Ne o r, G41 8 resistance).
  • dh fr genetic defects
  • the target gene can be selected even on a thymidine-free medium.
  • a signal sequence suitable for the host is added to the N-terminal side of the protein of the present invention. If the host is Escherichia, Pho A signal sequence, OmpA signal sequence, etc., if the host is Bacillus, ⁇ — amylase signal sequence, subtilisin signal sequence, etc. In the case of yeast, MFa signal sequence, SUC2 signal sequence, etc. In the case of host animal cells, use of insulin signal sequence, one interferon, signal sequence, antibody molecule, signal sequence, etc. it can.
  • a transformant can be produced.
  • Escherichia bacteria for example, Escherichia bacteria, Bacillus bacteria, yeast, insect cells, insects, animal cells, and the like are used.
  • Esdieric hia coli Kl 2 ⁇ DH1 Procedures of the national academy 'ob' (Proc. Natl. Acad. ScI. USA), Vol. 60, 160 (1968)], JM103 [Nucleic Acids Research], (Nucleic Acids Research), Vol. 9, 309 (1981)], JA2 21 [Journal of Molecular Biology, 120, 517 (1978)], HB 101 [Journal of Molecular Biology, Biology, 41, 459 (1969)] ], C600 [Genetics, 39, 440 (1954)] and the like.
  • Bacillus bacteria examples include, for example, Bacillus subtilis MI 114 [Gene, 24, 255 (1983)], 207-21 [Journal of Biochemistry, 95] , 87 (1 984)].
  • yeast examples include, for example, Saccharomyces cerevisiae AH22, AH22R-, NA87-11A, DKD- 5D, 20B-12, Shizosaccharomyces' bomb (Sdiizosaccharomyces pombe) NCYC 1913, NCYC 2036, Pichia 'Pichia pastoris KM71 etc. is used.
  • Insect cells include, for example, when the virus is Ac NPV, a cell line derived from the larvae of night moth (Spodoptera frugiperda cell; S f cell); High Five TM cells, cells derived from Mamestra b rassicae or cells derived from Estigmena acrea are used.
  • S f cell a cell line derived from silkworm (Bombyx mori N cell; BmN cell) is used.
  • Sf cell include Sf9 cell (ATCC CRL1711), Sf21 cell (Vaughn, JL et al., In Vivo, 13, 213-217, (1977)) and the like. Used.
  • insects for example, silkworm larvae are used [Maeda et al., Nature, 315, 592 (1985)].
  • animal cells examples include monkey cells COS-7, Vero, Chinese Hamster cells CHO (hereinafter abbreviated as CHO cells), dh fr gene-deficient Chinese Hamster Yuichi cells CHO (hereinafter CHO (dhfr_) cells) Abbreviations), mouse L cells, mouse AtT-20, mouse myeloma cells, rat GH3, human FL cells, H9c2 cells, etc. are used.
  • Transformation of Escherichia sp. Is described, for example, in Proc. Natl. Acad. Sci. USA, Proc. Natl. Acad. Sci. , 2110 (1972) and Gene, 17, 107 (1982). Transformation of Bacillus spp. Can be performed, for example, according to the method described in Molecular & General Genetics, Vol. 168, 111 (1979).
  • Insect cells or insects can be transformed, for example, according to the method described in Bio Z Technology (Bio / Technology), 6, 47-55 (1988). In order to transform animal cells, for example, Cell Engineering Separate Volume 8 New Cell Engineering Experiment Protocol. 263—267 (1995) (published by Shujunsha), Virology, 52, 456 (1973) Can be performed according to the method described in (1).
  • a liquid medium is suitable as the medium used for the culturing, and a carbon source necessary for the growth of the transformant is contained therein.
  • the carbon source include glucose, dextrin, soluble starch, and sucrose.
  • the nitrogen source include ammonium salts, nitrates, corn chip liquor, peptone, casein, meat extract, soybean meal, potato extract, and the like.
  • the inorganic or organic substance and the inorganic substance include calcium chloride, sodium dihydrogen phosphate, magnesium chloride and the like.
  • yeast extract, vitamins, growth promoting factors and the like may be added.
  • the H of the medium is preferably about 5-8.
  • a medium for cultivating a bacterium belonging to the genus Escherichia for example, an M9 medium containing glucose and casamino acid [Miller, Journal of Exp. lar Genetics), 431-433, Cold Spring Harbor Laboratory, New York 1972].
  • a drug such as 3-indolylacrylic acid can be added to make the promoter work efficiently.
  • cultivation is usually performed at about 15 to 43 ° C for about 3 to 24 hours, and if necessary, aeration and stirring can be applied.
  • culturing is usually performed at about 30 to 40 ° C for about 6 to 24 hours, and if necessary, aeration and stirring may be added.
  • a transformant in which the host is yeast
  • Burkholder's minimal medium Bostian, KL, et al., Processings * of the National Academy of Sciences] Proc. Natl. Acad. Sci. USA, 77, 4505 (1980)] and SD medium containing 0.5% casamino acid, CBiUer, GA et al. -The National Academy of Sciences of the USA (Proc. Natl. Acad. Sci. USA), 81, 5330 (1984)].
  • the pH of the medium is preferably adjusted to about 5-8.
  • the cultivation is usually performed at about 20 t to 35 ° C for about 24 to 72 hours, and aeration and agitation are added as necessary.
  • the culture medium used was a 10% strain immobilized in Grace's Insect Medium (Grace, TC, Nature, 195, 788 (1962)). Those to which additives such as serum are appropriately added are used. It is preferable to adjust the ⁇ ⁇ of the culture medium to about 6.2 to 6.4. Culture is usually performed at about 27 ° C for about 3 to 5 days, and aeration and agitation are added as necessary.
  • examples of the medium include a MEM medium containing about 5 to 20% fetal bovine serum [Science, 122, 501 (1952)], a DMEM medium [Virology, 8, 396 (1959)], RPM I 1640 medium [The Journal of the American Medical Association at ion, 199, 519 (1967) )], And 199 medium [Proceding of the Society for the Biological Medicine, Vol. 73, 1 (1950)].
  • the pH is about 6-8.
  • Cultivation is usually carried out at about 30 to 40 ° C for about 15 to 60 hours, and aeration and agitation are added as necessary.
  • the protein of the present invention can be produced in the cytoplasm or extracellularly 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
  • the buffer may contain a protein denaturant such as urea or guanidine hydrochloride, or a surfactant such as Triton X-100 TM .
  • a protein denaturant such as urea or guanidine hydrochloride
  • a surfactant such as Triton X-100 TM .
  • Purification of the protein contained in the culture supernatant or extract obtained in this manner can be performed by appropriately combining known separation and purification methods.
  • These known separation and purification methods include methods utilizing solubility such as salting-out and solvent precipitation, dialysis, ultrafiltration, gel filtration, and SDS-polyacrylamide gel electrophoresis, mainly molecular weight.
  • Method using difference in charge such as ion exchange chromatography, method using specific affinity such as affinity chromatography, hydrophobicity such as reversed-phase high-performance liquid chromatography, etc.
  • a method using a difference in gender, a method using a difference in isoelectric point such as isoelectric focusing, and the like are used.
  • the protein thus obtained When the protein thus obtained is obtained as a free form, it can be converted to a salt by a known method or a method analogous thereto, and conversely, when the protein is obtained as a salt, a known method or analogous method Depending on the method, it can be converted into a free form or another salt.
  • the protein produced by the recombinant can be arbitrarily modified or the polypeptide can be partially removed by the action of an appropriate protein-modifying enzyme before or after purification.
  • an appropriate protein-modifying enzyme for example, trypsin, chymotrypsin, arginyl endopeptidase, protein kinase, glycosidase and the like are used.
  • the presence of the protein of the present invention thus produced is determined using a specific antibody.
  • Antibodies against the protein or partial peptide of the present invention or a salt thereof are described in Any polyclonal antibody or monoclonal antibody may be used as long as it can recognize the protein or partial peptide or its salt.
  • an antibody against the protein or partial peptide of the present invention or a salt thereof (hereinafter, these may be simply abbreviated to the protein of the present invention in the description of the antibody) is obtained by using the protein of the present invention as an antigen.
  • the antibody or antiserum can be produced according to the following method.
  • the protein of the present invention is administered to a warm-blooded animal at a site where the antibody can be produced by administration to itself or together with a carrier or a diluent.
  • Complete Freund's adjuvant or incomplete Freund's adjuvant may be administered in order to enhance the antibody-producing ability upon administration. Administration is usually performed once every 2 to 6 weeks, for a total of 2 to 10 times.
  • Examples of the warm-blooded animal to be used include monkeys, egrets, dogs, guinea pigs, mice, rats, sheep, goats, and chickens, and mice and rats are preferably used.
  • a warm-blooded animal immunized with an antigen for example, an individual with an antibody titer is selected from a mouse, and the spleen or lymph node is collected 2 to 5 days after the final immunization and contained in them.
  • an individual with an antibody titer is selected from a mouse, and the spleen or lymph node is collected 2 to 5 days after the final immunization and contained in them.
  • a monoclonal antibody-producing hybridoma can be prepared.
  • the antibody titer in the antiserum can be measured, for example, by reacting the labeled protein described below with the antiserum, and then measuring the activity of the labeling agent bound to the antibody.
  • the fusion operation can be performed according to a known method, for example, the method of Kellar and Milstein (Nature, 256, 495 (1975)).
  • the fusion promoter include polyethylene glycol (PEG) and Sendai virus, but PEG is preferably used.
  • PEG polyethylene glycol
  • myeloma cells include myeloma cells of warm-blooded animals such as NS-1, P3U1, SP 2/0, and AP-1, but P3U1 is preferably used.
  • the preferred ratio between the number of antibody-producing cells (spleen cells) used and the number of myeloma cells used is about 1: 1 to 20: 1, and PEG (preferably PEG 1000 to PEG6000) Is added at a concentration of about 10 to 80%, and incubating at 20 to 40 ° (preferably at 30 to 37 for 1 to 10 minutes) enables efficient cell fusion.
  • PEG preferably PEG 1000 to PEG6000
  • Various methods can be used to screen for produced hybridomas. For example, the hybridoma culture supernatant is added to a solid phase (eg, a microplate) on which protein antigens are directly or adsorbed together with a carrier, and then radioactive substances are added.
  • Method to detect monoclonal antibody bound to solid phase by adding anti-immunoglobulin antibody (anti-mouse immunoglobulin antibody is used if the cell used for cell fusion is mouse) or protein A labeled with proteins or enzymes Then, add the hybridoma culture supernatant to the solid phase to which the anti-immunoglobulin antibody or protein A is adsorbed, and use radioactive substances, enzymes, etc.
  • a method of detecting a monoclonal antibody bound to a solid phase by adding a labeled protein may be used.
  • the selection of the monoclonal antibody can be performed according to a known method or a method analogous thereto. Usually, it can be performed in an animal cell culture medium supplemented with HAT (hypoxanthine, aminopterin, thymidine).
  • HAT hyperxanthine, aminopterin, thymidine
  • any medium can be used as long as it can grow a hybridoma.
  • RPMI 1640 medium containing 1 to 20%, preferably 10 to 20% fetal bovine serum, GIT medium containing 1 to 10% fetal bovine serum (Wako Pure Chemical Industries, Ltd.)
  • a serum-free medium for hybridoma culture SFM-101, Nissui Pharmaceutical Co., Ltd.
  • the culture temperature is usually from 20 to 40 ° C, preferably about 37 ° C.
  • the culture time is generally 5 days to 3 weeks, preferably 1 week to 2 weeks.
  • the culture can be usually performed under 5% carbon dioxide gas.
  • the antibody titer of the hybridoma culture supernatant can be measured in the same manner as the measurement of the antibody titer in the antiserum described above.
  • Monoclonal antibodies can be separated and purified by known methods, for example, immunoglobulin separation and purification methods [eg, salting out, alcohol precipitation, isoelectric point precipitation, electrophoresis, ion exchangers (eg, DEAE) ), Ultracentrifugation, gel filtration, antigen-binding solid phase or specific antibody purification by collecting only the antibody with an active adsorbent such as protein A or protein G, and dissociating the bond to obtain the antibody). It can be carried out (Preparation of polyclonal antibody)
  • the polyclonal antibody of the present invention can be produced according to a known method or a method analogous thereto. For example, a complex of an immunizing antigen (an antigen such as the protein of the present invention) and 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.
  • the antibody can be produced by collecting an antibody-containing substance against the antibody and separating and purifying the antibody.
  • the type of carrier protein and the mixing ratio of carrier and hapten are determined by the antibody against hapten immunized by cross-linking with the carrier. If it can be efficiently performed, any kind may be crosslinked at any ratio.
  • any kind may be crosslinked at any ratio.
  • serum serum albumin, thyroglobulin, hemocyanin, etc. may be used in a weight ratio of about 0 to 1 for hapten.
  • a method of condensing at a rate of 1 to 20, preferably about 1 to 5 is used. Further, various condensing agents can be used for force coupling between the hapten and the carrier.
  • an active ester reagent containing a daltaraldehyde, a carbodiimide, a maleimide active ester, a thiol group or a dithioviridyl group is used.
  • the condensation product is administered to a warm-blooded animal itself or together with a carrier or diluent at a site where antibody production is possible.
  • Complete Freund's adjuvant or incomplete Freund's adjuvant may be administered in order to enhance the antibody-producing ability upon administration. The administration is usually made once every about 2 to 6 weeks, for a total of about 3 to 10 times.
  • the polyclonal antibody can be collected from the blood, ascites, etc., preferably from the blood of a warm-blooded animal immunized by the above method.
  • the measurement of the polyclonal antibody titer in the antiserum can be performed in the same manner as the measurement of the antibody titer in the antiserum described above. Separation and purification of the polyclonal antibody can be carried out according to the same immunoglobulin separation and purification method as the above-mentioned separation and purification of the monoclonal antibody.
  • DNAs encoding the protein or partial peptide of the present invention are abbreviated as DNAs of the present invention in the description of antisense nucleotides
  • Antisense nucleotides having a base sequence or a part thereof are complementary or substantially complementary to the DNA of the present invention. Any antisense nucleotide may be used as long as it has a complementary base sequence or a part thereof and has an action capable 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 partial base 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 nucleotide sequence having a homology of about 97% or more, preferably about 98% or more, and more preferably about 99% or more with the sequence is exemplified.
  • the complementary sequence of the base sequence for example, the base sequence near the start codon
  • Antisense nucleotides having a homology of 7% or more, preferably about 98% or more, more preferably about 99% or more are suitable.
  • the antisense nucleotide is usually composed of about 10 to 40 bases, preferably about 15 to 30 bases.
  • the phosphate residues (phosphates) of each nucleotide constituting the antisense nucleotide are, for example, chemically modified phosphates such as phosphorothioate, methylphosphonate, and phosphorodithionate. It may be substituted with a residue.
  • These antisense nucleotides can be produced using a known DNA synthesizer or the like.
  • an antisense polynucleotide capable of inhibiting the replication or expression of the protein gene of the present invention is cloned or a DNA encoding a determined protein is obtained. It can be designed and synthesized on the basis of the base sequence information.
  • a polynucleotide can hybridize with the RNA of the protein gene of the present invention, inhibit the synthesis or function of the RNA, or inhibit the interaction with the protein-related RNA of the present invention.
  • the expression of the protein gene of the present invention can be regulated and controlled.
  • Polynucleotides that are complementary to a selected sequence of a protein-associated RNA of the present invention, and polynucleotides that can specifically hybridize with a protein-associated RNA of the present invention can be used in vivo and in vitro. It is useful for regulating and controlling the expression of a protein gene in a plant, and is also useful for treating or diagnosing a disease or the like.
  • Term "corresponding The term “is” means having homology or being complementary to a specific sequence of nucleotides, base sequences or nucleic acids including genes.
  • nucleotide, base sequence or nucleic acid and a peptide (protein) usually refers to the amino acid of the peptide (protein) under instructions derived from the nucleotide (nucleic acid) sequence or its complement. ing. 5 'end hairpin loop of protein gene, 5' end 6-base spare repeat, 5 'end untranslated region, polypeptide translation start codon, protein code region, ORF translation stop codon, 3' end untranslated The region, the 3 'end parindrome region, and the 3' end hairpin loop may be selected as preferred regions of interest, but any region within the protein gene may be selected.
  • the relationship between the nucleic acid of interest and a polynucleotide complementary to at least a part of the target region can be said to be "antisense" with the polynucleotide capable of hybridizing with the target.
  • Antisense polynucleotides include polynucleotides containing 2-dexoxy D-reports, polynucleotides containing D-reports, other types of polynucleotides that are N-daricosides of purine or pyrimidine bases, Alternatively, other polymers having non-nucleotide backbones (eg, commercially available protein nucleic acids and synthetic sequence-specific nucleic acid polymers) or other polymers containing special bonds ( ⁇ , such polymers may be found in DNA or RNA) (Including a nucleotide having a configuration that allows base pairing and base attachment as found)).
  • RNA hybrids can be double-stranded DNA, single-stranded DNA, double-stranded RNA, single-stranded RNA, and even DNA: RNA hybrids, and can be unmodified polynucleotides (or unmodified polynucleotides). (Nucleotides), and also those with known modifications, such as those with labels, caps, methylated, and one or more natural nucleotides known in the art.
  • Substituted, or modified with an intramolecular nucleotide for example, having an uncharged bond (eg, methylphosphonate, phosphotriester, phosphoramidate, olebamate, etc.), a charged bond or Those having a sulfur-containing bond (eg, phosphorothioate, phosphorodithioate, etc.), such as proteins (nucleases, nucleases / inhibitors) , Toxins, antibodies, signal peptides, poly -L one lysine, etc.) or a sugar (e.g., Mo No.3, etc., which have side chain groups, etc.
  • an uncharged bond eg, methylphosphonate, phosphotriester, phosphoramidate, olebamate, etc.
  • Those having a sulfur-containing bond eg, phosphorothioate, phosphorodithioate, etc.
  • proteins nucleases, nucleases / inhibitors
  • nucleic acid of the type I may include not only those containing purine and pyrimidine bases but also those having other modified heterocyclic bases. Such modifications may include methylated purines and pyrimidines, acylated purines and pyrimidines, or other heterocycles.
  • Modified nucleotides and modified nucleotides may also be modified at the sugar moiety, e.g., where one or more hydroxyl groups have been replaced with halogens, aliphatic groups, etc., or functionalities such as ethers, amines, etc. It may be converted to a group.
  • the antisense polynucleotide (nucleic acid) of the present invention is an RNA, a DNA, or a modified nucleic acid (RNA, DNA).
  • modified nucleic acid include, but are not limited to, sulfur derivatives of nucleic acids, thiophosphate derivatives, and those resistant to degradation of polynucleoside amides and oligonucleoside amides.
  • the antisense nucleic acid of the present invention can be preferably designed according to the following policy. That is, to make the antisense nucleic acid more stable in the cell, to increase the cell permeability of the antisense nucleic acid, to have a greater affinity for the target sense strand, and to be more toxic if it is toxic. Make sense nucleic acid less toxic.
  • the antisense nucleic acids of the present invention may contain altered or modified sugars, bases, or bonds, may be provided in special forms such as ribosomes or microspheres, may be applied by gene therapy, It could be given in additional form. In this way, the charge in the phosphate skeleton is used in the additional form.
  • Polycationic substances such as polylysine, which acts to neutralize, and crude water-based substances, such as lipids (eg, phospholipids, cholesterol, etc.), which enhance the interaction with cell membranes and increase the uptake of nucleic acids.
  • Preferred lipids for addition include cholesterol and its derivatives (eg, cholesteryl chromate formate, cholic acid, etc.).
  • nucleic acid can be attached via a base, sugar, or intramolecular nucleoside bond.
  • Other groups include cap groups specifically located at the 3 'or 5' end of nucleic acids that prevent degradation by nucleases such as exonucleases and RNases. .
  • capping groups include, but are not limited to, hydroxyl-protecting groups known in the art, such as dalicol such as polyethylene glycol and tetraethylene dalicol.
  • the inhibitory activity of the antisense nucleic acid can be examined using the transformant of the present invention, the in vivo or in vitro gene expression system of the present invention, or the in vivo or in vitro translation system of the protein of the present invention.
  • the nucleic acid can be applied to cells by various known methods.
  • the protein or partial peptide used in the present invention or a salt thereof (hereinafter, sometimes abbreviated as the protein of the present invention), the DNA encoding the protein or the partial peptide of the present invention (hereinafter, 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), and an antisense nucleotide of the DNA of the present invention (hereinafter, the present invention) May be abbreviated as “antisense nucleotide”).
  • the expression of the protein of the present invention increases in the heart at the stage of heart failure transition (heart failure decompensation / heart failure decompensation) after myocardial infarction, it can be used as a disease marker. That is, early diagnosis of diseases characterized by decreased cardiac function (eg, heart failure after myocardial infarction; angina pectoris; cardiomyopathy; heart diseases such as heart failure resulting from diseases such as angina pectoris and cardiomyopathy). It is useful as a marker for determining the severity of symptoms and predicting disease progression.
  • diseases characterized by decreased cardiac function eg, heart failure after myocardial infarction; angina pectoris; cardiomyopathy; heart diseases such as heart failure resulting from diseases such as angina pectoris and cardiomyopathy. It is useful as a marker for determining the severity of symptoms and predicting disease progression.
  • Antisense nucleotide of a gene encoding the protein of the present invention Pharmaceuticals containing a compound that regulates the activity of a protein or a salt thereof or an antibody against the protein of the present invention include, for example, diseases characterized by decreased cardiac function (eg, heart failure after myocardial infarction; angina pectoris; cardiomyopathy) Useful in the treatment and prevention of heart diseases such as heart failure due to diseases such as angina pectoris and cardiomyopathy).
  • diseases characterized by decreased cardiac function eg, heart failure after myocardial infarction; angina pectoris; cardiomyopathy
  • Hearts such as diseases characterized by reduced cardiac function (eg, heart failure after myocardial infarction; angina pectoris; cardiomyopathy; heart diseases such as heart failure resulting from diseases such as angina pectoris and cardiomyopathy) It can be used as a treatment and prevention drug for diseases.
  • the protein of the present invention is useful as a reagent for screening a compound or its salt that regulates the activity of the protein of the present invention.
  • a method for screening a compound or a salt thereof that regulates the activity of the protein of the present invention, characterized by using the protein of the present invention, is provided.
  • a cell capable of producing the protein of the present invention is preferably tested under the condition of low oxygen conditions when stimulated with extension, and (ii) A cell capable of producing the protein of the present invention is tested. It is intended to provide a method for screening a compound or a salt thereof that regulates the activity of the protein of the present invention, which is characterized by comparing a mixture of compounds with a case where a stretching stimulus is applied, preferably under hypoxic conditions.
  • the above-mentioned screening method is characterized in that, for example, in (i) and (ii), the gene expression levels of the protein of the present invention are measured and compared.
  • the activity of the protein of the present invention includes, for example, an activity of promoting cardiac function decline accompanying decompensation failure, an effect of suppressing an excessive compensation mechanism, and the like.
  • the low-oxygen conditions e.g. 2 0% 0 2 or less of oxygen concentration, for example, 2% (Neichiya primary, third 9 Volume 4, 4 8 5 page one 4 9 0 p., 1 9 9 8 years) Means the following conditions.
  • Stretch stimulation means that cardiomyocytes are cultured on a stretchable silicon membrane, It is a stimulus that applies a mechanical load by pulling the membrane (JBC, '271, 33 592-33597, 1996, Saccuration, 89, 2204-2211, 1994) , JBC, 271: 3221-3228, 1996).
  • Stretch stimulation means that cardiomyocytes are cultured on a stretchable silicon membrane, It is a stimulus that applies a mechanical load by pulling the membrane (JBC, '271, 33 592-33597, 1996, Saccuration, 89, 2204-2211, 1994) , JBC, 271: 3221-3228, 1996).
  • JBC JBC, '271, 33 592-33597, 1996, Saccur
  • the cytoprotective action in the cases (iii) and (iv) and the expression level of the gene encoding the protein of the present invention are measured by a known method and compared.
  • Cytoprotective action can be shown by cardiomyocyte activation or viability.
  • MTT 3- (4,5-Dimethyl-2-t iazolyl) -2,5-di henyl -2H-tetrazol iuni) method, which can measure commonly used respiratory activity, and trypan-bleed staining It can be measured by the TUNNEL staining method (Terminal deoxytransferase-mediated d UTP-X nick end labeling, Cell, Vol. 97, pp. 189-198, 1999).
  • Appropriate expression enhancement at the time of cell death or cell damage can be expected to have a cytoprotective effect.
  • overexpression of the gene encoding the protein of the present invention is considered to cause excessive activation of cells and accelerate cell exhaustion. Therefore, it is considered important to appropriately control the expression level of the gene encoding the protein of the present invention. It is thought that cell damage is caused by promotion of expression of the gene encoding the protein of the present invention, for example, during the heart failure decompensation period and the heart failure decompensation period.
  • an inhibitor a compound that inhibits the activity of the protein of the present invention or a salt thereof
  • an enhancer a compound that promotes the activity of the protein of the present invention or a salt thereof
  • the DNA (gene) encoding the protein of the present invention has a function of positively regulating the expression of a heart-specific gene, extreme decrease in the expression of the gene encoding the protein of the present invention impairs cell function. Damage and, conversely, overexpression may cause more activation of cells than necessary, resulting in cell damage.
  • the function of the protein of the present invention, the DNA of the present invention, or the function of the protein of the present invention obtained by the above-described screening method is considered.
  • a compound or a salt thereof which will be specifically described later
  • the disorder of the cell function can be prevented or treated, and conversely, an excess of the protein of the present invention or the DNA of the present invention can be obtained.
  • administration of a compound that inhibits the function of the protein of the present invention obtained by the above-described screening method or a salt thereof will increase the cell function. Disability can be prevented and treated.
  • a gene whose expression is positively regulated by the protein of the present invention e.g., atrial sodium diuretic peptide, cardiac adoriamycinresvonshib 'protein (CARP), myosin light chain 2V (MLC 2v), Nkx 2.5, GAT A-4, MEF2 C, N-myc, HAND-1, Ventricular natriuretic peptide (BBR, Vol. 270, pp. 1074-1079, 2000; Development, Vol. 126, 1269-1280 , 1999)), a compound that regulates the activity of the protein of the present invention, characterized by measuring the enzymatic activity of a reporter gene in a reporter gene using a promoter such as It also provides a method for screening the salt.
  • the protein of the present invention e.g., atrial sodium diuretic peptide, cardiac adoriamycinresvonshib 'protein (CARP), myosin light chain 2V (MLC 2v), Nkx 2.5, GAT A
  • the reporter gene assay is, for example, a promoter region of a gene whose expression is positively regulated by the protein of the present invention [eg, the promoter region of the atrial natriuretic peptide gene (JBC, Vol. 272, 22800). -22808, 1997; Development, Vol. 124, pp.
  • plasmid containing a DNA encoding the protein of the present invention
  • a method for producing a transformant transformed with the recombinant vector It can be performed according to a similar method.
  • the compound or salt thereof obtained by the screening method is a substance that affects the expression level and activation of DNA (gene) encoding the protein of the present invention. Any of a compound that promotes the activity of the protein and a compound that inhibits the activity of the protein of the present invention can be selected.
  • Test compounds include, for example, peptides, proteins, non-peptidic compounds derived from living organisms (such as carbohydrates and lipids), synthetic compounds, microbial cultures, cell extracts, and plant extracts Liquid, animal tissue extract and the like. These compounds may be novel compounds or known compounds.
  • cells having the ability to produce the protein of the present invention are cultured in a medium suitable for screening.
  • the medium may be any as long as it does not affect the gene expression of the protein of the present invention.
  • Examples of cells having the ability to produce the protein of the present invention include, for example, primary cardiomyocytes having the ability to produce the protein of the present invention or vectors containing DNA encoding the protein of the present invention described above.
  • a host (transformant) transformed with is used.
  • the host for example, animal cells such as H9c2 cells are preferably used.
  • a transformant in which the protein of the present invention is expressed in a cell by culturing by the method described above is preferably used.
  • the expression level of the gene of the present invention can be determined by a known method, for example, Northern blotting, Reverse transcript ion-polymerase chain react ion (RT-PCR), or a real-time PCR analysis system (Ad, TadMan polymerase chain react ion).
  • RT-PCR Reverse transcript ion-polymerase chain react ion
  • Ad TadMan polymerase chain react ion
  • the gene expression level in the case (ii) is inhibited or promoted by about 20% or more, preferably 30% or more, more preferably about 50% or more, as compared with the case (i).
  • the test compound to be tested can be selected as a compound that inhibits or promotes the activity of the protein of the present invention.
  • the cytoprotective effect and the expression level of the gene encoding the protein of the present invention were measured, and the gene expression level was reduced, resulting in cell damage.
  • a compound that increases the expression level under the conditions is used as a compound that promotes the activity of the protein of the present invention.
  • a compound that reduces the expression level is used as a compound that inhibits the activity of the protein of the present invention, and can be used as a compound that has a cytoprotective action.
  • the enzyme activity of the reporter gene in the case of the above (V) is about 20% or more, preferably 30% or more, more preferably about 30% or more of that in the case of the above (iV).
  • a test compound that inhibits or promotes 50% or more can be selected as a compound that inhibits or promotes the activity of the protein of the present invention.
  • the compound or a salt thereof that inhibits the activity of the protein of the present invention selected by the above screening method is used during the heart failure decompensation phase and the decompensation phase in which the expression of the DNA (gene) encoding the protein of the present invention is enhanced.
  • the administration can be expected to have a cardiac function recovery effect.
  • the compound or salt thereof which promotes the activity of the protein of the present invention selected by the above screening method enhances the compensatory mechanism by being administered in the acute phase of heart failure where expression is reduced, thereby protecting cardiomyocytes. A heart protective effect can be expected.
  • the screening kit of the present invention contains the protein or partial peptide used in the present invention or a salt thereof, or a cell capable of producing the protein or partial peptide used in the present invention.
  • the compound (a compound that promotes or inhibits the activity of the protein of the present invention) or a salt thereof obtained by using the screening method or the screening kit of the present invention may be a test compound as described above, for example, a peptide, a protein, a non-peptide derived from a living body.
  • Compounds eg, carbohydrates, lipids, etc.
  • synthetic compounds, compounds or salts thereof selected from microbial cultures, fermentation products, cell extracts, plant extracts, animal tissue extracts, plasma, etc.
  • a compound or a salt thereof that regulates (promotes or inhibits) the activity of the protein of the present invention eg, activity to lower cardiac function).
  • salt of the compound those similar to the aforementioned salts of the protein of the present invention are used.
  • Compounds or salts thereof that regulate (promote or inhibit) the activity of the protein of the present invention include, for example, diseases characterized by reduced cardiac function (eg, heart failure after myocardial infarction; angina pectoris; cardiomyopathy; angina pectoris) And heart diseases such as heart failure caused by diseases such as cardiomyopathy).
  • diseases characterized by reduced cardiac function eg, heart failure after myocardial infarction; angina pectoris; cardiomyopathy; angina pectoris
  • heart diseases such as heart failure caused by diseases such as cardiomyopathy.
  • a compound or a salt thereof obtained by using the screening method or the screening kit of the present invention is used as the above-mentioned therapeutic or prophylactic agent, it can be formulated according to a conventional method.
  • tablets, capsules, elixirs, micron capsules, sterile solutions, suspensions and the like can be used.
  • the preparations obtained in this way are safe and have low toxicity, for example, warm-blooded animals (for example, humans, mice, rats, puppies, sheep, bush, puppies, puppies, birds, cats, dogs) , Monkeys, chimpanzees, etc.) orally or parenterally.
  • the dose of the compound or a salt thereof varies depending on its action, target disease, subject of administration, route of administration, and the like.
  • a compound or a salt thereof that regulates the activity of the protein of the present invention for the purpose of treating heart failure When the compound is orally administered, generally in an adult (assuming a body weight of 60 kg), the compound or a salt thereof is used in an amount of about 0.1 to 10 Omg, preferably about 1.0 to 50 mg, more preferably about 0.1 to 50 mg per day. 1.
  • the single dose of the compound or a salt thereof varies depending on the administration subject, target disease, and the like.
  • a compound or a compound thereof that regulates the activity of the protein of the present invention for the purpose of treating heart failure When the salt is administered to an adult (as 60 kg) usually in the form of an injection, the compound or a salt thereof is used in an amount of about 0.01 to 3 Omg per day, preferably about 0.1 to 2 Omg, more preferably about 0.1 to 2 Omg. It is convenient to administer about 0.1 to 1 Omg by intravenous injection. In the case of other animals, the amount converted per 60 kg can be administered.
  • 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, particularly It can be used for quantification by sandwich immunoassay.
  • 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 F (ab ') 2 , Fab', or Fab fraction of the antibody molecule may be used.
  • the method for quantifying the protein of the present invention using the antibody of the present invention is not particularly limited, and may be an antibody, an antigen, or an antibody-antigen complex corresponding to the amount of antigen (eg, the amount of protein) in the test solution. Any measurement method may be used as long as the amount of the body is detected by chemical or physical means, and this is calculated from a standard curve prepared using a standard solution containing a known amount of antigen. . For example, nephrometry, a competition method, an immunometric method and a sandwich method are suitably used, but it is particularly preferable to use a sandwich method described later in terms of sensitivity and specificity.
  • a labeling agent used in a measuring method using a labeling substance for example, a radioisotope, an enzyme, a fluorescent substance, a luminescent substance and the like are used. Radioisotopes, if example embodiment, [1 2 5 I], [1 3 1 I], [3 H], and [1 4 C] used.
  • the above-mentioned enzyme those which are stable and have a large specific activity are preferable. For example, i3-galactosidase,) 8-darcosidase, alkaline phosphatase, peroxidase, and lignoic acid dehydrogenase are used.
  • fluorescent substance for example, fluorescein, fluorescein isothiosinate and the like are used.
  • luminescent substance for example, luminol, luminol derivative, luciferin, lucigenin and the like are used.
  • a biotin-avidin system can be used for binding the antibody or antigen to the labeling agent.
  • Carriers include insoluble polysaccharides such as agarose, dextran, and cellulose; and synthetic resins such as polystyrene, polyacrylamide, and silicon. Or 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).
  • primary reaction the insolubilized monoclonal antibody of the present invention
  • secondary reaction another labeled monoclonal antibody of the present invention
  • the primary reaction and the secondary reaction may be performed in the reverse order, may be performed simultaneously, or may be performed at staggered times.
  • the labeling agent and the method of insolubilization can be in accordance with those described above.
  • the antibody used for the solid phase antibody or the labeling antibody does not necessarily need to be one kind, and a mixture of two or more kinds of antibodies is used for the purpose of improving measurement sensitivity and the like. May be used.
  • the monoclonal antibody of the present invention used in the primary reaction and the secondary reaction is an antibody having a different site to which the protein of the present invention binds. It is preferably used. That is, the antibody used in the primary reaction and the secondary reaction is, for example, when the antibody used in the secondary reaction recognizes the C-terminal of the protein of the present invention, the antibody used in the primary reaction is Preferably, an antibody that recognizes other than the C-terminal, for example, the N-terminal, is used.
  • the monoclonal antibody of the present invention can be used in a measurement system other than the sandwich method, for example, a competition method, an immunometric method, a nephelometry method, or the like.
  • a competition method an antigen in a test solution and a labeled antigen are applied to the antibody.
  • the unreacted labeled antigen (F) and the labeled antigen (B) bound to the antibody are separated (B / F separation), and the labeling amount of either B or F is measured.
  • the amount of antigen in the test solution is quantified.
  • a soluble antibody is used as an antibody
  • B / F separation is performed using a polyethylene glycol
  • a liquid phase method using a second antibody against the antibody or a solid phase antibody is used as the first antibody.
  • an immobilization method using a soluble first antibody and an immobilized antibody as the second antibody is used.
  • the antigen in the test solution and the immobilized antigen are subjected to a competitive reaction with a certain amount of labeled antibody, and then the solid phase and the liquid phase are separated. Reaction with an excess amount of the labeled antibody, and then adding immobilized antigen to the unreacted labeled antibody. After binding the body to the solid phase, the solid and liquid phases are separated. Next, the amount of label in either phase is measured to determine the amount of antigen in the test solution.
  • nephrometry the amount of insoluble sediment resulting from an antigen-antibody reaction in a gel or in a solution is measured. Even when the amount of antigen in the test solution is small and only a small amount of sediment is obtained, laser nephrometry utilizing laser scattering is preferably used.
  • the protein measurement system of the present invention may be constructed by adding ordinary technical considerations to those skilled in the art to the ordinary conditions and procedures in each method. For details of these general technical means, reference can be made to reviews and written documents.
  • the protein of the present invention can be quantified with high sensitivity by using the antibody of the present invention.
  • 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, a disease characterized by decreased cardiac function (Eg, heart failure after myocardial infarction; angina; cardiomyopathy; heart disease such as heart failure from diseases such as angina and cardiomyopathy) or will be affected in the future Can be diagnosed as having a high possibility.
  • a disease characterized by decreased cardiac function Eg, heart failure after myocardial infarction; angina; cardiomyopathy; heart disease such as heart failure from diseases such as angina and cardiomyopathy
  • the antibody of the present invention can be used for detecting the protein of the present invention present in a subject such as a body fluid or a tissue.
  • a subject such as a body fluid or a tissue.
  • the detection of the protein of the present invention in each fraction during purification, and the analysis of the behavior of the protein of the present invention in test cells, etc. Can be used.
  • the DNA of the present invention can be used, for example, as a probe to produce warm-blooded animals (eg, humans, rats, mice, guinea pigs, egrets, birds, higgies, bush, horses, dogs, cats, dogs, DNA or mRNA encoding the protein of the present invention or its partial peptide (monkey, chimpanzee, etc.) can be detected (gene abnormality).
  • warm-blooded animals eg, humans, rats, mice, guinea pigs, egrets, birds, higgies, bush, horses, dogs, cats, dogs, DNA or mRNA encoding the protein of the present invention or its partial peptide (monkey, chimpanzee, etc.) can be detected (gene abnormality).
  • the DNA or mRNA is damaged, mutated or expressed. It is useful as a gene diagnostic agent for a decrease, an increase in the DNA or mRNA, or an overexpression.
  • the above-described genetic diagnosis using the DNA of the present invention can be performed, for example, by the well-known Northern hybridization or PCR-SSCP method (Genomics, Vol. 5, p. 874-879 (1989). Proceedings of the National Academy of Sciences of the United States of America; Vol. 86, Vol. 27, Proceedings of the National Academy of Sciences of the United States of America; 66-27770 (11989)), etc.
  • Northern hybridization or DNA mutation by PCR-SSCP method Is detected, it is possible to diagnose that the disease is highly likely to be a disease such as a heart disease accompanied by a decrease in cardiac function, etc.
  • the antisense nucleotide of the present invention which complementarily binds to the DNA of the present invention and can suppress the expression of the DNA, has low toxicity, and is characterized by having the protein of the present invention or the DNA of the present invention in vivo in vivo. It can regulate (inhibit) activities and functions (eg, cardiac dysfunction promoting activity), for example, diseases characterized by reduced cardiac function (eg, heart failure after myocardial infarction; angina pectoris; myocardium Sickness; derived from diseases such as angina pectoris and cardiomyopathy It can be used as a treatment and prevention agent for heart diseases such as heart failure.
  • the above-mentioned antisense nucleotide is used as the above-mentioned therapeutic or prophylactic agent, it can be formulated and administered according to a known method.
  • the antisense nucleotide when used, the antisense nucleotide is inserted alone or into an appropriate vector such as a retrovirus vector, an adenovirus vector, an adenovirus associated virus vector, and the like.
  • an appropriate vector such as a retrovirus vector, an adenovirus vector, an adenovirus associated virus vector, and the like.
  • Oral or parenteral to warm-blooded animals e.g., humans, rats, mice, guinea pigs, egrets, birds, higgins, pigs, pigs, dogs, cats, dogs, monkeys, chimpanzees, etc.
  • the antisense nucleotide can be administered as it is or in the form of a formulation together with a physiologically acceptable carrier such as an adjuvant for promoting uptake, and then administered using a gene gun or a catheter such as a hydrogel catheter.
  • the dose of the antisense nucleotide varies depending on the disease to be treated, the subject to be administered, the administration route, and the like.
  • the antisense nucleotide of the present invention is orally administered for the purpose of treating heart failure, generally the adult (body weight) is used. At 6 O kg), about 0.1 to 10 O mg of the antisense nucleotide is administered per day.
  • the antisense nucleotide can also be used as a diagnostic oligonucleotide probe for examining the presence of the DNA of the present invention in tissues or cells and the state of its expression.
  • the present invention further provides
  • 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 polynucleotide, and can inhibit the expression of the peptide of the present invention in vivo. It can regulate (inhibit) the activity or function of the polynucleotide (eg, DNA) of the present invention (eg, the activity of promoting cardiac function decline), for example, a disease (eg, a disease characterized by decreased cardiac function). Heart failure after myocardial infarction All; angina; cardiomyopathy; heart diseases such as heart failure derived from diseases such as angina and cardiomyopathy).
  • Double-stranded RNA can be produced by designing based on the sequence of the polynucleotide of the present invention according to a known method (eg, Nature, 411, 494, 2001).
  • the lipozyme can be designed and manufactured based on the sequence of the polynucleotide of the present invention according to a known method (eg, TRENDS in Molecular Medicine, 7, 221, 2001). For example, it can be produced by linking a known lipozyme to a part of the RNA encoding the peptide of the present invention.
  • a part of the RNA encoding the peptide of the present invention includes a portion (RNA fragment) close to the cleavage site on the RNA of the present invention, which can be cleaved by a known lipozyme.
  • RNA or lipozyme When the above double-stranded RNA or lipozyme is used as the above-mentioned prophylactic / therapeutic agent, it can be formulated and administered in the same manner as an antisense polynucleotide.
  • the antibody of the present invention which has the activity of neutralizing the activity of the protein of the present invention, is used for a disease characterized by a decrease in cardiac function (eg, heart failure after myocardial infarction; angina; cardiomyopathy; angina, cardiac muscle) It can be used as a prophylactic or therapeutic agent for diseases such as heart failure caused by diseases such as sickness.
  • the agent for preventing or treating the above-mentioned diseases which contains the antibody of the present invention, has low toxicity, and can be used as it is as a liquid or as a pharmaceutical composition of an appropriate dosage form, as a warm-blooded animal (eg, human, rat, mouse, It can be administered orally or parenterally to guinea pigs, egrets, birds, sheep, pigs, puppies, pomas, cats, dogs, monkeys, chimpanzees, etc.
  • the dosage varies depending on the administration subject, target disease, symptoms, administration route and the like.For example, when used for the treatment or prevention of heart failure in adults, the antibody of the present invention is usually administered as a single dose.
  • the antibodies of the present invention can be administered by themselves or as a suitable pharmaceutical composition.
  • the pharmaceutical composition used for the administration contains the antibody or a salt thereof and a pharmacologically acceptable carrier, diluent or excipient.
  • Such compositions are provided in dosage forms suitable for oral or parenteral administration.
  • compositions for oral administration include solid or liquid dosage forms, specifically tablets (including sugar-coated tablets and film-coated tablets), pills, granules, powders, capsules (soft capsules and the like). ), Syrups, emulsions, suspensions and the like.
  • Such a composition is produced by a known method and contains a carrier, diluent or excipient commonly used in the field of pharmaceuticals. For example, lactose, starch, sucrose, magnesium stearate and the like are used as carriers and excipients for tablets.
  • 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, drip injections, etc. Is included.
  • Such injections are prepared according to known methods, for example, by dissolving, suspending or emulsifying the antibody or a salt thereof in a sterile aqueous or oily liquid commonly used for injections.
  • aqueous solution for injection for example, physiological saline, isotonic solution containing glucose and other adjuvants and the like are used, and a suitable solubilizing agent, for example, alcohol (eg, ethanol), polyalcohol (eg, Propylene glycol, polyethylene glycol), non-ionic surfactants (eg, polysorbate 80, HC-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)), etc. Good.
  • a suitable solubilizing agent for example, alcohol (eg, ethanol), polyalcohol (eg, Propylene glycol, polyethylene glycol), non-ionic surfactants (eg, polysorbate 80, HC-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)), etc.
  • 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 so
  • the above-mentioned oral or parenteral pharmaceutical composition is conveniently prepared in a unit dosage form adapted to the dose of the active ingredient.
  • dosage unit forms include tablets, pills, capsules, injections (ampoules), and suppositories.
  • Each dosage unit usually contains 5 to 500 mg, especially 5 to 100 mg for injections, and 10 to 25 mg for other dosage forms. preferable.
  • compositions may contain another active ingredient as long as the composition does not cause an undesirable interaction with the above-mentioned antibody.
  • the protein of the present invention is used as a drug such as a vaccine for producing the antibody of the present invention.
  • a vaccine for producing the antibody of the present invention can be produced by a known method using the protein of the present invention.
  • Recombinant N obtained by linking a nuclear localization signal sequence (antenapedia: a nuclear localization peptide of Drosophi la, a report by Ito et al. Of Tokyo Medical and Dental University: 2nd Molecular Cardiovascular Conference) to the protein of the present invention.
  • the reduced protein of the present invention can be supplemented by administering kx2.5 systemically or directly to the heart during a cardiac catheterization test.
  • the medicament containing DNA of the present invention is used for gene therapy of heart failure.
  • One of the functions of the protein of the present invention is to enhance the expression of heart-specific genes. We believe that this will result in enhanced compensatory mechanisms following myocardial infarction. Therefore, during the period when the expression of the protein of the present invention is decreased, such as in the acute phase of heart failure, the DNA of the present invention can be administered so that the protein of the present invention maintains an appropriate expression level.
  • transgenic animals expressing the protein of the present invention can be prepared.
  • animals include mammals (for example, rats, mice, rabbits, rabbits, sheep, pigeons, pigs, cats, dogs, monkeys, etc.) (hereinafter sometimes abbreviated as animals).
  • mammals for example, rats, mice, rabbits, rabbits, sheep, pigeons, pigs, cats, dogs, monkeys, etc.
  • animals include mammals (for example, rats, mice, rabbits, rabbits, sheep, pigeons, pigs, cats, dogs, monkeys, etc.) (hereinafter sometimes abbreviated as animals).
  • the DNA is used as a gene construct linked downstream of a promoter capable of being expressed in animal cells. Is generally advantageous.
  • a gene construct in which the DNA of the present invention derived from an animal having a high homology with the DNA is linked to a downstream of various promoters capable of expressing the same in animal cells is used, for example.
  • a DNA-introduced animal of the present invention that produces high levels of protein can be produced.
  • a ubiquitous expression promoter such as a virus-derived promoter or meta-mouth thionein may be used, but preferably, an NGF gene promoter, a genolases gene promoter, etc., which are specifically expressed in the brain. Is used.
  • 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 animal.
  • the presence of the protein of the present invention in the germinal cells of the animal after the introduction of DNA means that all the offspring of the animal have the protein of the present invention in all of the germinal and somatic cells.
  • the progeny of such animals that have inherited the gene will have the protein of the invention in all of their germinal and somatic cells.
  • the animal having the DNA can be reared in an ordinary breeding environment as the DNA-bearing animal. Furthermore, by crossing male and female animals having the target DNA, homozygous animals having the transgene on both homologous chromosomes are obtained, and by crossing the male and female animals, all the offspring will have the DNA Breeding to have Since the protein of the present invention is highly expressed in the animal into which the DNA of the present invention has been introduced, it is useful as an agonist or an animal for screening an gonist against the protein of the present invention.
  • the DNA-introduced animal of the present invention can also be used as a cell source for tissue culture.
  • tissue culture for example, by directly analyzing DNA or RNA in the tissue of the DNA-introduced mouse of the present invention, or by analyzing the tissue in which the receptor protein of the present invention expressed by a gene is present, the present invention can be used. Analyze protein.
  • the cells of a tissue having the protein of the present invention can be cultured by standard tissue culture techniques, and these can be used to study the function of cells from generally difficult-to-cultivate tissues such as those derived from brain and peripheral tissues. . Also, use the cells Thus, for example, it is possible to select a medicine that enhances the function of various tissues. Further, if there is a high expression cell line, the protein of the present invention can be isolated and purified therefrom.
  • a test compound is administered to the DNA-introduced animal of the present invention, and the heart function, electrocardiogram, heart weight, and the like of the animal are measured.
  • Heart weight is the parameter of hypertrophy. Specifically, the heart structure can be examined by calculating the heart weight per body weight, the left ventricular weight per body weight, and the left ventricular weight per right ventricle weight. Since the above parameters increase when cardiac hypertrophy occurs, test compounds can be evaluated using the suppression of this increase as an index. After administering a test compound to the DNA-introduced animal of the present invention, a myocardial infarction is performed, and the heart function, electrocardiogram, heart weight, and the like of the animal are measured.
  • the infarction progress-inhibiting activity of the test compound can be examined by weighing the infarct layer.
  • Administration of the test compound may be after infarction surgery.
  • the animal can be crossed with a genetic hypertension model rat such as an SHR rat to create a new heart failure model. The compound is administered to the cardiac failure model thus prepared, and the animal is examined for cardiac function, electrocardiogram, cardiac weight, activity for suppressing infarction progression, and the like.
  • the present invention provides a non-human mammalian embryonic stem cell in which the DNA of the present invention is 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 3-galactosidase gene derived from Escherichia coli), and the reporter gene is transformed into the DN of the present invention.
  • a reporter gene eg, a 3-galactosidase gene derived from Escherichia coli
  • the non-human mammal according to the above (6) which can be expressed under the control of a promoter for A.
  • non-human mammalian embryonic stem cells in which the DNA of the present invention has been inactivated are artificially mutated to the DNA of the present invention possessed by the non-human mammal to suppress the DNA expression ability, Alternatively, by substantially eliminating the activity of the protein of the present invention encoded by the DNA, the DNA does not substantially have the ability to express the protein of the present invention (hereinafter referred to as the knockout DNA of the present invention).
  • Non-human mammalian embryonic stem cells hereinafter abbreviated as ES cells).
  • non-human mammal those similar to the above can be 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 produced by, for example, shifting the codon reading frame or disrupting the function of the promoter or exon by these mutations.
  • non-human mammalian embryonic stem cells of the present invention in which DNA is inactivated include, for example, The non-human mammal DNA of the present invention is isolated and its exon portion is a drug resistance gene typified by a neomycin resistance gene, a hygromycin resistance gene, or lacZ (13-galactosidase gene), A DNA sequence that disrupts exon function by inserting a reporter gene, such as cat (chloramphenicylacetyltransferase gene), or terminates gene transcription in the intron portion between exons (for example, po 1 gene, resulting in disruption of gene synthesis by preventing the synthesis of complete mRNA
  • a targeting vector A DNA strand that was constructed so that A DNA strand (hereinafter, abbreviated as a targeting vector) is introduced into the chromosome of the animal by, for example, a
  • ES cells from which the DNA of the present invention is inactivated by the homologous recombination method or the like for example, those already established as described above may be used, or the known methods of Evans and Kaufman may be used. It may be newly established according to. For example, in the case of mouse ES cells, currently, 129 ES cells are generally used, but since the immunological background is not clear, a pure line that substitutes them can be used for immunological inheritance.
  • BDFi mice for the purpose of obtaining ES cells with a clear background, BDFi mice (C57BLZ6 and DBAZ2 and C57BLZ6) BDFi mice can be used satisfactorily.Because BDFi mice have a high number of eggs collected and their eggs are durable, they have C57BLZ6 mice as their background.
  • the obtained ES cells can be advantageously used when a pathological model mouse is created, since the genetic background can be replaced by C57BL / 6 mice by backcrossing with C57BLZ6 mice.
  • blastocysts 3.5 days after fertilization are generally used. Early embryos can be obtained.
  • male ES cells are generally more convenient for producing a germline chimera. 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. Using this method, it has traditionally required about 10 6 cell numbers to perform a karyotype analysis. On the other hand, the number of ES cells in one colony (approximately 50) is sufficient, so the primary selection of ES cells in the early stage of culture can be performed by gender discrimination. By making selection possible, labor in the initial stage of culture can be significantly reduced.
  • Embryonic stem cell lines obtained in this way usually have very good proliferative properties, but they must be carefully subcultured because they tend to lose their ability to generate individuals.
  • a suitable feeder cell such as STO fibroblasts
  • a carbon dioxide incubator preferably 5% carbon dioxide, 95% air or 5%
  • LIF 1-1000 OU / ml
  • trypsin / EDTA solution usually 0.001-0.5% trypsin / 0 1-5mM EDTA (preferably about 0.1% trypsin ZlmM EDTA) is used to convert the cells into single cells and seed them on freshly prepared feeder cells.
  • Such subculture is usually performed every 11 to 13 days. At this time, it is desirable to observe the cells, and if morphologically abnormal cells are found, discard the cultured cells.
  • ES cells can be cultured in monolayers at high densities or in suspension cultures to form cell clumps under appropriate conditions to produce various types of cells such as parietal, visceral, and cardiac muscles.
  • MJ Evans and MH Kaufman Nature 292, 154, 1981; GR Martin Proceedings 'Ob' National 'Academy of Science' Natl. Acad. Sci. USA, Vol. 78, p. 7634, 1981; TC Doetschman et al., Journal of Obelimborgologi 'and' Experimental Morphology, Vol. 87, 27. P., 1985]
  • the DNA-deficient cells of the present invention obtained by differentiating the ES cells of the present invention are cells of the protein of the present invention in the mouth of the intestine Useful in biological studies.
  • the non-human mammal deficient in DNA expression of the present invention can be distinguished from a normal animal by measuring the mRNA level of the animal using a known method and indirectly comparing the expression level. It is possible.
  • non-human mammal those similar to the aforementioned can be used.
  • the non-human mammal deficient in DNA expression of the present invention can be obtained, for example, by introducing the targeting vector prepared as described above into a mouse embryonic stem cell or a mouse egg cell, and introducing the DNA of the present invention into the evening targeting vector.
  • the inactivated DNA sequence can be knocked out by homologous recombination of the DNA of the present invention on the chromosome of mouse embryonic stem cells or mouse egg cells by homologous recombination. it can.
  • Cells in which the DNA of the present invention has been knocked out are combined with a DNA sequence on a Southern hybridization analysis or a targeting vector using a DNA sequence on or near the DNA of the present invention as a probe, and a targeting vector.
  • the determination can be made by PCR analysis using the DNA sequence of the neighboring region other than the DNA of the present invention derived from the mouse used as the primer.
  • the cell line in which the DNA of the present invention has been inactivated is cloned by homologous gene recombination, and the cell line is cloned at an appropriate time, for example, at the 8-cell stage.
  • the chimeric embryo is injected into a human mammalian embryo or blastocyst, and the resulting chimeric embryo is transplanted into the uterus of the pseudo-pregnant non-human mammal.
  • the produced animal is a chimeric animal composed of both cells having the normal DNA locus of the present invention and cells having the artificially altered DNA locus of the present invention.
  • all tissues are artificially mutated from a population obtained by crossing such a chimeric individual with a normal individual. It can be obtained by selecting individuals composed of cells having the added DNA locus of the present invention, for example, by judging coat color or the like.
  • the individuals obtained in this manner are usually individuals with heterozygous expression of the protein of the present invention, which are mated with individuals with 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 in which the targeting vector has been introduced into the chromosome can be obtained by injecting a DNA solution into the chromosome of the present invention. It can be obtained by selecting those with mutations at the NA locus.
  • 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 a plurality of homozygous animals are obtained.
  • homozygous and heterozygous animals having the inactivated DNA are bred and subcultured.
  • non-human mammalian embryonic stem cells in which the DNA of the present invention has been inactivated are extremely useful for producing a non-human mammal deficient in expressing the DNA of the present invention.
  • the non-human mammal deficient in expression of the DNA of the present invention lacks various biological activities that can be induced by the protein of the present invention, a disease caused by inactivation of the biological activity of the protein of the present invention. It can be a model, so it is useful for studying the causes of these diseases and examining treatment methods.
  • the non-human mammal deficient in expression of the DNA of the present invention can be used for screening for a compound having a therapeutic / preventive effect against diseases caused by the deficiency or damage of the DNA of the present invention.
  • the present invention is characterized in that a test compound is administered to a non-human mammal deficient in expression of the DNA of the present invention, and changes in the animal are observed and measured.
  • a method for screening a compound or a salt thereof having a therapeutic or preventive effect on a disease caused by the disease includes 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.
  • 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 the organs, tissues, disease symptoms, etc. of the animal are used as indices.
  • the test compound can be tested for its therapeutic and prophylactic effects. .
  • test compound for example, oral administration, intravenous injection and the like are used, and it can be appropriately selected according to the symptoms of the test animal, the properties of the test compound, and the like.
  • the dose of the test compound can be appropriately selected according to the administration method, the properties of the test compound, and the like.
  • a test compound is administered to a non-human mammal deficient in the expression of DNA of the present invention, and the heart function, electrocardiogram, heart weight and the like of the animal are measured.
  • Heart weight is a parameter of cardiac hypertrophy.
  • the heart structure can be examined by calculating the heart weight per body weight, the left ventricular weight per body weight, and the left ventricular weight per right ventricle weight.
  • test compounds can be evaluated using the suppression of this increase as an index.
  • a myocardial infarction is performed, and the heart function, electrocardiogram, heart weight, and the like of the animal are measured.
  • the infarct growth inhibitory activity of the test compound can be examined by weighing the infarct layer.
  • Administration of the test compound may be post-infarct surgery.
  • a new heart failure model can be created by crossing the animal with a genetically high blood model rat such as an SHR rat.
  • the compound is administered to the heart failure model thus prepared, and the animal is examined for cardiac function, electrocardiogram, heart weight, infarction progress inhibitory activity and the like.
  • the compound obtained by using the screening method is a compound selected from the test compounds described above, and has a prophylactic / therapeutic effect against a disease caused by deficiency or damage of the protein of the present invention. It can be used as a safe and low toxic prophylactic and therapeutic agent. Further, a compound derived from the compound obtained by the above-mentioned screening can also be used.
  • 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, etc.).
  • Such salts include, for example, salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.), or organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid) Salts with acids, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc. are used.
  • inorganic acids eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.
  • organic acids eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid
  • Salts with acids 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, and can be used, for example, in humans or other mammals (eg, rats, mice, guinea pigs, egrets, higgs, bush, foxes, dogs, 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 is orally administered, it is generally used in adults (with a body weight of 6 kg).
  • the compound is administered at about 0.1 to 10 Omg, preferably about 1.0 to 5 Omg, more preferably about 1.0 to 20 mg per day.
  • the single dose of the compound varies depending on the administration subject, target disease, and the like.
  • the compound is usually administered in the form of an injection in an adult (as 60 kg) patient with heart disease.
  • the present invention provides a test compound administered to a non-human mammal deficient in expression of a DNA of the present invention, and detects or enhances the expression of a reporter gene.
  • the non-human mammal deficient in expression of the DNA of the present invention may be one of the above-mentioned non-human mammals deficient in expression of the DNA of the present invention, in which the DNA of the present invention is not introduced by introducing a reporter gene. Those which are activated and which can express the reporter gene under the control of the promoter for the DNA of the present invention are used.
  • test compound examples include the same compounds as described above.
  • the same gene as described above is used, and a) -galactosidase gene (1 ac Z), a soluble alkaline phosphatase gene or a luciferase gene is suitable.
  • the reporter gene is under the control of the promoter for the DNA of the present invention. By tracing the expression, the activity of the promoter can be detected.
  • a tissue that originally expresses the protein of the present invention may be used.
  • j3-galactosidase is expressed instead of the protein of the present invention.
  • a reagent that serves as a substrate for j8-galactosidase such as 5-promote 4-monocloth-3- ⁇ f-indolyl-j8-galactovyranoside (X-gal)
  • X-gal 5-promote 4-monocloth-3- ⁇ f-indolyl-j8-galactovyranoside
  • the protein-deficient mouse of the present invention or a tissue section thereof is fixed with dartalaldehyde or the like, washed with phosphate buffered saline (PBS), and then stained with X-ga1 at room temperature or at 37 ° C. After reacting for about 30 minutes to 1 hour at about ° C, the tissue sample was washed with ImM EDTA / PBS solution to stop the 3-galactosidase reaction and to observe the coloration. I just think.
  • mRNA encoding 1 ac Z may be detected according to a conventional method.
  • 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 DNA promoter activity 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). Salts are used, and physiologically acceptable acid addition salts are particularly preferred.
  • Such salts include, for example, inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.) Salts or salts with organic acids (such as 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.) Are used.
  • the compound of the present invention or a salt thereof which promotes the promoter activity for DNA can promote the expression of the protein of the present invention and promote the function of the protein, for example, it is characterized by a decrease in cardiac function. It is useful as a drug for the prevention and treatment of diseases (eg, heart failure after myocardial infarction; angina pectoris; cardiomyopathy; heart diseases such as heart failure derived from diseases such as angina pectoris and cardiomyopathy).
  • diseases eg, heart failure after myocardial infarction; angina pectoris; cardiomyopathy; heart diseases such as heart failure derived from diseases such as angina pectoris and cardiomyopathy).
  • the compound of the present invention or a salt thereof that inhibits the activity of a promoter for DNA can inhibit the expression of the protein of the present invention and inhibit the function of the protein. It is useful as a drug for the prevention and treatment of diseases (eg, heart failure after myocardial infarction; angina pectoris; cardiomyopathy; heart diseases such as heart failure derived from diseases such as angina pectoris and cardiomyopathy). .
  • diseases eg, heart failure after myocardial infarction; angina pectoris; cardiomyopathy; heart diseases such as heart failure derived from diseases such as angina pectoris and cardiomyopathy.
  • a compound derived from the compound obtained by the above screening can also be used.
  • 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 or a salt thereof.
  • the preparations obtained in this way are safe and low toxic, and thus can be used, for example, in humans or other mammals (eg, rats, mice, guinea pigs, egrets, sheep, Pigs, pests, pomas, 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 overnight activity against DNA is orally administered, Generally, in an adult (assuming a body weight of 60 kg) patient with heart 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 per day is used.
  • Administer in an adult (assuming a body weight of 60 kg) patient with heart 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 per day is used.
  • Administer in an adult (assuming a body weight of 60 kg) patient with heart
  • the single dose of the compound varies depending on the administration subject, target disease, and the like.
  • a compound that promotes the promoter activity for DNA of the present invention is usually administered in the form of an injection to an adult.
  • the compound When administered to a patient with a heart disease of about 60 kg (as 60 kg), the compound is administered in an amount of about 0.01 to 3 Omg per day, preferably about 0.1 to 20 mg, more preferably about 0.1 to 1 mg per day. It is convenient to administer about Omg 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 administered parenterally, generally, in an adult (assuming a body weight of 6 O kg) heart disease patients, the compound is reduced to about 0 per day. 1 to 10 Omg, preferably about 1.0 to 50 mg, more preferably about 1.0 to 20 mg.
  • the single dose of the compound varies depending on the subject of administration, the target disease, and the like.
  • the compound of the present invention that inhibits the promoter activity on DNA may be administered in the form of an injection.
  • the compound When administered to a normal adult (as 6 O kg) patient with a heart disease, the compound is administered in an amount of about 0.01 to 3 Omg per day, preferably about 0.1 to 2 Omg, more preferably about 0.1 to 2 Omg. It is convenient to administer about 0.1 to 1 Omg by intravenous injection. In the case of other animals, the dose can be administered in terms of 60 kg.
  • the non-human mammal deficient in expression of the DNA of the present invention is extremely useful for screening a compound or a salt thereof that promotes or inhibits the activity of the promoter for the DNA of the present invention.
  • Investigating or preventing the causes of various diseases caused by insufficient expression of DNA can greatly contribute to the development of therapeutic drugs.
  • genes encoding various proteins are ligated downstream thereof and injected into egg cells of an animal to produce a so-called transgenic animal (gene). Introduced animal) It is also possible to synthesize the polypeptide in an appropriate manner and to examine the action in the living body. Furthermore, by binding an appropriate repo overnight gene to a part of the above-mentioned promoter and establishing a cell line in which the gene is expressed, the action of specifically promoting or suppressing the ability of the protein itself of the present invention to produce in the body is achieved. It can be used as a search system for low molecular weight compounds with.
  • bases, amino acids, and the like are indicated by abbreviations based on the abbreviations of the IU PAC-IUB Commission on Biochemical Nomenclature or commonly used abbreviations in the art, and examples thereof are described below.
  • amino acids may have optical isomers, L-form shall be indicated unless otherwise specified.
  • Example 2 shows the amino acid sequence of the protein of the present invention (derived from rat) obtained in Example 1.
  • Example 1 shows the nucleotide sequence of a primer used in Example 1.
  • Example 1 shows the nucleotide sequence of a primer used in Example 1.
  • Example 1 shows the nucleotide sequence of a primer used in Example 1.
  • Example 1 shows the nucleotide sequence of the gene fragment obtained in Example 1.
  • Example 1 shows the nucleotide sequence of a primer used in Example 1.
  • Example 1 shows the nucleotide sequence of a primer used in Example 1.
  • Example 1 shows the nucleotide sequence of a primer used in Example 1.
  • Example 1 shows the nucleotide sequence of a primer used in Example 1.
  • Example 1 shows the entire nucleotide sequence including the non-coding region of DNA (gene) encoding the amino acid sequence represented by SEQ ID NO: 1 obtained in Example 1.
  • Example 1 shows the nucleotide sequence of a primer used in Example 1.
  • Example 1 shows the nucleotide sequence of a primer used in Example 1.
  • Example 3 shows the nucleotide sequence of a primer used in Example 2.
  • Example 3 shows the nucleotide sequence of a primer used in Example 2.
  • Example 4 shows the nucleotide sequence of a probe used in Example 2.
  • nucleotide sequence represented by SEQ ID NO: 2 this shows the 391st to 402nd partial nucleotide sequence from the 5 'end.
  • this shows the 490th to 495th partial nucleotide sequence from the 5 'end.
  • this shows the 796th to 804th partial nucleotide sequence from the 5 'end.
  • this shows the 829th to 846th partial nucleotide sequence from the 5 'end.
  • the transformant Escherichia coli DH5 o; / pTB2165 obtained in Example 1 described below has been used since October 19, 2000 at 1-1-1 Tsukuba-Higashi, Ibaraki, Japan 1 6 (Postal Code 305-8566) at the National Institute of Advanced Industrial Science and Technology (AIST), the Patent Organism Depositary Center (formerly Ministry of International Trade and Industry, National Institute of Advanced Industrial Science and Technology (NI BH)) under the deposit number FERM BP-7327.
  • Rats 1 week, 8 weeks, 20 weeks, and 30 weeks after surgery were subjected to thoracotomy under pentobarbital anesthesia, the heart was removed, and the coronary artery was perfused retrograde from the aorta with saline. Blood was washed away. After removing tissues other than the left ventricle from the extracted heart with scissors, infarct formation was confirmed and then the infarct region (scar formation site) was removed, leaving only the non-infarcted region. This was cut into small pieces with scissors, and Tota1 RNA was extracted using ISOGEN (Wako Pure Chemical).
  • reaction was performed using TaKaRa La Taa with GC buffer (Takara Shuzo) on a thermal cycler gene amp PGR syst em 9700 (manufactured by PerkinElmer), for 30 seconds at 95, 30 seconds at 62 ° C, and 3 at 72. 33 cycles were repeated with one minute as one cycle.
  • a DNA consisting of the following base sequence was synthesized, and PCR was carried out using a marathon lady rat heart cDNA library 1 (Clontech) as type III.
  • the nucleotide sequence of the obtained gene fragment was decoded to obtain a DNA comprising the nucleotide sequence represented by SEQ ID NO: 12 and containing the DNA encoding the novel Nkx2.5 analogous protein of the present invention.
  • the DNA consisting of the nucleotide sequence represented by SEQ ID NO: 2 obtained in Example 1 was designated as type III, and the nucleotides represented by SEQ ID NO: 15 and SEQ ID NO: 16 Using the DNA consisting of the sequence as a primer, PCR was performed in the same manner as in Example 1- (3) to prepare a probe (SEQ ID NO: 17).
  • Rat MTN Blot manufactured by Clontech was used as a membrane for northern blotting.
  • Prehybridization was performed at 68 ° C using Express Hyb Hybridization solution (Clontech) as a hybridization solution.
  • the Nkx2.5-like gene fragment prepared above was labeled as a probe using [ ⁇ - 32 P] dCTP and BcaBEST Labeling Kit (Takara Shuzo). Hybridization was performed in Express Hyb Hybridization solution (Clontech) containing the labeled probe at 68 ° C for 1 hour. The membrane was finally washed at 50 ° C in 0.1 XSSC, 0.1% SDS solution, and BAS-2000 (Fujifilm) was used for detection. As a result, it was found that the heart was the main expression site of DNA encoding the novel Nkx2.5 analogous protein of the present invention.
  • Example 1 Tota1 RNA derived from the non-infarcted region of the left ventricle of the rat after 1 week, 8 weeks, 20 weeks and 30 weeks after myocardial infarction described in (2) Eight weeks after the operation, cDNA was synthesized from the left ventricle-derived Tota 1 RNA using TadM an Reverse Transcription Reagents (manufactured by PE Applied Biosystems). Next, using a TadMan Rodent G3PDH control reagent VIC 'probe (manufactured by PE Applied Biosystems), the copy number of glycerol triphosphate dehydrogenase was determined by PCR using ABI Prism 7700 seauence Detectio. Performed by n System.
  • reaction was performed using TaKaRa La Ta with GC buffer (Takara Shuzo) in a samurai cycler gene amp PCR syst em 9700 (manufactured by PerkinElmer) at 95 ° C for 30 seconds and at 62 ° C.
  • the cycle was performed at 72 ° C for 30 seconds and 3 minutes as one cycle.
  • sampling was performed for each cycle from 30 to 40.
  • the obtained DNA encoding the novel Nkx2.5 analogous protein of the present invention was subjected to electrophoresis with 2% agarose, and the band detected by ethidium bromide staining was analyzed with an image analyzer (Fluorlmager 595, manufactured by Molecular Dynamics). Quantified.
  • the vertical axis indicates the copy number (expression level) of the DNA encoding the novel Nkx2.5-like protein of the present invention in the left ventricle, which is the number of copies of the glycerol triphosphate dehydrogenase gene, which is a housekeeping gene.
  • the numerical value obtained by dividing by the measurement value of the sham operation group was shown, and this was indicated as fold increase.
  • the time (weeks) on the horizontal axis shows the progress of each sample of the heart failure model used.
  • sham 8w for sham operation group MI lw for 1 week after surgery, MI 8w for 8 weeks after surgery, MI 20w for 20 weeks after surgery, and Ml 30 w for heart after 30 weeks after surgery Indicates the sample name.
  • the DNA encoding the novel Nkx2.5 analogous protein of the present invention showed a marked decrease (0.29-fold) at 1 week after surgery and an increasing tendency (1.135-fold) at 8 weeks after surgery. However, it increased at 20 weeks after the operation (2 times) and returned to the sham operation group level at 30 weeks.
  • One week after the operation is considered to be the time when infarction is being formed. It is presumed that myocardial cells in the downstream region from the coronary artery died and died rapidly, and inflammation was caused by lymphocyte infiltration. Since 20 to 30 weeks after the operation is immediately before death is observed, 8 weeks after the operation is the time when the compensation mechanism is operating, and after 20 weeks after the operation, sufficient compensation is available. It is likely that the order is not working, or that there is a compensatory failure due to excessive compensation mechanisms. Therefore, one week after the operation was considered the acute phase (acute heart failure), eight weeks after the surgery was considered the chronic phase (heart failure decompensation), and 20 weeks after the surgery was considered the end stage (heart failure decompensation).
  • the compensatory mechanism involved in the transition from myocardial infarction to heart failure is considered as follows.
  • the myocardial cells fall off (necrosis or apoptosis)
  • the remaining myocardial cells become enlarged to compensate for the functions of the lost myocardial cells throughout the heart, and the heart is remodeled with diastolic and fibrotic changes (cardiac). Remodeling) occurs.
  • the molecules involved in compensatory failure itself have not yet been identified, and the mechanism has not been elucidated.
  • the novel N kx of the present invention which is a gene highly homologous to N kX 2.5 which is a positive transcription regulator of the heart-specific gene group, is used.
  • An expression profile for DNA encoding the 2.5 analogous protein was generated. This gene decreased remarkably in the acute phase, and increased from the chronic phase to the end phase.
  • the detailed expression profile of the DNA encoding the novel N kx 2.5 analogous protein of the present invention which has been clarified in the above example, can be considered as follows.
  • Nkx2.5 induces the expression of atrial sodium diuretic peptide and ventricular sodium diuretic peptide, which are thought to protect the heart.
  • Myosin one of the cardiac contractile proteins It has been reported to positively regulate light chain 2V expression. Therefore, there is a possibility that the Nkx2.5-like gene has a similar function, and it is considered that the decreased expression in the acute phase is directly linked to the worsening of the disease state.
  • CARP diatsk-adriamycin-resbonsib 'protein
  • the drug of the present invention that regulates the expression of DNA encoding the novel Nkx2.5-like protein and the function of the gene product is useful as a new agent for preventing and treating heart disease.
  • the protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a salt thereof is novel, and a compound or a salt thereof which regulates the activity of the protein or the salt thereof;
  • An antibody that regulates the activity of the salt can be used, for example, as a prophylactic or therapeutic agent for heart disease and the like.
  • An antisense nucleotide having a nucleotide sequence complementary or substantially complementary to DNA encoding the protein or a salt thereof can suppress the expression of the protein or a salt thereof. It can be used as a therapeutic agent.

Landscapes

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

Abstract

It is intended to provide a novel protein, its DNA, etc. A compound controlling the activity of the above-described protein or its salt and a neutralizing antibody controlling the activity of this protein are useful as, for example, preventives and remedies for heart diseases.

Description

新規疾患関連遺伝子およびその用途 技術分野  New disease-related genes and their uses
本発明は、 疾患関連遺伝子の用途に関する。 さらに詳しくは、 該疾患関連遺伝 子産物を用いる薬剤のスクリーニング方法、 心筋症、 心筋梗塞、 心不全、 狭心症 などの心疾患の診断マ一カーとして有用な疾患関連遺伝子に対するアンチセンス ヌクレオチド、 該疾患関連遺伝子産物に対する抗体などに関する。 背景技術  The present invention relates to uses of disease-related genes. More specifically, a method for screening a drug using the disease-related gene product, an antisense nucleotide against a disease-related gene useful as a marker for diagnosis of heart diseases such as cardiomyopathy, myocardial infarction, heart failure, and angina; It relates to an antibody against a related gene product and the like. Background art
心不全とは、 心筋の収縮不全と考えられている。 発症の機序としては、 次のよ うなものが考えられる。 心筋の障害、 心臓ポンプの機械的、 機能的異常、 高血圧 及び肺高血圧による圧負荷、 貧血、 急性腎炎など容量負荷などが原因となり、 生 体の需要に応じた血液量を心臓が拍出し得なくなる状態が生じる。 これに対して 、 交感神経系、 神経一体液一内分泌系などの代償機序が作動し、 生体の恒常性の 維持に向かう。 心不全の代償機序としては、 1 ) 前述の負荷が増大して心臓の収 縮力が増し、 サルコメァの長さの増大に伴って心拡大が生じる、 2 ) 心筋の収縮 単位が増し、 その結果として心筋肥大が生じる、 3 ) 全身に必要な血液を駆出で きない状態を補うため神経 f夜性因子が活性化され、 局所的には心筋繊維化が進展 する。 基本的には与えられた負荷に、 適応しょうとする機序であるが、 不十分な 作動によって心不全が促進する場合もあれば、 逆に過剰な作動によって心筋傷害 を生じ、 心不全を悪化させる場合もある。 代償機序が作動した結果として、 心筋 細胞が肥大し、 心肥大が生じる。 しかしながら、 前述の障害あるいは負荷が慢性 的に継続された場合、 その代償機序が破綻する。 つまり、 肥大した心筋細胞に十 分な量の血液が供給されずに虚血に陥り、 これが原因で心筋収縮不全などの心筋 障害が生じ、 心拍出量の低下、 臓器循環障害、 静脈鬱血、 体液貯留などを伴う心 不全症候群を来すことになる。 これに対する治療としては、 心筋細胞障害の改善 、 心保護作用の強化、 心筋収縮不全による心機能低下の回復及びその原因である 生体の代償破綻の抑制あるいは過剰な代償機序の改善が必要となる。 現在、 該心 不全症候群の治療には、 臨床的には強心薬として 1 . ジゴキシンなどの強心配 糖体、 2 . ドブ夕ミンなどの交感神経作動薬、 3 . アムリノンなどのホスホジェ ステラ一ゼ阻害薬が、 また血管拡張薬としてはヒドララジン、 カルシウム拮抗薬 及びアンジォテンシン I変換酵素阻害薬、 アンジォテンシン I I受容体拮抗薬な どが使用されている。 また、 他拡張型心筋症の治療には、 /3ブロッカーなどが使 用されている。 Heart failure is considered to be inadequate contraction of the heart muscle. The mechanism of the onset may be as follows. Myocardial damage, mechanical and functional abnormalities of the heart pump, pressure overload due to hypertension and pulmonary hypertension, volume overload such as anemia and acute nephritis can cause the heart to pump out blood volume according to the demands of the organism A state of disappearance occurs. On the other hand, the sympathetic nervous system, the nervous system, the endocrine system and other compensatory mechanisms operate to maintain the homeostasis of the body. The compensatory mechanisms of heart failure include: 1) the above-mentioned load increases and the contractility of the heart increases, and the heart expands as the length of the sarcomer increases.2) The contraction unit of the myocardium increases, resulting in Myocardial hypertrophy occurs as a result. 3) Nerve f nocturnal factors are activated to compensate for the inability to excrete the blood necessary for the whole body, and myocardial fibrosis progresses locally. Basically, the mechanism is to adapt to a given load.However, insufficient operation may promote heart failure, while excessive operation may cause myocardial injury and exacerbate heart failure There is also. As a result of activation of the compensatory mechanism, myocardial cells become enlarged and cardiac hypertrophy occurs. However, if the aforementioned obstacles or loads are chronically sustained, the compensatory mechanism will fail. In other words, ischemia occurs without a sufficient amount of blood being supplied to the enlarged cardiomyocytes, resulting in myocardial damage such as myocardial contractile dysfunction, decreased cardiac output, impaired organ circulation, venous congestion, This results in heart failure syndrome with fluid retention. Treatment for this requires improvement of myocardial cell damage, enhancement of cardioprotective action, recovery of cardiac dysfunction due to myocardial contractile dysfunction, and suppression of decompensation of the living organism, which is the cause, or improvement of excessive compensation mechanisms . Currently, the heart For the treatment of insufficiency syndrome, clinically cardiotonic drugs include: 1. cardiac glycosides such as digoxin, 2. sympathomimetics such as dobuyumin, and 3. phosphogesterase inhibitors such as amrinone. As vasodilators, hydralazine, calcium antagonists, angiotensin I converting enzyme inhibitors, angiotensin II receptor antagonists and the like have been used. For treatment of other dilated cardiomyopathy, a / 3 blocker is used.
しかしながら、 速やかな代償機序の作動、 過剰な代償機序を抑制あるいは代償 破綻を抑制 (アポトーシス抑制を含む) し得る治療薬はない。 速やかな代償機序 の作動や過剰な代償機序あるいは代償破綻の抑制という観点から治療薬の開発が 望まれている。 発明の開示  However, there are no therapeutic agents that can activate the compensatory mechanism promptly, suppress excessive compensatory mechanisms or suppress compensatory failure (including suppression of apoptosis). There is a need for the development of therapeutic agents from the viewpoint of prompt activation of the compensation mechanism and suppression of excessive compensation mechanisms or failure. 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、 冠動脈結 紮による心筋梗塞モデルラットの心不全発症時において発現が増加する遺伝子を 見出した。 この、 配列番号: 2で表される塩基配列を有する mR NAの発現プロ ファイルを詳細に検討した結果、 この mR NAは、 術後' 1週で顕著に減少し、 術 後 8週から 2 0週で増加傾向を示し、 3 0週で偽手術群と同レベルに戻る発現プ 口ファイルを示すことが明らかとなった。 本遺伝子は、 ショウジヨウバエで報告 されているチンマンの哺乳類カウンターパートと考えられている N k x 2 . 5 ( 別名 Csx, 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. Nat l. Acad. Sci. USA) , 第 90巻 , 第 8145- 8149頁、 1993年;デベロプメント、 第 119巻、 第 419- 431頁、 1993年; 医学のあゆみ、 第 185巻、 第 1 -7頁、 1998年) の類似遺伝子としてラット心臓 c D NAライブラリ一からクローニングした。 N k x 2 . 5は心臓の発生過程で重 要な役割を演じる転写因子として機能解析が進められているが、 心不全病態との 関連性は明らかになっていない。 一方で今回クローニングした N k x 2 . 5類似 遺伝子は心不全発症時に増加した。 これらの知見に基づいて、 さらに検討を重ね た結果、 本発明を完成するに至った。  The present inventors have conducted intensive studies in order to solve the above-described problems, and as a result, have found a gene whose expression increases when heart failure occurs in a myocardial infarction model rat due to coronary artery ligation. As a result of detailed examination of the expression profile of mRNA having the nucleotide sequence represented by SEQ ID NO: 2, this mRNA was significantly reduced one week after the operation, and decreased from 8 weeks after the operation to 20 weeks. Weeks showed an increasing trend, and at week 30 it was evident that the expression port file returned to the same level as the sham-operated group. This gene is believed to be the chimney mammalian counterpart reported in Drosophila, Nkx2.5 (also known as Csx, Processings of the National Academy of Sciences'). Proc. Natl. Acad. Sci. USA, Vol. 90, pp. 8145-8149, 1993; Development, Vol. 119, pp. 419-431, 1993; History of Medicine 185, pp. 1-7, 1998) and cloned from a rat heart cDNA library. Functional analysis of Nkx2.5 as a transcription factor that plays an important role in the developmental process of the heart is ongoing, but its relationship to heart failure pathology has not been clarified. On the other hand, the Nkx2.5 similar gene cloned this time increased at the onset of heart failure. As a result of further studies based on these findings, the present invention was completed.
すなわち、 本発明は、 (1) 配 J番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質またはその塩、 That is, the present invention (1) a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by arrangement J number: 1, or a salt thereof;
(2) 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またはその  (2) a protein containing the amino acid sequence represented by SEQ ID NO: 1 or a protein thereof;
'つ  '
(3) 前記 (1) 記載のタンパク質の部分ペプチドまたはその塩、  (3) a partial peptide of the protein according to (1) or a salt thereof,
(4) 前記 (1) 記載のタンパク質または前記 (3) 記載の部分ペプチドをコ 一ドするポリヌクレオチドを含有するポリヌクレオチド、  (4) a polynucleotide comprising a polynucleotide encoding the protein of (1) or the partial peptide of (3),
(5) DNAである前記 (4) 記載のポリヌクレオチド、  (5) the polynucleotide according to (4), which is a DNA,
(6) 配列番号: 2で表される塩基配列を含有する前記 (5) 記載の DNA、 (7) 前記 (4) 記載のポリヌクレオチドを含有する組換えべクタ一、 (6) a DNA according to the above (5) containing the nucleotide sequence represented by SEQ ID NO: 2, (7) a recombinant vector containing the polynucleotide according to the above (4),
(8) 前記 (7) 記載の組換えベクターで形質転換された形質転換体、(8) a transformant transformed with the recombinant vector according to (7),
(9) 前記 (8) 記載の形質転換体を培養し、 前記 (1) 記載のタンパク質も しくはその塩または前記 (3) 記載の部分ペプチドまたはその塩を生成、 蓄積せ しめ、 これを採取することを特徴とする前記 (1) 記載のタンパク質もしくはそ の塩または前記 (3) 記載の部分ペプチドもしくはその塩の製造方法、 (9) The transformant according to (8) is cultured, and the protein or salt thereof according to (1) or the partial peptide or salt thereof according to (3) is produced, accumulated, and collected. A method for producing the protein according to the above (1) or a salt thereof, or the partial peptide according to the above (3) or a salt thereof,
(10) 前記 (1) 記載のタンパク質もしくはその塩または前記 (3) 記載の 部分べプチドもしくはその塩を含有してなる医薬、  (10) a pharmaceutical comprising the protein or salt thereof according to (1) or the partial peptide or salt thereof according to (3);
(11) 前記 (1) 記載のタンパク質もしくはその塩または前記 (3) 記載の 部分べプチドもしくはその塩に対する抗体、  (11) an antibody against the protein or salt thereof according to (1) or the partial peptide or salt thereof according to (3),
(12) 前記 (11) 記載の抗体を含有する診断薬、  (12) a diagnostic agent comprising the antibody according to (11),
(13) 前記 (1) 記載のタンパク質もしくはその塩または前記 (3) 記載の 部分ペプチドもしくはその塩を用いることを特徴とする、 前記 (1) 記載のタン パク質もしくはその塩または前記 (3) 記載の部分ペプチドもしくはその塩の活 性を調節する化合物またはその塩のスクリーニング方法、  (13) The protein or salt thereof according to (1), or the salt thereof, wherein the protein or salt thereof according to (1) or the partial peptide or salt thereof according to (3) is used. A method for screening a compound or a salt thereof that regulates the activity of the partial peptide or a salt thereof according to the above,
(14) 前記 (1) 記載のタンパク質もしくはその塩または前記 (3) 記載の 部分ペプチドもしくはその塩が、 前記 (1) 記載のタンパク質または前記 (3) 記載の部分ペプチドをコードする D N Aを含有する D N Aで形質転換された形質 転換体の細胞質内に発現されたものである前記 (13) 記載のスクリーニング方 法、 (15) 前記 (1) 記載のタンパク質もしくはその塩または前記 (3) 記載の 部分ペプチドもしくはその塩を含有することを特徴とする、 前記 (1) 記載の夕 ンパク質もしくはその塩または前記 (3) 記載の部分ペプチドもしくはその塩の 活性を調節する化合物またはその塩のスクリーニング用キット、 (14) The protein or salt thereof according to (1) or the partial peptide or salt thereof according to (3) contains a DNA encoding the protein according to (1) or the partial peptide according to (3). The screening method according to the above (13), which is expressed in the cytoplasm of a transformant transformed with the DNA; (15) The protein or salt thereof according to (1), which comprises the protein or salt thereof according to (1) or the partial peptide or salt thereof according to (3). A) a kit for screening a compound or a salt thereof that regulates the activity of the partial peptide or a salt thereof described above;
(16) 前記 (13) 記載のスクリーニング方法または前記 (15) 記載のス クリーニング用キットを用いて得られる、 前記 (1) 記載のタンパク質もしくは その塩または前記 (3) 記載の部分ペプチドもしくはその塩の活性を調節する化 合物またはその塩、  (16) The protein or salt thereof according to (1) or the partial peptide or salt thereof according to (3), which is obtained by using the screening method according to (13) or the screening kit according to (15). A compound or a salt thereof that regulates the activity of
(17) 前記 (16) 記載の化合物またはその塩を含有してなる医薬、 (18) 心疾患の予防 ·治療剤である前記 (17) 記載の医薬、  (17) a medicament comprising the compound according to (16) or a salt thereof, (18) a medicament according to (17), which is a prophylactic or therapeutic agent for heart disease.
(19) 前記 (1) 記載のタンパク質または前記 (3) 記載の部分ペプチドを コードする DNAに相補的もしくは実質的に相補的な塩基配列を有するァンチセ ンスヌクレオチド、  (19) an antisense nucleotide having a base sequence complementary or substantially complementary to DNA encoding the protein according to (1) or the partial peptide according to (3),
(20) 前記 (19) 記載のアンチセンスヌクレオチドを含有してなる医薬、 (21) 前記 (4) 記載のポリヌクレオチドを含有してなる医薬、  (20) a medicine comprising the antisense nucleotide according to (19), (21) a medicine comprising the polynucleotide according to (4),
(22) 前記 (4) 記載のポリヌクレオチドを含有してなる診断薬、  (22) a diagnostic agent comprising the polynucleotide according to (4),
(23) 哺乳動物に対して、 前記 (16) 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心疾患の予防 ·治療方法、  (23) a method for preventing or treating heart disease, which comprises administering to a mammal an effective amount of the compound or a salt thereof according to (16);
(24) 心疾患の予防 ·治療剤を製造するための前記 (16) 記載の化合物ま たはその塩の使用などを提供する。  (24) Use of the compound or a salt thereof according to the above (16) for producing an agent for preventing or treating heart disease.
さらに、 本発明は、  Further, the present invention provides
(25) 前記 (1) 記載のタンパク質をコードする DNAまたはその変異 DN Aを有する非ヒト DNA導入動物、  (25) a DNA encoding the protein according to the above (1) or a non-human DNA transgenic animal having a mutant DNA thereof,
(26) 非ヒト動物がゲッ歯動物である前記 (25) 記載の動物、  (26) the animal according to (25), wherein the non-human animal is a rodent;
(27) ゲッ歯動物がマウスまたはラッ卜である前記 (26) 記載の動物、 (27) The animal according to (26), wherein the rodent is a mouse or a rat.
(28) 本発明の DNAが不活性化された非ヒト哺乳動物胚幹細胞、 (28) a non-human mammalian embryonic stem cell in which the DNA of the present invention has been inactivated,
(29) 該 DNAがレポーター遺伝子を導入することにより不活性化された前 記 (28) 記載の胚幹細胞、  (29) The embryonic stem cell according to the above (28), wherein the DNA is inactivated by introducing a reporter gene,
(30) ネオマイシン耐性である前記 (28) 記載の胚幹細胞、 (31) 非ヒト哺乳動物がゲッ歯動物である前記 (28) 記載の胚幹細胞、 (32) ゲッ歯動物がマウスである前記 (31) 記載の胚幹細胞、 (30) The embryonic stem cell according to (28), which is neomycin-resistant, (31) the embryonic stem cell according to (28), wherein the non-human mammal is a rodent; (32) the embryonic stem cell according to (31), wherein the rodent is a mouse;
(33) 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、 (34) 該 DNA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 レポ一夕一遺伝子が本発明の D N Aに対するプロモーターの制御下で発現しうる 前記 (33) 記載の非ヒ卜哺乳動物、  (33) a non-human mammal deficient in expression of the DNA in which the DNA of the present invention has been inactivated, (34) the DNA has been inactivated by introducing a repo overnight gene, and the repo overnight gene is The non-human mammal according to the above (33), which can be expressed under the control of a promoter for the DNA of the present invention,
(35) 非ヒ卜哺乳動物がゲッ歯動物である前記 (34) 記載の非ヒト哺乳動 物、  (35) the non-human mammal according to the above (34), wherein the non-human mammal is a rodent;
(36) ゲッ歯動物がマウスである前記 (35) 記載の非ヒト哺乳動物、 およ び  (36) the non-human mammal according to the above (35), wherein the rodent is a mouse; and
(37) 前記 (36) 記載の動物に、 試験化合物を投与し、 レポーター遺 β子 の発現を検出することを特徴とする本発明の D N Aに対するプロモータ一活性を 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なども提供する。 図面の簡単な説明  (37) A compound or a salt thereof, which comprises administering a test compound to the animal according to (36) and detecting the expression of a reporter gene β, which promotes or inhibits the activity of a promoter against DNA of the present invention. Also provided are screening methods and the like. BRIEF DESCRIPTION OF THE FIGURES
図 1は実施例 1で示した本発明の新規 Nkx2.5類似タンパク質をコードする DN A (NolO rCsx full) と AF006664 (AF006664Csx. SE) のァライメントを示す (図 2に続く) 。  FIG. 1 shows an alignment of DNA (NolOrCsx full) and AF006664 (AF006664Csx. SE) encoding the novel Nkx2.5 analogous protein of the present invention shown in Example 1 (continued from FIG. 2).
図 2は実施例 1で示した本発明の新規 Nkx2.5類似夕ンパク質をコードする DN A (NolO rCsx full) と AF006664 (AF006664Csx. SE) のァライメントを示す (図 1の続き、 図 3に続く) 。  FIG. 2 shows an alignment of DNA (NolOrCsx full) and AF006664 (AF006664Csx.SE) encoding the novel Nkx2.5-like protein of the present invention shown in Example 1 (continued from FIG. 1 and FIG. Continue) .
図 3は実施例 1で示した本発明の新規 Nkx2.5類似夕ンパク質をコードする DN A (NolO rCsx full) と AF006664 (AF006664Csx. SE) のァライメントを示す (図 2の続き、 図 4に続く) 。  FIG. 3 shows an alignment of DNA (NolOrCsx full) and AF006664 (AF006664Csx. SE) encoding the novel Nkx2.5-like protein of the present invention shown in Example 1 (continued from FIG. 2 and FIG. Continue) .
図 4は実施例 1で示した本発明の新規 Nkx2.5類似夕ンパク質をコードする DN A (NolO rCsx full) と AF006664 (AF006664Csx. SE) のァライメントを示す (図 3の続き、 図 5に続く) 。  FIG. 4 shows the alignment of DNA (NolOrCsx full) and AF006664 (AF006664Csx.SE) encoding the novel Nkx2.5-like protein of the present invention shown in Example 1 (continued from FIG. 3, FIG. Continue) .
図 5は実施例 1で示した本発明の新規 Nkx2.5類似タンパク質をコードする DN A (NolO rCsx full) と AF006664 (AF006664Csx. SE) のァライメントを示す (図 4の続き、 図 6に続く) 。 FIG. 5 shows the alignment of DNA (NolOrcsx full) and AF006664 (AF006664Csx. SE) encoding the novel Nkx2.5 analogous protein of the present invention shown in Example 1 (FIG. (Continued from Figure 4 and continued from Figure 6).
図 6は実施例 1で示した本発明の新規 Nkx2. 5類似タンパク質をコ一ドする D N A (NolO rCsx ful l) と AF006664 (AF006664Csx. SE) のァライメントを示す (図 5の続き、 図 7に続く) 。  FIG. 6 shows an alignment between DNA (NolO rCsx ful) encoding the novel Nkx2.5 analogous protein of the present invention shown in Example 1 and AF006664 (AF006664Csx.SE) (continuation of FIG. 5 and FIG. 7). Continue) .
図 7は実施例 1で示した本発明の新規 Nkx2. 5類似タンパク質をコードする D N A (NolO rCsx ful l) と删 6664 (AF006664Csx. SE) のァライメン卜を示す (図 6の続き、 図 8に続く) 。  FIG. 7 shows an alignment between DNA (NolO rCsx ful) encoding the novel Nkx2.5 analogous protein of the present invention and 删 6664 (AF006664Csx.SE) shown in Example 1 (continuation of FIG. 6 and FIG. Continue) .
図 8は実施例 1で示した本発明の新規 Nkx2. 5類似夕ンパク質をコードする D N A (NolO rCsx ful l) と AF006664 (AF006664Csx. SE) のァライメン卜を示す (図 7の続き、 図 9に続く) 。  FIG. 8 shows an alignment between DNA (NolO rCsx ful) encoding the novel Nkx2.5 analogous protein of the present invention and AF006664 (AF006664Csx. SE) shown in Example 1 (continuation of FIG. 7, FIG. followed by) .
図 9は実施例 1で示した本発明の新規 Nkx2. 5類似夕ンパク質をコードする D N A (NolO rCsx ful l) と AF006664 (AF006664Csx. SE) のァライメントを示す (図 8の続き) 。  FIG. 9 shows an alignment of DNA (NolO rCsx ful) encoding the novel Nkx2.5 analogous protein of the present invention shown in Example 1 and AF006664 (AF006664Csx. SE) (continuation of FIG. 8).
図 1 0は心筋梗塞モデルラットでの本発明の新規 Nkx2. 5類似タンパク質をコー ドする D N Aの経時変化を示す。 発明を実施するため最良の形態  FIG. 10 shows the time course of DNA encoding the novel Nkx2.5-like protein of the present invention in a myocardial infarction model rat. BEST MODE FOR CARRYING OUT THE INVENTION
本発明の配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の アミノ酸配列を含有するタンパク質 (以下、 本発明のタンパク質または本発明で 用いられるタンパク質と称することもある。 また、 本発明のタンパク質、 本発明 で用いられるタンパク質は、 そのアミド体、 エステル体も包含する意味で用いら れる場合がある。 ) は、 温血動物 (例えば、 ヒ卜、 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、 膝臓 |3細胞、 骨髄細胞、 メサンギゥム細胞、 ランゲル八ンス細胞、 表皮細胞、 上皮細胞、 杯細胞、 内皮細胞、 平滑筋細胞、 繊 維芽細胞、 繊維細胞、 筋細胞、 脂肪細胞、 免疫細胞 (例、 マクロファージ、 T細 胞、 B細胞、 ナチュラルキラー細胞、 肥満細胞、 好中球、 好塩基球、 好酸球、 単 球) 、 巨核球、 滑膜細胞、 軟骨細胞、 骨細胞、 骨芽細胞、 破骨細胞、 乳腺細胞、 肝細胞もしくは間質細胞、 またはこれら細胞の前駆細胞、 幹細胞もしくはガン細 胞など) もしくはそれらの細胞が存在するあらゆる組織、 例えば、 脳、 脳の各部 位 (例、 嗅球、 扁桃核、 大脳基底球、 海馬、 視床、 視床下部、 大脳皮質、 延髄、 小脳) 、 脊髄、 下垂体、 胃、 膝臓、 腎臓、 肝臓、 生殖腺、 甲状腺、 胆のう、 骨髄 、 副腎、 皮膚、 筋肉、 肺、 消化管 (例、 大腸、 小腸) 、 血管、 心臓、 胸腺、 脾臓 、 顎下腺、 末梢血、 前立腺、 睾丸、 卵巣、 胎盤、 子宮、 骨、 関節、 骨格筋などに 由来するタンパク質であってもよく、 合成タンパク質であってもよい。 A protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 of the present invention (hereinafter, also referred to as the protein of the present invention or the protein used in the present invention. The protein of the present invention and the protein used in the present invention may be used in the sense that their amides and esters are also included.) Are warm-blooded animals (for example, humans, guinea pigs, rats, mice, chickens, Egret, bush, sheep, wedge, monkey, etc.) cells (eg, hepatocytes, spleen cells, nerve cells, glial cells, knees | 3 cells, bone marrow cells, mesangial cells, Langer's cells, epidermal cells, Epithelial cells, goblet cells, endothelial cells, smooth muscle cells, fibroblasts, fiber cells, muscle cells, fat cells, immune cells (eg, macrophages) , T cells, B cells, natural killer cells, mast cells, neutrophils, basophils, eosinophils, monocytes), megakaryocytes, synovial cells, chondrocytes, bone cells, osteoblasts, ruptures Osteocytes, breast cells, hepatocytes or stromal cells, or their precursors, stem cells or cancer cells Vesicles) or any tissue where these cells are present, such as the brain, various parts of the brain (eg, olfactory bulb, amygdala, basal nucleus, hippocampus, thalamus, hypothalamus, cerebral cortex, medulla, cerebellum), spinal cord, Pituitary, stomach, knee, kidney, liver, gonad, thyroid, gall bladder, bone marrow, adrenal gland, skin, muscle, lung, digestive tract (eg, large intestine, small intestine), blood vessels, heart, thymus, spleen, submandibular gland, It may be a protein derived from peripheral blood, prostate, testis, ovary, placenta, uterus, bone, joint, skeletal muscle, or the like, or may be a synthetic protein.
配列番号: 1で表されるアミノ酸配列と実質的に同一のアミノ酸配列としては 、 配列番号: 1で表されるアミノ酸配列と約 9 7 %以上、 好ましくは約 9 8 %以 上、 さらに好ましくは約 9 9 %以上の相同性を有するアミノ酸配列などが挙げら れる。  The amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 is about 97% or more, preferably about 98% or more, more preferably the amino acid sequence represented by SEQ ID NO: 1. Examples include amino acid sequences having about 99% or more homology.
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。 実質的に同質の活性としては、 例えば、 心機能低下促進活性などが挙げられる 。 実質的に同質とは、 その活性が性質的に (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、 心機能低下促進活性が同等 (例、 約 0 . 0 1〜1 0 0倍、 好ましくは約 0 . 1〜1 0倍、 より好ましくは 0 . 5〜2倍) であることが好ましいが、 この活性の程度、 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 the protein having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 include, for example, a protein having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 described above. However, a protein having substantially the same activity as the protein having the amino acid sequence represented by SEQ ID NO: 1 is preferable. Examples of the activity of substantially the same quality include, for example, activity for promoting cardiac function decline. Substantially identical indicates that the activity is qualitatively (eg, physiologically or pharmacologically) equivalent. Therefore, it is preferable that the activity to promote cardiac function decline is equivalent (eg, about 0.01 to 100 times, preferably about 0.1 to 10 times, more preferably 0.5 to 2 times). However, quantitative factors such as the degree of this activity and the molecular weight of the protein may be different.
心機能低下促進活性などの活性の測定は、 心エコー測定装置 (セル、 第 9 7巻 、 1 8 9頁一 1 9 8頁、 1 9 9 9年) または心臓カテーテルによる心機能測定 ( サーキュレーションリサ一チ、 第 6 9巻、 3 7 0— 3 7 7頁、 1 9 9 1年) によ つて行うことができる。 更に例えばアンジォテンシン I変換酵素 (A C E) など のレニンアンジォテンシン系 (RA S ) の亢進を市販の測定キット (例、 ぺニン スラ一社製、 フェニックス社製など) を用いて測定したり、 血中カテコラミンの 増加活性 (東ソ一社製、 全自動カテコールアミン分析計) などを指標に該活性を 測定することができる。  Measurement of activity such as cardiac dysfunction promoting activity can be measured using an echocardiograph (Cell, Vol. 97, pp. 189-199, 1989) or cardiac function measurement using a cardiac catheter (circulation) (Research, Vol. 69, p. 370—p. 377, 1991). Further, for example, the enhancement of renin-angiotensin system (RAS) such as angiotensin I converting enzyme (ACE) is measured using a commercially available assay kit (eg, manufactured by Peninsula Inc., Phoenix Inc.). The activity can be measured by using, as an index, the activity of increasing catecholamine in blood (a full-automatic catecholamine analyzer manufactured by Tosoh Corporation).
また、 本発明のタンパク質としては、 例えば、 ①配列番号: 1で表されるアミ ノ酸配列中の 1または 2個以上 (好ましくは 1〜1 0個程度、 さらに好ましくは 数 (1〜5 ) 個) のアミノ酸が欠失したアミノ酸配列、 ②配列番号: 1で表され るアミノ酸配列に 1または 2個以上 (好ましくは 1〜1 0個程度、 さらに好まし くは数 (1〜5 ) 個) のアミノ酸が付加したアミノ酸配列; ③配列番号: 1で表 されるアミノ酸配列に 1または 2個以上 (好ましくは 1 ~ 1 0個程度、 さらに好 ましくは数 (1〜5 ) 個) のアミノ酸が挿入されたアミノ酸配列、 ④配列番号: 1で表されるアミノ酸配列中の 1または 2個以上 (好ましくは好ましくは 1〜 1 0個程度、 さらに好ましくは数 (1〜5 ) 個) のアミノ酸が他のアミノ酸で置換 されたアミノ酸配列、 または⑤それらを組み合わせたアミノ酸配列を含有する夕 ンパク質などのいわゆるムティンも含まれる。 Examples of the protein of the present invention include: (1) an amino acid represented by SEQ ID NO: 1; An amino acid sequence in which one or two or more (preferably about 1 to 10 and more preferably (1 to 5)) amino acids have been deleted from the amino acid sequence; 2) the amino acid represented by SEQ ID NO: 1 An amino acid sequence obtained by adding one or more (preferably about 1 to 10, more preferably a number of (1 to 5)) amino acids to the sequence; ③ the amino acid sequence represented by SEQ ID NO: 1 An amino acid sequence into which 1 or 2 or more (preferably about 1 to 10, more preferably a number (1 to 5)) amino acids have been inserted; ④ in the amino acid sequence represented by SEQ ID NO: 1 An amino acid sequence in which one or more (preferably preferably about 1 to 10 and more preferably 1 to 5) amino acids have been substituted with another amino acid, or an amino acid sequence combining them So-called muti, such as contained protein It is also included.
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 (カルボキシル末端) である。 配列番号: 1で表さ れるアミノ酸配列を含有するタンパク質をはじめとする、 本発明で用いられる夕 ンパク質は、 C末端が力ルポキシル基 (一 C O O H) 、 カルポキシレート(一 C O O— ) 、 アミド (― C O NH2) またはエステル (一 C O O R) のいずれであ つてもよい。 In the protein in the present specification, the left end is the N-terminus (amino terminus) and the right end is the C-terminus (carboxyl terminus) according to the convention of peptide labeling. The proteins used in the present invention, including the protein containing the amino acid sequence represented by SEQ ID NO: 1, include C-terminal lipoxyl group (one COOH), carboxylate (one COO—), and amide It may be either (—CONH 2 ) or ester (one COOR).
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—プチルなどの 6アルキル基、 例えば、 シクロペンチル 、 シクロへキシルなどの C 38シクロアルキル基、 例えば、 フエニル、 α—ナフ チルなどの C 61 2ァリール基、 例えば、 ベンジル、 フエネチルなどのフエニル 一 アルキル基もしくはひ一ナフチルメチルなどのひ一ナフチルー ァ ルキル基などの C 7 _ 4ァラルキル基、 ビバロイルォキシメチル基などが用いら れる。 Here, as R in the ester, e.g., methyl, Echiru, n _ propyl, isopropyl, 6 alkyl group, such as n- heptyl, for example, C 3, such as cyclohexyl cyclopentyl, cyclohexylene - 8 cycloalkyl group, for example, phenyl, α- naphthyl, such as C 6 - 1 2 Ariru group, e.g., benzyl, C 7 _ 4 Ararukiru groups such as flying one Nafuchiru § alkyl group such as a phenyl primary alkyl group or flying one naphthylmethyl such phenethyl, Bibaroiruo A xymethyl group or the like is used.
本発明で用いられるタンパク質が C末端以外にカルボキシル基 (または力ルポ キシレート) 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で用いられる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。  When the protein used in the present invention has a carboxyl group (or propyloxylate) other than the C-terminal, the protein used in the present invention includes amidated or esterified lipoxyl group. . As the ester in this case, for example, the above-mentioned C-terminal ester and the like are used.
さらに、 本発明で用いられるタンパク質には、 N末端のアミノ酸残基 (例、 メ チォニン残基) のァミノ基が保護基 (例えば、 ホルミル基、 ァセチル基などの C ト 6アルカノィルなどの 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末端のダル夕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 (例えば一 OH、 一 S H、 アミノ基、 イミダゾ ール基、 インドール基、 グァニジノ基など) が適当な保護基 (例えば、 ホルミル 基、 ァセチル基などの アルカノィル基などの 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令タン パク質なども含まれる。 Further, in the protein used in the present invention, the amino group of the N-terminal amino acid residue (eg, methionine residue) has a protecting group (eg, formyl group, acetyl group, etc.). It is protected with a protecting such Ashiru group such bets 6 Arukanoiru), which dull evening Min residues of N-terminal region is cleaved in vivo to form pin hole dull evening Min oxide, the side chains of amino acids in the molecular The above substituents (eg, 1 OH, 1 SH, amino group, imidazole group, indole group, guanidino group, etc.) are suitable protecting groups (eg, alkenyl groups such as alkanoyl groups such as formyl group, acetyl group, etc.) Protected proteins, or multiproteins such as so-called glycoproteins to which sugar chains are bound are also included.
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で表されるラッ 卜由来のタンパク質などがあげられる。  Specific examples of the protein of the present invention include, for example, the rat-derived protein represented by SEQ ID NO: 1.
本発明のタンパク質の部分ペプチド (以下、 本発明の部分ペプチドと称する場 合がある。 また、 本発明の部分ペプチドは、 そのアミド体、 エステル体も包含す る意味で用いられる場合がある。 ) としては、 前記した本発明のタンパク質の部 分ペプチドであって、 好ましくは、 前記した本発明のタンパク質と同様の活性を 有するものであればいずれのものでもよい。  Partial peptide of the protein of the present invention (hereinafter sometimes referred to as the partial peptide of the present invention. Further, the partial peptide of the present invention may be used in the sense of including its amide form and ester form.) Is a partial peptide of the above-mentioned protein of the present invention, and preferably any peptide having the same activity as the above-mentioned protein of the present invention.
例えば、 本発明のタンパク質の構成アミノ酸配列のうち少なくとも 2 0個以上 、 好ましくは 5 0個以上、 さらに好ましくは 7 0個以上、 より好ましくは 1 0 0 個以上、 最も好ましくは 1 5 0個以上のアミノ酸配列を有するペプチドなどが用 いられる。  For example, at least 20 or more, preferably 50 or more, more preferably 70 or more, more preferably 100 or more, most preferably 150 or more of the constituent amino acid sequences of the protein of the present invention. Peptides having the amino acid sequence of
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〜 5個) のアミノ酸が欠失し、 または、 そのアミノ酸配列に 1また は 2個以上 (好ましくは、 1〜2 0個程度、 より好ましくは 1〜1 0個程度、 さ らに好ましくは数 1〜5個のアミノ酸が付加し、 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、 1〜5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、 1〜5個) のアミノ酸が他の アミノ酸で置換されていてもよい。  In the partial peptide of the present invention, one or more (preferably 1 to 5) amino acids in the amino acid sequence are deleted, or one or two or more ( Preferably, about 1 to 20 amino acids, more preferably about 1 to 10 amino acids, and even more preferably about 1 to 5 amino acids are added, or 1 or 2 or more (preferably , 1 to 5) amino acids may be inserted, or one or more (preferably 1 to 5) amino acids in the amino acid sequence may be substituted with another amino acid.
より具体的には、 本発明の部分ペプチドとすれば、 配列番号: 1で表されるァ ミノ酸配列中、 ① N末端から第 5 3番目 (G 1 u ) 〜第 5 5番目 (T y r ) のァ ミノ酸残基からなる部分アミノ酸配列、 ② N末端から第 1 3 1番目 (A r g ) 〜 第 1 3 4番目 (A l a ) のアミノ酸残基からなる部分アミノ酸配列、 ③ N末端か ら第 1 6 4番目 (A l a ) 〜第 1 6 5番目 (P r o ) のアミノ酸残基からなる部 分アミノ酸配列、 ④ N末端から第 2 6 6番目 (A 1 a ) 〜第 2 6 8番目 (A 1 a ) のアミノ酸残基からなる部分アミノ酸配列、 および⑤ N末端から第 2 7 7番目 (G i n ) 〜第 2 8 2番目 (A l a ) のアミノ酸残基からなる部分アミノ酸配列 から選ばれる部分アミノ酸配列のいずれかまたは複数 (2〜5 ) 個を有する前記 した本発明の夕ンパク質の部分べプチドなどがあげられる。 More specifically, as the partial peptide of the present invention, in the amino acid sequence represented by SEQ ID NO: 1, (1) the 53rd (G1u) to 55th (Tyr) )) A partial amino acid sequence consisting of amino acid residues, ② a partial amino acid sequence consisting of the 131st (Arg) to 134th (Ala) amino acid residues from the N-terminal, ③ And the partial amino acid sequence consisting of the 164th (Ala) to 165th (Pro) amino acid residues, and ④ the 266th (A1a) to 266th from the N-terminal. A partial amino acid sequence consisting of the (A1a) amino acid residue and a partial amino acid sequence consisting of the 277th (Gin) to 282nd (Ala) amino acid residue from the N-terminal. The above-mentioned partial peptides of the protein of the present invention having any or a plurality (2 to 5) of the selected partial amino acid sequences are exemplified.
また、 本発明の部分ペプチドは C末端は力ルポキシル基 (一 C O OH) 、 カル ポキシレート (一 C O O— ) 、 アミド (― C O NH 2) またはエステル (一 C O O R) (Rは上記と同意義を示す) の何れであってもよい。 In the partial peptide, the C-terminus of the present invention the force Rupokishiru group (one CO OH), Cal Pokishireto (one COO-), amide (- CO NH 2) or an ester (one COOR) (R is as defined above ).
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、 N末端のアミノ酸残基 (例、 メチォニン残基) のァミノ基が保護基で保護されて いるもの、 N端側が生体内で切断され生成したグルタミン残基がピログルタミン 酸化したもの、 分子内のアミノ酸の側鎖上の置換基が適当な保護基で保護されて いるもの、 あるいは糖鎖が結合したいわゆる糖ペプチドなどの複合ペプチドなど も含まれる。  Further, similar to the protein of the present invention, the partial peptide of the present invention has an amino terminal residue at the N-terminal (eg, methionine residue) in which the amino group is protected by a protecting group, and the N-terminal side has a native amino acid residue. Glutamine residues generated by cleavage in the body and pyroglutamine oxidized, those in which the substituents on the side chains of the amino acids in the molecule are protected with appropriate protecting groups, and so-called glycopeptides to which sugar chains are bonded Also included are composite peptides.
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。 本発明の夕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. Preferred are acid addition salts that are chemically acceptable. 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ペプチドまたはその塩は、 前述したヒト または温血動物の細胞または組織より公知のタンパク質の精製方法により製造、 またはタンパク質をコードする D N Aを含有する形質転換体を培養することによ り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 may be produced by a known protein purification method from human or warm-blooded animal cells or tissues as described above, or by culturing a transformant containing DNA encoding the protein. It can be manufactured by Further, it can be produced according to the peptide synthesis method described later.
ヒトまたは温血動物の組織または細胞から製造する場合、 ヒトまたは温血動物 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 —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 manufactured from human or warm-blooded animal tissues or cells, human or warm-blooded animal After homogenizing the tissue or cells, the extract can be extracted with an acid or the like, and the extract can be purified and isolated by a combination of chromatography such as reverse phase chromatography and ion exchange 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、 4一べンジルォキシベンジルアルコール 樹脂、 4一メチルベンズヒドリルァミン樹脂、 P AM樹脂、 4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脂、 4一 ( 2 ' , 4, ー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 Fm o cアミノエチル) フエノキシ樹脂などを 挙げることができる。 このような樹脂を用い、 ひー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を切り出すと 同時に各種保護基を除去し、 さらに高希釈溶液中で分子内ジスルフイド結合形成 反応を実施し、 目的のタンパク質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 usually used. Such resins include, for example, chloromethyl resin, hydroxymethyl resin, benzhydrylamine resin, aminomethyl resin, 4-benzyloxybenzyl alcohol resin, 4-methylbenzhydrylamine resin, PAM resin, 4-hydroxymethyl resin Methyl phenylacetamidomethyl resin, polyacrylamide resin, 4- (2 ', 4, dimethoxyphenyl-hydroxymethyl) phenoxy resin, 4-(2', 4 'dimethoxyphenyl-Fmocaminoethyl) phenoxy resin And so on. Using such a resin, an amino acid having a suitably protected amino group and side chain functional group is condensed on the resin according to the sequence of the target protein according to various known condensation methods. At the end of the reaction, the protein is cleaved from the resin, and at the same time, various protecting groups are removed. Further, an intramolecular disulfide bond formation reaction is carried out in a highly diluted solution to obtain a target protein or partial peptide or an amide thereof.
上記した保護アミノ酸の縮合に関しては、 タンパク質合成に使用できる各種活 性化試薬を用いることができるが、 特に、 カルポジイミド類がよい。 カルポジィ 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 ' — (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〇 B t, HO O B t ) とともに保護アミノ酸を直接樹脂に添加するかまたは、 対称酸無水物または HO B tエステルあるいは HO O B tエステルとしてあらかじめ保護アミノ酸の活性 化を行った後に樹脂に添加することができる。  For 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. Examples of the carpoimides include DCC, N, N'-diisopropylcarpo- imide, N-ethyl N '-(3-dimethylaminoprolyl) carbopimide. For these activations, the protected amino acid may be added directly to the resin along with a racemization inhibitor additive (eg, H〇Bt, HOOBt) or as a symmetrical anhydride or HOBtester or HOOBtester. After the protected amino acid is activated in advance, it can be added to the resin.
保'護アミノ酸の活性化や樹脂との縮合に用いられる溶媒としては、 タンパク質 縮合反応に使用しうることが知られている溶媒から適宜選択されうる。 例えば、 N, N—ジメチルホルムアミド, N, N—ジメチルァセトアミド, N—メチルピ 口リドンなどの酸アミド類、 塩化メチレン, クロ口ホルムなどのハロゲン化炭化 水素類、 トリフルォロエタノールなどのアルコール類、 ジメチルスルホキシドな どのスルホキシド類、 ピリジン, ジォキサン, テトラヒドロフランなどのェ一テ ル類、 ァセトニトリル, プロピオ二トリルなどの二トリル類、 酢酸メチル, 酢酸 ェチルなどのエステル類あるいはこれらの適宜の混合物などが用いられる。 反応 温度はタンパク質結合形成反応に使用され得ることが知られている範囲から適宜 選択され、 通常約一 2 0 °C〜 5 0での範囲から適宜選択される。 活性化されたァ ミノ酸誘導体は通常 1 . 5〜4倍過剰で用いられる。 ニンヒドリン反応を用いた テス卜の結果、 縮合が不十分な場合には保護基の脱離を行なうことなく縮合反応 を繰り返すことにより十分な縮合を行なうことができる。 反応を繰り返しても十 分な縮合が得られないときには、 無水酢酸またはァセチルイミダゾールを用いて 未反応アミノ酸をァセチル化することによって、 後の反応に影響を与えないよう にすることができる。 The solvent used for activating the protective amino acid or condensing with the resin can be appropriately selected from solvents known to be usable for the protein condensation reaction. For example, acid amides such as N, N-dimethylformamide, N, N-dimethylacetamide, N-methylpiperidone, and halogenated carbons such as methylene chloride and chloroform. Hydrogens, alcohols such as trifluoroethanol, sulfoxides such as dimethyl sulfoxide, ethers such as pyridine, dioxane, and tetrahydrofuran; nitriles such as acetonitrile and propionitrile; methyl acetate and ethyl acetate; Esters or an appropriate mixture thereof are used. The reaction temperature is appropriately selected from a range that is known to be usable for a protein bond formation reaction, and is usually appropriately selected from a range of about 120 ° C. to 50 ° C. The activated amino acid derivative is usually used in a 1.5 to 4-fold excess. As a result of the test using the ninhydrin reaction, when the condensation is insufficient, sufficient condensation can be performed by repeating the condensation reaction without removing the protecting group. When a sufficient condensation cannot be obtained even by repeating the reaction, unreacted amino acids can be acetylated using acetic anhydride or acetylimidazole to 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—ペンチルォキシ カルボニル、 イソポルニルォキシカルポニル、 4ーメトキシベンジルォキシカル ポニル、 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二ル、 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, isopolnyoxycarbonyl, 4-methoxybenzyloxycarbonyl, C11Z, Br-Z, and adaman. Tyloxycarbonyl, trifluoroacetyl, phthaloyl, formyl, 2-nitrophenylsulfenyl, diphenylphosphinothioyl, Fmoc and the like are used.
カルボキシル基は、 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—ブチル、 シクロペンチル、 シクロへキシル、 シクロヘプ 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ァ 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— ニトロべンジルエステル、 4—メトキシベンジルエステル、 4—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 シカルボニルヒドラジド化、 t—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 carboxyl group may be, for example, alkyl-esterified (eg, methyl, ethyl, propyl, butyl, t-butyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, 2-adamantyl, etc.). Alkyl esterification), aralkyl esterification (eg, benzyl ester, 4-nitrobenzyl ester, 4-methoxybenzyl ester, 4-chlorobenzyl ester, benzhydryl esterification), phenacyl esterification, benzyloxycarbonyl It can be protected by hydrazide, t-butoxycarbonyl hydrazide, trityl hydrazide and the like.
セリンの水酸基は、 例えば、 エステル化またはエーテル化によって保護するこ とができる。 このエステル化に適する基としては、 例えば、 ァセチル基などの低 級 ((: アルカノィル基、 ベンゾィル基などのァロイル基、 ベンジルォキシ カルポニル基、 エトキシカルポニル基などの炭酸から誘導される基などが用いら れる。 また、 ェ一テル化に適する基としては、 例えば、 ベンジル基、 テトラヒド ロピラニル基、 t-ブチル基などである。 The hydroxyl group of serine can be protected, for example, by esterification or etherification. Examples of a group suitable for the esterification include a lower group such as an acetyl group ((: an aroyl group such as an alkanoyl group and a benzoyl group, a group derived from carbonic acid such as a benzyloxycarbonyl group and an ethoxycarbonyl group). It is. Examples of groups 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 し C 12- B z K 2_ニトロベンジル、 B r— Z、 t一プチルなどが用いられる。 The protecting group of the phenolic hydroxyl group of tyrosine, for example, Bz and C 1 2 - B z K 2_ nitrobenzyl, B r- Z, such as t one-heptyl is used.
ヒスチジンのイミダゾールの保護基としては、 例えば、 To s、 4—メトキシ —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、 T r t、 Fmo cなどが用いられる。  As the protecting group for imidazole of histidine, for example, Tos, 4-methoxy-2,3,6-trimethylbenzenesulfonyl, DNP, benzyloxymethyl, Bum, Boc, Trt, Fmoc and the like are used.
原料の力ルポキシル基の活性化されたものとしては、 例えば、 対応する酸無水 物、 アジド、 活性エステル 〔アルコール (例えば、 ペン夕クロ口フエノール、 2 , 4, 5—トリクロ口フエノール、 2, 4—ジニトロフエノール、 シァノメチル アルコール、 パラニトロフエノ一ル、 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、 対応するリン酸アミドが用い られる。  Examples of the activated form of the carboxylic group of the raw material include, for example, corresponding acid anhydrides, azides, and active esters [alcohols (eg, phenol, 2,4,5-trichlorophenol, 2,4 —Dinitrophenol, cyanomethyl alcohol, paranitrophenol, HONB, N-hydroxysuccinimide, N-hydroxyphthalimide, ester with HOB t)]. As the activated amino group of the raw material, for example, a corresponding phosphoric amide is used.
保護基の除去 (脱離) 方法としては、 例えば、 Pd—黒あるいは Pd—炭素な どの触媒の存在下での水素気流中での接触還元や、 また、 無水フッ化水素、 メタ ンスルホン酸、 トリフルォロメ夕ンスルホン酸、 トリフルォロ酢酸あるいはこれ らの混合液などによる酸処理や、 ジィソプロピルェチルァミン、 トリェチルアミ ン、 ピぺリジン、 ピぺラジンなどによる塩基処理、 また液体アンモニア中ナトリ ゥムによる還元なども用いられる。 上記酸処理による脱離反応は、 一般に約一 2 0°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、 チオア二ソール、 メタクレゾール、 パラクレゾ一ル、 ジメチルスル フイド、 1, 4—ブタンジチオール、 1, 2—エタンジチオールなどのような力 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 、 トリプトファンのィンドール保護基として用いられるホルミル基は上記の 1, 2—エタンジチオール、 1, 4一ブタンジチオールなどの存在下の酸処理による 脱保護以外に、 希水酸化ナトリウム溶液、 希アンモニアなどによるアルカリ処理 によっても除去される。 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。 Methods for removing (eliminating) protecting groups include, for example, catalytic reduction in a stream of hydrogen in the presence of a catalyst such as Pd-black or Pd-carbon, or hydrogen fluoride anhydride, methanesulfonic acid, or trifluoromethane. 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 Also used. The elimination reaction by the above-mentioned acid treatment is generally performed at a temperature of about 120 ° C to 40 ° C. In the acid treatment, for example, anisol, phenol, thioanisole, methacresol, paracresol, dimethylsulfur It is effective to add a force-thione scavenger such as FID, 1,4-butanedithiol, 1,2-ethanedithiol, etc. In addition, the 2,4-dinitrophenyl group used as an imidazole protecting group of histidine is removed by thiophenol treatment, and the formyl group used as an indole protecting group of tryptophan is the above 1,2-ethanedithiol, 1,4-1. In addition to deprotection by acid treatment in the presence of butanedithiol, etc., it is also removed by alkali treatment with dilute sodium hydroxide solution, dilute ammonia, etc. The protection of the functional group which should not be involved in the reaction of the raw materials, the protecting 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、 カルボキシ末端アミノ酸のひ一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、 該ぺプ チド鎖の N末端の α—アミノ基の保護基のみを除いたタンパク質または部分ぺプ チドと C末端のカルボ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べ 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ペ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 タンパク質またはペ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, after amidating and protecting a lipoxyl group of a carboxy terminal amino acid, a peptide (protein) chain having a desired chain length on the amino group side is obtained. And a protein or partial peptide from which only the protecting group at the N-terminal α-amino group of the peptide chain has been removed and a protein or partial peptide from which only the protecting group at the C-terminal carboxyl group has been removed. Are produced, and these proteins or peptides are condensed in a mixed solvent as described above. Details of the condensation reaction are the same as described above. After purifying the protected protein or peptide obtained by the condensation, all the protecting groups are removed by the above-mentioned method, and a desired crude protein or peptide can be obtained. The crude protein or peptide is purified by using various known purification means, and the main fraction is lyophilized to obtain an amide of the desired protein or peptide.
タンパク質またはペプチドのエステル体を得るには、 例えば、 カルボキシ末端 アミノ酸のひ—カルボキシル基を所望のアルコール類と縮合しアミノ酸エステル とした後、 タンパク質またはペプチドのアミド体と同様にして、 所望のタンパク 質またはべプチドのエステル体を得ることができる。  To obtain an ester of a protein or peptide, for example, after condensing the carboxyl group of the carboxy terminal amino acid with a desired alcohol to form an amino acid ester, the desired protein can be obtained in the same manner as the amide of a protein or peptide. Alternatively, an ester of a peptide can be obtained.
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は、 公知のペプチドの 合成法に従って、 あるいは本発明のタンパク質を適当なぺプチダ一ゼで切断する ことによって製造することができる。 ペプチドの合成法としては、 例えば、 固相 合成法、 液相合成法のいずれによっても良い。 すなわち、 本発明の部分ペプチド 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ぺ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ば、 例えば、 以下の①〜⑤に 記載された方法が挙げられる。  The protein or 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 peptidase. As a method for synthesizing a peptide, for example, any of a solid phase synthesis method and a liquid phase synthesis method may be used. That is, the desired peptide can be produced by condensing a partial peptide or amino acid capable of constituting the partial peptide of the present invention with the remaining portion, and if the product has a protective group, removing the protective group to produce the desired peptide. Can be. Known condensation methods and elimination of protecting groups include, for example, the methods described in the following ① to ⑤.
ΦΜ. Bodanszkyおよび M. A. Onde t t i、 ペプチド ·シンセシス (Pept ide Synthe s is) , Interscience Publ ishers, New York (1966年) ② Sdiroederおよび Luebke、 ザ ·ペプチド(The Peptide), Academic Press, New York (1965年) ΦΜ. Bodanszky and MA Ondetti, Peptide Synthesis, Interscience Publ ishers, New York (1966) ② Sdiroeder and Luebke, The Peptide, Academic Press, New York (1965)
③泉屋信夫他、 ペプチド合成の基礎と実験、 丸善 (株) (1975年)  (3) Nobuo Izumiya et al. Basics and experiments on peptide synthesis, Maruzen Co., Ltd. (1975)
④矢島治明 および榊原俊平、 生化学実験講座 1、 タンパク質の化学 IV、 205、 ( 1977年)  治 Haruaki Yajima and Shunpei Sakakibara, Biochemistry Laboratory Lecture 1, Protein Chemistry IV, 205, (1977)
⑤矢島治明監修、 続医薬品の開発、 第 14巻、 ペプチド合成、 広川書店  治 Supervised by Haruaki Yajima, Development of Continuing Drugs, Volume 14, Peptide Synthesis, Hirokawa Shoten
また、 反応後は通常の精製法、 例えば、 溶媒抽出 ·蒸留 ·カラムクロマトダラ フィー ·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で用いられ る部分べプチドを精製単離することができる。 上記方法で得られるタンパク質ま たは部分べプチドが遊離体である場合は、 公知の方法あるいはそれに準じる方法 によって適当な塩に変換することができるし、 逆に塩で得られた場合は、 公知の 方法あるいはそれに準じる方法によって遊離体または他の塩に変換することがで さる。  After the reaction, the partial peptide used in the present invention can be purified and isolated by a combination of ordinary purification methods such as solvent extraction, distillation, column chromatography, liquid chromatography, and recrystallization. When the protein or partial peptide obtained by the above method is a free form, it can be converted to an appropriate salt by a known method or a method analogous thereto, and conversely, when the protein or the partial peptide is obtained by a salt, it is known. It can be converted to the free form or other salts by the method of or a method analogous thereto.
本発明のタ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 のタンパク質をコ一ドする塩基配列 (DNAまたは RNA、 好ましくは DNA) 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 DNA、 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 base sequence (DNA or RNA, preferably DNA) encoding the protein of the present invention. . 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. If single-stranded, it may be the sense strand (ie, the coding strand) or the antisense strand (ie, the 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、 公知の方法、 例 えば、 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。  Using a polynucleotide encoding the protein of the present invention, the method described in a known method, for example, “Experimental Medicine Special Edition,“ New PCR and Its Application ”15 (7), 1997, or a method analogous thereto, is used. The mRNA of the protein of the invention can be quantified.
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 c DNA、 前記した細胞 '組織由来の c DNAライブラリ一、 合成 DNAのいずれ でもよい。 The DNA encoding the protein of the present invention may be any DNA as long as it contains the above-described nucleotide sequence encoding the protein of the present invention. In addition, any of genomic DNA, genomic DNA library, the above-mentioned cells, tissue-derived cDNA, the above-mentioned cell 組織 tissue-derived cDNA library, and synthetic DNA May be.
ライブラリー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、 ファージミドなどいずれであってもよい。 また、 前記した細胞 ·組織より total R N Aまたは mRN A画分を調製したものを用いて直接 Reverse Transcrip tase Polymerase Chain React ion (以下、 R T— P C R法と略称する) によって 増幅することもできる。  The vector used for the library may be any of bacteriophage, plasmid, cosmid, phagemid and the like. Alternatively, amplification can be directly performed by reverse transcription polymerase chain reaction (hereinafter abbreviated as RT-PCR method) using a total RNA or mRNA fraction prepared from the cells and tissues described above.
本発明のタンパク質をコードする D N Aとしては、 例えば、 配列番号: 2で表 される塩基配列を含有する D NA、 または配列番号: 2で表される塩基配列を有 する D NA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 D N Aを含有 し、 本発明のタンパク質と実質的に同質の性質を有するタンパク質をコードする D N Aであれば何れのものでもよい。  Examples of the DNA encoding the protein of the present invention include a DNA having the nucleotide sequence represented by SEQ ID NO: 2 or a DNA having the nucleotide sequence represented by SEQ ID NO: 2 and a high stringency. Any DNA may be used as long as it contains DNA that hybridizes under the conditions and encodes a protein having substantially the same properties as the protein of the present invention.
配列番号: 2で表される塩基配列を含有する D NA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 D NAとしては、 例えば、 配列番号: 2で表され る塩基配列と約 9 7 %以上、 好ましくは約 9 8 %以上、 さらに好ましくは約 9 9 %以上の相同性を有する塩基配列を含有する D N Aなどが用いられる。  Examples of a DNA that can hybridize under high stringent conditions with a DNA containing the nucleotide sequence represented by SEQ ID NO: 2 include, for example, about 97% or more of the nucleotide sequence represented by SEQ ID NO: 2, Preferably, a DNA containing a base sequence having a homology of about 98% or more, more preferably about 99% or more is used.
ハイプリダイゼーシヨンは、 公知の方法あるいはそれに準じる方法、 例えば、 モレキュラー ·クローニング (Molecular Cloning) 2 n d (J. Sambrook et al ., Cold Spring Harbor Lab. Press, 1989) に記載の方法などに従って行なうこ とができる。 また、 市販のライブラリーを使用する場合、 添付の使用説明書に記 載の方法に従って行うことができる。 より好ましくは、 ハイストリンジェン卜な 条件に従って行うことができる。  Hybridization can be performed according to a known method or a method analogous thereto, for example, the method described in Molecular Cloning 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). Can be. 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 stringent conditions.
ハイストリンジェン卜な条件とは、 例えば、 ナトリウム濃度が約 1 9〜4 0 m M、 好ましくは約 1 9〜 2 0 mMで、 温度が約 5 0〜 7 0 ° (:、 好ましくは約 6 0 〜6 5 の条件を示す。 特に、 ナトリウム濃度が約 1 9 mMで温度が約 6 5 °Cの 場合が最も好ましい。  High stringent conditions include, for example, a sodium concentration of about 19 to 40 mM, preferably about 19 to 20 mM, and a temperature of about 50 to 70 ° (: The conditions are from 0 to 65. Particularly preferred is a sodium concentration of about 19 mM and a temperature of about 65 ° C.
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 D NAとして、 配列番号: 2で表される塩基配列を含有する D NA などが用いられる。  More specifically, as the DNA encoding the protein containing the amino acid sequence represented by SEQ ID NO: 1, a DNA containing the base sequence represented by SEQ ID NO: 2 or the like is used.
本発明の部分ペプチドをコードする D NAとしては、 前記した本発明の部分べ プチドをコードする塩基配列を含有する DN A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 c DNAライブラリー、 合成 DNAの いずれでもよい。 Examples of the DNA encoding the partial peptide of the present invention include the aforementioned partial peptide of the present invention. Any DNA containing a nucleotide sequence encoding the peptide may be used. Further, any of genomic DNA, genomic DNA library, cDNA derived from the above-described cells and tissues, cDNA library derived from the above-described cells and tissues, and synthetic DNA may be used.
本発明の部分ペプチドをコードする DNAとしては、 例えば、 配列番号: 2で 表される塩基配列を含有する D N Aの部分塩基配列を有する D N A、 または配列 番号: 2で表される塩基配列を含有する DN A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 DNAを有し、 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と実質的に同質の活性を有するタンパク質をコードする DN Aの部分塩基配列を有する D N Aなどが用いられる。  Examples of the DNA encoding the partial peptide of the present invention include a DNA having a partial base sequence of DNA containing the base sequence represented by SEQ ID NO: 2, or a base sequence represented by SEQ ID NO: 2 A partial nucleotide sequence of a DNA encoding a protein having a DNA that hybridizes with DNA under high stringent conditions and having substantially the same activity as a protein containing the amino acid sequence represented by SEQ ID NO: 1 DNA or the like is used.
配列番号: 2で表される塩基配列を含有する DNAとハイストリンジェントな 条件下でハイプリダイズできる DNAとしては、 例えば配列番号: 2で表される 塩基配列と約 97%以上、 好ましくは約 98%、 より好ましくは約 99%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 DNA containing the nucleotide sequence represented by SEQ ID NO: 2 under high stringent conditions include, for example, about 97% or more, preferably about 98%, of the nucleotide sequence represented by SEQ ID NO: 2 %, More preferably a DNA containing a base sequence having a homology of about 99% or more is used.
より具体的には、 本発明の部分ペプチドをコードする DNAとすれば、 配列番 号: 1で表されるアミノ酸配列中、 ① N末端から第 53番目 (G 1 u) 〜第 55 番目 (Ty r) のアミノ酸残基からなる部分アミノ酸配列をコードする塩基配列 (例、 配列番号: 2で表される塩基配列中、 5'.末端から第 157番目から第 1 65番目の部分塩基配列 (配列番号: 18) ) 、 ② N末端から第 131番目 (A r g) 〜第 134番目 (A l a) のアミノ酸残基からなる部分アミノ酸配列をコ ードする塩基配列 (例、 配列番号: 2で表される塩基配列中、 5' 末端から第 3 91番目から第 402番目の部分塩基配列 (配列番号: 19) ) 、 ③ N末端から 第 164番目 (A 1 a) 〜第 165番目 (P r o) のアミノ酸残基からなる部分 アミノ酸配列をコードする塩基配列 (例、 配列番号: 2で表される塩基配列中、 5' 末端から第 490番目から第 495番目の部分塩基配列 (配列番号: 20) ) 、 ④ N末端から第 266番目 (A 1 a) 〜第 268番目 (A 1 a) のアミノ酸 残基からなる部分アミノ酸配列をコードする塩基配列 ( (例、 配列番号: 2で表 される塩基配列中、 5 ' 末端から第 796番目から第 804番目の部分塩基配列 (配列番号: 21) ) 、 および⑤ N末端から第 277番目 (G 1 n) 〜第 282 番目 (A l a) のアミノ酸残基からなる部分アミノ酸配列をコードする塩基配列 (例、 配列番号: 2で表される塩基配列中、 5' 末端から第 829番目から第 8 46番目の部分塩基配列 (配列番号: 22) ) から選ばれる塩基配列のいずれか または複数 (2〜5) 個を有する前記した本発明のタンパク質の部分ペプチドを コードする DNAなどがあげられる。 More specifically, assuming that the DNA encodes the partial peptide of the present invention, in the amino acid sequence represented by SEQ ID NO: 1, (1) 53rd (G1u) to 55th (Ty r) the base sequence encoding the partial amino acid sequence consisting of the amino acid residues (eg, in the base sequence represented by SEQ ID NO: 2, the partial base sequence from the 157th to the 165th from the 5 'end (sequence No .: 18)), ② Nucleotide sequence encoding the partial amino acid sequence consisting of the 131st (Arg) to 134th (Ala) amino acid residues from the N-terminus (eg, SEQ ID NO: 2 391st to 402th partial nucleotide sequence from the 5 'end (SEQ ID NO: 19)), ③ 164th (A1a) to 165th (Pro) from the N-terminal A nucleotide sequence encoding an amino acid sequence (eg, 5 ′ in the nucleotide sequence represented by SEQ ID NO: 2) 490th to 495th partial base sequence from the end (SEQ ID NO: 20)), 部分 partial amino acid consisting of 266th (A1a) to 268th (A1a) amino acid residues from the N-terminus A nucleotide sequence encoding the sequence (eg, in the nucleotide sequence represented by SEQ ID NO: 2, a partial nucleotide sequence from 796th to 804th from the 5 'end (SEQ ID NO: 21)); 277th (G1n) to 282nd A nucleotide sequence encoding a partial amino acid sequence consisting of the amino acid residue at position (A la) (eg, in the nucleotide sequence represented by SEQ ID NO: 2, the 829th to 846th partial nucleotide sequence from the 5 'end) (SEQ ID NO: 22)) and a DNA encoding a partial peptide of the protein of the present invention having any one or a plurality (2 to 5) of base sequences selected from the group consisting of:
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トな条件は前記と同 様のものが用いられる。  Hybridization methods and high stringency conditions are the same as described above.
本発明のタンパク質または本発明の部分ペプチド (以下、 これらをコードする DN Aのクローニングおよび発現の説明においては、 これらを単に本発明のタン パク質と略記する場合がある) を完全にコードする DNAのクローエングの手段 としては、 本発明のタンパク質をコードする塩基配列の一部分を有する合成 DN Aプライマーを用いて PC R法によって増幅するか、 または適当なベクターに組 み込んだ DN Aを本発明のタンパク質の一部または全領域をコードする DN A断 片もしくは合成 DNAを用いて標識したものとのハイブリダィゼーシヨンによつ て選別することができる。 ハイブリダィゼーシヨンの方法は、 例えば、 モレキュ ラー -クローニング (Molecular Cloning) 2nd (J. Sambrook et al. , Cold Spring Harbor Lab. Press, 1989) に記載の方法などに従って行なうことができ る。 また、 市販のライブラリーを使用する場合、 添付の使用説明書に記載の方法 に従って行なうことができる。  DNA that completely encodes the protein of the present invention or the partial peptide of the present invention (hereinafter, in the description of the cloning and expression of DNAs encoding them, these may be simply abbreviated as the protein of the present invention). Cloning means may be amplified by the PCR method using a synthetic DNA primer having a part of the nucleotide sequence encoding the protein of the present invention, or the DNA incorporated into an appropriate vector may be used as the DNA of the present invention. Selection can be performed by hybridization with a DNA fragment encoding a part or the whole region of the protein or labeled with a synthetic DNA. Hybridization can be carried out, for example, according to the method described in Molecular-Cloning (Molecular Cloning) 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). When a commercially available library is used, it can be performed according to the method described in the attached instruction manual.
DNAの塩基配列の変換は、 PCRや公知のキット、 例えば、 Mu t anTM -s u e r Exp r e s s Km (宝酒造) 、 Mu t a n™-K (宝酒造) などを用いて、 〇DA— LA P CR法、 Gapp e d du l e x法、 Ku n k e 1法など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 using PCR or a known kit, for example, Mutan -suer Expression Km (Takara Shuzo), Mutan ™ -K (Takara Shuzo), etc. The method can be carried out according to a known method such as the Gap ed dulex method or the Knuke 1 method or a method analogous thereto.
クローン化されたタンパク質をコードする DNAは目的によりそのまま、 また は所望により制限酵素で消化したり、 リンカーを付加して使用することができる 。 該 DNAはその 5, 末端側に翻訳開始コドンとしての ATGを有し、 また 3, 末端側には翻訳終止コドンとしての TAA、 TGAまたは TAGを有していても よい。 これらの翻訳開始コドンや翻訳終止コドンは、 適当な合成 DNAアダプタ 一を用いて付加することもできる。 The DNA encoding the cloned protein can be used as it is depending on the purpose, or can be used by digesting with a restriction enzyme or adding a linker if desired. The DNA may have ATG as a translation initiation codon on its 5, terminal side, and may have TAA, TGA or TAG as a translation termination codon on its 3, terminal side. These translation start codons and translation stop codons are It can also be added using one.
本発明のタンパク質の発現ベクターは、 例えば、 (ィ) 本発明のタンパク質を コードする DNAから目的とする DNA断片を切り出し、 (口) 該 DNA断片を 適当な発現ベクター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 for the protein of the present invention includes, for example, (a) cutting out a DNA fragment of interest from DNA encoding the protein of the present invention, and (mouth) ligating the DNA fragment downstream of a promoter in an appropriate expression vector. It can be manufactured by
ベクターとしては、 大腸菌由来のプラスミド (例、 pBR322, pBR 32 5, pUC 12, pUC 13) 、 枯草菌由来のプラスミド (例、 pUB 110, TP 5, pC 194) 、 酵母由来プラスミド (例、 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, ワクシニアウィルス , バキュロウィルスなどの動物ウィルスなどの他、 pAl— 11、 XT 1, ρ Rc/CMV, p Rc/RS V, p c D NA I ZN e oなどが用いられる。 本発明で用いられるプロモータ一としては、 遺伝子の発現に用いる宿主に対応 して適切なプロモーターであればいかなるものでもよい。 例えば、 動物細胞を宿 主として用いる場合は、 SRaプロモーター、 SV40プロモーター、 LTRプ 口モーター、 CMVプロモーター、 HS V-TKプロモーターなどが挙げられる これらのうち、 CMV (サイトメガロウィルス) プロモーター、 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 ρプロモーター、 l a cプロモーター、 r e cAプロモーター、 APLプロモー ター、 1 p pプロモーター、 T 7プロモ一夕一などが、 宿主がバチルス属菌であ る場合は、 SPO lプロモー夕一、 SPO 2プロモーター、 p enPプロモータ 一など、 宿主が酵母である場合は、 PH05プロモ一夕一、 PGKプロモ一夕一 、 GAPプロモー夕一、 ADHプロモータ一などが好ましい。 宿主が昆虫細胞で ある場合は、 ポリヘドリンプロモーター、 P 10プロモーターなどが好ましい。 発現ベクターには、 以上の他に、 所望によりェンハンサー、 スプライシングシ ダナル、 ポリ A付加シグナル、 選択マーカー、 SV40複製オリジン (以下、 S V40 o r iと略称する場合がある) などを含有しているものを用いることがで きる。 選択マーカ一としては、 例えば、 ジヒドロ葉酸還元酵素 (以下、 dh f r と略称する場合がある) 遺伝子 〔メソトレキセート (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 orと略称する場合がある、 G41 8耐性) 等が挙げられる。 特に、 dh f r遺伝子欠損チャイニーズハムスター細胞を用いて dh f r遺伝子 を選択マ一力一として使用する場合、 目的遺伝子をチミジンを含まない培地によ つても選択できる。 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 (eg, pSH 19, pSH15), bacteriophages such as λ phage, animal viruses such as retrovirus, vaccinia virus, baculovirus, etc., pAl-11, XT1, ρRc / CMV, pRc / RSV, pcD NA I ZN eo or the like is used. The promoter used in the present invention may be any promoter as long as it is appropriate for the host used for gene expression. For example, when animal cells are used as host, SRa promoter, SV40 promoter, LTR motor, CMV promoter, HSV-TK promoter, etc. are mentioned. Among them, CMV (cytomegalovirus) promoter, SRa promoter It is preferable to use one or the like. When the host is Eshierihia genus bacteria, tr [rho promoter, lac promoter, re cA promoter, AP L promoter, if 1 pp promoter, T 7 including promoter Isseki one is, host Ru der Bacillus, When the host is yeast, such as SPOL promoter, SPO2 promoter, penP promoter, etc., PH05 promoter, PGK promoter, GAP promoter, ADH promoter, etc. are preferable. When the host is an insect cell, a polyhedrin promoter, a P10 promoter and the like are preferable. In addition to the above, the expression vector may further include an enhancer, a splicing signal, a polyA addition signal, a selection marker, an SV40 replication origin (hereinafter, sometimes abbreviated as SV40 ori), and the like, if desired. Can be used. Selection markers include, for example, dihydrofolate reductase (hereinafter sometimes abbreviated as dh fr) gene (methotrexate (MTX) resistance), ampicillin Phosphorus resistant gene (hereinafter sometimes abbreviated as Amp 1 "), neomycin resistance gene (hereinafter sometimes abbreviated as Ne o r, G41 8 resistance). In particular, dh fr genetic defects When the dhfr gene is used as a selection method using Chinese hamster cells, the target gene can be selected even on a thymidine-free medium.
また、 必要に応じて、 宿主に合ったシグナル配列を、 本発明のタンパク質の N 端末側に付加する。 宿主がェシヱリヒア属菌である場合は、 P h o A ·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、 《— 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、 MF a ·シグナル配列、 SUC 2 ·シグナル配列など、 宿主が動物 細胞である場合には、 インシュリン ·シグナル配列、 一インターフェロン,シ グナル配列、 抗体分子 ·シグナル配列などがそれぞれ利用できる。  If necessary, a signal sequence suitable for the host is added to the N-terminal side of the protein of the present invention. If the host is Escherichia, Pho A signal sequence, OmpA signal sequence, etc., if the host is Bacillus, 《— amylase signal sequence, subtilisin signal sequence, etc. In the case of yeast, MFa signal sequence, SUC2 signal sequence, etc. In the case of host animal cells, use of insulin signal sequence, one interferon, signal sequence, antibody molecule, signal sequence, etc. it can.
このようにして構築された本発明のタンパク質をコードする DN Aを含有する ベクターを用いて、 形質転換体を製造することができる。  Using the vector containing the DNA encoding the protein of the present invention thus constructed, a transformant can be produced.
宿主としては、 例えば、 ェシエリヒア属菌、 バチルス属菌、 酵母、 昆虫細胞、 昆虫、 動物細胞などが用いられる。  As the host, for example, Escherichia bacteria, Bacillus bacteria, yeast, insect cells, insects, animal cells, and the like are used.
ェシエリヒア属菌の具体例としては、 例えば、 ェシエリヒア 'コリ (Esdieric hia coli) Kl 2 · DH1 〔プロシージングズ.ォブ ·ザ ·ナショナル ·ァカデ ミー 'ォブ 'サイェンシィズ 'ォブ ·ザ ·ユーエスェ一 (Proc. Natl. Acad. Sc i. USA) , 60巻, 1 60 (1 968)〕 , JM103 〔ヌクイレック 'ァシッズ 'リサ—チ, (Nucleic Acids Research) , 9巻, 309 (1 981)〕 , JA2 21 〔ジャーナル ·ォブ ·モレキュラー ·バイオロジー (Journal of Molecular Biology) , 120巻, 51 7 (1978)〕 , HB 101 〔ジャーナル ·ォブ · モレキユラ一 .バイオロジー, 41巻, 459 (1969)〕 , C 600 〔ジエネ ティックス (Genetics) , 39巻, 440 (1954)〕 などが用いられる。  Specific examples of the genus Escherichia include, for example, Esdieric hia coli Kl 2 · DH1 [Procedures of the national academy 'ob' (Proc. Natl. Acad. ScI. USA), Vol. 60, 160 (1968)], JM103 [Nucleic Acids Research], (Nucleic Acids Research), Vol. 9, 309 (1981)], JA2 21 [Journal of Molecular Biology, 120, 517 (1978)], HB 101 [Journal of Molecular Biology, Biology, 41, 459 (1969)] ], C600 [Genetics, 39, 440 (1954)] and the like.
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 ·バイオケミストリー (Journal of Biochemistry) , 95巻, 87 (1 984)〕 などが用いられる。 酵母としては、 例えば、 サッカロマイセス ·セレピシェ (Saccharomyces cere visiae) AH22, AH22R―, NA87 - 11 A, DKD— 5D, 20 B- 12、 シゾサッカロマイセス 'ボンべ (Sdiizosaccharomyces pombe) NCYC 1913, NCYC 2036, ピキア 'パス卜リス (Pichia pastoris) KM 7 1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 (1 984)]. Examples of yeast include, for example, Saccharomyces cerevisiae AH22, AH22R-, NA87-11A, DKD- 5D, 20B-12, Shizosaccharomyces' bomb (Sdiizosaccharomyces pombe) NCYC 1913, NCYC 2036, Pichia 'Pichia pastoris KM71 etc. is used.
昆虫細胞としては、 例えば、 ウィルスが Ac NPVの場合は、 夜盗蛾の幼虫由 来株化細胞 (Spodoptera frugiperda cell; S f細胞) 、 Trichoplusia niの中 腸由来の MGl細胞、 Trichoplusia niの卵由来の High Five™細胞、 Mamestra b rassicae由来の細胞または Estigmena acrea由来の細胞などが用いられる。 ウイ ルスが 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 ·ヴイボ (In Vivo) , 13, 213- 217, (1977)) などが用いられる。  Insect cells include, for example, when the virus is Ac NPV, a cell line derived from the larvae of night moth (Spodoptera frugiperda cell; S f cell); High Five ™ cells, cells derived from Mamestra b rassicae or cells derived from Estigmena acrea are used. When the virus is BmNPV, a cell line derived from silkworm (Bombyx mori N cell; BmN cell) is used. Examples of the Sf cell include Sf9 cell (ATCC CRL1711), Sf21 cell (Vaughn, JL et al., In Vivo, 13, 213-217, (1977)) and the like. Used.
昆虫としては、 例えば、 カイコの幼虫などが用いられる 〔前田ら、 ネイチヤー (Nature) , 315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 ro, チャイニーズハ ムスター細胞 CHO (以下、 CHO細胞と略記) , dh f r遺伝子欠損チヤィニ ーズハムス夕一細胞 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 FL 細胞、 H 9 c 2細胞などが用いられる。  Examples of animal cells include monkey cells COS-7, Vero, Chinese Hamster cells CHO (hereinafter abbreviated as CHO cells), dh fr gene-deficient Chinese Hamster Yuichi cells CHO (hereinafter CHO (dhfr_) cells) Abbreviations), mouse L cells, mouse AtT-20, mouse myeloma cells, rat GH3, human FL cells, H9c2 cells, etc. are used.
ェシエリヒア属菌を形質転換するには、 例えば、 プロシージングズ ·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 ·ユーエスエー ( Proc. Natl. Acad. Sci. USA) , 69巻, 2110 (1972) 、 ジーン (Gene ) , 17巻, 107 (1982)などに記載の方法に従って行なうことができる。 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 ·ジエネティックス (Molecular & General Genetics) , 168巻, 111 ( 1979)などに記載の方法に従って行うことができる。  Transformation of Escherichia sp. Is described, for example, in Proc. Natl. Acad. Sci. USA, Proc. Natl. Acad. Sci. , 2110 (1972) and Gene, 17, 107 (1982). Transformation of Bacillus spp. Can be performed, for example, according to the method described in Molecular & General Genetics, Vol. 168, 111 (1979).
酵母を形質転換するには、 例えば、 メソッズ ·イン ·ェンザィモロジ一 (Meth ods in Enzymology) , 194巻, 182— 187 (1991) 、 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 · ュ一エスェ一 (Proc. Natl. Acad. Sci. USA) , 75巻, 1929 (1978) などに記載の方法に従って行なうことができる。 To transform yeast, see, for example, Methods in Enzymology, 194, 182-187 (1991), According to the method described in, for example, Goods-of-the-National Academy of Things-of-the-Sciences-of-the-Us. (Proc. Natl. Acad. Sci. USA), Vol. 75, 1929 (1978) Can do it.
昆虫細胞または昆虫を形質転換するには、 例えば、 バイオ Zテクノロジー (Bi o/Technology) ,6, 47-55(1988) などに記載の方法に従って行うことができる。 動物細胞を形質転換するには、 例えば、 細胞工学別冊 8 新細胞工学実験プロ トコ一ル. 263— 267 (1995) (秀潤社発行) 、 ヴイロロジー (Virolo gy) , 52巻, 456 (1973)に記載の方法に従って行うことができる。  Insect cells or insects can be transformed, for example, according to the method described in Bio Z Technology (Bio / Technology), 6, 47-55 (1988). In order to transform animal cells, for example, Cell Engineering Separate Volume 8 New Cell Engineering Experiment Protocol. 263—267 (1995) (published by Shujunsha), Virology, 52, 456 (1973) Can be performed according to the method described in (1).
このようにして、 タンパク質をコードする DNAを含有する発現ベクターで形 質転換された形質転換体を得ることができる。  Thus, a transformant transformed with the expression vector containing the DNA encoding the protein can be obtained.
宿主がェシエリヒア属菌、 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 · リカー、 ペプトン 、 カゼイン、 肉エキス、 大豆粕、 バレイショ抽出液などの無機または有機物質、 無機物としては、 例えば、 塩化カルシウム、 リン酸二水素ナトリウム、 塩化マグ ネシゥムなどが挙げられる。 また、 酵母エキス、 ビタミン類、 生長促進因子など を添加してもよい。 培地の; Hは約 5〜 8が望ましい。  When culturing a transformant whose host is a bacterium belonging to the genus Escherichia or Bacillus, a liquid medium is suitable as the medium used for the culturing, and a carbon source necessary for the growth of the transformant is contained therein. , Nitrogen sources, inorganic substances and others. Examples of the carbon source include glucose, dextrin, soluble starch, and sucrose. Examples of the nitrogen source include ammonium salts, nitrates, corn chip liquor, peptone, casein, meat extract, soybean meal, potato extract, and the like. Examples of the inorganic or organic substance and the inorganic substance include calcium chloride, sodium dihydrogen phosphate, magnesium chloride and the like. In addition, yeast extract, vitamins, growth promoting factors and the like may be added. The H of the medium is preferably about 5-8.
ェシエリヒア属菌を培養する際の培地としては、 例えば、 グルコース、 カザミ ノ酸を含む M 9培地 〔ミラ一 (Miller) , ジャーナル ·ォブ ·ェクスペリメンッ •イン -モレキユラ一 ·ジエネティックス (Journal of Experiments in Molecu lar Genetics) , 431— 433, Cold Spring Harbor Laboratory, New York 1972〕 が好ましい。 ここに必要によりプロモーターを効率よく働かせるため に、 例えば、 3 —インドリルアクリル酸のような薬剤を加えることができる。 宿主がェシェリヒア属菌の場合、 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。  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 Exp. lar Genetics), 431-433, Cold Spring Harbor Laboratory, New York 1972]. Here, if necessary, a drug such as 3-indolylacrylic acid can be added to make the promoter work efficiently. When the host is a bacterium belonging to the genus Escherichia, cultivation is usually performed at about 15 to 43 ° C for about 3 to 24 hours, and if necessary, aeration and stirring can be applied.
宿主がバチルス属菌の場合、 培養は通常約 30〜 40 °Cで約 6〜 24時間行な レ、、 必要により通気や撹拌を加えることもできる。 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 ·ュ一エス エー (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 CBiUer, G. A. ら、 プロシージングズ-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) , 81巻, 5330 (1984) 〕 カ 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 t〜 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 the host is a bacterium belonging to the genus Bacillus, culturing is usually performed at about 30 to 40 ° C for about 6 to 24 hours, and if necessary, aeration and stirring may be added. When culturing a transformant in which the host is yeast, for example, Burkholder's minimal medium [Bostian, KL, et al., Processings * of the National Academy of Sciences] Proc. Natl. Acad. Sci. USA, 77, 4505 (1980)] and SD medium containing 0.5% casamino acid, CBiUer, GA et al. -The National Academy of Sciences of the USA (Proc. Natl. Acad. Sci. USA), 81, 5330 (1984)]. The pH of the medium is preferably adjusted to about 5-8. The cultivation is usually performed at about 20 t to 35 ° C for about 24 to 72 hours, and aeration and agitation are added as necessary.
宿主が昆虫細胞または昆虫である形質転換体を培養する際、 培地としては、 Gr ace's Insect Medium (Grace, T. C. ,ネイチヤー (Nature) , 195, 788 (1962)) に非動化した 10 %ゥシ血清等の添加物を適宜加えたものなどが用いられる。 培 地の ρ Ηは約 6. 2〜 6. 4に調整するのが好ましい。 培養は通常約 27 °Cで約 3〜5日間行ない、 必要に応じて通気や撹拌を加える。  When culturing an insect cell or a transformant in which the host is an insect, the culture medium used was a 10% strain immobilized in Grace's Insect Medium (Grace, TC, Nature, 195, 788 (1962)). Those to which additives such as serum are appropriately added are used. It is preferable to adjust the ρ 培 of the culture medium 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卷, 501 (1952)] , DMEM培地 〔ヴイロロジ一 (Virology) , 8巻, 396 (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 Associ at ion) 199巻, 519 (1967)〕 , 199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 (Proceeding of the Society for the Biological Medicine) , 73巻, 1 (1950)〕 などが 用いられる。 pHは約 6〜8であるのが好ましい。 培養は通常約 30 〜 40°C で約 15〜60時間行ない、 必要に応じて通気や撹拌を加える。  When culturing a transformant in which the host is an animal cell, examples of the medium include a MEM medium containing about 5 to 20% fetal bovine serum [Science, 122, 501 (1952)], a DMEM medium [Virology, 8, 396 (1959)], RPM I 1640 medium [The Journal of the American Medical Association at ion, 199, 519 (1967) )], And 199 medium [Proceding of the Society for the Biological Medicine, Vol. 73, 1 (1950)]. Preferably, the pH is about 6-8. Cultivation is usually carried out at about 30 to 40 ° C for about 15 to 60 hours, and aeration and agitation are added as necessary.
以上のようにして、 形質転換体の細胞質内または細胞外に本発明のタンパク質 を生成せしめることができる。  As described above, the protein of the present invention can be produced in the cytoplasm or extracellularly 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When extracting the protein of the present invention from cultured cells or cells, The cells or cells are collected by a known method, suspended in an appropriate buffer, and
、 リゾチームおよび Zまたは凍結融解などによって菌体あるいは細胞を破壊した のち、 遠心分離やろ過によりタンパク質の粗抽出液を得る方法などが適宜用いら れる。 緩衝液の中に尿素や塩酸グァニジンなどのタンパク質変性剤や、 トリトン X - 1 0 0 TMなどの界面活性剤が含まれていてもよい。 培養液中にタンパク質 が分泌される場合には、 培養終了後、 公知の方法で菌体あるいは細胞と上清とを 分離し、 上清を集める。 After the cells or cells are disrupted by lysozyme, Z or freeze-thawing, a method of obtaining a crude protein extract by centrifugation or filtration is used as appropriate. The buffer may contain a protein denaturant such as urea or guanidine hydrochloride, or a surfactant such as Triton X-100 . When the protein is secreted into the culture solution, after completion of the culture, the supernatant is separated from the cells or cells by a 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 S D S—ポリアクリルアミドゲル電気 泳動法などの主として分子量の差を利用する方法、 イオン交換クロマトグラフィ 一などの荷電の差を利用する方法、 ァフィ二ティ一クロマトグラフィーなどの特 異的親和性を利用する方法、 逆相高速液体ク口マトグラフィ一などの疎水性の差 を利用する方法、 等電点電気泳動法などの等電点の差を利用する方法などが用い られる。  Purification of the protein contained in the culture supernatant or extract obtained in this manner can be performed by appropriately combining known separation and purification methods. These known separation and purification methods include methods utilizing solubility such as salting-out and solvent precipitation, dialysis, ultrafiltration, gel filtration, and SDS-polyacrylamide gel electrophoresis, mainly molecular weight. Method using difference in charge, such as ion exchange chromatography, method using specific affinity such as affinity chromatography, hydrophobicity such as reversed-phase high-performance liquid chromatography, etc. A method using a difference in gender, a method using 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。  When the protein thus obtained is obtained as a free form, it can be converted to a salt by a known method or a method analogous thereto, and conversely, when the protein is obtained as a salt, a known method or analogous method Depending on the method, it can be converted into a free form or another salt.
なお、 組換え体が産生するタンパク質を、 精製前または精製後に適当なタンパ ク質修飾酵素を作用させることにより、 任意に修飾を加えたり、 ポリペプチドを 部分的に除去することもできる。 タンパク質修飾酵素としては、 例えば、 トリプ シン、 キモトリブシン、 アルギニルエンドべプチダーゼ、 プロテインキナーゼ、 グリコシダーゼなどが用いられる。  The protein produced by the recombinant can be arbitrarily modified or the polypeptide can be partially removed by the action of an appropriate protein-modifying enzyme before or after purification. As the protein modifying enzyme, for example, trypsin, chymotrypsin, arginyl endopeptidase, protein kinase, glycosidase and the like are used.
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 さる。  The presence of the protein of the present invention thus produced is determined using a specific antibody.
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、 モノクローナル抗体の何れであってもよい。 Antibodies against the protein or partial peptide of the present invention or a salt thereof are described in Any polyclonal antibody or monoclonal antibody may be used as long as it can recognize the protein or partial peptide or its salt.
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 に従っ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 to the protein of the present invention in the description of the antibody) is obtained by using the protein of the present invention as an antigen. The antibody or antiserum can be produced according to the following method.
〔モノクローナル抗体の作製〕  [Preparation of monoclonal antibody]
(a) モノクローナル抗体産生細胞の作製  (a) Preparation of monoclonal antibody-producing cells
本発明の夕ンパク質は、 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や不完全フロイントアジュバントを投 与してもよい。 投与は通常 2〜 6週毎に 1回ずつ、 計 2〜10回程度行われる。 用いられる温血動物としては、 例えば、 サル、 ゥサギ、 ィヌ、 モルモット、 マウ ス、 ラット、 ヒッジ、 ャギ、 ニヮトリが挙げられるが、 マウスおよびラッ卜が好 ましく用いられる。  The protein of the present invention is administered to a warm-blooded animal at a site where the antibody can be produced by administration to itself or together with a carrier or a diluent. Complete Freund's adjuvant or incomplete Freund's adjuvant may be administered in order to enhance the antibody-producing ability upon administration. Administration is usually performed once every 2 to 6 weeks, for a total of 2 to 10 times. Examples of the warm-blooded animal to be used include monkeys, egrets, dogs, guinea pigs, mice, rats, sheep, goats, and chickens, and mice and rats are preferably used.
モノクローナル抗体産生細胞の作製に際しては、 抗原で免疫された温血動物、 例えばマウスから抗体価の認められた個体を選択し最終免疫の 2〜 5日後に脾臓 またはリンパ節を採取し、 それらに含まれる抗体産生細胞を同種または異種動物 の骨髄腫細胞と融合させることにより、 モノクローナル抗体産生ハイブリドーマ を調製することができる。 抗血清中の抗体価の測定は、 例えば、 後記の標識化夕 ンパク質と抗血清とを反応させたのち、 抗体に結合した標識剤の活性を測定する ことにより行なうことができる。 融合操作は既知の方法、 例えば、 ケ一ラ一とミ ルスタインの方法 〔ネイチヤー (Nature), 256、 495 (1975)) に従い実施するこ とができる。 融合促進剤としては、 例えば、 ポリエチレングリコール (PEG) やセンダイウィルスなどが挙げられるが、 好ましくは PEGが用いられる。 骨髄腫細胞としては、 例えば、 NS— 1、 P 3U1、 S P 2/0, AP— 1な どの温血動物の骨髄腫細胞が挙げられるが、 P 3 U 1が好ましく用いられる。 用 いられる抗体産生細胞 (脾臓細胞) 数と骨髄腫細胞数との好ましい比率は 1 : 1 〜20 : 1程度であり、 PEG (好ましくは PEG 1000〜PEG6000) が 1 0〜8 0 %程度の濃度で添加され、 2 0〜4 0 ° (、 好ましくは 3 0〜3 7 で 1〜 1 0分間ィンキュベートすることにより効率よく細胞融合を'実施できる。 モノクローナル抗体産生ハイプリドーマのスクリーニングには種々の方法が使 用できるが、 例えば、 タンパク質抗原を直接あるいは担体とともに吸着させた固 相 (例、 マイクロプレート) にハイプリドーマ培養上清を添加し、 次に放射性物 質や酵素などで標識した抗免疫グロプリン抗体 (細胞融合に用いられる細胞がマ ウスの場合、 抗マウス免疫グロブリン抗体が用いられる) またはプロテイン Aを 加え、 固相に結合したモノクローナル抗体を検出する方法、 抗免疫グロブリン抗 体またはプロテイン Aを吸着させた固相にハイプリドーマ培養上清を添加し、 放 射性物質や酵素などで標識したタンパク質を加え、 固相に結合したモノクローナ ル抗体を検出する方法などが挙げられる。 When preparing monoclonal antibody-producing cells, a warm-blooded animal immunized with an antigen, for example, an individual with an antibody titer is selected from a mouse, and the spleen or lymph node is collected 2 to 5 days after the final immunization and contained in them. By fusing the antibody-producing cells obtained with myeloma cells of the same or different species, a monoclonal antibody-producing hybridoma can be prepared. The antibody titer in the antiserum can be measured, for example, by reacting the labeled protein described below with the antiserum, and then measuring the activity of the labeling agent bound to the antibody. The fusion operation can be performed according to a known method, for example, the method of Kellar and Milstein (Nature, 256, 495 (1975)). Examples of the fusion promoter include polyethylene glycol (PEG) and Sendai virus, but PEG is preferably used. Examples of myeloma cells include myeloma cells of warm-blooded animals such as NS-1, P3U1, SP 2/0, and AP-1, but P3U1 is preferably used. The preferred ratio between the number of antibody-producing cells (spleen cells) used and the number of myeloma cells used is about 1: 1 to 20: 1, and PEG (preferably PEG 1000 to PEG6000) Is added at a concentration of about 10 to 80%, and incubating at 20 to 40 ° (preferably at 30 to 37 for 1 to 10 minutes) enables efficient cell fusion. Various methods can be used to screen for produced hybridomas. For example, the hybridoma culture supernatant is added to a solid phase (eg, a microplate) on which protein antigens are directly or adsorbed together with a carrier, and then radioactive substances are added. Method to detect monoclonal antibody bound to solid phase by adding anti-immunoglobulin antibody (anti-mouse immunoglobulin antibody is used if the cell used for cell fusion is mouse) or protein A labeled with proteins or enzymes Then, add the hybridoma culture supernatant to the solid phase to which the anti-immunoglobulin antibody or protein A is adsorbed, and use radioactive substances, enzymes, etc. A method of detecting a monoclonal antibody bound to a solid phase by adding a labeled protein may be used.
モノクローナル抗体の選別は、 公知あるいはそれに準じる方法に従って行なう ことができる。 通常 HA T (ヒポキサンチン、 アミノプテリン、 チミジン) を添 加した動物細胞用培地で行なうことができる。 選別および育種用培地としては、 ハイプリドーマが生育できるものならばどのような培地を用いても良い。 例えば 、 1〜2 0 %、 好ましくは 1 0〜2 0 %の牛胎児血清を含む R P M I 1 6 4 0 培地、 1〜 1 0 %の牛胎児血清を含む G I T培地 (和光純薬工業 (株) ) あるい はハイプリドーマ培養用無血清培地 (S F M— 1 0 1、 日水製薬 (株) ) などを 用いることができる。 培養温度は、 通常 2 0〜4 0 °C、 好ましくは約 3 7 °Cであ る。 培養時間は、 通常 5日〜 3週間、 好ましくは 1週間〜 2週間である。 培養は 、 通常 5 %炭酸ガス下で行なうことができる。 ハイプリドーマ培養上清の抗体価 は、 上記の抗血清中の抗体価の測定と同様にして測定できる。  The selection of the monoclonal antibody can be performed according to a known method or a method analogous thereto. Usually, it can be performed in an animal cell culture medium supplemented with HAT (hypoxanthine, aminopterin, thymidine). As a selection and breeding medium, any medium can be used as long as it can grow a hybridoma. For example, RPMI 1640 medium containing 1 to 20%, preferably 10 to 20% fetal bovine serum, GIT medium containing 1 to 10% fetal bovine serum (Wako Pure Chemical Industries, Ltd.) Alternatively, a serum-free medium for hybridoma culture (SFM-101, Nissui Pharmaceutical Co., Ltd.) can be used. The culture temperature is usually from 20 to 40 ° C, preferably about 37 ° C. The culture time is generally 5 days to 3 weeks, preferably 1 week to 2 weeks. The culture can be usually performed under 5% carbon dioxide gas. The antibody titer of the hybridoma culture supernatant can be measured in the same manner as the measurement of the antibody titer in the antiserum described above.
( b ) モノクローナル抗体の精製  (b) Purification of monoclonal antibody
モノクロ一ナル抗体の分離精製は、 公知の方法、 例えば、 免疫グロブリンの分 離精製法 〔例、 塩析法、 アルコール沈殿法、 等電点沈殿法、 電気泳動法、 イオン 交換体 (例、 D E A E ) による吸脱着法、 超遠心法、 ゲルろ過法、 抗原結合固相 あるいはプロティン 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 〔ポリクローナル抗体の作製〕 Monoclonal antibodies can be separated and purified by known methods, for example, immunoglobulin separation and purification methods [eg, salting out, alcohol precipitation, isoelectric point precipitation, electrophoresis, ion exchangers (eg, DEAE) ), Ultracentrifugation, gel filtration, antigen-binding solid phase or specific antibody purification by collecting only the antibody with an active adsorbent such as protein A or protein G, and dissociating the bond to obtain the antibody). It can be carried out (Preparation of polyclonal antibody)
本発明のポリクローナル抗体は、 公知あるいはそれに準じる方法に従って製造 することができる。 例えば、 免疫抗原 (本発明のタンパク質等の抗原) とキヤリ ァ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 antibody of the present invention can be produced according to a known method or a method analogous thereto. For example, a complex of an immunizing antigen (an antigen such as the protein of the present invention) and 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. The antibody can be produced by collecting an antibody-containing substance against the antibody 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ータンパク質との複 合体に関し、 キヤリァータンパク質の種類およびキヤリァ一とハプテンとの混合 比は、 キャリアー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ブリン、 へモシァニン等を重量比でハプテン 1に対し、 約 0 . 1〜2 0、 好ましくは約 1〜5の割合で縮合させる方法が用いられる。 また、 ハプテンとキャリアーの力プリングには、 種々の縮合剤を用いることが できるが、 ダルタルアルデヒドやカルポジイミド、 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卜や不完全フロイントアジュバントを投与してもよい。 投 与は、 通常約 2〜 6週毎に 1回ずつ、 計約 3〜1 0回程度行なわれる。  Regarding the complex of immunizing antigen and carrier protein used for immunizing warm-blooded animals, the type of carrier protein and the mixing ratio of carrier and hapten are determined by the antibody against hapten immunized by cross-linking with the carrier. If it can be efficiently performed, any kind may be crosslinked at any ratio.For example, serum serum albumin, thyroglobulin, hemocyanin, etc. may be used in a weight ratio of about 0 to 1 for hapten. A method of condensing at a rate of 1 to 20, preferably about 1 to 5 is used. Further, various condensing agents can be used for force coupling between the hapten and the carrier. For example, an active ester reagent containing a daltaraldehyde, a carbodiimide, a maleimide active ester, a thiol group or a dithioviridyl group is used. The condensation product is administered to a warm-blooded animal itself or together with a carrier or diluent at a site where antibody production is possible. Complete Freund's adjuvant or incomplete Freund's adjuvant may be administered in order to enhance the antibody-producing ability upon administration. The administration is usually made once every about 2 to 6 weeks, for a total of about 3 to 10 times.
ポリクローナル抗体は、 上記の方法で免疫された温血動物の血液、 腹水など、 好ましくは血液から採取することができる。  The polyclonal antibody can be collected from 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、 上記のモノクローナ ル抗体の分離精製と同様の免疫グロプリンの分離精製法に従って行うことができ る。  The measurement of the polyclonal antibody titer in the antiserum can be performed in the same manner as the measurement of the antibody titer in the antiserum described above. Separation and purification of the polyclonal antibody can be carried out according to the same immunoglobulin separation and purification method as the above-mentioned separation and purification of the monoclonal antibody.
本発明のタンパク質または部分ペプチドをコードする D NA (以下、 アンチセ ンス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) 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 チセンスヌクレオチドとしては、 本発明の D NA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。 DNAs encoding the protein or partial peptide of the present invention (hereinafter, these DNAs are abbreviated as DNAs of the present invention in the description of antisense nucleotides) or are substantially complementary to the DNAs. Antisense nucleotides having a base sequence or a part thereof are complementary or substantially complementary to the DNA of the present invention. Any antisense nucleotide may be used as long as it has a complementary base sequence or a part thereof and has an action capable 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) の全塩基配列または部 分塩基配列と約 9 7 %以上、 好ましくは約 9 8 %以上、 より好ましくは約 9 9 % 以上、 の相同性を有する塩基配列などが挙げられる。 特に、 本発明の D NAの相 補鎖の全塩基配列うち、 本発明のタンパク質の N末端部位をコードする部分の塩 基配列 (例えば、 開始コドン付近の塩基配列など) の相補鎖と約 9 7 %以上、 好 ましくは約 9 8 %以上、 より好ましくは約 9 9 %以上の相同性を有するアンチセ ンスヌクレオチドが好適である。  The nucleotide sequence substantially complementary to the DNA of the present invention is, for example, the entire nucleotide sequence or a partial base 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 nucleotide sequence having a homology of about 97% or more, preferably about 98% or more, and more preferably about 99% or more with the sequence is exemplified. In particular, of the entire base sequence of the complementary chain of the DNA of the present invention, the complementary sequence of the base sequence (for example, the base sequence near the start codon) of the portion encoding the N-terminal site of the protein of the present invention is about 9%. Antisense nucleotides having a homology of 7% or more, preferably about 98% or more, more preferably about 99% or more are suitable.
アンチセンスヌクレオチドは通常、 1 0〜4 0個程度、 好ましくは 1 5〜3 0 個程度の塩基から構成される。  The antisense nucleotide is usually composed of about 10 to 40 bases, preferably about 15 to 30 bases.
ヌクレアーゼなどの加水分解酵素による分解を防ぐために、 ァンチセンスヌク レオチドを構成する各ヌクレオチドのリン酸残基 (ホスフェート) は、 例えば、 ホスホロチォエート、 メチルホスホネート、 ホスホロジチォネートなどの化学修 飾リン酸残基に置換されていてもよい。 これらのアンチセンスヌクレオチドは、 公知の D NA合成装置などを用いて製造することができる。  In order to prevent degradation by hydrolases such as nucleases, the phosphate residues (phosphates) of each nucleotide constituting the antisense nucleotide are, for example, chemically modified phosphates such as phosphorothioate, methylphosphonate, and phosphorodithionate. It may be substituted with a residue. These antisense nucleotides can be produced using a known DNA synthesizer or the like.
本発明に従えば、 本発明のタンパク質遺伝子の複製または発現を阻害すること のできるアンチセンス ·ポリヌクレオチド (核酸) を、 クロ一ン化した、 あるい は決定されたタンパク質をコードする D NAの壌基配列情報に基づき設計し、 合 成しうる。 かかるポリヌクレオチド (核酸) は、 本発明のタンパク質遺伝子の 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 N Aとの相互作用を介して本 発明のタンパク質遺伝子の発現を調節 ·制御することができる。 本発明のタンパ ク質関連 R NA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と特異的にハイブリダィ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 たは核酸とペプチド (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ペプチド (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—べ一スペア · リピート、 5 ' 端非翻訳領域、 ポリペプチド翻訳開始コド ン、 タンパク質コ一ド領域、 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、 および 3 ' 端ヘアピンループは好ましい対象領域として選 択しうるが、 タンパク質遺伝子内の如何なる領域も対象として選択しうる。 According to the present invention, an antisense polynucleotide (nucleic acid) capable of inhibiting the replication or expression of the protein gene of the present invention is cloned or a DNA encoding a determined protein is obtained. It can be designed and synthesized on the basis of the base sequence information. Such a polynucleotide (nucleic acid) can hybridize with the RNA of the protein gene of the present invention, inhibit the synthesis or function of the RNA, or inhibit the interaction with the protein-related RNA of the present invention. Thus, the expression of the protein gene of the present invention can be regulated and controlled. Polynucleotides that are complementary to a selected sequence of a protein-associated RNA of the present invention, and polynucleotides that can specifically hybridize with a protein-associated RNA of the present invention can be used in vivo and in vitro. It is useful for regulating and controlling the expression of a protein gene in a plant, and is also useful for treating or diagnosing a disease or the like. Term "corresponding The term "is" means having homology or being complementary to a specific sequence of nucleotides, base sequences or nucleic acids including genes. The “correspondence” between a nucleotide, base sequence or nucleic acid and a peptide (protein) usually refers to the amino acid of the peptide (protein) under instructions derived from the nucleotide (nucleic acid) sequence or its complement. ing. 5 'end hairpin loop of protein gene, 5' end 6-base spare repeat, 5 'end untranslated region, polypeptide translation start codon, protein code region, ORF translation stop codon, 3' end untranslated The region, the 3 'end parindrome region, and the 3' end hairpin loop may be selected as preferred regions of interest, but any region within the protein gene may be selected.
目的核酸と、 対象領域の少なくとも一部に相補的なポリヌクレオチドとの関係 は、 対象物とハイブリダィズすることができるポリヌクレオチドとの関係は、 「 アンチセンス」 であるということができる。 アンチセンス ·ポリヌクレオチドは 、 2—デォキシー D—リポースを含有しているポリヌクレオチド、 D—リポース を含有しているポリヌクレオチド、 プリンまたはピリミジン塩基の N—ダリコシ ドであるその他のタイプのポリヌクレオチド、 あるいは非ヌクレオチド骨格を有 するその他のポリマー (例えば、 市販の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 (伹し、 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や塩基の付着を許容 する配置をもつヌクレオチドを含有する) などが挙げられる。 それらは、 二本鎖 D NA、 一本鎖 D NA、 二本鎖 R NA、 一本鎖 R NA、 さらに D NA : R NAハ イブリツドであることができ、 さらに非修飾ポリヌクレオチド (または非修飾ォ リゴヌクレオチド) 、 さらには公知の修飾の付加されたもの、 例えば当該分野で 知られた標識のあるもの、 キャップの付いたもの、 メチル化されたもの、 1個以 上の天然のヌクレオチドを類縁物で置換したもの、 分子内ヌクレオチド修飾のさ れたもの、 例えば非荷電結合 (例えば、 メチルホスホネート、 ホスホトリエステ ル、 ホスホルアミデート、 力ルバメートなど) を持つもの、 電荷を有する結合ま たは硫黄含有結合 (例えば、 ホスホロチォエート、 ホスホロジチォエー卜など) を持つもの、 例えばタンパク質 (ヌクレアーゼ、 ヌクレアーゼ ·インヒビター、 トキシン、 抗体、 シグナルペプチド、 ポリ—L一リジンなど) や糖 (例えば、 モ ノサッカライドなど) などの側鎖基を有しているもの、 インターカレント化合物The relationship between the nucleic acid of interest and a polynucleotide complementary to at least a part of the target region can be said to be "antisense" with the polynucleotide capable of hybridizing with the target. Antisense polynucleotides include polynucleotides containing 2-dexoxy D-reports, polynucleotides containing D-reports, other types of polynucleotides that are N-daricosides of purine or pyrimidine bases, Alternatively, other polymers having non-nucleotide backbones (eg, commercially available protein nucleic acids and synthetic sequence-specific nucleic acid polymers) or other polymers containing special bonds (伹, such polymers may be found in DNA or RNA) (Including a nucleotide having a configuration that allows base pairing and base attachment as found)). They can be double-stranded DNA, single-stranded DNA, double-stranded RNA, single-stranded RNA, and even DNA: RNA hybrids, and can be unmodified polynucleotides (or unmodified polynucleotides). (Nucleotides), and also those with known modifications, such as those with labels, caps, methylated, and one or more natural nucleotides known in the art. Substituted, or modified with an intramolecular nucleotide, for example, having an uncharged bond (eg, methylphosphonate, phosphotriester, phosphoramidate, olebamate, etc.), a charged bond or Those having a sulfur-containing bond (eg, phosphorothioate, phosphorodithioate, etc.), such as proteins (nucleases, nucleases / inhibitors) , Toxins, antibodies, signal peptides, poly -L one lysine, etc.) or a sugar (e.g., Mo No.3, etc., which have side chain groups, etc.
(例えば、 ァクリジン、 ソラレンなど) を持つもの、 キレ一ト化合物 (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。 こうした修飾物は、 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、 あるいはェ 一テル、 ァミンなどの官能基に変換されていてよい。 (E.g., acridine, psoralen, etc.), chelate compounds (e.g., metals, radioactive metals, boron, oxidizable metals, etc.), those containing alkylating agents, modification (Eg, a nucleic acid of the type I). Here, the “nucleoside”, “nucleotide” and “nucleic acid” may include not only those containing purine and pyrimidine bases but also those having other modified heterocyclic bases. Such modifications may include methylated purines and pyrimidines, acylated purines and pyrimidines, or other heterocycles. Modified nucleotides and modified nucleotides may also be modified at the sugar moiety, e.g., where one or more hydroxyl groups have been replaced with halogens, aliphatic groups, etc., or functionalities such as ethers, amines, etc. It may be converted to a group.
本発明のアンチセンス ·ポリヌクレオチド (核酸) は、 R NA、 D NA、 ある いは修飾された核酸 (R NA、 D NA) である。 修飾された核酸の具体例として は核酸の硫黄誘導体ゃチォホスフェート誘導体、 そしてポリヌクレオシドアミド やオリゴヌクレオシドアミドの分解に抵抗性のものが挙げられるが、 それに限定 されるものではない。 本発明のアンチセンス核酸は次のような方針で好ましく設 計されうる。 すなわち、 細胞内でのアンチセンス核酸をより安定なものにする、 アンチセンス核酸の細胞透過性をより高める、 目標とするセンス鎖に対する親和 性をより大きなものにする、 そしてもし毒性があるならアンチセンス核酸の毒性 をより小さなものにする。  The antisense polynucleotide (nucleic acid) of the present invention is an RNA, a DNA, or a modified nucleic acid (RNA, DNA). Specific examples of the modified nucleic acid include, but are not limited to, sulfur derivatives of nucleic acids, thiophosphate derivatives, and those resistant to degradation of polynucleoside amides and oligonucleoside amides. The antisense nucleic acid of the present invention can be preferably designed according to the following policy. That is, to make the antisense nucleic acid more stable in the cell, to increase the cell permeability of the antisense nucleic acid, to have a greater affinity for the target sense strand, and to be more toxic if it is toxic. Make sense nucleic acid less toxic.
こうして修飾は当該分野で数多く知られており、 例えば J. Kawakami et al. , Pharm Tech Japan, Vol. 8, pp. 247, 1992 ; Vol. 8, pp. 395, 1992 ; S. T. Cro oke et al. ed., Ant i sense Research and Appl icat ions, CRC Press, 1993 な どに開示がある。  Thus, many modifications are known in the art, for example, J. Kawakami et al., Pharm Tech Japan, Vol. 8, pp. 247, 1992; Vol. 8, pp. 395, 1992; ST Crooke et al. ed., Ant isense Research and Applicat ions, CRC Press, 1993.
本発明のアンチセンス核酸は、 変化せしめられたり、 修飾された糖、 塩基、 結 合を含有していて良く、 リボゾーム、 ミクロスフエアのような特殊な形態で供与 されたり、 遺伝子治療により適用されたり、 付加された形態で与えられることが できうる。 こうして付加形態で用いられるものとしては、 リン酸基骨格の電荷を 中和するように働くポリリジンのようなポリカチォン体、 細胞膜との相互作用を 高めたり、 核酸の取込みを増大せしめるような脂質 (例えば、 ホスホリピド、 コ レステロールなど) といった粗水性のものが挙げられる。 付加するに好ましい脂 質としては、 コレステロールやその誘導体 (例えば、 コレステリルクロ口ホルメ ート、 コール酸など) が挙げられる。 こうしたものは、 核酸の 3 ' 端あるいは 5 ' 端に付着させることができ、 塩基、 糖、 分子内ヌクレオシド結合を介して付着 させることができうる。 その他の基としては、 核酸の 3 ' 端あるいは 5 ' 端に特 異的に配置されたキャップ用の基で、 ェキソヌクレアーゼ、 R N a s eなどのヌ クレアーゼによる分解を阻止するためのものが挙げられる。 こうしたキャップ用 の基としては、 ポリエチレングリコール、 テトラエチレンダリコールなどのダリ コールをはじめとした当該分野で知られた水酸基の保護基が挙げられるが、 それ に限定されるものではない。 The antisense nucleic acids of the present invention may contain altered or modified sugars, bases, or bonds, may be provided in special forms such as ribosomes or microspheres, may be applied by gene therapy, It could be given in additional form. In this way, the charge in the phosphate skeleton is used in the additional form. Polycationic substances such as polylysine, which acts to neutralize, and crude water-based substances, such as lipids (eg, phospholipids, cholesterol, etc.), which enhance the interaction with cell membranes and increase the uptake of nucleic acids. Preferred lipids for addition include cholesterol and its derivatives (eg, cholesteryl chromate formate, cholic acid, etc.). These can be attached to the 3 'end or 5' end of the nucleic acid, and can be attached via a base, sugar, or intramolecular nucleoside bond. Other groups include cap groups specifically located at the 3 'or 5' end of nucleic acids that 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, such as dalicol such as polyethylene glycol and tetraethylene dalicol.
アンチセンス核酸の阻害活性は、 本発明の形質転換体、 本発明の生体内や生体 外の遺伝子発現系、 あるいは本発明のタンパク質の生体内や生体外の翻訳系を用 いて調べることができる。 該核酸は公知の各種の方法で細胞に適用できる。  The inhibitory activity of the antisense nucleic acid can be examined using the transformant of the present invention, the in vivo or in vitro gene expression system of the present invention, or the in vivo or in vitro translation system of the protein of the present invention. The nucleic acid can be applied to cells by various known methods.
以下に、 本発明で用いられるタンパク質もしくは部分ペプチドまたはその塩 ( 以下、 本発明のタンパク質と略記する場合がある) 、 本発明のタンパク質または 部分ペプチドをコードする D NA (以下、 本発明の D NAと略記する場合がある ) 、 本発明のタンパク質もしくは部分ペプチドまたはその塩に対する抗体 (以下 、 本発明の抗体と略記する場合がある) 、 および本発明の D NAのアンチセンス ヌクレオチド (以下、 本発明のアンチセンスヌクレオチドと略記する場合がある ) の用途を説明する。  Hereinafter, the protein or partial peptide used in the present invention or a salt thereof (hereinafter, sometimes abbreviated as the protein of the present invention), the DNA encoding the protein or the partial peptide of the present invention (hereinafter, 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), and an antisense nucleotide of the DNA of the present invention (hereinafter, the present invention) May be abbreviated as “antisense nucleotide”).
本発明のタンパク質は心筋梗塞後の心不全移行期 (心不全代償期/心不全非代 償期) の心臓に発現が上昇するので、 疾患マーカ一として利用することができる 。 すなわち、 心機能の低下を特徴とする疾病 (例、 心筋梗塞後の心不全;狭心症 ;心筋症;狭心症、 心筋症などの疾患に由来する心不全などの心疾患など) の早 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。  Since the expression of the protein of the present invention increases in the heart at the stage of heart failure transition (heart failure decompensation / heart failure decompensation) after myocardial infarction, it can be used as a disease marker. That is, early diagnosis of diseases characterized by decreased cardiac function (eg, heart failure after myocardial infarction; angina pectoris; cardiomyopathy; heart diseases such as heart failure resulting from diseases such as angina pectoris and cardiomyopathy). It is useful as a marker for determining the severity of symptoms and predicting disease progression.
本発明のタンパク質をコードする遺伝子のアンチセンスヌクレオチド、 本発明 のタンパク質の活性を調節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、 心機能の低下を特徴とする疾病 (例、 心筋梗塞後の心不全;狭心症;心筋症;狭心症、 心筋症などの疾患に由来する心 不全などの心疾患など) などの治療 ·予防剤として有用である。 Antisense nucleotide of a gene encoding the protein of the present invention; Pharmaceuticals containing a compound that regulates the activity of a protein or a salt thereof or an antibody against the protein of the present invention include, for example, diseases characterized by decreased cardiac function (eg, heart failure after myocardial infarction; angina pectoris; cardiomyopathy) Useful in the treatment and prevention of heart diseases such as heart failure due to diseases such as angina pectoris and cardiomyopathy).
〔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
本発明の夕ンパク質は心筋梗塞後の心機能低下とともに (心不全代償期 Z心不 全非代償) 発現が増加するので、 本発明のタンパク質の活性を調節する化合物ま たはその塩は、 例えば、 心機能の低下を特徴とする疾病 (例、 心筋梗塞後の心不 全;狭心症;心筋症;狭心症、 心筋症などの疾患に由来する心不全などの心疾患 など) などの心疾患の治療 ·予防薬として使用できる。  Since the expression of the protein of the present invention increases with a decrease in cardiac function after myocardial infarction (heart failure decompensation Z cardiac decompensation), compounds or salts thereof that regulate the activity of the protein of the present invention include, for example, Hearts such as diseases characterized by reduced cardiac function (eg, heart failure after myocardial infarction; angina pectoris; cardiomyopathy; heart diseases such as heart failure resulting from diseases such as angina pectoris and cardiomyopathy) It can be used as a treatment and prevention drug for diseases.
したがって、 本発明のタンパク質は、 本発明のタンパク質の活性を調節する化 合物またはその塩のスクリーニングのための試薬として有用である。  Therefore, the protein of the present invention is useful as a reagent for screening a compound or its salt that regulates the activity of the protein of the present invention.
すなわち、 本発明は、  That is, the present invention
( 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 を調節する化合物またはその塩のスクリーニング方法等を提供する。  (1) A method for screening a compound or a salt thereof that regulates the activity of the protein of the present invention, characterized by using the protein of the present invention, is provided.
具体的には、 例えば  Specifically, for example,
( 2 ) (i) 本発明のタンパク質を産生する能力を有する細胞を、 好ましくは低酸 素条件下で伸展刺激を加えた場合と (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、 好ましくは低酸素条件下で伸展刺激を加 えた場合との比較を行うことを#徴とする本発明のタンパク質の活性を調節する 化合物またはその塩のスクリーニング方法を提供する。  (2) (i) A cell capable of producing the protein of the present invention is preferably tested under the condition of low oxygen conditions when stimulated with extension, and (ii) A cell capable of producing the protein of the present invention is tested. It is intended to provide a method for screening a compound or a salt thereof that regulates the activity of the protein of the present invention, which is characterized by comparing a mixture of compounds with a case where a stretching stimulus is applied, preferably under hypoxic conditions.
より具体的には、 上記スクリーニング方法においては、 例えば、 (i) と (i i ) の場合における、 本発明のタンパク質の遺伝子発現量を測定して、 比較するこ とを特徴とするものである。  More specifically, the above-mentioned screening method is characterized in that, for example, in (i) and (ii), the gene expression levels of the protein of the present invention are measured and compared.
本発明のタンパク質の活性としては、 例えば、 代償破綻に伴う心機能低下促進 活性、 過剰な代償機序の抑制作用などが挙げられる。  The activity of the protein of the present invention includes, for example, an activity of promoting cardiac function decline accompanying decompensation failure, an effect of suppressing an excessive compensation mechanism, and the like.
ここで、 上記低酸素条件下とは例えば 2 0 % 02以下の酸素濃度で例えば 2 % (ネィチヤ一、 第 3 9 4巻、 4 8 5頁一 4 9 0頁、 1 9 9 8年) の条件を意味す る。 また、 伸展刺激とは心筋細胞を伸展可能なシリコン膜上に培養し、 シリコン 膜を引っ張ることで機械的負荷を加える刺激である (J. B.C.、 '第 271巻、 33 592頁— 33597頁、 1996年、 サ一キユレ一ション、 第 89巻、 220 4頁— 2211頁、 1994年、 J.B. C.、 第 271巻、 3221頁— 3228頁 、 1996年) 。 さらには、 Here, the low-oxygen conditions e.g. 2 0% 0 2 or less of oxygen concentration, for example, 2% (Neichiya primary, third 9 Volume 4, 4 8 5 page one 4 9 0 p., 1 9 9 8 years) Means the following conditions. Stretch stimulation means that cardiomyocytes are cultured on a stretchable silicon membrane, It is a stimulus that applies a mechanical load by pulling the membrane (JBC, '271, 33 592-33597, 1996, Saccuration, 89, 2204-2211, 1994) , JBC, 271: 3221-3228, 1996). Moreover,
(3) (iii) 本発明のタンパク質を産生する能力を有する細胞または本発明の タンパク質をコードする c DNAを導入した細胞を致死的な条件下で培養を行つ た場合 (具体例としては、 血清除去下あるいは心筋細胞に比較的毒性の強いアド リアマイシンなどの抗癌剤を加えて培養した場合) と、 (iv) 本発明のタンパク 質を産生する能力を有する細胞または本発明のタンパク質をコ一ドする c DNA を導入した細胞と試験化合物の混合物を致死的な条件下で培養を行った場合 (具 体例としては、 血清除去下あるいは心筋細胞に比較的毒性の強いァドリァマイシ ンなどの抗癌剤を加えて培養した場合) との比較を行うことを特徴とする本発明 のタンパク質の活性を調節する化合物またはその塩のスクリーニング方法を提供 する。  (3) (iii) When cells having the ability to produce the protein of the present invention or cells into which cDNA encoding the protein of the present invention has been introduced are cultured under lethal conditions (as specific examples, (When serum is removed or cardiomyocytes are cultured with an anticancer agent such as adriamycin which is relatively toxic) and (iv) cells having the ability to produce the protein of the present invention or the protein of the present invention. When a mixture of cells into which the cDNA has been introduced and the test compound are cultured under lethal conditions (for example, adding anticancer drugs such as adriamicin, which is relatively toxic to serum cells or cardiomyocytes, is relatively toxic to cardiomyocytes). And a method for screening a compound or a salt thereof that regulates the activity of the protein of the present invention.
上記スクリーニング方法においては、 例えば、 (iii) と (iv) の場合における 、 細胞保護作用や、 本発明のタンパク質をコードする遺伝子の発現量を公知の方 法などで測定して、 比較する。  In the above-mentioned screening method, for example, the cytoprotective action in the cases (iii) and (iv) and the expression level of the gene encoding the protein of the present invention are measured by a known method and compared.
細胞保護作用は、 心筋細胞の活性化あるいは生存率によつて示すことができる 。 具体的には一般によく用いられる呼吸活性を測定することができる MTT (3- (4, 5-Dimethyl-2-t iazolyl)-2, 5-di henyl -2H-tetrazol iuni) 法やトリパンブル 一染色法あるいは TUNNEL染色法 (Terminal deoxytransferase- mediated d UTP-X nick end labeling, セル、 第 97巻、 189頁一 198頁、 1999年 ) で測定することができる。  Cytoprotective action can be shown by cardiomyocyte activation or viability. Specifically, the MTT (3- (4,5-Dimethyl-2-t iazolyl) -2,5-di henyl -2H-tetrazol iuni) method, which can measure commonly used respiratory activity, and trypan-bleed staining It can be measured by the TUNNEL staining method (Terminal deoxytransferase-mediated d UTP-X nick end labeling, Cell, Vol. 97, pp. 189-198, 1999).
細胞死または細胞障害時の適度な発現増強は、 細胞保護作用が期待できる。 ま た、 本発明のタンパク質をコードする遺伝子の過剰な発現増強は、 細胞の過剰な 活性化を引き起こし、 細胞の疲弊を加速すると考えられる。 従って、 本発明の夕 ンパク質をコードする遺伝子の発現量を適切にコン卜ロールすることが大切であ ると考えられる。 例えば心不全代償期および心不全非代償期のように、 本発明の タンパク質をコードする遺伝子の発現促進によって細胞障害が生じていると考え られる場合には、 阻害薬 (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) を投与し、 例えば、 心不全急性期などのように、 本発明のタンパク質をコ —ドする遺伝子の発現低下によって細胞障害が生じていると考えられる場合には 、 促進薬 (本発明のタンパク質の活性を促進する化合物またはその塩) を投与す る。 Appropriate expression enhancement at the time of cell death or cell damage can be expected to have a cytoprotective effect. In addition, overexpression of the gene encoding the protein of the present invention is considered to cause excessive activation of cells and accelerate cell exhaustion. Therefore, it is considered important to appropriately control the expression level of the gene encoding the protein of the present invention. It is thought that cell damage is caused by promotion of expression of the gene encoding the protein of the present invention, for example, during the heart failure decompensation period and the heart failure decompensation period. If administered, an inhibitor (a compound that inhibits the activity of the protein of the present invention or a salt thereof) is administered to reduce the expression of a gene encoding the protein of the present invention, for example, in the acute phase of heart failure. If cell damage is considered to have occurred, an enhancer (a compound that promotes the activity of the protein of the present invention or a salt thereof) is administered.
本発明のタンパク質をコードする DN A (遺伝子) は心臓特異的な遺伝子の発 現を正に調節する機能を有するため、 極度の本発明のタンパク質をコードする遺 伝子の発現低下は細胞機能を障害し、 逆に過剰な発現は必要以上の細胞の活性化 を惹起し、 その結果として細胞障害が生じるものと考えられる。  Since the DNA (gene) encoding the protein of the present invention has a function of positively regulating the expression of a heart-specific gene, extreme decrease in the expression of the gene encoding the protein of the present invention impairs cell function. Injury and, conversely, overexpression may cause more activation of cells than necessary, resulting in cell damage.
従って、 本発明のタンパク質または本発明の DNAの過剰な発現低下が見出さ れた場合には、 本発明のタンパク質もしくは本発明の DNA、 または上記のスク リーニング方法によって得られた本発明のタンパク質の機能を促進する化合物ま たはその塩 (具体的には後述) を投与することにより、 該細胞機能の障害を予防 •治療することができ、 逆に本発明のタンパク質または本発明の DN Aの過剰な 発現が見出された場合には、 上記のスクリーニング方法によって得られた本発明 のタンパク質の機能を阻害する化合物またはその塩 (具体的には後述) を投与す ることにより、 該細胞機能の障害を予防 ·治療することができる。  Therefore, when an excessive decrease in the expression of the protein of the present invention or the DNA of the present invention is found, the function of the protein of the present invention, the DNA of the present invention, or the function of the protein of the present invention obtained by the above-described screening method is considered. By administering a compound or a salt thereof (which will be specifically described later), which promotes cell growth, the disorder of the cell function can be prevented or treated, and conversely, an excess of the protein of the present invention or the DNA of the present invention can be obtained. If any expression is found, administration of a compound that inhibits the function of the protein of the present invention obtained by the above-described screening method or a salt thereof (specifically, described below) will increase the cell function. Disability can be prevented and treated.
さらに、 本発明は、  Further, the present invention provides
(4) 本発明のタンパク質によって発現が正に調節されている遺伝子 〔例、 心房 性ナトリゥム利尿べプチド、 カーディアツク ·ァドリァマイシン ·レスボンシブ 'プロテイン (CARP) 、 ミオシン軽鎖 2 V (MLC 2 v) 、 Nkx 2. 5、 GAT A— 4、 MEF2 C、 N— myc、 HAND— 1、 心室性ナトリウム利尿 ペプチド (B.B.R. , 第 270巻, 1074-1079頁, 2000年;デベロプメント, 第 126 巻, 1269- 1280頁, 1999年) など〕 などのプロモーターを用いるレポ一夕一 ·ジ ーン ·アツセィにおいて、 レポーター ·ジーンの酵素活性を測定することを特徴 とする、 本発明のタンパク質の活性を調節する化合物またはその塩のスクリー二 ング法も提供する。 これは、 本発明のタンパク質の発現量および活性化に依存し てレポ一夕一 ·ジーンの酵素活性が上昇することを利用するものである。 具体的 には、 初代心筋細胞、 H 9 c 2細胞株 (ATCC No. CRL— 1446) ま たは N k x 2 . 5遺伝子を導入した初代心筋細胞もしくは H 9 c 2細胞株などを 宿主細胞としてレポ一夕一 ·ジーン ·アツセィを行う。 レポーター ·ジーン ·ァ ッセィは、 例えば、 本発明のタンパク質によって発現が正に調節されている遺伝 子のプロモーター領域 〔例、 心房性ナトリウム利尿ペプチド遺伝子のプロモー夕 一領域 (J. B. C. , 第 272巻, 22800- 22808頁, 1997年;デベロプメント, 第 124巻 , 793-804頁, 1997年) など〕 の下流にレポ一夕一 ·ジーン (例、 ;8ガラクトシ ダーゼ、 クロラムフエニコールァセチルトランスフェラーゼ、 ルシフェラーゼな ど) を連結させたプラスミドを構築し、 該プラスミドを心筋細胞等に公知の方法 により導入した細胞を用いて行う。 該プラスミドの構築、 プラスミドの導入など は、 公知の方法、 例えば、 上記の本発明のタンパク質をコードする D NAを含有 する組み換えベクター、 および該組み換えベクターで形質転換された形質転換体 の製造方法と同様の方法に従って行うことができる。 (4) a gene whose expression is positively regulated by the protein of the present invention (e.g., atrial sodium diuretic peptide, cardiac adoriamycinresvonshib 'protein (CARP), myosin light chain 2V (MLC 2v), Nkx 2.5, GAT A-4, MEF2 C, N-myc, HAND-1, Ventricular natriuretic peptide (BBR, Vol. 270, pp. 1074-1079, 2000; Development, Vol. 126, 1269-1280 , 1999)), a compound that regulates the activity of the protein of the present invention, characterized by measuring the enzymatic activity of a reporter gene in a reporter gene using a promoter such as It also provides a method for screening the salt. This utilizes the fact that the enzymatic activity of the repo overnight gene is increased depending on the expression level and activation of the protein of the present invention. Specifically, primary cardiomyocytes, H9c2 cell line (ATCC No. CRL-1446), Alternatively, the primary cardiomyocyte or the H9c2 cell line into which the Nkx2.5 gene has been introduced is used as a host cell to perform repo overnight gene access. The reporter gene assay is, for example, a promoter region of a gene whose expression is positively regulated by the protein of the present invention [eg, the promoter region of the atrial natriuretic peptide gene (JBC, Vol. 272, 22800). -22808, 1997; Development, Vol. 124, pp. 793-804, 1997)) and a repo overnight gene (eg, 8 galactosidase, chloramphenicol acetyltransferase, luciferase). Is constructed by using a cell in which the plasmid has been introduced into cardiomyocytes or the like by a known method. Construction of the plasmid, introduction of the plasmid, and the like can be performed by known methods, for example, a method for producing a recombinant vector containing a DNA encoding the protein of the present invention, and a method for producing a transformant transformed with the recombinant vector. It can be performed according to a similar method.
具体例としては、  As a specific example,
(v) 本発明のタンパク質によって発現が正に調節されている遺伝子のプロモー 夕一領域の下流にレポーター ·ジーンを連結させたプラスミドを導入した細胞に (v) Promoter of a gene whose expression is positively regulated by the protein of the present invention In a cell into which a plasmid having a reporter gene ligated downstream of the promoter region is introduced.
、 本発明のタンパク質を混合または接触させた場合と、 (vi) 本発明のタンパク 質によって発現が正に調節されている遺伝子のプロモーター領域の下流にレポ一 ター 'ジーンを連結させたプラスミドを導入した細胞に、 本発明のタンパク質お よび試験化合物を混合または接触させた場合との、 レポ一夕一 ·ジーンの酵素活 性の比較を行うことを特徴とする本発明のタンパク質の活性を調節する化合物ま たはその塩のスクリーニング方法を提供する。 レポーター ·ジーンの酵素活性は 公知の方法に従って測定する。 And (vi) introducing a plasmid into which a reporter gene has been ligated downstream of the promoter region of a gene whose expression is positively regulated by the protein of the present invention. The activity of the protein of the present invention is compared with the case where the protein of the present invention and a test compound are mixed or brought into contact with the isolated cells, and the enzymatic activity of the repo overnight gene is compared. A method for screening a compound or a salt thereof is provided. The enzyme activity of the reporter gene is measured according to a known method.
該スクリーニング方法によって得られる化合物またはその塩は、 本発明のタン パク質をコードする D NA (遺伝子) の発現量および活性化に影響を与える物質 であることから、 該スクリーニング方法によって本発明のタンパク質の活性を促 進する化合物および本発明のタンパク質の活性を阻害する化合物のいずれをも選 択することができる。  The compound or salt thereof obtained by the screening method is a substance that affects the expression level and activation of DNA (gene) encoding the protein of the present invention. Any of a compound that promotes the activity of the protein and a compound that inhibits the activity of the protein of the present invention can be selected.
試験化合物としては、 例えば、 ペプチド、 タンパク質、 生体由来非ペプチド性 化合物 (糖質、 脂質など) 、 合成化合物、 微生物培養物、 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化合物であってもよい し、 公知の化合物であってもよい。 Test compounds include, for example, peptides, proteins, non-peptidic compounds derived from living organisms (such as carbohydrates and lipids), synthetic compounds, microbial cultures, cell extracts, and plant extracts Liquid, animal tissue extract and the like. These compounds may be novel compounds or known compounds.
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培地を用いて培養する。 培地は、 本発 明のタンパク質の遺伝子発現に影響を与えないものであればいずれでもよい。 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、 本来本発 明のタンパク質を産生する能力を有する初代心筋細胞あるいは前述した本発明の タンパク質をコードする D NAを含有するべク夕一で形質変換された宿主 (形質 転換体) などが用いられる。 宿主としては、 例えば、 H 9 c 2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内に発現させた形質転換体が好まし く用いられる。  To carry out the above-described screening method, cells having the ability to produce the protein of the present invention are cultured in a medium suitable for screening. The medium may be any as long as it does not affect the gene expression of the protein of the present invention. Examples of cells having the ability to produce the protein of the present invention include, for example, primary cardiomyocytes having the ability to produce the protein of the present invention or vectors containing DNA encoding the protein of the present invention described above. A host (transformant) transformed with is used. As the host, for example, animal cells such as H9c2 cells are preferably used. For the screening, for example, a transformant in which the protein of the present invention is expressed in a cell by culturing by the method described above is preferably used.
本発明の遺伝子の発現量は、 公知の方法、 例えば、 ノーザンブロッテイングや Reverse transcript ion— polymerase chain react ion (R T— P C R) やリアレ タイム P C R解析システム (ABI社製、 TadMan polymerase chain react ion) 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。  The expression level of the gene of the present invention can be determined by a known method, for example, Northern blotting, Reverse transcript ion-polymerase chain react ion (RT-PCR), or a real-time PCR analysis system (Ad, TadMan polymerase chain react ion). The measurement can be performed according to any method or a method according to the method.
例えば、 上記 (i i) の場合における遺伝子発現量を、 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 るあるいは促進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あるいは 促進する化合物として選択することができる。  For example, the gene expression level in the case (ii) is inhibited or promoted by about 20% or more, preferably 30% or more, more preferably about 50% or more, as compared with the case (i). The test compound to be tested can be selected as a compound that inhibits or promotes the activity of the protein of the present invention.
また、 上記 (i i i) と (iv) の場合における、 細胞保護作用および本発明の夕 ンパク質をコードする遺伝子の発現量を測定して、 遺伝子発現量が低下し、 細胞 障害を生じているアツセィ条件下で発現量を増加させる化合物が本発明のタンパ ク質の活性を促進する化合物として用いられる。 また発現が増強し、 細胞障害が 生じているアツセィ系では発現量を低下させる化合物が本発明のタンパク質の活 性を阻害する化合物として用いられ、 細胞保護作用を有する化合物として用いる ことができる。  In addition, in the cases (iii) and (iv) above, the cytoprotective effect and the expression level of the gene encoding the protein of the present invention were measured, and the gene expression level was reduced, resulting in cell damage. A compound that increases the expression level under the conditions is used as a compound that promotes the activity of the protein of the present invention. In an Atsushi system in which expression is enhanced and cell damage has occurred, a compound that reduces the expression level is used as a compound that inhibits the activity of the protein of the present invention, and can be used as a compound that has a cytoprotective action.
さらに、 上記 (V) の場合におけるレポ一ター 'ジーンの酵素活性を、 上記 (i 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 %以上阻害するまたは促進する試験化合物を本発明のタンパク質の活性を阻 害するまたは促進する化合物として選択することができる。 Furthermore, the enzyme activity of the reporter gene in the case of the above (V) is about 20% or more, preferably 30% or more, more preferably about 30% or more of that in the case of the above (iV). A test compound that inhibits or promotes 50% or more can be selected as a compound that inhibits or promotes the activity of the protein of the present invention.
上記のスクリーニング方法により選択された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は、 本発明のタンパク質をコードする D NA (遺伝子) の発現増強が認められる心不全代償期および非代償期期に投与することにより心 機能回復効果が期待できる。 また上記のスクリーニング方法により選択された本 発明のタンパク質の活性を促進する化合物またはその塩は発現低下が認められる 心不全急性期に投与することにより代償機序を強化し、 心筋細胞を保護すること による心保護効果 (heart protect ive ef fect) が期待できる。  The compound or a salt thereof that inhibits the activity of the protein of the present invention selected by the above screening method is used during the heart failure decompensation phase and the decompensation phase in which the expression of the DNA (gene) encoding the protein of the present invention is enhanced. The administration can be expected to have a cardiac function recovery effect. In addition, the compound or salt thereof which promotes the activity of the protein of the present invention selected by the above screening method enhances the compensatory mechanism by being administered in the acute phase of heart failure where expression is reduced, thereby protecting cardiomyocytes. A heart protective effect can be expected.
本発明のスクリーニング用キットは、 本発明で用いられるタンパク質もしくは 部分ペプチドまたはその塩、 または本発明で用いられるタンパク質もしくは部分 ペプチドを産生する能力を有する細胞を含有するものである。  The screening kit of the present invention contains the protein or partial peptide used in the present invention or a salt thereof, or a cell capable of producing the protein or partial peptide used in the present invention.
本発明のスクリーニング方法またはスクリーニング用キットを用いて得られる 化合物 (本発明のタンパク質の活性を促進または阻害する化合物) またはその塩 は、 上記した試験化合物、 例えば、 ペプチド、 タンパク質、 生体由来非ペプチド 性化合物 (例、 糖質、 脂質など) 、 合成化合物、 微生物培養物、 発酵生産物、 細 胞抽出液、 植物抽出液、 動物組織抽出液、 血漿などから選ばれた化合物またはそ の塩であり、 本発明のタンパク質の活性 (例、 心機能低下促進活性など) を調節 (促進または阻害) する化合物またはその塩である。  The compound (a compound that promotes or inhibits the activity of the protein of the present invention) or a salt thereof obtained by using the screening method or the screening kit of the present invention may be a test compound as described above, for example, a peptide, a protein, a non-peptide derived from a living body. Compounds (eg, carbohydrates, lipids, etc.), synthetic compounds, compounds or salts thereof selected from microbial cultures, fermentation products, cell extracts, plant extracts, animal tissue extracts, plasma, etc. A compound or a salt thereof that regulates (promotes or inhibits) the activity of the protein of the present invention (eg, activity to lower cardiac function).
該化合物の塩としては、 前記した本発明のタンパク質の塩と同様のものが用い られる。  As the salt of the compound, those similar to the aforementioned salts of the protein of the present invention are used.
本発明のタンパク質の活性を調節 (促進または阻害) する化合物またはその塩 は、 例えば、 心機能の低下を特徴とする疾病 (例、 心筋梗塞後の心不全;狭心症 ;心筋症;狭心症、 心筋症などの疾患に由来する心不全などの心疾患など) など に対する治療 ·予防剤などの医薬として有用である。  Compounds or salts thereof that regulate (promote or inhibit) the activity of the protein of the present invention include, for example, diseases characterized by reduced cardiac function (eg, heart failure after myocardial infarction; angina pectoris; cardiomyopathy; angina pectoris) And heart diseases such as heart failure caused by diseases such as cardiomyopathy).
本発明のスクリーニング方法またはスクリーニング用キットを用いて得られる 化合物またはその塩を上述の治療 ·予防剤として使用する場合、 常套手段に従つ て製剤化することができる。 例えば、 錠剤、 カプセル剤、 エリキシル剤、 マイク 口カプセル剤、 無菌性溶液、 懸濁液剤などとすることができる。 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、 温血動物 ( 例えば、 ヒト、 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。 When a compound or a salt thereof obtained by using the screening method or the screening kit of the present invention is used as the above-mentioned therapeutic or prophylactic agent, it can be formulated according to a conventional method. For example, tablets, capsules, elixirs, micron capsules, sterile solutions, suspensions and the like can be used. The preparations obtained in this way are safe and have low toxicity, for example, warm-blooded animals (for example, humans, mice, rats, puppies, sheep, bush, puppies, puppies, birds, cats, dogs) , Monkeys, chimpanzees, etc.) orally or parenterally.
該化合物またはその塩の投与量は、 その作用、 対象疾患、 投与対象、 投与ル一 トなどにより差異はあるが、 例えば、 心不全治療の目的で本発明のタンパク質の 活性を調節する化合物またはその塩を経口投与する場合、 一般的に成人 (体重 6 0 kgとして) においては、 一日につき該化合物またはその塩を約 0. 1〜10 Omg、 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、 心不全治療の目的で本発明の タンパク質の活性を調節する化合物またはその塩を注射剤の形で通常成人 (60 kg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〜 3 Omg程度、 好ましくは約 0. 1〜2 Omg程度、 より好ましくは約 0. 1〜 1 Omg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、 60 k g当たりに換算した量を投与することができる。  The dose of the compound or a salt thereof varies depending on its action, target disease, subject of administration, route of administration, and the like.For example, a compound or a salt thereof that regulates the activity of the protein of the present invention for the purpose of treating heart failure When the compound is orally administered, generally in an adult (assuming a body weight of 60 kg), the compound or a salt thereof is used in an amount of about 0.1 to 10 Omg, preferably about 1.0 to 50 mg, more preferably about 0.1 to 50 mg per day. 1. Administer 0 to 20 mg. When administered parenterally, the single dose of the compound or a salt thereof varies depending on the administration subject, target disease, and the like.For example, a compound or a compound thereof that regulates the activity of the protein of the present invention for the purpose of treating heart failure When the salt is administered to an adult (as 60 kg) usually in the form of an injection, the compound or a salt thereof is used in an amount of about 0.01 to 3 Omg per day, preferably about 0.1 to 2 Omg, more preferably about 0.1 to 2 Omg. It is convenient to administer about 0.1 to 1 Omg by intravenous injection. In the case of other animals, the amount converted per 60 kg can be administered.
〔2〕 本発明のタンパク質、 その部分ペプチドまたはその塩の定量  [2] Quantification of the protein of the present invention, its partial peptide or its salt
本発明のタンパク質に対する抗体 (以下、 本発明の抗体と略記する場合がある ) は、 本発明のタンパク質を特異的に認識することができるので、 被検液中の本 発明のタンパク質の定量、 特にサンドイ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, particularly 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タンパク質の割合を測定 することを特徴とする被検液中の本発明のタンパク質の定量法、 および  (i) reacting the antibody of the present invention with a test solution and a labeled protein of the present invention competitively, and measuring the ratio of the labeled protein of the present invention bound to the antibody. A method for quantifying the protein of the present invention in a test solution, and
(ii) 被検液と担体上に不溶化した本発明の抗体および標識化された本発明の別 の抗体とを同時あるいは連続的に反応させたのち、 不溶化担体上の標識剤の活性 を測定することを特徴とする被検液中の本発明のタンパク質の定量法を提供する 上記 (i i) の定量法においては、 一方の抗体が本発明のタンパク質の N端部を 認識する抗体で、 他方の抗体が本発明のタンパク質の C端部に反応する抗体であ ることが望ましい。 (ii) After reacting the test solution with the antibody of the present invention insoluble on the carrier and another labeled antibody of the present invention simultaneously or continuously, the activity of the labeling agent on the insoluble carrier is measured. To provide a method for quantifying the protein of the present invention in a test solution, characterized in that: 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. .
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 ( a b ' ) 2、 F a b '、 あるいは F a b画分を用いてもよい。 In addition, the protein of the present invention can be quantified using a monoclonal antibody against the protein of the present invention (hereinafter sometimes referred to as the monoclonal antibody of the present invention), and can also be detected by tissue staining or the like. . For these purposes, the antibody molecule itself may be used, or the F (ab ') 2 , Fab', or Fab fraction of the antibody molecule may be used.
本発明の抗体を用いる本発明のタンパク質の定量法は、 特に制限されるべきも のではなく、 被測定液中の抗原量 (例えば、 タンパク質量) に対応した抗体、 抗 原もしくは抗体一抗原複合体の量を化学的または物理的手段により検出し、 これ を既知量の抗原を含む標準液を用いて作製した標準曲線より算出する測定法であ れば、 いずれの測定法を用いてもよい。 例えば、 ネフロメトリ一、 競合法、 ィム ノメトリック法およびサンドイッチ法が好適に用いられるが、 感度、 特異性の点 で、 後述するサンドイッチ法を用いるのが特に好ましい。  The method for quantifying the protein of the present invention using the antibody of the present invention is not particularly limited, and may be an antibody, an antigen, or an antibody-antigen complex corresponding to the amount of antigen (eg, the amount of protein) in the test solution. Any measurement method may be used as long as the amount of the body is detected by chemical or physical means, and this is calculated from a standard curve prepared using a standard solution containing a known amount of antigen. . For example, nephrometry, a competition method, an immunometric method and a sandwich method are suitably used, but it is particularly preferable to use a sandwich method described later in terms of sensitivity and specificity.
標識物質を用いる測定法に用いられる標識剤としては、 例えば、 放射性同位元 素、 酵素、 蛍光物質、 発光物質などが用いられる。 放射性同位元素としては、 例 えば、 〔1 2 5 I〕 、 〔1 3 1 I〕 、 〔3 H〕 、 〔1 4 C〕 などが用いられる。 上記酵 素としては、 安定で比活性の大きなものが好ましく、 例えば、 i3—ガラクトシダ ーゼ、 )8—ダルコシダ一ゼ、 アルカリフォスファタ一ゼ、 パーォキシダーゼ、 リ ンゴ酸脱水素酵素などが用いられる。 蛍光物質としては、 例えば、 フルォレス力 ミン、 フルォレツセンイソチオシァネートなどが用いられる。 発光物質としては 、 例えば、 ルミノール、 ルミノール誘導体、 ルシフェリン、 ルシゲニンなどが用 いられる。 さらに、 抗体あるいは抗原と標識剤との結合にピオチン一アビジン系 を用いることもできる。 As a labeling agent used in a measuring method using a labeling substance, for example, a radioisotope, an enzyme, a fluorescent substance, a luminescent substance and the like are used. Radioisotopes, if example embodiment, [1 2 5 I], [1 3 1 I], [3 H], and [1 4 C] used. As the above-mentioned enzyme, those which are stable and have a large specific activity are preferable. For example, i3-galactosidase,) 8-darcosidase, alkaline phosphatase, peroxidase, and lignoic acid dehydrogenase are used. Can be As the fluorescent substance, for example, fluorescein, fluorescein isothiosinate and the like are used. As the luminescent substance, for example, luminol, luminol derivative, luciferin, lucigenin and the like are used. Further, a biotin-avidin system can be used for binding the antibody or antigen to the labeling agent.
抗原あるいは抗体の不溶化に当っては、 物理吸着を用いてもよく、 また通常夕 ンパク質あるいは酵素等を不溶化、 固定化するのに用いられる化学結合を用いる 方法でもよい。 担体としては、 ァガロース、 デキストラン、 セルロースなどの不 溶性多糖類、 ポリスチレン、 ポリアクリルアミド、 シリコン等の合成樹脂、 ある いはガラス等が挙げられる。 For the insolubilization of the antigen or antibody, physical adsorption may be used, or a method using a chemical bond usually used for insolubilizing and immobilizing proteins or enzymes may be used. Carriers include insoluble polysaccharides such as agarose, dextran, and cellulose; and synthetic resins such as polystyrene, polyacrylamide, and silicon. Or glass.
サンドイッチ法においては不溶化した本発明のモノクローナル抗体に被検液を 反応させ (1次反応) 、 さらに標識化した別の本発明のモノクローナル抗体を反 応させ (2次反応) たのち、 不溶化担体上の標識剤の活性を測定することにより 被検液中の本発明のタンパク質量を定量することができる。 1次反応と 2次反応 は逆の順序に行っても、 また、 同時に行なってもよいし時間をずらして行なって もよい。 標識化剤および不溶化の方法は前記のそれらに準じることができる。 ま た、 サンドイッチ法による免疫測定法において、 固相用抗体あるいは標識用抗体 に用いられる抗体は必ずしも 1種類である必要はなく、 測定感度を向上させる等 の目的で 2種類以上の抗体の混合物を用いてもよい。  In the sandwich method, the test solution is reacted with the insolubilized monoclonal antibody of the present invention (primary reaction), and further reacted with another labeled monoclonal antibody of the present invention (secondary reaction). By measuring the activity of the labeling agent, the amount of the protein of the present invention in the test solution can be determined. The primary reaction and the secondary reaction may be performed in the reverse order, may be performed simultaneously, or may be performed at staggered times. The labeling agent and the method of insolubilization can be in accordance with those described above. Also, in the immunoassay by the sandwich method, the antibody used for the solid phase antibody or the labeling antibody does not necessarily need to be one kind, and a mixture of two or more kinds of antibodies is used for the purpose of improving measurement sensitivity and the like. May be used.
本発明のサンドイツチ法による本発明の夕ンパク質の測定法においては、 1次 反応と 2次反応に用いられる本発明のモノクローナル抗体は、 本発明の夕ンパク 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 according to the San Deutsch method of the present invention, the monoclonal antibody of the present invention used in the primary reaction and the secondary reaction is an antibody having a different site to which the protein of the present invention binds. It is preferably used. That is, the antibody used in the primary reaction and the secondary reaction is, for example, when the antibody used in the secondary reaction recognizes the C-terminal of the protein of the present invention, the antibody used in the primary reaction is Preferably, an antibody that recognizes other than the C-terminal, for example, the N-terminal, is used.
本発明のモノクローナル抗体をサンドイッチ法以外の測定システム、 例えば、 競合法、 ィムノメトリック法あるいはネフロメトリ一などに用いることができる 競合法では、 被検液中の抗原と標識抗原とを抗体に対して競合的に反応させた のち、 未反応の標識抗原(F)と、 抗体と結合した標識抗原 (B ) とを分離し (B /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。  The monoclonal antibody of the present invention can be used in a measurement system other than the sandwich method, for example, a competition method, an immunometric method, a nephelometry method, or the like.In a competition method, an antigen in a test solution and a labeled antigen are applied to the antibody. After the competitive reaction, the unreacted labeled antigen (F) and the labeled antigen (B) bound to the antibody are separated (B / F separation), and the labeling amount of either B or F is measured. The amount of antigen in the test solution is quantified. In this reaction method, a soluble antibody is used as an antibody, B / F separation is performed using a polyethylene glycol, a liquid phase method using a second antibody against the antibody, or a solid phase antibody is used as the first antibody. Alternatively, an immobilization method using a soluble first antibody and an immobilized antibody as the second antibody is used.
ィムノメトリック法では、 被検液中の抗原と固相化抗原とを一定量の標識化抗 体に対して競合反応させた後固相と液相を分離するか、 あるいは、 被検液中の抗 原と過剰量の標識化抗体とを反応させ、 次に固相化抗原を加え未反応の標識化抗 体を固相に結合させたのち、 固相と液相を分離する。 次に、 いずれかの相の標識 量を測定し被検液中の抗原量を定量する。 In the immunometric method, the antigen in the test solution and the immobilized antigen are subjected to a competitive reaction with a certain amount of labeled antibody, and then the solid phase and the liquid phase are separated. Reaction with an excess amount of the labeled antibody, and then adding immobilized antigen to the unreacted labeled antibody. After binding the body to the solid phase, the solid and liquid phases are separated. Next, the amount of label in either phase is measured to determine the amount of antigen in the test solution.
また、 ネフロメトリ一では、 ゲル内あるいは溶液中で抗原抗体反応の結果生じ た不溶性の沈降物の量を測定する。 被検液中の抗原量が僅かであり、 少量の沈降 物しか得られない場合にもレーザー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 nephrometry, the amount of insoluble sediment resulting from an antigen-antibody reaction in a gel or in a solution is measured. Even when the amount of antigen in the test solution is small and only a small amount of sediment is obtained, laser nephrometry utilizing laser scattering is preferably used.
これら個々の免疫学的測定法を本発明の定量方法に適用するにあたっては、 特 別の条件、 操作等の設定は必要とされない。 それぞれの方法における通常の条件 、 操作法に当業者の通常の技術的配慮を加えて本発明のタンパク質の測定系を構 築すればよい。 これらの一般的な技術手段の詳細については、 総説、 成書などを 参照することができる。  In applying these individual immunological measurement methods to the quantification method of the present invention, no special conditions, operations, and the like need to be set. The protein measurement system of the present invention may be constructed by adding ordinary technical considerations to those skilled in the art to the ordinary conditions and procedures in each method. For details of these general technical means, reference can be made to reviews and written documents.
例えば、 入江 寛編 「ラジオィムノアツセィ」 (講談社、 昭和 4 9年発行) 、 入江 寛編 「続ラジオィムノアツセィ」 (講談社、 昭和 5 4年発行) 、 石川栄治 ら編 「酵素免疫測定法」 (医学書院、 昭和 5 3年発行) 、 石川栄治ら編 「酵素免 疫測定法」 (第 2版) (医学書院、 昭和 5 7年発行) 、 石川栄治ら編 「酵素免疫 測定法」 (第 3版) (医学書院、 昭和 6 2年発行) 、 「Methods in ENZYMOLOGYj Vol. 70 (Immunochemical Tec ni ues (Part A) ) ^ 同書 Vol. 73 (Immunochemical TechniQues (Part B) )、 同書 Vol. 74 (Immunochemical Techniaues (Part C) )、 同書 Vol. 84 (Immunochemical Techniaues (Part D: Selected Immunoassays) )、 同書 Vol. 92 (Immunochefflical Techniaues (Part E: Monoclonal Ant ibodies and General Immunoassay Methods) ) > 同書 Vol. 121 (Immunochemical Techniaues (P art I '.Hybridoma Technology and Monoclonal Ant ibodies) ) (以上、 ァカデミツ クプレス社発行) などを参照することができる。  For example, edited by Hiro Irie "Radio Nonotsusei" (Kodansha, published in 1949), edited by Hiro Irie "Radio Imunoatsusei" (Kodansha, published in 1954), Eiji Ishikawa et al. "Measurement Method" (Medical Shoin, published in 1958), Eiishi Ishikawa et al., "Enzyme Immunoassay" (Second Edition) (Medical Publishing, published in 1977), Eiji Ishikawa, "Enzyme Immunoassay" (3rd edition) (Medical Shoin, published in 1962), "Methods in ENZYMOLOGYj Vol. 70 (Immunochemical Tec niues (Part A)) ^ Ibid. 73 (Immunochemical TechniQues (Part B)), Ibid. Vol. 74 (Immunochemical Techniaues (Part C)), ibid.Vol. 84 (Immunochemical Techniaues (Part D: Selected Immunoassays)), ibid.Vol. 92 (Immunochefflical Techniaues (Part E: Monoclonal Ant ibodies and General Immunoassay Methods))> ibid. 121 (Immunochemical Techniaues (P art I '.Hybridoma Technology and Monoclonal Ant ibodies)) ( The above can be referred to, for example, 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夕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、 心機能の 低下を特徴とする疾病 (例、 心筋梗塞後の心不全;狭心症;心筋症;狭心症、 心 筋症などの疾患に由来する心不全などの心疾患など) である、 または将来罹患す る可能性が高いと診断することができる。 Furthermore, when an increase 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, a disease characterized by decreased cardiac function (Eg, heart failure after myocardial infarction; angina; cardiomyopathy; heart disease such as heart failure from diseases such as angina and cardiomyopathy) or will be affected in the future Can be diagnosed as having a high possibility.
また、 本発明の抗体は、 体液や組織などの被検体中に存在する本発明のタンパ ク質を検出するために使用することができる。 また、 本発明のタンパク質を精製 するために使用する抗体カラムの作製、 精製時の各分画中の本発明のタンパク質 の検出、 被検細胞内における本発明のタンパク質の挙動の分析などのために使用 することができる。  Further, the antibody of the present invention can be used for detecting the protein of the present invention present in a subject such as a body fluid or a tissue. In addition, for the preparation of an antibody column used for purifying the protein of the present invention, the detection of the protein of the present invention in each fraction during purification, and the analysis of the behavior of the protein of the present invention in test cells, etc. Can be used.
〔3〕 遺伝子診断剤  (3) Gene diagnostic agent
本発明の D N Aは、 例えば、 プローブとして使用することにより、 温血動物 ( 例えば、 ヒト、 ラッ卜、 マウス、 モルモット、 ゥサギ、 トリ、 ヒッジ、 ブ夕、 ゥ シ、 ゥマ、 ネコ、 ィヌ、 サル、 チンパンジーなど) における本発明のタンパク質 またはその部分ペプチドをコードする D N Aまたは mR N Aの異常 (遺伝子異常 ) を検出することができるので、 例えば、 該 D NAまたは mR NAの損傷、 突然 変異あるいは発現低下や、 該 D N Aまたは m R N Aの増加あるいは発現過多など の遺伝子診断剤として有用である。  The DNA of the present invention can be used, for example, as a probe to produce warm-blooded animals (eg, humans, rats, mice, guinea pigs, egrets, birds, higgies, bush, horses, dogs, cats, dogs, DNA or mRNA encoding the protein of the present invention or its partial peptide (monkey, chimpanzee, etc.) can be detected (gene abnormality). For example, the DNA or mRNA is damaged, mutated or expressed. It is useful as a gene diagnostic agent for a decrease, an increase in the DNA or mRNA, or an overexpression.
本発明の D NA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プ 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巻, 8 7 4〜 8 7 9頁 (1 9 8 9年) 、 プロシージングズ ·ォブ ·ザ ·ナショナル · アカデミー ·ォブ ·サイェンシィズ ·ォブ ·ユーエスエー (Proceedings of the Nat ional Academy of Sciences of the Uni ted States of America; , 第 8 6 卷, 2 7 6 6〜2 7 7 0頁 (1 9 8 9年) ) などにより実施することができる。 例えば、 ノーザンハイブリダィゼーシヨンにより発現過多が検出された場合や P C R— S S C P法により D N Aの突然変異が検出された場合は、 例えば、 心機 能低下を伴う心疾患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。 〔4〕 アンチセンスヌクレオチドを含有する医薬  The above-described genetic diagnosis using the DNA of the present invention can be performed, for example, by the well-known Northern hybridization or PCR-SSCP method (Genomics, Vol. 5, p. 874-879 (1989). Proceedings of the National Academy of Sciences of the United States of America; Vol. 86, Vol. 27, Proceedings of the National Academy of Sciences of the United States of America; 66-27770 (11989)), etc. For example, when overexpression is detected by Northern hybridization or DNA mutation by PCR-SSCP method Is detected, it is possible to diagnose that the disease is highly likely to be a disease such as a heart disease accompanied by a decrease in cardiac function, etc. [4] A drug containing an antisense nucleotide
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 発明のァンチセンスヌクレオチドは低毒性であり、 生体内における本発明の夕ン パク質または本発明の D NAの活性や機能 (例、 心機能低下促進活性など) を調 節 (阻害) することができるので、 例えば、 心機能の低下を特徴とする疾病 (例 、 心筋梗塞後の心不全;狭心症;心筋症;狭心症、 心筋症などの疾患に由来する 心不全などの心疾患など) などの治療 ·予防剤として使用することができる。 上記アンチセンスヌクレオチドを上記の治療 ·予防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。 The antisense nucleotide of the present invention, which complementarily binds to the DNA of the present invention and can suppress the expression of the DNA, has low toxicity, and is characterized by having the protein of the present invention or the DNA of the present invention in vivo in vivo. It can regulate (inhibit) activities and functions (eg, cardiac dysfunction promoting activity), for example, diseases characterized by reduced cardiac function (eg, heart failure after myocardial infarction; angina pectoris; myocardium Sickness; derived from diseases such as angina pectoris and cardiomyopathy It can be used as a treatment and prevention agent for heart diseases such as heart failure. When the above-mentioned antisense nucleotide is used as the above-mentioned therapeutic or prophylactic agent, it can be formulated and administered according to a known method.
例えば、 該アンチセンスヌクレオチドを用いる場合、 該アンチセンスヌクレオ チドを単独あるいはレトロウイルスベクタ一、 アデノウイルスベクタ一、 アデノ ウィルスァソシエーテッドウィルスベクターなどの適当なベクターに挿入した後 、 常套手段に従って、 温血動物 (例えば、 ヒト、 ラッ卜、 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。 該アンチセンスヌ クレオチドは、 そのままで、 あるいは摂取促進のために補助剤などの生理学的に 認められる担体とともに製剤化し、 遺伝子銃やハイドロゲルカテーテルのような カテーテルによって投与できる。  For example, when the antisense nucleotide is used, the antisense nucleotide is inserted alone or into an appropriate vector such as a retrovirus vector, an adenovirus vector, an adenovirus associated virus vector, and the like. Oral or parenteral to warm-blooded animals (e.g., humans, rats, mice, guinea pigs, egrets, birds, higgins, pigs, pigs, dogs, cats, dogs, monkeys, chimpanzees, etc.) Can be administered. The antisense nucleotide can be administered as it is or in the form of a formulation together with a physiologically acceptable carrier such as an adjuvant for promoting uptake, and then administered using a gene gun or a catheter such as a hydrogel catheter.
該アンチセンスヌクレオチドの投与量は、 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、 心不全の治療の目的で本発明のアンチセンスヌ クレオチドを経口投与する場合、 一般的に成人 (体重 6 O k g ) においては、 一 日につき該アンチセンスヌクレオチドを約 0 . 1〜1 0 O m g投与する。  The dose of the antisense nucleotide varies depending on the disease to be treated, the subject to be administered, the administration route, and the like. For example, when the antisense nucleotide of the present invention is orally administered for the purpose of treating heart failure, generally the adult (body weight) is used. At 6 O kg), about 0.1 to 10 O mg of the antisense nucleotide is administered per day.
さらに、 該アンチセンスヌクレオチドは、 組織や細胞における本発明の D NA の存在やその発現状況を調べるための診断用オリゴヌクレオチドプローブとして 使用することもできる。  Furthermore, the antisense nucleotide can also be used as a diagnostic oligonucleotide probe for examining the presence of the DNA of the present invention in tissues or cells and the state of its expression.
本発明は、 さらに  The present invention further provides
①本発明のタンパク質をコードする R NAの一部を含有する二重鎖 R NA、 (1) a double-stranded RNA containing a part of the RNA encoding the protein of the present invention,
②前記二重鎖 R N Aを含有してなる医薬、 (2) a medicament comprising the double-chain RNA,
③本発明のタンパク質をコードする RN Aの一部を含有するリポザィム、 (3) a lipozyme containing a part of the RNA encoding the protein of the present invention;
④前記リポザィムを含有してなる医薬を提供する。 (4) To provide a medicine containing the lipozyme.
これらの二重鎖 R NA、 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、 心機能低下促進活性など) を調節 (阻害) すること ができるので、 例えば、 心機能の低下を特徴とする疾病 (例、 心筋梗塞後の心不 全;狭心症;心筋症;狭心症、 心筋症などの疾患に由来する心不全などの心疾患 など) などの治療 ·予防剤として使用することができる。 These 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 polynucleotide, and can inhibit the expression of the peptide of the present invention in vivo. It can regulate (inhibit) the activity or function of the polynucleotide (eg, DNA) of the present invention (eg, the activity of promoting cardiac function decline), for example, a disease (eg, a disease characterized by decreased cardiac function). Heart failure after myocardial infarction All; angina; cardiomyopathy; heart diseases such as heart failure derived from diseases such as angina and cardiomyopathy).
二重鎖 RNA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。  Double-stranded RNA can be produced by designing based on the sequence of the polynucleotide of the present invention according to a known method (eg, Nature, 411, 494, 2001).
リポザィムは、 公知の方法 (例、 TRENDS in Molecular Medicine, 7巻, 221頁 , 2001年) に準じて、 本発明のポリヌクレオチドの配列を基に設計して製造する ことができる。 例えば、 本発明のペプチドをコードする R NAの一部に公知のリ ポザィムを連結することによつて製造することができる。 本発明のペプチドをコ ードする RN Aの一部としては、 公知のリポザィムによって切断され得る本発明 の R NA上の切断部位に近接した部分 (R NA断片) が挙げられる。  The lipozyme can be designed and manufactured based on the sequence of the polynucleotide of the present invention according to a known method (eg, TRENDS in Molecular Medicine, 7, 221, 2001). For example, it can be produced by linking a known lipozyme to a part of the RNA encoding the peptide of the present invention. A part of the RNA encoding the peptide of the present invention includes a portion (RNA fragment) close to the cleavage site on the RNA of the present invention, which can be cleaved by a known lipozyme.
上記の二重鎖 RNAまたはリポザィムを上記予防 ·治療剤として使用する場合 、 アンチセンスポリヌクレオチドと同様にして製剤化し、 投与することができる  When the above double-stranded RNA or lipozyme is used as the above-mentioned prophylactic / therapeutic agent, it can be formulated and administered in the same manner as an antisense polynucleotide.
〔5〕 本発明の抗体を含有する医薬 [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、 心機能の 低下を特徴とする疾病 (例、 心筋梗塞後の心不全;狭心症;心筋症;狭心症、 心 筋症などの疾患に由来する心不全などの心疾患など) などの予防 ·治療剤として 使用することができる。  The antibody of the present invention, which has the activity of neutralizing the activity of the protein of the present invention, is used for a disease characterized by a decrease in cardiac function (eg, heart failure after myocardial infarction; angina; cardiomyopathy; angina, cardiac muscle) It can be used as a prophylactic or therapeutic agent for diseases such as heart failure caused by diseases such as sickness.
本発明の抗体を含有する上記疾病の予防'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、 温血動物 (例えば、 ヒト、 ラ ット、 マウス、 モルモット、 ゥサギ、 トリ、 ヒッジ、 ブタ、 ゥシ、 ゥマ、 ネコ、 ィヌ、 サル、 チンパンジーなど) 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、 成人の心不全の治療 ·予防のために使用する場合には、 本発明 の抗体を 1回量として、 通常 0. 0 1〜2 O m g/ k g体重程度、 好ましくは 0. 1〜1 O m g /k g体重程度、 さらに好ましくは 0. l〜5 m g / k g体重程度 を、 1日 1〜5回程度、 好ましくは 1日 1〜3回程度、 静脈注射により投与する のが好都合である。 他の非経口投与および経口投与の場合もこれに準ずる量を投 与することができる。 症状が特に重い場合には、 その症状に応じて増量してもよ い。 The agent for preventing or treating the above-mentioned diseases, which contains the antibody of the present invention, has low toxicity, and can be used as it is as a liquid or as a pharmaceutical composition of an appropriate dosage form, as a warm-blooded animal (eg, human, rat, mouse, It can be administered orally or parenterally to guinea pigs, egrets, birds, sheep, pigs, puppies, pomas, cats, dogs, monkeys, chimpanzees, etc. The dosage varies depending on the administration subject, target disease, symptoms, administration route and the like.For example, when used for the treatment or prevention of heart failure in adults, the antibody of the present invention is usually administered as a single dose. 0.0 1-2 O mg / kg body weight, preferably 0.1-1 O mg / kg body weight, more preferably 0.1-5 mg / kg body weight, about 1-5 times a day, It is convenient to administer by intravenous injection preferably 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 according to the symptoms. No.
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ゥ ムなどが用いられる。  That is, for example, compositions for oral administration include solid or liquid dosage forms, specifically tablets (including sugar-coated tablets and film-coated tablets), pills, granules, powders, capsules (soft capsules and the like). ), Syrups, emulsions, suspensions and the like. Such a composition is produced by a known method and contains a carrier, diluent or excipient commonly used in the field of pharmaceuticals. For example, lactose, starch, sucrose, magnesium stearate and the like are used as carriers and excipients for tablets.
非経口投与のための組成物としては、 例えば、 注射剤、 坐剤などが用いられ、 注射剤は静脈注射剤、 皮下注射剤、 皮内注射剤、 筋肉注射剤、 点滴注射剤などの 剤形を包含する。 かかる注射剤は、 公知の方法に従って、 例えば、 上記抗体また はその塩を通常注射剤に用いられる無菌の水性もしくは油性液に溶解、 懸濁また は乳化することによって調製する。 注射用の水性液としては、 例えば、 生理食塩 水、 ブドウ糖やその他の補助薬を含む等張液などが用いられ、 適当な溶解補助剤 、 例えば、 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 卜 8 0、 H C〇一 5 0 (polyoxyethylene (50 mol) adduct of hydrogenated ca s 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, drip injections, etc. Is included. Such injections are prepared according to known methods, for example, by dissolving, suspending or emulsifying the antibody or a salt thereof in a sterile aqueous or oily liquid commonly used for injections. As an aqueous solution for injection, for example, physiological saline, isotonic solution containing glucose and other adjuvants and the like are used, and a suitable solubilizing agent, for example, alcohol (eg, ethanol), polyalcohol (eg, Propylene glycol, polyethylene glycol), non-ionic surfactants (eg, polysorbate 80, HC-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)), etc. Good. As the oily liquid, for example, sesame oil, soybean oil, and the like are used, and benzyl benzoate, benzyl alcohol, and the like may be used in combination as a solubilizing agent. The prepared injection is usually filled in an appropriate ampoule. A suppository for rectal administration is prepared by mixing the above antibody or a salt thereof with a 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〜 5 0 0 m g、 とりわけ注射剤では 5〜1 0 O m g、 その他の剤形では 1 0〜2 5 O m gの上記抗体が含有されていることが 好ましい。 The above-mentioned oral or parenteral pharmaceutical composition is conveniently prepared in a unit dosage form adapted to the dose of the active ingredient. Examples of such dosage unit forms include tablets, pills, capsules, injections (ampoules), and suppositories. Each dosage unit usually contains 5 to 500 mg, especially 5 to 100 mg for injections, and 10 to 25 mg for other dosage forms. preferable.
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。  Each of the above-mentioned compositions may contain another active ingredient as long as the composition does not cause an undesirable interaction with the above-mentioned antibody.
〔6〕 本発明のタンパク質を含有する医薬  [6] A drug containing the protein of the present invention
( 1 ) 本発明のタンパク質は、 本発明の抗体を産生するためのワクチン等の医薬 として用いられる。 本発明の抗体を産生するためのワクチンは、 本発明のタンパ ク質を用いて、 公知の方法により製造することができる。  (1) The protein of the present invention is used as a drug such as a vaccine for producing the antibody of the present invention. A vaccine for producing the antibody of the present invention can be produced by a known method using the protein of the present invention.
( 2 ) 本発明のタンパク質に核移行シグナル配列 (antenapedia:Drosophi laの核 移行ペプチド、 東京医科歯科大学の伊藤らの学会報告:第 2回 Molecular Cardi ovascular Conference) を連結させた遺伝子組換え型 N k x 2 . 5を全身に投与 したり、 心臓カテ一テル検査の際に心臓へ直接投与することによって、 低下した 本発明のタンパク質を補うことができる。  (2) Recombinant N obtained by linking a nuclear localization signal sequence (antenapedia: a nuclear localization peptide of Drosophi la, a report by Ito et al. Of Tokyo Medical and Dental University: 2nd Molecular Cardiovascular Conference) to the protein of the present invention. The reduced protein of the present invention can be supplemented by administering kx2.5 systemically or directly to the heart during a cardiac catheterization test.
〔7〕 本発明の D NAを含有する医薬  (7) a drug containing the DNA of the present invention
本発明の D N Aを含有する医薬は、 心不全の遺伝子治療に用いられる。 本発明 のタンパク質の機能の一つとして、 心臓特異的な遺伝子群の発現増強がある。 こ の結果として心筋梗塞後の代償機序の強化が生じると考えている。 従って心不全 急性期のような本発明のタンパク質の発現が低下している時期には、 本発明の夕 ンパク質が適切な発現量を維持するように、 本発明の D NAを投与することがで さる。  The medicament containing DNA of the present invention is used for gene therapy of heart failure. One of the functions of the protein of the present invention is to enhance the expression of heart-specific genes. We believe that this will result in enhanced compensatory mechanisms following myocardial infarction. Therefore, during the period when the expression of the protein of the present invention is decreased, such as in the acute phase of heart failure, the DNA of the present invention can be administered so that the protein of the present invention maintains an appropriate expression level. Monkey
〔8〕 本発明の D NAを有する動物の作製 .  (8) Preparation of an animal having the DNA of the present invention.
本発明の D N Aを用いて、 本発明のタンパク質を発現するトランスジエニック 動物を作製することができる。 動物としては、 哺乳動物 (例えば、 ラット、 マウ' ス、 ゥサギ、 ヒッジ、 ブタ、 ゥシ、 ネコ、 ィヌ、 サルなど) など (以下、 動物と 略記する場合がある) が挙げられるが、 特に、 マウス、 ゥサギなどが好適である 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遺伝子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、 ゥサギ由来の本発明の D NAを導入させる場合 、 これと相同性が高い動物由来の本発明の D N Aを動物細胞で発現させうる各種 プロモーターの下流に結合した遺伝子コンストラクトを、 例えば、 ゥサギ受精卵 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夕ンパク質を高産生する D NA導入動物を作出できる。 このプロモ一夕一としては、 例えば、 ウィルス由 来プロモーター、 メタ口チォネイン等のュビキアスな発現プロモーターも使用し うるが、 好ましくは脳で特異的に発現する N G F遺伝子プロモーターゃェノラ一 ゼ遺伝子プロモータ一などが用いられる。 Using the DNA of the present invention, transgenic animals expressing the protein of the present invention can be prepared. Examples of animals include mammals (for example, rats, mice, rabbits, rabbits, sheep, pigeons, pigs, cats, dogs, monkeys, etc.) (hereinafter sometimes abbreviated as animals). When introducing the DNA of the present invention into a target animal, the DNA is used as a gene construct linked downstream of a promoter capable of being expressed in animal cells. Is generally advantageous. For example, when a DNA of the present invention derived from a egret is introduced, a gene construct in which the DNA of the present invention derived from an animal having a high homology with the DNA is linked to a downstream of various promoters capable of expressing the same in animal cells is used, for example. By introducing microinjection into eggs, a DNA-introduced animal of the present invention that produces high levels of protein can be produced. As such a promoter, for example, a ubiquitous expression promoter such as a virus-derived promoter or meta-mouth thionein may be used, but preferably, an NGF gene promoter, a genolases gene promoter, etc., which are specifically expressed in the brain. Is used.
受精卵細胞段階における本発明の D N Aの導入は、 対象動物の胚芽細胞および 体細胞の全てに存在するように確保される。 D N A導入後の作出動物の胚芽細胞 において本発明のタンパク質が存在することは、 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タンパク質を有することを意味する。 遺伝子 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 タンパク質を有する。  Introduction of the 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 animal. The presence of the protein of the present invention in the germinal cells of the animal after the introduction of DNA means that all the offspring of the animal have the protein of the present invention in all of the germinal and somatic cells. The progeny of such animals that have inherited the gene will have the protein of the invention in all of their germinal and somatic cells.
本発明の D N A導入動物は、 交配により遺伝子を安定に保持することを確認し て、 該 D NA保有動物として通常の飼育環境で飼育継代を行うことができる。 さ らに、 目的 D NAを保有する雌雄の動物を交配することにより、 導入遺伝子を相 同染色体の両方に持つホモザィゴート動物を取得し、 この雌雄の動物を交配する ことによりすべての子孫が該 D N Aを有するように繁殖継代することができる。 本発明の D NAが導入された動物は、 本発明のタンパク質が高発現させられて いるので、 本発明のタンパク質に対するァゴニストまたはアン夕ゴニス卜のスク リーニング用の動物などとして有用である。  After confirming that the DNA-introduced animal of the present invention stably retains the gene by breeding, the animal having the DNA can be reared in an ordinary breeding environment as the DNA-bearing animal. Furthermore, by crossing male and female animals having the target DNA, homozygous animals having the transgene on both homologous chromosomes are obtained, and by crossing the male and female animals, all the offspring will have the DNA Breeding to have Since the protein of the present invention is highly expressed in the animal into which the DNA of the present invention has been introduced, it is useful as an agonist or an animal for screening an gonist against the protein of the present invention.
本発明の D N A導入動物を、 組織培養のための細胞源として使用することもで きる。 例えば、 本発明の D NA導入マウスの組織中の D NAもしくは R NAを直 接分析するか、 あるいは遺伝子により発現された本発明のレセプタータンパク質 が存在する組織を分析することにより、 本発明の夕ンパク質について分析するこ とができる。 本発明のタンパク質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、 例えば、 脳や末梢組織由来のような一般に培養困難 な組織からの細胞の機能を研究することができる。 また、 その細胞を用いること により、 例えば、 各種組織の機能を高めるような医薬の選択も可能である。 また 、 高発現細胞株があれば、 そこから、 本発明のタンパク質を単離精製することも 可能である。 The DNA-introduced animal of the present invention can also be used as a cell source for tissue culture. For example, by directly analyzing DNA or RNA in the tissue of the DNA-introduced mouse of the present invention, or by analyzing the tissue in which the receptor protein of the present invention expressed by a gene is present, the present invention can be used. Analyze protein. The cells of a tissue having the protein of the present invention can be cultured by standard tissue culture techniques, and these can be used to study the function of cells from generally difficult-to-cultivate tissues such as those derived from brain and peripheral tissues. . Also, use the cells Thus, for example, it is possible to select a medicine that enhances the function of various tissues. Further, if there is a high expression cell line, the protein of the present invention can be isolated and purified therefrom.
本発明の DN A導入動物に試験化合物を投与し、 該動物の心機能、 心電図、 心 重量などを測定する。 心重量は心肥大のパラメ一夕一である。 具体的には体重当 たりの心臓重量、 体重当たりの左心室重量、 右心室重量当たりの左心室重量を算 出することによつて心臓構造を調べることができる。 心肥大が生じると上記パラ メーターは増加するため、 この増加を抑制することを指標として試験化合物を評 価することができる。 本発明の DNA導入動物に試験化合物を投与した後、 心筋 梗塞形成手術を行い、 該動物の心機能、 心電図、 心重量などを測定する。 また心 筋梗塞手術を行つた後、 梗塞層を秤量することによつて試験化合物の梗塞進展抑 制活性を調べることができる。 試験化合物の投与は、 梗塞形成手術後であっても よい。 また該動物と例えば SHRラットなど遺伝的高血圧モデルラッ卜と交配さ せ、 新しい心不全モデルを作成することができる。 このようにして作成した心不 全モデルに化合物を投与し、 該動物の心機能、 心電図、 心重量、 梗塞進展抑制活 性などを調べる。  A test compound is administered to the DNA-introduced animal of the present invention, and the heart function, electrocardiogram, heart weight, and the like of the animal are measured. Heart weight is the parameter of hypertrophy. Specifically, the heart structure can be examined by calculating the heart weight per body weight, the left ventricular weight per body weight, and the left ventricular weight per right ventricle weight. Since the above parameters increase when cardiac hypertrophy occurs, test compounds can be evaluated using the suppression of this increase as an index. After administering a test compound to the DNA-introduced animal of the present invention, a myocardial infarction is performed, and the heart function, electrocardiogram, heart weight, and the like of the animal are measured. In addition, after performing cardiomyocardial infarction surgery, the infarction progress-inhibiting activity of the test compound can be examined by weighing the infarct layer. Administration of the test compound may be after infarction surgery. In addition, the animal can be crossed with a genetic hypertension model rat such as an SHR rat to create a new heart failure model. The compound is administered to the cardiac failure model thus prepared, and the animal is examined for cardiac function, electrocardiogram, cardiac weight, activity for suppressing infarction progression, and the like.
〔9〕 ノックアウト動物  [9] Knockout animal
本発明は、 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 is inactivated and a non-human mammal deficient in expression of the DNA of the present invention.
すなわち、 本発明は、  That is, the present invention
(1) 本発明の DNAが不活性化された非ヒ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 ]3—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 the above (1), wherein the DNA is inactivated by introducing a reporter gene (eg, a 3-galactosidase enzyme gene derived from Escherichia 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発現不全非ヒ卜哺乳動物、 (6) a DNA-deficient non-human mammal in which the DNA of the present invention has been inactivated,
(7) 該 DNAがレポーター遺伝子 (例、 大腸菌由来の ]3—ガラクトシダーゼ遺 伝子) を導入することにより不活性化され、 該レポ一夕一遺伝子が本発明の DN Aに対するプロモーターの制御下で発現しうる上記 (6) 記載の非ヒト哺乳動物 (7) The DNA is inactivated by introducing a reporter gene (eg, a 3-galactosidase gene derived from Escherichia coli), and the reporter gene is transformed into the DN of the present invention. The non-human mammal according to the above (6), which can be expressed under the control of a promoter for A.
(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(9) The non-human mammal according to (8), wherein the rodent is a mouse; and
(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 D N Aに対するプロモータ一活性を促進 または阻害する化合物またはその塩のスクリーニング方法を提供する。 (10) Screening for a compound or a salt thereof that promotes or inhibits the activity of the promoter of the DNA of the present invention, which comprises administering a test compound to the animal according to (7) and detecting the expression of a reporter gene. Provide a way.
本発明の DNAが不活性化された非ヒ卜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 D N A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、 DN Aが実質的に本発明のタンパク質の発現 能を有さない (以下、 本発明のノックアウト DNAと称することがある) 非ヒト 哺乳動物の胚幹細胞 (以下、 ES細胞と略記する) をいう。  The non-human mammalian embryonic stem cells in which the DNA of the present invention has been inactivated are artificially mutated to the DNA of the present invention possessed by the non-human mammal to suppress the DNA expression ability, Alternatively, by substantially eliminating the activity of the protein of the present invention encoded by the DNA, the DNA does not substantially have the ability to express the protein of the present invention (hereinafter referred to as the knockout DNA of the present invention). Non-human mammalian embryonic stem cells (hereinafter abbreviated as ES cells).
非ヒ卜哺乳動物としては、 前記と同様のものが用いられる。  As the non-human mammal, those similar to the above can be 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 DNA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、 プロモーターあるいはェキソンの機能を破壌することに より本発明のノックァゥト 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 produced by, for example, shifting the codon reading frame or disrupting the function of the promoter or exon by these mutations.
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 (以下、 本発明の D NA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 ac Z (13—ガラクトシダ一ゼ遺伝子 ) 、 c a t (クロラムフエニコ一ルァセチルトランスフェラーゼ遺伝子) を代表 とするレポー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ィントロン部分に遺伝子の転写を終結させる DN A配列 ( 例えば、 p o 1 yA付加シグナルなど) を揷入し、 完全な mRNA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、 ターゲッティングベクターと略記する) を、 例えば相同組 換え法により該動物の染色体に導入し、 得られた ES細胞について本発明の DN^ A上あるいはその近傍の DNA配列をプローブとしたサザンハイブリダィゼ一シ ョン解析あるいは夕一ゲッティングベクタ一上の DNA配列と夕ーゲッティング ベクタ一作製に使用した本発明の D N A以外の近傍領域の D N A配列をプライマ —とした PC R法により解析し、 本発明のノックアウト ES細胞を選別すること により得ることができる。 Specific examples of the non-human mammalian embryonic stem cells of the present invention in which DNA is 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 non-human mammal DNA of the present invention is isolated and its exon portion is a drug resistance gene typified by a neomycin resistance gene, a hygromycin resistance gene, or lacZ (13-galactosidase gene), A DNA sequence that disrupts exon function by inserting a reporter gene, such as cat (chloramphenicylacetyltransferase gene), or terminates gene transcription in the intron portion between exons (for example, po 1 gene, resulting in disruption of gene synthesis by preventing the synthesis of complete mRNA Have a DNA sequence that was constructed so that A DNA strand (hereinafter, abbreviated as a targeting vector) is introduced into the chromosome of the animal by, for example, a homologous recombination method, and the obtained ES cell is treated with a DNA sequence on or near DN ^ A of the present invention as a probe. PCR method using 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 preparation as primers By analyzing 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 D N Aを不活化させる元の E S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、 また公知の Ev ansと Kaufman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 E S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 学的背景がはつ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 ES細胞を取得するなどの目的で例えば、 C57 BLZ6マウスや C 57 BLZ6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 BLZ6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 BLZ6マウスを背景に持つので、 これを用いて得られた E S細胞は病態モデルマウスを作出したとき、 C 57 BLZ6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 BL/ 6マウスに代えることが可能である 点で有利に用い得る。  As the ES cells from which the DNA of the present invention is inactivated by the homologous recombination method or the like, for example, those already established as described above may be used, or the known methods of Evans and Kaufman may be used. It may be newly established according to. For example, in the case of mouse ES cells, currently, 129 ES cells are generally used, but since the immunological background is not clear, a pure line that substitutes them can be used for immunological inheritance. For example, for the purpose of obtaining ES cells with a clear background, BDFi mice (C57BLZ6 and DBAZ2 and C57BLZ6) BDFi mice can be used satisfactorily.Because BDFi mice have a high number of eggs collected and their eggs are durable, they have C57BLZ6 mice as their background. The obtained ES cells can be advantageously used when a pathological model mouse is created, since the genetic background can be replaced by C57BL / 6 mice by backcrossing with C57BLZ6 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 establishing ES cells, 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。  Although either male or female ES cells may be used, male ES cells are generally more convenient for producing a germline chimera. 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コロニー程度の ES細胞数 (約 50個) で済むので、 培養 初期における E S細胞の第一次セレクションを雌雄の判別で行なうことが可能で あり、 早期に雄細胞の選定を可能にしたことにより培養初期の手間は大幅に削減 できる。 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. Using this method, it has traditionally required about 10 6 cell numbers to perform a karyotype analysis. On the other hand, the number of ES cells in one colony (approximately 50) is sufficient, so the primary selection of ES cells in the early stage of culture can be performed by gender discrimination. By making selection possible, labor in the initial stage of culture can be significantly reduced.
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 E S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. It is desirable that the number of chromosomes in the obtained ES cells is 100% of the normal number. However, if it is difficult due to physical operations at the time of establishment, after knocking out the gene of the ES cells, normal cells (for example, in mice) It is desirable to clone again into cells with 2 n = 40 chromosomes.
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、 S TO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- 1000 OU/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 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. 00 1 - 0. 5 %トリプシン/ 0. 1— 5mM EDTA, 好ましくは約 0. 1 %トリプシ ン ZlmM EDTA) 処理により単細胞化し、 新たに用意したフィーダ一細胞 上に播種する方法などがとられる。 このような継代は、 通常 1一 3日毎に行なう が、 この際に細胞の観察を行い、 形態的に異常な細胞が見受けられた場合はその 培養細胞は放棄することが望まれる。  Embryonic stem cell lines obtained in this way usually have very good proliferative properties, but they must be carefully subcultured because they tend to lose their ability to generate individuals. For example, on a suitable feeder cell, such as STO fibroblasts, in a carbon dioxide incubator (preferably 5% carbon dioxide, 95% air or 5%) in the presence of LIF (1-1000 OU / ml). Culture at about 37% oxygen, 5% carbon dioxide, 90% air) at the time of subculture. At the time of subculture, for example, trypsin / EDTA solution (usually 0.001-0.5% trypsin / 0 1-5mM EDTA (preferably about 0.1% trypsin ZlmM EDTA) is used to convert the cells into single cells and seed them on freshly prepared feeder cells. Such subculture is usually performed every 11 to 13 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. Kaufm an, ネイチヤー (Nature) 第 292巻、 154頁、 1981年; G. R. Martin プロシーデ イングス 'ォブ'ナショナル 'アカデミー ·ォブ ·サイエンス 'ュ一エスェ一 ( 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 'ェクスペリメンタル ·モル フォロジ一、 第 87巻、 27頁、 1985年〕 、 本発明の E S細胞を分化させて得られる 本発明の 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 can be cultured in monolayers at high densities or in suspension cultures to form cell clumps under appropriate conditions to produce various types of cells such as parietal, visceral, and cardiac muscles. [MJ Evans and MH Kaufman, Nature 292, 154, 1981; GR Martin Proceedings 'Ob' National 'Academy of Science' Natl. Acad. Sci. USA, Vol. 78, p. 7634, 1981; TC Doetschman et al., Journal of Obelimborgologi 'and' Experimental Morphology, Vol. 87, 27. P., 1985], the DNA-deficient cells of the present invention obtained by differentiating the ES cells of the present invention are cells of the protein of the present invention in the mouth of the intestine Useful in biological studies.
本発明の D NA発現不全非ヒ卜哺乳動物は、 該動物の mR NA量を公知の方法 を用いて測定して間接的にその発現量を比較することにより、 正常動物と区別す ることが可能である。  The non-human mammal deficient in DNA expression of the present invention can be distinguished from a normal animal by measuring the mRNA level of the animal using a known method and indirectly comparing the expression level. It is possible.
該非ヒ卜哺乳動物としては、 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 NA発現不全非ヒト哺乳動物は、 例えば、 前述のようにして作製し たターゲッティングベクターをマウス胚幹細胞またはマウス卵細胞に導入し、 導 入により夕ーゲッティングベクターの本発明の 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 N Aを ノックアウトさせることができる。  The non-human mammal deficient in DNA expression of the present invention can be obtained, for example, by introducing the targeting vector prepared as described above into a mouse embryonic stem cell or a mouse egg cell, and introducing the DNA of the present invention into the evening targeting vector. The inactivated DNA sequence can be knocked out by homologous recombination of the DNA of the present invention on the chromosome of mouse embryonic stem cells or mouse egg cells by homologous recombination. it can.
本発明の D NAがノックアウトされた細胞は、 本発明の D NA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ーション解析またはター ゲッティングベクタ一上の D NA配列と、 タ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配列とをプライマ一とした 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 NA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細胞期の非ヒト哺乳動物胚ま たは胚盤胞に注入し、 作製したキメラ胚を偽妊娠させた該非ヒト哺乳動物の子宮 に移植する。 作出された動物は正常な本発明の D NA座をもつ細胞と人為的に変 異した本発明の D NA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。 該キメラ動物の生殖細胞の一部が変異した本発明の D N A座をもつ場合、 この ようなキメラ個体と正常個体を交配することにより得られた個体群より、 全ての 組織が人為的に変異を加えた本発明の D NA座をもつ細胞で構成された個体を、 例えば、 コートカラー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。  Cells in which the DNA of the present invention has been knocked out are combined with a DNA sequence on a Southern hybridization analysis or a targeting vector using a DNA sequence on or near the DNA of the present invention as a probe, and a targeting vector. The determination can be made by PCR analysis using the DNA sequence of the neighboring region other than the DNA of the present invention derived from the mouse used as the primer. When a non-human mammalian embryonic stem cell is used, the cell line in which the DNA of the present invention has been inactivated is cloned by homologous gene recombination, and the cell line is cloned at an appropriate time, for example, at the 8-cell stage. The chimeric embryo is injected into a human mammalian embryo or blastocyst, and the resulting chimeric embryo is transplanted into the uterus of the pseudo-pregnant non-human mammal. The produced animal is a chimeric animal composed of both cells having the normal DNA locus of the present invention and cells having the artificially altered DNA locus of the present invention. When a part of the germ cells of the chimeric animal has a mutated DNA locus of the present invention, all tissues are artificially mutated from a population obtained by crossing such a chimeric individual with a normal individual. It can be obtained by selecting individuals composed of cells having the added DNA locus of the present invention, for example, by judging coat color or the like. The individuals obtained in this manner are usually individuals with heterozygous expression of the protein of the present invention, which are mated with individuals with 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 N A溶液を注入することによりターゲッティングベクタ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、 これらのトランスジ エニック非ヒト哺乳動物に比べて、 遺伝子相同組換えにより本発明の D NA座に 変異のあるものを選択することにより得られる。 When using egg cells, for example, microinjection into the nucleus of the egg cell A transgenic non-human mammal in which the targeting vector has been introduced into the chromosome can be obtained by injecting a DNA solution into the chromosome of the present invention. It can be obtained by selecting those with mutations at the NA locus.
このようにして本発明の D NAがノックアウトされている個体は、 交配により 得られた動物個体も該 D N Aがノックアウトされていることを確認して通常の飼 育環境で飼育継代を行なうことができる。  In the individual knocked out of the DNA of the present invention in this manner, it is possible to confirm that the DNA has been knocked out in the animal individual obtained by mating, and to carry out rearing in an ordinary rearing environment. 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ート動物を取得しうる。 得られたホモザィゴ —ト動物は、 母親動物に対して、 正常個体 1, ホモザィゴート複数になるような 状態で飼育することにより効率的に得ることができる。 ヘテロザィゴ一卜動物の 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 ロザィゴ一卜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 a plurality of homozygous animals 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 NA 発現不全非ヒ卜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 a non-human mammal deficient in expressing the DNA of the present invention.
また、 本発明の D NA発現不全非ヒト哺乳動物は、 本発明のタンパク質により 誘導され得る種々の生物活性を欠失するため、 本発明のタンパク質の生物活性の 不活性化を原因とする疾病のモデルとなり得るので、 これらの疾病の原因^ E明及 び治療法の検討に有用である。  In addition, since the non-human mammal deficient in expression of the DNA of the present invention lacks various biological activities that can be induced by the protein of the present invention, a disease caused by inactivation of the biological activity of the protein of the present invention. It can be a model, so it is useful for studying the causes of these diseases and examining treatment methods.
( 9 a ) 本発明の D NAの欠損や損傷などに起因する疾病に対して治療 ·予防効 果を有する化合物のスクリーニング方法  (9a) A method for screening a compound having a therapeutic / preventive effect on diseases caused by DNA deficiency or damage of the present invention.
本発明の D NA発現不全非ヒト哺乳動物は、 本発明の D NAの欠損や損傷など に起因する疾病に対して治療 ·予防効果を有する化合物のスクリーニングに用い ることができる。  The non-human mammal deficient in expression of the DNA of the present invention can be used for screening for a compound having a therapeutic / preventive effect against diseases caused by the deficiency or damage of the DNA of the present invention.
すなわち、 本発明は、 本発明の D NA発現不全非ヒト哺乳動物に試験化合物を 投与し、 該動物の変化を観察 ·測定することを特徴とする、 本発明の D N Aの欠 損や損傷などに起因する疾病に対して治療 ·予防効果を有する化合物またはその 塩のスクリーニング方法を提供する。 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。 That is, the present invention is characterized in that a test compound is administered to a non-human mammal deficient in expression of the DNA of the present invention, and changes in the animal are observed and measured. Provided is a method for screening a compound or a salt thereof having a therapeutic or preventive effect on a disease caused by the disease. The non-human mammal deficient in DNA expression of the present invention used in the screening method includes the same ones as described above.
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、 公知の化合物であつ てもよい。  Test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and plasma.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 、 無処理の对照動物と比較し、 該動物の各器官、 組織、 疾病の症状などの変化を 指標として試験化合物の治療 ·予防効果を試験することができる。.  Specifically, a non-human mammal deficient in expression of the DNA of the present invention is treated with a test compound and compared with an untreated control animal, and changes in the organs, tissues, disease symptoms, etc. of the animal are used as indices. The test compound can be tested for its therapeutic and prophylactic effects. .
試験動物を試験化合物で処理する方法としては、 例えば、 経口投与、 静脈注射 などが用いられ、 試験動物の症状、 試験化合物の性質などにあわせて適宜選択す ることができる。 また、 試験化合物の投与量は、 投与方法、 試験化合物の性質な どにあわせて適宜選択することができる。  As a method for treating a test animal with a test compound, for example, oral administration, intravenous injection and the like are used, and it can be appropriately selected according to the symptoms of the test animal, the properties of the test compound, and the like. The dose of the test compound can be appropriately selected according to the administration method, the properties of the test compound, and the like.
例えば、 心機能の低下を特徴とする疾病 (例、 心筋梗塞後の心不全;狭心症; 心筋症;狭心症、 心筋症などの疾患に由来する心不全などの心疾患など) 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試験化合物を投与し、 該動物の心機能、 心電図、 心重量 などを測定する。 心重量は心肥大のパラメ一ターである。 具体的には体重当たり の心臓重量、 体重当たりの左心室重量、 右心室重量当たりの左心室重量を算出す ることによって心臓構造を調べることができる。 心肥大が生じると上記パラメ一 夕一は増加するため、 この増加を抑制することを指標として試験化合物を評価す ることができる。 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 た後、 心筋梗塞形成手術を行い、 該動物の心機能、 心電図、 心重量などを測定す る。 また心筋梗塞手術を行った後、 梗塞層を秤量することによって試験化合物の 梗塞進展抑制活性を調べることができる。 試験化合物の投与は、 梗塞形成手術後 であってもよい。 また該動物と例えば S H Rラッ卜など遺伝的高血庄モデルラッ 卜と交配させ、 新しい心不全モデルを作成することができる。 このようにして作 成した心不全モデルに化合物を投与し、 該動物の心機能、 心電図、 心重量、 梗塞 進展抑制活性などを調べる。 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、 上記スクリー二 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。 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。 For example, to prevent diseases characterized by reduced cardiac function (eg, heart failure after myocardial infarction; angina pectoris; cardiomyopathy; heart diseases such as heart failure derived from diseases such as angina pectoris and cardiomyopathy). · When screening for a compound having a therapeutic effect, a test compound is administered to a non-human mammal deficient in the expression of DNA of the present invention, and the heart function, electrocardiogram, heart weight and the like of the animal are measured. Heart weight is a parameter of cardiac hypertrophy. Specifically, the heart structure can be examined by calculating the heart weight per body weight, the left ventricular weight per body weight, and the left ventricular weight per right ventricle weight. Since the above parameters increase when cardiac hypertrophy occurs, test compounds can be evaluated using the suppression of this increase as an index. After administering the test compound to the non-human mammal deficient in DNA expression of the present invention, a myocardial infarction is performed, and the heart function, electrocardiogram, heart weight, and the like of the animal are measured. After the myocardial infarction operation, the infarct growth inhibitory activity of the test compound can be examined by weighing the infarct layer. Administration of the test compound may be post-infarct surgery. In addition, a new heart failure model can be created by crossing the animal with a genetically high blood model rat such as an SHR rat. The compound is administered to the heart failure model thus prepared, and the animal is examined for cardiac function, electrocardiogram, heart weight, infarction progress inhibitory activity and the like. The compound obtained by using the screening method is a compound selected from the test compounds described above, and has a prophylactic / therapeutic effect against a disease caused by deficiency or damage of the protein of the present invention. It can be used as a safe and low toxic prophylactic and therapeutic agent. Further, a compound derived from the compound obtained by the above-mentioned screening can also be used. 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, etc.). And the like, and a physiologically acceptable acid addition salt is particularly preferable. Such salts include, for example, salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.), or organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid) Salts with acids, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc. are used.
該スクリーニング方法で得られた化合物またはその塩を含有する医薬は、 前記 した本発明のタンパク質を含有する医薬と同様にして製造することができる。 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 はその他の哺乳動物 (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。 該化合物またはその塩の投与量は、 対象疾患、 投与対象、 投与ルートなどによ り差異はあるが、 例えば、 該化合物を経口投与する場合、 一般的に成人 (体重 6 O kgとして) の心疾患の患者においては、 一日につき該化合物を約 0. 1〜1 0 Omg、 好ましくは約 1. 0〜5 Omg、 より好ましくは約 1. 0〜20mg 投与する。 非経口的に投与する場合は、 該化合物の 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 (60 kgとして) の心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程度、 好ましくは約 0. l〜20mg程度、 より好ましくは約 0. 1 〜1 Omg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 、 60 kg当たりに換算した量を投与することができる。  A drug containing the compound or a salt thereof obtained by the screening method can be produced in the same manner as the above-mentioned drug containing the protein of the present invention. The preparations obtained in this way are safe and low toxic, and can be used, for example, in humans or other mammals (eg, rats, mice, guinea pigs, egrets, higgs, bush, foxes, dogs, 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 is orally administered, it is generally used in adults (with a body weight of 6 kg). In a patient suffering from a disease, the compound is administered at about 0.1 to 10 Omg, preferably about 1.0 to 5 Omg, more preferably about 1.0 to 20 mg per day. When administered parenterally, the single dose of the compound varies depending on the administration subject, target disease, and the like. For example, the compound is usually administered in the form of an injection in an adult (as 60 kg) patient with heart disease. About 0.01 to 30 mg, preferably about 0.1 to 20 mg, more preferably about 0.1 to 1 mg per day, by intravenous injection of the compound. is there. In the case of other animals, it can be administered in an amount converted per 60 kg.
(9 b) 本発明の DN Aに対するプロモー夕一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 (9b) a compound that promotes or inhibits the activity of promoter of DNA of the present invention. Screening method
本発明は、 本発明の DNA発現不全非ヒト哺乳動物に、 試験化合物を投与し、 レポーター遺伝子の発現を検出することを特徴とする本発明の DN Aに対するプ 口モータ—の活性を促進または阻害する化合物またはその塩のスクリーニング方 法を提供する。  The present invention provides a test compound administered to a non-human mammal deficient in expression of a DNA of the present invention, and detects or enhances the expression of a reporter gene. A method for screening a compound or a salt thereof.
上記スクリーニング方法において、 本発明の DN A発現不全非ヒト哺乳動物と しては、 前記した本発明の DNA発現不全非ヒト哺乳動物の中でも、 本発明の D NAがレポーター遺伝子を導入することにより不活性化され、 該レポーター遺伝 子が本発明の D N Aに対するプロモーターの制御下で発現しうるものが用いられ る。  In the above-described screening method, the non-human mammal deficient in expression of the DNA of the present invention may be one of the above-mentioned non-human mammals deficient in expression of the DNA of the present invention, in which the DNA of the present invention is not introduced by introducing a reporter gene. Those which are activated and which can express the reporter gene under the control of the 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.
レポ一夕一遺伝子としては、 前記と同様のものが用いられ、 )3—ガラクトシダ —ゼ遺伝子 (1 ac Z) 、 可溶性アルカリフォスファターゼ遺伝子またはルシフ ェラ一ゼ遺伝子などが好適である。  As the repo overnight gene, the same gene as described above is used, and a) -galactosidase gene (1 ac Z), a soluble alkaline phosphatase gene or a luciferase gene is suitable.
本発明の DNAをレポーター遺伝子で置換された本発明の DN A発現不全非ヒ ト哺乳動物では、 レポーター遺伝子が本発明の DN Aに対するプロモーターの支 配下に存在するので、 レポーター遺伝子がコードする物質の発現をトレースする ことにより、 プロモーターの活性を検出することができる。  In a non-human mammal deficient in expression of the DNA of the present invention in which the DNA of the present invention has been replaced with a reporter gene, the reporter gene is under the control of the promoter for the DNA of the present invention. By tracing the expression, the activity of the promoter can be detected.
例えば、 本発明のタンパク質をコードする DNA領域の一部を大腸菌由来の ]3 一ガラクトシダ一ゼ遺伝子 (1 a c Z) で置換している場合、 本来、 本発明の夕 ンパク質の発現する組織で、 本発明のタンパク質の代わりに j3—ガラクトシダ一 ゼが発現する。 従って、 例えば、 5—プロモー 4一クロ口— 3—^ f ンドリル— j8 —ガラクトビラノシド (X— g a l) のような j8—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 (PBS) で洗浄後、 X— g a 1を含む染色液で、 室温または 37°C付近 で、 約 30分ないし 1時間反応させた後、 組織標本を ImM EDTA/PBS 溶液で洗浄することによって、 ;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 NAを検出しても よい。 For example, when a part of the DNA region encoding the protein of the present invention is replaced with a [3-galactosidase gene (1 ac Z) derived from Escherichia coli, a tissue that originally expresses the protein of the present invention may be used. However, j3-galactosidase is expressed instead of the protein of the present invention. Thus, for example, by staining with a reagent that serves as a substrate for j8-galactosidase, such as 5-promote 4-monocloth-3- ^ f-indolyl-j8-galactovyranoside (X-gal) The state of expression of the protein of the present invention in an animal body can be easily observed. Specifically, the protein-deficient mouse of the present invention or a tissue section thereof is fixed with dartalaldehyde or the like, washed with phosphate buffered saline (PBS), and then stained with X-ga1 at room temperature or at 37 ° C. After reacting for about 30 minutes to 1 hour at about ° C, the tissue sample was washed with ImM EDTA / PBS solution to stop the 3-galactosidase reaction and to observe the coloration. I just think. In addition, mRNA encoding 1 ac Z may be detected according to a conventional method.
上記スクリーニング方法を用いて得られる化合物またはその塩は、 上記した試 験化合物から選ばれた化合物であり、 本発明の D N Aに対するプロモーター活性 を促進または阻害する化合物である。  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 DNA promoter activity of the present invention.
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 レ^ この様な塩としては、 例えば、 無機酸 (例えば、 塩酸、 リン酸、 臭化水素酸 、 硫酸など) との塩、 あるいは有機酸 (例えば、 酢酸、 ギ酸、 プロピオン酸、 フ マル酸、 マレイン酸、 コハク酸、 酒石酸、 クェン酸、 リンゴ酸、 蓚酸、 安息香酸 、 メタンスルホン酸、 ベンゼンスルホン酸など) との塩などが用いられる。 本発明の D N Aに対するプロモータ一活性を促進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促進し、 該タンパク質の機能を促進することができ るので、 例えば、 心機能の低下を特徴とする疾病 (例、 心筋梗塞後の心不全;狭 心症;心筋症;狭心症、 心筋症などの疾患に由来する心不全などの心疾患など) などの予防 ·治療剤などの医薬として有用である。  The compound obtained by the screening method may form a salt. Examples of the salt of the compound include physiologically acceptable acids (eg, inorganic acids) and bases (eg, organic acids). Salts are used, and physiologically acceptable acid addition salts are particularly preferred. ^ Such salts include, for example, inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.) Salts or salts with organic acids (such as 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.) Are used. Since the compound of the present invention or a salt thereof which promotes the promoter activity for DNA can promote the expression of the protein of the present invention and promote the function of the protein, for example, it is characterized by a decrease in cardiac function. It is useful as a drug for the prevention and treatment of diseases (eg, heart failure after myocardial infarction; angina pectoris; cardiomyopathy; heart diseases such as heart failure derived from diseases such as angina pectoris and cardiomyopathy).
また、 本発明の D N Aに対するプロモータ一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、 例えば心機能の低下を特徴とする疾病 (例、 心筋梗塞後の心不全 ;狭心症;心筋症;狭心症、 心筋症などの疾患に由来する心不全などの心疾患な ど) などの予防 ·治療剤などの医薬として有用である。  In addition, the compound of the present invention or a salt thereof that inhibits the activity of a promoter for DNA can inhibit the expression of the protein of the present invention and inhibit the function of the protein. It is useful as a drug for the prevention and treatment of diseases (eg, heart failure after myocardial infarction; angina pectoris; cardiomyopathy; heart diseases such as heart failure derived from diseases such as angina pectoris and cardiomyopathy). .
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 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 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 はその他の哺乳動物 (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 DNAに対するプロモ一夕一活性を促進する 化合物を経口投与する場合、 一般的に成人 (体重 60 k gとして) の心疾患の患 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 DNA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 (60 kgとして) の心疾患の患者に投与する場合、 一日につき 該化合物を約 0. 01〜3 Omg程度、 好ましくは約 0. l〜20mg程度、 よ り好ましくは約 0. 1〜1 Omg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、 60 kg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 DN Aに対するプロモーター活性を阻害する化合物を 経口投与する場合、 一般的に成人 (体重 6 O kgとして) の心疾患の患者におい ては、 一日につき該化合物を約 0. 1〜10 Omg、 好ましくは約 1. 0〜50 mg、 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 (6 O kgとして) の心疾患の患者に投与する場合、 一日につき該化合物 を約 0. 01〜3 Omg程度、 好ましくは約 0. 1〜 2 Omg程度、 より好まし くは約 0. 1〜1 Omg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、 60 k g当たりに換算した量を投与することができる。 The preparations obtained in this way are safe and low toxic, and thus can be used, for example, in humans or other mammals (eg, rats, mice, guinea pigs, egrets, sheep, Pigs, pests, pomas, 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 overnight activity against DNA is orally administered, Generally, in an adult (assuming a body weight of 60 kg) patient with heart 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 per day is used. Administer. 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 promotes the promoter activity for DNA of the present invention is usually administered in the form of an injection to an adult. When administered to a patient with a heart disease of about 60 kg (as 60 kg), the compound is administered in an amount of about 0.01 to 3 Omg per day, preferably about 0.1 to 20 mg, more preferably about 0.1 to 1 mg per day. It is convenient to administer about Omg 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 6 O kg) heart disease patients, the compound is reduced to about 0 per day. 1 to 10 Omg, preferably about 1.0 to 50 mg, more preferably about 1.0 to 20 mg. When administered parenterally, the single dose of the compound varies depending on the subject of administration, the target disease, and the like. For example, the compound of the present invention that inhibits the promoter activity on DNA may be administered in the form of an injection. When administered to a normal adult (as 6 O kg) patient with a heart disease, the compound is administered in an amount of about 0.01 to 3 Omg per day, preferably about 0.1 to 2 Omg, more preferably about 0.1 to 2 Omg. It is convenient to administer about 0.1 to 1 Omg 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 DNA発現不全非ヒト哺乳動物は、 本発明の DNAに対 するプロモーターの活性を促進または阻害する化合物またはその塩をスクリ一二 ングする上で極めて有用であり、 本発明の DN 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 for the DNA of the present invention. Investigating or preventing the causes of various diseases caused by insufficient expression of DNA can greatly contribute to the development of therapeutic drugs.
また、 本発明のタンパク質のプロモータ一領域を含有する DNAを使って、 そ の下流に種々のタンパクをコードする遺伝子を連結し、 これを動物の卵細胞に注 入していわゆるトランスジエニック動物 (遺伝子導入動物) を作成すれば、 特異 的にそのポリペプチドを合成させ、 その生体での作用を検討することも可能とな る。 さらに上記プロモ一夕一部分に適当なレポ一夕遺伝子を結合させ、 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。 本明細書および図面において、 塩基やアミノ酸などを略号で表示する場合、 IU PAC-IUB Commission on Biochemical Nomenclatureによる略号あるいは当該分野 における慣用略号に基づくものであり、 その例を下記する。 またアミノ酸に関し 光学異性体があり得る場合は、 特に明示しなければ L体を示すものとする。 In addition, using a DNA containing a 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 (gene). Introduced animal) It is also possible to synthesize the polypeptide in an appropriate manner and to examine the action in the living body. Furthermore, by binding an appropriate repo overnight gene to a part of the above-mentioned promoter and establishing a cell line in which the gene is expressed, the action of specifically promoting or suppressing the ability of the protein itself of the present invention to produce in the body is achieved. It can be used as a search system for low molecular weight compounds with. In the present specification and drawings, bases, amino acids, and the like are indicated by abbreviations based on the abbreviations of the IU PAC-IUB Commission on Biochemical Nomenclature or commonly used abbreviations in the art, and examples thereof are described below. When amino acids may have optical isomers, L-form shall be indicated unless otherwise specified.
DNA デォキシリポ核酸 DNA deoxylipo nucleic acid
c DNA 相補的デォキシリポ核酸  c DNA complementary deoxylipo nucleic acid
A アデニン  A adenine
T チミン  T thymine
G グァニン  G Guanin
C C
RNA リボ核酸 RNA ribonucleic acid
mRNA ―リポ核酸  mRNA-liponucleic acid
dATP '三リン酸  dATP 'Triphosphate
dTTP デォキシチミジン三リン酸  dTTP Deoxythymidine triphosphate
dGTP デォキシグァノシン三リン酸  dGTP Deoxyguanosine triphosphate
dCTP デォキシシチジン三リン酸  dCTP Deoxycytidine triphosphate
ATP アデノシン三リン酸  ATP Adenosine triphosphate
EDTA エチレンジァミン四酢酸  EDTA ethylenediaminetetraacetic acid
SDS ドデシル硫酸ナトリウム  SDS sodium 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 L e u Va 1 Valine L eu
I 1 e  I 1 e
S e r セリン  S e r serine
Th r スレオニン  Th r threonine
Cy s  Cy s
Me t メチォニン  Me t Methionin
G 1 u グルタミン酸  G 1 u Glutamic acid
As p ァスパラギン酸  As p Aspartic acid
L y s リジン  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 トリプ卜ファン  T r p Triple fan
P r o プロリン  Pro proline
A s n ァスパラギン  A s n asparagine
G 1 n グルタミン  G 1 n Glutamine
p G 1 u ピログルタミン酸 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。  pG1u pyroglutamic acid 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 tethyl group
Bu ブチル基  Bu butyl group
P h フエニル基  P h phenyl group
TC チアゾリジン— 4 (R) 一力ルポキサミド基  TC thiazolidine-4 (R) one-pot lipoxamide group
T o s p—トルエンスルフォニル  T os p—toluenesulfonyl
CHO ホルミル B z 1 CHO Holmill B z 1
C 12-B z 1 2, 6—ジクロロべンジル C 1 2 -B z 1 2, 6- dichloro base Njiru
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 -Z 2—クロ口べンジルォキシカルボニル  C 1 -Z 2 -Brozen benzyloxycarbonyl
B r - Z 2一ブロモベンジルォキシカルボニル  B r -Z 2 bromobenzyloxycarbonyl
B o c t一ブトキシカルボニル  B oc t-butoxycarbonyl
DNP ジニトロフエニル  DNP dinitrophenyl
T r t 卜リテル  T r t Trittel
Bum t一ブトキシメチル  Bum t-butoxymethyl
Fmo c N— 9 _フルォレニルメトキシカルポニル  Fmo c N— 9 _fluorenylmethoxycarbonyl
HOB t 1—ヒドロキシベンズトリアゾ一ル  HOB t 1—Hydroxybenztriazole
HOOB t 3, 4—ジヒドロー 3—ヒドロキシ一 4一ォキソ一  HOOB t 3,4-Dihydro-3-hydroxy-1 4-oxo-1
1, 2, 3—ベンゾ卜リアジン  1,2,3-benzotriazine
HONB 1 -ヒドロキシ -5-ノルポルネン -2, 3-ジカルボキシィ ド、 DCC 本願明細書の配列表の配列番号は、 以下の配列を示す。 〔配列番号: 1〕  HONB 1-Hydroxy-5-norporene-2,3-dicarboxide, DCC SEQ ID NOs in the Sequence Listing of the present specification indicate the following sequences. [SEQ ID NO: 1]
実施例 1で得られた本発明のタンパク質 (ラッ卜由来) のアミノ酸配列を示す  2 shows the amino acid sequence of the protein of the present invention (derived from rat) obtained in Example 1.
〔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コードする DNA (遺伝子) の塩基配 列を示す。  This shows the base sequence of DNA (gene) encoding the amino acid sequence represented by SEQ ID NO: 1.
〔配列番号: 3〕  [SEQ ID NO: 3]
実施例 1で用いられたプライマーの塩基配列を示す。  1 shows the nucleotide sequence of a primer used in Example 1.
〔配列番号: 4〕  [SEQ ID NO: 4]
実施例 1で用いられたプライマーの塩基配列を示す。 〔配列番号: 5〕 1 shows the nucleotide sequence of a primer used in Example 1. [SEQ ID NO: 5]
実施例 1で用いられたプライマーの塩基配列を示す。 1 shows the nucleotide sequence of a primer used in Example 1.
〔配列番号: 6〕  [SEQ ID NO: 6]
実施例 1で用いられたプライマーの塩基配列を示す。 1 shows the nucleotide sequence of a primer used in Example 1.
〔配列番号: 7〕  [SEQ ID NO: 7]
実施例 1で得られた遺伝子断片の塩基配列を示す。  1 shows the nucleotide sequence of the gene fragment obtained in Example 1.
〔配列番号: 8〕  [SEQ ID NO: 8]
実施例 1で用いられたプライマーの塩基配列を示す。  1 shows the nucleotide sequence of a primer used in Example 1.
〔配列番号: 9〕  [SEQ ID NO: 9]
実施例 1で用いられたプライマーの塩基配列を示す。  1 shows the nucleotide sequence of a primer used in Example 1.
〔配列番号: 10〕  [SEQ ID NO: 10]
実施例 1で用いられたプライマーの塩基配列を示す。  1 shows the nucleotide sequence of a primer used in Example 1.
〔配列番号: 1 1〕  [SEQ ID NO: 11]
実施例 1で用いられたプライマーの塩基配列を示す。  1 shows the nucleotide sequence of a primer used in Example 1.
〔配列番号: 12〕  [SEQ ID NO: 12]
実施例 1で得られた配列番号: 1で表されるアミノ酸配列をコードする DN A (遺伝子) の非コード領域を含む全塩基配列を示す。  1 shows the entire nucleotide sequence including the non-coding region of DNA (gene) encoding the amino acid sequence represented by SEQ ID NO: 1 obtained 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の塩基配列を示す。  1 shows the nucleotide sequence of a primer 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の塩基配列を示す。  1 shows the nucleotide sequence of a primer used in Example 1.
〔配列番号: 15〕  [SEQ ID NO: 15]
実施例 2で用いられたプライマーの塩基配列を示す。  3 shows the nucleotide sequence of a primer used in Example 2.
〔配列番号: 16〕  [SEQ ID NO: 16]
実施例 2で用いられたプライマーの塩基配列を示す。  3 shows the nucleotide sequence of a primer used in Example 2.
〔配列番号: 17〕  [SEQ ID NO: 17]
実施例 2で用いられたプローブの塩基配列を示す。  4 shows the nucleotide sequence of a probe used in Example 2.
〔配列番号: 18〕  [SEQ ID NO: 18]
配列番号: 2で表される塩基配列中、 5' 末端から第 157番目から第 165 番目の部分塩基配列を示す。 In the nucleotide sequence represented by SEQ ID NO: 2, from the 157th to the 165th from the 5 'end The third partial base sequence is shown.
〔配列番号: 19〕  [SEQ ID NO: 19]
配列番号: 2で表される塩基配列中、 5' 末端から第 391番目から第 402 番目の部分塩基配列を示す。  In the nucleotide sequence represented by SEQ ID NO: 2, this shows the 391st to 402nd partial nucleotide sequence from the 5 'end.
〔配列番号: 20〕  [SEQ ID NO: 20]
配列番号: 2で表される塩基配列中、 5' 末端から第 490番目から第 495 番目の部分塩基配列を示す。  In the nucleotide sequence represented by SEQ ID NO: 2, this shows the 490th to 495th partial nucleotide sequence from the 5 'end.
〔配列番号: 21〕  [SEQ ID NO: 21]
配列番号: 2で表される塩基配列中、 5' 末端から第 796番目から第 804 番目の部分塩基配列を示す。  In the nucleotide sequence represented by SEQ ID NO: 2, this shows the 796th to 804th partial nucleotide sequence from the 5 'end.
〔配列番号: 22〕  [SEQ ID NO: 22]
配列番号: 2で表される塩基配列中、 5' 末端から第 829番目から第 846 番目の部分塩基配列を示す。 後述の実施例 1で得られた形質転換体ェシエリヒア ·コリ (Es che r i c h i a co l i) DH5 o;/pTB2165は、 2000年 10月 19日から 日本国茨城県つくば巿東 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 (旧:通商産業省 工業技術院生命工学工業技術研究所 (NI BH) ) に寄託番号 FERM BP— 7327として、 2000年 9月 26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 (I FO ) に寄託番号 I FO 16481として寄託されている。 実施例  In the nucleotide sequence represented by SEQ ID NO: 2, this shows the 829th to 846th partial nucleotide sequence from the 5 'end. The transformant Escherichia coli DH5 o; / pTB2165 obtained in Example 1 described below has been used since October 19, 2000 at 1-1-1 Tsukuba-Higashi, Ibaraki, Japan 1 6 (Postal Code 305-8566) at the National Institute of Advanced Industrial Science and Technology (AIST), the Patent Organism Depositary Center (formerly Ministry of International Trade and Industry, National Institute of Advanced Industrial Science and Technology (NI BH)) under the deposit number FERM BP-7327. From September 26, 2000, deposited with the Fermentation Research Institute (I FO) at 2-173, Jusanhoncho, Yodogawa-ku, Osaka-shi, Osaka (zip code 532-8686) as deposit number I FO 16481 Have been. Example
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。 なお、 大腸菌を用いての遺伝子操作法は、 モレキユラ 一 ·クロ一ニング (Molecular cloning) , 2nd, J. Sambrook et al. , Cold Spri ng Harbor Lab. Press, 1989年に記載されている方法に従った。 実施例 1 Hereinafter, the present invention will be described more specifically with reference to Examples, but the present invention is not limited thereto. Genetic manipulation using Escherichia coli is performed according to the method described in Molecular cloning, 2nd, J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989. Was. Example 1
(1) 心筋梗塞モデルラットの作製  (1) Preparation of myocardial infarction model rat
渡邊らの報告 (サーキュレーションリサーチ、 第 69巻、 370— 377頁、 1991年) に従い雄性ウィスターラット (11週齢:体重 300— 400 g) をペントバルビタール (50mg/kg, i. p. ) で麻酔し、 人工呼吸下で正中にて開胸し た。 心嚢膜を切開後、 心臓を露出させた。 冠動脈の左前下行枝起始部で、 糸付き 縫合針 (エルプ社、 5_0シルク) にて心筋ごと冠動脈を絹糸で縛った後閉胸し た。 偽手術群は糸を縛らずに閉胸した。 麻酔から回復後、 通常飼育した。  According to the report of Watanabe et al. (Circulation Research, Vol. 69, pp. 370-377, 1991), male Wistar rats (11 weeks old, weighing 300-400 g) were anesthetized with pentobarbital (50 mg / kg, ip). The chest was opened midline under artificial respiration. After incision of the pericardium, the heart was exposed. At the root of the left anterior descending branch of the coronary artery, the coronary artery and myocardium were tied with a suture needle with a thread (ELP, 5_0 silk) and the chest was closed. The sham surgery group closed the chest without tying the thread. After recovery from anesthesia, they were usually reared.
(2) To t a l RNAの抽出  (2) Total RNA extraction
術後 1週経過、 8週経過、 20週経過、 30週経過したラットをペントバルビ タール麻酔下で開胸し、 心臓を摘出した後、 生理食塩水で大動脈より逆行性に冠 動脈を灌流して血液を洗い流した。 摘出した心臓からハサミで左心室以外の組織 を取り除いた後、 梗塞形成を確認した後に梗塞領域 (スカー形成部位) を取り除 き、 非梗塞領域のみとした。 これをハサミで細かく細断した後、 I SOGEN ( 和光純薬) を用いて To t a 1 RNAを抽出した  Rats 1 week, 8 weeks, 20 weeks, and 30 weeks after surgery were subjected to thoracotomy under pentobarbital anesthesia, the heart was removed, and the coronary artery was perfused retrograde from the aorta with saline. Blood was washed away. After removing tissues other than the left ventricle from the extracted heart with scissors, infarct formation was confirmed and then the infarct region (scar formation site) was removed, leaving only the non-infarcted region. This was cut into small pieces with scissors, and Tota1 RNA was extracted using ISOGEN (Wako Pure Chemical).
(3) 新規ラット Nkx 2. 5類似遺伝子のクローニング  (3) Cloning of a novel rat Nkx2.5-like gene
ジーンバンクに登録されていたマウス Nk X 2. 5 c DNAの塩基配列 (プ ロセッシング ·ォブ ·ナチョナル ·アカデミック ·サイエンス、 U.S.A. 第 90 卷、 第 8145 - 8149頁、 1993年、 ァクセッションナンパ一: AF091351) から P CRプライマーとして配列番号: 3および配列番号: 4で表される塩基配列から なる DNAを合成し、 マラソンレディラット心臓 cDNAライブラリー (クロン テック) を铸型として PC Rを行った。 反応は TaKaRa La Taa with GC buffer ( 宝酒造) を用いてサーマルサイクラ一 g e n e amp PGR s y s t em 9700 (パーキンエルマ一社製) にて行い、 95 で 30秒、 62°Cで 30秒 、 72でで 3分を 1サイクルとして 33サイクルを繰り返した。 得られた DNA 断片を pT 7— Tベクター (宝酒造) にクローニングした後、 さらに公知の合成 プライマー (Τ7プライマー (配列番号: 5) と U— 19プライマ一 (配列番号 : 6) ) を用い、 ΡΕアプライドバイオシステムズ社のサイクルシーケンスキッ トによって反応を行い、 蛍光 DNAシーケンサ一 (AB I PR I SM 377 , パーキンエルマ一社製) で分析して、 塩基配列を解読した結果、 配列番号: 7 で表される塩基配列からなる DNAを得た。 本配列は、 公知のマウス Nkx 2. 5 c DNA配列と高いホモロジ一を示した。 そこで 5, RACE法および 3' RACE法にて cDNA配列の全長配列の決定を行った。 5, RACE用のブラ イマ一として配列番号: 8と配列番号: 9で表される塩基配列からなる DNAを 合成し、 3, RACE用のプライマーとして配列番号: 10と配列番号: 11で 表される塩基配列からなる DNAを合成し、 マラソンレディラット心臓 cDNA ライブラリ一 (クロンテック) を鍀型として PCRを行った。 得られた遺伝子断 片の塩基配列を解読し、 本発明の新規 Nkx 2. 5類似タンパク質をコードする DNAを含有する、 配列番号: 12で表される塩基配列からなる DNAを得た。 次にマラソンレディラット心臓 cDNAライブラリー (クロンテック) を铸型と して配列番号: 12で表される塩基配列を参考にして合成した 2つのプライマー (配列番号: 13と配列番号: 14) を用いて PC Rを行い、 本発明の新規 Nk X 2. 5類似タンパク質をコードする DNAの全長を取得した。 このようにして 得られた本発明の新規 Nkx 2. 5類似タンパク質をコードする DNAを pT 7 一 Τベクタ一 (宝酒造) にクロ一ニングし、 本ベクター (プラスミド) を ρΤΒ 2165と命名した。 さらに公知の合成プライマー (Τ7プライマ一 (配列番号 : 5) と U— 19プライマー (配列番号: 6) ) を用い、 ΡΕアプライドバイオ システムズ社のサイクルシーケンスキットによって反応を行い、 蛍光 DNAシ一 ケンサ一 (AB I PR I SM 377, パーキンエルマ一社製) で分析し塩基 配列を解読した結果、 本発明の新規 Nkx 2. 5類似タンパク質をコードする D NA (配列番号: 2) を得た。 上記 pTB 2165を大腸菌に導入した形質変換 体をェシエリヒア .コリ (E s c h e r i c h i a c o l i ) DH5 α/ρ T Β 2165と命名した。 Nucleotide sequence of mouse NkX2.5c DNA registered in GeneBank (Processing, Ob, Nachonar, Academic Science, USA, Vol. 90, pp. 8145-8149, 1993, Accession Nampa) : AF091351), a DNA consisting of the nucleotide sequence represented by SEQ ID NO: 3 and SEQ ID NO: 4 was synthesized as a PCR primer, and PCR was performed using a marathon lady rat heart cDNA library (Clontech) as type II. . The reaction was performed using TaKaRa La Taa with GC buffer (Takara Shuzo) on a thermal cycler gene amp PGR syst em 9700 (manufactured by PerkinElmer), for 30 seconds at 95, 30 seconds at 62 ° C, and 3 at 72. 33 cycles were repeated with one minute as one cycle. After cloning the obtained DNA fragment into the pT7-T vector (Takara Shuzo), using known synthetic primers (Τ7 primer (SEQ ID NO: 5) and U-19 primer-1 (SEQ ID NO: 6)), The reaction was performed using the Applied Biosystems cycle sequence kit, and a fluorescent DNA sequencer (AB IPRISM 377 As a result of analyzing the nucleotide sequence, a DNA consisting of the nucleotide sequence represented by SEQ ID NO: 7 was obtained. This sequence showed a high homology with the known mouse Nkx2.5 cDNA sequence. Therefore, the full-length sequence of the cDNA sequence was determined by the 5, RACE method and the 3 'RACE method. 5. Synthesizes a DNA consisting of the nucleotide sequence represented by SEQ ID NO: 8 and SEQ ID NO: 9 as a primer for RACE, and 3, represented by SEQ ID NO: 10 and SEQ ID NO: 11 as primers for RACE A DNA consisting of the following base sequence was synthesized, and PCR was carried out using a marathon lady rat heart cDNA library 1 (Clontech) as type III. The nucleotide sequence of the obtained gene fragment was decoded to obtain a DNA comprising the nucleotide sequence represented by SEQ ID NO: 12 and containing the DNA encoding the novel Nkx2.5 analogous protein of the present invention. Next, two primers (SEQ ID NO: 13 and SEQ ID NO: 14) were synthesized using the marathon lady rat heart cDNA library (Clontech) as a type II and referring to the nucleotide sequence represented by SEQ ID NO: 12. PCR was performed to obtain the full-length DNA encoding the novel NkX2.5 analogous protein of the present invention. The thus-obtained DNA encoding the novel Nkx2.5 analogous protein of the present invention was cloned into pT71-vector (Takara Shuzo), and the vector (plasmid) was named ρ1652165. Further, using known synthetic primers (Τ7 primer (SEQ ID NO: 5) and U-19 primer (SEQ ID NO: 6)), the reaction was carried out using a cycle sequence kit of Applied Biosystems, Inc. to obtain a fluorescent DNA sequencer. (AB IPRISM 377, manufactured by PerkinElmer Inc.), and the nucleotide sequence was decoded. As a result, DNA (SEQ ID NO: 2) encoding the novel Nkx2.5 analogous protein of the present invention was obtained. The transformant in which the above-mentioned pTB2165 was introduced into Escherichia coli was named Escherichiacoli DH5α / ρTΒ2165.
次に、 明らかとなった塩基配列を基に公のデータベースである Ge η e.b 1 e データベースを用いて B 1 a s t Nによるホモロジ一検索を行ったところ、 Ratt us norvegicus tin腿 homolog(rNkx-2.5) (AF006664)と高い相同性を示したが、 本発明の新規 Nkx 2. 5類似タンパク質をコードする DNA (配列番号: 2) は新規な Nkx 2. 5類似遺伝子であることが分かった (図 1〜図 9) 。 実施例 2 Next, a homology search using B 1 ast N was performed using the public database Ge η eb 1 e database based on the revealed base sequence, and Ratt us norvegicus tin thigh homolog (rNkx-2.5) (AF006664), the DNA encoding the novel Nkx2.5-like protein of the present invention (SEQ ID NO: 2) was found to be a novel Nkx2.5-like gene (FIG. 1). ~ Figure 9). Example 2
正常ラットにおける本発明の新規 Nkx 2. 5類似タンパク質をコードする D N Aの組織分布の解析  Analysis of tissue distribution of DNA encoding the novel Nkx2.5 analogous protein of the present invention in normal rats
ノーザンブロッテイング用のプローブを得るために、 実施例 1で得られた配列 番号: 2で表される塩基配列からなる DNAを铸型とし、 配列番号: 15および 配列番号: 16で表される塩基配列からなる DNAをプライマーとして用いて実 施例 1— (3) の方法と同様の方法で PC Rを行い、 プローブ (配列番号: 17 ) を調製した。 ノ一ザンブロッテイング用膜はクロンテック社製ラット MTN Blot を用いた。 ハイブリダィゼーシヨン溶液として Express Hyb Hybridization solu tion (クロンテック) を使用して、 68°Cでプレハイブリダィゼ一シヨンを行つ た。 一方、 プローブとして上記で調製した Nkx 2. 5類似遺伝子断片を [α- 32 P]dCTPと BcaBEST Labeling Kit (宝酒造) を用いて標識した。 ハイブリダィゼー シヨンは標識プローブを含む Express Hyb Hybridization solution (クロンテツ ク) 中で 68 °C、 1時間の条件で行つた。 膜は最終的に 0. 1 X S S C , 0. 1 %SD S液中 50°Cで洗浄し、 検出には BAS— 2000 (フジフィルム) を用 いた。 その結果、 心臓が本発明の新規 Nkx 2. 5類似タンパク質をコードする DN Aの主要発現部位であることが分かった。 実施例 3 In order to obtain a probe for Northern blotting, the DNA consisting of the nucleotide sequence represented by SEQ ID NO: 2 obtained in Example 1 was designated as type III, and the nucleotides represented by SEQ ID NO: 15 and SEQ ID NO: 16 Using the DNA consisting of the sequence as a primer, PCR was performed in the same manner as in Example 1- (3) to prepare a probe (SEQ ID NO: 17). Rat MTN Blot manufactured by Clontech was used as a membrane for northern blotting. Prehybridization was performed at 68 ° C using Express Hyb Hybridization solution (Clontech) as a hybridization solution. On the other hand, the Nkx2.5-like gene fragment prepared above was labeled as a probe using [α- 32 P] dCTP and BcaBEST Labeling Kit (Takara Shuzo). Hybridization was performed in Express Hyb Hybridization solution (Clontech) containing the labeled probe at 68 ° C for 1 hour. The membrane was finally washed at 50 ° C in 0.1 XSSC, 0.1% SDS solution, and BAS-2000 (Fujifilm) was used for detection. As a result, it was found that the heart was the main expression site of DNA encoding the novel Nkx2.5 analogous protein of the present invention. Example 3
心筋梗塞モデルラッ卜での本発明の新規 Nkx 2. 5類似タンパク質をコード する D N Aの経時変化の解析  Analysis of the time course of DNA encoding the novel Nkx2.5 analogous protein of the present invention in a myocardial infarction model rat
実施例 1一 (2) で記載した心筋梗塞形成術後 1週、 8週、 20週、 30週経 過ラットの左心室の非梗塞領域由来の To t a 1 RNAと、 その対象として用 いた偽手術した後 8週経過した左心室由来の To t a 1 RN Aをそれぞれ TadM an Reverse Transcription Reagents (PEアプライドバイオシステムズ社製) を用いて cDNAを合成した。 次に TadMan Rodent G3PDH control reagent VIC ' probe (P Eアプライドバイオシステムズ社製) を用いて P CRによるグリセ口 ール 3リン酸脱水素酵素のコピー数の定量を ABI Prism 7700 seauence Detectio n Systemによって行った。 この反応は、 TadMan PCR Core Reagents kit (PEァ プライドバイオシステムズ社製) を使用し、 添付されている説明書に従って一連 の操作を行った。 その結果得られたグリセロール 3リン酸脱水素酵素のコピー数 で各 c DNAライプラリーをノーマライズした後、 RT— PCRによって本発明 の新規 Nkx2. 5類似タンパク質をコードする DNAの発現量を測定した。 次に、 本発明の新規 Nkx 2. 5類似タンパク質をコードする DNAを特異的 に増幅することができるプライマー (配列番号: 16と配列番号: 17) を合成 し、 次に示す方法で PC Rを行った。 即ち、 反応は TaKaRa La Ta with GC buff er (宝酒造) を用いて、 サ一マルサイクラ一 g e n e amp PCR s y s t em 9700 (パーキンエルマ一社製) にて行い、 95°Cで 30秒、 62°C で 30秒、 72°Cで 3分を 1サイクルとして行った。 なお、 30から 40までの 各サイクルごとにサンプリングした。 得られた本発明の新規 Nkx 2. 5類似夕 ンパク質をコードする DNAを 2 %ァガロースによる電気泳動し、 ェチジュゥム ブロマイド染色によって検出したバンドを画像解析装置 (Fluorlmager 595, Mole cular Dynamics社製) で定量した。 片対数プロット上、 プラ! ^一に達していない 直線性のあるサイクル時の測定値から発現量を算出した。 次に偽手術群の測定値 を 1とした時の経時的な発現量を図 10に示した。 図中、 縦軸は、 左心室におけ る本発明の新規 Nkx2.5類似タンパク質をコードする DNAのコピー数 (発現量) をハウスキーピング遺伝子であるグリセロール 3リン酸脱水素酵素遺伝子のコピ 一数で割ることにより補正した後、 偽手術群の測定値で割ることによって得た数 値を示し、 これをフォールドインクリースとして表示した。 横軸の時間 (週) は 用いた心不全モデルのサンプルごとの経過を示した。 sham 8wは偽手術群、 MI lw は手術後 1週経過、 MI 8wは手術後 8週経過、 MI 20wは手術後 20週経過、 Ml 30 wは手術後 30週経過した心臓を分析した際のサンプル名を示す。 Example 1 Tota1 RNA derived from the non-infarcted region of the left ventricle of the rat after 1 week, 8 weeks, 20 weeks and 30 weeks after myocardial infarction described in (2) Eight weeks after the operation, cDNA was synthesized from the left ventricle-derived Tota 1 RNA using TadM an Reverse Transcription Reagents (manufactured by PE Applied Biosystems). Next, using a TadMan Rodent G3PDH control reagent VIC 'probe (manufactured by PE Applied Biosystems), the copy number of glycerol triphosphate dehydrogenase was determined by PCR using ABI Prism 7700 seauence Detectio. Performed by n System. In this reaction, a series of operations were performed using TadMan PCR Core Reagents kit (manufactured by PE Applied Biosystems) according to the attached instructions. After normalizing each cDNA library with the copy number of glycerol triphosphate dehydrogenase obtained as a result, the expression level of the DNA encoding the novel Nkx2.5 analogous protein of the present invention was measured by RT-PCR. Next, primers (SEQ ID NO: 16 and SEQ ID NO: 17) capable of specifically amplifying the DNA encoding the novel Nkx2.5 analogous protein of the present invention were synthesized, and PCR was performed by the following method. went. That is, the reaction was performed using TaKaRa La Ta with GC buffer (Takara Shuzo) in a samurai cycler gene amp PCR syst em 9700 (manufactured by PerkinElmer) at 95 ° C for 30 seconds and at 62 ° C. The cycle was performed at 72 ° C for 30 seconds and 3 minutes as one cycle. In addition, sampling was performed for each cycle from 30 to 40. The obtained DNA encoding the novel Nkx2.5 analogous protein of the present invention was subjected to electrophoresis with 2% agarose, and the band detected by ethidium bromide staining was analyzed with an image analyzer (Fluorlmager 595, manufactured by Molecular Dynamics). Quantified. On a semi-log plot, plastic! ^ The expression level was calculated from the measured value during a cycle that did not reach one. Next, the amount of expression over time when the measured value of the sham operation group was set to 1 is shown in FIG. In the figure, the vertical axis indicates the copy number (expression level) of the DNA encoding the novel Nkx2.5-like protein of the present invention in the left ventricle, which is the number of copies of the glycerol triphosphate dehydrogenase gene, which is a housekeeping gene. After correcting by dividing by, the numerical value obtained by dividing by the measurement value of the sham operation group was shown, and this was indicated as fold increase. The time (weeks) on the horizontal axis shows the progress of each sample of the heart failure model used. sham 8w for sham operation group, MI lw for 1 week after surgery, MI 8w for 8 weeks after surgery, MI 20w for 20 weeks after surgery, and Ml 30 w for heart after 30 weeks after surgery Indicates the sample name.
これより、 本発明の新規 Nkx 2. 5類似タンパク質をコードする DNAは術 後 1週で顕著に低下 (0. 29倍) した後、 術後 8週で増加傾向 (1. 135倍 ) を示し、 術後 20週で増加 (2倍) 、 30週で偽手術群レベルに戻ることが明 らかとなつた。  Thus, the DNA encoding the novel Nkx2.5 analogous protein of the present invention showed a marked decrease (0.29-fold) at 1 week after surgery and an increasing tendency (1.135-fold) at 8 weeks after surgery. However, it increased at 20 weeks after the operation (2 times) and returned to the sham operation group level at 30 weeks.
手術直後から 1週経過時は、 梗塞が形成されつつある時期と考えられ、 結窄さ れた冠動脈から下流領域の心筋細胞が急速に死滅、 脱落し、 リンパ球の浸潤によ り炎症が生じていると推測される。 また術後 2 0週から 3 0週は死亡例が見られ る直前であることから術後 8週は代償機序が作動している時期であり、 術後 2 0 週以降は十分な代償機序が作動していないか、 あるいは過剰な代償機序により代 償破綻が生じている時期であると考えられる。 そこで術後 1週経過時を急性期 ( 心不全急性期) 、 術後 8週経過時を慢性期 (心不全代償期) 、 術後 2 0週以降を 末期 (心不全非代償期) と考えた。 心筋梗塞から心不全への移行に関わる代償機 序は、 次のように考えられる。 心筋細胞が脱落 (壊死あるいはアポトーシス) す ると失った心筋細胞の有していた機能を心臓全体で代償するために残存心筋細胞 は肥大し、 心拡張や線維化を伴う心臓の再構築 (心リモデリング) が生じる。 こ れによって機能的に心機能は代償されることになるが、 一方でこの心リモデリン グあるいは過剰な代償機序そのものが心不全を発症する危険性をはらんでいると 考えられている (内科、 第 7 9巻、 2— 2 0頁、 1997年) 。 しかしながら代償破 綻そのものに関与する分子は未だ同定されておらず、 従ってそのメカニズムも明 らかにされていない。 上記実施例で示したとおり、 上記ラット心筋梗塞モデルを 用いて、 心臓特異的遺伝子群の正の転写調節因子である N k X 2 . 5と相同性の 高い遺伝子である本発明の新規 N k x 2 . 5類似タンパク質をコードする D NA の発現プロファイルを作成した。 本遺伝子は急性期に顕著に低下し、 慢性期から 末期で増加傾向を示した。 上記実施例で明らかになった本発明の新規 N k x 2 . 5類似夕ンパク質をコードする D N Aの詳細な発現プロフアイルは次のように考 察することができる。 N k x 2 . 5は、 心臓に対して保護的に働くと考えられて いる心房性ナトリゥム利尿べプチドと心室性ナトリゥム利尿べプチドの発現を誘 導すること、 心収縮タンパク質の一つであるミオシンライトチェーン 2 Vの発現 を正に調節することが報告されている。 従って N k x 2 . 5類似遺伝子が同様の 機能を有している可能性があり、 急性期における発現低下は病態悪化に直接結び つくものと考えられる。 一方で慢性期から末期にかけてその発現増強は、 心臓特 異的な負の調節因子と考えられている力一ディアツク ·アドリアマイシン -レス ボンシブ 'プロテイン (CARP) (J. B 、 第 272巻、 22800- 22808頁、 1997年、 デ ベロプメント、 第 124巻、 793- 804頁、 1997年、 デベロプメント、 第 126巻、 4223- 4234頁、 1999年)の発現を増強することになり、 この期の発現増強は病態悪化に 直接結びつくものと考えられる。 心疾患予防'治療薬としては、 急性期に速やか に代償機序を作動させることと代償破綻を抑制することが重要である。 本発明の 新規 N k x 2 . 5類似タンパク質をコードする D N Aの発現を適切にコント口一 ルすることは、 この課題を解決する上で重要である。 発現低下は代償機序を抑制 し、 発現増加は代償破綻を加速すると考えられる。 従って本発明の新規 N k x 2 . 5類似タンパク質をコードする D NAの発現や遺伝子産物の機能を調節する薬 剤は心疾患の新たな予防 ·治療剤として有用である。 産業上の利用可能性 One week after the operation is considered to be the time when infarction is being formed. It is presumed that myocardial cells in the downstream region from the coronary artery died and died rapidly, and inflammation was caused by lymphocyte infiltration. Since 20 to 30 weeks after the operation is immediately before death is observed, 8 weeks after the operation is the time when the compensation mechanism is operating, and after 20 weeks after the operation, sufficient compensation is available. It is likely that the order is not working, or that there is a compensatory failure due to excessive compensation mechanisms. Therefore, one week after the operation was considered the acute phase (acute heart failure), eight weeks after the surgery was considered the chronic phase (heart failure decompensation), and 20 weeks after the surgery was considered the end stage (heart failure decompensation). The compensatory mechanism involved in the transition from myocardial infarction to heart failure is considered as follows. When the myocardial cells fall off (necrosis or apoptosis), the remaining myocardial cells become enlarged to compensate for the functions of the lost myocardial cells throughout the heart, and the heart is remodeled with diastolic and fibrotic changes (cardiac). Remodeling) occurs. This functionally compensates for cardiac function, but it is thought that this cardiac remodeling or excessive compensatory mechanism itself carries the risk of developing heart failure. Vol. 79, pp. 2-20, 1997). However, the molecules involved in compensatory failure itself have not yet been identified, and the mechanism has not been elucidated. As shown in the above Examples, using the rat myocardial infarction model described above, the novel N kx of the present invention, which is a gene highly homologous to N kX 2.5 which is a positive transcription regulator of the heart-specific gene group, is used. An expression profile for DNA encoding the 2.5 analogous protein was generated. This gene decreased remarkably in the acute phase, and increased from the chronic phase to the end phase. The detailed expression profile of the DNA encoding the novel N kx 2.5 analogous protein of the present invention, which has been clarified in the above example, can be considered as follows. Nkx2.5 induces the expression of atrial sodium diuretic peptide and ventricular sodium diuretic peptide, which are thought to protect the heart.Myosin, one of the cardiac contractile proteins It has been reported to positively regulate light chain 2V expression. Therefore, there is a possibility that the Nkx2.5-like gene has a similar function, and it is considered that the decreased expression in the acute phase is directly linked to the worsening of the disease state. On the other hand, from the chronic stage to the end stage, the expression is enhanced by the ability of diatsk-adriamycin-resbonsib 'protein (CARP), which is considered to be a cardiac-specific negative regulator (J.B, 272, 22800- 22808, 1997, Development, 124, 793-804, 1997, Development, 126, 4223- 4234, 1999), which is thought to directly lead to worsening of the disease state. As a therapeutic agent for heart disease prevention, it is important to actuate the compensatory mechanism promptly in the acute phase and to suppress compensatory failure. Appropriate control of the expression of DNA encoding the novel Nkx2.5 analogous protein of the present invention is important for solving this problem. Decreased expression suppresses the compensatory mechanism, and increased expression is thought to accelerate compensation failure. Therefore, the drug of the present invention that regulates the expression of DNA encoding the novel Nkx2.5-like protein and the function of the gene product is useful as a new agent for preventing and treating heart disease. Industrial applicability
配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のアミノ酸 配列を有するタンパク質またはその塩は新規であり、 該タンパク質またはその塩 の活性を調節する化合物またはその塩、 および該タンパク質またはその塩の活性 を調節する抗体は、 例えば、 心疾患などの予防 ·治療剤として使用することがで きる。 該タンパク質またはその塩をコードする D NAに相補もしくは実質的に相 補な塩基配列を有するアンチセンスヌクレオチドは、 該タンパク質またはその塩 の発現を抑制することができ、 例えば、 心疾患などの予防 ·治療剤として使用す ることができる。  The protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a salt thereof is novel, and a compound or a salt thereof which regulates the activity of the protein or the salt thereof; An antibody that regulates the activity of the salt can be used, for example, as a prophylactic or therapeutic agent for heart disease and the like. An antisense nucleotide having a nucleotide sequence complementary or substantially complementary to DNA encoding the protein or a salt thereof can suppress the expression of the protein or a salt thereof. It can be used as a therapeutic agent.

Claims

請求の範囲 The scope of the claims
1 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のアミ ノ酸配列を含有するタンパク質またはその塩。  1. A protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1, or a salt thereof.
2 . 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またはその塩 。  2. A protein containing an amino acid sequence represented by SEQ ID NO: 1 or a salt thereof.
3 . 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。  3. The partial protein of claim 1 or a salt thereof.
4. 請求項 1記載のタンパク質または請求項 3記載の部分べプチドをコードす るポリヌクレオチドを含有するポリヌクレオチド。  4. A polynucleotide containing the polynucleotide encoding the protein according to claim 1 or the partial peptide according to claim 3.
5 . D N Aである請求項 4記載のポリヌクレオチド。  5. The polynucleotide according to claim 4, which is DNA.
6 . 配列番号: 2で表される塩基配列を含有する請求項 5記載の D NA。  6. The DNA according to claim 5, which comprises the nucleotide sequence represented by SEQ ID NO: 2.
7 . 請求項 4記載のポリヌクレオチドを含有する組換えベクター。  7. A recombinant vector containing the polynucleotide according to claim 4.
8 . 請求項 7記載の組換えべクタ一で形質転換された形質転換体。  8. A transformant transformed by the recombinant vector according to claim 7.
9 . 請求項 8記載の形質転換体を培養し、 請求項 1記載のタンパク質もしくは その塩または請求項 3記載の部分ペプチドまたはその塩を生成、 蓄積せしめ、 こ れを採取することを特徴とする請求項 1記載のタンパク質もしくはその塩または 請求項 3記載の部分べプチドもしくはその塩の製造方法。  9. The method according to claim 8, wherein the transformant according to claim 8 is cultured to produce and accumulate the protein according to claim 1 or a salt thereof or the partial peptide according to claim 3 or a salt thereof, and collect the same. A method for producing the protein of claim 1 or a salt thereof or the partial peptide of claim 3 or a salt thereof.
1 0 . 請求項 1記載のタンパク質もしくはその塩または請求項 3記載の部分べ プチドもしくはその塩を含有してなる医薬。  10. A medicament comprising the protein of claim 1 or a salt thereof or the partial peptide of claim 3 or a salt thereof.
1 1 . 請求項 1記載のタンパク質もしくはその塩または請求項 3記載の部分べ プチドもしくはその塩に対する抗体。  11. An antibody against the protein according to claim 1 or a salt thereof, or the partial peptide according to claim 3 or a salt thereof.
1 2 . 請求項 1 1記載の抗体を含有する診断薬。  12. A diagnostic agent comprising the antibody according to claim 11.
1 3 . 請求項 1記載のタンパク質もしくはその塩または請求項 3記載の部分べ プチドもしくはその塩を用いるごとを特徴とする、 請求項 1記載のタンパク質も しくはその塩または請求項 3記載の部分ぺプチドもしくはその塩の活性を調節す る化合物またはその塩のスクリーニング方法。  13. The protein according to claim 1, a salt thereof, or the partial peptide according to claim 3 or a salt thereof, wherein the protein according to claim 1, a salt thereof, or the portion according to claim 3 is used. A method for screening a compound or a salt thereof that regulates the activity of the peptide or a salt thereof.
1 4. 請求項 1記載のタンパク質もしくはその塩または請求項 3記載の部分べ プチドもしくはその塩が、 請求項 1記載のタンパク質または請求項 3記載の部分 ペプチドをコードする D N Aを含有する D N Aで形質転換された形質転換体の細 胞質内に発現されたものである請求項 1 3記載のスクリーニング方法。 1 4. A protein or a salt thereof according to claim 1 or a partial peptide or a salt thereof according to claim 3 is characterized by a DNA containing a DNA encoding the protein according to claim 1 or the partial peptide according to claim 3. 14. The screening method according to claim 13, which is expressed in the cells of the transformed transformant.
1 5 . 請求項 1記載のタンパク質もしくはその塩または請求項 3記載の部分べ プチドもしくはその塩を含有することを特徴とする、 請求項 1記載のタンパク質 もしくはその塩または請求項 3記載の部分べプチドもしくはその塩の活性を調節 する化合物またはその塩のスクリーニング用キット。 15. The protein according to claim 1, or a salt thereof, or the partial peptide according to claim 3, or a salt thereof, wherein the protein according to claim 1, or a salt thereof, or the salt according to claim 3, is contained. A kit for screening a compound or a salt thereof that regulates the activity of a peptide or a salt thereof.
1 6 . 請求項 1 3記載のスクリーニング方法または請求項 1 5記載のスクリ一 ニング用キットを用いて得られる、 請求項 1記載のタンパク質もしくはその塩ま たは請求項 3記載の部分べプチドもしくはその塩の活性を調節する化合物または その塩。  16. The protein according to claim 1, or a salt thereof, or the partial peptide or the partial peptide according to claim 3, obtained by using the screening method according to claim 13 or the screening kit according to claim 15. A compound or a salt thereof that regulates the activity of the salt.
1 7 . 請求項 1 6記載の化合物またはその塩を含有してなる医薬。  17. A medicament comprising the compound according to claim 16 or a salt thereof.
1 8 . 心疾患の予防 ·治療剤である請求項 1 7記載の医薬。  18. The medicament according to claim 17, which is an agent for preventing or treating heart disease.
1 9 . 請求項 1記載のタンパク質または請求項 3記載の部分ペプチドをコード する D NAに相補的もしくは実質的に相補的な塩基配列を有するアンチセンスヌ クレオチド。  19. An antisense nucleotide having a nucleotide sequence complementary or substantially complementary to DNA encoding the protein according to claim 1 or the partial peptide according to claim 3.
2 0 . 請求項 1 9記載のアンチセンスヌクレオチドを含有してなる医薬。  20. A medicament comprising the antisense nucleotide according to claim 19.
2 1 . 請求項 4記載のポリヌクレオチドを含有してなる医薬。 21. A pharmaceutical comprising the polynucleotide according to claim 4.
2 2 . 請求項 4記載のポリヌクレオチドを含有してなる診断薬。  22. A diagnostic agent comprising the polynucleotide according to claim 4.
2 3 . 哺乳動物に対して、 請求項 1 6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心疾患の予防 ·治療方法。  23. A method for preventing and treating heart disease, comprising administering an effective amount of the compound according to claim 16 or a salt thereof to a mammal.
2 4. 心疾患の予防 ·治療剤を製造するための請求項 1 6記載の化合物または その塩の使用。  2 4. Use of the compound according to claim 16 or a salt thereof for producing an agent for preventing or treating heart disease.
PCT/JP2001/009477 2000-10-30 2001-10-29 Novel disease-associated gene and use thereof WO2002036772A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2002210960A AU2002210960A1 (en) 2000-10-30 2001-10-29 Novel disease-associated gene and use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2000-331362 2000-10-30
JP2000331362 2000-10-30

Publications (1)

Publication Number Publication Date
WO2002036772A1 true WO2002036772A1 (en) 2002-05-10

Family

ID=18807716

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2001/009477 WO2002036772A1 (en) 2000-10-30 2001-10-29 Novel disease-associated gene and use thereof

Country Status (2)

Country Link
AU (1) AU2002210960A1 (en)
WO (1) WO2002036772A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1379691A4 (en) * 2001-02-23 2005-12-14 Univ Johns Hopkins Med Differentially methylated sequences in pancreatic cancer

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001048150A1 (en) * 1999-12-28 2001-07-05 Kyowa Hakko Kogyo Co., Ltd. Cells capable of differentiating into heart muscle cells
WO2001048151A1 (en) * 1999-12-28 2001-07-05 Kyowa Hakko Kogyo Co., Ltd. Cells capable of differentiating into heart muscle cells
WO2001048149A1 (en) * 1999-12-28 2001-07-05 Kyowa Hakko Kogyo Co., Ltd. Adult bone marrow-origin cell capable of differentiating into heart muscle cell

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001048150A1 (en) * 1999-12-28 2001-07-05 Kyowa Hakko Kogyo Co., Ltd. Cells capable of differentiating into heart muscle cells
WO2001048151A1 (en) * 1999-12-28 2001-07-05 Kyowa Hakko Kogyo Co., Ltd. Cells capable of differentiating into heart muscle cells
WO2001048149A1 (en) * 1999-12-28 2001-07-05 Kyowa Hakko Kogyo Co., Ltd. Adult bone marrow-origin cell capable of differentiating into heart muscle cell

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
KASAHARA H. ET AL.: "Cardiac and extracardiac expression of Csx/Nkx2.5 homeodomain protein", CIRC. RES., vol. 82, no. 9, 18 May 1998 (1998-05-18), pages 936 - 946, XP002907877 *
KOMURO I. AND IZUMO S.: "Csx: a murine homeobox-containing gene specifically expressed in the developing heart", PROC. NATL. ACAD. SCI. USA, vol. 90, no. 17, 1 September 1993 (1993-09-01), pages 8145 - 8149, XP002907875 *
LINTS T.J. ET AL.: "Nkx-2.5: a novel murine homeobox gene expressed in early heart progenitor cells and their myogenic descendants", DEVELOPMENT, vol. 119, no. 3, November 1993 (1993-11-01), pages 969, XP002907876 *
TURBAY D. ET AL.: "Molecular cloning, chromosomal mapping and characterization of the human carciac-specific homeobox gene hCsx", MOL. MED., vol. 2, no. 1, January 1996 (1996-01-01), pages 86 - 96, XP002943590 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1379691A4 (en) * 2001-02-23 2005-12-14 Univ Johns Hopkins Med Differentially methylated sequences in pancreatic cancer

Also Published As

Publication number Publication date
AU2002210960A1 (en) 2002-05-15

Similar Documents

Publication Publication Date Title
JP2002112772A (en) New polypeptide and its dna
WO2002053738A1 (en) Novel proteins and dnas thereof
US7235531B2 (en) Tachykinin-like polypeptides and use thereof
US7256279B2 (en) Protein having ribonucleotide reductase activity and DNA thereof
WO2002036772A1 (en) Novel disease-associated gene and use thereof
WO2001048203A1 (en) Novel protein and dna thereof
WO2004028558A1 (en) Preventives/remedies for neurodegenerative disease
WO2002036763A1 (en) Novel gene overexpressed in heart and skeletal muscle and use tehreof
JP2001029090A (en) Novel polypeptide
JP2001228146A (en) Use of disease-associated gene
JP4300008B2 (en) Novel protein and its DNA
WO2003097686A1 (en) Novel protein, its dna and use thereof
JPH11266870A (en) New protein and its dna
JP2001299363A (en) New polypeptide and dna encoding the same
JP2002348299A (en) Irap binding protein
JP2004026692A (en) New use of mepe
JP2001299364A (en) New polypeptide and dna encoding the same
WO2002016428A1 (en) Irap-binding protein
JPH10108683A (en) New protein and its dna
JP2002355064A (en) New disease-associated gene and its use
JP2000053700A (en) New protein and its dna
WO2002033082A1 (en) Novel disease-associated gene and use thereof
JP2003000272A (en) New protein and its dna
JP2002355063A (en) New disease-associated gene and its use
JP2001211890A (en) New transcription factor and its dna

Legal Events

Date Code Title Description
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
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP