WO2003085106A1 - Preventives and/or remedies for cancer - Google Patents

Preventives and/or remedies for cancer Download PDF

Info

Publication number
WO2003085106A1
WO2003085106A1 PCT/JP2003/004272 JP0304272W WO03085106A1 WO 2003085106 A1 WO2003085106 A1 WO 2003085106A1 JP 0304272 W JP0304272 W JP 0304272W WO 03085106 A1 WO03085106 A1 WO 03085106A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
dna
nucleotide sequence
receptor protein
present
Prior art date
Application number
PCT/JP2003/004272
Other languages
French (fr)
Japanese (ja)
Inventor
Yuichi Hikichi
Koji Yoshimura
Isao Kaieda
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 AU2003236358A priority Critical patent/AU2003236358A1/en
Publication of WO2003085106A1 publication Critical patent/WO2003085106A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/711Natural deoxyribonucleic acids, i.e. containing only 2'-deoxyriboses attached to adenine, guanine, cytosine or thymine and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to antisense oligonucleotides useful for cancer prevention and treatment, screening for cancer prevention and treatment agents, and the like.
  • Antisense oligonucleotides when introduced into cells, hybridize with RNA having a complementary sequence and induce RNA degradation by RNaseH to inhibit protein translation, or hybridize to inhibit direct protein synthesis. Also bring. Since it is possible to specifically suppress the function of the target gene, it is frequently used as a means for analyzing the function of genes, and some antisense oligonucleotides are being developed for clinical application.
  • Protein tyrosine kinase genes form the largest family of oncogenes and are deeply involved in cell proliferation and differentiation and morphogenesis (Harvey Lecto. 94, 81-119, 1998- 1999), it has been picked up as a good target gene for developing botanicals.
  • drug development targeting the EGF receptor, HER2 receptor or VEGF receptor which is classified as receptor tyrosine kinase (RTK)
  • RTK receptor tyrosine kinase
  • RTK receptor tyrosine kinases
  • RTK is considered to be one of the subfamilies with a high probability of success as pharmaceuticals.
  • Eph erythropoietin producing human hepatocellular carcinoma cell line
  • the Ephr in gene family identified as ligands for the Eph receptor, has also been reported to consist of at least eight members.
  • extremely localized expression in embryos and the brain suggests their involvement in morphogenesis during development and proper projection of nerve axons ( Annu. Rev. Neurosci. 21, 309-345 (1998)), but its role in cancer has also attracted attention in recent years. For example, increased expression of Eph receptor in various cancers has been reported
  • the present inventors have conducted intensive studies to solve the above-mentioned problems, and as a result, found that cancer cells cause apoptosis by suppressing the expression of NEPHA gene expressed in cancer cells. Further studies based on this finding have led to the completion of the present invention.
  • an oligonucleotide having a nucleotide sequence identical or substantially identical to the nucleotide sequence represented by SEQ ID NO: 13 or SEQ ID NO: 14,
  • the medicament according to the above (6) which is an agent for preventing and / or treating cancer.
  • a medicament comprising the compound according to (17) or a salt thereof, (18a) a medicament comprising the compound according to (17a) or a salt thereof, (19) the medicament according to the above (18), which is an agent for preventing and / or treating cancer; (19a) the medicament according to the above (18a), which is an agent for preventing and / or treating cancer;
  • a method for preventing and treating cancer which comprises administering to an animal an effective amount of the oligonucleotide according to (1).
  • Oligonucleotide containing a base sequence substantially identical to the base sequence represented by SEQ ID NO: 13 or SEQ ID NO: 14 (hereinafter, abbreviated as oligonucleotide of the present invention or antisense oligonucleotide of the present invention) Is an Eph receptor protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 11 (hereinafter, the receptor protein of the present invention or the Eph receptor protein of the present invention) (May be abbreviated as) and can inhibit the synthesis or function of the RNA, or through the interaction with the receptor protein-related RNA of the present invention.
  • the expression of the receptor protein gene can be regulated and / or controlled.
  • Oligonucleotides containing a base sequence substantially identical to the base sequence represented by SEQ ID NO: 13 or SEQ ID NO: 14 include nucleotides represented by SEQ ID NO: 13 or SEQ ID NO: 14. Any oligonucleotide having a nucleotide sequence that hybridizes under stringent conditions and having substantially the same activity as the oligonucleotide having the nucleotide sequence represented by SEQ ID NO: 13 or SEQ ID NO: 14 It may be something.
  • substantially equivalent activities include, for example, the activity of inhibiting the expression of the receptor protein of the present invention.
  • the expression inhibiting activity include transcription inhibition, splicing inhibition of precursor RNA, inhibition of mRNA translocation to the cytoplasm, and inhibition of translation into protein.
  • activities such as the above-mentioned expression inhibitory activity are equivalent (eg, about 0.01 to 100 times, preferably about (0.5 to 20 times, more preferably about 0.5 to 2 times).
  • the measurement of the above-mentioned inhibitory activity and the like can be performed according to a known method.
  • the inhibitory activity is determined by using a transformant containing the receptor protein of the present invention, an in vivo or in vitro gene expression system of the receptor protein of the present invention, or an in vivo or in vitro translation system of the receptor protein of the present invention.
  • DNA that can hybridize with the nucleotide sequence represented by SEQ ID NO: 13 or SEQ ID NO: 14 under high stringent conditions include, for example, SEQ ID NO: 13 or SEQ ID NO: 14 A base having about 60% or more, more preferably about 70% or more, more preferably about 80% or more, particularly preferably about 90% or more, and most preferably about 95% or more homology with the base sequence. DNA containing a sequence is used. .
  • Hybridization can be performed by a method known per se or a method analogous thereto, for example, a method described in Molecular Cloning 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). Etc.
  • the procedure can be performed according to the method described in the attached instruction manual. More preferably, it can be carried out under high stringency conditions.
  • High stringency conditions include, for example, a sodium concentration of about 19 to 40 mM, preferably about 19 to 20 mM, and a temperature of about 50 to 70, preferably about 60 to 60. The condition of 5 t is shown. Particularly, the case where the sodium concentration is about 19 mM and the temperature is about 65 is most preferable.
  • Oligonucleotides are polydeoxynucleotides containing 2-dexoxy-D-reports, polydeoxynucleotides containing D-reports, N-glycosides of purine or pyrimidine bases.
  • Other types of oligonucleotides or other polymers with non-nucleotide backbones eg, commercially available protein nucleic acids and synthetic sequence-specific nucleic acid polymers
  • specialty Other polymers containing a suitable bond these polymers include base pairing as found in DNA and RNA, and contain a nucleotide having a configuration permitting base attachment).
  • RNA hybrids can be double-stranded DNA, single-stranded DNA, double-stranded RNA, single-stranded RNA, and even DNA: RNA hybrids, and can also be unmodified oligonucleotides (or unmodified oligonucleotides). Nucleotides), as well as those with known modifications, such as labeled, capped, methylated, one or more natural nucleotides, as known in the art.
  • Is substituted with an analog, modified with an intramolecular nucleotide for example, has an uncharged bond (eg, methylphosphonate, phosphotriester, phosphoramidite, thiolbamate, etc.), a charged bond or sulfur
  • bonds for example, phosphorothioate, phosphorodithioate, etc.
  • proteins for example, proteins (nucleases, nucleases / inhibitors) , Toxins, antibodies, signal peptides, poly-L-lysine, etc.) or sugars (for example, monosaccharides), etc., or those having an intermolecular compound (for example, acridine, psoralen, etc.)
  • Compounds containing chelating compounds eg, metals, radioactive metals, boron, oxidizable metals, etc.
  • alkylating agents those with modified bonds
  • eg, ⁇ -anomers Type nucleic acid
  • nucleoside may include not only those containing purine and pyrimidine bases but also those having other modified heterocyclic bases. Such modifications may include methylated purines and pyrimidines, acylated purines and pyrimidines, or other heterocycles. Modified nucleotides and modified nucleotides may also be modified at the sugar moiety, e.g., where one or more hydroxyl groups have been replaced with halogens, aliphatic groups, etc., or functional groups such as ethers, amines, etc. It may be converted to a base.
  • the oligonucleotide of the present invention may be a modified nucleic acid (RNA, DNA).
  • modified nucleic acid include a sulfur derivative, a thiophosphate derivative, and a nucleic acid that is resistant to degradation of polynucleoside amide or oligonucleoside amide.
  • oligonucleotide of the present invention a base represented by SEQ ID NO: 13 DNA containing a sequence, DNA containing the base sequence represented by SEQ ID NO: 14 and the like are used.
  • the oligonucleotide of the present invention is produced according to a known method, for example, a solid phase or liquid phase synthesis method (eg, an automated nucleic acid synthesizer, liquid phase technology, etc.). Alternatively, it may be produced according to known gene engineering techniques (eg, use of reverse transcriptase, PCR synthesis, etc.).
  • amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 11 include, for example, about 55% or more, and preferably about 60% or more of the amino acid sequence represented by SEQ ID NO: 11, More preferably, an amino acid sequence having a homology of about 70% or more, more preferably about 80% or more, particularly preferably about 90% or more, and most preferably about 95% or more. .
  • Examples of a protein having an amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 11 include, for example, a protein having an amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 11 A protein having substantially the same activity as the amino acid sequence represented by SEQ ID NO: 11 is preferred.
  • Substantially the same activity includes, for example, ligand binding activity, signal transduction activity, cell death inhibitory activity, cell adhesion activity, and cell motility regulating activity. Substantially the same indicates that their activities are the same in nature. Therefore, activities such as ligand binding activity, signal transduction activity, cell death inhibitory activity, cell adhesion activity and cell motility control activity are equivalent (eg, about 0.01 to 100 times, preferably Is preferably about 0.5 to 20 times, more preferably about 0.5 to 2 times), but the quantitative factors such as the degree of activity and the molecular weight of the protein may be different.
  • the amino acid sequence of the receptor protein is the N-terminus (amino terminus) at the left end and the C-terminus (aminopropyl end) at the right end, according to the convention of peptide notation.
  • Receptor protein of the present invention, C-terminal, the force Rupokishiru group (-C00H), Karupokishire bets (-C00-), amide (- C0NH 2) or ester (- C00R) may be either.
  • R in the ester e.g., methyl, Echiru, n- propyl, C 1H5 alkyl group such as isopropyl or n- butyl, cyclopentyl Le, C 3 _ 8 cycloalkyl group such as cyclohexyl, for example, phenyl, (3 6 _ 12 Ariru groups, such as single-naphthyl shed, for example, benzyl, phenylene route C DOO 2 alkyl or ⁇ - naphthylmethyl etc.
  • ⁇ - naphthyl such as phenethyl - C, such as _ 2 alkyl groups in addition to C 7 _ 14 Ararukiru group, Viva port Iruo Kishimechiru group commonly used as an oral ester.
  • the receptor protein of the present invention When the receptor protein of the present invention has a lipoxyl group (or carboxylate) other than the C-terminus, the receptor protein of the present invention also includes those in which the lipoxyl group is amidated or esterified. .
  • the ester in this case, for example, the above-mentioned C-terminal ester and the like are used.
  • the receptor protein of the present invention is the protein mentioned above, Amino group protecting groups methylate Onin residues of N-terminal (e.g., formyl group, C 2 such Asechiru - such Ashiru group such as 6 Al Kanoiru group ), The N-terminal side is cleaved in vivo, and the daryumil group formed is pyroglutamine-oxidized, the substituent on the side chain of the amino acid in the molecule (eg, -0H, -SH, amino group, imidazole group, indole group, Guanijino group, etc.) a suitable protecting group (e.g., formyl group, C 2 such Asechiru - those protected by 6 etc. Ashiru groups such Arukanoiru group), a sugar chain Complex proteins such as bound so-called glycoproteins are also included.
  • Amino group protecting groups methylate Onin residues of N-terminal e.g., formyl group, C 2 such Asechir
  • receptor protein of the present invention include, for example, a receptor protein containing the amino acid sequence represented by SEQ ID NO: 11 and the like.
  • a partial peptide of the receptor protein of the present invention (hereinafter, sometimes abbreviated as the partial peptide of the present invention) can also be used.
  • the partial peptide of the present invention may be any peptide as long as it is a partial peptide of the above-described receptor protein of the present invention.
  • the receptor protein molecules of the present invention For example, a portion having a substantially identical activity or a portion located inside the cell membrane is used.
  • substantially the same activity refers to, for example, ligand binding activity, signal information transmission activity, cell death inhibitory activity, cell adhesion activity, and cell motility control activity.
  • Ligand binding activity, signal transduction activity, cell death suppressing activity, measurements, such as cell adhesion activity and cell motility control activity, the amino acid number of the partial base peptide of the c the present invention can be carried out according to known methods, Peptides having an amino acid sequence of at least 5 or more, preferably 10 or more, preferably 50 or more, more preferably 100 or more of the amino acid sequences of the above-described receptor protein of the present invention are preferable. .
  • a substantially identical amino acid sequence is at least about 50%, preferably at least about 60%, more preferably at least about 70%, still more preferably at least about 80%, and most preferably at least about 50% of these amino acid sequences.
  • An amino acid sequence having about 90% or more, most preferably about 95% or more homology is shown.
  • the partial peptide of the present invention comprises: (i) one or more (preferably about 1 to 10, more preferably several (1 to 5)) amino acids in the above amino acid sequence; (Ii) one or two or more (preferably about 1 to 20, more preferably about 1 to 10, and more preferably several (1 to 5)) (Iii) one or more (preferably about 1 to 10, more preferably several, more preferably about 1 to 5) amino acids in the above amino acid sequence It may be substituted with an amino acid.
  • Specific examples of the partial peptide of the present invention include the 60th to 73rd or 491st to 504th amino acid sequences of the amino acid sequence represented by SEQ ID NO: 11. Examples of such peptides include:
  • the C-terminus carboxyl group of the present invention (- C00H), Karupoki Shireto (- C00-), amide (- C0NH 2) or esters may be a (-C00R) (R is as defined above Is shown).
  • R is as defined above Is shown.
  • the partial peptide of the present invention has a carboxyl group (or carboxylate) other than the C-terminus, those in which the carbonyl group is amidated or esterified are also included in the partial peptide of the present invention.
  • the ester in this case, for example, the above-mentioned C-terminal ester and the like are used.
  • the partial peptide of the present invention has a N-terminal methionine residue in which the amino group of the methionine residue is protected with a protecting group, and the N-terminal side is cleaved in vivo to produce Dartamyl group is pyroglutamine-oxidized, amino acid side chain in the molecule is protected by a suitable protecting group, or sugar chain Conjugated peptides, such as so-called glycopeptides, to which is bound.
  • Examples of the salt of the receptor protein or its partial peptide of the present invention include a physiologically acceptable salt with an acid or a base, 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) or organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, succinic acid) Acids, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid) and the like are used.
  • inorganic acids eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid
  • organic acids eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, succinic acid
  • the receptor protein of the present invention or a salt thereof can be produced from the above-described human or mammalian cells or tissues by a known method for purifying a receptor protein, or encodes the receptor protein of the present invention described later. It can also be produced by culturing a transformant containing DNA. Also, the protein can be produced by the protein synthesis method described later or according to this method.
  • the human or mammalian tissues or cells are homogenized and then extracted with an acid or the like, and the resulting extract is subjected to reverse phase chromatography, ion exchange chromatography, etc. Purification and isolation can be carried out by combining the above chromatography methods.
  • a commercially available resin for protein synthesis can be usually used.
  • resins include chloromethyl resin, hydroxymethyl resin, benzhydrylamine resin, aminomethyl resin, 4-benzyloxybenzyl alcohol resin, 4-methylbenzhydrylamine resin, PAM resin, and 4-hydroxy resin.
  • Methylmethylphenylacetamidomethyl resin, polyacrylamide resin, 4- (2 ', 4'dimethoxyphenylhydroxymethyl) phenoxy resin, 4- (2', 4'dimethyloxyphenyl Fmocaminoethyl) Phenoxy resins and the like can be mentioned.
  • amino acids having appropriately protected amino groups and side chain functional groups are condensed on the resin according to various known condensation methods in accordance with the amino acid sequence of the target protein or peptide.
  • a protein or peptide is cleaved from the resin, and at the same time, various protecting groups are removed.
  • an intramolecular disulfide bond formation reaction is performed in a highly diluted solution to obtain a target protein or partial peptide. Or its amide form.
  • various activating reagents that can be used for protein synthesis can be used, and carbodiimides are particularly preferable.
  • the carbopimides include DCC, ⁇ , ⁇ ′-diisopropyl carbopimide, and ⁇ -ethyl- ⁇ ′-(3-dimethylaminoprolyl) carbopimide.
  • Activation by these involves the addition of a protected amino acid directly to the resin along with a racemization inhibitor additive (eg, HOBt, HOOBt), or a symmetrical acid anhydride or HOBt ester or H ⁇ Bt ester. Can be added to the resin after activation of the protected amino acid in advance.
  • the solvent used for activating the protected amino acid or for 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, and N-methylpyrrolidone
  • halogenated hydrocarbons such as methylene chloride and chloroform
  • alcohols such as trifluoroethanol.
  • sulfoxides such as dimethylsulfoxide, ethers such as pyridine, dioxane and tetrahydrofuran, nitriles such as acetonitrile and propionitrile, esters such as methyl acetate and ethyl acetate, or an appropriate mixture thereof.
  • the reaction temperature is appropriately selected from the range known to be usable for the protein bond formation reaction, and is usually selected from the range of about 20 ° 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, succinyl-pentyloxycarbonyl, isoporiloxycarbonyl, 4-methoxybenzyloxycarbonyl, C1-Z, Br-Z, Adamantyloxycarbonyl, trifluoroacetyl, phthaloyl, formyl, 2-ditrophenylsulfenyl, diphenylphosphinothioyl, Fmoc and the like are used.
  • the lipoxyl group may be, for example, an alkyl esterified (eg, methyl, ether, propyl, butyl, tert-butyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, 2-adamantyl, etc.) Or cyclic alkyl esterification), aralkyl esterification (eg, benzyl ester, 4-nitrobenzyl ester, 4-methoxybenzyl ester, 4-cyclobenzyl ester, benzhydryl esterification), phenacyl ester Can be protected by benzyloxycarbonyl hydrazide, short-lived butoxycarbonyl hydrazide, trityl hydrazide, etc.
  • alkyl esterified eg, methyl, ether, propyl, butyl, tert-butyl, cyclopentyl, cyclohexyl,
  • the hydroxyl group of serine can be protected, for example, by esterification or etherification.
  • esterification for example, a lower alkanol group such as an acetyl group, an aroyl group such as a benzoyl group, a group derived from carbonic acid such as a benzyloxycarbonyl group, an ethoxycarbonyl group and the like are used.
  • groups suitable for etherification include, for example, a benzyl group, a tetrahydropyranyl group, and a t-peptizole group.
  • the protecting group of the phenolic hydroxyl group of tyrosine for example, B zl, C 1 2 - B z 1, 2 twelve Torobenjiru, B r- Z, such as tertiary butyl is used.
  • Examples of the protecting group for histidine imidazole include Tos, 4-methoxy-2,3,6-trimethylbenzenesulfonyl, DNP, benzyloxymethyl, Bum, Boc, Trt, and Fmoc.
  • Activated carbonyl groups of the raw materials include, for example, corresponding acid anhydrides, azides, active esters [alcohols (eg, pentachlorophenol, 2,4,5-trichloromouth phenol, 2,4-dinitro Phenol, cyanomethyl alcohol, paranitrophenol, H ⁇ NB, N-hydroxysuccinimide, N-hydroxyphthalimide, and esters with HOBt).
  • active esters eg, pentachlorophenol, 2,4,5-trichloromouth phenol, 2,4-dinitro Phenol, cyanomethyl alcohol, paranitrophenol, H ⁇ NB, N-hydroxysuccinimide, N-hydroxyphthalimide, and esters with HOBt.
  • active esters eg, pentachlorophenol, 2,4,5-trichloromouth phenol, 2,4-dinitro Phenol, cyanomethyl alcohol, paranitrophenol, H ⁇ NB, N-hydroxysuccinimide, N-hydroxyphthalimide, and esters with H
  • Methods for removing (eliminating) the protecting group include, for example, catalytic reduction in a hydrogen stream in the presence of a catalyst such as Pd-black or Pd-carbon, or hydrogen fluoride anhydride or methanesulfonic acid. , Trifluoromethanesulfonic acid, trifluoroacetic acid or this Acid treatment with a mixed solution thereof, base treatment with diisopropylethylamine, triethylamine, piperidine, piperazine, and the like, and reduction with sodium in liquid ammonia are also used.
  • the elimination reaction by the above acid treatment generally takes about one
  • the reaction is carried out at a temperature of 20 ° C to 40 ° C.
  • the 2,4-dinitrophenyl group used as the imidazole protecting group of histidine is removed by thiophenol treatment, and the formyl group used as the indole protecting group of tributofan is 1,2-ethanedithiol, 1,4 —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.
  • an amide form of a protein for example, first, after amidating and protecting the ⁇ -hydroxyl group of the carboxy-terminal amino acid, a peptide (protein) chain is added to the amino group side to a desired chain length. After the elongation, a protein was prepared by removing only the protecting group of the ⁇ -amino group at the ⁇ -terminal of the peptide chain and a protein was obtained by removing only the protecting group of the carboxy group at the C-terminus. In such a mixed solvent. Details of the condensation reaction are the same as described above. After purifying the protected protein obtained by the condensation, all the protecting groups are removed by the above-mentioned method, and a desired crude protein can be obtained. This crude protein is purified by various known purification means, and the main fraction is freeze-dried to obtain an amide of the desired protein.
  • ester for example, after condensing the carboxyl group of the carboxy terminal amino acid with a desired alcohol to form an amino acid ester, the ester of the desired protein is converted in the same manner as the amide of the protein. Obtainable.
  • the partial peptide of the receptor protein of the present invention or a salt thereof can be obtained by cleaving the receptor protein of the present invention with an appropriate peptidase according to a known peptide synthesis method. And can be manufactured by As a peptide synthesis method, for example, any of a solid phase synthesis method and a liquid phase synthesis method may be used. That is, the objective peptide is produced by condensing a partial peptide or amino acid capable of constituting the receptor protein of the present invention with the remaining portion, and if the product has a protecting group, removing the protecting group to produce the desired peptide. Can be.
  • Known condensation methods and elimination of protecting groups include, for example, the following (i) to
  • the partial peptide of the present invention can be purified and isolated by a combination of ordinary purification methods, for example, solvent extraction, distillation, column chromatography, 'liquid chromatography', and recrystallization.
  • the partial peptide obtained by the above method is a free form, it can be converted to an appropriate salt by a known method, and conversely, when it is obtained as a salt, it is converted to a free form by a known method. be able to.
  • the polynucleotide encoding the receptor protein of the present invention is not particularly limited as long as it contains the above-described nucleotide sequence (DNA or RNA, preferably DNA) encoding the receptor protein of the present invention. Is also good.
  • the polynucleotide is RNA such as DNA or mRNA encoding the receptor protein of the present invention, and may be double-stranded or single-stranded. In the case of double-stranded, it may be double-stranded DNA, double-stranded RNA or DNA: RNA hybrid. In the case of a single strand, it may be a sense strand (ie, a coding strand) or an antisense strand (ie, a non-coding strand).
  • Examples of the DNA encoding the receptor protein of the present invention include genomic DNA, genomic DNA library, cDNA derived from the above-described cells and tissues, and cells derived from the above-described cells. It may be either a conventional cDNA library or synthetic DNA.
  • the vector used for the library may be any of bacteriophage, plasmid, cosmid, phagemid and the like.
  • DNA may be directly amplified by reverse transcriptase polymerase chain reaction (hereinafter abbreviated as RT-PCR) using a preparation of all RNA or mRNA fractions from the above-mentioned cells' tissues. it can.
  • RT-PCR reverse transcriptase polymerase chain reaction
  • a DNA containing the nucleotide sequence represented by SEQ ID NO: 12 or the nucleotide represented by SEQ ID NO: 12 Has DNA that hybridizes under high stringent conditions to DNA containing the sequence, and has substantially the same activity as the receptor protein of the present invention (eg, ligand binding activity, signal transduction activity, cell death)
  • Any DNA may be used as long as it encodes a receptor protein having inhibitory activity, cell adhesion activity, and cell motility regulating activity.
  • Examples of the DNA which hybridizes with the DNA having the nucleotide sequence represented by SEQ ID NO: 12 under high stringent conditions include, for example, about 55% or more of the nucleotide sequence represented by SEQ ID NO: 12; Preferably containing a base sequence having a homology of about 60% or more, preferably about 70% or more, preferably about 80% or more, more preferably about 90% or more, and still more preferably about 95% or more. DNA or the like is used.
  • Hybridization can be performed by a known method or a method analogous thereto, for example, according to the method described in Molecular Cloning 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). When a commercially available library is used, it can be carried out according to the method described in the attached instruction manual. More preferably, hybridization can be performed under high stringency conditions.
  • the high stringency conditions include, for example, a sodium concentration of about 19 to 40 mM, preferably about 19 to 20 mM, and a temperature of about 50 to 70 ° C, preferably about 60 to 70 ° C.
  • the condition of 65 T: is shown. In particular, the case where the sodium concentration is about 19 mM and the temperature is about 65 ° C is most preferable.
  • the DNA encoding the partial peptide of the present invention may be any DNA containing the above-described nucleotide sequence encoding the partial peptide of the present invention. Also, 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.
  • the vector used for the library may be any of bacteriophage, plasmid, cosmid, phagemid and the like. DNA can also be directly amplified by the RT-PCR method using an mRNA fraction prepared from the cells and tissues described above.
  • the DNA encoding the partial peptide of the present invention includes, for example, DNA having a partial nucleotide sequence of DNA containing the nucleotide sequence represented by SEQ ID NO: 12, or DNA represented by SEQ ID NO: 12.
  • a DNA having a partial nucleotide sequence of the DNA to be used is used.
  • a DNA encoding the receptor protein of the present invention may be used.
  • Amplification by the PCR method using a synthetic DNA primer having a partial nucleotide sequence of the nucleotide sequence, or DNA incorporating the DNA into a suitable vector is performed for partial or whole region of the receptor protein of the present invention. Selection can be performed by hybridization with a DNA fragment to be coded or labeled with a synthetic DNA.
  • the DNA base sequence can be replaced by ODA-LA PCR using PCR or a known kit, for example, Mutan TM -super Express Km (Takara Shuzo Co., Ltd.), Mutan TM -K (Takara Shuzo Co., Ltd.), etc.
  • the method can be performed according to known methods such as the gapped duplex method and the Kunkel method, or a method analogous thereto.
  • the DNA encoding the cloned receptor 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 at the 5 'end, and TAA, TGA or TAG as a translation stop codon at the 3' end. These translation initiation codon and translation termination codon can also be added using a suitable synthetic DNA adapter.
  • the DNA encoding the receptor protein of the present invention includes, for example, (a) a DNA fragment encoding the receptor protein of the present invention; Can be incorporated into an expression vector by ligating the DNA downstream of the promoter in an appropriate expression vector.
  • vectors for cloning or expression include plasmids derived from E. coli (eg, pCR4, pCR2.K pBR322, pBR325, pUC12, pUC13), and plasmids derived from Bacillus subtilis (eg, pUB110, pTP5, pC194) ), Yeast-derived plasmids (eg, pSH19,
  • PSH15 bacteriophage
  • bacteriophage such as ⁇ phage
  • animal viruses such as retrovirus, vaccinia virus, baculovirus, etc.
  • pAl-11 XTK pRc / CMV, pRc / RSV, pcDNAI / Neo, etc.
  • pAl-11 XTK pRc / CMV, pRc / RSV, pcDNAI / Neo, etc.
  • the promoter may be any promoter as long as it is appropriate for the host used for gene expression.
  • SRo when an animal cell is used as a host, SRo; a promoter, an SV40 promoter, an LTR promoter, a CMV promoter, an HSV-TK promoter, and the like can be mentioned. Of these, it is preferable to use CMV promoter, SRa promoter and the like.
  • S S When the host is Escherichia, trp promoter, 1 ac promoter, recA promoter, ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ promoter, 1 ⁇ promoter, etc., and when the host is Bacillus, S S When the host is yeast, such as ⁇ 1 promoter, S ⁇ 2 promoter, pe ⁇ promoter, etc., ⁇ 05 promoter, PGK promoter, GAP promoter, ADH promoter and the like are preferable. If the host is an insect cell, the polyhedrin promoter and the P10 promoter
  • expression vectors include, if desired, an enhancer, a splicing signal, a polyA addition signal, a selection marker, and an SV40 replication origin (hereinafter, referred to as the
  • Examples of the c- selectable marker that can be used include a gene for dihydrofolate reductase (hereinafter sometimes abbreviated as dhfr) gene [methotrexate (MTX) resistance ], An ampicillin resistance gene (hereinafter sometimes abbreviated as Amp 1 ), a neomycin resistance gene (hereinafter sometimes abbreviated as Ne of, G418 resistance), and the like.
  • dhfr gene for dihydrofolate reductase
  • MTX metalhotrexate
  • Amp 1 An ampicillin resistance gene
  • Ne of, G418 resistance neomycin resistance gene
  • the target gene can be selected even with a thym-free medium.
  • a signal sequence suitable for the host can be added to the N-terminal side of the receptor protein of the present invention. If the host is a genus Escherichia, the Ph A signal sequence, the ⁇ mp A signal sequence, etc., if the host is a Bacillus genus, the ⁇ -amylase signal sequence, subtilisin signal sequence, etc. If the host is yeast, MF, signal sequence, SUC2, signal sequence, etc.If the host is an animal cell, insulin signal sequence, Hi-interferon, signal sequence, antibody molecule, signal sequence, etc. Are available respectively.
  • a transformant can be produced.
  • Examples of the host used in the present invention include Escherichia bacteria, Bacillus bacteria, yeast, insect cells, insects, animal cells, and the like.
  • Escherichia coli K12-DH1 Escherichia coli K12-DH1 [Proc. Natl. Acad. Sci. USA, 60, 160 (1968)], JM103.
  • Bacillus bacteria include, for example, Bacillus subtilis (Bacillus).
  • yeast examples include, for example, Saccharomyces cerevisiae AH22, AH22R-, ONA87-11A, DKD-5D, 20B-12, Schizosaccharomyces pombe NCYC1913, NCYC2036 Pichia pastoris (Pichia pastoris) ) Is used.
  • Insect cells include, for example, when the virus is AcNPV, a cell line derived from a larva of Spodoptera (Spodoptera frugiperda cell; Si cell), or the midgut of Trichoplusia ni Original MG1 cells, High Five TM cells derived from Trichoplus ia ni eggs, Mames tra
  • Sf cells derived from brass icae or cells derived from Estigmena acrea are used.
  • viruses When the virus is BmNPV, a cell line derived from silkworm (Bombyx mori N; BmN cell) is used.
  • Sf cells for example, Sf9 cells (ATCC CRL1711), Sf21 cells (hereinafter, In Vivo, 13, 213-217 (1977)) and the like are used.
  • insects for example, silkworm larvae are used [Nature, vol. 315, 592 (1985)].
  • animal cells examples include monkey cells COS-7, Vero, Chinese Hams Yuichi cell CH0 (hereinafter abbreviated as CH0 cells), dhfr gene-deficient Chinese Hams Yuichi cells CH0 (hereinafter abbreviated as CHO (dhfr) cells). ), Mouse L cells, mouse AtT-20, mouse myeloma cells, rat GH3, human FL cells, and the like.
  • Transformation of a bacterium belonging to the genus Escherichia can be performed, for example, according to the method described in Proc. Natl. Acad. Sci. USA, 69, 2110 (1972) or Gene, 17, 107 (1982). it can.
  • Transformation of Bacillus can be performed, for example, according to the method described in Molecular & General Genetics, Vol. 168, 111 (1979).
  • Transformation of yeast can be performed, for example, according to the method described in Methods in Enzymology, Vol. 194, 182-187 (1991), Proc. Natl. Acad. Sci. USA, Vol. 75, 1929 (1978). Can be.
  • Insect cells or insects can be transformed, for example, according to the method described in Bio / Technology, 6, 47-55 (1988).
  • a liquid medium is suitable as a medium used for the cultivation, and a carbon source necessary for the growth of the transformant is contained therein.
  • Nitrogen sources inorganic substances and others.
  • a carbon source For example, glucose, dextrin, soluble starch, sucrose, etc.Nitrogen sources include, for example, inorganic or nitric acid salts such as ammonium salts, nitrates, corn steep liquor, peptone, power zein, meat extract, soybean meal, potato extract, etc.
  • the 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 pH of the medium is preferably about 5-8.
  • an M9 medium containing glucose and casamino acids is preferable. If necessary, an agent such as 3-indolylacrylic acid can be added to make the promoter work efficiently.
  • the cultivation is usually performed at about 15 to 43 ° C for about 3 to 24 hours, and if necessary, aeration and stirring may be added.
  • the cultivation is usually performed for about 30 to 4 (TC for about 6 to 24 hours, and if necessary, aeration and stirring may be applied.
  • the culture medium When culturing a transformant in which the host is yeast, the culture medium may be, for example, a Burkholder minimum medium [Proc. Natl. Acad. Sci. USA, 77, 4505 (1980)] or 0.5 Natl. Acad. Sci. USA, 81, 5330 (1984)]].
  • the pH of the medium is preferably adjusted to about 5-8. Cultivation is usually carried out at about 20 ° C to 35 for about 24 to 72 hours, with aeration and agitation as necessary.
  • a medium such as Grace's Insect Medium (Nature, 195, 788 (1962)) is supplemented as appropriate with an additive such as immobilized 10% serum.
  • an additive such as immobilized 10% serum.
  • the pH of the medium is adjusted to about 6.2-4.
  • Culture is usually performed at about 27 ° C. for about 3 to 5 days, and if necessary, aeration and agitation are added.
  • a MEM medium containing about 5 to 20% fetal bovine serum [Science, 122, 501 (1952)], a DMEM medium [Virology , 8 vol., 396 (1959)], RPMI1640 medium [The Journal of the Amer i can Med i cal Assoc i ation, Volume 199, 519 (1967)], 199 medium [Proceeding of the Socie ty for or the Biologi cal Med ici ne, Volume 73, 1 (1950) ] Etc. are used.
  • the pH is about 6-8.
  • Culture is usually performed at about 30 ° C to 4 (TC for about 15 to 60 hours, and aeration and agitation are added as necessary.
  • the receptor protein of the present invention can be produced in the cells, in the cell membrane, or outside the cells of the transformant.
  • Separation and purification of the receptor protein of the present invention from the above culture can be performed, for example, by the following method.
  • the cells or cells are collected by a known method, suspended in an appropriate buffer, and subjected to ultrasonication, lysozyme and / or lysozyme.
  • a method of destroying bacterial cells or cells by freeze-thawing or the like, and then obtaining a crude extract of the receptor protein 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 TM.
  • Purification of the receptor protein contained in the thus obtained culture supernatant or extract can be carried out by appropriately combining known separation and purification methods.
  • These known separation and purification methods include methods that utilize solubility such as salting out and solvent precipitation, and methods that mainly use the difference in molecular weight, such as dialysis, ultrafiltration, and gel filtration.
  • a method that utilizes a difference in charge such as ion exchange chromatography; a method that utilizes a specific affinity such as affinity chromatography; a method that utilizes a difference in hydrophobicity such as reverse-phase high-performance liquid chromatography;
  • a method utilizing the difference between isoelectric points such as point electrophoresis is used.
  • the receptor protein thus obtained When the receptor 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 receptor protein is obtained as a salt, the known method is used. Alternatively, it can be converted to a free form or another salt by a method analogous thereto.
  • the receptor protein produced by the recombinant can be arbitrarily modified or partially modified by the action of an appropriate protein-modifying enzyme before or after purification. Can also be removed.
  • an appropriate protein-modifying enzyme for example, trypsin, chymotrypsin, arginyl endopeptidase, protein kinase, glycosidase and the like are used.
  • the activity of the receptor protein of the present invention or a salt thereof thus produced can be measured by a binding experiment with a labeled ligand, an enzymimnoassay using a specific antibody, or the like.
  • the antibody against the receptor protein or its partial peptide or its salt of the present invention may be any of a polyclonal antibody and a monoclonal antibody as long as it is an antibody that can recognize the receptor protein or its partial peptide or its salt of the present invention. Is also good.
  • An antibody against the receptor protein of the present invention or a partial peptide thereof or a salt thereof may be a known antibody using the receptor protein or the like of the present invention as an antigen. Alternatively, it can be produced according to a method for producing an antiserum.
  • the receptor protein or the like of the present invention is administered to a mammal at a site capable of producing an antibody by administration itself or together with a carrier or a diluent.
  • Complete Freund's adjuvant / incomplete Freund's adjuvant may be administered in order to enhance antibody production upon administration.
  • the administration is usually performed once every 2 to 6 weeks, for a total of about 2 to 10 times. Examples of mammals to be used include monkeys, rabbits, dogs, guinea pigs, mice, rats, sheep, and goats, and mice and rats are preferably used.
  • a warm-blooded animal immunized with the antigen for example, a mouse 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.
  • a monoclonal antibody-producing hybridoma By fusing the antibody-producing cells contained in the above with myeloma cells, a monoclonal antibody-producing hybridoma can be prepared.
  • the antibody titer in the antiserum can be measured, for example, by reacting the labeled receptor protein or the like described below with antiserum, and then measuring the activity of the labeling agent bound to the antibody. .
  • the fusion operation is performed in a known manner, for example, in the case of Keller and Milstein. Act (Nature, 256, 495, 1975).
  • the fusion promoter include polyethylene glycol (PEG) and Sendai virus, and PEG is preferably used.
  • myeloma cells examples include NS-1, P3UK SP2 / 0 and the like, and P3U1 is preferably used.
  • the preferred ratio between the number of somatic cells (spleen cells) and the number of myeloma cells used is about 1: 1 to 20: 1, and PEG (preferably PEG1000 to
  • PEG6000 is added at a concentration of about 10 to 80%, and the cell fusion can be carried out efficiently by incubating at about 20 to 40 ° (: preferably at about 30 to 37 ° C for about 1 to 10 minutes).
  • hybridomas are cultured on a solid phase (eg, microplate) onto which an antigen such as a receptor protein is adsorbed directly or together with a carrier. Then, an anti-immunoglobulin antibody (anti-mouse immunoglobulin antibody is used if the cell used for cell fusion is a mouse) or protein A, which is labeled with a radioactive substance or an enzyme, is added.
  • a solid phase eg, microplate
  • an antigen such as a receptor protein
  • an anti-immunoglobulin antibody anti-mouse immunoglobulin antibody is used if the cell used for cell fusion is a mouse
  • protein A which is labeled with a radioactive substance or an enzyme
  • a method for detecting monoclonal antibodies bound to a solid phase adding a hybridoma culture supernatant to a solid phase to which an anti-immunoglobulin antibody or protein A is adsorbed, and adding a receptor protein or the like labeled with a radioactive substance, an enzyme, etc. And a method for detecting a monoclonal antibody bound to a solid phase.
  • the selection of the monoclonal antibody can be carried out according to a known method or a method analogous thereto. Usually, it can be carried out in a medium for animal cells to which HAT (hypoxanthine, aminopterin, thymidine) is added.
  • HAT hyperxanthine, aminopterin, thymidine
  • any medium can be used as long as it can grow a hybridoma.
  • RPMI 1640 medium containing 1-20%, preferably 10-20% fetal bovine serum, GIT medium containing 1-10% fetal bovine serum (Wako Pure Chemical Industries, Ltd.) or hybridoma culture medium Serum medium (SFM-101, Nissui Pharmaceutical Co., Ltd.) or the like can be used.
  • the cultivation temperature is usually 20 to 40 ° (:, preferably about 37.
  • the cultivation time is usually 5 days to 3 weeks, preferably 1 week to 2 weeks.
  • the cultivation is usually 5% CO 2 gas.
  • the antibody titer of the culture supernatant of the hybridoma 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 in the same manner as normal polyclonal antibodies.
  • salting out alcohol precipitation, isoelectric focusing, electrophoresis, ion exchangers (ex. , DEAE) adsorption / desorption method, ultracentrifugation method, gel filtration method, antigen-binding solid phase or specific purification by collecting only the antibody using an active adsorbent such as protein A or protein G and dissociating the bond to obtain the antibody Act].
  • 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 receptor protein of the present invention) and a carrier-protein is formed, and a mammal 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 receptor protein or the like and separating and purifying the antibody.
  • the type of carrier protein and the mixing ratio of carrier to hapten are determined by the ratio of hapten immunized by cross-linking to carrier protein. Any antibody may be cross-linked at any ratio as long as it can be efficiently produced.
  • serum serum alpmin, shishiro glopurin, keyhole, limpet, hemocyanin, etc. are weight ratios.
  • a method of coupling the hapten 1 at a ratio of about 0.1 to 20 and preferably about 1 to 5 is used.
  • condensation product is administered to a warm-blooded animal itself or together with a carrier or diluent at a site where antibody production is possible.
  • Complete Freund's adjuvant / incomplete Freund's adjuvant may be administered in order to enhance the antibody-producing ability upon administration. The administration can usually be performed once every about 2 to 6 weeks, for a total of about 3 to 10 times.
  • the polyclonal antibody can be collected from blood, ascites, etc., preferably from blood, of the mammal 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 serum 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.
  • non-human mammals e.g., mouse, rat, rabbit, sheep, pigeon, pig, cat, dog, monkey, etc., more specifically, dementia rat, obese mouse, atherosclerotic rabbit, For cancer-bearing mice
  • human cells e.g., human cells
  • a specific organ eg., brain, lung, large intestine, etc.
  • a tissue or cell isolated from the organ is obtained.
  • the mRNA of the receptor protein of the present invention or its partial peptide contained in the obtained cells is quantified by, for example, extracting mRNA from cells or the like by a usual method, and using, for example, a technique such as TaqMan PCR. It can also be analyzed by performing Northern blotting by known means.
  • a transformant expressing the receptor protein of the present invention or a partial peptide thereof is prepared according to the above method, and the mRNA of the receptor protein of the present invention or the partial peptide thereof contained in the transformant is prepared. It can be quantified and analyzed in the same way.
  • the antibody of the present invention can specifically recognize the receptor protein of the present invention and the like. Can be used for quantification of the receptor protein and the like, particularly for quantification by sandwich immunoassay. For example,
  • one antibody is an antibody that recognizes the N-terminal of the receptor protein or the like of the present invention
  • the other antibody is an antibody that reacts with the C-terminal of the receptor protein or the like of the present invention. Is preferred.
  • the receptor protein and the like of the present invention can be measured using a monoclonal antibody against the receptor protein and the like of the present invention (hereinafter sometimes referred to as the monoclonal antibody of the present invention), and detection by tissue staining and the like can be performed. You can do it too.
  • the antibody molecule itself may be used, or F (ab ') 2 , Fab', or Fab fraction of the antibody molecule may be used.
  • the measurement method using an antibody against the receptor protein or the like 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 an antigen (for example, the amount of a receptor protein) in a liquid to be measured.
  • any measurement method may be used as long as the amount of the body is detected by chemical or physical means, and the amount is calculated from a standard curve prepared using a standard solution containing a known amount of antigen.
  • nephrometry, a competitive method, an immunometric method, and a sandwich method are suitably used, but in terms of sensitivity and specificity, it is particularly preferable to use a sandwich method described later.
  • a labeling agent used in a measurement method using a labeling substance for example, a radioisotope, an enzyme, a fluorescent substance, a luminescent substance and the like are used. Radioisotopes, if example embodiment, [125 1], 131 1], 3 ⁇ 4], "c], etc.
  • the enzyme large preferably stable and specific activity, e.g., beta -.
  • Galactosidase one ⁇ -glucosidase, alkaline phosphatase, peroxidase, malate dehydrogenase, etc.
  • the fluorescent substance for example, fluorescamine, fluorescein isothiocyanate, etc., is used.
  • a biotin-avidin system may be used for binding an antibody or antigen to a labeling agent.
  • insolubilization of the antigen or antibody physical adsorption may be used.
  • a method using a chemical bond used for insolubilizing or immobilizing white matter or an enzyme may be used.
  • the carrier for example, insoluble polysaccharides such as agarose, dextran, and cellulose; synthetic resins such as polystyrene, polyacrylamide, and silicon; and glass are used.
  • the test solution is reacted with the insolubilized monoclonal antibody of the present invention (primary reaction), and further reacted with the labeled monoclonal antibody of the present invention (secondary reaction).
  • primary reaction the insolubilized monoclonal antibody of the present invention
  • secondary reaction the 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 is not necessarily one kind, and a mixture of two or more kinds of antibodies is used for the purpose of improving measurement sensitivity and the like. May be used.
  • the monoclonal antibody of the present invention used in the primary reaction and the secondary reaction is preferably an antibody having a different binding site to the receptor protein or the like.
  • 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 receptor protein, the antibody used in the primary reaction is preferably the C-terminal.
  • an antibody that recognizes other than the N-terminal part is used.
  • the monoclonal body of the present invention can be used in a measurement system other than the sandwich method, for example, a competition method, an immunometric method, or a nephrometry.
  • a competition method the antigen in the test solution and the labeled antigen are allowed to react competitively with the antibody, and then the unreacted labeled antigen is separated from (F) and the labeled antigen (B) bound to the antibody. Then (BZF separation), the amount of labeling of either B or F is measured, and the amount of Bobara in the test solution is quantified.
  • a soluble antibody is used as the antibody
  • B / F separation is performed using polyethylene glycol
  • a liquid phase method using a second antibody against the above antibody or a solid phase antibody is used as the first antibody.
  • a solid phase method using a soluble first antibody and a solid phase antibody as the second antibody is used.
  • the antigen in the test solution and the immobilized antigen After a competitive reaction with the body, the solid phase and the liquid phase are separated, or the antigen in the test solution is allowed to react with an excessive amount of the labeled antibody, and then the immobilized antigen is added and unreacted After binding the labeled antibody to the solid phase, the solid phase and the liquid phase are separated. Next, the amount of label in any phase is measured to determine the amount of antigen in the test solution.
  • nephelometry the amount of insoluble sediment generated as a result of 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 precipitate is obtained, laser nephrometry utilizing laser scattering is preferably used.
  • the receptor protein of the present invention or a salt thereof can be quantified with high sensitivity.
  • a drug containing an antisense oligonucleotide The oligonucleotide of the present invention is stable in cells, has high cell permeability, has high affinity for the target sense strand, and has low toxicity.
  • the polynucleotide of the present invention may be, for example, a disease associated with overexpression of the receptor protein of the present invention, such as cancer (eg, non-small cell lung cancer, ovarian cancer, prostate cancer, gastric cancer, bladder cancer, breast cancer, cervix) Cancer, colon cancer, rectal cancer, etc.), and can be used as a medicament such as a prophylactic or Z or therapeutic agent.
  • cancer eg, non-small cell lung cancer, ovarian cancer, prostate cancer, gastric cancer, bladder cancer, breast cancer, cervix
  • Cancer colon cancer
  • rectal cancer etc.
  • the antisense oligonucleotides of the present invention may contain altered or modified sugars, bases, or bonds, may be provided in special forms such as ribosomes, microspheres, or may be applied by gene therapy. , Can be provided in an added form. Additional forms include polycations such as polylysine, which acts to neutralize the charge on the phosphate backbone, and lipids that enhance interaction with cell membranes or increase nucleic acid uptake (eg, Hydrophobic substances such as phospholipids and cholesterol). Preferred lipids for addition include cholesterol and its derivatives (eg, cholesteryl chloroformate, cholic acid, etc.).
  • nucleic acids 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 capping groups specifically arranged at the 3 'end or 5' end of nucleic acids for preventing degradation by nucleases such as exonuclease and RNase. Examples of such capping groups include, but are not limited to, hydroxyl-protecting groups known in the art, including glycols such as polyethylene glycol and tetraethylene glycol.
  • the antisense oligonucleotide of the present invention When used as the above medicine, it can be formulated and administered according to a known method.
  • the antisense oligonucleotide when used, the antisense oligonucleotide 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 non-human administration to human or non-human mammals eg, rats, gray egrets, sheep, higgs, bush, red sea lions, cats, dogs, monkeys, etc.
  • human or non-human mammals eg, rats, gray egrets, sheep, higgs, bush, red sea lions, cats, dogs, monkeys, etc.
  • the antisense oligonucleotide 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.
  • a physiologically acceptable carrier such as an adjuvant for promoting uptake
  • the dose of the antisense oligonucleotide varies depending on the target disease, the subject of administration, the route of administration, and the like.
  • the antisense oligonucleotide of the present invention may be administered to an organ (eg, liver, lung, When topically administered to the heart, kidney, etc., generally, for an adult (body weight 60 kg), about 0.1 to 100 mg of the antisense oligonucleotide is administered per day.
  • the antisense oligonucleotide can also be used as a diagnostic oligonucleotide probe for examining the presence of the polynucleotide (eg, DNA) of the present invention in tissues or cells and the state of expression thereof.
  • the polynucleotide eg, DNA
  • the present invention provides a transformant transformed with a recombinant DNA in which a reporter gene is ligated downstream (under expression control) of the transcriptional regulatory region of the gene of the receptor protein of the present invention in the presence of a test compound.
  • a method for screening for a compound that promotes or inhibits the transcriptional regulatory activity comprising measuring and comparing each repo overnight activity when cultured in the absence and presence of the same, and a script for this method.
  • a kit for single use is also provided.
  • Examples of the transcription regulatory region of the gene of the receptor protein of the present invention include a DNA having the same or substantially the same nucleotide sequence as the nucleotide sequence represented by SEQ ID NO: 20.
  • the DNA containing a base sequence substantially identical to the base sequence represented by SEQ ID NO: 20 has a base sequence that hybridizes under high stringency end conditions, and has substantially the same sequence as SEQ ID NO: 20. Any DNA may be used as long as it has the same transcriptional regulatory activity (eg, promoter-one activity).
  • DNA that can hybridize with the nucleotide sequence represented by SEQ ID NO: 20 under high stringent conditions include, for example, the nucleotide sequence represented by SEQ ID NO: 20 A base having a homology of about 50% or more, preferably about 60% or more, more preferably about 70% or more, more preferably about 80% or more, particularly preferably about 90% or more, and most preferably about 95% or more.
  • a DNA containing the sequence is used.
  • Hybridization is performed according to a method known per se or a method analogous thereto, for example, the method described in Molecular Cloning 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). be able to. When a commercially available library is used, it can be performed according to the method described in the attached instruction manual. More preferably, the reaction can be performed under high stringency conditions.
  • the high stringent end conditions include, for example, a sodium concentration of about 19 to 40 mM, preferably about 19 to 2 OmM, and a temperature of about 50 to 70: preferably about 60 to 65.
  • a sodium concentration of about 19 to 40 mM, preferably about 19 to 2 OmM, and a temperature of about 50 to 70: preferably about 60 to 65.
  • the sodium concentration is about 19 mM and the temperature is about 65 ° C.
  • the transcription regulatory region eg, DNA containing a nucleotide sequence substantially identical to the nucleotide sequence represented by SEQ ID NO: 20
  • the transcription regulatory region is substantially the same as the nucleotide sequence represented by SEQ ID NO: 20
  • a sequence obtained by further adding a genomic DNA sequence at the 5 ′ upstream to the same base sequence (preferably a promoter region) is also included.
  • the number of bases to be added is, for example, about 10 Kb or less, preferably about 5 Kb or less, more preferably about 2 Kb or less, and most preferably about 1 Kb or less.
  • reporter genes include, for example, lacZ (yogalactosidase gene), chloramphenicol acetyltransferase (CAT), luciferase, growth factor,] 3-glucuronidase, alphospholiphosphatase, Green fluorescent protein (GFP), / 3-lacquerase and the like are used.
  • a test compound that increases the amount of the repo overnight gene product can be used as a test compound for the receptor protein of the present invention.
  • a compound having an action of controlling (particularly promoting) transcriptional regulatory activity preferably promoter activity
  • ie, promoting expression of the receptor protein of the present invention can be selected as a compound having a certain activity.
  • a test compound that reduces the amount of a reporter gene product is a compound that has the activity of controlling (particularly inhibiting) the transcriptional regulatory activity (preferably, overnight promoter) of the receptor protein of the present invention, that is, the compound of the present invention.
  • the compound can be selected as a compound having an activity of inhibiting the expression of the receptor protein.
  • the test compound is, for example, a compound selected from peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, plasma, and the like.
  • the salt of the compound those similar to the aforementioned salts of the receptor protein of the present invention are used.
  • the transformant can be cultured in the same manner as the above-described transformant containing the receptor protein of the present invention.
  • the vector construction of the repo overnight gene and the Atsey method can be performed according to known techniques (eg, Molecular Biotechnology 13, 29-43, 1999).
  • the compound having an activity of promoting the expression of the receptor protein of the present invention or a salt thereof is useful as a safe and low-toxic drug.
  • a compound having an activity of inhibiting the expression of the peptide of the present invention or a salt thereof is a safe and low-toxic drug for suppressing the physiological activity of the receptor protein of the present invention, for example, cancer (eg, non-small cell lung cancer) Ovarian cancer, prostate cancer, gastric cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, rectal cancer, etc.).
  • cancer eg, non-small cell lung cancer
  • salt those similar to the aforementioned salts of the receptor protein of the present invention are used.
  • the above compound when used as a medicine, it can be formulated according to conventional means.
  • the compound can be used as a sugar-coated tablet, capsule, elixir, microcapsule or the like as needed, orally, or aseptic solution with water or another pharmaceutically acceptable liquid. It can be used parenterally or in the form of injections such as suspensions.
  • the compound is mixed with known physiologically acceptable carriers, flavoring agents, excipients, vehicles, preservatives, stabilizers, binders, and the like in a unit dosage form generally required for the practice of pharmaceutical preparations. It can be manufactured by The amount of the active ingredient in these preparations is such that a suitable dosage in the specified range can be obtained.
  • prophylactic and therapeutic agents examples include, but are not limited to, a buffer (eg, a phosphate buffer, a sodium acetate buffer), a soothing agent (eg, benzalkonidum chloride, proforce hydrochloride, etc.), a stabilizer (eg, , Human serum albumin, polyethylene glycol, etc.), preservatives (eg, benzyl alcohol, phenol, etc.), antioxidants and the like.
  • a buffer eg, a phosphate buffer, a sodium acetate buffer
  • a soothing agent eg, benzalkonidum chloride, proforce hydrochloride, etc.
  • a stabilizer eg, Human serum albumin, polyethylene glycol, etc.
  • preservatives eg, benzyl alcohol, phenol, etc.
  • antioxidants and the like examples include, but are not limited to, a buffer (eg, a phosphate buffer, a sodium acetate buffer), a soothing agent (eg
  • the preparations obtained in this way are safe and low toxic, so they can be used, for example, in humans and mammals (eg, rats, mice, egrets, higgs, bushes, cats, cats, dogs, dogs, etc.). Can be administered.
  • mammals eg, rats, mice, egrets, higgs, bushes, cats, cats, dogs, dogs, etc.
  • the dose of the compound or a salt thereof varies depending on the administration subject, target organ, symptom, administration method, and the like.
  • oral administration for example, in a cancer patient (with a body weight of 60 kg), About 0.1 to 100 nig per day, preferably about 1.0 to 50 mg, more preferably about 1.0 to 20 nig.
  • parenteral administration the single dose varies depending on the subject of administration, target organ, symptoms, administration method, etc.
  • injection it is usually used, for example, in cancer patients (with a body weight of 60 kg). It is convenient to administer about 0.01 to 30 mg, preferably about 0.1 to 20 mg, more preferably about 0.1 to 10 mg per day by intravenous injection.
  • the dose can be administered in terms of weight per 60 kg.
  • a base, an amino acid, or the like is indicated by an abbreviation, the indication is based on an abbreviation by IUPAC-IUB Communication on Biochemical Nomenclature or a conventional abbreviation in the field. An example is shown below.
  • an amino acid can have optical isomers, the L-form is indicated unless otherwise specified.
  • Trp Tryptophan
  • FIG. 9 shows a part of the nucleotide sequence (3 ′ side) of cDNA obtained in Reference Example 3 below.
  • the nucleotide sequence of cDNA which encodes a novel receptor protein NEPHA derived from human brain is shown.
  • Example 1 shows the base sequence of the oligonucleotide used in Example 1 below.
  • Example 1 shows the base sequence of the oligonucleotide used in Example 1 below.
  • the human brain cDNA (CL0NTECH) contained in MTC Panel I was type III, and PCR was carried out using two primers, primer 1 (SEQ ID NO: 2) and primer 2 (SEQ ID NO: 3). .
  • the composition of the reaction solution used in the reaction was the above cDNA 0.51 as type III, 1 U of Pfu Turbo DNA Polymerase (STRATAGENE), primer 1 (SEQ ID NO: 2) and primer 1 (SEQ ID NO: 3) 1 M each, 200 l of dNTPs, and 101 of 2XGC Buf ferl (Takara Shuzo) were added to give a liquid volume of 201.
  • the PCR reaction was repeated 96 times at 96X3 for 1 minute, followed by a cycle of 96 ° C, 1 minute, 60 ° C, 1 minute, 72 ° C, and 1 minute 40 times.
  • the PCR reaction product was purified by agarose gel electrophoresis, and then purified using the plasmid vector pCR-Bluntil-T0P0 (Invitrogen) according to the prescription of Zero Blunt T0P0 PCR Cloning Kit (Invitrogen). ).
  • This was introduced into E. coli TOP10 and selected on LB agar medium containing kanamycin.
  • SEQ ID NO: 1 nucleotide sequence of cDNA encoding a part of the novel receptor protein was obtained.
  • PCR was carried out using two primers, primer 3 (SEQ ID NO: 5) and primer 4 (SEQ ID NO: 6).
  • the composition of the reaction solution used in the reaction was the above-mentioned cDNA 11 as type III, and 2.5 U of Pfu Turbo DNA Polymerase (STRATAGENE), primer 3 (SEQ ID NO: 5) and primer 4 (SEQ ID NO: 6).
  • STRATAGENE Pfu Turbo DNA Polymerase
  • primer 3 SEQ ID NO: 5
  • primer 4 SEQ ID NO: 6
  • PCR was performed using two primers, Primer 5 (SEQ ID NO: 8) and Primer 1 (SEQ ID NO: 9).
  • the composition of the reaction solution used in the reaction was the above cDNA II as type III, and 2.5 U of Pfu Turbo DNA Polymerase (STRATAGENE), Primer 5 (SEQ ID NO: 8) and Primer 6 (SEQ ID NO: 9) 1 M, dNTPs were added to 200 XM, and 2XGC Buffer1 (Takara Shuzo) to make a liquid volume of 501.
  • the PCR reaction was repeated 40 times at 96, 1 minute, and 60 times for 1 minute, 72 ° C, and 2 minutes. Next, 5 l of the PCR reaction product was used as type ⁇ , and Pfu Turbo DNA Polymerase was used.
  • SEQ ID NO: 12 Based on the nucleotide sequence of the partial cDNA encoding the novel receptor protein obtained in Reference Example 1, Reference Example 2 and Reference Example 3 (SEQ ID NO: 1, SEQ ID NO: 4 and SEQ ID NO: 7), The sequence (SEQ ID NO: 12) was determined. SEQ ID NO: 12 corresponds to the 71st to 789th sequence of SEQ ID NO: 1, the 284th to 1134th sequence of SEQ ID NO: 4 and the 473th to 1926th sequence of SEQ ID NO: 7 . The amino acid sequence of the protein predicted from the nucleotide sequence of the full-length cDNA (SEQ ID NO: 12) is shown in SEQ ID NO: 11.
  • NEPHA novel EphA
  • NEPHA and the human Ephr in receptor EphA7 have 50.7% homology at the amino acid level.
  • the gene encoding the full length is obtained by cleaving the cDNA fragments obtained in Reference Examples 1, 2 and 3 with an appropriate restriction enzyme and ligating them with T4 ligase or the like.
  • a primer is designed to amplify the cDNA encoding the full length based on the nucleotide sequences identified in Reference Examples 1 and 3, and the primers are obtained by PCR.
  • Reference example 5
  • a plasmid containing the nucleotide sequences of the cDNAs obtained in Reference Examples 2 and 3 was digested with restriction enzymes.
  • the composition of the reaction mixture was lg, Xol (Takara Shuzo) 5U, and ⁇ ⁇ Buf, containing a plasmid containing the nucleotide sequence of the cDNA represented by SEQ ID NO: 4 or a plasmid containing the nucleotide sequence of the cDNA represented by SEQ ID NO: 7.
  • fer (Takara Shuzo) 5 x I was added to make a liquid volume of 501.
  • the restriction enzyme reaction was performed at 37 ° C.
  • the nucleotide sequence of cDNA represented by SEQ ID NO: 13 was obtained. Further, a plasmid containing the nucleotide sequence of the cDNA obtained in Reference Example 1 (SEQ ID NO: 1) and a plasmid containing the nucleotide sequence of the cDNA represented by SEQ ID NO: 13 were digested with restriction enzymes.
  • the composition of the reaction solution is lfig, Pvul (Takara Shuzo) 10 U, and plasmid containing the nucleotide sequence of the cDNA represented by SEQ ID NO: 1 or plasmid containing the nucleotide sequence of the cDNA represented by SEQ ID NO: 13.
  • the plasmid containing the nucleotide sequence of the cDNA represented by SEQ ID NO: 1 contains 743 bp and the nucleotide sequence of the cDNA represented by SEQ ID NO: 13
  • the reaction product corresponding to 5977 bp was purified from the plasmid by agarose gel electrophoresis, and these were ligated according to the prescription of DNA Ligation Kit Ver.2 (Takara Shuzo).
  • breast cancer cell line ZR-75-1 (Cancer Res, 38 (10), 3352-3364, purchased from ATCC, 1978) was used. Falcon).
  • OligofectAMINE (Invitrogen) was used and the protocol was followed. Twenty hours after the introduction, RNA was extracted according to the protocol of the RNeasy mini kit (Qiagen), and cDNA was prepared using the TaqManRT kit (Perkin-Elmer). At the same time, the same reaction was carried out without adding reverse transcriptase to obtain a non-reverse transcription control.
  • a Cell death detection ELISA (Roche) was used to examine the effect on the apoptosis when the expression of the NEPHA gene was suppressed by introducing the antisense oligonucleotide.
  • the control oligonucleotide was used to compare with the sample introduced with the oligonucleotide.
  • the expression of the NEPHA gene was 20 hours after the introduction of the antisense oligonucleotide compared to the control oligonucleotide (SEQ ID NO: 15).
  • the amount of expression of the NEPHA gene was reduced to 57% when the antisense (SEQ ID NO: 14) was used, compared to the control oligonucleotide (SEQ ID NO: 16).
  • apoptosis was calculated to be 224% when the control oligonucleotide (SEQ ID NO: 13) was introduced and the control oligonucleotide (SEQ ID NO: 13) was assumed to be 100%. Assuming that the oligonucleotide (SEQ ID NO: 16) was 100%, it was increased to 185% when the antisense oligonucleotide (SEQ ID NO: 14) was introduced.
  • the oligonucleotide of the present invention has low toxicity and is useful as an excellent agent for preventing and / or treating cancer. Further, it can be obtained by screening using a transformant transformed with a recombinant DNA in which a repo overnight gene is ligated downstream (under expression control) of the transcription regulatory region of the receptor protein gene of the present invention.
  • a compound, preferably a compound having an activity of inhibiting the expression of the receptor protein of the present invention or a salt thereof is also low toxicity and is useful as an excellent agent for preventing and / or treating cancer.

Abstract

Oligonucleotides having base sequences which are the same or substantially the same as a base sequence represented by SEQ ID NO:13 or 14 are useful as excellent anticancer agents.

Description

明細 がんの予防および/または治療剤 技術分野  Description Cancer preventive and / or therapeutic agent
本発明は、 がんの予防 ·治療に有用なアンチセンスオリゴヌクレオチド、 がん の予防 ·治療剤のスクリーニングなどに関する。 背景技術  The present invention relates to antisense oligonucleotides useful for cancer prevention and treatment, screening for cancer prevention and treatment agents, and the like. Background art
がんの化学療法では新しい抗がん剤の開発により延命効果が向上し、 治癒に向 かうケースも増えてきている。しかしながら、 現在使用されている抗がん剤は DNA に傷害を与えたり、 細胞分裂を阻害する細胞毒性の強い薬剤が殆どであり、 この ため正常細胞に対しても少なからず傷害を与え、 特に細胞分裂の盛んな骨髄など にしばしば強い副作用が現れる。これらのことから、 がん細胞に特異的に発現す る分子を標的とし、 がん細胞の増殖阻害、 またはアポトーシス誘導を行える薬剤 が切望されている。  In cancer chemotherapy, the development of new anticancer drugs has improved the life-span effect and is increasing the number of cases toward cure. However, most of the currently used anticancer drugs are highly cytotoxic drugs that damage DNA or inhibit cell division. Strong side effects often appear in the rapidly dividing bone marrow. For these reasons, there is a strong need for drugs that target molecules that are specifically expressed in cancer cells and that can inhibit the growth of cancer cells or induce apoptosis.
アンチセンスオリゴヌクレオチドは、 細胞内に導入されると相補的な配列を有 する RNAとハイブリダィズし、 RNas eHによる RNAの分解を誘導して蛋白質翻訳を阻 害する、 あるいはハイブリダィズによる庳接的蛋白質合成阻害ももたらす。 目的 の遺伝子機能を特異的に抑えることが可能なことから、 遺伝子の機能解析手段と して頻用されていると共に、 いくつかのアンチセンスオリゴヌクレオチドは臨床 応用開発が進んでいる。  Antisense oligonucleotides, when introduced into cells, hybridize with RNA having a complementary sequence and induce RNA degradation by RNaseH to inhibit protein translation, or hybridize to inhibit direct protein synthesis. Also bring. Since it is possible to specifically suppress the function of the target gene, it is frequently used as a means for analyzing the function of genes, and some antisense oligonucleotides are being developed for clinical application.
プロテインチロシンキナーゼ遺伝子群は、 癌遺伝子の中でも最大の遺伝子ファ ミリ一を形成し、 細胞の増殖分化や形態形成に深く関与している (Harvey Lec t . 94巻, 81- 1 19頁, 1998- 99年) ことから、 坊癌剤開発の格好の標的遺伝子として 取りあげられてきた。 なかでも受容体型チロシンキナーゼ (RTK) に分類される EGF受容体、 HER2受容体または VEGF受容体を標的とした医薬品開発は、 ヒトに現 実に試されるレベルの抗癌医薬品を生み出してきており、 プロテインチロシンキ ナーゼのなかでも RTKは医薬品として成功確率の高いサブファミリ一であると考 えられ、 RTKの中でも Eph (erythropoietin producing human hepatocellular carcinoma cell line) 受容体遺伝子ファミリ一は現時点で 14種類のメンバ一か ら構成され、 最大規模を有する (Cell 90巻, 403- 404頁, 1997年) 。 Eph受容体 のリガンドとして同定された Ephr i n遺伝子ファミリーもまた、 少なくとも 8種類 の構成メンバーからなることが報告されている。 Eph受容体および Ephr inリガン ドの生理的機能については、 胎生期や脳における極めて限局的な発現様式から発 生期の形態形成やあるいは神経軸索の適切な投射における関与が示唆されている (Annu. Rev. Neurosci. 21巻, 309- 345頁, 1998年) が、 近年癌における役割も 注目されつつある。 例えば、 様々の癌における Eph受容体の発現亢進が報告 Protein tyrosine kinase genes form the largest family of oncogenes and are deeply involved in cell proliferation and differentiation and morphogenesis (Harvey Lecto. 94, 81-119, 1998- 1999), it has been picked up as a good target gene for developing botanicals. In particular, drug development targeting the EGF receptor, HER2 receptor or VEGF receptor, which is classified as receptor tyrosine kinase (RTK), has produced anticancer drugs at a level that can be tested in humans. Among the tyrosine kinases, RTK is considered to be one of the subfamilies with a high probability of success as pharmaceuticals. At present, among the RTKs, the Eph (erythropoietin producing human hepatocellular carcinoma cell line) receptor gene family is currently composed of 14 members and has the largest size (Cell 90, 403-404, 1997). ). The Ephr in gene family, identified as ligands for the Eph receptor, has also been reported to consist of at least eight members. Regarding the physiological functions of Eph receptors and Ephr in ligands, extremely localized expression in embryos and the brain suggests their involvement in morphogenesis during development and proper projection of nerve axons ( Annu. Rev. Neurosci. 21, 309-345 (1998)), but its role in cancer has also attracted attention in recent years. For example, increased expression of Eph receptor in various cancers has been reported
(Oncogene 19巻, 5614-5619頁, 2000年) されている。 さらに、 血管新生におけ る関与も示唆されてきている (Genes & Dev. 13巻, 1055-1066頁, 1999年) 。 がん細胞に発現する分子を標的とし、 がん細胞の増殖阻害、 またはアポトーシ ス誘導を行いうる薬剤が切望されている。 発明の開示  (Oncogene 19, 5614-5619, 2000). Furthermore, its involvement in angiogenesis has been suggested (Genes & Dev. 13, 1055-1066, 1999). There is a strong need for drugs that target molecules expressed in cancer cells and that can inhibit the growth of cancer cells or induce apoptosis. 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、 がん細胞 に発現している NEPHA遺伝子の発現を抑制することにより、 がん細胞がアポトー シスを起こすことを見出し、 この知見に基づいて、 さらに検討を重ねた結果、 本 発明を完成するに至った。  The present inventors have conducted intensive studies to solve the above-mentioned problems, and as a result, found that cancer cells cause apoptosis by suppressing the expression of NEPHA gene expressed in cancer cells. Further studies based on this finding have led to the completion of the present invention.
すなわち、 本発明は、  That is, the present invention
(1) 配列番号: 1 3または配列番号: 14で表される塩基配列と同一または 実質的に同一の塩基配列を含有するオリゴヌクレオチド、  (1) an oligonucleotide having a nucleotide sequence identical or substantially identical to the nucleotide sequence represented by SEQ ID NO: 13 or SEQ ID NO: 14,
(2) 配列番号: 1 3で表される塩基配列からなるオリゴヌクレオチド、 (2) an oligonucleotide having a base sequence represented by SEQ ID NO: 13;
(3) 配列番号: 14で表される塩基配列からなるオリゴヌクレオチド、 (4) アンチセンスオリゴヌクレオチドである上記 (1) 記載のオリゴヌクレ ォチド、 (3) an oligonucleotide consisting of the nucleotide sequence represented by SEQ ID NO: 14, (4) the oligonucleotide according to (1) above, which is an antisense oligonucleotide,
(5) DN Aである上記 (1) 記載のオリゴヌクレオチド、  (5) the oligonucleotide according to the above (1), which is DNA;
(6) 上記 (1) 記載のオリゴヌクレオチドを含有してなる医薬、  (6) a medicine comprising the oligonucleotide according to the above (1),
(7) がんの予防および または治療剤である上記 (6) 記載の医薬、 (8) 配列番号: 20で表される塩基配列と同一または実質的に同一の塩基配 列を含有する DNA、 (7) The medicament according to the above (6), which is an agent for preventing and / or treating cancer. (8) a DNA containing the same or substantially the same base sequence as the base sequence represented by SEQ ID NO: 20,
(9) 配列番号: 12で表される塩基配列と同一または実質的に同一の塩基配 列を含有する DNAの転写調節領域である上記 (8) 記載の DNA、  (9) The DNA according to the above (8), which is a transcription control region of a DNA containing the same or substantially the same nucleotide sequence as the nucleotide sequence represented by SEQ ID NO: 12.
(10) 転写調節領域がプロモーター領域である上記 (9) 記載の DNA、 (10) the DNA according to the above (9), wherein the transcription regulatory region is a promoter region;
(11) 上記 (8) 記載の DNAを含有する組換えベクター、 (11) a recombinant vector containing the DNA of (8) above,
(12) 配列番号: 12で表される塩基配列と同一または実質的に同一の塩基 配列を含有する DN Aの転写調節領域の下流にレポ一ター遺伝子を有する DN A を含有する上記 (11) 記載の組換えべクタ一、  (12) The method according to the above (11), wherein the DNA comprises a DNA having a reporter gene downstream of a transcriptional regulatory region of the DNA having the same or substantially the same nucleotide sequence as the nucleotide sequence represented by SEQ ID NO: 12. Described recombination vector,
(13) 上記 (11) 記載の組換えべクタ一を含有する形質転換体、  (13) a transformant containing the recombinant vector according to the above (11),
(14) 上記 (8) 記載の DNAを用いることを特徴とする、 配列番号: 12 で表される塩基配列と同一または実質的に同一の塩基配列を含有する DN Aの転 写調節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、 (14) The use of the DNA according to (8) above, which promotes the transcription control activity of DNA containing the same or substantially the same nucleotide sequence as the nucleotide sequence represented by SEQ ID NO: 12. Or a method of screening for a compound or a salt thereof that inhibits
(15) 上記 (13) 記載の形質転換体を用 る上記 (14) 記載のスクリ一 ニング方法、 (15) The screening method according to the above (14) using the transformant according to the above (13),
(16) 上記 (8) 記載の DNAを含有することを特徴とする、 配列番号: 1 2で表される塩基配列と同一または実質的に同一の塩基配列を含有する DNAの 転写調節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用キッ 卜、  (16) Promotes the activity of regulating the transcription of DNA having the same or substantially the same nucleotide sequence as the nucleotide sequence represented by SEQ ID NO: 12, characterized by containing the DNA of (8) above. Or a screening kit for a compound that inhibits or a salt thereof,
(17) 上記 (14) 記載のスクリーニング方法または上記 (16) 記載のス クリーニング用キットを用いて得られる配列番号: 12で表される塩基配列と同 一または実質的に同一の塩基配列を含有する DNAの転写調節活性を促進または 阻害する化合物またはその塩、  (17) Contains the same or substantially the same nucleotide sequence as the nucleotide sequence represented by SEQ ID NO: 12 obtained by using the screening method according to (14) or the screening kit according to (16). Compounds or salts thereof that promote or inhibit the activity of regulating the transcription of DNA,
(17 a) 上記 (14) 記載のスクリーニング方法または上記 (16) 記載の スクリーニング用キットを用いて得られる配列番号: 12で表される塩基配列と 同一または実質的に同一の塩基配列を含有する D N Aの転写調節活性を阻害する 化合物またはその塩、  (17a) It contains the same or substantially the same nucleotide sequence as the nucleotide sequence represented by SEQ ID NO: 12 obtained by using the screening method described in (14) or the screening kit described in (16). A compound or a salt thereof that inhibits the transcription regulation activity of DNA,
(18) 上記 (17) 記載の化合物またはその塩を含有してなる医薬、 (18 a) 上記 (17 a) 記載の化合物またはその塩を含有してなる医薬、 (19) がんの予防および Zまたは治療剤である上記 (18) 記載の医薬、 (19 a) がんの予防および/または治療剤である上記 (18 a) 記載の医薬、 (20) 哺乳動物に対して、 上記 (1) 記載のオリゴヌクレオチドの有効量を 投与することを特徴とするがんの予防 ·治療方法、 (18) a medicament comprising the compound according to (17) or a salt thereof, (18a) a medicament comprising the compound according to (17a) or a salt thereof, (19) the medicament according to the above (18), which is an agent for preventing and / or treating cancer; (19a) the medicament according to the above (18a), which is an agent for preventing and / or treating cancer; A method for preventing and treating cancer, which comprises administering to an animal an effective amount of the oligonucleotide according to (1).
(21) がんの予防 ·治療剤を製造するための上記 (1) 記載のオリゴヌクレ ォチドの使用などに関する。 発明を実施するための最良の形態  (21) Use of the oligonucleotide described in (1) above for the manufacture of an agent for preventing or treating cancer. BEST MODE FOR CARRYING OUT THE INVENTION
配列番号: 13または配列番号: 14で表される塩基配列と実質的に同一の塩 基配列を含有するオリゴヌクレオチド (以下、 本発明のオリゴヌクレオチドまた は本発明のアンチセンスオリゴヌクレオチドと略記することもある) は、 配列番 号: 11で表されるアミノ酸配列と同一または実質的に同一のアミノ酸配列を含 有する Eph受容体蛋白質 (以下、 本発明の受容体蛋白質または本発明の Eph 受容体蛋白質と略記することもある) の遺伝子の RNAとハイブリダィズするこ とができ、 該 RN Aの合成または機能を阻害することができる、 あるいは本発明 の受容体蛋白質関連 RN Aとの相互作用を介して該受容体蛋白質遺伝子の発現を 調節および (または) 制御することができる。  Oligonucleotide containing a base sequence substantially identical to the base sequence represented by SEQ ID NO: 13 or SEQ ID NO: 14 (hereinafter, abbreviated as oligonucleotide of the present invention or antisense oligonucleotide of the present invention) Is an Eph receptor protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 11 (hereinafter, the receptor protein of the present invention or the Eph receptor protein of the present invention) (May be abbreviated as) and can inhibit the synthesis or function of the RNA, or through the interaction with the receptor protein-related RNA of the present invention. The expression of the receptor protein gene can be regulated and / or controlled.
配列番号: 13または配列番号: 14で表される塩基配列と実質的に同一の塩 基配列を含有するオリゴヌクレオチドとしては、 配列番号: 13または配列番 号: 14で表される塩基配列とハイストリンジェン卜な条件下でハイブリダィズ する塩基配列を有し、 配列番号: 13または配列番号: 14で表される塩基配列 を有するオリゴヌクレオチドと実質的に同質の活性を有するオリゴヌクレオチド であれば何れのものでもよい。  Oligonucleotides containing a base sequence substantially identical to the base sequence represented by SEQ ID NO: 13 or SEQ ID NO: 14 include nucleotides represented by SEQ ID NO: 13 or SEQ ID NO: 14. Any oligonucleotide having a nucleotide sequence that hybridizes under stringent conditions and having substantially the same activity as the oligonucleotide having the nucleotide sequence represented by SEQ ID NO: 13 or SEQ ID NO: 14 It may be something.
実質的に同質の活性としては、 例えば、 本発明の受容体蛋白質の発現阻害活性 などが挙げられる。 発現阻害活性としては、 例えば転写阻害、 前駆型 RNAのス プライシング阻害、 mRN Aの細胞質移行阻害、 蛋白質への翻訳阻害などが挙げ られる。  Examples of substantially equivalent activities include, for example, the activity of inhibiting the expression of the receptor protein of the present invention. Examples of the expression inhibiting activity include transcription inhibition, splicing inhibition of precursor RNA, inhibition of mRNA translocation to the cytoplasm, and inhibition of translation into protein.
実質的に同質とは、 その活性が性質的に同質であることを示す。 したがって、 上記発現阻害活性などの活性が同等 (例、 約 0. 01〜100倍、 好ましくは約 0 . 5〜2 0倍、 より好ましくは約 0 . 5〜2倍) であることが好ましい。 上記阻害活性などの測定は、 公知の方法に準じて行うことができる。 阻害活性 は、 本発明の受容体蛋白質を含有する形質転換体、 本発明の受容体蛋白質の生体 内または生体外の遺伝子発現系、 あるいは本発明の受容体蛋白質の生体内または 生体外の翻訳系を用いて調べることができる。 例えば、 後述の (A) 本発明の受 容体蛋白質またはその部分ペプチドの mR N A量の測定法、 (B ) 本発明の受容 体蛋白質もしくはその部分べプチドまたはその塩の定量法に記載に方法により、 測定できる。 Substantially the same indicates that the activity is the same in nature. Therefore, activities such as the above-mentioned expression inhibitory activity are equivalent (eg, about 0.01 to 100 times, preferably about (0.5 to 20 times, more preferably about 0.5 to 2 times). The measurement of the above-mentioned inhibitory activity and the like can be performed according to a known method. The inhibitory activity is determined by using a transformant containing the receptor protein of the present invention, an in vivo or in vitro gene expression system of the receptor protein of the present invention, or an in vivo or in vitro translation system of the receptor protein of the present invention. Can be checked using For example, the method described in (A) the method for measuring the mRNA amount of the receptor protein of the present invention or the partial peptide thereof, and the method described in (B) the method for quantifying the receptor protein or the partial peptide or a salt thereof of the present invention described below are used. , Can be measured.
配列番号: 1 3または配列番号: 1 4で表される塩基配列とハイストリンジェ ントな条件下でハイブリダィズできる D NAとしては、 例えば、 配列番号: 1 3 または配列番号: 1 4で表される塩基配列と約 6 0 %以上、 さらに好ましくは約 7 0 %以上、 より好ましくは約 8 0 %以上、 特に好ましくは約 9 0 %以上、 最も 好ましくは約 9 5 %以上の相同性を有する塩基配列を含有する D N Aなどが用い られる。 。  Examples of DNA that can hybridize with the nucleotide sequence represented by SEQ ID NO: 13 or SEQ ID NO: 14 under high stringent conditions include, for example, SEQ ID NO: 13 or SEQ ID NO: 14 A base having about 60% or more, more preferably about 70% or more, more preferably about 80% or more, particularly preferably about 90% or more, and most preferably about 95% or more homology with the base sequence. DNA containing a sequence is used. .
ハイブリダィゼ一シヨンは、 自体公知の方法あるいはそれに準じる方法、 例え ば、 モレキュラー 'クロ一ニング (Molecular Cloning) 2 nd (J. Sambrook et al. , Cold Spr ing Harbor Lab. Press, 1989) に記載の方法などに従って行なう ことができる。 また、 市販のライブラリーを使用する場合、 添付の使用説明書に 記載の方法に従って行なうことができる。 より好ましくは、 ハイストリンジェン トな条件に従って行なうことができる。  Hybridization can be performed by a method known per se or a method analogous thereto, for example, a method described in Molecular Cloning 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). Etc. When a commercially available library is used, the procedure can be performed according to the method described in the attached instruction manual. More preferably, it can be carried out under high stringency 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 t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 の 場合が最も好ましい。  High stringency conditions include, for example, a sodium concentration of about 19 to 40 mM, preferably about 19 to 20 mM, and a temperature of about 50 to 70, preferably about 60 to 60. The condition of 5 t is shown. Particularly, the case where the sodium concentration is about 19 mM and the temperature is about 65 is most preferable.
「オリゴヌクレオチド」 としては、 2—デォキシ一 D—リポースを含有してい るポリデォキシヌクレオチド、 D—リポースを含有しているポリデォキシヌクレ ォチド、 プリンまたはピリミジン塩基の N—グリコシドであるその他のタイプの オリゴヌクレオチド、 あるいは非ヌクレオチド骨格を有するその他のポリマ一 (例えば、 市販の蛋白質核酸および合成配列特異的な核酸ポリマー) または特殊 な結合を含有するその他のポリマー (伹し、 該ポリマーは D N Aや R NA中に見 出されるような塩基のペアリングゃ塩基の付着を許容する配置をもつヌクレオチ ドを含有する) などが挙げられる。 これらは、 二本鎖 D NA、 一本鎖 D NA、 二 本鎖 R NA、 一本鎖 R NA、 さらに D N A: R NAハイブリッドであることがで き、 さらに非修飾オリゴヌクレオチド (または非修飾オリゴヌクレオチド) 、 さ らには公知の修飾の付加されたもの、 例えば当該分野で知られた標識化されたも 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、 ホスホルアミデ一 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、 ホスホロジチォェ一トなど) を持つもの、 例えば蛋 白質 (ヌクレアーゼ、 ヌクレア一ゼ ·インヒビター、 トキシン、 抗体、 シグナル ペプチド、 ポリ一 L _リジンなど) や糖 (例えば、 モノサッカライドなど) など の側鎖基を有しているもの、 インターカレント化合物 (例えば、 ァクリジン、 プ ソラレンなど) を持つもの、 キレート化合物 (例えば、 金属、 放射活性をもつ金 属、 ホウ素、 酸化性の金属など) を含有するもの、 アルキル化剤を含有するもの、 修飾された結合を持つもの (例えば、 α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、 あるいはエーテル、 ァミンなどの官能基 に変換されていてよい。 また、 本発明のオリゴヌクレオチドは、 修飾された核酸 (R NA、 D NA) であってもよい。 修飾された核酸の具体例としては、 核酸の 硫黄誘導体、 チォホスフェート誘導体、 あるいはポリヌクレオシドアミドまたは オリゴヌクレオシドアミドの分解に抵抗性のものなどが挙げられる。 "Oligonucleotides" are polydeoxynucleotides containing 2-dexoxy-D-reports, polydeoxynucleotides containing D-reports, N-glycosides of purine or pyrimidine bases. Other types of oligonucleotides or other polymers with non-nucleotide backbones (eg, commercially available protein nucleic acids and synthetic sequence-specific nucleic acid polymers) or specialty Other polymers containing a suitable bond (these polymers include base pairing as found in DNA and RNA, and contain a nucleotide having a configuration permitting base attachment). . These can be double-stranded DNA, single-stranded DNA, double-stranded RNA, single-stranded RNA, and even DNA: RNA hybrids, and can also be unmodified oligonucleotides (or unmodified oligonucleotides). Nucleotides), as well as those with known modifications, such as labeled, capped, methylated, one or more natural nucleotides, as known in the art. Is substituted with an analog, modified with an intramolecular nucleotide, for example, has an uncharged bond (eg, methylphosphonate, phosphotriester, phosphoramidite, thiolbamate, etc.), a charged bond or sulfur Those having contained bonds (for example, phosphorothioate, phosphorodithioate, etc.), for example, proteins (nucleases, nucleases / inhibitors) , Toxins, antibodies, signal peptides, poly-L-lysine, etc.) or sugars (for example, monosaccharides), etc., or those having an intermolecular compound (for example, acridine, psoralen, etc.) , Compounds containing chelating compounds (eg, metals, radioactive metals, boron, oxidizable metals, etc.), those containing alkylating agents, those with modified bonds (eg, α-anomers) Type nucleic acid). Here, “nucleoside”, “nucleotide” and “nucleic acid” may include not only those containing purine and pyrimidine bases but also those having other modified heterocyclic bases. Such modifications may include methylated purines and pyrimidines, acylated purines and pyrimidines, or other heterocycles. Modified nucleotides and modified nucleotides may also be modified at the sugar moiety, e.g., where one or more hydroxyl groups have been replaced with halogens, aliphatic groups, etc., or functional groups such as ethers, amines, etc. It may be converted to a base. The oligonucleotide of the present invention may be a modified nucleic acid (RNA, DNA). Specific examples of the modified nucleic acid include a sulfur derivative, a thiophosphate derivative, and a nucleic acid that is resistant to degradation of polynucleoside amide or oligonucleoside amide.
本発明のオリゴヌクレオチドの具体例として、 配列番号: 1 3で表される塩基 配列を含有する D NA、 配列番号: 1 4で表される塩基配列を含有する D N Aな どが用いられる。 As a specific example of the oligonucleotide of the present invention, a base represented by SEQ ID NO: 13 DNA containing a sequence, DNA containing the base sequence represented by SEQ ID NO: 14 and the like are used.
本発明のオリゴヌクレオチドは、 公知の方法、 例えば、 固相または液相合成法 (例、 自動化核酸合成器、 液相技術等) に従って製造する。 あるいは、 公知の遺 伝子工学技術 (例、 逆転写酵素、 P C R合成などの使用等) に従って製造しても よい。  The oligonucleotide of the present invention is produced according to a known method, for example, a solid phase or liquid phase synthesis method (eg, an automated nucleic acid synthesizer, liquid phase technology, etc.). Alternatively, it may be produced according to known gene engineering techniques (eg, use of reverse transcriptase, PCR synthesis, etc.).
配列番号: 1 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、 例えば、 配列番号: 1 1で表されるアミノ酸配列と約 5 5 %以上、 好ましく は約 6 0 %以上、 より好ましくは約 7 0 %以上、 さらに好ましくは約 8 0 %以上、 なかでも好ましくは約 9 0 %以上、 最も好ましくは約 9 5 %以上の相同性を有す るァミノ酸配列などが挙げられる。  Examples of the amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 11 include, for example, about 55% or more, and preferably about 60% or more of the amino acid sequence represented by SEQ ID NO: 11, More preferably, an amino acid sequence having a homology of about 70% or more, more preferably about 80% or more, particularly preferably about 90% or more, and most preferably about 95% or more. .
配列番号: 1 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 する蛋白質としては、 例えば、 配列番号: 1 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有し、 配列番号: 1 1で表されるアミノ酸配列と実質的 に同質の活性を有する蛋白質などが好ましい。  Examples of a protein having an amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 11 include, for example, a protein having an amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 11 A protein having substantially the same activity as the amino acid sequence represented by SEQ ID NO: 11 is preferred.
実質的に同質の活性としては、 例えば、 リガンド結合活性、 シグナル情報伝達 作用、 細胞死抑制活性、 細胞接着活性および細胞運動能制御活性などが挙げられ る。 実質的に同質とは、 それらの活性が性質的に同質であることを示す。 したが つて、 リガンド結合活性、 シグナル情報伝達作用、 細胞死抑制活性、 細胞接着活 性および細胞運動能制御活性などの活性が同等 (例、 約 0 . 0 1〜1 0 0倍、 好 ましくは約 0 . 5〜2 0倍、 より好ましくは約 0 . 5〜2倍) であることが好ま しいが、 これらの活性の程度や蛋白質の分子量などの量的要素は異なっていても よい。  Substantially the same activity includes, for example, ligand binding activity, signal transduction activity, cell death inhibitory activity, cell adhesion activity, and cell motility regulating activity. Substantially the same indicates that their activities are the same in nature. Therefore, activities such as ligand binding activity, signal transduction activity, cell death inhibitory activity, cell adhesion activity and cell motility control activity are equivalent (eg, about 0.01 to 100 times, preferably Is preferably about 0.5 to 20 times, more preferably about 0.5 to 2 times), but the quantitative factors such as the degree of activity and the molecular weight of the protein may be different.
リガンド結合活性、 シグナル情報伝達作用、 細胞死抑制活性、 細胞接着活性お よび細胞運動能制御活性などの測定は、 公知の方法に準じて行うことができる。 本明細書における受容体蛋白質のアミノ酸配列は、 ぺプチド表記の慣例に従つ て、 左端が N末端 (ァミノ末端) 、 右端が C末端 (力ルポキシル末端) である。 本発明の受容体蛋白質は、 C末端が力ルポキシル基 (-C00H) 、 カルポキシレー ト (-C00—) 、 アミド (- C0NH2) またはエステル (- C00R) の何れであってもよい。 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、 イソプロピルもしくは n—ブチルなどの C 1H5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 C3_8シクロアルキル基、 例えば、 フエニル、 ひ一ナフ チルなどの (36_12ァリール基、 例えば、 ベンジル、 フエネチルなどのフエ二ルー C ト 2アルキル基もしくは α—ナフチルメチルなどの α—ナフチル— C ,_2アルキル基 などの C 7_14ァラルキル基のほか、 経口用エステルとして汎用されるビバ口ィルォ キシメチル基などが用いられる。 Measurement of ligand binding activity, signal transduction activity, cell death inhibitory activity, cell adhesion activity, cell motility control activity, and the like can be performed according to known methods. In the present specification, the amino acid sequence of the receptor protein is the N-terminus (amino terminus) at the left end and the C-terminus (aminopropyl end) at the right end, according to the convention of peptide notation. Receptor protein of the present invention, C-terminal, the force Rupokishiru group (-C00H), Karupokishire bets (-C00-), amide (- C0NH 2) or ester (- C00R) may be either. Here, as R in the ester, e.g., methyl, Echiru, n- propyl, C 1H5 alkyl group such as isopropyl or n- butyl, cyclopentyl Le, C 3 _ 8 cycloalkyl group such as cyclohexyl, for example, phenyl, (3 6 _ 12 Ariru groups, such as single-naphthyl shed, for example, benzyl, phenylene route C DOO 2 alkyl or α- naphthylmethyl etc. α- naphthyl such as phenethyl - C, such as _ 2 alkyl groups in addition to C 7 _ 14 Ararukiru group, Viva port Iruo Kishimechiru group commonly used as an oral ester.
本発明の受容体蛋白質が C末端以外に力ルポキシル基 (またはカルボキシレー ト) を有している場合、 力ルポキシル基がアミド化またはエステル化されている ものも本発明の受容体蛋白質に含まれる。 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 receptor protein of the present invention has a lipoxyl group (or carboxylate) other than the C-terminus, the receptor protein of the present invention also includes those in which the lipoxyl group is amidated or esterified. . As the ester in this case, for example, the above-mentioned C-terminal ester and the like are used.
さらに、 本発明の受容体蛋白質には、 上記した蛋白質において、 N末端のメチ ォニン残基のァミノ基が保護基 (例えば、 ホルミル基、 ァセチルなどの C26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、 N端側が生体内で 切断され生成したダル夕ミル基がピログルタミン酸化したもの、 分子内のアミノ 酸の側鎖上の置換基 (例、 -0H、 -SH、 アミノ基、 イミダゾール基、 インドール基、 グァニジノ基など) が適当な保護基 (例えば、 ホルミル基、 ァセチルなどの C26 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蛋白質などの複合蛋白質なども含まれる。 Furthermore, the receptor protein of the present invention is the protein mentioned above, Amino group protecting groups methylate Onin residues of N-terminal (e.g., formyl group, C 2 such Asechiru - such Ashiru group such as 6 Al Kanoiru group ), The N-terminal side is cleaved in vivo, and the daryumil group formed is pyroglutamine-oxidized, the substituent on the side chain of the amino acid in the molecule (eg, -0H, -SH, amino group, imidazole group, indole group, Guanijino group, etc.) a suitable protecting group (e.g., formyl group, C 2 such Asechiru - those protected by 6 etc. Ashiru groups such Arukanoiru group), a sugar chain Complex proteins such as bound so-called glycoproteins are also included.
本発明の受容体蛋白質の具体例としては、 例えば、 配列番号: 1 1で表される アミノ酸配列を含有する受容体蛋白質などが用いられる。  Specific examples of the receptor protein of the present invention include, for example, a receptor protein containing the amino acid sequence represented by SEQ ID NO: 11 and the like.
また本発明の受容体蛋白質の部分ペプチド (以下、 本発明の部分ペプチドと略 記する場合がある) を用いることもできる。 本発明の部分ペプチドとしては、 上 記した本発明の受容体蛋白質の部分ペプチドであれば何れのものであってもよい が、 例えば、 本発明の受容体蛋白質分子のうち、 細胞膜の外に露出している部分 や細胞膜の内側に位置する部分であって、 実質的に同質の活性を有するものなど が用いられる。  Further, a partial peptide of the receptor protein of the present invention (hereinafter, sometimes abbreviated as the partial peptide of the present invention) can also be used. The partial peptide of the present invention may be any peptide as long as it is a partial peptide of the above-described receptor protein of the present invention. For example, of the receptor protein molecules of the present invention, For example, a portion having a substantially identical activity or a portion located inside the cell membrane is used.
ここで、 「実質的に同質の活性」 とは、 例えばリガンド結合活性、 シグナル情 報伝達作用、 細胞死抑制活性、 細胞接着活性および細胞運動能制御活性などを示 す。 リガンド結合活性、 シグナル情報伝達作用、 細胞死抑制活性、 細胞接着活性 および細胞運動能制御活性などの測定は、 公知の方法に準じて行うことができる c 本発明の部分べプチドのアミノ酸数は、 上記した本発明の受容体蛋白質の構成 アミノ酸配列のうち少なくとも 5個以上、 好ましくは 1 0個以上、 好ましくは 5 0個以上、 より好ましくは 1 0 0個以上のアミノ酸配列を有するペプチドなどが 好ましい。 実質的に同一のアミノ酸配列とは、 これらアミノ酸配列と約 5 0 % 以上、 好ましくは約 6 0 %以上、 より好ましくは約 7 0 %以上、 さらに好ましく は約 8 0 %以上、 なかでも好ましくは約 9 0 %以上、 最も好ましくは約 9 5 %以 上の相同性を有するアミノ酸配列を示す。 Here, “substantially the same activity” refers to, for example, ligand binding activity, signal information transmission activity, cell death inhibitory activity, cell adhesion activity, and cell motility control activity. You. Ligand binding activity, signal transduction activity, cell death suppressing activity, measurements, such as cell adhesion activity and cell motility control activity, the amino acid number of the partial base peptide of the c the present invention can be carried out according to known methods, Peptides having an amino acid sequence of at least 5 or more, preferably 10 or more, preferably 50 or more, more preferably 100 or more of the amino acid sequences of the above-described receptor protein of the present invention are preferable. . A substantially identical amino acid sequence is at least about 50%, preferably at least about 60%, more preferably at least about 70%, still more preferably at least about 80%, and most preferably at least about 50% of these amino acid sequences. An amino acid sequence having about 90% or more, most preferably about 95% or more homology is shown.
また、 本発明の部分ペプチドは、 (i) 上記アミノ酸配列中の 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個 (1〜5個) ) のアミ ノ酸が欠失し、 (i i) 上記アミノ酸配列に 1または 2個以上 (好ましくは、 1〜 20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個 (1〜5 個) ) のアミノ酸が付加し、 または (i i i) 上記アミノ酸配列中の 1または 2個 以上 (好ましくは、 1〜1 0個程度、 より好ましくは数個、 さらに好ましくは 1 〜5個程度) のアミノ酸が他のアミノ酸で置換されていてもよい。 本発明の部分 ペプチドの具体例としては、 配列番号: 1 1で表されるアミノ酸配列の第 6 0番 目〜第 7 3番目または第 4 9 1番目〜第 5 0 4番目のアミノ酸配列を含有するべ プチドなどが挙げられる。  In addition, the partial peptide of the present invention comprises: (i) one or more (preferably about 1 to 10, more preferably several (1 to 5)) amino acids in the above amino acid sequence; (Ii) one or two or more (preferably about 1 to 20, more preferably about 1 to 10, and more preferably several (1 to 5)) (Iii) one or more (preferably about 1 to 10, more preferably several, more preferably about 1 to 5) amino acids in the above amino acid sequence It may be substituted with an amino acid. Specific examples of the partial peptide of the present invention include the 60th to 73rd or 491st to 504th amino acid sequences of the amino acid sequence represented by SEQ ID NO: 11. Examples of such peptides include:
また、 本発明の部分ペプチドは C末端がカルボキシル基 (- C00H) 、 カルポキ シレート (- C00-) 、 アミド (- C0NH2) またはエステル (-C00R) であってもよい (Rは前記と同意義を示す) 。 本発明の部分ペプチドが C末端以外にカルポキシ ル基 (またはカルボキシレート) 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の部分ペプチドに含まれる。 この場合 のエステルとしては、 例えば上記した C末端のエステルなどが用いられる。 In the partial peptide, the C-terminus carboxyl group of the present invention (- C00H), Karupoki Shireto (- C00-), amide (- C0NH 2) or esters may be a (-C00R) (R is as defined above Is shown). When the partial peptide of the present invention has a carboxyl group (or carboxylate) other than the C-terminus, those in which the carbonyl group is amidated or esterified are also included in the partial peptide of the present invention. As the ester in this case, for example, the above-mentioned C-terminal ester and the like are used.
さらに、 本発明の部分ペプチドには、 上記した本発明の受容体蛋白質と同様に、 N末端の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ダルタミル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 above-described receptor protein of the present invention, the partial peptide of the present invention has a N-terminal methionine residue in which the amino group of the methionine residue is protected with a protecting group, and the N-terminal side is cleaved in vivo to produce Dartamyl group is pyroglutamine-oxidized, amino acid side chain in the molecule is protected by a suitable protecting group, or sugar chain Conjugated peptides, such as so-called glycopeptides, to which is bound.
本発明の受容体蛋白質またはその部分ペプチドの塩としては、 酸または塩基と の生理学的に許容される塩が挙げられ、 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。  Examples of the salt of the receptor protein or its partial peptide of the present invention include a physiologically acceptable salt with an acid or a base, 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) or organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, succinic acid) Acids, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid) and the like are used.
本発明の受容体蛋白質またはその塩は、 上記したヒトゃ哺乳動物の細胞または 組織から公知の受容体蛋白質の精製方法によって製造することもできるし、 後に 記載する本発明の受容体蛋白質をコードする D NAを含有する形質転換体を培養 することによつても製造することができる。 また、 後に記載する蛋白質合成法ま たはこれに準じて製造することもできる。  The receptor protein of the present invention or a salt thereof can be produced from the above-described human or mammalian cells or tissues by a known method for purifying a receptor protein, or encodes the receptor protein of the present invention described later. It can also be produced by culturing a transformant containing DNA. Also, the protein can be produced by the protein synthesis method described later or according to this method.
ヒトゃ哺乳動物の組織または細胞から製造する場合、 ヒトゃ哺乳動物の組織ま たは細胞をホモジナイズした後、 酸などで抽出を行ない、 該抽出液を逆相クロマ トグラフィ一、 イオン交換クロマトグラフィーなどのクロマトグラフィ一を組み 合わせることにより精製単離することができる。  When producing from human or mammalian tissues or cells, the human or mammalian tissues or cells are homogenized and then extracted with an acid or the like, and the resulting extract is subjected to reverse phase chromatography, ion exchange chromatography, etc. Purification and isolation can be carried out by combining the above chromatography methods.
本発明の受容体蛋白質もしくはその部分べプチドまたはその塩またはそのアミ ド体の合成には、 通常市販の蛋白質合成用樹脂を用いることができる。 そのよう な樹脂としては、 例えば、 クロロメチル樹脂、 ヒドロキシメチル樹脂、 ベンズヒ ドリルァミン樹脂、 アミノメチル樹脂、 4—ベンジルォキシベンジルアルコール 樹脂、 4—メチルベンズヒドリルァミン樹脂、 P AM樹脂、 4—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ーヒドロキシメチル) フエノキシ樹脂、 4一 ( 2 ' , 4 'ージメ卜キシフエ二ルー F m o cアミノエチル) フエノキシ樹脂などを挙げ ることができる。 このような樹脂を用い、 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、 目的とする蛋白質またはペプチドのアミノ酸配列通りに、 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蛋白質ま たはペプチドを切り出すと同時に各種保護基を除去し、 さらに高希釈溶液中で分 子内ジスルフィド結合形成反応を実施し、 目的の蛋白質もしくは部分ペプチドま たはそのアミド体を取得する。 For the synthesis of the receptor protein of the present invention, its partial peptide, its salt or its amide, a commercially available resin for protein synthesis can be usually used. Examples of such resins include chloromethyl resin, hydroxymethyl resin, benzhydrylamine resin, aminomethyl resin, 4-benzyloxybenzyl alcohol resin, 4-methylbenzhydrylamine resin, PAM resin, and 4-hydroxy resin. Methylmethylphenylacetamidomethyl resin, polyacrylamide resin, 4- (2 ', 4'dimethoxyphenylhydroxymethyl) phenoxy resin, 4- (2', 4'dimethyloxyphenyl Fmocaminoethyl) Phenoxy resins and the like can be mentioned. Using such a resin, amino acids having appropriately protected amino groups and side chain functional groups are condensed on the resin according to various known condensation methods in accordance with the amino acid sequence of the target protein or peptide. At the end of the reaction, a protein or peptide is cleaved from the resin, and at the same time, various protecting groups are removed.In addition, an intramolecular disulfide bond formation reaction is performed in a highly diluted solution to obtain a target protein or partial peptide. Or its amide form.
上記した保護アミノ酸の縮合に関しては、 蛋白質合成に使用できる各種活性化 試薬を用いることができるが、 特に、 カルポジイミド類がよい。 カルポジイミド 類としては、 D C C、 Ν, Ν'—ジイソプロピルカルポジイミド、 Ν—ェチル—Ν '一 (3—ジメチルァミノプロリル) カルポジイミドなどが用いられる。 これら による活性化にはラセミ化抑制添加剤 (例えば、 H O B t、 H O O B t ) ととも に保護アミノ酸を直接樹脂に添加するか、 または、 対称酸無水物または H O B t エステルあるいは H〇〇B tエステルとしてあらかじめ保護アミノ酸の活性化を 行なつた後に樹脂に添加することができる。  Regarding the condensation of the above protected amino acids, various activating reagents that can be used for protein synthesis can be used, and carbodiimides are particularly preferable. Examples of the carbopimides include DCC, Ν, Ν′-diisopropyl carbopimide, and ェ -ethyl-Ν ′-(3-dimethylaminoprolyl) carbopimide. Activation by these involves the addition of a protected amino acid directly to the resin along with a racemization inhibitor additive (eg, HOBt, HOOBt), or a symmetrical acid anhydride or HOBt ester or H〇〇Bt ester. Can be added to the resin after activation of the protected amino acid in advance.
保護アミノ酸の活性化や樹脂との縮合に用いられる溶媒としては、 蛋白質縮合 反応に使用しうることが知られている溶媒から適宜選択されうる。 例えば、 N, N—ジメチルホルムアミド, N,N—ジメチルァセトアミド, N—メチルピロリ ドンなどの酸アミド類、 塩化メチレン, クロ口ホルムなどのハロゲン化炭化水素 類、 トリフルォロエタノールなどのアルコール類、 ジメチルスルホキシドなどの スルホキシド類、 ピリジン, ジォキサン, テトラヒドロフランなどのエーテル類、 ァセトニトリル, 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 蛋白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 20°C〜50°Cの範囲から適宜選択される。 活性化されたアミノ酸誘導体は 通常 1. 5〜4倍過剰で用いられる。 ニンヒドリン反応を用いたテストの結果、 縮 合が不十分な場合には保護基の脱離を行うことなく縮合反応を繰り返すことによ り十分な縮合を行なうことができる。 反応を繰り返しても十分な縮合が得られな いときには、 無水酢酸またはァセチルイミダゾ一ルを用いて未反応アミノ酸をァ セチル化することができる。  The solvent used for activating the protected amino acid or for condensing with the resin 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, and N-methylpyrrolidone; halogenated hydrocarbons such as methylene chloride and chloroform; alcohols such as trifluoroethanol. And sulfoxides such as dimethylsulfoxide, ethers such as pyridine, dioxane and tetrahydrofuran, nitriles such as acetonitrile and propionitrile, esters such as methyl acetate and ethyl acetate, or an appropriate mixture thereof. The reaction temperature is appropriately selected from the range known to be usable for the protein bond formation reaction, and is usually selected from the range of about 20 ° 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, the unreacted amino acid can be acetylated using acetic anhydride or acetylimidazole.
原料のァミノ基の保護基としては、 例えば、 Z、 B o c、 夕一シャリ一ペンチ ルォキシカルポニル、 ィソポル二ルォキシカルポニル、 4ーメトキシベンジルォ キシカルポニル、 C 1— Z、 B r— Z、 ァダマンチルォキシカルボニル、 トリフ ルォロアセチル、 フタロイル、 ホルミル、 2—二トロフエニルスルフエ二ル、 ジ フエニルホスフィノチオイル、 Fm o cなどが用いられる。 力ルポキシル基は、 例えば、 アルキルエステル化 (例えば、 メチル、 エヂル、 プロピル、 ブチル、 夕一シャリーブチル、 シクロペンチル、 シクロへキシル、 シ クロへプチル、 シクロォクチル、 2—ァダマンチルなどの直鎖状、 分枝状もしく は環状アルキルエステル化) 、 ァラルキルエステル化 (例えば、 ベンジルエステ ル、 4—ニトロべンジルエステル、 4ーメトキシベンジルエステル、 4一クロ口 ベンジルエステル、 ベンズヒドリルエステル化) 、 フエナシルエステル化、 ベン ジルォキシカルポニルヒドラジド化、 夕一シャリーブトキシカルポニルヒドラジ ド化、 トリチルヒドラジド化などによつて保護することができる。 Examples of the protecting group for the amino group of the starting material include Z, Boc, succinyl-pentyloxycarbonyl, isoporiloxycarbonyl, 4-methoxybenzyloxycarbonyl, C1-Z, Br-Z, Adamantyloxycarbonyl, trifluoroacetyl, phthaloyl, formyl, 2-ditrophenylsulfenyl, diphenylphosphinothioyl, Fmoc and the like are used. The lipoxyl group may be, for example, an alkyl esterified (eg, methyl, ether, propyl, butyl, tert-butyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, 2-adamantyl, etc.) Or cyclic alkyl esterification), aralkyl esterification (eg, benzyl ester, 4-nitrobenzyl ester, 4-methoxybenzyl ester, 4-cyclobenzyl ester, benzhydryl esterification), phenacyl ester Can be protected by benzyloxycarbonyl hydrazide, short-lived butoxycarbonyl hydrazide, trityl hydrazide, etc.
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、 エトキシカルボニル基などの炭酸から誘導される基などが用いられる。 また、 エーテル化に適する基としては、 例えば、 ベンジル基、 テトラヒドロピラニル基、 t一プチゾレ基などである。  The hydroxyl group of serine can be protected, for example, by esterification or etherification. As a group suitable for the esterification, for example, a lower alkanol group such as an acetyl group, an aroyl group such as a benzoyl group, a group derived from carbonic acid such as a benzyloxycarbonyl group, an ethoxycarbonyl group and the like are used. In addition, groups suitable for etherification include, for example, a benzyl group, a tetrahydropyranyl group, and a t-peptizole group.
チロシンのフエノール性水酸基の保護基としては、 例えば、 B z l、 C 1 2- B z 1、 2一二トロベンジル、 B r— Z、 ターシャリーブチルなどが用いられる。 ヒスチジンのイミダゾ一ルの保護基としては、 例えば、 T o s、 4ーメトキシ — 2, 3 , 6—トリメチルベンゼンスルホニル、 D N P、 ベンジルォキシメチル、 B um、 B o c、 T r t、 F m o cなどが用いられる。 The protecting group of the phenolic hydroxyl group of tyrosine, for example, B zl, C 1 2 - B z 1, 2 twelve Torobenjiru, B r- Z, such as tertiary butyl is used. Examples of the protecting group for histidine imidazole include Tos, 4-methoxy-2,3,6-trimethylbenzenesulfonyl, DNP, benzyloxymethyl, Bum, Boc, Trt, and Fmoc. Can be
原料の力ルポキシル基の活性化されたものとしては、 例えば、 対応する酸無水 物、 アジド、 活性エステル 〔アルコール (例えば、 ペンタクロロフエノール、 2, 4, 5—トリクロ口フエノール、 2, 4ージニトロフエノール、 シァノメチルアル コール、 パラニトロフエノール、 H〇N B、 N—ヒドロキシスクシミド、 N—ヒ ドロキシフタルイミド、 HO B t ) とのエステル〕 などが用いられる。 原料のァ ミノ基の活性化されたものとしては、 例えば、 対応するリン酸アミドが用いられ る。  Activated carbonyl groups of the raw materials include, for example, corresponding acid anhydrides, azides, active esters [alcohols (eg, pentachlorophenol, 2,4,5-trichloromouth phenol, 2,4-dinitro Phenol, cyanomethyl alcohol, paranitrophenol, H〇NB, N-hydroxysuccinimide, N-hydroxyphthalimide, and esters with HOBt). 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、 P d—黒あるいは P d—炭素な どの触媒の存在下での水素気流中での接触還元や、 また、 無水フッ化水素、 メタ ンスルホン酸、 トリフルォロメタンスルホン酸、 トリフルォロ酢酸あるいはこれ らの混合液などによる酸処理や、 ジイ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Methods for removing (eliminating) the protecting group include, for example, catalytic reduction in a hydrogen stream in the presence of a catalyst such as Pd-black or Pd-carbon, or hydrogen fluoride anhydride or methanesulfonic acid. , Trifluoromethanesulfonic acid, trifluoroacetic acid or this Acid treatment with a mixed solution thereof, base treatment with diisopropylethylamine, triethylamine, piperidine, piperazine, and the like, and reduction with sodium in liquid ammonia are also used. The elimination reaction by the above acid treatment generally takes about one
20°C〜40°Cの温度で行なわれるが、 酸処理においては、 例えば、 ァニソ一ル、 フ エノール、 チオアニソール、 メタクレゾール、 パラクレゾール、 ジメチルスルフ イド、 1, 4一ブタンジチオール、 1 , 2—エタンジチオールなどのようなカチォ ン捕捉剤の添加が有効である。 また、 ヒスチジンのイミダゾ一ル保護基として用 いられる 2, 4—ジニトロフエニル基はチォフエノール処理により除去され、 ト リブトファンのィンドール保護基として用いられるホルミル基は上記の 1, 2— エタンジチオール、 1 , 4—ブタンジチオールなどの存在下の酸処理による脱保 護以外に、 希水酸化ナトリウム溶液、 希アンモニアなどによるアルカリ処理によ つても除去される。 The reaction is carried out at a temperature of 20 ° C to 40 ° C. In the acid treatment, for example, anisol, phenol, thioanisole, metacresol, paracresol, dimethylsulfide, 1,4-butanedithiol, 1,2 —Addition of cation scavenger such as ethanedithiol is effective. The 2,4-dinitrophenyl group used as the imidazole protecting group of histidine is removed by thiophenol treatment, and the formyl group used as the indole protecting group of tributofan is 1,2-ethanedithiol, 1,4 —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.
原料の反応に関与すべきでない官能基の保護ならびに保護基、 およびその保護 基の脱離、 反応に関与する官能基の活性化などは公知の基または公知の手段から 適宜選択しうる。  The protection of the functional group which should not be involved in the reaction of the raw materials, the 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.
蛋白質のアミド体を得る別の方法としては、 例えば、 まず、 カルボキシ末端ァ ミノ酸の α—力ルポキシル基をアミド化して保護した後、 アミノ基側にペプチド (蛋白質) 鎖を所望の鎖長まで延ばした後、 該ペプチド鎖の Ν末端の α—ァミノ 基の保護基のみを除いた蛋白質と C末端の力ルポキシル基の保護基のみを除去し た蛋白質とを製造し、 この両蛋白質を上記したような混合溶媒中で縮合させる。 縮合反応の詳細については上記と同様である。 縮合により得られた保護蛋白質を 精製した後、 上記方法によりすベての保護基を除去し、 所望の粗蛋白質を得るこ とができる。 この粗蛋白質は既知の各種精製手段を駆使して精製し、 主要画分を ' 凍結乾燥することで所望の蛋白質のアミド体を得ることができる。  As another method for obtaining an amide form of a protein, for example, first, after amidating and protecting the α-hydroxyl group of the carboxy-terminal amino acid, a peptide (protein) chain is added to the amino group side to a desired chain length. After the elongation, a protein was prepared by removing only the protecting group of the α-amino group at the Ν-terminal of the peptide chain and a protein was obtained by removing only the protecting group of the carboxy group at the C-terminus. In such a mixed solvent. Details of the condensation reaction are the same as described above. After purifying the protected protein obtained by the condensation, all the protecting groups are removed by the above-mentioned method, and a desired crude protein can be obtained. This crude protein is purified by various known purification means, and the main fraction is freeze-dried to obtain an amide of the desired protein.
蛋白質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一カル ポキシル基を所望のアルコール類と縮合しアミノ酸エステルとした後、 蛋白質の アミド体と同様にして、 所望の蛋白質のエステル体を得ることができる。  In order to obtain a protein ester, for example, after condensing the carboxyl group of the carboxy terminal amino acid with a desired alcohol to form an amino acid ester, the ester of the desired protein is converted in the same manner as the amide of the protein. Obtainable.
本発明の受容体蛋白質の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、 あるいは本発明の受容体蛋白質を適当なぺプチダーゼで切断するこ とによって製造することができる。 ペプチドの合成法としては、 例えば、 固相合 成法、 液相合成法のいずれによっても良い。 すなわち、 本発明の受容体蛋白質を 構成し得る部分ペプチドもしくはアミノ酸と残余部分とを縮合させ、 生成物が保 護基を有する場合は保護基を脱離することにより目的のぺプチドを製造すること ができる。 公知の縮合方法や保護基の脱離としては、 例えば、 以下の (i) 〜 The partial peptide of the receptor protein of the present invention or a salt thereof can be obtained by cleaving the receptor protein of the present invention with an appropriate peptidase according to a known peptide synthesis method. And can be manufactured by As a peptide synthesis method, for example, any of a solid phase synthesis method and a liquid phase synthesis method may be used. That is, the objective peptide is produced by condensing a partial peptide or amino acid capable of constituting the receptor protein of the present invention with the remaining portion, and if the product has a protecting group, removing the protecting group to produce the desired peptide. Can be. Known condensation methods and elimination of protecting groups include, for example, the following (i) to
(V) に記載された方法が挙げられる。  (V).
(i) . Bodanszkyおよび M.A. Ondetti、 ペプチド シンセシス (Peptide Synthesis; , Interscience Publishers, New York (1966年)  (i). Bodanszky and M.A. Ondetti, Peptide Synthesis;, Interscience Publishers, New York (1966)
(ii) Schroederおよび Luebke、 ザペプチド(The Peptide), Academic Press, New York (1965年)  (ii) Schroeder and Luebke, The Peptide, Academic Press, New York (1965)
(iii) 泉屋信夫他、 ペプチド合成の基礎と実験、 丸善 (株) (1975年)  (iii) Nobuo Izumiya et al., Fundamentals and experiments of peptide synthesis, Maruzen Co., Ltd. (1975)
(iv) 矢島治明 および榊原俊平、 生化学実験講座 1、 蛋白質の化学 IV、 205、 (1977年)  (iv) Haruaki Yajima and Shunpei Sakakibara, Laboratory of Biochemistry 1, Protein Chemistry IV, 205, (1977)
(V) 矢島治明監修、 続医薬品の開発第 14巻ペプチド合成広川書店  (V) Supervised by Haruaki Yajima, Continuing Drug Development
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 フィ一'液体クロマトグラフィー '再結晶などを組み合わせて本発明の部分ぺプ チドを精製単離することができる。 上記方法で得られる部分べプチドが遊離体で ある場合は、 公知の方法によって適当な塩に変換することができるし、 逆に塩で 得られた場合は、 公知の方法によって遊離体に変換することができる。  After the reaction, the partial peptide of the present invention can be purified and isolated by a combination of ordinary purification methods, for example, solvent extraction, distillation, column chromatography, 'liquid chromatography', and recrystallization. When the partial peptide obtained by the above method is a free form, it can be converted to an appropriate salt by a known method, and conversely, when it is obtained as a salt, it is converted to a free form by a known method. be able to.
本発明の受容体蛋白質をコードするポリヌクレオチドとしては、 上記した本発 明の受容体蛋白質をコードする塩基配列 (DNAまたは RNA、 好ましくは DN A) を含有するものであればいかなるものであってもよい。 該ポリヌクレオチド としては、 本発明の受容体蛋白質をコードする 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: RN Aのハイプリッドでもよい。 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 receptor protein of the present invention is not particularly limited as long as it contains the above-described nucleotide sequence (DNA or RNA, preferably DNA) encoding the receptor protein of the present invention. Is also good. The polynucleotide is RNA such as DNA or mRNA encoding the receptor protein of the present invention, and may be double-stranded or single-stranded. In the case of double-stranded, it may be double-stranded DNA, double-stranded RNA or DNA: RNA hybrid. In the case of a single strand, it may be a sense strand (ie, a coding strand) or an antisense strand (ie, a non-coding strand).
本発明の受容体蛋白質をコードする DNAとしては、 ゲノム DNA、 ゲノム D NAライブラリー、 上記した細胞'組織由来の cDNA、 上記した細胞'組織由 来の c D N Aライブラリ一、 合成 D N Aのいずれでもよい。 ライブラリ一に使用 するベクターは、 バクテリオファージ、 プラスミド、 コスミド、 ファージミドな どいずれであってもよい。 また、 上記した細胞'組織より全 R NAまたは mR N A画分を調製したものを用いて直接 Reverse Transcr iptase Polymerase Chain Reac t ion (以下、 RT- PCR法と略称する) によって D N Aを増幅することもできる。 具体的には、 本発明の受容体蛋白質をコードする D NAとしては、 例えば、 配 列番号: 1 2で表される塩基配列を含有する D NA、 または配列番号: 1 2で表 される塩基配列を含有する D N 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 D NAを有し、 本発明の受容体蛋白質と実質的に同質の活性 (例えば、 リガンド結合活性、 シグナル情報伝達作用、 細胞死抑制活性、 細胞接着活性およ び細胞運動能制御活性など) を有する受容体蛋白質をコ一ドする D NAなどであ れば何れのものでもよい。 Examples of the DNA encoding the receptor protein of the present invention include genomic DNA, genomic DNA library, cDNA derived from the above-described cells and tissues, and cells derived from the above-described cells. It may be either a conventional cDNA library or synthetic DNA. The vector used for the library may be any of bacteriophage, plasmid, cosmid, phagemid and the like. Alternatively, DNA may be directly amplified by reverse transcriptase polymerase chain reaction (hereinafter abbreviated as RT-PCR) using a preparation of all RNA or mRNA fractions from the above-mentioned cells' tissues. it can. Specifically, as the DNA encoding the receptor protein of the present invention, for example, a DNA containing the nucleotide sequence represented by SEQ ID NO: 12 or the nucleotide represented by SEQ ID NO: 12 Has DNA that hybridizes under high stringent conditions to DNA containing the sequence, and has substantially the same activity as the receptor protein of the present invention (eg, ligand binding activity, signal transduction activity, cell death) Any DNA may be used as long as it encodes a receptor protein having inhibitory activity, cell adhesion activity, and cell motility regulating activity.
配列番号: 1 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ィズする D NAとしては、 例えば、 配列番号: 1 2で表され る塩基配列と約 5 5 %以上、 好ましくは約 6 0 %以上、 好ましくは約 7 0 %以上、 好ましくは約 8 0 %以上、 より好ましくは約 9 0 %以上、 さらに好ましくは約 9 5 %以上の相同性を有する塩基配列を含有する D N Aなどが用いられる。  Examples of the DNA which hybridizes with the DNA having the nucleotide sequence represented by SEQ ID NO: 12 under high stringent conditions include, for example, about 55% or more of the nucleotide sequence represented by SEQ ID NO: 12; Preferably containing a base sequence having a homology of about 60% or more, preferably about 70% or more, preferably about 80% or more, more preferably about 90% or more, and still more preferably about 95% or more. DNA or the like is used.
ハイブリダィゼ一シヨンは、 公知の方法あるいはそれに準じる方法、 例えば、 モレキュラー ·クロ一ニング (Molecular Cloning) 2 nd (J. Sambrook et al ., Cold Spring Harbor Lab. Press, 1989) に記載の方法などに従って、 また、 市販のライブラリーを使用する場合、 添付の使用説明書に記載の方法に従って行 なうことができる。 より好ましくは、 ハイストリンジェントな条件下でハイプリ ダイゼ一シ 3ンを行なうことができる。  Hybridization can be performed by a known method or a method analogous thereto, for example, according to the method described in Molecular Cloning 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). When a commercially available library is used, it can be carried out according to the method described in the attached instruction manual. More preferably, hybridization can be performed under high stringency conditions.
該ハイストリンジェントな条件とは、 例えば、 ナトリウム濃度が約 1 9〜4 0 mM、 好ましくは約 1 9〜 2 0 mMで、 温度が約 5 0〜 7 0 °C、 好ましくは約 6 0〜6 5 T: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。  The high stringency conditions include, for example, a sodium concentration of about 19 to 40 mM, preferably about 19 to 20 mM, and a temperature of about 50 to 70 ° C, preferably about 60 to 70 ° C. The condition of 65 T: is shown. In particular, the case where the sodium concentration is about 19 mM and the temperature is about 65 ° C is most preferable.
本発明の部分べプチドをコードする D N Aとしては、 上記した本発明の部分べ プチドをコ一ドする塩基配列を含有するものであればいかなるものであってもよ レ^ また、 ゲノム DNA、 ゲノム DNAライブラリー、 上記した細胞 ·組織由来 の cDNA、 上記した細胞 ·組織由来の c DNAライブラリー、 合成 DNAのい ずれでもよい。 ライブラリ一に使用するベクターは、 バクテリオファージ、 ブラ 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、 上記した細 胞 ·組織より mRNA画分を調製したものを用いて直接 RT— PC R法によって DNAを増幅することもできる。 The DNA encoding the partial peptide of the present invention may be any DNA containing the above-described nucleotide sequence encoding the partial peptide of the present invention. Also, 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. The vector used for the library may be any of bacteriophage, plasmid, cosmid, phagemid and the like. DNA can also be directly amplified by the RT-PCR method using an mRNA fraction prepared from the cells and tissues described above.
具体的には、 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: 12で表される塩基配列を含有する DNAの部分塩基配列を有する DN A、 または配列番号: 12で表される DNAとハイストリンジエンドな条件下で ハイブリダィズする DNAを含有し、 本発明の受容体蛋白質と実質的に同質の活 性、 例えば、 リガンド結合活性、 シグナル情報伝達作用などを有する蛋白質をコ ードする DN Aの部分塩基配列を有する DN Aなどが用いられる。  Specifically, the DNA encoding the partial peptide of the present invention includes, for example, DNA having a partial nucleotide sequence of DNA containing the nucleotide sequence represented by SEQ ID NO: 12, or DNA represented by SEQ ID NO: 12. A protein containing DNA that hybridizes under high stringent conditions with the DNA to be obtained and having substantially the same activity as the receptor protein of the present invention, for example, ligand binding activity, signal transduction activity, etc. For example, a DNA having a partial nucleotide sequence of the DNA to be used is used.
本 明の受容体蛋白質またはその部分ペプチド (以下、 本発明の受容体蛋白質 と略記する場合がある) を完全にコードする DNAのクローニングの手段として は、 本発明の受容体蛋白質をコードする DNAの塩基配列の部分塩基配列を有す る合成 DNAプライマ一を用いて PC R法によって増幅するか、 または適当なベ クタ一に組み込んだ DN Aを本発明の受容体蛋白質の一部あるいは全領域をコー ドする D N A断片もしくは合成 D N Aを用いて標識したものとのハイブリダイゼ ーシヨンによって選別することができる。  As a means for cloning a DNA that completely encodes the receptor protein of the present invention or a partial peptide thereof (hereinafter, sometimes abbreviated as the receptor protein of the present invention), a DNA encoding the receptor protein of the present invention may be used. Amplification by the PCR method using a synthetic DNA primer having a partial nucleotide sequence of the nucleotide sequence, or DNA incorporating the DNA into a suitable vector is performed for partial or whole region of the receptor protein of the present invention. Selection can be performed by hybridization with a DNA fragment to be coded or labeled with a synthetic DNA.
DNAの塩基配列の置換は、 PCRや公知のキット、 例えば、 Mu tan™— super Express Km (宝酒造 (株) ) 、 Mutan™— K (宝酒造 (株) ) 等を用いて、 ODA— LA PCR法、 gapped duplex法、 Kunkel法等の公知の方法あるいはそれらに準じる方法 に従って行なうことができる。  The DNA base sequence can be replaced by ODA-LA PCR using PCR or a known kit, for example, Mutan ™ -super Express Km (Takara Shuzo Co., Ltd.), Mutan ™ -K (Takara Shuzo Co., Ltd.), etc. The method can be performed according to known methods such as the gapped duplex method and the Kunkel method, or a method analogous thereto.
クローン化された受容体蛋白質をコードする DNAは目的によりそのまま、 ま たは所望により制限酵素で消化したり、 リンカ一を付加したりして使用すること ができる。 該 DN A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。 本発明の受容体蛋白質をコードする DNAは、 例えば、 (ィ) 本発明の受容体 蛋白質をコードする DNAを含む、 例えば c DNAから目的とする DNA部分の 断片を切り出し、 (口) 該 DNA断片を適当な発現べクタ一中のプロモ一夕一の 下流に連結することにより発現ベクターへ組み込むことができる。 The DNA encoding the cloned receptor 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 at the 5 'end, and TAA, TGA or TAG as a translation stop codon at the 3' end. These translation initiation codon and translation termination codon can also be added using a suitable synthetic DNA adapter. The DNA encoding the receptor protein of the present invention includes, for example, (a) a DNA fragment encoding the receptor protein of the present invention; Can be incorporated into an expression vector by ligating the DNA downstream of the promoter in an appropriate expression vector.
クローン化のための、 あるいは発現のためのベクタ一としては、 大腸菌由来の プラスミド (例、 pCR4、 pCR2. K pBR322, pBR325、 pUC12、 pUC13) 、 枯草菌由来 のプラスミド (例、 pUB110、 pTP5、 pC194) 、 酵母由来プラスミド (例、 pSH19、 Examples of vectors for cloning or expression include plasmids derived from E. coli (eg, pCR4, pCR2.K pBR322, pBR325, pUC12, pUC13), and plasmids derived from Bacillus subtilis (eg, pUB110, pTP5, pC194) ), Yeast-derived plasmids (eg, pSH19,
PSH15) 、 λファ一ジなどのバクテリオファ一ジ、 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、 XTK pRc/CMV, pRc/RSV, pcDNAI/Neoなどが用いられる。 PSH15), bacteriophage such as λ phage, animal viruses such as retrovirus, vaccinia virus, baculovirus, etc., as well as pAl-11, XTK pRc / CMV, pRc / RSV, pcDNAI / Neo, etc. .
上記プロモーターとしては、 遺伝子の発現に用いる宿主に対応して適切なプロ モータ—であればいかなるものでもよい。 例えば、 動物細胞を宿主として用いる 場合は、 SRo;プロモータ一、 SV40プロモーター、 LTRプロモーター、 C MVプロモ一夕一、 HS V— TKプロモ一夕一などが挙げられる。 これらのうち. CMVプロモーター、 SRaプロモーターなどを用いるのが好ましい。  The promoter may be any promoter as long as it is appropriate for the host used for gene expression. For example, when an animal cell is used as a host, SRo; a promoter, an SV40 promoter, an LTR promoter, a CMV promoter, an HSV-TK promoter, and the like can be mentioned. Of these, it is preferable to use CMV promoter, SRa promoter and the like.
宿主がェシエリヒア属菌である場合は、 t r pプロモーター、 1 a cプロモー 夕一、 r e cAプロモ一夕一、 λΡ^プロモーター、 1 ρ ρプロモータ一などが、 宿主がバチルス属菌である場合は、 S Ρ〇 1プロモータ一、 S ΡΟ 2プロモータ 一、 p e η Ρプロモーターなど、 宿主が酵母である場合は、 ΡΗ05プロモータ 一、 PGKプロモー夕一、 GAPプロモータ一、 ADHプロモーターなどが好ま しい。 宿主が昆虫細胞である場合は、 ポリヘドリンプロモータ一、 P 10プロモ When the host is Escherichia, trp promoter, 1 ac promoter, recA promoter, λ プ ロ モ ー タ ー ^ promoter, 1 ρ promoter, etc., and when the host is Bacillus, S S When the host is yeast, such as 〇1 promoter, SΡΟ2 promoter, peηΡ promoter, etc., ΡΗ05 promoter, PGK promoter, GAP promoter, ADH promoter and the like are preferable. If the host is an insect cell, the polyhedrin promoter and the P10 promoter
—夕一などが好ましい。 —I prefer evening.
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、  In addition to the above, expression vectors include, if desired, an enhancer, a splicing signal, a polyA addition signal, a selection marker, and an SV40 replication origin (hereinafter, referred to as the
SV40ori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 c 選択マーカーとしては、 例えば、 ジヒドロ葉酸還元酵素 (以下、 dhfr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 ofと略称する場合がある、 G418耐性) 等が挙げられる。 特に、 CHO (dhfr) 細胞を用いて dhfr遺伝子を選択マーカ一として使用する場合、 チミ を含まない培地によっても目的遺伝子を選択できる。 Examples of the c- selectable marker that can be used include a gene for dihydrofolate reductase (hereinafter sometimes abbreviated as dhfr) gene [methotrexate (MTX) resistance ], An ampicillin resistance gene (hereinafter sometimes abbreviated as Amp 1 ), a neomycin resistance gene (hereinafter sometimes abbreviated as Ne of, G418 resistance), and the like. When the dhfr gene is used as a selection marker using (dhfr) cells, the target gene can be selected even with a thym-free medium.
また、 必要に応じて、 宿主に合ったシグナル配列を、 本発明の受容体蛋白質の N端末側に付加することができる。 宿主がェシエリヒア属菌である場合は、 Ph o Aシグナル配列、 〇mp Aシグナル配列などが、 宿主がバチルス属菌である場 合は、 α—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ひ ·シグナル配列、 SUC 2 ·シグナル配列など、 宿主が動物細胞である場合には、 インシュリン ·シグナル配列、 ひ一インターフ エロン ·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 can be added to the N-terminal side of the receptor protein of the present invention. If the host is a genus Escherichia, the Ph A signal sequence, the 〇mp A signal sequence, etc., if the host is a Bacillus genus, the α-amylase signal sequence, subtilisin signal sequence, etc. If the host is yeast, MF, signal sequence, SUC2, signal sequence, etc.If the host is an animal cell, insulin signal sequence, Hi-interferon, signal sequence, antibody molecule, signal sequence, etc. Are available respectively.
このようにして構築された本発明の受容体蛋白質をコードする DNA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 receptor protein of the present invention thus constructed, a transformant can be produced.
本発明に用いる宿主としては、 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、 動物細胞などがあげられる。  Examples of the host used in the present invention include Escherichia bacteria, Bacillus bacteria, yeast, insect cells, insects, animal cells, and the like.
ェシエリヒア属菌の具体例としては、 ェシエリヒア 'コリ (Escherichia coli) K12 - DH1 [Proc. Natl. Acad. Sci. USA, 60巻, 160 (1968)〕 , JM103 Specific examples of the bacterium belonging to the genus Escherichia include Escherichia coli K12-DH1 [Proc. Natl. Acad. Sci. USA, 60, 160 (1968)], JM103.
CNucleic Acids Research, 9巻, 309 (1981)] , JA221 [Journal of Molecular Biology, 120巻, 517 (1978)] , HB101 [Journal of Molecular Biology, 41巻, 459 (1969)〕 , C600 [Genetics, 39卷, 440 (1954)] , DH5 a [Gene, 96, 23-28 (1990)〕 , DHIOB [Proc. Natl. Acad. Sci. USA, 87巻, 645-4649 (1990)] な どが用いられる。 CNucleic Acids Research, 9, 309 (1981)], JA221 [Journal of Molecular Biology, 120, 517 (1978)], HB101 [Journal of Molecular Biology, 41, 459 (1969)], C600 [Genetics, 39 Vol., 440 (1954)], DH5a [Gene, 96, 23-28 (1990)], DHIOB [Proc. Natl. Acad. Sci. USA, 87, 645-4649 (1990)], etc. .
バチルス属菌としては、 例えば、 バチルス ·ズブチルス (Bacillus  Bacillus bacteria include, for example, Bacillus subtilis (Bacillus
subtilis) MI114 [Gene, 24巻, 255 (1983)] , 207-21 [Journal of subtilis) MI114 [Gene, 24, 255 (1983)], 207-21 [Journal of
Biochemistry, 95卷, 87 (1984)) などが用いられる。 Biochemistry, 95, 87 (1984)).
酵母としては、 例えば、 サッカロマイセス セレピシェ (Saccharomyces cerevisiae) AH22、 AH22R―、 ONA87-11A, DKD-5D, 20B- 12、 シゾサッカロマイセ ス ボンべ (Schizosaccharomyces pombe) NCYC1913, NCYC2036 ピキア パス トリス (Pichia pastoris) などが用いられる。  Examples of yeast include, for example, Saccharomyces cerevisiae AH22, AH22R-, ONA87-11A, DKD-5D, 20B-12, Schizosaccharomyces pombe NCYC1913, NCYC2036 Pichia pastoris (Pichia pastoris) ) Is used.
昆虫細胞としては、 例えば、 ウィルスが AcNPVの場合は、 ョトウガの幼虫由来 株化細胞 (Spodoptera frugiperda cell; Si細胞) 、 Trichoplusia niの中腸由 来の MG1細胞、 Tr ichoplus ia niの卵由来の High Five™細胞、 Mames tra Insect cells include, for example, when the virus is AcNPV, a cell line derived from a larva of Spodoptera (Spodoptera frugiperda cell; Si cell), or the midgut of Trichoplusia ni Original MG1 cells, High Five ™ cells derived from Trichoplus ia ni eggs, Mames tra
brass icae由来の細胞または Es t igmena 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 mor i N; BmN細胞) などが用い られる。 該 Sf細胞としては、 例えば、 Sf9細胞 (ATCC CRL1711) 、 Sf21細胞 (以 上、 In Vivo, 13, 213-217 (1977) ) などが用いられる。 Cells derived from brass icae or cells derived from Estigmena acrea are used. When the virus is BmNPV, a cell line derived from silkworm (Bombyx mori N; BmN cell) is used. As the Sf cells, for example, Sf9 cells (ATCC CRL1711), Sf21 cells (hereinafter, In Vivo, 13, 213-217 (1977)) and the like are used.
昆虫としては、 例えば、 カイコの幼虫などが用いられる 〔Nature, 315巻, 592 (1985) ] 。  As insects, for example, silkworm larvae are used [Nature, vol. 315, 592 (1985)].
動物細胞としては、 例えば、 サル細胞 COS- 7、 Vero、 チャイニーズハムス夕一 細胞 CH0 (以下、 CH0細胞と略記) 、 dhfr遺伝子欠損チャイニーズハムス夕一細胞 CH0 (以下、 CHO (dhf r)細胞と略記) 、 マウス L細胞, マウス AtT - 20、 マウスミエ ローマ細胞、 ラット GH3、 ヒト FL細胞などが用いられる。  Examples of animal cells include monkey cells COS-7, Vero, Chinese Hams Yuichi cell CH0 (hereinafter abbreviated as CH0 cells), dhfr gene-deficient Chinese Hams Yuichi cells CH0 (hereinafter abbreviated as CHO (dhfr) cells). ), Mouse L cells, mouse AtT-20, mouse myeloma cells, rat GH3, human FL cells, and the like.
ェシエリヒア属菌を形質転換するには、 例えば、 Proc. Nat l . Acad. Sc i . USA, 69巻, 2110 (1972)や 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。  Transformation of a bacterium belonging to the genus Escherichia can be performed, for example, according to the method described in Proc. Natl. Acad. Sci. USA, 69, 2110 (1972) or Gene, 17, 107 (1982). it can.
バチルス属菌を形質転換するには、 例えば、 Molecular & General Genet ics, 168巻, 111 (1979)などに記載の方法に従って行なうことができる。  Transformation of Bacillus can be performed, for example, according to the method described in Molecular & General Genetics, Vol. 168, 111 (1979).
酵母を形質転換するには、 例えば、 Methods in Enzymology, 194巻, 182— 187 (1991) 、 Proc. Nat l . 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。  Transformation of yeast can be performed, for example, according to the method described in Methods in Enzymology, Vol. 194, 182-187 (1991), Proc. Natl. Acad. Sci. USA, Vol. 75, 1929 (1978). Can be.
昆虫細胞または昆虫を形質転換するには、 例えば、 Bio/Technology, 6, 47-55 (1988)などに記載の方法に従つて行なうことができる。  Insect cells or insects can be transformed, for example, according to the method described in Bio / Technology, 6, 47-55 (1988).
動物細胞を形質転換するには、 例えば、 細胞工学別冊 8 新細胞工学実験プロ トコ一ル. 263- 267 (1995) (秀潤社発行) 、 Virology, 52巻, 456 (1973)に記 載の方法に従って行なうことができる。  To transform animal cells, for example, see Cell Engineering Separate Volume 8 New Cell Engineering Experimental Protocol. 263-267 (1995) (published by Shujunsha), Virology, Vol. 52, 456 (1973). It can be performed according to the method.
このようにして、 E p h受容体蛋白質をコードする D NAを含有する発現べク タ一で形質転換された形質転換体が得られる。  Thus, a transformant transformed with the expression vector containing the DNA encoding the Eph receptor protein is 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、 コ一ンスチープ' リカー、 ペプトン、 力 ゼイン、 肉エキス、 大豆粕、 バレイショ抽出液などの無機または有機物質、 無機 物としては、 例えば、 塩化カルシウム、 リン酸二水素ナトリウム、 塩化マグネシ ゥムなどが挙げられる。 また、 酵母エキス、 ビタミン類、 生長促進因子などを添 加してもよい。 培地の pHは約 5〜 8が望ましい。 When culturing a transformant whose host is a bacterium belonging to the genus Escherichia or Bacillus, a liquid medium is suitable as a medium used for the cultivation, and a carbon source necessary for the growth of the transformant is contained therein. , Nitrogen sources, inorganic substances and others. As a carbon source, For example, glucose, dextrin, soluble starch, sucrose, etc.Nitrogen sources include, for example, inorganic or nitric acid salts such as ammonium salts, nitrates, corn steep liquor, peptone, power zein, meat extract, soybean meal, potato extract, etc. Examples of the 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 pH of the medium is preferably about 5-8.
ェシエリヒア属菌を培養する際の培地としては、 例えば、 グルコース、 カザミ ノ酸を含む M 9培地 (Journal of Experiments in Molecular Genetics, 431— 433, Cold Spring Harbor Laboratory, New York 1972) が好ましい。 ここに必 要によりプロモーターを効率よく働かせるために、 例えば、 3 —インドリルァ クリル酸のような薬剤を加えることができる。  As a medium for culturing the genus Escherichia, for example, an M9 medium containing glucose and casamino acids (Journal of Experiments in Molecular Genetics, 431-433, Cold Spring Harbor Laboratory, New York 1972) is preferable. If necessary, an agent such as 3-indolylacrylic acid can be added to make the promoter work efficiently.
宿主がェシエリヒア属菌の場合、 培養は通常約 15〜43°Cで約 3〜24時間行ない、 必要により、 通気や撹拌を加えることもできる。  When the host is a bacterium belonging to the genus Escherichia, the cultivation is usually performed at about 15 to 43 ° C for about 3 to 24 hours, and if necessary, aeration and stirring may be added.
宿主がバチルス属菌の場合、 培養は通常約 30〜4(TCで約 6〜24時間行ない、 必 要により通気や撹拌を加えることもできる。  When the host is a bacterium belonging to the genus Bacillus, the cultivation is usually performed for about 30 to 4 (TC for about 6 to 24 hours, and if necessary, aeration and stirring may be applied.
宿主が酵母である形質転換体を培養する際、 培地としては、 例えば、 バ一クホ 一ルダー (Burkholder) 最小培地 [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 CProc. Natl. Acad. Sci. USA, 81巻, 5330 (1984) 」 〕 が挙げられる。 培地の pHは約 5〜8に調整するのが好まし い。 培養は通常約 20°C〜35 で約 24〜72時間行ない、 必要に応じて通気や撹拌を 加える。  When culturing a transformant in which the host is yeast, the culture medium may be, for example, a Burkholder minimum medium [Proc. Natl. Acad. Sci. USA, 77, 4505 (1980)] or 0.5 Natl. Acad. Sci. USA, 81, 5330 (1984)]]. The pH of the medium is preferably adjusted to about 5-8. Cultivation is usually carried out at about 20 ° C to 35 for about 24 to 72 hours, with aeration and agitation as necessary.
宿主が昆虫細胞または昆虫である形質転換体を培養する際、 培地としては、 Grace's Insect Medium (Nature, 195, 788 (1962)) に非動化した 10%ゥシ血清 等の添加物を適宜加えたものなどが用いられる。 培地の pHは約 6.2〜 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、 必要に応じて通気ゃ撹 拌を加える。  When culturing an insect cell or a transformant in which the host is an insect, a medium such as Grace's Insect Medium (Nature, 195, 788 (1962)) is supplemented as appropriate with an additive such as immobilized 10% serum. Are used. Preferably, the pH of the medium is adjusted to about 6.2-4. Culture is usually performed at about 27 ° C. for about 3 to 5 days, and if necessary, aeration and agitation are added.
宿主が動物細胞である形質転換体を培養する際、 培地としては、 例えば、 約 5 〜20%の胎児牛血清を含む MEM培地 [Science, 122巻, 501 (1952)) , DMEM培 地 [Virology, 8卷, 396 (1959)〕 , RPMI1640培地 [The Journal of the Amer i can Med i cal Assoc i at i on, 199卷, 519 (1967)〕 , 199培地 [Proceed i ng o f the Soc i e ty f or the Bio l ogi cal Med i c i ne, 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°C〜4(TC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 whose animal host is an animal cell, for example, a MEM medium containing about 5 to 20% fetal bovine serum [Science, 122, 501 (1952)], a DMEM medium [Virology , 8 vol., 396 (1959)], RPMI1640 medium [The Journal of the Amer i can Med i cal Assoc i ation, Volume 199, 519 (1967)], 199 medium [Proceeding of the Socie ty for or the Biologi cal Med ici ne, Volume 73, 1 (1950) ] Etc. are used. Preferably, the pH is about 6-8. Culture is usually performed at about 30 ° C to 4 (TC for about 15 to 60 hours, and aeration and agitation are added as necessary.
以上のようにして、 形質転換体の細胞内、 細胞膜または細胞外に本発明の受容 体蛋白質を生成することができる。  As described above, the receptor protein of the present invention can be produced in the cells, in the cell membrane, or outside the cells of the transformant.
上記培養物から本発明の受容体蛋白質の分離精製は、 例えば、 下記の方法によ り行なうことができる。  Separation and purification of the receptor protein of the present invention from the above culture can be performed, for example, by the following method.
本発明の受容体蛋白質を培養菌体あるいは細胞から抽出するには、 培養後、 公 知の方法で菌体あるいは細胞を集め、 これを適当な緩衝液に懸濁し、 超音波、 リ ゾチームおよび/または凍結融解などによつて菌体ぁるいは細胞を破壊したのち、 遠心分離やろ過により受容体蛋白質の粗抽出液を得る方法などが適宜用いられる。 緩衝液の中に尿素や塩酸グァニジンなどの蛋白質変性剤や、 トリトン X- 100™など の界面活性剤が含まれていてもよい。 培養液中に受容体蛋白質が分泌される場合 には、 培養終了後、 公知の方法で該受容体蛋白質を含む培養上清を集める。  In order to extract the receptor protein of the present invention from cultured cells or cells, after culturing, the cells or cells are collected by a known method, suspended in an appropriate buffer, and subjected to ultrasonication, lysozyme and / or lysozyme. Alternatively, a method of destroying bacterial cells or cells by freeze-thawing or the like, and then obtaining a crude extract of the receptor protein 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 receptor protein is secreted into the culture solution, after the culture is completed, a culture supernatant containing the receptor protein is collected by a known method.
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受容体蛋白質 の精製は、 公知の分離 ·精製法を適切に組み合わせて行なうことができる。 これ らの公知の分離、 精製法としては、 塩析ゃ溶媒沈澱法などの溶解度を利用する方 法、 透析法、 限外ろ過法、 およびゲルろ過法などの主として分子量の差を利用す る方法、 イオン交換クロマトグラフィーなどの荷電の差を利用する方法、 ァフィ 二ティークロマトグラフィーなどの特異的親和性を利用する方法、 逆相高速液体 クロマトグラフィーなどの疎水性の差を利用する方法、 等電点電気泳動法などの 等電点の差を利用する方法などが用いられる。  Purification of the receptor protein contained in the thus obtained culture supernatant or extract can be carried out by appropriately combining known separation and purification methods. These known separation and purification methods include methods that utilize solubility such as salting out and solvent precipitation, and methods that mainly use the difference in molecular weight, such as dialysis, ultrafiltration, and gel filtration. A method that utilizes a difference in charge such as ion exchange chromatography; a method that utilizes a specific affinity such as affinity chromatography; a method that utilizes a difference in hydrophobicity such as reverse-phase high-performance liquid chromatography; A method utilizing the difference between isoelectric points such as point electrophoresis is used.
このようにして得られる受容体蛋白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 receptor 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 receptor protein is obtained as a salt, the known method is used. Alternatively, it can be converted to a free form or another salt by a method analogous thereto.
なお、 組換え体が産生する受容体蛋白質を、 精製前または精製後に適当な蛋白 修飾酵素を作用させることにより、 任意に修飾を加えたり、 ポリペプチドを部分 的に除去することもできる。 蛋白修飾酵素としては、 例えば、 トリプシン、 キモ トリプシン、 アルギニルェンドぺプチダーゼ、 プロテインキナーゼ、 グリコシダ ーゼなどが用いられる。 The receptor protein produced by the recombinant can be arbitrarily modified or partially modified by the action of an appropriate protein-modifying enzyme before or after purification. Can also be removed. 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受容体蛋白質またはその塩の活性は、 標識し たリガンドとの結合実験および特異抗体を用いたェンザィムィムノアッセィなど により測定することができる。  The activity of the receptor protein of the present invention or a salt thereof thus produced can be measured by a binding experiment with a labeled ligand, an enzymimnoassay using a specific antibody, or the like.
本発明の受容体蛋白質もしくはその部分ペプチドまたはその塩に対する抗体は、 本発明の受容体蛋白質もしくはその部分べプチドまたはその塩を認識し得る抗体 であれば、 ポリクローナル抗体、 モノクローナル抗体の何れであってもよい。  The antibody against the receptor protein or its partial peptide or its salt of the present invention may be any of a polyclonal antibody and a monoclonal antibody as long as it is an antibody that can recognize the receptor protein or its partial peptide or its salt of the present invention. Is also good.
本発明の受容体蛋白質もしくはその部分ペプチドまたはその塩 (以下、 本発明 の受容体蛋白質等と略記する場合がある) に対する抗体は、 本発明の受容体蛋白 質等を抗原として用い、 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 receptor protein of the present invention or a partial peptide thereof or a salt thereof (hereinafter, sometimes abbreviated as the receptor protein of the present invention) may be a known antibody using the receptor protein or the like of the present invention as an antigen. Alternatively, it can be produced according to a method for producing an antiserum.
〔モノクローナル抗体の作製〕  [Preparation of monoclonal antibody]
(a) モノクローナル抗体産生細胞の作製  (a) Preparation of monoclonal antibody-producing cells
本発明の受容体蛋白質等は、 哺乳動物に対して投与により抗体産生が可能な部 位にそれ自体あるいは担体、 希釈剤とともに投与される。 投与に際して抗体産生 能を高めるため、 完全フロイントアジュバントゃ不完全フロイントアジュバント を投与してもよい。 投与は通常 2〜 6週毎に 1回ずつ、 計 2〜1 0回程度行なわ れる。 用いられる哺乳動物としては、 例えば、 サル、 ゥサギ、 ィヌ、 モルモット、 マウス、 ラット、 ヒッジ、 ャギが挙げられるが、 マウスおよびラットが好ましく 用いられる。  The receptor protein or the like of the present invention is administered to a mammal at a site capable of producing an antibody by administration itself or together with a carrier or a diluent. Complete Freund's adjuvant / incomplete Freund's adjuvant may be administered in order to enhance antibody production upon administration. The administration is usually performed once every 2 to 6 weeks, for a total of about 2 to 10 times. Examples of mammals to be used include monkeys, rabbits, dogs, guinea pigs, mice, rats, sheep, and goats, 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、 抗原を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、 後記の標識化受容体蛋白質等と抗血 清とを反応させたのち、 抗体に結合した標識剤の活性を測定することにより行な うことができる。 融合操作は既知の方法、 例えば、 ケ一ラーとミルスタインの方 法 (Nature、 256巻、 495頁、 1975年) 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。 When producing monoclonal antibody-producing cells, a warm-blooded animal immunized with the antigen, for example, a mouse 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. By fusing the antibody-producing cells contained in the above with myeloma cells, a monoclonal antibody-producing hybridoma can be prepared. The antibody titer in the antiserum can be measured, for example, by reacting the labeled receptor protein or the like described below with antiserum, and then measuring the activity of the labeling agent bound to the antibody. . The fusion operation is performed in a known manner, for example, in the case of Keller and Milstein. Act (Nature, 256, 495, 1975). Examples of the fusion promoter include polyethylene glycol (PEG) and Sendai virus, and PEG is preferably used.
骨髄腫細胞としては、 例えば、 NS- 1、 P3UK SP2/0などが挙げられるが、 P3U1 が好ましく用いられる。 用いられる钪体産生細胞 (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、 PEG1000〜  Examples of myeloma cells include NS-1, P3UK SP2 / 0 and the like, and P3U1 is preferably used. The preferred ratio between the number of somatic cells (spleen cells) and the number of myeloma cells used is about 1: 1 to 20: 1, and PEG (preferably PEG1000 to
PEG6000) が 10〜80%程度の濃度で添加され、 約 20〜40° (:、 好ましくは約 30〜 37°Cで約 1〜10分間ィンキュベー卜することにより効率よく細胞融合を実施でき る。 PEG6000) is added at a concentration of about 10 to 80%, and the cell fusion can be carried out efficiently by incubating at about 20 to 40 ° (: preferably at about 30 to 37 ° C for about 1 to 10 minutes).
モノクローナル抗体産生ハイプリド一マのスクリーニングには種々の方法が使 用できるが、 例えば、 受容体蛋白質等の抗原を直接あるいは担体とともに吸着さ せた固相 (例、 マイクロプレート) にハイブリド一マ培養上清を添加し、 次に放 射性物質や酵素などで標識した抗免疫グロプリン抗体 (細胞融合に用いられる細 胞がマウスの場合、 抗マウス免疫グロブリン抗体が用いられる) またはプロティ ン Aを加え、 固相に結合したモノクローナル抗体を検出する方法、 抗免疫グロブ 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受容体蛋白質等を加え、 固相に結合したモ ノクローナル抗体を検出する方法などが挙げられる。  Various methods can be used to screen for monoclonal antibodies producing hybridomas. For example, hybridomas are cultured on a solid phase (eg, microplate) onto which an antigen such as a receptor protein is adsorbed directly or together with a carrier. Then, an anti-immunoglobulin antibody (anti-mouse immunoglobulin antibody is used if the cell used for cell fusion is a mouse) or protein A, which is labeled with a radioactive substance or an enzyme, is added. A method for detecting monoclonal antibodies bound to a solid phase, adding a hybridoma culture supernatant to a solid phase to which an anti-immunoglobulin antibody or protein A is adsorbed, and adding a receptor protein or the like labeled with a radioactive substance, an enzyme, etc. And a method for detecting a monoclonal antibody bound to a solid phase.
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 ことができるが、 通常は HAT (ヒポキサンチン、 アミノプテリン、 チミジン) を 添加した動物細胞用培地など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、 1〜20%、 好ましくは 10〜20%の牛胎児血清を含む RPMI 1640培地、 1〜 10%の牛胎児血清を含む GIT培地 (和光純薬工業 (株) ) またはハイプリドーマ 培養用無血清培地 (SFM-101、 日水製薬 (株) ) などを用いることができる。 培 養温度は、 通常 20〜40° (:、 好ましくは約 37 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。 ハイブリド一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。 (b) モノクローナル抗体の精製 The selection of the monoclonal antibody can be carried out according to a known method or a method analogous thereto. Usually, it can be carried out in a medium for animal cells to which HAT (hypoxanthine, aminopterin, thymidine) is added. 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 1-20%, preferably 10-20% fetal bovine serum, GIT medium containing 1-10% fetal bovine serum (Wako Pure Chemical Industries, Ltd.) or hybridoma culture medium Serum medium (SFM-101, Nissui Pharmaceutical Co., Ltd.) or the like can be used. The cultivation temperature is usually 20 to 40 ° (:, preferably about 37. The cultivation time is usually 5 days to 3 weeks, preferably 1 week to 2 weeks. The cultivation is usually 5% CO 2 gas. The antibody titer of the culture supernatant of the hybridoma can be measured in the same manner as the measurement of the antibody titer in the antiserum described above. (b) Purification of monoclonal antibody
モノクローナル抗体の分離精製は、 通常のポリクローナル抗体の分離精製と同 様に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、 DEAE) による吸脱着法、 超遠心法、 ゲルろ 過法、 抗原結合固相またはプロテイン Aあるいはプロテイン Gなどの活性吸着剤 により抗体のみを採取し、 結合を解離させて抗体を得る特異的精製法〕 に従って 行なうことができる。  Monoclonal antibodies can be separated and purified in the same manner as normal polyclonal antibodies. [Examples: salting out, alcohol precipitation, isoelectric focusing, electrophoresis, ion exchangers (ex. , DEAE) adsorption / desorption method, ultracentrifugation method, gel filtration method, antigen-binding solid phase or specific purification by collecting only the antibody using an active adsorbent such as protein A or protein G and dissociating the bond to obtain the antibody Act].
〔ポリクローナル抗体の作製〕  (Preparation of polyclonal antibody)
本発明のポリクローナル抗体は、 公知あるいはそれに準じる方法にしたがって 製造することができる。 例えば、 免疫抗原 (本発明の受容体蛋白質等の抗原) と キャリア一蛋白質との複合体をつくり、 上記のモノクローナル抗体の製造法と同 様に哺乳動物に免疫を行ない、 該免疫動物から本発明の受容体蛋白質等に対する 抗体含有物を採取して、 抗体の分離精製を行なうことにより製造できる。  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 receptor protein of the present invention) and a carrier-protein is formed, and a mammal 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 receptor protein or the like and separating and purifying the antibody.
哺乳動物を免疫するために用いられる免疫抗原とキヤリァ一蛋白質との複合体 に関し、 キャリアー蛋白質の種類およびキャリア一とハプテンとの混合比は、 キ ャリァ一蛋白質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、 ゥシ血清アルプミ ン、 ゥシサイログロプリン、 キーホール · リンぺット ·へモシァニン等を重量比 でハプテン 1に対し、 約 0 . 1〜2 0、 好ましくは約 1〜 5の割合でカプルさせ る方法が用いられる。  Regarding the complex of an immunizing antigen and a carrier protein used for immunizing mammals, the type of carrier protein and the mixing ratio of carrier to hapten are determined by the ratio of hapten immunized by cross-linking to carrier protein. Any antibody may be cross-linked at any ratio as long as it can be efficiently produced.For example, serum serum alpmin, shishiro glopurin, keyhole, limpet, hemocyanin, etc. are weight ratios. A method of coupling the hapten 1 at a ratio of about 0.1 to 20 and preferably about 1 to 5 is used.
また、 ハプテンとキャリア一蛋白質の力プリングには、 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トゃ不完全フロイントアジュバントを投与してもよい。 投 与は、 通常約 2〜 6週毎に 1回ずつ、 計約 3〜10回程度行なうことができる。  Various condensing agents can be used for force-coupling between the hapten and the carrier-protein, but daltaraldehyde, carbodiimide, maleimide active ester, an active ester reagent containing a thiol group or a dithioviridyl group, etc. are used. . The condensation product is administered to a warm-blooded animal itself or together with a carrier or diluent at a site where antibody production is possible. Complete Freund's adjuvant / incomplete Freund's adjuvant may be administered in order to enhance the antibody-producing ability upon administration. The administration can usually be performed once every about 2 to 6 weeks, for a total of about 3 to 10 times.
ポリクローナル抗体は、 上記の方法で免疫された哺乳動物の血液、 腹水など、 好ましくは血液から採取することができる。 抗血清中のポリクローナル抗体価の測定は、 上記の血清中の抗体価の測定と同 様にして測定できる。 ポリクロ一ナル抗体の分離精製は、 上記のモノクローナル 抗体の分離精製と同様の免疫グロプリンの分離精製法に従って行なうことができ る。 The polyclonal antibody can be collected from blood, ascites, etc., preferably from blood, of the mammal 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 serum 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.
(A) 本発明の受容体蛋白質またはその部分ペプチドの mR N A量の測定 本発明の受容体蛋白質またはその部分ペプチドの mR N A量の測定の具体例と しては、 以下の方法が挙げられる。  (A) Measurement of mRNA Amount of Receptor Protein or Partial Peptide of the Receptor Protein of the Present Invention Specific examples of measurement of mRNA amount of the receptor protein of the present invention or a partial peptide thereof include the following methods.
(0 正常あるいは疾患モデル非ヒト哺乳動物 (例えば、 マウス、 ラット、 ゥ サギ、 ヒッジ、 ブタ、 ゥシ、 ネコ、 ィヌ、 サルなど、 より具体的には痴呆ラット、 肥満マウス、 動脈硬化ゥサギ、 担癌マウスなど) またはヒト細胞に対して、 薬剤 (0 normal or disease model non-human mammals (e.g., mouse, rat, rabbit, sheep, pigeon, pig, cat, dog, monkey, etc., more specifically, dementia rat, obese mouse, atherosclerotic rabbit, For cancer-bearing mice) or human cells
(例えば、 抗痴呆薬、 血圧低下薬、 抗癌剤、 抗肥満薬など) あるいは物理的スト レス (例えば、 浸水ストレス、 電気ショック、 明暗、 低温など) などを与え、 一 定時間経過した後に、 血液、 あるいは特定の臓器 (例えば、 脳、 肺、 大腸など) 、 または臓器から単離した組織、 あるいは細胞を得る。 (E.g., anti-dementia drugs, anti-hypertensive drugs, anti-cancer drugs, anti-obesity drugs, etc.) or physical stress (e.g., flooding stress, electric shock, light / dark, low temperature, etc.) Alternatively, a specific organ (eg, brain, lung, large intestine, etc.) or a tissue or cell isolated from the organ is obtained.
得られた細胞に含まれる本発明の受容体蛋白質またはその部分ペプチドの mR NAは、 例えば、 通常の方法により細胞等から mR NAを抽出し、 例えば、 TaqMan PCRなどの手法を用いることにより定量することができ、 公知の手段によ りノザンブロットを行うことにより解析することもできる。  The mRNA of the receptor protein of the present invention or its partial peptide contained in the obtained cells is quantified by, for example, extracting mRNA from cells or the like by a usual method, and using, for example, a technique such as TaqMan PCR. It can also be analyzed by performing Northern blotting by known means.
(i i) 本発明の受容体蛋白質もしくはその部分べプチドを発現する形質転換体 を上記の方法に従い作製し、 該形質転換体に含まれる本発明の受容体蛋白質また はその部分ペプチドの mR N Aを同様にして定量、 解析することができる。  (ii) A transformant expressing the receptor protein of the present invention or a partial peptide thereof is prepared according to the above method, and the mRNA of the receptor protein of the present invention or the partial peptide thereof contained in the transformant is prepared. It can be quantified and analyzed in the same way.
(B ) 本発明の受容体蛋白質もしくはその部分ペプチドまたはその塩の定量 本発明の抗体は、 本発明の受容体蛋白質等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受容体蛋白質等の定量、 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。 例えば、  (B) Quantification of the receptor protein of the present invention or a partial peptide thereof or a salt thereof The antibody of the present invention can specifically recognize the receptor protein of the present invention and the like. Can be used for quantification of the receptor protein and the like, particularly for quantification by sandwich immunoassay. For example,
(i) 本発明の钪体と、 被検波および標識化受容体蛋白質等とを競合的に反応 させ、 該抗体に結合した標識化受容体蛋白質等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受容体蛋白質等の定量法、  (i) reacting the antibody of the present invention with a test wave and a labeled receptor protein or the like in a competitive manner, and measuring the ratio of the labeled receptor protein or the like bound to the antibody; Method for quantifying the receptor protein of the present invention in a test solution,
(i i) 被検液と担体上に不溶化した本発明の抗体および檩識化された本発明の 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受容体蛋白質等の定量法などが挙 げられる。 (ii) the antibody of the present invention insolubilized on the test solution and the carrier and the antibody of the present invention A method for measuring the activity of a labeling agent on an insolubilized carrier after simultaneous or continuous reaction with an antibody, and a method for quantifying the receptor protein of the present invention in a test solution, and the like.
上記 (i i) においては、 一方の抗体が本発明の受容体蛋白質等の N端部を認識 する抗体で、 他方の抗体が本発明の受容体蛋白質等の C端部に反応する抗体であ ることが好ましい。  In the above (ii), one antibody is an antibody that recognizes the N-terminal of the receptor protein or the like of the present invention, and the other antibody is an antibody that reacts with the C-terminal of the receptor protein or the like of the present invention. Is preferred.
本発明の受容体蛋白質等に対するモノクローナル抗体 (以下、 本発明のモノク 口一ナル抗体と称する場合がある) を用いて本発明の受容体蛋白質等の測定を行 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 ' 、 また は F a b画分を用いてもよい。 本発明の受容体蛋白質等に対する抗体を用いる測 定法は、 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、 受容 体蛋白質量) に対応した抗体、 抗原もしくは抗体—抗原複合体の量を化学的また は物理的手段により検出し、 これを既知量の抗原を含む標準液を用いて作製した 標準曲線より算出する測定法であれば、 いずれの測定法を用いてもよい。 例えば、 ネフロメトリ一、 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、 後に記載するサンドイッチ法を用いるのが特 に好ましい。 The receptor protein and the like of the present invention can be measured using a monoclonal antibody against the receptor protein and the like of the present invention (hereinafter sometimes referred to as the monoclonal antibody of the present invention), and detection by tissue staining and the like can be performed. You can do it too. For these purposes, the antibody molecule itself may be used, or F (ab ') 2 , Fab', or Fab fraction of the antibody molecule may be used. The measurement method using an antibody against the receptor protein or the like 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 an antigen (for example, the amount of a receptor protein) in a liquid to be measured. Any measurement method may be used as long as the amount of the body is detected by chemical or physical means, and the amount is calculated from a standard curve prepared using a standard solution containing a known amount of antigen. . For example, nephrometry, a competitive method, an immunometric method, and a sandwich method are suitably used, but in terms of sensitivity and specificity, it is particularly preferable to use a sandwich method described later.
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、 〔1251〕 、 1311〕 、 ¾〕 、 "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、 β —ガラクトシダ一ゼ、 β—グ ルコシダーゼ、 アルカリホスファターゼ、 バーオキシダーゼ、 リンゴ酸脱水素酵 素などが用いられる。 蛍光物質としては、 例えば、 フルォレスカミン、 フルォレ ッ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—アビジン系を用いることも できる。 As a labeling agent used in a measurement method using a labeling substance, for example, a radioisotope, an enzyme, a fluorescent substance, a luminescent substance and the like are used. Radioisotopes, if example embodiment, [125 1], 131 1], ¾], "c], etc. As the enzyme, large preferably stable and specific activity, e.g., beta -. Galactosidase one , Β-glucosidase, alkaline phosphatase, peroxidase, malate dehydrogenase, etc. As the fluorescent substance, for example, fluorescamine, fluorescein isothiocyanate, etc., is used. For example, luminol, luminol derivatives, luciferin, lucigenin, etc. In addition, a biotin-avidin system may be used for binding an antibody or antigen to a labeling agent.
抗原また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、 蛋 白質または酵素等を不溶化、 固定化するのに用いられる化学結合を用いる方法 もよい。 担体としては、 例えば、 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 るいはガラス等が用いられる。 For insolubilization of the antigen or antibody, physical adsorption may be used. A method using a chemical bond used for insolubilizing or immobilizing white matter or an enzyme may be used. As the carrier, for example, insoluble polysaccharides such as agarose, dextran, and cellulose; synthetic resins such as polystyrene, polyacrylamide, and silicon; and glass are used.
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 (一次反応) 、 さらに標識化した本発明のモノクローナル抗体を反応さ せ (二次反応) た後、 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 中の本発明の受容体蛋白質量を定量することができる。 一次反応と二次反応は逆 の順序に行なっても、 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上記のそれらに準じることができる。  In the sandwich method, the test solution is reacted with the insolubilized monoclonal antibody of the present invention (primary reaction), and further reacted with the labeled monoclonal antibody of the present invention (secondary reaction). By measuring the activity of the receptor protein, the amount of the receptor 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.
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 immunoassay by the sandwich method, the antibody used for the solid phase antibody or the labeling antibody is not necessarily one kind, and a mixture of two or more kinds of antibodies is used for the purpose of improving measurement sensitivity and the like. May be used.
本発明のサンドィツチ法による受容体蛋白質等の測定法においては、 一次反応 と二次反応に用いられる本発明のモノク口一ナル抗体は受容体蛋白質等の結合す る部位が相異なる抗体が好ましく用いられる。 すなわち、 一次反応および二次反 応に用いられる抗体は、 例えば、 二次反応で用いられる抗体が、 受容体蛋白質の C端部を認識する場合、 一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 a receptor protein or the like by the sandwich method of the present invention, the monoclonal antibody of the present invention used in the primary reaction and the secondary reaction is preferably an antibody having a different binding site to the receptor protein or the like. Can be 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 receptor protein, the antibody used in the primary reaction is preferably the C-terminal. For example, an antibody that recognizes other than the N-terminal part is used.
本発明のモノクローナル坊体をサンドイッチ法以外の測定システム、 例えば、 競合法、 ィムノメトリック法あるいはネフロメトリーなどに用いることができる。 競合法では、 被検液中の抗原と標識抗原とを抗体に対して競合的に反応させたの ち、 未反応の標識抗原と (F ) と抗体と結合した標識抗原 (B ) とを分離し (B Z F分離) 、 B、 F何れかの標識量を測定し、 被検液中の坊原量を定量する。 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一 ル、 上記抗体に対する第 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 body of the present invention can be used in a measurement system other than the sandwich method, for example, a competition method, an immunometric method, or a nephrometry. In the competition method, the antigen in the test solution and the labeled antigen are allowed to react competitively with the antibody, and then the unreacted labeled antigen is separated from (F) and the labeled antigen (B) bound to the antibody. Then (BZF separation), the amount of labeling of either B or F is measured, and the amount of Bobara in the test solution is quantified. In this reaction method, a soluble antibody is used as the antibody, B / F separation is performed using polyethylene glycol, a liquid phase method using a second antibody against the above antibody, or a solid phase antibody is used as the first antibody. Alternatively, a solid phase method using a soluble first antibody and a solid phase antibody as the second antibody is used.
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坊 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、 何れかの相の標識量 を測定し被検液中の抗原量を定量する。 In the trick method, the antigen in the test solution and the immobilized antigen After a competitive reaction with the body, the solid phase and the liquid phase are separated, or the antigen in the test solution is allowed to react with an excessive amount of the labeled antibody, and then the immobilized antigen is added and unreacted After binding the labeled antibody to the solid phase, the solid phase and the liquid phase are separated. Next, the amount of label in any phase is measured to determine the amount of antigen in the test solution.
また、 ネフロメトリーでは、 ゲル内あるいは溶液中で抗原抗体反応の結果、 生 じた不溶性の沈降物の量を測定する。 被検液中の抗原量が僅かであり、 少量の沈 降物しか得られない場合にもレーザ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 nephelometry, the amount of insoluble sediment generated as a result of 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 precipitate 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測定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受容体蛋白質またはその塩 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 〔例えば、 入江 寛編 「ラジオィムノアッセ ィ」 (講談社、 昭和 49年発行) 、 入江 寛編 「続ラジオィムノアツセィ」 (講談 社、 昭和 54年発行) 、 石川栄治ら編 「酵素免疫測定法」 (医学書院、 昭和 53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 57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 62年発行) 、 「メ ソッズ 'イン 'ェンザィモロジ一 (Methods in ENZYMOLOGY) 」 Vol . 70  In applying these individual immunological measurement methods to the measurement method of the present invention, no special conditions, operations, and the like need to be set. What is necessary is just to construct the measuring system of the receptor protein of the present invention or its salt by adding ordinary technical considerations of those skilled in the art to ordinary conditions and operation methods in each method. For details of these general technical means, it is possible to refer to reviews and compendiums. [For example, Hiroshi Irie “Radio Nonoassy” (Kodansha, published in 1974), Hiroshi Irie “ "Radio Imnoatsay" (Kodansha, published in 1979), "Enzyme immunoassay" edited by Eiji Ishikawa et al. (Medical Publishing, published in 1978), "Enzyme immunoassay" edited by Eiji Ishikawa et al. (Edition) (Medical Shoin, published in 1982), edited by Eiji Ishikawa et al., "Enzyme Immunoassay" (3rd edition) (published in 1987), "Methods in ENZYMOLOGY" Vol. 70
(Immunochemical Techniques (Part A) )、 同書 Vol . 73 (Immunochemical Techniques (Par t B))、 同書 Vol . 74 (Immunochemical Techniques (Part C) )、 同書 Vol. 84 (Immunochemical Techniques (Part D: Selected Immunoassays) ) , 同書 Vol. 92 (Immunochemi cal Techniques (Part E :Monocl onal Ant ibodi es and General Immunoassay Methods) )、 同書 Vol . 121 (Immunochemical (Immunochemical Techniques (Part A)), ibid.Vol. 73 (Immunochemical Techniques (Part B)), ibid.Vol. 74 (Immunochemical Techniques (Part C)), ibid.Vol. 84 (Immunochemical Techniques (Part D: Selected Immunoassays)) ), Ibid., Vol. 92 (Immunochemi cal Techniques (Part E: Monoclonal Ant ibodies and General Immunoassay Methods)), ibid., Vol. 121 (Immunochemical).
Techniques (Par t I: Hybr idoma Techno l oGy and Monoclonal Ant ibodies) ) (以上、 アカデミックプレス社発行)など参照〕 。 Techniques (Part I: Hybridoma TechnoGy and Monoclonal Ant ibodies)) (see Academic Press).
以上のように、 本発明の抗体を用いることによって、 本発明の受容体蛋白質ま たはその塩を感度良く定量することができる。  As described above, by using the antibody of the present invention, the receptor protein of the present invention or a salt thereof can be quantified with high sensitivity.
( 1 ) アンチセンスオリゴヌクレオチドを含有する医薬 本発明のオリゴヌクレオチドは、 細胞内で安定であり、 細胞透過性が高く、 目 標とするセンス鎖に対する親和性が大きく、 低毒性である。 (1) a drug containing an antisense oligonucleotide The oligonucleotide of the present invention is stable in cells, has high cell permeability, has high affinity for the target sense strand, and has low toxicity.
本発明のポリヌクレオチドは、 例えば、 本発明の受容体蛋白質の過剰発現に関 連する疾患、 例えば癌 (例、 非小細胞肺癌、 卵巣癌、 前立腺癌、 胃癌、 膀胱癌、 乳癌、 子宮頸部癌、 結腸癌、 直腸癌など) などの疾患に対する予防および Zまた は治療剤などの医薬として使用することができる。  The polynucleotide of the present invention may be, for example, a disease associated with overexpression of the receptor protein of the present invention, such as cancer (eg, non-small cell lung cancer, ovarian cancer, prostate cancer, gastric cancer, bladder cancer, breast cancer, cervix) Cancer, colon cancer, rectal cancer, etc.), and can be used as a medicament such as a prophylactic or Z or therapeutic agent.
本発明のアンチセンスオリゴヌクレオチドは、 変化せしめられたり、 修飾され た糖、 塩基、 結合を含有していて良く、 リボゾーム、 ミクロスフエアのような特 殊な形態で供与されたり、 遺伝子治療により適用されたり、 付加された形態で与 えられることができうる。 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) といった疎水性のものが挙げられる。 付加するに好ま しい脂質としては、 コレステロールやその誘導体 (例えば、 コレステリルクロ口 ホルメート、 コール酸など) が挙げられる。 これらは、 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 ァ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一ル、 テトラエチレングリコールなどのグリコ一 ルをはじめとした当該分野で知られた水酸基の保護基が挙げられるが、 それに限 定されるものではない。  The antisense oligonucleotides of the present invention may contain altered or modified sugars, bases, or bonds, may be provided in special forms such as ribosomes, microspheres, or may be applied by gene therapy. , Can be provided in an added form. Additional forms include polycations such as polylysine, which acts to neutralize the charge on the phosphate backbone, and lipids that enhance interaction with cell membranes or increase nucleic acid uptake (eg, Hydrophobic substances such as phospholipids and cholesterol). Preferred lipids for addition include cholesterol and its derivatives (eg, cholesteryl chloroformate, 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 capping groups specifically arranged at the 3 'end or 5' end of nucleic acids for preventing degradation by nucleases such as exonuclease and RNase. Examples of such capping groups include, but are not limited to, hydroxyl-protecting groups known in the art, including glycols such as polyethylene glycol and tetraethylene glycol.
本発明のアンチセンスオリゴヌクレオチドを上記の医薬として使用する場合、 公知の方法に従って製剤化し、 投与することができる。  When the antisense oligonucleotide of the present invention is used as the above medicine, it can be formulated and administered according to a known method.
例えば、 該アンチセンスオリゴヌクレオチドを用いる場合、 該アンチセンスォ リゴヌクレオチドを単独あるいはレトロウイルスベクタ一、 アデノウイルスべク タ一、 アデノウイルスァソシエーテツドウィルスベクタ一などの適当なベクタ一 に挿入した後、 常套手段に従って、 ヒトまたは非ヒト哺乳動物 (例、 ラット、 ゥ サギ、 ヒッジ、 ブ夕、 ゥシ、 ネコ、 ィヌ、 サルなど) に対して経口的または非経 口的に投与することができる。 該アンチセンスオリゴヌクレオチドは、 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 oligonucleotide is used, the antisense oligonucleotide 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 non-human administration to human or non-human mammals (eg, rats, gray egrets, sheep, higgs, bush, red sea lions, cats, dogs, monkeys, etc.) It can be administered orally. The antisense oligonucleotide 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.
該アンチセンスオリゴ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、 がん治療の目的で本発明のアンチセンス オリゴヌクレオチドを臓器 (例、 肝臓、 肺、 心臓、 腎臓など) に局所投与する場 合、 一般的に成人 (体重 60kg) においては、 一日につき該アンチセンスオリゴヌ クレオチドを約 0. l〜100mg投与する。  The dose of the antisense oligonucleotide varies depending on the target disease, the subject of administration, the route of administration, and the like. For example, the antisense oligonucleotide of the present invention may be administered to an organ (eg, liver, lung, When topically administered to the heart, kidney, etc., generally, for an adult (body weight 60 kg), about 0.1 to 100 mg of the antisense oligonucleotide is administered per day.
さらに、 該アンチセンスオリゴヌクレオチドは、 組織や細胞における本発明の ポリヌクレオチド (例、 D NA) の存在やその発現状況を調べるための診断用ォ 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 oligonucleotide can also be used as a diagnostic oligonucleotide probe for examining the presence of the polynucleotide (eg, DNA) of the present invention in tissues or cells and the state of expression thereof.
( 2 ) レポ一夕一ジーンアツセィ (2) Repo One Night One Gene Atsushi
さらに、 本発明は、 本発明の受容体蛋白質の遺伝子の転写調節領域の下流 (発 現制御下) にレポーター遺伝子を連結した組換え D NAで形質転換した形質転換 体を、 試験化合物の存在下および非存在下で培養した場合における、 それぞれの レポ一夕一活性を測定し、 比較することを特徴とする該転写調節活性を促進また は阻害する化合物のスクリーニング方法、 およびこの方法のためのスクリ一ニン グ用キットも提供する。  Furthermore, the present invention provides a transformant transformed with a recombinant DNA in which a reporter gene is ligated downstream (under expression control) of the transcriptional regulatory region of the gene of the receptor protein of the present invention in the presence of a test compound. And a method for screening for a compound that promotes or inhibits the transcriptional regulatory activity, comprising measuring and comparing each repo overnight activity when cultured in the absence and presence of the same, and a script for this method. A kit for single use is also provided.
本発明の受容体蛋白質の遺伝子の転写調節領域としては、 配列番号: 2 0で表 される塩基配列と同一または実質的に同一な塩基配列を含有する D NAなどが挙 げられる。  Examples of the transcription regulatory region of the gene of the receptor protein of the present invention include a DNA having the same or substantially the same nucleotide sequence as the nucleotide sequence represented by SEQ ID NO: 20.
配列番号: 2 0で表される塩基配列と実質的に同一な塩基配列を含有する D N Aとしては、 ハイストリンジエンドな条件下でハイブリダィズする塩基配列を有 し、 配列番号: 2 0と実質的に同質の転写調節活性 (例、 プロモータ一活性な ど) を有する D N Aであれば何れのものでもよい。  The DNA containing a base sequence substantially identical to the base sequence represented by SEQ ID NO: 20 has a base sequence that hybridizes under high stringency end conditions, and has substantially the same sequence as SEQ ID NO: 20. Any DNA may be used as long as it has the same transcriptional regulatory activity (eg, promoter-one activity).
配列番号: 2 0で表される塩基配列とハイストリンジェントな条件下でハイブ リダィズできる D N Aとしては、 例えば、 配列番号: 2 0で表される塩基配列と 約 50%以上、 好ましくは約 60%以上、 さらに好ましくは約 70%以上、 より 好ましくは約 80%以上、 特に好ましくは約 90%以上、 最も好ましくは約 9 5%以上の相同性を有する塩基配列を含有する DN Aなどが用いられる。 Examples of DNA that can hybridize with the nucleotide sequence represented by SEQ ID NO: 20 under high stringent conditions include, for example, the nucleotide sequence represented by SEQ ID NO: 20 A base having a homology of about 50% or more, preferably about 60% or more, more preferably about 70% or more, more preferably about 80% or more, particularly preferably about 90% or more, and most preferably about 95% or more. A DNA containing the sequence is used.
ハイブリダィゼ一シヨンは、 自体公知の方法あるいはそれに準じる方法、 例え ば、 モレキユラ一 '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、 市販のライブラリ一を使用する場合、 添付の使用説明書に 記載の方法に従って行なうことができる。 より好ましくは、 ハイストリンジェン トな条件に従つて行なうことができる。  Hybridization is performed according to a method known per se or a method analogous thereto, for example, the method described in Molecular Cloning 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). be able to. When a commercially available library is used, it can be performed according to the method described in the attached instruction manual. More preferably, the reaction can be performed under high stringency conditions.
ハイストリンジエンドな条件とは、 例えば、 ナトリウム濃度が約 19〜40m M、 好ましくは約 19〜2 OmMで、 温度が約 50〜70 :、 好ましくは約 60 〜65 の条件を示す。 特に、 ナトリウム濃度が約 19 mMで温度が約 65°Cの 場合が最も好ましい。  The high stringent end conditions include, for example, a sodium concentration of about 19 to 40 mM, preferably about 19 to 2 OmM, and a temperature of about 50 to 70: preferably about 60 to 65. In particular, it is most preferable that the sodium concentration is about 19 mM and the temperature is about 65 ° C.
また、 転写調節領域 (例、 配列番号: 20で表される塩基配列と実質的に同一 な塩基配列を含有する DN A等) としては、 配列番号: 20で表される塩基配列 と実質的に同一な塩基配列に、 さらに 5' 上流のゲノム DNA配列が付加された 配列なども挙げられる (好ましくはプロ一モーター領域) 。 付加する塩基配列の 塩基数は、 例えば約 10 Kb以下、 好ましくは約 5 Kb以下、 さらに好ましくは 約 2Kb以下、 最も好ましくは約 1Kb以下の配列数が挙げられる。 さらに、 配 列番号: 20で表される塩基配列と実質的に同一な塩基配列に、 さらに本発明の 受容体蛋白質の遺伝子のイントロンの配列が付加された配列なども挙げられる。 レポーター遺伝子としては、 例えば、 l a c Z (ヨーガラクトシダーゼ遺伝 子) 、 クロラムフエニコールァセチルトランスフェラーゼ (CAT) 、 ルシフェラ ーゼ、 成長因子、 ]3—グルクロニダ一ゼ、 アル力リホスファタ一ゼ、 Green fluorescent protein (GFP), /3—ラク夕マ一ゼなどが用いられる。  In addition, the transcription regulatory region (eg, DNA containing a nucleotide sequence substantially identical to the nucleotide sequence represented by SEQ ID NO: 20) is substantially the same as the nucleotide sequence represented by SEQ ID NO: 20 A sequence obtained by further adding a genomic DNA sequence at the 5 ′ upstream to the same base sequence (preferably a promoter region) is also included. The number of bases to be added is, for example, about 10 Kb or less, preferably about 5 Kb or less, more preferably about 2 Kb or less, and most preferably about 1 Kb or less. Furthermore, a sequence in which an intron sequence of the gene of the receptor protein of the present invention is further added to the nucleotide sequence substantially the same as the nucleotide sequence represented by SEQ ID NO: 20, etc. Reporter genes include, for example, lacZ (yogalactosidase gene), chloramphenicol acetyltransferase (CAT), luciferase, growth factor,] 3-glucuronidase, alphospholiphosphatase, Green fluorescent protein (GFP), / 3-lacquerase and the like are used.
レポ一夕一遺伝子産物 (例、 mRNA、 蛋白質) の量を公知の方法を用いて測 定することによって、 レポ一夕一遺伝子産物の量を増加させる試験化合物を本発 明の受容体蛋白質の転写調節活性 (好ましくはプロモーター活性) を制御 (特に 促進) する作用を有する化合物、 すなわち本発明の受容体蛋白質の発現を促進す る活性を有する化合物として選択できる。 逆に、 レポーター遺伝子産物の量を減 少させる試験化合物を本発明の受容体蛋白質の転写調節活性 (好ましくはプロモ 一夕一活性) を制御 (特に阻害) する作用を有する化合物、 すなわち本発明の受 容体蛋白質の発現を阻害する活性を有する化合物として選択することができる。 試験化合物としては、 例えば、 ペプチド、 蛋白質、 非ペプチド性化合物、 合成 化合物、 発酵生産物、 細胞抽出液、 植物抽出液、 動物組織抽出液、 血漿などから 選ばれた化合物である。 該化合物の塩としては、 前記の本発明の受容体蛋白質の 塩と同様のものが用いられる。 By measuring the amount of the repo overnight gene product (eg, mRNA, protein) using a known method, a test compound that increases the amount of the repo overnight gene product can be used as a test compound for the receptor protein of the present invention. A compound having an action of controlling (particularly promoting) transcriptional regulatory activity (preferably promoter activity), ie, promoting expression of the receptor protein of the present invention. Can be selected as a compound having a certain activity. Conversely, a test compound that reduces the amount of a reporter gene product is a compound that has the activity of controlling (particularly inhibiting) the transcriptional regulatory activity (preferably, overnight promoter) of the receptor protein of the present invention, that is, the compound of the present invention. The compound can be selected as a compound having an activity of inhibiting the expression of the receptor protein. The test compound is, for example, a compound selected from peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, plasma, and the like. As the salt of the compound, those similar to the aforementioned salts of the receptor protein of the present invention are used.
形質転換体の培養は、 前記の本発明の受容体蛋白質を含有する形質転換体と同 様にして行うことができる。 レポ一夕一遺伝子のベクター構築やアツセィ法は公 知の技術に従うことができる (例、 Molecular Biotechnology 13, 29-43, 1999) 。  The transformant can be cultured in the same manner as the above-described transformant containing the receptor protein of the present invention. The vector construction of the repo overnight gene and the Atsey method can be performed according to known techniques (eg, Molecular Biotechnology 13, 29-43, 1999).
本発明の受容体蛋白質の発現を促進する活性を有する化合物またはその塩は、 安全で低毒性な医薬として有用である。  The compound having an activity of promoting the expression of the receptor protein of the present invention or a salt thereof is useful as a safe and low-toxic drug.
本発明のペプチドの発現を阻害する活性を有する化合物またはその塩は、 本発 明の受容体蛋白質の生理活性を抑制するための安全で低毒性な医薬、 例えば、 癌 (例、 非小細胞肺癌、 卵巣癌、 前立腺癌、 胃癌、 膀胱癌、 乳癌、 子宮頸部癌、 結 腸癌、 直腸癌など) の予防および Zまたは治療剤として有用である。  A compound having an activity of inhibiting the expression of the peptide of the present invention or a salt thereof is a safe and low-toxic drug for suppressing the physiological activity of the receptor protein of the present invention, for example, cancer (eg, non-small cell lung cancer) Ovarian cancer, prostate cancer, gastric cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, rectal cancer, etc.).
該塩としては、 前記の本発明の受容体蛋白質の塩と同様のものが用いられる。 上記化合物を医薬として使用する場合は、 常套手段に従って製剤化することが できる。  As the salt, those similar to the aforementioned salts of the receptor protein of the present invention are used. When the above compound is used as a medicine, it can be formulated according to conventional means.
例えば、 該化合物は、 必要に応じて糖衣を施した錠剤、 カプセル剤、 エリキシ ル剤、 マイクロカプセル剤などとして経口的に、 あるいは水もしくはそれ以外の 薬学的に許容し得る液との無菌性溶液、 または懸濁液剤などの注射剤の形で非経 口的に使用できる。 例えば、 該化合物を生理学的に認められる公知の担体、 香味 剤、 賦形剤、 べヒクル、 防腐剤、 安定剤、 結合剤などとともに一般に認められた 製剤実施に要求される単位用量形態で混和することによって製造することができ る。 これら製剤における有効成分量は指示された範囲の適当な用量が得られるよ うにするものである。 また、 上記予防およびノまたは治療剤は、 例えば、 緩衝剤 (例えば、 リン酸塩 緩衝液、 酢酸ナトリウム緩衝液) 、 無痛化剤 (例えば、 塩化ベンザルコニゥム、 塩酸プロ力インなど) 、 安定剤 (例えば、 ヒト血清アルブミン、 ポリエチレング リコールなど) 、 保存剤 (例えば、 ベンジルアルコール、 フエノールなど) 、 酸 化防止剤などと配合してもよい。 調製された注射液は通常、 適当なアンプルに充 填される。 For example, the compound can be used as a sugar-coated tablet, capsule, elixir, microcapsule or the like as needed, orally, or aseptic solution with water or another pharmaceutically acceptable liquid. It can be used parenterally or in the form of injections such as suspensions. For example, the compound is mixed with known physiologically acceptable carriers, flavoring agents, excipients, vehicles, preservatives, stabilizers, binders, and the like in a unit dosage form generally required for the practice of pharmaceutical preparations. It can be manufactured by The amount of the active ingredient in these preparations is such that a suitable dosage in the specified range can be obtained. Examples of the prophylactic and therapeutic agents include, but are not limited to, a buffer (eg, a phosphate buffer, a sodium acetate buffer), a soothing agent (eg, benzalkonidum chloride, proforce hydrochloride, etc.), a stabilizer (eg, , Human serum albumin, polyethylene glycol, etc.), preservatives (eg, benzyl alcohol, phenol, etc.), antioxidants and the like. The prepared injection solution is usually filled into a suitable ampoule.
このようにして得られる製剤は安全で低毒性であるので、 例えば、 ヒトゃ哺乳 動物 (例えば、 ラット、 マウス、 ゥサギ、 ヒッジ、 ブ夕、 ゥシ、 ネコ、 ィヌ、 サ ルなど) に対して投与することができる。  The preparations obtained in this way are safe and low toxic, so they can be used, for example, in humans and mammals (eg, rats, mice, egrets, higgs, bushes, cats, cats, dogs, dogs, etc.). Can be administered.
該化合物またはその塩の投与量は、 投与対象、 対象臓器、 症状、 投与方法など により差異はあるが、 経口投与の場合、 一般的に例えば、 がん患者 (体重 60kgと して) においては、 一日につき約 0. l〜100nig、 好ましくは約 1. 0〜50mg、 より好 ましくは約 1. 0〜20nigである。 非経口的に投与する場合は、 その 1回投与量は投 与対象、 対象臓器、 症状、 投与方法などによっても異なるが、 例えば、 注射剤の 形では通常例えば、 癌症患者 (体重 60kgとして) においては、 一日につき約 0. 01 〜30mg、 好ましくは約 0. l〜20mg、 より好ましくは約 0. l〜10mgを静脈注射により 投与するのが好都合である。 他の動物の場合も、 体重 60kg当たりに換算した量を 投与することができる。 本明細書において、 塩基やアミノ酸などを略号で表示する場合、 その表示は、 IUPAC-IUB Commiss ion on Biochemi cal Nomenc latureによる略号あるいは当該分 野における慣用略号に基づくものである。 その例を以下に示す。 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。  The dose of the compound or a salt thereof varies depending on the administration subject, target organ, symptom, administration method, and the like. However, in the case of oral administration, for example, in a cancer patient (with a body weight of 60 kg), About 0.1 to 100 nig per day, preferably about 1.0 to 50 mg, more preferably about 1.0 to 20 nig. In the case of parenteral administration, the single dose varies depending on the subject of administration, target organ, symptoms, administration method, etc. For example, in the case of injection, it is usually used, for example, in cancer patients (with a body weight of 60 kg). It is convenient to administer about 0.01 to 30 mg, preferably about 0.1 to 20 mg, more preferably about 0.1 to 10 mg per day by intravenous injection. In the case of other animals, the dose can be administered in terms of weight per 60 kg. In the present specification, when a base, an amino acid, or the like is indicated by an abbreviation, the indication is based on an abbreviation by IUPAC-IUB Communication on Biochemical Nomenclature or a conventional abbreviation in the field. An example is shown below. When an amino acid can have optical isomers, the L-form is indicated unless otherwise specified.
Gly :グリシン  Gly: Glycine
Ala :ァラニン  Ala: Alanin
Val : ) リン  Val:) Lynn
Leu : ロイシン  Leu: Leucine
l ie :イソロイ  l ie: Isolloy
Ser :セリン Thr :スレオニン Ser: Serine Thr: Threonine
Cys :システィン  Cys: Cystine
Met :メチォニン  Met: Methionin
Glu :グルタミン酸  Glu: Glutamic acid
Asp :ァスパラギン酸  Asp: Aspartic acid
Lys : リジン  Lys: Lysine
Arg :アルギニン  Arg: Arginine
Hi s : ヒスチジン  Hi s: Histidine
Phe :フエ二ルァラニン  Phe: feniralanin
Tyr :チロシン  Tyr: Tyrosine
Trp : トリプトファン  Trp: Tryptophan
Pro :プロリン  Pro: Proline
Asn :ァスパラギン  Asn: Asparagine
Gin :ダル夕ミン  Gin: Dal Yu Min
pGlu : ピログルタミン酸  pGlu: pyroglutamic acid
Me :メチル基  Me: methyl group
Et :ェチル基  Et: ethyl group
Bu :ブチル基  Bu: butyl group
Ph :フエニル基  Ph: phenyl group
TC :チアゾリジン- 400 -カルボキサミド  TC: Thiazolidine-400-carboxamide
また、 本明細書中で繁用される置換基、 保護基および試薬を下記の記号で表記 する。  The substituents, protecting groups and reagents frequently used in the present specification are represented by the following symbols.
Tos : p -トルエンスルフォニル  Tos: p-toluenesulfonyl
CH0 :ホルミル  CH0: Formyl
Bz l  Bz l
Cl2Bz l 2, 6 -ジクロ口べンジル Cl 2 Bz l 2, 6-
Bom ベンジルォキシメチル  Bom benzyloxymethyl
Z ベンジルォキシカルポニル  Z benzyloxycarponyl
Cl-Z : 22—-クロ口べンジルォキシカルポニル Br-Z 2—ブロモベンジルォキシカルポニル Cl-Z: 22--Ventyloxycarponyl Br-Z 2-bromobenzyloxycarbonyl
Boc t -ブトキシカルボニル Boc t-butoxycarbonyl
DNP ジニトロフエノール DNP dinitrophenol
Tr t 卜リチル  Tr t Trityl
Bum t-ブトキシメチル  Bum t-butoxymethyl
Fmoc N - 9_フルォレニルメトキシカルポニル  Fmoc N-9_fluorenylmethoxycarbonyl
HOBt HOBt
HOOBt 3, 4-ジヒドロ- 3-ヒドロキシ -4-ォキソ -1, 2, 3-ベンゾトリアジン HONB 1 -ヒドロキシ- 5-ノルポルネン _2, 3 -ジカルポキシイミド HOOBt 3,4-Dihydro-3-hydroxy-4-oxo-1,2,3-benzotriazine HONB 1-Hydroxy-5-norporene-2,3-dicarpoxyimide
DCC Ν, Ν' -ジシクロへキシルカルポジイミド 本願明細書の配列表の配列番号は、 以下の配列を示す。  DCC Ν, Ν'-dicyclohexylcarposimide SEQ ID NOs in the Sequence Listing of the present specification indicate the following sequences.
〔配列番号: 1〕 [SEQ ID NO: 1]
以下の参考例 1で得られた c D N Αの塩基配列 (5 '側) の一部を示す。  The following shows a part of the nucleotide sequence (5 'side) of cDNΑ obtained in Reference Example 1 below.
〔配列番号: 2〕 [SEQ ID NO: 2]
以下の参考例 1における P C R反応で使用したプライマ一 1の塩基配列を示す c 〔配列番号: 3〕  Indicates the base sequence of primer 11 used in the PCR reaction in Reference Example 1 below c [SEQ ID NO: 3]
以下の参考例 1における P C R反応で使用したプライマー 2の塩基配列を示す c 〔配列番号: 4〕  Indicates the nucleotide sequence of primer 2 used in the PCR reaction in Reference Example 1 below c [SEQ ID NO: 4]
以下の参考例 2で得られた c D NAの塩基配列の一部 (中央部) を示す。  A part (center) of the nucleotide sequence of cDNA obtained in Reference Example 2 below is shown.
〔配列番号: 5〕 [SEQ ID NO: 5]
以下の参考例 2における P C R反応で使用したプライマー 3の塩基配列を示す c 〔配列番号: 6〕  Indicates the base sequence of primer 3 used in the PCR reaction in Reference Example 2 below c [SEQ ID NO: 6]
以下の参考例 2における P C R反応で使用したプライマー 4の塩基配列を示す c 〔配列番号: 7〕  Indicates the base sequence of primer 4 used in the PCR reaction in Reference Example 2 below c [SEQ ID NO: 7]
以下の参考例 3で得られた c D N Aの塩基配列 (3 '側) の一部を示す。  FIG. 9 shows a part of the nucleotide sequence (3 ′ side) of cDNA obtained in Reference Example 3 below.
〔配列番号: 8〕 [SEQ ID NO: 8]
以下の参考例 3における P C R反応で使用したプライマー 5の塩基配列を示す。 〔配列番号: 9〕 以下の参考例 3における P C R反応で使用したプライマ一 6の塩基配列を示す 〔配列番号: 10〕 The base sequence of primer 5 used in the PCR reaction in Reference Example 3 below is shown. [SEQ ID NO: 9] This shows the base sequence of primer 16 used in the PCR reaction in Reference Example 3 below [SEQ ID NO: 10]
以下の参考例 3における P C R反応で使用したプライマー 7の塩基配列を示す c 〔配列番号: 11〕 C represents the base sequence of primer 7 used in the PCR reaction in Reference Example 3 below [SEQ ID NO: 11]
参考例 5で得られたヒト脳由来新規受容体蛋白質 NEPHAのァミノ酸配列を 示す。  7 shows the amino acid sequence of the human brain-derived novel receptor protein NEPHA obtained in Reference Example 5.
〔配列番号: 12〕  [SEQ ID NO: 12]
ヒト脳由来新規受容体蛋白質 NEPHAをコードする cDN Aの塩基配列を示 す。  The nucleotide sequence of cDNA which encodes a novel receptor protein NEPHA derived from human brain is shown.
〔配列番号: 13〕  [SEQ ID NO: 13]
本発明のアンチセンスオリゴヌクレオチドの塩基配列を示す。  1 shows the base sequence of the antisense oligonucleotide of the present invention.
〔配列番号: 14〕  [SEQ ID NO: 14]
本発明のアンチセンスオリゴヌクレオチドの塩基配列を示す。  1 shows the base sequence of the antisense oligonucleotide of the present invention.
〔配列番号: 15〕  [SEQ ID NO: 15]
以下の実施例 1で使用したオリゴヌクレオチドの塩基配列を示す。  1 shows the base sequence of the oligonucleotide used in Example 1 below.
〔配列番号: 16〕  [SEQ ID NO: 16]
以下の実施例 1で使用したオリゴヌクレオチドの塩基配列を示す。  1 shows the base sequence of the oligonucleotide used in Example 1 below.
〔配列番号: 17〕  [SEQ ID NO: 17]
以下の実施例 1における P C R反応で使用したプライマ一の塩基配列を示す。 〔配列番号: 18〕  1 shows the nucleotide sequence of a primer used in the PCR reaction in Example 1 below. [SEQ ID NO: 18]
以下の実施例 1における PC R反応で使用したプライマーの塩基配列を示す。 〔配列番号: 19〕 . 以下の実施例 1における P C R反応で使用したプローブの塩基配列を示す。  The base sequence of the primer used in the PCR reaction in Example 1 below is shown. [SEQ ID NO: 19] This shows the base sequence of the probe used in the PCR reaction in Example 1 below.
〔配列番号: 20〕  [SEQ ID NO: 20]
NEPHAの転写調節領域の塩基配列を示す。 後述の参考例 5で取得された大腸菌 (Escherichia coli) TOP10/pTB2230は、 2001年 5月 24日から、 日本国茨城県つくば巿東卜 1-1 中央第 6 (郵便番号 305 - 8566) の独立行政法人産業技術総合研究所特許生物寄託センターに寄託番号 FERM BP- 7606として、 2001年 4月 24日から、 大阪府大阪市淀川区十三本町 2-17 - 85 (郵便番号 532- 8686) の財団法人 発酵研究所 (IF0) に受託番号 IF0 16623とし て寄託されている。 以下において、 参考例および実施例により本発明をより具体的にするが、 この 発明はこれらに限定されるものではない。 2 shows the nucleotide sequence of the transcription control region of NEPHA. Escherichia coli TOP10 / pTB2230 obtained in Reference Example 5 described below has been established on May 24, 2001 by the establishment of 1-1 Chukoku No. 6 Tsukuba Totoro, Ibaraki, Japan (zip code 305-8566). Deposited number at Patent Organism Depositary, National Institute of Advanced Industrial Science and Technology From April 24, 2001, FERM BP-7606 was given to the Fermentation Research Institute (IF0) of 2-17-85, Jusanhoncho, Yodogawa-ku, Osaka-shi (Osaka Prefecture) under the accession number IF0-16623. Has been deposited. Hereinafter, the present invention will be more specifically described with reference examples and examples, but the present invention is not limited thereto.
参考例 1 Reference example 1
ヒト脳由来新規受容体蛋白質をコ一ドする cDNAの 5'部分塩基配列の決定 Encoding a novel receptor protein from human brain. Determination of the 5 'partial nucleotide sequence of cDNA.
MTC Pane l Iに含まれるヒト脳 cDNA (CL0NTECH社) を铸型とし、 2個のプライマ 一、 プライマー 1 (配列番号: 2 ) およびプライマー 2 (配列番号: 3 ) を用い て PCR反応を行った。 該反応における反応液の組成は上記 cDNA 0. 5 1を铸型とし て使用し、 Pfu Turbo DNA Polymerase (STRATAGENE社) 1U、 プライマー 1 (配列 番号: 2 ) およびプライマ一 2 (配列番号: 3 ) を各 1 M、 dNTPsを 200 Μ、 お よび 2XGC Buf ferl (宝酒造) を 10 1加え、 20 1の液量とした。 PCR反応は、 96X3、 1分の後、 96°C、 1分、 60°C、 1分、 72°C、 1分のサイクルを 40回繰り返し行 つた。 次にァガロースゲル電気泳動にて該 PCR反応産物を精製した後、 これを Zero Blunt T0P0 PCRクロ一ニングキット (Invi trogen社) の処方に従いプラス ミドベクタ一 pCR-Bl unt i l- T0P0 (Invi t rogen社) へサブクロ一ニングした。 これ を大腸菌 TOP 10に導入し、 カナマイシンを含む LB寒天培地中で選択した。 個々の クローンの配列を解析した結果、 新規受容体蛋白質の一部をコードする cDNAの塩 基配列 (配列番号: 1 ) を得た。 参考例 2  The human brain cDNA (CL0NTECH) contained in MTC Panel I was type III, and PCR was carried out using two primers, primer 1 (SEQ ID NO: 2) and primer 2 (SEQ ID NO: 3). . The composition of the reaction solution used in the reaction was the above cDNA 0.51 as type III, 1 U of Pfu Turbo DNA Polymerase (STRATAGENE), primer 1 (SEQ ID NO: 2) and primer 1 (SEQ ID NO: 3) 1 M each, 200 l of dNTPs, and 101 of 2XGC Buf ferl (Takara Shuzo) were added to give a liquid volume of 201. The PCR reaction was repeated 96 times at 96X3 for 1 minute, followed by a cycle of 96 ° C, 1 minute, 60 ° C, 1 minute, 72 ° C, and 1 minute 40 times. Next, the PCR reaction product was purified by agarose gel electrophoresis, and then purified using the plasmid vector pCR-Bluntil-T0P0 (Invitrogen) according to the prescription of Zero Blunt T0P0 PCR Cloning Kit (Invitrogen). ). This was introduced into E. coli TOP10 and selected on LB agar medium containing kanamycin. As a result of analyzing the sequence of each clone, a nucleotide sequence (SEQ ID NO: 1) of cDNA encoding a part of the novel receptor protein was obtained. Reference example 2
ヒト脳由来新規受容体蛋白質をコードする cDNAの中央部分塩基配列の決定 Determination of the central partial nucleotide sequence of cDNA encoding a novel receptor protein from human brain.
ヒト脳 cDNA (CL0NTECH¾ MTC Panel I) を铸型とし、 2個のプライマ一、 プライ マ一 3 (配列番号: 5 ) およびプライマー 4 (配列番号: 6 ) を用いて PCR反応 を行った。 該反応における反応液の組成は上記 cDNA 1 1を铸型として使用し、 P fu Turbo DNA Polymerase (STRATAGENE社) 2. 5U、 プライマー 3 (配列番号: 5 ) およびプライマー 4 (配列番号: 6 ) を各 1 M、 dNTPsを 200 M、 および 2XGC Bufferl (宝酒造) を 25 1加え、 50^1の液量とした。 PCR反応は、 96 、 1 分の後、 96°C、 1分、 60°C、 1分、 72 、 2分のサイクルを 40回繰り返し行った。 次にァガロースゲル電気泳動にて該 PCR反応産物を精製した後、 これを Zero Blunt T0P0 PCRクロ一ニングキット (Invi trogen社) の処方に従いプラスミドべ クタ一 pCR-Bl until- T0P0 (Invi trogen社) へサブクロ一ニングした。 これを大腸 菌 T0P10に導入し、 カナマイシンを含む LB寒天培地中で選択した。 個々のクロ一 ンの配列を解析した結果、 新規受容体蛋白質の一部をコ一ドする cDNAの塩基配列 (配列番号: 4) を得た。 参考例 3 Using human brain cDNA (CL0NTECH II MTC Panel I) as type II, PCR was carried out using two primers, primer 3 (SEQ ID NO: 5) and primer 4 (SEQ ID NO: 6). The composition of the reaction solution used in the reaction was the above-mentioned cDNA 11 as type III, and 2.5 U of Pfu Turbo DNA Polymerase (STRATAGENE), primer 3 (SEQ ID NO: 5) and primer 4 (SEQ ID NO: 6). 1 M each, 200 M dNTPs, and Add 25 1 of 2XGC Bufferl (Takara Shuzo) to make a volume of 50 ^ 1. After 96 minutes, the PCR reaction was repeated 40 times at 96 ° C, 1 minute, 60 ° C, 1 minute, 72 minutes and 2 minutes. Next, after purifying the PCR reaction product by agarose gel electrophoresis, the PCR product is transferred to a plasmid vector pCR-Bl until-T0P0 (Invitrogen) according to the instructions of Zero Blunt T0P0 PCR Cloning Kit (Invitrogen). Sub-cloned. This was introduced into E. coli T0P10 and selected on LB agar medium containing kanamycin. As a result of analyzing the sequence of each clone, a nucleotide sequence (SEQ ID NO: 4) of a cDNA encoding a part of the novel receptor protein was obtained. Reference example 3
ヒ卜脳由来新規受容体蛋白質をコードする cDNAの 3'部分塩基配列の決定 Determination of 3 'partial nucleotide sequence of cDNA encoding novel receptor protein from human brain
ヒト脳 cDNA (CL0NTECH社 MTC Panell) を铸型とし、 2個のプライマー、 プライ マー 5 (配列番号: 8) およびプライマ一 6 (配列番号: 9) を用いて PCR反応 を行った。 該反応における反応液の組成は上記 cDNA l lを铸型として使用し、 Pfu Turbo DNA Polymerase (STRATAGENE社) 2.5U、 プライマ一 5 (配列番号: 8) およびプライマ一 6 (配列番号: 9) を各 1 M、 dNTPsを 200 XM、 および 2XGC Bufferl (宝酒造) を 加え、 50 1の液量とした。 PCR反応は、 96°C、 1 分の後、 96 、 1分、 60で、 1分、 72°C、 2分のサイクルを 40回繰り返し行った。 次に該 PCR反応産物 5 lを铸型として使用し、 Pfu Turbo DNA Polymerase  Using human brain cDNA (CL0NTECH MTC Panel1) as type I, PCR was performed using two primers, Primer 5 (SEQ ID NO: 8) and Primer 1 (SEQ ID NO: 9). The composition of the reaction solution used in the reaction was the above cDNA II as type III, and 2.5 U of Pfu Turbo DNA Polymerase (STRATAGENE), Primer 5 (SEQ ID NO: 8) and Primer 6 (SEQ ID NO: 9) 1 M, dNTPs were added to 200 XM, and 2XGC Buffer1 (Takara Shuzo) to make a liquid volume of 501. After 1 minute at 96 ° C, the PCR reaction was repeated 40 times at 96, 1 minute, and 60 times for 1 minute, 72 ° C, and 2 minutes. Next, 5 l of the PCR reaction product was used as type 、, and Pfu Turbo DNA Polymerase was used.
(STRATAGENE社) 2.5U、 プライマ一 5 (配列番号: 8) およびプライマ一 7 (配 列番号: 10) を各 1 M、 dNTPsを 200 M、 および 2XGC Bufferl (宝酒造) を 25 l加え、 50 1の液量とした。 PCR反応は、 96 、 1分の後、 96°C、 1分、 60 、 1 分、 72°C、 2分のサイクルを 45回繰り返し行った。 次にァガロースゲル電気泳動 にて該 PCR反応産物を精製した後、 これを Zero Blunt T0P0 PCRクローニングキッ ト (Invitrogen社) の処方に従いプラスミドベクター pCR- BluntII-T0P0  (STRATAGENE) Add 2.5 U, Primer-5 (SEQ ID NO: 8) and Primer-7 (SEQ ID NO: 10) at 1 M each, dNTPs at 200 M, and 25 l of 2XGC Bufferl (Takara Shuzo), and add 50 1 Liquid volume. The PCR reaction was repeated 45 times at 96 ° C., 1 minute, 60 ° C., 1 minute, 72 ° C., 2 minutes after 96 minutes. Next, the PCR reaction product was purified by agarose gel electrophoresis, and then purified using the plasmid vector pCR-BluntII-T0P0 according to the procedure of Zero Blunt T0P0 PCR Cloning Kit (Invitrogen).
(Invi trogen社) へサブクロ一ニングした。 これを大腸菌 T0P10に導入し、 カナ マイシンを含む LB寒天培地中で選択した。 個々のクローンの配列を解析した結果、 新規受容体蛋白質の一部をコードする cDNAの塩基配列 (配列番号: 7) を得た。 参考例 4 (Invitrogen). This was introduced into E. coli T0P10 and selected on LB agar medium containing kanamycin. As a result of analyzing the sequence of each clone, the nucleotide sequence (SEQ ID NO: 7) of cDNA encoding a part of the novel receptor protein was obtained. Reference example 4
ヒ卜脳由来新規受容体蛋白質をコードする全長 cMAの塩基配列の決定 Determination of the nucleotide sequence of full-length cMA encoding a novel receptor protein from human brain
参考例 1、 参考例 2および参考例 3により得られた新規受容体蛋白質をコード する部分 cDNAの塩基配列 (配列番号: 1、 配列番号: 4および配列番号: 7 ) を 基に全長 cDNAの塩基配列 (配列番号: 1 2 ) を決定した。 配列番号: 1 2は配列 番号: 1の 71番目から 789番目までの配列、 配列番号: 4の 284番目から 1134番目 までの配列および配列番号: 7の 473番目から 1926番目までの配列に該当する。 全長 cDNAの塩基配列 (配列番号: 1 2 ) から予想される蛋白質のアミノ酸配列は 配列番号: 1 1に示す。 配列番号: 1 1で表されるアミノ酸配列を含有する蛋白 質を NEPHA (novel EphA) と命名した。 NEPHAとヒト Ephr in受容体 EphA7とは、 ァ ミノ酸レベルで 50. 7%の相同性を有する。 全長をコードする遺伝子は参考例 1、 参考例 2および参考例 3で得た cDNA断片を適当な制限酵素により切断後、 T4リガ ーゼなどで結合することにより取得する。 あるいは、 参考例 1および参考例 3で 明らかにした塩基配列を基に全長をコードする cDNAを増幅するようなプライマー を設計し PCR法により取得する。 参考例 5  Based on the nucleotide sequence of the partial cDNA encoding the novel receptor protein obtained in Reference Example 1, Reference Example 2 and Reference Example 3 (SEQ ID NO: 1, SEQ ID NO: 4 and SEQ ID NO: 7), The sequence (SEQ ID NO: 12) was determined. SEQ ID NO: 12 corresponds to the 71st to 789th sequence of SEQ ID NO: 1, the 284th to 1134th sequence of SEQ ID NO: 4 and the 473th to 1926th sequence of SEQ ID NO: 7 . The amino acid sequence of the protein predicted from the nucleotide sequence of the full-length cDNA (SEQ ID NO: 12) is shown in SEQ ID NO: 11. The protein containing the amino acid sequence represented by SEQ ID NO: 11 was named NEPHA (novel EphA). NEPHA and the human Ephr in receptor EphA7 have 50.7% homology at the amino acid level. The gene encoding the full length is obtained by cleaving the cDNA fragments obtained in Reference Examples 1, 2 and 3 with an appropriate restriction enzyme and ligating them with T4 ligase or the like. Alternatively, a primer is designed to amplify the cDNA encoding the full length based on the nucleotide sequences identified in Reference Examples 1 and 3, and the primers are obtained by PCR. Reference example 5
ヒト脳由来新規受容体蛋白質をコ一ドする全長 cDNAの塩基配列を有するプラスミ ドの作製 Construction of a plasmid having a full-length cDNA sequence encoding a novel receptor protein derived from human brain
参考例 2および参考例 3により得られた cDNAの塩基配列 (配列番号: 4および 配列番号: 7 ) を含むプラスミドを制限酵素により切断した。 反応液の組成は配 列番号: 4で表される cDNAの塩基配列を含むプラスミドもしくは配列番号: 7で 表される cDNAの塩基配列を含むプラスミドを l g、 X ol (宝酒造) 5U、 および ΙΟχΗ Buf fer (宝酒造) 5 x Iを加え 50 1の液量とした。 制限酵素反応は 37°C、 1 時間にて行い、 さらに該反応産物に Bs tEI I (宝酒造) 5Uを加え 60°C、 1時間の反 応を行った。 次に配列番号: 4で表される cMAの塩基配列を含むプラスミドから は 941bpおよび配列番号: 7で表される cDNAの塩基配列を含むプラスミドからは 5273bpに相当する該反応産物をァガロース電気泳動にて精製した後、 これらを DNA L igat i on Ki t Ver. 2 (宝酒造) の処方に従い結合させた。 これを大腸菌 ΤΟΡίΟに導入し、 カナマイシンを含む LB寒天培地中で選択した。 個々のクローン の配列を解析した結果、 配列番号: 13で表される cDNAの塩基配列を得た。 さら に、 参考例 1により得られた cDNAの塩基配列 (配列番号: 1) を含むプラスミド および配列番号: 1 3で表される cDNAの塩基配列を含むプラスミドを制限酵素に より切断した。 反応液の組成は配列番号: 1で表される cDNAの塩基配列を含むプ ラスミドもしくは配列番号: 13で表される cDNAの塩基配列を含むプラスミドを lfig、 Pvul (宝酒造) 10U、 および ΙΟχΚ Buffer (宝酒造) を加え 50〃1の液量と した。 制限酵素反応は 37° (、 1時間にて行った。 次に配列番号: 1で表される cDNAの塩基配列を含むプラスミドからは 743bpおよび配列番号: 13で表される cDNAの塩基配列を含むプラスミドからは 5977bpに相当する該反応産物をァガ口一 ス電気泳動にて精製した後、 これらを DNA Ligation Kit Ver.2 (宝酒造) の処方 に従い結合させた。 これを大腸菌 TOP10に導入し、 カナマイシンを含む LB寒天培 地中で選択した。 個々のクローンの配列を解析した結果、 ヒト脳由来新規受容体 蛋白質をコードする全長 cDNAの塩基配列 (配列番号: 12) を有するプラスミド を得た。 ここで得られた配列番号: 12を有するプラスミドを PTB2230と命名し, 得られた形質転換体を大腸菌 (Escherichia coli) TOP10/pTB2230と命名した。 実施例 1 A plasmid containing the nucleotide sequences of the cDNAs obtained in Reference Examples 2 and 3 (SEQ ID NO: 4 and SEQ ID NO: 7) was digested with restriction enzymes. The composition of the reaction mixture was lg, Xol (Takara Shuzo) 5U, and 含 む Buf, containing a plasmid containing the nucleotide sequence of the cDNA represented by SEQ ID NO: 4 or a plasmid containing the nucleotide sequence of the cDNA represented by SEQ ID NO: 7. fer (Takara Shuzo) 5 x I was added to make a liquid volume of 501. The restriction enzyme reaction was performed at 37 ° C. for 1 hour, and 5 U of BstEI I (Takara Shuzo) was added to the reaction product, followed by a reaction at 60 ° C. for 1 hour. Next, the reaction product corresponding to 941 bp from the plasmid containing the nucleotide sequence of cMA represented by SEQ ID NO: 4 and 5273 bp from the plasmid containing the nucleotide sequence of cDNA represented by SEQ ID NO: 7 was subjected to agarose electrophoresis. After purification, they were bound according to the prescription of DNA Ligation Kit Ver. 2 (Takara Shuzo). This is E. coli And selected in LB agar medium containing kanamycin. As a result of analyzing the sequence of each clone, the nucleotide sequence of cDNA represented by SEQ ID NO: 13 was obtained. Further, a plasmid containing the nucleotide sequence of the cDNA obtained in Reference Example 1 (SEQ ID NO: 1) and a plasmid containing the nucleotide sequence of the cDNA represented by SEQ ID NO: 13 were digested with restriction enzymes. The composition of the reaction solution is lfig, Pvul (Takara Shuzo) 10 U, and plasmid containing the nucleotide sequence of the cDNA represented by SEQ ID NO: 1 or plasmid containing the nucleotide sequence of the cDNA represented by SEQ ID NO: 13. (Takara Shuzo) to make a liquid volume of 50〃1. The restriction enzyme reaction was performed at 37 ° (1 hour. Next, the plasmid containing the nucleotide sequence of the cDNA represented by SEQ ID NO: 1 contains 743 bp and the nucleotide sequence of the cDNA represented by SEQ ID NO: 13 The reaction product corresponding to 5977 bp was purified from the plasmid by agarose gel electrophoresis, and these were ligated according to the prescription of DNA Ligation Kit Ver.2 (Takara Shuzo). Selection was performed in LB agar medium containing kanamycin As a result of analyzing the sequence of each clone, a plasmid having the nucleotide sequence of the full-length cDNA encoding the novel human brain-derived receptor protein (SEQ ID NO: 12) was obtained. The resulting plasmid having SEQ ID NO: 12 was named PTB2230, and the resulting transformant was named Escherichia coli TOP10 / pTB2230.
NEPHA遺伝子のアポ 1 シスに及ぼす影響を解析するために、 アンチセンスオリ ゴヌクレオチド導入実験を行つた。  In order to analyze the effect of the NEPHA gene on apo1 cis, an antisense oligonucleotide introduction experiment was performed.
まず、 配列番号: 12で表される塩基配列に対するアンチセンス (配列番号: 13および配列番号: 14) を設計後、 phosphorothioate化オリゴヌクレオチド を合成し、 HPLC精製して導入実験に用いた。 コントロールオリゴヌクレオチドと しては、 配列番号: 13で示される塩基配列のリバース配列 (配列番号: 1 5) および配列番号: 14で示される塩基配列のリバース配列 (配列番号: 1 6) を, 同様に phosphorothioate化し、 HPLC精製して用いた。  First, after designing an antisense (SEQ ID NO: 13 and SEQ ID NO: 14) for the base sequence represented by SEQ ID NO: 12, a phosphorothioated oligonucleotide was synthesized, purified by HPLC, and used for an introduction experiment. As control oligonucleotides, the reverse sequence of the base sequence represented by SEQ ID NO: 13 (SEQ ID NO: 15) and the reverse sequence of the base sequence represented by SEQ ID NO: 14 (SEQ ID NO: 16) were similarly used. And purified by HPLC for use.
被験細胞として乳がん細胞株 ZR- 75-1 (Cancer Res、 38(10)巻、 3352 - 3364頁、 1978年、 ATCCより購入) を用い、 オリゴヌクレオチド導入前日に 2x10個の細胞を 24ゥエルプレー卜 (Falcon社製) に播種した。 オリゴヌクレオチド導入には、 OligofectAMINE (Invi trogen社製) を使用し、 そのプロトコ一ルに従った。 導入 後 20時間で RNeasy mini kit (Qiagen社製) のプロトコ一ルに従って RNAを抽出し、 TaqManRTキット (Perkin- Elmer社製) を用いて cDNAを調製した。 また、 同時に逆 転写酵素を添加せずに同様の反応を行い、 非逆転写コン卜ロールとした。 As test cells, breast cancer cell line ZR-75-1 (Cancer Res, 38 (10), 3352-3364, purchased from ATCC, 1978) was used. Falcon). For oligonucleotide introduction, OligofectAMINE (Invitrogen) was used and the protocol was followed. Twenty hours after the introduction, RNA was extracted according to the protocol of the RNeasy mini kit (Qiagen), and cDNA was prepared using the TaqManRT kit (Perkin-Elmer). At the same time, the same reaction was carried out without adding reverse transcriptase to obtain a non-reverse transcription control.
2種のプライマ一 (配列番号: 17および配列番号: 1 8) 、 およびプローブ (配列番号: 19)を用いて NEPHA遺伝子の発現量を、 また補正のために TaqMan /3-actin Control Reagents (Perkin- Elmer社製) を用いて i3ァクチン発現量を 見ることとし、 ABI 7900 (Perkin- Elmer社製) のマニュアルに従い PCRを行った c NEPHA遺伝子の発現量は、 cDNAを用いて得られたデータから非逆転写コントロー ルを用いて得られたデータを差し引いた後、 i3ァクチン発現量で補正して比較し た。 Using two primers (SEQ ID NO: 17 and SEQ ID NO: 18) and a probe (SEQ ID NO: 19), the expression level of the NEPHA gene was corrected, and TaqMan / 3-actin Control Reagents (Perkin - using Elmer Co., Ltd.) and watching i3 Akuchin expression level, expression level of c NEPHA gene PCR was performed according to the manual of ABI 7900 (Perkin Elmer Inc.) from the data obtained using the cDNA After subtracting the data obtained using the non-reverse transcription control, the results were corrected for i3 actin expression and compared.
一方、 アンチセンスオリゴヌクレオチドを導入して NEPHA遺伝子の発現を抑え た際のアポトーシスに及ぼす影響を見るために、 アンチセンスオリゴヌクレオチ ド導入後 3日目に、 Cell death detection ELISA (Roche社製) を用いてコント口 —ルオリゴヌクレオチド導入サンプルと比較した。  On the other hand, on the third day after the introduction of the antisense oligonucleotide, a Cell death detection ELISA (Roche) was used to examine the effect on the apoptosis when the expression of the NEPHA gene was suppressed by introducing the antisense oligonucleotide. The control oligonucleotide was used to compare with the sample introduced with the oligonucleotide.
その結果、 NEPHA遺伝子に対するアンチセンス (配列番号: 13) を用いた場 合、 アンチセンスオリゴヌクレオチド導入後 20時間で、 そのコント口一ルオリゴ ヌクレオチド (配列番号: 1 5) に比べ、 NEPHA遺伝子の発現量は 53%に減少し ており、 アンチセンス (配列番号: 14) を用いた場合も、 そのコントロールォ リゴヌクレオチド (配列番号: 16) に比べ、 NEPHA遺伝子の発現量は 57%に減 少していた。  As a result, when the antisense to the NEPHA gene (SEQ ID NO: 13) was used, the expression of the NEPHA gene was 20 hours after the introduction of the antisense oligonucleotide compared to the control oligonucleotide (SEQ ID NO: 15). The amount of expression of the NEPHA gene was reduced to 57% when the antisense (SEQ ID NO: 14) was used, compared to the control oligonucleotide (SEQ ID NO: 16). Was.
また、 アンチセンスオリゴヌクレオチド導入後 3日目で、 アポト一シスは、 コ ントロールオリゴヌクレオチド (配列番号: 15) を 100%とするとアンチセン スオリゴヌクレオチド (配列番号: 13) 導入時には 224%に、 コントロールォ リゴヌクレオチド (配列番号: 16) を 100%とするとアンチセンスオリゴヌク レオチド (配列番号: 14) 導入時には 185%に増加していた。  On the third day after the introduction of the antisense oligonucleotide, apoptosis was calculated to be 224% when the control oligonucleotide (SEQ ID NO: 13) was introduced and the control oligonucleotide (SEQ ID NO: 13) was assumed to be 100%. Assuming that the oligonucleotide (SEQ ID NO: 16) was 100%, it was increased to 185% when the antisense oligonucleotide (SEQ ID NO: 14) was introduced.
これらのことから、 がん細胞に発現している NEPHA遺伝子の発現を抑制するこ とにより、 がん細胞がアポトーシスを起こすことが確認され、 NEPHA遺伝子がが ん細胞の増殖やアポト一シスに関与していることが明らかとなった。 実施例 2 These results confirm that suppressing the expression of the NEPHA gene expressed in cancer cells causes cancer cells to undergo apoptosis, and that the NEPHA gene is involved in cancer cell proliferation and apoptosis. It became clear that we were doing. Example 2
NEPHAの 5,上流域の解析  NEPHA 5, upstream analysis
配列番号: 1で表される塩基配列を用いて、 NCBIのヒトゲノム配列に対して BLAST検索を行ったところ、 配列番号: 1の 1番から 176番の塩基配列がヒト 1番 染色体のゲノム配列 (ACCESSION番号は NT#004568、 VERSIONは ΝΤ#004568· 8  When a BLAST search was performed on the NCBI human genomic sequence using the nucleotide sequence represented by SEQ ID NO: 1, the nucleotide sequence from No. 1 to 176 of SEQ ID NO: 1 was the genomic sequence of human chromosome 1 ( ACCESSION number is NT # 004568, VERSION is ΝΤ # 004568 · 8
(GI: 18548377) 、 DEFINITIONは Homo sapiens chromosome 1 working draft sequence segment) と一致した。 そこで、 このゲノム配列から NEPHAの開始コド ンの 5'上流 (約 2Kb) のゲノム配列 (配列番号: 20) を取得し、 転写因子予測ソ フト TRANFACの Matrix version 1.3にて転写因子結合領域を予想した。 その結果、 表 1に示すように、 各種の転写因子の結合位置が予測され、 この領域が NEPHAの 転写調節領域であることが示唆された。  (GI: 18548377), DEFINITION was consistent with Homo sapiens chromosome 1 working draft sequence segment). Therefore, the genomic sequence (SEQ ID NO: 20) 5 'upstream (about 2 Kb) of the NEPHA start codon was obtained from this genomic sequence, and the transcription factor binding region was predicted using the transcription factor prediction software Matrix version 1.3 of TRANFAC. did. As a result, as shown in Table 1, the binding positions of various transcription factors were predicted, suggesting that this region is a transcriptional regulatory region of NEPHA.
〔表 1〕 ス卜 コア マ卜リック 認識配列 転写因子 [Table 1] Storage core matrix recognition sequence Transcription factor
マッチ スマッチ  Match match
4 (+) 0.993 0.97 tcagAGGAAgtct c-Ets-l(p54)  4 (+) 0.993 0.97 tcagAGGAAgtct c-Ets-l (p54)
5 (+) 1 0.924 cagAGGAAgtctgt c-Ets-2  5 (+) 1 0.924 cagAGGAAgtctgt c-Ets-2
6 ω 1 0.95 agAGGAAgtctgtat c-Ets-1  6 ω 1 0.95 agAGGAAgtctgtat c-Ets-1
7 (+) 0.837 0.785 gaGGAAGtct Ncx  7 (+) 0.837 0.785 gaGGAAGtct Ncx
14 (-) 0.885 0.913 tctgtATTTTgttt FAC1  14 (-) 0.885 0.913 tctgtATTTTgtgttt FAC1
14 (-) 1 0.937 tctGTATTttgttt MRF-2  14 (-) 1 0.937 tctGTATTttgttt MRF-2
16 (-) 0.926 0.865 tgtatTTTGTttta FAC1  16 (-) 0.926 0.865 tgtatTTTGTttta FAC1
18 (-) 0.966 0.961 tatTTTGTttta SRY  18 (-) 0.966 0.961 tatTTTGTttta SRY
18 (-) 0.842 0.812 tATTTTgttttagca Evi-1  18 (-) 0.842 0.812 tATTTTgttttagca Evi-1
19 (-) 0.978 0.868 attttGTTTTagca FAC1  19 (-) 0.978 0.868 attttGTTTTagca FAC1
24 (-) 0.859 0.89 gttttaGCACCga C/EBP  24 (-) 0.859 0.89 gttttaGCACCga C / EBP
44 (+) 0.777 0.725 aacctctgGGCTGaagtc Pax-8  44 (+) 0.777 0.725 aacctctgGGCTGaagtc Pax-8
67 (-) 0.94 0.936 agcttcaGTAAGaa C/EBPalpha  67 (-) 0.94 0.936 agcttcaGTAAGaa C / EBPalpha
72 (+) 0.993 0.903 caGTAAGaag Ncx 74 (-) 0.893 0.766 gtaagaagccATGATtggt Pax- 272 (+) 0.993 0.903 caGTAAGaag Ncx 74 (-) 0.893 0.766 gtaagaagccATGATtggt Pax- 2
83 (+) 0.997 0.991 catGATTGgt GATA-183 (+) 0.997 0.991 catGATTGgt GATA-1
84 (-) 1 0.961 atgATTGGtgc NF-Y84 (-) 1 0.961 atgATTGGtgc NF-Y
93 (+) 0.908 0.752 gccttagaGCCTGctaag Pax-893 (+) 0.908 0.752 gccttagaGCCTGctaag Pax-8
104 (+) 0.836 0.803 tgCTAAGggg Ncx104 (+) 0.836 0.803 tgCTAAGggg Ncx
110 (+) 1 0.99 gggGATGGaa GATA-1110 (+) 1 0.99 gggGATGGaa GATA-1
127 (+) 0.905 0.761 ttaaagagGAGTGagggg Pax-8127 (+) 0.905 0.761 ttaaagagGAGTGagggg Pax-8
129 (+) 0.897 0.902 aaagaggagTGAGG GKLF129 (+) 0.897 0.902 aaagaggagTGAGG GKLF
130 (+) 1 0.977 aagaGGAGTgaggg p300130 (+) 1 0.977 aagaGGAGTgaggg p300
131 (+) 0.981 0.887 agaggagtgAGGGG GKLF131 (+) 0.981 0.887 agaggagtgAGGGG GKLF
133 (+) 0.796 0.736 aggaGTGAGgggtggtgga Pax- 2133 (+) 0.796 0.736 aggaGTGAGgggtggtgga Pax- 2
133 ω 0.881 0.852 aggagTGAGGgg t gg t ggag t Pax- 4133 ω 0.881 0.852 aggagTGAGGgg t gg t ggag t Pax-4
134 (+) 0.87 0.736 ggagtgagGGGTGgtgga Pax-8134 (+) 0.87 0.736 ggagtgagGGGTGgtgga Pax-8
135 (+) 1 0.893 gagtgAGGGGtgg MZF1135 (+) 1 0.893 gagtgAGGGGtgg MZF1
141 (-) 1 0.983 gGGGTGgtggag Pax-4141 (-) 1 0.983 gGGGTGgtggag Pax-4
142 (+) 0.905 0.729 gggtggtgGAGTGaggac Pax-8142 (+) 0.905 0.729 gggtggtgGAGTGaggac Pax-8
1 4 (-) 0.903 0.893 gTGGTGgagtga Pax-41 4 (-) 0.903 0.893 gTGGTGgagtga Pax-4
145 (+) 1 0.952 tggtGGAGTgagga p300145 (+) 1 0.952 tggtGGAGTgagga p300
148 (+) 0.796 0.732 tggaGTGAGgactcatttt Pax - 2148 (+) 0.796 0.732 tggaGTGAGgactcatttt Pax-2
155 (+) 0.918 0.918 aggaCTCATtttgaaat YY1155 (+) 0.918 0.918 aggaCTCATtttgaaat YY1
160 (+) 0.996 0.975 tcattttGAAATga C/EBPbeta160 (+) 0.996 0.975 tcattttGAAATga C / EBPbeta
160 (-) 0.929 0.936 tcattttGAAATga C/EBPalpha160 (-) 0.929 0.936 tcattttGAAATga C / EBPalpha
172 (+) 1 0.95 gaaaGGAGTcctgg p300172 (+) 1 0.95 gaaaGGAGTcctgg p300
175 (+) 0.825 0.73 aggaGTCCTgggtctctaa Pax - 2175 (+) 0.825 0.73 aggaGTCCTgggtctctaa Pax-2
194 (+) 0.807 gcagcttctaaAAATAccatct MEF-2194 (+) 0.807 gcagcttctaaAAATAccatct MEF-2
194 (+) 0.898 gcagcttctaaAAATAccatct MEF-2194 (+) 0.898 gcagcttctaaAAATAccatct MEF-2
197 (+) 0.902 gcttcTAAAAataccatc aMEF-2197 (+) 0.902 gcttcTAAAAataccatc aMEF-2
205 (+) 0.949 aaatACCATctcccaga YY1205 (+) 0.949 aaatACCATctcccaga YY1
208 (-) 0.992 taCCATCtcc GATA-1208 (-) 0.992 taCCATCtcc GATA-1
220 (-) 0.903 0.897 gAGGTGgaggca Pax-4220 (-) 0.903 0.897 g AGGTGgaggca Pax-4
226 (+) 1 0.869 gaggcAGGGGgag MZF1 227 (-) 0.992 0.888 aggcaGGGGGag AP-2226 (+) 1 0.869 gaggcAGGGGgag MZF1 227 (-) 0.992 0.888 aggcaGGGGGag AP-2
231 (+) 0.927 0.924 agggGGAGGggag Spl231 (+) 0.927 0.924 agggGGAGGggag Spl
231 (+) 0.956 0.911 agggGGAGGggaga GC box231 (+) 0.956 0.911 agggGGAGGggaga GC box
232 (+) 1 0.931 gggggAGGGGaga MZF1232 (+) 1 0.931 gggggAGGGGaga MZF1
232 (+) 0.935 0.939 gggggAGGGGaga AZR232 (+) 0.935 0.939 gggggAGGGGaga AZR
242 (+) 0.758 0.795 agaaaTCGAGcgtgaggagaa Pax- 4242 (+) 0.758 0.795 agaaaTCGAGcgtgaggagaa Pax- 4
242 (+) 0.715 0.782 agaaATCGAgc gt gaggag Pax - 2242 (+) 0.715 0.782 agaaATCGAgc gt gaggag Pax-2
243 (-) 0.84 0.798 gaaatcgagcGTGAGgaga Pax- 2243 (-) 0.84 0.798 gaaatcgagcGTGAGgaga Pax- 2
243 (+) 1 0.862 gaaatcgaGCGTGaggag Pax- 8243 (+) 1 0.862 gaaatcgaGCGTGaggag Pax- 8
243 (-) 0.75 0.713 gaaatCGAGCgt gaggag Pax- 8243 (-) 0.75 0.713 gaaatCGAGCgt gaggag Pax- 8
244 (-) 1 0.864 aaa t cgagCGTGAggagaatg Pax- 3244 (-) 1 0.864 aaa t cgagCGTGAggagaatg Pax- 3
252 (+) 0.868 0.73 cgtgaggaGAATGaagat Pax - 8252 (+) 0.868 0.73 cgtgaggaGAATGaagat Pax-8
254 (+) 0.952 0.872 t gaggagaATGAAg t ggaaagg OCT- 1254 (+) 0.952 0.872 t gaggagaATGAAg t ggaaagg OCT- 1
259 (+) 0.673 0.707 agaatgaaGATGGaaagg Pax - 8259 (+) 0.673 0.707 agaatgaaGATGGaaagg Pax-8
260 (+) 0.948 0.86 gaa t gaagATGGAaaggagg t gg OCT - 1260 (+) 0.948 0.86 gaa t gaagATGGAaaggagg t gg OCT-1
260 (-) 1 0.974 gaatgaagATGGAaagg YY1260 (-) 1 0.974 gaatgaagATGGAaagg YY1
264 (+) 1 0.985 gaaGATGGaa GATA-1264 (+) 1 0.985 gaaGATG Gaa GATA-1
264 (+) 0.888 0.876 gaagatGGAAAggag OCT-1264 (+) 0.888 0.876 gaagatGGAAAggag OCT-1
265 (+) 0.852 0.867 aaGATGGaaagga C/EBP265 (+) 0.852 0.867 aaGATGGaaagga C / EBP
268 (+) 0.979 0.964 atggAAAGGaggtgg Barbie Box268 (+) 0.979 0.964 atggAAAGGaggtgg Barbie Box
276 (-) 0.903 0.897 gAGGTGgcagtg Pax - 4276 (-) 0.903 0.897 gAGGTGgcagtg Pax-4
277 (+) 0.846 0.719 aggtggcaGTGTGagaac Pax-8277 (+) 0.846 0.719 aggtggcaGTGTGagaac Pax-8
278 (+) 0.944 0.937 ggtgGCAGTgtgag p300278 (+) 0.944 0.937 ggtgGCAGTgtgag p300
284 (+) 0.94 0.909 agTGTGAgaaccc C/EBP284 (+) 0.94 0.909 agTGTGAgaaccc C / EBP
296 (+) 0.81 0.738 cttcCTCAGgtaggtctgg Pax-2296 (+) 0.81 0.738 cttcCTCAGgtaggtctgg Pax-2
299 (-) 1 0.965 cctCAGGTaggtc AREB6299 (-) 1 0.965 cctCAGGTaggtc AREB6
303 (+) 1 0.949 aggtaggTCTGGtttt Handl/E47303 (+) 1 0.949 aggtaggTCTGGtttt Handl / E47
305 (+) 0.822 0.777 gtagGTCTGgttttagaag Pax-2305 (+) 0.822 0.777 gtagGTCTGgttttagaag Pax-2
319 (+) 0.833 0.744 agaaGTCCCgcctgcatcc Pax-2319 (+) 0.833 0.744 agaaGTCCCgcctgcatcc Pax-2
319 (-) 0.984 0.931 agaagtcCCGCCtgc Spzl319 (-) 0.984 0.931 agaagtcCCGCCtgc Spzl
320 (+) 0.908 0.726 gaagtcccGCCTGcatcc Pax-8 b b o 〇 320 (+) 0.908 0.726 gaagtcccGCCTGcatcc Pax-8 bbo 〇
Figure imgf000046_0001
Figure imgf000046_0001
532 (-) 1 0.926 cacaagcACACAcgca Poly A 534 (-) 0.986 0.942 caaGCACAcacgcagaaa! GR 537 (-) 1 0.857 gcacaCACGCagaaatgg Pax - 8 543 (-) 0.998 0.937 acgcagaaATGGCacat YY1 548 (-) 0.857 0.782 gaaat ggc acATGAAagcg Pax- 2 556 (+) 0.86 0.709 acatgaaaGCGGGttgta Pax- 8 566 (+) 0.872 0.88 ggGTTGTaaatgg C/EBP 567 (-) 0.998 0.963 ggttgtaaATGGCcagt YY1 569 (+) 0.969 0.948 ttGTAAAtgg Ncx 572 (-) 0.902 0.733 taaatggccaGTGATctac Pax-2 586 (+) 0.75 0.707 tctacgatGCCAGgaatc Pax-8 604 (-) 0.993 0.9 ctcCTTACac Ncx 630 (+) 1 0.935 tatggacTCTGGcaga Handl/E47 642 (+) 1 0.709 cagagacaGCGTGgttca Pax-8 663 (-) 0.993 0.985 ggGCATCcgc GATA-1 671 (-) 0.986 0.951 gc aGCACAgagc at ggc gc GR 673 (+) 0.963 0.752 age ac agaGCATGgc gcc Pax-8 675 (-) 0.998 0.908 cacagagcATGGCgccc YY1 690 (+) 0.777 0.708 ccagtgagGGCTGggtag Pax-8 702 (+) 0.87 0.728 gggtagagGGGTGaggga Pax-8 703 (+) 1 0.916 ggtagAGGGGtga MZF1 706 (+) 0.765 0.718 agaggggtGAGGGataca Pax-8 709 (-) 1 0.971 gGGGTGagggat Pax - 4 710 (+) 0.844 0.847 gggtgAGGGAtac MZF1 711 (+) 0.847 0.822 ggtgagggATACAtatatgcaaa OCT - 1 719 (+) 1 0.936 atacatatATGCAaaagcacaga OCT-1 723 (-) 0.972 0.941 atatatGCAAAag C/EBP 723 (+) 1 0.892 atatatGCAAAagca OCT-1 724 (+) 0.917 0.925 taTATGCaaaagc C/EBP 732 (-) 0.986 0.938 aaaGCACAgacac tcagac GR 741 (一) 0.753 0.706 acactCAGACcgcagggg Pax-8 CO to t 532 (-) 1 0.926 cacaagcACACAcgca Poly A 534 (-) 0.986 0.942 caaGCACAcacgcagaaa! GR 537 (-) 1 0.857 gcacaCACGCagaaatgg Pax-8 543 (-) 0.998 0.937 acgcagaaATGGCacat YY1 548 (-) 0.857 0.782 gGAtagcagGAcAgcAgAgAgcAgAgC +) 0.86 0.709 acatgaaaGCGGGttgta Pax- 8 566 (+) 0.872 0.88 ggGTTGTaaatgg C / EBP 567 (-) 0.998 0.963 ggttgtaaATGGCcagt YY1 569 (+) 0.969 0.948 ttGTAAAtgg Ncx 572 (-) 0.902 0.733 taaatggccaTG7 0.75 tctacgatGCCAGgaatc Pax-8 604 (-) 0.993 0.9 ctcCTTACac Ncx 630 (+) 1 0.935 tatggacTCTGGcaga Handl / E47 642 (+) 1 0.709 cagagacaGCGTGgttca Pax-8 663 (-) 0.993 0.985 ggGCATCcgc GATA-1671g (GATA-1671g) at ggc gc GR 673 (+) 0.963 0.752 age ac agaGCATGgc gcc Pax-8 675 (-) 0.998 0.908 cacagagcATGGCgccc YY1 690 (+) 0.777 0.708 ccagtgagGGCTGggtag Pax-8 702 (+) 0.87 0.728 gggtagagGGGTGaggga Pax-8 0.916 ggtagAGGGGtga MZF1 706 (+) 0.765 0.718 agaggggtGAGGGataca Pax-8 709 (-) 1 0.971 gGGGTGagggat Pax-4 710 (+) 0.844 0.847 gggtgA GGGAtac MZF1 711 (+) 0.847 0.822 ggtgagggATACAtatatgcaaa OCT-1 719 (+) 1 0.936 atacatatATGCAaaagcacaga OCT-1 723 (-) 0.972 0.941 atatatGCAAAag C / EBP 723 (+) 1 0.892 atatatGCAAAagca OCT-1719c TACTA 0.99 Tg / EBP 732 (-) 0.986 0.938 aaaGCACAgacac tcagac GR 741 (one) 0.753 0.706 acactCAGACcgcagggg Pax-8 CO to t
o o  o o
oo  oo
' tO  '' tO
oo oo
—4 oo oo —4 oo oo
Figure imgf000048_0001
Figure imgf000048_0001
•■0 no p Szl  • ■ 0 no p Szl
GC b GC b
p Sl 圈 p Sl circle
i OAV; i OAV;
Figure imgf000049_0001
Figure imgf000049_0001
CO  CO
O O OO O O
O O
1155 U. ( aggggaacccしし し [c ig Γ d-Λ L1155 U. (aggggaaccc, then [c ig Γ d-Λ L
1 C Π QQ7 1 C Π QQ7
llo Π9 -) 0.897 C t C I g¾bし rAC L し/ DDIllo Π9-) 0.897 C t C I g¾b then rAC L then / DDI
11
Figure imgf000050_0001
11
Figure imgf000050_0001
η71η 7 1
11.808 o 0.989 u. yio atguLrlLr l I Ig rrtg rraag π· ς)L丄 11.808 o 0.989 u.yio atguLrlLr l I Ig rrtg rraag π) 丄) L 丄
 ,
1189 1 u. yb i b llrgt tglg Γ dX 寸  1189 1 u.yb i b llrgt tglg Γ dX
1194 0.796 Π 70  1194 0.796 Π 70
U. 丄 ggt I ilrAijglglgCl C I Γαλ U. 丄 ggt I ilrAijglglgCl C IΓαλ
0.881 π 00 c 0.881 π 00 c
1194 ggL Igl bA IgtgCtgCllg P r vX— /1  1194 ggL Igl bA IgtgCtgCllg P r vX— / 1
ヽ r>  ヽ r>
1202 0.908 Γ ΤΤΡ†  1202 0.908 Γ ΤΤΡ †
U. ί DO r dX v o ヽ  U. ί DO r dX v o ヽ
1209 (-) 0.979 U. oU9 tgcttgtatcii し age a a v—  1209 (-) 0.979 U.oU9 tgcttgtatcii then age a a v—
1217 0.823 U. ί 4 tc t tgaca rし iiragcaa i d o 1217 0.823 U.ί 4 tc t tgaca r and iiragcaa i d o
 ヽ
1245 0.864 U. oy aACCGAtcgt CDP CR1  1245 0.864 U.oy aACCGAtcgt CDP CR1
/ ヽ  / ヽ
1247 、- 0.858 u. oy ccgaTCGTTc CDP CR1  1247, -0.858 u.oy ccgaTCGTTc CDP CR1
/,ヽ  / 、 ヽ
1249 1 gatcgttcATGCAaaaccgccac OCT - 1 1249 1 gatcgttcATGCAaaaccgccac OCT-1
1250 0.857 n n atcgTTCATgcaaaaccgc Pax-2 1250 0.857 n n atcgTTCATgcaaaaccgc Pax-2
( ヽ  (ヽ
1251 -) 0.72 Λ u.7Ωyo0 tcgi icatgcAAAACcgcc rax / fl π n 7  1251-) 0.72 Λ u.7Ωyo0 tcgi icatgcAAAACcgcc rax / fl π n 7
I25l 、- 0.963 tcgttCATGCaaaaccgc Pax-8  I25l, -0.963 tcgttCATGCaaaaccgc Pax-8
 ヽ
1252 一) 0.984 I).94o cgttcatGCAAAac C/EBP 1252 i) 0.984 I) .94o cgttcatGCAAAac C / EBP
1253 (+) 1 oyo gttcatGCAAAaccg OCT-1 o ヽ n no o 1253 (+) 1 oyo gttcatGCAAAaccg OCT-1 o ヽ n no o
1258 0.984 u. y o tgcaaaaCCGCCacc Spzl  1258 0.984 u.y o tgcaaaaCCGCCacc Spzl
 ヽ
1263 (+) 0.92 U.91b aaccgcCACCGc Pax - 4 、  1263 (+) 0.92 U.91b aaccgcCACCGc Pax-4,
1266 、- 1 u. y l cgccaCCGCCccca GC box  1266, -1 u.y l cgccaCCGCCccca GC box
( ヽ  (ヽ
l oT 1 Λ QQ7 gccaCCGCCccca Spl i ft i* ヽ l oT 1 Λ QQ7 gccaCCGCCccca Spl i ft i * ヽ
1276 1 cccaTCCCAgcg Ik - 2 o n r f ヽ Q  1276 1 cccaTCCCAgcg Ik-2 o n r f ヽ Q
1285 ) 1 n O gcgtccacATGGTttcc YY1 1285) 1 n O gcgtccacATGGTttcc YY1
1 o * 、 Π QQ/1 1 o *, Π QQ / 1
1286 K-) 0.938 U. yo4 cgtccACATGgttt USF ヽ  1286 K-) 0.938 U. yo4 cgtccACATGgttt USF ヽ
1286 (+) 0.938 u. yo cgtcCACATggttt USF ヽ Q Q 0 n QQ 0  1286 (+) 0.938 u.yo cgtcCACATggttt USF ヽ Q Q 0 n QQ 0
u. yo tgGTTTCccagcctcgg ' RFXl o n r ヽ QQ 1  u.yo tgGTTTCccagcctcgg 'RFXl on r ヽ QQ 1
1295 (-) 1 ggttTCCCAgcc Ik-2 1295 (-) 1 ggttTCCCAgcc Ik-2
1303 (+) 1 0.941 agcctCGGAAgcgc Elk-11303 (+) 1 0.941 agcctCGGAAgcgc Elk-1
1311 (-) 1 0.857 aaGCGCGttggta ZF51311 (-) 1 0.857 aaGCGCGttggta ZF5
1318 (+) 0.823 0.724 ttggtaggGCAGGacccc Pax-8 1333 (-) 0.765 0.707 cccgtCCCTCactttgct Pax - 81318 (+) 0.823 0.724 ttggtaggGCAGGacccc Pax-8 1333 (-) 0.765 0.707 cccgtCCCTCactttgct Pax-8
1333 (-) 0.945 0.901 CCCGTccctcactt GKLF 1333 (-) 0.945 0.901 CCCGTccctcactt GKLF
1353 (-) 0.978 0.964 ccttTCCCGagc Ik - 2 ' 1353 (-) 0.978 0.964 ccttTCCCGagc Ik-2 '
1371 (+) 0.995 0.992 cggGATCGtt GATA-11371 (+) 0.995 0.992 cggGATCGtt GATA-1
1381 (-) 1 0.87 cggCCCCTccttc MZF1 1381 (-) 1 0.87 cggCCCCTccttc MZF1
1383 (-) 1 0.929 gcccctccTTCCTgc c-Ets - 1 1383 (-) 1 0.929 gcccctccTTCCTgc c-Ets-1
1384 (-) 0.981 0.923 CCCCTccttcctgc GKLF 1384 (-) 0.981 0.923 CCCCTccttcctgc GKLF
1388 (-) 0.823 0.738 tccttCCTGCcgctgtgt Pax - 8 1388 (-) 0.823 0.738 tccttCCTGCcgctgtgt Pax-8
1434 (+) 0.846 0.705 gtgtgtgtGTGTGatgag Pax - 81434 (+) 0.846 0.705 gtgtgtgtGTGTGatgag Pax-8
1441 (+) 0.765 0.707 tgtgtgatGAGGGcggga Pax-81441 (+) 0.765 0.707 tgtgtgatGAGGGcggga Pax-8
1441 (+) 0.94 0.942 tgTGTGAtgaggg C/EBP1441 (+) 0.94 0.942 tgTGTGAtgaggg C / EBP
1445 (+) 0.86 0.716 tgatgaggGCGGGaagag Pax-81445 (+) 0.86 0.716 tgatgaggGCGGGaagag Pax-8
1448 (+) 1 0.928 tgagGGCGGgaag Spl 1448 (+) 1 0.928 tgagGGCGGgaag Spl
1449 (+) 0.984 0.937 gagGGCGGgaagaga Spzl  1449 (+) 0.984 0.937 gagGGCGGgaagaga Spzl
1454 (+) 0.968 0.891 cgggaagagAAAGG GKLF  1454 (+) 0.968 0.891 cgggaagagAAAGG GKLF
1455 (+) 1 0.907 gggaagagaAAGGG GKLF  1455 (+) 1 0.907 gggaagagaAAGGG GKLF
1456 (+) 0.981 0.92 ggaagagaaAGGGG GKLF  1456 (+) 0.981 0.92 ggaagagaaAGGGG GKLF
1457 (+) 0.946 0.925 gaagagaaaGGGGG GKLF  1457 (+) 0.946 0.925 gaagagaaaGGGGG GKLF
1458 (+) 0.946 0.915 aagagaaagGGGGG GKLF  1458 (+) 0.946 0.915 aagagaaagGGGGG GKLF
1459 (+) 0.946 0.939 agagaaaggGGGGG GKLF  1459 (+) 0.946 0.939 agagaaaggGGGGG GKLF
1462 (+) 0.85 0.831 gaaagGGGGGgat MZF1  1462 (+) 0.85 0.831 gaaagGGGGGgat MZF1
1463 (+) 0.998 0.925 aaaGGGGGggatgag Spzl  1463 (+) 0.998 0.925 aaaGGGGGggatgag Spzl
1464 (+) 0.998 0.943 aagGGGGGgatgagc Spzl  1464 (+) 0.998 0.943 aagGGGGGgatgagc Spzl
1501 (+) 0.993 0.969 acacAGGAAcaga c-Ets-l(p54) 1501 (+) 0.993 0.969 acacAGGAAcaga c-Ets-l (p54)
1535 (-) 0.924 0.883 agctccCCAGAgc C/EBP1535 (-) 0.924 0.883 agctccCCAGAgc C / EBP
1538 (+) 0.908 0.762 tccccagaGCTTGgggtc Pax-81538 (+) 0.908 0.762 tccccagaGCTTGgggtc Pax-8
1549 (+) 0.97 0.775 tgggGTCAGgacataaagc Pax- 21549 (+) 0.97 0.775 tgggGTCAGgacataaagc Pax- 2
1554 (-) 0.989 0.972 tcaGGACAtaaagcttcaa GR 1554 (-) 0.989 0.972 tcaGGACAtaaagcttcaa GR
1558 (-) 0.672 0.734 gacataaagcTTCAAaacc Pax - 2 1558 (-) 0.672 0.734 gacataaagcTTCAAaacc Pax-2
1571 (-) 0.86 0.702 aaaacCCCGCaagtcctg Pax-81571 (-) 0.86 0.702 aaaacCCCGCaagtcctg Pax-8
1577 (+) 0.843 0.803 ccGCAAGtcc Ncx 1584 (+) 1 0.98 tccTGGGAacaa Ik— 2 1594 (-) 0.783 0.789 aaGGGAGct t tea ZF5 1621 (+) 0.843 0.818 ctGCAAGtgt Ncx 1648 (-) 1 0.915 tcgCCCCTcctta MZF1 1654 (-) 0.867 0.795 ctcCTTAAgc Ncx 1664 (+) 0.903 0.9 caagccCACCTt Pax - 4 1668 (+) 0.908 0.743 cccaccttGCCTGcctac Pax - 8 1670 (-) 0.843 0.793 cacCTTGCct Ncx 1684 (+) 0.81 0.858 acttCTCAGgaatgaggag Pax - 2 1685 (-) 0.831 0.748 cttctcaggaATGAGgagc Pax - 2 1685 (+) 0.868 0.757 cttctcagGAATGaggag Pax - 8 1690 (+) 0.844 0.782 caGGAATgag Ncx 1709 (+) 0.944 0.92 ttTAGCGctctg E2F 1733 (-) 0.868 0.737 cagctCATTCccctctcg Pax - 8 1736 (-) 0.844 0.791 ctcATTCCcc Ncx 1739 (-) 1 0.947 attCCCCTctcgc MZF1 1742 (-) 0.843 0.796 cccctCTCGCagaagccc Pax-8 1775 (-) 0.954 0.957 ctgccCCACCcccc GC box 1775 (+) 1 0.989 ctgcccCACCCc Pax-4 1776 (-) 0.87 0.755 tgcccCACCCcccacggt Pax-8 1776 (-) 0.915 0.932 tgccCCACCcccc Spl 1777 (-) 0.998 0.96 gccccacCCCCCacg Spzl 1780 (+) 0.984 0.952 ccaccccccaCGGTTt E2 1780 (+) 0.986 0.961 ccaccccccaCGGTTt E2 1791 (+) 1 0.966 ggTTTGGagatct C/EBP 1796 (+) 0.86 0.78 ggagatctGCGGGacccc Pax-8 1810 (-) 0.981 0.907 CCCCTtcctttccc GKLF 1811 (-) 1 0.927 CCCTTcctttcccc GKLF 1818 (+) 0.903 0.905 tttcccCACCAg Pax-4 1825 (-) 1 0.978 acCAGTTggg c-Myb 1831 (-) 1 0.901 tGGGTGttccgc Pax-4 cv。。。1577 (+) 0.843 0.803 ccGCAAGtcc Ncx 1584 (+) 1 0.98 tccTGGGAacaa Ik-- 2 1594 (-) 0.783 0.789 aaGGGAGct t tea ZF5 1621 (+) 0.843 0.818 ctGCAAGtgt Ncx 1648 (-) 1 0.915 tcgCCCCTcctta MZF1 1654 (-) 0.867 0.795 ctcCTTAAgc Ncx16 caagccCACCTt Pax-4 1668 (+) 0.908 0.743 cccaccttGCCTGcctac Pax-8 1670 (-) 0.843 0.793 cacCTTGCct Ncx 1684 (+) 0.81 0.858 acttCTCAGgaatgaggag Pax-2 1685 (-) 0.831 0.748 cttctcaggaATGAGgaggx Pax -2 1685 (-) -8 1690 (+) 0.844 0.782 caGGAATgag Ncx 1709 (+) 0.944 0.92 ttTAGCGctctg E2F 1733 (-) 0.868 0.737 cagctCATTCccctctcg Pax-8 1736 (-) 0.844 0.791 ctcATTCCcc Ncx 1739 (-) 1 0.947 attCCCCT1cc96c cccctCTCGCagaagccc Pax-8 1775 (-) 0.954 0.957 ctgccCCACCcccc GC box 1775 (+) 1 0.989 ctgcccCACCCc Pax-4 1776 (-) 0.87 0.755 tgcccCACCCcccacggt Pax-8 1776 (-) 0.915 0.932 tgccCCACCcccccccccccccccccccccc1cc (+) 0.984 0.952 ccaccccccaCGGTTt E2 1780 (+) 0.986 0.961 ccaccccccaCGGTTt E2 1791 (+) 1 0.966 ggTTTGGagatct C / EBP 1796 (+) 0.86 0.78 ggagatctGCGGGacccc Pax-8 1810 (-) 0.981 0.907 CCCCTtcctttccc GKLF 1811 (-) 1 0.927 CCCTTcctttcccc GKLF 1818 (+) 0.903 0.905 tttcccCACCAg Pax-18g -Myb 1831 (-) 1 0.901 tGGGTGttccgc Pax-4 cv. . .
Figure imgf000053_0001
P
Figure imgf000053_0001
P
bD 一
Figure imgf000053_0002
bD one
Figure imgf000053_0002
Figure imgf000053_0003
Figure imgf000053_0003
o O 〇  o O 〇
CO CO
1949 (-) 1 0.951 gactCCGCCcccc Spl 1949 (-) 1 0.951 gactCCGCCcccc Spl
1953 (-) 0,915 0.944 ccgcccccCGCTGgctct AP-4 1953 (-) 0,915 0.944 ccgcccccCGCTGgctct AP-4
1957 (+) 1 0.93 ccCCCGCtggct AP-2 1957 (+) 1 0.93 ccCCCGCtggct AP-2
1968 (+) 1 0.937 tctggggTCTGGgggc Handl/E47 1968 (+) 1 0.937 tctggggTCTGGgggc Handl / E47
1970 (+) 0.822 0.735 tgggGTCTGggggcattgc Pax- 2 1970 (+) 0.822 0.735 tgggGTCTGggggcattgc Pax- 2
1973 (-) 0.992 0.885 ggtctGGGGGca AP-2 1973 (-) 0.992 0.885 ggtctGGGGGca AP-2
1982 (+) 1 0.919 gcATTGCtcagcgg C/EBP 1982 (+) 1 0.919 gcATTGCtcagcgg C / EBP
1982 (+) 0.897 0.894 gcATTGCtcagcg C/EBP 1982 (+) 0.897 0.894 gcATTGCtcagcg C / EBP
2003 (+) 0.908 0.714 ctggcgcgGCTTGagccg Pax- 8 2003 (+) 0.908 0.714 ctggcgcgGCTTGagccg Pax- 8
2027 (-) J 0.974 gacTGACAgct TGIF 2027 (-) J 0.974 gacTGACAgct TGIF
産業上の利用可能性 Industrial applicability
本発明のオリゴヌクレオチドは、 低毒性で、 優れたがんの予防および/または 治療剤として有用である。 また、 本発明の受容体蛋白質の遺伝子の転写調節領域 の下流 (発現制御下) にレポ一夕一遺伝子を連結した組換え DNAで形質転換し た形質転換体を使用したスクリ一ニングで得られる化合物、 好ましくは本発明の 受容体蛋白質の発現を阻害する活性を有する化合物またはその塩も、 低毒性で、 優れたがんの予防および Zまたは治療剤として有用である。  The oligonucleotide of the present invention has low toxicity and is useful as an excellent agent for preventing and / or treating cancer. Further, it can be obtained by screening using a transformant transformed with a recombinant DNA in which a repo overnight gene is ligated downstream (under expression control) of the transcription regulatory region of the receptor protein gene of the present invention. A compound, preferably a compound having an activity of inhibiting the expression of the receptor protein of the present invention or a salt thereof is also low toxicity and is useful as an excellent agent for preventing and / or treating cancer.

Claims

請求 の 範 囲 The scope of the claims
1. 配列番号: 13または配列番号: 14で表される塩基配列と同一または実 質的に同一の塩基配列を含有するオリゴヌクレオチド。 1. An oligonucleotide containing the same or substantially the same nucleotide sequence as the nucleotide sequence represented by SEQ ID NO: 13 or SEQ ID NO: 14.
2. 配列番号: 13で表される塩基配列からなるオリゴヌクレオチド。  2. An oligonucleotide having a base sequence represented by SEQ ID NO: 13.
3. 配列番号: 14で表される塩基配列からなるオリゴヌクレオチド。  3. An oligonucleotide having a base sequence represented by SEQ ID NO: 14.
4. ァンチセンスオリゴヌクレオチドである請求項 1記載のオリゴヌクレオチ ド、' 4. The oligonucleotide according to claim 1, which is an antisense oligonucleotide.
5. DN Aである請求項 1記載のオリゴヌクレオチド。  5. The oligonucleotide according to claim 1, which is a DNA.
6. 請求項 1記載のオリゴヌクレオチドを含有してなる医薬。 6. A pharmaceutical comprising the oligonucleotide according to claim 1.
7. がんの予防および/または治療剤である請求項 6記載の医薬。 7. The medicament according to claim 6, which is an agent for preventing and / or treating cancer.
8. 配列番号: 20で表される塩基配列と同一または実質的に同一の塩基配列 を含有する DNA。 8. DNA containing a nucleotide sequence identical or substantially identical to the nucleotide sequence represented by SEQ ID NO: 20.
9. 配列番号: 12で表される塩基配列と同一または実質的に同一の塩基配列 を含有する D N Aの転写調節領域である請求項 8記載の D N A。  9. The DNA according to claim 8, which is a DNA transcriptional regulatory region containing the same or substantially the same nucleotide sequence as the nucleotide sequence represented by SEQ ID NO: 12.
10. 転写調節領域がプロモーター領域である請求項 9記載の DNA。  10. The DNA according to claim 9, wherein the transcription control region is a promoter region.
11. 請求項 8記載の DNAを含有する組換えべクタ一。  11. A recombinant vector containing the DNA according to claim 8.
12. 配列番号: 12で表される塩基配列と同一または実質的に同一の塩基配 列を含有する D N Aの転写調節領域の下流にレポ一夕一遺伝子を有する D N Aを 含有する請求項 1 1記載の組換えべクタ一。  12. The method according to claim 11, which comprises a DNA having a repo all-in-one gene downstream of a transcription regulatory region of a DNA containing the same or substantially the same base sequence as the base sequence represented by SEQ ID NO: 12. Recombination vector.
13. 請求項 1 1記載の組換えべクタ一を含有する形質転換体。  13. A transformant containing the recombinant vector according to claim 11.
14. 請求項 8記載の DNAを用いることを特徴とする、 配列番号: 12で表 される塩基配列と同一または実質的に同一の塩基配列を含有する D N Aの転写調 節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。  14. Use of the DNA of claim 8 to promote or inhibit the transcriptional regulation activity of DNA containing the same or substantially the same nucleotide sequence as the nucleotide sequence represented by SEQ ID NO: 12 A method for screening a compound or a salt thereof.
15. 請求項 13記載の形質転換体を用いる請求項 14記載のスクリーニング 方法。  15. The screening method according to claim 14, wherein the transformant according to claim 13 is used.
16. 請求項 8記載の DNAを含有することを特徴とする、 配列番号: 12で 表される塩基配列と同一または実質的に同一の塩基配列を含有する DN Aの転写 調節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用キット。 16. Promoting or inhibiting the transcriptional regulatory activity of a DNA containing the same or substantially the same nucleotide sequence as the nucleotide sequence represented by SEQ ID NO: 12, characterized by containing the DNA according to claim 8. For screening a compound or a salt thereof.
1 7 . 請求項 1 4記載のスクリーニング方法または請求項 1 6記載のスクリ一 ニング用キットを用いて得られる配列番号: 1 2で表される塩基配列と同一また は実質的に同一の塩基配列を含有する D NAの転写調節活性を促進または阻害す る化合物またはその塩。 17. Nucleotide sequence identical or substantially identical to the nucleotide sequence represented by SEQ ID NO: 12 obtained by using the screening method according to claim 14 or the screening kit according to claim 16 A compound or a salt thereof which promotes or inhibits the transcriptional regulatory activity of DNA, comprising:
1 8 . 請求項 1 7記載の化合物またはその塩を含有してなる医薬。  18. A medicament comprising the compound according to claim 17 or a salt thereof.
1 9 . がんの予防および Zまたは治療剤である請求項 1 8記載の医薬。  19. The medicament according to claim 18, which is a prophylactic and / or therapeutic agent for cancer.
2 0 . 哺乳動物に対して、 請求項 1記載のオリゴヌクレオチドの有効量を投与 することを特徴とするがんの予防 ·治療方法。  20. A method for preventing and treating cancer, comprising administering an effective amount of the oligonucleotide according to claim 1 to a mammal.
2 1 . がんの予防 ·治療剤を製造するための請求項 1記載のオリゴヌクレオチ ドの使用。  21. Use of the oligonucleotide according to claim 1 for producing an agent for preventing or treating cancer.
PCT/JP2003/004272 2002-04-05 2003-04-03 Preventives and/or remedies for cancer WO2003085106A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2003236358A AU2003236358A1 (en) 2002-04-05 2003-04-03 Preventives and/or remedies for cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2002-103497 2002-04-05
JP2002103497A JP2003289876A (en) 2002-04-05 2002-04-05 Preventive and/or therapeutic agent for cancer

Publications (1)

Publication Number Publication Date
WO2003085106A1 true WO2003085106A1 (en) 2003-10-16

Family

ID=28786304

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2003/004272 WO2003085106A1 (en) 2002-04-05 2003-04-03 Preventives and/or remedies for cancer

Country Status (3)

Country Link
JP (1) JP2003289876A (en)
AU (1) AU2003236358A1 (en)
WO (1) WO2003085106A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001038503A2 (en) * 1999-11-24 2001-05-31 Sugen, Inc. Novel human protein kinases and protein kinase-like enzymes
WO2001074851A2 (en) * 2000-03-30 2001-10-11 Curagen Corporation Novel proteins and nucleic acids encoding same
WO2002008253A2 (en) * 2000-07-26 2002-01-31 Merck Patent Gmbh A NOVEL MEMBER OF THE EphA RECEPTOR FAMILY
WO2002008399A2 (en) * 2000-07-21 2002-01-31 Incyte Genomics, Inc. Human kinases
US20020025570A1 (en) * 2000-06-09 2002-02-28 Pe Corporation (Ny) Isolated human kinase proteins, nucleic acid molecules encoding human kinase proteins, and uses thereof
WO2002083735A1 (en) * 2001-04-06 2002-10-24 Takeda Chemical Industries, Ltd. Novel receptor protein and dna thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001038503A2 (en) * 1999-11-24 2001-05-31 Sugen, Inc. Novel human protein kinases and protein kinase-like enzymes
WO2001074851A2 (en) * 2000-03-30 2001-10-11 Curagen Corporation Novel proteins and nucleic acids encoding same
US20020025570A1 (en) * 2000-06-09 2002-02-28 Pe Corporation (Ny) Isolated human kinase proteins, nucleic acid molecules encoding human kinase proteins, and uses thereof
WO2002008399A2 (en) * 2000-07-21 2002-01-31 Incyte Genomics, Inc. Human kinases
WO2002008253A2 (en) * 2000-07-26 2002-01-31 Merck Patent Gmbh A NOVEL MEMBER OF THE EphA RECEPTOR FAMILY
WO2002083735A1 (en) * 2001-04-06 2002-10-24 Takeda Chemical Industries, Ltd. Novel receptor protein and dna thereof

Also Published As

Publication number Publication date
JP2003289876A (en) 2003-10-14
AU2003236358A1 (en) 2003-10-20

Similar Documents

Publication Publication Date Title
US20050240008A1 (en) Novel protein, dna thereof and process for producing the same
WO2001016309A1 (en) Novel g protein-coupled receptor protein and dna thereof
US20110104705A1 (en) Musclin receptor and use thereof
US7235531B2 (en) Tachykinin-like polypeptides and use thereof
US20100008926A1 (en) Novel proteins and use thereof
JP3792655B2 (en) Novel oncogene, recombinant protein derived from the oncogene, and uses thereof
WO2003085106A1 (en) Preventives and/or remedies for cancer
WO2003055506A1 (en) Preventives/remedies for cancer
WO2003091434A1 (en) Novel protein and dna thereof
JP4761812B2 (en) Musclin receptor and use thereof
JP4530631B2 (en) Novel protein and cancer preventive / therapeutic agent
JPWO2005118631A1 (en) Novel protein complex and its use
US20070275888A1 (en) Preventive/Remedy for Cancer
US20040091960A1 (en) Novel physiologically active peptides and use thereof
US20090047282A1 (en) Prophylactic/Therapeutic Agent for Cancer
WO2003087155A1 (en) Novel protein and dna thereof
JP2003277289A (en) Preventive/remedial agent for cancer
WO2004002514A1 (en) Preventives/remedies for cancer
JP2004089182A (en) Preventive/therapeutic agent for cancer
JP2005015460A (en) Use of sglt homolog
WO2002088182A1 (en) Nove g protein-coupled receptor protein and dna thereof
JP2003189873A (en) Prophylactic/therapeutic agent for cancer
JP2001340094A (en) New protein, dna encoding the same and method of producing the same
US20050208494A1 (en) Novel proteins and dnas thereof
WO2004058969A1 (en) Preventives/remedies for cancer

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

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