WO2004071534A1 - Preventives/remedies for kidney diseases - Google Patents

Preventives/remedies for kidney diseases Download PDF

Info

Publication number
WO2004071534A1
WO2004071534A1 PCT/JP2004/001645 JP2004001645W WO2004071534A1 WO 2004071534 A1 WO2004071534 A1 WO 2004071534A1 JP 2004001645 W JP2004001645 W JP 2004001645W WO 2004071534 A1 WO2004071534 A1 WO 2004071534A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
present
dna
amino acid
acid sequence
Prior art date
Application number
PCT/JP2004/001645
Other languages
French (fr)
Japanese (ja)
Inventor
Hiroyuki Miya
Takanori Matsuo
Hiroko Tsuge
Original Assignee
Takeda Pharmaceutical Company Limited
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 Pharmaceutical Company Limited filed Critical Takeda Pharmaceutical Company Limited
Publication of WO2004071534A1 publication Critical patent/WO2004071534A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy

Definitions

  • the present invention relates to a preventive / therapeutic agent for renal diseases such as diabetic nephropathy, a diagnostic agent, and a screening of a preventive / therapeutic agent for renal diseases such as diabetic nephropathy.
  • kidneys excrete ⁇ Maintain the body's homeostasis by regulating the concentration of body fluid components through reabsorption, and activate vitamins! ), Is an extremely important tissue that also functions as an endocrine organ that produces and secretes physiologically active substances such as erythropoietin and renin. Impaired renal function, which plays such an important role, can lead to a variety of conditions and a wide variety of renal diseases. These kidney diseases include nephropathy closely related to diabetes, obesity, and abnormal lipid metabolism.In particular, diabetic nephropathy, which is a primary disease of diabetes, requires strict control of diabetes after diagnosis of nephropathy.
  • Wistar Fatty rats show symptoms characteristic of diabetes in addition to renal dysfunction, so they are used as a diabetic nephropathy model (Front iers in diabetes research). , lessons from animal diabetes II, 535-541, 1988), and Zucker Fatty rats (ZF rats) show diabetic nephropathy because they exhibit renal dysfunction and diabetes as well as WF rats.
  • WF rats Wistar Fatty rats
  • ZF rats Zucker Fatty rats
  • SHC rats spontaneously hypercholesterolemia rats
  • human type lib Na / Pi cotransporter sodium phosphate cotransporter type lib
  • transpomf amyl sodium-dependent phosphate transport activity
  • high activity at low pH This point is consistent with the results of the phosphate uptake assay using the rabbit and mouse small intestine, suggesting that it may be a molecule responsible for phosphorus uptake in the small intestine (Biochemical and Biophysical Research Communi cat ions, 258, 578-582, 1999).
  • treatment of diseases related to the function of human Type Lib Na / Picot ransporter such as treatment of abnormal conditions of serum phosphate concentration such as hypophosphatemia and hyperphosphatemia, etc.
  • U.S. Pat.No. 6,319,688 states that angiogonists and agonists of human Type I ib Na / Picotransporter are useful in treating chronic renal failure, end-stage renal disease, uremic bone disease, and cancer. It is described as useful for preventive care.
  • the present inventors have conducted intensive studies to solve the above problems, and as a result, analyzed model animals such as WF rat, ZF rat, SHC rat, and found a gene whose expression is significantly increased, As a result of further studies based on this finding, the present invention has been completed.
  • a renal disease comprising a compound which inhibits the activity of a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof, or a salt thereof, or a salt thereof.
  • an agent for diagnosing renal disease comprising a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a polynucleotide encoding a partial peptide thereof,
  • Inorganic phosphate is added to a synthetic lipid bilayer containing a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof.
  • Inorganic phosphate transport activity was measured when the buffer containing salt was brought into contact with the buffer containing inorganic phosphate, and (ii) when the buffer containing inorganic phosphate was brought into contact with the synthetic lipid bilayer in the presence of the test compound. Above to measure
  • a prophylactic / therapeutic agent for kidney disease comprising a compound that inhibits the activity of a salt or a salt thereof,
  • a prophylactic and / or therapeutic agent for renal disease characterized by containing a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof.
  • the renal disease is diabetic nephropathy or focal glomerulosclerosis (1) 5) the screening method described,
  • a prophylactic / therapeutic agent for renal disease comprising the compound or salt thereof according to (17a) above,
  • a compound that inhibits the activity of a protein, or a partial peptide or a salt thereof containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 in mammals Or an effective amount of a compound or a salt thereof, or a compound or a salt thereof that inhibits the expression of the gene of the protein or its partial peptide or a salt thereof, or a method for preventing or treating renal disease,
  • FIG. 1 is a graph showing the time course of the expression level of rat Typelib Na / Pi Cotransporter.
  • the vertical axis represents the relative expression level of rat Typelib Na / Pi Cotransporter gene expression value to rat GAPDH expression value
  • the horizontal axis represents the age at which kidneys were collected. ⁇ indicates the results of the experimental group (WF rats), and the mouth indicates the results of the control group (WL rats).
  • FIG. 2 is a graph showing the time course of the expression level of rat Typelib Na / Pi Cotransporter.
  • the vertical axis indicates the relative expression level of the rat Type Lib Na / Pi Co transporter gene expression value to the rat GAPDH expression value
  • the horizontal axis indicates the age at which the kidney was collected.
  • shows the results of the experimental group (ZF rats) and the mouth shows the results of the control group (ZL rats).
  • FIG. 3 is a diagram showing a time-dependent change in the expression level of rat Typelib Na / Pi Cotransporter.
  • the vertical axis indicates the relative expression level of the rat Type Lib Na / Pi Cotransporter gene expression value to the rat GAPDH expression value
  • the horizontal axis indicates the age at which the kidney was collected. The results are for the experimental group (SHC rats) and the control group (SD rats) for the mouth.
  • FIG. 4 is a graph showing the effect of TCV116 on rat Typelib Na / Pi Cotransporter expression levels.
  • the vertical axis indicates the relative expression level of the rat Typelib Na / Pi Cotransporter gene expression value to the rat GAPDH expression value.
  • the horizontal axis shows the type of rat used in the experiment and the presence or absence of TCV116 administration.
  • ZF shows the results of ZF rats to which vehicle was administered
  • ZF (TCV116) shows the results of ZF rats to which TCV116 was administered
  • ZL (Vehicle) shows the results of ZL rats to which vehicle was administered.
  • FIG. 5 is a diagram showing the expression levels of human Typelib Na / Pi Cotransporter in various normal human cells.
  • the vertical axis indicates the relative expression level of the human Typelib Na / Pi Cotransporter gene expression value to the GAPDH expression value.
  • Lane 1 on the horizontal axis is umbilical vein endothelial cells
  • lane 2 is aortic endothelial cells
  • lane 3 is coronary endothelial cells
  • lane 4 is skin microvascular endothelial cells
  • lane 5 is lung microvascular endothelial cells
  • lane 6 is pulmonary artery blood vessels.
  • Endothelial cells lane 7 aortic smooth muscle cells, lane 8 coronary artery flat muscle cells, lane 9 uterine smooth muscle cells, lane 10 bronchial smooth muscle cells, lane 11 skeletal muscle cells, lane 12 mammary epithelium Cells, lane 13 is bronchial epithelial cells, lane 14 is bronchiolar epithelial cells, lane 15 is lung fibroblasts, lane 16 is renal proximal tubule cells, lane 17 is mezzanine cells, lane 18 is renal cortical cells, lane 19 shows renal epithelial cells, lane 20 shows mesenchymal stem cells, lane 21 shows chondrocytes, lane 22 shows osteoblasts, lane 23 shows prostate stromal cells, and lane 24 shows epidermal keratinocytes.
  • a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 may be a human or a human.
  • Cells of warm-blooded animals eg, guinea pigs, rats, mice, chicks, etc., egrets, pigs, sheep, pigs, monkeys, etc.
  • hepatocytes, spleen cells, nerve cells, glial cells, kidney / 3 cells, bone marrow cells, mesangial cells, Langerhans cells, epidermal cells, epithelial cells, goblet cells, endothelial cells, smooth muscle cells, fibroblasts, fiber cells, muscle cells, fat cells, immune cells
  • the brain various parts of the brain (eg, olfactory bulb) , Amygdala, basal ganglia, hippocampus, thalamus, hypothalamus, cerebral cortex, medulla oblongata, cerebellum), spinal cord, pituitary, stomach, victory, kidney, liver, gonad, thyroid, gall bladder, bone marrow, adrenal gland, skin , Muscle, lung, gastrointestinal tract (eg, large intestine, small intestine), blood vessels, heart, thymus,
  • the amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 is about 60% or more, preferably about 70% or more, preferably about 80% or more of the amino acid sequence represented by SEQ ID NO: 1. % Or more, preferably about 90% or more, preferably about Amino acid sequences having a homology of 95% or more.
  • Examples of the protein having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 include, for example, a protein having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 described above. However, a protein having substantially the same activity as the protein containing the amino acid sequence represented by SEQ ID NO: 1 is preferable.
  • Examples of the protein containing an amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 1 include, for example, a protein containing the amino acid sequence represented by SEQ ID NO: 3, and a protein represented by SEQ ID NO: 5 And a protein containing an amino acid sequence.
  • substantially the same activity examples include an inorganic phosphate uptake activity and an inorganic phosphate transport activity.
  • substantially identical indicates that the properties are qualitatively (eg, physiologically or pharmacologically) equivalent. Therefore, inorganic phosphate uptake activity, inorganic phosphate transport activity, etc. are equivalent (eg, about 0.01 to 100 times, preferably about 0.1 to 10 times, more preferably 0.5 to 2 times). ), But the quantitative factors such as the degree of these activities and the molecular weight of the protein may be different.
  • Inorganic phosphate uptake activity can be measured according to a method known per se, for example, the method described in Methods in Enzymology, 191: 494-505, 1990 or a method analogous thereto.
  • a labeled inorganic phosphate eg, an inorganic phosphate labeled with a radioisotope
  • a labeled inorganic phosphate is added to cells expressing the protein of the present invention, and after incubation for a certain time, intracellular This is performed by measuring the amount of inorganic phosphoric acid.
  • the reaction is performed in an appropriate buffer, and the inorganic phosphate that has not been taken up by cells is washed away before measurement.
  • the labeling agent is a radioisotope
  • the uptake activity is measured by a known method, for example, using a radioactivity measuring device.
  • the inorganic phosphate transport activity can be measured according to a method known per se, for example, the method described in Biochimi ca et Biophysica ca Acta, Vol. 1236, pp. 339-344, 1995, or a method analogous thereto. Can be.
  • inorganic phosphate is added to the synthetic lipid bilayer containing the protein of the present invention.
  • a buffer containing a salt is added, and the change in membrane current that occurs when inorganic phosphate is transported by the protein of the present invention is measured.
  • the membrane current is measured using a known method, for example, a membrane potential fixing method.
  • the synthetic lipid bilayer containing the protein of the present invention can be obtained by, for example, using a purified protein obtained from a membrane fraction of cells expressing the protein of the present invention according to a known membrane protein purification method, using a phospholipid or the like.
  • the material can be manufactured according to a known method, for example, a method described in "Patch Clamp Experimental Technique" (Yoshioka Shoten, published in 1996).
  • Examples of the protein used in the present invention include: (i) one or two or more (for example, 1 to 10) in the amino acid sequence represented by SEQ ID NO: 1, 'SEQ ID NO: 3 or SEQ ID NO: 5; An amino acid sequence in which about 0, preferably about 1 to 30, preferably about 1 to 10, and more preferably about (1 to 5) amino acids have been deleted; (ii) SEQ ID NO: 1, 1 or 2 or more amino acid sequences represented by SEQ ID NO: 3 or SEQ ID NO: 5 (for example, about 1 to 100, preferably about 1 to 30 and preferably about 1 to 10) (Iii) one or two amino acids in the amino acid sequence represented by SEQ ID NO: 1, SEQ ID NO: 3 or SEQ ID NO: 5, and more preferably (1 to 5) amino acids.
  • a 7-amino acid sequence into which a number (1 to 5) of amino acids have been inserted (iv) one or more of the amino acid sequences represented by SEQ ID NO: 1, SEQ ID NO: 3 or SEQ ID NO: 5 Or two or more (for example, about 1 to 100, preferably about 1 to 30, preferably about 1 to 10, and more preferably about 1 to 5) amino acids.
  • mucins such as proteins containing an amino acid sequence substituted with an amino acid of (a) or (v) an amino acid sequence obtained by combining them are also included.
  • the position of the insertion, deletion or substitution is not particularly limited.
  • the left end is the N-terminus (amino terminus) and the right end is the C-terminus (potassium terminus) according to the convention of peptide notation.
  • the protein used in the present invention including the protein containing the amino acid sequence represented by SEQ ID NO: 1, has a C-terminal lipoxyl group (-C00H), It may be any of (—COO—), amide (—C0NH 2 ) and ester (—C00R).
  • R in the ester e.g., methyl, Echiru, n- propyl Le, isopropyl, CM alkyl group, such as n- butyl, cyclopentyl Le, C 3 _ 8 cycloalkyl group such as cyclohexyl, for example, phenyl, alpha-Na C 6 _ 12 7 aryl group such as Fuchiru, for example, benzyl, C 7, such as single Nafuchiru C M Al kill groups such as phenyl one CM alkyl group or single naphthylmethyl such phenethyl - 14 Ararukiru group, A pivaloyloxymethyl group or the like is used.
  • CM alkyl group such as n- butyl, cyclopentyl Le, C 3 _ 8 cycloalkyl group such as cyclohexyl, for example, phenyl, alpha-Na C 6 _ 12 7 aryl group such
  • the protein used in the present invention has a lipoxyl group (or carboxylate) other than the C-terminal
  • the protein in which the lipoxyl group is amidated or esterified is also included in the protein used in the present invention.
  • the ester in this case for example, the above-mentioned C-terminal ester or the like is used.
  • the protein used in the present invention the amino acid residue (e.g., Mechionin residues) of N-terminal Amino group protecting groups (e.g., c Bok such c Bok 6 Al force Noiru such formyl group, Asechiru group 6- Glycyl group), N-terminal glutamine residue generated by cleavage in vivo, and pyroglutamine oxidation, substituent on the side chain of amino acid in the molecule (e.g.,- 0H, -SH, amino group, imidazole group, indole group, guanidino group, etc. are protected with appropriate protecting groups (for example, C-silyl groups such as alkanoyl groups such as formyl group and acetyl group). And complex proteins such as so-called glycoproteins to which sugar chains are bound.
  • N-terminal Amino group protecting groups e.g., c Bok such c Bok 6 Al force Noiru such formyl group, Asechiru group
  • protein used in the present invention include, for example, a protein containing the amino acid sequence represented by SEQ ID NO: 1, a protein containing the amino acid sequence represented by SEQ ID NO: 3, and a protein containing the amino acid sequence represented by SEQ ID NO: 5. Proteins containing the represented amino acid sequence are exemplified.
  • the partial peptide of the protein used in the present invention is the partial peptide of the protein used in the present invention described above, and is preferably any one having the same properties as the protein used in the present invention described above. Even things Good.
  • amino acids represented by SEQ ID NO: 1 to 1 to 101, 1 87 to 360, and 57 A peptide having an amino acid sequence of the 5th to 69th amino acid sequence, the 1st to 101st, the 187th to 360th, and the 575th to 6995th amino acid sequence represented by SEQ ID NO: 3
  • the partial peptide used in the present invention may have one or two or more amino acids in its amino acid sequence (for example, about 1 to 20, preferably about 1 to 10, more preferably about 1 to 5). Amino acids are deleted, or one or more amino acids are added to the amino acid sequence (for example, about 1 to 20, preferably about 1 to 10, more preferably about 1 to 5 )) Or one or more amino acids (eg, about 1 to 20, preferably about 1 to 10, more preferably number (1 to 5)) to the amino acid sequence. Or one or more amino acids in the amino acid sequence (for example, about 1 to 20 amino acids, preferably about 1 to 10 amino acids, more preferably about 1 to 5 amino acids). An amino acid may be substituted with another amino acid.
  • the partial peptide used in the present invention may be either.
  • the partial peptide used in the present invention has a lipoxyl group (or carboxylate) other than the C-terminal, and the N-terminal amino acid residue.
  • the amino group of a group eg, methionine residue
  • the amino acid residue has been oxidized with pip dar, the substituent on the side chain of the amino acid in the molecule is protected with an appropriate protecting group, or the so-called sugar peptide to which a sugar chain is bound.
  • Complex peptides and the like are also included.
  • the partial peptide used in the present invention can also be used as an antigen for producing an antibody.
  • salts with physiologically acceptable acids eg, inorganic acids, organic acids
  • bases eg, alkali metals
  • Acceptable acid addition salts are preferred.
  • Such salts include, for example, salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid) or organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, Salts with succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid) are used.
  • inorganic acids eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid
  • organic acids eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid
  • the protein or its partial peptide or a salt thereof used in the present invention can be produced from the above-mentioned human or warm-blooded animal cells or tissues by a known method for purifying a protein, or a protein encoding D It can also be produced by culturing a transformant containing NA. It can also be produced according to the peptide synthesis method described below.
  • the human or mammalian tissues or cells are homogenized, then extracted with an acid or the like, and the resulting extract is subjected to reversed phase chromatography, ion exchange chromatography, etc. Purification and isolation can be performed by combining the above chromatography.
  • 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, 4 —Hydroxymethylmethylphenylacetamidomethyl resin, polyacrylamide resin, 4- (2,, 4'-dimethoxyphenyl-hydroxymethyl) phenoxy resin, 4- (2,, 4,1-dimethoxyphenyl) F mo Ami Noethyl) phenoxy resin and the like.
  • amino acids having a suitably protected amino group and side chain functional group are condensed on the resin in accordance with the sequence of the target protein in accordance with various known condensation methods.
  • the protein or partial peptide is cleaved from the resin, and at the same time, various protecting groups are removed.
  • an intramolecular disulfide bond formation reaction is performed in a highly diluted solution to obtain the target protein or partial peptide or an amide thereof. I do.
  • various activating reagents that can be used for protein synthesis can be used, and carbodiimides are particularly preferable.
  • disulfide diimides DCC, N, N, -diisopropylcarpo- imide, N-ethyl- ⁇ '-(3-dimethylaminoprolyl) carbopimide, and the like are used.
  • the protected amino acid may be added directly to the resin with a racemization inhibitor additive (e.g., HOB t, HOOB t), or may be pre-protected as a symmetrical anhydride or HOB t ester or H ⁇ OB t ester
  • a racemization inhibitor additive e.g., HOB t, HOOB t
  • the amino acid can be added to the resin after activation.
  • the solvent used for activating the protected amino acid or condensing with the resin may be appropriately selected from solvents known to be usable for the protein condensation reaction.
  • acid amides such as N, N-dimethylformamide, N, N-dimethylacetoamide, N-methylpyrrolidone, halogenated hydrocarbons such as methylene chloride, chloroform, trifluoroethanol, etc.
  • Alcohols, sulphoxides 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. Used.
  • the reaction temperature is appropriately selected from a range known to be usable for the protein bond formation reaction, and is usually appropriately selected from a 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 raw material include ⁇ , Boc, t-pentyloxycarponyl, isopolnyloxycarbonyl, 4-methoxybenzyloxyl-ponyl, C 1 _Z, Br—Z, and adaman Tyloxycarbonyl, trifluoroacetyl, phthaloyl, formyl, 2-nitrophenylsulfenyl, diphenylphosphinothioyl, Fmoc and the like are used.
  • Lepoxyl groups are, for example, alkyl esterified (e.g., methyl, ethyl, propyl, butyl, t-butyl, cyclopentyl, cyclohexyl, cyclopentyl, cyclooctyl, 2-adamantyl, etc.).
  • alkyl esterified e.g., methyl, ethyl, propyl, butyl, t-butyl, cyclopentyl, cyclohexyl, cyclopentyl, cyclooctyl, 2-adamantyl, etc.
  • branched or cyclic alkyl esterification branched or cyclic alkyl esterification
  • aralkyl esterification eg, benzyl ester, 412 trobenzyl ester, 4-methoxybenzyl ester, 4-chlorobenzyl ester, benzhydryl esterification
  • phene It can be protected by nasylation, benzyloxycarbonyl hydrazide, t-butoxycarbonyl hydrazide, trityl hydrazide, or the like.
  • the hydroxyl group of serine can be protected, for example, by esterification or etherification.
  • a group suitable for the esterification for example, a group derived from carbonic acid such as a lower (C 6 ) alkanol group such as an acetyl group, an aroyl group such as a benzoyl group, a benzyloxycarbonyl group, and an ethoxycarbonyl group can be used.
  • a group suitable for etherification include a benzyl group, a tetrahydropyranyl group, and a t-butyl group.
  • protecting group for the phenolic hydroxyl group of tyrosine for example, Bz1, C12-Bzl, 2-nitrobenzyl, Br-Z, t-butyl and the like are used.
  • protecting group for imidazole of histidine for example, Tos, 4-methoxy-1,2,3,6-trimethylbenzenesulfonyl, DNP, benzyloxymethyl, Bum, Boc, Trt, Fmoc and the like are used.
  • Examples of the activated carbonyl group of the raw material include, for example, a corresponding acid anhydride, azide, and an active ester [alcohol (eg, pentachlorophenol, 2,4,5-trichlorophenol, 2 , 4-dinitrophenol, shear Nomethyl alcohol, p-nitrophenol, H ⁇ NB, N-hydroxysuccinimide, N-hydroxyfurimide, ester with HOBt)].
  • an active ester eg, pentachlorophenol, 2,4,5-trichlorophenol, 2 , 4-dinitrophenol, shear Nomethyl alcohol, p-nitrophenol, H ⁇ NB, N-hydroxysuccinimide, N-hydroxyfurimide, ester with HOBt
  • an active ester alcohol (eg, pentachlorophenol, 2,4,5-trichlorophenol, 2 , 4-dinitrophenol, shear Nomethyl alcohol, p-nitrophenol, H ⁇ NB, N-hydroxysuccinimide, N-hydroxyfurimide, ester with
  • Methods for removing (eliminating) protecting groups 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.
  • the elimination reaction by the above acid treatment is generally carried out at a temperature of about 120 ° C. to about 40 ° C.
  • anisol for example, anisol, phenol, thioanisole, methacrylol, paracresol, etc. It is effective to add a cation scavenger such as toluene, dimethyl sulfide, 1,4-butanedithiol, or 1,2-ethanedithiol.
  • a cation scavenger such as toluene, dimethyl sulfide, 1,4-butanedithiol, or 1,2-ethanedithiol.
  • the 2,4-dinitrophenyl group used as an imidazole protecting group of histidine is removed by thiophenol treatment, and the formyl group used as an indole protecting group of tryptophan is substituted with 1,2-ethanedithiol, 1,4-
  • 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 material, the protection group, the elimination of the protective group, and the activation of the functional group involved in the reaction can be appropriately selected from known groups or known means.
  • Another method for obtaining an amide form of a protein or partial peptide is, for example, first protecting a carboxy-terminal amino acid with the desired amino acid by amidating and protecting the amino acid side of the peptide (protein) chain. After lengthening, the protein or partial peptide from which only the ⁇ ; -amino protecting group at the N-terminus of the peptide chain has been removed, and the protein or partial protein from which only the protecting group at the C-terminal carbonyl group has been removed. A peptide is produced, and these proteins or peptides are condensed in a mixed solvent as described above. The details of the condensation reaction are the same as above. is there.
  • an ester of a protein or peptide for example, after condensing the ⁇ -lipoxyl group of the terminal amino acid with a desired alcohol to form an amino acid ester, in the same manner as the amide of a protein or peptide, The desired ester of the protein or peptide can be obtained.
  • the partial peptide used in the present invention or a salt thereof can be produced according to a peptide synthesis method known per se, or by cleaving the protein used in the present invention with an appropriate peptidase.
  • a method for synthesizing a peptide for example, any of a solid phase synthesis method and a liquid phase synthesis method may be used. That is, the partial peptide or amino acid that can constitute the partial peptide used in the present invention is condensed with the remaining portion, and when the product has a protecting group, the protecting group is eliminated to produce the desired peptide. can do.
  • Examples of the known condensation method and elimination of the protecting group include the methods described in the following [omega] to (V).
  • the polynucleotide encoding the protein used in the present invention may be any polynucleotide containing the above-described nucleotide sequence encoding the protein used in the present invention.
  • it is DNA.
  • the DNA may be any of genomic DNA, genomic DNA library, the above-described cell / tissue-derived cDNA, the above-described cell / tissue-derived cDNA library, and synthetic DNA.
  • the vector used for the library may be any of pacteriophage, plasmid, cosmid, and phagemid. Alternatively, it can also be directly amplified by Reverse Transcriptase Polymerase Chain Reaction (hereinafter abbreviated as RT-PCR method) using a total RNA or mRNA fraction prepared from the cells and tissues described above.
  • RT-PCR method Reverse Transcriptase Polymerase Chain Reaction
  • the DNA encoding the protein used in the present invention includes, for example, a DNA containing the nucleotide sequence represented by SEQ ID NO: 2 or a DNA hybridizing with the nucleotide sequence represented by SEQ ID NO: 2 under high stringency conditions. Any DNA may be used as long as it contains a soybean base sequence and encodes a protein having substantially the same properties as the protein containing the amino acid sequence represented by SEQ ID NO: 1.
  • Examples of the DNA that can hybridize with the nucleotide sequence represented by SEQ ID NO: 2 under high stringency conditions include, for example, about 60% or more, preferably about 70% or more of the nucleotide sequence represented by SEQ ID NO: 2.
  • DNA containing a base sequence having a homology of about 80% or more, preferably about 90% or more, and preferably about 95% or more is used.
  • Specific examples include a DNA containing the base sequence represented by SEQ ID NO: 4, a DNA containing the base sequence represented by SEQ ID NO: 6, and the like.
  • Hybridization is carried out by a method known per se or a method analogous thereto, for example, Molecular Cloning 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). When a commercially available library is used, it can be performed according to the method described in the attached instruction manual. More preferably, the reaction can be performed under high stringent conditions.
  • the high stringent conditions refer to, 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 t: 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. More specifically, as the DNA encoding the protein containing the amino acid sequence represented by SEQ ID NO: 1, the DNA containing the base sequence represented by SEQ ID NO: 2; As the DNA encoding the protein containing the amino acid sequence represented by SEQ ID NO: 4, a DNA containing the nucleotide sequence represented by SEQ ID NO: 4 and a protein containing the amino acid sequence represented by SEQ ID NO: 5 are encoded. As the DNA to be used, a DNA containing the base sequence represented by SEQ ID NO: 6 and the like are used.
  • the polynucleotide (e.g., DNA) encoding the partial peptide used in the present invention may be any polynucleotide containing a base sequence encoding the partial peptide used in the present invention described above. Is also good. Further, it may be any of genomic DNA, genomic DNA library, the above-described cell-tissue-derived cDNA, the above-described cell-tissue-derived cDNA library, and synthetic DNA. Examples of the DNA encoding the partial peptide used in the present invention include, for example, a DNA having a part of the DNA containing the nucleotide sequence represented by SEQ ID NO: 2, or SEQ ID NO: 2. DNAs that contain a base sequence that hybridizes with a base sequence under high stringency conditions and that contain a part of a DNA encoding a protein having substantially the same activity as the protein of the present invention are used.
  • DNA capable of hybridizing with the nucleotide sequence represented by SEQ ID NO: 2 has the same significance as described above.
  • the DNA and the partial peptide used in the present invention may be used to completely encode the DNA.
  • the DNA is amplified by PCR using a synthetic DNA primer having a part of the nucleotide sequence encoding the protein of the present invention, or the DNA incorporated into an appropriate vector is used as the cloning DNA of the present invention. Selection can be carried out by hybridization with a DNA fragment coding for a part or the entire region of the quality or labeled with a synthetic DNA.
  • the hybridization method can be performed, for example, according to the method described in Molecular Cloning 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). When a commercially available library is used, it can be performed according to the method described in the attached instruction manual.
  • the DNA base sequence can be converted using the ODA-LA PCR method using PCR, a known kit, for example, Mutan TM -super Express Km (Takara Shuzo Co., Ltd.), Mutan TM -K (Takara Shuzo Co., Ltd.), etc. It can be carried out according to a method known per se such as the gapped duplex method, the Kunke method, or a method analogous thereto.
  • the DNA encoding the cloned protein can be used as it is depending on the purpose, or can be digested with a restriction enzyme or added with 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 termination codon at the 3' end. These translation initiation codon and translation termination codon can also be added using an appropriate synthetic DNA adapter.
  • the expression vector for the protein of the present invention includes, for example, (a) cutting out a DNA fragment of interest from DNA encoding the protein of the present invention, and (mouth) downstream of the DNA fragment from a promoter in an appropriate expression vector. It can be manufactured by connecting to
  • the vector examples include a plasmid derived from Escherichia coli (eg, pBR322, pBR325, pUC12, pUC13), a plasmid derived from Bacillus subtilis (eg, pUB110, pTP5, pC194), a plasmid derived from yeast ( Example, pSHl 9, pSH 15), Bacteriophages such as ⁇ phage, animal viruses such as retrovirus, vaccinia virus, baculovirus, etc., pAl-11, pXT1, pRc / CMV, pRc / RSV, pcDNA I / Neo, etc. Is used. '
  • the promoter used in the present invention may be any promoter as long as it is appropriate for the host used for gene expression.
  • SR promoter when an animal cell is used as a host, SR promoter, SV40 promoter, LTR promoter, CMV promoter, HSV-TK promoter and the like can be mentioned.
  • the CMV (cytomegalovirus) promoter the SRa promoter, and the like.
  • the host is a genus Escherichia
  • the host is a Bacillus genus such as trp promoter, lac promoter, recA promoter, ⁇ ⁇ promoter, ⁇ ⁇ promoter, and c7 promoter.
  • the S ⁇ 1 promoter overnight, the SPO2 promoter, the penP promoter, and the like.
  • yeast the PH05 promoter, the PGK promoter, the GAP promoter, the ADH promoter, and the like are preferable.
  • the host is an insect cell, a polyhedrin promoter, a P10 promoter and the like are preferable.
  • the expression vector may contain, in addition to the above, an enhancer, a splicing signal, a poly-A addition signal, a selection marker, an SV40 replication origin (hereinafter sometimes abbreviated as SV40 ori), and the like, if desired.
  • the selection marker one, for example, dihydrofolate reductase (hereinafter sometimes abbreviated as dh fr) gene [methotrexate (MTX) resistant], ampicillin resistant gene (hereinafter sometimes abbreviated as Amp r) , Ne Omaishin resistant gene (hereinafter sometimes abbreviated as Ne 0 1 ", G418 resistant) and the like.
  • dh fr dihydrofolate reductase
  • MTX metalhotrexate
  • Amp r ampicillin resistant gene
  • Ne Omaishin resistant gene hereinafter sometimes abbreviated as Ne 0 1 ", G418 resistant
  • the target gene can be selected using a thymidine-free medium.
  • a signal sequence suitable for the host may be added to the protein of the present invention. Add to N terminal side. If the host is a genus Escherichia, the PhoA 'signal sequence, OmpA signal sequence, etc., if the host is a Bacillus genus, the amylase * signal sequence, subtilisin signal sequence, etc. In some cases, MFa signal sequence, SUC2 signal sequence, etc. In the case where the host is an animal cell, insulin signal sequence, -interferin signal sequence, antibody molecule, signal sequence, etc. can be used, respectively. .
  • a transformant can be produced.
  • Escherichia bacteria for example, Escherichia bacteria, Bacillus bacteria, yeast, insect cells, insects, animal cells and the like are used.
  • Escherichia include, for example, Escherichia coli.
  • Bacillus subtilis MI114 Gene, 24, 255 (1983)]
  • 207-21 Journal of Biochemistry, 95, 87 (1984)] and the like are used.
  • yeast examples include, for example, Saccharomyces cerevisiae AH22, AH22 R-, NA87-11A, DKD-5D, 2OB-12, Schizosaccharomyces pombe N CYC 1913, NCYC 2036, Pichia pastori (Pichia pastoris) KM 71 or the like is used.
  • Insect cells include, for example, when the virus is Ac NPV, a cell line derived from a larva of night moth (Spodoptera frugiperda cell; S f cell), MG1 cell derived from the midgut of Trichoplusia ni, High derived from egg of Trichoplusia ni Five TM cells,
  • Sf cells derived from Mamestra brassicae or cells derived from Estigmena acrea are used. If the virus is BmNP V, a silkworm-derived cell line (Bombyx mori N cells) Vesicles; BmN cells). Examples of the Sf cells include Sf9 cells (ATCC CRL1711) and Sf21 cells (Vaughn, JL et al., In Vipo).
  • insects for example, silkworm larvae are used [Nature, vol. 315, 592 (1985)].
  • animal cells examples include monkey cells COS-7, Vero, Chinese hamster cells CH ⁇ (hereinafter abbreviated as CHO cells), dh fr gene-deficient Chinese hamster cells CHO (hereinafter, CHO (dh fr—) cells Abbreviations), mouse L cells, mouse AtT-20, mouse myeloid cells, mouse ATDC 5 cells, rat GH3, human FL cells, etc. are used.
  • Transformation of a genus Escherichia can be performed, for example, according to the method described in Proc. Natl. Acad. Sci. USA, 69, 2110 (1972), Gene, 17, 107 (1982). it can.
  • Transformation of Bacillus spp. can be carried out, for example, according to the method described in Molecular 'and General Genetics (Molecular & General Genetics), vol. 168, 111 (1979).
  • the yeast can be transformed according to the method described in, for example, Methods in Enzymology, Vol. 194, 182-187 (1991), Proc. Natl. Acad. Sci. USA, Vol. 75, 1929 (1978). 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 to be used for cultivation. It contains carbon sources, nitrogen sources, inorganic substances, etc., necessary for the growth of sorghum.
  • the carbon source include glucose, dextrin, soluble starch, and sucrose.
  • the nitrogen source include ammonium salts, nitrates, corn chips, lica, peptone, casein, meat extract, and soybean meal.
  • Inorganic or organic substances such as potato extract and inorganic substances include, for example, 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.
  • a medium for culturing a bacterium belonging to 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.
  • a drug such as, for example, 3; 8-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 applied.
  • the cultivation is usually performed at about 30 to 40 ° C for about 6 to 24 hours, and if necessary, aeration and stirring can be applied.
  • the culture medium When culturing an insect cell or a transformant whose host is an insect, the culture medium may be supplemented with Grace's Insect Medium (Nature, 195, 788 (1962)) with appropriate addition of immobilized 10% serum or other additives. Are used.
  • the pH of the medium is preferably adjusted to about 6.2 to 6.4. Culture is usually performed at about 27 ° C for about 3 to 5 days, and aeration and agitation are added as necessary.
  • the medium When culturing a transformant in which the host is an animal cell, the medium may be, for example, MEM medium containing about 5 to 20% fetal bovine serum [Science, 122 vol., 501 (1952)], DM EM medium [Virology, 8 volumes, 396 (1959)], RPMI 1640 medium [The Journal of the American Medical Association 199, 519 (1967)], 199 medium
  • the pH is preferably about 6 to 8.
  • Culture is usually performed at about 30 to 40 ° C. Run for 15-60 hours, adding aeration and / or agitation as needed.
  • the protein of the present invention can be produced in the cells of the transformant, in the cell membrane, or outside the cells.
  • the protein of the present invention can be separated and purified from the culture by, for example, the following method.
  • the cells or cells are collected by a known method, suspended in an appropriate buffer, and subjected to ultrasonic wave, lysozyme, and / or freeze-thawing. After the cells or cells are destroyed by the method, a method of obtaining a crude protein extract by centrifugation or filtration is appropriately used.
  • a protein modifier such as urea or hydrochloric guanidine in the buffers may contain a surfactant such as preparative Litton X- 1 0 0 TM.
  • the protein contained in the culture supernatant or extract obtained in this manner can be purified by appropriately combining known separation and purification methods.
  • known separation and purification methods include methods using solubility such as salting out and solvent precipitation, dialysis, ultrafiltration, gel filtration, and SDS-polyacrylamide gel electrophoresis, mainly for molecular weight analysis.
  • Method using difference method using charge difference such as ion exchange chromatography, method using specific affinity such as affinity chromatography, hydrophobicity such as reversed-phase high-performance liquid chromatography, etc.
  • a method utilizing a difference, a method utilizing an isoelectric point difference such as an isoelectric focusing method, and the like are used.
  • the protein thus obtained when it is obtained in a free form, it can be converted to a salt by a method known per se or a method analogous thereto, and conversely, when it is obtained as a salt, a method known per se or analogous thereto Depending on the free form or other Can be converted to a salt.
  • the protein produced by the recombinant can be arbitrarily modified or the polypeptide can be partially removed by applying an appropriate protein modifying enzyme before or after purification.
  • an appropriate protein modifying enzyme for example, trypsin, chymotrypsin, arginyl endopeptidase, protein kinase, dalicosidase and the like are used.
  • the presence of the thus-produced protein of the present invention can be measured by enzyme immunoassay using a specific antibody, Western blotting, or the like.
  • the antibody against the protein or partial peptide or a salt thereof used in the present invention may be a polyclonal antibody or a monoclonal antibody as long as it can recognize the protein or partial peptide or a salt thereof used in the present invention. Is also good.
  • an antibody against the protein or partial peptide or a salt thereof (hereinafter, these may be simply referred to as the protein of the present invention) used in the present invention is obtained by using the protein of the present invention as an antigen.
  • the antibody or antiserum can be produced according to a known method for producing an antibody or antiserum.
  • the protein of the present invention is administered to a warm-blooded animal itself or together with a carrier or a diluent at a site capable of producing an antibody upon administration.
  • Complete Freund's adjuvant or incomplete Freund's adjuvant may be administered in order to enhance antibody production upon administration.
  • the administration is usually performed once every 2 to 6 weeks, for a total of about 2 to 10 times.
  • warm-blooded animals to be used include monkeys, rabbits, rabbits, dogs, guinea pigs, mice, rats, sheep, goats, and chickens, and mice and rats are preferably used.
  • a warm-blooded animal immunized with an antigen for example, an individual with an antibody titer is selected from a mouse, and the spleen or lymph node is collected 2 to 5 days after the final immunization.
  • Allogeneic or antibody-producing cells contained in By fusing with a myeloma cell of a heterologous animal, a monoclonal antibody-producing hybridoma can be prepared.
  • the antibody titer in the antiserum can be measured, for example, by reacting a labeled protein described below with the antiserum, and then measuring the activity of a labeling agent bound to the antibody.
  • the fusion operation can be carried out according to a known method, for example, the method of Kohler and Milstein [Nature, 256, 495 (1975)].
  • the fusion promoter include polyethylene glycol (PEG) and Sendai virus, and PEG is preferably used.
  • myeloma cells examples include myeloma cells of warm-blooded animals such as NS-1, P3U1, SP2 / 0, and AP-1, but P3U1 is preferably used.
  • the preferred ratio between the number of antibody-producing cells (spleen cells) and the number of myeloma cells used is about 1: 1 to 20: 1, and PEG (preferably PEG 1000 to PEG 6000) is used at a concentration of about 10 to 80%.
  • Cell fusion can be carried out efficiently by adding the mixture and incubating at 20 to 40 ° C, preferably 30 to 37 ° C for 1 to 10 minutes.
  • a hybridoma culture supernatant is added to a solid phase (eg, a microplate) on which a protein antigen is adsorbed directly or together with a carrier.
  • a solid phase eg, a microplate
  • An anti-immunoglobulin antibody labeled with a radioactive substance or enzyme is used if the cells used for cell fusion are mice) or protein A is added, and the monoclonal antibody bound to the solid phase is added.
  • Hybridoma culture supernatant is added to the solid phase to which the anti-immunoglobulin antibody or protein A is adsorbed, and proteins labeled with radioactive substances or enzymes are added, and the monoclonal antibody bound to the solid phase is detected. And the like.
  • the selection of the monoclonal antibody can be carried out according to a method known per se or a method analogous thereto. Usually, it can be performed in a medium for animal cells supplemented with HAT (hypoxanthine, aminopterin, thymidine).
  • HAT hyperxanthine, aminopterin, thymidine
  • any medium can be used as long as it can grow a hybridoma.
  • RP containing 1-20%, preferably 10-20% fetal calf serum MI 1640 medium, GIT medium containing 1 to 10% fetal bovine serum (Wako Pure Chemical Industries, Ltd.) or serum-free medium for hybridoma culture (SFM-101, Nissui Pharmaceutical Co., Ltd. )) Etc.
  • SFM-101 serum-free medium for hybridoma culture
  • the culture temperature is usually from 20 to 40 ° C, preferably about 37 ° C.
  • the culturing time is usually 5 days to 3 weeks, preferably 1 week to 2 weeks.
  • the culture can be usually performed under 5% carbon dioxide gas.
  • the antibody titer of the hybridoma culture supernatant can be measured in the same manner as the measurement of the antibody titer in the antiserum described above.
  • Monoclonal antibodies can be separated and purified by methods known per se, for example, immunoglobulin separation and purification methods (e.g., salting out method, alcohol precipitation method, isoelectric point precipitation method, electrophoresis method, ion exchanger (e.g., DEAE) Adsorption / desorption method, ultracentrifugation method, gel filtration method, specific purification method of collecting antibody only with antigen-bound solid phase or active adsorbent such as protein A or protein G and dissociating the bond to obtain antibody) Can do it.
  • immunoglobulin separation and purification methods e.g., salting out method, alcohol precipitation method, isoelectric point precipitation method, electrophoresis method, ion exchanger (e.g., DEAE)
  • Adsorption / desorption method e.g., ultracentrifugation method
  • gel filtration method e.g., specific purification method of collecting antibody only with antigen-bound solid phase or active adsorbent such as protein A
  • the polyclonal antibody of the present invention can be produced by a method known per se or a method analogous thereto. For example, an immunizing antigen (protein antigen) itself or a complex thereof with a carrier protein is formed, and immunization is performed on a warm-blooded animal in the same manner as in the method for producing a monoclonal antibody described above.
  • the antibody can be produced by collecting an antibody-containing substance against the protein of the present invention and separating and purifying the antibody.
  • the type of carrier protein and the mixing ratio between carrier and hapten depend on the efficiency of the antibody against hapten immunized by cross-linking with a carrier. If possible, any kind may be crosslinked at any ratio.For example, serum serum albumin, thyroglobulin, hemocyanin, etc. are used in a weight ratio of about 0.1 to 1 for hapten. A method of coupling at a rate of 1 to 20, preferably about 1 to 5 is used.
  • various condensing agents must be used for force coupling between the hapten and carrier.
  • an active ester reagent containing daltaraldehyde, carbodiimide, a maleimide active ester, a thiol group, or a dithiopyridyl group can be used, for example, an active ester reagent containing daltaraldehyde, carbodiimide, a maleimide active ester, a thiol group, or a dithiopyridyl group.
  • the condensation product is administered to a warm-blooded animal at a site where antibody production is possible or together with a carrier or diluent.
  • Complete Freund's adjuvant or incomplete Freund's adjuvant may be administered in order to enhance antibody production ability upon administration.
  • the administration is usually performed once every about 2 to 6 weeks, for a total of about 3 to 10 times.
  • the polyclonal antibody can be preferably collected from blood, such as blood or ascites, of a warm-blooded animal immunized by the above method.
  • the polyclonal antibody titer in the antiserum can be measured in the same manner as the measurement of the antibody titer in the antiserum described above.
  • the separation and purification of the polyclonal antibody can be performed according to the same immunoglobulin separation and purification method as the above-described separation and purification of the monoclonal antibody.
  • Polynucleotides encoding the protein or partial peptide used in the present invention e.g., DNA (hereinafter, these DNAs may be abbreviated as the DNA of the present invention in the description of antisense polynucleotides)
  • Any antisense polynucleotide may be used as long as it has a basic nucleotide sequence or a part thereof and has an action capable of suppressing the expression of the DNA, but antisense DNA is preferable.
  • the nucleotide sequence substantially complementary to the DNA of the present invention is, for example, the entire nucleotide sequence or a part of the nucleotide sequence complementary to the DNA of the present invention (that is, the complementary strand of the DNA of the present invention).
  • a base sequence having about 70% or more, preferably about 80% or more, more preferably about 90% or more, and most preferably about 95% or more homology with the base sequence is exemplified.
  • the nucleotide sequence of the portion encoding the N-terminal site of the protein of the present invention for example, , A base sequence near the start codon, etc.
  • the nucleotide sequence of the portion encoding the N-terminal site of the protein of the present invention for example, , A base sequence near the start codon, etc.
  • about 70% or more preferably about 80% or more, more preferably about 70% or more.
  • the antisense polynucleotide having a homology of 90% or more, most preferably about 95% or more is (mouth) an antisense polynucleotide which directs RNA degradation by RNase H, the present invention containing an intron About 70% or more, preferably about 80% or more, more preferably about 90% or more, and most preferably about 95% or more homology with the complementary strand of the entire base sequence of the DNA. Nucleotides are respectively preferred.
  • nucleotide sequence complementary to or substantially complementary to the nucleotide sequence of DNA containing the nucleotide sequence represented by SEQ ID NO: 2, SEQ ID NO: 4 or SEQ ID NO: 6, or a part thereof preferably a nucleotide sequence complementary to the nucleotide sequence of the DNA containing the nucleotide sequence represented by SEQ ID NO: 2, SEQ ID NO: 4 or SEQ ID NO: 6, or a part thereof.
  • Antisense polynucleotide having a nucleotide sequence (more preferably, a nucleotide sequence complementary to the nucleotide sequence of the DNA containing the nucleotide sequence represented by SEQ ID NO: 2, SEQ ID NO: 4 or SEQ ID NO: 6, or a part thereof) Sense polynucleotide) and the like.
  • An antisense polynucleotide is usually composed of about 10 to 40 bases, preferably about 15 to 30 bases.
  • the phosphate residues (phosphates) of each nucleotide constituting the antisense DNA are, for example, chemically modified phosphate residues such as phosphorothioate, methylphosphonate, and phosphorodithionate. It may be substituted by a group.
  • the sugar (deoxy lipose) of each nucleotide may be substituted with a chemically modified sugar structure such as 2′-dimethylation, and the base (pyrimidine, purine) may also be chemically modified. And any one that hybridizes to DNA having the base sequence represented by SEQ ID NO: 2.
  • These antisense polynucleotides can be produced using a known DNA synthesizer or the like.
  • the antisense polynucleotide (nucleic acid) corresponding to the protein gene of the present invention which can inhibit the replication or expression of the gene, is cloned or D-encoding the determined protein.
  • NA base sequence Can be designed and synthesized based on information.
  • Such antisense polynucleotides can hybridize to RNA of the protein gene of the present invention, inhibit the synthesis or function of the RNA, or interact with the protein-related RNA of the present invention through interaction with the RNA.
  • the expression of the protein gene of the present invention can be regulated and controlled.
  • Polynucleotides complementary to the selected sequence of the protein-linked RNA of the present invention can be used in vivo and in vitro. It is useful for regulating and controlling the expression of protein genes, and is also useful for treating or diagnosing diseases.
  • the term "corresponding" means having homology or being complementary to a nucleotide, base sequence or a specific sequence of a nucleic acid including a gene.
  • nucleotide, nucleotide sequence or nucleic acid and a protein generally refers to the amino acid of the protein (as specified in the directive) derived from the nucleotide (nucleic acid) sequence or its complement.
  • the terminal palindrome region or the 3, terminal hairpin loop can be selected as a preferable target region, but any region in the protein gene can be selected as the target.
  • Antisense polynucleotides include polydexoxyribonucleotides containing 2-deoxy D-report, polyliponucleotides containing D-lipose, and other types of N-glycosides of purine or pyrimidine bases.
  • polymers having a non-nucleotide backbone eg, commercially available protein nucleic acids and synthetic sequence-specific nucleic acid polymers
  • polymers containing special bonds provided that the polymer is DNA or RNA
  • nucleotides having a configuration that allows base pairing and base attachment as found in the above It May be a double-stranded DNA, a single-stranded DNA, a double-stranded RNA, a single-stranded RNA, a DNA: RNA hybrid, and may further comprise an unmodified polynucleotide (or an unmodified oligonucleotide).
  • Nucleotides” and “nucleic acids” may include those containing not only purine and pyrimidine bases, but also other modified heterocyclic bases. Such modifications may include those containing 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.
  • the antisense polynucleotide of the present invention is RNA, DNA or a modified nucleic acid (RNA, DNA).
  • modified nucleic acid include sulfur derivatives of nucleic acids, thiophosphate derivatives, and polynucleoside amides that are resistant to degradation of oligonucleoside amides.
  • the antisense polynucleotide of the present invention can be designed, for example, as follows. That is, an antisense polynucleotide that makes an antisense polynucleotide more stable in a cell.
  • the antisense polynucleotide of the present invention may contain altered or modified sugars, bases, or bonds, and may be provided in a special form such as ribosome or microsphere, applied by gene therapy, It can be provided in an added form.
  • additional forms include polycations, such as polylysine, which act to neutralize the charge on the phosphate backbone, and lipids, which enhance interaction with cell membranes and increase nucleic acid uptake.
  • Preferred lipids for addition include cholesterol and its derivatives (eg, cholesteryl chromate formate, cholic acid, etc.). These can be attached to the 3 'or 5' end of the nucleic acid and can be attached via a base, sugar, or intramolecular nucleoside linkage.
  • Other groups include capping groups specifically located at the 3 'or 5' end of the nucleic acid that prevent degradation by nucleases such as exonucleases and RNases. . Such capping groups include, but are not limited to, hydroxyl-protecting groups known in the art, including glycols such as polyethylene glycol and tetraethylene glycol.
  • the inhibitory activity of the antisense polynucleotide can be measured using the transformant of the present invention, the in vivo or in vitro gene expression system of the present invention, or the protein in vivo or in vitro translation system of the present invention. You can find out.
  • a protein or partial peptide of the present invention or a salt thereof (hereinafter, sometimes abbreviated as the protein of the present invention), a polynucleotide encoding the protein or partial peptide of the present invention (eg, DNA (hereinafter, referred to as DNA) May be abbreviated as the DNA of the present invention))), the protein or partial peptide of the present invention or (Hereinafter sometimes abbreviated as the antibody of the present invention) and a DNA antisense polynucleotide of the present invention (hereinafter sometimes abbreviated as the antisense polynucleotide of the present invention).
  • DNA hereinafter, referred to as DNA
  • DNA DNA antisense polynucleotide of the present invention
  • the expression of the protein of the present invention is increased in the kidney tissues of Wistar Fatty rats (WF rats), Zucker Fatty rats (ZF rats) and spontaneously hypercholesterolemic rats (SHC rats). It can be used as one of the best. In other words, it is useful as a marker for early diagnosis of renal dysfunction, judgment of symptom severity, and prediction of disease progression.
  • Compounds or salts thereof that regulate (preferably inhibit) the activity of the protein of the present invention include, for example, renal diseases (eg, diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome, Mesangium Proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy, membranous proliferative nephritis, crescentic adult nephritis, lupus nephritis, amyloid kidney, tubular interstitial nephritis, acute tubular necrosis, It can be used as a preventive or therapeutic agent for acute renal failure, renal sclerosis, renal sclerosis, renal glomerulosclerosis,
  • the protein of the present invention is useful as a reagent for screening a compound or a salt thereof that regulates (inhibits or promotes) the activity of the protein of the present invention.
  • the present invention provides a compound which regulates (promotes or inhibits) the activity of the protein of the present invention (eg, an inorganic phosphate uptake activity, an inorganic phosphate transporting activity, etc.) characterized by using the protein of the present invention, or a compound thereof. How to screen salt Provide the law.
  • inorganic phosphate uptake activity is determined by a known method, for example, the method described in Methods in Enzymology, vol. 191, pp. 494-505, 1990, or Measure and compare according to an equivalent method.
  • is obtained by adding a labeled inorganic phosphate to cells capable of expressing the protein of the present invention; and (ii) cells capable of expressing the protein of the present invention in the presence of a test compound.
  • the amount of inorganic phosphate taken into cells when a labeled inorganic phosphate is added is measured, and a compound or a salt thereof that regulates (promotes or inhibits) the activity of the protein of the present invention is screened. . Perform this reaction in an appropriate buffer, and wash off inorganic phosphate that has not been taken up by cells before measurement.
  • Examples of the labeled inorganic phosphate include an inorganic phosphate labeled with a radioisotope (eg, [ 32 P], [ 33 P], etc.).
  • a radioisotope eg, [ 32 P], [ 33 P], etc.
  • the activity of the cells to take up the inorganic phosphate is measured by a known method for measuring the amount of radioactivity, for example, using a radioactivity measuring device.
  • the labeled inorganic phosphate may be reacted with cells expressing the protein of the present invention after being mixed with the test compound, and the labeled inorganic phosphate may be added to cells expressing the protein of the present invention. After contacting, the test compound may be added.
  • the cells capable of expressing the protein of the present invention described above include cells encoding a gene encoding the protein of the present invention in appropriate cells, for example, COS-7 cells, CH0 cells, HEK293 cells, African Xenopus oocytes, and the like.
  • the protein of the present invention is expressed on a cell membrane by being introduced into an animal cell or the like and cultured.
  • the method for culturing cells capable of expressing the protein of the present invention is the same as the above-described method for culturing the transformant of the present invention.
  • the inorganic phosphate transport activity can be determined by a known method, for example, Biochimica et al.
  • a compound that regulates (inhibits or promotes) the activity of the protein of the present invention by measuring the inorganic phosphate transport activity when a buffer solution containing an inorganic phosphate is brought into contact with a protein-containing synthetic lipid bilayer. Or screen the salt.
  • a change in the membrane current that occurs when inorganic phosphate is transported by the protein of the present invention is measured using a known method, for example, a membrane potential fixing method.
  • the synthetic lipid bilayer containing the protein of the present invention can be prepared by, for example, using a purified protein obtained from a membrane fraction of a cell expressing the protein of the present invention according to a known membrane protein purification method, using a phospholipid or the like.
  • the material can be produced according to a known method, for example, a method described in "Patch Clamp Experimental Technique” (Yoshioka Shoten, published in 1996).
  • the inorganic phosphate may be reacted with the lipid bilayer containing the protein of the present invention after being mixed with the test compound, and the inorganic phosphate ion may be added to the lipid bilayer containing the protein of the present invention. After bringing into contact, the test compound may be added.
  • Test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and the like.
  • the inorganic phosphate uptake activity in the case of the above (ii) is about 20% or more, preferably 30% or more, more preferably
  • a test compound that reduces about 50% or more can be selected as a compound that inhibits the activity of the protein of the present invention.
  • the inorganic phosphate transport activity in the case of the above (ii ′) is about 20% or more, preferably 30% or more, more preferably as compared with the case of the above ( ⁇ ).
  • the compound having the activity of promoting the activity of the protein of the present invention is useful as a safe and low-toxic drug for enhancing the physiological activity of the protein of the present invention.
  • the compound having an activity of inhibiting the activity of the protein of the present invention is a safe and low toxic drug for suppressing the physiological activity of the protein of the present invention, for example, renal disease
  • nephropathy nephropathy
  • focal glomerulosclerosis chronic glomerulonephritis
  • minimally altered Nef osteomyelitis mesangial proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy, membranous Proliferative nephritis, crescentic nephritis, lupus nephritis, amyloid kidney, tubulointerstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, renal tumor, chronic rejection after kidney transplantation
  • it is a prophylactic / therapeutic agent for glycemic nephropathy, focal glomerulosclerosis and the like.
  • Compounds or salts thereof obtained using the screening method or the screening kit of the present invention include, for example, peptides, proteins, non-peptide compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissues It is a compound selected from extracts, plasma, etc.
  • the salt of the compound those similar to the aforementioned salts of the peptide of the present invention are used.
  • the gene encoding the protein of the present invention is also expressed in kidney tissues of Wistar Fatty rats (WF rats), Zucker Fatty rats (ZF rats) and spontaneously hypercholesterolemia (SHC rats).
  • Compounds that modulate (preferably inhibit) the expression of the gene encoding the protein of the present invention because they are elevated include, for example, renal diseases (eg, diabetic nephropathy, focal glomerulosclerosis, Chronic glomerulonephritis, minimal change nephrotic syndrome, mesangial proliferative nephritis, intraluminal proliferative nephritis, IgA nephropathy, membranous nephropathy, membranous proliferative nephritis, crescentic nephritis, lupus nephritis, amyloid kidney, urine It can be used as a prophylactic / therapeutic agent for tubulointerstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, renal tumors, chronic rejection after renal transplantation, etc.).
  • renal diseases eg, diabetic nephropathy, focal glomer
  • the polynucleotide (eg, DNA) of the present invention can be used as a reagent for screening a compound or a salt thereof that regulates (preferably, inhibits) the expression of a gene encoding the protein of the present invention.
  • a compound or a salt thereof that regulates (preferably, inhibits) the expression of a gene encoding the protein of the present invention.
  • the screening methods include (iii) culturing cells capable of producing the protein of the present invention, and (iv) culturing cells capable of producing the protein used in the present invention in the presence of a test compound.
  • a screening method characterized by performing a comparison with the case where the method is performed.
  • the expression level of the gene (specifically, the amount of the protein of the present invention or the amount of the mRNA encoding the protein) in the cases (iii) and (iv) is measured and compared.
  • test compound and cells having the ability to produce the protein of the present invention include the same cells as described above.
  • the amount of the protein is measured by a known method, for example, using an antibody recognizing the protein of the present invention, and analyzing the protein present in a cell extract or the like according to a method such as Western analysis, ELISA, or a method analogous thereto. Can be measured.
  • the amount of mRNA can be measured by a known method, for example, Northern hybridization or PCR using a nucleic acid containing SEQ ID NO: 2, SEQ ID NO: 4 or SEQ ID NO: 6 or a part thereof as a probe, or PCR. Measure according to the method Can be
  • a test compound that promotes the expression of the gene encoding the protein of the present invention a test compound that inhibits about 20% or more, preferably 30% or more, more preferably about 50% or more, is obtained by coding the protein of the present invention.
  • the screening kit of the present invention contains the protein or partial peptide used in the present invention or a salt thereof, or a cell capable of producing the protein or partial peptide used in the present invention.
  • Compounds or salts thereof obtained using the screening method or screening kit of the present invention include the test compounds described above, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, and plants. It is a compound selected from an extract, an animal tissue extract, and plasma, or a salt thereof.
  • the salt of the compound those similar to the aforementioned salts of the protein of the present invention are used.
  • a compound or a salt thereof that regulates (preferably inhibits) the activity of the protein of the present invention and a compound or a salt thereof that regulates (preferably inhibits) the expression of a gene encoding the protein of the present invention may be, for example, a renal disease ( E.g., diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome, mesangial proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy, membranous proliferative nephritis , Crescent-forming nephritis, lupus nephritis, amyloid kidney, tubular interstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, renal tumors, chronic rejection after renal transplantation, etc.) It is useful
  • compositions for oral administration include solid or liquid dosage forms, specifically tablets (including dragees and film-coated tablets), pills, granules, powders, capsules (including soft capsules). ), Syrups, emulsions, suspensions, etc. I can do it.
  • Such a composition is produced by a method known per se and contains a carrier, diluent or excipient commonly used in the field of pharmaceuticals. For example, lactose, starch, sucrose, magnesium stearate and the like are used as carriers and excipients for tablets.
  • compositions for parenteral administration for example, injections, suppositories, etc. are used.
  • Injections are intravenous, subcutaneous, intradermal, intramuscular, intravenous, intraarticular. Includes dosage forms such as injections.
  • Such injections are prepared according to a method known per se, for example, by dissolving, suspending or emulsifying the antibody or a salt thereof in a sterile aqueous or oily liquid commonly used for injections.
  • aqueous liquid for injection examples include physiological saline, isotonic solution containing pudose and other adjuvants, and a suitable solubilizing agent such as alcohol (eg, ethanol), Use in combination with polyalcohols (eg, propylene glycol, polyethylene glycol), nonionic surfactants (eg, polysorbate 80, HCO-50 (polyoxyethylen (50iol) adduc tof hydrogenated cas tor oil)), etc. You may.
  • alcohol eg, ethanol
  • polyalcohols eg, propylene glycol, polyethylene glycol
  • nonionic surfactants eg, polysorbate 80, HCO-50 (polyoxyethylen (50iol) adduc tof hydrogenated cas tor oil)
  • oily liquid for example, sesame oil, soybean oil, and the like are used, and benzyl benzoate, benzyl alcohol, and the like may be used in combination as a solub
  • the above-mentioned oral or parenteral pharmaceutical composition is conveniently prepared in the form of a dosage unit so as to conform to the dose of the active ingredient.
  • the dosage unit include tablets, pills, capsules, injections (ampoules), suppositories, etc., and usually 5 to 500 mg, especially 5 to 100 mg for each dosage unit.
  • other dosage forms contain 10 to 250 mg of the above compound.
  • compositions may contain other active ingredients as long as the compound and the above-mentioned compound do not cause an undesirable interaction.
  • the preparations obtained in this way are safe and low toxic, for example, in humans or warm-blooded animals (eg, mice, rats, puppies, higgies, bush, puppies, puppies, To birds, cats, dogs, monkeys, chimpanzees, etc.) orally or parenterally.
  • warm-blooded animals eg, mice, rats, puppies, higgies, bush, puppies, puppies, To birds, cats, dogs, monkeys, chimpanzees, etc.
  • the dose of the compound or a salt thereof varies depending on its action, target disease, subject of administration, route of administration, and the like.
  • the activity of the protein of the present invention is regulated for the purpose of treating diabetic nephropathy ( When a compound or a salt thereof is orally administered, the compound or a salt thereof is generally administered in an amount of about 0.1 to 100 mg, preferably about 1. 0 to 50 mg, more preferably about 1.0 to 20 mg is administered.
  • the single dose of the compound or a salt thereof varies depending on the administration subject, target disease, and the like.
  • the activity of the protein of the present invention is regulated for the purpose of treating diabetic nephropathy.
  • a compound (or a salt thereof) which is preferably (inhibited) is administered to an adult (with a body weight of 60 kg) usually in the form of an injection
  • the compound or a salt thereof is used in an amount of about 0.01 to 30 mg, preferably about 0.1 to 30 mg per day.
  • 1-20 mg, more preferably about 0.1-10 mg will be administered by intravenous injection.
  • the dose can be administered in terms of weight per 60 kg.
  • An antibody against the protein of the present invention (hereinafter sometimes abbreviated as the antibody of the present invention) can specifically recognize the protein of the present invention, so that the quantification of the protein of the present invention in a test solution can be performed.
  • the antibody of the present invention can be used for quantification by the San Germanti immunoassay.
  • one of the antibodies is the N-terminal of the protein of the present invention.
  • the other antibody is an antibody that reacts with the C-terminal of the protein of the present invention.
  • the protein of the present invention can be quantified using a monoclonal antibody against the protein of the present invention (hereinafter sometimes referred to as the monoclonal antibody of the present invention), and can also be detected by tissue staining or the like.
  • the antibody molecule itself may be used, or F (ab ') 2 , Fab' or Fab fraction of the antibody molecule may be used.
  • the method for quantifying the protein of the present invention using the antibody of the present invention is not particularly limited, and may be an antibody, antigen or antibody-antigen complex corresponding to the amount of antigen (eg, the amount of protein) in the test solution. Any method can be used as long as the amount is determined 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 preferably used, but it is particularly preferable to use a sandwich method described later in terms of sensitivity and specificity.
  • Examples of the labeling agent used in the assay method using the labeling substance for example, a radioactive isotope (e.g., [125 iota], [131 iota], [3 ⁇ 4], TO, TM, [33 P], a [35 s] ), Fluorescent substances (eg, cyanine fluorescent dyes (eg, Cy2, Cy3, Cy5, Cy5.5, Cy7 (manufactured by Amersham Bioscience)), fluorescamine, fluorescein isothiocynate, etc.), enzymes (examples) , ⁇ -lactosidase,) 3-darcosidase, alkaline phosphatase, peroxidase, malate dehydrogenase, etc., luminescent substances (eg, luminol, luminol derivatives, reluciferin, lucigenin, etc.), element of laninide Are used. Further, a biotin-avidin system can be used for binding the antibody
  • the carrier include insoluble polysaccharides such as agarose, dextran, and cellulose; synthetic resins such as polystyrene, polyacrylamide, and silicon; and glass.
  • test solution was added to the insolubilized monoclonal antibody of the present invention.
  • Primary reaction and further reacted with another labeled monoclonal antibody of the present invention (secondary reaction), and then the activity of the labeling agent on the insolubilized carrier is measured. Can be quantified.
  • the primary reaction and the secondary reaction may be performed in the reverse order, may be performed simultaneously, or may be performed at staggered times.
  • the labeling agent and the method of insolubilization can be the same as those described above.
  • the antibody used for the solid phase antibody or the labeling antibody does not necessarily need to be one kind, and two or more kinds of antibodies are used for the purpose of improving the measurement sensitivity and the like. Mixtures may be used.
  • the monoclonal antibody of the present invention used in the primary reaction and the secondary reaction is an antibody having a different site to which the protein of the present invention binds. It is preferably used. That is, when the antibody used in the primary reaction and the secondary reaction is, for example, the antibody used in the secondary reaction recognizes the C-terminal of the protein of the present invention, the antibody used in the primary reaction is preferably Is an antibody that recognizes other than the C-terminal, for example, the N-terminal.
  • the monoclonal antibody of the present invention can be used in a measurement system other than the sandwich method, for example, a competition method, an immunometric method, or a nephrometry method.
  • the antigen in the test solution and the labeled antigen are allowed to react competitively with the antibody, and then the unreacted labeled antigen (F) and the labeled antigen (B) bound to the antibody are separated.
  • BZF separation The labeling amount of either B or F is measured, and the amount of antigen in the test solution is quantified.
  • a soluble phase is used as the antibody
  • B / F separation is carried out 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
  • An immobilization method using a soluble antibody as the first antibody and an immobilized antibody as the second antibody is used.
  • the ability to separate the solid phase from the liquid phase after a competitive reaction between the antigen in the test wave and the immobilized antigen for a certain amount of the labeled antibody, or the antigen in the test solution Is allowed to react with an excess amount of the labeled antibody, and then the immobilized antigen is added to bind the unreacted labeled antibody to the solid phase, and then the solid phase and the liquid phase are separated. Then either In nephrometry, 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 sediment is obtained, laser nephrometry utilizing laser scattering is preferably used.
  • the system for measuring the protein of the present invention may be constructed by adding ordinary technical considerations to those skilled in the art to the ordinary conditions and procedures in each method. For details of these general technical means, reference can be made to reviews and documents.
  • the protein of the present invention can be quantified with high sensitivity by using the antibody of the present invention.
  • renal disease eg, diabetic nephropathy
  • Focal glomerulosclerosis chronic glomerulonephritis
  • minimal change nephrotic syndrome mesangial proliferative nephritis
  • endoproliferative nephritis IgA nephropathy, membranous nephropathy, membranous proliferative nephritis, crescent-forming nephritis
  • Lupus nephritis amyloid Renal disease
  • tubulointerstitial nephritis acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, renal tumors, chronic rejection after renal transplantation, etc.
  • renal disease eg, diabetic nephropathy
  • Focal glomerulosclerosis chronic glomerulonephritis
  • minimal change nephrotic syndrome mesangial proliferative nephritis
  • the antibody of the present invention can be used for detecting the protein of the present invention present in a subject such as a body fluid or a tissue.
  • preparation of an antibody column used for purifying the protein of the present invention, detection of the protein of the present invention in each fraction at the time of purification, analysis of the behavior of the protein of the present invention in test cells, etc. can be used for (3) Gene diagnostics
  • the DNA of the present invention can be used, for example, in humans or warm-blooded animals (eg, rats, mice, guinea pigs, egrets, birds, higgies, pigs, pigs, dogs, cats, dogs, monkeys) by using them as probes. , Chimpanzees, etc.) can detect abnormalities (gene abnormalities) in DNA or mRNA encoding the protein of the present invention or a partial peptide thereof.
  • warm-blooded animals eg, rats, mice, guinea pigs, egrets, birds, higgies, pigs, pigs, dogs, cats, dogs, monkeys
  • abnormalities gene abnormalities
  • the above-described gene diagnosis using the DNA of the present invention can be performed, for example, by the known Northern hybridization, PCR-SSCP method (Genomics, Vol. 5, pp. 874-879, 1989, Proceedings of the National Academy of Sciences of the United
  • renal disease eg, diabetic nephropathy, focal thread
  • Glomerulosclerosis chronic glomerulonephritis, minimal change nephrotic syndrome, mesangial proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy, membranous proliferative nephritis, crescentic nephritis, lupus nephritis, Diagnosed as having a high likelihood of amyloid kidney, tubular interstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, renal tumor, chronic rejection after kidney transplantation, etc.
  • the antisense polynucleotide of the present invention which can complementarily bind to the DNA of the present invention and suppress the expression of the DNA, has low toxicity, and functions of the protein of the present invention or the DNA of the present invention in vivo.
  • renal diseases eg, diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome, mesangial proliferative nephritis, endoproliferative nephritis, IgA Nephropathy, membranous nephropathy, membranous proliferative nephritis, crescentic nephritis, lupus nephritis, amyloid kidney, tubulointerstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema ⁇ Can be used as a prophylactic or therapeutic agent for kidney tumors, chronic rejection after kidney transplantation, etc.).
  • renal diseases eg, diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome, mesangial proliferative nephriti
  • the antisense polynucleotide When used as the above-mentioned prophylactic / therapeutic agent, it can be formulated and administered according to a method known per se.
  • the above-mentioned antisense polynucleotide is used alone or in a suitable vector such as a retrovirus vector, an adenovirus vector, an adenovirus associated virus vector, and the like, and thereafter, human or mammal (for example, it can be administered orally or parenterally to rats, puppies, sheep, sheep, bush, puppies, cats, dogs, monkeys, etc.).
  • the antisense polynucleotide can be administered as it is or in the form of a formulation together with a physiologically acceptable carrier such as an auxiliary agent for promoting uptake, and can be administered by a gene gun or a catheter such as a hydrogel catheter. Alternatively, they can be aerosolized and administered topically into the trachea as an inhalant.
  • the above antisense polynucleotide is formulated alone or together with a carrier such as ribosome (injection), and is intravenously, subcutaneously, It may be administered to a lesion or the like.
  • the dosage of the antisense polynucleotide varies depending on the target disease, the administration subject, the administration route, and the like.For example, when the antisense polynucleotide of the present invention is administered for the purpose of treating diabetic nephropathy, In general, for adults (body weight 60 kg), about 0.1 to 100 mg of the antisense polynucleotide is administered per day. You.
  • the antisense polynucleotide can also be used as a diagnostic oligonucleotide probe for examining the presence or expression of the DNA of the present invention in tissues or cells.
  • RNA containing a part of the RNA encoding the protein of the present invention a double-stranded RNA containing a part of the RNA encoding the protein of the present invention, a lipozyme containing a part of the RNA encoding the protein of the present invention, etc.
  • renal diseases eg, Nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome group, mesangial proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy, membranous proliferative nephritis, crescent formation
  • renal diseases eg, Nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome group, mesangial proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy, membranous proliferative nephritis, crescent formation
  • lupus nephritis amyloid kidney, tubular interstitial nephritis, acute tubular necrosis,
  • the double-stranded RNA can be designed and manufactured based on the sequence of the polynucleotide of the present invention according to a known method (eg, Nature, 411, 494, 2001).
  • the lipozyme can be designed and manufactured based on the sequence of the polynucleotide of the present invention according to a known method (eg, TRENDS in Molecular Medicine, Vol. 7, p. 221, 2001). For example, it can be produced by linking a known lipozyme to a part of RNA encoding the protein of the present invention.
  • a part of the RNA encoding the protein of the present invention includes a portion (RNA fragment) adjacent to the cleavage site on the RNA of the present invention which can be cleaved by a known lipozyme.
  • RNA or lipozyme When the above-described double-stranded RNA or lipozyme is used as the above-mentioned prophylactic / therapeutic agent, it can be formulated and administered in the same manner as an antisense polynucleotide.
  • the antibody of the present invention which has the activity of neutralizing the activity of the protein of the present invention, includes, for example, For example, renal diseases (eg, diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, microdegenerative nephrotic syndrome, mesangial proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy, Membranous proliferative nephritis, crescentic nephritis, lupus nephritis, amyloid kidney, tubulointerstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, renal tumor, chronic after renal transplantation Rejection) can be used as a prophylactic or therapeutic agent.
  • renal diseases eg, diabetic nephropathy, focal glomerulosclerosis, chronic glomerul
  • the antibodies of the present invention can be administered by themselves or as a suitable pharmaceutical composition.
  • the pharmaceutical composition used for the administration contains the antibody or a salt thereof and a pharmacologically acceptable carrier, diluent or excipient.
  • Such compositions are provided in dosage forms suitable for oral or parenteral administration.
  • compositions for oral administration include solid or liquid dosage forms, specifically tablets (including sugar-coated tablets and film-coated tablets), pills, granules, powders, capsules (soft capsules) And syrups, emulsions, suspensions and the like.
  • Such a composition is produced by a known method and contains a carrier, diluent or excipient commonly used in the pharmaceutical field.
  • a carrier for example, lactose, starch, sucrose, magnesium stearate and the like are used as carriers and excipients for tablets.
  • compositions for parenteral administration for example, injections, suppositories, etc. are used.
  • Injections include intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections, intravenous injections, etc. Include shapes.
  • Such injections are prepared according to known methods, for example, by dissolving, suspending or emulsifying the antibody or a salt thereof in a sterile aqueous or oily liquid commonly used for injections.
  • aqueous liquid for injection examples include physiological saline, isotonic solution containing pudose and other adjuvants, and a suitable solubilizing agent such as alcohol (eg, ethanol) and polyalcohol ( For example, propylene glycol, polyethylene glycol), a nonionic surfactant [eg, polysorbate 80, HCO-50 (polyoxyethylene (50 mol) addoct of hydrogenated catalyst)], etc. may be used in combination.
  • alcohol eg, ethanol
  • polyalcohol For example, propylene glycol, polyethylene glycol
  • a nonionic surfactant eg, polysorbate 80, HCO-50 (polyoxyethylene (50 mol) addoct of hydrogenated catalyst)
  • oily liquid for example, sesame oil, soybean oil, and the like are used, and benzyl benzoate, benzyl alcohol, and the like may be used in combination as a solubilizing agent.
  • the prepared injection solution is usually Filled into ampou
  • the above-mentioned oral or parenteral pharmaceutical composition is conveniently prepared in the form of a dosage unit so as to conform to the dose of the active ingredient.
  • dosage unit forms include tablets, pills, capsules, injections (ampoules), and suppositories, and usually 5 to 500 mg per dosage unit form, especially 5 to 500 mg for injections.
  • 100 mg, and other dosage forms contain 10 to 250 mg of the above antibody.
  • compositions may contain another active ingredient as long as the composition does not cause an undesirable interaction with the above-mentioned antibody.
  • the prophylactic / therapeutic agent for the above-mentioned diseases containing the antibody of the present invention has low toxicity and can be used as it is as a liquid or as a pharmaceutical composition of an appropriate dosage form, in humans or mammals.
  • the antibody of the present invention is used as a single dose.
  • about 0.01 to 20 mg / kg body weight, preferably about 0.1 to 10 mg / kg body weight, more preferably about 0.1 to 5 mg / kg body weight, about 1 to 5 times a day, preferably 1 to 5 times a day It is convenient to administer as an injection about three times. In the case of other parenteral administration and oral administration, an equivalent dose can be administered. If the symptoms are particularly severe, the dose may be increased accordingly.
  • the antibody of the present invention can be used, for example, for renal diseases (eg, diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome, mesangial proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy, membranous proliferative nephritis, crescentic nephritis, lupus nephritis, amyloid kidney, tubulointerstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal It is also useful as a diagnostic agent for edema, kidney tumors, chronic rejection after kidney transplantation, etc.).
  • renal diseases eg, diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome, mes
  • the present invention relates to a DNA encoding the exogenous protein of the present invention (hereinafter abbreviated as the exogenous DNA of the present invention) or a mutant DNA thereof (sometimes abbreviated as the exogenous mutant DNA of the present invention). And a non-human mammal having the formula:
  • Non-human mammals having the exogenous DNA of the present invention or the mutant DNA thereof include unfertilized eggs, fertilized eggs, germ cells including spermatozoa and their progenitor cells, and the like.
  • a calcium phosphate method, an electric pulse method, a ribofection method It can be produced by transferring the desired DNA by an agglutination method, microinjection method, particle gun method, DEAE-dextran method, or the like.
  • the exogenous DNA of the present invention can be transferred to somatic cells, organs of living organisms, tissue cells, and the like, and can be used for cell culture, tissue culture, and the like.
  • the DNA-transferred animal of the present invention can also be produced by fusing cells with the above-mentioned germ cells by a cell fusion method known per se.
  • non-human mammals for example, porcupines, pigs, higgins, goats, magpies, dogs, cats, guinea pigs, hamsters, mice, rats and the like are used.
  • mice for example, pure strains such as C57BL / 6 strain, DBA2 strain, etc.
  • "Mammals" in a recombinant vector that can be expressed in mammals include humans and the like in addition to the above-mentioned non-human mammals.
  • the exogenous DNA of the present invention refers not to the DNA of the present invention originally possessed by non-human mammals but to the DNA of the present invention once isolated and extracted from the mammal.
  • Examples of the mutant DNA of the present invention include those in which a mutation (for example, mutation) has occurred in the base sequence of the original DNA of the present invention, specifically, addition or deletion of a base, substitution with another base, or the like.
  • the DNA in which the occurrence has occurred is used, and the abnormal DNA is also included.
  • the abnormal DNA refers to a DNA that expresses an abnormal protein of the present invention, and for example, a DNA that expresses a protein that suppresses the function of the normal protein of the present invention is used.
  • the exogenous DNA of the present invention may be derived from a mammal that is the same or different from the animal of interest.
  • various mammals having the DNA of the present invention having a high homology with the human for example, egret, dog, cat, guinea pig, hamster, rat, mouse, etc.
  • Microinjection of a DNA construct e.g., vector, etc.
  • a human DNA of the present invention is bound downstream of one of various promoters capable of expressing DNA of origin from a fertilized egg of a target mammal, for example, a mouse fertilized egg
  • a DNA-transferred mammal that highly expresses the DNA of the present invention can be produced.
  • Examples of the expression vector of the protein of the present invention include plasmids derived from Escherichia coli, plasmids derived from Bacillus subtilis, plasmids derived from yeast, bacteriophages such as ⁇ phage, retroviruses such as Moroni leukemia virus, and vaccinia viruses. Alternatively, animal viruses such as baculovirus are used. Among them, a plasmid derived from Escherichia coli, a plasmid derived from Bacillus subtilis or a plasmid derived from yeast are preferably used.
  • promoters that regulate the above DNA expression include: (i) virus (eg, simian virus, cytomegalovirus, Moroni leukemia virus) Promoters of DNA derived from humans, JC virus, breast cancer virus, poliovirus, etc.), and (ii) Promoters derived from various mammals (human, egret, dog, cat, guinea pig, hamster, rat, mouse, etc.) 1.For example, albumin, insulin II, perobrakin II, elas ubiquitin, erythropoietin, endothelin, muscle creatine kinase, glial fibrillary acidic protein, gliubithion S-transferase, platelet-derived growth factor / 3, Keratin Kl, K10 and ⁇ 14, collagen type I and type II, cyclic AMP-dependent protein kinase i3 I subunit, dystrophin, tartrate-resistant lipophilic phosphatase, atrial sodium diuretic factor
  • a cytomegalovirus promoter that can be highly expressed in the whole body, a promoter of a human peptide chain elongation factor 1a (EF-1), a human and a chicken / 3-actin promoter are preferable. is there.
  • EF-1 human peptide chain elongation factor 1a
  • a human and a chicken / 3-actin promoter are preferable. is there.
  • the vector preferably has a sequence that terminates transcription of a messenger RNA of interest in a DNA-transferred mammal (generally called terminator).
  • terminator a sequence that terminates transcription of a messenger RNA of interest in a DNA-transferred mammal
  • each vector may be derived from a virus or a mammal.
  • a DNA sequence can be used, and preferably, SV40 terminator of simian virus or the like is used.
  • splicing signals of each DNA, enhancer region, and part of eukaryotic DNA introns for the purpose of further expressing the desired foreign DNA. It is also possible to connect a promoter 5 ′ upstream of the promoter region, between the promoter region and the translation region, or 3 ′ downstream of the translation region.
  • the normal translation region of the protein of the present invention is human or various mammals (eg, egrets, dogs, cats, guinea pigs, eight musters, rats, mice, etc.).
  • Liver, kidney, thyroid cells, fibroblasts All or part of genomic DNA from cell-derived DNA and various commercially available genomic DNA libraries, or complementary DNA prepared by known methods from dried, kidney, thyroid, or fibroblast-derived RNA. It can be obtained as a raw material.
  • a foreign abnormal DNA can produce a translation region obtained by mutating the translation region of a normal protein obtained from the above-described cells or tissues by a point mutagenesis method.
  • the translation region can be prepared as a DNA construct that can be expressed in a transposed animal by a conventional DNA engineering technique in which it is ligated downstream of the above-mentioned promoter and, if desired, upstream of the transcription termination site.
  • Transfer of the exogenous DNA of the present invention at the fertilized egg cell stage is ensured to be present in all germ cells and somatic cells of the target mammal.
  • the presence of the exogenous DNA of the present invention in the germ cells of the produced animal after DNA transfer means that all the progeny of the produced animal retain the exogenous DNA of the present invention in all of the germ cells and somatic cells Means that.
  • the progeny of such animals inheriting the exogenous DNA of the present invention have the exogenous DNA of the present invention in all of their germinal and somatic cells.
  • the non-human mammal to which the exogenous normal DNA of the present invention has been transferred is confirmed to stably maintain exogenous DNA by mating, and should be subcultured as an animal having the DNA in a normal breeding environment. Can be done.
  • Transfer of the exogenous DNA of the present invention at the fertilized egg cell stage is ensured to be present in excess in all germ cells and somatic cells of the target mammal.
  • Excessive presence of the exogenous DNA of the present invention in the germinal cells of the animal after transfer of the DNA indicates that the offspring of the animal in which the animal is produced excessively contains the exogenous DNA of the present invention in all of its germ cells and somatic cells. Means to have. The offspring of an animal of this species that has inherited the exogenous DNA of the present invention, In excess.
  • the non-human mammal having the normal DNA of the present invention expresses the normal DNA of the present invention at a high level, and eventually promotes the function of endogenous normal DNA to thereby finally produce the protein of the present invention. May develop hyperfunction, and can be used as a model animal for the disease. For example, using the normal DNA-transferred animal of the present invention, it is possible to elucidate the pathological mechanism of the hyperactivity of the protein of the present invention and diseases associated with the protein of the present invention, and to examine a method for treating these diseases. It is possible.
  • a prophylactic / therapeutic agent for a disease associated with the protein of the present invention such as kidney Diseases (e.g., diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimally altered nephritis syndrome, mesangial proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy , Membranous proliferative nephritis, crescentic nephritis, lupus nephritis, amyloid kidney, tubular interstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, renal tumor, after kidney transplantation It can also be used for screening tests for prophy
  • kidney Diseases e.g., diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis,
  • the non-human mammal having the foreign abnormal DNA of the present invention should be subcultured in a normal breeding environment as an animal having the DNA after confirming that the foreign DNA is stably retained by the crossing. Can be done. Further, the desired foreign DNA can be incorporated into the above-mentioned plasmid and used as a source substance.
  • the DNA construct with the promoter can be prepared by ordinary DNA engineering techniques. The transfer of the abnormal DNA of the present invention at the fertilized egg cell stage is ensured to be present in all germ cells and somatic cells of the target mammal.
  • the presence of the abnormal DNA of the present invention in the germinal cells of a produced animal after DNA transfer means that the offspring of the produced animal have the abnormal DNA of the present invention in all of its germinal and somatic cells.
  • the offspring of this type of animal that inherited the exogenous DNA of the present invention All germ cells and somatic cells have the abnormal DNA of the present invention.
  • the abnormal DNA of the present invention is highly expressed, and the function of the protein of the present invention is ultimately reduced by inhibiting the function of endogenous normal DNA.
  • Inactive refractory disease may occur, and it can be used as a disease model animal. For example, it is possible to elucidate the pathological mechanism of the functionally inactive refractory state of the protein of the present invention and to examine a method for treating this disease using the abnormal DNA transgenic animal of the present invention.
  • the abnormal DNA highly expressing animal of the present invention can be used to inhibit the function of a normal protein by the abnormal protein of the present invention in the function-inactive refractory disease of the protein of the present invention (dominant negative activity). Action).
  • the preventive / therapeutic agent for the protein or functionally inactive type refractory of the present invention is used.
  • renal diseases eg, diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome, mesangial proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy, Membranous proliferative nephritis, crescentic adult nephritis, lupus nephritis, amyloid kidney, tubular interstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, renal tumor, after kidney transplantation It can also be used for screening tests for preventive
  • the DNA-transferred animal of the present invention it is possible to examine the clinical symptoms of diseases related to the protein of the present invention, including the inactive type refractory type of the protein of the present invention, and the like. It is possible to obtain more detailed pathological findings in each organ of the disease model related to the protein of the present invention, to develop new treatment methods, and to contribute to the research and treatment of secondary diseases caused by the disease. it can.
  • the DNA-transferred animal of the present invention in order to use the DNA-transferred animal of the present invention to develop a therapeutic agent for a disease associated with the protein of the present invention, including the inactive type refractory type of the protein of the present invention, Using a test method and a quantitative method, it is possible to provide an effective and rapid screening method for a therapeutic agent for the disease. Furthermore, using the DNA transfer product of the present invention or the exogenous DNA expression vector of the present invention, it is possible to study and develop a method for treating a DNA associated disease of the present invention for DNA.
  • the present invention provides a non-human mammalian embryonic stem cell in which the DNA of the present invention has been inactivated, and a non-human mammal deficient in expression of the DNA of the present invention.
  • the present invention (1) a non-human mammalian embryonic stem cell in which the DNA of the present invention has been inactivated,
  • the DNA is inactivated by introducing a repo overnight gene (eg, an 8-galactosidase gene derived from Escherichia coli), and the repo overnight gene is under the control of a promoter for the DNA of the present invention.
  • a repo overnight gene eg, an 8-galactosidase gene derived from Escherichia coli
  • a compound that promotes or inhibits the activity of the promoter of DNA of the present invention which comprises administering a test compound to the animal described in (7) and detecting the expression of a repo overnight gene; A method for screening the salt is provided.
  • a non-human mammalian embryonic stem cell in which the DNA of the present invention has been inactivated refers to a DNA of the present invention possessed by the non-human mammal, which is artificially mutated to suppress the expression ability of the DNA, or By substantially losing the activity of the protein of the present invention encoded by the DNA, the DNA substantially does not have the ability to express the protein of the present invention (hereinafter, referred to as the knockout DNA of the present invention).
  • ES cells non-human mammal embryonic stem cells
  • non-human mammal the same one as described above is used.
  • the method of artificially mutating the DNA of the present invention can be performed, for example, by deleting a part or all of the DNA sequence and inserting or substituting another DNA by a genetic engineering technique.
  • the knockout DNA of the present invention may be prepared by shifting the reading frame of codons or disrupting the function of the promoter or exon.
  • Non-human mammalian embryonic stem cells in which the DNA of the present invention has been inactivated include, for example, isolating the DNA of the present invention from a non-human mammal of interest, and adding a neomycin resistance gene to its exon portion.
  • a drug resistance gene typified by the idalomycin resistance gene, or a repo allele gene typified by lacZ (3-galactosidase gene) and cat (chloramphenicol acetyltransferase gene) Or the insertion of a DNA sequence that terminates gene transcription (for example, a po1yA addition signal) into the intron between the exons, resulting in a complete messenger RNA.
  • a DNA strand having a DNA sequence constructed so as to result in the disruption of the gene by preventing the synthesis of ) Is introduced into the chromosome of the animal by, for example, homologous recombination, and the obtained ES cells are subjected to Southern hybridization using the DNA sequence on or near the DNA of the present invention as a probe.
  • the knockout ES cells of the present invention are selected by analyzing the DNA sequence on the targeting vector and the DNA sequence of the neighboring region other than the DNA of the present invention used for the preparation of the targeting vector by using the primers as primers.
  • ES cells from which the DNA of the present invention is inactivated by the homologous recombination method or the like for example, those already established as described above may be used, or the method described in the known Evans and Kaufma method may be used. A newly established one may be used.
  • mouse ES cells generally, 129 ES cells are currently used, but since the immunological background is not clear, it is a pure line that substitutes the immunological genetic background.
  • the BDFt mice C57BLZ6 and DBAZ2 BDFi mice can be used satisfactorily because they have the advantage of high number of eggs collected and robust eggs, and have C57BLZ6 mice as a background.
  • ES cells can be advantageously used when a pathological model mouse is created, because the genetic background can be replaced by C57BLZ6 mice by backcrossing with C57BLZ6 mice.
  • blastocysts 3.5 days after fertilization are generally used, but efficiency is also improved by collecting embryos at the 8-cell stage and culturing them up to blastocysts. Many early embryos can be obtained well.
  • Either male or female ES cells may be used, but male ES cells are generally more convenient for producing a breeding line chimera. It is also desirable to discriminate between males and females as soon as possible in order to reduce the complexity of culturing.
  • An example of a method for determining the sex of ES cells is a method of amplifying and detecting a gene in the sex-determining region on the Y chromosome by PCR.
  • this method conventionally, for example G-banding method, it requires about 1 0 6 cells for karyotype analysis, the need in ES cell number of about 1 colony (about 5 0) Therefore, the primary selection of ES cells in the early stage of culture can be performed by discriminating between male and female, and the early stage of culture can be greatly reduced by enabling the selection of male cells at an early stage.
  • the secondary selection can be performed, for example, by confirming the number of chromosomes by the G-banding method.
  • Embryonic stem cell lines obtained in this way usually have very good proliferative potential, but must be carefully subcultured because they tend to lose their ontogenetic potential.
  • a suitable feeder cell such as STO fibroblasts
  • a carbon dioxide incubator preferably 5% carbon dioxide, 95% air or 5% oxygen
  • LIF 1-10000 U / ml
  • 5% CO 2, 90% air at about 37.
  • trypsin ZEDTA solution usually 0.001 to 0.5% trypsin ZO. L
  • 5 mM EDTA preferably, about 0.1% trypsin / ImM EDTA
  • ES cells are differentiated into various types of cells, such as parietal, visceral, and cardiac muscle, by monolayer culture up to high density or suspension culture until cell clumps are formed under appropriate conditions.
  • MJ Evans and MH Kaufman Nature, Vol. 292, pp. 154, 1981; GR Martin Proceedings of the National Academy of Sciences' Ob Science Sciences Proc. Natl. Acad. Sci. USA) 78, 7634, 1981; TC Doetschman et al., Journal of Embrio Morphology, Vol. 87, p. 27. 1985].
  • the DNA-deficient cells of the present invention obtained by differentiating the ES cells of the present invention are useful in the cell biology of the protein of the present invention in the mouth of in vivo.
  • the non-human mammal deficient in DNA expression of the present invention can be distinguished from a normal animal by measuring the mRNA amount of the animal using a known method and indirectly comparing the expression levels.
  • non-human mammal the same one as described above is used.
  • the non-human mammal deficient in expression of the DNA of the present invention may be obtained, for example, by introducing the evening-getting vector prepared as described above into a mouse embryonic stem cell or a mouse egg cell, and introducing the evening-getting vector into the DNA of the present invention.
  • the cells in which the DNA of the present invention has been knocked out are subjected to Southern hybridization analysis using the DNA sequence on or near the DNA of the present invention as a probe or the DNA sequence on the overnight getter vector, and the target DNA.
  • the DNA can be determined by a PCR method using the DNA sequence of a neighboring region other than the DNA of the present invention derived from the mouse used as the primer vector as a primer.
  • a non-human mammalian embryonic stem cell is used, a cell line in which the DNA of the present invention has been inactivated by gene homologous recombination is cloned, and the cell is used at an appropriate time, for example, at the 8-cell stage.
  • the chimeric embryo is injected into a non-human mammal embryo or blastocyst, and the resulting chimeric embryo is transplanted into the uterus of the pseudo-pregnant non-human mammal.
  • the produced animal also has a normal DNA locus of the present invention. This is a chimera animal composed of both an artificial cell and an artificially mutated cell having the DNA locus of the present invention.
  • all tissues are artificially obtained from the population obtained by crossing such a chimeric individual with a normal individual. It can be obtained by selecting individuals composed of cells having the DNA locus of the present invention to which a mutation has been added, for example, by judging coat color.
  • the individual obtained in this manner is usually an individual having a heterozygous expression of the protein of the present invention, which is crossed with an individual having a heterozygous expression of the protein of the present invention. A homozygous deficient individual can be obtained.
  • a transgenic non-human mammal having a chromosome into which the evening-getting vector has been introduced can be obtained by injecting a DNA solution into the nucleus of the egg cell by a microinjection method.
  • the animal obtained by the crossing should be confirmed to be knocked out of the DNA and subjected to rearing in a normal rearing environment. Can be.
  • the germline can be obtained and maintained according to a conventional method. That is, by crossing male and female animals having the inactivated DNA, homozygous animals having the inactivated DNA on both homologous chromosomes can be obtained.
  • the obtained homozygous animal can be efficiently obtained by rearing the mother animal in such a manner that one normal individual and a plurality of homozygous animals are obtained.
  • By mating male and female heterozygous goats, a homozygote having the inactivated DNA is obtained.
  • the non-human mammalian embryonic stem cells in which the DNA of the present invention has been inactivated are the DN of the present invention.
  • the non-human mammal deficient in expression of the DNA of the present invention lacks various biological activities that can be induced by the protein of the present invention
  • the non-human mammal may be inactivated by the inactivation of the biological activity of the protein of the present invention.
  • the non-human mammal deficient in DNA expression of the present invention can be used for screening for a compound having a therapeutic / preventive effect against diseases caused by DNA deficiency or damage of the present invention.
  • the present invention is characterized in that a test compound is administered to a non-human mammal deficient in expression of the DNA of the present invention, and changes in the animal are observed and measured.
  • renal diseases eg, diabetic nephropathy, focal glomerulosclerosis, chronic glomer
  • Examples of the non-human mammal deficient in DNA expression of the present invention used in the screening method include those described above.
  • Test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and plasma.These compounds are novel compounds. Or a known compound.
  • a non-human mammal deficient in expression of the DNA of the present invention is treated with a test compound and compared with a non-treated control animal to examine changes in organs, tissues, disease symptoms and the like of the animal.
  • the therapeutic / preventive effect of the test compound can be tested as an index.
  • test compound for example, oral administration, intravenous injection, etc. are used, and the method can be appropriately selected according to the symptoms of the test animal, the properties of the test compound, and the like.
  • the dose of the test compound can be appropriately selected according to the administration method, the properties of the test compound, and the like.
  • a test compound when screening for a compound having a therapeutic / preventive effect on diabetic nephropathy, etc., a test compound is administered to a non-human mammal deficient in expression of the DNA of the present invention, and the test compound non-administration group and the renal function Observe, over time, differences in urine protein excretion and glomerular filtration rate, which are indicators, or glomerular sclerosis rate, which is an indicator of renal dysfunction.
  • the disease symptom of the test animal is improved by about 10% or more, preferably about 30% or more, more preferably about 50% or more. It can be selected as a compound having a therapeutic / preventive effect on the above diseases.
  • the compound obtained by using the screening method is a compound selected from the test compounds described above, and has a therapeutic / preventive effect against a disease caused by deficiency or damage of the protein of the present invention. It can be used as a medicament such as a safe and low toxic prophylactic / therapeutic agent for the disease. Further, a compound derived from the compound obtained by the above screening can also be used.
  • the compound obtained by the screening method may form a salt.
  • the salt of the compound include physiologically acceptable acids (eg, inorganic acids, organic acids, etc.) and bases (eg, alkali metals). And the like, and especially preferred are physiologically acceptable acid addition salts.
  • physiologically acceptable acids eg, inorganic acids, organic acids, etc.
  • bases eg, alkali metals
  • physiologically acceptable acid addition salts include salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.) and organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, etc.). Acid, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc.).
  • a drug containing the compound or a salt thereof obtained by the screening method can be produced in the same manner as the above-mentioned drug containing the protein of the present invention.
  • the preparations obtained in this way are safe and have low toxicity, for example, in humans or mammals (eg, rats, mice, guinea pigs, egrets, sheep, sheep, butter, horseshoe, horses, cats, cats). And monkeys).
  • the dose of the compound or a salt thereof varies depending on the target disease, the subject of administration, the administration route, and the like. For example, when the compound is orally administered, generally the diabetic kidney of an adult (with a body weight of 60 kg) is used. For patients with sickness, about 0.1 to 100 mg, preferably about 1.0 to 50 mg, more preferably about 1.0 to 20 mg of the compound is administered per day.
  • the single dose of the compound varies depending on the administration subject, target disease and the like.
  • the compound is usually in the form of an injection and is usually used for adult (as 60 kg) sugar urine.
  • the compound When administered to patients with pathologic nephropathy, about 0.01 to 30 mg, preferably about 0.1 to 2 Omg, and more preferably about 0.1 to 1 Omg of the compound is administered intravenously per day. It is conveniently administered by In the case of other animals, it is also possible to administer the amount converted per 60 kg.
  • the present invention relates to the activity of a promoter for the DNA of the present invention, which comprises administering a test compound to a non-human mammal deficient in expressing the DNA of the present invention and detecting the expression of a repo allele.
  • a method for screening a compound or a salt thereof that promotes or inhibits cell growth comprises administering a test compound to a non-human mammal deficient in expressing the DNA of the present invention and detecting the expression of a repo allele.
  • the non-human mammal deficient in expression of the DNA of the present invention includes, among the aforementioned non-human mammals deficient in the expression of the DNA of the present invention, the DNA of the present invention obtained by introducing a repo overnight gene. Those inactivated and capable of expressing the repo overnight gene under the control of the promoter for the DNA of the present invention are used.
  • test compound examples include the same compounds as described above.
  • the reporter gene the same one as described above is used, and a ⁇ -galactosidase gene (1 ac Z), a soluble alkaline phosphatase gene or a luciferase gene is preferable.
  • the reporter gene is under the control of the promoter of the DNA of the present invention.
  • the activity of the promoter can be detected. For example, when a part of the DNA region encoding the protein of the present invention is replaced by the iS_galactosidase gene (1 ac Z) derived from Escherichia coli, a tissue that originally expresses the protein of the present invention may 3-galactosidase is expressed instead of this protein.
  • a reagent that serves as a substrate for i3-galactosidase such as 5-bromo-4-chloro-3-indolyl-j8-galactopyranoside (X-gal)
  • X-gal 5-bromo-4-chloro-3-indolyl-j8-galactopyranoside
  • the expression state of the protein of the present invention in an animal body can be observed.
  • the protein-deficient mouse of the present invention or a tissue section thereof is fixed with dartalaldehyde or the like, washed with phosphate buffered saline (PBS), and then stained with X-ga1 at room temperature or at room temperature.
  • PBS phosphate buffered saline
  • the / 3_-galactosidase reaction can be stopped by washing the tissue specimen with 1 mM ED TAZ PBS solution, and the coloration can be observed.
  • mRNA encoding lacZ may be detected according to a conventional method.
  • the compound or a salt thereof obtained by using the above-mentioned screening method is a compound selected from the above-mentioned test compounds, and is a compound that promotes or inhibits the promoter activity for DNA of the present invention.
  • the compound obtained by the screening method may form a salt.
  • the salt of the compound include physiologically acceptable acids (eg, inorganic acids, organic acids) and bases.
  • salts include, for example, salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.) or organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, Salts such as maleic acid, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc. are used.
  • inorganic acids eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.
  • organic acids eg, acetic acid, formic acid, propionic acid, fumaric acid
  • Salts such as maleic acid, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc. are used.
  • a compound or a salt thereof that promotes or inhibits (preferably inhibits) the promoter activity of the DNA of the present invention can regulate the expression of the protein of the present invention and regulate the function of the protein.
  • Kidney disease e.g., diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome, mesangial proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy, membranous Proliferative nephritis, Crescentic nephritis, lupus nephritis, amyloid kidney, tubular interstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, renal tumors, chronic rejection after kidney transplantation, etc.
  • Useful as a prophylactic 'therapeutic Useful as a
  • a drug containing the compound or a salt thereof obtained by the screening method can be produced in the same manner as the above-mentioned drug containing the protein of the present invention or a salt thereof.
  • the preparations obtained in this way are safe and low toxic, for example, in humans or mammals (e.g., rats, mice, guinea pigs, egrets, sheep, sheep, horses, horses, cats, cats, cats). And monkeys and monkeys).
  • the dose of the compound or a salt thereof varies depending on the target disease, the subject of administration, the route of administration, and the like.For example, when the compound of the present invention that inhibits the promoter activity against DNA is orally administered, it is generally used. In an adult (assuming a body weight of 60 kg) diabetic nephropathy, about 0.1 to 100 mg, preferably about 1.0 to 50 nig, more preferably about 1.0 to 20 mg of the compound is administered per day.
  • the single dose of the compound varies depending on the administration subject, target disease, and the like.
  • the compound of the present invention that inhibits the promoter activity against DNA is usually in the form of an injection.
  • an adult (as 60 kg) diabetic nephropathy patient about 0.01 to 30 mg, preferably about 0.1 to 20 mg, more preferably about 0.1 to 1 Onig of the compound per day is administered intravenously. It is convenient to administer by injection. In the case of other animals, the dose can be administered in terms of 60 kg.
  • the non-human mammal deficient in DNA expression of the present invention is extremely useful for screening a compound or a salt thereof that promotes or inhibits the activity of the promoter of DNA of the present invention
  • the present invention can greatly contribute to the investigation of the cause of various diseases caused by DNA expression deficiency or the development of preventive and therapeutic agents.
  • genes encoding various proteins are ligated downstream thereof and injected into egg cells of an animal to produce a so-called transgenic animal (transgenic animal). )
  • transgenic animal transgenic animal
  • a low molecule having an action of specifically promoting or suppressing the production ability of the protein of the present invention itself in the body can be used as a search system for compounds.
  • bases, amino acids, and the like are indicated by abbreviations based on the abbreviations by the IUPAC-IUB Commission on Biochemical Nomenclature or commonly used abbreviations in the relevant field, and examples thereof are described below.
  • amino acids can have optical isomers, the L-form is indicated unless otherwise specified.
  • DNA Deoxylipo nucleic acid
  • RNA ribonucleic acid
  • mRNA messenger ribonucleic acid
  • a 1 a Alanine
  • Trt Trityl
  • HONB Tohydroxy-5-norporene-2,3-dicarboximide DC C
  • sequence numbers in the sequence listing in the present specification show the following sequences.
  • the amino acid rooster sequence (GenBank Accession No. AAC98695) of human Type lib Na / Pi Cotransporter is shown.
  • Example 3 shows the nucleotide sequence of a primer used in Example 2.
  • Example 3 shows the nucleotide sequence of a primer used in Example 2.
  • Example 3 shows the nucleotide sequence of a probe used in Example 2.
  • Example 3 shows the nucleotide sequence of a primer used in Example 3.
  • Example 3 shows the nucleotide sequence of a primer used in Example 3.
  • Example 3 shows the nucleotide sequence of a probe used in Example 3.
  • the present invention will be more specifically described with reference to examples, but the present invention is not limited thereto.
  • the disease model animals are Wistar Fatty rat (WF Rad), Zucker Fatty rat (ZF rat), and spontaneously hypercholesterolemic rat (SHC rat).
  • Disease model animals and control animals are bred under the same conditions. Rats and WL rats are 13 weeks, 20 weeks, 40 weeks and 68 weeks old. At week 5, kidneys were collected from a minimum of 5 individuals in each group at 8 and 27 weeks of age for ZF and ZL rats and at 6, 12, 20, and 30 weeks of age for SHC and SD rats.
  • Total RNA was prepared for each individual according to the method described in the instruction manual of IS0GEN (Nitsubon Gene). Among the prepared samples, in WF rats and WL rats, samples from the WF rats at the age of 40 weeks, at which renal damage was progressing in the WF rats, and 68 weeks after birth, at which kidney function was further advanced and renal function was reduced, were compared with the ZF rat. For ZL rats and ZL rats, a 27-week-old sample with renal impairment in ZF rats was developed, and for SHC and SD rats, a 30-week-old sample with decreased renal function in SHC rats was used for the following comprehensive genes: It was used for expression analysis experiments. Table 1 shows the experimental configuration.
  • rat type lib Na / Pi Cotransporter was used by TaqMan PCR System.
  • Example 1 (Genbank Accession No. AF157026) in each individual kidney was measured. Using the total kidney RNA of each individual used in Example 1 as type II RM, the expression fluctuation was analyzed by comparing the expression levels of the experimental group and the control group.
  • the experimental groups were WF rats, ZF rats and SHC rats, and the corresponding control groups were WL rats, ZL rats and SD rats.
  • RNA for each individual prepared in Example 1 was prepared in Example 1 as a type III using a TaqMan Gold RT-PCR Kit (Applied Biosystems) to prepare cDNA.
  • the composition of the reaction solution Individual kidney Total RNA lng prepared in Example 1, 5.5mM MgCl 2, 0.5mM dNTPs , 2.5 zM Ramdoni Hexamers, 0.4U / 1 RNase Inhibitor,
  • the composition of the reaction solution was cDNA 5 ⁇ (corresponding to 2.4 ng of Total RNA) prepared by the above method, synthetic DNA primer — (SEQ ID NO: 7) 0.2 M, synthetic DNA primer (SEQ ID NO: 4), synthetic DNA probe (SEQ ID NO: 4). : 5) 0.2 M and TaqMan Universal PCR Master Mix
  • the relative expression level is calculated as the number of copies of the Type lib Na / Pi Cotransporter gene per unit volume. The value was divided by one copy of the GAPDH gene and multiplied by 100.
  • TCV116 (generic name: Candesartan cilexetil) (Kidney international 49, SI 15-SI 18, 1996, CI inical Experimental Nephrology 4, 207-214, So-called 0 year, Japan Journal of Pharmacology, 88, 300-306, 2002) was administered daily at 10 mg / kg body weight daily for 18 weeks to 9 weeks after birth.
  • the non-administration group received only the same amount of vehicle (0.5% methylcellulose lOOcP solution) as the administration group.
  • Kidneys were collected at 27 weeks of age from the three groups of ZF rats, vehicle-administered ZF rats, vehicle-administered ZF rats, and vehicle-administered ZL rats. Total RNA was prepared for each individual, and the relative expression was analyzed in the same manner as above.
  • the expression level of the human Type lib Na / Pi Cotransporter gene was measured by TaqMan PCR using the cDNA prepared by the above method as type III.
  • the composition of the reaction solution was as follows: various normal human cell-derived cDNA solutions 21 (equivalent to 4 ng of total RNA), synthetic DNA primers (SEQ ID NO: 10) 0.4 M, synthetic DNA primers (SEQ ID NO: 11) 0.4 ⁇ 0.2 l of the synthetic DNA probe (SEQ ID NO: 12) and 51 of TaqMan Universal PCR Master Mix (Applied Biosystems) were added to make the total volume 10 1.
  • the reaction was carried out using a 7900HT Sequence Detection System (Applied Biosystems) at 50 ° C for 2 minutes and at 95 ° C for 10 minutes, followed by a cycle of 95 ° C for 15 seconds and 60 ° C for 1 minute. Repeated times.
  • the expression value of dariceraldehyde triphosphate dehydrogenase (GAPDH) was measured for each sample, and the expression level of the human Type lib Na / Pi Cotransporter gene was measured. It was calculated as the relative expression level to the human GAPDH expression level.
  • the relative expression level was expressed as 100 times the value obtained by dividing the number of copies of the human Type lib Na / Pi Cotransporter gene per unit liquid volume by the number of copies of the human GAPDH gene per unit liquid volume.
  • TaqMan GAPDH Control Reagents (Applied Biosystems) was used to measure the expression value of human GAPDH.
  • measurement was performed twice for each sample, and the average value was used as the expression value.
  • Fig. 5 shows the results.
  • kidney proximal tubule cells Among the 24 types of normal human cells, kidney proximal tubule cells and human type lib
  • the proteins and polynucleotides of the present invention include, for example, renal diseases (eg, diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome group, mesangial proliferative nephritis, endoproliferative nephritis) , IgA nephropathy, membranous nephropathy, membranous proliferative nephritis, crescentic nephritis, Rubus nephritis, amyloid kidney, tubulointerstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis And a renal edema, a renal tumor, a chronic rejection after kidney transplantation, etc.), and a modulator obtained by screening using the protein, the polynucleotide, or an antibody against the protein.
  • renal diseases eg, diabetic nephropathy,
  • an inhibitor is less toxic, for example, a renal disease (eg, diabetic nephropathy, Glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome, mesangial proliferative nephritis, intraluminal proliferative nephritis, IgA nephropathy, membranous nephropathy, membranous proliferative nephritis, crescent-forming nephritis , Lupus nephritis, amyloid kidney, tubulointerstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, kidney tumor, chronic rejection after kidney transplantation, etc.
  • a renal disease eg, diabetic nephropathy, Glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome, mesangial proliferative nephritis
  • the polynucleotide of the present invention, the antisense polynucleotide of the present invention, and the like can be used, for example, in renal diseases (eg, diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, microdegenerative nephrotic syndrome, mesangium) Proliferative nephritis, intraluminal proliferative nephritis, IgA nephropathy, membranous nephropathy, membranous proliferative nephritis, crescentic nephritis, lupus nephritis, amyloid kidney, tubular interstitial nephritis, acute tubular necrosis, acute kidney It is useful for diagnosis, prevention or treatment of insufficiency, renal sclerosis, renal edema, renal tumors, chronic rejection after renal transplantation, etc.).
  • renal diseases eg, diabetic nephro

Abstract

As preventives/remedies for kidney diseases such as diabetic nephropathy, use can be made of a compound controlling (preferably inhibiting) the activity of a protein having an amino acid sequence, which is the same or substantially the same as the amino acid sequence represented by SEQ ID NO:1, or the expression of a gene of this protein; an antisense polynucleotide having a base sequence, which is complementary or substantially complementary with the base sequence of DNA encoding the above protein or its peptide fragment, or a part thereof; an antibody against the above protein or its peptide fragment, and so on.

Description

明 細 書 腎疾患の予防 ·治療剤 技術分野  Description Prevention and treatment of renal disease Technical field
本発明は、 糖尿病性腎症などの腎疾患の予防 ·治療剤、 診断薬、 および糖尿 病性腎症などの腎疾患の予防 ·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 a preventive / therapeutic agent for renal diseases such as diabetic nephropathy, a diagnostic agent, and a screening of a preventive / therapeutic agent for renal diseases such as diabetic nephropathy. Background art
· 腎は、 排泄 ·再吸収を通して体液成分の濃度を調節することで生体の恒常性 を保つとともに、 活性型ビタミン!)、 エリトロポェチン、 レニンなどの生理活性 物質を産生 ·分泌する内分泌器官としての機能も持つ極めて重要な組織である 。 このような重要な役割を担う腎の機能が障害されると、 種々の病態が引き起 こされ、 多種多様の腎疾患が発生する。 これら腎疾患の中には、 糖尿病、 肥満 および脂質代謝異常と密接に関連した腎症も含まれ、 特に、 糖尿病を原疾患と する糖尿病性腎症は、 腎症診断後は糖尿病のコントロールを厳密に行っても進 展阻止が困難で、 しかも腎不全に陥るケースが多く、 また患者数も少なくない 。 また、 腎疾患のモデル動物として、 Wis tar Fat ty ラット (WFラット) は、 腎 機能低下症状に加え糖尿病に特徴的は症状を示すことから、 糖尿病性腎症モデ レとして (Front iers in diabetes research, lessons from animal diabetes I I, 535-541 , 1988年) 、 Zucker Fat tyラット (ZFラッ卜) は、 WFラッ卜と同様 に腎機能低下と糖尿病の症状を合わせて呈することから、 糖尿病性腎症モデル として (Kidney internat ional, 52巻, S218 - S220頁, 1997年) 、 自然発症高コ レステロール血症ラット (SHCラット) は、 ヒト巣状糸球体硬化症と類似の症状 を示すことから、 巣状糸球体硬化症モデルとして (日本腎臓学界誌, 37巻, 91- 99頁, 1995年) 報告されている。 · The kidneys excrete · Maintain the body's homeostasis by regulating the concentration of body fluid components through reabsorption, and activate vitamins! ), Is an extremely important tissue that also functions as an endocrine organ that produces and secretes physiologically active substances such as erythropoietin and renin. Impaired renal function, which plays such an important role, can lead to a variety of conditions and a wide variety of renal diseases. These kidney diseases include nephropathy closely related to diabetes, obesity, and abnormal lipid metabolism.In particular, diabetic nephropathy, which is a primary disease of diabetes, requires strict control of diabetes after diagnosis of nephropathy. However, it is difficult to prevent progress, and in many cases renal failure occurs, and the number of patients is not small. As a model animal for renal disease, Wistar Fatty rats (WF rats) show symptoms characteristic of diabetes in addition to renal dysfunction, so they are used as a diabetic nephropathy model (Front iers in diabetes research). , lessons from animal diabetes II, 535-541, 1988), and Zucker Fatty rats (ZF rats) show diabetic nephropathy because they exhibit renal dysfunction and diabetes as well as WF rats. As a model (Kidney international, 52, S218-S220, 1997), spontaneously hypercholesterolemia rats (SHC rats) show symptoms similar to those of human focal glomerulosclerosis. It has been reported as a model of glomerulosclerosis (Japanese Journal of Nephrology, Vol. 37, pp. 91-99, 1995).
一方、 ヒト Type l ib Na/Pi cotranspor ter (リン酸ナトリウム共輸送体タイ プ l ib) は、 ナトリウムに依存したリン酸の輸送活性を有するトランスポ一タフ アミリーに属する分子で、 小腸で発現が認められることと、 低 pHで高活性を示 す点がゥサギまたはマウス小腸を用いたリン酸取り込みアツセィの結果に一致 することから、 小腸でのリン取り込みを担う分子である可能性が報告されてい る (Biochemi cal and Bi ophys i cal Research Communi cat i ons, 258巻, 578-582 頁, 1999年) 。 また、 ヒト Type l ib Na/P i cot ransporterの機能に関連する疾 患の状態、 例えば低リン酸血症および高リン酸血症などの血清リン酸塩濃度の 異常を来たす状態の治療法などが、 特開 2000- 245488号公報に開示されている。 USP 6, 319, 688号公報には、 ヒト Type l ib Na/P i co transpor terのアン夕ゴニス トおよびァゴニストが、 慢性腎不全、 最終病期の腎疾患、 尿毒性性骨疾患、 癌 の予防治 ¾に有用であると記載されている。 On the other hand, human type lib Na / Pi cotransporter (sodium phosphate cotransporter type lib) is a molecule belonging to transpomf amyl, which has sodium-dependent phosphate transport activity, and is expressed in the small intestine. And high activity at low pH This point is consistent with the results of the phosphate uptake assay using the rabbit and mouse small intestine, suggesting that it may be a molecule responsible for phosphorus uptake in the small intestine (Biochemical and Biophysical Research Communi cat ions, 258, 578-582, 1999). In addition, treatment of diseases related to the function of human Type Lib Na / Picot ransporter, such as treatment of abnormal conditions of serum phosphate concentration such as hypophosphatemia and hyperphosphatemia, etc. Are disclosed in Japanese Patent Application Laid-Open No. 2000-245488. U.S. Pat.No. 6,319,688 states that angiogonists and agonists of human Type I ib Na / Picotransporter are useful in treating chronic renal failure, end-stage renal disease, uremic bone disease, and cancer. It is described as useful for preventive care.
上述の原疾患に起因する腎病変に関しては、 発症機序と病態生理の解明を目 的として細胞生物学 ·分子生物学の手法を取り入れた研究が精力的に行われ、 新しい知見も得られつつあるが、 現時点では依然として未解明の課題が山積し ている状態である。 安全で優れた腎疾患の予防《治療剤が求められている。 発明の開示  Regarding renal lesions caused by the above-mentioned underlying diseases, studies that incorporate techniques of cell biology and molecular biology are being vigorously conducted with the aim of elucidating the pathogenesis and pathophysiology, and new knowledge is being obtained. However, there are still many unsolved issues at this time. There is a need for safe and excellent prevention of renal diseases << therapeutic agents. 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、 WFラッ ト、 ZFラット、 SHCラットなどのモデル動物を解析し、 発現が顕著に増加する遺 伝子を見出し、 この知見に基づいて、 さらに検討を重ねた結果、 本発明を完成 するに至った。  The present inventors have conducted intensive studies to solve the above problems, and as a result, analyzed model animals such as WF rat, ZF rat, SHC rat, and found a gene whose expression is significantly increased, As a result of further studies based on this finding, the present invention has been completed.
すなわち、 本発明は、  That is, the present invention
( 1 ) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の アミノ酸配列を含有するタンパク質もしくはその部分ペプチドまたはその塩の 活性を阻害する化合物またはその塩を含有してなる腎疾患の予防 ·治療剤、 (1) A renal disease comprising a compound which inhibits the activity of a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof, or a salt thereof, or a salt thereof Prevention and treatment of
( 1 a ) 腎疾患が、 糖尿病性腎症または巣状糸球体硬化症である上記 (1 ) 記載の予防 ·治療剤、 (1a) the prophylactic or therapeutic agent according to (1), wherein the renal disease is diabetic nephropathy or focal glomerulosclerosis;
( 2 ) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の アミノ酸配列を含有するタンパク質もしくはその部分ペプチドまたはその塩の 遺伝子の発現を阻害する化合物またはその塩を含有してなる腎疾患の予防 ·治 療剤、 (2 a) 腎疾患が、 糖尿病性腎症または巣状糸球体硬化症である上記 (2) 記載の予防 ·治療剤、 (2) a compound containing a protein or a partial peptide thereof having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a compound thereof or a salt thereof which inhibits gene expression Prevention and treatment of kidney disease, (2a) the prophylactic or therapeutic agent according to (2) above, wherein the renal disease is diabetic nephropathy or focal glomerulosclerosis;
(3) 腎疾患が、 糖尿病性腎症または巣状糸球体硬化症である上記 (1) ま たは (2) 記載の予防 ·治療剤、  (3) The prophylactic and / or therapeutic agent according to (1) or (2) above, wherein the renal disease is diabetic nephropathy or focal glomerulosclerosis.
(4) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の アミノ酸配列を含有するタンパク質またはその部分ペプチドをコードするポリ ヌクレオチドの塩基配列に相補的もしくは実質的に相補的な塩基配列またはそ の一部分を含有するァンチセンスポリヌクレオチド、  (4) a base complementary or substantially complementary to the base sequence of a polynucleotide encoding a protein or a partial peptide thereof having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 An antisense polynucleotide containing the sequence or a portion thereof,
(5) 上記 (4) 記載のアンチセンスポリヌクレオチドを含有してなる医 薬、  (5) a medicine comprising the antisense polynucleotide according to (4),
(6) 腎疾患の予防 ·治療剤である上記 (5) 記載の医薬、  (6) the medicament according to the above (5), which is an agent for preventing or treating renal disease;
(6 a) 腎疾患が、 糖尿病性腎症または巣状糸球体硬化症である上記 (6) 記載の予防 ·治療剤、  (6a) the prophylactic or therapeutic agent according to (6) above, wherein the renal disease is diabetic nephropathy or focal glomerulosclerosis;
(7) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の アミノ酸配列を含有するタンパク質またはその部分ペプチドまたはその塩に対 する抗体、  (7) an antibody against a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1, a partial peptide thereof, or a salt thereof;
(8) 上記 (7) 記載の抗体を含有してなる医薬、  (8) a pharmaceutical comprising the antibody according to (7) above,
(9) 腎疾患の予防 ·治療剤である上記 (8) 記載の医薬、  (9) The medicament according to the above (8), which is an agent for preventing or treating kidney disease.
(9 a) 腎疾患が、 糖尿病性腎症または巣状糸球体硬化症である上記 (9) 記載の予防 ·治療剤、  (9a) The preventive / therapeutic agent according to (9) above, wherein the renal disease is diabetic nephropathy or focal glomerulosclerosis.
(10) 上記 (7) 記載の抗体を含有してなる診断薬、  (10) a diagnostic agent comprising the antibody according to (7),
(1 1) 腎疾患の診断薬である上記 (10) 記載の診断薬、  (11) The diagnostic agent according to the above (10), which is a diagnostic agent for renal disease,
(12) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一 のァミノ酸配列を含有するタンパク質またはその部分べプチドをコードするポ リヌクレオチドを含有してなる腎疾患の診断薬、  (12) an agent for diagnosing renal disease comprising a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a polynucleotide encoding a partial peptide thereof,
(13) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一 のアミノ酸配列を含有するタンパク質もしくはその部分ペプチドまたはその塩 を用いることを特徴とする腎疾患の予防 ·治療剤のスクリーニング方法、 (13) Screening of a prophylactic and therapeutic agent for renal disease, characterized by using a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof. Method,
(13 a) (i) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的 に同一のァミノ酸配列を含有するタンパク質もしくはその部分べプチドまたは その塩を発現しうる細胞に、 標識された無機リン酸塩を加えた場合と、 (ii) 試験化合物の存在下、 上記細胞に、 標識された無機リン酸塩を加えた場合の、 細胞内に取り込まれた無機リン酸量をそれぞれ測定する上記 (13) 記載のス クリーニング方法、 (13a) (i) identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 (A) adding a labeled inorganic phosphate to a cell capable of expressing a protein containing the same amino acid sequence or its partial peptide or a salt thereof, and (ii) applying the labeled amino phosphate to the cell in the presence of a test compound. The screening method according to the above (13), wherein the amount of the inorganic phosphate taken into the cells when the labeled inorganic phosphate is added is measured,
(13 b) 標識された無機リン酸塩が、 放射性同位元素 ( 〔32P〕 、 〔33P〕 な ど) で標識された無機リン酸塩である上記 (13 a) 記載のスクリーニング方 法、 (13 b) labeled inorganic phosphate, radioisotope ([32 P], [33 P], etc.) screening how the (13 a), wherein the labeled inorganic phosphate in,
(13 c) (i) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的 に同一のアミノ酸配列を含有するタンパク質もしくはその部分ペプチドまたは その塩を含有する合成脂質二重層に、 無機リン酸塩を含む緩衝液を接触させた 場合と、 (ii) 試験化合物の存在下、 上記合成脂質二重層に、 無機リン酸塩を 含む緩衝液を接触させた場合の、 無機リン酸輸送活性をそれぞれ測定する上記 (13c) (i) Inorganic phosphate is added to a synthetic lipid bilayer containing a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof. Inorganic phosphate transport activity was measured when the buffer containing salt was brought into contact with the buffer containing inorganic phosphate, and (ii) when the buffer containing inorganic phosphate was brought into contact with the synthetic lipid bilayer in the presence of the test compound. Above to measure
(13) 記載のスクリーニング方法、 (13) the screening method described,
(13 d) 腎疾患が、 糖尿病性腎症または巣状糸球体硬化症である上記 (1 3) 記載のスクリーニング方法、  (13d) the screening method according to (13), wherein the kidney disease is diabetic nephropathy or focal glomerulosclerosis;
(13 e) 上記 (13) 記載のスクリーニング方法を用いて得られる、 配列 番号: 1で表されるァミノ酸配列と同一もしくは実質的に同一のァミノ酸配列 を含有するタンパク質もしくはその部分ペプチドまたはその塩の活性を阻害す る化合物またはその塩を含有してなる腎疾患の予防 ·治療剤、  (13e) a protein or a partial peptide thereof containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 obtained by using the screening method described in the above (13). A prophylactic / therapeutic agent for kidney disease comprising a compound that inhibits the activity of a salt or a salt thereof,
(14) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一 のアミノ酸配列を含有するタンパク質もしくはその部分ペプチドまたはその塩 を含有することを特徴とする腎疾患の予防 ·治療剤のスクリーニング用キッ 卜、  (14) A prophylactic and / or therapeutic agent for renal disease characterized by containing a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof. Screening kit,
(15) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一 のアミノ酸配列を含有するタンパク質またはその部分ペプチドをコードするポ リヌクレオチドを用いることを特徴とする腎疾患の予防 ·治療剤のスクリ一二 ング方法、  (15) Prevention and treatment of renal disease, characterized by using a polynucleotide encoding a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof. How to screen the agent,
(15 a) 腎疾患が、 糖尿病性腎症または巣状糸球体硬化症である上記 (1 5) 記載のスクリーニング方法、 (15a) The renal disease is diabetic nephropathy or focal glomerulosclerosis (1) 5) the screening method described,
(16) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一 のアミノ酸配列を含有するタンパク質またはその部分ペプチドをコードするポ リヌクレオチドを含有することを特徴とする腎疾患の予防 ·治療剤のスクリ一 ニング用キッ卜、  (16) Prevention of renal disease characterized by containing a polynucleotide encoding a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof, Kit for screening therapeutic agents,
(17) 腎疾患が、 糖尿病性腎症または巣状糸球体硬化症である上記 (1 3) または (15) 記載のスクリーニング方法、  (17) The screening method according to the above (13) or (15), wherein the renal disease is diabetic nephropathy or focal glomerulosclerosis,
(17 a) 上記 (13) もしくは (15) 記載のスクリーニング方法または 上記 (14) もしくは (16) 記載のスクリーニング用キットを用いて得られ る腎疾患の予防 ·治療作用を有する化合物またはその塩、  (17a) A compound having a prophylactic or therapeutic effect on renal disease or a salt thereof, which is obtained by using the screening method according to (13) or (15) or the screening kit according to (14) or (16).
(17 b) 上記 (17 a) 記載の化合物またはその塩を含有してなる腎疾患 の予防 ·治療剤、  (17b) a prophylactic / therapeutic agent for renal disease comprising the compound or salt thereof according to (17a) above,
(18) 哺乳動物に対して、 配列番号: 1で表されるアミノ酸配列と同一も しくは実質的に同一のァミノ酸配列を含有するタンパク質もしくはその部分べ プチドまたはその塩の活性を阻害する化合物もしくはその塩、 または該タンパ ク質もしくはその部分ペプチドまたはその塩の遣伝子の発現を阻害する化合物 もしくはその塩の有効量を投与することを特徴とする、 腎疾患の予防 ·治療 法、  (18) a compound that inhibits the activity of a protein, or a partial peptide or a salt thereof, containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 in mammals Or an effective amount of a compound or a salt thereof, or a compound or a salt thereof that inhibits the expression of the gene of the protein or its partial peptide or a salt thereof, or a method for preventing or treating renal disease,
(18 a) 腎疾患が、 糖尿病性腎症または巣状糸球体硬化症である上記 (1 8) 記載の予防 ·治療法、  (18a) The method of (18), wherein the renal disease is diabetic nephropathy or focal glomerulosclerosis,
(19) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一 のァミノ酸配列を含有するタンパク質もしくはその部分ペプチドまたはその塩 の活性を阻害する、 または該夕ンパク質もしくはその部分べプチドまたはその 塩の遺伝子の発現を阻害することを特徴とする、 腎疾患の予防 ·治療法、 (19 a) 腎疾患が、 糖尿病性腎症または巣状糸球体硬化症である上記 (1 9) 記載の予防,治療法、  (19) inhibits the activity of a protein or a partial peptide thereof or a salt thereof containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1, or inhibits the activity of the protein or a partial thereof; Prevention and treatment of renal disease, characterized by inhibiting the expression of the peptide or its salt gene. (19a) The renal disease is diabetic nephropathy or focal glomerulosclerosis (19) ) Described prevention, treatment,
(20) 腎疾患の予防 ·治療剤を製造するための配列番号: 1で表されるァ ミノ酸配列と同一もしくは実質的に同一のアミノ酸配列を含有するタンパク質 もしくはその部分べプチドまたはその塩の活性を阻害する化合物もしくはその 塩、 または該タンパク質もしくはその部分ペプチドまたはその塩の遺伝子の発 現を阻害する化合物もしくはその塩の使用、 (20) Prevention of renal disease · A protein containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof for producing a therapeutic agent A compound that inhibits the activity or its Use of a salt, or a compound or a salt thereof that inhibits expression of the gene for the protein or its partial peptide or a salt thereof;
( 2 0 a ) 腎疾患が、 糖尿病性腎症または巣状糸球体硬化症である上記 (2 0 ) 記載の使用などに関する。 図面の簡単な説明  (20a) The use according to (20), wherein the renal disease is diabetic nephropathy or focal glomerulosclerosis. BRIEF DESCRIPTION OF THE FIGURES
図 1は、 ラット Type l ib Na/Pi Cotransporter発現量の経時変化を表す図で ある。 図中、 縦軸はラット Type l ib Na/Pi Cotransporter遺伝子発現値のラッ ト GAPDH発現値に対する相対的発現量を、 横軸は腎臓を採取した週齢を示す。 鱷は実験群 (WFラット) 、 口は対照群 (WLラット) の結果である。  FIG. 1 is a graph showing the time course of the expression level of rat Typelib Na / Pi Cotransporter. In the figure, the vertical axis represents the relative expression level of rat Typelib Na / Pi Cotransporter gene expression value to rat GAPDH expression value, and the horizontal axis represents the age at which kidneys were collected.鱷 indicates the results of the experimental group (WF rats), and the mouth indicates the results of the control group (WL rats).
図 2は、 ラット Type l ib Na/Pi Cotransporter発現量の経時変化を表す図で ある。 図中、 縦軸はラット Type l ib Na/P i Co transpor ter遣伝子発現値のラッ ト GAPDH発現値に対する相対的発現量を、 横軸は腎臓を採取した週齢を示す。 ■は実験群 (ZFラット) 、 口は対照群 (ZLラット) の結果である。  FIG. 2 is a graph showing the time course of the expression level of rat Typelib Na / Pi Cotransporter. In the figure, the vertical axis indicates the relative expression level of the rat Type Lib Na / Pi Co transporter gene expression value to the rat GAPDH expression value, and the horizontal axis indicates the age at which the kidney was collected. ■ shows the results of the experimental group (ZF rats) and the mouth shows the results of the control group (ZL rats).
図 3は、 ラット Type l ib Na/Pi Cotranspor ter発現量の経時変化を表す図で ある。 図中、 縦軸はラッ卜 Type l ib Na/Pi Cotransporter遺伝子発現値のラッ ト GAPDH発現値に対する相対的発現量を、 横軸は腎臓を採取した週齢を示す。 國は実験群 (SHCラット) 、 口は対照群 (SDラット) の結果である。  FIG. 3 is a diagram showing a time-dependent change in the expression level of rat Typelib Na / Pi Cotransporter. In the figure, the vertical axis indicates the relative expression level of the rat Type Lib Na / Pi Cotransporter gene expression value to the rat GAPDH expression value, and the horizontal axis indicates the age at which the kidney was collected. The results are for the experimental group (SHC rats) and the control group (SD rats) for the mouth.
図 4は、 TCV116のラット Type l ib Na/Pi Cotransporter発現量に対する影響 を表す図である。 図中、 縦軸はラット Type l ib Na/Pi Cotransporter遺伝子発 現値のラット GAPDH発現値に対する相対的発現量を示す。 横軸は実験に用いたラ ッ卜の種類と TCV116投与の有無を示す。 ZF (Vehic le) は賦形剤を投与した ZFラ ット、 ZF (TCV116) は TCV116を投与した ZFラット、 ZL (Vehicle) は賦形剤を投 与した ZLラットの結果を示す。  FIG. 4 is a graph showing the effect of TCV116 on rat Typelib Na / Pi Cotransporter expression levels. In the figure, the vertical axis indicates the relative expression level of the rat Typelib Na / Pi Cotransporter gene expression value to the rat GAPDH expression value. The horizontal axis shows the type of rat used in the experiment and the presence or absence of TCV116 administration. ZF (Vehicle) shows the results of ZF rats to which vehicle was administered, ZF (TCV116) shows the results of ZF rats to which TCV116 was administered, and ZL (Vehicle) shows the results of ZL rats to which vehicle was administered.
図 5は、 ヒト Type l ib Na/P i Cotransporterの各種正常ヒト細胞での発現量 を表す図である。 図中、 縦軸はヒト Type l ib Na/Pi Cotransporter遺伝子発現 値の GAPDH発現値に対する相対的発現量を示す。 横軸のレーン 1は臍帯静脈内皮 細胞、 レーン 2は大動脈血管内皮細胞、 レーン 3は冠状動脈内皮細胞、 レーン 4は 皮膚微小血管内皮細胞、 レーン 5は肺微小血管内皮細胞、 レーン 6は肺動脈血管 内皮細胞、 レーン 7は大動脈平滑筋細胞、 レーン 8は冠状動脈平潸筋細胞、 レー ン 9は子宮平滑筋細胞、 レーン 10は気管支平滑筋細胞、 レーン 11は骨格筋細胞、 レーン 12は乳腺上皮細胞、 レーン 13は気管支上皮細胞、 レーン 14は細気管支上 皮細胞、 レーン 15は肺繊維芽細胞、 レーン 16は腎臓近位尿細管細胞、 レーン 17 はメザンギゥム細胞、 レーン 18は腎臓皮質細胞、 レーン 19は腎上皮細胞、 レー ン 20は間葉性幹細胞、 レーン 21は軟骨細胞、 レーン 22は骨芽細胞、 レーン 23は 前立腺間質細胞、 レーン 24は表皮角化細胞をそれぞれ示す。 発明を実施するための最良の形態 FIG. 5 is a diagram showing the expression levels of human Typelib Na / Pi Cotransporter in various normal human cells. In the figure, the vertical axis indicates the relative expression level of the human Typelib Na / Pi Cotransporter gene expression value to the GAPDH expression value. Lane 1 on the horizontal axis is umbilical vein endothelial cells, lane 2 is aortic endothelial cells, lane 3 is coronary endothelial cells, lane 4 is skin microvascular endothelial cells, lane 5 is lung microvascular endothelial cells, and lane 6 is pulmonary artery blood vessels. Endothelial cells, lane 7 aortic smooth muscle cells, lane 8 coronary artery flat muscle cells, lane 9 uterine smooth muscle cells, lane 10 bronchial smooth muscle cells, lane 11 skeletal muscle cells, lane 12 mammary epithelium Cells, lane 13 is bronchial epithelial cells, lane 14 is bronchiolar epithelial cells, lane 15 is lung fibroblasts, lane 16 is renal proximal tubule cells, lane 17 is mezzanine cells, lane 18 is renal cortical cells, lane 19 shows renal epithelial cells, lane 20 shows mesenchymal stem cells, lane 21 shows chondrocytes, lane 22 shows osteoblasts, lane 23 shows prostate stromal cells, and lane 24 shows epidermal keratinocytes. BEST MODE FOR CARRYING OUT THE INVENTION
配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァミノ 酸配列を含有するタンパク質 (以下、 本発明のタンパク質または本発明で用い られるタンパク質と称することもある) は、 ヒ卜や温血動物 (例、 モルモッ ト、 ラット、 マウス、 ニヮ卜リ.. ゥサギ、 ブタ、 ヒッジ、 ゥシ、 サルなど) の 細胞 (例、 肝細胞、 脾細胞、 神経細胞、 グリア細胞、 塍臓 /3細胞、 骨髄細胞、 メサンギゥム細胞、 ランゲルハンス細胞、 表皮細胞、 上皮細胞、 杯細胞、 内皮 細胞、 平滑筋細胞、 繊維芽細胞、 繊維細胞、 筋細胞、 脂肪細胞、 免疫細胞  A protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 (hereinafter, also referred to as the protein of the present invention or the protein used in the present invention) may be a human or a human. Cells of warm-blooded animals (eg, guinea pigs, rats, mice, chicks, etc., egrets, pigs, sheep, pigs, monkeys, etc.) (eg, hepatocytes, spleen cells, nerve cells, glial cells, kidney) / 3 cells, bone marrow cells, mesangial cells, Langerhans cells, epidermal cells, epithelial cells, goblet cells, endothelial cells, smooth muscle cells, fibroblasts, fiber cells, muscle cells, fat cells, immune cells
(例、 マクロファージ、 T細胞、 B細胞、 ナチュラルキラー細胞、 肥満細胞、 好中球、 好塩基球、 好酸球、 単球) 、 巨核球、 滑膜細胞、 軟骨細胞、 骨細胞、 骨芽細胞、 破骨細胞、 乳腺細胞、 肝細胞もしくは間質細胞、 またはこれら細胞 の前駆細胞、 幹細胞もしくはガン細胞など) もしくはそれらの細胞が存在する あらゆる組織、 例えば、 脳、 脳の各部位 (例、 嗅球、 扁桃核、 大脳基底球、 海 馬、 視床、 視床下部、 大脳皮質、 延髄、 小脳) 、 脊髄、 下垂体、 胃、 勝臓、 腎 臓、 肝臓、 生殖腺、 甲状腺、 胆のう、 骨髄、 副腎、 皮膚、 筋肉、 肺、 消化管 (例、 大腸、 小腸) 、 血管、 心臓、 胸腺、 脾臓、 顎下腺、 末梢血、 前立腺、 睾 丸、 卵巣、 胎盤、 子宮、 骨、 関節、 骨格筋などに由来するタンパク質であって もよく、 合成タンパク質であってもよい。  (Eg, macrophages, T cells, B cells, natural killer cells, mast cells, neutrophils, basophils, eosinophils, monocytes), megakaryocytes, synovial cells, chondrocytes, bone cells, osteoblasts , Osteoclasts, mammary gland cells, hepatocytes or stromal cells, or precursors of these cells, stem cells or cancer cells, or any tissue in which those cells are present, for example, the brain, various parts of the brain (eg, olfactory bulb) , Amygdala, basal ganglia, hippocampus, thalamus, hypothalamus, cerebral cortex, medulla oblongata, cerebellum), spinal cord, pituitary, stomach, victory, kidney, liver, gonad, thyroid, gall bladder, bone marrow, adrenal gland, skin , Muscle, lung, gastrointestinal tract (eg, large intestine, small intestine), blood vessels, heart, thymus, spleen, submandibular gland, peripheral blood, prostate, testicle, ovary, placenta, uterus, bone, joint, skeletal muscle, etc. The protein May it, may be a synthetic protein.
配列番号: 1で表されるアミノ酸配列と実質的に同一のアミノ酸配列として は、 配列番号: 1で表わされるアミノ酸配列と約 6 0 %以上、 好ましくは約 7 0 %以上、 好ましくは約 8 0 %以上、 好ましくは約 9 0 %以上、 好ましくは約 9 5 %以上の相同性を有するアミノ酸配列などが挙げられる。 The amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 is about 60% or more, preferably about 70% or more, preferably about 80% or more of the amino acid sequence represented by SEQ ID NO: 1. % Or more, preferably about 90% or more, preferably about Amino acid sequences having a homology of 95% or more.
配列番号: 1で表されるアミノ酸配列と実質的に同一のアミノ酸配列を含有 するタンパク質としては、 例えば、 前記の配列番号: 1で表されるアミノ酸配 列と実質的に同一のアミノ酸配列を含有し、 配列番号: 1で表されるアミノ酸 配列を含有するタンパク質と実質的に同質の活性を有するタンパク質などが好 ましい。 配列番号: 1で表されるアミノ酸配列と実質的に同一のアミノ酸配列 を含有するタンパク質としては、 例えば、 配列番号: 3で表されるアミノ酸配 列を含有するタンパク質、 配列番号: 5で表されるアミノ酸配列を含有するタ ンパク質などが挙げられる。  Examples of the protein having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 include, for example, a protein having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 described above. However, a protein having substantially the same activity as the protein containing the amino acid sequence represented by SEQ ID NO: 1 is preferable. Examples of the protein containing an amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 1 include, for example, a protein containing the amino acid sequence represented by SEQ ID NO: 3, and a protein represented by SEQ ID NO: 5 And a protein containing an amino acid sequence.
実質的に同質の活性としては、 例えば、 無機リン酸取り込み活性、 無機リン 酸輸送活性などが挙げられる。 実質的に同質とは、 それらの性質が性質的に (例、 生理学的に、 または薬理学的に) 同質であることを示す。 したがって、 無機リン酸取り込み活性、 無機リン酸輸送活性などが同等 (例、 約 0 . 0 1〜 1 0 0倍、 好ましくは約 0 . 1〜1 0倍、 より好ましくは 0 . 5〜2倍) であ ることが好ましいが、 これらの活性の程度、 タンパク質の分子量などの量的要 素は異なっていてもよい。  Examples of substantially the same activity include an inorganic phosphate uptake activity and an inorganic phosphate transport activity. Substantially identical indicates that the properties are qualitatively (eg, physiologically or pharmacologically) equivalent. Therefore, inorganic phosphate uptake activity, inorganic phosphate transport activity, etc. are equivalent (eg, about 0.01 to 100 times, preferably about 0.1 to 10 times, more preferably 0.5 to 2 times). ), But the quantitative factors such as the degree of these activities and the molecular weight of the protein may be different.
無機リン酸取り込み活性の測定は、 自体公知の方法、 例えば、 Me thods in Enzymol ogy, 191巻, 494-505頁, 1990年などに記載の方法またはそれに準じる 方法に従つて測定することができる。  Inorganic phosphate uptake activity can be measured according to a method known per se, for example, the method described in Methods in Enzymology, 191: 494-505, 1990 or a method analogous thereto.
具体的には、 本発明のタンパク質を発現する細胞に、 標識された無機リン酸 塩 (例、 放射性同位元素で標識された無機リン酸塩など) を加え、 一定時間保 温後、 細胞内の無機リン酸量を測定することにより行う。 反応は適当な緩衝液 中で行い、 細胞に取り込まれなかった無機リン酸は測定前に洗浄除去する。 標 識剤が放射性同位元素である場合、 取り込み活性は、 放射活性量を公知の方 法、 例えば放射活性測定装置などを使用して測定する。  Specifically, a labeled inorganic phosphate (eg, an inorganic phosphate labeled with a radioisotope) is added to cells expressing the protein of the present invention, and after incubation for a certain time, intracellular This is performed by measuring the amount of inorganic phosphoric acid. The reaction is performed in an appropriate buffer, and the inorganic phosphate that has not been taken up by cells is washed away before measurement. When the labeling agent is a radioisotope, the uptake activity is measured by a known method, for example, using a radioactivity measuring device.
無機リン酸輸送活性の測定は、 自体公知の方法、 例えば、 Bi ochimi ca et Bi ophys i ca Ac ta, 1236巻, 339- 344頁, 1995年などに記載の方法またはそれに 準じる方法に従って測定することができる。  The inorganic phosphate transport activity can be measured according to a method known per se, for example, the method described in Biochimi ca et Biophysica ca Acta, Vol. 1236, pp. 339-344, 1995, or a method analogous thereto. Can be.
具体的には、 本発明のタンパク質を含有する合成脂質二重層に、 無機リン酸 塩を含む緩衝液を添加し、 無機リン酸が本発明のタンパク質によって輸送され た際に生じる膜電流の変化を測定する。 膜電流の測定は、 公知の方法、 例えば 膜電位固定法などを使用して測定する。 本発明のタンパク質を含有する合成脂 質二重層は、 例えば、 本発明のタンパク質を発現させた細胞の膜画分から、 公 知の膜タンパク質精製方法に従って得られた精製タンパク質を用い、 リン脂質 などを材料にして、 公知の方法、 例えば 「パッチクランプ実験技術法」 (吉岡 書店、 1996年発行) 等に記載された方法に従って製造できる。 Specifically, inorganic phosphate is added to the synthetic lipid bilayer containing the protein of the present invention. A buffer containing a salt is added, and the change in membrane current that occurs when inorganic phosphate is transported by the protein of the present invention is measured. The membrane current is measured using a known method, for example, a membrane potential fixing method. The synthetic lipid bilayer containing the protein of the present invention can be obtained by, for example, using a purified protein obtained from a membrane fraction of cells expressing the protein of the present invention according to a known membrane protein purification method, using a phospholipid or the like. The material can be manufactured according to a known method, for example, a method described in "Patch Clamp Experimental Technique" (Yoshioka Shoten, published in 1996).
また、 本発明で用いられるタンパク質としては、 例えば、 (i) 配列番号: 1、'配列番号: 3または配列番号: 5で表されるアミノ酸配列中の 1または 2 個以上 (例えば 1〜1 0 0個程度、 好ましくは 1〜3 0個程度、 好ましくは 1 〜1 0個程度、 さらに好ましくは数 ( 1〜5 ) 個) のアミノ酸が欠失したアミ ノ酸配列、 (i i) 配列番号: 1、 配列番号: 3または配列番号: 5で表される アミノ酸配列に 1または 2個以上 (例えば 1〜1 0 0個程度、 好ましくは 1〜 3 0個程度、 好ましくは 1〜1 0個程度、 さらに好ましくは数 ( 1〜5 ) 個) のアミノ酸が付加したアミノ酸配列、 (i i i) 配列番号: 1、 配列番号: 3また は配列番号: 5で表されるァミノ酸配列に 1または 2個以上 (例えば 1〜1 0 0個程度、 好ましくは 1〜3 0個程度、 好ましくは 1〜1 0個程度、 さらに好 ましくは数 (1〜5 ) 個) のァミノ酸が挿入された 7ミノ酸配列、 (iv) 配列 番号: 1、 配列番号: 3または配列番号: 5で表されるアミノ酸配列中の 1ま たは 2個以上 (例えば 1〜1 0 0個程度、 好ましくは 1〜 3 0個程度、 好まし くは 1〜1 0個程度、 さらに好ましくは数 (1〜5 ) 個) のアミノ酸が他のァ ミノ酸で置換されたアミノ酸配列、 または (v) それらを組み合わせたアミノ酸 配列を含有するタンパク質などのいわゆるムティンも含まれる。  Examples of the protein used in the present invention include: (i) one or two or more (for example, 1 to 10) in the amino acid sequence represented by SEQ ID NO: 1, 'SEQ ID NO: 3 or SEQ ID NO: 5; An amino acid sequence in which about 0, preferably about 1 to 30, preferably about 1 to 10, and more preferably about (1 to 5) amino acids have been deleted; (ii) SEQ ID NO: 1, 1 or 2 or more amino acid sequences represented by SEQ ID NO: 3 or SEQ ID NO: 5 (for example, about 1 to 100, preferably about 1 to 30 and preferably about 1 to 10) (Iii) one or two amino acids in the amino acid sequence represented by SEQ ID NO: 1, SEQ ID NO: 3 or SEQ ID NO: 5, and more preferably (1 to 5) amino acids. Above (for example, about 1 to 100 pieces, preferably about 1 to 30 pieces, preferably about 1 to 10 pieces, Preferably, a 7-amino acid sequence into which a number (1 to 5) of amino acids have been inserted, (iv) one or more of the amino acid sequences represented by SEQ ID NO: 1, SEQ ID NO: 3 or SEQ ID NO: 5 Or two or more (for example, about 1 to 100, preferably about 1 to 30, preferably about 1 to 10, and more preferably about 1 to 5) amino acids. So-called mucins such as proteins containing an amino acid sequence substituted with an amino acid of (a) or (v) an amino acid sequence obtained by combining them are also included.
上記のようにアミノ酸配列が挿入、 欠失または置換されている場合、 その挿 入、 欠失または置換の位置としては、 とくに限定されない。  When the amino acid sequence is inserted, deleted or substituted as described above, the position of the insertion, deletion or substitution is not particularly limited.
本明細書におけるタンパク質は、 ペプチド標記の慣例に従って左端が N末端 (ァミノ末端) 、 右端が C末端 (力ルポキシル末端) である。 配列番号: 1で 表わされるアミノ酸配列を含有するタンパク質をはじめとする、 本発明で用い られるタンパク質は、 C末端が力ルポキシル基 (- C00H) 、 力ルポキシレ一ト (-COO—) 、 アミド (- C0NH2) またはエステル (- C00R) の何れであってもよ い。 In the protein in the present specification, the left end is the N-terminus (amino terminus) and the right end is the C-terminus (potassium terminus) according to the convention of peptide notation. The protein used in the present invention, including the protein containing the amino acid sequence represented by SEQ ID NO: 1, has a C-terminal lipoxyl group (-C00H), It may be any of (—COO—), amide (—C0NH 2 ) and ester (—C00R).
ここでエステルにおける Rとしては、 例えば、 メチル、 ェチル、 n—プロピ ル、 イソプロピル、 n—ブチルなどの C Mアルキル基、 例えば、 シクロペンチ ル、 シクロへキシルなどの C 3_8シクロアルキル基、 例えば、 フエニル、 α—ナ フチルなどの C 6_127リール基、 例えば、 ベンジル、 フエネチルなどのフエニル 一 C Mアルキル基もしくは 一ナフチルメチルなどの 一ナフチルー C Mアル キル基などの C 714ァラルキル基、 ピバロィルォキシメチル基などが用いられ る。 Here, as R in the ester, e.g., methyl, Echiru, n- propyl Le, isopropyl, CM alkyl group, such as n- butyl, cyclopentyl Le, C 3 _ 8 cycloalkyl group such as cyclohexyl, for example, phenyl, alpha-Na C 6 _ 12 7 aryl group such as Fuchiru, for example, benzyl, C 7, such as single Nafuchiru C M Al kill groups such as phenyl one CM alkyl group or single naphthylmethyl such phenethyl - 14 Ararukiru group, A pivaloyloxymethyl group or the like is used.
本発明で用いられるタンパク質が C末端以外に力ルポキシル基 (またはカル ポキシレー卜) を有している場合、 力ルポキシル基がアミド化またはエステル 化されているものも本発明で用いられるタンパク質に含まれる。 この場合のェ ステルとしては、 例えば上記した C末端のエステルなどが用いられる。  When the protein used in the present invention has a lipoxyl group (or carboxylate) other than the C-terminal, the protein in which the lipoxyl group is amidated or esterified is also included in the protein used in the present invention. . As the ester in this case, for example, the above-mentioned C-terminal ester or the like is used.
さらに、 本発明で用いられるタンパク質には、 N末端のアミノ酸残基 (例、 メチォニン残基) のァミノ基が保護基 (例えば、 ホルミル基、 ァセチル基など の c卜 6アル力ノィルなどの c卜 6ァシル基など) で保護されているもの、 生体内 で切断されて生成する N末端のグルタミン残基がピログルタミン酸化したも の、 分子内のァ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、 ァセチル基などの アルカノィル基などの C ァシル基など) で保護 されているもの、 あるいは糖鎖が結合したいわゆる糖タンパク質などの複合夕 ンパク質なども含まれる。 Furthermore, the protein used in the present invention, the amino acid residue (e.g., Mechionin residues) of N-terminal Amino group protecting groups (e.g., c Bok such c Bok 6 Al force Noiru such formyl group, Asechiru group 6- Glycyl group), N-terminal glutamine residue generated by cleavage in vivo, and pyroglutamine oxidation, substituent on the side chain of amino acid in the molecule (e.g.,- 0H, -SH, amino group, imidazole group, indole group, guanidino group, etc. are protected with appropriate protecting groups (for example, C-silyl groups such as alkanoyl groups such as formyl group and acetyl group). And complex proteins such as so-called glycoproteins to which sugar chains are bound.
本発明で用いられるタンパク質の具体例としては、 例えば、 配列番号: 1で 表されるアミノ酸配列を含有するタンパク質、 配列番号: 3で表されるァミノ 酸配列を含有するタンパク質、 配列番号: 5で表されるアミノ酸配列を含有す るタンパク質などがあげられる。  Specific examples of the protein used in the present invention include, for example, a protein containing the amino acid sequence represented by SEQ ID NO: 1, a protein containing the amino acid sequence represented by SEQ ID NO: 3, and a protein containing the amino acid sequence represented by SEQ ID NO: 5. Proteins containing the represented amino acid sequence are exemplified.
本発明で用いられるタンパク質の部分ペプチドとしては、 前記した本発明で 用いられるタンパク質の部分ペプチドであって、 好ましくは、 前記した本発明 で用いられるタンパク質と同様の性質を有するものであればいずれのものでも よい。 The partial peptide of the protein used in the present invention is the partial peptide of the protein used in the present invention described above, and is preferably any one having the same properties as the protein used in the present invention described above. Even things Good.
具体的には、 後述する本発明の抗体を調製する目的には、 配列番号: 1で表 されるアミノ酸配列において第 1〜1 0 1番目、 第 1 8 7〜 3 6 0番目、 第 5 7 5〜6 9 0番目のアミノ酸配列を有するペプチド、 配列番号: 3で表される アミノ酸配列において第 1〜1 0 1番目、 第 1 8 7〜 3 6 0番目、 第 5 7 5〜 6 9 5番目のアミノ酸配列を有するペプチド、 配列番号: 5で表されるァミノ 酸配列において第 1〜1 0 1番目、 第 1 8 7〜3 6 0番目、 第 5 7 5〜 6 9 7 番目のアミノ酸配列を有するペプチドなどがあげられる。 例えば、 本発明で用 いられるタンパク質の構成アミノ酸配列のうち少なくとも 2 0個以上、 好まし くは 5 0個以上、 さらに好ましくは 7 0個以上、 より好ましくは 1 0 0個以 上、 最も好ましくは 2 0 0個以上のアミノ酸配列を有するペプチドなどが用い ら  Specifically, for the purpose of preparing the antibody of the present invention described later, the amino acids represented by SEQ ID NO: 1 to 1 to 101, 1 87 to 360, and 57 A peptide having an amino acid sequence of the 5th to 69th amino acid sequence, the 1st to 101st, the 187th to 360th, and the 575th to 6995th amino acid sequence represented by SEQ ID NO: 3 A peptide having the amino acid sequence at position No. 1-101, 187-360, and 575-699 in the amino acid sequence represented by SEQ ID NO: 5; And the like. For example, at least 20 or more, preferably 50 or more, more preferably 70 or more, more preferably 100 or more, and most preferably, of the constituent amino acid sequences of the protein used in the present invention. Is a peptide having 200 or more amino acid sequences.
また、 本発明で用いられる部分ペプチドは、 そのアミノ酸配列中の 1または 2個以上 (例えば 1〜 2 0個程度、 好ましくは 1〜1 0個程度、 さらに好まし くは数 (1〜5 ) 個) のアミノ酸が欠失し、 または、 そのアミノ酸配列に 1ま たは 2個以上 (例えば 1〜 2 0個程度、 好ましくは 1〜1 0個程度、 さらに好 ましくは数 (1〜5 ) 個) のアミノ酸が付加し、 または、 そのアミノ酸配列に 1または 2個以上 (例えば 1〜 2 0個程度、 好ましくは 1〜1 0個程度、 さら に好ましくは数 (1〜5 ) 個) のアミノ酸が揷入され、 または、 そのアミノ酸 配列中の 1または 2個以上 (例えば 1〜2 0個程度、 好ましくは 1〜1 0個程 度、 さらに好ましくは数 (1〜5 ) 個) のアミノ酸が他のアミノ酸で置換され ていてもよい。  The partial peptide used in the present invention may have one or two or more amino acids in its amino acid sequence (for example, about 1 to 20, preferably about 1 to 10, more preferably about 1 to 5). Amino acids are deleted, or one or more amino acids are added to the amino acid sequence (for example, about 1 to 20, preferably about 1 to 10, more preferably about 1 to 5 )) Or one or more amino acids (eg, about 1 to 20, preferably about 1 to 10, more preferably number (1 to 5)) to the amino acid sequence. Or one or more amino acids in the amino acid sequence (for example, about 1 to 20 amino acids, preferably about 1 to 10 amino acids, more preferably about 1 to 5 amino acids). An amino acid may be substituted with another amino acid.
また、 本発明で用いられる部分ペプチドは C末端が力ルポキシル基 (- C00H) 、 カルポキシレート (-COO") 、 アミド (- C0NH2) またはエステル (- C00R) の何れであってもよい。 The partial peptide used in the present invention, the C-terminus force Rupokishiru group (- C00H), Karupokishireto (-COO "), amido (- C0NH 2) or ester (- C00R) may be either.
さらに、 本発明で用いられる部分ペプチドには、 前記した本発明で用いられ るタンパク質と同様に、 C末端以外に力ルポキシル基.(またはカルポキシレー ト) を有しているもの、 N末端のアミノ酸残基 (例、 メチォニン残基) のアミ 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 夕ミン残基がピ口ダル夕ミン酸化したもの、 分子内のァ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 ぺプチドなどの複合ペプチドなども含まれる。 Further, similar to the protein used in the present invention, the partial peptide used in the present invention has a lipoxyl group (or carboxylate) other than the C-terminal, and the N-terminal amino acid residue. When the amino group of a group (eg, methionine residue) is protected by a protecting group, the dal formed by cleavage of the N-terminal side in vivo The amino acid residue has been oxidized with pip dar, the substituent on the side chain of the amino acid in the molecule is protected with an appropriate protecting group, or the so-called sugar peptide to which a sugar chain is bound. Complex peptides and the like are also included.
本発明で用いられる部分ペプチドは抗体作成のための抗原としても用いるこ とができる。  The partial peptide used in the present invention can also be used as an antigen for producing an antibody.
本発明で用いられるタンパク質または部分ペプチドの塩としては、 生理学的 に許容される酸 (例、 無機酸、 有機酸) や塩基 (例、 アルカリ金属) などとの 塩が用いられ、 とりわけ生理学的に許容される酸付加塩が好ましい。 この様な 塩としては、 例えば、 無機酸 (例えば、 塩酸、 リン酸、 臭化水素酸、 硫酸) と の塩、 あるいは有機酸 (例えば、 酢酸、 ギ酸、 プロピオン酸、 フマル酸、 マレ イン酸、 コハク酸、 酒石酸、 クェン酸、 リンゴ酸、 蓚酸、 安息香酸、 メタンス ルホン酸、 ベンゼンスルホン酸) との塩などが用いられる。  As the salt of the protein or partial peptide used in the present invention, salts with physiologically acceptable acids (eg, inorganic acids, organic acids) and bases (eg, alkali metals) are used. Acceptable acid addition salts are preferred. Such salts include, for example, salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid) or organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, Salts with succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid) are used.
本発明で用いられるタンパク質もしくはその部分べプチドまたはその塩は、 前述したヒ卜や温血動物の細胞または組織から自体公知のタンパク質の精製方 法によって製造することもできるし、 タンパク質をコードする D NAを含有す る形質転換体を培養することによつても製造することができる。 また、 後述の ぺプチド合成法に準じて製造することもできる。  The protein or its partial peptide or a salt thereof used in the present invention can be produced from the above-mentioned human or warm-blooded animal cells or tissues by a known method for purifying a protein, or a protein encoding D It can also be produced by culturing a transformant containing NA. It can also be produced according to the peptide synthesis method described below.
ヒトゃ哺乳動物の組織または細胞から製造する場合、 ヒトゃ哺乳動物の組織 または細胞をホモジナイズした後、 酸などで抽出を行ない、 該抽出液を逆相ク ロマ卜グラフィ一、 イオン交換クロマトグラフィーなどのクロマトグラフィー を組み合わせることにより精製単離することができる。  When producing from human or mammalian tissues or cells, the human or mammalian tissues or cells are homogenized, then extracted with an acid or the like, and the resulting extract is subjected to reversed phase chromatography, ion exchange chromatography, etc. Purification and isolation can be performed by combining the above chromatography.
本発明で用いられるタンパク質もしくは部分ペプチドまたはその塩、 または そのアミド体の合成には、 通常市販のタンパク質合成用樹脂を用いることがで きる。 そのような樹脂としては、 例えば、 クロロメチル樹脂、 ヒドロキシメチ ル樹脂、 ベンズヒドリルァミン樹脂、 アミノメチル樹脂、 4—ベンジルォキシ ベンジルアルコール樹脂、 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―ヒドロキシメチ ル) フエノキシ樹脂、 4一 (2, , 4, 一ジメトキシフエ二ルー F m o。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—アミノ基と側鎖官能基を適当に保護したアミノ酸を、 目的とするタン パク質の配列通りに、 自体公知の各種縮合方法に従い、 樹脂上で縮合させる。 反応の最後に樹脂からタンパク質または部分ペプチドを切り出すと同時に各種 保護基を除去し、 さらに高希釈溶液中で分子内ジスルフイド結合形成反応を実 施し、 目的のタンパク質もしくは部分ペプチドまたはそれらのアミド体を取得 する。 For the synthesis of the protein or partial peptide or a salt thereof, or an amide thereof used in the present invention, a commercially available resin for protein synthesis can be usually used. Examples of such resins include chloromethyl resin, hydroxymethyl resin, benzhydrylamine resin, aminomethyl resin, 4-benzyloxybenzyl alcohol resin, 4-methylbenzhydrylamine resin, PAM resin, 4 —Hydroxymethylmethylphenylacetamidomethyl resin, polyacrylamide resin, 4- (2,, 4'-dimethoxyphenyl-hydroxymethyl) phenoxy resin, 4- (2,, 4,1-dimethoxyphenyl) F mo Ami Noethyl) phenoxy resin and the like. Using such a resin, amino acids having a suitably protected amino group and side chain functional group are condensed on the resin in accordance with the sequence of the target protein in accordance with various known condensation methods. At the end of the reaction, the protein or partial peptide is cleaved from the resin, and at the same time, various protecting groups are removed.In addition, an intramolecular disulfide bond formation reaction is performed in a highly diluted solution to obtain the target protein or partial peptide or an amide thereof. I do.
上記した保護アミノ酸の縮合に関しては、 タンパク質合成に使用できる各種 活性化試薬を用いることができるが、 特に、 カルポジイミド類がよい。 力ルポ ジイミド類としては、 D C C、 N, N, —ジイソプロピルカルポジイミド、 N 一ェチル _ 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 HO B tエステルあるいは H〇 O B tエステルとしてあらかじめ保護 アミノ酸の活性化を行なった後に樹脂に添加することができる。  For the condensation of the protected amino acids described above, various activating reagents that can be used for protein synthesis can be used, and carbodiimides are particularly preferable. As the disulfide diimides, DCC, N, N, -diisopropylcarpo- imide, N-ethyl-Ν '-(3-dimethylaminoprolyl) carbopimide, and the like are used. For these activations, the protected amino acid may be added directly to the resin with a racemization inhibitor additive (e.g., HOB t, HOOB t), or may be pre-protected as a symmetrical anhydride or HOB t ester or H〇OB t ester The amino acid can be added to the resin after activation.
保護アミノ酸の活性化や樹脂との縮合に用いられる溶媒としては、 タンパク 質縮合反応に使用しうることが知られてレゝ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。 反応温度は夕ンパク質結合形成反応に使用され得ることが知られ ている範囲から適宜選択され、 通常約— 2 0 °C〜5 0 °Cの範囲から適宜選択さ れる。 活性化されたアミノ酸誘導体は通常 1 . 5〜4倍過剰で用いられる。 ニン ヒドリン反応を用いたテストの結果、 縮合が不十分な場合には保護基の脱離を 行なうことなく縮合反応を繰り返すことにより十分な縮合を行なうことができ る。 反応を繰り返しても十分な縮合が得られないときには、 無水酢酸またはァ セチルイミダゾールを用いて未反応アミノ酸をァセチル化することによって、 後の反応に影響を与えないようにすることができる。 The solvent used for activating the protected amino acid or condensing with the resin may be appropriately selected from solvents known to be usable for the protein condensation reaction. For example, acid amides such as N, N-dimethylformamide, N, N-dimethylacetoamide, N-methylpyrrolidone, halogenated hydrocarbons such as methylene chloride, chloroform, trifluoroethanol, etc. Alcohols, sulphoxides 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. Used. The reaction temperature is appropriately selected from a range known to be usable for the protein bond formation reaction, and is usually appropriately selected from a 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, if the condensation is insufficient, sufficient condensation can be performed by repeating the condensation reaction without removing the protecting group. If sufficient condensation is not obtained even after repeating the reaction, use acetic anhydride or By acetylating unreacted amino acids using cetyl imidazole, it is possible to prevent the subsequent reactions from being affected.
原料のァミノ基の保護基としては、 例えば、 τ、 B o c、 t一ペンチルォキ シカルポニル、 イソポルニルォキシカルボニル、 4ーメトキシベンジルォキシ 力ルポニル、 C 1 _ Z、 B r— Z、 ァダマンチルォキシカルポニル、 トリフル ォロアセチル、 フタロイル、 ホルミル、 2—ニトロフエニルスルフエ二ル、 ジ フエニルホスフィノチオイル、 Fm o cなどが用いられる。  Examples of the protecting group for the amino group of the raw material include τ, Boc, t-pentyloxycarponyl, isopolnyloxycarbonyl, 4-methoxybenzyloxyl-ponyl, C 1 _Z, Br—Z, and adaman Tyloxycarbonyl, trifluoroacetyl, phthaloyl, formyl, 2-nitrophenylsulfenyl, diphenylphosphinothioyl, Fmoc and the like are used.
力ルポキシル基は、 例えば、 アルキルエステル化 (例えば、 メチル、 ェチ ル、 プロピル、 ブチル、 t -ブチル、 シクロペンチル、 シクロへキシル、 シク 口へプチル、 シクロォクチル、 2—ァダマンチルなどの直鎖状、 分枝状もしく は環状アルキルエステル化) 、 ァラルキルエステル化 (例えば、 ベンジルエス テル、 4一二トロべンジルエステル、 4—メトキシベンジルエステル、 4ーク ロロべンジルエステル、 ベンズヒドリルエステル化) 、 フエナシルエステル 化、 ベンジルォキシカルポニルヒドラジド化、 t一ブトキシカルポニルヒドラ ジド化、 トリチルヒドラジド化などによつて保護することができる。  Lepoxyl groups are, for example, alkyl esterified (e.g., methyl, ethyl, propyl, butyl, t-butyl, cyclopentyl, cyclohexyl, cyclopentyl, cyclooctyl, 2-adamantyl, etc.). Branched or cyclic alkyl esterification), aralkyl esterification (eg, benzyl ester, 412 trobenzyl ester, 4-methoxybenzyl ester, 4-chlorobenzyl ester, benzhydryl esterification), phene It can be protected by nasylation, benzyloxycarbonyl hydrazide, t-butoxycarbonyl hydrazide, trityl hydrazide, or the like.
セリンの水酸基は、 例えば、 エステル化またはエーテル化によって保護する ことができる。 このエステル化に適する基としては、 例えば、 ァセチル基など の低級 ( Cト 6) アルカノィル基、 ベンゾィル基などのァロイル基、 ベンジルォ キシカルポニル基、 エトキシカルポニル基などの炭酸から誘導される基などが 用いられる。 また、 エーテル化に適する基としては、 例えば、 ベンジル基、 テ トラヒドロピラニル基、 t一ブチル基などである。 The hydroxyl group of serine can be protected, for example, by esterification or etherification. As a group suitable for the esterification, for example, a group derived from carbonic acid such as a lower (C 6 ) alkanol group such as an acetyl group, an aroyl group such as a benzoyl group, a benzyloxycarbonyl group, and an ethoxycarbonyl group can be used. Can be Examples of a group suitable for etherification include a benzyl group, a tetrahydropyranyl group, and a t-butyl group.
チロシンのフエノ一ル性水酸基の保護基としては、 例えば、 B z 1、 C 1 2 — B z l、 2—ニトロベンジル、 B r—Z、 t -ブチルなどが用いられる。 ヒスチジンのイミダゾールの保護基としては、 例えば、 T o s、 4—メトキ シ一 2, 3, 6—トリメチルベンゼンスルホニル、 D N P、 ベンジルォキシメ チル、 B u m、 B o c、 T r t、 Fm o cなどが用いられる。  As the protecting group for the phenolic hydroxyl group of tyrosine, for example, Bz1, C12-Bzl, 2-nitrobenzyl, Br-Z, t-butyl and the like are used. As the protecting group for imidazole of histidine, for example, Tos, 4-methoxy-1,2,3,6-trimethylbenzenesulfonyl, DNP, benzyloxymethyl, Bum, Boc, Trt, Fmoc and the like are used.
原料の力ルポキシル基の活性化されたものとしては、 例えば、 対応する酸無 水物、 アジド、 '活性エステル 〔アルコール (例えば、 ペンタクロロフエノ一 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—ヒドロキシフ夕ルイミド、 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。 Examples of the activated carbonyl group of the raw material include, for example, a corresponding acid anhydride, azide, and an active ester [alcohol (eg, pentachlorophenol, 2,4,5-trichlorophenol, 2 , 4-dinitrophenol, shear Nomethyl alcohol, p-nitrophenol, H〇NB, N-hydroxysuccinimide, N-hydroxyfurimide, ester with HOBt)]. As the activated amino group of the raw material, for example, a corresponding phosphoramide is used.
保護基の除去 (脱離) 方法としては、 例えば、 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、 トリフルォロメ夕ンスルホン酸、 トリフルォロ酢酸あるい はこれらの混合液などによる酸処理や、 ジイソプロピルェチルァミン、 トリエ チルァミン、 ピぺリジン、 ピペラジンなどによる塩基処理、 また液体アンモニ ァ中ナトリウムによる還元なども用いられる。 上記酸処理による脱離反応は、 一般に約一 2 0 Τ:〜4 0 °Cの温度で行なわれるが、 酸処理においては、 例え ば、 ァニソール、 フエノール、 チオアニソ一ル、 メタクレゾ一ル、 パラクレゾ ール、 ジメチルスルフィド、 1, 4ープ夕ンジチオール、 1 , 2—エタンジチォー ルなどのようなカチオン捕捉剤の添加が有効である。 また、 ヒスチジンのイミ ダゾール保護基として用いられる 2 , 4—ジニトロフエニル基はチオフエノ一 ル処理により除去され、 トリプトファンのィンドール保護基として用いられる ホルミル基は上記の 1, 2—エタンジチオール、 1, 4一ブタンジチオールなどの 存在下の酸処理による脱保護以外に、 希水酸化ナトリウム溶液、 希アンモニア などによるアルカリ処理によっても除去される。  Methods for removing (eliminating) protecting groups include, for example, catalytic reduction in a hydrogen stream in the presence of a catalyst such as Pd-black or Pd-carbon, or hydrogen fluoride anhydride or methanesulfonic acid. Acid treatment with trifluoromethanesulfonic acid, trifluoroacetic acid or a mixture thereof, base treatment with diisopropylethylamine, triethylamine, piperidine, piperazine, etc., and reduction with sodium in liquid ammonia, etc. Used. The elimination reaction by the above acid treatment is generally carried out at a temperature of about 120 ° C. to about 40 ° C. In the acid treatment, for example, anisol, phenol, thioanisole, methacrylol, paracresol, etc. It is effective to add a cation scavenger such as toluene, dimethyl sulfide, 1,4-butanedithiol, or 1,2-ethanedithiol. In addition, the 2,4-dinitrophenyl group used as an imidazole protecting group of histidine is removed by thiophenol treatment, and the formyl group used as an indole protecting group of tryptophan is substituted with 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 material, the protection group, the elimination of the protective group, and the activation of the functional group involved in the reaction can be appropriately selected from known groups or known means. ·
タンパク質または部分ペプチドのアミド体を得る別の方法としては、 例え ば、 まず、 カルポキシ末端アミノ酸のひ一力ルポキシル基をアミド化して保護 した後、 アミノ基側にペプチド (タンパク質) 鎖を所望の鎖長まで延ばした 後、 該ペプチド鎖の N末端の ο;—ァミノ基の保護基のみを除いたタンパク質ま たは部分ペプチドと C末端の力ルポキシル基の保護基のみを除去したタンパク 質または部分べプチドとを製造し、 これらのタンパク質またはべプチドを上記 したような混合溶媒中で縮合させる。 縮合反応の詳細については上記と同様で ある。 縮合により得られた保護タンパク質またはペプチドを精製した後、 上記 方法によりすベての保護基を除去し、 所望の粗タンパク質またはペプチドを得 ることができる。 この粗夕ンパク質またはべプチドは既知の各種精製手段を駆 使して精製し、 主要画分を凍結乾燥することで所望のタンパク質またはべプチ ドのアミド体を得ることができる。 Another method for obtaining an amide form of a protein or partial peptide is, for example, first protecting a carboxy-terminal amino acid with the desired amino acid by amidating and protecting the amino acid side of the peptide (protein) chain. After lengthening, the protein or partial peptide from which only the ο; -amino protecting group at the N-terminus of the peptide chain has been removed, and the protein or partial protein from which only the protecting group at the C-terminal carbonyl group has been removed. A peptide is produced, and these proteins or peptides are condensed in a mixed solvent as described above. The details of the condensation reaction are the same as above. is there. After purifying the protected protein or peptide obtained by the condensation, all the protecting groups are removed by the above method, and a desired crude protein or peptide can be obtained. This crude protein or peptide can be purified using various known purification means, and the main fraction can be lyophilized to obtain an amide of the desired protein or peptide.
タンパク質またはペプチドのエステル体を得るには、 例えば、 力ルポキシ末 端アミノ酸の α—力ルポキシル基を所望のアルコール類と縮合しアミノ酸エス テルとした後、 タンパク質またはペプチドのアミド体と同様にして、 所望の夕 ンパク質またはぺプチドのエステル体を得ることができる。  To obtain an ester of a protein or peptide, for example, after condensing the α-lipoxyl group of the terminal amino acid with a desired alcohol to form an amino acid ester, in the same manner as the amide of a protein or peptide, The desired ester of the protein or peptide can be obtained.
本発明で用いられる部分ペプチドまたはそれらの塩は、 自体公知のペプチド の合成法に従って、 あるいは本発明で用いられるタンパク質を適当なぺプチダ ーゼで切断することによって製造することができる。 ぺプチドの合成法として は、 例えば、 固相合成法、 液相合成法のいずれによっても良い。 すなわち、 本 発明で用いられる部分べプチドを構成し得る部分べプチドもしくはアミノ酸と 残余部分とを縮合させ、 生成物が保護基を有する場合は保護基を脱離すること により目的のぺプチドを製造することができる。 公知の縮合方法や保護基の脱 離としては、 例えば、 以下の ω 〜 (V) に記載された方法が挙げられる。  The partial peptide used in the present invention or a salt thereof can be produced according to a peptide synthesis method known per se, or by cleaving the protein used in the present invention with an appropriate peptidase. As a method for synthesizing a peptide, for example, any of a solid phase synthesis method and a liquid phase synthesis method may be used. That is, the partial peptide or amino acid that can constitute the partial peptide used in the present invention is condensed with the remaining portion, and when the product has a protecting group, the protecting group is eliminated to produce the desired peptide. can do. Examples of the known condensation method and elimination of the protecting group include the methods described in the following [omega] to (V).
(0 M. Bodanszkyおよび M. A. Ondet t i, ぺプチド ·シンセシス (Pept i de Synthes is) , Interscience Publ ishers, New York (1966年)  (0 M. Bodanszky and M.A. Ondet t i, Pept i de Synthes is, Interscience Publ ishers, New York (1966)
(i i) Schroederおよび Luebkeゝ ザ ·ペプチド(The Pept ide) , Academic Press New York (1965年)  (ii) Schroeder and Luebke ゝ The Peptide, Academic Press New York (1965)
(i i i) 泉屋信夫他、 ペプチド合成の基礎と実験、 丸善 (株) (1975年) (i i i) 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、 タンパク質の化学 IV、 205、 (1977年) (iv) Haruaki Yajima and Shunpei Sakakibara, Laboratory of Biochemistry 1, Protein Chemistry IV, 205, (1977)
(V) 矢島治明監修、 続医薬品の開発、 第 14巻、 ペプチド合成、 広川書店 また、 反応後は通常の精製法、 例えば、 溶媒抽出 ·蒸留 ·カラムクロマトグ ラフィ一 '液体クロマトグラフィー '再結晶などを組み合わせて本発明で用い られる部分べプチドを精製単離することができる。 上記方法で得られる部分べ プチドが遊離体である場合は、 公知の方法あるいはそれに準じる方法によって 適当な塩に変換することができるし、 逆に塩で得られた場合は、 公知の方法あ るいはそれに準じる方法によって遊離体または他の塩に変換することができ る。 (V) Supervised by Haruaki Yajima, Development of Continuing Pharmaceuticals, Vol. 14, Peptide Synthesis, Hirokawa Shoten In addition, after the reaction, conventional purification methods, for example, solvent extraction, distillation, column chromatography, 'liquid chromatography' recrystallization The partial peptides used in the present invention can be purified and isolated by combining these methods. When the partial peptide obtained by the above method is a free form, it may be obtained by a known method or a method analogous thereto. The compound can be converted to an appropriate salt, or conversely, when obtained as a salt, can be converted to a free form or another salt by a known method or a method analogous thereto.
本発明で用いられるタンパク質をコードするポリヌクレオチドとしては、 前 述した本発明で用いられるタンパク質をコードする塩基配列を含有するもので あればいかなるものであってもよい。 好ましくは DNAである。 DNAとして は、 ゲノム DNA、 ゲノム DNAライブラリ一、 前記した細胞 ·組織由来の c DNA、 前記した細胞 ·組織由来の c DNAライブラリー、 合成 DNAのいず れでもよい。  The polynucleotide encoding the protein used in the present invention may be any polynucleotide containing the above-described nucleotide sequence encoding the protein used in the present invention. Preferably it is DNA. The DNA may be any of genomic DNA, genomic DNA library, the above-described cell / tissue-derived cDNA, the above-described cell / tissue-derived cDNA library, and synthetic DNA.
ライブラリーに使用するべク夕一は、 パクテリオファージ、 プラスミド、 コ スミド、 ファ一ジミドなどいずれであってもよい。 また、 前記した細胞 ·組織 より totalRNAまたは mRNA画分を調製したものを用いて直接 Reverse Transcriptase Polymerase Chain Reaction (以下、 RT— PCR法と略称す る) によって増幅することもできる。  The vector used for the library may be any of pacteriophage, plasmid, cosmid, and phagemid. Alternatively, it can also be directly amplified by Reverse Transcriptase Polymerase Chain Reaction (hereinafter abbreviated as RT-PCR method) using a total RNA or mRNA fraction prepared from the cells and tissues described above.
本発明で用いられるタンパク質をコードする DNAとしては、 例えば、 配列 番号: 2で表される塩基配列を含有する DNA、 または配列番号: 2で表され る塩基配列とハイストリンジェントな条件下でハイプリダイズする塩基配列を 含有し、 配列番号: 1で表されるアミノ酸配列を含有するタンパク質と実質的 に同質の性質を有するタンパク質をコードする D N Aであれば何れのものでも よい。  The DNA encoding the protein used in the present invention includes, for example, a DNA containing the nucleotide sequence represented by SEQ ID NO: 2 or a DNA hybridizing with the nucleotide sequence represented by SEQ ID NO: 2 under high stringency conditions. Any DNA may be used as long as it contains a soybean base sequence and encodes a protein having substantially the same properties as the protein containing the amino acid sequence represented by SEQ ID NO: 1.
配列番号: 2で表される塩基配列とハイストリンジェントな条件下でハイブ リダィズできる DNAとしては、 例えば、 配列番号: 2で表される塩基配列と 約 60%以上、 好ましくは約 70%以上、 好ましくは約 80%以上、 好ましく は約 90 %以上、 好ましくは約 95 %以上の相同性を有する塩基配列を含有す る DNAなどが用いられる。 具体的には、 配列番号: 4で表される塩基配列を 含有する DNA、 配列番号: 6で表される塩基配列を含有する DNAなどが挙 げられる。  Examples of the DNA that can hybridize with the nucleotide sequence represented by SEQ ID NO: 2 under high stringency conditions include, for example, about 60% or more, preferably about 70% or more of the nucleotide sequence represented by SEQ ID NO: 2. DNA containing a base sequence having a homology of about 80% or more, preferably about 90% or more, and preferably about 95% or more is used. Specific examples include a DNA containing the base sequence represented by SEQ ID NO: 4, a DNA containing the base sequence represented by SEQ ID NO: 6, and the like.
ハイブリダィゼーシヨンは、 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 'クロー二.ング (Molecular Cloning) 2nd (J. Sambrook et al . , Col d 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 is carried out by a method known per se or a method analogous thereto, for example, Molecular Cloning 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). When a commercially available library is used, it can be performed according to the method described in the attached instruction manual. More preferably, the reaction can be performed under high stringent conditions.
ハイストリンジェン卜な条件とは、 例えば、 ナトリウム濃度が約 19〜40mM、 好ましくは約 19〜20mMで、 温度が約 50〜70t:、 好ましくは約 60〜65 の条件を 示す。 特に、 ナトリゥム濃度が約 19mMで温度が約 65°Cの場合が最も好ましい。 より具体的には、 配列番号: 1で表されるアミノ酸配列を含有するタンパク 質をコードする D NAとしては、 配列番号: 2で表される塩基配列を含有する D NA、 配列番号: 3で表されるアミノ酸配列を含有するタンパク質をコード する D NAとしては、 配列番号: 4で表される塩基配列を含有する D NA、 配 列番号: 5で表されるアミノ酸配列を含有するタンパク質をコードする D NA としては、 配列番号: 6で表される塩基配列を含有する D NAなどが用いられ る。  The high stringent conditions refer to, 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 t: 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. More specifically, as the DNA encoding the protein containing the amino acid sequence represented by SEQ ID NO: 1, the DNA containing the base sequence represented by SEQ ID NO: 2; As the DNA encoding the protein containing the amino acid sequence represented by SEQ ID NO: 4, a DNA containing the nucleotide sequence represented by SEQ ID NO: 4 and a protein containing the amino acid sequence represented by SEQ ID NO: 5 are encoded. As the DNA to be used, a DNA containing the base sequence represented by SEQ ID NO: 6 and the like are used.
本発明で用いられる部分べプチドをコ一ドするポリヌクレオチド (例、 D N A) としては、 前述した本発明で用いられる部分ペプチドをコードする塩基配 列を含有するものであればいかなるものであってもよい。 また、 ゲノム D N A、 ゲノム D NAライプラリー、 前記した細胞 ·組織由来の c D NA、 前記し た細胞 ·組織由来の c D NAライブラリー、 合成 D N Aのいずれでもよい。 本発明で用いられる部分ペプチドをコードする D NAとしては、 例えば、 配 列番号: 2で表される塩基配列を含有する D NAの一部分を有する D NA、 ま たは配列番号: 2で表される塩基配列とハイストリンジェントな条件下でハイ ブリダィズする塩基配列を含有し、 本発明のタンパク質と実質的に同質の活性 を有するタンパク質をコードする D N Aの一部分を含有する D N Aなどが用い られる。  The polynucleotide (e.g., DNA) encoding the partial peptide used in the present invention may be any polynucleotide containing a base sequence encoding the partial peptide used in the present invention described above. Is also good. Further, it may be any of genomic DNA, genomic DNA library, the above-described cell-tissue-derived cDNA, the above-described cell-tissue-derived cDNA library, and synthetic DNA. Examples of the DNA encoding the partial peptide used in the present invention include, for example, a DNA having a part of the DNA containing the nucleotide sequence represented by SEQ ID NO: 2, or SEQ ID NO: 2. DNAs that contain a base sequence that hybridizes with a base sequence under high stringency conditions and that contain a part of a DNA encoding a protein having substantially the same activity as the protein of the present invention are used.
配列番号: 2で表される塩基配列とハイブリダィズできる D NAは、 前記と 同意義を示す。  DNA capable of hybridizing with the nucleotide sequence represented by SEQ ID NO: 2 has the same significance as described above.
ハイブリダィゼーションの方法および八イストリンジェントな条件は前記と 同様のものが用いられる。 本発明で用いられるタンパク質、 部分ペプチド (以下、 これらをコードする DN Aのクローニングおよび発現の説明においては、 これらを単に本発明の夕 ンパク質と略記する場合がある) を完全にコードする DNAのクローニングの 手段としては、 本発明のタンパク質をコードする塩基配列の一部分を有する合 成 DNAプライマ一を用いて PCR法によって増幅するか、 または適当なべク 夕一に組み込んだ D N Aを本発明の夕ンパク質の一部あるいは全領域をコ一ド する DN A断片もしくは合成 DN Aを用いて標識したものとのハイブリダィゼ ーションによって選別することができる。 ハイブリダィゼ一シヨンの方法は、 例えば、 モレキュラー ·クローニング (Molecular Cloning) 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989) に記載の方法などに従って行 なうことができる。 また、 市販のライブラリーを使用する場合、 添付の使用説 明書に記載の方法に従って行なうことができる。 The same hybridization method and eight stringent conditions as described above are used. The DNA and the partial peptide used in the present invention (hereinafter, in the description of the cloning and expression of the DNAs encoding them, these may be simply abbreviated to the protein of the present invention) may be used to completely encode the DNA. As a means for cloning, the DNA is amplified by PCR using a synthetic DNA primer having a part of the nucleotide sequence encoding the protein of the present invention, or the DNA incorporated into an appropriate vector is used as the cloning DNA of the present invention. Selection can be carried out by hybridization with a DNA fragment coding for a part or the entire region of the quality or labeled with a synthetic DNA. The hybridization method can be performed, for example, according to the method described in Molecular Cloning 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). When a commercially available library is used, it can be performed according to the method described in the attached instruction manual.
D N Aの塩基配列の変換は、 PCR、 公知のキット、 例えば、 Mutan™-super Express Km (宝酒造 (株) ) 、 Mutan™- K (宝酒造 (株) ) 等を用いて、 ODA- LA PCR法、 Gapped duplex法、 Kunke 1法等の自体公知の方法あるいはそれらに準じ る方法に従つて行なうことができる  The DNA base sequence can be converted using the ODA-LA PCR method using PCR, a known kit, for example, Mutan ™ -super Express Km (Takara Shuzo Co., Ltd.), Mutan ™ -K (Takara Shuzo Co., Ltd.), etc. It can be carried out according to a method known per se such as the gapped duplex method, the Kunke method, or a method analogous thereto.
クローン化されたタンパク質をコードする DNAは目的によりそのまま、 ま たは所望により制限酵素で消化したり、 リンカ一を付加したりして使用するこ とができる。 該 DNAはその 5' 末端側に翻訳開始コドンとしての ATGを有 し、 また 3' 末端側には翻訳終止コドンとしての TAA、 TGAまたは TAG を有していてもよい。 これらの翻訳開始コドンや翻訳終止コドンは、 適当な合 成 DN Aアダプタ一を用いて付加することもできる。  The DNA encoding the cloned protein can be used as it is depending on the purpose, or can be digested with a restriction enzyme or added with 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 termination codon at the 3' end. These translation initiation codon and translation termination codon can also be added using an appropriate synthetic DNA adapter.
本発明のタンパク質の発現ベクターは、 例えば、 (ィ) 本発明のタンパク質 をコードする DNAから目的とする DNA断片を切り出し、 (口) 該 DNA断 片を適当な発現ベクター中のプロモ一ターの下流に連結することにより製造す ることができる。  The expression vector for the protein of the present invention includes, for example, (a) cutting out a DNA fragment of interest from DNA encoding the protein of the present invention, and (mouth) downstream of the DNA fragment from a promoter in an appropriate expression vector. It can be manufactured by connecting to
ベクタ一としては、 大腸菌由来のプラスミド (例、 pBR 322, pBR 3 25, pUC l 2, pUC 13) 、 枯草菌由来のプラスミド (例、 pUB 11 0, pTP 5, pC 194) 、 酵母由来プラスミド (例、 pSHl 9, pSH 15) 、 λファージなどのバクテリオファージ、 レトロウイルス, ワクシニア ウィルス, バキュロウィルスなどの動物ウィルスなどの他、 pAl— 11、 p XT1、 pRc/CMV、 pR c/RS V, p c DNA I /N e oなどが用い られる。 ' Examples of the vector include a plasmid derived from Escherichia coli (eg, pBR322, pBR325, pUC12, pUC13), a plasmid derived from Bacillus subtilis (eg, pUB110, pTP5, pC194), a plasmid derived from yeast ( Example, pSHl 9, pSH 15), Bacteriophages such as λ phage, animal viruses such as retrovirus, vaccinia virus, baculovirus, etc., pAl-11, pXT1, pRc / CMV, pRc / RSV, pcDNA I / Neo, etc. Is used. '
本発明で用いられるプロモータ一としては、 遺伝子の発現に用いる宿主に対 応して適切なプロモーターであればいかなるものでもよい。 例えば、 動物細胞 を宿主として用いる場合は、 SR プロモーター、 SV40プロモ一ター、 L TRプロモーター、 CMVプロモーター、 H S V-T Kプロモーターなどが挙げ られる。  The promoter used in the present invention may be any promoter as long as it is appropriate for the host used for gene expression. For example, when an animal cell is used as a host, SR promoter, SV40 promoter, LTR promoter, CMV promoter, HSV-TK promoter and the like can be mentioned.
これらのうち、 CMV (サイトメガロウィルス) プロモ一夕一、 SRaプロ モーターなどを用いるのが好ましい。 宿主がェシエリヒア属菌である場合は、 t r pプロモーター、 l acプロモータ一、 r e cAプロモータ一、 λΡ^プ ロモ—夕—、 ιρρプロモーター、 丁 7プロモー夕一などが、 宿主がバチルス 属菌である場合は、 S ΡΟ 1プロモ一夕一、 SPO 2プロモーター、 p enP プロモーターなど、 宿主が酵母である場合は、 PH05プロモータ一、 PGK プロモーター、 GAPプロモータ一、 ADHプロモーターなどが好ましい。 宿 主が昆虫細胞である場合は、 ボリへドリンプロモーター、 P 10プロモーター などが好ましい。 Of these, it is preferable to use the CMV (cytomegalovirus) promoter, the SRa promoter, and the like. If the host is a genus Escherichia, the host is a Bacillus genus such as trp promoter, lac promoter, recA promoter, λΡ ^ promoter, ι ρρ promoter, and c7 promoter. In this case, the SΡΟ1 promoter overnight, the SPO2 promoter, the penP promoter, and the like. When the host is yeast, the PH05 promoter, the PGK promoter, the GAP promoter, the ADH promoter, and the like are preferable. When the host is an insect cell, a polyhedrin promoter, a P10 promoter and the like are preferable.
発現ベクターには、 以上の他に、 所望によりェンハンサー、 スプライシング シグナル、 ポリ A付加シグナル、 選択マ一カー、 S V40複製オリジン (以 下、 S V40 o r iと略称する場合がある) などを含有しているものを用いる ことができる。 選択マーカ一としては、 例えば、 ジヒドロ葉酸還元酵素 (以 下、 dh f rと略称する場合がある) 遺伝子 〔メソトレキセート (MTX) 耐 性〕 、 アンピシリン耐性遺伝子 (以下、 Amp rと略称する場合がある) 、 ネ ォマイシン耐性遺伝子 (以下、 Ne 01"と略称する場合がある、 G418耐 性) 等が挙げられる。 特に、 dh f r遺伝子欠損チャイニーズハムスター細胞 を用いて dh f r遺伝子を選択マーカーとして使用する場合、 目的遺伝子をチ ミジンを含まない培地によっても選択できる。 The expression vector may contain, in addition to the above, an enhancer, a splicing signal, a poly-A addition signal, a selection marker, an SV40 replication origin (hereinafter sometimes abbreviated as SV40 ori), and the like, if desired. Can be used. The selection marker one, for example, dihydrofolate reductase (hereinafter sometimes abbreviated as dh fr) gene [methotrexate (MTX) resistant], ampicillin resistant gene (hereinafter sometimes abbreviated as Amp r) , Ne Omaishin resistant gene (hereinafter sometimes abbreviated as Ne 0 1 ", G418 resistant) and the like. in particular, when used as a selection marker dh fr gene using dh fr gene-deficient Chinese hamster cells Alternatively, the target gene can be selected using a thymidine-free medium.
また、 '必要に応じて、 宿主に合ったシグナル配列を、 本発明のタンパク質の N端末側に付加する。 宿主がェシエリヒア属菌である場合は、 PhoA 'シグナル 配列、 OmpA ·シグナル配列などが、 宿主がバチルス属菌である場合は、 ーァ ミラーゼ*シグナル配列、 サブチリシン ·シグナル配列などが、 宿主が酵母で ある場合は、 MFa ·シグナル配列、 SUC2 ·シグナル配列など、 宿主が動 物細胞である場合には、 インシュリン ·シグナル配列、 —インターフエ口 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。 In addition, if necessary, a signal sequence suitable for the host may be added to the protein of the present invention. Add to N terminal side. If the host is a genus Escherichia, the PhoA 'signal sequence, OmpA signal sequence, etc., if the host is a Bacillus genus, the amylase * signal sequence, subtilisin signal sequence, etc. In some cases, MFa signal sequence, SUC2 signal sequence, etc. In the case where the host is an animal cell, insulin signal sequence, -interferin signal sequence, antibody molecule, signal sequence, etc. can be used, respectively. .
このようにして構築された本発明のタンパク質をコードする DN Aを含有す るベクターを用いて、 形質転換体を製造することができる。  Using the vector containing the DNA encoding the protein of the present invention thus constructed, a transformant can be produced.
宿主としては、 例えば、 ェシエリヒア属菌、 バチルス属菌、 酵母、 昆虫細 胞、 昆虫、 動物細胞などが用いられる。  As the host, for example, Escherichia bacteria, Bacillus bacteria, yeast, insect cells, insects, animal cells and the like are used.
ェシエリヒア属菌の具体例としては、 例えば、 ェシエリヒア 'コリ  Specific examples of the genus Escherichia include, for example, Escherichia coli.
(Escherichia coli) K 12 · DH 1 [Proc. Natl. Acad. Sci. USA, 60 巻, 160 (1968)〕 , J M 103 [Nucleic Acids Research, 9卷, 309 (1981)〕 , J A221 [Journal of Molecular Biology, 120巻, 517 (1978)] , HB 101 [Journal of Molecular Biology, 41巻, 459 (1969)〕 , C 600 [Genetics, 39 巻,440 (1954)〕 などが用いられる。  (Escherichia coli) K 12 · DH 1 [Proc. Natl. Acad. Sci. USA, 60, 160 (1968)], JM 103 [Nucleic Acids Research, 9, 309 (1981)], J A221 [Journal of Molecular Biology, 120, 517 (1978)], HB101 [Journal of Molecular Biology, 41, 459 (1969)], C600 [Genetics, 39, 440 (1954)] and the like are used.
バチルス属菌としては、 例えば、 バチルス ·サブチルス (Bacillus subtilis) M I 114 〔Gene, 24卷, 255 (1983)〕 , 207 - 21 [Journal of Biochemistry, 95巻, 87 (1984)〕 などが用いられる。  As the Bacillus sp., For example, Bacillus subtilis MI114 [Gene, 24, 255 (1983)], 207-21 [Journal of Biochemistry, 95, 87 (1984)] and the like are used.
酵母としては、 例えば、 サッカロマイセス セレビシェ (Saccharomyces cerevisiae) AH22, AH22 R-, NA87 - 11 A, DKD— 5D, 2 OB— 12、 シゾサッカロマイセス ボンべ (Schizosaccharomyces pombe) N CYC 1913, NCYC 2036、 ピキア パストリス (Pichia pastoris) KM 71などが用いられる。  Examples of yeast include, for example, Saccharomyces cerevisiae AH22, AH22 R-, NA87-11A, DKD-5D, 2OB-12, Schizosaccharomyces pombe N CYC 1913, NCYC 2036, Pichia pastori (Pichia pastoris) KM 71 or the like is used.
昆虫細胞としては、 例えば、 ウィルスが Ac NPVの場合は、 夜盗蛾の幼虫 由来株化細胞 (Spodoptera frugiperda cell; S f細胞) 、 Trichoplusia niの 中腸由来の MG1細胞、 Trichoplusia niの卵由来の High Five™細胞、  Insect cells include, for example, when the virus is Ac NPV, a cell line derived from a larva of night moth (Spodoptera frugiperda cell; S f cell), MG1 cell derived from the midgut of Trichoplusia ni, High derived from egg of Trichoplusia ni Five ™ cells,
Mamestra brassicae由来の細胞または Estigmena acrea由来の細胞などが用いら れる。 ウィルスが BmNP Vの場合は、 蚕由来株化細胞 (Bombyx mori N細 胞; BmN細胞) などが用いられる。 該 S f細胞としては、 例えば、 S f 9細 胞 (ATCC CRL1711) 、 S f 21細胞 (以上、 Vaughn, J.L.ら、 イン ·ヴィポCells derived from Mamestra brassicae or cells derived from Estigmena acrea are used. If the virus is BmNP V, a silkworm-derived cell line (Bombyx mori N cells) Vesicles; BmN cells). Examples of the Sf cells include Sf9 cells (ATCC CRL1711) and Sf21 cells (Vaughn, JL et al., In Vipo).
(In Vivo) , 13, 213-217, (1977)) などが用いられる。 (In Vivo), 13, 213-217, (1977)).
昆虫としては、 例えば、 カイコの幼虫などが用いられる 〔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, Ve r o, チャイニーズ ハムスター細胞 CH〇 (以下、 CHO細胞と略記) , dh f r遺伝子欠損チヤ ィニーズハムスター細胞 CHO (以下、 CHO (dh f r— ) 細胞と略記) , マウス L細胞, マウス A t T- 20, マウスミエ口一マ細胞, マウス ATDC 5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 hamster cells CH〇 (hereinafter abbreviated as CHO cells), dh fr gene-deficient Chinese hamster cells CHO (hereinafter, CHO (dh fr—) cells Abbreviations), mouse L cells, mouse AtT-20, mouse myeloid cells, mouse ATDC 5 cells, rat GH3, human FL cells, etc. are used.
ェシエリヒア属菌を形質転換するには、 例えば、 Proc. Natl. Acad. Sci. USA, 69巻, 2110 (1972)、 Gene, 17巻, 107 (1982)などに記載の方法に従つて行なう ことができる。  Transformation of a genus Escherichia can be performed, for example, according to the method described in Proc. Natl. Acad. Sci. USA, 69, 2110 (1972), Gene, 17, 107 (1982). it can.
バチルス属菌を形質転換するには、 例えば、 モレキュラー 'アンド ·ジエネ ラル-ジェネティックス (Molecular & General Genetics) , 168巻, 11 1 (1979 )などに記載の方法に従つて行なうことができる。  Transformation of Bacillus spp. Can be carried out, for example, according to the method described in Molecular 'and General Genetics (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. Natl. Acad. Sci. USA, 75巻, 1929 (1978)などに記載の方法に 従って行なうことができる。  The yeast can be transformed according to the method described in, for example, Methods in Enzymology, Vol. 194, 182-187 (1991), Proc. Natl. Acad. Sci. USA, Vol. 75, 1929 (1978). 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 口トコ一ル. 263 - 267 (1995) (秀潤社発行) 、 ヴイロロジ一 (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 Experiment Protocol, 263-267 (1995) (published by Shujunsha), Virology, 52, 456 (1973). )).
このようにして、 タンパク質をコードする DN Aを含有する発現ベクターで 形質転換された形質転換体を得ることができる。  Thus, a transformant transformed with the expression vector containing the DNA encoding the protein can be obtained.
宿主がェシエリヒア属菌、 バチルス属菌である形質転換体を培養する際、 培 養に使用される培地としては液体培地が適当であり、 その中には該形質転換体 の生育に必要な炭素源、 窒素源、 無機物その他が含有せしめられる。 炭素源と しては、 例えば、 グルコース、 デキス卜リン、 可溶性澱粉、 ショ糖など、 窒素 源としては、 例えば、 アンモニゥム塩類、 硝酸塩類、 コーンスチープ'リカ 一、 ペプトン、 カゼイン、 肉エキス、 大豆粕、 バレイショ抽出液などの無機ま たは有機物質、 無機物としては、 例えば、 塩化カルシウム、 リン酸二水素ナト リウム、 塩化マグネシウムなどが挙げられる。 また、 酵母エキス、 ビタミン 類、 生長促進因子などを添加してもよい。 培地の pHは約 5〜 8が望ましい。 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 Genet ics, 431- 433, Cold Spring Harbor Laboratory, New York 1972) が好ましい。 ここに必 要によりプロモーターを効率よく働かせるために、 例えば、 3 ;8—インドリル アクリル酸のような薬剤を加えることができる。 When culturing a transformant whose host is a bacterium belonging to the genus Escherichia or Bacillus, a liquid medium is suitable as a medium to be used for cultivation. It contains carbon sources, nitrogen sources, inorganic substances, etc., necessary for the growth of sorghum. Examples of the carbon source include glucose, dextrin, soluble starch, and sucrose. Examples of the nitrogen source include ammonium salts, nitrates, corn chips, lica, peptone, casein, meat extract, and soybean meal. Inorganic or organic substances such as potato extract and inorganic substances include, for example, 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. As a medium for culturing a bacterium belonging to 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, a drug such as, for example, 3; 8-indolylacrylic acid can be added to make the promoter work efficiently.
宿主がェシェリヒア属菌の場合、 培養は通常約 1 5〜 43 °Cで約 3〜 24時 間行ない、 必要により、 通気や撹拌を加えることもできる。  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 applied.
宿主がバチルス属菌の場合、 培養は通常約 30〜 40 °Cで約 6〜 24時間行 ない、 必要により通気や撹拌を加えることもできる。  When the host is a bacterium belonging to the genus Bacillus, the cultivation is usually performed at about 30 to 40 ° C for about 6 to 24 hours, and if necessary, aeration and stirring can be applied.
宿主が酵母である形質転換体を培養する際、 培地としては、 例えば、 バーク ホ一ルダ一 (Burkholder) 最小培地 [Proc. Natl. Acad. Sci. USA, 77  When culturing a transformant in which the host is yeast, for example, Burkholder's minimal medium [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 roc. Natl. Acad. Sci. USA, 81巻, 5330(1984)〕 が挙げられる。 培地の p Hは約 5〜 8に調整する のが好ましい。 培養は通常約 20 〜 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。 Vol. 4505 (1980)] and SD medium containing 0.5% casamino acid roc. Natl. Acad. Sci. USA, 81, 5330 (1984)]. The pH of the medium is preferably adjusted to about 5-8. Culture is usually performed at about 20 to 35 for about 24 to 72 hours, and aeration and agitation are added as necessary.
宿主が昆虫細胞または昆虫である形質転換体を培養する際、 培地としては、 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. 2〜6. 4に調整するのが好ましい。 培養は通常約 27°Cで約 3〜 5日間行ない、 必要 に応じて通気や撹拌を加える。  When culturing an insect cell or a transformant whose host is an insect, the culture medium may be supplemented with Grace's Insect Medium (Nature, 195, 788 (1962)) with appropriate addition of immobilized 10% serum or other additives. Are used. The pH of the medium is preferably adjusted to about 6.2 to 6.4. Culture is usually performed at about 27 ° C for about 3 to 5 days, and aeration and agitation are added as necessary.
宿主が動物細胞である形質転換体を培養する際、 培地としては、 例えば、 約 5〜20%の胎児牛血清を含む MEM培地 [Science, 122卷, 501 (1952)〕 , DM E M培地 [Virology, 8卷, 396 (1959)〕 , R P M I 1 6 4 0培地 [The Journal of the American Medical Assoc iat ion 199巻, 519 (1967)〕 , 1 9 9培地 When culturing a transformant in which the host is an animal cell, the medium may be, for example, MEM medium containing about 5 to 20% fetal bovine serum [Science, 122 vol., 501 (1952)], DM EM medium [Virology, 8 volumes, 396 (1959)], RPMI 1640 medium [The Journal of the American Medical Association 199, 519 (1967)], 199 medium
(Proceeding of the Society for the Biological Medic ine, 73卷, 1 (1950)〕 などが用いられる。 p Hは約 6〜8であるのが好ましい。 培養は通常約 3 0 〜4 0 °Cで約 1 5〜6 0時間行ない、 必要に応じて通気や撹拌を加える。  (Proceeding of the Society for the Biological Medicine, Vol. 73, 1 (1950)), etc. The pH is preferably about 6 to 8. Culture is usually performed at about 30 to 40 ° C. Run for 15-60 hours, adding aeration and / or agitation as needed.
以上のようにして、 形質転換体の細胞内、 細胞膜または細胞外に本発明の夕 ンパク質を生成せしめることができる。  As described above, the protein of the present invention can be produced in the cells of the transformant, in the cell membrane, or outside the cells.
上記培養物から本発明のタンパク質を分離精製するには、 例えば、 下記の方 法により行なうことができる。  The protein of the present invention can be separated and purified from the culture by, for example, the following method.
本発明のタンパク質を培養菌体あるいは細胞から抽出するに際しては、 培養 後、 公知の方法で菌体あるいは細胞を集め、 これを適当な緩衝液に懸濁し、 超 音波、 リゾチームおよび/または凍結融解などによって菌体あるいは細胞を破 壊したのち、 遠心分離やろ過によりタンパク質の粗抽出液を得る方法などが適 宜用いられる。 緩衝液の中に尿素や塩酸グァニジンなどの蛋白質変性剤や、 ト リトン X— 1 0 0 TMなどの界面活性剤が含まれていてもよい。 培養液中にタン パク質が分泌される場合には、 培養終了後、 それ自体公知の方法で菌体あるい は細胞と上清とを分離し、 上清を集める。 When extracting the protein of the present invention from the cultured cells or cells, after the culture, the cells or cells are collected by a known method, suspended in an appropriate buffer, and subjected to ultrasonic wave, lysozyme, and / or freeze-thawing. After the cells or cells are destroyed by the method, a method of obtaining a crude protein extract by centrifugation or filtration is appropriately used. A protein modifier such as urea or hydrochloric guanidine in the buffers may contain a surfactant such as preparative Litton X- 1 0 0 TM. When the protein is secreted into the culture solution, after the culture is completed, the cells or cells are separated from the supernatant by a method known per se, and the supernatant is collected.
このようにして得られた培養上清、 あるいは抽出液中に含まれるタンパク質 の精製は、 自体公知の分離 ·精製法を適切に組み合わせて行なうことができ る。 これらの公知の分離、 精製法としては、 塩析ゃ溶媒沈澱法などの溶解度を 利用する方法、 透析法、 限外ろ過法、 ゲルろ過法、 および S D S—ポリアクリ ルアミドゲル電気泳動法などの主として分子量の差を利用する方法、 イオン交 換クロマトグラフィ一などの荷電の差を利用する方法、 ァフィ二ティークロマ トグラフィーなどの特異的親和性を利用する方法、 逆相高速液体クロマトダラ フィ一などの疎水性の差を利用する方法、 等電点電気泳動法などの等電点の差 を利用する方法などが用いられる。  The protein contained in the culture supernatant or extract obtained in this manner can be purified by appropriately combining known separation and purification methods. These known separation and purification methods include methods using solubility such as salting out and solvent precipitation, dialysis, ultrafiltration, gel filtration, and SDS-polyacrylamide gel electrophoresis, mainly for molecular weight analysis. Method using difference, method using charge difference such as ion exchange chromatography, method using specific affinity such as affinity chromatography, hydrophobicity such as reversed-phase high-performance liquid chromatography, etc. A method utilizing a difference, a method utilizing an isoelectric point difference such as an isoelectric focusing method, and the like are used.
かくして得られるタンパク質が遊離体で得られた場合には、 自体公知の方法 あるいはそれに準じる方法によって塩に変換することができ、 逆に塩で得られ た場合には自体公知の方法あるいはそれに準じる方法により、 遊離体または他 の塩に変換することができる。 When the protein thus obtained is obtained in a free form, it can be converted to a salt by a method known per se or a method analogous thereto, and conversely, when it is obtained as a salt, a method known per se or analogous thereto Depending on the free form or other Can be converted to a salt.
なお、 組換え体が産生するタンパク質を、 精製前または精製後に適当な蛋白 修飾酵素を作用させることにより、 任意に修飾を加えたり、 ポリペプチドを部 分的に除去することもできる。 蛋白修飾酵素としては、 .例えば、 トリプシン、 キモトリブシン、 アルギニルエンドべプチダーゼ、 プロテインキナーゼ、 ダリ コシダーゼなどが用いられる。  The protein produced by the recombinant can be arbitrarily modified or the polypeptide can be partially removed by applying an appropriate protein modifying enzyme before or after purification. As the protein-modifying enzyme, for example, trypsin, chymotrypsin, arginyl endopeptidase, protein kinase, dalicosidase and the like are used.
かくして生成する本発明のタンパク質の存在は、 特異抗体を用いたェンザィ ムィムノアッセィやウエスタンブロッテイングなどにより測定することができ る。  The presence of the thus-produced protein of the present invention can be measured by enzyme immunoassay using a specific antibody, Western blotting, or the like.
本発明で用いられるタンパク質もしくは部分ペプチドまたはその塩に対する 抗体は、 本発明で用いられるタンパク質もしくは部分べプチドまたはその塩を 認識し得る抗体であれば、 ポリクロ一ナル抗体、 モノクローナル抗体の何れで あってもよい。  The antibody against the protein or partial peptide or a salt thereof used in the present invention may be a polyclonal antibody or a monoclonal antibody as long as it can recognize the protein or partial peptide or a salt thereof used in the present invention. Is also good.
本発明で用いられる夕ンパク質もしくは部分べプチドまたはその塩 (以下、 抗体の説明においては、 これらを単に本発明のタンパク質と略記する場合があ る) に対する抗体は、 本発明のタンパク質を抗原として用い、 自体公知の抗体 または抗血清の製造法に従って製造することができる。  An antibody against the protein or partial peptide or a salt thereof (hereinafter, these may be simply referred to as the protein of the present invention) used in the present invention is obtained by using the protein of the present invention as an antigen. The antibody or antiserum can be produced according to a known method for producing an antibody or antiserum.
〔モノクローナル抗体の作製〕  [Preparation of monoclonal antibody]
( a ) モノクローナル抗体産生細胞の作製  (a) Preparation of monoclonal antibody-producing cells
本発明のタンパク質は、 温血動物に対して投与により抗体産生が可能な部位 にそれ自体あるいは担体、 希釈剤とともに投与される。 投与に際して抗体産生 能を高めるため、 完全フロイントアジュバントゃ不完全フロイントアジュバン トを投与してもよい。 投与は通常 2〜 6週毎に 1回ずつ、 計 2〜1 0回程度行 われる。 用いられる温血動物としては、 例えば、 サル、 ゥサギ、 ィヌ、 モルモ ット、 マウス、 ラット、 ヒッジ、 ャギ、 ニヮトリが挙げられるが、 マウスおよ びラットが好ましく用いられる。  The protein of the present invention is administered to a warm-blooded animal itself or together with a carrier or a diluent at a site capable of producing an antibody upon administration. Complete Freund's adjuvant or incomplete Freund's adjuvant may be administered in order to enhance 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 warm-blooded animals to be used include monkeys, rabbits, rabbits, dogs, guinea pigs, mice, rats, sheep, goats, and chickens, and mice and rats are preferably used.
モノクローナル抗体産生細胞の作製に際しては、 抗原で免疫された温血動 物、 例えばマウスから抗体価の認められた個体を選択し最終免疫の 2〜 5曰後 に脾臓またはリンパ節を採取し、 それらに含まれる抗体産生細胞を同種または 異種動物の骨髄腫細胞と融合させることにより、 モノクローナル抗体産生ハイ プリドーマを調製することができる。 抗血清中の抗体価の測定は、 例えば、 後 記の標識化タンパク質と抗血清とを反応させたのち、 抗体に結合した標識剤の 活性を測定することにより行なうことができる。 融合操作は既知の方法、 例え ば、 ケーラーとミルスタインの方法 [: Nature, 256、 495 (1975)〕 に従い実施す ることができる。 融合促進剤としては、 例えば、 ポリエチレングリコール (P EG) 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 preparing monoclonal antibody-producing cells, a warm-blooded animal immunized with an antigen, for example, an individual with an antibody titer is selected from a mouse, and the spleen or lymph node is collected 2 to 5 days after the final immunization. Allogeneic or antibody-producing cells contained in By fusing with a myeloma cell of a heterologous animal, a monoclonal antibody-producing hybridoma can be prepared. The antibody titer in the antiserum can be measured, for example, by reacting a labeled protein described below with the antiserum, and then measuring the activity of a labeling agent bound to the antibody. The fusion operation can be carried out according to a known method, for example, the method of Kohler and Milstein [Nature, 256, 495 (1975)]. Examples of the fusion promoter include polyethylene glycol (PEG) and Sendai virus, and PEG is preferably used.
骨髄腫細胞としては、 例えば、 NS— 1、 P 3U1、 S P 2/0、 AP— 1 などの温血動物の骨髄腫細胞が挙げられるが、 P 3 U 1が好ましく用いられ る。 用いられる抗体産生細胞 (脾臓細胞) 数と骨髄腫細胞数との好ましい比率 は 1 : 1〜20 : 1程度であり、 PEG (好ましくは PEG 1000〜PEG 6000) が 10〜 80 %程度の濃度で添加され、 20〜 40°C、 好ましくは 30〜37°Cで 1〜10分間インキュベートすることにより効率よく細胞融合 を実施できる。  Examples of myeloma cells include myeloma cells of warm-blooded animals such as NS-1, P3U1, SP2 / 0, and AP-1, but P3U1 is preferably used. The preferred ratio between the number of antibody-producing cells (spleen cells) and the number of myeloma cells used is about 1: 1 to 20: 1, and PEG (preferably PEG 1000 to PEG 6000) is used at a concentration of about 10 to 80%. Cell fusion can be carried out efficiently by adding the mixture and incubating at 20 to 40 ° C, preferably 30 to 37 ° C for 1 to 10 minutes.
モノクローナル抗体産生ハイブリドーマのスクリ一ニングには種々の方法が 使用できるが、 例えば、 タンパク質抗原を直接あるいは担体とともに吸着させ た固相 (例、 マイクロプレート) にハイプリドーマ培養上清を添加し、 次に放 射性物質や酵素などで標識した抗免疫グロプリン抗体 (細胞融合に用いられる 細胞がマウスの場合、 抗マウス免疫グロブリン抗体が用いられる) またはプロ ティン 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 グ口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ブリドーマ培養上 清を添加し、 放射性物質や酵素などで標識したタンパク質を加え、 固相に結合 したモノクローナル抗体を検出する方法などが挙げられる。  Various methods can be used to screen monoclonal antibody-producing hybridomas. For example, a hybridoma culture supernatant is added to a solid phase (eg, a microplate) on which a protein antigen is adsorbed directly or together with a carrier. An anti-immunoglobulin antibody labeled with a radioactive substance or enzyme (anti-mouse immunoglobulin antibody is used if the cells used for cell fusion are mice) or protein A is added, and the monoclonal antibody bound to the solid phase is added. Hybridoma culture supernatant is added to the solid phase to which the anti-immunoglobulin antibody or protein A is adsorbed, and proteins labeled with radioactive substances or enzymes are added, and the monoclonal antibody bound to the solid phase is detected. And the like.
. モノクロ一ナル抗体の選別は、 自体公知あるいはそれに準じる方法に従って 行なうことができる。 通常 HAT (ヒポキサンチン、 アミノプテリン、 チミジ ン) を添加した動物細胞用培地で行なうことができる。 選別および育種用培地 としては、 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 RP M I 1 6 4 0培地、 1〜1 0 %の牛胎児血清を含む G I T培地 (和光純薬工業 (株) ) あるいはハイプリドーマ培養用無血清培地 (S F M— 1 0 1、 日水製 薬 (株) ) などを用いることができる。 培養温度は、 通常 2 0〜4 0 °C、 好ま しくは約 3 7 °Cである。 培養時間は、 通常 5日〜 3週間、 好ましくは 1週間〜 2週間である。 培養は、 通常 5 %炭酸ガス下で行なうことができる。 ハイプリ ドーマ培養上清の抗体価は、 上記の抗血清中の抗体価の測定と同様にして測定 できる。 The selection of the monoclonal antibody can be carried out according to a method known per se or a method analogous thereto. Usually, it can be performed in a medium for animal cells supplemented with HAT (hypoxanthine, aminopterin, thymidine). As a selection and breeding medium, any medium can be used as long as it can grow a hybridoma. For example, RP containing 1-20%, preferably 10-20% fetal calf serum MI 1640 medium, GIT medium containing 1 to 10% fetal bovine serum (Wako Pure Chemical Industries, Ltd.) or serum-free medium for hybridoma culture (SFM-101, Nissui Pharmaceutical Co., Ltd. )) Etc. can be used. The culture temperature is usually from 20 to 40 ° C, preferably about 37 ° C. The culturing time is usually 5 days to 3 weeks, preferably 1 week to 2 weeks. The culture can be usually performed under 5% carbon dioxide gas. The antibody titer of the hybridoma culture supernatant can be measured in the same manner as the measurement of the antibody titer in the antiserum described above.
( b ) モノクローナル扰体の精製  (b) Purification of monoclonal antibody
モノクローナル抗体の分離精製は、 自体公知の方法、 例えば、 免疫グロプリ ンの分離精製法 〔例、 塩析法、 アルコール沈殿法、 等電点沈殿法、 電気泳動 法、 イオン交換体 (例、 D E A E) による吸脱着法、 超遠心法、 ゲルろ過法、 抗原結合固相あるいはプロテイン Aあるいはプロティン Gなどの活性吸着剤に より抗体のみを採取し、 結合を解離させて抗体を得る特異的精製法〕 に従って 行なうことができる。  Monoclonal antibodies can be separated and purified by methods known per se, for example, immunoglobulin separation and purification methods (e.g., salting out method, alcohol precipitation method, isoelectric point precipitation method, electrophoresis method, ion exchanger (e.g., DEAE) Adsorption / desorption method, ultracentrifugation method, gel filtration method, specific purification method of collecting antibody only with antigen-bound solid phase or active adsorbent such as protein A or protein G and dissociating the bond to obtain antibody) Can do it.
〔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、 それ自体公知あるいはそれに準じる方法に 従って製造することができる。 例えば、 免疫抗原 (タンパク質抗原) 自体、 あ るいはそれとキヤリァー蛋白質との複合体をつくり、 上記のモノク口一ナル抗 体の製造法と同様に温血動物に免疫を行ない、 該免疫動物から本発明のタンパ ク質に対する抗体含有物を採取して、 抗体の分離精製を行なうことにより製造 することができる。  The polyclonal antibody of the present invention can be produced by a method known per se or a method analogous thereto. For example, an immunizing antigen (protein antigen) itself or a complex thereof with a carrier protein is formed, and immunization is performed on a warm-blooded animal in the same manner as in the method for producing a monoclonal antibody described above. The antibody can be produced by collecting an antibody-containing substance against the protein of the present invention and separating and purifying the antibody.
温血動物を免疫するために用いられる免疫抗原とキヤリァー蛋白質との複合 体に関し、 キヤリァー蛋白質の種類およびキヤリァ一とハプテンとの混合比 は、 キャリアーに架橋させて免疫したハプテンに対して抗体が効率良くできれ ば、 どの様なものをどの様な比率で架橋させてもよいが、 例えば、 ゥシ血清ァ ルブミンゃゥシサイログロブリン、 へモシァニン等を重量比でハプテン 1に対 し、 約 0 . 1〜2 0、 好ましくは約 1〜5の割合でカプルさせる方法が用いら れる。  Regarding the complex of immunizing antigen and carrier protein used to immunize warm-blooded animals, the type of carrier protein and the mixing ratio between carrier and hapten depend on the efficiency of the antibody against hapten immunized by cross-linking with a carrier. If possible, any kind may be crosslinked at any ratio.For example, serum serum albumin, thyroglobulin, hemocyanin, etc. are used in a weight ratio of about 0.1 to 1 for hapten. A method of coupling at a rate of 1 to 20, preferably about 1 to 5 is used.
また、 ハプテンとキャリアーの力プリングには、 種々の縮合剤を用いること ができるが、 ダルタルアルデヒドやカルポジイミド、 マレイミド活性エステ ル、 チオール基、 ジチォピリジル基を含有する活性エステル試薬等が用いられ る。 In addition, various condensing agents must be used for force coupling between the hapten and carrier. However, there can be used, for example, an active ester reagent containing daltaraldehyde, carbodiimide, a maleimide active ester, a thiol group, or a dithiopyridyl group.
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パントゃ不完全フロイントアジュバントを投与してもよ い。 投与は、 通常約 2〜 6週毎に 1回ずつ、 計約 3〜1 0回程度行なわれる。 ポリクローナル抗体は、 上記の方法で免疫された温血動物の血液、 腹水な ど、 好ましくは血液から採取することができる。  The condensation product is administered to a warm-blooded animal at a site where antibody production is possible or together with a carrier or diluent. Complete Freund's adjuvant or incomplete Freund's adjuvant may be administered in order to enhance antibody production ability upon administration. The administration is usually performed once every about 2 to 6 weeks, for a total of about 3 to 10 times. The polyclonal antibody can be preferably collected from blood, such as blood or ascites, of a warm-blooded animal immunized by the above method.
抗血清中のポリクロ一ナル抗体価の測定は、 上記の抗血清中の抗体価の測定 と同様にして測定できる。 ポリクローナル抗体の分離精製は、 上記のモノクロ ーナル抗体の分離精製と同様の免疫グロブリンの分離精製法に従って行なうこ とができる。  The polyclonal antibody titer in the antiserum can be measured in the same manner as the measurement of the antibody titer in the antiserum described above. The separation and purification of the polyclonal antibody can be performed according to the same immunoglobulin separation and purification method as the above-described separation and purification of the monoclonal antibody.
本発明で用いられる夕ンパク質または部分べプチドをコ一ドするボリヌクレ ォチド (例、 D NA (以下、 アンチセンスポリヌクレオチドの説明において は、 これらの D N Aを本発明の D N Aと略記する場合がある) ) の塩基配列に 相補的な、 または実質的に相補的な塩基配列またはその一部を有するアンチセ ンスポリヌクレオチドとしては、 本発明の D N Aの塩基配列に相補的な、 また は実質的に相補的な塩基配列またはその一部を有し、 該 D NAの発現を抑制し 得る作用を有するものであれば、 いずれのアンチセンスポリヌクレオチドであ つてもよいが、 アンチセンス D NAが好ましい。  Polynucleotides encoding the protein or partial peptide used in the present invention (e.g., DNA (hereinafter, these DNAs may be abbreviated as the DNA of the present invention in the description of antisense polynucleotides) The antisense polynucleotide having a nucleotide sequence complementary to or substantially complementary to the nucleotide sequence of)) or a nucleotide sequence complementary to or substantially complementary to the nucleotide sequence of the DNA of the present invention. Any antisense polynucleotide may be used as long as it has a basic nucleotide sequence or a part thereof and has an action capable of suppressing the expression of the DNA, but antisense DNA is preferable.
本発明の D NAに実質的に相補的な塩基配列とは、 例えば、 本発明の D NA に相補的な塩基配列 (すなわち、 本発明の D NAの相補鎖) の全塩基配列ある いは部分塩基配列と約 7 0 %以上、 好ましくは約 8 0 %以上、 より好ましくは 約 9 0 %以上、 最も好ましくは約 9 5 %以上の相同性を有する塩基配列などが 挙げられる。 特に、 本発明の D NAの相補鎖の全塩基配列うち、 (ィ) 翻訳阻 害を指向したアンチセンスポリヌクレオチドの場合は、 本発明のタンパク質の N末端部位をコードする部分の塩基配列 (例えば、 開始コドン付近の塩基配列 など) の相補鎖と約 7 0 %以上、 好ましくは約 8 0 %以上、 より好ましくは約 9 0 %以上、 最も好ましくは約 9 5 %以上の相同性を有するアンチセンスポリ ヌクレオチドが、 (口) R N a s e Hによる R NA分解を指向するアンチセン スポリヌクレオチドの場合は、 イントロンを含む本発明の D N Aの全塩基配列 の相補鎖と約 7 0 %以上、 好ましくは約 8 0 %以上、 より好ましくは約' 9 0 % 以上、 最も好ましくは約 9 5 %以上の相同性を有するアンチセンスポリヌクレ ォチドがそれぞれ好適である。 The nucleotide sequence substantially complementary to the DNA of the present invention is, for example, the entire nucleotide sequence or a part of the nucleotide sequence complementary to the DNA of the present invention (that is, the complementary strand of the DNA of the present invention). A base sequence having about 70% or more, preferably about 80% or more, more preferably about 90% or more, and most preferably about 95% or more homology with the base sequence is exemplified. In particular, of the total nucleotide sequence of the complementary strand of the DNA of the present invention, (a) in the case of an antisense polynucleotide directed to translation inhibition, the nucleotide sequence of the portion encoding the N-terminal site of the protein of the present invention (for example, , A base sequence near the start codon, etc.) and about 70% or more, preferably about 80% or more, more preferably about 70% or more. When the antisense polynucleotide having a homology of 90% or more, most preferably about 95% or more is (mouth) an antisense polynucleotide which directs RNA degradation by RNase H, the present invention containing an intron About 70% or more, preferably about 80% or more, more preferably about 90% or more, and most preferably about 95% or more homology with the complementary strand of the entire base sequence of the DNA. Nucleotides are respectively preferred.
具体的には、 配列番号: 2、 配列番号: 4または配列番号: 6で表わされる 塩基配列を含有する D NAの塩基配列に相補的な、 もしくは実質的に相補的な 塩基配列、 またはその一部分を有するアンチセンスポリヌクレオチド、 好まし くは例えば、 配列番号: 2、 配列番号: 4または配列番号: 6で表わされる塩 基配列を含有する D N Aの塩基配列に相補な塩基配列、 またはその一部分を有 するアンチセンスポリヌクレオチド (より好ましくは、 配列番号: 2、 配列番 号: 4または配列番号: 6で表わされる塩基配列を含有する D N Aの塩基配列 に相補な塩基配列、 またはその一部分を有するアンチセンスポリヌクレオチ ド) などが挙げられる。  Specifically, a nucleotide sequence complementary to or substantially complementary to the nucleotide sequence of DNA containing the nucleotide sequence represented by SEQ ID NO: 2, SEQ ID NO: 4 or SEQ ID NO: 6, or a part thereof And preferably a nucleotide sequence complementary to the nucleotide sequence of the DNA containing the nucleotide sequence represented by SEQ ID NO: 2, SEQ ID NO: 4 or SEQ ID NO: 6, or a part thereof. Antisense polynucleotide having a nucleotide sequence (more preferably, a nucleotide sequence complementary to the nucleotide sequence of the DNA containing the nucleotide sequence represented by SEQ ID NO: 2, SEQ ID NO: 4 or SEQ ID NO: 6, or a part thereof) Sense polynucleotide) and the like.
アンチセンスポリヌクレオチドは通常、 1 0〜4 0個程度、 好ましくは 1 5 〜 3 0個程度の塩基から構成される。  An antisense polynucleotide is usually composed of about 10 to 40 bases, preferably about 15 to 30 bases.
ヌクレアーゼなどの加水分解酵素による分解を防ぐために、 アンチセンス D N Aを構成する各ヌクレオチドのりん酸残基 (ホスフェート) は、 例えば、 ホ スホロチォエート、 メチルホスホネート、 ホスホロジチォネートなどの化学修 飾りん酸残基に置換されていてもよい。 また、 各ヌクレオチドの糖 (デォキシ リポース) は、 2 ' —〇一メチル化などの化学修飾糖構造に置換されていても よいし、 塩基部分 (ピリミジン、 プリン) も化学修飾を受けたものであっても よく、 配列番号: 2で表わされる塩基配列を有する D NAにハイブリダィズす るものであればいずれのものでもよい。 これらのアンチセンスポリヌクレオチ ドは、 公知の D N A合成装置などを用いて製造することができる。  To prevent degradation by hydrolases such as nucleases, the phosphate residues (phosphates) of each nucleotide constituting the antisense DNA are, for example, chemically modified phosphate residues such as phosphorothioate, methylphosphonate, and phosphorodithionate. It may be substituted by a group. In addition, the sugar (deoxy lipose) of each nucleotide may be substituted with a chemically modified sugar structure such as 2′-dimethylation, and the base (pyrimidine, purine) may also be chemically modified. And any one that hybridizes to DNA having the base sequence represented by SEQ ID NO: 2. These antisense polynucleotides can be produced using a known DNA synthesizer or the like.
本発明に従えば、 本発明のタンパク質遺伝子の複製または発現を阻害するこ とのできる該遺伝子に対応するアンチセンスポリヌクレオチド (核酸) を、 ク ローン化した、 あるいは決定されたタンパク質をコードする D NAの塩基配列 情報に基づき設計し、 合成しうる。 かかるアンチセンスポリヌクレオチドは、 本発明のタンパク質遺伝子の R N Aとハイブリダイズすることができ、 該 R N Aの合成または機能を阻害することができるか、 あるいは本発明のタンパク質 関連 R NAとの相互作用を介して本発明のタンパク質遺伝子の発現を調節 ·制 御することができる。 本発明のタンパク質闋連 R N Aの選択された配列に相補 的なポリヌクレオチド、 および本発明のタンパク質関連 R N Aと特異的にハイ プリダイズすることができるポリヌクレオチドは、 生体内および生体外で本発 明のタンパク質遺伝子の発現を調節 ·制御するのに有用であり、 また病気など の治療または診断に有用である。 用語 「対応する」 とは、 遺伝子を含めたヌク レオチド、 塩基配列または核酸の特定の配列に相同性を有するあるいは相補的 であることを意味する。 ヌクレオチド、 塩基配列または核酸とタンパク質との 間で 「対応する」 とは、 ヌクレオチド (核酸) の配列またはその相補体から誘 導される (指令にある) タンパク質のアミノ酸を通常指している。 タンパク質 遺伝子の 5, 端ヘアピンループ、 5 ' 端 6—ベースペア ' リピート、 5, 端非 翻訳領域、 ポリペプチド翻訳開始コドン、 タンパク質コード領域、 O R F翻訳 終止コドン、 3 ' 端非翻訳領域、 3, 端パリンドローム領域または 3, 端ヘア ピンループなどは、 好ましい対象領域として選択しうるが、 タンパク質遺伝子 内の如何なる領域も対象として選択しうる。 According to the present invention, the antisense polynucleotide (nucleic acid) corresponding to the protein gene of the present invention, which can inhibit the replication or expression of the gene, is cloned or D-encoding the determined protein. NA base sequence Can be designed and synthesized based on information. Such antisense polynucleotides can hybridize to RNA of the protein gene of the present invention, inhibit the synthesis or function of the RNA, or interact with the protein-related RNA of the present invention through interaction with the RNA. Thus, the expression of the protein gene of the present invention can be regulated and controlled. Polynucleotides complementary to the selected sequence of the protein-linked RNA of the present invention, and polynucleotides capable of specifically hybridizing with the protein-related RNA of the present invention, can be used in vivo and in vitro. It is useful for regulating and controlling the expression of protein genes, and is also useful for treating or diagnosing diseases. The term "corresponding" means having homology or being complementary to a nucleotide, base sequence or a specific sequence of a nucleic acid including a gene. The "correspondence" between a nucleotide, nucleotide sequence or nucleic acid and a protein generally refers to the amino acid of the protein (as specified in the directive) derived from the nucleotide (nucleic acid) sequence or its complement. 5 'end hairpin loop of protein gene, 5' end 6—base pair 'repeat, 5, end untranslated region, polypeptide translation start codon, protein coding region, ORF translation stop codon, 3' end untranslated region, 3, The terminal palindrome region or the 3, terminal hairpin loop can be selected as a preferable target region, but any region in the protein gene can be selected as the target.
目的核酸と、 対象領域の少なくとも一部に相補的なポリヌクレオチドとの関 係については、 目的核酸が対象領域と八ィプリダイズすることができる場合 は、 その目的核酸は、 当該対象領域のポリヌクレオチドに対して 「アンチセン ス」 であるということができる。 アンチセンスポリヌクレオチドは、 2ーデォ キシー D—リポースを含有しているポリデォキシリボヌクレオチド、 D—リポ —スを含有しているポリリポヌクレオチド、 プリンまたはピリミジン塩基の N ーグリコシドであるその他のタイプのポリヌクレオチド、 非ヌクレオチド骨格 を有するその他のポリマー (例えば、 市販のタンパク質核酸および合成配列特 異的な核酸ポリマー) または特殊な結合を含有するその他のポリマ一 (但し、 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や塩基の 付着を許容する配置をもつヌクレオチドを含有する) などが挙げられる。 それ らは、 2本鎖 D NA、 1本鎖 D NA、 2本鎖 R NA、 1本鎖 R NA、 D NA: R NAハイブリッドであってもよく、 さらに非修飾ポリヌクレオチド (または 非修飾オリゴヌクレオチド) 、 公知の修飾の付加されたもの、 例えば当該分野 で知られた標識のあるもの、 キャップの付いたもの、 メチル化されたもの、 1 個以上の天然のヌクレオチドを類緣物で置換したもの、 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、 メチルホスホネ一卜、 ホスホト リエステル、 ホスホルアミデート、 力ルバメ一トなど) を持つもの、 電荷を有 する結合または硫黄含有結合 (例、 ホスホロチォエー卜、 ホスホロジチォエー 卜など) を持つもの、 例えばタンパク質 (例、 ヌクレアーゼ、 ヌクレア一ゼ' インヒビター、 トキシン、 抗体、 シグナルペプチド、 ポリ— L一リジンなど) や糖 (例、 モノサッカライドなど) などの側鎖基を有しているもの、 インタ一 カレント化合物 (例、 ァクリジン、 ソラレンなど) を持つもの、 キレート化合 物 (例えば、 金属、 放射活性をもつ金属、 ホウ素、 酸化性の金属など) を含有 するもの、 アルキル化剤を含有するもの、 修飾された結合を持つもの (例え ば、 αァノマー型の核酸など) であってもよい。 ここで 「ヌクレオシド」 、Regarding the relationship between the target nucleic acid and a polynucleotide complementary to at least a part of the target region, if the target nucleic acid can be hybridized with the target region, the target nucleic acid is included in the polynucleotide of the target region. On the other hand, it can be said that it is “antisense”. Antisense polynucleotides include polydexoxyribonucleotides containing 2-deoxy D-report, polyliponucleotides containing D-lipose, and other types of N-glycosides of purine or pyrimidine bases. Or other polymers having a non-nucleotide backbone (eg, commercially available protein nucleic acids and synthetic sequence-specific nucleic acid polymers) or other polymers containing special bonds (provided that the polymer is DNA or RNA) (Including nucleotides having a configuration that allows base pairing and base attachment as found in the above). It May be a double-stranded DNA, a single-stranded DNA, a double-stranded RNA, a single-stranded RNA, a DNA: RNA hybrid, and may further comprise an unmodified polynucleotide (or an unmodified oligonucleotide). ), Those with known modifications, such as those with a label, capped, methylated, and one or more natural nucleotides replaced with analogs known in the art Those having an intramolecular nucleotide modification, for example, those having an uncharged bond (eg, methylphosphonate, phosphotriester, phosphoramidate, calcium phosphate, etc.), a charged bond or a sulfur-containing bond ( (Eg, phosphorothioate, phosphorodithioate, etc.), such as proteins (eg, nucleases, nucleases' inhibitors, toxins, antibodies, signa) Peptides, poly-L-lysine, etc.) or sugars (eg, monosaccharides, etc.) with side-chain groups, inter-current compounds (eg, acridine, psoralen, etc.), chelating compounds (eg, For example, those containing metals, radioactive metals, boron, oxidizing metals, etc., those containing alkylating agents, those with modified bonds (eg, alpha-anomeric nucleic acids, etc.) There may be. Where "nucleoside",
「ヌクレオチド」 および 「核酸」 とは、 プリンおよびピリミジン塩基を含有す るのみでなく、 修飾されたその他の複素環型塩基をもつようなものを含んでい て良い。 このような修飾物は、 メチル化されたプリンおよびピリミジン、 ァシ ル化されたプリンおよびピリミジン、 あるいはその他の複素環を含むものであ つてよい。 修飾されたヌクレオチドおよび修飾されたヌクレオチドはまた糖部 分が修飾されていてよく、 例えば、 1個以上の水酸基がハロゲンとか、 脂肪族 基などで置換されていたり、 またはエーテル、 ァミンなどの官能基に変換され ていてよい。 “Nucleotides” and “nucleic acids” may include those containing not only purine and pyrimidine bases, but also other modified heterocyclic bases. Such modifications may include those containing 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.
本発明のアンチセンスポリヌクレオチドは、 RN A、 D NAまたは修飾され た核酸 (R NA、 D NA) である。 修飾された核酸の具体例としては、 核酸の 硫黄誘導体、 チォホスフェート誘導体、 ポリヌクレオシドアミドゃオリゴヌク レオシドアミドの分解に抵抗性のものなどが挙げられる。 本発明のアンチセン スポリヌクレオチドは、 例えば、 以下のように設計されうる。 すなわち、 細胞 内でのアンチセンスポリヌクレオチドをより安定なものにする、 アンチセンス ポリヌクレオチドの細胞透過性をより高める、 目標とするセンス鎖に対する親 和性をより大きなものにする、 また、 もし毒性があるような場合はアンチセン スポリヌクレオチドの毒性をより小さなものにする。 このような修飾は、 例え ば Pharm Tech Japan, 8卷, 247頁または 395頁, 1992年、 Ant i sense Research and Appl icat i ons, CRC Press, 1993年などで数多く報告されている。 The antisense polynucleotide of the present invention is RNA, DNA or a modified nucleic acid (RNA, DNA). Specific examples of the modified nucleic acid include sulfur derivatives of nucleic acids, thiophosphate derivatives, and polynucleoside amides that are resistant to degradation of oligonucleoside amides. The antisense polynucleotide of the present invention can be designed, for example, as follows. That is, an antisense polynucleotide that makes an antisense polynucleotide more stable in a cell. Increase the cell permeability of the polynucleotide, increase the affinity for the sense strand of interest, and, if so, reduce the toxicity of the antisense polynucleotide. Many such modifications have been reported, for example, in Pharm Tech Japan, vol. 8, p. 247 or p. 395, 1992, Antisense Research and Appli- cations, CRC Press, 1993.
本発明のアンチセンスポリヌクレオチドは、 変化せしめられたり、 修飾され た糖、 塩基、 結合を含有していて良く、 リボゾーム、 ミクロスフエアのような 特殊な形態で供与されたり、 遺伝子治療により適用されたり、 付加された形態 で与えられることができうる。 こうして付加形態で用いられるものとしては、 リン酸基骨格の電荷を中和するように働くポリリジンのようなポリカチオン 体、 細胞膜との相互作用を高めたり、 核酸の取込みを増大せしめるような脂質 The antisense polynucleotide of the present invention may contain altered or modified sugars, bases, or bonds, and may be provided in a special form such as ribosome or microsphere, applied by gene therapy, It can be provided in an added form. Such additional forms include polycations, such as polylysine, which act to neutralize the charge on the phosphate backbone, and lipids, which enhance interaction with cell membranes and increase nucleic acid uptake.
(例、 ホスホリピド、 コレステロールなど) などの疎水性のものが挙げられ る。 付加するに好ましい脂質としては、 コレステロールやその誘導体 (例、 コ レステリルクロ口ホルメート、 コール酸など) が挙げられる。 こうしたもの は、 核酸の 3 ' 端または 5 ' 端に付着させることができ、 塩基、 糖、 分子内ヌ クレオシド結合を介して付着させることができうる。 その他の基としては、 核 酸の 3 ' 端または 5 ' 端に特異的に配置されたキャップ用の基で、 ェキソヌク レアーゼ、 R N a s eなどのヌクレア一ゼによる分解を阻止するためのものが 挙げられる。 こうしたキャップ用の基としては、 ポリエチレングリコ一ル、 テ トラエチレングリコ一ルなどのグリコールをはじめとした当該分野で知られた 水酸基の保護基が挙げられるが、 それに限定されるものではない。 (Eg, phospholipids, cholesterol, etc.). Preferred lipids for addition include cholesterol and its derivatives (eg, cholesteryl chromate formate, cholic acid, etc.). These can be attached to the 3 'or 5' end of the nucleic acid and can be attached via a base, sugar, or intramolecular nucleoside linkage. Other groups include capping groups specifically located at the 3 'or 5' end of the nucleic acid that prevent degradation by nucleases such as exonucleases and RNases. . Such capping groups include, but are not limited to, hydroxyl-protecting groups known in the art, including glycols such as polyethylene glycol and tetraethylene glycol.
アンチセンスポリヌクレオチドの阻害活性は、 本発明の形質転換体、 本発明 の生体内や生体外の遣伝子発現系、 または本発明の夕ンパク質の生体内や生体 外の翻訳系を用いて調べることができる。 以下に、 本発明のタンパク質もしくは部分ペプチドまたはその塩 (以下、 本 発明のタンパク質と略記する場合がある) 、 本発明のタンパク質または部分べ プチドをコ一ドするポリヌクレオチド (例、 D NA (以下、 本発明の D NAと 略記する場合がある) ) 、 本発明のタンパク質もしくは部分ペプチドまたはそ の塩に対する抗体 (以下、 本発明の抗体と略記する場合がある) 、 および本発 明の D NAのアンチセンスポリヌクレオチド (以下、 本発明のアンチセンスポ リヌクレオチドと略記する場合がある) の用途を説明する。 The inhibitory activity of the antisense polynucleotide can be measured using the transformant of the present invention, the in vivo or in vitro gene expression system of the present invention, or the protein in vivo or in vitro translation system of the present invention. You can find out. Hereinafter, a protein or partial peptide of the present invention or a salt thereof (hereinafter, sometimes abbreviated as the protein of the present invention), a polynucleotide encoding the protein or partial peptide of the present invention (eg, DNA (hereinafter, referred to as DNA) May be abbreviated as the DNA of the present invention))), the protein or partial peptide of the present invention or (Hereinafter sometimes abbreviated as the antibody of the present invention) and a DNA antisense polynucleotide of the present invention (hereinafter sometimes abbreviated as the antisense polynucleotide of the present invention). The application will be described.
本発明のタンパク質は、 Wi s tar Fat ty ラット (WFラット) 、 Zucker Fat tyラ ット (ZFラット) および自然発症高コレステロール血症ラット (SHCラット) の 腎組織で発現が上昇するので、 疾患マ一力一として利用することが出来る。 す なわち、 腎臓の機能障害の早期診断、 症状の重症度の判定、 疾患進行の予測の ためのマーカーとして有用である。 本発明のタンパク質の活性を調節 (好まし <は阻害) する化合物もしくはその塩、 本発明の抗体、 本発明のアンチセンス ポリヌクレオチドなどは、 例えば、 糖尿病性腎症などの腎疾患の予防,治療剤 として使用することができる。  The expression of the protein of the present invention is increased in the kidney tissues of Wistar Fatty rats (WF rats), Zucker Fatty rats (ZF rats) and spontaneously hypercholesterolemic rats (SHC rats). It can be used as one of the best. In other words, it is useful as a marker for early diagnosis of renal dysfunction, judgment of symptom severity, and prediction of disease progression. A compound or a salt thereof that regulates (preferably inhibits) the activity of the protein of the present invention, the antibody of the present invention, the antisense polynucleotide of the present invention, and the like, for example, for the prevention and treatment of renal diseases such as diabetic nephropathy It can be used as an agent.
( 1 ) 疾病に対する医薬候補化合物のスクリーニング (1) Screening of drug candidate compounds for diseases
本発明のタンパク質は、 Wi s tar Fat ty ラット (WFラット) 、 Zucker Fat tyラ ット (ZFラッ卜) および自然発症高コレステロール血症ラット (SHCラット) の 腎組織で発現が上昇するので、 本発明のタンパク質の活性を調節 (好ましくは 阻害) する化合物またはその塩は、 例えば、 腎疾患 (例、 糖尿病性腎症、 巣状 糸球体硬化症、 慢性糸球体腎炎、 微小変化型ネフローゼ症候群、 メサンギゥム 増殖性腎炎、 管内増殖性腎炎、 IgA腎症、 膜性腎症、 膜性増殖性腎炎、 半月体形 成性腎炎、 ループス腎炎、 アミロイド腎、 尿細管間質性腎炎、 急性尿細管壊 死、 急性腎不全、 腎硬化症、 腎性浮腫、 腎腫瘍、 腎移植後の慢性拒絶など) な どの予防 ·治療剤として使用することができる。 好ましくは、 糖尿病性腎症、 巣状糸球体硬化症などの予防 ·治療剤である。  Since the expression of the protein of the present invention is increased in kidney tissues of Wistar Fatty rats (WF rats), Zucker Fatty rats (ZF rats) and spontaneously hypercholesterolemic rats (SHC rats), Compounds or salts thereof that regulate (preferably inhibit) the activity of the protein of the present invention include, for example, renal diseases (eg, diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome, Mesangium Proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy, membranous proliferative nephritis, crescentic adult nephritis, lupus nephritis, amyloid kidney, tubular interstitial nephritis, acute tubular necrosis, It can be used as a preventive or therapeutic agent for acute renal failure, renal sclerosis, renal edema, renal tumor, chronic rejection after renal transplantation, etc. Preferred are prophylactic and therapeutic agents for diabetic nephropathy, focal glomerulosclerosis and the like.
したがって、 本発明のタンパク質は、 本発明のタンパク質の活性を調節 (阻 害または促進) する化合物またはその塩のスクリーニングのための試薬として 有用である。  Therefore, the protein of the present invention is useful as a reagent for screening a compound or a salt thereof that regulates (inhibits or promotes) the activity of the protein of the present invention.
すなわち、 本発明は、 本発明のタンパク質を用いることを特徴とする本発明 のタンパク質の活性 (例えば、 無機リン酸取り込み活性、 無機リン酸輸送活性 など) を調節 (促進または阻害) する化合物またはその塩のスクリーニング方 法を提供する。 That is, the present invention provides a compound which regulates (promotes or inhibits) the activity of the protein of the present invention (eg, an inorganic phosphate uptake activity, an inorganic phosphate transporting activity, etc.) characterized by using the protein of the present invention, or a compound thereof. How to screen salt Provide the law.
具体的には、 (i) 本発明のタンパク質を含有する細胞の無機リン酸取り込み 活性と、 (i i) 本発明のタンパク質を含有する細胞と試験化合物の混合物の無 機リン酸取り込み活性との比較をすることを特徴する本発明のタンパク質の活 性を調節 (促進または阻害) する化合物またはその塩のスクリーニング方法、 Specifically, a comparison between (i) the inorganic phosphate uptake activity of cells containing the protein of the present invention and (ii) the inorganic phosphate uptake activity of a mixture of cells containing the protein of the present invention and a test compound. A method for screening a compound or a salt thereof that regulates (promotes or inhibits) the activity of the protein of the present invention,
(ί ' ) 本発明のタンパク質を含有する合成脂質二重層における無機リン酸輸送 活性と、 (i i ' ) 本発明のタンパク質を含有する合成脂質二重層および試験化合 物の混合物の無機リン酸輸送活性の比較をすることを特徴する本発明のタンパ ク質の活性を調節 (促進または阻害) する化合物またはその塩のスクリーニン グ方法などが挙げられる。 (ί ′) Inorganic phosphate transport activity in the synthetic lipid bilayer containing the protein of the present invention, and (ii ′) Inorganic phosphate transport activity of the mixture of the synthetic lipid bilayer containing the protein of the present invention and the test compound And a method of screening for a compound or its salt that regulates (promotes or inhibits) the activity of the protein of the present invention, which is characterized by comparing the above.
上記スクリーニング方法においては、 例えば (0 と (i i) の場合において、 無機リン酸取り込み活性を公知の方法、 例えば、 Methods in Enzymo logy, 191 巻, 494- 505頁, 1990年などに記載の方法またはそれに準じる方法に従って測定 し、 比較する。  In the above screening method, for example, in the case of (0 and (ii), inorganic phosphate uptake activity is determined by a known method, for example, the method described in Methods in Enzymology, vol. 191, pp. 494-505, 1990, or Measure and compare according to an equivalent method.
具体的には、 ω本発明のタンパク質を発現しうる細胞に、 標識された無機リ ン酸塩を加えた場合と、 (i i) 試験化合物の存在下、 本発明のタンパク質を発 現しうる細胞に、 標識された無機リン酸塩を加えた場合の、 細胞内に取り込ま れた無機リン酸量をそれぞれ測定し、 本発明のタンパク質の活性を調節 (促進 または阻害) する化合物またはその塩をスクリーニングする。 本反応は適当な 緩衝液中で行い、 細胞に取り込まれなかった無機リン酸は、 測定前に洗浄除去 する。  Specifically, ω is obtained by adding a labeled inorganic phosphate to cells capable of expressing the protein of the present invention; and (ii) cells capable of expressing the protein of the present invention in the presence of a test compound. The amount of inorganic phosphate taken into cells when a labeled inorganic phosphate is added is measured, and a compound or a salt thereof that regulates (promotes or inhibits) the activity of the protein of the present invention is screened. . Perform this reaction in an appropriate buffer, and wash off inorganic phosphate that has not been taken up by cells before measurement.
標識された無機リン酸塩としては、 放射性同位元素 (例、 〔32P〕 、 〔33P〕 な ど) などで標識された無機リン酸塩などが挙げられる。 Examples of the labeled inorganic phosphate include an inorganic phosphate labeled with a radioisotope (eg, [ 32 P], [ 33 P], etc.).
放射性同位元素で標識された無機リン酸塩を用いた場合、 細胞の無機リン酸 塩取り込み活性は、 放射活性量を公知の方法、 例えばに放射活性測定装置など を使用して測定する。  When an inorganic phosphate labeled with a radioisotope is used, the activity of the cells to take up the inorganic phosphate is measured by a known method for measuring the amount of radioactivity, for example, using a radioactivity measuring device.
標識された無機リン酸塩は、 試験化合物と混合した後に本発明のタンパク質 を発現する細胞と反応させてもよく、 また、 本発明のタンパク質を発現する細 胞に標識された無機リン酸塩を接触させた後、 試験化合物を添加してもよい。 上記の本発明の夕ンパク質を発現しうる細胞は、 本発明のタンパク質をコー ドする遺伝子を適当な細胞、 例えば、 COS- 7細胞、 CH0細胞、 HEK293細胞、 ァフリ カツメガエル卵母細胞などの動物細胞などに導入し、 培養することにより、 本 発明のタンパク質を細胞膜上に発現させる。 本発明のタンパク質を発現し得る 細胞の培養方法は、 前記した本発明の形質変換体の培養法と同様である。 The labeled inorganic phosphate may be reacted with cells expressing the protein of the present invention after being mixed with the test compound, and the labeled inorganic phosphate may be added to cells expressing the protein of the present invention. After contacting, the test compound may be added. The cells capable of expressing the protein of the present invention described above include cells encoding a gene encoding the protein of the present invention in appropriate cells, for example, COS-7 cells, CH0 cells, HEK293 cells, African Xenopus oocytes, and the like. The protein of the present invention is expressed on a cell membrane by being introduced into an animal cell or the like and cultured. The method for culturing cells capable of expressing the protein of the present invention is the same as the above-described method for culturing the transformant of the present invention.
また、 上記スクリーニング方法においては、 例えば (ΐ ' ) と (i f ) の場合に おいて、 無機リン酸輸送活性を公知の方法、 例えば、 Biochimica et  In the above screening method, for example, in the case of (ΐ ′) and (if), the inorganic phosphate transport activity can be determined by a known method, for example, Biochimica et al.
Biophys ica Acta, 1236巻, 339- 344頁, 1995年などに記載の方法またはそれに 準じる方法に従って測定し、 比較する。 Measure and compare according to the method described in Biophysica Acta, Vol. 1236, pp. 339-344, 1995, or a method analogous thereto.
具体的には、 (i ' )本発明のタンパク質を含有する合成脂質二重層に、 無機リ ン酸塩を含む緩衝液を接触させた場合と、 (i i) 試験化合物の存在下、 本発明 のタンパク質を含有する合成脂質二重層に、 無機リン酸塩を含む緩衝液を接触 させた場合の、 無機リン酸輸送活性をそれぞれ測定し、 本発明のタンパク質の 活性を調節 (阻害または促進) する化合物またはその塩をスクリ一ニングす る。  Specifically, (i ′) the case where a buffer solution containing an inorganic phosphate is brought into contact with the synthetic lipid bilayer containing the protein of the present invention; and (ii) the case where the present invention is used in the presence of a test compound. A compound that regulates (inhibits or promotes) the activity of the protein of the present invention by measuring the inorganic phosphate transport activity when a buffer solution containing an inorganic phosphate is brought into contact with a protein-containing synthetic lipid bilayer. Or screen the salt.
無機リン酸輸送活性の測定は、 無機リン酸が本発明のタンパク質によって輸 送された際に生じる膜電流の変化を、 公知の方法、 例えば膜電位固定法などを 使用して測定する。  In the measurement of the inorganic phosphate transport activity, a change in the membrane current that occurs when inorganic phosphate is transported by the protein of the present invention is measured using a known method, for example, a membrane potential fixing method.
本発明のタンパク質を含有する合成脂質二重層は、 例えば、 本発明のタンパ ク質を発現させた細胞の膜画分から、 公知の膜タンパク質精製方法に従って得 られた精製タンパク質を用い、 リン脂質などを材料にして、 公知の方法、 例え ば 「パッチクランプ実験技術法」 (吉岡書店、 1996年発行) 等に記載された方 法に従って製造できる。  The synthetic lipid bilayer containing the protein of the present invention can be prepared by, for example, using a purified protein obtained from a membrane fraction of a cell expressing the protein of the present invention according to a known membrane protein purification method, using a phospholipid or the like. The material can be produced according to a known method, for example, a method described in "Patch Clamp Experimental Technique" (Yoshioka Shoten, published in 1996).
無機リン酸塩は、 試験化合物と混合した後に本発明のタンパク質を含有する 脂質二重膜層と反応させてもよく、 また、 本発明のタンパク質を含有する脂質 二重膜層に無機リン酸イオンを接触させた後、 試験化合物を添加してもよい。 試験化合物としては、 例えばペプチド、 タンパク質、 非ペプチド性化合物、 合成化合物、 発酵生産物、 細胞抽出液、 植物抽出液、 動物組織抽出液などがあ げられる。 例えば、 上記 (i i) の場合における無機リン酸取り込み活性が上記 (i) の場 合に比べて、 約 20%以上、 好ましくは 30%以上、 より好ましくは約 50%以上上 昇させる試験化合物を、 本発明のタンパク質の活性を促進する化合物として、 上記 (i i) の場合における無機リン酸取り込み活性が上記 (i) の場合に比べ て、 約 20%以上、 好ましくは 30%以上、 より好ましくは約 50%以上減少させる 試験化合物を本発明のタンパク質の活性を阻害する化合物として選択すること ができる。 The inorganic phosphate may be reacted with the lipid bilayer containing the protein of the present invention after being mixed with the test compound, and the inorganic phosphate ion may be added to the lipid bilayer containing the protein of the present invention. After bringing into contact, the test compound may be added. Test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and the like. For example, a test compound that increases the inorganic phosphate uptake activity in the case of the above (ii) by about 20% or more, preferably 30% or more, more preferably about 50% or more compared to the case of the above (i) As the compound that promotes the activity of the protein of the present invention, the inorganic phosphate uptake activity in the case of the above (ii) is about 20% or more, preferably 30% or more, more preferably A test compound that reduces about 50% or more can be selected as a compound that inhibits the activity of the protein of the present invention.
例えば、 上記 (i i ' ) の場合における無機リン酸輸送活性が上記 (Γ ) の場合 に比べて、 約 20%以上、 好ましくは 30%以上、 より好ましくは約 50 %以上上昇 させる試験化合物を、 本発明のタンパク質の活性を促進する化合物として、 上 記 (i i ' ) の場合における無機リン酸輸送活性が上記 (Γ ) の場合に比べて、 約 20%以上、 好ましくは 30%以上、 より好ましくは約 50%以上減少させる試験ィ匕 合物を本発明のタンパク質の活性を阻害する化合物として選択することができ る。  For example, a test compound that increases the inorganic phosphate transport activity in the case of the above (ii ′) by about 20% or more, preferably 30% or more, more preferably about 50% or more as compared with the case of the above (II), As the compound that promotes the activity of the protein of the present invention, the inorganic phosphate transport activity in the case of the above (ii ′) is about 20% or more, preferably 30% or more, more preferably as compared with the case of the above (Γ). Can be selected as compounds that inhibit the activity of the protein of the present invention.
本発明のタンパク質の活性を促進する活性を有する化合物は、 本発明のタン パク質の生理活性を増強するための安全で低毒性な医薬として有用である。 本発明のタンパク質の活性を阻害する活性を有する化合物は、 本発明のタン パク質の生理活性を抑制するための安全で低毒性な医薬、 例えば、 腎疾患  The compound having the activity of promoting the activity of the protein of the present invention is useful as a safe and low-toxic drug for enhancing the physiological activity of the protein of the present invention. The compound having an activity of inhibiting the activity of the protein of the present invention is a safe and low toxic drug for suppressing the physiological activity of the protein of the present invention, for example, renal disease
(例、 糖尿病性腎症、 巣状糸球体硬化症、 慢性糸球体腎炎、 微小変化型ネフ口 —ゼ症候群、 メサンギゥム増殖性腎炎、 管内増殖性腎炎、 IgA腎症、 膜性腎症、 膜性増殖性腎炎、 半月体形成性腎炎、 ループス腎炎、 アミロイド腎、 尿細管間 質性腎炎、 急性尿細管壊死、 急性腎不全、 腎硬化症、 腎性浮腫、 腎腫瘍、 腎移 植後の慢性拒絶など) などの予防 ·治療剤として有用である。 好ましくは、 糖 尿病性腎症、 巣状糸球体硬化症などの予防 ·治療剤である。  (E.g., diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimally altered Nef osteomyelitis, mesangial proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy, membranous Proliferative nephritis, crescentic nephritis, lupus nephritis, amyloid kidney, tubulointerstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, renal tumor, chronic rejection after kidney transplantation It is useful as a preventive and therapeutic agent. Preferably, it is a prophylactic / therapeutic agent for glycemic nephropathy, focal glomerulosclerosis and the like.
本発明のスクリーニング方法またはスクリーニング用キットを用いて得られ る化合物またはその塩は、 例えば、 ペプチド、 タンパク、 非ペプチド性化合 物、 合成化合物、 発酵生産物、 細胞抽出液、 植物抽出液、 動物組織抽出液、 血 漿などから選ばれた化合物である。 該化合物の塩としては、 前記した本発明の ペプチドの塩と同様のものが用いられる。 さらに、 本発明のタンパク質をコードする遺伝子も、 Wis tar Fat tyラット (WFラット) 、 Zucker Fat tyラット (ZFラット) および自然発症高コレステロ 一ル血症ラット (SHCラット) の腎組織で発現が上昇するので、 本発明のタンパ ク質をコードする遺伝子の発現を調節 (好ましくは阻害) する化合物またはそ の塩は、 例えば、 腎疾患 (例、 糖尿病性腎症、 巣状糸球体硬化症、 慢性糸球体 腎炎、 微小変化型ネフローゼ症候群、 メサンギゥム増殖性腎炎、 管内増殖性腎 炎、 IgA腎症、 膜性腎症、 膜性増殖性腎炎、 半月体形成性腎炎、 ループス腎炎、 アミロイド腎、 尿細管間質性腎炎、 急性尿細管壊死、 急性腎不全、 腎硬化症、 腎性浮腫、 腎腫瘍、 腎移植後の慢性拒絶など) などの予防 ·治療剤として使用 することができる。 Compounds or salts thereof obtained using the screening method or the screening kit of the present invention include, for example, peptides, proteins, non-peptide compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissues It is a compound selected from extracts, plasma, etc. As the salt of the compound, those similar to the aforementioned salts of the peptide of the present invention are used. Furthermore, the gene encoding the protein of the present invention is also expressed in kidney tissues of Wistar Fatty rats (WF rats), Zucker Fatty rats (ZF rats) and spontaneously hypercholesterolemia (SHC rats). Compounds that modulate (preferably inhibit) the expression of the gene encoding the protein of the present invention because they are elevated include, for example, renal diseases (eg, diabetic nephropathy, focal glomerulosclerosis, Chronic glomerulonephritis, minimal change nephrotic syndrome, mesangial proliferative nephritis, intraluminal proliferative nephritis, IgA nephropathy, membranous nephropathy, membranous proliferative nephritis, crescentic nephritis, lupus nephritis, amyloid kidney, urine It can be used as a prophylactic / therapeutic agent for tubulointerstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, renal tumors, chronic rejection after renal transplantation, etc.).
したがって、 本発明のポリヌクレオチド (例、 D NA) は、 本発明のタンパ ク質をコ一ドする遣伝子の発現を調節 (好ましくは阻害) する化合物またはそ の塩のスクリーニングのための試薬として有用である。  Accordingly, the polynucleotide (eg, DNA) of the present invention can be used as a reagent for screening a compound or a salt thereof that regulates (preferably, inhibits) the expression of a gene encoding the protein of the present invention. Useful as
スクリーニング方法としては、 (i i i) 本発明のタンパク質を産生する能力を 有する細胞を培養した場合と、 (iv) 試験化合物の存在下、 本発明で用いられ るタンパク質を産生する能力を有する細胞を培養した場合との比較を行うこと を特徴とするスクリーニング方法が挙げられる。  The screening methods include (iii) culturing cells capable of producing the protein of the present invention, and (iv) culturing cells capable of producing the protein used in the present invention in the presence of a test compound. A screening method characterized by performing a comparison with the case where the method is performed.
上記方法において、 (i i i) と (iv) の場合における、 前記遺伝子の発現量 (具体的には、 本発明のタンパク質量または前記タンパク質をコードする mRNA 量) を測定して、 比較する。  In the above method, the expression level of the gene (specifically, the amount of the protein of the present invention or the amount of the mRNA encoding the protein) in the cases (iii) and (iv) is measured and compared.
試験化合物および本発明のタンパク質を産生する能力を有する細胞として は、 上記と同様のものが挙げられる。  Examples of the test compound and cells having the ability to produce the protein of the present invention include the same cells as described above.
タンパク質量の測定は、 公知の方法、 例えば、 本発明のタンパク質を認識す る抗体を用いて、 細胞抽出液中などに存在する前記タンパク質を、 ウエスタン 解析、 ELISA法などの方法またはそれに準じる方法に従い測定することができ る。  The amount of the protein is measured by a known method, for example, using an antibody recognizing the protein of the present invention, and analyzing the protein present in a cell extract or the like according to a method such as Western analysis, ELISA, or a method analogous thereto. Can be measured.
mRNA量の測定は、 公知の方法、 例えば、 プローブとして配列番号: 2、 配列 蕃号: 4もしくは配列番号: 6またはその一部分を含有する核酸を用いるノー ザンハイブリダィゼーシヨンまたは PCR法またはそれに準じる方法に従い測定す ることができる。 The amount of mRNA can be measured by a known method, for example, Northern hybridization or PCR using a nucleic acid containing SEQ ID NO: 2, SEQ ID NO: 4 or SEQ ID NO: 6 or a part thereof as a probe, or PCR. Measure according to the method Can be
例えば、 上記 (iv) の場合における遺伝子発現量を、 上記 (i i i) の場合に比 ベて、 約 20%以上、 好ましくは 30%以上、 より好ましくは約 50%以上上昇させ る試験化合物を、 本発明のタンパク質をコードする遺伝子の発現を促進する化 合物として、 約 20%以上、 好ましくは 30%以上、 より好ましくは約 50%以上阻 害する試験化合物を、 本発明のタンパク質をコ一ドする遺伝子の発現を抑制す る化合物として選択することができる。  For example, a test compound that increases the gene expression level in the case of the above (iv) by about 20% or more, preferably 30% or more, more preferably about 50% or more compared to the case of the above (iii), As a compound that promotes the expression of the gene encoding the protein of the present invention, a test compound that inhibits about 20% or more, preferably 30% or more, more preferably about 50% or more, is obtained by coding the protein of the present invention. Can be selected as a compound that suppresses the expression of a gene that causes the expression.
本発明のスクリーニング用キットは、 本発明で用いられるタンパク質もしく は部分ペプチドまたはその塩、 または本発明で用いられるタンパク質もしくは 部分べプチドを産生する能力を有する細胞を含有するものである。  The screening kit of the present invention contains the protein or partial peptide used in the present invention or a salt thereof, or a cell capable of producing the protein or partial peptide used in the present invention.
本発明のスクリーニング方法またはスクリーニング用キットを用いて得られ る化合物またはその塩は、 上記した試験化合物、 例えば、 ペプチド、 タンパ ク、 非ペプチド性化合物、 合成化合物、 発酵生産物、 細胞抽出液、 植物抽出 液、 動物組織抽出液、 血漿などから選ばれた化合物またはその塩である。 該化 合物の塩としては、 前記本発明のタンパク質の塩と同様のものが用いられる。 本発明のタンパク質の活性を調節 (好ましくは阻害) する化合物またはその 塩、 および本発明のタンパク質をコードする遺伝子の発現を調節 (好ましくは 阻害) する化合物またはその塩はそれぞれ、 例えば、 腎疾患 (例、 糖尿病性腎 症、 巣状糸球体硬化症、 慢性糸球体腎炎、 微小変化型ネフローゼ症候群、 メサ ンギゥム増殖性腎炎、 管内増殖性腎炎、 IgA腎症、 膜性腎症、 膜性増殖性腎炎、 半月体形成性腎炎、 ループス腎炎、 アミロイド腎、 尿細管間質性腎炎、 急性尿 細管壊死、 急性腎不全、 腎硬化症、 腎性浮腫、 腎腫瘍、 腎移植後の慢性拒絶な ど) などの予防,治療剤として有用である。  Compounds or salts thereof obtained using the screening method or screening kit of the present invention include the test compounds described above, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, and plants. It is a compound selected from an extract, an animal tissue extract, and plasma, or a salt thereof. As the salt of the compound, those similar to the aforementioned salts of the protein of the present invention are used. A compound or a salt thereof that regulates (preferably inhibits) the activity of the protein of the present invention and a compound or a salt thereof that regulates (preferably inhibits) the expression of a gene encoding the protein of the present invention may be, for example, a renal disease ( E.g., diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome, mesangial proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy, membranous proliferative nephritis , Crescent-forming nephritis, lupus nephritis, amyloid kidney, tubular interstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, renal tumors, chronic rejection after renal transplantation, etc.) It is useful as a prophylactic and therapeutic agent.
本発明のスクリーニング方法またはスクリーニング用キットを用いて得られ る化合物またはその塩を上述の予防 ·治療剤として使用する場合、 常套手段に 従って製剤化することができる。 '  When a compound or a salt thereof obtained by using the screening method or the screening kit of the present invention is used as the above-mentioned prophylactic / therapeutic agent, it can be formulated into a preparation according to a conventional method. '
例えば、 経口投与のための組成物としては、 固体または液体の剤形、 具体的 - には錠剤 (糖衣錠、 フィルムコーティング錠を含む) 、 丸剤、 顆粒剤、 散剤、 カプセル剤 (ソフトカプセル剤を含む) 、 シロップ剤、 乳剤、 懸濁剤などがあ げられる。 かかる組成物は自体公知の方法によって製造され、 製剤分野におい て通常用いられる担体、 希釈剤もしくは賦形剤を含有するものである。 例え ば、 錠剤用の担体、 賦形剤としては、 乳糖、 でんぷん、 蔗糖、 ステアリン酸マ グネシゥムなどが用いられる。 For example, compositions for oral administration include solid or liquid dosage forms, specifically tablets (including dragees and film-coated tablets), pills, granules, powders, capsules (including soft capsules). ), Syrups, emulsions, suspensions, etc. I can do it. Such a composition is produced by a method known per se and contains a carrier, diluent or excipient commonly used in the field of pharmaceuticals. For example, lactose, starch, sucrose, magnesium stearate and the like are used as carriers and excipients for tablets.
非経口投与のための組成物としては、 例えば、 注射剤、 坐剤などが用いら れ、 注射剤は静脈注射剤、 皮下注射剤、 皮内注射剤、 筋肉注射剤、 点滴注射 剤、 関節内注射剤などの剤形を包含する。 かかる注射剤は、 自体公知の方法に 従って、 例えば、 上記抗体またはその塩を通常注射剤に用いられる無菌の水性 もしくは油性液に溶解、 懸濁または乳化することによって調製する。 注射用の 水性液と'しては、 例えば、 生理食塩水、 プドウ糖やその他の補助薬を含む等張 液などが用いられ、 適当な溶解補助剤、 例えば、 アルコール (例、 エタノー ル) 、 ポリアルコール (例、 プロピレングリコール、 ポリエチレングリコ一 ル) 、 非イオン界面活性剤 〔例、, ポリソルベート 80、 HCO-50 (po lyoxye thy l ene (50iol) adduc t o f hydrogenated cas tor o i l) 〕 などと併用してもよい。 油 性液としては、 例えば、 ゴマ油、 大豆油などが用いられ、 溶解補助剤として安 息香酸ベンジル、 ベンジルアルコールなどを併用してもよい。 調製された注射 液は、 通常、 適当なアンプルに充填される。 直腸投与に用いられる坐剤は、 上 記化合物またはその塩を通常の坐薬用基剤に混合することによって調製され る。  As compositions for parenteral administration, for example, injections, suppositories, etc. are used. Injections are intravenous, subcutaneous, intradermal, intramuscular, intravenous, intraarticular. Includes dosage forms such as injections. Such injections are prepared according to a method known per se, for example, by dissolving, suspending or emulsifying the antibody or a salt thereof in a sterile aqueous or oily liquid commonly used for injections. Examples of the aqueous liquid for injection include physiological saline, isotonic solution containing pudose and other adjuvants, and a suitable solubilizing agent such as alcohol (eg, ethanol), Use in combination with polyalcohols (eg, propylene glycol, polyethylene glycol), nonionic surfactants (eg, polysorbate 80, HCO-50 (polyoxyethylen (50iol) adduc tof hydrogenated cas tor oil)), etc. You may. As the oily liquid, for example, sesame oil, soybean oil, and the like are used, and benzyl benzoate, benzyl alcohol, and the like may be used in combination as a solubilizing agent. The prepared injection solution is usually filled in a suitable ampoule. Suppositories used for rectal administration are prepared by mixing the above compound or a salt thereof with a usual suppository base.
上記の経口用または非経口用医薬組成物は、 活性成分の投与量に適合するよ うな投薬単位の剤形に調製されることが好都合である。 かかる投薬単位の剤形 としては、 錠剤、 丸剤、 カプセル剤、 注射剤 (アンプル) 、 坐剤などが例示さ れ、 それぞれの投薬単位剤形当たり通常 5〜500mg、 とりわけ注射剤では 5〜 100mg、 その他の剤形では 10〜250mgの上記化合物が含有されていることが好ま しい。  The above-mentioned oral or parenteral pharmaceutical composition is conveniently prepared in the form of a dosage unit so as to conform to the dose of the active ingredient. Examples of the dosage unit include tablets, pills, capsules, injections (ampoules), suppositories, etc., and usually 5 to 500 mg, especially 5 to 100 mg for each dosage unit. Preferably, other dosage forms contain 10 to 250 mg of the above compound.
なお前記した各組成物は、 上記化合物との配合により好ましくない相互作用 を生じない限り他の活性成分を含有してもよい。  Each of the above-mentioned compositions may contain other active ingredients as long as the compound and the above-mentioned compound do not cause an undesirable interaction.
このようにして得られる製剤は安全で低毒性であるので、 例えば、 ヒトまた は温血動物 (例えば、 マウス、 ラット、 ゥサギ、 ヒッジ、 ブ夕、 ゥシ、 ゥマ、 トリ、 ネコ、 ィヌ、 サル、 チンパンジーなど) に対して経口的にまたは非経口 的に投与することができる。 The preparations obtained in this way are safe and low toxic, for example, in humans or warm-blooded animals (eg, mice, rats, puppies, higgies, bush, puppies, puppies, To birds, cats, dogs, monkeys, chimpanzees, etc.) orally or parenterally.
該化合物またはその塩の投与量は、 その作用、 対象疾患、 投与対象、 投与ル ートなどにより差異はあるが、 例えば、 糖尿病性腎症の治療の目的で本発明の タンパク質の活性を調節 (好ましくは阻害) する化合物またはその塩を経口投 与する場合、 一般的に成人 (体重 60kgとして) においては、 一日につき該化合 物またはその塩を約 0. l〜100mg、 好ましくは約 1. 0〜50mg、 より好ましくは約 1. 0〜20mg投与する。 非経口的に投与する場合は、 該化合物またはその塩の 1回 投与量は投与対象、 対象疾患などによっても異なるが、 例えば、 糖尿病性腎症 の治療の目的で本発明のタンパク質の活性を調節 (好ましくは阻害) する化合 物またはその塩を注射剤の形で通常成人 (体重 60kgとして) に投与する場合、 一日につき該化合物またはその塩を約 0. 01〜30mg、 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。  The dose of the compound or a salt thereof varies depending on its action, target disease, subject of administration, route of administration, and the like. For example, the activity of the protein of the present invention is regulated for the purpose of treating diabetic nephropathy ( When a compound or a salt thereof is orally administered, the compound or a salt thereof is generally administered in an amount of about 0.1 to 100 mg, preferably about 1. 0 to 50 mg, more preferably about 1.0 to 20 mg is administered. When administered parenterally, the single dose of the compound or a salt thereof varies depending on the administration subject, target disease, and the like.For example, the activity of the protein of the present invention is regulated for the purpose of treating diabetic nephropathy. When a compound (or a salt thereof) which is preferably (inhibited) is administered to an adult (with a body weight of 60 kg) usually in the form of an injection, the compound or a salt thereof is used in an amount of about 0.01 to 30 mg, preferably about 0.1 to 30 mg per day. Conveniently, 1-20 mg, more preferably about 0.1-10 mg, will be administered by intravenous injection. In the case of other animals, the dose can be administered in terms of weight per 60 kg.
( 2 ) 本発明のタンパク質、 その部分べプチドまたはその塩の定量 (2) Quantification of the protein of the present invention, its partial peptide or its salt
本発明のタンパク質に対する抗体 (以下、 本発明の抗体と略記する場合があ る) は、 本発明のタンパク質を特異的に認識することができるので、 被検液中 の本発明のタンパク質の定量、 特にサンドイツチ免疫測定法による定量などに 使用することができる。  An antibody against the protein of the present invention (hereinafter sometimes abbreviated as the antibody of the present invention) can specifically recognize the protein of the present invention, so that the quantification of the protein of the present invention in a test solution can be performed. In particular, it can be used for quantification by the San Germanti immunoassay.
すなわち、 本発明は、  That is, the present invention
(i) 本発明の抗体と、 被検液および標識化された本発明のタンパク質とを競合 的に反応させ、 該抗体に結合した標識化された本発明のタンパク質の割合を測 定することを特徴とする被検液中の本発明のタンパク質の定量法、 および  (i) reacting the antibody of the present invention with a test solution and the labeled protein of the present invention competitively, and measuring the ratio of the labeled protein of the present invention bound to the antibody. A method for quantifying the protein of the present invention in a test solution, and
(ϋ) 被検波と担体上に不溶化した本発明の抗体および標識化された本発明の 別の抗体とを同時あるいは連続的に反応させたのち、 不溶化担体上の標識剤の 活性を測定することを特徴とする被検波中の本発明のタンパク質の定量法を提 供する。  (ii) Measuring the activity of the labeling agent on the insolubilized carrier after simultaneously or consecutively reacting the test wave with the antibody of the present invention insolubilized on the carrier and another labeled antibody of the present invention on the carrier. A method for quantifying the protein of the present invention in a test wave is provided.
上記 (i i) の定量法においては、 一方の抗体が本発明のタンパク質の N端部 を認識する抗体で、 他方の抗体が本発明のタンパク質の C端部に反応する抗体 であることが望ましい。 In the quantification method (ii), one of the antibodies is the N-terminal of the protein of the present invention. Preferably, the other antibody is an antibody that reacts with the C-terminal of the protein of the present invention.
また、 本発明のタンパク質に対するモノクローナル抗体 (以下、 本発明のモ ノクローナル抗体と称する場合がある) を用いて本発明のタンパク質の定量を 行なえるほか、 組織染色等による検出を行なうこともできる。 これらの目的に は、 抗体分子そのものを用いてもよく、 また、 抗体分子の F (ab' ) 2 、 Fab'ある いは Fab画分を用いてもよい。 In addition, the protein of the present invention can be quantified using a monoclonal antibody against the protein of the present invention (hereinafter sometimes referred to as the monoclonal antibody of the present invention), and can also be detected by tissue staining or the like. For these purposes, the antibody molecule itself may be used, or F (ab ') 2 , Fab' or Fab fraction of the antibody molecule may be used.
本発明の抗体を用いる本発明のタンパク質の定量法は、 特に制限されるべき ものではなく、 被測定液中の抗原量 (例えば、 タンパク質量) に対応した抗 体、 抗原もしくは抗体—抗原複合体の量を化学的または物理的手段により検出 し、 これを既知量の抗原を含む標準液を用いて作製した標準曲線より算出する 測定法であれば、 いずれの測定法を用いてもよい。 例えば、 ネフロメトリ一、 競合法 ィムノメトリック法およびサンドイッチ法が好適に用いられるが 感 度、 特異性の点で、 後述するサンドイッチ法を用いるのが特に好ましい。  The method for quantifying the protein of the present invention using the antibody of the present invention is not particularly limited, and may be an antibody, antigen or antibody-antigen complex corresponding to the amount of antigen (eg, the amount of protein) in the test solution. Any method can be used as long as the amount is determined 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 preferably used, but it is particularly preferable to use a sandwich method described later in terms of sensitivity and specificity.
標識物質を用いる測定法に用いられる標識剤としては、 例えば、 放射性同位 元素 (例、 〔125ι〕 、 〔131 ι〕 、 〔¾〕 、 TO 、 ™ 、 〔33P〕 、 〔35s〕 な ど) 、 蛍光物質 〔例、 シァニン蛍光色素 (例、 Cy2、 Cy3、 Cy5、 Cy5. 5、 Cy7 (ァ マシャムバイオサイエンス社製) など) 、 フルォレスカミン、 フルォレツセン イソチオシァネートなど〕 、 酵素 (例、 β— ラクトシダ一ゼ、 )3—ダルコシ ダーゼ、 アルカリフォスファターゼ、 パーォキシダーゼ、 リンゴ酸脱水素酵素 など) 、 発光物質 (例、 ルミノール、 ルミノール誘導体、 リレシフェリン、 ルシ ゲニンなど) 、 ラン夕二ド元素などが用いられる。 さらに、 抗体あるいは抗原 と標識剤との結合にピオチン一アビジン系を用いることもできる。 Examples of the labeling agent used in the assay method using the labeling substance, for example, a radioactive isotope (e.g., [125 iota], [131 iota], [¾], TO, ™, [33 P], a [35 s] ), Fluorescent substances (eg, cyanine fluorescent dyes (eg, Cy2, Cy3, Cy5, Cy5.5, Cy7 (manufactured by Amersham Bioscience)), fluorescamine, fluorescein isothiocynate, etc.), enzymes (examples) , Β-lactosidase,) 3-darcosidase, alkaline phosphatase, peroxidase, malate dehydrogenase, etc., luminescent substances (eg, luminol, luminol derivatives, reluciferin, lucigenin, etc.), element of laninide Are used. Further, a biotin-avidin system can be used for binding the antibody or antigen to the labeling agent.
抗原あるいは抗体の不溶化に当っては、 物理吸着を用いてもよく、 また通常 タンパク質あるいは酵素等を不溶化、 固定化するのに用いられる化学結合を用 いる方法でもよい。 担体としては、 ァガロース、 デキストラン、 セルロースな どの不溶性多糖類、 ポリスチレン、 ポリアクリルアミド、 シリコン等の合成樹 脂、 あるいはガラス等が挙げられる。  For the insolubilization of an antigen or an antibody, physical adsorption may be used, or a method using a chemical bond usually used for insolubilizing and immobilizing a protein or an enzyme may be used. Examples of the carrier include insoluble polysaccharides such as agarose, dextran, and cellulose; synthetic resins such as polystyrene, polyacrylamide, and silicon; and glass.
サンドイッチ法においては不溶化した本発明のモノクローナル抗体に被検液 を反応させ (1次反応) 、 さらに標識化した別の本発明のモノクローナル抗体 を反応させ (2次反応) たのち、 不溶化担体上の標識剤の活性を測定すること により被検波中の本発明の夕ンパク質量を定量することができる。 1次反応と 2次反応は逆の順序に行っても、 また、 同時に行なってもよいし時間をずらし て行なってもよい。 標識化剤および不溶化の方法は前記のそれらに準じること ができる。 また、 サンドイッチ法による免疫測定法において、 固相用抗体ある いは標識用抗体に用いられる抗体は必ずしも 1種類である必要はなく、 測定感 度を向上させる等の目的で 2種類以上の抗体の混合物を用いてもよい。 In the sandwich method, test solution was added to the insolubilized monoclonal antibody of the present invention. (Primary reaction), and further reacted with another labeled monoclonal antibody of the present invention (secondary reaction), and then the activity of the labeling agent on the insolubilized carrier is measured. Can be quantified. The primary reaction and the secondary reaction may be performed in the reverse order, may be performed simultaneously, or may be performed at staggered times. The labeling agent and the method of insolubilization can be the same as those described above. In addition, in the immunoassay by the sandwich method, the antibody used for the solid phase antibody or the labeling antibody does not necessarily need to be one kind, and two or more kinds of antibodies are used for the purpose of improving the measurement sensitivity and the like. Mixtures may be used.
本発明のサンドイッチ法による本発明のタンパク質の測定法においては、 1 次反応と 2次反応に用いられる本発明のモノクロ一ナル抗体は、 本発明のタン パク質の結合する部位が相異なる抗体が好ましく用いられる。 すなわち、 1次 反応および 2次反応に用いられる抗体は、 例えば、 2次反応で用いられる抗体' が、 本発明のタンパク質の C端部を認識する場合、 1次反応で用いられる抗体 は、 好ましくは C端部以外、 例えば N端部を認識する抗体が用いられる。  In the method for measuring the protein of the present invention by the sandwich method of the present invention, the monoclonal antibody of the present invention used in the primary reaction and the secondary reaction is an antibody having a different site to which the protein of the present invention binds. It is preferably used. That is, when the antibody used in the primary reaction and the secondary reaction is, for example, the antibody used in the secondary reaction recognizes the C-terminal of the protein of the present invention, the antibody used in the primary reaction is preferably Is an antibody that recognizes other than the C-terminal, for example, the N-terminal.
本発明のモノク口一ナル抗体をサンドィッチ法以外の測定システム、 例え ば、 競合法、 ィムノメトリック法あるいはネフロメトリ一などに用いることが できる。  The monoclonal antibody of the present invention can be used in a measurement system other than the sandwich method, for example, a competition method, an immunometric method, or a nephrometry method.
競合法では、 被検液中の抗原と標識抗原とを抗体に対して競合的に反応させ たのち、 未反応の標識抗原(F ) と、 抗体と結合した標識抗原 (B) とを分離し (BZ F分離) 、 B , Fいずれかの標識量を測定し、 被検液中の抗原量を定量 する。 本反応法には、 抗体として可溶性抗体を用い、 B/F分離をポリエチレ ングリコール、 前記抗体に対する第 2抗体などを用いる液相法、 および、 第 1 抗体として固相化抗体を用いるか、 あるいは、 第 1抗体は可溶性のものを用い 第 2抗体として固相化抗体を用いる固相化法とが用いられる。  In the competitive method, the antigen in the test solution and the labeled antigen are allowed to react competitively with the antibody, and then the unreacted labeled antigen (F) and the labeled antigen (B) bound to the antibody are separated. (BZF separation) The labeling amount of either B or F is measured, and the amount of antigen in the test solution is quantified. In this reaction method, a soluble phase is used as the antibody, B / F separation is carried out 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, or An immobilization method using a soluble antibody as the first antibody and an immobilized antibody as the second antibody is used.
ィムノメトリック法では、 被検波中の抗原と固相化抗原とを一定量の標識化 抗体に対して競合反応させた後固相と液相を分離する力、 あるいは、 被検液中 の抗原と過剰量の標識化抗体とを反応させ、 次に固相化抗原を加え未反応の標 識化抗体を固相に結合させたのち、 固相と液相を分離する。 次に、 いずれかの また、 ネフロメトリ一では、 ゲル内あるいは溶液中で抗原抗体反応の結果生 じた不溶性の沈降物の量を測定する。 被検液中の抗原量が僅かであり、 少量の 沈降物しか得られない場合にもレーザーの散乱を利用するレーザーネフロメト リーなどが好適に用いられる。 In the immunometric method, the ability to separate the solid phase from the liquid phase after a competitive reaction between the antigen in the test wave and the immobilized antigen for a certain amount of the labeled antibody, or the antigen in the test solution Is allowed to react with an excess amount of the labeled antibody, and then the immobilized antigen is added to bind the unreacted labeled antibody to the solid phase, and then the solid phase and the liquid phase are separated. Then either In nephrometry, 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 sediment is obtained, laser nephrometry utilizing laser scattering is preferably used.
' これら個々の免疫学的測定法を本発明の定量方法に適用するにあたっては、 特別の条件、 操作等の設定は必要とされない。 それぞれの方法における通常の 条件、 操作法に当業者の通常の技術的配慮を加えて本発明のタンパク質の測定 系を構築すればよい。 これらの一般的な技術手段の詳細については、 総説、 成 書などを参照することができる。 'No special conditions, procedures, etc. are required to apply these individual immunological assays to the quantification method of the present invention. The system for measuring the protein of the present invention may be constructed by adding ordinary technical considerations to those skilled in the art to the ordinary conditions and procedures in each method. For details of these general technical means, reference can be made to reviews and documents.
例えば、 入江 寛編 「ラジオィムノアツセィ」 (講談社、 昭和 4 9年発 行) 、 入江 寛編 「続ラジオィムノアツセィ」 (講談社、 昭和 5 4年発行) 、 石川栄治ら編 「酵素免疫測定法」 (医学書院、 昭和 5 3年発行) 、 石川栄治ら 編 「酵素免疫測定法」 (第 2版) (医学書院., 昭和 5 7年発行) 、 石川栄治ら 編 「酵素免疫測定法」 (第 3版) (医学書院、 昭和 6 2年発行) 、 「Methods in ENZYMOLOGYJ Vol . 70 (Immunochemical Techniques (Part A))、 同書 Vol . 73 (Immunochemical Techniques (Part B))、 同書 Vo l . 74 (Immunochemical Techniques (Part C) )、 同書 Vol. 84 (Immunochemical Techniques (Part D: Selec ted Immunoassays) ) 同書 Vol. 92 (Immunochemical Techniques (Part E : Monoc lonal Ant ibodies and General Immunoassay Methods))、 同書 Vol . 121 (Immunochemical Techniques (Part I :Hybridoma Technology and  For example, edited by Hiro Irie "Radio Nono Atsue" (Kodansha, published in 1949), edited by Hiro Irie "Radio Imno Atsue" (Kodansha, published in 1954), edited by Eiji Ishikawa et al. "Immunoassay" (Medical Shoin, published in 1975), edited by Eiji Ishikawa et al., "Enzyme Immunoassay" (2nd edition) (Medical Publishing, published in 1977), Eiji Ishikawa, edited by "Enzyme Immunoassay" Law (3rd edition) (Issue Shoin, published in 1962), "Methods in ENZYMOLOGYJ Vol. 70 (Immunochemical Techniques (Part A))", Vol. 73 (Immunochemical Techniques (Part B)), Vol. 74 (Immunochemical Techniques (Part C)), ibid.Vol. 84 (Immunochemical Techniques (Part D: Selected Immunoassays)) ibid.Vol. 92 (Immunochemical Techniques (Part E: Monoclonal Ant ibodies and General Immunoassay Methods)), ibid. Vol. 121 (Immunochemical Techniques (Part I: Hybridoma Technology and
Monoclonal Ant ibodi es) ) (以上、 アカデミックプレス社発行)などを参照するこ とができる。  Monoclonal Ant ibodi es)) (above, published by Academic Press).
以上のようにして、 本発明の抗体を用いることによって、 本発明のタンパク 質を感度良く定量することができる。  As described above, the protein of the present invention can be quantified with high sensitivity by using the antibody of the present invention.
さらには、 本発明の抗体を用いて本発明のタンパク質の濃度を定量すること によって、 本発明のタンパク質の濃度の増加または減少が検出された場合、 例 えば、 腎疾患 (例、 糖尿病性腎症、 巣状糸球体硬化症、 慢性糸球体腎炎、 微小 変化型ネフローゼ症候群、 メサンギゥム増殖性腎炎、 管内増殖性腎炎、 IgA腎 症、 膜性腎症、 膜性増殖性腎炎、 半月体形成性腎炎、 ループス腎炎、 アミロイ ド腎、 尿細管間質性腎炎、 急性尿細管壊死、 急性腎不全、 腎硬化症、 腎性浮 腫、 腎腫瘍、 腎移植後の慢性拒絶など) などである、 または将来罹患する可能 性が高いと診断することができる。 Furthermore, when an increase or decrease in the concentration of the protein of the present invention is detected by quantifying the concentration of the protein of the present invention using the antibody of the present invention, for example, renal disease (eg, diabetic nephropathy) , Focal glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome, mesangial proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy, membranous proliferative nephritis, crescent-forming nephritis, Lupus nephritis, amyloid Renal disease, tubulointerstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, renal tumors, chronic rejection after renal transplantation, etc., or may be affected in the future It can be diagnosed as high.
また、 本発明の抗体は、 体液や組織などの被検体中に存在する本発明のタン パク質を検出するために使用することができる。 また、 本発明のタンパク質を 精製するために使用する抗体カラムの作製、 精製時の各分画中の本発明のタン パク質の検出、 被検細胞内における本発明のタンパク質の挙動の分析などのた めに使用することができる。 ( 3 ) 遺伝子診断薬  Further, the antibody of the present invention can be used for detecting the protein of the present invention present in a subject such as a body fluid or a tissue. In addition, preparation of an antibody column used for purifying the protein of the present invention, detection of the protein of the present invention in each fraction at the time of purification, analysis of the behavior of the protein of the present invention in test cells, etc. Can be used for (3) Gene diagnostics
本発明の D N Aは、 例えば、 プローブとして使用することにより、 ヒトまた は温血動物 (例えば、 ラット、 マウス、 モルモット、 ゥサギ、 トリ、 ヒッジ、 ブタ、 ゥシ、 ゥマ、 ネコ、 ィヌ、 サル、 チンパンジーなど) における本発明の タンパク質またはその部分ペプチドをコードする D N Aまたは m R N Aの異常 (遣伝子異常) を検出することができるので、 例えば、 該 D NAまたは mR N The DNA of the present invention can be used, for example, in humans or warm-blooded animals (eg, rats, mice, guinea pigs, egrets, birds, higgies, pigs, pigs, dogs, cats, dogs, monkeys) by using them as probes. , Chimpanzees, etc.) can detect abnormalities (gene abnormalities) in DNA or mRNA encoding the protein of the present invention or a partial peptide thereof.
Aの損傷、 突然変異あるいは発現低下や、 該 D NAまたは mR NAの増加ある いは発現過多などの遺伝子診断薬として有用である。 It is useful as a gene diagnostic agent for A damage, mutation or decreased expression, and increase or excessive expression of the DNA or mRNA.
本発明の D N Aを用いる上記の遗伝子診断は、 例えば、 自体公知のノーザン ハイブリダィゼ一シヨン、 PCR-SSCP法 (Genomi cs, 第 5巻, 874〜879頁, 1989 年、 Proceedings of the Nat i onal Academy of Sc i ences of the Uni ted The above-described gene diagnosis using the DNA of the present invention can be performed, for example, by the known Northern hybridization, PCR-SSCP method (Genomics, Vol. 5, pp. 874-879, 1989, Proceedings of the National Academy of Sciences of the United
States o f Amer i ca, 第 86巻, 2766〜2770頁, 1989年) などにより実施すること ができる。 States of America, Vol. 86, pp. 2766-2770, 1989).
例えば、 ノーザンハイブリダイゼ一シヨンにより発現過多または減少が検出 された場合、 PCR- SSCP法により DNAの突然変異が検出された場合は、 例えば、 腎 疾患 (例、 糖尿病性腎症、 巣状糸球体硬化症、 慢性糸球体腎炎、 微小変化型ネ フローゼ症候群、 メサンギゥム増殖性腎炎、 管内増殖性腎炎、 IgA腎症、 膜性腎 症、 膜性増殖性腎炎、 半月体形成性腎炎、 ループス腎炎、 アミロイド腎、 尿細 管間質性腎炎、 急性尿細管壊死、 急性腎不全、 腎硬化症、 腎性浮腫、 腎腫瘍、 腎移植後の慢性拒絶など) などである可能性が高いと診断することができる。 ( 4 ) アンチセンスポリヌクレオチドを含有する医薬 For example, when overexpression or decrease is detected by Northern hybridization, or when DNA mutation is detected by PCR-SSCP method, for example, renal disease (eg, diabetic nephropathy, focal thread) Glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome, mesangial proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy, membranous proliferative nephritis, crescentic nephritis, lupus nephritis, Diagnosed as having a high likelihood of amyloid kidney, tubular interstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, renal tumor, chronic rejection after kidney transplantation, etc. Can be. (4) a drug containing an antisense polynucleotide
本発明の D N Aに相補的に結合し、 該 D N Aの発現を抑制することができる 本発明のアンチセンスポリヌクレオチドは低毒性であり、 生体内における本発 明のタンパク質または本発明の D NAの機能を抑制することができるので、 例 えば、 腎疾患 (例、 糖尿病性腎症、 巣状糸球体硬化症、 慢性糸球体腎炎、 微小 変化型ネフローゼ症候群、 メサンギゥム増殖性腎炎、 管内増殖性腎炎、 IgA腎 症、 膜性腎症、 膜性増殖性腎炎、 半月体形成性腎炎、 ループス腎炎、 アミロイ ド腎、 尿細管間質性腎炎、 急性尿細管壊死、 急性腎不全、 腎硬化症、 腎性浮 齓 腎腫瘍、 腎移植後の慢性拒絶など) などの予防 ·治療剤として使用するこ とができる。  The antisense polynucleotide of the present invention, which can complementarily bind to the DNA of the present invention and suppress the expression of the DNA, has low toxicity, and functions of the protein of the present invention or the DNA of the present invention in vivo. For example, renal diseases (eg, diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome, mesangial proliferative nephritis, endoproliferative nephritis, IgA Nephropathy, membranous nephropathy, membranous proliferative nephritis, crescentic nephritis, lupus nephritis, amyloid kidney, tubulointerstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema齓 Can be used as a prophylactic or therapeutic agent for kidney tumors, chronic rejection after kidney transplantation, etc.).
上記ァンチセンスポリヌクレオチドを上記の予防 ·治療剤として使用する場 合、 自体公知の方法に従って製剤化し、 投与することができる。  When the antisense polynucleotide is used as the above-mentioned prophylactic / therapeutic agent, it can be formulated and administered according to a method known per se.
また、 例えば、 前記のアンチセンスポリヌクレオチドを単独あるいはレトロ ウィルスベクター、 アデノウイルスベクター、 アデノウイルスァソシエーテツ ドウィルスベクターなどの適当なベクターに揷入した後、 常套手段に従って、 ヒトまたは哺乳動物 (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。 該ァ ンチセンスポリヌクレオチドは、 そのままで、 あるいは摂取促進のために補助 剤などの生理学的に認められる担体とともに製剤化し、 遺伝子銃やハイドロゲ ルカテ一テルのようなカテーテルによって投与できる。 あるいは、 エアロゾル 化して吸入剤として気管内に局所投与することもできる。  Further, for example, the above-mentioned antisense polynucleotide is used alone or in a suitable vector such as a retrovirus vector, an adenovirus vector, an adenovirus associated virus vector, and the like, and thereafter, human or mammal ( For example, it can be administered orally or parenterally to rats, puppies, sheep, sheep, bush, puppies, cats, dogs, monkeys, etc.). The antisense polynucleotide can be administered as it is or in the form of a formulation together with a physiologically acceptable carrier such as an auxiliary agent for promoting uptake, and can be administered by a gene gun or a catheter such as a hydrogel catheter. Alternatively, they can be aerosolized and administered topically into the trachea as an inhalant.
さらに、 体内動態の改良、 半減期の長期化、 細胞内取り込み効率の改善を目 的に、 前記のアンチセンスポリヌクレオチドを単独またはリボゾームなどの担 体とともに製剤 (注射剤) 化し、 静脈、 皮下、 病変部等に投与してもよい。 該アンチセンスポリヌクレオチドの投与量は、 対象疾患、 投与対象、 投与ル ートなどにより差異はあるが、 例えば、 糖尿病性腎症の治療の目的で本発明の アンチセンスポリヌクレオチドを投与する場合、 一般的に成人 (体重 60kg) に おいては、 一日につき該アンチセンスポリヌクレオチドを約 0. l〜100mg投与す る。 Furthermore, in order to improve pharmacokinetics, prolong the half-life, and improve the efficiency of intracellular uptake, the above antisense polynucleotide is formulated alone or together with a carrier such as ribosome (injection), and is intravenously, subcutaneously, It may be administered to a lesion or the like. The dosage of the antisense polynucleotide varies depending on the target disease, the administration subject, the administration route, and the like.For example, when the antisense polynucleotide of the present invention is administered for the purpose of treating diabetic nephropathy, In general, for adults (body weight 60 kg), about 0.1 to 100 mg of the antisense polynucleotide is administered per day. You.
さらに、 該アンチセンスポリヌクレオチドは、 組織や細胞における本発明の D NAの存在やその発現状況を調べるための診断用オリゴヌクレオチドプロ一 ブとして使用することもできる。  Furthermore, the antisense polynucleotide can also be used as a diagnostic oligonucleotide probe for examining the presence or expression of the DNA of the present invention in tissues or cells.
上記アンチセンスポリヌクレオチドと同様に、 本発明のタンパク質をコード する R NAの一部を含有する二重鎖 R NA、 本発明のタンパク質をコードする R NAの一部を含有するリポザィムなども、 本発明の遺伝子の発現を抑制する ことができ、 生体内における本発明で用いられるタンパク質または本発明で用 いられる D N Aの機能を抑制することができるので、 例えば、 腎疾患 (例、 糖 尿病性腎症、 巣状糸球体硬化症、 慢性糸球体腎炎、 微小変化型ネフローゼ症候 群、 メサンギゥム増殖性腎炎、 管内増殖性腎炎、 IgA腎症、 膜性腎症、 膜性増殖 性腎炎、 半月体形成性腎炎 ループス腎炎、 アミロイド腎、 尿細管間質性腎 炎、 急性尿細管壊死、 急性腎不全、 腎硬化症、 腎性浮腫、 腎腫瘍、 腎移植後の 慢性拒絶など) などの予防 ·治療剤などとして使用することができる。  Similarly to the above-mentioned antisense polynucleotide, a double-stranded RNA containing a part of the RNA encoding the protein of the present invention, a lipozyme containing a part of the RNA encoding the protein of the present invention, etc. Since the expression of the gene of the present invention can be suppressed, and the function of the protein used in the present invention or the DNA used in the present invention in a living body can be suppressed, for example, renal diseases (eg, Nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome group, mesangial proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy, membranous proliferative nephritis, crescent formation Prevention and treatment of lupus nephritis, amyloid kidney, tubular interstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, renal tumors, chronic rejection after kidney transplantation, etc.) It can be used as such.
二重鎖 R NAは、 公知の方法 (例、 Nature, 411巻, 494頁, 2001年) に準じ て、 本発明のポリヌクレオチドの配列を基に設計して製造することができる。 リポザィムは、 公知の方法 (例、 TRENDS in Molecul ar Medi c ine, 7巻, 221 頁, 2001年) に準じて、 本発明のポリヌクレオチドの配列を基に設計して製造 することができる。 例えば、 本発明のタンパク質をコードする R N Aの一部に 公知のリポザィムを連結することによつて製造することができる。 本発明の夕 ンパク質をコードする R NAの一部としては、 公知のリポザィムによつて切断 され得る本発明の R N A上の切断部位に近接した部分 (R NA断片) が挙げら れる。  The double-stranded RNA can be designed and manufactured based on the sequence of the polynucleotide of the present invention according to a known method (eg, Nature, 411, 494, 2001). The lipozyme can be designed and manufactured based on the sequence of the polynucleotide of the present invention according to a known method (eg, TRENDS in Molecular Medicine, Vol. 7, p. 221, 2001). For example, it can be produced by linking a known lipozyme to a part of RNA encoding the protein of the present invention. A part of the RNA encoding the protein of the present invention includes a portion (RNA fragment) adjacent to the cleavage site on the RNA of the present invention which can be cleaved by a known lipozyme.
上記の二重鎖 R NAまたはリポザィムを上記予防 ·治療剤として使用する場 合、 アンチセンスポリヌクレオチドと同様にして製剤化し、 投与することがで さる。  When the above-described double-stranded RNA or lipozyme is used as the above-mentioned prophylactic / therapeutic agent, it can be formulated and administered in the same manner as an antisense polynucleotide.
( 5 ) 本発明の抗体を含有する医薬 (5) a drug containing the antibody of the present invention
本発明のタンパク質の活性を中和する作用を有する本発明の抗体は、 例え ば、 腎疾患 (例、 糖尿病性腎症、 巣状糸球体硬化症、 慢性糸球体腎炎、 微小変 化型ネフローゼ症候群、 メサンギゥム増殖性腎炎、 管内増殖性腎炎、 IgA腎症、 膜性腎症、 膜性増殖性腎炎、 半月体形成性腎炎、 ループス腎炎、 アミロイド 腎、 尿細管間質性腎炎、 急性尿細管壊死、 急性腎不全、 腎硬化症、 腎性浮腫、 腎腫瘍、 腎移植後の慢性拒絶など) などの予防'治療剤として使用することが できる。 The antibody of the present invention, which has the activity of neutralizing the activity of the protein of the present invention, includes, for example, For example, renal diseases (eg, diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, microdegenerative nephrotic syndrome, mesangial proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy, Membranous proliferative nephritis, crescentic nephritis, lupus nephritis, amyloid kidney, tubulointerstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, renal tumor, chronic after renal transplantation Rejection) can be used as a prophylactic or therapeutic agent.
本発明の抗体は、 それ自体または適当な医薬組成物として投与することがで きる。 上記投与に用いられる医薬組成物は、 上記抗体またはその塩と薬理学的 に許容され得る担体、 希釈剤もしくは賦形剤とを含むものである。 かかる組成 物は、 経口または非経口投与に適する剤形として提供される。  The antibodies of the present invention can be administered by themselves or as a suitable pharmaceutical composition. The pharmaceutical composition used for the administration contains the antibody or a salt thereof and a pharmacologically acceptable carrier, diluent or excipient. Such compositions are provided in dosage forms suitable for oral or parenteral administration.
すなわち、 例えば、 経口投与のための組成物としては、 固体または液体の剤 形、 具体的には錠剤 (糖衣錠、 フィルムコ一ティング錠を含む) 、 丸剤、 顆粒 剤、 散剤、 カプセル剤 (ソフトカプセル剤を含む) 、 シロップ剤、 乳剤、 懸濁 剤などがあげられる。 かかる組成物は公知の方法によって製造され、 製剤分野 において通常用いられる担体、 希釈剤もしくは賦形剤を含有するものである。 例えば、 錠剤用の担体、 賦形剤としては、 乳糖、 でんぷん、 蔗糖、 ステアリン 酸マグネシウムなどが用いられる。  That is, for example, compositions for oral administration include solid or liquid dosage forms, specifically tablets (including sugar-coated tablets and film-coated tablets), pills, granules, powders, capsules (soft capsules) And syrups, emulsions, suspensions and the like. Such a composition is produced by a known method and contains a carrier, diluent or excipient commonly used in the pharmaceutical field. For example, lactose, starch, sucrose, magnesium stearate and the like are used as carriers and excipients for tablets.
非経口投与のための組成物としては、 例えば、 注射剤、 坐剤などが用いら れ、 注射剤は静脈注射剤、 皮下注射剤、 皮内注射剤、 筋肉注射剤、 点滴注射剤 などの剤形を包含する。 かかる注射剤は、 公知の方法に従って、 例えば、 上記 抗体またはその塩を通常注射剤に用いられる無菌の水性もしくは油性液に溶 解、 懸濁または乳化することによって調製する。 注射用の水性液としては、 例 えば、 生理食塩水、 プドウ糖やその他の補助薬を含む等張液などが用いられ、 適当な溶解補助剤、 例えば、 アルコール (例、 エタノール) 、 ポリアルコール (例、 プロピレングリコール、 ポリエチレングリコール) 、 非イオン界面活性 剤 〔例、 ポリソルべ一ト 80、 HCO-50 (polyoxyethylene (50mol) addoct of hydrogenated cas tor oi l) 〕 などと併用してもよい。 油性液としては、 例え ば、 ゴマ油、 大豆油などが用いられ、 溶解補助剤として安息香酸ベンジル、 ベ ンジルアルコールなどを併用してもよい。 調製された注射液は、 通常、 適当な アンプルに充填される。 直腸投与に用いられる坐剤は、 上記抗体またはその塩 を通常の坐薬用基剤に混合することによって調製される。 As compositions for parenteral administration, for example, injections, suppositories, etc. are used. Injections include intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections, intravenous injections, etc. Include shapes. Such injections are prepared according to known methods, for example, by dissolving, suspending or emulsifying the antibody or a salt thereof in a sterile aqueous or oily liquid commonly used for injections. Examples of the aqueous liquid for injection include physiological saline, isotonic solution containing pudose and other adjuvants, and a suitable solubilizing agent such as alcohol (eg, ethanol) and polyalcohol ( For example, propylene glycol, polyethylene glycol), a nonionic surfactant [eg, polysorbate 80, HCO-50 (polyoxyethylene (50 mol) addoct of hydrogenated catalyst)], etc. may be used in combination. As the oily liquid, for example, sesame oil, soybean oil, and the like are used, and benzyl benzoate, benzyl alcohol, and the like may be used in combination as a solubilizing agent. The prepared injection solution is usually Filled into ampoules. A suppository for rectal administration is prepared by mixing the above antibody or a salt thereof with a usual suppository base.
上記の経口用または非経口用医薬組成物は、 活性成分の投与量に適合するよ うな投薬単位の剤形に調製されることが好都合である。 かかる投薬単位の剤形 としては、 錠剤、 丸剤、 カプセル剤、 注射剤 (アンプル) 、 坐剤などが例示さ れ、 それぞれの投薬単位剤形当たり通常 5〜500mg、 とりわけ注射剤では 5〜  The above-mentioned oral or parenteral pharmaceutical composition is conveniently prepared in the form of a dosage unit so as to conform to the dose of the active ingredient. Examples of such dosage unit forms include tablets, pills, capsules, injections (ampoules), and suppositories, and usually 5 to 500 mg per dosage unit form, especially 5 to 500 mg for injections.
100mg、 その他の剤形では 10〜250mgの上記抗体が含有されていることが好まし い。 It is preferred that 100 mg, and other dosage forms, contain 10 to 250 mg of the above antibody.
なお前記した各組成物は、 上記抗体との配合により好ましくない相互作用を 生じない限り他の活性成分を含有してもよい。  Each of the above-mentioned compositions may contain another active ingredient as long as the composition does not cause an undesirable interaction with the above-mentioned antibody.
本発明の抗体を含有する上記疾患の予防 ·治療剤は低毒性であり、 そのまま 液剤として、 または適当な剤型の医薬組成物として、 ヒトまたは哺乳動物  The prophylactic / therapeutic agent for the above-mentioned diseases containing the antibody of the present invention has low toxicity and can be used as it is as a liquid or as a pharmaceutical composition of an appropriate dosage form, in humans or mammals.
(例、 ラット、 ゥサギ、 ヒッジ、 ブ夕、 ゥシ、 ネコ、 ィヌ、 サルなど) に対し て経口的または非経口的 (例、 静脈投与) に投与することができる。 投与量 は、 投与対象、 対象疾患、 症状、 投与ルートなどによっても異なるが、 例え ば、 成人の糖尿病性腎症の治療のために使用する場合には、 本発明の抗体を 1 回量として、 通常 0. 01〜20mg/kg体重程度、 好ましくは 0. l〜10mg/kg体重程度、 さらに好ましくは 0. 〜 5mg/kg体重程度を、 1日 1〜5回程度、 好ましくは 1日 1〜3 回程度、 注射剤として投与するのが好都合である。 他の非経口投与および経口 投与の場合もこれに準ずる量を投与することができる。 症状が特に重い場合に は、 その症状に応じて増量してもよい。  It can be administered orally or parenterally (eg, intravenously) to animals (eg, rats, egrets, sheep, sheep, bush, squirrels, cats, dogs, monkeys, etc.). The dose varies depending on the administration subject, target disease, symptoms, administration route, and the like.For example, when used for the treatment of diabetic nephropathy in adults, the antibody of the present invention is used as a single dose. Usually, about 0.01 to 20 mg / kg body weight, preferably about 0.1 to 10 mg / kg body weight, more preferably about 0.1 to 5 mg / kg body weight, about 1 to 5 times a day, preferably 1 to 5 times a day It is convenient to administer as an injection about three times. In the case of other parenteral administration and oral administration, an equivalent dose can be administered. If the symptoms are particularly severe, the dose may be increased accordingly.
また、 本発明の抗体は、 例えば、 腎疾患 (例、 糖尿病性腎症、 巣状糸球体硬 化症、 慢性糸球体腎炎、 微小変化型ネフローゼ症候群、 メサンギゥム増殖性腎 炎、 管内増殖性腎炎、 IgA腎症、 膜性腎症、 膜性増殖性腎炎、 半月体形成性腎 炎、 ループス腎炎、 アミロイド腎、 尿細管間質性腎炎、 急性尿細管壊死、 急性 腎不全、 腎硬化症、 腎性浮腫、 腎腫瘍、 腎移植後の慢性拒絶など) などの診断 薬としても有用である。 (6) DNA転移動物 Further, the antibody of the present invention can be used, for example, for renal diseases (eg, diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome, mesangial proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy, membranous proliferative nephritis, crescentic nephritis, lupus nephritis, amyloid kidney, tubulointerstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal It is also useful as a diagnostic agent for edema, kidney tumors, chronic rejection after kidney transplantation, etc.). (6) DNA transfer animal
本発明は、 外来性の本発明のタンパク質をコードする DNA (以下、 本発明 の外来性 DN Aと略記する) またはその変異 DN A (本発明の外来性変異 DN Aと略記する場合がある) を有する非ヒ卜哺乳動物を提供する。  The present invention relates to a DNA encoding the exogenous protein of the present invention (hereinafter abbreviated as the exogenous DNA of the present invention) or a mutant DNA thereof (sometimes abbreviated as the exogenous mutant DNA of the present invention). And a non-human mammal having the formula:
すなわち、 本発明は、  That is, the present invention
(1) 本発明の外来性 DNAまたはその変異 DNAを有する非ヒト哺乳動物、 (1) a non-human mammal having the exogenous DNA of the present invention or a mutant DNA thereof,
(2) 非ヒト哺乳動物がゲッ歯動物である第 (1)記載の動物、 (2) the animal according to (1), wherein the non-human mammal is a rodent;
(3) ゲッ歯動物がマウスまたはラッ卜である第 (2) 記載の動物、 および (3) The animal according to (2), wherein the rodent is a mouse or a rat, and
(4) 本発明の外来性 DNAまたはその変異 DNAを含有し、 哺乳動物におい て発現しうる組換えべクタ一を提供するものである。 (4) It is intended to provide a recombinant vector containing the exogenous DNA of the present invention or its mutant DNA, which can be expressed in mammals.
本発明の外来性 DNAまたはその変異 DNAを有する非ヒト哺乳動物 (以 下、 本発明の DNA転移動物と略記する) は、 未受精卵、 受精卵、 精子および その始原細胞を含む胚芽細胞などに対して、 好ましくは、 非ヒト哺乳動物の発 生における胚発生の段階 (さらに好ましくは、 単細胞または受精卵細胞の段階 でかつ一般に 8細胞期以前) に、 リン酸カルシウム法、 電気パルス法、 リボフ ェクション法、 凝集法、 マイクロインジェクション法、 パ一ティクルガン法、 DEAE—デキストラン法などにより目的とする DN Aを転移することによつ て作出することができる。 また、 該 DNA転移方法により、 体細胞、 生体の臓 器、 組織細胞などに目的とする本発明の外来性 DNAを転移し、 細胞培養、 組 織培養などに利用することもでき、 さらに、 これら細胞を上述の胚芽細胞と自 体公知の細胞融合法により融合させることにより本発明の D N A転移動物を作 出することもできる。  Non-human mammals having the exogenous DNA of the present invention or the mutant DNA thereof (hereinafter abbreviated as the DNA transgenic animal of the present invention) include unfertilized eggs, fertilized eggs, germ cells including spermatozoa and their progenitor cells, and the like. In contrast, preferably, during the stage of embryonic development in the development of a non-human mammal (more preferably, at the stage of a single cell or a fertilized egg and generally before the 8-cell stage), a calcium phosphate method, an electric pulse method, a ribofection method, It can be produced by transferring the desired DNA by an agglutination method, microinjection method, particle gun method, DEAE-dextran method, or the like. Further, by the DNA transfer method, the exogenous DNA of the present invention can be transferred to somatic cells, organs of living organisms, tissue cells, and the like, and can be used for cell culture, tissue culture, and the like. The DNA-transferred animal of the present invention can also be produced by fusing cells with the above-mentioned germ cells by a cell fusion method known per se.
非ヒト哺乳動物としては、 例えば、 ゥシ、 ブタ、 ヒッジ、 ャギ、 ゥサギ、 ィ ヌ、 ネコ、 モルモット、 ハムスター、 マウス、 ラットなどが用いられる。 なか でも、 病体動物モデル系の作成の面から個体発生および生物サイクルが比較的 短く、 また、 繁殖が容易なゲッ歯動物、 とりわけマウス (例えば、 純系とし て、 C57BL/6系統, DBA2系統など、 交雑系として、 B6C3F,系統, BDF【系統, B6D2F,系統, BALB/c系統, ICR系統など) またはラット (例えば、 Wistar, SDな ど) などが好ましい。 哺乳動物において発現しうる組換えベクターにおける 「哺乳動物」 として は、 上記の非ヒト哺乳動物の他にヒトなどがあげられる。 As non-human mammals, for example, porcupines, pigs, higgins, goats, magpies, dogs, cats, guinea pigs, hamsters, mice, rats and the like are used. Above all, rodents with relatively short ontogeny and biological cycles in terms of the creation of disease animal model systems, and easy to breed rodents, especially mice (for example, pure strains such as C57BL / 6 strain, DBA2 strain, etc.) As the crossing strain, B6C3F, strain, BDF [strain, B6D2F, strain, BALB / c strain, ICR strain, etc.) or rat (eg, Wistar, SD, etc.) is preferable. "Mammals" in a recombinant vector that can be expressed in mammals include humans and the like in addition to the above-mentioned non-human mammals.
本発明の外来性 D NAとは、 非ヒ卜哺乳動物が本来有している本発明の D N Aではなく、 いったん哺乳動物から単離 ·抽出された本発明の D N Aをいう。 本発明の変異 D NAとしては、 元の本発明の D N Aの塩基配列に変異 (例え ば、 突然変異など) が生じたもの、 具体的には、 塩基の付加、 欠損、 他の塩基 へ 置換などが生じた D NAなどが用いられ、 また、 異常 D NAも含まれる。 該異常 D N Aとレては、 異常な本発明のタンパク質を発現させる D N Aを意 味し、 例えば、 正常な本発明のタンパク質の機能を抑制するタンパク質を発現 させる D N Aなどが用いられる。  The exogenous DNA of the present invention refers not to the DNA of the present invention originally possessed by non-human mammals but to the DNA of the present invention once isolated and extracted from the mammal. Examples of the mutant DNA of the present invention include those in which a mutation (for example, mutation) has occurred in the base sequence of the original DNA of the present invention, specifically, addition or deletion of a base, substitution with another base, or the like. The DNA in which the occurrence has occurred is used, and the abnormal DNA is also included. The abnormal DNA refers to a DNA that expresses an abnormal protein of the present invention, and for example, a DNA that expresses a protein that suppresses the function of the normal protein of the present invention is used.
本発明の外来性 D NAは、 対象とする動物と同種あるいは異種のどちらの哺 乳動物由来のものであってもよい。 本発明の D N Aを対象動物に転移させるに あたっては、 該 D N Aを動物細胞で発現させうるプロモーターの下流に結合し た D NAコンストラクトとして用いるのが一般に有利である。 例えば、 本発明 のヒ卜 D NAを転移させる場合、 これと相同性が高い本発明の D NAを有する 各種哺乳動物 (例えば、 ゥサギ、 ィヌ、 ネコ、 モルモット、 ハムスター、 ラッ ト、 マウスなど) 由来の D NAを発現させうる各種プロモータ一の下流に、 本 発明のヒト D NAを結合した D NAコンストラクト (例、 ベクターなど) を対 象哺乳動物の受精卵、 例えば、 マウス受精卵へマイクロインジェクションする ことによつて本発明の D N Aを高発現する D N A転移哺乳動物を作出すること ができる。  The exogenous DNA of the present invention may be derived from a mammal that is the same or different from the animal of interest. In transferring the DNA of the present invention to a target animal, it is generally advantageous to use the DNA as a DNA construct downstream of a promoter capable of being expressed in animal cells. For example, when the human DNA of the present invention is transferred, various mammals having the DNA of the present invention having a high homology with the human (for example, egret, dog, cat, guinea pig, hamster, rat, mouse, etc.) Microinjection of a DNA construct (e.g., vector, etc.) to which a human DNA of the present invention is bound downstream of one of various promoters capable of expressing DNA of origin from a fertilized egg of a target mammal, for example, a mouse fertilized egg By doing so, a DNA-transferred mammal that highly expresses the DNA of the present invention can be produced.
本発明のタンパク質の発現べクタ一としては、 大腸菌由来のプラスミド、 枯 草菌由来のプラスミド、 酵母由来のプラスミド、 λファージなどのバクテリオ ファ一ジ、 モロニ一白血病ウィルスなどのレトロウイルス、 ワクシニアウィル スまたはバキュロウィルスなどの動物ウィルスなどが用いられる。 なかでも、 大腸菌由来のプラスミド、 枯草菌由来のプラスミドまたは酵母由来のプラスミ ドなどが好ましく用いられる。  Examples of the expression vector of the protein of the present invention include plasmids derived from Escherichia coli, plasmids derived from Bacillus subtilis, plasmids derived from yeast, bacteriophages such as λ phage, retroviruses such as Moroni leukemia virus, and vaccinia viruses. Alternatively, animal viruses such as baculovirus are used. Among them, a plasmid derived from Escherichia coli, a plasmid derived from Bacillus subtilis or a plasmid derived from yeast are preferably used.
上記の D N A発現調節を行なうプロモータ一としては、 例えば、 (i) ウィル ス (例、 シミアンウィルス、 サイトメガロウィルス、 モロニ一白血病ウィル ス、 J Cウィルス、 乳がんウィルス、 ポリオウイルスなど) に由来する DNA のプロモーター、 (ii) 各種哺乳動物 (ヒト、 ゥサギ、 ィヌ、 ネコ、 モルモッ ト、 ハムスター、 ラット、 マウスなど) 由来のプロモ一夕一、 例えば、 アルブ ミン、 インスリン I I、 ゥロブラキン I I、 エラス夕一ゼ、 エリス口ポェチ ン、 エンドセリン、 筋クレアチンキナーゼ、 グリア線維性酸性タンパク質、 グ ル夕チオン S—トランスフェラーゼ、 血小板由来成長因子 /3、 ケラチン K l, K1 0および Κ14、 コラーゲン I型および I I型、 サイクリック AMP依存 タンパク質キナーゼ i3 Iサブユニット、 ジストロフィン、 酒石酸抵抗性アル力 リフォスファターゼ、 心房ナトリゥム利尿性因子、 内皮レセプターチ口シンキ ナーゼ (一般に T i e 2と略される) 、 ナトリゥムカリゥムアデノシン 3リン 酸化酵素 (Na, K-ATP a s e) 、 ニューロフィラメント軽鎖、 メタロチ ォネイン Iおよび I I A、 メ夕ロプロティナ一ゼ 1組織インヒビター、 MHC クラス I抗原 (H- 2 L) 、 1-1— r a s、 レニン、 ド一パミン jS—水酸化醇 素、 甲状腺ペルォキシダ一ゼ (TPO) 、 ペプチド鎖延長因子 l a (EF- 1 ) 、 βァクチン、 «および |Sミオシン重鎖、 ミオシン軽鎖 1および 2、 ミエ リン基礎タンパク質、 チログロブリン、 Thy— 1、 免疫グロブリン、 H鎖可 変部 (VNP) 、 血清アミロイド Pコンポーネント、 ミオグロビン、 トロポニ ン〇、 平滑筋ひァクチン-. プレプロエンケフアリン A、 バソプレシンなどのプ 口モー夕—などが用いられる。 なかでも、 全身で高発現することが可能なサイ トメガロウィルスプロモ一ター、 ヒ卜ぺプチド鎖延長因子 1 a (EF- 1 ) のプロモーター、 ヒトおよびニヮトリ /3ァクチンプロモータ一などが好適であ る。 Examples of promoters that regulate the above DNA expression include: (i) virus (eg, simian virus, cytomegalovirus, Moroni leukemia virus) Promoters of DNA derived from humans, JC virus, breast cancer virus, poliovirus, etc.), and (ii) Promoters derived from various mammals (human, egret, dog, cat, guinea pig, hamster, rat, mouse, etc.) 1.For example, albumin, insulin II, perobrakin II, elas ubiquitin, erythropoietin, endothelin, muscle creatine kinase, glial fibrillary acidic protein, gliubithion S-transferase, platelet-derived growth factor / 3, Keratin Kl, K10 and Κ14, collagen type I and type II, cyclic AMP-dependent protein kinase i3 I subunit, dystrophin, tartrate-resistant lipophilic phosphatase, atrial sodium diuretic factor, endothelial receptor ostium synkinase ( (Generally abbreviated as Tie 2) Pharmacium adenosine 3-phosphate oxidase (Na, K-ATPase), neurofilament light chain, metallotionin I and IIA, meloproteinase 1 tissue inhibitor, MHC class I antigen (H-2L), 1 -1—ras, renin, dopamine jS—hydroxyl, thyroid peroxidase (TPO), peptide chain elongation factor la (EF-1), β-actin, «and | S myosin heavy chain, myosin light chain 1 and 2, myelin base protein, thyroglobulin, Thy-1, immunoglobulin, heavy chain variable region (VNP), serum amyloid P component, myoglobin, troponin II, smooth muscle actin-. Preproenkephalin A And vasopressin and the like. Among them, a cytomegalovirus promoter that can be highly expressed in the whole body, a promoter of a human peptide chain elongation factor 1a (EF-1), a human and a chicken / 3-actin promoter are preferable. is there.
上記べクターは、 D N A転移哺乳動物において目的とするメッセンジャー R N Aの転写を終結する配列 (一般にターミネ夕一と呼ばれる) を有しているこ とが好ましく、 例えば、 ウィルス由来および各種哺乳動物由来の各 DNAの配 列を用いることができ、 好ましくは、 シミアンウィルスの SV40ターミネタ —などが用いられる。  The vector preferably has a sequence that terminates transcription of a messenger RNA of interest in a DNA-transferred mammal (generally called terminator). For example, each vector may be derived from a virus or a mammal. A DNA sequence can be used, and preferably, SV40 terminator of simian virus or the like is used.
その他、 目的とする外来性 DNAをさらに高発現させる目的で各 DNAのス プライシングシグナル、 ェンハンサー領域、 真核 DN Aのイントロンの一部な どをプロモーター領域の 5 ' 上流、 プロモーター領域と翻訳領域間あるいは翻 訳領域の 3 ' 下流 に連結することも目的により可能である。 In addition, splicing signals of each DNA, enhancer region, and part of eukaryotic DNA introns for the purpose of further expressing the desired foreign DNA. It is also possible to connect a promoter 5 ′ upstream of the promoter region, between the promoter region and the translation region, or 3 ′ downstream of the translation region.
正常な本発明のタンパク質の翻訳領域は、 ヒトまたは各種哺乳動物.(例え ば、 ゥサギ、 ィヌ、 ネコ、 モルモット、 八ムスター、 ラット、 マウスなど) 由 来の肝臓、 腎臓、 甲状腺細胞、 線維芽細胞由来 D N Aおよび市販の各種ゲノム D NAライブラリーよりゲノム D NAの全てあるいは一部として、 または干 臓、 腎臓、 甲状腺細胞、 線維芽細胞由来 R NAより公知の方法により調製され た相補 D NAを原料として取得することが出来る。 また、 外来性の異常 D NA は、 上記の細胞または組織より得られた正常なタンパク質の翻訳領域を点突然 変異誘発法により変異した翻訳領域を作製することができる。  The normal translation region of the protein of the present invention is human or various mammals (eg, egrets, dogs, cats, guinea pigs, eight musters, rats, mice, etc.). Liver, kidney, thyroid cells, fibroblasts All or part of genomic DNA from cell-derived DNA and various commercially available genomic DNA libraries, or complementary DNA prepared by known methods from dried, kidney, thyroid, or fibroblast-derived RNA. It can be obtained as a raw material. In addition, a foreign abnormal DNA can produce a translation region obtained by mutating the translation region of a normal protein obtained from the above-described cells or tissues by a point mutagenesis method.
該翻訳領域は転移動物において発現しうる D NAコンストラクトとして、 前 記のプロモーターの下流および所望により転写終結部位の上流に連結させる通 常の D N A工学的手法により作製することができる。  The translation region can be prepared as a DNA construct that can be expressed in a transposed animal by a conventional DNA engineering technique in which it is ligated downstream of the above-mentioned promoter and, if desired, upstream of the transcription termination site.
受精卵細胞段階における本発明の外来性 D NAの転移は、 対象哺乳動物の胚 芽細胞および体細胞のすべてに存在するように確保される。 D N A転移後の作 出動物の胚芽細胞において、 本発明の外来性 D N Aが存在することは、 作出動 物の後代がすべて、 その胚芽細胞および体細胞のすべてに本発明の外来性 D N Aを保持することを意味する。 本発明の外来性 D N Aを受け継いだこの種の動 物の子孫はその胚芽細胞および体細胞のすべてに本発明の外来性 D NAを有す る。  Transfer of the exogenous DNA of the present invention at the fertilized egg cell stage is ensured to be present in all germ cells and somatic cells of the target mammal. The presence of the exogenous DNA of the present invention in the germ cells of the produced animal after DNA transfer means that all the progeny of the produced animal retain the exogenous DNA of the present invention in all of the germ cells and somatic cells Means that. The progeny of such animals inheriting the exogenous DNA of the present invention have the exogenous DNA of the present invention in all of their germinal and somatic cells.
本発明の外来性正常 D NAを転移させた非ヒト哺乳動物は、 交配により外来 性 D N Aを安定に保持することを確認して、 該 D N A保有動物として通常の飼 育環境で継代飼育することが出来る。  The non-human mammal to which the exogenous normal DNA of the present invention has been transferred is confirmed to stably maintain exogenous DNA by mating, and should be subcultured as an animal having the DNA in a normal breeding environment. Can be done.
受精卵細胞段階における本発明の外来性 D N Aの転移は、 対象哺乳動物の胚 芽細胞および体細胞の全てに過剰に存在するように確保される。 D NA転移後 の作出動物の胚芽細胞において本発明の外来性 D N Aが過剰に存在すること は、 作出動物の子孫が全てその胚芽細胞および体細胞の全てに本発明の外来性 D NAを過剰に有することを意味する。 本発明の外来性 D N Aを受け継いだこ の種の動物の子孫はその胚芽細胞および体細胞の全てに本発明の外来性 D N A を過剰に有する。 Transfer of the exogenous DNA of the present invention at the fertilized egg cell stage is ensured to be present in excess in all germ cells and somatic cells of the target mammal. Excessive presence of the exogenous DNA of the present invention in the germinal cells of the animal after transfer of the DNA indicates that the offspring of the animal in which the animal is produced excessively contains the exogenous DNA of the present invention in all of its germ cells and somatic cells. Means to have. The offspring of an animal of this species that has inherited the exogenous DNA of the present invention, In excess.
導入 D N Aを相同染色体の両方に持つホモザィゴート動物を取得し、 この雌 雄の動物を交配することによりすべての子孫が該 D N Aを過剰に有するように 繁殖継代することができる。  By obtaining a homozygous animal having the introduced DNA on both homologous chromosomes, and mating the male and female animals, it is possible to breed so that all offspring have the DNA in excess.
本発明の正常 D NAを有する非ヒト哺乳動物は、 本発明の正常 D NAが高発 現させられており、 内在性の正常 D N Aの機能を促進することにより最終的に 本発明の夕ンパク質の機能亢進症を発症することがあり、 その病態モデル動物 として利用することができる。 例えば、 本発明の正常 D N A転移動物を用い て、 本発明のタンパク質の機能亢進症や、 本発明のタンパク質が関連する疾患 の病態機序の解明およびこれらの疾患の治療方法の検討を行なうことが可能で める。  The non-human mammal having the normal DNA of the present invention expresses the normal DNA of the present invention at a high level, and eventually promotes the function of endogenous normal DNA to thereby finally produce the protein of the present invention. May develop hyperfunction, and can be used as a model animal for the disease. For example, using the normal DNA-transferred animal of the present invention, it is possible to elucidate the pathological mechanism of the hyperactivity of the protein of the present invention and diseases associated with the protein of the present invention, and to examine a method for treating these diseases. It is possible.
また、 本発明の外来性正常 D NAを転移させた哺乳動物は、 遊離した本発明 のタンパク質の増加症状.を有することから、 本発明のタンパク質に関連する疾 患に対する予防 治療剤、 例えば、 腎疾患 (例、 糖尿病性腎症、 巣状糸球体硬 化症、 慢性糸球体腎炎、 微小変化型ネフ口一ゼ症候群、 メサンギゥム増殖性腎 炎、 管内増殖性腎炎、 IgA腎症、 膜性腎症、 膜性増殖性腎炎、 半月体形成性腎 炎、 ループス腎炎、 アミロイド腎、 尿細管間質性腎炎、 急性尿細管壊死、 急性 腎不全、 腎硬化症、 腎性浮腫、 腎腫瘍、 腎移植後の慢性拒絶など) などの予 防 ·治療剤のスクリーニング試験にも利用可能である。  In addition, since the mammal to which the exogenous normal DNA of the present invention has been transferred has an increased symptom of the free protein of the present invention, a prophylactic / therapeutic agent for a disease associated with the protein of the present invention, such as kidney Diseases (e.g., diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimally altered nephritis syndrome, mesangial proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy , Membranous proliferative nephritis, crescentic nephritis, lupus nephritis, amyloid kidney, tubular interstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, renal tumor, after kidney transplantation It can also be used for screening tests for prophylactic and therapeutic agents such as chronic rejection.
一方、 本発明の外来性異常 D NAを有する非ヒト哺乳動物は、 交配により外 来性 D N Aを安定に保持することを確認して該 D N A保有動物として通常の飼 育環境で継代飼育することが出来る。 さらに、 目的とする外来 D NAを前述の プラスミドに組み込んで原科として用いることができる。 プロモーターとの D N Aコンストラク卜は、 通常の D NA工学的手法によって作製することができ る。 受精卵細胞段階における本発明の異常 D N Aの転移は、 対象哺乳動物の胚 芽細胞および体細胞の全てに存在するように確保される。 D NA転移後の作出 動物の胚芽細胞において本発明の異常 D N Aが存在することは、 作出動物の子 孫が全てその胚芽細胞および体細胞の全てに本発明の異常 D N Aを有すること を意味する。 本発明の外来性 D NAを受け継いだこの種の動物の子孫は、 その 胚芽細胞および体細胞の全てに本発明の異常 D N Aを有する。 導入 D NAを相 同染色体の両方に持つホモザィゴート動物を取得し、 この雌雄の動物を交配す ることによりすべての子孫が該 D N Aを有するように繁殖継代することができ る。 On the other hand, the non-human mammal having the foreign abnormal DNA of the present invention should be subcultured in a normal breeding environment as an animal having the DNA after confirming that the foreign DNA is stably retained by the crossing. Can be done. Further, the desired foreign DNA can be incorporated into the above-mentioned plasmid and used as a source substance. The DNA construct with the promoter can be prepared by ordinary DNA engineering techniques. The transfer of the abnormal DNA of the present invention at the fertilized egg cell stage is ensured to be present in all germ cells and somatic cells of the target mammal. The presence of the abnormal DNA of the present invention in the germinal cells of a produced animal after DNA transfer means that the offspring of the produced animal have the abnormal DNA of the present invention in all of its germinal and somatic cells. The offspring of this type of animal that inherited the exogenous DNA of the present invention All germ cells and somatic cells have the abnormal DNA of the present invention. By obtaining a homozygous animal having the introduced DNA on both homologous chromosomes, and crossing the male and female animals, it is possible to breed subculture so that all offspring have the DNA.
本発明の異常 D NAを有する非ヒト哺乳動物は、 本発明の異常 D NAが高発 現させられており、 内在性の正常 D N Aの機能を阻害することにより最終的に 本発明のタンパク質の機能不活性型不応症となることがあり、 その病態モデル 動物として利用することができる。 例えば、 本発明の異常 D N A転移動物を用 いて、 本発明のタンパク質の機能不活性型不応症の病態機序の解明およびこの 疾患を治療方法の検討を行なうことが可能である。  In the non-human mammal having the abnormal DNA of the present invention, the abnormal DNA of the present invention is highly expressed, and the function of the protein of the present invention is ultimately reduced by inhibiting the function of endogenous normal DNA. Inactive refractory disease may occur, and it can be used as a disease model animal. For example, it is possible to elucidate the pathological mechanism of the functionally inactive refractory state of the protein of the present invention and to examine a method for treating this disease using the abnormal DNA transgenic animal of the present invention.
また、 具体的な利用可能性としては、 本発明の異常 D N A高発現動物は、 本 発明のタンパク質の機能不活性型不応症における本発明の異常タンパク質によ る正常タンパク質の機能阻害 (dominant negat ive作用) を解明するモデルとな る。  In addition, as a specific possibility, the abnormal DNA highly expressing animal of the present invention can be used to inhibit the function of a normal protein by the abnormal protein of the present invention in the function-inactive refractory disease of the protein of the present invention (dominant negative activity). Action).
また、 本発明の外来異常 D NAを転移させた哺乳動物は、 遊離した本発明の 夕ンパク質の増加症状を有することから、 本発明のタンパク質または機能不活 性型不応症に対する予防 ·治療剤、 例えば、 腎疾患 (例、 糖尿病性腎症、 巣状 糸球体硬化症、 慢性糸球体腎炎、 微小変化型ネフローゼ症候群、 メサンギゥム 増殖性腎炎、 管内増殖性腎炎、 IgA腎症、 膜性腎症、 膜性増殖性腎炎、 半月体形 成性腎炎、 ループス腎炎、 アミロイド腎、 尿細管間質性腎炎、 急性尿細管壊 死、 急性腎不全、 腎硬化症、 腎性浮腫、 腎腫瘍、 腎移植後の慢性拒絶など) な どの予防 ·治療剤のスクリーニング試験にも利用可能である。  In addition, since the mammal to which the foreign abnormal DNA of the present invention has been transferred has an increased symptom of the released protein of the present invention, the preventive / therapeutic agent for the protein or functionally inactive type refractory of the present invention is used. , For example, renal diseases (eg, diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome, mesangial proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy, Membranous proliferative nephritis, crescentic adult nephritis, lupus nephritis, amyloid kidney, tubular interstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, renal tumor, after kidney transplantation It can also be used for screening tests for preventive and therapeutic agents such as chronic rejection.
また、 上記 2種類の本発明の D N A転移動物のその他の利用可能性として、 例えば、  In addition, other possible uses of the above two types of DNA transgenic animals of the present invention include, for example,
(i) 組織培養のための細胞源としての使用、  (i) use as a cell source for tissue culture,
(i i) 本発明の D NA転移動物の組織中の D NAもしくは R N Aを直接分析す るか、 または D NAにより発現されたペプチド組織を分析することによる、 本 発明のタンパク質により特異的に発現あるいは活性化するペプチドとの関連性 についての解析、 (i i i) D NAを有する組織の細胞を標準組織培養技術により培養し、 これらを 使用して、 一般に培養困難な組織からの細胞の機能の研究、 (ii) Direct analysis of DNA or RNA in the tissue of the DNA-transferred animal of the present invention, or analysis of peptide tissue expressed by the DNA to specifically express or express the protein of the present invention. Analysis of the relationship with the activating peptide, (iii) culturing cells of a tissue having DNA by standard tissue culture techniques, and using these to study the function of cells from tissues that are generally difficult to culture,
(iv) 上記 (i i i) 記載の細胞を用いることによる細胞の機能を高めるような薬 剤のスクリーニング、 および  (iv) screening for a drug that enhances cell function by using the cell described in (iii) above, and
(V) 本発明の変異タンパク質を単離精製およびその抗体作製などが考えられ る。  (V) Isolation and purification of the mutant protein of the present invention and production of its antibody may be considered.
さらに、 本発明の D NA転移動物を用いて、 本発明のタンパク質の機能不活 性型不応症などを含む、 本発明のタンパク質に関連する疾患の臨床症状を調べ ることができ、 また、 本発明のタンパク質に関連する疾患モデルの各臓器にお けるより詳細な病理学的所見が得られ、 新しい治療方法の開発、 さらには、 該 疾患による二次的疾患の研究および治療に貢献することができる。  Furthermore, using the DNA-transferred animal of the present invention, it is possible to examine the clinical symptoms of diseases related to the protein of the present invention, including the inactive type refractory type of the protein of the present invention, and the like. It is possible to obtain more detailed pathological findings in each organ of the disease model related to the protein of the present invention, to develop new treatment methods, and to contribute to the research and treatment of secondary diseases caused by the disease. it can.
また、 本発明の D N A転移動物から各臓器を取り出し、 細切後、 トリプシン などのタンパク質分解酵素により、 遊離した D N A転移細胞の取得、 その培養 またはその培養細胞の系統化を行なうことが可能である。 さらに、 本発明の夕 ンパク質産生細胞の特定化、 アポトーシス、 分化あるいは増殖との関連性、 ま たはそれらにおけるシグナル伝達機構を調べ、 それらの異常を調べることなど ができ、 本発明のタンパク質およびその作用解明のための有効な研究材料とな る。  In addition, it is possible to take out each organ from the DNA-transferred animal of the present invention, shred it, and then use a protease such as trypsin to obtain free DNA-transferred cells, culture them, or systematize the cultured cells. . Furthermore, it is possible to identify the protein-producing cells of the present invention, examine their relationship with apoptosis, differentiation or proliferation, or investigate their signal transduction mechanisms, and investigate their abnormalities. It is an effective research material for elucidating its action.
さらに、 本発明の D NA転移動物を用いて、 本発明のタンパク質の機能不活 性型不応症を含む、 本発明のタンパク質に関連する疾患の治療薬の開発を行な うために、 上述の検査法および定量法などを用いて、 有効で迅速な該疾患治療 薬のスクリーニング法を提供することが可能となる。 また、 本発明の D NA転 移動物または本発明の外来性 D N A発現べクターを用いて、 本発明の夕ンパク 質が関連する疾患の D N A治療法を検討、 開発することが可能である。  Further, in order to use the DNA-transferred animal of the present invention to develop a therapeutic agent for a disease associated with the protein of the present invention, including the inactive type refractory type of the protein of the present invention, Using a test method and a quantitative method, it is possible to provide an effective and rapid screening method for a therapeutic agent for the disease. Furthermore, using the DNA transfer product of the present invention or the exogenous DNA expression vector of the present invention, it is possible to study and develop a method for treating a DNA associated disease of the present invention for DNA.
( 7 ) ノックアウト動物 (7) Knockout animal
本発明は、 本発明の D NAが不活性化された非ヒト哺乳動物胚幹細胞および 本発明の D NA発現不全非ヒト哺乳動物を提供する。  The present invention provides a non-human mammalian embryonic stem cell in which the DNA of the present invention has been inactivated, and a non-human mammal deficient in expression of the DNA of the present invention.
すなわち、 本発明は、 ( 1 ) 本発明の D N Aが不活性化された非ヒト哺乳動物胚幹細胞、 That is, the present invention (1) a non-human mammalian embryonic stem cell in which the DNA of the present invention has been inactivated,
(2) 該 DNAがレポ一ター遺伝子 (例、 大腸菌由来の ιδ—ガラクトシダ一ゼ 遺伝子) を導入することにより不活性化された第 (1) 項記載の胚幹細胞、 (2) The embryonic stem cell according to (1), wherein the DNA is inactivated by introducing a reporter gene (eg, an ιδ-galactosidase gene derived from Escherichia coli).
(3) ネオマイシン耐性である第 (1) 項記載の胚幹細胞、 (3) The embryonic stem cell according to (1), which is neomycin-resistant,
(4) 非ヒト哺乳動物がゲッ歯動物である第 (1) 項記載の胚幹細胞、 (4) The embryonic stem cell according to (1), wherein the non-human mammal is a rodent,
(5) ゲッ歯動物がマウスである第 (4) 項記載の胚幹細胞、 (5) The embryonic stem cell according to (4), wherein the rodent is a mouse,
(6) 本発明の DN Αが不活性化された該 DN Α発現不全非ヒト哺乳動物、 (6) a non-human mammal deficient in expression of DN の of the present invention, wherein the DNΑ is inactivated;
(7) 該 DNAがレポ一夕一遺伝子 (例、 大腸菌由来の ;8—ガラクトシダーゼ 遺伝子) を導入することにより不活性化され、 該レポ一夕一遺伝子が本発明の DNAに対するプロモーターの制御下で発現しうる第 (6) 項記載の非ヒト哺 乳動物、 (7) The DNA is inactivated by introducing a repo overnight gene (eg, an 8-galactosidase gene derived from Escherichia coli), and the repo overnight gene is under the control of a promoter for the DNA of the present invention. A non-human mammal according to (6), which is capable of expressing;
(8) 非ヒト哺乳動物がゲッ歯動物である第 (6) 項記載の非ヒ卜哺乳動物、 (8) the non-human mammal according to (6), wherein the non-human mammal is a rodent;
(9) ゲッ歯動物がマウスである第 (8) 項記載の非ヒト哺乳動物、 および(9) the non-human mammal according to (8), wherein the rodent is a mouse; and
(10) 第 (7) 項記載の動物に、 試験化合物を投与し、 レポ一夕一遺伝子の 発現を検出することを特徴とする本発明の DN Aに対するプロモータ一活性を 促進または阻害する化合物またはその塩のスクリーニング方法を提供する。 本発明の DNAが不活性化された非ヒト哺乳動物胚幹細胞とは、 該非ヒト哺 乳動物が有する本発明の DNAに人為的に変異を加えることにより、 DNAの 発現能を抑制するか、 もしくは該 DNAがコードしている本発明のタンパク質 の活性を実質的に喪失させることにより、 DN Aが実質的に本発明のタンパク 質の発現能を さない (以下、 本発明のノックアウト DNAと称することがあ る) 非ヒト哺乳動物の胚幹細胞 (以下、 ES細胞と略記する) をいう。 (10) A compound that promotes or inhibits the activity of the promoter of DNA of the present invention, which comprises administering a test compound to the animal described in (7) and detecting the expression of a repo overnight gene; A method for screening the salt is provided. A non-human mammalian embryonic stem cell in which the DNA of the present invention has been inactivated refers to a DNA of the present invention possessed by the non-human mammal, which is artificially mutated to suppress the expression ability of the DNA, or By substantially losing the activity of the protein of the present invention encoded by the DNA, the DNA substantially does not have the ability to express the protein of the present invention (hereinafter, referred to as the knockout DNA of the present invention). Refers to non-human mammal embryonic stem cells (hereinafter abbreviated as ES cells).
非ヒト哺乳動物としては、 前記と同様のものが用いられる。  As the non-human mammal, the same one as described above is used.
本発明の DNAに人為的に変異を加える方法としては、 例えば、 遺伝子工学 的手法により該 DN A配列の一部又は全部の削除、 他 DNAを挿入または置換 させることによって行なうことができる。 これらの変異により、 例えば、 コド ンの読み取り枠をずらしたり、 プロモータ一あるいはェキソンの機能を破壊す ることにより本発明のノックアウト D N Aを作製すればよい。  The method of artificially mutating the DNA of the present invention can be performed, for example, by deleting a part or all of the DNA sequence and inserting or substituting another DNA by a genetic engineering technique. By these mutations, for example, the knockout DNA of the present invention may be prepared by shifting the reading frame of codons or disrupting the function of the promoter or exon.
本発明の DN Aが不活性化された非ヒ卜哺乳動物胚幹細胞 (以下、 本発明の D N A不活性化 E S細胞または本発明のノックアウト E S細胞と略記する) の 具体例としては、 例えば、 目的とする非ヒト哺乳動物が有する本発明の DNA を単離し、 そのェキソン部分にネオマイシン耐性遺伝子、 八イダロマイシン耐 性遺伝子を代表とする薬剤耐性遺伝子、 あるいは l a c Z ( 3—ガラクトシダ ーゼ遺伝子) 、 c a t (クロラムフエニコールァセチルトランスフェラ一ゼ遺 伝子) を代表とするレポ一夕一遺伝子等を挿入することによりェキソンの機能 を破壊するか、 あるいはェキソン間のイントロン部分に遺伝子の転写を終結さ せる DNA配列 (例えば、 p o 1 yA付加シグナルなど) を挿入し、 完全なメ ッセンジャー RN Aを合成できなくすることによって、 結果的に遺伝子を破壊 するように構築した DNA配列を有する DNA鎖 (以下、 夕ーゲッティングべ クタ一と略記する) を、 例えば相同組換え法により該動物の染色体に導入し、 得られた ES細胞について本発明の DN A上あるいはその近傍の DN A配列を プロープとしたサザンハイブリダィゼーション解析あるいはターゲッティング ベクター上の DNA配列とターゲッティングベクタ一作製に使用した本発明の D N A以外の近傍領域の D N A配列をプライマ一とした P C R法により解析 し、 本発明のノックアウト ES細胞を選別することにより得ることができる。 また、 相同組換え法等により本発明の DNAを不活化させる元の ES細胞と しては、 例えば、 前述のような既に樹立されたものを用いてもよく、 また公知 Evansと Kaufmaの方法に準じて新しく樹立したものでもよい。 例えば、 マウスの ES細胞の場合、 現在、 一般的にば 129系の ES細胞が使用されているが、 免疫学的背景がはっきりしていないので、 これに代わる純系で免疫学的に遺伝 的背景が明らかな ES細胞を取得するなどの目的で例えば、 C 57BLZ6マ ウスや C 57 BL/ 6の採卵数の少なさを DBAZ 2との交雑により改善した BDFtマウス (C 57 BLZ6と DBAZ2との を用いて樹立したもの なども良好に用いうる。 BDFiマウスは、 採卵数が多く、 かつ、 卵が丈夫で あるという利点に加えて、 C 57BLZ6マウスを背景に持つので、 これを用 いて得られた ES細胞は病態モデルマウスを作出したとき、 C 57BLZ6マ ウスとバッククロスすることでその遺伝的背景を C 57 B LZ 6マウスに代え ることが可能である点で有利に用い得る。 また、 E S細胞を樹立する場合、 一般には受精後 3 . 5日目の胚盤胞を使用す るが、 これ以外に 8細胞期胚を採卵し胚盤胞まで培養して用いることにより効 率よく多数の初期胚を取得することができる。 Non-human mammalian embryonic stem cells in which the DNA of the present invention has been inactivated (hereinafter referred to as the present invention) Specific examples of the DNA-inactivated ES cells or the knockout ES cells of the present invention) include, for example, isolating the DNA of the present invention from a non-human mammal of interest, and adding a neomycin resistance gene to its exon portion. A drug resistance gene typified by the idalomycin resistance gene, or a repo allele gene typified by lacZ (3-galactosidase gene) and cat (chloramphenicol acetyltransferase gene) Or the insertion of a DNA sequence that terminates gene transcription (for example, a po1yA addition signal) into the intron between the exons, resulting in a complete messenger RNA. A DNA strand having a DNA sequence constructed so as to result in the disruption of the gene by preventing the synthesis of ) Is introduced into the chromosome of the animal by, for example, homologous recombination, and the obtained ES cells are subjected to Southern hybridization using the DNA sequence on or near the DNA of the present invention as a probe. The knockout ES cells of the present invention are selected by analyzing the DNA sequence on the targeting vector and the DNA sequence of the neighboring region other than the DNA of the present invention used for the preparation of the targeting vector by using the primers as primers. Can be obtained by As the ES cells from which the DNA of the present invention is inactivated by the homologous recombination method or the like, for example, those already established as described above may be used, or the method described in the known Evans and Kaufma method may be used. A newly established one may be used. For example, in the case of mouse ES cells, generally, 129 ES cells are currently used, but since the immunological background is not clear, it is a pure line that substitutes the immunological genetic background. For example, for the purpose of obtaining ES cells for which C57BLZ6 and C57BLZ6 mice have a low number of eggs, the BDFt mice (C57BLZ6 and DBAZ2 BDFi mice can be used satisfactorily because they have the advantage of high number of eggs collected and robust eggs, and have C57BLZ6 mice as a background. ES cells can be advantageously used when a pathological model mouse is created, because the genetic background can be replaced by C57BLZ6 mice by backcrossing with C57BLZ6 mice. In addition, when ES cells are established, blastocysts 3.5 days after fertilization are generally used, but efficiency is also improved by collecting embryos at the 8-cell stage and culturing them up to blastocysts. Many early embryos can be obtained well.
また、 雌雄いずれの E S細胞を用いてもよいが、 通常雄の E S細胞の方が生 殖系列キメラを作出するのに都合が良い。 また、 煩雑な培養の手間を削減する ためにもできるだけ早く雌雄の判別を行なうことが望ましい。  Either male or female ES cells may be used, but male ES cells are generally more convenient for producing a breeding line chimera. It is also desirable to discriminate between males and females as soon as possible in order to reduce the complexity of culturing.
E S細胞の雌雄の判定方法としては、 例えば、 P C R法により Y染色体上の 性決定領域の遺伝子を増幅、 検出する方法が、 その 1例としてあげることがで きる。 この方法を使用すれば、 従来、 核型分析をするのに約 1 0 6個の細胞数 を要していたのに対して、 1コロニー程度の E S細胞数 (約 5 0個) で済むの で、 培養初期における E S細胞の第一次セレクションを雌雄の判別で行なうこ とが可能であり、 早期に雄細胞の選定を可能にしたことにより培養初期の手間 は大幅に削減できる。 An example of a method for determining the sex of ES cells is a method of amplifying and detecting a gene in the sex-determining region on the Y chromosome by PCR. Using this method, conventionally, for example G-banding method, it requires about 1 0 6 cells for karyotype analysis, the need in ES cell number of about 1 colony (about 5 0) Therefore, the primary selection of ES cells in the early stage of culture can be performed by discriminating between male and female, and the early stage of culture can be greatly reduced by enabling the selection of male cells at an early stage.
また、 第二次セレクションとしては、 例えば、 G—バンデイング法による染 色体数の確認等により行うことができる。 得られる E S細胞の染色体数は正常 数の 1 0 0 %が望ましいが、 樹立の際の物理的操作等の関係上困難な場合は、 E S細胞の遗伝子をノックアウトした後、 正常細胞 (例えば、 マウスでは染色 体数が 2 n = 4 0である細胞) に再びクローニングすることが望ましい。  The secondary selection can be performed, for example, by confirming the number of chromosomes by the G-banding method. The number of chromosomes in the obtained ES cells is preferably 100% of the normal number, but if it is difficult due to physical operations at the time of establishment, knock out the ES cell gene and On the other hand, it is desirable to clone again into cells in which the number of chromosomes is 2 n = 40 in mice).
このようにして得られた胚幹細胞株は、 通常その増殖性は大変良いが、 個体 発生できる能力を失いやすいので、 注意深く継代培養することが必要である。 例えば、 S T O繊維芽細胞のような適当なフィーダ一細胞上で L I F (1〜 10000U/ml) 存在下に炭酸ガス培養器内 (好ましくは、 5 %炭酸ガス、 9 5 %空 気または 5 %酸素、 5 %炭酸ガス、 9 0 %空気) で約 3 7 で培養するなどの 方法で培養し、 継代時には、 例えば、 トリプシン ZE D TA溶液 (通常 0. 001〜 0. 5%トリプシン ZO. l〜5mM EDTA、 好ましくは約 0. 1 %トリプシン/ ImM EDTA) 処理により単細胞化し、 新たに用意したフィ一ダー細胞上に播種する方法など がとられる。 このような継代は、 通常 1一 3日毎に行なうが、 この際に細胞の 観察を行い、 形態的に異常な細胞が見受けられた場合はその培養細胞は放棄す ることが望まれる。 ES細胞は、 適当な条件により、 高密度に至るまで単層培養するか、 または 細胞集塊を形成するまで浮遊培養することにより、 頭頂筋、 内臓筋、 心筋など の種々のタイプの細胞に分化させることが可能であり 〔M. J. Evans及び M. H. Kaufman, ネィチヤ一 (Nature) 第 292卷、 154頁、 1981年; G. R. Martin プロ シーディングス ·ォブ ·ナショナル ·アカデミー 'ォブ ·サイエンス ·ユーェ スェ一 (Proc. Natl. Acad. Sci. U.S.A.) 第 78巻、 7634頁、 1981年; T. C. Doetschman ら、 ジャーナル ·ォブ.ェンブリオ口ジ一 .アンド .ェクスぺリメ ンタル ·モルフォロジ一、 第 87巻、 27頁、 1985年〕 、 本発明の ES細胞を分化 させて得られる本発明の DN A発現不全細胞は、 インビト口における本発明の 夕ンパク質の細胞生物学的検討において有用である。 Embryonic stem cell lines obtained in this way usually have very good proliferative potential, but must be carefully subcultured because they tend to lose their ontogenetic potential. For example, on a suitable feeder cell, such as STO fibroblasts, in a carbon dioxide incubator (preferably 5% carbon dioxide, 95% air or 5% oxygen) in the presence of LIF (1-10000 U / ml) , 5% CO 2, 90% air) at about 37. At the time of subculture, for example, trypsin ZEDTA solution (usually 0.001 to 0.5% trypsin ZO. L) To 5 mM EDTA (preferably, about 0.1% trypsin / ImM EDTA), and seeding the cells on freshly prepared feeder cells. Such subculture is usually performed every 11 to 13 days. At this time, it is desirable to observe the cells and discard the cultured cells if morphologically abnormal cells are found. ES cells are differentiated into various types of cells, such as parietal, visceral, and cardiac muscle, by monolayer culture up to high density or suspension culture until cell clumps are formed under appropriate conditions. [MJ Evans and MH Kaufman, Nature, Vol. 292, pp. 154, 1981; GR Martin Proceedings of the National Academy of Sciences' Ob Science Sciences Proc. Natl. Acad. Sci. USA) 78, 7634, 1981; TC Doetschman et al., Journal of Embrio Morphology, Vol. 87, p. 27. 1985]. The DNA-deficient cells of the present invention obtained by differentiating the ES cells of the present invention are useful in the cell biology of the protein of the present invention in the mouth of in vivo.
本発明の DNA発現不全非ヒト哺乳動物は、 該動物の mRN A量を公知方法 を用いて測定して間接的にその発現量を比較することにより、 正常動物と区別 することが可能である。  The non-human mammal deficient in DNA expression of the present invention can be distinguished from a normal animal by measuring the mRNA amount of the animal using a known method and indirectly comparing the expression levels.
該非ヒト哺乳動物としては、 前記と同様のものが用いられる。  As the non-human mammal, the same one as described above is used.
本発明の DNA発現不全非ヒト哺乳動物は、 例えば、 前述のようにして作製 した夕ーゲッティングベクターをマウス胚幹細胞またはマウス卵細胞に導入 し、 導入により夕一ゲッティングベクタ一の本発明の D N Aが不活性化された DNA配列が遺伝子相同組換えにより、 マウス胚幹細胞またはマウス卵細胞の 染色体上の本発明の DN Aと入れ換わる相同組換えをさせることにより、 本発 明の DNAをノックアウトさせることができる。  The non-human mammal deficient in expression of the DNA of the present invention may be obtained, for example, by introducing the evening-getting vector prepared as described above into a mouse embryonic stem cell or a mouse egg cell, and introducing the evening-getting vector into the DNA of the present invention. Knockout of the DNA of the present invention by homologous recombination in which the DNA sequence in which the DNA has been inactivated replaces the DNA of the present invention on the chromosome of mouse embryonic stem cells or mouse egg cells by gene homologous recombination. Can be.
本発明の DN Aがノックアウトされた細胞は、 本発明の DN A上またはその 近傍の DN A配列をプローブとしたサザンハイブリダイゼーション解析または 夕一ゲッティングベクタ一上の DN A配列と、 タ一ゲッティングベクターに使 用したマウス由来の本発明の DN A以外の近傍領域の DN A配列とをプライマ 一とした PCR法による解析で判定することができる。 非ヒ卜哺乳動物胚幹細 胞を用いた場合は、 遺伝子相同組換えにより、 本発明の DNAが不活性化され た細胞株をクローニングし、 その細胞を適当な時期、 例えば、 8細胞期の非ヒ ト哺乳動物胚または胚盤胞に注入し、 作製したキメラ胚を偽妊娠させた該非ヒ ト哺乳動物の子宮に移植する。 作出された動物は正常な本発明の DN A座をも つ細胞と人為的に変異した本発明の D NA座をもつ細胞との両者から構成され るキメラ動物である。 The cells in which the DNA of the present invention has been knocked out are subjected to Southern hybridization analysis using the DNA sequence on or near the DNA of the present invention as a probe or the DNA sequence on the overnight getter vector, and the target DNA. The DNA can be determined by a PCR method using the DNA sequence of a neighboring region other than the DNA of the present invention derived from the mouse used as the primer vector as a primer. When a non-human mammalian embryonic stem cell is used, a cell line in which the DNA of the present invention has been inactivated by gene homologous recombination is cloned, and the cell is used at an appropriate time, for example, at the 8-cell stage. The chimeric embryo is injected into a non-human mammal embryo or blastocyst, and the resulting chimeric embryo is transplanted into the uterus of the pseudo-pregnant non-human mammal. The produced animal also has a normal DNA locus of the present invention. This is a chimera animal composed of both an artificial cell and an artificially mutated cell having the DNA locus of the present invention.
該キメラ動物の生殖細胞の一部が変異した本発明の D N A座をもつ場合、 こ のようなキメラ個体と正常個体を交配ずることにより得られた個体群より、 全 ての組織が人為的に変異を加えた本発明の D N A座をもつ細胞で構成された個 体を、 例えば、 コートカラーの判定等により選別することにより得られる。 こ のようにして得られた個体は、 通常、 本発明のタンパク質のヘテロ発現不全個 体であり、 本発明のタンパク質のヘテロ発現不全個体同志を交配し、 それらの 産仔から本発明のタンパク質のホモ発現不全個体を得ることができる。  When a part of the germ cells of the chimeric animal has a mutated DNA locus of the present invention, all tissues are artificially obtained from the population obtained by crossing such a chimeric individual with a normal individual. It can be obtained by selecting individuals composed of cells having the DNA locus of the present invention to which a mutation has been added, for example, by judging coat color. The individual obtained in this manner is usually an individual having a heterozygous expression of the protein of the present invention, which is crossed with an individual having a heterozygous expression of the protein of the present invention. A homozygous deficient individual can be obtained.
卵細胞を使用する場合は、 例えば、 卵細胞核内にマイクロインジェクション 法で D N A溶液を注入することにより夕ーゲッティングベクターを染色体内に 導入したトランスジエニック非ヒト哺乳動物を得ることができ、 これらのトラ ンスジエニック非ヒト哺乳動物に比べて、 遺伝子相同組換えにより本発明の D When using an egg cell, for example, a transgenic non-human mammal having a chromosome into which the evening-getting vector has been introduced can be obtained by injecting a DNA solution into the nucleus of the egg cell by a microinjection method. Compared to transgenic non-human mammals, the homologous recombination of the D
N A座に変異のあるものを選択することにより得られる。 Obtained by selecting those with mutations at the NA locus.
このようにして本発明の D NAがノックアウトされている個体は、 交配によ り得られた動物個体も該 D N Aがノックァゥトされていることを確認して通常 の飼育環境で飼育継代を行なうことができる。  When the DNA of the present invention is knocked out in this manner, the animal obtained by the crossing should be confirmed to be knocked out of the DNA and subjected to rearing in a normal rearing environment. Can be.
さらに、 生殖系列の取得および保持についても常法に従えばよい。 すなわ ち、 該不活化 D NAの保有する雌雄の動物を交配することにより、 該不活化 D N Aを相同染色体の両方に持つホモザィゴート動物を取得しうる。 得られたホ モザィゴート動物は、 母親動物に対して、 正常個体 1, ホモザィゴート複数に なるような状態で飼育することにより効率的に得ることができる。 ヘテロザィ ゴート動物の雌雄を交配することにより、 該不活化 D N Aを有するホモザィゴ Furthermore, the germline can be obtained and maintained according to a conventional method. That is, by crossing male and female animals having the inactivated DNA, homozygous animals having the inactivated DNA on both homologous chromosomes can be obtained. The obtained homozygous animal can be efficiently obtained by rearing the mother animal in such a manner that one normal individual and a plurality of homozygous animals are obtained. By mating male and female heterozygous goats, a homozygote having the inactivated DNA is obtained.
―トおよびへテロザィゴート動物を繁殖継代する。 -Breeding and breeding wild and heterozygous animals.
本発明の D NAが不活性化された非ヒト哺乳動物胚幹細胞は、 本発明の D N The non-human mammalian embryonic stem cells in which the DNA of the present invention has been inactivated are the DN of the present invention.
A発現不全非ヒト哺乳動物を作出する上で、 非常に有用である。 It is very useful in creating non-human mammals deficient in A expression.
また、 本発明の D NA発現不全非ヒ卜哺乳動物は、 本発明のタンパク質によ り誘導され得る種々の生物活性を欠失するため、 本発明のタンパク質の生物活 性の不活性化を原因とする疾病のモデルとなり得るので、 これらの疾病の原因 究明及び治療法の検討に有用である。 In addition, since the non-human mammal deficient in expression of the DNA of the present invention lacks various biological activities that can be induced by the protein of the present invention, the non-human mammal may be inactivated by the inactivation of the biological activity of the protein of the present invention. Can be a model of the disease It is useful for investigation and treatment.
( 7 a ) 本発明の D NAの欠損や損傷などに起因する疾病に対して治療 ·予防 効果を有する化合物のスクリーニング方法  (7a) A method for screening a compound having a therapeutic / preventive effect against diseases caused by DNA deficiency or damage of the present invention
本発明の D N A発現不全非ヒト哺乳動物は、 本発明の D N Aの欠損や損傷な どに起因する疾病に対して治療 ·予防効果を有する化合物のスクリーニングに 用いることができる。  The non-human mammal deficient in DNA expression of the present invention can be used for screening for a compound having a therapeutic / preventive effect against diseases caused by DNA deficiency or damage of the present invention.
すなわち、 本発明は、 本発明の D NA発現不全非ヒト哺乳動物に試験化合物 を投与し、 該動物の変化を観察 ·測定することを特徵とする、 本発明の D NA の欠損や損傷などに起因する疾病、 例えば、 腎疾患 (例、 糖尿病性腎症、 巣状 糸球体硬化症、 慢性糸球体腎炎、 微小変化型ネフ口一ゼ症候群、 メサンギゥム 増殖性腎炎、 管内増殖性腎炎、 IgA腎症、 膜性腎症、 膜性増殖性腎炎、 半月体形 成性腎炎、 ループス腎炎、 アミロイド腎、 尿細管間質性腎炎、 急性尿細管壊 死、 急性腎不全、 腎硬化症、 腎性浮腫、 腎腫瘍、 腎移植後の慢性拒絶など) な どに対して治療 ·予防効果を有する化合物またはその塩のスクリ一ニング方法 を提供する。  That is, the present invention is characterized in that a test compound is administered to a non-human mammal deficient in expression of the DNA of the present invention, and changes in the animal are observed and measured. Diseases caused by, for example, renal diseases (eg, diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimally altered nephrotic syndrome, mesangium, proliferative nephritis, endoproliferative nephritis, IgA nephropathy , Membranous nephropathy, membranous proliferative nephritis, crescentic adult nephritis, lupus nephritis, amyloid kidney, tubular interstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, kidney To provide a method for screening a compound or a salt thereof having a therapeutic / preventive effect on tumors, chronic rejection after kidney transplantation, etc.).
該スクリーニング方法において用いられる本発明の D NA発現不全非ヒト哺 乳動物としては、 前記と同様のものがあげられる。  Examples of the non-human mammal deficient in DNA expression of the present invention used in the screening method include those described above.
試験化合物としては、 例えば、 ペプチド、 タンパク、 非ペプチド性化合物、 合成化合物、 発酵生産物、 細胞抽出液、 植物抽出液、 動物組織抽出液、 血漿な どがあげられ、 これら化合物は新規な化合物であつてもよいし、 公知の化合物 であってもよい。  Test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and plasma.These compounds are novel compounds. Or a known compound.
具体的には、 本発明の D NA発現不全非ヒ卜哺乳動物を、 試験化合物で処理 し、 無処理の対照動物と比較し、 該動物の各器官、 組織、 疾病の症状などの変 化を指標として試験化合物の治療 ·予防効果を試験することができる。  Specifically, a non-human mammal deficient in expression of the DNA of the present invention is treated with a test compound and compared with a non-treated control animal to examine changes in organs, tissues, disease symptoms and the like of the animal. The therapeutic / preventive effect of the test compound can be tested as an index.
試験動物を試験化合物で処理する方法としては、 例えば、 経口投与、 静 J ^注 射などが用いられ、 試験動物の症状、 試験化合物の性質などにあわせて適宜選 択することができる。 また、 試験化合物の投与量は、 投与方法、 試験化合物の 性質などにあわせて適宜選択することができる。 例えば糖尿病性腎症などに対して治療 ·予防効果を有する化合物をスクリー エングする場合、 本発明の D NA発現不全非ヒト哺乳動物に試験化合物を投与 し、 試験化合物非投与群と、 腎機能の指標である尿中タンパク排泄量や糸球体 濾過率、 または腎機能障害の指標である糸球体硬化率の違いなどを経時的に観 察する。 As a method of treating a test animal with a test compound, for example, oral administration, intravenous injection, etc. are used, and the method can be appropriately selected according to the symptoms of the test animal, the properties of the test compound, and the like. The dose of the test compound can be appropriately selected according to the administration method, the properties of the test compound, and the like. For example, when screening for a compound having a therapeutic / preventive effect on diabetic nephropathy, etc., a test compound is administered to a non-human mammal deficient in expression of the DNA of the present invention, and the test compound non-administration group and the renal function Observe, over time, differences in urine protein excretion and glomerular filtration rate, which are indicators, or glomerular sclerosis rate, which is an indicator of renal dysfunction.
該スクリーニング方法において、 試験動物に試験化合物を投与した場合、 該 試験動物の上記疾患症状が約 10%以上、 好ましくは約 30%以上、 より好ましく は約 50%以上改善した場合、 該試験化合物を上記の疾患に対して治療 ·予防効 果を有する化合物として選択することができる。  In the screening method, when the test compound is administered to a test animal, the disease symptom of the test animal is improved by about 10% or more, preferably about 30% or more, more preferably about 50% or more. It can be selected as a compound having a therapeutic / preventive effect on the above diseases.
該スクリーニング方法を用いて得られる化合物は、 上記した試験化合物から 選ばれた化合物であり、 本発明のタンパク質の欠損や損傷などによって引き起 こされる疾患に対して治療 ·予防効果を有するので、 該疾患に対する安全で低 毒性な予防 ·治療剤などの医薬として使用することができる。 さらに、 上記ス クリ一ニングで得られた化合物から誘導される化合物も同様に用いることがで きる。  The compound obtained by using the screening method is a compound selected from the test compounds described above, and has a therapeutic / preventive effect against a disease caused by deficiency or damage of the protein of the present invention. It can be used as a medicament such as a safe and low toxic prophylactic / therapeutic agent for the disease. Further, a compound derived from the compound obtained by the above screening can also be used.
該スクリーニング方法で得られた化合物は塩を形成していてもよく、 該化合 物の塩としては、 生理学的に許容される酸 (例、 無機酸、 有機酸など) や塩基 (例、 アルカリ金属など) などとの塩が用いられ、 とりわけ生理学的に許容さ れる酸付加塩が好ましい。 この様な塩としては、 例えば、 無機酸 (例えば、 塩 酸、 リン酸、 臭化水素酸、 硫酸など) との塩、 あるいは有機酸 (例えば、 酢 酸、 ギ酸、 プロピオン酸、 フマル酸、 マレイン酸、 コハク酸、 酒石酸、 クェン 酸、 リンゴ酸、 蓚酸、 安息香酸、 メタンスルホン酸、 ベンゼンスルホン酸な ど) との塩などが用いられる。  The compound obtained by the screening method may form a salt. Examples of the salt of the compound include physiologically acceptable acids (eg, inorganic acids, organic acids, etc.) and bases (eg, alkali metals). And the like, and especially preferred are physiologically acceptable acid addition salts. Examples of such salts include salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.) and organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, etc.). Acid, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc.).
該スクリーニング方法で得られた化合物またはその塩を含有する医薬は、 前 記した本発明のタンパク質を含有する医薬と同様にして製造することができ る。  A drug containing the compound or a salt thereof obtained by the screening method can be produced in the same manner as the above-mentioned drug containing the protein of the present invention.
このようにして得られる製剤は、 安全で低毒性であるので、 例えば、 ヒトま たは哺乳動物 (例えば、 ラット、 マウス、 モルモット、 ゥサギ、 ヒッジ、 ブ タ、 ゥシ、 ゥマ、 ネコ、 ィヌ、 サルなど) に対して投与することができる。 該化合物またはその塩の投与量は、 対象疾患、 投与対象、 投与ルートなどに より差異はあるが、 例えば、 該化合物を経口投与する場合、 一般的に成人 (体 重 60kgとして) の糖尿病性腎症患者においては、 一日につき該ィ匕合物を約 0. 1〜 100mg、 好ましくは約 1. 0〜50mg、 より好ましくは約 1. 0〜20mg投与する。 非経口 的に投与する場合は、 該化合物の 1回投与量は投与対象、 対象疾患などによつ ても異なるが、 例えば、 該化合物を注射剤の形で通常成人 (60kgとして) の糖 尿病性腎症患者に投与する場合、 一日につき該ィヒ合物を約 0. 01〜30mg、 好まし くは約 0. 〜 2 Omg、 より好ましくは約 0. 1〜 1 Omgを静脈注射により投与するのが 好都合である。 他の動物の場合も、 60kg当たりに換算した量を投与することが でさる。 The preparations obtained in this way are safe and have low toxicity, for example, in humans or mammals (eg, rats, mice, guinea pigs, egrets, sheep, sheep, butter, horseshoe, horses, cats, cats). And monkeys). The dose of the compound or a salt thereof varies depending on the target disease, the subject of administration, the administration route, and the like. For example, when the compound is orally administered, generally the diabetic kidney of an adult (with a body weight of 60 kg) is used. For patients with sickness, about 0.1 to 100 mg, preferably about 1.0 to 50 mg, more preferably about 1.0 to 20 mg of the compound is administered per day. In the case of parenteral administration, the single dose of the compound varies depending on the administration subject, target disease and the like. For example, the compound is usually in the form of an injection and is usually used for adult (as 60 kg) sugar urine. When administered to patients with pathologic nephropathy, about 0.01 to 30 mg, preferably about 0.1 to 2 Omg, and more preferably about 0.1 to 1 Omg of the compound is administered intravenously per day. It is conveniently administered by In the case of other animals, it is also possible to administer the amount converted per 60 kg.
( 7 b ) 本発明の D N Aに対するプロモータ一の活性を促進または阻害するィ匕 合物をスクリーニング方法  (7b) A method for screening a conjugate which promotes or inhibits the activity of a promoter for DNA of the present invention
本発明は、 本発明の D NA発現不全非ヒト哺乳動物に、 試験化合物を投与 し、 レポ一夕一遗伝子の発現を検出することを特徴とする本発明の D N Aに対 するプロモーターの活性を促進または阻害する化合物またはその塩のスクリ一 ニング方法を提供する。  The present invention relates to the activity of a promoter for the DNA of the present invention, which comprises administering a test compound to a non-human mammal deficient in expressing the DNA of the present invention and detecting the expression of a repo allele. To provide a method for screening a compound or a salt thereof that promotes or inhibits cell growth.
上記スクリーニング方法において、 本発明の D NA発現不全非ヒト哺乳動物 としては、 前記した本発明の D NA発現不全非ヒト哺乳動物の中でも、 本発明 の D N Aがレポ一夕一遺伝子を導入することにより不活性化され、 該レポ一夕 —遺伝子が本発明の D N Aに対するプロモーターの制御下で発現しうるものが 用いられる。  In the above screening method, the non-human mammal deficient in expression of the DNA of the present invention includes, among the aforementioned non-human mammals deficient in the expression of the DNA of the present invention, the DNA of the present invention obtained by introducing a repo overnight gene. Those inactivated and capable of expressing the repo overnight gene under the control of the promoter for the DNA of the present invention are used.
試験化合物としては、 前記と同様のものがあげられる。  Examples of the test compound include the same compounds as described above.
レポーター遺伝子としては、 前記と同様のものが用いられ、 β—ガラクトシ ダーゼ遺伝子 (1 a c Z ) 、 可溶性アルカリフォスファターゼ遺伝子またはル シフェラーゼ遺伝子などが好適である。  As the reporter gene, the same one as described above is used, and a β-galactosidase gene (1 ac Z), a soluble alkaline phosphatase gene or a luciferase gene is preferable.
本発明の D NAをレポ一夕一遺伝子で置換された本発明の D N A発現不全非 ヒト哺乳動物では、 レポーター遺伝子が本発明の D N Aに対するプロモ一夕一 の支配下に存在するので、 レポーター遺伝子がコ一ドする物質の発現をトレー スすることにより、 プロモーターの活性を検出することができる。 例えば、 本発明のタンパク質をコードする D N A領域の一部を大腸菌由来の iS _ガラクトシダ一ゼ遺伝子 (1 a c Z ) で置換している場合、 本来、 本発明 のタンパク質の発現する組織で、 本発明のタンパク質の代わりに 3—ガラクト シダ一ゼが発現する。 従って、 例えば、 5—ブロモー 4—クロ口— 3—インド リル一 j8—ガラクトピラノシド (X—g a l ) のような i3—ガラクトシダ一ゼ の基質となる試薬を用いて染色することにより、 簡便に本発明のタンパク質の 動物生体内における発現状態を観察することができる。 具体的には、 本発明の タンパク質欠損マウスまたはその組織切片をダルタルアルデヒドなどで固定 し、 リン酸緩衝生理食塩液 ( P B S ) で洗浄後、 X— g a 1を含む染色液で、 室温または 3 7 °C付近で、 約 3 0分ないし 1時間反応させた後、 組織標本を 1 mM E D TAZ P B S溶液で洗浄することによって、 /3 _ガラクトシダーゼ反 応を停止させ、 呈色を観察すればよい。 また、 常法に従い、 l a c Zをコード する mR N Aを検出してもよい。 In a non-human mammal deficient in DNA expression of the present invention in which the DNA of the present invention has been replaced with a reporter overnight gene, the reporter gene is under the control of the promoter of the DNA of the present invention. By tracing the expression of the coding substance, the activity of the promoter can be detected. For example, when a part of the DNA region encoding the protein of the present invention is replaced by the iS_galactosidase gene (1 ac Z) derived from Escherichia coli, a tissue that originally expresses the protein of the present invention may 3-galactosidase is expressed instead of this protein. Therefore, for example, by staining with a reagent that serves as a substrate for i3-galactosidase, such as 5-bromo-4-chloro-3-indolyl-j8-galactopyranoside (X-gal), it is convenient In addition, the expression state of the protein of the present invention in an animal body can be observed. Specifically, the protein-deficient mouse of the present invention or a tissue section thereof is fixed with dartalaldehyde or the like, washed with phosphate buffered saline (PBS), and then stained with X-ga1 at room temperature or at room temperature. After reacting at about 7 ° C for about 30 minutes to 1 hour, the / 3_-galactosidase reaction can be stopped by washing the tissue specimen with 1 mM ED TAZ PBS solution, and the coloration can be observed. . In addition, mRNA encoding lacZ may be detected according to a conventional method.
上記スクリーニング方法を用いて得られる化合物またはその塩は、 上記した 試験化合物から選ばれた化合物であり、 本発明の D NAに対するプロモーター 活性を促進または阻害する化合物である。  The compound or a salt thereof obtained by using the above-mentioned screening method is a compound selected from the above-mentioned test compounds, and is a compound that promotes or inhibits the promoter activity for DNA of the present invention.
該スクリーニング方法で得られた化合物は塩を形成していてもよく、 該化合 物の塩としては、 生理学的に許容される酸 (例、 無機酸、 有機酸) や塩基  The compound obtained by the screening method may form a salt. Examples of the salt of the compound include physiologically acceptable acids (eg, inorganic acids, organic acids) and bases.
(例、 アルカリ金属など) などとの塩が用いられ、 とりわけ生理学的に許容さ れる酸付加塩が好ましい。 この様な塩としては、 例えば、 無機酸 (例えば、 塩 酸、 リン酸、 臭化水素酸、 硫酸など) との塩、 あるいは有機酸 (例えば、 酢 '酸、 ギ酸、 プロピオン酸、 フマル酸、 マレイン酸、 コハク酸、 酒石酸、 クェン 酸、 リンゴ酸、 蓚酸、 安息香酸、 メタンスルホン酸、 ベンゼンスルホン酸な ど) との塩などが用いられる。  (Eg, alkali metals) and the like, and physiologically acceptable acid addition salts are particularly preferable. Such salts include, for example, salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.) or organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, Salts such as maleic acid, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc. are used.
本発明の D NAに対するプロモーター活性を促進または阻害 (好ましくは阻 害) する化合物またはその塩は、 本発明のタンパク質の発現の調節、 該タンパ ク質の機能を調節することができるので、 例えば、 腎疾患 (例、 糖尿病性腎 症、 巣状糸球体硬化症、 慢性糸球体腎炎、 微小変化型ネフローゼ症候群、 メサ ンギゥム増殖性腎炎、 管内増殖性腎炎、 IgA腎症、 膜性腎症、 膜性増殖性腎炎、 半月体形成性腎炎、 ループス腎炎、 アミロイド腎、 尿細管間質性腎炎、 急性尿 細管壊死、 急性腎不全、 腎硬化症、 腎性浮腫、 腎腫瘍、 腎移植後の慢性拒絶な ど) などの予防'治療剤として有用である。 A compound or a salt thereof that promotes or inhibits (preferably inhibits) the promoter activity of the DNA of the present invention can regulate the expression of the protein of the present invention and regulate the function of the protein. Kidney disease (e.g., diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome, mesangial proliferative nephritis, endoproliferative nephritis, IgA nephropathy, membranous nephropathy, membranous Proliferative nephritis, Crescentic nephritis, lupus nephritis, amyloid kidney, tubular interstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, renal tumors, chronic rejection after kidney transplantation, etc.) Useful as a prophylactic 'therapeutic.
さらに、 上記スクリーニングで得られた化合物から誘導される化合物も同様 に用いることができる。 .  Further, compounds derived from the compounds obtained by the above screening can be used in the same manner. .
該スクリーニング方法で得られた化合物またはその塩を含有する医薬は、 前 記した本発明のタンパク質またはその塩を含有する医薬と同様にして製造する ことができる。  A drug containing the compound or a salt thereof obtained by the screening method can be produced in the same manner as the above-mentioned drug containing the protein of the present invention or a salt thereof.
このようにして得られる製剤は、 安全で低毒性であるので、 例えば、 ヒトま たは哺乳動物 (例えば、 ラット、 マウス、 モルモット、 ゥサギ、 ヒッジ、 ブ 夕、 ゥシ、 ゥマ、 ネコ、 ィヌ、 サルなど) に対して投与することができる。 該化合物またはその塩の投与量は、 対象疾患、 投与対象、 投与ルー卜などに より差異はあるが、 例えば、 本発明の D NAに対するプロモーター活性を阻害 する化合物を経口投与する場合、 一般的に成人 (体重 60kgとして) の糖尿病性 腎症患者においては、 一日につき該化合物を約 0. l〜100mg、 好ましくは約 1. 0〜 50nig、 より好ましくは約 1. 0〜20mg投与する。 非経口的に投与する場合は、 該化 合物の 1回投与量は投与対象、 対象疾患などによっても異なるが、 例えば、 本 発明の D N Aに対するプロモーター活性を阻害する化合物を注射剤の形で通常 成人 (60kgとして) の糖尿病性腎症患者に投与する場合、 一日につき該化合物 を約 0. 01〜30mg、 好ましくは約 0. l〜20mg、 より好ましくは約 0. 1〜 1 Onigを静脈 注射により投与するのが好都合である。 他の動物の場合も、 60kg当たりに換算 した量を投与することができる。  The preparations obtained in this way are safe and low toxic, for example, in humans or mammals (e.g., rats, mice, guinea pigs, egrets, sheep, sheep, horses, horses, cats, cats, cats). And monkeys and monkeys). The dose of the compound or a salt thereof varies depending on the target disease, the subject of administration, the route of administration, and the like.For example, when the compound of the present invention that inhibits the promoter activity against DNA is orally administered, it is generally used. In an adult (assuming a body weight of 60 kg) diabetic nephropathy, about 0.1 to 100 mg, preferably about 1.0 to 50 nig, more preferably about 1.0 to 20 mg of the compound is administered per day. When administered parenterally, the single dose of the compound varies depending on the administration subject, target disease, and the like.For example, the compound of the present invention that inhibits the promoter activity against DNA is usually in the form of an injection. When administered to an adult (as 60 kg) diabetic nephropathy patient, about 0.01 to 30 mg, preferably about 0.1 to 20 mg, more preferably about 0.1 to 1 Onig of the compound per day is administered intravenously. It is convenient to administer by injection. In the case of other animals, the dose can be administered in terms of 60 kg.
このように、 本発明の D NA発現不全非ヒト哺乳動物は、 本発明の D NAに 対するプロモータ一の活性を促進または阻害する化合物またはその塩をスクリ 一二ングする上で極めて有用であり、 本発明の D N A発現不全に起因する各種 疾患の原因究明または予防 ·治療剤の開発に大きく貢献することができる。 また、 本発明のタンパク質のプロモータ一領域を含有する D N Aを使って、 その下流に種々のタンパクをコードする遺伝子を連結し、 これを動物の卵細胞 に注入していわゆるトランスジエニック動物 (遺伝子移入動物) を作成すれ ば、 特異的にそのタンパク質を合成させ、 その生体での作用を検討することも. 可能となる。 さらに上記プロモーター部分に適当なレポーター遺伝子を結合さ せ、 これが発現するような細胞株を樹立すれば、 本発明のタンパク質そのもの の体内での産生能力を特異的に促進もしくは抑制する作用を持つ低分子化合物 の探索系として使用できる。 本明細書において、 塩基やアミノ酸などを略号で表示する場合、 IUPAC-IUB Commission on Biochemical Nomenclature による略号あるいは当該分野におけ る慣用略号に基づくものであり、 その例を下記する。 またアミノ酸に関し光学 異性体があり得る場合は、 特に明示しなければ L体を示すものとする。 As described above, the non-human mammal deficient in DNA expression of the present invention is extremely useful for screening a compound or a salt thereof that promotes or inhibits the activity of the promoter of DNA of the present invention, The present invention can greatly contribute to the investigation of the cause of various diseases caused by DNA expression deficiency or the development of preventive and therapeutic agents. In addition, using a DNA containing a promoter region of the protein of the present invention, genes encoding various proteins are ligated downstream thereof and injected into egg cells of an animal to produce a so-called transgenic animal (transgenic animal). ) For example, it would be possible to specifically synthesize the protein and examine its action in living organisms. Furthermore, by binding an appropriate reporter gene to the above promoter portion and establishing a cell line capable of expressing the same, a low molecule having an action of specifically promoting or suppressing the production ability of the protein of the present invention itself in the body. It can be used as a search system for compounds. In the present specification, bases, amino acids, and the like are indicated by abbreviations based on the abbreviations by the IUPAC-IUB Commission on Biochemical Nomenclature or commonly used abbreviations in the relevant field, and examples thereof are described below. When amino acids can have optical isomers, the L-form is indicated unless otherwise specified.
DNA :デォキシリポ核酸  DNA: Deoxylipo nucleic acid
c DNA :相補的デォキシリポ核酸  c DNA: Complementary deoxylipo nucleic acid
A :アデニン  A: Adenine
T :チミン  T: Thymine
G : グァニン  G: Guanin
C : シ卜シン  C: Shitoshin
RNA : リボ核酸  RNA: ribonucleic acid
mRNA : メッセンジャーリボ核酸  mRNA: messenger ribonucleic acid
dATP :デォキシアデノシン三リン酸  dATP: Deoxyadenosine triphosphate
dTTP :デォキシチミジン三リン酸  dTTP: Deoxythymidine triphosphate
dGTP :デォキシグアノシン三リン酸  dGTP: Deoxyguanosine triphosphate
d CTP :デォキシシチジン三リン酸  d CTP: Deoxycytidine triphosphate
ATP :アデノシン三リン酸  ATP: Adenosine triphosphate
EDTA :エチレンジァミン四酢酸  EDTA: Ethylenediaminetetraacetic acid
SDS : ドデシル硫酸ナトリウム  SDS: Sodium dodecyl sulfate
G 1 y :グリシン  G 1 y: glycine
A 1 a :ァラニン  A 1 a: Alanine
V a 1 :パリン  V a 1: Palin
Le u : ロイシン I 1 e :イソロイシン Le u: Leucine I 1 e: Isoleucine
S e r :セリン  S e r: Serine
T r :スレオニン  T r: Threonine
Cy s :システィン  Cy s: Cystine
Me t :メチォニン  Me t: Methionin
G 1 u :グルタミン酸  G 1 u: Glutamic acid
As :ァスパラギン酸  As: Aspartic acid
L y s : リジン  L y s: lysine
A r g :アルギニン  A r g: Arginine
H i s : ヒスチジン  H i s: Histidine
Ph e :フエニルァラニン  Ph e: Phenylalanine
Ty r :チロシン  Ty r: Tyrosine
T r p : 卜リブトフアン  T r p: Tributofuan
P r o :プロリン  Pro: Proline
A s n :ァスパラギン  A s n: Asparagine
G 1 n : グル夕ミン  G 1 n: Guru Yu Min
P G 1 u : ピログルタミン酸  P G 1 u: Pyroglutamic acid
S e c :セレノシスティン (selenocysteine) また、 本明細 f中で繁用される置換基  S e c: selenocysteine Also, a substituent frequently used in this specification f
記する。 Write.
Me メチル基  Me methyl group
E t ェチル基  Etethyl group
Bu ブチル基  Bu butyl group
Ph フエニル基  Ph phenyl group
TC チアゾリジン一 4 (R) 一力ルポキサミド基 To s p—トルエンスルフォニル  TC thiazolidine-1 (R) One-pot lipoxamide group To s p-toluenesulfonyl
CHO ホルミル  CHO Holmill
B z 1  B z 1
Cl2- Bzl 2, 6—ジクロロべンジル Bom :ベンジルォキシメチル Cl 2 -Bzl 2, 6-dichlorobenzyl Bom: benzyloxymethyl
Z :ベンジルォキシカルポニル  Z: benzyloxycarponyl
C 1一 z : 2—クロ口べンジルォキシカルボニル  C 1 z: 2-cyclobenzoyloxycarbonyl
B r -Z : 2—ブロモベンジルォキシカルボニル  B r -Z: 2-bromobenzyloxycarbonyl
B o c : t—ブトキシカルポニル  B o c: t-butoxycarponyl
DNP :ジニトロフエニル  DNP: dinitrophenyl
T r t : トリチル  Trt: Trityl
Bum : t—ブトキシメチル  Bum: t-butoxymethyl
Fmo c : N— 9 _フルォレニルメトキシカルポニル  Fmo c: N—9_fluorenylmethoxycarbonyl
HOB t : 1ーヒドロキシベンズトリァゾ一ル  HOB t: 1-hydroxybenztriazole
HOOB t : 3, 4ージヒドロ一 3—ヒドロキシ一4—ォキソ一  HOOB t: 3,4-dihydro-3-hydroxy-1-oxo
1-2, 3一べンゾ卜リアジン  1-2, 3 Benzotriazine
HONB :トヒドロキシ -5-ノルポルネン- 2, 3 -ジカルボキシイミド DC C 本願明細書の配列表の配列番号は、 以下の配列を示す。  HONB: Tohydroxy-5-norporene-2,3-dicarboximide DC C The sequence numbers in the sequence listing in the present specification show the following sequences.
〔配列番号: 1〕  [SEQ ID NO: 1]
ヒト Type lib Na/Pi Cotransporterのアミノ酸酉己列 (GenBank Accession No. AAC98695) を示す。 The amino acid rooster sequence (GenBank Accession No. AAC98695) of human Type lib Na / Pi Cotransporter is shown.
〔配列番号: 2〕  [SEQ ID NO: 2]
ヒト Type lib Na/Pi Cotransporterの塩基配列 (GenBank Accession No. Nucleotide sequence of human Type lib Na / Pi Cotransporter (GenBank Accession No.
AF111856) を示す。 AF111856).
〔配列番号: 3〕  [SEQ ID NO: 3]
ラット Type lib Na/Pi Cotransporterのアミノ酸配列 (GenBank Accession No. AAF76291) を示す。 2 shows the amino acid sequence (GenBank Accession No. AAF76291) of rat Type lib Na / Pi Cotransporter.
〔配列番号: 4〕  [SEQ ID NO: 4]
ラット Type lib Na/Pi Cotransporterの塩基配列 (GenBank Accession No. AF157026) を示す。 The nucleotide sequence of rat Type lib Na / Pi Cotransporter (GenBank Accession No. AF157026) is shown.
〔配列番号: 5〕 - マウス Type lib Na/Pi Cotransporterのアミノ酸配列 (GenBank Accession No. AAC80007) を示す。 [SEQ ID NO: 5]- The amino acid sequence (GenBank Accession No. AAC80007) of mouse Type lib Na / Pi Cotransporter is shown.
〔配列番号: 6〕  [SEQ ID NO: 6]
マウス Type lib Na/Pi Cotransporterの塩基配列 (GenBank Accession No. AF081499) を示す。 The nucleotide sequence of mouse Type lib Na / Pi Cotransporter (GenBank Accession No. AF081499) is shown.
〔配列番号: 7〕  [SEQ ID NO: 7]
実施例 2で用いられたプライマーの塩基配列を示す。 3 shows the nucleotide sequence of a primer used in Example 2.
〔配列番号: 8〕  [SEQ ID NO: 8]
実施例 2で用いられたプライマーの塩基配列を示す。 3 shows the nucleotide sequence of a primer used in Example 2.
〔配列番号: 9〕  [SEQ ID NO: 9]
実施例 2で用いられたプローブの塩基配列を示す。 3 shows the nucleotide sequence of a probe used in Example 2.
〔配列番号: 10〕  [SEQ ID NO: 10]
実施例 3で用いられたプライマーの塩基配列を示す。 3 shows the nucleotide sequence of a primer used in Example 3.
〔配列番号: 11〕  [SEQ ID NO: 11]
実施例 3で用いられたプライマ一の塩基配列を示す。 3 shows the nucleotide sequence of a primer used in Example 3.
〔配列番号: 12〕  [SEQ ID NO: 12]
実施例 3で用いられたプロ一ブの塩基配列を示す。 以下において、 実施例により本発明をより具体的にするが、 この発明はこれ らに限定されるものではない。 3 shows the nucleotide sequence of a probe used in Example 3. Hereinafter, the present invention will be more specifically described with reference to examples, but the present invention is not limited thereto.
実施例 1 Example 1
腎疾患モデルラット腎組織で発現変動するチャネル · トランスポー夕の中か ら創薬標的を発掘する目的で以下のような実験を行った。  The following experiments were performed to discover drug targets from the channels and transport pathways that fluctuate in the kidney tissue of rat kidney model.
疾患モデル動物としては、 Wistar Fatty ラット (WFラッド) 、 Zucker Fatty ラット (ZFラット) 、 自然発症高コレステロール血症ラット (SHCラット) の 3 種類を用い、 それぞれに対応する対照として、 ラットに対しては Wistar Lean Rat (WLラット) 、 ZFラットに対しては Zucker Lean Rat (ZLラット) 、 SHCラッ トに対しては SD Rat (SDラ'ット) を使用した。 疾患モデル動物と対照動物を同 じ条件で飼育し、 ラットおよび WLラットでは生後 13週、 20週、 40週および 68 週に、 ZFラットおよび ZLラットでは生後 8週および 27週に、 SHCラットおよび SD ラットでは生後 6週、 12週、 20週および 30週に、 各群最低 5の個体から腎臓を採 取し、 IS0GEN (二ツボンジーン社) 使用説明書記載の方法に従って個体別に Total RNAを調製した。 調製した試料のうち、 WFラットおよび WLラットでは、 WF ラットで腎障害が進展しつつある生後 40週と、 腎障害がさらに進んで腎機能の 低下が認められる生後 68週の試料を、 ZFラットおよび ZLラットでは、 ZFラット で腎障害が進展しつつある生後 27週の試料を、 SHCラットおよび SDラットでは、 SHCラットで腎機能の低下が認められる生後 30週の試料を以下の網羅的遺伝子発 現解析実験に用いた。 実験構成を表 1に示す。 The disease model animals are Wistar Fatty rat (WF Rad), Zucker Fatty rat (ZF rat), and spontaneously hypercholesterolemic rat (SHC rat). Wistar Lean Rat (WL rat), Zucker Lean Rat (ZL rat) for ZF rat, and SD Rat (SD rat) for SHC rat. Disease model animals and control animals are bred under the same conditions. Rats and WL rats are 13 weeks, 20 weeks, 40 weeks and 68 weeks old. At week 5, kidneys were collected from a minimum of 5 individuals in each group at 8 and 27 weeks of age for ZF and ZL rats and at 6, 12, 20, and 30 weeks of age for SHC and SD rats. Total RNA was prepared for each individual according to the method described in the instruction manual of IS0GEN (Nitsubon Gene). Among the prepared samples, in WF rats and WL rats, samples from the WF rats at the age of 40 weeks, at which renal damage was progressing in the WF rats, and 68 weeks after birth, at which kidney function was further advanced and renal function was reduced, were compared with the ZF rat. For ZL rats and ZL rats, a 27-week-old sample with renal impairment in ZF rats was developed, and for SHC and SD rats, a 30-week-old sample with decreased renal function in SHC rats was used for the following comprehensive genes: It was used for expression analysis experiments. Table 1 shows the experimental configuration.
〔表 1〕  〔table 1〕
Figure imgf000072_0001
本実験では、 各群の個体別腎臓 Total RNAを等量ずつ混合した総量 40 gの Total RNAをひとつの試料として (例えば、 生後 40週の WFラッ卜の場合、 この群 の個体数は 5であるから 1個体当たり 8 n gを混合して総量 40 nigとした) 、 Oligonucleotide microarray (Rat Genome U34A, U34B, U34C; Af fymetr ix社) を用いて遺伝子発現解析を行った。 実験方法は、 Affymetrix社の実験手引き書 (Expression analysis technical manual) に従った。 実験で得られた
Figure imgf000072_0001
In this experiment, a total of 40 g of total RNA obtained by mixing equal amounts of kidney total RNA for each individual in each group was used as one sample (for example, in the case of a WF rat 40 weeks old, the number of individuals in this group is 5 Therefore, gene expression analysis was performed using an Oligonucleotide microarray (Rat Genome U34A, U34B, U34C; Af fymetrix) in which 8 ng per individual was mixed to give a total amount of 40 nig. The experimental method followed Affymetrix's experiment guide (Expression analysis technical manual). Obtained in the experiment
Microarray掲載各配列の発現測定値を Expression analysis software Expression analysis software
(GeneSpring: Silicon Genetics社) で解析し、 発現変動を示す配列を抽出し た。  (GeneSpring: Silicon Genetics) to extract sequences showing expression fluctuations.
その結果、 ラット Type lib Na/Pi Cotransporter (Genbank Accession No. AF157026) が 3種のモデルいずれでも発現亢進していた。 特に SHCラットではラ ッ卜 Type lib Na/Pi Cotransporterの変動が顕著であった。 実施例 2 As a result, the expression of rat Type lib Na / Pi Cotransporter (Genbank Accession No. AF157026) was enhanced in all three models. Especially in the SHC rats, the rat Type lib Na / Pi Cotransporter fluctuated significantly. Example 2
実施例 1で示した Oligonucleotide microarrayでの解析結果を精查する目的 で、 TaqMan PCR Systemにより、 ラット Type lib Na/Pi Cotransporter  In order to refine the analysis results using the Oligonucleotide microarray described in Example 1, rat type lib Na / Pi Cotransporter was used by TaqMan PCR System.
(Genbank Accession No. AF157026) の各個体腎臓での発現量を測定した。 铸 型 RMとして、 実施例 1で用いた個体別腎臓 Total RNAを用い、 実験群と対照群 の発現量を比較することによって発現変動を解析した。 実験群は WFラット、 ZF ラットおよび SHCラットで、 各々に対応する対照群は WLラット、 ZLラットおよび SDラットである。  (Genbank Accession No. AF157026) in each individual kidney was measured. Using the total kidney RNA of each individual used in Example 1 as type II RM, the expression fluctuation was analyzed by comparing the expression levels of the experimental group and the control group. The experimental groups were WF rats, ZF rats and SHC rats, and the corresponding control groups were WL rats, ZL rats and SD rats.
まず、 実施例 1で調製した個体別 RNAを铸型として TaqMan Gold RT-PCR Kit (Applied Biosys terns 社) によって逆転写反応を行い、 cDNAを調製した。 反応 液の組成は、 実施例 1で調製した個体別腎臓 Total RNA lng、 5.5mM MgCl2、 0.5mM dNTPs, 2.5 zM Ramdoni Hexamers, 0.4U/ 1 RNase Inhibitor, First, a reverse transcription reaction was performed using the RNA for each individual prepared in Example 1 as a type III using a TaqMan Gold RT-PCR Kit (Applied Biosystems) to prepare cDNA. The composition of the reaction solution, Individual kidney Total RNA lng prepared in Example 1, 5.5mM MgCl 2, 0.5mM dNTPs , 2.5 zM Ramdoni Hexamers, 0.4U / 1 RNase Inhibitor,
1.25U/ /1 MultiScribe Reverse Transcriptaseおよびキットに付属の逆転写反 応用緩衝液で、 総液量を 50 xlとした。 反応は、 サーマルサイクラ一 (PE1.25 U / / 1 MultiScribe Reverse Transcriptase and reverse transcription application buffer supplied with the kit were used to make the total volume 50 xl. The reaction was performed using a thermal cycler (PE
Biosystems社) を用い、 25°C · 10分間、 48°C · 30分間、 95°C · 5分間保温するこ とで行った。 次いで、 以上の方法で調製した cDNAを铸型としてラット Type lib Na/Pi Cotranspor ter遺伝子の発現量を TaqMan PCRで測定した。 反応液の組成 は、 上記方法で調製した cDNA 5 Ι (Total RNA 2.4ng相当) 、 合成 DNAプライマ — (配列番号: 7) 0.2 M、 合成 DNAプライマー (配列番号: 4) 合成 DNAプローブ (配列番号: 5) 0.2 Mおよび TaqMan Universal PCR Master MixUsing Biosystems), the temperature was kept at 25 ° C for 10 minutes, at 48 ° C for 30 minutes, and at 95 ° C for 5 minutes. Next, the cDNA prepared by the above method was used as type III, and the expression level of the rat Type lib Na / Pi Cotransporter gene was measured by TaqMan PCR. The composition of the reaction solution was cDNA 5 Ι (corresponding to 2.4 ng of Total RNA) prepared by the above method, synthetic DNA primer — (SEQ ID NO: 7) 0.2 M, synthetic DNA primer (SEQ ID NO: 4), synthetic DNA probe (SEQ ID NO: 4). : 5) 0.2 M and TaqMan Universal PCR Master Mix
(Applied Biosystems 社) を 7.5 1加え、 総液量を 15 1とした。 反応は、 7900HT Sequence Detection System (Applied Biosystems 社) を用い、 50°C · 2分間、 95°C · 10分間の保温の後、 95 · 15秒、 60°C · 1分のサイクルを 40回繰 り返した。.試料間での铸型 cDNA添加量の多少を補正するため、 各試料について ダリセルアルデヒド 3リン酸デヒドロゲナ一ゼ (GAPDH) の発現値を測定してお いて、 ラット Type lib Na/Pi Cotranspor ter遺伝子発現量をラット GAPDH発現値 に対する相対的発現量として算出した。 相対的発現量は、 単位液量あたりのラ ット Type lib Na/Pi Cotransporter遺伝子のコピー数を同単位液量あたりのラ ット GAPDH遺伝子のコピ一数で除した値を 100倍した値として表した。 ラット GAPDHの発現値測定には、 TaqMan Rodent GAPDH Control Reagents VIC Probe(Applied Biosystems) and 7.5 1 were added to make the total volume 151. The reaction was carried out using a 7900HT Sequence Detection System (Applied Biosystems) at 50 ° C for 2 minutes and at 95 ° C for 10 minutes, followed by a cycle of 95 ° 15 seconds and 60 ° C for 1 minute 40 times. I returned. In order to compensate for the amount of type II cDNA added between samples, the expression value of dariceraldehyde triphosphate dehydrogenase (GAPDH) was measured for each sample, and rat Type lib Na / Pi Cotransporter was used. The gene expression level was calculated as the relative expression level to the rat GAPDH expression level. The relative expression level is calculated as the number of copies of the Type lib Na / Pi Cotransporter gene per unit volume. The value was divided by one copy of the GAPDH gene and multiplied by 100. TaqMan Rodent GAPDH Control Reagents VIC Probe
(Applied Biosystems 社) を用いた。 また、 ラット Type lib Na/Pi (Applied Biosystems) was used. Also, rat Type lib Na / Pi
Cotransporter遺伝子およびラット GAPDH遺伝子ともに、 測定は各試料 2回行い、 その平均値を発現値とした。 For both the Cotransporter gene and the rat GAPDH gene, measurement was performed twice for each sample, and the average value was used as the expression value.
結果を図 1〜図 3に示す。  The results are shown in FIGS.
以上の実験で得られた相対的発現量を解析した結果、 実験群である WF、 ZF、 SHCのいずれの腎疾患モデルラットにおいても病態の進行とともに、 ラット Type lib Na/Pi Cotransporter遺伝子の発現量が対照群に対して亢進することが再確 認され、 実施例 1の結果が裏付けられた。  As a result of analyzing the relative expression levels obtained in the above experiments, it was found that the expression level of the rat Type lib Na / Pi Cotransporter Was confirmed to be higher than that in the control group, confirming the results of Example 1.
また、 ZFラットに腎保護作用が報告されている TCV116 (—般名: Candesartan cilexetil) (Kidney international 49巻, SI 15 - SI 18頁, 1996年、 CI inical Experimental Nephrology 4巻, 207-214頁, 謂 0年、 Japan Journal of Pharmacology 88巻, 300- 306頁, 2002年等) を生後 18週から 9週間、 一日に体重 1kgあたり 10mg、 毎日投与した。 無投与群には投与群と同量の賦形剤 (0.5% メ チルセルロース lOOcP溶液) のみを投与した。 賦形剤投与 ZFラット、 TCV116投与 ZFラッ卜、 賦形剤投与 ZLラッ卜の 3群の個体から、 生後 27週目に腎臓を採取し、 IS0GE (二ツボンジーン社) 使用説明書記載の方法に従って個体別に Total RNA を調製し、 上記と同様に相対的発現量を解析した。  TCV116 (generic name: Candesartan cilexetil) (Kidney international 49, SI 15-SI 18, 1996, CI inical Experimental Nephrology 4, 207-214, So-called 0 year, Japan Journal of Pharmacology, 88, 300-306, 2002) was administered daily at 10 mg / kg body weight daily for 18 weeks to 9 weeks after birth. The non-administration group received only the same amount of vehicle (0.5% methylcellulose lOOcP solution) as the administration group. Kidneys were collected at 27 weeks of age from the three groups of ZF rats, vehicle-administered ZF rats, vehicle-administered ZF rats, and vehicle-administered ZL rats. Total RNA was prepared for each individual, and the relative expression was analyzed in the same manner as above.
結果を、 図 4に示す。  The results are shown in FIG.
図 4の結果から、 ZFラットに TCV116を腎障害発症前から投与しておくと、 ラ ット Type lib Na/Pi Cotransporter遺伝子の発現量が対照の ZLラットと同程度 の値になり、 TCV116無投与の ZFラットで認められるような発現宂進が起こらな くなることがわかる。 一方、 TCV116を投与した ZFラットでは、 尿中タンパク量 が ZLラット並みの正常値を示し、 腎機能低下が抑制されていた。 以上のよう に、 腎機能低下と連動してラット Type lib Na/Pi Cotransporter遺伝子の発現 亢進が認められるので、 ラット Type lib Na/Pi Cotransporter遺伝子の発現亢 進と腎機能低下とは関連性があると考えられる。 実施例 3 From the results in Fig. 4, when TCV116 was administered to ZF rats before the onset of renal injury, the expression level of the rat Type lib Na / Pi Cotransporter gene was similar to that of the control ZL rats, indicating that TCV116 was not present. It can be seen that the expression enhancement as observed in the administered ZF rats does not occur. On the other hand, in ZF rats to which TCV116 was administered, the urinary protein level was as normal as that of ZL rats, and renal function decline was suppressed. As described above, increased expression of the rat Type lib Na / Pi Cotransporter gene is observed in association with decreased renal function, and therefore, increased expression of the rat Type lib Na / Pi Cotransporter gene is associated with decreased renal function. it is conceivable that. Example 3
ヒト Type l ib Na/Pi Cot ranspor t erの細胞レベルでの発現を調べる目的で、 各種正常ヒト培養細胞での発現解析を行った。 正常ヒト細胞は Cambrex Bi osc i ence Walkersvi l l社製品を購入し、 製品添付の使用説明書記載の方法に 従って培養した。 実験に使用した細胞と各々の細胞の培養に用いた培地を表 2 に示す。  For the purpose of examining the expression of human Type Lib Na / Pi Cotransporter at the cell level, expression analysis was performed on various normal human cultured cells. Normal human cells were purchased from Cambrex Bioscience Walkersville and cultured according to the method described in the instruction manual attached to the product. Table 2 shows the cells used in the experiment and the medium used to culture each cell.
〔表 2〕  (Table 2)
No. 細胞名 培地  No. Cell name Medium
1 臍帯静脈血管内皮細胞 CC- 25Π ブレットキット EGM CC-3124 1 Umbilical vein vascular endothelial cell CC-25Π bullet kit EGM CC-3124
2 大動脈血管内皮細胞 CC- 2535 ブレットキット EGM- 2 CC-31622 Aortic vascular endothelial cells CC-2535 Bullet kit EGM-2 CC-3162
3 冠状動脈血管内皮細胞 CC- 2585 ブレットキット EGM-2MV CC-32023 Coronary vascular endothelial cells CC-2585 Bullet kit EGM-2MV CC-3202
4 皮膚微小血管内皮細胞 CC-2543 プレットキット EGM- 2MV CC-32024 Skin microvascular endothelial cells CC-2543 Pret kit EGM-2MV CC-3202
5 肺微小血管内皮細胞 CC- 2527 ブレットキット EGM-2MV CC-32025 Lung microvascular endothelial cells CC-2527 Bullet kit EGM-2MV CC-3202
6 肺動脈血管内皮細胞 CC-2530 ブレットキット EGM CC-31246 Pulmonary artery endothelial cells CC-2530 Bullet kit EGM CC-3124
7 大動脈平滑筋細胞 CC-2571 プレットキット SmGM-2 CC- 31827 Aortic smooth muscle cells CC-2571 Pret kit SmGM-2 CC-3182
8 冠状動脈平滑筋細胞 CC-2583 ブレットキット SmGM-2 CC- 31828 Coronary artery smooth muscle cells CC-2583 Bullet kit SmGM-2 CC-3182
9 子宮平滑筋細胞 CC- 2562 ブレットキット SmGM-2 CC-31829 Uterine smooth muscle cells CC-2562 Bullet kit SmGM-2 CC-3182
10 気管支平滑筋細胞 CC-2576 ブレットキット SmGM-2 CC-318210 Bronchial smooth muscle cells CC-2576 Bullet Kit SmGM-2 CC-3182
11 骨格筋衛星細胞 CC- 2561 ブレットキット SkGM CC-316011 Skeletal muscle satellite cells CC-2561 Bullet kit SkGM CC-3160
12 乳腺上皮細胞 CC- 2551 ブレットキット MEGM CC-315012 Breast epithelial cells CC-2551 Bullet kit MEGM CC-3150
13 気管支上皮細胞 CC- 2541 ブレットキット SAGM CC-311813 Bronchial epithelial cells CC-2541 Bullet kit SAGM CC-3118
14 肺細気管支上皮細胞 CC- 2547 プレットキット SAGM CC-311814 Lung bronchiolar epithelial cells CC-2547 Pret kit SAGM CC-3118
15 肺繊維芽細胞 CC- 2512 ブレットキット FGM-2 CC-313215 Lung fibroblasts CC-2512 Bullet kit FGM-2 CC-3132
16 腎臓近位尿細管上皮細胞 CC- 2553 ブレットキット REGM CC-319016 Kidney proximal tubular epithelial cells CC-2553 Bullet kit REGM CC-3190
17 メサンギゥム細胞 CC-2559 ブレットキット MsGM CC-314617 Mesangium cells CC-2559 Bullet kit MsGM CC-3146
18 腎臓皮質上皮細胞 CC- 2554 ブレットキット REGM CC-319018 Kidney cortical epithelial cells CC-2554 Bullet kit REGM CC-3190
19 腎上皮細胞 CC- 2556 ブレットキット REGM CC-319019 Renal epithelial cell CC-2556 Bullet kit REGM CC-3190
20 間葉系幹細胞 PT- 2501 ブレットキット MSCGM PT-300120 Mesenchymal stem cells PT-2501 Bullet kit MSCGM PT-3001
21 膝関節軟骨細胞 CC- 2550 ブレツトキット CGM CC-321621 Knee chondrocytes of the knee joint CC-2550 Bullet kit CGM CC-3216
22 骨芽細胞 CC-2538 プレツトキット 0GM CC-320722 Osteoblast CC-2538 Preset kit 0GM CC-3207
23 前立腺間質細胞 CC-2508 ブレットキット SCGM CC-320523 Prostate stromal cells CC-2508 Bullet kit SCGM CC-3205
24 表皮角化細胞 CC-2501 ブレットキット KGM-2 CC-3107 各細胞を 75cm2培養フラスコにサブコンフルェントになるよう培養して、 トリ プシン- EDTA処理により細胞を回収した 6 回収した細胞から、 IS0GEN (二ツボン ジーン社製) または RNeasy Mini. Kit (キアゲン社製) を用いて total RNAを調 製した。 調製した total RNA に対して TaqMan Revease Transcription Reagents (アプライドバイオシステムズ社製) を用いて逆転写反応を行い cDNAを調製し た。 次いで、 以上の方法で調製した cDNAを铸型としてヒト Type lib Na/Pi Cotransporter遺伝子の発現量を TaqMan PCRで測 した。 反応液の組成は、 各種 正常ヒト細胞由来 cDNA溶液 2 1 (Total RNA 4ng相当) 、 合成 DNAプライマー (配列番号: 1 0) 0.4 M、 合成 DNAプライマー (配列番号: 1 1) 0·4 χΜ、 合 成 DNAプローブ (配列番号: 1 2) 0.2 Μおよび TaqMan Universal PCR Master Mix (Applied Biosystems 社) を 5 1加え、 総液量を 10 1とした。 反応は、 7900HT Sequence Detection System (Applied Biosystems 社) を用い、 50°C · 2分間、 95°C · 10分間の保温の後、 95°C · 15秒、 60°C · 1分のサイクルを 40回繰 り返した。 試料間での铸型 cDNA添加量の多少を補正するため、 各試料について ダリセルアルデヒド 3リン酸デヒドロゲナ一ゼ (GAPDH) の発現値を測定し、 ヒ ト Type lib Na/Pi Cotransporter遺伝子発現量をヒト GAPDH発現値に対する相対 的発現量として算出した。 相対的発現量は、 単位液量あたりのヒト Type lib Na/Pi Cotransporter遺伝子のコピ一数を同単位液量あたりのヒト GAPDH遺伝子 のコピー数で除した値を 100倍した値として表した。 ヒト GAPDHの発現値測定に は、 TaqMan GAPDH Control Reagents (Applied Biosystems 社) を用いた。 ま た、 ヒト Type lib Na/Pi Cotransporter遺伝子およびヒト GAPDH遺伝子ともに、 測定は各試料 2回行い、 その平均値を発現値とした。 24 Epidermal keratinocytes CC-2501 Bullet kit KGM-2 CC-3107 Each cell was cultured in a 75 cm 2 culture flask so as to become subconfluent, and the cells were recovered by trypsin-EDTA treatment. 6 From the recovered cells, IS0GEN (manufactured by Futtsubon Gene) or RNeasy Mini. Kit (Qiagen) Was used to prepare total RNA. The prepared total RNA was subjected to a reverse transcription reaction using TaqMan Revease Transcription Reagents (manufactured by Applied Biosystems) to prepare cDNA. Next, the expression level of the human Type lib Na / Pi Cotransporter gene was measured by TaqMan PCR using the cDNA prepared by the above method as type III. The composition of the reaction solution was as follows: various normal human cell-derived cDNA solutions 21 (equivalent to 4 ng of total RNA), synthetic DNA primers (SEQ ID NO: 10) 0.4 M, synthetic DNA primers (SEQ ID NO: 11) 0.4 · 0.2 l of the synthetic DNA probe (SEQ ID NO: 12) and 51 of TaqMan Universal PCR Master Mix (Applied Biosystems) were added to make the total volume 10 1. The reaction was carried out using a 7900HT Sequence Detection System (Applied Biosystems) at 50 ° C for 2 minutes and at 95 ° C for 10 minutes, followed by a cycle of 95 ° C for 15 seconds and 60 ° C for 1 minute. Repeated times. To compensate for the amount of type II cDNA added between samples, the expression value of dariceraldehyde triphosphate dehydrogenase (GAPDH) was measured for each sample, and the expression level of the human Type lib Na / Pi Cotransporter gene was measured. It was calculated as the relative expression level to the human GAPDH expression level. The relative expression level was expressed as 100 times the value obtained by dividing the number of copies of the human Type lib Na / Pi Cotransporter gene per unit liquid volume by the number of copies of the human GAPDH gene per unit liquid volume. TaqMan GAPDH Control Reagents (Applied Biosystems) was used to measure the expression value of human GAPDH. In addition, for both the human Type lib Na / Pi Cotransporter gene and the human GAPDH gene, measurement was performed twice for each sample, and the average value was used as the expression value.
結果を図 5に示す。  Fig. 5 shows the results.
24種類の正常ヒ卜細胞の中では、 腎臓近位尿細管細胞で、 ヒト Type lib Among the 24 types of normal human cells, kidney proximal tubule cells and human type lib
Na/Pi Cotransporter遺伝子の比較的高い発現が認められた (GAPDHの約 2%) 。 これより、 ヒト Type lib Na/Pi Cotransporter遺伝子が腎臓で機能していると 考えられる。 産業上の利用可能性 Relatively high expression of the Na / Pi Cotransporter gene was observed (about 2% of GAPDH). This suggests that the human Type lib Na / Pi Cotransporter gene functions in the kidney. Industrial applicability
本発明のタンパク質およびポリヌクレオチドは、 例えば、 腎疾患 (例、 糖尿 病性腎症、 巣状糸球体硬化症、 慢性糸球体腎炎、 微小変化型ネフローゼ症候 群、 メサンギゥム増殖性腎炎、 管内増殖性腎炎、 IgA腎症、 膜性腎症、 膜性増殖 性腎炎、 半月体形成性腎炎、 ル一ブス腎炎、 アミロイド腎、 尿細管間質性腎 炎、 急性尿細管壊死、 急性腎不全、 腎硬化症、 腎性浮腫、 腎腫瘍、 腎移植後の 慢性拒絶など) などの診断マ一力一等として有用であり、 該タンパク質、 ポリ ヌクレオチドまたは該タンパク質に対する抗体などを用いるスクリーニングに より得られる調節剤 (好ましくは阻害剤) 、 該タンパク質に対する抗体、 本発 明のアンチセンスポリヌクレオチドなどは、 低毒性な、 例えば、 腎疾患 (例、 糖尿病性腎症、 巣状糸球体硬化症、 慢性糸球体腎炎、 微小変化型ネフローゼ症 候群、 メサンギゥム増殖性腎炎、 管内増殖性腎炎、 IgA腎症、 膜性腎症、 膜性増 殖性腎炎、 半月体形成性腎炎、 ループス腎炎、 アミロイド腎、 尿細管間質性腎 炎、 急性尿細管壊死、 急性腎不全、 腎硬化症、 腎性浮腫、 腎腫瘍、 腎移植後の 慢性拒絶など) などの予防 ·治療剤として使用することができる。 さらに、 本 発明のポリヌクレオチド、 本発明のアンチセンスポリヌクレオチドなどは、 例 えば腎疾患 (例、 糖尿病性腎症、 巣状糸球体硬化症、 慢性糸球体腎炎、 微小変 化型ネフローゼ症候群、 メサンギゥム増殖性腎炎、 管内増殖性腎炎、 IgA腎症、 膜性腎症、 膜性増殖性腎炎、 半月体形成性腎炎、 ループス腎炎、 アミロイド 腎、 尿細管間質性腎炎、 急性尿細管壊死、 急性腎不全、 腎硬化症、 腎性浮腫、 腎腫瘍、 腎移植後の慢性拒絶など) などの診断、 予防または治療に有用であ る。  The proteins and polynucleotides of the present invention include, for example, renal diseases (eg, diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome group, mesangial proliferative nephritis, endoproliferative nephritis) , IgA nephropathy, membranous nephropathy, membranous proliferative nephritis, crescentic nephritis, Rubus nephritis, amyloid kidney, tubulointerstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis And a renal edema, a renal tumor, a chronic rejection after kidney transplantation, etc.), and a modulator obtained by screening using the protein, the polynucleotide, or an antibody against the protein. Preferably an inhibitor), an antibody against the protein, an antisense polynucleotide of the present invention, and the like, are less toxic, for example, a renal disease (eg, diabetic nephropathy, Glomerulosclerosis, chronic glomerulonephritis, minimal change nephrotic syndrome, mesangial proliferative nephritis, intraluminal proliferative nephritis, IgA nephropathy, membranous nephropathy, membranous proliferative nephritis, crescent-forming nephritis , Lupus nephritis, amyloid kidney, tubulointerstitial nephritis, acute tubular necrosis, acute renal failure, renal sclerosis, renal edema, kidney tumor, chronic rejection after kidney transplantation, etc.) Can be used. Furthermore, the polynucleotide of the present invention, the antisense polynucleotide of the present invention, and the like can be used, for example, in renal diseases (eg, diabetic nephropathy, focal glomerulosclerosis, chronic glomerulonephritis, microdegenerative nephrotic syndrome, mesangium) Proliferative nephritis, intraluminal proliferative nephritis, IgA nephropathy, membranous nephropathy, membranous proliferative nephritis, crescentic nephritis, lupus nephritis, amyloid kidney, tubular interstitial nephritis, acute tubular necrosis, acute kidney It is useful for diagnosis, prevention or treatment of insufficiency, renal sclerosis, renal edema, renal tumors, chronic rejection after renal transplantation, etc.).

Claims

請求 の 範囲 The scope of the claims
1 . 配列番号: 1で表されアミノ酸配列と同一もしくは実質的に同一のアミ ノ酸配列を含有するタンパク質もしくはその部分べプチドまたはその塩の活性 を阻害する化合物またはその塩を含有してなる腎疾患の予防 ·治療剤。 1. Kidney comprising a compound or a salt thereof that inhibits the activity of a protein having the same or substantially the same amino acid sequence as SEQ ID NO: 1 or a partial peptide thereof or a salt thereof Disease prevention and treatment.
2 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質もしくはその部分ペプチドまたはその塩の遺 伝子の発現を阻害する化合物またはその塩を含有してなる腎疾患の予防 ·治療 剤。  2. Contains a compound or a salt thereof that inhibits the expression of a protein or a partial peptide thereof or a salt thereof that has the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1. Prevention and treatment of renal disease.
3 . 腎疾患が、 糖尿病性腎症または巣状糸球体硬化症である請求項 1または 請求項 2記載の予防 ·治療剤。 3. The preventive / therapeutic agent according to claim 1, wherein the renal disease is diabetic nephropathy or focal glomerulosclerosis.
4. 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質またはその部分べプチドをコードするポリヌ クレオチドの塩基配列に相補的もしくは実質的に相補的な塩基配列またはその 一部分を含有するアンチセンスポリヌクレオチド。  4. Complementary or substantially complementary to the base sequence of a polynucleotide encoding a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof Antisense polynucleotide containing a unique base sequence or a part thereof.
5 . 請求項 4記載のァンチセンスポリヌクレオチドを含有してなる医薬。 5. A medicament comprising the antisense polynucleotide according to claim 4.
6 . 腎疾患の予防 ·治療剤である請求項 5記載の医薬。 6. The medicament according to claim 5, which is an agent for preventing or treating renal disease.
7 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質またはその部分ペプチドまたはその塩に対す る抗体。  7. An antibody against a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1, a partial peptide thereof, or a salt thereof.
8 . 請求項 7記載の抗体を含有してなる医薬。  8. A drug comprising the antibody according to claim 7.
9 . 腎疾患の予防 *治療剤である請求項 8記載の医薬。  9. The medicament according to claim 8, which is an agent for preventing and treating renal disease.
1 0 . 請求項 7記載の抗体を含有してなる診断薬。  10. A diagnostic agent comprising the antibody according to claim 7.
1 1 . 腎疾患の診断薬である請求項 1 0記載の診断薬。  11. The diagnostic agent according to claim 10, which is a diagnostic agent for kidney disease.
1 2 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の アミノ酸配列を含有するタンパク質またはその部分ペプチドをコードするポリ ヌクレオチドを含有してなる腎疾患の診断薬。 12. A diagnostic agent for renal disease comprising a polynucleotide encoding a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof.
1 3 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の アミノ酸配列を含有するタンパク質もしくはその部分ペプチドまたはその塩を 用いることを特徴とする腎疾患の予防 ·.治療剤のスクリーニング方法。 1 3. A protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof Prevention of renal disease characterized by using a method for screening a therapeutic agent.
1 4 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の アミノ酸配列を含有するタンパク質もしくはその部分ペプチドまたはその塩を 含有することを特徴とする腎疾患の予防 ·治療剤のスクリーニング用キッ卜。  14. A prophylactic or therapeutic agent for renal disease characterized by containing a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof. Screening kit.
1 5 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の ァミノ酸配列を含有するタンパク質またはその部分べプチドをコードするポリ ヌクレオチドを用いることを特徴とする腎疾患の予防 ·治療剤のスクリ一ニン グ方法。  15 5. Prevention of renal disease characterized by using a polynucleotide encoding a protein containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof · Screening method for therapeutic agents.
1 6 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の ァミノ酸配列を含有するタンパク質またはその部分ペプチドをコードするポリ ヌクレオチドを含有することを特徴とする腎疾患の予防 ·治療剤のスクリ一二 ング用キット。  16. Prevention of renal disease characterized by containing a polynucleotide encoding a protein having an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof · Kit for screening therapeutic agents.
1 7 . 腎疾患が、 糖尿病性腎症または巣状糸球体硬化症である請求項 1 3ま たは請求項 1 5記載のスクリ一二ング方法。  17. The screening method according to claim 13 or claim 15, wherein the renal disease is diabetic nephropathy or focal glomerulosclerosis.
1 8 . 哺乳動物に対して、 配列番号: 1で表されるアミノ酸配列と同一もし くは実質的に同一のアミノ酸配列を含有するタンパク質もしくはその部分ぺプ チドまたはその塩の活性を阻害する化合物もしくはその塩、 または該夕ンパク 質もしくはその部分べプチドまたはその塩の遺伝子の発現を阻害する化合物も しくはその塩の有効量を投与することを特徴とする、 腎疾患の予防 ·治療法。  18. A compound that inhibits the activity of a protein, a partial peptide thereof, or a salt thereof having the same or substantially the same amino acid sequence as that shown in SEQ ID NO: 1, in mammals Or a method for preventing or treating renal disease, which comprises administering an effective amount of a compound or a salt thereof, or a compound or a salt thereof, which inhibits the expression of a gene of the protein or its partial peptide or a salt thereof.
1 9 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の ァミノ酸配列を含有するタンパク質もしくはその部分べプチドまたはその塩の 活性を阻害する、 または該タンパク質もしくはその部分べプチドまたはその塩 の遺伝子の発現を阻害することを特徴とする、 腎疾患の予防 ·治療法。  1 9. Inhibits the activity of a protein or a partial peptide or a salt thereof containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1, or the protein or a partial peptide thereof Or a method for preventing or treating kidney disease, which comprises inhibiting the expression of a salt gene.
2 0 . 腎疾患の予防 ·治療剤を製造するための配列番号: 1で表されるアミ ノ酸配列と同一もしくは実質的に同一のアミノ酸配列を含有するタンパク質も しくはその部分べプチドまたはその塩の活性を阻害する化合物もしくはその 塩、 または該タンパク質もしくはその部分ペプチドまたはその塩の遺伝子の発 現を阻害する化合物もしくはその塩の使用。 20. A protein or a partial peptide or a partial peptide thereof containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 for producing an agent for preventing or treating renal disease Use of a compound that inhibits the activity of a salt or a salt thereof, or a compound or a salt thereof that inhibits the expression of a gene of the protein or its partial peptide or a salt thereof.
PCT/JP2004/001645 2003-02-17 2004-02-16 Preventives/remedies for kidney diseases WO2004071534A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2003-038657 2003-02-17
JP2003038657 2003-02-17

Publications (1)

Publication Number Publication Date
WO2004071534A1 true WO2004071534A1 (en) 2004-08-26

Family

ID=32866400

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2004/001645 WO2004071534A1 (en) 2003-02-17 2004-02-16 Preventives/remedies for kidney diseases

Country Status (1)

Country Link
WO (1) WO2004071534A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0622077A1 (en) * 1993-04-22 1994-11-02 Takeda Chemical Industries, Ltd. Angiotensin II antagonists as a prophylactic or therapeutic drug for renal diseases
JPH09227365A (en) * 1996-02-27 1997-09-02 Kureha Chem Ind Co Ltd Zingerol-containing synthesis suppressor for hsp47
EP0875569A1 (en) * 1997-04-28 1998-11-04 Smithkline Beecham Corporation A human sodium dependent phosphate transporter (IPT-1)
EP1180686A1 (en) * 1999-05-27 2002-02-20 Tanabe Seiyaku Co., Ltd. Remedies for kidney diseases and method for screening the same

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0622077A1 (en) * 1993-04-22 1994-11-02 Takeda Chemical Industries, Ltd. Angiotensin II antagonists as a prophylactic or therapeutic drug for renal diseases
JPH09227365A (en) * 1996-02-27 1997-09-02 Kureha Chem Ind Co Ltd Zingerol-containing synthesis suppressor for hsp47
EP0875569A1 (en) * 1997-04-28 1998-11-04 Smithkline Beecham Corporation A human sodium dependent phosphate transporter (IPT-1)
EP1180686A1 (en) * 1999-05-27 2002-02-20 Tanabe Seiyaku Co., Ltd. Remedies for kidney diseases and method for screening the same

Similar Documents

Publication Publication Date Title
EP1241257A1 (en) Novel tachykinin-like polypeptides and use thereof
WO2003055506A1 (en) Preventives/remedies for cancer
WO2003091434A1 (en) Novel protein and dna thereof
JP2004073182A (en) Insulin resistance-improving agent
WO2004071534A1 (en) Preventives/remedies for kidney diseases
JP4353697B2 (en) Cancer preventive / therapeutic agent
EP1482038B1 (en) Organic anion transporter (oatp/lst) proteins and dna thereof
JP4300008B2 (en) Novel protein and its DNA
JP2004089182A (en) Preventive/therapeutic agent for cancer
JP2004269512A (en) Preventing and treating agent of renal disease
WO2004024760A1 (en) Novel protein and dna thereof
WO2003087155A1 (en) Novel protein and dna thereof
JP2004026692A (en) New use of mepe
JP2005015460A (en) Use of sglt homolog
WO2003054190A1 (en) Novel proteins and dnas thereof
WO2004002515A1 (en) Diagnostics/preventives/remedies for respiratory diseases
JP2003189873A (en) Prophylactic/therapeutic agent for cancer
WO2003097686A1 (en) Novel protein, its dna and use thereof
JP2001299364A (en) New polypeptide and dna encoding the same
JP2005151826A (en) Use of c1qtnf5
JP2004121245A (en) New protein and its dna
US20050208494A1 (en) Novel proteins and dnas thereof
WO2004039974A1 (en) Novel protein and dna thereof
JP2004323405A (en) Agent for preventing and treating renal disease
WO2004000346A1 (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 BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW 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

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP