WO2005033700A1 - Method of screening insulin resistance-improving agent - Google Patents

Method of screening insulin resistance-improving agent Download PDF

Info

Publication number
WO2005033700A1
WO2005033700A1 PCT/JP2004/014802 JP2004014802W WO2005033700A1 WO 2005033700 A1 WO2005033700 A1 WO 2005033700A1 JP 2004014802 W JP2004014802 W JP 2004014802W WO 2005033700 A1 WO2005033700 A1 WO 2005033700A1
Authority
WO
WIPO (PCT)
Prior art keywords
present
amino acid
receptor
salt
protein
Prior art date
Application number
PCT/JP2004/014802
Other languages
French (fr)
Japanese (ja)
Inventor
Hideaki Tojo
Hiroyuki Sumi
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 WO2005033700A1 publication Critical patent/WO2005033700A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • A61P5/50Drugs for disorders of the endocrine system of the pancreatic hormones for increasing or potentiating the activity of insulin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value

Definitions

  • the present invention relates to an insulin resistance improving agent, a diabetes preventive / therapeutic agent, and the like, and the like.
  • Insulin resistance is a condition in which the sensitivity of insulin to tissues decreases, and in type II diabetes in particular, it is a major etiological factor involved in the onset and progression of diabetes in addition to insulin secretion deficiency. In general, insulin resistance is considered to be closely related to obesity, since many diabetic patients with obesity generally exhibit insulin resistance. Furthermore, it is known that insulin resistance is observed not only in diabetes but also in diseases caused by abnormal lipid metabolism such as arteriosclerosis (Saltiel, AR, Cell, vol. 104, pp. 517-529, 2001). .
  • the mechanism of action of insulin resistance remains largely unknown, but one idea suggests that it is similar to the mechanism of inflammation at the animal level or at the experimental level using cultured cell lines.
  • LPS lipopolysaccharide
  • the offspring develop obesity and insulin resistance after growth (Nilsson et al, Endocrinol, 142, 262-2630, 2001).
  • TNF- a which is one of the inflammatory site power-ins, is produced and secreted more in obese adipocytes and inhibits the insulin action (Hotamisligil GS et al., Science, Vol.
  • thiazolidine derivatives which are insulin ameliorating drugs, have anti-inflammatory activity
  • ob / ob mice which are obese model mice, have been altered in the expression of several proteins involved in the inflammatory response, including LPS-binding protein (Soukas, A. et al, Genes Develop, 14 Vol. 963-980, 2000).
  • Triggering receptor expressed on myeloid cells 2 Is a single transmembrane membrane protein belonging to the immunoglobulin superfamily found as a homologue protein of ⁇ -1 which is thought to be involved in non-patent literature 1 (Non-Patent Document 1 Bauchon, A. et al., J. Immunol., 164, 4991-4995, 2000, Daws MR et al, Eur J Immunol, 31, 783-791, 2001).
  • TREM-l is abundantly expressed in neutrophils and monocytes, and its expression has the effect of inflammation by promoting secretion of lipopolysaccharide-induced TNF- a and interleukin-lj8. In addition, inhibiting this can suppress the acute inflammatory response in mice
  • TREM-2 like TREM-1, signals by interacting with DAP12
  • DAP12 Daws MR et al, Eur J Immunol, Vol. 31, pp. 783-791, 2001
  • CCR7 CC chemokine receptor 7
  • LPS lipopolysaccharide
  • LTA lipoticoic acid
  • dextran sulfate dextran sulfate
  • the present inventors have already a model animal exhibiting obesity and insulin resistance KKA y mice (Nishimura, ⁇ ⁇ , Exp Animal , 18 Certificates, 147 - 157 p., 1969) in adipose cells, the expression TREM-2 has been found among the upregulated genes, and it has been reported that suppression of TREM-2 function improves diabetes (Patent Document 1: Japanese Patent Application No. 2003-144204). Disclosure of the invention
  • gandarioside is a ligand of TREM-2.Based on this finding, the present inventors have further studied and found that The invention has been completed.
  • the present invention (1) using (a) 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, and (b) using a glycoconjugate A method for screening a compound or a salt thereof that changes the binding property between the protein or a salt thereof and the complex bran,
  • glycoconjugate is 3'-sialyl lactose, 5, -sialyl ratatoose, sialyl oleylis X, sialyl lewis A, sialyl lacto-N-tetraose a, sialyl ratato-N-tetraose b, sialyl ratato-N-
  • the screening method according to any one of the above (1) to (5), wherein the screening method is tetraose c or disialyl lacto-N-tetraose.
  • (10) (a) a protein or a salt thereof having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1; and (b) a complex saccharide.
  • a preventive / therapeutic agent for hypoglycemia comprising a complex carbohydrate
  • an insulin sensitizer comprising the compound of the above (21) or a salt thereof
  • FIG. 1 shows the results of quantification of the fluorescent label GM3 bound to mTREM2-Fc.
  • the garden shows the amount of binding to Protein G agarose to which mTREM2-Fc was bound, and the mouth shows the amount of binding to Protein G agarose to which mTREM2_Fc was not immobilized.
  • the horizontal axis shows the amount of the fluorescent label GM3 added, and the vertical axis shows the fluorescence intensity of GM3 contained in the eluate.
  • FIG. 2 is a diagram showing the results of quantification of the fluorescent label GM3 bound to mTREM1-Fc, mTREM2-Fc, hTREM1-Fc and hTREM2-Fc.
  • the mouth indicates the amount bound to mTREMl-Fc
  • the garden indicates mT
  • the amount of binding to REM2-Fc, ⁇ indicates the amount of binding to hTREM1-Fc, and indicates the amount of binding to hTREM2-Fc.
  • the horizontal axis indicates the amount of the fluorescent label GM3 added, and the vertical axis indicates the fluorescence intensity of the fluorescent GM3 contained in the eluate.
  • a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 (hereinafter sometimes referred to as the receptor of the present invention, the protein of the present invention, or the protein used in the present invention)
  • are cells of human warm-blooded animals eg, guinea pigs, rats, mice, chickens, egrets, stags, hidges, horses, monkeys, etc.
  • Organ cells eg, hepatocytes, spleen cells, nerve cells, glial cells, knees
  • Organ 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 (eg, macrophages, T cells, B cells, natural killer cells, mast cells, neutrophils, basophils, eosinophils, monocytes), megakaryocytes,
  • the amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 includes, for example, about 60% or more, preferably about 70% or more, of the amino acid sequence represented by SEQ ID NO: 1.
  • the amino acid sequence preferably has about 80% or more, preferably about 90% or more, and preferably about 95% or more homology.
  • Examples of the protein containing an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 include, for example, a protein containing the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 above. However, a protein having substantially the same activity as the protein having the amino acid sequence represented by SEQ ID NO: 1 is preferred. Examples of the protein containing the amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 1 include a protein containing the amino acid sequence represented by SEQ ID NO: 2, and the like.
  • substantially equivalent activities include, for example, signal transduction activity (eg, intracellular signal transduction activity of the protein of the present invention (preferably TREM-2)), ligand binding activity
  • binding activity between the protein of the present invention preferably TREM-2
  • a ligand or a small molecule etc.
  • the properties are qualitatively (eg, physiologically or pharmacologically) equivalent. Therefore, signal transduction activity, ligand binding 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). However, the quantitative factors such as the degree of these activities and the molecular weight of the protein may be different.
  • the signal transduction activity and the ligand binding activity can be measured according to a method known per se, for example, the method described in J. Exp. Med. 194, .1111-1112, 2001 or a method analogous thereto. it can.
  • the signal of the protein (preferably TREM-2) of the present invention causes, for example, phosphorylation of TREM-2 or DAP12 (DNAX adapter protein 12), activates ERK (extracellular signal-related protein), and induces inflammatory cytokines ( Eg, TNF-secretion).
  • the above-mentioned signal transduction activity is, for example, as compared to the protein-expressing cells of the present invention (eg, TREM-2 expressing animal cells), as necessary, (a) microbial cell lysate, microbial culture supernatant, eukaryotic cell lysate, A solution containing a ligand such as eukaryotic cell culture supernatant, (b) the ligand itself, ( c ) a substance having the same binding activity as the natural ligand to the protein of the present invention, or (d) a protein of the present invention (eg, By adding an antibody that activates TREM-2), (1) the amount of phosphorylated ERK produced, (2) the amount of extracellular TNF-a produced or (3) the phosphorylated TREM- Measure the yield of 2.
  • the protein-expressing cells of the present invention eg, TREM-2 expressing animal cells
  • ligand a complex saccharide described later is used.
  • the amount of phosphorylated ERK or TNF-a produced can be measured by a known method (eg, Western blotting, EIA, etc.) using an anti-phosphorylated ERK antibody or an anti-TNF-A antibody.
  • the amount of phosphorylated TREM-2 produced was measured using an anti-TREM-2 antibody and an anti-phosphorylated tyrosine antibody, and the amount of phosphorylated DAP12 was produced using an anti-DAP12 antibody and an anti-phosphorylated antibody.
  • a known method eg, immunoprecipitation method, Western blot method, etc.
  • Immunoprecipitation of TREM-2 or DAP12 is performed by expressing each protein in animal cells as a recombinant protein tagged with, for example, FLAG, His, V5, myc, HA, etc., and using each anti-tag antibody. You can do it too.
  • the ligand binding activity can be measured using the protein (preferably TREM-2) of the present invention and a ligand, for example, by an immunoprecipitation method, a protein affinity purification method, a yeast two-hybrid method, or the like.
  • the receptor of the present invention includes (1) (i) one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 1 (for example, about 1 to 100, preferably 1 to 100). About 30 amino acids, preferably about 1 to 10 amino acids, and more preferably about 1 to 5 amino acids, and (ii) 1 amino acid sequence represented by SEQ ID NO: 1. Also Has two or more amino acids (for example, 1 to: about L00 ', preferably about 1 to 30, preferably about 1 to 10, and more preferably about 1 to 5).
  • Amino acid sequence (iii) one or more amino acid sequences represented by SEQ ID NO: 1 (eg, 1 to: about L00, preferably about 1 to 30, preferably 1 to 1) An amino acid sequence having about 0, more preferably a number (1 to 5) amino acids inserted, and (iv) one or more amino acids in the amino acid sequence represented by SEQ ID NO: 1 (for example, 1 to 1) An amino acid sequence in which about 100, preferably about 1 to 30, preferably about 1 to 10, and more preferably a number (1 to 5) of amino acids have been substituted with another amino acid, or V) so-called muteins, such as proteins containing amino acid sequences combining them; (2) (i) SEQ ID NO: 2 1 or 2 or more in the amino acid sequence (for example, about 1 to 100, preferably about 1 to 30, preferably about 1 to 10, and more preferably about 1 to 5) (Ii) 1 or 2 or more amino acids in the amino acid sequence represented by SEQ ID NO: 2 (for example, about 1 to 100, preferably about 1
  • amino acids in the amino acid sequence represented by SEQ ID NO: 2 for example, about 1 to 100, preferably about 1 to 30, preferably about 1 to 10, more preferably (Iv) one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 2 (for example, about 1 to 100 amino acids, preferably (1 to 5) amino acids) Is an amino acid sequence in which about 1 to 30 amino acids, preferably about 1 to 10 amino acids, and more preferably several (1 to 5) amino acids have been substituted with other amino acids; or
  • the position of the insertion, deletion or substitution is not particularly limited.
  • receptor of the present invention examples include, for example, a protein containing the amino acid sequence represented by SEQ ID NO: 1 (human TREM-2) and a protein containing the amino acid sequence represented by SEQ ID NO: 2 ( Mouse TREM-2).
  • Partial peptide of the receptor of the present invention (hereinafter sometimes referred to as the partial peptide of the present invention) ) May be any partial peptide that can be used in a screening method for a drug or the like described below.
  • the protein molecule of the present invention is exposed outside the cell membrane. Sites having substantially the same ligand binding activity and the like are used.
  • the partial peptide of the present invention lacks one or more (preferably about 1 to 10, preferably 1 to 5) amino acids in its amino acid sequence, Alternatively, one or more (preferably about 1 to 20, more preferably about 1 to 10, and more preferably about 1 to 5) amino acids are added to the amino acid sequence. Or 1 or 2 or more (preferably: about! 20, more preferably about 1 to 10, and more preferably a number (1 to 5)) of amino acids in the amino acid sequence. Or one or more (preferably about 1 to 10, more preferably several, and more preferably about 1 to 5) amino acids in the amino acid sequence are replaced with other amino acids. May be.
  • the partial peptide of the present invention also contains the extracellular domain of the receptor of the present invention.
  • the extracellular domain include a peptide having the 14th to 16th amino acid sequence or the 19th to 17th amino acid sequence of the amino acid sequence represented by SEQ ID NO: 1; And a peptide having an amino acid sequence at the 14th to 170th amino acid sequence or at the 19th to 170th amino acid sequence.
  • the left end is the N-terminus (amino terminus) and the right end is the C-terminus (carboxyl terminus) according to the convention of peptide labeling.
  • C-terminal carboxy (-C00H), carboxylate (- C00-), amide (-C0NH 2) or ester - may be a (C00R).
  • R in the ester e.g., methyl, Echiru, n- propyl, alkyl such as isopropyl or n- heptyl, for example, cyclopentyl, C 3 _ 8 cycloalkyl such as cyclohexyl, for example, phenyl, - a Nafuchinore which C 6 _ 12 Ariru Piparoiruo, for example, benzyl, which are widely used as well, an ester for oral administration of C 7 _ 14 Ararukiru such phenylene Lou al kills or ⁇ - naphthylmethyl etc. ⁇ - Nafuchiru alkyl such phenethyl Xymethyl is used.
  • the receptor of the present invention and the partial peptide of the present invention have carboxy (or carboxylate) other than at the C-terminus
  • those in which carboxy is amidated or esterified also include those of the present invention. It is included in the partial peptide of the present invention.
  • the ester in this case, for example, the above-mentioned C-terminal ester and the like are used.
  • the receptor of the present invention and the partial peptide of the present invention include an N-terminal amino acid residue (eg, a methionine residue).
  • the amino group is protected by a protecting group (for example, alkyl such as formyl or acetyl, etc.), the glutamine residue at the N-terminus generated by cleavage in vivo is pyroglutamine-oxidized,
  • a protecting group for example, alkyl such as formyl or acetyl, etc.
  • the glutamine residue at the N-terminus generated by cleavage in vivo is pyroglutamine-oxidized
  • Substituents on the side chains of amino acids for example, _0H, -SH, amino group, imidazole group, indole group, guanidino group, etc.
  • suitable protecting groups for example, C w alkynyl such as formyl, acetyl, etc.
  • complex proteins such as so-called glycoproteins to which sugar chains are bound.
  • salts with physiologically acceptable acids eg, inorganic acids, organic acids
  • bases eg, alkali metal salts
  • physiologically acceptable acids eg, inorganic acids, organic acids
  • bases eg, alkali metal salts
  • Physiologically 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) And succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid and benzenesulfonic acid.
  • inorganic acids eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid
  • organic acids eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic
  • the glycoconjugate (hereinafter sometimes abbreviated as the ligand of the present invention) may be any as long as it specifically binds to the receptor of the present invention.
  • the sugar chain portion of the complex carbohydrate specifically binds to the receptor of the present invention
  • the sugar chain of the complex carbohydrate is A chain portion is also included in the ligand of the present invention.
  • the dissociation constant of the binding to the receptor of the present invention is 10 nM or less, preferably 2 M or less, more preferably 1 M or less, particularly preferably 200 nM or less, and most preferably 100 nM or less. nM or less.
  • Examples of the ligand of the present invention include gandarioside (eg, GM1, GM2, GM3, GD3, GDla, etc.), sialyloligosugar (eg, 3, -sialylratose, 5′-sialylratathose, sialylluis X, sialylluis A, sialyllac) G-N-tetraoose 3, sialyl lacto-N-tetraose b, sialyl ratato-N-tetraose c, disialyl lacto-N-tetraose, etc., lipopolysaccharide (LPS) (eg, LPS from Escherichia coli, LPS from Pseudomonas aeruginosa, etc.) Lipotic acid (LTA) (eg, LTA derived from Staphylococcus aureus, LTA derived from Bacillus subtilis),
  • the ligand of the present invention includes (a) a partial structure constituting ganglioside, sialyloligosaccharide, lipopolysaccharide, lipoticoic acid or dextran sulfate, and (b)
  • Preferred ligands of the present invention include, for example, gandariosides (eg, GM1, GM2, GM3, GD3, GDla, etc.), sialyloligosaccharides (eg, 3, -sialylactose, 5, -sialyllatase, sialyl Louis X, sialyl Lewis A, sialyl lactone) G-N-tetraose a, sialyl lacto-N-tetraose b, sialyl lacto-N-tetraose 0, disialyl lacto-N-tetraose, etc.). More preferably, it is GM3.
  • sialyloligosaccharides eg, 3, -sialylactose, 5, -sialyllatase, sialyl Louis X, sialyl Lewis A, sialyl lactone
  • Labeled glycoconjugates are also included in the ligand of the present invention.
  • the labeling substance a radioactive isotope (e.g., [125 1], [131 1], [], [14 C], [32 P], [33 P] and [35 S]), fluorescent substances [e.g. , Cyanine fluorescent dye (eg, Cy2,
  • Cy3, Cy5, Cy5.5, Cy7 (manufactured by Amersham Bioscience), fluorescamine, fluorescein isothiocyanate, NBD (7-nitrobenz-2-oxa-1,3-diazol), etc.), Enzymes (eg, ⁇ -galactosidase, mono-dalcosidase, alkaline phosphatase, peroxidase, malate dehydrogenase, etc.), luminescent substances (eg, noreminol, luminol derivatives, luciferin, lucigenin, etc.), biotin And lanthanide elements. Among them, a fluorescent substance is preferable. More NBD is preferred.
  • the labeled ligand is a gandarioside labeled with a fluorescent substance, and more preferably GM3 labeled with NBD.
  • the receptor of the present invention and the partial peptide of the present invention can be produced from the above-mentioned cells or tissues of humans or warm-blooded animals by a known method for purifying polypeptides, It can also be produced by culturing a transformant transformed with DNA encoding the polypeptide. Further, it can be produced according to the peptide synthesis method. For example, Genomics, 56, 12-21, 1999, Biochim.
  • the human or mammalian tissues or cells are homogenized and then extracted with an acid or the like, and the extract is subjected to reverse phase chromatography, ion exchange chromatography, etc. Purification and isolation can be performed by combining the above chromatography.
  • a commercially available resin for synthesizing a polypeptide can be usually used.
  • a resin include chloromethyl resin, hydroxymethyl resin, benzhydrylamine resin, aminomethyl resin, 4-benzyloxybenzyl alcohol resin, 4-methylbenzylhydramine resin, PAM resin, and the like.
  • an amino acid appropriately protected with a monoamino group and a side chain functional group can be synthesized according to the sequence of the target polypeptide. Is then condensed on a resin according to various condensation methods known per se. At the end of the reaction, the polypeptide is cleaved from the resin, and at the same time, various protecting groups are removed.In addition, an intramolecular disulfide bond formation reaction is carried out in a highly diluted solution to obtain the desired polypeptide, receptor, partial peptide or their amino acids.
  • the protected Amino acids c above can be used a variety of activation reagents that can be used in polypeptide synthesis, in particular, it is Karupojiimi earth.
  • Karposi Examples of imids include DCC, ⁇ , ' ⁇ '-diisopropyl carbodiimide, and ⁇ -ethyl ⁇ , 1- (3-dimethylaminoprolyl) carbodiimide.
  • Protected amino acids can be added directly to the resin along with an antioxidant (eg, ⁇ B t, HOOB t), or the protected amino acid can be pre-activated as a symmetric anhydride or HOB t ester or HO ⁇ B t ester. After that, it can be added to the resin.
  • the solvent used for activating the protected amino acid or condensing with the resin can be appropriately selected from solvents known to be usable for the polypeptide condensation reaction.
  • acid amides such as N, N-dimethylformamide, N, N-dimethylacetamide, N-methylvirolidone, halogenated hydrocarbons such as methylene chloride, methylform, and trifluoromethyl Alcohols such as ethanol, sulfoxides such as dimethyl sulfoxide, ethers such as pyridine, dioxane and tetrahydrofuran; nitriles such as acetonitrile and propionitrile; esters such as methyl acetate and ethyl acetate; A mixture or the like is used.
  • the reaction temperature is appropriately selected from a range known to be usable for the polypeptide bond formation reaction, and is usually appropriately selected from a range of about 120 ° C to 50 ° C.
  • the activated amino acid derivative is usually used in a 1.5 to 4-fold excess.
  • Examples of the protecting group for the starting amino group include Z, Boc, t-pentyloxycarbo-nore, isoborninoleoxycarbonyl, 4-methoxybenzyloxycanolebonyl, C11Z, Br-Z
  • adamantyl oxycarbonyl, trifluoroacetinole, phthaloynole, honoleminole, 2-butenophenorelesnorefueinole, difuinore phosphinochioil, Fmoc and the like can be used.
  • the carboxyl group is, for example, alkyl esterified (for example, methyl, ethyl, propynole, petitnole, t-butynole, cyclopentynole, cyclohexynole, cyclohexyl).
  • Linear, branched or cyclic alkyl esterification such as tyl, cyclooctyl, 2-adamantyl, etc., aralkyl esterification (eg, benzyl ester, 4-nitrobenzinoleestenole, 4-methoxypentinole) Estenole, 4-methylbenzene ester, benzhydryl ester), phenacyl ester, benzyloxycarbonyl hydrazide, t-butoxycarbol hydrazide, trityl hydrazide and the like.
  • aralkyl esterification eg, benzyl ester, 4-nitrobenzinoleestenole, 4-methoxypentinole
  • Estenole 4-methylbenzene ester, benzhydryl ester
  • phenacyl ester benzyloxycarbonyl hydrazide
  • t-butoxycarbol hydrazide trityl hydrazide
  • 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 (Cw) 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. It is possible.
  • Examples of a group suitable for ethereal dan include a benzyl group, a tetrahydrovinyl group, a t_butyl group, and the like.
  • the protecting group of the phenolic hydroxyl group of tyrosine for example, B zl, C l 2 _
  • Examples of the protecting group for imidazole of histidine include Tos, 4-methoxy-12,3,6-trimethylbenzenesulfonyl, DNP, benzyloxymethyl,
  • Examples of the activated carboxyl groups of the raw material include, for example, corresponding acid anhydrides, azides, and activated decay esters [alcohols (eg, pentachlorophenol, 2,4,5-trichlorophenol, 2 , 4-dinitrophenol, cyanomethinorea, phenolic phenol, HONB, N-hydroxysuccinimide, N-hydroxyphthalimid, HOB tester).
  • alcohols eg, pentachlorophenol, 2,4,5-trichlorophenol, 2 , 4-dinitrophenol, cyanomethinorea, phenolic phenol, HONB, N-hydroxysuccinimide, N-hydroxyphthalimid, HOB tester.
  • As the activated amino group of the raw material for example, a corresponding phosphoric amide is used.
  • Methods for removing (eliminating) protecting groups include, for example, catalytic reduction in a hydrogen stream in the presence of a catalyst such as Pd-black or Pd-carbon, or hydrogen fluoride anhydride or methanesulfonic acid.
  • a catalyst such as Pd-black or Pd-carbon, or hydrogen fluoride anhydride or methanesulfonic acid.
  • Reduction by pum is also used.
  • the elimination reaction by the above-mentioned acid treatment is generally carried out at a temperature of about 120 ° C to 40 ° C.
  • the acid treatment for example, anisol, phenanol, thioanisole, methacrylone, paracrezonole, dimethinoresolefide, It is effective to add a force-thione scavenger such as 1,4-butanedithiol, 1,2-ethanedithiol and the like.
  • a force-thione scavenger such as 1,4-butanedithiol, 1,2-ethanedithiol and the like.
  • 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 tributofan is treated with the above 1,1,1-ethanedithiol, 1,4-butanedithiol.
  • alkali treatment with dilute sodium hydroxide solution, dilute ammonia, etc.
  • the protection of the functional group which should not be involved in the reaction of the raw materials, the protecting group, the elimination of the protective group, the activation of the functional group involved in the reaction, and the like can be appropriately selected from known groups or known means.
  • a peptide (polypeptide) chain is desired on the amino group side. Then, a polypeptide is obtained by removing only the protecting group of the N-terminal monoamino group of the peptide chain and a polypeptide from which only the protecting group of the C-terminal carboxyl group is removed.
  • the two polypeptides are condensed in a mixed solvent as described above. The details of the condensation reaction are the same as described above.
  • all the protecting groups are removed by the above-mentioned method to obtain a desired crude polypeptide. This crude polypeptide is purified by various known purification means, and the main fraction is freeze-dried to obtain the desired receptor or an amide of the partial peptide.
  • an ester of the receptor or partial peptide of the present invention or a salt thereof for example, after condensing a monocarboxylic group of the carboxy terminal amino acid with a desired alcohol to form an amino acid ester, the receptor or a portion thereof is obtained.
  • an ester form of the desired receptor or a partial peptide thereof can be obtained.
  • the receptor or partial peptide of the present invention can be obtained by a known method for synthesizing a peptide.
  • a partial peptide of the receptor can be produced by cleaving the receptor 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, a partial peptide or amino acid capable of constituting the receptor or partial peptide of the present invention is condensed with the remaining portion, and when the product has a protecting group, the protecting group is eliminated to remove the desired peptide. Can be manufactured. Examples of the known condensation method and elimination of the protecting group include the methods described in the following (i) to (V).
  • the receptor or partial peptide of the present invention can be purified and isolated by combining crystals and the like.
  • the receptor or partial peptide obtained by the above method is a free form, it can be converted into an appropriate salt by a known method or a method analogous thereto. It can be converted to a free form or another salt by a known method or a method analogous thereto.
  • the polynucleotide encoding the receptor or partial peptide of the present invention may be any polynucleotide containing the above-described nucleotide sequence encoding the receptor or partial peptide of the present invention. Good. Of these, DNA is preferable, and the DNA may be any of genomic DNA, genomic DNA library, cDNA derived from the above-described cells and tissues, cDNA library derived from the above-described cells and tissues, and synthetic DNA.
  • the vectors used for the library are pateriophage, plasmid, And phagemid. Alternatively, it can be directly amplified by reverse transcriptase polymerase chain reaction (hereinafter abbreviated as RT-PCR method) using a preparation of a total RNA or mRNA fraction from the cells and tissues described above.
  • RT-PCR method reverse transcriptase polymerase chain reaction
  • Examples of the DNA encoding the receptor of the present invention include a DNA containing the nucleotide sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4, or the nucleotide sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4 Encodes a receptor having a nucleotide sequence that hybridizes under high stringent conditions to a protein having substantially the same activity as a protein having the amino acid sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4. Any DNA may be used.
  • Examples of the DNA that can hybridize with the nucleotide sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4 under high stringency conditions include, for example, the nucleotide sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4 DNA containing a base sequence having a homology of 60% or more, preferably about 70% or more, preferably about 80% or more, preferably about 90% or more, and preferably about 95% or more. Is used.
  • Hybridization is carried out by a method known per se or a method analogous thereto, for example, by the method of Molecular Cloning 2nd Edition (J. Sambrook et al., Cold Spring
  • the high stringent conditions include, for example, a sodium concentration of about 19 to 40 mM, preferably about 19 to 20 mM, and a temperature of about 50 to 70 ° C, preferably about 60 ° C.
  • the condition of ⁇ 65 ° C is shown.
  • the case where the sodium concentration is about 19 mM and the temperature is about 65 is most preferable.
  • examples of the DNA encoding the receptor containing the amino acid sequence represented by SEQ ID NO: include a DNA containing the base sequence represented by SEQ ID NO: 3, and the like.
  • DNA encoding the receptor containing the amino acid sequence represented by 2 DNA containing the base sequence represented by SEQ ID NO: 4 or the like is used.
  • the DNA encoding the partial peptide of the present invention may be any DNA containing a base sequence encoding the partial peptide of the receptor of the present invention.
  • any of genomic DNA, genomic DNA library, cDNA derived from the above-described cells and tissues, cDNA library derived from the above-described cells and tissues, and synthetic DNA may be used.
  • a DNA having a partial base sequence of a DNA having the base sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4, or a base sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4 Encoding a receptor having a nucleotide sequence that hybridizes under high stringent conditions and having substantially the same activity as a protein containing the amino acid sequence represented by SEQ ID NO: 1 or SEQ ID NO: 2 DNA having a partial nucleotide sequence of DNA to be used is used.
  • the DNA hybridizable to the nucleotide sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4 has the same significance as described above.
  • the polynucleotide (eg, DNA) encoding the receptor or partial peptide of the present invention may be labeled by a method known per se. Labeling substances include radioisotopes, fluorescent substances (eg, fluorescein, etc.), luminescent substances, enzymes, biotin, lanthanide elements, and the like.
  • a PCR method known per se using a synthetic DNA primer having a partial nucleotide sequence of the receptor or partial peptide of the present invention is used.
  • Hybridization with DNA amplified or incorporated into an appropriate vector, labeled with a DNA fragment encoding a part or all of the receptor or partial peptide of the present invention or with synthetic DNA can be sorted out.
  • Hypridease Chillon methods are described, for example, Molecuiar Cloning 2 nd Edition (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989) can be carried out according to the method described in. When a commercially available library is used, the procedure can be performed according to the method described in the attached instruction manual.
  • the DNA encoding the cloned receptor can be used as it is depending on the purpose, or digested with a restriction enzyme or added with a linker as desired.
  • the DNA may have ATG as a translation initiation codon at the 5 'end and TAA, TGA or TAG as a translation stop codon at the 3' end. These translation initiation codon and translation termination codon can be added using an appropriate synthetic DNA adapter;
  • the expression vector for the receptor or partial peptide of the present invention includes, for example, (a) cutting out a DNA fragment of interest from DNA encoding the receptor or partial peptide of the present invention, and (b) the DNA fragment Is ligated downstream of the promoter in an appropriate expression vector.
  • Plasmids derived from E. coli eg, pBR322, pBR325, pUC12, pUC13
  • Bacillus subtilis-derived plasmids eg, pUB110, pTP5, pC194
  • Plasmid derived from yeast eg, ⁇ pSH19, pSH15
  • Bacteriophage such as ⁇ phage, animal viruses such as retroinoles, vaccinia virus, baculovirus, etc., pAlll, pXTl, p RcZCMV, pRc / RSV, pcDNAI / Neo and the like are 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.
  • SRa promoter when animal cells are used as host, SRa promoter, SV40 promoter, HIV 'LTR promoter, CMV promoter, HSV-TK promoter and the like can be mentioned.
  • CMV site's megalovirus
  • SRCK SRCK promoter
  • the host is Escherichia, the trp promoter, lac promoter, recA promoter, LPL promoter, lpp promoter, T7 promoter, etc .; if the host is Bacillus, SP ⁇ 1 promoter, SPO
  • yeast such as 2 promoters, penP promoters, etc., PHO5 promoter, PGK promoter, GAP promoter, ADH promoter and the like are preferable.
  • a polyhedrin promoter, a P10 promoter and the like are preferable.
  • the expression vector may further contain, if desired, an enhancer, a splicing signal, a polyA addition signal, a selection marker, an SV40 replication origin (hereinafter sometimes abbreviated as SV40 ori), and the like.
  • an enhancer for example, dihydrofolate reductase (hereinafter sometimes abbreviated as dh fr) gene [methotrexate (MTX) resistance], ampicillin phosphorus resistant gene (hereinafter sometimes abbreviated as Amp r), Neomycin resistance gene (hereinafter sometimes abbreviated as Neo T , G418 resistance) and the like.
  • dhfr gene is used as a selection marker using Chinese hamster cells deficient in the dhfr gene
  • the target gene can be selected using a thymidine-free medium.
  • a signal sequence suitable for the host is added to the N-terminal side of the receptor of the present invention. If the host is a genus Escherichia, a Pho A signal sequence, an Omp A signal sequence, etc., if the host is a Bacillus genus, an ⁇ -amylase. Signal sequence, a subtilisin signal sequence, etc. If the host is yeast, MFa signal sequence, SUC2 signal sequence, etc.If the host is an animal cell, insulin signal sequence, Hi-interferon signal sequence, antibody molecule, signal sequence, etc. Are available respectively.
  • a transformant can be produced using the thus-constructed DNA-containing vector encoding the receptor or partial peptide of the present invention.
  • Escherichia bacteria for example, Escherichia bacteria, Bacillus bacteria, yeast, insect cells, insects, animal cells, and the like are used.
  • Specific examples of the genus Escherichia include, for example, Escherichia coli.
  • Bacillus bacteria include, for example, Bacillus.
  • subtilis MI 114 (Gene, 24, 255 983)], 207-21 [Journal of Biochemistry, 95, 87 (1984)] and the like.
  • yeast examples include, for example, Saccharomyces cerevisiae AH 22, AH 22 R ", NA87-11A, DKD-5D, 20B-12, Schizosaccharomyces pombe NC YC 1913, NCYC 2036 And Pichia pastoris K # 71.
  • insect cells for example, when the virus is Ac NPV, Spodoptera frugiperda cell lines (Spodoptera frugiperda cells), MG1 cells derived from the midgut of Trichoplusia, and High cells derived from eggs of Trichoplusia ni Five TM cells, cells derived from Mamestra brassicae or cells derived from Estigmena acrea are used.
  • a silkworm-derived cell line Boombyx raori N cell; BmN cell
  • Sf cell include Sf9 cell (ATCC CRL1711), Sf21 cell (Vaughn, J.L., et al., In Vivo).
  • insects for example, silkworm larvae are used [Maeda et al., Nature, Vol. 315, 592 (1985)].
  • animal cells examples include monkey cells COS-7 (COS 7), Vero, Chinese hamster cells CHO (hereinafter abbreviated as CH ⁇ cells), dh fr gene-deficient Chinese hamster cells CHO (hereinafter CHO ( dhfr-) cells), mouse L cells, mouse AtT-20, mouse myeloma cells, rat GH3, and human FL cells.
  • 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.
  • Transformation of insect cells or insects can be performed, for example, according to the method described in Bio / Technology, 6, 47-55 (1988).
  • Transformation of animal cells can be performed, for example, according to the method described in Cell Engineering Separate Volume 8 New Cell Engineering Experimental Protocol. 263-267 (1995) (published by Shujunsha), Virology, 52, 456 (1973). Can do it.
  • a liquid medium is suitable as a medium used for the culturing, and a carbon medium necessary for the growth of the transformant is contained therein.
  • the carbon source include glucose, dextrin, soluble starch, and sucrose.
  • examples of the nitrogen source include ammonium salts, nitrates, corn chip liquor, peptone, potato zein, meat extract, soybean meal, and potato extract.
  • Inorganic or organic substances such as liquids 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.
  • an M9 medium containing glucose and casamino acids As a medium for cultivating a bacterium belonging to the genus Escherichia, for example, an M9 medium containing glucose and casamino acids [Miller, Journal of Experiments in
  • an agent such as 3-indolylacrylic acid can be added to make the promoter work efficiently.
  • the cultivation is usually carried out at about 15 to 43 ° C for about 3 to 24 hours, and if necessary, aeration and stirring can be applied.
  • the cultivation is usually carried out at about 30 to 40 ° C for about 6 to 24 hours, and if necessary, aeration and stirring may be added.
  • the host is yeast
  • Burkholder's minimum medium Bostian, KL et al., Proc. Natl. Acad. Sci. USA, 77, 4505 (1980)
  • SD medium containing 0.5% casamino acid Bitter, GA et al., Proc. Natl. Acad. Sci. USA, 81, 5330 (1984)
  • the pH of the medium is adjusted to about 5-8.
  • the cultivation is usually performed at about 20 ° C to 35 ° C for about 24 to 72 hours, and aeration and stirring are added as necessary.
  • the medium used is 10% serum serum immobilized in Grace's Insect Medium (Grace, TCC, Nature, 195, 788 (1962)). And the like to which additives such as the above are appropriately added 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.
  • a MEM medium containing about 5 to 20% fetal bovine serum for example, a MEM medium containing about 5 to 20% fetal bovine serum [Science, 122 vol., 501 (1952)], a DMEM medium [ Virology, 8 volumes, 396 (1959)], RPMI 1640 medium [The Journal of the American Medical Association 199 volumes, 519 (1967)], 199 medium
  • the pH is about 6-8.
  • Cultivation is usually carried out at about 30 ° C to 40 ° C for about 15 to 60 hours, and aeration and stirring are added as necessary.
  • the receptor or partial peptide of the present invention can be produced in the cells, cell membrane, or extracellular cells of the transformant.
  • the receptor or partial peptide of the present invention can be separated and purified from the above culture by, for example, the following method.
  • the receptor or partial peptide of the present invention When extracting the receptor or partial peptide of the present invention from cultured cells or cells, after culturing, cells or cells are collected by a known method, and the cells or cells are suspended in an appropriate buffer, and then subjected to ultrasound, Cells or by lysozyme and / or freeze-thaw After the cells are disrupted, a method of obtaining a crude polypeptide extract by centrifugation or filtration is used as appropriate.
  • the buffer may contain a protein denaturant such as urea or guanidine hydrochloride, or a surfactant such as Triton X-100 TM.
  • the receptor or partial peptide contained in the culture supernatant or the extract obtained in this way can be purified by appropriately combining known separation and purification methods.
  • These known separation and purification methods mainly include methods utilizing solubility such as salting out and solvent precipitation, dialysis, ultrafiltration, gel filtration, and SDS-polyacrylamide gel electrophoresis.
  • Method using difference in molecular weight method using charge difference such as ion exchange chromatography, method using specific affinity such as affinity chromatography, reverse phase high-performance liquid chromatography
  • a method utilizing a difference in hydrophobicity such as a method of isoelectric point electrophoresis, or a method utilizing a difference in isoelectric point, such as isoelectric focusing electrophoresis, may be used.
  • the thus obtained receptor or partial peptide When the thus obtained receptor or partial peptide is obtained as a free form, it can be converted into a salt by a method known per se or a method analogous thereto, and conversely, when the receptor or a partial peptide is obtained as a salt, It can be converted into a free form or another salt by a known method or a method analogous thereto.
  • Receptors or partial peptides produced by recombinants can be arbitrarily modified or polypeptides can be partially removed before or after purification by the action of an appropriate protein-modifying enzyme.
  • an appropriate protein-modifying enzyme for example, trypsin, chymotrypsin, arginyl endopeptidase, protein kinase, glycosidase and the like are used.
  • a commercially available product can be used as it is, or can be extracted or produced according to a method known per se or a method analogous thereto.
  • an antibody against a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof (hereinafter, may be simply referred to as the antibody of the present invention) Is an antibody against the receptor of the present invention.
  • Any antibody that can be recognized may be a polyclonal antibody or a monoclonal antibody.
  • the antibody against the receptor of the present invention include an antibody that inactivates signal transduction of a receptor, an antibody that activates signal transduction of a receptor, and the like.
  • An antibody against the receptor of the present invention can be produced according to a known method for producing an antibody or antiserum using the receptor of the present invention as an antigen.
  • the receptor of the present invention is administered to a warm-blooded animal itself or together with a carrier or diluent at a site where antibody production is possible upon administration.
  • Complete Freund's adjuvant / incomplete Freund's adjuvant may be administered in order to enhance the antibody-producing ability upon administration. Administration is usually performed once every 2 to 6 weeks, for a total of about 2 to 10 times.
  • warm-blooded animals to be used include monkeys, rabbits, rabbits, dogs, guinea pigs, mice, rats, sheep, goats, and chickens, and mice and rats are preferably used.
  • a warm-blooded animal immunized with an antigen for example, an individual with an antibody titer is selected from a mouse, and the spleen or lymph node is collected 2 to 5 days after the final immunization and included in them.
  • an individual with an antibody titer is selected from a mouse, and the spleen or lymph node is collected 2 to 5 days after the final immunization and included in them.
  • a monoclonal antibody-producing hybridoma can be prepared.
  • the antibody titer in the antiserum can be measured, for example, by reacting the labeled polypeptide described below with the antiserum, and then measuring the activity of the labeling agent bound to the antibody.
  • the fusion operation can be performed according to a known method, for example, the method of Koehler and Milstein [Nature, 256, 495 (1975)].
  • the fusion promoter include polyethylene glycol (PEG) and Sendai virus, and PEG is preferably used.
  • PEG polyethylene glycol
  • myeloma cells include myeloma cells of warm-blooded animals such as NS_1, P3U1, SP2Z0, and AP-1, and P3U1 is preferably used.
  • the preferred ratio between the number of antibody-producing cells (spleen cells) used and the number of myeloma cells used is about 1: 1 to 20: 1, and PEG (preferably PEG1000 to PEG6000) Is added at a concentration of about 10 to 80%, and efficient cell fusion is performed by incubating at 20 to 40 ° (preferably at 30 to 37 ° C for 1 to 10 minutes).
  • PEG preferably PEG1000 to PEG6000
  • a variety of methods can be used to screen monoclonal antibody-producing hybridomas. For example, hybridoma culture supernatants can be applied to a solid phase (eg, microplate) on which polypeptide (protein) antigen is adsorbed directly or together with a carrier.
  • an anti-immunoglobulin antibody (anti-mouse immunoglobulin antibody is used if the cells used for cell fusion are mice) or peptide A, labeled with a radioactive substance or enzyme, and then add the solid phase.
  • an anti-immunoglobulin antibody anti-mouse immunoglobulin antibody is used if the cells used for cell fusion are mice
  • peptide A labeled with a radioactive substance or enzyme
  • Monoclonal antibodies can be selected according to known methods or modifications thereof. Usually, it can be performed in an animal cell culture medium supplemented with HAT (hypoxanthine, aminopterin, thymidine).
  • HAT hyperxanthine, aminopterin, thymidine
  • any medium can be used as long as it can grow a hybridoma. For example: !! 20%, preferably RPMI 1640 medium containing 10 to 20% fetal bovine serum, GIT medium containing 1 to 10% fetal bovine serum (Wako Pure Chemical Industries, Ltd. ) Or a serum-free medium for hybridoma cultivation (SFM-101, Nissui Pharmaceutical Co., Ltd.).
  • the culturing temperature is usually 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 known methods, for example, immunoglobulin separation and purification methods (eg, salting out method, alcohol precipitation method, isoelectric point precipitation method, electrophoresis method, ion exchanger (eg, DEAE)).
  • immunoglobulin separation and purification methods eg, salting out method, alcohol precipitation method, isoelectric point precipitation method, electrophoresis method, ion exchanger (eg, DEAE)
  • Adsorption / desorption method, ultracentrifugation method, gel filtration method, antigen-bound solid phase or specific purification method in which the antibody is collected using an active adsorbent such as protein A or protein G and the bond is dissociated to obtain the antibody) Can be done.
  • an active adsorbent such as protein A or protein G and the bond is dissociated to obtain the antibody
  • the polyclonal antibody of the present invention can be produced according to a known method or a method analogous thereto.
  • a immunizing antigen polypeptide antigen itself or a complex thereof with a carrier protein is formed, and a warm-blooded animal is immunized in the same manner as in the above-described method for producing a monoclonal antibody.
  • the antibody can be produced by collecting an antibody-containing substance against the antibody and separating and purifying the antibody.
  • the type of the carrier protein and the mixing ratio of the carrier and the hapten are determined by the antibody against the hapten immunized by cross-linking the carrier.
  • any kind may be cross-linked at any ratio.
  • serum albumin, thyroglobulin, hemocyanin, etc. in a weight ratio of about 0.1 to 2 per hapten per hapten.
  • a method of pulling force at 0, preferably about 1 to 5 is used.
  • Various condensing agents can be used for force coupling between the hapten and the carrier.
  • daltaraldehyde ⁇ carbodiimide, a maleimide active ester, an active ester reagent containing a thiol group or a dithioviridyl group, or the like is used.
  • the condensation product is administered to a warm-blooded animal itself or together with a carrier or diluent at a site where antibody production is possible.
  • Complete Freund's adjuvant / incomplete Freund's adjuvant may be administered in order to enhance the antibody-producing ability upon administration. The administration is usually made once every about 2 to 6 weeks, for a total of about 3 to 10 times.
  • the polyclonal antibody can be collected from the blood, ascites, etc., preferably from the blood, of the warm-blooded animal immunized by the above method.
  • the measurement of the polyclonal antibody titer in the antiserum can be performed in the same manner as the measurement of the antibody titer in the antiserum described above.
  • the separation and purification of the polyclonal antibody can be performed according to the same method for separation and purification of immunoglobulin as in the above-described separation and purification of the monoclonal antibody.
  • SEQ ID NO: 1 Complementary or substantially complementary to a polynucleotide (eg, DNA) 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 or a salt thereof
  • a polynucleotide eg, DNA
  • Any polynucleotide (antisense polynucleotide) which has a nucleotide sequence complementary to or substantially complementary to a nucleotide or a part thereof and has an action capable of suppressing the expression of the polynucleotide. It may be.
  • DNA a polynucleotide encoding the receptor of the present invention
  • these DNAs may be abbreviated as the DNA of the present invention.
  • Antisense DNA having complementary base sequence or part thereof
  • antisense DNAs having a nucleotide sequence complementary to or substantially complementary to the DNA of the present invention or a part thereof, Any antisense DNA may be used as long as it has an effect of suppressing the expression of the DNA.
  • the nucleotide sequence substantially complementary to the DNA of the present invention is, for example, about the entire nucleotide sequence or a partial nucleotide sequence of the nucleotide sequence complementary to the DNA of the present invention (that is, the complementary strand of the DNA of the present invention).
  • the complementary sequence of the base sequence of the portion encoding the N-terminal part of the receptor of the present invention is approximately 70%. % Or more, preferably about 80% or more, more preferably about 90% or more, and most preferably about 95% or more.
  • These antisense DNAs can be produced using a known DNA synthesizer or the like.
  • nucleotide sequence complementary to or substantially complementary to the nucleotide sequence of DNA having the nucleotide sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4, or an antisense having a portion thereof A polynucleotide, an antisense polynucleotide having a nucleotide sequence complementary to or substantially complementary to the nucleotide sequence of DNA having the nucleotide sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4, or a portion thereof And so on.
  • an antisense polynucleotide having a base sequence complementary to the base sequence of DNA having the base sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4, or a part thereof SEQ ID NO: 3 or SEQ ID NO: : Has a base sequence complementary to the base sequence of DNA having the base sequence represented by 4, or a part thereof Antisense polynucleotides 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. May be substituted.
  • These antisense polynucleotides can be produced using a known DNA synthesizer or the like.
  • the ligand of the present invention has a DAP12 phosphorylation promoting activity, an ERK phosphorylation promoting activity, an insulin signaling suppression activity, a TNF ct production promoting activity, a glucose uptake inhibiting activity, and the like.
  • the binding between the receptor of the present invention and the ligand of the present invention can be improved.
  • peptides, proteins, antibodies, non-peptide compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, serum, etc. or salts thereof are efficiently screened. be able to.
  • the compound or a salt thereof includes (i) cell stimulating activity via the receptor of the present invention (eg, arachidonic acid release, acetylcholine release, intracellular Ca 2+ release, intracellular cAMP generation, intracellular Inhibition of cAMP production, intracellular cGMP production, inositol phosphate production, cell membrane potential fluctuation, intracellular protein phosphorylation, activation of c_fos, decrease in pH, GTP7S binding activity, cAMP dependent Activating protein kinase, c GMP-dependent protein kinase, phospholipid-dependent protein kinase, microtubule-associated protein kinase (MAP kinase), etc.
  • cell stimulating activity via the receptor of the present invention eg, arachidonic acid release, acetylcholine release, intracellular Ca 2+ release, intracellular cAMP generation, intracellular Inhibition of cAMP production, intracellular cGMP production, inositol phosphat
  • a comparison is made between (i) the case where the receptor of the present invention is brought into contact with the ligand of the present invention and (ii) the case where the receptor of the present invention is brought into contact with the ligand of the present invention and a test compound.
  • Perform The comparison is performed, for example, by measuring the binding amount of the ligand of the present invention to the receptor of the present invention, cell stimulating activity, and the like.
  • screening method of the present invention include, for example,
  • the method comprises measuring and comparing the amount of the ligand of the present invention bound to the cell or the membrane fraction when the ligand is brought into contact with the cell or the membrane fraction of the cell.
  • the ligand of the present invention is a labeled ligand; a receptor binding assay system such as the above-mentioned (a) to (c) screen-Jung method;
  • the receptor of the present invention a membrane fraction of an organ of a human or a warm-blooded animal is preferably used.
  • the receptor of the present invention and the like which is expressed in large amounts using a recombinant, is suitable for screening.
  • the above-described method for producing the receptor of the present invention and the like are used.
  • the preparation method described later when cells containing the receptor of the present invention or the cell membrane fraction are used, the preparation method described later may be followed.
  • the cell When a cell containing the receptor of the present invention is used, the cell may be immobilized with datalaldehyde, formalin, or the like.
  • the immobilization method can be performed according to a method known per se.
  • the cell containing the receptor of the present invention refers to a host cell expressing the receptor of the present invention.
  • the host cell include the aforementioned Escherichia coli, Bacillus subtilis, yeast, insect cells, animal cells, and the like. .
  • the manufacturing method is the same as described above.
  • the membrane fraction refers to a fraction containing a large amount of cell membrane obtained by a method known per se after cell framing.
  • Methods for crushing cells include crushing cells with a Potter-Elvehjem homogenizer, Waring Plender ⁇ Polytron.
  • fractionation by centrifugal force such as fractionation centrifugation and density gradient centrifugation, is mainly used.
  • the cell lysate may be reduced at low speed (500 rpm to 3000 rpm). Centrifuge for about 1 minute to 10 minutes (usually about 1 to 10 minutes)
  • the membrane fraction is rich in the expressed receptor of the present invention and membrane components such as cell-derived phospholipids and membrane proteins.
  • the amount of the receptor of the present invention in cells or in the membrane fraction containing the receptor of the main invention is preferably from 1 0 3 to 1 0 8 molecules per cell, 1 0 5-1 0 7 molecules It is preferred that The higher the expression level, the higher the ligand binding activity (specific activity) per membrane fraction, which not only enables the construction of a highly sensitive screening system, but also enables the measurement of a large number of samples in the same port. become.
  • the receptor fraction of the present invention and the ligand of the present invention are used.
  • the receptor fraction of the present invention a naturally occurring receptor fraction of the present invention or a recombinant receptor fraction of the present invention having an activity equivalent thereto is desirable.
  • equivalent activity refers to equivalent ligand binding activity and the like.
  • the labeled ligand e.g., a radioactive isotope (e.g., [125 1], [131 1], [3 ⁇ 4], [14 C], [32 P], [33 P] and [3 3 ⁇ 4]), Fluorescent substances [eg, cyanine fluorescent dyes (eg, Cy2, Cy3, Cy5, Cy5.5, Cy7 (manufactured by Amersham Biosciences), etc.), fluorescamine, phnoleorescene isothiocynate, NBD, etc.], enzymes (eg, , / 3-galactosidase, 1-dalcosidase, al-force phosphatase, peroxidase, malate dehydrogenase, etc., luminous substances (eg, luminol, luminol derivatives, luciferin, lucigenin, etc.), piotin, lanthanide
  • a ligand labeled with an element or the like can be used.
  • a compound of a compound that changes the binding between the ligand of the present invention and the receptor of the present invention is used.
  • the buffer may be any buffer such as a phosphate buffer having a pH of 4 to 10 (preferably pH 6 to 8) or a buffer of Tris-monohydrochloride, which does not inhibit the binding between the ligand and the receptor.
  • a buffer for the purpose of reducing non-specific binding, CHAPS, Tween-80 TM (Kaoichi Atlas), digitonin, dexcholate, etc. Can be added to the buffer.
  • a protease inhibitor such as PMSF, leptin, E-64 (manufactured by Peptide Research Laboratories) and pepstatin can be added.
  • a fixed amount 5000 to 500,000 cpm
  • 10 to 1 M 10 to 7 M of the test compound are allowed to coexist.
  • a reaction tube containing a large excess of the unlabeled ligand of the present invention is also prepared to determine the non-specific binding amount (NSB).
  • the reaction is carried out at 0 ° C to 50 ° C, preferably at 4 ° C to 37 ° C for 20 minutes to 24 hours, preferably 30 minutes to 3 hours.
  • the mixture is filtered through a glass fiber filter or the like, washed with an appropriate amount of the same buffer, and the radioactivity remaining on the glass fiber filter is measured with a liquid scintillation counter or a ⁇ _counter.
  • the specific binding amount (B-NSB) is 50%, for example. % Or less of the test compound can be selected as a candidate substance having a competitive inhibition ability.
  • compounds that bind to the receptor of the present invention can be screened by utilizing the surface plasmon sensor technology.
  • the receptor of the present invention is immobilized on the surface of a sensor chip of Biacore 3000 (Biacore)
  • the surface of the chip surface when a test compound dissolved in phosphate buffer solution (PBS) or the like is flowed.
  • a test compound that binds to the receptor of the present invention is selected by measuring the change in the plasmon.
  • a test compound that gives a measured value of change in surface plasmon of 5 or more resonance units is selected as a substance having a binding property to the receptor of the present invention.
  • the increase or decrease in fluorescence intensity and fluorescence polarization can be measured to screen for a compound that binds to the receptor of the present invention.
  • the cell-stimulating activity via the receptor of the present invention for example, arachidonic acid release, acetylcholine release, intracellular Ca 2+ release, intracellular CAM P production, inhibition of intracellular cAMP production, intracellular cGMP production, inositol phosphate production, fluctuation of cell membrane potential, phosphorylation of intracellular protein, activation of c-fos, decrease of pH, GTPTS binding activity, c AM P dependency Activities that promote or inhibit protein kinase activation, c GMP-dependent protein kinase activation, phospholipid-dependent protein kinase activation, microtubule-associated protein kinase (MAP kinase) activation, etc.
  • MAP kinase microtubule-associated protein kinase
  • cells containing the receptor of the present invention are cultured in a multiwell plate or the like.c When performing screening, replace with a fresh medium or an appropriate buffer that is not toxic to cells. After adding the test compound and incubating for a certain period of time, extract the cells or collect the supernatant, and quantitate the product produced according to each method. If the production of a substance (for example, arachidonic acid) as an indicator of cell stimulating activity is difficult to be assayed by a degrading enzyme contained in a cell, an inhibitor for the degrading enzyme may be added to perform the assay. In addition, activities such as inhibition of cAMP production can be detected as an activity of inhibiting production of cells whose basic production has been increased by forskolin or the like.
  • a substance for example, arachidonic acid
  • cells expressing the appropriate receptor of the present invention are required.
  • the above-mentioned cell lines expressing the receptor of the present invention and the like are desirable.
  • Test compounds include, for example, peptides, proteins, antibodies, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and serum.
  • a compound that alters the binding between the ligand of the present invention and the receptor of the present invention is screened by measuring the stimulating activity of the ligand of the present invention on the cells expressing the receptor of the present invention. be able to.
  • the case where the ligand of the present invention is brought into contact with the receptor of the present invention and the DAP12 co-expressing cell and the case where the ligand of the present invention and the test compound are brought into contact with the receptor of the present invention and the DAP12 co-expressing cell Measure the degree of DAP12 phosphate in
  • a compound that changes the binding property between the ligand of the present invention and the receptor of the present invention is screened.
  • the degree of DAP12 phosphoric acid is measured by a known method, for example, a Western blot method using an anti-phosphorylated tyrosine antibody.
  • the receptor of the present invention and VAP-tagged DAP12 co-expression are produced by a known method.
  • the cells were seeded at 5xl0 4 cell / well in 24-well plates, and cultured for 48 hours.
  • the ligand of the present invention and a test compound are added, and the cells are cultured for 10 minutes.
  • Cells are lysed with a cell lysate [30 mM Tris-HCl (pH 7.4), 150 mM NaCl, 10 mM EDTA, 1% NP-40, 50 mM NaF, 1 mM Na-Vanadate], and then sonicated. Crush the membrane.
  • the cell lysate is immunoprecipitated with an anti-V5 antibody according to a known method, and the immunoprecipitated sample is analyzed by Western blotting.
  • DAP12 total protein in a sample is detected by a densitometer using an anti-V5 antibody (Invitrogen).
  • the degree of phosphorylation of DAP12 when cells were stimulated only with the ligand of the present invention was compared with the degree of phosphorylation of MP12 when the test compound and ⁇ : ligand of the invention were added to the cells.
  • a test compound that prevents the enhanced phosphorylation of DAP12 by the ligand is selected as an antagonist.
  • the production of intracellular cAMP is suppressed by stimulation with the ligand of the present invention.
  • the stimulatory activity of the ligand of the present invention on the cell expressing the receptor of the present invention is measured to screen for a compound that alters the binding between the ligand of the present invention and the receptor of the present invention. Can be.
  • the ligand of the present invention when the ligand of the present invention is brought into contact with the receptor-expressing cell of the present invention in the presence of a substance that increases the amount of intracellular cAMP, the ligand of the present invention and the test compound express the receptor of the present invention.
  • Compounds that alter the binding between the ligand of the present invention and the receptor of the present invention are screened by measuring and comparing the inhibitory activity of the cells on the production of intracellular cAMP when brought into contact with the cells.
  • Substances that increase the amount of intracellular cAMP include, for example, forskolin, calcito Nin and the like are used. +
  • the amount of cAMP produced in the cells expressing the receptor of the present invention was determined using an anti-cAMP antibody obtained by immunizing mice, rats, rabbits, goats, rabbits, etc., and [ 125 1] -labeled cAMP (both commercially available products). Can be measured by RIA system or EIA system combining anti-cAMP antibody and labeled cAMP. Moreover, the anti-cAMP antibody, SPA to use the beads and [125 1] -labeled cAMP scintillant containing fixed using such antibodies to such animal IgG used for protein A or an anti-cAMP antibody production (Scintillation Proximity Assay) is also possible (using a kit from Amersham-Pharmacia Biotech).
  • a test compound showing an activity of inhibiting the cAMP production-suppressing activity of the receptor-expressing cell of the present invention by the ligand of the present invention can be selected as a candidate substance having a competitive inhibition ability.
  • a compound showing agonist activity can be screened by bringing only the test compound into contact with the receptor-expressing cell of the present invention and examining the cAMP production inhibitory activity.
  • Cells expressing the receptor of the present invention are seeded on a 24-well plate at 5 ⁇ 10 4 cells / well, and cultured for 48 hours. Wash the cells with Hanks buffer ( ⁇ 7.4) containing 0.2raM 3-isobutyl-methylxanthine, 0.05% BSA and 20mM HEPES (hereinafter abbreviated as reaction buffer). Then, add 0.5 ml of reaction buffer and incubate for 30 minutes in the incubator.
  • Hanks buffer ⁇ 7.4
  • reaction buffer containing 0.2raM 3-isobutyl-methylxanthine, 0.05% BSA and 20mM HEPES
  • reaction buffer 0.25 ml of the reaction buffer was added to the cells, and 1 ⁇ L of the ligand of the present invention or 1 ⁇ L of the ligand of the present invention and the test compound were added thereto. ⁇ ⁇ Add 0.25 ml of reaction buffer containing forskolin to the cells, and incubate at 37 ° C for 24 minutes. Stop the reaction by adding ⁇ of 20% perchloric acid, then place on ice for 1 hour to extract intracellular cAMP. Measure the amount of cAMP in the extract using the cAMP EIA kit (Amersham Pharmacia Biotech).
  • a test compound for the cAMP production inhibitory activity of the ligand of the present invention Calculate the effect of A test compound that inhibits the activity of the ligside of the present invention and has a cAMP production activity of, for example, 50% or more can be selected as a candidate substance having a competitive inhibition ability.
  • the receptor-expressing cells of the present invention showing the property of increasing the intracellular cAMP level upon stimulation of the ligand of the present invention
  • the case where the ligand of the present invention is brought into contact with the receptor-expressing cells of the present invention is compared with
  • the ligand of the present invention and a test compound are brought into contact with a cell that expresses the receptor of the present invention
  • the activity of the cell for promoting intracellular cAMP production is measured and compared.
  • Compounds that alter the binding to the receptor can be screened.
  • a test compound showing an activity of inhibiting the cAMP production promoting activity of the receptor-expressing cell of the present invention by the ligand of the present invention can be selected as a candidate substance having a competitive inhibition ability.
  • a compound exhibiting agonist activity can be screened by bringing only the test compound into contact with the receptor-expressing cell of the present invention and examining cAMP production promoting activity.
  • the cAMP production promoting activity can be measured by adding the ligand of the present invention or the ligand of the present invention to cells expressing the receptor of the present invention (eg, animal cells such as CH0 cells) without adding forskolin in the above-described screening method.
  • CAMP produced by adding the compound is quantified and measured by the method described above.
  • the binding between the ligand of the present invention and the receptor of the present invention is changed by measuring the stimulating activity of the ligand of the present invention on cells expressing the receptor of the present invention using the CRE-reporter gene vector. Compounds to be screened.
  • DNA containing a CRE (cAMP response element) is inserted upstream of the reporter gene of the vector to obtain a CRE-reporter gene vector.
  • a CRE cAMP response element
  • stimulation accompanied by an increase in cAMP induces CRE-mediated reporter gene expression and subsequent production of a reporter gene gene product (protein). Induce.
  • protein reporter gene gene product
  • the ligand of the present invention is brought into contact with the receptor-expressing cell of the present invention into which a CRE-reporter gene vector has been introduced in the presence of a substance that increases the amount of intracellular cAMP.
  • the compound is brought into contact with a CRE-reporter-gene vector-transfected receptor-expressing cell of the present invention, the enzymatic activity of the reporter gene protein is measured and compared to obtain the ligand of the present invention and the ligand of the present invention. Screen for compounds that alter the binding to the receptor.
  • a substance that increases the amount of intracellular cAMP for example, phonorescoline, calcitonin, and the like are used.
  • the vector for example, Pitka Gene Basic Vector, Pikken Gene Enhancer Vector (Toyo Ink Manufacturing Co., Ltd.) and the like are used.
  • the DNA containing the CRE, the reporter gene of the vector for example ⁇ the multiple cloning site of the luciferase gene on stream, CRE-in reporter gene vector to £ method, enzyme reporter monogenic protein by the ligand of the present invention
  • a test compound that restores activity suppression can be selected as a candidate substance capable of competitive inhibition.
  • agonists can be screened by bringing only the test compound into contact with cells expressing the receptor of the present invention and measuring the same suppression of the amount of luminescence increased by forskolin stimulation as with the ligand of the present invention.
  • the cells expressing the receptor of the present invention into which the CRE-reporter gene (luciferase) has been introduced are seeded on a 24-well plate at 5 ⁇ 10 3 cells / well, and cultured for 48 hours. Wash the cells with Hank's buffer ( ⁇ 7.4) containing 0.2raM 3-isobutyl-methylxanthine, 0.05% BSA and 20mM HEPES (hereinafter abbreviated as reaction buffer). Then add 0.5 ml of reaction buffer and keep incubator for 30 minutes. After removing the reaction buffer and adding 0.25 ml of the reaction buffer to the cells, add 2 ⁇ l of the ligand of the present invention or 1 ⁇ l of the ligand of the present invention and the test compound.
  • Hank's buffer ⁇ 7.4
  • reaction buffer containing 0.2raM 3-isobutyl-methylxanthine, 0.05% BSA and 20mM HEPES
  • the ligand of the present invention suppresses an increase in luminescence by luciferase based on forskolin stimulation.
  • a compound that restores the inhibition can be selected as a candidate substance having competitive inhibitory ability.
  • reporter genes include alkaline phosphatase, chloramphenicol transferase (chloramphenicol)
  • a gene such as acetyltransferase) or j3-galactosidase may be used.
  • the enzyme activity of these reporter gene proteins is measured according to a known method or using a commercially available measurement kit.
  • Alkaline phosphatase activity can be measured using, for example, Lumi-Phos 530 manufactured by Wako Pure Chemical
  • chloramphen-chol 'acetyltransferase activity can be measured using, for example, FAST CAT chrolamphenicol Acetyltransferase Assay KiT manufactured by Wako Pure Chemical.
  • the receptor-expressing cells of the present invention release TNF o; outside the cells upon stimulation with the ligand of the present invention.
  • TNF a when the ligand of the present invention is brought into contact with the receptor-expressing cell of the present invention and when the ligand and the test compound of the present invention are brought into contact with the receptor-expressing cell of the present invention.
  • the compounds that change the binding between the ligand of the present invention and the receptor of the present invention are screened by measuring and comparing the release activities of the compounds.
  • a test compound that inhibits the activity of releasing the TNF by the ligand of the present invention can be selected as a candidate substance having a competitive inhibition ability.
  • the agonist activity was also determined by contacting the test compound alone with the receptor-expressing cells of the present invention and examining the TNF-releasing activity of the receptor-expressing cells of the present invention by a known method. Screening of compounds.
  • the receptor-expressing cells of the present invention were seeded at 5xl0 4 C ell / we ll in 24-well plates, 24 hr culture Yogo, Rigando of the present invention the ligand or a final concentration of 10 / M of the present invention at a final concentration of 10 M And test compounds are added to each well. After incubation at 37 ° C for 10 minutes, the cells were lysed with cell lysate (30 mM Tris-HCl (pH 7.4), 150 mM NaCl, 10 mM EDTA, 1% NP-40, 50 mM NaF, 1 mM Na-Vanadate ] And disintegrate the membrane by sonication.
  • the amount of TNFa contained in the cell lysate is measured according to the known EIA method.
  • the amount of TNFa is 0%, and the reaction buffer contains 10 / M of the ligand of the present invention.
  • test compound having a TNFa releasing activity of, for example, 50% or less can be selected as a candidate substance capable of competitive inhibition.
  • the Ca concentration in the cell is increased by the stimulation of the ligand of the present invention.
  • the intracellular in the case where the ligand of the present invention is brought into contact with the receptor-expressing cell of the present invention and the case where the ligand of the present invention and the test compound are brought into contact with the receptor-expressing cell of the present invention By measuring and comparing the calcium concentration increasing activity, a compound that changes the binding property between the ligand of the present invention and the receptor of the present invention is screened. The measurement is performed according to a known method.
  • a test compound that suppresses an increase in intracellular calcium concentration due to the ligand of the present invention can be selected as a candidate substance having a competitive inhibition ability.
  • agonists can be screened by measuring the increase in fluorescence intensity due to the addition of the test compound alone.
  • the receptor-expressing cell of the present invention was seeded on a sterilized microscope cover glass, and two days later, Replace the culture solution with HBSS in which 4raM Fura-2 AM (Dojindo Laboratories) is suspended, and leave at room temperature for 2 hours and 30 minutes. After washing with HBSS, place the cover glass on the cuvette, add the ligand of the present invention or the ligand of the present invention and the test compound, and measure the increase in the ratio of the fluorescence intensity at 505 nm at the excitation wavelengths of 340 nm and 380 nm by fluorescence measurement. Measure with a tester and compare.
  • FLIPR Molecular Devices
  • Fluo-3 AM manufactured by Dojindo Laboratories
  • the supernatant is washed several times by centrifugation, and the cells are transferred to a 96-well plate. Sow. Set in a FLIPR device, add the ligand of the present invention or the ligand of the present invention and a test compound in the same manner as in the case of Fura-2, measure the increase in the ratio of fluorescence intensity with a fluorimeter, and compare. I do.
  • a gene eg, aequorin
  • a gene protein eg, aequorin or the like
  • becomes Ca-binding and emits light it is also possible to screen for a compound that alters the binding between the ligand of the present invention and the receptor of the present invention.
  • a receptor-expressing cell of the present invention in which a gene for a protein that emits light by increasing intracellular Ca ions, is seeded on a 96-well plate, and the ligand of the present invention or the ligand of the present invention, Add the test compound, measure the increase in the ratio of fluorescence intensity with a fluorimeter, and compare.
  • a test compound that suppresses an increase in fluorescence intensity due to the ligand of the present invention can be selected as a candidate substance having a competitive inhibitory ability.
  • the intracellular inositol triphosphate concentration increases.
  • a compound that changes the binding property between the ligand of the present invention and the receptor of the present invention is screened by utilizing the activity of the ligand of the present invention to produce intracellular inositol triphosphate in the cells expressing the receptor of the present invention. be able to.
  • the ligand of the present invention when the ligand of the present invention is brought into contact with the receptor-expressing cell of the present invention in the presence of labeled inositol, By measuring and comparing the inositol triphosphate productivity when the cells are brought into contact with the receptor-expressing cells of the present invention, compounds that alter the binding between the ligand of the present invention and the receptor of the present invention are screened. The measurement is performed according to a known method. In this method, a test compound that suppresses inositol triphosphate-producing activity can be selected as a candidate substance capable of competitive inhibition.
  • agonists can be screened by bringing only the test compound into contact with the receptor-expressing cell of the present invention and measuring the increase in inositol triphosphate production.
  • the cells expressing the receptor of the present invention are seeded on a 24-well plate and cultured for one day. After that, the cells are cultured in a medium supplemented with myo- [2- 3 H] inositol (2.5 ⁇ Ci / well) for 1 day, and the cells are thoroughly washed with a medium free of radioactive inositol. After adding the ligand of the present invention or the ligand of the present invention and the test compound, 10% perchloric acid is added to stop the reaction. 1. neutralized with 5M hydroxide force Riumu and 60 mM HEPES solution and then passed through a column packed with AGlx8 resin (Bio- Rad) in 0.
  • the radioactivity eluted with 1 M ammonium formate and 0.1 M formic acid is measured using a liquid scintillation counter.
  • the radioactivity when the ligand of the present invention is not added is 0%, and the radioactivity when the ligand of the present invention is added is 100%, and the ligand of the present invention and the receptor of the present invention are the test compounds. Calculate the effect on binding.
  • test compound having an inositol triphosphate-producing activity of, for example, 50% or less can be selected as a candidate substance having an antagonistic ability.
  • the binding between the ligand of the present invention and the receptor of the present invention is changed by measuring the stimulating activity of the ligand of the present invention on cells expressing the receptor of the present invention using the TRE-reporter gene vector. Compounds to be screened.
  • DNA containing a TRE is inserted into the vector upstream of the reporter gene to obtain a TRE-reporter gene vector.
  • TPA response element DNA containing a TRE (TPA response element) is inserted into the vector upstream of the reporter gene to obtain a TRE-reporter gene vector.
  • stimulation accompanied by an increase in intracellular calcium concentration is caused by TRE-mediated reporter gene expression and subsequent reporter gene expression.
  • the ligand of the present invention when the ligand of the present invention is brought into contact with a TRE-reporter-gene vector-introduced receptor-expressing cell, the ligand of the present invention and a test compound are expressed in a TRE-reporter-gene vector.
  • a compound that changes the binding between the ligand of the present invention and the receptor of the present invention by measuring and comparing the enzymatic activity of the reporter gene protein when the cell is brought into contact with the cells expressing the receptor of the present invention. Screen.
  • the vector for example, Pitka Gene Basic Vector, Pitka Gene Enhancer Vector-1 (Toyo Ink Mfg. Co., Ltd.) and the like are used.
  • the DNA containing the TRE, the reporter gene of the vector for example ⁇ the multiple cloning site of the luciferase gene on stream, TRE-in c the method of the reporter gene vector, the enzymatic activity of the reporter gene protein by the ligand of the present invention
  • the test compound to be suppressed can be selected as a candidate substance having competitive inhibitory ability.
  • agonist screening is performed by bringing only the test compound into contact with the TRE-reporter gene vector-transfected receptor-expressing cell of the present invention and measuring the increase in luminescence in the same manner as the ligand of the present invention. You can also.
  • the TRE-reporter gene (luciferase) -introduced receptor-expressing cells of the present invention are seeded on a 24-well plate at 5 ⁇ 10 3 cells / well and cultured for 48 hours. After washing the cells with Hanks buffer (pH 7.4) containing 0.05% BSA and 20 mM HEPES, add ⁇ ligand of the present invention or ⁇ ligand of the present invention and a test compound, and add at 37 ° C. And react for 60 minutes.
  • Hanks buffer pH 7.4
  • BSA Hanks buffer
  • ⁇ ligand of the present invention or ⁇ ligand of the present invention and a test compound add at 37 ° C. And react for 60 minutes.
  • Cell lysing agent for Pitka Gene Toyo Ink Manufacturing
  • a luminescent substrate (Toyo Ink Mfg. Co., Ltd.) to the solution. Luminescence from Luciferase is measured with a noreminometer, liquid scintillation counter or top counter.
  • a noreminometer liquid scintillation counter or top counter.
  • the increase in intracellular calcium by the ligand of the present invention increases the amount of luminescence by luciferase.
  • a compound that suppresses this increase can be selected as a candidate substance capable of competitive inhibition.
  • Reporter genes include, for example, alkaline phosphatase, chloramphenicol transferase, and chloramphenicol.
  • acetyltransferase -A gene such as galactosidase may be used.
  • the enzyme activity of these reporter gene proteins is measured according to a known method or using a commercially available measurement kit. Alkaline phosphatase activity was measured using, for example, Lumi-Phos 530 manufactured by Wako Pure Chemical, and chloramphenicolone acetyltransferase activity was measured using, for example, FAST CAT chrolamphenicol Acetyltransferase Assay KiT manufactured by Wako Pure Chemical. The activity is measured using, for example, Aurora Gal-XE manufactured by Wako Pure Chemical Industries.
  • the MAP kinase is activated by the stimulation of the ligand of the present invention, and the cells expressing the receptor of the present invention proliferate. Utilizing this reaction, the stimulatory activity of the ligand of the present invention on the cells expressing the receptor of the present invention is measured to screen for a compound that changes the binding between the ligand of the present invention and the receptor of the present invention.
  • the t specifically capable of, the ligand of the present invention and when contacted with receptor-expressing cells of the present invention, a ligand and a test compound of the present invention, brought into contact with receptor-expressing cells of the present invention
  • a compound that changes the binding between the ligand of the present invention and the receptor of the present invention is screened.
  • the growth of the receptor-expressing cells of the present invention may be measured, for example, by measuring MAP kinase activity, thymidine uptake activity, cell number, and the like.
  • MAP kinase activity after adding the ligand of the present invention or the ligand of the present invention and a test compound to the receptor-expressing cells of the present invention, an anti-MAP kinase antibody was used from a cell lysate. After obtaining the MAP kinase fractions by immunoprecipitation, a known method, for example, manufactured by Wako pure Chemical Industries, Ltd. MAP of kinase Assay Kit and ⁇ - [32 P] - using ATP to measure MAP kinase activity, and compared.
  • the cells expressing the receptor of the present invention were seeded on a 24-well plate, cultured, and added with the ligand of the present invention or the ligand and the test compound of the present invention, and then thymidine labeled with radioactivity (eg, [methyl-II] -thymidine), then lyse the cells and measure the thymidine uptake activity by counting the radioactivity of the thymidine incorporated into the cells with a liquid scintillation counter. I do.
  • radioactivity eg, [methyl-II] -thymidine
  • the cells expressing the receptor of the present invention were seeded on a 24-well plate, cultured, supplemented with the ligand of the present invention or the ligand and the test compound of the present invention, and then treated with MTT (3- (4 , 5-dimethyto 2-thiazolyl)-2, 5-diphenyl-2H-tetrazolium bromide).
  • MTT 3- (4 , 5-dimethyto 2-thiazolyl)-2, 5-diphenyl-2H-tetrazolium bromide.
  • MTT formazan in which MTT has been changed by being taken into cells is dissolved in an aqueous solution of isopropanol acidified with hydrochloric acid, and then measured by absorption at 570 nm for comparison.
  • a test compound that suppresses the growth of the receptor-expressing cell of the present invention can be selected as a candidate substance having a competitive inhibition ability.
  • agonists can be screened by bringing only the test compound into contact with the receptor-expressing cell of the present invention and measuring the same cell proliferation activity as the ligand of the present invention.
  • the receptor-expressing cells of the present invention are seeded at 5,000 cells / well in a 24-well plate, and cultured for 1 day ( then in a serum-free medium for 2 days).
  • the ligand of the present invention or the ligand of the present invention and a test compound to the cells and culture for 24 hours, and then add [methyl-3 ⁇ 4] -thymidine at 0.015 MBq / ml.
  • wash the cells with PBS add methanol and leave for 10 minutes, then add 5% trichloroacetic acid and leave for 15 minutes to fix the cells. Wash the cells four times with distilled water 0.3 Lyse the cells with 3N sodium hydroxide solution, and measure the radioactivity in the lysate with a liquid scintillation counter.
  • a test compound that suppresses an increase in radioactivity when the ligand of the present invention is added can be selected as a candidate substance capable of competitive inhibition.
  • a compound that alters the binding between the ligand of the present invention and the receptor of the present invention is screened by measuring the stimulating activity of the ligand of the present invention on the cells expressing the receptor of the present invention. be able to.
  • ERK1 / 2 phosphorylation in the case of contacting the ligand of the present invention with the receptor-expressing cell of the present invention and in the case of contacting the ligand of the present invention and a test compound with the receptor-expressing cell of the present invention.
  • a compound that changes the binding between the ligand of the present invention and the receptor of the present invention is screened.
  • the degree of phosphorylation of ERK1 / 2 is measured by a known method, for example, Western blotting using an anti-phosphorylated ERK antibody.
  • Cells expressing the receptor of the present invention are prepared by a known method (the cells are seeded on a 24-well plate at 5 ⁇ 10 4 cells / well, and cultured for 48 hours. Add the ligand of the invention and the test compound, and incubate for 10 minutes Cell lysate [30 mM Tris-HCl (pH 7.4), 150 mM NaCl, 10 mM EDTA, 1% NP-40, 50 mM NaF , 1 was dissolved in mM Na-vanadate], further source - Keshiyon by ⁇ Re the t cells broken ⁇ to Yabu ⁇ a film by a known method, were immunoprecipitated with anti-V5 antibody, immunoprecipitated samples ⁇ We stamp lot Detect the total ERK1 / 2 protein in the sample by a known method using a densitometer using an anti-ERK1 / 2 antibody (Sigma) while using an anti-phosphorylated ERK antibody (Sigma).
  • the cells expressing the receptor of the present invention react with the ligand of the present invention to change extracellular pH. Utilizing this reaction, the stimulatory activity of the ligand of the present invention on the cells expressing the receptor of the present invention is measured, whereby the binding between the ligand of the present invention and the receptor of the present invention is measured. Compounds that alter compatibility can be screened.
  • the cells obtained when the ligand of the present invention was brought into contact with the receptor-expressing cell of the present invention and the cells obtained when the ligand and the test compound were brought into contact with the receptor-expressing cell of the present invention.
  • a compound that alters the binding between the ligand of the present invention and the receptor of the present invention is screened by measuring and comparing the change in external pH. The extracellular pH change is measured, for example, using a Cytosensor device (Molecular Devices).
  • a test compound that suppresses the extracellular pH change caused by the ligand of the present invention can be selected as a candidate substance capable of competitive inhibition.
  • agonists can be screened by bringing only the test compound into contact with the receptor-expressing cell of the present invention and measuring the extracellular pH change as in the ligand of the present invention.
  • the receptor-expressing cell of the present invention is cultured overnight in a capsule for a Cytosensor device, set in a chamber of the device, and RPMI1640 medium containing 0.1% BSA (molecular device) for about 2 hours until the extracellular pH is stabilized. Perfusion). After the pH has stabilized, the cells of the present invention or a medium containing a ligand of the present invention and a test compound are perfused onto the cells. The pH change of the medium caused by perfusion is measured and compared.
  • a compound that suppresses the extracellular pH change caused by the ligand of the present invention can be selected as a candidate substance having a competitive inhibition ability.
  • the receptor of the present invention (eg, TREM-2) is prepared by combining the extracellular domain of the receptor of the present invention (eg, TREM-2) and the fusion protein expression protein of CDSzeta with the NFAT-reporter gene. 2)
  • the signal can be detected by replacing the signal with the CD3 signal. Utilizing this reaction, the stimulatory activity of the ligand of the present invention on the cells expressing the receptor of the present invention is measured to screen for a compound that alters the binding between the ligand of the present invention and the receptor of the present invention. can do.
  • a receptor of the present invention eg, TREM-2
  • an extracellular domain of a receptor of the present invention eg, TREM-2
  • an expression vector of a fusion protein of CD3zeta and an NFAT-reporter gene eg, luciferase
  • a receptor of the present invention eg, luciferase
  • TREM-22 Activation of CD3zeta induces NFAT-mediated reporter gene expression and subsequent production of reporter gene gene products (proteins). That is, by measuring the enzymatic activity of the reporter gene protein, the signal intensity of the receptor of the present invention (eg, TREM-2) of the NFAT-reporter gene vector-introduced cells can be detected.
  • the ligand of the present invention is introduced into the expression cell into which the extracellular domain of the receptor of the present invention (eg, TREM-2) and the CD3zeta fusion protein expression vector and the NFAT-reporter gene have been introduced.
  • the ligand of the present invention and the test compound are introduced into the cells in which the extracellular domain of the receptor of the present invention (eg, TREM-2) and the fusion protein expression vector of CD3zeta and the NFAT-reporter gene have been introduced.
  • TREM-2 extracellular domain of the receptor of the present invention
  • a compound that changes the binding between the ligand of the present invention and the receptor of the present invention eg, TREM-2 is screened. I do.
  • Receptor of the present invention (Example, TREM- 2) of the extracellular domain and CD3zeta fusion protein expression vector and NFAT- reporter gene (luciferase) and introduced expression cells of at 5xl0 4 cell / well in 24-well plates Seed and incubate for 48 hours.
  • reaction buffer 1 Except for the reaction buffer 1, 0.25 ml of the reaction buffer was added to the cells, and then 1 ⁇ of the ligand of the present invention or 1 ⁇ of the ligand of the present invention and the test compound were added. Add 25 ml of application buffer to the cells and incubate at 37 ° C for 10 minutes. Lyse the cells with a cell lysing agent for Pitka Gene (Toyo Ink Mfg. Co., Ltd.) and add a luminescent substrate (Toyo Ink Mfg. Co., Ltd.) to the lysate. Luminescence from luciferase is measured with a luminometer, liquid scintillation counter or top counter.
  • a cell lysing agent for Pitka Gene Toyo Ink Mfg. Co., Ltd.
  • luminescent substrate Toyo Ink Mfg. Co., Ltd.
  • the amount of luminescence by luciferase in the case where only the ligand of the present invention is added and the case where 1 ⁇ M of the ligand of the present invention and the test compound are added are measured and compared.
  • the receptor gene RNA of the present invention is injected into Xenopus oocytes and stimulated with the ligand of the present invention, the cellular calcium concentration increases, and calcium-activated chloride current is generated. This can be considered as a change in the membrane potential (even when there is a change in the K ion concentration gradient).
  • the ligand of the present invention when the ligand of the present invention is brought into contact with the receptor gene RNA-introduced Xenopus laevis oocytes, the ligand of the present invention and the test compound are introduced into the receptor gene of the present invention.
  • the ligand of the present invention and the test compound are introduced into the receptor gene of the present invention.
  • a compound that changes the binding between the ligand of the present invention and the receptor of the present invention is screened.
  • a test compound that suppresses a change in cell membrane potential can be selected as a candidate substance having a competitive inhibition ability.
  • the receptor gene mRNA of the present invention may be prepared from a tissue or a cell, or may be transcribed from a plasmid in vitro.
  • the receptor gene mRNA of the present invention was cultured in MBS solution at 20 ° C. for 3 days, and this was placed in the cavity of a voltage clamp device in which Ringer solution was flowing, and the potential was measured. Insert the glass microelectrode for fixation and the glass microelectrode for potential measurement into the cell, and place the (-) electrode outside the cell. When the potential is stabilized, a change in potential is recorded by flowing a Ringer solution containing the ligand of the present invention or the ligand of the present invention and a test compound.
  • test compound was determined by comparing the change in the cell membrane potential of the oocyte of the receptor gene of the present invention into which RNA of the present invention was transfected with that of the Ringer solution containing only the ligand of the present invention. Can be measured.
  • a compound that suppresses the change in cell membrane potential can be selected as a candidate substance having a competitive inhibition ability.
  • a screening kit for a compound or a salt thereof that alters the binding property between the ligand of the present invention and the receptor of the present invention includes a receptor of the present invention or a cell containing the receptor of the present invention or a membrane fraction of the cell, And a ligand of the invention.
  • screening kit of the present invention examples include the following. 1. Screening reagent
  • the solution may be sterilized by filtration through a 0.45 / zm filter and stored at 4 ° C, or may be prepared at use.
  • CHO cells expressing the receptor of the present invention 1 2-well plates and passaged 5 X 1 0 5 cells / well, cultured for 2 days at 3 7 ° C, 5% C 0 2, 9 5% air What you did.
  • a radioisotope such as [], [ 125 1], [ 14 C], [ 32 P], [ 33 P], [ 35 S] in a suitable solvent or buffer Up to 4 ° C
  • the ligand of the present invention is dissolved to a concentration of 1 mM in PBS containing 0.1% ⁇ serum albumin (manufactured by Sigma) and stored at 120 ° C.
  • a compound or a salt thereof obtained by using the screening method or the screening kit of the present invention is a compound that alters the binding between the ligand of the present invention and the receptor of the present invention or the activity of the receptor of the present invention.
  • a compound that promotes or inhibits specifically, (i) a compound having a cell stimulating activity via the receptor of the present invention or a salt thereof (receptor agonist of the present invention); and (ii) a compound having the stimulating activity.
  • a compound that promotes the binding strength between the receptor of the present invention and the ligand of the present invention (iv) a binding force between the receptor of the present invention and the ligand of the present invention.
  • Such compounds include peptides, proteins, antibodies, Examples include compounds selected from non-peptide compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and serum.These compounds may be novel compounds. It may be a known compound.
  • salt of the compound those similar to the aforementioned salts of the receptor of the present invention are used.
  • the method of evaluating whether the receptor agonist of the present invention is an antagonist or an antagonist may be, for example, according to the following (i) or (ii).
  • a compound having a cell stimulating activity or a salt thereof is the receptor agonist (agonist) of the present invention, and a compound having no such activity or a salt thereof is a receptor antagonist (antagonist) of the present invention:
  • test compound is brought into contact with a cell containing the receptor of the present invention, and the cell stimulating activity of the receptor of the present invention is measured.
  • the compound having a cell stimulating activity or a salt thereof is the receptor agonist of the present invention.
  • the ligand of the present invention has a DAP12 phosphorylation promoting activity, an ERK phosphorylation promoting activity, an insulin signaling inhibitory activity, a TNF production promoting activity, a glucose uptake inhibiting activity, and the like.
  • the receptor agonist of the present invention has the physiological activity of the ligand of the present invention.
  • DAP12 phosphorylation promoting activity ERK phosphorylation promoting activity
  • insulin signaling inhibitory activity TNF a production promoting activity
  • glucose uptake inhibitory activity etc.
  • it is useful as an agent for preventing or treating hypoglycemia.
  • Physiological activity possessed by the ligand of the present invention eg, DAP12 phosphorylation promoting activity, ERK phosphorylation promoting activity, insulin signaling inhibition activity, TNF o; production promoting activity, glucose uptake inhibiting activity, etc.
  • safe and low toxic drugs such as insulin resistance improver, impaired bran tolerance, diabetes, obesity, hyperlipidemia, arteriosclerosis, prevention of hypertension or heart disease ⁇ Useful as a therapeutic agent.
  • the compound that promotes the binding strength between the receptor of the present invention and the ligand of the present invention is useful as a safe and low-toxicity drug, for example, an agent for preventing or treating hypoglycemia.
  • Compounds that inhibit the binding between the receptor of the present invention and the ligand of the present invention are safe and low-toxic drugs, such as insulin resistance improvers, impaired glucose tolerance, diabetes, obesity, hyperlipidemia, and arteries. It is useful as a preventive and therapeutic agent for sclerosis, hypertension or heart disease.
  • Compounds or salts thereof obtained using the screening method or screening kit of the present invention include, for example, peptides, proteins, antibodies, non-peptide compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissues A compound selected from the group consisting of extract, serum and the like, which compound alters the binding between the receptor of the present invention and the ligand of the present invention, and a compound which promotes or inhibits the activity or function of the receptor of the present invention And compounds that promote or inhibit (increase or decrease the expression level) the expression of the receptor gene of the present invention.
  • salt of the compound those similar to the aforementioned salts of the receptor of the present invention are used.
  • the compound or a salt thereof can be used, for example, as a sugar-coated tablet, capsule, elixir, microcapsule, etc., orally, or with water or another pharmaceutically acceptable liquid. It can be used parenterally in the form of injectable preparations such as sterile solutions or suspensions.
  • injectable preparations such as sterile solutions or suspensions.
  • the compound or a salt thereof is combined with a physiologically acceptable carrier, flavoring agent, excipient, vehicle, preservative, stabilizer, binder and the like. It can be manufactured by mixing in the unit dosage form generally required for the practice of preparations. The amount of active ingredient in these preparations is such that a suitable dosage in the specified range can be obtained.
  • Excipients that can be incorporated into tablets, forceps, etc. include, for example, binders such as gelatin, corn starch, tragacanth, gum arabic, excipients such as crystalline cellulose, corn starch, gelatin, alginic acid
  • binders such as gelatin, corn starch, tragacanth, gum arabic
  • excipients such as crystalline cellulose, corn starch, gelatin, alginic acid
  • swelling agents such as sucrose, lubricating agents such as magnesium stearate, sweetening agents such as sucrose, lactose or saccharin, and flavoring agents such as peppermint, cocoa oil or cherry are used.
  • a liquid carrier such as oils and fats can be further contained in the above-mentioned type of material.
  • Sterile compositions for injection are formulated according to standard pharmaceutical practice, such as dissolving or suspending the active substance in vehicles such as water for injection, and naturally occurring vegetable oils such as sesame oil and coconut oil. Can
  • aqueous solutions for injection include physiological saline, isotonic solutions containing glucose and other adjuvants (eg, D-sorbitol, D-mannitol, sodium chloride, etc.).
  • Agents for example, alcohols (eg, ethanol), polyalcohols (eg, propylene glycol, polyethylene glycol, etc.), nonionic surfactants (eg, polysorbate 80 TM, HCO-50, etc.) You can use it together.
  • the oily liquid include sesame oil and soybean oil, which may be used in combination with solubilizing agents such as benzyl benzoate and benzyl alcohol.
  • the compound or a salt thereof is treated with a buffer (eg, phosphate buffer, sodium acetate buffer, etc.), a soothing agent (eg, benzalkonium chloride, procaine hydrochloride, etc.), a stabilizer. Serum albumin, polyethylene glycol, etc.), preservatives (eg, benzyl alcohol, phenol, etc.), antioxidants and the like.
  • a buffer eg, phosphate buffer, sodium acetate buffer, etc.
  • a soothing agent eg, benzalkonium chloride, procaine hydrochloride, etc.
  • a stabilizer e.g, benzyl alcohol, phenol, etc.
  • the prepared injection solution is usually filled in an appropriate sample.
  • the preparations obtained in this way are safe and low toxic, and can be used, for example, in humans or warm-blooded animals (eg, mice, rats, puppies, sheep, pigs, puppies, pumas, birds, cats, dogs, Monkeys, chimpanzees, etc.). There is a difference depending on the dose of the compound or its salt, its action, target disease, subject of administration, route of administration and the like.
  • a diabetic patient per 6 kg of body weight
  • the compound when administered parenterally, for example, when the receptor antagonist of the present invention is administered to a diabetic patient (body weight: 60 kg) in the form of an injection, the compound can be administered in an amount of about 0.01 to about 0.01 per day. It is convenient to administer 30 mg, preferably about 0.:!2 Omg, more preferably about 0.1-1 Omg, by intravenous injection. In the case of other animals, the dose can be administered per 60 kg body weight.
  • the antibody of the present invention can specifically recognize the receptor of the present invention, it can be used for quantification of the receptor of the present invention in a test solution, particularly for quantification by a sandwich immunoassay. .
  • a method for quantifying the receptor of the present invention in a test solution comprising:
  • the activity of the labeling agent on the insolubilized carrier is measured.
  • a method for quantifying the receptor of the present invention in a test solution is provided.
  • one antibody may be an antibody that recognizes the N-terminal of the receptor of the present invention, and the other antibody may be an antibody that reacts with the C-terminal of the receptor of the present invention. desirable.
  • the receptor of the present invention can be quantified using a monoclonal antibody against the receptor of the present invention, and can also be detected by tissue staining or the like.
  • the antibody molecule itself may be used, or the F (ab ′) 2 , Fab ′, or Fab fraction of the antibody molecule may be used.
  • the method for quantifying the receptor of the present invention using the antibody of the present invention is not particularly limited, and may be an antibody, an antigen, or an antibody corresponding to the amount of antigen (eg, the amount of polypeptide) in the test solution. Any method that detects the amount of the complex by chemical or physical means and calculates this from a standard curve prepared using a standard solution containing a known amount of antigen can be used. Good. For example, a nephelometry, a competition method, an immunometric method, and a sandwich method are preferably used, and in terms of sensitivity and specificity, it is particularly preferable to use a sandwich method described later.
  • a labeling agent used in a measurement method using a labeling substance for example, a radioisotope, an enzyme, a fluorescent substance, a luminescent substance and the like are used. Radioisotopes, if example embodiment, [125 1], [131 1], [], are used like [14 c].
  • the above-mentioned enzymes those which are stable and have a large specific activity are preferable, and for example, ⁇ -galatatosidase, ⁇ -glucosidase, lipophosphatase, and c. Oxidase, malate dehydrogenase and the like are used.
  • the fluorescent substance for example, fluorescamine, fluorescein isothiocyanate and the like are used.
  • the luminescent substance for example, luminol, luminol derivative, luciferin, lucigenin and the like are used.
  • a biotin-avidin system can be used for binding the antibody or antigen to the labeling agent.
  • the carrier include insoluble polysaccharides such as agarose, dextran, and cellulose; synthetic resins such as polystyrene, polyacrylamide, and silicon; and glass.
  • the test solution is reacted with the insolubilized monoclonal antibody of the present invention (primary reaction), and further reacted with another labeled monoclonal antibody of the present invention (secondary reaction).
  • primary reaction the insolubilized monoclonal antibody of the present invention
  • secondary reaction another labeled monoclonal antibody of the present invention
  • the primary reaction and the secondary reaction may be performed in the reverse order, or may be performed simultaneously or at staggered times.
  • the labeling agent and the method of insolubilization can be in accordance with those described above. It is also used as an antibody for solid phase or an antibody for labeling in immunoassay by the sandwich method.
  • One type of antibody is not necessarily required, and a mixture of two or more types of antibodies may be used for the purpose of improving measurement sensitivity and the like.
  • the monoclonal antibody of the present invention used in the primary reaction and the secondary reaction is preferably an antibody having a different site to which the receptor of the present invention binds.
  • the antibody used in the primary reaction and the secondary reaction is, for example, when the antibody used in the secondary reaction recognizes the C-terminal of the receptor of the present invention, the antibody used in the primary reaction is Preferably, an antibody which recognizes other than the C-terminal, for example, the N-terminal, is used.
  • the monoclonal antibody of the present invention can be used in a measurement system other than the sandwich method, for example, a competition method, an immunometric method, or a nephrometry.
  • a competition method after the antigen in the test solution and the labeled antigen are allowed to react competitively with the antibody, the unreacted labeled antigen (F) and the labeled antigen (B) bound to the antibody are separated. (B / F separation)
  • the labeling amount of either B or F is measured, and the amount of antigen in the test solution is quantified.
  • a soluble antibody is used as the antibody
  • BZF separation is performed using polyethylene glycol
  • a liquid phase method using a second antibody against the antibody or an immobilized antibody is used as the first antibody.
  • an immobilization method using a soluble first antibody and an immobilized antibody as the second antibody is used.
  • the antigen in the test solution and the immobilized antigen are subjected to a competitive reaction with a certain amount of labeled antibody, and then the solid phase and the liquid phase are separated. After reacting the antigen with an excess amount of the labeled antibody, the immobilized antigen is added to bind the unreacted labeled antibody to the solid phase, and then the solid phase and the liquid phase are separated. Next, the amount of the label in either phase is measured to determine the amount of the antigen in the test solution.
  • nephelometry the amount of insoluble sediment resulting from an antigen-antibody reaction in a gel or in a solution is measured. Even when the amount of antigen in the test solution is small and only a small amount of sediment is obtained, laser nephrometry utilizing laser scattering is preferably used.
  • the receptor of the present invention can be quantified with good sensitivity by using the antibody of the present invention.
  • an increase in the concentration of the receptor of the present invention is detected by quantifying the concentration of the receptor of the present invention using the antibody of the present invention, for example, insulin resistance, impaired glucose tolerance, etc. It can be diagnosed as a disease such as diabetes, obesity, hyperlipidemia, arteriosclerosis, hypertension or heart disease, or is likely to be affected in the future. In addition, when a decrease in the concentration of the receptor of the present invention is detected, it can be diagnosed that the disease is, for example, a disease such as hypoglycemia, or that the disease is likely to be caused in the future.
  • the antibody of the present invention can be used for detecting the receptor 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 receptor of the present invention, detection of the receptor of the present invention in each fraction during purification, analysis of the behavior of the receptor of the present invention in test cells, etc. can be used for
  • An agent for preventing and treating hypoglycemia comprising the ligand of the present invention
  • the ligand of the present invention has MP12 phosphorylation promoting activity, ERK phosphorylation promoting activity, insulin signaling inhibition activity, TNF ⁇ production promoting activity, glucose uptake inhibitory activity, and the like. Therefore, when the ligand of the present invention is abnormal or defective, for example, hypoglycemia develops.
  • the ligand of the present invention can be used as a safe medicament such as an agent for preventing or treating hypoglycemia and the like.
  • the ligand of the present invention When used as the above-mentioned therapeutic / prophylactic agent, it should be purified to at least 90%, preferably 95% or more, more preferably 98% or more, and still more preferably 99% or more. Is preferred.
  • the ligand of the present invention can be used, for example, in the form of tablets, capsules, elixirs, microcapsenoles, and the like, which are sugar-coated as required, or with water or other pharmaceutically acceptable liquids. It can be used parenterally in the form of injections, such as sterile solutions or suspensions.
  • the ligand of the present invention is mixed with physiologically acceptable carriers, flavors, excipients, vehicles, preservatives, stabilizers, binders, and the like in a unit dosage form generally required for the practice of pharmaceutical preparations. It can be manufactured by The amount of the active ingredient in these preparations is such that a suitable dosage in the specified range can be obtained.
  • Additives that can be incorporated into tablets, capsules, etc. include, for example, binders such as gelatin, corn starch, tragacanth, gum arabic, excipients such as crystalline cellulose, corn starch, gelatin, alginic acid Lubricating agents such as magnesium stearate, sweetening agents such as sucrose, lactose or saccharin, and flavoring agents such as peppermint, cocoa oil or cherry.
  • a liquid carrier such as oils and fats can be further contained in the above-mentioned type of material.
  • Sterile compositions for injection can be formulated according to normal pharmaceutical practice, such as dissolving or suspending the active substance in vehicles such as water for injection, and naturally occurring vegetable oils such as sesame oil and coconut oil. it can.
  • Aqueous liquids for injection include, for example, saline, isotonic solutions containing dextrose and other auxiliaries (eg, D-sorbitol, D-mannitol, sodium salt, etc.)
  • Suitable solubilizers for example, alcohols (eg, ethanol, etc.), polyalcohols (eg, propylene glycol, polyethylene glycol, etc.), nonionic surfactants (eg, Polysorbate 80 TM, HC0 -50 etc.).
  • oily liquor include sesame oil and soybean oil, and may be used in combination with solubilizers such as benzyl benzoate and benzyl alcohol.
  • buffers eg, phosphate buffer, sodium acetate buffer, etc.
  • soothing agents eg, benzalkonium chloride, proforce hydrochloride, etc.
  • stabilizers eg, human serum albumin, polyethylene glycol, etc.
  • Preservatives eg, benzyl alcohol, phenol, etc.
  • antioxidants eg, antioxidants and the like.
  • the prepared injection is usually filled in an appropriate ampoule.
  • the preparations obtained in this way are safe and have low toxicity, for example, warm-blooded animals (e.g. humans, rats, mice, guinea pigs, egrets, birds, higgies, pigs, pests, pomas, cats, animals Dogs, monkeys, chimpanzees, etc.).
  • warm-blooded animals e.g. humans, rats, mice, guinea pigs, egrets, birds, higgies, pigs, pests, pomas, cats, animals Dogs, monkeys, chimpanzees, etc.
  • the dose of the ligand of the present invention varies depending on the target disease, the subject of administration, the administration route, and the like.
  • the ligand of the present invention is orally administered for the treatment of hypoglycemia, it is generally required for an adult (body weight 60 kg).
  • the ligand is 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 per day.
  • the single dose of the ligand varies depending on the administration subject, target disease, and the like.
  • the ligand of the present invention is formed in the form of an injection for the treatment of hypoglycemia.
  • the dose When administered to a human (with a body weight of 60 kg), about 0.01 to 30 mg, preferably about 0.:! To 20 mg, more preferably about 0.1 to 10 mg of the ligand per day is injected into the affected area. It is more convenient to administer. In the case of other animals, the dose can be administered in terms of weight per 60 kg.
  • the “compound or salt thereof that inhibits the activity of the ligand of the present invention on the protein of the present invention” is a compound or a salt thereof that inhibits the activating effect of the ligand of the present invention on the protein of the present invention. Any one may be used, for example, peptide, protein, antibody, non-peptide compound, synthetic compound, fermentation product, cell extract, plant extract, animal tissue extract, serum and the like. When the compound or a salt thereof is used as the above agent, it may be used in the same manner as in the above [1].
  • a prophylactic / therapeutic agent for hypoglycemia comprising a compound or a salt thereof that promotes the activating effect of the ligand of the present invention on the protein of the present invention.
  • the “compound or salt thereof that promotes the activating action of the ligand of the present invention on the protein of the present invention” includes a compound or a salt thereof that promotes the activating action of the ligand of the present invention on the protein of the present invention. Any of them may be used, for example, peptides. Proteins, antibodies, non-peptide compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, serum, and the like. When the compound or a salt thereof is used as the above agent, it may be used in the same manner as in the above [1].
  • bases, amino acids, and the like are indicated by abbreviations based on the abbreviations by the IUPAC-IUB Commission on Biochemical Nomenclature or conventional abbreviations in the relevant field.
  • optical isomer of an amino acid the L-form is indicated unless otherwise specified.
  • Y Thymine (T) or cytosine (C)
  • M Aden (A) or cytosine (C)
  • B Guanine (G), Guanine (G) or Thymine (T)
  • V Adenine (A), guanine (G) or cytosine (C)
  • N Aden (A), guanine (G), cytosine (C) or thymine (T) or unknown or other salt
  • RNA ribonucleic acid
  • mRNA messenger ribonucleic acid
  • DIEA Disopropylethylamine G 1 y or G: Glycine ''
  • H is or H
  • sequence numbers in the sequence listing in the present specification indicate the following sequences.
  • the amino acid sequence of human TREM-2 is shown.
  • [SEQ ID NO: 13] 7 shows the nucleotide sequence of a primer used in Reference Example 6.
  • Example 7 shows the nucleotide sequence of a primer used in Reference Example 6.
  • Epididymal adipose tissue was removed from KKA y mice (14 weeks old, os) and C57BL / 6 mice (14 weeks old, os) used as controls, and homogenized in IS0GEN reagent (Wako Pure Chemical Industries). After chilling, total RA was extracted from each tissue by black-mouth extraction and isopropanol precipitation, and poly (A) + RNA was further purified by Oligotex dT30 (Takara Bio).
  • cDNA is synthesized by Reverse Transcriptase (Superscript RTII; Invitrogen), and KKA y mouse adipose tissue is synthesized using PCR-Select cDNA Subtraction Kit (Clontech).
  • the cDNA group whose expression level was increased or decreased was selectively amplified as a PCR fragment.
  • the amplified cDNA group was ligated to pT7Blue-T, and cloned by transforming Escherichia coli DH5 ⁇ ( each insertion sequence was vector sequence M13 primer ⁇ 7 (SEQ ID NO: 7).
  • the TREM-2 full-length coding region cDNA was cloned from human adipocyte and mouse 3T3-L1 adipocyte cDNA libraries by PCR to confirm the expression fluctuation of TREM-2 and to obtain experimental materials for functional analysis.
  • the reaction was performed using the Advantage-2 cDNA PCR Kit (Clontech) at 35 cycles of 98 ° C for 20 seconds and 68 ° C for 1 minute and 30 seconds.
  • the reaction was performed using Pfu Turbo DNA polymerase (Stratagene) at 35 cycles of 95 ° C for 20 seconds, 65 ° C for 40 seconds, and 72 ° C for 1 minute.
  • TREM-2 isolated from human adipocytes was consistent with known human TREM-2 (AF213457).
  • mouse TREM-2 has three known cDNA sequences, TREM-2a (AY024348), TREM-2b (AY024349), and TREM_2c (AF213458), and TREM-2 obtained from 3T3-L1 adipocytes. All of the 6 clones were consistent with TREM-2a.
  • Reference example 3
  • RT-PCR was performed on RNA extracted from epididymal adipose tissue of KKA y mice (14 weeks old, os) and C57BL / 6 mice (14 weeks old, os).
  • 0.5 ig of total RNA extracted from each tissue was synthesized with AMV (Avian Myeloblastosis Virus) reverse transcriptase (TakaraPio) using 01igo-dT-adapter primer (TakaraVio) and cDNA was synthesized.
  • PCR was performed using primers (SEQ ID NO: 9 and SEQ ID NO: 10) for TREM-2 closing.
  • the reaction was performed using Advan1: age_2 cDNA PCR Kit (Clontech) at 98 ° C for 20 seconds and 68 ° C for 1 minute and 30 seconds in 25 cycles.
  • Advan1 age_2 cDNA PCR Kit (Clontech)
  • the expression level of TREM-2 in the adipose tissue of the KKA y mouse was similar to the expression level of TREM-2 in the C57BL / 6 mouse, similar to the result of the microarray in Reference Example 1. A difference of more than 5 times could be confirmed.
  • a heron was immunized with a peptide composed of the 133rd Leu to the 147th Ser of the mouse TREM-2 sequence [SEQ ID NO: 2] as an antigen to obtain an anti-TREM-2 polyclonal antibody.
  • KKA y mice and C57BL / 6 mice epididymis adipose tissue using excised each 2 0 mu ⁇
  • the antibody obtained by homogenizing the Toko filtrate was analyzed by Western plotting, TREM-2 from the KKA y mice tissues Band was detected, but not detected in the tissue derived from C57BL / 6 mouse.
  • RNA was extracted from epididymal adipose tissue, mesenteric adipose tissue, skeletal muscle, liver, testis, spleen, brain, and kidney of KKA y mice and C57BL / 6 mice, respectively, and the RT-PCR method described in Reference Example 3 was used. Examination of the distribution of TREM-2 expression tissues revealed that KKA y mice had remarkable expression in epididymal fat tissue and mesenteric fat tissue. Meanwhile, these fat groups Almost no expression was observed in tissues other than tissues.
  • the copy number of TREM-2 mRNA per 1 ng of total RNA was measured by quantitative RT-PCR.
  • C RT-PCR was performed using SYBR Green RT-PCR reagent kit (Applied Biosystems) according to the attached protocol.
  • Automatic detection ⁇ Quantification system Quantification was performed using ABI PRISM 7700 (Applied Biosystems).
  • the following primers were used for quantitative RT-PCR of mouse TREM-2.
  • the following primers were used for quantitative RT-PCR of human TREM-2.
  • the following primers were used for quantitative RT-PCR of mouse TNF- ⁇ .
  • TREM-2 expression was quantified by the quantitative RT-PCR method described in Reference Example 6 (1), KKA y In the epididymal adipose tissue and mesenteric adipose tissue of the mouse, the expression level was increased 10 times or more compared to the C57BL / 6 mouse. Furthermore, TREM-2 mRNA expression in epididymal adipose tissue of KKA y mice at 7, 14, and 28 weeks of age was measured in the same manner, and the expression increased in proportion to the increase in blood glucose with age. . At this time, TNF is an indicator of insulin resistance properties - mRNA expression level of a is increased similarly to TREM- 2, TREM - 2 and TNF - correlation coefficient of the expression of a (R 2 value) 0. 8802.
  • Total RNA was extracted from subcutaneous adipose tissue from a human diabetic patient, and the amount of TREM-2 mRNA contained in the total RNA was compared with that from a non-diabetic patient by the quantitative RT-PCR method described in Reference Example 6 (1). did.
  • TREM-2 extracellular domain (hereinafter abbreviated as Sol TREM-2) neutralizing TREM2 ligand
  • Sol TREM-2 recombines at the N-terminus of the polypeptide consisting of 147 amino acids from Ala at position 18 to Glu at position 164, which corresponds to the extracellular domain in the mouse TREM-2 sequence (SEQ ID NO: 2).
  • One having four amino acids (Gly-Ser-His-Met) added at the time of constructing the protein expression vector was prepared as an E. coli recombinant protein.
  • a total of 400 zg Sol TREM of KKA y mice (14 weeks old, male) at lOO ⁇ ug 3 days before, 2 days before, 1 day before glucose tolerance test and 4 times each on the day of glucose tolerance test -2 was administered intraperitoneally. After administration of Sol TREM-2, glucose was loaded by intraperitoneally administering 1 g / kg of glucose to each mouse, and blood glucose levels were measured up to 2 hours after glucose loading.
  • mice treated with Sol TREM-2 showed a remarkable decrease in blood glucose 30 minutes after the glucose load compared to the control mice. A decrease in blood glucose of about 100 rag / dL was observed from 60 minutes to 120 minutes.
  • the mice administered Sol TREM-2 showed approximately a lower value than control mice. A significant decrease in AUC value of 70% was observed.
  • the mRNA expression level of TNF- ⁇ in epididymal white adipose tissue after Sol TREM2 administration was reduced to 50% or less as a result of measurement by the quantitative RT-PCR method described in Reference Example 6 (1). Furthermore, by increasing the number of administrations to 8 times from 7 days before the glucose tolerance test to the day of the glucose tolerance test, the blood glucose lowering effect became more remarkable, and the fasting blood glucose level of the group without Sol TREM-2 was reduced to about In contrast to 250 mg / dL, the fasting blood glucose level of the Sol TREM-2 administration group decreased to almost a normal level of about 140 mg / dL.
  • TREM-2 is the amino acid sequence of mouse TREM-2 (SEQ ID NO: 2), which is the polypeptide N consisting of 147 amino acids from Ala at the 18th position to Glu at the 164th position corresponding to the extracellular domain.
  • -A His-Tag ligated end was expressed in E. coli under the control of the T7 promoter.
  • the target protein was purified from the cell lysate using a nickel chelate column (Bulmacier) (hereinafter abbreviated as N-His-TREM2).
  • GM3 used was Hytest (# 8G16-4h).
  • the GM3 content of the eluate increased to about 0.3, 1.0 and 1.6 at the absorbance at 492 nm.
  • the absorbance at 492 nm when GM3 was not added was below the detection limit.
  • TREM-2 and LPS bind, that TREM-2 and LPS binding are inhibited by GM3, and that GM3, which inhibits the binding of TREM-2 and LPS, competes with LPS for TREM- 2 was shown to be bound.
  • Example 2 Competitive inhibition of gangliosides (GMl, GM2, GM3, GD3), lipoteichoic acid, and dextran sulfate for binding of TREM-2 and LPS
  • the TREM2 protein used in the binding test was a polypeptide of the 147 amino acids from the 18th Ala to the 164th Glu corresponding to the extracellular domain in the amino acid sequence of mouse TREM-2 (SEQ ID NO: 2).
  • -A human immunoglobulin Fc fragment ligated to the end was expressed as a secreted protein in animal cells (mouse myeloma cells) under the control of the CMV promoter.
  • the target protein was purified from the culture using Protein A column (Pharmacia) (hereinafter abbreviated as mTREM2-Fc).
  • GM3 and 0_ (acetamido-3, 5-dideoxy-D-glycero- -D-galacto-2-nonulopyranosylonic acid)-(2 ⁇ 3)-0_ ⁇ -D-galactopyranosyl- (1 ⁇ 4) -0 - ⁇ -D- glucopyranosyl- (1 ⁇ 1)-(2S, 3R, 4E)-2-N- (7-nitrobenz-2-oxa-1, 3-diazol-4 "yl) aminooctanamido-4-octadecene- l, 3-diol (Peptide Laboratories, Inc .; hereinafter abbreviated as fluorescent label GM3) was used.
  • secondary selection is performed by selecting a substance that retains the antagonist activity using DAP12 phosphorylation as an index.
  • GM3 and a test compound are added to TREM-2 and VAP-tagged DAP12 co-expressing CH0 cells prepared by a known method, and cultured for 10 minutes. Cells are lysed with a cell lysate [30 mM Tris-HCl (pH 7.4), 150 mM NaCl, 10 mM EDTA, 1% NP-40, 50 mM NaF, 1 mM Na-Vanadate], and then sonicated. Stele the membrane.
  • the cell lysate is immunoprecipitated with an anti-V5 antibody, and the immunoprecipitated sample is analyzed by Western blotting.
  • DAP12 total protein in the sample is detected with a densitometer using an anti-V5 antibody (invitrogen).
  • C) Detect DAP12 phosphorylated with an oxidized tyrosine antibody (Sigma) by the same method.
  • C) Compare the two to detect the degree of tyrosine phosphorylation per DAP12 protein. The degree of phosphorylation of DAP12 when cells were stimulated with GM3 alone without the addition of the test compound was compared with the degree of phosphorylation of DAP12 when the test compound and GM3 were added.
  • the test compound that interferes is secondarily selected as an antagonist.
  • the second-selected test compound is administered to a diabetes model mouse such as a KKA y mouse, and a blood glucose level, a glucose tolerance test, a blood insulin level, a blood lipid level, etc. are measured to determine whether insulin resistance is improved. Confirm.
  • a diabetes model mouse such as a KKA y mouse
  • a blood glucose level, a glucose tolerance test, a blood insulin level, a blood lipid level, etc. are measured to determine whether insulin resistance is improved.
  • the human TREM-2 protein used in the binding test consists of 154 amino acids from the 14th Glu to the 167th Glu corresponding to the extracellular domain in the amino acid sequence of human TREM-2 (SEQ ID NO: 1).
  • Human immunoglobulin Fc fragment conjugated to the C-terminus of the polypeptide was expressed as a secreted protein in animal cells (mouse myeloma cells) under the control of the CMV promoter.
  • the target protein was purified from the culture using a protein A column (Pharmacia) (hereinafter abbreviated as hTREM2-Fc).
  • the mouse TREM-2 protein used in Example 3 was used.
  • Human TREM-1 protein is Recombinant Human TREM-1 / Fc Chimera (R & D System; hereinafter abbreviated as hTREMl-Fc), and mouse TREM-1 protein is Recombinant Mouse TRE-l / Fc Chimera (R & D System; mTREMl-Fc).
  • step 8 each protein and the fluorescent label GM3 were eluted from Protein G agarose. For each eluate, the fluorescence intensity at 535 nm with respect to the excitation light at 485 nm was measured, and the fluorescent label GM3 bound to each protein was quantified.
  • mice TREM- 2 Oyopi person TREM- 2 it forces s Wachikararu having substantially binding force to the equivalent GM3.
  • the protein eg, human TREM-2, mouse TREM-2, etc.
  • glycoconjugates preferably, gandarioside, more preferably, GM3
  • gandarioside preferably, GM3
  • GM3 glycoconjugates

Abstract

It is intended to provide a method of screening an insulin-resistance improving agent. More specifically, a method characterized by comprising using (a) 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, its partial peptide or its salt and (b) a complex carbohydrate capable of binding specifically to the above protein, to thereby screen a compound or its salt changing the binding properties of the above protein or its salt to the above complex carbohydrate; a kit for the screening; a compound or its salt obtained by the screening; and an insulin-resistance improving agent, a preventive/a remedy for diabetes, etc. comprising the compound or its salt.

Description

インスリン抵抗性改善剤のスクリ—ニング方法 技術分野  Screening method of insulin sensitizer
本発明は、 インスリン抵抗性の改善剤、 糖尿病の予防 ·治療剤などのスタリー ニングなどに関する。 背景技術  The present invention relates to an insulin resistance improving agent, a diabetes preventive / therapeutic agent, and the like, and the like. Background art
ィンスリン抵抗性は、 組織でのィンスリンの感受性が低下する病態で、 特に II 型糖尿病では、 インスリン分泌不全に加え、 糖尿病の発症や進展に関わる主な病 因となっている。 一般的に肥満を伴う糖尿病患者の多くがインスリン抵抗性を呈 していることから、 インスリン抵抗性は肥満と深く関わっていると考えられてい る。 さらに、 糖尿病のみならず動脈硬化等の脂質代謝異常に起因する疾病にもィ ンスリン抵抗性が見られることが知られている (Saltiel, A. R., Cell, 104卷, 517- 529頁, 2001年) 。  Insulin resistance is a condition in which the sensitivity of insulin to tissues decreases, and in type II diabetes in particular, it is a major etiological factor involved in the onset and progression of diabetes in addition to insulin secretion deficiency. In general, insulin resistance is considered to be closely related to obesity, since many diabetic patients with obesity generally exhibit insulin resistance. Furthermore, it is known that insulin resistance is observed not only in diabetes but also in diseases caused by abnormal lipid metabolism such as arteriosclerosis (Saltiel, AR, Cell, vol. 104, pp. 517-529, 2001). .
インスリン抵抗性の作用機序には未だ不明の点が多いが、 ひとつの考え方とし て、 動物実験または培養細胞系を用いた実験レベルでは炎症のメカニズムとの類 似が示唆されている。 例えば、 妊娠ラットにリポ多糖 (LPS)を投与した場合、 子 が生育した後に肥満およびィンスリン抵抗性を示すようになること(Nilsson et al, Endocrinol, 142巻, 2622- 2630頁, 2001年)、 また、 炎症性サイト力インの 一つである TNF- aが肥満の脂肪細胞において、 より多く産生 '分泌され、 ィンス リン作用を阻害すること(Hotamisligil G. S. et al. , Science, 259卷, 87- 91頁, 1993年)、 さらに、 インスリン抵抗生改善薬であるチアゾリジン誘導体が抗炎症 作用を持つこと(Pasceri, V. et al. , Circulation, 101卷, 235- 238頁, 2000 年)などが報告されている。 また、 肥満モデルマウスである ob/obマウスでも LPS- binding proteinをはじめとするいくつかの炎症反応に関与する蛋白質の発現変 動が認められている (Soukas, A. et al, Genes Develop, 14卷, 963 - 980頁, 2000年)。 The mechanism of action of insulin resistance remains largely unknown, but one idea suggests that it is similar to the mechanism of inflammation at the animal level or at the experimental level using cultured cell lines. For example, when lipopolysaccharide (LPS) is administered to pregnant rats, the offspring develop obesity and insulin resistance after growth (Nilsson et al, Endocrinol, 142, 262-2630, 2001). In addition, TNF- a, which is one of the inflammatory site power-ins, is produced and secreted more in obese adipocytes and inhibits the insulin action (Hotamisligil GS et al., Science, Vol. 259, 87- 91, 1993), and the fact that thiazolidine derivatives, which are insulin ameliorating drugs, have anti-inflammatory activity (Pasceri, V. et al., Circulation, 101, 235-238, 2000). Have been. Also, ob / ob mice, which are obese model mice, have been altered in the expression of several proteins involved in the inflammatory response, including LPS-binding protein (Soukas, A. et al, Genes Develop, 14 Vol. 963-980, 2000).
Triggering receptor expressed on myeloid cells 2 (TREM- 2) は、 炎;)正反/心 に関与していると考えられている ίΈΕΜ - 1のホモログ蛋白質として見いだされた免 疫グロプリンスーパ一フアミリーに属する一回膜貫通型膜蛋白質である (非特許 文献 1 Bauchon, A. et al. , J. Immunol., 164卷, 4991 - 4995頁, 2000年、 Daws M. R. et al, Eur J Immunol, 31巻, 783- 791頁, 2001年)。 TREM-lは、 好中球、 単球で多量に発現しており、 その発現はリポ多糖で誘導される TNF- aやインター ロイキン - l j8の分泌を促すことにより炎症の増幅作用を持つこと、 さらに、 これ を阻害することによりマウスで急性炎症応答を抑えることができること Triggering receptor expressed on myeloid cells 2 (TREM-2) Is a single transmembrane membrane protein belonging to the immunoglobulin superfamily found as a homologue protein of ίΈΕΜ-1 which is thought to be involved in non-patent literature 1 (Non-Patent Document 1 Bauchon, A. et al., J. Immunol., 164, 4991-4995, 2000, Daws MR et al, Eur J Immunol, 31, 783-791, 2001). TREM-l is abundantly expressed in neutrophils and monocytes, and its expression has the effect of inflammation by promoting secretion of lipopolysaccharide-induced TNF- a and interleukin-lj8. In addition, inhibiting this can suppress the acute inflammatory response in mice
(Bauchon, A. et al. , Nature, 410卷, 1103- 1107頁, 2001年) が知られている ( TREM - 2は、 TREM- 1と同様に DAP12と相互作用することによりシグナル伝達を行う こと(Daws M. R. et al, Eur J Immunol, 31巻, 783 - 791頁, 2001年) 、 樹状細胞 またはマクロファージでは CC chemokine receptor 7 (CCR7) の発現誘導を引き 起こし樹状細胞の成熟に関与していること (Bouchon, A. et al. , J. Exp. Med. 194巻, 1111- 1122頁, 2001年)、 NOの産生に関与していること(Daws M. R. et al, Eur J Immunol, 31巻, 783- 791頁, 2001年) が報告されている。 また、 TREM- 2の リガンドとして、 リポ多糖 (LPS) 、 リポティコ酸 (LTA) 、 デキストラン硫酸が 示唆されている (非特許文献 2 Daws, M. R. et al. , J Immunol, 171巻, 594- 599頁, 2003年) 。 (Bauchon, A. et al., Nature, Vol. 410, pp. 1103-1107, 2001) ( TREM-2, like TREM-1, signals by interacting with DAP12) (Daws MR et al, Eur J Immunol, Vol. 31, pp. 783-791, 2001), induce dendritic cells or macrophages to induce the expression of CC chemokine receptor 7 (CCR7) and are involved in maturation of dendritic cells. (Bouchon, A. et al., J. Exp. Med. 194, 1111-1122, 2001), and involvement in NO production (Daws MR et al., Eur J Immunol, 31). Pp. 783-791, 2001. In addition, lipopolysaccharide (LPS), lipoticoic acid (LTA), and dextran sulfate have been suggested as ligands of TREM-2 (Non-Patent Document 2 Daws). , MR et al., J Immunol, 171: 594-599, 2003).
本発明者らは、 既に、 肥満およびインスリン抵抗性を呈するモデル動物である KKAyマウス (Nishimura, Μ·, Exp Animal, 18卷, 147 - 157頁, 1969年) の脂肪細 胞において、 発現の亢進している遺伝子群の中から、 TREM- 2を見出し、 TREM-2の 機能を抑制することにより糖尿病病態が改善されることを報告している (特許文 献 1 特願 2003 - 144204) 。 発明の開示 The present inventors have already a model animal exhibiting obesity and insulin resistance KKA y mice (Nishimura, Μ ·, Exp Animal , 18 Certificates, 147 - 157 p., 1969) in adipose cells, the expression TREM-2 has been found among the upregulated genes, and it has been reported that suppression of TREM-2 function improves diabetes (Patent Document 1: Japanese Patent Application No. 2003-144204). Disclosure of the invention
インスリン抵抗性を改善する安全で優れた医薬が切望されている。  There is a long-felt need for safe and superior medicines that improve insulin resistance.
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、 ガンダリ オシドが TREM - 2のリガンドであることを見出し、 この知見に基づいて、 さらに検 討を重ねた結果、 本発明を完成するに至った。  The present inventors have conducted intensive studies to solve the above-described problems, and as a result, have found that gandarioside is a ligand of TREM-2.Based on this finding, the present inventors have further studied and found that The invention has been completed.
すなわち、 本発明は、 ( 1 ) (a) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一 のアミノ酸配列を含有する蛋白質もしくはその部分べプチドまたはその塩、 およ び (b) 複合糖質を用いることを特徴とする、 該蛋白質またはその塩と該複合糠 質との結合性を変化させる化合物またはその塩のスクリーェング方法、 That is, the present invention (1) using (a) 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, and (b) using a glycoconjugate A method for screening a compound or a salt thereof that changes the binding property between the protein or a salt thereof and the complex bran,
( l a ) (a) 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 —のアミノ酸配列を含有する蛋白質もしくはその部分べプチドまたはその塩、 お よび (b) 複合糖質を用いることを特徴とする、 該蛋白質またはその塩と該複合 糖質との結合を阻害する化合物またはその塩のスクリ一二ング方法、  (la) (a) a protein containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide or a salt thereof, and (b) a glycoconjugate A method for screening a compound or a salt thereof that inhibits binding between the protein or a salt thereof and the glycoconjugate;
( 2 ) (a) 複合糖質を、 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蛋白質もしくはその部分ぺプチドまたは その塩に接触させた場合と、 (b) 複合糖質および試験化合物を、 該蛋白質もし くはその部分べプチドまたはその塩に接触させた場合における、 該複合糖質の該 蛋白質もしくはその部分べプチドまたはその塩に対する結合量を測定し、 比較す る、 上記 (1 ) 記載のスクリーニング方法、  (2) (a) contacting a glycoconjugate with 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; (B) measuring the amount of the glycoconjugate bound to the protein or its partial peptide or its salt when the glycoconjugate and the test compound are brought into contact with the protein or its partial peptide or its salt; Screening method according to (1) above,
( 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蛋白質もしくはその部分ぺプチドまたはその塩が、 該蛋白質 もしくはその部分べプチドまたはその塩をコードする D N Aを含有する形質転換 体を培養することによって細胞膜上に発現した蛋白質もしくはその部分べプチド またはその塩である上記 (1 ) 記載のスクリーニング方法、  (3) 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 is substituted with the protein or a partial peptide or a salt thereof; The screening method according to the above (1), wherein the protein or its partial peptide or a salt thereof is expressed on a cell membrane by culturing a transformant containing the encoding DNA,
( 4 ) 複合糖質が、 標識した複合糖質である上記 (1 ) 記載のスクリーニング 方法、  (4) The screening method according to the above (1), wherein the glycoconjugate is a labeled glycoconjugate.
( 5 ) (a) 複合糖質を、 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蛋白質もしくはその部分ぺプチドまたは その塩に接触させた場合と、 (b) 複合糖質および試験化合物を、 該蛋白質もし くはその部分ぺプチドまたはその塩に接触させた場合における、 該蛋白質もしぐ はその部分ペプチドまたはその塩を介した細胞刺激活性を測定し、 比較する、 上 記 (1 ) 記載のスクリーニング方法、  (5) (a) contacting the glycoconjugate with 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; (B) When a glycoconjugate and a test compound are brought into contact with the protein or its partial peptide or a salt thereof, the cell stimulating activity of the protein or its partial peptide or its salt via the partial peptide or its salt is measured. Comparing the screening method according to the above (1),
( 6 ) 複合糖質が、 ガンダリオシド、 シァリルオリゴ糖、 リポ多糖、 リポティコ 酸またはデキストラン硫酸である上記 (1 ) 〜 (5 ) 記載のスクリーニング方法、 (6 a) 複合糖質が、 ガンダリオシドまたはシァリルオリゴ糖である上記 (1) 〜 (5) 記載のスクリーニング方法、 (6) The screening method according to any one of the above (1) to (5), wherein the complex carbohydrate is gandarioside, sialyl oligosaccharide, lipopolysaccharide, lipotic acid or dextran sulfate. (6a) the screening method according to any one of the above (1) to (5), wherein the glycoconjugate is a gandarioside or a sialyl oligosaccharide;
(7) 複合糖質が、 ガングリオシドである上記 (1) 〜 (5) 記載のスクリー二 ング方法、  (7) The screening method according to the above (1) to (5), wherein the glycoconjugate is a ganglioside;
(7 a) 複合糖質が、 GM1、 GM2、 GM3、 GD 3または GD 1 aである上 記 (1) 〜 (5) 記載のスクリーニング方法、  (7a) The screening method according to any one of (1) to (5), wherein the glycoconjugate is GM1, GM2, GM3, GD3 or GD1a.
(8) 複合糖質が、 GM 3である上記 (1) 〜 (5) 記載のスクリーニング方法、 (8 a) 複合糖質が、 シァリルオリゴ糖である上記 (1) 〜 (5) 記載のスクリ 一ユング方法、  (8) The screening method according to any one of the above (1) to (5), wherein the glycoconjugate is GM3; (8a) the screening method according to the above (1) to (5), wherein the glycoconjugate is a sialyl oligosaccharide. Jung method,
(8 b) 複合糖質が、 3' -シァリルラクトース、 5,-シァリルラタトース、 シァリ ノレルイス X、 シァリルルイス A、 シァリルラクト -N-テトラオース a、 シァリルラタ ト- N-テトラオース b、 シァリルラタト- N-テトラオース c、 またはジシァリルラク ト -N-テトラオースである上記 (1) 〜 (5) 記載のスクリーニング方法、  (8b) When the glycoconjugate is 3'-sialyl lactose, 5, -sialyl ratatoose, sialyl oleylis X, sialyl lewis A, sialyl lacto-N-tetraose a, sialyl ratato-N-tetraose b, sialyl ratato-N- The screening method according to any one of the above (1) to (5), wherein the screening method is tetraose c or disialyl lacto-N-tetraose.
( 9 ) 配列番号: 1で表されるァミノ酸配列と実質的に同一のァミノ酸配列が、 配列番号: 2で表されるアミノ酸配列である上記 ( 1 ) 記載のスクリ一ユング方 法、  (9) The method according to (1), wherein the amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 1 is the amino acid sequence represented by SEQ ID NO: 2.
(10) (a) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同 一のアミノ酸配列を含有する蛋白質またはその塩および (b) 複合糖質を含有す ることを特徴とする、 該蛋白質またはその塩と該複合糖質との結合性を変化させ る化合物またはその塩のスクリーニング用キット、  (10) (a) a protein or a salt thereof having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1; and (b) a complex saccharide. A screening kit for a compound or a salt thereof that alters the binding property between the protein or a salt thereof and the glycoconjugate;
(1 1) 複合糖質の、 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 的に同一のァミノ酸配列を含有する蛋白質もしくはその部分べプチドまたはその 塩に対する活性化作用を阻害する化合物またはその塩を含有してなるインスリン 抵抗性改善剤、  (11) A compound that inhibits the activating action of glycoconjugates on 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 Or an insulin sensitizer comprising a salt thereof,
(1 2) 複合糖質の、 配列番号: 1で表されるァミノ酸配列と同一もしくは実質 的に同一のァミノ酸配列を含有する蛋白質もしくはその部分ぺプチドまたはその 塩に対する活性ィヒ作用を阻害する化合物またはその塩を含有してなる耐糖能異常、 糖尿病、 肥満症、 髙脂血症、 動脈硬化、 高血圧症または心疾患の予防 ·治療剤、 (1 3) 複合糖質の、 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'る蛋白質もしくはその部分べプチドまたはその 塩に対する活性ィヒ作用を促進する化合物またはその塩を含有してなる低血糖の予 防 ·治療剤、 (12) Inhibits the activity of glycoconjugates on 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 For preventing or treating impaired glucose tolerance, diabetes, obesity, lipidemia, arteriosclerosis, hypertension or heart disease, comprising a compound or a salt thereof, (1 3) glycoconjugate, SEQ ID NO: Identical or substantially identical to the amino acid sequence represented by 1 A prophylactic / therapeutic agent for hypoglycemia, comprising a compound or a salt thereof that promotes the activity of a protein or a partial peptide thereof or a salt thereof containing the same amino acid sequence.
(14) 複合糖質を含有してなる低血糖の予防 ·治療剤、  (14) a preventive / therapeutic agent for hypoglycemia comprising a complex carbohydrate,
(1 5) 複合糖質が、 ガングリオシド、 シァリルォリゴ糖、 リポ多糖、 リポティ コ酸またはデキストラン硫酸である上記 (1 4) 記載の予防 ·治療剤、  (15) The prophylactic or therapeutic agent according to the above (14), wherein the glycoconjugate is ganglioside, sialyloligosaccharide, lipopolysaccharide, lipotic acid or dextran sulfate.
(1 6) (a) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同 一のアミノ酸配列を含有する蛋白質もしくはその部分ぺプチドまたはその塩、 お よび (b) 複合糖質を用いることを特徴とする、 インスリン抵抗性改善作用を有 する化合物またはその塩のスクリ一二ング方法、  (16) (a) 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, and (b) a glycoconjugate. A method of screening a compound having an insulin sensitizing effect or a salt thereof, which comprises using the compound;
(1 7) (a) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同 一のァミノ酸配列を含有する蛋白質もしくはその部分べプチドまたはその塩、 お よび (b) 複合糖質を用いることを特徴とする、 糖尿病、 肥満症、 髙脂血症、 動 脈硬化症、 高血圧症または心疾患の予防 ·治療作用を有する化合物またはその塩 のスクリーニング方法、  (17) (a) 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, and (b) a glycoconjugate A method for screening a compound or a salt thereof having a preventive / therapeutic action for diabetes, obesity, hypolipidemia, arteriosclerosis, hypertension or heart disease,
(1 8) 複合糖質の、 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 的に同一のァミノ酸配列を含有する蛋白質もしくはその部分べプチドまたはその 塩に対する活性化作用を阻害することを特徴とするインスリン抵抗性改善方法、 (18) Inhibiting the activating action of glycoconjugates on 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 A method for improving insulin resistance,
(1 9) 上記 ( 1 ) 記載のスクリ一ユング方法または上記 (1 0) 記載のスクリ 一ユング用キットを用いて得られうる化合物またはその塩、 (19) a compound or a salt thereof obtainable by using the screening method according to (1) or the kit for screening according to (10);
(20) 化合物が、 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蛋白質もしくはその部分ぺプチドまたはその塩 と複合糖質との結合を阻害する化合物またはその塩である上記 (1 9) 記載の化 合物またはその塩、  (20) The compound, wherein the compound inhibits the binding 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 or a salt thereof to a glycoconjugate Or a salt thereof, or the compound according to the above (19) or a salt thereof;
(2 1) アンタゴニストである上記 (20) 記載の化合物またはその塩、 (22) ァゴニストである上記 (20) 記載の化合物またはその塩、  (21) the compound of the above (20), which is an antagonist, or a salt thereof; (22) the compound of the above (20), which is an agonist, or a salt thereof,
(2 3) 上記 (2 1) 記載の化合物またはその塩を含有してなるインスリン抵抗 性改善剤、  (23) an insulin sensitizer comprising the compound of the above (21) or a salt thereof,
(24) 上記 (2 1) 記載の化合物またはその塩を含有してなる耐糠能異常、 糖 尿病、 肥満症、 高脂血症、 動脈硬化症、 高血圧症または心疾患の予防 ·治療剤、 ( 2 5 ) 上記 (2 1 ) .記載の化合物またはその塩を含有してなる糖尿病の予防' 治療剤、 (24) Abnormal bran tolerance, sugar comprising the compound or salt thereof according to (21) above (25) A preventive or therapeutic agent for urinary disease, obesity, hyperlipidemia, arteriosclerosis, hypertension or heart disease, (25) prevention of diabetes comprising the compound or salt thereof according to (21). '' Therapeutic agents,
( 2 6 ) 上記 (2 2 ) 記載の化合物またはその塩を含有してなる低血糠の予防 治療剤などを提供する。 図面の簡単な説明 図 1は、 mTREM2- Fcに結合した蛍光ラベル GM3の定量結果を表す図である。 図中、 園は mTREM2- Fcが結合した Protein Gァガロースへの結合量を、 口は、 mTREM2_Fc を固定化していない Protein Gァガロースへの結合量を示す。 横軸は、 蛍光ラベ ル GM3の添加量を、 縦軸は、 溶出液に含まれる GM3の蛍光強度を示す。  (26) A prophylactic / therapeutic agent for low blood bran comprising the compound according to (22) or a salt thereof is provided. BRIEF DESCRIPTION OF THE DRAWINGS FIG. 1 shows the results of quantification of the fluorescent label GM3 bound to mTREM2-Fc. In the figure, the garden shows the amount of binding to Protein G agarose to which mTREM2-Fc was bound, and the mouth shows the amount of binding to Protein G agarose to which mTREM2_Fc was not immobilized. The horizontal axis shows the amount of the fluorescent label GM3 added, and the vertical axis shows the fluorescence intensity of GM3 contained in the eluate.
図 2は、 mTREMl- Fc、 mTREM2- Fc、 hTREMl- Fcおよび hTREM2- Fcに結合した蛍光ラ ベル GM3の定量結果を表す図である。 図中、 口は mTREMl- Fcへの結合量を、 園は mT FIG. 2 is a diagram showing the results of quantification of the fluorescent label GM3 bound to mTREM1-Fc, mTREM2-Fc, hTREM1-Fc and hTREM2-Fc. In the figure, the mouth indicates the amount bound to mTREMl-Fc, and the garden indicates mT
REM2- Fcへの結合量を、 △は hTREMl- Fcへの結合量を、 ▲は hTREM2- Fcへの結合量 を示す。 横軸は、 蛍光ラベル GM3の添加量を、 縦軸は、 溶出液に含まれる蛍光 GM3 の蛍光強度を示す。 発明を実施するための最良の形態 The amount of binding to REM2-Fc, Δ indicates the amount of binding to hTREM1-Fc, and indicates the amount of binding to hTREM2-Fc. The horizontal axis indicates the amount of the fluorescent label GM3 added, and the vertical axis indicates the fluorescence intensity of the fluorescent GM3 contained in the eluate. 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アミノ酸 配列を含有する蛋白質 (以下、 本発明の受容体、 本発明の蛋白質または本発明で 用いられる蛋白質と称することもある) は、 ヒトゃ温血動物 (例えば、 モルモッ ト、 ラット、 マウス、 ニヮトリ、 ゥサギ、 プタ、 ヒッジ、 ゥシ、 サルなど) の細 胞 (例えば、 肝細胞、 脾細胞、 神経細胞、 グリア細胞、 膝臓 細胞、 骨髄細胞、 メサンギゥム細胞、 ランゲルハンス細胞、 表皮細胞、 上皮細胞、 杯細胞、 内皮細 胞、 平滑筋細胞、 繊維芽細胞、 繊維細胞、 筋細胞、 脂肪細胞、 免疫細胞 (例、 マ クロファージ、 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、 瞎臓、 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 (例 赤血球、 白血球、 血小板) 、 前立腺、 睾丸、 卵巣、 胎盤、 子宮、 骨、 関節、 骨格筋、 白色脂肪組織、 褐色脂肪組織など に由来する蛋白質であってもよく、 合成蛋白質であってもよい。 A protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 (hereinafter sometimes referred to as the receptor of the present invention, the protein of the present invention, or the protein used in the present invention) Are cells of human warm-blooded animals (eg, guinea pigs, rats, mice, chickens, egrets, stags, hidges, horses, monkeys, etc.) (eg, hepatocytes, spleen cells, nerve cells, glial cells, knees) Organ 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 (eg, macrophages, T cells, B cells, natural killer cells, mast cells, neutrophils, basophils, eosinophils, monocytes), megakaryocytes, synovial cells, chondrocytes, bone cells, Osteoblasts, osteoclasts, mammary cells, hepatocytes or stromal cells, or their precursors, stem cells or cancer cells, or any tissue in which these cells are present, eg For example, brain, various parts of the brain (eg, olfactory bulb, amygdala, basal sphere, hippocampus, thalamus, hypothalamus, cerebral cortex, medulla, cerebellum), spinal cord, pituitary, stomach, spleen, kidney, liver, gonad , Thyroid, gall bladder, bone marrow, adrenal gland, skin, muscle, lung, digestive tract (eg, large intestine, small intestine), blood vessels, heart, thymus, spleen, submandibular gland, peripheral blood (eg, red blood cells, white blood cells, platelets), prostate It may be a protein derived from, testis, ovary, placenta, uterus, bone, joint, skeletal muscle, white adipose tissue, brown adipose tissue, or a synthetic protein.
配列番号: 1で表されるァミノ酸配列と実質的に同一のァミノ酸配列としては、 配列番号: 1で表されるァミノ酸配列と例えば約 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 includes, for example, about 60% or more, preferably about 70% or more, of the amino acid sequence represented by SEQ ID NO: 1. The amino acid sequence preferably has about 80% or more, preferably about 90% or more, and preferably about 95% or more homology.
アミノ酸配列の相同性は、 相同性計算アルゴリズム NCBI BLAST (National Center for Biotechnology Information Basic Local Alignment search Tool) を用い、 以下の条件 (期待値 = 10;ギヤップを許す;マトリタス =BL0SUM62; フ ィルタリング =0FF) にて計算することができる。  The amino acid sequence homology was determined using the homology calculation algorithm NCBI BLAST (National Center for Biotechnology Information Basic Local Alignment search Tool) under the following conditions (expected value = 10; allow gaps; Matritas = BL0SUM62; filtering = 0FF) Can be calculated.
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 る蛋白質としては、 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 する蛋白質と実質的に同質の活性を有する蛋白質などが好ましい。 配列番号: 1 で表されるァミノ酸配列と実質的に同一のァミノ酸配列を含有する蛋白質として は、 例えば、 配列番号: 2で表されるアミノ酸配列を含有する蛋白質などが挙げ ら; る。  Examples of the protein containing an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 include, for example, a protein containing the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 above. However, a protein having substantially the same activity as the protein having the amino acid sequence represented by SEQ ID NO: 1 is preferred. Examples of the protein containing the amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 1 include a protein containing the amino acid sequence represented by SEQ ID NO: 2, and the like.
実質的に同質の活性としては、 例えば、 シグナル伝達活性 〔例、 本発明の蛋白 質 (好ましくは TREM-2) の細胞内シグナル伝達活性など〕 、 リガンド結合活性 Examples of substantially equivalent activities include, for example, signal transduction activity (eg, intracellular signal transduction activity of the protein of the present invention (preferably TREM-2)), ligand binding activity
〔例、 本発明の蛋白質 (好ましくは TREM-2) とリガンドまたは低分子との結合活 性など〕 などが挙げられる。 実質的に同質とは、 それらの性質が性質的に (例、 生理学的に、 または薬理学的に) 同質であることを示す。 したがって; シグナル 伝達活性、 リガンド結合活性などが同等 (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、 蛋白質の分子量などの量的要素は異なつていてもよい。 シグナル伝達活性およびリガンド結合活性の測定は、 自体公知の方法、 例えば J. Exp. Med. 194巻, . 1111- 1122頁, 2001年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。 [Examples: binding activity between the protein of the present invention (preferably TREM-2) and a ligand or a small molecule, etc.]. Substantially identical indicates that the properties are qualitatively (eg, physiologically or pharmacologically) equivalent. Therefore, signal transduction activity, ligand binding 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). However, the quantitative factors such as the degree of these activities and the molecular weight of the protein may be different. The signal transduction activity and the ligand binding activity can be measured according to a method known per se, for example, the method described in J. Exp. Med. 194, .1111-1112, 2001 or a method analogous thereto. it can.
本発明の蛋白質 (好ましくは TREM-2) のシグナルは、 例えば TREM- 2または DAP12 (DNAX adaptor protein 12) にリン酸化を生じさせ、 ERK (extracellular signal-related protein) を活性化し、 炎症性サイトカイン (例、 TNF-ひ等) の 分泌を促進する。 従って、 上記シグナル伝達活性は、 例えば本発明の蛋白質発現 細胞 (例、 TREM- 2発現動物細胞) に対し、 必要に応じ (a) 微生物細胞破碎液、 微生物培養上清、 真核細胞破碎液、 真核細胞培養上清などリガンドが含まれる液、 (b) リガンド自身、 (c) 天然のリガンドと同等に本発明の蛋白質に結合活性を 有する物質、 または (d) 本発明の蛋白質 (例、 TREM-2) を活性化する抗体を添 加して、 (1) リン酸化 ERKの生成量、 (2) 細胞外に分泌生産される TNF- aの生 成量または (3) リン酸化 TREM-2の生成量を測定する。 The signal of the protein (preferably TREM-2) of the present invention causes, for example, phosphorylation of TREM-2 or DAP12 (DNAX adapter protein 12), activates ERK (extracellular signal-related protein), and induces inflammatory cytokines ( Eg, TNF-secretion). Therefore, the above-mentioned signal transduction activity is, for example, as compared to the protein-expressing cells of the present invention (eg, TREM-2 expressing animal cells), as necessary, (a) microbial cell lysate, microbial culture supernatant, eukaryotic cell lysate, A solution containing a ligand such as eukaryotic cell culture supernatant, (b) the ligand itself, ( c ) a substance having the same binding activity as the natural ligand to the protein of the present invention, or (d) a protein of the present invention (eg, By adding an antibody that activates TREM-2), (1) the amount of phosphorylated ERK produced, (2) the amount of extracellular TNF-a produced or (3) the phosphorylated TREM- Measure the yield of 2.
リガンドとしては、 後述する複合糖質などが用いられる。  As the ligand, a complex saccharide described later is used.
上記のリン酸化 ERKまたは TNF- aの生成量は、 抗リン酸化 ERK抗体または抗 TNF- ひ抗体を用いて、 公知の方法 (例、 ウェスタンブロッテイング法、 EIA法など) により測定できる。 リン酸化 TREM-2の生成量は、 抗 TREM- 2抗体および抗リン酸化 チロシン抗体を用いて、 またリン酸化 DAP12の生成量は抗 DAP12抗体おょぴ抗リン 酸ィ匕チ口シン抗体を用いて、 公知の方法 (例、 免疫沈降法、 ウェスタンブロット 法など) により測定できる。 TREM-2または DAP12の免疫沈降は、 それぞれの蛋白 質を、 例えば FLAG、 His, V5、 myc、 HA等のタグを付けた組換え型蛋白質として動 物細胞で発現し、 それぞれの抗タグ抗体で行うこともできる。  The amount of phosphorylated ERK or TNF-a produced can be measured by a known method (eg, Western blotting, EIA, etc.) using an anti-phosphorylated ERK antibody or an anti-TNF-A antibody. The amount of phosphorylated TREM-2 produced was measured using an anti-TREM-2 antibody and an anti-phosphorylated tyrosine antibody, and the amount of phosphorylated DAP12 was produced using an anti-DAP12 antibody and an anti-phosphorylated antibody. Can be measured by a known method (eg, immunoprecipitation method, Western blot method, etc.). Immunoprecipitation of TREM-2 or DAP12 is performed by expressing each protein in animal cells as a recombinant protein tagged with, for example, FLAG, His, V5, myc, HA, etc., and using each anti-tag antibody. You can do it too.
リガンド結合活性は、 本発明の蛋白質 (好ましくは TREM-2) とリガンドを用い て、 例えば、 免疫沈降法、 蛋白質ァフィ二ティー精製法、 酵母 Two- hybrid法など により測定できる。  The ligand binding activity can be measured using the protein (preferably TREM-2) of the present invention and a ligand, for example, by an immunoprecipitation method, a protein affinity purification method, a yeast two-hybrid method, or the like.
また、 本発明の受容体としては、 (1) (i) 配列番号: 1で表されるアミ.ノ酸 配列中の 1または 2個以上 (例えば 1〜 1 0 0個程度、 好ましくは 1〜 3 0個程 度、 好ましくは 1 〜 1 0個程度、 さらに好ましくは数 (1〜 5 ) 個) のアミノ酸 が欠失したアミノ酸配列、 (i i) 配列番号: 1で表されるアミノ酸配列に 1また は 2個以上 (例えば 1〜: L 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、 (iii) 配列番号: 1で表されるアミノ酸配列に 1または 2個以上 ' (例えば 1〜: L 0 0個程度、 好ましくは 1〜 3 0個程度、 好ましくは 1〜 1 0個 程度、 さらに好ましくは数 (1〜5 ) 個) のアミノ酸が挿入されたアミノ酸配列、 (iv) 配列番号: 1で表されるァミノ酸配列中の 1または 2個以上 (例えば 1〜 1 0 0個程度、 好ましくは 1〜3 0個程度、 好ましくは 1〜1 0個程度、 さらに 好ましくは数 (1〜5 ) 個) のアミノ酸が他のアミノ酸で置換されたアミノ酸配 列、 または (V) それらを組み合わせたアミノ酸配列を含有する蛋白質などのい わゆるムテイン、 (2) (i) 配列番号: 2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ァミノ 酸配列、 (ii) 配列番号: 2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) のアミノ酸が付カ卩したアミノ酸配列、 Further, the receptor of the present invention includes (1) (i) one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 1 (for example, about 1 to 100, preferably 1 to 100). About 30 amino acids, preferably about 1 to 10 amino acids, and more preferably about 1 to 5 amino acids, and (ii) 1 amino acid sequence represented by SEQ ID NO: 1. Also Has two or more amino acids (for example, 1 to: about L00 ', preferably about 1 to 30, preferably about 1 to 10, and more preferably about 1 to 5). Amino acid sequence, (iii) one or more amino acid sequences represented by SEQ ID NO: 1 (eg, 1 to: about L00, preferably about 1 to 30, preferably 1 to 1) An amino acid sequence having about 0, more preferably a number (1 to 5) amino acids inserted, and (iv) one or more amino acids in the amino acid sequence represented by SEQ ID NO: 1 (for example, 1 to 1) An amino acid sequence in which about 100, preferably about 1 to 30, preferably about 1 to 10, and more preferably a number (1 to 5) of amino acids have been substituted with another amino acid, or V) so-called muteins, such as proteins containing amino acid sequences combining them; (2) (i) SEQ ID NO: 2 1 or 2 or more in the amino acid sequence (for example, about 1 to 100, preferably about 1 to 30, preferably about 1 to 10, and more preferably about 1 to 5) (Ii) 1 or 2 or more amino acids in the amino acid sequence represented by SEQ ID NO: 2 (for example, about 1 to 100, preferably about 1 to 30; Is an amino acid sequence obtained by adding about 1 to 10 amino acids, more preferably a number (1 to 5) of amino acids;
(iii) 配列番号: 2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) のアミノ酸が揷入されたアミノ酸配列、 (iv) 配列 番号: 2で表されるアミノ酸配列中の 1または 2個以上 (例えば 1〜1 0 0個程 度、 好ましくは 1〜 3 0個程度、 好ましくは 1〜 1 0個程度、 さらに好ましくは 数 (1〜5 ) 個) のアミノ酸が他のアミノ酸で置換されたアミノ酸配列、 または (iii) 1 or 2 or more amino acids in the amino acid sequence represented by SEQ ID NO: 2 (for example, about 1 to 100, preferably about 1 to 30, preferably about 1 to 10, more preferably (Iv) one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 2 (for example, about 1 to 100 amino acids, preferably (1 to 5) amino acids) Is an amino acid sequence in which about 1 to 30 amino acids, preferably about 1 to 10 amino acids, and more preferably several (1 to 5) amino acids have been substituted with other amino acids; or
(V) それらを組み合わせたアミノ酸配列を含有する蛋白質などのいわゆるムテ インも含まれる。 (V) So-called muteins such as proteins containing 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.
本発明の受容体の具体例としては、 例えば、 配列番号: 1で表されるアミノ酸 配列を含有する蛋白質 (ヒ ト TREM - 2) 、 配列番号: 2で表されるアミノ酸配列を 含有する蛋白質 (マウス TREM - 2) などがあげられる。  Specific examples of the receptor of the present invention include, for example, a protein containing the amino acid sequence represented by SEQ ID NO: 1 (human TREM-2) and a protein containing the amino acid sequence represented by SEQ ID NO: 2 ( Mouse TREM-2).
本発明の受容体の部分ペプチド (以下、 本発明の部分ペプチドと称する場合が ある) としては、 後述の医薬等の'スクリーニング方法に用いることのできる部分 ペプチドであれば、 いかなるものであってもよく、 例えば、 本発明の蛋白質分子 のうち、 細胞膜の外に露出している部位であって、 実質的に同質のリガンド結合 活性などを有するものなどが用いられる。 Partial peptide of the receptor of the present invention (hereinafter sometimes referred to as the partial peptide of the present invention) ) May be any partial peptide that can be used in a screening method for a drug or the like described below. For example, the protein molecule of the present invention is exposed outside the cell membrane. Sites having substantially the same ligand binding activity and the like are used.
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 3 3〜1 4 7番目のアミノ酸配列を有するぺプチ ド、 配列番号: 2で表されるアミノ酸配列において第 1 3 3〜1 4 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蛋白 質の構成ァ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ぺプチドなどが用いられる。  Specifically, for the purpose of preparing the antibody of the present invention described below, a peptide having the amino acid sequence at positions 133 to 147 in the amino acid sequence represented by SEQ ID NO: 1, SEQ ID NO: And a peptide having the amino acid sequence at positions 133 to 147 in the amino acid sequence represented by 2. For example, at least 20 or more, preferably 50 or more, more preferably 70 or more, more preferably 100 or more, most preferably the constituent amino acid sequences of the protein used in the present invention. Is a peptide having an amino acid sequence of 200 or more.
また、 本発明の部分ペプチドは、 そのアミノ酸配列中の 1または 2個以上 (好 ましくは、 1〜1 0個程度、 さらに好ましくは数 (1〜5 ) 個) のアミノ酸が欠 失し、 または、 そのアミノ酸配列に 1または 2個以上 (好ましくは、 1〜2 0個 程度、 より好ましくは 1〜1 0個程度、 さらに好ましくは数 (1〜5 ) 個) のァ ミノ酸が付加し、 または、 そのアミノ酸配列に 1または 2個以上 (好ましくは、 :!〜 2 0個程度、 より好ましくは 1〜 1 0個程度、 さらに好ましくは数 ( 1〜 5 ) 個) のアミノ酸が揷入され、 または、 そのアミノ酸配列中の 1または 2個以 上 (好ましくは、 1〜1 0個程度、 より好ましくは数個、 さらに好ましくは 1〜 5個程度) のアミノ酸が他のアミノ酸で置換されていてもよい。  In addition, the partial peptide of the present invention lacks one or more (preferably about 1 to 10, preferably 1 to 5) amino acids in its amino acid sequence, Alternatively, one or more (preferably about 1 to 20, more preferably about 1 to 10, and more preferably about 1 to 5) amino acids are added to the amino acid sequence. Or 1 or 2 or more (preferably: about! 20, more preferably about 1 to 10, and more preferably a number (1 to 5)) of amino acids in the amino acid sequence. Or one or more (preferably about 1 to 10, more preferably several, and more preferably about 1 to 5) amino acids in the amino acid sequence are replaced with other amino acids. May be.
また、 本発明の部分ペプチドには、 本発明の受容体の細胞外ドメインも含む。 該細胞外ドメインとしては、 例えば、 配列番号: 1で表されるアミノ酸配列の第 1 4〜1 6 7番目または第 1 9〜1 7 3番目のアミノ酸配列を有するペプチド、 配列番号: 2で表されるアミノ酸配列の第 1 4〜1 7 0番目または第 1 9〜1 7 0番目のアミノ酸配列を有するペプチドなどが挙げられる。  The partial peptide of the present invention also contains the extracellular domain of the receptor of the present invention. Examples of the extracellular domain include a peptide having the 14th to 16th amino acid sequence or the 19th to 17th amino acid sequence of the amino acid sequence represented by SEQ ID NO: 1; And a peptide having an amino acid sequence at the 14th to 170th amino acid sequence or at the 19th to 170th amino acid sequence.
本発明の受容体および本発明の部分ぺプチドは、 ぺプチド標記の慣例に従って 左端が N末端 (ァミノ末端) 、 右端が C末端 (カルボキシル末端) である。 C末 端はカルボキシ (-C00H) 、 カルボキシレート (- C00— ) 、 アミド (-C0NH2) また はエステル (- C00R) であってもよい。 ここでエステルにおける Rとしては、 例えば、 メチル、 ェチル、 n—プロピル、 イソプロピルもしくは n—プチルなどの アルキル、 例えば、 シクロペンチル、 シクロへキシルなどの C3_8シクロアルキル、 例えば、 フエニル、 —ナフチノレな どの C6_12ァリール、 例えば、 ベンジル、 フエネチルなどのフエ二ルー アル キルもしくは α—ナフチルメチルなどの α—ナフチルー アルキルなどの C7_ 14ァラルキルのほか、 経口用エステルとして汎用されるピパロィルォキシメチル などが用いられる。 In the receptor of the present invention and the partial peptide of the present invention, the left end is the N-terminus (amino terminus) and the right end is the C-terminus (carboxyl terminus) according to the convention of peptide labeling. C-terminal carboxy (-C00H), carboxylate (- C00-), amide (-C0NH 2) or ester - may be a (C00R). Here, as R in the ester, e.g., methyl, Echiru, n- propyl, alkyl such as isopropyl or n- heptyl, for example, cyclopentyl, C 3 _ 8 cycloalkyl such as cyclohexyl, for example, phenyl, - a Nafuchinore which C 6 _ 12 Ariru Piparoiruo, for example, benzyl, which are widely used as well, an ester for oral administration of C 7 _ 14 Ararukiru such phenylene Lou al kills or α- naphthylmethyl etc. α- Nafuchiru alkyl such phenethyl Xymethyl is used.
本発明の受容体および本発明の部分べプチドが C末端以外にカルボキシ (また はカルボキシレート) を有している場合、 カルボキシがアミ ド化またはエステル 化されているものも本発明の受容体おょぴ本発明の部分ペプチドに含まれる。 こ の場合のエステルとしては、 例えば上記した C末端のエステルなどが用いられる c さらに、 本発明の受容体および本発明の部分ペプチドには、 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、 ァセチル などの アルカノィルなどの ァシルなど) で保護されているもの、 生体内 で切断されて生成する N末端のグルタミン残基がピログルタミン酸化したもの、 分子内のアミノ酸の側鎖上の置換基 (例えば、 _0H、 - SH、 アミノ基、 イミダゾー ル基、 インドール基、 グァニジノ基など) が適当な保護基 (例えば、 ホルミル、 ァセチルなどの C wアル力ノィルなどの ァシルなど) で保護されているもの、 あるいは糖鎖が結合したいわゆる糖蛋白質などの複合蛋白質なども含まれる。 本発明の受容体または本発明の部分ペプチドの塩としては、 生理学的に許容さ れる酸 (例、 無機酸、 有機酸) や塩基 (例、 アルカリ金属塩) などとの塩が用い られ、 とりわけ生理学的に許容される酸付加塩が好ましい。 このような塩として 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。 When the receptor of the present invention and the partial peptide of the present invention have carboxy (or carboxylate) other than at the C-terminus, those in which carboxy is amidated or esterified also include those of the present invention. It is included in the partial peptide of the present invention. As the ester in this case, for example, the above-mentioned C-terminal ester and the like are used. C Further, the receptor of the present invention and the partial peptide of the present invention include an N-terminal amino acid residue (eg, a methionine residue). The amino group is protected by a protecting group (for example, alkyl such as formyl or acetyl, etc.), the glutamine residue at the N-terminus generated by cleavage in vivo is pyroglutamine-oxidized, Substituents on the side chains of amino acids (for example, _0H, -SH, amino group, imidazole group, indole group, guanidino group, etc.) are suitable protecting groups (for example, C w alkynyl such as formyl, acetyl, etc.). And complex proteins such as so-called glycoproteins to which sugar chains are bound. As the salt of the receptor of the present invention or the partial peptide of the present invention, salts with physiologically acceptable acids (eg, inorganic acids, organic acids) and bases (eg, alkali metal salts) are used. Physiologically 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) And succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid and benzenesulfonic acid.
複合糖質 (以下、 本発明のリガンドと略記することもある) としては、 本発明 の受容体と特異的に結合するものであれば、 何れの物であってもよい。 また、 複 合糖質の糖鎖部分が本発明の受容体と特異的に結合する場合は、 該複合糖質の糖 鎖部分も、 本発明のリガンドに含まれる。 例えば、 本発明の受容体との結合の解 離定数が 1 0 n M以下、 好ましくは 2 M以下、 さらに好ましくは 1 M以下、 特に好ましくは 2 0 0 n M以下、 最も好ましくは 1 0 0 n M以下である物などが 挙げられる。 The glycoconjugate (hereinafter sometimes abbreviated as the ligand of the present invention) may be any as long as it specifically binds to the receptor of the present invention. When the sugar chain portion of the complex carbohydrate specifically binds to the receptor of the present invention, the sugar chain of the complex carbohydrate is A chain portion is also included in the ligand of the present invention. For example, the dissociation constant of the binding to the receptor of the present invention is 10 nM or less, preferably 2 M or less, more preferably 1 M or less, particularly preferably 200 nM or less, and most preferably 100 nM or less. nM or less.
本発明のリガンドとしては、 例えば、 ガンダリオシド (例、 GM1、 GM2、 GM3、 GD3、 GDla等) 、 シァリルォリゴ糖 (例、 3, -シァリルラタ トース、 5' -シァリル ラタ トース、 シァリルルイス X、 シァリルルイス A、 シァリルラク ト -N-テトラオ 一ス3、 シァリルラクト- N-テトラオース b、 シァリルラタト- N-テトラオース c、 ジシァリルラクト -N-テトラオース等) 、 リポ多糖 (LPS) (例、 大腸菌由来 LPS、 緑膿菌由来 LPS等) 、 リポティコ酸 (LTA) (例、 スタフイロコッカス ·ァウレゥ ス由来 LTA、 枯草菌由来 LTA等) 、 デキストラン硫酸などが挙げられる。  Examples of the ligand of the present invention include gandarioside (eg, GM1, GM2, GM3, GD3, GDla, etc.), sialyloligosugar (eg, 3, -sialylratose, 5′-sialylratathose, sialylluis X, sialylluis A, sialyllac) G-N-tetraoose 3, sialyl lacto-N-tetraose b, sialyl ratato-N-tetraose c, disialyl lacto-N-tetraose, etc., lipopolysaccharide (LPS) (eg, LPS from Escherichia coli, LPS from Pseudomonas aeruginosa, etc.) Lipotic acid (LTA) (eg, LTA derived from Staphylococcus aureus, LTA derived from Bacillus subtilis), dextran sulfate, and the like.
本発明のリガンドには、 (a) ガングリオシド、 シァリルォリゴ糖、 リポ多糖、 リポティコ酸またはデキストラン硫酸を構成する部分構造、 および (b) 上記 The ligand of the present invention includes (a) a partial structure constituting ganglioside, sialyloligosaccharide, lipopolysaccharide, lipoticoic acid or dextran sulfate, and (b)
(a) の部分構造を含有する化合物なども含まれる。 Compounds containing the partial structure (a) are also included.
本発明のリガンドとして好ましくは、 例えばガンダリオシド (例、 GM1、 GM2、 GM3、 GD3、 GDla等) 、 シァリルォリゴ糖 (例、 3, -シァリルラク トース、 5, -シ ァリルラタ トース、 シァリルルイス X、 シァリルルイス A、 シァリルラク ト- N-テ トラオース a、 シァリルラク ト- N-テトラオース b、 シァリルラク ト -N-テトラオ一 ス0、 ジシァリルラクト -N-テトラオース等) などである。 さらに好ましくは GM3 などである。  Preferred ligands of the present invention include, for example, gandariosides (eg, GM1, GM2, GM3, GD3, GDla, etc.), sialyloligosaccharides (eg, 3, -sialylactose, 5, -sialyllatase, sialyl Louis X, sialyl Lewis A, sialyl lactone) G-N-tetraose a, sialyl lacto-N-tetraose b, sialyl lacto-N-tetraose 0, disialyl lacto-N-tetraose, etc.). More preferably, it is GM3.
標識された複合糖質も、 本発明のリガンドに含まれる。  Labeled glycoconjugates are also included in the ligand of the present invention.
標識物質としては、 放射性同位元素 (例、 〔1251〕 、 〔1311〕 、 〔 〕 、 [14C] 、 〔32P〕 、 〔33P〕 、 〔35S〕 など) 、 蛍光物質 〔例、 シァニン蛍光色素 (例、 Cy2、The labeling substance, a radioactive isotope (e.g., [125 1], [131 1], [], [14 C], [32 P], [33 P] and [35 S]), fluorescent substances [e.g. , Cyanine fluorescent dye (eg, Cy2,
Cy3、 Cy5、 Cy5. 5、 Cy7 (アマシャムバイオサイエンス社製) など) 、 フルォレス カミン、 フルォレツセンイソチオシァネート、 NBD (7- nitrobenz- 2- oxa- 1, 3- diazol)など〕 、 酵素 (例、 β—ガラク トシダーゼ、 一ダルコシダーゼ、 アル カリフォスファタ一ゼ、 パーォキシダーゼ、 リンゴ酸脱水素酵素など) 、 発光物 質 (例、 ノレミノール、 ルミノール誘導体、 ルシフェリン、 ルシゲニンなど) 、 ビ ォチン、 ランタニド元素などがあげられる。 中でも、 蛍光物質が好ましい。 さら に NBDが好ましい。 Cy3, Cy5, Cy5.5, Cy7 (manufactured by Amersham Bioscience), fluorescamine, fluorescein isothiocyanate, NBD (7-nitrobenz-2-oxa-1,3-diazol), etc.), Enzymes (eg, β-galactosidase, mono-dalcosidase, alkaline phosphatase, peroxidase, malate dehydrogenase, etc.), luminescent substances (eg, noreminol, luminol derivatives, luciferin, lucigenin, etc.), biotin And lanthanide elements. Among them, a fluorescent substance is preferable. More NBD is preferred.
標識したリガンドとして好ましくは、 蛍光物質で標識されたガンダリオシド、 さらに好ましくは、 NBDで標識された GM3などが挙げられる。  Preferably, the labeled ligand is a gandarioside labeled with a fluorescent substance, and more preferably GM3 labeled with NBD.
本発明の受容体およぴ本発明の部分べプチドは、 前述したヒ トや温血動物の細 胞または組織から自体公知のポリぺプチドの精製方法によつて製造することもで きるし、 ポリべプチドをコ一ドする D N Aで形質転換された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、 ペプチド合成法に準じて製造す ることもできる。 例えば、 Genomics、 56巻、 12- 21頁、 1999年、 Biochim.  The receptor of the present invention and the partial peptide of the present invention can be produced from the above-mentioned cells or tissues of humans or warm-blooded animals by a known method for purifying polypeptides, It can also be produced by culturing a transformant transformed with DNA encoding the polypeptide. Further, it can be produced according to the peptide synthesis method. For example, Genomics, 56, 12-21, 1999, Biochim.
Biophys. Acta, 1446卷、 57- 70頁、 1999年などに記載の方法またはこれに準じた 方法により、 製造することもできる。 It can also be produced by the method described in Biophys. Acta, Vol. 1446, pp. 57-70, 1999 or a method analogous thereto.
ヒトゃ哺乳動物の組織または細胞から製造する場合、 ヒトゃ哺乳動物の組織ま たは細胞をホモジナイズした後、 酸などで抽出を行ない、 該抽出液を逆相クロマ トグラフィー、 イオン交換クロマトグラフィーなどのクロマトグラフィーを組み 合わせることにより精製単離することができる。  When producing from human or mammalian tissues or cells, the human or mammalian tissues or cells are homogenized and then extracted with an acid or the like, and the extract is subjected to reverse phase chromatography, ion exchange chromatography, etc. Purification and isolation can be performed by combining the above chromatography.
本発明の受容体または部分ペプチドもしくはそれらの塩の合成には、 通常、 市 販のポリぺプチド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、 クロロメチル榭脂、 ヒドロキシメチル樹脂、 ベンズヒ ドリルアミン樹月旨、 アミノメチル樹脂、 4一べンジルォキシベンジルアルコール樹脂、 4一メチルベ ンズヒ ドリルァミン樹脂、 P AM樹脂、 4—ヒ ドロキシメチルメチルフエュルァ 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 ' —ジメ トキ シフエ二ルーヒ ドロキシメチル) フエノキシ樹脂、 4一 (2, , 4, 一ジメ トキ シフエ二ル一 F m o cアミノエチル) フエノキシ樹脂などをあげることができる c このような樹脂を用い、 ひ一ァミノ基と側鎖官能基を適当に保護したアミノ酸を、 目的とするポリペプチドの配列通りに、 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ポリペプチドを切り出すと同時に各種保護 基を除去し、 さらに高希釈溶液中で分子内ジスルフィ ド結合形成反応を実施し、 目的のポリペプチド、 受容体、 部分ペプチドまたはそれらのアミ ド体を取得する c 上記した保護ァミノ酸の縮合に関しては、 ポリぺプチド合成に使用できる各種 活性化試薬を用いることができるが、 特に、 カルポジイミ ド類がよい。 カルポジ イミ ド類としては、 D C C、 Ν, 'Ν ' —ジイソプロピルカルポジイミ ド、 Ν—ェ チル一 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、 Η〇 B t , H O O B t ) とともに保護アミノ酸を直接樹脂に添加するかまたは、 対称酸無水物または H O B tエステルあるいは H O〇B tエステルとしてあらかじめ保護アミノ酸の活性 化を行なった後に樹脂に添加することができる。 For the synthesis of the receptor or partial peptide of the present invention or a salt thereof, a commercially available resin for synthesizing a polypeptide can be usually used. Examples of such a resin include chloromethyl resin, hydroxymethyl resin, benzhydrylamine resin, aminomethyl resin, 4-benzyloxybenzyl alcohol resin, 4-methylbenzylhydramine resin, PAM resin, and the like. 4-Hydroxymethylmethylphenylacetamide methyl resin, polyacrylamide resin, 4- (2 ', 4'-dimethyloxyphenyloxymethyl) phenoxy resin, 4- (2,, 4,1-dimethyl) Phenyl (Fmoc aminoethyl) phenoxy resin, etc. c. Using such a resin, an amino acid appropriately protected with a monoamino group and a side chain functional group can be synthesized according to the sequence of the target polypeptide. Is then condensed on a resin according to various condensation methods known per se. At the end of the reaction, the polypeptide is cleaved from the resin, and at the same time, various protecting groups are removed.In addition, an intramolecular disulfide bond formation reaction is carried out in a highly diluted solution to obtain the desired polypeptide, receptor, partial peptide or their amino acids. for condensation of the protected Amino acids c above to obtain the de body, it can be used a variety of activation reagents that can be used in polypeptide synthesis, in particular, it is Karupojiimi earth. Karposi Examples of imids include DCC, Ν, 'Ν'-diisopropyl carbodiimide, and ミ -ethyl ェ, 1- (3-dimethylaminoprolyl) carbodiimide. Protected amino acids can be added directly to the resin along with an antioxidant (eg, Η〇B t, HOOB t), or the protected amino acid can be pre-activated as a symmetric anhydride or HOB t ester or HO〇B t ester. After that, it can be added to the resin.
保護アミノ酸の活性化や樹脂との縮合に用いられる溶媒としては、 ポリべプチ ド縮合反応に使用しうることが知られている溶媒から適宜選択されうる。 例えば、 N , N—ジメチルホルムアミド, N, N—ジメチルァセトアミ ド, N—メチルビ ロリ ドンなどの酸アミ ド類、 塩ィ匕メチレン, クロ口ホルムなどのハロゲン化炭化 水素類、 トリフルォロエタノールなどのアルコール類、 ジメチルスルホキシドな どのスルホキシド類、 ピリジン, ジォキサン, テトラヒ ドロフランなどのエーテ ル類、 ァセトニトリル, プロピオ二トリルなどの二トリル類、 酢酸メチル, 酢酸 ェチルなどのエステル類あるいはこれらの適宜の混合物などが用いられる。 反応 温度はポリぺプチド結合形成反応に使用され得ることが知られている範囲から適 宜選択され、 通常約一 2 0 °C〜5 0 °Cの範囲から適宜選択される。 活性化された アミノ酸誘導体は通常 1 . 5〜4倍過剰で用いられる。 ニンヒ ドリン反応を用い たテストの結果、 縮合が不十分な場合には保護基の脱離を行なうことなく縮合反 応を繰り返すことにより十分な縮合を行なうことができる。 反応を繰り返しても 十分な縮合が得られないときには、 無水酢酸またはァセチルイミダゾールを用い て未反応アミノ酸をァセチル化することによって、 後の反応に影響を与えないよ うにすることができる。  The solvent used for activating the protected amino acid or condensing with the resin can be appropriately selected from solvents known to be usable for the polypeptide condensation reaction. For example, acid amides such as N, N-dimethylformamide, N, N-dimethylacetamide, N-methylvirolidone, halogenated hydrocarbons such as methylene chloride, methylform, and trifluoromethyl Alcohols such as ethanol, sulfoxides such as dimethyl sulfoxide, ethers such as pyridine, dioxane and tetrahydrofuran; nitriles such as acetonitrile and propionitrile; esters such as methyl acetate and ethyl acetate; A mixture or the like is used. The reaction temperature is appropriately selected from a range known to be usable for the polypeptide bond formation reaction, and is usually appropriately selected from a range of about 120 ° C to 50 ° C. The activated amino acid derivative is usually used in a 1.5 to 4-fold excess. As a result of the test using the ninhydrin reaction, if the condensation is insufficient, sufficient condensation can be performed by repeating the condensation reaction without removing the protecting group. When a sufficient condensation cannot be obtained even by repeating the reaction, unreacted amino acids can be acetylated using acetic anhydride or acetylimidazole to prevent the subsequent reaction from being affected.
原料のァミノ基の保護基としては、 例えば、 Z、 B o c、 t—ペンチルォキシ カルボ-ノレ、 イソボルニノレオキシカルボニル、 4—メ トキシベンジルォキシカノレ ボニル、 C 1 一 Z、 B r— Z、 ァダマンチルォキシ力ルボニル、 トリフルォロア セチノレ、 フタロイノレ、 ホノレミノレ、 2—ュト口フエニノレスノレフエ二ノレ、 ジフエ二ノレ ホスフイノチオイル、 F m o cなどが用いられる。  Examples of the protecting group for the starting amino group include Z, Boc, t-pentyloxycarbo-nore, isoborninoleoxycarbonyl, 4-methoxybenzyloxycanolebonyl, C11Z, Br-Z For example, adamantyl oxycarbonyl, trifluoroacetinole, phthaloynole, honoleminole, 2-butenophenorelesnorefueinole, difuinore phosphinochioil, Fmoc and the like can be used.
カルボキシル基は、 例えば、 アルキルエステル化 (例えば、 メチル、 ェチル、 プロピノレ、 プチノレ、 tーブチノレ、 シクロペンチノレ、 シクロへキシノレ、 シクロへプ チル、 シクロォクチル、 2—ァダマンチルなどの直鎖状、 分枝状もしくは環状ァ ルキルエステル化) 、 ァラルキルエステル化 (例えば、 ベンジルエステル、 4一 ニトロべンジノレエステノレ、 4ーメ トキシペンジノレエステノレ、 4一クロ口べンジノレ エステル、 ベンズヒ ドリルエステル化) 、 フエナシルエステル化、 ベンジルォキ シカルボニルヒ ドラジド化、 t一ブトキシカルポ-ルヒ ドラジド化、 トリチルヒ ドラジド化などによつて保護することができる。 The carboxyl group is, for example, alkyl esterified (for example, methyl, ethyl, propynole, petitnole, t-butynole, cyclopentynole, cyclohexynole, cyclohexyl). Linear, branched or cyclic alkyl esterification such as tyl, cyclooctyl, 2-adamantyl, etc., aralkyl esterification (eg, benzyl ester, 4-nitrobenzinoleestenole, 4-methoxypentinole) Estenole, 4-methylbenzene ester, benzhydryl ester), phenacyl ester, benzyloxycarbonyl hydrazide, t-butoxycarbol hydrazide, trityl hydrazide and the like.
セリンの水酸基は、 例えば、 エステル化またはエーテル化によって保護するこ とができる。 このエステル化に適する基としては、 例えば、 ァセチル基などの低 級 (Cw) 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 ルポ二ル基、 エトキシカルボニル基などの炭酸から誘導される基などが用いられ る。 また、 エーテルィ匕に適する基としては、 例えば、 ベンジル基、 テトラヒドロ ビラ二ル基、 t_プチル基などである。  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 (Cw) 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. It is possible. Examples of a group suitable for ethereal dan include a benzyl group, a tetrahydrovinyl group, a t_butyl group, and the like.
チロシンのフエノール性水酸基の保護基としては、 例えば、 B z l、 C l2_The protecting group of the phenolic hydroxyl group of tyrosine, for example, B zl, C l 2 _
B z l、 2—二トロベンジル、 B r— Z、 t一ブチルなどが用いられる。 Bzl, 2-nitrobenzyl, Br-Z, t-butyl and the like are used.
ヒスチジンのイミダゾールの保護基としては、 例えば、 To s、 4ーメ トキシ 一 2, 3, 6—トリメチルベンゼンスルホニル、 DNP、 ベンジルォキシメチル、 Examples of the protecting group for imidazole of histidine include Tos, 4-methoxy-12,3,6-trimethylbenzenesulfonyl, DNP, benzyloxymethyl,
Bum, B o c、 T r t、 Fmo cなどが用いられる。 Bum, Boc, Trt, Fmoc and the like are used.
原料のカルボキシル基の活十生化されたものとしては、 例えば、 対応する酸無水 物、 アジド、 活十生エステル 〔アルコール (例えば、 ペンタクロロフエノール、 2, 4, 5—トリクロロフエノ一ノレ、 2, 4—ジニトロフエノール、 シァノメチノレア ノレコーノレ、 パラ二ト口フエノール、 HONB、 N—ヒ ドロキシスクシミ ド、 N— ヒ ドロキシフタルイミ ド、 HOB t) とのエステル〕 などが用いられる。 原料の ァミノ基の活性化されたものとしては、 例えば、 対応するリン酸アミ ドが用いら れる。  Examples of the activated carboxyl groups of the raw material include, for example, corresponding acid anhydrides, azides, and activated decay esters [alcohols (eg, pentachlorophenol, 2,4,5-trichlorophenol, 2 , 4-dinitrophenol, cyanomethinorea, phenolic phenol, HONB, N-hydroxysuccinimide, N-hydroxyphthalimid, HOB tester). As the activated amino group of the raw material, for example, a corresponding phosphoric amide is used.
保護基の除去 (脱離) 方法としては、 例えば、 P d—黒あるいは P d—炭素な どの触媒の存在下での水素気流中での接触還元や、 また、 無水フッ化水素、 メタ ンスルホン酸、 トリフルォロメタンスルホン酸、 トリフルォロ酢酸あるいはこれ らの混合液などによる酸処理や、 ジィソプロピルェチルァミン、 トリェチルァミ ン、 ピぺリジン、 ピぺラジンなどによる塩基処理、 また液体アンモニア中ナトリ ゥムによる還元なども用いられる。 上記酸処理による脱離反応は、 一般に約一 2 0 °C〜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、 フエノーノレ、 チオアニソール、 メタクレゾーノレ、 パラクレゾーノレ、 ジメチノレスノレ フイ ド、 1 , 4ーブタンジチオール、 1 , 2 _エタンジチオールなどのような力 チオン捕捉剤の添加が有効である。 また、 ヒスチジンのイミダゾール保護基とし て用いられる 2 , 4—ジニトロフエニル基はチォフエノール処理により除去され、 トリブトファンのインドール保護基として用いられるホルミル基は上記の 1 , 2 一エタンジチオール、 1 , 4—ブタンジチオールなどの存在下の酸処理による脱 保護以外に、 希水酸化ナトリウム溶液、 希アンモニ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. Reduction by pum is also used. The elimination reaction by the above-mentioned acid treatment is generally carried out at a temperature of about 120 ° C to 40 ° C. In the acid treatment, for example, anisol, phenanol, thioanisole, methacrylone, paracrezonole, dimethinoresolefide, It is effective to add a force-thione scavenger such as 1,4-butanedithiol, 1,2-ethanedithiol and the like. Also, 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 tributofan is treated with the above 1,1,1-ethanedithiol, 1,4-butanedithiol. In addition to deprotection by acid treatment in the presence of, etc., it is also removed by alkali treatment with dilute sodium hydroxide solution, dilute ammonia, etc.
原料の反応に関与すべきでない官能基の保護ならびに保護基、 およびその保護 基の脱離、 反応に関与する官能基の活性化などは公知の基または公知の手段から 適宜選択しうる。  The protection of the functional group which should not be involved in the reaction of the raw materials, the protecting group, the elimination of the protective group, the activation of the functional group involved in the reaction, and the like can be appropriately selected from known groups or known means.
本発明の受容体または部分ペプチドを得る別の方法としては、 例えば、 まず、 カルボキシ末端アミノ酸のひ一カルボキシル基をアミ ド化して保護した後、 アミ ノ基側にペプチド (ポリペプチド) 鎖を所望の鎖長まで延ばした後、 該ペプチド 鎖の N末端のひ一ァミノ基の保護基のみを除いたポリべプチドと C末端のカルボ キシル基の保護基のみを除去したポリぺプチドとを製造し、 この両ポリぺプチド 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ポリペプチドを精製した後、 上記方法により すべての保護基を除去し、 所望の粗ポリぺプチドを得ることができる。 この粗ポ リベ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 とで所望の受容体またはその部分べプチドのァミ ド体を得ることができる。  As another method for obtaining the receptor or partial peptide of the present invention, for example, first, after protecting a single carboxyl group of the carboxy terminal amino acid by amidation, a peptide (polypeptide) chain is desired on the amino group side. Then, a polypeptide is obtained by removing only the protecting group of the N-terminal monoamino group of the peptide chain and a polypeptide from which only the protecting group of the C-terminal carboxyl group is removed. The two polypeptides are condensed in a mixed solvent as described above. The details of the condensation reaction are the same as described above. After purifying the protected polypeptide obtained by the condensation, all the protecting groups are removed by the above-mentioned method to obtain a desired crude polypeptide. This crude polypeptide is purified by various known purification means, and the main fraction is freeze-dried to obtain the desired receptor or an amide of the partial peptide.
本発明の受容体または部分べプチドもしくはそれらの塩のエステル体を得るに は、 例えば、 カルボキシ末端アミノ酸のひ一カルボキシル基を所望のアルコール 類と縮合しアミノ酸エステルとした後、 受容体またはその部分ペプチドのアミ ド 体と同様にして、 所望の受容体またはその部分ペプチドのエステル体を得ること ができる。  In order to obtain an ester of the receptor or partial peptide of the present invention or a salt thereof, for example, after condensing a monocarboxylic group of the carboxy terminal amino acid with a desired alcohol to form an amino acid ester, the receptor or a portion thereof is obtained. In the same manner as in the amide form of the peptide, an ester form of the desired receptor or a partial peptide thereof can be obtained.
本発明の受容体または部分べプチドは、 自体公知のぺプチドの合成法に従って、 あるいは受容体の部分べプチドにづいては、 受容体を適当なぺプチダーゼで切断 することによって製造することができる。 ペプチドの合成法としては、 例えば、 固相合成法、 液相合成法のいずれによっても良い。 すなわち、 本発明受容体また は部分ぺ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 (i) 〜 (V) に記載された方法があげられる。 The receptor or partial peptide of the present invention can be obtained by a known method for synthesizing a peptide. Alternatively, a partial peptide of the receptor can be produced by cleaving the receptor 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, a partial peptide or amino acid capable of constituting the receptor or partial peptide of the present invention is condensed with the remaining portion, and when the product has a protecting group, the protecting group is eliminated to remove the desired peptide. Can be manufactured. Examples of the known condensation method and elimination of the protecting group include the methods described in the following (i) to (V).
(i) M. Bodanszkyおよび M. A. 0ndetti、 Peptide Synthesis, Interscience Publishers, New York (1966年)  (i) M. Bodanszky and M.A. 0ndetti, Peptide Synthesis, Interscience Publishers, New York (1966)
(i i) Schroederおよぴ Luebke、 The Peptide, Academic Press, New York (1965 年)  (ii) Schroeder and Luebke, The Peptide, Academic Press, New York (1965)
(ii i) 泉屋信夫他、 ペプチド合成の基礎と実験、 丸善 (株) (1975年)  (ii 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、 蛋白質の化学 IV、 東京化 学同人(1977年)  (iv) Haruaki Yajima and Shunpei Sakakibara, Laboratory of Biochemistry 1, Protein Chemistry IV, Tokyo Kagaku Dojin (1977)
(v) 矢島治明監修、 続医薬品の開発、 第 14卷、 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、 カラムクロマトダラ フィ一、 液体クロマトグラフィ一、 再結晶などを組み合わせて本発明の受容体ま たは部分ぺプチドを精製単離することができる。 上記方法で得られる受容体また は部分べプチドが遊離体である場合は、 公知の方法あるいはそれに準じる方法に よって適当な塩に変換することができるし、 逆に塩で得られた場合は、 公知の方 法あるいはそれに準じる方法によって遊離体または他の塩に変換することができ る。  (v) Supervised by Haruaki Yajima, Development of Continuing Pharmaceuticals, Vol. 14, Peptide Synthesis, Hirokawa Shoten After the reaction, conventional purification methods such as solvent extraction, distillation, column chromatography, liquid chromatography, and liquid chromatography The receptor or partial peptide of the present invention can be purified and isolated by combining crystals and the like. When the receptor or partial peptide obtained by the above method is a free form, it can be converted into an appropriate salt by a known method or a method analogous thereto. It can be converted to a free form or another salt by a known method or a method analogous thereto.
本発明の受容体または部分ぺプチドをコ一ドするポリヌクレオチドとしては、 前述した本発明の受容体または部分ぺプチドをコ一ドする塩基配列を含有するも のであればいかなるものであってもよい。 このうち D N Aが好ましく、 該 D N A は、 ゲノム D NA、 ゲノム D NAライブラリー、 前記した細胞 ·組織由来の c D N A、 前記した細胞 ·組織由来の c D N Aライブラリー、 合成 D N Aのいずれで もよい。  The polynucleotide encoding the receptor or partial peptide of the present invention may be any polynucleotide containing the above-described nucleotide sequence encoding the receptor or partial peptide of the present invention. Good. Of these, DNA is preferable, and the DNA may be any of genomic DNA, genomic DNA library, cDNA derived from the above-described cells and tissues, cDNA library derived from the above-described cells and tissues, and synthetic DNA.
ライブラリーに使用するベクターは、 パクテリオファージ、 プラスミド、 コス ミド、 ファージミドなどいずれであってもよい。 また、 前記した細胞 · ,組織より total RNAまたは 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 vectors used for the library are pateriophage, plasmid, And phagemid. Alternatively, it can be directly amplified by reverse transcriptase polymerase chain reaction (hereinafter abbreviated as RT-PCR method) using a preparation of a total RNA or mRNA fraction from the cells and tissues described above.
本発明の受容体をコードする D NAとしては、 例えば配列番号: 3または配列 番号: 4で表される塩基配列を含有する D N A、 または配列番号: 3または配列 番号: 4で表される塩基配列とハイストリンジェントな条件下でハイプリダイズ する塩基配列を有し配列番号: 3または配列番号.: 4で表されるアミノ酸配列を 含有する蛋白質と実質的に同質の活性を有する受容体をコードする D N Aなどで あれば何れのものでもよい。  Examples of the DNA encoding the receptor of the present invention include a DNA containing the nucleotide sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4, or the nucleotide sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4 Encodes a receptor having a nucleotide sequence that hybridizes under high stringent conditions to a protein having substantially the same activity as a protein having the amino acid sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4. Any DNA may be used.
配列番号: 3または配列番号: 4で表される塩基配列とハイストリンジユント な条件下でハイブリダィズできる D N Aとしては、 例えば、 配列番号: 3または 配列番号: 4で表される塩基配列と例えば約 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塩基配列を含有する D N Aなどが用いられる。  Examples of the DNA that can hybridize with the nucleotide sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4 under high stringency conditions include, for example, the nucleotide sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4 DNA containing a base sequence having a homology of 60% or more, preferably about 70% or more, preferably about 80% or more, preferably about 90% or more, and preferably about 95% or more. Is used.
塩基配列の相同性は、 相同性計算アルゴリズム NCBI BLAST (National Center for Biotechnology Information Basic Local Alignment Search i'ool j を用い. 以下の条件 (期待値 = 10;ギャップを許す;フィルタリング =0N;マッチスコア = 1 ; ミスマッチスコア =-3) にて計算することができる。  The homology of the nucleotide sequences was determined using the homology calculation algorithm NCBI BLAST (National Center for Biotechnology Information Basic Local Alignment Search i'ool j. The following conditions (expected value = 10; gap allowed; filtering = 0N; match score = 1; mismatch score = -3).
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 は、 olecular Cloning 2nd Edition (J. Sambrook et al., Cold Spring  Hybridization is carried out by a method known per se or a method analogous thereto, for example, by the method of Molecular Cloning 2nd Edition (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、 市販のライプ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つ 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。 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, it can be performed under high stringent conditions.
ハイストリンジヱントな条件とは、 例えば、 ナトリウム濃度が約 1 9〜 4 0 m M、 好ましくは約 1 9〜 2 0 mMで、 温度が約 5 0〜 7 0 °C、 好ましくは約 6 0 〜6 5 °Cの条件を示す。 特に、 ナトリゥム濃度が約 1 9 mMで温度が約 6 5 の 場合が最も好ましい。 より具体的には、 配列番号: で表されるアミノ酸配列を含有する受容体をコ ードする DNAとしては、 配列番号: 3で表される塩基配列を含有する DNAな どが、 配列番号: 2で表されるアミノ酸配列を含有する受容体をコードする DN Aとしては、 配列番号: 4で表される塩基配列を含有する DNAなどが用いられ る。 The high stringent conditions include, for example, a sodium concentration of about 19 to 40 mM, preferably about 19 to 20 mM, and a temperature of about 50 to 70 ° C, preferably about 60 ° C. The condition of ~ 65 ° C is shown. In particular, the case where the sodium concentration is about 19 mM and the temperature is about 65 is most preferable. More specifically, examples of the DNA encoding the receptor containing the amino acid sequence represented by SEQ ID NO: include a DNA containing the base sequence represented by SEQ ID NO: 3, and the like. As the DNA encoding the receptor containing the amino acid sequence represented by 2, DNA containing the base sequence represented by SEQ ID NO: 4 or the like is used.
本発明の部分ぺプチドをコ一ドする DNAとしては、 本発明の受容体の部分べ 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 レ、。 また、 ゲノム DNA、 ゲノム DNAライブラリー、 前記した細胞'組織由来 の cDNA、 前記した細胞 '組織由来の c DNAライブラリー、 合成 DNAのい ずれでもよレ、。 具体的には、 配列番号: 3または配列番号: 4で表される塩基配 列を有する DNAの部分塩基配列を有する DNA、 または配列番号: 3または配 列番号: 4で表される塩基配列とハイストリンジヱントな条件下でハイブリダイ ズする塩基配列を有し、 配列番号: 1または配列番号: 2で表されるアミノ酸配 列を含有する蛋白質と実質的に同質の活性を有する受容体をコードする DN Aの 部分塩基配列を有する D N Aなどが用いられる。  The DNA encoding the partial peptide of the present invention may be any DNA containing a base sequence encoding the partial peptide of the receptor of the present invention. In addition, any of genomic DNA, genomic DNA library, cDNA derived from the above-described cells and tissues, cDNA library derived from the above-described cells and tissues, and synthetic DNA may be used. Specifically, a DNA having a partial base sequence of a DNA having the base sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4, or a base sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4 Encoding a receptor having a nucleotide sequence that hybridizes under high stringent conditions and having substantially the same activity as a protein containing the amino acid sequence represented by SEQ ID NO: 1 or SEQ ID NO: 2 DNA having a partial nucleotide sequence of DNA to be used is used.
配列番号: 3または配列番号: 4で表される塩基配列とハイブリダイズできる DNAは、 前記と同意義を示す。  The DNA hybridizable to the nucleotide sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4 has the same significance as described above.
ハイブリダイゼーションの方法およぴハイストリンジェントな条件は前記と同 様のものが用いられる。  The same hybridization method and high stringency conditions as described above are used.
本発明の受容体または部分ペプチドをコードするポリヌクレオチド (例、 DN A) は、 自体公知の方法で標識化されていてもよい。 標識物質としては、 放射性 同位元素、 蛍光物質 (例、 フルォレセインなど) 、 発光物質、 酵素、 ピオチン、 ランタ二ド元素などがあげられる。  The polynucleotide (eg, DNA) encoding the receptor or partial peptide of the present invention may be labeled by a method known per se. Labeling substances include radioisotopes, fluorescent substances (eg, fluorescein, etc.), luminescent substances, enzymes, biotin, lanthanide elements, and the like.
本発明の受容体または部分べプチドを完全にコードする DNAのクローニング の手段としては、 本発明の受容体または部分べプチドの部分塩基配列を有する合 成 DNAプライマーを用いて自体公知の PCR法によって増幅するか、 または適 当なベクターに組み込んだ DNAを本発明の受容体または部分べプチドの一部あ るいは全領域をコードする DNA断片もしくは合成 DNAを用いて標識したもの とのハイプリダイゼーションによって選別することができる。 ハイプリダイゼー シヨンの方法は、 例えば、 Molecuiar Cloning 2nd Edition (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。 As a means for cloning DNA that completely encodes the receptor or partial peptide of the present invention, a PCR method known per se using a synthetic DNA primer having a partial nucleotide sequence of the receptor or partial peptide of the present invention is used. Hybridization with DNA amplified or incorporated into an appropriate vector, labeled with a DNA fragment encoding a part or all of the receptor or partial peptide of the present invention or with synthetic DNA Can be sorted out. Hypridease Chillon methods are described, for example, Molecuiar Cloning 2 nd Edition (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989) can be carried out according to the method described in. When a commercially available library is used, the procedure can be performed according to the method described in the attached instruction manual.
DN Aの塩基酉己列の変換は、 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、 0M- LA PCR法、 Gapped duplex法、 Kunkel法等の自体公知の方法あるいはそれらに準じる方法に 従って行なうことができる。 Conversion of the base Rooster himself column of DN A is known kit, for example, Mutan ™ - super Express Km (Takara Shuzo Co., Ltd.), using a Mut an, ™ _K (Takara Shuzo) or the like, 0M- LA PCR method The method can be performed according to a method known per se, such as the gapped duplex method and the Kunkel method, or a method analogous thereto.
クローン化された受容体をコードする 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 DNAァダ プターを用いて付; D口することもできる。  The DNA encoding the cloned receptor can be used as it is depending on the purpose, or digested with a restriction enzyme or added with a linker as desired. The DNA may have ATG as a translation initiation codon at the 5 'end and TAA, TGA or TAG as a translation stop codon at the 3' end. These translation initiation codon and translation termination codon can be added using an appropriate synthetic DNA adapter;
本発明の受容体または部分ペプチドの発現ベクターは、 例えば、 (a) 本発明 の受容体または部分ぺプチドをコードする DN Aから目的とする DN A断片を切 り出し、 (b) 該 DNA断片を適当な発現ベクター中のプロモーターの下流に連 結することにより製造することができる。 '  The expression vector for the receptor or partial peptide of the present invention includes, for example, (a) cutting out a DNA fragment of interest from DNA encoding the receptor or partial peptide of the present invention, and (b) the DNA fragment Is ligated downstream of the promoter in an appropriate expression vector. '
ベクターとしては、 大腸菌由来のプラスミ ド (例、 pBR322, p BR 32 5, pUC 1 2, pUC 13) 、 枯草菌由来のプラスミ ド (例、 pUB 1 10, p TP 5, p C 1 94) 、 酵母由来プラスミ ド (例、■ p SH 1 9, p SH 1 5) . λファージなどのバクテリオファージ、 レトロゥイノレス, ワクシニアウイ ス, バキュロウィルスなどの動物ウィルスなどの他、 pAl— l l、 pXT l、 p R cZCMV、 p R c/R S V, p c DNA I /N e oなどが用いられる。  Plasmids derived from E. coli (eg, pBR322, pBR325, pUC12, pUC13), Bacillus subtilis-derived plasmids (eg, pUB110, pTP5, pC194), Plasmid derived from yeast (eg, ■ pSH19, pSH15). Bacteriophage such as λ phage, animal viruses such as retroinoles, vaccinia virus, baculovirus, etc., pAlll, pXTl, p RcZCMV, pRc / RSV, pcDNAI / Neo and the like are used.
本発明で用いられるプロモーターとしては、 遺伝子の発現に用いる宿主に対応 して適切なプロモーターであればいかなるものでもよい。 例えば、 動物細胞を宿 主として用いる場合は、 SRaプロモーター、 SV40プロモーター、 H I V ' LTRプロモーター、 CMVプロモーター、 HSV-TKプロモーターなどがあ げられる。 これらのうち、 CMV (サイ ト'メガロウィルス) プロモーター、 S R CKプロモ 一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 a cプロモーター、 r e cAプロモーター、 LPLプロモー ター、 l p pプロモーター、 T 7プロモーターなどが、 宿主がバチルス属菌であ る場合は、 SP〇 1プロモーター、 SPO 2プロモーター、 p e nPプロモータ 一など、 宿主が酵母である場合は、 PHO 5プロモーター、 PGKプロモーター、 GAPプロモーター、 ADHプロモーターなどが好ましい。 宿主が昆虫細胞であ る場合は、 ポリヘドリンプロモーター、 P 10プロモーターなどが好ましい。 発現ベクターには、 以上の他に、 所望によりェンハンサー、 スプライシングシ ダナル、 ポリ A付加シグナル、 選択マーカー、 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。 選択マーカーとしては、 例えば、 ジヒ ドロ葉酸還元酵素 (以下、 dh f r と略称する場合がある) 遺伝子 〔メソトレキセート (MTX) 耐性〕 、 アンピシ リン耐性遺伝子 (以下、 Amprと略称する場合がある) 、 ネオマイシン耐性遺 伝子 (以下、 N e oTと略称する場合がある、 G418耐性) 等があげられる。 特に、 d h f r遺伝子欠損チャイニーズハムスター細胞を用いて d.h 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 つても選択できる。 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 animal cells are used as host, SRa promoter, SV40 promoter, HIV 'LTR promoter, CMV promoter, HSV-TK promoter and the like can be mentioned. Among them, it is preferable to use a CMV (site's megalovirus) promoter, an SRCK promoter and the like. If the host is Escherichia, the trp promoter, lac promoter, recA promoter, LPL promoter, lpp promoter, T7 promoter, etc .; if the host is Bacillus, SP 属 1 promoter, SPO When the host is yeast, such as 2 promoters, penP promoters, etc., PHO5 promoter, PGK promoter, GAP promoter, ADH promoter and the like are preferable. When the host is an insect cell, a polyhedrin promoter, a P10 promoter and the like are preferable. In addition to the above, the expression vector may further contain, if desired, an enhancer, a splicing signal, a polyA addition signal, a selection marker, an SV40 replication origin (hereinafter sometimes abbreviated as SV40 ori), and the like. Can be used. As the selection marker, for example, dihydrofolate reductase (hereinafter sometimes abbreviated as dh fr) gene [methotrexate (MTX) resistance], ampicillin phosphorus resistant gene (hereinafter sometimes abbreviated as Amp r), Neomycin resistance gene (hereinafter sometimes abbreviated as Neo T , G418 resistance) and the like. In particular, when the dhfr gene is used as a selection marker using Chinese hamster cells deficient in the dhfr gene, 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、 本発明の受容体の N端末 側に付加する。 宿主がェシヱリヒア属菌である場合は、 P h o A · シグナル配列、 Omp A ·シグナル配列などが、 宿主がバチルス属菌である場合は、 α—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、 MF a ·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、 ィンシュリン ·シグナル配列、 ひ一インターフェロン ·シグナル 配列、 抗体分子 ·シグナル配列などがそれぞれ利用できる。  If necessary, a signal sequence suitable for the host is added to the N-terminal side of the receptor of the present invention. If the host is a genus Escherichia, a Pho A signal sequence, an Omp A signal sequence, etc., if the host is a Bacillus genus, an α-amylase. Signal sequence, a subtilisin signal sequence, etc. If the host is yeast, MFa signal sequence, SUC2 signal sequence, etc.If the host is an animal cell, insulin signal sequence, Hi-interferon signal sequence, antibody molecule, signal sequence, etc. Are available respectively.
このようにして構築された本発明の受容体または部分ぺプチドをコ一ドする D N Aを含有するベクターを用いて、 形質転換体を製造することができる。  A transformant can be produced using the thus-constructed DNA-containing vector encoding the receptor or partial peptide of the present invention.
宿主としては、 例えば、 ェシエリヒア属菌、 バチルス属菌、 酵母、 昆虫細胞、 昆虫、 動物細胞などが用いられる。 ェシェリヒァ属菌の具体例としては、 例えば、 ェシェリヒア ' コリ As the host, for example, Escherichia bacteria, Bacillus bacteria, yeast, insect cells, insects, animal cells, and the like are used. Specific examples of the genus Escherichia include, for example, Escherichia coli.
(Escherichia coli) K 12 · DH 1 [Proc. Natl. Acad. Sci. USA, 60 巻, 160(1968)〕 , JM103 [Nucleic Acids Research, 9巻, 309(1981)〕 , J A 221 [Journal of Molecular Biology, 120卷, 517 (1978)〕 , HB 101  (Escherichia coli) K 12 · DH 1 [Proc. Natl. Acad. Sci. USA, 60, 160 (1968)], JM103 [Nucleic Acids Research, 9, 309 (1981)], JA 221 [Journal of Molecular] Biology, 120, 517 (1978)], HB 101
[Journal of Molecular Biology, 41¾ 459 (1969) ) , C 600 [Genetics, 39 卷, 440(1954)〕 などが用いられる。  [Journal of Molecular Biology, 41¾459 (1969)), C 600 [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  Bacillus bacteria include, for example, Bacillus.
subtilis) MI 1 14 〔Gene, 24卷, 255 983)〕 , 207-21 [Journal of Biochemistry, 95卷, 87(1984)〕 などが用いられる。 subtilis) MI 114 (Gene, 24, 255 983)], 207-21 [Journal of Biochemistry, 95, 87 (1984)] and the like.
酵母としては、 例えば、 サッカロマイセス セレビシェ (Saccharomyces cerevisiae) AH 22, AH 22 R", NA87— 1 1A, DKD— 5D, 20 B— 12、 シゾサッカロマイセス ボンべ (Schizosaccharomyces pombe) NC YC 191 3, NCYC 2036、 ピキア パストリス (Pichia pastoris) K Μ71などが用いられる。  Examples of yeast include, for example, Saccharomyces cerevisiae AH 22, AH 22 R ", NA87-11A, DKD-5D, 20B-12, Schizosaccharomyces pombe NC YC 1913, NCYC 2036 And Pichia pastoris K # 71.
昆虫細胞としては、 例えば、 ウィルスが Ac NPVの場合は、 ョ トウガの幼虫 由来株化細胞 (Spodoptera frugiperda cell; S f 細胞) 、 Trichoplusia の 中腸由来の MG l細胞、 Trichoplusia niの卵由来の High Five™細胞、 Mamestra brassicae由来の細胞または Estigmena acrea由来の細胞などが用いられる。 ウイ ルスが BmNP Vの場合は、 蚕由来株化細胞 (Bombyx raori N細胞; BmN細 胞) などが用いられる。 該 S f 細胞としては、 例えば、 S f 9細胞 (ATCC CRL1711) 、 S f 21細胞 (以上、 Vaughn, J.L.ら、 イン · ヴイボ (In  As insect cells, for example, when the virus is Ac NPV, Spodoptera frugiperda cell lines (Spodoptera frugiperda cells), MG1 cells derived from the midgut of Trichoplusia, and High cells derived from eggs of Trichoplusia ni Five ™ cells, cells derived from Mamestra brassicae or cells derived from Estigmena acrea are used. When the virus is BmNPV, a silkworm-derived cell line (Bombyx raori N cell; BmN cell) is used. Examples of the Sf cell include Sf9 cell (ATCC CRL1711), Sf21 cell (Vaughn, J.L., et al., In Vivo).
Vivo) ,13, 213-217, (1977)) などが用いられる。 Vivo), 13, 213-217, (1977)).
昆虫としては、 例えば、 カイコの幼虫などが用いられる 〔前田ら、 Nature, 315 巻, 592(1985)〕 。  As insects, for example, silkworm larvae are used [Maeda et al., Nature, Vol. 315, 592 (1985)].
動物細胞としては、 例えば、 サル細胞 COS— 7 (COS 7) , Ve r o, チ ャィニーズハムスター細胞 CHO (以下、 CH〇細胞と略記) , dh f r遺伝子 欠損チャイニーズハムスター細胞 CHO (以下、 CHO (d h f r— ) 細胞と略 記) , マウス L細胞, マウス A t T— 20, マウスミエローマ細胞, ラット GH 3, ヒト FL細胞などが用いられる。 ェシエリヒア属菌を形質転換するには、 例えば、 Proc. Natl. Acad. Sci. Examples of animal cells include monkey cells COS-7 (COS 7), Vero, Chinese hamster cells CHO (hereinafter abbreviated as CH〇 cells), dh fr gene-deficient Chinese hamster cells CHO (hereinafter CHO ( dhfr-) cells), mouse L cells, mouse AtT-20, mouse myeloma cells, rat GH3, and human FL cells. To transform Escherichia sp., For example, Proc. Natl. Acad. Sci.
USA, 69巻, 2110 (1972)や Gene, 17卷, 107 (1982)などに記載の方法に従って行なうこ とができる。 USA, 69, 2110 (1972) or Gene, 17, 107 (1982).
バチルス属菌を形質転換するには、 例えば、 Molecular & General  To transform Bacillus, for example, use Molecular & General
Genetics, 168巻, 111 (1979)などに記載の方法に従って行なうことができる。 Genetics, Vol. 168, 111 (1979) can be used.
酵母を形質転換するには、 例えば、 Methods in Enzymology, 194卷, 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って行なうことができる。  Transformation of insect cells or insects can be performed, for example, according to the method described in Bio / Technology, 6, 47-55 (1988).
動物細胞を形質転換するには、 例えば、 細胞工学別冊 8 新細胞工学実験プロ トコール. 263-267 (1995) (秀潤社発行) 、 Virology, 52巻, 456 (1973)に記載の方 法に従って行なうことができる。  Transformation of animal cells can be performed, for example, according to the method described in Cell Engineering Separate Volume 8 New Cell Engineering Experimental Protocol. 263-267 (1995) (published by Shujunsha), Virology, 52, 456 (1973). Can do it.
このようにして、 受容体または部分ぺプチドをコードする D N Aを含有する発 現ベクターで形質転換された形質転換体を得ることができる。  Thus, a transformant transformed with the expression vector containing DNA encoding the receptor or the partial peptide can be obtained.
宿主がエシ リヒア属菌、 バチルス属菌である形質転換体を培養する際、 培養 に使用される培地としては液体培地が適当であり、 その中には該形質転換体の生 育に必要な炭素源、 窒素源、 無機物その他が含有せしめられる。 炭素源としては、 例えば、 グルコース、 デキス トリン、 可溶性澱粉、 ショ糖など、 窒素源としては、 例えば、 アンモニゥム塩類、 硝酸塩類、 コーンスチープ' リカー、 ペプトン、 力 ゼイン、 肉エキス、 大豆粕、 パレイショ抽出液などの無機または有機物質、 無機 物としては、 例えば、 塩化カルシウム、 リン酸二水素ナトリウム、 塩化マグネシ ゥムなどがあげられる。 また、 酵母エキス、 ビタミン類、 成長促進因子などを添 加してもよい。 培地の p Hは約 5〜 8が望ましい。  When culturing a transformant whose host is a genus Escherichia or Bacillus, a liquid medium is suitable as a medium used for the culturing, and a carbon medium necessary for the growth of the transformant is contained therein. Sources, nitrogen sources, minerals and others. Examples of the carbon source include glucose, dextrin, soluble starch, and sucrose. Examples of the nitrogen source include ammonium salts, nitrates, corn chip liquor, peptone, potato zein, meat extract, soybean meal, and potato extract. Inorganic or organic substances such as liquids and inorganic substances include, for example, calcium chloride, sodium dihydrogen phosphate, magnesium chloride and the like. Also, yeast extract, vitamins, growth promoting factors and the like may be added. The pH of the medium is preferably about 5-8.
ェシエリヒア属菌を培養する際の培地としては、 例えば、 グルコース、 カザミ ノ酸を含む M 9培地 〔ミラー (Miller) , Journal of Experiments in  As a medium for cultivating a bacterium belonging to the genus Escherichia, for example, an M9 medium containing glucose and casamino acids [Miller, Journal of Experiments in
Molecular Genetics, 431 - 433, Cold Spring Harbor Laboratory, New York 1972〕 が好ましい。 ここに必要によりプロモーターを効率よく働かせるために、 例えば、 3 一インドリルアクリル酸のような薬剤を加えることができる。 宿主がェシェリヒァ属菌の場合、 培養は通常約 1 5〜 4 3 °Cで約 3〜 2 4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。 Molecular Genetics, 431-433, Cold Spring Harbor Laboratory, New York 1972]. If necessary, an agent such as 3-indolylacrylic acid can be added to make the promoter work efficiently. When the host is a bacterium belonging to the genus Escherichia, the cultivation is usually carried out at about 15 to 43 ° C for about 3 to 24 hours, and if necessary, aeration and stirring can be applied.
宿主がバチルス属菌の場合、 培養は通常約 3 0〜 4 0 °Cで約 6〜 2 4時間行な レ、、 必要により通気や撹拌を加えることもできる。  When the host is a bacterium belonging to the genus Bacillus, the cultivation is usually carried out at about 30 to 40 ° C for about 6 to 24 hours, and if necessary, aeration and stirring may be added.
宿主が酵母である形質転換体を培養する際、 培地としては、 例えば、 バークホ 一ノレダー (Burkholder) 最小培地 [Bostian, K. L. ら、 Proc. Natl. Acad. Sci. USA, 77卷, 4505 (1980)〕 や 0 . 5 %カザミノ酸を含有する S D培地 〔Bitter, G. A. ら、 Proc. Natl. Acad. Sci. USA, 81卷, 5330 (1984)〕 があげられる。 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 2 0 °C〜 3 5 °Cで約 2 4〜 7 2時間行ない、 必要に応じて通気や撹拌を加える。  When culturing a transformant in which the host is yeast, for example, Burkholder's minimum medium [Bostian, KL et al., Proc. Natl. Acad. Sci. USA, 77, 4505 (1980) And SD medium containing 0.5% casamino acid [Bitter, GA et al., Proc. Natl. Acad. Sci. USA, 81, 5330 (1984)]. Preferably, the pH of the medium is adjusted to about 5-8. The cultivation is usually performed at about 20 ° C to 35 ° C for about 24 to 72 hours, and aeration and stirring are added as necessary.
宿主が昆虫細胞または昆虫である形質転換体を培養する際、 培地としては、 Grace' s Insect Medium (Grace, T. C. C. , Nature, 195, 788 (1962) ) に非動化した 1 0 %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 p Hは約 6 . 2〜 6 . 4に調整するのが好ましい。 培養は通常約 2 7 °Cで約 3〜 5日間行 ない、 必要に応じて通気や撹拌を加える。  When culturing an insect cell or a transformant in which the host is an insect, the medium used is 10% serum serum immobilized in Grace's Insect Medium (Grace, TCC, Nature, 195, 788 (1962)). And the like to which additives such as the above are appropriately added 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 〜2 0 %の胎児牛血清を含む M E M培地 [Science, 122卷, 501 (1952)〕 , DME M培地 [: Virology, 8卷, 396 (1959)〕 , R P M I 1 6 4 0培地 〔The Journal of the American Medical Association 199卷, 519 (1967)〕 , 1 9 9培地  When culturing a transformant in which the host is an animal cell, for example, a MEM medium containing about 5 to 20% fetal bovine serum [Science, 122 vol., 501 (1952)], a DMEM medium [ Virology, 8 volumes, 396 (1959)], RPMI 1640 medium [The Journal of the American Medical Association 199 volumes, 519 (1967)], 199 medium
[Proceeding of the Society for the Biological Medicine, 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 °C〜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)]. Preferably, the pH is about 6-8. Cultivation is usually carried out at about 30 ° C to 40 ° C for about 15 to 60 hours, and aeration and stirring are added as necessary.
以上のようにして、 形質転換体の細胞内、 細胞膜または細胞外などに本発明の 受容体または部分べプチドを生成せしめることができる。  As described above, the receptor or partial peptide of the present invention can be produced in the cells, cell membrane, or extracellular cells of the transformant.
上記培養物から本発明の受容体または部分ぺプチド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 receptor or partial peptide of the present invention can be separated and purified from the above culture by, for example, the following method.
本発明の受容体または部分ペプチド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ポリべプチド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™などの界面活性剤が含まれていてもよい。 培養液中に ポリペプチド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ぁ るいは細胞と上清とを分離し、 上清を集める。 When extracting the receptor or partial peptide of the present invention from cultured cells or cells, after culturing, cells or cells are collected by a known method, and the cells or cells are suspended in an appropriate buffer, and then subjected to ultrasound, Cells or by lysozyme and / or freeze-thaw After the cells are disrupted, a method of obtaining a crude polypeptide extract by centrifugation or filtration is used as appropriate. The buffer may contain a protein denaturant such as urea or guanidine hydrochloride, or a surfactant such as Triton X-100 ™. When the polypeptide 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受容体または 部分ぺプチドの精製は、 自体公知の分離 ·精製法を適宜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 リルァミ ドゲル電気泳動法などの主として分子量の差を利用する方法、 ィオン交 換ク口マトグラフィーなどの荷電の差を利用する方法、 ァフィ二ティーク口マト グラフィ一などの特異的親和性を利用する方法、 逆相高速液体ク口マトグラフィ 一などの疎水性の差を利用する方法、 等電点電気泳動法などの等電点の差を利用 する方法などが用いられる。  The receptor or partial peptide contained in the culture supernatant or the extract obtained in this way can be purified by appropriately combining known separation and purification methods. These known separation and purification methods mainly include methods utilizing solubility such as salting out and solvent precipitation, dialysis, ultrafiltration, gel filtration, and SDS-polyacrylamide gel electrophoresis. Method using difference in molecular weight, method using charge difference such as ion exchange chromatography, method using specific affinity such as affinity chromatography, reverse phase high-performance liquid chromatography For example, a method utilizing a difference in hydrophobicity, such as a method of isoelectric point electrophoresis, or a method utilizing a difference in isoelectric point, such as isoelectric focusing electrophoresis, may be used.
かくして得られる受容体または部分ペプチド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つ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 thus obtained receptor or partial peptide is obtained as a free form, it can be converted into a salt by a method known per se or a method analogous thereto, and conversely, when the receptor or a partial peptide is obtained as a salt, It can be converted into a free form or another salt by a known method or a method analogous thereto.
なお、 組換え体が産生する受容体または部分ペプチドを、 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、 ポリべ プチドを部分的に除去することもできる。 蛋白修飾酵素としては、 例えば、 トリ プシン、 キモトリプシン、 アルギニルェンドぺプチダーゼ、 プロティンキナーゼ, グリコシダーゼなどが用いられる。  Receptors or partial peptides produced by recombinants can be arbitrarily modified or polypeptides can be partially removed before or after purification by the action of an appropriate protein-modifying enzyme. it can. As the protein modifying enzyme, for example, trypsin, chymotrypsin, arginyl endopeptidase, protein kinase, glycosidase and the like are used.
本発明のリガンドは、 市販されている場合には市販品をそのまま用いることも でき、 自体公知の方法またはこれらに準じた方法に従って抽出または製造するこ ともできる。  When the ligand of the present invention is commercially available, a commercially available product can be used as it is, or can be extracted or produced according to a method known per se or a method analogous thereto.
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 配列を含有する蛋白質もしくはその部分べプチドまたはその塩に対する抗体 (以 下、 単に本発明の抗体と称する場合がある) は、 本発明の受容体に対する抗体を 認識し得る抗体であれば、 ポリクローナル抗体、 モノクローナル抗体の何れであ つてもよい。 本発明の受容体に対する抗体としては、 受容体のシグナル伝達を不 活性化する抗体、 受容体のシグナル伝達を活性化する抗体などが挙げられる。 本発明の受容体に対する抗体は、 本発明の受容体を抗原として用い、 公知の抗 体または抗血清の製造法に従つて製造することができる。 An antibody against 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 (hereinafter, may be simply referred to as the antibody of the present invention) Is an antibody against the receptor of the present invention. Any antibody that can be recognized may be a polyclonal antibody or a monoclonal antibody. Examples of the antibody against the receptor of the present invention include an antibody that inactivates signal transduction of a receptor, an antibody that activates signal transduction of a receptor, and the like. An antibody against the receptor of the present invention can be produced according to a known method for producing an antibody or antiserum using the receptor of the present invention as an antigen.
〔モノクローナル抗体の作製〕 [Preparation of monoclonal antibody]
(a) モノクローナル抗体産生細胞の作製  (a) Preparation of monoclonal antibody-producing cells
本発明の受容体は、 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 2〜10回程度行われる。 用い られる温血動物としては、 例えば、 サル、 ゥサギ、 ィヌ、 モルモット、 マウス、 ラット、 ヒッジ、 ャギ、 ニヮトリがあげられるが、 マウスおよびラットが好まし く用いられる。  The receptor of the present invention is administered to a warm-blooded animal itself or together with a carrier or diluent at a site where antibody production is possible upon administration. Complete Freund's adjuvant / incomplete Freund's adjuvant may be administered in order to enhance the antibody-producing ability upon administration. Administration is usually performed once every 2 to 6 weeks, for a total of about 2 to 10 times. Examples of warm-blooded animals to be used include monkeys, rabbits, rabbits, dogs, guinea pigs, mice, rats, sheep, goats, and chickens, and mice and rats are preferably used.
モノクローナル抗体産生細胞の作製に際しては、 抗原で免疫された温血動物、 例えばマウスから抗体価の認められた個体を選択し最終免疫の 2〜 5日後に脾臓 またはリンパ節を採取し、 それらに含まれる抗体産生細胞を同種または異種動物 の骨髄腫細胞と融合させることにより、 モノクローナル抗体産生ハイプリ ドーマ を調製することができる。 抗血清中の抗体価の測定は、 例えば、 後記の標識化ポ リぺプチド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 〔ネイチヤー (Nature)、 256、 495 (1975)〕 に従い実施する ことができる。 融合促進剤としては、 例えば、 ポリエチレングリコール (PE G) やセンダイウィルスなどがあげられるが、 好ましくは PEGが用いられる。 骨髄腫細胞としては、 例えば、 NS_1、 P 3U1、 SP2Z0、 AP—lな どの温血動物の骨髄腫細胞があげられるが、 P 3U1が好ましく用いられる。 用 いられる抗体産生細胞 (脾臓細胞) 数と骨髄腫細胞数との好ましい比率は 1 : 1 〜20 : 1程度であり、 PEG (好ましくは PEG1000〜PEG6000) が 1 0〜 8 0 %程度の濃度で添加され、 2 0〜 4 0 ° (:、 好ましくは 3 0〜 3 7 °C で 1〜 1 0分間イン^ュベートすることにより効率よく細胞融合を実施できる。 モノクローナル抗体産生ハイプリ ドーマのスクリーニングには種々の方法が使 用できるが、 例えば、 ポリペプチド (蛋白質) 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 口ティン Aを加え、 固相に結合したモノクローナレ抗体を検出する方法、 抗免疫 グロブリン抗体またはプロテイン Aを吸着させた固相にハイブリ 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ポリペプチドを加え、 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あげられる。 When preparing monoclonal antibody-producing cells, a warm-blooded animal immunized with an antigen, for example, an individual with an antibody titer is selected from a mouse, and the spleen or lymph node is collected 2 to 5 days after the final immunization and included in them. By fusing the antibody-producing cells obtained with myeloma cells of the same or different species, a monoclonal antibody-producing hybridoma can be prepared. The antibody titer in the antiserum can be measured, for example, by reacting the labeled polypeptide described below with the antiserum, and then measuring the activity of the labeling agent bound to the antibody. The fusion operation can be performed according to a known method, for example, the method of Koehler and Milstein [Nature, 256, 495 (1975)]. Examples of the fusion promoter include polyethylene glycol (PEG) and Sendai virus, and PEG is preferably used. Examples of myeloma cells include myeloma cells of warm-blooded animals such as NS_1, P3U1, SP2Z0, and AP-1, and P3U1 is preferably used. The preferred ratio between the number of antibody-producing cells (spleen cells) used and the number of myeloma cells used is about 1: 1 to 20: 1, and PEG (preferably PEG1000 to PEG6000) Is added at a concentration of about 10 to 80%, and efficient cell fusion is performed by incubating at 20 to 40 ° (preferably at 30 to 37 ° C for 1 to 10 minutes). A variety of methods can be used to screen monoclonal antibody-producing hybridomas. For example, hybridoma culture supernatants can be applied to a solid phase (eg, microplate) on which polypeptide (protein) antigen is adsorbed directly or together with a carrier. Then, add an anti-immunoglobulin antibody (anti-mouse immunoglobulin antibody is used if the cells used for cell fusion are mice) or peptide A, labeled with a radioactive substance or enzyme, and then add the solid phase. For detection of monoclonal antibody bound to DNA, Add hybridoma culture supernatant to solid phase to which anti-immunoglobulin antibody or protein A is adsorbed, and release In addition a polypeptide labeled with a sex substance or an enzyme, such as a method of detecting a monoclonal antibody bound to the solid phase.
モノクロ一ナル抗体の選別は、 公知あるいはそれに準じる方法に従って行なう ことができる。 通常 H A T (ヒポキサンチン、 アミノプテリン、 チミジン) を添 加した動物細胞用培地で行なうことができる。 選別および育種用培地としては、 ハイプリ ドーマが生育できるものならばどのような培地を用いても良い。 例えば、 :!〜 2 0 %、 好ましくは 1 0〜 2 0 %の牛胎児血清を含む R P M I 1 6 4 0培 地、 1〜 1 0 %の牛胎児血清を含む G I T培地 (和光純薬工業 (株) ) あるいは ハイプリ ドーマ培養用無血清培地 (S F M— 1 0 1、 日水製薬 (株) ) などを用 いることができる。 培養温度は、 通常 2 0〜4 0 °C、 好ましくは約 3 7 °Cである。 培養時間は、 通常 5日〜 3週間、 好ましくは 1週間〜 2週間である。 培養は、 通 常 5 %炭酸ガス下で行なうことができる。 ハイプリ ドーマ培養上清の抗体価は、 上記の抗血清中の抗体価の測定と同様にして測定できる。  Monoclonal antibodies can be selected according to known methods or modifications thereof. Usually, it can be performed in an animal cell culture medium supplemented with HAT (hypoxanthine, aminopterin, thymidine). As a selection and breeding medium, any medium can be used as long as it can grow a hybridoma. For example: !! 20%, preferably RPMI 1640 medium containing 10 to 20% fetal bovine serum, GIT medium containing 1 to 10% fetal bovine serum (Wako Pure Chemical Industries, Ltd. ) Or a serum-free medium for hybridoma cultivation (SFM-101, Nissui Pharmaceutical Co., Ltd.). The culturing temperature is usually 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 ) モノクローナル抗体の精製  (b) Purification of monoclonal antibodies
モノクローナル抗体の分離精製は、 公知の方法、 例えば、 免疫グロブリンの分 離精製法 〔例、 塩析法、 アルコール沈殿法、 等電点沈殿法、 電気泳動法、 イオン 交換体 (例、 D E A E ) による吸脱着法、 超遠心法、 ゲルろ過法、 抗原結合固相 あるいはプロテイン Aあるいはプロテイン Gなどの活性吸着剤により抗体のみを 採取し、 結合を解離させて抗体を得る特異的精製法〕 に従って行なうことができ る。 〔ポリクローナル抗体の作製〕 Monoclonal antibodies can be separated and purified by known methods, for example, immunoglobulin separation and purification methods (eg, salting out method, alcohol precipitation method, isoelectric point precipitation method, electrophoresis method, ion exchanger (eg, DEAE)). Adsorption / desorption method, ultracentrifugation method, gel filtration method, antigen-bound solid phase or specific purification method in which the antibody is collected using an active adsorbent such as protein A or protein G and the bond is dissociated to obtain the antibody) Can be done. (Preparation of polyclonal antibody)
本発明のポリクローナル抗体は、 公知あるいはそれに準じる方法に従って製造 することができる。 例えば、 免疫抗原 (ポリペプチド抗原) 自体、 あるいはそれ とキャリアー蛋白質との複合体をつくり、 上記のモノクローナル抗体の製造法と 同様に温血動物に免疫を行い、 該免疫動物から本発明の受容体に対する抗体含有 物を採取して、 抗体の分離精製を行なうことにより製造することができる。 温血動物を免疫するために用いられる免疫抗原とキャリアー蛋白質との複合体 に関し、 キャリアー蛋白質の種類およびキャリアーとハプテンとの混合比は、 キ ャリァ一に架橋させて免疫したハプテンに対して抗体が効率良くできれば、 どの 様なものをどの様な比率で架橋させてもよいが、 例えば、 ゥシ血清アルブミンや ゥシサイログロブリン、 へモシァニン等を重量比でハプテン 1に対し、 約 0 . 1 〜 2 0、 好ましくは約 1〜 5の割合で力プルさせる方法が用レ、られる。  The polyclonal antibody of the present invention can be produced according to a known method or a method analogous thereto. For example, a immunizing antigen (polypeptide antigen) itself or a complex thereof with a carrier protein is formed, and a warm-blooded animal is immunized in the same manner as in the above-described method for producing a monoclonal antibody. The antibody can be produced by collecting an antibody-containing substance against the antibody and separating and purifying the antibody. Regarding a complex of an immunizing antigen and a carrier protein used for immunizing a warm-blooded animal, the type of the carrier protein and the mixing ratio of the carrier and the hapten are determined by the antibody against the hapten immunized by cross-linking the carrier. As long as it can be efficiently performed, any kind may be cross-linked at any ratio.For example, serum albumin, thyroglobulin, hemocyanin, etc., in a weight ratio of about 0.1 to 2 per hapten per hapten. A method of pulling force at 0, 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ゃカルポジイミド、 マレイミ ド活性エステル、 チ オール基、 ジチオビリジル基を含有する活性エステル試薬等が用いられる。 縮合生成物は、 温血動物に対して、 抗体産生が可能な部位にそれ自体あるいは 担体、 希釈剤とともに投与される。 投与に際して抗体産生能を高めるため、 完全 フロイントアジュバントゃ不完全フロイントアジュバントを投与してもよい。 投 与は、 通常約 2〜 6週毎に 1回ずつ、 計約 3〜 1 0回程度行なわれる。  Various condensing agents can be used for force coupling between the hapten and the carrier. For example, daltaraldehyde ゃ carbodiimide, a maleimide active ester, an active ester reagent containing a thiol group or a dithioviridyl group, or the like is used. The condensation product is administered to a warm-blooded animal itself or together with a carrier or diluent at a site where antibody production is possible. Complete Freund's adjuvant / incomplete Freund's adjuvant may be administered in order to enhance the antibody-producing ability upon administration. The administration is usually made once every about 2 to 6 weeks, for a total of about 3 to 10 times.
ポリクローナル抗体は、 上記の方法で免疫された温血動物の血液、 腹水など、 好ましくは血液から採取することができる。  The polyclonal antibody can be collected from the blood, ascites, etc., preferably from the blood, of the warm-blooded animal immunized by the above method.
抗血清中のポリクローナル抗体価の測定は、 上記の抗血清中の抗体価の測定と 同様にして測定できる。 ポリクローナル抗体の分離精製は、 上記のモノクローナ ル抗体の分離精製と同様の免疫グロプリンの分離精製法に従って行なうことがで さる。  The measurement of the polyclonal antibody titer in the antiserum can be performed in the same manner as the measurement of the antibody titer in the antiserum described above. The separation and purification of the polyclonal antibody can be performed according to the same method for separation and purification of immunoglobulin as in the above-described separation and purification of the monoclonal antibody.
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 配列を含有する蛋白質もしくはその部分ぺプチドまたはその塩をコードするポリ ヌクレオチド (例、 D NA) に相補的な、 または実質的に相補的な塩基配列また はその一部を含有するポリヌクレオチド (例、 D NA) としては、 該ポリヌクレ ォチドに相補的な、 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 ポリヌクレオチドの発現を抑制し得る作用を有するものであれば、 いずれのポリ ヌクレオチド (アンチセンスポリヌクレオチド) であってもよレ、。 SEQ ID NO: 1. Complementary or substantially complementary to a polynucleotide (eg, DNA) 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 or a salt thereof As a polynucleotide (eg, DNA) containing a partially complementary base sequence or a part thereof, the polynucleotide Any polynucleotide (antisense polynucleotide) which has a nucleotide sequence complementary to or substantially complementary to a nucleotide or a part thereof and has an action capable of suppressing the expression of the polynucleotide. It may be.
具体的には、 本発明の受容体をコードするポリヌクレオチド (例、 D NA) (以下、 これらの D NAを本発明の D NAと略記する場合がある) に相補的な、 または実質的に相補的な塩基配列またはその一部を有するアンチセンス D N A Specifically, it is complementary to or substantially complementary to a polynucleotide encoding the receptor of the present invention (eg, DNA) (hereinafter, these DNAs may be abbreviated as the DNA of the present invention). Antisense DNA having complementary base sequence or part thereof
(以下、 これらの D NAをアンチセンス D NAと略記する場合がある) が挙げら れ、 本発明の D N Aに相補的な、 または実質的に相補的な塩基配列またはその一 部を有し、 該 D N Aの発現を抑制し得る作用を有するものであれば、 いずれのァ ンチセンス D NAであってもよい。 (Hereinafter, these DNAs may be abbreviated as antisense DNAs), having a nucleotide sequence complementary to or substantially complementary to the DNA of the present invention or a part thereof, Any antisense DNA may be used as long as it has an effect of suppressing the expression of the DNA.
本発明の D N Aに実質的に相補的な塩基配列とは、 例えば、 本発明の D N Aに 相補的な塩基配列 (すなわち、 本癸明の D N A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あげら れる。 特に、 本発明の D N Aの相補鎖の全塩基配列うち、 本発明の受容体の 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 D N Aが好適で ある。 これらのアンチセンス D NA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。  The nucleotide sequence substantially complementary to the DNA of the present invention is, for example, about the entire nucleotide sequence or a partial nucleotide sequence of the nucleotide sequence complementary to the DNA of the present invention (that is, the complementary strand of the DNA of the present invention). Base sequences having a homology of 70% or more, preferably about 80% or more, more preferably about 90% or more, and most preferably about 95% or more. In particular, of the entire base sequence of the complementary strand of the DNA of the present invention, the complementary sequence of the base sequence of the portion encoding the N-terminal part of the receptor of the present invention (for example, the base sequence near the start codon) is approximately 70%. % Or more, preferably about 80% or more, more preferably about 90% or more, and most preferably about 95% or more. These antisense DNAs can be produced using a known DNA synthesizer or the like.
具体的には、 配列番号: 3または配列番号: 4で表される塩基配列を有する 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 部分を有するァンチセンスポリヌクレオチド、 配列番号: 3または配列番号: 4 で表される塩基配列を有する 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など が挙げられる。 好ましくは例えば、 配列番号: 3または配列番号: 4で表される 塩基配列を有する D N Aの塩基配列に相補な塩基配列、 またはその一部分を有す るァンチセンスポリヌクレオチド、 配列番号: 3または配列番号: 4で表される 塩基配列を有する D N Aの塩基配列に相補な塩基配列、 またはその一部分を有す るアンチセンスポリヌクレオチドなどが挙げられる。 Specifically, a nucleotide sequence complementary to or substantially complementary to the nucleotide sequence of DNA having the nucleotide sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4, or an antisense having a portion thereof A polynucleotide, an antisense polynucleotide having a nucleotide sequence complementary to or substantially complementary to the nucleotide sequence of DNA having the nucleotide sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4, or a portion thereof And so on. Preferably, for example, an antisense polynucleotide having a base sequence complementary to the base sequence of DNA having the base sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4, or a part thereof, SEQ ID NO: 3 or SEQ ID NO: : Has a base sequence complementary to the base sequence of DNA having the base sequence represented by 4, or a part thereof Antisense polynucleotides 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。 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 hydrolytic enzymes 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. May be substituted. These antisense polynucleotides can be produced using a known DNA synthesizer or the like.
以下に、 本発明の受容体および本発明のリガンドなどの用途を説明する。  The use of the receptor of the present invention and the ligand of the present invention will be described below.
〔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
本発明のリガンドは、 DAP12リン酸化促進活性、 ERKリン酸化促進活性、 ィンス リンシグナル伝達抑制活性、 TNF ct産生促進活性、 グルコース取り込み阻害活性 などを有する。  The ligand of the present invention has a DAP12 phosphorylation promoting activity, an ERK phosphorylation promoting activity, an insulin signaling suppression activity, a TNF ct production promoting activity, a glucose uptake inhibiting activity, and the like.
本発明の受容体を用い、 または組換え型本発明の受容体の発現系を用いたリガ ンドレセプターアツセィ系を用いることにより、 本発明の受容体と、 本発明のリ ガンドとの結合性を変化させる化合物 (例えば、 ペプチド、 蛋白質、 抗体、 非ぺ 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、 血清など) またはその塩を効率よくスクリーニングすることができる。 該化合物またはその塩には、 (i) 本発明の受容体を介して細胞刺激活性 (例、 ァラキドン酸遊離、 アセチルコリン遊離、 細胞内 C a 2+遊離、 細胞内 c AM P生 成、 細胞内 c AM P産生抑制、 細胞内 c GM P生成、 ィノシトールリン酸産生、 細胞膜電位変動、 細胞内蛋白質のリン酸化、 c _ f o sの活性化、 p Hの低下、 G T P 7 S結合活性、 c AM P依存性プロティンキナーゼの活性化、 c GMP依 存性プロテインキナーゼの活性化、 リン脂質依存性プロテインキナーゼの活性化、 微小管結合蛋白質リン酸化酵素 (MA Pキナーゼ) の活性化などを促進する活性 など) を有する化合物 (ァゴ二スト) 、 (ii) 上記細胞刺激活性を有しない化合 物 (アンタゴニスト) 、 (iii) 本発明の受容体と本発明のリガンドとの結合力 を促進する化合物、 (iv) 本発明の受容体と本発明のリガンドとの結合力を阻害 する化合物などが含まれる。 ' By using the receptor of the present invention or a ligand receptor atsey system using the recombinant expression system of the receptor of the present invention, the binding between the receptor of the present invention and the ligand of the present invention can be improved. (Eg, peptides, proteins, antibodies, non-peptide compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, serum, etc.) or salts thereof are efficiently screened. be able to. The compound or a salt thereof includes (i) cell stimulating activity via the receptor of the present invention (eg, arachidonic acid release, acetylcholine release, intracellular Ca 2+ release, intracellular cAMP generation, intracellular Inhibition of cAMP production, intracellular cGMP production, inositol phosphate production, cell membrane potential fluctuation, intracellular protein phosphorylation, activation of c_fos, decrease in pH, GTP7S binding activity, cAMP dependent Activating protein kinase, c GMP-dependent protein kinase, phospholipid-dependent protein kinase, microtubule-associated protein kinase (MAP kinase), etc. (Ii) a compound having no cell stimulating activity (antagonist), (iii) a compound that promotes the binding force between the receptor of the present invention and the ligand of the present invention, (iv) ) Receptor of the present invention Inhibits the binding force between the ligand of the present invention and And the like. '
具体的には、 (i) 本発明の受容体に、 本発明のリガンドを接触させた場合と (ii) 本発明の受容体に、 本発明のリガンドおよび試験化合物を接触させた場合 との比較を行なう。 比較は、 例えば、 本発明の受容体に対する本発明のリガンド の結合量、 細胞刺激活性などを測定して行う。  Specifically, a comparison is made between (i) the case where the receptor of the present invention is brought into contact with the ligand of the present invention and (ii) the case where the receptor of the present invention is brought into contact with the ligand of the present invention and a test compound. Perform The comparison is performed, for example, by measuring the binding amount of the ligand of the present invention to the receptor of the present invention, cell stimulating activity, and the like.
本発明のスクリ一二ング方法としての具体例としては、 例えば、  Specific examples of the screening method of the present invention include, for example,
(a) 本発明のリガンドを本発明の受容体に接触させた場合と、 本発明のリガン ドおよび試験化合物を本発明の受容体に接触させた場合における、 本発明のリガ ンドの本発明の受容体に対する結合量を測定し、 比較することを特徴とする、 本 発明のリガンドと本発明の受容体との結合性を変化させる化合物またはその塩の スクリーユング方法、  (a) When the ligand of the present invention is brought into contact with the receptor of the present invention, and when the ligand of the present invention and the test compound are brought into contact with the receptor of the present invention, Measuring the amount of binding to the receptor and comparing the results, and a method for screening a compound or a salt thereof that changes the binding property between the ligand of the present invention and the receptor of the present invention,
(b) 本発明のリガンドを、 本発明の受容体を含有する細胞または該細胞の膜画 分に接触させた場合と、 本発明のリガンドぉよぴ試験化合物を本発明の受容体を 含有する細胞または該細胞の膜画分に接触させた場合における、 本発明のリガン ドの該細胞または該膜画分に対する結合量を測定し、 比較することを特徴とする. 本発明のリガンドと本発明の受容体との結合性を変化させる化合物またはその塩 のスクリーニング方法、 および  (b) a case where the ligand of the present invention is brought into contact with a cell containing the receptor of the present invention or a membrane fraction of the cell; and a case where the ligand of the present invention and the test compound contain the receptor of the present invention. The method comprises measuring and comparing the amount of the ligand of the present invention bound to the cell or the membrane fraction when the ligand is brought into contact with the cell or the membrane fraction of the cell. A method for screening a compound or a salt thereof that alters the binding to a receptor, and
(c) 本発明の受容体が、 本宪明の受容体をコードする D N Aを含有する形質転 換体を培養することによつて細胞膜上に発現した本発明の受容体である上記  (c) the receptor of the present invention, which is expressed on a cell membrane by culturing a transformant containing DNA encoding the receptor of the present invention;
(b) 記載のスクリーニング方法、  (b) the screening method described,
(d) 本発明のリガンドが、 標識したリガンドである'上記 (a) 〜 (c) のスクリ 一ユング方法などのレセプター結合ァッセィ系、  (d) the ligand of the present invention is a labeled ligand; a receptor binding assay system such as the above-mentioned (a) to (c) screen-Jung method;
(e) 本発明のリガンドを本発明の受容体に接触させた場合と、 本発明のリガン ドおよび試験化合物を本発明の受容体に接触させた場合における、 本発明の受容 体を介した細胞刺激活性を測定し、 比較することを特徴とする、 本発明のリガン ドと本発明の受容体との結合性を変化させる化合物またはその塩のスクリーニン グ方法、  (e) cells mediated by the receptor of the present invention when the ligand of the present invention is brought into contact with the receptor of the present invention and when the ligand of the present invention and the test compound are brought into contact with the receptor of the present invention A method for screening a compound or a salt thereof that changes the binding between the ligand of the present invention and the receptor of the present invention, which comprises measuring and comparing the stimulating activities;
(f) 本発明のリガンドを本発明の受容体を含有する細胞または該細胞の膜画分 に接触させた場合と、 本発明のリガンドおよび試験化合物を本発明の受容体を含 有する細胞または該細胞の膜画分に接触させた場合における、 本発明の受容体を 介した細胞刺激活性を測定し、 比較することを特徴とする、 本発明のリガンドと 本発明の受容体との結合性を変化させる化合物またはその塩のスクリーニング方 法、 および (f) when the ligand of the present invention is brought into contact with a cell containing the receptor of the present invention or a membrane fraction of the cell, and the ligand of the present invention and the test compound contain the receptor of the present invention. A ligand of the present invention and a receptor of the present invention, wherein the cell stimulating activity mediated by the receptor of the present invention when the cell is brought into contact with a cell or a membrane fraction of the cell is measured and compared. A method of screening for a compound or a salt thereof that alters the binding of
(g) 本発明の受容体が、 本発明の受容体をコードする D NAを含有する形質転 換体を培養することによつて細胞膜上に発現した本発明の受容体である上記 (f) のスクリ一ユング方法などの細胞刺激ァッセィ系などが挙げられる。  (g) The receptor according to (f) above, wherein the receptor of the present invention is a receptor of the present invention expressed on a cell membrane by culturing a transformant containing DNA encoding the receptor of the present invention. Cell stimulation assay systems such as the screening method.
本発明のスクリ一二ング方法の具体的な説明を以下にする。 .  A specific description of the screening method of the present invention will be described below. .
本発明の受容体としては、 ヒ トゃ温血動物の臓器の膜画分などが好適に用いら れる。 しかし、 特にヒト由来の臓器は入手が極めて困難なことから、 スクリー二 ングに用いられるものとしては、 組換え体を用いて大量発現させた本発明の受容 体などが適している。  As the receptor of the present invention, a membrane fraction of an organ of a human or a warm-blooded animal is preferably used. However, since it is particularly difficult to obtain human-derived organs, the receptor of the present invention and the like, which is expressed in large amounts using a recombinant, is suitable for screening.
本発明の受容体を製造するには、 前述の本発明の受容体の製造方法などが用い ら る。  To produce the receptor of the present invention, the above-described method for producing the receptor of the present invention and the like are used.
本発明のスクリーニング方法において、 本発明の受容体を含有する細胞あるい は該細胞膜画分などを用いる場合、 後述の調製法に従えばよい。  In the screening method of the present invention, when cells containing the receptor of the present invention or the cell membrane fraction are used, the preparation method described later may be followed.
本発明の受容体を含有する細胞を用いる場合、 該細胞をダルタルアルデヒ ド、 ホルマリンなどで固定化してもよい。 固定化方法はそれ自体公知の方法に従って 行うことができる。  When a cell containing the receptor of the present invention is used, the cell may be immobilized with datalaldehyde, formalin, or the like. The immobilization method can be performed according to a method known per se.
本発明の受容体を含有する細胞としては、 本発明の受容体を発現した宿主細胞 をいうが、 該宿主細胞としては、 前述の大腸菌、 枯草菌、 酵母、 昆虫細胞、 動物 細胞などが挙げられる。 製造方法は前述と同様である。  The cell containing the receptor of the present invention refers to a host cell expressing the receptor of the present invention. Examples of the host cell include the aforementioned Escherichia coli, Bacillus subtilis, yeast, insect cells, animal cells, and the like. . The manufacturing method is the same as described above.
膜画分としては、 細胞を破枠した後、 それ自体公知の方法で得られる細胞膜が 多く含まれる画分のことをいう。 細胞の破砕方法としては、 Potter— Elvehjem型 ホモジナイザーで細胞を押し潰す方法、 ワーリングプレンダーゃポリ トロン The membrane fraction refers to a fraction containing a large amount of cell membrane obtained by a method known per se after cell framing. Methods for crushing cells include crushing cells with a Potter-Elvehjem homogenizer, Waring Plender ゃ Polytron.
(Kinematica社製) による破砕、 超音波による破碎、 フレンチプレスなどで加圧 しながら細胞を細いノズルから噴出させることによる破碎などがあげられる。 細 胞膜の分画には、 分画遠心分離法や密度勾配遠心分離法などの遠心力による分画 法が主として用いられる。 例えば、 細胞破砕液を低速 (500rpm〜3000rpm) で短 時間 (通常、 約 1分〜 1 0分) 遠心し、 上清をさらに高速 (15000rpn!〜 (Kinematica), crushing by ultrasonic waves, crushing by ejecting cells from a thin nozzle while applying pressure with a French press, etc. For fractionation of cell membranes, fractionation by centrifugal force, such as fractionation centrifugation and density gradient centrifugation, is mainly used. For example, the cell lysate may be reduced at low speed (500 rpm to 3000 rpm). Centrifuge for about 1 minute to 10 minutes (usually about 1 to 10 minutes)
30000rpm) で通常 3 0分〜 2時間遠心し、 得られる沈澱を膜画分とする。 該膜画 分中には、 発現した本発明の受容体と細胞由来のリン脂質や膜蛋白質などの膜成 分が多く含まれる。 (30,000 rpm) for 30 minutes to 2 hours, and the resulting precipitate is used as the membrane fraction. The membrane fraction is rich in the expressed receptor of the present invention and membrane components such as cell-derived phospholipids and membrane proteins.
該本発明の受容体を含有する細胞や膜画分中の本発明の受容体の量は、 1細胞 当たり 1 0 3〜1 08分子であるのが好ましく、 1 0 5〜1 0 7分子であるのが好適 である。 なお、 発現量が多いほど膜画分当たりのリガンド結合活性 (比活性) が 高くなり、 高感度なスクリーニング系の構築が可能になるばかりでなく、 同一口 ットで大量の試料を測定できるようになる。 The amount of the receptor of the present invention in cells or in the membrane fraction containing the receptor of the main invention is preferably from 1 0 3 to 1 0 8 molecules per cell, 1 0 5-1 0 7 molecules It is preferred that The higher the expression level, the higher the ligand binding activity (specific activity) per membrane fraction, which not only enables the construction of a highly sensitive screening system, but also enables the measurement of a large number of samples in the same port. become.
前記のレセプター結合アツセィ系や細胞刺激アツセィ系などのスクリーニング 方法を実施するためには、 例えば、 本発明の受容体画分と、 本発明のリガンド (例、 標識した本発明のリガンド) などが用いられる。 本発明の受容体画分とし ては、 天然型の本発明の受容体画分か、 またはそれと同等の活性を有する組換え 型本発明の受容体画分などが望ましい。 ここで、 同等の活性とは、 同等のリガン ド結合活性などを示す。 標識したリガンドとしては、 例えば、 放射性同位元素 (例、 〔1251〕 、 〔1311〕 、 〔¾〕 、 〔14C〕 、 〔32P〕 、 〔33P〕 、 〔3¾〕 など) 、 蛍 光物質 〔例、 シァニン蛍光色素 (例、 Cy2、 Cy3、 Cy5、 Cy5. 5、 Cy7 (アマシャム バイオサイエンス社製) など) 、 フルォレスカミン、 フノレオレツセンイソチオシ ァネート、 NBDなど〕 、 酵素 (例、 /3—ガラクトシダーゼ、 一ダルコシダーゼ, アル力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、 ルシフ 'エリン、 ルシゲニンなど) , ピオチン、 ランタ二ド元素などで標識されたリガンドなどを用いることができる 具体的には、 本発明のリガンドと本発明の受容体との結合性を変化させる化合 物のスクリーニングを行うには、 本発明の受容体を含有する細胞または細胞の膜 画分を、 スクリーニングに適したバッファーに懸濁することによりレセプター標 品を調製する。 バッファーには、 pH4〜10 (望ましくは pH6〜8) のリン酸パッフ ァー、 トリス一塩酸パッファーなどのリガンドと受容体との結合を阻害しないパ ッファーであればいずれでもよい。 また、 非特異的結合を低減させる目的で、 CHAPS, Tween-80™ (花王一アトラス社) 、 ジギトニン、 デォキシコレートなど の界面活性剤をバッファーに加えることもできる。 さらに、 プロテアーゼによる 本発明の受容体の分解を抑える目的で PMSF、 ロイぺプチン、 E-64 (ペプチド研究 所製) 、 ぺプスタチンなどのプロテアーゼ阻害剤を添加することもできる。 0. 01 〜 10mlの該レセプター溶液に、 一定量 (5000〜500000cpm) の標識した本発明の リガンドを添加し、 同時に 10— 1C)〜10— 7Mの試験化合物を共存させる。 非特異的結合 量 (NSB) を知るために大過剰の未標識の本発明のリガンドを加えた反応チュー プも用意する。 反応は 0°C〜50°C、 望ましくは 4°C〜37°Cで 20分〜 24時間、 望まし くは 30分〜 3時間行う。 反応後、 ガラス繊維濾紙等で濾過し、 適量の同バッファ 一で洗浄した後、 ガラス繊維濾紙に残存する放射活性を液体シンチレーションカ ゥンターまたは γ _カウンターで計測する。 拮抗する物質がない場合のカウント (Β。) から非特異的結合量 (NSB) を引いたカウント (Β。- NSB) を 100%とした時、 特異的結合量 (B- NSB) が例えば 50%以下になる試験化合物を拮抗阻害能力のあ る候補物質として選択することができる。 In order to carry out the above-mentioned screening method for the receptor binding assay system or the cell stimulation assay system, for example, the receptor fraction of the present invention and the ligand of the present invention (eg, a labeled ligand of the present invention) are used. Can be As the receptor fraction of the present invention, a naturally occurring receptor fraction of the present invention or a recombinant receptor fraction of the present invention having an activity equivalent thereto is desirable. Here, “equivalent activity” refers to equivalent ligand binding activity and the like. The labeled ligand, e.g., a radioactive isotope (e.g., [125 1], [131 1], [¾], [14 C], [32 P], [33 P] and [3 ¾]), Fluorescent substances [eg, cyanine fluorescent dyes (eg, Cy2, Cy3, Cy5, Cy5.5, Cy7 (manufactured by Amersham Biosciences), etc.), fluorescamine, phnoleorescene isothiocynate, NBD, etc.], enzymes (eg, , / 3-galactosidase, 1-dalcosidase, al-force phosphatase, peroxidase, malate dehydrogenase, etc., luminous substances (eg, luminol, luminol derivatives, luciferin, lucigenin, etc.), piotin, lanthanide A ligand labeled with an element or the like can be used. Specifically, a compound of a compound that changes the binding between the ligand of the present invention and the receptor of the present invention is used. To perform the Ningu, cells or cell membrane fraction containing the receptor of the present invention, to prepare the receptor preparation by suspending in a buffer appropriate for screening. The buffer may be any buffer such as a phosphate buffer having a pH of 4 to 10 (preferably pH 6 to 8) or a buffer of Tris-monohydrochloride, which does not inhibit the binding between the ligand and the receptor. In addition, for the purpose of reducing non-specific binding, CHAPS, Tween-80 ™ (Kaoichi Atlas), digitonin, dexcholate, etc. Can be added to the buffer. Further, for the purpose of suppressing the degradation of the receptor of the present invention by a protease, a protease inhibitor such as PMSF, leptin, E-64 (manufactured by Peptide Research Laboratories) and pepstatin can be added. To 0.01 to 10 ml of the receptor solution, a fixed amount (5000 to 500,000 cpm) of the labeled ligand of the present invention is added, and at the same time, 10 to 1 M ) to 10 to 7 M of the test compound are allowed to coexist. A reaction tube containing a large excess of the unlabeled ligand of the present invention is also prepared to determine the non-specific binding amount (NSB). The reaction is carried out at 0 ° C to 50 ° C, preferably at 4 ° C to 37 ° C for 20 minutes to 24 hours, preferably 30 minutes to 3 hours. After the reaction, the mixture is filtered through a glass fiber filter or the like, washed with an appropriate amount of the same buffer, and the radioactivity remaining on the glass fiber filter is measured with a liquid scintillation counter or a γ_counter. When the count (Β.-NSB) obtained by subtracting the non-specific binding amount (NSB) from the count when no antagonist is present (Β.) Is 100%, the specific binding amount (B-NSB) is 50%, for example. % Or less of the test compound can be selected as a candidate substance having a competitive inhibition ability.
さらに、 表面プラズモンセンサー技術を利用することによって、 本発明の受容 体に結合する化合物をスクリーニングすることもできる。  Furthermore, compounds that bind to the receptor of the present invention can be screened by utilizing the surface plasmon sensor technology.
具体的には、 ビアコア 3000 (ビアコア社) のセンサーチップ表面に、 本発明の 受容体を固定化後、 チップ表面にリン酸緩衝液 (PBS) などに溶解した試験化合 物を流したときの表面プラズモンの変化を測定することにより、 本発明の受容体 に結合する試験化合物を選択する。 例えば、 表面プラズモンの変化の測定値が 5 レゾナンスュニット以上与える試験化合物を本発明の受容体に結合性を有する物 質として選択する。  Specifically, after the receptor of the present invention is immobilized on the surface of a sensor chip of Biacore 3000 (Biacore), the surface of the chip surface when a test compound dissolved in phosphate buffer solution (PBS) or the like is flowed. A test compound that binds to the receptor of the present invention is selected by measuring the change in the plasmon. For example, a test compound that gives a measured value of change in surface plasmon of 5 or more resonance units is selected as a substance having a binding property to the receptor of the present invention.
さらに、 蛍光物質で標識されたリガンドを用いた場合、 蛍光強度、 蛍光偏光の 増減を測定して、 本発明の受容体に結合する化合物をスクリーニングすることも できる。  Further, when a ligand labeled with a fluorescent substance is used, the increase or decrease in fluorescence intensity and fluorescence polarization can be measured to screen for a compound that binds to the receptor of the present invention.
前記の細胞刺激アツセィ系のスクリ ニング方法を実施するためには、 本発明 の受容体を介する細胞刺激活性 (例えば、 ァラキドン酸遊離、 アセチルコリン遊 離、 細胞内 C a 2+遊離、 細胞内 c AM P生成、 細胞内 c AM P産生抑制、 細胞内 c GM P生成、 イノシトールリン酸産生、 細胞膜電位変動、 細胞内蛋白質のリン 酸化、 c一 f o sの活性化、 p Hの低下、 G T P T S結合活性、 c AM P依存性 プロテインキナーゼの活性化、 c GMP依存性プロテインキナーゼの活性化、 リ ン脂質依存性プロテインキナーゼの活性化、 微小管結合蛋白質リン酸化酵素 (M A Pキナーゼ) の活性化などを促進する活性または抑制する活性など) を、 自体 公知の方法または市販の測定用キットを用いて測定することができる。 具体的に は、 まず、 本発明の受容体を含有する細胞をマルチウヱルプレート等に培養する c スクリーニングを行うにあたっては前もつて新鮮な培地あるいは細胞に毒性を示 さない適当なバッファーに交換し、 試験化合物などを添カ卩して一定時間ィンキュ ペートした後、 細胞を抽出あるいは上清液を回収レて、 生成した産物をそれぞれ の方法に従って定量する。 細胞刺激活性の指標とする物質 (例えば、 ァラキドン 酸など) の生成が、 細胞が含有する分解酵素によって検定困難な場合は、 該分解 酵素に対する阻害剤を添加してアツセィを行なってもよい。 また、 c AM P産生 抑制などの活性については、 フォルスコリンなどで細胞の基礎的産生量を増大さ せておいた細胞に対する産生抑制作用として検出することができる。 In order to carry out the above-described screening method for the cell-stimulating Atsei system, the cell-stimulating activity via the receptor of the present invention (for example, arachidonic acid release, acetylcholine release, intracellular Ca 2+ release, intracellular CAM P production, inhibition of intracellular cAMP production, intracellular cGMP production, inositol phosphate production, fluctuation of cell membrane potential, phosphorylation of intracellular protein, activation of c-fos, decrease of pH, GTPTS binding activity, c AM P dependency Activities that promote or inhibit protein kinase activation, c GMP-dependent protein kinase activation, phospholipid-dependent protein kinase activation, microtubule-associated protein kinase (MAP kinase) activation, etc. ) Can be measured using a method known per se or a commercially available measurement kit. Specifically, first, cells containing the receptor of the present invention are cultured in a multiwell plate or the like.c When performing screening, replace with a fresh medium or an appropriate buffer that is not toxic to cells. After adding the test compound and incubating for a certain period of time, extract the cells or collect the supernatant, and quantitate the product produced according to each method. If the production of a substance (for example, arachidonic acid) as an indicator of cell stimulating activity is difficult to be assayed by a degrading enzyme contained in a cell, an inhibitor for the degrading enzyme may be added to perform the assay. In addition, activities such as inhibition of cAMP production can be detected as an activity of inhibiting production of cells whose basic production has been increased by forskolin or the like.
細胞刺激活性を測定してスクリーニングを行なうには、 適当な本発明の受容体 を発現した細胞が必要である。 本発明の受容体を発現した細胞としては、 前述の 本発明の受容体発現細胞株などが望ましい。  In order to perform screening by measuring the cell stimulating activity, cells expressing the appropriate receptor of the present invention are required. As the cells expressing the receptor of the present invention, the above-mentioned cell lines expressing the receptor of the present invention and the like are desirable.
試験化合物としては、 例えばペプチド、 蛋白質、 抗体、 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、 血清など があげられる。  Test compounds include, for example, peptides, proteins, antibodies, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and serum.
上記細胞刺激アツセィ系のスクリーニング方法について、 さらに具体的に以下 ( 1 ) 〜 (1 2 ) に記載する。 '  The screening method for the cell-stimulating Atssay system is described more specifically in the following (1) to (12). '
( 1 ) 本発明の受容体発現細胞が受容体ァゴニストにより刺激されると、 細胞内 の DAP12 (DNAX adaptor protein 12) のチロシン残基がリン酸化される。  (1) When the receptor-expressing cell of the present invention is stimulated by a receptor agonist, a tyrosine residue of DAP12 (DNAX adapter protein 12) in the cell is phosphorylated.
この反応を利用して、 本発明のリガンドの本発明の受容体発現細胞に対する刺 激活性を測定することにより、 本発明のリガンドと本発明の受容体との結合性を 変化させる化合物をスクリーニングすることができる。  Using this reaction, a compound that alters the binding between the ligand of the present invention and the receptor of the present invention is screened by measuring the stimulating activity of the ligand of the present invention on the cells expressing the receptor of the present invention. be able to.
具体的には、 本発明のリガンドを、 本発明の受容体および DAP12共発現細胞に 接触した場合と、 本発明のリガンドおよび試験化合物を、 本発明の受容体および DAP12共発現細胞に接触した場合における、 DAP12リン酸ィヒの程度を測定し、 比較 することにより、 本発明のリガン'ドと本発明の受容体との結合性を変化させる化 合物をスクリーニングする。 DAP12のリン酸ィヒの程度は、 公知の方法、 例えば抗 リン酸化チロシン抗体を用いたウェスタンプロット法で測定する。 Specifically, the case where the ligand of the present invention is brought into contact with the receptor of the present invention and the DAP12 co-expressing cell, and the case where the ligand of the present invention and the test compound are brought into contact with the receptor of the present invention and the DAP12 co-expressing cell Measure the degree of DAP12 phosphate in Thus, a compound that changes the binding property between the ligand of the present invention and the receptor of the present invention is screened. The degree of DAP12 phosphoric acid is measured by a known method, for example, a Western blot method using an anti-phosphorylated tyrosine antibody.
スクリーニング法の一具体例を以下に述べる。  One specific example of the screening method is described below.
本発明の受容体および V5タグ付き DAP12共発現 (例、 CH0などの動物細胞) を公 知の手法により作製する。 該細胞を、 24穴プレートに 5xl04cell/wellで播種し、 48時間培養する。 この細胞に、 本発明のリガンドおよび試験化合物を添加し、 10 分間培養する。 細胞を、 細胞溶解液 [30 mM Tris-HCl (pH7. 4) , 150 mM NaCl, 10 mM EDTA, 1% NP-40, 50 mM NaF, 1 mM Na - Vanadate] で溶解し、 さらにソニ ケーシヨンにより膜を破碎する。 細胞破碎液を公知の方法に従い抗 V5抗体で免疫 沈降し、 免疫沈降された試料をウェスタンプロット法で解析する。 まず、 抗 V5抗 体 (インビトロジェン) を用いて試料中の DAP12総蛋白質をデンシトメ一ターで 検出する。 抗リン酸化チロシン抗体 (シグマ) を用いてリン酸化された DAP12を 同様の手法で検出する。 両者を比較することにより DAP12蛋白質あたりのチロシ ンリン酸ィヒの程度が検出される。 The receptor of the present invention and VAP-tagged DAP12 co-expression (eg, animal cells such as CH0) are produced by a known method. The cells were seeded at 5xl0 4 cell / well in 24-well plates, and cultured for 48 hours. To the cells, the ligand of the present invention and a test compound are added, and the cells are cultured for 10 minutes. Cells are lysed with a cell lysate [30 mM Tris-HCl (pH 7.4), 150 mM NaCl, 10 mM EDTA, 1% NP-40, 50 mM NaF, 1 mM Na-Vanadate], and then sonicated. Crush the membrane. The cell lysate is immunoprecipitated with an anti-V5 antibody according to a known method, and the immunoprecipitated sample is analyzed by Western blotting. First, DAP12 total protein in a sample is detected by a densitometer using an anti-V5 antibody (Invitrogen). Detect phosphorylated DAP12 using an anti-phosphorylated tyrosine antibody (Sigma) using the same method. By comparing the two, the degree of tyrosin phosphate per DAP12 protein is detected.
本発明のリガンドのみで細胞を刺激した場合の DAP12のリン酸化の程度と、 試 験化合物および^:発明のリガンドを添カ卩した場合の MP12のリン酸化の程度を比 較し、 本発明のリガンドによる DAP12のリン酸化亢進を妨げる試験化合物をアン タゴニストとして選択する。  The degree of phosphorylation of DAP12 when cells were stimulated only with the ligand of the present invention was compared with the degree of phosphorylation of MP12 when the test compound and ^: ligand of the invention were added to the cells. A test compound that prevents the enhanced phosphorylation of DAP12 by the ligand is selected as an antagonist.
( 2 ) 本発明の受容体発現細胞は、 本発明のリガンドの刺激により、 細胞内 cAMP の産生が抑制される。 この反応を利用して、 本発明のリガンドの本発明の受容体 発現細胞に対する刺激活性を測定することにより、 本発明のリガンドと本発明の 受容体との結合性を変化させる化合物をスクリーニングすることができる。  (2) In the cells expressing the receptor of the present invention, the production of intracellular cAMP is suppressed by stimulation with the ligand of the present invention. By utilizing this reaction, the stimulatory activity of the ligand of the present invention on the cell expressing the receptor of the present invention is measured to screen for a compound that alters the binding between the ligand of the present invention and the receptor of the present invention. Can be.
具体的には、 細胞内 cAMP量を増加させる物質の存在下、 本発明のリガンドを本 発明の受容体発現細胞に接触させた場合と、 本発明のリガンドおよび試験化合物 を本発明の受容体発現細胞に接触させた場合における、 該細胞の細胞内 cAMPの産 生抑制活性を測定し、 比較することにより、 本発明のリガンドと本発明の受容体 との結合性を変化させる化合物をスクリーニングする。  Specifically, when the ligand of the present invention is brought into contact with the receptor-expressing cell of the present invention in the presence of a substance that increases the amount of intracellular cAMP, the ligand of the present invention and the test compound express the receptor of the present invention. Compounds that alter the binding between the ligand of the present invention and the receptor of the present invention are screened by measuring and comparing the inhibitory activity of the cells on the production of intracellular cAMP when brought into contact with the cells.
細胞内 cAMP量を増加させる物質としては、 例えば、 フオルスコリン、 カルシト ニンなどが用いられる。 + Substances that increase the amount of intracellular cAMP include, for example, forskolin, calcito Nin and the like are used. +
本発明の受容体発現細胞内の cAMP産生量は、 マウス、 ラット、 ゥサギ、 ャギ、 ゥシなどを免疫して得られた抗 cAMP抗体と 〔1251〕 標識 cAMP (ともに市販品) を 使用することによる RIA系、 または抗 cAMP抗体と標識 cAMPとを組み合わせた EIA系 で測定することができる。 また、 抗 cAMP抗体を、 protein Aまたは抗 cAMP抗体産 生に用いた動物の IgGなどに対する抗体などを使用して固定したシンチラントを 含むビーズと 〔1251〕 標識 cAMPとを使用する SPA (Scintillation Proximity Assay) 法による定量も可能である (アマシャムフアルマシアバイオテク社製の キットを使用する) 。 The amount of cAMP produced in the cells expressing the receptor of the present invention was determined using an anti-cAMP antibody obtained by immunizing mice, rats, rabbits, goats, rabbits, etc., and [ 125 1] -labeled cAMP (both commercially available products). Can be measured by RIA system or EIA system combining anti-cAMP antibody and labeled cAMP. Moreover, the anti-cAMP antibody, SPA to use the beads and [125 1] -labeled cAMP scintillant containing fixed using such antibodies to such animal IgG used for protein A or an anti-cAMP antibody production (Scintillation Proximity Assay) is also possible (using a kit from Amersham-Pharmacia Biotech).
本方法において、 本発明のリガンドによる本発明の受容体発現細胞の cAMP産生 抑制活性を阻害する活性を示す試験化合物を、 拮抗阻害能力のある候補物質とし て選択することができる。  In this method, a test compound showing an activity of inhibiting the cAMP production-suppressing activity of the receptor-expressing cell of the present invention by the ligand of the present invention can be selected as a candidate substance having a competitive inhibition ability.
一方、 試験化合物のみを本発明の受容体発現細胞に接触させて、 cAMP産生抑制 活性を調べることによりァゴニスト活性を示す化合物のスクリ一二ングを行なう ことができる。  On the other hand, a compound showing agonist activity can be screened by bringing only the test compound into contact with the receptor-expressing cell of the present invention and examining the cAMP production inhibitory activity.
スクリーユング法の一具体例を以下に述べる。  One specific example of the SCREENG method is described below.
本発明の受容体発現細胞 (例、 CH0細胞などの動物細胞) を 24穴プレートに 5xl04cell/wellで播種し、 48時間培養する。 細胞を 0· 2raM 3-イソブチル-メチル キサンチン、 0· 05% BSAおよび 20mM HEPESを含むハンクスバッファー (ρΗ7· 4) で洗浄する (以下、 反応用バッファーと略記する) 。 その後、 0. 5mlの反応用バ ッファーを加えて 30分間培養器で保温する。 反応用バッファーを除き、 新たに 0. 25mlの反応用バッファ一を細胞に加えた後、 1 μ Μの本発明のリガンドまたは 1 Μの本発明のリガンドおよび試験化合物を添カ卩した 2 /1 Μ フオルスコリンを含 む 0. 25mlの反応用バッファーを、 細胞に加え、 37°Cで 24分間反応させる。 ΙΟΟ μ Ι の 20%過塩素酸を加えて反応を停止させ、 その後氷上で 1時間置くことにより細 胞内 cAMPを抽出する。 抽出液中の cAMP量を、 cAMP EIAキット (アマシャムフアル マシアバイオテク) を用いて測定する。 フオルスコリンの刺激によって産生され た cAMP量を 100%とし、 1 Mの本発明のリガンドの添加によつて抑制された cAMP 量を 0%として、 本発明のリガンドによる cAMP産生抑制活性に対する試験化合物 の影響を算出する。 本発明のリガシドの活性を阻害して、 cAMP産生活性が例えば 50%以上になる試験化合物を、 拮抗阻害能力のある候補物質として選択すること ができる。 Cells expressing the receptor of the present invention (eg, animal cells such as CH0 cells) are seeded on a 24-well plate at 5 × 10 4 cells / well, and cultured for 48 hours. Wash the cells with Hanks buffer (ρΗ7.4) containing 0.2raM 3-isobutyl-methylxanthine, 0.05% BSA and 20mM HEPES (hereinafter abbreviated as reaction buffer). Then, add 0.5 ml of reaction buffer and incubate for 30 minutes in the incubator. After removing the reaction buffer, 0.25 ml of the reaction buffer was added to the cells, and 1 μL of the ligand of the present invention or 1 μL of the ligand of the present invention and the test compound were added thereto.加 え Add 0.25 ml of reaction buffer containing forskolin to the cells, and incubate at 37 ° C for 24 minutes. Stop the reaction by adding ΙΟΟμΙ of 20% perchloric acid, then place on ice for 1 hour to extract intracellular cAMP. Measure the amount of cAMP in the extract using the cAMP EIA kit (Amersham Pharmacia Biotech). Assuming that the amount of cAMP produced by forskolin stimulation is 100% and the amount of cAMP suppressed by the addition of 1 M of the ligand of the present invention is 0%, a test compound for the cAMP production inhibitory activity of the ligand of the present invention Calculate the effect of A test compound that inhibits the activity of the ligside of the present invention and has a cAMP production activity of, for example, 50% or more can be selected as a candidate substance having a competitive inhibition ability.
また、 本発明のリガンドの刺激により、 細胞内 cAMP量が増加する性質を示す本 発明の受容体発現細胞を使用する場合、 本発明のリガンドを本発明の受容体発現 細胞に接触させた場合と、 本発明のリガンドおよび試験化合物を本発明の受容体 発現細胞に接触させた場合における、 該細胞の細胞内 cAMPの産生促進活性を測定 し、 比較することにより、 本発明のリガンドと本発明の受容体との結合性を変化 させる化合物をスクリーニングすることができる。  Further, when using the receptor-expressing cells of the present invention showing the property of increasing the intracellular cAMP level upon stimulation of the ligand of the present invention, the case where the ligand of the present invention is brought into contact with the receptor-expressing cells of the present invention is compared with When the ligand of the present invention and a test compound are brought into contact with a cell that expresses the receptor of the present invention, the activity of the cell for promoting intracellular cAMP production is measured and compared. Compounds that alter the binding to the receptor can be screened.
本方法において、 本発明のリガンドによる本発明の受容体発現細胞の cAMP産生 促進活性を阻害する活性を示す試験化合物を、 拮抗阻害能力のある候補物質とし て選択することができる。  In this method, a test compound showing an activity of inhibiting the cAMP production promoting activity of the receptor-expressing cell of the present invention by the ligand of the present invention can be selected as a candidate substance having a competitive inhibition ability.
一方、 試験化合物のみを本発明の受容体発現細胞に接触させて cAMP産生促進活 性を調べることによりァゴニスト活性を示す化合物のスクリーユングを行なうこ とができる。  On the other hand, a compound exhibiting agonist activity can be screened by bringing only the test compound into contact with the receptor-expressing cell of the present invention and examining cAMP production promoting activity.
cAMP産生促進活性は、 上記のスクリ一ユング法においてフォルスコリンを添加 せずに本発明の受容体発現細胞 (例、 CH0細胞などの動物細胞) に本発明のリガ ンドまたは本発明のリガンドおよび試験化合物を添加して産生された cAMPを上記 の方法で定量して測定する。  The cAMP production promoting activity can be measured by adding the ligand of the present invention or the ligand of the present invention to cells expressing the receptor of the present invention (eg, animal cells such as CH0 cells) without adding forskolin in the above-described screening method. CAMP produced by adding the compound is quantified and measured by the method described above.
( 3 ) CRE-レポーター遺伝子ベクターを用いて、 本発明のリガンドの本発明の受 容体発現細胞に対する刺激活性を測定することにより、 本発明のリガンドと本発 明の受容体との結合性を変化させる化合物をスクリーニングすることができる。  (3) The binding between the ligand of the present invention and the receptor of the present invention is changed by measuring the stimulating activity of the ligand of the present invention on cells expressing the receptor of the present invention using the CRE-reporter gene vector. Compounds to be screened.
CRE (cAMP response element) を含む DNAを、 ベクターのレポーター遺伝子上 流に挿入し、 CRE-レポーター遺伝子ベクターを得る。 CRE -レポーター遺伝子べク ターを導入した本発明の受容体発現細胞において、 cAMPの上昇を伴う刺激は、 CREを介したレポーター遺伝子発現と、 それに引き続くレポーター遺伝子の遺伝 子産物 (蛋白質) の産生を誘導する。 つまり、 レポーター遺伝子蛋白質の酵素活 性を測定することにより、 CRE -レポーター遺伝子ベクター導入細胞内の cAMP量の 変動を検出することができる。 具体的には、 細胞内 cAMP量を増加させる物質の存在下、 本発明のリガンドを、 CRE -レポーター遺伝子ベクター導入本発明の受容体発現細胞に接触させた場合と. 本発明のリガンドぉよび試験化合物を、 CRE-レポータ一遺伝子べクタ一導入本発 明の受容体発現細胞に接触させた場合における、 レポーター遺伝子蛋白質の酵素 活性を測定し、 比較することにより、 本発明のリガンドと本発明の受容体との結 合性を変化させる化合物をスクリーニングする。 DNA containing a CRE (cAMP response element) is inserted upstream of the reporter gene of the vector to obtain a CRE-reporter gene vector. In the cells expressing the receptor of the present invention into which the CRE-reporter gene vector has been introduced, stimulation accompanied by an increase in cAMP induces CRE-mediated reporter gene expression and subsequent production of a reporter gene gene product (protein). Induce. In other words, by measuring the enzymatic activity of the reporter gene protein, it is possible to detect a change in the amount of cAMP in the cells into which the CRE-reporter gene vector has been introduced. Specifically, the case where the ligand of the present invention is brought into contact with the receptor-expressing cell of the present invention into which a CRE-reporter gene vector has been introduced in the presence of a substance that increases the amount of intracellular cAMP. When the compound is brought into contact with a CRE-reporter-gene vector-transfected receptor-expressing cell of the present invention, the enzymatic activity of the reporter gene protein is measured and compared to obtain the ligand of the present invention and the ligand of the present invention. Screen for compounds that alter the binding to the receptor.
細胞内 cAMP量を増加させる物質としては、 例えば、 フオノレスコリン、 カルシト ニンなどが用いられる。  As a substance that increases the amount of intracellular cAMP, for example, phonorescoline, calcitonin, and the like are used.
ベクターとしては、 例えば、 ピツカジーン べィシックベクター、 ピッ力ジー ン ェンハンサーベクター (東洋インキ製造 (株) ) などが用いられる。 CREを 含む DNAを、 上記ベクターのレポーター遺伝子、 例えばルシフェラーゼ遺伝子上 流のマルチクローニングサイ トに揷入し、 CRE-レポーター遺伝子ベクターとする £ 本方法において、 本発明のリガンドによるレポータ一遺伝子蛋白質の酵素活性 抑制を回復させる試験化合物を、 拮抗阻害能力のある候補物質として選択するこ とができる。 As the vector, for example, Pitka Gene Basic Vector, Pikken Gene Enhancer Vector (Toyo Ink Manufacturing Co., Ltd.) and the like are used. The DNA containing the CRE, the reporter gene of the vector, for example揷入the multiple cloning site of the luciferase gene on stream, CRE-in reporter gene vector to £ method, enzyme reporter monogenic protein by the ligand of the present invention A test compound that restores activity suppression can be selected as a candidate substance capable of competitive inhibition.
一方、 試験化合物のみを本発明の受容体発現細胞に接触させて、 フオルスコリ ン刺激によって上昇した発光量の本発明のリガンドと同様な抑制を測定すること によりァゴニストのスクリーニングを行なうこともできる。  On the other hand, agonists can be screened by bringing only the test compound into contact with cells expressing the receptor of the present invention and measuring the same suppression of the amount of luminescence increased by forskolin stimulation as with the ligand of the present invention.
レポーター遺伝子として、 ルシフェラーゼを利用する例を用いて、 このスクリ 一二ング方法の具体例を以下に述べる。  A specific example of this screening method will be described below using an example using luciferase as a reporter gene.
CRE-レポーター遺伝子 (ルシフェラーゼ) を導入した本発明の受容体発現細胞 を、 24穴プレートに 5xl03cell/wellで播種し、 48時間培養する。 細胞を 0. 2raM 3- イソブチル-メチルキサンチン、 0. 05% BSAおよび 20mM HEPESを含むハンクスノ ッファー (ρΗ7· 4) で洗浄する (以下、 反応用バッファーと略記ずる) 。 その後 0. 5mlの反応用バッファーを加えて 30分間培養器で保温する。 反応用バッファー を除き、 新たに 0. 25mlの反応用バッファーを細胞に加えた後、 Ι μ Μの本発明のリ ガンドまたは 1 μ Μの本発明のリガンドおよぴ試験化合物を添加した 2 Μ フオル スコリンを含む 0. 25mlの反応用バッファーを、 細胞に加え、 37°Cで 24分間反応さ せる。 細胞をピツカジーン用細胞溶解剤 (東洋インキ製造 (株) ) で溶かし、 溶 解液に発光基質 (東洋インキ製造 '(株) ) を添加する。 ルシフェラーゼによる発 光は、 ルミノメーター、 液体シンチレーシヨンカウンターまたはトップカウンタ 一により測定する。 本発明のリガンド単独を添カ卩した場合と、 Ι μ Μの本発明のリ ガンドぉよび試験化合物を添加した場合のルシフヱラーゼによる発光量を測定し て、 比較する。 The cells expressing the receptor of the present invention into which the CRE-reporter gene (luciferase) has been introduced are seeded on a 24-well plate at 5 × 10 3 cells / well, and cultured for 48 hours. Wash the cells with Hank's buffer (ρΗ7.4) containing 0.2raM 3-isobutyl-methylxanthine, 0.05% BSA and 20mM HEPES (hereinafter abbreviated as reaction buffer). Then add 0.5 ml of reaction buffer and keep incubator for 30 minutes. After removing the reaction buffer and adding 0.25 ml of the reaction buffer to the cells, add 2 μl of the ligand of the present invention or 1 μl of the ligand of the present invention and the test compound. Add 0.25 ml of reaction buffer containing forskolin to the cells and incubate at 37 ° C for 24 minutes. Lyse the cells with a cell lysing agent for Pitka Gene (Toyo Ink Mfg. Co., Ltd.) Add a luminescent substrate (Toyo Ink Manufacturing Co., Ltd.) to the solution. Luminescence from luciferase is measured with a luminometer, liquid scintillation counter or top counter. The amount of luminescence by luciferase in the case where the ligand of the present invention alone is added and the case where the ligand and test compound of the present invention are added in an amount of ΙμΜ are measured and compared.
本発明のリガンドは、 フオルスコリン刺激に基づくルシフェラーゼによる発光 量の増加を抑制する。 該抑制を回復させる化合物を拮抗阻害能力のある候補物質 として選択することができる。  The ligand of the present invention suppresses an increase in luminescence by luciferase based on forskolin stimulation. A compound that restores the inhibition can be selected as a candidate substance having competitive inhibitory ability.
レポーター遺伝子として、 例えば、 アルカリフォスファターゼ、 クロラムフエ ニコーノレ ·ァセチノレトランスフェラーゼ (chloramphenicol  Examples of reporter genes include alkaline phosphatase, chloramphenicol transferase (chloramphenicol)
acetyltransferase) 、 j3—ガラクトシダーゼなどの遺伝子を用いてもよい。 こ れらのレポーター遺伝子蛋白質の酵素活性は、 公知の方法に従い、 または市販の 測定キットを用いて測定する。 アルカリフォスファターゼ活性は、 例えば和光純 薬製 Lumi-Phos 530を用いて、 クロラムフエ-コール 'ァセチルトランスフェラ ーゼ活性は、 例えば和光純薬製 FAST CAT chrolamphenicol Acetyltransferase Assay KiTを用いて、 —ガラクトシダーゼ活性は、 例えば和光純薬製 Aurora Gal - XEを用いて測定する。 A gene such as acetyltransferase) or j3-galactosidase may be used. The enzyme activity of these reporter gene proteins is measured according to a known method or using a commercially available measurement kit. Alkaline phosphatase activity can be measured using, for example, Lumi-Phos 530 manufactured by Wako Pure Chemical, and chloramphen-chol 'acetyltransferase activity can be measured using, for example, FAST CAT chrolamphenicol Acetyltransferase Assay KiT manufactured by Wako Pure Chemical. For example, it is measured using Aurora Gal-XE manufactured by Wako Pure Chemical.
( 4 ) 本発明の受容体発現細胞は、 本発明のリガンドの刺激により、 TNF o;を細 胞外に放出する。 この反応を利用して、 本発明のリガンドの本発明の受容体発現 細胞に対する刺激活性を測定することにより、 本発明のリガンドと本発明の受容 体との結合性を変化させる化合物をスクリーニングすることができる。  (4) The receptor-expressing cells of the present invention release TNF o; outside the cells upon stimulation with the ligand of the present invention. By utilizing this reaction to measure the stimulating activity of the ligand of the present invention on the cells expressing the receptor of the present invention, it is possible to screen for a compound that alters the binding between the ligand of the present invention and the receptor of the present invention. Can be.
具体的には、 本発明のリガンドを、 本発明の受容体発現細胞に接触させた場合 と、 本発明のリガンドおよび試験化合物を、 本発明の受容体発現細胞に接触させ た場合における、 TNF aの放出活性を測定し、 比較することにより、 本発明のリ ガンドと本発明の受容体との結合性を変化させる化合物をスクリーニングする。 本方法において、 本発明のリガンドによる TNFひ放出活性を阻害する試験化合 物を、 拮抗阻害能力のある候補物質として選択することができる。  Specifically, TNF a when the ligand of the present invention is brought into contact with the receptor-expressing cell of the present invention and when the ligand and the test compound of the present invention are brought into contact with the receptor-expressing cell of the present invention. The compounds that change the binding between the ligand of the present invention and the receptor of the present invention are screened by measuring and comparing the release activities of the compounds. In this method, a test compound that inhibits the activity of releasing the TNF by the ligand of the present invention can be selected as a candidate substance having a competitive inhibition ability.
また、 試験化合物のみを本発明の受容体発現細胞に接触させ、 本発明の受容体 発現細胞の TNFひ放出活性を公知の方法で調べることによりァゴニスト活性を示 す化合物のスクリーニングを行なうこともできる。 The agonist activity was also determined by contacting the test compound alone with the receptor-expressing cells of the present invention and examining the TNF-releasing activity of the receptor-expressing cells of the present invention by a known method. Screening of compounds.
スクリ一二ング法の一具体例を以下に述べる。  One specific example of the screening method is described below.
本発明の受容体発現細胞を 24穴プレートに 5xl04 Cell/wellで播種し、 24時間培 養後、 終濃度 10 Mの本発明のリガンドまたは終濃度 10 / Mの本発明のリガンドぉ よび試験化合物を各 wellに添加する。 37°Cで 10分間インキュベートした後、 細胞 を細胞溶解液 〔30 mM Tris-HCl (pH7. 4) , 150 mM NaCl, 10 mM EDTA, 1% NP-40, 50 mM NaF, 1 mM Na- Vanadate] で溶解し、 さらにソニケーションにより膜を破 砕する。 細胞破碎液に対し、 公知の EIA法に準じ、 含有する TNF a量を測定する。 反応用バッファー 500 μ 1のみを添加した場合 (本発明のリガンド非添力卩 ·試 験化合物非添加) の TNF aの量を 0%、 10 / Mの本発明のリガンドを含む反応用バ ッファーを添カ卩した場合 (試験化合物非添加) の TNF o;の量を 100%として、 試験 化合物を添カ卩した場合の TNFひの量を算出する。 The receptor-expressing cells of the present invention were seeded at 5xl0 4 C ell / we ll in 24-well plates, 24 hr culture Yogo, Rigando of the present invention the ligand or a final concentration of 10 / M of the present invention at a final concentration of 10 M And test compounds are added to each well. After incubation at 37 ° C for 10 minutes, the cells were lysed with cell lysate (30 mM Tris-HCl (pH 7.4), 150 mM NaCl, 10 mM EDTA, 1% NP-40, 50 mM NaF, 1 mM Na-Vanadate ] And disintegrate the membrane by sonication. The amount of TNFa contained in the cell lysate is measured according to the known EIA method. When only 500 μl of the reaction buffer is added (without the ligand of the present invention and without the test compound), the amount of TNFa is 0%, and the reaction buffer contains 10 / M of the ligand of the present invention. Using the amount of TNF o; when the test compound is added (without adding the test compound) as 100%, calculate the amount of TNF when the test compound is added.
TNF a放出活性が、 例えば 50%以下になる試験化合物を拮抗阻害能力のある候 補物質として選択することができる。  A test compound having a TNFa releasing activity of, for example, 50% or less can be selected as a candidate substance capable of competitive inhibition.
( 5 ) 本発明の受容体発現細胞は、 本発明のリガンドの刺激により、 細胞内の Ca 濃度が上昇する。 この反応を利用して、 本発明のリガンドの本発明の受容体発現 細胞に対する刺激活性を測定することにより、 本発明のリガンドと本発明の受容 体との結合性を変化させる化合物をスクリーニングすることができる。  (5) In the receptor-expressing cell of the present invention, the Ca concentration in the cell is increased by the stimulation of the ligand of the present invention. By utilizing this reaction to measure the stimulating activity of the ligand of the present invention on the cells expressing the receptor of the present invention, it is possible to screen for a compound that alters the binding between the ligand of the present invention and the receptor of the present invention. Can be.
具体的には、 本発明のリガンドを、 本発明の受容体発現細胞に接触させた場合 と、 本発明のリガンドおよび試験化合物を、 本発明の受容体発現細胞に接触させ た場合における、 細胞内カルシウム濃度上昇活性を測定し、 比較することにより、 本発明のリガンドと本発明の受容体との結合性を変化させる化合物をスクリ一- ングする。 測定は公知の方法に従って行う。  Specifically, the intracellular in the case where the ligand of the present invention is brought into contact with the receptor-expressing cell of the present invention and the case where the ligand of the present invention and the test compound are brought into contact with the receptor-expressing cell of the present invention By measuring and comparing the calcium concentration increasing activity, a compound that changes the binding property between the ligand of the present invention and the receptor of the present invention is screened. The measurement is performed according to a known method.
本方法において、 本発明のリガンドによる細胞内カルシウム濃度の上昇を抑制 する試験化合物を、 拮抗阻害能力のある候補物質として選択することができる。 —方、 試験化合物のみの添加による蛍光強度の上昇を測定することによってァ ゴニストのスクリ一ユングを行なうこともできる。  In this method, a test compound that suppresses an increase in intracellular calcium concentration due to the ligand of the present invention can be selected as a candidate substance having a competitive inhibition ability. On the other hand, agonists can be screened by measuring the increase in fluorescence intensity due to the addition of the test compound alone.
スクリ一二ング法の一具体例を以下に述べる。  One specific example of the screening method is described below.
本発明の受容体発現細胞を、 滅菌した顕微鏡用カバーグラス上に播き、 2日後、 培養液を、 4raM Fura-2 AM (同仁化学研究所) を縣濁した HBSSに置換し、 室温で 2 時間 30分おく。 HBSSで洗浄した後、 キュベットにカバーグラスをセットし、 本発 明のリガンドまたは本発明のリガンドおよび試験化合物を添加し、 励起波長 340nmおよび 380nmでの、 505nmの蛍光強度の比の上昇を蛍光測定器で測定し、 比 較する。 The receptor-expressing cell of the present invention was seeded on a sterilized microscope cover glass, and two days later, Replace the culture solution with HBSS in which 4raM Fura-2 AM (Dojindo Laboratories) is suspended, and leave at room temperature for 2 hours and 30 minutes. After washing with HBSS, place the cover glass on the cuvette, add the ligand of the present invention or the ligand of the present invention and the test compound, and measure the increase in the ratio of the fluorescence intensity at 505 nm at the excitation wavelengths of 340 nm and 380 nm by fluorescence measurement. Measure with a tester and compare.
また、 FLIPR (モレキュラーデバイス社製) を使って行ってもよい。 本発明の 受容体発現細胞縣濁液に Fluo- 3 AM (同仁化学研究所製) を添加し、 細胞に取り 込ませた後、 上清を遠心により数度洗浄後、 96穴プレートに細胞を播く。 FLIPR 装置にセットし、 Fura- 2の場合と同様に、 本発明のリガンドまたは本発明のリガ ンドおよび試験化合物を添カ卩し、 蛍光強度の比の上昇を蛍光測定器で測定し、 比 較する。  Alternatively, FLIPR (Molecular Devices) may be used. After adding Fluo-3 AM (manufactured by Dojindo Laboratories) to the receptor-expressing cell suspension of the present invention and allowing the cells to take up, the supernatant is washed several times by centrifugation, and the cells are transferred to a 96-well plate. Sow. Set in a FLIPR device, add the ligand of the present invention or the ligand of the present invention and a test compound in the same manner as in the case of Fura-2, measure the increase in the ratio of fluorescence intensity with a fluorimeter, and compare. I do.
さらに、 本発明の受容体発現細胞に、 細胞内 Caイオンの上昇によって発光する ような蛋白質の遺伝子 (例、 aequorinなど) を共発現させておき、 細胞内 Caィォ ン濃度の上昇によって、 該遺伝子蛋白質 (例、 aequorinなど) が Ca結合型となり 発光することを利用して、 本発明のリガンドと本発明の受容体との結合性を変ィ匕 させる化合物をスクリーニングすることもできる。  Further, a gene (eg, aequorin) of a protein that emits light due to an increase in intracellular Ca ions is co-expressed in the receptor-expressing cell of the present invention, and the intracellular Ca ion concentration increases. Utilizing the fact that a gene protein (eg, aequorin or the like) becomes Ca-binding and emits light, it is also possible to screen for a compound that alters the binding between the ligand of the present invention and the receptor of the present invention.
細胞内 Caイオンの上昇によって発光するような蛋白質の遺伝子を共発現させた 本発明の受容体発現細胞を、 96穴プレートに播き、 上記と同様に、 本発明のリガ ンドまたは本発明のリガンドおよび試験化合物を添加し、 蛍光強度の比の上昇を 蛍光測定器で測定し、 比較する。  A receptor-expressing cell of the present invention, in which a gene for a protein that emits light by increasing intracellular Ca ions, is seeded on a 96-well plate, and the ligand of the present invention or the ligand of the present invention, Add the test compound, measure the increase in the ratio of fluorescence intensity with a fluorimeter, and compare.
本発明のリガンドによる蛍光強度の上昇を、 抑制する試験化合物を拮抗阻害能 力のある候補物質として選択することができる。  A test compound that suppresses an increase in fluorescence intensity due to the ligand of the present invention can be selected as a candidate substance having a competitive inhibitory ability.
( 6 ) 受容体を発現する細胞に、 受容体ァゴニストを添加すると、 細胞内イノシ トール三リン酸濃度は上昇する。 本発明のリガンドの、 本発明の受容体発現細胞 における細胞内ィノシトール三リン酸産生活性を利用することにより、 本発明の リガンドと本発明の受容体との結合性を変化させる化合物をスクリーニングする ことができる。  (6) When a receptor agonist is added to a cell that expresses the receptor, the intracellular inositol triphosphate concentration increases. A compound that changes the binding property between the ligand of the present invention and the receptor of the present invention is screened by utilizing the activity of the ligand of the present invention to produce intracellular inositol triphosphate in the cells expressing the receptor of the present invention. be able to.
具体的には、 標識したイノシトールの存在下、 本発明のリガンドを、 本発明の 受容体発現細胞に接触させた場合と、 本発明のリガンドおよび試験化合物を、 本 発明の受容体発現細胞に接触させ 場合における、 イノシトール三リン酸産生活 性を測定し、 比較することにより、 本発明のリガンドと本発明の受容体との結合 性を変化させる化合物をスクリーニングする。 測定は公知の方法に従って行う。 本方法において、 イノシトール三リン酸産生活性を抑制する試験化合物を、 拮 抗阻害能力のある候補物質として選択することができる。 Specifically, when the ligand of the present invention is brought into contact with the receptor-expressing cell of the present invention in the presence of labeled inositol, By measuring and comparing the inositol triphosphate productivity when the cells are brought into contact with the receptor-expressing cells of the present invention, compounds that alter the binding between the ligand of the present invention and the receptor of the present invention are screened. The measurement is performed according to a known method. In this method, a test compound that suppresses inositol triphosphate-producing activity can be selected as a candidate substance capable of competitive inhibition.
一方、 試験化合物のみを本発明の受容体発現細胞に接触させ、 イノシトール三 リン酸産生上昇を測定することによってァゴニストのスクリーニングを行なうこ ともできる。  On the other hand, agonists can be screened by bringing only the test compound into contact with the receptor-expressing cell of the present invention and measuring the increase in inositol triphosphate production.
スクリーニング法の一具体例を以下に述べる。  One specific example of the screening method is described below.
本発明の受容体発現細胞を 24穴プレートに播き、 1日間培養する。 その後、 myo-[2-3H] inositol (2. 5 μ Ci/well) を添加した培地で 1日間培養し、 細胞を放 射活性を有するィノシトールを無添加の培地でよく洗浄する。 本発明のリガンド または本発明のリガンドおよび試験化合物を添加後、 10%過塩素酸を加え、 反応 を止める。 1. 5M水酸化力リゥムおよび 60mM HEPES溶液で中和し、 0. 5mlの AGlx8 樹脂 (Bio- Rad) を詰めたカラムに通し、 5mM 四ホウ酸ナトリウム (Na2B407) お ょぴ 60mM ギ酸アンモニゥムで洗浄した後、 1M ギ酸アンモニゥムおよび 0. 1M ギ 酸で溶出した放射活性を、 液体シンチレーシヨンカウンターで測定する。 本発明 のリガンドを添カ卩しない場合の放射活性を 0%、 本発明のリガンドを添カ卩した場 合の放射活性を 100%とし、 試験化合物の、 本発明のリガンドと本発明の受容体 の結合に対する影響を算出する。 The cells expressing the receptor of the present invention are seeded on a 24-well plate and cultured for one day. After that, the cells are cultured in a medium supplemented with myo- [2- 3 H] inositol (2.5 μCi / well) for 1 day, and the cells are thoroughly washed with a medium free of radioactive inositol. After adding the ligand of the present invention or the ligand of the present invention and the test compound, 10% perchloric acid is added to stop the reaction. 1. neutralized with 5M hydroxide force Riumu and 60 mM HEPES solution and then passed through a column packed with AGlx8 resin (Bio- Rad) in 0. 5 ml, 5 mM sodium tetraborate (Na 2 B 4 0 7) Contact Yopi After washing with 60 mM ammonium formate, the radioactivity eluted with 1 M ammonium formate and 0.1 M formic acid is measured using a liquid scintillation counter. The radioactivity when the ligand of the present invention is not added is 0%, and the radioactivity when the ligand of the present invention is added is 100%, and the ligand of the present invention and the receptor of the present invention are the test compounds. Calculate the effect on binding.
ィノシトール三リン酸産生活性が、 例えば 50%以下になる試験化合物を拮抗阻 害能力のある候補物質として選択することができる。  A test compound having an inositol triphosphate-producing activity of, for example, 50% or less can be selected as a candidate substance having an antagonistic ability.
( 7 ) TRE-レポーター遺伝子ベクターを用いて、 本発明のリガンドの本発明の受 容体発現細胞に対する刺激活性を測定することにより、 本発明のリガンドと本発 明の受容体との結合性を変化させる化合物をスクリーニングすることができる。  (7) The binding between the ligand of the present invention and the receptor of the present invention is changed by measuring the stimulating activity of the ligand of the present invention on cells expressing the receptor of the present invention using the TRE-reporter gene vector. Compounds to be screened.
TRE (TPA response element) を含む DNA 、 ベクターのレポーター遺伝子上流 に挿入し、 TRE-レポーター遺伝子ベクターを得る。 TRE-レポーター遺伝子べクタ 一を導入した本発明の受容体発現細胞において、 細胞内カルシウム濃度の上昇を 伴う刺激は、 TREを介したレポーター遺伝子発現と、 それに引き続くレポーター 遺伝子の遺伝子産物 (蛋白質) の產生を誘導する。 つまり、 レポーター遺伝子蛋 白質の酵素活性を測定することにより、 TRE-レポーター遺伝子ベクター導入細胞 内のカルシウム量の変動を検出することができる。 DNA containing a TRE (TPA response element) is inserted into the vector upstream of the reporter gene to obtain a TRE-reporter gene vector. In the receptor-expressing cells of the present invention into which the TRE-reporter gene vector has been introduced, stimulation accompanied by an increase in intracellular calcium concentration is caused by TRE-mediated reporter gene expression and subsequent reporter gene expression. Induces the production of gene products (proteins) of genes. That is, by measuring the enzymatic activity of the reporter gene protein, it is possible to detect a change in the amount of calcium in the cells into which the TRE-reporter gene vector has been introduced.
具体的には、 本発明のリガンドを、 TRE -レポータ一遺伝子べクタ一導入本発明 の受容体発現細胞に接触させた場合と、 本発明のリガンドおよび試験化合物を、 TRE-レポータ一遺伝子べクタ一導入本発明の受容体発現細胞に接触させた場合に おける、 レポーター遺伝子蛋白質の酵素活性を測定し、 比較することにより、 本 発明のリガンドと本発明の受容体との結合性を変化させる化合物をスクリーニン グする。  Specifically, when the ligand of the present invention is brought into contact with a TRE-reporter-gene vector-introduced receptor-expressing cell, the ligand of the present invention and a test compound are expressed in a TRE-reporter-gene vector. A compound that changes the binding between the ligand of the present invention and the receptor of the present invention by measuring and comparing the enzymatic activity of the reporter gene protein when the cell is brought into contact with the cells expressing the receptor of the present invention. Screen.
ベクターとしては、 例えば、 ピツカジーン べィシックベクター、 ピツカジー ン ェンハンサーベクタ一 (東洋インキ製造 (株) ) などが用いられる。 TREを 含む DNAを、 上記ベクターのレポーター遺伝子、 例えばルシフェラーゼ遺伝子上 流のマルチクローニングサイトに揷入し、 TRE-レポーター遺伝子ベクターとする c 本方法において、 本発明のリガンドによるレポーター遺伝子蛋白質の酵素活性 を抑制する試験化合物を、 拮抗阻害能力のある候補物質として選択することがで さる。 As the vector, for example, Pitka Gene Basic Vector, Pitka Gene Enhancer Vector-1 (Toyo Ink Mfg. Co., Ltd.) and the like are used. The DNA containing the TRE, the reporter gene of the vector, for example揷入the multiple cloning site of the luciferase gene on stream, TRE-in c the method of the reporter gene vector, the enzymatic activity of the reporter gene protein by the ligand of the present invention The test compound to be suppressed can be selected as a candidate substance having competitive inhibitory ability.
一方、 試験化合物のみを TRE-レポーター遺伝子ベクター導入本発明の受容体発 現細胞に接触させ、 本発明のリガンドと同様な発光量の増加を測定することによ りァゴニストのスクリ一エングを行なうこともできる。  On the other hand, agonist screening is performed by bringing only the test compound into contact with the TRE-reporter gene vector-transfected receptor-expressing cell of the present invention and measuring the increase in luminescence in the same manner as the ligand of the present invention. You can also.
レポーター遺伝子として、 ルシフェラーゼを利用する例を用いて、 このスクリ 一ユング方法の具体例を以下に述べる。  A specific example of this screening method will be described below using an example using luciferase as a reporter gene.
TRE-レポーター遺伝子 (ルシフェラーゼ) を導入した本発明の受容体発現細胞 を、 24穴プレートに 5xl03cell/wellで捧種し、 48時間培養する。 細胞を 0. 05% BSAおよび 20mM HEPESを含むハンクスバッファー (pH7. 4) で洗浄した後、 ΙΟηΜの 本発明のリガンドまたは ΙΟηΜの本発明のリガンドおよび試験化合物を添カ卩し、 ' 37°Cで 60分間反応させる。 細胞をピツカジーン用細胞溶解剤 (東洋インキ製造 The TRE-reporter gene (luciferase) -introduced receptor-expressing cells of the present invention are seeded on a 24-well plate at 5 × 10 3 cells / well and cultured for 48 hours. After washing the cells with Hanks buffer (pH 7.4) containing 0.05% BSA and 20 mM HEPES, add ΙΟηΙΟ ligand of the present invention or ΙΟηΜ ligand of the present invention and a test compound, and add at 37 ° C. And react for 60 minutes. Cell lysing agent for Pitka Gene (Toyo Ink Manufacturing)
(株)) で溶かし、 溶解液に発光基質 (東洋インキ製造 (株)) を添加する。 ルシフ エラーゼによる発光は、 ノレミノメーター、 液体シンチレーシヨンカウンターまた はトップカゥンターにより測定する。本発明のリガンドを添カ卩した場合と、 ΙΟηΜ の本発明のリガンドおよび試験化合物を添カ卩した場合のルシフエラーゼによる発 光量を測定して、 比較する。 And add a luminescent substrate (Toyo Ink Mfg. Co., Ltd.) to the solution. Luminescence from Luciferase is measured with a noreminometer, liquid scintillation counter or top counter. When the ligand of the present invention is added, The amount of light emitted by luciferase when the ligand of the present invention and the test compound are added is measured and compared.
本発明のリガンドによる細胞内カルシウムの上昇によって、 ルシフェラーゼに よる発光量が増加する。 この増加を抑制する化合物を拮抗阻害能力のある候補物 質として選択することができる。  The increase in intracellular calcium by the ligand of the present invention increases the amount of luminescence by luciferase. A compound that suppresses this increase can be selected as a candidate substance capable of competitive inhibition.
レポーター遺伝子として、 例えば、 アルカリフォスファターゼ、 クロラムフエ ニコーノレ 'ァセチノレトランスフェラーセ (chloramphenicol  Reporter genes include, for example, alkaline phosphatase, chloramphenicol transferase, and chloramphenicol.
acetyltransferase) 、 —ガラクトシダーゼなどの遺伝子を用いてもよい。 こ れらのレポーター遺伝子蛋白質の酵素活性は、 公知の方法に従い、 または市販の 測定キットを用いて測定する。 アルカリフォスファターゼ活性は、 例えば和光純 薬製 Lumi-Phos 530を用いて、 クロラムフエニコーノレ 'ァセチルトランスフェラ ーゼ活性は、 例えば和光純薬製 FAST CAT chrolamphenicol Acetyltransferase Assay KiTを用いて、 —ガラクトシダーゼ活性は、 例えば和光純薬製 Aurora Gal- XEを用いて測定する。 acetyltransferase), -A gene such as galactosidase may be used. The enzyme activity of these reporter gene proteins is measured according to a known method or using a commercially available measurement kit. Alkaline phosphatase activity was measured using, for example, Lumi-Phos 530 manufactured by Wako Pure Chemical, and chloramphenicolone acetyltransferase activity was measured using, for example, FAST CAT chrolamphenicol Acetyltransferase Assay KiT manufactured by Wako Pure Chemical. The activity is measured using, for example, Aurora Gal-XE manufactured by Wako Pure Chemical Industries.
( 8 ) 本発明の受容体発現細胞は、 本発明のリガンドの刺激により、 MAPキナー ゼが活性ィヒされ、 増殖する。 この反応を利用して、 本発明のリガンドの本発明の 受容体発現細胞に対する刺激活性を測定することにより、 本発明のリガンドと本 発明の受容体との結合性を変ィ匕させる化合物をスクリーニングすることができる t 具体的には、 本発明のリガンドを、 本発明の受容体発現細胞に接触させた場合 と、 本発明のリガンドおよび試験化合物を、 本発明の受容体発現細胞に接触させ た場合における、 細胞増殖を測定し、 比較することにより、 本発明のリガンドと 本発明の受容体との結合性を変化させる化合物をスクリーニングする。 (8) The MAP kinase is activated by the stimulation of the ligand of the present invention, and the cells expressing the receptor of the present invention proliferate. Utilizing this reaction, the stimulatory activity of the ligand of the present invention on the cells expressing the receptor of the present invention is measured to screen for a compound that changes the binding between the ligand of the present invention and the receptor of the present invention. the t specifically capable of, the ligand of the present invention, and when contacted with receptor-expressing cells of the present invention, a ligand and a test compound of the present invention, brought into contact with receptor-expressing cells of the present invention By measuring and comparing the cell proliferation in each case, a compound that changes the binding between the ligand of the present invention and the receptor of the present invention is screened.
本発明の受容体発現細胞の増殖は、 例えば、 MAPキナーゼ活性、 チミジン取り 込み活性、 細胞数などを測定すればよい。  The growth of the receptor-expressing cells of the present invention may be measured, for example, by measuring MAP kinase activity, thymidine uptake activity, cell number, and the like.
具体例としては、 MAPキナーゼ活性については、 本発明のリガンドまたは本発 明のリガンドおよび試験化合物を、 本発明の受容体発現細胞に添加した後、 細胞 溶解液から抗 MA Pキナーゼ抗体を用いた免疫沈降により MAPキナーゼ分画を得 た後、 公知の方法、 例えば和光純薬製 MAP Kinase Assay Kitおよび γ - [32P] - ATP を使用して MAPキナーゼ活性を測定し、 比較する。 チミジン取り込み活性についてほ、 本発明の受容体発現細胞を 24穴プレートに 播種し、 培養し、 本発明のリガンドまたは本発明のリガンドおよび試験化合物を 添加した後、 放射活性により標識したチミジン (例、 [methyl- ¾]-チミジンな ど) を加え、 その後、 細胞を溶解し、 細胞内に取り込まれたチミジンの放射活性 を、 液体シンチレーシヨンカウンターで計数することにより、 チミジン取り込み 活性を測定し、 比較する。 As a specific example, for the MAP kinase activity, after adding the ligand of the present invention or the ligand of the present invention and a test compound to the receptor-expressing cells of the present invention, an anti-MAP kinase antibody was used from a cell lysate. after obtaining the MAP kinase fractions by immunoprecipitation, a known method, for example, manufactured by Wako pure Chemical Industries, Ltd. MAP of kinase Assay Kit and γ - [32 P] - using ATP to measure MAP kinase activity, and compared. Regarding the thymidine incorporation activity, the cells expressing the receptor of the present invention were seeded on a 24-well plate, cultured, and added with the ligand of the present invention or the ligand and the test compound of the present invention, and then thymidine labeled with radioactivity (eg, [methyl-II] -thymidine), then lyse the cells and measure the thymidine uptake activity by counting the radioactivity of the thymidine incorporated into the cells with a liquid scintillation counter. I do.
細胞数の測定については、 本発明の受容体発現細胞を 24穴プレートに播種し、 培養し、 本発明のリガンドまたは本発明のリガンドおよび試験化合物を添カ卩した 後、 MTT (3- (4, 5-dimethyト 2 - thiazolyl) - 2, 5-diphenyl - 2H - tetrazolium bromide) を添加する。 細胞内に取り込まれて MTTが変化した MTTホルマザンを、 塩酸にて酸性としたィソプロパノール水溶液で細胞を溶解した後、 570nmの吸収 によって測定し、 比較する。  For the measurement of the cell number, the cells expressing the receptor of the present invention were seeded on a 24-well plate, cultured, supplemented with the ligand of the present invention or the ligand and the test compound of the present invention, and then treated with MTT (3- (4 , 5-dimethyto 2-thiazolyl)-2, 5-diphenyl-2H-tetrazolium bromide). MTT formazan in which MTT has been changed by being taken into cells is dissolved in an aqueous solution of isopropanol acidified with hydrochloric acid, and then measured by absorption at 570 nm for comparison.
本方法において、 本発明の受容体発現細胞の増殖を抑制する試験化合物を、 拮 抗阻害能力のある候補物質として選択することができる。  In this method, a test compound that suppresses the growth of the receptor-expressing cell of the present invention can be selected as a candidate substance having a competitive inhibition ability.
—方、 試験化合物のみを本発明の受容体発現細胞に接触させ、 本発明のリガン ドと同様な細胞増殖活性を測定することによりァゴニストのスクリーニングを行 なうこともできる。  On the other hand, agonists can be screened by bringing only the test compound into contact with the receptor-expressing cell of the present invention and measuring the same cell proliferation activity as the ligand of the present invention.
チミジン取り込み活性を利用するスクリーニング法の一具体例を以下に述べる ( 本発明の受容体発現細胞を 24穴プレートに 5000個/ゥエル播き、 1日間培養する ( 次に血清を含まない培地で 2日間培養し、 細胞を飢餓状態にする。 本発明のリガ ンドまたは本発明のリガンドおよび試験化合物を、 細胞に添加して 24時間培養し た後、 [methyl- ¾]-チミジンをゥヱル当たり 0. 015MBq添カ卩し、 6時間培養する。 細胞を PBSで洗った後、 メタノールを添加して 10分間放置する。 次に 5%トリクロ 口酢酸を添カ卩して 15分間放置後、 固定された細胞を蒸留水で 4回洗う。 0. 3N水酸 化ナトリゥム溶液で細胞を溶解し、 溶解液中の放射活性を液体シンチレーシヨン カウンターで測定する。 One specific example of the screening method utilizing the thymidine incorporation activity is described below ( the receptor-expressing cells of the present invention are seeded at 5,000 cells / well in a 24-well plate, and cultured for 1 day ( then in a serum-free medium for 2 days). After culturing to starve the cells, add the ligand of the present invention or the ligand of the present invention and a test compound to the cells and culture for 24 hours, and then add [methyl-¾] -thymidine at 0.015 MBq / ml. After culturing for 6 hours, wash the cells with PBS, add methanol and leave for 10 minutes, then add 5% trichloroacetic acid and leave for 15 minutes to fix the cells. Wash the cells four times with distilled water 0.3 Lyse the cells with 3N sodium hydroxide solution, and measure the radioactivity in the lysate with a liquid scintillation counter.
本発明のリガンドを添加した場合の放射活性の増加を抑制する試験化合物を、 拮抗阻害能力のある候補物質として選択することができる。  A test compound that suppresses an increase in radioactivity when the ligand of the present invention is added can be selected as a candidate substance capable of competitive inhibition.
( 9 ) 本発明の受容体発現細胞が受容体ァゴニストによって刺激されると、 細胞 内の ERK (extracellular signal-regulated kinase) 1/2のチロシン残基がリン 酸化される。 (9) When the receptor-expressing cell of the present invention is stimulated by a receptor agonist, Tyrosine residues of ERK (extracellular signal-regulated kinase) 1/2 are phosphorylated.
この反応を利用して、 本発明のリガンドの本発明の受容体発現細胞に対する刺 激活性を測定することにより、 本発明のリガンドと本発明の受容体との結合性を 変化させる化合物をスクリーニングすることができる。  Using this reaction, a compound that alters the binding between the ligand of the present invention and the receptor of the present invention is screened by measuring the stimulating activity of the ligand of the present invention on the cells expressing the receptor of the present invention. be able to.
具体的には、 本発明のリガンドを本発明の受容体発現細胞に接触した場合と、 本発明のリガンドおよび試験化合物を本発明の受容体発現細胞に接触した場合に おける、 ERK1/2リン酸化の程度を測定し、 比較することにより、 本発明のリガン ドと本発明の受容体との結合性を変化させる化合物をスクリーニングする。  Specifically, ERK1 / 2 phosphorylation in the case of contacting the ligand of the present invention with the receptor-expressing cell of the present invention and in the case of contacting the ligand of the present invention and a test compound with the receptor-expressing cell of the present invention. By measuring and comparing the levels of the compounds, a compound that changes the binding between the ligand of the present invention and the receptor of the present invention is screened.
ERK1/2のリン酸化の程度は、 公知の方法、 例えば抗リン^化 ERK抗体を用いた ウェスタンブロット法で測定する。  The degree of phosphorylation of ERK1 / 2 is measured by a known method, for example, Western blotting using an anti-phosphorylated ERK antibody.
スクリーニング法の一具体例を以下に述べる。  One specific example of the screening method is described below.
本発明の受容体発現細胞 (例、 CH0などの動物細胞) を公知の手法で作製する ( 該細胞を、 24穴プレートに 5xl04cell/wellで播種し、 48時間培養する。 この細胞 に本発明のリガンドおよび試験化合物を添加し、 10分間培養する。 細胞を細胞溶 解液 〔30 mM Tris-HCl (pH7. 4) , 150 mM NaCl, 10 mM EDTA, 1% NP-40, 50 mM NaF, 1 mM Na-Vanadate] で溶解し、 さらにソ-ケーシヨンにより膜を破碎する t 細胞破碎液を公知の方法に従レ、抗 V5抗体で免疫沈降し、 免疫沈降された試料をゥ ヱスタンプロット法で解析する。 抗 ERK1/2抗体 (シグマ) を用いて試料中の ERK1/2総蛋白質をデンシトメーターを用いた公知の手法で検出する。 一方抗リン 酸化 ERK抗体 (シグマ) を用いてリン酸化された ERK1/2を同様の手法で検出する t 両者を比較することにより、 ERK1/2蛋白質あたりのチロシンリン酸化の程度が検 出される。 本発明のリガンドのみで細胞を刺激した場合の ERK1/2のリン酸化の程 度と、 試験化合物おょぴ本発明のリガンドを添加した場合の ERK1/2のリン酸化の 程度を比較し、 本発明のリガンドによる ERK1/2のリン酸化亢進を妨げる試験化合 物をアンタゴニストとして選択する。 Cells expressing the receptor of the present invention (eg, animal cells such as CH0) are prepared by a known method (the cells are seeded on a 24-well plate at 5 × 10 4 cells / well, and cultured for 48 hours. Add the ligand of the invention and the test compound, and incubate for 10 minutes Cell lysate [30 mM Tris-HCl (pH 7.4), 150 mM NaCl, 10 mM EDTA, 1% NP-40, 50 mM NaF , 1 was dissolved in mM Na-vanadate], further source - Keshiyon by従Re the t cells broken碎液to Yabu碎a film by a known method, were immunoprecipitated with anti-V5 antibody, immunoprecipitated samples © We stamp lot Detect the total ERK1 / 2 protein in the sample by a known method using a densitometer using an anti-ERK1 / 2 antibody (Sigma) while using an anti-phosphorylated ERK antibody (Sigma). by comparing the t both to detect the ERK1 / 2 phosphorylated in the same manner, Ciro per ERK1 / 2 protein The degree of phosphorylation of ERK1 / 2 when cells are stimulated only with the ligand of the present invention and the degree of ERK1 / 2 when cells containing the test compound and the ligand of the present invention are added are detected. The degree of phosphorylation is compared, and a test compound that prevents enhanced phosphorylation of ERK1 / 2 by the ligand of the present invention is selected as an antagonist.
( 1 0 ) 本発明の受容体発現細胞が本発明のリガンドに反応し、 細胞外の pHが変 化する。 この反応を利用して、 本発明のリガンドの本発明の受容体発現細胞に対 する刺激活性を測定することにより、 本発明のリガンドと本発明の受容体との結 合性を変化させる化合物をスクリ一二ングすることができる。 (10) The cells expressing the receptor of the present invention react with the ligand of the present invention to change extracellular pH. Utilizing this reaction, the stimulatory activity of the ligand of the present invention on the cells expressing the receptor of the present invention is measured, whereby the binding between the ligand of the present invention and the receptor of the present invention is measured. Compounds that alter compatibility can be screened.
具体的には、 本努明のリガンドを、 本発明の受容体発現細胞に接触させた場合 と、 本発明のリガンドおよび試験化合物を、 本発明の受容体発現細胞に接触させ た場合における、 細胞外の pH変化を測定し、 比較することにより、 本発明のリガ ンドと本発明の受容体との結合性を変化させる化合物をスクリーニングする。 細胞外 pH変化は、 例えば、 Cytosensor装置 (モレキュラーデバイス社) を使用 して測定する。  Specifically, the cells obtained when the ligand of the present invention was brought into contact with the receptor-expressing cell of the present invention, and the cells obtained when the ligand and the test compound were brought into contact with the receptor-expressing cell of the present invention. A compound that alters the binding between the ligand of the present invention and the receptor of the present invention is screened by measuring and comparing the change in external pH. The extracellular pH change is measured, for example, using a Cytosensor device (Molecular Devices).
本方法において、 本発明のリガンドによる細胞外 pH変化を抑制する試験化合物 を、 拮抗阻害能力のある候補物質として選択することができる。  In this method, a test compound that suppresses the extracellular pH change caused by the ligand of the present invention can be selected as a candidate substance capable of competitive inhibition.
一方、 試験化合物のみを本発明の受容体発現細胞に接触させ、 本発明のリガン ドと同様な細胞外 pH変化を測定することによりァゴニストのスクリーニングを行 なうこともできる。  On the other hand, agonists can be screened by bringing only the test compound into contact with the receptor-expressing cell of the present invention and measuring the extracellular pH change as in the ligand of the present invention.
スクリーニング法の一具体例を以下に述べる。  One specific example of the screening method is described below.
本発明の受容体発現細胞を Cytosensor装置用のカプセル内で終夜培養し、 装置 のチャンバ一にセットして細胞外 pHが安定するまで約 2時間、 0. 1% BSAを含む RPMI1640培地 (モレキュラーデバイス社製) を灌流させる。 pHが安定した後、 本 発明のリガンドまたは本発明のリガンドおよび試験化合物を含む培地を細胞上に 灌流させる。 灌流によって生じた培地の pH変化を測定し、 比較する。  The receptor-expressing cell of the present invention is cultured overnight in a capsule for a Cytosensor device, set in a chamber of the device, and RPMI1640 medium containing 0.1% BSA (molecular device) for about 2 hours until the extracellular pH is stabilized. Perfusion). After the pH has stabilized, the cells of the present invention or a medium containing a ligand of the present invention and a test compound are perfused onto the cells. The pH change of the medium caused by perfusion is measured and compared.
本発明のリガンドによる細胞外 pH変化を抑制する化合物を拮抗阻害能力のある 候補物質として選択することができる。  A compound that suppresses the extracellular pH change caused by the ligand of the present invention can be selected as a candidate substance having a competitive inhibition ability.
( 1 1 ) 本発明の受容体 (例、 TREM- 2) の細胞外ドメインおよび CDSzetaの融合 蛋白質発現べクタ一と NFAT—レポータ一遺伝子とを組み合わせて、 本発明の受容 体 (例、 TREM-2) シグナルを CD3シグナルと置き換えて検出することができる。 この反応を利用して、 本発明のリガンドの本発明の受容体発現細胞に対する刺激 活性を測定することにより、 本発明のリガンドと本発明の受容体との結合性を変 ィ匕させる化合物をスクリーニングすることができる。  (11) The receptor of the present invention (eg, TREM-2) is prepared by combining the extracellular domain of the receptor of the present invention (eg, TREM-2) and the fusion protein expression protein of CDSzeta with the NFAT-reporter gene. 2) The signal can be detected by replacing the signal with the CD3 signal. Utilizing this reaction, the stimulatory activity of the ligand of the present invention on the cells expressing the receptor of the present invention is measured to screen for a compound that alters the binding between the ligand of the present invention and the receptor of the present invention. can do.
本発明の受容体 (例、 TREM- 2) の細胞外ドメインおよび CD3zetaの融合蛋白質 発現ベクターと NFAT—レポーター遺伝子 (例、 ルシフェラーゼなど) とを導入し た発現細胞において、 本発明の受容体 (例、 TREM- 2) の活性化は、 同時に CD3zetaの活性化となり、 NFATを介したレポーター遺伝子発現と、 それに引き続 くレポーター遺伝子の遺伝子産物 (蛋白質) の産生を誘導する。 つまり、 レポ一 ター遺伝子蛋白質の酵素活性を測定することにより、 NFAT—レポーター遺伝子べ クタ一導入細胞の本発明の受容体 (例、 TREM-2) シグナル強度を検出することが できる。 In an expression cell into which an extracellular domain of a receptor of the present invention (eg, TREM-2) and an expression vector of a fusion protein of CD3zeta and an NFAT-reporter gene (eg, luciferase) are introduced, a receptor of the present invention (eg, luciferase) is used. , TREM-2) Activation of CD3zeta induces NFAT-mediated reporter gene expression and subsequent production of reporter gene gene products (proteins). That is, by measuring the enzymatic activity of the reporter gene protein, the signal intensity of the receptor of the present invention (eg, TREM-2) of the NFAT-reporter gene vector-introduced cells can be detected.
具体的には、 本発明の受容体 (例、 TREM-2) の細胞外ドメインおよび CD3zeta の融合蛋白質発現べクタ一と NFAT—レポ一タ一遺伝子を導入した発現細胞に本発 明のリガンドを接触させた場合と、 本発明の受容体 (例、 TREM - 2) の細胞外ドメ ィンおよび CD3zetaの融合蛋白質発現ベクターと NFAT—レポータ一遺伝子を導入 した発現細胞に本発明のリガンドおよび試験化合物を接触させた場合における、 レポーター遺伝子の蛋白質活性をそれぞれ測定し、 比較することにより、 本発明 のリガンドと本発明の受容体 (例、 TREM-2) との結合性を変化させる化合物をス クリーニングする。  Specifically, the ligand of the present invention is introduced into the expression cell into which the extracellular domain of the receptor of the present invention (eg, TREM-2) and the CD3zeta fusion protein expression vector and the NFAT-reporter gene have been introduced. The ligand of the present invention and the test compound are introduced into the cells in which the extracellular domain of the receptor of the present invention (eg, TREM-2) and the fusion protein expression vector of CD3zeta and the NFAT-reporter gene have been introduced. By measuring and comparing the protein activity of the reporter gene in the case of contact with the receptor, a compound that changes the binding between the ligand of the present invention and the receptor of the present invention (eg, TREM-2) is screened. I do.
レポーター遺伝子として、 ノレシフェラーゼを利用する例を用いて、 このスクリ 一ユング方法の具体例を以下に述べる。  A specific example of this screening method will be described below using an example in which noreciferase is used as a reporter gene.
本発明の受容体 (例、 TREM- 2) の細胞外ドメインおよび CD3zetaの融合蛋白質 発現ベクターと NFAT—レポーター遺伝子 (ルシフェラーゼ) とを導入した発現細 胞を、 24穴プレートに 5xl04cell/wellで播種し、 48時間培養する。 細胞を 0· 2ιηΜ 3 -イソブチル -メチルキサンチン、 0. 05% BSAおよび 20mM HEPESを含むハンクス バッファー (ΡΗ7· 4) で洗浄する (以下、 反応用バッファーと略記する) 。 その 後 0. 5mlの反応用バッファーを加えて 30分間培養器で保温する。 反応用バッファ 一を除き、 新たに 0. 25mlの反応用バッファーを細胞に加えた後、 1 Μの本発明の リガンドまたは 1 μ Μの本発明のリガン.ドぉよび試験化合物を添加した 0. 25mlの反 応用バッファーを、 細胞に加え、 37°Cで 10分間反応させる。 細胞をピツカジーン 用細胞溶解剤 (東洋インキ製造 (株) ) で溶かし、 溶解液に発光基質 (東洋イン キ製造 (株) ) を添加する。 ルシフェラーゼによる発光は、 ルミノメーター、 液 体シンチレーションカウンターまたはトップカウンタ一により測定する。 本発明 のリガンドのみを添加した場合と、 1 μ Mの本発明のリガンドおよび試験化合物を 添加した場合におけるルシフェラーゼによる発光量を測定して、 比較する。 ( 1 2 ) 本発明の受容体遺伝子 RNAをアフリカッメガエル卵母細胞に注入し、 本 発明のリガンドによって刺激すると細胞カルシウム濃度が上昇して、 calcium - activated chloride currentが生じる。 これは、 膜電位の変化としてとらえるこ とができる (Kイオン濃度勾配に変化がある場合も同様) 。 本発明のリガンドに よつて生じる本発明の受容体導入ァフリカッメガエル卵母細胞における上記反応 を利用して、 本発明のリガンドの本発明の受容体発現細胞に対する刺激活性を測 定することにより、 本発明のリガンドと本発明の受容体との結合性を変化させる 化合物をスクリーニングすることができる。 Receptor of the present invention (Example, TREM- 2) of the extracellular domain and CD3zeta fusion protein expression vector and NFAT- reporter gene (luciferase) and introduced expression cells of at 5xl0 4 cell / well in 24-well plates Seed and incubate for 48 hours. Cells 0 · 2ιηΜ 3 - isobutyl - washing with Hanks buffer Η7 · 4) containing methylxanthines, 0. 05% BSA and 20 mM HEPES (hereinafter, abbreviated as a reaction buffer). Then add 0.5 ml of reaction buffer and keep incubator for 30 minutes. Except for the reaction buffer 1, 0.25 ml of the reaction buffer was added to the cells, and then 1 本 of the ligand of the present invention or 1 μΜ of the ligand of the present invention and the test compound were added. Add 25 ml of application buffer to the cells and incubate at 37 ° C for 10 minutes. Lyse the cells with a cell lysing agent for Pitka Gene (Toyo Ink Mfg. Co., Ltd.) and add a luminescent substrate (Toyo Ink Mfg. Co., Ltd.) to the lysate. Luminescence from luciferase is measured with a luminometer, liquid scintillation counter or top counter. The amount of luminescence by luciferase in the case where only the ligand of the present invention is added and the case where 1 μM of the ligand of the present invention and the test compound are added are measured and compared. (12) When the receptor gene RNA of the present invention is injected into Xenopus oocytes and stimulated with the ligand of the present invention, the cellular calcium concentration increases, and calcium-activated chloride current is generated. This can be considered as a change in the membrane potential (even when there is a change in the K ion concentration gradient). Measuring the stimulatory activity of the ligand of the present invention on cells expressing the receptor of the present invention utilizing the above-mentioned reaction in the oocytes of the receptor-introduced A. facsimile produced by the ligand of the present invention. Thus, a compound that changes the binding between the ligand of the present invention and the receptor of the present invention can be screened.
具体的には、 本発明のリガンドを、 本発明の受容体遺伝子 RNA導入アフリカッ メガエル卵母細胞に接触させた場合と、 本発明のリガンドおよび試験化合物を、 本発明の受容体遺伝子 R A導入アフリカッメガエル卵母細胞に接触させた場合に おける、 細胞膜電位の変化を測定し、 比較することにより、 本発明のリガンドと 本発明の受容体との結合性を変化させる化合物をスクリーニングする。  Specifically, when the ligand of the present invention is brought into contact with the receptor gene RNA-introduced Xenopus laevis oocytes, the ligand of the present invention and the test compound are introduced into the receptor gene of the present invention. By measuring and comparing the change in cell membrane potential when brought into contact with the melanogaster oocyte, a compound that changes the binding between the ligand of the present invention and the receptor of the present invention is screened.
本方法において、 細胞膜電位変化を抑制する試験化合物を、 拮抗阻害能力のあ る候補物質として選択することができる。  In this method, a test compound that suppresses a change in cell membrane potential can be selected as a candidate substance having a competitive inhibition ability.
一方、 試験化合物のみを本発明の受容体遺伝子 RNA導入ァフリカツメガエル卵 母細胞に接触させ、 本発明のリガンドと同様な細胞膜電位変化を測定することに よりァゴニストのスタリ一二ングを行なうこともできる。  On the other hand, it is also possible to perform agonist staring by contacting the test compound alone with the receptor gene RNA-transfected A. frog oocytes and measuring a change in cell membrane potential similar to that of the ligand of the present invention. it can.
スクリーニング法の一具体例を以下に述べる。  One specific example of the screening method is described below.
氷冷して動けなくなった雌のアフリカッメガエルから取り出した、 卵母細胞塊 を、 MBS液 (88mM NaCl, ImM KC1, 0. 41mM CaCl2, 0. 33mM Ca (N03) 2, 0. 82mM MgS04, 2. 4mM NaHC03, lOmM HEPES; pH7. 4) に溶かしたコラーゲナーゼ Taken from female African Tsu mega El became stuck ice cooling, the oocytes mass, MBS solution (88mM NaCl, ImM KC1, 0. 41mM CaCl 2, 0. 33mM Ca (N0 3) 2, 0. 82mM MgS0 4, 2. 4mM NaHC0 3 , lOmM HEPES;. pH7 collagenase dissolved in 4)
(0. 5mg/ml) で卵塊がほぐれるまで 19°C、 ;!〜 6時間、 150rpmで処理する。 外液を MBS液に置換することで 3度洗浄し、 マイクロマニピュレーターで本発明の受容体 遺伝子 poly A付カロ cRNA (50ng/50nl) を卵母細胞にマイクロインジェクションす る。  (0.5 mg / ml) at 19 ° C until the egg mass is loosened; Process at 150 rpm for ~ 6 hours. The external solution is replaced with an MBS solution to wash the cells three times, and the oocytes are microinjected with calo cRNA (50 ng / 50 nl) having the receptor gene polyA of the present invention using a micromanipulator.
本発明の受容体遺伝子 mRNAは、 組織や細胞から調製してもよく、 プラスミドか ら in vitroで転写してもよい。 本発明の受容体遺伝子 mRNAを MBS液中で 20°Cで 3日 培養し、 これを Ringer液を流している voltage clamp装置のくぼみに置き、 電位 固定用ガラス微小電極およぴ電位測定用ガラス微小電極を細胞内に刺入し、 (-) 極は細胞外に置く。 電位が安定したら、 本発明のリガンドまたは本発明の リガンドおよび試験化合物を含む Ringer液を流して電位変化を記録する。 試験化 合物の影響は、 本発明の受容体遺伝子 RNA導入ァフリカッメガエル卵母細胞の細 胞膜電位変化を、 本発明のリガンドのみ含む Ringer液を流した場合と比較するこ とによって測定することができる。 The receptor gene mRNA of the present invention may be prepared from a tissue or a cell, or may be transcribed from a plasmid in vitro. The receptor gene mRNA of the present invention was cultured in MBS solution at 20 ° C. for 3 days, and this was placed in the cavity of a voltage clamp device in which Ringer solution was flowing, and the potential was measured. Insert the glass microelectrode for fixation and the glass microelectrode for potential measurement into the cell, and place the (-) electrode outside the cell. When the potential is stabilized, a change in potential is recorded by flowing a Ringer solution containing the ligand of the present invention or the ligand of the present invention and a test compound. The effect of the test compound was determined by comparing the change in the cell membrane potential of the oocyte of the receptor gene of the present invention into which RNA of the present invention was transfected with that of the Ringer solution containing only the ligand of the present invention. Can be measured.
細胞膜電位変化を抑制する化合物を拮抗阻害能力のある候補物質として選択す ることができる。  A compound that suppresses the change in cell membrane potential can be selected as a candidate substance having a competitive inhibition ability.
上記の系において、 電位の変化量を増大させると、 測定しやすくなるため、 各 種の G蛋白質遺伝子の poly A付加 RNAを導入してもよい。 また、 カルシウム存在下 で発光を生じるような蛋白質 (例、 aequorinなど) の遺伝子の poly A付加 RNAを 共ィンジェクシヨンすることにより、 膜電位変化ではなく発光量を測定すること もできる。  In the above-mentioned system, when the amount of change in the potential is increased, the measurement becomes easier, so that poly A-added RNA of each G protein gene may be introduced. Also, by co-injecting poly A-added RNA of a gene of a protein (eg, aequorin, etc.) that emits light in the presence of calcium, the amount of luminescence can be measured instead of a change in membrane potential.
本発明のリガンドと本発明の受容体との結合性を変化させる化合物またはその 塩のスクリーニング用キットは、 本発明の受容体または本発明の受容体を含有す る細胞もしくは細胞の膜画分、 および本発明のリガンドを含有する。  A screening kit for a compound or a salt thereof that alters the binding property between the ligand of the present invention and the receptor of the present invention includes a receptor of the present invention or a cell containing the receptor of the present invention or a membrane fraction of the cell, And a ligand of the invention.
本発明のスクリーユング用キットの例としては、 次のものが挙げられる。 1 . スクリーニング用試薬  Examples of the screening kit of the present invention include the following. 1. Screening reagent
(i) 測定用緩衝液および洗浄用緩衝液  (i) Measurement buffer and washing buffer
Hanks' Balanced Salt Solution (ギブコ社製) に、 0 . 0 5 %のゥシ血清アル プミン (シグマ社製) を加えたもの。  Hanks' Balanced Salt Solution (manufactured by Gibco) supplemented with 0.05% serum albumin (Sigma).
孔径 0 . 4 5 /z mのフィルターで濾過滅菌し、 4 °Cで保存するか、 または用時 調製しても良い。  The solution may be sterilized by filtration through a 0.45 / zm filter and stored at 4 ° C, or may be prepared at use.
(ii) 本発明の受容体標品  (ii) Receptor preparation of the present invention
本発明の受容体を発現させた C H O細胞を、 1 2穴プレートに 5 X 1 0 5個/ 穴で継代し、 3 7 °C、 5 % C 02、 9 5 % a i rで 2日間培養したもの。 CHO cells expressing the receptor of the present invention, 1 2-well plates and passaged 5 X 1 0 5 cells / well, cultured for 2 days at 3 7 ° C, 5% C 0 2, 9 5% air What you did.
(iii) 標識リガンド  (iii) Labeled ligand
〔 〕 、 〔1251〕 、 〔14C〕 、 〔32P〕 、 〔33P〕 、 〔35S〕 などの放射性同位元素で 標識した本発明のリガンドを適当な溶媒または緩衝液に溶解したものを、 4 °Cま たは一 20°Cにて保存し、 用時に測定用緩衝液にて 1 に希釈する。 A solution of the ligand of the present invention labeled with a radioisotope such as [], [ 125 1], [ 14 C], [ 32 P], [ 33 P], [ 35 S] in a suitable solvent or buffer Up to 4 ° C Alternatively, store at 20 ° C and dilute to 1 with assay buffer before use.
(iv) リガンド標準液  (iv) Ligand standard solution
本発明のリガンドを 0. 1 %ゥシ血清アルブミン (シグマ社製) を含む P B S で 1 mMとなるように溶解し、 一 20 °Cで保存する。  The ligand of the present invention is dissolved to a concentration of 1 mM in PBS containing 0.1% 社 serum albumin (manufactured by Sigma) and stored at 120 ° C.
2. 測定法 2. Measurement method
(i) 12穴組織培養用プレートにて培養した本発明の受容体を発現させた細胞 を、 測定用緩衝液 1 m 1で 2回洗浄した後、 490 ^ 1の測定用緩衝液を各穴に 加える。  (i) The cells expressing the receptor of the present invention cultured on a 12-well tissue culture plate were washed twice with 1 ml of the measurement buffer, and 490 ^ 1 of the measurement buffer was added to each well. Add to
(ii) 10一3〜 1 CT1QMの試験化合物溶液を 5 μ 1加えた後、 標識した本発明の リガンドを 5 μ 1加え、 室温にて 1時間反応させる。 非特異的結合量を知るため には試験化合物の代わりに 10— 3Μの本発明のリガンドを 5 μ 1加えておく。(ii) 10 one 3 ~ 1 CT 1Q M after addition test compound solution 5 mu 1, the ligands of the present invention labeled 5 mu 1 was added and reacted at room temperature for 1 hour. A supplementary 5 mu 1 ligand of the present invention the 10- 3 Micromax in place of the test compound to determine the amount of non-specific binding.
(iii) 反応液を除去し、 lm 1の洗浄用緩衝液で 3回洗浄する。 細胞に結合し た標識された本発明のリガンドを 0. 2 N N a OH- 1 % S D Sで溶解し、 4 m 1の液体シンチレーター A (和光純薬製) と混合する。 (iii) Remove the reaction solution and wash three times with lm1 of washing buffer. The labeled ligand of the present invention bound to cells is dissolved in 0.2 N NaOH-1% SDS and mixed with 4 ml of liquid scintillator A (manufactured by Wako Pure Chemical Industries, Ltd.).
(iv) 液体シンチレーションカウンター (ベックマン社製) を用いて放射活性を 測定し、 Percent Maximum Binding (PMB) を次式で求める。  (iv) The radioactivity is measured using a liquid scintillation counter (manufactured by Beckman), and the Percent Maximum Binding (PMB) is determined by the following equation.
PMB= [ (B-NS B) / (B0— NSB) ] X 100 PMB = [(B-NS B) / (B 0 — NSB)] X 100
PMB : Percent Maximum Binding  PMB: Percent Maximum Binding
B :検体を加えた時の値  B: Value when the sample is added
NSB : Non-specific Binding (非特異的結合量)  NSB: Non-specific Binding
B。 :最大結合量 ·  B. : Maximum binding amount ·
本発明のスクリーユ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う る化合物またはその塩は、 本発明のリガンドと本発明の受容体との結合を変化さ せる化合物あるいは本発明の受容体の活性を促進または阻害する化合物であり、 具体的には、 (i) 本発明の受容体を介して細胞刺激活性を有する化合物または その塩 (本発明の受容体ァゴニスト) 、 (ii) 該刺激活性を有しない化合物 (本 発明の受容体アンタゴニスト) 、 (iii) 本発明の受容体と本発明のリガンドと の結合力を促進する化合物、 (iv) 本発明の受容体と本発明のリガンドとの結合 力を阻害する化合物などである。 該化合物としては、 ペプチド、 蛋白質、 抗体、 非ぺプチド性化合物、 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、 血清などから選ばれた化合物などが挙げられ、 これら化合物は新規 な化合物であってもよいし、 公知の化合物であってもよい。 A compound or a salt thereof obtained by using the screening method or the screening kit of the present invention is a compound that alters the binding between the ligand of the present invention and the receptor of the present invention or the activity of the receptor of the present invention. A compound that promotes or inhibits, specifically, (i) a compound having a cell stimulating activity via the receptor of the present invention or a salt thereof (receptor agonist of the present invention); and (ii) a compound having the stimulating activity. (Iii) a compound that promotes the binding strength between the receptor of the present invention and the ligand of the present invention; (iv) a binding force between the receptor of the present invention and the ligand of the present invention. And the like. Such compounds include peptides, proteins, antibodies, Examples include compounds selected from non-peptide compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and serum.These compounds may be novel compounds. It may be a known compound.
該化合物の塩としては、 前記した本発明の受容体の塩と同様のものが用いられ る。  As the salt of the compound, those similar to the aforementioned salts of the receptor of the present invention are used.
上記本発明の受容体ァゴニストであるか、 またはアンタゴニストであるかの評 価方法は、 例えば、 以下の (i) または (i i) に従えばよい。  The method of evaluating whether the receptor agonist of the present invention is an antagonist or an antagonist may be, for example, according to the following (i) or (ii).
(i) 前記 (a) 〜 (c) のスクリ一二ング方法で示されるバインディング .アツ セィを行い、 本発明のリガンドと本発明の受容体との結合性を変化させる (特に. 結合を阻害する) 化合物を得た後、 該化合物が上記した本発明の受容体を介する 細胞刺激活性を有しているか否かを測定する。 細胞刺激活性を有する化合物また はその塩は本発明の受容体ァゴニスト (ァゴ二スト) であり、 該活性を有しない 化合物またはその塩は本発明の受容体アンタゴニスト (アンタゴニスト) である: (i) binding by the screening method described in the above (a) to (c), which is performed to change the binding between the ligand of the present invention and the receptor of the present invention. After obtaining the compound, it is determined whether or not the compound has the above-described receptor-mediated cell stimulating activity of the present invention. A compound having a cell stimulating activity or a salt thereof is the receptor agonist (agonist) of the present invention, and a compound having no such activity or a salt thereof is a receptor antagonist (antagonist) of the present invention:
(i i) (a)試験化合物を本発明の受容体を含有する細胞に接触させ、 本発明の受 容体を介した細胞刺激活性を測定する。 細胞刺激活性を有する化合物またはその 塩は本発明の受容体ァゴニストである。 (ii) (a) The test compound is brought into contact with a cell containing the receptor of the present invention, and the cell stimulating activity of the receptor of the present invention is measured. The compound having a cell stimulating activity or a salt thereof is the receptor agonist of the present invention.
(b)本発明のリガンドを本発明の受容体を含有する細胞に接触させた場合と、 本 発明のリガンドおよび試験化合物を本発明の受容体を含有する細胞に接触させた 場合における、 本発明の受容体を介した細胞刺激活性を測定し、 比較する。 本発 明の受容体を活性化する化合物による細胞刺激活性を減少させ得る化合物または その塩は本発明の受容体アンタゴニストである。 '  (b) The present invention in the case where the ligand of the present invention is brought into contact with a cell containing the receptor of the present invention, and in the case where the ligand of the present invention and a test compound are brought into contact with a cell containing the receptor of the present invention. The cell stimulating activity mediated by the receptor is measured and compared. A compound or a salt thereof capable of decreasing the cell stimulating activity by the compound activating the receptor of the present invention is the receptor antagonist of the present invention. '
前述したように、 本発明のリガンドは、 DAP12リン酸化促進活性、 ERKリン酸ィ匕 促進活性、 インスリンシグナル伝達抑制活性、 TNFひ産生促進活性、 グルコース 取り込み阻害活性などを有する。  As described above, the ligand of the present invention has a DAP12 phosphorylation promoting activity, an ERK phosphorylation promoting activity, an insulin signaling inhibitory activity, a TNF production promoting activity, a glucose uptake inhibiting activity, and the like.
従って、 本発明の受容体ァゴニス トは、 本発明のリガンドが有する生理活性 Therefore, the receptor agonist of the present invention has the physiological activity of the ligand of the present invention.
(例、 DAP12リン酸化促進活性、 ERKリン酸化促進活性、 インスリンシグナル伝達 抑制活性、 TNF a産生促進活性、 グルコース取り込み阻害活性など) と同様の作 用を有しており、 安全で低毒性な医薬、 例えば、 低血糖の予防 ·治療剤などとし て有用である。 本発明の受容体アンタゴニストほ、 本発明のリガンドが有する生理活性 (例、 DAP12リン酸化促進活性、 ERKリン酸化促進活性、 インスリンシグナル伝達抑制活 性、 TNF o;産生促進活性、 グルコース取り込み阻害活性など) を抑制することが できるので、 安全で低毒性な医薬、 例えば、 インスリン抵抗性の改善剤、 耐糠能 異常、 糖尿病、 肥満症、 高脂血症、 動脈硬化、 高血圧症または心疾患の予防 ·治 療剤などとして有用である。 (E.g., DAP12 phosphorylation promoting activity, ERK phosphorylation promoting activity, insulin signaling inhibitory activity, TNF a production promoting activity, glucose uptake inhibitory activity, etc.) For example, it is useful as an agent for preventing or treating hypoglycemia. Physiological activity possessed by the ligand of the present invention (eg, DAP12 phosphorylation promoting activity, ERK phosphorylation promoting activity, insulin signaling inhibition activity, TNF o; production promoting activity, glucose uptake inhibiting activity, etc.) ) Can be controlled by safe and low toxic drugs, such as insulin resistance improver, impaired bran tolerance, diabetes, obesity, hyperlipidemia, arteriosclerosis, prevention of hypertension or heart disease · Useful as a therapeutic agent.
本発明の受容体と本発明のリガンドとの結合力を促進する化合物は、 安全で低 毒性な医薬、 例えば、 低血糖の予防 ·治療剤などとして有用である。  The compound that promotes the binding strength between the receptor of the present invention and the ligand of the present invention is useful as a safe and low-toxicity drug, for example, an agent for preventing or treating hypoglycemia.
本発明の受容体と本発明のリガンドとの結合力を阻害する化合物は、 安全で低 毒性な医薬、 例えばインスリン抵抗性の改善剤、 耐糖能異常、 糖尿病、 肥満症、 高脂血症、 動脈硬化、 高血圧症または心疾患の予防 ·治療剤などとして有用であ る。  Compounds that inhibit the binding between the receptor of the present invention and the ligand of the present invention are safe and low-toxic drugs, such as insulin resistance improvers, impaired glucose tolerance, diabetes, obesity, hyperlipidemia, and arteries. It is useful as a preventive and therapeutic agent for sclerosis, hypertension or heart disease.
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、 蛋白質、 抗体、 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、 血清など 力 ら選ばれた化合物であり、 本発明の受容体と本発明のリガンドとの結合性を変 ィ匕させる化合物、 本発明の受容体め活性または機能を促進または阻害する化合物、 本発明の受容体の遺伝子の発現を促進または阻害 (発現量を増加または減少) す る化合物などである。  Compounds or salts thereof obtained using the screening method or screening kit of the present invention include, for example, peptides, proteins, antibodies, non-peptide compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissues A compound selected from the group consisting of extract, serum and the like, which compound alters the binding between the receptor of the present invention and the ligand of the present invention, and a compound which promotes or inhibits the activity or function of the receptor of the present invention And compounds that promote or inhibit (increase or decrease the expression level) the expression of the receptor gene of the present invention.
該化合物の塩としては、 前記した本発明の受容体の塩と同様のものが用いられ る。  As the salt of the compound, those similar to the aforementioned salts of the receptor of the present invention are used.
本発明のスクリーニング方法またはスクリーニング用キットを用いて得られる 化合物またはその塩を上記の医薬 (予防 ·治療剤など) として使用する場合、 常 套手段に従つて実施することができる。  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 medicament (prophylactic / therapeutic agent, etc.), it can be carried out according to a conventional method.
該化合物またはその塩は、 例えば、 必要に応じて糖衣を施した錠剤、 カプセル 剤、 エリキシル剤、 マイクロカプセル剤などとして経口的に、 あるいは水もしく はそれ以外の薬学的に許容し得る液との無菌性溶液、 または懸濁液剤などの注射 剤の形で非経口的に使用できる。 例えば、 該化合物またはその塩を生理学的に認 められる担体、 香味剤、 賦形剤、 べヒクル、 防腐剤、 安定剤、 結合剤などととも に一般に認められた製剤実施に要汆される単位用量形態で混和することによって 製造することができる。 これら製剤における有効成分量は指示された範囲の適当 な用量が得られるようにするものである。 The compound or a salt thereof can be used, for example, as a sugar-coated tablet, capsule, elixir, microcapsule, etc., orally, or with water or another pharmaceutically acceptable liquid. It can be used parenterally in the form of injectable preparations such as sterile solutions or suspensions. For example, the compound or a salt thereof is combined with a physiologically acceptable carrier, flavoring agent, excipient, vehicle, preservative, stabilizer, binder and the like. It can be manufactured by mixing in the unit dosage form generally required for the practice of preparations. The amount of active ingredient in these preparations is such that a suitable dosage in the specified range can be obtained.
錠剤、 力プセル剤などに混和することができる添加剤としては、 例えば、 ゼラ チン、 コーンスターチ、 トラガント、 アラビアゴムのような結合剤、 結晶性セル ロースのような賦形剤、 コーンスターチ、 ゼラチン、 アルギン酸などのような膨 ィ匕剤、 ステアリン酸マグネシゥムのような潤滑剤、 ショ糖、 乳糖またはサッカリ ンのような甘味剤、 ペパーミント、 ァカモノ油またはチェリーのような香味剤な どが用いられる。 調剤単位形態がカプセルである場合には、 前記タイプの材料に さらに油脂のような液状担体を含有することができる。 注射のための無菌組成物 は注射用水のようなべヒクル中の活性物質、 胡麻油、 椰子油などのような天然産 出植物油などを溶解または懸濁させるなどの通常の製剤実施に従つて処方するこ とができる。  Excipients that can be incorporated into tablets, forceps, etc. include, for example, binders such as gelatin, corn starch, tragacanth, gum arabic, excipients such as crystalline cellulose, corn starch, gelatin, alginic acid For example, swelling agents such as sucrose, lubricating agents such as magnesium stearate, sweetening agents such as sucrose, lactose or saccharin, and flavoring agents such as peppermint, cocoa oil or cherry are used. When the unit dosage form is a capsule, a liquid carrier such as oils and fats can be further contained in the above-mentioned type of material. Sterile compositions for injection are formulated according to standard pharmaceutical practice, such as dissolving or suspending the active substance in vehicles such as water for injection, and naturally occurring vegetable oils such as sesame oil and coconut oil. Can be.
注射用の水性液としては、 例えば、 生理食塩水、 ブドウ糖やその他の補助薬を 含む等張液 (例えば、 D—ソルビトール、 D—マンニトール、 塩化ナトリウムな ど) などがあげられ、 適当な溶解補助剤、 例えば、 アルコール (例えば、 ェタノ 一ノレなど) 、 ポリアルコール (例えば、 プロピレングリコーノレ、 ポリエチレング リコールなど) 、 非イオン性界面活性剤 (例えば、 ポリソルベート 8 0™、 H C O— 5 0など) などと併用してもよレ、。 油性液としては、 例えば、 ゴマ油、 大豆 油などがあげられ、 溶解補助剤として安息香酸ベンジル、 ベンジルアルコールな どと併用してもよレ、。 また、 該化合物またはその塩を、 緩衝剤 (例えば、 リン酸 塩緩衝液、 酢酸ナトリウム緩衝液など) 、 無痛化剤 (例えば、 塩化ベンザルコニ ゥム、 塩酸プロカインなど) 、 安定剤 . (例えば、 ヒ ト血清アルブミン、 ポリェチ レングリコールなど) 、保存剤 (例えば、 ベンジルアルコール、 フエノールな ど) 、 酸ィ匕防止剤などと配合してもよい。 調製された注射液は、 通常、 適当なァ ンプルに充填される。  Examples of aqueous solutions for injection include physiological saline, isotonic solutions containing glucose and other adjuvants (eg, D-sorbitol, D-mannitol, sodium chloride, etc.). Agents, for example, alcohols (eg, ethanol), polyalcohols (eg, propylene glycol, polyethylene glycol, etc.), nonionic surfactants (eg, polysorbate 80 ™, HCO-50, etc.) You can use it together. Examples of the oily liquid include sesame oil and soybean oil, which may be used in combination with solubilizing agents such as benzyl benzoate and benzyl alcohol. In addition, the compound or a salt thereof is treated with a buffer (eg, phosphate buffer, sodium acetate buffer, etc.), a soothing agent (eg, benzalkonium chloride, procaine hydrochloride, etc.), a stabilizer. Serum albumin, polyethylene glycol, etc.), preservatives (eg, benzyl alcohol, phenol, etc.), antioxidants and the like. The prepared injection solution is usually filled in an appropriate sample.
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、 ブタ、 ゥシ、 ゥマ、 トリ、 ネコ、 ィヌ、 サル、 チンパンジーなど) に対して投与することができる。 該化合物またはその塩の投与量ほ、 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。 The preparations obtained in this way are safe and low toxic, and can be used, for example, in humans or warm-blooded animals (eg, mice, rats, puppies, sheep, pigs, puppies, pumas, birds, cats, dogs, Monkeys, chimpanzees, etc.). There is a difference depending on the dose of the compound or its salt, its action, target disease, subject of administration, route of administration and the like.
例えば、 糖尿病患者 (体重 6 O k g当たり) に、 一日につき本発明の受容体ァ ンタゴニストを約 0. 1〜: 100mg、 好ましくは約 1. 0〜5 Qmg、 より好 ましくは約 1. 0〜2 Omg経口投与する。 非経口的に投与する場合、 例えば、 本発明の受容体アンタゴ-ストを注射剤の形で糖尿病患者 (体重 60 k g当た り) 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. :!〜 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、 体重 60 k g当たりに換算した量を 投与することができる。  For example, in a diabetic patient (per 6 kg of body weight), about 0.1 to 100 mg, preferably about 1.0 to 5 Qmg, more preferably about 1.1, of the receptor antagonist of the present invention per day. Administer orally 0-2 Omg. When administered parenterally, for example, when the receptor antagonist of the present invention is administered to a diabetic patient (body weight: 60 kg) in the form of an injection, the compound can be administered in an amount of about 0.01 to about 0.01 per day. It is convenient to administer 30 mg, preferably about 0.:!2 Omg, more preferably about 0.1-1 Omg, by intravenous injection. In the case of other animals, the dose can be administered per 60 kg body weight.
〔2〕 本発明の受容体の定量 (2) Quantification of the receptor of the present invention
本発明の抗体は、 本発明の受容体を特異的に認識することができるので、 被検 液中の本発明の受容体の定量、 特にサンドィ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。  Since the antibody of the present invention can specifically recognize the receptor of the present invention, it can be used for quantification of the receptor of the present invention in a test solution, particularly for quantification by a sandwich immunoassay. .
すなわち、 本発明は、  That is, the present invention
(i) 本発明の抗体と、 被検液および標識化された本発明の受容体とを競合的に 反応させ、 該抗体に結合した標識化された本発明の受容体の割合を測定すること を特徴とする被検液中の本発明の受容体の定量法、 および  (i) Competitively reacting the antibody of the present invention with a test solution and a labeled receptor of the present invention, and measuring the ratio of the labeled receptor of the present invention bound to the antibody. A method for quantifying the receptor of the present invention in a test solution, comprising:
(ii) 被検液と担体上に不溶化した本発明の抗体および標識化された本発明の別 の抗体とを同時あるいは連続的に反応させたのち、 不溶化担体上の標識剤の活性 を測定することを特徴とする被検液中の本発明の受容体の定量法を提供する。 上記 (ii) の定量法においては、 一方の抗体が本発明の受容体の N端部を認識 する抗体で、 他方の抗体が本発明の受容体の C端部に反応する抗体であることが 望ましい。  (ii) After reacting the test solution with the antibody of the present invention insolubilized on the carrier and another labeled antibody of the present invention simultaneously or continuously, the activity of the labeling agent on the insolubilized carrier is measured. A method for quantifying the receptor of the present invention in a test solution is provided. In the quantification method (ii), one antibody may be an antibody that recognizes the N-terminal of the receptor of the present invention, and the other antibody may be an antibody that reacts with the C-terminal of the receptor of the present invention. desirable.
また、 本発明の受容体に対するモノクローナル抗体を用いて本発明の受容体の 定量を行うことができるほか、 組織染色等による検出を行なうこともできる。 こ れらの目的には、 抗体分子そのものを用いてもよく、 また、 抗体分子の F(a b ')2、 F a b'、 あるいは F a b画分を用いてもよい。 本発明の抗体を用いる本発明の ¾容体の定量法は、 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、 ポリペプチド量) に対応した抗体、 抗 原もしくは抗体一抗原複合体の量を化学的または物理的手段により検出し、 これ を既知量の抗原を含む標準液を用いて作製した標準曲線より算出する測定法であ れば、 いずれの測定法を用いてもよい。 例えば、 ネフロメ トリー、 競合法、 ィム ノメトリック法おょぴサンドイッチ法が好適に用いられるが、 感度、 特異性の点 で、 後述するサンドィツチ法を用いるのが特に好ましい。 In addition, the receptor of the present invention can be quantified using a monoclonal antibody against the receptor of the present invention, and can also be detected by tissue staining or the like. For these purposes, the antibody molecule itself may be used, or the F (ab ′) 2 , Fab ′, or Fab fraction of the antibody molecule may be used. The method for quantifying the receptor of the present invention using the antibody of the present invention is not particularly limited, and may be an antibody, an antigen, or an antibody corresponding to the amount of antigen (eg, the amount of polypeptide) in the test solution. Any method that detects the amount of the complex by chemical or physical means and calculates this from a standard curve prepared using a standard solution containing a known amount of antigen can be used. Good. For example, a nephelometry, a competition method, an immunometric method, and a sandwich method are preferably used, and in terms of sensitivity and specificity, it is particularly preferable to use a sandwich method described later.
標識物質を用いる測定法に用いられる標識剤としては、 例えば、 放射性同位元 素、 酵素、 蛍光物質、 発光物質などが用いられる。 放射性同位元素としては、 例 えば、 〔1251〕 、 〔1311〕 、 〔 〕 、 〔14c〕 などが用いられる。 上記酵素としては、 安定で比活性の大きなものが好ましく、 例えば、 β—ガラタトシダーゼ、 β一グ ルコシダーゼ、 アル力リフォスファタ一ゼ、 ハ。ーォキシダーゼ、 リンゴ酸脱水素 酵素などが用いられる。 蛍光物質としては、 例えば、 フルォレスカミン、 フルォ レツセンィソチオシァネートなどが用いられる。 発光物質としては、 例えば、 ル ミノール、 ルミノール誘導体、 ルシフェリン、 ルシゲニンなどが用いられる。 さ らに、 抗体あるいは抗原と標識剤との結合にピオチン一ァビジン系を用いること もできる。 As a labeling agent used in a measurement method using a labeling substance, for example, a radioisotope, an enzyme, a fluorescent substance, a luminescent substance and the like are used. Radioisotopes, if example embodiment, [125 1], [131 1], [], are used like [14 c]. As the above-mentioned enzymes, those which are stable and have a large specific activity are preferable, and for example, β-galatatosidase, β-glucosidase, lipophosphatase, and c. Oxidase, malate dehydrogenase and the like are used. As the fluorescent substance, for example, fluorescamine, fluorescein isothiocyanate and the like are used. As the luminescent substance, for example, luminol, luminol derivative, luciferin, lucigenin and the like are used. Furthermore, a biotin-avidin system can be used for binding the antibody or antigen to the labeling agent.
抗原あるいは抗体の不溶化に当っては、 物理吸着を用いてもよく、 また通常ポ リペプチド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 るいはガラス等があげられる。  For the insolubilization of the antigen or antibody, physical adsorption may be used, or a method using a chemical bond usually used for insolubilizing or immobilizing a polypeptide or an enzyme may be used. Examples of the carrier include insoluble polysaccharides such as agarose, dextran, and cellulose; synthetic resins such as polystyrene, polyacrylamide, and silicon; and glass.
サンドィツチ法においては不溶化した本発明のモノクローナル抗体に被検液を 反応させ (1次反応) 、 さらに標識化した別の本発明のモノクローナル抗体を反 応させ (2次反応) たのち、 不溶化担体上の標識剤の活性を測定することにより 被検液中の受容体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レ、。 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、 サンドィ ツ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種類である必要ほなく、 測定感度を向上させる等の目的で 2種 類以上の抗体の混合物を用いてもよい。 In the sandwich method, the test solution is reacted with the insolubilized monoclonal antibody of the present invention (primary reaction), and further reacted with another labeled monoclonal antibody of the present invention (secondary reaction). By measuring the activity of the labeling agent, the amount of the receptor in the test solution can be quantified. The primary reaction and the secondary reaction may be performed in the reverse order, or may be performed simultaneously or at staggered times. The labeling agent and the method of insolubilization can be in accordance with those described above. It is also used as an antibody for solid phase or an antibody for labeling in immunoassay by the sandwich method. One type of antibody is not necessarily required, and a mixture of two or more types of antibodies may be used for the purpose of improving measurement sensitivity and the like.
本発明のサンドィツチ法による本発明の受容体の測定法においては、 1次反応 と 2次反応に用いられる本発明のモノクローナル抗体は、 本発明の受容体の結合 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次反応おょぴ 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、 本発明の受容 体の 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 、られる。  In the method for measuring the receptor of the present invention by the sandwich method of the present invention, the monoclonal antibody of the present invention used in the primary reaction and the secondary reaction is preferably an antibody having a different site to which the receptor of the present invention binds. Can be That is, the antibody used in the primary reaction and the secondary reaction is, for example, when the antibody used in the secondary reaction recognizes the C-terminal of the receptor of the present invention, the antibody used in the primary reaction is Preferably, an antibody which recognizes other than the C-terminal, for example, the N-terminal, is used.
本発明のモノクローナル抗体をサンドィツチ法以外の測定システム、 例えば、 競合法、 ィムノメ トリック法あるいはネフロメトリーなどに用いることができる。 競合法では、 被検液中の抗原と標識抗原とを抗体に対して競合的に反応させた のち、 未反応の標識抗原 (F ) と、 抗体と結合した標識抗原 (B ) とを分離し (B /F分離) 、 B , Fいずれかの標識量を測定し、 被検液中の抗原量を定量す る。 本反応法には、 抗体として可溶性抗体を用い、 BZ F分離をポリエチレング リコール、 前記抗体に対する第 2抗体などを用いる液相法、 および、 第 1抗体と して固相化抗体を用いるか、 あるいは、 第 1抗体は可溶性のものを用い第 2抗体 として固相化抗体を用いる固相化法とが用いられる。  The monoclonal antibody of the present invention can be used in a measurement system other than the sandwich method, for example, a competition method, an immunometric method, or a nephrometry. In the competition method, after the antigen in the test solution and the labeled antigen are allowed to react competitively with the antibody, the unreacted labeled antigen (F) and the labeled antigen (B) bound to the antibody are separated. (B / F separation) The labeling amount of either B or F is measured, and the amount of antigen in the test solution is quantified. In this reaction method, a soluble antibody is used as the antibody, BZF separation is performed using polyethylene glycol, a liquid phase method using a second antibody against the antibody, or an immobilized antibody is used as the first antibody. Alternatively, an immobilization method using a soluble first antibody and an immobilized antibody as the second antibody is used.
ィムノメトリック法では、 被検液中の抗原と固相化抗原とを一定量の標識化抗 体に対して競合反応させた後固相と液相を分離するか、 あるいは、 被検液中の抗 原と過剰量の標識化抗体とを反応させ、 次に固相化抗原を加え未反応の標識化抗 体を固相に結合させたのち、 固相と液相を分離する。 次に、 いずれかの相の標識 量を測定し被検液中の抗原量を定量する。  In the immunometric method, the antigen in the test solution and the immobilized antigen are subjected to a competitive reaction with a certain amount of labeled antibody, and then the solid phase and the liquid phase are separated. After reacting the antigen with an excess amount of the labeled antibody, the immobilized antigen is added to bind the unreacted labeled antibody to the solid phase, and then the solid phase and the liquid phase are separated. Next, the amount of the label in either phase is measured to determine the amount of the antigen in the test solution.
また、 ネフロメ トリーでは、 ゲル内あるいは溶液中で抗原抗体反応の結果生じ た不溶性の沈降物の量を測定する。 被検液中の抗原量が僅かであり、 少量の沈降 物しか得られない場合にもレーザーの散乱を利用するレーザーネフロメトリーな どが好適に用いられる。  In nephelometry, the amount of insoluble sediment resulting from an antigen-antibody reaction in a gel or in a solution is measured. Even when the amount of antigen in the test solution is small and only a small amount of sediment is obtained, laser nephrometry utilizing laser scattering is preferably used.
これら個々の免疫学的測定法を本発明の定量方法に適用するにあたっては、 特 別の条件、 操作等の設定は必要とされない。 それぞれの方法における通常の条件、 操作法に当業者の通常の技術的配慮を加えて本発明の受容体の測定系を構築すれ ばよレ、。 これらの一般的な技術手段の詳細については、 総説、 成書などを参照す ることができる。 In applying these individual immunoassays to the quantification method of the present invention, no special conditions, operations, and the like need to be set. The system for measuring the receptor of the present invention can be constructed by adding ordinary technical considerations to those skilled in the art to the ordinary conditions and procedures in each method. Bye, For details of these general technical means, reference can be made to reviews and written documents.
例えば、 入江 寛編 「ラジオィムノアツセィ」 (講談社、 昭和 4 9年発行) 、 入江 寛編 「続ラジオィムノアツセィ」 (講談社、 昭和 5 4年発行) 、 石川栄治 ら編 「酵素免疫測定法」 (医学書院、 昭和 5 3年発行) 、 石川栄治ら編 「酵素免 疫測定法」 (第 2版) (医学書院、 昭和 5 7年発行) 、 石川栄治ら編 「酵素免疫 測定法」 (第 3版) (医学書院、 昭和 6 2年発行) 、 「Methods in  For example, edited by Hiro Irie, "Radio Nonotsusei" (Kodansha, published in Showa 49), edited by Hiroshi Irie, "Radio Imunoatsushi" (Kodansha, published in 1954), Eiji Ishikawa et al. "Measurement Method" (Medical Shoin, published in 1958), edited by Eiji Ishikawa et al., "Enzyme Immunoassay" (Second Edition) (Medical School, published in 1977), Eiji Ishikawa, et al., "Enzyme Immunoassay" (3rd edition) (Issue Shoin, published in 1962), "Methods in
ENZYM0L0GY」 Vol. 70 (Immunochemical Techniques (Part A) ) 同書 Vol. ENZYM0L0GY '' Vol. 70 (Immunochemical Techniques (Part A)) Ibid.
73 (Immunochemical Techniques (Part B) )、 同書 Vol. 74 (Immunochemical 73 (Immunochemical Techniques (Part B)), ibid.Vol. 74 (Immunochemical Techniques)
Techniques (Part し) )、 同書 Vol. 84 (Immunochemical Techniques (Part D : selected Immunoassays) ) 同書 Vol. 92 (Immunochemical Techniques (Part E : Monoclonal Antibodies and Genera丄 Immunoassay Methods) )、 同 Vol. Vol. 84 (Immunochemical Techniques (Part D: selected Immunoassays)), Vol. 92 (Immunochemical Techniques (Part E: Monoclonal Antibodies and Genera and Immunoassay Methods)), ibid.
121 (Immunochemical Techniques (Part I : Hybridoma Technology and 121 (Immunochemical Techniques (Part I: Hybridoma Technology and
Monoclonal Antibodies) ) (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 Antibodies)) (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、 本発明の受容体を感 度良く定量することができる。  As described above, the receptor of the present invention can be quantified with good 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受容体の濃度を定量することによつ て、 本発明の受容体の濃度の増加が検出された場合、 例えば、 インスリン抵抗性、 耐糖能異常、 糖尿病、 肥満症、 高脂血症、 動脈硬化、 高血圧症または心疾患など の疾病である、 または将来罹患する可能性が高いと診断することができる。 また、 本発明の受容体の濃度の減少が検出された場合には、 例えば、 低血糖などの疾病 である、 または将来罹患する可能性が高いと診断することができる。  Furthermore, when an increase in the concentration of the receptor of the present invention is detected by quantifying the concentration of the receptor of the present invention using the antibody of the present invention, for example, insulin resistance, impaired glucose tolerance, etc. It can be diagnosed as a disease such as diabetes, obesity, hyperlipidemia, arteriosclerosis, hypertension or heart disease, or is likely to be affected in the future. In addition, when a decrease in the concentration of the receptor of the present invention is detected, it can be diagnosed that the disease is, for example, a disease such as hypoglycemia, or that the disease is likely to be caused in the future.
また、 本発明の抗体は、 体液や組織などの被検体中に存在する本発明の受容体 を検出するために使用することができる。 また、 本発明の受容体を精製するため 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受容体の検出、 被検 細胞内における本発明の受容体の挙動の分析などのために使用することができる。  Further, the antibody of the present invention can be used for detecting the receptor 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 receptor of the present invention, detection of the receptor of the present invention in each fraction during purification, analysis of the behavior of the receptor of the present invention in test cells, etc. Can be used for
〔3〕 本発明のリガンドを含有してなる低血糖の予防 ·治療剤 本発明のリガンドは、 MP12リン酸化促進活性、 ERKリン酸化促進活性、 インス リンシグナル伝達抑制活性、 TNF α産生促進活性、 グルコース取り込み阻害活性 などを有する。 したがって、 本発明のリガンドに異常があったり、 欠損している 場合には、 例えば、 低血糖などが発症する。 [3] An agent for preventing and treating hypoglycemia comprising the ligand of the present invention The ligand of the present invention has MP12 phosphorylation promoting activity, ERK phosphorylation promoting activity, insulin signaling inhibition activity, TNFα production promoting activity, glucose uptake inhibitory activity, and the like. Therefore, when the ligand of the present invention is abnormal or defective, for example, hypoglycemia develops.
したがって、 本発明のリガンドは、:例えば低血糖などの予防 ·治療剤などの安 全な医薬として使用することができる。  Therefore, the ligand of the present invention can be used as a safe medicament such as an agent for preventing or treating hypoglycemia and the like.
本発明のリガンドを上記の治療 ·予防剤として使用する場合は、 少なくとも 90%、 好ましくは 95%以上、 より好ましくは 98%以上、 さらに好ましくは 99%以 上に精製されたものを使用するのが好ましい。  When the ligand of the present invention is used as the above-mentioned therapeutic / prophylactic agent, it should be purified to at least 90%, preferably 95% or more, more preferably 98% or more, and still more preferably 99% or more. Is preferred.
本発明のリガンドは、 例えば、 必要に応じて糖衣を施した錠剤、 カプセル剤、 エリキシノレ剤、 マイクロカプセノレ剤などとして経口的に、 あるいは水もしくはそ れ以外の薬学的に許容し得る液との無菌性溶液、 または懸濁液剤などの注射剤の 形で非経口的に使用できる。 例えば、 本発明のリガンドを生理学的に認められる 担体、 香味剤、 賦形剤、 べヒクル、 防腐剤、 安定剤、 結合剤などとともに一般に 認められた製剤実施に要求される単位用量形態で混和することによって製造する ことができる。 これら製剤における有効成分量は指示された範囲の適当な用量が 得られるようにするものである。  The ligand of the present invention can be used, for example, in the form of tablets, capsules, elixirs, microcapsenoles, and the like, which are sugar-coated as required, or with water or other pharmaceutically acceptable liquids. It can be used parenterally in the form of injections, such as sterile solutions or suspensions. For example, the ligand of the present invention is mixed with physiologically acceptable carriers, flavors, excipients, vehicles, preservatives, stabilizers, binders, and the like in a unit dosage form generally required for the practice of pharmaceutical preparations. It can be manufactured by The amount of the active ingredient in these preparations is such that a suitable dosage in the specified range can be obtained.
錠剤、 カプセル剤などに混和することができる添加剤としては、 例えば、 ゼラ チン、 コーンスターチ、 トラガント、 アラビアゴムのような結合剤、 結晶性セル ロースのような賦形剤、 コーンスターチ、 ゼラチン、 アルギン酸などのような月彭 化剤、 ステアリン酸マグネシウムのような潤滑剤、 ショ糖、 乳糖またはサッカリ ンのような甘味剤、 ペパーミント、 ァカモノ油またはチェリーのような香味剤な どが用いられる。 調剤単位形態がカプセルである場合には、 前記タイプの材料に さらに油脂のような液状担体を含有することができる。 注射のための無菌組成物 は注射用水のようなべヒクル中の活性物質、 胡麻油、 椰子油などのような天然産 出植物油などを溶解または懸濁させるなどの通常の製剤実施に従って処方するこ とができる。  Additives that can be incorporated into tablets, capsules, etc. include, for example, binders such as gelatin, corn starch, tragacanth, gum arabic, excipients such as crystalline cellulose, corn starch, gelatin, alginic acid Lubricating agents such as magnesium stearate, sweetening agents such as sucrose, lactose or saccharin, and flavoring agents such as peppermint, cocoa oil or cherry. When the unit dosage form is a capsule, a liquid carrier such as oils and fats can be further contained in the above-mentioned type of material. Sterile compositions for injection can be formulated according to normal pharmaceutical practice, such as dissolving or suspending the active substance in vehicles such as water for injection, and naturally occurring vegetable oils such as sesame oil and coconut oil. it can.
注射用の水性液としては、 例えば、 生理食塩水、 ブドウ糖やその他の補助薬を 含む等張液 (例えば、 D-ソルビトール、 D -マンニトール、 塩ィヒナトリウムなど) などが挙げられ、 適当な溶解補助剤、 例えば、 アルコール (例えば、 エタノール など) 、 ポリアルコール (例えば、 プロピレングリコール、 ポリエチレングリコ ールなど) 、 非イオン性界面活性剤 (例えば、 ポリソルベート 80™、 HC0- 50な ど) などと併用してもよい。 油生液としては、 例えば、 ゴマ油、 大豆油などが挙 げられ、 溶解補助剤として安息香酸ベンジル、 ベンジルアルコールなどと併用し てもよい。 また、 緩衝剤 (例えば、 リン酸塩緩衝液、 酢酸ナトリウム緩衝液な ど) 、 無痛化剤 (例えば、 塩化ベンザルコニゥム、 塩酸プロ力インなど) 、 安定 剤 (例えば、 ヒト血清アルブミン、 ポリエチレングリコールなど) 、 保存剤 (例 えば、 ベンジルアルコール、 フエノールなど) 、 酸化防止剤などと配合してもよ い。 調製された注射液は、 通常、 適当なアンプルに充填される。 Aqueous liquids for injection include, for example, saline, isotonic solutions containing dextrose and other auxiliaries (eg, D-sorbitol, D-mannitol, sodium salt, etc.) Suitable solubilizers, for example, alcohols (eg, ethanol, etc.), polyalcohols (eg, propylene glycol, polyethylene glycol, etc.), nonionic surfactants (eg, Polysorbate 80 ™, HC0 -50 etc.). Examples of oily liquor include sesame oil and soybean oil, and may be used in combination with solubilizers such as benzyl benzoate and benzyl alcohol. In addition, buffers (eg, phosphate buffer, sodium acetate buffer, etc.), soothing agents (eg, benzalkonium chloride, proforce hydrochloride, etc.), stabilizers (eg, human serum albumin, polyethylene glycol, etc.) , Preservatives (eg, benzyl alcohol, phenol, etc.), antioxidants and the like. The prepared injection is usually filled in an appropriate ampoule.
このようにして得られる製剤は、 安全で低毒性であるので、 例えば、 温血動物 (例えば、 ヒト、 ラット、 マウス、 モルモット、 ゥサギ、 トリ、 ヒッジ、 ブタ、 ゥシ、 ゥマ、 ネコ、 ィヌ、 サル、 チンパンジーなど) に対して投与することがで きる。  The preparations obtained in this way are safe and have low toxicity, for example, warm-blooded animals (e.g. humans, rats, mice, guinea pigs, egrets, birds, higgies, pigs, pests, pomas, cats, animals Dogs, monkeys, chimpanzees, etc.).
本発明のリガンドの投与量は、 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、 低血糖の治療目的で本発明のリガンド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、 一日につき該リガンドを約 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、 該リガンドの 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、 低血糖の治療目的で本発明のリガンドを注射剤の形で成 人 (体重 60kgとして) に投与する場合、 一日につき該リガンドを約 0. 01〜30mg、 好ましくは約 0.:!〜 20mg、 より好ましくは約 0· l〜10mgを患部に注射することによ り投与するのが好都合である。 他の動物の場合も、 体重 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 ligand of the present invention varies depending on the target disease, the subject of administration, the administration route, and the like. For example, when the ligand of the present invention is orally administered for the treatment of hypoglycemia, it is generally required for an adult (body weight 60 kg). ), The ligand is 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 per day. When administered parenterally, the single dose of the ligand varies depending on the administration subject, target disease, and the like. For example, the ligand of the present invention is formed in the form of an injection for the treatment of hypoglycemia. When administered to a human (with a body weight of 60 kg), about 0.01 to 30 mg, preferably about 0.:! To 20 mg, more preferably about 0.1 to 10 mg of the ligand per day is injected into the affected area. It is more convenient to administer. In the case of other animals, the dose can be administered in terms of weight per 60 kg.
〔4 a〕 「本発明のリガンドの、 本発明の蛋白質に対する活性ィ匕作用を阻害す る化合物またはその塩を含有してなるインスリン抵抗性改善剤」 および 「本発 明のリガンドの、 本発明の蛋白質にに対する活性化作用を阻害する化合物また はその塩を含有してなる耐糖能異常、 糖尿病、 肥満症、 高脂血症、 動脈硬化、 高血圧症または心疾患の予防 ·治療剤」 [4a] “Insulin sensitizer comprising a compound that inhibits the activity of the ligand of the present invention on the protein of the present invention or a salt thereof” and “Insulin sensitizer of the present invention, Impaired glucose tolerance, diabetes, obesity, hyperlipidemia, arteriosclerosis, Prevention and treatment of hypertension or heart disease "
「本発明のリガンドの、 本発明の蛋白質に対する活性ィヒ作用を阻害する化合物 またはその塩」 としては、 本発明のリガンドの、 本発明の蛋白質に対する活性化 作用を阻害する化合物またはその塩であればいずれでもよく、 例えば、 ペプチド. 蛋白質、 抗体、 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、 血清などから選ばれる。 該化合物またはその塩を、 上記の剤として使用する場合は、 上記 〔1〕 と同様に使用すればよい。  The “compound or salt thereof that inhibits the activity of the ligand of the present invention on the protein of the present invention” is a compound or a salt thereof that inhibits the activating effect of the ligand of the present invention on the protein of the present invention. Any one may be used, for example, peptide, protein, antibody, non-peptide compound, synthetic compound, fermentation product, cell extract, plant extract, animal tissue extract, serum and the like. When the compound or a salt thereof is used as the above agent, it may be used in the same manner as in the above [1].
〔4 b〕 「本発明のリガンドの、 本発明の蛋白質に対する活性化作用を促進す る化合物またはその塩を含有してなる低血糖の予防 ·治療剤」 [4b] “A prophylactic / therapeutic agent for hypoglycemia comprising a compound or a salt thereof that promotes the activating effect of the ligand of the present invention on the protein of the present invention”
「本発明のリガンドの、 本発明の蛋白質に対する活性化作用を促進する化合物 またはその塩」 としては、 本発明のリガンドの、 本発明の蛋白質に対する活性化 作用を促進する化合物またはその塩であればいずれでもよく、 例えば、 ぺプチド. 蛋白質、 抗体、 非ぺプチド性化合物、 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、 血清などから選ばれる。 該化合物またはその塩を、 上記の剤として使用する場合は、 上記 〔1〕 と同様に使用すればよい。  The “compound or salt thereof that promotes the activating action of the ligand of the present invention on the protein of the present invention” includes a compound or a salt thereof that promotes the activating action of the ligand of the present invention on the protein of the present invention. Any of them may be used, for example, peptides. Proteins, antibodies, non-peptide compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, serum, and the like. When the compound or a salt thereof is used as the above agent, it may be used in the same manner as in the above [1].
本明細書および図面において、 塩基やアミノ酸などを略号で表示する場合、 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。  In the present specification and the drawings, bases, amino acids, and the like are indicated by abbreviations based on the abbreviations by the IUPAC-IUB Commission on Biochemical Nomenclature or conventional abbreviations in the relevant field. When there is an optical isomer of an amino acid, the L-form is indicated unless otherwise specified.
D N A デォキシリボ核酸  D N A Deoxyribonucleic acid
c D NA 相補的デォキシリボ核酸  c DNA Complementary deoxyribonucleic acid
A アデニン  A adenine
T チミン  T thymine
G グァニン  G Guanin
C シトシン  C cytosine
I :イノシン  I: Inosine
R :アデニン (A) またはグァニン (G)  R: Adenine (A) or Guanine (G)
Y :チミン (T) またはシトシン ( C ) M :アデ-ン (A) またはシトシン (C) Y: Thymine (T) or cytosine (C) M: Aden (A) or cytosine (C)
K : グァニン (G) またはチミン (T)  K: Guanine (G) or thymine (T)
S : グァ-ン (G) またはシトシン (C)  S: Guan (G) or cytosine (C)
W :アデ-ン (A) またはチミン (T)  W: Adene (A) or thymine (T)
B : グァニン (G) 、 グァニン (G) またはチミン (T) B: Guanine (G), Guanine (G) or Thymine (T)
D :了デニン (A) 、 グァニン (G) またはチミン (T)D: Ladenine (A), Guanine (G) or Thymine (T)
V :アデ二ン (A) 、 グァニン (G) またはシトシン (C)V: Adenine (A), guanine (G) or cytosine (C)
N :アデ-ン (A) 、 グァニン (G) 、 シトシン (C) もしく はチミン (T) または不明もしくは他の塩N: Aden (A), guanine (G), cytosine (C) or thymine (T) or unknown or other salt
RNA : リボ核酸 RNA: ribonucleic acid
mRNA :メッセンジャーリボ核酸  mRNA: messenger ribonucleic acid
d ATP  d ATP
:フォキンァ —リン酸  : Fokina-phosphoric acid
d TTP :デォキシチミジン三リン酸  d TTP: Deoxythymidine triphosphate
d GTP :デォキシグアノシン三リン酸  d GTP: Deoxyguanosine triphosphate
d CT P :デォキシシチジン三リン酸  d CT P: Deoxycytidine triphosphate
ATP :アデノシン三リン酸  ATP: Adenosine triphosphate
EDTA :エチレンジアミン四酢酸  EDTA: Ethylenediaminetetraacetic acid
SD S : ドデシル硫酸ナトリウム  SD S: Sodium dodecyl sulfate
BHA :ベンズヒ ドリノレアミン  BHA: Benzhi Dorinoleamine
pMBHA : p—メチノレべンズヒ ドリノレアミン  pMBHA: p-methinolebenzhydrinoleamine
T o s : p一トノレエンスノレフォニノレ  T o s: p-one
B z 1 :ベンジノレ  B z 1: Benginole
B om :ベンジノレオキシメチノレ  B om: Benzinoleoxymethinole
B o c : tーブチノレオキシカノレボニノレ  Boc: t-butynoleoxycanoleboninole
DCM : ジクロロメタン  DCM: dichloromethane
HOB t : 1ーヒ ドロキシベンズトリァゾール HOB t: 1-Hydroxybenztriazole
DCC : N, N,一ジシク口へキシルカルボジィミ ドDCC: N, N, hexyl carbyl imide
TF A : トリフルォロ酢酸 TF A: trifluoroacetic acid
D I E A : ジィソプロピルェチルァミン G 1 y又は G : グリシン ' DIEA: Disopropylethylamine G 1 y or G: Glycine ''
A 1 a又は A ァラニン A 1 a or A alanine
V a 1又は V パリン V a 1 or V Palin
L e u又は ロイシン  Leu or leucine
I 1 e又は I イソロイシン  I 1 e or I isoleucine
S e r又は S セリン  S e r or S serine
Th r又は T スレオニン Th r or T threonine
C y s又は C システィン Cys or C cysteine
Me t又は M メチォニン Me t or M methionine
G 1 u又は E グノレタミン酸 G 1 u or E Gunoletamic acid
A s p又は D ァスパラギン酸 Asp or D aspartic acid
Ly s又は K リジン Lys or K lysine
A r g又は R アルギニン A r g or R Arginine
H i s又は HH is or H
11 8又は? フエニノレアラニン  11 8 or? Pheninoleanine
Ty r又は Y チロシン Ty r or Y tyrosine
T r p又は W トリプトファン Trp or W tryptophan
P r o又は P プロリン Pro or P proline
A s n又は N ァスパラギン Asn or Nasparagine
G 1 n又は Q グノレタミン G 1 n or Q gnoletamine
p G 1 u ピログノレタミン酸 p G 1 u Pyrognoletamic acid
Ty r (I) 3—ョードチロシン Ty r (I) 3—Edo tyrosine
DMF N, N—ジメチルホルムアミ ド DMF N, N-dimethylformamide
Fm o c N— 9—フノレオレニノレメ トキシカノレポ-ノレ Fm o c N— 9—Funoolereninoleme Toxicanorepo
T r t トリチル T r t Trityl
P b f 2, 2, 4, 6, 7—ペンタメチノレジヒ ドロべンゾ フランー 5—スノレホニノレ P b f 2,2,4,6,7-pentamethinolesig Drobenzo furan 5-snolehoninole
C 1 t 2—クロ口 トリチル  C 1 t 2—Black mouth Trityl
B u* t—ブチノレ M e t (O) :メチォニンスルフォキシド B u * t—Buchinore M et (O): methionine sulfoxide
本願明細書の配列表の配列番号は、 以下の配列を示す。  The sequence numbers in the sequence listing in the present specification indicate the following sequences.
〔配列番号: 1〕  [SEQ ID NO: 1]
ヒ ト TREM - 2のァミノ酸配列を示す。  The amino acid sequence of human TREM-2 is shown.
〔配列番号: 2〕  [SEQ ID NO: 2]
マウス TREM- 2のァミノ酸配列を示す。  2 shows the amino acid sequence of mouse TREM-2.
〔配列番号: 3〕  [SEQ ID NO: 3]
配列番号: 1で表されるアミノ酸配列を有するヒト TREM - 2をコードする DNAの 塩基配列を示す。  This shows the base sequence of DNA encoding human TREM-2 having the amino acid sequence represented by SEQ ID NO: 1.
〔配列番号: 4〕  [SEQ ID NO: 4]
配列番号: 2で表されるァミノ酸配列を有するマウス TREM - 2をコードする DNA の塩基配列を示す。  This shows the nucleotide sequence of DNA encoding mouse TREM-2 having the amino acid sequence represented by SEQ ID NO: 2.
〔配列番号: 5〕  [SEQ ID NO: 5]
参考例 1で用いられたプライマ一の塩基配列を示す。  1 shows the nucleotide sequence of a primer used in Reference Example 1.
〔配列番号: 6〕  [SEQ ID NO: 6]
参考例 1で用いられたプライマーの塩基配列を示す。  2 shows the nucleotide sequence of a primer used in Reference Example 1.
〔配列番号: 7〕  [SEQ ID NO: 7]
参考例 2で用いられたプライマーの塩基配列を示す。  7 shows the nucleotide sequence of a primer used in Reference Example 2.
〔配列番号: 8〕  [SEQ ID NO: 8]
参考例 2で用いられたプライマーの塩基配列を示す。  7 shows the nucleotide sequence of a primer used in Reference Example 2.
〔配列番号: 9〕  [SEQ ID NO: 9]
参考例 2および参考例 3で用いられたプライマーの塩基配列を示す。  The base sequences of the primers used in Reference Examples 2 and 3 are shown.
〔配列番号: 1 0〕  [SEQ ID NO: 10]
参考例 2および参考例 3で用いられたプライマーの塩基配列を示す。  The base sequences of the primers used in Reference Examples 2 and 3 are shown.
〔配列番号: 1 1〕  [SEQ ID NO: 11]
参考例 6で用いられたプライマ一の塩基配列を示す。  7 shows the nucleotide sequence of a primer used in Reference Example 6.
〔配列番号: 1 2〕  [SEQ ID NO: 1 2]
参考例 6で用いられたプライマーの塩基配列を示す。  7 shows the nucleotide sequence of a primer used in Reference Example 6.
〔配列番号: 1 3〕 参考例 6で用いられたプライマ一の塩基配列を示す。 [SEQ ID NO: 13] 7 shows the nucleotide sequence of a primer used in Reference Example 6.
〔配列番号: 1 4〕  [SEQ ID NO: 14]
参考例 6で用いられたプライマーの塩基配列を示す。  7 shows the nucleotide sequence of a primer used in Reference Example 6.
〔配列番号: 1 5〕  [SEQ ID NO: 15]
参考例 6で用いられたプライマーの塩基配列を示す。  7 shows the nucleotide sequence of a primer used in Reference Example 6.
〔配列番号: 1 6〕  [SEQ ID NO: 16]
参考例 6で用いられたプライマーの塩基配列を示す。 実 施 例  7 shows the nucleotide sequence of a primer used in Reference Example 6. Example
以下に参考例おょぴ実施例を挙げて本発明を更に具体的に説明するが、 本発明 はそれに限定されるものではない。  Hereinafter, the present invention will be described more specifically with reference to Reference Examples and Examples, but the present invention is not limited thereto.
発現変動遺伝子の検索 Search for genes with altered expression
KKAyマウス (14週齢、 ォス) およびコントロールとして用いた C57BL/6マウス (14週齢、 ォス) より副睾丸脂肪組織を摘出し、 IS0GEN試薬 (和光純薬) 中でホ モジナイゼーシヨンした後、 クロ口ホルム抽出、 イソプロパノール沈殿によりそ れぞれの組織より Total R Aを抽出し、 さらに Oligotex dT30 (タカラバイオ) に より poly (A)+ RNAを精製した。 これらの poly (A)+RNAを 2 gを出発材料として Reverse Transcriptase (Superscript RTII ; インビ卜ロジェン)により cDNAを合 成し、 PCR - Select cDNA Subtraction Kit (クロンテック)を用いて、 KKAyマウス 脂肪組織で発現量の上昇あるいは低下している cDNA群を選択的に PCR断片として 増幅した。 増幅された cDNA群はク口一ニングべクタ一 pT7Blue - Tにラィゲーショ ンした後、 Escherichia coli DH5 αを形質転換することによりクローニングした ( それぞれの挿入配列をベクター配列である M13 primer Ρ7 (配列番号: 5 ) およ び M13 primer P8 (配列番号: 6 ) (いずれも東洋紡) で増幅し、 得られた PCR断 片をガラススライド上にスポッティングしてマイクロアレイを作製した。 検出プ ローブとしてそれぞれのマウス脂肪組織由来の poly (A)+ RNAをランダムプライ ム法により Cy 5または Cy3で蛍光ラベルしたものを作製し、 これをマイクロアレイ に対し 15時間ハイブリダィズして ¾現量の差の顕著な遺伝子群を同定した。 コン ト口ールである C57BL/6マウス発現遺伝子と比較して KKAyにおいて 4倍以上の発現 上昇遺伝子として 29遺伝子、 一方で 4倍以下の発現低下遺伝子として 69遺伝子が 同定された。 マウス型 TREM- 2は発現上昇遺伝子群の中から 5. 32倍上昇する遺伝子 として同定された。 参考例 2 Epididymal adipose tissue was removed from KKA y mice (14 weeks old, os) and C57BL / 6 mice (14 weeks old, os) used as controls, and homogenized in IS0GEN reagent (Wako Pure Chemical Industries). After chilling, total RA was extracted from each tissue by black-mouth extraction and isopropanol precipitation, and poly (A) + RNA was further purified by Oligotex dT30 (Takara Bio). Using 2 g of these poly (A) + RNAs as starting materials, cDNA is synthesized by Reverse Transcriptase (Superscript RTII; Invitrogen), and KKA y mouse adipose tissue is synthesized using PCR-Select cDNA Subtraction Kit (Clontech). The cDNA group whose expression level was increased or decreased was selectively amplified as a PCR fragment. The amplified cDNA group was ligated to pT7Blue-T, and cloned by transforming Escherichia coli DH5α ( each insertion sequence was vector sequence M13 primer Ρ7 (SEQ ID NO: 7). : 5) and M13 primer P8 (SEQ ID NO: 6) (both from Toyobo) were amplified, and the PCR fragments obtained were spotted on glass slides to prepare microarrays. Poly (A) + RNA derived from adipose tissue was fluorescently labeled with Cy5 or Cy3 by the random prime method. The hybrids were hybridized for 15 hours to identify a gene group having a remarkable difference in the present amount. 29 gene as expression increased gene least four times in the KKA y, whereas 69 genes as 4 times the decreased expression gene were identified by comparison with C57BL / 6 mice expressed gene is a con preparative port Lumpur. Mouse TREM-2 was identified as a 5.32-fold upregulated gene from a group of upregulated genes. Reference example 2
TREM- 2の脂肪細胞からのクロ一ユング  Cloung from fat cells of TREM-2
TREM- 2の発現変動の確認および機能解析のための実験材料獲得のため TREM-2の 全長コ一ド領域 cDNAを、 ヒト脂肪細胞およびマウス 3T3- L1脂肪細胞 cDNAライブラ リーより PCRでクローニングした。  The TREM-2 full-length coding region cDNA was cloned from human adipocyte and mouse 3T3-L1 adipocyte cDNA libraries by PCR to confirm the expression fluctuation of TREM-2 and to obtain experimental materials for functional analysis.
ヒト TREM - 2のクローニングには次の配列をプライマーとして用いた。  The following sequences were used as primers for the cloning of human TREM-2.
5' -ATGGAGCCTCTCCGGCTGCTCATC-3 ' (配列番号: 7 )  5'-ATGGAGCCTCTCCGGCTGCTCATC-3 '(SEQ ID NO: 7)
5, -TCACGTGTCTCTCAGCCCTGGCAG-3 ' (配列番号: 8 )  5, -TCACGTGTCTCTCAGCCCTGGCAG-3 '(SEQ ID NO: 8)
反応は Advantage-2 cDNA PCR Kit (クロンテック) を用い、 98°C 20秒、 68°C 1分 30秒を 35サイクルで行った。  The reaction was performed using the Advantage-2 cDNA PCR Kit (Clontech) at 35 cycles of 98 ° C for 20 seconds and 68 ° C for 1 minute and 30 seconds.
マウス TREM-2のクローニングには次の配列をプライマーとして用いた。  For cloning of mouse TREM-2, the following sequences were used as primers.
5, - ATGGGACCTCTCCACCAGTTTCTCCTG - 3, (配列番号: 9 )  5,-ATGGGACCTCTCCACCAGTTTCTCCTG-3, (SEQ ID NO: 9)
5' -TCACGTACCTCCGGGTCCAGTGAG-3 ' (配列番号: 1 0 )  5'-TCACGTACCTCCGGGTCCAGTGAG-3 '(SEQ ID NO: 10)
反応は Pfu Turbo DNA polymerase (ストラタジーン) を用い、 95°C 20秒、 65°C 40秒、 72°C 1分を 35サイクルで行った。  The reaction was performed using Pfu Turbo DNA polymerase (Stratagene) at 35 cycles of 95 ° C for 20 seconds, 65 ° C for 40 seconds, and 72 ° C for 1 minute.
得られた cDNA断片はヒト TREM- 2についてはそのまま、 一方、 マウス TREM- 2につ いては TAKARA Ex-Taq (タカラバイオ) で 72°C、 10分間反応して PCR断片の両側の 3 ' -末端に Aを付加した後、 それぞれ pCR2. 1ベクター (インビトロジェン) にク ローニングして DNA sequencingを行った。 ヒト脂肪細胞より単離した TREM-2は公 知のヒト TREM-2 (AF213457) と一致した。 一方、 マウス TREM- 2は公知配列として TREM- 2a (AY024348) 、 TREM- 2b (AY024349) および TREM_2c (AF213458) の 3種類 の cDNAが存在するが、 3T3- L1脂肪細胞より得られた TREM-2は 6クローンのうちす ベてが TREM - 2aと一致した。 参考例 3 The obtained cDNA fragment was used as is for human TREM-2, while mouse TREM-2 was reacted with TAKARA Ex-Taq (Takara Bio) at 72 ° C for 10 minutes and the 3'- After adding A to the end, each was cloned into pCR2.1 vector (Invitrogen) to perform DNA sequencing. TREM-2 isolated from human adipocytes was consistent with known human TREM-2 (AF213457). On the other hand, mouse TREM-2 has three known cDNA sequences, TREM-2a (AY024348), TREM-2b (AY024349), and TREM_2c (AF213458), and TREM-2 obtained from 3T3-L1 adipocytes. All of the 6 clones were consistent with TREM-2a. Reference example 3
RT - PCRによる発現変動の確認  Confirmation of expression fluctuation by RT-PCR
KKAyマウス (14週齢、 ォス) および C57BL/6マウス (14週齢、 ォス) の副睾丸 脂肪組織より抽出した RNAに対して RT- PCRを行つた。 それぞれの組織より抽出し た 0. 5 i gの Total RNAについて、 01igo-dT-アダプタープライマー (タカラバイ ォ) を用いて AMV (Avian Myeloblastosis Virus)由来逆転写酵素 (タカラパイ ォ) で cDNAを合成し、 マウス TREM- 2クローユングのためのプライマー (配列番 号: 9および配列番号: 1 0 ) を用いて PCRを行った。 反応は Advan1:age_2 cDNA PCR Kit (クロンテック) を用い、 98°C 20秒、 68°C 1分 30秒を 25サイクルで行つ た。 反応産物をァガロースゲル電気泳動で検出したところ、 参考例 1のマイクロ アレイの結果と同様に、 KKAyマウス脂肪組織の TREM-2発現量は、 C57BL/6マウス の TREM- 2発現量と比較して、 5倍以上の差が確認できた。 参考例 4 RT-PCR was performed on RNA extracted from epididymal adipose tissue of KKA y mice (14 weeks old, os) and C57BL / 6 mice (14 weeks old, os). 0.5 ig of total RNA extracted from each tissue was synthesized with AMV (Avian Myeloblastosis Virus) reverse transcriptase (TakaraPio) using 01igo-dT-adapter primer (TakaraVio) and cDNA was synthesized. PCR was performed using primers (SEQ ID NO: 9 and SEQ ID NO: 10) for TREM-2 closing. The reaction was performed using Advan1: age_2 cDNA PCR Kit (Clontech) at 98 ° C for 20 seconds and 68 ° C for 1 minute and 30 seconds in 25 cycles. When the reaction products were detected by agarose gel electrophoresis, the expression level of TREM-2 in the adipose tissue of the KKA y mouse was similar to the expression level of TREM-2 in the C57BL / 6 mouse, similar to the result of the microarray in Reference Example 1. A difference of more than 5 times could be confirmed. Reference example 4
抗マウス TREM- 2抗体の作製とマウス脂肪組織における蛋白質レベルでの TREM- 2 の発現解析  Production of anti-mouse TREM-2 antibody and expression analysis of TREM-2 at the protein level in mouse adipose tissue
マウス TREM- 2配列 〔配列番号: 2〕 の 133番目の Leu〜147番目の Serで構成され るぺプチドを抗原としてゥサギを免疫し、 抗 TREM- 2ポリクローナル抗体を取得し た。 KKAyマウスならびに C57BL/6マウスの副睾丸脂肪組織を摘出しホモジナイズ したもの各20 μ §をこの抗体を用いてウェスタンプロッティングで解析したとこ ろ、 KKAyマウス由来の組織からは TREM-2のバンドが検出されたが、 C57BL/6マウ ス由来の組織からは検出されなかった。 A heron was immunized with a peptide composed of the 133rd Leu to the 147th Ser of the mouse TREM-2 sequence [SEQ ID NO: 2] as an antigen to obtain an anti-TREM-2 polyclonal antibody. KKA y mice and C57BL / 6 mice epididymis adipose tissue using excised each 2 0 mu § The antibody obtained by homogenizing the Toko filtrate was analyzed by Western plotting, TREM-2 from the KKA y mice tissues Band was detected, but not detected in the tissue derived from C57BL / 6 mouse.
参考例 5 Reference Example 5
TREM- 2のマウスにおける発現組織分布  Tissue distribution of TREM-2 in mice
KKAyマウスならびに C57BL/6マウスの副睾丸脂肪組織、 腸間膜脂肪組織、 骨格 筋、 肝臓、 精巣、 脾臓、 脳、 腎臓からそれぞれ total RNAを抽出し、 参考例 3に 記載の RT- PCR法で TREM-2の発現組織分布を検討したところ KKAyマゥスの副睾丸脂 肪組織ならびに腸間膜脂肪組織で顕著な発現が認められた。 一方、 これら脂肪組 織以外の組織での発現はほとんど認められなかつた。 参考例 6 Total RNA was extracted from epididymal adipose tissue, mesenteric adipose tissue, skeletal muscle, liver, testis, spleen, brain, and kidney of KKA y mice and C57BL / 6 mice, respectively, and the RT-PCR method described in Reference Example 3 was used. Examination of the distribution of TREM-2 expression tissues revealed that KKA y mice had remarkable expression in epididymal fat tissue and mesenteric fat tissue. Meanwhile, these fat groups Almost no expression was observed in tissues other than tissues. Reference example 6
TREM-2の糖尿病モデルマウスにおける発現の定量  Quantification of TREM-2 expression in diabetic model mice
( 1 ) 定量的 RT- PCR法  (1) Quantitative RT-PCR method
Total RNA 1 ngあたりの TREM- 2 mRNAのコピー数を定量的 RT- PCR法で測定した c RT-PCRは SYBR Green RT-PCR試薬キット (アプライドバイオシステムズ) を用い て添付プロトコールに従って行い、 PCRプロダクト自動検出 ·定量システム ABI PRISM 7700 (アプライドバイオシステムズ) で定量した。 The copy number of TREM-2 mRNA per 1 ng of total RNA was measured by quantitative RT-PCR. C RT-PCR was performed using SYBR Green RT-PCR reagent kit (Applied Biosystems) according to the attached protocol. Automatic detection · Quantification system Quantification was performed using ABI PRISM 7700 (Applied Biosystems).
マウス TREM- 2の定量的 RT-PCRには、 以下のプライマーを用いた。  The following primers were used for quantitative RT-PCR of mouse TREM-2.
5' - ACACCCTTGCTGGAACCGTCAC-3' (配列番号: 1 1 )  5 '-ACACCCTTGCTGGAACCGTCAC-3' (SEQ ID NO: 1 1)
5' - GTCCTCCAGCACCTCCACCAGTA- 3, (配列番号: 1 2 )  5 '-GTCCTCCAGCACCTCCACCAGTA-3, (SEQ ID NO: 12)
ヒト TREM- 2の定量的 RT- PCRには、 以下のプライマーを用いた。  The following primers were used for quantitative RT-PCR of human TREM-2.
5' -GAGTCTGAGAGCTTCGAGGATG-3' (配列番号: 1 3 )  5'-GAGTCTGAGAGCTTCGAGGATG-3 '(SEQ ID NO: 13)
5' -CTGGCTGCTAGAATCTTGATGA-3' (配列番号: 1 4 ) 、  5'-CTGGCTGCTAGAATCTTGATGA-3 '(SEQ ID NO: 14),
マウス TNF- αの定量的 RT- PCRには、 以下のプライマーを用いた。  The following primers were used for quantitative RT-PCR of mouse TNF-α.
5' -AAGGGATGAGAAGTTCCCAAA-3 ' (配列番号: 1 5 )  5'-AAGGGATGAGAAGTTCCCAAA-3 '(SEQ ID NO: 15)
5' - CTCCACTTGGTGGTTTGCTAC-3 ' (配列番号: 1 6 )  5'-CTCCACTTGGTGGTTTGCTAC-3 '(SEQ ID NO: 16)
( 2 ) 糖尿病動物モデルにおける糖尿病病態の進行と TREM- 2の発現の関連 参考例 6 ( 1 ) に記載の定量的 RT- PCR法で TREM-2 mRNAの発現量を定量したと ころ、 KKAyマウスの副睾丸脂肪組織おょぴ腸間膜脂肪組織では C57BL/6マウスと 比較して 10倍以上の発現量亢進が認められた。 さらに、 7、 14および 28週齢の KKAyマウス副睾丸脂肪組織の TREM-2 mRNA発現量を同様の方法で測定したところ、 週齢に伴う血糖値の上昇と比例して発現量が増大した。 このとき、 インスリン抵 抗性の指標である TNF - aの mRNA発現量は TREM- 2と同様に増大し、 TREM - 2と TNF - a の発現量の相関係数 (R2値) は 0. 8802であった。 (2) Relationship between TREM-2 expression and progression of diabetic pathology in a diabetic animal model When the quantitative expression of TREM-2 mRNA was quantified by the quantitative RT-PCR method described in Reference Example 6 (1), KKA y In the epididymal adipose tissue and mesenteric adipose tissue of the mouse, the expression level was increased 10 times or more compared to the C57BL / 6 mouse. Furthermore, TREM-2 mRNA expression in epididymal adipose tissue of KKA y mice at 7, 14, and 28 weeks of age was measured in the same manner, and the expression increased in proportion to the increase in blood glucose with age. . At this time, TNF is an indicator of insulin resistance properties - mRNA expression level of a is increased similarly to TREM- 2, TREM - 2 and TNF - correlation coefficient of the expression of a (R 2 value) 0. 8802.
—方、 11、 20、 40週齢の db/dbマウスの副睾丸脂肪組織で TREM-2 mRNAの発現を 上記と同様の手法で定量した場合も KKAyマウスと同様に、 週齢に伴う血糖値の上 昇と比例して発現が増大した。 参考例 7 -When TREM-2 mRNA expression in epididymal adipose tissue of 11, 20, and 40-week-old db / db mice was quantified by the same method as above, blood glucose with age was similar to that of KKA y mice. Expression increased in proportion to increasing values. Reference Example 7
インスリン抵抗性状態での 3T3- L1脂肪細胞における TREM- 2の発現解析  Expression analysis of TREM-2 in 3T3-L1 adipocytes in insulin resistance state
3T3- L1脂肪細胞に持続的にィンスリンを作用させることにより実験的に組織培 養レベルでのィンスリン抵抗性状態を作り出すことができる (Diabetologia, 38 卷, 1148- 1156頁, 1995年; J. Biol. Chem. , 272卷, 7759-7764頁, 1997年)。 こ の手法でィンスリン抵抗性状態にした 3T3-L1細胞での TREM- 2発現量を参考例 6 ( 1 ) に記載の定量的 RT-PCR法で測定した。  By continuously insulin acting on 3T3-L1 adipocytes, an insulin-resistant state at the tissue culture level can be experimentally created (Diabetologia, 38, 1148-1156, 1995; J. Biol. Chem., 272, 7759-7764, 1997). The expression level of TREM-2 in 3T3-L1 cells, which had been made insulin-resistant by this method, was measured by the quantitative RT-PCR method described in Reference Example 6 (1).
3Τ3- L1脂肪細胞にインスリンを 100 ηΜで 48時間添加した場合に TREM- 2 mRNA発 現量が 3倍以上、 またィンスリンを 2 Mで 48時間添力卩した場合は TREM-2 mRNA発現 量力 S6倍以上増加した。 参考例 8  3Τ3- TREM-2 mRNA expression more than 3 times when insulin is added to L1 adipocytes at 100 ηΜ for 48 hours, and TREM-2 mRNA expression capacity when insulin is added at 2 M for 48 hours S6 More than doubled. Reference Example 8
正常ヒト組織における TREM- 2の発現解析  Expression analysis of TREM-2 in normal human tissues
正常ヒト由来の脳、 大腸、 心臓、 腎臓、 白血球、 肝臓、 肺、 卵巣、 膝臓、 前立 腺、 胎盤、 骨格筋、 小腸、 脾臓、 精巣および胸腺で TREM-2 mRNAの発現を参考例 2に記載の cDNAクローニングに用いたプライマーで参考例 3に記載の方法に準じ て RT- PCRを行った。  See TREM-2 mRNA expression in brain, colon, heart, kidney, leukocyte, liver, lung, ovary, knee, prostate, placenta, skeletal muscle, small intestine, spleen, testis and thymus from normal humans Reference Example 2 RT-PCR was performed according to the method described in Reference Example 3 using the primers used for the cDNA cloning described in (1).
いずれの組織でも TREM-2 mRNAの発現はほとんど確認できなかった。 参考例 9  In all tissues, TREM-2 mRNA expression was hardly confirmed. Reference Example 9
ヒト糖尿病患者脂肪組織における TREM - 2の発現解析  Expression analysis of TREM-2 in adipose tissue of human diabetic patients
ヒト糖尿病患者由来皮下脂肪組織よ.り total RNAを抽出し、 これに含まれる TREM-2 mRNA量を、 非糖尿病患者由来のものと参考例 6 ( 1 ) 記載の定量的 RT - PCR法で比較した。  Total RNA was extracted from subcutaneous adipose tissue from a human diabetic patient, and the amount of TREM-2 mRNA contained in the total RNA was compared with that from a non-diabetic patient by the quantitative RT-PCR method described in Reference Example 6 (1). did.
非糖尿病患者 6例では、 いずれもほとんど TREM- 2の発現が認められなかったが, 糖尿病患者では 8例中 3例で非糖尿病患者と比較して 10倍以上の顕著な TREM-2の発 現亢進が認められた。 参考例 1 0 In 6 non-diabetic patients, almost no TREM-2 expression was observed, but in 3 out of 8 diabetic patients, TREM-2 expression was more than 10 times higher than that in non-diabetic patients. Enhancement was noted. Reference example 10
TREM-2細胞外ドメィンの糖尿病モデル動物における血糖低下作用の検討  Investigation of the hypoglycemic effect of TREM-2 extracellular domain in diabetes model animals
TREM-2細胞外ドメイン (以下 Sol TREM-2と略記) が TREM2リガンドを中和する ことによる TREM- 2の機能抑制検討のため、 KKAyマウスへの投与実験を行った。 In order to examine the suppression of TREM-2 function by TREM-2 extracellular domain (hereinafter abbreviated as Sol TREM-2) neutralizing TREM2 ligand, administration experiments to KKA y mice were performed.
Sol TREM-2はマウス TREM- 2配列 (配列番号: 2 ) のうち細胞外ドメィンに相当 する 18番目の Ala〜164番目の Gluの 147ァミノ酸からなるポリぺプチドの N -末端に- 組換え蛋白質発現ベクター構築の際に付カ卩された 4アミノ酸 (Gly- Ser-His- Met) を持つものを大腸菌組換え型蛋白質として調製した。 KKAyマウス (14週齢、 ォ ス) に対し、 糖負荷試験の 3日前、 2日前、 1日前および糖負荷試験当日の 4回、 そ れぞれ lOO ^u gずつ、 合計 400 z gの Sol TREM-2を腹腔内投与した。 Sol TREM-2投与 後、 それぞれのマウスに 1 g/kgのブドウ糖を腹腔内投与することにより糖負荷を 行い、 糖負荷後 2時間までの血糖値を測定した。 Sol TREM-2 recombines at the N-terminus of the polypeptide consisting of 147 amino acids from Ala at position 18 to Glu at position 164, which corresponds to the extracellular domain in the mouse TREM-2 sequence (SEQ ID NO: 2). One having four amino acids (Gly-Ser-His-Met) added at the time of constructing the protein expression vector was prepared as an E. coli recombinant protein. A total of 400 zg Sol TREM of KKA y mice (14 weeks old, male) at lOO ^ ug 3 days before, 2 days before, 1 day before glucose tolerance test and 4 times each on the day of glucose tolerance test -2 was administered intraperitoneally. After administration of Sol TREM-2, glucose was loaded by intraperitoneally administering 1 g / kg of glucose to each mouse, and blood glucose levels were measured up to 2 hours after glucose loading.
Sol TREM- 2投与マウスはコントロールマウスと比較して糖負荷後 30分より顕著 な血糖値低下が認められた。 60分より 120分にかけて約 100 rag/dLの血糖値の低下 が認められた。 また、 この実験で糖負荷直後から 120分までの血糖値の変動曲線 下面積 (Area under glucose curve ; AUG) 値を比較した場合、 Sol TREM-2投与マ ウスはコント口ールマウスと比較して約 70%の有意な AUC値低下が認められた。  The mice treated with Sol TREM-2 showed a remarkable decrease in blood glucose 30 minutes after the glucose load compared to the control mice. A decrease in blood glucose of about 100 rag / dL was observed from 60 minutes to 120 minutes. In this experiment, when comparing the area under the glucose curve (AUG) from immediately after glucose load to 120 minutes, the mice administered Sol TREM-2 showed approximately a lower value than control mice. A significant decrease in AUC value of 70% was observed.
Sol TREM2投与後の副睾丸白色脂肪組織における TNF- αの mR A発現量は参考例 6 ( 1 ) に記載の定量的 RT- PCR法による測定の結果、 50%以下に低下していた。 さらに、 投与回数を、 糖負荷試験の 7日前から糖負荷試験当日までの 8回に増加 したことにより、 血糖低下作用はさらに顕著になり、 Sol TREM-2非投与群の空腹 時血糖値が約 250 mg/dLであったのに対し、 Sol TREM-2投与群の空腹時血糖値は 約 140 mg/dLとほぼ正常レベルにまで低下した。  The mRNA expression level of TNF-α in epididymal white adipose tissue after Sol TREM2 administration was reduced to 50% or less as a result of measurement by the quantitative RT-PCR method described in Reference Example 6 (1). Furthermore, by increasing the number of administrations to 8 times from 7 days before the glucose tolerance test to the day of the glucose tolerance test, the blood glucose lowering effect became more remarkable, and the fasting blood glucose level of the group without Sol TREM-2 was reduced to about In contrast to 250 mg / dL, the fasting blood glucose level of the Sol TREM-2 administration group decreased to almost a normal level of about 140 mg / dL.
このとき、 Sol TREM- 2投与群の血中インスリン値は、 Sol TREM- 2非投与群の 73%に低下した。 参考例 1 1  At this time, the blood insulin level in the group administered Sol TREM-2 was reduced to 73% of that in the group not administered Sol TREM-2. Reference example 1 1
TREM- 2細胞外ドメィンの糖尿病モデル動物における血中脂質低下作用の検討 参考例 1 0に記載の方法で KKAyマウスに Sol TREM2を 8回投与した後、 採血を行 .ぃ、 血中の脂質を測定した。 Investigation of blood lipid lowering effect of TREM-2 extracellular domain in diabetic model animals KKA y mice were administered Sol TREM2 eight times by the method described in Reference Example 10, and blood was collected. . ぃ, Blood lipids were measured.
Sol TREM- 2投与群の KKAyマウスでは、 Sol TREM- 2非投与群と比較して、 血中ト リグリセリ ドが 63%に、 血中遊離脂肪酸が 68%に低下した。 実施例 1 In the KKA y mice in the Sol TREM-2 administration group, the blood triglycerides and blood free fatty acids were reduced to 63% and 68%, respectively, as compared with the Sol TREM-2 non-administration group. Example 1
LPSと TREM - 2の結合試験およぴ GM3による競合阻害実験  Binding test between LPS and TREM-2 and competitive inhibition experiment with GM3
TREM- 2はマウス TREM- 2のァミノ酸配列 (配列番号: 2 ) のうち、 細胞外ドメイ ンに相当する 18番目の Ala〜164番目の Gluまでの 147ァミノ酸からなるポリぺプチ ドの N-末端に、 His- Tagを結合したものを、 大腸菌で T7プロモーターの制御下で 発現させた。 目的蛋白質は細胞破碎液をニッケルキレートカラム (ブアルマシ ァ) を用いて精製した (以下、 N - His - TREM2と略記する) 。  TREM-2 is the amino acid sequence of mouse TREM-2 (SEQ ID NO: 2), which is the polypeptide N consisting of 147 amino acids from Ala at the 18th position to Glu at the 164th position corresponding to the extracellular domain. -A His-Tag ligated end was expressed in E. coli under the control of the T7 promoter. The target protein was purified from the cell lysate using a nickel chelate column (Bulmacier) (hereinafter abbreviated as N-His-TREM2).
LPSは大腸菌由来 LPS (SIGMA L6529)を EZ- Link Biotin化試薬 (PIERCE) で Biotinラベルしたものを調整し、 用いた (以下 Biotin-LPSと略記する) 。  LPS was used after preparing Escherichia coli-derived LPS (SIGMA L6529) labeled with Biotin using EZ-Link Biotinization Reagent (PIERCE) (hereinafter abbreviated as Biotin-LPS).
GM3は Hytest社製 (#8G16-4h) を用いた。  GM3 used was Hytest (# 8G16-4h).
ニッケルキレートビーズ (Novagen) に 0. 36 μ gの N- His-TREM2を固定化したの ち、 Biotin- LPSを 0-50 μ g加えると濃度依存的に N- His- TREM2への結合が認められ た。 次に、 50 // gの Biotin_Lおに対し、 GM3を 50 §、 100 μ gおよび 300 /z g添加し、 4°Cでー晚反応させた。 ビーズを PBSで洗浄後、 グリシン塩酸緩衝液 (pH 2. 8) で 溶出し、 溶出液中の LPS含量を HRP標識ストレプトアビジンで、 また GM3含量を抗 GM3抗体 (生化学工業) でそれぞれ定量した。 50 gの LPSに対し、 GM3を 50 §、 100 μ gおよび 150 g添カ卩した場合の溶出液の LPS含量は、 GM3を添カ卩しない場合に 対し、 それぞれ約 60%、 30%および 20%に低下した。 一方、 溶出液の GM3含量は 492 nmの吸光度で約 0. 3、 1. 0および 1. 6と増加した。 なお、 GM3を添加しない場合 の 492 nmの吸光度は検出限界以下であった。 After immobilizing 0.36 μg of N-His-TREM2 on nickel chelate beads (Novagen), adding 0-50 μg of Biotin-LPS showed binding to N-His-TREM2 in a concentration-dependent manner. Was done. Next, 50 §, 100 μg, and 300 / zg of GM3 were added to 50 // g of Biotin_L, and reacted at 4 ° C. After washing the beads with PBS, the beads were eluted with glycine hydrochloride buffer (pH 2.8), and the LPS content in the eluate was quantified with HRP-labeled streptavidin, and the GM3 content was quantified with an anti-GM3 antibody (Seikagaku Corporation). . 50 to LPS of g, GM3 and 50 §, the LPS content of the eluate in the case of 100 mu g and 0.99 g添Ka卩, against when not添Ka卩the GM3, about 60%, respectively, 30% and 20 %. On the other hand, the GM3 content of the eluate increased to about 0.3, 1.0 and 1.6 at the absorbance at 492 nm. The absorbance at 492 nm when GM3 was not added was below the detection limit.
以上の結果より、 TREM - 2と LPSが結合すること、 TREM - 2と LPSの結合は GM3で阻 害されること、 および TREM-2と LPSの結合を阻害した GM3は LPSと競合的に TREM- 2 に結合していることが示された。 実施例 2 TREM-2と LPSの結合に対するガングリオシド (GMl, GM2, GM3, GD3) 、 リポテ ィコ酸、 デキストラン硫酸の競合阻害実験 These results indicate that TREM-2 and LPS bind, that TREM-2 and LPS binding are inhibited by GM3, and that GM3, which inhibits the binding of TREM-2 and LPS, competes with LPS for TREM- 2 was shown to be bound. Example 2 Competitive inhibition of gangliosides (GMl, GM2, GM3, GD3), lipoteichoic acid, and dextran sulfate for binding of TREM-2 and LPS
実施例 1に記載の方法で二ッケルキレートビーズに、 0. 36 μ gの N_Hi s- TREM2を 固定化したのち、 50 z gの Biotin- LPSに対し、 GM1、 GM2、 GM3および GD3を、 それ ぞれ lOO / g添加し、 溶出液中の LPS含量がガンダリオシド未添加の場合と比較し てどの程度に減少するかを検討した。  After immobilizing 0.36 μg of N_His-TREM2 on Nickel chelate beads by the method described in Example 1, GM1, GM2, GM3 and GD3 were added to 50 zg of Biotin-LPS. Each lOO / g was added, and it was examined how much the LPS content in the eluate was reduced as compared to the case where gandarioside was not added.
GM3添加の時の LPS結合量が非添加の約 30%であったのに対し、 GD3が約 38%、 GM1が約 35%、 GM2が約 20%と、 いずれも同程度の TREM2と LPSの結合阻害作用を示 した。 同様の実験系でそれぞれ 250 z gのデキストラン硫酸 (平均分子量 5, 000 ; SIGMA, D7037) 、 デキストラン硫酸 (平均分子量 500, 000 ; SIGMA, D6001) 、 お よぴリポティコ酸 (SIGMA, L2515) を添カ卩した場合も未添加と比較して、 溶出液 の LPS含量が、 それぞれ約 60%、 約 50%、 約 5%に低下した。  When GM3 was added, the amount of LPS bound was about 30% of that when no GM3 was added, whereas GD3 was about 38%, GM1 was about 35%, and GM2 was about 20%. It showed a binding inhibitory effect. In a similar experimental system, 250 zg of dextran sulfate (average molecular weight: 5,000; SIGMA, D7037), dextran sulfate (average molecular weight: 500,000; SIGMA, D6001), and lipotetic acid (SIGMA, L2515) were added. Even in the case of cultivation, the LPS content in the eluate was reduced to about 60%, about 50%, and about 5%, respectively, as compared to the case where no addition was made.
以上のことより、 TREM- 2と LPSの結合検出系を用いて TREM-2のァゴニストまた はアンタゴニストが検出できることが示された。 実施例 3  From the above, it was shown that the agonist or antagonist of TREM-2 can be detected using the binding detection system of TREM-2 and LPS. Example 3
GM3と TREM- 2の結合試験  GM3 and TREM-2 binding test
結合試験に用いた TREM2蛋白質はマウス TREM- 2のァミノ酸配列 (配列番号: 2 ) のうち、 細胞外ドメインに相当する 18番目の Alaから 164番目の Gluまでの 147 アミノ酸からなるポリペプチドの C -末端に、 ヒ ト免疫グロブリン Fcフラグメント を結合したものを、 動物細胞 (マウスミエローマ細胞) で CMVプロモーターの制 御下で分泌型蛋白質として発現させた。 目的蛋白質は培養液からプロテイン A力 ラム (フアルマシア) を用いて精製した (以下 mTREM2- Fcと略記する) 。  The TREM2 protein used in the binding test was a polypeptide of the 147 amino acids from the 18th Ala to the 164th Glu corresponding to the extracellular domain in the amino acid sequence of mouse TREM-2 (SEQ ID NO: 2). -A human immunoglobulin Fc fragment ligated to the end was expressed as a secreted protein in animal cells (mouse myeloma cells) under the control of the CMV promoter. The target protein was purified from the culture using Protein A column (Pharmacia) (hereinafter abbreviated as mTREM2-Fc).
GM3とし飞は、 0_ (acetamido-3, 5-dideoxy-D-glycero- -D-galacto-2- nonulopyranosylonic acid) - (2→3) - 0_ β -D-galactopyranosyl- (1→4) -0- β - D- glucopyranosyl- (1→ 1) - (2S, 3R, 4E) - 2 - N- (7 - nitrobenz - 2 - oxa - 1, 3 - diazol - 4「 yl) aminooctanamido-4-octadecene-l, 3- diol (株式会社ぺプチド研究所;以下、 蛍光ラベル GM3と略記する) を用いた。  GM3 and 0_ (acetamido-3, 5-dideoxy-D-glycero- -D-galacto-2-nonulopyranosylonic acid)-(2 → 3)-0_ β-D-galactopyranosyl- (1 → 4) -0 -β-D- glucopyranosyl- (1 → 1)-(2S, 3R, 4E)-2-N- (7-nitrobenz-2-oxa-1, 3-diazol-4 "yl) aminooctanamido-4-octadecene- l, 3-diol (Peptide Laboratories, Inc .; hereinafter abbreviated as fluorescent label GM3) was used.
100 mlの Protein G ァガロース (PIERCE社) に 10 gの mTREM2- Fcを添カロし、 4°Cで 3時間反応し固定ィヒした。 リン酸緩衝液 (PBS) で 3回洗浄し、 Protein Gに 結合していない mTREM2_Fcを除去した後、 蛍光ラベル GM3を 0. 2〜3. 2 g添カロし、 4°Cでー晚結合反応させた。 PBSで 4回洗浄し、 未反応の蛍光ラベル GM3を除去した 後、 グリシン塩酸緩衝液 (pH 2. 8) で Protein Gァガロースから mTREM2-Fcおよび 蛍光ラベル GM3を溶出した。 それぞれの溶出液について、 485 nmの励起光に対す る 535 nmの蛍光強度を測定し、 mTREM2 - Fcに結合した蛍光ラベル GM3の定量を行つ た。 なお、 Protein Gァガロースへの非特異的な GM3吸着量を測定するため、 mTREM2-Fcを固定化していない Protein Gァガロースに対し、 それぞれ同様の実験 を行った。 Add 100 g of Protein G agarose (PIERCE) with 10 g of mTREM2-Fc, The reaction was carried out at 4 ° C for 3 hours, and the mixture was fixed. After washing with phosphate buffer (PBS) three times to remove mTREM2_Fc not bound to Protein G, 0.2 to 3.2 g of fluorescent label GM3 was added, and the binding reaction was performed at 4 ° C. I let it. After washing with PBS four times to remove unreacted fluorescent label GM3, mTREM2-Fc and fluorescent label GM3 were eluted from Protein G agarose with glycine hydrochloride buffer (pH 2.8). For each eluate, the fluorescence intensity at 535 nm with respect to the excitation light at 485 nm was measured, and the fluorescent label GM3 bound to mTREM2-Fc was quantified. In addition, in order to measure the amount of nonspecific GM3 adsorption to Protein G agarose, the same experiment was performed for Protein G agarose on which mTREM2-Fc was not immobilized.
結果を図 1に示す。  The results are shown in Figure 1.
これより、 GM3は濃度依存的に TREM-2に結合することがわかった。 実施例 4  This indicates that GM3 binds to TREM-2 in a concentration-dependent manner. Example 4
TREM-2アンタゴニス 1、のスクリ一ニンク、、  Screening of TREM-2 Antagonis 1,
実施例 3に記載した TREM-2と GM3の結合試験を利用して一次スクリ一二ングを 行う。  Primary screening is performed using the binding test of TREM-2 and GM3 described in Example 3.
固定化した TREM2- Fcに対し、 GM3およぴ試験化合物を混在した試料を添加し、 PBSで洗浄後、 固定化された TREM-2に結合している GM3を蛍光強度で測定する。 GM3のみを添加した場合と比較して、 蛍光強度を低下せしめる試験化合物を選択 し、 TREM-2と GM3の結合を阻害する化合物として一次選択する (一次選択化合 物) 。 '  Add a sample mixed with GM3 and a test compound to the immobilized TREM2-Fc, wash with PBS, and measure the GM3 bound to the immobilized TREM-2 by fluorescence intensity. Select a test compound that reduces the fluorescence intensity as compared to the case where only GM3 is added, and select it as the primary compound that inhibits the binding between TREM-2 and GM3 (primary selection compound). '
一次選択化合物について、 DAP12のリン酸化を指標にしてアンタゴニスト活性 を保持する可能物を選択することにより、 二次選択を行う。 公知の手法で作製し た TREM-2およぴ V5タグ付き DAP12共発現 CH0細胞に、 GM3および試験化合物を添加 し、 10分間培養する。 細胞を細胞溶解液 [30 mM Tris-HCl (pH7. 4) , 150 mM NaCl, 10 mM EDTA, 1% NP-40, 50 mM NaF, 1 mM Na - Vanadate] で溶解し、 さら にソニケーションにより膜を破碑する。 細胞破碎液を抗 V5抗体で免疫沈降し、 免 疫沈降された試料をウェスタンプロット法で解析する。 まず抗 V5抗体 (インビト ロジェン) で試料中の DAP12総蛋白質をデンシトメ一ターで検出し、 一方、 抗リ ン酸化チロシン抗体 (シグマ) でリン酸化された DAP12を同様の手法で検出する c 両者を比較することにより DAP12蛋白質あたりのチロシンリン酸ィ匕の程度を検出 する。 試験化合物を添加せず GM3のみで細胞を刺激した場合の DAP12のリン酸化の 程度と、 試験化合物および GM3を添加した場合の DAP12のリン酸化の程度を比較し、 GM3による DAP12のリン酸化亢進を妨げる試験化合物をアンタゴニストとして二次 選択する。 For the primary selection compound, secondary selection is performed by selecting a substance that retains the antagonist activity using DAP12 phosphorylation as an index. GM3 and a test compound are added to TREM-2 and VAP-tagged DAP12 co-expressing CH0 cells prepared by a known method, and cultured for 10 minutes. Cells are lysed with a cell lysate [30 mM Tris-HCl (pH 7.4), 150 mM NaCl, 10 mM EDTA, 1% NP-40, 50 mM NaF, 1 mM Na-Vanadate], and then sonicated. Stele the membrane. The cell lysate is immunoprecipitated with an anti-V5 antibody, and the immunoprecipitated sample is analyzed by Western blotting. First, DAP12 total protein in the sample is detected with a densitometer using an anti-V5 antibody (invitrogen). C) Detect DAP12 phosphorylated with an oxidized tyrosine antibody (Sigma) by the same method. C) Compare the two to detect the degree of tyrosine phosphorylation per DAP12 protein. The degree of phosphorylation of DAP12 when cells were stimulated with GM3 alone without the addition of the test compound was compared with the degree of phosphorylation of DAP12 when the test compound and GM3 were added. The test compound that interferes is secondarily selected as an antagonist.
二次選択された試験化合物は KKAyマウス等の糖尿病モデルマゥスに投与し、 血 糖値、 耐糖能試験、 血中インスリン量、 血中脂質量等を測定しインスリン抵抗性 等が改善されるかを確認する。 実施例 5 The second-selected test compound is administered to a diabetes model mouse such as a KKA y mouse, and a blood glucose level, a glucose tolerance test, a blood insulin level, a blood lipid level, etc. are measured to determine whether insulin resistance is improved. Confirm. Example 5
ヒ ト TREM_2、 マウス TREM - 2、 ヒ ト TREM- 1およびマウス TREM- 1と GM3との結合試 藍  Human TREM_2, mouse TREM-2, human TREM-1 and mouse TREM-1 binding assay with GM3
結合試験に用いたヒ ト TREM - 2蛋白質はヒト TREM- 2のアミノ酸配列 (配列番号: 1 ) のうち、 細胞外ドメインに相当する 14番目の Gluから 167番目の Gluまでの 154 ァミノ酸からなるポリぺプチドの C-末端に、 ヒト免疫グロプリン Fcフラグメント を結合したものを、 動物細胞 (マウスミエローマ細胞) で CMVプロモーターの制 御下で分泌型蛋白質として発現させた。目的蛋白質は培養液からプロテイン Aカラ ム (フアルマシア社) を用いて精製した (以下 hTREM2- Fcと略記する) 。 マウス TREM- 2蛋白質は実施例 3に記載のものを使用した。  The human TREM-2 protein used in the binding test consists of 154 amino acids from the 14th Glu to the 167th Glu corresponding to the extracellular domain in the amino acid sequence of human TREM-2 (SEQ ID NO: 1). Human immunoglobulin Fc fragment conjugated to the C-terminus of the polypeptide was expressed as a secreted protein in animal cells (mouse myeloma cells) under the control of the CMV promoter. The target protein was purified from the culture using a protein A column (Pharmacia) (hereinafter abbreviated as hTREM2-Fc). The mouse TREM-2 protein used in Example 3 was used.
ヒト TREM-1蛋白質は、 Recombinant Human TREM- 1/Fc Chimera (R&Dシステム社 ;以下 hTREMl - Fcと略記する) 、 マウス TREM - 1蛋白質は、 Recombinant Mouse TRE -l/Fc Chimera (R&Dシステム社;以下 mTREMl- Fcと略記する) を用いた。  Human TREM-1 protein is Recombinant Human TREM-1 / Fc Chimera (R & D System; hereinafter abbreviated as hTREMl-Fc), and mouse TREM-1 protein is Recombinant Mouse TRE-l / Fc Chimera (R & D System; mTREMl-Fc).
ΙΟΟ μ Ιの Protein Gァガロース (PIERCE社) に、 それぞれ 3 /_t gの mTREMl_Fc、 raTREM2- Fc、 hTREMl- Fcおよび hTREM2_Fcを添カ卩し、 4°Cで 3時間反応し Protein Gァ ガロースにそれぞれの蛋白質を固定化した。リン酸緩衝液 (PBS) で Protein Gァ ガロースを 3回洗浄し、 Protein Gに結合していない蛋白質を除去した後、 実施例 3に記载の蛍光ラべル6¾13を0. 4〜1. 6 §添加し、 4°Cでー晚結合反応させた。 PBS で 4回洗浄し、 未反応の蛍光ラベル GM3を除去した後、 グリシン塩酸緩衝液 (pH 2. 8) で Protein Gァガロースから各蛋白質および蛍光ラベル GM3を溶出した。 そ れぞれの溶出液について 485 nmの励起光に対する 535 nmの蛍光強度を測定し、 各 蛋白質に結合した蛍光ラベル GM3の定量を行なった。 Add 3 / _t g of mTREMl_Fc, raTREM2-Fc, hTREMl-Fc and hTREM2_Fc to ΙΟΟμΙ of Protein G agarose (PIERCE) and react at 4 ° C for 3 hours to react with Protein G agarose, respectively. Was immobilized. After washing Protein G agarose three times with phosphate buffered saline (PBS) to remove proteins not bound to Protein G, the fluorescent label 6-13 described in Example 3 was replaced with 0.4-1. 6 § was added and allowed to undergo a 晚 -bonding reaction at 4 ° C. After washing four times with PBS to remove unreacted fluorescent label GM3, glycine hydrochloride buffer (pH 2. In step 8), each protein and the fluorescent label GM3 were eluted from Protein G agarose. For each eluate, the fluorescence intensity at 535 nm with respect to the excitation light at 485 nm was measured, and the fluorescent label GM3 bound to each protein was quantified.
結果を図 2に示す。  The result is shown in figure 2.
これにより GM3は TREM-2特異的に濃度に依存して結合することがわかる。 さら に、 マウス TREM- 2およぴヒト TREM- 2は、 ほぼ同等の GM3への結合力を有すること 力 sわ力る。 実施例 6 This indicates that GM3 specifically binds to TREM-2 in a concentration-dependent manner. In addition, mice TREM- 2 Oyopi person TREM- 2, it forces s Wachikararu having substantially binding force to the equivalent GM3. Example 6
TREM2_Fcおよび蛍光ラベル GM3複合体に対する、 非ラベル GM3による GM3置換実  GM3 substitution by unlabeled GM3 for TREM2_Fc and fluorescently labeled GM3 complex
100 1の Protein Gァガロース (PIERCE社) に 3 μ gの hTREM2- Fcを添加し、 4°C で 3時間反応し固定化した。リン酸緩衝液 (PBS) で 3回洗浄し、 Protein Gに結合 していない蛋白質を除去した後、 蛍光ラベル GM3を 1. 6 g添カ卩し、 4°Cで 3時間反 応させた。 リン酸緩衝液 (PBS) で 4回洗浄し未反応の蛍光ラベル GM3を除去した 後、 0. 8〜6. 4 /_^の6¾13 (HyTest社;以下非ラベル GM3と記載する) を添加し 4°Cで ー晚結合反応させた。 PBSで 4回洗浄し、.未反応の蛍光ラベル GM3および非ラベル GM3を除去した後、 グリシン塩酸緩衝液 (pH 2. 8) で Protein Gァガロースから hTREM2- Fc、 蛍光ラベル GM3およぴ非ラベル GM3を溶出した。 それぞれの溶出液に ついて 485 nraの励起光に対する 535 nmの蛍光強度を測定し、 hTREM2- Fcに結合し た蛍光ラベル GM3の定量を行なった。 100 μl of Protein G agarose (PIERCE) was added with 3 μg of hTREM2-Fc, and reacted at 4 ° C. for 3 hours for immobilization. After washing three times with a phosphate buffer (PBS) to remove proteins that were not bound to Protein G, 1.6 g of fluorescent label GM3 was added and reacted at 4 ° C for 3 hours. After washing with phosphate buffer (PBS) four times to remove unreacted fluorescent label GM3, add 0.8 to 6.4 / _ ^ of 6¾13 (HyTest; hereinafter referred to as unlabeled GM3). A binding reaction was performed at 4 ° C. After washing with PBS four times to remove unreacted fluorescent label GM3 and unlabeled GM3, hTREM2-Fc, fluorescent label GM3 and unlabeled from Protein G agarose with glycine HCl buffer (pH 2.8) GM3 eluted. For each eluate, the fluorescence intensity at 535 nm against 485 nra of excitation light was measured, and the fluorescent label GM3 bound to hTREM2-Fc was quantified.
非ラベル GM3を添カ卩しない実験の TREM - 2結合蛍光ラベル GM3量を 100%としたと き、 0. 8 gの非ラベル GM3を添カ卩した場合に約 78%、 1. 6 μ gの場合に約 57%、 6. 4 μ gの場合に約 33%に TREM-2結合蛍光ラベル GM3量がそれぞれ低下した。  When the amount of TREM-2 bound fluorescent label GM3 in the experiment without unlabeled GM3 was 100% and the amount of 0.8 g of unlabeled GM3 was added, about 78%, 1.6 μg The amount of the TREM-2-binding fluorescent label GM3 was reduced to about 57% in the case of, and to about 33% in the case of 6.4 μg.
これより、 この実験系の非ラベル GM3を試験化合物と置き換えて同様に実施す ることにより、 TREM-2と GM3の結合を阻害する化合物のスクリーニングが蛍光強 度の測定により簡便に行えることがわかる。 産業上の利用可能性 This indicates that screening for compounds that inhibit the binding of TREM-2 to GM3 can be easily performed by measuring the fluorescence intensity by replacing the unlabeled GM3 in this experimental system with the test compound and performing the same procedure. . Industrial applicability
(a) 配列番号: Γ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蛋白質もしくはその部分ぺプチドまたはその塩、 および (b) 複合糖質を用いる、 該蛋白質またはその塩と該複合糖質との結合性を変化 させる化合物またはその塩 (好ましくは、 該複合糖質の該蛋白質に対する活性化 作用を阻害する化合物またはその塩) は、 例えば、 インスリン抵抗性改善剤とし て、 さらには糖尿病、 肥満症、 高脂血症、 動脈硬化症、 高血圧症または心疾患の 予防 ·治療剤として有用である。 また、 該蛋白質 (例、 ヒ ト TREM- 2、 マウス TREM- 2など) および複合糖質 (好ましくはガンダリオシド、 さらに好ましくは GM3) は、 インスリン抵抗性改善作用、 糖尿病、 肥満症、 高脂血症、 動脈硬化症、 高血圧症または心疾患の予防 ·治療作用を有する化合物またはその塩のスクリー ユングにとって有用である。  (a) a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: も し く は or a partial peptide thereof or a salt thereof; and (b) a protein using a glycoconjugate Or a compound that changes the binding property between the salt and the complex saccharide or a salt thereof (preferably, a compound or a salt thereof that inhibits the activating action of the complex saccharide on the protein) is, for example, an insulin sensitizer. It is also useful as a prophylactic or therapeutic agent for diabetes, obesity, hyperlipidemia, arteriosclerosis, hypertension or heart disease. In addition, the protein (eg, human TREM-2, mouse TREM-2, etc.) and glycoconjugates (preferably, gandarioside, more preferably, GM3) have an effect of improving insulin resistance, diabetes, obesity, and hyperlipidemia. It is useful for the screening of a compound or a salt thereof which has a preventive / therapeutic action for arteriosclerosis, hypertension or heart disease.

Claims

'請求の範囲 'The scope of the claims
1 . (a) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一 のアミノ酸配列を含有する蛋白質もしくはその部分べプチドまたはその塩、 およ 5 び (b) 複合糖質を用いることを特徴とする、 該蛋白質またはその塩と該複合糖 質との結合性を変化させる化合物またはその塩のスクリーニング方法。 1. (a) using 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, and (b) using a glycoconjugate A method for screening a compound or a salt thereof, which alters the binding property between the protein or a salt thereof and the glycoconjugate.
2 . (a) 複合糖質を、 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蛋白質もしくはその部分ぺプチドまたは その塩に接触させた場合と、 (b) 複合糖質および試験化合物を、 該蛋白質もし 10 くはその部分ペプチドまたはその塩に接触させた場合における、 該複合糠質の該 蛋白質もしくはその部分べプチドまたはその塩に対する結合量を測定し、 比較す る、 請求項 1記載のスクリーニング方法。  2. (a) When the glycoconjugate is contacted with 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. (B) when the glycoconjugate and the test compound are brought into contact with the protein or its partial peptide or its salt, the binding amount of the complex bran to the protein or its partial peptide or its salt is determined. 2. The screening method according to claim 1, wherein the method is measured and compared.
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蛋白質もしくはその部分ぺプチドまたはその塩が、 該蛋白質 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 is obtained.
15 もしくはその部分ぺプチドまたはその塩をコードする D N Aを含有する形質転換 体を培養することによって細胞膜上に発現した蛋白質もしくはその部分べプチド またはその塩である請求項 1記載のスクリーユング方法。 2. The screening method according to claim 1, wherein the protein or a partial peptide or a salt thereof is a protein expressed on a cell membrane by culturing a transformant containing DNA encoding 15 or a partial peptide or a salt thereof.
4 . 複合糖質が、 標識した複合糖質である請求項 1記載のスクリーニング方法。 4. The screening method according to claim 1, wherein the glycoconjugate is a labeled glycoconjugate.
5 . (a) 複合糖質を、 配列番号: 1で表されるアミノ酸配列と同一もしくは 20 実質的に同一のアミノ酸配列を含有する蛋白質もしくはその部分べプチドまたは その塩に接触させた場合と、 (b) 複合糖質および試験化合物を、 該蛋白質もし くはその部分ぺプチドまたはその塩に接触させた場合における、 該蛋白質もしく ■ はその部分ペプチドまたはその塩を介した細胞刺激活性を測定し、 比較する、 請 求項 1記載のスクリーニング方法。 5. (a) contacting the glycoconjugate with a protein having the same or substantially 20 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; (B) measuring the cell stimulating activity of the glycoconjugate or the test compound via the protein or its partial peptide or its salt when the glycoconjugate or the test compound is brought into contact with the protein or its partial peptide or its salt; The screening method according to claim 1, wherein the screening method is performed.
25 6 . 複合糖質が、 ガンダリオシド、 シァリルォリゴ糖、 リポ多糖、 リポティコ 酸またはデキストラン硫酸である請求項 1〜 5記載のスクリーニング方法。 256. The screening method according to any one of claims 1 to 5, wherein the complex carbohydrate is gandarioside, sialyloligosaccharide, lipopolysaccharide, lipoticoic acid or dextran sulfate.
7 . 複合糖質が、 ガンダリオシドである請求項 1〜 5記載のスクリーニング方 法。  7. The screening method according to any one of claims 1 to 5, wherein the glycoconjugate is gandarioside.
8 . 複合糖質が、 GM 3である請求項 1〜 5記載のスクリーニング方法。 8. The screening method according to any one of claims 1 to 5, wherein the glycoconjugate is GM3.
9 . 配列番号: 1で表されるァミノ酸配列と実質的に同一のァミノ酸配列が、 配列番号: 2で表されるァミノ酸配列である請求項 1記載のスクリーニング方法。 9. The screening method according to claim 1, wherein the amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 1 is the amino acid sequence represented by SEQ ID NO: 2.
1 0 . (a) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同 一のアミノ酸配列を含有する蛋白質またはその塩および (b) 複合糖質を含有す ることを特徴とする、 該蛋白質またはその塩と該複合糖質との結合性を変化させ る化合物またはその塩のスクリーニング用キット。  10. (A) a protein or a salt thereof having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1, and (b) a complex saccharide A screening kit for a compound or a salt thereof that alters the binding property between the protein or a salt thereof and the glycoconjugate.
1 1 . 複合糖質の、 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蛋白質もしくはその部分べプチドまたはその 塩に対する活性化作用を阻害する化合物またはその塩を含有してなるインスリン 抵抗性改善剤。  11. A compound that inhibits the activating action of a glycoconjugate on 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 or a salt thereof, or An insulin sensitizer containing the salt thereof.
1 2 . 複合糖質の、 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蛋白質もしくはその部分ぺプチドまたはその 塩に対する活性化作用を阻害する化合物またはその塩を含有してなる耐糖能異常、 糖尿病、 肥満症、 高脂血症、 動脈硬化、 高血圧症または心疾患の予防 ·治療剤。  12. Compounds that inhibit the activating effect of glycoconjugates on proteins containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1, its partial peptides or salts thereof, or Prevention and treatment of impaired glucose tolerance, diabetes, obesity, hyperlipidemia, arteriosclerosis, hypertension or heart disease containing salts.
1 3 . 複合糖質の、 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 的に同一のァミノ酸配列を含有する蛋白質もしくはその部分べプチドまたはその 塩に対する活性化作用を促進する化合物またはその塩を含有してなる低血糖の予 防 ·治療剤。  13. A compound that promotes the activating action of a glycoconjugate on 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 An agent for preventing or treating hypoglycemia containing the salt.
1 4 . 複合糖質を含有してなる低血糖の予防 ·治療剤。  1 4. Prevention and treatment of hypoglycemia containing complex carbohydrates.
1 5 . 複合糖質が、 ガンダリオシド、 シァリルオリゴ糖、 リポ多糖、 リポティ コ酸またはデキストラン硫酸である請求項 1 4記載の予防 ·治療剤。  15. The prophylactic / therapeutic agent according to claim 14, wherein the complex carbohydrate is gandarioside, sialyl oligosaccharide, lipopolysaccharide, lipotic acid or dextran sulfate.
1 6 . (a) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同 一のァミノ酸配列を含有する蛋白質もレくはその部分べプチドまたはその塩、 お よび (b) 複合糖質を用いることを特徴とする、 インスリン抵抗性改善作用を有 する化合物またはその塩のスクリーニング方法。  16. (a) 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 or a salt thereof, and (b) a complex A method for screening a compound having an insulin resistance improving action or a salt thereof, which comprises using a saccharide.
1 7 . (a) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同 一のァミノ酸配列を含有する蛋白質もしくはその部分べプチドまたはその塩、 お ょぴ (b) 複合糖質を用いることを特徴とする、 耐糖能異常、 糖尿病、 肥満症、 高脂血症、 動脈硬化症、 高血圧症または心疾患の予防 ·治療作用を有する化合物 またはその塩のスクリーニング方法。 17. (a) 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; A compound having a preventive / therapeutic effect on impaired glucose tolerance, diabetes, obesity, hyperlipidemia, arteriosclerosis, hypertension or heart disease, characterized by using Or a method of screening for a salt thereof.
1 8 . 複合糖質の、 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 的に同一のァミノ酸配列を含有する蛋白質もしくはその部分ぺプチドまたはその 塩への活性化作用を阻害することを特徴とするインスリン抵抗性改善方法。  18. Inhibiting the activating action of glycoconjugates on proteins containing amino acid sequences identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 or partial peptides thereof or salts thereof A method for improving insulin resistance, comprising:
PCT/JP2004/014802 2003-10-03 2004-09-30 Method of screening insulin resistance-improving agent WO2005033700A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2003-346246 2003-10-03
JP2003346246 2003-10-03

Publications (1)

Publication Number Publication Date
WO2005033700A1 true WO2005033700A1 (en) 2005-04-14

Family

ID=34419501

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2004/014802 WO2005033700A1 (en) 2003-10-03 2004-09-30 Method of screening insulin resistance-improving agent

Country Status (1)

Country Link
WO (1) WO2005033700A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108196054A (en) * 2017-07-27 2018-06-22 数字本草中医药检测有限公司 A kind of test strips for detecting glycyrrhizic acid and its preparation method and application
WO2020194317A1 (en) * 2019-03-28 2020-10-01 Yeda Research And Development Co. Ltd. Method of treating lipid-related disorders

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH03170491A (en) * 1989-11-14 1991-07-24 Fidia Spa Improved ganglioside and its functional group derivative
JP2003503676A (en) * 1999-06-01 2003-01-28 プロティオーム・サイエンシィズ・ピーエルシー Methods and compositions relating to insulin resistance disease
WO2003099331A1 (en) * 2002-05-24 2003-12-04 Takeda Pharmaceutical Company Limited Insulin resistance improving agents

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH03170491A (en) * 1989-11-14 1991-07-24 Fidia Spa Improved ganglioside and its functional group derivative
JP2003503676A (en) * 1999-06-01 2003-01-28 プロティオーム・サイエンシィズ・ピーエルシー Methods and compositions relating to insulin resistance disease
WO2003099331A1 (en) * 2002-05-24 2003-12-04 Takeda Pharmaceutical Company Limited Insulin resistance improving agents

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SALTIEL ALAN R.: "New perspectives into the molecular pathogenesis and treatment of type 2 diabetes", CELL, vol. 104, 23 February 2001 (2001-02-23), pages 517 - 529, XP002904502 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108196054A (en) * 2017-07-27 2018-06-22 数字本草中医药检测有限公司 A kind of test strips for detecting glycyrrhizic acid and its preparation method and application
WO2020194317A1 (en) * 2019-03-28 2020-10-01 Yeda Research And Development Co. Ltd. Method of treating lipid-related disorders

Similar Documents

Publication Publication Date Title
EP1479768B1 (en) Novel screening method
WO2000024890A1 (en) Novel g protein-coupled receptor proteins, dnas thereof and ligands to the same
WO2002006483A1 (en) Novel physiologically active peptide and use thereof
CA2401290A1 (en) Novel g protein-coupled receptor protein and dna thereof
KR20010103766A (en) Novel g protein- coupled receptor protein and dna thereof
EP0845529A2 (en) Human G-protein coupled receptor protein cloned form fetal brain CDNA library
JPH08193099A (en) New g protein conjugated receptor protein, its production and use
EP1207201A1 (en) Novel g protein-coupled receptor protein and dna thereof
WO2005033700A1 (en) Method of screening insulin resistance-improving agent
EP1118620A1 (en) Novel g protein-coupled receptor protein and dna thereof
EP1698697A2 (en) SSD-containing protein, process for producing the same and use thereof
WO2000008053A1 (en) Novel g-protein coupled receptor protein and dna thereof
EP1559721B1 (en) Fprl1 ligands and use thereof
JPH08245697A (en) Novel g protein-conjugated receptor protein, its production and use
JP4188720B2 (en) New screening method
JP2005128010A (en) Method of screening insulin sensitizer
JP2004073182A (en) Insulin resistance-improving agent
EP1138693A1 (en) Novel g protein-coupled receptor protein and dna thereof
EP1126029A1 (en) Novel g protein-coupled receptor proteins and dnas thereof
EP1459755A1 (en) Body weight gain inhibitor
US20090227501A1 (en) Agents for preventing and/or treating upper digestive tract disorders
JP2003047472A (en) New g protein conjugated receptor protein and its dna
EP1207169A1 (en) Novel g protein-coupled receptor protein and dna thereof
JP2002223762A (en) New protein and dna encoding the same
US20040048263A1 (en) Novel g protein-coupled receptor protein and dna thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR 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 NA 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 PL 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