[go: up one dir, main page]
More Web Proxy on the site http://driver.im/

WO2004065633A1 - Method of using peptidyl arginine deiminase type iv - Google Patents

Method of using peptidyl arginine deiminase type iv Download PDF

Info

Publication number
WO2004065633A1
WO2004065633A1 PCT/JP2004/000430 JP2004000430W WO2004065633A1 WO 2004065633 A1 WO2004065633 A1 WO 2004065633A1 JP 2004000430 W JP2004000430 W JP 2004000430W WO 2004065633 A1 WO2004065633 A1 WO 2004065633A1
Authority
WO
WIPO (PCT)
Prior art keywords
gene
protein
padi4
expression level
test substance
Prior art date
Application number
PCT/JP2004/000430
Other languages
French (fr)
Japanese (ja)
Inventor
Kazuhiko Yamamoto
Ryo Yamada
Akari Horiguchi
Xiaotian Chang
Hidehiko Furukawa
Mitsuru Ono
Reimi Mikami
Makiko Nakayama
Masahiko Ohtsuki
Masakatsu Suzuki
Miyuki Nagasaki
Original Assignee
Riken
Sankyo 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 Riken, Sankyo Company, Limited filed Critical Riken
Priority to JP2005508091A priority Critical patent/JP4503531B2/en
Publication of WO2004065633A1 publication Critical patent/WO2004065633A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/78Hydrolases (3) acting on carbon to nitrogen bonds other than peptide bonds (3.5)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y305/00Hydrolases acting on carbon-nitrogen bonds, other than peptide bonds (3.5)
    • C12Y305/03Hydrolases acting on carbon-nitrogen bonds, other than peptide bonds (3.5) in linear amidines (3.5.3)
    • C12Y305/03015Protein-arginine deiminase (3.5.3.15)
    • 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
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/573Immunoassay; Biospecific binding assay; Materials therefor for enzymes or isoenzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/172Haplotypes

Definitions

  • the present invention relates to a method for evaluating the effect of a therapeutic agent for rheumatoid arthritis, and a method for predicting the risk of developing rheumatoid arthritis, using peptidylarginine diminase 'type IV.
  • RA Rheumatoid arthritis
  • RA Rheumatoid arthritis
  • RA pathologically begins with a specific immune response to an unknown antigen that has reached the joints, causing chronic inflammation and synovial proliferation by blood lymphocytes, macrophages, and neutrophils that have infiltrated the joints, Osteoclasts derived from inflammatory granulation tissue and bone and cartilage destruction by proteases occur.
  • RA pathologically begins with a specific immune response to an unknown antigen that has reached the joints, causing chronic inflammation and synovial proliferation by blood lymphocytes, macrophages, and neutrophils that have infiltrated the joints, Osteoclasts derived from inflammatory granulation tissue and bone and cartilage destruction by proteases occur.
  • RA RA-associated rheumatoid factor
  • anti-Sa protein antibody anti-perinuclear factor antibody
  • anti-keratin antibody anti-filaggrin antibody
  • anti-citrullinated peptide antibody and the like are known (Mart inus AM van Boekel). et al., Arthr it is Research (2002) 4 (2), p87-93)
  • Anti-citrullinated peptide antibody is detected in about 76% of sera of RA patients, but only about 4% is detected in sera of patients who show rheumatic-like symptoms but are not determined to be RA. It is an autoantibody (Gerard A.
  • Peptidyl citrulline is an amino acid produced by the conversion of arginine residues in proteins by peptidyl arginine diminase (hereinafter referred to as “PADI”).
  • PADI peptidyl arginine diminase
  • rodents type I is epidermis and uterus
  • type II is various tissues such as muscle, uterus, salivary glands, and knees
  • type II I is In epidermal and hair root cysts
  • type IV is expressed in various tissues (Ahied Abu Rus'd et al., European Journal of Biochemistry (1999) 259 (3), p660-669).
  • genes of type I, II, III, and V have the highest homology to rodent types I, II, III, and IV, respectively, and are registered on Gene Information Bank.
  • type I II (Takuya Kanno et al., Journal of Investigative Dermatology (2000) 115 (5), p813-823) and type V (Katsuhiko Nakashiia et al., Journal of Biological Chemistry (1999) ) Other than 274 (39), p27786-27792), no detailed report has been made.
  • Type III of human PADI is the same as that of rodents in hair root cysts (Takuya Kanno et al., Journal of Investigative Dermatology (2000) 115 (5), p813-823), and evening V is favorable (Hiroaki Asaga et al., Journal of Leukocyte Biology (2001) 70 (1), p46-51), respectively, but direct association between PADI and RA has been reported. Unknown.
  • type V human PADI is referred to as type IV in this specification.
  • a major factor that determines the constitution of an individual is a genetic polymorphism.
  • Recent advances in pharmacogenomics research have elucidated the relationship between genetic polymorphisms and the effects or side effects of drugs, and the effects or side effects of the drugs using genetic diagnosis It is becoming possible to predict before administration.
  • by examining the relationship between genetic polymorphisms and diseases it will be possible to make preliminary diagnoses and determine prognosis for some diseases.
  • An example of the former is a gene polymorphism of a drug metabolizing enzyme.
  • drug metabolizing enzymes whose activity is increased or decreased in association with polymorphism include cytochrome P4501A2, cytochrome P4502A6, cytochrome P4502C9, cytochrome P4502C19, cytochrome P4502D6, Cytochrome P450 2 E 1 and the like are known.
  • Genetic polymorphisms also occur in a group of enzymes called conjugating enzymes, such as thiopurine methyltransferase, N-acetyltransferase, UDP-dalcuronosyltransferase, and daryuthion S-transferase. It has been reported that the activity is reduced by polymorphism (Yusuke Nakamura, ed., “Strategy for SNP gene polymorphism”, (2000), Nakayama Shoten, Tokyo).
  • HLA as a causative gene for ulcerative colitis
  • TCR as a causative gene for rheumatoid arthritis
  • AP0E4 as a causative gene for Alzheimer's disease
  • Dopamine D3 receptor as a causative gene for schizophrenia
  • tributophan hydroxylase as a causative gene for manic depression
  • angiotensin precursor as a causative gene for albuminuria
  • myocardial infarction Blood coagulation factor VII is known as a causative gene for leukemia
  • lebutin is known as a causative gene for obesity (L. Kruglyak, Nature Genetics (1999) 22 (2), pl39-144).
  • TNF-a is thought to be an important substance involved in the pathogenesis of various inflammatory diseases. Recently, however, the expression of the TNF-h gene has occurred in the 5'-terminal upstream region of the TNF- ⁇ gene. (T. Higuchi et al., Tissue Antigens (1998) 51 (6), p605-612). Since these polymorphisms enhance the expression level of the TNF-a gene, they can be diagnostic markers for diseases involving TNF-0; such as juvenile rheumatoid arthritis, rheumatoid arthritis, and diabetes. It is believed that. Disclosure of the invention
  • the present invention relates to the effects of a therapeutic agent for rheumatoid arthritis or the onset of rheumatoid arthritis.
  • the purpose is to provide a new method for assessing risk.
  • the present inventors found that PADI4 protein and its mRNA are highly expressed specifically in the lesion site of RA patients, and that the expression of PADI4 Increased levels and contributed to RA development. Based on these findings, the present inventors considered that it is possible to evaluate a therapeutic agent for rheumatoid arthritis using the inhibitory effect on the expression level or activity of PADI4 protein or its mRNA as an index.
  • the present invention relates to a method for evaluating the effect of a test substance as a therapeutic agent for chronic rheumatoid arthritis, using as an index the inhibitory effect on the PADI4 gene or its orthologous gene, or the protein encoded by the gene.
  • the evaluation method of the present invention includes the following steps:
  • test substance as a therapeutic agent for rheumatoid arthritis is evaluated based on the difference in the expression amount under the conditions of administration or non-administration of the test substance.
  • the expression level of the gene can be determined, for example, by using a nucleic acid hybridization method, an RT-PCR method, a real-time PCR using a solid-phased sample selected from a gene chip, a cDNA array, and a membrane filter. , A subtraction method, a differential display method, a differential hybridization method, and a cross-hybridization method.
  • the evaluation method of the present invention comprises the following steps:
  • test substance as a therapeutic agent for rheumatoid arthritis is evaluated based on the difference in the expression amount under the conditions of administration or non-administration of the test substance.
  • the expression level of the protein is determined by, for example, Detected by one of the following methods: blot plot, slot blot, ELISA, and RIA.
  • the cells used in the evaluation method of the present invention are preferably blood-derived cells, synovial cells, spleen cells, and peritoneal cells.
  • the animal used in the evaluation method of the present invention is preferably a mouse.
  • the detection target is preferably the mouse ortholog of the PADI4 gene: the mouse PADI4 gene, or a protein encoded by the gene.
  • the evaluation method of the present invention comprises the following steps:
  • test substance as a therapeutic agent for rheumatoid arthritis is evaluated based on the difference in the above activities under the conditions of addition or non-addition of the test substance.
  • the activity of the protein is preferably measured by quantification of the reaction product using a fluorescence method or an absorption method.
  • the PADI4 protein is preferably a human-derived PADI4 protein.
  • the present inventors have found that there is a haplotype on the PADI4 gene that frequently appears in RA patients. They found that this haplotype encodes a mutant PAD 14 protein having a different amino acid sequence and enzyme activity from the normal PAD 14 protein, and that this difference is closely related to the onset of RA. Based on these findings, the present inventors thought that the use of the mutant PADI4 protein and its gene would enable prediction of the risk of developing RA and evaluation of the therapeutic effect on RA.
  • the present invention provides a method for chronically treating a subject who has provided a specimen based on the expression level of a mutant PADI4 protein having the amino acid sequence shown in SEQ ID NO: 21 or the gene (SEQ ID NO: 20) in the specimen.
  • a method for predicting the risk of developing rheumatoid arthritis is provided.
  • the prediction method of the present invention comprises the following steps:
  • the expression level of the gene may be determined, for example, by using a nucleic acid hybridization method using a solid-phased sample selected from a gene chip, a cDNA array, and a membrane fill sample, an RT-PCR method, a real-time PCR method, It is detected by any one of the subtraction method, the differential display method, the differential hybridization method, and the cross-hybridization method.
  • the prediction method of the invention comprises the following steps:
  • the expression level of the protein is detected by, for example, any one method selected from Western blotting, dot blotting, slot blotting, ELISA, and RIA.
  • the present invention provides a kit for predicting the risk of developing rheumatoid arthritis, comprising at least one selected from the group consisting of the following a) to e).
  • b) Mutant type consisting of the nucleotide sequence shown in SEQ ID NO: 20 A continuous polynucleotide probe of 20-1500 base length that specifically hybridizes to the PADI4 gene and detects the gene.
  • the present invention relates to a variant comprising the amino acid sequence of SEQ ID NO: 21.
  • the method includes, for example, the following steps:
  • test substance as a therapeutic agent for rheumatoid arthritis is evaluated based on the difference in the above activities under the conditions of addition or non-addition of the test substance.
  • the present invention provides a mutant PADI4 protein consisting of the amino acid sequence shown in SEQ ID NO: 21.
  • the protein is useful as a predictive marker for predicting the risk of developing rheumatoid arthritis.
  • ADVANTAGE OF THE INVENTION According to this invention, the simple screening of the therapeutic agent for rheumatoid arthritis is possible.
  • the risk of developing a subject with rheumatoid arthritis can be determined at an early stage by detecting an indicator of the expression level of the mutant PADI4 protein and the gene according to the present invention.
  • Figure 1 is a graph comparing the mRNA stability (resolution) of one haplotype of human PADI4: V9 and V18.
  • FIG. 2 is a graph showing (A) a result of PADI4 enzyme activity measurement by a fluorescence method, and (B) a graph showing a result of PADI4 enzyme activity measurement by an absorption method.
  • FIG. 3 is a graph showing the results of comparing the enzyme activities of two haplotypes of PADI4: V9 and V18 by an absorption method.
  • FIG. 4 is a photograph showing the results of In situ RT-PCR in which the expression of PADI4 mA in RA patient synovial tissues was observed. The two right ones in the figure are negative controls.
  • FIG. 5 is a graph showing the expression levels of mPADI4 gene in synovial tissue and spleen tissue in the mouse collagen-induced arthritis (CIA) group and the normal group.
  • Figure 6 shows (A) immunohistochemical staining of RA patient synovium with anti-PADI4 antibody, and (B) is a photograph showing immunohistochemical staining of the synovium of an OA patient using both an antibody that specifically recognizes PADI4 and an antibody that recognizes all subtypes of PADI.
  • Figure 7 shows (A) immunohistochemical staining of RA patient synovium using anti-PADI4 antibody and (B) immunohistochemical staining of RA patient synovium using anti-citrullinated protein antibody. It is a photograph shown.
  • FIG. 8 is a graph comparing the stability of human PADI4 V9 and PADI4 V18 mRNA. This description includes part or all of the contents as disclosed in the description of Japanese Patent Application No. 2003-12738 and Japanese Patent Application No. 2003-12774, which are the basis of the priority of the present application. BEST MODE FOR CARRYING OUT THE INVENTION
  • Peptidyl arginine diminase (hereinafter referred to as “PADI”) is an enzyme that converts arginine residues in a protein to citrulline to produce peptidyl citrulline.
  • PADI Peptidyl arginine diminase
  • the present inventors have confirmed that the PADI4 protein and the PADI4 gene (mRNA) are specifically highly expressed in the lesion tissue of RA patients, and that the citrullinated peptide is detected at a high level. Citrullinated peptides can cause RA lesions through the production of autoantibodies (anti-citrullinated peptide antibodies). Therefore, it is possible to evaluate the effect of a test substance as a therapeutic agent for rheumatoid arthritis using the inhibitory effect on the expression level and activity of the PADI4 protein or its gene as an index.
  • the genes used in the evaluation method of the present invention include not only the PADI4 gene but also its orthologous gene.
  • "Osolog gene” means "gene of common ancestry” Genes of different species having the same function and evolved from them ”.
  • orthologs of PADI4 include mouse PADI4.
  • the PADI4 gene and its ortholog gene are collectively referred to as “PADI4 gene of the present invention”.
  • the origin of the gene is not particularly limited, but is preferably derived from mammals, more preferably primates or rodents.
  • the most preferred examples include the human PADI4 gene and the mouse PADI4 gene.
  • the sequence of cDNA encoding human PADI4 is shown in SEQ ID NO: 16, but is not limited to this sequence, and as long as it encodes human PADI4, the gene is included in the human PADI4 gene.
  • the present inventors have confirmed that human PADI4 has a plurality of haplotypes, and these haplotypes are also included in the human PADI4 gene.
  • the sequence of the cDNA encoding mouse PADI4 is shown in SEQ ID NO: 18, but is not limited to this sequence. Is included in the mouse PADI4 gene.
  • the term “gene” includes not only DNA but also its mRNA, cDNA and cDNA. Therefore, the gene of the present invention includes all of these DNAs, mRNAs, cDNAs, and cDNAs.
  • all nucleotide sequences in the sequence listing are described as DNA sequences. When the sequence indicates RNA, the base symbol “t” in the sequence listing is replaced with “u”.
  • the protein (PADI4 protein) used in the evaluation method of the present invention includes, in addition to the PADI4 protein encoded by the PADI4 gene, the protein encoded by the orthologous gene.
  • PADI4 protein of the present invention includes, in addition to the PADI4 protein encoded by the PADI4 gene, the protein encoded by the orthologous gene.
  • these proteins are collectively referred to as “PADI4 protein of the present invention”.
  • the origin of the protein is not particularly limited, but is preferably derived from mammals, and more preferably derived from primates and rodents.
  • the most preferred examples of the present invention include human PADI4 protein and mouse PADI4 protein.
  • the amino acid sequence of human PADI4 is shown in SEQ ID NO: 17, but is not limited thereto, and is represented by a sequence in which one or several amino acids have been deleted, substituted or added. As long as the peptide also has a function as human PADI4, it is included in the human PADI4 protein.
  • mouse PADI4 amino acid sequence of mouse PADI4 is shown in SEQ ID NO: 18, but is not limited thereto, and the peptide is represented by a sequence in which one or several amino acids have been deleted, substituted or added in the sequence. As long as they also have a function as mouse PADI4, they are included in the mouse PADI4 protein.
  • the present inventors have found that there are a plurality of polymorphisms that frequently appear in patients with rheumatoid arthritis on the human PADI4 gene, and these are almost completely linked (98.9 to 100%). To make up one haplotype. Furthermore, it was confirmed that this haplotype encodes a mutant PADI4 protein having a different amino acid sequence and enzyme activity from a normal PADI4 protein, and that the difference is closely related to the onset of RA.
  • mutant PADI4 protein is a protein having the amino acid sequence represented by SEQ ID NO: 21, and the amino acid sequence of normal PADI4 (SEQ ID NO: 17) Amino acid 5 (substituted from Ser to G1y), Amino acid 82 (substituted from A1a to Va1), Amino acid 112 (substituted from A1a to G1y) Are different. Due to this difference, mutant PADI4 protein has higher enzymatic activity than normal PAD protein and enhances the production of citrullinated protein that contributes to RA lesions.
  • the “mutant PADI4 gene” has a cDNA sequence represented by SEQ ID NO: 20.
  • a normal PADI4 gene is defined as the third intron of the third intron in the genome sequence. 6th base (substitute cytosine for thymine), 2nd exon 7 1st base (substitute adenine for guanine), 2nd exon 15 3rd base (substitute cytosine for thymine), 3rd exon
  • the second base (substitution of cytosine to guanine) of the fourth and the ninth base of exon 4 substitution of cytosine to thymine: this substitution does not involve amino acid substitution) are different.
  • the mRNA of the mutant PADI4 gene is more stable than the mRNA of the normal PADI4 gene, and may increase the production of citrullinated proteins that contribute to RA lesions. 2 Methods for evaluating the effects of test substances as therapeutics for rheumatoid arthritis
  • the present invention provides a method for evaluating the effect of a test substance as a therapeutic agent for rheumatoid arthritis, using an inhibitory effect on the PADI4 gene of the present invention or the protein encoded by the gene as an index.
  • the above method may be a method for comparatively evaluating the inhibitory effect of one test substance under the administration (addition) and non-administration conditions, or a similar comparative evaluation of two or more test substances. You may. Alternatively, if a correlation between the expression level or activity of the gene or protein and the RA therapeutic effect is established empirically, an absolute evaluation without a comparative control may be performed based on the relationship.
  • the inhibitory effect on the PADI4 gene or PADI4 protein of the present invention may be evaluated using the expression level of the PADI4 gene or PADI4 protein of the present invention as an index, or the inhibitory effect of the PADI4 gene or PADI4 protein of the present invention.
  • Activity (including stability) may be evaluated as an index.
  • the evaluation system may be an in vivo system or an in vitro system.
  • the in vivo evaluation method of the present invention using the PADI4 gene expression level as an index includes, for example, the following steps.
  • Step 1 Animals are bred under the conditions of administration or non-administration of the test substance.
  • Step 2 detecting the expression level of the PADI4 gene of the present invention in the blood or cells of the above animal.
  • Step 3 Evaluate the effect of the test substance as a therapeutic agent for RA on the basis of the difference in the expression level under the conditions of administration or non-administration of the test substance.
  • Process 1 Animal rearing
  • the “animal” used in the evaluation method of the present invention is not particularly limited, but a model animal exhibiting RA pathology is preferable. Such animals may be commercially available or may be produced according to known methods.
  • Models exhibiting such RA pathology include, for example, spontaneous arthritis model, MRL mouse arthritis, NZB / KN mouse arthritis, SKG mouse arthritis; inducible arthritis model, rat adjuvant arthritis, rat mouse collagen Arthritis (CIA rat 'mouse), pristine arthritis, SCID cell-transplanted arthritis, SCID mouse tissue transplant arthritis; and HTLV-1 transgenic mice, IL-1 receptor Yuichi antagonist K0 mice, etc.
  • the animals are bred for an appropriate period under the conditions of administration or non-administration of the test substance.
  • the dose of the test substance to the animal is not particularly limited, and an appropriate dose may be set according to the properties of the test substance and the weight of the animal.
  • the method and period of administration of the test substance to the animal are also not particularly limited, and the administration route and the administration period may be appropriately set according to the properties of the test substance.
  • blood or cells are isolated from animals bred under the conditions of administration or non-administration of the test substance, and the expression level of the PADI4 gene of the present invention in the blood or cells is detected.
  • the method for detecting the PADI4 gene of the present invention includes, for example, a method of first extracting total RNA from isolated blood or cells and detecting the expression level of mRNA in the total RNA.
  • RNA extraction solvent examples include those containing a component having an action to inactivate liponucleases such as phenol (for example,
  • RNA thiocyanate / cesium chloride ultracentrifugation method guanidine thiocyanate / hot phenol method
  • guanidine hydrochloride method guanidine acid thiocyanate phenol / clonal form method
  • the guanidine acid thiocyanate / phenol / chloroform method is preferred.
  • the extracted total RNA may be further purified only to mRNA if necessary.
  • the purification method is not particularly limited, most mRNAs present in the cytoplasm of eukaryotic cells have a poly (A) sequence at the 3 ′ end, and this characteristic can be used, for example, as follows. can do. First, a biotinylated oligo (dT) probe is added to all the extracted RNA to adsorb poly (A) + RNA. Next, a paramagnetic particle carrier on which streptavidin is immobilized is added, and poly (A) + RNA is captured using the binding between biotin and streptavidin. After washing, poly (A) + RNA is eluted from the oligo (dT) probe.
  • a method of adsorbing poly (A) + RNA using an oligo (dT) cellulose column and eluting the same may be used for purification.
  • the eluted poly (A) + RNA may be further fractionated by sucrose density gradient centrifugation or the like.
  • the expression level of the PADI4 gene of the present invention in the total RNA under the conditions of administration or non-administration of the test substance is detected.
  • the gene expression level can be detected as signal intensity by preparing cRNA or cDNA from the obtained total RNA and labeling it with an appropriate labeling compound.
  • the method of detecting the expression level of the gene is described below in detail by dividing it into i) an analysis method using a solid-phased sample, ii) RT-PCR method (real-time PCR method), and iii) other analysis methods. . ii)) Analytical method using solid-phase sample preparation
  • Cc DD NNAA or cc RR NNAA (hereinafter referred to as "" “" ").
  • the mark labeling probe described above even with the mmRR NNAA chlorochloroform of PPAADDII44 of the present invention, labels all mmRR NNAA expressing and expressing. You may be aware of it. .
  • As a starting and starting material for making prop rooves it is possible to use mmRR NN AA, which may or may not be purified. However, it is better to use the poporily ((AA)) ++ RR NN AA purified and produced by the method described above. .
  • the analysis and analysis method using various types of test samples for solid-phase solidification are explained. .
  • the genetic gene chip used in the present invention is a method in which a genetic gene, which is an object to be detected and detected, is solid-phase-phased. If it is a thing that is sold, even if it is a thing that is marketed and sold, it is a publicly known method ((LLiippsshhuuttzz ,, RR .. JJ .. eett aall .. ((11999999)) NNaattuurree ggeenneett .. 2211 ,, ssuuppppll iimmeenntt , 2200--2244)). . Detection and detection by genetic genetic test can be performed in accordance with the conventional method. .
  • the protocol attached to the product may be used.
  • By detecting, analyzing and analyzing the emitted light it is possible to obtain the amount of expression of the genetic gene. .
  • the array or membrane fill used in the present invention may be a commercially available array (for example, Intelligin (Takara Shuzo), a filter microarray atlas system) as long as the gene to be detected is immobilized. (Clontech, etc.)) or may be produced based on a known method.
  • the gene to be immobilized is a cDNA or RT-PCR product cloned by performing a reverse transcriptase reaction or PCR with primers created based on sequence information such as Gen Bank. .
  • a radioisotope e.g., 32 P, 33 P
  • a labeled probe is prepared by the addition, and hybridization is performed by a conventional method.
  • detection is performed using an analyzer (for example, atlas image: manufactured by Clontech). Perform analysis.
  • RT PCR method (Real-time PCR method)
  • the RT-PCR method and one of them, the real-time PCR (TaciMan PCR) method are suitable for the evaluation method of the present invention in that they can detect minute amounts of DNA with high sensitivity and quantitatively.
  • the real-time PCR (TaqMan PCR) method an oligonucleotide probe that hybridizes to a specific region of the target gene and has a 5 'end labeled with a fluorescent dye (Repo Ichiichi) and a 3' end labeled with a fluorescent dye (Quenchia I) is used. used. In this probe, the fluorescence of the reporter is normally suppressed by quenching. With the fluorescent probe completely hybridized to the target gene, PCR is performed from outside using Tad DNA polymerase.
  • the exonuclease activity hydrolyzes the fluorescent probe from the 5 'end, releasing the repo dye overnight to emit fluorescence.
  • the real-time PCR method can accurately quantify the initial amount of type I DNA by monitoring the fluorescence intensity in real time.
  • methods for analyzing gene expression levels include, for example, the subtraction method (Sive, HL and John, T. St. (1988) Nucleic Acids Research 16, 10937, Wang, Z., and Brown, DD (1991) Proc. Natl. Acad. Sci. USA 88, 11505-11509), Differential 'Display Method (Liang, P., and Pardee, AB (1992) Science 257, 967-971, Liang, P., Averboukh, L., Keyomarsi, K., Sager, R., and Pardee, AB (1992) Cancer Research 52, 6966-6968), differential 'hybridization' Method (John, T.
  • a single-stranded cDNA was prepared from total RNA, and this was hybridized with all RNA obtained from another cell, and then a single-stranded cDNA was not hybridized with a hydroxyapatite column.
  • a method for isolating strand DNA and preparing a cDNA library from this cDNA (Bio Manual Series 3, Genetic Cloning Experiments, Yodosha (1993), Current 'Protocols in' Molecular ⁇ Biology) or a cDNA library was prepared first, and single-stranded DNA was prepared from this library using helper phage, etc., and biotin was added to this single-stranded DNA and all RNA obtained from another cell.
  • RNA or total RNA is purified for each sample under the administration or non-administration conditions of the test substance, the total RNA purified from the sample under the administration condition is converted into type II, and cDNA is synthesized using reverse transcriptase. I do.
  • CDNA can also be labeled by adding [a- 32 P] dNTP during synthesis.
  • the labeled cDNA and total RNA in the form of a ⁇ form a stable double-stranded DNA-RNA hybrid.
  • high-temperature treatment in the presence of heat degrades only RNA and generates single-stranded cDNA.
  • a double-stranded DNA-RNA hybrid is formed that is more stable than the nucleotide sequence complementarity. That is, the cDNA expressed under non-administration conditions forms a hybrid, but the cDNA expressed specifically as RNA under the administration conditions remains single-stranded.
  • the double-stranded DNA-RNA hybrid and the single-stranded cDNA are separated on a hydroxyapatite column, and only the single-stranded cDNA is purified. By repeating this step, cDNA specific for the target tissue can be concentrated.
  • the concentrated specific cDNA When the concentrated specific cDNA is labeled with a radioisotope or the like, it can be used as a probe for screening a cDNA library. This operation can also be performed using a commercially available kit (for example, PCR Select cDNA Subtraction Kit: manufactured by Clontech, etc.).
  • RNA or total RNA is extracted from two samples to be compared (in the case of the present invention, a sample under the administration or non-administration of the test substance), and this is converted into single-stranded cDNA using reverse transcriptase. Convert.
  • PCR is performed using the obtained single-stranded cDNA as a type II and using appropriate primers.
  • a random primer a primer of about 10 to 12 mers consisting of an arbitrary sequence
  • an uncard primer anchored primer
  • an arbitrary primer arbitrary primer
  • T oligo d
  • n 11 to 12
  • V guanine, adenine or cytosine
  • a gene that is specifically expressed in any of the specimens can be selected, and a cDNA fragment thereof can be isolated.
  • This method can also be performed using a commercially available kit (for example, RNA Image Kit 1, manufactured by Jenhan Yuichi).
  • the cDNA library one prepared from total RNA purified from the tissue of interest, and screened with 32 P-labeled c DNA probe synthesized from total RNA of eye tissue and a control tissue, and clones are selected that seen Haiburidizu the probe of target tissue Is the way.
  • a cDNA library is prepared from total RNA purified from a sample under non-administration conditions according to a conventional method, and two sets of replica filters are prepared from the library.
  • cDNA is synthesized with reverse transcriptase using the total RNA purified from the sample under the non-administration condition as type III.
  • [- 32 P] labeling c DNA by adding dNTPs are examples of the total RNA purified from the sample under the non-administration condition.
  • the labeled cDNA and total RNA that has become type II are degraded to single-stranded DNA by high-temperature treatment in the presence of alkali that forms a stable double-stranded DNA-RNA hybrid. Purify the cDNA. Similarly, a single-stranded cDNA is prepared in which all RNA purified from a sample under the test substance administration conditions is labeled with 32 P in a small form. Using both labeled cDNAs as probes, perform hybridization with a filter prepared from a sample under non-administration conditions.
  • a gene that is specifically expressed under the administration conditions of the test substance can be identified. Can be cloned.
  • Positive clones were obtained by hybridizing the cDNA library derived from either the test substance-administered or non-administered sample with low DNA stringency using appropriate DNA as a probe. obtain. Using the obtained positive clone as a probe, Northern hybridization is performed on all RNAs derived from each sample, and a clone expressing only one of them is selected.
  • the effect of the test substance as an RA therapeutic is evaluated based on the difference in the expression level of the PADI4 gene of the present invention between administration and non-administration of the test substance. That is, when the expression level of the PADI4 gene of the present invention is significantly reduced under the administration condition of the test substance as compared with the non-administration condition, it can be evaluated that the test substance has an effect as a therapeutic agent for RA.
  • “significantly reduced” means that, for example, a statistically significant difference (p ⁇ 0.05) in the expression level of the PADI4 gene of the present invention under the administration and non-administration conditions of the test substance. It means there is.
  • the in vivo evaluation method using the expression level of the PADI4 protein of the present invention as an index includes, for example, the following steps.
  • Step 1 The animals are bred under the conditions of administration or non-administration of the test substance.
  • Step 2 The expression level of the PADI4 protein of the present invention in the blood or cells of the animal is detected using an antibody that specifically binds to the protein.
  • Step 3 The effect of the test substance as an RA therapeutic agent is evaluated based on the difference in the expression level between the administration and non-administration of the test substance.
  • Process 1 Animal rearing
  • Step 2 Detection of expression level of PADI4 protein of the present invention
  • the expression level of the PADI4 protein of the present invention in the blood or cells of the animal is detected using an antibody that specifically binds to the protein.
  • Methods for detecting proteins using antibodies are not particularly limited, but include Western blot, dot blot, slot blot, ELISA, and RIA.
  • the method is any one selected from the following methods.
  • these detection methods will be described specifically from sample preparation to detection.
  • blood or cells in which the PADI4 protein of the present invention is highly expressed are preferable, and therefore, blood-derived cells such as neutrophils and eosinophils, synovial cells, spleen cells, and intraperitoneal cells are preferable.
  • blood or cells used as a cell extract
  • ELISA / RIA samples or Western blot samples as follows. To be prepared.
  • the collected serum may be used as it is, or may be appropriately diluted with a buffer.
  • Western blot for electrophoresis
  • a cell extract as it is or dilute appropriately with a buffer to prepare a sample buffer containing 2_mercatorethanol for SDS-polyacrylamide gel electrophoresis ( Sigma).
  • a sample buffer containing 2_mercatorethanol for SDS-polyacrylamide gel electrophoresis Sigma.
  • a sample for dot / slot plotting for example, a recovered cell extract itself or a sample appropriately diluted with a buffer is directly adsorbed to a membrane using a blotting apparatus.
  • polypeptide in a sample containing the PADI4 protein of the present invention is immobilized on a membrane or the bottom surface of a well of a 96-well plate.
  • the membrane to be used include a nitrocellulose membrane (for example, manufactured by Bio-Rad), a nylon membrane (for example, Hybond-ECL (manufactured by Amersham-Pharmacia)), a cotton membrane (for example, blotting sorbent filter (for example, Biorad) and polyvinylidene difluoride (PVDF) membrane (for example, Biorad).
  • nitrocellulose membrane for example, manufactured by Bio-Rad
  • a nylon membrane for example, Hybond-ECL (manufactured by Amersham-Pharmacia)
  • a cotton membrane for example, blotting sorbent filter (for example, Biorad) and polyvinylidene difluoride (PVDF) membrane (for example, Biorad).
  • PVDF polyvinylidene difluoride
  • the plotting method used is an ⁇ ET-type blotting method (CURRENT PROTOCOLS IN IMMUNOLOGY volume 2 ed by JE Coligan, AM Krui sbeek, DH Margulies, EM Shevach, W. Strober), semi-dry blotting method (see CURRENT PROTOCOLS IN IMMUNOLOGY volume 2 above), and the like.
  • examples of the method for immobilization on a 96-well plate include enzyme-linked immunosorbent assay (ELISA) and radioisotope immunoassay (RIA).
  • the immobilization may be performed, for example, by subjecting the 96-well plate (for example, Imnoplate Maxi Soap (manufactured by Nunc) or the like) to a sample or a dilution thereof (for example, phosphate buffered saline containing 0.05% sodium azide). Add a saline solution (hereinafter, referred to as “PBS”) diluted, and incubate at 4 to room temperature overnight or at 37 ° C for 1 to 3 hours to adsorb the polypeptide on the bottom of the well. You can do it.
  • PBS saline solution
  • anti-PADI4 antibody The “antibody that specifically binds to the ⁇ 4 protein of the present invention (hereinafter referred to as“ anti-PADI4 antibody ”)” used in this step may be prepared according to a known method, or may be commercially available. May be used.
  • the antibody can be prepared by a conventional method (for example, Shinsei Kagaku Kenkyusho 1, Protein 1, P. 389-397, 1992), using the PADI4 protein of the present invention as an antigen or any polypeptide selected from the amino acid sequence thereof. It can be obtained by immunizing an animal with the antibody and collecting and purifying an antibody produced in the living body of the animal. In addition, publicly known methods (for example, Kohler and Children, Nature 256, 495-497, 1975, Kennet, R. ed., Monoclonal Ant ibody p. 365-367, 1980, Prenum Press, NY) were used. Thus, a hybridoma can be established by fusing the antibody-producing cells producing the anti-PADI4 antibody with myeloma cells, and a monoclonal antibody can be obtained therefrom.
  • a conventional method for example, Shinsei Kagaku Kenkyusho 1, Protein 1, P. 389-397, 1992
  • PADI4 protein of the present invention as an antigen or any
  • Antigens for producing antibodies include the PADI4 protein of the present invention or a polypeptide comprising at least six consecutive partial amino acid sequences thereof, or any amino acid sequence or carrier thereof (for example, a keyhole limpet to which an N-terminal is added). And derivatives to which mosyanine has been added.
  • the antigen polypeptide can be obtained by producing a PADI4 protein of the present invention in a host cell by genetic manipulation. Specifically, a vector capable of expressing the PADI4 gene of the present invention is prepared, and this is introduced into a host cell to transform the gene. It may be expressed.
  • the host cell examples include prokaryotic cells, such as Escherichia coli and Bacillus subtilis.
  • the host cells are transformed with a plasmid vector that contains a regulatory element and a replicon, or replication origin, from a species compatible with the host.
  • the vector is preferably one having a sequence capable of imparting phenotypic (phenotypic) selectivity to transformed cells.
  • the K12 strain or the like is often used, and the vector is generally a pBR322 or pUC-type plasmid, but is not limited thereto, and may use various known strains or vectors. it can.
  • examples of a promoter used in Escherichia coli include a tryptophan (trp) promoter, a lactose (lac) promoter, a tryptophan lactose (tac) promoter, a lipoprotein (lpp) promoter, and a polypeptide.
  • trp tryptophan
  • lac lactose
  • tac tryptophan lactose
  • lpp lipoprotein
  • strain 207-25 is preferable, and pT UB 228 (Ohmura, K. et al. (1984) J. Biochem. 95, 87-93) is used as a vector. It is not limited to this.
  • extracellular secretory expression can be achieved by ligating the DN ⁇ sequence encoding the signal peptide sequence of ⁇ -amylase of Bacillus subtilis to the vector.
  • Eukaryotic host cells include cells such as vertebrates, insects, and yeast.
  • Vertebrate cells include, for example, dihydrofolate reduction of monkey COS cells (Gluzman, Y. (1981) Cell 23, 175-182, ATCC CL-1650) and CHO cells (ATCC CCL-61).
  • Enzyme-deficient strains (Urlaub, G. and Chasin, LA (1980) Pro Natl. Acad. Sci. USA 77, 4126-4220) are frequently used, but are not limited thereto.
  • a vector having a promoter, an RNA splice site, a polyadenylation site, a transcription termination sequence, and the like located upstream of a gene to be normally expressed can be used.
  • it may have a replication origin if desired.
  • the expression vector include pCR3.1 (manufactured by Invitrogen) having the cytomegalovirus early promoter and the SV40 V2 dh ⁇ r having an early promoter (Subramani, S. et al. (1981) Mol. Cell. Biol. 1, 854-864), and the like, but are not limited thereto.
  • the expression vector has an SV40 origin of replication, is capable of autonomous growth in COS cells, and has a transcription promoter, a transcription termination signal, and Those having an RNA splice site can be suitably used.
  • the expression vector is prepared by the method of getylaminoethyl (DEAE) -dextran method (Luthman, H. and Magnus son, G. (1983) Nucleic Acids Res, 11, 1295-1308), calcium phosphate-DNA coprecipitation method (Graham, FL) and van der Eb, AJ (1973) Virology 52, 456-457) and electric pulse perforation (Neumann, E. et al. (1982) EMB0 J. 1, 841-845)
  • DEAE getylaminoethyl
  • vectors that can express the neo gene functioning as an antibiotic G418 resistance marker together with the expression vector for example, pRSVneo (Saibrook, J. et. al. (1989): “Molecular Cloning A Laboratory Manual Cold Spring Harbor Laboratory, NY) and p S V2 neo (Southern, PJ and Berg, P. (1982) J. Mol. Appl. Genet. 1, 327-341)
  • Transfected cells that stably produce the desired polypeptide can be obtained by co-transfecting them and selecting G418-resistant colonies.
  • cell lines derived from ovarian cells of Spodoptera frugiperda (S f — 9 or S ⁇ ⁇ 21) of the family Lepidoptera and the High Five cells derived from Trichoplusia ni eggs (Wickham, TJ) et al, (1992) Biotechnol. Prog, i: 391-396) is often used as a host cell, and as a Bacchus virus transfer vector, the polyhedrin protein of autographa nucleopolyhedrovirus (AcNPV) is used. PVL 1392/1393 using the Promo overnight is often used (Kidd, i.
  • baculoviruses as expression vectors. Applied Biochemistry and Biotechnology 420, 137-159).
  • vectors using baculovirus P10 and a promoter of the same basic protein can also be used.
  • AcN The recombinant protein can be expressed as a secreted protein by connecting the secretory signal sequence of the PV envelope surface protein GP67 to the N-terminal side of the target protein (Zhe-mei Wang, et al. (1998) Biol. Chem., 379, 167-174).
  • yeast As an expression system using a eukaryotic microorganism as a host cell, yeast is generally well known. Among them, yeast of the genus Saccharomyces, for example, baker's yeast Saccharomyces cerevis iae and petroleum yeast Pichia pastoris are preferable.
  • expression vectors for eukaryotic microorganisms such as the yeast include, for example, a promoter for an alcohol dehydrogenase gene (Bennettzen, JL and Hall, BD (1982) J. Biol. Chem. 257, 3018-3025) and Acid phosphatase gene promoter (Miyanohara, A. et al. (1983) Proc. Natl. Acad. Sci.
  • the transformant obtained as described above can be cultured according to a conventional method, and the culture produces the target polypeptide inside or outside the cell.
  • the medium used for the culture various types of commonly used media can be appropriately selected depending on the host cell used.
  • serum components such as fetal calf serum were added to a medium such as RPMI 1640 medium or Dulbecco's modified Eagle medium (hereinafter referred to as “DMEM”) as necessary. Anything can be used.
  • DMEM Dulbecco's modified Eagle medium
  • the recombinant protein produced inside or outside the cells of the transformant by the above culture is separated and purified by a known separation operation method utilizing the physical and chemical properties of the protein. be able to.
  • Examples of the method include treatment with a protein precipitant, ultrafiltration, molecular sieve chromatography (gel filtration), adsorption chromatography, ion exchange chromatography, affinity chromatography, high-performance liquid chromatography (HPLC ) Etc.
  • Raffy and dialysis can be used alone or in combination. If a histidine consisting of 6 residues is linked to the recombinant protein to be expressed, it can be purified efficiently with a nickel affinity column.
  • the target PADI4 protein of the present invention can be easily produced in high yield and high purity by appropriately combining the methods described above.
  • the anti-PADI4 antibody obtained by the method described in the above (3) may be directly labeled, or may be used as a primary antibody to specifically recognize the primary antibody (antibody derived from the animal from which the antibody was prepared) Recognition) Used for detection in cooperation with a labeled secondary antibody.
  • the type of the label include an enzyme (alkaline phosphatase or horseradish peroxidase) or piotin (however, an operation of binding an enzyme-labeled streptavidin to the secondary antibody piotin is added).
  • an enzyme alkaline phosphatase or horseradish peroxidase
  • piotin an operation of binding an enzyme-labeled streptavidin to the secondary antibody piotin is added.
  • an enzyme alkaline phosphatase or horseradish peroxidase
  • piotin an operation of binding an enzyme-labeled streptavidin to the secondary antibody piotin is added.
  • the expression level of the antigen is measured.
  • substrates that emit color or emit light by the catalyst of these enzymes are commercially available.
  • a color-developing substrate it can be detected visually using Western plotting or dot / slot plotting.
  • detection can be performed by Western radiography or dot / slot plotting by autoradiography using an X-ray film or imaging plate, or by photographing using an instant camera. Can be. Also, densitometry and molecular imagers — Quantification using Fx system (manufactured by Bio-Rad) is also possible.
  • a luminescent substrate is used in the ELISA method, the enzyme activity is measured using a luminescent microplate reader (for example, Bio-Rad).
  • the membrane in order to prevent non-specific adsorption of antibodies, the membrane must be treated with a substance that inhibits such non-specific adsorption (skim milk, casein, serum albumin, gelatin, etc.). (Polyvinylpyrrolidone, etc.) for a certain period of time (blocking).
  • a substance that inhibits such non-specific adsorption skim milk, casein, serum albumin, gelatin, etc.
  • PBS phosphate buffered saline
  • TBS Tris buffered saline
  • Block Ace (Dainippon Pharmaceutical); -10% serum albumin, 0.5-3% gelatin or 1% polyvinylpyrrolidone may be used.
  • the blocking time is 4 to 16 hours at 4 or 1 to 3 hours at room temperature.
  • washing solution PBS or TBS containing 0.05 to 0.1% Tween 20 (hereinafter referred to as “washing solution”) to remove excess blocking solution, and then diluted appropriately with the blocking solution.
  • An anti-PAD14 antibody is immersed in the solution for a certain period of time to allow the antibody to bind to the antigen on the membrane.
  • the dilution ratio of the antibody at this time can be determined, for example, by performing a preliminary pastelotting experiment using a serial dilution of the recombinant antigen as a sample.
  • This antibody reaction operation is preferably performed at room temperature for 2 hours. After the antibody reaction operation, wash the membrane with washing solution.
  • the detection operation can be performed immediately.
  • an unlabeled antibody performs a secondary antibody reaction. For example, when using a commercially available labeled secondary antibody, dilute it with a blocking solution at a ratio of 2000 to 20000 times. (If a suitable dilution ratio is described in the attached instruction sheet, follow the description.) ). After washing and removing the primary antibody, immerse the membrane in the secondary antibody solution at room temperature for 45 minutes to 1 hour, wash with the washing solution, and perform the detection operation according to the labeling method. The washing operation is performed, for example, by first shaking the membrane in the washing solution for 15 minutes, then replacing the washing solution with a new one. After shaking for 5 minutes, change the washing solution again and shake for 5 minutes. If necessary, the washing solution may be further exchanged for washing.
  • blocking is performed in advance as in the case of Western blot.
  • the blocking conditions are as described in the section of Western plot.
  • washing solution PBS or TBS containing 0.05 to 0.1% Tween 20 (hereinafter referred to as “washing solution”) to remove excess blocking solution, and then appropriately diluted with the washing solution.
  • An anti-PADI4 antibody is dispensed and incubated for a certain period of time to bind the antibody to the antigen.
  • the dilution ratio of the antibody at this time can be determined, for example, by conducting a preliminary ELISA experiment using a sample obtained by serially diluting the recombinant antigen. This antibody reaction operation is preferably performed at room temperature for about 1 hour. After completion of the antibody reaction procedure, wash the inside of the well with a washing solution.
  • the detection operation can be performed immediately. If an unlabeled antibody is used, perform a secondary antibody reaction.
  • a commercially available labeled secondary antibody for example, dilute it to 2000 to 2000 times with a washing solution and use it (when a suitable dilution ratio is described in the attached instruction sheet). Is subject to that statement). Dispense the secondary antibody solution into the wells after washing and removing the primary antibody, incubate at room temperature for 1 to 3 hours, wash with the washing solution, and perform the detection operation according to the labeling method.
  • the washing operation is performed, for example, by first dispensing the washing solution into the well, shaking for 5 minutes, replacing the washing solution with a new one, shaking for 5 minutes, replacing the washing solution again, and shaking for 5 minutes. . If necessary, the washing solution may be further exchanged for washing.
  • the so-called sandwich ELISA can be performed by the method described below.
  • two hydrophilic regions are selected from each amino acid sequence of the PADI4 protein of the present invention.
  • a partial peptide comprising at least 6 amino acid residues in each region is synthesized, and two types of antibodies using the partial peptide as an antigen are obtained.
  • One of the antibodies is labeled.
  • the unlabeled antibody was immobilized on the bottom of the well of a 96-well ELISA plate. I do. After blocking, place the sample solution in a well and incubate at room temperature for 1 hour. After washing the wells, dispense the labeled antibody diluent into each well and incubate. After washing the inside of the well again, perform the detection operation according to the labeling method.
  • Step 3 Evaluation of test substance
  • the test substance when the expression level of the protein is significantly reduced under the test substance administration condition compared to the non-administration condition, the test substance can be evaluated as having an effect as a RA therapeutic agent.
  • “significantly reduced” means, for example, that there is a statistically significant difference (p ⁇ 0.05) in the expression level of the PADI4 protein of the present invention under the test substance administration and non-administration conditions. Means that.
  • a method for evaluating the effect of a test substance as an RA therapeutic agent in vitro using PADI4 activity as an index includes, for example, the following steps. , Step 1: Add PADI4 protein to the reaction mixture containing the arginine-containing compound with or without the addition of the test substance.
  • Step 2 PADI4 protein activity is measured by quantifying the amount of the reaction product in the reaction solution.
  • Step 3 The effect of the test substance as a therapeutic agent for rheumatoid arthritis is evaluated based on the difference in the activity under the conditions of addition or non-addition of the test substance.
  • Step 1 Preparation of reaction solution
  • the reaction solution used in this method contains an arginine-containing compound that can serve as an enzyme substrate for PADI4 protein in a suitable buffer.
  • Arginine-containing compounds include, for example, arginine such as BAEE (Na-benzoyl-L-arginine e thyl ester), N--Benzoyl-L-arginine, dansyl-glycy L-citruline, etc. Compound If it is a thing, it will not be specifically limited.
  • the amount of the test substance to be added to the reaction solution and the method of addition are not particularly limited, and may be appropriately set according to the properties of the test substance. PAD 14 and the substrate are allowed to react at 25 to 5 (around TC for about 1 to 2 hours. Step 2:
  • the activity of the PADI4 protein is measured by quantifying the amount of a reaction product generated by the conversion of arginine residues in the substrate to citrulline by PADI4.
  • the “reaction product” may be a reaction product directly generated from a substrate by an enzymatic reaction, or may be a reaction product generated as a by-product.
  • two methods, a fluorescence method and an absorption method will be described as preferable measurement methods to be used in the present method.
  • the present invention is not limited thereto, and other known methods: for example, fluorescently labeled dansyl-glycyl -Using L-arginine to quantitatively determine the enzymatic reaction product by HPLC (T. Cikura et al., Analytical Biochemistry 285, p230-234, (2000)) be able to.
  • reaction solution for example, a solution is prepared by adding lOmM BAEE or ⁇ -benzoy arginine as a substrate to a lOOmM Tris / HCl (pH 7.8) buffer containing 10 mM CaCl 2 and 5 M DTT.
  • a reaction solution an appropriate amount of PADI4 protein is added, and the reaction is performed, for example, at 37 ° C. for about 1 to 2 hours.
  • stop the reaction by adding an appropriate amount of EDTA, and measure the wavelengths of the excitation wavelength of 413 ⁇ and the fluorescence wavelength of 476 nm using a fluorescence microplate reader.
  • the measurement results were obtained by subtracting the measured value of the reaction solution containing no PADI4 protein as a background, and drawing a calibration curve from the measured values of the reaction solution to which ammonium sulfate was added for the preparation of a calibration curve. Determine the concentration.
  • the activity of the PADI4 protein is evaluated using the ammonium ion concentration as an index.
  • reaction solution for example, in lOOmM Tr is / HCl (pH7. 6) buffer containing 10iM CaCl 2 5mM DTT, a solution is prepared by adding lOmM BAEE (Sigma) as substrate. To this reaction solution, an appropriate amount of PADI4 protein is added, and the reaction is performed, for example, at 37 ° C. for about 1 to 2 hours.
  • the measurement result was calculated by subtracting the measured value of the reaction solution containing no enzyme as a background, and drawing a calibration curve from the measured value of the reaction solution to which citrulline for preparing a calibration curve was added, to obtain the amount of citrulline generated by the reaction. Find the concentration.
  • the activity of PADI4 protein is evaluated using the citrulline concentration as an index.
  • the test substance when the activity of the gene or the protein is significantly reduced under the conditions of the addition of the test substance as compared with the non-addition conditions, the test substance can be evaluated as having an effect as a therapeutic agent for RA.
  • the term “significantly reduced” refers to, for example, a statistically significant difference (p ⁇ 0) between the expression level of the PADI4 gene of the present invention or the PADI4 protein of the present invention under the conditions of adding or not adding a test substance. 0 5).
  • a method for evaluating the effect of a test substance as an RA therapeutic agent in vivo using cultured cells includes, for example, the following steps.
  • the evaluation method preferably includes the following steps.
  • Step 1 Cells are cultured under the condition of adding or not adding a test substance.
  • Step 2 The expression level of the PADI4 gene of the present invention in the above cells is detected, or the expression level of the PADI4 protein of the present invention is detected using an antibody that specifically binds to the protein.
  • Step 3 The effect of the test substance as a therapeutic agent for RA is evaluated based on the difference in the expression level under the conditions of addition or non-addition of the test substance.
  • Step 1 Cell culture
  • the cells used in the evaluation method of the present invention are not particularly limited as long as they are mammalian cells that highly express the PADI4 gene of the present invention.
  • Cultured cells derived from mammals are preferred, and cultured cells derived from blood, synovial tissue, and spleen tissue (preferably, primary cultured cells) are particularly preferred.
  • the mammal is preferably a human, a mouse, a rat or a hamster, and more preferably a human or a mouse.
  • artificially transformed cells such as a cell into which the PADI4 gene of the present invention is introduced together with its promoter region and which is highly expressed, may be used.
  • the cells are cultured with or without the addition of the test substance.
  • the culture method is not particularly limited, and a culture method suitable for the cells may be appropriately selected.
  • the method and amount of the test substance to be added to the cultured cells are not particularly limited either.
  • the test substance is added to the culture medium and cultured for a certain period of time before or at an appropriate time during the culture of the cells.
  • the culture period after the addition of the test substance is not particularly limited, but is preferably 30 minutes to 24 hours.
  • Step 2 Detection of the expression level of the PADI4 gene of the present invention or the PADI4 protein of the present invention
  • the difference in the expression level of the PADI4 gene of the present invention in the above cells under the conditions of addition or non-addition of the test substance is detected.
  • the difference in the expression level of the PADI4 protein of the present invention is detected using an antibody that specifically binds to the protein.
  • the detection of the gene may be basically performed according to the method described in 2.1. Detection using an antibody may be performed according to the method described in 2.2.
  • the present invention uses a gene (hereinafter referred to as “reporter gene”) that allows detection of the promoter activity under the control of the promoter of the PADI4 gene of the present invention.
  • reporter gene a gene that allows detection of the promoter activity under the control of the promoter of the PADI4 gene of the present invention.
  • the expression of the PADI4 gene and the PADI4 protein of the present invention can also be detected.
  • a detection method using the repo overnight gene will be described.
  • the repo overnight gene encodes a reporter protein that is specifically distinguishable from any other protein that the host cell may produce in the course of this test method. Anything can be used.
  • the cells before transformation do not have a gene encoding the same or similar protein as the reporter protein.
  • the reporter protein is toxic to the cell or confers resistance to a susceptible antibiotic, the presence or absence of expression of the repo overnight gene can be determined. It can be determined by the survival rate of the cells.
  • a more preferable reporter gene used in the present invention can specifically and quantitatively detect the expression level (for example, when a specific antibody against the protein encoded by the reporter gene is obtained). It is a structural gene.
  • a gene encoding an enzyme or the like which specifically reacts with a foreign substrate to produce a metabolite which can be easily measured quantitatively examples include, for example, genes encoding the following proteins, but the present invention is not limited thereto.
  • PCAT3-BASIC vector (Promega) is commercially available as a vector that can be used to prepare a vector for repo assay simply by incorporating a promoter.
  • pGL3-BASIC vector-1 (promega) is commercially available.
  • Some substrates can be measured by color reaction, fluorescence or chemiluminescence, respectively.
  • p / 3gal-Basic is commercially available.
  • pSEAP2-Basic (Clontech) is commercially available.
  • Green-fluorescent protein Although it is not an enzyme, it can quantify directly because it emits fluorescence. Similarly, pEGFP-1 (Clontech) is commercially available as a vector for repo overnight.
  • a reporter gene expression plasmid and a recombinant vector in which the PADI4 gene of the present invention of the present invention can be expressed in mammalian cells are prepared, and these are cotransfected into cells.
  • the vector pCR3.1 (manufactured by Invitrogen), pCMV-Script (manufactured by Straugene), and the like can be preferably used, but are not limited thereto.
  • Methods for introducing an expression plasmid into cells include the DEAE-dextran method (Luthman, H. and Magnus son, G. (1983) Nucleic Acids Res. 11, 1295-1308), calcium phosphate-DNA coprecipitation method (Graham, FL and van der Eb, AJ (1973) Virology 52, 456-457), electric pulse perforation (Neumann, E. eUl. (1982) EMBO J. 1, 841-845), ribofection (Lopata et al. (1984) ) Nucl. Acids Res. 12, 5707-5717, Sussman and Milman (1984) Mol. Cell. Biol. 4, 1641-1643).
  • the cells are so-called floating cells, it is preferable to use a method other than the calcium phosphate-DNA coprecipitation method. In any of these methods, optimized transfection conditions are used according to the cells used. It is necessary to use.
  • the “conditions under which the reporter gene can be expressed” are defined as conditions under which the cells transfected by the repo overnight expression vector survive and can produce a transcript (reporter protein) of the reporter gene. I just need.
  • the medium is suitable for the cell line used (or may contain serum components such as fetal bovine serum) and contains 4 to 6% (most preferably 5%) carbon dioxide.
  • Air presence Incubate at 36-38 ° C (most preferably 37 ° C) for 2-3 days (most preferably 2 days).
  • a cell obtained by double-transforming a host cell with the expression vector for the repo allelic gene and the PADI4 gene of the present invention is used. Test methods that have been adopted can also be adopted.
  • an expression vector such as pIND (manufactured by Invitrogen) or pTet-On (manufactured by Clontech) is used to induce the expression of the reporter gene under conditions that induce the expression of the PADI4 gene of the present invention. It is necessary to establish a cell line that promotes expression of the gene. In producing transformed cells, it is desirable that the gene to be introduced be integrated into the chromosome of the host cell and be stably retained in the host cell even after repeated passages.
  • a transgene is ligated with a selectable marker such as an antibiotic resistance or the like (for example, neomycin (or G418) resistance gene neo or the like) and transfected.
  • a selectable marker such as an antibiotic resistance or the like (for example, neomycin (or G418) resistance gene neo or the like)
  • test substance as a therapeutic agent for RA is evaluated based on the difference in the expression level of the PADI4 gene of the present invention or the PADI4 protein of the present invention under the conditions of adding or not adding the test substance.
  • the test substance when the expression level of the gene or protein is significantly reduced under the conditions of the addition of the test substance as compared with the non-addition conditions, the test substance can be evaluated as having an effect as a therapeutic agent for RA.
  • “significantly decreases” means, for example, It means that there is a statistically significant difference (p ⁇ 0.05) in the expression level of the PADI4 gene of the present invention or the PADI4 protein of the present invention under the conditions of adding or not adding the test substance.
  • kits for performing the evaluation method of the present invention include a kit containing at least one of the following a) to: O 2.
  • the primer described in a) above can be readily designed and amplified according to a conventional method, for example, using commercially available primer design software, based on the nucleotide sequence of the above gene (SEQ ID NO: 16 or 17).
  • primers include, for example, oligonucleotides having the nucleotide sequences of SEQ ID NOs: 7 to 10.
  • the probe described in b) above is preferably a single-stranded oligonucleotide or double-stranded DNA having a length of about 20 bases in the case of the Northern hybridization method.
  • a double-stranded DNA having a length of about 100 to 1500 bases or a single-stranded oligonucleotide having a length of about 20 to 100 bases is suitably used.
  • a single-stranded oligo with a length of about 25 bases is preferred.
  • These are preferably designed as probes that specifically hybridize to a portion of the PADI4 gene of the present invention having high sequence specificity.
  • probes examples include, for example, an oligonucleotide having the nucleotide sequence of SEQ ID NO: 11.
  • probes and the aforementioned primers May be labeled with an appropriate label (for example, an enzyme label, a radioactive label, a fluorescent label, or the like), or may be modified with biotin, phosphoric acid, an amine, or the like.
  • the solid-phased sample described in c) above is produced by fixing the probe described in b) above to a solid phase such as a glass plate, a nylon membrane, microbeads, or a silicon chip.
  • a solid phase such as a glass plate, a nylon membrane, microbeads, or a silicon chip.
  • the immobilized sample and the method for preparing the same have already been described in 2.1, but examples thereof include a gene chip, a cDNA array, an oligo array, and a membrane filter.
  • the antibodies described in d) and e) and the protein in f) can be prepared by the method described in 2.2.
  • the antibody may be labeled with a suitable label (for example, an enzyme label, a radioactive label, a fluorescent label, or the like), or may be appropriately modified with biotin or the like.
  • the kit may be used for evaluation of the present invention, such as hybridization, probe labeling, or a reagent for detecting a label, a reaction buffer, and an enzyme substrate, if necessary. It may optionally contain other reagents and the like necessary for carrying out the method.
  • the present invention relates to a mutant PADI4 protein comprising the amino acid sequence shown in SEQ ID NO: 21 in a sample, or a gene thereof (SEQ ID NO: 20).
  • the present invention provides a method for predicting the risk of developing rheumatoid arthritis (hereinafter referred to as “RA”) in a subject who has provided the test sample, based on the expression level of the test sample.
  • RA rheumatoid arthritis
  • specimen used in the above-mentioned method means any sample such as blood, body fluid, tissue, or excrement isolated from a subject, and includes blood (blood-derived cells), synovial tissue (synovial cells) ), Spleen tissue (spleen cells), and cells containing peritoneal cells are preferred, and blood cells (neutrophils, eosinophils) and synovial tissue (synovial cells) are more preferred.
  • the risk of onset may be predicted using the expression level of the mutant PADI4 protein as an index or the expression level of the mutant PADI4 gene (mRNA) as an index.
  • Step 1 preparing all RNAs from each sample isolated from a subject and a normal person;
  • Step 2 detecting the expression level of mRNA of the mutant PADI4 gene in all the above RNAs;
  • Step 3 Analyze the difference in the above expression level between the subject and the normal person, and predict the risk of developing the subject's chronic joint rheumatism.
  • the evaluation was performed using the ratio of the expression level of the mutant PADI4 gene and the appropriate control gene (for example, 3-actin gene ⁇ liposomal RNA, etc.) in the sample, and subjected to the above analysis. Is also good.
  • the method using the expression level of the mutant PADI4 protein as an index includes, for example, the following steps. Step 1: detecting the expression level of the mutant PADI4 protein in a sample isolated from a subject and a normal person using an antibody capable of specifically binding to the protein; Step 2: detecting the expression level in the subject and a normal person The difference in the expression level is analyzed to predict the risk of developing RA in the subject.
  • detection and analysis of the expression level of the mutant PADI4 protein can be performed according to the description in 2.2.
  • the expression level of the mutant PADI4 protein in the subject is significantly higher than that in a normal person, the subject can be evaluated as having a high risk of developing RA.
  • “significantly high” means, for example, a statistically significant difference in the expression level of mutant PADI4 protein between a subject and a normal person (p ⁇ 0.
  • Kit for predicting the risk of developing rheumatoid arthritis The present invention also provides a kit for predicting the risk of developing rheumatoid arthritis.
  • the kit includes, for example, at least one of the following a) to e). a) a 15- to 30-base-length continuous oligonucleotide primer for specifically amplifying a mutant PADI4 gene consisting of the base sequence shown in SEQ ID NO: 20 b) the base shown in SEQ ID NO: 20 A continuous polynucleotide probe with a length of 20 to 150 bases for specifically hybridizing to the mutant PADI4 gene comprising the sequence and detecting the gene.
  • the primer described in a) is a commercially available primer design software based on the nucleotide sequence of the above gene (SEQ ID NO: 20 or 21). It can be easily designed and amplified according to the usual method such as using.
  • a single-stranded oligonucleotide or double-stranded DNA having a length of about 20 bases is suitably used as the probe described in b) above.
  • a microarray a double-stranded DNA having a length of about 100 to 150 bases or a single-stranded oligonucleotide having a length of about 200 to 100 bases is suitably used.
  • a single-stranded oligo with a length of about 25 bases is preferred.
  • These are preferably designed as probes that specifically hybridize to a portion having high sequence specificity of the mutant PADI4 gene.
  • Examples of such a probe include, for example, an oligonucleotide having the base sequence of SEQ ID NO: 4.
  • These probes and the aforementioned primers may be labeled with an appropriate label (for example, an enzyme label, a radioactive label, a fluorescent label, or the like), or may be modified with biotin, phosphoric acid, an amine, or the like.
  • the solid-phased sample described in c) above is prepared by fixing the probe described in b) above to a solid phase such as a glass plate, a nylon membrane, micropease, or a silicon chip.
  • a solid phase such as a glass plate, a nylon membrane, micropease, or a silicon chip.
  • immobilized samples and their preparation methods have already been described in 2.1.
  • the antibody may be labeled with an appropriate label (for example, an enzyme label, a radioactive label, a fluorescent label, or the like), or may be appropriately modified with biotin or the like.
  • an appropriate label for example, an enzyme label, a radioactive label, a fluorescent label, or the like
  • the kit is, in addition to the components described above, necessary for the implementation of the method of the present invention, such as a hybridization, a reagent for detecting the label of the probe or a label, a reaction buffer, etc. Further, other reagents and the like may be appropriately contained.
  • Mutant PADI4 protein ⁇ The gene was newly discovered by the present inventors, and the functional difference between these mutant PADI4 protein ⁇ gene and normal PADI4 protein ⁇ gene is It is closely related to RA onset. That is, a mutant PADI4 protein consisting of the amino acid sequence shown in SEQ ID NO: 21 and a mutant PADI4 gene consisting of the nucleotide sequence shown in SEQ ID NO: 20 are used as diagnostic markers for RA (for example, anti-mutant PADI4 Useful as an antigen for producing antibodies). The present invention also provides RA diagnostic markers using such mutant PADI4.
  • the mutant PADI4 protein has a higher enzyme activity than the normal PADI4 protein, and may be one of the causes of RA onset through enhanced production of citrullinated peptides. Therefore, by using the specific inhibitory effect on the mutant PADI4 protein as an index, screening for RA therapeutic agents is possible.
  • the present invention provides a method for evaluating the effect of a test substance as a therapeutic drug for rheumatoid arthritis using the inhibitory effect on the mutant PADI4 protein consisting of the amino acid sequence shown in SEQ ID NO: 21 as an index.
  • the method includes the following steps.
  • Step 1 Add the mutant PADI4 protein to the reaction solution containing the arginine-containing compound, with or without the addition of the test substance;
  • Step 2 measuring the activity of the mutant PADI4 protein by quantifying the amount of the reaction product in the reaction solution;
  • Step 3 The effect of the test substance as a therapeutic agent for rheumatoid arthritis is evaluated based on the difference in the activity under the conditions of addition or non-addition of the test substance.
  • the mutant PADI4 protein can be prepared as described in 2.2, and steps 1 to 3 can be performed as described in 2.3.
  • the human PADI4 gene and PADI4 protein are specifically highly expressed in diseased tissues (eg, synovial tissue) of RA patients, and may cause RA through the production of citrullinated proteins. Therefore, by measuring the expression levels of the PADI4 gene and PADI4 protein in the specimen isolated from the subject as well as the mutant PADI4 described above, the RA pathology of the subject can be predicted. 5.2 RA model animal
  • an animal eg, a mouse or the like
  • the PADI4 gene or the mutant PADI4 gene of the present invention is incorporated into an appropriate vector and introduced into an animal, and if phenotypic changes such as inflammatory symptoms typified by human RA appear, the animal is used. Research on the condition of RA and its remedies.
  • a model animal exhibiting an RA disease state can be produced.
  • the present invention will be described more specifically with reference to Examples, but the present invention is not limited thereto.
  • Genomes from 846 RA patients and 658 non-patients from clinical facilities around Japan Blood for MA extraction was collected. All sample providers are Japanese. The donors gave their consent for informed consent by the Senyuichi Ethics Committee for Genetic Polymorphism Research at RIKEN. For RA patients, those who met at least three of the following seven criteria of the revised criteria of the American College of Rheumatology for the classification of rheumatoid arthritis were selected as RA patients.
  • the non-patient group was selected from the general Japanese population independently of patients. Sampling was performed so that there was no difference between the RA patient group and the non-patient group except for the presence or absence of the eight diagnosis.
  • Genomic DNA was extracted from leukocytes separated from peripheral blood of RA patients and non-patients as a starting material.
  • blood was collected in a blood collection tube (Benoject) containing EDTA'2Na, and the QIAa DNA Blood Kit (QIAGEN) was used. The absorbance at 260 nm of each sample was measured, and the DNA concentration of each sample was calculated. Thereafter, the prepared DNA was dissolved in lOmM Tris-HCl buffer (pH 8.0) containing lmMEDTA'2Na, and stored at -80 until use.
  • lOmM Tris-HCl buffer pH 8.0
  • Invader probe 5'-CTGCGCACAGGGAGATTTCTGAAATCCCATCAT-3 '(SEQ ID NO: 2)
  • FAM probe for cytosine detection 5'-CTGCGCACAGGGAGATTTCTGAAATCCCATCAT-3 '(SEQ ID NO: 2)
  • Invader Atsushi is prepared by adding an invader reaction solution (Applied Biosystems, Inc.) containing 96-well (Nihon Genetics) or 384-well plates (ABI) containing a 4-fold dilution of the PCR reaction solution 21 and the above three probes. ) was added, and the mixture was incubated at 95 ° C for 5 minutes, and then reacted at 63 ° C for 20 to 80 minutes until a signal was separated. The signal was detected using the ABI PRISM 7700 Seauence Detector System (Applied Biosystems) for 96-well, and the ABI PRISM 7900HT Sequence Detector System (Applied Biosystems) for 384-well.
  • a case-control-related test was conducted for 846 case patients (RA patients) and 658 controls (non-patients) for the single nucleotide polymorphism at the 3rd intron 2 13 6 Was.
  • the test was performed by comparing the allele frequency distribution between the two groups, comparing the genotype distribution using the dominant inheritance model, and comparing the dienotype frequency using the recessive inheritance model.
  • a 2 ⁇ 2 contingency table ⁇ 2 test was used as a test method. Table 1 shows the analysis results.
  • GenBank accession number An attempt was made to detect a novel gene polymorphism on the PADI4 gene, which is present at nucleotides 415,085 to 470,600 in the genome sequence shown in NTJ30584.2 by the following method.
  • PCR was performed in 96-well PCR plates, and 1 ng of 4 ng / ⁇ 1 genomic DNA was used for each well.
  • the reaction 26.5mM (NH 4) 2 S0 4 , 7.2mM Tris-HCl (pH8.8), 3.2 mM Mg 2+ , 1.5 mM dNTPs, 1.6 mM b-Mercapto ethanol, 10% DMSO (If DMSO is not used, substitute with ⁇ ⁇ 0), IM primers, 2.OU DNA polymerase (ExTaa (TaKaRa) ),
  • the experiment was performed under the solution conditions of 0.04 xM Taa Start Ant ibody (Clontech). After denaturation at 94 ° C for 2 minutes, the extension reaction was performed at 94T for 15 seconds, at an annealing temperature optimized for each reaction for 45 seconds, and at 72 for 1 or 3 minutes. 37 cycles were performed using an ABI9700 thermal cycler (Applied Biosystems). The amplified DNA sequence was reacted using BigDye TM Terminator RR Mix (Applied Biosystems) and the nucleotide sequence was determined using ABI 3700 seciuencer. 2. Result
  • nucleotide sequences of 48 regions were compared for each region, and the nucleotides with polymorphism were regarded as single nucleotide polymorphisms. As a result, the following three new exon single nucleotide polymorphisms with amino acid substitution and one new exon polymorphism without amino acid substitution were found.
  • PADI4 3rd intron 2 1 3 6th (1-15) strongly associated with RA Pairwise Linkage Disequilibrium Index (D ') between the nucleotide single nucleotide polymorphism (PADI4 intron 3-15 T / C) and four new single nucleotide polymorphisms on PADI4 was calculated using the EM algorithm. 2. Result
  • the EM algorithm shows that the four single nucleotide polymorphisms are almost completely linked to each other (98.9 to 100%), and are also almost completely linked to PADI4 intron 3 -15 T / C. Haplotype analysis.
  • the known single nucleotide polymorphisms in the PADI2 and PADI1 genes near PADI4 and the PADI4 intron 3-15 T / C almost reached linkage equilibrium.
  • cDNA synthesized by first strand cDNA synthes is kit (Amersham Pharmacia) using bone marrow total RNA (Clontech) was cloned into DONR201 (Invitrogen). The obtained cDNA was further recloned into PDEST14 (Invitrogen) containing the T7 promoter, and pV18-T7 And PV9-T7 vectors were prepared and sequenced.
  • pV18-T7 and pV9-T7 vectors were digested with Clal, and in vitro transcription was performed using RiboMax TM Large Scale RNA Product ion System-T7 (Promega Chido) to produce V9 and V18 mRNA. .
  • Bacterial cells were 30m1 of 50mM Tris-HCl (pH 7.6) (Wako Pure Chemical), lOOmM sodium chloride (Wako Pure Chemical), 2mM dithiothreitol (DTT: Wako Pure Chemical), specified amount of complete protease
  • the suspension was suspended in a lysis buffer containing an inhibitor cocktail (Roche Diagnostics), sonicated at 4 ° C., and centrifuged at 97,000 ⁇ g for 30 minutes to collect the supernatant.
  • the supernatant was applied to a Mi Ilex-HV (Millipore) filter.
  • buffer B 50 mM Tris-HCl (pH 7.6) (Wako Pure Chemical Industries), 0.1 M sodium chloride (Wako Pure Chemical Industries), 0.1% Triton X-100 ⁇ And added to a Ni-NTA super-flow column (QIAGEN). The column was washed with buffer B containing 20 mM imidazole and eluted with buffer B containing 200 mM imidazole.
  • a 0-50 nM ammonium sulfate solution was also added to another well to prepare a calibration curve. Further, 1 ml of 50 mM boric acid and 50 xl of an equal volume mixed solution of 50 mM o-phthalaldehyde and 50 mM DTT were added to these wells, and reacted at room temperature for 1 hour. The reaction solution was transferred to a white plate (Coaster Coating Co., Ltd.), and the excitation wavelength of 413 nm and the fluorescence wavelength of 476 nm were measured using Spectra max GEMINI (Molecular Devices).
  • the measured value of the well without enzyme was subtracted as the background, and the concentration of ammonium ion generated by the reaction was determined by drawing a calibration curve from the measured value of the well with the addition of ammonium sulfate for preparing the calibration curve (Fig. 2A). ;).
  • a 0-400 M L-citrulline solution was also added to another well, and a mixed solution of solution A and solution B was similarly added.
  • the mixed solution was reacted using a thermal cycler (GeneAmp TM PCR System 9700, Applied Biosystems) at 95 for 15 minutes and then at room temperature for 10 minutes.
  • the solution after the reaction was transferred at 150 ⁇ 1 minute to a 96-well plate (Corning), and the absorbance at 540 nm was measured using Spectra max 250 (Molecular Devices).
  • the concentration of citrulline produced by the reaction was determined by subtracting the measured value of the well containing no enzyme as a background and drawing a calibration curve from the measured value of the well to which citrulline for preparing the calibration curve was added (Fig. 2B). ).
  • the MESACUP ACF test for the anti-citrullinated peptide antibody titer of RA patient sera was performed using a kit commissioned by the Institute of Medical Biology (Nagoya).
  • the MESACUP ACF test is a method for measuring antibody titer by ELISA using citrullinated filaggrin, which is obtained by converting recombinant human filadrin peptide into citrulline by PAD, as an antigen.
  • serum was prepared from peripheral blood of a patient, and 10 zl was added to 1 mL of a reaction buffer and diluted 101-fold.
  • the anti-filaggrin antibody standard serum 2, the standard serum 1, and the diluted sample were added to the primary reaction preparation microplate at 150 / xL in the same manner as in the actual case. Thereafter, 100 L was transferred to the antigen-sensitized microcup using a multi-pip. After allowing the reaction to stand at room temperature (20 to 25 ° C) for 1 hour, the plate was washed four times with a washing solution, and the enzyme-labeled antibody was added by a multipipette.
  • reaction was allowed to stand at room temperature (20 to 25 ° C) for 1 hour, and the reaction terminating solution was added by a multipipette.
  • the sample after each reaction was measured for OD450 using a spectrophotometer, and the Index value was calculated using the following equation.
  • A450 of sample A450 of standard serum 1
  • Pro STAR HF (Stratagenes) was added to a piece of tissue prepared from the synovial tissue of RA patient, and one step in situ-RT-PCR was performed under the following conditions using Hybaid Omni slide (Hybaid Ltd., Middlesex, UK). Implemented above.
  • PCR was performed on the hPADI4 V18 cDNA (SEQ ID NO: 16) using the following primers for PADI4 amplification, and the resulting 335-base amplification product was labeled with digoxigenin (DIG) to detect PADI4. A probe was used.
  • DIG labeling was performed using PCR DIG probe synthes is kit (Roche Diagnostics). To the sample after reverse transcription, the above-mentioned primer was added to a final concentration of 0.34 M, covered with a slide glass, and prehybridized at 37 ° C for 1 hour. Next, a DIG-labeled probe denatured at 94 ° C for 5 minutes was added, and hybridization was performed at 37 ° C for 12 hours. After the reaction, the slide was washed, and the PADI4 gene was detected using digoxygenin detect ion kit (Roche Diagnostics). As a negative control, detection was performed in the same manner with no primer added and no reaction enzyme added.
  • CIA Collagen-induced arthritis
  • mPADI4 mouse peptidylarginine diminase 'type IV
  • MPADM is a mouse ortholog of human peptidylarginine diminase 'type IV (hPADI4).
  • the cDNA sequence and amino acid sequence of mPADI4 are shown in SEQ ID NO: 18 and SEQ ID NO:
  • the expression of the mPADI4 gene was examined by RT-PCR using ABI PRIZM 7700 System (Applied Biosystems).
  • the used primers for amplifying the mPADI4 gene and the probe for detecting the mPADI4 gene are as follows.
  • the expression level of mPADI4 gene was corrected by the expression level of mouse / 3-glucuronidase (mGUS) gene used as an internal standard.
  • the sequences of the primer for amplifying the MGUS4 gene and the probe for detecting the MGUS4 gene are as follows.
  • the results are shown in FIG.
  • the expression level of mPADI4 was increased 19.5-fold in the synovial tissue of the CIA group compared with the healthy group.
  • the expression level of mPAD was three times higher than in the spleen tissue of the healthy group. From the above, it was confirmed that mPAD gene expression was significantly increased in synovial tissue and spleen tissue in the CIA group.
  • a heron Kbl'JW, Oss, age: retired
  • FCA Full's complete adjuvant
  • DIFC0 incomplete adjuvant
  • FIA incomplete Freund's adjuvant
  • a synovial tissue piece was obtained from the dissected tissue obtained during the total knee arthroplasty.
  • the obtained synovial tissue piece was immediately immersed in liquid nitrogen and stored at -80 until preparation of a thin section on a slide glass.
  • fix the frozen tissue section with 10% neutralized buffered formalin, and add 70% ethanol, 85% ethanol, 90% ethanol, and 100% ethanol (twice for 100% ethanol only).
  • the tissue was treated once with 50% ethanol and 50% paraffin solution for 30 minutes and twice with 100% paraffin solution for 30 minutes to paraffinize the tissue pieces. Thereafter, the tissue pieces were embedded in paraffin. Paraffin-embedded tissue slices were sliced to a thickness of 4 m with Leica Microtome RM2165 and fixed on slide glasses.
  • the mRNAs of the two PADI4 haplotypes were prepared by the following method.
  • a plasmid in which the gene encoding V9 or V18 was connected downstream of the CMV promoter was transfected into human K562 cells using Lipofectamine 2000 (Invitrogen) according to the attached protocol. After culturing at 37 ° C for 4 hours, PMA (Sigma) was added to a final concentration of 50 ng / ml. After culturing for 40 hours, actinomycin D (Sigma) was added to a final concentration of 5 x g / ml, and cells were collected after 0, 2, and 4 hours, respectively.
  • the remaining human PADI4 V9 or V18 mRNA is measured using ABI PRISM 7700 Sequence Detection System (Applied Biosystems) using TaqMan One-step RT-PCR Master Mix (Applied Biosystems) and following the specified protocol. did.
  • the present invention can be used for screening drugs for treating rheumatoid arthritis in drug development.
  • the present invention can be used in the medical field for early determination of the potential risk of a subject to develop rheumatoid arthritis. Sequence listing free text
  • SEQ ID NO: 1 Human PADI4 gene fragment (first intron-1000 to fourth intron) Description of one mutation: Single nucleotide polymorphism due to substitution of (A) and (G)

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biomedical Technology (AREA)
  • Analytical Chemistry (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Physics & Mathematics (AREA)
  • Urology & Nephrology (AREA)
  • Medicinal Chemistry (AREA)
  • Hematology (AREA)
  • Pathology (AREA)
  • Food Science & Technology (AREA)
  • General Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Biophysics (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Enzymes And Modification Thereof (AREA)

Abstract

A method of evaluating the therapeutic effect of a remedy for rheumatoid arthritis and a method of estimating the risk for onset of rheumatoid arthritis by using peptidyl arginine deiminase type IV. Namely, it is intended to provide a method of evaluating the effect of a test substance as a remedy for rheumatoid arthritis by using its inhibitory effect on a peptidyl arginine deiminase type IV gene, an orthologous gene thereof or a protein encoded by the gene as an indication. It is also intended to provide a method of estimating the risk for onset of rheumatoid arthritis in a subject based on the expression amount of a mutant peptidyl arginine deiminase type IV protein comprising the amino acid sequence represented by SEQ ID NO:21 or its gene (SEQ ID NO:20) in a specimen which is provided by the subject.

Description

ぺプチジルアルギニン ·ディミナ一ゼ ·タイプ I Vの利用方法 技術 分 野  Peptidyl arginine · Diminase · Type IV usage technology Technology
本発明は、 ぺプチジルアルギニン ·ディミナ一ゼ'タイプ I Vを利用した、 慢性関節リゥマチ治療薬の効果を評価する方法、 および慢性関節リゥマチの発 症危険度を予測する方法に関する。  The present invention relates to a method for evaluating the effect of a therapeutic agent for rheumatoid arthritis, and a method for predicting the risk of developing rheumatoid arthritis, using peptidylarginine diminase 'type IV.
 Light
背 景技術 Background technology
慢性関節リウマチ (R A) は関節の滑膜書組織を病変の主座とする慢性炎症性 疾患であり、 有病率が人口の約 1 %を占める疾患である。 R Aは、 初期には滑 膜炎を来し、 次第に軟骨や骨が侵され、 進行すると関節が破壌され変形する。 症状の経過は、 関節炎の寛解 ·再燃を繰り返し、 完治する例や急速に進行する 例など多彩であるが、 早期発見と早期治療が R A患者の生活の質を改良する上 からも重要である。  Rheumatoid arthritis (RA) is a chronic inflammatory disease that mainly involves the synovial tissue of the joints, with a prevalence of about 1% of the population. RA initially develops synovitis, gradually invades cartilage and bones, and as it progresses, the joints rupture and deform. The course of symptoms varies, including repeated cures and relapses of arthritis, complete cure, and rapid progression. Early detection and early treatment are also important for improving the quality of life of RA patients.
R Aは病態的には関節に達した未知の抗原に対する特異的な免疫応答に始ま り、 関節に浸潤した血中リンパ球、 マクロファージ、 好中球による慢性炎症と 滑膜増殖を生じ、 その結果、 炎症性肉芽組織由来の破骨細胞やプロテアーゼに よる骨,軟骨破壊が起きる。 R Aの病因は不明であるが、 遺伝的要因と微生物 感染などの環境要因の両者が関与していると考えられている。  RA pathologically begins with a specific immune response to an unknown antigen that has reached the joints, causing chronic inflammation and synovial proliferation by blood lymphocytes, macrophages, and neutrophils that have infiltrated the joints, Osteoclasts derived from inflammatory granulation tissue and bone and cartilage destruction by proteases occur. Although the etiology of RA is unknown, both genetic and environmental factors, such as microbial infection, are thought to be involved.
R Aの診断は主に症状によってなされるが、 早期診断には R A患者の血中に 見出される自己抗体の検出が今のところ有用である。 そのような自己抗体とし ては、 例えば、 リウマトイド因子、 抗 Sa蛋白抗体、 抗 perinuclear factor抗 体、 抗ケラチン抗体、 抗フィラグリン抗体、 抗シトルリン化ペプチド抗体など が知られている(Mart inus A. M. van Boekel et al. , Arthr i t is Research (2002) 4 (2) , p87-93)  The diagnosis of RA is mainly based on symptoms, but detection of autoantibodies found in the blood of RA patients is currently useful for early diagnosis. As such autoantibodies, for example, rheumatoid factor, anti-Sa protein antibody, anti-perinuclear factor antibody, anti-keratin antibody, anti-filaggrin antibody, anti-citrullinated peptide antibody and the like are known (Mart inus AM van Boekel). et al., Arthr it is Research (2002) 4 (2), p87-93)
抗シトルリン化ペプチド抗体は R A患者血清の約 7 6 %から検出されるが、 リゥマチ様症状を呈するが R Aとは判定されない患者血清からは約 4 %しか検 出されない、 R Aに極めて特異性の高い自己抗体である(Gerard A. Schel lekens et al. , The Journal of Cl inical Invest igat ion (1998) 101 (1) , 273-281 ; ZhiJ ie Zhou and Henri-Andre Menard, Current Opinion in Rheumatology (2002) 14 (3), p250-253 また、 R A患者の滑膜にはシトルリン化されたタンパクが 検出されることから、 シトルリンを含むェピトープの R Aの病原性への関与が 指摘されている (Wal ter J van Venrooij and Ger J. M. Pruijn, Arthri t is Research (2000) 2 (4) , p249-251 ; Christ ine Masson-BessiD et al., Journal of Immunology (2001) 166 (6) , p4177-4184)0 Anti-citrullinated peptide antibody is detected in about 76% of sera of RA patients, but only about 4% is detected in sera of patients who show rheumatic-like symptoms but are not determined to be RA. It is an autoantibody (Gerard A. Schel lekens et al., The Journal of Clinical Investigation (1998) 101 (1), 273-281; ZhiJie Zhou and Henri-Andre Menard, Current Opinion in Rheumatology (2002) 14 (3), p250-253 Citrullinated proteins are detected in the synovium of RA patients, suggesting that epitopes containing citrulline may be involved in the pathogenesis of RA (Walter J van Venrooij and Ger JM Pruijn, Arthrit is Research (2000) 2 (4), p249-251; Christine Masson-BessiD et al., Journal of Immunology (2001) 166 (6), p4177-4184) 0
ぺプチジルシトルリンはタンパク内のアルギニン残基がぺプチジルアルギニ ン ·ディミナ一ゼ (以下、 「PADI」 という。) で変換されて生ずるアミノ酸であ る。 げっ歯類においては、 現在まで 4つのサブタイプの PADIが知られており、 タイプ Iは表皮や子宮で、 タイプ IIは筋肉、 子宮、 唾液腺、 膝臓など様々な組 織で、 タイプ II Iは表皮や毛根嚢胞で、 タイプ I Vは様々な組織で発現してい る (Ahied Abu Rus' d et al. , European Journal of Biochemistry (1999) 259 (3), p660- 669)。 ヒトにおいては、 げっ歯類のタイプ I、 Π、 ΠΙ、 I Vに最も相同 性が高い遺伝子としてそれぞれタイプ I、 II、 III、 Vの遺伝子が遺伝子情報デ —夕ベース G e n B a n k上に登録されていたが、タイプ I II (Takuya Kanno et al. , Journal of Invest igat ive Dermatology (2000) 115 (5) , p813-823) およ びタイプ V (Katsuhiko Nakashiia et al. , Journal of Biological Chemistry (1999) 274 (39) , p27786-27792)以外については詳細な報告はなされていない。 ヒ卜の PADIのタイプ IIIはげつ歯類と同じく毛根嚢胞部分で (Takuya Kanno et al. , Journal of Invest igat ive Dermatology (2000) 115 (5) , p813 - 823)、 夕 ィプ Vは好中球や好酸球で (Hiroaki Asaga et al. , Journal of Leukocyte Biology (2001) 70 (1) , p46-51) それぞれ発現していることが報告されている が、 PADIと R Aの直接の関連は不明である。  Peptidyl citrulline is an amino acid produced by the conversion of arginine residues in proteins by peptidyl arginine diminase (hereinafter referred to as “PADI”). To date, four subtypes of PADI are known in rodents: type I is epidermis and uterus, type II is various tissues such as muscle, uterus, salivary glands, and knees; type II I is In epidermal and hair root cysts, type IV is expressed in various tissues (Ahied Abu Rus'd et al., European Journal of Biochemistry (1999) 259 (3), p660-669). In humans, genes of type I, II, III, and V have the highest homology to rodent types I, II, III, and IV, respectively, and are registered on Gene Information Bank. However, type I II (Takuya Kanno et al., Journal of Investigative Dermatology (2000) 115 (5), p813-823) and type V (Katsuhiko Nakashiia et al., Journal of Biological Chemistry (1999) ) Other than 274 (39), p27786-27792), no detailed report has been made. Type III of human PADI is the same as that of rodents in hair root cysts (Takuya Kanno et al., Journal of Investigative Dermatology (2000) 115 (5), p813-823), and evening V is favorable (Hiroaki Asaga et al., Journal of Leukocyte Biology (2001) 70 (1), p46-51), respectively, but direct association between PADI and RA has been reported. Unknown.
なお、 本願基礎出願時においてタイプ Vと言われていた遺伝子は、 その後マ ウスタイプ I Vのォーソログであることが確認されたため、 本明細書中におい ては、 ヒト PADIのタイプ Vをタイプ I Vと呼ぶこととする。  In addition, since the gene which was said to be type V at the time of the basic application of the present application was subsequently confirmed to be a mouse type IV ortholog, the type V human PADI is referred to as type IV in this specification. And
個人の体質を規定する大きな要因として遺伝子多型がある。 最近のファーマ コジエノミクス研究の進展により、 遺伝子多型と薬物の効果、 あるいは副作用 との関係が解明され、 当該薬物の効果、 あるいは副作用を遺伝子診断を用いて 投与前に予測することが可能になりつつある。 また、 遺伝子多型と疾患との関 係を調べることにより、 一部の疾患の事前診断や予後の判定も可能になりつつ める。 A major factor that determines the constitution of an individual is a genetic polymorphism. Recent advances in pharmacogenomics research have elucidated the relationship between genetic polymorphisms and the effects or side effects of drugs, and the effects or side effects of the drugs using genetic diagnosis It is becoming possible to predict before administration. In addition, by examining the relationship between genetic polymorphisms and diseases, it will be possible to make preliminary diagnoses and determine prognosis for some diseases.
前者の例としては、 薬物代謝酵素の遺伝子多型が挙げられる。 例えば、 多型 に関連して活性が増加、 あるいは、 減少する薬物代謝酵素として、 シトクロム P 4501 A2, シ卜クロム P4502A6、 シ卜クロム P4502C 9、 シ 卜クロム P 4502 C 19, シトクロム P 4502D 6、 シ卜クロム P450 2 E 1などが知られている。 また、 チォプリンメチルトランスフェラーゼ、 N -ァセチルトランスフエラーゼ、 UDP-ダルクゥロノシルトランスフェラー ゼ、 および、 ダル夕チオン S—トランスフェラ一ゼなどの抱合酵素と呼ばれる 一群の酵素群にも遺伝子多型が存在し、 多型により活性が減少することが報告 されている (中村祐輔編, 「SNP遺伝子多型の戦略」 , (2000), 中山書店, 東 京)。  An example of the former is a gene polymorphism of a drug metabolizing enzyme. For example, drug metabolizing enzymes whose activity is increased or decreased in association with polymorphism include cytochrome P4501A2, cytochrome P4502A6, cytochrome P4502C9, cytochrome P4502C19, cytochrome P4502D6, Cytochrome P450 2 E 1 and the like are known. Genetic polymorphisms also occur in a group of enzymes called conjugating enzymes, such as thiopurine methyltransferase, N-acetyltransferase, UDP-dalcuronosyltransferase, and daryuthion S-transferase. It has been reported that the activity is reduced by polymorphism (Yusuke Nakamura, ed., “Strategy for SNP gene polymorphism”, (2000), Nakayama Shoten, Tokyo).
後者の例としては、 多型解析研究により見出された疾患原因遺伝子が挙げら れる。 例えば、 (1)潰瘍性大腸炎の原因遺伝子としての HLA、 (2)慢性関節リ ゥマチの原因遺伝子としての TCR 、 (3)アルツハイマー病の原因遺伝子とし ての A P 0 E 4、 (4)精神分裂症の原因遺伝子としてのドーパミン D 3受容体、 (5)躁鬱病の原因遺伝子としてのトリブトファン水酸化酵素、 (6)アルブミン尿 症の原因遺伝子としてのアンジォテンシン前駆体、(7)心筋梗塞の原因遺伝子と しての血液凝固因子 V I I、(8)肥満の原因遺伝子としてのレブチンなどが知ら れている (L. Kruglyak, Nature Genetics (1999) 22 (2), pl39-144)。  An example of the latter is a disease-causing gene found by a polymorphism analysis study. For example, (1) HLA as a causative gene for ulcerative colitis, (2) TCR as a causative gene for rheumatoid arthritis, (3) AP0E4 as a causative gene for Alzheimer's disease, (4) mental health Dopamine D3 receptor as a causative gene for schizophrenia, (5) tributophan hydroxylase as a causative gene for manic depression, (6) angiotensin precursor as a causative gene for albuminuria, (7) myocardial infarction Blood coagulation factor VII is known as a causative gene for leukemia, and (8) lebutin is known as a causative gene for obesity (L. Kruglyak, Nature Genetics (1999) 22 (2), pl39-144).
一方、 TNF— aは、 各種炎症性疾患の病態形成に関与する重要な物質と考 えられているが、 最近、 TNF— α遺伝子の 5' 末端側上流域に TNF—ひ遺 伝子の発現を亢進する多型が見出された (T. Higuchi et al. , Tissue Antigens (1998) 51 (6), p605- 612)。 これらの多型は、 TN F— a遺伝子の発現量を亢進 させるものであるため、 若年性関節リウマチ、 慢性関節リウマチ、 糖尿病など の TNF— 0;が関与する疾患の診断マーカ一になりうるものと考えられている。 発 明 の 開 示  On the other hand, TNF-a is thought to be an important substance involved in the pathogenesis of various inflammatory diseases. Recently, however, the expression of the TNF-h gene has occurred in the 5'-terminal upstream region of the TNF-α gene. (T. Higuchi et al., Tissue Antigens (1998) 51 (6), p605-612). Since these polymorphisms enhance the expression level of the TNF-a gene, they can be diagnostic markers for diseases involving TNF-0; such as juvenile rheumatoid arthritis, rheumatoid arthritis, and diabetes. It is believed that. Disclosure of the invention
本発明は、 慢性関節リゥマチ治療薬の効果あるいは慢性関節リゥマチの発症 危険度を評価するための新規な方法を提供することを目的とする。 The present invention relates to the effects of a therapeutic agent for rheumatoid arthritis or the onset of rheumatoid arthritis. The purpose is to provide a new method for assessing risk.
上記課題を解決するために鋭意検討した結果、 本発明者らは R A患者の病変 部位特異的に、 PADI4タンパクやその mRNAが高発現すること、そしてこの PADI4 の髙発現がシ卜ルリン化タンパクのレベルを亢進させ、 R A発症の一因となり うることを見出した。 これらの知見から、 本発明者らは、 PADI4 タンパクまた はその mR N Aの発現量や活性に対する阻害効果を指標として、 慢性関節リゥ マチ治療薬の評価が可能であると考えた。  As a result of intensive studies to solve the above problems, the present inventors found that PADI4 protein and its mRNA are highly expressed specifically in the lesion site of RA patients, and that the expression of PADI4 Increased levels and contributed to RA development. Based on these findings, the present inventors considered that it is possible to evaluate a therapeutic agent for rheumatoid arthritis using the inhibitory effect on the expression level or activity of PADI4 protein or its mRNA as an index.
すなわち、 本発明は、 PADI4遺伝子若しくはそのォーソログ遺伝子、 または 該遺伝子にコードされる蛋白質に対する阻害効果を指標として、 被験物質の慢 性関節リゥマチ治療薬としての効果を評価する方法に関する。  That is, the present invention relates to a method for evaluating the effect of a test substance as a therapeutic agent for chronic rheumatoid arthritis, using as an index the inhibitory effect on the PADI4 gene or its orthologous gene, or the protein encoded by the gene.
ある実施態様において、 本発明の評価方法は下記の工程を含む:  In one embodiment, the evaluation method of the present invention includes the following steps:
1 ) 動物を被験物質の投与または非投与条件下で飼育する;  1) keep the animals under the conditions of administration or non-administration of the test substance;
2 ) 上記動物の血液または細胞中における PADI4遺伝子またはそのォーソログ 遺伝子の発現量を検出する;  2) detecting the expression level of the PADI4 gene or its orthologous gene in the blood or cells of the animal;
3 ) 被験物質の投与または非投与条件下における、 上記発現量の相違に基づい て、 該被験物質の慢性関節リゥマチ治療薬としての効果を評価する。 3) The effect of the test substance as a therapeutic agent for rheumatoid arthritis is evaluated based on the difference in the expression amount under the conditions of administration or non-administration of the test substance.
前記方法において、 遺伝子の発現量は、 例えば、 遺伝子チップ、 c D NAァ レイ、 およびメンブレンフィルタ一から選ばれる固相化試料を用いた核酸ハイ ブリダィゼ一シヨン法、 R T- P C R法、 リアルタイム P C R法、サブトラクシ ヨン法、 ディファレンシャル ·ディスプレイ法、 ディファレンシャル ·ハイブ リダイゼーション法、 ならびにクロスハイプリダイゼーション法から選ばれる いずれか一つの方法によって検出される。  In the above method, the expression level of the gene can be determined, for example, by using a nucleic acid hybridization method, an RT-PCR method, a real-time PCR using a solid-phased sample selected from a gene chip, a cDNA array, and a membrane filter. , A subtraction method, a differential display method, a differential hybridization method, and a cross-hybridization method.
別な実施態様において、 本発明の評価方法は下記の工程を含む:  In another embodiment, the evaluation method of the present invention comprises the following steps:
1 ) 動物を被験物質の投与または非投与条件下で飼育する;  1) keep the animals under the conditions of administration or non-administration of the test substance;
2 ) 上記動物の血液または細胞中において、 PADI4遺伝子またはそのォーソロ グ遺伝子にコ一ドされる蛋白質の発現量を、 該蛋白質に特異的に結合する抗体 を用いて検出する;  2) detecting the expression level of the protein encoded by the PADI4 gene or its orthologous gene in the blood or cells of the animal using an antibody that specifically binds to the protein;
3 ) 被験物質の投与または非投与条件下における、 上記発現量の相違に基づい て、 該被験物質の慢性関節リゥマチ治療薬としての効果を評価する。  3) The effect of the test substance as a therapeutic agent for rheumatoid arthritis is evaluated based on the difference in the expression amount under the conditions of administration or non-administration of the test substance.
前記方法において、 蛋白質の発現量は、 例えば、 ウエスタンプロット法、 ド ットプロット法、 スロットブロット法、 E L I S A法、 および R I A法から選 ばれるいずれか一つの方法によって検出される。 In the above method, the expression level of the protein is determined by, for example, Detected by one of the following methods: blot plot, slot blot, ELISA, and RIA.
本発明の評価方法で用いられる細胞としては、 血液由来細胞、 滑膜細胞、 脾 臓細胞、 および腹腔浸潤細胞が好ましい。 また、 本発明の評価方法で用いられ る動物としては、 マウスが好ましく、 この場合、 検出対象は、 PADI4遺伝子の マウスォーソログ:マウス PADI4遺伝子、 または該遺伝子にコードされる蛋白 質であることが好ましい。  The cells used in the evaluation method of the present invention are preferably blood-derived cells, synovial cells, spleen cells, and peritoneal cells. In addition, the animal used in the evaluation method of the present invention is preferably a mouse. In this case, the detection target is preferably the mouse ortholog of the PADI4 gene: the mouse PADI4 gene, or a protein encoded by the gene.
さらに別な実施態様において、 本発明の評価方法は下記の工程を含む: In still another embodiment, the evaluation method of the present invention comprises the following steps:
1 ) アルギニン含有化合物を含む反応液に、 被験物質の添加または非添加条件 下で、 PADI4蛋白質を加える。 1) Add PADI4 protein to the reaction mixture containing the arginine-containing compound with or without the test substance.
2 ) 上記反応液中における反応生成物の量を定量することにより、 PADI4蛋白 質の活性を測定する。  2) The activity of PADI4 protein is measured by quantifying the amount of the reaction product in the above reaction solution.
3 )被験物質の添加または非添加条件下における、上記活性の相違に基づいて、 該被験物質の慢性関節リゥマチ治療薬としての効果を評価する。  3) The effect of the test substance as a therapeutic agent for rheumatoid arthritis is evaluated based on the difference in the above activities under the conditions of addition or non-addition of the test substance.
前記方法において、 例えば、 蛋白質の活性は、 蛍光法または吸光法を用いた 反応生成物の定量によって測定されることが好ましい。 また PADI4蛋白質はヒ ト由来の PADI4蛋白質であることが好ましい。 さらに、 本発明者らは PADI4遺伝子上に R A患者に高い頻度で現れるハプロ タイプが存在することを見出した。 そしてこのハプロタイプは、 正常型 PAD 14 蛋白質とはァミノ酸配列や酵素活性が異なる変異型 PAD 14蛋白質をコードし、 この違いが R Aの発症と深い関連を有することを見出した。これらの知見から、 本発明者らは、 変異型 PADI4タンパク質やその遺伝子を利用すれば、 R Aの発 症危険度の予測や R Aに対する治療効果の評価が可能であると考えた。  In the above method, for example, the activity of the protein is preferably measured by quantification of the reaction product using a fluorescence method or an absorption method. The PADI4 protein is preferably a human-derived PADI4 protein. In addition, the present inventors have found that there is a haplotype on the PADI4 gene that frequently appears in RA patients. They found that this haplotype encodes a mutant PAD 14 protein having a different amino acid sequence and enzyme activity from the normal PAD 14 protein, and that this difference is closely related to the onset of RA. Based on these findings, the present inventors thought that the use of the mutant PADI4 protein and its gene would enable prediction of the risk of developing RA and evaluation of the therapeutic effect on RA.
すなわち、 本発明は、 検体中の配列番号 2 1に示されるアミノ酸配列からな る変異型 PADI4蛋白質、 またはその遺伝子 (配列番号 2 0 ) の発現量に基づい て、 該検体を提供した被験者の慢性関節リゥマチの発症危険度を予測する方法 を提供する。  That is, the present invention provides a method for chronically treating a subject who has provided a specimen based on the expression level of a mutant PADI4 protein having the amino acid sequence shown in SEQ ID NO: 21 or the gene (SEQ ID NO: 20) in the specimen. A method for predicting the risk of developing rheumatoid arthritis is provided.
ある実施態様において、 本発明の予測方法は以下の工程を含む:  In one embodiment, the prediction method of the present invention comprises the following steps:
1 ) 被験者および正常人から単離された各検体より全 R NAを調製する; 2) 上記全 RNA中における変異型 PADI4遺伝子の mRNAの発現量を検出す る; 1) Prepare total RNA from each sample isolated from subjects and normal persons; 2) detecting the expression level of the mutant PADI4 gene mRNA in the total RNA;
3) 被験者と正常人における上記発現量の相違を解析し、 被験者の慢性関節リ ゥマチの発症危険度を予測する。  3) Analyze the difference in the expression level between the subject and normal person and predict the risk of developing rheumatoid arthritis in the subject.
前記方法において、 遺伝子の発現量は、 例えば、 遺伝子チップ、 cDNAァ レイ、 およびメンブレンフィル夕一から選ばれる固相化試料を用いた核酸ハイ ブリダィゼ一シヨン法、 RT- PCR法、 リアルタイム PCR法、 サブトラクシ ヨン法、 ディファレンシャル ·ディスプレイ法、 ディファレンシャル ·ハイブ リダィゼ一シヨン法、 ならびにクロスハイブリダィゼーシヨン法から選ばれる いずれか一つの方法によって検出される。  In the above method, the expression level of the gene may be determined, for example, by using a nucleic acid hybridization method using a solid-phased sample selected from a gene chip, a cDNA array, and a membrane fill sample, an RT-PCR method, a real-time PCR method, It is detected by any one of the subtraction method, the differential display method, the differential hybridization method, and the cross-hybridization method.
別な実施態様において、 本発明の予測方法は以下の工程を含む:  In another embodiment, the prediction method of the invention comprises the following steps:
1) 被験者および正常人から単離された検体中における、 変異型 PADI4蛋白質 の発現量を該蛋白質に特異的に結合しうる抗体を用いて検出する;  1) detecting the expression level of the mutant PADI4 protein in a sample isolated from a subject and a normal person using an antibody capable of specifically binding to the protein;
2) 被験者と正常人における上記発現量の相違を解析し、 該被験者の慢性関節 リゥマチの発症危険度を予測する。  2) Analyze the difference in the expression level between the test subject and normal subject, and predict the risk of developing rheumatoid arthritis in the test subject.
前記方法において、 蛋白質の発現量は、 例えば、 ウエスタンプロット法、 ド ットブロット法、 スロットブロット法、 EL I SA法、 および RI A法から選 ばれるいずれか一つの方法によって検出される。  In the above method, the expression level of the protein is detected by, for example, any one method selected from Western blotting, dot blotting, slot blotting, ELISA, and RIA.
また、 本発明は、 以下の a) ~e) からなる群より選ばれる少なくとも 1以 上を含む、慢性関節リウマチの発症危険度を予測するためのキッ卜を提供する。 a) 配列番号 20に示される塩基配列からなる変異型 PADI4遺伝子を特異的に 増幅するための、 15〜 30塩基長の連続したオリゴヌクレオチドプライマー b) 配列番号 20に示される塩基配列からなる変異型 PADI4遺伝子に特異的に ハイブリダィズし、 該遺伝子を検出するための 20-1500塩基長の連続し たポリヌクレオチドプローブ  Further, the present invention provides a kit for predicting the risk of developing rheumatoid arthritis, comprising at least one selected from the group consisting of the following a) to e). a) Mutant type consisting of the nucleotide sequence shown in SEQ ID NO: 20 A continuous oligonucleotide primer having a length of 15 to 30 nucleotides for specifically amplifying the PADI4 gene b) Mutant type consisting of the nucleotide sequence shown in SEQ ID NO: 20 A continuous polynucleotide probe of 20-1500 base length that specifically hybridizes to the PADI4 gene and detects the gene.
c) 上記 b) 記載のポリヌクレオチドプローブが固定された固相化試料 d) 配列番号 21に示されるアミノ酸配列からなる変異型 PADI4蛋白質に特異 的に結合し、 該蛋白質を検出するための抗体 c) An immobilized sample on which the polynucleotide probe described in b) above is immobilized.d) An antibody specifically binding to the mutant PADI4 protein having the amino acid sequence shown in SEQ ID NO: 21 and detecting the protein.
e) 上記 d) 記載の抗体に特異的に結合しうる二次抗体 e) secondary antibody capable of specifically binding to the antibody described in d) above
さらに、 本発明は、 配列番号 2 1に示されるアミノ酸配列からなる変異型 PADI4蛋白質に対する阻害効果を指標として、 被験物質の慢性関節リウマチ治 療薬としての効果を評価する方法を提供する。 Further, the present invention relates to a variant comprising the amino acid sequence of SEQ ID NO: 21. Provided is a method for evaluating the effect of a test substance as a therapeutic drug for rheumatoid arthritis using an inhibitory effect on PADI4 protein as an index.
前記方法は、 例えば、 以下の工程を含む:  The method includes, for example, the following steps:
1 ) アルギニン含有化合物を含む反応液に、 被験物質の添加または非添加条件 下で、 変異型 PADI4蛋白質を加える;  1) Add the mutant PADI4 protein to the reaction solution containing the arginine-containing compound, with or without the addition of the test substance;
2 )上記反応液中における反応生成物の量を定量することにより、変異型 PADI4 蛋白質の活性を測定する;  2) measuring the activity of the mutant PADI4 protein by quantifying the amount of the reaction product in the reaction solution;
3 )被験物質の添加または非添加条件下における、上記活性の相違に基づいて、 該被験物質の慢性関節リウマチ治療薬としての効果を評価する。  3) The effect of the test substance as a therapeutic agent for rheumatoid arthritis is evaluated based on the difference in the above activities under the conditions of addition or non-addition of the test substance.
さらにまた、 本発明は、 配列番号 2 1に示されるアミノ酸配列からなる変異 型 PADI4蛋白質を提供する。 該蛋白質は慢性関節リウマチの発症危険度を予測 するための予測用マーカーとして有用である。 本発明によれば、 慢性関節リゥマチ治療薬の簡便なスクリーニングが可能と なる。 また、 本発明にかかる変異型 PADI4蛋白質および該遺伝子の発現量を指 標を検出することにより、 被験者が慢性関節リウマチを発症する危険度を早期 に判定することができる。 図面の簡単な説明  Furthermore, the present invention provides a mutant PADI4 protein consisting of the amino acid sequence shown in SEQ ID NO: 21. The protein is useful as a predictive marker for predicting the risk of developing rheumatoid arthritis. ADVANTAGE OF THE INVENTION According to this invention, the simple screening of the therapeutic agent for rheumatoid arthritis is possible. In addition, the risk of developing a subject with rheumatoid arthritis can be determined at an early stage by detecting an indicator of the expression level of the mutant PADI4 protein and the gene according to the present invention. BRIEF DESCRIPTION OF THE FIGURES
図 1は、 ヒト PADI4 の Iつのハプロタイプ: V9と V18の各 mRNA安定性 (分 解度) を比較したグラフである。  Figure 1 is a graph comparing the mRNA stability (resolution) of one haplotype of human PADI4: V9 and V18.
図 2は、 (A)蛍光法による PADI4酵素活性測定の結果を示すグラフ、 および ( B ) 吸光法による PADI4酵素活性測定の結果を示すグラフである。  FIG. 2 is a graph showing (A) a result of PADI4 enzyme activity measurement by a fluorescence method, and (B) a graph showing a result of PADI4 enzyme activity measurement by an absorption method.
図 3は、 PADI4 の 2つのハプロタイプ: V9および V18の酵素活性を吸光法に より比較した結果を示すグラフである。  FIG. 3 is a graph showing the results of comparing the enzyme activities of two haplotypes of PADI4: V9 and V18 by an absorption method.
図 4は、 R A患者滑膜組織における PADI4 m A発現をみた In s i tu RT-PCR の結果を示す写真である。 図中右の 2つは陰性対照である。  FIG. 4 is a photograph showing the results of In situ RT-PCR in which the expression of PADI4 mA in RA patient synovial tissues was observed. The two right ones in the figure are negative controls.
図 5は、 マウスコラーゲン誘導関節炎 (CIA) 群と正常 (normal) 群における 滑膜組織および脾臓組織における mPADI4遺伝子発現量を示すグラフである。 図 6は、 (A)抗 PADI4抗体を用いた R A患者滑膜での免疫組織染色像、およ び(B) PADI4を特異的に認識する抗体と PADIの全てのサブタイプを認識する 抗体の両者を用いた OA患者滑膜での免疫組織染色像を示す写真である。 FIG. 5 is a graph showing the expression levels of mPADI4 gene in synovial tissue and spleen tissue in the mouse collagen-induced arthritis (CIA) group and the normal group. Figure 6 shows (A) immunohistochemical staining of RA patient synovium with anti-PADI4 antibody, and (B) is a photograph showing immunohistochemical staining of the synovium of an OA patient using both an antibody that specifically recognizes PADI4 and an antibody that recognizes all subtypes of PADI.
図 7は、 (A)抗 PADI4抗体を用いた R A患者滑膜での免疫組織染色像、 およ び (B ) 抗シトルリン化タンパク質抗体を用いた RA患者滑膜での免疫組織染 色像を示す写真である。  Figure 7 shows (A) immunohistochemical staining of RA patient synovium using anti-PADI4 antibody and (B) immunohistochemical staining of RA patient synovium using anti-citrullinated protein antibody. It is a photograph shown.
図 8は、 ヒト PADI4 V9と PADI4 V18の mRNAの安定性を比較したグラフであ る。 本明細書は、 本願の優先権の基礎である特願 2003-12738 号および特願 2003-12774号の明細書に記載された内容を包含する。 発明を実施するための最良の形態  FIG. 8 is a graph comparing the stability of human PADI4 V9 and PADI4 V18 mRNA. This description includes part or all of the contents as disclosed in the description of Japanese Patent Application No. 2003-12738 and Japanese Patent Application No. 2003-12774, which are the basis of the priority of the present application. BEST MODE FOR CARRYING OUT THE INVENTION
1 . ぺプチジルアルギニン ·ディミナーゼ ·タイプ I V  1. Peptidyl arginine diminase type IV
I . 1 ぺプチジルアルギニン ·ディミナーゼ ·タイプ I V (PAD 14)  I.1 Peptidylarginine diminase type IV (PAD 14)
ぺプチジルアルギニン ·ディミナーゼ (以下、 「PADI」 と記載する。) は、 蛋 白質内のアルギニン残基をシ卜ルリンに変換し、 ぺプチジルシ卜ルリンを生成 させる酵素である。 PADI には複数のサブタイプが存在し、 ヒトではタイプ I、 Peptidyl arginine diminase (hereinafter referred to as “PADI”) is an enzyme that converts arginine residues in a protein to citrulline to produce peptidyl citrulline. There are several subtypes of PADI, including type I and
II、 II I、 Vの 4つのサブタイプ遺伝子が G e n B a n k上に登録されているが、 その詳細な機能は不明である。 マウスでは、 タイプ I、 I I、 III、 I Vの 4つの サブタイプが報告されており、 このうちタイプ I Vはヒト PADI4のマウスォー ソログである。 Four subtype genes, II, II I, and V, are registered on GenBank, but their detailed functions are unknown. In mice, four subtypes have been reported, type I, II, III, and IV, of which type IV is the mouse ortholog of human PADI4.
本発明者らは、 R A患者の病変組織において PADI4蛋白質や PADI4遺伝子 (m R NA) が特異的に高発現し、 シトルリン化ペプチドが高レベルで検出される ことを確認した。 シトルリン化ペプチドは自己抗体 (抗シトルリン化ペプチド 抗体) の産生を介して、 R A病変の原因となりうるものである。 したがって、 PADI4蛋白質またはその遺伝子の発現量や活性に対する阻害効果を指標として、 被験物質の慢性関節リゥマチ治療薬としての効果を評価することが可能となる。  The present inventors have confirmed that the PADI4 protein and the PADI4 gene (mRNA) are specifically highly expressed in the lesion tissue of RA patients, and that the citrullinated peptide is detected at a high level. Citrullinated peptides can cause RA lesions through the production of autoantibodies (anti-citrullinated peptide antibodies). Therefore, it is possible to evaluate the effect of a test substance as a therapeutic agent for rheumatoid arthritis using the inhibitory effect on the expression level and activity of the PADI4 protein or its gene as an index.
( 1 ) 本発明の PADI4遺伝子  (1) PADI4 gene of the present invention
本発明の評価方法で用いられる遺伝子には、 PADI4遺伝子のほか、 そのォー ソログ遺伝子も含まれる。 「ォ一ソログ遺伝子」 とは、 「共通の祖先の遺伝子か ら進化した、 同じ機能を有する、 異なる種の遺伝子」 であって、 例えば、 PADI4 のォーソログとしては、 マウス PADI4等が挙げられる。 以下、 PADI4遺伝子と そのォーソログ遺伝子をまとめて、 「本発明の PADI4遺伝子」 という。 The genes used in the evaluation method of the present invention include not only the PADI4 gene but also its orthologous gene. "Osolog gene" means "gene of common ancestry" Genes of different species having the same function and evolved from them ”. For example, orthologs of PADI4 include mouse PADI4. Hereinafter, the PADI4 gene and its ortholog gene are collectively referred to as “PADI4 gene of the present invention”.
前記遺伝子の由来は特に限定されないが、 哺乳動物由来のものが好ましく、 霊長類、 ゲッ歯動物由来のものがより好ましい。 特に、 最も好適な例として、 ヒト PADI4遺伝子およびマウス PADI4遺伝子を挙げることができる。  The origin of the gene is not particularly limited, but is preferably derived from mammals, more preferably primates or rodents. In particular, the most preferred examples include the human PADI4 gene and the mouse PADI4 gene.
本発明の PADI4遺伝子のうち、 ヒト PADI4をコードする c D NAの配列を配 列番号 1 6に示すが、 この配列に限定されず、 ヒト PADI4をコードする限り、 その遺伝子はヒト PADI4遺伝子に含まれるものとする。例えば、本発明者らは、 ヒト PADI4には複数のハプロタイプが存在することを確認したが、 これらハプ 口タイプもヒト PADI4遺伝子に含まれる。 同様に、 本発明にかかるォ一ソログ 遺伝子のうち、 マウス PADI4をコードする c D NAの配列を配列番号 1 8に示 すが、 この配列に限定されず、 マウス PADI4をコードする限り、 その遺伝子は マウス PADI4遺伝子に含まれるものとする。  Among the PADI4 genes of the present invention, the sequence of cDNA encoding human PADI4 is shown in SEQ ID NO: 16, but is not limited to this sequence, and as long as it encodes human PADI4, the gene is included in the human PADI4 gene. Shall be For example, the present inventors have confirmed that human PADI4 has a plurality of haplotypes, and these haplotypes are also included in the human PADI4 gene. Similarly, among the orthologous genes according to the present invention, the sequence of the cDNA encoding mouse PADI4 is shown in SEQ ID NO: 18, but is not limited to this sequence. Is included in the mouse PADI4 gene.
なお本明細書中において、 「遺伝子」 という用語には、 D N Aのみならずそ の mR NA、 c D NAおよび c R NAも含むものとする。 したがって、 本発明 の遺伝子には、 これらの D NA、 mR NA、 c D NA, および c R NAの全て が含まれる。 また、 配列表の塩基配列はすべて D NA配列として記載するが、 該配列が R NAを示す場合は、 配列表中の塩基記号 「t」 は 「u」 に読み替え るものとする。  In this specification, the term “gene” includes not only DNA but also its mRNA, cDNA and cDNA. Therefore, the gene of the present invention includes all of these DNAs, mRNAs, cDNAs, and cDNAs. In addition, all nucleotide sequences in the sequence listing are described as DNA sequences. When the sequence indicates RNA, the base symbol “t” in the sequence listing is replaced with “u”.
( 2 ) 本発明の PADI4蛋白質 (2) PADI4 protein of the present invention
本発明の評価方法で用いられる蛋白質(PADI4蛋白質) には、 PADI4遺伝子に よってコードされる PADI4蛋白質のほか、 前記ォーソログ遺伝子によってコ一 ドされる蛋白質も含まれる。以下、これらの蛋白質をまとめて、「本発明の PADI4 蛋白質」 という。  The protein (PADI4 protein) used in the evaluation method of the present invention includes, in addition to the PADI4 protein encoded by the PADI4 gene, the protein encoded by the orthologous gene. Hereinafter, these proteins are collectively referred to as “PADI4 protein of the present invention”.
前記蛋白質の由来は特に限定されないが、 哺乳動物由来のものが好ましく、 霊長類、 ゲッ歯動物由来のものがより好ましい。 特に、 本発明における最も好 適な例として、 ヒト PADI4蛋白質およびマウス PADI4蛋白質を挙げることがで きる。 本発明の蛋白質のうち、ヒト PADI4のアミノ酸配列を配列番号 1 7に示すが、 この配列に限定されず、 該配列において 1または数個のアミノ酸が欠失、 置換 または付加された配列で示されるペプチドもヒト PADI4としての機能を有する 限り、 ヒト PADI4蛋白質に含まれるものとする。 同様に、 マウス PADI4のアミ ノ酸配列を配列番号 1 8に示すが、 この配列に限定されず、 該配列において 1 または数個のアミノ酸が欠失、 置換または付加された配列で示されるぺプチド もマウス PADI4としての機能を有する限り、 マウス PADI4蛋白質に含まれるも のとする。 1 . 2 変異型べプチジルアルギニン ·ディミナ一ゼ ·タイプ I V The origin of the protein is not particularly limited, but is preferably derived from mammals, and more preferably derived from primates and rodents. In particular, the most preferred examples of the present invention include human PADI4 protein and mouse PADI4 protein. Among the proteins of the present invention, the amino acid sequence of human PADI4 is shown in SEQ ID NO: 17, but is not limited thereto, and is represented by a sequence in which one or several amino acids have been deleted, substituted or added. As long as the peptide also has a function as human PADI4, it is included in the human PADI4 protein. Similarly, the amino acid sequence of mouse PADI4 is shown in SEQ ID NO: 18, but is not limited thereto, and the peptide is represented by a sequence in which one or several amino acids have been deleted, substituted or added in the sequence. As long as they also have a function as mouse PADI4, they are included in the mouse PADI4 protein. 1.2 Mutant beptidyl arginine, diminase, type IV
本発明者らは、 ヒト PADI4遺伝子上に慢性関節リゥマチ患者に高い頻度で現 れる複数の遺伝子多型が存在し、 これらがほぼ完全連鎖(98. 9〜100%) の関係 にあって、 一つのハプロタイプを構成することを確認した。 さらに、 このハプ 口タイプは、 正常な PADI4蛋白質とはアミノ酸配列や酵素活性が異なる変異型 PADI4蛋白質をコードし、 その違いが R Aの発症と深い関連を有することを確 認した。  The present inventors have found that there are a plurality of polymorphisms that frequently appear in patients with rheumatoid arthritis on the human PADI4 gene, and these are almost completely linked (98.9 to 100%). To make up one haplotype. Furthermore, it was confirmed that this haplotype encodes a mutant PADI4 protein having a different amino acid sequence and enzyme activity from a normal PADI4 protein, and that the difference is closely related to the onset of RA.
すなわち、 本発明にかかる 「変異型 PADI4蛋白質」 とは、 配列番号 2 1で示 されるアミノ酸配列を有する蛋白質であって、正常な PADI4のアミノ酸配列(配 列番号 1 7 ) とは、 第 5 5番アミノ酸(S e rから G 1 yに置換)、 第 8 2番ァ ミノ酸(A 1 aから V a 1に置換)、第 1 1 2番アミノ酸(A 1 aから G 1 yに 置換)が異なっている。 この違いにより、変異型 PADI4蛋白質は、正常な PAD 蛋白質よりも酵素活性が高く、 R A病変の一因となるシトルリン化蛋白質の生 成を亢進させる。  That is, the “mutant PADI4 protein” according to the present invention is a protein having the amino acid sequence represented by SEQ ID NO: 21, and the amino acid sequence of normal PADI4 (SEQ ID NO: 17) Amino acid 5 (substituted from Ser to G1y), Amino acid 82 (substituted from A1a to Va1), Amino acid 112 (substituted from A1a to G1y) Are different. Due to this difference, mutant PADI4 protein has higher enzymatic activity than normal PAD protein and enhances the production of citrullinated protein that contributes to RA lesions.
また、 本発明にかかる 「変異型 PADI4遺伝子」 とは、 配列番号 2 0で示され る c D NA配列を有し、 正常な PADI4遺伝子とは、 ゲノム配列上、 第 3イント ロンの 2 1 3 6番目の塩基(シトシンからチミンに置換)、第 2ェクソンの 7 1 番目の塩基(アデニンからグァニンに置換)、第 2ェクソンの 1 5 3番目の塩基 (シトシンからチミンに置換)、第 3ェクソンの 6 2番目の塩基(シトシンから グァニンに置換)、および第 4ェクソンの 9番目の塩基(シトシンからチミンに 置換:この置換はアミノ酸置換を伴わない)が異なっている。この違いにより、 変異型 PADI4遺伝子の mR N Aは、 正常な PADI4遺伝子の mR N Aよりも安定 性が高く、 R A病変の一因となるシトルリン化蛋白質の生成を亢進させる可能 性がある。 2 被験物質の慢性関節リゥマチ治療薬としての効果を評価する方法 Further, the “mutant PADI4 gene” according to the present invention has a cDNA sequence represented by SEQ ID NO: 20. A normal PADI4 gene is defined as the third intron of the third intron in the genome sequence. 6th base (substitute cytosine for thymine), 2nd exon 7 1st base (substitute adenine for guanine), 2nd exon 15 3rd base (substitute cytosine for thymine), 3rd exon The second base (substitution of cytosine to guanine) of the fourth and the ninth base of exon 4 (substitution of cytosine to thymine: this substitution does not involve amino acid substitution) are different. Due to this difference, The mRNA of the mutant PADI4 gene is more stable than the mRNA of the normal PADI4 gene, and may increase the production of citrullinated proteins that contribute to RA lesions. 2 Methods for evaluating the effects of test substances as therapeutics for rheumatoid arthritis
本発明は、 本発明の PADI4遺伝子、 または該遺伝子にコードされる蛋白質に 対する阻害効果を指標として、 被験物質の慢性関節リゥマチ治療薬としての効 果を評価する方法を提供する。  The present invention provides a method for evaluating the effect of a test substance as a therapeutic agent for rheumatoid arthritis, using an inhibitory effect on the PADI4 gene of the present invention or the protein encoded by the gene as an index.
前記方法は、 1つの被験物質について、 その投与 (添加) および非投与条件 下における、 阻害効果を比較評価するものであってもよいし、 2つ以上の被験 物質についての同様な比較評価であってもよい。 あるいは、 前記遺伝子や蛋白 質の発現量や活性と R A治療効果との相関関係が経験的に確立されれば、 その 関係に基づいて、 比較対照なしに絶対評価するものであってもよい。  The above method may be a method for comparatively evaluating the inhibitory effect of one test substance under the administration (addition) and non-administration conditions, or a similar comparative evaluation of two or more test substances. You may. Alternatively, if a correlation between the expression level or activity of the gene or protein and the RA therapeutic effect is established empirically, an absolute evaluation without a comparative control may be performed based on the relationship.
本発明の評価方法において、 本発明の PADI4遺伝子や PADI4蛋白質に対する 阻害効果は、 本発明の PADI4遺伝子や PADI4蛋白質の発現量を指標として評価 してもよいし、 本発明の PADI4遺伝子や PADI4蛋白質の活性 (安定性含む) を 指標として評価しても良い。 また、 評価系は in vivo系であってもよいし、 in vi tro 系であってもよい。 以下、 本発明の評価方法の具体的な実施方法につい て説明する。  In the evaluation method of the present invention, the inhibitory effect on the PADI4 gene or PADI4 protein of the present invention may be evaluated using the expression level of the PADI4 gene or PADI4 protein of the present invention as an index, or the inhibitory effect of the PADI4 gene or PADI4 protein of the present invention. Activity (including stability) may be evaluated as an index. The evaluation system may be an in vivo system or an in vitro system. Hereinafter, a specific implementation method of the evaluation method of the present invention will be described.
2 . 1 本発明の PADI4遺伝子の発現量を指標とした評価方法 2.1 Evaluation method using expression level of PADI4 gene of the present invention as an index
本発明の PADI4遺伝子の発現量を指標とした in vivoにおける評価方法は、 例えば下記の工程を含む。  The in vivo evaluation method of the present invention using the PADI4 gene expression level as an index includes, for example, the following steps.
工程 1 :動物を被験物質の投与または非投与条件下で飼育する。 Step 1: Animals are bred under the conditions of administration or non-administration of the test substance.
工程 2 :上記動物の血液または細胞中における本発明の PADI4遺伝子の発現量 を検出する。 Step 2: detecting the expression level of the PADI4 gene of the present invention in the blood or cells of the above animal.
工程 3 :被験物質の投与または非投与条件下における、 上記発現量の相違に基 づいて、 該被験物質の R A治療薬としての効果を評価する。 工程 1 :動物の飼育 本発明の評価方法で用いられる 「動物」 は特に限定されないが、 R A病態を 呈するモデル動物が好ましい。 そのような動物は市販のものであっても、 公知 の方法にしたがって作製されたものでもよい。 そのような R A病態を呈するモ デルとしては、例えば、自然発症関節炎モデルである、 MRLマウス関節炎、 NZB/KN マウス関節炎、 SKGマウス関節炎;誘導性関節炎モデルである、 ラットアジュ バント関節炎、 ラット ·マウスコラーゲン関節炎 (CIAラット 'マウス)、 プリ スタン関節炎、 SCID細胞移入関節炎、 SCID マウス組織移植関節炎;および、 HTLV-1 トランスジエニックマウス、 IL-1レセプ夕一アンタゴニスト K0マウス 等を挙げることができる (関節炎モデル 日本医学館、監修 ·京極方久、編集 · 安倍千之、 澤井高志 参照)。 なかでも一般的なのは、 RL マウス関節炎、 ラッ トアジュバント関節炎、 ラット ·マウスコラーゲン関節炎、 およびプリスタン 関節炎である。 Step 3: Evaluate the effect of the test substance as a therapeutic agent for RA on the basis of the difference in the expression level under the conditions of administration or non-administration of the test substance. Process 1: Animal rearing The “animal” used in the evaluation method of the present invention is not particularly limited, but a model animal exhibiting RA pathology is preferable. Such animals may be commercially available or may be produced according to known methods. Models exhibiting such RA pathology include, for example, spontaneous arthritis model, MRL mouse arthritis, NZB / KN mouse arthritis, SKG mouse arthritis; inducible arthritis model, rat adjuvant arthritis, rat mouse collagen Arthritis (CIA rat 'mouse), pristine arthritis, SCID cell-transplanted arthritis, SCID mouse tissue transplant arthritis; and HTLV-1 transgenic mice, IL-1 receptor Yuichi antagonist K0 mice, etc. Model supervised by Nippon Medical Center, Hakuyo Kyogoku, editor, Chiyuki Abe, Takashi Sawai). The most common are RL mouse arthritis, rat adjuvant arthritis, rat-mouse collagen arthritis, and pristane arthritis.
前記動物は、 被験物質の投与または非投与条件下で適当な期間飼育を行う。 動物への被験物質の投与量は特に限定されず、 被験物質の性状や動物の体重に 合わせて、 適宜用量を設定すればよい。 また、 動物への被験物質の投与方法お よび投与期間も特に限定されず、 被験物質の性状に合わせて、 適宜その投与経 路と投与期間を設定すればよい。 工程 2 :本発明の PADI4遺伝子の検出  The animals are bred for an appropriate period under the conditions of administration or non-administration of the test substance. The dose of the test substance to the animal is not particularly limited, and an appropriate dose may be set according to the properties of the test substance and the weight of the animal. The method and period of administration of the test substance to the animal are also not particularly limited, and the administration route and the administration period may be appropriately set according to the properties of the test substance. Step 2: Detection of the PADI4 gene of the present invention
次に、 被験物質の投与または非投与条件下で飼育された動物から血液または 細胞を単離し、 該血液または細胞中の本発明の PADI4遺伝子の発現量を検出す る。  Next, blood or cells are isolated from animals bred under the conditions of administration or non-administration of the test substance, and the expression level of the PADI4 gene of the present invention in the blood or cells is detected.
検出対象とする細胞としては、 本発明の PADI4遺伝子が高発現している細胞 が好ましく、 したがって、 好中球や好酸球等の血液由来細胞、 滑膜細胞、 脾臓 細胞、 および腹腔浸潤細胞が好ましい。 本発明の PADI4遺伝子の検出方法とし ては、 例えば、 単離された血液または細胞からまず全 R NAを抽出し、 該全 R N A中における mR N Aの発現量を検出する方法を挙げることができる。  As the cells to be detected, cells in which the PADI4 gene of the present invention is highly expressed are preferable. preferable. The method for detecting the PADI4 gene of the present invention includes, for example, a method of first extracting total RNA from isolated blood or cells and detecting the expression level of mRNA in the total RNA.
( 1 ) 全 R NAの抽出 (1) Extraction of total RNA
全 R NAの抽出は、 公知の方法にしたがい、 単離された血液または細胞より RNA抽出用溶媒を用いて抽出する。 該抽出溶媒としては、 例えば、 フエノー ル等のリポヌクレアーゼを不活性化する作用を有する成分を含むもの(例えば、Extraction of total RNA is performed according to known methods from isolated blood or cells. Extract using the RNA extraction solvent. Examples of the extraction solvent include those containing a component having an action to inactivate liponucleases such as phenol (for example,
TRIzol試薬:ギブコ ·ピーアールエル社製等) が好ましい。 RN Aの抽出方法 は特に限定されず、例えば、チォシアン酸グァニジン ·塩化セシウム超遠心法、 チォシアン酸グァニジン ·ホットフエノール法、 グァニジン塩酸法、 酸性チォ シアン酸グァニジン'フエノール 'クロ口ホルム法(Chomczynski, P. and Sacchi, N. , (1987) Anal. Bioche . , 162, 156-159) 等を採用することができる。 なか でも、 酸性チォシアン酸グァニジン ·フエノール ·クロ口ホルム法が好適であ る。 TRIzol reagent: manufactured by Gibco PRL) is preferable. The method of extracting RNA is not particularly limited. For example, guanidine thiocyanate / cesium chloride ultracentrifugation method, guanidine thiocyanate / hot phenol method, guanidine hydrochloride method, guanidine acid thiocyanate phenol / clonal form method (Chomczynski, P. and Sacchi, N., (1987) Anal. Bioche., 162, 156-159) and the like can be employed. Of these, the guanidine acid thiocyanate / phenol / chloroform method is preferred.
抽出された全 RNAは、 必要に応じてさらに mRNAのみに精製して用いて もよい。 精製方法は特に限定されないが、 真核細胞の細胞質に存在する mRN Aの多くは、その 3'末端にポリ (A)配列を持っため、 この特徴を利用して、 例えば、 以下のように実施することができる。 まず、 抽出した全 RN Aにピオ チン化オリゴ (dT) プローブを加えてポリ (A) +RNAを吸着させる。 次 に、 ストレプトアビジンを固定化した常磁性粒子担体を加え、 ピオチン/スト レプトアビジン間の結合を利用して、 ポリ (A) +RNAを捕捉させる。 洗浄 操作の後、 最後にオリゴ (dT) プローブからポリ (A) +RNAを溶出する。 この方法のほか、 オリゴ (dT) セルロースカラムを用いてポリ (A) +RN Aを吸着させ、 これを溶出して精製する方法も採用してもよい。 溶出されたポ リ (A) +RNAは、 さらに、 ショ糖密度勾配遠心法等により分画してもよい。 The extracted total RNA may be further purified only to mRNA if necessary. Although the purification method is not particularly limited, most mRNAs present in the cytoplasm of eukaryotic cells have a poly (A) sequence at the 3 ′ end, and this characteristic can be used, for example, as follows. can do. First, a biotinylated oligo (dT) probe is added to all the extracted RNA to adsorb poly (A) + RNA. Next, a paramagnetic particle carrier on which streptavidin is immobilized is added, and poly (A) + RNA is captured using the binding between biotin and streptavidin. After washing, poly (A) + RNA is eluted from the oligo (dT) probe. In addition to this method, a method of adsorbing poly (A) + RNA using an oligo (dT) cellulose column and eluting the same may be used for purification. The eluted poly (A) + RNA may be further fractionated by sucrose density gradient centrifugation or the like.
(2) 本発明の PADI4遺伝子の検出 (2) Detection of PADI4 gene of the present invention
次に、 被験物質の投与または非投与条件下における、 全 RNA中の本発明の PADI4遺伝子の発現量を検出する。 遺伝子の発現量は、 得られた全 RNAより c RNAまたは cDNAを調製し、 これを適当な標識化合物でラベルすること により、 そのシグナル強度として検出することができる。 以下、 遺伝子の発現 量の検出方法について、 i) 固相化試料を用いた解析方法、 ii) RT-PCR 法 (リアルタイム PCR法) 、 iii) その他の解析方法に分けて、 具体的に説明 する。 ii )) 固固相相化化試試料料をを用用いいたた解解析析方方法法 Next, the expression level of the PADI4 gene of the present invention in the total RNA under the conditions of administration or non-administration of the test substance is detected. The gene expression level can be detected as signal intensity by preparing cRNA or cDNA from the obtained total RNA and labeling it with an appropriate labeling compound. The method of detecting the expression level of the gene is described below in detail by dividing it into i) an analysis method using a solid-phased sample, ii) RT-PCR method (real-time PCR method), and iii) other analysis methods. . ii)) Analytical method using solid-phase sample preparation
公公知知のの遺遺伝伝子子をを固固定定ししたた固固相相化化試試料料にに、、 投投与与ままたたはは非非投投与与条条件件下下ににおおけけるる標標 識識ししたた cc DD NNAAままたたはは cc RR NNAA ((以以下下、、 「「標標識識ププロローーブブ」」 とと記記載載すするる。。)) をを、、 同同 じじ条条件件でで別別個個にに、、ああるるいいはは混混合合ししてて同同時時ににハハイイブブリリダダィィズズささせせるる((BBrroowwnn,, PP.. 00.. eett aall.. ((11..999999)) NNaattuurree ggeenneett.. 2211,, ssuuppppll iimmeenntt,, 3333--3377))。。 前前記記標標識識ププロローーブブはは、、 本本発発明明のの PPAADDII44のの mmRR NNAAククロロ一一ンンででもも、、 発発現現ししてていいるる全全ててのの mmRR NNAAをを標標識識 ししたたももののででももよよいい。。 ププロローーブブ作作製製ののたためめのの出出発発材材料料ととししててはは、、 精精製製ししてていいなないい mmRR NN AAをを用用いいててももよよいいがが、、 前前述述のの方方法法でで精精製製ししたたポポリリ ((AA)) ++ RR NN AAをを用用いい るるここととががよよりり好好ままししいい。。 以以下下、、 各各種種固固相相化化試試料料をを用用いいたた解解析析方方法法ににつついいてて説説明明 すするる。。  A solid-phased sample material in which a known genetic gene has been fixed and fixed to the solid-phase-phased sample material, with or without administration. Cc DD NNAA or cc RR NNAA (hereinafter referred to as "" "" "). Under the same conditions, separately and / or mixed together at the same time so that they can be combined at the same time (( BBrroowwnn ,, PP .. 00 .. eett aall .. ((11..999999)) NNaattuurree ggeenneett .. 2211 ,, ssuupppplliimmeenntt ,, 3333--3377)). . The mark labeling probe described above, even with the mmRR NNAA chlorochloroform of PPAADDII44 of the present invention, labels all mmRR NNAA expressing and expressing. You may be aware of it. . As a starting and starting material for making prop rooves, it is possible to use mmRR NN AA, which may or may not be purified. However, it is better to use the poporily ((AA)) ++ RR NN AA purified and produced by the method described above. . In the following, the analysis and analysis method using various types of test samples for solid-phase solidification are explained. .
aa )) 遺遺伝伝子子チチッッフフ:: aa)) Genetic Gene Chichipfoff ::
本本発発明明でで用用いいらられれるる遺遺伝伝子子チチッッププはは、、 検検出出対対象象ででああるる遺遺伝伝子子がが固固相相化化さされれてて いいるるももののででああれればば、、市市販販ののももののででああっっててもも、、公公知知のの方方法法((LLiippsshhuuttzz,, RR.. JJ.. eett aall.. ((11999999)) NNaattuurree ggeenneett.. 2211,, ssuuppppll iimmeenntt,, 2200--2244)) にに基基づづきき作作製製さされれたたももののででああ つつててももよよいい。。 遺遺伝伝子子チチッッププにによよるる検検出出はは、、 常常法法ににししたたががっってて実実施施すするるここととががでで ききるる。。 例例ええばば、、 ァァフフィィメメトトリリククスス社社製製チチッッププをを用用いいるる場場合合ででああれればば、、 製製品品にに添添 付付さされれたたププロロトトココ一一ルルににししたたががいい、、 ピピオオチチンン標標識識ししたた cc RR NN AAププロローーブブをを調調製製 すするる。。 次次いいでで、、 該該ププロロトトココーールルににししたたががいいハハイイブブリリダダィィゼゼーーシシヨヨンンをを行行いい、、 ァァ ビビジジンンにによよるる発発光光をを検検出出、、 解解析析すすれればば遺遺伝伝子子のの発発現現量量をを求求めめるるここととががででききるる。。
Figure imgf000016_0001
The genetic gene chip used in the present invention is a method in which a genetic gene, which is an object to be detected and detected, is solid-phase-phased. If it is a thing that is sold, even if it is a thing that is marketed and sold, it is a publicly known method ((LLiippsshhuuttzz ,, RR .. JJ .. eett aall .. ((11999999)) NNaattuurree ggeenneett .. 2211 ,, ssuuppppll iimmeenntt ,, 2200--2244)). . Detection and detection by genetic genetic test can be performed in accordance with the conventional method. . For example, if a chip manufactured by Affiliate Metrics Rix Co., Ltd. is used, the protocol attached to the product may be used. Prepare a cc RR NN AA probe labeled with pipiootintin and prepare it. . Next, follow the procedure described in the above, and perform the following procedure, and perform the following steps. By detecting, analyzing and analyzing the emitted light, it is possible to obtain the amount of expression of the genetic gene. .
Figure imgf000016_0001
本発明で用いられるアレイまたはメンブレンフィル夕一は、 検出対象である 遺伝子が固相化されているものであれば、 市販のもの (例えば、 インテリジー ン (宝酒造社製)、 フィルター製マイクロアレイ アトラスシステム (クローン テック社製等))であっても、公知の方法に基づいて作製されたものであっても よい。 固相化される遺伝子は、 G e n B a n k等の配列情報をもとに作製され たプライマーで逆転写酵素反応や P C Rを実施することによりクローン化され た c D NAまたは R T— P C R産物を用いる。  The array or membrane fill used in the present invention may be a commercially available array (for example, Intelligin (Takara Shuzo), a filter microarray atlas system) as long as the gene to be detected is immobilized. (Clontech, etc.)) or may be produced based on a known method. The gene to be immobilized is a cDNA or RT-PCR product cloned by performing a reverse transcriptase reaction or PCR with primers created based on sequence information such as Gen Bank. .
アレイを用いた検出では、 逆転写酵素反応でポリ (A) + R NAから c D N Aを作製する際に、 蛍光色素 (例えば、 C y 3、 C y 5等) で標識された d— U T P等を加えることにより標識プローブを調製する。 被験物質の投与条件下 におけるポリ (A) +RNAと被験物質の非投与条件下におけるポリ (A) +R NAはそれぞれ異なる色素で標識し、 両者を混合してハイブリダィズさせ、 蛍 光シグナル検出器を用いて検出する。 得られる蛍光シグナルは投与および非投 与条件下における遺伝子発現量の相対比を示す (Brown, P. 0. et al. (1999) Nature genet. 1, s卿 liment、 33 - 37)。 In the detection using an array, when producing cDNA from poly (A) + RNA by reverse transcriptase reaction, d- UTP, etc. labeled with a fluorescent dye (for example, Cy3, Cy5, etc.) To prepare a labeled probe. Test substance administration conditions In the above, poly (A) + RNA and poly (A) + RNA under the non-administration condition of the test substance are respectively labeled with different dyes, the two are mixed and hybridized, and detected using a fluorescent signal detector. The resulting fluorescence signal indicates the relative ratio of gene expression under the administration and non-administration conditions (Brown, P. 0. et al. (1999) Nature genet. 1, s liment, 33-37).
メンブレンフィルターを用いた検出では、 逆転写酵素反応でポリ (A) +R NAから cDNAを作製する際に、 放射性同位元素 (例えば、 32P、 33P) で標 識された d— C T P等を加えることにより標識プローブを調製し、 常法により ハイブリダィゼーションを行う。 市販のフィルター製マイクロアレイである、 アトラスシステム (クローンテック社製) を用いてハイブリダィゼーションぉ よび洗浄を行った後、 解析装置 (例えば、 アトラスイメージ:クローンテック 社製等) を用いて検出、 解析を行う。 ii) RT— PCR法 (リアルタイム PC R法) In the detection using a membrane filter, in making cDNA from poly (A) + R NA with reverse transcriptase reaction, a radioisotope (e.g., 32 P, 33 P) at the-labeled the d-CTP, etc. A labeled probe is prepared by the addition, and hybridization is performed by a conventional method. After performing hybridization and washing using an atlas system (manufactured by Clontech), which is a commercially available filter microarray, detection is performed using an analyzer (for example, atlas image: manufactured by Clontech). Perform analysis. ii) RT—PCR method (Real-time PCR method)
RT— PCR法やその 1つであるリアルタイム PCR (TaciMan PCR) 法は微 量な DN Aを高感度かつ定量的に検出できるという点で本発明の評価方法に好 適である。 リアルタイム PCR (TaqMan PCR) 法では、 5' 端を蛍光色素 (レポ 一夕一) で、 3' 端を蛍光色素 (クェンチヤ一) で標識した、 目的遺伝子の特定 領域にハイプリダイズするオリゴヌクレオチドプローブが使用される。 該プロ ーブは、 通常の状態ではクェンチヤ一によつてレポーターの蛍光が抑制されて いる。 この蛍光プローブを目的遺伝子に完全にハイブリダィズさせた状態で、 その外側から Tad DNAポリメラ一ゼを用いて P CRを行う。 Taci DNAポリメラ —ゼによる伸長反応が進むと、 そのェキソヌクレァーゼ活性により蛍光プロ一 ブが 5' 端から加水分解され、 レポ一夕一色素が遊離し、 蛍光を発する。 リア ルタイム PC R法は、 この蛍光強度をリアルタイムでモニタリングすることに より、 踌型 DN Aの初期量を正確に定量することができる。 iii) その他の解析方法  The RT-PCR method and one of them, the real-time PCR (TaciMan PCR) method, are suitable for the evaluation method of the present invention in that they can detect minute amounts of DNA with high sensitivity and quantitatively. In the real-time PCR (TaqMan PCR) method, an oligonucleotide probe that hybridizes to a specific region of the target gene and has a 5 'end labeled with a fluorescent dye (Repo Ichiichi) and a 3' end labeled with a fluorescent dye (Quenchia I) is used. used. In this probe, the fluorescence of the reporter is normally suppressed by quenching. With the fluorescent probe completely hybridized to the target gene, PCR is performed from outside using Tad DNA polymerase. As the elongation reaction of the Taci DNA polymerase progresses, the exonuclease activity hydrolyzes the fluorescent probe from the 5 'end, releasing the repo dye overnight to emit fluorescence. The real-time PCR method can accurately quantify the initial amount of type I DNA by monitoring the fluorescence intensity in real time. iii) Other analysis methods
上記以外に、 遺伝子発現量を解析する方法としては、 例えば、 サブトラクシ ヨン法 (Sive, H. L. and John, T. St. (1988) Nucleic Acids Research 16, 10937、 Wang, Z. , and Brown, D. D. (1991) Proc. Natl. Acad. Sci. U. S. A. 88, 11505- 11509)、ディファレンシャル 'ディスプレイ法 (Liang, P., and Pardee, A. B. (1992) Science 257, 967 - 971、 Liang, P. , Averboukh, L. , Keyomarsi, K. , Sager, R. , and Pardee, A. B. (1992) Cancer Research 52, 6966 - 6968)、 デ ィファレンシャル 'ハイブリダィゼーシヨン法 (John, T. St., and Davis, R. W. Cell (1979) 16, 443-452)、 また、 適当なプローブを用いたクロスハイプリ ダイゼ一シヨン法(" Molecular Cloning, A Laboratory Manual" Maniatis, T., Fritsch, E. F. , Sambrook, J. (1982) Cold Spring Harbor Laboratory Press) 等を挙げることができる。 In addition to the above, methods for analyzing gene expression levels include, for example, the subtraction method (Sive, HL and John, T. St. (1988) Nucleic Acids Research 16, 10937, Wang, Z., and Brown, DD (1991) Proc. Natl. Acad. Sci. USA 88, 11505-11509), Differential 'Display Method (Liang, P., and Pardee, AB (1992) Science 257, 967-971, Liang, P., Averboukh, L., Keyomarsi, K., Sager, R., and Pardee, AB (1992) Cancer Research 52, 6966-6968), differential 'hybridization' Method (John, T. St., and Davis, RW Cell (1979) 16, 443-452) and a cross-hybridization method using an appropriate probe ("Molecular Cloning, A Laboratory Manual" Maniatis, T. , Fritsch, EF, Sambrook, J. (1982) Cold Spring Harbor Laboratory Press).
a) サブトラクシヨンクローニング法: a) Subtraction cloning method:
特定の細胞に特異的に発現する遺伝子の c DNAを取得し、 該 c DNAをプ ローブとして c DN Aライブラリ一をスクリーニングすることにより遺伝子を クローニングする方法である。 サブトラクシヨンの方法としては、 全 RNAか ら一本鎖 c DNAを作製し、 これと別の細胞から得られた全 RN Aをハイプリ ダイズさせた後、 ハイドロキシアパタイトカラムでハイブリダィズしなかった 一本鎖 DNAを単離し、 この c DNAから c DN Aライブラリ一を作製する方 法(バイオマニュアルシリーズ 3、遣伝子クローニング実験法、羊土社 (1993), カレン卜 'プロトコールズ ·イン 'モレキュラー ·バイオロジー) や、 cDN Aライブラリーをまず作製し、 このライブラリーからヘルパーファージ等を用 いて一本鎖 DNAを調製し、 この一本鎖 DNAと別の細胞から得られた全 RN Aにビォチン標識したものとをハイプリダイズさせた後、 ァビジンを利用して ハイブリダイズしなかつた一本鎖 D N Aを単離し、 D N Aポリメラ一ゼによつ て二本鎖に戻して c D N Aライブラリ一を作製する方法 (Tanaka, H. , Yoshimura, Y. , Nishina, Υ. , Nozaki, Μ. , Noj iia, Η. , and Nishimune, Υ. (1994) FEBS Lett. 355, 4-10) 等が挙げられる。  This is a method in which cDNA of a gene specifically expressed in a specific cell is obtained, and the gene is cloned by screening a cDNA library using the cDNA as a probe. As a subtraction method, a single-stranded cDNA was prepared from total RNA, and this was hybridized with all RNA obtained from another cell, and then a single-stranded cDNA was not hybridized with a hydroxyapatite column. A method for isolating strand DNA and preparing a cDNA library from this cDNA (Bio Manual Series 3, Genetic Cloning Experiments, Yodosha (1993), Current 'Protocols in' Molecular · Biology) or a cDNA library was prepared first, and single-stranded DNA was prepared from this library using helper phage, etc., and biotin was added to this single-stranded DNA and all RNA obtained from another cell. After hybridizing with the labeled product, single-stranded DNA that did not hybridize was isolated using avidin, returned to double-stranded DNA by DNA polymerase, and cDNA A method for preparing a slurry (Tanaka, H., Yoshimura, Y., Nishina, Υ., Nozaki, zaki., Nojiia, Η., And Nishimune, Υ. (1994) FEBS Lett. 355, 4-10) And the like.
具体的には、 まず被験物質の投与または非投与条件下の検体それぞれについ て mRNAまたは全 RNAを精製し、 投与条件下の検体から精製した全 R N A を铸型とし、 逆転写酵素で cDNAを合成する。 合成時に [a- 32P] dNT Pを加えることで c DNAを標識することもできる。 標識された c DNAと铸 型となった全 RNAは安定な二本鎖 DNA- RNAハイプリッドを形成してい るが、 アル力リ存在下で高温処理することにより RNAのみを分解し一本鎖 c DNAを生成する。 この一本鎖 cDNAと、 非投与条件下の検体から抽出した RNAとを混合し、 適当な条件下で静置すると、 ヌクレオチド配列の相補性よ り安定な二本鎖 DNA- RNAハイプリッドを形成する。すなわち、非投与条件 下でも発現している cDNAはハイブリッドを形成するが、 投与条件下でのみ 特異的に発現している RNAを铸型とした c DNAは一本鎖のままである。 次 いで、ハイドロキシァパタイトカラムで二本鎖 DNA- RNAハイブリッドとー 本鎖 cDNAとを分離し、 一本鎖 c DN Aのみを精製する。 このステップを繰 り返すことで目的とした組織に特異的な c D N Aを濃縮することができる。 濃 縮された特異的 cDN Aは放射性同位元素等で標識されている場合は、 cDN Aライブラリーをスクリーニングするプローブとして使用することができる。 なお、 この操作は市販のキット (例えば、 P CRセレクト c DNAサブトラク シヨンキット:クローンテック社製等) を利用して行うこともできる。 Specifically, first, mRNA or total RNA is purified for each sample under the administration or non-administration conditions of the test substance, the total RNA purified from the sample under the administration condition is converted into type II, and cDNA is synthesized using reverse transcriptase. I do. CDNA can also be labeled by adding [a- 32 P] dNTP during synthesis. The labeled cDNA and total RNA in the form of a 铸 form a stable double-stranded DNA-RNA hybrid. However, high-temperature treatment in the presence of heat degrades only RNA and generates single-stranded cDNA. When this single-stranded cDNA and RNA extracted from a sample under non-administration conditions are mixed and allowed to stand under appropriate conditions, a double-stranded DNA-RNA hybrid is formed that is more stable than the nucleotide sequence complementarity. . That is, the cDNA expressed under non-administration conditions forms a hybrid, but the cDNA expressed specifically as RNA under the administration conditions remains single-stranded. Next, the double-stranded DNA-RNA hybrid and the single-stranded cDNA are separated on a hydroxyapatite column, and only the single-stranded cDNA is purified. By repeating this step, cDNA specific for the target tissue can be concentrated. When the concentrated specific cDNA is labeled with a radioisotope or the like, it can be used as a probe for screening a cDNA library. This operation can also be performed using a commercially available kit (for example, PCR Select cDNA Subtraction Kit: manufactured by Clontech, etc.).
b) ディファレンシャル'ディスプレイ法: b) Differential 'display method:
Liangらの方法 (Science (1992) 257, 967-971) に準じ、 例えば、 以下のよ うに実施できる。 まず比較する 2つの試料 (本発明の場合は被験物質の投与ま たは非投与条件下の検体) から mRNAまたは全 RNAを抽出し、 逆転写酵素 を用いてこれを一本鎖 c DN Aに変換する。 次いで、 得られた一本鎖 cDNA を铸型として、適当なプライマーを用いて P CRを行う。プライマーとしては、 例えば、 ランダムプライマー (任意の配列からなる約 10〜 12me rのプラ イマ一) を用いることができる。 あるいは、 アンカードプライマ一 (anchored primer) およびァーピトラリ一プライマー (arbitrary primer) 各一種ずつを 組み合わせて用いてもよい。 アンカ一ドプライマ一としては、 オリゴ d (T) nVX [n= 1 1〜12 ; V =グァニン、 アデニンまたはシトシン; X==グァ ニン、 アデニン、 チミンまたはシ卜シン] からなるプライマーを用いることが できる。 また、 ァービトラリープライマーとしては、 任意の配列からなる約 1 Ome rのランダムプライマーを用いることができる。 このような PCRを、 種々のプライマーを組み合わせて行うことで、 より広い範囲の遺伝子群をスク リーニングすることが可能となる。 続いて、 得られた PC R産物をゲル電気泳 動し、 ゲル上に展開 (ディスプレイ) される全 RN Aの発現パターン (フィン ガープリント) を比較解析することにより、 いずれかの検体で特異的に発現し ている遺伝子 (本発明の PADI4遺伝子) を選択し、 その cDNA断片を単離す ることができる。 なお、 この方法は、 市販されているキット (例えば、 RNA イメージ ·キッ 1、:ジェンハン夕一社製等) を用いて行うこともできる。 According to the method of Liang et al. (Science (1992) 257, 967-971), for example, it can be carried out as follows. First, mRNA or total RNA is extracted from two samples to be compared (in the case of the present invention, a sample under the administration or non-administration of the test substance), and this is converted into single-stranded cDNA using reverse transcriptase. Convert. Next, PCR is performed using the obtained single-stranded cDNA as a type II and using appropriate primers. As the primer, for example, a random primer (a primer of about 10 to 12 mers consisting of an arbitrary sequence) can be used. Alternatively, an uncard primer (anchored primer) and an arbitrary primer (arbitrary primer) may be used in combination. As an anchor primer, use a primer consisting of oligo d (T) n VX [n = 11 to 12; V = guanine, adenine or cytosine; X == guanine, adenine, thymine or cytosine] Can be done. In addition, as an arbitrary primer, a random primer of about 1 Omer having an arbitrary sequence can be used. By performing such PCR by combining various primers, it becomes possible to screen a wider range of gene groups. Subsequently, the obtained PCR product is subjected to gel electrophoresis, and the expression pattern (fins) of all RNAs spread (displayed) on the gel is displayed. By performing a comparative analysis of the garprint, a gene (PADI4 gene of the present invention) that is specifically expressed in any of the specimens can be selected, and a cDNA fragment thereof can be isolated. This method can also be performed using a commercially available kit (for example, RNA Image Kit 1, manufactured by Jenhan Yuichi).
c) ディファレンシャル ·ハイブリダィゼーシヨン法: c) Differential hybridization method:
目的の組織から精製した全 RNAから作製した cDNAライブラリ一を、 目 的組織および対照組織の全 R N Aから合成した32 P標識 c D N Aプローブで スクリーニングし、 目的組織のプローブとのみハイブリダィズするクローンを 選択する方法である。 例えば、 まず非投与条件下の検体から精製した全 RNA から常法に従い cDN Aライブラリーを作製し、 そのライブラリーから 2組の レプリカフィルターを作製する。 次に、 該非投与条件下の検体から精製した全 RNAを铸型として、 逆転写酵素で c DN Aを合成する。 合成時に [ -32 P] dNTPを加えることで c DNAを標識する。 標識された c DNAと铸型とな つた全 RNAは安定な二本鎖 DNA-RNAハイプリッドを形成している力 ァ ルカリ存在下で高温処理することにより全 RN Aのみを分解し、 一本鎖 cDN Aを精製する。 同様に、 被験物質投与条件下の検体から精製した全 RN Aを铸 型に32 Pで標識された一本鎖 c D N Aを作製する。両標識 c D N Aをそれぞれ プロ一プとして、 非投与条件下の検体から作製したフィルターとハイプリダイ ゼーシヨンを行う。 X線フィルムのオートラジオグラフィー像を比較し、 投与 または非投与条件下の c D N Aプローブの一方にのみハイブリダィズするクロ ーンを選ぶことにより、 被験物質の投与条件下で特異的に発現する遺伝子をク ローニングすることができる。 The cDNA library one prepared from total RNA purified from the tissue of interest, and screened with 32 P-labeled c DNA probe synthesized from total RNA of eye tissue and a control tissue, and clones are selected that seen Haiburidizu the probe of target tissue Is the way. For example, first, a cDNA library is prepared from total RNA purified from a sample under non-administration conditions according to a conventional method, and two sets of replica filters are prepared from the library. Next, cDNA is synthesized with reverse transcriptase using the total RNA purified from the sample under the non-administration condition as type III. During synthesis [- 32 P] labeling c DNA by adding dNTPs. The labeled cDNA and total RNA that has become type II are degraded to single-stranded DNA by high-temperature treatment in the presence of alkali that forms a stable double-stranded DNA-RNA hybrid. Purify the cDNA. Similarly, a single-stranded cDNA is prepared in which all RNA purified from a sample under the test substance administration conditions is labeled with 32 P in a small form. Using both labeled cDNAs as probes, perform hybridization with a filter prepared from a sample under non-administration conditions. By comparing the autoradiographic images of the X-ray film and selecting a clone that hybridizes only to one of the cDNA probes under the administration or non-administration conditions, a gene that is specifically expressed under the administration conditions of the test substance can be identified. Can be cloned.
d) クロスハイブリダイゼ一ション法: d) Cross-hybridization method:
被験物質の投与または非投与条件下の検体のいずれかに由来する c DNAラ イブラリーに対して、 適当な DNAをプローブとして、 ストリンジエンシーの 低い条件でハイブリダィゼーシヨンを行い、 陽性クローンを得る。 得られた陽 性クローンをプローブとして、 それぞれの検体に由来する全 R N Aに対してノ —ザンハイブリダィゼーシヨンを行い、 一方にのみ発現しているクローンを選 択する。  Positive clones were obtained by hybridizing the cDNA library derived from either the test substance-administered or non-administered sample with low DNA stringency using appropriate DNA as a probe. obtain. Using the obtained positive clone as a probe, Northern hybridization is performed on all RNAs derived from each sample, and a clone expressing only one of them is selected.
こうして得られた c DNAをプローブとして、 投与または非投与条件下の検 体の全 R NAに対してノーザンプロッティングを行い、 選択した遺伝子の全 R NAが、 投与条件下で特異的に発現していることを確認できる。 工程 3 :被験物質の評価 Using the cDNA obtained in this way as a probe, testing under administration or non-administration conditions By performing Northern plotting on all RNAs of the body, it can be confirmed that all the RNAs of the selected gene are specifically expressed under the administration conditions. Step 3: Evaluation of test substance
最後に、 被験物質の投与または非投与における、 本発明の PADI4遺伝子の発 現量の相違に基づいて、 該被験物質の R A治療薬としての効果を評価する。 すなわち、 被験物質の投与条件下で非投与条件下よりも本発明の PADI4遺伝 子の発現量が有意に減少している場合、 該被験物質は R A治療薬としての効果 を有すると評価できる。 ここで、 「有意に減少している」 とは、 例えば、 被験 物質の投与および非投与条件下での本発明の PADI4遺伝子の発現量に統計的有 意差 (p < 0 . 0 5 ) があることを意味する。  Finally, the effect of the test substance as an RA therapeutic is evaluated based on the difference in the expression level of the PADI4 gene of the present invention between administration and non-administration of the test substance. That is, when the expression level of the PADI4 gene of the present invention is significantly reduced under the administration condition of the test substance as compared with the non-administration condition, it can be evaluated that the test substance has an effect as a therapeutic agent for RA. Here, “significantly reduced” means that, for example, a statistically significant difference (p <0.05) in the expression level of the PADI4 gene of the present invention under the administration and non-administration conditions of the test substance. It means there is.
2 . 2 本発明の PADI4蛋白質の発現量を指標とした評価方法 2.2 Evaluation method using expression level of PADI4 protein of the present invention as an index
本発明の PADI4蛋白質の発現量を指標とした in vivoにおける評価方法は、 例えば下記の工程を含む。  The in vivo evaluation method using the expression level of the PADI4 protein of the present invention as an index includes, for example, the following steps.
工程 1 :動物を被験物質の投与または非投与の条件下で飼育する。 Step 1: The animals are bred under the conditions of administration or non-administration of the test substance.
工程 2 :上記動物の血液または細胞中における本発明の PADI4蛋白質の発現量 を、 該蛋白質に特異的に結合する抗体を用いて検出する。 Step 2: The expression level of the PADI4 protein of the present invention in the blood or cells of the animal is detected using an antibody that specifically binds to the protein.
工程 3:被験物質の投与または非投与における、上記発現量の相違に基づいて、 該被験物質の R A治療薬としての効果を評価する。 工程 1 :動物の飼育 Step 3: The effect of the test substance as an RA therapeutic agent is evaluated based on the difference in the expression level between the administration and non-administration of the test substance. Process 1: Animal rearing
動物は、 前項 2 . 1に記載した方法にしたがい、 被験物質の投与または非投 与の条件下で飼育する。 工程 2 :本発明の PADI4蛋白質の発現量の検出  Animals are to be bred under the conditions of administration or non-administration of the test substance according to the method described in 2.1 above. Step 2: Detection of expression level of PADI4 protein of the present invention
次に、 上記動物の血液または細胞中における本発明の PADI4蛋白質の発現量 を、 該蛋白質に特異的に結合する抗体を用いて検出する。  Next, the expression level of the PADI4 protein of the present invention in the blood or cells of the animal is detected using an antibody that specifically binds to the protein.
抗体を利用した蛋白質の検出方法は特に限定されないが、 ウエスタンブロッ ト法、 ドットブロット法、 スロットブロット法、 E L I S A法、 および R I A 法から選ばれるいずれか一の方法であることが好ましい。 以下、 これらの検出 方法について、 試料の調製から検出までを具体的に説明する。 Methods for detecting proteins using antibodies are not particularly limited, but include Western blot, dot blot, slot blot, ELISA, and RIA. Preferably, the method is any one selected from the following methods. Hereinafter, these detection methods will be described specifically from sample preparation to detection.
( 1 ) 試料の調製  (1) Sample preparation
検体としては、 血液または本発明の PADI4蛋白質が高発現している細胞が好 ましく、 したがって好中球や好酸球等の血液由来細胞、 滑膜細胞、 脾臓細胞、 および腹腔浸潤細胞が好ましい。 これらの血液または細胞 (細胞抽出液として 使用する) は、 必要に応じて高速遠心を行うことにより不溶性の物質を除去し た後、 以下のように E L I S A/R I A用試料やウェスタンブロット用試料と して調製する。  As the specimen, blood or cells in which the PADI4 protein of the present invention is highly expressed are preferable, and therefore, blood-derived cells such as neutrophils and eosinophils, synovial cells, spleen cells, and intraperitoneal cells are preferable. . These blood or cells (used as a cell extract) are subjected to high-speed centrifugation as necessary to remove insoluble substances, and then used as ELISA / RIA samples or Western blot samples as follows. To be prepared.
E L I S A/R I A用試料は、例えば、回収した血清をそのまま使用するか、 緩衝液で適宜希釈したものを用いる。 ウエスタンプロット用 (電気泳動用) 試 料は、 例えば、 細胞抽出液をそのまま使用するか、 緩衝液で適宜希釈して、 S D S—ボリアクリルアミドゲル電気泳動用の 2 _メルカトルエタノールを含む ンプル緩衝液 (シグマ社製等) と混合したものを用いる。 ドット /スロット プロット用試料は、 例えば、 回収した細胞抽出液そのもの、 または緩衝液で適 宜希釈したものを、 ブロッテイング装置を使用するなどして、 直接メンブレン へ吸着させたものを用いる。  As the sample for ELISA / RIA, for example, the collected serum may be used as it is, or may be appropriately diluted with a buffer. For Western blot (for electrophoresis), for example, use a cell extract as it is or dilute appropriately with a buffer to prepare a sample buffer containing 2_mercatorethanol for SDS-polyacrylamide gel electrophoresis ( Sigma). As the sample for dot / slot plotting, for example, a recovered cell extract itself or a sample appropriately diluted with a buffer is directly adsorbed to a membrane using a blotting apparatus.
( 2 ) 試料の固相化  (2) Immobilization of sample
上記方法では、 まず、 本発明の PADI4蛋白質が含まれる試料中のボリべプチ ドをメンブレンあるいは 9 6穴プレートのゥエル内底面等に固相化する。  In the above method, first, polypeptide in a sample containing the PADI4 protein of the present invention is immobilized on a membrane or the bottom surface of a well of a 96-well plate.
メンブレンに固相化する方法としては、 試料のポリアクリルアミドゲル電気 泳動を経てメンブレンにポリぺプチドを転写する方法(ウエスタンプロット法) と、 直接メンブレンに試料またはその希釈液を染み込ませる方法 (ドットプロ ット法やスロットプロット法) を挙げることができる。 用いられるメンブレン としては、 ニトロセルロースメンブレン (例えば、 バイオラッド社製等)、 ナイ ロンメンブレン (例えば、 ハイボンド一 E C L (アマシャム ·フアルマシア社 製)等)、コットンメンブレン (例えば、ブロットァブソーベントフィルター (バ ィォラッド社製) 等) またはポリビニリデン ·ジフルオリド (P V D F ) メン プレン (例えば、 バイオラッド社製等) 等を挙げることができる。 また、 プロ ッティング方法としては、ゥエツト式ブロッティング法(CURRENT PROTOCOLS IN IMMUNOLOGY volume 2 ed by J. E. Col igan, A. M. Krui sbeek, D. H. Margul ies, E. M. Shevach, W. Strober) , セミドライ式ブロッテイング法 (上記 CURRENT PROTOCOLS IN IMMUNOLOGY volume 2 参照) 等を挙げることができる。 There are two methods for immobilizing the sample on the membrane: transferring the polypeptide to the membrane via polyacrylamide gel electrophoresis of the sample (Western plot method), and directly impregnating the sample or its diluent into the membrane (dot prototyping). Slot method or slot plot method). Examples of the membrane to be used include a nitrocellulose membrane (for example, manufactured by Bio-Rad), a nylon membrane (for example, Hybond-ECL (manufactured by Amersham-Pharmacia)), a cotton membrane (for example, blotting sorbent filter (for example, Biorad) and polyvinylidene difluoride (PVDF) membrane (for example, Biorad). In addition, the plotting method used is an 式 ET-type blotting method (CURRENT PROTOCOLS IN IMMUNOLOGY volume 2 ed by JE Coligan, AM Krui sbeek, DH Margulies, EM Shevach, W. Strober), semi-dry blotting method (see CURRENT PROTOCOLS IN IMMUNOLOGY volume 2 above), and the like.
一方、 9 6穴プレートに固相化する方法としては、 固相酵素免疫定量法 (E L I S A法) や放射性同位元素免疫定量法 (R I A法) 等を挙げることができ る。 固相化は、 例えば、 前記 9 6穴プレート (例えば、 ィムノプレート ·マキ シソープ(ヌンク社製)等) に試料またはその希釈液(例えば、 0 . 0 5 % ァ ジ化ナトリウムを含むリン酸緩衝生理食塩水 (以下「P B S」 と記載する。) で 希釈したもの) を入れて 4で〜室温で一晩、 または 3 7 °Cで 1〜3時間静置し て、 ゥエル底面にポリペプチドを吸着させればよい。  On the other hand, examples of the method for immobilization on a 96-well plate include enzyme-linked immunosorbent assay (ELISA) and radioisotope immunoassay (RIA). The immobilization may be performed, for example, by subjecting the 96-well plate (for example, Imnoplate Maxi Soap (manufactured by Nunc) or the like) to a sample or a dilution thereof (for example, phosphate buffered saline containing 0.05% sodium azide). Add a saline solution (hereinafter, referred to as “PBS”) diluted, and incubate at 4 to room temperature overnight or at 37 ° C for 1 to 3 hours to adsorb the polypeptide on the bottom of the well. You can do it.
( 3 ) 本発明の PADI4蛋白質に特異的に結合する抗体  (3) an antibody that specifically binds to the PADI4 protein of the present invention
本工程で用いられる「本発明の Ρ Π4蛋白質に特異的に結合する抗体 (以下、 「抗 PADI4抗体」 と記載する。)」 は、 公知の方法にしたがって調製してもよい し、 市販のものを用いてもよい。  The “antibody that specifically binds to the Δ4 protein of the present invention (hereinafter referred to as“ anti-PADI4 antibody ”)” used in this step may be prepared according to a known method, or may be commercially available. May be used.
前記抗体は、 常法により (例えば、 新生化学実験講座 1、 タンパク質 1、 P. 389-397、 1992)、 抗原となる本発明の PADI4蛋白質、 あるいはそのアミノ酸 配列から選択される任意のポリペプチドを用いて動物を免疫し、 該動物生体内 に産生される抗体を採取、 精製することによって得ることができる。 また、 公 知の方法 (例えば、 Kohler and Mi ls te in, Nature 256, 495-497, 1975, Kennet, R. ed. , Monoclonal Ant ibody p. 365-367, 1980, Prenum Press, N. Y. ) にした がって、 抗 PADI4抗体を産生する抗体産生細胞とミエローマ細胞とを融合させ ることによりハイプリドーマを樹立し、 これよりモノクローナル抗体を得るこ ともできる。  The antibody can be prepared by a conventional method (for example, Shinsei Kagaku Kenkyusho 1, Protein 1, P. 389-397, 1992), using the PADI4 protein of the present invention as an antigen or any polypeptide selected from the amino acid sequence thereof. It can be obtained by immunizing an animal with the antibody and collecting and purifying an antibody produced in the living body of the animal. In addition, publicly known methods (for example, Kohler and Children, Nature 256, 495-497, 1975, Kennet, R. ed., Monoclonal Ant ibody p. 365-367, 1980, Prenum Press, NY) were used. Thus, a hybridoma can be established by fusing the antibody-producing cells producing the anti-PADI4 antibody with myeloma cells, and a monoclonal antibody can be obtained therefrom.
抗体作製用の抗原としては、 本発明の PADI4蛋白質またはその少なくとも 6 個の連続した部分アミノ酸配列からなるポリペプチド、 あるいはこれらに任意 のアミノ酸配列や担体 (例えば、 N末端付加するキーホールリンペットへモシ ァニン) が付加された誘導体を挙げることができる。  Antigens for producing antibodies include the PADI4 protein of the present invention or a polypeptide comprising at least six consecutive partial amino acid sequences thereof, or any amino acid sequence or carrier thereof (for example, a keyhole limpet to which an N-terminal is added). And derivatives to which mosyanine has been added.
前記抗原ポリペプチドは、 本発明の PADI4蛋白質を遺伝子操作により宿主細 胞に産生させることによって得ることができる。 具体的には、 本発明の PADI4 遺伝子を発現可能なベクターを作製し、 これを宿主細胞に導入して該遺伝子を 発現させればよい。 The antigen polypeptide can be obtained by producing a PADI4 protein of the present invention in a host cell by genetic manipulation. Specifically, a vector capable of expressing the PADI4 gene of the present invention is prepared, and this is introduced into a host cell to transform the gene. It may be expressed.
前記宿主細胞としては、 原核細胞であれば、 例えば、 大腸菌 (Escherichia coli) や枯草菌 (Bacillus subtilis) 等が挙げられる。 目的の遺伝子をこれら の宿主細胞内で形質転換させるには、 宿主と適合し得る種由来のレブリコンす なわち複製起点と、 調節配列を含んでいるプラスミドベクターで宿主細胞を形 質転換させる。 該ベクタ一としては、 形質転換細胞に表現形質 (表現型) の選 択性を付与しうる配列を有するものが好ましい。  Examples of the host cell include prokaryotic cells, such as Escherichia coli and Bacillus subtilis. To transform a gene of interest into these host cells, the host cells are transformed with a plasmid vector that contains a regulatory element and a replicon, or replication origin, from a species compatible with the host. The vector is preferably one having a sequence capable of imparting phenotypic (phenotypic) selectivity to transformed cells.
例えば、 大腸菌であれば、 K 12株等がよく用いられ、 ベクターとしては、 一般に p BR 322や pUC系のプラスミドが用いられるが、 これらに限定さ れず、 公知の各種菌株やべクタ一を使用できる。 また、 大腸菌で用いられるプ 口モーターとしては、 例えば、 トリプ卜ファン (trp) プロモーター、 ラク卜一 ス (lac) プロモーター、 トリプトファン ·ラクト一ス (tac) プロモーター、 リポプロテイン (lpp) プロモーター、 ポリペプチド鎖伸張因子 Tu (tufB) プ 口モータ一等を挙げることができ、 いずれも好適に用いることができる。  For example, in the case of Escherichia coli, the K12 strain or the like is often used, and the vector is generally a pBR322 or pUC-type plasmid, but is not limited thereto, and may use various known strains or vectors. it can. In addition, examples of a promoter used in Escherichia coli include a tryptophan (trp) promoter, a lactose (lac) promoter, a tryptophan lactose (tac) promoter, a lipoprotein (lpp) promoter, and a polypeptide. Chain elongation factor Tu (tufB) peptide motor and the like can be mentioned, and any of them can be suitably used.
また、 枯草菌であれば、 207— 25株が好ましく、 ベクターとしては pT UB 228 (Ohmura, K. et al. (1984) J. Bioc em. 95, 87-93) 等が用いら れるが、 これに限定されるものではない。 なお、 ベクターに枯草菌の α—アミ ラ一ゼのシグナルべプチド配列をコードする DN Α配列を連結することにより、 菌体外での分泌発現も可能となる。  For Bacillus subtilis, strain 207-25 is preferable, and pT UB 228 (Ohmura, K. et al. (1984) J. Biochem. 95, 87-93) is used as a vector. It is not limited to this. In addition, extracellular secretory expression can be achieved by ligating the DNΑ sequence encoding the signal peptide sequence of α-amylase of Bacillus subtilis to the vector.
真核細胞の宿主細胞としては、脊椎動物、昆虫、酵母等の細胞が挙げられる。 脊椎動物細胞としては、 例えば、 サルの細胞である COS細胞 (Gluzman, Y. (1981) Cell 23, 175-182、 ATCC C L-1650) や CHO細胞 (ATCC CCL-61) のジ ヒドロ葉酸還元酵素欠損株 (Urlaub, G. and Chasin, L. A. (1980) Pro Natl. Acad. Sci. USA 77, 4126-4220) 等がよく用いられているが、 これらに限定さ れない。  Eukaryotic host cells include cells such as vertebrates, insects, and yeast. Vertebrate cells include, for example, dihydrofolate reduction of monkey COS cells (Gluzman, Y. (1981) Cell 23, 175-182, ATCC CL-1650) and CHO cells (ATCC CCL-61). Enzyme-deficient strains (Urlaub, G. and Chasin, LA (1980) Pro Natl. Acad. Sci. USA 77, 4126-4220) are frequently used, but are not limited thereto.
脊椎動物細胞の発現ベクターとしては、 通常発現させようとする遺伝子の上 流に位置するプロモーター、 RN Aのスプライス部位、 ポリアデニル化部位、 および転写終結配列等を有するものを使用できる。 さらに、 これは必要により 複製起点を有してもよい。 該発現ベクターの例としては、 サイトメガロウィル ス初期プロモータ一を有する p CR 3. 1 (Invitrogen 社製)、 SV40の初 期プロモーターを有する p S V2 d h ί r (Subramani, S. et al. (1981) Mol. Cell. Biol. 1, 854-864) 等が挙げられるが、 これらに限定されない。 As an expression vector for a vertebrate cell, a vector having a promoter, an RNA splice site, a polyadenylation site, a transcription termination sequence, and the like located upstream of a gene to be normally expressed can be used. In addition, it may have a replication origin if desired. Examples of the expression vector include pCR3.1 (manufactured by Invitrogen) having the cytomegalovirus early promoter and the SV40 V2 dhί r having an early promoter (Subramani, S. et al. (1981) Mol. Cell. Biol. 1, 854-864), and the like, but are not limited thereto.
宿主細胞として、 COS細胞を用いる場合を例に挙げると、 発現ベクターと しては、 S V 40複製起点を有し、 COS細胞において自立増殖が可能であり、 さらに、 転写プロモーター、 転写終結シグナル、 および RN Aスプライス部位 を備えたものを好適に用いることができる。 該発現べクタ一は、 ジェチルアミ ノエチル(DEAE) —デキストラン法(Luthman, H. and Magnus son, G. (1983) Nucleic Acids Res, 11, 1295-1308), リン酸カルシウム— D N A共沈殿法 (Graham, F. L. and van der Eb, A. J. (1973) Virology 52, 456 - 457)、 お よび電気パルス穿孔法 (Neumann, E. et al. (1982) EMB0 J. 1, 841-845) 等 により COS細胞に取り込ませることができ、 かくして所望の形質転換細胞を 得ることができる。  Taking the case where COS cells are used as host cells as an example, the expression vector has an SV40 origin of replication, is capable of autonomous growth in COS cells, and has a transcription promoter, a transcription termination signal, and Those having an RNA splice site can be suitably used. The expression vector is prepared by the method of getylaminoethyl (DEAE) -dextran method (Luthman, H. and Magnus son, G. (1983) Nucleic Acids Res, 11, 1295-1308), calcium phosphate-DNA coprecipitation method (Graham, FL) and van der Eb, AJ (1973) Virology 52, 456-457) and electric pulse perforation (Neumann, E. et al. (1982) EMB0 J. 1, 841-845) Thus, desired transformed cells can be obtained.
また、 宿主細胞として CH〇細胞を用いる場合には、 発現べクタ一と共に、 抗生物質 G418耐性マーカ一として機能する n e o遺伝子を発現し得るべク 夕一、 例えば、 pRSVn e o (Saibrook, J. et al. (1989) : "Molecular Cloning A Laboratory Manual Cold Spring Harbor Laboratory, NY) や p S V2 n e o (Southern, P. J. and Berg, P. (1982) J. Mol. Appl. Genet. 1, 327-341)等をコ ' トランスフエクトし、 G418耐性のコロニーを選択するこ とにより、 目的のポリペプチドを安定に産生する形質転換細胞を得ることがで きる。  When CH〇 cells are used as host cells, vectors that can express the neo gene functioning as an antibiotic G418 resistance marker together with the expression vector, for example, pRSVneo (Saibrook, J. et. al. (1989): "Molecular Cloning A Laboratory Manual Cold Spring Harbor Laboratory, NY) and p S V2 neo (Southern, PJ and Berg, P. (1982) J. Mol. Appl. Genet. 1, 327-341) Transfected cells that stably produce the desired polypeptide can be obtained by co-transfecting them and selecting G418-resistant colonies.
昆虫細胞を宿主細胞として用いる場合には、 鱗翅類ャガ科の Spodoptera frugiperda の卵巣細胞由来株化細胞 ( S f — 9または S ί— 2 1 ) や Trichoplusia niの卵細胞由来 High Five細胞 (Wickham, T. J. et al, (1992) Biotechnol. Prog, i: 391-396) 等が宿主細胞としてよく用いられ、 バキュ口 ウィルストランスファーベクターとしてはオートグラファ核多角体ウィルス (Ac NP V) のポリへドリン夕ンパクのプロモ一夕一を利用した pVL 13 92/1393がよく用いられる (Kidd, i . Μ· and V. C. Emery (1993) The use of baculoviruses as expression vectors. Applied Biochemistry and Biotechnology 420, 137-159)。 この他にも、 バキュロウィルスの P 10や同塩 基性蛋白質のプロモータ一を利用したベクタ一も使用できる。 さらに、 AcN P Vのエンベロープ表面蛋白質 G P 6 7の分泌シグナル配列を目的蛋白質の N 末端側に繋げることにより、 組換え蛋白質を分泌蛋白質として発現させること も可能である (Zhe-mei Wang, et al. (1998) Biol. Chem. , 379, 167 - 174)。 When insect cells are used as host cells, cell lines derived from ovarian cells of Spodoptera frugiperda (S f — 9 or S ί − 21) of the family Lepidoptera and the High Five cells derived from Trichoplusia ni eggs (Wickham, TJ) et al, (1992) Biotechnol. Prog, i: 391-396) is often used as a host cell, and as a Bacchus virus transfer vector, the polyhedrin protein of autographa nucleopolyhedrovirus (AcNPV) is used. PVL 1392/1393 using the Promo overnight is often used (Kidd, i. Μ · and VC Emery (1993) The use of baculoviruses as expression vectors. Applied Biochemistry and Biotechnology 420, 137-159). In addition, vectors using baculovirus P10 and a promoter of the same basic protein can also be used. In addition, AcN The recombinant protein can be expressed as a secreted protein by connecting the secretory signal sequence of the PV envelope surface protein GP67 to the N-terminal side of the target protein (Zhe-mei Wang, et al. (1998) Biol. Chem., 379, 167-174).
真核微生物を宿主細胞とした発現系としては、 酵母が一般によく知られてお り、 その中でもサッカロミセス属酵母、 例えば、 パン酵母 Saccharomyces cerevis iaeや石油酵母 Pichia pas tor isが好ましい。 該酵母等の真核微生物の 発現べクタ一としては、 例えば、 アルコール脱水素酵素遺伝子のプロモーター (Benne tzen, J. L. and Hal l, B. D. (1982) J. Biol. Chem. 257, 3018-3025) や酸性フォスファターゼ遺伝子のプロモー夕一 (Miyanohara, A. et al. (1983) Proc. Nat l. Acad. Sci. USA 80, 1-5) 等を好ましく利用できる。 また、 分泌 型蛋白質として発現させる場合には、 分泌シグナル配列と宿主細胞の持つ内在 性プロテアーゼあるいは既知のプ口テアーゼの切断部位を N末端側に持つ組換 え体として発現させることも可能である。 例えば、 トリプシン型セリンプロテ ァーゼのヒトマスト細胞トリプターゼを石油酵母で発現させた系では、 N末端 側に酵母の enファクタ一の分泌シグナル配列と石油酵母の持つ K E X 2プロテ ァーゼの切断部位をつなぎ発現させることにより、 活性型トリプ夕ーゼが培地 中に分泌されることが知られている (Andrew, L. Ni les, et al. (1998) Biotechnol. Appl. Biochem. 28, 125-131)。  As an expression system using a eukaryotic microorganism as a host cell, yeast is generally well known. Among them, yeast of the genus Saccharomyces, for example, baker's yeast Saccharomyces cerevis iae and petroleum yeast Pichia pastoris are preferable. Examples of expression vectors for eukaryotic microorganisms such as the yeast include, for example, a promoter for an alcohol dehydrogenase gene (Bennettzen, JL and Hall, BD (1982) J. Biol. Chem. 257, 3018-3025) and Acid phosphatase gene promoter (Miyanohara, A. et al. (1983) Proc. Natl. Acad. Sci. USA 80, 1-5) can be preferably used. When expressed as a secretory protein, it can also be expressed as a recombinant having a secretory signal sequence and a cleavage site for an endogenous protease or a known protease of the host cell at the N-terminal side. . For example, in a system in which human mast cell tryptase, a trypsin-type serine protease, is expressed in petroleum yeast, expression must be performed by connecting the secretory signal sequence of yeast en factor-1 to the N-terminal end of the KEX2 protease cleavage site of petroleum yeast. It is known that activated trypsinase is secreted into the culture medium (Andrew, L. Niles, et al. (1998) Biotechnol. Appl. Biochem. 28, 125-131).
上記のようにして得られる形質転換体は、 常法にしたがい培養することがで き、 該培養により細胞内、 または細胞外に目的のポリペプチドが産生される。 該培養に用いられる培地としては、 採用した宿主細胞に応じて慣用される各種 の培地を適宜選択できる。 例えば、 上記 C O S細胞であれば、 R P M I 1 6 4 0培地やダルベッコ変法イーグル培地 (以下 「D M E M」 と記載する) 等の培 地に、 必要に応じゥシ胎児血清等の血清成分を添加したものを使用できる。 上記培養により、 形質転換体の細胞内または細胞外に産生される組換え蛋白 質は、 該蛋白質の物理的性質や化学的性質等を利用した公知の分離操作法によ り、 分離 ·精製することができる。 該方法としては、 例えば、 タンパク沈殿剤 による処理、 限外濾過、 分子ふるいクロマトグラフィー (ゲル濾過)、 吸着クロ マトグラフィ一、 イオン交換クロマ卜グラフィー、 ァフィ二ティークロマトグ ラフィ一、 高速液体クロマトグラフィー (H P L C ) 等の各種液体クロマトグ ラフィー、 透析法、 を単独あるいは組合せて利用できる。 また、 発現させる組 換え蛋白質に 6残基からなるヒスチジンを繋げれば、 ニッケルァフィ二ティー カラムで効率的に精製することができる。 目的とする本発明の PADI4タンパク は、 以上に記載した方法を適宜組み合わせることにより、 容易に高収率、 高純 度で製造できる。 The transformant obtained as described above can be cultured according to a conventional method, and the culture produces the target polypeptide inside or outside the cell. As the medium used for the culture, various types of commonly used media can be appropriately selected depending on the host cell used. For example, in the case of the above COS cells, serum components such as fetal calf serum were added to a medium such as RPMI 1640 medium or Dulbecco's modified Eagle medium (hereinafter referred to as “DMEM”) as necessary. Anything can be used. The recombinant protein produced inside or outside the cells of the transformant by the above culture is separated and purified by a known separation operation method utilizing the physical and chemical properties of the protein. be able to. Examples of the method include treatment with a protein precipitant, ultrafiltration, molecular sieve chromatography (gel filtration), adsorption chromatography, ion exchange chromatography, affinity chromatography, high-performance liquid chromatography (HPLC ) Etc. Raffy and dialysis can be used alone or in combination. If a histidine consisting of 6 residues is linked to the recombinant protein to be expressed, it can be purified efficiently with a nickel affinity column. The target PADI4 protein of the present invention can be easily produced in high yield and high purity by appropriately combining the methods described above.
( 4 ) 検出 '  (4) Detection ''
上記 (3 ) 記載の方法で得られる抗 PADI4抗体は、 それを直接標識するか、 または該抗体を一次抗体とし、 該一次抗体を特異的に認識する (抗体を作製し た動物由来の抗体を認識する) 標識二次抗体と協同で検出に用いられる。  The anti-PADI4 antibody obtained by the method described in the above (3) may be directly labeled, or may be used as a primary antibody to specifically recognize the primary antibody (antibody derived from the animal from which the antibody was prepared) Recognition) Used for detection in cooperation with a labeled secondary antibody.
前記標識の種類として好ましいものは、 酵素 (アルカリホスファターゼまた は西洋ヮサビペルォキシダーゼ) またはピオチン (ただし二次抗体のピオチン にさらに酵素標識ストレプトアビジンを結合させる操作が加わる) であるが、 これらに限定されない。 標識二次抗体 (または標識ストレプトアビジン) とし ては、 予め標識された抗体 (またはストレプトアビジン) が、 各種市販されて いる。なお、 R I Aの場合は 125 1等の放射性同位元素で標識された抗体を用い、 測定は液体シンチレ一シヨンカウン夕一等を用いて行う。 Preferable examples of the type of the label include an enzyme (alkaline phosphatase or horseradish peroxidase) or piotin (however, an operation of binding an enzyme-labeled streptavidin to the secondary antibody piotin is added). Not limited. As the labeled secondary antibody (or labeled streptavidin), various types of pre-labeled antibodies (or streptavidin) are commercially available. In the case of RIA using antibodies labeled with a radioactive isotope such as 125 1, the measurement is performed using a liquid scintillation one Shiyonkaun evening first prize.
これら標識された酵素の活性を検出することにより、 抗原の発現量が測定さ れる。 アル力リホスファタ一ゼまたは西洋ヮサビペルォキシダーゼで標識する 場合、これら酵素の触媒により発色する基質や発光する基質が市販されている。 発色する基質を用いた場合、 ウエスタンプロット法やドット/スロットプロ ット法を利用すれば、 目視で検出できる。 E L I S A法では、 市販のマイクロ プレートリーダーを用いて各ゥエルの吸光度 (測定波長は基質により異なる) を測定し、 定量することが好ましい。 また上述の抗体作製に使用した抗原の希 釈系列を調製し、これを標準抗原試料として他の試料と同時に検出操作を行い、 標準抗原濃度と測定値をプロットした標準曲線を作成することにより、 他の試 料中の抗原濃度を定量することも可能である。  By detecting the activity of these labeled enzymes, the expression level of the antigen is measured. In the case of labeling with alkaline phosphatase or horseradish peroxidase, substrates that emit color or emit light by the catalyst of these enzymes are commercially available. When a color-developing substrate is used, it can be detected visually using Western plotting or dot / slot plotting. In the ELISA method, it is preferable to measure and quantify the absorbance (measurement wavelength varies depending on the substrate) of each well using a commercially available microplate reader. Also, by preparing a dilution series of the antigen used in the above antibody preparation, using this as a standard antigen sample, performing detection operations simultaneously with other samples, and creating a standard curve plotting the standard antigen concentration and the measured value, It is also possible to quantify the concentration of antigen in other samples.
一方、 発光する基質を使用した場合は、 ウエスタンプロット法やドット/ス ロットプロット法においては、 X線フィルムまたはイメージングプレートを用 いたォートラジオグラフィ一や、 インスタントカメラを用いた写真撮影により 検出することができる。 また、 デンシトメトリ一やモレキュラー ·イメージャ — Fxシステム(バイオラッド社製)等を利用した定量も可能である。さらに、 EL I S A法で発光基質を用いる場合は、 発光マイクロプレートリーダー (例 えば、 バイオラッド社製等) を用いて酵素活性を測定する。 On the other hand, when a substrate that emits light is used, detection can be performed by Western radiography or dot / slot plotting by autoradiography using an X-ray film or imaging plate, or by photographing using an instant camera. Can be. Also, densitometry and molecular imagers — Quantification using Fx system (manufactured by Bio-Rad) is also possible. When a luminescent substrate is used in the ELISA method, the enzyme activity is measured using a luminescent microplate reader (for example, Bio-Rad).
(5) 測定操作  (5) Measurement operation
a) ウエスタンブロット、 ドットプロットまたはスロットブロットの場合 まず、 抗体の非特異的吸着を阻止するため、 予めメンブレンをそのような非 特異的吸着を阻害する物質 (スキムミルク、 カゼイン、 ゥシ血清アルブミン、 ゼラチン、 ポリビニルピロリドン等) を含む緩衝液中に一定時間浸しておく操 作 (ブロッキング) を行う。 ブロッキング溶液の組成は、 例えば、 5% スキ ムミルク、 0. 05〜0. 1 % ツイ一ン 20を含むリン酸緩衝生理食塩水(P BS) またはトリス緩衝生理食塩水 (TBS) が用いられる。 スキムミルクの 代わりに、 ブロックエース (大日本製薬)、 ;!〜 10 %のゥシ血清アルブミン、 0.5〜3 %のゼラチンまたは 1 %のポリビニルピロリドン等を用いてもよい。 ブロッキングの時間は、 4でで 16〜24時間、 または室温で 1〜3時間であ る。 a) In the case of Western blot, dot plot or slot blot First, in order to prevent non-specific adsorption of antibodies, the membrane must be treated with a substance that inhibits such non-specific adsorption (skim milk, casein, serum albumin, gelatin, etc.). (Polyvinylpyrrolidone, etc.) for a certain period of time (blocking). As the composition of the blocking solution, for example, phosphate buffered saline (PBS) or Tris buffered saline (TBS) containing 5% skim milk, 0.05 to 0.1% Tween 20 is used. Instead of skim milk, Block Ace (Dainippon Pharmaceutical); -10% serum albumin, 0.5-3% gelatin or 1% polyvinylpyrrolidone may be used. The blocking time is 4 to 16 hours at 4 or 1 to 3 hours at room temperature.
次に、 メンブレンを 0. 05〜0. 1 % ツイーン 20を含む PBSまたは TBS (以下 「洗浄液」 と記載する) で洗浄して余分なブロッキング溶液を除 去した後、 ブロッキング溶液で適宜希釈した溶液中に抗 PAD 14抗体を一定時間 浸して、 メンブレン上の抗原に該抗体を結合させる。 このときの抗体の希釈倍 率は、 例えば、 前記組換え抗原を段階希釈したものを試料とした予備的なゥェ スタンプロッティング実験を行って決定することができる。 この抗体反応操作 は、 好ましくは室温で 2時間行う。 抗体反応操作終了後、 メンブレンを洗浄液 で洗浄する。 ここで、 用いた抗体が標識されたものである場合は、 ただちに検 出操作を行うことができる。 未標識の抗体を用いた場合には、 引き続き二次抗 体反応を行う。 標識二次抗体は、 例えば、 市販のものを使用する場合はプロッ キング溶液で 2000〜 20000倍に希釈して用いる (添付の指示書に好適 な希釈倍率が記載されている場合は、その記載に従う)。一次抗体を洗浄除去し た後のメンブレンを二次抗体溶液に室温で 45分〜 1時間浸し、 洗浄液で洗浄 してから、 標識方法に合わせた検出操作を行う。 洗浄操作は、 例えば、 まずメ ンブレンを洗浄液中で 15分間振盪してから、 洗浄液を新しいものに交換して 5分間振盪した後、 再度洗浄液を交換して 5分間振盪することにより行う。 必 要に応じてさらに洗浄液を交換して洗浄してもよい。 Next, the membrane is washed with PBS or TBS containing 0.05 to 0.1% Tween 20 (hereinafter referred to as “washing solution”) to remove excess blocking solution, and then diluted appropriately with the blocking solution. An anti-PAD14 antibody is immersed in the solution for a certain period of time to allow the antibody to bind to the antigen on the membrane. The dilution ratio of the antibody at this time can be determined, for example, by performing a preliminary pastelotting experiment using a serial dilution of the recombinant antigen as a sample. This antibody reaction operation is preferably performed at room temperature for 2 hours. After the antibody reaction operation, wash the membrane with washing solution. Here, if the antibody used is labeled, the detection operation can be performed immediately. If an unlabeled antibody is used, perform a secondary antibody reaction. For example, when using a commercially available labeled secondary antibody, dilute it with a blocking solution at a ratio of 2000 to 20000 times. (If a suitable dilution ratio is described in the attached instruction sheet, follow the description.) ). After washing and removing the primary antibody, immerse the membrane in the secondary antibody solution at room temperature for 45 minutes to 1 hour, wash with the washing solution, and perform the detection operation according to the labeling method. The washing operation is performed, for example, by first shaking the membrane in the washing solution for 15 minutes, then replacing the washing solution with a new one. After shaking for 5 minutes, change the washing solution again and shake for 5 minutes. If necessary, the washing solution may be further exchanged for washing.
b ) E L I S A/R I A b) E L I S A / R I A
まず、 試料を固相化させたプレートのゥエル内底面への抗体の非特異的吸着 を阻止するため、 ウエスタンプロットの塲合と同様、 予めブロッキングを行つ ておく。 ブロッキングの条件については、 ウエスタンプロットの項に記載した 通りである。  First, in order to prevent non-specific adsorption of the antibody to the inner bottom surface of the plate on which the sample is immobilized, blocking is performed in advance as in the case of Western blot. The blocking conditions are as described in the section of Western plot.
次に、 ゥエル内を 0 . 0 5〜0 . 1 % ツイーン 2 0を含む P B Sまたは T B S (以下 「洗浄液」 と記載する) で洗浄して余分なブロッキング溶液を除去 した後、 洗浄液で適宜希釈した抗 PADI4抗体を分注して一定時間ィンキュベー シヨンし、抗原に該抗体を結合させる。このときの抗体の希釈倍率は、例えば、 上記組換え抗原を段階希釈したものを試料とした予備的な E L I S A実験を行 つて決定することができる。 この抗体反応操作は、 好ましくは室温で 1時間程 度行う。 抗体反応操作終了後、 ゥエル内を洗浄液で洗浄する。 ここで、 用いた 抗体が標識されたものである場合は、 ただちに検出操作を行うことができる。 未標識の抗体を用いた場合には、 引き続き二次抗体反応を行う。 標識二次抗体 は、 例えば、 市販のものを使用する場合は洗浄液で 2 0 0 0〜2 0 0 0 0倍に 希釈して用いる (添付の指示書に好適な希釈倍率が記載されている場合は、 そ の記載に従う)。一次抗体を洗浄除去した後のゥエルに二次抗体溶液を分注して 室温で 1〜3時間インキュベーションし、 洗浄液で洗浄してから、 標識方法に 合わせた検出操作を行う。 洗浄操作は、 例えば、 まずゥエル内に洗浄液を分注 して 5分間振盪してから、 洗浄液を新しいものに交換して 5分間振盪した後、 再度洗浄液を交換して 5分間振盪することにより行う。 必要に応じてさらに洗 浄液を交換して洗浄してもよい。  Next, the inside of the well was washed with PBS or TBS containing 0.05 to 0.1% Tween 20 (hereinafter referred to as “washing solution”) to remove excess blocking solution, and then appropriately diluted with the washing solution. An anti-PADI4 antibody is dispensed and incubated for a certain period of time to bind the antibody to the antigen. The dilution ratio of the antibody at this time can be determined, for example, by conducting a preliminary ELISA experiment using a sample obtained by serially diluting the recombinant antigen. This antibody reaction operation is preferably performed at room temperature for about 1 hour. After completion of the antibody reaction procedure, wash the inside of the well with a washing solution. Here, if the antibody used is labeled, the detection operation can be performed immediately. If an unlabeled antibody is used, perform a secondary antibody reaction. When using a commercially available labeled secondary antibody, for example, dilute it to 2000 to 2000 times with a washing solution and use it (when a suitable dilution ratio is described in the attached instruction sheet). Is subject to that statement). Dispense the secondary antibody solution into the wells after washing and removing the primary antibody, incubate at room temperature for 1 to 3 hours, wash with the washing solution, and perform the detection operation according to the labeling method. The washing operation is performed, for example, by first dispensing the washing solution into the well, shaking for 5 minutes, replacing the washing solution with a new one, shaking for 5 minutes, replacing the washing solution again, and shaking for 5 minutes. . If necessary, the washing solution may be further exchanged for washing.
例えば、 本発明において、 いわゆるサンドイッチ法の E L I S Aは以下に記 載する方法により実施することができる。 まず、 本発明の PADI4蛋白質の各ァ ミノ酸配列より、 親水性に富む領域をそれぞれ 2箇所選択する。 次に、 各領域 中のアミノ酸 6残基以上からなる部分べプチドを合成し、 該部分べプチドを抗 原とした 2種類の抗体を取得する。 このうち一方の抗体を標識しておく。 標識 しなかった方の抗体は、 9 6穴 E L I S A用プレートのゥエル内底面に固相化 する。 ブロッキングの後、 試料液をゥエル内に入れて常温で 1時間インキュべ ーシヨンする。 ゥエル内を洗浄後、 標識した方の抗体希釈液を各ゥエルに分注 してインキュベーションする。 再びゥエル内を洗浄後、 標識方法に合わせた検 出操作を行う。 工程 3 :被験物質の評価 For example, in the present invention, the so-called sandwich ELISA can be performed by the method described below. First, two hydrophilic regions are selected from each amino acid sequence of the PADI4 protein of the present invention. Next, a partial peptide comprising at least 6 amino acid residues in each region is synthesized, and two types of antibodies using the partial peptide as an antigen are obtained. One of the antibodies is labeled. The unlabeled antibody was immobilized on the bottom of the well of a 96-well ELISA plate. I do. After blocking, place the sample solution in a well and incubate at room temperature for 1 hour. After washing the wells, dispense the labeled antibody diluent into each well and incubate. After washing the inside of the well again, perform the detection operation according to the labeling method. Step 3: Evaluation of test substance
最後に、 被験物質の投与または非投与条件下における、 本発明の PADI4蛋白 質の発現量の相違に基づいて、 該被験物質の R A治療薬としての効果を評価す る。  Finally, based on the difference in the expression level of the PADI4 protein of the present invention under the conditions of administration or non-administration of the test substance, the effect of the test substance as an RA therapeutic is evaluated.
すなわち、 被験物質の投与条件下で非投与条件下よりも上記蛋白質の発現量 が有意に減少している場合、 該被験物質は R A治療薬としての効果を有すると 評価できる。 ここで、 「有意に減少している」 とは、 例えば、 被験物質の投与 および非投与条件下での本発明の PADI4蛋白質の発現量に統計的有意差 (p < 0 . 0 5 ) があることを意味する。  That is, when the expression level of the protein is significantly reduced under the test substance administration condition compared to the non-administration condition, the test substance can be evaluated as having an effect as a RA therapeutic agent. Here, “significantly reduced” means, for example, that there is a statistically significant difference (p <0.05) in the expression level of the PADI4 protein of the present invention under the test substance administration and non-administration conditions. Means that.
2 . 3 PADI4蛋白質の活性を指標とした評価方法 2.3 Evaluation method based on PADI4 protein activity
PADI4活性を指標とした in vi t roにおける被験物質の R A治療薬としての効 果を評価する方法は、 例えば下記の工程を含む。 , 工程 1 :アルギニン含有化合物を含む反応液に、 被験物質の添加または非添加 条件下で、 PADI4蛋白質を加える。  A method for evaluating the effect of a test substance as an RA therapeutic agent in vitro using PADI4 activity as an index includes, for example, the following steps. , Step 1: Add PADI4 protein to the reaction mixture containing the arginine-containing compound with or without the addition of the test substance.
工程 2 :上記反応液中における反応生成物の量を定量することにより、 PADI4 蛋白質の活性を測定する。 Step 2: PADI4 protein activity is measured by quantifying the amount of the reaction product in the reaction solution.
工程 3 :被験物質の添加または非添加条件下における、 上記活性の相違に基づ いて、 該被験物質の慢性関節リゥマチ治療薬としての効果を評価する。 工程 1 :反応液の調製 Step 3: The effect of the test substance as a therapeutic agent for rheumatoid arthritis is evaluated based on the difference in the activity under the conditions of addition or non-addition of the test substance. Step 1: Preparation of reaction solution
本方法で用いられる反応液は適当な緩衝液中に PADI4蛋白質の酵素基質とな りうるアルギニン含有化合物を含むものである。 アルギニン含有化合物とは、 例 え は 、 BAEE ( N- a -benzoyl-L-arginine e thyl es ter ) 、 N- -Benzoyl-L-arginine, Dansyl- glycy卜 L- c i trul ine等のアルギニンを含む化合 物であれば特に限定されない。 また、 反応液中に添加する被験物質の量や添加 方法も特に限定されず、被験物質の性状に合わせて適宜設定すればよい。 PAD 14 と基質は 25〜5(TC付近で、 1〜2時間程度反応させることが好ましい。 工程 2 : The reaction solution used in this method contains an arginine-containing compound that can serve as an enzyme substrate for PADI4 protein in a suitable buffer. Arginine-containing compounds include, for example, arginine such as BAEE (Na-benzoyl-L-arginine e thyl ester), N--Benzoyl-L-arginine, Dansyl-glycy L-citruline, etc. Compound If it is a thing, it will not be specifically limited. The amount of the test substance to be added to the reaction solution and the method of addition are not particularly limited, and may be appropriately set according to the properties of the test substance. PAD 14 and the substrate are allowed to react at 25 to 5 (around TC for about 1 to 2 hours. Step 2:
次に、 上記基質中のアルギニン残基が PADI4によってシトルリンに変換され て生じる反応生成物の量を定量することにより、 PADI4蛋白質の活性を測定す る。 ここで 「反応生成物」 は、 酵素反応によって基質から直接生じる反応生成 物のほか、 副次的に生じる反応生成物であってもよい。 以下に、 本方法で用い られる好適な測定方法として、 蛍光法および吸光法の 2つの方法について説明 するが、 これらに限定されず、 他の公知の方法:例えば、 蛍光標識された Dans yl-glycyl-L-arginineを用いてその酵素反応生成物を HPLCを用いて定量的に 調べる方法(T. C ikura et al. , Anal lyt ical Biochemistry 285, p230-234, (2000) ) 等を任意に用いることができる。  Next, the activity of the PADI4 protein is measured by quantifying the amount of a reaction product generated by the conversion of arginine residues in the substrate to citrulline by PADI4. Here, the “reaction product” may be a reaction product directly generated from a substrate by an enzymatic reaction, or may be a reaction product generated as a by-product. Hereinafter, two methods, a fluorescence method and an absorption method, will be described as preferable measurement methods to be used in the present method. However, the present invention is not limited thereto, and other known methods: for example, fluorescently labeled dansyl-glycyl -Using L-arginine to quantitatively determine the enzymatic reaction product by HPLC (T. Cikura et al., Analytical Biochemistry 285, p230-234, (2000)) be able to.
( 1 ) 蛍光法  (1) Fluorescence method
反応液として、 例えば、 10mM CaCl2, 5 M DTTを含む lOOmM Tr is/HCl (pH7. 8) 緩衝液に、 基質として lOmM BAEEまたは Ν_ α- Benzoy卜卜 arginineを加えた溶液 を調製する。 この反応液に、 適当量の PADI4蛋白質を加え、 例えば、 37°Cで 1〜2時間程度反応させる。 反応後、 適当量の EDTAを添加して反応を停止させ、 蛍光マイクロプレートリーダーを用いて励起波長 413ηηι、 蛍光波長 476nmの各波 長を測定する。測定結果は、 PADI4蛋白質を含まない反応液での測定値をバック グラウンドとして差し引き、 検量線作成用硫酸アンモニゥムを添加した反応液 での測定値から検量線を描くことにより、 生成したアンモニゥムイオンの濃度 を求める。このアンモニゥムイオンイオン濃度を指標として PADI4蛋白質の活性 を評価する。 As a reaction solution, for example, a solution is prepared by adding lOmM BAEE or Να-benzoy arginine as a substrate to a lOOmM Tris / HCl (pH 7.8) buffer containing 10 mM CaCl 2 and 5 M DTT. To this reaction solution, an appropriate amount of PADI4 protein is added, and the reaction is performed, for example, at 37 ° C. for about 1 to 2 hours. After the reaction, stop the reaction by adding an appropriate amount of EDTA, and measure the wavelengths of the excitation wavelength of 413ηηι and the fluorescence wavelength of 476 nm using a fluorescence microplate reader. The measurement results were obtained by subtracting the measured value of the reaction solution containing no PADI4 protein as a background, and drawing a calibration curve from the measured values of the reaction solution to which ammonium sulfate was added for the preparation of a calibration curve. Determine the concentration. The activity of the PADI4 protein is evaluated using the ammonium ion concentration as an index.
( 2 ) 吸光法  (2) Absorption method
反応液として、 例えば、 10iM CaCl2 5mM DTTを含む lOOmM Tr is/HCl (pH7. 6) 緩衝液に、 基質として lOmM BAEE (Sigma) を加えた溶液を調製する。 この反応 液に、適当量の PADI4蛋白質を加え、例えば、 37°Cで 1〜2時間程度反応させる。 反応後、 δΟπιΜ Diacetyl monoxime ( 3 - hydroxyimino 2 - butane ) , 2mM thiosemicarbaz ideを含む A液と、 3M H3P04、 6M H2S04、 2mM N¾Fe (S04) 2を含む B液を 1 : 3の比率で混合した溶液を適当量添加し、サーマルサイクラ一(例え ば、 GeneAmp™ PCR Sys tem 9700 (Appl ied B iosys tems社) 等) を使用して、 例 えば、 95°C 15分次いで室温で 10分、 反応させる。反応後、 吸光度計(例えば、 Spect ra max 250 (Mol ecul ar Devices社) 等) を用いて 540nmの吸光度を測定 する。 測定結果は、 酵素を含まない反応液での測定値をバックグラウンドとし て差し引き、 さらに検量線作成用シトルリンを添加した反応液での測定値から 検量線を描くことにより、 反応によって生成したシトルリンの濃度を求める。 このシトルリン濃度を指標として PADI4蛋白質の活性を評価する。 工程 3 :被験物質の評価 As the reaction solution, for example, in lOOmM Tr is / HCl (pH7. 6) buffer containing 10iM CaCl 2 5mM DTT, a solution is prepared by adding lOmM BAEE (Sigma) as substrate. To this reaction solution, an appropriate amount of PADI4 protein is added, and the reaction is performed, for example, at 37 ° C. for about 1 to 2 hours. After the reaction, δΟπιΜ Diacetyl monoxime (3-hydroxyimino 2-butane), 2 mM and A solution containing thiosemicarbaz ide, 3M H 3 P0 4 , 6M H 2 S0 4, 2mM N¾Fe (S0 4) the B solution containing 2 1: adding an appropriate amount of mixed solution at a ratio of 3, a thermal cycler one (Eg, GeneAmp ™ PCR System 9700 (Applied Biosystems)), for example, and react at 95 ° C for 15 minutes and then at room temperature for 10 minutes. After the reaction, the absorbance at 540 nm is measured using an absorbance meter (eg, Spectra max 250 (Molecular Devices)). The measurement result was calculated by subtracting the measured value of the reaction solution containing no enzyme as a background, and drawing a calibration curve from the measured value of the reaction solution to which citrulline for preparing a calibration curve was added, to obtain the amount of citrulline generated by the reaction. Find the concentration. The activity of PADI4 protein is evaluated using the citrulline concentration as an index. Step 3: Evaluation of test substance
最後に、 被験物質の添加または非添加条件下における、 本発明の PADI4遺伝 子または本発明の PADI4蛋白質の活性の相違に基づいて、 該被験物質の R A治 療薬としての効果を評価する。  Finally, based on the difference in the activity of the PADI4 gene of the present invention or the PADI4 protein of the present invention under the conditions of adding or not adding a test substance, the effect of the test substance as an RA therapeutic drug is evaluated.
すなわち、 被験物質の添加条件下で非添加条件下よりも前記遺伝子または蛋 白質の活性が有意に減少している場合、 該被験物質は R A治療薬としての効果 を有すると評価できる。 ここで、 「有意に減少している」 とは、 例えば、 被験 物質の添加または非添加条件下での本発明の PADI4 遺伝子または本発明の PADI4蛋白質の発現量に統計的有意差(p < 0 . 0 5 )があることを意味する。  That is, when the activity of the gene or the protein is significantly reduced under the conditions of the addition of the test substance as compared with the non-addition conditions, the test substance can be evaluated as having an effect as a therapeutic agent for RA. Here, the term “significantly reduced” refers to, for example, a statistically significant difference (p <0) between the expression level of the PADI4 gene of the present invention or the PADI4 protein of the present invention under the conditions of adding or not adding a test substance. 0 5).
2 . 4 培養細胞を用いた評価方法 2.4 Evaluation method using cultured cells
培養細胞を用いた in vivoにおける被験物質の R A治療薬としての効果を評 価する方法は、 例えば下記の工程を含む。  A method for evaluating the effect of a test substance as an RA therapeutic agent in vivo using cultured cells includes, for example, the following steps.
評価方法は、 下記の工程を含むことが好ましい。 The evaluation method preferably includes the following steps.
工程 1 :細胞を被検物質の添加または非添加条件下で培養する。 Step 1: Cells are cultured under the condition of adding or not adding a test substance.
工程 2 :上記細胞中の本発明の PADI4遺伝子の発現量を検出するか、 または、 本発明の PADI4蛋白質の発現量を該蛋白質に特異的に結合する抗体を用いて検 出する。 Step 2: The expression level of the PADI4 gene of the present invention in the above cells is detected, or the expression level of the PADI4 protein of the present invention is detected using an antibody that specifically binds to the protein.
工程 3 :被検物質の添加または非添加条件下における、 上記発現量の相違に基 づいて、 該被験物質の R A治療薬としての効果を評価する。 工程 1 :細胞の培養 Step 3: The effect of the test substance as a therapeutic agent for RA is evaluated based on the difference in the expression level under the conditions of addition or non-addition of the test substance. Step 1: Cell culture
本発明の評価方法で用いられる細胞は、 本発明の PADI4遺伝子を高発現する 哺乳動物細胞であれば特に限定されない。 好ましくは哺乳動物由来の培養細胞 であって、 特に血液、 滑膜組織、 脾臓組織に由来する培養細胞 (好ましくは、 初代培養細胞) が好ましい。 前記哺乳動物としては、 ヒト、 マウス、 ラットま たはハムスター等が好ましく、 ヒトまたはマウスがより好ましい。 また、 本発 明の PADI4遺伝子をそのプロモー夕一領域とともに導入し、 該遺伝子を高発現 するように組換えた細胞など、人為的に形質転換された細胞を使用してもよい。 細胞は、 被検物質の添加または非添加条件下で培養する。 培養方法は特に限 定されず、 当該細胞に適した培養方法を適宜選択すればよい。 培養細胞への被 検物質の添加方法や添加量も特に限定されず、 上記細胞の培養前あるいは培養 期間中の適当な時点で、 被検物質を培養培地に添加し一定期間培養する。 被験 物質添加後の培養期間も特に限定されないが、 好ましくは 3 0分〜 2 4時間で ある。 工程 2 :本発明の PADI4遺伝子または本発明の PADI4蛋白質の発現量の検出 次に、 被検物質の添加または非添加条件下における、 上記細胞中の本発明の PADI4遺伝子の発現量の相違を検出するか、 または、 本発明の PADI4蛋白質の 発現量の相違を該蛋白質に特異的に結合する抗体を用いて検出する。  The cells used in the evaluation method of the present invention are not particularly limited as long as they are mammalian cells that highly express the PADI4 gene of the present invention. Cultured cells derived from mammals are preferred, and cultured cells derived from blood, synovial tissue, and spleen tissue (preferably, primary cultured cells) are particularly preferred. The mammal is preferably a human, a mouse, a rat or a hamster, and more preferably a human or a mouse. Alternatively, artificially transformed cells, such as a cell into which the PADI4 gene of the present invention is introduced together with its promoter region and which is highly expressed, may be used. The cells are cultured with or without the addition of the test substance. The culture method is not particularly limited, and a culture method suitable for the cells may be appropriately selected. The method and amount of the test substance to be added to the cultured cells are not particularly limited either. The test substance is added to the culture medium and cultured for a certain period of time before or at an appropriate time during the culture of the cells. The culture period after the addition of the test substance is not particularly limited, but is preferably 30 minutes to 24 hours. Step 2: Detection of the expression level of the PADI4 gene of the present invention or the PADI4 protein of the present invention Next, the difference in the expression level of the PADI4 gene of the present invention in the above cells under the conditions of addition or non-addition of the test substance is detected. Alternatively, the difference in the expression level of the PADI4 protein of the present invention is detected using an antibody that specifically binds to the protein.
遺伝子の検出は、 基本的に 2 . 1に記載した方法にしたがって行えばよい。 また、 抗体を用いた検出は、 2 . 2に記載した方法にしたがって行えばよい。 上記方法のほか、 本発明の PADI4遺伝子のプロモーター支配下に、 該プロモ —ター活性の検出を可能にする遺伝子(以下「レポーター遺伝子」と記載する。) を利用して、 間接的に本発明の PADI4遺伝子や本発明の PADI4蛋白質の発現を 検出することもできる。 以下、 レポ一夕一遺伝子を利用した検出方法について 説明する。  The detection of the gene may be basically performed according to the method described in 2.1. Detection using an antibody may be performed according to the method described in 2.2. In addition to the above-described methods, the present invention uses a gene (hereinafter referred to as “reporter gene”) that allows detection of the promoter activity under the control of the promoter of the PADI4 gene of the present invention. The expression of the PADI4 gene and the PADI4 protein of the present invention can also be detected. Hereinafter, a detection method using the repo overnight gene will be described.
( 1 ) レポーター遺伝子  (1) Reporter gene
レポ一夕一遺伝子は、 宿主細胞が本試験方法の一連の過程において産生し得 る他のいかなる蛋白質とも特異的に区別可能な、 レポーター蛋白質をコードす るものであればよい。 好ましくは、 形質転換前の細胞が該レポーター蛋白質と 同一または類似の蛋白質をコードする遺伝子を持たないようなものがよい。 例 えば、 レポーター蛋白質が該細胞に対して毒性を有するようなものや、 該細胞 が感受性を有する抗生物質の耐性を付与するものであるような場合でも、 レポ 一夕一遺伝子の発現の有無は細胞の生存率で判定することが可能である。 しか しながら、 本発明で用いられるレポーター遺伝子としてより好ましいものは、 発現量を特異的かつ定量的に検出することができる (例えば、 該レポーター遺 伝子にコードされる蛋白質に対する特異的抗体が取得されているような) 構造 遺伝子である。 より好ましくは、 外来の基質と特異的に反応することにより定 量的測定が容易な代謝産物を生じるような酵素等をコードする遺伝子である。 そのようなレポーター遺伝子としては、 例えば、 以下の蛋白質をコードする遺 伝子を例示することができるが、 本発明はそれらに限定されない。 The repo overnight gene encodes a reporter protein that is specifically distinguishable from any other protein that the host cell may produce in the course of this test method. Anything can be used. Preferably, the cells before transformation do not have a gene encoding the same or similar protein as the reporter protein. For example, even when the reporter protein is toxic to the cell or confers resistance to a susceptible antibiotic, the presence or absence of expression of the repo overnight gene can be determined. It can be determined by the survival rate of the cells. However, a more preferable reporter gene used in the present invention can specifically and quantitatively detect the expression level (for example, when a specific antibody against the protein encoded by the reporter gene is obtained). It is a structural gene. More preferably, it is a gene encoding an enzyme or the like which specifically reacts with a foreign substrate to produce a metabolite which can be easily measured quantitatively. Examples of such a reporter gene include, for example, genes encoding the following proteins, but the present invention is not limited thereto.
a) クロラムフエニコールァセチルトランスフェラーゼ: a) Chloramphenicol acetyltransferase:
クロラムフエ二コールにァセチル基を付加する酵素で、 いわゆる CATアツ セィ等で検出することができる。 プロモーターを組み込むだけでレポ一夕ーァ ッセィ用のベクターを調製できるベクタ一として、 pCAT3— B a s i cベ クタ一 (プロメガ社製) が市販されている。  An enzyme that adds an acetyl group to chloramphenicol, and can be detected by so-called CAT technology. PCAT3-BASIC vector (Promega) is commercially available as a vector that can be used to prepare a vector for repo assay simply by incorporating a promoter.
b) ホタルルシフェラーゼ: b) Firefly luciferase:
ルシフェリンを代謝した際に生じる生物発光を測定することにより定量でき る。 レポ一夕一アツセィ用のベクターとしては、 p GL 3— B a s i cベクタ 一 (プロメガ社製) が市販されている。  It can be quantified by measuring the bioluminescence generated when luciferin is metabolized. As a vector for repo overnight, pGL3-BASIC vector-1 (promega) is commercially available.
c) β一ガラクトシダーゼ: c) β-galactosidase:
呈色反応、 蛍光または化学発光でそれぞれ測定可能な基質がある。 レポ一夕 一アツセィ用のベクターとしては、 p/3 g a l— B a s i c (プロメガ社製) が市販されている。  Some substrates can be measured by color reaction, fluorescence or chemiluminescence, respectively. As a vector for repo overnight, p / 3gal-Basic (promega) is commercially available.
d) 分泌型アルカリホスファターゼ: d) Secreted alkaline phosphatase:
呈色反応、 生物発光または化学発光でそれぞれ測定可能な基質がある。 レポ 一夕一アツセィ用のベクタ一としては、 pSEAP 2— B a s i c (クロンテ ック社製) が市販されている。  There are substrates that can be measured by color reaction, bioluminescence, or chemiluminescence, respectively. As a vector for repo overnight, pSEAP2-Basic (Clontech) is commercially available.
e ) 緑色堂光蛋白質 (green-fluorescent protein) : 酵素ではないが、 自らが蛍光を発するので直接定量できる。 同じくレポ一夕 一アツセィ用のベクタ一として pEGFP— 1 (クロンテック社製) が市販さ れている。 e) Green-fluorescent protein: Although it is not an enzyme, it can quantify directly because it emits fluorescence. Similarly, pEGFP-1 (Clontech) is commercially available as a vector for repo overnight.
(2) レポーター遺伝子の導入  (2) Introduction of reporter gene
公知の方法に従い、 レポーター遺伝子発現プラスミドと、 本発明の本発明の PADI4 遺伝子を哺乳類細胞で発現可能にした組換えベクターを作製し、 これら を細胞に同時トランスフエクシヨンする。ベクタ一としては、 pCR 3. 1 (ィ ンビトロジェン社製)、 pCMV— S c r i p t (ストラ夕ジ一ン社製)等を好 適に用いることができるが、 これらに限定されない。  According to a known method, a reporter gene expression plasmid and a recombinant vector in which the PADI4 gene of the present invention of the present invention can be expressed in mammalian cells are prepared, and these are cotransfected into cells. As the vector, pCR3.1 (manufactured by Invitrogen), pCMV-Script (manufactured by Straugene), and the like can be preferably used, but are not limited thereto.
細胞に発現プラスミドを導入する方法としては、 DEAE—デキストラン法 (Luthman, H. and Magnus son, G. (1983) Nucleic Acids Res. 11, 1295-1308)、 リン酸カルシウム一 DNA共沈殿法 (Graham, F. L. and van der Eb, A. J. (1973) Virology 52, 456-457)、 電気パルス穿孔法 (Neumann, E. eUl. (1982) EMBO J. 1, 841-845), リボフェクシヨン法 (Lopata et al. (1984) Nucl. Acids Res. 12, 5707-5717, Sussman and Milman (1984) Mol. Cell. Biol. 4, 1641-1643)等を挙げることができる。 ただし、 細胞がいわゆる浮遊細胞である 場合は、 リン酸カルシウム—DNA共沈殿法以外の方法を用いることが好まし レ^ いずれの方法においても、 用いる細胞に応じて、 至適化されたトランスフ ェクション条件を用いることが必要である。  Methods for introducing an expression plasmid into cells include the DEAE-dextran method (Luthman, H. and Magnus son, G. (1983) Nucleic Acids Res. 11, 1295-1308), calcium phosphate-DNA coprecipitation method (Graham, FL and van der Eb, AJ (1973) Virology 52, 456-457), electric pulse perforation (Neumann, E. eUl. (1982) EMBO J. 1, 841-845), ribofection (Lopata et al. (1984) ) Nucl. Acids Res. 12, 5707-5717, Sussman and Milman (1984) Mol. Cell. Biol. 4, 1641-1643). However, if the cells are so-called floating cells, it is preferable to use a method other than the calcium phosphate-DNA coprecipitation method. In any of these methods, optimized transfection conditions are used according to the cells used. It is necessary to use.
(3) 評価  (3) Evaluation
本発明の PADI4遺伝子の発現ベクターと、 レポーター発現ベクターを同時ト ランスフエクションした細胞を培養すると、 該遺伝子の発現依存的にレポ一夕 一遺伝子の転写が促進される。 したがって、 レポ一夕一遺伝子の発現が可能な 条件下において、 培地中に任意の被検物質を添加した場合と添加しない場合で のレポーター遺伝子の発現量変化をみれば、 本発明の PADI4遺伝子の発現量が 評価できる。 ここで、 「レポーター遺伝子の発現が可能な条件」 とは、 レポ一夕 一発現ベクターによってトランスフエク卜された細胞が生存して、 レポーター 遺伝子の転写産物 (レポーター蛋白質) の生産が可能な条件であればよい。 好 ましくは、 使用される細胞株に適合した培地 (ゥシ胎児血清等の血清成分を添 加してもよい) で、 4〜6% (最も好適には 5%) の炭酸ガスを含む空気存在 下、 3 6〜 3 8 °C (最も好適には 3 7 °C) で 2〜 3日間 (最も好適には 2日間) 培養する。 When cells transfected with the PADI4 gene expression vector of the present invention and a reporter expression vector are co-transfected, transcription of the repo overnight gene is promoted depending on the expression of the gene. Therefore, the change in the expression level of the reporter gene with and without the addition of any test substance in the medium under conditions that allow for the expression of the repo overnight gene indicates the PADI4 gene of the present invention. The expression level can be evaluated. Here, the “conditions under which the reporter gene can be expressed” are defined as conditions under which the cells transfected by the repo overnight expression vector survive and can produce a transcript (reporter protein) of the reporter gene. I just need. Preferably, the medium is suitable for the cell line used (or may contain serum components such as fetal bovine serum) and contains 4 to 6% (most preferably 5%) carbon dioxide. Air presence Incubate at 36-38 ° C (most preferably 37 ° C) for 2-3 days (most preferably 2 days).
( 4 ) その他 (形質転換細胞株の樹立)  (4) Others (establishment of transformed cell line)
以上のような、 一過的な遺伝子導入法を利用した試験方法とは別に、 レポ一 夕一遺伝子、 および本発明の PADI4遺伝子の発現ベクターで、 宿主細胞を二重 に形質転換した細胞を利用した試験方法も採択可能である。 この場合には、 p I ND (インビトロジェン社製) や p T e t— O n (クロンテック社製) 等の 発現ベクターを利用して、 本発明の PADI4遺伝子の発現を誘導する条件下で該 レポ一夕一遺伝子の発現が促進されるような細胞株を樹立することが必要とな る。 形質転換細胞の作出においては、 導入される遺伝子は、 宿主細胞の染色体 に組み込まれ、継代を重ねても宿主細胞に安定的に保持されることが望ましい。 そのように形質転換された細胞を選択するために、 導入遺伝子に抗生物質耐性 等の選択マーカー (例えば、 ネオマイシン (または G 4 1 8 ) 耐性遺伝子 n e o等) を連結してトランスフエクシヨンしたり、 もしくは別個に調製した該選 択マーカーと導入遺伝子とを同時トランスフエクシヨンすることが好ましい。 その後は、 該選択マーカ一の特性を利用することにより、 安定的に形質転換さ れた細胞を選択することができる。  Apart from the test method using the transient gene transfer method as described above, a cell obtained by double-transforming a host cell with the expression vector for the repo allelic gene and the PADI4 gene of the present invention is used. Test methods that have been adopted can also be adopted. In this case, an expression vector such as pIND (manufactured by Invitrogen) or pTet-On (manufactured by Clontech) is used to induce the expression of the reporter gene under conditions that induce the expression of the PADI4 gene of the present invention. It is necessary to establish a cell line that promotes expression of the gene. In producing transformed cells, it is desirable that the gene to be introduced be integrated into the chromosome of the host cell and be stably retained in the host cell even after repeated passages. In order to select cells transformed in such a manner, a transgene is ligated with a selectable marker such as an antibiotic resistance or the like (for example, neomycin (or G418) resistance gene neo or the like) and transfected. Alternatively, it is preferable to simultaneously transfect the separately prepared selectable marker and the transgene. Thereafter, by utilizing the characteristics of the selectable marker, stably transformed cells can be selected.
こうして得られた細胞株を、 本発明の PADI4遺伝子の発現が誘導される条件 下におくと、 該遺伝子の発現依存的にレポーター遺伝子の転写が促進される。 したがって、 レポーター遺伝子の発現が可能な条件下において、 培地中に任意 の被検物質を添加した場合と添加しない場合でのレポーター遺伝子の発現量変 化をみれば、 本発明の PADI4の発現量が評価できる。 工程 3 :被験物質の評価  When the cell line thus obtained is placed under conditions that induce the expression of the PADI4 gene of the present invention, the transcription of the reporter gene is promoted in a manner dependent on the expression of the gene. Therefore, under the conditions where the reporter gene can be expressed, the expression level of the PADI4 of the present invention can be seen from the change in the expression level of the reporter gene with and without the addition of any test substance in the medium. Can be evaluated. Step 3: Evaluation of test substance
最後に、 被験物質の添加または非添加条件下における、 本発明の PADI4遺伝 子または本発明の PADI4蛋白質の発現量の相違に基づいて、 該被験物質の R A 治療薬としての効果を評価する。  Finally, the effect of the test substance as a therapeutic agent for RA is evaluated based on the difference in the expression level of the PADI4 gene of the present invention or the PADI4 protein of the present invention under the conditions of adding or not adding the test substance.
すなわち、 被験物質の添加条件下で非添加条件下よりも前記遺伝子または蛋 白質の発現量が有意に減少している場合、 該被験物質は R A治療薬としての効 果を有すると評価できる。 ここで、 「有意に減少している」 とは、 例えば、 被 験物質の添加または非添加条件下での本発明の PADI4遺伝子または本発明の PADI4蛋白質の発現量に統計的有意差(p<0. 05)があることを意味する。 That is, when the expression level of the gene or protein is significantly reduced under the conditions of the addition of the test substance as compared with the non-addition conditions, the test substance can be evaluated as having an effect as a therapeutic agent for RA. Here, “significantly decreases” means, for example, It means that there is a statistically significant difference (p <0.05) in the expression level of the PADI4 gene of the present invention or the PADI4 protein of the present invention under the conditions of adding or not adding the test substance.
2. 5 被験物質の RA治療薬としての効果を評価するためのキット 2.5 Kits for evaluating the effects of test substances as RA therapeutics
本発明の評価方法を実施するためのキットとして、例えば、 以下の a)〜: O の少なくとも一つ以上を含むキットを挙げることができる。  Examples of the kit for performing the evaluation method of the present invention include a kit containing at least one of the following a) to: O 2.
a) ヒト PADI4遺伝子 (配列番号 16) またはマウス PADI4遺伝子 (配列番号a) Human PADI4 gene (SEQ ID NO: 16) or mouse PADI4 gene (SEQ ID NO: 16)
17) を特異的に増幅するための、 15〜30塩基長の連続したオリゴヌクレ ォチドプライマ一 17) A continuous oligonucleotide primer of 15 to 30 bases in length to specifically amplify
b) ヒト PADI4遺伝子またはマウス PADI4遺伝子に特異的に結合し、 該遺伝子 を検出するための 20〜1500塩基長の連続したポリヌクレオチドプロ一ブ c) 上記 b) 記載のポリヌクレオチドプローブが固定された固相化試料 d) ヒト PADI4蛋白質 (配列番号 17) またはマウス PADI4蛋白質 (配列番号b) A continuous polynucleotide probe of 20 to 1500 bases in length that specifically binds to the human PADI4 gene or mouse PADI4 gene and detects the gene c) The polynucleotide probe described in b) above was immobilized Immobilized sample d) Human PADI4 protein (SEQ ID NO: 17) or mouse PADI4 protein (SEQ ID NO:
18) に特異的に結合し、 該蛋白質を検出するための抗体 18) an antibody that specifically binds to and detects the protein
e) 上記 d) 記載の抗体に特異的に結合しうる二次抗体 e) secondary antibody capable of specifically binding to the antibody described in d) above
f ) ヒト PADI4蛋白質  f) Human PADI4 protein
前記 a) 記載のプライマーは、 上記遺伝子の塩基配列 (配列番号 16または 17) に基づき、 市販のプライマー設計ソフトを用いる等、 常法にしたがい容 易に設計し、 増幅することができる。 このようなプライマ一の例としては、 例 えば、 配列番号 7〜 10に記載の塩基配列を有するォリゴヌクレオチドを挙げ ることができる。  The primer described in a) above can be readily designed and amplified according to a conventional method, for example, using commercially available primer design software, based on the nucleotide sequence of the above gene (SEQ ID NO: 16 or 17). Examples of such primers include, for example, oligonucleotides having the nucleotide sequences of SEQ ID NOs: 7 to 10.
前記 b) 記載のプローブは、 ノーザンハイプリダイゼ一シヨン法であれば、 20塩基長程度の 1本鎖オリゴヌクレオチドか 2本鎖 DNAが好適に用いられ る。 マイクロアレイであれば、 100〜1500塩基長程度の 2本鎖 DNA、 または 20〜100塩基長程度の 1本鎖オリゴヌクレオチドが好適に用いられ る。 Affimetrix社の Gene Chipシステムであれば 25塩基長程度の 1本鎖オリ ゴがよい。 これらは、 特に本発明の PADI4遺伝子の配列特異性が高い部分に特 異的にハイブリダィズするプローブとして設計することが好ましい。 このよう なプローブの例としては、 例えば配列番号 11に記載の塩基配列を有するオリ ゴヌクレオチドを挙げることができる。 これらのプローブや前述のプライマー は、 適当な標識によりラベル (例えば、 酵素標識、 放射性標識、 蛍光標識等) されていてもよく、 またピオチン、 リン酸、 アミン等により修飾されていても よい。 The probe described in b) above is preferably a single-stranded oligonucleotide or double-stranded DNA having a length of about 20 bases in the case of the Northern hybridization method. In the case of a microarray, a double-stranded DNA having a length of about 100 to 1500 bases or a single-stranded oligonucleotide having a length of about 20 to 100 bases is suitably used. For the Affimetrix Gene Chip system, a single-stranded oligo with a length of about 25 bases is preferred. These are preferably designed as probes that specifically hybridize to a portion of the PADI4 gene of the present invention having high sequence specificity. Examples of such a probe include, for example, an oligonucleotide having the nucleotide sequence of SEQ ID NO: 11. These probes and the aforementioned primers May be labeled with an appropriate label (for example, an enzyme label, a radioactive label, a fluorescent label, or the like), or may be modified with biotin, phosphoric acid, an amine, or the like.
前記 c ) 記載の固相化試料は、 前記 b ) 記載のプローブをガラス板、 ナイ口 ンメンプレン、 マイクロビーズ、 シリコンチップ等の固相に固定することによ り作製される。 このような固相化試料とその作製方法については、 既に 2 . 1 で説明したが、 例えば、 遺伝子チップ、 c D NAアレイ、 オリゴアレイ、 メン プレンフィルター等を挙げることができる。  The solid-phased sample described in c) above is produced by fixing the probe described in b) above to a solid phase such as a glass plate, a nylon membrane, microbeads, or a silicon chip. The immobilized sample and the method for preparing the same have already been described in 2.1, but examples thereof include a gene chip, a cDNA array, an oligo array, and a membrane filter.
前記 d ) および e ) 記載の抗体や f ) の蛋白質は、 2 . 2に記載した方法に より作製することができる。 抗体は、 適当な標識によりラベル (例えば、 酵素 標識、 放射性標識、 蛍光標識等) されていてもよいし、 ピオチン等により適当 に修飾されていてもよい。  The antibodies described in d) and e) and the protein in f) can be prepared by the method described in 2.2. The antibody may be labeled with a suitable label (for example, an enzyme label, a radioactive label, a fluorescent label, or the like), or may be appropriately modified with biotin or the like.
前記キットは上記した構成要素のほか、 必要に応じて、 ハイブリダィゼーシ ョン、プローブの標識、あるいはラベル体の検出のための試薬、反応用緩衝液、 酵素基質等、 本発明の評価方法の実施に必要な、 その他の試薬等を適宜含んで いてもよい。  In addition to the components described above, the kit may be used for evaluation of the present invention, such as hybridization, probe labeling, or a reagent for detecting a label, a reaction buffer, and an enzyme substrate, if necessary. It may optionally contain other reagents and the like necessary for carrying out the method.
3 . 変異型 PADI4を利用した慢性関節リゥマチの発症危険度を予測する方法 本発明は、 検体中の配列番号 2 1に示されるアミノ酸配列からなる変異型 PADI4蛋白質、 またはその遺伝子 (配列番号 2 0 ) の発現量に基づいて、 該検 体を提供した被験者の慢性関節リウマチ (以下、 「R A」 と記載する。) の発症 危険度を予測する方法を提供する。 3. Method for Predicting Risk of Onset of Rheumatoid Arthritis Using Mutant PADI4 The present invention relates to a mutant PADI4 protein comprising the amino acid sequence shown in SEQ ID NO: 21 in a sample, or a gene thereof (SEQ ID NO: 20). The present invention provides a method for predicting the risk of developing rheumatoid arthritis (hereinafter referred to as “RA”) in a subject who has provided the test sample, based on the expression level of the test sample.
前記方法において用いられる 「検体」 とは、 被験者から単離された、 血液、 体液、組織または排泄物等のすべての試料を意味するが、血液(血液由来細胞)、 滑膜組織 (滑膜細胞)、 脾臓組織 (脾臓細胞)、 腹腔浸潤細胞を含むものが好ま しく、 特に血球 (好中球、 好酸球)、 滑膜組織 (滑膜細胞) がより好ましい。 ま た、 発症危険度の予測は、 変異型 PADI4蛋白質の発現量を指標として行っても よいし、変異型 PADI4遺伝子(mR NA)の発現量を指標として行ってもよい。 3 . 1 変異型 PADI4遺伝子の発現量を指標とした方法 PADI4遺伝子の発現量を指標とした予測方法は、 例えば下記の工程を含む。 工程 1 :被験者および正常人から単離された各検体より全 R NAを調製する; 工程 2 :上記全 R NA中における変異型 PADI4遺伝子の mR NAの発現量を検 出する; The term “specimen” used in the above-mentioned method means any sample such as blood, body fluid, tissue, or excrement isolated from a subject, and includes blood (blood-derived cells), synovial tissue (synovial cells) ), Spleen tissue (spleen cells), and cells containing peritoneal cells are preferred, and blood cells (neutrophils, eosinophils) and synovial tissue (synovial cells) are more preferred. The risk of onset may be predicted using the expression level of the mutant PADI4 protein as an index or the expression level of the mutant PADI4 gene (mRNA) as an index. 3.1 Method based on the expression level of mutant PADI4 gene The prediction method using the expression level of the PADI4 gene as an index includes, for example, the following steps. Step 1: preparing all RNAs from each sample isolated from a subject and a normal person; Step 2: detecting the expression level of mRNA of the mutant PADI4 gene in all the above RNAs;
工程 3 :被験者と正常人における上記発現量の相違を解析し、 被験者の慢性関 節リゥマチの発症危険度を予測する。 Step 3: Analyze the difference in the above expression level between the subject and the normal person, and predict the risk of developing the subject's chronic joint rheumatism.
前記方法において、 全 R NAの抽出および変異型 PADI4遺伝子の mR NAの 発現量の検出と解析は、 2 . 1の記載にしたがって実施することができる。 そ の結果、 被験者において変異型 PADI4の mR NAの発現量が正常人に比較して 有意に高い場合、 該被験者は R Aを発症する危険度が高いと評価できる。 ここ で、 「有意に高い」 とは、 例えば、 被験者と正常人の間で変異型 PADI4の mR NAの発現量に統計的有意差 (p < 0 . 0 5 ) があることを意味する。 なお、 前述したように、 評価は変異型 PADI4遺伝子と検体内の適当なコントロール遺 伝子 (例えば、 3ァクチン遺伝子ゃリポゾ一マル R NA等) との発現量比とし て、 上記解析に供してもよい。  In the above method, extraction of all RNAs and detection and analysis of the mRNA expression level of the mutant PADI4 gene can be performed according to the description in 2.1. As a result, if the expression level of the mutant PADI4 mRNA is significantly higher in a subject than in a normal person, the subject can be evaluated as having a high risk of developing RA. Here, “significantly higher” means that, for example, there is a statistically significant difference (p <0.05) in the expression level of the mutant PADI4 mRNA between the subject and a normal person. As described above, the evaluation was performed using the ratio of the expression level of the mutant PADI4 gene and the appropriate control gene (for example, 3-actin gene ゃ liposomal RNA, etc.) in the sample, and subjected to the above analysis. Is also good.
3 . 2 変異型 PADI4蛋白質の発現量を指標とした評価方法 3.2 Evaluation method based on the expression level of mutant PADI4 protein
変異型 PADI4蛋白質の発現量を指標とした方法は、例えば下記の工程を含む。 工程 1 :被験者および正常人から単離された検体中における、 変異型 PADI4蛋 白質の発現量を該蛋白質に特異的に結合しうる抗体を用いて検出する; 工程 2 :被験者と正常人における上記発現量の相違を解析し、 該被験者の RA の発症危険度を予測する。  The method using the expression level of the mutant PADI4 protein as an index includes, for example, the following steps. Step 1: detecting the expression level of the mutant PADI4 protein in a sample isolated from a subject and a normal person using an antibody capable of specifically binding to the protein; Step 2: detecting the expression level in the subject and a normal person The difference in the expression level is analyzed to predict the risk of developing RA in the subject.
前記方法において、 変異型 PADI4蛋白質の発現量の検出と解析は、 2 . 2の 記載にしたがって実施することができる。 その結果、 被験者において変異型 PADI4蛋白質の発現量が正常人に比較して有意に高い場合、 該被験者は RAを 発症する危険度が高いと評価できる。 ここで、 「有意に高い」 とは、 例えば、 被験者と正常人の間で変異型 PADI4蛋白質の発現量に統計的有意差 (p < 0 . In the above method, detection and analysis of the expression level of the mutant PADI4 protein can be performed according to the description in 2.2. As a result, when the expression level of the mutant PADI4 protein in the subject is significantly higher than that in a normal person, the subject can be evaluated as having a high risk of developing RA. Here, “significantly high” means, for example, a statistically significant difference in the expression level of mutant PADI4 protein between a subject and a normal person (p <0.
0 5 ) があることを意味する。 3 . 3 慢性関節リウマチの発症危険度を予測するためのキット 本発明はまた、 慢性関節リウマチの発症危険度を予測するためのキットを提 供する。該キットは、例えば、以下の a ) ~ e )の少なくとも一つ以上を含む。 a ) 配列番号 2 0に示される塩基配列からなる変異型 PADI4遺伝子を特異的に 増幅するための、 1 5〜3 0塩基長の連続したオリゴヌクレオチドプライマ一 b ) 配列番号 2 0に示される塩基配列からなる変異型 PADI4遺伝子に特異的に ハイブリダィズし、 該遺伝子を検出するための 2 0〜1 5 0 0塩基長の連続し たボリヌクレオチドプローブ 0 5). 3.3 Kit for predicting the risk of developing rheumatoid arthritis The present invention also provides a kit for predicting the risk of developing rheumatoid arthritis. The kit includes, for example, at least one of the following a) to e). a) a 15- to 30-base-length continuous oligonucleotide primer for specifically amplifying a mutant PADI4 gene consisting of the base sequence shown in SEQ ID NO: 20 b) the base shown in SEQ ID NO: 20 A continuous polynucleotide probe with a length of 20 to 150 bases for specifically hybridizing to the mutant PADI4 gene comprising the sequence and detecting the gene.
c ) 上記 b ) 記載のポリヌクレオチドプローブが固定された固相化試料 d ) 配列番号 2 1に示されるアミノ酸配列からなる変異型 PADI4蛋白質に特異 的に結合し、 該蛋白質を検出するための抗体 c) An immobilized sample on which the polynucleotide probe described in b) above is immobilized d) An antibody specifically binding to the mutant PADI4 protein having the amino acid sequence shown in SEQ ID NO: 21 and detecting the protein
e ) 上記 d ) 記載の抗体に特異的に結合しうる二次抗体 前記 a ) 記載のプライマーは、 上記遺伝子の塩基配列 (配列番号 2 0または 2 1 ) に基づき、 市販のプライマ一設計ソフトを用いる等、 常法にしたがい容 易に設計し、 増幅することができる。 また、 前記 b ) 記載のプローブは、 ノー ザンハイブリダィゼーシヨン法であれば、 2 0塩基長程度の 1本鎖オリゴヌク レオチドか 2本鎖 D NAが好適に用いられる。 マイクロアレイであれば、 1 0 0〜1 5 0 0塩基長程度の 2本鎖 D NA、 または 2 0〜1 0 0塩基長程度の 1 本鎖ォリゴヌクレオチドが好適に用いられる。 Af me t r ix社の Gene Ch ipシス テムであれば 2 5塩基長程度の 1本鎖オリゴがよい。 これらは、 特に変異型 PADI4遺伝子の配列特異性が高い部分に特異的にハイブリダィズするプローブ として設計することが好ましい。 このようなプローブの例としては、 例えば配 列番号 4に記載の塩基配列を有するオリゴヌクレオチドを挙げることができる。 これらのプローブや前述のプライマーは、 適当な標識によりラベル (例えば、 酵素標識、 放射性標識、 蛍光標識等) されていてもよく、 またピオチン、 リン 酸、 アミン等により修飾されていてもよい。 e) A secondary antibody capable of specifically binding to the antibody described in d) above. The primer described in a) is a commercially available primer design software based on the nucleotide sequence of the above gene (SEQ ID NO: 20 or 21). It can be easily designed and amplified according to the usual method such as using. In the Northern hybridization method, a single-stranded oligonucleotide or double-stranded DNA having a length of about 20 bases is suitably used as the probe described in b) above. In the case of a microarray, a double-stranded DNA having a length of about 100 to 150 bases or a single-stranded oligonucleotide having a length of about 200 to 100 bases is suitably used. In the case of the Gene Chip system of Afmetrix, a single-stranded oligo with a length of about 25 bases is preferred. These are preferably designed as probes that specifically hybridize to a portion having high sequence specificity of the mutant PADI4 gene. Examples of such a probe include, for example, an oligonucleotide having the base sequence of SEQ ID NO: 4. These probes and the aforementioned primers may be labeled with an appropriate label (for example, an enzyme label, a radioactive label, a fluorescent label, or the like), or may be modified with biotin, phosphoric acid, an amine, or the like.
前記 c ) 記載の固相化試料は、 前記 b ) 記載のプローブをガラス板、 ナイ口 ンメンプレン、 マイクロピーズ、 シリコンチップ等の固相に固定することによ り作製される。 このような固相化試料とその作製方法については、 既に 2 . 1 で説明したが、 例えば、 遺伝子チップ、 c D NAアレイ、 オリゴアレイ、 メン プレンフィルタ一等を挙げることができる。前記 d )および e )記載の抗体は、The solid-phased sample described in c) above is prepared by fixing the probe described in b) above to a solid phase such as a glass plate, a nylon membrane, micropease, or a silicon chip. Such immobilized samples and their preparation methods have already been described in 2.1. For example, gene chips, cDNA arrays, oligo arrays, One example is a plain filter. The antibody according to d) and e),
2 . 2に記載した方法により作製することができる。 抗体は、 適当な標識によ りラベル(例えば、酵素標識、放射性標識、蛍光標識等) されていてもよいし、 ピオチン等により適当に修飾されていてもよい。 It can be produced by the method described in 2.2. The antibody may be labeled with an appropriate label (for example, an enzyme label, a radioactive label, a fluorescent label, or the like), or may be appropriately modified with biotin or the like.
前記キットは上記した構成要素のほか、 必要に応じて、 ハイブリダィゼーシ ヨン、 プローブの標識、 あるいはラベル体の検出のための試薬、 反応用緩衝液 等、 本発明の方法の実施に必要な、 その他の試薬等を適宜含んでいてもよい。  The kit is, in addition to the components described above, necessary for the implementation of the method of the present invention, such as a hybridization, a reagent for detecting the label of the probe or a label, a reaction buffer, etc. Further, other reagents and the like may be appropriately contained.
3 . 4 R A診断用マーカー . 3.4 RA diagnostic marker.
変異型 PADI4蛋白質ゃ該遺伝子は、 本発明者らによって新規に見出されたも のであり、 これら変異型 PADI4蛋白質ゃ該遺伝子と正常型 PADI4蛋白質ゃ該遺 伝子との機能的な違いは、 R A発症と深い関連を有する。 すなわち、 配列番号 2 1に示されるアミノ酸配列からなる変異型 PADI4蛋白質や、 配列番号 2 0に 示される塩基配列からなる変異型 PADI4遺伝子は、 R Aの診断用マーカーとし て (例えば、 抗変異型 PADI4抗体作製のための抗原として) 有用である。 本発 明はそのような変異型 PADI4を利用した R A診断用マーカーも提供する。  Mutant PADI4 protein ゃ The gene was newly discovered by the present inventors, and the functional difference between these mutant PADI4 protein ゃ gene and normal PADI4 protein ゃ gene is It is closely related to RA onset. That is, a mutant PADI4 protein consisting of the amino acid sequence shown in SEQ ID NO: 21 and a mutant PADI4 gene consisting of the nucleotide sequence shown in SEQ ID NO: 20 are used as diagnostic markers for RA (for example, anti-mutant PADI4 Useful as an antigen for producing antibodies). The present invention also provides RA diagnostic markers using such mutant PADI4.
4. 変異型 PADI4を利用した R A治療薬のスクリ一ニング方法 4. Screening method of RA therapeutic drug using mutant PADI4
変異型 PADI4蛋白質は正常型 PADI4蛋白質に比較して酵素活性が高く、 シト ルリン化ペプチド生成の亢進を介して、 R A発症の一つの原因となりうる。 し たがって、 変異型 PADI4蛋白質に対する特異的な阻害効果を指標とすることに より、 R A治療薬のスクリーニングも可能となる。  The mutant PADI4 protein has a higher enzyme activity than the normal PADI4 protein, and may be one of the causes of RA onset through enhanced production of citrullinated peptides. Therefore, by using the specific inhibitory effect on the mutant PADI4 protein as an index, screening for RA therapeutic agents is possible.
すなわち、 本発明は、 配列番号 2 1に示されるアミノ酸配列からなる変異型 PADI4蛋白質に対する阻害効果を指標として、 被験物質の慢性関節リウマチ治 療薬としての効果を評価する方法を提供する。  That is, the present invention provides a method for evaluating the effect of a test substance as a therapeutic drug for rheumatoid arthritis using the inhibitory effect on the mutant PADI4 protein consisting of the amino acid sequence shown in SEQ ID NO: 21 as an index.
例えば、 前記方法は下記の工程を含む。  For example, the method includes the following steps.
工程 1 :アルギニン含有化合物を含む反応液に、 被験物質の添加または非添加 条件下で、 変異型 PADI4蛋白質を加える; Step 1: Add the mutant PADI4 protein to the reaction solution containing the arginine-containing compound, with or without the addition of the test substance;
工程 2 :上記反応液中における反応生成物の量を定量することにより、 変異型 PADI4蛋白質の活性を測定する; 工程 3 :被験物質の添加または非添加条件下における、 上記活性の相違に基づ いて、 該被験物質の慢性関節リゥマチ治療薬としての効果を評価する。 Step 2: measuring the activity of the mutant PADI4 protein by quantifying the amount of the reaction product in the reaction solution; Step 3: The effect of the test substance as a therapeutic agent for rheumatoid arthritis is evaluated based on the difference in the activity under the conditions of addition or non-addition of the test substance.
前記方法において、 変異型 PADI4蛋白質は、 2 . 2の記載にしたがって調製 することができ、 工程 1〜3は 2 . 3の記載にしたがって実施することができ る。  In the above method, the mutant PADI4 protein can be prepared as described in 2.2, and steps 1 to 3 can be performed as described in 2.3.
5 . その他 5. Other
5 . 1 PADI4を利用した R A病態の予測方法  5.1 Prediction method of RA disease state using PADI4
ヒ卜 PADI4遺伝子や PADI4蛋白質は、 R A患者の病変組織 (例えば、 滑膜組 織) で特異的に高発現し、 シトルリン化タンパク質の生成を通じて R Aの原因 となりうる。 したがって、 前述した変異型 PADI4のみならず、 被験者から単離 された検体中の、 PADI4遺伝子や PADI4蛋白質の発現量を測定すれば、 該被験 者の R A病態を予測することもできると考えられる。 5 . 2 R Aモデル動物  The human PADI4 gene and PADI4 protein are specifically highly expressed in diseased tissues (eg, synovial tissue) of RA patients, and may cause RA through the production of citrullinated proteins. Therefore, by measuring the expression levels of the PADI4 gene and PADI4 protein in the specimen isolated from the subject as well as the mutant PADI4 described above, the RA pathology of the subject can be predicted. 5.2 RA model animal
本発明の PADI4遺伝子や変異型 PADI4遺伝子を人為的に高発現させることに より、 R A病態を呈する動物 (例えば、 マウス等) を作製することも可能であ る。 例えば、 本発明の PADI4遺伝子または変異型 PADI4遺伝子を適当なベクタ 一に組み込んで、 動物に導入し、 ヒト R Aに代表される炎症症状等の表現的変 化が現れれば、 該動物を利用して R Aの病態やその治療薬の研究を行うことが できる。 同様に、 本発明の PADI4蛋白質または変異型 PADI4蛋白質を直接動物 に導入することによって、 R A病態を呈するモデル動物を作製することも可能 と考えられる。 以下に実施例を用いて本発明をさらに具体的に説明するが、 本発明はこれら に限定されるものではない。  By artificially expressing the PADI4 gene or mutant PADI4 gene of the present invention at a high level, an animal (eg, a mouse or the like) exhibiting an RA pathological condition can be produced. For example, the PADI4 gene or the mutant PADI4 gene of the present invention is incorporated into an appropriate vector and introduced into an animal, and if phenotypic changes such as inflammatory symptoms typified by human RA appear, the animal is used. Research on the condition of RA and its remedies. Similarly, by introducing the PADI4 protein or the mutant PADI4 protein of the present invention directly into an animal, a model animal exhibiting an RA disease state can be produced. Hereinafter, the present invention will be described more specifically with reference to Examples, but the present invention is not limited thereto.
〔実施例 1〕 PADI4第 3イントロン中の一塩基多型と R A罹患とのァソシェ ーション解析  [Example 1] Association analysis between single nucleotide polymorphism in PADI4 third intron and RA disease
1 . 解析対象患者の選択 .  1.Select patient for analysis.
日本各地の臨床施設より 846 人の RA患者と 658 人の非患者とからゲノム MA抽出用の血液を採取した。 すべての検体提供者は日本人である。 提供者か らは、 理化学研究遺伝子多型研究セン夕一倫理委員会によるインフォームドコ ンセントに対する同意を得た。 RA患者については、 The revised criteria of the American College of Rheumatology for the classification of rheumatoid arthritis の以下の 7つの基準のうち少なくとも 3項目に該当する者を R A患 者群として選択した。 Genomes from 846 RA patients and 658 non-patients from clinical facilities around Japan Blood for MA extraction was collected. All sample providers are Japanese. The donors gave their consent for informed consent by the Senyuichi Ethics Committee for Genetic Polymorphism Research at RIKEN. For RA patients, those who met at least three of the following seven criteria of the revised criteria of the American College of Rheumatology for the classification of rheumatoid arthritis were selected as RA patients.
(1) 朝のこわばり  (1) Morning stiffness
(2) 3領域以上の関節炎  (2) Arthritis in three or more areas
(3) 手関節炎  (3) Wrist arthritis
(4) 対称性関節炎  (4) symmetric arthritis
(5) リウマトイド結節  (5) Rheumatoid nodules
(6) リウマトイド因子  (6) Rheumatoid factor
(7) X線所見  (7) X-ray findings
非患者群については、 日本人一般集団から患者とは独立に選抜した。 八の 診断の有無以外では R A患者群、 非患者群の間に差がないようにサンプリング した。  The non-patient group was selected from the general Japanese population independently of patients. Sampling was performed so that there was no difference between the RA patient group and the non-patient group except for the presence or absence of the eight diagnosis.
2. ゲノム DNAの調製 2. Preparation of genomic DNA
R A患者および非患者の末梢血から分離した白血球を出発材料とし、 ゲノム DNAを抽出した。 ゲノム DNAの抽出にあたっては、 EDTA'2Na入り採血菅 (べノ ジェクト) に採血し、 QIAa即 DNA Blood kit (QIAGEN)を使用した。 得られた各 試料の 260 nmの吸光度を測定し、各試料の DN A濃度を算出した。その後、 調製した DNAは、 lmMEDTA'2Naを含む lOmM Tris-塩酸緩衝液 (pH8.0)に溶解し、 使用するまで、 -80でで保存した。  Genomic DNA was extracted from leukocytes separated from peripheral blood of RA patients and non-patients as a starting material. For extraction of genomic DNA, blood was collected in a blood collection tube (Benoject) containing EDTA'2Na, and the QIAa DNA Blood Kit (QIAGEN) was used. The absorbance at 260 nm of each sample was measured, and the DNA concentration of each sample was calculated. Thereafter, the prepared DNA was dissolved in lOmM Tris-HCl buffer (pH 8.0) containing lmMEDTA'2Na, and stored at -80 until use.
3. 第 3イントロン中の一塩基多型の検出 3. Detection of single nucleotide polymorphism in the third intron
以下に示す 3種のプローブを用いて、 PADI4の第 3イントロンの 2136 (— 15) 番目の位置 (配列番号 1の 6177番目) における塩基がシトシンであ るかチミンであるかの検出を行った。  Using the three types of probes shown below, we detected whether the base at position 2136 (-15) of the third intron of PADI4 (position 6177 in SEQ ID NO: 1) was cytosine or thymine. .
ィンべ一ダープローブ: 5' -CTGCGCACAGGGAGATTTCTGAAATCCCATCAT-3' (配列番号 2 ) シトシン検出用 F AMプロ一ブ: Invader probe: 5'-CTGCGCACAGGGAGATTTCTGAAATCCCATCAT-3 '(SEQ ID NO: 2) FAM probe for cytosine detection:
5' -ATGACGTGGCAGACGGTAAGAGGAGAGGTTGGY-3' (配列番号 3 )  5'-ATGACGTGGCAGACGGTAAGAGGAGAGGTTGGY-3 '(SEQ ID NO: 3)
チミン検出用 V I Cプローブ: VIC probe for thymine detection:
5' -CGCGCCGAGGAGTAAGAGGAGAGGTTGGY-3' (配列番号 4 ) 5'-CGCGCCGAGGAGTAAGAGGAGAGGTTGGY-3 '(SEQ ID NO: 4)
各プローブは Thi rd Wave Technologies, Inc.に委託して設計 ·合成後、 試 薬として混合されたものを使用した。 インベーダーアツセィは、 96 ゥエル(日 本ジエネティックス)か 384ゥエルプレ一ト(ABI)に、 PCR反応液の 4倍希釈液 2 1 および上記の 3 種のプローブの入ったインベーダー反応液 (Appl ied Biosystems社) を 3 1加え、 95°Cにて 5分間インキュベートした後、 63°Cに て 20分から 80分、シグナルが分離するまで反応させた。 96ゥエルの場合は ABI PRISM 7700 Seauence Detector Sys tem (Appl ied Biosystems社)、 384ゥエル の場合は ABI PRISM 7900HT Sequence Detector Sys tem (Appl ied Biosystems 社) を使ってシグナルを検出した。  Each probe was commissioned to Third Wave Technologies, Inc., designed and synthesized, and used as a mixed reagent. Invader Atsushi is prepared by adding an invader reaction solution (Applied Biosystems, Inc.) containing 96-well (Nihon Genetics) or 384-well plates (ABI) containing a 4-fold dilution of the PCR reaction solution 21 and the above three probes. ) Was added, and the mixture was incubated at 95 ° C for 5 minutes, and then reacted at 63 ° C for 20 to 80 minutes until a signal was separated. The signal was detected using the ABI PRISM 7700 Seauence Detector System (Applied Biosystems) for 96-well, and the ABI PRISM 7900HT Sequence Detector System (Applied Biosystems) for 384-well.
4. 統計解析 4. Statistical analysis
ケース群 (R A患者群) 846人とコントロール群 (非患者群) 658人を対象と し、第 3イントロン 2 1 3 6番目の塩基の 1塩基多型についてケース'コント口 ール関連検定を行った。 検定は 2群間のアレル頻度分布比較 ·優性遺伝形式モ デルによるジエノタイプ分布比較 ·劣性遺伝形式モデルによるジエノタイプ頻 度比較にて行った。検定方法は 2X2分割表 χ 2検定を用いた。解析結果を表 1に 示す。 A case-control-related test was conducted for 846 case patients (RA patients) and 658 controls (non-patients) for the single nucleotide polymorphism at the 3rd intron 2 13 6 Was. The test was performed by comparing the allele frequency distribution between the two groups, comparing the genotype distribution using the dominant inheritance model, and comparing the dienotype frequency using the recessive inheritance model. As a test method, a 2 × 2 contingency table χ2 test was used. Table 1 shows the analysis results.
〔表 1〕 〔table 1〕
解析結果 (PADI4 第 3イントロン 2136番塩基の一塩基多型) Analysis results (PADI4 3rd intron Single nucleotide polymorphism at base No. 2136)
Figure imgf000045_0001
Figure imgf000045_0001
Odds Ratio TT/CC 1.97(95% CI 1.44-2.69)  Odds Ratio TT / CC 1.97 (95% CI 1.44-2.69)
TT/TC 1.38(95% CI 1.03-1.86) TT / TC 1.38 (95% CI 1.03-1.86)
TC/CC 1.43(95% CI 1.13-1 ,80) TC / CC 1.43 (95% CI 1.13-1,80)
Figure imgf000045_0002
以上のとおり、 PADI4遺伝子の第 3イントロンの 2136番目 (配列番号 1 の 6177番目) の位置の遺伝子多型と慢性関節リウマチ罹患との間に統計的 に有意な相関が認められた。
Figure imgf000045_0002
As described above, a statistically significant correlation was found between the polymorphism at position 2136 of the third intron of the PADI4 gene (position 6177 in SEQ ID NO: 1) and the incidence of rheumatoid arthritis.
〔実施例 2〕 PADI4遺伝子中の新規ェクソンー塩基多型の検出 [Example 2] Detection of novel exon-nucleotide polymorphism in PADI4 gene
1. 検出方法  1. Detection method
GenBank のァクセシヨン番号 : NTJ30584.2 に示されるゲノム配列の第 415, 085塩基から第 470, 600塩基に存在する、 PADI4遺伝子上の新規遺伝子多型 の検出を以下の方法により試みた。  GenBank accession number: An attempt was made to detect a novel gene polymorphism on the PADI4 gene, which is present at nucleotides 415,085 to 470,600 in the genome sequence shown in NTJ30584.2 by the following method.
まず、 該遺伝子の全ェクソン、 およびェクソン近傍イントロン、 5'- flankin g領域を約 500 塩基対ごとに分割して増幅領域と設定し、 PCR 増幅した。 PCR 用のプライマ一は Primer3 (http: //www-genome, wi. mit. edu/ge匪 e_sof tware /ot er/pr imer3.html)ソフトウェアを用いてデザインした。 PCR条件は丽 SOの 濃度と、 ァニール温度を変えることで最適化した。  First, the entire exon, the intron near the exon, and the 5'-flanking region of the gene were divided every about 500 base pairs and set as an amplification region, followed by PCR amplification. Primers for PCR were designed using Primer3 (http: // www-genome, wi. Mit. Edu / ge band e_software / oter / primer3.html) software. PCR conditions were optimized by changing the concentration of SO and the annealing temperature.
PCRは 96ゥエル PCRプレートで行い、 各ゥエルに 4 ng/^1ゲノム DNAを 1 a 1ずつ使用した。 反応は 26.5mM (NH4) 2S04, 7.2mM Tris-HCl (pH8.8), 3.2 mM Mg2+, 1. 5mM dNTPs, 1. 6mM b-Mercapto ethanol, 10% DMSO (DMSOを用いない場 合には、 ¾0にて代用), I M pr imers , 2. OU DNA polymerase (ExTaa (TaKaRa) ) ,PCR was performed in 96-well PCR plates, and 1 ng of 4 ng / ^ 1 genomic DNA was used for each well. The reaction 26.5mM (NH 4) 2 S0 4 , 7.2mM Tris-HCl (pH8.8), 3.2 mM Mg 2+ , 1.5 mM dNTPs, 1.6 mM b-Mercapto ethanol, 10% DMSO (If DMSO is not used, substitute with に て 0), IM primers, 2.OU DNA polymerase (ExTaa (TaKaRa) ),
0. 04 xM Taa Start Ant ibody (Clontech)の溶液条件で行った。 反応時間および 反応温度の条件として、 94°Cにて 2分間変性させた後、 94T: 15 秒、 反応ごと に最適化したァニール温度で 45秒, 72でにて 1 あるいは 3 分の伸張反応を 37 サイクル、 ABI9700 thermal cycler (Appl ied Biosystems社)を用いて行った。 増幅された DNA配列は、 BigDye™ Terminator RR Mix (Appl ied Biosystems社) を使って反応を行い、 ABI 3700 seciuencerを用いて塩基配列を決定した。 2 . 結果 The experiment was performed under the solution conditions of 0.04 xM Taa Start Ant ibody (Clontech). After denaturation at 94 ° C for 2 minutes, the extension reaction was performed at 94T for 15 seconds, at an annealing temperature optimized for each reaction for 45 seconds, and at 72 for 1 or 3 minutes. 37 cycles were performed using an ABI9700 thermal cycler (Applied Biosystems). The amplified DNA sequence was reacted using BigDye ™ Terminator RR Mix (Applied Biosystems) and the nucleotide sequence was determined using ABI 3700 seciuencer. 2. Result
各々の領域につき 48人分の塩基配列を比較し、多型性の認められた塩基を 1 塩基多型とみなした。 この結果、 アミノ酸の置換を伴う以下の 3種の新規ェク ソン一塩基多型とアミノ酸の置換を伴わない 1種の新規ェクソン多型が見出さ れた。  The nucleotide sequences of 48 regions were compared for each region, and the nucleotides with polymorphism were regarded as single nucleotide polymorphisms. As a result, the following three new exon single nucleotide polymorphisms with amino acid substitution and one new exon polymorphism without amino acid substitution were found.
1 ) 第 5 5番アミノ酸の置換 (Ser/Gly) を伴う多型:  1) Polymorphism with substitution of amino acid 5 (Ser / Gly):
第 2ェクソンの 7 1番目 (配列番号 1の 1 0 7 1番目) の塩基がアデニンまた はグァニンである一塩基多型 Single nucleotide polymorphism in which the 71st base of the second exon (1071st in SEQ ID NO: 1) is adenine or guanine
2 ) 第 8 2番アミノ酸の置換 (Al a/Val) を伴う多型:  2) Polymorphism with substitution of amino acid 8 (Al a / Val):
第 2ェクソンの 1 5 3番目 (配列番号 1の 1 1 5 3番目) の塩基がシトシンま たはチミンである一塩基多型 Single nucleotide polymorphism in which the 153rd base of the second exon (115rd base in SEQ ID NO: 1) is cytosine or thymine
3 ) 第 1 1 2番アミノ酸の置換 (Al a/Gly) を伴う多型:  3) Polymorphism involving substitution of amino acids 1 and 2 (Al a / Gly):
第 3ェクソンの 6 2番目 (配列番号 1の 4 0 3 6番目) の塩基がシトシンまた はグァニンである一塩基多型 Single nucleotide polymorphism in which the 3rd exon's 6 2nd base (SEQ ID NO: 1 at 4036) is cytosine or guanine
4 ) 第 1 1 7番アミノ酸 (Leu) における、 アミノ酸の置換を伴わない多型: 第 4ェクソンの 9番目 (配列番号 1の 6 2 0 0番目) の塩基がシトシンまたは チミンである一塩基多型  4) Polymorphism without amino acid substitution in the 117th amino acid (Leu): Single nucleotide polymorphism in which the 9th base of the fourth exon (620000th in SEQ ID NO: 1) is cytosine or thymine Type
〔実施例 3〕 PADI4遺伝子上の各一塩基多型の連鎖解析 [Example 3] Linkage analysis of each single nucleotide polymorphism on PADI4 gene
1 . 解析方法  1. Analysis method
R Aと強い関連が認められた PADI4第 3イントロン 2 1 3 6番目 (一 1 5番 目) のヌクレオチドの一塩基多型 (以下、 「PADI4 intron 3 - 15 T/C」 と示す。) と PADI4 上の 4つの新規一塩基多型との Pairwise Linkage Disequi l ibr ium Index (D')を EMアルゴリズムを用いて算出した。 2 . 結果 PADI4 3rd intron 2 1 3 6th (1-15) strongly associated with RA Pairwise Linkage Disequilibrium Index (D ') between the nucleotide single nucleotide polymorphism (PADI4 intron 3-15 T / C) and four new single nucleotide polymorphisms on PADI4 Was calculated using the EM algorithm. 2. Result
その結果、 4つの一塩基多型は相互にほぼ完全連鎖(98. 9〜100%) の関係に あり、 かつ PADI4 intron 3 -15 T/Cともほぼ完全連鎖の関係にあることが、 EM アルゴリズムによるハプロタイプ解析によって確認された。 一方、 PADI4近傍 の PADI2、 PADI 1遺伝子内の公知の一塩基多型と PADI4 intron 3 -15 T/Cにつ いては、 ほとんど連鎖平衡に達していた。  As a result, the EM algorithm shows that the four single nucleotide polymorphisms are almost completely linked to each other (98.9 to 100%), and are also almost completely linked to PADI4 intron 3 -15 T / C. Haplotype analysis. On the other hand, the known single nucleotide polymorphisms in the PADI2 and PADI1 genes near PADI4 and the PADI4 intron 3-15 T / C almost reached linkage equilibrium.
つまり、 日本人集団では、 R A患者群と非患者群の両群において、 55 (Ser) -82 (Al a) -112 (Al a) タイプと 55 (Gly) - 82 (Val) - 112 (Gly) タイプの 2ぺ プチドタイプのみが存在し、 このうち後者のタイプと R Aとの間に、 PADI4 intron 3 -15 T/C と同様のアソシエーションがあることが確認された。 以下、 前者を 「PADI4 V18」 (あるいは単に 「V18」)、 後者を 「PADI4 V9」 (あるいは単 に 「V9」) と記載する。 V18の cDNA配列は配列表の配列番号 1 6に、 アミノ酸 配列は同配列番号 1 7に記載した。 また、 V9の cDNA配列は配列表の配列番号 2 0に、 アミノ酸配列は同配列番号 2 1に記載した。  In other words, in the Japanese population, 55 (Ser) -82 (Al a) -112 (Al a) type and 55 (Gly)-82 (Val)-112 (Gly It was confirmed that only the 2 type peptide exists, and that the latter type and RA have the same association as PADI4 intron 3 -15 T / C. Hereinafter, the former is referred to as “PADI4 V18” (or simply “V18”), and the latter is referred to as “PADI4 V9” (or simply “V9”). The cDNA sequence of V18 is shown in SEQ ID NO: 16 and the amino acid sequence is shown in SEQ ID NO: 17 in the sequence listing. The cDNA sequence of V9 is shown in SEQ ID NO: 20 in the sequence listing, and the amino acid sequence is shown in SEQ ID NO: 21.
以上より、 PADI4遺伝子のジエノタイプと R A発症との間には関連があり、 その関連は PADI4遺伝子に存在する 2つのハプロタイプに由来するものと考え られた。そして、 R Aの発症に関連するハプロタイプは V9であり、 R Aの発症 と関連がないハプロタイプは V18であると考えられた。  From the above, it was concluded that there was a relationship between the dienotype of the PADI4 gene and RA onset, and that the relationship was derived from two haplotypes present in the PADI4 gene. The haplotype associated with the onset of RA was V9, and the haplotype not associated with the onset of RA was V18.
〔実施例 4〕 ヒト PADI4 mR NAの安定性比較 [Example 4] Comparison of stability of human PADI4 mRNA
1 . 分解度の測定 1. Degradation measurement
2つの PAD 14ハプロタイプをコードする遺伝子は以下の方法で調製した。 まず、 Bone marrow total RNA (Clontech 社) を用いて f irs t strand cDNA synthes is ki t (Amersham Pharmacia 社) により合成した cDNA を DONR201 (Invi trogen社) にクローニングした。 得られた cDNAを、 さらに T7プロモー 夕一を含む PDEST14 (Invi trogen社) にリクローニングし、 それぞれ pV18 - T7 および PV9-T7 ベクターを作製し、それらについてシークェンシングを行った。 次いで、 pV18- T7 および pV9- T7 ベクターを Clal で消化し、 RiboMax™ Large Scale RNA Product ion System - T7 (Promega千土)を用レて in vitro transcription を行い、 V9、 および V18の mRNAを作製した。 Genes encoding two PAD 14 haplotypes were prepared in the following manner. First, cDNA synthesized by first strand cDNA synthes is kit (Amersham Pharmacia) using bone marrow total RNA (Clontech) was cloned into DONR201 (Invitrogen). The obtained cDNA was further recloned into PDEST14 (Invitrogen) containing the T7 promoter, and pV18-T7 And PV9-T7 vectors were prepared and sequenced. Then, the pV18-T7 and pV9-T7 vectors were digested with Clal, and in vitro transcription was performed using RiboMax ™ Large Scale RNA Product ion System-T7 (Promega Chido) to produce V9 and V18 mRNA. .
Whole cell extractを調製するために、 HL-60細胞を PBSで洗浄し、 抽出バ ッ フ ァー (0.5% NP-40; 20mM HEPES pH8.0; 20%glycerol (v/v); 400mM NaClcontaining 0.5mM DTT; 0.2mM EDTA; 1% proteinase inhibitor cocktail (Nacal a i) )に懸濁した。氷上で 30分ィンキュベーシヨンした後、 4°Cで遠心し、 上清を 80°Cで保存した。  To prepare Whole cell extract, wash HL-60 cells with PBS and extract buffer (0.5% NP-40; 20 mM HEPES pH 8.0; 20% glycerol (v / v); 400 mM NaCl containing 0.5 mM DTT; 0.2 mM EDTA; 1% proteinase inhibitor cocktail (Nacal ai)). After incubating on ice for 30 minutes, the mixture was centrifuged at 4 ° C, and the supernatant was stored at 80 ° C.
V9、および V18の各 mRNA5 gZreactionに 1000倍希釈した前述の Whole cell extract 3 xlを混ぜ、室温でィンキュベートした。 0, 5, 10分反応後、 formamide dyeを加えて反応を止め、 68でで 10分間加熱し、 氷冷した。 転写産物はノザン プロットハイブリダイゼ一シヨンにより検出した。スキャニングは DocuCentre color 500cp (Fuj i_Xerox社)を用いて行い、シグナル強度は obePhotoshop 6.0 (Adobe社)により測定した。  The above-mentioned Whole cell extract 3 xl diluted 1000-fold was mixed with 5 g of each V9 and V18 mRNA and incubated at room temperature. After reacting for 0, 5, and 10 minutes, the reaction was stopped by adding formamide dye, heated at 68 for 10 minutes, and cooled on ice. Transcripts were detected by Northern plot hybridization. Scanning was performed using DocuCentre color 500cp (Fuji iXerox), and the signal intensity was measured by obePhotoshop 6.0 (Adobe).
結果を図 1に示す。図 1から明らかなように、 R A病変に関連した V9の mRNA は R A病変に関連しない V18の mRNAに比べて有意に分解し難い傾向にあること がわかった。 (*T検定より 5分後で p=0.048, 10分後で p=0.013)。 〔実施例 5〕 ヒト PADI4タンパクの調製  The results are shown in Figure 1. As is evident from FIG. 1, it was found that V9 mRNA associated with RA lesions was significantly less likely to be degraded than V18 mRNA not associated with RA lesions. (P = 0.048 5 minutes after * T test, p = 0.013 after 10 minutes). [Example 5] Preparation of human PADI4 protein
1. 大腸菌による H i S- 1 ag付加 PAD 14タンパク質の発現  1. Expression of PAD 14 protein with Hi S-1 ag by Escherichia coli
His- tagを付加した 2つの PADI4ハプロタイプ断片: RGS6-HisV18 (配列番号 5)、または RGS6- HisV9 (配列番号 6 )が pETlld (Stratagene社)の NcoI/BamHI へ挿入された発現プラスミドを作製した。 このプラスミドで形質転換した大腸 菌 BL2卜 codonPlus (DE3)- RIL cell (Strategene 社)をアンピシリン 50 g/ml (Sigma社) とクロラムフエ二コール 50/ g/ml (Sigma社) を添加した LB培地 (Invitrogen社) に接種し、 37°Cで一晩培養した。 10mLの培養液を 1Lの 2%グ ルコース (和光純薬)、 アンピシリン 50 ig/ml (Sigma社) を添加した 2倍濃度 の LB培地 (Invitrogen社) に添加し、 600nm波長の吸光度が 0.9 になるまで 37°Cで培養した。終濃度 0.1 になるようにイソプロピルチオ- 0- D-ガラクト シド (IPTG :和光純薬) を添加し、 20°Cで 4時間培養した。 培養液を 6000 X g で 10分遠心して、菌体を回収した。菌体を 30m 1の 50mM トリス-塩酸 (pH7. 6) (和光純薬)、 lOOmM塩化ナトリゥム(和光純薬)、 2mM ジチォスレイトール (DTT: 和光純薬)、規定量の完全蛋白分解酵素阻害剤カクテル(Roche Diagnos t ics社) を含む溶解用緩衝液へ懸濁して、 4°Cで超音波破砕し、 97000X gで 30分遠心し て上清を回収した。 さらにこの上清を Mi I lex- HV (ミリポア社)フィルターにか けた。 An expression plasmid was prepared in which two PADI4 haplotype fragments to which His-tags were added: RGS6-HisV18 (SEQ ID NO: 5) or RGS6-HisV9 (SEQ ID NO: 6) were inserted into NcoI / BamHI of pETlld (Stratagene). Escherichia coli BL2 codonPlus (DE3) -RIL cell (Strategene) transformed with this plasmid was added to LB medium (ampicillin 50 g / ml (Sigma) and chloramphenicol 50 / g / ml (Sigma)). Invitrogen) and cultured overnight at 37 ° C. 10 mL of the culture solution was added to a double-concentration LB medium (Invitrogen) supplemented with 1 L of 2% glucose (Wako Pure Chemical Industries) and ampicillin 50 ig / ml (Sigma), and the absorbance at 600 nm was reduced to 0.9. The cells were cultured at 37 ° C until complete. Isopropylthio-0-D-galacto to a final concentration of 0.1 Cid (IPTG: Wako Pure Chemical Industries) was added, and the cells were cultured at 20 ° C for 4 hours. The culture was centrifuged at 6000 X g for 10 minutes to collect the cells. Bacterial cells were 30m1 of 50mM Tris-HCl (pH 7.6) (Wako Pure Chemical), lOOmM sodium chloride (Wako Pure Chemical), 2mM dithiothreitol (DTT: Wako Pure Chemical), specified amount of complete protease The suspension was suspended in a lysis buffer containing an inhibitor cocktail (Roche Diagnostics), sonicated at 4 ° C., and centrifuged at 97,000 × g for 30 minutes to collect the supernatant. The supernatant was applied to a Mi Ilex-HV (Millipore) filter.
2 . His- tag付加 PADI4 タンパク質の精製 2. Purification of His-tagged PADI4 protein
前項で調製した上清を予めバッファー 50mM トリス-塩酸(pH7. 6) (和光 純薬)、 lmM エチレンヂアミンテトラ酢酸 (EDTA :同仁化学) 1で平衡化した Sephadex- G25 (アマシャム ·バイオサイエンス社) に供し、 0. 1Mの塩酸ナトリ ゥムが入ったバッファ一 Aで分画した。 活性が認められた画分をバッファー A で予め平衡化した DEAE-Sepharose (アマシャム'バイオサイエンス社)に供し、 0. 1Mの塩酸ナトリウムが入ったバッファー Aで洗浄した後、 バッファー Aへ添 加した塩酸ナトリウム濃度を 0. 1Mから 0. 2Mへ徐々に変化させて溶出し、 分画 した。 活性が認められた画分に三倍量のバッファ一 B { 50mM トリス-塩酸 (pH7. 6) (和光純薬)、 0. 1M塩化ナトリウム(和光純薬)、 0. 1% トライトン X-100} を加え、 Ni- NTAスーパ一フローカラム (QIAGEN社) に供した。 カラム を 20mMのィミダゾールが入ったバッファ一 Bで洗浄し、 200mMのィミダゾール が入ったバッファ一 Bで溶出した。活性が認められた画分をバッファー C { 10mM 卜リス-塩酸 (pH7. 6) (和光純薬)、 0. 15M塩化ナトリウム (和光純薬)、 2mM DTT (和 光純薬)、 lmM EDTA (同仁化学) } で透析し、 濃縮後、 終濃度 10%になるように グリセロール (Sigma社) を添加して、 _80°Cに保存した。  Sephadex-G25 (Amersham Biosciences, Inc.) previously equilibrated the supernatant prepared in the previous section with buffer 50 mM Tris-HCl (pH 7.6) (Wako Pure Chemical Industries) and lmM ethylenediaminetetraacetic acid (EDTA: Dojindo Chemical) 1 ) And fractionated with buffer 1A containing 0.1 M sodium chloride. The fraction in which the activity was observed was applied to DEAE-Sepharose (Amersham Bioscience) pre-equilibrated with buffer A, washed with buffer A containing 0.1 M sodium chloride, and then added to buffer A. Elution was performed by gradually changing the sodium chloride concentration from 0.1 M to 0.2 M, and fractionation was performed. A three-fold amount of buffer B (50 mM Tris-HCl (pH 7.6) (Wako Pure Chemical Industries), 0.1 M sodium chloride (Wako Pure Chemical Industries), 0.1% Triton X-100 } And added to a Ni-NTA super-flow column (QIAGEN). The column was washed with buffer B containing 20 mM imidazole and eluted with buffer B containing 200 mM imidazole. The fractions showing the activity were analyzed using buffer C {10 mM Tris-HCl (pH 7.6) (Wako Pure Chemical), 0.15 M sodium chloride (Wako Pure Chemical), 2 mM DTT (Wako Pure Chemical), lmM EDTA (Dojindo) After concentrating, glycerol (Sigma) was added to a final concentration of 10% and stored at _80 ° C.
〔実施例 6〕 ヒト PADI4夕ンパクの活性測定 [Example 6] Activity measurement of human PADI4 protein
1 . PADI4活性の測定方法 (蛍光法)  1. Method for measuring PADI4 activity (fluorescence method)
酵素反応用の緩衝液として、 lOOmM Tri s/HCl (pH7. 8)、 難 CaCl2、 5mM DTT、 lOmM BAEE (Sigma) または N- a - Benzoy卜卜 arginine (東京化成) を含む溶液 を調製した。この緩衝液40 1と、実施例5で調整した¥18酵素原液(113 1111 ) の希釈列 10/i 1を PCR反応プレート (Applied Biosystems社) 内にて混合し、 37°Cで 2時間反応させた。 反応後、 lO/ilの 200mMEDTAを添加して反応を停止 した。 このうち 分を deep well plate (日本ジエネテイクス社) に移し た。 なお同時に、 検量線作成のため、 0- 50nMの硫酸アンモニゥム溶液も別のゥ エルへ添加した。 さらにこれらのゥエルへ、 50mMホウ酸を lml、 および 50mM o-phthalaldehydeと 50mM DTTの等量混合溶液を 50 xlずつ添加し、 室温で 1 時間反応させた。反応溶液は 分を白色プレート (コースター ·コ一ニン グ社) に移し、 Spectra max GEMINI (Molecular Devices 社) を用いて励起波 長 413nm、 蛍光波長 476nmの各波長を測定した。 酵素を含まないゥエルの測定 値をバックグラウンドとして差し引き、 検量線作成用硫酸アンモニゥムを添加 したゥエルでの測定値から検量線を描くことにより、 反応によって生成したァ ンモニゥムイオンの濃度を求めた (図 2A;)。 As a buffer solution for enzyme reaction, lOOmM Tri s / HCl (pH7 8.), Flame CaCl 2, 5mM DTT, lOmM BAEE (Sigma) or N-a - A solution containing Benzoy Bokuboku arginine (Tokyo Kasei) was prepared . This buffer solution 401 and the undiluted enzyme solution of ¥ 18 prepared in Example 5 (113 1111) Was diluted in a PCR reaction plate (Applied Biosystems) and reacted at 37 ° C for 2 hours. After the reaction, the reaction was stopped by adding lO / il of 200 mM EDTA. A portion of this was transferred to a deep well plate (Nihon Genetics). At the same time, a 0-50 nM ammonium sulfate solution was also added to another well to prepare a calibration curve. Further, 1 ml of 50 mM boric acid and 50 xl of an equal volume mixed solution of 50 mM o-phthalaldehyde and 50 mM DTT were added to these wells, and reacted at room temperature for 1 hour. The reaction solution was transferred to a white plate (Coaster Coating Co., Ltd.), and the excitation wavelength of 413 nm and the fluorescence wavelength of 476 nm were measured using Spectra max GEMINI (Molecular Devices). The measured value of the well without enzyme was subtracted as the background, and the concentration of ammonium ion generated by the reaction was determined by drawing a calibration curve from the measured value of the well with the addition of ammonium sulfate for preparing the calibration curve (Fig. 2A). ;).
2. PADI4活性の測定方法 (吸光法) 2. PADI4 activity measurement method (absorption method)
酵素反応用の緩衝液として、 lOOmMTris/HCl (pH7.6)、 10mMCaCl2、 5mMDTT、 lOmM BAEE (Sigma) を含む溶液を調製した。 この緩衝液 40 1 と、
Figure imgf000050_0001
の実施例 5で調整した V18酵素原液(113/ g/ml)の希釈列 10 lを PCR反応プ レート (Applied Biosystems社) 内にて混合し、 50°Cもしくは 371:で 2時間反 )¾ヽさせた。反応後、 A液(SOmMDiacetyl monoxime (3— hydroxy imino 2— butane)、 2mM t iosemicarbazide) を含む) と B液 (3M H3P04、 6M ¾S04、 2mM NH4Fe (S04) 2 を含む) を 1: 3の比率で混合した溶液を 150 1ずつ添加した。 なお同時に、 検量線作成のため、 0— 400 Mの L-シトルリン溶液も別のゥェルへ添加し、 同 様に A液と B液の混合溶液を添加した。 混合溶液は、 サーマルサイクラ一 (GeneAmp™ PCR System 9700、 Applied Biosystems社) を使用して 95で15分 の後室温で 10分反応させた。反応後の溶液は 150 X 1分を 96ゥエルプレート(コ 一二ング社) に移し、 Spectra max 250 (Molecular Devices社) を用いて 540nm の吸光度を測定した。 酵素を含まないゥエルの測定値をバックグラウンドとし て差し引き、 検量線作成用シトルリンを添加したゥエルでの測定値から検量線 を描くことにより、反応によって生成したシトルリンの濃度を求めた(図 2 B)。 3 . ヒト PADI4ハプロタイプ V9および V18の酵素活性
A solution containing 100 mM Tris / HCl (pH 7.6), 10 mM CaCl 2 , 5 mM DTT, and 10 mM BAEE (Sigma) was prepared as a buffer for the enzyme reaction. This buffer 40 1 and
Figure imgf000050_0001
10 l of the diluted series of V18 enzyme stock solution (113 / g / ml) prepared in Example 5 was mixed in a PCR reaction plate (Applied Biosystems), and the mixture was reacted at 50 ° C or 371: for 2 hours.ヽAfter the reaction, including solution A (SOmMDiacetyl monoxime (3- hydroxy imino 2- butane), 2mM t iosemicarbazide) containing) and B solution (3M H 3 P0 4, 6M ¾S0 4, 2mM NH 4 Fe (S0 4) 2 ) In a 1: 3 ratio were added in 150 1 increments. At the same time, to prepare a calibration curve, a 0-400 M L-citrulline solution was also added to another well, and a mixed solution of solution A and solution B was similarly added. The mixed solution was reacted using a thermal cycler (GeneAmp ™ PCR System 9700, Applied Biosystems) at 95 for 15 minutes and then at room temperature for 10 minutes. The solution after the reaction was transferred at 150 × 1 minute to a 96-well plate (Corning), and the absorbance at 540 nm was measured using Spectra max 250 (Molecular Devices). The concentration of citrulline produced by the reaction was determined by subtracting the measured value of the well containing no enzyme as a background and drawing a calibration curve from the measured value of the well to which citrulline for preparing the calibration curve was added (Fig. 2B). ). 3. Enzyme activity of human PADI4 haplotype V9 and V18
吸光度法を用いてヒト PAD 14ハプロタイプ V9および V 18の酵素活性を測定し、 比較した (図 3 )。 図 3から明らかなように、 V9は V18に比べて有意に酵素活 性が高いことが確認された (Pく 0. 01)。 以上より、 R A病変に関連した PADI4夕 イブ (V9) は正常 PADI4タイプ (V18) よりもその酵素活性が高く、 こうした酵 素活性の違いが R A発症と何らかの関連を有することが推測された。  The enzymatic activities of human PAD 14 haplotypes V9 and V18 were measured using the absorbance method and compared (Figure 3). As is clear from FIG. 3, it was confirmed that V9 had significantly higher enzyme activity than V18 (P 0.01). Based on the above, PADI4 Eve (V9) associated with RA lesions had higher enzyme activity than normal PADI4 type (V18), suggesting that such a difference in enzyme activity might have some association with RA onset.
〔実施例 7〕 R A患者の PADI4ジエノタイプと抗シトルリン化フィラグリン 抗体価との関連 [Example 7] Association between PADI4 dienotype and anti-citrullinated filaggrin antibody titer in RA patients
1 . 抗シトルリン化ペプチド抗体価の測定方法  1. Anti-citrullinated peptide antibody titer measurement method
R A患者血清の抗シトルリン化ぺプチド抗体価を MESACUP ACFテストは、 医 学生物学研究所(名古屋)委託して作製したキットを用いて行った。 MESACUP ACF テス卜とは、リコンビナント ヒトフイラダリンぺプチドを PADによりシトルリ ン化した、 シトルリン化フィラグリンを抗原として、 ELISA法により抗体価を 測定する方法である。  The MESACUP ACF test for the anti-citrullinated peptide antibody titer of RA patient sera was performed using a kit commissioned by the Institute of Medical Biology (Nagoya). The MESACUP ACF test is a method for measuring antibody titer by ELISA using citrullinated filaggrin, which is obtained by converting recombinant human filadrin peptide into citrulline by PAD, as an antigen.
まず、患者末梢血より血清を調製し、 10 z lを反応用緩衝液 lmLに加えて 101 倍希釈した。 抗フィラグリン抗体標準血清 2、 標準血清 1および希釈した検体 を 150 /x L ずつ一次反応準備用マイクロプレートに実際と同じように添加した。 その後、抗原感作マイクロカップにマルチピぺッ卜を用い 100 Lずつ移した。 室温 (20〜25°C) で 1時間静置反応させた後、 洗浄液で 4回洗浄し、 酵素標識 抗体をマルチピペットで ずつ添加した。 その後、 室温 (20〜25°C) で 1時間静置反応させ、 反応停止液をマルチピペットで ずつ添加した。 各反応後のサンプルを分光吸光度計を用いて O D 4 5 0を測定し、 Index 値 を下式を用いて算出した。  First, serum was prepared from peripheral blood of a patient, and 10 zl was added to 1 mL of a reaction buffer and diluted 101-fold. The anti-filaggrin antibody standard serum 2, the standard serum 1, and the diluted sample were added to the primary reaction preparation microplate at 150 / xL in the same manner as in the actual case. Thereafter, 100 L was transferred to the antigen-sensitized microcup using a multi-pip. After allowing the reaction to stand at room temperature (20 to 25 ° C) for 1 hour, the plate was washed four times with a washing solution, and the enzyme-labeled antibody was added by a multipipette. Thereafter, the reaction was allowed to stand at room temperature (20 to 25 ° C) for 1 hour, and the reaction terminating solution was added by a multipipette. The sample after each reaction was measured for OD450 using a spectrophotometer, and the Index value was calculated using the following equation.
検体の A450—標準血清 1の A450  A450 of sample—A450 of standard serum 1
Index値 = X 100 抗フィラグリン抗体標準血清 2の A450—標準血清 1の A450  Index value = X100 A450 of anti-filaggrin antibody standard serum 2—A450 of standard serum 1
2 . P AD 14ジエノタイプと抗シトルリン化フィラグリン抗体価との関連 2. Association between PAD 14 dienotype and titer of anti-citrullinated filaggrin
PADI4 int ron 3 -15 T/C ジエノタイプの決められた、 R A患者 57人につき、 血清中の抗合成シトルリン化フィラグリン抗体価を測定した。 カットオフ値 10にて、 分割表検定を行ったところ、 下表 2のようになった。 〔表 2〕
Figure imgf000052_0001
Serum anti-synthetic citrullinated filaggrin antibody titers in 57 RA patients with defined PADI4 intron 3 -15 T / C dienotype were measured. A contingency table test with a cutoff value of 10 gave the results shown in Table 2 below. (Table 2)
Figure imgf000052_0001
罹患ホモ接合体 (TT) のジエノタイプと他のジエノタイプとの間の比較を Fisher's 正確検定(One- s ided)にて行ったところ、 p=0. 0284を得た。すなわち、 R A患者内において、 罹患ホモ接合体 (TT) のジエノタイプを有する患者では 抗シトルリン化フイラダリン抗体価が他のジエノタイプを有する患者に比較し て有意に高いことが示された。 この結果 5 oと実施例 4、 6の結果より、 PADI4 ジ エノタイプの違いは、 niRNAの安定性や酵素活性といった PADI4フエノタイプの 変化をもたらし、 シトルリン化タンパクのレベルの亢進につながる可能性が高 いことが推測された。  Comparison between the dienotype of affected homozygotes (TT) and other dienotypes by Fisher's exact test (One-sided) gave p = 0.0284. That is, among RA patients, it was shown that the patients with the affected homozygote (TT) dienotype had significantly higher anti-citrullinated filadarin antibody titers than the patients with other dienotypes. From the results of 5 o and the results of Examples 4 and 6, differences in the PADI4 genotype are likely to lead to changes in the PADI4 phenotype such as niRNA stability and enzyme activity, leading to an increase in citrullinated protein levels. That was speculated.
〔実施例 8〕 R A患者滑膜組織における PADI4発現量の検討 [Example 8] Examination of PADI4 expression level in synovial tissue of RA patient
1 . In s i tu-RT-PCR  1. In situ-RT-PCR
R A患者の滑膜組織から調整した組織片に Pro STAR HF (Stratagenes社) を 添加し、 以下の条件で One step In s i tu-RT-PCR を Hybaid Omni sl ide (Hybaid Ltd. , Middlesex, UK)上にて実施した。  Pro STAR HF (Stratagenes) was added to a piece of tissue prepared from the synovial tissue of RA patient, and one step in situ-RT-PCR was performed under the following conditions using Hybaid Omni slide (Hybaid Ltd., Middlesex, UK). Implemented above.
(1) 42°C 30 分  (1) 42 ° C 30 minutes
(2) 94t: 2 分, 55°C 45秒, 68°C 2分  (2) 94t: 2 minutes, 55 ° C 45 seconds, 68 ° C 2 minutes
(3) 94t 45秒, 55°C 45秒, 68°C 2分、 25サイクル  (3) 94t 45 seconds, 55 ° C 45 seconds, 68 ° C 2 minutes, 25 cycles
PCR反応後のスライドグラスは PBS にて 5分間ずつ 2回洗浄した。  The slide glass after the PCR reaction was washed twice with PBS for 5 minutes each.
hPADI4 V18の cDNA (配列番号 1 6 ) を铸型に、 以下の PADI4増幅用プライマ —を用いて PCRを行い、得られた 335塩基長の増幅産物をジゴキシゲニン (DIG) 標識して、 PADI4検出用プローブとした。  PCR was performed on the hPADI4 V18 cDNA (SEQ ID NO: 16) using the following primers for PADI4 amplification, and the resulting 335-base amplification product was labeled with digoxigenin (DIG) to detect PADI4. A probe was used.
Forward Primer : 5' -GACCAGAAGGTTCAGATTTCATACTAC-3' (配列番号 7 ) Forward Primer: 5 '-GACCAGAAGGTTCAGATTTCATACTAC-3' (SEQ ID NO: 7)
Reverse Pr imer : 5' -ACCAGTGTATGGTTTGTGAAGAAGT-3' (配列番号 8 ) Reverse Primer: 5'-ACCAGTGTATGGTTTGTGAAGAAGT-3 '(SEQ ID NO: 8)
なお、 DIG標識は PCR DIG probe synthes is ki t (Roche Diagnost ics社)を用 いて行った。 逆転写後のサンプルに、 前述のプライマーを最終濃度 0. 34 Mとなるように 加え、スライドグラスを被せて 37°C 1時間プレハイブリダィゼーシヨンを行つ た。次いで、 94°Cで 5分間変性させた DIG標識プローブを加え、 37°Cで 12時間 ハイブリダィゼーシヨンを行った。 反応後のスライドを洗浄し、 digoxygenin detect ion ki t (Roche Diagnost ics社)を用いて PADI4遺伝子の検出を行った。 陰性対照として、 プライマー非添加、 反応酵素非添加の条件で同様に検出を行 つた。 DIG labeling was performed using PCR DIG probe synthes is kit (Roche Diagnostics). To the sample after reverse transcription, the above-mentioned primer was added to a final concentration of 0.34 M, covered with a slide glass, and prehybridized at 37 ° C for 1 hour. Next, a DIG-labeled probe denatured at 94 ° C for 5 minutes was added, and hybridization was performed at 37 ° C for 12 hours. After the reaction, the slide was washed, and the PADI4 gene was detected using digoxygenin detect ion kit (Roche Diagnostics). As a negative control, detection was performed in the same manner with no primer added and no reaction enzyme added.
2 . 結果 2. Result
結果を図 4に示した。 PAD 14 の転写産物は R A患者滑膜組織の表層(l ining) と滑膜表層下 (sub-l ining layer) 付近に局在していた。 また、 シグナルは繊 維芽細胞様細胞や血管周囲の球状核細胞 (round- nuclear cel l) 中で観察され た。 〔実施例 9〕 マウスコラーゲン誘導関節炎 (C I A) における PADI4遺伝子 発現量の検討  The results are shown in FIG. The transcript of PAD 14 was localized in the synovial tissue of RA patients at the surface of the synovial tissue (lining) and near the subsurface of the synovium (sub-lining layer). The signal was also observed in fibroblast-like cells and in round-nucleated cells around blood vessels. [Example 9] Examination of PADI4 gene expression level in mouse collagen-induced arthritis (CIA)
マウスにコラーゲン誘導関節炎(CIA) を誘導し、その滑膜組織および脾臓組 織における、マウス ぺプチジルアルギニン ·ディミナ一ゼ'タイプ IV (mPADI4) 遺伝子の発現量について検討した。 なお、 mPADM は、 ヒト ぺプチジルアルギ ニン ·ディミナ一ゼ'タイプ I V (hPADI4)のマウスォーソログである。 mPADI4 の cDNA配列とアミノ酸配列はそれぞれ配列表の配列番号 1 8および配列番号 Collagen-induced arthritis (CIA) was induced in mice, and the expression level of mouse peptidylarginine diminase 'type IV (mPADI4) gene in synovial tissue and spleen tissue was examined. MPADM is a mouse ortholog of human peptidylarginine diminase 'type IV (hPADI4). The cDNA sequence and amino acid sequence of mPADI4 are shown in SEQ ID NO: 18 and SEQ ID NO:
1 9に記載した。 Described in 19.
1 . マウス C I Aモデルの作製  1. Creation of mouse CIA model
マウス (5週齢の雌、 日本チヤ一ルスリバ一(株) より購入) は、 CIA 群 (N=l l) と健常群 (N=l l) に分けた。 CIA群は訓化後、 タイプ IIコラーゲン Mice (five-week-old females, purchased from Nippon Charlriva Co., Ltd.) were divided into CIA group (N = l l) and healthy group (N = l l). CIA group after training, type II collagen
2mg/ml (コラーゲン技術研修会)および ADJUVANT COMPLETE FREUND (DIFC0)を等 量の割合で混ぜ、超音波によりェマルジヨン化したものを、 1匹あたり 100 1、 一週間の間隔をおいて計 2回 尾根部に皮内投与した。最終的に、 CIA群は発症 が見られた 5匹 7肢を、 健常群は 11匹 22肢を以下の実験に供した。 2 . 滑膜組織および脾臓組織における、 mPADI4遺伝子発現量の測定 2mg / ml (collagen technology workshop) and ADJUVANT COMPLETE FREUND (DIFC0) mixed in equal proportions and emulsified by ultrasound, 100 1 per animal, 2 times at one week intervals twice It was intradermally administered to the part. Finally, the CIA group was subjected to the following experiment with 7 limbs of 5 animals with onset, and the healthy group with 22 limbs of 11 animals. 2. Measurement of mPADI4 gene expression in synovial tissue and spleen tissue
マウスは 2回目の投与後 7週目に断頭放血により致死させ、 滑膜組織および 脾臓組織をハサミおよびピンセットで摘出した。 摘出した組織より、 RNeasy mini ki t または RNeasy midi ki t (QIAGEN社)を用いて、 添付のプロトコ一ル に従い全 RNAを調製した。 調製した全 RNAより、 TaKaRa RNA PCR™ Ki t (AMV) Ver. 2. 1 (TaKaRa) を用いて、 添付プロトコールに従い cDNAを合成した。  The mice were sacrificed 7 weeks after the second administration by decapitation and the synovial tissue and spleen tissue were removed with scissors and tweezers. Total RNA was prepared from the excised tissues using RNeasy mini kit or RNeasy midi kit (QIAGEN) according to the attached protocol. CDNA was synthesized from the prepared total RNA using TaKaRa RNA PCR ™ Kit (AMV) Ver. 2.1 (TaKaRa) according to the attached protocol.
mPADI4遺伝子の発現は、 ABI PRIZM 7700 System (Appl ied Biosystems社) を用いた RT-PCRにより検討した。 用いた mPADI4遺伝子増幅用プライマー、 お よび mPADI4遺伝子検出用プローブは以下のとおりである。  The expression of the mPADI4 gene was examined by RT-PCR using ABI PRIZM 7700 System (Applied Biosystems). The used primers for amplifying the mPADI4 gene and the probe for detecting the mPADI4 gene are as follows.
Forward Primer : 5' -ACGCTGCCTGTGGTCTTTG-3' (配列番号 9 ) Forward Primer: 5'-ACGCTGCCTGTGGTCTTTG-3 '(SEQ ID NO: 9)
Reverse Primer : 5' -CCAGCCCAGTGAGCTCTGA-3' (配列番号 1 0 ) Reverse Primer: 5'-CCAGCCCAGTGAGCTCTGA-3 '(SEQ ID NO: 10)
Probe : 5' -CCTGAAGGATTTCCCTGTCAAGCGAGTT-3' (配列番号 1 1 ) Probe: 5'-CCTGAAGGATTTCCCTGTCAAGCGAGTT-3 '(SEQ ID NO: 11)
なお、 mPADI4 遺伝子発現量は、 内部標準として用いたマウス /3 -glucuronidase (mGUS)遺伝子の発現量で補正した。 MGUS4遺伝子増幅用プライ マー、 および MGUS4遺伝子検出用プローブの配列は以下のとおりである。  The expression level of mPADI4 gene was corrected by the expression level of mouse / 3-glucuronidase (mGUS) gene used as an internal standard. The sequences of the primer for amplifying the MGUS4 gene and the probe for detecting the MGUS4 gene are as follows.
Forward Primer : 5' -TACGGGATTGTGGTCATCGA-3' (配列番号 1 2 ) Forward Primer: 5'-TACGGGATTGTGGTCATCGA-3 '(SEQ ID NO: 12)
Reverse Primer : 5' -CGAACCAGCTCCTCCATCAC-3' (配列番号 1 3 ) Reverse Primer: 5'-CGAACCAGCTCCTCCATCAC-3 '(SEQ ID NO: 13)
Probe: 5' -CAACGAGTCACTTCGGCACCACCTAGAG-3' (配列番号 1 4 ) 3 . 結果 Probe: 5'-CAACGAGTCACTTCGGCACCACCTAGAG-3 '(SEQ ID NO: 14) 3. Results
結果を図 5に示した。 CIA群滑膜組織では健常群滑膜組織に比べ mPADI4の発 現量は 19. 5倍に上昇していた。 また、 CIA群脾臓組織では健常群脾臓組織に比 ベ mPAD の発現量は 3倍に上昇していた。以上より CIA群では、滑膜組織およ び脾臓組織において mPAD 遺伝子発現が著しく上昇することが確認された。  The results are shown in FIG. The expression level of mPADI4 was increased 19.5-fold in the synovial tissue of the CIA group compared with the healthy group. In the spleen tissue of the CIA group, the expression level of mPAD was three times higher than in the spleen tissue of the healthy group. From the above, it was confirmed that mPAD gene expression was significantly increased in synovial tissue and spleen tissue in the CIA group.
〔実施例 1 0〕 抗 PADI4抗体 (抗 PADI4ペプチド抗血清) の調製 [Example 10] Preparation of anti-PADI4 antibody (anti-PADI4 peptide antiserum)
ヒト PADI4由来のぺプチド配列 ( PAKKKSTGSSTWP_Cys:配列番号 1 5 ) を合 成し (オリエンタル酵母北山ラベス, ペプチド合成機:アプライドバイオシス テムズジャパン株式会社, Pioneer, 合成法: Fmocアミノ酸を用いた固相合成)、 KHL (Keyhole l impet heinocyanin, CALBIOCHEM )を MBS (PIERCE)法を用いてコ ンジユゲーションした。 Synthesize peptide sequence (PAKKKSTGSSTWP_Cys: SEQ ID NO: 15) derived from human PADI4 (Oriental Yeast Kitayama Labes, Peptide Synthesizer: Applied Biosystems Japan, Pioneer, Synthesis: Solid phase synthesis using Fmoc amino acid ), KHL (Keyhole l impet heinocyanin, CALBIOCHEM) using MBS (PIERCE) method It was conjugated.
免疫動物はゥサギ (Kbl 'JW, ォス, 週齢:リタイア)を使用し、初回免疫におい て、 FCA (フロイント完全アジュバント, DIFC0)と抗原との等量混合液を背部皮 下に注射した。初回免疫はこの抗原を 600 ; gペプチド/匹 用いて行った。 2回 目以降は FIA (フロイントの不完全アジュバント, DIFC0)と抗原との等量混合液 を 2週間おきに 3回背部皮下に注射した。 2次免疫以降はこの抗原を 300 / g ペプチド/匹 用いて行った。 最終免疫から 2週間後に全採血し、 3000rpm にて 20分間、遠心し、血清を分離回収したのち、 NaN3 (和光純薬) を 0. 05% (w/v) 添 加し、 4°C (冷蔵) にて保存した。 For the immunized animals, a heron (Kbl'JW, Oss, age: retired) was used. In the first immunization, an equal mixture of FCA (Freund's complete adjuvant, DIFC0) and an antigen was injected subcutaneously in the back. The first immunization was performed using this antigen at 600; g peptide / animal. From the second dose onwards, an equal volume mixture of FIA (incomplete Freund's adjuvant, DIFC0) and antigen was injected subcutaneously three times every two weeks into the back. After the second immunization, this antigen was used at 300 / g peptide / animal. Two weeks after the final immunization, the whole blood was collected, centrifuged at 3000 rpm for 20 minutes, and the serum was separated and collected, and NaN 3 (Wako Pure Chemical) was added at 0.05% (w / v) and the temperature was 4 ° C (Refrigerated).
〔実施例 1 1〕 R A患者関節滑膜組織片の免疫染色 [Example 11] Immunostaining of synovial tissue section of RA patient joint
1 . 関節滑膜組織切片の調製  1. Preparation of articular synovial tissue section
人工膝関節置換術に際して得られた切離組織より、 関節滑膜組織片を得た。 得られた滑膜組織片は即座に液体窒素中に漬け、 スライドグラス上薄層切片作 製まで- 80でで保存した。 薄層切片化に際し、 凍結組織片を 10%中和緩衝ホル マリンにて固定し、 70%エタノール、 85%エタノール、 90%エタノール、 100%エタ ノール(100%エタノールの場合に限り 2回)にて各 1時間脱水処理し、 ついで、 50%エタノール 50%パラフィン溶液で 30分間にて 1回、 100%パラフィン溶液で 30分間にて 2回、 処理し、 組織片をパラフィン化した。 その後、 組織片をパラ フィン中に包埋した。パラフィン包埋組織片はライカミクロト一ム RM2165で 4 mの厚さにスライスし、 スライドグラスに固定した。  A synovial tissue piece was obtained from the dissected tissue obtained during the total knee arthroplasty. The obtained synovial tissue piece was immediately immersed in liquid nitrogen and stored at -80 until preparation of a thin section on a slide glass. For thin sectioning, fix the frozen tissue section with 10% neutralized buffered formalin, and add 70% ethanol, 85% ethanol, 90% ethanol, and 100% ethanol (twice for 100% ethanol only). After dehydration for 1 hour each, the tissue was treated once with 50% ethanol and 50% paraffin solution for 30 minutes and twice with 100% paraffin solution for 30 minutes to paraffinize the tissue pieces. Thereafter, the tissue pieces were embedded in paraffin. Paraffin-embedded tissue slices were sliced to a thickness of 4 m with Leica Microtome RM2165 and fixed on slide glasses.
2 . 免疫染色 2. Immunostaining
免疫染色に先立ち、 前項で調製したパラフィン包埋切片をキシレンに 3分間 ずつ 2回つけ、脱パラフィン化した後、 100%エタノールに 1分間ずつ 2回、 95% エタノ一ルに 1分間ずつ 2回、 さらに PBS (0. 13M NaCL 8. 6mM K2HP04, 1. 5mM KH2P04)に 3分間ずつ 2回つけ、 再水和した。 30%¾02メ夕ノール溶液で 3分間 処理し、 内在性ピオチンを除去した。 PBS にて 5分間ずつ 2回、 洗浄し、 1000 倍希釈したそれぞれの抗体:抗シトルリン化タンパク質抗体 (Biogenesis社) と実施例 1 0で調製した抗 PADI4抗体とを加えて 4でにて 12時間反応させた。 反応後、 PBSにて 5分間ずつ 2回洗浄し、 SimpleStain MAX-P0 (Nichirei)を 1 スライドにっき ずつ加え、 25 :にて 30分間反応させた。 その後、 PBS にて 5 分間ずつ 2回洗浄を行った。 基質として、 SimpleStain AEC (Nichirei) を 1スライドにっき 100 1ずつ加え、 シグナルが検出されるまで 5分から 20 分反応させた。 反応は超純水につけて停止させた。 . 糸口果 Prior to immunostaining, attach the paraffin-embedded section prepared in the previous section to xylene twice for 3 minutes each, and then deparaffinize.Then, 100% ethanol twice for 1 minute, and 95% ethanol twice for 1 minute Then, the cells were immersed twice in PBS (0.13 M NaCL 8.6 mM K2HP04, 1.5 mM KH2P04) for 3 minutes each and rehydrated. 30% ¾0 2 main evening treated with Nord solution 3 minutes to remove endogenous Piochin. After washing twice with PBS for 5 minutes each and diluting 1000-fold, add each antibody: anti-citrullinated protein antibody (Biogenesis) and the anti-PADI4 antibody prepared in Example 10 and 4 for 12 hours. Reacted. After the reaction, the plate was washed twice with PBS for 5 minutes each, and SimpleStain MAX-P0 (Nichirei) was added to each slide, followed by reaction at 25: for 30 minutes. Thereafter, the cells were washed twice with PBS for 5 minutes each. As a substrate, SimpleStain AEC (Nichirei) was added 100 per slide, and allowed to react for 5 to 20 minutes until a signal was detected. The reaction was stopped by immersing in ultrapure water. .
結果を図 6および図 7に示す。 RA患者の滑膜組織では PADI4 を特異的に認 識する抗体による反応 (Aの枠で囲った箇所の赤い部分) が認められたが、 関 節リウマチの陰性対照である変形性関節症 (OA) 患者の滑膜組織には PADI4 を認識する抗体による反応は認められなかった(図 6 B)。 また、 RA患者の滑 膜組織では、 PADI4 とほぼ同じような部分にシトルリン化夕ンパク質が認めら れた (図 7) 〔実施例 1 2〕 ヒト PADI4 mRNAの安定性比較  The results are shown in FIGS. In the synovial tissue of RA patients, a reaction with an antibody that specifically recognizes PADI4 was observed (red area in box A), but osteoarthritis (OA), a negative control for rheumatoid arthritis, was observed. ) No reaction was observed in the synovial tissue of the patient due to antibodies recognizing PADI4 (Fig. 6B). In the synovial tissue of RA patients, citrullinated protein was observed in almost the same area as PADI4 (Fig. 7) [Example 12] Comparison of human PADI4 mRNA stability
1. mRNAの調製  1. Preparation of mRNA
2つの PADI4ハプロタイプの mRNAは、 以下の方法で調製した。  The mRNAs of the two PADI4 haplotypes were prepared by the following method.
V9もしくは V18をコードする遺伝子を CMVプロモーターの下流につないだブラ スミドを、 リポフエクトァミン 2000 (Invitrogen社) を用い、 添付のプロトコ —ルに従ってヒト K562細胞への遺伝子導入を行った。 37°Cで 4時間培養した後、 PMA(Sigma社)を最終濃度 50ng/mlとなるように添加した。 40時間培養した後、 ァクチノマイシン D(Sigma社)を最終濃度 5 x g/mlとなるように添加し、 0、 2、 4時間後にそれぞれ細胞を回収した。 A plasmid in which the gene encoding V9 or V18 was connected downstream of the CMV promoter was transfected into human K562 cells using Lipofectamine 2000 (Invitrogen) according to the attached protocol. After culturing at 37 ° C for 4 hours, PMA (Sigma) was added to a final concentration of 50 ng / ml. After culturing for 40 hours, actinomycin D (Sigma) was added to a final concentration of 5 x g / ml, and cells were collected after 0, 2, and 4 hours, respectively.
次いで、全 RNA抽出用試薬(RNeasyMiniおよび RNase- Free Dnase set: QIAGEN 社) を添付のプロトコ一ルに従って用いることにより、 全 RNAを抽出した。 な お、 回収した全 RNAは- 80°Cに保存した。 残存したヒト PADI4 V9もしくは V18 の mRNAを、 TaqMan One-step RT-PCR Master Mix (Applied Biosystems社) を用い、 指定のプロトコールに従って、 ABI PRISM 7700 Se uence Detection System (Appl ied Biosystems社) を用いて測定した。 なお、 得られたデータは リボゾ一マル RNA (18S) の値を用いて補正した。 乙 · ホロ宋 Next, total RNA was extracted using a total RNA extraction reagent (RNeasyMini and RNase-Free Dnase set: QIAGEN) according to the attached protocol. The collected total RNA was stored at -80 ° C. The remaining human PADI4 V9 or V18 mRNA is measured using ABI PRISM 7700 Sequence Detection System (Applied Biosystems) using TaqMan One-step RT-PCR Master Mix (Applied Biosystems) and following the specified protocol. did. The obtained data was corrected using the value of ribosomal RNA (18S). Otsu Holo Song
図 8に示すように、 PADI4 V9の niRNAは、 PADI4 V18に比べて有意に分解し難 い傾向にあることが分かった (T検定より、 4時間後で p=0. 03) 本明細書中で引用した全ての刊行物、 特許及び特許出願をそのまま参考とし て本明細書中にとり入れるものとする。 産業上の利用の可能性  As shown in FIG. 8, it was found that PADI4 V9 niRNA tended to be significantly harder to degrade than PADI4 V18 (p = 0.03 after 4 hours from T test). All publications, patents and patent applications cited in, are incorporated herein by reference in their entirety. Industrial potential
本発明は、 医薬開発において、 慢性関節リウマチ治療薬のスクリーニングに 利用できる。 また、 本発明は、 医療の分野において、 被験者が慢性関節リウマ チを発症する潜在的危険度の早期判定に利用できる。 配列表フリーテキスト  INDUSTRIAL APPLICABILITY The present invention can be used for screening drugs for treating rheumatoid arthritis in drug development. In addition, the present invention can be used in the medical field for early determination of the potential risk of a subject to develop rheumatoid arthritis. Sequence listing free text
配列番号 1ーヒト PADI4遺伝子断片(第 1イントロン- 1000〜第 4イントロン) 一変異の説明: (A)と(G)の置換による一塩基多型  SEQ ID NO: 1—Human PADI4 gene fragment (first intron-1000 to fourth intron) Description of one mutation: Single nucleotide polymorphism due to substitution of (A) and (G)
一変異の説明: (C)と(T)の置換による一塩基多型  Explanation of one mutation: Single nucleotide polymorphism due to substitution of (C) and (T)
一変異の説明: (C)と(G)の置換による一塩基多型  Explanation of one mutation: Single nucleotide polymorphism due to substitution of (C) and (G)
一変異の説明: (C)と(T)の置換による一塩基多型  Explanation of one mutation: Single nucleotide polymorphism due to substitution of (C) and (T)
一変異の説明: (C)と(T)の置換による一塩基多型  Explanation of one mutation: Single nucleotide polymorphism due to substitution of (C) and (T)
配列番号 2—人工配列の説明:プローブ  SEQ ID NO: 2—Description of Artificial Sequence: Probe
配列番号 3—人工配列の説明:プローブ  SEQ ID NO: 3—Description of artificial sequence: probe
配列番号 4一人工配列の説明:プローブ  SEQ ID NO: 4 Description of Artificial Sequence: Probe
配列番号 7—人工配列の説明:プライマー  SEQ ID NO: 7—Description of artificial sequence: primer
配列番号 8—人工配列の説明:プライマー  SEQ ID NO: 8—Description of artificial sequence: primer
配列番号 9一人工配列の説明:プライマ一  SEQ ID NO: 9 Description of artificial sequence: Primer
配列番号 1 0—人工配列の説明:プライマー  SEQ ID NO: 10—Description of artificial sequence: primer
配列番号 1 1一人工配列の説明:プローブ  SEQ ID NO: 1 Description of Artificial Sequence: Probe
配列番号 1 2—人工配列の説明:プライマー  SEQ ID NO: 12—Description of artificial sequence: primer
配列番号 1 3—人工配列の説明:プライマー  SEQ ID NO: 13—Description of artificial sequence: primer
配列番号 1 4一人工配列の説明:プローブ 配列番号 1 5 —人工配列の説明: ヒト PADI4由来べプチド SEQ ID NO: 14 Description of Artificial Sequence: Probe SEQ ID NO: 15—Description of artificial sequence: Human PADI4 derived peptide

Claims

1. ぺプチジルアルギニン ·ディミナーゼ ·タイプ I V遺伝子若しくはそのォ ーソログ遺伝子、 または該遺伝子にコードされる蛋白質に対する阻害効果 を指標として、 被験物質の慢性関節リウマチ治療薬としての効果を評価す る方法。 1. A method for evaluating the effect of a test substance as a therapeutic agent for rheumatoid arthritis using an inhibitory effect on a peptidylarginine diminase type IV gene or an ortholog gene thereof, or a protein encoded by the gene as an index.
2. 下記の工程を含む、 請求項 1に記載の方法:  2. The method of claim 1, comprising the following steps:
1 ) 動物を被験物質の投与請または非投与条件下で飼育する;  1) breed animals under conditions that allow or prevent administration of the test substance;
2 ) 上記動物の血液または細胞中におけるべプチジルアルギニン ·デイミ ナーゼ ·タイプ I V遺伝子またはそのォーソログ遺伝子の発現量を検出す る;  2) detecting the expression level of the beptidylarginine deiminase type IV gene or its ortholog gene in the blood or cells of the animal;
3 ) 被験物質の投与または非投与条件下にお囲ける、 上記発現量の相違に基 づいて、 該被験物質の慢性関節リウマチ治療薬としての効果を評価する。 3) The effect of the test substance as a therapeutic agent for rheumatoid arthritis is evaluated based on the difference in the expression level under the conditions of administration or non-administration of the test substance.
3. 遺伝子の発現量が、 遺伝子チップ、 c D NAアレイ、 およびメンブレンフ ィルターから選ばれる固相化試料を用いた核酸ハイブリダィゼ一シヨン法、3. The nucleic acid hybridization method using a solid-phased sample selected from a gene chip, a cDNA array, and a membrane filter,
R T- P C R法、 リアルタイム P C R法、 サブトラクシヨン法、 ディファレ ンシャル ·ディスプレイ法、 ディファレンシャル ·ハイブリダィゼーショ ン法、 ならびにクロスハイブリダイゼーション法から選ばれるいずれか一 つの方法によって検出されることを特徴とする、 請求項 2に記載の方法。 RT-PCR, real-time PCR, subtraction, differential display, differential hybridization, and cross-hybridization. The method of claim 2, wherein the method is characterized by:
4. 遺伝子の発現量が、 R T- P C R法によって検出されることを特徴とする、 請求項 3に記載の方法。 4. The method according to claim 3, wherein the expression level of the gene is detected by an RT-PCR method.
5. 下記の工程を含む、 請求項 1に記載の方法:  5. The method of claim 1, comprising the following steps:
1 ) 動物を被験物質の投与または非投与条件下で飼育する;  1) keep the animals under the conditions of administration or non-administration of the test substance;
2 ) 上記動物の血液または細胞中において、 ぺプチジルアルギニン ·ディ ミナーゼ 'タイプ I V遺伝子またはそのォーソログ遺伝子にコードされる 蛋白質の発現量を、 該蛋白質に特異的に結合する抗体を用いて検出する; 2) In the blood or cells of the above animal, the expression level of the protein encoded by the peptidylarginine diminase type IV gene or its ortholog gene is detected using an antibody that specifically binds to the protein. ;
3 ) 被験物質の投与または非投与条件下における、 上記発現量の相違に基 づいて、 該被験物質の慢性関節リウマチ治療薬としての効果を評価する。3) The effect of the test substance as a therapeutic agent for rheumatoid arthritis is evaluated based on the difference in the expression level under the conditions of administration or non-administration of the test substance.
6. 蛋白質の発現量が、 ウエスタンプロット法、 ドットプロット法、 スロット ブロット法、 E L I S A法、 および R I A法から選ばれるいずれか一つの 方法によって検出されることを特徴とする、 請求項 5に記載の方法。 6. The protein expression level is selected from one of the following: Western plot, dot plot, slot blot, ELISA, and RIA. The method according to claim 5, wherein the method is detected by a method.
7. 蛋白質の発現量が E L I S A法によって検出されることを特徴とする、 請 求項 6に記載の方法。  7. The method according to claim 6, wherein the expression level of the protein is detected by an ELISA method.
8. 細胞が血液由来細胞、 滑膜細胞、 脾臓細胞、 および腹腔浸潤細胞から選ば れることを特徴とする、 請求項 2〜 7のいずれか一項に記載の方法。  8. The method according to any one of claims 2 to 7, wherein the cells are selected from blood-derived cells, synovial cells, spleen cells, and peritoneal cells.
9. 動物がマウスであり、 マウスべプチジルアルギニン ·ディミナ一ゼ'タイ プ I V遺伝子、 または該遺伝子にコードされる蛋白質の発現量を検出する ことを特徴とする、 請求項 2〜 7のいずれか一項に記載の方法。  9. The animal according to claim 2, wherein the animal is a mouse, and the expression level of a mouse beptidylarginine-diminase 'type IV gene or a protein encoded by the gene is detected. The method according to any one of the preceding claims.
10. 下記の工程を含む、 請求項 1に記載の方法:  10. The method of claim 1, comprising the following steps:
1 ) アルギニン含有化合物を含む反応液に、 被験物質の添加または非添加 条件下で、 ぺプチジルアルギニン ·ディミナーゼ ·タイプ I V蛋白質を加 える。  1) Add peptidylarginine / diminase / type IV protein to the reaction solution containing the arginine-containing compound with or without the test substance.
2 ) 上記反応液中における反応生成物の量を定量することにより、 ぺプチ ジルアルギニン ·ディミナ一ゼ ·タイプ I V蛋白質の活性を測定する。 3 ) 被験物質の添加または非添加条件下における、 上記活性の相違に基づ いて、 該被験物質の慢性関節リゥマチ治療薬としての効果を評価する。 2) The activity of peptidylarginine / diminase / type IV protein is determined by quantifying the amount of the reaction product in the above reaction solution. 3) Evaluate the effect of the test substance as a therapeutic agent for rheumatoid arthritis based on the difference in the above activities under the conditions of addition or non-addition of the test substance.
11. 蛋白質の活性が、 蛍光法または吸光法を利用して測定されることを特徴と する請求項 1 0に記載の方法。 11. The method according to claim 10, wherein the activity of the protein is measured by using a fluorescence method or an absorption method.
12. ぺプチジルアルギニン ·ディミナ一ゼ ·タイプ I V蛋白質がヒトぺプチジ ルアルギニン ·ディミナ一ゼ ·タイプ I V蛋白質である、 請求項 1 0また は 1 1記載の方法。  12. The method according to claim 10 or 11, wherein the peptidyl arginine diminase type IV protein is a human peptidyl arginine diminase type IV protein.
13. 検体中の配列番号 2 1に示されるアミノ酸配列からなる変異型べプチジル アルギニン ·ディミナ一ゼ ·タイプ I V蛋白質、 またはその遺伝子 (配列 番号 2 0 ) の発現量に基づいて、 該検体を提供した被験者の慢性関節リウ マチの発症危険度を予測する方法。  13. Provide the sample based on the expression level of the mutant beptidyl arginine, diminase, type IV protein consisting of the amino acid sequence shown in SEQ ID NO: 21 or the gene (SEQ ID NO: 20) in the sample or the gene To predict the risk of developing rheumatoid arthritis in affected subjects.
14. 以下の工程を含む、 請求項 1 3に記載の方法:  14. The method of claim 13, comprising the following steps:
1 ) 被験者および正常人から単離された各検体より全 R NAを調製する; 1) Prepare total RNA from each sample isolated from subjects and normal persons;
2 )上記全 R NA中における変異型べプチジルアルギニン ·ディミナ一ゼ · タイプ I V遺伝子の mR N Aの発現量を検出する; 2) detecting the expression level of mRNA of the mutant beptidylarginine / diminase / type IV gene in all the RNAs;
3 ) 被験者と正常人における上記発現量の相違を解析し、 被験者の慢性関 節リゥマチの発症危険度を予測する。 3) Analyze the difference between the above expression levels between the subjects and normal subjects, and Predict the risk of developing rheumatoid arthritis.
15. 遺伝子の発現量が、 遺伝子チップ、 cDNAアレイ、 およびメンブレンフ ィルターから選ばれる固相化試料を用いた核酸ハイブリダイゼーシヨン法、 RT-PCR法、 リアルタイム PC R法、 サブトラクシヨン法、 ディファレ ンシャル ·ディスプレイ法、 ディファレンシャル ·ハイブリダィゼーショ ン法、 ならびにクロスハイブリダィゼーシヨン法から選ばれるいずれか一 つの方法によって検出されることを特徴とする、請求項 14に記載の方法。 15. Gene expression level is determined by nucleic acid hybridization method, RT-PCR method, real-time PCR method, subtraction method using immobilized sample selected from gene chip, cDNA array and membrane filter. 15. The method according to claim 14, wherein the detection is performed by any one method selected from a differential display method, a differential hybridization method, and a cross-hybridization method.
16. 遺伝子の発現量が、 RT-PCR法によって検出されることを特徴とする、 請求項 15に記載の方法。 16. The method according to claim 15, wherein the expression level of the gene is detected by an RT-PCR method.
17. 以下の工程を含む、 請求項 13に記載の方法:  17. The method of claim 13, comprising the following steps:
1) 被験者および正常人から単離された検体中における、 変異型べプチジ ルアルギニン ·ディミナーゼ ·タイプ I V蛋白質の発現量を該蛋白質に特 異的に結合しうる抗体を用いて検出する;  1) detecting the expression level of a mutant beptidylarginine / diminase / type IV protein in a sample isolated from a subject or a normal person using an antibody capable of binding specifically to the protein;
2) 被験者と正常人における上記発現量の相違を解析し、 該被験者の慢性 関節リゥマチの発症危険度を予測する。  2) Analyze the difference in the expression level between the test subject and the normal person, and predict the risk of developing rheumatoid arthritis in the test subject.
18. 蛋白質の発現量が、 ウエスタンプロット法、 ドットプロット法、 スロット ブロット法、 EL I SA法、 および R I A法から選ばれるいずれか一つの 方法によって検出されることを特徴とする、 請求項 17に記載の方法。 18. The method according to claim 17, wherein the expression level of the protein is detected by any one method selected from Western plotting, dot plotting, slot blotting, ELISA, and RIA. The described method.
19. 蛋白質の発現量が EL I S A法によって検出されることを特徴とする、 請 求項 18に記載の方法。 19. The method according to claim 18, wherein the expression level of the protein is detected by an ELISA method.
20. 以下の a) 〜e) からなる群より選ばれる少なくとも 1以上を含む、 慢性 関節リウマチの発症危険度を予測するためのキット。  20. A kit for predicting the risk of developing rheumatoid arthritis, comprising at least one selected from the group consisting of: a) to e) below.
a) 配列番号 20に示される塩基配列からなる変異型べプチジルアルギニ ン-ディミナ一ゼ*タイプ IV遺伝子を特異的に増幅するための、 15〜3 0塩基長の連続したオリゴヌクレオチドプライマ一  a) A continuous oligonucleotide primer having a length of 15 to 30 bases for specifically amplifying a mutant beptidylarginine-diminase * type IV gene consisting of the base sequence shown in SEQ ID NO: 20
b) 配列番号 20に示される塩基配列からなる変異型べプチジルアルギニ ン 'ディミナ一ゼ 'タイプ I V遺伝子に特異的にハイプリダイズし、 該遺伝 子を検出するための 20〜1500塩基長の連続したポリヌクレオチドプ ローブ  b) A 20- to 1500-base-long continuous polynucleotide for specifically hybridizing to the mutant beptidyl arginine 'diminase' type IV gene having the nucleotide sequence shown in SEQ ID NO: 20 and detecting the gene. Nucleotide probe
c) 上記 b) 記載のポリヌクレオチドプローブが固定された固相化試料 d )配列番号 2 1に示されるアミノ酸配列からなる変異型べプチジルアルギ ニン ·デイミナーゼ ·タイプ I V蛋白質に特異的に結合し、 該蛋白質を検出 するための抗体 c) Immobilized sample on which the polynucleotide probe described in b) above is immobilized d) an antibody that specifically binds to a mutant beptidylarginine deiminase type IV protein consisting of the amino acid sequence represented by SEQ ID NO: 21 and detects the protein
e ) 上記 d ) 記載の抗体に特異的に結合しうる二次抗体  e) secondary antibody capable of specifically binding to the antibody according to d) above
21. 配列番号 2 1に示されるアミノ酸配列からなる変異型べプチジルアルギニ ン ·デイミナーゼ 'タイプ I V蛋白質に対する阻害効果を指標として、 被験 物質の慢性関節リウマチ治療薬としての効果を評価する方法。 21. A method for evaluating the effect of a test substance as a therapeutic agent for rheumatoid arthritis using an inhibitory effect on a mutant type beptidylarginine deiminase 'type IV protein having the amino acid sequence shown in SEQ ID NO: 21 as an index.
22. 下記の工程を含む、 請求項 2 1に記載の方法:  22. The method of claim 21 comprising the following steps:
1 )アルギニン含有化合物を含む反応液に、被験物質の添加または非添加条 件下で、 変異型べプチジルアルギニン ·ディミナ一ゼ'タイプ I V蛋白質を 加える;  1) To a reaction solution containing an arginine-containing compound, a mutant beptidylarginine-diminase 'type IV protein is added under the conditions of adding or not adding a test substance;
2 )上記反応液中における反応生成物の量を定量することにより、変異型べ プチジルアルギニン ·ディミナ一ゼ'タイプ I V蛋白質の活性を測定する; 2) measuring the activity of the mutant beptidylarginine-diminase 'type IV protein by quantifying the amount of the reaction product in the reaction solution;
3 )被験物質の添加または非添加条件下における、上記活性の相違に基づい て、 該被験物質の慢性関節リウマチ治療薬としての効果を評価する。 3) The effect of the test substance as a therapeutic agent for rheumatoid arthritis is evaluated based on the difference in the activity under the conditions of addition or non-addition of the test substance.
23. 配列番号 2 1に示されるアミノ酸配列からなる変異型べプチジルアルギニ ン ·デイミナーゼ ·タイプ I V蛋白質。  23. A mutant beptidylarginine deiminase type IV protein consisting of the amino acid sequence shown in SEQ ID NO: 21.
PCT/JP2004/000430 2003-01-21 2004-01-20 Method of using peptidyl arginine deiminase type iv WO2004065633A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2005508091A JP4503531B2 (en) 2003-01-21 2004-01-20 How to use peptidylarginine deiminase type IV

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
JP2003012774 2003-01-21
JP2003-012738 2003-01-21
JP2003-012774 2003-01-21
JP2003012738 2003-01-21

Publications (1)

Publication Number Publication Date
WO2004065633A1 true WO2004065633A1 (en) 2004-08-05

Family

ID=32775167

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2004/000430 WO2004065633A1 (en) 2003-01-21 2004-01-20 Method of using peptidyl arginine deiminase type iv

Country Status (3)

Country Link
JP (1) JP4503531B2 (en)
TW (1) TW200502398A (en)
WO (1) WO2004065633A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008029619A1 (en) * 2006-09-07 2008-03-13 Daiichi Sankyo Company, Limited Ena antisense oligonucleotide having sequence-specific action
JP2009156615A (en) * 2007-12-25 2009-07-16 Tokyo Metropolitan Foundation For Social Welfare & Public Health Method for measuring pad 4 and anti-pad 4 antibody and method of detecting articular rheumatism
WO2010115745A3 (en) * 2009-03-30 2010-12-16 INSERM (Institut National de la Santé et de la Recherche Médicale) Biomarkers, methods and kits for the diagnosis of rheumatoid arthritis

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
CORNELIS F. ET AL: "New susceptibility locus for rheumatoid arthritis suggested by a genome-wide linkage study", PNAS, vol. 95, no. 18, 1998, pages 10746 - 10750, XP002979061 *
GIRBAL-NEUHAUSER E. ET AL: "The epitopes targeted by the rheumatoid arthritis-associated antifilaggrin autoantibodies are posttranslationally generated on various sites of (pro)filaggrin by deimination of arginine residues", J. IMMUNOL., vol. 162, no. 1, 1999, pages 585 - 584, XP002108878 *
NAKASHIMA K. ET AL: "Molecular characterization of peptidylarginine deiminase in HL-60 cells induced by retinoic acid and 1alpha,25-dihydroxyvitamin D3", EUR. J. CHEM., vol. 274, no. 39, 1999, pages 27786 - 27792, XP002177280 *
RUS'D A.A. ET AL: "Molecular cloning of cDNAs of mouse peptidylarginine deiminase type I, type III and type IV, and the expression pattern of type I in mouse", EUR. J. BIOCHEM., vol. 259, no. 3, 1999, pages 660 - 669, XP002177600 *
SUZUKI A. ET AL: "Functional haplotypes of PADI4, encoding citrullinating enzyme peptidylarginine deiminase 4, are associated with rheumatoid arthritis", NATURE GENETICS, vol. 34, no. 4, August 2003 (2003-08-01), pages 395 - 402, XP002979063 *
VAN VENROOIJ W.J. ET AL: "Citrullination: a small change for a protein with great consequences for rheumatoid arthritis", ARTHRITIS RES., vol. 2, no. 4, 2000, pages 249 - 251, XP002979062 *
YAMADA R. ET AL: "Peptidylarginine deiminase type 4: identification of a rheumatoid arthritis-susceptible gene", TRENDS MOL. MED., vol. 9, no. 11, November 2003 (2003-11-01), pages 503 - 508, XP002979064 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008029619A1 (en) * 2006-09-07 2008-03-13 Daiichi Sankyo Company, Limited Ena antisense oligonucleotide having sequence-specific action
JP2009156615A (en) * 2007-12-25 2009-07-16 Tokyo Metropolitan Foundation For Social Welfare & Public Health Method for measuring pad 4 and anti-pad 4 antibody and method of detecting articular rheumatism
WO2010115745A3 (en) * 2009-03-30 2010-12-16 INSERM (Institut National de la Santé et de la Recherche Médicale) Biomarkers, methods and kits for the diagnosis of rheumatoid arthritis
AU2010233926B2 (en) * 2009-03-30 2014-01-16 Inserm (Institut National De La Sante Et De La Recherche Medicale) Biomarkers, methods and kits for the diagnosis of Rheumatoid Arthritis

Also Published As

Publication number Publication date
JPWO2004065633A1 (en) 2006-05-18
JP4503531B2 (en) 2010-07-14
TW200502398A (en) 2005-01-16

Similar Documents

Publication Publication Date Title
DK2414543T3 (en) Genetic markers for risk management of atrial fibrillation and stroke
JP2009544314A (en) Mutation of PKD and its evaluation
US8114592B2 (en) Genetic markers associated with age-related macular degeneration, methods of detection and uses thereof
JP2005510251A (en) Evaluation method and prognostication method of sarcoidosis
EP2297325A1 (en) Methods of stratifying, prognosing and diagnosing schizophrenia, mutant nucleic acid molecules and polypeptides
Banno et al. Association of genetic polymorphisms of endothelin-converting enzyme-1 gene with hypertension in a Japanese population and rare missense mutation in preproendothelin-1 in Japanese hypertensives
WO2004065633A1 (en) Method of using peptidyl arginine deiminase type iv
US8129142B2 (en) Mutations in ion channels
JP4688492B2 (en) Determination method of inflammatory diseases
US20070249518A1 (en) Compositions and Methods for Treating Mental Disorders
JP5034086B2 (en) Test method for inflammatory diseases and screening method for therapeutic agents for inflammatory diseases
JP2006526986A (en) Diagnosis method for inflammatory bowel disease
JP4668792B2 (en) Method for determining inflammatory disease using single nucleotide polymorphism in galectin-2 gene
JP4546397B2 (en) How to diagnose kidney disease or predisposition
US20190309366A1 (en) CXR4 as a Susceptibility Locus in Juvenile Idiopathic Arthritis (JIA) and Methods of Use Thereof for the Treatment and Diagnosis of the Same
JP2009535033A (en) Compositions and methods for detection of Cripto-3
US8067158B2 (en) Methods and compositions for the diagnosis and treatment of schizophrenia
JP5644009B2 (en) Determination method of inflammatory disease using single nucleotide polymorphism
JP2004154136A (en) METHOD FOR EVALUATING beta CELL DYSFUNCTION IMPROVING AGENT
JP4421889B2 (en) How to predict the risk of developing rheumatoid arthritis
JP4801596B2 (en) Method for examining inflammatory disease and screening method for inflammatory disease therapeutic agent
JP4982771B2 (en) Determination method of inflammatory diseases
JP2005516626A (en) Polymorphisms in the human gene of TPMT and methods of use thereof in diagnostic and therapeutic applications
WO2003072824A1 (en) Markers for predicting pathological conditions in haert failure and method of using the same
JP2004000155A (en) Marker for predicting condition of cardiac failure and method for utilizing the same

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2005508091

Country of ref document: JP

122 Ep: pct application non-entry in european phase