[go: up one dir, main page]

WO2016031996A1 - Agent prophylactique/thérapeutique contre l'arthrite, trousse d'essai pour l'arthrite, et procédé de criblage d'agent prophylactique/thérapeutique contre l'arthrite - Google Patents

Agent prophylactique/thérapeutique contre l'arthrite, trousse d'essai pour l'arthrite, et procédé de criblage d'agent prophylactique/thérapeutique contre l'arthrite Download PDF

Info

Publication number
WO2016031996A1
WO2016031996A1 PCT/JP2015/074556 JP2015074556W WO2016031996A1 WO 2016031996 A1 WO2016031996 A1 WO 2016031996A1 JP 2015074556 W JP2015074556 W JP 2015074556W WO 2016031996 A1 WO2016031996 A1 WO 2016031996A1
Authority
WO
WIPO (PCT)
Prior art keywords
tet
arthritis
gene
nucleic acid
expression
Prior art date
Legal status (The legal status 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 status listed.)
Ceased
Application number
PCT/JP2015/074556
Other languages
English (en)
Japanese (ja)
Inventor
和久 中野
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Occupational and Environmental Health Japan
Original Assignee
University of Occupational and Environmental Health Japan
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 University of Occupational and Environmental Health Japan filed Critical University of Occupational and Environmental Health Japan
Priority to JP2016545659A priority Critical patent/JPWO2016031996A1/ja
Publication of WO2016031996A1 publication Critical patent/WO2016031996A1/fr
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • 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
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • G01N33/5023Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects on expression patterns
    • 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
    • G01N33/564Immunoassay; Biospecific binding assay; Materials therefor for pre-existing immune complex or autoimmune disease, i.e. systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, rheumatoid factors or complement components C1-C9
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering nucleic acids [NA]
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/10Musculoskeletal or connective tissue disorders
    • G01N2800/101Diffuse connective tissue disease, e.g. Sjögren, Wegener's granulomatosis
    • G01N2800/102Arthritis; Rheumatoid arthritis, i.e. inflammation of peripheral joints

Definitions

  • the present invention relates to a prophylactic / therapeutic agent for arthritis, particularly rheumatoid arthritis (RA), and a screening method for a prophylactic / therapeutic agent for arthritis, particularly rheumatoid arthritis. More specifically, arthritis, particularly a prophylactic and / or therapeutic agent for rheumatoid arthritis, which contains a substance that inhibits the function of Tet 3 (Ten-Eleven tranlocation 3), and arthritis using Tet 3 function inhibition as an index, In particular, the present invention relates to a method for screening a candidate substance for a prophylactic / therapeutic agent for rheumatoid arthritis.
  • RA rheumatoid arthritis
  • Rheumatoid arthritis is an intractable autoimmune disease that causes chronic inflammation in joints throughout the body.
  • the synovial tissue in the joint grows and progressively destroys cartilage and bone.
  • Rheumatoid arthritis synovial fibroblasts FLS are activated by stimulation (inflammatory cytokines such as TNF ⁇ and IL-1 ⁇ ) by macrophages and lymphocytes, and activated synovial fibroblasts While chemokine production and substrate-degrading enzyme secretion cause joint destruction, it also has properties similar to cancer cells, such as increased proliferation and invasion and decreased apoptosis sensitivity.
  • Non-patent Document 1 Rheumatoid arthritis treatment targeting inflammatory cytokines with biologics has made remission induction familiar, but there is a so-called “Window of Opportunity” in rheumatoid arthritis treatment, and the delay in treatment start has a therapeutic effect. Limited.
  • Tet Te-Eleven transcription
  • an object of the present invention is to clarify the function of Tet protein, particularly Tet3 protein, in arthritis, particularly rheumatoid arthritis, and based on this function, a prophylactic / therapeutic agent and test kit for arthritis targeting Tet3 And a means for searching for a novel substance having arthritis preventive and / or therapeutic activity using Tet 3 function regulation as an index.
  • the present inventor has expressed the expression level of Tet protein family and DNA in synovial fibroblasts (FLS) derived from rheumatoid arthritis patients stimulated with synovium or inflammatory cytokines derived from rheumatoid arthritis patients. Methylation levels were measured.
  • FLS synovial fibroblasts
  • OA osteoarthritis
  • a prophylactic and / or therapeutic agent for arthritis which contains an inhibitor of expression of Tet 3 (Ten-Eleven transcription 3).
  • An expression inhibitor of Tet 3 is (A) an antisense nucleic acid against a transcript of the Tet 3 gene, The agent according to [1], which is (b) a ribozyme nucleic acid for a transcription product of the Tet 3 gene, or (c) a nucleic acid having RNAi activity for a transcription product of the Tet 3 gene or a precursor thereof.
  • the agent according to [1] or [2], wherein the arthritis is rheumatoid arthritis, psoriatic arthritis or spondyloarthritis.
  • a screening method for a prophylactic and / or therapeutic drug for arthritis comprising the following steps (1) to (3): (1) contacting a cell containing a nucleic acid encoding a Tet 3 gene or a reporter protein under the control of a transcriptional regulatory region of the gene with a test substance; (2) measuring the expression level of the Tet 3 gene, Tet 3 protein or reporter protein in the cell, (3) A test substance in which the expression level of the Tet 3 gene, Tet 3 protein, or reporter protein is reduced as compared with the case where it is measured in the absence of the test substance is used as a prophylactic and / or therapeutic drug for arthritis. Selecting as a candidate.
  • the method further includes applying a test substance selected as a candidate for a prophylactic and / or therapeutic agent for arthritis to an arthritis model and testing whether or not to suppress an inflammatory reaction in the model. The method described.
  • a screening method for a prophylactic and / or therapeutic agent for arthritis comprising the following steps (1) to (3): (1) a step of bringing synovial fibroblasts into contact with a test substance; (2) measuring the degree of demethylation or invasiveness of 5-methylcytosine (5 mC) in the genome of the cell, (3) Compared to the measurement in the absence of the test substance, the test substance that suppresses the demethylation or invasiveness of 5-methylcytosine (5mC) in the genome of the cell is used to prevent arthritis and And / or selecting as a candidate for a therapeutic agent.
  • a method for examining arthritis comprising detecting or quantifying a transcription product or translation product of the Tet 3 gene from a sample derived from a subject using the following (a) or (b): (A) a nucleic acid probe or nucleic acid primer capable of specifically detecting a transcription product of the Tet 3 gene; and (b) an antibody that specifically recognizes a translation product of the Tet 3 gene.
  • the kit according to [11] wherein the arthritis is rheumatoid arthritis, psoriatic arthritis or spondyloarthritis.
  • a method for preventing and / or treating arthritis comprising administering to a subject an effective amount of an expression inhibitor of Tet 3 (Ten-Eleven transcription 3).
  • Tet 3 is involved in the pathological deterioration of rheumatoid arthritis
  • arthritis can be treated or prevented by inhibiting the expression or function of Tet 3.
  • therapeutic or prophylactic agents for arthritis can be screened using Tet 3 expression or function inhibition as an index.
  • arthritis can be examined using the expression of Tet 3 as an index.
  • FIG. 1 shows expression of Tet protein family (Tet1,2,3), DNA methylation (5-methylcytosine (5 mC), 5-hydroxymethylcytosine (5 hmC)) in synovial tissue derived from patients with rheumatoid arthritis (RA). It is an immunohistochemical dyeing
  • FIG. 2 shows the expression of Tet protein family (Tet1,2,3), DNA methylation (5-methylcytosine (5 mC), 5-hydroxymethylcytosine (5 hmC) in synovial tissue from osteoarthritis (OA) patients. It is an immunohistochemical staining image showing)).
  • FIG. 1 shows expression of Tet protein family (Tet1,2,3), DNA methylation (5-methylcytosine (5 mC), 5-hydroxymethylcytosine (5 hmC) in synovial tissue from osteoarthritis (OA) patients. It is an immunohistochemical staining image showing)).
  • FIG. 1 shows expression of Tet protein family (Tet
  • FIG. 3 shows immunity showing double staining of Tet3 protein (blue) and CD55 protein (brown) and double staining of Tet3 protein (blue) and CD68 protein (brown) in synovial tissue from patients with rheumatoid arthritis (RA). It is a histochemical staining image.
  • FIG. 4 is a diagram showing the relative mRNA expression levels of the Tet family (Tet1, Tet 2, Tet 3) in cytokine-unstimulated FLS (normal subjects, OA, RA).
  • FIG. 5 is an immunohistochemically stained image showing the expression of the Tet family (green) (Tet1, Tet 2, Tet 3) and the nucleus (blue) in cytokine-unstimulated FLS (OA, RA).
  • FIG. 7 is a graph showing the change over time in the N / C ratio of Tet3 protein in FLS after stimulation with TNF ⁇ .
  • FIG. 8A is a diagram showing the protein expression level of Tet3 in OA-derived or healthy subject-derived FLS (OA, Nr) after stimulation with TNF ⁇ .
  • FIG. 8B shows the protein expression level of Tet 3 in AR-derived FLS after stimulation with TNF ⁇ .
  • FIG. 9A is a diagram showing a test process of Example 4.
  • FIG. 9B is a dot blot image showing 5 hmC levels in AR-derived FLS after stimulation with TNF ⁇ .
  • FIG. 9C is a graph showing the 5hmC level in AR-derived FLS.
  • FIG. 10A is a diagram showing a test process of Example 5.
  • FIG. 10A is a diagram showing a test process of Example 5.
  • FIG. 10B is a graph showing the secretion level of inflammatory cytokines in AR-derived FLS after knocking down Tet3 and stimulation with TNF ⁇ .
  • FIG. 11A is a diagram showing a test process of Example 6.
  • FIG. 11B is a micrograph showing Scratch assay of AR-derived FLS after knocking down Tet3 and stimulation with TNF ⁇ .
  • FIG. 11C is a graph showing the number of infiltrating cells per unit area of AR-derived FLS after knocking down Tet 3, stimulating with TNF ⁇ , and 24 hours after scratch.
  • the present invention is based, at least in part, on the discovery that Tet 3 contributes to exacerbation of rheumatoid arthritis.
  • This finding indicates that Tet 3 can be used not only as a marker for arthritis, particularly rheumatoid arthritis, but also as a drug discovery target for arthritis, particularly rheumatoid arthritis. That is, a known inhibitor of Tet 3 is useful for prevention and / or treatment of arthritis, and a novel Tet 3 inhibitor, and thus prevention / treatment of arthritis, using Tet3 protein and cells / animals expressing it. You can also search for substances that will be drugs. I. Tet 3 or nucleic acid encoding it In this specification, Tet 3 is a known protein, and Genbank Accession No.
  • Tet 3 A protein containing the amino acid sequence of human Tet 3 represented by SEQ ID NO: 2, or the amino acid sequence substantially identical thereto, known as 043151.
  • proteins and peptides are described with the N-terminus (amino terminus) at the left end and the C-terminus (carboxyl terminus) at the right end according to the convention of peptide notation.
  • Tet 3 refers to cells of humans and other warm-blooded animals (eg, mice, rats, cows, monkeys, dogs, pigs, sheep, rabbits, guinea pigs, hamsters, chickens, etc.) [eg, synovial fibroblasts.
  • amino acid sequence represented by SEQ ID NO: 2 or an amino acid sequence substantially identical thereto include the following (a) to (e): (A) the amino acid sequence represented by SEQ ID NO: 2; (B) In the amino acid sequence represented by SEQ ID NO: 2, one or more amino acids are deleted, added, inserted or substituted, and demethylating activity of 5-methylcytosine (5 mC) or synovial fibroblasts An amino acid sequence having invasion promoting activity; (C) an amino acid having 90% or more homology with the amino acid sequence represented by SEQ ID NO: 2 and having a demethylating activity of 5-methylcytosine (5 mC) or an activity of promoting invasion of synovial fibroblasts Sequence; (D) an amino acid sequence encoded by DNA having the base sequence represented by
  • the human Tet 3 protein consisting of the amino acid sequence represented by SEQ ID NO: 2 or the amino acid sequence of an ortholog in mammals or the human Tet 3 protein consisting of the amino acid sequence represented by SEQ ID NO: 2 or The amino acid sequence in the splice variant, allelic variant or polymorphic variant of the ortholog may be mentioned.
  • “homology” refers to an optimal alignment when two amino acid sequences are aligned using a mathematical algorithm known in the art (preferably the algorithm uses a sequence of sequences for optimal alignment). The percentage of identical and similar amino acid residues relative to all overlapping amino acid residues in which one or both of the gaps can be considered).
  • Similar amino acids mean amino acids that are similar in physicochemical properties, such as aromatic amino acids (Phe, Trp, Tyr), aliphatic amino acids (Ala, Leu, Ile, Val), polar amino acids (Gln, Asn). ), Basic amino acids (Lys, Arg, His), acidic amino acids (Glu, Asp), amino acids having hydroxyl groups (Ser, Thr), amino acids with small side chains (Gly, Ala, Ser, Thr, Met), etc. Examples include amino acids classified into groups. It is expected that substitution with such similar amino acids will not change the phenotype of the protein (ie, is a conservative amino acid substitution).
  • the stringent conditions in (e) above are, for example, the conditions described in Current Protocols in Molecular Biology, John Wiley & Sons, 6.3.1-6.3.6, 1999, for example, 6 ⁇ SSC ( sodium chloride / sodium citrate) / hybridization at 45 ° C., followed by one or more washings at 0.2 ⁇ SSC / 0.1% SDS / 50 to 65 ° C. Hybridization conditions that give the same stringency can be selected as appropriate.
  • amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 2 is about 90% or more, preferably about 95% or more, More preferred is an amino acid sequence having an identity of about 96% or more, more preferably about 97% or more, particularly preferably about 98% or more, and most preferably about 99% or more.
  • a protein comprising an amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 2 comprises an amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 2, and SEQ ID NO: 2 is a protein having substantially the same function as the protein consisting of the amino acid sequence represented by 2.
  • substantially the same function means that the properties are qualitatively the same, for example, physiologically or pharmacologically, and the degree of function (eg, about 0.1 to about Quantities such as about 10 times, preferably 0.5 to 2 times) and the molecular weight of the protein may be different.
  • Tet3 has a function of converting 5-methylcytosine (5mC) into 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC) or 5-carboxyl cytosine (5CaC) (hereinafter referred to as “5 -Demethylation activity of methylcytosine (5 mC) ”), promotion activity of synovial fibroblast invasion, etc., and a protein having the above activity is regarded as“ a protein having substantially the same function ” be able to.
  • the demethylation activity of 5-methylcytosine (5 mC) and the promotion activity of infiltration of synovial fibroblasts can be measured, for example, as described in Examples described later.
  • Tet3 protein in the present invention examples include (i) 1 to 30, preferably 1 to 10, more preferably 1 to the number (5, 4, 3, or 2) in the amino acid sequence represented by SEQ ID NO: 2. ) An amino acid sequence in which one amino acid is deleted, (ii) 1 to 30, preferably 1 to 10, more preferably 1 to a number (5, 4, 3, or 3) in the amino acid sequence represented by SEQ ID NO: 2. 2) an amino acid sequence to which one amino acid is added, (iii) 1 to 30, preferably 1 to 10, more preferably 1 to a number (5, 4, 3, or 3) in the amino acid sequence represented by SEQ ID NO: 2.
  • amino acid sequence in which one amino acid is inserted (iv) 1 to 30, preferably 1 to 10, more preferably 1 to a number (5, 4, or 4) in the amino acid sequence represented by SEQ ID NO: 2. 3 or 2) substitution of another amino acid with another amino acid
  • the amino acid sequence was, or (v) is also included, such as protein comprising the amino acid sequence comprising a combination thereof.
  • the position of the insertion, deletion, addition or substitution is determined by demethylation of 5-methylcytosine (5 mC) or synovial membrane. There is no particular limitation as long as fibroblast infiltration can be promoted.
  • a technique for artificially performing amino acid deletion, addition, insertion or substitution for example, conventional site-directed mutagenesis is applied to DNA encoding the amino acid sequence represented by SEQ ID NO: 2. Thereafter, a technique for expressing this DNA by a conventional method can be mentioned.
  • a site-specific mutagenesis method for example, a method utilizing amber mutation (gapped duplex method, Nucleic Acids Res., 12, 9441-9456 (1984)), a PCR method using a mutagenesis primer.
  • Preferred examples of Tet 3 include, for example, a human protein consisting of the amino acid sequence represented by SEQ ID NO: 2 (Genbank Accession No.
  • Nucleic acid encoding Tet 3 is a nucleic acid comprising the amino acid sequence represented by SEQ ID NO: 2 shown in the above (a) to (e) or a base sequence encoding an amino acid sequence substantially identical thereto. Represents.
  • the gene may be DNA such as cDNA or genomic DNA, or RNA such as mRNA, and is a concept including both a single-stranded nucleic acid sequence and a double-stranded nucleic acid sequence.
  • the nucleic acid sequence shown in SEQ ID NO: 1 or the like is a DNA sequence for convenience, but when an RNA sequence such as mRNA is shown, thymine (T) is interpreted as uracil (U).
  • Preferred examples of the nucleic acid encoding Tet 3 include, for example, human Tet 3 cDNA (Genbank Accession No.
  • NM_001287491 consisting of the base sequence represented by SEQ ID NO: 1, or its orthologs and allelic variants in other mammals And polymorphic variants [for example, single nucleotide polymorphisms (SNPs)] and the like.
  • the present invention provides a prophylactic and / or therapeutic agent for arthritis, comprising a substance that inhibits the expression of Tet 3. II.
  • Substances that inhibit the expression of Tet 3 refers to any transcription level of nucleic acid encoding Tet3 (Tet3 gene), level of post-transcriptional regulation, level of translation into Tet 3 protein, level of post-translational modification, etc. It may act in stages.
  • substances that inhibit Tet3 expression include, for example, substances that inhibit Tet3 gene transcription (eg, antigenes), substances that inhibit the processing of early transcription products into mRNA, and mRNA transport to the cytoplasm.
  • substances that inhibit translation of Tet 3 from mRNA eg, antisense nucleic acid, miRNA
  • substances that degrade mRNA eg, siRNA, ribozyme
  • substances that inhibit post-translational modification of the initial translation product It is. Any substance acting at any stage can be preferably used, but more preferably, a substance selected from the group consisting of the following (1) to (3) is exemplified.
  • an antisense nucleic acid against a transcript of the Tet 3 gene (2) a ribozyme nucleic acid for the transcription product of the Tet 3 gene, (3) A nucleic acid having RNAi activity for a transcription product of the Tet3 gene or a precursor thereof.
  • a preferable example of the transcription product is mRNA.
  • the base sequence is complementary or substantially complementary to the base sequence of these mRNAs or its A nucleic acid containing a part is mentioned.
  • the base sequence substantially complementary to the base sequence of mRNA of Tet 3 gene is the physiological condition of Tet 3 producing cells (eg, synovial fibroblasts, synovial surface cells) in the mammal to be administered.
  • the base sequence complementary or substantially complementary to the base sequence of mRNA of the Tet 3 gene includes the following (k) or (l): (K) a base sequence complementary or substantially complementary to the base sequence represented by SEQ ID NO: 1; (L) a base sequence that hybridizes with a complementary strand sequence of the base sequence represented by SEQ ID NO: 1 under stringent conditions, and has a demethylation activity of 5-methylcytosine (5mC) or synovial fiber
  • 5mC 5-methylcytosine
  • synovial fiber A base sequence complementary to or substantially complementary to a sequence encoding a protein having an activity of promoting blast infiltration; Is mentioned.
  • the stringent conditions are as described above.
  • Tet 3 gene mRNA examples include human Tet 3 mRNA comprising the nucleotide sequence represented by SEQ ID NO: 1 (Genbank Accession No. NM_001287491), or their orthologs in other mammals, Examples include splice variants, allelic variants, and polymorphic variants.
  • the nucleotide sequence of the Tet 3 gene mRNA and the “part of the complementary or substantially complementary nucleotide sequence” are capable of specifically binding to the Tet 3 gene mRNA and of the protein from the mRNA.
  • the length and position are not particularly limited as long as they can inhibit translation (or degrade the mRNA). However, from the viewpoint of sequence specificity, at least a portion complementary or substantially complementary to the target sequence is required.
  • the nucleic acid containing any one of the following (1) to (3) is preferable as a nucleic acid containing a base sequence complementary to or substantially complementary to the base sequence of mRNA of the Tet 3 gene or a part thereof. Illustrated: (1) an antisense nucleic acid against mRNA of the Tet 3 gene, (2) a ribozyme nucleic acid against mRNA of the Tet 3 gene, (3) A nucleic acid having RNAi activity against Tet 3 gene mRNA or a precursor thereof.
  • Antisense nucleic acid against Tet 3 gene mRNA is a nucleic acid comprising a base sequence complementary to or substantially complementary to the base sequence of the mRNA or a part thereof, which is specific to the target mRNA and It has a function of suppressing protein synthesis by forming a stable double chain and binding.
  • Antisense nucleic acids are polydeoxyribonucleotides containing 2-deoxy-D-ribose, polyribonucleotides containing D-ribose, other types of polynucleotides that are N-glycosides of purine or pyrimidine bases, Other polymers with non-nucleotide backbones (eg, commercially available protein nucleic acids and synthetic sequence specific nucleic acid polymers) or other polymers containing special linkages, provided that the polymer is a base as found in DNA or RNA And a nucleotide having a configuration that allows attachment of a base).
  • RNA double-stranded DNA, single-stranded DNA, double-stranded RNA, single-stranded RNA, DNA: RNA hybrids, unmodified polynucleotides (or unmodified oligonucleotides), known modifications Additions, such as those with labels known in the art, capped, methylated, one or more natural nucleotides replaced with analogs, intramolecular nucleotide modifications Such as those having uncharged bonds (eg methylphosphonates, phosphotriesters, phosphoramidates, carbamates, etc.), charged bonds or sulfur-containing bonds (eg phosphorothioates, phosphorodithioates, etc.) Things such as proteins (eg, nucleases, nuclease inhibitors, toxins, antibodies, Null peptide, poly-L-lysine, etc.) and sugars (eg, monosaccharides), etc., side chain groups, intercurrent compounds (eg, acridine, ps
  • nucleoside may include not only purine and pyrimidine bases but also those having other modified heterocyclic bases. Such modifications may include methylated purines and pyrimidines, acylated purines and pyrimidines, or other heterocycles. Modified nucleosides and modified nucleotides may also be modified at the sugar moiety, for example, one or more hydroxyl groups are replaced by halogens, aliphatic groups, etc., or functional groups such as ethers, amines, etc. It may be converted. As described above, the antisense nucleic acid may be DNA or RNA, or may be a DNA / RNA chimera.
  • the RNA DNA hybrid formed by the target RNA and the antisense DNA can be recognized by endogenous RNase H and cause selective degradation of the target RNA. Therefore, in the case of antisense DNA directed to degradation by RNase H, the target sequence may be not only the sequence in mRNA but also the sequence of the intron region in the initial translation product of the Tet3 gene.
  • the intron sequence can be determined by comparing the genomic sequence and the cDNA base sequence of the Tet3 gene using a homology search program such as BLAST or FASTA.
  • the target region of the antisense nucleic acid of the present invention is not particularly limited in length as long as the antisense nucleic acid hybridizes, and as a result, the translation into the protein: Tet 3 is inhibited. May be the entire sequence or a partial sequence of mRNA that encodes, and may include a short sequence of about 10 bases and a long sequence of mRNA or the initial transcript. In view of easiness of synthesis, antigenicity, intracellular migration, etc., an oligonucleotide consisting of about 10 to about 40 bases, particularly about 15 to about 30 bases is preferred, but is not limited thereto.
  • a 3 ′ end palindromic region or a 3 ′ end hairpin loop or the like may be selected as a preferred target region of an antisense nucleic acid, but is not limited thereto.
  • the antisense nucleic acid of the present invention not only hybridizes with Tet 3 gene mRNA and initial transcription products to inhibit translation into proteins, but also binds to these genes that are double-stranded DNA to form triplex. It may be a (triplex) -forming (antigene) that can inhibit transcription to RNA.
  • the nucleotide molecules constituting the antisense nucleic acid may be natural DNA or RNA, but various chemicals may be used to improve stability (chemical and / or enzyme) and specific activity (affinity with RNA). Modifications can be included.
  • the phosphate residue (phosphate) of each nucleotide constituting the antisense nucleic acid is chemically modified, for example, phosphorothioate (PS), methylphosphonate, phosphorodithionate, etc. It can be substituted with a phosphate residue.
  • PS phosphorothioate
  • methylphosphonate methylphosphonate
  • phosphorodithionate etc. It can be substituted with a phosphate residue.
  • the 2′-position hydroxyl group of the sugar (ribose) of each nucleotide is changed to —OR (R ⁇ CH 3 (2'-O-Me), CH 2 CH 2 OCH 3 (2'-O-MOE), CH 2 CH 2 NHC (NH) NH 2 , CH 2 CONHCH 3 , CH 2 CH 2 CN, etc.) may be substituted.
  • the base moiety pyrimidine, purine
  • RNA The conformation of the sugar part of RNA is dominated by C2′-endo (S type) and C3′-endo (N type).
  • S type C2′-endo
  • N type C3′-endo
  • BNA BNA
  • Imanishi RNA derivative in which the conformation of the sugar moiety is fixed to the N-type by cross-linking 2 ′ oxygen and 4 ′ carbon
  • the antisense oligonucleotide of the present invention determines the target sequence of mRNA or initial transcript based on the cDNA sequence or genomic DNA sequence of the Tet 3 gene, and is a commercially available DNA / RNA automatic synthesizer (Applied Biosystems, Beckman). Etc.) can be prepared by synthesizing a complementary sequence thereto.
  • any of the above-described antisense nucleic acids containing various modifications can be chemically synthesized by a method known per se.
  • Ribozyme nucleic acid against Tet 3 gene mRNA As another preferred example of a nucleic acid comprising a base sequence complementary to or substantially complementary to the base sequence of mRNA of the Tet 3 gene or a part thereof, a ribozyme capable of specifically cleaving the mRNA within the coding region A nucleic acid is mentioned. “Ribozyme” refers to RNA having an enzyme activity that cleaves nucleic acids in a narrow sense, but in this specification, it is used as a concept including DNA as long as it has sequence-specific nucleic acid cleaving activity.
  • the most versatile ribozyme nucleic acid is self-splicing RNA found in infectious RNA such as viroid and virusoid, and hammerhead type and hairpin type are known.
  • the hammerhead type exhibits enzyme activity at about 40 bases, and a few bases at both ends adjacent to the part having the hammerhead structure (about 10 bases in total) are made into a sequence complementary to the desired cleavage site of mRNA. By doing so, it is possible to specifically cleave only the target mRNA.
  • This type of ribozyme nucleic acid has the additional advantage of not attacking genomic DNA because it uses only RNA as a substrate.
  • the target sequence is made single-stranded by using a hybrid ribozyme linked with an RNA motif derived from a viral nucleic acid that can specifically bind to an RNA helicase.
  • a hybrid ribozyme linked with an RNA motif derived from a viral nucleic acid that can specifically bind to an RNA helicase [Proc. Natl. Acad. Sci. USA, 98 (10): 5572-5577 (2001)].
  • the ribozyme is used in the form of an expression vector containing the DNA encoding the ribozyme, in order to promote the transfer of the transcription product to the cytoplasm, the ribozyme should be a hybrid ribozyme further linked with a tRNA-modified sequence.
  • siRNA against Tet 3 gene mRNA a double-stranded RNA composed of an oligo RNA complementary to Tet 3 gene mRNA and its complementary strand, so-called siRNA, is also complementary or substantially complementary to the base sequence of Tet 3 gene mRNA. Defined as being encompassed by a nucleic acid comprising a basic nucleotide sequence or a portion thereof.
  • RNAi RNA interference
  • siRNA can be obtained, for example, from Elbashir et al. (Genes Dev., 15, 188-200 (2001)), Teramoto et al. (FEBS Lett. 579 (13): p2878-82 (2005)). Can be designed according to the rules proposed by The target sequence of siRNA has a length of 15 to 50 bases, preferably 19 to 49 bases, more preferably 19 to 27 bases in principle.
  • the nucleic acid of the present invention may have an additional base at the 5 ′ or 3 ′ end.
  • the length of the additional base is usually about 2 to 4 bases, and the total length of siRNA is 19 bases or more.
  • the additional base may be DNA or RNA, but the use of DNA may improve the stability of the nucleic acid.
  • siRNA may have an overhang
  • the siRNA may have a different number of bases in the sense strand and the antisense strand, for example, “aiRNA” in which the antisense strand has a protruding portion sequence (overhang) at the 3 ′ end and the 5 ′ end.
  • a typical aiRNA has an antisense strand consisting of 21 bases, a sense strand consisting of 15 bases, and has an overhang structure of 3 bases at each end of the antisense strand. (Sun, X. et al., Nature Biotechnology Vol. 26 No. 12 p1379, International Publication No. WO2009 / 029688 pamphlet).
  • the position of the target sequence is not particularly limited, but it is desirable to select the target sequence from the 5′-UTR and the start codon to about 50 bases and from a region other than the 3′-UTR.
  • BLAST http: //www.ncbi.nlm Investigate using homology search software such as .nih.gov / BLAST /
  • a sense strand having a 3 'end overhang of TT or UU at 19-21 bases after AA (or NA), a sequence complementary to the 19-21 bases and TT or A double-stranded RNA consisting of an antisense strand having a 3 ′ terminal overhang of UU may be designed as siRNA.
  • siRNA short hairpin RNA
  • an arbitrary linker sequence for example, about 5-25 bases
  • the sense strand and the antisense strand are combined with each other. It can be designed by linking via a linker sequence.
  • siRNA Target Finders provided by Ambion (Http://www.ambion.com/jp/techlib/misc/siRNA_finder.html) and pSilencer® Expression Vector insert design tool (Http://www.ambion.com/jp/techlib/misc/psilencer_converter.html)
  • RNAi Codex GeneSee provided by RNAi Codex (Http://codex.cshl.edu/scripts/newsearchhairpin.cgi), but is not limited thereto.
  • the ribonucleoside molecule constituting the siRNA may also be modified in the same manner as in the above-described antisense nucleic acid in order to improve stability, specific activity and the like.
  • siRNA if all ribonucleoside molecules in natural RNA are replaced with a modified form, RNAi activity may be lost, and therefore it is necessary to introduce a minimum modified nucleoside that allows the RISC complex to function. .
  • natural DNA, stability (chemical and / or enzyme) and specific activity (affinity with RNA) as the modification.
  • RNAs can be substituted with various chemically modified RNAs (see Usman and Cedergren, 1992, TIBS 17, 34; Usman et al., 1994, Nucleic Acids Sym. Ser. 31, 163).
  • a phosphate residue (phosphate) of each nucleotide constituting siRNA is changed to a chemically modified phosphate such as phosphorothioate (PS), methylphosphonate, phosphorodithionate, etc. It can be substituted with a residue.
  • PS phosphorothioate
  • PS methylphosphonate
  • phosphorodithionate etc. It can be substituted with a residue.
  • the 2′-position hydroxyl group of the sugar (ribose) of each nucleotide is changed to —OR (R ⁇ CH 3 (2'-O-Me), CH 2 CH 2 OCH 3 (2'-O-MOE), CH 2 CH 2 NHC (NH) NH 2 , CH 2 CONHCH 3 , CH 2 CH 2 CN, etc.) and a fluorine atom (-F) may be substituted.
  • the base moiety pyrimidine, purine
  • the method for modifying an antisense nucleic acid described in (1) above can be used. Or you may give the chemical modification (2'-deoxylation, 2'-H) which substitutes a part of RNA in siRNA with DNA.
  • the 2′-position and 4′-position of sugar (ribose) are —O—CH 2
  • An artificial nucleic acid (LNA: Locked Nucleic Acid) in which the conformation is fixed to N-type by crosslinking with ⁇ may be used.
  • the sense strand and antisense strand constituting siRNA are linked via a linker to a ligand, peptide, sugar chain, antibody, lipid, positive charge, or molecular structure that specifically recognizes a receptor present on the cell surface.
  • the siRNA is synthesized by synthesizing the sense strand and antisense strand of the target sequence on the mRNA with a DNA / RNA automatic synthesizer and denatured in an appropriate annealing buffer at about 90 to about 95 ° C. for about 1 minute, It can be prepared by annealing at about 30 to about 70 ° C. for about 1 to about 8 hours. It can also be prepared by synthesizing a short hairpin RNA (shRNA) serving as a precursor of siRNA and cleaving it with a dicer.
  • shRNA short hairpin RNA
  • a nucleic acid designed to generate siRNA against Tet3 gene mRNA in vivo also includes a nucleotide sequence complementary or substantially complementary to the nucleotide sequence of Tet3 gene mRNA or a nucleotide sequence thereof. Defined as encompassed by a portion of nucleic acid. Examples of such a nucleic acid include an expression vector constructed so as to express the above-mentioned shRNA and siRNA.
  • the shRNA is an oligo comprising a base sequence in which a sense sequence and an antisense strand of a target sequence on mRNA are inserted by interposing a spacer sequence having a length (for example, about 5 to 25 bases) capable of forming an appropriate loop structure.
  • RNA can be prepared by designing RNA and synthesizing it with an automatic DNA / RNA synthesizer.
  • RNA synthesizer There are tandem type and stem loop (hairpin) type in vectors expressing shRNA.
  • siRNA sense strand expression cassette and antisense strand expression cassette are connected in tandem, and each strand is expressed and annealed in the cell to form double stranded siRNA (dsRNA).
  • dsRNA double stranded siRNA
  • shRNA expression cassette is inserted into a vector, in which shRNA is expressed in cells and processed by dicer to form dsRNA.
  • a pol II promoter for example, a CMV immediate early promoter
  • a pol III promoter is generally used in order to cause transcription of a short RNA accurately.
  • the polIII promoter include mouse and human U6-snRNA promoter, human H1-RNase P RNA promoter, human valine-tRNA promoter, and the like.
  • a sequence in which 4 or more Ts are continuous is used as a transcription termination signal.
  • the siRNA or shRNA expression cassette thus constructed is then inserted into a plasmid vector or viral vector.
  • virus vectors such as retrovirus, lentivirus, adenovirus, adeno-associated virus, herpes virus, Sendai virus, animal cell expression plasmids and the like are used.
  • the siRNA is based on nucleotide sequence information, for example, 394 Applied Biosystems, Inc. It can be chemically synthesized according to a conventional method using an automatic DNA / RNA synthesizer such as a synthesizer. For example, Caruthers et al. , 1992, Methods in Enzymology 211, 3-19, Thompson et al. , International Publication No. 99/54459, Wincott et al. , 1995, Nucleic Acids Res.
  • nucleic acid protecting group for example, dimethoxytrityl group at the 5 ′ end
  • a coupling group for example, phosphoramidite at the 3 ′ end
  • RNA for example, 2′-O-methyl nucleotide, 2′-deoxy-2′-fluoronucleotide
  • a borage reagent 3H-1,2-benzodithiol-3-one 1,1-dioxide
  • oligonucleotides may be synthesized separately and joined together after synthesis, for example by ligation (Moore et al., 1992, Science 256, 9923; Draper et al.
  • siRNA molecules can also be synthesized by tandem synthesis. That is, both siRNA strands are synthesized as a single continuous oligonucleotide separated by a cleavable linker, which is then cleaved to generate separate siRNA fragments that are hybridized and purified. .
  • the linker may be a polynucleotide linker or a non-nucleotide linker.
  • the synthesized siRNA molecules can be purified using methods known to those skilled in the art. For example, a method of purification by gel electrophoresis or a method of purification using high performance liquid chromatography (HPLC) can be mentioned.
  • the oligo RNA complementary to the Tet 3 gene mRNA that constitutes the siRNA against the Tet 3 gene and its complementary strand include a sequence comprising SEQ ID NO: 3 and a sequence comprising SEQ ID NO: 4, respectively.
  • the sequence consisting of SEQ ID NO: 3 and the sequence consisting of SEQ ID NO: 4 are exemplified.
  • a nucleic acid containing a base sequence complementary to or substantially complementary to the base sequence of mRNA of the Tet 3 gene or a part thereof is micro RNA (miRNA) targeting the mRNA.
  • miRNA miRNA targeting human Tet 3 mRNA
  • human miRNA targeting human Tet 3 mRNA include hsa-miR-22, hsa-miR-26a, hsa-miR-301a, hsa-miR-301b, hsa-miR-372, hsa-miR-29c. Hsa-miR-29a, hsa-miR-29b, hsa-miR-374b, hsa-miR-374a, and the like.
  • miRNA can also be prepared according to the method described for siRNA.
  • a nucleic acid containing a nucleotide sequence complementary to or substantially complementary to the nucleotide sequence of Tet 3 gene mRNA or a part thereof is provided in a special form such as a liposome or a microsphere, or other molecules are added.
  • Can be provided in different forms. Examples of additional forms that can be used include polycationic substances such as polylysine that act to neutralize the charge of the phosphate group skeleton, and lipids that enhance interaction with cell membranes and increase nucleic acid uptake. Hydrophobic substances such as (eg, phospholipid, cholesterol, etc.) can be mentioned.
  • Preferred lipids for addition include cholesterol and derivatives thereof (eg, cholesteryl chloroformate, cholic acid, etc.). Such can be attached to the 3 'or 5' end of the nucleic acid and can be attached via a base, sugar, intramolecular nucleoside bond.
  • the other group include a cap group specifically arranged at the 3 ′ end or 5 ′ end of a nucleic acid, which prevents degradation by a nuclease such as exonuclease or RNase.
  • capping groups include, but are not limited to, hydroxyl protecting groups known in the art, including glycols such as polyethylene glycol and tetraethylene glycol.
  • Tet 3 expression inhibitory activity of these nucleic acids can be determined by transforming a nucleic acid encoding Tet 3 into a Tet 3 gene expression system in vivo or in vitro, or a translation system for Tet 3 protein in vivo or in vitro. Can be used to investigate.
  • the substance that inhibits the expression of Tet3 in the present invention is not limited to a nucleic acid containing a base sequence complementary to or substantially complementary to the base sequence of mRNA of Tet3 gene as described above, or a part thereof. Other substances such as small molecule compounds may be used as long as production is directly or indirectly inhibited. Such a substance can be obtained, for example, by the screening method of the present invention described later.
  • a substance that inhibits the expression of Tet 3 exhibits the activity of suppressing the demethylation activity of 5-methylcytosine (5mC) possessed by Tet 3 or the activity of promoting the invasion of synovial fibroblasts. Useful for treatment. Therefore, a medicament containing a substance that inhibits the expression of Tet 3 can be used as an agent for preventing and / or treating arthritis.
  • Medicament containing antisense nucleic acid, ribozyme nucleic acid, siRNA and precursor thereof An antisense nucleic acid of the present invention that can complementarily bind to a transcription product of the Tet 3 gene and suppress protein translation from the transcription product, or a homology (or complementary) to the transcription product (mRNA) of the Tet 3 gene.
  • SiRNA (or ribozyme) having a nucleotide sequence and capable of cleaving the transcription product as a target
  • shRNA that is a precursor of the siRNA (hereinafter, comprehensively referred to as “the nucleic acid of the present invention”) May suppress the expression of Tet3 in vivo and suppress the activity of promoting the demethylation of 5-methylcytosine (5mC) or the infiltration of synovial fibroblasts, and thus preventing arthritis and / or It can be used as a therapeutic agent.
  • 5mC 5-methylcytosine
  • the medicament containing the nucleic acid of the present invention has low toxicity and can be used as a liquid or as a pharmaceutical composition of an appropriate dosage form as a human or non-human mammal (eg, rat, rabbit, sheep, pig, cow, cat, It can be administered orally or parenterally (eg, intravascular administration, subcutaneous administration, etc.) to dogs, monkeys, etc.
  • a human or non-human mammal eg, rat, rabbit, sheep, pig, cow, cat
  • parenterally eg, intravascular administration, subcutaneous administration, etc.
  • the nucleic acid of the present invention is inserted alone or in a functional manner into an appropriate expression vector for mammalian cells such as a retrovirus vector, adenovirus vector, adenovirus associated virus vector, etc., and then formulated according to conventional means. can do.
  • the nucleic acid can be administered as it is or together with an auxiliary agent for promoting intake by a catheter such as a gene gun or a hydrogel catheter. Alternatively, it can be aerosolized and locally administered into the trachea as an inhalant.
  • the nucleic acid may be formulated (injection) alone or with a carrier such as liposome and administered intravenously, subcutaneously, etc. .
  • the nucleic acids of the invention may be administered per se or as a suitable pharmaceutical composition.
  • the pharmaceutical composition used for administration may contain the nucleic acid of the present invention and a pharmacologically acceptable carrier, diluent or excipient.
  • Such pharmaceutical compositions are provided as dosage forms suitable for oral or parenteral administration.
  • a composition for parenteral administration for example, injections, suppositories and the like are used.
  • Injections are dosage forms such as intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections, infusions, and the like. May be included.
  • Such an injection can be prepared according to a known method.
  • As a method for preparing an injection it can be prepared, for example, by dissolving, suspending or emulsifying the nucleic acid of the present invention in a sterile aqueous liquid or oily liquid usually used for injection.
  • an aqueous solution for injection for example, an isotonic solution containing physiological saline, glucose and other adjuvants, and the like are used, and suitable solubilizers such as alcohol (eg, ethanol), polyalcohol (eg, Propylene glycol, polyethylene glycol), nonionic surfactants (eg, polysorbate 80, HCO-50 (polyoxyethylene (50 mol) additive of hydrogenated castor oil)), and the like.
  • suitable solubilizers such as alcohol (eg, ethanol), polyalcohol (eg, Propylene glycol, polyethylene glycol), nonionic surfactants (eg, polysorbate 80, HCO-50 (polyoxyethylene (50 mol) additive of hydrogenated castor oil)), and the like.
  • the oily liquid for example, sesame oil, soybean oil and the like are used, and benzyl benzoate, benzyl alcohol and the like may be used in combination as a solubilizing agent.
  • the prepared injection solution is
  • compositions for oral administration include solid or liquid dosage forms, specifically tablets (including dragees and film-coated tablets), pills, granules, powders, capsules (including soft capsules), syrups Agents, emulsions, suspensions and the like.
  • Such a composition is produced by a known method and may contain a carrier, a diluent or an excipient usually used in the pharmaceutical field.
  • a carrier and excipient for tablets for example, lactose, starch, sucrose, and magnesium stearate are used.
  • the above parenteral or oral pharmaceutical compositions are conveniently prepared in dosage unit form to suit the dosage of the active ingredient.
  • Examples of the dosage form of such a dosage unit include tablets, pills, capsules, injections (ampoules), and suppositories.
  • the nucleic acid of the present invention is preferably contained, for example, usually 5 to 500 mg per dosage unit form, especially 5 to 100 mg for injections and 10 to 250 mg for other dosage forms.
  • the dose of the above-mentioned medicament containing the nucleic acid of the present invention varies depending on the administration subject, target disease, symptom, administration route, etc., but for example, when used for the treatment / prevention of rheumatoid arthritis,
  • the amount of nucleic acid is usually about 0.01 to 20 mg / kg body weight, preferably about 0.1 to 10 mg / kg body weight, more preferably about 0.1 to 5 mg / kg body weight 1 to 5 times a day. It is convenient to administer by intravenous injection to a degree, preferably about 1 to 3 times a day. In the case of other parenteral administration and oral administration, an equivalent amount can be administered. If symptoms are particularly severe, the dose may be increased according to the symptoms.
  • compositions may appropriately contain other active ingredients as long as an undesirable interaction is not caused by blending with the nucleic acid of the present invention.
  • the pharmaceutical composition containing the above-mentioned antisense nucleic acid, ribozyme nucleic acid, siRNA and its precursor to Tet 3, or a low-molecular compound that suppresses the expression of Tet 3, etc. is used for the treatment, prevention, or progression of arthritis.
  • arthritis include rheumatoid arthritis, psoriatic arthritis, spondyloarthritis (eg, ankylosing spondylitis), and preferably rheumatoid arthritis.
  • any disease in which Tet 3 is involved in the deterioration of the disease state is included in the target disease of the present invention.
  • the pharmaceutical composition containing the above-mentioned antisense nucleic acid, ribozyme nucleic acid, siRNA and its precursor against Tet 3 or a low molecular weight compound that suppresses the expression of Tet 3 is used for the treatment or prevention of arthritis May be used alone or in combination with one or more drugs having anti-inflammatory activity.
  • the drug used in combination is not particularly limited.
  • mesalazine corticosteroids (eg, betamethasone, prednisolone, hydrocortisone, dexamethasone, etc.), nonsteroidal anti-inflammatory drugs (NSAIDs; eg, salicylic acid, anthranilic acid) , Aryl acids, propionates, oxicams, pilins), anti-TNF ⁇ antibodies (influximab, adalimumab), anti-rheumatic drugs (eg, immunomodulators such as actarit, immunosuppressants such as methotrexate, anti-TNF ⁇ antibodies And biological preparations such as etanercept).
  • NSAIDs nonsteroidal anti-inflammatory drugs
  • NSAIDs eg, salicylic acid, anthranilic acid
  • Aryl acids propionates
  • oxicams oxicams
  • pilins anti-TNF ⁇ antibodies
  • anti-rheumatic drugs eg, immunomodulators such as actarit, immunos
  • Tet 3 can be used as a tool for screening for preventive and / or therapeutic drugs for arthritis by using the expression level and / or function of Tet 3 (or Tet 3 gene) as an index. it can.
  • the screening method involves culturing cells capable of producing Tet 3 in the presence and absence of a test substance, 3 comparing the expression level or degree of function.
  • a compound that inhibits the function of Tet 3 can also be screened by testing its ability to bind to purified Tet 3 protein.
  • Cells having the ability to produce Tet 3 used in the above screening methods include human or other mammalian cells (eg, synovial fibroblasts, synovial surface cells, etc.) that naturally express them, or those If it is a biological sample containing (for example: synovium (especially synovial surface cell layer) etc.) etc., there will be no restriction
  • synovial membranes derived from non-human animals they may be isolated from the living body and cultured, or the test substance may be administered to the living body itself, and the biological sample may be isolated after a certain period of time. .
  • various transformants prepared by known and commonly used genetic engineering techniques can also be used as the cells having the ability to produce Tet 3.
  • animal cells such as H4IIE-C3 cells, HepG2 cells, HEK293 cells, COS7 cells and CHO cells are preferably used. Specifically, it hybridizes under stringent conditions with a DNA encoding Tet3 (that is, the base sequence represented by SEQ ID NO: 1 or a base sequence complementary to the base sequence, and SEQ ID NO: A DNA comprising a base sequence encoding a polypeptide having the same function as that of the protein consisting of the amino acid sequence represented by 2) is introduced downstream into a promoter in an appropriate expression vector and introduced into a host animal cell. Can be prepared. A method for preparing a gene encoding Tet 3 will be described below.
  • the gene encoding Tet 3 can be obtained by a conventional genetic engineering method (for example, Sambrook J., Frisch EF, Maniatis T., Molecular Cloning 2nd edition), published by Cold Spring Harbor Laboratory ( The method can be obtained according to the method described in Cold Spring Harbor Laboratory Press). That is, DNA encoding Tet 3 is derived from, for example, a cell or tissue that produces Tet 3 described above by synthesizing an appropriate oligonucleotide as a probe or primer based on the nucleotide sequence represented by SEQ ID NO: 1. Cloning can be performed from cDNA or cDNA library using hybridization method or PCR method.
  • Hybridization can be performed, for example, according to the method described in Molecular Cloning 2nd edition (above). When a commercially available library is used, hybridization can be performed according to the method described in the instruction manual attached to the library.
  • the nucleotide sequence of DNA can be determined using a known kit such as Mutan. TM -Super Express Km (Takara Shuzo Co., Ltd.), Mutan TM -K (Takara Shuzo Co., Ltd.) and the like can be converted according to a method known per se such as the ODA-LA PCR method, the Gapped duplex method, the Kunkel method, or the like.
  • the cloned DNA can be used as it is or after digestion with a restriction enzyme or addition of a linker, if desired.
  • the DNA may have ATG as a translation initiation codon on the 5 ′ end side, and may have TAA, TGA, or TAG as a translation termination codon on the 3 ′ end side. These translation initiation codon and translation termination codon can be added using an appropriate synthetic DNA adapter.
  • Tet3 gene cells expressing the Tet3 protein can be produced and obtained according to a normal genetic engineering method. For example, a plasmid is prepared so that the Tet 3 gene can be expressed in the host cell, this is introduced into the host cell, transformed, and the transformed host cell (transformant) is cultured.
  • Examples of the plasmid include a promoter that can replicate autonomously in a host cell, can replicate autonomously, can be easily isolated and purified from the host cell, and can function in the host cell.
  • Preferred examples include those in which a gene encoding Tet 3 is introduced into an expression vector having a detectable marker.
  • Various types of expression vectors are commercially available.
  • an expression vector used for expression in E. coli is an expression vector containing a promoter such as lac, trp, tac, etc., and these are commercially available from Pharmacia, Takara Bio and the like.
  • Restriction enzymes used for introducing a gene encoding Tet 3 into the expression vector are also commercially available from Takara Bio and others. If it is necessary to direct further expression, a ribosome binding region may be linked upstream of the DNA encoding Tet3. Examples of the ribosome binding region used include Guarente L. et al. (Cell 20, p543) and Taniguchi et al. (Genetics of Industrial Microorganisms, p202, Kodansha).
  • animal cell expression plasmids eg, pA1-11, pXT1, pRc / CMV, pRc / RSV, pcDNAI / Neo
  • bacteriophages such as ⁇ phage
  • animal virus vectors such as retrovirus, vaccinia virus, adenovirus, etc. It can also be used.
  • the promoter may be any promoter as long as it is appropriate for the host used for gene expression.
  • SR ⁇ promoter for example, SR ⁇ promoter, SV40 promoter, LTR promoter, CMV (cytomegalovirus) promoter, RSV (rous sarcoma virus) promoter, MoMuLV (Moloney murine leukemia virus) LTR, HSV-TK (herpes simplex virus thymidine kinase) promoter, ⁇ actin A gene promoter, aP2 gene promoter, etc. are used.
  • EF- ⁇ promoter, CAG promoter, CMV promoter, SR ⁇ promoter and the like are preferable.
  • an expression vector containing an enhancer, a splicing signal, a poly A addition signal, a selection marker, an SV40 origin of replication (hereinafter sometimes abbreviated as SV40ori) and the like is used as desired.
  • the selectable marker include a dihydrofolate reductase gene (hereinafter abbreviated as dhfr, methotrexate (MTX) resistance), an ampicillin resistance gene (hereinafter amp).
  • dhfr dihydrofolate reductase gene
  • MTX methotrexate
  • amp ampicillin resistance gene
  • r A neomycin resistance gene hereinafter, neo
  • the target gene can be selected using a medium not containing thymidine.
  • a Tet 3 expressing cell can be produced by transforming a host with an expression vector containing the DNA encoding Tet 3 described above. Examples of host cells include prokaryotic or eukaryotic microbial cells, insect cells, or mammalian cells.
  • mammalian cells examples include HepG2 cells, HEK293 cells, HeLa cells, human FL cells, monkey COS-7 cells, monkey Vero cells, Chinese hamster ovary cells (hereinafter abbreviated as CHO cells), dhfr gene-deficient CHO cells ( Hereinafter, CHO (dhfr ⁇ Abbreviated as cells), mouse L cells, mouse AtT-20 cells, mouse myeloma cells, rat H4IIE-C3 cells, rat GH3 cells, and the like.
  • the plasmid obtained as described above can be introduced into the host cell by an ordinary genetic engineering method.
  • the transformant can be cultured by a conventional method used for culturing microorganisms, insect cells or mammalian cells.
  • culturing is performed in a medium appropriately containing an appropriate carbon source, nitrogen source, and micronutrients such as vitamins.
  • the culture method may be any of solid culture and liquid culture, and preferred examples include liquid culture such as aeration and agitation culture. Transformation can be performed by calcium phosphate coprecipitation method, PEG method, electroporation method, microinjection method, lipofection method and the like.
  • the method described in Cell Engineering Supplement 8 New Cell Engineering Experiment Protocol, 263-267 (1995) (published by Shujunsha), Virology, Volume 52, 456 (1973) can be used.
  • a transformed cell obtained as described above, a mammalian cell having an ability to produce Tet 3 or a tissue / organ containing the cell is, for example, a minimum essential medium containing about 5 to 20% fetal calf serum ( (MEM) [Science, 122, 501 (1952)], Dulbecco's modified Eagle medium (DMEM) [Virology, 8, 396 (1959)], RPMI 1640 medium [The Journal of the American Medical Association, 199 (199) 1967)] and 199 medium [Proceeding of the Society for the Biological Medicine, Vol. 73, 1 (1950)].
  • the pH of the medium is preferably about 6-8. Culture is usually performed at about 30 to 40 ° C., and aeration and agitation are added as necessary.
  • the Tet 3 protein may be obtained by combining methods commonly used for general protein isolation / purification.
  • the transformant obtained by the above culture may be removed by centrifugation or the like, and Tet3 may be purified from the culture supernatant in the same manner as described above.
  • Tet3 protein may be solubilized and purified by singly or in combination with various chromatographic processes such as ion exchange, hydrophobicity, and gel filtration. An operation of restoring the higher order structure of the purified protein may be further performed.
  • test substance examples include proteins, peptides, non-peptide compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and the like. These substances may be novel or may be known ones.
  • a control cell that is not brought into contact with the test substance can also be used as a comparative control.
  • “do not contact the test substance” means that the same amount of solvent (blank) as the test substance is added instead of the test substance, the expression level of Tet 3 or Tet 3 gene, or the function of Tet 3
  • a negative control substance that does not affect the above is also included.
  • the contact of the test substance with the cells is, for example, the above-mentioned medium or various buffers (for example, HEPES buffer, phosphate buffer, phosphate buffered saline, Tris-HCl buffer, borate buffer, acetic acid).
  • the test substance can be added to a buffer solution or the like, and the cells can be incubated for a certain time.
  • the concentration of the test substance to be added varies depending on the type of compound (solubility, toxicity, etc.), but is appropriately selected within the range of about 0.1 nM to about 100 ⁇ M, for example.
  • Examples of the incubation time include about 10 minutes to about 24 hours.
  • cells producing Tet 3 are provided in the form of a non-human mammal individual, the state of the animal individual is not particularly limited.
  • an arthritis model animal in which arthritis is induced by a drug or genetic modification for example, collagen RA model animals such as induced arthritis (CIA) mice, SKG mice, PD-1 knockout mice, K / BxN mice, Synoviolin Tg mice, etc.).
  • the administration route is not particularly limited, and examples thereof include intravenous administration, intraarterial administration, subcutaneous administration, intradermal administration, intraperitoneal administration, oral administration, intratracheal administration, and rectal administration.
  • the dose is not particularly limited. For example, a dose of about 0.5 to 20 mg / kg can be administered 1 to 5 times a day, preferably 1 to 3 times a day for 1 to 14 days. .
  • the above screening method may be performed by contacting a test substance with an extract of the cells or Tet 3 isolated and purified from the cells, instead of the cells having the ability to produce Tet 3. it can.
  • the present invention relates to a prophylactic and / or therapeutic agent for arthritis, characterized by comparing the expression of the protein (gene) in cells having the ability to produce Tet 3 in the presence and absence of a test substance.
  • a screening method is provided. The cells used in this method, the type of test substance, the mode of contact between the test substance and cells, etc. are the same as described above.
  • the expression level of Tet 3 is a nucleic acid that can hybridize with the above-described DNA encoding Tet 3 under stringent conditions, that is, the base sequence represented by SEQ ID NO: 1 or a complementary base sequence and stringent. Can be measured at the RNA level by detecting mRNA of the Tet 3 gene using a nucleic acid (DNA) that can hybridize under various conditions (hereinafter sometimes referred to as “the nucleic acid for detection of the present invention”). it can. Alternatively, the expression level can also be measured at the protein level by detecting these proteins using the above-described antibody against Tet 3 (hereinafter sometimes referred to as “the detection antibody of the present invention”). .
  • the present invention (A) Cells having the ability to produce Tet 3 are cultured in the presence and absence of a test substance, and the amount of mRNA encoding the protein under both conditions is determined using the nucleic acid for detection of the present invention.
  • a screening method for a prophylactic and / or therapeutic agent for arthritis is provided. That is, screening for a substance that changes the expression level of Tet 3 can be performed as follows.
  • test substance administered to a normal or disease model (eg, RA model animal) non-human mammal (eg, mouse, rat, rabbit, sheep, pig, cow, cat, dog, monkey, etc.) After a certain period of time (after 30 minutes to 3 days, preferably 1 hour to 2 days, more preferably 1 hour to 24 hours), blood or a specific organ (for example, synovial membrane), Alternatively, tissue or cells isolated from the organ are obtained.
  • Tet 3 mRNA can be quantified by extracting mRNA from cells or the like by a conventional method, or can be quantified by Northern blot analysis known per se.
  • Tet 3 protein can be quantified using Western blot analysis or various immunoassay methods described in detail below.
  • a test substance when a cell expressing Tet 3 gene for example, a synovial fibroblast, a synovial surface cell, or a transformant introduced with Tet3 is prepared according to the above method and cultured according to a conventional method Is added to the medium or buffer, and after incubation for a certain period of time (from 1 day to 7 days, preferably from 1 day to 3 days, more preferably from 2 days to 3 days), Tet3 contained in the cells or the code thereof is encoded.
  • MRNA can be quantified and analyzed in the same manner as (i) above.
  • Detection and quantification of the expression level of the Tet3 gene can be performed by a known method such as Northern blotting or RT-PCR using RNA prepared from the cells or a complementary polynucleotide transcribed therefrom. Specifically, by using a polynucleotide having at least 15 bases continuous in the base sequence of the Tet 3 gene and / or a complementary polynucleotide thereof as a primer or a probe, the presence or absence of expression of the Tet 3 gene in RNA, Its expression level can be detected and measured.
  • Such a probe or primer can be designed based on the base sequence of the Tet 3 gene, for example, using primer 3 (http://primer3.sourceforge.net/) or vector NTI (manufactured by Infomax). it can.
  • the primer or probe is a radioisotope ( 32 P, 33 P or the like: labeled with RI) or a fluorescent substance, and hybridized with cell-derived RNA transferred to a nylon membrane or the like according to a conventional method, and then the primer or probe (DNA or RNA) and RNA formed And a method of detecting and measuring a double strand with a radiation detector (BAS-1800II, manufactured by Fuji Film Co., Ltd.) or a fluorescence detector as a signal derived from the primer or probe label (RI or fluorescent substance) can do.
  • a radiation detector BAS-1800II, manufactured by Fuji Film Co., Ltd.
  • a fluorescence detector as a signal derived from the primer or probe label (RI or fluorescent substance) can do.
  • the probe is labeled according to the protocol, hybridized with cell-derived RNA, and then the signal derived from the probe label is multibiotic.
  • a method of detecting and measuring with a major STORM860 can also be used.
  • cDNA was prepared from cell-derived RNA according to a conventional method, and was prepared based on the Tet 3 gene sequence so that the target Tet 3 gene region could be amplified using this as a template.
  • a pair of primers (a normal strand that binds to the cDNA ( ⁇ strand) and a reverse strand that binds to the + strand) are hybridized with this, and PCR is performed according to a conventional method, and the resulting amplified double-stranded DNA is detected.
  • the method of doing can be illustrated.
  • the detection of the amplified double-stranded DNA was performed by a method for detecting the labeled double-stranded DNA produced by performing the PCR using a primer previously labeled with RI or a fluorescent substance.
  • a method may be used in which double-stranded DNA is transferred to a nylon membrane or the like according to a conventional method, and the labeled primer is used as a probe to hybridize with this to detect it.
  • the produced labeled double-stranded DNA product can be measured with an Agilent 2100 Bioanalyzer (manufactured by Yokogawa Analytical Systems).
  • an RT-PCR reaction solution is prepared according to the protocol using SYBR Green RT-PCR Reagents (manufactured by Applied Biosystems), and reacted with ABI PRIME 7900 Sequence Detection System (manufactured by Applied Biosystems).
  • the “transcriptional regulatory region” usually refers to a range of several kb to several tens of kb upstream of the chromosomal gene.
  • 5′-race method for example, 5
  • Ii a step of determining the 5 ′ end by a conventional method such as oligocap method, S1 primer mapping, etc.
  • a reporter protein expression vector is constructed by linking a nucleic acid encoding a reporter protein (hereinafter referred to as “reporter gene”) in a functional form downstream of the transcriptional regulatory region of the Tet 3 gene.
  • reporter gene a reporter protein
  • Reporter proteins include ⁇ -glucuronidase (GUS), luciferase, chloramphenicol transacetylase (CAT), ⁇ -galactosidase (GAS), green fluorescent protein (GFP), yellow fluorescent protein (YFP), blue fluorescent protein ( CFP), red fluorescent protein (RFP) and the like.
  • a reporter gene in which the transcriptional regulatory region of the prepared Tet 3 gene is operably linked is inserted into a vector that can be used in a cell into which the reporter gene is introduced, using a normal genetic engineering technique, and a plasmid Can be prepared and introduced into a suitable host cell.
  • Stable transformed cells can be obtained by culturing in a medium with selection conditions according to the selection marker gene mounted on the vector.
  • a reporter gene in which the transcriptional regulatory region of the Tet 3 gene is operably linked may be transiently expressed in the host cell.
  • a method for measuring the expression level of the reporter gene a method corresponding to each reporter gene may be used as a method for measuring the expression level of the reporter gene may be used.
  • luciferase gene when used as a reporter gene, the transformed cell is cultured for several days, an extract of the cell is obtained, and then the extract is reacted with luciferin and ATP to cause chemiluminescence, and the luminescence intensity Promoter activity can be detected by measuring.
  • a commercially available luciferase reaction detection kit such as Picker Gene Dual Kit (registered trademark; manufactured by Toyo Ink) can be used.
  • a method for measuring the protein amount of Tet 3 specifically, for example, (I)
  • the detection antibody of the present invention is reacted competitively with the sample solution and labeled Tet3, and the labeled protein bound to the antibody is detected to quantify Tet3 in the sample solution.
  • (Ii) The labeling agent on the insolubilized carrier after reacting the sample solution with the detection antibody of the present invention insolubilized on the carrier and another labeled detection antibody of the present invention simultaneously or continuously For example, a method of quantifying Tet 3 in a sample solution by measuring the amount (activity) of. Detection and quantification of the protein expression level of Tet 3 can be quantified according to a known method such as Western blotting using an antibody recognizing Tet3. Western blotting uses an antibody that recognizes Tet 3 as a primary antibody, and then uses a secondary antibody as a secondary antibody.
  • Labeled with an antibody that binds to a primary antibody labeled with a radioisotope such as I, a fluorescent substance, an enzyme such as horseradish peroxidase (HRP), etc., and signals derived from these labeled substances are measured with a radiation measuring instrument (BAI-1800II: It can be carried out by measuring with a fluorescence detector or the like.
  • a radiation measuring instrument BAI-1800II: It can be carried out by measuring with a fluorescence detector or the like.
  • detection is performed according to the protocol using an ECL Plus Western Blotting Detection System (Amersham Pharmacia Biotech), and a multi-biometric STORM860 (Amersham Pharmacia Biotech) Can also be measured.
  • the form of the antibody is not particularly limited, and may be a polyclonal antibody having Tet 3 as an immunogen or a monoclonal antibody, and further, at least a sequence of amino acid sequences constituting Tet 3 is continuous.
  • an antibody having antigen-binding property against a polypeptide consisting of 8 amino acids, preferably 15 amino acids, more preferably 20 amino acids can also be used.
  • Methods for producing these antibodies are already well known, and the antibodies of the present invention can also be produced according to these conventional methods (Current protocol in Molecular Biology edit. Ausubel et al. (1987) Publ. John Wiley and Sons. Section 11.12-11.13). In the above quantification method (ii), it is desirable that the two types of antibodies recognize different portions of Tet 3.
  • one antibody recognizes the N-terminal part of Tet 3, one that reacts with the C-terminal part of the protein can be used as the other antibody.
  • a labeling agent used in a measurement method using a labeling substance for example, a radioisotope, an enzyme, a fluorescent substance, a luminescent substance, or the like is used. Examples of radioisotopes include [ 125 I], [ 131 I], [ 3 H], [ 14 C] and the like are used.
  • the enzyme a stable enzyme having a large specific activity is preferable.
  • ⁇ -galactosidase ⁇ -glucosidase, alkaline phosphatase, peroxidase, malate dehydrogenase and the like are used.
  • fluorescent substance for example, fluorescamine, fluorescein isothiocyanate and the like are used.
  • luminescent substance for example, luminol, luminol derivatives, luciferin, lucigenin and the like are used.
  • a biotin- (strept) avidin system can also be used for binding of an antibody or antigen and a labeling agent.
  • the Tet 3 quantification method using the detection antibody of the present invention is not particularly limited, and the amount of antibody, antigen or antibody-antigen complex corresponding to the amount of antigen in the sample solution is chemically or physically determined. Any measurement method may be used as long as it is a measurement method that is detected by a standard means and calculated from a standard curve prepared using a standard solution containing a known amount of antigen. For example, nephrometry, competition method, immunometric method and sandwich method are preferably used. In view of sensitivity and specificity, for example, the sandwich method described later is preferably used. In the insolubilization of the antigen or antibody, physical adsorption may be used, or a chemical bond usually used for insolubilizing and immobilizing proteins or enzymes may be used.
  • the carrier examples include insoluble polysaccharides such as agarose, dextran, and cellulose, synthetic resins such as polystyrene, polyacrylamide, and silicon, or glass.
  • the sample solution is reacted with the insolubilized detection antibody of the present invention (primary reaction), and further labeled with another detection antibody of the present invention (secondary reaction), and then on the insolubilized carrier.
  • secondary reaction By measuring the amount or activity of the labeling agent, Tet 3 in the sample solution can be quantified.
  • the primary reaction and the secondary reaction may be performed in the reverse order, may be performed simultaneously, or may be performed at different times.
  • the labeling agent and the insolubilization method can be the same as those described above.
  • the antibody used for the immobilized antibody or the labeled antibody is not necessarily one type, and a mixture of two or more types of antibodies is used for the purpose of improving measurement sensitivity. May be.
  • the detection antibody of the present invention can also be used in measurement systems other than the sandwich method, such as a competitive method, an immunometric method, or nephrometry.
  • Tet 3 in a sample solution and labeled Tet 3 are reacted competitively with an antibody, and then an unreacted labeled antigen (F) and a labeled antigen (B) bound to the antibody are combined.
  • B / F separation Separation (B / F separation) and the amount of labeling of either B or F is measured to quantify Tet 3 in the sample solution.
  • a soluble antibody is used as an antibody
  • B / F separation is performed using polyethylene glycol or a secondary antibody against the antibody (primary antibody)
  • a solid phase is used as the primary antibody.
  • Either an antibody is used (direct method), or a primary antibody is soluble, and a solid phase antibody is used as a secondary antibody (indirect method).
  • Tet 3 in a sample solution and Tet 3 immobilized on a solid phase are competitively reacted with a certain amount of labeled antibody, and then the solid phase and the liquid phase are separated, or the Tet 3 in the sample solution is separated.
  • Tet 3 is reacted with an excess amount of labeled antibody, and then solid-phased Tet 3 is added to bind unreacted labeled antibody to the solid phase, and then the solid phase and the liquid phase are separated. Next, the amount of label in any phase is measured to quantify the amount of antigen in the sample solution. In nephrometry, the amount of insoluble precipitate produced as a result of the antigen-antibody reaction in a gel or solution is measured. Even when the amount of Tet 3 in the sample solution is very small and only a small amount of precipitate can be obtained, laser nephrometry using laser scattering is preferably used. In applying these individual immunological measurement methods to the quantification method of the present invention, special conditions, operations and the like are not required to be set.
  • the amount of Tet3 in a cell can be quantified with high sensitivity by using the detection antibody of the present invention.
  • the expression level of Tet 3 (mRNA level or protein level) in the presence of the test substance is about 20% or more, preferably about 30%, compared to the case in the absence of the test substance.
  • the test substance can be selected as a Tet 3 expression inhibitor, and thus as a candidate for a prophylactic and / or therapeutic drug for arthritis.
  • a cell containing a reporter gene under the control of the transcriptional regulatory region in the Tet3 gene can be used instead of the cell expressing the Tet3 gene.
  • Such cells may be cells, tissues, organs or individuals of transgenic animals into which a reporter gene (eg, luciferase, GFP, etc.) under the control of the transcriptional regulatory region of the Tet 3 gene has been introduced.
  • a reporter gene eg, luciferase, GFP, etc.
  • the expression level of Tet 3 can be evaluated by measuring the expression level of the reporter gene using a conventional method.
  • the screening method of the present invention can also be performed using as an index whether or not the test substance inhibits the function of Tet3. Since Tet 3 is a demethylated protein, it is considered that a substance capable of binding to Tet 3 protein can inhibit the function of Tet 3 by suppressing the demethylation activity. Therefore, a candidate for a Tet 3 function inhibitor can be screened using the binding ability to Tet 3 as an index. For example, a test substance is adsorbed to each well of a well plate, a Tet 3 solution labeled with an appropriate labeling agent is added to each well and incubated, the liquid phase is removed, and the amount of label bound to the solid phase after washing is determined.
  • a test substance having the ability to bind to Tet 3 can be detected.
  • Tet 3 it is also possible to detect Tet 3 bound to the solid phase using a labeled anti-Tet 3 antibody.
  • a test substance solution is passed through a carrier on which Tet 3 is immobilized (for example, an affinity column), and the test substance retained on the carrier is used as a substance capable of binding to Tet 3, ie, prevention of arthritis and It can also be selected as a candidate for a therapeutic agent.
  • Whether or not the candidate substance thus obtained actually has an anti-inflammatory action is confirmed by applying the candidate substance to an arthritis model and testing whether or not the inflammatory reaction in the model is suppressed. be able to.
  • in vivo and in vitro models can be used as such arthritis models.
  • in vivo models include CIA models (can be prepared by immunizing non-human animals with type II collagen emulsified with complete Freund's adjuvant), CAIA models (recognizing epitopes in CB11 of type II collagen) RA models such as those that can be prepared by injecting a monoclonal antibody cocktail into a non-human animal) can be used, but are not limited thereto.
  • examples of in vitro models include culture systems of target cells in arthritis (for example, synovial cells in RA) (for example, culture systems of synovial fibroblasts derived from synovial tissue of RA patients). However, it is not limited to these.
  • inflammatory cytokines such as TNF ⁇ as necessary, or complex cultures with inflammatory cytokine producing cells such as monocytes, macrophages, neutrophils (eg, transwells).
  • TM Culture using target culture cells eg, synovial fibroblasts
  • inflammatory cytokine-producing cells microphage-like THP-1 cells, RAW264.7 cells
  • Whether or not the candidate substance has an anti-inflammatory action can be determined by whether or not the inflammatory reaction in the arthritis model is suppressed by the addition of the candidate substance.
  • the presence or absence and / or degree of arthritis treatment effect can be determined by using synovitis, the degree of inflammatory cell infiltration, the presence or absence of rheumatoid factor, and the like as indices.
  • synovitis the degree of inflammatory cell infiltration
  • rheumatoid factor the degree of inflammatory cell infiltration
  • rheumatoid factor the degree of inflammatory cell infiltration
  • rheumatoid factor rheumatoid factor
  • the degree of inflammatory reaction can be evaluated using as an index.
  • the in vitro arthritis model can be used to screen in one step a substance that inhibits the function of Tet 3 and exhibits an effect of preventing and / or treating arthritis.
  • the method includes the following steps (1) to (3). (1) a step of bringing synovial fibroblasts into contact with a test substance; (2) measuring the degree of demethylation or invasiveness of 5-methylcytosine (5 mC) in the genome of the cell, (3) Compared to the measurement in the absence of the test substance, the test substance that suppresses the demethylation or invasiveness of 5-methylcytosine (5mC) in the genome of the cell is used to prevent arthritis and And / or selecting as a candidate for a therapeutic agent.
  • the method may further include a step of inducing inflammation at the same time as or before or after the step (1), if necessary.
  • methods for inducing inflammation include stimulation with inflammatory cytokines such as TNF ⁇ , and complex culture with inflammatory cytokine-producing cells such as monocytes, macrophages and neutrophils.
  • transwell TM A method of culturing target cells (eg, synovial fibroblasts) in the upper compartment and inflammatory cytokine-producing cells (macrophage-like THP-1 cells, RAW264.7 cells) in the lower compartment using a culture system or the like, respectively. Is mentioned.
  • the test substance is usually added to the medium in the lower compartment.
  • a test substance can be added to the medium in the upper compartment.
  • the degree of demethylation of 5-methylcytosine (5mC) in the cell genome is measured according to a known method such as Western blotting using an antibody that recognizes 5hmC or 5mC using the genomic DNA prepared from the cell. Can be quantified.
  • the Western blot method used in this screening method may be the same as the Western blot method described in the above-described method for measuring Tet 3 protein.
  • the form of the antibody recognizing 5hmC or 5mC is not particularly limited, and may be a polyclonal antibody having 5hmC or 5mC as an immunogen or a monoclonal antibody. Moreover, the measurement of the invasive degree of a cell can be measured according to the method as described in the Example mentioned later, for example.
  • cells that are brought into contact with or not in contact with the test substance are cultured in a medium (10% FCS-containing DMEM (which may contain inflammatory cytokine (eg, TNF ⁇ ) if necessary)) ( For example, after 96 hours), scraping the surface of the culture dish to which the cells adhere, and measuring the number of cells that infiltrate the scratched surface while culturing again, thereby measuring and comparing the degree of invasiveness of the cells.
  • a substance that inhibits the expression or function of Tet 3 obtained by using any one of the screening methods of the present invention is useful as a medicament for the prevention and / or treatment of inflammatory diseases.
  • the compound obtained by using the screening method of the present invention when used as the above-mentioned prophylactic / therapeutic agent, it can be formulated in the same manner as the low molecular weight compound that inhibits the expression or function of Tet 3, and the same administration route And administered orally or parenterally to humans or mammals (eg, mice, rats, rabbits, sheep, pigs, cows, horses, cats, dogs, monkeys, chimpanzees, etc.) Can do. V.
  • mammals eg, mice, rats, rabbits, sheep, pigs, cows, horses, cats, dogs, monkeys, chimpanzees, etc.
  • Tet 3 was high in synovial fibroblasts derived from rheumatoid arthritis patients stimulated with a synovial surface cell layer derived from rheumatoid arthritis patients and inflammatory cytokines. Therefore, arthritis can be examined by using the expression level of Tet 3 (or Tet 3 gene) in a sample derived from a subject as an index.
  • the present invention provides a method for examining arthritis, which comprises detecting or quantifying a Tet 3 gene transcription product or translation product from a subject-derived sample using a Tet 3 detection substance.
  • Tet3 detection substance include the following (a) or (b): (A) a nucleic acid probe or a nucleic acid primer capable of specifically detecting a transcript of the Tet 3 gene (B) An antibody that specifically recognizes the translation product of the Tet 3 gene.
  • arthritis include rheumatoid arthritis, psoriatic arthritis, spondyloarthritis (eg, ankylosing spondylitis), and preferably rheumatoid arthritis.
  • the sample used in the test method of the present invention contains a Tet3 gene product (eg, RNA, protein, its degradation product, etc.) that is to be detected or quantified and that is separated from the test subject.
  • any tissue or cell that can be used is not particularly limited. Examples thereof include synovium, synovial fibroblasts, and synovial surface cells.
  • Detection or quantification of Tet 3 in a sample isolated from a subject is performed by preparing a RNA (eg, total RNA, mRNA) fraction from the subject and detecting or quantifying a transcript of the Tet 3 gene contained in the fraction. Can be examined. Therefore, in one embodiment, the test method of the present invention is characterized by measuring using a nucleic acid probe or a nucleic acid primer capable of specifically detecting a transcript of the Tet 3 gene.
  • the RNA fraction can be prepared using a known method such as guanidine-CsCl ultracentrifugation, AGPC, etc., but using a commercially available RNA extraction kit (eg, RNeasy Mini Kit; manufactured by QIAGEN, etc.) High-purity total RNA can be prepared quickly and easily from a small amount of sample.
  • a means for detecting the transcript of the Tet 3 gene in the RNA fraction for example, a method using hybridization (Northern blot, dot blot, DNA chip analysis, etc.) or PCR (RT-PCR, competitive PCR, real-time PCR) Etc.).
  • Quantitative PCR methods such as competitive PCR and real-time PCR are preferable because they can detect changes in the expression of the Tet 3 gene quickly and easily from a small amount of sample with high quantitativeness.
  • the nucleic acid probe or nucleic acid primer capable of specifically detecting the transcript of the Tet 3 gene and the hybridization method using the nucleic acid probe or nucleic acid primer are the above-described screening methods for preventive and / or therapeutic agents for arthritis of the present invention. It may be the same as the nucleic acid for detection and the hybridization method of the present invention described.
  • the detection or quantification of Tet 3 in a sample isolated from a subject comprises preparing a protein fraction from the specimen and detecting or quantifying the translation product of the gene (ie, Tet3 protein) contained in the fraction. Can be examined. Detection or quantification of Tet3 can also be performed by an immunoassay (eg, ELISA, FIA, RIA, Western blot, etc.) using an antibody that specifically recognizes Tet3 protein. Therefore, in one embodiment, the test method of the present invention is characterized by measuring using an antibody capable of specifically detecting the translation product of Tet 3.
  • an immunoassay eg, ELISA, FIA, RIA, Western blot, etc.
  • the antibody specifically recognizing the translation product of Tet 3 and the immunological measurement method using the antibody are the detection antibody of the present invention described in the screening method for the prevention and / or treatment of arthritis of the present invention. And it may be the same as the immunological measurement method.
  • arthritis inspection method of the present invention arthritis can be examined by detecting or quantifying Tet3. Specifically, it may be a method including the following steps. (1) detecting or quantifying Tet 3 for a sample separated from a control group and a subject; (2) A step of comparing Tet3 detected or quantified in the control group with Tet3 detected or quantified in the subject.
  • the Tet3 concentration in the synovium is higher in patients with rheumatoid arthritis compared to osteoarthritis patients (control group).
  • Arthritis testing is based on such a positive correlation between the concentration of Tet3 and the prevalence of arthritis.
  • the concentration of Tet 3 in a sample from a control group and subject that does not develop arthritis is quantified, and the concentration of Tet 3 in a sample from the subject is compared to the concentration of Tet 3 in a sample from the control group.
  • a correlation diagram between the concentration of Tet 3 and the presence or absence of the onset of arthritis may be prepared in advance, and the Tet 3 concentration in the subject may be compared with the correlation diagram.
  • the concentration comparison is preferably performed based on the presence or absence of a significant difference.
  • Tet 3 When Tet 3 is detected or quantified at a higher value in the subject than in the control group, it can be determined that the possibility of developing arthritis as described above is high. Therefore, in addition to the steps (1) and (2) above, the test method of the present invention (3) develops arthritis when Tet3 is detected or quantified in subjects at a higher value than the control group. It may also include a step of determining that it is.
  • the present invention extends to a kit (diagnostic agent) for arthritis examination.
  • the arthritis test kit of the present invention is not particularly limited as long as it is a kit for simply carrying out the above-described test method of the present invention.
  • the test kit is (A) a nucleic acid probe or nucleic acid primer capable of specifically detecting a transcript of the Tet 3 gene, and / or (B) an antibody that specifically recognizes the translation product of the Tet 3 gene It contains.
  • the determination kit includes two or more of the above-described nucleic acids and / or antibodies, each nucleic acid or antibody specifically recognizes a different part of the base sequence of the Tet 3 gene from each other, or is a translation product of the Tet 3 gene. Different epitopes can be specifically recognized.
  • kits of the present invention includes the reagent containing the nucleic acid (a) as a component
  • examples of the nucleic acid include the probe nucleic acid or the primer oligonucleotide described above in the test method of the present invention.
  • the nucleic acid capable of detecting the expression of the Tet 3 gene can be provided as a solid in a dry state or in an alcohol precipitate state, or dissolved in water or an appropriate buffer (eg, TE buffer). Can also be provided.
  • the nucleic acid can be provided in a state of being previously labeled with any of the above-mentioned labeling substances, or can be provided separately from the labeling substance and can be used after labeling.
  • the nucleic acid can be provided in a state immobilized on an appropriate solid phase.
  • the solid phase include, but are not limited to, glass, silicon, plastic, nitrocellulose, nylon, polyvinylidene difluoride, and the like.
  • a functional group such as an amino group, an aldehyde group, an SH group, or biotin is introduced into a nucleic acid in advance, and a functional group capable of reacting with the nucleic acid on a solid phase (eg, aldehyde) Group, amino group, SH group, streptavidin, etc.), and the solid phase and the nucleic acid are cross-linked by covalent bond between the two functional groups, or the polyanionic nucleic acid is coated with the polycation and the static phase is coated.
  • a solid phase eg, aldehyde
  • the method include immobilization of nucleic acid using electric coupling, but are not limited thereto.
  • the nucleic acid contained in the test kit is constructed so that the expression of the Tet 3 gene can be detected by the same method (eg, Northern blot, dot blot, DNA array technology, quantitative RT-PCR, etc.). It is particularly preferable.
  • the kit of the present invention contains the reagent containing the antibody (b) as a component, examples of the antibody include the antibodies described above in the test method of the present invention.
  • the reagent constituting the kit of the present invention is a nucleic acid or antibody that can detect the expression of the Tet 3 gene and other substances necessary for the reaction for detecting the expression of the gene, and is stored in a coexisting state. Thus, a substance that does not adversely influence the reaction can be further contained.
  • the reagent may be provided with a separate reagent containing other substances necessary in the reaction to detect the expression of the Tet 3 gene.
  • the other substance include a reaction buffer, dNTPs, and a heat-resistant DNA polymerase.
  • competing PCR or real-time PCR it can further include a competitor nucleic acid, a fluorescent reagent (such as the above intercalator and fluorescent probe), and the like.
  • the reaction for detecting the expression of the Tet 3 gene is an antigen-antibody reaction
  • examples of the other substance include a reaction buffer, a competitor antibody, and a labeled secondary antibody (for example, the primary antibody is a rabbit anti-antibody reaction).
  • the primary antibody is a rabbit anti-antibody reaction.
  • human Tet 3 antibody mouse anti-rabbit IgG labeled with peroxidase, alkaline phosphatase, etc.), blocking solution, etc. may be mentioned.
  • the following examples are merely to illustrate the present invention more specifically, and do not limit the scope of the present invention.
  • Tet 1, Tet 2 and Tet 3 expression level of Tet protein family in synovium Tet protein family has three subtypes, Tet 1, Tet 2 and Tet 3, and the expression level varies from cell to cell. It is known that the genetic locus to be different is different. Therefore, the expression level of the Tet protein family was first examined by immunohistochemical staining in synovium from rheumatoid arthritis (RA) patients and osteoarthritis (OA) patients as controls. As for the synovium, a part of synovial tissue obtained at the time of joint surgery was cryopreserved and a tissue section was prepared later.
  • RA rheumatoid arthritis
  • OA osteoarthritis
  • the antibodies are Goat-anti-human TET1 Ab (Santa Cruz biotech, sc-1634443), mouse anti-human TET2 Ab (sc-136926), rabbit anti-human TET3 Ab (sc-139itx) 5-hmC) polyclonal Ab (ActiveMotif), rabbit anti-5-methylcytosine (5-mC) polyclonal Ab (ActiveMotif), mouse monoclonal anti-CD89A (Ab7470 ),
  • the secondary antibody was used MACH 2 double stain 2 (mouse-HRP + rabbit-AP). For observation, BIOREVO (keyence, Japan) was used.
  • Example 2 Expression level of Tet protein family in synovial fibroblasts (FLS) In subcultured FLS, there is no direct effect of inflammatory cytokines, but RA-specific DNA methylation profile is maintained. (Nakano K et al. Ann Rheum Dis 2013). Therefore, the expression levels (mRNA and protein) of the Tet family when cultured in the absence of inflammatory cytokines were compared between RA, OA, and healthy human FLS. As FLS, synovial cells extracted by adding collagenase treatment to the synovial tissue of Example 1 were used for passage 4-6, and FLS in which purity of 99% or more was confirmed was used. mRNA expression was analyzed by real-time PCR using StepOnePlus TM .
  • TET1 Hs00286756_m1
  • TET2 Hs00325999_m1
  • TET3 (00379125_m1) from TaqMan Gene Expression Assays were used, and the expression level was corrected with GAPDH (Hs9999999_m1).
  • Goat-anti-human TET1 Ab (Santa Cruz biotech, sc-1634443), mouse anti-human TET2 Ab (sc-136926), rabbit anti-human TET3 Ab, sc-19616 FITC-conjugated anti-goat IgG, FITC-conjugated anti-mouse IgG, and FITC-conjugated anti-rabbit IgG were used as antibodies.
  • BIOREVO keyence, Japan
  • BZ-II Viewer software (keyence) was used for image analysis.
  • TNF ⁇ and IL-1 ⁇ which are considered to be mainly involved in the pathological condition of RA in FLS of Example 2
  • Changes in expression level (mRNA, protein) of the Tet protein family were examined.
  • TNF ⁇ was stimulated with recombinant human TNF-alpha (R & D)
  • IL-1 ⁇ was stimulated with 50 ng / ml and 1 ng / ml, respectively, using recombinant human IL-1 ⁇ (RELIATech GmbH).
  • Example 5 Secretion level of inflammatory cytokine in FLS after suppression of Tet 3 expression Inhibition of Tet 3 expression using Tet 3 siRNA, followed by stimulation with TNF ⁇ and secretion of RA-derived FLS Evaluated.
  • FLS obtained by knocking down Tet 3 with siRNAs consisting of the nucleotide sequences of SEQ ID NO: 3 and SEQ ID NO: 4 was cultured in a medium containing TNF ⁇ (1 ng / ml) (10% FCS-containing DMEM) for 96 hours. After washing, the cells were further cultured for 48 hours in DMEM containing 0.1% FCS without TNF ⁇ . Tet 1 siRNA was used as a control.
  • siRNA IDs HSS129586, HSS123253 for TET1, HSS123253, HSS1233Hor TET2, HSS15364T used.
  • Low GC Duplex Invitrogen
  • Stealth TM RNAi Negative Control Duplexes was used as a control.
  • the siRNA gene was introduced by lipofection using Lipofectamine RNAiMAX Transfection Reagent (Life technologies). As a result of preliminary examination of gene transfer efficiency, it was decided to use HSS1533381 as TET3 siRNA and HSS129578 as TET1 siRNA.
  • the BD TM Cytometric Bead Array (CBA, BD, Japan) asked the SRL to measure the secreted amount of MMP3 (the test method was the LTIA method) .
  • the expression of adhesion molecules ICAM-1 and VCAM-1 was measured by flow-Cytometry method (FACSVerse TM, BD), and analyzed with FlowJo (Miltenyi Biotec).
  • IL-6, IL-8, VEGF, CCL2, MMP3, etc. were found to induce TNF ⁇ -dependent expression.
  • CCL2 inhibited TNF ⁇ -dependent expression induction by Tet 3 knockdown (FIG. 10). This phenomenon was also confirmed at the mRNA and secretory protein levels.
  • TAM ⁇ -dependent expression induction of ICAM1 was similarly inhibited by Tet 3 knockdown. It is known that CCL2 and ICAM-1 are involved in the invasion of FLS into bone and cartilage tissues and deeply involved in the pathogenesis of RA.
  • Tet 3 knockdown is effective in the prevention and treatment of RA by suppressing the expression of CCL2 and ICAM-1 through maintenance of DNA methylation level.
  • detection was attempted for IL-1 ⁇ , TNF ⁇ , and IL-17 ⁇ , they could not be detected because the values were below detection sensitivity.
  • Example 6 Invasion ability of FLS after suppression of Tet 3 expression After inhibiting Tet 3 expression using Tet 3 siRNA, stimulation with TNF ⁇ was performed, and the invasion ability of RA-derived FLS was evaluated by Scratch assay.
  • an agent for preventing and / or treating arthritis or a diagnostic kit targeting Tet 3, which is a factor different from the target of conventional therapeutic agents is provided.
  • therapeutic drugs with new mechanisms of action
  • Better treatment can also be provided for arthritic patients who have acquired resistance to the therapeutic agent.
  • the present invention can also be used for the purpose of preventing arthritis in advance, and for the purpose of preventing recurrence of disease in patients whose arthritis has been temporarily ameliorated.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Hematology (AREA)
  • Biochemistry (AREA)
  • Physics & Mathematics (AREA)
  • General Engineering & Computer Science (AREA)
  • Urology & Nephrology (AREA)
  • Microbiology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Rheumatology (AREA)
  • Plant Pathology (AREA)
  • Food Science & Technology (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Analytical Chemistry (AREA)
  • Cell Biology (AREA)
  • Rehabilitation Therapy (AREA)
  • Pain & Pain Management (AREA)
  • Dermatology (AREA)

Abstract

La présente invention concerne : un agent prophylactique/thérapeutique contre l'arthrite dont la polyarthrite rhumatoïde, qui comprend un inhibiteur de l'expression de Tet3; une trousse pour le diagnostic de l'arthrite dont la polyarthrite rhumatoïde; et un moyen de recherche d'une nouvelle substance ayant une activité prophylactique et/ou thérapeutique sur l'arthrite dont la polyarthrite rhumatoïde.
PCT/JP2015/074556 2014-08-28 2015-08-24 Agent prophylactique/thérapeutique contre l'arthrite, trousse d'essai pour l'arthrite, et procédé de criblage d'agent prophylactique/thérapeutique contre l'arthrite Ceased WO2016031996A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2016545659A JPWO2016031996A1 (ja) 2014-08-28 2015-08-24 関節炎の予防・治療剤、検査キット、並びに関節炎予防・治療薬のスクリーニング方法

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2014174638 2014-08-28
JP2014-174638 2014-08-28

Publications (1)

Publication Number Publication Date
WO2016031996A1 true WO2016031996A1 (fr) 2016-03-03

Family

ID=55399876

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2015/074556 Ceased WO2016031996A1 (fr) 2014-08-28 2015-08-24 Agent prophylactique/thérapeutique contre l'arthrite, trousse d'essai pour l'arthrite, et procédé de criblage d'agent prophylactique/thérapeutique contre l'arthrite

Country Status (2)

Country Link
JP (1) JPWO2016031996A1 (fr)
WO (1) WO2016031996A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020206055A1 (fr) * 2019-04-02 2020-10-08 Yale University Méthodes de traitement de maladies et de troubles associés à un taux de tet accru, un taux de h19 accru, un taux accru de signalisation par tgf, ou toute combinaison de ceux-ci
CN113970641A (zh) * 2021-10-27 2022-01-25 南通大学附属医院 Cdc37在类风湿关节炎预防或治疗方面的应用

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2013021932A (ja) * 2011-07-15 2013-02-04 Chiba Univ 関節リウマチに対する抗il−6受容体抗体療法の有効性の予測方法

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2013021932A (ja) * 2011-07-15 2013-02-04 Chiba Univ 関節リウマチに対する抗il−6受容体抗体療法の有効性の予測方法

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
"Merck Manual 18th edition Japanese language Edition", 25 April 2007, NIKKEI BUSINESS PUBLICATIONS, INC., ISBN: 978-4-8222-0398-6, pages: 299 *
HIROSHI MAKINO ET AL.: "Kakusan Iyaku Application of nucleic acid medicine for arthritis", JAPANESE JOURNAL OF CLINICAL MEDICINE (SPECIAL EXTRA) KANSETSU RHEUMATOID, vol. 68, no. 5, 2010, pages 633 - 638, ISSN: 0047-1852 *
ISAO MATSUMOTO: "Kansetsuen Model", THE JOURNAL OF THE JAPANESE SOCIETY OF INTERNAL MEDICINE, vol. 101, no. 10, 10 October 2012 (2012-10-10), pages 2839 - 2843, ISSN: 0021-5384 *
KAZUHISA NAKANO ET AL.: "Sagyo Kanrensei Kinkokkakukei Shikkan ni Okeru Epigenetic Biomarker no Tansaku", JOURNAL OF UOEH, vol. 36, no. 1, 1 March 2014 (2014-03-01), pages 77, ISSN: 0387-821X *
KAZUKO SHIOZAWA ET AL.: "Idenshi Chiryo Total na Kansetsu Rheumatoid no Chiryo wa Kano ka", PHARMA MEDICA, vol. 17, no. 10, 1999, pages 101 - 113, ISSN: 0289-5803 *
SHUN'ICHI KUBO ET AL.: "Kansetsuen Model Rat ni Taisuru siRNA Chiryo no Koka", INFLAMMATION & IMMUNOLOGY, vol. 18, no. 2, 2010, pages 125 - 129, ISSN: 0918-8371 *
YOSHIHIRO KITAMURA: "Jikken Koza Reporter Assay", SURGERY FRONTIER, vol. 13, no. 4, 2006, pages 426 - 430, ISSN: 1340-5594 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020206055A1 (fr) * 2019-04-02 2020-10-08 Yale University Méthodes de traitement de maladies et de troubles associés à un taux de tet accru, un taux de h19 accru, un taux accru de signalisation par tgf, ou toute combinaison de ceux-ci
US20220160751A1 (en) * 2019-04-02 2022-05-26 Yale University Methods of treatment of diseases and disorders associated with increased tet level, increased h19 level, increased level of tgf signaling, or any combination thereof
CN113970641A (zh) * 2021-10-27 2022-01-25 南通大学附属医院 Cdc37在类风湿关节炎预防或治疗方面的应用

Also Published As

Publication number Publication date
JPWO2016031996A1 (ja) 2017-06-08

Similar Documents

Publication Publication Date Title
US12066429B2 (en) Method for inhibiting the expression of inflammation promoting factors
JP7175526B2 (ja) 細胞遊走調節に関する疾患の予防・治療剤および肺間質の疾患の疾患活動性判定・予後評価
JP5794514B2 (ja) 腫瘍血管新生阻害剤
JP2015173601A (ja) がん幹細胞の増殖抑制物質のスクリーニング方法
JP2011032236A (ja) 腫瘍血管新生阻害剤
WO2016031996A1 (fr) Agent prophylactique/thérapeutique contre l'arthrite, trousse d'essai pour l'arthrite, et procédé de criblage d'agent prophylactique/thérapeutique contre l'arthrite
JP6226315B2 (ja) 炎症性疾患の予防・治療剤、並びに炎症性疾患予防・治療薬のスクリーニング方法
JP6653120B2 (ja) Ikaros阻害に基づく抗炎症薬
EP2153847A1 (fr) Gène sensible à une maladie des os/articulations et son utilisation
JP2011502108A (ja) 癌の予防・治療剤
WO2010008084A1 (fr) Nouvelle application d'utilisation de récepteur reconnaissant une chaîne de sucre
EP2123303A1 (fr) Agent améliorant pour la résistance à l'insuline
JP5669271B2 (ja) 腫瘍血管新生阻害剤
JP2008263851A (ja) 熱射病の治療ターゲット
US8309687B2 (en) Biomarker specific for cancer
JP2023055804A (ja) Card14を用いた治療、診断およびスクリーニング
WO2011115271A1 (fr) Inhibiteur d'angiogenèse de tumeur
WO2011019065A1 (fr) Inhibiteur de l'angiogenèse tumorale
JP2011105669A (ja) 抗腫瘍剤および抗腫瘍剤のスクリーニング方法
EP1577387A1 (fr) Medicaments preventifs/remedes pour le cancer
WO2011115268A1 (fr) Inhibiteur d'angiogenèse de tumeur

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15835149

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2016545659

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15835149

Country of ref document: EP

Kind code of ref document: A1