WO2016031996A1 - Prophylactic/therapeutic agent for arthritis, test kit for arthritis, and method for screening for prophylactic/therapeutic agent for arthritis - Google Patents

Prophylactic/therapeutic agent for arthritis, test kit for arthritis, and method for screening for prophylactic/therapeutic agent for arthritis 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
Application number
PCT/JP2015/074556
Other languages
French (fr)
Japanese (ja)
Inventor
和久 中野
Original Assignee
学校法人産業医科大学
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 学校法人産業医科大学 filed Critical 学校法人産業医科大学
Priority to JP2016545659A priority Critical patent/JPWO2016031996A1/en
Publication of WO2016031996A1 publication Critical patent/WO2016031996A1/en

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 N.A.
    • 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.

Abstract

The present invention provides: a prophylactic/therapeutic agent for arthritis including rheumatoid arthritis, which comprises a Tet3 expression inhibitor; a kit for diagnosing arthritis including rheumatoid arthritis; and a means for searching for a novel substance having a prophylactic and/or therapeutic activity on arthritis including rheumatoid arthritis.

Description

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Arthritis prevention / treatment agent, test kit, and screening method for arthritis prevention / treatment agent
 本発明は、関節炎、特に関節リウマチ(rheumatoid arthritis;RA)の予防・治療剤、並びに関節炎、特に関節リウマチ予防・治療薬のスクリーニング方法に関する。より詳しくは、Tet 3(Ten−Eleven translocation 3)の機能を阻害する物質を含有する、関節炎、特に関節リウマチの予防剤及び/又は治療剤、並びにTet 3の機能阻害を指標とする、関節炎、特に関節リウマチ予防・治療薬の候補物質をスクリーニングする方法に関する。 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.
 関節リウマチは、全身の関節に慢性炎症を生じる難治性の自己免疫疾患である。関節内の滑膜組織が増殖し、進行性に軟骨と骨が破壊される。関節リウマチの滑膜線維芽細胞(FLS)はマクロファージやリンパ球により刺激(TNFα、IL−1βなどの炎症性サイトカイン)されて活性化され、活性化した滑膜線維芽細胞は、炎症性サイトカインやケモカイン産生、基質分解酵素分泌により関節破壊を起こす一方、増殖能・浸潤能の亢進、アポトーシス感受性低下などの癌細胞と似た性質も持つ。近年、GWAS(Genome−Wide Association Study)の結果より約30の遺伝子多型が関節リウマチの発症や重症度に関わることが知られるようになってきた。その一方で、エピジェネティクスの異常が関節リウマチの発症や重症度に深く関わることが示唆されるようになった。DNAのメチル化はエピジェネティクスの代表的な機構の一つであり、関節リウマチにおいてもゲノム全体、もしくは特定の遺伝子のプロモーター領域のDNAメチル化異常が存在することが報告されてきた。発明者は近年、ゲノム網羅的DNAメチル化解析により、関節リウマチ患者由来滑膜線維芽細胞には疾患に特有なDNAメチル化パターンが存在し、異常メチル化を示した遺伝子の多くが関節リウマチの病態に深く関わることを示した(非特許文献1)。
 生物学的製剤による炎症性サイトカインを標的とした関節リウマチ治療は、寛解導入を身近なものにしたが、関節リウマチ治療にはいわゆる「Window of Opportunity」が存在し、治療開始の遅れは治療効果を限定的なものとする。このことは、炎症環境の持続自体が、滑膜炎症部位に存在する細胞をより攻撃的でかつ治療抵抗性な表現型に変質させている可能性を示唆するが、この詳細なメカニズムは不詳であった。発明者は、関節リウマチにおける滑膜炎症・骨関節破壊の中心的役割を担うTNFαやIL−1βが、FLSにおいてDNAメチル化酵素(DNMT)の発現を低下させ、受動的脱メチル化を促進することを示し、疾患特有のDNAメチル化パターン形成の分子機構の一端を明らかにした(非特許文献2)。
 しかしながら、これまでは能動的なDNA脱メチル化の機構が不詳であったため、DNAメチル化・脱メチル化のダイナミズムの全貌は不明であった。近年、DNA脱メチル化酵素として機能するTet(Ten−Eleven translocation)タンパク質ファミリーが同定され、Tetタンパク質が5−メチルシトシン(5mC)を水酸化して5−ヒドロキシメチルシトシン(5hmC)を合成することが報告され、ES細胞などにおけるDNAメチル化のダイナミズムの研究は急速な進展を見せている。
 ただ、関節リウマチにおけるTetタンパク質の関与については未だ明らかにされておらず、Tetタンパク質が関節リウマチの治療標的となり得るか否かは全く不明のままであった。
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 TNFα and IL-1β) 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. In recent years, from the results of GWAS (Genome-Wide Association Study), it has become known that about 30 gene polymorphisms are related to the onset and severity of rheumatoid arthritis. On the other hand, it has been suggested that abnormalities in epigenetics are deeply related to the onset and severity of rheumatoid arthritis. DNA methylation is one of the typical mechanisms of epigenetics, and it has been reported that rheumatoid arthritis also has abnormal DNA methylation in the entire genome or the promoter region of a specific gene. The inventors have recently conducted genome-wide DNA methylation analyses, and there are DNA methylation patterns peculiar to diseases in synovial fibroblasts derived from rheumatoid arthritis patients, and many of the genes exhibiting abnormal methylation have been found in rheumatoid arthritis. It was shown to be deeply involved in the pathological condition (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. This suggests that the persistence of the inflammatory environment itself may transform the cells present at the site of synovial inflammation into a more aggressive and treatment-resistant phenotype, but this detailed mechanism is unknown. there were. The inventor found that TNFα and IL-1β, which play a central role in synovial inflammation and bone joint destruction in rheumatoid arthritis, decrease the expression of DNA methylase (DNMT) in FLS and promote passive demethylation This indicates that the molecular mechanism of disease-specific DNA methylation pattern formation was clarified (Non-patent Document 2).
However, since the mechanism of active DNA demethylation was unknown until now, the whole picture of the dynamism of DNA methylation / demethylation was unknown. Recently, a Tet (Ten-Eleven transcription) protein family that functions as a DNA demethylase has been identified, and the Tet protein hydroxylates 5-methylcytosine (5mC) to synthesize 5-hydroxymethylcytosine (5hmC). The research on DNA methylation dynamism in ES cells and the like is making rapid progress.
However, the involvement of Tet protein in rheumatoid arthritis has not yet been clarified, and it remains unclear whether Tet protein can be a therapeutic target for rheumatoid arthritis.
 従って、本発明の目的は、関節炎、特に関節リウマチにおけるTetタンパク質、特にTet3タンパク質の機能を明らかにすることであり、当該機能に基づいてTet 3を標的とする関節炎の予防・治療剤、検査キットを提供すること、並びにTet 3の機能調節を指標として、関節炎の予防及び/又は治療活性を有する新規な物質を探索する手段を提供することである。 Accordingly, 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.
 本発明者は、上記の目的を達成すべく、関節リウマチ患者由来の滑膜や炎症性サイトカインで刺激した、関節リウマチ患者由来の滑膜線維芽細胞(FLS)におけるTetタンパク質ファミリーの発現レベルおよびDNAメチル化レベルを測定した。その結果、健常者や変形性関節症(OA)患者に比べて、関節リウマチ患者由来の滑膜表層細胞層や炎症性サイトカインで刺激した、関節リウマチ患者由来の滑膜線維芽細胞(FLS)では、Tet 3の発現レベルが高く、DNAメチル化レベルも低いことを見出した。また、関節リウマチ患者由来のFLSのTet3をノックダウンした場合、FLS自身の組織(軟骨、骨)浸潤に関与するケモカインCCL2や接着分子ICAM1の発現が阻害され、また、該FLSの浸潤能も抑制された。これらの結果は、Tet 3の選択的阻害が、DNAメチル化レベルの維持を介してCCL2やICAM1の発現を阻害し、それに伴って浸潤性に代表される滑膜線維芽細胞の活性化を阻害することによって、関節炎、特に関節リウマチの新規治療につながることを示唆するものである。
 本発明者は、これらの知見に基づいてさらに検討を重ねた結果、本発明を完成するに至った。
 即ち、本発明は以下の通りである。
[1]Tet 3(Ten−Eleven translocation 3)の発現阻害物質を含有する、関節炎の予防及び/又は治療剤。
[2]Tet 3の発現阻害物質が、
(a)Tet 3遺伝子の転写産物に対するアンチセンス核酸、
(b)Tet 3遺伝子の転写産物に対するリボザイム核酸、又は
(c)Tet 3遺伝子の転写産物に対してRNAi活性を有する核酸もしくはその前駆体である、[1]に記載の剤。
[3]関節炎が関節リウマチ、乾癬性関節炎または脊椎関節炎である、[1]または[2]に記載の剤。
[4]以下の(1)~(3)の工程を含む、関節炎の予防及び/又は治療薬のスクリーニング方法:
(1)Tet 3遺伝子もしくは該遺伝子の転写調節領域の制御下にあるレポータータンパク質をコードする核酸を含む細胞を、被検物質に接触させる工程、
(2)前記細胞におけるTet 3遺伝子もしくはTet 3タンパク質又はレポータータンパク質の発現量を測定する工程、
(3)被検物質の非存在下において測定した場合と比較して、Tet 3遺伝子もしくはTet 3タンパク質又はレポータータンパク質の発現量を低下させた被検物質を、関節炎の予防及び/又は治療薬の候補として選択する工程。
[5]Tet 3と被検物質とを接触させ、Tet 3と結合能を有する被検物質を関節炎の予防及び/又は治療薬の候補として選択することを特徴とする、関節炎の予防及び/又は治療薬のスクリーニング方法。
[6]関節炎の予防及び/又は治療薬の候補として選択された被検物質を関節炎モデルに適用し、該モデルにおける炎症反応を抑制するか否かを検定することをさらに含む、[5]に記載の方法。
[7]以下の(1)~(3)の工程を含む、関節炎の予防及び/又は治療薬のスクリーニング方法:
(1)滑膜線維芽細胞を、被検物質に接触させる工程、
(2)前記細胞のゲノムの5−メチルシトシン(5mC)の脱メチル化または浸潤性の程度を測
定する工程、
(3)被検物質の非存在下において測定した場合と比較して、前記細胞のゲノムの5−メチルシトシン(5mC)の脱メチル化または浸潤性を抑制した被検物質を、関節炎の予防及び/又は治療薬の候補として選択する工程。
[8]関節炎が関節リウマチ、乾癬性関節炎または脊椎関節炎である、[4]~[7]のいずれか1つに記載の方法。
[9]被験者由来の試料から、下記(a)または(b)を用いてTet 3遺伝子の転写産物または翻訳産物を検出または定量することを含む、関節炎の検査方法:
(a)Tet 3遺伝子の転写産物を特異的に検出し得る核酸プローブまたは核酸プライマー
(b)Tet 3遺伝子の翻訳産物を特異的に認識する抗体。
[10]関節炎が関節リウマチ、乾癬性関節炎または脊椎関節炎である、[9]に記載の方法。
[11]下記(a)および/または(b):
(a)Tet 3遺伝子の転写産物を特異的に検出し得る核酸プローブまたは核酸プライマー
(b)Tet 3遺伝子の翻訳産物を特異的に認識する抗体
を含有してなる、関節炎検査用キット。
[12]関節炎が関節リウマチ、乾癬性関節炎または脊椎関節炎である、[11]に記載のキット。
[13]Tet 3(Ten−Eleven translocation 3)の発現阻害物質の有効量を対象に投与することを含む、関節炎の予防及び/又は治療方法。
[14]関節炎の予防及び/又は治療に使用するための、Tet 3(Ten−Eleven translocation 3)の発現阻害物質。
[15]関節炎の予防及び/又は治療剤を製造するための、Tet 3(Ten−Eleven translocation 3)の発現阻害物質の使用。
In order to achieve the above object, 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. As a result, compared with healthy subjects and osteoarthritis (OA) patients, the synovial fibroblasts (FLS) derived from rheumatoid arthritis patients stimulated with rheumatoid arthritis patient-derived synovial surface cell layers and inflammatory cytokines We found that the expression level of Tet 3 was high and the DNA methylation level was low. In addition, when Tet3 of FLS derived from patients with rheumatoid arthritis is knocked down, expression of chemokine CCL2 and adhesion molecule ICAM1 involved in FLS's own tissue (cartilage, bone) infiltration is inhibited, and the invasion ability of FLS is also suppressed. It was done. These results indicate that selective inhibition of Tet 3 inhibits the expression of CCL2 and ICAM1 through the maintenance of DNA methylation levels, and accordingly inhibits the activation of synovial fibroblasts represented by invasiveness. This suggests that this may lead to a novel treatment for arthritis, particularly rheumatoid arthritis.
As a result of further studies based on these findings, the present inventors have completed the present invention.
That is, the present invention is as follows.
[1] A prophylactic and / or therapeutic agent for arthritis, which contains an inhibitor of expression of Tet 3 (Ten-Eleven transcription 3).
[2] 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.
[3] The agent according to [1] or [2], wherein the arthritis is rheumatoid arthritis, psoriatic arthritis or spondyloarthritis.
[4] 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.
[5] Contact with Tet 3 and a test substance, and select a test substance capable of binding to Tet 3 as a prophylactic and / or therapeutic drug for arthritis, and / or prevention of arthritis A screening method for therapeutic drugs.
[6] 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.
[7] 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.
[8] The method according to any one of [4] to [7], wherein the arthritis is rheumatoid arthritis, psoriatic arthritis or spondyloarthritis.
[9] 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.
[10] The method according to [9], wherein the arthritis is rheumatoid arthritis, psoriatic arthritis or spondyloarthritis.
[11] The following (a) and / or (b):
(A) a nucleic acid probe or nucleic acid primer capable of specifically detecting a Tet 3 gene transcription product; and (b) an arthritis test kit comprising an antibody that specifically recognizes a translation product of the Tet 3 gene.
[12] The kit according to [11], wherein the arthritis is rheumatoid arthritis, psoriatic arthritis or spondyloarthritis.
[13] 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).
[14] An expression inhibitor of Tet 3 (Ten-Eleven transcription 3) for use in the prevention and / or treatment of arthritis.
[15] Use of a Tet 3 (Ten-Eleven tranlation 3) expression inhibitor for the manufacture of an agent for preventing and / or treating arthritis.
 本発明により、Tet 3は関節リウマチの病態悪化に関与することが明らかとなったので、Tet 3の発現もしくは機能を阻害することにより、関節炎を治療又は予防することができる。また、Tet 3の発現もしくは機能阻害を指標として、関節炎の治療又は予防薬をスクリーニングすることができる。さらに、Tet 3の発現を指標として、関節炎を検査することができる。 Since the present invention has revealed that 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. In addition, therapeutic or prophylactic agents for arthritis can be screened using Tet 3 expression or function inhibition as an index. Furthermore, arthritis can be examined using the expression of Tet 3 as an index.
 図1は関節リウマチ(RA)患者由来の滑膜組織における、Tetタンパク質ファミリー(Tet1,2,3)の発現、DNAメチル化(5−メチルシトシン(5mC)、5−ヒドロキシメチルシトシン(5hmC))を示す免疫組織化学染色像である。
 図2は変形性関節症(OA)患者由来の滑膜組織における、Tetタンパク質ファミリー(Tet1,2,3)の発現、DNAメチル化(5−メチルシトシン(5mC)、5−ヒドロキシメチルシトシン(5hmC))を示す免疫組織化学染色像である。
 図3は関節リウマチ(RA)患者由来の滑膜組織における、Tet3タンパク質(青色)とCD55タンパク質(茶色)の二重染色、Tet3タンパク質(青色)とCD68タンパク質(茶色)の二重染色を示す免疫組織化学染色像である。
 図4はサイトカイン未刺激のFLS(健常人、OA、RA)における、Tetファミリー(Tet1,Tet 2,Tet 3)の相対的mRNA発現レベルを示す図である。
 図5はサイトカイン未刺激のFLS(OA、RA)における、Tetファミリー(緑色)(Tet1,Tet 2,Tet 3)の発現と核(青色)を示す免疫組織化学染色像である。
 図6はTNFαやIL−1βによる刺激後における、FLS(n=4(RA 2,OA 2))におけるTetファミリー(Tet 1,Tet 2,Tet 3)のmRNA発現レベルの経時変化を示す図である。
 図7はTNFα刺激後における、FLSにおけるTet3タンパク質のN/C比の経時変化を示す図である。
 図8AはTNFαによる刺激後における、OA由来または健常者由来FLS(OA,Nr)におけるTet3のタンパク質発現レベルを示す図である。図8BはTNFαによる刺激後における、AR由来FLSにおけるTet 3のタンパク質発現レベルを示す図である。
 図9Aは実施例4の試験過程を示す図である。図9BはTNFαによる刺激後における、AR由来FLSにおける5hmCレベルを示すドットブロット像である。図9CはAR由来FLSにおける5hmCレベルを示すグラフである。
 図10Aは実施例5の試験過程を示す図である。図10BはTet3をノックダウンし、TNFαによる刺激後における、AR由来FLSにおける炎症性サイトカインの分泌レベルを示すグラフである。
 図11Aは実施例6の試験過程を示す図である。図11BはTet3をノックダウンし、TNFαによる刺激後の、AR由来FLSのScratch assayを示す顕微鏡像である。図11CはTet 3をノックダウンし、TNFαによって刺激し、scratch後24時間における、AR由来FLSの単位面積当たりの浸潤細胞数を示すグラフである。
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 | staining image which shows.
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. 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. 6 is a graph showing temporal changes in the mRNA expression level of the Tet family (Tet 1, Tet 2, Tet 3) in FLS (n = 4 (RA 2, OA 2)) after stimulation with TNFα or IL-1β. is there.
FIG. 7 is a graph showing the change over time in the N / C ratio of Tet3 protein in FLS after stimulation with TNFα.
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 TNFα. FIG. 8B shows the protein expression level of Tet 3 in AR-derived FLS after stimulation with TNFα.
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 TNFα. 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. 10B is a graph showing the secretion level of inflammatory cytokines in AR-derived FLS after knocking down Tet3 and stimulation with TNFα.
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 TNFα. 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 TNFα, and 24 hours after scratch.
 本発明は、少なくとも部分的には、Tet 3が関節リウマチの増悪に寄与していることの発見に基づく。当該知見は、Tet 3が関節炎、特に関節リウマチマーカーとして利用できるだけでなく、関節炎、特に関節リウマチの創薬標的ともなり得ることを示すものである。即ち、Tet 3の既知の阻害薬は関節炎の予防及び/又は治療に有用であるとともに、Tet3タンパク質やそれを発現する細胞・動物を用いて、新規なTet 3阻害薬、ひいては関節炎の予防・治療薬となる物質を探索することもできる。
I.Tet 3又はこれをコードする核酸
 本明細書において、Tet 3は公知のタンパク質であり、Genbank Accession No.:043151として知られている、配列番号:2で表されるヒトTet 3のアミノ酸配列、あるいはこれと実質的に同一のアミノ酸配列を含むタンパク質である。本明細書において、タンパク質およびペプチドは、ペプチド表記の慣例に従って左端がN末端(アミノ末端)、右端がC末端(カルボキシル末端)で記載される。
 本明細書において、Tet 3はヒトや他の温血動物(例えば、マウス、ラット、ウシ、サル、イヌ、ブタ、ヒツジ、ウサギ、モルモット、ハムスター、ニワトリなど)の細胞[例えば、滑膜線維芽細胞、滑膜表層細胞など]又は組織[例えば、滑膜(特に滑膜表層細胞層)など]等から、公知のタンパク質分離精製技術により単離・精製されるものであってよい。
 「配列番号:2で表されるアミノ酸配列またはこれと実質的に同一のアミノ酸配列」としては、以下の(a)~(e)が挙げられる:
(a)配列番号:2で示されるアミノ酸配列;
(b)配列番号:2で示されるアミノ酸配列において、1もしくは複数のアミノ酸が欠失、付加、挿入もしくは置換され、かつ5−メチルシトシン(5mC)の脱メチル化活性または滑膜線維芽細胞の浸潤の促進活性を有するアミノ酸配列;
(c)配列番号:2で示されるアミノ酸配列と90%以上の相同性を有し、かつ5−メチルシトシン(5mC)の脱メチル化活性または滑膜線維芽細胞の浸潤の促進活性を有するアミノ酸配列;
(d)配列番号1で示される塩基配列を有するDNAによりコードされるアミノ酸配列;
(e)配列番号1で示される塩基配列の相補鎖配列を有するDNAとストリンジェントな条件下でハイブリダイズするDNAによりコードされ、かつ5−メチルシトシン(5mC)の脱メチル化活性または滑膜線維芽細胞の浸潤の促進活性を有するアミノ酸配列。
 具体的には、配列番号:2で表されるアミノ酸配列からなるヒトTet 3タンパク質の他の哺乳動物におけるオルソログのアミノ酸配列、または配列番号:2で表されるアミノ酸配列からなるヒトTet 3タンパク質もしくはそのオルソログのスプライスバリアント、アレル変異体もしくは多型バリアントにおけるアミノ酸配列が挙げられる。
 ここで「相同性」とは、当該技術分野において公知の数学的アルゴリズムを用いて2つのアミノ酸配列をアラインさせた場合の、最適なアラインメント(好ましくは、該アルゴリズムは最適なアラインメントのために配列の一方もしくは両方へのギャップの導入を考慮し得るものである)における、オーバーラップする全アミノ酸残基に対する同一アミノ酸および類似アミノ酸残基の割合(%)を意味する。「類似アミノ酸」とは物理化学的性質において類似したアミノ酸を意味し、例えば、芳香族アミノ酸(Phe、Trp、Tyr)、脂肪族アミノ酸(Ala、Leu、Ile、Val)、極性アミノ酸(Gln、Asn)、塩基性アミノ酸(Lys、Arg、His)、酸性アミノ酸(Glu、Asp)、水酸基を有するアミノ酸(Ser、Thr)、側鎖の小さいアミノ酸(Gly、Ala、Ser、Thr、Met)などの同じグループに分類されるアミノ酸が挙げられる。このような類似アミノ酸による置換はタンパク質の表現型に変化をもたらさない(即ち、保存的アミノ酸置換である)ことが予測される。保存的アミノ酸置換の具体例は当該技術分野で周知であり、種々の文献に記載されている(例えば、Bowieら,Science,247:1306−1310(1990)を参照)。
 本明細書におけるアミノ酸配列の相同性は、相同性計算アルゴリズムNCBI BLAST(National Center for Biotechnology Information Basic Local Alignment Search Tool)を用い、以下の条件(期待値=10;ギャップを許す;マトリクス=BLOSUM62;フィルタリング=OFF)にて計算することができる。アミノ酸配列の相同性を決定するための他のアルゴリズムとしては、例えば、Karlinら,Proc.Natl.Acad.Sci.USA,90:5873−5877(1993)に記載のアルゴリズム[該アルゴリズムはNBLASTおよびXBLASTプログラム(version 2.0)に組み込まれている(Altschulら,Nucleic Acids Res.,25:3389−3402(1997))]、Needlemanら,J.Mol.Biol.,48:444−453(1970)に記載のアルゴリズム[該アルゴリズムはGCGソフトウェアパッケージ中のGAPプログラムに組み込まれている]、MyersおよびMiller,CABIOS,4:11−17(1988)に記載のアルゴリズム[該アルゴリズムはCGC配列アラインメントソフトウェアパッケージの一部であるALIGNプログラム(version 2.0)に組み込まれている]、Pearsonら,Proc.Natl.Acad.Sci.USA,85:2444−2448(1988)に記載のアルゴリズム[該アルゴリズムはGCGソフトウェアパッケージ中のFASTAプログラムに組み込まれている]等が挙げられ、それらも同様に好ましく用いられ得る。
 上記(e)におけるストリンジェントな条件とは、例えば、Current Protocols in Molecular Biology,John Wiley & Sons,6.3.1−6.3.6,1999に記載される条件、例えば、6×SSC(sodium chloride/sodium citrate)/45℃でのハイブリダイゼーション、次いで0.2×SSC/0.1% SDS/50~65℃での一回以上の洗浄等が挙げられるが、当業者であれば、これと同等のストリンジェンシーを与えるハイブリダイゼーションの条件を適宜選択することができる。
 より好ましくは、「配列番号:2で表されるアミノ酸配列と実質的に同一のアミノ酸配列」として、配列番号:2で表されるアミノ酸配列と、約90%以上、好ましくは約95%以上、より好ましくは約96%以上、いっそう好ましくは約97%以上、特に好ましくは約98%以上、最も好ましくは約99%以上の同一性を有するアミノ酸配列が挙げられる。
 「配列番号:2で表されるアミノ酸配列と実質的に同一のアミノ酸配列を含むタンパク質」は、配列番号:2で表されるアミノ酸配列と実質的に同一のアミノ酸配列を含み、かつ配列番号:2で表されるアミノ酸配列からなるタンパク質と実質的に同質の機能を有するタンパク質である。
 ここで「実質的に同質の機能」とは、例えば生理学的に、あるいは薬理学的にみて、その性質が定性的に同じであることを意味し、機能の程度(例、約0.1~約10倍、好ましくは0.5~2倍)や、タンパク質の分子量などの量的要素は異なっていてもよい。また、Tet3の機能としては、5−メチルシトシン(5mC)を5−ヒドロキシメチルシトシン(5hmC)、5−フォルミルシトシン(5fC)または5−カルボキシルシトシン(5CaC)に変換する活性(以下、「5−メチルシトシン(5mC)の脱メチル化活性」)、滑膜線維芽細胞の浸潤の促進活性等が挙げられ、上記の活性を有するタンパク質を、「実質的に同質の機能を有するタンパク質」とみなすことができる。
 ここで、5−メチルシトシン(5mC)の脱メチル化活性および滑膜線維芽細胞の浸潤の促進活性は、例えば、後述する実施例に記載の通りに測定することができる。
 本発明におけるTet3タンパク質として、例えば、(i)配列番号:2で表されるアミノ酸配列中の1~30個、好ましくは1~10個、より好ましくは1~数(5、4、3もしくは2)個のアミノ酸が欠失したアミノ酸配列、(ii)配列番号:2で表されるアミノ酸配列に1~30個、好ましくは1~10個、より好ましくは1~数(5、4、3もしくは2)個のアミノ酸が付加したアミノ酸配列、(iii)配列番号:2で表されるアミノ酸配列に1~30個、好ましくは1~10個、より好ましくは1~数(5、4、3もしくは2)個のアミノ酸が挿入されたアミノ酸配列、(iv)配列番号:2で表されるアミノ酸配列中の1~30個、好ましくは1~10個、より好ましくは1~数(5、4、3もしくは2)個のアミノ酸が他のアミノ酸で置換されたアミノ酸配列、または(v)それらを組み合わせたアミノ酸配列を含有するタンパク質なども含まれる。
 上記のようにアミノ酸配列が挿入、欠失、付加または置換されている場合、その挿入、欠失、付加または置換の位置は、タンパク質が、5−メチルシトシン(5mC)の脱メチル化または滑膜線維芽細胞の浸潤を促進し得る限り、特に限定されない。
 ここでアミノ酸の欠失、付加、挿入または置換を人為的に行う場合の手法としては、例えば、配列番号:2で示されるアミノ酸配列をコードするDNAに対して慣用の部位特異的変異導入を施し、その後このDNAを常法により発現させる手法が挙げられる。ここで部位特異的変異導入法としては、例えば、アンバー変異を利用する方法(ギャップド・デュプレックス法、Nucleic Acids Res.,12,9441−9456(1984))、変異導入用プライマーを用いたPCRによる方法等が挙げられる。
 Tet 3の好ましい例としては、例えば、配列番号:2で表されるアミノ酸配列からなるヒトタンパク質(Genbank Accession No.043151)、あるいは他の哺乳動物におけるそのオルソログ、アレル変異体、多型バリアント〔例えば一塩基多型(SNPs)〕などがあげられる。
 「Tet 3をコードする核酸」は、上記(a)~(e)で示される、配列番号:2で表されるアミノ酸配列又はこれと実質的に同一のアミノ酸配列をコードする塩基配列を含む核酸を表す。具体的には、以下の(f)~(j):
(f)配列番号:2で示されるアミノ酸配列をコードする塩基配列、
(g)配列番号:2で示されるアミノ酸配列において、1もしくは複数のアミノ酸が欠失、付加、挿入もしくは置換され、かつ5−メチルシトシン(5mC)の脱メチル化活性または滑膜線維芽細胞の浸潤の促進活性を有するアミノ酸配列をコードする塩基配列、
(h)配列番号:2で示されるアミノ酸配列と90%以上の相同性を有し、かつ5−メチルシトシン(5mC)の脱メチル化活性または滑膜線維芽細胞の浸潤の促進活性を有するアミノ酸配列をコードする塩基配列、
(i)配列番号1で示される塩基配列、
(j)配列番号1で示される塩基配列の相補鎖配列を有するDNAとストリンジェントな条件下でハイブリダイズする塩基配列であって、かつ5−メチルシトシン(5mC)の脱メチル化活性または滑膜線維芽細胞の浸潤の促進活性を有するアミノ酸配列をコードする塩基配列、
を有する核酸が挙げられる。
 尚、ここで遺伝子とは、cDNAもしくはゲノムDNA等のDNA、またはmRNA等のRNAのいずれでもよく、また一本鎖の核酸配列および二本鎖の核酸配列を共に含む概念である。また、本明細書において、配列番号:1等に示される核酸配列は、便宜的にDNA配列であるが、mRNAなどRNA配列を示す場合には、チミン(T)をウラシル(U)として解する。
 Tet 3をコードする核酸の好ましい例としては、例えば、配列番号:1で表される塩基配列からなるヒトTet 3 cDNA(Genbank Accession No.NM_001287491)、あるいは他の哺乳動物におけるそのオルソログ、アレル変異体、多型バリアント〔例えば一塩基多型(SNPs)〕などがあげられる。
 本発明は、Tet 3の発現を阻害する物質を含有してなる、関節炎の予防及び/又は治療剤を提供する。
II.Tet 3の発現を阻害する物質
 本発明において「Tet3の発現を阻害する物質」とは、Tet3をコードする核酸(Tet3遺伝子)の転写レベル、転写後調節のレベル、Tet 3タンパク質への翻訳レベル、翻訳後修飾のレベル等のいかなる段階で作用するものであってもよい。従って、Tet3の発現を阻害する物質としては、例えば、Tet3遺伝子の転写を阻害する物質(例、アンチジーン)、初期転写産物からmRNAへのプロセッシングを阻害する物質、mRNAの細胞質への輸送を阻害する物質、mRNAからTet 3の翻訳を阻害するか(例、アンチセンス核酸、miRNA)あるいはmRNAを分解する物質(例、siRNA、リボザイム)、初期翻訳産物の翻訳後修飾を阻害する物質などが含まれる。いずれの段階で作用するものであっても好ましく用いることができるが、より好ましくは、以下の(1)~(3)からなる群より選択される物質が例示される。
(1)Tet 3遺伝子の転写産物に対するアンチセンス核酸、
(2)Tet 3遺伝子の転写産物に対するリボザイム核酸、
(3)Tet3遺伝子の転写産物に対してRNAi活性を有する核酸もしくはその前駆体。
 ここで転写産物の好ましい例としては、mRNAが挙げられる。
 Tet 3遺伝子のmRNAからTet 3への翻訳を特異的に阻害する(あるいはmRNAを分解する)物質として、好ましくは、これらのmRNAの塩基配列と相補的もしくは実質的に相補的な塩基配列またはその一部を含む核酸が挙げられる。
 Tet 3遺伝子のmRNAの塩基配列と実質的に相補的な塩基配列とは、投与対象となる哺乳動物におけるTet 3産生細胞(例、滑膜線維芽細胞、滑膜表層細胞)の生理的条件下において、該mRNAの標的配列に結合してその翻訳を阻害し得る(あるいは該標的配列を切断する)程度の相補性を有する塩基配列を意味し、具体的には、例えば、該mRNAの塩基配列と完全相補的な塩基配列(すなわち、mRNAの相補鎖の塩基配列)と、オーバーラップする領域に関して、約90%以上、好ましくは約95%以上、より好ましくは約97%以上の相同性を有する塩基配列である。
 本発明における「塩基配列の相同性」は、相同性計算アルゴリズムNCBI BLAST(National Center for Biotechnology Information Basic Local Alignment Search Tool)を用い、以下の条件(期待値=10;ギャップを許す;フィルタリング=ON;マッチスコア=1;ミスマッチスコア=−3)にて計算することができる。
 より具体的には、Tet 3遺伝子のmRNAの塩基配列と相補的もしくは実質的に相補的な塩基配列としては、以下の(k)または(l):
(k)配列番号:1で表される塩基配列と相補的もしくは実質的に相補的な塩基配列;
(l)配列番号:1で表される塩基配列の相補鎖配列とストリンジェントな条件下でハイブリダイズする塩基配列であって、かつ5−メチルシトシン(5mC)の脱メチル化活性または滑膜線維芽細胞の浸潤の促進活性を有するタンパク質をコードする配列と、相補的もしくは実質的に相補的な塩基配列;
が挙げられる。
 ストリンジェントな条件は、前述のとおりである。
 Tet 3遺伝子のmRNAの好ましい例としては、配列番号:1で表される塩基配列(Genbank Accession No.NM_001287491)を含むヒトTet 3のmRNA、あるいは他の哺乳動物におけるそれらのオルソログ、さらにはそれらのスプライスバリアント、アレル変異体、多型バリアント等が挙げられる。
 Tet 3遺伝子のmRNAの塩基配列と「相補的もしくは実質的に相補的な塩基配列の一部」とは、Tet 3遺伝子のmRNAに特異的に結合することができ、且つ該mRNAからのタンパク質の翻訳を阻害(あるいは該mRNAを分解)し得るものであれば、その長さや位置に特に制限はないが、配列特異性の面から、標的配列に相補的もしくは実質的に相補的な部分を少なくとも10塩基以上、好ましくは約15塩基以上を含むものである。
 具体的には、Tet 3遺伝子のmRNAの塩基配列と相補的もしくは実質的に相補的な塩基配列またはその一部を含む核酸として、以下の(1)~(3)のいずれかのものが好ましく例示される:
(1)Tet 3遺伝子のmRNAに対するアンチセンス核酸、
(2)Tet 3遺伝子のmRNAに対するリボザイム核酸、
(3)Tet 3遺伝子のmRNAに対してRNAi活性を有する核酸もしくはその前駆体。
(1)Tet 3遺伝子のmRNAに対するアンチセンス核酸
 本発明における「Tet3遺伝子のmRNAに対するアンチセンス核酸」とは、該mRNAの塩基配列と相補的もしくは実質的に相補的な塩基配列またはその一部を含む核酸であって、標的mRNAと特異的かつ安定した二重鎖を形成して結合することにより、タンパク質合成を抑制する機能を有するものである。
 アンチセンス核酸は、2−デオキシ−D−リボースを含有しているポリデオキシリボヌクレオチド、D−リボースを含有しているポリリボヌクレオチド、プリンまたはピリミジン塩基のN−グリコシドであるその他のタイプのポリヌクレオチド、非ヌクレオチド骨格を有するその他のポリマー(例えば、市販のタンパク質核酸および合成配列特異的な核酸ポリマー)または特殊な結合を含有するその他のポリマー(但し、該ポリマーはDNAやRNA中に見出されるような塩基のペアリングや塩基の付着を許容する配置をもつヌクレオチドを含有する)などが挙げられる。それらは、二本鎖DNA、一本鎖DNA、二本鎖RNA、一本鎖RNA、DNA:RNAハイブリッドであってもよく、さらに非修飾ポリヌクレオチド(または非修飾オリゴヌクレオチド)、公知の修飾の付加されたもの、例えば当該分野で知られた標識のあるもの、キャップの付いたもの、メチル化されたもの、1個以上の天然のヌクレオチドを類縁物で置換したもの、分子内ヌクレオチド修飾のされたもの、例えば非荷電結合(例えば、メチルホスホネート、ホスホトリエステル、ホスホルアミデート、カルバメートなど)を持つもの、電荷を有する結合または硫黄含有結合(例、ホスホロチオエート、ホスホロジチオエートなど)を持つもの、例えばタンパク質(例、ヌクレアーゼ、ヌクレアーゼ・インヒビター、トキシン、抗体、シグナルペプチド、ポリ−L−リジンなど)や糖(例、モノサッカライドなど)などの側鎖基を有しているもの、インターカレント化合物(例、アクリジン、ソラレンなど)を持つもの、キレート化合物(例えば、金属、放射活性をもつ金属、ホウ素、酸化性の金属など)を含有するもの、アルキル化剤を含有するもの、修飾された結合を持つもの(例えば、αアノマー型の核酸など)であってもよい。ここで「ヌクレオシド」、「ヌクレオチド」および「核酸」とは、プリンおよびピリミジン塩基を含有するのみでなく、修飾されたその他の複素環型塩基をもつようなものを含んでいて良い。このような修飾物は、メチル化されたプリンおよびピリミジン、アシル化されたプリンおよびピリミジン、あるいはその他の複素環を含むものであってよい。修飾されたヌクレオシドおよび修飾されたヌクレオチドはまた糖部分が修飾されていてよく、例えば、1個以上の水酸基がハロゲンとか、脂肪族基などで置換されていたり、またはエーテル、アミンなどの官能基に変換されていたりしてよい。
 上記の通り、アンチセンス核酸はDNAであってもRNAであってもよく、あるいはDNA/RNAキメラであってもよい。アンチセンス核酸がDNAの場合、標的RNAとアンチセンスDNAとによって形成されるRNA:DNAハイブリッドは、内在性RNase Hに認識されて標的RNAの選択的な分解を引き起こすことができる。したがって、RNase Hによる分解を指向するアンチセンスDNAの場合、標的配列は、mRNA中の配列だけでなく、Tet3遺伝子の初期翻訳産物におけるイントロン領域の配列であってもよい。イントロン配列は、ゲノム配列と、Tet3遺伝子のcDNA塩基配列とをBLAST、FASTA等のホモロジー検索プログラムを用いて比較することにより、決定することができる。
 本発明のアンチセンス核酸の標的領域は、該アンチセンス核酸がハイブリダイズすることにより、結果としてタンパク質:Tet 3への翻訳が阻害されるものであればその長さに特に制限はなく、Tet 3をコードするmRNAの全配列であっても部分配列であってもよく、短いもので約10塩基程度、長いものでmRNAもしくは初期転写産物の全配列が挙げられる。合成の容易さや抗原性、細胞内移行性の問題等を考慮すれば、約10~約40塩基、特に約15~約30塩基からなるオリゴヌクレオチドが好ましいが、それに限定されない。具体的には、Tet3遺伝子の5’端ヘアピンループ、5’端6−ベースペア・リピート、5’端非翻訳領域、翻訳開始コドン、タンパク質コード領域、ORF翻訳終止コドン、3’端非翻訳領域、3’端パリンドローム領域または3’端ヘアピンループなどが、アンチセンス核酸の好ましい標的領域として選択しうるが、それらに限定されない。
 さらに、本発明のアンチセンス核酸は、Tet 3遺伝子のmRNAや初期転写産物とハイブリダイズしてタンパク質への翻訳を阻害するだけでなく、二本鎖DNAであるこれらの遺伝子と結合して三重鎖(トリプレックス)を形成し、RNAへの転写を阻害し得るもの(アンチジーン)であってもよい。
 アンチセンス核酸を構成するヌクレオチド分子は、天然型のDNAもしくはRNAでもよいが、安定性(化学的および/または対酵素)や比活性(RNAとの親和性)を向上させるために、種々の化学修飾を含むことができる。例えば、ヌクレアーゼなどの加水分解酵素による分解を防ぐために、アンチセンス核酸を構成する各ヌクレオチドのリン酸残基(ホスフェート)を、例えば、ホスホロチオエート(PS)、メチルホスホネート、ホスホロジチオネートなどの化学修飾リン酸残基に置換することができる。また、各ヌクレオチドの糖(リボース)の2’位の水酸基を、−OR(R=CH(2’−O−Me)、CHCHOCH(2’−O−MOE)、CHCHNHC(NH)NH、CHCONHCH、CHCHCN等)に置換してもよい。さらに、塩基部分(ピリミジン、プリン)に化学修飾を施してもよく、例えば、ピリミジン塩基の5位へのメチル基やカチオン性官能基の導入、あるいは2位のカルボニル基のチオカルボニルへの置換などが挙げられる。
 RNAの糖部のコンフォメーションはC2’−endo(S型)とC3’−endo(N型)の2つが支配的であり、一本鎖RNAではこの両者の平衡として存在するが、二本鎖を形成するとN型に固定される。したがって、標的RNAに対して強い結合能を付与するために、2’酸素と4’炭素を架橋することにより、糖部のコンフォメーションをN型に固定したRNA誘導体であるBNA(LNA)(Imanishi,T.et al.,Chem.Commun.,1653−9,2002;Jepsen,J.S.et al.,Oligonucleotides,14,130−46,2004)やENA(Morita,K.et al.,Nucleosides Nucleotides Nucleic Acids,22,1619−21,2003)もまた、好ましく用いられ得る。
 本発明のアンチセンスオリゴヌクレオチドは、Tet 3遺伝子のcDNA配列もしくはゲノミックDNA配列に基づいてmRNAもしくは初期転写産物の標的配列を決定し、市販のDNA/RNA自動合成機(アプライド・バイオシステムズ社、ベックマン社等)を用いて、これに相補的な配列を合成することにより調製することができる。また、上記した各種修飾を含むアンチセンス核酸も、いずれも自体公知の手法により、化学的に合成することができる。
(2)Tet 3遺伝子のmRNAに対するリボザイム核酸
 Tet 3遺伝子のmRNAの塩基配列と相補的もしくは実質的に相補的な塩基配列またはその一部を含む核酸の他の好ましい例としては、該mRNAをコード領域の内部で特異的に切断し得るリボザイム核酸が挙げられる。「リボザイム」とは、狭義には、核酸を切断する酵素活性を有するRNAをいうが、本明細書では配列特異的な核酸切断活性を有する限りDNAをも包含する概念として用いるものとする。リボザイム核酸として最も汎用性の高いものとしては、ウイロイドやウイルソイド等の感染性RNAに見られるセルフスプライシングRNAがあり、ハンマーヘッド型やヘアピン型等が知られている。ハンマーヘッド型は約40塩基程度で酵素活性を発揮し、ハンマーヘッド構造をとる部分に隣接する両端の数塩基ずつ(合わせて約10塩基程度)をmRNAの所望の切断部位と相補的な配列にすることにより、標的mRNAのみを特異的に切断することが可能である。このタイプのリボザイム核酸は、RNAのみを基質とするので、ゲノムDNAを攻撃することがないというさらなる利点を有する。Tet 3遺伝子のmRNAが自身で二本鎖構造をとる場合には、RNAヘリカーゼと特異的に結合し得るウイルス核酸由来のRNAモチーフを連結したハイブリッドリボザイムを用いることにより、標的配列を一本鎖にすることができる[Proc.Natl.Acad.Sci.USA,98(10):5572−5577(2001)]。さらに、リボザイムを、それをコードするDNAを含む発現ベクターの形態で使用する場合には、転写産物の細胞質への移行を促進するために、tRNAを改変した配列をさらに連結したハイブリッドリボザイムとすることもできる[Nucleic Acids Res.,29(13):2780−2788(2001)]。
(3)Tet 3遺伝子のmRNAに対するsiRNA
 本明細書においては、Tet 3遺伝子のmRNAに相補的なオリゴRNAとその相補鎖とからなる二本鎖RNA、いわゆるsiRNAもまた、Tet 3遺伝子のmRNAの塩基配列と相補的もしくは実質的に相補的な塩基配列またはその一部を含む核酸に包含されるものとして定義される。短い二本鎖RNAを細胞内に導入するとそのRNAに相補的なmRNAが分解される、いわゆるRNA干渉(RNAi)と呼ばれる現象は、以前から線虫、昆虫、植物等で知られていたが、この現象が動物細胞でも広く起こることが確認されて以来[Nature,411(6836):494−498(2001)]、上記のアンチセンス核酸やリボザイムの代替技術として汎用されている。
 siRNAは、標的遺伝子のcDNA配列情報に基づいて、例えば、Elbashirら(Genes Dev.,15,188−200(2001))、Teramotoら(FEBS Lett.579(13):p2878−82(2005))の提唱する規則に従って設計することができる。siRNAの標的配列は、原則的には15~50塩基、好ましくは19~49塩基、更に好ましくは19~27塩基の長さを有しており、例えばAA+(N)19(AAに続く、19塩基の塩基配列)、AA+(N)21(AAに続く、21塩基の塩基配列)もしくはA+(N)21(Aに続く、21塩基の塩基配列)であってもよい。
 本発明の核酸は、5’または3’末端に、付加的な塩基を有していてもよい。該付加的塩基の長さは、通常2~4塩基程度であり、siRNAの全長として19塩基以上である。該付加的塩基は、DNAでもRNAでもよいが、DNAを用いると核酸の安定性を向上させることができる場合がある。このような付加的塩基の配列としては、例えばug−3’、uu−3’、tg−3’、tt−3’、ggg−3’、guuu−3’、gttt−3’、ttttt−3’、uuuuu−3’などの配列が挙げられるが、これらに限定されるものではない。
 また、siRNAは、3’末端に突出部配列(オーバーハング)を有していてもよく、具体的には、dTdT(dTはデオキシリボ核酸のデオキシチミジン残基を表わす)を付加したものが挙げられる。また、末端付加がない平滑末端(ブラントエンド)であってもよい。
 また、siRNAは、センス鎖とアンチセンス鎖が異なる塩基数であってもよく、例えば、アンチセンス鎖が3’末端および5’末端に突出部配列(オーバーハング)を有している「aiRNA」を挙げることができる。典型的なaiRNAは、アンチセンス鎖が21塩基からなり、センス鎖が15塩基からなり、アンチセンス鎖の両端で各々3塩基のオーバーハング構造をとる
(Sun,X.ら著、Nature Biotechnology Vol26 No.12 p1379、国際公開第WO2009/029688号パンフレット)。
 標的配列の位置は特に制限されるわけではないが、5’−UTRおよび開始コドンから約50塩基まで、並びに3’−UTR以外の領域から標的配列を選択することが望ましい。上述の規則その他に基づいて選択された標的配列の候補群について、標的以外のmRNAにおいて16−17塩基の連続した配列に相同性がないかどうかを、BLAST(http://www.ncbi.nlm.nih.gov/BLAST/)等のホモロジー検索ソフトを用いて調べ、選択した標的配列の特異性を確認する。特異性の確認された標的配列について、AA(もしくはNA)以降の19−21塩基にTTもしくはUUの3’末端オーバーハングを有するセンス鎖と、該19−21塩基に相補的な配列およびTTもしくはUUの3’末端オーバーハングを有するアンチセンス鎖とからなる2本鎖RNAをsiRNAとして設計してもよい。また、siRNAの前駆体であるショートヘアピンRNA(shRNA)は、ループ構造を形成しうる任意のリンカー配列(例えば、5−25塩基程度)を適宜選択し、上記センス鎖とアンチセンス鎖とを該リンカー配列を介して連結することにより設計することができる。
 siRNAおよび/またはshRNAの配列は、種々のwebサイト上に無料で提供される検索ソフトを用いて検索が可能である。このようなサイトとしては、例えば、Ambionが提供するsiRNA Target Finder
(http://www.ambion.com/jp/techlib/misc/siRNA_finder.html)およびpSilencer(登録商標)Expression Vector用インサートデザインツール
(http://www.ambion.com/jp/techlib/misc/psilencer_converter.html)、RNAi Codexが提供するGeneSeer
(http://codex.cshl.edu/scripts/newsearchhairpin.cgi)があるがこれらに限定されない。
 siRNAを構成するリボヌクレオシド分子もまた、安定性、比活性などを向上させるために、上記のアンチセンス核酸の場合と同様の修飾を受けていてもよい。但し、siRNAの場合、天然型RNA中のすべてのリボヌクレオシド分子を修飾型で置換すると、RNAi活性が失われる場合があるので、RISC複合体が機能できる最小限の修飾ヌクレオシドの導入が必要である。
 当該修飾として具体的には、siRNAを構成するヌクレオチド分子の一部を、天然型のDNAや、安定性(化学的および/または対酵素)や比活性(RNAとの親和性)を向上させるために、種々の化学修飾を施したRNAに置換することができる(Usman and Cedergren,1992,TIBS 17,34;Usman et al.,1994,Nucleic Acids Symp.Ser.31,163を参照)。例えば、ヌクレアーゼなどの加水分解酵素による分解を防ぐために、siRNAを構成する各ヌクレオチドのリン酸残基(ホスフェート)を、例えば、ホスホロチオエート(PS)、メチルホスホネート、ホスホロジチオネートなどの化学修飾リン酸残基に置換することができる。また、各ヌクレオチドの糖(リボース)の2’位の水酸基を、−OR(R=CH(2’−O−Me)、CHCHOCH(2’−O−MOE)、CHCHNHC(NH)NH、CHCONHCH、CHCHCN等)、フッ素原子(−F)に置換してもよい。さらに、塩基部分(ピリミジン、プリン)に化学修飾を施してもよく、例えば、ピリミジン塩基の5位へのメチル基やカチオン性官能基の導入、あるいは2位のカルボニル基のチオカルボニルへの置換などが挙げられる。その他上記(1)に記載されたアンチセンス核酸における修飾方法を用いることができる。あるいは、siRNAにおけるRNAの一部をDNAに置換する化学修飾(2’−デオキシ化、2’−H)を施してもよい。また、糖(リボース)の2’位と4’位を−O−CH−で架橋しコンフォメーションをN型に固定した人工核酸(LNA:Locked Nucleic Acid)を用いてもよい。
 また、siRNAを構成するセンス鎖およびアンチセンス鎖は、リンカーを介し、細胞表層に存在する受容体を特異的に認識するリガンド、ペプチド、糖鎖、抗体、脂質や正電荷や分子構造的に細胞膜表層に吸着し貫通するオリゴアルギニン、Tatペプチド、RevペプチドまたはAntペプチドなどと化学結合していてもよい。
 siRNAは、mRNA上の標的配列のセンス鎖およびアンチセンス鎖をDNA/RNA自動合成機でそれぞれ合成し、適当なアニーリング緩衝液中、約90~約95℃で約1分程度変性させた後、約30~約70℃で約1~約8時間アニーリングさせることにより調製することができる。また、siRNAの前駆体となるショートヘアピンRNA(shRNA)を合成し、これを、ダイサー(dicer)を用いて切断することにより調製することもできる。
 本明細書においては、生体内でTet3遺伝子のmRNAに対するsiRNAを生成し得るようにデザインされた核酸もまた、Tet 3遺伝子のmRNAの塩基配列と相補的もしくは実質的に相補的な塩基配列またはその一部を含む核酸に包含されるものとして定義される。そのような核酸としては、上記したshRNAやsiRNAを発現するように構築された発現ベクターなどが挙げられる。shRNAは、mRNA上の標的配列のセンス鎖およびアンチセンス鎖を適当なループ構造を形成しうる長さ(例えば5~25塩基程度)のスペーサー配列を間に挿入して連結した塩基配列を含むオリゴRNAをデザインし、これをDNA/RNA自動合成機で合成することにより調製することができる。shRNAを発現するベクターには、タンデムタイプとステムループ(ヘアピン)タイプとがある。前者はsiRNAのセンス鎖の発現カセットとアンチセンス鎖の発現カセットをタンデムに連結したもので、細胞内で各鎖が発現してアニーリングすることにより2本鎖のsiRNA(dsRNA)を形成するというものである。一方、後者はshRNAの発現カセットをベクターに挿入したもので、細胞内でshRNAが発現しdicerによるプロセシングを受けてdsRNAを形成するというものである。プロモーターとしては、polII系プロモーター(例えば、CMV前初期プロモーター)を使用することもできるが、短いRNAの転写を正確に行わせるために、polIII系プロモーターを使用するのが一般的である。polIII系プロモーターとしては、マウスおよびヒトのU6−snRNAプロモーター、ヒトH1−RNase P RNAプロモーター、ヒトバリン−tRNAプロモーターなどが挙げられる。また、転写終結シグナルとして4個以上Tが連続した配列が用いられる。
 このようにして構築したsiRNAもしくはshRNA発現カセットを、次いでプラスミドベクターやウイルスベクターに挿入する。このようなベクターとしては、レトロウイルス、レンチウイルス、アデノウイルス、アデノ随伴ウイルス、ヘルペスウイルス、センダイウイルスなどのウイルスベクターや、動物細胞発現プラスミドなどが用いられる。
 上記siRNAは、ヌクレオチド配列の情報に基づいて、例えば394 Applied Biosystems,Inc.合成機等のDNA/RNA自動合成機を用いて常法に従って化学的に合成することができる。例えば、Caruthers et al.,1992,Methods in Enzymology 211,3−19、Thompson et al.,国際公開99/54459、Wincott et al.,1995,Nucleic Acids Res.23,2677−2684、Wincott et al.,1997,Methods Mol.Bio.,74,59、Brennan et al.,1998,Biotechnol Bioeng.,61,33−45、Usman et al.,1987
J.Am.Chem.Soc.,109,7845、Scaringe et al.,1990 Nucleic Acids Res.,18,5433、および米国特許第6001311号に記載される方法等が挙げられる。具体的には、当業者に公知の核酸保護基(例えば5’末端にジメトキシトリチル基)およびカップリング基(例えば3’末端にホスホルアミダイト)を用いて合成できる。すなわち、5’末端の保護基を、TCA(トリクロロ酢酸)等の酸で脱保護し、カップリング反応を行う。ついでアセチル基でキャッピングを行った後、次の核酸の縮合反応を行う。修飾されたRNAやDNAを含むsiRNAの場合には、原料として修飾されたRNA(例えば、2’−O−メチルヌクレオチド、2’−デオキシ−2’−フルオロヌクレオチド)を用いればよく、カップリング反応の条件は適宜調整できる。また、リン酸結合部分が修飾されたホスホロチオエート結合を導入する場合には、ボーケージ試薬(3H−1,2−ベンゾジチオール−3−オン1,1−ジオキシド)を用いることができる。
 あるいは、オリゴヌクレオチドは、別々に合成し、合成後に例えばライゲーションにより一緒につなげてもよいし(Moore et al.,1992,Science 256,9923;Draper et al.国際公開WO93/23569;Shabarova et al.,1991,Nucleic Acids Research 19,4247;Bellon et al.,1997,Nucleosides&Nucleotides,16,951:Bellon et al.,1997,Nucleosides&Nucleotides,Bellon et al.,1997,Bioconjugate Chem.8,204)、または合成および/または脱保護の後にハイブリダイゼーションにより、一緒につなげてもよい。siRNA分子はまたタンデム合成法により合成することができる。すなわち、両方のsiRNA鎖を、切断可能なリンカーにより分離された単一の連続するオリゴヌクレオチドとして合成し、次にこれを切断して別々のsiRNAフラグメントを生成し、これをハイブリダイズさせて精製する。リンカーはポリヌクレオチドリンカーであっても非ヌクレオチドリンカーであってもよい。
 合成したsiRNA分子は、当業者に公知の方法を用いて精製できる。例えばゲル電気泳動により精製する方法、または高速液体クロマトグラフィー(HPLC)を用いて精製する方法が挙げられる。
 本発明において、Tet 3遺伝子に対するsiRNAを構成するTet 3遺伝子のmRNAに相補的なオリゴRNAおよびその相補鎖としては、それぞれ配列番号:3を含む配列および配列番号:4を含む配列が挙げられ、好ましくは、それぞれ配列番号:3からなる配列および配列番号:4からなる配列が挙げられる。
 Tet 3遺伝子のmRNAの塩基配列と相補的もしくは実質的に相補的な塩基配列またはその一部を含む核酸の別の好ましい例として、該mRNAを標的とするmicro RNA(miRNA)が挙げられる。ヒトTet 3 mRNAを標的とするヒトmiRNAとしては、例えば、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等が挙げられる。miRNAも、上述のsiRNAについて記載した方法に準じて調製することができる。
 Tet 3遺伝子のmRNAの塩基配列と相補的もしくは実質的に相補的な塩基配列またはその一部を含む核酸は、リポソーム、ミクロスフェアのような特殊な形態で提供されたり、他の分子が付加された形態で提供されうる。付加形態で用いられるものとしては、リン酸基骨格の電荷を中和するように働くポリリジンのようなポリカチオン体、細胞膜との相互作用を高めたり、核酸の取込みを増大させたりするような脂質(例、ホスホリピド、コレステロールなど)などの疎水性のものが挙げられる。付加するに好ましい脂質としては、コレステロールやその誘導体(例、コレステリルクロロホルメート、コール酸など)が挙げられる。こうしたものは、核酸の3’端または5’端に付着させることができ、塩基、糖、分子内ヌクレオシド結合を介して付着させることができうる。その他の基としては、核酸の3’端または5’端に特異的に配置されたキャップ用の基で、エキソヌクレアーゼ、RNaseなどのヌクレアーゼによる分解を阻止するためのものが挙げられる。こうしたキャップ用の基としては、ポリエチレングリコール、テトラエチレングリコールなどのグリコールをはじめとした当該分野で知られた水酸基の保護基が挙げられるが、それに限定されるものではない。
 これらの核酸のTet 3発現阻害活性は、Tet 3をコードする核酸を導入した形質転換体、生体内や生体外のTet 3遺伝子発現系、または生体内や生体外のTet 3タンパク質の翻訳系を用いて調べることができる。
 本発明におけるTet3の発現を阻害する物質は、上記のようなTet3遺伝子のmRNAの塩基配列と相補的もしくは実質的に相補的な塩基配列またはその一部を含む核酸に限定されず、Tet 3の産生を直接的または間接的に阻害する限り、低分子化合物などの他の物質であってもよい。そのような物質は、例えば、後述する本発明のスクリーニング方法により取得することができる。
 Tet 3の発現を阻害する物質は、Tet 3の有する5−メチルシトシン(5mC)の脱メチル化活性または滑膜線維芽細胞の浸潤の促進活性を抑制する活性を示すことから、関節炎の予防及び/又は治療に有用である。
 従って、Tet 3の発現を阻害する物質を含有する医薬は、関節炎の予防及び/又は治療剤として使用することができる。
III.アンチセンス核酸、リボザイム核酸、siRNAおよびその前駆体を含有する医薬
 Tet 3遺伝子の転写産物に相補的に結合し、該転写産物からのタンパク質の翻訳を抑制することができる本発明のアンチセンス核酸や、Tet 3遺伝子の転写産物(mRNA)と相同な(もしくは相補的な)塩基配列を有し、当該転写産物を標的として該転写産物を切断し得るsiRNA(もしくはリボザイム)、さらに該siRNAの前駆体であるshRNAなど(以下、包括的に「本発明の核酸」という場合がある)は、生体内におけるTet3の発現を抑制し、5−メチルシトシン(5mC)の脱メチル化または滑膜線維芽細胞の浸潤の促進活性を抑制するので、関節炎の予防及び/又は治療剤として使用することができる。
 本発明の核酸を含有する医薬は低毒性であり、そのまま液剤として、または適当な剤型の医薬組成物として、ヒトまたは非ヒト哺乳動物(例、ラット、ウサギ、ヒツジ、ブタ、ウシ、ネコ、イヌ、サルなど)に対して経口的または非経口的(例、血管内投与、皮下投与など)に投与することができる。
 本発明の核酸を上記の関節炎の予防及び/又は治療剤として使用する場合、自体公知の方法に従って製剤化し、投与することができる。即ち、本発明の核酸を、単独あるいはレトロウイルスベクター、アデノウイルスベクター、アデノウイルスアソシエーテッドウイルスベクターなどの適当な哺乳動物細胞用の発現ベクターに機能可能な態様で挿入した後、常套手段に従って製剤化することができる。該核酸は、そのままで、あるいは摂取促進のための補助剤とともに、遺伝子銃やハイドロゲルカテーテルのようなカテーテルによって投与することができる。あるいは、エアロゾル化して吸入剤として気管内に局所投与することもできる。
 さらに、体内動態の改良、半減期の長期化、細胞内取り込み効率の改善を目的に、前記核酸を単独またはリポソームなどの担体とともに製剤(注射剤)化し、静脈、皮下等に投与してもよい。
 本発明の核酸は、それ自体を投与してもよいし、または適当な医薬組成物として投与してもよい。投与に用いられる医薬組成物としては、本発明の核酸と薬理学的に許容され得る担体、希釈剤もしくは賦形剤とを含むものであってよい。このような医薬組成物は、経口または非経口投与に適する剤形として提供される。
 非経口投与のための組成物としては、例えば、注射剤、坐剤等が用いられ、注射剤は静脈注射剤、皮下注射剤、皮内注射剤、筋肉注射剤、点滴注射剤等の剤形を包含しても良い。このような注射剤は、公知の方法に従って調製できる。注射剤の調製方法としては、例えば、上記本発明の核酸を通常注射剤に用いられる無菌の水性液、または油性液に溶解、懸濁または乳化することによって調製できる。注射用の水性液としては、例えば、生理食塩水、ブドウ糖やその他の補助薬を含む等張液等が用いられ、適当な溶解補助剤、例えば、アルコール(例、エタノール)、ポリアルコール(例、プロピレングリコール、ポリエチレングリコール)、非イオン界面活性剤〔例、ポリソルベート80、HCO−50(polyoxyethylene(50mol)adduct of hydrogenated castor oil)〕等と併用してもよい。油性液としては、例えば、ゴマ油、大豆油等が用いられ、溶解補助剤として安息香酸ベンジル、ベンジルアルコール等を併用してもよい。調製された注射液は、適当なアンプルに充填されることが好ましい。直腸投与に用いられる坐剤は、上記核酸を通常の坐薬用基剤に混合することによって調製されてもよい。
 経口投与のための組成物としては、固体または液体の剤形、具体的には錠剤(糖衣錠、フィルムコーティング錠を含む)、丸剤、顆粒剤、散剤、カプセル剤(ソフトカプセル剤を含む)、シロップ剤、乳剤、懸濁剤等が挙げられる。このような組成物は公知の方法によって製造され、製剤分野において通常用いられる担体、希釈剤もしくは賦形剤を含有していても良い。錠剤用の担体、賦形剤としては、例えば、乳糖、でんぷん、蔗糖、ステアリン酸マグネシウムが用いられる。
 上記の非経口用または経口用医薬組成物は、活性成分の投与量に適合するような投薬単位の剤形に調製されることが好都合である。このような投薬単位の剤形としては、例えば、錠剤、丸剤、カプセル剤、注射剤(アンプル)、坐剤が挙げられる。本発明の核酸は、例えば、投薬単位剤形当たり通常5~500mg、とりわけ注射剤では5~100mg、その他の剤形では10~250mg含有されていることが好ましい。
 本発明の核酸を含有する上記医薬の投与量は、投与対象、対象疾患、症状、投与ルートなどによっても異なるが、例えば、関節リウマチの治療・予防のために使用する場合には、本発明の核酸を1回量として、通常0.01~20mg/kg体重程度、好ましくは0.1~10mg/kg体重程度、さらに好ましくは0.1~5mg/kg体重程度を、1日1~5回程度、好ましくは1日1~3回程度、静脈注射により投与するのが好都合である。他の非経口投与および経口投与の場合もこれに準ずる量を投与することができる。症状が特に重い場合には、その症状に応じて増量してもよい。
 なお前記した各組成物は、本発明の核酸との配合により好ましくない相互作用を生じない限り適宜他の活性成分を含有してもよい。
 上述のTet 3に対するアンチセンス核酸、リボザイム核酸、siRNAおよびその前駆体を含有する医薬や、Tet 3の発現を抑制する低分子化合物等を含有する医薬組成物は、関節炎の治療、予防、または進行防止に用いることができる。関節炎として、具体的には、関節リウマチ、乾癬性関節炎、脊椎関節炎(例えば、強直性脊椎炎など)などが挙げられ、好ましくは、関節リウマチが挙げられる。また、Tet 3がその病態の増悪に関与しているいかなる疾患も、本発明の対象疾患に包含される。
 上述のTet 3に対するアンチセンス核酸、リボザイム核酸、siRNAおよびその前駆体を含有する医薬や、Tet 3の発現を抑制する低分子化合物等を含有する医薬組成物を関節炎の治療または予防に使用する場合には、単独で使用してもよいが、1種または2種以上の抗炎症作用を有する薬剤と併用してもよい。
 併用する薬剤としては、特に限定されるものではないが、例えば、メサラジン、副腎皮質ステロイド(例、ベタメタゾン、プレドニゾロン、ヒドロコルチゾン、デキサメタゾン等)、非ステロイド抗炎症薬(NSAIDs;例、サリチル酸系、アントラニル酸系、アリール酸系、プロピオン酸系、オキシカム系、ピリン系)、抗TNFα抗体(インフルキシマブ、アダリムマブ)、抗リウマチ薬(例、アクタリット等の免疫調節薬、メトトレキサート等の免疫抑制薬、抗TNFα抗体やエタネルセプト等の生物学的製剤)などが挙げられる。
IV.疾病に対する医薬候補化合物のスクリーニング
 上述の通り、Tet 3の発現及び/又は機能を阻害すると、5−メチルシトシン(5mC)の脱メチル化が抑制され、活性化された滑膜線維芽細胞の最も重要な攻撃的な表現型の一つである浸潤能が抑制される。このことは、Tet3の選択的阻害は、DNAメチル化レベルの維持を介してCCL2やICAM1の発現を阻害し、それに伴って浸潤性に代表される滑膜線維芽細胞の活性化を阻害することによって、関節炎の治療につながることを意味する。従って、Tet 3の発現及び/又は機能を阻害する化合物は、関節炎の予防及び/又は治療剤として使用することができる。
 したがって、Tet 3を産生する細胞は、Tet 3(又はTet 3遺伝子)の発現量及び/又は機能を指標とすることにより、関節炎の予防及び/又は治療薬のスクリーニングのためのツールとして用いることができる。
 Tet 3の発現又は機能を阻害する化合物をスクリーニングする場合、該スクリーニング方法は、Tet 3を産生する能力を有する細胞を、被検物質の存在下および非存在下に培養し、両条件下におけるTet 3の発現量又は機能の程度を比較することを含む。また、Tet 3の機能を阻害する化合物は、精製したTet 3タンパク質への結合能を試験することによっても、スクリーニングすることができる。
 上記のスクリーニング方法において用いられるTet 3を産生する能力を有する細胞としては、それらを生来発現しているヒトもしくは他の哺乳動物細胞(例:滑膜線維芽細胞、滑膜表層細胞等)またはそれを含む生体試料(例:滑膜(特に滑膜表層細胞層)等)であれば特に制限はない。非ヒト動物由来の滑膜等の場合は、それらを生体から単離して培養してもよいし、あるいは生体自体に被検物質を投与し、一定時間経過後にそれら生体試料を単離してもよい。
 また、Tet 3を産生する能力を有する細胞としては、公知慣用の遺伝子工学的手法により作製された各種の形質転換体も使用することができる。宿主としては、例えば、H4IIE−C3細胞、HepG2細胞、HEK293細胞、COS7細胞、CHO細胞などの動物細胞が好ましく用いられる。
 具体的には、Tet3をコードするDNA(即ち、配列番号:1で表される塩基配列または該塩基配列に対し相補性を有する塩基配列とストリンジェントな条件下でハイブリダイズし、且つ配列番号:2で表されるアミノ酸配列からなるタンパク質と同質の機能を有するポリペプチドをコードする塩基配列を含むDNA)を、適当な発現ベクター中のプロモーターの下流に連結して宿主動物細胞に導入することにより調製することができる。
 Tet 3をコードする遺伝子の調製方法について、以下に説明する。
 Tet 3をコードする遺伝子は、通常の遺伝子工学的方法(例えば、Sambrook J.,Frisch E.F.,Maniatis T.著、モレキュラークローニング第2版(Molecular Cloning 2nd edition)、コールド スプリング ハーバー ラボラトリー発行(Cold Spring Harbor Laboratory press)等に記載されている方法)に準じて取得することができる。すなわち、Tet 3をコードするDNAは、例えば、配列番号:1で表される塩基配列に基づいて、適当なオリゴヌクレオチドをプローブもしくはプライマーとして合成し、前記したTet 3を産生する細胞・組織由来のcDNAもしくはcDNAライブラリーから、ハイブリダイゼーション法やPCR法を用いてクローニングすることができる。ハイブリダイゼーションは、例えば、モレキュラー・クローニング(Molecular Cloning)第2版(上記)に記載の方法などに従って行なうことができる。また、市販のライブラリーを使用する場合、ハイブリダイゼーションは、該ライブラリーに添付された使用説明書に記載の方法に従って行なうことができる。
 DNAの塩基配列は、公知のキット、例えば、MutanTM−super Express Km(宝酒造(株))、MutanTM−K(宝酒造(株))等を用いて、ODA−LA PCR法、Gapped duplex法、Kunkel法等の自体公知の方法あるいはそれらに準じる方法に従って変換することができる。
 クローン化されたDNAは、目的によりそのまま、または所望により制限酵素で消化するか、リンカーを付加した後に、使用することができる。該DNAはその5’末端側に翻訳開始コドンとしてのATGを有し、また3’末端側には翻訳終止コドンとしてのTAA、TGAまたはTAGを有していてもよい。これらの翻訳開始コドンや翻訳終止コドンは、適当な合成DNAアダプターを用いて付加することができる。
 次いで、得られたTet3遺伝子を用いて、通常の遺伝子工学的方法に準じてTet3タンパク質を発現する細胞を製造・取得することができる。
 例えば、Tet 3遺伝子が宿主細胞中で発現できるようなプラスミドを作製し、これを宿主細胞に導入して形質転換し、さらに形質転換された宿主細胞(形質転換体)を培養することでTet 3タンパク質を発現する細胞を取得すればよい。上記プラスミドとしては、例えば、宿主細胞中で複製可能な遺伝情報を含み、自律的に複製できるものであって、宿主細胞からの単離・精製が容易であり、宿主細胞中で機能可能なプロモーターを有し、検出可能なマーカーをもつ発現ベクターに、Tet 3をコードする遺伝子が導入されたものを好ましく挙げることができる。尚、発現ベクターとしては、各種のものが市販されている。
 例えば、大腸菌での発現に使用される発現ベクターは、lac、trp、tacなどのプロモーターを含む発現ベクターであって、これらはファルマシア社、タカラバイオ等から市販されている。当該発現ベクターにTet 3をコードする遺伝子を導入するために用いられる制限酵素もタカラバイオ等から市販されている。さらなる高発現を導くことが必要な場合には、Tet 3をコードするDNAの上流にリボソーム結合領域を連結してもよい。用いられるリボソーム結合領域としては、Guarente L.ら(Cell 20,p543)や谷口ら(Genetics of Industrial Microorganisms,p202,講談社)による報告に記載されたものを挙げることができる。
 また、動物細胞発現プラスミド(例:pA1−11、pXT1、pRc/CMV、pRc/RSV、pcDNAI/Neo);λファージなどのバクテリオファージ;レトロウイルス、ワクシニアウイルス、アデノウイルスなどの動物ウイルスベクターなどを用いることもできる。プロモーターとしては、遺伝子の発現に用いる宿主に対応して適切なプロモーターであればいかなるものでもよい。例えば、SRαプロモーター、SV40プロモーター、LTRプロモーター、CMV(サイトメガロウイルス)プロモーター、RSV(ラウス肉腫ウイルス)プロモーター、MoMuLV(モロニーマウス白血病ウイルス)LTR、HSV−TK(単純ヘルペスウイルスチミジンキナーゼ)プロモーター、βアクチン遺伝子プロモーター、aP2遺伝子プロモーターなどが用いられる。なかでも、EF−αプロモーター、CAGプロモーター、CMVプロモーター、SRαプロモーターなどが好ましい。
 発現ベクターとしては、上記の他に、所望によりエンハンサー、スプライシングシグナル、ポリA付加シグナル、選択マーカー、SV40複製起点(以下、SV40oriと略称する場合がある)などを含有しているものを用いることができる。
 選択マーカーとしては、例えば、ジヒドロ葉酸還元酵素遺伝子(以下、dhfrと略称する場合がある、メソトレキセート(MTX)耐性)、アンピシリン耐性遺伝子(以下、ampと略称する場合がある)、ネオマイシン耐性遺伝子(以下、neoと略称する場合がある、G418耐性)等が挙げられる。特に、dhfr遺伝子欠損チャイニーズハムスター細胞を用い、dhfr遺伝子を選択マーカーとして使用する場合、チミジンを含まない培地によって目的遺伝子を選択することもできる。
 上記したTet 3をコードするDNAを含む発現ベクターで宿主を形質転換することにより、Tet 3発現細胞を製造することができる。
 宿主細胞としては、原核生物もしくは真核生物である微生物細胞、昆虫細胞または哺乳動物細胞等を挙げることができる。哺乳動物細胞としては、例えば、HepG2細胞、HEK293細胞、HeLa細胞、ヒトFL細胞、サルCOS−7細胞、サルVero細胞、チャイニーズハムスター卵巣細胞(以下、CHO細胞と略記)、dhfr遺伝子欠損CHO細胞(以下、CHO(dhfr)細胞と略記)、マウスL細胞、マウスAtT−20細胞、マウスミエローマ細胞、ラットH4IIE−C3細胞、ラットGH3細胞などが用いられ得る。
 前記のようにして得られたプラスミドは、通常の遺伝子工学的方法により前記宿主細胞に導入することができる。形質転換体の培養は、微生物培養、昆虫細胞もしくは哺乳動物細胞の培養に使用される通常の方法によって行うことができる。例えば大腸菌の場合、適当な炭素源、窒素源およびビタミン等の微量栄養物を適宜含む培地中で培養を行う。培養方法としては、固体培養、液体培養のいずれの方法でもよく、好ましくは、通気撹拌培養法等の液体培養を挙げることができる。
 形質転換は、リン酸カルシウム共沈殿法、PEG法、エレクトロポレーション法、マイクロインジェクション法、リポフェクション法などにより行うことができる。例えば、細胞工学別冊8 新細胞工学実験プロトコール,263−267(1995)(秀潤社発行)、ヴィロロジー(Virology),52巻,456(1973)に記載の方法を用いることができる。
 上記のようにして得られる形質転換細胞や生来Tet 3を産生する能力を有する哺乳動物細胞または該細胞を含む組織・臓器は、例えば、約5~20%の胎仔牛血清を含む最小必須培地(MEM) 〔Science,122巻,501(1952)〕、ダルベッコ改変イーグル培地(DMEM)〔Virology,8巻,396(1959)〕、RPMI 1640培地〔The Journal of the American Medical Association,199巻,519(1967)〕、199培地〔Proceeding of the Society for the Biological Medicine,73巻,1(1950)〕などの培地中で培養することができる。培地のpHは約6~8であるのが好ましい。培養は通常約30~40℃で行ない、必要に応じて通気や撹拌を加える。
 Tet 3タンパク質の取得は、一般のタンパク質の単離・精製に通常使用される方法を組み合わせて実施すればよい。例えば、前記の培養により得られた形質転換体を遠心分離などで除去し、培養上清からTet3を前記と同様にして精製してもよい。また、Tet 3タンパク質が前記の培養により得られた形質転換体の細胞内に蓄積する場合には、例えば、該形質転換体を遠心分離等で集めた後、細胞を破砕または溶解せしめ、必要であればタンパク質の可溶化を行い、イオン交換、疎水、ゲルろ過等の各種クロマトグラフィーを用いた工程を単独で、若しくは組み合わせることにより精製すればよい。精製されたタンパク質の高次構造を復元する操作をさらに行ってもよい。
 本発明のスクリーニングを実施するに当たり、被検物質としては、例えばタンパク質、ペプチド、非ペプチド性化合物、合成化合物、発酵生産物、細胞抽出液、植物抽出液、動物組織抽出液などが挙げられ、これらの物質は新規なものであってもよいし、公知のものであってもよい。
 また、Tet 3もしくはTet 3遺伝子の発現量を低下させる物質、またはTet 3の機能を低下させる物質を選択する際に、被検物質を接触させない対照細胞を比較対照として用いることもできる。ここで「被検物質を接触させない」とは、被検物質の代わりに被検物質と同量の溶媒(ブランク)を添加する場合や、Tet 3もしくはTet 3遺伝子の発現量またはTet 3の機能に影響を与えないネガティブコントロール物質を添加する場合も含まれる。
 被検物質の上記細胞との接触は、例えば、上記の培地や各種緩衝液(例えば、HEPES緩衝液、リン酸緩衝液、リン酸緩衝生理食塩水、トリス塩酸緩衝液、ホウ酸緩衝液、酢酸緩衝液など)の中に被検物質を添加して、細胞を一定時間インキュベートすることにより実施することができる。添加される被検物質の濃度は化合物の種類(溶解度、毒性等)により異なるが、例えば、約0.1nM~約100μMの範囲で適宜選択される。インキュベート時間としては、例えば、約10分~約24時間が挙げられる。
 Tet 3を産生する細胞が、非ヒト哺乳動物個体の形態で提供される場合、該動物個体の状態は特に制限されないが、例えば、薬剤もしくは遺伝子改変により関節炎を誘起した関節炎モデル動物(例えば、コラーゲン誘導関節炎(CIA)マウス、SKGマウス、PD−1ノックアウトマウス、K/BxNマウス、シノビオリンTgマウス等のRAモデル動物など)であってもよい。使用される動物の飼育条件に特に制限はないが、SPFグレード以上の環境下で飼育されたものであることが好ましい。被検物質の該細胞との接触は、該動物個体への被検物質の投与によって行われる。投与経路は特に制限されないが、例えば、静脈内投与、動脈内投与、皮下投与、皮内投与、腹腔内投与、経口投与、気道内投与、直腸投与等が挙げられる。投与量も特に制限はないが、例えば、1回量として約0.5~20mg/kgを、1日1~5回、好ましくは1日1~3回、1~14日間投与することができる。
 あるいは、上記のスクリーニング方法は、Tet 3を産生する能力を有する細胞に代えて、該細胞の抽出液、あるいは該細胞から単離精製したTet 3に、被検物質を接触させることにより行うこともできる。
(Tet 3遺伝子またはTet 3の発現量の測定)
 本発明は、Tet 3を産生する能力を有する細胞における該タンパク質(遺伝子)の発現を、被検物質の存在下と非存在下で比較することを特徴とする、関節炎の予防及び/又は治療薬のスクリーニング方法を提供する。本方法において用いられる細胞、被検物質の種類、被検物質と細胞との接触の態様などは、上記と同様である。
 Tet 3の発現量は、前記したTet 3をコードするDNAとストリンジェントな条件下でハイブリダイズし得る核酸、即ち、配列番号:1で表される塩基配列もしくはそれと相補的な塩基配列とストリンジェントな条件下でハイブリダイズし得る核酸(DNA)(以下、「本発明の検出用核酸」という場合がある)を用いて、Tet 3遺伝子のmRNAを検出することにより、RNAレベルで測定することができる。あるいは、該発現量は、前記したTet 3に対する抗体(以下、「本発明の検出用抗体」という場合がある)を用いて、これらのタンパク質を検出することにより、タンパク質レベルで測定することもできる。
 従って、より具体的には、本発明は、
(a)Tet 3を産生する能力を有する細胞を被検物質の存在下および非存在下に培養し、両条件下における該タンパク質をコードするmRNAの量を、本発明の検出用核酸を用いて測定、比較することを特徴とする、関節炎の予防及び/又は治療薬のスクリーニング方法、および
(b)Tet 3を産生する能力を有する細胞を被検物質の存在下および非存在下に培養し、両条件下における該タンパク質の量を、本発明の検出用抗体を用いて測定、比較することを特徴とする、関節炎の予防及び/又は治療薬のスクリーニング方法を提供する。
 すなわち、Tet 3の発現量を変化させる物質のスクリーニングは、以下のようにして行うことができる。
(i)正常あるいは疾患モデル(例えば、RAモデル動物など)非ヒト哺乳動物(例えば、マウス、ラット、ウサギ、ヒツジ、ブタ、ウシ、ネコ、イヌ、サルなど)に対して被検物質を投与し、一定時間経過した後(30分後~3日後、好ましくは1時間後~2日後、より好ましくは1時間後~24時間後)に、血液、あるいは特定の臓器(例えば、滑膜等)、あるいは臓器から単離した組織または細胞を得る。
 Tet 3のmRNAは、通常の方法により細胞等からmRNAを抽出して定量することができ、あるいは自体公知のノーザンブロット解析により定量することもできる。一方、Tet 3のタンパク質量は、ウェスタンブロット解析や以下に詳述する各種イムノアッセイ法を用いて定量することができる。
(ii)Tet 3遺伝子を発現する細胞(例えば、滑膜線維芽細胞、滑膜表層細胞、Tet3を導入した形質転換体)を上記の方法に従って作製し、常法に従って培養する際に被検物質を培地もしくは緩衝液中に添加し、一定時間インキュベート後(1日後~7日後、好ましくは1日後~3日後、より好ましくは2日後~3日後)、該細胞中に含まれるTet3あるいはそれをコードするmRNAを、上記(i)と同様にして定量、解析することができる。
 Tet3遺伝子(mRNA)の発現レベルの検出および定量は、前記細胞から調製したRNAまたはそれから転写された相補的なポリヌクレオチドを用いて、ノーザンブロット法、RT−PCR法など公知の方法で実施できる。具体的には、Tet 3遺伝子の塩基配列において連続する少なくとも15塩基を有するポリヌクレオチドおよび/またはその相補的なポリヌクレオチドをプライマーまたはプローブとして用いることによって、RNA中のTet 3遺伝子の発現の有無やその発現レベルを検出、測定することができる。そのようなプローブもしくはプライマーは、Tet 3遺伝子の塩基配列をもとに、例えばprimer 3(http://primer3.sourceforge.net/)あるいはベクターNTI(Infomax社製)を利用して設計することができる。
 ノーザンブロット法を利用する場合、前記プライマーもしくはプローブを放射性同位元素(32P、33Pなど:RI)や蛍光物質などで標識し、それを、常法に従ってナイロンメンブレン等にトランスファーした細胞由来のRNAとハイブリダイズさせた後、形成された前記プライマーもしくはプローブ(DNAまたはRNA)とRNAとの二重鎖を、前記プライマーもしくはプローブの標識物(RI若しくは蛍光物質)に由来するシグナルとして放射線検出器(BAS−1800II、富士フィルム社製)または蛍光検出器で検出、測定する方法を例示することができる。また、AlkPhos Direct Labelling and Detection System(Amersham PharamciaBiotech社製)を用いて、該プロトコールに従って前記プローブを標識し、細胞由来のRNAとハイブリダイズさせた後、前記プローブの標識物に由来するシグナルをマルチバイオイメージャーSTORM860(Amersham Pharmacia Biotech社製)で検出、測定する方法を使用することもできる。
 RT−PCR法を利用する場合は、細胞由来のRNAから常法に従ってcDNAを調製して、これを鋳型として標的のTet 3遺伝子の領域が増幅できるように、Tet 3遺伝子の配列に基づき調製した一対のプライマー(上記cDNA(−鎖)に結合する正鎖、+鎖に結合する逆鎖)をこれとハイブリダイズさせて、常法に従ってPCR法を行い、得られた増幅二本鎖DNAを検出する方法を例示することができる。なお、増幅された二本鎖DNAの検出は、上記PCRを予めRIや蛍光物質で標識しておいたプライマーを用いて行うことによって産生される標識二本鎖DNAを検出する方法、産生された二本鎖DNAを常法に従ってナイロンメンブレン等にトランスファーさせて、標識した前記プライマーをプローブとして使用してこれとハイブリダイズさせて検出する方法などを用いることができる。なお、生成された標識二本鎖DNA産物はアジレント2100バイオアナライザ(横河アナリティカルシステムズ社製)などで測定することができる。また、SYBR Green RT−PCR Reagents(Applied Biosystems社製)で該プロトコールに従ってRT−PCR反応液を調製し、ABI PRIME 7900 Sequence Detection System(Applied Biosystems社製)で反応させて、該反応物を検出することもできる。
 また、Tet 3の発現量を変化させる物質のスクリーニングは、Tet 3遺伝子の転写調節領域を用いたレポーター遺伝子アッセイで行うことも可能である。ここで、「転写調節領域」とは、通常、当該染色体遺伝子の上流数kbから数十kbの範囲を指し、例えば、(i)5’−レース法(5’−RACE法)(例えば、5’−full Race Core Kit(タカラバイオ社製)等を用いて実施されうる)、オリゴキャップ法、S1プライマーマッピング等の通常の方法により、5’末端を決定するステップ;(ii)Genome Walker Kit(クローンテック社製)等を用いて5’−上流領域を取得し、得られた上流領域について、プロモーター活性を測定するステップ;を含む手法等により同定することが出来る。
 Tet 3遺伝子の転写調節領域の下流に機能可能な形でレポータータンパク質をコードする核酸(以下、「レポーター遺伝子」という)を連結して、レポータータンパク質発現ベクターを構築する。該ベクターは当業者に公知の方法で調製すればよい。すなわち、「Molecular Cloning:A Laboratory Manual 2nd edition」(1989),Cold Spring Harbor Laboratory Press、「Current Protocols In Molecular Biology」(1987),John Wiley & Sons,Inc.等に記載される通常の遺伝子工学的手法に従って切り出されたTet 3遺伝子の転写調節領域を、レポーター遺伝子を含むプラスミド上に組み込むことができる。
 レポータータンパク質としては、β−グルクロニダーゼ(GUS)、ルシフェラーゼ、クロラムフェニコールトランスアセチラーゼ(CAT)、β−ガラクトシダーゼ(GAS)、緑色蛍光タンパク質(GFP)、黄色蛍光タンパク質(YFP)、青色蛍光タンパク質(CFP)、赤色蛍光タンパク質(RFP)等が挙げられる。
 調製したTet 3遺伝子の転写調節領域を機能可能な形で連結されてなるレポーター遺伝子を、通常の遺伝子工学的手法を用いて、当該レポーター遺伝子を導入する細胞において使用可能なベクターに挿入し、プラスミドを作製し、適当な宿主細胞へ導入することができる。ベクターに搭載される選択マーカー遺伝子に応じた選抜条件の培地で培養することにより、安定な形質転換細胞を得ることができる。あるいは、Tet 3遺伝子の転写調節領域を機能可能な形で連結されてなるレポーター遺伝子は、宿主細胞内に一過的に発現させてもよい。
 また、レポーター遺伝子の発現量を測定する方法としては、個々のレポーター遺伝子に応じた方法を利用すればよい。例えば、レポーター遺伝子としてルシフェラーゼ遺伝子を用いる場合には、前記形質転換細胞を数日間培養後、当該細胞の抽出物を得、次いで当該抽出物をルシフェリンおよびATPと反応させて化学発光させ、その発光強度を測定することによりプロモーター活性を検出することができる。この際、ピッカジーンデュアルキット(登録商標;東洋インキ製)等の市販のルシフェラーゼ反応検出キットを用いることができる。
 Tet 3のタンパク質量の測定方法としては、具体的には、例えば、
(i)本発明の検出用抗体と、試料液および標識化されたTet3とを競合的に反応させ、該抗体に結合した標識化されたタンパク質を検出することにより試料液中のTet3を定量する方法や、
(ii)試料液と、担体上に不溶化した本発明の検出用抗体および標識化された別の本発明の検出用抗体とを、同時あるいは連続的に反応させた後、不溶化担体上の標識剤の量(活性)を測定することにより、試料液中のTet 3を定量する方法等が挙げられる。
 Tet 3のタンパク質発現レベルの検出および定量は、Tet3を認識する抗体を用いたウェスタンブロット法等の公知方法に従って定量できる。ウェスタンブロット法は、一次抗体としてTet 3を認識する抗体を用いた後、二次抗体として125Iなどの放射性同位元素、蛍光物質、ホースラディッシュペルオキシダーゼ(HRP)等の酵素等で標識した一次抗体に結合する抗体を用いて標識し、これら標識物質由来のシグナルを放射線測定器(BAI−1800II:富士フィルム社製など)、蛍光検出器などで測定することによって実施できる。また、一次抗体としてTet 3を認識する抗体を用いた後、ECL Plus Western Blotting Detection System(アマシャム ファルマシアバイオテク社製)を利用して該プロトコールに従って検出し、マルチバイオメージャーSTORM860(アマシャム ファルマシアバイオテク社製)で測定することもできる。
 上記の抗体は、その形態に特に制限はなく、Tet 3を免疫原とするポリクローナル抗体であっても、またモノクローナル抗体であってもよく、さらにはTet 3を構成するアミノ酸配列のうち少なくとも連続する、通常8アミノ酸、好ましくは15アミノ酸、より好ましくは20アミノ酸からなるポリペプチドに対して抗原結合性を有する抗体を用いることもできる。
 これらの抗体の製造方法は、すでに周知であり、本発明の抗体もこれらの常法に従って製造することができる(Current protocols in Molecular Biology edit.Ausubel et al.(1987)Publish.John Wiley and Sons.Section 11.12~11.13)。
 上記(ii)の定量法においては、2種の抗体はTet 3の異なる部分を認識するものであることが望ましい。例えば、一方の抗体がTet 3のN端部を認識する抗体であれば、他方の抗体として該タンパク質のC端部と反応するものを用いることができる。
 標識物質を用いる測定法に用いられる標識剤としては、例えば、放射性同位元素、酵素、蛍光物質、発光物質などが用いられる。放射性同位元素としては、例えば、〔125I〕、〔131I〕、〔H〕、〔14C〕などが用いられる。上記酵素としては、安定で比活性の大きなものが好ましく、例えば、β−ガラクトシダーゼ、β−グルコシダーゼ、アルカリフォスファターゼ、パーオキシダーゼ、リンゴ酸脱水素酵素などが用いられる。蛍光物質としては、例えば、フルオレスカミン、フルオレッセンイソチオシアネートなどが用いられる。発光物質としては、例えば、ルミノール、ルミノール誘導体、ルシフェリン、ルシゲニンなどが用いられる。さらに、抗体あるいは抗原と標識剤との結合にビオチン−(ストレプト)アビジン系を用いることもできる。
 本発明の検出用抗体を用いるTet 3の定量法は、特に制限されるべきものではなく、試料液中の抗原量に対応した、抗体、抗原もしくは抗体−抗原複合体の量を化学的または物理的手段により検出し、これを既知量の抗原を含む標準液を用いて作製した標準曲線より算出する測定法であれば、いずれの測定法を用いてもよい。例えば、ネフロメトリー、競合法、イムノメトリック法およびサンドイッチ法が好適に用いられる。感度、特異性の点で、例えば、後述するサンドイッチ法を用いるのが好ましい。
 抗原あるいは抗体の不溶化にあたっては、物理吸着を用いてもよく、また通常タンパク質あるいは酵素等を不溶化・固定化するのに用いられる化学結合を用いてもよい。担体としては、アガロース、デキストラン、セルロースなどの不溶性多糖類、ポリスチレン、ポリアクリルアミド、シリコン等の合成樹脂、あるいはガラス等があげられる。
 サンドイッチ法においては不溶化した本発明の検出用抗体に試料液を反応させ(1次反応)、さらに標識化した別の本発明の検出用抗体を反応させた(2次反応)後、不溶化担体上の標識剤の量もしくは活性を測定することにより、試料液中のTet 3を定量することができる。1次反応と2次反応は逆の順序で行っても、また、同時に行ってもよいし、時間をずらして行ってもよい。標識化剤および不溶化の方法は前記のそれらに準じることができる。また、サンドイッチ法による免疫測定法において、固相化抗体あるいは標識化抗体に用いられる抗体は必ずしも1種類である必要はなく、測定感度を向上させる等の目的で2種類以上の抗体の混合物を用いてもよい。
 本発明の検出用抗体は、サンドイッチ法以外の測定システム、例えば、競合法、イムノメトリック法あるいはネフロメトリーなどにも用いることができる。
 競合法では、試料液中のTet 3と標識したTet 3とを抗体に対して競合的に反応させた後、未反応の標識抗原(F)と、抗体と結合した標識抗原(B)とを分離し(B/F分離)、B,Fいずれかの標識量を測定することにより、試料液中のTet 3を定量する。本反応法には、抗体として可溶性抗体を用い、ポリエチレングリコールや前記抗体(1次抗体)に対する2次抗体などを用いてB/F分離を行う液相法、および、1次抗体として固相化抗体を用いるか(直接法)、あるいは1次抗体は可溶性のものを用い、2次抗体として固相化抗体を用いる(間接法)固相化法とが用いられる。
 イムノメトリック法では、試料液中のTet 3と固相化したTet 3とを一定量の標識化抗体に対して競合反応させた後、固相と液相を分離するか、あるいは試料液中のTet 3と過剰量の標識化抗体とを反応させ、次に固相化したTet 3を加えて未反応の標識化抗体を固相に結合させた後、固相と液相を分離する。次に、いずれかの相の標識量を測定し試料液中の抗原量を定量する。
 また、ネフロメトリーでは、ゲル内あるいは溶液中で抗原抗体反応の結果生じた不溶性の沈降物の量を測定する。試料液中のTet 3の量がわずかであり、少量の沈降物しか得られない場合にもレーザーの散乱を利用するレーザーネフロメトリーなどが好適に用いられる。
 これら個々の免疫学的測定法を本発明の定量方法に適用するにあたっては、特別の条件、操作等の設定は必要とされない。それぞれの方法における通常の条件、操作法に当業者の通常の技術的配慮を加えてTet 3の測定系を構築すればよい。これらの一般的な技術手段の詳細については、総説、成書などを参照することができる。
 例えば、入江 寛編「ラジオイムノアッセイ」(講談社、昭和49年発行)、入江 寛編「続ラジオイムノアッセイ」(講談社、昭和54年発行)、石川栄治ら編「酵素免疫測定法」(医学書院、昭和53年発行)、石川栄治ら編「酵素免疫測定法」(第2版)(医学書院、昭和57年発行)、石川栄治ら編「酵素免疫測定法」(第3版)(医学書院、昭和62年発行)、「Methods in ENZYMOLOGY」Vol.70(Immunochemical Techniques(Part A))、同書Vol.73(Immunochemical Techniques(Part B))、同書Vol.74(Immunochemical Techniques(Part C))、同書Vol.84(Immunochemical Techniques(Part D:Selected Immunoassays))、同書Vol.92(Immunochemical Techniques(Part E:Monoclonal Antibodies and General Immunoassay Methods))、同書Vol.121(Immunochemical Techniques(Part I:Hybridoma Technology and Monoclonal Antibodies))(以上、アカデミックプレス社発行)などを参照することができる。
 以上のようにして、本発明の検出用抗体を用いることによって、細胞におけるTet3の量を感度よく定量することができる。
 例えば、上記スクリーニング法において、被検物質の存在下におけるTet 3の発現量(mRNA量またはタンパク質量)が、被検物質の非存在下における場合に比べて、約20%以上、好ましくは約30%以上、より好ましくは約50%以上阻害された場合、該被検物質を、Tet 3の発現阻害物質、従って、関節炎の予防及び/又は治療薬の候補として選択することができる。
 あるいは、上記スクリーニング法において、Tet 3遺伝子を発現する細胞に代えて、Tet3遺伝子の内在の転写調節領域の制御下にあるレポーター遺伝子を含む細胞を用いることができる。このような細胞は、Tet 3遺伝子の転写調節領域の制御下にあるレポーター遺伝子(例、ルシフェラーゼ、GFPなど)を導入したトランスジェニック動物の細胞、組織、臓器もしくは個体であってもよい。かかる細胞を用いる場合には、Tet 3の発現量は、レポーター遺伝子の発現レベルを、常法を用いて測定することにより評価することができる。
(Tet 3の機能の測定)
 本発明のスクリーニング方法は、被検物質がTet 3の機能を阻害するか否かを指標として行うこともできる。
 Tet 3は脱メチル化タンパク質であるので、Tet 3タンパク質に結合能を有する物質は、脱メチル化活性を抑制することにより、Tet 3の機能を阻害し得ると考えられる。従って、Tet 3への結合能を指標として、Tet 3の機能阻害物質の候補をスクリーニングすることができる。
 例えば、被検物質をウェルプレートの各ウェルに吸着させ、適当な標識剤で標識したTet 3溶液を各ウェルに添加してインキュベートした後液相を除き、洗浄後に固相に結合した標識量を測定することにより、Tet 3との結合能を有する被検物質を検出することができる。Tet 3を直接標識する代わりに、標識した抗Tet 3抗体を用いて固相に結合したTet 3を検出することもできる。あるいは、Tet 3を固定化した担体(例、アフィニティーカラム)に被検物質の溶液を通し、該担体に保持された被検物質を、Tet 3との結合能を有する物質、即ち関節炎の予防及び/又は治療薬の候補として選択することもできる。
 このようにして得られた候補物質が実際に抗炎症作用を有するか否かは、該候補物質を関節炎モデルに適用し、該モデルにおける炎症反応を抑制するか否かを検定することにより確認することができる。そのような関節炎モデルとしては、in vivo及びin vitroのモデルを用いることができる。in vivoモデルとしては、例えばCIAモデル(完全フロイントアジュバントとエマルジョン化したII型コラーゲンで非ヒト動物を免疫することにより調製することができる)、CAIAモデル(II型コラーゲンのCB11内のエピトープを認識するモノクローナル抗体カクテルを非ヒト動物に注射することにより調製することができる)等のRAモデル等を用いることができるが、これらに限定されない。一方、in vitroモデルとしては、関節炎における標的細胞(例えば、RAにおける滑膜細胞等)の培養系(例えば、RA患者の滑膜組織由来の滑膜線維芽細胞の培養系等)などが挙げられるが、これらに限定されない。これらのin vitroモデルは、必要に応じてTNFα等の炎症性サイトカイン等による刺激、あるいは単球、マクロファージ、好中球等の炎症性サイトカイン産生細胞との複合培養(例えば、トランスウェルTM培養システム等を用い、上部コンパートメントに標的細胞(例、滑膜線維芽細胞)、下部コンパートメントに炎症性サイトカイン産生細胞(マクロファージ様のTHP−1細胞、RAW264.7細胞)をそれぞれ単層培養する培養系)などにより、炎症反応を惹起することができる。
 候補物質が抗炎症作用を有するか否かは、上記の関節炎モデルにおける炎症反応が候補物質の添加により抑制されたか否かにより判定することができる。例えば、上記のRAモデル動物であれば、滑膜炎、炎症細胞浸潤の程度、リウマトイド因子の有無などを指標にして、関節炎治療効果の有無及び/又はその程度を判定することができる。一方、in vitro関節炎モデルである滑膜線維芽細胞の単層培養系を用いた場合、後述の実施例の通り、ゲノムの5−メチルシトシン(5mC)の脱メチル化の程度またはscratch assayの結果を指標にして炎症反応の程度を評価することができる。
 本発明のさらに別の実施態様においては、上記in vitro関節炎モデルを用いて、Tet 3の機能を阻害して関節炎の予防及び/又は治療効果を示す物質を、ワンステップでスクリーニングすることもできる。当該方法は以下の(1)~(3)の工程を含む。
(1)滑膜線維芽細胞を、被検物質に接触させる工程、
(2)前記細胞のゲノムの5−メチルシトシン(5mC)の脱メチル化または浸潤性の程度を測定する工程、
(3)被検物質の非存在下において測定した場合と比較して、前記細胞のゲノムの5−メチルシトシン(5mC)の脱メチル化または浸潤性を抑制した被検物質を、関節炎の予防及び/又は治療薬の候補として選択する工程。
 当該方法は、必要に応じて、上記工程(1)と同時もしくはその前後において、炎症を惹起する工程をさらに含み得る。炎症を惹起する方法としては、例えば、TNFα等の炎症性サイトカイン等による刺激、あるいは単球、マクロファージ、好中球等の炎症性サイトカイン産生細胞との複合培養等が挙げられる。好ましい一実施態様においては、例えば、トランスウェルTM培養システム等を用い、上部コンパートメントに標的細胞(例、滑膜線維芽細胞)、下部コンパートメントに炎症性サイトカイン産生細胞(マクロファージ様のTHP−1細胞、RAW264.7細胞)をそれぞれ単層培養する方法が挙げられる。被検物質は通常、下部コンパートメントの培地に添加されるが、例えば、腸管吸収されてTet 3の機能を阻害し得る食品中に含有される成分や、経口投与可能なTet 3機能阻害薬をスクリーニングすることを目的とする場合等においては、上部コンパートメントの培地に被検物質が添加され得る。
 細胞のゲノムの5−メチルシトシン(5mC)の脱メチル化の程度の測定は、前記細胞から調製したゲノムDNAを用いて、5hmCや5mCを認識する抗体を用いたウェスタンブロット法等の公知方法に従って定量できる。本スクリーニング方法に用いるウェスタンブロット法は、前記のTet 3タンパク質の測定方法に記載のウェスタンブロット法と同様であってよい。5hmCや5mCを認識する抗体は、その形態に特に制限はなく、5hmCや5mCを免疫原とするポリクローナル抗体であっても、またモノクローナル抗体であってもよい。
 また、細胞の浸潤性の程度の測定は、例えば、後述する実施例に記載の方法に従って測定することができる。具体的には、被験物質に接触させた細胞または接触させていない細胞を培地(10%FCS含有DMEM(必要に応じて炎症性サイトカイン(例えば、TNFα)を含有してもよい))で培養(例えば96時間)後、細胞が接着した培養皿の表面を擦過し、再度培養しながら、細胞が擦過表面に浸潤する細胞数を測定することによって、細胞の浸潤性の程度の測定・比較することができる。
 本発明の上記いずれかのスクリーニング方法を用いて得られる、Tet 3の発現または機能を阻害する物質は、炎症性疾患の予防及び/又は治療用の医薬として有用である。
 本発明のスクリーニング方法を用いて得られる化合物を上述の予防・治療剤として使用する場合、上記Tet 3の発現または機能を阻害する低分子化合物と同様に製剤化することができ、同様の投与経路および投与量で、ヒトまたは哺乳動物(例えば、マウス、ラット、ウサギ、ヒツジ、ブタ、ウシ、ウマ、ネコ、イヌ、サル、チンパンジーなど)に対して、経口的にまたは非経口的に投与することができる。
V.疾病に対する検査方法
 上述の通り、関節リウマチ患者由来の滑膜表層細胞層や炎症性サイトカインで刺激した、関節リウマチ患者由来の滑膜線維芽細胞では、Tet 3の発現レベルが高いことを見出した。
 したがって、被験者由来の試料のTet 3(又はTet 3遺伝子)の発現量を指標とすることにより、関節炎を検査することができる。
 本発明は、被験者由来の試料から、Tet3の検出物質を用いてTet 3遺伝子の転写産物または翻訳産物を検出または定量することを含む、関節炎の検査方法を提供する。Tet3の検出物質としては、具体的には、以下の(a)または(b)が挙げられる:
(a)Tet 3遺伝子の転写産物を特異的に検出し得る核酸プローブまたは核酸プライマー
(b)Tet 3遺伝子の翻訳産物を特異的に認識する抗体。
 関節炎としては、具体的には、関節リウマチ、乾癬性関節炎、脊椎関節炎(例えば、強直性脊椎炎など)などが挙げられ、好ましくは、関節リウマチが挙げられる。
 本発明の検査方法に用いられる試料としては、検査対象である被験者から分離されるものであって、検出または定量する対象であるTet3遺伝子産物(例、RNA、タンパク質、その分解産物など)を含有し得る組織または細胞等であれば特に制限されない。例えば、滑膜、滑膜線維芽細胞、滑膜表層細胞などが挙げられる。
 被験者から分離した試料におけるTet 3の検出または定量は、該被験者からRNA(例:全RNA、mRNA)画分を調製し、該画分中に含まれるTet 3遺伝子の転写産物を検出または定量することにより調べることができる。
 従って、一実施態様において、本発明の検査方法は、Tet 3遺伝子の転写産物を特異的に検出し得る核酸プローブまたは核酸プライマーを用いて測定することを特徴とする。
 RNA画分の調製は、グアニジン−CsCl超遠心法、AGPC法など公知の手法を用いて行うことができるが、市販のRNA抽出用キット(例:RNeasy Mini Kit;QIAGEN製等)を用いて、微量検体から迅速且つ簡便に高純度の全RNAを調製することができる。RNA画分中のTet 3遺伝子の転写産物を検出する手段としては、例えば、ハイブリダイゼーション(ノーザンブロット、ドットブロット、DNAチップ解析等)を用いる方法、あるいはPCR(RT−PCR、競合PCR、リアルタイムPCR等)を用いる方法などが挙げられる。微量検体から迅速且つ簡便に定量性よくTet 3遺伝子の発現変動を検出できる点で競合PCRやリアルタイムPCRなどの定量的PCR法が好ましい。
 Tet 3遺伝子の転写産物を特異的に検出し得る核酸プローブまたは核酸プライマーおよび該核酸プローブまたは核酸プライマーを用いたハイブリダイゼーション方法は、前記の本発明の関節炎の予防及び/又は治療薬のスクリーニング方法において記載した本発明の検出用核酸およびハイブリダイゼーション方法と同様であってよい。
 あるいは、被験者から分離した試料におけるTet 3の検出または定量は、該検体からタンパク質画分を調製し、該画分中に含まれる該遺伝子の翻訳産物(即ち、Tet3タンパク質)を検出または定量することにより調べることができる。Tet3の検出または定量は、Tet 3タンパク質を特異的に認識する抗体を用いて、免疫学的測定法(例:ELISA、FIA、RIA、ウェスタンブロット等)によって行うこともできる。
 従って、一実施態様において、本発明の検査方法は、Tet 3の翻訳産物を特異的に検出し得る抗体を用いて測定することを特徴とする。
 Tet 3の翻訳産物を特異的に認識する抗体および該抗体を用いた免疫学的測定方法は、前記の本発明の関節炎の予防及び/又は治療薬のスクリーニング方法において記載した本発明の検出用抗体および免疫学的測定方法と同様であってよい。
 本発明の関節炎の検査方法において、Tet 3の検出又は定量により、関節炎の検査を行うことができる。具体的には、以下の工程を含む方法であってよい。
(1)対照群および被験者から分離した試料についてTet 3を検出または定量する工程、
(2)対照群で検出または定量されたTet3と被験者で検出または定量されたTet3を比較する工程。
 後述の実施例に示されるように、変形性関節症患者(対照群)と比較して関節リウマチを発症した患者において滑膜中のTet3濃度が高い。関節炎の検査は、Tet3の濃度と関節炎への罹患率との間のこのような正の相関に基づき行われる。
 例えば、関節炎を発症していない対照群及び被験者からの試料におけるTet 3の濃度を定量し、被験者からの試料におけるTet 3の濃度を、対照群からの試料におけるTet 3の濃度と比較する。あるいは、Tet 3の濃度と関節炎の発症の有無との相関図をあらかじめ作成しておき、被験者におけるTet 3濃度をその相関図と比較してもよい。濃度の比較は、好ましくは、有意差の有無に基づいて行われる。
 そして、被験者においてTet 3が、対照群に比べて高値で検出若しくは定量された場合には、上記のような関節炎を発症している可能性が高いと判断することができる。従って、本発明の検査方法は、上記(1)、(2)の工程に加えて、(3)被験者においてTet3が、対照群に比べて高値で検出若しくは定量された場合には、関節炎を発症していると判断する工程を含んでもよい。
 さらに、本発明は、関節炎検査用のキット(診断剤)にも及ぶ。本発明の関節炎検査用キットは、上述の本発明の検査方法を簡便に実施するためのキットであればよく、特に限定されない。該検査するためのキットは、
(a)Tet 3遺伝子の転写産物を特異的に検出し得る核酸プローブまたは核酸プライマー、および/または
(b)Tet 3遺伝子の翻訳産物を特異的に認識する抗体
を含有してなる。該判定するためのキットが2以上の上記核酸および/または抗体を含む場合、各核酸または抗体は互いにTet 3遺伝子の塩基配列上の異なる部分を特異的に認識、またはTet 3遺伝子の翻訳産物の異なるエピトープを特異的に認識し得るものである。
 本発明のキットが前記(a)の核酸を含有する試薬を構成として含む場合、該核酸としては、本発明の検査方法において前記したプローブ用核酸もしくはプライマー用オリゴヌクレオチドが挙げられる。
 Tet 3遺伝子の発現を検出し得る核酸は、乾燥した状態もしくはアルコール沈澱の状態で、固体として提供することもできるし、水もしくは適当な緩衝液(例:TE緩衝液等)中に溶解した状態で提供することもできる。標識プローブとして用いられる場合、該核酸は予め上記のいずれかの標識物質で標識した状態で提供することもできるし、標識物質とそれぞれ別個に提供され、用時標識して用いることもできる。
 あるいは、該核酸は、適当な固相に固定化された状態で提供することもできる。固相としては、例えば、ガラス、シリコン、プラスチック、ニトロセルロース、ナイロン、ポリビニリデンジフロリド等が挙げられるが、これらに限定されない。また、固定化手段としては、予め核酸にアミノ基、アルデヒド基、SH基、ビオチンなどの官能基を導入しておき、一方、固相上にも該核酸と反応し得る官能基(例:アルデヒド基、アミノ基、SH基、ストレプトアビジンなど)を導入し、両官能基間の共有結合で固相と核酸を架橋したり、ポリアニオン性の核酸に対して、固相をポリカチオンコーティングして静電結合を利用して核酸を固定化するなどの方法が挙げられるが、これらに限定されない。
 該検査するためのキットに含有される核酸は、同一の方法(例:ノーザンブロット、ドットブロット、DNAアレイ技術、定量RT−PCR等)によりTet 3遺伝子の発現を検出し得るように構築されていることが特に好ましい。
 本発明のキットが前記(b)の抗体を含有する試薬を構成として含む場合、該抗体としては、本発明の検査方法において前記した抗体が挙げられる。
 本発明のキットを構成する試薬は、Tet 3遺伝子の発現を検出し得る核酸や抗体に加えて、該遺伝子の発現を検出するための反応において必要な他の物質であって、共存状態で保存することにより反応に悪影響を及ぼさない物質をさらに含有することができる。あるいは、該試薬は、Tet 3遺伝子の発現を検出するための反応において必要な他の物質を含有する別個の試薬とともに提供されてもよい。例えば、Tet3遺伝子の発現を検出するための反応がPCRの場合、当該他の物質としては、例えば、反応緩衝液、dNTPs、耐熱性DNAポリメラーゼ等が挙げられる。競合PCRやリアルタイムPCRを用いる場合は、competitor核酸や蛍光試薬(上記インターカレーターや蛍光プローブ等)などをさらに含むことができる。また、Tet 3遺伝子の発現を検出するための反応が抗原抗体反応の場合、当該他の物質としては、例えば、反応緩衝液、competitor抗体、標識された二次抗体(例えば、一次抗体がウサギ抗ヒトTet 3抗体の場合、ペルオキシダーゼやアルカリホスファターゼ等で標識されたマウス抗ウサギIgGなど)、ブロッキング液等が挙げられる。
 以下の実施例は、単に本発明をより具体的に例示するためのものであって、本発明の範囲を制限するものではない。
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. : 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. In the present specification, 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.
As used herein, 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. Cells, synovial surface cells, etc.] or tissues [e.g., synovial membranes (especially, synovial surface cell layers), etc.] and the like may be isolated and purified by known protein separation and purification techniques.
Examples of the “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 SEQ ID NO: 1;
(E) Demethylation activity of 5-methylcytosine (5 mC) or synovial fiber encoded by DNA that hybridizes under stringent conditions with DNA having a complementary strand sequence of the base sequence represented by SEQ ID NO: 1 An amino acid sequence having an activity of promoting blast infiltration.
Specifically, 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.
As used herein, “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). Specific examples of conservative amino acid substitutions are well known in the art and are described in various literature (see, for example, Bowie et al., Science, 247: 1306-1310 (1990)).
The homology of the amino acid sequences in this specification is determined using the homology calculation algorithm NCBI BLAST (National Center for Biotechnology Information Local Alignment Search Tool), and the following conditions (expectation value = 10; allow gap; matrix = BLOSUM62; filtering) = OFF). Other algorithms for determining amino acid sequence homology include, for example, Karlin et al., Proc. Natl. Acad. Sci. USA, 90: 5873-5877 (1993) [the algorithm is incorporated into the NBLAST and XBLAST programs (version 2.0) (Altschul et al., Nucleic Acids Res., 25: 3389-3402 (1997)]. )], Needleman et al., J. MoI. Mol. Biol. 48: 444-453 (1970) [the algorithm is incorporated into the GAP program in the GCG software package], Myers and Miller, CABIOS, 4: 11-17 (1988) [ The algorithm is incorporated into the ALIGN program (version 2.0) which is part of the CGC sequence alignment software package], Pearson et al., Proc. Natl. Acad. Sci. USA, 85: 2444-2448 (1988) [the algorithm is incorporated in the FASTA program in the GCG software package] and the like, and they can be preferably used as well.
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.
More preferably, as “an amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO: 2”, 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.
Here, “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.
Here, 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.
Examples of the Tet3 protein in the present invention 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. 2) an 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.
When the amino acid sequence is inserted, deleted, added or substituted as described above, 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.
Here, as 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. Here, as 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. Etc.
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. 0431151), or an ortholog, allelic variant, and polymorphic variant thereof in other mammals [eg, Single nucleotide polymorphisms (SNPs)] and the like.
“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. Specifically, the following (f) to (j):
(F) a base sequence encoding the amino acid sequence represented by SEQ ID NO: 2,
(G) 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 A base sequence encoding an amino acid sequence having an invasion promoting activity,
(H) 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 A base sequence encoding the sequence,
(I) the base sequence represented by SEQ ID NO: 1,
(J) a base sequence that hybridizes with a DNA having a complementary strand sequence of the base sequence represented by SEQ ID NO: 1 under stringent conditions, and the demethylation activity or synovium of 5-methylcytosine (5 mC) A base sequence encoding an amino acid sequence having an activity of promoting fibroblast infiltration,
The nucleic acid which has is mentioned.
Here, 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. In this specification, 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
In the present invention, the “substance that inhibits the expression of Tet3” 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. Therefore, 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), and 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.
(1) 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.
Here, a preferable example of the transcription product is mRNA.
As a substance that specifically inhibits the translation of Tet 3 gene from mRNA to Tet 3 (or degrades mRNA), it is preferable that 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 having a degree of complementarity capable of binding to the target sequence of the mRNA and inhibiting its translation (or cleaving the target sequence), specifically, for example, the base sequence of the mRNA About 90% or more, preferably about 95% or more, more preferably about 97% or more of the homologous base sequence (that is, the base sequence of the complementary strand of mRNA) with the overlapping region. It is a base sequence.
The “base sequence homology” in the present invention uses the homology calculation algorithm NCBI BLAST (National Center for Biotechnology Information Basic Alignment Search Tool) and the following conditions (expectation value = 10; allow gap; filtering = ON; Match score = 1; mismatch score = -3).
More specifically, 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 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.
Preferred examples of Tet 3 gene mRNA 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. It contains 10 bases or more, preferably about 15 bases or more.
Specifically, 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.
(1) Antisense nucleic acid against Tet 3 gene mRNA
The “antisense nucleic acid against Tet3 gene mRNA” in the present invention 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). They may be 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, psoralen, etc.), chelate compounds (eg, , Metals, radioactive metals, boron, oxidizing metals, etc.), alkylating agents, and modified bonds (eg, alpha anomeric nucleic acids) Also good. Here, the “nucleoside”, “nucleotide” and “nucleic acid” 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. When the antisense nucleic acid is DNA, 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. Specifically, 5 ′ end hairpin loop, 5 ′ end 6-base pair repeat, 5 ′ end untranslated region, translation start codon, protein coding region, ORF translation stop codon, 3 ′ end untranslated region of Tet3 gene 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.
Furthermore, 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. For example, in order to prevent degradation by a hydrolase such as nuclease, 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. In addition, 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. Furthermore, the base moiety (pyrimidine, purine) may be chemically modified, for example, introduction of a methyl group or a cationic functional group at the 5-position of the pyrimidine base, or substitution of the carbonyl group at the 2-position with thiocarbonyl, etc. Is mentioned.
The conformation of the sugar part of RNA is dominated by C2′-endo (S type) and C3′-endo (N type). In single-stranded RNA, it exists as an equilibrium between the two, but double-stranded Is fixed to the N type. Therefore, in order to give strong binding ability to the target RNA, BNA (LNA) (Imanishi), which is an RNA derivative in which the conformation of the sugar moiety is fixed to the N-type by cross-linking 2 ′ oxygen and 4 ′ carbon , T. et al., Chem. Commun., 1653-9, 2002; Jepsen, JS et al., Oligonucleotides, 14, 130-46, 2004) and ENA (Morita, K. et al., Nucleosides). Nucleotides Nucleic Acids, 22, 1619-21, 2003) can also be preferably used.
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. In addition, any of the above-described antisense nucleic acids containing various modifications can be chemically synthesized by a method known per se.
(2) 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. When the Tet 3 gene mRNA has a double-stranded structure by itself, 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. [Proc. Natl. Acad. Sci. USA, 98 (10): 5572-5577 (2001)]. Furthermore, when 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. [Nucleic Acids Res. 29 (13): 2780-2788 (2001)].
(3) siRNA against Tet 3 gene mRNA
In the present specification, 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. When a short double-stranded RNA is introduced into a cell, a phenomenon called RNA interference (RNAi), in which mRNA complementary to the RNA is degraded, has been known in nematodes, insects, plants, etc. Since it has been confirmed that this phenomenon occurs widely in animal cells [Nature, 411 (6836): 494-498 (2001)], it has been widely used as an alternative technique to the above-described antisense nucleic acids and ribozymes.
Based on the cDNA sequence information of the target gene, 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. For example, AA + (N) 19 (following AA, 19 Base sequence), AA + (N) 21 (base sequence of 21 bases following AA) or A + (N) 21 (base sequence of 21 bases following A).
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. Examples of such an additional base sequence include ug-3 ′, uu-3 ′, tg-3 ′, tt-3 ′, ggg-3 ′, guuu-3 ′, gttt-3 ′, and tttt-3. Examples of such sequences include, but are not limited to, ', uuuu-3'.
Moreover, siRNA may have an overhang | projection part sequence (overhang) in 3 'terminal, and specifically, what added dTdT (dT represents the deoxythymidine residue of deoxyribonucleic acid) is mentioned. . Further, it may be a blunt end (blunt end) without end addition.
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. Can be mentioned. 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. For a candidate group of target sequences selected based on the above-mentioned rules and others, whether or not there is a homology in a 16-17 base sequence in mRNA other than the target is determined by BLAST (http: //www.ncbi.nlm Investigate using homology search software such as .nih.gov / BLAST /) to confirm the specificity of the selected target sequence. For the target sequence confirmed to be specific, 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. In addition, for the short hairpin RNA (shRNA) that is a precursor of siRNA, an arbitrary linker sequence (for example, about 5-25 bases) capable of forming a loop structure is appropriately selected, and the sense strand and the antisense strand are combined with each other. It can be designed by linking via a linker sequence.
The sequence of siRNA and / or shRNA can be searched using search software provided free of charge on various web sites. Such sites include, for example, 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), 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. However, in the case of 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. .
Specifically, in order to improve a part of the nucleotide molecule constituting siRNA, natural DNA, stability (chemical and / or enzyme) and specific activity (affinity with RNA) as the modification. 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). For example, in order to prevent degradation by a hydrolase such as nuclease, 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. In addition, 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. Furthermore, the base moiety (pyrimidine, purine) may be chemically modified, for example, introduction of a methyl group or a cationic functional group at the 5-position of the pyrimidine base, or substitution of the carbonyl group at the 2-position with thiocarbonyl, etc. Is mentioned. In addition, 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. In addition, 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.
In addition, 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. It may be chemically bonded to oligoarginine, Tat peptide, Rev peptide, Ant peptide or the like that adsorbs and penetrates the surface layer.
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.
In the present specification, 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. It can be prepared by designing RNA and synthesizing it with an automatic DNA / RNA synthesizer. There are tandem type and stem loop (hairpin) type in vectors expressing shRNA. In the former, 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). It is. On the other hand, the latter is one in which an shRNA expression cassette is inserted into a vector, in which shRNA is expressed in cells and processed by dicer to form dsRNA. As a promoter, a pol II promoter (for example, a CMV immediate early promoter) can be used, but a pol III promoter is generally used in order to cause transcription of a short RNA accurately. Examples of the polIII promoter include mouse and human U6-snRNA promoter, human H1-RNase P RNA promoter, human valine-tRNA promoter, and the like. Further, 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. As such vectors, 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. 23, 2677-2684, Wincott et al. , 1997, Methods Mol. Bio. 74, 59, Brennan et al. , 1998, Biotechnol Bioeng. 61, 33-45, Usman et al. , 1987
J. et al. Am. Chem. Soc. 109, 7845; Scaringe et al. 1990 Nucleic Acids Res. , 18, 5433, and the method described in US Pat. No. 6,00131111. Specifically, it can be synthesized using a nucleic acid protecting group (for example, dimethoxytrityl group at the 5 ′ end) and a coupling group (for example, phosphoramidite at the 3 ′ end) known to those skilled in the art. That is, the protecting group at the 5 ′ end is deprotected with an acid such as TCA (trichloroacetic acid) and a coupling reaction is performed. Then, after capping with an acetyl group, the next nucleic acid condensation reaction is performed. In the case of siRNA containing modified RNA or DNA, a modified RNA (for example, 2′-O-methyl nucleotide, 2′-deoxy-2′-fluoronucleotide) may be used as a raw material, and the coupling reaction These conditions can be adjusted as appropriate. When introducing a phosphorothioate bond in which the phosphate binding moiety is modified, a borage reagent (3H-1,2-benzodithiol-3-one 1,1-dioxide) can be used.
Alternatively, oligonucleotides may be synthesized separately and joined together after synthesis, for example by ligation (Moore et al., 1992, Science 256, 9923; Draper et al. International Publication WO 93/23569; Shabarova et al. , 1991, Nucleic Acids Research 19, 4247; Bellon et al., 1997, Nucleosides & Nucleotides, 16, 951: Bellon et al., 1997, Nucleosides & Nucleotides, j. Et al. And / or connected together by hybridization after deprotection It may be. 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.
In the present invention, 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. Preferably, the sequence consisting of SEQ ID NO: 3 and the sequence consisting of SEQ ID NO: 4 are exemplified.
Another preferred example of 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. Examples of 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. Examples of 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. Such capping groups include, but are not limited to, hydroxyl protecting groups known in the art, including glycols such as polyethylene glycol and tetraethylene glycol.
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.
III. 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, and 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.
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.
When the nucleic acid of the present invention is used as a prophylactic and / or therapeutic agent for the arthritis, it can be formulated and administered according to a method known per se. That is, 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.
Furthermore, for the purpose of improving the pharmacokinetics, prolonging the half-life, and improving the efficiency of cellular uptake, 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.
As 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. As 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. As the oily liquid, for example, sesame oil, soybean oil and the like are used, and benzyl benzoate, benzyl alcohol and the like may be used in combination as a solubilizing agent. The prepared injection solution is preferably filled in a suitable ampoule. Suppositories used for rectal administration may be prepared by mixing the nucleic acid with a normal suppository base.
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. As the 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.
Each of the above-described 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. Can be used for prevention. Specific examples of arthritis include rheumatoid arthritis, psoriatic arthritis, spondyloarthritis (eg, ankylosing spondylitis), and preferably rheumatoid arthritis. In addition, 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.
When 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. For example, mesalazine, corticosteroids (eg, betamethasone, prednisolone, hydrocortisone, dexamethasone, etc.), nonsteroidal anti-inflammatory drugs (NSAIDs; eg, salicylic acid, anthranilic acid) , Aryl acids, propionates, oxicams, pilins), anti-TNFα antibodies (influximab, adalimumab), anti-rheumatic drugs (eg, immunomodulators such as actarit, immunosuppressants such as methotrexate, anti-TNFα antibodies And biological preparations such as etanercept).
IV. Screening drug candidate compounds for disease
As mentioned above, inhibition of Tet 3 expression and / or function suppresses the demethylation of 5-methylcytosine (5mC), which is the most important aggressive phenotype of activated synovial fibroblasts One invasive ability is suppressed. This indicates that selective inhibition of Tet3 inhibits the expression of CCL2 and ICAM1 through the maintenance of DNA methylation levels, and accordingly, inhibits the activation of synovial fibroblasts represented by invasiveness. Means that it leads to treatment of arthritis. Therefore, a compound that inhibits the expression and / or function of Tet 3 can be used as a prophylactic and / or therapeutic agent for arthritis.
Therefore, cells that produce 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.
When screening for a compound that inhibits the expression or function of Tet 3, 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 | limiting. In the case of 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. .
Moreover, 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. As the host, for example, 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.
Next, using the obtained 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. What is necessary is just to acquire the cell which expresses protein. 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.
For example, 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).
In addition, animal cell expression plasmids (eg, pA1-11, pXT1, pRc / CMV, pRc / RSV, pcDNAI / Neo); bacteriophages such as λ 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. For example, SRα 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, β actin A gene promoter, aP2 gene promoter, etc. are used. Of these, EF-α promoter, CAG promoter, CMV promoter, SRα promoter and the like are preferable.
In addition to the above, 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. it can.
Examples of the selectable marker include a dihydrofolate reductase gene (hereinafter abbreviated as dhfr, methotrexate (MTX) resistance), an ampicillin resistance gene (hereinafter amp). r A neomycin resistance gene (hereinafter, neo) r G418 resistance) and the like. In particular, when dhfr gene-deficient Chinese hamster cells are used and the dhfr gene is used as a selection marker, 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. Examples of mammalian cells 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. For example, in the case of Escherichia coli, 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. For example, 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. For example, 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. Further, when the Tet 3 protein accumulates in the cells of the transformant obtained by the above culture, for example, the transformant is collected by centrifugation or the like, and then the cells are disrupted or lysed. If there is, the 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.
In conducting the screening of the present invention, examples of the test substance 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.
In addition, when selecting a substance that decreases the expression level of Tet 3 or the Tet 3 gene or a substance that decreases the function of Tet 3, a control cell that is not brought into contact with the test substance can also be used as a comparative control. Here, “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 The case where a negative control substance that does not affect the above is added 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 μM, for example. Examples of the incubation time include about 10 minutes to about 24 hours.
When 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. For example, 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.). There are no particular restrictions on the breeding conditions of the animals used, but it is preferable that the animals are raised in an environment of SPF grade or higher. Contact of the test substance with the cells is carried out by administering the test substance to the animal individual. 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. .
Alternatively, 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.
(Measurement of expression level of Tet 3 gene or Tet 3)
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”). .
Therefore, more specifically, 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 method for screening a prophylactic and / or therapeutic agent for arthritis, characterized by measuring and comparing; and
(B) Cells having the ability to produce Tet 3 are cultured in the presence and absence of a test substance, and the amount of the protein under both conditions is measured and compared using the detection antibody 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.
(I) administering a test substance 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. On the other hand, the amount of Tet 3 protein can be quantified using Western blot analysis or various immunoassay methods described in detail below.
(Ii) 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 (mRNA) 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.
When using Northern blotting, 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. In addition, using AlkPhos Direct Labeling and Detection System (manufactured by Amersham Pharmacia Biotech), 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 (manufactured by Amersham Pharmacia Biotech) can also be used.
When using the RT-PCR method, 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. In addition, 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. For example, 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). Also, 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). You can also
Screening for a substance that changes the expression level of Tet 3 can also be performed by a reporter gene assay using the transcriptional regulatory region of Tet 3 gene. Here, the “transcriptional regulatory region” usually refers to a range of several kb to several tens of kb upstream of the chromosomal gene. For example, (i) 5′-race method (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. (ii) Genome Walker Kit (which can be carried out using “-full Race Core Kit (manufactured by Takara Bio Inc.), etc.); The 5′-upstream region can be obtained using Clontech, etc., and the obtained upstream region can be identified by a technique including a step of measuring promoter activity.
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. The vector may be prepared by a method known to those skilled in the art. That is, “Molecular Cloning: A Laboratory Manual 2nd edition” (1989), Cold Spring Harbor Laboratory Press, “Current Protocols in Molecular Biology” (1987), John Ins. The transcriptional regulatory region of the Tet 3 gene excised according to the usual genetic engineering technique described in the above can be incorporated on a plasmid containing a reporter gene.
Reporter proteins include β-glucuronidase (GUS), luciferase, chloramphenicol transacetylase (CAT), β-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. Alternatively, 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.
In addition, as a method for measuring the expression level of the reporter gene, a method corresponding to each reporter gene may be used. For example, when a luciferase gene is 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. In this case, a commercially available luciferase reaction detection kit such as Picker Gene Dual Kit (registered trademark; manufactured by Toyo Ink) can be used.
As 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. How,
(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. 125 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. In addition, after using an antibody that recognizes Tet 3 as a primary antibody, 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. In general, 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. For example, if 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.
As a labeling agent used in a measurement method using a labeling substance, for example, a radioisotope, an enzyme, a fluorescent substance, a luminescent substance, or the like is used. Examples of radioisotopes include [ 125 I], [ 131 I], [ 3 H], [ 14 C] and the like are used. As the enzyme, a stable enzyme having a large specific activity is preferable. For example, β-galactosidase, β-glucosidase, alkaline phosphatase, peroxidase, malate dehydrogenase and the like are used. As the fluorescent substance, for example, fluorescamine, fluorescein isothiocyanate and the like are used. As the luminescent substance, for example, luminol, luminol derivatives, luciferin, lucigenin and the like are used. Furthermore, 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. Examples of the carrier include insoluble polysaccharides such as agarose, dextran, and cellulose, synthetic resins such as polystyrene, polyacrylamide, and silicon, or glass.
In the sandwich method, 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. 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. Further, in the immunoassay by the sandwich method, 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.
In the competition method, 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. Separation (B / F separation) and the amount of labeling of either B or F is measured to quantify Tet 3 in the sample solution. In this reaction method, 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), and 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).
In the immunometric 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. What is necessary is just to construct | assemble the measurement system of Tet3, adding the usual technical consideration of those skilled in the art to the usual conditions and operation method in each method. For details of these general technical means, it is possible to refer to reviews, books and the like.
For example, Hiroshi Irie “Radioimmunoassay” (Kodansha, published in 1974), Hiroshi Irie “Continue Radioimmunoassay” (published in Kodansha, 1979), “Enzyme Immunoassay” edited by Eiji Ishikawa et al. 53)), edited by Eiji Ishikawa et al. "Enzyme Immunoassay" (2nd edition) (Medical Shoin, published in 1982), edited by Eiji Ishikawa et al. "Enzyme Immunoassay" (3rd edition) (Medical Shoin, Showa 62)), “Methods in ENZYMOLOGY” Vol. 70 (Immunochemical Technologies (Part A)), ibid., Vol. 73 (Immunochemical Technologies (Part B)), ibid., Vol. 74 (Immunochemical Technologies (Part C)), ibid., Vol. 84 (Immunochemical Technologies (Part D: Selected Immunoassays)), ibid., Vol. 92 (Immunochemical Technologies (Part E: Monoclonal Antibodies and General Immunoassay Methods)), Vol. 121 (Immunochemical Technologies (Part I: Hybridoma Technology and Monoclonal Antibodies)) (published by Academic Press).
As described above, the amount of Tet3 in a cell can be quantified with high sensitivity by using the detection antibody of the present invention.
For example, in the above screening method, 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. %, More preferably about 50% or more, 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.
Alternatively, in the screening method described above, 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. When such cells are used, the expression level of Tet 3 can be evaluated by measuring the expression level of the reporter gene using a conventional method.
(Measurement of Tet 3 function)
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. By measuring, a test substance having the ability to bind to Tet 3 can be detected. Instead of directly labeling Tet 3, it is also possible to detect Tet 3 bound to the solid phase using a labeled anti-Tet 3 antibody. Alternatively, 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. Examples of 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. On the other hand, 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. These in vitro models can be stimulated by inflammatory cytokines such as TNFα 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) 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, etc. System) and the like can induce an inflammatory reaction.
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. For example, in the case of the above RA model animal, 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. On the other hand, when a synovial fibroblast monolayer culture system, which is an in vitro arthritis model, was used, the degree of demethylation of genomic 5-methylcytosine (5 mC) or the result of scratch assay, as described in the examples below. The degree of inflammatory reaction can be evaluated using as an index.
In yet another embodiment of the present invention, 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. Examples of methods for inducing inflammation include stimulation with inflammatory cytokines such as TNFα, and complex culture with inflammatory cytokine-producing cells such as monocytes, macrophages and neutrophils. In a preferred embodiment, for example, 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. For example, screening for ingredients contained in foods that can be absorbed into the intestine and inhibit the function of Tet 3, and orally administrable Tet 3 function inhibitors In the case where it is intended to do so, 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. Specifically, 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, TNFα) 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. Can do.
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.
When the compound obtained by using the screening method of the present invention is 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. Testing method for disease
As described above, it was found that the expression level of 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. Specific examples of the 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.
Specific examples of 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. As 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.
Alternatively, 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.
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.
In the 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.
As shown in Examples described later, 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.
For example, 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. Alternatively, 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.
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.
Furthermore, 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. When 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.
When the kit 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. When used as a labeled probe, 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.
Alternatively, the nucleic acid can be provided in a state immobilized on an appropriate solid phase. Examples of the solid phase include, but are not limited to, glass, silicon, plastic, nitrocellulose, nylon, polyvinylidene difluoride, and the like. As immobilization means, 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. Examples of 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.
When 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. Alternatively, 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. For example, when the reaction for detecting the expression of the Tet3 gene is PCR, examples of the other substance include a reaction buffer, dNTPs, and a heat-resistant DNA polymerase. When competing PCR or real-time PCR is used, it can further include a competitor nucleic acid, a fluorescent reagent (such as the above intercalator and fluorescent probe), and the like. In addition, when 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). In the case of 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.
(実施例1)滑膜におけるTetタンパク質ファミリーの発現レベル
 Tetタンパク質ファミリーとしてはTet 1,Tet 2,Tet 3の3種類のサブタイプが存在し、細胞ごとに発現レベルが異なること、またそれぞれが標的にする遺伝子座が異なることが知られる。そのため、まずは関節リウマチ(RA)患者、対照となる変形性関節症(OA)患者由来の滑膜における、Tetタンパク質ファミリーの発現レベルを免疫組織化学染色法によって調べた。滑膜は関節手術の際に得られた滑膜組織の一部を凍結保存して後に組織切片を作成した。抗体はGoat−anti−human TET1 Ab(Santa Cruz biotech,sc−1634443),mouse anti−human TET2 Ab(sc−136926),rabbit anti−huuman TET3 Ab(sc−139186),rabbit anti−5−hydroxymethylcytosine(5−hmC)polyclonal Ab(ActiveMotif),rabbit anti−5−methylcytosine(5−mC)polyclonal Ab(ActiveMotif)を用い、二重染色にはmouse monoclonal anti−CD55 Ab(ab89190)、mouse monoclonal anti−CD68 Ab(ab74704)、2次抗体としてはMACH 2 double stain 2(mouse−HRP + rabbit−AP)を使用した。観察にはBIOREVO(keyence,Japan)を使用した。
 その結果、OA、RAともにTet 1、Tet 2の発現レベルには差が認められず、Tet 3は滑膜表層細胞層において、OAよりもRAで強い発現が認められた(図1~3)。
 また、RA、OA由来の滑膜におけるDNAメチル化レベルおよびTetタンパク質ファミリーによる脱メチル化レベルを調べるために、5−メチルシトシン(5mC)および5−ヒドロキシメチルシトシン(5hmC)を免疫組織化学染色した。
 その結果、RA、OA共に、5mCのレベルは低く、5hmCのレベルは中等度で滑膜表層でより強い印象であった(図1、2)。
(実施例2)滑膜線維芽細胞(FLS)におけるTetタンパク質ファミリーの発現レベル
 継代培養したFLSではすでに炎症性サイトカインの直接的な影響はないが、RA特有のDNAメチル化プロファイルは保たれている(Nakano K et al.Ann Rheum Dis 2013)。そこで、炎症性サイトカイン非存在下で培養した場合のTetファミリーの発現レベル(mRNA、タンパク質)を、RA、OA、健常人由来FLSで比較した。FLSとしては、実施例1の滑膜組織にコラゲナーゼ処理を加えて抽出した滑膜細胞を4−6継代して、99%以上の純度が確認されたFLSを用いた。mRNAの発現はStepOnePlusTMを使用してリアルタイムPCRで解析した。primer/probeはTaqMan Gene Expression AssaysのTET1(Hs00286756_m1),TET2(Hs00325999_m1),TET3(00379125_m1)を用い、発現レベルをGAPDH(Hs99999905_m1)で補正した。蛍光染色には一次抗体としてGoat−anti−human TET1 Ab(Santa Cruz biotech,sc−1634443),mouse anti−human TET2 Ab(sc−136926),rabbit anti−human TET3 Ab(sc−139186)、二次抗体としてFITC−conjugated anti−goat IgG,FITC−conjugated anti−mouse IgG,FITC−conjugated anti−rabbit IgGをそれぞれ使用した。観察にはBIOREVO(keyence,Japan)を、画像解析にはBZ−II Viewer software(keyence)を使用した。
 その結果、炎症性サイトカインによる刺激がないFLSにおけるTetタンパク質ファミリーのmRNA発現レベルは、RA、OA、健常人の間で差を認めなかった(図4)。一方、タンパク質発現レベルでは、Tet1、Tet2の発現レベルは非常に低く、Tet3はTet 1、Tet 2に比べて強い発現がOA、RA由来FLSで認められた(図5)。
(実施例3)炎症性サイトカイン刺激後のFLSにおけるTetタンパク質ファミリーの発現レベル
 実施例2のFLSにRAの病態に中心的に関与するとされる炎症性サイトカインTNFαやIL−1βを加えて培養した場合の、Tetタンパク質ファミリーの発現レベルの変化(mRNA、タンパク質)を調べた。TNFαはrecombinant human TNF−alpha(R&D),IL−1βはrecombinant human IL−1β(RELIATech GmbH)を使用して、それぞれ50ng/ml,1ng/mlで刺激した。また、蛍光染色を行い、ランダムに20個の細胞の核内と細胞質の蛍光強度を解析し、蛍光強度の比をN/C比とした。核タンパクの抽出にはNuclear Extraction Kit(Affymetrix,Fremont,CA)を使用し、Western blot法でタンパク質の発現を解析した。
 その結果、TNFαやIL−1βによってFLSを刺激すると、OA、RAに関わらず、mRNA発現レベルに関しては、Tet 1が刺激後2時間で顕著に低下したことに対して、Tet3は刺激後2時間で有意に発現が増加した。Tet2の発現の変化は認めなかった(図6)。タンパク質発現レベルに関しては、健常者、OA、RAに関わらず、TNFα刺激後24時間からFLSの核内のTet 3発現レベルは有意に増加し、刺激後96時間で顕著となった(図7、8)。
(実施例4)炎症性サイトカイン刺激後のFLSにおけるDNAメチル化レベル
 RA由来FLSにTNFαによる刺激を加え、DNAの脱メチル化レベルを調べるために5hmCのレベル(genomic DNA)の変化を調べた。gDNAの抽出にはSpinclean genomic DNA purification kit(m.biotech)を用い、Dot Blot法でgDNA中の5hmCの発現レベルを解析した。一次抗体にはrabbit anti−5−methylcytosine(5−mC)polyclonal Ab(ActiveMotif)、二次抗体にはECLTM Anti−Rabbit IgG,HRP linked whole antibody(GE healthcare,UK)、検出にはECLTM Prime Western blotting detection reagent(GE healthcare,UK)を用いた。コントロールとしては5−Methylcytosine & 5−Hydroxymethylcytosine DNA Standard Setを使用した。画像撮影にはLight−capture(AE−6972,Atto,Tokyo,Japan)を使用し、画像解析にはCS Analyzer,version 3.0(Atto)を用いて定量化を行った。
 その結果、TNFαによって刺激したFLSは刺激後96時間で有意に5hmCのレベルが増加した(図9)。
(実施例5)Tet 3発現抑制後のFLSにおける炎症性サイトカインの分泌レベル
 Tet 3 siRNAを用いてTet 3発現を阻害した上で、TNFαによって刺激を加え、RA由来FLSが分泌する炎症性サイトカインレベルを評価した。具体的には、配列番号:3および配列番号4の塩基配列からなるsiRNAでTet 3をノックダウンしたFLSをTNFα(1ng/ml)を含む培地(10%FCS含有DMEM)で96時間培養後、洗浄して、さらにTNFαを含まない0.1%FCS含有DMEMで48時間の培養を行った。コントロールとしてTet 1のsiRNAを使用した。予備的検討で使用したsiRNA(いずれもLife technologies社のPrimer Set Human)はそれぞれの遺伝子について2種類ずつ、すなわち、siRNA ID:HSS129586,HSS129587 for TET1,HSS123253,HSS123254 for TET2,HSS153381,HSS176459 for TET3を使用した。コントロールとしてはStealthTM RNAi Negative Control DuplexesよりLow GC Duplex(Invitrogen)を使用した。Lipofectamine RNAiMAX Transfection Reagent(Life technologies)を用いてリポフェクション法でsiRNA遺伝子を導入した。遺伝子導入効率の予備的検討の結果より、TET3のsiRNAとしてはHSS153381、TET1のsiRNAとしてはHSS129587を使用することとした。IL−6、IL−8、VEGF、CCL2などのタンパク分泌量についてはBDTMCytometric Bead Array(CBA,BD,Japan)で、MMP3の分泌量はSRLに測定を依頼した(検査方法はLTIA法)。またICAM−1やVCAM−1の接着分子の発現はflow−cytometry法(FACSVerseTM,BD)で測定し、FlowJo(Miltenyi Biotec)で解析した。CCL2のmRNAの発現解析にはTaqMan(R)GeneExpress Assays CCL2(Hs00234140_m1)を用いた。
 その結果、RAの病態への関連が示唆されてきた多数のサイトカイン、ケモカイン、MMPsの中で、IL−6、IL−8、VEGF、CCL2、MMP3などはTNFα依存性の発現誘導が認められたが、これらの中でCCL2のみはTet 3ノックダウンによりTNFα依存性の発現誘導が阻害された(図10)。この現象はmRNAと分泌タンパクレベルでも確認された。また、接着分子の中でICAM1も同様にTet 3ノックダウンによりTNFα依存性の発現誘導が阻害された。CCL2やICAM−1はFLSの骨・軟骨組織への浸潤に関わり、RAの病態形成に深く関与することが知られている。従って、Tet 3ノックダウンは、DNAのメチル化レベルの維持を介して、CCL2やICAM−1の発現を抑制することによって、RAの予防および治療に有効であることが示唆された。なお、IL−1β、TNFα、IL−17αについても検出を試みたが、値が検出感度以下であったため、検出することができなかった。
(実施例6)Tet 3発現抑制後のFLSの浸潤能
 Tet 3 siRNAを用いてTet 3発現を阻害した上で、TNFαによって刺激を加え、RA由来FLSの浸潤能をScratch assayで評価した。具体的には、siRNAでTet 3をノックダウンしたFLSをTNFα(1ng/ml)を含む培地(10%FCS含有DMEM)で96時間培養後、洗浄して、FLSが接着したdish表面にscratchを施行し、TNFαを含まない0.1%FCS含有DMEMで培養しながら、最長48時間の観察を行った。コントロールとしてTet 1のsiRNAを使用した。
 その結果、Tet 3ノックダウンによりTNFα依存性のFLS浸潤誘導が完全に阻害された(図11)。Tet 3の選択的阻害が関節リウマチ患者の滑膜線維芽細胞において見られる最も重要な攻撃的な表現型の一つである浸潤性を阻害することで、RAの予防および治療につながることが示唆された。
(Example 1) 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. 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.
As a result, there was no difference in the expression levels of Tet 1 and Tet 2 in both OA and RA, and Tet 3 was more strongly expressed in RA than OA in the synovial surface cell layer (FIGS. 1 to 3). .
In addition, in order to examine the DNA methylation level in the RA, OA-derived synovium and the demethylation level by the Tet protein family, 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) were immunohistochemically stained. .
As a result, the level of 5 mC was low for both RA and OA, and the level of 5 hmC was moderate, giving a stronger impression on the synovial surface (FIGS. 1 and 2).
(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 . For primer / probe, TET1 (Hs00286756_m1), TET2 (Hs00325999_m1), and TET3 (00379125_m1) from TaqMan Gene Expression Assays were used, and the expression level was corrected with GAPDH (Hs9999999_m1). For fluorescent staining, as primary antibodies, 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) was used for observation, and BZ-II Viewer software (keyence) was used for image analysis.
As a result, there was no difference in the mRNA expression level of the Tet protein family in FLS without stimulation by inflammatory cytokine among RA, OA, and healthy individuals (FIG. 4). On the other hand, at the protein expression level, the expression levels of Tet1 and Tet2 were very low, and Tet3 was observed to be stronger in OA and RA-derived FLS than Tet1 and Tet2 (FIG. 5).
(Example 3) Expression level of Tet protein family in FLS after stimulation with inflammatory cytokines In the case of culturing with addition of inflammatory cytokines TNFα and IL-1β 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. TNFα was stimulated with recombinant human TNF-alpha (R & D), and IL-1β was stimulated with 50 ng / ml and 1 ng / ml, respectively, using recombinant human IL-1β (RELIATech GmbH). In addition, fluorescence staining was performed, and the fluorescence intensity of the nucleus and cytoplasm of 20 cells was randomly analyzed, and the ratio of the fluorescence intensity was defined as the N / C ratio. Nucleo Extraction Kit (Affymetrix, Fremont, Calif.) Was used for the extraction of nucleoprotein, and protein expression was analyzed by Western blot method.
As a result, when FLS was stimulated with TNFα or IL-1β, Tet 1 significantly decreased 2 hours after stimulation, whereas Tet 3 2 hours after stimulation, regardless of OA and RA. The expression increased significantly. There was no change in the expression of Tet2 (FIG. 6). Regarding the protein expression level, regardless of healthy subjects, OA, and RA, the Tet 3 expression level in the FLS nucleus increased significantly from 24 hours after TNFα stimulation and became prominent at 96 hours after stimulation (FIG. 7, 8).
(Example 4) DNA methylation level in FLS after stimulation with inflammatory cytokines In order to examine the DNA demethylation level, stimulation with TNFα was performed on RA-derived FLS, and the change in the level of 5hmC (genomic DNA) was examined. For the extraction of gDNA, the Spingen genomic DNA purification kit (m. biotech) was used, and the expression level of 5hmC in gDNA was analyzed by the Dot Blot method. Rabbit anti-5-methylcytosine (5-mC) polyclonal Ab (ActiveMotif) for primary antibodies, ECL TM Anti-Rabbit IgG for secondary antibodies, HRP linked whole antibody, GE health EC Blotting detection reagent (GE healthcare, UK) was used. As controls, 5-Methylcytosine & 5-Hydroxymethylcytosine DNA Standard Set was used. Quantification was performed using Light-capture (AE-6972, Atto, Tokyo, Japan) for image capturing, and CS Analyzer, version 3.0 (Atto) for image analysis.
As a result, FLS stimulated with TNFα significantly increased the level of 5 hmC at 96 hours after stimulation (FIG. 9).
(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 TNFα and secretion of RA-derived FLS Evaluated. Specifically, 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 TNFα (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 TNFα. Tet 1 siRNA was used as a control. Two types of siRNA (Primer Set Human from Life technologies) used in the preliminary study were used for each gene, that is, siRNA IDs: HSS129586, HSS123253 for TET1, HSS123253, HSS1233Hor TET2, HSS15364T used. As a control, Low GC Duplex (Invitrogen) from Stealth RNAi Negative Control Duplexes was used. 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. Regarding the amount of secreted protein such as IL-6, IL-8, VEGF, CCL2, etc., the BD 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). TaqMan (R) GeneExpress Assays CCL2 (Hs00234140_m1) was used for expression analysis of CCL2 mRNA.
As a result, among many cytokines, chemokines, and MMPs that have been suggested to be related to the pathological condition of RA, IL-6, IL-8, VEGF, CCL2, MMP3, etc. were found to induce TNFα-dependent expression. However, among these, only CCL2 inhibited TNFα-dependent expression induction by Tet 3 knockdown (FIG. 10). This phenomenon was also confirmed at the mRNA and secretory protein levels. In addition, among the adhesion molecules, TAMα-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. Therefore, it was suggested that 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. Although detection was attempted for IL-1β, TNFα, and IL-17α, 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 TNFα was performed, and the invasion ability of RA-derived FLS was evaluated by Scratch assay. Specifically, FLS obtained by knocking down Tet 3 with siRNA was cultured for 96 hours in a medium (10% FCS-containing DMEM) containing TNFα (1 ng / ml), washed, and scratch was applied to the dish surface to which FLS was adhered. The observation was performed for a maximum of 48 hours while culturing in 0.1% FCS-containing DMEM not containing TNFα. Tet 1 siRNA was used as a control.
As a result, TNF3 knockdown completely inhibited TNFα-dependent induction of FLS infiltration (FIG. 11). Suggested that selective inhibition of Tet 3 leads to the prevention and treatment of RA by inhibiting invasiveness, one of the most important aggressive phenotypes found in synovial fibroblasts of rheumatoid arthritis patients It was done.
 本発明により、これまでの治療剤の標的とは異なる因子であるTet 3を標的とした、関節炎の予防及び/又は治療剤あるいは診断キットが提供される。新たな作用機序を有する治療薬が提供されることにより、既存の治療法では病状回復効果が認められない、又は病状回復効果が十分でない関節炎患者、及び既存の治療薬を継続使用することで該治療薬について耐性を獲得した関節炎患者に対してもより良い治療を提供し得る。さらに本発明は、関節炎を未然に防ぐ予防目的、及び関節炎が一時寛解した患者が病気の再発を予防する目的にも使用され得る。また、本発明によれば、Tet 3の発現又は機能を阻害することで関節炎の予防及び/又は治療効果を発揮する、関節炎の新規予防・治療薬の候補物質をスクリーニングすることが可能である。
 本出願は、日本で出願された特願2014−174638(出願日:平成26年8月28日)を基礎としており、その内容はすべて本明細書に包含されるものとする。
According to the present invention, 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. By providing therapeutic drugs with new mechanisms of action, patients with arthritis who do not have a pathological recovery effect with existing therapies or who do not have sufficient pathological recovery effects, and continue to use existing drugs Better treatment can also be provided for arthritic patients who have acquired resistance to the therapeutic agent. Furthermore, 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. In addition, according to the present invention, it is possible to screen for a candidate substance for a novel prophylactic / therapeutic agent for arthritis that exhibits an effect of preventing and / or treating arthritis by inhibiting the expression or function of Tet3.
This application is based on Japanese Patent Application No. 2014-174638 (filing date: August 28, 2014) filed in Japan, the contents of which are incorporated in full herein.

Claims (15)

  1.  Tet 3(Ten−Eleven translocation 3)の発現阻害物質を含有する、関節炎の予防及び/又は治療剤。 A preventive and / or therapeutic agent for arthritis, which contains an expression inhibitor of Tet 3 (Ten-Eleven tranlocation 3).
  2.  Tet 3の発現阻害物質が、
    (a)Tet 3遺伝子の転写産物に対するアンチセンス核酸、
    (b)Tet 3遺伝子の転写産物に対するリボザイム核酸、又は
    (c)Tet 3遺伝子の転写産物に対してRNAi活性を有する核酸もしくはその前駆体である、請求項1に記載の剤。
    Tet 3 expression inhibitor is
    (A) an antisense nucleic acid against a transcript of the Tet 3 gene,
    The agent according to claim 1, which is (b) a ribozyme nucleic acid for a transcription product of Tet 3 gene, or (c) a nucleic acid having RNAi activity for a transcription product of Tet 3 gene or a precursor thereof.
  3.  関節炎が関節リウマチ、乾癬性関節炎または脊椎関節炎である、請求項1または2に記載の剤。 The agent according to claim 1 or 2, wherein the arthritis is rheumatoid arthritis, psoriatic arthritis or spondyloarthritis.
  4.  以下の(1)~(3)の工程を含む、関節炎の予防及び/又は治療薬のスクリーニング方法:
    (1)Tet 3遺伝子もしくは該遺伝子の転写調節領域の制御下にあるレポータータンパク質をコードする核酸を含む細胞を、被検物質に接触させる工程、
    (2)前記細胞におけるTet 3遺伝子もしくはTet 3タンパク質又はレポータータンパク質の発現量を測定する工程、
    (3)被検物質の非存在下において測定した場合と比較して、Tet 3遺伝子もしくはTet 3タンパク質又はレポータータンパク質の発現量を低下させた被検物質を、関節炎の予防及び/又は治療薬の候補として選択する工程。
    A method for screening a prophylactic and / or therapeutic agent 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.
  5.  Tet 3と被検物質とを接触させ、Tet 3と結合能を有する被検物質を関節炎の予防及び/又は治療剤の候補として選択することを特徴とする、関節炎の予防及び/又は治療薬のスクリーニング方法。 A prophylactic and / or therapeutic drug for arthritis, characterized by contacting Tet 3 with a test substance and selecting a test substance capable of binding to Tet 3 as a prophylactic and / or therapeutic drug for arthritis. Screening method.
  6.  関節炎の予防及び/又は治療薬の候補として選択された被検物質を関節炎モデルに適用し、該モデルにおける炎症反応を抑制するか否かを検定することをさらに含む、請求項5に記載の方法。 The method according to claim 5, further comprising applying a test substance selected as a candidate for a prophylactic and / or therapeutic agent for arthritis to an arthritis model and testing whether to suppress an inflammatory response in the model. .
  7.  以下の(1)~(3)の工程を含む、関節炎の予防及び/又は治療薬のスクリーニング方法:
    (1)滑膜線維芽細胞を、被検物質に接触させる工程、
    (2)前記細胞のゲノムの5−メチルシトシン(5mC)の脱メチル化または浸潤性の程度を測定する工程、
    (3)被検物質の非存在下において測定した場合と比較して、前記細胞のゲノムの5−メチルシトシン(5mC)の脱メチル化または浸潤性を抑制した被検物質を、関節炎の予防及び/又は治療薬の候補として選択する工程。
    A method for screening 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.
  8.  関節炎が関節リウマチ、乾癬性関節炎または脊椎関節炎である、請求項4~7のいずれか1項に記載の方法。 The method according to any one of claims 4 to 7, wherein the arthritis is rheumatoid arthritis, psoriatic arthritis or spondyloarthritis.
  9.  被験者由来の試料から、下記(a)または(b)を用いてTet 3遺伝子の転写産物または翻訳産物を検出または定量することを含む、関節炎の検査方法:
    (a)Tet 3遺伝子の転写産物を特異的に検出し得る核酸プローブまたは核酸プライマー
    (b)Tet 3遺伝子の翻訳産物を特異的に認識する抗体。
    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.
  10.  関節炎が関節リウマチ、乾癬性関節炎または脊椎関節炎である、請求項9に記載の方法。 The method according to claim 9, wherein the arthritis is rheumatoid arthritis, psoriatic arthritis or spondyloarthritis.
  11.  下記(a)および/または(b):
    (a)Tet 3遺伝子の転写産物を特異的に検出し得る核酸プローブまたは核酸プライマー
    (b)Tet 3遺伝子の翻訳産物を特異的に認識する抗体
    を含有してなる、関節炎検査用キット。
    (A) and / or (b) below:
    (A) a nucleic acid probe or nucleic acid primer capable of specifically detecting a Tet 3 gene transcription product; and (b) an arthritis test kit comprising an antibody that specifically recognizes a translation product of the Tet 3 gene.
  12.  関節炎が関節リウマチ、乾癬性関節炎または脊椎関節炎である、請求項11に記載のキット。 The kit according to claim 11, wherein the arthritis is rheumatoid arthritis, psoriatic arthritis or spondyloarthritis.
  13.  Tet 3(Ten−Eleven translocation 3)の発現阻害物質の有効量を対象に投与することを含む、関節炎の予防及び/又は治療方法。 A method for preventing and / or treating arthritis, comprising administering to a subject an effective amount of an expression-inhibiting substance of Tet 3 (Ten-Eleven tranlocation 3).
  14.  関節炎の予防及び/又は治療に使用するための、Tet 3(Ten−Eleven translocation 3)の発現阻害物質。 Expression inhibitor of Tet 3 (Ten-Eleven tranlation 3) for use in the prevention and / or treatment of arthritis.
  15.  関節炎の予防及び/又は治療剤を製造するための、Tet 3(Ten−Eleven translocation 3)の発現阻害物質の使用。 Use of an expression-inhibiting substance of Tet 3 (Ten-Eleven tranlocation 3) to produce an agent for preventing and / or treating arthritis.
PCT/JP2015/074556 2014-08-28 2015-08-24 Prophylactic/therapeutic agent for arthritis, test kit for arthritis, and method for screening for prophylactic/therapeutic agent for arthritis WO2016031996A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2016545659A JPWO2016031996A1 (en) 2014-08-28 2015-08-24 Arthritis prevention / treatment agent, test kit, and screening method for arthritis prevention / treatment agent

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 (en) 2016-03-03

Family

ID=55399876

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2015/074556 WO2016031996A1 (en) 2014-08-28 2015-08-24 Prophylactic/therapeutic agent for arthritis, test kit for arthritis, and method for screening for prophylactic/therapeutic agent for arthritis

Country Status (2)

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

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020206055A1 (en) * 2019-04-02 2020-10-08 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

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2013021932A (en) * 2011-07-15 2013-02-04 Chiba Univ Method for predicting efficacy of anti-il-6 receptor antibody therapy to rheumatoid arthritis

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2013021932A (en) * 2011-07-15 2013-02-04 Chiba Univ Method for predicting efficacy of anti-il-6 receptor antibody therapy to rheumatoid arthritis

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 (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020206055A1 (en) * 2019-04-02 2020-10-08 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

Also Published As

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

Similar Documents

Publication Publication Date Title
JP7175526B2 (en) Preventive/therapeutic agents for diseases related to cell migration regulation and disease activity assessment/prognostic evaluation for pulmonary interstitial diseases
JP2015173601A (en) Screening method for cancer stem cell proliferation inhibition material
JP7177439B2 (en) Pro-inflammatory factor expression inhibitor, screening method for active ingredient thereof, expression cassette useful for said method, diagnostic agent, and diagnostic method
JP2011032236A (en) Inhibitor for tumor neovascularization
JP5794514B2 (en) Tumor angiogenesis inhibitor
WO2016031996A1 (en) Prophylactic/therapeutic agent for arthritis, test kit for arthritis, and method for screening for prophylactic/therapeutic agent for arthritis
JP6226315B2 (en) Inflammatory disease preventive / therapeutic agent and screening method for inflammatory disease prophylactic / therapeutic agent
EP2153847A1 (en) Gene sensitive to bone/joint disease and use thereof
WO2010008084A1 (en) Novel use application of sugar chain-recognizing receptor
JP2011502108A (en) Cancer preventive / therapeutic agent
EP2123303A1 (en) Ameliorating agent for insulin resistance
JP6653120B2 (en) Anti-inflammatory drugs based on IKAROS inhibition
JP5669271B2 (en) Tumor angiogenesis inhibitor
WO2011115271A1 (en) Tumor angiogenesis inhibitor
US8309687B2 (en) Biomarker specific for cancer
JP2023055804A (en) Therapy, diagnosis and screening using card 14
WO2011019065A1 (en) Tumor angiogenesis inhibitor
WO2011115268A1 (en) Tumor angiogenesis inhibitor
JP2008263851A (en) Treatment target of heatstroke
JP2011105669A (en) Antitumor agent and method of screening antitumor agent
EP1577387A1 (en) Preventives/remedies for cancer
JP2010111623A (en) New use of sugar chain recognition receptor

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