WO2011019074A1 - Nucleic acid that controls fibrosis of cells or organs - Google Patents

Nucleic acid that controls fibrosis of cells or organs Download PDF

Info

Publication number
WO2011019074A1
WO2011019074A1 PCT/JP2010/063728 JP2010063728W WO2011019074A1 WO 2011019074 A1 WO2011019074 A1 WO 2011019074A1 JP 2010063728 W JP2010063728 W JP 2010063728W WO 2011019074 A1 WO2011019074 A1 WO 2011019074A1
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
fibrosis
cell
microrna
expression
Prior art date
Application number
PCT/JP2010/063728
Other languages
French (fr)
Japanese (ja)
Inventor
伸晃 江藤
将矩 兼松
Original Assignee
協和発酵キリン株式会社
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 協和発酵キリン株式会社 filed Critical 協和発酵キリン株式会社
Publication of WO2011019074A1 publication Critical patent/WO2011019074A1/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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • 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
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • 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.
    • C12N2310/141MicroRNAs, miRNAs
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/08Hepato-biliairy disorders other than hepatitis
    • G01N2800/085Liver diseases, e.g. portal hypertension, fibrosis, cirrhosis, bilirubin

Definitions

  • the present invention relates to a cell or organ fibrosis control agent using a nucleic acid, a diagnostic or therapeutic agent for a disease caused by a fibrotic disorder, a cell or organ fibrosis control method, and a screening method for a cell or organ fibrosis control agent About.
  • MicroRNA which is a kind of nucleic acid, is a small non-coding single-stranded RNA consisting of about 22 nucleotides that are not translated into protein, and is known to exist in many organisms including humans (Non-Patent Documents). 1, 2). MicroRNAs are generated from genes that are transcribed into single or clustered microRNA precursors. In other words, the primary transcript, primary-microRNA (pri-miRNA), is first transcribed from the gene, and then in a stepwise process from pri-miRNA to mature microRNA, a precursor of about 70 bases with a characteristic hairpin structure. -microRNA (pre-miRNA) is generated from pri-miRNA. Furthermore, mature microRNA is generated from pre-miRNA by Dicer-mediated processing (Non-patent Document 3).
  • Mature microRNAs are thought to be involved in post-transcriptional control of gene expression by binding complementarily to the target mRNA and suppressing mRNA translation, or by degrading the mRNA.
  • the mechanism by which microRNA suppresses the expression of target mRNA is not completely clarified, an outline has been elucidated by recent studies.
  • MicroRNA binds to a partially complementary sequence in the 3'-untranslated region (3'-UTR) of the target mRNA, suppresses its translation, or suppresses expression by degrading the target mRNA To do.
  • 3'-UTR 3'-untranslated region
  • the above complementarity may not be perfect, it has been shown that complementarity of the second to eighth bases from the 5′-end side of the microRNA is particularly important.
  • Non-patent Document 4 Is sometimes referred to as a “seed sequence” (Non-patent Document 4). It has been shown that microRNAs with a common seed sequence suppress the expression of a common target mRNA even if other sequences are different. Therefore, RNA having microRNA-like activity can be designed by using a sequence complementary to the sequence present at the 3′-end of any mRNA as a seed sequence. Unlike siRNA, microRNA usually refers only to RNA that is “naturally present in cells”. Therefore, a microRNA-like sequence designed in this way may be particularly referred to as “artificial microRNA”.
  • the microRNA database miRBase (http://microrna.sanger.ac.uk/) contains 706 human, 547 mouse, and 286 rat microRNAs. .
  • miR-181 Non-patent Document 5
  • miR-375 involved in insulin secretion
  • Non-patent document 6 is only a small part, and many of them have unclear physiological activity.
  • studies using nematodes and Drosophila have revealed that microRNAs play various important roles in the development and differentiation of living organisms. There has been a report suggesting a relationship (Non-patent Document 7).
  • Fibrosis is a phenomenon in which the part called connective tissue that constitutes tissues such as internal organs grows abnormally and occurs in various tissues and organs such as lung, kidney, liver, heart, skin, blood vessels, and causes disease It is known. For example, when fibrosis occurs in the myocardium, abnormalities occur in the function of the heart, and symptoms such as dyspnea and increased heart rate (palpitations) appear. In addition, bone atrophy and degeneration in rheumatoid arthritis and the pathology of cirrhosis, which indicates fibrosis of the entire liver, are well known as examples of abnormal growth of the connective tissue, that is, fibrosis.
  • Tissue fibrosis is mainly caused by abnormal production of the extracellular matrix of fibroblasts present in the tissue, but the epithelial cells constituting the tissue are transformed into mesenchymal cells together with the extracellular matrix. It has been clarified that starting to produce a substrate also contributes, and these are called epithelial-mesenchymal transition (EMT) (Non-patent Document 8).
  • EMT epithelial-mesenchymal transition
  • MicroRNA research so far has been progressing mainly in cancer research, and in many cancers, microRNA expression is different from normal tissues, and cancer can be classified by microRNA expression profile analysis.
  • Non-Patent Document 9 It is also known that about half of the human microRNAs found so far exist in chromosomal abnormalities or fragile sites of chromosomes known in human cancer (Non-patent Document 10).
  • general immunosuppressive drugs such as corticosteroids and other anti-inflammatory treatments are currently used for treatment of organ fibrosis.
  • the mechanisms involved in controlling fibrosis appear to be a separate phenomenon from that of inflammation, and anti-inflammatory therapy is not always effective in reducing or preventing fibrosis. It cannot be said that the clinical needs are satisfied (Non-patent Document 11).
  • Non-patent Document 11 Thus, there is still a need to develop new therapies that modulate fibrotic disorders, including reducing or preventing fibrosis.
  • microRNA Since microRNA is involved in the control of the expression of various genes, abnormalities in microRNA are expected to be involved in various human diseases.
  • miR-15a / miR-16 cluster is included in chromosome 13q14, which is deleted in miR-29cB cell chronic lymphocytic leukemia (B-CLL), and this deletion is one of the causes of B-CLL. That are predicted to be connected (Non-patent Document 12).
  • B-CLL miR-29 and miR-181 expression is further reduced, and one of the targets is Tcl1, which is known as a proto-oncogene. (Non-patent Document 13) is known.
  • Non-patent Documents 14 and 15 expression of Let-7, which is one of microRNAs, is decreased, and one of its targets is Ras known as a proto-oncogene (Non-patent Documents 14 and 15).
  • microRNAs such as miR-127 and miR-124a is reduced by hypermethylation modification of genes in cancer, and Bcl6 and Cdk6 known as proto-oncogenes are the targets (Non-patent Document 16, 17).
  • Many microRNAs have decreased expression in cancer cells, but there are also microRNAs in which gene amplification and overexpression are seen in cancer cells. For example, in a region where gene amplification is observed in malignant lymphoma, there are 6 types of microRNA clusters (miR-17-92).
  • Non-patent Document 18 a gene called BIC, which has been previously overexpressed in Hodgkin's lymphoma and was a candidate oncogene that does not encode a protein, encodes miR-155 (Non-patent Document 19).
  • microRNAs whose expression changes in fibrotic organs, analyzing their functions, and elucidating the relationship with diseases.
  • finding microRNAs that promote or suppress fibrosis of cells or organs not only helps understand the mechanism of fibrosis, but also develops diagnostic and therapeutic agents for human organ fibrosis, and further It is expected to lead to new diagnostic methods and treatments for the fibrosis used.
  • An object of the present invention is to provide a nucleic acid and a fibrosis control agent useful for controlling fibrosis of cells or organs, and methods for using them.
  • the fibrosis regulator refers to a fibrosis inhibitor and a fibrosis promoter.
  • a cell or organ fibrosis control agent comprising any one of the following nucleic acids (a) to (h) as an active ingredient.
  • Nucleic acid comprising a base sequence represented by any of SEQ ID NOs: 1-27, 28-594
  • b Containing a nucleic acid comprising a base sequence represented by any of SEQ ID NOs: 1-27, 28-594
  • a nucleic acid consisting of a base sequence having a nucleotide sequence of 90% or more
  • c a sequence represented by any one of SEQ ID NOs: 1 to 27 and 28 to 594
  • e a base represented by any one of SEQ ID NOs: 1 to 27 and 28
  • a cell or organ fibrosis control agent comprising, as an active ingredient, a nucleic acid having a base sequence complementary to the base sequence of the nucleic acid described in [1].
  • a cell or organ fibrosis control agent comprising a vector that expresses the nucleic acid according to any one of [1] to [3] as an active ingredient.
  • a cell or organ fibrosis control agent comprising, as an active ingredient, a substance that suppresses the expression of the gene having the target base sequence of the nucleic acid according to [1].
  • a cell or organ fibrosis control agent comprising as an active ingredient a substance that promotes expression of a gene having the target base sequence of the nucleic acid according to [1].
  • [7] The cell or organ fibrosis regulator according to [5] or [6] above, wherein the substance that suppresses or promotes expression is a nucleic acid.
  • a cell or organ fibrosis control agent comprising the vector expressing the nucleic acid according to [7] as an active ingredient.
  • the nucleic acid according to any one of [1] to [3] above, the vector according to [4] above, or the substance according to any of [5] to [8] above as an active ingredient A therapeutic or diagnostic agent for diseases caused by fibrosis of cells or organs.
  • [11] A disease caused by fibrosis of a cell or an organ containing as an active ingredient a reagent for detecting the expression level of the nucleic acid according to [1], a mutation of the nucleic acid, or a mutation of a genome encoding the nucleic acid Diagnostics.
  • a method for treating a disease caused by fibrosis of a cell or organ characterized by comprising: [15] Diagnosis of a disease caused by fibrosis of a cell or organ, wherein the expression level of the nucleic acid according to [1], a mutation of the nucleic acid, or a mutation of a genome encoding the nucleic acid is detected Method.
  • nucleic acid according to any one of [1] to [3] above, the vector according to [4] above, or the above [for manufacturing a therapeutic agent for a disease caused by fibrosis of a cell or organ Use of the substance according to any one of [5] to [8].
  • [20] Diseases caused by fibrosis of cells or organs are acute renal failure, glomerulonephritis, vasculitis, diabetic nephropathy, hypertensive nephrosclerosis, HIV nephropathy, IgA nephropathy, lupus nephritis, stroma.
  • An agent for controlling expression of a target gene of a nucleic acid which is an active ingredient of a fibrosis regulator for cells or organs, containing the nucleic acid according to any one of [1] to [3] as an active ingredient.
  • a nucleic acid target gene expression control agent which is an active ingredient of a cell or organ fibrosis control agent, comprising the vector according to [4] as an active ingredient.
  • a method for controlling fibrosis of a cell or organ comprising using the nucleic acid according to any one of [1] to [3] above.
  • a method for controlling fibrosis of a cell or organ characterized by using the vector according to [4] above.
  • a method for controlling fibrosis of a cell or organ comprising using a substance that suppresses or promotes the expression of the target gene of the nucleic acid according to [1].
  • [26] The method for controlling fibrosis of a cell or organ according to the above [25], wherein the substance that suppresses or promotes expression is a nucleic acid.
  • [30] A method for controlling expression of a target gene of a nucleic acid, which is an active ingredient of a fibrosis control agent for cells or organs, using the vector according to [4] above.
  • [31] A screening method for a cell or organ fibrosis regulator using as an index the promotion or suppression of the expression or function of the nucleic acid according to [1].
  • a fibrosis inhibitor or fibrosis promoter for cells or organs a diagnostic or therapeutic agent for diseases caused by abnormal extracellular matrix production of cells or EMT, and an expression inhibitor for target genes of nucleic acids such as microRNA
  • an expression promoter, a method for suppressing cell fibrosis, or a method for promoting fibrosis can be provided.
  • FIG. 1 shows the results of analysis of collagen-I gene expression by real-time RT-PCR.
  • the expression of Collagen I in the liver after administration of control RNA, miR-30b, miR-486, let-7i is shown as a relative ratio to the control.
  • FIG. 2 shows the results of analysis of TGF- ⁇ gene expression by real-time RT-PCR.
  • the expression of TGF- ⁇ in the liver after administration of control microRNA, miR-30b, miR-486, let-7i is shown as a relative ratio to the control.
  • the nucleic acid used in the present invention may be any molecule as long as nucleotides and molecules having functions equivalent to the nucleotides are polymerized, such as RNA that is a ribonucleotide polymer, DNA that is a deoxyribonucleotide polymer. , A polymer in which ribonucleotides and deoxyribonucleotides are mixed, and a nucleotide polymer containing a nucleotide analog, and further a nucleotide polymer containing a nucleic acid derivative.
  • the nucleic acid in the present invention may be a single-stranded nucleic acid or a double-stranded nucleic acid.
  • the double-stranded nucleic acid also includes a double-stranded nucleic acid in which one strand is hybridized under stringent conditions to the other strand.
  • nucleotide analogues are used to improve nuclease resistance, to stabilize, to increase affinity with complementary strand nucleic acids, to increase cell permeability, or to be visualized as compared to RNA or DNA.
  • any molecule may be used as long as it is a molecule obtained by modifying ribonucleotides, deoxyribonucleotides, RNA, or DNA. Examples thereof include sugar moiety-modified nucleotide analogs and phosphodiester bond-modified nucleotide analogs.
  • the sugar moiety-modified nucleotide analog may be any one obtained by adding or substituting any chemical structural substance to a part or all of the chemical structure of the sugar of the nucleotide.
  • any chemical structural substance for example, 2'-O-methyl Nucleotide analogues substituted with ribose, nucleotide analogues substituted with 2'-O-propylribose, nucleotide analogues substituted with 2'-methoxyethoxyribose, substituted with 2'-O-methoxyethylribose Nucleotide analogues, nucleotide analogues substituted with 2'-O- [2- (guanidinium) ethyl] ribose, nucleotide analogues substituted with 2'-O-fluororibose, introducing a bridging structure into the sugar moiety Bridged Nucleic Acid (BNA) having two circular structures, more specifically,
  • the phosphodiester bond-modified nucleotide analog may be any one obtained by adding or substituting an arbitrary chemical substance to a part or all of the chemical structure of a phosphodiester bond of a nucleotide.
  • Examples include nucleotide analogues substituted with thioate linkages, nucleotide analogues substituted with N3'-P5 'phosphoramidate linkages [Cell engineering, 16 , 1463-1473 (1997)] [RNAi method And Antisense, Kodansha (2005)].
  • nucleic acid derivative in order to improve nuclease resistance, to stabilize, to increase affinity with a complementary strand nucleic acid, to increase cell permeability or to be visualized as compared with a nucleic acid, a different chemical is used. Any molecule can be used as long as it is a molecule to which a substance is added, for example, 5′-polyamine addition derivative, cholesterol addition derivative, steroid addition derivative, bile acid addition derivative, vitamin addition derivative, Cy5 addition derivative, Cy3 addition derivative, 6-FAM Examples include addition derivatives, biotin addition derivatives, and the like.
  • nucleic acid examples include nucleic acids represented by the following (a) to (k).
  • Nucleic acid comprising a base sequence represented by any of SEQ ID NOs: 1 to 594 (b) Nucleic acid comprising 17 to 28 bases comprising a nucleic acid comprising a base sequence represented by any of SEQ ID NOs: 1 to 594 (C) a nucleic acid comprising a base sequence represented by any one of SEQ ID NOs: 1 to 594 and having a nucleotide sequence having 90% or more identity (d) comprising a base sequence represented by any of SEQ ID NOs: 1 to 594 Nucleic acid that hybridizes with a complementary strand of nucleic acid under stringent conditions (e) A nucleic acid comprising the second to eighth base sequences of the base sequence represented by any one of SEQ ID NOs: 1 to 594 (f) (a) to ( a double-stranded nucleic acid comprising the nucleic acid of e) and a nucleic acid
  • a single-stranded nucleic acid or a nucleic acid containing the single-stranded nucleic acid (i) a nucleic acid consisting of a base sequence represented by any of SEQ ID NOs: 595 to 1281 (j) represented by any of SEQ ID NOs: 595 to 1281
  • a nucleic acid comprising a nucleotide sequence having 90% or more identity to the nucleotide sequence (k) a nucleic acid that hybridizes under stringent conditions with a complementary strand of a nucleic acid comprising the nucleotide sequence represented by any of SEQ ID NOs: 595 to 1281 .
  • microRNA refers to single-stranded RNA having a length of 17 to 28 bases.
  • the surrounding genomic sequence containing the microRNA sequence has a sequence that can form a hairpin structure, and the microRNA can be excised from either strand of the hairpin.
  • MicroRNAs complementarily bind to their target mRNA and suppress mRNA translation, or promote post-transcriptional control of gene expression by promoting mRNA degradation.
  • microRNA examples include human microRNA having a base sequence represented by any of SEQ ID NOs: 1 to 27. Furthermore, as a microRNA having the same function as the human microRNA consisting of the base sequence represented by any of SEQ ID NOs: 1-27, it is represented by any of SEQ ID NOs: 28-486, which is an ortholog of the human microRNA. A nucleic acid having a base sequence can be mentioned. As a specific example, the ortholog of the human microRNA of SEQ ID NO: 1 includes those consisting of the base sequences represented by SEQ ID NOs: 28 to 33 and 37 to 39. Table 1 shows a correspondence table between microRNAs having the base sequences represented by SEQ ID NOs: 1 to 27 and their orthologs.
  • hsa Homo sapiens, humans; mmu, Mus musculus, mice; rno, Rattus norvegicus, rats; cgr, Cricetulus griseus, Chinese hamsters; age, Ateles geoffroyi, red spider monkeys; Mnea catamarin; mml, Macaca mulatta, rhesus monkey; mne, Macaca nemestrina, pigtail monkey; pbi, Pygathrix bieti, black goldfish; ggo, Gorilla gorilla, gorilla; ppa, Pan paniscus, bonobo; ptr, Pan trogopy; Ssy, Symphalangus syndactylus, octopus gibbon; lca, Lemur catta, ring-tailed lemur; oan, Ornithorhynchus anatinus
  • microRNA As a mechanism by which microRNA suppresses translation of mRNA of its target gene, mRNA having a base sequence complementary to the 2-8th base sequence on the 5 'end side of microRNA is recognized as a microRNA target gene. [Current Biology, 15 , R458-R460 (2005)]. By this mechanism, the expression of the mRNA is suppressed by the microRNA. Accordingly, microRNAs having the same base sequence on the 2nd to 8th positions on the 5 ′ end side have the same function by suppressing the expression of the same mRNA.
  • a base represented by any one of SEQ ID NOs: 487 to 594 as a microRNA having the same base sequence at the 5 'end side from the microRNA having the base sequence represented by any one of SEQ ID NOs: 1 to 27 A nucleic acid consisting of a sequence can be mentioned.
  • the microRNA for the microRNA consisting of the base sequence represented by SEQ ID NO: 1, microRNA consisting of the base sequence represented by SEQ ID NOs: 487 and 488 can be mentioned.
  • Artificial microRNA is also included in microRNA.
  • Table 2 shows a correspondence table between the microRNAs having the base sequences represented by SEQ ID NOs: 1 to 27 and the microRNAs having the same base sequence at the second to eighth positions on the 5 ′ end. MicroRNAs having a common seed sequence are considered to have the same function because their target base sequences are considered identical.
  • a microRNA precursor is also preferably used as the nucleic acid.
  • the microRNA precursor is a nucleic acid having a length of about 50 to about 200 bases, more preferably about 70 to about 100 bases including the above-mentioned nucleic acid, and can form a hairpin structure.
  • MicroRNA is produced from a microRNA precursor through processing by a protein called Dicer.
  • microRNA precursor examples include a nucleic acid having a base sequence represented by SEQ ID NO: 595 for human microRNA of SEQ ID NO: 1.
  • a nucleic acid having a base sequence represented by any of SEQ ID NOs: 2 to 594 a nucleic acid having a base sequence represented by any of SEQ ID NOs: 596 to 1281 can be exemplified.
  • Table 3 shows the correspondence between the microRNA and the microRNA precursor in the present invention.
  • the microRNA precursor includes an artificial microRNA precursor.
  • a nucleic acid having 90% or more identity with the nucleotide sequence represented by any of SEQ ID NOs: 1-1811 is BLAST [J. Mol. Biol., 215 , 403 (1990)] or FASTA [ Methods in Enzymology, 183 , 63 (1990)], etc., and at least 90% or more, preferably 93% of the nucleic acid consisting of the base sequence represented by any of SEQ ID NOs: 1-1811 It means that the nucleic acid has the identity of 95% or more, more preferably 96% or more, particularly preferably 97% or more, and most preferably 98% or more.
  • the stringent conditions in the above are, for example, 7.5 mL, 1 M Na 2 HPO 4 (pH 7.2) 0.6 mL, 10% SDS 21 mL, 50x Denhardt's solution 0.6 mL, 10 Add the other strand labeled with 32 P-ATP to the Hybridization buffer consisting of 0.3 mL of mg / mL sonicated salmon sperm DNA, react at 50 ° C overnight, then react at 50 ° C for 10 minutes, 5x SSC / 5% SDS solution And then washed with 1 ⁇ SSC / 1% SDS solution at 50 ° C. for 10 minutes, and then the membrane is taken out and exposed to an X-ray film to detect the signal.
  • Those skilled in the art can appropriately select the hybridization conditions that give the same stringency.
  • any method for detecting the expression of a nucleic acid such as microRNA using a nucleic acid can be used as long as it can detect the presence of a nucleic acid such as microRNA or a microRNA precursor in a sample.
  • a nucleic acid such as microRNA or a microRNA precursor in a sample.
  • any method can be used as long as it can detect the presence of a nucleic acid such as microRNA or a microRNA precursor in a sample.
  • any method can be used as long as it can detect a mutation in the base sequence of a nucleic acid such as microRNA or a microRNA precursor in a sample.
  • a method for detecting a heteroduplex formed by hybridization of a nucleic acid having a non-mutated base sequence and a nucleic acid having a mutant base sequence, or by directly sequencing a base sequence derived from a specimen A method for detecting the presence or absence can be given.
  • the vector expressing nucleic acid may be any vector as long as it is designed to biosynthesize nucleic acid such as microRNA by being introduced into a cell and transcribed.
  • vectors capable of expressing nucleic acids such as microRNA in cells include pCDNA6.2-GW / miR (Invitrogen), pSilencer4.1-CMV (Ambion), pSINsi-hH1 DNA (Takara Bio), pSINsi-hU6UDNA (Takara Bio), pENTR / U6 (Invitrogen), microRNA Archive (Takara Bio), pMIRRNA1 (System Bioscience), etc. it can.
  • a target gene As a method for suppressing the expression of a gene having a target base sequence of a nucleic acid such as microRNA (hereinafter referred to as a target gene), the activity of suppressing the expression of mRNA having a target base sequence of a nucleic acid such as microRNA is utilized. Any method may be used as long as it suppresses the expression of a gene having the target base sequence.
  • suppression of expression here includes a case where the translation of mRNA is suppressed, and a case where the amount of protein translated from mRNA is reduced by cleaving or decomposing mRNA.
  • Specific examples of substances that suppress the expression of mRNA having the target base sequence include nucleic acids such as siRNA and antisense oligonucleotides.
  • the siRNA can be prepared based on the continuous sequence information of the mRNA [Genes Dev., 13 , 3191 (1999)].
  • the number of residues of the base constituting one strand of siRNA is preferably 17 to 30 residues, more preferably 18 to 25 residues, still more preferably 19 to 23 residues.
  • the microRNA is a single-stranded RNA having a length of 17 to 28 bases containing a sequence complementary to a continuous 7 base sequence existing in the 3 ′ untranslated region of the mRNA of the target gene as the second to eighth base sequences. Some artificial microRNAs are also included. When the sequence including the sequence and extending it back and forth forms a hairpin structure, and the microRNA sequence is RNA that can be excised from any one strand of the hairpin structure in the cell by the microRNA biosynthetic pathway, The extended sequence is called an artificial microRNA precursor. Artificial microRNAs and artificial microRNA precursors can be designed using the method as described above using a gene whose expression is to be suppressed as a target gene.
  • the target base sequence of nucleic acid such as microRNA is a base sequence consisting of several bases recognized by nucleic acid such as microRNA in the present invention, and the expression of mRNA having the base sequence is such as microRNA in the present invention.
  • the base sequence complementary to the 2-8th base sequence on the 5 ′ end side of the microRNA is that translation of mRNA having the base sequence is suppressed by the microRNA [Current Biology, 15 , R458-R460]. (2005)], a base sequence complementary to the second to eighth base sequences on the 5 ′ end side of nucleic acids such as microRNA in the present invention can be mentioned as the target base sequence.
  • an mRNA containing a sequence that perfectly matches the 3 'UTR base sequence group of human mRNAs by preparing a target sequence complementary to the 2-8th base sequence on the 5' end side of the microRNA. Can be determined by selecting by a method such as a character string search.
  • the 3'UTR base sequence group of human mRNA should be prepared using the genome sequence and gene position information that can be obtained from "UCSC Human Genome Browser Gateway (http://genome.ucsc.edu/cgi-bin/hgGateway)". Can do.
  • a method for expressing a nucleic acid such as microRNA in a cell includes a method using a nucleic acid that expresses a gene encoding the microRNA when introduced into the cell.
  • the nucleic acid in addition to DNA, RNA, or nucleotide analogues, these chimeric molecules or derivatives of the nucleic acids can also be used.
  • the nucleic acid can be designed in the same manner as Pre-miRTMmimiRNA Precursor Molecules (Ambion) or miRIDIAN microRNA Mimics (GE Healthcare) to express nucleic acids such as microRNA in cells. it can.
  • any method may be used as long as microRNA can finally be produced in the cell.
  • RNA in addition to introducing single-stranded RNA as a microRNA precursor, (2) There is a method for introducing microRNA itself and RNA consisting of complementary strands of microRNA and 100% -matched double-stranded RNA, and (3) double-stranded RNA assuming the state after microRNA is cleaved into Dicer.
  • Examples of products using such a method include miCENTURY OX Precursor (manufactured by B-Bridge), miCENTURY OX siMature (manufactured by B-Bridge), and miCENTURY OX miNatural (manufactured by B-Bridge).
  • the method for synthesizing the nucleic acid used in the present invention is not particularly limited, and it can be produced by a method using a known chemical synthesis, an enzymatic transcription method or the like.
  • methods using known chemical synthesis include phosphoramidite method, phosphorothioate method, phosphotriester method, etc.
  • synthesis with ABI3900 high-throughput nucleic acid synthesizer can do.
  • the enzymatic transcription method include transcription using a typical phage RNA polymerase, for example, T7, T3, or SP6 RNA polymerase, using a plasmid or DNA having the target base sequence as a template.
  • Examples of a method for screening for a substance that promotes or suppresses the expression or function of the nucleic acid using the nucleic acid used in the present invention include, for example, introducing a vector that expresses the nucleic acid into a cell, and expressing an mRNA having the target base sequence. Alternatively, a method of screening for a substance that promotes or suppresses the function can be mentioned.
  • the pharmaceutical comprising the nucleic acid used in the present invention as an active ingredient can be used for diagnosis or treatment of diseases caused by fibrosis of cells or organs, tissue hyperplasia, or the like.
  • the nucleic acid can also be used as a fibrosis control agent for cells or organs.
  • Cell or organ fibrosis as used herein refers to a state in which extracellular matrix production is abnormally increased in the living body or a state in which epithelial-mesenchymal transition (EMT) occurs.
  • EMT epithelial-mesenchymal transition
  • An agent refers to an inhibitor or an accelerator.
  • “fibrosis of cells or organs” is not limited to fibrosis of only one of cells or organs, but includes cases where both cells and organs are fibrotic.
  • diseases caused by cell or organ fibrosis or tissue hyperplasia include, for example, fibrosis associated with damage to fibrous tissue.
  • the fibrous tissue is not particularly limited as long as it has a connective tissue, for example, kidney, lung, liver, heart, pancreas, spleen, stomach, spinal cord, pituitary gland, gonad, thyroid, gallbladder, Bone marrow, adrenal gland, skin, digestive tract (eg, large intestine, small intestine), brain, brain parts (eg, olfactory bulb, amygdala, basal sphere, hippocampus, thalamus, hypothalamus, cerebral cortex, medulla, cerebellum), bladder , Testis, ovary, placenta, uterus, bone, joint, skin, blood vessel, adipose tissue, skeletal muscle and the like.
  • a connective tissue for example, kidney, lung, liver, heart, pancreas, spleen, stomach, spinal cord
  • the disease include, but are not limited to, acute renal failure, glomerulonephritis, vasculitis, diabetic nephropathy, hypertensive nephrosclerosis, HIV nephropathy, IgA nephropathy, lupus nephritis, interstitial Nephritis, fibrosis associated with renal disorders such as obstructed kidney due to ureteral obstruction, pulmonary fibrosis, cirrhosis, arteriosclerosis, scleroderma, coronary restenosis after percutaneous transluminal coronary vasodilation (PTCA), between Examples include interstitial myocarditis, interstitial cystitis, skin scarring after burn injury, and fibrosis associated with poisoning.
  • PTCA percutaneous transluminal coronary vasodilation
  • RNA RNA
  • Identification of microRNA Whether a small RNA sequence is microRNA is determined by RNA (RNA) ), 9 , 277-279 (2003).
  • RNA RNA
  • RNA predicted to be transcribed from the genomic sequence Predict secondary structure.
  • Genomic sequences are publicly available and are available, for example, from UCSC Genome Bioinformatics (http://genome.ucsc.edu/).
  • RNAfold Nucleic Acids Research, 31 , 3429-3431 (2003)
  • Mfold Nucleic Acids Research, 31 , 3406-3415 (2003)
  • miRBase a database that maps miRBase to existing microRNAs.
  • low molecular weight RNA can be obtained by 15% polyacrylamide gel electrophoresis in accordance with the method described in Jeans & Development (Genes & Development), 15 , 188-200 (2000). The method of separating is mentioned. From this, 5'-terminal dephosphorylation, 3'-adapter ligation, phosphorylation, 5'-adapter ligation, reverse transcription, PCR amplification, concatamerization, and ligation to vector are sequentially cloned, and the clone is cloned. Can be determined.
  • RNA can be cloned and the base sequence of the clone can be determined.
  • RNA Cloning Kit manufactured by Takara Bio Inc.
  • 1-3 Method for detecting expression level of nucleic acid such as microRNA
  • methods for detecting the expression level of nucleic acid such as microRNA and its precursor include (1) Northern hybridization and (2) dot blot high. Hybridization, (3) in situ hybridization, (4) quantitative PCR, (5) differential hybridization, (6) microarray, (7) ribonuclease protection assay.
  • Northern blotting is a method in which sample-derived RNA is separated by gel electrophoresis, then transferred to a support such as a nylon filter, a probe labeled appropriately based on the base sequence of the nucleic acid is prepared, and hybridization and washing are performed.
  • this is a method for detecting a band specifically bound to a nucleic acid. Specifically, for example, it can be performed according to the method described in Science, 294 , 853-858 (2001).
  • the labeled probe is a base sequence of a nucleic acid used in the present invention, for example, a radioisotope, biotin, digoxigenin, a fluorescent group, a chemiluminescent group, etc. by a method such as nick translation, random priming or phosphorylation at the 5 ′ end. It can be prepared by incorporating it into DNA or RNA having a complementary sequence to LNA or LNA. Since the binding amount of the labeled probe reflects the expression level of the nucleic acid, the expression level of the nucleic acid can be quantified by quantifying the amount of bound labeled probe. Electrophoresis, transfer to membrane, probe preparation, hybridization, and nucleic acid detection can be performed by the method described in Molecular Cloning 3rd Edition [Cold Spring Harbor Press, (2001) Cold Spring Harbor, NY] it can.
  • RNA extracted from tissues and cells is spot-fixed on a membrane in a dotted manner, and then hybridized with a labeled polynucleotide that serves as a probe to detect RNA that specifically hybridizes with the probe. It is a method to do.
  • the probe the same probe as in Northern hybridization can be used. Preparation of RNA, RNA spot, hybridization, and detection of RNA can be performed by the methods described in Molecular Cloning 3rd edition.
  • In situ hybridization uses a paraffin or cryostat section of tissue obtained from a living body or immobilized cells as a specimen, performs hybridization and washing steps with a labeled probe, and observes the tissue and nucleic acid of a nucleic acid by microscopic observation. It is a method for examining distribution and localization in cells [Methods in Enzymology, 254 , 419 (1995)].
  • the probe the same probe as in Northern hybridization can be used. Specifically, microRNA can be detected according to the method described in Nature Method, 3 , 27 (2006).
  • cDNA synthesized from a sample-derived RNA using a reverse transcription primer and a reverse transcriptase (hereinafter, the cDNA is referred to as a sample-derived cDNA) is used for measurement.
  • a reverse transcription primer used for cDNA synthesis a random primer or a specific RT primer can be used.
  • the specific RT primer refers to a primer having a sequence complementary to a nucleic acid and a base sequence corresponding to the surrounding genomic sequence.
  • a template-specific design designed from a nucleotide sequence corresponding to a nucleic acid such as microRNA or a microRNA precursor and its surrounding genomic sequence, or a nucleotide sequence corresponding to a reverse transcription primer PCR is performed using a typical primer, a cDNA fragment containing the nucleic acid is amplified, and the amount of the nucleic acid contained in the sample-derived RNA is detected from the number of cycles until a certain amount is reached.
  • an appropriate region corresponding to the nucleic acid and the surrounding genomic sequence is selected, and consists of a sequence of 15 to 40 residues, preferably 20 to 30 residues at the 5 ′ end of the base sequence of the region.
  • a set of DNA or LNA consisting of DNA or LNA and a sequence complementary to 15 to 40 residues, preferably 20 to 30 residues at the 3 ′ end can be used. Specifically, it can be performed according to the method described in Nucleic Acids Research, 32 , e43 (2004).
  • a specific RT primer having a stem-loop structure can also be used as a reverse transcription primer for cDNA synthesis. Specifically, the method described in Nucleic Acid Research, 33, e179 ( 2005) or can be performed using the TaqMan MicroRNA Assays (Applied Biosystems).
  • a reverse transcription reaction can also be performed by adding a polyA sequence to a sample-derived RNA with polyA polymerase and using a base sequence containing an oligo dT sequence as a primer for reverse transcription. .
  • it can be performed using miScript System (Qiagen), QuantiMir RT Kit (System Biosciences), PrimeScript 1st strand cDNA Synthesis (Takara Bio).
  • miScript System Qiagen
  • QuantiMir RT Kit System Biosciences
  • PrimeScript 1st strand cDNA Synthesis Takara Bio
  • Examples of the method based on such hybridization include a method using differential hybridization [Trends Genet., 7 , 314 (1991)] and a microarray [Genome Res., 6 , 639 (1996)].
  • the difference in the amount of nucleic acid between the control sample and the target sample can be accurately detected by immobilizing an internal control such as a base sequence corresponding to U6 RNA on a filter or substrate.
  • labeled cDNA synthesis using differently labeled dNTPs mixturetures of dATP, dGTP, dCTP, and dTTP
  • RNA derived from the control sample and the target sample and two filters on one filter or one substrate.
  • nucleic acid quantification can be performed by simultaneously hybridizing labeled cDNA. Furthermore, the nucleic acid can be quantified by directly labeling and hybridizing RNA derived from the control sample and / or the target sample. For example, using a microarray described in Proc. Natl. Acad. Sci. USA, 101 , 9740-9744 (2004), Nucleic Acid Research, 32 , e188 (2004), RNA, 13 , 151-159 (2007), etc. MicroRNA can be detected. Specifically, it can be detected or quantified in the same manner as mirVana miRNA Bioarray (Ambion) and miRNA microarray kit (Agilent Technology).
  • a promoter sequence such as T7 promoter and SP6 promoter is bound to the 3 ′ end of the nucleotide sequence corresponding to the nucleic acid or its surrounding genomic sequence, and labeled NTP (mixture of ATP, GTP, CTP, UTP) and Labeled antisense RNA is synthesized by an in vitro transcription system using RNA polymerase.
  • the labeled antisense RNA is bound to the sample-derived RNA to form an RNA-RNA hybrid, and then digested with ribonuclease A that degrades only single-stranded RNA.
  • RNA fragments protected from digestion by forming RNA-RNA hybrids are detected or quantified as nucleic acids. Specifically, it can be detected or quantified using mirVana miRNA Detection Kit (Ambion).
  • RNAs or microRNA precursors that are expressed in cells or fibrotic tissues with increased fibrosis, or whose expression is increased or decreased compared to normal tissues, etc.
  • DNA that is a polymer of deoxyribonucleotides
  • the base sequence of DNA can be determined based on the base sequence of microRNA identified in 1 above.
  • the base sequence of DNA corresponding to the base sequence of RNA can be uniquely determined by replacing U (uracil) contained in the RNA sequence with T (thymine).
  • a polymer in which ribonucleotides and deoxyribonucleotides are mixed, a polymer containing nucleotide analogues, and a nucleic acid derivative can be synthesized in the same manner.
  • the method for synthesizing the nucleic acid is not particularly limited, and the nucleic acid can be produced by a method using known chemical synthesis or an enzymatic transcription method.
  • methods using known chemical synthesis include phosphoramidite method, phosphorothioate method, phosphotriester method, etc.
  • synthesis with ABI3900 high-throughput nucleic acid synthesizer can do.
  • the enzymatic transcription method include a transcription method using a typical phage RNA polymerase such as T7, T3, or SP6 RNA polymerase using a plasmid or DNA having the target base sequence as a template.
  • Method for detecting the function of a nucleic acid such as microRNA or microRNA precursor As a method for detecting the function of a nucleic acid such as microRNA, there is a method for detecting whether or not the translation of mRNA having a target base sequence is suppressed. be able to.
  • microRNA suppresses translation of mRNA containing the target base sequence in the 3 ′ terminal untranslated region (3′UTR) [Current Biology, 15 , R458-R460 (2005)]. Therefore, a DNA in which the target base sequence for the single-stranded RNA to be measured is inserted into the 3′UTR of an appropriate reporter gene expression vector is prepared, introduced into a host cell suitable for the expression vector, and single-stranded into the cell. By measuring the expression of a reporter gene when RNA is expressed, it can be detected whether or not it has a function of microRNA.
  • the reporter gene expression vector may be any vector as long as it has a promoter upstream of the reporter gene and can express the reporter gene in the host cell.
  • Any reporter gene can be used as the reporter gene, for example, firefly luciferase gene, Renilla luciferase gene, chloramphenicol acetyltransferase gene, ⁇ -glucuronidase gene, ⁇ -galactosidase gene, ⁇ -Lactamase gene, aequorin gene, green fluorescent protein gene and DsRed fluorescent gene can be used.
  • Reporter gene expression vectors having such properties include, for example, psiCHECK-1 (Promega), psiCHECK-2 (Promega), pGL3-Control (Promega), pGL4 (Promega), pRNAi-GL ( (Takara Bio), pCMV-DsRed-Express (CLONTECH), pMIR-REPORT® System (Ambion). Single-stranded RNA can be expressed by the method described in 6 below.
  • the function of single-stranded RNA as a microRNA can be specifically detected as follows. First, host cells are cultured in a multi-well plate or the like to express a reporter gene expression vector having a target sequence and single-stranded RNA. Then, the reporter activity is measured, and the function of the single-stranded RNA as a microRNA is detected by measuring the reporter activity when the single-stranded RNA is expressed compared to the case where the single-stranded RNA is not expressed. can do.
  • Method for detecting mutations in nucleic acids such as microRNA and microRNA precursors As a method for detecting mutations in nucleic acids such as microRNAs and microRNA precursors, heterogeneous nucleic acids formed by hybridization of normal and mutant nucleic acids A method for detecting this strand can be used.
  • Methods for detecting heteroduplex include (1) heteroduplex detection by polyacrylamide gel electrophoresis [Trends genet., 7 , 5 (1991)], (2) single strand conformation polymorphism analysis [Genomics, 16 , 325-332 (1993)], (3) Chemical cleavage of mismatches (CCM) [Human Genetics (1996), Tom Strachan and Andrew P. Read, BIOS Scientific Publishers Limited ], (4) Enzymatic cleavage method of mismatch [Nature Genetics, 9 , 103-104 (1996)], (5) Denaturing gel electrophoresis [Mutat. Res., 288 , 103-112 (1993)], etc. There are methods.
  • the heteroduplex detection method by polyacrylamide gel electrophoresis is performed as follows, for example. First, using a specimen-derived DNA or a specimen-derived cDNA as a template, a primer designed based on the genomic base sequence including the nucleic acid base sequence is amplified as a fragment smaller than 200 ⁇ bp. When heteroduplexes are formed, the mobility is slower than homoduplexes without mutations, and they can be detected as extra bands. If the fragment is smaller than 200 bp, most insertions, deletions and substitutions of 1 base or more can be detected. Heteroduplex analysis is preferably performed on a single gel combined with single-strand conformation analysis described below.
  • SSCP analysis single-strand conformation polymorphism analysis
  • primers derived from the base sequence of the genome including the base sequence of the nucleic acid
  • sample-derived DNA or the sample-derived cDNA as a template
  • the DNA amplified as a fragment smaller than 200 bp is denatured and electrophoresed in a native polyacrylamide gel.
  • the amplified DNA can be detected as a band by labeling the primer with an isotope or a fluorescent dye, or silver-staining the unlabeled amplification product.
  • a fragment having a mutation can be detected from the difference in mobility.
  • CCM method DNA fragments amplified with primers designed based on the base sequence of the genome including the base sequence of the nucleic acid using the sample-derived DNA or the sample-derived cDNA as a template are used as isotopes or fluorescence in the nucleic acid.
  • a labeled nucleic acid incorporating a label By hybridizing with a labeled nucleic acid incorporating a label and treating with osmium tetroxide, one strand of DNA at a position where a mismatch exists can be cleaved to detect a mutation.
  • CCM is one of the most sensitive detection methods and can be applied to specimens of kilobase length.
  • the mismatch can be cleaved enzymatically by combining RNase A with an enzyme involved in mismatch repair in cells such as T4 phage resol base and endonuclease VII.
  • DGGE method DNA fragments amplified with primers designed based on genomic base sequences, including nucleic acid base sequences, are used chemically, using specimen-derived DNA or specimen-derived cDNA as a template. Electrophoresis is performed using a gel having a denaturant concentration gradient and a temperature gradient. The amplified DNA fragment moves in the gel to a position where it is denatured into a single strand and does not move after denaturation. Since there is a difference in the movement of the amplified DNA in the gel with and without the mutation, the presence of the mutation can be detected.
  • a poly (G: C) terminal is preferably attached to each primer.
  • nucleic acid mutations can be detected by directly determining and analyzing the base sequence of the sample-derived DNA or the sample-derived cDNA.
  • nucleic acids such as microRNA and microRNA precursors
  • RNA sequences and their precursor sequences are registered in a database called miRBase at the Sanger Center in the UK.
  • Nucleic acids such as RNA precursors can be made. It can also be prepared using the microRNA sequence obtained by the method described in 1.
  • the nucleic acid can be expressed by using a vector that is biosynthesized by introduction into a cell and transcription. Specifically, based on the base sequence of the nucleic acid or the genomic base sequence containing the base sequence, a DNA fragment containing the hairpin portion is prepared and inserted downstream of the promoter of the expression vector to construct an expression plasmid. Next, the nucleic acid can be expressed by introducing the expression plasmid into a host cell suitable for the expression vector.
  • a vector that can replicate autonomously in a host cell or can be integrated into a chromosome and contains a promoter at a position where a gene containing a nucleic acid base sequence can be transcribed is used.
  • Any promoter can be used as long as it can be expressed in the host cell.
  • a promoter etc. can be mention
  • the Pol II promoter include a promoter of cytomegalovirus (human CMV) IE (immediate early) gene, an early promoter of SV40, and the like.
  • Examples of expression vectors using them include pCDNA6.2-GW / miR (Invitrogen), pSilencer® 4.1-CMV (Ambion), and the like.
  • Examples of pol III promoters include U6 RNA, H1 RNA, and tRNA gene promoters.
  • Examples of expression vectors using them include pSINsi-hH1 DNA (Takara Bio), pSINsi-hU6 DNA (Takara Bio), and pENTR / U6 (Invitrogen).
  • a gene containing a nucleic acid base sequence is inserted downstream of a promoter in a viral vector to construct a recombinant viral vector, the vector is introduced into a packaging cell to produce a recombinant virus, and the nucleic acid base sequence is determined. Genes containing can also be expressed.
  • the packaging cell may be any cell as long as it can replenish the deficient protein of the recombinant viral vector deficient in any of the genes encoding the proteins required for virus packaging, for example, from human kidney HEK293 cells, mouse fibroblast NIH3T3-derived cells, and the like can be used.
  • Proteins supplemented by packaging cells include mouse retrovirus-derived gag, pol, env, etc. for retrovirus vectors, and HIV virus-derived gag, pol, env, vpr, vpu for lentiviral vectors.
  • Vif, tat, rev, nef, etc. in the case of adenovirus vectors, proteins such as E1A and E1B derived from adenovirus, and in the case of adeno-associated virus vectors, Rep (p5, p19, p40), Vp (Cap), etc. Can be used.
  • nucleic acid used in the present invention can be directly introduced into a cell without using a vector.
  • a nucleic acid used in this method in addition to DNA, RNA, or nucleotide analogues, these chimeric molecules or derivatives of the nucleic acids can be used.
  • nucleic acids such as microRNAs and microRNA precursors can be expressed in the same manner as Pre-miRTM miRNA Precursor Molecules (Ambion) or miRIDIAN microRNA Mimics (GE Healthcare). When microRNA is expressed, any method can be used as long as microRNA can finally be produced in the cell.
  • RNA in addition to introducing single-stranded RNA as a microRNA precursor, (2) There is a method of introducing microRNA itself and RNA consisting of a complementary strand of microRNA and 100% -matched double-stranded RNA, and (3) double-stranded RNA assuming a state after microRNA is cleaved into Dicer. can give. Products that use these methods include miCENTURY OX Precursor (B-Bridge), miCENTURY OX siMature (B-Bridge), and miCENTURY OX miNatural (B-Bridge).
  • B-Bridge miCENTURY OX Precursor
  • B-Bridge miCENTURY OX siMature
  • B-Bridge miCENTURY OX miNatural
  • nucleic acids such as microRNAs and microRNA precursors are antisense technologies [Bioscience and Industry, 50 , 322 (1992), Chemistry, 46 , 681 ( 1991), Biotechnology, 9 , 358 (1992), Trends in Biotechnology, 10 , 87 (1992), Trends in Biotechnology, 10 , 152 (1992), Cell Engineering, 16 , 1463 (1997)], Triple Helix Technology [ Trends in Biotechnology, 10 , 132 (1992)], ribozyme technology [Current Opinion in Chemical Biology, 3 , 274 (1999), FEMS Microbiology Reviews, 23 , 257 (1999), Frontiers in Bioscience, 4 , D497 (1999), Chemistry & Biology, 6 , R33 (1999), Nucleic Acids Research, 26 , 5237 (1998), Trends In Biotechnology, 16 , 438 (1998)], Decoy DNA method [Nippon Rinsho-Japanese Journal of Clinical Medicine, 56
  • Antisense refers to a nucleic acid that can specifically hybridize a nucleic acid having a base sequence complementary to the base sequence of a certain target nucleic acid to suppress the expression of the target nucleic acid.
  • the nucleic acid used for antisense in addition to DNA, RNA or nucleotide analogs, these chimeric molecules or derivatives of the nucleic acids can also be used.
  • antisense can be produced and expression can be suppressed by following the method described in Nature, 432 , 226 (2004) and the like.
  • the siRNA is a short double-stranded RNA containing a base sequence of a certain target nucleic acid and can suppress the expression of the target nucleic acid by RNA interference (RNAi).
  • RNAi RNA interference
  • sequence of the siRNA the literature of the nucleotide sequence [Genes Dev., 13, 3191 (1999)] targeting can be appropriately designed based on the conditions.
  • siRNA can be prepared by synthesizing and annealing two RNAs having a sequence of 19 bases selected and a sequence obtained by adding TT to the 3 ′ end of each complementary sequence and annealing.
  • siRNA expression vector such as pSilencer 1.0-U6 (Ambion) or pSUPER (OligoEngine)
  • a vector expressing siRNA can be prepared.
  • the siRNA that suppresses a nucleic acid such as microRNA used in the present invention may be any siRNA that can suppress the activity of the nucleic acid, but each of the nucleotide sequences represented by any of SEQ ID NOs: 1 to 594 SiRNA designed from continuous sequence information after the 9th base is preferred.
  • the number of residues of the base constituting one strand of siRNA is preferably 17 to 30 residues, more preferably 18 to 25 residues, still more preferably 19 to 23 residues.
  • MicroRNA expressed in fibrotic cells using antisense or siRNA specific to nucleic acids such as microRNA and microRNA precursors expressed in fibrotic cells or tissues (hereinafter also referred to as “fibrotic cells”) And the expression of microRNA precursors can be suppressed.
  • fibrotic cells fibrotic cells
  • antisense or siRNA specific to nucleic acids such as microRNA and microRNA precursors expressed in fibrotic cells or tissues
  • the expression of microRNA precursors can be suppressed.
  • the activity of the microRNA is suppressed and the action of the microRNA or microRNA precursor in fibrotic cells is controlled. can do.
  • fibrotic cells can be obtained by administering to the patient an antisense oligonucleotide or siRNA specific to the microRNA or its precursor.
  • the disease which develops by the said expression abnormality can be treated. That is, an antisense oligonucleotide or siRNA specific to the microRNA or a precursor thereof is useful as a therapeutic agent for diseases caused by abnormal production of extracellular matrix, that is, fibrosis.
  • the antisense oligonucleotide or siRNA specific to a nucleic acid such as microRNA or a precursor thereof
  • the antisense oligonucleotide or siRNA alone or the nucleic acid encoding them is a retroviral vector
  • an appropriate vector such as an adenovirus vector or an adeno-associated virus vector
  • nucleic acid such as microRNA is used for expression of mRNA having target base sequence. Any method may be used as long as it uses the inhibitory activity. For example, by expressing a nucleic acid such as microRNA and increasing the amount of nucleic acid such as microRNA in the cell, it is possible to suppress the translation of mRNA having the target sequence and to suppress the expression of the gene. .
  • the nucleic acid can be expressed by the method described in 5 above. Examples of the mRNA having the target base sequence of the nucleic acid consisting of the base sequence represented by any of SEQ ID NOs: 1 to 594 can include the gene groups shown in Table 4 above, for example.
  • the function of the target gene can be suppressed using siRNA for the target gene shown in Table 4.
  • Method for screening a substance that promotes or suppresses expression or function of nucleic acid using nucleic acid such as microRNA or microRNA precursor Using nucleic acid to promote expression or function of nucleic acid such as microRNA or its precursor Substances to be suppressed can be screened. For example, by selecting a base sequence to be screened from the base sequence of a nucleic acid and utilizing a cell that expresses a nucleic acid having the base sequence, the expression or function of the selected microRNA or its precursor is promoted or Substances to be suppressed can be screened.
  • a vector expressing a nucleic acid having the base sequence may be an animal cell, A transformed cell obtained by introduction into a host cell such as yeast, a cell into which a nucleic acid having the base sequence is directly introduced without using a vector, and the like can also be used.
  • Specific screening methods include methods that use changes in the expression level of nucleic acids such as microRNAs or their precursors to be screened as indicators, as well as mRNAs that have nucleic acid target sequences such as microRNAs, and encoded by them.
  • a method using the change in the expression level of the gene product as an index can be mentioned.
  • (A) Screening method using as an index a change in the expression level of a nucleic acid such as a microRNA or a precursor thereof as a target for screening The test substance is contacted with a cell expressing the nucleic acid, and the expression level of the selected nucleic acid is determined.
  • a substance that promotes or suppresses nucleic acids such as expression of microRNA and its precursor is obtained.
  • the expression level of the nucleic acid can be detected by the method described in 3 above.
  • (B) Screening method using as an index the change in the expression level of mRNA having a nucleic acid target sequence such as microRNA to be screened and the gene product encoded by it Contact test cells with cells expressing the mRNA
  • a substance that promotes or suppresses the expression or function of nucleic acids such as microRNA and its precursor is obtained using changes in the expression level of mRNA having the target sequence of the selected nucleic acid and the gene product encoded thereby as an index.
  • a DNA in which the target sequence of a nucleic acid such as microRNA is inserted into the 3′UTR of an appropriate reporter gene expression vector is prepared, introduced into a host cell suitable for the expression vector, and a test substance is contacted with the cell, Using a change in the expression level of the reporter gene as an indicator, a substance that promotes or suppresses the expression or function of nucleic acids such as microRNA and its precursor is obtained.
  • mRNA having a nucleic acid target sequence such as a microRNA comprising a base sequence represented by any of SEQ ID NOs: 1 to 594 include: Examples of the gene groups shown in Table 4 above can be given.
  • Cell fibrosis inhibitor or fibrosis promoter using nucleic acid such as microRNA or microRNA precursor Nucleic acid such as microRNA or microRNA precursor, and nucleic acid having a base sequence complementary to the base sequence are: By controlling the expression of a gene having a target sequence, it can be used as a cell fibrosis inhibitor or fibrosis promoter.
  • Examples of the organ or cell fibrosis inhibitor include the following nucleic acids mentioned in (a) to (h).
  • a nucleic acid having a nucleotide sequence of 17 to 28 nucleotides (c) having 90% or more identity with the nucleotide sequence represented by any of SEQ ID NOs: 1 to 23, 28 to 411, and 487 to 589
  • SEQ ID Nos. 595 to 618, 624 to 1073, and 1156 to 1277 Nucleic acid comprising a base sequence represented by any of the above (g) A nucleic acid comprising a base sequence represented by any one of SEQ ID NOs: 595 to 618, 624 to 1073, and 1156 to 1277 and having a nucleotide sequence of 90% or more (H) a nucleic acid that hybridizes under stringent conditions with a complementary strand of a nucleic acid comprising the base sequence represented by any of SEQ ID NOs: 619 to 623, 1074 to 1155, and 1278 to 1281.
  • organ or cell fibrosis promoters include nucleic acids listed in (i) to (p) below.
  • Nucleic acid consisting of a base sequence (l) Nucleic acid (m) sequence that hybridizes under stringent conditions with a complementary strand of a nucleic acid consisting of a base sequence represented by any one of SEQ ID NOs: 1 to 23, 28 to 411, and 487 to 589 Nucleic acids containing the second to eighth base sequences of the base sequences represented by any of Nos.
  • SEQ ID Nos. 619 to 623, 1074 to 1155, 12 Nucleic acid consisting of a base sequence represented by any one of 8 to 1281 (o) A base having 90% or more identity with the base sequence represented by any one of SEQ ID NOs: 619 to 623, 1074 to 1155, 1278 to 1281 Nucleic acid consisting of sequence (p) A nucleic acid that hybridizes under stringent conditions with a complementary strand of nucleic acid consisting of a base sequence represented by any of SEQ ID NOs: 595 to 618, 624 to 1073, and 1156 to 1277.
  • the nucleic acid microRNA or a microRNA precursor is preferably used as the nucleic acid.
  • Examples of the fibrosis promoter for cells or organs include nucleic acids having a base sequence complementary to the base sequences of the nucleic acids described in (a) to (h) above. Examples thereof include nucleic acids having a base sequence complementary to the base sequences of the nucleic acids described in (i) to (p) above.
  • the above-mentioned nucleic acids and vectors expressing nucleic acids complementary thereto can also be used as cell growth inhibitors or growth promoters.
  • a substance that suppresses the expression of a target gene of a nucleic acid such as the above microRNA, or a substance that promotes the expression of the target gene can also be used as a fibrosis inhibitor or a proliferation promoter of cells or organs.
  • a nucleic acid or a vector expressing it can also be used.
  • substances that suppress the expression of the target gene include siRNA against the mRNA of the target gene and antisense to the target gene.
  • Substances that promote the expression of the target gene include those against the target gene-specific microRNA. Examples include siRNA and antisense to the target gene.
  • the preparation form and administration method of the cell or organ fibrosis inhibitor or growth promoter are the same as those for diagnostic agents and therapeutic agents containing nucleic acids such as microRNA and microRNA precursor described later in 10.
  • nucleic acids such as microRNA and microRNA precursors control the expression of genes that have target sequences or control the expression of nucleic acids such as microRNAs By controlling, it can be used as a therapeutic agent for diseases caused by fibrosis of cells or organs, that is, abnormal production of extracellular matrix and EMT.
  • siRNA against the target gene of the nucleic acid controls fibrosis of cells or organs, that is, as a disease caused by abnormal production of extracellular matrix and EMT, etc. Examples include fibrosis associated with tissue damage.
  • a diagnostic agent containing a nucleic acid is a reagent necessary for quantifying a nucleic acid or detecting a mutation, such as a buffer, a salt, a reaction enzyme, a labeled protein that binds to the nucleic acid, depending on the target diagnostic method, And a coloring agent for detection.
  • a therapeutic agent containing a nucleic acid as an active ingredient can be administered alone, but usually mixed with one or more pharmacologically acceptable carriers and well known in the pharmaceutical arts. It is desirable to administer it as a pharmaceutical preparation produced by any method. It is desirable to use the most effective route for treatment, and oral administration or parenteral administration such as buccal, respiratory tract, rectal, subcutaneous, intramuscular and intravenous is desirable. Can be given intravenously.
  • Examples of the dosage form include sprays, capsules, tablets, granules, syrups, emulsions, suppositories, injections, ointments, tapes and the like.
  • Suitable formulations for oral administration include emulsions, syrups, capsules, tablets, powders, granules and the like.
  • Liquid preparations such as emulsions and syrups include sugars such as water, sucrose, sorbitol and fructose, glycols such as polyethylene glycol and propylene glycol, oils such as sesame oil, olive oil and soybean oil, p-hydroxybenzoic acid
  • Preservatives such as esters, and flavors such as strawberry flavor and peppermint can be used as additives.
  • excipients such as lactose, glucose, sucrose, mannitol, disintegrants such as starch and sodium alginate, lubricants such as magnesium stearate and talc, polyvinyl alcohol, hydroxy A binder such as propylcellulose and gelatin, a surfactant such as fatty acid ester, and a plasticizer such as glycerin can be used as additives.
  • Formulations suitable for parenteral administration include injections, suppositories, sprays and the like.
  • the injection is prepared using a carrier made of a salt solution, a glucose solution, or a mixture of both.
  • Suppositories are prepared using a carrier such as cacao butter, hydrogenated fat or carboxylic acid.
  • the spray is prepared using a carrier that does not irritate the recipient's oral cavity and airway mucosa, and that facilitates absorption by dispersing the active ingredient as fine particles.
  • the carrier include lactose and glycerin.
  • a formulation such as an aerosol or dry powder is possible.
  • the components exemplified as additives for oral preparations can also be added.
  • the dose or frequency of administration varies depending on the intended therapeutic effect, administration method, treatment period, age, weight, etc., but is usually 10 ⁇ g / kg to 20 mg / kg per day for an adult.
  • a therapeutic agent containing a nucleic acid as an active ingredient can also be produced by preparing a nucleic acid-expressing vector and a base used for the nucleic acid therapeutic agent [Nature Genet., 8, 42 (1994)].
  • the base used for the therapeutic agent may be any base as long as it is usually used for injections, such as distilled water, sodium chloride or a salt solution such as a mixture of sodium chloride and an inorganic salt, mannitol, lactose, dextran. And a solution such as glucose, an amino acid solution such as glycine and arginine, an organic acid solution or a mixed solution of a salt solution and a glucose solution, and the like.
  • these bases are mixed with an osmotic pressure adjusting agent, a pH adjusting agent, a vegetable oil such as sesame oil and soybean oil, or an auxiliary such as a surfactant such as lecithin or a nonionic surfactant.
  • An injection may be prepared as a suspension or dispersion. These injections can be prepared as preparations for dissolution at the time of use by operations such as pulverization and freeze-drying.
  • the therapeutic agent can be used for treatment as it is in the case of a liquid just before the treatment, or in the case of a solid, dissolved in the above sterilized base if necessary.
  • the nucleic acid-expressing vector examples include the recombinant virus vector prepared by the above method 5, and more specifically, a retrovirus vector and a lentivirus vector.
  • a virus vector can be prepared by combining a nucleic acid with a polylysine-conjugated antibody specific for an adenovirus hexon protein to produce a complex, and binding the resulting complex to an adenovirus vector.
  • the virus vector stably reaches a target cell, is taken up into the cell by an endosome, is degraded in the cell, and the nucleic acid can be efficiently expressed.
  • RNA vectors based on Sendai virus (-) strand RNA virus have also been developed (WO97 / 16538, WO97 / 16539), and using the Sendai virus, a Sendai virus incorporating a nucleic acid can be produced. Can do. Nucleic acids can also be transferred by non-viral nucleic acid transfer methods. For example, calcium phosphate coprecipitation method [Virology, 52 , 456-467 (1973); Science, 209 , 1414-1422 (1980)], microinjection method [Proc. Natl. Acad. Sci. USA, 77 , 5399-5403 ( Proc. Natl. Acad. Sci.
  • nucleic acid can be taken up and expressed locally in the tissue by administering the liposome preparation directly to the target tissue [Hum. Gene Ther., 3 , 399 (1992)].
  • Direct DNA uptake techniques are preferred for targeting DNA directly to a lesion.
  • receptor-mediated DNA transfer can be performed by binding DNA (usually in the form of a covalently closed supercoiled plasmid) to a protein ligand via polylysine. The ligand is selected based on the presence of the corresponding ligand receptor on the cell surface of the target cell or tissue.
  • the ligand-DNA conjugate can be injected directly into the blood vessel, if desired, and can be directed to a target tissue where receptor binding and internalization of the DNA-protein complex occurs.
  • adenovirus can be co-infected to disrupt endosomal function.
  • the method for measuring fibrosis is not particularly limited as long as it is a method capable of measuring increased production of extracellular matrix.
  • staining of extracellular matrix can be used.
  • a method for evaluating the extracellular matrix includes Masson's trichrome staining.
  • the nucleus can be dyed in black with iron hematoxylin, the cytoplasm in red with acidic fuchsin, and the collagen fibers in blue with aniline blue.
  • the blue region and the fibrosis region in the tissue are in a proportional relationship. It is recognized [J Clin Pathol. 59 (4) , 377-81 (2006)].
  • Method for measuring the degree of organ and cell fibrosis As a method for measuring the degree of organ and cell fibrosis, gene expression of the extracellular matrix can be analyzed by quantitative PCR. Specifically, the expression of Collagen and Fibronectin, which are extracellular substrates, is performed by PCR using appropriate reagents such as Syber-Green (QIAGEN) and Taqman-probe (Applied Biosystems), and the fluorescence intensity is measured. Methods and the like. There are also methods for quantifying extracellular matrix by Western blot, ELISA (Enzyme-Linked Immunosorbent Assay) method, colorimetric method and the like. More specifically, Sircol Soluble Collagen Assay (manufactured by Biocolor) and the like can be mentioned.
  • ELISA Enzyme-Linked Immunosorbent Assay
  • fibrosis can be measured only in EMT, which is said to be a particularly important phenomenon in the process of organ and cell fibrosis.
  • Methods for evaluating EMT include reduction of cell markers that are specifically expressed in epithelial cells such as ⁇ -catenin and ZO-1, E-cadherin, and mesenchymal cells such as FSP-1, ⁇ SMA, and Vimentin.
  • quantitative analysis is also possible by analyzing their gene expression by quantitative PCR [J Clin Invest., 117 , 482-91 (2007)].
  • microRNA expression of microRNA during renal fibrosis
  • UUO unilateral ureteral obstruction
  • the left renal ureter of 7-week-old BALB / c mice purchased from Charles River Japan
  • Trizol reagent Invitrogen
  • the product was added to the product.
  • Total RNA was extracted according to the method described.
  • mirVana registered trademark
  • miRNA Bioarrays V9.2 manufactured by Ambion
  • microRNA in the control mouse was set to 1.00, and compared with the expression level of microRNA in the ureter-ligated mouse, the expression was observed in both ureter-ligated mice.
  • Table 5 shows the average value of two urinary ligation mice relative to the control. As shown in Table 5, 31 candidate microRNAs were obtained.
  • mmu_miR_345 is SEQ ID NO: 28
  • hsa_miR_320 is SEQ ID NO: 2
  • hsa_miR_422b corresponds to hsa_miR_378, SEQ ID NO: 3
  • rno_miR_422b corresponds to rno_miR_378, SEQ ID NO: 45
  • hsa_miR_213 is SEQ ID NO: 4
  • mmu_miR_99h is SEQ ID NO: 4
  • hsa_miR_422a is SEQ ID NO: 7
  • hsa_miR_30e_5p is SEQ ID NO: 9
  • hsa_miR_30d is SEQ ID NO: 10
  • hsa_miR_30b is SEQ ID NO: 11
  • hsa_miR_30c is SEQ ID NO: 12
  • the microRNAs indicated by ambi_miR_3998, ambi_miR_7029, ambi_miR_13268, and ambi_miR_13143 are hsa_miR_213 (SEQ ID NO: 4), hsa_miR_451 (SEQ ID NO: 18), hsa_miR_140-3p (SEQ ID NO: 21) registered in miRBase, respectively. This corresponds to hsa_miR_1974 (SEQ ID NO: 23).
  • NRK-49F A rat renal fibroblast cell line, NRK-49F cells (hereinafter referred to as NRK-49F), is a DMEM medium (GIBCO) containing 5% fetal calf serum (GIBCO) and 5% CO 2 at 37 ° C. Cultured in a concentration incubator.
  • NRK-49F was seeded in a 6-well plate at 3 ⁇ 10 4 per well and cultured overnight in the above medium.
  • the medium was replaced with DMEM medium without serum.
  • the medium was replaced with a recombinant human TGF- ⁇ 1 (manufactured by R & D) diluted to a final concentration of 5 ng / ml using DMEM medium without serum, and the culture was continued.
  • the medium was changed to serum-free DMEM not containing TGF- ⁇ 1.
  • the medium was removed, and total RNA was extracted using Trizol reagent (Invitrogen) according to the method described in the product.
  • RNA concentration After measuring the RNA concentration, react with TaqMan MicroRNA assays (Applied Biosystems) according to the method described in the product, and use Real-time PCR with Applied Biosystems 7900HT Fast real-time PCR system (Applied Biosystems). Went.
  • the relative expression level of each control cell was calculated by setting the microRNA expression level of control cells to which TGF ⁇ -1 was not added to 1.00.
  • Table 6 it is clear that miR-378, 192, 30e-5p, 30b, 30c, 30d, 486, 185, 320, 451, 200a, 29c expression is reduced by TGF- ⁇ stimulation. It became.
  • miR-378, 30b, 30c, 486, 451, 29c showed an expression decrease of 50% or more.
  • miR-378 is SEQ ID NO: 3
  • miR-192 is SEQ ID NO: 6
  • miR-30e-5p is SEQ ID NO: 9
  • miR-30d is SEQ ID NO: 10
  • miR-30c is SEQ ID NO: 12
  • miR-30b is SEQ ID NO: 11
  • miR-486 is SEQ ID NO: 14
  • miR-185 is SEQ ID NO: 16
  • miR-320 is SEQ ID NO: 2
  • miR-451 is SEQ ID NO: 18
  • miR-200a is SEQ ID NO: 19
  • miR-29c is SEQ ID NO: The microRNA which has a base sequence represented by 20 is represented.
  • probes represented by human sequences were used for TaqMan MicroRNA assays.
  • MDCK cells which are canine renal epithelial cells.
  • MDCK was seeded in a 24-well plate at 1 ⁇ 10 4 per well and cultured overnight in the above medium. One day later, the medium was replaced with recombinant human TGF- ⁇ 1 (R & D) diluted with DMEM medium containing 10% fetal bovine serum to a final concentration of 10 ng / ml, and the culture was continued. .
  • Trizol reagent Invitrogen
  • TaqMan MicroRNA assays (Applied Biosystems)
  • Applied Biosystems 7900HT Fast real-time PCR system (Applied Biosystems)
  • expression of control cells that do not stimulate TGF- ⁇ and microRNA The amount was compared.
  • the relative expression level of each control cell was calculated by setting the microRNA expression level of the control cells to which TGF- ⁇ 1 was not added to 1.00. As a result, as shown in Table 7, it was revealed that miR-30b, 30c, and 486 expression was reduced by TGF- ⁇ stimulation.
  • miR-146b, 21, 223 expression was increased by TGF- ⁇ stimulation.
  • miR-30b is SEQ ID NO: 11
  • miR-30c is SEQ ID NO: 12
  • miR-486 is SEQ ID NO: 14
  • miR-146b is SEQ ID NO: 25
  • miR-21 is SEQ ID NO: 26
  • miR-223 is SEQ ID NO: The microRNA which has a base sequence represented by 27 is represented.
  • probes represented by human sequences were used for TaqMan MicroRNA assays.
  • Renal fibrosis is known to involve tubule cells, and the cell line NRK-52E is used as a model. Collagen I and the like are used as an index of tubule cell fibrosis [JASN., 16 , 2702-13 (2005)], [Journal of Cellular Biochemistry, 103 , 1999-2009 (2008)].
  • NRK-52E cells (hereinafter referred to as NRK-52E), a rat renal tubular epithelial cell line, are 5% CO at 37 ° C in DMEM medium (GIBCO) containing 5% fetal calf serum (GIBCO). The cells were cultured in a 2 concentration incubator. NRK-52E was seeded in a 24-well plate at 1 ⁇ 10 4 per well and cultured overnight in the above medium. One day later, the final concentration of pre-miR miRNA precursor molecules (Ambion) producing miR-21, 223, 30b, 30c, 486, 194, let-7i was determined by lipofection using lipofectamine 2000 (Invitrogen). Was introduced into NRK-52E to be 5 nM.
  • DMEM medium fetal calf serum
  • MiRNA precursor molecules-Negative control (Ambion) was also introduced as a negative control. Lipofection followed the method described in the instructions attached to the product. 24 hours after the introduction of the molecule by lipofection method, the medium was diluted with DMEM medium containing 5% fetal bovine serum to a final concentration of 2 ng / ml of recombinant human TGF- ⁇ 1 (R & D). The culture was continued. Two days after changing to a medium containing TGF- ⁇ 1, the medium was removed, and using Trizol reagent (Invitrogen), total RNA was extracted according to the method described in the product.
  • Trizol reagent Invitrogen
  • cDNA was synthesized according to the method described in the product.
  • the expression of fibrosis-related genes was analyzed by real-time RT-PCR.
  • Samples were prepared using the QuantiTect SYBR Green PCR Kit (QIAGEN) according to the method described in the product, and the expression level of collagen I was quantified using an Applied Biosystems 7900HT Fast real-time PCR system (Applied Biosystems).
  • fibroblasts forced to express microRNAs on extracellular matrix production Pre-miR miRNA precursor molecules (Ambion) were introduced into fibroblasts, and the effects of microRNAs generated from these molecules on collagen production were examined. It was. Renal fibrosis is known to involve fibroblasts in kidney tissue, and the model cell line NRK-49F is used as the model. Collagen I or the like is used as an index of fibrosis [JASN., 16 , 2702-13 (2005)], [Kidney International, 66 , 112-120 (2004)]. NRK-49F was seeded in a 24-well plate at 2 ⁇ 10 4 per well and cultured overnight in the above medium.
  • pre-miR miRNA precursor molecules that produce miR-21, 30b, 30c, 192, 486, let-7i is 5 nM by lipofection using lipofectamine 2000 (Invitrogen). Introduced into NRK-49F. MiRNA precursor molecules-Negative control (Ambion) was also introduced as a negative control. Lipofection followed the method described in the instructions attached to the product. Eight hours after the introduction of the molecule by the lipofection method, the medium was replaced with serum-free DMEM medium.
  • the medium was replaced with recombinant human TGF- ⁇ 1 (R & D) diluted to a final concentration of 1 ng / ml using DMEM medium without serum, and the culture was continued.
  • TGF- ⁇ 1 recombinant human TGF- ⁇ 1
  • the medium was removed, and using Trizol reagent (Invitrogen), total RNA was extracted according to the method described in the product.
  • Trizol reagent Invitrogen
  • total RNA was extracted according to the method described in the product.
  • PrimeScript 1st strand cDNA Synthesis Using Kit manufactured by Takara Bio Inc.
  • cDNA was synthesized according to the method described in the product.
  • the expression of fibrosis-related genes was analyzed by real-time RT-PCR.
  • MDCK epithelial-mesenchymal transition
  • ⁇ SMA ⁇ smooth muscle actin
  • MDCK was seeded in a 24-well plate at 1 ⁇ 10 4 per well and cultured overnight in the above medium.
  • pre-miR miRNA precursor molecules that generate miR-21, 146b, 223, 194, let-7i were made MDCK to a final concentration of 5 nM by lipofection using lipofectamine 2000 (Invitrogen). Introduced.
  • MiRNA precursor molecules-Negative control (Ambion) was also introduced as a negative control. Lipofection followed the method described in the instructions attached to the product. 24 hours after introduction of the molecule by the lipofection method, the medium was diluted with DMEM medium containing 10% fetal calf serum to a final concentration of 2 ng / ml of recombinant human TGF- ⁇ 1 (R & D). The culture was continued. Two days after replacement with a medium containing TGF- ⁇ 1, the medium was removed, and using Trizol reagent (Invitrogen), total RNA was extracted according to the method described in the product.
  • Trizol reagent Invitrogen
  • Li-90 cells which are human hepatic stellate cells, are DMEM medium (GIBCO) containing 20% fetal calf serum (GIBCO) and a 5% CO 2 concentration at 37 ° C.
  • the medium was replaced with recombinant human TGF- ⁇ 1 (manufactured by R & D) diluted to a final concentration of 1 ng / ml using DMEM medium without serum, and the culture was continued.
  • TGF- ⁇ 1 manufactured by R & D
  • Trizol reagent Invitrogen
  • total RNA was extracted according to the method described in the product.
  • cDNA was synthesized using superscript VILO (Invitrogen) according to the method described in the product. Using the prepared cDNA, the expression of fibrosis-related genes was analyzed by real-time RT-PCR.
  • microRNA effects of microRNA on forced extracellular matrix production of pulmonary fibroblasts
  • Pre-miR miRNA precursor molecules manufactured by Ambion
  • fibroblasts in lung tissue are involved in lung fibrosis, and collagen I and the like are used as an index of the fibrosis [Respiration, 77 , 311-319 (2009) ].
  • IMR-90 Human lung fibroblast IMR-90 cells
  • IMR-90 Human lung fibroblast IMR-90 cells
  • MEM medium containing 10% fetal calf serum (GIBCO), 2 mM L-glutamine, 0.1 mM NEAA, 1 mM pyruvate ( (Manufactured by GIBCO) at 37 ° C. in an incubator with 5% CO 2 concentration.
  • IMR-90 was seeded in a 24-well plate at 1.5 ⁇ 10 4 per well and cultured overnight in the above medium.
  • pre-miR miRNA precursor molecules that generate miR-30b, 30c, 486, and let-7i were added to IMR-90 by lipofection using lipofectamine 2000 (Invitrogen) to a final concentration of 10 nM. Introduced. MiRNA precursor molecules-Negative control (Ambion) was also introduced as a negative control. Lipofection followed the method described in the instructions attached to the product. Eight hours after the introduction of the molecule by the lipofection method, the medium was replaced with a serum-free MEM medium.
  • the medium was replaced with a recombinant human TGF- ⁇ 1 (manufactured by R & D) diluted to a final concentration of 0.5 ng / ml using DMEM medium without serum, and the culture was further continued.
  • TGF- ⁇ 1 human TGF- ⁇ 1
  • Trizol reagent Invitrogen
  • total RNA was extracted according to the method described in the product.
  • cDNA was synthesized using superscript VILO (Invitrogen) according to the method described in the product. Using the prepared cDNA, the expression of fibrosis-related genes was analyzed by real-time RT-PCR.
  • anti-miR miRNA inhibitors that inhibit miR-30c, 486, 320a, 378, and 451 were introduced into NRK-52E by lipofection using lipofectamine 2000 (Invitrogen) to a final concentration of 30 nM. did.
  • Anti-miR miRNA Inhibitors-Negative Control # 1 (Ambion) was also introduced as a negative control. Lipofection followed the method described in the instructions attached to the product. 24 hours after introduction of the molecule by the lipofection method, the medium was diluted with DMEM medium containing 5% fetal calf serum to a final concentration of 2 ng / ml of recombinant human TGF- ⁇ 1 (R & D) The culture was continued.
  • miR-320a SEQ ID NO: 2
  • miR-378 SEQ ID NO: 3
  • miR-30c SEQ ID NO: 12
  • miR-486 SEQ ID NO: 14
  • miR-451 It was revealed that the expression of collagen I was increased by suppressing the expression of SEQ ID NO: 18).
  • anti-miR miRNA inhibitors that inhibit miR-30c, 192, 320a, 378, and 451 were introduced into NRK-49F by lipofection using lipofectamine 2000 (Invitrogen) to a final concentration of 30 nM. did.
  • Anti-miR miRNA Inhibitors-Negative Control # 1 was also introduced as a negative control. Lipofection followed the method described in the instructions attached to the product. For let-7i and miR-486, miRIDIAN microRNA Hairpin Inhibitor (manufactured by Dharmacon) was introduced into NRK-49F by a lipofection method using lipofectamine 2000 to a final concentration of 10 nM.
  • MiRIDIAN microRNA Hairpin Inhibitor Negative Control # 2 (Dharmacon) was also introduced as a negative control. Eight hours after the introduction of the molecule by the lipofection method, the medium was replaced with serum-free DMEM medium. After another 16 hours, the medium was replaced with a recombinant human TGF- ⁇ 1 (manufactured by R & D) diluted to a final concentration of 1 ng / ml using DMEM medium without serum, and the culture was further continued. Two days after replacement with a medium containing TGF- ⁇ 1, the medium was removed, and using Trizol reagent (Invitrogen), total RNA was extracted according to the method described in the product.
  • Trizol reagent Invitrogen
  • miR-320a SEQ ID NO: 2
  • miR-378 SEQ ID NO: 3
  • miR-192 SEQ ID NO: 6
  • miR-30c SEQ ID NO: 12
  • miR-486 It was revealed that the expression of collagen I was increased by suppressing the expression of SEQ ID NO: 14), miR-451 (SEQ ID NO: 18), and let-7i (SEQ ID NO: 8).
  • Anti-miR miRNA Inhibitors-Negative Control # 1 was also introduced as a negative control. Lipofection followed the method described in the instructions attached to the product. 24 hours after introduction of the molecule by the lipofection method, the medium was diluted with DMEM medium containing 10% fetal bovine serum to a final concentration of 2 ng / ml of recombinant human TGF- ⁇ 1 (R & D). The culture was continued. Two days after replacement with a medium containing TGF- ⁇ 1, the medium was removed, and using Trizol reagent (Invitrogen), total RNA was extracted according to the method described in the product.
  • Trizol reagent Invitrogen
  • miR-320a SEQ ID NO: 2
  • miR-378 SEQ ID NO: 3
  • miR-192 SEQ ID NO: 6
  • miR-30c SEQ ID NO: 12
  • miR-486 It was revealed that the expression of ⁇ SMA was increased by suppressing the expression of SEQ ID NO: 14) and miR-451 (SEQ ID NO: 18).
  • microRNA administration on organ fibrosis
  • the microRNAs evaluated were miR-30b, miR-486, and let-7i.
  • miR-30b uses a nucleic acid (hsa-miR-30b) consisting of the base sequence represented by SEQ ID NO: 11 as an antisense strand, and is complementary to the first base to the 19th base on the 5 ′ side of the antisense strand.
  • miR-486 is obtained by annealing an antisense strand consisting of the base sequence represented by SEQ ID NO: 14 and a sense strand consisting of the base sequence represented by SEQ ID NO: 1283
  • let-7i is SEQ ID NO: 8
  • An antisense strand consisting of the base sequence represented by the above and an annealed sense strand consisting of the base sequence represented by SEQ ID NO: 1284 were used.
  • 21mer luciferase siRNA was used as a negative control.
  • RNA was synthesized by Hokkaido System Science and described in Stable nucleic acid lipid particle (SNALP), a lipid particle with high delivery efficiency to the liver, described in Mol Ther., 13 (3) , 494-505 (2006). After conjugation according to the established method, it was used for evaluation. Hepatology, 50 , 185-197 (2009), Am J Physiol Gastrointest Liver Physiol, 296 , G582-G592 using the Bile Duct Ligation (BDL) model known as a general animal model for liver fibrosis (2009)].
  • BDL Bile Duct Ligation
  • Bile ducts from 9-week-old male C57BL / 6J mice purchased from Charles River Japan were ligated under anesthesia and 7 SNALP conjugate microRNAs were administered at a dose of 150 ⁇ g / head immediately after surgery and 24 hours later, respectively. Administered. Thereafter, the liver was removed 48 hours later and quickly frozen in liquid nitrogen. Frozen liver is crushed using Trizol reagent (Invitrogen), added with chloroform (Wako Pure Chemicals) and centrifuged, and then the supernatant is described in the product using RNeasy kit (QIAGEN) Total RNA was extracted according to the method described.
  • cDNA was synthesized using Super Script VILO TM cDNA Synthesis Kit (manufactured by Invitrogen) according to the method described in the product.
  • Super Script VILO TM cDNA Synthesis Kit manufactured by Invitrogen
  • the expression of fibrosis-related genes was analyzed by real-time RT-PCR.
  • the expression level of TGF- ⁇ 1 was quantified.
  • an agent for controlling fibrosis of cells or organs a diagnostic or therapeutic agent for diseases caused by fibrosis of cells or organs, an agent for controlling expression of target genes of nucleic acids such as microRNA, a method for controlling cells or organs,
  • screening methods for cell or organ fibrosis control agents and the like are provided, which are useful in the prevention, diagnosis or treatment of diseases caused by cell or organ fibrosis.
  • control refers to suppression and promotion.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Urology & Nephrology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biochemistry (AREA)
  • Wood Science & Technology (AREA)
  • Hematology (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Engineering & Computer Science (AREA)
  • Physics & Mathematics (AREA)
  • Zoology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Biophysics (AREA)
  • Food Science & Technology (AREA)
  • Plant Pathology (AREA)
  • Cell Biology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Provided are a nucleic acid and fibrosis control agent that are useful in controlling fibrosis of cells or organs. Also provided is a method for using said nucleic acid and fibrosis control agent. The following are provided: a nucleic acid containing a base sequence represented by one of sequence numbers 1 to 1281; a nucleic acid at least 90% identical to the aforementioned nucleic acid; a nucleic acid hybridized with a complementary strand of the aforementioned nucleic acid under stringent conditions; a nucleic acid containing the second through eighth base sequences of a base sequence represented by one of sequence numbers 1 to 594; a cell or organ fibrosis control agent using a nucleic acid or the like comprising a base sequence that is complementary to the base sequence of an abovementioned nucleic acid; a diagnostic or therapeutic agent for disorders resulting from cell or organ fibrosis; an expression control agent for target genes of nucleic acids such as microRNAs; a control method or the like for cells and organs; and a method for screening for cell or organ fibrosis control agents.

Description

細胞または臓器の線維化を制御する核酸Nucleic acids that control fibrosis of cells or organs
 本発明は、核酸を用いた細胞または臓器の線維化制御剤、線維性障害に起因する疾患の診断薬または治療薬、細胞または臓器線維化制御方法、細胞または臓器の線維化制御剤のスクリーニング方法に関する。 The present invention relates to a cell or organ fibrosis control agent using a nucleic acid, a diagnostic or therapeutic agent for a disease caused by a fibrotic disorder, a cell or organ fibrosis control method, and a screening method for a cell or organ fibrosis control agent About.
 核酸の1種であるマイクロRNA(miRNA)は、蛋白質に翻訳されない約22ヌクレオチドからなる小さな非コード一本鎖RNAであり、ヒトを含む生物に多数存在することが知られている(非特許文献1、2)。マイクロRNAは、単一又はクラスター化されたマイクロRNA前駆体に転写される遺伝子から生成される。すなわち、まず遺伝子から一次転写産物であるprimary-microRNA(pri-miRNA)が転写され、次いでpri-miRNAから成熟型マイクロRNAへの段階的プロセシングにおいて、特徴的なヘアピン構造を有する約70塩基のprecursor-microRNA(pre-miRNA)がpri-miRNAから生成される。さらに、Dicer介在によるプロセシングによりpre-miRNAから成熟型マイクロRNAが生成される(非特許文献3)。 MicroRNA (miRNA), which is a kind of nucleic acid, is a small non-coding single-stranded RNA consisting of about 22 nucleotides that are not translated into protein, and is known to exist in many organisms including humans (Non-Patent Documents). 1, 2). MicroRNAs are generated from genes that are transcribed into single or clustered microRNA precursors. In other words, the primary transcript, primary-microRNA (pri-miRNA), is first transcribed from the gene, and then in a stepwise process from pri-miRNA to mature microRNA, a precursor of about 70 bases with a characteristic hairpin structure. -microRNA (pre-miRNA) is generated from pri-miRNA. Furthermore, mature microRNA is generated from pre-miRNA by Dicer-mediated processing (Non-patent Document 3).
 成熟型マイクロRNAは、標的となるmRNAに相補的に結合してmRNAの翻訳を抑制するか、あるいはmRNAを分解することにより遺伝子発現の転写後制御に関与していると考えられている。マイクロRNAが標的mRNAの発現を抑制するメカニズムについては完全には明らかになっていないが、近年の研究により概要が解明されつつある。マイクロRNAは標的となるmRNAの3’-非翻訳領域(3’-UTR)中の部分的に相補的な配列に結合し、その翻訳を抑制し、または標的mRNAを分解することで発現を抑制する。上記の相補性は完全でなくても良いが、特にマイクロRNAの5’-末端側から2番目から8番目までの塩基の相補性が重要であることが示されており、この領域をマイクロRNAの「シード配列(seed sequence)」と呼ぶこともある(非特許文献4)。シード配列が共通するマイクロRNAは他の配列が異なっていても、共通の標的mRNAの発現を抑制することが示されている。従って、任意のmRNAの3’-末端に存在する配列と相補的
な配列をシード配列となるようにすることで、マイクロRNA様の活性を有するRNAを設計することができる。マイクロRNAはsiRNAとは異なり、通常「細胞内に天然に存在する」RNAのみを指すことが多いため、このようにして設計したマイクロRNA様配列を特に「人工マイクロRNA」と呼ぶ場合がある。
Mature microRNAs are thought to be involved in post-transcriptional control of gene expression by binding complementarily to the target mRNA and suppressing mRNA translation, or by degrading the mRNA. Although the mechanism by which microRNA suppresses the expression of target mRNA is not completely clarified, an outline has been elucidated by recent studies. MicroRNA binds to a partially complementary sequence in the 3'-untranslated region (3'-UTR) of the target mRNA, suppresses its translation, or suppresses expression by degrading the target mRNA To do. Although the above complementarity may not be perfect, it has been shown that complementarity of the second to eighth bases from the 5′-end side of the microRNA is particularly important. Is sometimes referred to as a “seed sequence” (Non-patent Document 4). It has been shown that microRNAs with a common seed sequence suppress the expression of a common target mRNA even if other sequences are different. Therefore, RNA having microRNA-like activity can be designed by using a sequence complementary to the sequence present at the 3′-end of any mRNA as a seed sequence. Unlike siRNA, microRNA usually refers only to RNA that is “naturally present in cells”. Therefore, a microRNA-like sequence designed in this way may be particularly referred to as “artificial microRNA”.
 2009年3月現在、マイクロRNAのデータベースmiRBase(http://microrna.sanger.ac.uk/)には、ヒトで706種、マウスで547種、ラットでは286種のマイクロRNAが登録されている。ヒトを含む哺乳類で発現するマイクロRNAの中で、その生理的機能に関して知られているものは、血球系分化に関与するmiR-181(非特許文献5)やインシュリン分泌に関与するmiR-375(非特許文献6)などごく一部のみであり、多くはその生理的活性が未解明である。ただし、線虫やショウジョウバエを用いた研究からマイクロRNAが生物の発生、分化に様々な重要な役割を果たしていることが明らかとなってきており、ヒト疾患との関係においても、特に癌との深い関係を示唆する報告がされている(非特許文献7)。 As of March 2009, the microRNA database miRBase (http://microrna.sanger.ac.uk/) contains 706 human, 547 mouse, and 286 rat microRNAs. . Among the microRNAs expressed in mammals including humans, those known for their physiological functions include miR-181 (Non-patent Document 5) involved in blood cell differentiation and miR-375 (involved in insulin secretion). Non-patent document 6) is only a small part, and many of them have unclear physiological activity. However, studies using nematodes and Drosophila have revealed that microRNAs play various important roles in the development and differentiation of living organisms. There has been a report suggesting a relationship (Non-patent Document 7).
 線維化は内臓などの組織を構成している結合組織と呼ばれる部分が異常増殖する現象のことで、肺、腎臓、肝臓、心臓、皮膚、血管など各種組織・臓器において起こり、疾患の原因となることが知られている。たとえば、心筋に線維化が生じたときには心臓の働きに異常が起き、呼吸困難や心悸亢進(動悸)などの症状が出る。また関節リウマチにおける骨の萎縮や変性、肝臓全体の線維化を示す肝硬変の病態なども、結合組織が異常増殖すなわち線維化した例としてよく知られている。腎臓においても線維化によって間質の肥大が起こり、尿細管の壊死あるいは減少により最終的には腎不全へと進行する。組織の線維化は組織中に存在する線維芽細胞の細胞外基質の異常産生がおもな原因となるが、それとともに組織を構成する上皮系細胞が間葉系細胞に形質転換して細胞外基質を産生し始めることも寄与していることが明らかになっており、これらは上皮間葉転換(EMT:Epithelial Mesenchymal Transition)と呼ばれている(非特許文献8)。これまでのマイクロRNAの研究は主に癌研究において進展しており、多くの癌においてマイクロRNAの発現が正常組織と異なっていること、マイクロRNAの発現プロファイル解析により癌の分類が可能であることなどが報告されている(非特許文献9)。また、これまでに見出されたヒトのマイクロRNAの約半数が、ヒト癌で知られている染色体異常あるいは染色体の脆弱部位に存在することも知られている(非特許文献10)。一方、臓器の線維化に対する治療には、現在コルチコステロイドなどの一般的な免疫抑制薬剤、および他の抗炎症治療が利用されている。しかし、線維化の制御に関与する機構は、炎症のものとは別の現象であるようであり、抗炎症療法は、線維化を減少させるかまたは防止する際に、常に有効であるわけではなく、臨床ニーズを満たしているとは言えない(非特許文献11)。したがって、線維化を減少させあるいは防止することを含めて、線維性障害を調節する新しい治療を開発する必要性がなおも存在する。 Fibrosis is a phenomenon in which the part called connective tissue that constitutes tissues such as internal organs grows abnormally and occurs in various tissues and organs such as lung, kidney, liver, heart, skin, blood vessels, and causes disease It is known. For example, when fibrosis occurs in the myocardium, abnormalities occur in the function of the heart, and symptoms such as dyspnea and increased heart rate (palpitations) appear. In addition, bone atrophy and degeneration in rheumatoid arthritis and the pathology of cirrhosis, which indicates fibrosis of the entire liver, are well known as examples of abnormal growth of the connective tissue, that is, fibrosis. In the kidney, the stromal hypertrophy occurs due to fibrosis, and eventually progresses to renal failure due to necrosis or decrease of tubules. Tissue fibrosis is mainly caused by abnormal production of the extracellular matrix of fibroblasts present in the tissue, but the epithelial cells constituting the tissue are transformed into mesenchymal cells together with the extracellular matrix. It has been clarified that starting to produce a substrate also contributes, and these are called epithelial-mesenchymal transition (EMT) (Non-patent Document 8). MicroRNA research so far has been progressing mainly in cancer research, and in many cancers, microRNA expression is different from normal tissues, and cancer can be classified by microRNA expression profile analysis. Have been reported (Non-Patent Document 9). It is also known that about half of the human microRNAs found so far exist in chromosomal abnormalities or fragile sites of chromosomes known in human cancer (Non-patent Document 10). On the other hand, general immunosuppressive drugs such as corticosteroids and other anti-inflammatory treatments are currently used for treatment of organ fibrosis. However, the mechanisms involved in controlling fibrosis appear to be a separate phenomenon from that of inflammation, and anti-inflammatory therapy is not always effective in reducing or preventing fibrosis. It cannot be said that the clinical needs are satisfied (Non-patent Document 11). Thus, there is still a need to develop new therapies that modulate fibrotic disorders, including reducing or preventing fibrosis.
 マイクロRNAは様々な遺伝子の発現制御に関与することから、マイクロRNAの異常はヒトの種々の疾患に関与していることが予想される。がんにおいては、miR-29cB細胞慢性リンパ性白血病(B-CLL)で欠失が見られる染色体13q14にmiR-15a/miR-16クラスターが含まれ、その欠失がB-CLLの原因の一つになっていると予測されること(非特許文献12)、B-CLLにおいてはさらにmiR-29とmiR-181の発現が低下しており、その標的の一つが癌原遺伝子として知られるTcl1であること(非特許文献13)が知られている。肺癌ではマイクロRNAの一つであるLet-7の発現が低下しており、その標的の一つが癌原遺伝子として知られるRasであること(非特許文献14、15)が知られている。また、癌における遺伝子の高メチル化修飾によりmiR-127やmiR-124aといったマイクロRNAの発現が減少しており、癌原遺伝子として知られるBcl6やCdk6がその標的であること(非特許文献16、17)などがあげられる。多くのマイクロRNAは癌細胞で発現が低下しているが、逆に癌細胞で遺伝子増幅や過剰発現が見られるマイクロRNAも存在する。例えば、悪性リンパ腫で遺伝子増幅が見られる領域には6種のマイクロRNAからなるクラスター(miR-17-92)が存在し、このmiRNAクラスター遺伝子をヒトB細胞リンパ腫のモデルマウスに強制発現させるとリンパ腫の発生が促進されることが知られている(非特許文献18)。また、以前からホジキンリンパ腫で過剰発現する、蛋白質をコードしない癌遺伝子候補とされていたBICと呼ばれる遺伝子は、miR-155をコードしていることも明らかとなっている(非特許文献19)。 Since microRNA is involved in the control of the expression of various genes, abnormalities in microRNA are expected to be involved in various human diseases. In cancer, miR-15a / miR-16 cluster is included in chromosome 13q14, which is deleted in miR-29cB cell chronic lymphocytic leukemia (B-CLL), and this deletion is one of the causes of B-CLL. That are predicted to be connected (Non-patent Document 12). In B-CLL, miR-29 and miR-181 expression is further reduced, and one of the targets is Tcl1, which is known as a proto-oncogene. (Non-patent Document 13) is known. In lung cancer, expression of Let-7, which is one of microRNAs, is decreased, and one of its targets is Ras known as a proto-oncogene (Non-patent Documents 14 and 15). Moreover, the expression of microRNAs such as miR-127 and miR-124a is reduced by hypermethylation modification of genes in cancer, and Bcl6 and Cdk6 known as proto-oncogenes are the targets (Non-patent Document 16, 17). Many microRNAs have decreased expression in cancer cells, but there are also microRNAs in which gene amplification and overexpression are seen in cancer cells. For example, in a region where gene amplification is observed in malignant lymphoma, there are 6 types of microRNA clusters (miR-17-92). When this miRNA cluster gene is forcibly expressed in a model mouse of human B cell lymphoma, lymphoma It is known that the generation of the above is promoted (Non-patent Document 18). It has also been clarified that a gene called BIC, which has been previously overexpressed in Hodgkin's lymphoma and was a candidate oncogene that does not encode a protein, encodes miR-155 (Non-patent Document 19).
 一方、これまでに報告されている線維化とマイクロRNAの関係の例としては、心臓において左心冠動脈閉塞により心筋梗塞を惹起したマウスあるいはヒト心筋梗塞においてmiR-29の発現が低下しており、培養心線維芽細胞でmiR-29の発現を抑制させると、線維化関連分子の一つであるCollagen A1, A2, A3の発現が有意に上昇することを示している(非特許文献20)。同じく心線維化については、心不全が自然発症するβ1-アドレナリン受容体トランスジェニックマウスにおいて、心臓でのマイクロRNAの発現を解析したところ、miR-21が経時的に上昇することを見出した報告もある(非特許文献21)。同論文ではmiR-21のシード配列を用いてバイオインフォーマティクスによるデータベース解析を行って、miR-21の標的の一つとしてRas/ERKシグナルを阻害する分子であるSprouty-1(SPRY1)を見出し、線維化の進展においてmiR-21がSPRY1の発現を抑制して、ERK-MAPKシグナルを活性化し、線維芽細胞の生存を調節していることを明らかにしている。 On the other hand, as an example of the relationship between fibrosis and microRNA reported so far, miR-29 expression is decreased in mice or human myocardial infarction that caused myocardial infarction due to left coronary artery occlusion in the heart, It has been shown that when miR-29 expression is suppressed in cultured cardiac fibroblasts, the expression of Collagen A1, A2, A3, which is one of the fibrosis-related molecules, is significantly increased (Non-patent Document 20). Similarly, with regard to cardiac fibrosis, in a β1-adrenergic receptor transgenic mouse with spontaneous heart failure, analysis of microRNA expression in the heart found that miR-21 increased with time. (Non-patent document 21). In this paper, we performed database analysis by bioinformatics using miR-21 seed sequence and found Sprouty-1 (SPRY1), a molecule that inhibits Ras / ERK signal as one of miR-21 targets. In the progress of fibrosis, miR-21 suppresses the expression of SPRY1, activates the ERK-MAPK signal, and regulates the survival of fibroblasts.
 このように病態とマイクロRNAの関係は近年多数報告されているが、その多くは癌細胞での発現異常を示すものであり、マイクロRNAの機能を示した報告は、Let-7、miR-143やmiR-145を癌細胞株に強制発現させることで癌細胞株の増殖を阻害したという報告(非特許文献22、23、24)、miR-16やmiR-34を強制発現させることにより癌細胞株の細胞周期を停止させたという報告(非特許文献25、26)、miR-372やmiR-373を強制発現させることにより細胞の形質転換を引き起こしたという報告(非特許文献27)などがある。
 一方、体外からマイクロRNAあるいはその前駆体を投与することにより該マイクロRNAの発現を増大させること、あるいはそのアンチセンスオリゴヌクレオチドを投与してマイクロRNAの発現を減少させることで臓器の線維化病変を改善させたという報告はほとんどなく、先に述べた心臓での報告で、miR-29の前駆体投与により心臓でのCollagen A1, A2, A3の発現が有意に亢進した例(非特許文献20)やmiR-21のアンチセンスオリゴヌクレオチドを投与して心障害負荷を掛けると心線維化が亢進したという例(非特許文献21)はあるものの、腎臓の線維化について解析を行った報告は皆無であるのが現状である。
As described above, many relations between disease states and microRNAs have been reported in recent years, but many of them show abnormal expression in cancer cells, and reports showing the function of microRNAs include Let-7, miR-143. Report that the growth of cancer cell lines was inhibited by forcibly expressing miR-145 and miR-145 (Non-Patent Documents 22, 23, 24), cancer cells by forcibly expressing miR-16 and miR-34 There are reports that the cell cycle of the strain was stopped (Non-patent Documents 25 and 26), reports that cell transformation was caused by forced expression of miR-372 and miR-373 (Non-patent Document 27), and the like. .
On the other hand, administration of microRNA or a precursor thereof from outside the body increases the expression of the microRNA, or administration of the antisense oligonucleotide reduces the expression of the microRNA to reduce fibrosis of the organ. There has been almost no report of improvement, and in the above-mentioned report in the heart, an example in which the expression of Collagen A1, A2, A3 in the heart was significantly increased by administration of the precursor of miR-29 (Non-patent Document 20) Although there is an example (Non-patent Document 21) that cardiac fibrosis is enhanced when a cardiac disorder is applied by administering an antisense oligonucleotide of miR-21 or miR-21, there is no report analyzing the fibrosis of the kidney There is the present situation.
 線維化臓器で発現変動しているマイクロRNAを同定し、その機能を解析し、疾患との関係を解明することにより、新しい治療薬及び診断薬が開発されることが期待される。
 特に、細胞または臓器の線維化促進または抑制を引き起こすマイクロRNAを見出すことは、線維化のメカニズムの理解に役立つのみならず、ヒトの臓器線維化の診断薬や治療薬の開発、さらにはそれらを利用した線維化の新しい診断法や治療法につながることが期待される。特に尿細管間質の線維化を特徴とする急性腎不全や糸球体腎炎、血管炎、糖尿病性腎症、高血圧性腎硬化症、HIV腎症、IgA腎症、ループス腎炎、間質性腎炎、尿管閉塞による閉塞腎、さらには肺線維症、肝硬変、動脈硬化症、強皮症、経皮経管冠動脈血管拡張術(PTCA)後の冠動脈再狭窄、間質性心筋炎、間質性膀胱炎、熱傷後の皮膚瘢痕化、中毒等に伴う線維症等など細胞の細胞外基質産生の異常、組織の過形成等が原因となっている疾患の診断薬・治療薬やそれらを利用した診断法・治療法の開発に貢献することが期待される。
It is expected that new therapeutic agents and diagnostic agents will be developed by identifying microRNAs whose expression changes in fibrotic organs, analyzing their functions, and elucidating the relationship with diseases.
In particular, finding microRNAs that promote or suppress fibrosis of cells or organs not only helps understand the mechanism of fibrosis, but also develops diagnostic and therapeutic agents for human organ fibrosis, and further It is expected to lead to new diagnostic methods and treatments for the fibrosis used. Acute renal failure and glomerulonephritis characterized by fibrosis of tubulointerstitium, vasculitis, diabetic nephropathy, hypertensive nephropathy, HIV nephropathy, IgA nephropathy, lupus nephritis, interstitial nephritis, Obstructed kidney due to ureteral obstruction, as well as pulmonary fibrosis, cirrhosis, arteriosclerosis, scleroderma, coronary restenosis after percutaneous transluminal coronary vasodilation (PTCA), interstitial myocarditis, interstitial bladder Diagnostic and therapeutic agents for diseases caused by abnormalities in extracellular matrix production of cells, tissue hyperplasia, such as inflammation, skin scarring after burns, fibrosis associated with poisoning, etc., and diagnosis using them It is expected to contribute to the development of methods and treatments.
 本発明の目的は、細胞または臓器の線維化制御に有用な核酸及び線維化制御剤、それらの利用法を提供することにある。この場合、線維化制御剤とは線維化抑制剤および線維化促進剤を指す。 An object of the present invention is to provide a nucleic acid and a fibrosis control agent useful for controlling fibrosis of cells or organs, and methods for using them. In this case, the fibrosis regulator refers to a fibrosis inhibitor and a fibrosis promoter.
 本発明は以下の[1]~[31]に関する。
[1]以下の(a)~(h)のいずれかの核酸を有効成分として含有する、細胞または臓器の線維化制御剤。
(a)配列番号1~27、28~594のいずれかで表される塩基配列からなる核酸
(b)配列番号1~27、28~594のいずれかで表される塩基配列からなる核酸を含有する、17~28塩基の核酸
(c)配列番号1~27、28~594のいずれかで表される塩基配列と90%以上の同一性を有する塩基配列からなる核酸
(d)配列番号1~27、28~594のいずれかで表される塩基配列からなる核酸の相補鎖とストリンジェントな条件でハイブリダイズする核酸
(e)配列番号1~27、28~594のいずれかで表される塩基配列の2~8番目の塩基配列を含む核酸
(f)配列番号595~1281のいずれかで表される塩基配列からなる核酸
(g)配列番号595~1281のいずれかで表される塩基配列と90%以上の同一性を有する塩基配列からなる核酸
(h)配列番号595~1281のいずれかで表される塩基配列からなる核酸の相補鎖とストリンジェントな条件でハイブリダイズする核酸
[2]核酸がマイクロRNAまたはマイクロRNA前駆体である、上記[1]に記載の細胞または臓器の線維化制御剤。
[3]上記[1]に記載の核酸の塩基配列に対して相補的な塩基配列からなる核酸を有効成分として含有する、細胞または臓器の線維化制御剤。
[4]上記[1]~[3]のいずれかに記載の核酸を発現するベクターを有効成分として含有する、細胞または臓器の線維化制御剤。
[5]上記[1]に記載の核酸の標的塩基配列を有する遺伝子の発現を抑制する物質を有効成分として含有する、細胞または臓器の線維化制御剤。
[6]上記[1]に記載の核酸の標的塩基配列を有する遺伝子の発現を促進する物質を有効成分として含有する、細胞または臓器の線維化制御剤。
[7]発現を抑制または促進する物質が核酸である、上記[5]または[6]に記載の細胞または臓器の線維化制御剤。
[8]核酸がsiRNAまたはアンチセンスオリゴヌクレオチドである、上記[7]に記載の細胞または臓器の線維化制御剤。
[9]上記[7]に記載の核酸を発現するベクターを有効成分として含有する、細胞または臓器の線維化制御剤。
[10]上記[1]~[3]のいずれかに記載の核酸、上記[4]に記載のベクター、または上記[5]~[8]のいずれかに記載の物質を有効成分として含有する、細胞または臓器の線維化に起因する疾患の治療薬または診断薬。
[11]上記[1]に記載の核酸の発現量、該核酸の変異、または該核酸をコードするゲノムの変異を検出する試薬を有効成分として含有する、細胞または臓器の線維化に起因する疾患の診断薬。
[12]細胞または臓器の線維化に起因する疾患が、急性腎不全、糸球体腎炎、血管炎、糖尿病性腎症、高血圧性腎硬化症、HIV腎症、IgA腎症、ループス腎炎、間質性腎炎、尿管閉塞による閉塞腎、肺線維症、肝硬変、動脈硬化症、強皮症、経皮経管冠動脈血管拡張術(PTCA)後の冠動脈再狭窄、間質性心筋炎、間質性膀胱炎、熱傷後の皮膚瘢痕化、または中毒に伴う線維症である、上記[10]または[11]に記載の治療薬または診断薬。
[13]細胞または臓器の線維化に起因する疾患が、急性腎不全、糸球体腎炎、血管炎、糖尿病性腎症、高血圧性腎硬化症、HIV腎症、IgA腎症、ループス腎炎、間質性腎炎、および尿管閉塞による閉塞腎からなる群から選択される腎障害に伴う線維症である、上記[10]または[11]に記載の治療薬または診断薬。
[14]上記[1]~[3]のいずれかに記載の核酸、上記[4]に記載のベクター、または上記[5]~[8]のいずれかに記載の物質を有効量投与することを特徴とする、細胞または臓器の線維化に起因する疾患の治療方法。
[15]上記[1]に記載の核酸の発現量、該核酸の変異、または該核酸をコードするゲノムの変異を検出することを特徴とする、細胞または臓器の線維化に起因する疾患の診断方法。
[16]細胞または臓器の線維化に起因する疾患が、急性腎不全、糸球体腎炎、血管炎、糖尿病性腎症、高血圧性腎硬化症、HIV腎症、IgA腎症、ループス腎炎、間質性腎炎、尿管閉塞による閉塞腎、肺線維症、肝硬変、動脈硬化症、強皮症、経皮経管冠動脈血管拡張術(PTCA)後の冠動脈再狭窄、間質性心筋炎、間質性膀胱炎、熱傷後の皮膚瘢痕化、または中毒に伴う線維症である、上記[14]または[15]に記載の治療または診断方法。
[17]細胞または臓器の線維化に起因する疾患が、急性腎不全、糸球体腎炎、血管炎、糖尿病性腎症、高血圧性腎硬化症、HIV腎症、IgA腎症、ループス腎炎、間質性腎炎、および尿管閉塞による閉塞腎からなる群から選択される腎障害に伴う線維症である、上記[14]または[15]に記載の治療または診断方法。
[18]細胞または臓器の線維化に起因する疾患の治療薬の製造のための、上記[1]~[3]のいずれかに記載の核酸、上記[4]に記載のベクター、または上記[5]~[8]のいずれかに記載の物質の使用。
[19]細胞または臓器の線維化に起因する疾患が、急性腎不全、糸球体腎炎、血管炎、糖尿病性腎症、高血圧性腎硬化症、HIV腎症、IgA腎症、ループス腎炎、間質性腎炎、尿管閉塞による閉塞腎、肺線維症、肝硬変、動脈硬化症、強皮症、経皮経管冠動脈血管拡張術(PTCA)後の冠動脈再狭窄、間質性心筋炎、間質性膀胱炎、熱傷後の皮膚瘢痕化、または中毒に伴う線維症である、上記[18]に記載の使用。
[20]細胞または臓器の線維化に起因する疾患が、急性腎不全、糸球体腎炎、血管炎、糖尿病性腎症、高血圧性腎硬化症、HIV腎症、IgA腎症、ループス腎炎、間質性腎炎、および尿管閉塞による閉塞腎からなる群から選択される腎障害に伴う線維症である、上記[18]に記載の使用。
[21]上記[1]~[3]のいずれかに記載の核酸を有効成分として含有する、細胞または臓器の線維化制御剤の有効成分である核酸の標的遺伝子の発現制御剤。
[22]上記[4]に記載のベクターを有効成分として含有する、細胞または臓器の線維化制御剤の有効成分である核酸の標的遺伝子の発現制御剤。
[23]上記[1]~[3]のいずれかに記載の核酸を用いることを特徴とする、細胞または臓器の線維化制御方法。
[24]上記[4]に記載のベクターを用いることを特徴とする、細胞または臓器の線維化制御方法。
[25]上記[1]に記載の核酸の標的遺伝子の発現を抑制または促進する物質を用いることを特徴とする、細胞または臓器の線維化制御方法。
[26]発現を抑制または促進する物質が核酸である、上記[25]に記載の細胞または臓器の線維化制御方法。
[27]核酸がsiRNAまたはアンチセンスオリゴヌクレオチドである、上記[26]に記載の細胞または臓器の線維化制御方法。
[28]上記[26]に記載の核酸を発現するベクターを用いる、細胞または臓器の線維化制御方法。
[29]上記[1]~[3]のいずれかに記載の核酸を用いることを特徴とする、細胞または臓器の繊維化制御剤の有効成分である核酸の標的遺伝子の発現制御方法。
[30]上記[4]に記載のベクターを用いることを特徴とする、細胞または臓器の繊維化制御剤の有効成分である核酸の標的遺伝子の発現制御方法。
[31]上記[1]に記載の核酸の発現または機能を促進または抑制させることを指標とする、細胞または臓器の線維化制御剤のスクリーニング方法。
The present invention relates to the following [1] to [31].
[1] A cell or organ fibrosis control agent comprising any one of the following nucleic acids (a) to (h) as an active ingredient.
(A) Nucleic acid comprising a base sequence represented by any of SEQ ID NOs: 1-27, 28-594 (b) Containing a nucleic acid comprising a base sequence represented by any of SEQ ID NOs: 1-27, 28-594 A nucleic acid consisting of a base sequence having a nucleotide sequence of 90% or more (d) a sequence number 1 to 1 to a nucleic acid of 17 to 28 bases (c) a sequence represented by any one of SEQ ID NOs: 1 to 27 and 28 to 594 A nucleic acid that hybridizes under stringent conditions with a complementary strand of a nucleic acid comprising the nucleotide sequence represented by any one of 27 and 28 to 594 (e) a base represented by any one of SEQ ID NOs: 1 to 27 and 28 to 594 A nucleic acid comprising the second to eighth base sequences of the sequence (f) a nucleic acid comprising the base sequence represented by any one of SEQ ID NOs: 595 to 1281 (g) a base sequence represented by any one of the sequence numbers 595 to 1281; 90% or more identity (H) Nucleic acid that hybridizes under stringent conditions with the complementary strand of the nucleic acid consisting of the base sequence represented by any of SEQ ID NOs: 595 to 1281 [2] The nucleic acid is microRNA or microRNA precursor The cell or organ fibrosis regulator according to [1] above, which is a body.
[3] A cell or organ fibrosis control agent comprising, as an active ingredient, a nucleic acid having a base sequence complementary to the base sequence of the nucleic acid described in [1].
[4] A cell or organ fibrosis control agent comprising a vector that expresses the nucleic acid according to any one of [1] to [3] as an active ingredient.
[5] A cell or organ fibrosis control agent comprising, as an active ingredient, a substance that suppresses the expression of the gene having the target base sequence of the nucleic acid according to [1].
[6] A cell or organ fibrosis control agent comprising as an active ingredient a substance that promotes expression of a gene having the target base sequence of the nucleic acid according to [1].
[7] The cell or organ fibrosis regulator according to [5] or [6] above, wherein the substance that suppresses or promotes expression is a nucleic acid.
[8] The cell or organ fibrosis regulator according to [7] above, wherein the nucleic acid is siRNA or antisense oligonucleotide.
[9] A cell or organ fibrosis control agent comprising the vector expressing the nucleic acid according to [7] as an active ingredient.
[10] The nucleic acid according to any one of [1] to [3] above, the vector according to [4] above, or the substance according to any of [5] to [8] above as an active ingredient A therapeutic or diagnostic agent for diseases caused by fibrosis of cells or organs.
[11] A disease caused by fibrosis of a cell or an organ containing as an active ingredient a reagent for detecting the expression level of the nucleic acid according to [1], a mutation of the nucleic acid, or a mutation of a genome encoding the nucleic acid Diagnostics.
[12] Diseases caused by fibrosis of cells or organs are acute renal failure, glomerulonephritis, vasculitis, diabetic nephropathy, hypertensive nephrosclerosis, HIV nephropathy, IgA nephropathy, lupus nephritis, stroma Nephritis, obstructed kidney due to ureteral obstruction, pulmonary fibrosis, cirrhosis, arteriosclerosis, scleroderma, coronary restenosis after percutaneous transluminal coronary vasodilation (PTCA), interstitial myocarditis, interstitial The therapeutic or diagnostic agent according to the above [10] or [11], which is cystitis, skin scarring after burn injury, or fibrosis associated with poisoning.
[13] Diseases caused by fibrosis of cells or organs are acute renal failure, glomerulonephritis, vasculitis, diabetic nephropathy, hypertensive nephrosclerosis, HIV nephropathy, IgA nephropathy, lupus nephritis, stroma The therapeutic agent or diagnostic agent according to [10] or [11] above, which is fibrosis associated with renal disorder selected from the group consisting of pneumonia and obstructive kidney due to ureteral obstruction.
[14] Administering an effective amount of the nucleic acid according to any one of [1] to [3] above, the vector according to [4] above, or the substance according to any of [5] to [8] above. A method for treating a disease caused by fibrosis of a cell or organ, characterized by comprising:
[15] Diagnosis of a disease caused by fibrosis of a cell or organ, wherein the expression level of the nucleic acid according to [1], a mutation of the nucleic acid, or a mutation of a genome encoding the nucleic acid is detected Method.
[16] Diseases caused by fibrosis of cells or organs are acute renal failure, glomerulonephritis, vasculitis, diabetic nephropathy, hypertensive nephrosclerosis, HIV nephropathy, IgA nephropathy, lupus nephritis, stroma Nephritis, obstructed kidney due to ureteral obstruction, pulmonary fibrosis, cirrhosis, arteriosclerosis, scleroderma, coronary restenosis after percutaneous transluminal coronary vasodilation (PTCA), interstitial myocarditis, interstitial The method of treatment or diagnosis according to [14] or [15] above, which is cystitis, skin scarring after burn injury, or fibrosis associated with poisoning.
[17] Diseases caused by fibrosis of cells or organs are acute renal failure, glomerulonephritis, vasculitis, diabetic nephropathy, hypertensive nephrosclerosis, HIV nephropathy, IgA nephropathy, lupus nephritis, stroma The method of treatment or diagnosis according to [14] or [15] above, wherein the disease is fibrosis associated with renal disorder selected from the group consisting of nephritis and obstructed kidney due to ureteral obstruction.
[18] The nucleic acid according to any one of [1] to [3] above, the vector according to [4] above, or the above [for manufacturing a therapeutic agent for a disease caused by fibrosis of a cell or organ Use of the substance according to any one of [5] to [8].
[19] Diseases caused by fibrosis of cells or organs are acute renal failure, glomerulonephritis, vasculitis, diabetic nephropathy, hypertensive nephrosclerosis, HIV nephropathy, IgA nephropathy, lupus nephritis, stroma Nephritis, obstructed kidney due to ureteral obstruction, pulmonary fibrosis, cirrhosis, arteriosclerosis, scleroderma, coronary restenosis after percutaneous transluminal coronary vasodilation (PTCA), interstitial myocarditis, interstitial The use according to [18] above, which is fibrosis associated with cystitis, skin scarring after burns, or poisoning.
[20] Diseases caused by fibrosis of cells or organs are acute renal failure, glomerulonephritis, vasculitis, diabetic nephropathy, hypertensive nephrosclerosis, HIV nephropathy, IgA nephropathy, lupus nephritis, stroma. The use according to [18] above, which is fibrosis associated with renal disorder selected from the group consisting of pneumonia and obstructive kidney due to ureteral obstruction.
[21] An agent for controlling expression of a target gene of a nucleic acid, which is an active ingredient of a fibrosis regulator for cells or organs, containing the nucleic acid according to any one of [1] to [3] as an active ingredient.
[22] A nucleic acid target gene expression control agent, which is an active ingredient of a cell or organ fibrosis control agent, comprising the vector according to [4] as an active ingredient.
[23] A method for controlling fibrosis of a cell or organ, comprising using the nucleic acid according to any one of [1] to [3] above.
[24] A method for controlling fibrosis of a cell or organ, characterized by using the vector according to [4] above.
[25] A method for controlling fibrosis of a cell or organ, comprising using a substance that suppresses or promotes the expression of the target gene of the nucleic acid according to [1].
[26] The method for controlling fibrosis of a cell or organ according to the above [25], wherein the substance that suppresses or promotes expression is a nucleic acid.
[27] The method for controlling fibrosis of a cell or organ according to the above [26], wherein the nucleic acid is siRNA or antisense oligonucleotide.
[28] A method for controlling fibrosis of a cell or organ using the vector expressing the nucleic acid according to [26].
[29] A method for controlling expression of a target gene of a nucleic acid that is an active ingredient of a fibrosis control agent for cells or organs, wherein the nucleic acid according to any one of [1] to [3] above is used.
[30] A method for controlling expression of a target gene of a nucleic acid, which is an active ingredient of a fibrosis control agent for cells or organs, using the vector according to [4] above.
[31] A screening method for a cell or organ fibrosis regulator using as an index the promotion or suppression of the expression or function of the nucleic acid according to [1].
 本発明により、細胞または臓器の線維化抑制剤または線維化促進剤、細胞の細胞外基質産生異常またはEMTに起因する疾患の診断薬または治療薬、マイクロRNAなどの核酸の標的遺伝子の発現抑制剤または発現促進剤、細胞の線維化抑制方法または線維化促進方法を提供することができる。 According to the present invention, a fibrosis inhibitor or fibrosis promoter for cells or organs, a diagnostic or therapeutic agent for diseases caused by abnormal extracellular matrix production of cells or EMT, and an expression inhibitor for target genes of nucleic acids such as microRNA Alternatively, an expression promoter, a method for suppressing cell fibrosis, or a method for promoting fibrosis can be provided.
図1は、Collagen Iの遺伝子発現をreal-time RT-PCRにて解析した結果を示す。コントロールRNA、miR-30b、miR-486、let-7iを投与した後の肝臓におけるCollagen Iの発現をコントロールとの相対比で示す。FIG. 1 shows the results of analysis of collagen-I gene expression by real-time RT-PCR. The expression of Collagen I in the liver after administration of control RNA, miR-30b, miR-486, let-7i is shown as a relative ratio to the control. 図2は、TGF-βの遺伝子発現をreal-time RT-PCRにて解析した結果を示す。コントロールマイクロRNA、miR-30b、miR-486、let-7iを投与した後の肝臓におけるTGF-βの発現をコントロールとの相対比で示す。FIG. 2 shows the results of analysis of TGF-β gene expression by real-time RT-PCR. The expression of TGF-β in the liver after administration of control microRNA, miR-30b, miR-486, let-7i is shown as a relative ratio to the control.
 本発明で用いる核酸としては、ヌクレオチドおよび該ヌクレオチドと同等の機能を有する分子が重合した分子であればいかなるものでもよく、例えば、リボヌクレオチドの重合体であるRNA、デオキシリボヌクレオチドの重合体であるDNA、リボヌクレオチドおよびデオキシリボヌクレオチドが混合した重合体、および、ヌクレオチド類似体を含むヌクレオチド重合体をあげることができ、さらに、核酸誘導体を含むヌクレオチド重合体であってもよい。また本発明における核酸は、一本鎖核酸または二本鎖核酸であってもよい。また二本鎖核酸には、一方の鎖に対し、他方の鎖がストリンジェントな条件でハイブリダイズする二本鎖核酸も含まれる。 The nucleic acid used in the present invention may be any molecule as long as nucleotides and molecules having functions equivalent to the nucleotides are polymerized, such as RNA that is a ribonucleotide polymer, DNA that is a deoxyribonucleotide polymer. , A polymer in which ribonucleotides and deoxyribonucleotides are mixed, and a nucleotide polymer containing a nucleotide analog, and further a nucleotide polymer containing a nucleic acid derivative. The nucleic acid in the present invention may be a single-stranded nucleic acid or a double-stranded nucleic acid. The double-stranded nucleic acid also includes a double-stranded nucleic acid in which one strand is hybridized under stringent conditions to the other strand.
 本発明においてヌクレオチド類似体としては、RNAまたはDNAと比較して、ヌクレアーゼ耐性を向上させるため、安定化させるため、相補鎖核酸とのアフィニティーをあげるため、細胞透過性をあげるため、あるいは可視化させるために、リボヌクレオチド、デオキシリボヌクレオチド、RNAまたはDNAに修飾を施した分子であればいかなる分子であってもよい。例えば、糖部修飾ヌクレオチド類似体やリン酸ジエステル結合修飾ヌクレオチド類似体等があげられる。 In the present invention, nucleotide analogues are used to improve nuclease resistance, to stabilize, to increase affinity with complementary strand nucleic acids, to increase cell permeability, or to be visualized as compared to RNA or DNA. In addition, any molecule may be used as long as it is a molecule obtained by modifying ribonucleotides, deoxyribonucleotides, RNA, or DNA. Examples thereof include sugar moiety-modified nucleotide analogs and phosphodiester bond-modified nucleotide analogs.
 糖部修飾ヌクレオチド類似体としては、ヌクレオチドの糖の化学構造の一部あるいは全てに対し、任意の化学構造物質を付加あるいは置換したものであればいかなるものでもよく、例えば、2’-O-メチルリボースで置換されたヌクレオチド類似体、2’-O-プロピルリボースで置換されたヌクレオチド類似体、2’-メトキシエトキシリボースで置換されたヌクレオチド類似体、2’-O-メトキシエチルリボースで置換されたヌクレオチド類似体、2’-O-[2-(グアニジウム)エチル]リボースで置換されたヌクレオチド類似体、2’-O-フルオロリボースで置換されたヌクレオチド類似体、糖部に架橋構造を導入することにより2つの環状構造を有する架橋構造型人工核酸(Bridged Nucleic Acid)(BNA)、より具体的には、2’位の酸素原子と4’位の炭素原子がメチレンを介して架橋したロックト人工核酸(Locked Nucleic Acid)(LNA)、およびエチレン架橋構造型人工核酸(Ethylene bridged nucleic acid)(ENA)[Nucleic Acid Research, 32, e175(2004)]があげられ、さらにペプチド核酸(PNA)[Acc. Chem. Res., 32, 624 (1999)]、オキシペプチド核酸(OPNA)[J. Am. Chem. Soc., 123, 4653 (2001)]、およびペプチドリボ核酸(PRNA)[J. Am. Chem. Soc., 122, 6900 (2000)]等をあげることができる。 The sugar moiety-modified nucleotide analog may be any one obtained by adding or substituting any chemical structural substance to a part or all of the chemical structure of the sugar of the nucleotide. For example, 2'-O-methyl Nucleotide analogues substituted with ribose, nucleotide analogues substituted with 2'-O-propylribose, nucleotide analogues substituted with 2'-methoxyethoxyribose, substituted with 2'-O-methoxyethylribose Nucleotide analogues, nucleotide analogues substituted with 2'-O- [2- (guanidinium) ethyl] ribose, nucleotide analogues substituted with 2'-O-fluororibose, introducing a bridging structure into the sugar moiety Bridged Nucleic Acid (BNA) having two circular structures, more specifically, 2′-position oxygen atom and 4′-position charcoal Locked artificial nucleic acid in which atoms are bridged via methylene (Locked Nucleic Acid) (LNA) , and ethylene bridged structure type artificial nucleic acid (Ethylene bridged nucleic acid) (ENA ) [Nucleic Acid Research, 32, e175 (2004)] is mentioned Furthermore, peptide nucleic acids (PNA) [Acc. Chem. Res., 32 , 624 (1999)], oxypeptide nucleic acids (OPNA) [J. Am. Chem. Soc., 123 , 4653 (2001)], and peptides Ribonucleic acid (PRNA) [J. Am. Chem. Soc., 122 , 6900 (2000)] and the like can be mentioned.
 リン酸ジエステル結合修飾ヌクレオチド類似体としては、ヌクレオチドのリン酸ジエステル結合の化学構造の一部あるいは全てに対し、任意の化学物質を付加あるいは置換したものであればいかなるものでもよく、例えば、ホスフォロチオエート結合に置換されたヌクレオチド類似体、N3'-P5'ホスフォアミデート結合に置換されたヌクレオチド類似体等をあげることができる[細胞工学, 16, 1463-1473 (1997)][RNAi法とアンチセンス法、講談社(2005)]。 The phosphodiester bond-modified nucleotide analog may be any one obtained by adding or substituting an arbitrary chemical substance to a part or all of the chemical structure of a phosphodiester bond of a nucleotide. Examples include nucleotide analogues substituted with thioate linkages, nucleotide analogues substituted with N3'-P5 'phosphoramidate linkages [Cell engineering, 16 , 1463-1473 (1997)] [RNAi method And Antisense, Kodansha (2005)].
 核酸誘導体としては、核酸に比べ、ヌクレアーゼ耐性を向上させるため、安定化させるため、相補鎖核酸とのアフィニティーをあげるため、細胞透過性をあげるため、あるいは可視化させるために、該核酸に別の化学物質を付加した分子であればいかなる分子でもよく、例えば、5’-ポリアミン付加誘導体、コレステロール付加誘導体、ステロイド付加誘導体、胆汁酸付加誘導体、ビタミン付加誘導体、Cy5付加誘導体、Cy3付加誘導体、6-FAM付加誘導体、およびビオチン付加誘導体等をあげることができる。 As a nucleic acid derivative, in order to improve nuclease resistance, to stabilize, to increase affinity with a complementary strand nucleic acid, to increase cell permeability or to be visualized as compared with a nucleic acid, a different chemical is used. Any molecule can be used as long as it is a molecule to which a substance is added, for example, 5′-polyamine addition derivative, cholesterol addition derivative, steroid addition derivative, bile acid addition derivative, vitamin addition derivative, Cy5 addition derivative, Cy3 addition derivative, 6-FAM Examples include addition derivatives, biotin addition derivatives, and the like.
 核酸としては、例えば以下の(a)~(k)で示される核酸をあげることができる。
(a)配列番号1~594のいずれかで表される塩基配列からなる核酸
(b)配列番号1~594のいずれかで表される塩基配列からなる核酸を含有する、17~28塩基の核酸
(c)配列番号1~594のいずれかで表される塩基配列と90%以上の同一性を有する塩基配列からなる核酸
(d)配列番号1~594のいずれかで表される塩基配列からなる核酸の相補鎖とストリンジェントな条件でハイブリダイズする核酸
(e)配列番号1~594のいずれかで表される塩基配列の2~8番目の塩基配列を含む核酸
(f)(a)~(e)の核酸と、該核酸の塩基配列に対して相補的な塩基配列を含む核酸とからなる二本鎖核酸、または該二本鎖核酸を含有する核酸
(g)(a)~(e)の核酸と、該核酸とストリンジェントな条件でハイブリダイズする核酸とからなる二本鎖核酸、または該二本鎖核酸を含有する核酸
(h)(f)または(g)の二本鎖核酸が8~28塩基からなるスペーサーオリゴヌクレオチドで連結されたヘアピン構造を有する一本鎖核酸、または該一本鎖核酸を含有する核酸
(i)配列番号595~1281のいずれかで表される塩基配列からなる核酸
(j)配列番号595~1281のいずれかで表される塩基配列と90%以上の同一性を有する塩基配列からなる核酸
(k)配列番号595~1281のいずれかで表される塩基配列からなる核酸の相補鎖とストリンジェントな条件でハイブリダイズする核酸。
Examples of the nucleic acid include nucleic acids represented by the following (a) to (k).
(A) Nucleic acid comprising a base sequence represented by any of SEQ ID NOs: 1 to 594 (b) Nucleic acid comprising 17 to 28 bases comprising a nucleic acid comprising a base sequence represented by any of SEQ ID NOs: 1 to 594 (C) a nucleic acid comprising a base sequence represented by any one of SEQ ID NOs: 1 to 594 and having a nucleotide sequence having 90% or more identity (d) comprising a base sequence represented by any of SEQ ID NOs: 1 to 594 Nucleic acid that hybridizes with a complementary strand of nucleic acid under stringent conditions (e) A nucleic acid comprising the second to eighth base sequences of the base sequence represented by any one of SEQ ID NOs: 1 to 594 (f) (a) to ( a double-stranded nucleic acid comprising the nucleic acid of e) and a nucleic acid comprising a base sequence complementary to the base sequence of the nucleic acid, or a nucleic acid containing the double-stranded nucleic acid (g) (a) to (e) Nucleic acids that hybridize with these nucleic acids under stringent conditions Or a hairpin structure in which a double-stranded nucleic acid (h), (f) or (g) containing the double-stranded nucleic acid is linked with a spacer oligonucleotide consisting of 8 to 28 bases. A single-stranded nucleic acid or a nucleic acid containing the single-stranded nucleic acid (i) a nucleic acid consisting of a base sequence represented by any of SEQ ID NOs: 595 to 1281 (j) represented by any of SEQ ID NOs: 595 to 1281 A nucleic acid comprising a nucleotide sequence having 90% or more identity to the nucleotide sequence (k) a nucleic acid that hybridizes under stringent conditions with a complementary strand of a nucleic acid comprising the nucleotide sequence represented by any of SEQ ID NOs: 595 to 1281 .
 上記核酸としては、マイクロRNAが好ましく用いられる。マイクロRNAとは、17~28塩基長からなる一本鎖RNAをいう。マイクロRNAの該配列を含む周辺ゲノム配列はヘアピン構造を形成し得る配列を有しており、マイクロRNAは該ヘアピンのいずれか片鎖から切り出され得る。マイクロRNAは、その標的となるmRNAに相補的に結合してmRNAの翻訳を抑制し、あるいはmRNAの分解を促進することで遺伝子発現の転写後制御を行う。 As the nucleic acid, microRNA is preferably used. MicroRNA refers to single-stranded RNA having a length of 17 to 28 bases. The surrounding genomic sequence containing the microRNA sequence has a sequence that can form a hairpin structure, and the microRNA can be excised from either strand of the hairpin. MicroRNAs complementarily bind to their target mRNA and suppress mRNA translation, or promote post-transcriptional control of gene expression by promoting mRNA degradation.
 マイクロRNAとしては、例えば、配列番号1~27のいずれかで表される塩基配列からなるヒトマイクロRNAをあげることができる。さらに配列番号1~27のいずれかで表される塩基配列からなるヒトマイクロRNAと同一の機能を有するマイクロRNAとして、該ヒトマイクロRNAのオーソログである、配列番号28~486のいずれかで表される塩基配列からなる核酸をあげることができる。具体的な例として、配列番号1のヒトマイクロRNAのオーソログは配列番号28~33、37~39で示される塩基配列からなるものがあげられる。配列番号1~27で表される塩基配列からなるマイクロRNAとそのオーソログの対応表を表1に示す。 Examples of microRNA include human microRNA having a base sequence represented by any of SEQ ID NOs: 1 to 27. Furthermore, as a microRNA having the same function as the human microRNA consisting of the base sequence represented by any of SEQ ID NOs: 1-27, it is represented by any of SEQ ID NOs: 28-486, which is an ortholog of the human microRNA. A nucleic acid having a base sequence can be mentioned. As a specific example, the ortholog of the human microRNA of SEQ ID NO: 1 includes those consisting of the base sequences represented by SEQ ID NOs: 28 to 33 and 37 to 39. Table 1 shows a correspondence table between microRNAs having the base sequences represented by SEQ ID NOs: 1 to 27 and their orthologs.
Figure JPOXMLDOC01-appb-T000001

 
Figure JPOXMLDOC01-appb-T000001

 
 なお、表1及び後述する表2の最上段において示されている生物種はそれぞれ以下の通りである。hsa、Homo sapiens、ヒト; mmu、Mus musculus、マウス; rno、Rattus norvegicus、ラット; cgr、Cricetulus griseus、チャイニーズハムスター; age、Ateles geoffroyi、アカクモザル; lla、Lagothrix lagotricha、フンボルトウーリーモンキー; sla、Saguinus labiatus、ムネアカタマリン; mml、Macaca mulatta、アカゲザル; mne、Macaca nemestrina、ブタオザル; pbi、Pygathrix bieti、クロキンシコウ; ggo、Gorilla gorilla、ゴリラ; ppa、Pan paniscus、ボノボ; ptr、Pan troglodytes、チンパンジー; ppy、Pongo pygmaeus、オランウータン; ssy, Symphalangus syndactylus、フクロテナガザル; lca、Lemur catta、ワオキツネザル; oan、Ornithorhynchus anatinus、カモノハシ; cfa、Canis familiaris、イヌ; mdo、Monodelphis domestica、ハイイロジネズミオポッサム; gga、Gallus gallus、ニワトリ; xla、Xenopus laevis、アフリカツメガエル; xtr、Xenopus tropicalis、ニシツメガエル; bta、Bos taurus、ウシ; oar、Ovis aries、ヒツジ; ssc、Sus scrofa、ブタ; dre、Danio rerio、ゼブラフィッシュ; fru、Fugu rubripes、トラフグ; tni、Tetraodon nigroviridis、ミドリフグヒトマイクロRNAとそのオーソログは、その配列同一性が高いことから、同様の機能を有していると考えられる。また、マイクロRNA名末尾に付加されたアルファベットによりサブグループ化されているマイクロRNA同士もまた、その配列同一性が高いことから、同様の機能を有していると考えられる。 The species shown in the top row of Table 1 and Table 2 to be described later are as follows. hsa, Homo sapiens, humans; mmu, Mus musculus, mice; rno, Rattus norvegicus, rats; cgr, Cricetulus griseus, Chinese hamsters; age, Ateles geoffroyi, red spider monkeys; Mnea catamarin; mml, Macaca mulatta, rhesus monkey; mne, Macaca nemestrina, pigtail monkey; pbi, Pygathrix bieti, black goldfish; ggo, Gorilla gorilla, gorilla; ppa, Pan paniscus, bonobo; ptr, Pan trogopy; Ssy, Symphalangus syndactylus, octopus gibbon; lca, Lemur catta, ring-tailed lemur; oan, Ornithorhynchus anatinus, platypus; cfa, Canis familiaris, dog; mdo, Monodelphis Xla, Xenopus laevis, Xenopus laevis; xtr, Xenopus tropicalis, Nishimegaru; bta, Bos taurus, cattle; oar, Ovis aries, sheep; ssc, Sus scrofa, pig; dre, Danio rerio, zebrafish , Trufffish; tni, Tetraodon nigroviridis, greenfish human microRNA and its ortholog are considered to have similar functions because of their high sequence identity. In addition, microRNAs that are subgrouped by the alphabet added to the end of the microRNA name also have similar functions because of their high sequence identity.
 マイクロRNAがその標的遺伝子のmRNAの翻訳を抑制するメカニズムとして、マイクロRNAの5’末端側2~8番目の塩基配列に対して相補的な塩基配列を有するmRNAは、マイクロRNA標的遺伝子として認識されることが知られている[Current Biology, 15, R458-R460 (2005)]。このメカニズムにより、該mRNAの発現がマイクロRNAによって抑制される。従って、5’末端側2~8番目が同じ塩基配列を有するマイクロRNAは、同じmRNAの発現を抑制して同様の機能を有する。配列番号1~27のいずれかで表される塩基配列からなるマイクロRNAと5’末端側2~8番目が同じ塩基配列を有するマイクロRNAとして、配列番号487~594のいずれかで表される塩基配列からなる核酸をあげることができる。該マイクロRNAの具体的な例としては、配列番号1で表される塩基配列からなるマイクロRNAに対しては、配列番号487、488で表される塩基配列からなるマイクロRNAをあげることができる。また、人工マイクロRNAも、マイクロRNAに含まれる。配列番号1~27で表される塩基配列からなるマイクロRNAとその5’末端側2~8番目が同じ塩基配列を有するマイクロRNAの対応表を表2に示す。共通のシード配列を有するマイクロRNAは、その標的塩基配列が同一と考えられることから、同様の機能を有していると考えられる。 As a mechanism by which microRNA suppresses translation of mRNA of its target gene, mRNA having a base sequence complementary to the 2-8th base sequence on the 5 'end side of microRNA is recognized as a microRNA target gene. [Current Biology, 15 , R458-R460 (2005)]. By this mechanism, the expression of the mRNA is suppressed by the microRNA. Accordingly, microRNAs having the same base sequence on the 2nd to 8th positions on the 5 ′ end side have the same function by suppressing the expression of the same mRNA. A base represented by any one of SEQ ID NOs: 487 to 594 as a microRNA having the same base sequence at the 5 'end side from the microRNA having the base sequence represented by any one of SEQ ID NOs: 1 to 27 A nucleic acid consisting of a sequence can be mentioned. As a specific example of the microRNA, for the microRNA consisting of the base sequence represented by SEQ ID NO: 1, microRNA consisting of the base sequence represented by SEQ ID NOs: 487 and 488 can be mentioned. Artificial microRNA is also included in microRNA. Table 2 shows a correspondence table between the microRNAs having the base sequences represented by SEQ ID NOs: 1 to 27 and the microRNAs having the same base sequence at the second to eighth positions on the 5 ′ end. MicroRNAs having a common seed sequence are considered to have the same function because their target base sequences are considered identical.
Figure JPOXMLDOC01-appb-T000002

 
Figure JPOXMLDOC01-appb-T000002

 
 上記核酸としては、マイクロRNA前駆体もまた好ましく用いられる。マイクロRNA前駆体とは、上記核酸を含む約50~約200塩基長、より好ましくは約70~約100塩基長の核酸であり、かつ、ヘアピン構造を形成し得る核酸である。マイクロRNAは、Dicerと呼ばれる蛋白質によるプロセシングを経て、マイクロRNA前駆体から生成される。 As the nucleic acid, a microRNA precursor is also preferably used. The microRNA precursor is a nucleic acid having a length of about 50 to about 200 bases, more preferably about 70 to about 100 bases including the above-mentioned nucleic acid, and can form a hairpin structure. MicroRNA is produced from a microRNA precursor through processing by a protein called Dicer.
 マイクロRNA前駆体として、例えば、配列番号1のヒトマイクロRNAに対しては配列番号595で表される塩基配列からなる核酸をあげることができる。また、配列番号2~594のいずれかで表される塩基配列からなるマイクロRNAに対しては配列番号596~1281のいずれかで表される塩基配列からなる核酸をあげることができる。本発明におけるマイクロRNAとマイクロRNA前駆体の対応を表3に示す。また本発明において、マイクロRNA前駆体には人工マイクロRNA前駆体が含まれる。 Examples of the microRNA precursor include a nucleic acid having a base sequence represented by SEQ ID NO: 595 for human microRNA of SEQ ID NO: 1. In addition, for a microRNA having a base sequence represented by any of SEQ ID NOs: 2 to 594, a nucleic acid having a base sequence represented by any of SEQ ID NOs: 596 to 1281 can be exemplified. Table 3 shows the correspondence between the microRNA and the microRNA precursor in the present invention. In the present invention, the microRNA precursor includes an artificial microRNA precursor.
Figure JPOXMLDOC01-appb-T000003
Figure JPOXMLDOC01-appb-T000003
Figure JPOXMLDOC01-appb-T000004
Figure JPOXMLDOC01-appb-T000004
Figure JPOXMLDOC01-appb-T000005
Figure JPOXMLDOC01-appb-T000005
Figure JPOXMLDOC01-appb-T000006
Figure JPOXMLDOC01-appb-T000006
Figure JPOXMLDOC01-appb-T000007
Figure JPOXMLDOC01-appb-T000007
Figure JPOXMLDOC01-appb-T000008
Figure JPOXMLDOC01-appb-T000008
Figure JPOXMLDOC01-appb-T000009
Figure JPOXMLDOC01-appb-T000009
Figure JPOXMLDOC01-appb-T000010
Figure JPOXMLDOC01-appb-T000010
Figure JPOXMLDOC01-appb-T000011
Figure JPOXMLDOC01-appb-T000011
Figure JPOXMLDOC01-appb-T000012
Figure JPOXMLDOC01-appb-T000012
Figure JPOXMLDOC01-appb-T000013
Figure JPOXMLDOC01-appb-T000013
Figure JPOXMLDOC01-appb-T000014
Figure JPOXMLDOC01-appb-T000014
 本発明において、配列番号1~1281のいずれかで表される塩基配列と90%以上の同一性を有する核酸とは、BLAST〔J. Mol. Biol., 215, 403 (1990)〕やFASTA〔Methods in Enzymology, 183, 63 (1990)〕等の解析ソフトを用いて計算したときに、配列番号1~1281のいずれかで表される塩基配列からなる核酸と少なくとも90%以上、好ましくは93%以上、より好ましくは95%以上、さらに好ましくは96%以上、特に好ましくは97%以上、最も好ましくは98%以上の同一性を有する核酸であることを意味する。 In the present invention, a nucleic acid having 90% or more identity with the nucleotide sequence represented by any of SEQ ID NOs: 1-1811 is BLAST [J. Mol. Biol., 215 , 403 (1990)] or FASTA [ Methods in Enzymology, 183 , 63 (1990)], etc., and at least 90% or more, preferably 93% of the nucleic acid consisting of the base sequence represented by any of SEQ ID NOs: 1-1811 It means that the nucleic acid has the identity of 95% or more, more preferably 96% or more, particularly preferably 97% or more, and most preferably 98% or more.
 また上記においてストリンジェントな条件としては、例えば、一方の鎖をブロットしたメンブレンに対し、7.5 mL、1M Na2HPO4(pH7.2) 0.6 mL、10% SDS 21 mL、50xDenhardt's solution 0.6 mL、10 mg/mL sonicated salmon sperm DNA 0.3 mLから成るHybridization bufferに、32P-ATPで標識した他方の鎖を加え、50℃で一晩反応させた後、50℃で10分間、5xSSC/5%SDS液で洗浄し、更に50℃で10分間、1xSSC/1%SDS液で洗浄し、その後メンブレンを取り出し、X線フィルムに感光させることによりシグナルを検出できる条件である。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。 The stringent conditions in the above are, for example, 7.5 mL, 1 M Na 2 HPO 4 (pH 7.2) 0.6 mL, 10% SDS 21 mL, 50x Denhardt's solution 0.6 mL, 10 Add the other strand labeled with 32 P-ATP to the Hybridization buffer consisting of 0.3 mL of mg / mL sonicated salmon sperm DNA, react at 50 ° C overnight, then react at 50 ° C for 10 minutes, 5x SSC / 5% SDS solution And then washed with 1 × SSC / 1% SDS solution at 50 ° C. for 10 minutes, and then the membrane is taken out and exposed to an X-ray film to detect the signal. Those skilled in the art can appropriately select the hybridization conditions that give the same stringency.
 核酸を用いてマイクロRNAなどの核酸の発現を検出する方法としては、検体中のマイクロRNAまたはマイクロRNA前駆体などの核酸の存在が検出できる方法であれば、いかなる方法でもよく、例えば、(1)ノーザンハイブリダイゼーション、(2)ドットブロットハイブリダイゼーション、(3)in situハイブリダイゼーション、(4)定量的PCR、(5)ディファレンシャル・ハイブリダイゼーション、(6)マイクロアレイ、(7)リボヌクレアーゼ保護アッセイ等があげられる。 As a method for detecting the expression of a nucleic acid such as microRNA using a nucleic acid, any method can be used as long as it can detect the presence of a nucleic acid such as microRNA or a microRNA precursor in a sample. For example, (1 ) Northern hybridization, (2) dot blot hybridization, (3) in situ hybridization, (4) quantitative PCR, (5) differential hybridization, (6) microarray, (7) ribonuclease protection assay It is done.
 核酸を用いてマイクロRNAなどの核酸の変異を検出する方法としては、検体中のマイクロRNAまたはマイクロRNA前駆体などの核酸の塩基配列の変異が検出できる方法であればいかなる方法でもよく、例えば、非変異型塩基配列を有する核酸と変異型塩基配列を有する核酸とのハイブリダイズにより形成されるヘテロ二本鎖を検出する方法や、あるいは、検体由来の塩基配列を直接配列決定して、変異の有無を検出する方法等をあげることができる。 As a method for detecting a mutation in a nucleic acid such as microRNA using a nucleic acid, any method can be used as long as it can detect a mutation in the base sequence of a nucleic acid such as microRNA or a microRNA precursor in a sample. A method for detecting a heteroduplex formed by hybridization of a nucleic acid having a non-mutated base sequence and a nucleic acid having a mutant base sequence, or by directly sequencing a base sequence derived from a specimen, A method for detecting the presence or absence can be given.
 核酸を発現するベクターとしては、細胞内に導入して転写されることによりマイクロRNAなどの核酸が生合成されるように設計されたベクターであればいかなるものであってもよい。細胞内でマイクロRNAなどの核酸を発現することができるベクターとしては、具体的には、pCDNA6.2-GW/miR(Invitrogen社製)、pSilencer4.1-CMV(Ambion社製)、pSINsi-hH1 DNA(タカラバイオ社製)、pSINsi-hU6 DNA(タカラバイオ社製)、pENTR/U6(Invitrogen社製)、microRNA Archive(タカラバイオ社製)、pMIRRNA1(システムバイオサイエンス社製)等をあげることができる。 The vector expressing nucleic acid may be any vector as long as it is designed to biosynthesize nucleic acid such as microRNA by being introduced into a cell and transcribed. Specific examples of vectors capable of expressing nucleic acids such as microRNA in cells include pCDNA6.2-GW / miR (Invitrogen), pSilencer4.1-CMV (Ambion), pSINsi-hH1 DNA (Takara Bio), pSINsi-hU6UDNA (Takara Bio), pENTR / U6 (Invitrogen), microRNA Archive (Takara Bio), pMIRRNA1 (System Bioscience), etc. it can.
 マイクロRNAなどの核酸の標的塩基配列を有する遺伝子(以下、標的遺伝子という)の発現を抑制する方法としては、マイクロRNAなどの核酸の標的塩基配列を有するmRNAの発現を抑制する活性を利用して、該標的塩基配列を有する遺伝子の発現を抑制する方法であれば、いかなる方法でもよい。なお、ここで発現を抑制するとは、mRNAの翻訳を抑制させる場合、およびmRNAを切断あるいは分解することによって、結果としてmRNAから翻訳される蛋白質の量を減少させる場合も含まれる。該標的塩基配列を有するmRNAの発現を抑制する物質としては、具体的にはsiRNAやアンチセンスオリゴヌクレオチドなどの核酸があげられる。該siRNAは、該mRNAの連続した配列情報を基に作製することができる[Genes Dev., 13, 3191 (1999)]。siRNAの一方の鎖を構成する塩基の残基数は、好ましくは17~30残基、より好ましくは18~25残基、さらに好ましくは19~23残基である。 As a method for suppressing the expression of a gene having a target base sequence of a nucleic acid such as microRNA (hereinafter referred to as a target gene), the activity of suppressing the expression of mRNA having a target base sequence of a nucleic acid such as microRNA is utilized. Any method may be used as long as it suppresses the expression of a gene having the target base sequence. In addition, suppression of expression here includes a case where the translation of mRNA is suppressed, and a case where the amount of protein translated from mRNA is reduced by cleaving or decomposing mRNA. Specific examples of substances that suppress the expression of mRNA having the target base sequence include nucleic acids such as siRNA and antisense oligonucleotides. The siRNA can be prepared based on the continuous sequence information of the mRNA [Genes Dev., 13 , 3191 (1999)]. The number of residues of the base constituting one strand of siRNA is preferably 17 to 30 residues, more preferably 18 to 25 residues, still more preferably 19 to 23 residues.
 マイクロRNAとしては、標的遺伝子のmRNAの3’非翻訳領域に存在する連続する7塩基の配列と相補する配列を2~8番目の塩基配列として含む17~28塩基長からなる一本鎖RNAである人工マイクロRNAも含まれる。配列を含みそれを前後に延長させた配列がヘアピン構造を形成し、該マイクロRNA配列が細胞内でそのヘアピン構造のいずれか片鎖からマイクロRNAの生合成経路によって切り出され得るRNAである場合、その延長された配列を人工マイクロRNA前駆体と呼ぶ。発現抑制したい遺伝子を標的遺伝子として、上記のような方法を用いて人工マイクロRNAおよび人工マイクロRNA前駆体を設計することができる。 The microRNA is a single-stranded RNA having a length of 17 to 28 bases containing a sequence complementary to a continuous 7 base sequence existing in the 3 ′ untranslated region of the mRNA of the target gene as the second to eighth base sequences. Some artificial microRNAs are also included. When the sequence including the sequence and extending it back and forth forms a hairpin structure, and the microRNA sequence is RNA that can be excised from any one strand of the hairpin structure in the cell by the microRNA biosynthetic pathway, The extended sequence is called an artificial microRNA precursor. Artificial microRNAs and artificial microRNA precursors can be designed using the method as described above using a gene whose expression is to be suppressed as a target gene.
 マイクロRNAなどの核酸の標的塩基配列とは、本発明におけるマイクロRNAなどの核酸によって認識される数塩基からなる塩基配列で、かつ、該塩基配列を有するmRNAの発現が本発明におけるマイクロRNAなどの核酸により抑制される塩基配列をいう。マイクロRNAの5’末端側2~8番目の塩基配列に対して相補的な塩基配列は、該塩基配列を有するmRNAが該マイクロRNAによって翻訳が抑制されるので[Current Biology, 15, R458-R460 (2005)]、本発明におけるマイクロRNAなどの核酸の5’末端側2~8番目の塩基配列に対して相補的な塩基配列を標的塩基配列としてあげることができる。例えば、マイクロRNAの5’末端側2~8番目の塩基配列に対して相補的な標的配列を用意し、ヒトmRNAの3'UTR塩基配列群に対して、完全に一致する配列を含有するmRNAを、文字列探索などの方法で選抜することで決定することができる。ヒトmRNAの3'UTR塩基配列群は、「UCSC Human Genome Browser Gateway(http://genome.ucsc.edu/cgi-bin/hgGateway)」より取得できるゲノム配列および遺伝子位置情報を用いて作製することができる。配列番号1~27のいずれかで表される塩基配列からなるマイクロRNAの標的塩基配列を有した遺伝子の具体的な例としては、米国国立バイオテクノロジー情報センターNational Center for Biotechnology Information(NCBI)のEntreGeneデータベース(http://www.ncbi.nlm.nih.gov/Entrez/)で使われている名前(Official SymbolとGene ID)で表記された表4に記載の遺伝子をあげることができる。 The target base sequence of nucleic acid such as microRNA is a base sequence consisting of several bases recognized by nucleic acid such as microRNA in the present invention, and the expression of mRNA having the base sequence is such as microRNA in the present invention. A base sequence that is suppressed by a nucleic acid. The base sequence complementary to the 2-8th base sequence on the 5 ′ end side of the microRNA is that translation of mRNA having the base sequence is suppressed by the microRNA [Current Biology, 15 , R458-R460]. (2005)], a base sequence complementary to the second to eighth base sequences on the 5 ′ end side of nucleic acids such as microRNA in the present invention can be mentioned as the target base sequence. For example, an mRNA containing a sequence that perfectly matches the 3 'UTR base sequence group of human mRNAs by preparing a target sequence complementary to the 2-8th base sequence on the 5' end side of the microRNA. Can be determined by selecting by a method such as a character string search. The 3'UTR base sequence group of human mRNA should be prepared using the genome sequence and gene position information that can be obtained from "UCSC Human Genome Browser Gateway (http://genome.ucsc.edu/cgi-bin/hgGateway)". Can do. As a specific example of a gene having a target base sequence of microRNA consisting of the base sequence represented by any one of SEQ ID NOs: 1 to 27, EntreGene of National Center for Biotechnology Information (NCBI) Examples of the genes listed in Table 4 are the names (Official Symbol and Gene ID) used in the database (http://www.ncbi.nlm.nih.gov/Entrez/).
Figure JPOXMLDOC01-appb-T000015
Figure JPOXMLDOC01-appb-T000015
Figure JPOXMLDOC01-appb-T000016
Figure JPOXMLDOC01-appb-T000016
Figure JPOXMLDOC01-appb-T000017
Figure JPOXMLDOC01-appb-T000017
Figure JPOXMLDOC01-appb-T000018
Figure JPOXMLDOC01-appb-T000018
Figure JPOXMLDOC01-appb-T000019
Figure JPOXMLDOC01-appb-T000019
Figure JPOXMLDOC01-appb-T000020
Figure JPOXMLDOC01-appb-T000020
Figure JPOXMLDOC01-appb-T000021
Figure JPOXMLDOC01-appb-T000021
Figure JPOXMLDOC01-appb-T000022
Figure JPOXMLDOC01-appb-T000022
Figure JPOXMLDOC01-appb-T000023
Figure JPOXMLDOC01-appb-T000023
Figure JPOXMLDOC01-appb-T000024
Figure JPOXMLDOC01-appb-T000024
Figure JPOXMLDOC01-appb-T000025
Figure JPOXMLDOC01-appb-T000025
Figure JPOXMLDOC01-appb-T000026
Figure JPOXMLDOC01-appb-T000026
Figure JPOXMLDOC01-appb-T000027
Figure JPOXMLDOC01-appb-T000027
Figure JPOXMLDOC01-appb-T000028
Figure JPOXMLDOC01-appb-T000028
Figure JPOXMLDOC01-appb-T000029
Figure JPOXMLDOC01-appb-T000029
Figure JPOXMLDOC01-appb-T000030
Figure JPOXMLDOC01-appb-T000030
Figure JPOXMLDOC01-appb-T000031
Figure JPOXMLDOC01-appb-T000031
Figure JPOXMLDOC01-appb-T000032
Figure JPOXMLDOC01-appb-T000032
Figure JPOXMLDOC01-appb-T000033
Figure JPOXMLDOC01-appb-T000033
Figure JPOXMLDOC01-appb-T000034
Figure JPOXMLDOC01-appb-T000034
Figure JPOXMLDOC01-appb-T000035
Figure JPOXMLDOC01-appb-T000035
Figure JPOXMLDOC01-appb-T000036
Figure JPOXMLDOC01-appb-T000036
 本発明においてマイクロRNAなどの核酸を細胞で発現させる方法としては、細胞内に導入された際にマイクロRNAなどをコードする遺伝子が発現する核酸を用いる方法があげられる。該核酸としてはDNAやRNA、あるいはヌクレオチド類似体の他、これらのキメラ分子、あるいは該核酸の誘導体も用いることができる。具体的には、Pre-miRTM miRNA Precursor Molecules(Ambion社製)やmiRIDIAN microRNA Mimics(GEヘルスケア社製)と同様に該核酸を設計し、該マイクロRNAなどの核酸を細胞内で発現させることができる。マイクロRNAを発現させる場合は、細胞内で最終的にマイクロRNAができる状態になればいかなる方法でもよく、例えば、(1)マイクロRNA前駆体である一本鎖RNAを導入させる他、(2)マイクロRNAそのもの、およびマイクロRNAの相補鎖からなり、100%マッチの2本鎖からなるRNA、(3)マイクロRNAがDicerに切断された後の状態を想定した2本鎖RNAを導入させる方法があげられる。こうした方法を使用した製品としては、それぞれ例えば、miCENTURY OX Precursor(B-Bridge社製)、miCENTURY OX siMature(B-Bridge社製)、miCENTURY OX miNatural(B-Bridge社製)をあげることができる。 In the present invention, a method for expressing a nucleic acid such as microRNA in a cell includes a method using a nucleic acid that expresses a gene encoding the microRNA when introduced into the cell. As the nucleic acid, in addition to DNA, RNA, or nucleotide analogues, these chimeric molecules or derivatives of the nucleic acids can also be used. Specifically, the nucleic acid can be designed in the same manner as Pre-miRTMmimiRNA Precursor Molecules (Ambion) or miRIDIAN microRNA Mimics (GE Healthcare) to express nucleic acids such as microRNA in cells. it can. When microRNA is expressed, any method may be used as long as microRNA can finally be produced in the cell. For example, (1) In addition to introducing single-stranded RNA as a microRNA precursor, (2) There is a method for introducing microRNA itself and RNA consisting of complementary strands of microRNA and 100% -matched double-stranded RNA, and (3) double-stranded RNA assuming the state after microRNA is cleaved into Dicer. can give. Examples of products using such a method include miCENTURY OX Precursor (manufactured by B-Bridge), miCENTURY OX siMature (manufactured by B-Bridge), and miCENTURY OX miNatural (manufactured by B-Bridge).
 本発明で用いる核酸を合成する方法としては、特に限定されず、公知の化学合成を用いる方法、あるいは、酵素的転写法等にて製造することができる。公知の化学合成を用いる方法として、ホスホロアミダイト法、ホスフォロチオエート法、ホスホトリエステル法等をあげることができ、例えば、ABI3900ハイスループット核酸合成機(アプライドバイオシステムズ社製)により合成することができる。酵素的転写法としては、目的の塩基配列を有したプラスミドまたはDNAを鋳型として典型的なファージRNAポリメラーゼ、例えば、T7、T3、またはSP6 RNAポリメラーゼを用いた転写をあげることができる。 The method for synthesizing the nucleic acid used in the present invention is not particularly limited, and it can be produced by a method using a known chemical synthesis, an enzymatic transcription method or the like. Examples of methods using known chemical synthesis include phosphoramidite method, phosphorothioate method, phosphotriester method, etc. For example, synthesis with ABI3900 high-throughput nucleic acid synthesizer (Applied Biosystems) Can do. Examples of the enzymatic transcription method include transcription using a typical phage RNA polymerase, for example, T7, T3, or SP6 RNA polymerase, using a plasmid or DNA having the target base sequence as a template.
 本発明で用いる核酸により、該核酸の発現または機能を促進あるいは抑制させる物質をスクリーニングする方法としては、例えば、該核酸を発現するベクターを細胞に導入し、それらの標的塩基配列を有するmRNAの発現または機能を促進または抑制する物質をスクリーニングする方法があげられる。 Examples of a method for screening for a substance that promotes or suppresses the expression or function of the nucleic acid using the nucleic acid used in the present invention include, for example, introducing a vector that expresses the nucleic acid into a cell, and expressing an mRNA having the target base sequence. Alternatively, a method of screening for a substance that promotes or suppresses the function can be mentioned.
 本発明で用いる核酸を有効成分とする医薬は、細胞または臓器の線維化や組織の過形成等が原因となっている疾患の診断または治療に用いることができる。また核酸は、細胞または臓器の線維化制御剤として用いることもできる。ここでいう細胞または臓器の線維化とは、生体内において細胞外基質の産生が異常に亢進している状態、あるいは上皮間葉転換(EMT:Epithelial Mesenchymal Transition)が起きている状態をいい、制御剤とは抑制剤もしくは促進剤のことを指す。なお、本発明において、「細胞または臓器の線維化」とは、細胞又は臓器のいずれか一方のみの線維化に限定されず、細胞及び臓器が共に線維化されている場合を含む。 The pharmaceutical comprising the nucleic acid used in the present invention as an active ingredient can be used for diagnosis or treatment of diseases caused by fibrosis of cells or organs, tissue hyperplasia, or the like. The nucleic acid can also be used as a fibrosis control agent for cells or organs. Cell or organ fibrosis as used herein refers to a state in which extracellular matrix production is abnormally increased in the living body or a state in which epithelial-mesenchymal transition (EMT) occurs. An agent refers to an inhibitor or an accelerator. In the present invention, “fibrosis of cells or organs” is not limited to fibrosis of only one of cells or organs, but includes cases where both cells and organs are fibrotic.
 細胞または臓器の線維化や組織の過形成等が原因となっている疾患としては、例えば、繊維性の組織の障害に伴う繊維症等があげられる。ここで、繊維性の組織とは、結合組織を有する組織であれば特に限定されず、例えば、腎臓、肺、肝臓、心臓、膵臓、脾臓、胃、脊髄、下垂体、生殖腺、甲状腺、胆嚢、骨髄、副腎、皮膚、消化管(例、大腸、小腸)、脳、脳の各部位(例、嗅球、扁桃核、大脳基底球、海馬、視床、視床下部、大脳皮質、延髄、小脳)、膀胱、睾丸、卵巣、胎盤、子宮、骨、関節、皮膚、血管、脂肪組織、骨格筋等があげられる。上記疾患の具体例としては、以下に限定されないが、急性腎不全、糸球体腎炎、血管炎、糖尿病性腎症、高血圧性腎硬化症、HIV腎症、IgA腎症、ループス腎炎、間質性腎炎、尿管閉塞による閉塞腎等の腎障害に伴う線維症や、肺線維症、肝硬変、動脈硬化症、強皮症、経皮経管冠動脈血管拡張術(PTCA)後の冠動脈再狭窄、間質性心筋炎、間質性膀胱炎、熱傷後の皮膚瘢痕化、中毒等に伴う線維症等をあげることができる。 Examples of diseases caused by cell or organ fibrosis or tissue hyperplasia include, for example, fibrosis associated with damage to fibrous tissue. Here, the fibrous tissue is not particularly limited as long as it has a connective tissue, for example, kidney, lung, liver, heart, pancreas, spleen, stomach, spinal cord, pituitary gland, gonad, thyroid, gallbladder, Bone marrow, adrenal gland, skin, digestive tract (eg, large intestine, small intestine), brain, brain parts (eg, olfactory bulb, amygdala, basal sphere, hippocampus, thalamus, hypothalamus, cerebral cortex, medulla, cerebellum), bladder , Testis, ovary, placenta, uterus, bone, joint, skin, blood vessel, adipose tissue, skeletal muscle and the like. Specific examples of the disease include, but are not limited to, acute renal failure, glomerulonephritis, vasculitis, diabetic nephropathy, hypertensive nephrosclerosis, HIV nephropathy, IgA nephropathy, lupus nephritis, interstitial Nephritis, fibrosis associated with renal disorders such as obstructed kidney due to ureteral obstruction, pulmonary fibrosis, cirrhosis, arteriosclerosis, scleroderma, coronary restenosis after percutaneous transluminal coronary vasodilation (PTCA), between Examples include interstitial myocarditis, interstitial cystitis, skin scarring after burn injury, and fibrosis associated with poisoning.
 以下、本発明を詳細に説明する。
1.線維化した細胞で発現する、マイクロRNAおよびマイクロRNA前駆体などの核酸の発現を検出する方法
(1-1)マイクロRNAの同定
 低分子RNA配列がマイクロRNAであるかは、アール エヌ エイ(RNA), 9, 277-279 (2003)に記載の基準に従うか否かで判定できる。例えば、新たに取得して塩基配列を決定した低分子RNAの場合、以下のようにして行うことができる。
Hereinafter, the present invention will be described in detail.
1. Method for detecting expression of nucleic acid such as microRNA and microRNA precursor expressed in fibrotic cells (1-1) Identification of microRNA Whether a small RNA sequence is microRNA is determined by RNA (RNA) ), 9 , 277-279 (2003). For example, in the case of a low molecular weight RNA that has been newly acquired and whose base sequence has been determined, it can be performed as follows.
 取得した低分子RNA塩基配列に対応するDNA配列を5’末端側および3’末端側にそれぞれ50 nt程度伸ばした周辺ゲノム配列を取得し、そのゲノム配列から転写されることが予測されるRNAの2次構造を予測する。その結果、ヘアピン構造を有し、かつ該低分子RNAの塩基配列がヘアピンの片鎖に位置する場合、該低分子RNAはマイクロRNAであると判定できる。ゲノム配列は一般に公開されており、例えば、UCSC Genome Bioinformatics (http://genome.ucsc.edu/) から入手可能である。また、2次構造予測も様々なプログラムが公開されており、例えば、RNAfold [Nucleic Acids Research, 31, 3429-3431 (2003)]やMfold [Nucleic Acids Research, 31, 3406-3415 (2003)] 等を用いることができる。また、既存のマイクロRNA配列は英国サンガーセンターのmiRBaseというデータベースに登録されており、ここに記載の配列と同一か否かで、既存のマイクロRNAと同一か否かを判定することができる。
(1-2)低分子RNAの配列情報の取得
 細胞株、または臓器の線維化が見られる患者由来検体から全RNAを抽出し、該RNAを用い線維化細胞または線維化組織で発現するマイクロRNAを含有する低分子RNAを以下のようにして取得することができる。
A peripheral genomic sequence obtained by extending the DNA sequence corresponding to the obtained low molecular RNA base sequence by about 50 nt to the 5 'end side and 3' end side, respectively, is obtained, and RNA predicted to be transcribed from the genomic sequence Predict secondary structure. As a result, when it has a hairpin structure and the base sequence of the small RNA is located in one strand of the hairpin, it can be determined that the small RNA is a microRNA. Genomic sequences are publicly available and are available, for example, from UCSC Genome Bioinformatics (http://genome.ucsc.edu/). In addition, various programs for secondary structure prediction have been published, such as RNAfold [Nucleic Acids Research, 31 , 3429-3431 (2003)], Mfold [Nucleic Acids Research, 31 , 3406-3415 (2003)], etc. Can be used. Further, existing microRNA sequences are registered in a database called miRBase at Sanger Center in the UK, and it can be determined whether or not they are identical to existing microRNAs based on whether or not they are identical to the sequences described here.
(1-2) Acquisition of sequence information of low molecular weight RNA Extracting total RNA from a cell line or a specimen derived from a patient in which organ fibrosis is observed, and using the RNA, microRNA expressed in fibrotic cells or fibrotic tissue Small molecule RNA containing can be obtained as follows.
 低分子RNAの取得法としては、具体的には、ジーンズ アンド デベロップメント(Genes & Development), 15, 188-200 (2000)に記載の方法に準じて、15%ポリアクリルアミドゲル電気泳動により低分子RNAを分離する方法があげられる。これより5’末端脱リン酸化、3’-アダプターライゲーション、リン酸化、5’-アダプターライゲーション、逆転写、PCR増幅、コンカテマー化、ベクターへのライゲーションを順次経て、低分子RNAをクローニングし、そのクローンの塩基配列を決定することができる。あるいは、例えばサイエンス(Science), 294, 858-862 (2001)に記載の方法に準じて、5’-アデニル化、3’-アダプターライゲーション、5’-アダプターライゲーション、逆転写、PCR増幅、コンカテマー化、ベクターへのライゲーションを順次経て、低分子RNAをクローニングし、そのクローンの塩基配列を決定することもできる。 As a method for obtaining low molecular weight RNA, specifically, low molecular weight RNA can be obtained by 15% polyacrylamide gel electrophoresis in accordance with the method described in Jeans & Development (Genes & Development), 15 , 188-200 (2000). The method of separating is mentioned. From this, 5'-terminal dephosphorylation, 3'-adapter ligation, phosphorylation, 5'-adapter ligation, reverse transcription, PCR amplification, concatamerization, and ligation to vector are sequentially cloned, and the clone is cloned. Can be determined. Alternatively, for example, according to the method described in Science, 294 , 858-862 (2001), 5′-adenylation, 3′-adapter ligation, 5′-adapter ligation, reverse transcription, PCR amplification, concatamerization In addition, after sequentially ligating to a vector, low molecular RNA can be cloned and the base sequence of the clone can be determined.
 また、Nucleic Acids Research, 34, 1765-1771 (2006)に記載の方法により、5’末端脱リン酸化、3’-アダプターライゲーション、リン酸化、5’-アダプターライゲーション、逆転写、PCR増幅、マイクロビーズベクターへのライゲーションを順次経て、低分子RNAをクローニングし、そのマイクロビーズの塩基配列を読み取ることで塩基配列を決定することにより低分子RNAの塩基配列情報を取得することができる。 In addition, according to the method described in Nucleic Acids Research, 34 , 1765-1771 (2006), 5 ′ end dephosphorylation, 3′-adapter ligation, phosphorylation, 5′-adapter ligation, reverse transcription, PCR amplification, microbeads By sequentially ligating to a vector, low molecular RNA is cloned, and the base sequence information of the low molecular RNA can be obtained by determining the base sequence by reading the base sequence of the microbead.
 低分子RNAを取得するための他の方法として、small RNA Cloning Kit(タカラバイオ社製)を用いた方法があげられる。
(1-3)マイクロRNAなどの核酸の発現量の検出法
 マイクロRNAおよびその前駆体などの核酸の発現量を検出する方法としては、例えば、(1)ノーザンハイブリダイゼーション、(2)ドットブロットハイブリダイゼーション、(3)in situハイブリダイゼーション、(4)定量的PCR、(5)ディファレンシャル・ハイブリダイゼーション、(6)マイクロアレイ、(7)リボヌクレアーゼ保護アッセイなどがあげられる。
As another method for obtaining low molecular weight RNA, there is a method using a small RNA Cloning Kit (manufactured by Takara Bio Inc.).
(1-3) Method for detecting expression level of nucleic acid such as microRNA Examples of methods for detecting the expression level of nucleic acid such as microRNA and its precursor include (1) Northern hybridization and (2) dot blot high. Hybridization, (3) in situ hybridization, (4) quantitative PCR, (5) differential hybridization, (6) microarray, (7) ribonuclease protection assay.
 ノーザンブロット法とは、検体由来RNAをゲル電気泳動で分離後、ナイロンフィルター等の支持体に転写し、核酸の塩基配列をもとに適宜標識をしたプローブを作製し、ハイブリダイゼーションおよび洗浄をおこなうことで、核酸に特異的に結合したバンドを検出する方法であり、具体的には、例えば、Science, 294, 853-858 (2001)に記載の方法等に従って行うことができる。 Northern blotting is a method in which sample-derived RNA is separated by gel electrophoresis, then transferred to a support such as a nylon filter, a probe labeled appropriately based on the base sequence of the nucleic acid is prepared, and hybridization and washing are performed. Thus, this is a method for detecting a band specifically bound to a nucleic acid. Specifically, for example, it can be performed according to the method described in Science, 294 , 853-858 (2001).
 標識プローブは、例えば、ニック・トランスレーション、ランダム・プライミングまたは5'末端のリン酸化等の方法により放射性同位体、ビオチン、ジゴキシゲニン、蛍光基、化学発光基等を、本発明で用いる核酸の塩基配列と相補的な配列を有するDNAやRNA、あるいはLNAに取り込ませることで調製できる。標識プローブの結合量は該核酸の発現量を反映することから、結合した標識プローブの量を定量することで該核酸の発現量を定量することができる。電気泳動、メンブレンへの移行、プローブの調製、ハイブリダイゼーション、核酸の検出については、モレキュラー・クローニング第3版[Cold Spring Harbor Press, (2001) Cold Spring Harbor, NY]に記載の方法により行うことができる。 The labeled probe is a base sequence of a nucleic acid used in the present invention, for example, a radioisotope, biotin, digoxigenin, a fluorescent group, a chemiluminescent group, etc. by a method such as nick translation, random priming or phosphorylation at the 5 ′ end. It can be prepared by incorporating it into DNA or RNA having a complementary sequence to LNA or LNA. Since the binding amount of the labeled probe reflects the expression level of the nucleic acid, the expression level of the nucleic acid can be quantified by quantifying the amount of bound labeled probe. Electrophoresis, transfer to membrane, probe preparation, hybridization, and nucleic acid detection can be performed by the method described in Molecular Cloning 3rd Edition [Cold Spring Harbor Press, (2001) Cold Spring Harbor, NY] it can.
 ドットブロットハイブリダイゼーションは、組織や細胞から抽出したRNAを膜上に点状にスポットして固定し、プローブとなる標識したポリヌクレオチドとハイブリダイゼーションを行い、プローブと特異的にハイブリダイズするRNAを検出する方法である。プローブとしてはノーザンハイブリダイゼーションと同様のものを用いることができる。RNAの調製、RNAのスポット、ハイブリダイゼーション、RNAの検出については、モレキュラー・クローニング第3版に記載の方法により行なうことができる。 In dot blot hybridization, RNA extracted from tissues and cells is spot-fixed on a membrane in a dotted manner, and then hybridized with a labeled polynucleotide that serves as a probe to detect RNA that specifically hybridizes with the probe. It is a method to do. As the probe, the same probe as in Northern hybridization can be used. Preparation of RNA, RNA spot, hybridization, and detection of RNA can be performed by the methods described in Molecular Cloning 3rd edition.
 in situハイブリダイゼーションは、生体から取得した組織のパラフィンまたはクリオスタット切片、あるいは固定化した細胞を検体として用い、標識したプローブとのハイブリダイゼーションおよび洗浄の工程を行い、顕微鏡観察により、核酸の組織や細胞内での分布や局在を調べる方法である[Methods in Enzymology, 254, 419 (1995)]。プローブとしてはノーザンハイブリダイゼーションと同様のものを用いることができる。具体的には、Nature Method, 3, 27 (2006)に記載の方法に従って、マイクロRNAを検出することができる。 In situ hybridization uses a paraffin or cryostat section of tissue obtained from a living body or immobilized cells as a specimen, performs hybridization and washing steps with a labeled probe, and observes the tissue and nucleic acid of a nucleic acid by microscopic observation. It is a method for examining distribution and localization in cells [Methods in Enzymology, 254 , 419 (1995)]. As the probe, the same probe as in Northern hybridization can be used. Specifically, microRNA can be detected according to the method described in Nature Method, 3 , 27 (2006).
 定量的PCRでは、検体由来RNAから、逆転写用プライマーと逆転写酵素を用いて合成したcDNA(以後、該cDNAを検体由来cDNAと称する)を測定に用いる。cDNA合成に供する逆転写用プライマーとして、ランダムプライマーあるいは特異的RTプライマー等を用いることができる。特異的RTプライマーとは、核酸およびその周辺ゲノム配列に対応する塩基配列に相補する配列を有するプライマーをいう。 In quantitative PCR, cDNA synthesized from a sample-derived RNA using a reverse transcription primer and a reverse transcriptase (hereinafter, the cDNA is referred to as a sample-derived cDNA) is used for measurement. As a reverse transcription primer used for cDNA synthesis, a random primer or a specific RT primer can be used. The specific RT primer refers to a primer having a sequence complementary to a nucleic acid and a base sequence corresponding to the surrounding genomic sequence.
 例えば、検体由来cDNAを合成後、これを鋳型とし、マイクロRNAやマイクロRNA前駆体などの核酸およびその周辺ゲノム配列に対応する塩基配列、あるいは逆転写用プライマーに対応する塩基配列から設計した鋳型特異的なプライマーを用いてPCRを行い、該核酸を含むcDNAの断片を増幅させ、ある一定量に達するまでのサイクル数から検体由来RNAに含まれる該核酸の量を検出する。鋳型特異的なプライマーとしては、核酸およびその周辺ゲノム配列に対応する適当な領域を選択し、その領域の塩基配列の5'端15~40残基、好ましくは20~30残基の配列からなるDNAまたはLNA、および3'端15~40残基、好ましくは20~30残基と相補的な配列からなるDNAまたはLNAの組を用いることができる。具体的には、Nucleic Acids Research, 32, e43 (2004)に記載の方法等に準じて行うことができる。 For example, after synthesizing a specimen-derived cDNA, using this as a template, a template-specific design designed from a nucleotide sequence corresponding to a nucleic acid such as microRNA or a microRNA precursor and its surrounding genomic sequence, or a nucleotide sequence corresponding to a reverse transcription primer PCR is performed using a typical primer, a cDNA fragment containing the nucleic acid is amplified, and the amount of the nucleic acid contained in the sample-derived RNA is detected from the number of cycles until a certain amount is reached. As a template-specific primer, an appropriate region corresponding to the nucleic acid and the surrounding genomic sequence is selected, and consists of a sequence of 15 to 40 residues, preferably 20 to 30 residues at the 5 ′ end of the base sequence of the region. A set of DNA or LNA consisting of DNA or LNA and a sequence complementary to 15 to 40 residues, preferably 20 to 30 residues at the 3 ′ end can be used. Specifically, it can be performed according to the method described in Nucleic Acids Research, 32 , e43 (2004).
 また、cDNA合成に供する逆転写用プライマーとして、ステム・ループ構造を有した特異的RTプライマーを用いることもできる。具体的には、Nucleic Acid Research, 33, e179 (2005)に記載の方法、あるいは、TaqMan MicroRNA Assays(アプライドバイオシステムズ社製)を用いて行うことができる。 A specific RT primer having a stem-loop structure can also be used as a reverse transcription primer for cDNA synthesis. Specifically, the method described in Nucleic Acid Research, 33, e179 ( 2005) or can be performed using the TaqMan MicroRNA Assays (Applied Biosystems).
 更に別の検体由来cDNA合成法として、検体由来RNAに対してpolyAポリメラーゼによりpolyA配列を付加し、オリゴdT配列を含む塩基配列を逆転写用プライマーとして用いることにより、逆転写反応を行うこともできる。具体的には、miScript System(キアゲン社製)やQuantiMir RT Kit(System Biosciences社製)、PrimeScript 1st strand cDNA Synthesis Kit(タカラバイオ社製)を用いて行うことができる。更に、核酸を少なくとも1つ以上含む塩基配列に対応するDNAあるいはLNAを固定化させたフィルターあるいはスライドガラスやシリコンなどの基板に対して、検体由来RNAあるいはcDNAをハイブリダイゼーションし、洗浄を行うことにより、核酸の量の変動を検出することができる。 As yet another sample-derived cDNA synthesis method, a reverse transcription reaction can also be performed by adding a polyA sequence to a sample-derived RNA with polyA polymerase and using a base sequence containing an oligo dT sequence as a primer for reverse transcription. . Specifically, it can be performed using miScript System (Qiagen), QuantiMir RT Kit (System Biosciences), PrimeScript 1st strand cDNA Synthesis (Takara Bio). Furthermore, by subjecting the sample-derived RNA or cDNA to hybridization to a filter or slide glass or silicon substrate or the like to which DNA or LNA corresponding to a base sequence containing at least one nucleic acid is immobilized, and washing. The variation in the amount of nucleic acid can be detected.
 このようなハイブリダイゼーションに基づく方法には、ディファレンシャルハイブリダイゼーション[Trends Genet., 7, 314 (1991)]やマイクロアレイ[Genome Res., 6, 639 (1996)]を用いる方法があげられる。いずれの方法もフィルターあるいは基板上にU6 RNAに対応する塩基配列などの内部コントロールを固定化することで、対照検体と標的検体の間での核酸の量の違いを正確に検出することができる。また対照検体と標的検体由来のRNAをもとにそれぞれ異なる標識のdNTP(dATP、dGTP、dCTP、dTTPの混合物)を用いて標識cDNA合成を行い、1枚のフィルターあるいは1枚の基盤に2つの標識cDNAを同時にハイブリダイズさせることで正確な核酸の定量を行うことができる。更には、対照検体および/または標的検体由来のRNAを直接標識してハイブリダイズさせることで核酸の定量をおこなうこともできる。例えば、Proc. Natl. Acad. Sci. USA, 101, 9740-9744 (2004)やNucleic Acid Research, 32, e188 (2004)、RNA, 13, 151-159 (2007)等に記載のマイクロアレイを用いてマイクロRNAを検出することができる。具体的には、mirVana miRNA Bioarray(Ambion社製)、miRNA microarray kit(アジレント・テクノロジー社製)と同様にして検出または定量することができる。 Examples of the method based on such hybridization include a method using differential hybridization [Trends Genet., 7 , 314 (1991)] and a microarray [Genome Res., 6 , 639 (1996)]. In either method, the difference in the amount of nucleic acid between the control sample and the target sample can be accurately detected by immobilizing an internal control such as a base sequence corresponding to U6 RNA on a filter or substrate. In addition, labeled cDNA synthesis using differently labeled dNTPs (mixtures of dATP, dGTP, dCTP, and dTTP) based on RNA derived from the control sample and the target sample, and two filters on one filter or one substrate. Accurate nucleic acid quantification can be performed by simultaneously hybridizing labeled cDNA. Furthermore, the nucleic acid can be quantified by directly labeling and hybridizing RNA derived from the control sample and / or the target sample. For example, using a microarray described in Proc. Natl. Acad. Sci. USA, 101 , 9740-9744 (2004), Nucleic Acid Research, 32 , e188 (2004), RNA, 13 , 151-159 (2007), etc. MicroRNA can be detected. Specifically, it can be detected or quantified in the same manner as mirVana miRNA Bioarray (Ambion) and miRNA microarray kit (Agilent Technology).
 リボヌクレアーゼ保護アッセイでは、まず核酸あるいはその周辺ゲノム配列に対応する塩基配列の3'端にT7プロモーター、SP6プロモーターなどのプロモーター配列を結合し、標識したNTP(ATP、GTP、CTP、UTPの混合物)およびRNAポリメラーゼを用いたイン・ビトロの転写系により、標識したアンチセンスRNAを合成する。該標識アンチセンスRNAを、検体由来RNAと結合させて、RNA-RNAハイブリッドを形成させた後、1本鎖RNAのみを分解するリボヌクレアーゼAで消化する。該消化物をゲル電気泳動し、RNA-RNAハイブリッドを形成することにより消化から保護されたRNA断片を、核酸として、検出または定量する。具体的には、mirVana miRNA Detection Kit(Ambion社製)を用いて検出または定量することができる。 In the ribonuclease protection assay, first, a promoter sequence such as T7 promoter and SP6 promoter is bound to the 3 ′ end of the nucleotide sequence corresponding to the nucleic acid or its surrounding genomic sequence, and labeled NTP (mixture of ATP, GTP, CTP, UTP) and Labeled antisense RNA is synthesized by an in vitro transcription system using RNA polymerase. The labeled antisense RNA is bound to the sample-derived RNA to form an RNA-RNA hybrid, and then digested with ribonuclease A that degrades only single-stranded RNA. The digest is subjected to gel electrophoresis, and RNA fragments protected from digestion by forming RNA-RNA hybrids are detected or quantified as nucleic acids. Specifically, it can be detected or quantified using mirVana miRNA Detection Kit (Ambion).
2.核酸の合成
 上記1のようにして、線維化が亢進した細胞または線維化組織で発現している、あるいは正常組織と比較して発現が増大または減少しているマイクロRNAやマイクロRNA前駆体などの核酸が同定された後は、塩基配列に基づいてリボヌクレオチドの重合体であるRNAだけでなく、デオキシリボヌクレオチドの重合体であるDNAを合成することができる。例えば、上記1で同定したマイクロRNAの塩基配列をもとに、DNAの塩基配列を決定することができる。RNAの塩基配列に対応するDNAの塩基配列は、RNAの配列に含まれるU(ウラシル)をT(チミン)に読み替えることで一義的に決定できる。また、リボヌクレオチドとデオキシリボヌクレオチドが混合した重合体や、ヌクレオチド類似体を含む重合体や核酸の誘導体も同様にして合成することができる。
2. Synthesis of Nucleic Acid As described in 1 above, microRNAs or microRNA precursors that are expressed in cells or fibrotic tissues with increased fibrosis, or whose expression is increased or decreased compared to normal tissues, etc. After the nucleic acid is identified, not only RNA that is a polymer of ribonucleotides but also DNA that is a polymer of deoxyribonucleotides can be synthesized based on the base sequence. For example, the base sequence of DNA can be determined based on the base sequence of microRNA identified in 1 above. The base sequence of DNA corresponding to the base sequence of RNA can be uniquely determined by replacing U (uracil) contained in the RNA sequence with T (thymine). In addition, a polymer in which ribonucleotides and deoxyribonucleotides are mixed, a polymer containing nucleotide analogues, and a nucleic acid derivative can be synthesized in the same manner.
 核酸を合成する方法としては、特に限定されず、公知の化学合成を用いる方法、あるいは、酵素的転写法等にて製造することができる。公知の化学合成を用いる方法として、ホスホロアミダイト法、ホスフォロチオエート法、ホスホトリエステル法等をあげることができ、例えば、ABI3900ハイスループット核酸合成機(アプライドバイオシステムズ社製)により合成することができる。酵素的転写法としては、目的の塩基配列を有したプラスミドまたはDNAを鋳型として典型的なファージRNAポリメラーゼ、例えば、T7、T3、またはSP6 RNAポリメラーゼを用いた転写による方法をあげることができる。 The method for synthesizing the nucleic acid is not particularly limited, and the nucleic acid can be produced by a method using known chemical synthesis or an enzymatic transcription method. Examples of methods using known chemical synthesis include phosphoramidite method, phosphorothioate method, phosphotriester method, etc. For example, synthesis with ABI3900 high-throughput nucleic acid synthesizer (Applied Biosystems) Can do. Examples of the enzymatic transcription method include a transcription method using a typical phage RNA polymerase such as T7, T3, or SP6 RNA polymerase using a plasmid or DNA having the target base sequence as a template.
3.マイクロRNAやマイクロRNA前駆体などの核酸の機能を検出する方法
 マイクロRNAなどの核酸の機能を検出する方法としては、標的塩基配列を有するmRNAの翻訳を抑制するか否かで検出する方法をあげることができる。
3. Method for detecting the function of a nucleic acid such as microRNA or microRNA precursor As a method for detecting the function of a nucleic acid such as microRNA, there is a method for detecting whether or not the translation of mRNA having a target base sequence is suppressed. be able to.
 マイクロRNAは、その標的塩基配列を3’末端側untranslated region (3’UTR)に含むmRNAの翻訳を抑制することが知られている[Current Biology, 15, R458-R460 (2005)]。そこで測定しようとする一本鎖RNAに対する標的塩基配列を、適当なレポーター遺伝子発現ベクターの3’UTRに挿入したDNAを作製し、発現ベクターに適合した宿主細胞に導入し、その細胞に一本鎖RNAを発現させた時にレポーター遺伝子の発現を測定することで、マイクロRNAの機能を有しているか否かを検出することができる。 It is known that microRNA suppresses translation of mRNA containing the target base sequence in the 3 ′ terminal untranslated region (3′UTR) [Current Biology, 15 , R458-R460 (2005)]. Therefore, a DNA in which the target base sequence for the single-stranded RNA to be measured is inserted into the 3′UTR of an appropriate reporter gene expression vector is prepared, introduced into a host cell suitable for the expression vector, and single-stranded into the cell. By measuring the expression of a reporter gene when RNA is expressed, it can be detected whether or not it has a function of microRNA.
 レポーター遺伝子発現ベクターは、レポーター遺伝子の上流にプロモーターを有しており、宿主細胞においてレポーター遺伝子を発現できるものであればいかなるものでもよい。レポーター遺伝子としては、あらゆるレポーター遺伝子を使用することが可能であるが、例えば、ホタル・ルシフェラーゼ遺伝子、ウミシイタケ・ルシフェラーゼ遺伝子、クロラムフェニコール・アセチルトランスフェラーゼ遺伝子、β-グルクロニダーゼ遺伝子、β-ガラクトシダーゼ遺伝子、β-ラクタマーゼ遺伝子、エクオリン遺伝子、グリーン・フルオレッセント・プロテイン遺伝子およびDsRed蛍光遺伝子などが利用できる。こうした性質を有するレポーター遺伝子発現ベクターとして、例えば、psiCHECK-1(Promega社製)、psiCHECK-2(Promega社製)、pGL3-Control(Promega社製)、pGL4(Promega社製)、pRNAi-GL(タカラバイオ社製)、pCMV-DsRed-Express(CLONTECH社製)、pMIR-REPORT System(Ambion社製)などがあげられる。また、一本鎖RNAは、後述の6に記載した方法で発現させることができる。 The reporter gene expression vector may be any vector as long as it has a promoter upstream of the reporter gene and can express the reporter gene in the host cell. Any reporter gene can be used as the reporter gene, for example, firefly luciferase gene, Renilla luciferase gene, chloramphenicol acetyltransferase gene, β-glucuronidase gene, β-galactosidase gene, β -Lactamase gene, aequorin gene, green fluorescent protein gene and DsRed fluorescent gene can be used. Reporter gene expression vectors having such properties include, for example, psiCHECK-1 (Promega), psiCHECK-2 (Promega), pGL3-Control (Promega), pGL4 (Promega), pRNAi-GL ( (Takara Bio), pCMV-DsRed-Express (CLONTECH), pMIR-REPORT® System (Ambion). Single-stranded RNA can be expressed by the method described in 6 below.
 一本鎖RNAのマイクロRNAとしての機能は、具体的には以下のようにして検出することができる。まず宿主細胞をマルチウェルプレート等に培養し、標的配列を有したレポーター遺伝子発現ベクターと一本鎖RNAを発現させる。その後、レポーター活性を測定し、一本鎖RNAを発現させない場合に比べて、一本鎖RNAを発現させた場合のレポーター活性を測定することで、一本鎖RNAのマイクロRNAとしての機能を検出することができる。 The function of single-stranded RNA as a microRNA can be specifically detected as follows. First, host cells are cultured in a multi-well plate or the like to express a reporter gene expression vector having a target sequence and single-stranded RNA. Then, the reporter activity is measured, and the function of the single-stranded RNA as a microRNA is detected by measuring the reporter activity when the single-stranded RNA is expressed compared to the case where the single-stranded RNA is not expressed. can do.
4.マイクロRNAやマイクロRNA前駆体などの核酸の変異を検出する方法
 マイクロRNAやマイクロRNA前駆体などの核酸の変異を検出する方法として、正常型と変異型の核酸のハイブリダイズにより形成されるヘテロ二本鎖を検出する方法を用いることができる。
4). Method for detecting mutations in nucleic acids such as microRNA and microRNA precursors As a method for detecting mutations in nucleic acids such as microRNAs and microRNA precursors, heterogeneous nucleic acids formed by hybridization of normal and mutant nucleic acids A method for detecting this strand can be used.
 ヘテロ二本鎖を検出する方法としては、(1)ポリアクリルアミドゲル電気泳動によるヘテロ二本鎖検出法[Trends genet., 7, 5 (1991)]、(2)一本鎖コンフォメーション多型解析法[Genomics, 16, 325-332 (1993)]、(3)ミスマッチの化学的切断法(CCM, chemical cleavage of mismatches) [Human Genetics (1996), Tom Strachan and Andrew P. Read, BIOS Scientific Publishers Limited]、(4)ミスマッチの酵素的切断法[Nature Genetics, 9, 103-104 (1996)]、(5)変性ゲル電気泳動法[Mutat. Res., 288, 103-112 (1993)] 等の方法があげられる。 Methods for detecting heteroduplex include (1) heteroduplex detection by polyacrylamide gel electrophoresis [Trends genet., 7 , 5 (1991)], (2) single strand conformation polymorphism analysis [Genomics, 16 , 325-332 (1993)], (3) Chemical cleavage of mismatches (CCM) [Human Genetics (1996), Tom Strachan and Andrew P. Read, BIOS Scientific Publishers Limited ], (4) Enzymatic cleavage method of mismatch [Nature Genetics, 9 , 103-104 (1996)], (5) Denaturing gel electrophoresis [Mutat. Res., 288 , 103-112 (1993)], etc. There are methods.
 ポリアクリルアミドゲル電気泳動法によるヘテロ二本鎖検出法は、例えば、以下のようにして行う。まず、検体由来DNAあるいは検体由来cDNAをテンプレートとして、核酸の塩基配列を含むゲノムの塩基配列を基に設計したプライマーにより、200 bpよりも小さい断片として増幅する。ヘテロ二本鎖が形成された場合は、変異を持たないホモ二本鎖よりも移動度が遅く、それらは余分なバンドとして検出することができる。200 bpよりも小さい断片であれば、1塩基以上のほとんどの挿入、欠失、置換を検出することができる。ヘテロ二本鎖解析は、次に述べる一本鎖コンフォメーション解析と組み合わせた1枚のゲルで行うことが望ましい。 The heteroduplex detection method by polyacrylamide gel electrophoresis is performed as follows, for example. First, using a specimen-derived DNA or a specimen-derived cDNA as a template, a primer designed based on the genomic base sequence including the nucleic acid base sequence is amplified as a fragment smaller than 200 μbp. When heteroduplexes are formed, the mobility is slower than homoduplexes without mutations, and they can be detected as extra bands. If the fragment is smaller than 200 bp, most insertions, deletions and substitutions of 1 base or more can be detected. Heteroduplex analysis is preferably performed on a single gel combined with single-strand conformation analysis described below.
 一本鎖コンフォメーション多型解析(SSCP解析;single strand conformation polymorphism analysis)では、検体由来DNAあるいは検体由来cDNAをテンプレートにして、核酸の塩基配列を含むゲノムの塩基配列に基づき設計したプライマーを用いて、200bpよりも小さい断片として増幅したDNAを変性後に、未変性ポリアクリルアミドゲル中で泳動する。DNA増幅を行う際にプライマーを同位体あるいは蛍光色素で標識するか、または未標識の増幅産物を銀染色することにより、増幅したDNAをバンドとして検出することができる。野生型のパターンとの相違を明らかにするために、コントロールの検体も同時に泳動すると、移動度の違いから変異を持った断片を検出することができる。 In single-strand conformation polymorphism analysis (SSCP analysis; single-strand-conformation-polymorphism-analysis), using primers derived from the base sequence of the genome, including the base sequence of the nucleic acid, using the sample-derived DNA or the sample-derived cDNA as a template The DNA amplified as a fragment smaller than 200 bp is denatured and electrophoresed in a native polyacrylamide gel. When DNA amplification is performed, the amplified DNA can be detected as a band by labeling the primer with an isotope or a fluorescent dye, or silver-staining the unlabeled amplification product. In order to clarify the difference from the wild type pattern, when a control sample is also run at the same time, a fragment having a mutation can be detected from the difference in mobility.
 ミスマッチ化学的切断法(CCM法)では、検体由来DNAあるいは検体由来cDNAをテンプレートとして、核酸の塩基配列を含むゲノムの塩基配列に基づき設計したプライマーで増幅したDNA断片を、核酸に同位体あるいは蛍光標識をとり込ませた標識核酸とハイブリダイズさせ、四酸化オスミウムで処理することでミスマッチの存在する箇所のDNAの一方の鎖を切断させ、変異を検出することができる。CCMは最も感度の高い検出法の1つであり、キロベースの長さの検体にも適応できる。 In the mismatch chemical cleavage method (CCM method), DNA fragments amplified with primers designed based on the base sequence of the genome including the base sequence of the nucleic acid using the sample-derived DNA or the sample-derived cDNA as a template are used as isotopes or fluorescence in the nucleic acid. By hybridizing with a labeled nucleic acid incorporating a label and treating with osmium tetroxide, one strand of DNA at a position where a mismatch exists can be cleaved to detect a mutation. CCM is one of the most sensitive detection methods and can be applied to specimens of kilobase length.
 上記の四酸化オスミウムの代わりに、T4ファージリゾルベースとエンドヌクレアーゼVIIのような細胞内でミスマッチの修復に関与する酵素とRNaseAと組み合わせることで、酵素的にミスマッチを切断することもできる。 In place of the above osmium tetroxide, the mismatch can be cleaved enzymatically by combining RNase A with an enzyme involved in mismatch repair in cells such as T4 phage resol base and endonuclease VII.
 変性ゲル電気泳動法(denaturing gradient gel electrophoresis:DGGE法)では、検体由来DNAあるいは検体由来cDNAをテンプレートに、核酸の塩基配列を含むゲノムの塩基配列に基づき設計したプライマーで増幅したDNA断片を化学的変性剤の濃度勾配や温度勾配を有するゲルを用いて電気泳動する。増幅したDNA断片はゲル内を一本鎖に変性する位置まで移動し、変性後は移動しなくなる。変異がある場合とない場合では増幅したDNAのゲル内での移動が異なることから、変異の存在を検出することができる。検出感度を上げるにはそれぞれのプライマーにポリ(G:C)端末を付けるとよい。 In denaturing gel electrophoresis (DGGE method), DNA fragments amplified with primers designed based on genomic base sequences, including nucleic acid base sequences, are used chemically, using specimen-derived DNA or specimen-derived cDNA as a template. Electrophoresis is performed using a gel having a denaturant concentration gradient and a temperature gradient. The amplified DNA fragment moves in the gel to a position where it is denatured into a single strand and does not move after denaturation. Since there is a difference in the movement of the amplified DNA in the gel with and without the mutation, the presence of the mutation can be detected. In order to increase the detection sensitivity, a poly (G: C) terminal is preferably attached to each primer.
 また、検体由来DNAあるいは検体由来cDNAの塩基配列を直接的に決定し、解析することにより、核酸の変異を検出することもできる。 Also, nucleic acid mutations can be detected by directly determining and analyzing the base sequence of the sample-derived DNA or the sample-derived cDNA.
5.マイクロRNAやマイクロRNA前駆体などの核酸を発現させる方法
 既知のマイクロRNA配列、及びその前駆体の配列は英国サンガーセンターのmiRBaseというデータベースに登録されており、その配列を利用してマイクロRNAやマイクロRNA前駆体などの核酸を作製することができる。また、1で記載した方法により取得したマイクロRNAの配列を用いて作製することもできる。
5. Methods for expressing nucleic acids such as microRNA and microRNA precursors Known microRNA sequences and their precursor sequences are registered in a database called miRBase at the Sanger Center in the UK. Nucleic acids such as RNA precursors can be made. It can also be prepared using the microRNA sequence obtained by the method described in 1.
 核酸は、細胞内に導入して転写されることにより生合成されるようなベクターを用いることにより発現させることができる。具体的には、核酸の塩基配列あるいは、その塩基配列を含むゲノムの塩基配列をもとに、ヘアピン部分を含むDNA断片を調製し、発現ベクターのプロモーター下流に挿入して発現プラスミドを造成し、次に該発現プラスミドを、該発現ベクターに適合した宿主細胞に導入することにより、該核酸を発現させることができる。 The nucleic acid can be expressed by using a vector that is biosynthesized by introduction into a cell and transcription. Specifically, based on the base sequence of the nucleic acid or the genomic base sequence containing the base sequence, a DNA fragment containing the hairpin portion is prepared and inserted downstream of the promoter of the expression vector to construct an expression plasmid. Next, the nucleic acid can be expressed by introducing the expression plasmid into a host cell suitable for the expression vector.
 発現ベクターとしては、宿主細胞において自立複製可能または染色体中への組込みが可能で、核酸の塩基配列を含む遺伝子を転写できる位置にプロモーターを含有しているものが用いられる。プロモーターとしては、宿主細胞中で発現できるものであれば、いかなるものでもよく、例えば、RNA polymerase II(pol II)系プロモーターやU6 RNAやH1 RNAの転写系であるRNA polymerase III(pol III)系プロモーター等をあげることができる。Pol II系プロモーターとしては例えば、サイトメガロウィルス(ヒトCMV)のIE(immediate early)遺伝子のプロモーター、SV40の初期プロモーター等をあげることができる。それらを用いた発現ベクターとして、例えば、pCDNA6.2-GW/miR(Invitrogen社製)、pSilencer 4.1-CMV(Ambion社製)等をあげることができる。pol III系プロモーターとしてはU6 RNAやH1 RNAあるいはtRNA遺伝子のプロモーターをあげることができる。それらを用いた発現ベクターとして、例えば、pSINsi-hH1 DNA(タカラバイオ社製)、pSINsi-hU6 DNA(タカラバイオ社製)、pENTR/U6(Invitrogen社製)などをあげることができる。 As an expression vector, a vector that can replicate autonomously in a host cell or can be integrated into a chromosome and contains a promoter at a position where a gene containing a nucleic acid base sequence can be transcribed is used. Any promoter can be used as long as it can be expressed in the host cell. For example, RNA polymerase II (pol II) type promoter or U RNA or H1 RNA transcription system RNA polymerase III (pol III) system. A promoter etc. can be mention | raise | lifted. Examples of the Pol II promoter include a promoter of cytomegalovirus (human CMV) IE (immediate early) gene, an early promoter of SV40, and the like. Examples of expression vectors using them include pCDNA6.2-GW / miR (Invitrogen), pSilencer® 4.1-CMV (Ambion), and the like. Examples of pol III promoters include U6 RNA, H1 RNA, and tRNA gene promoters. Examples of expression vectors using them include pSINsi-hH1 DNA (Takara Bio), pSINsi-hU6 DNA (Takara Bio), and pENTR / U6 (Invitrogen).
 核酸の塩基配列を含む遺伝子をウィルスベクター内のプロモーター下流に挿入して組換えウィルスベクターを造成し、該ベクターをパッケージング細胞に導入して組換えウィルスを生産して、該核酸の塩基配列を含む遺伝子を発現させることもできる。 A gene containing a nucleic acid base sequence is inserted downstream of a promoter in a viral vector to construct a recombinant viral vector, the vector is introduced into a packaging cell to produce a recombinant virus, and the nucleic acid base sequence is determined. Genes containing can also be expressed.
 パッケージング細胞はウィルスのパッケージングに必要な蛋白質をコードする遺伝子のいずれかを欠損している組換えウィルスベクターの該欠損する蛋白質を補給できる細胞であればいずれの細胞でもよく、例えばヒト腎臓由来のHEK293細胞、マウス線維芽細胞NIH3T3由来の細胞などを用いることができる。パッケージング細胞で補給する蛋白質としては、レトロウィルスベクターの場合はマウスレトロウイルス由来のgag, pol, envなどの蛋白質が、レンチウィルスベクターの場合はHIVウィルス由来のgag, pol, env, vpr, vpu, vif, tat, rev, nefなどの蛋白質、アデノウィルスベクターの場合はアデノウィルス由来のE1A,E1Bなどの蛋白質、また、アデノ随伴ウィルスベクターの場合はRep(p5, p19, p40), Vp(Cap)などの蛋白質を用いることができる。 The packaging cell may be any cell as long as it can replenish the deficient protein of the recombinant viral vector deficient in any of the genes encoding the proteins required for virus packaging, for example, from human kidney HEK293 cells, mouse fibroblast NIH3T3-derived cells, and the like can be used. Proteins supplemented by packaging cells include mouse retrovirus-derived gag, pol, env, etc. for retrovirus vectors, and HIV virus-derived gag, pol, env, vpr, vpu for lentiviral vectors. , Vif, tat, rev, nef, etc., in the case of adenovirus vectors, proteins such as E1A and E1B derived from adenovirus, and in the case of adeno-associated virus vectors, Rep (p5, p19, p40), Vp (Cap), etc. Can be used.
 発現ベクターを用いる以外にも、本発明で用いる核酸をベクターを用いずに直接細胞に導入することもできる。本手法に用いる核酸としては、DNAやRNA、あるいはヌクレオチド類似体の他、これらのキメラ分子、あるいは該核酸の誘導体を用いることができる。具体的には、Pre-miRTM miRNA Precursor Molecules(Ambion社製)やmiRIDIAN microRNA Mimics(GEヘルスケア社製)と同様にして、マイクロRNAやマイクロRNA前駆体などの核酸を発現させることができる。マイクロRNAを発現させる場合は、細胞内で最終的にマイクロRNAができる状態になればいかなる方法でもよく、例えば、(1)マイクロRNA前駆体である一本鎖RNAを導入する他、(2)マイクロRNAそのもの、およびマイクロRNAの相補鎖からなり、100%マッチの二本鎖からなるRNA、(3)マイクロRNAがDicerに切断された後の状態を想定した二本鎖RNAを導入させる方法があげられる。こうした方法を使用した製品としては、miCENTURY OX Precursor(B-Bridge社製)、miCENTURY OX siMature(B-Bridge社製)、miCENTURY OX miNatural(B-Bridge社製)をあげることができる。 In addition to using an expression vector, the nucleic acid used in the present invention can be directly introduced into a cell without using a vector. As a nucleic acid used in this method, in addition to DNA, RNA, or nucleotide analogues, these chimeric molecules or derivatives of the nucleic acids can be used. Specifically, nucleic acids such as microRNAs and microRNA precursors can be expressed in the same manner as Pre-miRTM miRNA Precursor Molecules (Ambion) or miRIDIAN microRNA Mimics (GE Healthcare). When microRNA is expressed, any method can be used as long as microRNA can finally be produced in the cell. For example, (1) In addition to introducing single-stranded RNA as a microRNA precursor, (2) There is a method of introducing microRNA itself and RNA consisting of a complementary strand of microRNA and 100% -matched double-stranded RNA, and (3) double-stranded RNA assuming a state after microRNA is cleaved into Dicer. can give. Products that use these methods include miCENTURY OX Precursor (B-Bridge), miCENTURY OX siMature (B-Bridge), and miCENTURY OX miNatural (B-Bridge).
6.マイクロRNAやマイクロRNA前駆体などの核酸の活性を抑制する方法
 マイクロRNAやマイクロRNA前駆体などの核酸は、アンチセンス技術[バイオサイエンスとインダストリー, 50, 322 (1992)、化学, 46, 681 (1991)、Biotechnology, 9, 358 (1992)、Trends in Biotechnology, 10, 87 (1992)、Trends in Biotechnology, 10,152 (1992)、細胞工学, 16, 1463 (1997)]、トリプル・ヘリックス技術[Trends in Biotechnology, 10, 132 (1992)]、リボザイム技術[Current Opinion in Chemical Biology, 3, 274 (1999)、FEMS Microbiology Reviews, 23, 257 (1999)、Frontiers in Bioscience, 4, D497 (1999)、Chemistry & Biology, 6, R33 (1999)、Nucleic Acids Research, 26, 5237 (1998)、Trends In Biotechnology, 16, 438 (1998)]、デコイDNA法[Nippon Rinsho - Japanese Journal of Clinical Medicine, 56, 563 (1998)、Circulation Research, 82, 1023 (1998)、Experimental Nephrology, 5, 429 (1997)、Nippon Rinsho - Japanese Journal of Clinical Medicine, 54, 2583 (1996)]、あるいはsiRNA(short interfering RNA)を用いて、その活性を抑制することができる。
6). Methods for suppressing the activity of nucleic acids such as microRNAs and microRNA precursors Nucleic acids such as microRNAs and microRNA precursors are antisense technologies [Bioscience and Industry, 50 , 322 (1992), Chemistry, 46 , 681 ( 1991), Biotechnology, 9 , 358 (1992), Trends in Biotechnology, 10 , 87 (1992), Trends in Biotechnology, 10 , 152 (1992), Cell Engineering, 16 , 1463 (1997)], Triple Helix Technology [ Trends in Biotechnology, 10 , 132 (1992)], ribozyme technology [Current Opinion in Chemical Biology, 3 , 274 (1999), FEMS Microbiology Reviews, 23 , 257 (1999), Frontiers in Bioscience, 4 , D497 (1999), Chemistry & Biology, 6 , R33 (1999), Nucleic Acids Research, 26 , 5237 (1998), Trends In Biotechnology, 16 , 438 (1998)], Decoy DNA method [Nippon Rinsho-Japanese Journal of Clinical Medicine, 56 , 563 (1998), Circulation Research, 82 , 1023 (1998), Experimental Nephrology, 5 , 429 (1997), Nippon R insho-Japanese Journal of Clinical Medicine, 54 , 2583 (1996)], or siRNA (short interfering RNA) can be used to suppress the activity.
 アンチセンスとは、ある標的核酸の塩基配列に相補的な塩基配列を有する核酸を塩基配列特異的にハイブリダイゼーションさせ、該標的核酸の発現を抑制できるものをいう。アンチセンスに用いる核酸はDNAやRNAまたはヌクレオチド類似体の他、これらのキメラ分子、あるいは該核酸の誘導体も用いることができる。具体的には、Nature, 432, 226(2004)等に記載の方法に従うことでアンチセンスを作製し、発現を抑制することができる。また、Anti-miRTM miRNA Inhibitors(Ambion社製)やmiRIDIAN microRNA Hairpin Inhibitors(Dharmacon社製)と同様にしてマイクロRNAやマイクロRNA前駆体などの核酸の発現を抑制することができる。 Antisense refers to a nucleic acid that can specifically hybridize a nucleic acid having a base sequence complementary to the base sequence of a certain target nucleic acid to suppress the expression of the target nucleic acid. As the nucleic acid used for antisense, in addition to DNA, RNA or nucleotide analogs, these chimeric molecules or derivatives of the nucleic acids can also be used. Specifically, antisense can be produced and expression can be suppressed by following the method described in Nature, 432 , 226 (2004) and the like. Further, in the same manner as Anti-miRTM miRNA Inhibitors (manufactured by Ambion) and miRIDIAN microRNA Hairpin Inhibitors (manufactured by Dharmacon), the expression of nucleic acids such as microRNAs and microRNA precursors can be suppressed.
 siRNAとは、ある標的核酸の塩基配列を含む短い二本鎖RNAであり、RNA干渉(RNAi)により、該標的核酸の発現を抑制できるものをいう。siRNAの配列は、標的とする塩基配列から文献[Genes Dev., 13, 3191 (1999)]の条件に基づいて適宜設計することができる。例えば選択した19塩基の配列および相補的な配列それぞれの3'端にTTを付加した配列を有する2本のRNAを核酸合成機により合成し、アニーリングすることによりsiRNAを作製できる。また、pSilencer 1.0-U6 (Ambion社製)、pSUPER(OligoEngine社製)等のsiRNA発現用ベクターに上記の選択した19塩基の配列に相当するDNAを挿入することにより、該遺伝子の発現を抑制できるsiRNAを発現するベクターを作製することができる。本発明で用いるマイクロRNAなどの核酸を抑制するsiRNAとしては、該核酸の活性を抑制できるものであればいかなるものでもよいが、配列番号1~594のいずれかで表される塩基配列の、それぞれ9番目の塩基以降の連続した配列情報から設計されたsiRNAが好ましい。siRNAの一方の鎖を構成する塩基の残基数は、好ましくは17~30残基、より好ましくは18~25残基、さらに好ましくは19~23残基である。 The siRNA is a short double-stranded RNA containing a base sequence of a certain target nucleic acid and can suppress the expression of the target nucleic acid by RNA interference (RNAi). sequence of the siRNA, the literature of the nucleotide sequence [Genes Dev., 13, 3191 (1999)] targeting can be appropriately designed based on the conditions. For example, siRNA can be prepared by synthesizing and annealing two RNAs having a sequence of 19 bases selected and a sequence obtained by adding TT to the 3 ′ end of each complementary sequence and annealing. Further, by inserting a DNA corresponding to the selected 19-base sequence into a siRNA expression vector such as pSilencer 1.0-U6 (Ambion) or pSUPER (OligoEngine), the expression of the gene can be suppressed. A vector expressing siRNA can be prepared. The siRNA that suppresses a nucleic acid such as microRNA used in the present invention may be any siRNA that can suppress the activity of the nucleic acid, but each of the nucleotide sequences represented by any of SEQ ID NOs: 1 to 594 SiRNA designed from continuous sequence information after the 9th base is preferred. The number of residues of the base constituting one strand of siRNA is preferably 17 to 30 residues, more preferably 18 to 25 residues, still more preferably 19 to 23 residues.
 線維化した細胞または組織(以下、「線維化細胞」ともいう)で発現するマイクロRNAやマイクロRNA前駆体などの核酸に特異的なアンチセンスまたはsiRNAを用いて、線維化細胞で発現するマイクロRNAやマイクロRNA前駆体などの発現の抑制を行うことができる。例えば、該マイクロRNAまたは該マイクロRNA前駆体に特異的なアンチセンスDNAまたはsiRNAを投与することにより、該マイクロRNAの活性を抑制し、線維化細胞におけるマイクロRNAまたはマイクロRNA前駆体の作用を制御することができる。 MicroRNA expressed in fibrotic cells using antisense or siRNA specific to nucleic acids such as microRNA and microRNA precursors expressed in fibrotic cells or tissues (hereinafter also referred to as “fibrotic cells”) And the expression of microRNA precursors can be suppressed. For example, by administering antisense DNA or siRNA specific to the microRNA or the microRNA precursor, the activity of the microRNA is suppressed and the action of the microRNA or microRNA precursor in fibrotic cells is controlled. can do.
 また、線維化細胞で発現するマイクロRNAまたはその前駆体の発現異常による患者の場合、該マイクロRNAまたはその前駆体に特異的なアンチセンスオリゴヌクレオチドまたはsiRNAを患者に投与することにより、線維化細胞の機能を制御し、上記発現異常により発症する疾患の治療をすることができる。すなわち、該マイクロRNAまたはその前駆体に特異的なアンチセンスオリゴヌクレオチドまたはsiRNAは、細胞外基質の異常産生、すなわち線維化に起因する疾患の治療薬として有用である。 In the case of a patient with abnormal expression of microRNA or its precursor expressed in fibrotic cells, fibrotic cells can be obtained by administering to the patient an antisense oligonucleotide or siRNA specific to the microRNA or its precursor. The disease which develops by the said expression abnormality can be treated. That is, an antisense oligonucleotide or siRNA specific to the microRNA or a precursor thereof is useful as a therapeutic agent for diseases caused by abnormal production of extracellular matrix, that is, fibrosis.
 マイクロRNAまたはその前駆体などの核酸に特異的なアンチセンスオリゴヌクレオチドまたはsiRNAを上記の治療薬として使用する場合は、アンチセンスオリゴヌクレオチドまたはsiRNAを単独、あるいはこれらをコードする核酸をレトロウィルスベクター、アデノウィルスベクター、アデノ随伴ウィルスベクターなどの適当なベクターに挿入した後、下記9に記載した常法に従って医薬製剤とし、投与することができる。 When an antisense oligonucleotide or siRNA specific to a nucleic acid such as microRNA or a precursor thereof is used as the above therapeutic agent, the antisense oligonucleotide or siRNA alone or the nucleic acid encoding them is a retroviral vector, After being inserted into an appropriate vector such as an adenovirus vector or an adeno-associated virus vector, it can be administered as a pharmaceutical preparation according to the conventional method described in 9 below.
7.マイクロRNAやマイクロRNA前駆体などの核酸を用いて遺伝子の機能を抑制する方法
 核酸の標的遺伝子の機能または発現を抑制する方法としては、標的塩基配列を有するmRNAの発現をマイクロRNAなどの核酸が抑制する活性を利用する方法であればいかなる方法を用いてもよい。例えば、マイクロRNAなどの核酸を発現させ、細胞内のマイクロRNAなどの核酸の量を増加させることにより、標的配列を有するmRNAの翻訳を抑制し、遺伝子の発現を抑制する方法をあげることができる。なお、核酸を発現させるには、上記5で記載した方法により行なうことができる。配列番号1~594のいずれかで表される塩基配列からなる核酸の標的塩基配列を有するmRNAとしては、例えば、それぞれ前述の表4で示される遺伝子群を例示することができる。
7). Method of suppressing gene function using nucleic acid such as microRNA or microRNA precursor As a method of suppressing function or expression of target gene of nucleic acid, nucleic acid such as microRNA is used for expression of mRNA having target base sequence. Any method may be used as long as it uses the inhibitory activity. For example, by expressing a nucleic acid such as microRNA and increasing the amount of nucleic acid such as microRNA in the cell, it is possible to suppress the translation of mRNA having the target sequence and to suppress the expression of the gene. . The nucleic acid can be expressed by the method described in 5 above. Examples of the mRNA having the target base sequence of the nucleic acid consisting of the base sequence represented by any of SEQ ID NOs: 1 to 594 can include the gene groups shown in Table 4 above, for example.
 また、表4で示される標的遺伝子に対するsiRNAを用いて、該標的遺伝子の機能を抑制することができる。 Moreover, the function of the target gene can be suppressed using siRNA for the target gene shown in Table 4.
8.マイクロRNAやマイクロRNA前駆体などの核酸を用いて該核酸の発現または機能を促進または抑制させる物質をスクリーニングする方法
 核酸を用いて、マイクロRNAまたはその前駆体などの核酸の発現または機能を促進または抑制させる物質をスクリーニングすることができる。例えば、核酸の塩基配列から、スクリーニングの標的とする塩基配列を選択して、該塩基配列を有する核酸を発現する細胞を利用して、選択したマイクロRNAまたはその前駆体の発現または機能を促進または抑制させる物質をスクリーニングすることができる。
8). Method for screening a substance that promotes or suppresses expression or function of nucleic acid using nucleic acid such as microRNA or microRNA precursor Using nucleic acid to promote expression or function of nucleic acid such as microRNA or its precursor Substances to be suppressed can be screened. For example, by selecting a base sequence to be screened from the base sequence of a nucleic acid and utilizing a cell that expresses a nucleic acid having the base sequence, the expression or function of the selected microRNA or its precursor is promoted or Substances to be suppressed can be screened.
 スクリーニングに用いる、マイクロRNAおよびマイクロRNA前駆体などを発現する細胞としては、線維化が亢進した細胞のほか、上記5で記載したように、該塩基配列を有する核酸を発現するベクターを動物細胞や酵母などの宿主細胞に導入して得られる形質転換細胞や、該塩基配列を有する核酸をベクターを用いずに直接導入した細胞等を用いることもできる。 As a cell expressing microRNA and a microRNA precursor used for screening, in addition to a cell with enhanced fibrosis, as described in 5 above, a vector expressing a nucleic acid having the base sequence may be an animal cell, A transformed cell obtained by introduction into a host cell such as yeast, a cell into which a nucleic acid having the base sequence is directly introduced without using a vector, and the like can also be used.
 具体的なスクリーニング方法としては、スクリーニングの標的とするマイクロRNAまたはその前駆体などの核酸の発現量の変化を指標にする方法の他、マイクロRNAなどの核酸の標的配列を有するmRNAやそれにコードされる遺伝子産物の発現量の変化を指標にする方法があげることができる。
(a)スクリーニングの標的とするマイクロRNAまたはその前駆体などの核酸の発現量の変化を指標にするスクリーニング方法
 該核酸を発現する細胞に対し、試験物質を接触させ、選択した核酸の発現量の変化を指標に、マイクロRNAおよびその前駆体の発現などの核酸を促進または抑制させる物質を得る。核酸の発現量は、上記3で記載した方法により検出することができる。
(b)スクリーニングの標的とするマイクロRNAなどの核酸の標的配列を有するmRNAやそれにコードされる遺伝子産物の発現量の変化を指標にするスクリーニング方法
 該mRNAを発現する細胞に対し、試験物質を接触させ、選択した核酸の標的配列を有するmRNAやそれにコードされる遺伝子産物の発現量の変化を指標に、マイクロRNAおよびその前駆体などの核酸の発現または機能を促進または抑制させる物質を得る。または、マイクロRNAなどの核酸の標的配列を適当なレポーター遺伝子発現ベクターの3’UTRに挿入したDNAを作製して、発現ベクターに適合した宿主細胞に導入し、その細胞に試験物質を接触させ、レポーター遺伝子の発現量の変化を指標に、マイクロRNAおよびその前駆体などの核酸の発現または機能を促進または抑制させる物質を得る。
Specific screening methods include methods that use changes in the expression level of nucleic acids such as microRNAs or their precursors to be screened as indicators, as well as mRNAs that have nucleic acid target sequences such as microRNAs, and encoded by them. A method using the change in the expression level of the gene product as an index can be mentioned.
(A) Screening method using as an index a change in the expression level of a nucleic acid such as a microRNA or a precursor thereof as a target for screening The test substance is contacted with a cell expressing the nucleic acid, and the expression level of the selected nucleic acid is determined. Using the change as an indicator, a substance that promotes or suppresses nucleic acids such as expression of microRNA and its precursor is obtained. The expression level of the nucleic acid can be detected by the method described in 3 above.
(B) Screening method using as an index the change in the expression level of mRNA having a nucleic acid target sequence such as microRNA to be screened and the gene product encoded by it Contact test cells with cells expressing the mRNA Thus, a substance that promotes or suppresses the expression or function of nucleic acids such as microRNA and its precursor is obtained using changes in the expression level of mRNA having the target sequence of the selected nucleic acid and the gene product encoded thereby as an index. Alternatively, a DNA in which the target sequence of a nucleic acid such as microRNA is inserted into the 3′UTR of an appropriate reporter gene expression vector is prepared, introduced into a host cell suitable for the expression vector, and a test substance is contacted with the cell, Using a change in the expression level of the reporter gene as an indicator, a substance that promotes or suppresses the expression or function of nucleic acids such as microRNA and its precursor is obtained.
 標的配列の選択は、上記7で記載した方法により行なうことができ、配列番号1~594のいずれかで表される塩基配列からなるマイクロRNAなどの核酸の標的配列を有するmRNAとしては、例えば、それぞれ前述の表4で示される遺伝子群を例示することができる。 Selection of the target sequence can be performed by the method described in 7 above. Examples of mRNA having a nucleic acid target sequence such as a microRNA comprising a base sequence represented by any of SEQ ID NOs: 1 to 594 include: Examples of the gene groups shown in Table 4 above can be given.
9.マイクロRNAやマイクロRNA前駆体などの核酸を用いた細胞の線維化抑制剤または線維化促進剤
 マイクロRNAやマイクロRNA前駆体などの核酸、およびその塩基配列と相補的な塩基配列を有する核酸は、標的配列を有する遺伝子の発現を制御することにより、細胞の線維化抑制剤または線維化促進剤として利用することができる。
9. Cell fibrosis inhibitor or fibrosis promoter using nucleic acid such as microRNA or microRNA precursor Nucleic acid such as microRNA or microRNA precursor, and nucleic acid having a base sequence complementary to the base sequence are: By controlling the expression of a gene having a target sequence, it can be used as a cell fibrosis inhibitor or fibrosis promoter.
 臓器または細胞の線維化抑制剤としては、以下の(a)~(h)であげられる核酸があげられる。
(a)配列番号1~23、28~411、487~589のいずれかで表される塩基配列からなる核酸
(b)配列番号1~23、28~411、487~589のいずれかで表される塩基配列からなる核酸を含有する、17~28塩基の核酸
(c)配列番号1~23、28~411、487~589のいずれかで表される塩基配列と90%以上の同一性を有する塩基配列からなる核酸
(d)配列番号24~27、412~486、590~594のいずれかで表される塩基配列からなる核酸の相補鎖とストリンジェントな条件でハイブリダイズする核酸
(e)配列番号1~23、28~411、487~589のいずれかで表される塩基配列の2~8番目の塩基配列を含む核酸
(f)配列番号595~618、624~1073、1156~1277のいずれかで表される塩基配列からなる核酸
(g)配列番号595~618、624~1073、1156~1277のいずれかで表される塩基配列と90%以上の同一性を有する塩基配列からなる核酸
(h)配列番号619~623、1074~1155、1278~1281のいずれかで表される塩基配列からなる核酸の相補鎖とストリンジェントな条件でハイブリダイズする核酸。
Examples of the organ or cell fibrosis inhibitor include the following nucleic acids mentioned in (a) to (h).
(A) a nucleic acid comprising a base sequence represented by any one of SEQ ID NOs: 1 to 23, 28 to 411, and 487 to 589 (b) represented by any one of SEQ ID NOs: 1 to 23, 28 to 411, and 487 to 589 A nucleic acid having a nucleotide sequence of 17 to 28 nucleotides (c) having 90% or more identity with the nucleotide sequence represented by any of SEQ ID NOs: 1 to 23, 28 to 411, and 487 to 589 Nucleic acid consisting of a base sequence (d) Nucleic acid (e) that hybridizes under stringent conditions with a complementary strand of a nucleic acid consisting of the base sequence represented by any of SEQ ID NOs: 24-27, 412-486, 590-594 Nucleic acids comprising the second to eighth base sequences of the base sequences represented by any of Nos. 1 to 23, 28 to 411, and 487 to 589 (f) SEQ ID Nos. 595 to 618, 624 to 1073, and 1156 to 1277 Nucleic acid comprising a base sequence represented by any of the above (g) A nucleic acid comprising a base sequence represented by any one of SEQ ID NOs: 595 to 618, 624 to 1073, and 1156 to 1277 and having a nucleotide sequence of 90% or more (H) a nucleic acid that hybridizes under stringent conditions with a complementary strand of a nucleic acid comprising the base sequence represented by any of SEQ ID NOs: 619 to 623, 1074 to 1155, and 1278 to 1281.
 また臓器または細胞の線維化促進剤としては、以下の(i)~(p)であげられる核酸があげられる。
(i)配列番号24~27、412~486、590~594のいずれかで表される塩基配列からなる核酸
(j)配列番号24~27、412~486、590~594のいずれかで表される塩基配列からなる核酸を含有する、17~28塩基の核酸
(k)配列番号24~27、412~486、590~594のいずれかで表される塩基配列と90%以上の同一性を有する塩基配列からなる核酸
(l)配列番号1~23、28~411、487~589のいずれかで表される塩基配列からなる核酸の相補鎖とストリンジェントな条件でハイブリダイズする核酸
(m)配列番号24~27、412~486、590~594のいずれかで表される塩基配列の2~8番目の塩基配列を含む核酸
(n)配列番号619~623、1074~1155、1278~1281のいずれかで表される塩基配列からなる核酸
(o)配列番号619~623、1074~1155、1278~1281のいずれかで表される塩基配列と90%以上の同一性を有する塩基配列からなる核酸
(p)配列番号595~618、624~1073、1156~1277のいずれかで表される塩基配列からなる核酸の相補鎖とストリンジェントな条件でハイブリダイズする核酸。
 上記核酸としては、マイクロRNAまたはマイクロRNA前駆体が好適に用いられる。
Examples of organ or cell fibrosis promoters include nucleic acids listed in (i) to (p) below.
(I) a nucleic acid comprising a base sequence represented by any of SEQ ID NOs: 24-27, 412-486, 590-594 (j) represented by any of SEQ ID NOs: 24-27, 412-486, 590-594 A nucleic acid having a nucleotide sequence of 17 to 28 nucleotides (k) having 90% or more identity with the nucleotide sequence represented by any of SEQ ID NOs: 24 to 27, 412 to 486, and 590 to 594 Nucleic acid consisting of a base sequence (l) Nucleic acid (m) sequence that hybridizes under stringent conditions with a complementary strand of a nucleic acid consisting of a base sequence represented by any one of SEQ ID NOs: 1 to 23, 28 to 411, and 487 to 589 Nucleic acids containing the second to eighth base sequences of the base sequences represented by any of Nos. 24 to 27, 412 to 486, and 590 to 594 (n) SEQ ID Nos. 619 to 623, 1074 to 1155, 12 Nucleic acid consisting of a base sequence represented by any one of 8 to 1281 (o) A base having 90% or more identity with the base sequence represented by any one of SEQ ID NOs: 619 to 623, 1074 to 1155, 1278 to 1281 Nucleic acid consisting of sequence (p) A nucleic acid that hybridizes under stringent conditions with a complementary strand of nucleic acid consisting of a base sequence represented by any of SEQ ID NOs: 595 to 618, 624 to 1073, and 1156 to 1277.
As the nucleic acid, microRNA or a microRNA precursor is preferably used.
 細胞または臓器の線維化促進剤としては上記(a)~(h)に記載の核酸の塩基配列に対して相補的な塩基配列からなる核酸があげられ、細胞または臓器の線維化抑制剤としては上記(i)~(p)に記載の核酸の塩基配列に対して相補的な塩基配列からなる核酸があげられる。また上記の核酸や、これらと相補的な核酸を発現するベクターを細胞の増殖抑制剤または増殖促進剤として用いることもできる。 Examples of the fibrosis promoter for cells or organs include nucleic acids having a base sequence complementary to the base sequences of the nucleic acids described in (a) to (h) above. Examples thereof include nucleic acids having a base sequence complementary to the base sequences of the nucleic acids described in (i) to (p) above. In addition, the above-mentioned nucleic acids and vectors expressing nucleic acids complementary thereto can also be used as cell growth inhibitors or growth promoters.
 一方、上記マイクロRNAなどの核酸の標的遺伝子の発現を抑制する物質や、該標的遺伝子の発現を促進する物質を細胞または臓器の線維化抑制剤または増殖促進剤として用いることもできる。この物質として核酸やこれを発現するベクターを用いることもできる。また標的遺伝子の発現を抑制する物質としては、該標的遺伝子のmRNAに対するsiRNAや該標的遺伝子に対するアンチセンスがあげられ、標的遺伝子の発現を促進する物質としては、該標的遺伝子特異的なマイクロRNAに対するsiRNAや、該標的遺伝子に対するアンチセンスがあげられる。 On the other hand, a substance that suppresses the expression of a target gene of a nucleic acid such as the above microRNA, or a substance that promotes the expression of the target gene can also be used as a fibrosis inhibitor or a proliferation promoter of cells or organs. As this substance, a nucleic acid or a vector expressing it can also be used. Examples of substances that suppress the expression of the target gene include siRNA against the mRNA of the target gene and antisense to the target gene. Substances that promote the expression of the target gene include those against the target gene-specific microRNA. Examples include siRNA and antisense to the target gene.
 細胞または臓器の線維化抑制剤または増殖促進剤の製剤形態や、投与方法などについては、10で後述するマイクロRNAやマイクロRNA前駆体などの核酸を含有する診断薬および治療薬と同様である。 The preparation form and administration method of the cell or organ fibrosis inhibitor or growth promoter are the same as those for diagnostic agents and therapeutic agents containing nucleic acids such as microRNA and microRNA precursor described later in 10.
10.マイクロRNAやマイクロRNA前駆体などの核酸を含有する診断薬および治療薬
 マイクロRNAやマイクロRNA前駆体などの核酸は、標的配列を有する遺伝子の発現を制御したり、マイクロRNAなどの核酸の発現を制御することにより、細胞または臓器の線維化、すなわち細胞外基質の産生異常およびEMT等に起因する疾患の治療薬として利用することができる。さらに、該核酸の標的遺伝子に対するsiRNAは、当該遺伝子の発現を制御することにより、細胞または臓器の線維化、すなわち細胞外基質の産生異常およびEMT等に起因する疾患としては、上述した線維性の組織の障害に伴う線維症等があげられる。
10. Diagnostic and therapeutic agents that contain nucleic acids such as microRNA and microRNA precursors Nucleic acids such as microRNA and microRNA precursors control the expression of genes that have target sequences or control the expression of nucleic acids such as microRNAs By controlling, it can be used as a therapeutic agent for diseases caused by fibrosis of cells or organs, that is, abnormal production of extracellular matrix and EMT. Furthermore, siRNA against the target gene of the nucleic acid controls fibrosis of cells or organs, that is, as a disease caused by abnormal production of extracellular matrix and EMT, etc. Examples include fibrosis associated with tissue damage.
 また、核酸の定量または変異の検出により、細胞または臓器の線維化、すなわち細胞外基質の産生異常およびEMT等に起因する疾患の診断をすることができる。
 核酸を含有する診断薬は、目的の診断法に応じて、核酸の定量あるいは変異の検出を行うために必要な試薬、例えば緩衝剤、塩、反応用酵素、核酸と結合する標識された蛋白、および検出用発色剤等を含んでもよい。
Further, by quantifying nucleic acids or detecting mutations, it is possible to diagnose a disease caused by fibrosis of cells or organs, that is, abnormal production of extracellular matrix and EMT.
A diagnostic agent containing a nucleic acid is a reagent necessary for quantifying a nucleic acid or detecting a mutation, such as a buffer, a salt, a reaction enzyme, a labeled protein that binds to the nucleic acid, depending on the target diagnostic method, And a coloring agent for detection.
 核酸を有効成分として含有する治療薬は、単独で投与することもできるが、通常は薬理学的に許容される1つあるいはそれ以上の担体と一緒に混合し、製剤学の技術分野においてよく知られる任意の方法により製造した医薬製剤として投与するのが望ましい。
 投与経路は、治療に際し最も効果的なものを使用するのが望ましく、経口投与、または口腔内、気道内、直腸内、皮下、筋肉内および静脈内などの非経口投与をあげることができ、望ましくは静脈内投与をあげることができる。
A therapeutic agent containing a nucleic acid as an active ingredient can be administered alone, but usually mixed with one or more pharmacologically acceptable carriers and well known in the pharmaceutical arts. It is desirable to administer it as a pharmaceutical preparation produced by any method.
It is desirable to use the most effective route for treatment, and oral administration or parenteral administration such as buccal, respiratory tract, rectal, subcutaneous, intramuscular and intravenous is desirable. Can be given intravenously.
 投与形態としては、噴霧剤、カプセル剤、錠剤、顆粒剤、シロップ剤、乳剤、坐剤、注射剤、軟膏、テープ剤などがあげられる。
 経口投与に適当な製剤としては、乳剤、シロップ剤、カプセル剤、錠剤、散剤、顆粒剤などがあげられる。
 乳剤およびシロップ剤のような液体調製物は、水、ショ糖、ソルビトール、果糖などの糖類、ポリエチレングリコール、プロピレングリコールなどのグリコール類、ごま油、オリーブ油、大豆油などの油類、p-ヒドロキシ安息香酸エステル類などの防腐剤、ストロベリーフレーバー、ペパーミントなどのフレーバー類などを添加剤として用いて製造できる。
Examples of the dosage form include sprays, capsules, tablets, granules, syrups, emulsions, suppositories, injections, ointments, tapes and the like.
Suitable formulations for oral administration include emulsions, syrups, capsules, tablets, powders, granules and the like.
Liquid preparations such as emulsions and syrups include sugars such as water, sucrose, sorbitol and fructose, glycols such as polyethylene glycol and propylene glycol, oils such as sesame oil, olive oil and soybean oil, p-hydroxybenzoic acid Preservatives such as esters, and flavors such as strawberry flavor and peppermint can be used as additives.
 カプセル剤、錠剤、散剤、顆粒剤などは、乳糖、ブドウ糖、ショ糖、マンニトールなどの賦形剤、デンプン、アルギン酸ナトリウムなどの崩壊剤、ステアリン酸マグネシウム、タルクなどの滑沢剤、ポリビニルアルコール、ヒドロキシプロピルセルロース、ゼラチンなどの結合剤、脂肪酸エステルなどの界面活性剤、グリセリンなどの可塑剤などを添加剤として用いて製造できる。 For capsules, tablets, powders, granules, etc., excipients such as lactose, glucose, sucrose, mannitol, disintegrants such as starch and sodium alginate, lubricants such as magnesium stearate and talc, polyvinyl alcohol, hydroxy A binder such as propylcellulose and gelatin, a surfactant such as fatty acid ester, and a plasticizer such as glycerin can be used as additives.
 非経口投与に適当な製剤としては、注射剤、坐剤、噴霧剤などがあげられる。
 注射剤は、塩溶液、ブドウ糖溶液あるいは両者の混合物からなる担体などを用いて調製される。坐剤はカカオ脂、水素化脂肪またはカルボン酸などの担体を用いて調製される。また、噴霧剤は受容者の口腔および気道粘膜を刺激せず、かつ有効成分を微細な粒子として分散させ吸収を容易にさせる担体などを用いて調製される。
Formulations suitable for parenteral administration include injections, suppositories, sprays and the like.
The injection is prepared using a carrier made of a salt solution, a glucose solution, or a mixture of both. Suppositories are prepared using a carrier such as cacao butter, hydrogenated fat or carboxylic acid. The spray is prepared using a carrier that does not irritate the recipient's oral cavity and airway mucosa, and that facilitates absorption by dispersing the active ingredient as fine particles.
 担体として具体的には乳糖、グリセリンなどが例示される。核酸またはマイクロRNA前駆体、さらには用いる担体の性質により、エアロゾル、ドライパウダーなどの製剤が可能である。また、これらの非経口剤においても経口剤で添加剤として例示した成分を添加することもできる。
 投与量または投与回数は、目的とする治療効果、投与方法、治療期間、年齢、体重などにより異なるが、通常成人1日当たり10 μg/kg~20 mg/kgである。
Specific examples of the carrier include lactose and glycerin. Depending on the nature of the nucleic acid or microRNA precursor, as well as the carrier used, a formulation such as an aerosol or dry powder is possible. In these parenteral preparations, the components exemplified as additives for oral preparations can also be added.
The dose or frequency of administration varies depending on the intended therapeutic effect, administration method, treatment period, age, weight, etc., but is usually 10 μg / kg to 20 mg / kg per day for an adult.
 また、核酸を有効成分として含有する治療薬は、核酸を発現するベクターと核酸治療薬に用いる基剤とを調合することにより製造することもできる[Nature Genet., 8, 42(1994)]。
 治療薬剤に用いる基剤としては、通常注射剤に用いる基剤であればどのようなものでもよく、蒸留水、塩化ナトリウム又は塩化ナトリウムと無機塩との混合物等の塩溶液、マンニトール、ラクトース、デキストラン、グルコース等の溶液、グリシン、アルギニン等のアミノ酸溶液、有機酸溶液又は塩溶液とグルコース溶液との混合溶液等があげられる。また常法に従い、これらの基剤に浸透圧調整剤、pH調整剤、ゴマ油、ダイズ油等の植物油又はレシチンもしくは非イオン界面活性剤等の界面活性剤等の助剤を用いて、溶液、懸濁液、分散液として注射剤を調製してもよい。これらの注射剤を、粉末化、凍結乾燥等の操作により用時溶解用製剤として調製することもできる。治療薬は、治療の直前に液体の場合はそのままで、固体の場合は必要により滅菌処理をした上記の基剤に溶解して治療に使用することができる。
A therapeutic agent containing a nucleic acid as an active ingredient can also be produced by preparing a nucleic acid-expressing vector and a base used for the nucleic acid therapeutic agent [Nature Genet., 8, 42 (1994)].
The base used for the therapeutic agent may be any base as long as it is usually used for injections, such as distilled water, sodium chloride or a salt solution such as a mixture of sodium chloride and an inorganic salt, mannitol, lactose, dextran. And a solution such as glucose, an amino acid solution such as glycine and arginine, an organic acid solution or a mixed solution of a salt solution and a glucose solution, and the like. In addition, according to a conventional method, these bases are mixed with an osmotic pressure adjusting agent, a pH adjusting agent, a vegetable oil such as sesame oil and soybean oil, or an auxiliary such as a surfactant such as lecithin or a nonionic surfactant. An injection may be prepared as a suspension or dispersion. These injections can be prepared as preparations for dissolution at the time of use by operations such as pulverization and freeze-drying. The therapeutic agent can be used for treatment as it is in the case of a liquid just before the treatment, or in the case of a solid, dissolved in the above sterilized base if necessary.
 核酸を発現するベクターは、上記5の方法で作製した組換えウィルスベクターをあげることができ、より具体的には、レトロウィルスベクター及びレンチウィルスベクター等をあげることができる。
 例えば、核酸を、アデノウィルス・ヘキソン蛋白質に特異的なポリリジン-コンジュゲート抗体と組み合わせてコンプレックスを作製し、得られたコンプレックスをアデノウィルスベクターに結合させることにより、ウィルスベクターを調製することができる。該ウィルスベクターは安定に目的の細胞に到達し、エンドソームにより細胞内に取り込まれ、細胞内で分解され核酸を効率的に発現させることができる。
Examples of the nucleic acid-expressing vector include the recombinant virus vector prepared by the above method 5, and more specifically, a retrovirus vector and a lentivirus vector.
For example, a virus vector can be prepared by combining a nucleic acid with a polylysine-conjugated antibody specific for an adenovirus hexon protein to produce a complex, and binding the resulting complex to an adenovirus vector. The virus vector stably reaches a target cell, is taken up into the cell by an endosome, is degraded in the cell, and the nucleic acid can be efficiently expressed.
 また、(-)鎖RNAウィルスであるセンダイウィルスをベースにしたウィルスベクターも開発されており(WO97/16538、WO97/16539)、当該センダイウィルスを用いて、核酸を組み込んだセンダイウィルスを作製することができる。
 核酸は、非ウィルス核酸移入法によっても移入することができる。例えば、リン酸カルシウム共沈法[Virology, 52, 456-467 (1973);Science, 209, 1414-1422 (1980)]、マイクロインジェクション法[Proc. Natl. Acad. Sci. USA, 77, 5399-5403 (1980);Proc. Natl. Acad. Sci. USA, 77, 7380-7384 (1980);Cell, 27, 223-231 (1981);Nature, 294, 92-94 (1981)]、リポソームを介した膜融合-介在移入法[Proc. Natl. Acad. Sci. USA, 84, 7413-7417 (1987);Biochemistry, 28, 9508-9514 (1989);J. Biol. Chem., 264, 12126-12129 (1989);Hum. Gene Ther., 3, 267-275 (1992);Science, 249, 1285-1288 (1990);Circulation, 83, 2007-2011 (1992)]あるいは直接DNA取り込みおよび受容体-媒介DNA移入法[Science, 247, 1465-1468 (1990);J. Biol. Chem., 266, 14338-14342 (1991);Proc. Natl. Acad. Sci. USA, 87, 3655-3659 (1991);J. Biol. Chem., 264, 16985-16987 (1989);BioTechniques, 11, 474-485 (1991);Proc. Natl. Acad. Sci. USA, 87, 3410-3414 (1990);Proc. Natl. Acad. Sci. USA, 88, 4255-4259 (1991);Proc. Natl. Acad. Sci. USA, 87, 4033-4037 (1990);Proc. Natl. Acad. Sci. USA, 88, 8850-8854 (1991);Hum. Gene Ther., 3, 147-154 (1991)]等により移入することができる。
In addition, viral vectors based on Sendai virus (-) strand RNA virus have also been developed (WO97 / 16538, WO97 / 16539), and using the Sendai virus, a Sendai virus incorporating a nucleic acid can be produced. Can do.
Nucleic acids can also be transferred by non-viral nucleic acid transfer methods. For example, calcium phosphate coprecipitation method [Virology, 52 , 456-467 (1973); Science, 209 , 1414-1422 (1980)], microinjection method [Proc. Natl. Acad. Sci. USA, 77 , 5399-5403 ( Proc. Natl. Acad. Sci. USA, 77 , 7380-7384 (1980); Cell, 27 , 223-231 (1981); Nature, 294 , 92-94 (1981)], liposome-mediated membrane Fusion-mediated transfer [Proc. Natl. Acad. Sci. USA, 84 , 7413-7417 (1987); Biochemistry, 28 , 9508-9514 (1989); J. Biol. Chem., 264 , 12126-12129 (1989) Hum. Gene Ther., 3 , 267-275 (1992); Science, 249 , 1285-1288 (1990); Circulation, 83 , 2007-2011 (1992)] or direct DNA uptake and receptor-mediated DNA transfer [Science, 247 , 1465-1468 (1990); J. Biol. Chem., 266 , 14338-14342 (1991); Proc. Natl. Acad. Sci. USA, 87 , 3655-3659 (1991); Biol. Chem., 264 , 16985-16987 (1989); BioTechniques, 11 , 474-485 (1991); Proc. Natl. Acad. Sci. USA, 87 , 3410-3414 (1990); Proc. Natl. Acad. Sci. USA, 88 , 4255-4259 (1991 Proc. Natl. Acad. Sci. USA, 87 , 4033-4037 (1990); Proc. Natl. Acad. Sci. USA, 88 , 8850-8854 (1991); Hum. Gene Ther., 3 , 147- 154 (1991)].
 リポソームを介した膜融合-介在移入法では、リポソーム調製物を目的とする組織に直接投与することにより、核酸を当該組織の局所に取り込み、および発現させることができる[Hum. Gene Ther., 3, 399 (1992)]。DNAを病巣に直接ターゲッティングするには、直接DNA取り込み技術が好ましい。
 受容体-媒介DNA移入は、例えば、ポリリジンを介して、蛋白質リガンドにDNA(通常、共有的に閉環したスーパーコイル化プラスミドの形態をとる)を結合することによって行う方法をあげることができる。リガンドは、目的細胞または組織の細胞表面上の対応するリガンド受容体の存在に基づいて選択する。当該リガンド-DNAコンジュゲートは、所望により、血管に直接注射することができ、受容体結合およびDNA-蛋白質コンプレックスの内在化が起こる標的組織に指向し得る。DNAの細胞内破壊を防止するために、アデノウィルスを同時感染させて、エンドソーム機能を崩壊させることもできる。
In the membrane fusion-mediated transfer method via liposomes, nucleic acid can be taken up and expressed locally in the tissue by administering the liposome preparation directly to the target tissue [Hum. Gene Ther., 3 , 399 (1992)]. Direct DNA uptake techniques are preferred for targeting DNA directly to a lesion.
For example, receptor-mediated DNA transfer can be performed by binding DNA (usually in the form of a covalently closed supercoiled plasmid) to a protein ligand via polylysine. The ligand is selected based on the presence of the corresponding ligand receptor on the cell surface of the target cell or tissue. The ligand-DNA conjugate can be injected directly into the blood vessel, if desired, and can be directed to a target tissue where receptor binding and internalization of the DNA-protein complex occurs. In order to prevent intracellular destruction of DNA, adenovirus can be co-infected to disrupt endosomal function.
11.臓器および細胞の線維化を測定する方法
 線維化の測定方法としては、細胞外基質の産生亢進を測定できる方法であれば特に限定されない。細胞外基質の染色などを用いることができる。細胞外基質を評価する方法としては、マッソン・トリクローム染色が挙げられる。本法では核を鉄ヘマトキシリンで黒色、細胞質を酸性フクシンで赤色、膠原線維をアニリン青で青色に染め分けることができ、組織中の青色領域と線維化領域が比例関係にあることが一般的に認められている[J Clin Pathol. 59(4), 377-81(2006)]。
11. Method for Measuring Fibrosis of Organs and Cells The method for measuring fibrosis is not particularly limited as long as it is a method capable of measuring increased production of extracellular matrix. For example, staining of extracellular matrix can be used. A method for evaluating the extracellular matrix includes Masson's trichrome staining. In this method, the nucleus can be dyed in black with iron hematoxylin, the cytoplasm in red with acidic fuchsin, and the collagen fibers in blue with aniline blue. Generally, the blue region and the fibrosis region in the tissue are in a proportional relationship. It is recognized [J Clin Pathol. 59 (4) , 377-81 (2006)].
12.臓器および細胞の線維化の程度を測定する方法
 臓器および細胞の線維化の程度を測定する方法としては、細胞外基質の遺伝子発現を定量的PCRにより解析することができる。具体的には細胞外基質であるCollagenやFibronectinの発現をSyber-Green(QIAGEN社製)やTaqman-probe(Applied Biosystems社製)といった適切な試薬を用いてPCRを行い、その蛍光強度を測定する方法等が挙げられる。また、細胞外基質をWestern blotやELISA (Enzyme-Linked Immunosorbent Assay)法、比色法等により定量する方法もある。より具体的には、Sircol Soluble Collagen Assay (Biocolor社製)等が挙げられる。
12 Method for measuring the degree of organ and cell fibrosis As a method for measuring the degree of organ and cell fibrosis, gene expression of the extracellular matrix can be analyzed by quantitative PCR. Specifically, the expression of Collagen and Fibronectin, which are extracellular substrates, is performed by PCR using appropriate reagents such as Syber-Green (QIAGEN) and Taqman-probe (Applied Biosystems), and the fluorescence intensity is measured. Methods and the like. There are also methods for quantifying extracellular matrix by Western blot, ELISA (Enzyme-Linked Immunosorbent Assay) method, colorimetric method and the like. More specifically, Sircol Soluble Collagen Assay (manufactured by Biocolor) and the like can be mentioned.
 また、臓器および細胞の線維化の過程で特に重要な現象と言われるEMTに限定して線維化を測定することもできる。
 EMTを評価する方法としては、β-cateninやZO-1, E-cadherinといった上皮系細胞で特異的に発現する細胞マーカーの減少や、FSP-1, αSMA, Vimentinといった間葉系細胞で特異的に発現する細胞マーカーの増加をそれぞれの抗体で免疫染色することにより測定することができるほか、それらの遺伝子発現を定量的PCRによって解析すれば、定量化も可能である[J Clin Invest., 117, 482-91(2007)]。
In addition, fibrosis can be measured only in EMT, which is said to be a particularly important phenomenon in the process of organ and cell fibrosis.
Methods for evaluating EMT include reduction of cell markers that are specifically expressed in epithelial cells such as β-catenin and ZO-1, E-cadherin, and mesenchymal cells such as FSP-1, αSMA, and Vimentin. In addition to measuring the increase in cell markers expressed in each antibody by immunostaining with each antibody, quantitative analysis is also possible by analyzing their gene expression by quantitative PCR [J Clin Invest., 117 , 482-91 (2007)].
 以下に実施例により、本発明を具体的に説明する。ただし、本発明はこれらの実施例に限定されるものではない。 Hereinafter, the present invention will be described specifically by way of examples. However, the present invention is not limited to these examples.
腎線維化時におけるmicroRNAの発現
 腎線維化の一般的な動物モデルとして知られる一側尿管結紮(Unilateral ureteral obstruction : UUO)モデルを用いて線維化腎におけるmicroRNAの発現の違いについて調べた。
 7週齢のBALB/cマウス(チャールズリバージャパン社より購入)の左腎尿管を麻酔下で結紮し、4日後に左腎を摘出し、Trizol試薬(Invitrogen 社製)を加えて、製品に記載された方法に従って全RNAを抽出した。mirVana(登録商標)miRNA Bioarrays V9.2(Ambion 社製)を用いて、コントロールマウス、尿管結紮マウス各2匹についてmicroRNAマイクロアレイ解析を実施した。なお、これら解析に用いたサンプルはTGF-β, collagen, fibronectin等の線維化関連遺伝子の発現が上昇していることを確認している。
 コントロールマウスにおけるmicroRNAの発現を1.00として、尿管結紮マウスにおけるmicroRNAの発現量と比較し、尿管結紮マウス2匹ともにおいて発現変動のみられたものについて選抜した。表5にはコントロールに対する尿管結紮マウスの発現比の2匹の平均値を示す。表5にあるように31個の候補microRNAが得られた。
 表5中、mmu_miR_345は配列番号28、hsa_miR_320は配列番号2、hsa_miR_422bはhsa_miR_378に対応し配列番号3、rno_miR_422bはrno_miR_378に対応し配列番号45、hsa_miR_213は配列番号4、mmu_miR_99aは配列番号51、hsa_miR_192は配列番号6、hsa_miR_422aは配列番号7、hsa_miR_30e_5pは配列番号9、hsa_miR_30dは配列番号10、hsa_miR_30bは配列番号11、hsa_miR_30cは配列番号12、hsa_miR_30a_5pは配列番号13、hsa_miR_486は配列番号14、mmu_miR_187は配列番号266、hsa_miR_185は配列番号16、mmu_miR_203は配列番号289、hsa_miR_451は配列番号18、mmu_miR_451は配列番号301、hsa_miR_200aは配列番号19、hsa_miR_29cは配列番号20、rno_miR_140_ASは配列番号399、hsa_miR_200bは配列番号22、mmu_miR_192は配列番号72、hsa_miR_146bは配列番号25、hsa_miR_21は配列番号26、hsa_miR_223は配列番号27で表される塩基配列を有するマイクロRNAを表す。
 また、表5中、ambi_miR_3998、ambi_miR_7029、ambi_miR_13268及びambi_miR_13143で示されるマイクロRNAは、それぞれmiRBaseに登録されたhsa_miR_213(配列番号4)、hsa_miR_451(配列番号18)、hsa_miR_140-3p(配列番号21)、及びhsa_miR_1974(配列番号23)に対応する。
Expression of microRNA during renal fibrosis The difference in microRNA expression in fibrotic kidney was examined using a unilateral ureteral obstruction (UUO) model, which is known as a general animal model of renal fibrosis.
The left renal ureter of 7-week-old BALB / c mice (purchased from Charles River Japan) was ligated under anesthesia, 4 days later, the left kidney was removed, Trizol reagent (Invitrogen) was added, and the product was added to the product. Total RNA was extracted according to the method described. Using mirVana (registered trademark) miRNA Bioarrays V9.2 (manufactured by Ambion), microRNA microarray analysis was performed on each of two control mice and ureter-ligated mice. In addition, it was confirmed that the samples used for these analyzes showed increased expression of fibrosis-related genes such as TGF-β, collagen, fibronectin and the like.
The expression of microRNA in the control mouse was set to 1.00, and compared with the expression level of microRNA in the ureter-ligated mouse, the expression was observed in both ureter-ligated mice. Table 5 shows the average value of two urinary ligation mice relative to the control. As shown in Table 5, 31 candidate microRNAs were obtained.
In Table 5, mmu_miR_345 is SEQ ID NO: 28, hsa_miR_320 is SEQ ID NO: 2, hsa_miR_422b corresponds to hsa_miR_378, SEQ ID NO: 3, rno_miR_422b corresponds to rno_miR_378, SEQ ID NO: 45, hsa_miR_213 is SEQ ID NO: 4, mmu_miR_99h is SEQ ID NO: 4, SEQ ID NO: 6, hsa_miR_422a is SEQ ID NO: 7, hsa_miR_30e_5p is SEQ ID NO: 9, hsa_miR_30d is SEQ ID NO: 10, hsa_miR_30b is SEQ ID NO: 11, hsa_miR_30c is SEQ ID NO: 12, hsa_miR_30a_5p is SEQ ID NO: 13, hsa_miR_u is SEQ ID NO: 13 266, hsa_miR_185 is SEQ ID NO: 16, mmu_miR_203 is SEQ ID NO: 289, hsa_miR_451 is SEQ ID NO: 18, mmu_miR_451 is SEQ ID NO: 301, hsa_miR_29a is SEQ ID NO: 19, hsa_miR_29c is SEQ ID NO: 20, rno_miR_140_AS is SEQ ID NO: 399, hsa_miR mmu_miR_192 is SEQ ID NO: 72, hsa_miR_146b is SEQ ID NO: 25, hsa_miR_21 is SEQ ID NO: 26, and hsa_miR_223 is SEQ ID NO: It represents a micro RNA having the nucleotide sequence represented by 27.
In Table 5, the microRNAs indicated by ambi_miR_3998, ambi_miR_7029, ambi_miR_13268, and ambi_miR_13143 are hsa_miR_213 (SEQ ID NO: 4), hsa_miR_451 (SEQ ID NO: 18), hsa_miR_140-3p (SEQ ID NO: 21) registered in miRBase, respectively. This corresponds to hsa_miR_1974 (SEQ ID NO: 23).
Figure JPOXMLDOC01-appb-T000037

 
Figure JPOXMLDOC01-appb-T000037

 
細胞の線維化時におけるmicroRNAの発現量解析
 尿管結紮マウスにおけるmicroRNAの変動が線維化にかかわるものなのか更に調べるため、線維化の中心的ファクターであるTGF-βで腎線維芽細胞、腎尿細管上皮細胞を刺激した際のmicroRNAの発現の変動について定量的PCRを用いて調べた。
 ラット腎線維芽細胞株であるNRK-49F細胞(以下、NRK-49Fと称す)は、5 %ウシ胎児血清(GIBCO社製)を含むDMEM培地(GIBCO社製)で37℃の5 % CO2濃度のインキュベーター中で培養した。
 NRK-49Fを6穴プレートに1穴あたり3×104個となるように播種し、上記培地中で一晩培養した。1日後に培地に血清を含まないDMEM培地に交換した。さらに16時間後、培地をrecombinant human TGF-β1(R&D社製)を終濃度5 ng / mlとなるように血清を含まないDMEM培地を用いて希釈したものへ交換し、さらに培養を続けた。陰性コントロールとしては、TGF-β1を含まない無血清DMEMへ培地を交換した。
 TGF-β1の刺激48時間後に、培地を除き、Trizol試薬(Invitrogen社製)を用いて、製品に記載された方法に従って全RNAを抽出した。RNA濃度を測定した後、TaqMan MicroRNA assays(Applied Biosystems社製)を用いて、製品に記載された方法に従って反応させ、Applied Biosystems 7900HT FastリアルタイムPCRシステム(Applied Biosystems社製)を用いてreal-time PCRを行った。
 TGFβ-1を加えなかったコントロール細胞のmicroRNA発現量を1.00として、それぞれの相対発現量を計算した。その結果、表6に示すように、TGF-β刺激によってmiR-378, 192, 30e-5p, 30b, 30c, 30d, 486, 185, 320, 451, 200a, 29cの発現が低下することが明らかとなった。特に、miR-378, 30b, 30c, 486, 451, 29c については50%以上の発現低下がみられた。
 表6中、miR-378は配列番号3、miR-192は配列番号6、miR-30e-5pは配列番号9、miR-30dは配列番号10、miR-30cは配列番号12、miR-30bは配列番号11、miR-486は配列番号14、miR-185は配列番号16、miR-320は配列番号2、miR-451は配列番号18、miR-200aは配列番号19、miR-29cは配列番号20で表される塩基配列を有するマイクロRNAを表す。尚、TaqMan MicroRNA assaysにはヒト配列で表されるプローブを使用した。
Analysis of microRNA expression during cell fibrosis To further investigate whether microRNA fluctuations in ureter-ligated mice are involved in fibrosis, TGF-β, the central factor of fibrosis, was used to measure renal fibroblasts and renal urine. Changes in microRNA expression upon stimulation of tubule epithelial cells were examined using quantitative PCR.
A rat renal fibroblast cell line, NRK-49F cells (hereinafter referred to as NRK-49F), is a DMEM medium (GIBCO) containing 5% fetal calf serum (GIBCO) and 5% CO 2 at 37 ° C. Cultured in a concentration incubator.
NRK-49F was seeded in a 6-well plate at 3 × 10 4 per well and cultured overnight in the above medium. One day later, the medium was replaced with DMEM medium without serum. After another 16 hours, the medium was replaced with a recombinant human TGF-β1 (manufactured by R & D) diluted to a final concentration of 5 ng / ml using DMEM medium without serum, and the culture was continued. As a negative control, the medium was changed to serum-free DMEM not containing TGF-β1.
After 48 hours of stimulation with TGF-β1, the medium was removed, and total RNA was extracted using Trizol reagent (Invitrogen) according to the method described in the product. After measuring the RNA concentration, react with TaqMan MicroRNA assays (Applied Biosystems) according to the method described in the product, and use Real-time PCR with Applied Biosystems 7900HT Fast real-time PCR system (Applied Biosystems). Went.
The relative expression level of each control cell was calculated by setting the microRNA expression level of control cells to which TGFβ-1 was not added to 1.00. As a result, as shown in Table 6, it is clear that miR-378, 192, 30e-5p, 30b, 30c, 30d, 486, 185, 320, 451, 200a, 29c expression is reduced by TGF-β stimulation. It became. In particular, miR-378, 30b, 30c, 486, 451, 29c showed an expression decrease of 50% or more.
In Table 6, miR-378 is SEQ ID NO: 3, miR-192 is SEQ ID NO: 6, miR-30e-5p is SEQ ID NO: 9, miR-30d is SEQ ID NO: 10, miR-30c is SEQ ID NO: 12, and miR-30b is SEQ ID NO: 11, miR-486 is SEQ ID NO: 14, miR-185 is SEQ ID NO: 16, miR-320 is SEQ ID NO: 2, miR-451 is SEQ ID NO: 18, miR-200a is SEQ ID NO: 19, and miR-29c is SEQ ID NO: The microRNA which has a base sequence represented by 20 is represented. In addition, probes represented by human sequences were used for TaqMan MicroRNA assays.
Figure JPOXMLDOC01-appb-T000038

 
Figure JPOXMLDOC01-appb-T000038

 
 また、別の実験として、イヌの腎上皮細胞であるMDCK細胞を用いて同様に対象microRNAの発現解析を実施した。
 MDCKを24穴プレートに1穴あたり1×104個となるように播種し、上記培地中で一晩培養した。1日後に培地をrecombinant human TGF-β1(R&D社製)を終濃度10 ng / mlとなるように10 %ウシ胎児血清を含むDMEM培地を用いて希釈したものへ交換し、さらに培養を続けた。
 TGF-β1の刺激後72時間後に、培地を除き、Trizol試薬(Invitrogen社製)を用いて、製品に記載された方法に従って全RNAを抽出し、濃度測定後、TaqMan MicroRNA assays(Applied Biosystems社製)を用いて、製品に記載された方法に従って反応させ、Applied Biosystems 7900HT FastリアルタイムPCRシステム(Applied Biosystems社製)を用いてreal-time PCRを行い、TGF-β刺激をしないコントロール細胞とmicroRNAの発現量を比較した。
 TGF-β1を加えなかったコントロール細胞のmicroRNA発現量を1.00として、それぞれの相対発現量を計算した。その結果、表7に示すように、TGF-β刺激によってmiR-30b,30c,486の発現が低下することが明らかとなった。これはNRK-49Fにおける結果、UUOモデルにおける結果と一致するものである。また、miR-146b, 21, 223の発現はTGF-β刺激により上昇した。
 表7中、miR-30bは配列番号11、miR-30cは配列番号12、miR-486は配列番号14、miR-146bは配列番号25、miR-21は配列番号26、miR-223は配列番号27で表される塩基配列を有するマイクロRNAを表す。尚、TaqMan MicroRNA assaysにはヒト配列で表されるプローブを使用した。
As another experiment, expression analysis of the target microRNA was similarly performed using MDCK cells, which are canine renal epithelial cells.
MDCK was seeded in a 24-well plate at 1 × 10 4 per well and cultured overnight in the above medium. One day later, the medium was replaced with recombinant human TGF-β1 (R & D) diluted with DMEM medium containing 10% fetal bovine serum to a final concentration of 10 ng / ml, and the culture was continued. .
72 hours after stimulation with TGF-β1, the medium was removed, and total RNA was extracted using Trizol reagent (Invitrogen) according to the method described in the product.After concentration measurement, TaqMan MicroRNA assays (Applied Biosystems) ), Followed by real-time PCR using Applied Biosystems 7900HT Fast real-time PCR system (Applied Biosystems), and expression of control cells that do not stimulate TGF-β and microRNA The amount was compared.
The relative expression level of each control cell was calculated by setting the microRNA expression level of the control cells to which TGF-β1 was not added to 1.00. As a result, as shown in Table 7, it was revealed that miR-30b, 30c, and 486 expression was reduced by TGF-β stimulation. This is consistent with the result in the UUO model as a result of NRK-49F. In addition, miR-146b, 21, 223 expression was increased by TGF-β stimulation.
In Table 7, miR-30b is SEQ ID NO: 11, miR-30c is SEQ ID NO: 12, miR-486 is SEQ ID NO: 14, miR-146b is SEQ ID NO: 25, miR-21 is SEQ ID NO: 26, and miR-223 is SEQ ID NO: The microRNA which has a base sequence represented by 27 is represented. In addition, probes represented by human sequences were used for TaqMan MicroRNA assays.
Figure JPOXMLDOC01-appb-T000039

 
Figure JPOXMLDOC01-appb-T000039

 
microRNAを強制発現させた腎尿細管上皮細胞の細胞外マトリックス産生に対する影響
 microRNAの前駆体を腎尿細管上皮細胞に導入し、該分子から生成されるmicroRNAがコラーゲン産生に及ぼす影響を調べた。腎線維化には尿細管細胞が関与していることが知られており、そのモデルとして株化細胞であるNRK-52Eが用いられる。またその尿細管細胞の線維化の指標として、collagen I等が用いられる[JASN., 16, 2702-13(2005)], [Journal of Cellular Biochemistry, 103, 1999-2009(2008)]。
 ラット腎尿細管上皮細胞株であるNRK-52E細胞(以下、NRK-52Eと称す)は、5 %ウシ胎児血清(GIBCO社製)を含むDMEM培地(GIBCO社製)で37℃の5 % CO2濃度のインキュベーター中で培養した。
 NRK-52Eを24穴プレートに1穴あたり1×104個となるように播種し、上記培地中で一晩培養した。1日後、miR-21, 223, 30b, 30c, 486, 194, let-7iを生成するpre-miR miRNA precursor molecules (Ambion社製)をlipofectamine 2000(Invitrogen社製)を用いたリポフェクション法により終濃度が5 nMとなるようにNRK-52Eに導入した。また、miRNA precursor molecules-Negative control (Ambion社製)も陰性コントロールとして導入した。リポフェクションは、製品に添付された説明書に記載された方法に従った。
 リポフェクション法により該分子を導入した24時間後、培地をrecombinant human TGF-β1(R&D社製)を終濃度2 ng / mlとなるように5 %ウシ胎児血清を含むDMEM 培地を用いて希釈したものへ交換し、さらに培養を続けた。
 TGF-β1を含んだ培地に交換した2日後、培地を除き、Trizol試薬(Invitrogen社製)を用いて、製品に記載された方法に従って全RNAを抽出し、濃度測定後、等量のRNAよりPrimeScript 1st strand cDNA Synthesis Kit(タカラバイオ社製)を用いて、製品に記載された方法に従って、cDNAを合成した。
 作製したcDNAを用いて、real-time RT-PCR法により線維化関連遺伝子の発現を解析した。QuantiTect SYBR Green PCR Kit(QIAGEN社製)を用いて製品に記載された方法に従ってサンプルを調製し、Applied Biosystems 7900HT Fast リアルタイムPCRシステム(Applied Biosystems社製)を用いてcollagen Iの発現量を定量した。陰性コントロールとして用いたmiRNA precursor molecules-Negative controlを導入した細胞におけるcollagen Iの発現量を1.00としてそれぞれのmicroRNAを導入した細胞におけるcollagen Iの発現量を計算した結果、表8に示すように、let-7i(配列番号8)、miR-30b(配列番号11)、miR-30c(配列番号12)、miR-486(配列番号14)を生成する分子を導入することにより、collagen Iの発現が低下することが明らかとなった。またmiR-194(配列番号24)、miR-21(配列番号26)、miR-223(配列番号27)を生成する分子を導入することにより、collagen Iの発現が上昇することが明らかとなった。
Influence on extracellular matrix production of renal tubular epithelial cells in which microRNA was forcibly expressed A precursor of microRNA was introduced into renal tubular epithelial cells, and the influence of microRNA produced from the molecules on collagen production was examined. Renal fibrosis is known to involve tubule cells, and the cell line NRK-52E is used as a model. Collagen I and the like are used as an index of tubule cell fibrosis [JASN., 16 , 2702-13 (2005)], [Journal of Cellular Biochemistry, 103 , 1999-2009 (2008)].
NRK-52E cells (hereinafter referred to as NRK-52E), a rat renal tubular epithelial cell line, are 5% CO at 37 ° C in DMEM medium (GIBCO) containing 5% fetal calf serum (GIBCO). The cells were cultured in a 2 concentration incubator.
NRK-52E was seeded in a 24-well plate at 1 × 10 4 per well and cultured overnight in the above medium. One day later, the final concentration of pre-miR miRNA precursor molecules (Ambion) producing miR-21, 223, 30b, 30c, 486, 194, let-7i was determined by lipofection using lipofectamine 2000 (Invitrogen). Was introduced into NRK-52E to be 5 nM. MiRNA precursor molecules-Negative control (Ambion) was also introduced as a negative control. Lipofection followed the method described in the instructions attached to the product.
24 hours after the introduction of the molecule by lipofection method, the medium was diluted with DMEM medium containing 5% fetal bovine serum to a final concentration of 2 ng / ml of recombinant human TGF-β1 (R & D). The culture was continued.
Two days after changing to a medium containing TGF-β1, the medium was removed, and using Trizol reagent (Invitrogen), total RNA was extracted according to the method described in the product. Using PrimeScript 1st strand cDNA Synthesis Kit (manufactured by Takara Bio Inc.), cDNA was synthesized according to the method described in the product.
Using the prepared cDNA, the expression of fibrosis-related genes was analyzed by real-time RT-PCR. Samples were prepared using the QuantiTect SYBR Green PCR Kit (QIAGEN) according to the method described in the product, and the expression level of collagen I was quantified using an Applied Biosystems 7900HT Fast real-time PCR system (Applied Biosystems). As a result of calculating the expression level of collagen I in cells into which each microRNA was introduced, assuming that the expression level of collagen I in cells into which miRNA precursor molecules-Negative control used as negative control was introduced was 1.00, let -7i (SEQ ID NO: 8), miR-30b (SEQ ID NO: 11), miR-30c (SEQ ID NO: 12), and miR-486 (SEQ ID NO: 14) are introduced to reduce the expression of collagen I It became clear to do. It was also clarified that the expression of collagen I was increased by introducing molecules that generate miR-194 (SEQ ID NO: 24), miR-21 (SEQ ID NO: 26), and miR-223 (SEQ ID NO: 27). .
Figure JPOXMLDOC01-appb-T000040

 
Figure JPOXMLDOC01-appb-T000040

 
microRNAを強制発現させた線維芽細胞の細胞外マトリックス産生に対する影響
 pre-miR miRNA precursor molecules(Ambion社製)を線維芽細胞に導入し、該分子から生成されるmicroRNAがコラーゲン産生に及ぼす影響を調べた。腎線維化には腎組織中の線維芽細胞が関与していることが知られており、そのモデルとして株化細胞であるNRK-49Fが用いられる。またその線維化の指標としては、collagen I等が用いられる[JASN., 16, 2702-13(2005)], [Kidney International, 66, 112-120(2004)]。
 NRK-49Fを24穴プレートに1穴あたり2×104となるように播種し、上記培地中で一晩培養した。1日後、miR-21, 30b, 30c, 192, 486, let-7iを生成するpre-miR miRNA precursor molecules をlipofectamine 2000(Invitrogen社製)を用いたリポフェクション法により終濃度が5 nMとなるようにNRK-49Fに導入した。また、miRNA precursor molecules-Negative control(Ambion社製)も陰性コントロールとして導入した。リポフェクションは、製品に添付された説明書に記載された方法に従った。
 リポフェクション法により該分子を導入した8時間後、培地を血清を含まないDMEM培地に交換した。さらに16時間後、培地をrecombinant human TGF-β1(R&D社製)を終濃度1 ng/ ml となるように血清を含まないDMEM培地を用いて希釈したものへ交換し、さらに培養を続けた。
 TGF-β1を含んだ培地に交換した2日後、培地を除き、Trizol試薬(Invitrogen社製)を用いて、製品に記載された方法に従って全RNAを抽出し、濃度測定後、PrimeScript 1st strand cDNA Synthesis Kit(タカラバイオ社製)を用いて、製品に記載された方法に従って、cDNAを合成した。
 作製したcDNAを用いて、real-time RT-PCR法により線維化関連遺伝子の発現を解析した。QuantiTect SYBR Green PCR Kit(QIAGEN社製)を用いて製品に記載された方法に従ってサンプルを調製し、Applied Biosystems 7900HT Fast リアルタイムPCRシステム(Applied Biosystems社製)を用いてcollagen Iの発現量を定量した。その結果、表9に示すように、let-7i(配列番号8)、miR-30b(配列番号11)、miR-30c(配列番号12)、miR-192(配列番号6)、miR-486(配列番号14)を生成する分子を導入することにより、collagen Iの発現が低下することが明らかとなった。また、miR-21(配列番号26)を生成する分子を導入することにより、collagen Iの発現が上昇することが明らかとなった。
Effects of fibroblasts forced to express microRNAs on extracellular matrix production Pre-miR miRNA precursor molecules (Ambion) were introduced into fibroblasts, and the effects of microRNAs generated from these molecules on collagen production were examined. It was. Renal fibrosis is known to involve fibroblasts in kidney tissue, and the model cell line NRK-49F is used as the model. Collagen I or the like is used as an index of fibrosis [JASN., 16 , 2702-13 (2005)], [Kidney International, 66 , 112-120 (2004)].
NRK-49F was seeded in a 24-well plate at 2 × 10 4 per well and cultured overnight in the above medium. One day later, the final concentration of pre-miR miRNA precursor molecules that produce miR-21, 30b, 30c, 192, 486, let-7i is 5 nM by lipofection using lipofectamine 2000 (Invitrogen). Introduced into NRK-49F. MiRNA precursor molecules-Negative control (Ambion) was also introduced as a negative control. Lipofection followed the method described in the instructions attached to the product.
Eight hours after the introduction of the molecule by the lipofection method, the medium was replaced with serum-free DMEM medium. After another 16 hours, the medium was replaced with recombinant human TGF-β1 (R & D) diluted to a final concentration of 1 ng / ml using DMEM medium without serum, and the culture was continued.
Two days after replacement with a medium containing TGF-β1, the medium was removed, and using Trizol reagent (Invitrogen), total RNA was extracted according to the method described in the product. After measuring the concentration, PrimeScript 1st strand cDNA Synthesis Using Kit (manufactured by Takara Bio Inc.), cDNA was synthesized according to the method described in the product.
Using the prepared cDNA, the expression of fibrosis-related genes was analyzed by real-time RT-PCR. Samples were prepared using the QuantiTect SYBR Green PCR Kit (QIAGEN) according to the method described in the product, and the expression level of collagen I was quantified using an Applied Biosystems 7900HT Fast real-time PCR system (Applied Biosystems). As a result, as shown in Table 9, let-7i (SEQ ID NO: 8), miR-30b (SEQ ID NO: 11), miR-30c (SEQ ID NO: 12), miR-192 (SEQ ID NO: 6), miR-486 ( It was revealed that the expression of collagen I is reduced by introducing a molecule that generates SEQ ID NO: 14). It was also revealed that collagen I expression was increased by introducing a molecule that generates miR-21 (SEQ ID NO: 26).
Figure JPOXMLDOC01-appb-T000041

 
Figure JPOXMLDOC01-appb-T000041

 
microRNAを強制発現させた腎尿細管上皮細胞の上皮間葉転換 (EMT) に対する影響
 腎尿細管上皮細胞が間葉系細胞へと形質転換(EMT)し、細胞外マトリックス蛋白を過剰に産生する細胞となることが線維化を促進する原因の一端を担っていることが明らかとなっている。そこで、pre-miR miRNA precursor molecules (Ambion社製)を腎尿細管上皮細胞に導入し、該分子から生成されるmicroRNAがEMTに及ぼす影響を調べた。上皮間葉転換(EMT)の解析には、イヌの尿細管上皮細胞の細胞株であるMDCK細胞におけるα smooth muscle actin(αSMA)の発現上昇が用いられる[Am J Physiol Renal Physiol, 291, F1332-42 (2006), JASN., 18, 1497-1507, (2007)]。
 MDCKを24穴プレートに1穴あたり1×104となるように播種し、上記培地中で一晩培養した。1日後、miR-21, 146b, 223, 194, let-7iを生成するpre-miR miRNA precursor molecules をlipofectamine 2000(Invitrogen社製)を用いたリポフェクション法により終濃度が5 nMとなるようにMDCKに導入した。また、miRNA precursor molecules-Negative control(Ambion社製)も陰性コントロールとして導入した。リポフェクションは、製品に添付された説明書に記載された方法に従った。
 リポフェクション法により該分子を導入した24時間後、培地をrecombinant human TGF-β1 (R&D 社製)を終濃度2 ng/mlとなるように10 %ウシ胎児血清を含むDMEM培地を用いて希釈したものへ交換し、さらに培養を続けた。
 TGF-β1を含んだ培地に交換した2日後、培地を除き、Trizol試薬(Invitrogen社製)を用いて、製品に記載された方法に従って全RNAを抽出し、濃度測定後、PrimeScript 1st strand cDNA Synthesis Kit(タカラバイオ社製)を用いて、製品に記載された方法に従って、cDNAを合成した。
 作製したcDNAを用いて、real-time RT-PCR法により線維化関連遺伝子の発現を解析した。QuantiTect SYBR Green PCR Kit(QIAGEN社製)を用いて製品に記載された方法に従ってサンプルを調製し、Applied Biosystems 7900HT Fast リアルタイムPCRシステム(Applied Biosystems社製)を用いてEMTの指標であるαSMAの発現量を定量した。その結果、表10に示すように、let-7i(配列番号8)を生成する分子を導入することにより、αSMAの発現が低下することが明らかとなった。また、miR-194(配列番号24)、miR-146b(配列番号25)、miR-21(配列番号26)、miR-223(配列番号27)を生成する分子を導入することにより、αSMAの発現が上昇することが明らかとなった。
Effect on renal epithelial-mesenchymal transition (EMT) of renal tubular epithelial cells in which microRNA is forcibly expressed Cells in which renal tubular epithelial cells are transformed into mesenchymal cells (EMT), resulting in excessive production of extracellular matrix proteins It has become clear that this is part of the cause of promoting fibrosis. Therefore, pre-miR miRNA precursor molecules (manufactured by Ambion) were introduced into renal tubular epithelial cells, and the influence of microRNA produced from the molecules on EMT was examined. For the analysis of epithelial-mesenchymal transition (EMT), increased expression of α smooth muscle actin (αSMA) in MDCK cells, a canine tubular epithelial cell line, is used [Am J Physiol Renal Physiol, 291 , F1332- 42 (2006), JASN., 18 , 1497-1507, (2007)].
MDCK was seeded in a 24-well plate at 1 × 10 4 per well and cultured overnight in the above medium. One day later, pre-miR miRNA precursor molecules that generate miR-21, 146b, 223, 194, let-7i were made MDCK to a final concentration of 5 nM by lipofection using lipofectamine 2000 (Invitrogen). Introduced. MiRNA precursor molecules-Negative control (Ambion) was also introduced as a negative control. Lipofection followed the method described in the instructions attached to the product.
24 hours after introduction of the molecule by the lipofection method, the medium was diluted with DMEM medium containing 10% fetal calf serum to a final concentration of 2 ng / ml of recombinant human TGF-β1 (R & D). The culture was continued.
Two days after replacement with a medium containing TGF-β1, the medium was removed, and using Trizol reagent (Invitrogen), total RNA was extracted according to the method described in the product. After measuring the concentration, PrimeScript 1st strand cDNA Synthesis Using Kit (manufactured by Takara Bio Inc.), cDNA was synthesized according to the method described in the product.
Using the prepared cDNA, the expression of fibrosis-related genes was analyzed by real-time RT-PCR. Prepare samples using the QuantiTect SYBR Green PCR Kit (QIAGEN) according to the method described in the product, and use the Applied Biosystems 7900HT Fast real-time PCR system (Applied Biosystems) to express αSMA, which is an EMT indicator Was quantified. As a result, as shown in Table 10, it was clarified that the expression of αSMA is reduced by introducing a molecule that generates let-7i (SEQ ID NO: 8). In addition, by introducing molecules that generate miR-194 (SEQ ID NO: 24), miR-146b (SEQ ID NO: 25), miR-21 (SEQ ID NO: 26), miR-223 (SEQ ID NO: 27), expression of αSMA Became clear.
Figure JPOXMLDOC01-appb-T000042

 
Figure JPOXMLDOC01-appb-T000042

 
microRNAを強制発現させたヒト肝星細胞の細胞外マトリックス産生に対する影響
 ヒト肝星細胞にmicroRNAの前駆体を導入し、該分子から生成されるmicroRNAがコラーゲン産生に及ぼす影響を調べた。肝臓の線維化、コラーゲン産生には肝星細胞が中心的な役割を担っているとされており、その線維化の指標として、collagen I等が用いられる[HEPATOLOGY, 34, 89-100 (2001)]。
 ヒト肝星細胞であるLi-90細胞(以下、Li-90と称す)は、20 %ウシ胎児血清(GIBCO社製)を含むDMEM培地(GIBCO社製)で37℃の5 % CO2濃度のインキュベーター中で培養した。
 Li-90を24穴プレートに1穴あたり1.5×104個となるように播種し、上記培地中で一晩培養した。1日後、miR-30b, 486, let-7iを生成するpre-miR miRNA precursor molecules (Ambion社製)をlipofectamine 2000(Invitrogen社製)を用いたリポフェクション法により終濃度が10 nMとなるようにLi-90に導入した。また、miRNA precursor molecules-Negative control (Ambion社製)も陰性コントロールとして導入した。リポフェクションは、製品に添付された説明書に記載された方法に従った。
 リポフェクション法により該分子を導入した8時間後、培地を血清を含まないDMEM培地に交換した。さらに16時間後、培地をrecombinant human TGF-β1(R&D社製)を終濃度1 ng / mlとなるように血清を含まないDMEM培地を用いて希釈したものへ交換し、さらに培養を続けた。
 TGF-β1を含んだ培地に交換した2日後、培地を除き、Trizol試薬(Invitrogen社製)を用いて、製品に記載された方法に従って全RNAを抽出し、濃度測定後、等量のRNAよりsuperscript VILO(Invitrogen社製)を用いて、製品に記載された方法に従って、cDNAを合成した。
 作製したcDNAを用いて、real-time RT-PCR法により線維化関連遺伝子の発現を解析した。QuantiTect SYBR Green PCR Kit(QIAGEN社製)を用いて製品に記載された方法に従ってサンプルを調製し、Applied Biosystems 7900HT Fast リアルタイムPCRシステム(Applied Biosystems社製)を用いてcollagen Iの発現量を定量した。陰性コントロールとして用いたmiRNA precursor molecules-Negative controlを導入した細胞におけるcollagen Iの発現量を1.00としてそれぞれのmicroRNAを導入した細胞におけるcollagen Iの発現量を計算した結果、表11に示すように、let-7i(配列番号8)、miR-486(配列番号14)を生成する分子を導入することにより、collagen Iの発現が低下することが明らかとなった。
Effect on extracellular matrix production of human hepatic stellate cells in which microRNA was forcibly expressed A microRNA precursor was introduced into human hepatic stellate cells, and the influence of microRNA produced from the molecule on collagen production was examined. Hepatic stellate cells are said to play a central role in liver fibrosis and collagen production, and collagen I and the like are used as an index of fibrosis [HEPATOLOGY, 34 , 89-100 (2001) ].
Li-90 cells (hereinafter referred to as Li-90), which are human hepatic stellate cells, are DMEM medium (GIBCO) containing 20% fetal calf serum (GIBCO) and a 5% CO 2 concentration at 37 ° C. Cultured in an incubator.
Li-90 was seeded in a 24-well plate at 1.5 × 10 4 per well and cultured overnight in the above medium. One day later, pre-miR miRNA precursor molecules (manufactured by Ambion) that produce miR-30b, 486, let-7i were Lipofected using lipofectamine 2000 (manufactured by Invitrogen) to a final concentration of 10 nM. -90 introduced. MiRNA precursor molecules-Negative control (Ambion) was also introduced as a negative control. Lipofection followed the method described in the instructions attached to the product.
Eight hours after the introduction of the molecule by the lipofection method, the medium was replaced with serum-free DMEM medium. After another 16 hours, the medium was replaced with recombinant human TGF-β1 (manufactured by R & D) diluted to a final concentration of 1 ng / ml using DMEM medium without serum, and the culture was continued.
Two days after changing to a medium containing TGF-β1, the medium was removed, and using Trizol reagent (Invitrogen), total RNA was extracted according to the method described in the product. cDNA was synthesized using superscript VILO (Invitrogen) according to the method described in the product.
Using the prepared cDNA, the expression of fibrosis-related genes was analyzed by real-time RT-PCR. Samples were prepared using the QuantiTect SYBR Green PCR Kit (QIAGEN) according to the method described in the product, and the amount of collagen I expression was quantified using an Applied Biosystems 7900HT Fast real-time PCR system (Applied Biosystems). As a result of calculating the expression level of collagen I in the cells into which each microRNA was introduced, assuming that the expression level of collagen I in the cells into which miRNA precursor molecules-Negative control used as negative control was introduced was 1.00, let It was revealed that the expression of collagen I is reduced by introducing molecules that generate -7i (SEQ ID NO: 8) and miR-486 (SEQ ID NO: 14).
Figure JPOXMLDOC01-appb-T000043

 
Figure JPOXMLDOC01-appb-T000043

 
microRNAを強制発現させた肺線維芽細胞の細胞外マトリックス産生に対する影響
 線維化が病態の進行の要因となっている臓器である、肺におけるmicroRNAの作用をin vitroの系で検討した。pre-miR miRNA precursor molecules(Ambion社製)をヒト肺線維芽細胞に導入し、該分子から生成されるmicroRNAがコラーゲン産生に及ぼす影響を調べた。肺の線維化には肺組織中の線維芽細胞が関与していることが知られており、その線維化の指標としては、collagen I等が用いられる[Respiration, 77, 311-319 (2009)]。
 ヒト肺線維芽細胞であるIMR-90細胞(以下、IMR-90と称す)は、10 %ウシ胎児血清(GIBCO社製)、2 mM L-glutamine, 0.1 mM NEAA, 1mM pyruvateを含むMEM培地(GIBCO社製)で37℃の5 % CO2濃度のインキュベーター中で培養した。
 IMR-90を24穴プレートに1穴あたり1.5×104となるように播種し、上記培地中で一晩培養した。1日後、miR-30b, 30c, 486, let-7iを生成するpre-miR miRNA precursor molecules をlipofectamine 2000 (Invitrogen社製)を用いたリポフェクション法により終濃度が10 nMとなるようにIMR-90に導入した。また、miRNA precursor molecules-Negative control(Ambion社製)も陰性コントロールとして導入した。リポフェクションは、製品に添付された説明書に記載された方法に従った。
 リポフェクション法により該分子を導入した8時間後、培地を血清を含まないMEM培地に交換した。さらに16時間後、培地をrecombinant human TGF-β1(R&D社製)を終濃度0.5 ng / ml となるように血清を含まないDMEM培地を用いて希釈したものへ交換し、さらに培養を続けた。
 TGF-β1を含んだ培地に交換した2日後、培地を除き、Trizol試薬(Invitrogen社製)を用いて、製品に記載された方法に従って全RNAを抽出し、濃度測定後、等量のRNAよりsuperscript VILO(Invitrogen社製)を用いて、製品に記載された方法に従って、cDNAを合成した。
 作製したcDNAを用いて、real-time RT-PCR法により線維化関連遺伝子の発現を解析した。QuantiTect SYBR Green PCR Kit(QIAGEN社製)を用いて製品に記載された方法に従ってサンプルを調製し、Applied Biosystems 7900HT Fast リアルタイムPCRシステム(Applied Biosystems社製)を用いてcollagen Iの発現量を定量した。その結果、表12に示すように、let-7i(配列番号8)、miR-30b(配列番号11)、miR30c(配列番号12)、miR-486(配列番号14)を生成する分子を導入することにより、collagen Iの発現が低下することが明らかとなった。
Effects of microRNA on forced extracellular matrix production of pulmonary fibroblasts We examined the action of microRNA in the lung, an organ in which fibrosis is a cause of disease progression. Pre-miR miRNA precursor molecules (manufactured by Ambion) were introduced into human lung fibroblasts, and the influence of microRNA generated from the molecules on collagen production was examined. It is known that fibroblasts in lung tissue are involved in lung fibrosis, and collagen I and the like are used as an index of the fibrosis [Respiration, 77 , 311-319 (2009) ].
Human lung fibroblast IMR-90 cells (hereinafter referred to as IMR-90) are prepared in MEM medium containing 10% fetal calf serum (GIBCO), 2 mM L-glutamine, 0.1 mM NEAA, 1 mM pyruvate ( (Manufactured by GIBCO) at 37 ° C. in an incubator with 5% CO 2 concentration.
IMR-90 was seeded in a 24-well plate at 1.5 × 10 4 per well and cultured overnight in the above medium. One day later, pre-miR miRNA precursor molecules that generate miR-30b, 30c, 486, and let-7i were added to IMR-90 by lipofection using lipofectamine 2000 (Invitrogen) to a final concentration of 10 nM. Introduced. MiRNA precursor molecules-Negative control (Ambion) was also introduced as a negative control. Lipofection followed the method described in the instructions attached to the product.
Eight hours after the introduction of the molecule by the lipofection method, the medium was replaced with a serum-free MEM medium. After another 16 hours, the medium was replaced with a recombinant human TGF-β1 (manufactured by R & D) diluted to a final concentration of 0.5 ng / ml using DMEM medium without serum, and the culture was further continued.
Two days after changing to a medium containing TGF-β1, the medium was removed, and using Trizol reagent (Invitrogen), total RNA was extracted according to the method described in the product. cDNA was synthesized using superscript VILO (Invitrogen) according to the method described in the product.
Using the prepared cDNA, the expression of fibrosis-related genes was analyzed by real-time RT-PCR. Samples were prepared using the QuantiTect SYBR Green PCR Kit (QIAGEN) according to the method described in the product, and the expression level of collagen I was quantified using an Applied Biosystems 7900HT Fast real-time PCR system (Applied Biosystems). As a result, as shown in Table 12, molecules that generate let-7i (SEQ ID NO: 8), miR-30b (SEQ ID NO: 11), miR30c (SEQ ID NO: 12), miR-486 (SEQ ID NO: 14) are introduced. This revealed that the expression of collagen I was reduced.
Figure JPOXMLDOC01-appb-T000044

 
Figure JPOXMLDOC01-appb-T000044

 
microRNAの発現を抑制した腎尿細管上皮細胞における細胞外マトリックス産生に対する影響
 microRNAを相補するアンチセンスオリゴヌクレオチドを細胞に導入することによるmicroRNAの発現低下が細胞外マトリックス産生に与える影響を調べた。microRNAを相補するアンチセンスオリゴヌクレオチドとしては、Anti-miR miRNA inhibitors(Ambion社製)を用い、該分子によって抑制されるmicroRNAがコラーゲン産生に及ぼす影響を調べた。
 NRK-52Eを24穴プレートに1穴あたり1×104となるように播種し、上記培地中で一晩培養した。1日後、miR-30c, 486, 320a, 378, 451を抑制するAnti-miR miRNA inhibitorsをlipofectamine 2000 (Invitrogen社製)を用いたリポフェクション法により終濃度が30 nMとなるようにNRK-52Eに導入した。また、Anti-miR miRNA Inhibitors-Negative Control #1 (Ambion社製)も陰性コントロールとして導入した。リポフェクションは、製品に添付された説明書に記載された方法に従った。
 リポフェクション法により該分子を導入した24時間後、培地をrecombinant human TGF-β1 (R&D 社製)を終濃度2 ng / mlとなるように5 %ウシ胎児血清を含むDMEM培地を用いて希釈したものへ交換し、さらに培養を続けた。
 TGF-β1を含んだ培地に交換した2日後、培地を除き、Trizol試薬(Invitrogen社製)を用いて、製品に記載された方法に従って全RNAを抽出し、濃度測定後、PrimeScript 1st strand cDNA Synthesis Kit(タカラバイオ社製)を用いて、製品に記載された方法に従って、cDNAを合成した。
 作製したcDNAを用いて、real-time RT-PCR法により線維化関連遺伝子の発現を解析した。QuantiTect SYBR Green PCR Kit(QIAGEN社製)を用いて製品に記載された方法に従ってサンプルを調製し、Applied Biosystems 7900HT FastリアルタイムPCRシステム(Applied Biosystems社製)を用いてcollagen Iの発現量を定量した。その結果、表13に示すように、miR-320a(配列番号2)、miR-378(配列番号3)、miR-30c(配列番号12)、miR-486(配列番号14), miR-451(配列番号18)の発現を抑制することにより、collagen Iの発現が上昇することが明らかとなった。
Effect on extracellular matrix production in renal tubular epithelial cells in which microRNA expression was suppressed The effect of reduced microRNA expression on extracellular matrix production by introducing antisense oligonucleotides complementary to microRNA into cells was investigated. Anti-miR miRNA inhibitors (manufactured by Ambion) were used as antisense oligonucleotides complementary to microRNA, and the influence of microRNA suppressed by the molecule on collagen production was examined.
NRK-52E was seeded in a 24-well plate at 1 × 10 4 per well and cultured overnight in the above medium. One day later, anti-miR miRNA inhibitors that inhibit miR-30c, 486, 320a, 378, and 451 were introduced into NRK-52E by lipofection using lipofectamine 2000 (Invitrogen) to a final concentration of 30 nM. did. Anti-miR miRNA Inhibitors-Negative Control # 1 (Ambion) was also introduced as a negative control. Lipofection followed the method described in the instructions attached to the product.
24 hours after introduction of the molecule by the lipofection method, the medium was diluted with DMEM medium containing 5% fetal calf serum to a final concentration of 2 ng / ml of recombinant human TGF-β1 (R & D) The culture was continued.
Two days after changing to a medium containing TGF-β1, remove the medium, extract the total RNA according to the method described in the product using Trizol reagent (Invitrogen), measure the concentration, then PrimeScript 1st strand cDNA Synthesis Using Kit (manufactured by Takara Bio Inc.), cDNA was synthesized according to the method described in the product.
Using the prepared cDNA, the expression of fibrosis-related genes was analyzed by real-time RT-PCR. Samples were prepared using the QuantiTect SYBR Green PCR Kit (QIAGEN) according to the method described in the product, and the expression level of collagen I was quantified using an Applied Biosystems 7900HT Fast real-time PCR system (Applied Biosystems). As a result, as shown in Table 13, miR-320a (SEQ ID NO: 2), miR-378 (SEQ ID NO: 3), miR-30c (SEQ ID NO: 12), miR-486 (SEQ ID NO: 14), miR-451 ( It was revealed that the expression of collagen I was increased by suppressing the expression of SEQ ID NO: 18).
Figure JPOXMLDOC01-appb-T000045

 
Figure JPOXMLDOC01-appb-T000045

 
microRNAの発現を抑制した腎線維芽細胞における細胞外マトリックス産生に対する影響
 microRNAを相補するアンチセンスオリゴヌクレオチドを細胞に導入することによるmicroRNAの発現低下が細胞外マトリックス産生に与える影響を調べた。microRNAを相補するアンチセンスオリゴヌクレオチドとしては、Anti-miR miRNA inhibitors(Ambion社製)を用い、該分子によって抑制されるmicroRNAがコラーゲン産生に及ぼす影響を調べた。
 NRK-49Fを24穴プレートに1穴あたり2×104となるように播種し、上記培地中で一晩培養した。1日後、miR-30c, 192, 320a, 378 , 451を抑制するAnti-miR miRNA inhibitorsをlipofectamine 2000 (Invitrogen社製)を用いたリポフェクション法により終濃度が30 nMとなるようにNRK-49Fに導入した。また、Anti-miR miRNA Inhibitors-Negative Control #1(Ambion社製)も陰性コントロールとして導入した。リポフェクションは、製品に添付された説明書に記載された方法に従った。また、let-7i,miR-486については、miRIDIAN microRNA Hairpin Inhibitor(Dharmacon社製)を、lipofectamine 2000を用いたリポフェクション法により終濃度が10 nMとなるようにNRK-49Fに導入した。また、miRIDIAN microRNA Hairpin Inhibitor Negative Control #2(Dharmacon社製)も陰性コントロールとして導入した。
 リポフェクション法により該分子を導入した8時間後、培地を血清を含まないDMEM培地に交換した。さらに16時間後、培地をrecombinant human TGF-β1(R&D社製)を終濃度1 ng/ mlとなるように血清を含まないDMEM培地を用いて希釈したものへ交換し、さらに培養を続けた。
 TGF-β1を含んだ培地に交換した2日後、培地を除き、Trizol試薬(Invitrogen社製)を用いて、製品に記載された方法に従って全RNAを抽出し、濃度測定後、PrimeScript 1st strand cDNA Synthesis Kit(タカラバイオ社製)を用いて、製品に記載された方法に従って、cDNAを合成した。
 作製したcDNAを用いて、real-time RT-PCR法により線維化関連遺伝子の発現を解析した。QuantiTect SYBR Green PCR Kit(QIAGEN社製)を用いて製品に記載された方法に従ってサンプルを調製し、Applied Biosystems 7900HT FastリアルタイムPCRシステム(Applied Biosystems社製)を用いてcollagen Iの発現量を定量した。その結果、表14に示すように、miR-320a(配列番号2)、miR-378(配列番号3)、miR-192(配列番号6)、miR-30c(配列番号12)、miR-486(配列番号14)、miR-451(配列番号18)、let-7i(配列番号8)の発現を抑制することにより、collagen Iの発現が上昇することが明らかとなった。
Effect on extracellular matrix production in renal fibroblasts with suppressed expression of microRNA We investigated the effect of reduced expression of microRNA on extracellular matrix production by introducing antisense oligonucleotides complementary to microRNA into cells. Anti-miR miRNA inhibitors (manufactured by Ambion) were used as antisense oligonucleotides complementary to microRNA, and the influence of microRNA suppressed by the molecule on collagen production was examined.
NRK-49F was seeded in a 24-well plate at 2 × 10 4 per well and cultured overnight in the above medium. One day later, anti-miR miRNA inhibitors that inhibit miR-30c, 192, 320a, 378, and 451 were introduced into NRK-49F by lipofection using lipofectamine 2000 (Invitrogen) to a final concentration of 30 nM. did. Anti-miR miRNA Inhibitors-Negative Control # 1 (Ambion) was also introduced as a negative control. Lipofection followed the method described in the instructions attached to the product. For let-7i and miR-486, miRIDIAN microRNA Hairpin Inhibitor (manufactured by Dharmacon) was introduced into NRK-49F by a lipofection method using lipofectamine 2000 to a final concentration of 10 nM. MiRIDIAN microRNA Hairpin Inhibitor Negative Control # 2 (Dharmacon) was also introduced as a negative control.
Eight hours after the introduction of the molecule by the lipofection method, the medium was replaced with serum-free DMEM medium. After another 16 hours, the medium was replaced with a recombinant human TGF-β1 (manufactured by R & D) diluted to a final concentration of 1 ng / ml using DMEM medium without serum, and the culture was further continued.
Two days after replacement with a medium containing TGF-β1, the medium was removed, and using Trizol reagent (Invitrogen), total RNA was extracted according to the method described in the product. After measuring the concentration, PrimeScript 1st strand cDNA Synthesis Using Kit (manufactured by Takara Bio Inc.), cDNA was synthesized according to the method described in the product.
Using the prepared cDNA, the expression of fibrosis-related genes was analyzed by real-time RT-PCR. Samples were prepared using the QuantiTect SYBR Green PCR Kit (QIAGEN) according to the method described in the product, and the expression level of collagen I was quantified using an Applied Biosystems 7900HT Fast real-time PCR system (Applied Biosystems). As a result, as shown in Table 14, miR-320a (SEQ ID NO: 2), miR-378 (SEQ ID NO: 3), miR-192 (SEQ ID NO: 6), miR-30c (SEQ ID NO: 12), miR-486 ( It was revealed that the expression of collagen I was increased by suppressing the expression of SEQ ID NO: 14), miR-451 (SEQ ID NO: 18), and let-7i (SEQ ID NO: 8).
Figure JPOXMLDOC01-appb-T000046

 
Figure JPOXMLDOC01-appb-T000046

 
microRNAの発現を抑制させた腎尿細管上皮細胞の上皮間葉転換 (EMT) に対する影響
 microRNAを相補するアンチセンスオリゴヌクレオチドを細胞に導入することによるmicroRNAの発現低下がEMTに及ぼす影響を調べた。
 MDCKを24穴プレートに1穴あたり1×104となるように播種し、上記培地中で一晩培養した。1日後、miR-320a, 378, 192, 30c, 486, 451を抑制するAnti-miR miRNA inhibitorsをlipofectamine 2000(Invitrogen社製)を用いたリポフェクション法により終濃度が30 nMとなるようにMDCKに導入した。また、Anti-miR miRNA Inhibitors -Negative Control #1(Ambion社製)も陰性コントロールとして導入した。リポフェクションは、製品に添付された説明書に記載された方法に従った。
 リポフェクション法により該分子を導入した24時間後、培地をrecombinant human TGF-β1 (R&D社製)を終濃度2 ng / mlとなるように10 %ウシ胎児血清を含むDMEM培地を用いて希釈したものへ交換し、さらに培養を続けた。
 TGF-β1を含んだ培地に交換した2日後、培地を除き、Trizol試薬(Invitrogen社製)を用いて、製品に記載された方法に従って全RNAを抽出し、濃度測定後、PrimeScript 1st strand cDNA Synthesis Kit(タカラバイオ社製)を用いて、製品に記載された方法に従って、cDNAを合成した。
 作製したcDNAを用いて、real-time RT-PCR法により線維化関連遺伝子の発現を解析した。QuantiTect SYBR Green PCR Kit(QIAGEN社製)を用いて製品に記載された方法に従ってサンプルを調製し、Applied Biosystems 7900HT FastリアルタイムPCRシステム(Applied Biosystems社製)を用いてEMTの指標であるαSMAの発現量を定量した。その結果、表15に示すように、miR-320a(配列番号2)、miR-378(配列番号3)、miR-192(配列番号6)、miR-30c(配列番号12)、miR-486(配列番号14)、miR-451(配列番号18)の発現を抑制することにより、αSMAの発現が上昇することが明らかとなった。
Effects on renal epithelial-mesenchymal transition (EMT) of renal tubular epithelial cells with suppressed microRNA expression The effect of microRNA expression reduction on EMT by introduction of antisense oligonucleotide complementary to microRNA was examined.
MDCK was seeded in a 24-well plate at 1 × 10 4 per well and cultured overnight in the above medium. One day later, anti-miR miRNA inhibitors that inhibit miR-320a, 378, 192, 30c, 486, and 451 were introduced into MDCK to a final concentration of 30 nM by lipofection using lipofectamine 2000 (Invitrogen). did. Anti-miR miRNA Inhibitors-Negative Control # 1 (Ambion) was also introduced as a negative control. Lipofection followed the method described in the instructions attached to the product.
24 hours after introduction of the molecule by the lipofection method, the medium was diluted with DMEM medium containing 10% fetal bovine serum to a final concentration of 2 ng / ml of recombinant human TGF-β1 (R & D). The culture was continued.
Two days after replacement with a medium containing TGF-β1, the medium was removed, and using Trizol reagent (Invitrogen), total RNA was extracted according to the method described in the product. After measuring the concentration, PrimeScript 1st strand cDNA Synthesis Using Kit (manufactured by Takara Bio Inc.), cDNA was synthesized according to the method described in the product.
Using the prepared cDNA, the expression of fibrosis-related genes was analyzed by real-time RT-PCR. Prepare samples using the QuantiTect SYBR Green PCR Kit (QIAGEN) according to the method described in the product, and use the Applied Biosystems 7900HT Fast real-time PCR system (Applied Biosystems) to express αSMA, which is an EMT indicator Was quantified. As a result, as shown in Table 15, miR-320a (SEQ ID NO: 2), miR-378 (SEQ ID NO: 3), miR-192 (SEQ ID NO: 6), miR-30c (SEQ ID NO: 12), miR-486 ( It was revealed that the expression of αSMA was increased by suppressing the expression of SEQ ID NO: 14) and miR-451 (SEQ ID NO: 18).
Figure JPOXMLDOC01-appb-T000047

 
Figure JPOXMLDOC01-appb-T000047

 
microRNA投与による臓器線維化に対する影響
 microRNAの投与が臓器の線維化に影響を与えるかを調べるために、肝線維化モデルに対し3種のmicroRNAそれぞれを投与して評価を実施した。評価したmicroRNAはmiR-30b、miR-486、let-7iである。microRNAとして、miR-30bは、配列番号11で表される塩基配列からなる核酸(hsa-miR-30b)をアンチセンス鎖とし、そのアンチセンス鎖の5'側1塩基目から19塩基目に相補的な配列にTTオーバーハング配列を付加した、配列番号1282で表される塩基配列からなる核酸をセンス鎖として、それぞれ合成してアニーリングさせたものを用いた。miR-486は、配列番号14で表される塩基配列からなるアンチセンス鎖と、配列番号1283で表される塩基配列からなるセンス鎖とをアニーリングさせたものを用い、let-7iは配列番号8で表される塩基配列からなるアンチセンス鎖と、配列番号1284で表される塩基配列からなるセンス鎖とをアニーリングさせたものを用いた。陰性コントロールには21merからなるルシフェラーゼ siRNAを用いた。各RNAは北海道システムサイエンス社にて合成し、肝臓への送達効率が高い脂質粒子であるStable nucleic acid lipid particle (SNALP)に、Mol Ther., 13(3), 494-505 (2006)に記載された方法に従って抱合したのち、評価に用いた。
 肝線維化には一般的な動物モデルとして知られる胆管結紮(Bile Duct Ligation : BDL)モデルを用いた[Hepatology, 50, 185-197 (2009)、Am J Physiol Gastrointest Liver Physiol, 296, G582-G592 (2009)]。
 9週齢のオスC57BL/6Jマウス(チャールズリバージャパン社より購入)の胆管を麻酔下で結紮し、手術直後と24時間後にそれぞれ150μg/headの用量でSNALP抱合体のマイクロRNAを各群7匹に投与した。その後48時間後に肝臓を摘出し、液体窒素にて速やかに凍結した。凍結肝臓はTrizol試薬(Invitrogen社製)を用いて破砕し、クロロホルム(和光純薬社製)を添加して遠心した後、その上清からRNeasy kit(QIAGEN 社製)を用いて、製品に記載された方法に従って全RNAを抽出した。全RNAは濃度測定後、Super Script VILOTMcDNA Synthesis Kit(Invitrogen社製)を用いて、製品に記載された方法に従って、cDNAを合成した。作製したcDNAを用いて、real-time RT-PCR法により線維化関連遺伝子の発現を解析した。QuantiTect SYBR Green PCR Kit(QIAGEN社製)を用いて製品に記載された方法に従ってサンプルを調製し、Applied Biosystems 7900HT FastリアルタイムPCRシステム(Applied Biosystems社製)を用いて線維化の指標であるcollagen IおよびTGF-β1の発現量を定量した。その結果、miR-30b(配列番号11)、miR-486(配列番号14)、let-7i(配列番号8)を投与することにより、collagen I(図1)およびTGF-β1(図2)の発現は、それぞれ有意に抑制されることが明らかとなった。
Effects of microRNA administration on organ fibrosis In order to examine whether microRNA administration affects organ fibrosis, we evaluated each of the three microRNAs administered to the liver fibrosis model. The microRNAs evaluated were miR-30b, miR-486, and let-7i. As a microRNA, miR-30b uses a nucleic acid (hsa-miR-30b) consisting of the base sequence represented by SEQ ID NO: 11 as an antisense strand, and is complementary to the first base to the 19th base on the 5 ′ side of the antisense strand. A nucleic acid consisting of the base sequence represented by SEQ ID NO: 1282, in which a TT overhang sequence was added to a typical sequence, was synthesized as a sense strand and annealed. miR-486 is obtained by annealing an antisense strand consisting of the base sequence represented by SEQ ID NO: 14 and a sense strand consisting of the base sequence represented by SEQ ID NO: 1283, and let-7i is SEQ ID NO: 8 An antisense strand consisting of the base sequence represented by the above and an annealed sense strand consisting of the base sequence represented by SEQ ID NO: 1284 were used. As a negative control, 21mer luciferase siRNA was used. Each RNA was synthesized by Hokkaido System Science and described in Stable nucleic acid lipid particle (SNALP), a lipid particle with high delivery efficiency to the liver, described in Mol Ther., 13 (3) , 494-505 (2006). After conjugation according to the established method, it was used for evaluation.
Hepatology, 50 , 185-197 (2009), Am J Physiol Gastrointest Liver Physiol, 296 , G582-G592 using the Bile Duct Ligation (BDL) model known as a general animal model for liver fibrosis (2009)].
Bile ducts from 9-week-old male C57BL / 6J mice (purchased from Charles River Japan) were ligated under anesthesia and 7 SNALP conjugate microRNAs were administered at a dose of 150 μg / head immediately after surgery and 24 hours later, respectively. Administered. Thereafter, the liver was removed 48 hours later and quickly frozen in liquid nitrogen. Frozen liver is crushed using Trizol reagent (Invitrogen), added with chloroform (Wako Pure Chemicals) and centrifuged, and then the supernatant is described in the product using RNeasy kit (QIAGEN) Total RNA was extracted according to the method described. After measuring the concentration of total RNA, cDNA was synthesized using Super Script VILO cDNA Synthesis Kit (manufactured by Invitrogen) according to the method described in the product. Using the prepared cDNA, the expression of fibrosis-related genes was analyzed by real-time RT-PCR. Prepare samples using the QuantiTect SYBR Green PCR Kit (manufactured by QIAGEN) according to the method described in the product, and use the Applied Biosystems 7900HT Fast real-time PCR system (manufactured by Applied Biosystems) to identify collagen I and fibrosis indicators. The expression level of TGF-β1 was quantified. As a result, by administering miR-30b (SEQ ID NO: 11), miR-486 (SEQ ID NO: 14), let-7i (SEQ ID NO: 8), collagen I (FIG. 1) and TGF-β1 (FIG. 2) It was revealed that the expression was significantly suppressed.
 本発明により、細胞または臓器の線維化制御剤、細胞または臓器の線維化に起因する疾患の診断薬または治療薬、マイクロRNAなどの核酸の標的遺伝子の発現制御剤、細胞または臓器の制御方法、並びに細胞または臓器の線維化制御剤のスクリーニング方法等が提供され、これらは細胞または臓器の線維化に起因する疾患の予防、診断または治療等において有用である。この場合、制御とは抑制および促進を指す。 According to the present invention, an agent for controlling fibrosis of cells or organs, a diagnostic or therapeutic agent for diseases caused by fibrosis of cells or organs, an agent for controlling expression of target genes of nucleic acids such as microRNA, a method for controlling cells or organs, In addition, screening methods for cell or organ fibrosis control agents and the like are provided, which are useful in the prevention, diagnosis or treatment of diseases caused by cell or organ fibrosis. In this case, control refers to suppression and promotion.
 本出願は日本で出願された特願2009-187563(出願日:2009年8月12日)を基礎としており、その内容は本明細書に全て包含されるものである。 This application is based on Japanese Patent Application No. 2009-187563 filed in Japan (filing date: August 12, 2009), the contents of which are incorporated in full herein.

Claims (31)

  1. 以下の(a)~(h)のいずれかの核酸を有効成分として含有する、細胞または臓器の線維化制御剤。
    (a)配列番号1~27、28~594のいずれかで表される塩基配列からなる核酸
    (b)配列番号1~27、28~594のいずれかで表される塩基配列からなる核酸を含有する、17~28塩基の核酸
    (c)配列番号1~27、28~594のいずれかで表される塩基配列と90%以上の同一性を有する塩基配列からなる核酸
    (d)配列番号1~27、28~594のいずれかで表される塩基配列からなる核酸の相補鎖とストリンジェントな条件でハイブリダイズする核酸
    (e)配列番号1~27、28~594のいずれかで表される塩基配列の2~8番目の塩基配列を含む核酸
    (f)配列番号595~1281のいずれかで表される塩基配列からなる核酸
    (g)配列番号595~1281のいずれかで表される塩基配列と90%以上の同一性を有する塩基配列からなる核酸
    (h)配列番号595~1281のいずれかで表される塩基配列からなる核酸の相補鎖とストリンジェントな条件でハイブリダイズする核酸
    An agent for controlling fibrosis of cells or organs, which contains any of the following nucleic acids (a) to (h) as an active ingredient.
    (A) Nucleic acid comprising a base sequence represented by any of SEQ ID NOs: 1-27, 28-594 (b) Containing a nucleic acid comprising a base sequence represented by any of SEQ ID NOs: 1-27, 28-594 A nucleic acid consisting of a base sequence having a nucleotide sequence of 90% or more (d) a sequence number 1 to 1 to a nucleic acid of 17 to 28 bases (c) a sequence represented by any one of SEQ ID NOs: 1 to 27 and 28 to 594 A nucleic acid that hybridizes under stringent conditions with a complementary strand of a nucleic acid comprising the nucleotide sequence represented by any one of 27 and 28 to 594 (e) a base represented by any one of SEQ ID NOs: 1 to 27 and 28 to 594 A nucleic acid comprising the second to eighth base sequences of the sequence (f) a nucleic acid comprising the base sequence represented by any one of SEQ ID NOs: 595 to 1281 (g) a base sequence represented by any one of the sequence numbers 595 to 1281; 90% or more identity (H) a nucleic acid that hybridizes under stringent conditions with a complementary strand of a nucleic acid consisting of the base sequence represented by any of SEQ ID NOs: 595 to 1281
  2. 核酸がマイクロRNAまたはマイクロRNA前駆体である、請求項1に記載の細胞または臓器の線維化制御剤。 The cell or organ fibrosis regulator according to claim 1, wherein the nucleic acid is microRNA or a microRNA precursor.
  3. 請求項1に記載の核酸の塩基配列に対して相補的な塩基配列からなる核酸を有効成分として含有する、細胞または臓器の線維化制御剤。 A cell or organ fibrosis control agent comprising, as an active ingredient, a nucleic acid having a base sequence complementary to the base sequence of the nucleic acid according to claim 1.
  4. 請求項1~3のいずれか1項に記載の細胞または臓器の線維化制御剤の有効成分である核酸を発現するベクターを有効成分として含有する、細胞またはおよび臓器の線維化制御剤。 A cell or organ fibrosis control agent comprising, as an active ingredient, a vector that expresses a nucleic acid that is an active ingredient of the cell or organ fibrosis control agent according to any one of claims 1 to 3.
  5. 請求項1に記載の核酸の標的塩基配列を有する遺伝子の発現を抑制する物質を有効成分として含有する、細胞または臓器の線維化制御剤。 A cell or organ fibrosis control agent comprising a substance that suppresses the expression of a gene having the target base sequence of the nucleic acid according to claim 1 as an active ingredient.
  6. 請求項1に記載の核酸の標的塩基配列を有する遺伝子の発現を促進する物質を有効成分として含有する、細胞またはおよび臓器の線維化制御剤。 A cell or organ fibrosis control agent comprising a substance that promotes the expression of a gene having the target base sequence of the nucleic acid according to claim 1 as an active ingredient.
  7. 発現を抑制または促進する物質が核酸である、請求項5または6に記載の細胞または臓器の線維化制御剤。 The cell or organ fibrosis regulator according to claim 5 or 6, wherein the substance that suppresses or promotes expression is a nucleic acid.
  8. 核酸がsiRNAまたはアンチセンスオリゴヌクレオチドである、請求項7に記載の細胞または臓器の線維化制御剤。 The cell or organ fibrosis regulator of claim 7, wherein the nucleic acid is siRNA or antisense oligonucleotide.
  9. 請求項7に記載の核酸を発現するベクターを有効成分として含有する、細胞または臓器の線維化制御剤。 A cell or organ fibrosis control agent comprising the vector expressing the nucleic acid according to claim 7 as an active ingredient.
  10. 請求項1~3のいずれか1項に記載の核酸、請求項4に記載のベクター、または請求項5~8のいずれか1項に記載の物質を有効成分として含有する、細胞または臓器の線維化に起因する疾患の治療薬または診断薬。 A cell or organ fiber containing the nucleic acid according to any one of claims 1 to 3, the vector according to claim 4, or the substance according to any one of claims 5 to 8 as an active ingredient. Therapeutic or diagnostic agent for diseases caused by oxidization.
  11. 請求項1に記載の核酸の発現量、該核酸の変異、または該核酸をコードするゲノムの変異を検出する試薬を有効成分として含有する、細胞または臓器の線維化に起因する疾患の診断薬。 A diagnostic agent for a disease caused by fibrosis of a cell or organ, comprising as an active ingredient a reagent for detecting the expression level of the nucleic acid, the mutation of the nucleic acid, or the mutation of the genome encoding the nucleic acid.
  12. 細胞または臓器の線維化に起因する疾患が、急性腎不全、糸球体腎炎、血管炎、糖尿病性腎症、高血圧性腎硬化症、HIV腎症、IgA腎症、ループス腎炎、間質性腎炎、尿管閉塞による閉塞腎、肺線維症、肝硬変、動脈硬化症、強皮症、経皮経管冠動脈血管拡張術(PTCA)後の冠動脈再狭窄、間質性心筋炎、間質性膀胱炎、熱傷後の皮膚瘢痕化、または中毒に伴う線維症である、請求項10または11に記載の治療薬または診断薬。 Diseases resulting from cell or organ fibrosis are acute renal failure, glomerulonephritis, vasculitis, diabetic nephropathy, hypertensive nephrosclerosis, HIV nephropathy, IgA nephropathy, lupus nephritis, interstitial nephritis, Obstructed kidney due to ureteral obstruction, pulmonary fibrosis, cirrhosis, arteriosclerosis, scleroderma, coronary restenosis after percutaneous transluminal coronary vasodilation (PTCA), interstitial myocarditis, interstitial cystitis, The therapeutic or diagnostic agent according to claim 10 or 11, which is scarring after burn or fibrosis associated with poisoning.
  13. 細胞または臓器の線維化に起因する疾患が、急性腎不全、糸球体腎炎、血管炎、糖尿病性腎症、高血圧性腎硬化症、HIV腎症、IgA腎症、ループス腎炎、間質性腎炎、および尿管閉塞による閉塞腎からなる群から選択される腎障害に伴う線維症である、請求項10または11に記載の治療薬または診断薬。 Diseases resulting from cell or organ fibrosis are acute renal failure, glomerulonephritis, vasculitis, diabetic nephropathy, hypertensive nephrosclerosis, HIV nephropathy, IgA nephropathy, lupus nephritis, interstitial nephritis, The therapeutic or diagnostic agent according to claim 10 or 11, which is a fibrosis associated with a renal disorder selected from the group consisting of obstructive kidneys due to ureteral obstruction.
  14. 請求項1~3のいずれか1項に記載の核酸、請求項4に記載のベクター、または請求項5~8のいずれか1項に記載の物質を有効量投与することを特徴とする、細胞または臓器の線維化に起因する疾患の治療方法。 A cell characterized by administering an effective amount of the nucleic acid according to any one of claims 1 to 3, the vector according to claim 4, or the substance according to any one of claims 5 to 8. Alternatively, a method for treating a disease caused by organ fibrosis.
  15. 請求項1に記載の核酸の発現量、該核酸の変異、または該核酸をコードするゲノムの変異を検出することを特徴とする、細胞または臓器の線維化に起因する疾患の診断方法。 A method for diagnosing a disease caused by fibrosis of a cell or organ, comprising detecting the expression level of the nucleic acid according to claim 1, a mutation of the nucleic acid, or a mutation of a genome encoding the nucleic acid.
  16. 細胞または臓器の線維化に起因する疾患が、急性腎不全、糸球体腎炎、血管炎、糖尿病性腎症、高血圧性腎硬化症、HIV腎症、IgA腎症、ループス腎炎、間質性腎炎、尿管閉塞による閉塞腎、肺線維症、肝硬変、動脈硬化症、強皮症、経皮経管冠動脈血管拡張術(PTCA)後の冠動脈再狭窄、間質性心筋炎、間質性膀胱炎、熱傷後の皮膚瘢痕化、または中毒に伴う線維症である、請求項14または15に記載の治療または診断方法。 Diseases resulting from cell or organ fibrosis are acute renal failure, glomerulonephritis, vasculitis, diabetic nephropathy, hypertensive nephrosclerosis, HIV nephropathy, IgA nephropathy, lupus nephritis, interstitial nephritis, Obstructed kidney due to ureteral obstruction, pulmonary fibrosis, cirrhosis, arteriosclerosis, scleroderma, coronary restenosis after percutaneous transluminal coronary vasodilation (PTCA), interstitial myocarditis, interstitial cystitis, The method of treatment or diagnosis according to claim 14 or 15, wherein the skin scarring after burns or fibrosis associated with poisoning.
  17. 細胞または臓器の線維化に起因する疾患が、急性腎不全、糸球体腎炎、血管炎、糖尿病性腎症、高血圧性腎硬化症、HIV腎症、IgA腎症、ループス腎炎、間質性腎炎、および尿管閉塞による閉塞腎からなる群から選択される腎障害に伴う線維症である、請求項16に記載の治療または診断方法。 Diseases resulting from cell or organ fibrosis are acute renal failure, glomerulonephritis, vasculitis, diabetic nephropathy, hypertensive nephrosclerosis, HIV nephropathy, IgA nephropathy, lupus nephritis, interstitial nephritis, The treatment or diagnosis method according to claim 16, which is a fibrosis associated with renal disorder selected from the group consisting of obstructive kidneys due to ureteral obstruction.
  18. 細胞または臓器の線維化に起因する疾患の治療薬の製造のための、請求項1~3のいずれか1項に記載の核酸、請求項4に記載のベクター、または請求項5~8のいずれか1項に記載の物質の使用。 The nucleic acid according to any one of claims 1 to 3, the vector according to claim 4, or the vector according to any one of claims 5 to 8 for the manufacture of a therapeutic agent for a disease caused by fibrosis of cells or organs. Use of the substance according to claim 1.
  19. 細胞または臓器の線維化に起因する疾患が、急性腎不全、糸球体腎炎、血管炎、糖尿病性腎症、高血圧性腎硬化症、HIV腎症、IgA腎症、ループス腎炎、間質性腎炎、尿管閉塞による閉塞腎、肺線維症、肝硬変、動脈硬化症、強皮症、経皮経管冠動脈血管拡張術(PTCA)後の冠動脈再狭窄、間質性心筋炎、間質性膀胱炎、熱傷後の皮膚瘢痕化、または中毒に伴う線維症である、請求項18に記載の使用。 Diseases resulting from cell or organ fibrosis are acute renal failure, glomerulonephritis, vasculitis, diabetic nephropathy, hypertensive nephrosclerosis, HIV nephropathy, IgA nephropathy, lupus nephritis, interstitial nephritis, Obstructed kidney due to ureteral obstruction, pulmonary fibrosis, cirrhosis, arteriosclerosis, scleroderma, coronary restenosis after percutaneous transluminal coronary vasodilation (PTCA), interstitial myocarditis, interstitial cystitis, 19. Use according to claim 18, wherein the skin scarring after burns or fibrosis associated with poisoning.
  20. 細胞または臓器の線維化に起因する疾患が、急性腎不全、糸球体腎炎、血管炎、糖尿病性腎症、高血圧性腎硬化症、HIV腎症、IgA腎症、ループス腎炎、間質性腎炎、および尿管閉塞による閉塞腎からなる群から選択される腎障害に伴う線維症である、請求項18に記載の使用。 Diseases resulting from cell or organ fibrosis are acute renal failure, glomerulonephritis, vasculitis, diabetic nephropathy, hypertensive nephrosclerosis, HIV nephropathy, IgA nephropathy, lupus nephritis, interstitial nephritis, The use according to claim 18, which is a fibrosis associated with renal injury selected from the group consisting of obstructed kidneys due to ureteral obstruction.
  21. 請求項1~3のいずれか1項に記載の核酸を有効成分として含有する、細胞または臓器の線維化制御剤の有効成分である核酸の標的遺伝子の発現制御剤。 An expression control agent for a target gene of a nucleic acid, which is an active ingredient of a cell or organ fibrosis control agent, comprising the nucleic acid according to any one of claims 1 to 3 as an active ingredient.
  22. 請求項4に記載のベクターを有効成分として含有する、細胞または臓器の線維化制御剤の有効成分である核酸の標的遺伝子の発現制御剤。 An agent for controlling expression of a target gene of a nucleic acid, which is an active ingredient of a cell or organ fibrosis regulator, comprising the vector according to claim 4 as an active ingredient.
  23. 請求項1~3のいずれか1項に記載の核酸を用いることを特徴とする、細胞または臓器の線維化制御方法。 A method for controlling fibrosis of a cell or organ, characterized by using the nucleic acid according to any one of claims 1 to 3.
  24. 請求項4に記載のベクターを用いることを特徴とする、細胞または臓器の線維化制御方法。 A method for controlling fibrosis of a cell or organ, comprising using the vector according to claim 4.
  25. 請求項1に記載の核酸の標的遺伝子の発現を抑制または促進する物質を用いることを特徴とする、細胞または臓器の線維化制御方法。 A method for controlling fibrosis of a cell or organ, comprising using a substance that suppresses or promotes the expression of a target gene of the nucleic acid according to claim 1.
  26. 発現を抑制または促進する物質が核酸である、請求項25に記載の細胞または臓器の線維化制御方法。 26. The method for controlling fibrosis of a cell or organ according to claim 25, wherein the substance that suppresses or promotes expression is a nucleic acid.
  27. 核酸がsiRNAまたはアンチセンスオリゴヌクレオチドである、請求項26に記載の細胞または臓器の線維化制御方法。 27. The method for controlling fibrosis of a cell or organ according to claim 26, wherein the nucleic acid is siRNA or antisense oligonucleotide.
  28. 請求項26に記載の核酸を発現するベクターを用いる、細胞または臓器の線維化制御方法。 A method for controlling fibrosis of a cell or organ using the vector expressing the nucleic acid according to claim 26.
  29. 請求項1~3のいずれか1項に記載の核酸を用いることを特徴とする、細胞または臓器の線維化制御剤の有効成分である核酸の標的遺伝子の発現制御方法。 A method for controlling the expression of a target gene of a nucleic acid that is an active ingredient of a fibrosis control agent for cells or organs, wherein the nucleic acid according to any one of claims 1 to 3 is used.
  30. 請求項4に記載のベクターを用いることを特徴とする、細胞または臓器の線維化制御剤の有効成分である核酸の標的遺伝子の発現制御方法。 A method for controlling the expression of a target gene of a nucleic acid, which is an active ingredient of a cell or organ fibrosis regulator, characterized by using the vector according to claim 4.
  31. 請求項1に記載の核酸の発現または機能を促進または抑制させることを指標とする、細胞または臓器の線維化制御剤のスクリーニング方法。 A method for screening a fibrosis control agent for cells or organs, wherein the index is to promote or suppress the expression or function of the nucleic acid according to claim 1.
PCT/JP2010/063728 2009-08-12 2010-08-12 Nucleic acid that controls fibrosis of cells or organs WO2011019074A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2009-187563 2009-08-12
JP2009187563 2009-08-12

Publications (1)

Publication Number Publication Date
WO2011019074A1 true WO2011019074A1 (en) 2011-02-17

Family

ID=43586248

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2010/063728 WO2011019074A1 (en) 2009-08-12 2010-08-12 Nucleic acid that controls fibrosis of cells or organs

Country Status (1)

Country Link
WO (1) WO2011019074A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2011217617A (en) * 2010-04-05 2011-11-04 Kyoto Univ EXAMINATION METHOD OF FIBROSIS IN CHRONIC LIVER DISEASE USING miRNA
CN109852597A (en) * 2019-03-21 2019-06-07 云南师范大学 A kind of beta galactosidase galRBM20_1 and its preparation method and application
JP2019187413A (en) * 2018-04-23 2019-10-31 チャン グァン メモリアル ホスピタル,カオシュン Methods for detecting lupus nephritis or predicting its risk and applications thereof
WO2019246499A1 (en) * 2018-06-21 2019-12-26 Arizona Board Of Regents On Behalf Of The University Of Arizona Systems and methods for determining a treatment course of action
CN114015763A (en) * 2021-10-13 2022-02-08 深圳市人民医院 Application of miR-378-5p in mesangial proliferative glomerulonephritis
WO2022219404A1 (en) * 2021-04-13 2022-10-20 Oslo Universitetssykehus Hf Gene therapy for inflammatory conditions

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009018493A1 (en) * 2007-07-31 2009-02-05 The Board Of Regents Of The University Of Texas System A micro-rna family that modulates fibrosis and uses thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009018493A1 (en) * 2007-07-31 2009-02-05 The Board Of Regents Of The University Of Texas System A micro-rna family that modulates fibrosis and uses thereof

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
DUISTERS, R.F. ET AL.: "miR-133 and miR-30 regulate connective tissue growth factor: implications for a role of microRNAs in myocardial matrix remodeling", CIRC RES, vol. 104, 18 December 2008 (2008-12-18), pages 170 - 178, XP008102625, DOI: doi:10.1161/CIRCRESAHA.108.182535 *
KATO, M. ET AL.: "TGF-beta activates Akt kinase through a microRNA-dependent amplifying circuit targeting PTEN", NAT CELL BIOL, vol. 11, 21 June 2009 (2009-06-21), pages 881 - 889, XP055026246, DOI: doi:10.1038/ncb1897 *
LI, M. ET AL.: "MicroRNAs: control and loss of control in human physiology and disease", WORLD J SURG, vol. 33, no. 4, 22 November 2008 (2008-11-22), pages 667 - 684, XP019707084 *
LI, Z. ET AL.: "Biological functions of miR-29b contribute to positive regulation of osteoblast differentiation", J BIOL CHEM, vol. 284, 2 April 2009 (2009-04-02), pages 15676 - 15684, XP055038948, DOI: doi:10.1074/jbc.M809787200 *
ROOIJ, E.V. ET AL.: "Dysregulation of microRNAs after myocardial infarction reveals a role of miR-29 in cardiac fibrosis", PROC NATL ACAD SCI U S A, vol. 105, 22 August 2008 (2008-08-22), pages 13027 - 13032, XP002571306, DOI: doi:10.1073/pnas.0805038105 *
ROOIJ, E.V. ET AL.: "Searching for miR-acles in cardiac fibrosis", CIRC RES, vol. 104, 30 January 2009 (2009-01-30), pages 138 - 140, XP002591350, DOI: doi:10.1161/CIRCRESAHA.108.192492 *
THUM, T. ET AL.: "MicroRNA-21 contributes to myocardial disease by stimulating MAP kinase signalling in fibroblasts", NATURE, vol. 456, 30 November 2008 (2008-11-30), pages 980 - 984 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2011217617A (en) * 2010-04-05 2011-11-04 Kyoto Univ EXAMINATION METHOD OF FIBROSIS IN CHRONIC LIVER DISEASE USING miRNA
JP2019187413A (en) * 2018-04-23 2019-10-31 チャン グァン メモリアル ホスピタル,カオシュン Methods for detecting lupus nephritis or predicting its risk and applications thereof
WO2019246499A1 (en) * 2018-06-21 2019-12-26 Arizona Board Of Regents On Behalf Of The University Of Arizona Systems and methods for determining a treatment course of action
CN109852597A (en) * 2019-03-21 2019-06-07 云南师范大学 A kind of beta galactosidase galRBM20_1 and its preparation method and application
WO2022219404A1 (en) * 2021-04-13 2022-10-20 Oslo Universitetssykehus Hf Gene therapy for inflammatory conditions
CN114015763A (en) * 2021-10-13 2022-02-08 深圳市人民医院 Application of miR-378-5p in mesangial proliferative glomerulonephritis

Similar Documents

Publication Publication Date Title
EP2208499A1 (en) Nucleic acid capable of regulating the proliferation of cell
AU2007334502B2 (en) Compositions and methods to treat muscular & cardiovascular disorders
WO2009148137A1 (en) Nucleic acid capable of controlling degranulation of mast cell
EP2794881B1 (en) Mirna for treating head and neck cancer
EP2077326A1 (en) Novel nucleic acid
JPWO2008084319A1 (en) New nucleic acid
WO2011019074A1 (en) Nucleic acid that controls fibrosis of cells or organs
US20110118337A1 (en) Method of Using Compositions Comprising MIR-192 and/or MIR-215 for the Treatment of Cancer
EP2852669B1 (en) Therapeutic micro rna targets in chronic pulmonary diseases
EP2464730B1 (en) miRNA COMPOUNDS FOR TREATMENT OF PROSTATE CARCINOMA
WO2011111715A1 (en) Nucleic acid capable of regulating cell cycle
JPWO2010079819A1 (en) Pharmaceutical composition for the treatment of obesity or diabetes
US20100305195A1 (en) microrna mediator of cardiomyopathy and heart failure
KR101993894B1 (en) Double Stranded Oligo RNA Structure Comprising miRNA
WO2011010737A1 (en) Guide nucleic acid for cleavage of micro-rna
KR101861738B1 (en) Double Stranded Oligo RNA Structure Comprising miRNA
WO2010004816A1 (en) Inhibitor of degranulation of mast cell

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: 10808257

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10808257

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: JP