WO2010004816A1 - Inhibitor of degranulation of mast cell - Google Patents

Inhibitor of degranulation of mast cell Download PDF

Info

Publication number
WO2010004816A1
WO2010004816A1 PCT/JP2009/060220 JP2009060220W WO2010004816A1 WO 2010004816 A1 WO2010004816 A1 WO 2010004816A1 JP 2009060220 W JP2009060220 W JP 2009060220W WO 2010004816 A1 WO2010004816 A1 WO 2010004816A1
Authority
WO
WIPO (PCT)
Prior art keywords
degranulation
microrna
sirna
mast cells
substance
Prior art date
Application number
PCT/JP2009/060220
Other languages
French (fr)
Japanese (ja)
Inventor
陽史 山田
哲郎 吉田
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 WO2010004816A1 publication Critical patent/WO2010004816A1/en

Links

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
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • C12N15/1137Non-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 against enzymes
    • 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.

Definitions

  • the present invention relates to a degranulation inhibitor for mast cells, a method for inhibiting degranulation of mast cells, and a therapeutic agent for diseases caused by abnormal mast cells.
  • Non-patent Documents 1 and 2 The main ones are a wide variety of amines, arachidonic acid metabolites, proteases, cytokines, chemokines and the like.
  • mast cells are thought to play a major role in the pathogenesis of various allergic diseases, it is considered that allergic diseases can be treated by controlling the function of mast cells.
  • ⁇ 2-adrenergic receptor agonist isoproterenol has a low concentration of 10 nmol / l and suppresses the release of histamine, LT, PGD2, GM-CSF and MIP-1 ⁇ by 80% or more from cultured human mast cells.
  • Micro RNA is a small non-coding single-stranded RNA consisting of about 22 nucleotides that is not translated into protein, and is known to exist in many organisms including humans (Non-patent Documents 5 and 6). MicroRNAs are biosynthesized 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 biosynthesized from pri-miRNA.
  • Non-patent Document 7 It is known that Drosha and DGCR8 are involved in this processing. Thereafter, pre-miRNA is transported from the nucleus to the cytoplasm via Exportin-5, and then processed by Dicer and TRBP, and mature microRNA is biosynthesized from pre-miRNA (Non-patent Document 7).
  • Mature microRNAs are thought to be involved in post-transcriptional regulation of gene expression by binding complementarily to the target mRNA and suppressing mRNA translation or by degrading mRNA. It is known that microRNAs expressed in mammals including humans have an effect on mast cell degranulation (Patent Document 1). It is not known how to react like that.
  • An object of the present invention is to provide a degranulation inhibitor for mast cells, a method for inhibiting degranulation of mast cells, and a therapeutic agent for diseases caused by abnormal degranulation control of mast cells.
  • a mast cell degranulation inhibitor comprising as an active ingredient a substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor.
  • the degranulation inhibitor according to [1] wherein the substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor is a substance that suppresses the expression of Dicer1 gene.
  • the degranulation inhibitor according to [2], wherein the substance that suppresses the expression of the Dicer1 gene is a nucleic acid.
  • the degranulation inhibitor according to [3], wherein the nucleic acid is siRNA.
  • a mast cell degranulation inhibitor comprising, as an active ingredient, a vector that expresses siRNA that suppresses the expression of Dicer1 gene.
  • a method for inhibiting degranulation of mast cells comprising using a substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor.
  • the substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor is a substance that suppresses the expression of Dicer1 gene.
  • the siRNA is an siRNA having a base sequence represented by any one of SEQ ID NOs: 1 to 5 as a target sequence.
  • a therapeutic agent for a disease caused by abnormal mast cells comprising the degranulation inhibitor according to any one of [1] to [6] as an active ingredient.
  • the therapeutic agent according to [13], wherein the disease caused by abnormality of mast cells is a disease selected from the group consisting of atopic dermatitis, asthma, chronic obstructive pulmonary disease and allergic disease.
  • a method for treating a disease caused by abnormal mast cells comprising administering an effective amount of the degranulation inhibitor according to any one of [1] to [6] to a subject in need thereof.
  • a degranulation inhibitor for mast cells a method for inhibiting degranulation of mast cells, and a therapeutic drug for diseases caused by abnormal mast cells can be provided.
  • the present invention provides a mast cell degranulation inhibitor comprising as an active ingredient a substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor.
  • microRNA refers to single-stranded RNA having a length of 17 to 28 bases.
  • the surrounding genomic sequence including the sequence of the microRNA has a sequence capable of forming a hairpin structure, and the microRNA can be cut out from any one 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.
  • the microRNA of the present invention includes mature microRNA.
  • 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, and a hairpin structure is formed and microRNA is contained in one strand. Nucleic acid.
  • the microRNA precursors of the present invention include primary-microRNA (pri-miRNA) and precursor-microRNA (pre-miRNA).
  • a factor essential for biosynthesis of microRNA from the microRNA precursor As a substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor, a factor essential for biosynthesis of microRNA from the microRNA precursor (hereinafter also referred to as a factor essential for biosynthesis of microRNA). Any substance may be used as long as it inhibits the function of.
  • Factors essential for microRNA biosynthesis include, for example, Drosha and DGCR8, which are involved in the process of pre-miRNA biosynthesis from pri-miRNA, and the process of transport of pre-miRNA from the nucleus to the cytoplasm Examples include Dicer and TRBP involved in the process of biosynthesis of mature microRNA from Exportin-5 and pre-miRNA.
  • a substance that inhibits the function of a factor essential for microRNA biosynthesis for example, a substance that inhibits the expression of a gene encoding the factor, a substance that inhibits the function of the protein of the factor, a nucleic acid, an antibody, Any of low-molecular substances and the like may be used, but preferred is nucleic acid.
  • Substances that inhibit the function of factors essential for microRNA biosynthesis preferably substances that inhibit the expression of the gene encoding Dicer (hereinafter also referred to as Dicer gene), and substances that inhibit the function of Dicer protein Can be given.
  • Dicer gene examples include a Dicer1 gene, such as a human Dicer1 gene, and an ortholog that is a homologous gene of another species corresponding thereto, and preferably a human Dicer1 gene contained in the nucleotide sequence represented by SEQ ID NO: 6 or 7 can give.
  • Dicer1 is mentioned as a protein of Dicer, Preferably human Dicer1 is mention
  • substances that inhibit gene expression include substances that suppress the biosynthesis and translation of mRNA, and substances that reduce the amount of protein translated from mRNA by cleaving or decomposing mRNA.
  • nucleic acids that inhibit the function of factors essential for microRNA biosynthesis include the nucleic acids described in the following (a) to (c), and preferably the nucleic acids described in (a).
  • siRNA including a base sequence of a target nucleic acid of a gene encoding the factor short interferance RNA
  • B an antisense nucleic acid for the transcription product of the gene encoding the factor or a part thereof
  • a nucleic acid having a ribozyme activity that specifically cleaves the transcription product of the gene encoding the factor
  • the nucleic acid may be any molecule as long as it is a molecule obtained by polymerizing nucleotides or molecules having functions equivalent to those of the nucleotides.
  • nucleotides include RNAs that are polymers of ribonucleotides, DNAs that are polymers of deoxyribonucleotides, polymers in which RNA and DNA are mixed, and molecules in which molecules having functions equivalent to the nucleotides are polymerized. Examples thereof include a nucleotide polymer containing a nucleotide analog and a nucleotide polymer containing a nucleic acid derivative.
  • Nucleotide analogs include, for example, ribonucleic acid, to improve or stabilize nuclease resistance, increase affinity with complementary strand nucleic acids, increase cell permeability, or visualize, compared to RNA or DNA. Any molecule may be used as long as it is a modification of nucleotide, deoxyribonucleotide, 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), more specifically, 2′-position oxygen
  • 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 derivatives include atoms (for example, hydrogen atom, oxygen atom) or functional groups (for example, hydroxyl group, amino group) such as base portion, ribose portion, and phosphodiester bond portion of nucleic acid as other atoms (for example, hydrogen atom).
  • nucleic acid for example, lipid , Phospholipid, phenazine, folate, phenanthridine, anthraquinone, acridine, fluorescein, rhodamine, coumarin, and a molecule to which another chemical substance is added.
  • molecules obtained by adding another chemical substance to nucleic acid include 5′-polyamine addition derivatives, cholesterol addition derivatives, steroid addition derivatives, bile acid addition derivatives, vitamin addition derivatives, Cy5 addition derivatives, Cy3 addition derivatives, 6 -FAM addition derivatives, biotin addition derivatives, and the like are also included.
  • the siRNA containing the base sequence of the target nucleic acid of the gene encoding the factor essential for microRNA biosynthesis is a short double-stranded RNA containing the base sequence of a certain target nucleic acid, which is caused by RNA interference (RNAi). Any one can be used as long as it can suppress the expression of the target nucleic acid.
  • RNAi RNA interference
  • the number of bases constituting one strand of siRNA is preferably 17 to 30 bases, more preferably 18 to 25 bases, and still more preferably 19 to 23 bases.
  • siRNA may be composed only of RNA, but as long as the expression of the target nucleic acid can be suppressed, DNA, RNA and DNA mixed polymers, nucleotide polymers including nucleotide analogs, nucleotide polymers including nucleic acid derivatives Any nucleic acid may be used, such as a substituted one.
  • the siRNA may also contain mismatches, bumps, loops, or wobble base pairs as necessary in the double-stranded molecule in order to regulate the ability to mediate inhibition of target gene expression.
  • Such mismatches, bumps, loops or wobble base pairs are preferably placed in the terminal or internal region of the siRNA so that the siRNA does not significantly impair the ability to mediate inhibition of target gene expression.
  • the double-stranded RNA used in the present invention may have an overhang of several bases at the end.
  • the length and sequence of the base that forms this overhang are not particularly limited.
  • the overhang may be either DNA or RNA.
  • two base overhangs such as TT (two thymines), UU (two uracils), and other base overhangs.
  • TT two thymines
  • UU two uracils
  • a molecule having a 19-base double-stranded RNA and a 2-base (TT) overhang can be preferably used.
  • Double-stranded RNA includes molecules in which the base that forms the overhang is DNA.
  • the siRNA that suppresses the expression of the target nucleic acid is, for example, an algorithm for designing an siRNA for the gene based on the sequence [Nucleic Acids Res., 32, 936 (2004); Nature Biotechnology, 22, 326 (2004); Nature Biotechnology , 23, 995 (2005)].
  • SiRNAs designed using the above algorithm are commercially available from Qiagen, Applied Biosystems, Invitrogen, etc., and these may be used.
  • the siRNA preferably includes siRNA containing the base sequence of the target nucleic acid of human Dicer1 gene.
  • the siRNA may be any siRNA that suppresses the expression of the endogenous human Dicer1 gene by introducing the siRNA into the cell.
  • siRNA against human Dicer1 gene and the like can also be designed using the above algorithm.
  • the siRNA in which the expression level of the human Dicer1 gene is reduced or suppressed is preferably SEQ ID NO: 1, 2, 3 [Molecular Cell 24, 157 (2006)], 4 [Nature, 436, 740 (2005)] or 5 Examples include siRNAs having a base sequence represented by [Nature, 436, 740 (2005)] as a target sequence.
  • examples of the siRNA having the base sequence represented by SEQ ID NO: 1 as a target sequence include the base sequences shown below.
  • Antisense strand 5'-CUAGGAUCCAGAUAGCACAdTdT-3 '(SEQ ID NO: 8)
  • Sense strand 5'-UGUGCUAUCUGGAUCCUAGdTdT-3 '(SEQ ID NO: 9)
  • siRNA which makes the base sequence represented by the said sequence number 2 a target sequence
  • the base sequence shown below can be mention
  • Antisense strand 5'-UCCAGAGCUGCUUCAAGCAdTdT-3 '(SEQ ID NO: 10)
  • Sense strand 5'-UGCUUGAAGCAGCUCUGGAdTdT-3 '(SEQ ID NO: 11)
  • dT is T (overhang) of DNA, and the other is RNA.
  • the present invention provides a degranulation inhibitor for mast cells, containing as an active ingredient a vector that expresses siRNA that suppresses the expression of Dicer1 gene.
  • a vector that expresses siRNA that suppresses the expression of Dicer1 gene can easily prepare a vector capable of expressing the siRNA by a general genetic engineering technique.
  • the expression vector described in 3 below can be used.
  • the antisense nucleic acid for the transcription product of a gene encoding a factor essential for microRNA biosynthesis or a part thereof preferably has a sequence complementary to the gene encoding the factor or a part thereof.
  • the antisense nucleic acid may not be completely complementary as long as it can effectively suppress the expression of the gene encoding the factor, but is preferably 90% or more with respect to the transcription product of the gene encoding the factor. Preferably it has a complementarity of 95% or more.
  • the length of the antisense nucleic acid is at least 15 bases, preferably 100 bases or more, more preferably 500 bases or more.
  • an antisense nucleic acid if an antisense sequence complementary to the untranslated region near the 5 ′ end of the mRNA of the gene encoding the factor is designed, it is considered effective for inhibiting the translation of the gene.
  • a sequence complementary to the coding region or the 3 ′ untranslated region can also be used.
  • the antisense nucleic acid can be transformed into a desired animal using a known method after ligating, for example, downstream of an appropriate promoter, and preferably ligating a sequence containing a transcription termination signal on the 3 ′ side.
  • any RNA molecule having catalytic activity may be used, such as group I intron type or RNase P.
  • group I intron type or RNase P such as those having a size of 400 bases or more, such as the M1 RNA contained in, and those having an active domain of about 40 bases called hammerhead type or hairpin type.
  • the self-cleaving domain of the hammerhead ribozyme cleaves 3 'of C15 in the sequence G13U14C15, but base pairing between U14 and A9 is important for its activity. Instead of C15, A15 or U15 Can be cut [FEBS Lett., 228, 228 (1988)].
  • a ribozyme whose substrate binding site is complementary to the RNA sequence in the vicinity of the target site, it is possible to create a restriction enzyme-like RNA-cleaving ribozyme that recognizes the sequence UC, UU or UA in the target RNA [FEBSettLett , 239, 285 (1988); Protein Nucleic Acid Enzyme, 35, 2191 (1990); Nucl. Acids, Res 17, 7059 (1989)].
  • Hairpin ribozymes are found, for example, in the minus strand of satellite RNA of tobacco ring spot virus [Nature, 323, 349 (1986)].
  • an RNA-cleaving ribozyme specific for the base sequence of the target nucleic acid May be produced.
  • the substance that inhibits the function of the protein of the factor essential for microRNA biosynthesis include the antibody described in (a) below or the low molecular compound described in (b).
  • Antibodies that bind to factors essential for biosynthesis of microRNAs are prepared by methods known to those skilled in the art using recombinant proteins prepared using genetic recombination techniques in addition to the natural proteins of the factors. be able to.
  • a polyclonal antibody can be prepared as follows.
  • a small protein such as a rabbit is immunized with a recombinant protein expressed in a microorganism such as Escherichia coli or a partial peptide thereof as a protein of the factor or a fusion protein with GST to obtain serum.
  • This can be prepared, for example, by purification using ammonium sulfate precipitation, protein A, protein G column, DEAE ion exchange chromatography, an affinity column coupled with the protein of the factor or a synthetic peptide, or the like.
  • the protein of the factor or a partial peptide thereof is immunized to a small animal such as a mouse, the spleen is removed from the mouse, and this is ground to separate the cells, and the cells and mouse myeloma cells Are fused using a reagent such as polyethylene glycol, and a clone that produces an antibody that binds to the factor is selected from the fused cells (hybridoma).
  • a reagent such as polyethylene glycol
  • the obtained hybridoma was transplanted into the abdominal cavity of the mouse, and ascites was collected from the mouse, and the obtained monoclonal antibody was obtained by, for example, ammonium sulfate precipitation, protein A, protein G column, DEAE ion exchange chromatography, It can be prepared by purification using an affinity column coupled with protein or synthetic peptide.
  • the form of the antibody is not particularly limited, and as long as it binds to the factor, in addition to the polyclonal antibody and the monoclonal antibody, a human antibody, a humanized antibody by genetic recombination, an antibody fragment thereof, and an antibody modification product are also included. It is.
  • the protein of the factor used as a sensitizing antigen for antibody acquisition is not limited to the animal species from which it is derived, but is preferably a protein derived from a mammal such as a mouse or a human, and particularly preferably a protein derived from a human.
  • Human-derived proteins can be obtained using the gene sequences disclosed in the present specification.
  • the protein used as the sensitizing antigen may be a complete protein or a partial peptide of the protein.
  • the partial peptide of the protein include an amino group (N) terminal fragment and a carboxy (C) terminal fragment of the protein.
  • human lymphocytes such as human lymphocytes infected with EB virus are sensitized with proteins, protein-expressing cells or lysates thereof in vitro. Lymphocytes can be fused with human-derived myeloma cells having permanent mitotic activity, such as U266, to obtain hybridomas that produce desired human antibodies having protein-binding activity.
  • an antibody that binds to the protein of the factor is used for the purpose of administering it to the human body (antibody treatment)
  • a human antibody or a human type antibody is preferable in order to reduce immunogenicity.
  • Examples of the substance that can inhibit the function of the protein of the factor include low molecular weight substances that bind to the protein of the factor.
  • the low molecular weight substance that binds to the protein of the factor may be a natural or artificial compound. Usually, it is a compound that can be produced or obtained by using methods known to those skilled in the art.
  • Dicer is preferable, Dicer1 is more preferable, and human Dicer1 is more preferable.
  • the present invention also provides a method for suppressing degranulation of mast cells, characterized by using a substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor.
  • the substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor inhibits the process of biosynthesis of microRNA from the microRNA precursor in the degranulation inhibitor described above. It is the same as the substance to do.
  • the present invention also provides a method for inhibiting degranulation of mast cells, characterized by using a vector that expresses siRNA that suppresses the expression of Dicer1 gene.
  • the siRNA used in the degranulation suppressing method of the present invention is preferably an siRNA having a base sequence represented by any one of SEQ ID NOs: 1 to 5 as a target sequence.
  • the mast cell degranulation inhibitor provided by the present invention suppresses mast cell degranulation, and is therefore expected to have a therapeutic effect on diseases caused by mast cell abnormalities. Therefore, this invention relates to the therapeutic agent (pharmaceutical composition) of the disease resulting from the abnormality of a mast cell which contains the degranulation inhibitor of the mast cell of this invention as an active ingredient.
  • the degranulation inhibitor of the present invention can be used not only for treatment of diseases caused by abnormalities of mast cells but also for diagnosis.
  • the degranulation inhibitor provided by the present invention is expected to have a therapeutic effect on diseases caused by abnormalities of mast cells as described above, an effective amount of the degranulation inhibitor is required.
  • a disease caused by abnormal mast cells can be treated. That is, the present invention relates to a method for treating a disease caused by abnormal mast cells, comprising administering an effective amount of the degranulation inhibitor of the present invention to a subject in need.
  • the present invention also relates to the use of the degranulation inhibitor of the present invention for producing a therapeutic agent for diseases caused by abnormalities of mast cells.
  • the present invention also relates to the use of the degranulation inhibitor of the present invention in the manufacture of a therapeutic agent for diseases caused by abnormal mast cells.
  • abnormalities in mast cells include abnormalities such as mast cell differentiation and degranulation, inflammatory mediator production, cytokine production, and chemokine production.
  • diseases caused by abnormal mast cells include atopic dermatitis, asthma, chronic obstructive pulmonary disease, and allergic diseases.
  • the mast cells used in the present invention are preferably human mast cells.
  • Human mast cells are not particularly limited as long as they are safe and efficient, and for example, known methods from human lung, skin, fetal liver and the like [J. Immunol. Methods, 169, 153 (1994) J. Immunol., 138, 861 (1987); J. Allergy Clin. Immunol., 107, 322 (2001); J. Immunol. Methods., 240, 101 (2000)]. Further, known methods [J. Immunol., 157, 343, (1996); Blood, 91, 187 (1998); J. Allergy Clin.
  • a cell line established from human mast cells can also be used.
  • human mast cell lines LAD2 [Leuk. Res., 27, 671, which is known to retain the properties of human mast cells, is known. (2003); Leuk. Res., 27, 677 (2003)].
  • a substance has at least one of the following actions on mast cells: inhibition of activation, inhibition of degranulation, production of inflammatory mediators, inhibition of cytokine production and inhibition of chemokine production.
  • the mast cells were stimulated and released (i) Histamine and ⁇ -hexosaminidase as indicators of degranulation, (ii) Inflammatory mediators such as LTC4, LTD4, LTE4, PGD2, (iii) Cytokines such as TNF- ⁇ and GM-CSF, (iv) IL- 8, It can be confirmed by measuring chemokines such as I-309, MIP-1 ⁇ , etc., and comparing with a case where a substance that inhibits the process of biosynthesis of microRNA from the microRNA precursor of the present invention was not introduced. .
  • Methods for stimulating mast cells include, for example, a method of adding anti-IgE antibody after incubation with IgE, a method of adding compound 48/80, a method of adding polymyxin B, a method of adding dextran, calcium Method of adding ionophore, method of adding acetylcholine, method of adding carbachol, method of adding thrombin, method of adding concanavalin A, method of adding calcium ionophore, method of adding ATP, method of adding doxorubicin, Etc.
  • mast cells instead of degranulation, the activation of mast cells involves cytokine production such as TNF- ⁇ and GM-CSF, chemokine production such as IL-8, I-309, MIP-1 ⁇ , LTC4, LTD4, LTE4, PGD2, etc. It can also be examined by measuring inflammatory mediator production etc. [Blood, 100, 3861 (2002)].
  • cytokine production such as TNF- ⁇ and GM-CSF
  • chemokine production such as IL-8, I-309, MIP-1 ⁇ , LTC4, LTD4, LTE4, PGD2, etc. It can also be examined by measuring inflammatory mediator production etc. [Blood, 100, 3861 (2002)].
  • RNAs such as microRNAs
  • RNAs such as microRNAs
  • Trizol Invitrogen
  • ISOGEN Nippon Gene
  • mirVana TM miRNA Isolation Kit Ambion
  • miRNeasy Mini Kit Qiagen
  • low molecular weight RNA can be cloned from total RNA including low molecular weight RNA.
  • a method for cloning small RNA specifically, according to the method described in Genes & Development, 15, 188-200 (2000), separation and excision of low molecular RNA by 15% polyacrylamide gel electrophoresis, 5'-terminal dephosphorylation, 3'-adapter ligation, phosphorylation, 5'-adapter ligation, reverse transcription, PCR amplification, concatamerization, ligation to vector, and then cloning of small RNA, the base of the clone Examples include a method for determining the sequence.
  • RNA can also be obtained.
  • small RNAs can be cloned using small RNA Cloning Kit (Takara Bio Inc.).
  • RNA that is predicted to be transcribed from the genomic sequence Predict secondary structure.
  • Genome sequences are publicly available and can be obtained from, for example, UCSC Genome Bioinformatics (http://genome.ucsc.edu/).
  • RNAfold Nucleic Acids Research, 31, 3429-3431 (2003)
  • Mfold Nucleic Acids Research, 31, 3406-3415 (2003)]
  • miRBase http://microrna.sanger.ac.uk/
  • Methods for detecting the expression level of microRNA or its precursor include, for example, (1) Northern hybridization, (2) Dot blot high Hybridization, (3) in situ hybridization, (4) quantitative PCR, (5) differential hybridization, (6) microarray, (7) ribonuclease protection assay and the like.
  • Northern hybridization involves separating the sample-derived RNA by gel electrophoresis and then transferring it to a support such as a nylon filter to prepare a probe appropriately labeled based on the base sequence of the microRNA or its precursor. It is a method for detecting a band specifically bound to the microRNA or its precursor by performing hybridization and washing, and specifically, for example, the method described in Science, 294, 853-858 (2001) Etc. can be performed according to the above.
  • the labeled probe can be prepared by, for example, radioisotope, biotin, digoxigenin, fluorescent group, chemiluminescent group, etc. by the method such as nick translation, random priming, or phosphorylation at the 5 ′ end, and the micro or its RNA precursor. It can be prepared by incorporating it into DNA, RNA or LNA having a sequence complementary to the base sequence. Since the binding amount of the labeled probe reflects the expression level of the microRNA or its precursor, the expression level of the microRNA or its precursor can be determined by quantifying the amount of the labeled probe bound. . Electrophoresis, membrane transfer, probe preparation, hybridization, and nucleic acid detection can be performed by the methods described in Molecular Cloning 3rd edition.
  • RNA extracted from tissues or 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 paraffin or cryostat sections of tissue obtained from a living body, or immobilized cells as specimens, performs hybridization and washing steps with a labeled probe, and performs microscopic observation. This is a method for investigating the distribution and localization of RNA or its precursor in tissues and 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 also referred to as a sample-derived cDNA) is used for measurement.
  • a random primer or a specific RT primer can be used as a reverse transcription primer for cDNA synthesis.
  • the specific RT primer refers to a primer having a sequence complementary to a base sequence corresponding to the microRNA or a precursor thereof and a surrounding genomic sequence.
  • a template-specific primer designed from the base sequence corresponding to the microRNA or its precursor and its surrounding genomic sequence, or the base sequence corresponding to the reverse transcription primer PCR is used to amplify the cDNA fragment containing the micro or its RNA precursor, and the amount of the micro RNA or its precursor contained in the sample-derived RNA is detected from the number of cycles to reach a certain amount To do.
  • DNA or LNA comprising a sequence of 20 to 40 bases at the 5 ′ end of the base sequence of the region And a DNA or LNA pair consisting of a sequence complementary to 20 to 40 bases at the 3 ′ end can be used. Specifically, it can be carried out 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, it can be carried out using the method described in Nucleic® Acid Research, 33, 179 (2005) or TaqMan® MicroRNA Assays (Applied Biosystems).
  • sample-derived cDNA is hybridized to a filter or slide glass, silicon, or other substrate on which DNA corresponding to a base sequence containing at least one of the microRNAs or a precursor thereof, or LNA is immobilized, and washed.
  • a filter or slide glass, silicon, or other substrate on which DNA corresponding to a base sequence containing at least one of the microRNAs or a precursor thereof, or LNA is immobilized, and washed.
  • methods based on such hybridization include methods using differential hybridization [Trends Genet., 7, 314 (1991)] and microarrays [Genome Res., 6, 639 (1996)].
  • Each method immobilizes an internal control such as a nucleotide sequence corresponding to U6RNA on a filter or substrate to accurately detect the difference in the amount of the microRNA or its precursor between the control sample and the target sample. can do.
  • microRNA or a precursor thereof can be quantified.
  • microRNAs can be detected using a microarray described in Proc. Natl. Acad. Sci. USA, 101, 9740-9744 (2004), Nucleic Acid Research, 32, e188 (2004). Specifically, it can be detected or quantified in the same manner as mirVana miRNA Bioarray (Ambion).
  • a promoter sequence such as T7 promoter or SP6 promoter is bound to the 3 ′ end of the base sequence corresponding to the microRNA or a precursor thereof or the surrounding genomic sequence, and labeled NTP (ATP, GTP, CTP). , UTP mixture) and an in vitro transcription system using RNA polymerase to synthesize labeled antisense RNA.
  • 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 digested product is subjected to gel electrophoresis, and an RNA fragment protected from digestion by forming an RNA-RNA hybrid is detected or quantified as the nucleic acid or microRNA precursor of the present invention. Specifically, it can be detected or quantified using mirVana miRNA Detection Kit (Ambion).
  • MicroRNAs are transcribed from a gene into a primary transcript, primary-microRNA (pri-miRNA), followed by step-wise processing from pri-miRNA to mature microRNA.
  • primary-microRNA primary-microRNA
  • pre-miRNA precursor-microRNA
  • Mature microRNA is biosynthesized from pre-miRNA.
  • any method for inhibiting the biosynthesis of microRNA any method may be used as long as one of the above processes is inhibited and the amount of mature microRNA decreases.
  • a substance that inhibits the function of the factor essential for the above-described microRNA biosynthesis can be used in the method.
  • a vector that expresses siRNA can be prepared by inserting a DNA corresponding to a base sequence selected from a target base sequence into an expression vector.
  • an expression vector a vector that can replicate autonomously in a host cell, can be integrated into a chromosome, and contains a promoter at a position where a genomic gene containing a nucleic acid base sequence can be transcribed is used.
  • the promoter any promoter can be used so long as it can be expressed in the host cell.
  • RNA polymerase II (pol II) type promoter or U RNA or H1RNA transcription system such as RNA polymerase III (pol III) type promoter. Can give.
  • Examples of the pol II promoter include cytomegalovirus (human CMV) IE (immediate early) gene promoter, SV40 early promoter, 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 promoters. Examples of expression vectors using them include pSINsi-hH1 DNA (Takara Bio), pSINsi-hU6 DNA (Takara Bio), pENTR / U6 (Invitrogen) and the like.
  • a DNA corresponding to the selected base sequence is inserted downstream of the promoter in the viral vector to construct a recombinant viral vector, and the vector is introduced into a packaging cell to produce a recombinant virus.
  • the siRNA 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 viral packaging, such as human kidney.
  • HEK293 cells derived from mouse, mouse fibroblast NIH3T3, and the like can be used. Proteins supplemented by packaging cells include mouse retrovirus-derived gag, pol, env, etc.
  • HIV virus-derived gag, pol, env, vpr, vpu for lentiviral vectors.
  • Vif, tat, rev, nef, etc., adenovirus vectors, E1A, E1B, etc. Can be used.
  • Whether or not the biosynthesis of microRNA is actually inhibited can be confirmed by measuring the expression level for any microRNA according to the method described in 2 above.
  • the amount of mRNA and protein of factors essential for biosynthesis of microRNA can be measured by a real-time RT-PCR method or western blotting method, respectively, and it can be confirmed whether or not these amounts have decreased.
  • Mast cell degranulation inhibitor using a substance that inhibits microRNA biosynthesis The substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor according to the present invention is used as a mast cell degranulation inhibitor. Can be used. As described in 3 above, the substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor may be any substance as long as it inhibits the function of factors essential for biosynthesis. However, for example, siRNA for human Dicer1 gene can be mentioned.
  • mast cells which is a substance that inhibits the process of biosynthesis of microRNA from the microRNA precursor of the present invention
  • the mast cells After introducing into the mast cells a substance that inhibits the process of biosynthesis, the mast cells are stimulated and released (i) histamine and ⁇ -hexosaminidase, which are indicators of degranulation, (ii) LTC4, Measure inflammatory mediators such as LTD4, LTE4, PGD2, etc., (iii) cytokines such as TNF- ⁇ and GM-CSF, (iv) chemokines such as IL-8, I-309, MIP-1 ⁇ , etc. This can be confirmed by comparing with a case where a substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor was not introduced.
  • the formulation form and administration method of the mast cell degranulation inhibitor of the present invention are the same as those of the therapeutic agent containing a substance that inhibits biosynthesis of microRNA, which will be described later in 5.
  • Therapeutic drugs containing substances that inhibit the biosynthesis of microRNA Substances that inhibit the process of biosynthesis of microRNAs from microRNA precursors can suppress mast cell degranulation, thereby preventing abnormalities in mast cells. It can be used as a therapeutic agent for diseases caused by it. Abnormalities in mast cells include abnormalities such as mast cell differentiation and degranulation, inflammatory mediator production, cytokine production, chemokine production, etc. Diseases resulting from them include atopic dermatitis, asthma, chronic obstructive pulmonary disease And allergic diseases.
  • a therapeutic agent containing as an active ingredient a substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor can be administered alone, but usually one or more pharmacologically acceptable It is desirable to administer it as a pharmaceutical preparation prepared by any method well known in the pharmaceutical arts.
  • oral administration or parenteral administration such as buccal, respiratory tract, rectal, subcutaneous, intramuscular and intravenous is desirable. Can be given intravenously.
  • Administration forms include sprays, capsules, tablets, granules, syrups, emulsions, suppositories, injections, ointments, tapes and the like.
  • Preparations suitable 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.
  • Preparations suitable for parenteral administration include injections, suppositories, sprays and the like.
  • 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 cocoa 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.
  • preparations such as aerosols and dry powders are 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, body weight, etc., but is usually 10 ⁇ g / kg to 20 mg / kg per day for an adult.
  • the therapeutic agent containing the degranulation inhibitor of the present invention as an active ingredient such as a substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor, is used as a nucleic acid-expressing vector and nucleic acid therapeutic agent. It can also be produced by blending with a base [Nature Genet., 8, (42 (1994)].
  • the base used in the therapeutic agent of the present invention may be any base as long as it is usually used in injections, salt water such as distilled water, sodium chloride or a mixture of sodium chloride and an inorganic salt, mannitol, Examples thereof include a solution of lactose, dextran, glucose and the like, 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.
  • salt water such as distilled water, sodium chloride or a mixture of sodium chloride and an inorganic salt
  • mannitol examples thereof include a solution of lactose, dextran, glucose and the like, 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 of the present invention 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 vector that expresses the nucleic acid can be the recombinant virus vector prepared in 3 above. More specifically, a retrovirus vector, a lentivirus vector, etc. can be mentioned.
  • a viral vector can be prepared by preparing a complex by combining the above nucleic acid with a polylysine-conjugated antibody specific for an adenovirus hexon protein and binding the resulting complex to an adenovirus vector.
  • the virus vector stably reaches the target cell, is taken up into the cell by endosomes, is degraded in the cell, and the nucleic acid can be efficiently expressed.
  • the nucleic acid can also be transferred by non-viral nucleic acid transfer method.
  • non-viral nucleic acid transfer method For example, calcium phosphate coprecipitation method [Virology, 52, 456-467 (1973); Science, 209, 1414-1422 (1980)], microinjection method [Proc. Natl. Acad. Sci. USA, 77, 5399-5403-5 ( 1980); Proc. Natl. Acad. Sci. USA, 77, 7380-7384 (1980); Cell, 27, 223-231 (1981); Nature, 294, 92-94 (1981)], liposome-mediated membrane Fusion-Interventional Transfer [Proc. Natl. Acad. Sci. USA, 84, 13 7413-7417 (1987); Biochemistry, 28, 9508-9514 (1989); J. Biol.
  • Membrane fusion-mediated transfer via liposomes allows the nucleic acid to 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 the lesion.
  • Receptor-mediated DNA transfer can be performed, for example, by binding DNA (typically 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.
  • SiRNA that inhibits the process of biosynthesis of microRNA As a substance that inhibits the process of biosynthesis of microRNA, we focused on siRNA of human Dicer1 gene, which is one of the essential factors for biosynthesis of microRNA, and examined whether this actually reduces Dicer1 gene expression. .
  • siRNA for human Dicer1 gene is lipofection method, specifically, Lipofectamine2000 (Invitrogen) is used to achieve a final concentration of 20 nM.
  • Lipofectamine2000 Invitrogen
  • siRNA sequence for human Dicer1 gene Dicer-siRNA-1 having SEQ ID NO: 1 as a target sequence and Dicer-siRNA-2 having SEQ ID NO: 2 as a target sequence were purchased from Qiagen. Lipofection followed the method described in the instructions attached to the product. Moreover, the cell which carried out lipofection to the HeLa cell, without adding siRNA was set as control.
  • the cDNA synthesized above was used as a template for the PCR reaction, and Dicer1-specific PCR amplification was performed by SYBR-Green® PCR using ABI7900HT® Fast (Applied Biosystems) to quantify the amount of mRNA.
  • the amount of mRNA in the sample was evaluated as a relative ratio when the amount of Dicer1 mRNA was 1 in the control cells.
  • Dicer1 mRNA in Dicer-siRNA-1-introduced cells was 0.38
  • the relative expression level of Dicer1 in Dicer-siRNA-2-introduced cells was 0.24
  • expression of Dicer1 gene by siRNA introduction into Dicer1 It was confirmed that the amount was reduced.
  • LAD2 is a recently established human mast cell line that is well known to retain the properties of human mast cells [Leuk. Res., 27, 671 (2003); Leuk. Res., 27, 677 (2003)].
  • LAD2 was obtained from National Institute of Allergy and Infectious Diseases, National Institutes of Health (Bethesda, MD 20892-1881, USA), and cultured in Stem Pro-34 medium [manufactured by Invitrogen] containing 100 ng / mL SCF.
  • LAD2 is seeded in a 6-well plate at 5x10 5 per well, and siRNA (Dicer-siRNA-1) for human Dicer1 gene is lipofection method, specifically using Gene Silencer (Genlantis) The final concentration was 25 nM.
  • siRNA sequence for human Dicer1 gene Dicer-siRNA-1 (manufactured by Qiagen) having SEQ ID NO: 1 as a target sequence was used. Lipofection followed the method described in the instructions attached to the product.
  • the ⁇ -hexosaminidase activity was determined by using 4 mmol / L p-nitrophenyl N-acetyl- ⁇ -glucosaminide (Sigma) dissolved in 40 mmol / L citrate buffer (pH 4.5) in 50 ⁇ L of the collected supernatant. ), Incubate at 37 ° C for 1 hour, and then measure the absorbance at 405 nm of the sample to which 100 ⁇ L of 0.2 mol / L glycine (pH 10.7) has been added using a plate reader 1420 ARVOsx (Perkin Elmer). It was evaluated by.
  • the total ⁇ -hexosaminidase activity in LAD2 was measured by adding Triton X-100 having a final concentration of 1% instead of the rabbit anti-human IgE antibody and conducting the same experiment.
  • the percentage of degranulation was calculated as the percentage of ⁇ -hexosaminidase activity in the supernatant with respect to the total ⁇ -hexosaminidase activity, and the percentage of degranulation in the negative control (Gene Silencer only) for each.
  • the relative degranulation activity was calculated when 1.0 was 1.0.
  • human myeloma IgE (manufactured by Cosmo Bio) was added to a final concentration of 1.0 ⁇ g / mL and cultured overnight in an incubator at 37 ° C. with 5% CO 2 concentration. The following day, degranulation was induced by adding rabbit anti-human IgE antibody, and the degranulation rate was measured. For each, the degranulation relative activity when the degranulation rate in the negative control group (Gene Silencer only) was 1.0 Was calculated.
  • Table 1 shows the results of degranulation relative activity on the next day (that is, after 3 days and 8 days) after 2 days and 7 days after the introduction of the siRNA, respectively.
  • siRNA (SEQ ID NO: 1) as a target sequence (Dicer-siRNA-1)
  • siRNA (SEQ ID NO: 2) as a target sequence (Dicer-siRNA-2)
  • SEQ ID NO: 2 siRNA as a target sequence
  • LAD2 is seeded in a 6-well plate at 5x10 5 per well, and siRNA is introduced by lipofection method, specifically Gene Silencer (Genlantis) to a final concentration of 30 nM. did. Lipofection followed the method described in the instructions attached to the product.
  • the supernatant was collected by centrifugation, and the degree of degranulation was measured by measuring ⁇ -hexosaminidase activity in the supernatant.
  • the ⁇ -hexosaminidase activity was determined by using 4 mmol / L p-nitrophenyl N-acetyl- ⁇ -glucosaminide (Sigma) dissolved in 40 mmol / L citrate buffer (pH 4.5) in 50 ⁇ L of the collected supernatant.
  • the percentage of degranulation was calculated as the percentage of ⁇ -hexosaminidase activity in the supernatant with respect to the total ⁇ -hexosaminidase activity, and the percentage of degranulation in the negative control (Gene Silencer only) for each.
  • the relative degranulation activity was calculated when 1.0 was 1.0.
  • Table 2 shows the results of degranulation relative activity in which each siRNA was introduced.
  • LAD2 introduced with siRNA against human Dicer1 gene was stimulated with other than anti-IgE antibody, and the influence on degranulation was examined.
  • LAD2 is seeded in a 6-well plate at 5x10 5 per well, and siRNA (Dicer-siRNA-1) for human Dicer1 gene is lipofection method, specifically using Gene Silencer (Genlantis) The final concentration was 30 nM.
  • siRNA sequence for human Dicer1 gene Dicer-siRNA-1 (manufactured by Qiagen) having SEQ ID NO: 1 as a target sequence was used. Lipofection followed the method described in the instructions attached to the product.
  • the medium is removed by centrifugation, washed with Tyrode buffer, 1.5 mL of Tyrode buffer is added, the cells are suspended, and 1 is added to a 96-well plate. Dispense 100 ⁇ L per well.
  • compound 48/80 manufactured by Sigma-Aldrich was added to a final concentration of 0.5 ⁇ g / mL, and incubated for 20 minutes in an incubator at 37 ° C. with 5% CO 2 concentration to induce degranulation. The supernatant was collected by centrifugation, and the degree of degranulation was measured by measuring ⁇ -hexosaminidase activity in the supernatant.
  • the ⁇ -hexosaminidase activity was determined by using 4 mmol / L p-nitrophenyl N-acetyl- ⁇ -glucosaminide (Sigma) dissolved in 40 mmol / L citrate buffer (pH 4.5) in 50 ⁇ L of the collected supernatant. ), Incubate at 37 ° C for 1 hour, and then measure the absorbance at 405 nm of the sample to which 100 ⁇ L of 0.2 mol / L glycine (pH 10.7) has been added using a plate reader 1420 ARVOsx (Perkin Elmer). It was evaluated by.
  • the total ⁇ -hexosaminidase activity in LAD2 was measured by adding Triton X-100 having a final concentration of 1% instead of Compound 48/80 and conducting the same experiment.
  • the percentage of degranulation was calculated as the percentage of ⁇ -hexosaminidase activity in the supernatant with respect to the total ⁇ -hexosaminidase activity, and the percentage of degranulation in the negative control (Gene Silencer only) for each.
  • the relative degranulation activity was calculated when 1.0 was 1.0.
  • degranulation was measured by inducing degranulation by adding compound 48/80, and the degranulation ratio in the negative control group (Gene-Silencer only) was 1.0 for each. The relative degranulation activity was calculated.
  • Table 3 shows the results of degranulation relative activity 3 days and 7 days after the introduction of the siRNA, respectively.

Abstract

Disclosed are: an inhibitor of the degranulation of a mast cell, which comprises, as an active ingredient, a substance capable of inhibiting the process of biosynthesis of microRNA from a microRNA precursor, such as a substance capable of inhibiting the expression of Dicer1 gene; a method for inhibiting the degranulation of a mast cell; and a therapeutic agent for a disease induced by the abnormality in a mast cell.

Description

肥満細胞の脱顆粒抑制剤Mast cell degranulation inhibitor
 本発明は肥満細胞の脱顆粒抑制剤、肥満細胞の脱顆粒抑制方法、および肥満細胞の異常に起因する疾患の治療薬に関する。 The present invention relates to a degranulation inhibitor for mast cells, a method for inhibiting degranulation of mast cells, and a therapeutic agent for diseases caused by abnormal mast cells.
 肥満細胞は各種刺激により活性化され、脱顆粒を起こし、数多くの炎症性メディエーターを遊離、産生することが知られている(非特許文献1および2)。主なものはアミン類、アラキドン酸代謝産物、プロテアーゼ、サイトカイン、ケモカインなど多種多様である。例えば、肥満細胞に抗原が認識されると、脱顆粒によりヒスタミンやトリプターゼが速やかに放出されると共に、プロスタグランジンD2(PGD2)、ロイコトリエン(LT)、血小板活性化因子(PAF)などのケミカルメディエーター、およびマクロファージ炎症性蛋白質(MIP)-1α等の各種のケモカインや顆粒球マクロファージコロニー刺激因子(GM-CSF)等の各種サイトカインが新たに合成され、放出されることが知られている。また、これまで好酸球が作ると考えられていた細胞障害性蛋白質である主要塩基性蛋白質(major basic protein)に関しても、ヒトの場合は肥満細胞が多量につくることが最近明らかになっている(非特許文献3)。 It is known that mast cells are activated by various stimuli, cause degranulation, and release and produce many inflammatory mediators (Non-patent Documents 1 and 2). The main ones are a wide variety of amines, arachidonic acid metabolites, proteases, cytokines, chemokines and the like. For example, when an antigen is recognized by mast cells, histamine and tryptase are rapidly released by degranulation, and chemical mediators such as prostaglandin D2 (PGD2), leukotriene (LT), and platelet activating factor (PAF) It is known that various chemokines such as macrophage inflammatory protein (MIP) -1α and various cytokines such as granulocyte macrophage colony stimulating factor (GM-CSF) are newly synthesized and released. In addition, it has recently been clarified that the major basic protein (major basic protein), which is a cytotoxic protein previously thought to be produced by eosinophils, is produced in large amounts by mast cells in humans. (Non-Patent Document 3).
 このように、肥満細胞は種々のアレルギー疾患の病態形成において主要な役割を演じていると考えられることから、肥満細胞の機能を制御することにより、アレルギー疾患の治療が可能であると考えられる。 Thus, since mast cells are thought to play a major role in the pathogenesis of various allergic diseases, it is considered that allergic diseases can be treated by controlling the function of mast cells.
 肥満細胞で発現する遺伝子の情報は、種々のアレルギー疾患の治療薬のターゲットとなり得る点で重要であり、例えば、肥満細胞に発現しているGPCRに作用する薬剤が、ヒト培養肥満細胞からの炎症性メディエーターの産生を顕著に抑制することが知られている。具体的には、β2アドレナリン受容体刺激薬イソプロテレノールは10 nmol/lの低濃度で、ヒト培養肥満細胞からのヒスタミン、LT、PGD2、GM-CSFおよびMIP-1αの遊離を80%以上抑制する(非特許文献4)。 Information on genes expressed in mast cells is important in that they can be targets for therapeutic agents for various allergic diseases. For example, drugs that act on GPCRs expressed in mast cells are affected by inflammation from cultured human mast cells. It is known to significantly suppress the production of sex mediators. Specifically, the β2-adrenergic receptor agonist isoproterenol has a low concentration of 10 nmol / l and suppresses the release of histamine, LT, PGD2, GM-CSF and MIP-1α by 80% or more from cultured human mast cells. (Non-Patent Document 4).
 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5、6)。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合成される。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から生合成される。このプロセシングにはDroshaとDGCR8が関与することが知られている。その後、pre-miRNAは核内からExportin-5を介して細胞質に輸送された後、DicerとTRBPによってプロセシングされ、pre-miRNAから成熟型マイクロRNAが生合成される(非特許文献7)。 Micro RNA (miRNA) is a small non-coding single-stranded RNA consisting of about 22 nucleotides that is not translated into protein, and is known to exist in many organisms including humans (Non-patent Documents 5 and 6). MicroRNAs are biosynthesized 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 biosynthesized from pri-miRNA. It is known that Drosha and DGCR8 are involved in this processing. Thereafter, pre-miRNA is transported from the nucleus to the cytoplasm via Exportin-5, and then processed by Dicer and TRBP, and mature microRNA is biosynthesized from pre-miRNA (Non-patent Document 7).
 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。
 ヒトを含む哺乳類で発現するマイクロRNAが、肥満細胞の脱顆粒に影響を及ぼすことは知られている(特許文献1)が、マイクロRNAの生合成過程を広く阻害した場合に、肥満細胞がどのように反応するかついては知られていない。
Mature microRNAs are thought to be involved in post-transcriptional regulation of gene expression by binding complementarily to the target mRNA and suppressing mRNA translation or by degrading mRNA.
It is known that microRNAs expressed in mammals including humans have an effect on mast cell degranulation (Patent Document 1). It is not known how to react like that.
国際公開第2008/029790号パンフレットInternational Publication No. 2008/029790 Pamphlet
 本発明は、肥満細胞の脱顆粒抑制剤、肥満細胞の脱顆粒抑制方法、および肥満細胞の脱顆粒制御の異常に起因する疾患の治療薬の提供を課題とする。 An object of the present invention is to provide a degranulation inhibitor for mast cells, a method for inhibiting degranulation of mast cells, and a therapeutic agent for diseases caused by abnormal degranulation control of mast cells.
 本発明は以下に関する。
〔1〕 マイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質を有効成分として含有する、肥満細胞の脱顆粒抑制剤。
〔2〕 マイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質がDicer1遺伝子の発現を抑制する物質である、〔1〕に記載の脱顆粒抑制剤。
〔3〕 Dicer1遺伝子の発現を抑制する物質が核酸である、〔2〕に記載の脱顆粒抑制剤。
〔4〕 核酸がsiRNAである、〔3〕に記載の脱顆粒抑制剤。
〔5〕 Dicer1遺伝子の発現を抑制するsiRNAを発現するベクターを有効成分として含有する、肥満細胞の脱顆粒抑制剤。
〔6〕 siRNAが配列番号1~5のいずれかで表される塩基配列を標的配列とするsiRNAである、〔4〕または〔5〕に記載の脱顆粒抑制剤。
〔7〕 マイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質を用いることを特徴とする、肥満細胞の脱顆粒抑制方法。
〔8〕 マイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質が、Dicer1遺伝子の発現を抑制する物質である、〔7〕に記載の脱顆粒抑制方法。
〔9〕 Dicer1遺伝子の発現を抑制する物質が核酸である、〔8〕に記載の脱顆粒抑制方法。
〔10〕 核酸がsiRNAである、〔9〕に記載の脱顆粒抑制方法。
〔11〕 Dicer1遺伝子の発現を抑制するsiRNAを発現するベクターを用いることを特徴とする、肥満細胞の脱顆粒抑制方法。
〔12〕 siRNAが配列番号1~5のいずれかで表される塩基配列を標的配列とするsiRNAである、〔10〕または〔11〕に記載の脱顆粒抑制方法。
〔13〕 〔1〕~〔6〕のいずれかに記載の脱顆粒抑制剤を有効成分として含有する、肥満細胞の異常に起因する疾患の治療薬。
〔14〕 肥満細胞の異常に起因する疾患が、アトピー性皮膚炎、喘息、慢性閉塞性肺疾患およびアレルギー疾患からなる群から選ばれる疾患である、〔13〕に記載の治療薬。
〔15〕 〔1〕~〔6〕のいずれかに記載の脱顆粒抑制剤の有効量を、必要とする対象に投与することを含む、肥満細胞の異常に起因する疾患の治療方法。
〔16〕 肥満細胞の異常に起因する疾患が、アトピー性皮膚炎、喘息、慢性閉塞性肺疾患およびアレルギー疾患からなる群から選ばれる疾患である、〔15〕に記載の治療方法。
〔17〕 肥満細胞の異常に起因する疾患の治療薬の製造のための、〔1〕~〔6〕のいずれかに記載の脱顆粒抑制剤の使用。
〔18〕 肥満細胞の異常に起因する疾患が、アトピー性皮膚炎、喘息、慢性閉塞性肺疾患およびアレルギー疾患からなる群から選ばれる疾患である、〔17〕に記載の使用。
The present invention relates to the following.
[1] A mast cell degranulation inhibitor comprising as an active ingredient a substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor.
[2] The degranulation inhibitor according to [1], wherein the substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor is a substance that suppresses the expression of Dicer1 gene.
[3] The degranulation inhibitor according to [2], wherein the substance that suppresses the expression of the Dicer1 gene is a nucleic acid.
[4] The degranulation inhibitor according to [3], wherein the nucleic acid is siRNA.
[5] A mast cell degranulation inhibitor comprising, as an active ingredient, a vector that expresses siRNA that suppresses the expression of Dicer1 gene.
[6] The degranulation inhibitor according to [4] or [5], wherein the siRNA is a siRNA having a base sequence represented by any one of SEQ ID NOs: 1 to 5 as a target sequence.
[7] A method for inhibiting degranulation of mast cells, comprising using a substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor.
[8] The method for inhibiting degranulation according to [7], wherein the substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor is a substance that suppresses the expression of Dicer1 gene.
[9] The method for inhibiting degranulation according to [8], wherein the substance that suppresses the expression of the Dicer1 gene is a nucleic acid.
[10] The method for inhibiting degranulation according to [9], wherein the nucleic acid is siRNA.
[11] A method for inhibiting degranulation of mast cells, comprising using a vector that expresses siRNA that suppresses expression of Dicer1 gene.
[12] The method for inhibiting degranulation according to [10] or [11], wherein the siRNA is an siRNA having a base sequence represented by any one of SEQ ID NOs: 1 to 5 as a target sequence.
[13] A therapeutic agent for a disease caused by abnormal mast cells, comprising the degranulation inhibitor according to any one of [1] to [6] as an active ingredient.
[14] The therapeutic agent according to [13], wherein the disease caused by abnormality of mast cells is a disease selected from the group consisting of atopic dermatitis, asthma, chronic obstructive pulmonary disease and allergic disease.
[15] A method for treating a disease caused by abnormal mast cells, comprising administering an effective amount of the degranulation inhibitor according to any one of [1] to [6] to a subject in need thereof.
[16] The treatment method according to [15], wherein the disease caused by abnormality of mast cells is a disease selected from the group consisting of atopic dermatitis, asthma, chronic obstructive pulmonary disease, and allergic disease.
[17] Use of the degranulation inhibitor according to any one of [1] to [6] for the manufacture of a therapeutic agent for a disease caused by abnormality of mast cells.
[18] The use according to [17], wherein the disease caused by abnormality of mast cells is a disease selected from the group consisting of atopic dermatitis, asthma, chronic obstructive pulmonary disease, and allergic disease.
 本発明により、肥満細胞の脱顆粒抑制剤、肥満細胞の脱顆粒抑制方法、肥満細胞の異常に起因する疾患の治療薬を提供することができる。 According to the present invention, a degranulation inhibitor for mast cells, a method for inhibiting degranulation of mast cells, and a therapeutic drug for diseases caused by abnormal mast cells can be provided.
 本発明は、マイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質を有効成分として含有する、肥満細胞の脱顆粒抑制剤を提供する。 The present invention provides a mast cell degranulation inhibitor comprising as an active ingredient a substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor.
 本発明においてマイクロ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。本発明のマイクロRNAには成熟型マイクロRNAが含まれる。 In the present invention, microRNA refers to single-stranded RNA having a length of 17 to 28 bases. The surrounding genomic sequence including the sequence of the microRNA has a sequence capable of forming a hairpin structure, and the microRNA can be cut out from any one 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. The microRNA of the present invention includes mature microRNA.
 本発明においてマイクロRNA前駆体とは、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て一方の鎖にマイクロRNAが含有される核酸をいう。本発明のマイクロRNA前駆体には、primary-microRNA (pri-miRNA)、precursor-microRNA (pre-miRNA)が含まれる。 In the present invention, 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, and a hairpin structure is formed and microRNA is contained in one strand. Nucleic acid. The microRNA precursors of the present invention include primary-microRNA (pri-miRNA) and precursor-microRNA (pre-miRNA).
 マイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質としては、マイクロRNA前駆体からマイクロRNAの生合成に必須な因子(以下、マイクロRNAの生合成に必須な因子ともいう。)の機能を阻害する物質であれば、いかなる物質であってもよい。 As a substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor, a factor essential for biosynthesis of microRNA from the microRNA precursor (hereinafter also referred to as a factor essential for biosynthesis of microRNA). Any substance may be used as long as it inhibits the function of.
 マイクロRNAの生合成に必須な因子としては、例えば、pri-miRNAからpre-miRNAが生合成される過程に関与するDroshaおよびDGCR8、pre-miRNAが核内から細胞質に輸送される過程に関与するExportin-5、pre-miRNAから成熟型マイクロRNAが生合成される過程に関与するDicerおよびTRBPなどがあげられる。 Factors essential for microRNA biosynthesis include, for example, Drosha and DGCR8, which are involved in the process of pre-miRNA biosynthesis from pri-miRNA, and the process of transport of pre-miRNA from the nucleus to the cytoplasm Examples include Dicer and TRBP involved in the process of biosynthesis of mature microRNA from Exportin-5 and pre-miRNA.
 マイクロRNAの生合成に必須な因子の機能を阻害する物質としては、例えば該因子をコードする遺伝子の発現を阻害する物質、該因子の蛋白質の機能を阻害する物質であれば、核酸、抗体、低分子物質等、いずれであってもよいが、好ましくは核酸があげられる。 As a substance that inhibits the function of a factor essential for microRNA biosynthesis, for example, a substance that inhibits the expression of a gene encoding the factor, a substance that inhibits the function of the protein of the factor, a nucleic acid, an antibody, Any of low-molecular substances and the like may be used, but preferred is nucleic acid.
 マイクロRNAの生合成に必須な因子の機能を阻害する物質として、好ましくはDicerをコードする遺伝子(以下、Dicer遺伝子ともいう。)の発現を阻害する物質や、Dicerの蛋白質の機能を阻害する物質があげられる。 Substances that inhibit the function of factors essential for microRNA biosynthesis, preferably substances that inhibit the expression of the gene encoding Dicer (hereinafter also referred to as Dicer gene), and substances that inhibit the function of Dicer protein Can be given.
 Dicer遺伝子としては、Dicer1遺伝子、例えばヒトDicer1遺伝子、およびそれに対応する他生物種の相同遺伝子であるオーソログがあげられ、好ましくは配列番号6あるいは7で表される塩基配列に含まれるヒトDicer1遺伝子があげられる。また、Dicerの蛋白質としては、Dicer1があげられ、好ましくはヒトDicer1があげられる。 Examples of the Dicer gene include a Dicer1 gene, such as a human Dicer1 gene, and an ortholog that is a homologous gene of another species corresponding thereto, and preferably a human Dicer1 gene contained in the nucleotide sequence represented by SEQ ID NO: 6 or 7 can give. Moreover, Dicer1 is mentioned as a protein of Dicer, Preferably human Dicer1 is mention | raise | lifted.
 遺伝子の発現を阻害する物質としては、例えばmRNAの生合成や翻訳を抑制する物質、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物質があげられる。 Examples of substances that inhibit gene expression include substances that suppress the biosynthesis and translation of mRNA, and substances that reduce the amount of protein translated from mRNA by cleaving or decomposing mRNA.
 マイクロRNAの生合成に必須な因子の機能を阻害する核酸としては、例えば以下の(a)~(c)に記載の核酸があげられ、好ましくは(a)に記載の核酸があげられる。
(a)該因子をコードする遺伝子の標的核酸の塩基配列を含むsiRNA(短鎖干渉RNA;short interferance RNA)
(b)該因子をコードする遺伝子の転写産物またはその一部に対するアンチセンス核酸
(c)該因子をコードする遺伝子の転写産物を特異的に開裂するリボザイム活性を有する核酸
Examples of nucleic acids that inhibit the function of factors essential for microRNA biosynthesis include the nucleic acids described in the following (a) to (c), and preferably the nucleic acids described in (a).
(A) siRNA including a base sequence of a target nucleic acid of a gene encoding the factor (short interferance RNA)
(B) an antisense nucleic acid for the transcription product of the gene encoding the factor or a part thereof (c) a nucleic acid having a ribozyme activity that specifically cleaves the transcription product of the gene encoding the factor
 上記核酸としては、ヌクレオチドまたは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い。ヌクレオチドとしては、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、RNAおよびDNAが混合した重合体が、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としては、例えばヌクレオチド類似体を含むヌクレオチド重合体、核酸誘導体を含むヌクレオチド重合体が、それぞれあげられる。 The nucleic acid may be any molecule as long as it is a molecule obtained by polymerizing nucleotides or molecules having functions equivalent to those of the nucleotides. Examples of nucleotides include RNAs that are polymers of ribonucleotides, DNAs that are polymers of deoxyribonucleotides, polymers in which RNA and DNA are mixed, and molecules in which molecules having functions equivalent to the nucleotides are polymerized. Examples thereof include a nucleotide polymer containing a nucleotide analog and a nucleotide polymer containing a nucleic acid derivative.
 ヌクレオチド類似体としては、例えばRNAまたはDNAと比較して、ヌクレアーゼ耐性を向上または安定化させるため、相補鎖核酸とのアフィニティーをあげるため、あるいは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。 Nucleotide analogs include, for example, ribonucleic acid, to improve or stabilize nuclease resistance, increase affinity with complementary strand nucleic acids, increase cell permeability, or visualize, compared to RNA or DNA. Any molecule may be used as long as it is a modification of nucleotide, deoxyribonucleotide, 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), more specifically, 2′-position oxygen atom and 4′-position carbon Locked Nucleic Acid (LNA) and ethylene-bridged nucleic acid (ENA) [Nucleic Acid search Research, 32, e175 (2004)] 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.
 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、核酸の塩基部分、リボース部分、リン酸ジエステル結合部分等の原子(例えば、水素原子、酸素原子)もしくは官能基(例えば、水酸基、アミノ基)が他の原子(例えば、水素原子、硫黄原子)、官能基(例えば、アミノ基)、もしくは炭素数1~6のアルキル基で置換されたものまたは保護基(例えばメチル基またはアシル基)で保護されたもの、核酸に、例えば脂質、リン脂質、フェナジン、フォレート、フェナントリジン、アントラキノン、アクリジン、フルオレセイン、ローダミン、クマリン、色素など、別の化学物質を付加した分子等があげられる。また、核酸に別の化学物質を付加した分子としては、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もあげられる。 Examples of nucleic acid derivatives include atoms (for example, hydrogen atom, oxygen atom) or functional groups (for example, hydroxyl group, amino group) such as base portion, ribose portion, and phosphodiester bond portion of nucleic acid as other atoms (for example, hydrogen atom). , Sulfur atom), functional group (for example, amino group), or substituted with an alkyl group having 1 to 6 carbon atoms or protected with a protecting group (for example, methyl group or acyl group), nucleic acid, for example, lipid , Phospholipid, phenazine, folate, phenanthridine, anthraquinone, acridine, fluorescein, rhodamine, coumarin, and a molecule to which another chemical substance is added. In addition, examples of molecules obtained by adding another chemical substance to nucleic acid include 5′-polyamine addition derivatives, cholesterol addition derivatives, steroid addition derivatives, bile acid addition derivatives, vitamin addition derivatives, Cy5 addition derivatives, Cy3 addition derivatives, 6 -FAM addition derivatives, biotin addition derivatives, and the like are also included.
 マイクロRNAの生合成に必須な因子をコードする遺伝子の標的核酸の塩基配列を含むsiRNAとしては、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(RNA interferance;RNAi)により、該標的核酸の発現を抑制できるものであればいずれでもよい。siRNAの一方の鎖を構成する塩基数は、好ましくは17~30塩基、より好ましくは18~25塩基、さらに好ましくは19~23塩基である。 The siRNA containing the base sequence of the target nucleic acid of the gene encoding the factor essential for microRNA biosynthesis is a short double-stranded RNA containing the base sequence of a certain target nucleic acid, which is caused by RNA interference (RNAi). Any one can be used as long as it can suppress the expression of the target nucleic acid. The number of bases constituting one strand of siRNA is preferably 17 to 30 bases, more preferably 18 to 25 bases, and still more preferably 19 to 23 bases.
 siRNAは、RNAのみで構成されていてもよいが、標的核酸の発現を抑制できれば、DNA、RNAおよびDNAが混合した重合体、ヌクレオチド類似体を含むヌクレオチド重合体、核酸誘導体を含むヌクレオチド重合体に置換されたものなど、いずれの核酸であってもよい。 siRNA may be composed only of RNA, but as long as the expression of the target nucleic acid can be suppressed, DNA, RNA and DNA mixed polymers, nucleotide polymers including nucleotide analogs, nucleotide polymers including nucleic acid derivatives Any nucleic acid may be used, such as a substituted one.
 またsiRNAには、標的遺伝子発現の阻害を媒介する能力を調節するために、二重鎖分子中にミスマッチ、隆起、ループまたはゆらぎ塩基対を必要に応じて含ませてもよい。このようなミスマッチ、隆起、ループまたはゆらぎ塩基対は、siRNAが標的遺伝子発現の阻害を媒介する能力を有意に障害しないように、siRNAの末端領域または内部領域に配置することが好ましい。 The siRNA may also contain mismatches, bumps, loops, or wobble base pairs as necessary in the double-stranded molecule in order to regulate the ability to mediate inhibition of target gene expression. Such mismatches, bumps, loops or wobble base pairs are preferably placed in the terminal or internal region of the siRNA so that the siRNA does not significantly impair the ability to mediate inhibition of target gene expression.
 具体的には、末端に数塩基のオーバーハングを有する二本鎖RNAはRNAi効果が高いことが知られている。従って本発明で用いられる二本鎖RNAは、末端に数塩基のオーバーハングを有していてもよい。このオーバーハングを形成する塩基の長さ、および配列は特に制限されない。また、このオーバーハングは、DNAおよびRNAのいずれであってもよい。例えば2塩基のオーバーハングであるTT(チミンが2個)、UU(ウラシルが2個)、その他の塩基のオーバーハングがあげられる。例えば19塩基の二本鎖RNAと2塩基(TT)のオーバーハングを有する分子を好適に用いることができる。二本鎖RNAには、このようにオーバーハングを形成する塩基がDNAであるような分子も含まれる。 Specifically, it is known that a double-stranded RNA having an overhang of several bases at the end has a high RNAi effect. Therefore, the double-stranded RNA used in the present invention may have an overhang of several bases at the end. The length and sequence of the base that forms this overhang are not particularly limited. The overhang may be either DNA or RNA. For example, two base overhangs such as TT (two thymines), UU (two uracils), and other base overhangs. For example, a molecule having a 19-base double-stranded RNA and a 2-base (TT) overhang can be preferably used. Double-stranded RNA includes molecules in which the base that forms the overhang is DNA.
 標的核酸の発現を抑制させるsiRNAは、例えば該遺伝子に対するsiRNAを配列に基づいて設計するためのアルゴリズム[Nucleic Acids Res., 32, 936 (2004); Nature Biotechnology, 22, 326 (2004); Nature Biotechnology, 23, 995 (2005)]を用いることにより、設計することができる。上記アルゴリズムを用いて設計されたsiRNAはキアゲン社、Applied Biosystems社、Invitrogen社などから市販されており、これらを用いてもよい。 The siRNA that suppresses the expression of the target nucleic acid is, for example, an algorithm for designing an siRNA for the gene based on the sequence [Nucleic Acids Res., 32, 936 (2004); Nature Biotechnology, 22, 326 (2004); Nature Biotechnology , 23, 995 (2005)]. SiRNAs designed using the above algorithm are commercially available from Qiagen, Applied Biosystems, Invitrogen, etc., and these may be used.
 上記siRNAとして、好ましくはヒトDicer1遺伝子の標的核酸の塩基配列を含むsiRNAがあげられる。該siRNAとしては、細胞内にsiRNAが導入されることで内在するヒトDicer1遺伝子の発現抑制させるものであればいかなるものでもよい。 The siRNA preferably includes siRNA containing the base sequence of the target nucleic acid of human Dicer1 gene. The siRNA may be any siRNA that suppresses the expression of the endogenous human Dicer1 gene by introducing the siRNA into the cell.
 ヒトDicer1遺伝子等に対するsiRNAも、上記アルゴリズムを用いて設計することができる。ヒトDicer1遺伝子の発現量が低下するまたは抑制されるsiRNAとして、好ましくは、配列番号1、2、3[Molecular Cell 24, 157 (2006)]、4[Nature, 436, 740 (2005)]または5[Nature, 436, 740 (2005)]で表される塩基配列を標的配列とするsiRNAをあげることができる。 SiRNA against human Dicer1 gene and the like can also be designed using the above algorithm. The siRNA in which the expression level of the human Dicer1 gene is reduced or suppressed is preferably SEQ ID NO: 1, 2, 3 [Molecular Cell 24, 157 (2006)], 4 [Nature, 436, 740 (2005)] or 5 Examples include siRNAs having a base sequence represented by [Nature, 436, 740 (2005)] as a target sequence.
 例えば、上記配列番号1で表わされる塩基配列を標的配列とするsiRNAとしては、例えば以下に示す塩基配列をあげることができる。
アンチセンス鎖:5’-CUAGGAUCCAGAUAGCACAdTdT-3’(配列番号:8)
センス鎖:5’-UGUGCUAUCUGGAUCCUAGdTdT-3’ (配列番号:9)
For example, examples of the siRNA having the base sequence represented by SEQ ID NO: 1 as a target sequence include the base sequences shown below.
Antisense strand: 5'-CUAGGAUCCAGAUAGCACAdTdT-3 '(SEQ ID NO: 8)
Sense strand: 5'-UGUGCUAUCUGGAUCCUAGdTdT-3 '(SEQ ID NO: 9)
 また、上記配列番号2で表わされる塩基配列を標的配列とするsiRNAとしては、例えば以下に示す塩基配列をあげることができる。
アンチセンス鎖:5’-UCCAGAGCUGCUUCAAGCAdTdT-3’ (配列番号:10)
センス鎖:5’-UGCUUGAAGCAGCUCUGGAdTdT-3’ (配列番号:11)
Moreover, as siRNA which makes the base sequence represented by the said sequence number 2 a target sequence, the base sequence shown below can be mention | raise | lifted, for example.
Antisense strand: 5'-UCCAGAGCUGCUUCAAGCAdTdT-3 '(SEQ ID NO: 10)
Sense strand: 5'-UGCUUGAAGCAGCUCUGGAdTdT-3 '(SEQ ID NO: 11)
 なお上記配列番号:8~11における「dT」はDNAのT(オーバーハング)であり、その他はRNAである。 In the above SEQ ID NOs: 8 to 11, “dT” is T (overhang) of DNA, and the other is RNA.
 さらに本発明は、Dicer1遺伝子の発現を抑制するsiRNAを発現するベクターを有効成分として含有する、肥満細胞の脱顆粒抑制剤を提供する。上記siRNAを発現し得るベクターは、当業者においては、一般的な遺伝子工学技術により、容易に作製することができ、例えば後述の3に記載の発現ベクターを用いることができる。 Furthermore, the present invention provides a degranulation inhibitor for mast cells, containing as an active ingredient a vector that expresses siRNA that suppresses the expression of Dicer1 gene. Those skilled in the art can easily prepare a vector capable of expressing the siRNA by a general genetic engineering technique. For example, the expression vector described in 3 below can be used.
 マイクロRNAの生合成に必須な因子をコードする遺伝子の転写産物またはその一部に対するアンチセンス核酸としては、該因子をコードする遺伝子またはその一部と相補的な配列を有していることが好ましい。アンチセンス核酸は、該因子をコードする遺伝子の発現を有効に抑制できる限り、完全に相補的でなくてもよいが、該因子をコードする遺伝子の転写産物に対して好ましくは90%以上、さらに好ましくは95%以上の相補性を有する。アンチセンス核酸を用いて標的遺伝子の発現を効果的に抑制するには、アンチセンス核酸の長さは少なくとも15塩基以上であり、好ましくは100塩基以上であり、さらに好ましくは500塩基以上である。 The antisense nucleic acid for the transcription product of a gene encoding a factor essential for microRNA biosynthesis or a part thereof preferably has a sequence complementary to the gene encoding the factor or a part thereof. . The antisense nucleic acid may not be completely complementary as long as it can effectively suppress the expression of the gene encoding the factor, but is preferably 90% or more with respect to the transcription product of the gene encoding the factor. Preferably it has a complementarity of 95% or more. In order to effectively suppress the expression of a target gene using an antisense nucleic acid, the length of the antisense nucleic acid is at least 15 bases, preferably 100 bases or more, more preferably 500 bases or more.
 アンチセンス核酸としては、該因子をコードする遺伝子のmRNAの5'端近傍の非翻訳領域に相補的なアンチセンス配列を設計すれば、遺伝子の翻訳阻害に効果的と考えられる。また、コード領域もしくは3'側の非翻訳領域に相補的な配列も使用することができる。 As an antisense nucleic acid, if an antisense sequence complementary to the untranslated region near the 5 ′ end of the mRNA of the gene encoding the factor is designed, it is considered effective for inhibiting the translation of the gene. In addition, a sequence complementary to the coding region or the 3 ′ untranslated region can also be used.
 アンチセンス核酸は、例えば適当なプロモーターの下流に連結し、好ましくは3'側に転写終結シグナルを含む配列を連結した後に、公知の方法を用いて所望の動物へ形質転換することができる。 The antisense nucleic acid can be transformed into a desired animal using a known method after ligating, for example, downstream of an appropriate promoter, and preferably ligating a sequence containing a transcription termination signal on the 3 ′ side.
 マイクロRNAの生合成に必須な因子をコードする遺伝子の転写産物を特異的に開裂するリボザイム活性を有する核酸としては、触媒活性を有するRNA分子であればいずれでもよく、グループIイントロン型やRNase Pに含まれるM1 RNAのように400塩基以上の大きさのもの、ハンマーヘッド型やヘアピン型と呼ばれる40塩基程度の活性ドメインを有するもの等があげられる。 As a nucleic acid having ribozyme activity that specifically cleaves a transcript of a gene encoding a factor essential for microRNA biosynthesis, any RNA molecule having catalytic activity may be used, such as group I intron type or RNase P. Such as those having a size of 400 bases or more, such as the M1 RNA contained in, and those having an active domain of about 40 bases called hammerhead type or hairpin type.
 ハンマーヘッド型のリボザイムの自己切断ドメインは、G13U14C15という配列のC15の3'側を切断するが、その活性にはU14とA9との塩基対形成が重要とされ、C15の代わりにA15またはU15でも切断され得る[ FEBS Lett., 228, 228 (1988)]。基質結合部位が標的部位近傍のRNA配列と相補的なリボザイムを設計すれば、標的RNA中のUC、UUまたはUAという配列を認識する制限酵素的なRNA切断リボザイムを作出することができる[FEBS Lett., 239, 285 (1988); タンパク質核酸酵素, 35, 2191 (1990); Nucl. Acids Res, 17, 7059 (1989)]。 The self-cleaving domain of the hammerhead ribozyme cleaves 3 'of C15 in the sequence G13U14C15, but base pairing between U14 and A9 is important for its activity. Instead of C15, A15 or U15 Can be cut [FEBS Lett., 228, 228 (1988)]. By designing a ribozyme whose substrate binding site is complementary to the RNA sequence in the vicinity of the target site, it is possible to create a restriction enzyme-like RNA-cleaving ribozyme that recognizes the sequence UC, UU or UA in the target RNA [FEBSettLett , 239, 285 (1988); Protein Nucleic Acid Enzyme, 35, 2191 (1990); Nucl. Acids, Res 17, 7059 (1989)].
 ヘアピン型のリボザイムは、例えばタバコリングスポットウイルスのサテライトRNAのマイナス鎖に見出される[Nature, 323, 349 (1986)]。ヘアピン型のリボザイムから、公知の方法[Nucl. Acids Res, 19, 6751 (1991); 化学と生物, 30, 112 (1992)]を用いることにより、標的核酸の塩基配列に特異的なRNA切断リボザイムを作製してもよい。マイクロRNAの生合成に必須な因子の蛋白質の機能を阻害する物質としては、例えば以下の(a)に記載の抗体、または(b)に記載の低分子化合物をあげることができる。
(a)該因子と結合する抗体
(b)該因子と結合する低分子化合物
Hairpin ribozymes are found, for example, in the minus strand of satellite RNA of tobacco ring spot virus [Nature, 323, 349 (1986)]. By using a known method [Nucl. Acids Res, 19, 6751 (1991); Chemistry and Biology, 30, 112 (1992)] from a hairpin ribozyme, an RNA-cleaving ribozyme specific for the base sequence of the target nucleic acid May be produced. Examples of the substance that inhibits the function of the protein of the factor essential for microRNA biosynthesis include the antibody described in (a) below or the low molecular compound described in (b).
(A) an antibody that binds to the factor (b) a low molecular compound that binds to the factor
 マイクロRNAの生合成に必須な因子と結合する抗体は、該因子の天然の蛋白質の他、遺伝子組換え技術を利用して作製した組換え蛋白質を用いて、当業者に公知の方法により調製することができる。 Antibodies that bind to factors essential for biosynthesis of microRNAs are prepared by methods known to those skilled in the art using recombinant proteins prepared using genetic recombination techniques in addition to the natural proteins of the factors. be able to.
 ポリクローナル抗体であれば、例えば、次のようにして調製することができる。該因子の蛋白質、あるいはGSTとの融合蛋白質として大腸菌等の微生物において発現させた組換え蛋白質、またはその部分ペプチドをウサギ等の小動物に免疫し血清を得る。これを、例えば、硫安沈殿、プロテインA、プロテインGカラム、DEAEイオン交換クロマトグラフィー、該因子の蛋白質や合成ペプチドをカップリングしたアフィニティーカラム等により精製することにより調製できる。 For example, a polyclonal antibody can be prepared as follows. A small protein such as a rabbit is immunized with a recombinant protein expressed in a microorganism such as Escherichia coli or a partial peptide thereof as a protein of the factor or a fusion protein with GST to obtain serum. This can be prepared, for example, by purification using ammonium sulfate precipitation, protein A, protein G column, DEAE ion exchange chromatography, an affinity column coupled with the protein of the factor or a synthetic peptide, or the like.
 モノクローナル抗体であれば、例えば、該因子の蛋白質、もしくはその部分ペプチドをマウスなどの小動物に免疫を行い、同マウスより脾臓を摘出し、これをすりつぶして細胞を分離し、該細胞とマウスミエローマ細胞とをポリエチレングリコール等の試薬を用いて融合させ、これによりできた融合細胞(ハイブリドーマ)の中から、該因子と結合する抗体を産生するクローンを選択する。次いで、得られたハイブリドーマをマウス腹腔内に移植し、同マウスより腹水を回収し、得られたモノクローナル抗体を、例えば、硫安沈殿、プロテインA、プロテインGカラム、DEAEイオン交換クロマトグラフィー、該因子の蛋白質や合成ペプチドをカップリングしたアフィニティーカラム等により精製することで、調製できる。 In the case of a monoclonal antibody, for example, the protein of the factor or a partial peptide thereof is immunized to a small animal such as a mouse, the spleen is removed from the mouse, and this is ground to separate the cells, and the cells and mouse myeloma cells Are fused using a reagent such as polyethylene glycol, and a clone that produces an antibody that binds to the factor is selected from the fused cells (hybridoma). Next, the obtained hybridoma was transplanted into the abdominal cavity of the mouse, and ascites was collected from the mouse, and the obtained monoclonal antibody was obtained by, for example, ammonium sulfate precipitation, protein A, protein G column, DEAE ion exchange chromatography, It can be prepared by purification using an affinity column coupled with protein or synthetic peptide.
 上記抗体の形態には、特に制限はなく、該因子に結合する限り、上記ポリクローナル抗体、モノクローナル抗体のほかに、ヒト抗体、遺伝子組み換えによるヒト型化抗体、さらにその抗体断片や抗体修飾物も含まれる。 The form of the antibody is not particularly limited, and as long as it binds to the factor, in addition to the polyclonal antibody and the monoclonal antibody, a human antibody, a humanized antibody by genetic recombination, an antibody fragment thereof, and an antibody modification product are also included. It is.
 抗体取得の感作抗原として使用される該因子の蛋白質としては、その由来となる動物種に制限されないが哺乳動物、例えばマウス、ヒト由来の蛋白質が好ましく、特にヒト由来の蛋白質が好ましい。ヒト由来の蛋白質は、本明細書に開示される遺伝子配列を用いて得ることができる。 The protein of the factor used as a sensitizing antigen for antibody acquisition is not limited to the animal species from which it is derived, but is preferably a protein derived from a mammal such as a mouse or a human, and particularly preferably a protein derived from a human. Human-derived proteins can be obtained using the gene sequences disclosed in the present specification.
 上記感作抗原として使用される蛋白質は、完全な蛋白質あるいは蛋白質の部分ペプチドであってもよい。蛋白質の部分ペプチドとしては、例えば、蛋白質のアミノ基(N)末端断片やカルボキシ(C)末端断片があげられる。 The protein used as the sensitizing antigen may be a complete protein or a partial peptide of the protein. Examples of the partial peptide of the protein include an amino group (N) terminal fragment and a carboxy (C) terminal fragment of the protein.
 また、ヒト以外の動物に抗原を免疫して上記ハイブリドーマを得る他に、ヒトリンパ球、例えばEBウイルスに感染したヒトリンパ球をin vitroで蛋白質、蛋白質発現細胞またはその溶解物で感作し、感作リンパ球をヒト由来の永久分裂能を有するミエローマ細胞、例えばU266と融合させ、蛋白質への結合活性を有する所望のヒト抗体を産生するハイブリドーマを得ることもできる。 In addition to immunizing non-human animals with antigens to obtain the above hybridomas, human lymphocytes such as human lymphocytes infected with EB virus are sensitized with proteins, protein-expressing cells or lysates thereof in vitro. Lymphocytes can be fused with human-derived myeloma cells having permanent mitotic activity, such as U266, to obtain hybridomas that produce desired human antibodies having protein-binding activity.
 該因子の蛋白質と結合する抗体を人体に投与する目的(抗体治療)で使用する場合には、免疫原性を低下させるため、ヒト抗体やヒト型抗体が好ましい。 When an antibody that binds to the protein of the factor is used for the purpose of administering it to the human body (antibody treatment), a human antibody or a human type antibody is preferable in order to reduce immunogenicity.
 該因子の蛋白質の機能を阻害し得る物質としては、該因子の蛋白質に結合する低分子量物質もあげられる。該因子の蛋白質に結合する低分子量物質は、天然または人工の化合物であってもよい。通常、当業者に公知の方法を用いることによって製造または取得可能な化合物である。
 上記の抗体または低分子化合物が結合する因子としては、Dicerが好ましく、Dicer1がより好ましく、ヒトDicer1がさらに好ましい。
Examples of the substance that can inhibit the function of the protein of the factor include low molecular weight substances that bind to the protein of the factor. The low molecular weight substance that binds to the protein of the factor may be a natural or artificial compound. Usually, it is a compound that can be produced or obtained by using methods known to those skilled in the art.
As a factor to which the above antibody or low molecular weight compound binds, Dicer is preferable, Dicer1 is more preferable, and human Dicer1 is more preferable.
 また本発明は、マイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質を用いることを特徴とする、肥満細胞の脱顆粒抑制方法を提供する。 The present invention also provides a method for suppressing degranulation of mast cells, characterized by using a substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor.
 本発明の脱顆粒抑制方法において、マイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質は、上述の脱顆粒抑制剤におけるマイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質と同様である。 In the degranulation suppression method of the present invention, the substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor inhibits the process of biosynthesis of microRNA from the microRNA precursor in the degranulation inhibitor described above. It is the same as the substance to do.
 また本発明は、Dicer1遺伝子の発現を抑制するsiRNAを発現するベクターを用いることを特徴とする、肥満細胞の脱顆粒抑制方法を提供する。
 本発明の脱顆粒抑制方法において用いられるsiRNAは、好ましくは、配列番号1~5のいずれかで表される塩基配列を標的配列とするsiRNAである。
The present invention also provides a method for inhibiting degranulation of mast cells, characterized by using a vector that expresses siRNA that suppresses the expression of Dicer1 gene.
The siRNA used in the degranulation suppressing method of the present invention is preferably an siRNA having a base sequence represented by any one of SEQ ID NOs: 1 to 5 as a target sequence.
 また本発明によって提供される肥満細胞の脱顆粒抑制剤は、肥満細胞の脱顆粒を抑制することから、肥満細胞の異常に起因する疾患の治療効果を有することが期待される。従って本発明は、本発明の肥満細胞の脱顆粒抑制剤を有効成分として含有する、肥満細胞の異常に起因する疾患の治療薬(医薬組成物)に関する。本発明の脱顆粒抑制剤は、肥満細胞の異常に起因する疾患の治療だけでなく、診断にも用いることができる。 Also, the mast cell degranulation inhibitor provided by the present invention suppresses mast cell degranulation, and is therefore expected to have a therapeutic effect on diseases caused by mast cell abnormalities. Therefore, this invention relates to the therapeutic agent (pharmaceutical composition) of the disease resulting from the abnormality of a mast cell which contains the degranulation inhibitor of the mast cell of this invention as an active ingredient. The degranulation inhibitor of the present invention can be used not only for treatment of diseases caused by abnormalities of mast cells but also for diagnosis.
 また本発明によって提供される脱顆粒抑制剤は、上述のように肥満細胞の異常に起因する疾患の治療効果を有することが期待されることから、当該脱顆粒抑制剤の有効量を必要とする対象(例えば、肥満細胞の異常に起因する疾患を有する患者等)に投与することによって、肥満細胞の異常に起因する疾患を治療することができる。即ち本発明は、本発明の脱顆粒抑制剤の有効量を、必要とする対象に投与することを含む、肥満細胞の異常に起因する疾患の治療方法に関する。 Moreover, since the degranulation inhibitor provided by the present invention is expected to have a therapeutic effect on diseases caused by abnormalities of mast cells as described above, an effective amount of the degranulation inhibitor is required. By administering to a subject (for example, a patient having a disease caused by abnormal mast cells), a disease caused by abnormal mast cells can be treated. That is, the present invention relates to a method for treating a disease caused by abnormal mast cells, comprising administering an effective amount of the degranulation inhibitor of the present invention to a subject in need.
 また本発明は、肥満細胞の異常に起因する疾患の治療薬を製造するための、本発明の脱顆粒抑制剤の使用に関する。また本発明は、本発明の脱顆粒抑制剤の、肥満細胞の異常に起因する疾患の治療薬の製造における使用に関する。 The present invention also relates to the use of the degranulation inhibitor of the present invention for producing a therapeutic agent for diseases caused by abnormalities of mast cells. The present invention also relates to the use of the degranulation inhibitor of the present invention in the manufacture of a therapeutic agent for diseases caused by abnormal mast cells.
 肥満細胞の異常としては、例えば肥満細胞の分化や脱顆粒、炎症性メディエーター産生、サイトカイン産生、ケモカイン産生等の異常があげられる。肥満細胞の異常に起因する疾患としては、具体的には、アトピー性皮膚炎、喘息、慢性閉塞性肺疾患、およびアレルギー疾患等をあげることができる。 Examples of abnormalities in mast cells include abnormalities such as mast cell differentiation and degranulation, inflammatory mediator production, cytokine production, and chemokine production. Specific examples of diseases caused by abnormal mast cells include atopic dermatitis, asthma, chronic obstructive pulmonary disease, and allergic diseases.
 以下、本発明を詳細に説明する。
1.肥満細胞の活性化の程度を測定する方法
(1-1)肥満細胞の取得、培養
 本発明で用いられる肥満細胞は、ヒト肥満細胞であることが好ましい。
 ヒト肥満細胞は、安全かつ効率的に取得される方法であれば特に限定されないが、例えば、ヒトの肺、皮膚、胎児肝臓などから公知の方法[J. Immunol. Methods, 169, 153 (1994); J. Immunol., 138, 861 (1987); J. Allergy Clin. Immunol., 107, 322 (2001); J. Immunol. Methods., 240, 101 (2000)]により調製することができる。また、公知の方法[J. Immunol., 157, 343, (1996); Blood, 91, 187 (1998); J. Allergy Clin. Immunol., 106, 141 (2000); Blood, 97, 1016 (2001); Blood, 98, 1127 (2001); Blood, 100, 3861 (2002); Blood, 97, 2045 (2001)]に従って、ヒトの臍帯血、末梢血、骨髄、肺あるいは皮膚から調製した単核球を、幹細胞因子(以下、SCFと略す)の存在下で培養し、肥満細胞に分化させることにより、調製することができる。
Hereinafter, the present invention will be described in detail.
1. Method for measuring the degree of mast cell activation (1-1) Acquisition and culture of mast cells The mast cells used in the present invention are preferably human mast cells.
Human mast cells are not particularly limited as long as they are safe and efficient, and for example, known methods from human lung, skin, fetal liver and the like [J. Immunol. Methods, 169, 153 (1994) J. Immunol., 138, 861 (1987); J. Allergy Clin. Immunol., 107, 322 (2001); J. Immunol. Methods., 240, 101 (2000)]. Further, known methods [J. Immunol., 157, 343, (1996); Blood, 91, 187 (1998); J. Allergy Clin. Immunol., 106, 141 (2000); Blood, 97, 1016 (2001) ); Blood, 98, 1127 (2001); Blood, 100, 3861 (2002); Blood, 97, 2045 (2001)], mononuclear cells prepared from human umbilical cord blood, peripheral blood, bone marrow, lung or skin Can be prepared by culturing in the presence of stem cell factor (hereinafter abbreviated as SCF) and differentiating into mast cells.
 また、ヒト肥満細胞から樹立した細胞株を用いることもでき、ヒト肥満細胞株としては、ヒト肥満細胞の性質をよく保持していることが知られているLAD2[Leuk. Res., 27, 671 (2003); Leuk. Res., 27, 677 (2003)]等があげられる。 In addition, a cell line established from human mast cells can also be used. As human mast cell lines, LAD2 [Leuk. Res., 27, 671, which is known to retain the properties of human mast cells, is known. (2003); Leuk. Res., 27, 677 (2003)].
(1-2)肥満細胞の活性化程度の測定
 ある物質が肥満細胞に対し、活性化抑制、脱顆粒抑制、炎症性メディエーター産生抑制、サイトカイン産生抑制およびケモカイン産生抑制のうちの少なくとも1つの作用を有していることは、例えば、本発明のマイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質を肥満細胞に導入した後、肥満細胞を刺激して、放出された(i)脱顆粒の指標となるヒスタミンやβ-ヘキソサミニダーゼ、(ii)LTC4、LTD4、LTE4、PGD2等の炎症性メディエーター、(iii)TNF-αやGM-CSF等のサイトカイン、(iv)IL-8、I-309、MIP-1α等のケモカイン等を測定し、本発明のマイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質を導入しなかった場合と比較することにより確認できる。
(1-2) Measurement of the degree of activation of mast cells A substance has at least one of the following actions on mast cells: inhibition of activation, inhibition of degranulation, production of inflammatory mediators, inhibition of cytokine production and inhibition of chemokine production. Having, for example, after introducing a substance that inhibits the process of biosynthesis of microRNA from the microRNA precursor of the present invention into mast cells, the mast cells were stimulated and released (i) Histamine and β-hexosaminidase as indicators of degranulation, (ii) Inflammatory mediators such as LTC4, LTD4, LTE4, PGD2, (iii) Cytokines such as TNF-α and GM-CSF, (iv) IL- 8, It can be confirmed by measuring chemokines such as I-309, MIP-1α, etc., and comparing with a case where a substance that inhibits the process of biosynthesis of microRNA from the microRNA precursor of the present invention was not introduced. .
 肥満細胞を刺激する方法としては、例えばIgEを添加して培養した後に抗IgE抗体を添加する方法や、コンパウンド48/80を添加する方法、ポリミキシンBを添加する方法、デキストランを添加する方法、カルシウムイオノフォアを添加する方法、アセチルコリンを添加する方法、カルバコールを添加する方法、トロンビンを添加する方法、コンカナバリンAを添加する方法、カルシウムイオノフォアを添加する方法、ATPを添加する方法、ドキソルビシンを添加する方法、等があげられる。 Methods for stimulating mast cells include, for example, a method of adding anti-IgE antibody after incubation with IgE, a method of adding compound 48/80, a method of adding polymyxin B, a method of adding dextran, calcium Method of adding ionophore, method of adding acetylcholine, method of adding carbachol, method of adding thrombin, method of adding concanavalin A, method of adding calcium ionophore, method of adding ATP, method of adding doxorubicin, Etc.
 肥満細胞の活性化は、脱顆粒の代わりに、TNF-αやGM-CSF等のサイトカイン産生、IL-8、I-309、MIP-1α等のケモカイン産生、LTC4、LTD4、LTE4、PGD2等の炎症メディエーター産生等を測定することによっても調べることができる[Blood, 100, 3861(2002)]。 Instead of degranulation, the activation of mast cells involves cytokine production such as TNF-α and GM-CSF, chemokine production such as IL-8, I-309, MIP-1α, LTC4, LTD4, LTE4, PGD2, etc. It can also be examined by measuring inflammatory mediator production etc. [Blood, 100, 3861 (2002)].
2.肥満細胞で発現するマイクロRNA、マイクロRNA前駆体などの核酸の発現を検出する方法
(2-1)RNAの取得
 肥満細胞から全RNAを抽出する方法は、マイクロRNAなどの低分子RNAが含まれる方法であれば特に限定されないが、例えば、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うことができる。あるいは、Trizol(Invitrogen社製)、ISOGEN(ニッポンジーン社製)、mirVanaTM miRNA Isolation Kit(Ambion社製)、miRNeasy Mini Kit(キアゲン社製)などを用いて抽出することもできる。
2. Methods for detecting the expression of nucleic acids such as microRNAs and microRNA precursors expressed in mast cells (2-1) Acquisition of RNA Methods for extracting total RNA from mast cells include small RNAs such as microRNAs Although it will not specifically limit if it is a method, For example, it can carry out by the method as described in Molecular Cloning 3rd edition. Alternatively, extraction can be performed using Trizol (Invitrogen), ISOGEN (Nippon Gene), mirVana miRNA Isolation Kit (Ambion), miRNeasy Mini Kit (Qiagen), and the like.
 また、低分子RNAを含む全RNAから低分子RNAをクローニングすることもできる。低分子RNAのクローニング法としては、具体的には、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る低分子RNAの分離及び切り出し、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方法等があげられる。あるいは、例えば、Science, 294, 858-862 (2001)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る低分子RNAの分離および切り出し、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方法等があげられる。 Also, low molecular weight RNA can be cloned from total RNA including low molecular weight RNA. As a method for cloning small RNA, specifically, according to the method described in Genes & Development, 15, 188-200 (2000), separation and excision of low molecular RNA by 15% polyacrylamide gel electrophoresis, 5'-terminal dephosphorylation, 3'-adapter ligation, phosphorylation, 5'-adapter ligation, reverse transcription, PCR amplification, concatamerization, ligation to vector, and then cloning of small RNA, the base of the clone Examples include a method for determining the sequence. Alternatively, for example, according to the method described in Science, 294, 858-862 (2001), separation and excision of small RNA by 15% polyacrylamide gel electrophoresis, 5′-adenylation 3′-adapter ligation, 5 '-Adapter ligation, reverse transcription, PCR amplification, concatamerization, ligation to a vector, cloning of low molecular RNA, and determination of the base sequence of the clone, etc. can be mentioned.
 あるいは、Nucleic Acids Research, 34, 1765-1771 (2006)に記載の方法により、15%ポリアクリルアミドゲル電気泳動による低分子RNAの分離および切り出し、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を取得することもできる。 Alternatively, separation and excision of small RNA by 15% polyacrylamide gel electrophoresis, 5′-end dephosphorylation, 3′-adapter ligation, phosphorylation by the method described in Nucleic® Acids® Research, 34, 1765-1771 (2006) Low molecular weight is determined by cloning low molecular RNA through sequential oxidation, 5'-adapter ligation, reverse transcription, PCR amplification, and ligation to a microbead vector, and determining the base sequence by reading the base sequence of the microbead. RNA can also be obtained.
 また、small RNA Cloning Kit(タカラバイオ社製)を用いて低分子RNAをクローニングすることもできる。 Also, small RNAs can be cloned using small RNA Cloning Kit (Takara Bio Inc.).
(2-2)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、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。
(2-2) Identification of microRNA Whether a small RNA sequence is a microRNA can be determined by following the criteria described in 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塩基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、且つ該低分子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miRBase (http://microrna.sanger.ac.uk/) 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。 Obtain a peripheral genomic sequence obtained by extending the DNA sequence corresponding to the obtained low molecular RNA base sequence by about 50 bases to the 5 'end and 3' end, respectively, and RNA that is 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 low molecular weight RNA is located on one strand of the hairpin, it can be determined that the low molecular weight RNA is a microRNA. Genome sequences are publicly available and can be obtained from, for example, UCSC Genome Bioinformatics (http://genome.ucsc.edu/). Also, 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. In addition, existing microRNA sequences are registered in the database miRBase (http://microrna.sanger.ac.uk/) 、. It can be determined whether or not.
(2-3)マイクロRNAまたはその前駆体などの発現量の検出法
 マイクロRNAまたはその前駆体など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。
(2-3) Method for Detecting Expression Level of MicroRNA or its Precursor Examples Methods for detecting the expression level of microRNA or its precursor include, for example, (1) Northern hybridization, (2) Dot blot high Hybridization, (3) in situ hybridization, (4) quantitative PCR, (5) differential hybridization, (6) microarray, (7) ribonuclease protection assay and the like.
 ノーザンハイブリダイゼーションは、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、該マイクロRNAまたはその前駆体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、該マイクロRNAまたはその前駆体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 hybridization involves separating the sample-derived RNA by gel electrophoresis and then transferring it to a support such as a nylon filter to prepare a probe appropriately labeled based on the base sequence of the microRNA or its precursor. It is a method for detecting a band specifically bound to the microRNA or its precursor by performing hybridization and washing, and specifically, for example, the method described in Science, 294, 853-858 (2001) Etc. can be performed according to the above.
 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、該マイクロまたはそのRNA前駆体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マイクロRNAまたはその前駆体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、該マイクロRNAまたはその前駆体の発現量を定量することができる。電気泳動、メンブレンの移行、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版に記載の方法により行うことができる。 The labeled probe can be prepared by, for example, radioisotope, biotin, digoxigenin, fluorescent group, chemiluminescent group, etc. by the method such as nick translation, random priming, or phosphorylation at the 5 ′ end, and the micro or its RNA precursor. It can be prepared by incorporating it into DNA, RNA or LNA having a sequence complementary to the base sequence. Since the binding amount of the labeled probe reflects the expression level of the microRNA or its precursor, the expression level of the microRNA or its precursor can be determined by quantifying the amount of the labeled probe bound. . Electrophoresis, membrane transfer, probe preparation, hybridization, and nucleic acid detection can be performed by the methods described in Molecular Cloning 3rd edition.
 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をメンブラン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 or 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ハイブリダイゼーションならびに洗浄の工程を行い、顕微鏡観察を行うことにより、該マイクロRNAまたはその前駆体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 paraffin or cryostat sections of tissue obtained from a living body, or immobilized cells as specimens, performs hybridization and washing steps with a labeled probe, and performs microscopic observation. This is a method for investigating the distribution and localization of RNA or its precursor in tissues and 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ともいう)が測定に用いられる。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、該マイクロRNAまたはその前駆体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 also referred to as a sample-derived cDNA) is used for measurement. A random primer or a specific RT primer can be used as a reverse transcription primer for cDNA synthesis. The specific RT primer refers to a primer having a sequence complementary to a base sequence corresponding to the microRNA or a precursor thereof and a surrounding genomic sequence.
 例えば、検体由来cDNAを合成後、これを鋳型とし、該マイクロRNAまたはその前駆体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、該マイクロまたはそのRNA前駆体を含む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に含まれる該マイクロRNAまたはその前駆体の量を検出する。鋳型特異的なプライマーとしては、該マイクロRNAまたはその前駆体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20~40塩基の配列からなるDNAまたはLNA、および3'端20~40塩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 primer designed from the base sequence corresponding to the microRNA or its precursor and its surrounding genomic sequence, or the base sequence corresponding to the reverse transcription primer PCR is used to amplify the cDNA fragment containing the micro or its RNA precursor, and the amount of the micro RNA or its precursor contained in the sample-derived RNA is detected from the number of cycles to reach a certain amount To do. As a template-specific primer, an appropriate region corresponding to the microRNA or its precursor and its surrounding genomic sequence is selected, and DNA or LNA comprising a sequence of 20 to 40 bases at the 5 ′ end of the base sequence of the region And a DNA or LNA pair consisting of a sequence complementary to 20 to 40 bases at the 3 ′ end can be used. Specifically, it can be carried out according to the method described in Nucleic Acids Research, 32, e43 (2004).
 または、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。 Alternatively, a specific RT primer having a stem / loop structure can also be used as a reverse transcription primer for cDNA synthesis. Specifically, it can be carried out using the method described in Nucleic® Acid Research, 33, 179 (2005) or TaqMan® MicroRNA Assays (Applied Biosystems).
 更に、該マイクロRNAまたはその前駆体を少なくとも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盤に対して、検体由来cDNAをハイブリダイゼーションし、洗浄を行うことにより、該マイクロRNAまたはその前駆体の量の変動を検出することができる。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盤上にU6RNA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該マイクロRNAまたはその前駆体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、該マイクロRNAまたはその前駆体の定量を行うことができる。例えば、Proc.Natl.Acad.Sci. USA, 101, 9740-9744 (2004)やNucleic Acid Research, 32, e188 (2004)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)と同様にして検出または定量することができる。 Furthermore, sample-derived cDNA is hybridized to a filter or slide glass, silicon, or other substrate on which DNA corresponding to a base sequence containing at least one of the microRNAs or a precursor thereof, or LNA is immobilized, and washed. By performing the above, it is possible to detect fluctuations in the amount of the microRNA or its precursor. Examples of methods based on such hybridization include methods using differential hybridization [Trends Genet., 7, 314 (1991)] and microarrays [Genome Res., 6, 639 (1996)]. Each method immobilizes an internal control such as a nucleotide sequence corresponding to U6RNA on a filter or substrate to accurately detect the difference in the amount of the microRNA or its precursor between the control sample and the target sample. can do. 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. By simultaneously hybridizing the labeled cDNA, the microRNA or a precursor thereof can be quantified. For example, microRNAs can be detected using a microarray described in Proc. Natl. Acad. Sci. USA, 101, 9740-9744 (2004), Nucleic Acid Research, 32, e188 (2004). Specifically, it can be detected or quantified in the same manner as mirVana miRNA Bioarray (Ambion).
 リボヌクレアーゼ保護アッセイでは、まず該マイクロRNAまたはその前駆体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、一本鎖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、本発明の核酸またはマイクロRNA前駆体として、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 or SP6 promoter is bound to the 3 ′ end of the base sequence corresponding to the microRNA or a precursor thereof or the surrounding genomic sequence, and labeled NTP (ATP, GTP, CTP). , UTP mixture) and an in vitro transcription system using RNA polymerase to synthesize labeled antisense RNA. 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 digested product is subjected to gel electrophoresis, and an RNA fragment protected from digestion by forming an RNA-RNA hybrid is detected or quantified as the nucleic acid or microRNA precursor of the present invention. Specifically, it can be detected or quantified using mirVana miRNA Detection Kit (Ambion).
3.マイクロRNAの生合成される過程を阻害する方法
 マイクロRNAは、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から生合成され、その後、核内からExportin-5を介して細胞質に輸送された後、最終的にpre-miRNAから成熟型マイクロRNAが生合成される。マイクロRNAの生合成を阻害する方法としては、上記過程のいずれかが阻害されて、成熟型マイクロRNA量が減少すれば、いかなる方法でもよい。
3. How to inhibit the process of biosynthesis of microRNAs MicroRNAs are transcribed from a gene into a primary transcript, primary-microRNA (pri-miRNA), followed by step-wise processing from pri-miRNA to mature microRNA. About 70 bases of precursor-microRNA (pre-miRNA) with a characteristic hairpin structure was biosynthesized from pri-miRNA, then transported from the nucleus to the cytoplasm via Exportin-5, and finally Mature microRNA is biosynthesized from pre-miRNA. As a method for inhibiting the biosynthesis of microRNA, any method may be used as long as one of the above processes is inhibited and the amount of mature microRNA decreases.
 具体的には当該方法には、上述のマイクロRNA生合成に必須な因子の機能を阻害する物質を用いることができる。 Specifically, a substance that inhibits the function of the factor essential for the above-described microRNA biosynthesis can be used in the method.
 また、発現ベクターに標的塩基配列から選択した塩基配列に相当するDNAを挿入することにより、siRNAを発現するベクターを作製することもできる。発現ベクターとしては、宿主細胞において自立複製可能でかつ染色体中への組込みが可能で、核酸の塩基配列を含むゲノム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U6RNAやH1RNA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)等を例示することができる。pol III系プロモーターとしてはU6RNAやH1RNAあるいはtRNA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)等をあげることができる。 Also, a vector that expresses siRNA can be prepared by inserting a DNA corresponding to a base sequence selected from a target base sequence into an expression vector. As the expression vector, a vector that can replicate autonomously in a host cell, can be integrated into a chromosome, and contains a promoter at a position where a genomic gene containing a nucleic acid base sequence can be transcribed is used. As the promoter, any promoter can be used so long as it can be expressed in the host cell. For example, RNA polymerase II (pol II) type promoter or U RNA or H1RNA transcription system such as RNA polymerase III (pol III) type promoter. Can give. Examples of the pol II promoter include cytomegalovirus (human CMV) IE (immediate early) gene promoter, SV40 early promoter, 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 promoters. Examples of expression vectors using them include pSINsi-hH1 DNA (Takara Bio), pSINsi-hU6 DNA (Takara Bio), pENTR / U6 (Invitrogen) and the like.
 更には、上記の選択した塩基配列に相当するDNA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siRNAを発現させることもできる。パッケージング細胞はウィルスのパッケジ-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、マウス繊維芽細胞NIH3T3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。 Further, a DNA corresponding to the selected base sequence is inserted downstream of the promoter in the viral vector to construct a recombinant viral vector, and the vector is introduced into a packaging cell to produce a recombinant virus. The siRNA 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 viral packaging, such as human kidney. HEK293 cells derived from mouse, mouse fibroblast NIH3T3, 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., adenovirus vectors, E1A, E1B, etc. Can be used.
 実際にマイクロRNAの生合成が阻害されたかどうかは、任意のマイクロRNAに対する発現量を上記2に記載の方法に従って測定することで、確認することができる。また、マイクロRNAの生合成に必須な因子のmRNA量や蛋白量を、それぞれリアルタイムRT-PCR法やwestern blotting法にて測定し、それらの量が低下したか否かで確認することもできる。 Whether or not the biosynthesis of microRNA is actually inhibited can be confirmed by measuring the expression level for any microRNA according to the method described in 2 above. In addition, the amount of mRNA and protein of factors essential for biosynthesis of microRNA can be measured by a real-time RT-PCR method or western blotting method, respectively, and it can be confirmed whether or not these amounts have decreased.
4.マイクロRNAの生合成を阻害する物質を用いた肥満細胞の脱顆粒抑制剤
 本発明の、マイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質は、肥満細胞の脱顆粒抑制剤として利用することができる。マイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質は、上記3で記載したように、マイクロRNAが生合成に必須な因子の機能を阻害する物質であれば、いかなる物質でも良いが、例えば、ヒトDicer1遺伝子に対するsiRNAなどがあげられる。
4). Mast cell degranulation inhibitor using a substance that inhibits microRNA biosynthesis The substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor according to the present invention is used as a mast cell degranulation inhibitor. Can be used. As described in 3 above, the substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor may be any substance as long as it inhibits the function of factors essential for biosynthesis. However, for example, siRNA for human Dicer1 gene can be mentioned.
 本発明のマイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質の肥満細胞の脱顆粒抑制効果については、上記1で記載したように、本発明のマイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質を肥満細胞に導入した後、肥満細胞を刺激して、放出された(i)脱顆粒の指標となるヒスタミンやβ-ヘキソサミニダーゼ、(ii)LTC4、LTD4、LTE4、PGD2等の炎症性メディエーター、(iii)TNF-αやGM-CSF等のサイトカイン、(iv)IL-8、I-309、MIP-1α等のケモカイン等を測定し、本発明のマイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質を導入しなかった場合と比較することにより確認できる。 Regarding the degranulation inhibitory effect of mast cells, which is a substance that inhibits the process of biosynthesis of microRNA from the microRNA precursor of the present invention, as described in 1 above, microRNA from the microRNA precursor of the present invention After introducing into the mast cells a substance that inhibits the process of biosynthesis, the mast cells are stimulated and released (i) histamine and β-hexosaminidase, which are indicators of degranulation, (ii) LTC4, Measure inflammatory mediators such as LTD4, LTE4, PGD2, etc., (iii) cytokines such as TNF-α and GM-CSF, (iv) chemokines such as IL-8, I-309, MIP-1α, etc. This can be confirmed by comparing with a case where a substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor was not introduced.
 本発明の肥満細胞の脱顆粒抑制剤の製剤形態や、投与方法などについては、5で後述する、マイクロRNAの生合成を阻害する物質を含有する治療薬と同様である。 The formulation form and administration method of the mast cell degranulation inhibitor of the present invention are the same as those of the therapeutic agent containing a substance that inhibits biosynthesis of microRNA, which will be described later in 5.
5.マイクロRNAの生合成を阻害する物質を含有する治療薬
 マイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質は、肥満細胞の脱顆粒を抑制することにより、肥満細胞の異常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。肥満細胞の異常としては、肥満細胞の分化や脱顆粒、炎症性メディエーター産生、サイトカイン産生、ケモカイン産生等の異常があげられ、それに起因する疾患として、アトピー性皮膚炎、喘息、慢性閉塞性肺疾患、アレルギー性疾患等をあげることができる。
5). Therapeutic drugs containing substances that inhibit the biosynthesis of microRNA Substances that inhibit the process of biosynthesis of microRNAs from microRNA precursors can suppress mast cell degranulation, thereby preventing abnormalities in mast cells. It can be used as a therapeutic agent for diseases caused by it. Abnormalities in mast cells include abnormalities such as mast cell differentiation and degranulation, inflammatory mediator production, cytokine production, chemokine production, etc. Diseases resulting from them include atopic dermatitis, asthma, chronic obstructive pulmonary disease And allergic diseases.
 マイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質を有効成分として含有する治療剤は、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。 A therapeutic agent containing as an active ingredient a substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor can be administered alone, but usually one or more pharmacologically acceptable It is desirable to administer it as a pharmaceutical preparation prepared by any method well known in the pharmaceutical arts.
 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。 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、座剤、注射剤、軟膏、テープ剤などがあげられる。 Administration forms include sprays, capsules, tablets, granules, syrups, emulsions, suppositories, injections, ointments, tapes and the like.
 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。 Preparations suitable for oral administration include emulsions, syrups, capsules, tablets, powders, granules and the like.
 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。 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、座剤、噴霧剤などがあげられる。 Preparations suitable for parenteral administration include injections, suppositories, sprays and the like.
 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。座剤はカカオ脂、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。 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 cocoa 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。上記本発明の脱顆粒抑制剤、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。 Specific examples of the carrier include lactose and glycerin. Depending on the properties of the degranulation inhibitor of the present invention and the carrier used, preparations such as aerosols and dry powders are possible. In these parenteral preparations, the components exemplified as additives for oral preparations can also be added.
 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10μg/kg~20 mg/kgである。 The dose or frequency of administration varies depending on the intended therapeutic effect, administration method, treatment period, age, body weight, etc., but is usually 10 μg / kg to 20 mg / kg per day for an adult.
 また、マイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質などの上記本発明の脱顆粒抑制剤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)]。 The therapeutic agent containing the degranulation inhibitor of the present invention as an active ingredient, such as a substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor, is used as a nucleic acid-expressing vector and nucleic acid therapeutic agent. It can also be produced by blending with a base [Nature Genet., 8, (42 (1994)].
 本発明の治療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。本発明の治療剤は、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。 The base used in the therapeutic agent of the present invention may be any base as long as it is usually used in injections, salt water such as distilled water, sodium chloride or a mixture of sodium chloride and an inorganic salt, mannitol, Examples thereof include a solution of lactose, dextran, glucose and the like, 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 of the present invention 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.
 マイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質が核酸(例えばsiRNAなど)の場合、該核酸を発現するベクターは、上記3で作製した組換えウィルスベクター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。 When the substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor is a nucleic acid (for example, siRNA), the vector that expresses the nucleic acid can be the recombinant virus vector prepared in 3 above. More specifically, a retrovirus vector, a lentivirus vector, etc. can be mentioned.
 例えば、上記核酸を、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、エンドソームによる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。 For example, a viral vector can be prepared by preparing a complex by combining the above nucleic acid with a polylysine-conjugated antibody specific for an adenovirus hexon protein and binding the resulting complex to an adenovirus vector. The virus vector stably reaches the target cell, is taken up into the cell by endosomes, 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、該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。 In addition, viral vectors based on Sendai virus (-) strand RNA virus have been developed (WO97 / 16538, WO97 / 16539), and Sendai virus incorporating the nucleic acid is prepared using the Sendai virus. be able to.
 該核酸は、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。 The nucleic acid can also be transferred by non-viral nucleic acid transfer method. For example, calcium phosphate coprecipitation method [Virology, 52, 456-467 (1973); Science, 209, 1414-1422 (1980)], microinjection method [Proc. Natl. Acad. Sci. USA, 77, 5399-5403-5 ( 1980); Proc. Natl. Acad. Sci. USA, 77, 7380-7384 (1980); Cell, 27, 223-231 (1981); Nature, 294, 92-94 (1981)], liposome-mediated membrane Fusion-Interventional Transfer [Proc. Natl. Acad. Sci. USA, 84, 13 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 method [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 .Sc i. 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、該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。 Membrane fusion-mediated transfer via liposomes allows the nucleic acid to 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 the lesion.
 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。 Receptor-mediated DNA transfer can be performed, for example, by binding DNA (typically 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. To prevent intracellular destruction of DNA, adenovirus can be co-infected to disrupt endosomal function.
 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.
1.マイクロRNAが生合成される過程を阻害するsiRNA
 マイクロRNAが生合成される過程を阻害する物質として、マイクロRNAの生合成に必須な因子の一つであるヒトDicer1遺伝子のsiRNAに着目し、これが実際にDicer1遺伝子発現を低下するかを検討した。
1. SiRNA that inhibits the process of biosynthesis of microRNA
As a substance that inhibits the process of biosynthesis of microRNA, we focused on siRNA of human Dicer1 gene, which is one of the essential factors for biosynthesis of microRNA, and examined whether this actually reduces Dicer1 gene expression. .
 HeLa細胞を6穴プレートに1穴あたり2x105個になるように播種し、ヒトDicer1遺伝子に対するsiRNAをリポフェクション法、具体的には、Lipofectamine2000(Invitrogen社製)を用いて、終濃度が20 nMとなるよう導入した。ヒトDicer1遺伝子に対するsiRNAの配列は配列番号1を標的配列とするDicer-siRNA-1および配列番号2を標的配列とするDicer-siRNA-2をキアゲン社より購入して用いた。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、siRNAを添加せずに、HeLa細胞にリポフェクションをおこなった細胞をコントロールとした。 HeLa cells are seeded at a density of 2x10 5 per well in a 6-well plate, and siRNA for human Dicer1 gene is lipofection method, specifically, Lipofectamine2000 (Invitrogen) is used to achieve a final concentration of 20 nM. Introduced to be. For the siRNA sequence for human Dicer1 gene, Dicer-siRNA-1 having SEQ ID NO: 1 as a target sequence and Dicer-siRNA-2 having SEQ ID NO: 2 as a target sequence were purchased from Qiagen. Lipofection followed the method described in the instructions attached to the product. Moreover, the cell which carried out lipofection to the HeLa cell, without adding siRNA was set as control.
 siRNAを導入した2日後に、RNeasy(キアゲン社製)を用いてそれぞれの細胞からtotal RNAを抽出し、SuperScriptIII逆転写酵素(Invitrogen社製)を用いてcDNAを合成した。それぞれの実験手順は、製品に添付された説明書に記載された方法に従った。 Two days after the introduction of siRNA, total RNA was extracted from each cell using RNeasy (Qiagen), and cDNA was synthesized using SuperScriptIII reverse transcriptase (Invitrogen). Each experimental procedure followed the method described in the instructions attached to the product.
 上記で合成したcDNAをPCR反応の鋳型に用い、ABI7900HT Fast(アプライドバイオシステムズ社製)を用いたSYBR-Green PCRによりDicer1特異的なPCR増幅を行い、mRNA量の定量を行った。サンプルのmRNA量はコントロール細胞における、Dicer1のmRNA量を1としたときの相対的な割合として評価した。 The cDNA synthesized above was used as a template for the PCR reaction, and Dicer1-specific PCR amplification was performed by SYBR-Green® PCR using ABI7900HT® Fast (Applied Biosystems) to quantify the amount of mRNA. The amount of mRNA in the sample was evaluated as a relative ratio when the amount of Dicer1 mRNA was 1 in the control cells.
 その結果、Dicer-siRNA-1導入細胞でのDicer1のmRNA相対発現量は0.38、Dicer-siRNA-2導入細胞でのDicer1のmRNA相対発現量は0.24であり、Dicer1に対するsiRNA導入によりDicer1遺伝子の発現量が低下したことが確認された。 As a result, the relative expression level of Dicer1 mRNA in Dicer-siRNA-1-introduced cells was 0.38, and the relative expression level of Dicer1 in Dicer-siRNA-2-introduced cells was 0.24, and expression of Dicer1 gene by siRNA introduction into Dicer1 It was confirmed that the amount was reduced.
2.マイクロRNAが生合成される過程を阻害させたヒト肥満細胞における脱顆粒に及ぼす作用(1)
 ヒトDicer1遺伝子に対するsiRNAをヒト肥満細胞株であるLAD2に導入し、脱顆粒に及ぼす影響を調べた。
 LAD2は近年樹立されたヒト肥満細胞株で、ヒト肥満細胞の性質をよく保持していることが知られている[Leuk. Res., 27, 671 (2003); Leuk. Res., 27, 677 (2003)]。National Institute of Allergy and Infectious Diseases, National Institutes of Health (Bethesda, MD 20892-1881, USA)よりLAD2を入手し、100 ng/mLのSCFを含むStem Pro-34培地[Invitrogen社製]で培養した。
2. Effects on degranulation in human mast cells that inhibit the process of biosynthesis of microRNAs (1)
SiRNA for human Dicer1 gene was introduced into LAD2, a human mast cell line, and the effect on degranulation was examined.
LAD2 is a recently established human mast cell line that is well known to retain the properties of human mast cells [Leuk. Res., 27, 671 (2003); Leuk. Res., 27, 677 (2003)]. LAD2 was obtained from National Institute of Allergy and Infectious Diseases, National Institutes of Health (Bethesda, MD 20892-1881, USA), and cultured in Stem Pro-34 medium [manufactured by Invitrogen] containing 100 ng / mL SCF.
 LAD2を6穴プレートに1穴あたり5x105個になるように播種し、ヒトDicer1遺伝子に対するsiRNA(Dicer-siRNA-1)をリポフェクション法、具体的には、Gene Silencer(Genlantis社製)を用いて、終濃度が25 nMとなるよう導入した。ヒトDicer1遺伝子に対するsiRNAの配列は配列番号1を標的配列とするDicer-siRNA-1(キアゲン社製)を用いた。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。 LAD2 is seeded in a 6-well plate at 5x10 5 per well, and siRNA (Dicer-siRNA-1) for human Dicer1 gene is lipofection method, specifically using Gene Silencer (Genlantis) The final concentration was 25 nM. As the siRNA sequence for human Dicer1 gene, Dicer-siRNA-1 (manufactured by Qiagen) having SEQ ID NO: 1 as a target sequence was used. Lipofection followed the method described in the instructions attached to the product.
 リポフェクション法によりsiRNAを導入した2日後に、1.0μg/mLのヒトミエローマIgE(コスモバイオ社製)を添加して37℃の5%CO2濃度のインキュベーター中で一晩培養した。翌日、遠心分離により培地を除き、タイロード(Tyrode)緩衝液(126.1 mmol/L NaCl、4.0 mmol/L KCl、1.0 mmol/L CaCl2、0.6 mmol/L MgCl2、0.6 mmol/L KH2PO4、10 mM HEPES、5.6 mmol/L D-グルコース、0.1%ウシ血清アルブミン、pH7.4)で洗浄した後、タイロード緩衝液を1.5 mL添加して細胞を懸濁し、96穴プレートに1穴あたり100μLずつ分注した。次いで、終濃度10μg/mLとなるようウサギ抗ヒトIgE抗体(ダコ社製)を加え、37℃の5%CO2濃度のインキュベーター中で20分間インキュベートし、脱顆粒を誘導した。遠心分離により上清を回収し、上清中のβ-ヘキソサミニダーゼ活性を測定することにより、脱顆粒の程度を測定した。β-ヘキソサミニダーゼ活性は、回収した上清50μLに、40 mmol/Lクエン酸緩衝液(pH 4.5)に溶解した4 mmol/L p-ニトロフェニルN-アセチル-β-グルコサミニド(シグマ社製)を50μLを加え、37℃で1時間インキュベート後、0.2 mol/Lグリシン(pH 10.7)を100μL加えたサンプルの405 nmにおける吸光度をプレートリーダー1420 ARVOsx(パーキンエルマー社製)を用いて測定することにより評価した。また、ウサギ抗ヒトIgE抗体の代わりに最終濃度1%のトリトンX-100を添加して同様の実験を行うことにより、LAD2中の全β-ヘキソサミニダーゼ活性を測定した。脱顆粒の割合を、全β-ヘキソサミニダーゼ活性に対する上清中のβ-ヘキソサミニダーゼ活性の割合(%)で算出し、それぞれについて陰性対照区(Gene Silencerのみ)の脱顆粒の割合を1.0とした時の脱顆粒相対活性を計算した。 Two days after the introduction of siRNA by the lipofection method, 1.0 μg / mL human myeloma IgE (manufactured by Cosmo Bio) was added and cultured overnight in an incubator at 37 ° C. with 5% CO 2 concentration. The next day, the medium was removed by centrifugation, and Tyrode buffer (126.1 mmol / L NaCl, 4.0 mmol / L KCl, 1.0 mmol / L CaCl 2 , 0.6 mmol / L MgCl 2 , 0.6 mmol / L KH 2 PO 4. After washing with 10 mM HEPES, 5.6 mmol / L D-glucose, 0.1% bovine serum albumin, pH 7.4), add 1.5 mL of Tyrode's buffer to suspend the cells. 100 μL was dispensed per unit. Subsequently, a rabbit anti-human IgE antibody (manufactured by Dako) was added to a final concentration of 10 μg / mL and incubated for 20 minutes in an incubator at 37 ° C. with 5% CO 2 concentration to induce degranulation. The supernatant was collected by centrifugation, and the degree of degranulation was measured by measuring β-hexosaminidase activity in the supernatant. The β-hexosaminidase activity was determined by using 4 mmol / L p-nitrophenyl N-acetyl-β-glucosaminide (Sigma) dissolved in 40 mmol / L citrate buffer (pH 4.5) in 50 μL of the collected supernatant. ), Incubate at 37 ° C for 1 hour, and then measure the absorbance at 405 nm of the sample to which 100 µL of 0.2 mol / L glycine (pH 10.7) has been added using a plate reader 1420 ARVOsx (Perkin Elmer). It was evaluated by. Further, the total β-hexosaminidase activity in LAD2 was measured by adding Triton X-100 having a final concentration of 1% instead of the rabbit anti-human IgE antibody and conducting the same experiment. The percentage of degranulation was calculated as the percentage of β-hexosaminidase activity in the supernatant with respect to the total β-hexosaminidase activity, and the percentage of degranulation in the negative control (Gene Silencer only) for each. The relative degranulation activity was calculated when 1.0 was 1.0.
 また、該siRNAを導入した7日後にも、終濃度が1.0μg/mLとなるようヒトミエローマIgE(コスモバイオ社製)を添加し、37℃の5%CO2濃度のインキュベーター中で一晩培養し、翌日、ウサギ抗ヒトIgE抗体添加により脱顆粒を誘導して脱顆粒の割合を測定し、それぞれについて陰性対照区(Gene Silencerのみ)の脱顆粒の割合を1.0とした時の脱顆粒相対活性を計算した。 In addition, 7 days after the introduction of the siRNA, human myeloma IgE (manufactured by Cosmo Bio) was added to a final concentration of 1.0 μg / mL and cultured overnight in an incubator at 37 ° C. with 5% CO 2 concentration. The following day, degranulation was induced by adding rabbit anti-human IgE antibody, and the degranulation rate was measured. For each, the degranulation relative activity when the degranulation rate in the negative control group (Gene Silencer only) was 1.0 Was calculated.
 それぞれ該siRNAを導入した2日後および7日後の翌日(つまり3日後と8日後)における脱顆粒相対活性の結果を表1に示した。 Table 1 shows the results of degranulation relative activity on the next day (that is, after 3 days and 8 days) after 2 days and 7 days after the introduction of the siRNA, respectively.
Figure JPOXMLDOC01-appb-T000001
Figure JPOXMLDOC01-appb-T000001
 更に、ヒトDicer1遺伝子に対するsiRNAの普遍性を検討するため、配列番号1を標的配列とするsiRNA(Dicer-siRNA-1)および配列番号2を標的配列とするsiRNA(Dicer-siRNA-2)をLAD2細胞に導入し、6穴プレートを用いた脱顆粒活性測定を実施した。 Furthermore, in order to investigate the universality of siRNA against human Dicer1 gene, siRNA (SEQ ID NO: 1) as a target sequence (Dicer-siRNA-1) and siRNA (SEQ ID NO: 2) as a target sequence (Dicer-siRNA-2) are LAD2 The cells were introduced into cells, and degranulation activity was measured using a 6-well plate.
 LAD2を6穴プレートに1穴あたり5x105個になるように播種し、siRNA体をリポフェクション法、具体的には、Gene Silencer(Genlantis社製)を用いて、終濃度が30 nMとなるよう導入した。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。 LAD2 is seeded in a 6-well plate at 5x10 5 per well, and siRNA is introduced by lipofection method, specifically Gene Silencer (Genlantis) to a final concentration of 30 nM. did. Lipofection followed the method described in the instructions attached to the product.
 リポフェクション法によりsiRNAを導入した6日後に、1.0μg/mLのヒトミエローマIgE(コスモバイオ社製)を添加して37℃の5%CO2濃度のインキュベーター中で一晩培養した。翌日、遠心分離により培地を除き、タイロード(Tyrode)緩衝液で洗浄した後、タイロード緩衝液を1.5 mL添加して細胞を懸濁し、96穴プレートに1穴あたり100μLずつ分注した。次いで、終濃度15μg/mLとなるようウサギ抗ヒトIgE抗体(ダコ社製)を加え、37℃の5%CO2濃度のインキュベーター中で20分間インキュベートし、脱顆粒を誘導した。遠心により上清を回収し、上清中のβ-ヘキソサミニダーゼ活性を測定することにより、脱顆粒の程度を測定した。β-ヘキソサミニダーゼ活性は、回収した上清50μLに、40 mmol/Lクエン酸緩衝液(pH 4.5)に溶解した4 mmol/L p-ニトロフェニルN-アセチル-β-グルコサミニド(シグマ社製)を50μLを加え、37℃で1時間インキュベート後、0.2 mol/Lグリシン(pH 10.7)を100μL加えたサンプルの405 nmにおける吸光度をプレートリーダー1420 ARVOsx(パーキンエルマー社製)を用いて測定することにより評価した。また、ウサギ抗ヒトIgE抗体の代わりに最終濃度1%のトリトンX-100を添加して同様の実験を行うことにより、LAD2中の全β-ヘキソサミニダーゼ活性を測定した。脱顆粒の割合を、全β-ヘキソサミニダーゼ活性に対する上清中のβ-ヘキソサミニダーゼ活性の割合(%)で算出し、それぞれについて陰性対照区(Gene Silencerのみ)の脱顆粒の割合を1.0とした時の脱顆粒相対活性を計算した。 Six days after introduction of siRNA by the lipofection method, 1.0 μg / mL human myeloma IgE (manufactured by Cosmo Bio) was added and cultured overnight in an incubator at 37 ° C. with 5% CO 2 concentration. The next day, the medium was removed by centrifugation, and after washing with Tyrode buffer, 1.5 mL of Tyrode buffer was added to suspend the cells, and 100 μL per well was dispensed into a 96-well plate. Subsequently, a rabbit anti-human IgE antibody (manufactured by Dako) was added to a final concentration of 15 μg / mL and incubated for 20 minutes in an incubator at 37 ° C. with 5% CO 2 concentration to induce degranulation. The supernatant was collected by centrifugation, and the degree of degranulation was measured by measuring β-hexosaminidase activity in the supernatant. The β-hexosaminidase activity was determined by using 4 mmol / L p-nitrophenyl N-acetyl-β-glucosaminide (Sigma) dissolved in 40 mmol / L citrate buffer (pH 4.5) in 50 μL of the collected supernatant. ), Incubate at 37 ° C for 1 hour, and then measure the absorbance at 405 nm of the sample to which 100 µL of 0.2 mol / L glycine (pH 10.7) has been added using a plate reader 1420 ARVOsx (Perkin Elmer). It was evaluated by. Further, the total β-hexosaminidase activity in LAD2 was measured by adding Triton X-100 having a final concentration of 1% instead of the rabbit anti-human IgE antibody and conducting the same experiment. The percentage of degranulation was calculated as the percentage of β-hexosaminidase activity in the supernatant with respect to the total β-hexosaminidase activity, and the percentage of degranulation in the negative control (Gene Silencer only) for each. The relative degranulation activity was calculated when 1.0 was 1.0.
 表2に、それぞれのsiRNAを導入した脱顆粒相対活性の結果を示した。 Table 2 shows the results of degranulation relative activity in which each siRNA was introduced.
Figure JPOXMLDOC01-appb-T000002
Figure JPOXMLDOC01-appb-T000002
 これらの実験結果から、Dicer1遺伝子に対するsiRNA導入により脱顆粒の割合が減少することが認められた。 From these experimental results, it was confirmed that the rate of degranulation was reduced by the introduction of siRNA for Dicer1 gene.
3.マイクロRNAが生合成される過程を阻害させたヒト肥満細胞における脱顆粒に及ぼす作用(2)
 ヒトDicer1遺伝子に対するsiRNAを導入したLAD2を抗IgE抗体以外で刺激して、脱顆粒に及ぼす影響を調べた。
 LAD2を6穴プレートに1穴あたり5x105個になるように播種し、ヒトDicer1遺伝子に対するsiRNA (Dicer-siRNA-1)をリポフェクション法、具体的には、Gene Silencer(Genlantis社製)を用いて、終濃度が30 nMとなるよう導入した。ヒトDicer1遺伝子に対するsiRNAの配列は配列番号1を標的配列とするDicer-siRNA-1(キアゲン社製)を用いた。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。
3. Effects on degranulation in human mast cells that inhibit the process of biosynthesis of microRNAs (2)
LAD2 introduced with siRNA against human Dicer1 gene was stimulated with other than anti-IgE antibody, and the influence on degranulation was examined.
LAD2 is seeded in a 6-well plate at 5x10 5 per well, and siRNA (Dicer-siRNA-1) for human Dicer1 gene is lipofection method, specifically using Gene Silencer (Genlantis) The final concentration was 30 nM. As the siRNA sequence for human Dicer1 gene, Dicer-siRNA-1 (manufactured by Qiagen) having SEQ ID NO: 1 as a target sequence was used. Lipofection followed the method described in the instructions attached to the product.
 リポフェクション法によりsiRNAを導入した3日後に、遠心分離により培地を除き、タイロード(Tyrode)緩衝液で洗浄した後、タイロード緩衝液を1.5 mL添加して細胞を懸濁し、96穴プレートに1穴あたり100μLずつ分注した。次いで、終濃度0.5 μg/mLとなるようコンパウンド48/80(Sigma-Aldrich社製)を加え、37℃の5%CO2濃度のインキュベーター中で20分間インキュベートし、脱顆粒を誘導した。遠心分離により上清を回収し、上清中のβ-ヘキソサミニダーゼ活性を測定することにより、脱顆粒の程度を測定した。β-ヘキソサミニダーゼ活性は、回収した上清50μLに、40 mmol/Lクエン酸緩衝液(pH 4.5)に溶解した4 mmol/L p-ニトロフェニルN-アセチル-β-グルコサミニド(シグマ社製)を50μLを加え、37℃で1時間インキュベート後、0.2 mol/Lグリシン(pH 10.7)を100μL加えたサンプルの405 nmにおける吸光度をプレートリーダー1420 ARVOsx(パーキンエルマー社製)を用いて測定することにより評価した。また、コンパウンド48/80の代わりに最終濃度1%のトリトンX-100を添加して同様の実験を行うことにより、LAD2中の全β-ヘキソサミニダーゼ活性を測定した。脱顆粒の割合を、全β-ヘキソサミニダーゼ活性に対する上清中のβ-ヘキソサミニダーゼ活性の割合(%)で算出し、それぞれについて陰性対照区(Gene Silencerのみ)の脱顆粒の割合を1.0とした時の脱顆粒相対活性を計算した。 Three days after introduction of siRNA by the lipofection method, the medium is removed by centrifugation, washed with Tyrode buffer, 1.5 mL of Tyrode buffer is added, the cells are suspended, and 1 is added to a 96-well plate. Dispense 100 μL per well. Subsequently, compound 48/80 (manufactured by Sigma-Aldrich) was added to a final concentration of 0.5 μg / mL, and incubated for 20 minutes in an incubator at 37 ° C. with 5% CO 2 concentration to induce degranulation. The supernatant was collected by centrifugation, and the degree of degranulation was measured by measuring β-hexosaminidase activity in the supernatant. The β-hexosaminidase activity was determined by using 4 mmol / L p-nitrophenyl N-acetyl-β-glucosaminide (Sigma) dissolved in 40 mmol / L citrate buffer (pH 4.5) in 50 μL of the collected supernatant. ), Incubate at 37 ° C for 1 hour, and then measure the absorbance at 405 nm of the sample to which 100 µL of 0.2 mol / L glycine (pH 10.7) has been added using a plate reader 1420 ARVOsx (Perkin Elmer). It was evaluated by. Further, the total β-hexosaminidase activity in LAD2 was measured by adding Triton X-100 having a final concentration of 1% instead of Compound 48/80 and conducting the same experiment. The percentage of degranulation was calculated as the percentage of β-hexosaminidase activity in the supernatant with respect to the total β-hexosaminidase activity, and the percentage of degranulation in the negative control (Gene Silencer only) for each. The relative degranulation activity was calculated when 1.0 was 1.0.
 また、該siRNAを導入した7日後にも、コンパウンド48/80添加により脱顆粒を誘導して脱顆粒の割合を測定し、それぞれについて陰性対照区(Gene Silencerのみ)の脱顆粒の割合を1.0とした時の脱顆粒相対活性を計算した。 Also, 7 days after the introduction of the siRNA, degranulation was measured by inducing degranulation by adding compound 48/80, and the degranulation ratio in the negative control group (Gene-Silencer only) was 1.0 for each. The relative degranulation activity was calculated.
 それぞれ該siRNAを導入した3日後および7日後における脱顆粒相対活性の結果を表3に示した。 Table 3 shows the results of degranulation relative activity 3 days and 7 days after the introduction of the siRNA, respectively.
Figure JPOXMLDOC01-appb-T000003
Figure JPOXMLDOC01-appb-T000003
 この実験結果から、コンパウンド48/80刺激による脱顆粒の割合が、Dicer1遺伝子に対するsiRNA導入により減少することが認められた。 From this experimental result, it was confirmed that the ratio of degranulation by compound 48/80 stimulation was reduced by siRNA introduction to Dicer1 gene.

Claims (18)

  1.  マイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質を有効成分として含有する、肥満細胞の脱顆粒抑制剤。 A mast cell degranulation inhibitor containing as an active ingredient a substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor.
  2.  マイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質がDicer1遺伝子の発現を抑制する物質である、請求項1に記載の脱顆粒抑制剤。 The degranulation inhibitor according to claim 1, wherein the substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor is a substance that suppresses the expression of Dicer1 gene.
  3.  Dicer1遺伝子の発現を抑制する物質が核酸である、請求項2に記載の脱顆粒抑制剤。 The degranulation inhibitor according to claim 2, wherein the substance that suppresses the expression of Dicer1 gene is a nucleic acid.
  4.  核酸がsiRNAである、請求項3に記載の脱顆粒抑制剤。 4. The degranulation inhibitor according to claim 3, wherein the nucleic acid is siRNA.
  5.  Dicer1遺伝子の発現を抑制するsiRNAを発現するベクターを有効成分として含有する、肥満細胞の脱顆粒抑制剤。 A mast cell degranulation inhibitor containing, as an active ingredient, a vector that expresses siRNA that suppresses the expression of Dicer1 gene.
  6.  siRNAが配列番号1~5のいずれかで表される塩基配列を標的配列とするsiRNAである、請求項4または5に記載の脱顆粒抑制剤。 The degranulation inhibitor according to claim 4 or 5, wherein the siRNA is a siRNA having a base sequence represented by any one of SEQ ID NOs: 1 to 5 as a target sequence.
  7.  マイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質を用いることを特徴とする、肥満細胞の脱顆粒抑制方法。 A method for suppressing degranulation of mast cells, comprising using a substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor.
  8.  マイクロRNA前駆体からマイクロRNAが生合成される過程を阻害する物質が、Dicer1遺伝子の発現を抑制する物質である、請求項7に記載の脱顆粒抑制方法。 The method for inhibiting degranulation according to claim 7, wherein the substance that inhibits the process of biosynthesis of microRNA from a microRNA precursor is a substance that suppresses the expression of Dicer1 gene.
  9.  Dicer1遺伝子の発現を抑制する物質が核酸である、請求項8に記載の脱顆粒抑制方法。 The method for inhibiting degranulation according to claim 8, wherein the substance that suppresses the expression of Dicer1 gene is a nucleic acid.
  10.  核酸がsiRNAである、請求項9に記載の脱顆粒抑制方法。 The method for suppressing degranulation according to claim 9, wherein the nucleic acid is siRNA.
  11.  Dicer1遺伝子の発現を抑制するsiRNAを発現するベクターを用いることを特徴とする、肥満細胞の脱顆粒抑制方法。 A method for inhibiting degranulation of mast cells, characterized by using a vector that expresses siRNA that suppresses the expression of Dicer1 gene.
  12.  siRNAが配列番号1~5のいずれかで表される塩基配列を標的配列とするsiRNAである、請求項10または11に記載の脱顆粒抑制方法。 The method for suppressing degranulation according to claim 10 or 11, wherein the siRNA is a siRNA having a base sequence represented by any of SEQ ID NOs: 1 to 5 as a target sequence.
  13.  請求項1~6のいずれかに記載の脱顆粒抑制剤を有効成分として含有する、肥満細胞の異常に起因する疾患の治療薬。 A therapeutic agent for a disease caused by an abnormality in mast cells, comprising the degranulation inhibitor according to any one of claims 1 to 6 as an active ingredient.
  14.  肥満細胞の異常に起因する疾患が、アトピー性皮膚炎、喘息、慢性閉塞性肺疾患およびアレルギー疾患からなる群から選ばれる疾患である、請求項13に記載の治療薬。 The therapeutic agent according to claim 13, wherein the disease caused by an abnormality in mast cells is a disease selected from the group consisting of atopic dermatitis, asthma, chronic obstructive pulmonary disease, and allergic disease.
  15.  請求項1~6のいずれかに記載の脱顆粒抑制剤の有効量を、必要とする対象に投与することを含む、肥満細胞の異常に起因する疾患の治療方法。 A method for treating a disease caused by abnormal mast cells, comprising administering an effective amount of the degranulation inhibitor according to any one of claims 1 to 6 to a subject in need thereof.
  16.  肥満細胞の異常に起因する疾患が、アトピー性皮膚炎、喘息、慢性閉塞性肺疾患およびアレルギー疾患からなる群から選ばれる疾患である、請求項15に記載の治療方法。 The treatment method according to claim 15, wherein the disease caused by abnormality of mast cells is a disease selected from the group consisting of atopic dermatitis, asthma, chronic obstructive pulmonary disease, and allergic disease.
  17.  肥満細胞の異常に起因する疾患の治療薬の製造のための、請求項1~6のいずれかに記載の脱顆粒抑制剤の使用。 Use of the degranulation inhibitor according to any one of claims 1 to 6 for the manufacture of a therapeutic agent for a disease caused by an abnormality of mast cells.
  18.  肥満細胞の異常に起因する疾患が、アトピー性皮膚炎、喘息、慢性閉塞性肺疾患およびアレルギー疾患からなる群から選ばれる疾患である、請求項17に記載の使用。 The use according to claim 17, wherein the disease caused by abnormality of mast cells is a disease selected from the group consisting of atopic dermatitis, asthma, chronic obstructive pulmonary disease and allergic disease.
PCT/JP2009/060220 2008-07-07 2009-06-04 Inhibitor of degranulation of mast cell WO2010004816A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2008-177109 2008-07-07
JP2008177109A JP2011190176A (en) 2008-07-07 2008-07-07 Degranulation inhibitor for mast cell

Publications (1)

Publication Number Publication Date
WO2010004816A1 true WO2010004816A1 (en) 2010-01-14

Family

ID=41506938

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2009/060220 WO2010004816A1 (en) 2008-07-07 2009-06-04 Inhibitor of degranulation of mast cell

Country Status (2)

Country Link
JP (1) JP2011190176A (en)
WO (1) WO2010004816A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2006320327A (en) * 2002-07-26 2006-11-30 Chiron Corp Modified small interfering rna molecule and method of using the same
JP2007000002A (en) * 2003-07-16 2007-01-11 Genecare Research Institute Co Ltd Method for enhancing sensitivity to alkylating drug
WO2008029790A1 (en) * 2006-09-04 2008-03-13 Kyowa Hakko Kirin Co., Ltd. Novel nucleic acid

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2006320327A (en) * 2002-07-26 2006-11-30 Chiron Corp Modified small interfering rna molecule and method of using the same
JP2007000002A (en) * 2003-07-16 2007-01-11 Genecare Research Institute Co Ltd Method for enhancing sensitivity to alkylating drug
WO2008029790A1 (en) * 2006-09-04 2008-03-13 Kyowa Hakko Kirin Co., Ltd. Novel nucleic acid

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
ABE,T. ET AL.: "Eosinophil chemotactic factor released from murine bone marrow derived cultured mast cells", ARERUGI, vol. 39, no. 2, 1990, pages 69 - 74 *
BILEN,J. ET AL.: "MicroRNA pathways modulate polyglutamine-induced neurodegeneration", MOL CELL, vol. 24, no. 1, 2006, pages 157 - 163 *
CHENDRIMADA,T.P. ET AL.: "TRBP recruits the Dicer complex to Ago2 for microRNA processing and gene silencing", NATURE, vol. 436, no. 7051, 2005, pages 740 - 744 *
KOK,K.H. ET AL.: "Human TRBP and PACT directly interact with each other and associate with dicer to facilitate the production of small interfering RNA", J BIOL CHEM, vol. 282, no. 24, 2007, pages 17649 - 17657 *
KUEHN,H.S. ET AL.: "G protein-coupled receptors and the modification of FcepsilonRI-mediated mast cell activation", IMMUNOL LETT, vol. 113, no. 2, 2007, pages 59 - 69 *
MATSUDA,S. ET AL.: "Molecular cloning and characterization of a novel human gene (HERNA) which encodes a putative RNA-helicase", BIOCHIM BIOPHYS ACTA, vol. 1490, no. 1-2, 2000, pages 163 - 169 *
TEXTOR,B. ET AL.: "JunB is required for IgE- mediated degranulation and cytokine release of mast cells", J IMMUNOL, vol. 179, no. 10, 2007, pages 6873 - 6880 *

Also Published As

Publication number Publication date
JP2011190176A (en) 2011-09-29

Similar Documents

Publication Publication Date Title
EP3277815B1 (en) Oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders
CA2583375C (en) Regulation of oncogenes by micrornas
EP3190186B1 (en) Expression of mirnas in placental tissue
JP6414886B2 (en) Long non-coding RNA for anti-cancer therapy
WO2009148137A1 (en) Nucleic acid capable of controlling degranulation of mast cell
US20090220589A1 (en) Treatment, diagnostic, and method for discovering antagonist using sparc specific mirnas
JPWO2009044899A1 (en) Nucleic acids that control cell growth
EP2077326A1 (en) Novel nucleic acid
CN107267625B (en) Application of lncRNA as biomarker in liver cancer diagnosis and treatment
JP2011519951A (en) Enhancement of drug treatment by miRNA
JPWO2008084319A1 (en) New nucleic acid
WO2011019074A1 (en) Nucleic acid that controls fibrosis of cells or organs
US20100292299A1 (en) Nucleotide Motifs Providing Localization Elements and Methods of Use
CN112933112B (en) Application of graphene oxide or regulating and controlling molecule thereof in preparation of medicine for promoting diabetic wound repair
WO2011111715A1 (en) Nucleic acid capable of regulating cell cycle
EP2904102B1 (en) Modulation of rna activity and vascular permeability
WO2010004816A1 (en) Inhibitor of degranulation of mast cell
JP2006180710A (en) Rna inhibiting expression of klf5 gene
WO2011074652A1 (en) Nucleic acid capable of inhibiting expression of hif-2α
WO2011130464A1 (en) Translational regulation of sparc by the micrornas mir29a, b and c
WO2011010737A1 (en) Guide nucleic acid for cleavage of micro-rna
CN115666590A (en) Oral squamous carcinoma related biomarkers and methods of diagnosis and treatment
JP5263729B2 (en) Preventive and therapeutic agent for feline infectious peritonitis virus (FIPV)
JP2021075473A (en) Agents for preventing and/or treating mesothelioma
WO2013113252A1 (en) Method for regulating expression level of cyp gene and use thereof

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: 09794263

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

NENP Non-entry into the national phase

Ref country code: JP

122 Ep: pct application non-entry in european phase

Ref document number: 09794263

Country of ref document: EP

Kind code of ref document: A1