WO2012153854A1 - Cytokine-chemokine modulator - Google Patents
Cytokine-chemokine modulator Download PDFInfo
- Publication number
- WO2012153854A1 WO2012153854A1 PCT/JP2012/062230 JP2012062230W WO2012153854A1 WO 2012153854 A1 WO2012153854 A1 WO 2012153854A1 JP 2012062230 W JP2012062230 W JP 2012062230W WO 2012153854 A1 WO2012153854 A1 WO 2012153854A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- domain
- nucleotide
- represented
- sequence
- mrna
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Ceased
Links
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P29/00—Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/113—Non-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
- C12N15/1136—Non-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 growth factors, growth regulators, cytokines, lymphokines or hormones
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/10—Type of nucleic acid
- C12N2310/11—Antisense
- C12N2310/113—Antisense targeting other non-coding nucleic acids, e.g. antagomirs
Definitions
- the present invention relates to a nucleic acid (antisense nucleotide) that is complementary to a natural (endogenous) antisense transcript that is complementary to the mRNA of a cytokine chemokine involved in inflammation or infection, and changes the stability of the mRNA, and Utilization of its cytokine / chemokine mRNA stability regulating action, and a natural (endogenous) antisense transcript that is complementary to the cytokine / chemokine mRNA involved in inflammation and infection and changes the stability of the mRNA
- the present invention relates to a complementary nucleic acid (sense nucleotide) and use of its cytokine / chemokine mRNA stability regulating action.
- Natural (endogenous) antisense transcript (N atural A ntisense T ranscript; hereinafter also referred to as "NAT") and, inherent in the organism, a RNA having a nucleotide sequence complementary to mRNA, specifically Specifically, it is RNA synthesized using a DNA strand encoding a sense gene (that is, a non-template strand of mRNA) as a template.
- Sense-antisense RNA has the ability to form double strands. For example, double-stranded RNA is necessary for RNA interference, and is used to control protein translation by small RNAs called microRNAs (miRNAs). It is known that double-stranded RNA is involved.
- Non-patent Documents 1 and 2 Recent comprehensive cDNA analysis revealed that a considerable amount of NAT was transcribed (Non-patent Documents 1 and 2). For example, it has been suggested that about 2,500 pairs (Non-patent Document 3) exist in mice and about 2,600 pairs (Non-patent Document 4) exist in humans. Does not contain a lot of untranslatable NAT.
- Non-Patent Document 5 it has been reported that NAT complementary to endothelial nitric oxide synthase (eNOS) mRNA increases in the presence of a histone deacetylase inhibitor and decreases the amount of eNOS mRNA (non-patent literature). 6). Also in yeast, it is known that NAT of the PHO84 gene suppresses mRNA expression through histone deacetylation (Non-patent Document 7).
- eNOS endothelial nitric oxide synthase
- the present inventors have a NAT complementary to the 3′-untranslated region (UTR) of inducible nitric oxide synthase (iNOS) mRNA, and that this antisense RNA hybridizes with iNOS RNA mRNA.
- the mRNA is stabilized and the production amount of iNOS and the synthesis amount of NO increase, and the sense oligodeoxynucleotide (ODN) complementary to the NAT inhibits the binding of NAT to the iNOS mRNA. It was reported that iNOS production and NO synthesis by iNOS can be suppressed (Patent Documents 1 and 2, Non-Patent Document 8).
- Nishizawa and its collaborators have a complementary NAT in the region that forms a secondary structure with two stem-loops, which is important for the nuclear export of interferon (IFN) - ⁇ mRNA.
- Antisense RNA contributes to the stabilization of IFN- ⁇ mRNA, and NAT knockdown by sense ODN leads to mRNA instability, whereas overexpression of antisense RNA significantly stabilizes the mRNA (Patent Document 3).
- the object of the present invention was to search for NAT capable of regulating the expression of cytokines and chemokine genes related to inflammation and infection, and to use a sense oligonucleotide complementary to the NAT and an antisense oligonucleotide homologous to the NAT, It is to provide a novel means for preventing and treating diseases such as inflammatory diseases and viral infections.
- the present inventors selected 21 genes (including cytokines and chemokine genes) induced by IL-1 ⁇ and attempted to detect NAT complementary to the 3 ′ UTR of mRNA. As a result of strand-specific RT-PCR, it was found that NAT was transcribed from 16 out of 21 genes (76%). Next, we have 7 genes (CCL2, CCL20, CX3CL1, IL-23 p19 subunit, CD69, NF- ⁇ B p65 subunit, TNF- ⁇ ) in which both mRNA and NAT are abundantly transcribed.
- RNA sequences containing loop portions within the conserved region of the predicted secondary structure have altered (increased or decreased) the expression level of the target mRNA.
- a sense ODN designed outside the conserved region of the secondary structure did not affect the expression level of the target mRNA.
- the present inventors have a NAT containing a region corresponding to 3′UTR in many cytokine-chemokine genes induced by IL-1 ⁇ , and the expression of the gene is regulated by NAT. Furthermore, using a sense oligonucleotide having a sequence complementary to the region containing the loop portion of the secondary structure that can be taken by the NAT (hence, homologous to mRNA), expression control (up-regulation and down-regulation) of the gene is performed. It was concluded that (regulation) is possible, and the present invention was completed.
- the present invention is as follows. [1] Induced by IL-1 ⁇ , CCL2, CCL20, CX3CL1, IL-23A, CD69, NF- ⁇ B p65, TNF- ⁇ , Fam89a, Grk5, phosopholipid scramblase 1, Runx1, semaphorin 4A, Steap4, lymphphotoxin ⁇ Psmb10 Alternatively, an antisense nucleotide comprising a sequence complementary to the 3′UTR of the TLR2 gene and capable of regulating the expression of the gene.
- a sense oligonucleotide comprising a sequence complementary to an endogenous antisense transcript comprising a sequence complementary to the 3 ′ UTR of the TLR2 gene and capable of regulating expression of the gene.
- the sense oligonucleotide according to [6] above which is any of the following (a) to (g): (a) In the 3′UTR sequence of CCL2 mRNA represented by SEQ ID NO: 1, domain 1 represented by nucleotide numbers 25-51 or domain 2 represented by nucleotide numbers 103-175, or those of orthologs in other mammals Sense oligonucleotide capable of regulating the expression of the CCL2 gene, comprising a nucleotide sequence having 90% or more identity with a sequence comprising at least part of one or more loop structures in the corresponding domain (b) In the 3'UTR sequence of CCL20 mRNA represented by SEQ ID NO: 2, domain 1 represented by nucleotide numbers 65-107, domain 2 represented by nucleotide numbers 124-155, or domain 3 represented by nucleotide numbers 214-282 Or expression of the CCL20 gene comprising a nucleotide sequence having at least 90% identity with a sequence comprising at least
- the antisense nucleotide of the present invention can regulate the expression of the gene by interacting with the 3 'UTR of the mRNA in the same manner as NAT for mRNA of various genes induced by IL-1 ⁇ .
- the sense oligonucleotide of the present invention can regulate the expression of a target gene to a desired level by interacting with NAT to regulate its gene expression regulation action positively or negatively.
- FIG. 5 shows the position of a conserved region (indicated by domain “+”) containing one or more loop structures of 3′UTR of CCL2 mRNA and designed sense ODN (indicated by underline). It is a figure which shows the predicted secondary structure of 3'UTR of CCL20 * mRNA.
- FIG. 4 shows the position of a conserved region (indicated by domain “+”) containing one or more loop structures of 3′UTR of CCL20 mRNA and the designed sense ODN (indicated by underline). It is a figure which shows the predicted secondary structure of 3'UTR of CX3CL1 mRNA.
- FIG. 5 shows the position of a conserved region (indicated by domain “+”) containing one or more loop structures of 3′UTR of CCL2 mRNA and designed sense ODN (indicated by underline). It is a figure which shows the predicted secondary structure of 3'UTR of CX3CL1 mRNA.
- FIG. 3 shows the position of a conserved region (indicated by domain “+”) containing one or more loop structures of 3′UTR of CX3CL13 mRNA and the designed sense ODN (indicated by underline).
- FIG. 4 shows the predicted secondary structure of 3′UTR of IL-23A mRNA.
- FIG. 3 shows the position of a conserved region (indicated by domain “+”) containing one or more loop structures of 3′UTR of IL-23A mRNA and the designed sense ODN (indicated by underline).
- FIG. 3 shows the predicted secondary structure of 3 ′ UTR of CD69 mRNA.
- FIG. 3 shows the position of a conserved region (indicated by domain “+”) containing one or more loop structures of 3′UTR of CD69 mRNA and the designed sense ODN (indicated by underline). It is a figure which shows the predicted secondary structure of 3'UTR of NF- ⁇ B p65 mRNA.
- FIG. 3 shows the position of a conserved region (indicated by domain “+”) containing one or more loop structures of 3 ′ UTR of NF- ⁇ B p65 mRNA and designed sense ODN (indicated by underline). It is a figure which shows the predicted secondary structure of 3'UTR of TNF- (alpha) mRNA.
- 3 shows the position of a conserved region (indicated by domain “+”) containing one or more loop structures of 3′UTR of TNF- ⁇ mRNA and the designed sense ODN (indicated by underline). It is a figure which shows the expression fluctuation
- the present invention provides antisense nucleotides of these genes, which have an effect of regulating the expression of genes related to various inflammations / infections including cytokines and chemokines induced by IL-1 ⁇ .
- the target gene (mRNA) in the present invention is an endogenous antisense transcript containing a sequence complementary to its 3 ′ untranslated region (3′UTR) in the gene group induced by IL-1 ⁇ .
- chemokine (CC motif) ligand 2 CCL2
- chemokine (CC motif) ligand 20 CCL20
- chemokine (C-X3-C motif) ligand 1 CX3CL1
- IL-23A cluster of differentiation 69
- CD69 nuclear factor- ⁇ B p65 subunit
- NF- ⁇ B p65 nuclear factor- ⁇ B p65
- TNF- ⁇ tumor necrosis factor- ⁇
- family with sequence similarity 89 member A (Fam89a); G protein-coupled receptor kinase 5 (Grk5); phosopholipid scramblase 1; runt-related transcription factor 1 (Runx1); sema domain, immunoglobulin domain, transmembrane domain and short cytoplasmic domain 4A (semaphorin 4A); six-transmembrane epithelial antigen of prostate 4 (Stepa); chemokine (CC motif) ligand 2 (CCL2); chemokine (CC
- the antisense nucleotide of the present invention may be any nucleotide as long as it contains a sequence complementary to the 3 'UTR of the target mRNA and can regulate the expression (protein production) of the target gene.
- the “complementary” sequence is not only a sequence that is completely complementary to mRNA, but also 1 to a number (as long as it can hybridize with and interact with mRNA under physiological conditions of cells). 2, 3, 4 or 5) may contain base mismatches.
- the sequence complementary to the 3′UTR of the target mRNA is a stringent condition such as those described in Current Protocols in Molecular Biology, John Wiley & Sons, 6.3.1-6.3.6, 1999 For example, hybridization at 6 ⁇ SSC (sodium chloride / sodium citrate) / 45 ° C., followed by one or more washings at 0.2 ⁇ SSC / 0.1% SDS / 50-65 ° C.) It is a sequence that can hybridize with.
- SSC sodium chloride / sodium citrate
- the antisense nucleotides of the present invention may contain 3′UTR of the target mRNA. It is preferred to include a sequence complementary to a portion that is not thermodynamically stable therein.
- the portion that is not thermodynamically stable includes a region that is in a single-stranded state when the mRNA has a secondary structure (for example, a region corresponding to the loop portion of the stem-loop structure).
- the secondary structure of the 3'UTR of the target mRNA is represented by mfold (see GCG Software; Proc. Natl. Acad. Sci.
- nucleotide sequence information based on the nucleotide sequence information of the region. Can be predicted using an existing RNA secondary structure prediction program. Any of these nucleotide sequence information is readily available.
- CCL2, CCL20, CX3CL1, IL-23A, CD69, NF- ⁇ B p65 and TNF- ⁇ mRNA 3′UTR sequences are shown in SEQ ID NOs: 1-7, respectively. Show.
- the antisense nucleotide against CCL2 of the present invention is the domain 1 and / or nucleotide number 103-175 represented by nucleotide number 25-51 in the rat CCL2 mRNA 3'UTR sequence represented by SEQ ID NO: 1.
- a sequence containing at least a part of one or more loop structures (for example, 3 bases or more) in the domain corresponding to each domain in the rat CCL2 Containing a complementary nucleotide sequence is the domain 1 and / or nucleotide number 103-175 represented by nucleotide number 25-51 in the rat CCL2 mRNA 3'UTR sequence represented by SEQ ID NO: 1.
- a sequence containing at least a part of one or more loop structures for example, 3 bases or more
- the antisense nucleotide against CCL20 of the present invention in the rat CCL20 ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ mRNA 3'UTR sequence shown in SEQ ID NO: 2, is in domain 1 and / or nucleotide numbers 124-155 shown in nucleotide numbers 65-107 In the domain 2 shown and / or in the domain 3 shown by nucleotide numbers 214-282, in the CCL20 ortholog in other mammals, one or more loop structures in the domain corresponding to each domain in the rat CCL20 A nucleotide sequence complementary to a sequence containing at least a part (for example, 3 bases or more) is included.
- the antisense nucleotide against CX3CL1 of the present invention is the domain 1 and / or nucleotide number 1305-1369 represented by nucleotide number 1103-1202 in the 3′UTR sequence of rat CX3CL1 mRNA represented by SEQ ID NO: 3.
- domain 6 indicated by -1810 in the CX3CL1 ortholog in other mammals, the domain corresponding to each domain in the rat CX3CL1 is complementary to a sequence containing at least a part of one or more loop structures. Contains nucleotide sequence.
- the antisense nucleotide against IL-23A of the present invention is the domain 1 and / or nucleotide represented by nucleotide number 37-55 in the 3′UTR sequence of rat IL-23A mRNA represented by SEQ ID NO: 4
- the domain 2 shown by the number 193-303 and / or the domain 3 shown by the nucleotide number 380-448 and / or the domain 4 shown by the nucleotide number 555-600 in the IL-23A ortholog in other mammals
- the antisense nucleotide against CD69 of the present invention is the domain 1 and / or nucleotide number 120-186 represented by nucleotide number 15-32 in the 3′UTR sequence of rat CD69 mRNA represented by SEQ ID NO: 5.
- domain 6 shown by -786 in the CD69 ortholog in other mammals, the domain corresponding to each domain in rat CD69 is complementary to a sequence containing at least a part of one or more loop structures. Contains nucleotide sequence.
- the antisense nucleotide for NF- ⁇ B p65 of the present invention is the domain 1 and / or the nucleotides 161-301 in the 3′UTR sequence of rat NF- ⁇ B p65 mRNA shown in SEQ ID NO: 6. Or in domain 2 indicated by nucleotide number 343-380 and / or domain 3 indicated by nucleotide number 401-412 and / or domain 4 indicated by nucleotide number 473-523, to NF- ⁇ B p65 ortholog in other mammals In that case, it comprises a nucleotide sequence complementary to a sequence containing at least a part of one or more loop structures in the domain corresponding to each domain in the rat NF- ⁇ B p65.
- the antisense nucleotide against TNF- ⁇ of the present invention is the domain 1 and / or nucleotide represented by nucleotide number 304-405 in the 3′UTR sequence of rat TNF- ⁇ mRNA represented by SEQ ID NO: 7
- the nucleotide sequence of the peripheral region of the ARE motif common to multiple species is well conserved among mammalian species. Since the secondary structure is similar, the region can be preferably selected.
- Table 1 shows the number of ARE motifs present in the 3'UTR of the above-mentioned 7 genes of rat, mouse and human and the common ARE motifs between human-rat and among three species.
- the length of the antisense nucleotide of the present invention is not particularly limited, and a nucleotide sequence complementary to a sequence comprising at least a part of one or more loop structures in the domain in the full length of NAT or 3′UTR of the target mRNA. It may contain a partial sequence of NAT, but from the viewpoint of sequence specificity, it contains at least 10 bases, preferably about 12 bases or more, more preferably about 15 bases or more, a portion complementary to the target sequence. . In addition, from the viewpoint of ease of administration when used as a pharmaceutical, those having a base length of 500 bases or less, preferably 300 bases or less, more preferably 150 bases or less are mentioned.
- the antisense nucleotide of the present invention is an oligonucleotide having about 10 to 50 bases
- the nucleotide is a sequence that causes a sequence-nonspecific reaction (for example, 5′-CG-3 ′, 5′-GGGG-3 ', 5'-GGGGG-3' and the like are preferably selected, and are preferably selected from those having no similar complementary strand sequence in RNA other than the target mRNA.
- the absence of a similar complementary strand sequence in other RNAs can be confirmed by performing a homology search against the target mammalian genome sequence using the antisense oligonucleotide candidate sequence as a query. .
- homology search means known nucleic acid homology search software (for example, NCBI BLAST (National Center Biotechnology Information Basic Basic Local Alignment Search Tool) NBLAST and XBLAST programs (version 2.0), FASTA program in GCG software package Etc.) can be used.
- NCBI BLAST National Center Biotechnology Information Basic Basic Local Alignment Search Tool
- XBLAST XBLAST programs (version 2.0)
- FASTA program in GCG software package Etc. can be used as a genomic DNA data set.
- a genomic DNA data set for example, all human genome data provided by Celera can be used.
- the antisense nucleotide of the present invention is used in various forms depending on the method of introduction into cells.
- the antisense nucleotide when it is an oligonucleotide having about 10 to 50 bases, it may be any one of single-stranded DNA, single-stranded RNA, and DNA / RNA chimera, and it is further added with known modifications. There may be.
- the “nucleotide” may include not only purine and pyrimidine bases but also those having other modified heterocyclic bases.
- the nucleotide molecule constituting the antisense nucleotide may be natural DNA or RNA, but various chemicals may be used to improve stability (chemical and / or enzyme) and specific activity (affinity with RNA). Modifications can be included. For example, in order to prevent degradation by nuclease, etc., phosphate residues (phosphates) of each nucleotide constituting the antisense oligonucleotide may be changed to chemically modified phosphate residues such as phosphorothioate (PS), methylphosphonate, phosphorodithionate, etc. Substituents can be substituted.
- PS phosphorothioate
- methylphosphonate methylphosphonate
- phosphorodithionate etc.
- Substituents can be substituted.
- the 2′-position hydroxyl group of the sugar (ribose) of each nucleotide is represented by —OR (R is, for example, CH 3 (2′-O-Me), CH 2 CH 2 OCH 3 (2′-O-MOE), CH 2 CH 2 NHC (NH) NH 2 , CH 2 CONHCH 3 , CH 2 CH 2 CN and the like may be substituted).
- R is, for example, CH 3 (2′-O-Me), CH 2 CH 2 OCH 3 (2′-O-MOE), CH 2 CH 2 NHC (NH) NH 2 , CH 2 CONHCH 3 , CH 2 CH 2 CN and the like may be substituted).
- the base moiety pyrimidine, purine
- BNA LNA
- ENA ENA
- the antisense oligonucleotide of the present invention synthesizes a complementary sequence based on the target mRNA (cDNA) sequence using a commercially available DNA / RNA automatic synthesizer (Applied Biosystems, Beckman, etc.). Can be prepared.
- the antisense oligonucleotide of the present invention is provided in a special form such as a liposome or a microsphere, or a hydrophobic substance such as a polycationic substance such as polylysine or a lipid (eg, phospholipid, cholesterol, etc.) is added thereto. It can be provided in the form.
- the antisense oligonucleotide of the present invention is converted to a peptide having a membrane permeation function (for example, Drosophila-derived Antennapedia homeodomain (AntP), human immunodeficiency virus (HIV) -derived TAT, herpes simplex virus (HSV) -derived Modification with a cell-passing domain such as VP22) can promote the uptake of the oligonucleotide into cells.
- a membrane permeation function for example, Drosophila-derived Antennapedia homeodomain (AntP), human immunodeficiency virus (HIV) -derived TAT, herpes simplex virus (HSV) -derived Modification with a cell-passing domain such as VP22
- the antisense nucleotide of the present invention is a polynucleotide having a longer base length
- introduction of the nucleotide into a cell can be carried out using a gene transfer method known per se.
- double-stranded DNA is preferably used as the nucleotide.
- the antisense nucleotide of the present invention extracts, for example, total RNA from a cell that expresses the target gene NAT (for example, a cell that highly expresses target mRNA and NAT by IL-1 ⁇ stimulation).
- SEQ ID NOs: 1-7 Based on the sequence information of the target mRNA shown in the above, a primer that can amplify an appropriate region of the complementary strand sequence is designed, and RT-PCR is performed (see Examples below).
- the obtained cDNA is inserted into an appropriate expression vector containing a promoter that can function in host animal cells.
- expression vectors include retroviruses, lentiviruses, adenoviruses, adeno-associated viruses, herpes viruses, Sendai virus and other viral vectors, animal cell expression plasmids (eg, pA1-11, pXT1, pRc / CMV, pRc / RSV). , PcDNAI / Neo) or the like.
- Examples of the promoter used in the expression vector include EF1 ⁇ promoter, CAG promoter, SR ⁇ promoter, SV40 promoter, LTR promoter, CMV (cytomegalovirus) promoter, RSV (rous sarcoma virus) promoter, MoMuLV (Molone murine leukemia virus) LTR. HSV-TK (herpes simplex virus thymidine kinase) promoter and the like are used. Of these, EF1 ⁇ promoter, CAG promoter, MoMuLV LTR, CMV promoter, SR ⁇ promoter and the like are preferable.
- the expression vector may optionally contain an enhancer, a poly A addition signal, a selection marker gene, an SV40 replication origin, and the like.
- the selection marker gene include a dihydrofolate reductase gene, a neomycin resistance gene, a puromycin resistance gene, and the like.
- the antisense nucleotide of the present invention can regulate target gene expression (protein production) by interacting with the target mRNA.
- Various genes including cytokine / chemokine gene induced by IL-1 ⁇ have various physiological activities such as inflammation-inducing or anti-inflammatory action, infection defense, regulation of immune response, etc.
- the contained drug can be used as a target gene expression regulator for the prevention and / or treatment of various diseases including inflammatory diseases and infectious diseases.
- antisense nucleotides that can enhance the expression of CCL2 can be used for angiogenesis and wound healing, while CCL2 migrates to monocytes and eosinophils, so that it can suppress the expression of CCL2.
- Nucleotides are useful for the prevention and treatment of chronic inflammatory diseases including arteriosclerosis and allergic inflammation. Since CCL20 exhibits antibacterial activity against a wide range of microorganisms, antisense nucleotides that can enhance the expression of CCL20 are effective in preventing and treating various microbial infections. On the other hand, since CCL20 plays an important role in the initiation and maintenance of adaptive immunity, antisense nucleotides that can suppress the expression of CCL20 are bronchial asthma, multiple sclerosis, rheumatoid arthritis, various dermatitis, inflammatory It is effective for the prevention and treatment of autoimmune diseases such as bowel disease.
- Antisense nucleotides that can enhance CX3CL1 expression can be used for angiogenesis, while antisense nucleotides that can suppress CX3CL1 expression prevent or treat inflammatory diseases including arteriosclerosis and glomerulonephritis It is useful for the prevention and treatment of anti-rejection and rheumatoid arthritis.
- IL-23A is important for the maintenance of cellular immunity, and its overexpression causes multi-organ inflammation. Therefore, antisense nucleotides that can enhance IL-23A expression are effective in the prevention and treatment of infectious diseases.
- antisense nucleotides that can suppress the expression of IL-23A are useful for the prevention and treatment of autoimmune diseases and inflammatory diseases.
- CD69 plays an important role in the differentiation and activation of lymphocytes
- antisense nucleotides that can enhance the expression of CD69 are effective in the prevention and treatment of infectious diseases.
- CD69 is expressed in most inflammatory cells that are expressed locally, and an anti-antibody that can suppress the expression of CD69 due to detection of autoantibodies against CD69 from patients with rheumatoid arthritis and systemic lupus erythematosus (SLE).
- Nucleotides are useful for the prevention and treatment of autoimmune diseases and inflammatory diseases.
- NF- ⁇ B is a transcription factor having an important role in the immune system
- antisense nucleotides that can enhance the expression of NF- ⁇ B p65 are effective in preventing and treating infectious diseases.
- NF- ⁇ B is constantly activated in cancer cells and inhibits apoptosis, and is also involved in pathogenesis such as bone metabolism, bronchial asthma, arthritis, inflammatory bowel disease, sepsis, Antisense nucleotides that can suppress the expression of NF- ⁇ B p65 are useful for the prevention and treatment of cancer, osteoporosis, autoimmune diseases and inflammatory diseases. Since TNF- ⁇ shows protection against infection and anti-tumor action by inducing apoptosis and enhancing antibody production by inflammatory mediators and plasma cells, antisense nucleotides that can enhance the expression of TNF- ⁇ prevent infection and cancer. It is effective for treatment.
- TNF- ⁇ is involved in the pathogenesis of rheumatoid arthritis, psoriasis, diabetes, hyperlipidemia, sepsis, and osteoporosis
- antisense nucleotides that can suppress the expression of TNF- ⁇ are used to prevent and treat these diseases. Useful for.
- the medicament containing the antisense nucleotide of the present invention has low toxicity, and as such a solution or a pharmaceutical composition of an appropriate dosage form, can be used as a human or non-human mammal (eg, mouse, rat, guinea pig, rabbit, sheep, Pigs, cows, cats, dogs, monkeys, etc.) can be administered orally or parenterally (eg, inhalation administration, intravascular administration, subcutaneous administration, transmucosal administration, etc.).
- these nucleic acids are used as a prophylactic / therapeutic agent for the above-mentioned various diseases, they can be formulated and administered according to a method known per se.
- the antisense nucleotide of the present invention is inserted alone or in a functional manner into the appropriate expression vector for mammalian cells such as a retroviral vector, lentiviral vector, adenoviral vector, adeno-associated viral vector and the like. Then, it can be formulated according to conventional means.
- the nucleotide can be administered as it is or together with an auxiliary agent for promoting intake by a gene gun or a catheter such as a hydrogel catheter. Alternatively, it can be aerosolized and locally administered into the trachea as an inhalant.
- the nucleotide may be formulated (injection) alone or with a carrier such as a liposome and administered intravenously, subcutaneously, etc. .
- the antisense nucleotide of the present invention may be administered per se or as an appropriate pharmaceutical composition.
- the pharmaceutical composition used for administration may contain the antisense nucleotide of the present invention and a pharmacologically acceptable carrier, diluent or excipient.
- a pharmaceutical composition is provided as a dosage form suitable for oral or parenteral administration.
- compositions for parenteral administration for example, injections, aerosols, suppositories and the like are used, and injections are intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections, intravenous injections, etc. May be included.
- Such an injection can be prepared according to a known method. Aerosol formulations can be placed in compressed acceptable propellants such as dichlorodifluoromethane, propane, nitrogen and the like. Alternatively, it may be formulated as an incompressible pharmaceutical product such as a nebulizer or an atomizer.
- Suppositories used for rectal administration may be prepared by mixing the nucleic acid with a normal suppository base.
- compositions for oral administration include solid or liquid dosage forms, specifically tablets (including sugar-coated tablets and film-coated tablets), pills, granules, powders, capsules (including soft capsules), and syrups. Agents, emulsions, suspensions and the like.
- Such a composition is produced by a known method and may contain a carrier, a diluent or an excipient usually used in the pharmaceutical field.
- a carrier and excipient for tablets for example, lactose, starch, sucrose, and magnesium stearate are used.
- the above parenteral or oral pharmaceutical composition is conveniently prepared in a dosage unit form suitable for the dosage of the active ingredient.
- dosage forms include tablets, pills, capsules, injections (ampoules), aerosols, and suppositories.
- the antisense nucleotide of the present invention is preferably contained, for example, usually 5 to 500 mg per dosage unit dosage form, particularly 5 to 100 mg for injections, and 10 to 250 mg for other dosage forms.
- the dosage of the above-mentioned medicament containing the antisense nucleotide of the present invention varies depending on the administration subject, target disease, symptom, administration route, etc., but for example, the dosage of the antisense nucleotide of the present invention is usually 0.01 to 20 mg. It is convenient to administer about 0.1 kg / kg body weight, preferably about 0.1-10 mg / kg body weight, more preferably about 0.1-5 mg / kg body weight by intravenous injection or inhalation once to several times a day. In the case of other parenteral administration and oral administration, an equivalent amount can be administered. If symptoms are particularly severe, the dose may be increased according to the symptoms.
- the above-described pharmaceutical composition may contain other drugs as long as an undesirable interaction is not caused by the combination with the antisense nucleotide of the present invention.
- Other drugs include, for example, antiviral drugs, antitumor drugs, antibacterial drugs, antifungal drugs, antiprotozoal drugs, antibiotics, antiseptic drugs, antiseptic shock drugs, endotoxin antagonists, immunomodulators, non-steroidal drugs
- anti-inflammatory drugs steroid drugs, inflammatory mediator action inhibitors, inflammatory mediator production inhibitors, anti-inflammatory mediator action inhibitors, anti-inflammatory mediator production inhibitors.
- Oligonucleotides that contain a nucleotide sequence that is complementary to the nucleotide sequence of the endogenous antisense transcript (NAT) inhibits NAT's regulation of expression on the target mRNA. It is possible to regulate the stability of the target mRNA and regulate the expression of the target gene (protein production) positively or negatively. Therefore, the present invention also provides a sense oligonucleotide homologous to the 3 'UTR of a gene having an expression-regulating activity of a gene in which NAT is induced, induced by IL-1 ⁇ .
- the target gene (mRNA) is the same as described above for the antisense nucleotide of the present invention.
- the sense oligonucleotide of the present invention includes a sequence complementary to NAT (and thus a sequence homologous to the 3 ′ UTR), which contains a sequence complementary to the 3 ′ UTR of the target mRNA, and changes the stability of the target mRNA.
- Any oligonucleotide can be used as long as it can be used.
- the “complementary” sequence is not only a sequence that is completely complementary to NAT, but as long as it can hybridize with NAT under physiological conditions of cells and inhibit the action of NAT on mRNA, It may contain one to several (2, 3, 4 or 5) base mismatches.
- the sequence complementary to the target NAT refers to stringent conditions such as those described in Current Protocols in Molecular Biology, John Wiley & Sons, 6.3.1-6.3.6, 1999 (eg 6 ⁇ Under the condition of SSC (sodium chloride / sodium citrate) / 45 ° C hybridization, followed by 0.2 ⁇ SSC / 0.1% SDS / 50-65 ° C one or more washings).
- SSC sodium chloride / sodium citrate
- the antisense nucleotide against the target mRNA interacts with a thermodynamically unstable portion of the mRNA to regulate the stability of the mRNA
- the sense oligonucleotide of the present invention contains the target oligonucleotide in the target mRNA.
- the “homologous” sequence is not only a sequence that is completely identical to the specific partial nucleotide sequence of the target mRNA, but also the action of NAT on the mRNA by hybridizing with NAT on the mRNA under physiological conditions of the cell. 1 to several (2, 3, 4 or 5) bases may be different as long as they can be inhibited.
- the “homologous” sequence is 90% or more identity to the target mRNA in the overlapping region when the sequence of the target site of the target mRNA and the sequence of the sense oligonucleotide are aligned.
- the sense oligonucleotide of the present invention does not contain a sequence that causes a non-sequence-specific reaction (for example, 5′-CG-3 ′, 5′-GGGG-3 ′, 5′-GGGGG-3 ′, etc.). It is preferable to select from those having no similar sequence in RNA other than the target mRNA. The absence of similar sequences in other RNAs can be confirmed by methods similar to those described above for antisense oligonucleotides.
- the sense oligonucleotide for CCL2 NAT of the present invention in the rat CCL2 mRNA 3'UTR sequence shown in SEQ ID NO: 1, is represented by domain 1 shown by nucleotide numbers 25-51 or nucleotide numbers 103-175
- the sequence corresponding to each domain in the rat CCL2 includes at least a part of one or more loop structures (for example, 3 bases or more). Contains a homologous nucleotide sequence.
- TTAAGTAATGTTAAACTTAT (CCL2-Se1; SEQ ID NO: 8) that can enhance the expression of CCL2 mRNA as a sense oligonucleotide homologous to domain 1
- CCL2 mRNA as a sense oligonucleotide that is homologous to domain 2
- Examples include, but are not limited to, TCCATTTTTTTATTTCTCTG (CCL2-Se2; SEQ ID NO: 9) that can be enhanced.
- the sense oligonucleotide for CCL20 NAT of the present invention is represented by the domain 1 shown by nucleotide numbers 65-107 and nucleotide numbers 124-155 in the rat CCL20 mRNA 3'UTR sequence shown by SEQ ID NO: 2.
- domain 2 indicated by nucleotide 2 or nucleotide number 214-282, or in the CCL20 ortholog in other mammals at least a part of one or more loop structures in the domain corresponding to each domain in the rat CCL20 It includes a nucleotide sequence that is homologous to a sequence that includes (eg, 3 bases or more).
- GGTTTCACCTGCACATCACT (CCL20-Se1; SEQ ID NO: 10) can suppress the expression of CCL20 mRNA
- a sense oligonucleotide homologous to domain 3 expression of CCL20 mRNA Examples include, but are not limited to, GTTTAGCTATTTAATGTTAA (CCL2-Se2; SEQ ID NO: 11) that can be enhanced.
- the sense oligonucleotide for CX3CL1 NAT of the present invention in the 3′UTR sequence of rat CX3CL1 mRNA shown in SEQ ID NO: 3, is in domain 1, nucleotide numbers 1305-1369 shown as nucleotide numbers 1103-1202.
- the CX3CL1 ortholog in other mammals contains a nucleotide sequence homologous to a sequence containing at least a part of one or more loop structures in the domain corresponding to each domain in the rat CX3CL1.
- ACTTGTGCATGTGTGTACTT (CX3CL1-Se1; SEQ ID NO: 13) can enhance the expression of CX3CL1 mRNA as a sense oligonucleotide homologous to domain 1, and CX3CL1 mRNA as a sense oligonucleotide homologous to domain 2
- Examples include, but are not limited to, ACAAAGTGTCTACTGAAGCA (CX3CL1-Se2; SEQ ID NO: 14) and CTACTGAAGCAGAGAGCAGC (CX3CL1-Se3; SEQ ID NO: 15) that can suppress the expression of CX3CL1 mRNA.
- the sense oligonucleotide for IL-23A NAT of the present invention in the 3′UTR sequence of rat IL-23A mRNA shown in SEQ ID NO: 4, is the domain 1, nucleotide number shown by nucleotide numbers 37-55 In the domain 2 represented by 193-303, the domain 3 represented by nucleotide numbers 380-448 or the domain 4 represented by nucleotide numbers 555-600, and the IL-23A ortholog in other mammals, the rat IL- A nucleotide sequence homologous to a sequence comprising at least part of one or more loop structures within the domain corresponding to each domain of 23A.
- a sense oligonucleotide homologous to domain 1 AATCCATCAATGCAGACATC (IL23-Se1; SEQ ID NO: 16) capable of suppressing the expression of IL-23A mRNA
- IL-23A GAAGCTGGCAGACAGCTGCA IL23-Se2; SEQ ID NO: 17
- TCCTTCAGTTCTAACAGAAC IL23-Se3; SEQ ID NO: 18
- the sense oligonucleotide homologous to domain 4 include, but are not limited to, AACAGTTTAGAGGATTGTTA (IL23-Se4; SEQ ID NO: 19) that can suppress the expression of IL-23A mRNA.
- the sense oligonucleotide for CD69 NAT of the present invention is the domain 1 represented by nucleotide numbers 15-32 and nucleotide numbers 120-186 in the 3′UTR sequence of rat CD69 mRNA represented by SEQ ID NO: 5.
- domain 2 indicated in domain 3 indicated by nucleotide numbers 218-252, domain 4 indicated by nucleotide numbers 344-370, domain 5 indicated by nucleotide numbers 389-633 or domain 6 indicated by nucleotide numbers 656-786.
- the CD69 ortholog in other mammals contains a nucleotide sequence homologous to a sequence containing at least a part of one or more loop structures in the domain corresponding to each domain in the rat CD69.
- GCCAATGCTTATGAAAACA can enhance the expression of CD69 mRNA
- IL-23A ⁇ mRNA examples include, but are not limited to, GTGGCAGATCTCTGTCAGGA (CD69-Se3; SEQ ID NO: 22) that can enhance expression.
- the sense oligonucleotide for NF- ⁇ B p65 NAT of the present invention is the domain 1 represented by nucleotide numbers 161-301 in the 3 ′ UTR sequence of rat NF- ⁇ B p65 mRNA represented by SEQ ID NO: 6,
- domain 2 represented by nucleotide number 343-380
- domain 3 represented by nucleotide number 401-412 or domain 4 represented by nucleotide number 473-523
- the NF- ⁇ B p65 ortholog in other mammals A nucleotide sequence homologous to a sequence containing at least a part of one or more loop structures in the domain corresponding to each domain in rat NF- ⁇ B p65.
- a sense oligonucleotide homologous to domain 1 GAACTCTTGAGACCCTGCTT (p65-Se1; SEQ ID NO: 23) capable of enhancing the expression of NF- ⁇ B ⁇ p65 mRNA
- NF- GCAACGCTCCTAGGAGCAGC p65-Se3; SEQ ID NO: 25
- AACTCTCCATGCTGAGCAGT p65-Se4; sequence that can suppress the expression of NF- ⁇ B p65 mRNA as a homologous sense oligonucleotide to domain 4 No. 26
- the sense oligonucleotide for TNF- ⁇ NAT of the present invention is the domain 1, nucleotide number 430 represented by nucleotide number 304-405 in the 3′UTR sequence of rat TNF- ⁇ mRNA represented by SEQ ID NO: 7.
- the rat TNF- ⁇ A nucleotide sequence homologous to a sequence comprising at least part of one or more loop structures within the domain corresponding to each domain in For example, AGATGTCTCAGGCCTCCCTT (TNF-Se1; SEQ ID NO: 27) capable of enhancing the expression of TNF- ⁇ mRNA as a sense oligonucleotide homologous to domain 1, and TNF- ⁇ as a sense oligonucleotide homologous to domain 2 GGAACCCCCTATATTTATAA (TNF-Se2; SEQ ID NO: 28) that can enhance the expression of mRNA and TAATTGCACCTGTGACTATT (TNF-Se3; SEQ ID NO: 29) that can suppress the expression of TNF- ⁇ mRNA, as a sense oligon
- the length of the sense oligonucleotide of the present invention is not particularly limited, but from the viewpoint of sequence specificity, the portion complementary to the target sequence in the target NAT is at least 10 bases or more, preferably about 12 bases or more, more preferably Contains about 15 bases or more.
- the portion complementary to the target sequence in the target NAT is at least 10 bases or more, preferably about 12 bases or more, more preferably Contains about 15 bases or more.
- those having a base length of 50 bases or less, preferably 40 bases or less, more preferably 30 bases or less are mentioned.
- the sense oligonucleotide of the present invention may be any of single-stranded DNA, single-stranded RNA, and DNA / RNA chimera, and may be those with known modifications.
- the “nucleotide” may include not only purine and pyrimidine bases but also those having other modified heterocyclic bases.
- the sense oligonucleotide is DNA (ODN)
- the RNA: DNA hybrid formed by the target NAT and the sense ODN can be recognized by endogenous RNase H and cause selective degradation of the target NAT.
- the nucleotide molecule constituting the sense oligonucleotide may be natural DNA or RNA, but in order to improve stability (chemical and / or enzyme) and specific activity (affinity with RNA), the above antisense As with nucleotides, various chemical modifications can be included.
- the sense oligonucleotide of the present invention synthesizes a sequence homologous to the target mRNA (cDNA) sequence using a commercially available DNA / RNA automatic synthesizer (Applied Biosystems, Beckman, etc.). Can be prepared.
- the sense oligonucleotide of the present invention is provided in a special form such as a liposome or a microsphere, or a form to which a hydrophobic substance such as a polycationic substance such as polylysine or a lipid (eg, phospholipid, cholesterol, etc.) is added. Can be provided at.
- the sense oligonucleotide of the present invention is converted into a peptide having a membrane permeation function (for example, Drosophila-derived Antennapedia homeodomain (AntP), human immunodeficiency virus (HIV) -derived TAT, herpes simplex virus (HSV) -derived VP22. And the like can be promoted by incorporating the oligonucleotide into a cell.
- a membrane permeation function for example, Drosophila-derived Antennapedia homeodomain (AntP), human immunodeficiency virus (HIV) -derived TAT, herpes simplex virus
- the sense oligonucleotide of the present invention can change the target gene expression (protein production) regulating action by the NAT by interacting with the target NAT. Since various genes including cytokine / chemokine gene induced by IL-1 ⁇ have various physiological activities such as inflammation-inducing or anti-inflammatory action, infection defense, and regulation of immune response, the sense oligonucleotide of the present invention is used.
- the contained drug can be used as a target gene expression regulator for the prevention and / or treatment of various diseases including inflammatory diseases and infectious diseases.
- sense oligonucleotides that can enhance the expression of CCL2 can be used for angiogenesis and wound healing, while CCL2 migrates to monocytes and eosinophils, so that it can suppress the expression of CCL2.
- Nucleotides are useful for the prevention and treatment of chronic inflammatory diseases including arteriosclerosis and allergic inflammation. Since CCL20 exhibits antibacterial activity against a wide range of microorganisms, sense oligonucleotides that can enhance the expression of CCL20 are effective in the prevention and treatment of various microbial infections.
- sense oligonucleotides that can suppress the expression of CCL20 are bronchial asthma, multiple sclerosis, rheumatoid arthritis, various dermatitis, inflammatory It is effective for the prevention and treatment of autoimmune diseases such as bowel disease.
- Sense oligonucleotides that can enhance CX3CL1 expression can be used for angiogenesis, while sense oligonucleotides that can suppress CX3CL1 expression prevent or treat inflammatory diseases including arteriosclerosis and glomerulonephritis It is useful for the prevention and treatment of anti-rejection and rheumatoid arthritis.
- IL-23A is important for the maintenance of cellular immunity, and its overexpression causes multi-organ inflammation. Therefore, sense oligonucleotides that can enhance IL-23A expression are effective in the prevention and treatment of infectious diseases. On the other hand, sense oligonucleotides that can suppress the expression of IL-23A are useful for the prevention and treatment of autoimmune diseases and inflammatory diseases. Since CD69 plays an important role in the differentiation and activation of lymphocytes, a sense oligonucleotide that can enhance the expression of CD69 is effective in the prevention and treatment of infectious diseases.
- CD69 is expressed in most inflammatory cells that are expressed locally, and autoantibodies against CD69 are detected in patients with rheumatoid arthritis and systemic lupus erythematosus (SLE). Nucleotides are useful for the prevention and treatment of autoimmune diseases and inflammatory diseases. Since NF- ⁇ B is a transcription factor having an important role in the immune system, a sense oligonucleotide that can enhance the expression of NF- ⁇ B p65 is effective in the prevention and treatment of infectious diseases.
- NF- ⁇ B is constantly activated in cancer cells and inhibits apoptosis, and is also involved in pathogenesis such as bone metabolism, bronchial asthma, arthritis, inflammatory bowel disease, sepsis,
- a sense oligonucleotide capable of suppressing the expression of NF- ⁇ B p65 is useful for the prevention and treatment of cancer, osteoporosis, autoimmune diseases and inflammatory diseases.
- TNF- ⁇ shows protection against infection and anti-tumor action by inducing apoptosis and enhancing antibody production by inflammatory mediators and plasma cells. Therefore, sense oligonucleotides that can enhance the expression of TNF- ⁇ prevent infection and cancer. It is effective for treatment.
- TNF- ⁇ is involved in the pathogenesis of rheumatoid arthritis, psoriasis, diabetes, hyperlipidemia, sepsis, and osteoporosis
- sense oligonucleotides that can suppress the expression of TNF- ⁇ are used to prevent and treat these diseases. Useful for.
- the medicament containing the sense oligonucleotide of the present invention has low toxicity and can be used as a liquid or as a pharmaceutical composition of an appropriate dosage form as a human or non-human mammal (eg, mouse, rat, guinea pig, rabbit, sheep, Pigs, cows, cats, dogs, monkeys, etc.) can be administered orally or parenterally (eg, inhalation administration, intravascular administration, subcutaneous administration, transmucosal administration etc.).
- these nucleic acids are used as a prophylactic / therapeutic agent for the above-mentioned various diseases, they can be formulated and administered in the same manner as the antisense nucleotide of the present invention.
- the dose of the above-mentioned medicament containing the sense oligonucleotide of the present invention varies depending on the administration subject, target disease, symptom, administration route, etc.
- the dose of the sense oligonucleotide of the present invention is usually 0.01 to 20 mg. It is convenient to administer about 0.1 kg / kg body weight, preferably about 0.1-10 mg / kg body weight, more preferably about 0.1-5 mg / kg body weight by intravenous injection or inhalation once to several times a day. In the case of other parenteral administration and oral administration, an equivalent amount can be administered. If symptoms are particularly severe, the dose may be increased according to the symptoms.
- the above-described pharmaceutical composition may contain other drugs as long as no undesirable interaction is caused by the combination with the sense oligonucleotide of the present invention.
- Other drugs include, for example, antiviral drugs, antitumor drugs, antibacterial drugs, antifungal drugs, antiprotozoal drugs, antibiotics, antiseptic drugs, antiseptic shock drugs, endotoxin antagonists, immunomodulators, non-steroidal drugs
- anti-inflammatory drugs steroid drugs, inflammatory mediator action inhibitors, inflammatory mediator production inhibitors, anti-inflammatory mediator action inhibitors, anti-inflammatory mediator production inhibitors.
- the present invention also provides a method of screening for a substance that regulates target gene expression (protein production) by regulating the action of NAT on the target mRNA.
- the screening method of the present invention is characterized by detecting the hybridization between a target mRNA and NAT for the target mRNA in the presence and absence of a test substance and comparing the degree thereof.
- target mRNA and NAT are isolated by a conventional method, one of them is solid-phased and the other is labeled with an appropriate labeling agent, and the test is performed under conditions where RNA can form a physiological secondary structure.
- Examples include a method of hybridizing both in the presence and absence of a substance and comparing the amount of label bound to the solid phase under both conditions.
- mRNA and NAT the full length thereof may be used, respectively, or the 3 ′ UTR of mRNA and a fragment containing a NAT sequence complementary to the region may be used.
- the solid phase material examples include semiconductors such as silicon, inorganic materials such as glass and diamond, films mainly composed of high molecular substances such as polyethylene terephthalate and polypropylene, and the shape of the solid phase includes a slide glass, Examples include, but are not limited to, microwell plates, microbeads, and fiber types.
- functional groups such as amino group, aldehyde group, SH group, and biotin are introduced into the RNA in advance, while reacting with the RNA on the solid phase.
- Functional groups eg, aldehyde group, amino group, SH group, streptavidin, etc.
- the solid phase and RNA are cross-linked by covalent bond between the two functional groups, or the polyanionic RNA is immobilized.
- the method include, but are not limited to, a method of immobilizing RNA using polycation coating of a phase and electrostatic bonding.
- the solid-phase RNA preparation methods include the Affymetrix method, which synthesizes RNA one nucleotide at a time on a substrate (glass, silicon, etc.) using a photolithography method, the micro spotting method, the inkjet method, and the bubble jet (registered trademark) method
- the Stanford method in which RNA prepared in advance is spotted on the substrate using the above method, but considering the base length of the RNA to be used, it is preferable to use the Stanford method or a method combining both.
- a radioisotope for example, an enzyme, a fluorescent substance, a luminescent substance, or the like is used.
- the radioisotope for example, [ 32 P], [ 3 H], [ 14 C] and the like are used.
- the enzyme a stable enzyme having a large specific activity is preferable.
- ⁇ -galactosidase, ⁇ -glucosidase, alkaline phosphatase, peroxidase, malate dehydrogenase and the like are used.
- the fluorescent material for example, fluorescamine, fluorescein isothiocyanate, Cy3, Cy5 and the like are used.
- luminescent substance for example, luminol, luminol derivatives, luciferin, lucigenin and the like are used.
- biotin- (strept) avidin can also be used for binding between the probe and the labeling agent.
- the test substance may be any known substance or new substance, such as a nucleic acid, carbohydrate, lipid, protein, peptide, organic low molecular weight compound, compound library prepared using combinatorial chemistry techniques, solid phase Examples include random peptide libraries prepared by synthesis or phage display methods, or natural components derived from microorganisms, animals and plants, marine organisms, and the like.
- concentration of the test substance to be added varies depending on the type of compound (solubility, toxicity, etc.), but is appropriately selected within the range of, for example, about 0.1 nM to about 100 nM. Examples of the incubation time include about 1 to about 24 hours.
- the test substance After bringing the RNA on the solid phase into contact with the labeled RNA (and the test substance) and incubating, the RNA that did not bind to the solid phase is washed away, and the amount of RNA bound to the solid phase is detected.
- the test substance can be selected as a candidate for a substance that enhances the action of NAT on the target mRNA.
- the test substance can be selected as a candidate for a substance that suppresses the action of NAT on the target mRNA. it can.
- a target substance is contacted more directly by contacting a test substance with a cell that expresses the target mRNA and NAT, and measuring a change in the amount of the mRNA and / or protein encoded in the cell.
- Substances that enhance or suppress gene expression can be selected.
- the cell that expresses the target mRNA and NAT may be a cell that can naturally express both RNAs (for example, IL-1 ⁇ -stimulated hepatocytes), or a DNA that expresses either or both of them.
- the introduced recombinant cell may also be used.
- examples of host cells include animal cells such as H4IIE-C3 cells, HepG2 cells, 293T cells, HEK293 cells, COS7 cells, 2B4T cells, CHO cells, MCF-7 cells, and H295R cells.
- the target mRNA and DNA encoding NAT are both isolated by conventional methods, converted into double-stranded DNA by reverse transcription, etc., and then inserted into an expression vector having a promoter that can function in the host cell.
- it can be prepared by introducing this vector into a host cell by the calcium phosphate coprecipitation method, PEG method, electroporation method, microinjection method, lipofection method or the like.
- test substance those described above are used.
- a medium suitable for culturing the cell for example, a minimum essential medium (MEM) containing about 5 to 20% fetal calf serum, Dulbecco's modified Eagle medium (DMEM), RPMI1640 Medium, 199 medium, F12 medium, etc.
- various buffers eg, HEPES buffer, phosphate buffer, phosphate buffered saline, Tris-HCl buffer, borate buffer, acetate buffer, etc.
- HEPES buffer phosphate buffer, phosphate buffered saline, Tris-HCl buffer, borate buffer, acetate buffer, etc.
- the concentration of the test substance to be added varies depending on the type of compound (solubility, toxicity, etc.), but is appropriately selected within the range of, for example, about 0.1 nM to about 100 nM.
- Examples of the incubation time include about 1 to about 48 hours. If necessary, the cells may be infected with the virus during the incubation.
- RNA is extracted from the cells and the target mRNA level is measured by RT-PCR, real-time PCR or Northern blot analysis, or the culture supernatant is collected and various immunoassays known per se Or the amount of protein encoded by the target gene is measured by Western blotting or the like.
- the test substance can be selected as a candidate for a target gene expression enhancing substance.
- the test substance can be selected as a candidate for a target gene expression inhibitor.
- Substances capable of enhancing or suppressing the expression of the target gene selected by the screening method described above are used for the prevention and / or prevention of the above-mentioned various diseases in the same manner as antisense nucleotides and sense oligonucleotides that can enhance or suppress the expression of the gene. Alternatively, it can be used as a therapeutic drug.
- a medicine containing a substance selected by the above screening method has low toxicity, and can be used as a liquid or as a pharmaceutical composition of an appropriate dosage form as a human or non-human mammal (eg, mouse, rat, guinea pig, rabbit). , Sheep, pigs, cows, cats, dogs, monkeys, etc.) or orally (eg, inhalation administration, intravascular administration, subcutaneous administration, etc.).
- the pharmaceutical composition used for administration may comprise a selected substance and a pharmacologically acceptable carrier, diluent or excipient.
- compositions for parenteral administration for example, injections, aerosols, suppositories and the like are used, and injections are intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections, intravenous injections, etc. May be included.
- Such an injection can be prepared according to a known method.
- a method for preparing an injection it can be prepared, for example, by dissolving, suspending or emulsifying a substance for enhancing or suppressing the expression of a selected target gene in a sterile aqueous liquid or oily liquid that is usually used for injection.
- an aqueous solution for injection for example, an isotonic solution containing physiological saline, glucose and other adjuvants, and the like are used, and suitable solubilizers such as alcohol (eg, ethanol), polyalcohol (eg, Propylene glycol, polyethylene glycol), nonionic surfactants (eg, polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct-of-hydrogenated-castor-oil)) and the like may be used in combination.
- alcohol eg, ethanol
- polyalcohol eg, Propylene glycol, polyethylene glycol
- nonionic surfactants eg, polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct-of-hydrogenated-castor-oil)
- oily liquid for example, sesame oil, soybean oil and the like are used, and benzyl benzoate, benzyl alcohol and the like may be used in combination as a solub
- the prepared injection solution is preferably filled in a suitable ampoule.
- Aerosol formulations can be placed in compressed acceptable propellants such as dichlorodifluoromethane, propane, nitrogen and the like. Alternatively, it may be formulated as a non-compressible pharmaceutical product such as a nebulizer or an atomizer.
- a suppository used for rectal administration may be prepared by mixing a substance for enhancing or suppressing the expression of the target gene with a normal suppository base.
- compositions for oral administration include solid or liquid dosage forms, specifically tablets (including sugar-coated tablets and film-coated tablets), pills, granules, powders, capsules (including soft capsules), and syrups. Agents, emulsions, suspensions and the like.
- Such a composition is produced by a known method and may contain a carrier, a diluent or an excipient usually used in the pharmaceutical field.
- a carrier and excipient for tablets for example, lactose, starch, sucrose, and magnesium stearate are used.
- the above parenteral or oral pharmaceutical composition is conveniently prepared in a dosage unit form suitable for the dosage of the active ingredient.
- dosage forms include tablets, pills, capsules, injections (ampoules), aerosols, and suppositories.
- the substance for enhancing or suppressing the expression of the target gene is usually contained in an amount of 5 to 500 mg per dosage unit dosage form, particularly 5 to 100 mg for injections and 10 to 250 mg for other dosage forms.
- the dose of the above-mentioned pharmaceutical containing a substance that enhances or suppresses the expression of the target gene varies depending on the administration subject, target disease, symptom, administration route, etc., but for example, the substance is usually 0.01 to 20 mg per dose It is convenient to administer about 0.1 kg / kg body weight, preferably about 0.1 to 10 mg / kg body weight, more preferably about 0.1 to 5 mg / kg body weight by intravenous injection once to several times a day. In the case of other parenteral administration and oral administration, an equivalent amount can be administered. If symptoms are particularly severe, the dose may be increased according to the symptoms.
- Hepatocytes were isolated from male Wistar rats (SPF / VAF Crlj: WI; Charles River Japan) as described in Prostaglandins 1993; 45: 459-474 and seeded at 37 ° C. Incubated overnight. Hepatocytes (3.0 ⁇ 10 5 cells / dish) were transfected with sense ODN. Each ODN (1.5 ⁇ g) was mixed with 1.5 ⁇ L of magnet-assisted transfection A reagent (IBA), added to each well, incubated on the magnetic plate for 15 minutes, and then the medium was treated with fresh William's E medium (Sigma-Aldrich). ).
- IBA magnet-assisted transfection A reagent
- the cells were cultured overnight and treated with 1 nM human IL-1 ⁇ (Otsuka Pharmaceutical Co., Ltd.) for 4 hours (RNA preparation) or 8 hours (cell extraction). Animal experiments were conducted with the approval of the Animal Experiment Committee of Ritsumeikan University Biwako Kusatsu Campus.
- the secondary structure of 3'UTR of mRNA was predicted by the mfold program (Zuker). A common conserved region was selected from the predicted structure of the 3'UTR of mRNA. Each region contained at least one stem-loop structure.
- Sense ODN Sense ODN protected with a phosphothioate bond was designed according to the method described in J. Neurochem. 2003; 86: 374-382 (Gene Design).
- the designed sense ODN sequence corresponds to an mRNA sequence containing at least one loop of a common region conserved between human and rat, and CpG motifs and G-quartets were avoided.
- Tables 2 and 3 summarize the PCR primers, NAT reverse transcription primers and PCR primers used for mRNA amplification for the seven genes CCL2, CCL20, CX3CL1, IL-23A, CD69, NF- ⁇ B p65 and TNF- ⁇ . It was.
- the present inventors selected 7 genes (CCL2, CCL20, CX3CL1, IL-23A, CD69, NF- ⁇ B p65 and TNF- ⁇ ) in which both mRNA and NAT are abundantly transcribed, and applied NATRE technology. .
- the present inventors predicted the secondary structure of the 3′UTR of these mRNAs (FIGS. 1A-7A), and each sense ODN competes with the mRNA that leads to inhibition of the mRNA-NAT interaction.
- Sense ODNs were designed according to the rule of containing at least one loop of the storage region (FIGS. 1B-7B) (Table 5 and FIGS. 1B-7B).
- Elongation factor 1alpha (EF) mRNA was used as an internal standard, and the values in the graph indicate each mRNA / EF mRNA (%), and were expressed as a relative amount with respect to the expression level in non-sense ODN-introduced cells.
- Sense ODNs designed for RNA sequences that contain loop portions within conserved regions of predicted secondary structure have altered (increased or decreased) the expression level of the target mRNA, whereas Sense ODNs (TNF-Se4 and TNF-Se6) designed outside the conserved region of secondary structure did not affect the expression level of the target mRNA.
- the antisense nucleotide and sense oligonucleotide of the present invention are used as a regulator of the expression of an inflammation / infection-related gene in which an endogenous antisense transcript is present, which is induced by IL-1 ⁇ . Useful for treatment.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Genetics & Genomics (AREA)
- General Health & Medical Sciences (AREA)
- Engineering & Computer Science (AREA)
- Medicinal Chemistry (AREA)
- Molecular Biology (AREA)
- Pharmacology & Pharmacy (AREA)
- Veterinary Medicine (AREA)
- Animal Behavior & Ethology (AREA)
- Biomedical Technology (AREA)
- Public Health (AREA)
- General Engineering & Computer Science (AREA)
- Biotechnology (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Biophysics (AREA)
- Physics & Mathematics (AREA)
- Pain & Pain Management (AREA)
- Plant Pathology (AREA)
- Rheumatology (AREA)
- Microbiology (AREA)
- Endocrinology (AREA)
- Epidemiology (AREA)
- Biochemistry (AREA)
- Communicable Diseases (AREA)
- Oncology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Description
本発明は、炎症や感染に関与するサイトカイン・ケモカインのmRNAに相補的であり、かつ該mRNAの安定性を変化させる天然(内在性)アンチセンス転写物に相同な核酸(アンチセンスヌクレオチド)、及びそのサイトカイン・ケモカインmRNA安定性調節作用の利用、並びに、炎症や感染に関与するサイトカイン・ケモカインのmRNAに相補的であり、かつ該mRNAの安定性を変化させる天然(内在性)アンチセンス転写物に相補的な核酸(センスヌクレオチド)、及びそのサイトカイン・ケモカインmRNA安定性調節作用の利用に関する。 The present invention relates to a nucleic acid (antisense nucleotide) that is complementary to a natural (endogenous) antisense transcript that is complementary to the mRNA of a cytokine chemokine involved in inflammation or infection, and changes the stability of the mRNA, and Utilization of its cytokine / chemokine mRNA stability regulating action, and a natural (endogenous) antisense transcript that is complementary to the cytokine / chemokine mRNA involved in inflammation and infection and changes the stability of the mRNA The present invention relates to a complementary nucleic acid (sense nucleotide) and use of its cytokine / chemokine mRNA stability regulating action.
天然(内在性)アンチセンス転写物(Natural Antisense Transcript; 以下、「NAT」ともいう)とは、生物に内在する、mRNAに対して相補的なヌクレオチド配列を持ったRNAであり、具体的には、センス遺伝子をコードするDNA鎖(すなわちmRNAの非鋳型鎖)を鋳型として合成されたRNAである。センス-アンチセンスRNA間には2本鎖形成能があり、例えば、2本鎖RNAはRNA干渉が働く際に必要であること、及びマイクロRNA(miRNA)と呼ばれる小さなRNAによるタンパク質の翻訳制御に2本鎖RNAが関与することなどが知られている。最近の網羅的なcDNA解析により、かなり多くのNATが転写されていることが明らかとなった(非特許文献1、2)。例えば、マウスでは約2,500対(非特許文献3)、ヒトでは約2,600対(非特許文献4)のセンス-アンチセンスRNAペアが存在することが示唆されており、それらの中にはタンパク質をコードしない非翻訳性のNATが多く含まれている。 Natural (endogenous) antisense transcript (N atural A ntisense T ranscript; hereinafter also referred to as "NAT") and, inherent in the organism, a RNA having a nucleotide sequence complementary to mRNA, specifically Specifically, it is RNA synthesized using a DNA strand encoding a sense gene (that is, a non-template strand of mRNA) as a template. Sense-antisense RNA has the ability to form double strands. For example, double-stranded RNA is necessary for RNA interference, and is used to control protein translation by small RNAs called microRNAs (miRNAs). It is known that double-stranded RNA is involved. Recent comprehensive cDNA analysis revealed that a considerable amount of NAT was transcribed (Non-patent Documents 1 and 2). For example, it has been suggested that about 2,500 pairs (Non-patent Document 3) exist in mice and about 2,600 pairs (Non-patent Document 4) exist in humans. Does not contain a lot of untranslatable NAT.
NATの生理機能については十分に解明されていないが、これまでの研究から、NATは、mRNAの安定化もしくは不安定化、mRNAからの翻訳抑制など、異なる生理機能を有する多様な制御性RNAのグループに属することがわかってきている(非特許文献5)。
例えば、内皮型一酸化窒素合成酵素(eNOS)mRNAに相補的なNATは、ヒストン脱アセチル化酵素阻害剤の存在下で増加してeNOS mRNA量を減少させることが報告されている(非特許文献6)。また、酵母においても、PHO84遺伝子のNATはヒストン脱アセチル化を介してmRNAの発現を抑制することが知られている(非特許文献7)。
Although the physiological function of NAT has not been fully elucidated, NAT has shown that various regulatory RNAs with different physiological functions such as stabilization or destabilization of mRNA, translational suppression from mRNA, etc. It has been found that it belongs to a group (Non-Patent Document 5).
For example, it has been reported that NAT complementary to endothelial nitric oxide synthase (eNOS) mRNA increases in the presence of a histone deacetylase inhibitor and decreases the amount of eNOS mRNA (non-patent literature). 6). Also in yeast, it is known that NAT of the PHO84 gene suppresses mRNA expression through histone deacetylation (Non-patent Document 7).
一方、本発明者らは、誘導型一酸化窒素合成酵素(iNOS)mRNAの3’-非翻訳領域(UTR)に相補的なNATが存在し、このアンチセンスRNAがiNOS mRNAとハイブリダイズすることによって該mRNAが安定化されiNOSの産生量及びNOの合成量が増大すること、該NATに相補的なセンスオリゴデオキシヌクレオチド(ODN)は、NATのiNOS mRNAへの結合を阻害することにより該mRNAを不安定化し、iNOSの産生及びiNOSによるNO合成を抑制し得ることを報告した(特許文献1及び2、非特許文献8)。また、西澤とその共同研究者らは、インターフェロン(IFN)-α mRNAの核外輸送に重要な、2つのステム-ループを有する二次構造を形成する領域に相補的なNATが存在し、このアンチセンスRNAがIFN-α mRNAの安定化に寄与しており、センスODNによるNATノックダウンがmRNAの不安定化をもたらすのに対し、アンチセンスRNAの過剰発現により該mRNAが顕著に安定化されることを見出した(特許文献3)。本発明者らはこのセンスODNによりNATを制御する手法を「NAT-targeted REgulation(NATRE)技術」と名づけた。
On the other hand, the present inventors have a NAT complementary to the 3′-untranslated region (UTR) of inducible nitric oxide synthase (iNOS) mRNA, and that this antisense RNA hybridizes with iNOS RNA mRNA. The mRNA is stabilized and the production amount of iNOS and the synthesis amount of NO increase, and the sense oligodeoxynucleotide (ODN) complementary to the NAT inhibits the binding of NAT to the iNOS mRNA. It was reported that iNOS production and NO synthesis by iNOS can be suppressed (
ところで、肝細胞において、前炎症性サイトカインであるインターロイキン-1β(IL-1β)に応答して、サイトカインやケモカイン等の炎症や感染に関連する多くの遺伝子が発現するが、IL-1βにより誘導される初期応答遺伝子群からNATが転写されているか否かについてはこれまでに報告がなく、これら炎症/感染関連遺伝子においてセンスODNを用いた発現制御が可能か否かは全く不明のままである。 By the way, many genes related to inflammation and infection such as cytokines and chemokines are expressed in hepatocytes in response to the pro-inflammatory cytokine interleukin-1β (IL-1β), which is induced by IL-1β. It has not been reported so far whether or not NAT is transcribed from the early response gene group, and whether or not expression control using sense ODN is possible in these inflammation / infection-related genes remains unclear. .
本発明の目的は、炎症や感染に関連するサイトカイン・ケモカイン遺伝子の発現を調節し得るNATを探索し、該NATに相補的なセンスオリゴヌクレオチドや該NATと相同なアンチセンスオリゴヌクレオチドを利用した、炎症性疾患やウイルス感染症などの疾患の新規予防・治療手段を提供することである。 The object of the present invention was to search for NAT capable of regulating the expression of cytokines and chemokine genes related to inflammation and infection, and to use a sense oligonucleotide complementary to the NAT and an antisense oligonucleotide homologous to the NAT, It is to provide a novel means for preventing and treating diseases such as inflammatory diseases and viral infections.
本発明者らは、IL-1βにより誘導される21遺伝子(サイトカインおよびケモカイン遺伝子を含む)を選択し、mRNAの3’UTRに相補的なNATを検出しようと試みた。鎖特異的RT-PCRの結果、21遺伝子のうち16遺伝子(76%)からNATが転写されていることが判明した。次に、本発明者らは、mRNAとNATの両方が豊富に転写されている7遺伝子(CCL2、CCL20、CX3CL1、IL-23 p19サブユニット、CD69、NF-κB p65サブユニット、TNF-α)を選択し、これらのmRNAの3’UTRの二次構造を予測し、ヒト-ラット間で保存された領域内の少なくとも1つのループを含むRNA配列に相同なセンスODNを設計・合成して、ラット肝細胞に導入し、IL-1β刺激後の細胞における各遺伝子のmRNAレベルをセンスODN非導入細胞と比較した。
その結果、予測された二次構造の保存された領域内のループ部分を含むRNA配列に対して設計されたほとんどのセンスODNは、標的mRNAの発現レベルを変化(増加又は減少)させたのに対し、該二次構造の保存された領域外に対して設計されたセンスODNは標的mRNAの発現レベルに影響しなかった。
The present inventors selected 21 genes (including cytokines and chemokine genes) induced by IL-1β and attempted to detect NAT complementary to the 3 ′ UTR of mRNA. As a result of strand-specific RT-PCR, it was found that NAT was transcribed from 16 out of 21 genes (76%). Next, we have 7 genes (CCL2, CCL20, CX3CL1, IL-23 p19 subunit, CD69, NF-κB p65 subunit, TNF-α) in which both mRNA and NAT are abundantly transcribed. Predict the secondary structure of the 3'UTR of these mRNAs, design and synthesize a sense ODN that is homologous to an RNA sequence that contains at least one loop within a region conserved between human and rat, Introduced into rat hepatocytes, the mRNA level of each gene in the cells after stimulation with IL-1β was compared with that in cells not transfected with sense ODN.
As a result, most sense ODNs designed for RNA sequences containing loop portions within the conserved region of the predicted secondary structure have altered (increased or decreased) the expression level of the target mRNA. In contrast, a sense ODN designed outside the conserved region of the secondary structure did not affect the expression level of the target mRNA.
本発明者らは、これらの知見に基づいて、IL-1βにより誘導されるサイトカイン・ケモカイン遺伝子の多くに3’UTRに対応する領域を含むNATが存在し、当該遺伝子の発現がNATにより制御されており、さらに該NATがとりうる二次構造のループ部分を含む領域に相補的な(従って、mRNAに相同な)配列を有するセンスオリゴヌクレオチドを用いて、当該遺伝子の発現制御(アップレギュレーションおよびダウンレギュレーション)が可能であると結論して、本発明を完成するに至った。 Based on these findings, the present inventors have a NAT containing a region corresponding to 3′UTR in many cytokine-chemokine genes induced by IL-1β, and the expression of the gene is regulated by NAT. Furthermore, using a sense oligonucleotide having a sequence complementary to the region containing the loop portion of the secondary structure that can be taken by the NAT (hence, homologous to mRNA), expression control (up-regulation and down-regulation) of the gene is performed. It was concluded that (regulation) is possible, and the present invention was completed.
即ち、本発明は以下の通りである。
〔1〕IL-1βにより誘導される、CCL2、CCL20、CX3CL1、IL-23A、CD69、NF-κB p65、TNF-α、Fam89a、Grk5、phosopholipid scramblase 1、Runx1、semaphorin 4A、Steap4、lymphotoxin β Psmb10またはTLR2遺伝子の3’UTRに相補的な配列を含み、該遺伝子の発現を調節し得るアンチセンスヌクレオチド。
〔2〕IL-1βにより誘導される遺伝子が、CCL2、CCL20、CX3CL1、IL-23A、CD69、NF-κB p65またはTNF-αである、上記〔1〕記載のアンチセンスヌクレオチド。
〔3〕上記〔1〕記載のアンチセンスヌクレオチドを含有してなる、CCL2、CCL20、CX3CL1、IL-23A、CD69、NF-κB p65、TNF-α、Fam89a、Grk5、phosopholipid scramblase 1、Runx1、semaphorin 4A、Steap4、lymphotoxin β Psmb10またはTLR2の発現調節剤。
〔4〕炎症性疾患または感染症の予防又は治療用である、上記〔3〕記載の剤。
〔5〕IL-1βにより誘導される、CCL2、CCL20、CX3CL1、IL-23A、CD69、NF-κB p65、TNF-α、Fam89a、Grk5、phosopholipid scramblase 1、Runx1、semaphorin 4A、Steap4、lymphotoxin β Psmb10またはTLR2遺伝子の3’UTRに相補的な配列を含む内在性アンチセンス転写物に相補的な配列を含み、該遺伝子の発現を調節し得るセンスオリゴヌクレオチド。
〔6〕IL-1βにより誘導される遺伝子が、CCL2、CCL20、CX3CL1、IL-23A、CD69、NF-κB p65またはTNF-αである、上記〔5〕記載のセンスオリゴヌクレオチド。
〔7〕以下の(a)-(g)のいずれかである上記〔6〕記載のセンスオリゴヌクレオチド。
(a) 配列番号1に示されるCCL2 mRNAの3’UTR配列中、ヌクレオチド番号25-51で示されるドメイン1もしくはヌクレオチド番号103-175で示されるドメイン2、又は他の哺乳動物におけるオルソログのそれらに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列と90%以上の同一性を有するヌクレオチド配列を含んでなる、CCL2遺伝子の発現を調節し得るセンスオリゴヌクレオチド
(b) 配列番号2に示されるCCL20 mRNAの3’UTR配列中、ヌクレオチド番号65-107で示されるドメイン1、ヌクレオチド番号124-155で示されるドメイン2もしくはヌクレオチド番号214-282で示されるドメイン3、又は他の哺乳動物におけるオルソログのそれらに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列と90%以上の同一性を有するヌクレオチド配列を含んでなる、CCL20遺伝子の発現を調節し得るセンスオリゴヌクレオチド
(c) 配列番号3に示されるCX3CL1 mRNAの3’UTR配列中、ヌクレオチド番号1103-1202で示されるドメイン1、ヌクレオチド番号1305-1369で示されるドメイン2、ヌクレオチド番号1416-1482で示されるドメイン3、ヌクレオチド番号1633-1681で示されるドメイン4、ヌクレオチド番号1688-1745で示されるドメイン5、もしくはヌクレオチド番号1784-1810で示されるドメイン6、又は他の哺乳動物におけるオルソログのそれらに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列と90%以上の同一性を有するヌクレオチド配列を含んでなる、CX3CL1遺伝子の発現を調節し得るセンスオリゴヌクレオチド
(d) 配列番号4に示されるIL-23A mRNAの3’UTR配列中、ヌクレオチド番号37-55で示されるドメイン1、ヌクレオチド番号193-303で示されるドメイン2、ヌクレオチド番号380-448で示されるドメイン3、もしくはヌクレオチド番号555-600で示されるドメイン4、又は他の哺乳動物におけるオルソログのそれらに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列と90%以上の同一性を有するヌクレオチド配列を含んでなる、IL-23A遺伝子の発現を調節し得るセンスオリゴヌクレオチド
(e) 配列番号5に示されるCD69 mRNAの3’UTR配列中、ヌクレオチド番号15-32で示されるドメイン1、ヌクレオチド番号120-186で示されるドメイン2、ヌクレオチド番号218-252で示されるドメイン3、ヌクレオチド番号344-370で示されるドメイン4、ヌクレオチド番号389-633で示されるドメイン5、もしくはヌクレオチド番号656-786で示されるドメイン6、又は他の哺乳動物におけるオルソログのそれらに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列と90%以上の同一性を有するヌクレオチド配列を含んでなる、CD69遺伝子の発現を調節し得るセンスオリゴヌクレオチド
(f) 配列番号6に示されるNF-κB p65 mRNAの3’UTR配列中、ヌクレオチド番号161-301で示されるドメイン1、ヌクレオチド番号343-380で示されるドメイン2、ヌクレオチド番号401-412で示されるドメイン3、もしくはヌクレオチド番号473-523で示されるドメイン4、又は他の哺乳動物におけるオルソログのそれらに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列と90%以上の同一性を有するヌクレオチド配列を含んでなる、NF-κB p65遺伝子の発現を調節し得るセンスオリゴヌクレオチド
(g) 配列番号7に示されるTNF-α mRNAの3’UTR配列中、ヌクレオチド番号304-405で示されるドメイン1、ヌクレオチド番号430-546で示されるドメイン2、ヌクレオチド番号687-713で示されるドメイン3、もしくはヌクレオチド番号752-776で示されるドメイン4、又は他の哺乳動物におけるオルソログのそれらに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列と90%以上の同一性を有するヌクレオチド配列を含んでなる、TNF-α遺伝子の発現を調節し得るセンスオリゴヌクレオチド
〔8〕上記〔5〕記載のセンスオリゴヌクレオチドを含有してなる、CCL2、CCL20、CX3CL1、IL-23A、CD69、NF-κB p65、TNF-α、Fam89a、Grk5、phosopholipid scramblase 1、Runx1、semaphorin 4A、Steap4、lymphotoxin β Psmb10またはTLR2の発現調節剤。
〔9〕炎症性疾患または感染症の予防又は治療用である、上記〔8〕記載の剤。
〔10〕被験物質の存在下及び非存在下で、CCL2、CCL20、CX3CL1、IL-23A、CD69、NF-κB p65、TNF-α、Fam89a、Grk5、phosopholipid scramblase 1、Runx1、semaphorin 4A、Steap4、lymphotoxin β Psmb10またはTLR2遺伝子のmRNAとそれに対する内在性アンチセンス転写物とのハイブリダイゼーションを検出・比較することを特徴とする、該遺伝子の発現調節物質のスクリーニング方法。
〔11〕前記遺伝子のmRNAとそれに対する内在性アンチセンス転写物とを発現する細胞に被験物質を接触させ、該細胞における該mRNA量及び/又はそれにコードされるタンパク質量の変化を測定することを特徴とする、上記〔10〕記載の方法。
That is, the present invention is as follows.
[1] Induced by IL-1β, CCL2, CCL20, CX3CL1, IL-23A, CD69, NF-κB p65, TNF-α, Fam89a, Grk5, phosopholipid scramblase 1, Runx1, semaphorin 4A, Steap4, lymphphotoxin β Psmb10 Alternatively, an antisense nucleotide comprising a sequence complementary to the 3′UTR of the TLR2 gene and capable of regulating the expression of the gene.
[2] The antisense nucleotide according to [1] above, wherein the gene induced by IL-1β is CCL2, CCL20, CX3CL1, IL-23A, CD69, NF-κB p65 or TNF-α.
[3] CCL2, CCL20, CX3CL1, IL-23A, CD69, NF-κB p65, TNF-α, Fam89a, Grk5, phosopholipid scramblase 1, Runx1, semaphorin, comprising the antisense nucleotide described in [1] above 4A, Steap4, lymphphotoxin β Psmb10 or TLR2 expression regulator.
[4] The agent according to [3] above, which is used for prevention or treatment of inflammatory diseases or infectious diseases.
[5] Induced by IL-1β, CCL2, CCL20, CX3CL1, IL-23A, CD69, NF-κB p65, TNF-α, Fam89a, Grk5, phosopholipid scramblase 1, Runx1, semaphorin 4A, Steap4, lymphphotoxin β Psmb10 Alternatively, a sense oligonucleotide comprising a sequence complementary to an endogenous antisense transcript comprising a sequence complementary to the 3 ′ UTR of the TLR2 gene and capable of regulating expression of the gene.
[6] The sense oligonucleotide according to [5] above, wherein the gene induced by IL-1β is CCL2, CCL20, CX3CL1, IL-23A, CD69, NF-κB p65 or TNF-α.
[7] The sense oligonucleotide according to [6] above, which is any of the following (a) to (g):
(a) In the 3′UTR sequence of CCL2 mRNA represented by SEQ ID NO: 1, domain 1 represented by nucleotide numbers 25-51 or
(b) In the 3'UTR sequence of CCL20 mRNA represented by SEQ ID NO: 2, domain 1 represented by nucleotide numbers 65-107,
(c) In the 3′UTR sequence of CX3CL1 mRNA represented by SEQ ID NO: 3, domain 1 represented by nucleotide numbers 1103-1202,
(d) In the 3'UTR sequence of IL-23A mRNA shown in SEQ ID NO: 4, domain 1 shown by nucleotide numbers 37-55,
(e) In the 3'UTR sequence of CD69 mRNA represented by SEQ ID NO: 5, domain 1 represented by nucleotide numbers 15-32,
(f) In the 3′UTR sequence of NF-κB p65 mRNA represented by SEQ ID NO: 6, domain 1 represented by nucleotide numbers 161-301,
(g) In the 3′UTR sequence of TNF-α mRNA represented by SEQ ID NO: 7, domain 1 represented by nucleotide numbers 304-405,
[9] The agent according to [8] above, which is used for prevention or treatment of inflammatory diseases or infectious diseases.
[10] In the presence and absence of the test substance, CCL2, CCL20, CX3CL1, IL-23A, CD69, NF-κB p65, TNF-α, Fam89a, Grk5, phosopholipid scramblase 1, Runx1, semaphorin 4A, Steap4, A screening method for an expression regulator of the gene, comprising detecting and comparing the hybridization between the mRNA of the lymphotoxin β Psmb10 or TLR2 gene and the endogenous antisense transcript thereto.
[11] contacting a test substance with a cell expressing mRNA of the gene and an endogenous antisense transcript thereto, and measuring a change in the amount of the mRNA and / or protein encoded in the cell. The method according to [10] above, characterized in that it is characterized.
本発明のアンチセンスヌクレオチドは、IL-1βにより誘導される種々の遺伝子のmRNAに対するNATと同様に、該mRNAの3’UTRと相互作用することにより、該遺伝子の発現を調節することができる。一方、本発明のセンスオリゴヌクレオチドは、NATと相互作用することにより、その遺伝子発現調節作用を正もしくは負に調節して、標的遺伝子の発現を望ましいレベルに調節することができる。 The antisense nucleotide of the present invention can regulate the expression of the gene by interacting with the 3 'UTR of the mRNA in the same manner as NAT for mRNA of various genes induced by IL-1β. On the other hand, the sense oligonucleotide of the present invention can regulate the expression of a target gene to a desired level by interacting with NAT to regulate its gene expression regulation action positively or negatively.
本発明は、IL-1βにより誘導される、サイトカインやケモカインを含む種々の炎症/感染に関連する遺伝子の発現調節作用を有する、これら遺伝子のアンチセンスヌクレオチドを提供する。
本発明において標的となる遺伝子(mRNA)は、IL-1βにより誘導される遺伝子群の中で、その3’非翻訳領域(3’UTR)に相補的な配列を含む内在性アンチセンス転写物が存在する遺伝子であれば特に制限はないが、具体的には、例えば、chemokine (C-C motif) ligand 2 (CCL2); chemokine (C-C motif) ligand 20 (CCL20); chemokine (C-X3-C motif) ligand 1 (CX3CL1); interleukin-23 α-subunit p19 (IL-23A); cluster of differentiation 69 (CD69); nuclear factor-κB p65 subunit (NF-κB p65); tumor necrosis factor-α (TNF-α); family with sequence similarity 89 member A (Fam89a); G protein-coupled receptor kinase 5 (Grk5); phosopholipid scramblase 1; runt-related transcription factor 1 (Runx1); sema domain, immunoglobulin domain, transmembrane domain and short cytoplasmic domain 4A (semaphorin 4A); six-transmembrane epithelial antigen of prostate 4 (Steap4); lymphotoxin β; proteasome subunit, beta type 10 (Psmb10); Toll-like receptor 2 (TLR2)等が挙げられる。好ましくは、CCL2、CCL20、CX3CL1、IL-23A、CD69、NF-κB p65またはTNF-αである。
The present invention provides antisense nucleotides of these genes, which have an effect of regulating the expression of genes related to various inflammations / infections including cytokines and chemokines induced by IL-1β.
The target gene (mRNA) in the present invention is an endogenous antisense transcript containing a sequence complementary to its 3 ′ untranslated region (3′UTR) in the gene group induced by IL-1β. There is no particular limitation as long as it is a gene present, but specifically, for example, chemokine (CC motif) ligand 2 (CCL2); chemokine (CC motif) ligand 20 (CCL20); chemokine (C-X3-C motif) ligand 1 (CX3CL1); interleukin-23 α-subunit p19 (IL-23A); cluster of differentiation 69 (CD69); nuclear factor-κB p65 subunit (NF-κB p65); tumor necrosis factor-α (TNF-α) ; family with sequence similarity 89 member A (Fam89a); G protein-coupled receptor kinase 5 (Grk5); phosopholipid scramblase 1; runt-related transcription factor 1 (Runx1); sema domain, immunoglobulin domain, transmembrane domain and short cytoplasmic domain 4A (semaphorin 4A); six-transmembrane epithelial antigen of prostate 4 (Steap4); lymphotoxin β; proteasome subunit, beta type 10 (Psmb10); Toll-like recep and tor 2 (TLR2). CCL2, CCL20, CX3CL1, IL-23A, CD69, NF-κB p65 or TNF-α is preferable.
本発明のアンチセンスヌクレオチドは、標的mRNAの3’UTRに相補的な配列を含み、かつ標的遺伝子の発現(タンパク質の産生)を調節し得るヌクレオチドであれば、いかなるものであってもよい。ここで「相補的」な配列とは、mRNAに対して完全相補的な配列のみならず、細胞の生理的な条件下でmRNAとハイブリダイズしてmRNAと相互作用し得る限り、1ないし数(2, 3, 4 もしくは 5)塩基のミスマッチを含んでもよい。好ましくは、標的mRNAの3’UTRに相補的な配列とは、ストリンジェントな条件、例えば、Current Protocols in Molecular Biology, John Wiley & Sons,6.3.1-6.3.6, 1999に記載される条件(例えば、6×SSC(sodium chloride/sodium citrate)/45℃でのハイブリダイゼーション、次いで0.2×SSC/0.1% SDS/50-65℃での一回以上の洗浄等が挙げられる)下で、該mRNAとハイブリダイズし得る配列である。 The antisense nucleotide of the present invention may be any nucleotide as long as it contains a sequence complementary to the 3 'UTR of the target mRNA and can regulate the expression (protein production) of the target gene. Here, the “complementary” sequence is not only a sequence that is completely complementary to mRNA, but also 1 to a number (as long as it can hybridize with and interact with mRNA under physiological conditions of cells). 2, 3, 4 or 5) may contain base mismatches. Preferably, the sequence complementary to the 3′UTR of the target mRNA is a stringent condition such as those described in Current Protocols in Molecular Biology, John Wiley & Sons, 6.3.1-6.3.6, 1999 For example, hybridization at 6 × SSC (sodium chloride / sodium citrate) / 45 ° C., followed by one or more washings at 0.2 × SSC / 0.1% SDS / 50-65 ° C.) It is a sequence that can hybridize with.
具体的には、標的mRNAに対する内在アンチセンス転写物(NAT)は、該mRNAの熱力学的に安定でない部分と相互作用すると考えられるので、本発明のアンチセンスヌクレオチドは、標的mRNAの3’UTR中の熱力学的に安定でない部分に相補的な配列を含むことが好ましい。熱力学的に安定でない部分としては、mRNAが二次構造をとった際に一本鎖の状態にある(例えば、ステム-ループ構造のループ部分にあたる)領域が挙げられる。標的mRNAの3’UTRの二次構造は、該領域のヌクレオチド配列情報をもとに、mfold(GCG Software; Proc. Natl. Acad. Sci. USA, 86: 7706-10 (1989) 参照)に代表されるような既存のRNA二次構造予測プログラムを用いて予測することができる。それらのヌクレオチド配列情報はいずれも容易に入手可能である。例えば、CCL2、CCL20、CX3CL1、IL-23A、CD69、NF-κB p65およびTNF-α mRNAの3’UTR配列(但し、UはTに置き換えて記載している)を配列番号1-7にそれぞれ示す。
Specifically, since the endogenous antisense transcript (NAT) for a target mRNA is believed to interact with a thermodynamically unstable portion of the mRNA, the antisense nucleotides of the present invention may contain 3′UTR of the target mRNA. It is preferred to include a sequence complementary to a portion that is not thermodynamically stable therein. The portion that is not thermodynamically stable includes a region that is in a single-stranded state when the mRNA has a secondary structure (for example, a region corresponding to the loop portion of the stem-loop structure). The secondary structure of the 3'UTR of the target mRNA is represented by mfold (see GCG Software; Proc. Natl. Acad. Sci. USA, 86: 7706-10 (1989)) based on the nucleotide sequence information of the region. Can be predicted using an existing RNA secondary structure prediction program. Any of these nucleotide sequence information is readily available. For example, CCL2, CCL20, CX3CL1, IL-23A, CD69, NF-κB p65 and TNF-
好ましくは、本発明のCCL2に対するアンチセンスヌクレオチドは、配列番号1に示されるラットCCL2 mRNA 3’UTR配列にあっては、ヌクレオチド番号25-51で示されるドメイン1および/またはヌクレオチド番号103-175で示されるドメイン2内、他の哺乳動物におけるCCL2オルソログにあっては、前記ラットCCL2における各ドメインに対応するドメイン内の、1以上のループ構造の少なくとも一部(例えば、3塩基以上)を含む配列に相補的なヌクレオチド配列を含む。
Preferably, the antisense nucleotide against CCL2 of the present invention is the domain 1 and / or nucleotide number 103-175 represented by nucleotide number 25-51 in the rat CCL2 mRNA 3'UTR sequence represented by SEQ ID NO: 1. In the
好ましくは、本発明のCCL20に対するアンチセンスヌクレオチドは、配列番号2に示されるラットCCL20 mRNA 3’UTR配列にあっては、ヌクレオチド番号65-107で示されるドメイン1および/またはヌクレオチド番号124-155で示されるドメイン2および/またはヌクレオチド番号214-282で示されるドメイン3内、他の哺乳動物におけるCCL20オルソログにあっては、前記ラットCCL20における各ドメインに対応するドメイン内の、1以上のループ構造の少なくとも一部(例えば、3塩基以上)を含む配列に相補的なヌクレオチド配列を含む。
Preferably, the antisense nucleotide against CCL20 of the present invention, in the rat CCL20 ヌ ク レ オ チ ド mRNA 3'UTR sequence shown in SEQ ID NO: 2, is in domain 1 and / or nucleotide numbers 124-155 shown in nucleotide numbers 65-107 In the
好ましくは、本発明のCX3CL1に対するアンチセンスヌクレオチドは、配列番号3に示されるラットCX3CL1 mRNAの3’UTR配列にあっては、ヌクレオチド番号1103-1202で示されるドメイン1および/またはヌクレオチド番号1305-1369で示されるドメイン2および/またはヌクレオチド番号1416-1482で示されるドメイン3および/またはヌクレオチド番号1633-1681で示されるドメイン4および/またはヌクレオチド番号1688-1745で示されるドメイン5および/またはヌクレオチド番号1784-1810で示されるドメイン6内、他の哺乳動物におけるCX3CL1オルソログにあっては、前記ラットCX3CL1における各ドメインに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列に相補的なヌクレオチド配列を含む。
Preferably, the antisense nucleotide against CX3CL1 of the present invention is the domain 1 and / or nucleotide number 1305-1369 represented by nucleotide number 1103-1202 in the 3′UTR sequence of rat CX3CL1 mRNA represented by SEQ ID NO: 3.
好ましくは、本発明のIL-23Aに対するアンチセンスヌクレオチドは、配列番号4に示されるラットIL-23A mRNAの3’UTR配列にあっては、ヌクレオチド番号37-55で示されるドメイン1および/またはヌクレオチド番号193-303で示されるドメイン2および/またはヌクレオチド番号380-448で示されるドメイン3および/またはヌクレオチド番号555-600で示されるドメイン4内、他の哺乳動物におけるIL-23Aオルソログにあっては、前記ラットIL-23Aの各ドメインに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列に相補的なヌクレオチド配列を含む。
Preferably, the antisense nucleotide against IL-23A of the present invention is the domain 1 and / or nucleotide represented by nucleotide number 37-55 in the 3′UTR sequence of rat IL-23A mRNA represented by SEQ ID NO: 4 In the
好ましくは、本発明のCD69に対するアンチセンスヌクレオチドは、配列番号5に示されるラットCD69 mRNAの3’UTR配列にあっては、ヌクレオチド番号15-32で示されるドメイン1および/またはヌクレオチド番号120-186で示されるドメイン2および/またはヌクレオチド番号218-252で示されるドメイン3および/またはヌクレオチド番号344-370で示されるドメイン4および/またはヌクレオチド番号389-633で示されるドメイン5および/またはヌクレオチド番号656-786で示されるドメイン6内、他の哺乳動物におけるCD69オルソログにあっては、前記ラットCD69における各ドメインに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列に相補的なヌクレオチド配列を含む。
Preferably, the antisense nucleotide against CD69 of the present invention is the domain 1 and / or nucleotide number 120-186 represented by nucleotide number 15-32 in the 3′UTR sequence of rat CD69 mRNA represented by SEQ ID NO: 5. 2 and / or
好ましくは、本発明のNF-κB p65に対するアンチセンスヌクレオチドは、配列番号6に示されるラットNF-κB p65 mRNAの3’UTR配列にあっては、ヌクレオチド番号161-301で示されるドメイン1および/またはヌクレオチド番号343-380で示されるドメイン2および/またはヌクレオチド番号401-412で示されるドメイン3および/またはヌクレオチド番号473-523で示されるドメイン4内、他の哺乳動物におけるNF-κB p65オルソログにあっては、前記ラットNF-κB p65における各ドメインに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列に相補的なヌクレオチド配列を含む。
Preferably, the antisense nucleotide for NF-κB p65 of the present invention is the domain 1 and / or the nucleotides 161-301 in the 3′UTR sequence of rat NF-κB p65 mRNA shown in SEQ ID NO: 6. Or in
好ましくは、本発明のTNF-αに対するアンチセンスヌクレオチドは、配列番号7に示されるラットTNF-α mRNAの3’UTR配列にあっては、ヌクレオチド番号304-405で示されるドメイン1および/またはヌクレオチド番号430-546で示されるドメイン2および/またはヌクレオチド番号687-713で示されるドメイン3および/またはヌクレオチド番号752-776で示されるドメイン4内、他の哺乳動物におけるTNF-αオルソログにあっては、前記ラットTNF-αにおける各ドメインに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列に相補的なヌクレオチド配列を含む。
Preferably, the antisense nucleotide against TNF-α of the present invention is the domain 1 and / or nucleotide represented by nucleotide number 304-405 in the 3′UTR sequence of rat TNF-α mRNA represented by SEQ ID NO: 7 In the
ラット以外の哺乳動物におけるオルソログの対応するドメインとしては、3’UTRにAREモチーフを有する遺伝子の場合、複数種に共通なAREモチーフの周辺領域のヌクレオチド配列は哺乳動物種間でよく保存されており、二次構造も類似しているため、当該領域が好ましく選択され得る。例えば、ラット、マウスおよびヒトの上記7遺伝子の3’UTRに存在するAREモチーフ並びにヒト-ラット間および3種間で共通するAREモチーフの数を表1に示す。 As for the corresponding domain of orthologs in mammals other than rats, in the case of a gene having an ARE motif in 3'UTR, the nucleotide sequence of the peripheral region of the ARE motif common to multiple species is well conserved among mammalian species. Since the secondary structure is similar, the region can be preferably selected. For example, Table 1 shows the number of ARE motifs present in the 3'UTR of the above-mentioned 7 genes of rat, mouse and human and the common ARE motifs between human-rat and among three species.
本発明のアンチセンスヌクレオチドの長さに特に制限はなく、NATの全長もしくは標的mRNAの3’UTR中の前記ドメイン内の1以上のループ構造の少なくとも一部を含む配列に相補的なヌクレオチド配列を含むNATの部分配列を含むものであってよいが、配列特異性の面から、標的配列に相補的な部分を少なくとも10塩基以上、好ましくは約12塩基以上、より好ましくは約15塩基以上含むものである。また、医薬として用いる場合の投与のし易さ等の面から、500塩基以下、好ましくは300塩基以下、より好ましくは150塩基以下の塩基長を有するものが挙げられる。 The length of the antisense nucleotide of the present invention is not particularly limited, and a nucleotide sequence complementary to a sequence comprising at least a part of one or more loop structures in the domain in the full length of NAT or 3′UTR of the target mRNA. It may contain a partial sequence of NAT, but from the viewpoint of sequence specificity, it contains at least 10 bases, preferably about 12 bases or more, more preferably about 15 bases or more, a portion complementary to the target sequence. . In addition, from the viewpoint of ease of administration when used as a pharmaceutical, those having a base length of 500 bases or less, preferably 300 bases or less, more preferably 150 bases or less are mentioned.
さらに、本発明のアンチセンスヌクレオチドが10~50塩基程度のオリゴヌクレオチドの場合、該ヌクレオチドは、配列非特異的な反応を起こす配列(例えば、5’-CG-3’、5’-GGGG-3’、5’-GGGGG-3’等)を含まないものから選択することが好ましく、また、標的mRNA以外のRNA中に類似の相補鎖配列が存在しないものから選択することが好ましい。他のRNA中に類似の相補鎖配列が存在しないことは、アンチセンスオリゴヌクレオチドの候補配列をクエリーとして、対象とする哺乳動物のゲノム配列に対して相同性検索をかけることにより確認することができる。ここで相同性検索手段としては、公知の核酸の相同性検索ソフトウェア(例えば、NCBI BLAST(National Center for Biotechnology Information Basic Local Alignment Search Tool)NBLAST及びXBLASTプログラム(version 2.0)、GCGソフトウェアパッケージ中のFASTAプログラム等)を用いることができる。また、ゲノムDNAデータセットとしては、例えば、Celera社が提供する全ヒトゲノムデータを用いることができる。 Furthermore, when the antisense nucleotide of the present invention is an oligonucleotide having about 10 to 50 bases, the nucleotide is a sequence that causes a sequence-nonspecific reaction (for example, 5′-CG-3 ′, 5′-GGGG-3 ', 5'-GGGGG-3' and the like are preferably selected, and are preferably selected from those having no similar complementary strand sequence in RNA other than the target mRNA. The absence of a similar complementary strand sequence in other RNAs can be confirmed by performing a homology search against the target mammalian genome sequence using the antisense oligonucleotide candidate sequence as a query. . Here, as homology search means, known nucleic acid homology search software (for example, NCBI BLAST (National Center Biotechnology Information Basic Basic Local Alignment Search Tool) NBLAST and XBLAST programs (version 2.0), FASTA program in GCG software package Etc.) can be used. Moreover, as a genomic DNA data set, for example, all human genome data provided by Celera can be used.
本発明のアンチセンスヌクレオチドは、細胞への導入方法に応じて種々の形態で用いられる。例えば、該アンチセンスヌクレオチドが10~50塩基程度のオリゴヌクレオチドの場合、一本鎖DNA、一本鎖RNA、DNA/RNAキメラのいずれであってもよく、さらに公知の修飾の付加されたものであってもよい。ここで「ヌクレオチド」とは、プリン及びピリミジン塩基を含有するのみでなく、修飾されたその他の複素環型塩基をもつようなものを含んでいてもよい。 The antisense nucleotide of the present invention is used in various forms depending on the method of introduction into cells. For example, when the antisense nucleotide is an oligonucleotide having about 10 to 50 bases, it may be any one of single-stranded DNA, single-stranded RNA, and DNA / RNA chimera, and it is further added with known modifications. There may be. Here, the “nucleotide” may include not only purine and pyrimidine bases but also those having other modified heterocyclic bases.
アンチセンスヌクレオチドを構成するヌクレオチド分子は、天然型のDNAもしくはRNAでもよいが、安定性(化学的及び/又は対酵素)や比活性(RNAとの親和性)を向上させるために、種々の化学修飾を含むことができる。例えば、ヌクレアーゼなどによる分解を防ぐために、アンチセンスオリゴヌクレオチドを構成する各ヌクレオチドのリン酸残基(ホスフェート)を、例えば、ホスホロチオエート(PS)、メチルホスホネート、ホスホロジチオネートなどの化学修飾リン酸残基に置換することができる。また、各ヌクレオチドの糖(リボース)の2'位の水酸基を、-OR(Rは、例えばCH3(2'-O-Me)、CH2CH2OCH3(2'-O-MOE)、CH2CH2NHC(NH)NH2、CH2CONHCH3、CH2CH2CN等を示す)に置換してもよい。さらに、塩基部分(ピリミジン、プリン)に化学修飾を施してもよく、例えば、ピリミジン塩基の5位へのメチル基やカチオン性官能基の導入、あるいは2位のカルボニル基のチオカルボニルへの置換などが挙げられる。
RNAの糖部のコンフォーメーションはC2'-endo(S型)とC3'-endo(N型)の2つが支配的であり、一本鎖RNAではこの両者の平衡として存在するが、二本鎖を形成するとN型に固定される。したがって、標的mRNAに対して強い結合能を付与するために、2'酸素と4’炭素を架橋することにより、糖部のコンフォーメーションをN型に固定したRNA誘導体であるBNA (LNA)(Imanishi, T. et al., Chem. Commun., 1653-9, 2002; Jepsen, J.S. et al., Oligonucleotides, 14, 130-46, 2004)やENA(Morita, K. et al., Nucleosides Nucleotides Nucleic Acids, 22, 1619-21, 2003)もまた、好ましく用いられ得る。
The nucleotide molecule constituting the antisense nucleotide may be natural DNA or RNA, but various chemicals may be used to improve stability (chemical and / or enzyme) and specific activity (affinity with RNA). Modifications can be included. For example, in order to prevent degradation by nuclease, etc., phosphate residues (phosphates) of each nucleotide constituting the antisense oligonucleotide may be changed to chemically modified phosphate residues such as phosphorothioate (PS), methylphosphonate, phosphorodithionate, etc. Substituents can be substituted. In addition, the 2′-position hydroxyl group of the sugar (ribose) of each nucleotide is represented by —OR (R is, for example, CH 3 (2′-O-Me), CH 2 CH 2 OCH 3 (2′-O-MOE), CH 2 CH 2 NHC (NH) NH 2 , CH 2 CONHCH 3 , CH 2 CH 2 CN and the like may be substituted). Furthermore, the base moiety (pyrimidine, purine) may be chemically modified, for example, introduction of a methyl group or a cationic functional group at the 5-position of the pyrimidine base, or substitution of the carbonyl group at the 2-position with thiocarbonyl. Is mentioned.
The conformation of the sugar part of RNA is dominated by C2'-endo (S type) and C3'-endo (N type). In single-stranded RNA, it exists as an equilibrium between the two, but double-stranded Is fixed to the N type. Therefore, BNA (LNA) (Imanishi) is an RNA derivative in which the conformation of the sugar moiety is fixed to the N-type by bridging the 2 'oxygen and 4' carbon to give strong binding ability to the target mRNA. , T. et al., Chem. Commun., 1653-9, 2002; Jepsen, JS et al., Oligonucleotides, 14, 130-46, 2004) and ENA (Morita, K. et al., Nucleosides Nucleotides Nucleicides Nucleic Acids , 22, 1619-21, 2003) can also be preferably used.
本発明のアンチセンスオリゴヌクレオチドは、標的mRNA(cDNA)配列に基づいて、市販のDNA/RNA自動合成機(アプライド・バイオシステムズ社、ベックマン社等)を用いて、これに相補的な配列を合成することにより調製することができる。 The antisense oligonucleotide of the present invention synthesizes a complementary sequence based on the target mRNA (cDNA) sequence using a commercially available DNA / RNA automatic synthesizer (Applied Biosystems, Beckman, etc.). Can be prepared.
本発明のアンチセンスオリゴヌクレオチドは、リポソーム、ミクロスフェアのような特殊な形態で供与されたり、ポリリジンのようなポリカチオン体、脂質(例、ホスホリピド、コレステロールなど)などの疎水性物質が付加された形態で提供され得る。あるいはまた、本発明のアンチセンスオリゴヌクレオチドを、膜透過機能を有するペプチド(例えば、ショウジョウバエ由来のAntennapediaホメオドメイン(AntP)、ヒト免疫不全ウイルス(HIV)由来のTAT、単純ヘルペスウイルス(HSV)由来のVP22等の細胞通過ドメイン)などで修飾することにより、該オリゴヌクレオチドの細胞への取り込みを促進することができる。 The antisense oligonucleotide of the present invention is provided in a special form such as a liposome or a microsphere, or a hydrophobic substance such as a polycationic substance such as polylysine or a lipid (eg, phospholipid, cholesterol, etc.) is added thereto. It can be provided in the form. Alternatively, the antisense oligonucleotide of the present invention is converted to a peptide having a membrane permeation function (for example, Drosophila-derived Antennapedia homeodomain (AntP), human immunodeficiency virus (HIV) -derived TAT, herpes simplex virus (HSV) -derived Modification with a cell-passing domain such as VP22) can promote the uptake of the oligonucleotide into cells.
一方、本発明のアンチセンスヌクレオチドがより長い塩基長からなるポリヌクレオチドの場合、該ヌクレオチドの細胞への導入は、自体公知の遺伝子導入法を用いて実施することができる。この場合、該ヌクレオチドとしては二本鎖DNAが好ましく用いられる。本発明のアンチセンスヌクレオチドは、例えば、標的遺伝子のNATを発現する細胞(例えば、IL-1β刺激により標的mRNAおよびNATを高発現する細胞)から総RNAを抽出し、例えば、配列番号1-7に示される標的mRNAの配列情報に基づいて、その相補鎖配列の適当な領域を増幅し得るプライマーを設計し、RT-PCRを実施することにより取得することができる(後記実施例参照)。得られたcDNAは、宿主となる動物細胞で機能し得るプロモーターを含む適当な発現ベクターに挿入される。発現ベクターとしては、例えば、レトロウイルス、レンチウイルス、アデノウイルス、アデノ随伴ウイルス、ヘルペスウイルス、センダイウイルスなどのウイルスベクター、動物細胞発現プラスミド(例、pA1-11,pXT1,pRc/CMV,pRc/RSV,pcDNAI/Neo)などが用いられ得る。
発現ベクターにおいて使用されるプロモーターとしては、例えばEF1αプロモーター、CAGプロモーター、SRαプロモーター、SV40プロモーター、LTRプロモーター、CMV(サイトメガロウイルス)プロモーター、RSV(ラウス肉腫ウイルス)プロモーター、MoMuLV(モロニーマウス白血病ウイルス)LTR、HSV-TK(単純ヘルペスウイルスチミジンキナーゼ)プロモーターなどが用いられる。なかでも、EF1αプロモーター、CAGプロモーター、MoMuLV LTR、CMVプロモーター、SRαプロモーターなどが好ましい。
発現ベクターは、プロモーターの他に、所望によりエンハンサー、ポリA付加シグナル、選択マーカー遺伝子、SV40複製起点などを含有していてもよい。選択マーカー遺伝子としては、例えば、ジヒドロ葉酸還元酵素遺伝子、ネオマイシン耐性遺伝子、ピューロマイシン耐性遺伝子等が挙げられる。
On the other hand, when the antisense nucleotide of the present invention is a polynucleotide having a longer base length, introduction of the nucleotide into a cell can be carried out using a gene transfer method known per se. In this case, double-stranded DNA is preferably used as the nucleotide. The antisense nucleotide of the present invention extracts, for example, total RNA from a cell that expresses the target gene NAT (for example, a cell that highly expresses target mRNA and NAT by IL-1β stimulation). For example, SEQ ID NOs: 1-7 Based on the sequence information of the target mRNA shown in the above, a primer that can amplify an appropriate region of the complementary strand sequence is designed, and RT-PCR is performed (see Examples below). The obtained cDNA is inserted into an appropriate expression vector containing a promoter that can function in host animal cells. Examples of expression vectors include retroviruses, lentiviruses, adenoviruses, adeno-associated viruses, herpes viruses, Sendai virus and other viral vectors, animal cell expression plasmids (eg, pA1-11, pXT1, pRc / CMV, pRc / RSV). , PcDNAI / Neo) or the like.
Examples of the promoter used in the expression vector include EF1α promoter, CAG promoter, SRα promoter, SV40 promoter, LTR promoter, CMV (cytomegalovirus) promoter, RSV (rous sarcoma virus) promoter, MoMuLV (Molone murine leukemia virus) LTR. HSV-TK (herpes simplex virus thymidine kinase) promoter and the like are used. Of these, EF1α promoter, CAG promoter, MoMuLV LTR, CMV promoter, SRα promoter and the like are preferable.
In addition to the promoter, the expression vector may optionally contain an enhancer, a poly A addition signal, a selection marker gene, an SV40 replication origin, and the like. Examples of the selection marker gene include a dihydrofolate reductase gene, a neomycin resistance gene, a puromycin resistance gene, and the like.
本発明のアンチセンスヌクレオチドは、標的mRNAと相互作用することにより、標的遺伝子の発現(タンパク質の産生)を調節することができる。IL-1βにより誘導される、サイトカイン・ケモカイン遺伝子を含む種々の遺伝子は、炎症惹起もしくは抗炎症作用、感染防御、免疫応答の調節などの多様な生理活性を有するので、本発明のアンチセンスヌクレオチドを含有する医薬は、標的遺伝子の発現調節剤として、炎症性疾患や感染症を含む種々の疾患の予防及び/又は治療に利用することができる。
例えば、CCL2の発現を増強させ得るアンチセンスヌクレオチドは血管新生や創傷治癒などに利用することができ、一方、CCL2は単球や好酸球を遊走するため、CCL2の発現を抑制し得るアンチセンスヌクレオチドは、動脈硬化を含む慢性炎症性疾患やアレルギー性炎症の予防・治療に有用である。
CCL20は広範囲の微生物に対して抗菌活性を示すので、CCL20の発現を増強させ得るアンチセンスヌクレオチドは種々の微生物感染症の予防・治療に有効である。一方、CCL20は適応免疫の開始と持続に重要な役割を担っていることから、CCL20の発現を抑制し得るアンチセンスヌクレオチドは、気管支喘息、多発性硬化症、関節リウマチ、各種皮膚炎、炎症性腸疾患などの自己免疫疾患の予防・治療に有効である。
CX3CL1の発現を増強させ得るアンチセンスヌクレオチドは血管新生などに利用することができ、一方、CX3CL1の発現を抑制し得るアンチセンスヌクレオチドは、動脈硬化、糸球体腎炎を含む炎症性疾患の予防・治療や拒絶抑制、関節リウマチの予防・治療に有用である。
IL-23Aは細胞性免疫の維持に重要であり、その過剰発現は多臓器炎症を引き起こすことから、IL-23Aの発現を増強させ得るアンチセンスヌクレオチドは感染症の予防・治療に有効であり、一方、IL-23Aの発現を抑制し得るアンチセンスヌクレオチドは、自己免疫疾患や炎症性疾患の予防・治療に有用である。
CD69はリンパ球の分化や活性化に重要な役割を果たしていることから、CD69の発現を増強させ得るアンチセンスヌクレオチドは感染症の予防・治療に有効である。一方、CD69は炎症局所に発現する炎症細胞のほとんどで発現し、また関節リウマチや全身性エリテマトーデス(SLE)患者からCD69に対する自己抗体が検出されることなどから、CD69の発現を抑制し得るアンチセンスヌクレオチドは、自己免疫疾患や炎症性疾患の予防・治療に有用である。
NF-κBは免疫系に重要な役割を持つ転写因子であることから、NF-κB p65の発現を増強させ得るアンチセンスヌクレオチドは感染症の予防・治療に有効である。一方、NF-κBはがん細胞で恒常的に活性化してアポトーシスを阻害しており、また骨代謝や気管支喘息、関節炎、炎症性腸疾患、敗血症などの病態形成にも関与しているので、NF-κB p65の発現を抑制し得るアンチセンスヌクレオチドは、がん、骨粗鬆症、自己免疫疾患や炎症性疾患の予防・治療に有用である。
TNF-αはアポトーシス誘導、炎症メディエーターや形質細胞による抗体産生の亢進などを通じて感染防御や抗腫瘍作用を示すので、TNF-αの発現を増強させ得るアンチセンスヌクレオチドは感染症やがんの予防・治療に有効である。一方、TNF-αは関節リウマチ、乾癬、糖尿病、高脂血症、敗血症、骨粗鬆症の病態形成に関与するため、TNF-αの発現を抑制し得るアンチセンスヌクレオチドは、これらの疾患の予防・治療に有用である。
The antisense nucleotide of the present invention can regulate target gene expression (protein production) by interacting with the target mRNA. Various genes including cytokine / chemokine gene induced by IL-1β have various physiological activities such as inflammation-inducing or anti-inflammatory action, infection defense, regulation of immune response, etc. The contained drug can be used as a target gene expression regulator for the prevention and / or treatment of various diseases including inflammatory diseases and infectious diseases.
For example, antisense nucleotides that can enhance the expression of CCL2 can be used for angiogenesis and wound healing, while CCL2 migrates to monocytes and eosinophils, so that it can suppress the expression of CCL2. Nucleotides are useful for the prevention and treatment of chronic inflammatory diseases including arteriosclerosis and allergic inflammation.
Since CCL20 exhibits antibacterial activity against a wide range of microorganisms, antisense nucleotides that can enhance the expression of CCL20 are effective in preventing and treating various microbial infections. On the other hand, since CCL20 plays an important role in the initiation and maintenance of adaptive immunity, antisense nucleotides that can suppress the expression of CCL20 are bronchial asthma, multiple sclerosis, rheumatoid arthritis, various dermatitis, inflammatory It is effective for the prevention and treatment of autoimmune diseases such as bowel disease.
Antisense nucleotides that can enhance CX3CL1 expression can be used for angiogenesis, while antisense nucleotides that can suppress CX3CL1 expression prevent or treat inflammatory diseases including arteriosclerosis and glomerulonephritis It is useful for the prevention and treatment of anti-rejection and rheumatoid arthritis.
IL-23A is important for the maintenance of cellular immunity, and its overexpression causes multi-organ inflammation. Therefore, antisense nucleotides that can enhance IL-23A expression are effective in the prevention and treatment of infectious diseases. On the other hand, antisense nucleotides that can suppress the expression of IL-23A are useful for the prevention and treatment of autoimmune diseases and inflammatory diseases.
Since CD69 plays an important role in the differentiation and activation of lymphocytes, antisense nucleotides that can enhance the expression of CD69 are effective in the prevention and treatment of infectious diseases. On the other hand, CD69 is expressed in most inflammatory cells that are expressed locally, and an anti-antibody that can suppress the expression of CD69 due to detection of autoantibodies against CD69 from patients with rheumatoid arthritis and systemic lupus erythematosus (SLE). Nucleotides are useful for the prevention and treatment of autoimmune diseases and inflammatory diseases.
Since NF-κB is a transcription factor having an important role in the immune system, antisense nucleotides that can enhance the expression of NF-κB p65 are effective in preventing and treating infectious diseases. On the other hand, NF-κB is constantly activated in cancer cells and inhibits apoptosis, and is also involved in pathogenesis such as bone metabolism, bronchial asthma, arthritis, inflammatory bowel disease, sepsis, Antisense nucleotides that can suppress the expression of NF-κB p65 are useful for the prevention and treatment of cancer, osteoporosis, autoimmune diseases and inflammatory diseases.
Since TNF-α shows protection against infection and anti-tumor action by inducing apoptosis and enhancing antibody production by inflammatory mediators and plasma cells, antisense nucleotides that can enhance the expression of TNF-α prevent infection and cancer. It is effective for treatment. On the other hand, since TNF-α is involved in the pathogenesis of rheumatoid arthritis, psoriasis, diabetes, hyperlipidemia, sepsis, and osteoporosis, antisense nucleotides that can suppress the expression of TNF-α are used to prevent and treat these diseases. Useful for.
本発明のアンチセンスヌクレオチドを含有する医薬は低毒性であり、そのまま液剤として、又は適当な剤型の医薬組成物として、ヒト又は非ヒト哺乳動物(例、マウス、ラット、モルモット、ウサギ、ヒツジ、ブタ、ウシ、ネコ、イヌ、サルなど)に対して経口的又は非経口的(例、吸入投与、血管内投与、皮下投与、経粘膜投与など)に投与することができる。
これらの核酸を上記の各種疾患の予防・治療剤として使用する場合、自体公知の方法に従って製剤化し、投与することができる。即ち、本発明のアンチセンスヌクレオチドを、単独で、あるいはレトロウイルスベクター、レンチウイルスベクター、アデノウイルスベクター、アデノ随伴ウイルスベクターなどの上記の適当な哺乳動物細胞用の発現ベクターに機能可能な態様で挿入した後、常套手段に従って製剤化することができる。該ヌクレオチドは、そのままで、あるいは摂取促進のための補助剤とともに、遺伝子銃やハイドロゲルカテーテルのようなカテーテルによって投与することもできる。あるいは、エアロゾル化して吸入剤として気管内に局所投与することもできる。
さらに、体内動態の改良、半減期の長期化、細胞内取り込み効率の改善を目的に、前記ヌクレオチドを単独又はリポソームなどの担体とともに製剤(注射剤)化し、静脈、皮下等に投与してもよい。
The medicament containing the antisense nucleotide of the present invention has low toxicity, and as such a solution or a pharmaceutical composition of an appropriate dosage form, can be used as a human or non-human mammal (eg, mouse, rat, guinea pig, rabbit, sheep, Pigs, cows, cats, dogs, monkeys, etc.) can be administered orally or parenterally (eg, inhalation administration, intravascular administration, subcutaneous administration, transmucosal administration, etc.).
When these nucleic acids are used as a prophylactic / therapeutic agent for the above-mentioned various diseases, they can be formulated and administered according to a method known per se. That is, the antisense nucleotide of the present invention is inserted alone or in a functional manner into the appropriate expression vector for mammalian cells such as a retroviral vector, lentiviral vector, adenoviral vector, adeno-associated viral vector and the like. Then, it can be formulated according to conventional means. The nucleotide can be administered as it is or together with an auxiliary agent for promoting intake by a gene gun or a catheter such as a hydrogel catheter. Alternatively, it can be aerosolized and locally administered into the trachea as an inhalant.
Furthermore, for the purpose of improving pharmacokinetics, extending the half-life, and improving cellular uptake efficiency, the nucleotide may be formulated (injection) alone or with a carrier such as a liposome and administered intravenously, subcutaneously, etc. .
本発明のアンチセンスヌクレオチドは、それ自体を投与してもよいし、又は適当な医薬組成物として投与してもよい。投与に用いられる医薬組成物としては、本発明のアンチセンスヌクレオチドと薬理学的に許容され得る担体、希釈剤もしくは賦形剤とを含むものであってよい。このような医薬組成物は、経口又は非経口投与に適する剤形として提供される。 The antisense nucleotide of the present invention may be administered per se or as an appropriate pharmaceutical composition. The pharmaceutical composition used for administration may contain the antisense nucleotide of the present invention and a pharmacologically acceptable carrier, diluent or excipient. Such a pharmaceutical composition is provided as a dosage form suitable for oral or parenteral administration.
非経口投与のための組成物としては、例えば、注射剤、エアロゾル剤、坐剤等が用いられ、注射剤は静脈注射剤、皮下注射剤、皮内注射剤、筋肉注射剤、点滴注射剤等の剤形を包含してもよい。このような注射剤は、公知の方法に従って調製できる。エアロゾル製剤はジクロロジフロロメタン、プロパン、窒素などのような圧縮された許容しうる抛射薬内に入れることができる。あるいはネブライザーやアトマイザーのような非圧縮性製剤用医薬品として製剤化してもよい。直腸投与に用いられる坐剤は、上記核酸を通常の坐薬用基剤に混合することによって調製されてもよい。 As a composition for parenteral administration, for example, injections, aerosols, suppositories and the like are used, and injections are intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections, intravenous injections, etc. May be included. Such an injection can be prepared according to a known method. Aerosol formulations can be placed in compressed acceptable propellants such as dichlorodifluoromethane, propane, nitrogen and the like. Alternatively, it may be formulated as an incompressible pharmaceutical product such as a nebulizer or an atomizer. Suppositories used for rectal administration may be prepared by mixing the nucleic acid with a normal suppository base.
経口投与のための組成物としては、固体又は液体の剤形、具体的には錠剤(糖衣錠、フィルムコーティング錠を含む)、丸剤、顆粒剤、散剤、カプセル剤(ソフトカプセル剤を含む)、シロップ剤、乳剤、懸濁剤等が挙げられる。このような組成物は公知の方法によって製造され、製剤分野において通常用いられる担体、希釈剤もしくは賦形剤を含有していても良い。錠剤用の担体、賦形剤としては、例えば、乳糖、でんぷん、蔗糖、ステアリン酸マグネシウムが用いられる。 Compositions for oral administration include solid or liquid dosage forms, specifically tablets (including sugar-coated tablets and film-coated tablets), pills, granules, powders, capsules (including soft capsules), and syrups. Agents, emulsions, suspensions and the like. Such a composition is produced by a known method and may contain a carrier, a diluent or an excipient usually used in the pharmaceutical field. As the carrier and excipient for tablets, for example, lactose, starch, sucrose, and magnesium stearate are used.
上記の非経口用又は経口用医薬組成物は、活性成分の投与量に適合するような投薬単位の剤形に調製されることが好都合である。このような投薬単位の剤形としては、例えば、錠剤、丸剤、カプセル剤、注射剤(アンプル)、エアロゾル剤、坐剤が挙げられる。本発明のアンチセンスヌクレオチドは、例えば、投薬単位剤形当たり通常5~500mg、とりわけ注射剤では5~100mg、その他の剤形では10~250mg含有されていることが好ましい。 The above parenteral or oral pharmaceutical composition is conveniently prepared in a dosage unit form suitable for the dosage of the active ingredient. Examples of such dosage forms include tablets, pills, capsules, injections (ampoules), aerosols, and suppositories. The antisense nucleotide of the present invention is preferably contained, for example, usually 5 to 500 mg per dosage unit dosage form, particularly 5 to 100 mg for injections, and 10 to 250 mg for other dosage forms.
本発明のアンチセンスヌクレオチドを含有する上記医薬の投与量は、投与対象、対象疾患、症状、投与ルートなどによっても異なるが、例えば、本発明のアンチセンスヌクレオチドを1回量として、通常0.01~20mg/kg体重程度、好ましくは0.1~10mg/kg体重程度、さらに好ましくは0.1~5mg/kg体重程度を、1日1回~数回程度、静脈注射や吸入等により投与するのが好都合である。他の非経口投与及び経口投与の場合もこれに準ずる量を投与することができる。症状が特に重い場合には、その症状に応じて増量してもよい。 The dosage of the above-mentioned medicament containing the antisense nucleotide of the present invention varies depending on the administration subject, target disease, symptom, administration route, etc., but for example, the dosage of the antisense nucleotide of the present invention is usually 0.01 to 20 mg. It is convenient to administer about 0.1 kg / kg body weight, preferably about 0.1-10 mg / kg body weight, more preferably about 0.1-5 mg / kg body weight by intravenous injection or inhalation once to several times a day. In the case of other parenteral administration and oral administration, an equivalent amount can be administered. If symptoms are particularly severe, the dose may be increased according to the symptoms.
なお前記した医薬組成物は、本発明のアンチセンスヌクレオチドとの配合により好ましくない相互作用を生じない限り、他の薬剤を含有してもよい。他の薬剤としては、例えば、抗ウイルス薬、抗腫瘍薬、抗菌薬、抗真菌薬、抗原虫薬、抗生物質、抗セプシス薬、抗セプティックショック薬、エンドトキシン拮抗薬、免疫調節薬、非ステロイド性抗炎症薬、ステロイド薬、炎症性メディエーター作用抑制薬、炎症性メディエーター産生抑制薬、抗炎症性メディエーター作用抑制薬、抗炎症性メディエーター産生抑制薬などが挙げられる。 Note that the above-described pharmaceutical composition may contain other drugs as long as an undesirable interaction is not caused by the combination with the antisense nucleotide of the present invention. Other drugs include, for example, antiviral drugs, antitumor drugs, antibacterial drugs, antifungal drugs, antiprotozoal drugs, antibiotics, antiseptic drugs, antiseptic shock drugs, endotoxin antagonists, immunomodulators, non-steroidal drugs Examples include anti-inflammatory drugs, steroid drugs, inflammatory mediator action inhibitors, inflammatory mediator production inhibitors, anti-inflammatory mediator action inhibitors, anti-inflammatory mediator production inhibitors.
内在性アンチセンス転写物(NAT)のヌクレオチド配列に相補的なヌクレオチド配列を含むオリゴヌクレオチド(即ち、NAT転写される遺伝子のセンスオリゴヌクレオチド)は、標的mRNAに対するNATの発現調節作用を阻害することにより、標的mRNAの安定性を調節し、標的遺伝子の発現(タンパク質の産生)を正もしくは負に調節することができる。したがって、本発明はまた、IL-1βにより誘導される、NATが存在する遺伝子の発現調節活性を有する当該遺伝子の3’UTRと相同なセンスオリゴヌクレオチドを提供する。ここで標的となる遺伝子(mRNA)は本発明のアンチセンスヌクレオチドに関して前記したのと同様である。 Oligonucleotides that contain a nucleotide sequence that is complementary to the nucleotide sequence of the endogenous antisense transcript (NAT) (ie, the sense oligonucleotide of the NAT transcribed gene) inhibits NAT's regulation of expression on the target mRNA. It is possible to regulate the stability of the target mRNA and regulate the expression of the target gene (protein production) positively or negatively. Therefore, the present invention also provides a sense oligonucleotide homologous to the 3 'UTR of a gene having an expression-regulating activity of a gene in which NAT is induced, induced by IL-1β. Here, the target gene (mRNA) is the same as described above for the antisense nucleotide of the present invention.
本発明のセンスオリゴヌクレオチドは、標的mRNAの3’UTRに相補的な配列を含むNATに相補的な配列(従って、該3’UTRに相同な配列)を含み、かつ標的mRNAの安定性を変化させ得るオリゴヌクレオチドであれば、いかなるものであってもよい。ここで「相補的」な配列とは、NATに対して完全相補的な配列のみならず、細胞の生理的な条件下でNATとハイブリダイズしてmRNAへのNATの作用を阻害し得る限り、1ないし数(2, 3, 4 もしくは 5)塩基のミスマッチを含んでもよい。好ましくは、標的NATに相補的な配列とは、ストリンジェントな条件、例えば、Current Protocols in Molecular Biology, John Wiley & Sons,6.3.1-6.3.6, 1999に記載される条件(例えば、6×SSC(sodium chloride/sodium citrate)/45℃でのハイブリダイゼーション、次いで0.2×SSC/0.1% SDS/50~65℃での一回以上の洗浄等が挙げられる)下で、該NATとハイブリダイズし得る配列である。
上述のように、標的mRNAに対するアンチセンスヌクレオチドは、該mRNAの熱力学的に安定でない部分と相互作用して該mRNAの安定性を調節するので、本発明のセンスオリゴヌクレオチドは、標的mRNA中の熱力学的に安定でない部分と相同な配列を含むことが好ましい。熱力学的に安定でない部分としては、mRNAが二次構造をとった際に一本鎖の状態にある(例えば、ステムループ構造のループ部分にあたる)領域が挙げられる。
尚、「相同な」配列とは、標的mRNAの特定の部分ヌクレオチド配列と完全に同一な配列のみならず、該mRNAに対するNATと細胞の生理的な条件下でハイブリダイズしてmRNAに対するNATの作用を阻害し得る限り、1ないし数(2, 3, 4もしくは 5)個の塩基が異なっていてもよい。より好ましくは、「相同な」配列とは、標的mRNAの標的部位の配列とセンスオリゴヌクレオチドの配列とをalignさせたときに、オーバーラップする領域において、標的mRNAに対して90%以上の同一性を有するヌクレオチド配列を意味する。
The sense oligonucleotide of the present invention includes a sequence complementary to NAT (and thus a sequence homologous to the 3 ′ UTR), which contains a sequence complementary to the 3 ′ UTR of the target mRNA, and changes the stability of the target mRNA. Any oligonucleotide can be used as long as it can be used. Here, the “complementary” sequence is not only a sequence that is completely complementary to NAT, but as long as it can hybridize with NAT under physiological conditions of cells and inhibit the action of NAT on mRNA, It may contain one to several (2, 3, 4 or 5) base mismatches. Preferably, the sequence complementary to the target NAT refers to stringent conditions such as those described in Current Protocols in Molecular Biology, John Wiley & Sons, 6.3.1-6.3.6, 1999 (eg 6 × Under the condition of SSC (sodium chloride / sodium citrate) / 45 ° C hybridization, followed by 0.2 × SSC / 0.1% SDS / 50-65 ° C one or more washings). The sequence to get.
As described above, since the antisense nucleotide against the target mRNA interacts with a thermodynamically unstable portion of the mRNA to regulate the stability of the mRNA, the sense oligonucleotide of the present invention contains the target oligonucleotide in the target mRNA. It preferably contains a sequence homologous to a portion that is not thermodynamically stable. Examples of the portion that is not thermodynamically stable include a region that is in a single-stranded state when the mRNA has a secondary structure (for example, a region corresponding to the loop portion of the stem-loop structure).
The “homologous” sequence is not only a sequence that is completely identical to the specific partial nucleotide sequence of the target mRNA, but also the action of NAT on the mRNA by hybridizing with NAT on the mRNA under physiological conditions of the cell. 1 to several (2, 3, 4 or 5) bases may be different as long as they can be inhibited. More preferably, the “homologous” sequence is 90% or more identity to the target mRNA in the overlapping region when the sequence of the target site of the target mRNA and the sequence of the sense oligonucleotide are aligned. Means a nucleotide sequence having
さらに、本発明のセンスオリゴヌクレオチドは、配列非特異的な反応を起こす配列(例えば、5’-CG-3’、5’-GGGG-3’、5’-GGGGG-3’等)を含まないものから選択することが好ましく、また、標的mRNA以外のRNA中に類似の配列が存在しないものから選択することが好ましい。他のRNA中に類似の配列が存在しないことは、アンチセンスオリゴヌクレオチドについて上記したと同様の方法により確認することができる。 Furthermore, the sense oligonucleotide of the present invention does not contain a sequence that causes a non-sequence-specific reaction (for example, 5′-CG-3 ′, 5′-GGGG-3 ′, 5′-GGGGG-3 ′, etc.). It is preferable to select from those having no similar sequence in RNA other than the target mRNA. The absence of similar sequences in other RNAs can be confirmed by methods similar to those described above for antisense oligonucleotides.
好ましくは、本発明のCCL2 NATに対するセンスオリゴヌクレオチドは、配列番号1に示されるラットCCL2 mRNA 3’UTR配列にあっては、ヌクレオチド番号25-51で示されるドメイン1もしくはヌクレオチド番号103-175で示されるドメイン2内、他の哺乳動物におけるCCL2オルソログにあっては、前記ラットCCL2における各ドメインに対応するドメイン内の、1以上のループ構造の少なくとも一部(例えば、3塩基以上)を含む配列に相同なヌクレオチド配列を含む。例えば、ドメイン1に対して相同なセンスオリゴヌクレオチドとして、CCL2 mRNAの発現を増強し得るTTAAGTAATGTTAAACTTAT(CCL2-Se1; 配列番号8)、ドメイン2に対して相同なセンスオリゴヌクレオチドとして、CCL2 mRNAの発現を増強し得るTCCATTTTTTTATTTCTCTG(CCL2-Se2; 配列番号9)がそれぞれ挙げられるが、これらに限定されない。
Preferably, the sense oligonucleotide for CCL2 NAT of the present invention, in the rat CCL2 mRNA 3'UTR sequence shown in SEQ ID NO: 1, is represented by domain 1 shown by nucleotide numbers 25-51 or nucleotide numbers 103-175 In the CCL2 ortholog in other mammals in the
好ましくは、本発明のCCL20 NATに対するセンスオリゴヌクレオチドは、配列番号2に示されるラットCCL20 mRNA 3’UTR配列にあっては、ヌクレオチド番号65-107で示されるドメイン1、ヌクレオチド番号124-155で示されるドメイン2もしくはヌクレオチド番号214-282で示されるドメイン3内、他の哺乳動物におけるCCL20オルソログにあっては、前記ラットCCL20における各ドメインに対応するドメイン内の、1以上のループ構造の少なくとも一部(例えば、3塩基以上)を含む配列に相同なヌクレオチド配列を含む。例えば、ドメイン1に対して相同なセンスオリゴヌクレオチドとして、CCL20 mRNAの発現を抑制し得るGGTTTCACCTGCACATCACT(CCL20-Se1; 配列番号10)、ドメイン3に対して相同なセンスオリゴヌクレオチドとして、CCL20 mRNAの発現を増強し得るGTTTAGCTATTTAATGTTAA(CCL2-Se2; 配列番号11)がそれぞれ挙げられるが、これらに限定されない。
Preferably, the sense oligonucleotide for CCL20 NAT of the present invention is represented by the domain 1 shown by nucleotide numbers 65-107 and nucleotide numbers 124-155 in the rat CCL20 mRNA 3'UTR sequence shown by SEQ ID NO: 2. In
好ましくは、本発明のCX3CL1 NATに対するセンスオリゴヌクレオチドは、配列番号3に示されるラットCX3CL1 mRNAの3’UTR配列にあっては、ヌクレオチド番号1103-1202で示されるドメイン1、ヌクレオチド番号1305-1369で示されるドメイン2、ヌクレオチド番号1416-1482で示されるドメイン3、ヌクレオチド番号1633-1681で示されるドメイン4、ヌクレオチド番号1688-1745で示されるドメイン5もしくはヌクレオチド番号1784-1810で示されるドメイン6内、他の哺乳動物におけるCX3CL1オルソログにあっては、前記ラットCX3CL1における各ドメインに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列に相同なヌクレオチド配列を含む。例えば、ドメイン1に対して相同なセンスオリゴヌクレオチドとして、CX3CL1 mRNAの発現を増強し得るACTTGTGCATGTGTGTACTT(CX3CL1-Se1; 配列番号13)、ドメイン2に対して相同なセンスオリゴヌクレオチドとして、CX3CL1 mRNAの発現を増強し得るACAAAGTGTCTACTGAAGCA(CX3CL1-Se2; 配列番号14)およびCX3CL1 mRNAの発現を抑制し得るCTACTGAAGCAGAGAGCAGC(CX3CL1-Se3; 配列番号15)がそれぞれ挙げられるが、これらに限定されない。
Preferably, the sense oligonucleotide for CX3CL1 NAT of the present invention, in the 3′UTR sequence of rat CX3CL1 mRNA shown in SEQ ID NO: 3, is in domain 1, nucleotide numbers 1305-1369 shown as nucleotide numbers 1103-1202.
好ましくは、本発明のIL-23A NATに対するセンスオリゴヌクレオチドは、配列番号4に示されるラットIL-23A mRNAの3’UTR配列にあっては、ヌクレオチド番号37-55で示されるドメイン1、ヌクレオチド番号193-303で示されるドメイン2、ヌクレオチド番号380-448で示されるドメイン3もしくはヌクレオチド番号555-600で示されるドメイン4内、他の哺乳動物におけるIL-23Aオルソログにあっては、前記ラットIL-23Aの各ドメインに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列に相同なヌクレオチド配列を含む。例えば、ドメイン1に対して相同なセンスオリゴヌクレオチドとして、IL-23A mRNAの発現を抑制し得るAATCCATCAATGCAGACATC(IL23-Se1; 配列番号16)、ドメイン2に対して相同なセンスオリゴヌクレオチドとして、IL-23A mRNAの発現を増強し得るGAAGCTGGCAGACAGCTGCA(IL23-Se2; 配列番号17)、ドメイン3に対して相同なセンスオリゴヌクレオチドとして、IL-23A mRNAの発現を抑制し得るTCCTTCAGTTCTAACAGAAC(IL23-Se3; 配列番号18)、ドメイン4に対して相同なセンスオリゴヌクレオチドとして、IL-23A mRNAの発現を抑制し得るAACAGTTTAGAGGATTGTTA(IL23-Se4; 配列番号19)がそれぞれ挙げられるが、これらに限定されない。
Preferably, the sense oligonucleotide for IL-23A NAT of the present invention, in the 3′UTR sequence of rat IL-23A mRNA shown in SEQ ID NO: 4, is the domain 1, nucleotide number shown by nucleotide numbers 37-55 In the
好ましくは、本発明のCD69 NATに対するセンスオリゴヌクレオチドは、配列番号5に示されるラットCD69 mRNAの3’UTR配列にあっては、ヌクレオチド番号15-32で示されるドメイン1、ヌクレオチド番号120-186で示されるドメイン2、ヌクレオチド番号218-252で示されるドメイン3、ヌクレオチド番号344-370で示されるドメイン4、ヌクレオチド番号389-633で示されるドメイン5もしくはヌクレオチド番号656-786で示されるドメイン6内、他の哺乳動物におけるCD69オルソログにあっては、前記ラットCD69における各ドメインに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列に相同なヌクレオチド配列を含む。例えば、ドメイン5に対して相同なセンスオリゴヌクレオチドとして、CD69 mRNAの発現を増強し得るGACCAATGCTTATGAAAACA(CD69-Se2; 配列番号21)、ドメイン6に対して相同なセンスオリゴヌクレオチドとして、IL-23A mRNAの発現を増強し得るGTGGCAGATCTCTGTCAGGA(CD69-Se3; 配列番号22)がそれぞれ挙げられるが、これらに限定されない。
Preferably, the sense oligonucleotide for CD69 NAT of the present invention is the domain 1 represented by nucleotide numbers 15-32 and nucleotide numbers 120-186 in the 3′UTR sequence of rat CD69 mRNA represented by SEQ ID NO: 5. In
好ましくは、本発明のNF-κB p65 NATに対するセンスオリゴヌクレオチドは、配列番号6に示されるラットNF-κB p65 mRNAの3’UTR配列にあっては、ヌクレオチド番号161-301で示されるドメイン1、ヌクレオチド番号343-380で示されるドメイン2、ヌクレオチド番号401-412で示されるドメイン3もしくはヌクレオチド番号473-523で示されるドメイン4内、他の哺乳動物におけるNF-κB p65オルソログにあっては、前記ラットNF-κB p65における各ドメインに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列に相同なヌクレオチド配列を含む。例えば、ドメイン1に対して相同なセンスオリゴヌクレオチドとして、NF-κB p65 mRNAの発現を増強し得るGAACTCTTGAGACCCTGCTT(p65-Se1; 配列番号23)、ドメイン3に対して相同なセンスオリゴヌクレオチドとして、NF-κB p65 mRNAの発現を抑制し得るGCAACGCTCCTAGGAGCAGC(p65-Se3; 配列番号25)、ドメイン4に対して相同なセンスオリゴヌクレオチドとして、NF-κB p65 mRNAの発現を抑制し得るAACTCTCCATGCTGAGCAGT(p65-Se4; 配列番号26)がそれぞれ挙げられるが、これらに限定されない。
Preferably, the sense oligonucleotide for NF-κB p65 NAT of the present invention is the domain 1 represented by nucleotide numbers 161-301 in the 3 ′ UTR sequence of rat NF-κB p65 mRNA represented by SEQ ID NO: 6, In the
好ましくは、本発明のTNF-αNATに対するセンスオリゴヌクレオチドは、配列番号7に示されるラットTNF-α mRNAの3’UTR配列にあっては、ヌクレオチド番号304-405で示されるドメイン1、ヌクレオチド番号430-546で示されるドメイン2、ヌクレオチド番号687-713で示されるドメイン3もしくはヌクレオチド番号752-776で示されるドメイン4内、他の哺乳動物におけるTNF-αオルソログにあっては、前記ラットTNF-αにおける各ドメインに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列に相同なヌクレオチド配列を含む。例えば、ドメイン1に対して相同なセンスオリゴヌクレオチドとして、TNF-α mRNAの発現を増強し得るAGATGTCTCAGGCCTCCCTT(TNF-Se1; 配列番号27)、ドメイン2に対して相同なセンスオリゴヌクレオチドとして、TNF-α mRNAの発現を増強し得るGGAACCCCCTATATTTATAA(TNF-Se2; 配列番号28)およびTNF-α mRNAの発現を抑制し得るTAATTGCACCTGTGACTATT(TNF-Se3; 配列番号29)、ドメイン3に対して相同なセンスオリゴヌクレオチドとして、TNF-α mRNAの発現を抑制し得るAACAAGATATTTATCTAACC(TNF-Se5; 配列番号31)、ドメイン4に対して相同なセンスオリゴヌクレオチドとして、TNF-α mRNAの発現を抑制し得るACTGAACCTCTGCTCCCCAC(TNF-Se7; 配列番号33)がそれぞれ挙げられるが、これらに限定されない。
Preferably, the sense oligonucleotide for TNF-αNAT of the present invention is the domain 1, nucleotide number 430 represented by nucleotide number 304-405 in the 3′UTR sequence of rat TNF-α mRNA represented by SEQ ID NO: 7. In the
本発明のセンスオリゴヌクレオチドの長さに特に制限はないが、配列特異性の面から、標的NAT中の標的配列に相補的な部分を少なくとも10塩基以上、好ましくは約12塩基以上、より好ましくは約15塩基以上含むものである。また、合成の容易さ、製造コスト、投与のし易さ等の面から、50塩基以下、好ましくは40塩基以下、より好ましくは30塩基以下の塩基長を有するものが挙げられる。 The length of the sense oligonucleotide of the present invention is not particularly limited, but from the viewpoint of sequence specificity, the portion complementary to the target sequence in the target NAT is at least 10 bases or more, preferably about 12 bases or more, more preferably Contains about 15 bases or more. In addition, in view of ease of synthesis, production cost, ease of administration, and the like, those having a base length of 50 bases or less, preferably 40 bases or less, more preferably 30 bases or less are mentioned.
本発明のセンスオリゴヌクレオチドは、一本鎖DNA、一本鎖RNA、DNA/RNAキメラのいずれであってもよく、さらに公知の修飾の付加されたものであってもよい。ここで「ヌクレオチド」とは、プリン及びピリミジン塩基を含有するのみでなく、修飾されたその他の複素環型塩基をもつようなものを含んでいてもよい。センスオリゴヌクレオチドがDNA(ODN)の場合、標的NATとセンスODNとによって形成されるRNA:DNAハイブリッドは、内在性RNase Hに認識されて標的NATの選択的な分解を引き起こすことができる。 The sense oligonucleotide of the present invention may be any of single-stranded DNA, single-stranded RNA, and DNA / RNA chimera, and may be those with known modifications. Here, the “nucleotide” may include not only purine and pyrimidine bases but also those having other modified heterocyclic bases. When the sense oligonucleotide is DNA (ODN), the RNA: DNA hybrid formed by the target NAT and the sense ODN can be recognized by endogenous RNase H and cause selective degradation of the target NAT.
センスオリゴヌクレオチドを構成するヌクレオチド分子は、天然型のDNAもしくはRNAでもよいが、安定性(化学的及び/又は対酵素)や比活性(RNAとの親和性)を向上させるために、上記アンチセンスヌクレオチドの場合と同様に、種々の化学修飾を含むことができる。 The nucleotide molecule constituting the sense oligonucleotide may be natural DNA or RNA, but in order to improve stability (chemical and / or enzyme) and specific activity (affinity with RNA), the above antisense As with nucleotides, various chemical modifications can be included.
本発明のセンスオリゴヌクレオチドは、標的mRNA(cDNA)配列に基づいて、市販のDNA/RNA自動合成機(アプライド・バイオシステムズ社、ベックマン社等)を用いて、これに相同な配列を合成することにより調製することができる。 The sense oligonucleotide of the present invention synthesizes a sequence homologous to the target mRNA (cDNA) sequence using a commercially available DNA / RNA automatic synthesizer (Applied Biosystems, Beckman, etc.). Can be prepared.
本発明のセンスオリゴヌクレオチドは、リポソーム、ミクロスフェアのような特殊な形態で供与されたり、ポリリジンのようなポリカチオン体、脂質(例、ホスホリピド、コレステロールなど)などの疎水性物質が付加された形態で提供され得る。あるいはまた、本発明のセンスオリゴヌクレオチドを、膜透過機能を有するペプチド(例えば、ショウジョウバエ由来のAntennapediaホメオドメイン(AntP)、ヒト免疫不全ウイルス(HIV)由来のTAT、単純ヘルペスウイルス(HSV)由来のVP22等の細胞通過ドメイン)などで修飾することにより、該オリゴヌクレオチドの細胞への取り込みを促進することができる。 The sense oligonucleotide of the present invention is provided in a special form such as a liposome or a microsphere, or a form to which a hydrophobic substance such as a polycationic substance such as polylysine or a lipid (eg, phospholipid, cholesterol, etc.) is added. Can be provided at. Alternatively, the sense oligonucleotide of the present invention is converted into a peptide having a membrane permeation function (for example, Drosophila-derived Antennapedia homeodomain (AntP), human immunodeficiency virus (HIV) -derived TAT, herpes simplex virus (HSV) -derived VP22. And the like can be promoted by incorporating the oligonucleotide into a cell.
本発明のセンスオリゴヌクレオチドは、標的NATと相互作用することにより、該NATによる標的遺伝子の発現(タンパク質の産生)調節作用を変化させることができる。IL-1βにより誘導される、サイトカイン・ケモカイン遺伝子を含む種々の遺伝子は、炎症惹起もしくは抗炎症作用、感染防御、免疫応答の調節などの多様な生理活性を有するので、本発明のセンスオリゴヌクレオチドを含有する医薬は、標的遺伝子の発現調節剤として、炎症性疾患や感染症を含む種々の疾患の予防及び/又は治療に利用することができる。
例えば、CCL2の発現を増強させ得るセンスオリゴヌクレオチドは血管新生や創傷治癒などに利用することができ、一方、CCL2は単球や好酸球を遊走するため、CCL2の発現を抑制し得るセンスオリゴヌクレオチドは、動脈硬化を含む慢性炎症性疾患やアレルギー性炎症の予防・治療に有用である。
CCL20は広範囲の微生物に対して抗菌活性を示すので、CCL20の発現を増強させ得るセンスオリゴヌクレオチドは種々の微生物感染症の予防・治療に有効である。一方、CCL20は適応免疫の開始と持続に重要な役割を担っていることから、CCL20の発現を抑制し得るセンスオリゴヌクレオチドは、気管支喘息、多発性硬化症、関節リウマチ、各種皮膚炎、炎症性腸疾患などの自己免疫疾患の予防・治療に有効である。
CX3CL1の発現を増強させ得るセンスオリゴヌクレオチドは血管新生などに利用することができ、一方、CX3CL1の発現を抑制し得るセンスオリゴヌクレオチドは、動脈硬化、糸球体腎炎を含む炎症性疾患の予防・治療や拒絶抑制、関節リウマチの予防・治療に有用である。
IL-23Aは細胞性免疫の維持に重要であり、その過剰発現は多臓器炎症を引き起こすことから、IL-23Aの発現を増強させ得るセンスオリゴヌクレオチドは感染症の予防・治療に有効であり、一方、IL-23Aの発現を抑制し得るセンスオリゴヌクレオチドは、自己免疫疾患や炎症性疾患の予防・治療に有用である。
CD69はリンパ球の分化や活性化に重要な役割を果たしていることから、CD69の発現を増強させ得るセンスオリゴヌクレオチドは感染症の予防・治療に有効である。一方、CD69は炎症局所に発現する炎症細胞のほとんどで発現し、また関節リウマチや全身性エリテマトーデス(SLE)患者からCD69に対する自己抗体が検出されることなどから、CD69の発現を抑制し得るセンスオリゴヌクレオチドは、自己免疫疾患や炎症性疾患の予防・治療に有用である。
NF-κBは免疫系に重要な役割を持つ転写因子であることから、NF-κB p65の発現を増強させ得るセンスオリゴヌクレオチドは感染症の予防・治療に有効である。一方、NF-κBはがん細胞で恒常的に活性化してアポトーシスを阻害しており、また骨代謝や気管支喘息、関節炎、炎症性腸疾患、敗血症などの病態形成にも関与しているので、NF-κB p65の発現を抑制し得るセンスオリゴヌクレオチドは、がん、骨粗鬆症、自己免疫疾患や炎症性疾患の予防・治療に有用である。
TNF-αはアポトーシス誘導、炎症メディエーターや形質細胞による抗体産生の亢進などを通じて感染防御や抗腫瘍作用を示すので、TNF-αの発現を増強させ得るセンスオリゴヌクレオチドは感染症やがんの予防・治療に有効である。一方、TNF-αは関節リウマチ、乾癬、糖尿病、高脂血症、敗血症、骨粗鬆症の病態形成に関与するため、TNF-αの発現を抑制し得るセンスオリゴヌクレオチドは、これらの疾患の予防・治療に有用である。
The sense oligonucleotide of the present invention can change the target gene expression (protein production) regulating action by the NAT by interacting with the target NAT. Since various genes including cytokine / chemokine gene induced by IL-1β have various physiological activities such as inflammation-inducing or anti-inflammatory action, infection defense, and regulation of immune response, the sense oligonucleotide of the present invention is used. The contained drug can be used as a target gene expression regulator for the prevention and / or treatment of various diseases including inflammatory diseases and infectious diseases.
For example, sense oligonucleotides that can enhance the expression of CCL2 can be used for angiogenesis and wound healing, while CCL2 migrates to monocytes and eosinophils, so that it can suppress the expression of CCL2. Nucleotides are useful for the prevention and treatment of chronic inflammatory diseases including arteriosclerosis and allergic inflammation.
Since CCL20 exhibits antibacterial activity against a wide range of microorganisms, sense oligonucleotides that can enhance the expression of CCL20 are effective in the prevention and treatment of various microbial infections. On the other hand, since CCL20 plays an important role in the initiation and maintenance of adaptive immunity, sense oligonucleotides that can suppress the expression of CCL20 are bronchial asthma, multiple sclerosis, rheumatoid arthritis, various dermatitis, inflammatory It is effective for the prevention and treatment of autoimmune diseases such as bowel disease.
Sense oligonucleotides that can enhance CX3CL1 expression can be used for angiogenesis, while sense oligonucleotides that can suppress CX3CL1 expression prevent or treat inflammatory diseases including arteriosclerosis and glomerulonephritis It is useful for the prevention and treatment of anti-rejection and rheumatoid arthritis.
IL-23A is important for the maintenance of cellular immunity, and its overexpression causes multi-organ inflammation. Therefore, sense oligonucleotides that can enhance IL-23A expression are effective in the prevention and treatment of infectious diseases. On the other hand, sense oligonucleotides that can suppress the expression of IL-23A are useful for the prevention and treatment of autoimmune diseases and inflammatory diseases.
Since CD69 plays an important role in the differentiation and activation of lymphocytes, a sense oligonucleotide that can enhance the expression of CD69 is effective in the prevention and treatment of infectious diseases. On the other hand, CD69 is expressed in most inflammatory cells that are expressed locally, and autoantibodies against CD69 are detected in patients with rheumatoid arthritis and systemic lupus erythematosus (SLE). Nucleotides are useful for the prevention and treatment of autoimmune diseases and inflammatory diseases.
Since NF-κB is a transcription factor having an important role in the immune system, a sense oligonucleotide that can enhance the expression of NF-κB p65 is effective in the prevention and treatment of infectious diseases. On the other hand, NF-κB is constantly activated in cancer cells and inhibits apoptosis, and is also involved in pathogenesis such as bone metabolism, bronchial asthma, arthritis, inflammatory bowel disease, sepsis, A sense oligonucleotide capable of suppressing the expression of NF-κB p65 is useful for the prevention and treatment of cancer, osteoporosis, autoimmune diseases and inflammatory diseases.
TNF-α shows protection against infection and anti-tumor action by inducing apoptosis and enhancing antibody production by inflammatory mediators and plasma cells. Therefore, sense oligonucleotides that can enhance the expression of TNF-α prevent infection and cancer. It is effective for treatment. On the other hand, since TNF-α is involved in the pathogenesis of rheumatoid arthritis, psoriasis, diabetes, hyperlipidemia, sepsis, and osteoporosis, sense oligonucleotides that can suppress the expression of TNF-α are used to prevent and treat these diseases. Useful for.
本発明のセンスオリゴヌクレオチドを含有する医薬は低毒性であり、そのまま液剤として、又は適当な剤型の医薬組成物として、ヒト又は非ヒト哺乳動物(例、マウス、ラット、モルモット、ウサギ、ヒツジ、ブタ、ウシ、ネコ、イヌ、サルなど)に対して経口的又は非経口的(例、吸入投与、血管内投与、皮下投与、経粘膜投与など)に投与することができる。
これらの核酸を上記の各種疾患の予防・治療剤として使用する場合、上記の本発明のアンチセンスヌクレオチドと同様に製剤化し、投与することができる。
The medicament containing the sense oligonucleotide of the present invention has low toxicity and can be used as a liquid or as a pharmaceutical composition of an appropriate dosage form as a human or non-human mammal (eg, mouse, rat, guinea pig, rabbit, sheep, Pigs, cows, cats, dogs, monkeys, etc.) can be administered orally or parenterally (eg, inhalation administration, intravascular administration, subcutaneous administration, transmucosal administration etc.).
When these nucleic acids are used as a prophylactic / therapeutic agent for the above-mentioned various diseases, they can be formulated and administered in the same manner as the antisense nucleotide of the present invention.
本発明のセンスオリゴヌクレオチドを含有する上記医薬の投与量は、投与対象、対象疾患、症状、投与ルートなどによっても異なるが、例えば、本発明のセンスオリゴヌクレオチドを1回量として、通常0.01~20mg/kg体重程度、好ましくは0.1~10mg/kg体重程度、さらに好ましくは0.1~5mg/kg体重程度を、1日1回~数回程度、静脈注射や吸入等により投与するのが好都合である。他の非経口投与及び経口投与の場合もこれに準ずる量を投与することができる。症状が特に重い場合には、その症状に応じて増量してもよい。 The dose of the above-mentioned medicament containing the sense oligonucleotide of the present invention varies depending on the administration subject, target disease, symptom, administration route, etc. For example, the dose of the sense oligonucleotide of the present invention is usually 0.01 to 20 mg. It is convenient to administer about 0.1 kg / kg body weight, preferably about 0.1-10 mg / kg body weight, more preferably about 0.1-5 mg / kg body weight by intravenous injection or inhalation once to several times a day. In the case of other parenteral administration and oral administration, an equivalent amount can be administered. If symptoms are particularly severe, the dose may be increased according to the symptoms.
なお前記した医薬組成物は、本発明のセンスオリゴヌクレオチドとの配合により好ましくない相互作用を生じない限り、他の薬剤を含有してもよい。他の薬剤としては、例えば、抗ウイルス薬、抗腫瘍薬、抗菌薬、抗真菌薬、抗原虫薬、抗生物質、抗セプシス薬、抗セプティックショック薬、エンドトキシン拮抗薬、免疫調節薬、非ステロイド性抗炎症薬、ステロイド薬、炎症性メディエーター作用抑制薬、炎症性メディエーター産生抑制薬、抗炎症性メディエーター作用抑制薬、抗炎症性メディエーター産生抑制薬などが挙げられる。 The above-described pharmaceutical composition may contain other drugs as long as no undesirable interaction is caused by the combination with the sense oligonucleotide of the present invention. Other drugs include, for example, antiviral drugs, antitumor drugs, antibacterial drugs, antifungal drugs, antiprotozoal drugs, antibiotics, antiseptic drugs, antiseptic shock drugs, endotoxin antagonists, immunomodulators, non-steroidal drugs Examples include anti-inflammatory drugs, steroid drugs, inflammatory mediator action inhibitors, inflammatory mediator production inhibitors, anti-inflammatory mediator action inhibitors, anti-inflammatory mediator production inhibitors.
本発明はまた、標的mRNAに対するNATの作用を調節することにより標的遺伝子の発現(タンパク質の産生)を調節する物質をスクリーニングする方法を提供する。本発明のスクリーニング方法は、被験物質の存在下及び非存在下で、標的mRNAとそれに対するNATとのハイブリダイゼーションを検出し、その程度を比較することを特徴とする。
例えば、標的mRNAとNATとを常法により単離し、いずれか一方を固相化し、他方を適当な標識剤で標識して、RNAが生理的な二次構造を形成し得る条件下で、被験物質の存在下及び非存在下に両者をハイブリダイズさせ、固相に結合した標識量を両条件下で比較する方法が挙げられる。ここでmRNA及びNATとしては、それぞれその全長を用いてもよいし、あるいはmRNAの3’UTR、並びに該領域に相補的なNAT配列を含むそれらのフラグメントを用いてもよい。
The present invention also provides a method of screening for a substance that regulates target gene expression (protein production) by regulating the action of NAT on the target mRNA. The screening method of the present invention is characterized by detecting the hybridization between a target mRNA and NAT for the target mRNA in the presence and absence of a test substance and comparing the degree thereof.
For example, target mRNA and NAT are isolated by a conventional method, one of them is solid-phased and the other is labeled with an appropriate labeling agent, and the test is performed under conditions where RNA can form a physiological secondary structure. Examples include a method of hybridizing both in the presence and absence of a substance and comparing the amount of label bound to the solid phase under both conditions. Here, as mRNA and NAT, the full length thereof may be used, respectively, or the 3 ′ UTR of mRNA and a fragment containing a NAT sequence complementary to the region may be used.
固相の材料としては、シリコンなどの半導体、ガラス、ダイアモンドなどの無機物、ポリエチレンテレフタレート、ポリプロピレン等の高分子物質を主成分とするフィルムなどが挙げられ、また固相の形状としては、スライドガラス、マイクロウェルプレート、マイクロビーズ、繊維型などが挙げられるが、それらに制限されない。固相上にmRNAもしくはNATを固定化する方法としては、予め該RNAにアミノ基、アルデヒド基、SH基、ビオチンなどの官能基を導入しておき、一方、固相上にも該RNAと反応し得る官能基(例:アルデヒド基、アミノ基、SH基、ストレプトアビジンなど)を導入し、両官能基間の共有結合で固相とRNAを架橋したり、ポリアニオン性のRNAに対して、固相をポリカチオンコーティングして静電結合を利用してRNAを固定化するなどの方法が挙げられるが、これらに限定されない。固相化RNAの調製法としては、フォトリソグラフィー法を用いてRNAを基板(ガラス、シリコンなど)上で1ヌクレオチドずつ合成するAffymetrix方式と、マイクロスポッティング法、インクジェット法、バブルジェット(登録商標)法などを用いて、予め調製されたRNAを基板上にスポッティングするStanford方式とが挙げられるが、使用するRNAの塩基長を考慮すれば、Stanford方式あるいは両者を組み合わせた手法を用いるのが好ましい。 Examples of the solid phase material include semiconductors such as silicon, inorganic materials such as glass and diamond, films mainly composed of high molecular substances such as polyethylene terephthalate and polypropylene, and the shape of the solid phase includes a slide glass, Examples include, but are not limited to, microwell plates, microbeads, and fiber types. As a method for immobilizing mRNA or NAT on a solid phase, functional groups such as amino group, aldehyde group, SH group, and biotin are introduced into the RNA in advance, while reacting with the RNA on the solid phase. Functional groups (eg, aldehyde group, amino group, SH group, streptavidin, etc.) are introduced, and the solid phase and RNA are cross-linked by covalent bond between the two functional groups, or the polyanionic RNA is immobilized. Examples of the method include, but are not limited to, a method of immobilizing RNA using polycation coating of a phase and electrostatic bonding. The solid-phase RNA preparation methods include the Affymetrix method, which synthesizes RNA one nucleotide at a time on a substrate (glass, silicon, etc.) using a photolithography method, the micro spotting method, the inkjet method, and the bubble jet (registered trademark) method The Stanford method in which RNA prepared in advance is spotted on the substrate using the above method, but considering the base length of the RNA to be used, it is preferable to use the Stanford method or a method combining both.
標識剤としては、例えば、放射性同位元素、酵素、蛍光物質、発光物質などが用いられる。放射性同位元素としては、例えば、〔32P〕、〔3H〕、〔14C〕などが用いられる。酵素としては、安定で比活性の大きなものが好ましく、例えば、β-ガラクトシダーゼ、β-グルコシダーゼ、アルカリフォスファターゼ、パーオキシダーゼ、リンゴ酸脱水素酵素などが用いられる。蛍光物質としては、例えば、フルオレスカミン、フルオレッセンイソチオシアネート、Cy3、Cy5などが用いられる。発光物質としては、例えば、ルミノール、ルミノール誘導体、ルシフェリン、ルシゲニンなどが用いられる。さらに、プローブと標識剤との結合にビオチン-(ストレプト)アビジンを用いることもできる。 As the labeling agent, for example, a radioisotope, an enzyme, a fluorescent substance, a luminescent substance, or the like is used. As the radioisotope, for example, [ 32 P], [ 3 H], [ 14 C] and the like are used. As the enzyme, a stable enzyme having a large specific activity is preferable. For example, β-galactosidase, β-glucosidase, alkaline phosphatase, peroxidase, malate dehydrogenase and the like are used. As the fluorescent material, for example, fluorescamine, fluorescein isothiocyanate, Cy3, Cy5 and the like are used. As the luminescent substance, for example, luminol, luminol derivatives, luciferin, lucigenin and the like are used. Furthermore, biotin- (strept) avidin can also be used for binding between the probe and the labeling agent.
被験物質としては、いかなる公知物質及び新規物質であってもよく、例えば、核酸、糖質、脂質、タンパク質、ペプチド、有機低分子化合物、コンビナトリアルケミストリー技術を用いて作製された化合物ライブラリー、固相合成やファージディスプレイ法により作製されたランダムペプチドライブラリー、あるいは微生物、動植物、海洋生物等由来の天然成分などがあげられる。添加される被験物質の濃度は化合物の種類(溶解度、毒性等)により異なるが、例えば、約0.1nM~約100nMの範囲で適宜選択される。インキュベート時間としては、例えば、約1~約24時間が挙げられる。 The test substance may be any known substance or new substance, such as a nucleic acid, carbohydrate, lipid, protein, peptide, organic low molecular weight compound, compound library prepared using combinatorial chemistry techniques, solid phase Examples include random peptide libraries prepared by synthesis or phage display methods, or natural components derived from microorganisms, animals and plants, marine organisms, and the like. The concentration of the test substance to be added varies depending on the type of compound (solubility, toxicity, etc.), but is appropriately selected within the range of, for example, about 0.1 nM to about 100 nM. Examples of the incubation time include about 1 to about 24 hours.
固相上のRNAと、標識したRNA(及び被験物質)とを接触させて、インキュベートした後、固相に結合しなかったRNAを洗い流し、固相に結合したRNAの標識量を検出する。被験物質の存在下で、非存在下に比べて固相に結合した標識量が有意に増加した場合、該被験物質を標的mRNAに対するNATの作用を増強する物質の候補として選択することができる。一方、被験物質の存在下で、非存在下に比べて固相に結合した標識量が有意に減少した場合、該被験物質を標的mRNAに対するNATの作用を抑制する物質の候補として選択することができる。 After bringing the RNA on the solid phase into contact with the labeled RNA (and the test substance) and incubating, the RNA that did not bind to the solid phase is washed away, and the amount of RNA bound to the solid phase is detected. When the amount of the label bound to the solid phase is significantly increased in the presence of the test substance compared to the absence, the test substance can be selected as a candidate for a substance that enhances the action of NAT on the target mRNA. On the other hand, if the amount of label bound to the solid phase is significantly reduced in the presence of the test substance compared to the absence, the test substance can be selected as a candidate for a substance that suppresses the action of NAT on the target mRNA. it can.
好ましい実施態様においては、標的mRNAとNATとを発現する細胞に被験物質を接触させ、該細胞における該mRNA量及び/又はそれにコードされるタンパク質量の変化を測定することにより、より直接的に標的遺伝子の発現増強又は抑制物質を選択することができる。標的mRNAとNATとを発現する細胞は、両RNAを生来発現し得る細胞(例えば、IL-1β刺激した肝細胞等)であってもよいし、それらのいずれか一方もしくは両方を発現するDNAを導入した組換え細胞であってもよい。組換え細胞の場合、宿主細胞として、例えば、H4IIE-C3細胞、HepG2細胞、293T細胞、HEK293細胞、COS7細胞、2B4T細胞、CHO細胞、MCF-7細胞、H295R細胞などの動物細胞をあげることができる。標的mRNA及びNATをコードするDNAは、両RNAを常法により単離し、逆転写反応等によって二本鎖DNAに変換した後、宿主細胞内で機能しうるプロモーターを有する発現ベクターに挿入して、例えば、リン酸カルシウム共沈殿法、PEG法、エレクトロポレーション法、マイクロインジェクション法、リポフェクション法などにより、このベクターを宿主細胞に導入することにより作製することができる。 In a preferred embodiment, a target substance is contacted more directly by contacting a test substance with a cell that expresses the target mRNA and NAT, and measuring a change in the amount of the mRNA and / or protein encoded in the cell. Substances that enhance or suppress gene expression can be selected. The cell that expresses the target mRNA and NAT may be a cell that can naturally express both RNAs (for example, IL-1β-stimulated hepatocytes), or a DNA that expresses either or both of them. The introduced recombinant cell may also be used. In the case of recombinant cells, examples of host cells include animal cells such as H4IIE-C3 cells, HepG2 cells, 293T cells, HEK293 cells, COS7 cells, 2B4T cells, CHO cells, MCF-7 cells, and H295R cells. it can. The target mRNA and DNA encoding NAT are both isolated by conventional methods, converted into double-stranded DNA by reverse transcription, etc., and then inserted into an expression vector having a promoter that can function in the host cell. For example, it can be prepared by introducing this vector into a host cell by the calcium phosphate coprecipitation method, PEG method, electroporation method, microinjection method, lipofection method or the like.
被験物質としては、前記したとおりのものが用いられる。被験物質と上記細胞との接触は、例えば、該細胞の培養に適した培地(例えば、約5~20%の胎仔ウシ血清を含む最小必須培地(MEM)、ダルベッコ改変イーグル培地(DMEM)、RPMI1640培地、199培地、F12培地など)や各種緩衝液(例えば、HEPES緩衝液、リン酸緩衝液、リン酸緩衝生理食塩水、トリス塩酸緩衝液、ホウ酸緩衝液、酢酸緩衝液など)の中に被験物質を添加して、細胞を一定時間インキュベートすることにより実施することができる。添加される被験物質の濃度は化合物の種類(溶解度、毒性等)により異なるが、例えば、約0.1nM~約100nMの範囲で適宜選択される。インキュベート時間としては、例えば、約1~約48時間が挙げられる。必要に応じて、インキュベーションの際に上記細胞をウイルスに感染させてもよい。 As the test substance, those described above are used. The contact between the test substance and the cell is, for example, a medium suitable for culturing the cell (for example, a minimum essential medium (MEM) containing about 5 to 20% fetal calf serum, Dulbecco's modified Eagle medium (DMEM), RPMI1640 Medium, 199 medium, F12 medium, etc.) and various buffers (eg, HEPES buffer, phosphate buffer, phosphate buffered saline, Tris-HCl buffer, borate buffer, acetate buffer, etc.) It can be carried out by adding a test substance and incubating the cells for a certain period of time. The concentration of the test substance to be added varies depending on the type of compound (solubility, toxicity, etc.), but is appropriately selected within the range of, for example, about 0.1 nM to about 100 nM. Examples of the incubation time include about 1 to about 48 hours. If necessary, the cells may be infected with the virus during the incubation.
インキュベーション終了後(あるいは経時的に)、細胞からRNAを抽出してRT-PCR、リアルタイムPCRやノーザンブロット解析により標的mRNA量を測定するか、あるいは培養上清を回収して、自体公知の各種イムノアッセイやウェスタンブロッティング等により標的遺伝子にコードされるタンパク質量を測定する。被験物質の添加により細胞における標的mRNA量及び/又はタンパク質量が有意に増加した場合、該被験物質を標的遺伝子の発現増強物質の候補として選択することができる。一方、被験物質の添加により該mRNA量及び/又はタンパク質量が有意に減少した場合、該被験物質を標的遺伝子の発現抑制物質の候補として選択することができる。 After incubation (or over time), RNA is extracted from the cells and the target mRNA level is measured by RT-PCR, real-time PCR or Northern blot analysis, or the culture supernatant is collected and various immunoassays known per se Or the amount of protein encoded by the target gene is measured by Western blotting or the like. When the amount of target mRNA and / or protein in a cell is significantly increased by adding a test substance, the test substance can be selected as a candidate for a target gene expression enhancing substance. On the other hand, when the amount of mRNA and / or protein is significantly decreased by the addition of a test substance, the test substance can be selected as a candidate for a target gene expression inhibitor.
上記のスクリーニング法により選択された標的遺伝子の発現を増強または抑制し得る物質は、該遺伝子の発現を増強または抑制し得るアンチセンスヌクレオチドやセンスオリゴヌクレオチドと同様に、上記の各種疾患の予防および/または治療用の医薬として使用することができる。 Substances capable of enhancing or suppressing the expression of the target gene selected by the screening method described above are used for the prevention and / or prevention of the above-mentioned various diseases in the same manner as antisense nucleotides and sense oligonucleotides that can enhance or suppress the expression of the gene. Alternatively, it can be used as a therapeutic drug.
上記のスクリーニング法により選択された物質を含有する医薬は低毒性であり、そのまま液剤として、又は適当な剤型の医薬組成物として、ヒト又は非ヒト哺乳動物(例、マウス、ラット、モルモット、ウサギ、ヒツジ、ブタ、ウシ、ネコ、イヌ、サルなど)に対して経口的又は非経口的(例、吸入投与、血管内投与、皮下投与など)に投与することができる。投与に用いられる医薬組成物としては、選択された物質と薬理学的に許容され得る担体、希釈剤もしくは賦形剤とを含むものであってよい。 A medicine containing a substance selected by the above screening method has low toxicity, and can be used as a liquid or as a pharmaceutical composition of an appropriate dosage form as a human or non-human mammal (eg, mouse, rat, guinea pig, rabbit). , Sheep, pigs, cows, cats, dogs, monkeys, etc.) or orally (eg, inhalation administration, intravascular administration, subcutaneous administration, etc.). The pharmaceutical composition used for administration may comprise a selected substance and a pharmacologically acceptable carrier, diluent or excipient.
非経口投与のための組成物としては、例えば、注射剤、エアロゾル剤、坐剤等が用いられ、注射剤は静脈注射剤、皮下注射剤、皮内注射剤、筋肉注射剤、点滴注射剤等の剤形を包含してもよい。このような注射剤は、公知の方法に従って調製できる。注射剤の調製方法としては、例えば、選択された標的遺伝子の発現増強又は抑制物質を通常注射剤に用いられる無菌の水性液、又は油性液に溶解、懸濁又は乳化することによって調製できる。注射用の水性液としては、例えば、生理食塩水、ブドウ糖やその他の補助薬を含む等張液等が用いられ、適当な溶解補助剤、例えば、アルコール(例、エタノール)、ポリアルコール(例、プロピレングリコール、ポリエチレングリコール)、非イオン界面活性剤〔例、ポリソルベート80、HCO-50(polyoxyethylene(50mol)adduct of hydrogenated castor oil)〕等と併用してもよい。油性液としては、例えば、ゴマ油、大豆油等が用いられ、溶解補助剤として安息香酸ベンジル、ベンジルアルコール等を併用してもよい。調製された注射液は、適当なアンプルに充填されることが好ましい。エアロゾル製剤はジクロロジフロロメタン、プロパン、窒素などのような圧縮された許容しうる抛射薬内に入れることができる。あるいはネブライザーやアトマイザーのような非圧縮性製剤用医薬品して製剤化してもよい。直腸投与に用いられる坐剤は、上記標的遺伝子の発現増強又は抑制物質を通常の坐薬用基剤に混合することによって調製されても良い。 As a composition for parenteral administration, for example, injections, aerosols, suppositories and the like are used, and injections are intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections, intravenous injections, etc. May be included. Such an injection can be prepared according to a known method. As a method for preparing an injection, it can be prepared, for example, by dissolving, suspending or emulsifying a substance for enhancing or suppressing the expression of a selected target gene in a sterile aqueous liquid or oily liquid that is usually used for injection. As an aqueous solution for injection, for example, an isotonic solution containing physiological saline, glucose and other adjuvants, and the like are used, and suitable solubilizers such as alcohol (eg, ethanol), polyalcohol (eg, Propylene glycol, polyethylene glycol), nonionic surfactants (eg, polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct-of-hydrogenated-castor-oil)) and the like may be used in combination. As the oily liquid, for example, sesame oil, soybean oil and the like are used, and benzyl benzoate, benzyl alcohol and the like may be used in combination as a solubilizing agent. The prepared injection solution is preferably filled in a suitable ampoule. Aerosol formulations can be placed in compressed acceptable propellants such as dichlorodifluoromethane, propane, nitrogen and the like. Alternatively, it may be formulated as a non-compressible pharmaceutical product such as a nebulizer or an atomizer. A suppository used for rectal administration may be prepared by mixing a substance for enhancing or suppressing the expression of the target gene with a normal suppository base.
経口投与のための組成物としては、固体又は液体の剤形、具体的には錠剤(糖衣錠、フィルムコーティング錠を含む)、丸剤、顆粒剤、散剤、カプセル剤(ソフトカプセル剤を含む)、シロップ剤、乳剤、懸濁剤等が挙げられる。このような組成物は公知の方法によって製造され、製剤分野において通常用いられる担体、希釈剤もしくは賦形剤を含有していても良い。錠剤用の担体、賦形剤としては、例えば、乳糖、でんぷん、蔗糖、ステアリン酸マグネシウムが用いられる。 Compositions for oral administration include solid or liquid dosage forms, specifically tablets (including sugar-coated tablets and film-coated tablets), pills, granules, powders, capsules (including soft capsules), and syrups. Agents, emulsions, suspensions and the like. Such a composition is produced by a known method and may contain a carrier, a diluent or an excipient usually used in the pharmaceutical field. As the carrier and excipient for tablets, for example, lactose, starch, sucrose, and magnesium stearate are used.
上記の非経口用又は経口用医薬組成物は、活性成分の投与量に適合するような投薬単位の剤形に調製されることが好都合である。このような投薬単位の剤形としては、例えば、錠剤、丸剤、カプセル剤、注射剤(アンプル)、エアロゾル剤、坐剤が挙げられる。標的遺伝子の発現増強又は抑制物質は、投薬単位剤形当たり通常5~500mg、とりわけ注射剤では5~100mg、その他の剤形では10~250mg含有されていることが好ましい。 The above parenteral or oral pharmaceutical composition is conveniently prepared in a dosage unit form suitable for the dosage of the active ingredient. Examples of such dosage forms include tablets, pills, capsules, injections (ampoules), aerosols, and suppositories. It is preferable that the substance for enhancing or suppressing the expression of the target gene is usually contained in an amount of 5 to 500 mg per dosage unit dosage form, particularly 5 to 100 mg for injections and 10 to 250 mg for other dosage forms.
上記の標的遺伝子の発現増強又は抑制物質を含有する上記医薬の投与量は、投与対象、対象疾患、症状、投与ルートなどによっても異なるが、例えば、該物質を1回量として、通常0.01~20mg/kg体重程度、好ましくは0.1~10mg/kg体重程度、さらに好ましくは0.1~5mg/kg体重程度を、1日1回~数回程度、静脈注射により投与するのが好都合である。他の非経口投与及び経口投与の場合もこれに準ずる量を投与することができる。症状が特に重い場合には、その症状に応じて増量してもよい。 The dose of the above-mentioned pharmaceutical containing a substance that enhances or suppresses the expression of the target gene varies depending on the administration subject, target disease, symptom, administration route, etc., but for example, the substance is usually 0.01 to 20 mg per dose It is convenient to administer about 0.1 kg / kg body weight, preferably about 0.1 to 10 mg / kg body weight, more preferably about 0.1 to 5 mg / kg body weight by intravenous injection once to several times a day. In the case of other parenteral administration and oral administration, an equivalent amount can be administered. If symptoms are particularly severe, the dose may be increased according to the symptoms.
以下に実施例を挙げて本発明をさらに具体的に説明するが、これらは単なる例示であって本発明の範囲を何ら限定するものではない。 Hereinafter, the present invention will be described more specifically with reference to examples. However, these are merely examples and do not limit the scope of the present invention.
(材料と方法)
肝細胞のトランスフェクション
Prostaglandins 1993;45:459-474の記載に従って、雄性Wistarラット(SPF/VAF Crlj:WI; 日本チャールズ・リバー株式会社)から肝細胞を単離し、播種して、37℃で一晩インキュベートした。肝細胞(3.0x105細胞/ディッシュ)をセンスODNでトランスフェクトした。各ODN(1.5 μg)をmagnet-assisted transfection A reagent (IBA) 1.5 μLと混合し、各ウェルに添加して磁性プレート上で15分間インキュベートした後、培地を新鮮なWilliam’s E培地(シグマ-アルドリッチ社)に置換した。細胞を一晩培養し、1 nM ヒトIL-1β(大塚製薬株式会社)で4時間(RNA調製)もしくは8時間(細胞抽出)処理した。動物実験は、立命館大学びわこくさつキャンパスの動物実験委員会の承認を得て実施した。
(Materials and methods)
Transfection of hepatocytes Hepatocytes were isolated from male Wistar rats (SPF / VAF Crlj: WI; Charles River Japan) as described in Prostaglandins 1993; 45: 459-474 and seeded at 37 ° C. Incubated overnight. Hepatocytes (3.0 × 10 5 cells / dish) were transfected with sense ODN. Each ODN (1.5 μg) was mixed with 1.5 μL of magnet-assisted transfection A reagent (IBA), added to each well, incubated on the magnetic plate for 15 minutes, and then the medium was treated with fresh William's E medium (Sigma-Aldrich). ). The cells were cultured overnight and treated with 1 nM human IL-1β (Otsuka Pharmaceutical Co., Ltd.) for 4 hours (RNA preparation) or 8 hours (cell extraction). Animal experiments were conducted with the approval of the Animal Experiment Committee of Ritsumeikan University Biwako Kusatsu Campus.
mRNAの二次構造予測
mRNAの3’UTRの二次構造は、mfoldプログラム(Zuker)によって予測した。mRNAの3’UTRの予測された構造の中から共通の保存された領域を選択した。各領域は少なくとも1つのステム-ループ構造を含んでいた。
Secondary structure prediction of mRNA The secondary structure of 3'UTR of mRNA was predicted by the mfold program (Zuker). A common conserved region was selected from the predicted structure of the 3'UTR of mRNA. Each region contained at least one stem-loop structure.
センスODNのデザイン
ホスホチオエート結合で保護されたセンスODNを、J. Neurochem. 2003;86:374-382に記載の方法に従ってデザインした(ジーン・デザイン社)。デザインされたセンスODN配列は、ヒト-ラット間で保存された共通領域の少なくとも1つのループを含むmRNA配列に対応しており、CpGモチーフやG-カルテットは避けた。
Design of Sense ODN Sense ODN protected with a phosphothioate bond was designed according to the method described in J. Neurochem. 2003; 86: 374-382 (Gene Design). The designed sense ODN sequence corresponds to an mRNA sequence containing at least one loop of a common region conserved between human and rat, and CpG motifs and G-quartets were avoided.
鎖特異的逆転写-ポリメラーゼ連鎖反応(RT-PCR)
Sepasol I Super(ナカライテスク)を用いて全RNAを調製し、TURBO DNA-free kit(Ambion-Applied Biosystems)で処理した。mRNAについてはオリゴdTプライマーを用い、NATについては鎖特異的プライマーを用いて相補DNA(cDNA)を合成し、Genes Cells. 2000;5:111-125に記載の方法に従い、一対のプライマー(オペロン・バイオテクノロジーズ)を用いて、ステップダウンPCRを実施した。
CCL2、CCL20、CX3CL1、IL-23A、CD69、NF-κB p65およびTNF-αの7遺伝子について、mRNAの増幅に使用したPCRプライマー、NATの逆転写用プライマーおよびPCRプライマーを表2および3にまとめた。
Strand-specific reverse transcription-polymerase chain reaction (RT-PCR)
Total RNA was prepared using Sepasol I Super (Nacalai Tesque) and treated with TURBO DNA-free kit (Ambion-Applied Biosystems). A complementary DNA (cDNA) was synthesized using an oligo dT primer for mRNA and a strand-specific primer for NAT, and a pair of primers (operon and operon) were synthesized according to the method described in Genes Cells. 2000; 5: 111-125. Biotechnologies) was used to perform step-down PCR.
Tables 2 and 3 summarize the PCR primers, NAT reverse transcription primers and PCR primers used for mRNA amplification for the seven genes CCL2, CCL20, CX3CL1, IL-23A, CD69, NF-κB p65 and TNF-α. It was.
(結果)
mRNAとNATの両方を転写している遺伝子を探索すべく、本発明者らはIL-1βにより誘導される21遺伝子(サイトカインおよびケモカイン遺伝子を含む)を選択し、その3’UTRに相補的なNATを検出しようと試みた。即ち、RT用の遺伝子特異的センスプライマー(表3)を用いた鎖特異的RT-PCRにより、これらの21遺伝子からNATが転写されているかどうかを調べた。表4に示すとおり、鎖特異的RT-PCR分析の結果、21遺伝子中16遺伝子(76%)からNATが転写されていることが分かった。興味深いことに、3’UTRR中のAREモチーフは13のNATを転写する遺伝子(81%)中に見出された。これらのデータはmRNAの3’UTRに対応するNATがIL-1β誘導性遺伝子から高頻度に転写されることを示唆しており、NATが広く発現しているとのアイデアを支持するものであった。
(result)
In order to search for genes that transcribe both mRNA and NAT, we selected 21 genes (including cytokines and chemokine genes) induced by IL-1β and complementary to their 3 'UTRs. Attempted to detect NAT. That is, whether or not NAT was transcribed from these 21 genes was examined by chain-specific RT-PCR using a gene-specific sense primer for RT (Table 3). As shown in Table 4, as a result of the strand-specific RT-PCR analysis, it was found that NAT was transcribed from 16 out of 21 genes (76%). Interestingly, the ARE motif in 3'UTRR was found in 13 NAT-transcribed genes (81%). These data suggest that NAT corresponding to the 3'UTR of mRNA is frequently transcribed from IL-1β-inducible genes, supporting the idea that NAT is widely expressed. It was.
本発明者らはmRNAとNATの両方が豊富に転写されている7遺伝子(CCL2、CCL20、CX3CL1、IL-23A、CD69、NF-κB p65およびTNF-α)を選択し、NATRE技術を適用した。本発明者らは、これらのmRNAの3’UTRの二次構造を予測し(図1A-図7A)、各センスODNは、mRNA-NAT間相互作用の阻害を引きこすmRNAと競合する共通の保存領域(図1B-図7B)の少なくとも1つのループを含むというルールに従って、センスODNをデザインした(表5および図1B-図7B)。 The present inventors selected 7 genes (CCL2, CCL20, CX3CL1, IL-23A, CD69, NF-κB p65 and TNF-α) in which both mRNA and NAT are abundantly transcribed, and applied NATRE technology. . The present inventors predicted the secondary structure of the 3′UTR of these mRNAs (FIGS. 1A-7A), and each sense ODN competes with the mRNA that leads to inhibition of the mRNA-NAT interaction. Sense ODNs were designed according to the rule of containing at least one loop of the storage region (FIGS. 1B-7B) (Table 5 and FIGS. 1B-7B).
ラット肝細胞を各センスODNでトランスフェクトし、細胞をIL-1βで刺激した後、標的mRNAの発現レベルを、Hepatology, 2008;47:686-697に記載されるように、SYBR Green Iを用いたリアルタイムPCRにより測定した。結果を図8に示す。内部標準にElongation factor 1alpha (EF) mRNAを用い、グラフの値は、各mRNA/EF mRNA (%)を示し、センスODN非導入細胞における発現レベルに対する相対量として表した。予測された二次構造の保存された領域内のループ部分を含むRNA配列に対して設計されたほとんどのセンスODNは、標的mRNAの発現レベルを変化(増加又は減少)させたのに対し、該二次構造の保存された領域外に対して設計されたセンスODN(TNF-Se4およびTNF-Se6)は標的mRNAの発現レベルに影響しなかった。 After transfecting rat hepatocytes with each sense ODN and stimulating the cells with IL-1β, the expression level of the target mRNA was determined using SYBR Green I as described in Hepatology,; 2008; 47: 686-697. Measured by real-time PCR. The results are shown in FIG. Elongation factor 1alpha (EF) mRNA was used as an internal standard, and the values in the graph indicate each mRNA / EF mRNA (%), and were expressed as a relative amount with respect to the expression level in non-sense ODN-introduced cells. Most sense ODNs designed for RNA sequences that contain loop portions within conserved regions of predicted secondary structure have altered (increased or decreased) the expression level of the target mRNA, whereas Sense ODNs (TNF-Se4 and TNF-Se6) designed outside the conserved region of secondary structure did not affect the expression level of the target mRNA.
本発明のアンチセンスヌクレオチドおよびセンスオリゴヌクレオチドは、IL-1βにより誘導される、内在性アンチセンス転写物が存在する炎症/感染関連遺伝子の発現調節剤として、炎症性疾患や感染症などの予防及び/又は治療に有用である。 The antisense nucleotide and sense oligonucleotide of the present invention are used as a regulator of the expression of an inflammation / infection-related gene in which an endogenous antisense transcript is present, which is induced by IL-1β. Useful for treatment.
本出願は、2011年5月12日付で日本国に出願された、特願2011-107707を基礎としており、ここで言及することにより、その内容は全て本明細書に包含される。 This application is based on Japanese Patent Application No. 2011-107707, filed in Japan on May 12, 2011, the contents of which are hereby incorporated by reference.
Claims (11)
(a) 配列番号1に示されるCCL2 mRNAの3’UTR配列中、ヌクレオチド番号25-51で示されるドメイン1もしくはヌクレオチド番号103-175で示されるドメイン2、又は他の哺乳動物におけるオルソログのそれらに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列と90%以上の同一性を有するヌクレオチド配列を含んでなる、CCL2遺伝子の発現を調節し得るセンスオリゴヌクレオチド
(b) 配列番号2に示されるCCL20 mRNAの3’UTR配列中、ヌクレオチド番号65-107で示されるドメイン1、ヌクレオチド番号124-155で示されるドメイン2もしくはヌクレオチド番号214-282で示されるドメイン3、又は他の哺乳動物におけるオルソログのそれらに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列と90%以上の同一性を有するヌクレオチド配列を含んでなる、CCL20遺伝子の発現を調節し得るセンスオリゴヌクレオチド
(c) 配列番号3に示されるCX3CL1 mRNAの3’UTR配列中、ヌクレオチド番号1103-1202で示されるドメイン1、ヌクレオチド番号1305-1369で示されるドメイン2、ヌクレオチド番号1416-1482で示されるドメイン3、ヌクレオチド番号1633-1681で示されるドメイン4、ヌクレオチド番号1688-1745で示されるドメイン5、もしくはヌクレオチド番号1784-1810で示されるドメイン6、又は他の哺乳動物におけるオルソログのそれらに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列と90%以上の同一性を有するヌクレオチド配列を含んでなる、CX3CL1遺伝子の発現を調節し得るセンスオリゴヌクレオチド
(d) 配列番号4に示されるIL-23A mRNAの3’UTR配列中、ヌクレオチド番号37-55で示されるドメイン1、ヌクレオチド番号193-303で示されるドメイン2、ヌクレオチド番号380-448で示されるドメイン3、もしくはヌクレオチド番号555-600で示されるドメイン4、又は他の哺乳動物におけるオルソログのそれらに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列と90%以上の同一性を有するヌクレオチド配列を含んでなる、IL-23A遺伝子の発現を調節し得るセンスオリゴヌクレオチド
(e) 配列番号5に示されるCD69 mRNAの3’UTR配列中、ヌクレオチド番号15-32で示されるドメイン1、ヌクレオチド番号120-186で示されるドメイン2、ヌクレオチド番号218-252で示されるドメイン3、ヌクレオチド番号344-370で示されるドメイン4、ヌクレオチド番号389-633で示されるドメイン5、もしくはヌクレオチド番号656-786で示されるドメイン6、又は他の哺乳動物におけるオルソログのそれらに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列と90%以上の同一性を有するヌクレオチド配列を含んでなる、CD69遺伝子の発現を調節し得るセンスオリゴヌクレオチド
(f) 配列番号6に示されるNF-κB p65 mRNAの3’UTR配列中、ヌクレオチド番号161-301で示されるドメイン1、ヌクレオチド番号343-380で示されるドメイン2、ヌクレオチド番号401-412で示されるドメイン3、もしくはヌクレオチド番号473-523で示されるドメイン4、又は他の哺乳動物におけるオルソログのそれらに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列と90%以上の同一性を有するヌクレオチド配列を含んでなる、NF-κB p65遺伝子の発現を調節し得るセンスオリゴヌクレオチド
(g) 配列番号7に示されるTNF-α mRNAの3’UTR配列中、ヌクレオチド番号304-405で示されるドメイン1、ヌクレオチド番号430-546で示されるドメイン2、ヌクレオチド番号687-713で示されるドメイン3、もしくはヌクレオチド番号752-776で示されるドメイン4、又は他の哺乳動物におけるオルソログのそれらに対応するドメイン内の、1以上のループ構造の少なくとも一部を含む配列と90%以上の同一性を有するヌクレオチド配列を含んでなる、TNF-α遺伝子の発現を調節し得るセンスオリゴヌクレオチド The sense oligonucleotide according to claim 6, which is any one of the following (a) to (g).
(a) In the 3′UTR sequence of CCL2 mRNA represented by SEQ ID NO: 1, domain 1 represented by nucleotide numbers 25-51 or domain 2 represented by nucleotide numbers 103-175, or those of orthologs in other mammals Sense oligonucleotide capable of regulating the expression of the CCL2 gene, comprising a nucleotide sequence having 90% or more identity with a sequence comprising at least part of one or more loop structures in the corresponding domain
(b) In the 3'UTR sequence of CCL20 mRNA represented by SEQ ID NO: 2, domain 1 represented by nucleotide numbers 65-107, domain 2 represented by nucleotide numbers 124-155, or domain 3 represented by nucleotide numbers 214-282 Or expression of the CCL20 gene comprising a nucleotide sequence having at least 90% identity with a sequence comprising at least a portion of one or more loop structures within a domain corresponding to those of orthologs in other mammals Regulatable sense oligonucleotide
(c) In the 3′UTR sequence of CX3CL1 mRNA represented by SEQ ID NO: 3, domain 1 represented by nucleotide numbers 1103-1202, domain 2 represented by nucleotide numbers 1305-1369, and domain 3 represented by nucleotide numbers 1416-1482 In domain 4, represented by nucleotide number 1633-1681, domain 5 represented by nucleotide number 1688-1745, or domain 6 represented by nucleotide number 1784-1810, or a domain corresponding to those of orthologs in other mammals A sense oligonucleotide capable of regulating the expression of the CX3CL1 gene, comprising a nucleotide sequence having at least 90% identity with a sequence comprising at least a part of one or more loop structures
(d) In the 3'UTR sequence of IL-23A mRNA shown in SEQ ID NO: 4, domain 1 shown by nucleotide numbers 37-55, domain 2 shown by nucleotide numbers 193-303, and nucleotide numbers 380-448 90% or more identity with a sequence comprising at least part of one or more loop structures within domain 3, or domain 4 represented by nucleotide numbers 555-600, or domains corresponding to those of orthologs in other mammals Sense oligonucleotide capable of regulating IL-23A gene expression comprising a nucleotide sequence having
(e) In the 3'UTR sequence of CD69 mRNA represented by SEQ ID NO: 5, domain 1 represented by nucleotide numbers 15-32, domain 2 represented by nucleotide numbers 120-186, domain 3 represented by nucleotide numbers 218-252 In domain 4, indicated by nucleotide numbers 344-370, domain 5 indicated by nucleotide numbers 389-633, or domain 6 indicated by nucleotide numbers 656-786, or domains corresponding to those of orthologs in other mammals A sense oligonucleotide capable of regulating the expression of the CD69 gene, comprising a nucleotide sequence having 90% or more identity with a sequence comprising at least a part of one or more loop structures
(f) In the 3′UTR sequence of NF-κB p65 mRNA represented by SEQ ID NO: 6, domain 1 represented by nucleotide numbers 161-301, domain 2 represented by nucleotide numbers 343-380, represented by nucleotide numbers 401-412 90% or more identical to a sequence comprising at least a part of one or more loop structures in the domain 3, or the domain 4 indicated by nucleotide numbers 473-523, or the domain corresponding to those of orthologs in other mammals Sense oligonucleotide capable of regulating expression of NF-κB p65 gene, comprising a nucleotide sequence having sex
(g) In the 3′UTR sequence of TNF-α mRNA represented by SEQ ID NO: 7, domain 1 represented by nucleotide numbers 304-405, domain 2 represented by nucleotide numbers 430-546, and nucleotide numbers 687-713 90% or more identity with a sequence comprising at least part of one or more loop structures within domain 3, or domain 4 represented by nucleotide numbers 752-776, or domains corresponding to those of orthologs in other mammals A sense oligonucleotide capable of regulating the expression of the TNF-α gene, comprising a nucleotide sequence having
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP2011107707 | 2011-05-12 | ||
| JP2011-107707 | 2011-05-12 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2012153854A1 true WO2012153854A1 (en) | 2012-11-15 |
Family
ID=47139327
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/JP2012/062230 Ceased WO2012153854A1 (en) | 2011-05-12 | 2012-05-11 | Cytokine-chemokine modulator |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2012153854A1 (en) |
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2015026249A1 (en) | 2013-08-23 | 2015-02-26 | Instytut Biochemii I Biofizyki Pan | Use of a mir172 molecule for decreasing inflammation |
Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2007142303A1 (en) * | 2006-06-08 | 2007-12-13 | Amino Up Chemical Co., Ltd. | Method for controlling the amount of gene product, and agent for controlling the amount of gene product |
| WO2007142304A1 (en) * | 2006-06-08 | 2007-12-13 | Amino Up Chemical Co., Ltd. | SENSE OLIGONUCLEOTIDE CAPABLE OF CONTROLLING THE EXPRESSION OF iNOS AND COMPOSITION COMPRISING THE SAME |
| JP2010104280A (en) * | 2008-10-29 | 2010-05-13 | Ritsumeikan | INTERFERON-alpha EXPRESSION REGULATOR |
| JP2011504110A (en) * | 2007-11-23 | 2011-02-03 | パナジェン インコーポレイテッド | MicroRNA antisense PNA, composition containing the same, and use and evaluation method thereof |
-
2012
- 2012-05-11 WO PCT/JP2012/062230 patent/WO2012153854A1/en not_active Ceased
Patent Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2007142303A1 (en) * | 2006-06-08 | 2007-12-13 | Amino Up Chemical Co., Ltd. | Method for controlling the amount of gene product, and agent for controlling the amount of gene product |
| WO2007142304A1 (en) * | 2006-06-08 | 2007-12-13 | Amino Up Chemical Co., Ltd. | SENSE OLIGONUCLEOTIDE CAPABLE OF CONTROLLING THE EXPRESSION OF iNOS AND COMPOSITION COMPRISING THE SAME |
| JP2011504110A (en) * | 2007-11-23 | 2011-02-03 | パナジェン インコーポレイテッド | MicroRNA antisense PNA, composition containing the same, and use and evaluation method thereof |
| JP2010104280A (en) * | 2008-10-29 | 2010-05-13 | Ritsumeikan | INTERFERON-alpha EXPRESSION REGULATOR |
Non-Patent Citations (12)
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2015026249A1 (en) | 2013-08-23 | 2015-02-26 | Instytut Biochemii I Biofizyki Pan | Use of a mir172 molecule for decreasing inflammation |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| Wang et al. | CircUbe3a from M2 macrophage-derived small extracellular vesicles mediates myocardial fibrosis after acute myocardial infarction | |
| Yin et al. | MicroRNA-155 is an Epstein-Barr virus-induced gene that modulates Epstein-Barr virus-regulated gene expression pathways | |
| Jung et al. | c‐Myc‐mediated overexpression of miR‐17‐92 suppresses replication of hepatitis B virus in human hepatoma cells | |
| EP2458005A1 (en) | Fgf21 cis-element binding substance | |
| EP3760234A2 (en) | Rna interference for the treatment of gain-of-function disorders | |
| US20120115924A1 (en) | Micro-RNA Mediated Modulation of Colony Stimulating Factors | |
| WO2019196887A1 (en) | Novel small activating rna | |
| JP2025023131A (en) | SiRNA sequences targeting the expression of the human genes JAK1 or JAK3 for therapeutic use | |
| US10378015B2 (en) | Targeting hepatitis B virus (HBV) host factors | |
| Yang | miR-574-5p in epigenetic regulation and Toll-like receptor signaling | |
| WO2012153854A1 (en) | Cytokine-chemokine modulator | |
| US20230138103A1 (en) | Gene silencing agents for targeting coronavirus genes and uses thereof | |
| JP2010104280A (en) | INTERFERON-alpha EXPRESSION REGULATOR | |
| US9271997B2 (en) | Regulators of NFAT and/or store-operated calcium entry | |
| JP2014236717A (en) | Inhibitors of endogenous cox-2 antisense rna and its use and screening methods therefor | |
| US8338185B2 (en) | Methods of determining potency of chemically-synthesized oligonucleotides | |
| JP5794532B2 (en) | Interferon-α modulator | |
| US8048864B1 (en) | Regulators of NFAT and/or store-operated calcium entry | |
| JP4589794B2 (en) | Osteopontin siRNA | |
| US12458658B2 (en) | Composition for inhibiting replication of hepatitis B virus | |
| JP2010263917A (en) | Osteopontin siRNA | |
| JP2008237023A (en) | Multiple shRNA expression vector | |
| KR100992239B1 (en) | New Uses of the MIB12 Gene | |
| WO2024048528A1 (en) | Prophylactic agent and/or therapeutic agent for heart diseases and laminopathy | |
| JPWO2011115271A1 (en) | Tumor angiogenesis inhibitor |
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: 12782249 Country of ref document: EP Kind code of ref document: A1 |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| 122 | Ep: pct application non-entry in european phase |
Ref document number: 12782249 Country of ref document: EP Kind code of ref document: A1 |
|
| NENP | Non-entry into the national phase |
Ref country code: JP |