WO2014024820A1 - Pharmaceutical composition for treating new chronic kidney disease and method for screening medicine for new chronic kidney disease - Google Patents

Pharmaceutical composition for treating new chronic kidney disease and method for screening medicine for new chronic kidney disease Download PDF

Info

Publication number
WO2014024820A1
WO2014024820A1 PCT/JP2013/071109 JP2013071109W WO2014024820A1 WO 2014024820 A1 WO2014024820 A1 WO 2014024820A1 JP 2013071109 W JP2013071109 W JP 2013071109W WO 2014024820 A1 WO2014024820 A1 WO 2014024820A1
Authority
WO
WIPO (PCT)
Prior art keywords
gene
expression
substance
suppresses
galnt2
Prior art date
Application number
PCT/JP2013/071109
Other languages
French (fr)
Japanese (ja)
Inventor
六嶋 正知
知世 吉永
陽子 山口
義成 我原
満 能登谷
憲人 吉村
和彦 前川
Original Assignee
塩野義製薬株式会社
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 塩野義製薬株式会社 filed Critical 塩野義製薬株式会社
Publication of WO2014024820A1 publication Critical patent/WO2014024820A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • G01N33/5023Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects on expression patterns
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/34Genitourinary disorders
    • G01N2800/347Renal failures; Glomerular diseases; Tubulointerstitial diseases, e.g. nephritic syndrome, glomerulonephritis; Renovascular diseases, e.g. renal artery occlusion, nephropathy

Definitions

  • the present invention relates to a pharmaceutical composition for treating chronic kidney disease or fibrotic disease, which contains a substance that suppresses the expression of a specific gene.
  • the present invention relates to a pharmaceutical composition for the treatment of chronic kidney disease or fibrosis including a substance that suppresses expression of GALNT2, COL8A2, FRYL or TIMP1 gene, and expression of COL8A2, GALNT2, FRYL or TIMP1 gene
  • the present invention relates to a method for screening a therapeutic substance for chronic kidney disease or fibrotic disease based on fluctuation.
  • Chronic kidney disease refers to a condition in which "finding of kidney disease such as urine protein positivity” or "decreased renal function (glomerular filtration rate is less than 60 mL / min / 1.73 m2)" continues for 3 months or more.
  • Chronic kidney disease is not only a risk of progression of end-stage renal failure, but is also a risk factor for cardiovascular events, causing myocardial infarction, heart failure, cerebral infarction and death.
  • anti-hypertensive therapies such as angiotensin receptor antagonists and calcium antagonists are being used as preventive and therapeutic methods for chronic kidney disease.
  • anti-hypertensive therapies such as angiotensin receptor antagonists and calcium antagonists are being used as preventive and therapeutic methods for chronic kidney disease.
  • the effects obtained by existing therapies cannot be said to be sufficient, and there is a demand for better preventive / therapeutic agents for renal dysfunction.
  • the pathological features of chronic kidney disease include fibrosis of glomeruli and tubulointerstitium.
  • the pathological features of end stage renal failure are markedly parenchymal cell loss and fibrosis. It is known that patients who show tubulointerstitial fibrosis in patients with chronic kidney disease progress more rapidly in renal function deterioration than patients who do not show fibrosis.
  • Fibrosis can occur in any tissue, but can progress by a common mechanism, regardless of the type of trigger for its onset.
  • structures and structures of animal tissues and organs are maintained by fibers such as collagen, but when the tissues are injured in some way, they are restored to the original tissues by a wound healing process accompanied by collagen production.
  • the tissue is subjected to multiple immunological, chemical, mechanical, metabolic, or other injuries or the extent of the injuries is large, excessive fibrous connective tissue accumulation may occur.
  • Such accumulation of connective tissue is irreversible, and if fibers increase abnormally, a fibrotic disease in which tissues and organs do not function normally is caused.
  • RNA oligonucleotide strands of about 21 bases complementary to a desired gene (mRNA) and double-stranded RNA (small RNA interfering RNA) (siRNA) consisting of the antisense RNA oligonucleotide strand have been developed, and can be arbitrarily used in mammalian cells. It became possible to suppress the expression of the gene (Non-patent Document 1).
  • siRNA and antisense oligo technologies are actively used in life science research, but there are no disease treatment siRNA or antisense oligos sold as pharmaceuticals.
  • the object of the present invention is to suppress the expression of type 1 Collagen (COL1A1) gene by suppressing the expression of a novel target gene for chronic kidney disease or fibrosis, ie, its expression, thereby causing chronic kidney disease or fibrosis
  • a novel target gene for chronic kidney disease or fibrosis ie, its expression, thereby causing chronic kidney disease or fibrosis
  • GALNT2, COL8A2 are genes that significantly reduce the expression of COL1A1 gene by suppressing the gene expression by introducing a human gene siRNA library into human tubular epithelial cell line (HK2).
  • the present invention has been completed by identifying the FRYL and TIMP1 genes.
  • a pharmaceutical composition comprising a substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene, or a substance that suppresses the activity of a protein encoded by these genes.
  • the pharmaceutical composition according to [1] wherein the substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or the substance that suppresses the activity of the protein encoded by these genes is a nucleic acid.
  • the nucleic acid is siRNA or antisense oligonucleotide or an expression vector thereof.
  • a COL1A1 gene expression inhibitor comprising a substance that suppresses expression of GALNT2, COL8A2, FRYL, or TIMP1 gene, or a substance that suppresses the activity of a protein encoded by these genes.
  • the COL1A1 gene expression inhibitor according to [4] wherein the substance that suppresses the expression of the GALNT2, COL8A2, FRYL, or TIMP1 gene or the substance that suppresses the activity of the protein encoded by these genes is a nucleic acid.
  • a pharmaceutical composition for treating a disease associated with the COL1A1 gene comprising a substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene, or a substance that suppresses the activity of a protein encoded by these genes.
  • the pharmaceutical composition according to [7] wherein the substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or the substance that suppresses the activity of the protein encoded by these genes is a nucleic acid.
  • the nucleic acid is siRNA or antisense oligonucleotide or an expression vector thereof.
  • [12] (1) A step of contacting a cell with a test substance, (2) measuring the expression level of the GALNT2, COL8A2, FRYL, or TIMP1 gene in a cell contacted with the test substance, and comparing the expression level with the expression level of the gene in a control cell not contacted with the test substance; (3) selecting the test substance as a therapeutic substance for a disease associated with the COL1A1 gene when the expression of the gene in the cell contacted with the test substance is lower than the expression of the gene in a control cell; Including, A screening method for a therapeutic agent for a disease associated with the COL1A1 gene.
  • a method for screening a therapeutic substance for a disease associated with the COL1A1 gene which comprises using a polypeptide having N-acetylgalactosamine transfer activity.
  • Contacting a polypeptide having an amino acid sequence deleted, substituted, and / or added and having N-acetylgalactosamine transfer activity (2) comparing the N-acetylgalactosamine transfer activity of the polypeptide contacted with the test substance with the N-acetylgalactosamine transfer activity of the polypeptide not contacted with the test substance, and (3) When the N-acetylgalactosamine transfer activity of the polypeptide contacted with the test substance is lower than the N-acetylgalactosamine transfer activity of the polypeptide not contacted with the test substance, the test substance is transferred to the COL1A1 gene.
  • a screening method for a therapeutic agent for a disease associated with the COL1A1 gene comprising a step of administering a substance that suppresses expression of the GALNT2, COL8A2, FRYL, or TIMP1 gene or a substance that suppresses the activity of a protein encoded by these genes.
  • a method for treating chronic kidney disease or fibrotic disease comprising a step of administering a substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or a substance that suppresses the activity of a protein encoded by these genes.
  • a substance that suppresses the expression of a GALNT2, COL8A2, FRYL, or TIMP1 gene or a protein encoded by these genes which is used for treatment of chronic kidney disease or fibrotic disease.
  • the present invention it is possible to suppress the expression of COL1A1 gene by suppressing the expression of GALNT2, COL8A2, FRYL or TIMP1 gene, and the substance which suppresses the expression of GALNT2, COL8A2, FRYL or TIMP1 gene is a chronic kidney It can be used as an active ingredient of a pharmaceutical composition for treating diseases or fibrotic diseases.
  • the pharmaceutical composition according to the present invention provides a therapeutic agent having a new mechanism of action that specifically suppresses the expression of a specific gene, and is more toxic than conventional therapeutic agents for chronic kidney disease or fibrotic disease. It is a big feature that there is little.
  • the present invention also provides a method for screening a substance for treating chronic kidney disease or fibrosis using the GALNT2, COL8A2, FRYL or TIMP1 gene as a target. *
  • the present invention provides a therapeutic drug for a disease associated with the COL1A1 gene, comprising a substance that suppresses expression of the GALNT2, COL8A2, FRYL, or TIMP1 gene, or a substance that suppresses the activity of the protein encoded by these genes.
  • a composition is provided.
  • Diseases associated with the COL1A1 gene include, for example, chronic kidney disease such as chronic renal failure, chronic nephritis, tubulointerstitial disorder, nephrotic syndrome, polycystic kidney disease, diabetic nephropathy, and nephrosclerosis Disease, and scleroderma, hypertrophic scar, scar contracture, keloid disease, collagen disease, pulmonary fibrosis, fibrosis, silicosis, myelofibrosis, renal systemic fibrosis, Crohn's disease, myocardial fibrosis, and The disease included by fibrosis diseases, such as cirrhosis, can be mentioned.
  • chronic kidney disease such as chronic renal failure, chronic nephritis, tubulointerstitial disorder, nephrotic syndrome, polycystic kidney disease, diabetic nephropathy, and nephrosclerosis Disease, and scleroderma
  • hypertrophic scar such as chronic renal failure, chronic ne
  • GALNT2, COL8A2, FRYL or TIMP1 was found as a novel target molecule for chronic kidney disease or fibrotic disease.
  • the “GALNT2 gene” means a gene encoding GALNT2 protein.
  • the base sequence of the human GALNT2 gene and the amino acid sequence of the human GALNT2 protein are known.
  • the base sequence of the human GALNT2 gene and the human GALNT2 protein amino acid sequence are registered in GenBank (GenBank Accession No. NM_004481) and published.
  • the human GALNT2 protein includes not only a protein consisting of the amino acid sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual, and consists of the amino acid sequence registered in the above-mentioned GenBank.
  • the human GALNT2 gene includes not only a gene consisting of a base sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual, for example, from a base sequence registered in the above-mentioned GenBank. And a gene in which one or several bases are deleted, substituted, and / or added, and is generated by a mutation based on polymorphism or mutation.
  • the “GALNT2 gene” is 80% or more, for example, 85% or more, 90% or more, 95% or more, 97% or more, 98% or more, 99% with respect to the base sequence registered in the above GenBank.
  • the variant which consists of a nucleotide sequence which has 99.5% or more, 99.7% or more, or 99.9% or more identity is included.
  • the identity of the base sequence can be determined using a known algorithm such as BLAST or FASTA.
  • these mutant genes encode a protein having a function equivalent to that of the protein consisting of the amino acid sequence registered in the above GenBank.
  • the GALNT2 protein belongs to the family of GalNAc transferase (GALNT) consisting of at least 20 kinds of GALNT1 to GALNT14, GALNTL1, GALNTL2, GALNTL4, GALNTL5, GALNTL6, and WBSCR17 proteins.
  • GALNT family proteins are enzymes that add N-acetylgalactosamine to serine or threonine residues of a substrate protein and are known to be involved in the first step of substrate protein glycosylation.
  • COL1A1 gene means a gene encoding COL1A1 protein.
  • the base sequence of the human COL1A1 gene and the amino acid sequence of the human COL1A1 protein are known.
  • the base sequence of the human COL1A1 gene and the human COL1A1 protein amino acid sequence are registered in GenBank (GenBank Accession No. NM_000088) and published.
  • the human COL1A1 protein includes not only a protein consisting of the amino acid sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual, and consists of the amino acid sequence registered in the above-mentioned GenBank.
  • Proteins in which one amino acid or several amino acids are deleted, substituted and / or added in the protein, which are caused by mutations based on polymorphisms or mutations are included.
  • these mutant human COL1A1 proteins have functions equivalent to those of the protein consisting of the amino acid sequence registered in the above GenBank.
  • the human COL1A1 gene includes not only a gene having a base sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual. For example, from the base sequence registered in the above-mentioned GenBank And a gene in which one or several bases are deleted, substituted, and / or added, and is generated by a mutation based on polymorphism or mutation.
  • the “COL1A1 gene” is 80% or more, for example, 85% or more, 90% or more, 95% or more, 97% or more, 98% or more, 99% with respect to the base sequence registered in the above GenBank.
  • the variant which consists of a nucleotide sequence which has 99.5% or more, 99.7% or more, or 99.9% or more identity is included.
  • the identity of the base sequence can be determined using a known algorithm such as BLAST or FASTA.
  • these mutant genes encode a protein having a function equivalent to that of the protein consisting of the amino acid sequence registered in the above GenBank.
  • an expression inhibitor of a fibrosis-related gene such as the COL1A1 gene is a therapeutic agent for chronic kidney disease or fibrosis.
  • the “ ⁇ SMA gene” means a gene encoding an ⁇ SMA protein.
  • the nucleotide sequence of the human ⁇ SMA gene and the amino acid sequence of the human ⁇ SMA protein are known.
  • the nucleotide sequence of the human ⁇ SMA gene and the amino acid sequence of the human ⁇ SMA protein are registered in GenBank (GenBank Accession No. NM_001141945) and published.
  • the human ⁇ SMA protein includes not only a protein consisting of the amino acid sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual, and consists of the amino acid sequence registered in the above-mentioned GenBank.
  • Proteins in which one amino acid or several amino acids are deleted, substituted and / or added in the protein, which are caused by mutations based on polymorphisms or mutations are included.
  • these mutant human ⁇ SMA proteins have functions equivalent to those of the protein consisting of the amino acid sequence registered in the above-described GenBank.
  • the human ⁇ SMA gene includes not only a gene consisting of a base sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual, for example, from a base sequence registered in the above-mentioned GenBank. And a gene in which one or several bases are deleted, substituted, and / or added, and is generated by a mutation based on polymorphism or mutation.
  • the “ ⁇ SMA gene” is 80% or more, for example, 85% or more, 90% or more, 95% or more, 97% or more, 98% or more, 99% with respect to the base sequence registered in the above GenBank.
  • the variant which consists of a nucleotide sequence which has 99.5% or more, 99.7% or more, or 99.9% or more identity is included.
  • the identity of the base sequence can be determined using a known algorithm such as BLAST or FASTA.
  • these mutant genes encode a protein having a function equivalent to that of the protein consisting of the amino acid sequence registered in the above GenBank.
  • ⁇ SMA is known as a marker of fibrosis progression and is known to be upregulated in chronic kidney disease and fibrosis (Histopathology, 47, 276-280, 2005, J Am Soc Nephrol 15) 1-12, 2004, and Virchows Archiv, 450, 41-50, 2007).
  • COL8A2 gene means a gene encoding COL8A2 protein.
  • the base sequence of the human COL8A2 gene and the amino acid sequence of the human COL8A2 protein are known.
  • the base sequence of the human COL8A2 gene and the human COL8A2 protein amino acid sequence are registered in GenBank (GenBank Accession No. NM_005202) and published.
  • the human COL8A2 protein includes not only a protein consisting of the amino acid sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual, and consists of the amino acid sequence registered in the above-mentioned GenBank.
  • Proteins in which one amino acid or several amino acids are deleted, substituted and / or added in the protein, which are caused by mutations based on polymorphisms or mutations are included.
  • these mutant human COL8A2 proteins have functions equivalent to those of the protein consisting of the amino acid sequence registered in the above GenBank.
  • the human COL8A2 gene includes not only a gene having a base sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual, for example, from a base sequence registered in the above-mentioned GenBank. And a gene in which one or several bases are deleted, substituted, and / or added, and is generated by a mutation based on polymorphism or mutation.
  • the “COL8A2 gene” is 80% or more, for example, 85% or more, 90% or more, 95% or more, 97% or more, 98% or more, 99% with respect to the base sequence registered in the above GenBank.
  • the variant which consists of a nucleotide sequence which has 99.5% or more, 99.7% or more, or 99.9% or more identity is included.
  • the identity of the base sequence can be determined using a known algorithm such as BLAST or FASTA.
  • these mutant genes encode a protein having a function equivalent to that of the protein consisting of the amino acid sequence registered in the above GenBank.
  • FRYL gene means a gene encoding FRYL protein.
  • the nucleotide sequence of the human FRYL gene and the amino acid sequence of the human FRYL protein are known.
  • the nucleotide sequence of the human FRYL gene and the amino acid sequence of the human FRYL protein are registered in GenBank (GenBank Accession No. NM_015030) and published.
  • the human FRYL protein includes not only a protein consisting of the amino acid sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual, and consists of the amino acid sequence registered in the above-mentioned GenBank.
  • Proteins in which one amino acid or several amino acids are deleted, substituted and / or added in the protein, which are caused by mutations based on polymorphisms or mutations are included.
  • these mutant human FRYL proteins have functions equivalent to those of the protein consisting of the amino acid sequence registered in the above-described GenBank.
  • the human FRYL gene includes not only a gene consisting of a base sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual, for example, from a base sequence registered in the above-mentioned GenBank. And a gene in which one or several bases are deleted, substituted, and / or added, and is generated by a mutation based on polymorphism or mutation.
  • the “FRYL gene” is 80% or more, for example, 85% or more, 90% or more, 95% or more, 97% or more, 98% or more, 99% with respect to the base sequence registered in the above GenBank.
  • the variant which consists of a nucleotide sequence which has 99.5% or more, 99.7% or more, or 99.9% or more identity is included.
  • the identity of the base sequence can be determined using a known algorithm such as BLAST or FASTA.
  • these mutant genes encode a protein having a function equivalent to that of the protein consisting of the amino acid sequence registered in the above GenBank.
  • the “TIMP1 gene” means a gene encoding a TIMP1 protein.
  • the base sequence of the human TIMP1 gene and the amino acid sequence of the human TIMP1 protein are known.
  • the base sequence of the human TIMP1 gene and the human TIMP1 protein amino acid sequence are registered in GenBank (GenBank Accession No. NM_003254) and published.
  • the human TIMP1 protein includes not only a protein consisting of the amino acid sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual, and consists of the amino acid sequence registered in the above-mentioned GenBank.
  • the human TIMP1 gene includes not only a gene consisting of a base sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual. For example, from the base sequence registered in the above-mentioned GenBank And a gene in which one or several bases are deleted, substituted, and / or added, and is generated by a mutation based on polymorphism or mutation.
  • the “TIMP1 gene” is 80% or more, for example, 85% or more, 90% or more, 95% or more, 97% or more, 98% or more, 99% with respect to the base sequence registered in the above GenBank.
  • the variant which consists of a nucleotide sequence which has 99.5% or more, 99.7% or more, or 99.9% or more identity is included.
  • the identity of the base sequence can be determined using a known algorithm such as BLAST or FASTA.
  • these mutant genes encode a protein having a function equivalent to that of the protein consisting of the amino acid sequence registered in the above GenBank.
  • the “substance that suppresses gene expression” refers to a substance that suppresses the transcription of mRNA of the target gene, a substance that degrades the transcribed mRNA, or a substance that suppresses the translation of the protein from the mRNA.
  • examples of such substances include siRNA, antisense oligonucleotide, microRNA, ribozyme, and expression vectors thereof. Among them, siRNA or antisense oligonucleotide or an expression vector thereof is preferable, and siRNA or antisense oligonucleotide is particularly preferable.
  • “substances that suppress gene expression” include proteins, peptides, and other small molecules.
  • the target gene is COL8A2, GALNT2, FRYL, or TIMP1 gene.
  • the “substance that suppresses the activity of the protein encoded by the gene” is not particularly limited as long as it is a substance that suppresses the function inherent to the target protein.
  • examples of such substances include antibodies and antagonists.
  • the substance that suppresses the activity of the GALNT2 protein means a substance that inhibits the activity of the GALNT2 protein imparting N-acetylgalactosamine to the serine or threonine residue of the substrate.
  • ApoC-III, ANGPTL3 and the like are known (see Proceedings of the National Academy of Sciences of the USA, Vol. 109, pages 9893-9898, 2012).
  • siRNA is an RNA molecule having a double-stranded RNA portion consisting of about 15 to about 40 bases, and cleaves the mRNA of the target gene having a sequence complementary to the antisense strand of the siRNA. And has a function of suppressing the expression of the target gene.
  • the siRNA in the present invention is an antisense consisting of a sense RNA strand consisting of a sequence homologous to a continuous RNA sequence in mRNA of COL8A2, GALNT2, FRYL or TIMP1 gene, and a sequence complementary to the sense RNA sequence.
  • the length of the double-stranded RNA portion is about 15 to about 40 bases, preferably 15 to 30 bases, more preferably 15 to 25 bases, still more preferably 18 to 23 bases, and most preferably 19 to 21 bases as a base. It is.
  • the end structure of the sense strand or antisense strand of siRNA is not particularly limited and may be appropriately selected depending on the intended purpose. For example, it may have a blunt end or a protruding end (overhang) It is preferable that the 3 ′ end protrudes.
  • the siRNA having an overhang consisting of several bases, preferably 1 to 3 bases, more preferably 2 bases, at the 3 ′ end of the sense RNA strand and the antisense RNA strand suppresses the expression of the target gene. In many cases, the effect is large, which is preferable.
  • the type of the overhanging base is not particularly limited, and may be either a base constituting RNA or a base constituting DNA.
  • siRNA in which 1 to several nucleotides are deleted, substituted, inserted and / or added in one or both of the sense strand or the antisense strand of the siRNA is also used in the chronic kidney disease or fibrosis of the present invention. It can be used in a pharmaceutical composition for treating a disease.
  • the 1 to several bases are not particularly limited, but are preferably 1 to 4 bases, more preferably 1 to 3 bases, and most preferably 1 to 2 bases.
  • Such mutations include those in which the number of bases in the overhang portion at the 3 ′ end is 0 to 3, or the base sequence in the overhang portion at the 3 ′ end is changed to another base sequence, or Those in which the length of the sense RNA strand differs from that of the antisense RNA strand by 1 to 3 bases due to insertion, addition or deletion, or in which the base is replaced with another base in the sense strand and / or antisense strand For example, but not limited to. However, it is necessary that the sense strand and the antisense strand can hybridize in these mutant siRNAs, and that these mutant siRNAs have approximately the same level of gene expression suppression ability as siRNA having no mutation.
  • the siRNA may be a molecule having a closed structure at one end, for example, an siRNA having a hairpin structure (Short hairpin RNA; shRNA).
  • shRNA is a RNA comprising a sense strand RNA of a specific sequence of a target gene, an antisense strand RNA consisting of a sequence complementary to the sense strand RNA, and a linker sequence connecting both strands. The sense strand portion and the antisense strand portion Hybridize to form a double stranded RNA portion.
  • siRNA does not show a so-called off-target effect in clinical use.
  • the off-target effect refers to the action of suppressing the expression of another gene that is partially homologous to the siRNA used in addition to the target gene.
  • NCBI National Center for Biotechnology Information
  • RNA of the present invention In order to produce the siRNA of the present invention, a known method such as a method using chemical synthesis or a method using a gene recombination technique can be appropriately used.
  • double-stranded RNA can be synthesized by a conventional method based on sequence information.
  • an expression vector incorporating a sense strand sequence or an antisense strand sequence is constructed, and the sense strand RNA or antisense strand RNA generated by transcription after introducing the vector into a host cell. It can also be produced by acquiring each of the above.
  • Desired double-stranded RNA can also be prepared.
  • a part of the nucleic acid constituting the siRNA may be DNA.
  • the siRNA may be a nucleic acid in which all or part of the nucleic acid constituting the siRNA is modified as long as it has the activity of suppressing the expression of the target gene.
  • the modified nucleic acid means a nucleic acid having a structure different from that of a natural nucleic acid, in which a nucleoside (base site, sugar site) and / or internucleoside binding site is modified.
  • modified nucleoside constituting the modified nucleic acid
  • examples of the “modified nucleoside” constituting the modified nucleic acid include an abasic nucleoside; an arabino nucleoside, 2′-deoxyuridine, ⁇ -deoxyribonucleoside, ⁇ -L-deoxyribonucleoside, and other sugars
  • examples include nucleosides having modifications; peptide nucleic acids (PNA), peptide nucleic acids to which phosphate groups are bound (PHONA), locked nucleic acids (LNA), morpholino nucleic acids and the like.
  • nucleoside having a sugar modification examples include substituted pentasaccharides such as 2′-O-methylribose, 2′-deoxy-2′-fluororibose, and 3′-O-methylribose; 1 ′, 2′-deoxyribose Arabinose; substituted arabinose sugars; nucleosides with hexose and alpha-anomeric sugar modifications.
  • These nucleosides may be modified bases with modified base sites. Examples of such modified bases include pyrimidines such as 5-hydroxycytosine, 5-fluorouracil, 4-thiouracil; purines such as 6-methyladenine and 6-thioguanosine; and other heterocyclic bases.
  • modified internucleoside linkage constituting the modified nucleic acid
  • examples of the “modified internucleoside linkage” constituting the modified nucleic acid include, for example, alkyl linker, glyceryl linker, amino linker, poly (ethylene glycol) linkage, methylphosphonate internucleoside linkage; methylphosphonothioate, phosphotriester , Phosphothiotriester, phosphorothioate, phosphorodithioate, triester prodrug, sulfone, sulfonamide, sulfamate, formacetal, N-methylhydroxylamine, carbonate, carbamate, morpholino, boranophosphonate, phosphoramidate, etc.
  • Non-natural internucleoside linkages include, for example, alkyl linker, glyceryl linker, amino linker, poly (ethylene glycol) linkage, methylphosphonate internucleoside linkage; methylphosphonothioate
  • the sequences described in SEQ ID NOs: 2 to 25 in the sequence listing can be preferably used.
  • the nucleotide sequences of these siRNAs are shown in Table 1.
  • the uppercase letters indicate the sense RNA sequence and the antisense RNA sequence
  • the lowercase letters indicate the 3 ′ end overhang sequence.
  • the first sequence in Table 1 is a double-stranded siRNA composed of a sense strand shown in SEQ ID NO: 2 and an antisense strand shown in SEQ ID NO: 3, and tt at the 3 ′ end of SEQ ID NO: 2
  • ca at the 3 ′ end of SEQ ID NO: 3 is an overhang sequence. Since these siRNAs remarkably suppress the expression of the COL1A1 gene, the effect of the pharmaceutical composition for treating a disease associated with the COL1A1 gene containing these siRNAs is great.
  • Oligonucleotides complementary to the mRNA of the target gene are called “antisense oligonucleotides,” and the mRNA functions are suppressed by forming a double strand with the target gene (mRNA). .
  • Antisense oligonucleotides are not limited to those that are completely complementary to the target gene (mRNA), but may contain some mismatches as long as they can be stably hybridized with mRNA.
  • Antisense oligonucleotides may be modified. By applying an appropriate modification, the antisense oligonucleotide becomes difficult to be degraded in the living body, and the expression of the target gene can be inhibited more stably.
  • modified oligonucleotides include S-oligo type (phosphorothioate type), C-5 thiazole type, D-oligo type (phosphodiester type), M-oligo type (methyl phosphonate type), peptide nucleic acid
  • modified antisense oligonucleotides such as phosphodiester bond type, C-5 propynyl pyrimidine type, 2-O-propyl ribose, 2′-methoxyethoxy ribose type.
  • the antisense oligonucleotide may be one in which at least a part of the oxygen atom constituting the phosphate group is substituted or modified with a sulfur atom.
  • Such an antisense oligonucleotide is particularly excellent in nuclease resistance and affinity for RNA.
  • Examples of the antisense oligonucleotide in which at least a part of the oxygen atom constituting the phosphate group is substituted or modified with a sulfur atom include oligonucleotides such as S-oligo type.
  • An antisense oligonucleotide (or a derivative thereof) can be synthesized by a conventional method, and can be easily synthesized by, for example, a commercially available DNA synthesizer (for example, Applied Biosystems).
  • Examples of the synthesis method include a solid phase synthesis method using phosphoramidite and a solid phase synthesis method using hydrogen phosphonate.
  • RNA having an enzyme activity that cleaves nucleic acid refers to RNA having an enzyme activity that cleaves nucleic acid. Recently, it has been clarified that oligoDNA having the base sequence of the enzyme active site also has a nucleic acid cleaving activity. In the book, it is used as a concept including DNA as long as it has sequence-specific nucleic acid cleavage activity. Specifically, the ribozyme can specifically cleave mRNA or an initial transcription product encoding a target gene within the coding region (including an intron portion in the case of the initial transcription product). The most versatile ribozyme is self-splicing RNA found in infectious RNA such as viroid and virusoid, and hammerhead type and hairpin type are known.
  • the hammerhead type exhibits enzyme activity at about 40 bases, and several bases at both ends adjacent to the portion having the hammerhead structure (about 10 bases in total) are made complementary to the desired cleavage site of mRNA. By doing so, it is possible to specifically cleave only the target mRNA. Furthermore, when the ribozyme is used in the form of an expression vector containing the DNA encoding the ribozyme, in order to promote the transfer of the transcription product to the cytoplasm, it should be a hybrid ribozyme further linked with a tRNA-modified sequence. (Nucleic Acids Res., 29 (13): 2780-2788 (2001)).
  • Micro RNA is a small RNA that does not encode a protein, and micro RNA is a molecule having a function of controlling gene expression by binding to a complementary site on a target mRNA.
  • the substance that suppresses the expression of the target gene may be a nucleic acid molecule such as siRNA, antisense oligonucleotide, microRNA or ribozyme and an expression vector encoding the nucleic acid molecule.
  • the oligonucleotide or polynucleotide encoding the nucleic acid molecule must be operably linked to a promoter capable of exhibiting promoter activity in a mammalian cell to be administered.
  • the promoter to be used is not particularly limited as long as it can function in the mammal to be administered, but for example, polIII promoter (eg, tRNA promoter, U6 promoter, H1 promoter), mammalian promoter (eg, CMV promoter, CAG promoter, SV40 promoter) and the like.
  • the expression vector preferably contains a transcription termination signal, ie a terminator region, downstream of the oligo (poly) nucleotide encoding the nucleic acid molecule.
  • selectable marker genes for selection of transformed cells such as genes that confer resistance to drugs such as tetracycline, ampicillin, kanamycin, hygromycin, phosphinothricin, genes that complement auxotrophic mutations, etc.
  • the basic backbone vector used as the expression vector is not particularly limited, and examples thereof include a plasmid vector and a viral vector.
  • Suitable vectors for administration to mammals such as humans include viral vectors such as retrovirus, adenovirus, adeno-associated virus, herpes virus, vaccinia virus, poxvirus, poliovirus, Sindbis virus, Sendai virus, and the like. .
  • Examples of the expression vector of the present invention include siRNA, antisense oligonucleotide, microRNA, and ribozyme expression vectors. Among them, siRNA or antisense oligonucleotide expression vectors are preferable.
  • the nucleic acid sequence encoded by the expression vector for siRNA of the present invention is preferably the sequence shown in Table 1. Since the siRNA shown in Table 1 remarkably suppresses the expression of the COL1A1 gene, the effect of the pharmaceutical composition for treating a disease associated with the COL1A1 gene containing these siRNAs is great.
  • a substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or a substance that suppresses the activity of the protein encoded by these genes can be used as an active ingredient of a pharmaceutical composition.
  • the pharmaceutical composition of the present invention can be used as a pharmaceutical composition for treating a disease associated with the COL1A1 gene by administering the pharmaceutical composition in vivo.
  • a single gene expression inhibitor may be used as an active ingredient, or a plurality of gene expression inhibitors may be used as active ingredients.
  • the expression inhibitory substance of the pharmaceutical composition of the present invention is siRNA
  • one or more siRNAs may be used as the active ingredient.
  • the type of disease that is the subject of treatment with the pharmaceutical composition of the present invention is a disease associated with the COL1A1 gene.
  • diseases associated with the COL1A1 gene include chronic kidney disease such as chronic renal failure, chronic nephritis, tubulointerstitial disorder, nephrotic syndrome, polycystic kidney disease, diabetic nephropathy, and nephrosclerosis , And scleroderma, hypertrophic scar, scar contracture, keloidosis, collagen disease, pulmonary fibrosis, fibrosis, silicosis, myelofibrosis, renal systemic fibrosis, Crohn's disease, myocardial fibrosis, and cirrhosis Diseases included in fibrotic diseases such as
  • chronic kidney disease is a condition in which “finding of kidney disease such as urine protein positivity” or “reduced renal function (glomerular filtration rate less than 60 mL / min / 1.73 m2)” continues for 3 months or more
  • the pharmaceutical composition of the present invention can employ both oral and parenteral dosage forms.
  • the pharmaceutical composition of the present invention can be formulated according to a conventional method, and may contain a pharmaceutically acceptable carrier or additive.
  • Such carriers and additives include water, pharmaceutically acceptable organic solvents, collagen, polyvinyl alcohol, polyvinyl pyrrolidone, carboxyvinyl polymer, sodium carboxymethylcellulose, sodium polyacrylate, sodium alginate, water-soluble dextran, carboxymethyl.
  • additives examples include additives.
  • the additive is selected from the above alone or in appropriate combination depending on the dosage form of the pharmaceutical composition of the present invention.
  • the dosage form in the case of oral administration, it can be administered as a tablet, capsule, fine granule, powder, granule, liquid, syrup or the like, or in an appropriate dosage form.
  • pulmonary dosage forms for example, those using a nephriser etc.
  • nasal dosage forms for example, transdermal dosage forms (for example, ointments, creams), injection dosage forms and the like
  • injection dosage form it can be administered systemically or locally by intravenous injection such as infusion, intramuscular injection, intraperitoneal injection, subcutaneous injection or the like.
  • the substance that suppresses the expression of the target gene is a nucleic acid molecule such as siRNA, antisense oligonucleotide, microRNA or ribozyme, or an expression vector encoding the nucleic acid molecule
  • the expression is suppressed in phospholipid vesicles such as liposomes. It is also possible to introduce a substance and use the endoplasmic reticulum as the pharmaceutical composition of the present invention.
  • the dosage of the pharmaceutical composition of the present invention varies depending on age, sex, symptoms, administration route, administration frequency, and dosage form.
  • the administration method is appropriately selected depending on the age and symptoms of the patient.
  • the effective dose is 0.01 ⁇ g to 1000 mg, preferably 0.1 ⁇ g to 100 ⁇ g, per kg body weight.
  • the therapeutic agent is not limited to these doses.
  • a method for treating a disease associated with the COL1A1 gene comprising a step of administering a substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or a substance that suppresses the activity of a protein encoded by these genes, (2) A substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or the activity of a protein encoded by these genes, which is used for treatment of a disease associated with COL1A1 gene, (3) Use of a substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or a substance that suppresses the activity of a protein encoded by these genes for the manufacture of a therapeutic agent for a disease associated with the COL1A1 gene, (4) a method for treating chronic kidney disease or fibrotic disease, comprising a step of administering a substance that suppresses the expression of GALNT2, COL8A
  • the present invention relates to a method for screening a therapeutic agent for a disease associated with the COL1A1 gene, which is based on suppression of expression of the GALNT2, COL8A2, FRYL, or TIMP1 gene, or suppression of the activity of a protein encoded by these genes. I will provide a.
  • test substance to be subjected to the screening method may be any known compound and novel compound, for example, nucleic acid, carbohydrate, lipid, protein, peptide, low molecular organic compound, compound prepared using combinatorial chemistry technology
  • libraries random peptide libraries prepared by solid phase synthesis and phage display methods, or natural components derived from microorganisms, animals and plants, marine organisms, and the like.
  • the screening method of the present invention comprises: (1) a step of bringing cells into contact with a test substance; (2) measuring the expression level of the GALNT2, COL8A2, FRYL, or TIMP1 gene in a cell contacted with the test substance, and comparing the expression level with the expression level of the gene in a control cell not contacted with the test substance; (3) selecting the test substance as a therapeutic substance for a disease associated with the COL1A1 gene when the expression of the gene in the cell contacted with the test substance is lower than the expression of the gene in a control cell; Including, This is a screening method for a therapeutic substance for a disease associated with the COL1A1 gene.
  • the cell in step (1) of the above method means a cell capable of measuring the expression of GALNT2, COL8A2, FRYL or TIMP1 gene.
  • cells and a test substance are placed in contact with each other in a culture medium.
  • Cells capable of measuring the expression of GALNT2, COL8A2, FRYL or TIMP1 gene are directly or indirectly evaluated for the expression level of GALNT2, COL8A2, FRYL or TIMP1 gene products such as transcripts and translation products.
  • a cell capable of directly evaluating the expression level of the gene product can be a cell capable of naturally expressing the gene, while a cell capable of indirectly evaluating the expression level of the gene product includes: Examples include cells that allow reporter assay for the gene transcription regulatory region.
  • animal cells such as mice, rats, hamsters, guinea pigs, rabbits, dogs, monkeys or human mammalian cells can be used.
  • Human-derived cells, particularly human urine Tubular epithelial cell line HK2 cells and human primary tubular epithelial cells are preferred.
  • a cell enabling a reporter assay for a gene transcription regulatory region is a cell containing a target gene transcription regulatory region and a reporter gene operably linked to the region.
  • the target gene transcription regulatory region and the reporter gene can be inserted into an expression vector.
  • the transcriptional regulatory region of the target gene is not particularly limited as long as it can control the expression of the target gene. For example, a region from the transcription start point to about 2 kbp upstream, or one or more bases in the base sequence of the region Examples include a region consisting of a base sequence deleted, substituted or added and having the ability to control the transcription of the target gene.
  • the reporter gene may be any gene that encodes a detectable protein or an enzyme that produces a detectable substance.
  • the GFP green fluorescent protein
  • GUS ⁇ -glucuronidase
  • LUC luciferase
  • CAT Chloramphenicol acetyltransferase
  • a cell into which a gene transcription regulatory region and a reporter gene operably linked to the region are introduced can be used for quantitative analysis of the expression level of the reporter gene as long as the transcriptional regulatory function of the target gene can be evaluated. As long as it is, it is not particularly limited.
  • the culture medium in which the cells and the test substance are brought into contact is appropriately selected depending on the type of cells used.
  • a minimal essential medium (MEM) containing about 5 to 20% fetal bovine serum, Dulbecco's modification Minimum essential medium (DMEM), RPMI 1640 medium, 199 medium, and the like.
  • the culture conditions are also appropriately determined according to the type of cells to be used.
  • the pH of the medium is about 6 to about 8
  • the culture temperature is usually about 30 to about 40 ° C.
  • the culture time is About 12 to about 144 hours.
  • step (2) of the above method first, the expression level of the GALNT2, COL8A2, FRYL, or TIMP1 gene in the cells contacted with the test substance is measured.
  • the expression level can be measured by a method known per se in consideration of the type of cells used. For example, when a cell capable of naturally expressing the GALNT2, COL8A2, FRYL or TIMP1 gene is used as a cell capable of measuring the expression of the gene, the expression level is a gene product such as a transcription product or a translation product. Can be measured by a method known per se.
  • the expression level of the transcript can be measured by preparing total RNA from cells and performing RT-PCR, Northern blotting, or the like.
  • the expression level of the translation product can be measured by preparing an extract from the cells and using an immunological technique.
  • an immunological technique a radioisotope immunoassay (RIA method), an ELISA method (Methods in Enzymol. 70: 419-439 (1980)), a fluorescent antibody method, or the like can be used.
  • a cell capable of measuring a gene transcription regulatory region is used as a cell capable of measuring the expression of GALNT2, COL8A2, FRYL or TIMP1 gene
  • the expression level can be measured based on the signal intensity of the reporter.
  • step (3) of the above method the expression level of the gene in the cell contacted with the test substance is compared with the expression level of the gene in the control cell.
  • the comparison of expression levels is preferably performed based on the presence or absence of a significant difference.
  • the expression level of the gene in the control cell not contacted with the test substance is the expression level measured at the same time, even if it is the expression level measured in advance compared to the measurement of the gene expression level in the cell contacted with the test substance.
  • the expression level is preferably measured simultaneously from the viewpoint of the accuracy and reproducibility of the experiment.
  • the screening method of the present invention comprises: A polypeptide having the amino acid sequence represented by SEQ ID NO: 1, or an amino acid sequence in which one or several amino acids of the amino acid sequence represented by SEQ ID NO: 1 are deleted, substituted, and / or added; A method for screening a therapeutic substance for a disease associated with the COL1A1 gene, which comprises using a polypeptide having acetylgalactosamine transfer activity.
  • the screening method of the present invention comprises: (1) a substrate for GALNT2 protein, a test substance and 1: a polypeptide having the amino acid sequence represented by SEQ ID NO: 1, or 2: deletion of one or several amino acids of the amino acid sequence represented by SEQ ID NO: 1, Contacting a polypeptide having a substituted and / or added amino acid sequence and having N-acetylgalactosamine transfer activity; (2) a step of comparing the N-acetylgalactosamine transfer activity of the polypeptide contacted with the test substance with the N-acetylgalactosamine transfer activity of the polypeptide not contacted with the test substance, and (3) a poly contacted with the test substance.
  • the test substance is selected as a therapeutic agent for a disease associated with the COL1A1 gene Process.
  • the protein that can be used as a substrate for the GALNT2 protein in step (1) of the above method is as described above.
  • the polypeptide having the amino acid sequence represented by SEQ ID NO: 1 includes an isolated polypeptide, a polypeptide in a membrane fraction, and an intracellular polypeptide.
  • a screening target gene a group of genes that are highly likely to be drug discovery targets was narrowed down by expression information from all human genes. Specifically, in the expression database of 27 human normal tissues, a group of genes whose relative expression level is below the reference value in all of the heart, brain, spleen, and peripheral leukocytes is extracted as genes with relatively few side effects, and 10,723 genes Got. Next, in order to reduce the probability of false positives, total RNA is extracted from the human tubular epithelial cell line HK2 used for primary screening, and DNA microarray analysis is performed to remove genes whose levels cannot be detected in the same cells. This narrowed the target to 7,700 genes. From the above 7,700 genes, 1,994 genes were selected that are likely to be able to regulate activity with low molecular weight compounds and targets that are likely to be high molecular drugs.
  • the following experiment was performed as a primary screening. From the human whole gene siRNA library (Silencer Select Human Genome siRNA Library; manufactured by Ambion; 21,585 gene; 64,755 siRNA; 3 siRNA / gene), the focused siRNA library (5,982 siRNAs) of the above 1,994 gene was prepared by cherry picking . Using the focused siRNA library, we searched for a gene whose expression of COL1A1 gene is reduced by suppressing expression in human tubular epithelial cell line HK2. As a screening protocol, first, 1.5 ⁇ l (0.375 pmol, final 12.5 nM) of 0.25 ⁇ M library siRNA was spotted on a 384 well plate (Corning) coated with Poly-D-Lysine (PDL).
  • PDL Poly-D-Lysine
  • Negative Control # 1 siRNA (NT-1; manufactured by Ambion) was dispensed in the same amount on each plate as a negative control, and used for hit criteria setting. Meanwhile, Opti-MEM I (Gibco) 3.5 ⁇ l and siRNA introduction reagent RNAiMAX (Invitrogen) 0.1 ⁇ l were mixed, kept at room temperature for 5 minutes, and then 384-well plate dispensed with the above siRNA Added to.
  • Keratinocyte Serum Free Media Invitrogen, 0.05 mg / ml bovine pituitary extract and 5 ng / ml human rEGF
  • the cells were cultured for 48 hours in a 5% CO2 environment.
  • 30 ⁇ l of Keratinocyte Serum Free Media containing recombinant TGF ⁇ manufactured by R & D
  • TGF ⁇ recombinant TGF ⁇
  • the culture supernatant was removed by decantation, and 25 ⁇ l of Buffer FCW (Wash buffer) contained in FastLane Cell SYBR Green Kit (manufactured by Qiagen) was added. After removing Buffer FCW by decantation, 15 ⁇ l of Cell Processing Mix (Lysis buffer) also contained in Kit was added and kept at room temperature for 5 minutes to lyse the cells. A cell lysate heated at 75 ° C. for 5 minutes was used as a template for quantitative PCR. For quantitative PCR, FastLane Cell SYBR Green Kit was used, and the expression levels of COL1A1 gene as fibrosis-related gene and GAPDH gene and PPIA gene as internal standards were measured.
  • PCR primers used to amplify each gene were purchased from Takara Bio Inc. (catalog number; 5060D-40407, primer set ID; COL1A1, CH000497; GAPDH, HA067812; PPIA, HA067810).
  • a reaction solution was prepared, and quantitative PCR was performed using 7900HT Fast Real Time PCR System (Applied Biosystems).
  • Cycle threshold value (Ct value)
  • the expression of COL1A1 gene is reduced to 50% or less compared to the negative control siRNA on the same plate by either GAPDH gene correction or PPIA gene correction.
  • SiRNAs were extracted as hit candidate siRNAs.
  • 158 genes were obtained by using, as a primary screening hit candidate, a gene in which two or more of the three sequences of siRNAs for the same gene were hit candidates.
  • a focused siRNA library of the above 158 genes was prepared (474 siRNA, 3 siRNAs / gene), and the degree of expression reduction of the COL1A1 gene when each gene was suppressed by siRNA was evaluated using the same protocol as the primary screening.
  • the reproducibility of four genes, COL8A2, GALNT2, FRYL, and TIMP1 was confirmed, and the gene group was used as a primary screening hit gene.
  • the results of the primary screening reproducibility test are listed in Table 2.
  • GALNT2 which is considered to be the most promising target gene from the suppression data of COL1A1 gene expression, was evaluated from the second order onward. Specifically, in the secondary evaluation, the influence of other fibrosis markers on the expression of alpha-smooth muscle actin ( ⁇ SMA, ACTA2) protein was examined by Western blot.
  • siRNA used the same sequence as the primary screening (3 siRNA / gene), and Negative Control # 1 siRNA was used as a negative control.
  • RNAiMAX 56.75 ⁇ l of Opti-MEM I and 1.75 ⁇ l of RNAiMAX were mixed and kept at room temperature for 5 minutes, then mixed with 25 ⁇ l of 0.1 ⁇ M siRNA (final 5 nM) and kept at room temperature for another 20 minutes.
  • This solution was transferred to a 24-well plate, 416.5 ⁇ l of Keratinocyte Serum Free Media containing 50,000 HK2 cells was added to each well, and cultured for 48 hours at 37 ° C. in a 5% CO 2 environment. Next, 500 ⁇ l of Keratinocyte Serum Free Media containing 2 ng / ml recombinant TGF ⁇ was added by medium exchange, and further cultured for 24 hours.
  • the cells were washed with PBS, and the cells were lysed with 70 ⁇ l of RIPA buffer. After centrifuging the cell lysate, the protein concentration of the supernatant was measured, and 6 ⁇ g of total protein was subjected to SDS polyacrylamide gel electrophoresis. After electrophoresis, it was transferred to a PVDF membrane, blocked using Blocking One (manufactured by Nacalai Tesque), and then reacted with a primary antibody (mouse anti- ⁇ SMA antibody, manufactured by Sigma) at 6.5 ° C. overnight.
  • a primary antibody manufactured by Sigma
  • RPTEC human primary tubule epithelial cells
  • RPTEC Human primary tubule epithelial cells
  • NT-2 Negative Control # 2 siRNA
  • RNAiMAX RNAiMAX
  • Opti-MEM I and 0.7 ⁇ l of RNAiMAX were mixed and kept at room temperature for 5 minutes, then mixed with 30 ⁇ l of 0.1 ⁇ M siRNA (final 5 nM) and kept at room temperature for another 20 minutes.
  • This solution was transferred to a 24-well plate, 500 ⁇ l of REGM Bullet Kit (manufactured by LONZA) containing 75,000 RPTECs was added to each well, and cultured in a 37 ° C., 5% CO 2 environment.
  • siRNA # 2, # 3 Two types of siRNA (siRNA # 2, # 3) against GALNT2 in which COL1A1 expression-suppressing action was observed were introduced into HK2 cells, and after 48 hours, recombinant TGF ⁇ was added. Further, 24 hours later, total RNA was collected from the cells, and microarray data was obtained. Compared to the negative control siRNA, GALNT2 was associated with multiple fibrosis because COL4A2, P4HA2, SNAI1, etc. were included in addition to COL1A1 as a result of extracting genes that commonly decrease expression in two types of siRNAs against GALNT2. It became clear that the expression of the factor was controlled.
  • the present invention can be used in the field of pharmaceuticals, particularly in the field of development and production of therapeutic agents for chronic kidney disease or fibrotic diseases.

Abstract

The present invention relates to use of the GALNT2, COL8A2, FRYL or TIMP1 genes as a target for treating chronic kidney diseases or fibrotic diseases, a therapeutic agent for chronic kidney diseases or fibrotic diseases that targets the GALNT2, COL8A2, FRYL or TIMP1 genes, and a method for screening the therapeutic agent for chronic kidney diseases or fibrotic diseases that targets the GALNT2, COL8A2, FRYL or TIMP1 genes.

Description

新規慢性腎臓病治療用医薬組成物及び新規慢性腎臓病治療薬のスクリーニング方法Novel pharmaceutical composition for treating chronic kidney disease and screening method for novel therapeutic agent for chronic kidney disease
 本発明は、特定の遺伝子の発現を抑制する物質を含む慢性腎臓病又は線維化疾患の治療用医薬組成物に関する。具体的には、本発明は、GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質を含む慢性腎臓病又は線維化疾患の治療用医薬組成物、並びにCOL8A2、GALNT2、FRYL又はTIMP1遺伝子の発現変動に基づく慢性腎臓病又は線維化疾患の治療物質のスクリーニング方法、に関する。 The present invention relates to a pharmaceutical composition for treating chronic kidney disease or fibrotic disease, which contains a substance that suppresses the expression of a specific gene. Specifically, the present invention relates to a pharmaceutical composition for the treatment of chronic kidney disease or fibrosis including a substance that suppresses expression of GALNT2, COL8A2, FRYL or TIMP1 gene, and expression of COL8A2, GALNT2, FRYL or TIMP1 gene The present invention relates to a method for screening a therapeutic substance for chronic kidney disease or fibrotic disease based on fluctuation.
 腎機能障害を有する患者が世界的に増え続けており、透析医療を受ける末期腎不全患者数の増加は医療経済上の問題となっている。近年、慢性腎臓病(chronic kidney disease:CKD)という疾患概念が提唱され、その予防・治療の重要性に対する認識が高まっている。慢性腎臓病とは、「尿蛋白陽性などの腎疾患の存在する所見」、もしくは「腎機能低下(糸球体濾過量が60mL/min/1.73m2未満)」が3ヵ月以上続く状態を指す。 The number of patients with renal dysfunction continues to increase worldwide, and the increase in the number of end-stage renal failure patients receiving dialysis treatment is a medical economic problem. In recent years, the concept of chronic kidney disease (CKD) has been advocated, and awareness of the importance of prevention and treatment is increasing. Chronic kidney disease refers to a condition in which "finding of kidney disease such as urine protein positivity" or "decreased renal function (glomerular filtration rate is less than 60 mL / min / 1.73 m2)" continues for 3 months or more.
 慢性腎臓病の発症・進展に、高血圧、糖尿病、脂質代謝異常、喫煙などが危険因子となっており、ライフスタイルの変化によるメタボリックシンドロームの広がりから、今後も患者数の伸びが危惧されている。慢性腎臓病は、末期腎不全の進行リスクであるのみならず、心血管イベントの危険因子であり、心筋梗塞、心不全、脳梗塞および死亡の原因になっている。 Hypertension, diabetes, abnormal lipid metabolism, smoking, etc. are risk factors for the onset and progression of chronic kidney disease, and the spread of metabolic syndrome due to lifestyle changes is causing a concern that the number of patients will continue to grow. Chronic kidney disease is not only a risk of progression of end-stage renal failure, but is also a risk factor for cardiovascular events, causing myocardial infarction, heart failure, cerebral infarction and death.
 慢性腎臓病の予防・治療法としては、生活指導・食事指導に加えて、アンギオテンシン受容体拮抗薬やカルシウム拮抗薬などの降圧療法の治療が行われている。しかし、既存の治療法によって得られる効果は十分とは言えず、より優れた腎機能障害の予防・治療剤が求められている。 In addition to lifestyle guidance and dietary guidance, anti-hypertensive therapies such as angiotensin receptor antagonists and calcium antagonists are being used as preventive and therapeutic methods for chronic kidney disease. However, the effects obtained by existing therapies cannot be said to be sufficient, and there is a demand for better preventive / therapeutic agents for renal dysfunction.
 慢性腎臓病の病理学的特徴として糸球体や尿細管間質の線維化が挙げられる。末期腎不全の病理像は実質細胞の脱落と線維化が顕著である。慢性腎臓病患者において尿細管間質の線維化を示す患者は、線維化を示さない患者と比較してより腎機能悪化の進行が早いことが知られている。 The pathological features of chronic kidney disease include fibrosis of glomeruli and tubulointerstitium. The pathological features of end stage renal failure are markedly parenchymal cell loss and fibrosis. It is known that patients who show tubulointerstitial fibrosis in patients with chronic kidney disease progress more rapidly in renal function deterioration than patients who do not show fibrosis.
 線維化はあらゆる組織で起こりうるが、その発症の引き金の種類に関わらず、共通した機序で進行しうる。
 一方、動物の組織や臓器は、コラーゲン等の繊維により構造が維持されているが、組織が何らかの傷害を受けると、コラーゲン産生を伴う創傷治癒の過程により元の組織に修復される。しかしながら、組織が免疫的、化学的、機械的、代謝的、あるいはその他の傷害を複数回にわたって受けたり、その傷害の程度が大きいと、過剰な線維性結合組織の蓄積が生じる場合がある。このような結合組織の蓄積は不可逆的であり、繊維が異常に増えてしまうと、組織や臓器が正常な機能を果たさなくなる線維化疾患が引き起こされる。
Fibrosis can occur in any tissue, but can progress by a common mechanism, regardless of the type of trigger for its onset.
On the other hand, structures and structures of animal tissues and organs are maintained by fibers such as collagen, but when the tissues are injured in some way, they are restored to the original tissues by a wound healing process accompanied by collagen production. However, if the tissue is subjected to multiple immunological, chemical, mechanical, metabolic, or other injuries or the extent of the injuries is large, excessive fibrous connective tissue accumulation may occur. Such accumulation of connective tissue is irreversible, and if fibers increase abnormally, a fibrotic disease in which tissues and organs do not function normally is caused.
 近年、所望の遺伝子(mRNA)に相補する約21塩基程度のRNAオリゴヌクレオチド鎖及びそのアンチセンスRNAオリゴヌクレオチド鎖からなる二本鎖RNA(small interfering RNA;siRNA)が開発され、哺乳類細胞においても任意の遺伝子の発現を抑制する事が可能となった(非特許文献1)。現在、siRNAやアンチセンスオリゴの技術は、ライフサイエンスの研究において盛んに利用されているが、医薬品として販売されている疾患治療用siRNAやアンチセンスオリゴはまだ存在しない。 In recent years, RNA oligonucleotide strands of about 21 bases complementary to a desired gene (mRNA) and double-stranded RNA (small RNA interfering RNA) (siRNA) consisting of the antisense RNA oligonucleotide strand have been developed, and can be arbitrarily used in mammalian cells. It became possible to suppress the expression of the gene (Non-patent Document 1). Currently, siRNA and antisense oligo technologies are actively used in life science research, but there are no disease treatment siRNA or antisense oligos sold as pharmaceuticals.
 本発明の目的は、新規な慢性腎臓病又は線維化疾患のターゲット遺伝子、すなわちその発現を抑制することにより、1型Collagen(COL1A1)遺伝子の発現を抑制することで、慢性腎臓病又は線維化疾患の治療を可能とする遺伝子を見出して、慢性腎臓病又は線維化疾患の治療用医薬組成物を提供することである。 The object of the present invention is to suppress the expression of type 1 Collagen (COL1A1) gene by suppressing the expression of a novel target gene for chronic kidney disease or fibrosis, ie, its expression, thereby causing chronic kidney disease or fibrosis To provide a pharmaceutical composition for the treatment of chronic kidney disease or fibrotic disease.
 本発明者等は、ヒト尿細管上皮細胞株(HK2)に、ヒトの遺伝子のsiRNAライブラリーを導入することによる遺伝子発現の抑制により、COL1A1遺伝子の発現を顕著に減少させる遺伝子であるGALNT2、COL8A2、FRYL及びTIMP1遺伝子を同定することによって、本発明を完成するに至った。 The present inventors have introduced GALNT2, COL8A2 which are genes that significantly reduce the expression of COL1A1 gene by suppressing the gene expression by introducing a human gene siRNA library into human tubular epithelial cell line (HK2). The present invention has been completed by identifying the FRYL and TIMP1 genes.
 すなわち、本発明は以下の[1]~[21]を提供する。
[1]GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質を含む、医薬組成物。
[2]GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質が核酸である、[1]に記載の医薬組成物。
[3]前記核酸が、siRNA若しくはアンチセンスオリゴヌクレオチド又はこれらの発現ベクターである[2]に記載の医薬組成物。
[4]GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質を含む、COL1A1遺伝子の発現抑制剤。
[5]GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質が核酸である、[4]に記載のCOL1A1遺伝子の発現抑制剤。
[6]前記核酸が、siRNA若しくはアンチセンスオリゴヌクレオチド又はこれらの発現ベクターである[5]に記載のCOL1A1遺伝子の発現抑制剤。
[7]GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質を含む、COL1A1遺伝子が関連する疾患の治療用医薬組成物。
[8]GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質が核酸である、[7]に記載の医薬組成物。
[9]前記核酸が、siRNA若しくはアンチセンスオリゴヌクレオチド又はこれらの発現ベクターである[8]に記載の医薬組成物。
[10]COL1A1遺伝子が関連する疾患が慢性腎臓病又は線維化疾患である[7]~[9]のいずれかに記載の医薬組成物。
[11](1)細胞と被験物質とを接触させる工程、
(2)被験物質を接触させた細胞におけるGALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現量を測定し、該発現量を、被験物質を接触させない対照細胞における前記遺伝子の発現量と比較する工程、及び
(3)被験物質を接触させた細胞における前記遺伝子の発現が、対照細胞における前記遺伝子の発現よりも低下している場合に、被験物質をCOL1A1遺伝子の発現抑制剤として選択する工程を含む、
COL1A1遺伝子の発現抑制剤のスクリーニング方法。
[12](1)細胞と被験物質とを接触させる工程、
(2)被験物質を接触させた細胞におけるGALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現量を測定し、該発現量を、被験物質を接触させない対照細胞における前記遺伝子の発現量と比較する工程、及び
(3)被験物質を接触させた細胞における前記遺伝子の発現が、対照細胞における前記遺伝子の発現よりも低下している場合に、被験物質をCOL1A1遺伝子が関連する疾患の治療物質として選択する工程を含む、
COL1A1遺伝子が関連する疾患の治療物質のスクリーニング方法。
[13]配列番号1で表されるアミノ酸配列を有するポリペプチド、又は配列番号1で表されるアミノ酸配列の1又は数個のアミノ酸が欠失、置換、及び/若しくは付加されたアミノ酸配列を有し、N-アセチルガラクトサミン転移活性を有するポリペプチドを用いることを特徴とするCOL1A1遺伝子が関連する疾患の治療物質のスクリーニング方法。
[14](1) GALNT2タンパク質に対する基質、被験物質及び
1:配列番号1で表されるアミノ酸配列を有するポリペプチド、又は
2:配列番号1で表されるアミノ酸配列の1又は数個のアミノ酸が欠失、置換、及び/若しくは付加されたアミノ酸配列を有し、N-アセチルガラクトサミン転移活性を有するポリペプチドを接触させる工程、
(2) 被験物質を接触させたポリペプチドのN-アセチルガラクトサミン転移活性を、被験物質を接触させないポリペプチドのN-アセチルガラクトサミン転移活性と比較する工程、及び
(3) 被験物質を接触させたポリペプチドのN-アセチルガラクトサミン転移活性が、被験物質を接触させないポリペプチドのN-アセチルガラクトサミン転移活性と比較して低下している場合に、被験物質をCOL1A1遺伝子が関連する疾患の治療物質として選択する工程を含む、
COL1A1遺伝子が関連する疾患の治療物質のスクリーニング方法。
[15]COL1A1遺伝子が関連する疾患が慢性腎臓病又は線維化疾患である[12]~[14]のいずれかに記載の方法。
[16]GALNT2、COL8A2、FRYL又は TIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質を投与する工程を有する、COL1A1遺伝子が関連する疾患を治療する方法。
[17]COL1A1遺伝子が関連する疾患の治療に用いられる、GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質。
[18]COL1A1遺伝子が関連する疾患の治療剤の製造のための、GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質の使用。
[19]GALNT2、COL8A2、FRYL又は TIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質を投与する工程を有する、慢性腎臓病又は線維化疾患を治療する方法。
[20]慢性腎臓病又は線維化疾患の治療に用いられる、GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質。
[21]慢性腎臓病又は線維化疾患の治療剤の製造のための、GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質の使用。
That is, the present invention provides the following [1] to [21].
[1] A pharmaceutical composition comprising a substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene, or a substance that suppresses the activity of a protein encoded by these genes.
[2] The pharmaceutical composition according to [1], wherein the substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or the substance that suppresses the activity of the protein encoded by these genes is a nucleic acid.
[3] The pharmaceutical composition according to [2], wherein the nucleic acid is siRNA or antisense oligonucleotide or an expression vector thereof.
[4] A COL1A1 gene expression inhibitor comprising a substance that suppresses expression of GALNT2, COL8A2, FRYL, or TIMP1 gene, or a substance that suppresses the activity of a protein encoded by these genes.
[5] The COL1A1 gene expression inhibitor according to [4], wherein the substance that suppresses the expression of the GALNT2, COL8A2, FRYL, or TIMP1 gene or the substance that suppresses the activity of the protein encoded by these genes is a nucleic acid.
[6] The COL1A1 gene expression inhibitor according to [5], wherein the nucleic acid is siRNA, an antisense oligonucleotide, or an expression vector thereof.
[7] A pharmaceutical composition for treating a disease associated with the COL1A1 gene, comprising a substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene, or a substance that suppresses the activity of a protein encoded by these genes.
[8] The pharmaceutical composition according to [7], wherein the substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or the substance that suppresses the activity of the protein encoded by these genes is a nucleic acid.
[9] The pharmaceutical composition according to [8], wherein the nucleic acid is siRNA or antisense oligonucleotide or an expression vector thereof.
[10] The pharmaceutical composition according to any one of [7] to [9], wherein the disease associated with the COL1A1 gene is chronic kidney disease or fibrotic disease.
[11] (1) A step of bringing a cell into contact with a test substance,
(2) measuring the expression level of the GALNT2, COL8A2, FRYL, or TIMP1 gene in a cell contacted with the test substance, and comparing the expression level with the expression level of the gene in a control cell not contacted with the test substance; (3) including a step of selecting the test substance as a COL1A1 gene expression inhibitor when the expression of the gene in the cell contacted with the test substance is lower than the expression of the gene in a control cell;
A screening method for a COL1A1 gene expression inhibitor.
[12] (1) A step of contacting a cell with a test substance,
(2) measuring the expression level of the GALNT2, COL8A2, FRYL, or TIMP1 gene in a cell contacted with the test substance, and comparing the expression level with the expression level of the gene in a control cell not contacted with the test substance; (3) selecting the test substance as a therapeutic substance for a disease associated with the COL1A1 gene when the expression of the gene in the cell contacted with the test substance is lower than the expression of the gene in a control cell; Including,
A screening method for a therapeutic agent for a disease associated with the COL1A1 gene.
[13] A polypeptide having the amino acid sequence represented by SEQ ID NO: 1, or an amino acid sequence in which one or several amino acids of the amino acid sequence represented by SEQ ID NO: 1 are deleted, substituted, and / or added. And a method for screening a therapeutic substance for a disease associated with the COL1A1 gene, which comprises using a polypeptide having N-acetylgalactosamine transfer activity.
[14] (1) A substrate for GALNT2 protein, a test substance and 1: a polypeptide having the amino acid sequence represented by SEQ ID NO: 1, or 2: one or several amino acids of the amino acid sequence represented by SEQ ID NO: 1 Contacting a polypeptide having an amino acid sequence deleted, substituted, and / or added and having N-acetylgalactosamine transfer activity;
(2) comparing the N-acetylgalactosamine transfer activity of the polypeptide contacted with the test substance with the N-acetylgalactosamine transfer activity of the polypeptide not contacted with the test substance, and
(3) When the N-acetylgalactosamine transfer activity of the polypeptide contacted with the test substance is lower than the N-acetylgalactosamine transfer activity of the polypeptide not contacted with the test substance, the test substance is transferred to the COL1A1 gene. Selecting as a therapeutic agent for a disease associated with
A screening method for a therapeutic agent for a disease associated with the COL1A1 gene.
[15] The method according to any one of [12] to [14], wherein the disease associated with the COL1A1 gene is chronic kidney disease or fibrotic disease.
[16] A method for treating a disease associated with the COL1A1 gene, comprising a step of administering a substance that suppresses expression of the GALNT2, COL8A2, FRYL, or TIMP1 gene or a substance that suppresses the activity of a protein encoded by these genes.
[17] A substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene, or the activity of a protein encoded by these genes, used for treatment of a disease associated with the COL1A1 gene.
[18] Use of a substance that suppresses the expression of the GALNT2, COL8A2, FRYL, or TIMP1 gene or a substance that suppresses the activity of a protein encoded by these genes for the manufacture of a therapeutic agent for a disease associated with the COL1A1 gene.
[19] A method for treating chronic kidney disease or fibrotic disease, comprising a step of administering a substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or a substance that suppresses the activity of a protein encoded by these genes.
[20] A substance that suppresses the expression of a GALNT2, COL8A2, FRYL, or TIMP1 gene or a protein encoded by these genes, which is used for treatment of chronic kidney disease or fibrotic disease.
[21] Use of a substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or a substance that suppresses the activity of a protein encoded by these genes for the manufacture of a therapeutic agent for chronic kidney disease or fibrotic disease.
 本発明により、GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制することで、COL1A1遺伝子の発現を抑制することが可能であり、GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質は慢性腎臓病又は線維化疾患の治療用医薬組成物の有効成分として用いることが出来る。本発明による医薬組成物は、特定の遺伝子の発現を特異的に抑制するという新しい作用機序の治療剤を提供するものであり、従来の慢性腎臓病又は線維化疾患の治療剤に比べて毒性が少ないことを大きな特徴とする。さらに本発明により、GALNT2、COL8A2、FRYL又はTIMP1遺伝子をターゲットとして用いる、慢性腎臓病又は線維化疾患の治療用物質をスクリーニングする方法も提供される。   According to the present invention, it is possible to suppress the expression of COL1A1 gene by suppressing the expression of GALNT2, COL8A2, FRYL or TIMP1 gene, and the substance which suppresses the expression of GALNT2, COL8A2, FRYL or TIMP1 gene is a chronic kidney It can be used as an active ingredient of a pharmaceutical composition for treating diseases or fibrotic diseases. The pharmaceutical composition according to the present invention provides a therapeutic agent having a new mechanism of action that specifically suppresses the expression of a specific gene, and is more toxic than conventional therapeutic agents for chronic kidney disease or fibrotic disease. It is a big feature that there is little. Furthermore, the present invention also provides a method for screening a substance for treating chronic kidney disease or fibrosis using the GALNT2, COL8A2, FRYL or TIMP1 gene as a target. *
 本明細書において使用される用語は、特に言及する場合を除いて、当該分野で通常用いる意味で用いられる。 Unless otherwise stated, terms used in the present specification are used in the meaning normally used in this field.
 本発明は、1つの態様において、GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質を含む、COL1A1遺伝子が関連する疾患の治療用医薬組成物を提供する。COL1A1遺伝子が関連する疾患としては、例えば、慢性腎不全、慢性腎炎、尿細管間質障害、ネフローゼ症候群、多発性のう胞腎、糖尿病性腎症、及び腎硬化症などの慢性腎臓病に包含される疾患、並びに強皮症、肥厚性瘢痕、瘢痕拘縮、ケロイド症、膠原病、肺線維症、繊維性癒着、珪肺、骨髄線維症、腎性全身性線維症、クローン病、心筋線維症、及び肝硬変などの線維化疾患に包含される疾患を挙げることができる。 In one embodiment, the present invention provides a therapeutic drug for a disease associated with the COL1A1 gene, comprising a substance that suppresses expression of the GALNT2, COL8A2, FRYL, or TIMP1 gene, or a substance that suppresses the activity of the protein encoded by these genes. A composition is provided. Diseases associated with the COL1A1 gene include, for example, chronic kidney disease such as chronic renal failure, chronic nephritis, tubulointerstitial disorder, nephrotic syndrome, polycystic kidney disease, diabetic nephropathy, and nephrosclerosis Disease, and scleroderma, hypertrophic scar, scar contracture, keloid disease, collagen disease, pulmonary fibrosis, fibrosis, silicosis, myelofibrosis, renal systemic fibrosis, Crohn's disease, myocardial fibrosis, and The disease included by fibrosis diseases, such as cirrhosis, can be mentioned.
 本明細書の実施例1-3に示す通り、GALNT2、COL8A2、FRYL又はTIMP1を新規な慢性腎臓病又は線維化疾患の標的分子として見出した。 As shown in Example 1-3 of the present specification, GALNT2, COL8A2, FRYL or TIMP1 was found as a novel target molecule for chronic kidney disease or fibrotic disease.
 本明細書において、「GALNT2遺伝子」とは、GALNT2タンパク質をコードする遺伝子を意味する。ヒトGALNT2遺伝子の塩基配列及びヒトGALNT2タンパク質のアミノ酸配列は公知であり、例えば、ヒトGALNT2遺伝子の塩基配列およびヒトGALNT2タンパク質アミノ酸配列がGenBankに登録され(GenBank Accession No. NM_004481)、公表されている。
 本明細書において、ヒトGALNT2タンパク質は、前記のGenBankに登録されているアミノ酸配列からなるタンパク質のみならず、ヒト個体内で生じ得る変異体も含み、前記のGenBankに登録されているアミノ酸配列からなるタンパク質において1アミノ酸又は数アミノ酸が欠失、置換及び/又は付加されたタンパク質であって多型性や突然変異に基づく変異により生じるもの、が含まれる。ただし、これらの変異体ヒトGALNT2タンパク質は、前記のGenBankに登録されているアミノ酸配列からなるタンパク質と同等の機能を有するものである。
 本明細書において、ヒトGALNT2遺伝子は、前記のGenBankに登録されている塩基配列からなる遺伝子のみならず、ヒト個体内で生じ得る変異体も含み、例えば前記のGenBankに登録されている塩基配列からなる遺伝子において1塩基又は数塩基が欠失、置換及び/又は付加された遺伝子であって、多型性や突然変異に基づく変異により生じるもの、が含まれる。さらに、「GALNT2遺伝子」は、前記のGenBankに登録されている塩基配列に対して、80%以上、例えば、85%以上、90%以上、95%以上、97%以上、98%以上、99%以上、99.5%以上、99.7%以上または99.9%以上の同一性を有するヌクレオチド配列からなる変異体を含む。塩基配列の同一性は、BLAST、FASTAなどの公知のアルゴリズムを利用して決定できる。ただし、これらの変異体遺伝子は、前記のGenBankに登録されているアミノ酸配列からなるタンパク質と同等の機能を有するタンパク質をコードするものである。
 GALNT2タンパク質は、GALNT1からGALNT14、GALNTL1、GALNTL2、GALNTL4、GALNTL5、GALNTL6、WBSCR17タンパク質の少なくとも20種類からなるGalNAc transferase(GALNT)のファミリーに属している。GALNTファミリーのタンパク質は、Nアセチルガラクトサミンを基質タンパク質のセリンまたはスレオニン残基に付加する酵素であって、基質タンパク質のグリコシル化の最初のステップに関与していることが知られている。
 糖尿病性腎症の患者群において上昇する遺伝子が1,200ほど公開されているが(DIABETES、60巻、2354-2369頁、2011年のSupplementary data)、GALNT2遺伝子はその1,200あまりの遺伝子のうちの1つとして挙げられている。但し、GALNT2遺伝子の発現抑制及びGALNT2タンパク質の活性抑制と慢性腎臓病や線維化疾患との関連性については知られていない。
In the present specification, the “GALNT2 gene” means a gene encoding GALNT2 protein. The base sequence of the human GALNT2 gene and the amino acid sequence of the human GALNT2 protein are known. For example, the base sequence of the human GALNT2 gene and the human GALNT2 protein amino acid sequence are registered in GenBank (GenBank Accession No. NM_004481) and published.
In this specification, the human GALNT2 protein includes not only a protein consisting of the amino acid sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual, and consists of the amino acid sequence registered in the above-mentioned GenBank. Proteins in which one amino acid or several amino acids are deleted, substituted and / or added in the protein, which are caused by mutations based on polymorphisms or mutations are included. However, these mutant human GALNT2 proteins have functions equivalent to those of the protein consisting of the amino acid sequence registered in the above GenBank.
In this specification, the human GALNT2 gene includes not only a gene consisting of a base sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual, for example, from a base sequence registered in the above-mentioned GenBank. And a gene in which one or several bases are deleted, substituted, and / or added, and is generated by a mutation based on polymorphism or mutation. Furthermore, the “GALNT2 gene” is 80% or more, for example, 85% or more, 90% or more, 95% or more, 97% or more, 98% or more, 99% with respect to the base sequence registered in the above GenBank. As mentioned above, the variant which consists of a nucleotide sequence which has 99.5% or more, 99.7% or more, or 99.9% or more identity is included. The identity of the base sequence can be determined using a known algorithm such as BLAST or FASTA. However, these mutant genes encode a protein having a function equivalent to that of the protein consisting of the amino acid sequence registered in the above GenBank.
The GALNT2 protein belongs to the family of GalNAc transferase (GALNT) consisting of at least 20 kinds of GALNT1 to GALNT14, GALNTL1, GALNTL2, GALNTL4, GALNTL5, GALNTL6, and WBSCR17 proteins. GALNT family proteins are enzymes that add N-acetylgalactosamine to serine or threonine residues of a substrate protein and are known to be involved in the first step of substrate protein glycosylation.
There are about 1,200 genes that are elevated in the diabetic nephropathy patient group (DIABETES, 60, 2354-2369, 2011 Supplementary data), but the GALNT2 gene is one of over 1,200 genes. It is mentioned as. However, the relationship between suppression of GALNT2 gene expression and suppression of GALNT2 protein activity and chronic kidney disease or fibrosis is not known.
 本明細書において、「COL1A1遺伝子」とは、COL1A1タンパク質をコードする遺伝子を意味する。ヒトCOL1A1遺伝子の塩基配列及びヒトCOL1A1タンパク質のアミノ酸配列は公知であり、例えば、ヒトCOL1A1遺伝子の塩基配列およびヒトCOL1A1タンパク質アミノ酸配列がGenBankに登録され(GenBank Accession No. NM_000088)、公表されている。
 本明細書において、ヒトCOL1A1タンパク質は、前記のGenBankに登録されているアミノ酸配列からなるタンパク質のみならず、ヒト個体内で生じ得る変異体も含み、前記のGenBankに登録されているアミノ酸配列からなるタンパク質において1アミノ酸又は数アミノ酸が欠失、置換及び/又は付加されたタンパク質であって多型性や突然変異に基づく変異により生じるもの、が含まれる。ただし、これらの変異体ヒトCOL1A1タンパク質は、前記のGenBankに登録されているアミノ酸配列からなるタンパク質と同等の機能を有するものである。
 本明細書において、ヒトCOL1A1遺伝子は、前記のGenBankに登録されている塩基配列からなる遺伝子のみならず、ヒト個体内で生じ得る変異体も含み、例えば前記のGenBankに登録されている塩基配列からなる遺伝子において1塩基又は数塩基が欠失、置換及び/又は付加された遺伝子であって、多型性や突然変異に基づく変異により生じるもの、が含まれる。さらに、「COL1A1遺伝子」は、前記のGenBankに登録されている塩基配列に対して、80%以上、例えば、85%以上、90%以上、95%以上、97%以上、98%以上、99%以上、99.5%以上、99.7%以上または99.9%以上の同一性を有するヌクレオチド配列からなる変異体を含む。塩基配列の同一性は、BLAST、FASTAなどの公知のアルゴリズムを利用して決定できる。ただし、これらの変異体遺伝子は、前記のGenBankに登録されているアミノ酸配列からなるタンパク質と同等の機能を有するタンパク質をコードするものである。
 COL1A1遺伝子等の線維化関連遺伝子の発現が上昇し、細胞外マトリックスの過剰産生が起こることによって、慢性腎臓病や線維化疾患が引き起こされることが知られている(Nephron Experimental Nephrology, 102巻、e71-e75、2006年 参照)。
したがって、COL1A1遺伝子等の線維化関連遺伝子の発現抑制剤は、慢性腎臓病や線維化疾患の治療剤となる。
In the present specification, “COL1A1 gene” means a gene encoding COL1A1 protein. The base sequence of the human COL1A1 gene and the amino acid sequence of the human COL1A1 protein are known. For example, the base sequence of the human COL1A1 gene and the human COL1A1 protein amino acid sequence are registered in GenBank (GenBank Accession No. NM_000088) and published.
In this specification, the human COL1A1 protein includes not only a protein consisting of the amino acid sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual, and consists of the amino acid sequence registered in the above-mentioned GenBank. Proteins in which one amino acid or several amino acids are deleted, substituted and / or added in the protein, which are caused by mutations based on polymorphisms or mutations are included. However, these mutant human COL1A1 proteins have functions equivalent to those of the protein consisting of the amino acid sequence registered in the above GenBank.
In this specification, the human COL1A1 gene includes not only a gene having a base sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual. For example, from the base sequence registered in the above-mentioned GenBank And a gene in which one or several bases are deleted, substituted, and / or added, and is generated by a mutation based on polymorphism or mutation. Furthermore, the “COL1A1 gene” is 80% or more, for example, 85% or more, 90% or more, 95% or more, 97% or more, 98% or more, 99% with respect to the base sequence registered in the above GenBank. As mentioned above, the variant which consists of a nucleotide sequence which has 99.5% or more, 99.7% or more, or 99.9% or more identity is included. The identity of the base sequence can be determined using a known algorithm such as BLAST or FASTA. However, these mutant genes encode a protein having a function equivalent to that of the protein consisting of the amino acid sequence registered in the above GenBank.
It is known that expression of fibrosis-related genes such as the COL1A1 gene is increased and extracellular matrix overproduction occurs, leading to chronic kidney disease and fibrosis (Nephron Experimental Nephrology, Vol. 102, e71). -e75, see 2006).
Therefore, an expression inhibitor of a fibrosis-related gene such as the COL1A1 gene is a therapeutic agent for chronic kidney disease or fibrosis.
 本明細書において、「αSMA遺伝子」とは、αSMAタンパク質をコードする遺伝子を意味する。ヒトαSMA遺伝子の塩基配列及びヒトαSMAタンパク質のアミノ酸配列は公知であり、例えば、ヒトαSMA遺伝子の塩基配列およびヒトαSMAタンパク質アミノ酸配列がGenBankに登録され(GenBank Accession No. NM_001141945)、公表されている。
 本明細書において、ヒトαSMAタンパク質は、前記のGenBankに登録されているアミノ酸配列からなるタンパク質のみならず、ヒト個体内で生じ得る変異体も含み、前記のGenBankに登録されているアミノ酸配列からなるタンパク質において1アミノ酸又は数アミノ酸が欠失、置換及び/又は付加されたタンパク質であって多型性や突然変異に基づく変異により生じるもの、が含まれる。ただし、これらの変異体ヒトαSMAタンパク質は、前記のGenBankに登録されているアミノ酸配列からなるタンパク質と同等の機能を有するものである。
 本明細書において、ヒトαSMA遺伝子は、前記のGenBankに登録されている塩基配列からなる遺伝子のみならず、ヒト個体内で生じ得る変異体も含み、例えば前記のGenBankに登録されている塩基配列からなる遺伝子において1塩基又は数塩基が欠失、置換及び/又は付加された遺伝子であって、多型性や突然変異に基づく変異により生じるもの、が含まれる。さらに、「αSMA遺伝子」は、前記のGenBankに登録されている塩基配列に対して、80%以上、例えば、85%以上、90%以上、95%以上、97%以上、98%以上、99%以上、99.5%以上、99.7%以上または99.9%以上の同一性を有するヌクレオチド配列からなる変異体を含む。塩基配列の同一性は、BLAST、FASTAなどの公知のアルゴリズムを利用して決定できる。ただし、これらの変異体遺伝子は、前記のGenBankに登録されているアミノ酸配列からなるタンパク質と同等の機能を有するタンパク質をコードするものである。
 αSMAは線維化進行のマーカーとして知られており、慢性腎臓病や線維化疾患において発現上昇することが知られている(Histopathology, 47巻、276-280頁、2005年、J Am Soc Nephrol 15巻、1-12頁、2004年、及びVirchows Archiv, 450巻、41-50頁、2007年 参照)。
In the present specification, the “αSMA gene” means a gene encoding an αSMA protein. The nucleotide sequence of the human αSMA gene and the amino acid sequence of the human αSMA protein are known. For example, the nucleotide sequence of the human αSMA gene and the amino acid sequence of the human αSMA protein are registered in GenBank (GenBank Accession No. NM_001141945) and published.
In this specification, the human αSMA protein includes not only a protein consisting of the amino acid sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual, and consists of the amino acid sequence registered in the above-mentioned GenBank. Proteins in which one amino acid or several amino acids are deleted, substituted and / or added in the protein, which are caused by mutations based on polymorphisms or mutations are included. However, these mutant human αSMA proteins have functions equivalent to those of the protein consisting of the amino acid sequence registered in the above-described GenBank.
In the present specification, the human αSMA gene includes not only a gene consisting of a base sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual, for example, from a base sequence registered in the above-mentioned GenBank. And a gene in which one or several bases are deleted, substituted, and / or added, and is generated by a mutation based on polymorphism or mutation. Furthermore, the “αSMA gene” is 80% or more, for example, 85% or more, 90% or more, 95% or more, 97% or more, 98% or more, 99% with respect to the base sequence registered in the above GenBank. As mentioned above, the variant which consists of a nucleotide sequence which has 99.5% or more, 99.7% or more, or 99.9% or more identity is included. The identity of the base sequence can be determined using a known algorithm such as BLAST or FASTA. However, these mutant genes encode a protein having a function equivalent to that of the protein consisting of the amino acid sequence registered in the above GenBank.
αSMA is known as a marker of fibrosis progression and is known to be upregulated in chronic kidney disease and fibrosis (Histopathology, 47, 276-280, 2005, J Am Soc Nephrol 15) 1-12, 2004, and Virchows Archiv, 450, 41-50, 2007).
 本明細書において、「COL8A2遺伝子」とは、COL8A2タンパク質をコードする遺伝子を意味する。ヒトCOL8A2遺伝子の塩基配列及びヒトCOL8A2タンパク質のアミノ酸配列は公知であり、例えば、ヒトCOL8A2遺伝子の塩基配列およびヒトCOL8A2タンパク質アミノ酸配列がGenBankに登録され(GenBank Accession No. NM_005202)、公表されている。
 本明細書において、ヒトCOL8A2タンパク質は、前記のGenBankに登録されているアミノ酸配列からなるタンパク質のみならず、ヒト個体内で生じ得る変異体も含み、前記のGenBankに登録されているアミノ酸配列からなるタンパク質において1アミノ酸又は数アミノ酸が欠失、置換及び/又は付加されたタンパク質であって多型性や突然変異に基づく変異により生じるもの、が含まれる。ただし、これらの変異体ヒトCOL8A2タンパク質は、前記のGenBankに登録されているアミノ酸配列からなるタンパク質と同等の機能を有するものである。
 本明細書において、ヒトCOL8A2遺伝子は、前記のGenBankに登録されている塩基配列からなる遺伝子のみならず、ヒト個体内で生じ得る変異体も含み、例えば前記のGenBankに登録されている塩基配列からなる遺伝子において1塩基又は数塩基が欠失、置換及び/又は付加された遺伝子であって、多型性や突然変異に基づく変異により生じるもの、が含まれる。さらに、「COL8A2遺伝子」は、前記のGenBankに登録されている塩基配列に対して、80%以上、例えば、85%以上、90%以上、95%以上、97%以上、98%以上、99%以上、99.5%以上、99.7%以上または99.9%以上の同一性を有するヌクレオチド配列からなる変異体を含む。塩基配列の同一性は、BLAST、FASTAなどの公知のアルゴリズムを利用して決定できる。ただし、これらの変異体遺伝子は、前記のGenBankに登録されているアミノ酸配列からなるタンパク質と同等の機能を有するタンパク質をコードするものである。
In the present specification, “COL8A2 gene” means a gene encoding COL8A2 protein. The base sequence of the human COL8A2 gene and the amino acid sequence of the human COL8A2 protein are known. For example, the base sequence of the human COL8A2 gene and the human COL8A2 protein amino acid sequence are registered in GenBank (GenBank Accession No. NM_005202) and published.
In this specification, the human COL8A2 protein includes not only a protein consisting of the amino acid sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual, and consists of the amino acid sequence registered in the above-mentioned GenBank. Proteins in which one amino acid or several amino acids are deleted, substituted and / or added in the protein, which are caused by mutations based on polymorphisms or mutations are included. However, these mutant human COL8A2 proteins have functions equivalent to those of the protein consisting of the amino acid sequence registered in the above GenBank.
In this specification, the human COL8A2 gene includes not only a gene having a base sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual, for example, from a base sequence registered in the above-mentioned GenBank. And a gene in which one or several bases are deleted, substituted, and / or added, and is generated by a mutation based on polymorphism or mutation. Furthermore, the “COL8A2 gene” is 80% or more, for example, 85% or more, 90% or more, 95% or more, 97% or more, 98% or more, 99% with respect to the base sequence registered in the above GenBank. As mentioned above, the variant which consists of a nucleotide sequence which has 99.5% or more, 99.7% or more, or 99.9% or more identity is included. The identity of the base sequence can be determined using a known algorithm such as BLAST or FASTA. However, these mutant genes encode a protein having a function equivalent to that of the protein consisting of the amino acid sequence registered in the above GenBank.
 本明細書において、「FRYL遺伝子」とは、FRYLタンパク質をコードする遺伝子を意味する。ヒトFRYL遺伝子の塩基配列及びヒトFRYLタンパク質のアミノ酸配列は公知であり、例えば、ヒトFRYL遺伝子の塩基配列およびヒトFRYLタンパク質アミノ酸配列がGenBankに登録され(GenBank Accession No. NM_015030)、公表されている。
 本明細書において、ヒトFRYLタンパク質は、前記のGenBankに登録されているアミノ酸配列からなるタンパク質のみならず、ヒト個体内で生じ得る変異体も含み、前記のGenBankに登録されているアミノ酸配列からなるタンパク質において1アミノ酸又は数アミノ酸が欠失、置換及び/又は付加されたタンパク質であって多型性や突然変異に基づく変異により生じるもの、が含まれる。ただし、これらの変異体ヒトFRYLタンパク質は、前記のGenBankに登録されているアミノ酸配列からなるタンパク質と同等の機能を有するものである。
 本明細書において、ヒトFRYL遺伝子は、前記のGenBankに登録されている塩基配列からなる遺伝子のみならず、ヒト個体内で生じ得る変異体も含み、例えば前記のGenBankに登録されている塩基配列からなる遺伝子において1塩基又は数塩基が欠失、置換及び/又は付加された遺伝子であって、多型性や突然変異に基づく変異により生じるもの、が含まれる。さらに、「FRYL遺伝子」は、前記のGenBankに登録されている塩基配列に対して、80%以上、例えば、85%以上、90%以上、95%以上、97%以上、98%以上、99%以上、99.5%以上、99.7%以上または99.9%以上の同一性を有するヌクレオチド配列からなる変異体を含む。塩基配列の同一性は、BLAST、FASTAなどの公知のアルゴリズムを利用して決定できる。ただし、これらの変異体遺伝子は、前記のGenBankに登録されているアミノ酸配列からなるタンパク質と同等の機能を有するタンパク質をコードするものである。
In the present specification, “FRYL gene” means a gene encoding FRYL protein. The nucleotide sequence of the human FRYL gene and the amino acid sequence of the human FRYL protein are known. For example, the nucleotide sequence of the human FRYL gene and the amino acid sequence of the human FRYL protein are registered in GenBank (GenBank Accession No. NM_015030) and published.
In this specification, the human FRYL protein includes not only a protein consisting of the amino acid sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual, and consists of the amino acid sequence registered in the above-mentioned GenBank. Proteins in which one amino acid or several amino acids are deleted, substituted and / or added in the protein, which are caused by mutations based on polymorphisms or mutations are included. However, these mutant human FRYL proteins have functions equivalent to those of the protein consisting of the amino acid sequence registered in the above-described GenBank.
In the present specification, the human FRYL gene includes not only a gene consisting of a base sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual, for example, from a base sequence registered in the above-mentioned GenBank. And a gene in which one or several bases are deleted, substituted, and / or added, and is generated by a mutation based on polymorphism or mutation. Furthermore, the “FRYL gene” is 80% or more, for example, 85% or more, 90% or more, 95% or more, 97% or more, 98% or more, 99% with respect to the base sequence registered in the above GenBank. As mentioned above, the variant which consists of a nucleotide sequence which has 99.5% or more, 99.7% or more, or 99.9% or more identity is included. The identity of the base sequence can be determined using a known algorithm such as BLAST or FASTA. However, these mutant genes encode a protein having a function equivalent to that of the protein consisting of the amino acid sequence registered in the above GenBank.
 本明細書において、「TIMP1遺伝子」とは、TIMP1タンパク質をコードする遺伝子を意味する。ヒトTIMP1遺伝子の塩基配列及びヒトTIMP1タンパク質のアミノ酸配列は公知であり、例えば、ヒトTIMP1遺伝子の塩基配列およびヒトTIMP1タンパク質アミノ酸配列がGenBankに登録され(GenBank Accession No. NM_003254)、公表されている。
 本明細書において、ヒトTIMP1タンパク質は、前記のGenBankに登録されているアミノ酸配列からなるタンパク質のみならず、ヒト個体内で生じ得る変異体も含み、前記のGenBankに登録されているアミノ酸配列からなるタンパク質において1アミノ酸又は数アミノ酸が欠失、置換及び/又は付加されたタンパク質であって多型性や突然変異に基づく変異により生じるもの、が含まれる。ただし、これらの変異体ヒトTIMP1タンパク質は、前記のGenBankに登録されているアミノ酸配列からなるタンパク質と同等の機能を有するものである。
 本明細書において、ヒトTIMP1遺伝子は、前記のGenBankに登録されている塩基配列からなる遺伝子のみならず、ヒト個体内で生じ得る変異体も含み、例えば前記のGenBankに登録されている塩基配列からなる遺伝子において1塩基又は数塩基が欠失、置換及び/又は付加された遺伝子であって、多型性や突然変異に基づく変異により生じるもの、が含まれる。さらに、「TIMP1遺伝子」は、前記のGenBankに登録されている塩基配列に対して、80%以上、例えば、85%以上、90%以上、95%以上、97%以上、98%以上、99%以上、99.5%以上、99.7%以上または99.9%以上の同一性を有するヌクレオチド配列からなる変異体を含む。塩基配列の同一性は、BLAST、FASTAなどの公知のアルゴリズムを利用して決定できる。ただし、これらの変異体遺伝子は、前記のGenBankに登録されているアミノ酸配列からなるタンパク質と同等の機能を有するタンパク質をコードするものである。
In the present specification, the “TIMP1 gene” means a gene encoding a TIMP1 protein. The base sequence of the human TIMP1 gene and the amino acid sequence of the human TIMP1 protein are known. For example, the base sequence of the human TIMP1 gene and the human TIMP1 protein amino acid sequence are registered in GenBank (GenBank Accession No. NM_003254) and published.
In this specification, the human TIMP1 protein includes not only a protein consisting of the amino acid sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual, and consists of the amino acid sequence registered in the above-mentioned GenBank. Proteins in which one amino acid or several amino acids are deleted, substituted and / or added in the protein, which are caused by mutations based on polymorphisms or mutations are included. However, these mutant human TIMP1 proteins have functions equivalent to those of the protein consisting of the amino acid sequence registered in the above GenBank.
In this specification, the human TIMP1 gene includes not only a gene consisting of a base sequence registered in the above-mentioned GenBank but also a variant that can occur in a human individual. For example, from the base sequence registered in the above-mentioned GenBank And a gene in which one or several bases are deleted, substituted, and / or added, and is generated by a mutation based on polymorphism or mutation. Furthermore, the “TIMP1 gene” is 80% or more, for example, 85% or more, 90% or more, 95% or more, 97% or more, 98% or more, 99% with respect to the base sequence registered in the above GenBank. As mentioned above, the variant which consists of a nucleotide sequence which has 99.5% or more, 99.7% or more, or 99.9% or more identity is included. The identity of the base sequence can be determined using a known algorithm such as BLAST or FASTA. However, these mutant genes encode a protein having a function equivalent to that of the protein consisting of the amino acid sequence registered in the above GenBank.
 本明細書において、「遺伝子の発現を抑制する物質」とは、標的遺伝子のmRNAの転写を抑制する物質、転写されたmRNAを分解する物質、またはmRNAからのタンパク質の翻訳を抑制する物質であれば特に限定されるものでない。かかる物質として、siRNA、アンチセンスオリゴヌクレオチド、マイクロRNA若しくはリボザイム又はこれらの発現ベクターなどが例示される。其の中でも、siRNA若しくはアンチセンスオリゴヌクレオチド又はその発現ベクターが好ましく、特にsiRNA又はアンチセンスオリゴヌクレオチドが好ましい。「遺伝子の発現を抑制する物質」としては、上記のほか、タンパク質やペプチド、あるいは他の小分子も含まれる。なお、本発明において標的遺伝子は、COL8A2、GALNT2、FRYL又はTIMP1遺伝子である。 In the present specification, the “substance that suppresses gene expression” refers to a substance that suppresses the transcription of mRNA of the target gene, a substance that degrades the transcribed mRNA, or a substance that suppresses the translation of the protein from the mRNA. There is no particular limitation. Examples of such substances include siRNA, antisense oligonucleotide, microRNA, ribozyme, and expression vectors thereof. Among them, siRNA or antisense oligonucleotide or an expression vector thereof is preferable, and siRNA or antisense oligonucleotide is particularly preferable. In addition to the above, “substances that suppress gene expression” include proteins, peptides, and other small molecules. In the present invention, the target gene is COL8A2, GALNT2, FRYL, or TIMP1 gene.
 本明細書において、「遺伝子がコードするタンパク質の活性を抑制する物質」とは、標的タンパク質が本来有する機能を抑制する物質であれば特に限定されるものではない。かかる物質として、抗体やアンタゴニストなどが例示される。
 例えば、GALNT2タンパク質の活性を抑制する物質とは、GALNT2タンパク質がNアセチルガラクトサミンを基質のセリンまたはスレオニン残基に付与する活性を阻害する物質を意味する。GALNT2タンパク質の基質としては、ApoC-IIIやANGPTL3等が知られている(Proceedings of the National Academy of Sciences of the USA, 109巻、9893-9898頁、2012年参照)。
In the present specification, the “substance that suppresses the activity of the protein encoded by the gene” is not particularly limited as long as it is a substance that suppresses the function inherent to the target protein. Examples of such substances include antibodies and antagonists.
For example, the substance that suppresses the activity of the GALNT2 protein means a substance that inhibits the activity of the GALNT2 protein imparting N-acetylgalactosamine to the serine or threonine residue of the substrate. As a substrate of GALNT2 protein, ApoC-III, ANGPTL3 and the like are known (see Proceedings of the National Academy of Sciences of the USA, Vol. 109, pages 9893-9898, 2012).
 本明細書において、「siRNA」とは、約15~約40塩基からなる二本鎖RNA部分を有するRNA分子であり、前記siRNAのアンチセンス鎖と相補的な配列をもつ標的遺伝子のmRNAを切断し、標的遺伝子の発現を抑制する機能を有する。詳細には、本発明におけるsiRNAは、COL8A2、GALNT2、FRYL若しくはTIMP1遺伝子のmRNA中の連続したRNA配列と相同な配列からなるセンスRNA鎖と、該センスRNA配列に相補的な配列からなるアンチセンスRNA鎖とからなる二本鎖RNA部分を含んでなるRNAである。かかるsiRNAおよび後述の変異体siRNAの設計および製造は当業者の技量の範囲内である。 In the present specification, “siRNA” is an RNA molecule having a double-stranded RNA portion consisting of about 15 to about 40 bases, and cleaves the mRNA of the target gene having a sequence complementary to the antisense strand of the siRNA. And has a function of suppressing the expression of the target gene. Specifically, the siRNA in the present invention is an antisense consisting of a sense RNA strand consisting of a sequence homologous to a continuous RNA sequence in mRNA of COL8A2, GALNT2, FRYL or TIMP1 gene, and a sequence complementary to the sense RNA sequence. An RNA comprising a double-stranded RNA portion comprising an RNA strand. The design and production of such siRNAs and mutant siRNAs described below are within the skill of the artisan.
 二本鎖RNA部分の長さは、塩基として、約15~約40塩基、好ましくは15~30塩基、より好ましくは15~25塩基、更に好ましくは18~23塩基、最も好ましくは19~21塩基である。siRNAのセンス鎖またはアンチセンス鎖の末端構造としては、特に制限はなく、目的に応じて適宜選択することができ、例えば、平滑末端を有するものであってもよいし、突出末端(オーバーハング)を有するものであってもよく、3’末端が突き出したタイプが好ましい。センスRNA鎖およびアンチセンスRNA鎖の3’末端に数個の塩基、好ましくは1~3個の塩基、さらに好ましくは2個の塩基からなるオーバーハングを有するsiRNAは、標的遺伝子の発現を抑制する効果が大きい場合が多く、好ましいものである。オーバーハングの塩基の種類は特に制限はなく、RNAを構成する塩基あるいはDNAを構成する塩基のいずれであってもよい。 The length of the double-stranded RNA portion is about 15 to about 40 bases, preferably 15 to 30 bases, more preferably 15 to 25 bases, still more preferably 18 to 23 bases, and most preferably 19 to 21 bases as a base. It is. The end structure of the sense strand or antisense strand of siRNA is not particularly limited and may be appropriately selected depending on the intended purpose. For example, it may have a blunt end or a protruding end (overhang) It is preferable that the 3 ′ end protrudes. The siRNA having an overhang consisting of several bases, preferably 1 to 3 bases, more preferably 2 bases, at the 3 ′ end of the sense RNA strand and the antisense RNA strand suppresses the expression of the target gene. In many cases, the effect is large, which is preferable. The type of the overhanging base is not particularly limited, and may be either a base constituting RNA or a base constituting DNA.
 さらに、上記siRNAのセンス鎖またはアンチセンス鎖の一方または両方において1~数個までのヌクレオチドが欠失、置換、挿入及び/又は付加されているsiRNAもまた、本発明の慢性腎臓病又は線維化疾患の治療用医薬組成物に用いることができる。ここで、1~数塩基とは、特に限定されるものではないが、好ましくは1~4塩基、さらに好ましくは1~3塩基、最も好ましくは1~2塩基である。かかる変異の具体例としては、3’末端のオーバーハング部分の塩基数を0~3個としたもの、3’末端のオーバーハング部分の塩基配列を他の塩基配列に変更したもの、あるいは塩基の挿入、付加または欠失により上記センスRNA鎖とアンチセンスRNA鎖の長さが1~3塩基異なるもの、センス鎖および/またはアンチセンス鎖において塩基が別の塩基にて置換されているもの等が挙げられるが、これらに限定されない。ただし、これらの変異体siRNAにおいてセンス鎖とアンチセンス鎖がハイブリダイゼーションしうること、ならびにこれらの変異体siRNAが変異を有しないsiRNAとほぼ同程度の遺伝子発現抑制能を有することが必要である。 Furthermore, siRNA in which 1 to several nucleotides are deleted, substituted, inserted and / or added in one or both of the sense strand or the antisense strand of the siRNA is also used in the chronic kidney disease or fibrosis of the present invention. It can be used in a pharmaceutical composition for treating a disease. Here, the 1 to several bases are not particularly limited, but are preferably 1 to 4 bases, more preferably 1 to 3 bases, and most preferably 1 to 2 bases. Specific examples of such mutations include those in which the number of bases in the overhang portion at the 3 ′ end is 0 to 3, or the base sequence in the overhang portion at the 3 ′ end is changed to another base sequence, or Those in which the length of the sense RNA strand differs from that of the antisense RNA strand by 1 to 3 bases due to insertion, addition or deletion, or in which the base is replaced with another base in the sense strand and / or antisense strand For example, but not limited to. However, it is necessary that the sense strand and the antisense strand can hybridize in these mutant siRNAs, and that these mutant siRNAs have approximately the same level of gene expression suppression ability as siRNA having no mutation.
 さらに、該siRNAは、一方の端が閉じた構造の分子、例えば、ヘアピン構造を有するsiRNA(Short hairpin RNA;shRNA)であってもよい。shRNAは、標的遺伝子の特定配列のセンス鎖RNA、該センス鎖RNAに相補的な配列からなるアンチセンス鎖RNA及びその両鎖を繋ぐリンカー配列を含むRNAであり、センス鎖部分とアンチセンス鎖部分がハイブリダイズし、二本鎖RNA部分を形成する。 Furthermore, the siRNA may be a molecule having a closed structure at one end, for example, an siRNA having a hairpin structure (Short hairpin RNA; shRNA). A shRNA is a RNA comprising a sense strand RNA of a specific sequence of a target gene, an antisense strand RNA consisting of a sequence complementary to the sense strand RNA, and a linker sequence connecting both strands. The sense strand portion and the antisense strand portion Hybridize to form a double stranded RNA portion.
 siRNAは、臨床使用の際には、いわゆるoff-target効果を示さないことが望ましい。off-target効果とは、標的遺伝子以外に、使用したsiRNAに部分的にホモロジーのある別の遺伝子の発現を抑制する作用をいう。off-target効果を避けるために、候補siRNAについて、予めDNAマイクロアレイなどを利用して交差反応がないことを確認することが可能である。また、NCBI(National Center for Biotechnology Information)などが提供する公知のデータベースを用いて、標的となる遺伝子以外に候補siRNAの配列と相同性が高い部分を含む遺伝子が存在しないかを確認する事によって、off-target効果を避けることが可能である。 It is desirable that siRNA does not show a so-called off-target effect in clinical use. The off-target effect refers to the action of suppressing the expression of another gene that is partially homologous to the siRNA used in addition to the target gene. In order to avoid the off-target effect, it is possible to confirm in advance that there is no cross-reaction for the candidate siRNA using a DNA microarray or the like. In addition, by using a known database provided by NCBI (National Center for Biotechnology Information), etc., by confirming that there is no gene containing a portion having high homology with the candidate siRNA sequence other than the target gene, It is possible to avoid the off-target effect.
 本発明のsiRNAを作製するには、化学合成による方法及び遺伝子組換え技術を用いる方法等、公知の方法を適宜用いることができる。合成による方法では、配列情報に基づき、常法により二本鎖RNAを合成することができる。また、遺伝子組換え技術を用いる方法では、センス鎖配列やアンチセンス鎖配列を組み込んだ発現ベクターを構築し、該ベクターを宿主細胞に導入後、転写により生成されたセンス鎖RNAやアンチセンス鎖RNAをそれぞれ取得することによって作製することもできる。また、標的遺伝子の特定配列のセンス鎖RNA、該センス鎖RNAに相補的な配列からなるアンチセンス鎖RNA及びその両鎖を繋ぐリンカー配列を含み、ヘアピン構造を形成するshRNAを発現させることにより、所望の二本鎖RNAを作製することもできる。 In order to produce the siRNA of the present invention, a known method such as a method using chemical synthesis or a method using a gene recombination technique can be appropriately used. In the synthesis method, double-stranded RNA can be synthesized by a conventional method based on sequence information. In the method using gene recombination technology, an expression vector incorporating a sense strand sequence or an antisense strand sequence is constructed, and the sense strand RNA or antisense strand RNA generated by transcription after introducing the vector into a host cell. It can also be produced by acquiring each of the above. In addition, by expressing a sense strand RNA of a specific sequence of the target gene, an antisense strand RNA consisting of a sequence complementary to the sense strand RNA, and a linker sequence connecting both strands, and expressing a shRNA that forms a hairpin structure, Desired double-stranded RNA can also be prepared.
 siRNAは、標的遺伝子の発現抑制活性を有する限りにおいては、siRNAを構成する核酸の一部がDNAであっても良い。また、siRNAは、標的遺伝子の発現抑制活性を有する限りにおいては、siRNAを構成する核酸の全体又はその一部が修飾された核酸であってもよい。
 修飾された核酸とは、ヌクレオシド(塩基部位、糖部位)及び/又はヌクレオシド間結合部位に修飾が施されていて、天然の核酸と異なった構造を有するものを意味する。修飾された核酸を構成する「修飾されたヌクレオシド」としては、例えば、無塩基(abasic)ヌクレオシド;アラビノヌクレオシド、2’-デオキシウリジン、α-デオキシリボヌクレオシド、β-L-デオキシリボヌクレオシド、その他の糖修飾を有するヌクレオシド;ペプチド核酸(PNA)、ホスフェート基が結合したペプチド核酸(PHONA)、ロックド核酸(LNA)、モルホリノ核酸等が挙げられる。前記糖修飾を有するヌクレオシドには、2’-O-メチルリボース、2’-デオキシ-2’-フルオロリボース、3’-O-メチルリボース等の置換五単糖;1’,2’-デオキシリボース;アラビノース;置換アラビノース糖;六単糖およびアルファ-アノマーの糖修飾を有するヌクレオシドが含まれる。これらのヌクレオシドは塩基部位が修飾された修飾塩基であってもよい。このような修飾塩基には、例えば、5-ヒドロキシシトシン、5-フルオロウラシル、4-チオウラシル等のピリミジン;6-メチルアデニン、6-チオグアノシン等のプリン;及び他の複素環塩基等が挙げられる。
 修飾された核酸を構成する「修飾されたヌクレオシド間結合」としては、例えば、アルキルリンカー、グリセリルリンカー、アミノリンカー、ポリ(エチレングリコール)結合、メチルホスホネートヌクレオシド間結合;メチルホスホノチオエート、ホスホトリエステル、ホスホチオトリエステル、ホスホロチオエート、ホスホロジチオエート、トリエステルプロドラッグ、スルホン、スルホンアミド、サルファメート、ホルムアセタール、N-メチルヒドロキシルアミン、カルボネート、カルバメート、モルホリノ、ボラノホスホネート、ホスホルアミデートなどの非天然ヌクレオシド間結合が挙げられる。
As long as the siRNA has an activity of suppressing the expression of the target gene, a part of the nucleic acid constituting the siRNA may be DNA. The siRNA may be a nucleic acid in which all or part of the nucleic acid constituting the siRNA is modified as long as it has the activity of suppressing the expression of the target gene.
The modified nucleic acid means a nucleic acid having a structure different from that of a natural nucleic acid, in which a nucleoside (base site, sugar site) and / or internucleoside binding site is modified. Examples of the “modified nucleoside” constituting the modified nucleic acid include an abasic nucleoside; an arabino nucleoside, 2′-deoxyuridine, α-deoxyribonucleoside, β-L-deoxyribonucleoside, and other sugars Examples include nucleosides having modifications; peptide nucleic acids (PNA), peptide nucleic acids to which phosphate groups are bound (PHONA), locked nucleic acids (LNA), morpholino nucleic acids and the like. Examples of the nucleoside having a sugar modification include substituted pentasaccharides such as 2′-O-methylribose, 2′-deoxy-2′-fluororibose, and 3′-O-methylribose; 1 ′, 2′-deoxyribose Arabinose; substituted arabinose sugars; nucleosides with hexose and alpha-anomeric sugar modifications. These nucleosides may be modified bases with modified base sites. Examples of such modified bases include pyrimidines such as 5-hydroxycytosine, 5-fluorouracil, 4-thiouracil; purines such as 6-methyladenine and 6-thioguanosine; and other heterocyclic bases.
Examples of the “modified internucleoside linkage” constituting the modified nucleic acid include, for example, alkyl linker, glyceryl linker, amino linker, poly (ethylene glycol) linkage, methylphosphonate internucleoside linkage; methylphosphonothioate, phosphotriester , Phosphothiotriester, phosphorothioate, phosphorodithioate, triester prodrug, sulfone, sulfonamide, sulfamate, formacetal, N-methylhydroxylamine, carbonate, carbamate, morpholino, boranophosphonate, phosphoramidate, etc. Non-natural internucleoside linkages.
 本発明の二本鎖siRNAに含まれる核酸配列としては、配列表の配列番号2~25に記載の配列を好ましく用いることができる。これらのsiRNAのヌクレオチド配列を表1に示す。表1中、大文字で示されるのはセンスRNA配列およびアンチセンスRNA配列であり、小文字で示されるのは3’末端オーバーハング配列である。
 例えば、表1の1番上の配列は、配列番号:2に示すセンス鎖と配列番号:3に示すアンチセンス鎖とからなる2本鎖siRNAであり、配列番号:2の3’末端のttおよび配列番号:3の3’末端のcaがオーバーハング配列である。
 これらのsiRNAは、COL1A1遺伝子の発現を顕著に抑制するので、これらのsiRNAを含むCOL1A1遺伝子が関連する疾患の治療用医薬組成物の効果は大きいものである。

As the nucleic acid sequence contained in the double-stranded siRNA of the present invention, the sequences described in SEQ ID NOs: 2 to 25 in the sequence listing can be preferably used. The nucleotide sequences of these siRNAs are shown in Table 1. In Table 1, the uppercase letters indicate the sense RNA sequence and the antisense RNA sequence, and the lowercase letters indicate the 3 ′ end overhang sequence.
For example, the first sequence in Table 1 is a double-stranded siRNA composed of a sense strand shown in SEQ ID NO: 2 and an antisense strand shown in SEQ ID NO: 3, and tt at the 3 ′ end of SEQ ID NO: 2 In addition, ca at the 3 ′ end of SEQ ID NO: 3 is an overhang sequence.
Since these siRNAs remarkably suppress the expression of the COL1A1 gene, the effect of the pharmaceutical composition for treating a disease associated with the COL1A1 gene containing these siRNAs is great.

 標的遺伝子のmRNAに対して相補的なオリゴヌクレオチドを「アンチセンスオリゴヌクレオチド」と呼び、当該アンチセンスオリゴヌクレオチドが標的とする遺伝子(mRNA)と二本鎖を形成することによりmRNAの働きを抑制する。「アンチセンスオリゴヌクレオチド」には、標的となる遺伝子(mRNA)と完全に相補的であるもののみならず、mRNAと安定にハイブリダイズできる限り、多少のミスマッチが存在してもよい。 Oligonucleotides complementary to the mRNA of the target gene are called “antisense oligonucleotides,” and the mRNA functions are suppressed by forming a double strand with the target gene (mRNA). . “Antisense oligonucleotides” are not limited to those that are completely complementary to the target gene (mRNA), but may contain some mismatches as long as they can be stably hybridized with mRNA.
 アンチセンスオリゴヌクレオチドは、修飾されていてもよい。適当な修飾を施すことにより、当該アンチセンスオリゴヌクレオチドは生体内で分解されにくくなり、より安定して標的遺伝子の発現を阻害できるようになる。このような修飾されたオリゴヌクレオチドとしては、S-オリゴ型(ホスホロチオエート型)、C-5チアゾール型、D-オリゴ型(ホスホジエステル型)、M-オリゴ型(メチルフォスフォネイト型)、ペプチド核酸型、リン酸ジエステル結合型、C-5プロピニルピリミジン型、2-O-プロピルリボース、2'-メトキシエトキシリボース型等の修飾型のアンチセンスオリゴヌクレオチドが挙げられる。さらに、アンチセンスオリゴヌクレオチドとしては、リン酸基を構成する酸素原子の少なくとも一部がイオウ原子に置換、修飾されているものでもよい。このようなアンチセンスオリゴヌクレオチドは、ヌクレアーゼ耐性、RNAへの親和性に特に優れている。リン酸基を構成する酸素原子の少なくとも一部がイオウ原子に置換、修飾されたアンチセンスオリゴヌクレオチドとしては、例えば、S-オリゴ型等のオリゴヌクレオチドが挙げられる。
 アンチセンスオリゴヌクレオチド(又はその誘導体)は常法によって合成することができ、例えば、市販のDNA合成装置(例えばAppliedBiosystems社製など)によって容易に合成することができる。合成法はホスホロアミダイトを用いた固相合成法、ハイドロジェンホスホネートを用いた固相合成法などがある。
Antisense oligonucleotides may be modified. By applying an appropriate modification, the antisense oligonucleotide becomes difficult to be degraded in the living body, and the expression of the target gene can be inhibited more stably. Such modified oligonucleotides include S-oligo type (phosphorothioate type), C-5 thiazole type, D-oligo type (phosphodiester type), M-oligo type (methyl phosphonate type), peptide nucleic acid And modified antisense oligonucleotides such as phosphodiester bond type, C-5 propynyl pyrimidine type, 2-O-propyl ribose, 2′-methoxyethoxy ribose type. Furthermore, the antisense oligonucleotide may be one in which at least a part of the oxygen atom constituting the phosphate group is substituted or modified with a sulfur atom. Such an antisense oligonucleotide is particularly excellent in nuclease resistance and affinity for RNA. Examples of the antisense oligonucleotide in which at least a part of the oxygen atom constituting the phosphate group is substituted or modified with a sulfur atom include oligonucleotides such as S-oligo type.
An antisense oligonucleotide (or a derivative thereof) can be synthesized by a conventional method, and can be easily synthesized by, for example, a commercially available DNA synthesizer (for example, Applied Biosystems). Examples of the synthesis method include a solid phase synthesis method using phosphoramidite and a solid phase synthesis method using hydrogen phosphonate.
 「リボザイム」とは核酸を切断する酵素活性を有するRNAをいうが、最近では当該酵素活性部位の塩基配列を有するオリゴDNAも同様に核酸切断活性を有することが明らかになっているので、本明細書では配列特異的な核酸切断活性を有する限りDNAをも包含する概念として用いる。具体的には、リボザイムは、標的遺伝子をコードするmRNAまたは初期転写産物を、コード領域の内部(初期転写産物の場合はイントロン部分を含む)で特異的に切断し得る。リボザイムで最も汎用性の高いものとしては、ウイロイドやウイルソイド等の感染性RNAに見られるセルフスプライシングRNAがあり、ハンマーヘッド型やヘアピン型等が知られている。ハンマーヘッド型は約40塩基程度で酵素活性を発揮し、ハンマーヘッド構造をとる部分に隣接する両端の数塩基ずつ(合わせて約10塩基程度)をmRNAの所望の切断部位と相補的な配列にすることにより、標的mRNAのみを特異的に切断することが可能である。さらに、リボザイムを、それをコードするDNAを含む発現ベクターの形態で使用する場合には、転写産物の細胞質への移行を促進するために、tRNAを改変した配列をさらに連結したハイブリッドリボザイムとすることもできる(Nucleic Acids Res., 29(13): 2780-2788 (2001))。 “Ribozyme” refers to RNA having an enzyme activity that cleaves nucleic acid. Recently, it has been clarified that oligoDNA having the base sequence of the enzyme active site also has a nucleic acid cleaving activity. In the book, it is used as a concept including DNA as long as it has sequence-specific nucleic acid cleavage activity. Specifically, the ribozyme can specifically cleave mRNA or an initial transcription product encoding a target gene within the coding region (including an intron portion in the case of the initial transcription product). The most versatile ribozyme is self-splicing RNA found in infectious RNA such as viroid and virusoid, and hammerhead type and hairpin type are known. The hammerhead type exhibits enzyme activity at about 40 bases, and several bases at both ends adjacent to the portion having the hammerhead structure (about 10 bases in total) are made complementary to the desired cleavage site of mRNA. By doing so, it is possible to specifically cleave only the target mRNA. Furthermore, when the ribozyme is used in the form of an expression vector containing the DNA encoding the ribozyme, in order to promote the transfer of the transcription product to the cytoplasm, it should be a hybrid ribozyme further linked with a tRNA-modified sequence. (Nucleic Acids Res., 29 (13): 2780-2788 (2001)).
 マイクロRNA(「miRNA」といわれる)は、タンパク質をコードしない低分子RNAであり、マイクロRNAは、標的mRNA上の相補部位に結合することによって遺伝子発現を制御する機能を有する分子である。 Micro RNA (referred to as “miRNA”) is a small RNA that does not encode a protein, and micro RNA is a molecule having a function of controlling gene expression by binding to a complementary site on a target mRNA.
 標的遺伝子の発現を抑制する物質は、siRNA、アンチセンスオリゴヌクレオチド、マイクロRNA又はリボザイムなどの核酸分子および、該核酸分子をコードする発現ベクターでもよい。当該発現ベクターは、上記の核酸分子をコードするオリゴヌクレオチドもしくはポリヌクレオチドが、投与対象である哺乳動物の細胞内でプロモーター活性を発揮し得るプロモーターに機能的に連結されていなければならない。使用されるプロモーターは、投与対象である哺乳動物で機能し得るものであれば特に制限はないが、例えば、polIIIプロモーター(例、tRNAプロモーター、U6プロモーター、H1プロモーター)、哺乳動物用プロモーター(例、CMVプロモーター、CAGプロモーター、SV40プロモーター)などが挙げられる。
 発現ベクターは、好ましくは核酸分子をコードするオリゴ(ポリ)ヌクレオチドの下流に転写終結シグナル、すなわちターミネーター領域を含有する。さらに、形質転換細胞選択のための選択マーカー遺伝子(テトラサイクリン、アンピシリン、カナマイシン、ハイグロマイシン、ホスフィノスリシン等の薬剤に対する抵抗性を付与する遺伝子、栄養要求性変異を相補する遺伝子等)をさらに含有することもできる。
 発現ベクターとして使用される基本骨格のベクターは特に制限されないが、例えば、プラスミドベクター、ウイルスベクターが挙げられる。ヒト等の哺乳動物への投与に好適なベクターとしては、レトロウイルス、アデノウイルス、アデノ随伴ウイルス、ヘルペスウイルス、ワクシニアウイルス、ポックスウイルス、ポリオウイルス、シンドビスウイルス、センダイウイルス等のウイルスベクターが挙げられる。
The substance that suppresses the expression of the target gene may be a nucleic acid molecule such as siRNA, antisense oligonucleotide, microRNA or ribozyme and an expression vector encoding the nucleic acid molecule. In the expression vector, the oligonucleotide or polynucleotide encoding the nucleic acid molecule must be operably linked to a promoter capable of exhibiting promoter activity in a mammalian cell to be administered. The promoter to be used is not particularly limited as long as it can function in the mammal to be administered, but for example, polIII promoter (eg, tRNA promoter, U6 promoter, H1 promoter), mammalian promoter (eg, CMV promoter, CAG promoter, SV40 promoter) and the like.
The expression vector preferably contains a transcription termination signal, ie a terminator region, downstream of the oligo (poly) nucleotide encoding the nucleic acid molecule. In addition, selectable marker genes for selection of transformed cells (such as genes that confer resistance to drugs such as tetracycline, ampicillin, kanamycin, hygromycin, phosphinothricin, genes that complement auxotrophic mutations, etc.) You can also
The basic backbone vector used as the expression vector is not particularly limited, and examples thereof include a plasmid vector and a viral vector. Suitable vectors for administration to mammals such as humans include viral vectors such as retrovirus, adenovirus, adeno-associated virus, herpes virus, vaccinia virus, poxvirus, poliovirus, Sindbis virus, Sendai virus, and the like. .
 本発明の発現ベクターとしては、siRNA、アンチセンスオリゴヌクレオチド、マイクロRNA又はリボザイムの発現ベクターが例示されるが、其の中でも、siRNA又はアンチセンスオリゴヌクレオチドの発現ベクターが好ましい。
 本発明のsiRNAの発現ベクターがコードする核酸配列としては、表1に記載の配列が好ましい。表1記載のsiRNAは、COL1A1遺伝子の発現を顕著に抑制するので、これらのsiRNAを含むCOL1A1遺伝子が関連する疾患の治療用医薬組成物の効果は大きいものである。
Examples of the expression vector of the present invention include siRNA, antisense oligonucleotide, microRNA, and ribozyme expression vectors. Among them, siRNA or antisense oligonucleotide expression vectors are preferable.
The nucleic acid sequence encoded by the expression vector for siRNA of the present invention is preferably the sequence shown in Table 1. Since the siRNA shown in Table 1 remarkably suppresses the expression of the COL1A1 gene, the effect of the pharmaceutical composition for treating a disease associated with the COL1A1 gene containing these siRNAs is great.
 本発明において、GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質は、医薬組成物の有効成分として使用することができる。本発明の医薬組成物は、当該医薬組成物を生体内に投与することにより、COL1A1遺伝子が関連する疾患の治療用医薬組成物として使用することができる。 In the present invention, a substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or a substance that suppresses the activity of the protein encoded by these genes can be used as an active ingredient of a pharmaceutical composition. The pharmaceutical composition of the present invention can be used as a pharmaceutical composition for treating a disease associated with the COL1A1 gene by administering the pharmaceutical composition in vivo.
 本発明の医薬組成物は、単一の遺伝子発現抑制物質を有効成分としてもよいし、複数の遺伝子発現抑制物質を有効成分としても良い。例えば、本発明の医薬組成物の発現抑制物質がsiRNAである場合には、1種またはそれ以上のsiRNAを有効成分としてもよい。 In the pharmaceutical composition of the present invention, a single gene expression inhibitor may be used as an active ingredient, or a plurality of gene expression inhibitors may be used as active ingredients. For example, when the expression inhibitory substance of the pharmaceutical composition of the present invention is siRNA, one or more siRNAs may be used as the active ingredient.
 本発明の医薬組成物の治療の対象である疾患の種類は、COL1A1遺伝子が関連する疾患である。COL1A1遺伝子が関連する疾患の例として、慢性腎不全、慢性腎炎、尿細管間質障害、ネフローゼ症候群、多発性のう胞腎、糖尿病性腎症、及び腎硬化症などの慢性腎臓病に包含される疾患、並びに強皮症、肥厚性瘢痕、瘢痕拘縮、ケロイド症、膠原病、肺線維症、繊維性癒着、珪肺、骨髄線維症、腎性全身性線維症、クローン病、心筋線維症、及び肝硬変などの線維化疾患に包含される疾患を挙げることができる。
 ここで、慢性腎臓病とは、「尿蛋白陽性などの腎疾患の存在する所見」、もしくは「腎機能低下(糸球体濾過量が60mL/min/1.73m2未満)」が3ヵ月以上続く状態を指す。
The type of disease that is the subject of treatment with the pharmaceutical composition of the present invention is a disease associated with the COL1A1 gene. Examples of diseases associated with the COL1A1 gene include chronic kidney disease such as chronic renal failure, chronic nephritis, tubulointerstitial disorder, nephrotic syndrome, polycystic kidney disease, diabetic nephropathy, and nephrosclerosis , And scleroderma, hypertrophic scar, scar contracture, keloidosis, collagen disease, pulmonary fibrosis, fibrosis, silicosis, myelofibrosis, renal systemic fibrosis, Crohn's disease, myocardial fibrosis, and cirrhosis Diseases included in fibrotic diseases such as
Here, chronic kidney disease is a condition in which “finding of kidney disease such as urine protein positivity” or “reduced renal function (glomerular filtration rate less than 60 mL / min / 1.73 m2)” continues for 3 months or more. Point to.
 本発明の医薬組成物は、経口投与及び非経口投与のいずれの剤形をも採用することができる。 The pharmaceutical composition of the present invention can employ both oral and parenteral dosage forms.
 本発明の医薬組成物は常法にしたがって製剤化することができ、医薬的に許容される担体や添加物を含むものであってもよい。このような担体及び添加物として、水、医薬的に許容される有機溶剤、コラーゲン、ポリビニルアルコール、ポリビニルピロリドン、カルボキシビニルポリマー、カルボキシメチルセルロースナトリウム、ポリアクリル酸ナトリウム、アルギン酸ナトリウム、水溶性デキストラン、カルボキシメチルスターチナトリウム、ペクチン、メチルセルロース、エチルセルロース、キサンタンガム、アラビアゴム、カゼイン、寒天、ポリエチレングリコール、ジグリセリン、グリセリン、プロピレングリコール、ワセリン、パラフィン、ステアリルアルコール、ステアリン酸、ヒト血清アルブミン、マンニトール、ソルビトール、ラクトース、医薬添加物として許容される界面活性剤等が挙げられる。
 添加物は、本発明の医薬組成物の剤形に応じて上記の中から単独で又は適宜組み合わせて選ばれる。剤形としては、経口投与の場合は、錠剤、カプセル剤、細粒剤、粉末剤、顆粒剤、液剤、シロップ剤等として、または適当な剤形により投与が可能である。非経口投与の場合は、経肺剤形(例えばネフライザーなどを用いたもの)、経鼻投与剤形、経皮投与剤形(例えば軟膏、クリーム剤)、注射剤形等が挙げられる。注射剤形の場合は、例えば点滴等の静脈内注射、筋肉内注射、腹腔内注射、皮下注射等により全身又は局部的に投与することができる。
The pharmaceutical composition of the present invention can be formulated according to a conventional method, and may contain a pharmaceutically acceptable carrier or additive. Such carriers and additives include water, pharmaceutically acceptable organic solvents, collagen, polyvinyl alcohol, polyvinyl pyrrolidone, carboxyvinyl polymer, sodium carboxymethylcellulose, sodium polyacrylate, sodium alginate, water-soluble dextran, carboxymethyl. Sodium starch, pectin, methylcellulose, ethylcellulose, xanthan gum, gum arabic, casein, agar, polyethylene glycol, diglycerin, glycerin, propylene glycol, petrolatum, paraffin, stearyl alcohol, stearic acid, human serum albumin, mannitol, sorbitol, lactose, pharmaceutical Examples of acceptable surfactants include additives.
The additive is selected from the above alone or in appropriate combination depending on the dosage form of the pharmaceutical composition of the present invention. As for the dosage form, in the case of oral administration, it can be administered as a tablet, capsule, fine granule, powder, granule, liquid, syrup or the like, or in an appropriate dosage form. In the case of parenteral administration, pulmonary dosage forms (for example, those using a nephriser etc.), nasal dosage forms, transdermal dosage forms (for example, ointments, creams), injection dosage forms and the like can be mentioned. In the case of an injection dosage form, it can be administered systemically or locally by intravenous injection such as infusion, intramuscular injection, intraperitoneal injection, subcutaneous injection or the like.
 標的遺伝子の発現を抑制する物質がsiRNA、アンチセンスオリゴヌクレオチド、マイクロRNA若しくはリボザイムなどの核酸分子、又は該核酸分子をコードする発現ベクターである場合は、リポソームなどのリン脂質小胞体に当該発現抑制物質を導入し、その小胞体を本発明の医薬組成物とすることも可能である。 When the substance that suppresses the expression of the target gene is a nucleic acid molecule such as siRNA, antisense oligonucleotide, microRNA or ribozyme, or an expression vector encoding the nucleic acid molecule, the expression is suppressed in phospholipid vesicles such as liposomes. It is also possible to introduce a substance and use the endoplasmic reticulum as the pharmaceutical composition of the present invention.
 本発明の医薬組成物の投与量は、年齢、性別、症状、投与経路、投与回数、剤形によって異なる。投与方法は、患者の年齢、症状により適宜選択する。有効投与量は、一回につき体重1kgあたり0.01μg~1000mg、好ましくは0.1μg~100μgである。但し、上記治療剤はこれらの投与量に制限されるものではない。 The dosage of the pharmaceutical composition of the present invention varies depending on age, sex, symptoms, administration route, administration frequency, and dosage form. The administration method is appropriately selected depending on the age and symptoms of the patient. The effective dose is 0.01 μg to 1000 mg, preferably 0.1 μg to 100 μg, per kg body weight. However, the therapeutic agent is not limited to these doses.
 本発明は、さらなる態様において、
(1)GALNT2、COL8A2、FRYL又は TIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質を投与する工程を有する、COL1A1遺伝子が関連する疾患を治療する方法、
(2)COL1A1遺伝子が関連する疾患の治療に用いられる、GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質、
(3)COL1A1遺伝子が関連する疾患の治療剤の製造のための、GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質の使用、
(4)GALNT2、COL8A2、FRYL又は TIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質を投与する工程を有する、慢性腎臓病又は線維化疾患を治療する方法、
(5)慢性腎臓病又は線維化疾患の治療に用いられる、GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質、ならびに
(6)慢性腎臓病又は線維化疾患の治療剤の製造のための、GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質の使用
を提供する。
 上記「遺伝子の発現を抑制する物質」「遺伝子がコードするタンパク質の活性を抑制する物質」については、先述のとおりである。
The invention in a further aspect,
(1) A method for treating a disease associated with the COL1A1 gene, comprising a step of administering a substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or a substance that suppresses the activity of a protein encoded by these genes,
(2) A substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or the activity of a protein encoded by these genes, which is used for treatment of a disease associated with COL1A1 gene,
(3) Use of a substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or a substance that suppresses the activity of a protein encoded by these genes for the manufacture of a therapeutic agent for a disease associated with the COL1A1 gene,
(4) a method for treating chronic kidney disease or fibrotic disease, comprising a step of administering a substance that suppresses the expression of GALNT2, COL8A2, FRYL or TIMP1 gene or a substance that suppresses the activity of a protein encoded by these genes;
(5) a substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or a substance that suppresses the activity of a protein encoded by these genes, and (6) chronic that is used for the treatment of chronic kidney disease or fibrotic disease There is provided use of a substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or a substance that suppresses the activity of a protein encoded by these genes for the manufacture of a therapeutic agent for kidney disease or fibrotic disease.
The “substance that suppresses gene expression” and “substance that suppresses the activity of the protein encoded by the gene” are as described above.
 本発明は、もう1つの態様において、GALNT2、COL8A2、FRYL若しくはTIMP1遺伝子の発現の抑制又はこれらの遺伝子がコードするタンパク質の活性抑制を指標とするCOL1A1遺伝子が関連する疾患の治療物質をスクリーニングする方法を提供する。 In another aspect, the present invention relates to a method for screening a therapeutic agent for a disease associated with the COL1A1 gene, which is based on suppression of expression of the GALNT2, COL8A2, FRYL, or TIMP1 gene, or suppression of the activity of a protein encoded by these genes. I will provide a.
 スクリーニング方法に供される被験物質は、いかなる公知化合物および新規化合物であってもよく、例えば、核酸、糖質、脂質、蛋白質、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等が挙げられる。 The test substance to be subjected to the screening method may be any known compound and novel compound, for example, nucleic acid, carbohydrate, lipid, protein, peptide, low molecular organic compound, compound prepared using combinatorial chemistry technology Examples include libraries, random peptide libraries prepared by solid phase synthesis and phage display methods, or natural components derived from microorganisms, animals and plants, marine organisms, and the like.
 一実施形態では、本発明のスクリーニング方法は、
(1)細胞と被験物質とを接触させる工程、
(2)被験物質を接触させた細胞におけるGALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現量を測定し、該発現量を、被験物質を接触させない対照細胞における前記遺伝子の発現量と比較する工程、及び
(3)被験物質を接触させた細胞における前記遺伝子の発現が、対照細胞における前記遺伝子の発現よりも低下している場合に、被験物質をCOL1A1遺伝子が関連する疾患の治療物質として選択する工程を含む、
COL1A1遺伝子が関連する疾患の治療物質のスクリーニング方法である。
In one embodiment, the screening method of the present invention comprises:
(1) a step of bringing cells into contact with a test substance;
(2) measuring the expression level of the GALNT2, COL8A2, FRYL, or TIMP1 gene in a cell contacted with the test substance, and comparing the expression level with the expression level of the gene in a control cell not contacted with the test substance; (3) selecting the test substance as a therapeutic substance for a disease associated with the COL1A1 gene when the expression of the gene in the cell contacted with the test substance is lower than the expression of the gene in a control cell; Including,
This is a screening method for a therapeutic substance for a disease associated with the COL1A1 gene.
 上記方法の工程(1)における、細胞とは、GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を測定することが可能な細胞のことを意味する。該工程では、細胞と被験物質とが培養培地中で接触条件下におかれる。 The cell in step (1) of the above method means a cell capable of measuring the expression of GALNT2, COL8A2, FRYL or TIMP1 gene. In this step, cells and a test substance are placed in contact with each other in a culture medium.
 GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を測定することが可能な細胞とは、GALNT2、COL8A2、FRYL又はTIMP1遺伝子の産物、例えば、転写産物、翻訳産物の発現レベルを直接的または間接的に評価可能な細胞をいう。該遺伝子の産物の発現レベルを直接的に評価可能な細胞は、該遺伝子を天然で発現可能な細胞であり得、一方、該遺伝子の産物の発現レベルを間接的に評価可能な細胞としては、該遺伝子転写調節領域についてレポーターアッセイを可能とする細胞などが挙げられる。該遺伝子の発現を測定可能な細胞は、動物細胞、例えばマウス、ラット、ハムスター、モルモット、ウサギ、イヌ、サルあるいはヒトの哺乳動物細胞を用いることができ、ヒト由来の細胞、なかでも、ヒト尿細管上皮細胞株HK2細胞やヒト・プライマリー尿細管上皮細胞が好ましい。 Cells capable of measuring the expression of GALNT2, COL8A2, FRYL or TIMP1 gene are directly or indirectly evaluated for the expression level of GALNT2, COL8A2, FRYL or TIMP1 gene products such as transcripts and translation products. A possible cell. A cell capable of directly evaluating the expression level of the gene product can be a cell capable of naturally expressing the gene, while a cell capable of indirectly evaluating the expression level of the gene product includes: Examples include cells that allow reporter assay for the gene transcription regulatory region. As the cells capable of measuring the expression of the gene, animal cells such as mice, rats, hamsters, guinea pigs, rabbits, dogs, monkeys or human mammalian cells can be used. Human-derived cells, particularly human urine Tubular epithelial cell line HK2 cells and human primary tubular epithelial cells are preferred.
 遺伝子転写調節領域についてレポーターアッセイを可能とする細胞は、標的遺伝子転写調節領域、当該領域に機能可能に連結されたレポーター遺伝子を含む細胞である。標的遺伝子転写調節領域およびレポーター遺伝子は、発現ベクター中に挿入することが出来る。標的遺伝子の転写調節領域は、標的遺伝子の発現を制御し得る領域である限り特に限定されないが、例えば、転写開始点から上流約2kbpまでの領域、あるいは該領域の塩基配列において1以上の塩基が欠失、置換若しくは付加された塩基配列からなり、且つ標的遺伝子の転写を制御する能力を有する領域などが挙げられる。レポーター遺伝子は、検出可能な蛋白質または検出可能な物質を生成する酵素をコードする遺伝子であればよく、例えばGFP(緑色蛍光蛋白質)遺伝子、GUS(β-グルクロニダーゼ)遺伝子、LUC(ルシフェラーゼ)遺伝子、CAT(クロラムフェニコルアセチルトランスフェラーゼ)遺伝子等が挙げられる。 A cell enabling a reporter assay for a gene transcription regulatory region is a cell containing a target gene transcription regulatory region and a reporter gene operably linked to the region. The target gene transcription regulatory region and the reporter gene can be inserted into an expression vector. The transcriptional regulatory region of the target gene is not particularly limited as long as it can control the expression of the target gene. For example, a region from the transcription start point to about 2 kbp upstream, or one or more bases in the base sequence of the region Examples include a region consisting of a base sequence deleted, substituted or added and having the ability to control the transcription of the target gene. The reporter gene may be any gene that encodes a detectable protein or an enzyme that produces a detectable substance. For example, the GFP (green fluorescent protein) gene, GUS (β-glucuronidase) gene, LUC (luciferase) gene, CAT (Chloramphenicol acetyltransferase) gene and the like.
 遺伝子転写調節領域、当該領域に機能可能に連結されたレポーター遺伝子が導入される細胞は、標的となる遺伝子の転写調節機能を評価できる限り、即ち、該レポーター遺伝子の発現量が定量的に解析可能である限り特に限定されない。 A cell into which a gene transcription regulatory region and a reporter gene operably linked to the region are introduced can be used for quantitative analysis of the expression level of the reporter gene as long as the transcriptional regulatory function of the target gene can be evaluated. As long as it is, it is not particularly limited.
 細胞と被験物質とが接触される培養培地は、用いられる細胞の種類などに応じて適宜選択されるが、例えば、約5~20%のウシ胎仔血清を含む最少必須培地(MEM)、ダルベッコ改変最少必須培地(DMEM)、RPMI1640培地、199培地などである。培養条件もまた、用いられる細胞の種類などに応じて適宜決定されるが、例えば、培地のpHは約6~約8であり、培養温度は通常約30~約40℃であり、培養時間は約12~約144時間である。 The culture medium in which the cells and the test substance are brought into contact is appropriately selected depending on the type of cells used. For example, a minimal essential medium (MEM) containing about 5 to 20% fetal bovine serum, Dulbecco's modification Minimum essential medium (DMEM), RPMI 1640 medium, 199 medium, and the like. The culture conditions are also appropriately determined according to the type of cells to be used. For example, the pH of the medium is about 6 to about 8, the culture temperature is usually about 30 to about 40 ° C., and the culture time is About 12 to about 144 hours.
 上記方法の工程(2)では、先ず、被験物質を接触させた細胞におけるGALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現量が測定される。発現量の測定は、用いた細胞の種類などを考慮し、自体公知の方法により行われ得る。例えば、該遺伝子の発現を測定可能な細胞として、GALNT2、COL8A2、FRYL又はTIMP1遺伝子を天然で発現可能な細胞を用いた場合、発現量は、遺伝子の産物、例えば、転写産物または翻訳産物を対象として自体公知の方法により測定できる。例えば、転写産物の発現量は、細胞からtotal RNAを調製し、RT-PCR、ノーザンブロッティング等により測定され得る。また、翻訳産物の発現量は、細胞から抽出液を調製し、免疫学的手法により測定され得る。免疫学的手法としては、放射性同位元素免疫測定法(RIA法)、ELISA法(Methods in Enzymol. 70: 419-439 (1980))、蛍光抗体法などが使用できる。一方、GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を測定可能な細胞として、遺伝子転写調節領域についてレポーターアッセイを可能とする細胞を用いた場合、発現量は、レポーターのシグナル強度に基づき測定され得る。 In step (2) of the above method, first, the expression level of the GALNT2, COL8A2, FRYL, or TIMP1 gene in the cells contacted with the test substance is measured. The expression level can be measured by a method known per se in consideration of the type of cells used. For example, when a cell capable of naturally expressing the GALNT2, COL8A2, FRYL or TIMP1 gene is used as a cell capable of measuring the expression of the gene, the expression level is a gene product such as a transcription product or a translation product. Can be measured by a method known per se. For example, the expression level of the transcript can be measured by preparing total RNA from cells and performing RT-PCR, Northern blotting, or the like. The expression level of the translation product can be measured by preparing an extract from the cells and using an immunological technique. As an immunological technique, a radioisotope immunoassay (RIA method), an ELISA method (Methods in Enzymol. 70: 419-439 (1980)), a fluorescent antibody method, or the like can be used. On the other hand, when a cell capable of measuring a gene transcription regulatory region is used as a cell capable of measuring the expression of GALNT2, COL8A2, FRYL or TIMP1 gene, the expression level can be measured based on the signal intensity of the reporter.
 上記方法の工程(3)では、被験物質を接触させた細胞における該遺伝子の発現量が、対照細胞における遺伝子の発現量と比較される。
 発現量の比較は、好ましくは、有意差の有無に基づいて行なわれる。被験物質を接触させない対照細胞における遺伝子の発現量は、被験物質を接触させた細胞における遺伝子の発現量の測定に対し、事前に測定した発現量であっても、同時に測定した発現量であってもよいが、実験の精度、再現性の観点から同時に測定した発現量であることが好ましい。
In step (3) of the above method, the expression level of the gene in the cell contacted with the test substance is compared with the expression level of the gene in the control cell.
The comparison of expression levels is preferably performed based on the presence or absence of a significant difference. The expression level of the gene in the control cell not contacted with the test substance is the expression level measured at the same time, even if it is the expression level measured in advance compared to the measurement of the gene expression level in the cell contacted with the test substance. However, the expression level is preferably measured simultaneously from the viewpoint of the accuracy and reproducibility of the experiment.
 他の一実施形態では、本発明のスクリーニング方法は、
配列番号1で表されるアミノ酸配列を有するポリペプチド、又は配列番号1で表されるアミノ酸配列の1又は数個のアミノ酸が欠失、置換、及び/若しくは付加されたアミノ酸配列を有し、N-アセチルガラクトサミン転移活性を有するポリペプチドを用いることを特徴とするCOL1A1遺伝子が関連する疾患の治療物質のスクリーニング方法である。
In another embodiment, the screening method of the present invention comprises:
A polypeptide having the amino acid sequence represented by SEQ ID NO: 1, or an amino acid sequence in which one or several amino acids of the amino acid sequence represented by SEQ ID NO: 1 are deleted, substituted, and / or added; A method for screening a therapeutic substance for a disease associated with the COL1A1 gene, which comprises using a polypeptide having acetylgalactosamine transfer activity.
 また、他の一実施形態では、本発明のスクリーニング方法は、
(1)GALNT2タンパク質に対する基質、被験物質及び
1:配列番号1で表されるアミノ酸配列を有するポリペプチド、又は
2:配列番号1で表されるアミノ酸配列の1又は数個のアミノ酸が欠失、置換、及び/若しくは付加されたアミノ酸配列を有し、N-アセチルガラクトサミン転移活性を有するポリペプチドを接触させる工程、
(2)被験物質を接触させたポリペプチドのN-アセチルガラクトサミン転移活性を、被験物質を接触させないポリペプチドのN-アセチルガラクトサミン転移活性と比較する工程、及び
(3)被験物質を接触させたポリペプチドのN-アセチルガラクトサミン転移活性が、被験物質を接触させないポリペプチドのN-アセチルガラクトサミン転移活性と比較して低下している場合に、被験物質をCOL1A1遺伝子が関連する疾患の治療物質として選択する工程を含む。
In another embodiment, the screening method of the present invention comprises:
(1) a substrate for GALNT2 protein, a test substance and 1: a polypeptide having the amino acid sequence represented by SEQ ID NO: 1, or 2: deletion of one or several amino acids of the amino acid sequence represented by SEQ ID NO: 1, Contacting a polypeptide having a substituted and / or added amino acid sequence and having N-acetylgalactosamine transfer activity;
(2) a step of comparing the N-acetylgalactosamine transfer activity of the polypeptide contacted with the test substance with the N-acetylgalactosamine transfer activity of the polypeptide not contacted with the test substance, and (3) a poly contacted with the test substance. When the N-acetylgalactosamine transfer activity of a peptide is reduced compared to the N-acetylgalactosamine transfer activity of a polypeptide that does not contact the test substance, the test substance is selected as a therapeutic agent for a disease associated with the COL1A1 gene Process.
 上記方法の工程(1)における、GALNT2タンパク質に対する基質として用いることができるタンパク質は先述したとおりである。また、配列番号1で表されるアミノ酸配列を有するポリペプチドは、単離されたポリペプチド、膜画分中のポリペプチド及び細胞内のポリペプチドを含む。 The protein that can be used as a substrate for the GALNT2 protein in step (1) of the above method is as described above. The polypeptide having the amino acid sequence represented by SEQ ID NO: 1 includes an isolated polypeptide, a polypeptide in a membrane fraction, and an intracellular polypeptide.
 以下、実施例により本発明を具体的に説明するが、本発明はこれらの実施例に限定されるものではない。 Hereinafter, the present invention will be specifically described by way of examples, but the present invention is not limited to these examples.
 阻害することで線維化関連遺伝子の発現を抑制可能な遺伝子を見出す目的で、以下のスクリーニングを実施した。
 まず、スクリーニング対象遺伝子として、ヒト全遺伝子の中から創薬標的となる可能性の高い遺伝子群を発現情報により絞り込んだ。具体的には、ヒト27正常組織の発現データベースにおいて、心臓と脳、脾臓、末梢白血球のすべてで相対発現量が基準値以下の遺伝子群を、副作用懸念の比較的少ない遺伝子として抽出し、10,723遺伝子を得た。
 次に、擬陽性の確率を低減するため、一次スクリーニングに使用するヒト尿細管上皮細胞株HK2から全RNAを抽出し、DNAマイクロアレイ解析を行い、同細胞にて発現が検出できないレベルの遺伝子を除去することで、対象を7,700遺伝子に絞り込んだ。
 上記7,700遺伝子の中から、低分子化合物で活性を調節できる可能性が高そうなものや、高分子医薬の可能性が高そうなターゲットとなる、1,994遺伝子を選定した。
The following screening was performed with the aim of finding genes that can suppress the expression of fibrosis-related genes by inhibition.
First, as a screening target gene, a group of genes that are highly likely to be drug discovery targets was narrowed down by expression information from all human genes. Specifically, in the expression database of 27 human normal tissues, a group of genes whose relative expression level is below the reference value in all of the heart, brain, spleen, and peripheral leukocytes is extracted as genes with relatively few side effects, and 10,723 genes Got.
Next, in order to reduce the probability of false positives, total RNA is extracted from the human tubular epithelial cell line HK2 used for primary screening, and DNA microarray analysis is performed to remove genes whose levels cannot be detected in the same cells. This narrowed the target to 7,700 genes.
From the above 7,700 genes, 1,994 genes were selected that are likely to be able to regulate activity with low molecular weight compounds and targets that are likely to be high molecular drugs.
 一次スクリーニングとして以下の実験を実施した。ヒト全遺伝子siRNAライブラリー(Silencer Select Human Genome siRNA Library;Ambion 社製;21,585遺伝子;64,755 siRNA;3 siRNA/遺伝子)から、上記1,994遺伝子のフォーカストsiRNA ライブラリー(5,982 siRNAs)をチェリーピッキングにより作製した。同フォーカストsiRNA ライブラリーを用い、ヒト尿細管上皮細胞株HK2において発現抑制することでCOL1A1遺伝子の発現が低下する遺伝子を探索した。スクリーニングのプロトコールとしては、まず、Poly-D-Lysine(PDL)でコートした384ウェルプレート(Corning社製)に、0.25μMのライブラリーsiRNAを1.5μl(0.375 pmol, final 12.5 nM)ずつスポットした。また、各プレートに陰性コントロールとしてNegative Control #1 siRNA(NT-1;Ambion社製)も同量分注し、ヒットクライテリア設定に使用した。
 一方で、Opti-MEM I(Gibco社製)3.5μlとsiRNA導入試薬であるRNAiMAX(Invitrogen社製)0.1μlを混合し、室温で5分間保持した後、前述のsiRNAを分注した384ウェルプレートに添加した。室温で20分間インキュベーションした後、HK2細胞を5,000個含むKeratinocyte Serum Free Media(Invitrogen社製、0.05 mg/ml ウシ脳下垂体抽出物と5 ng/ml human rEGFを含む)を25μl添加し、37℃、5%CO2環境下で48時間培養した。その後、20 ng/ml の濃度でrecombinant TGFβ(R&D社製)を含むKeratinocyte Serum Free Mediaを30μl添加し、さらに24時間培養した。
 培養上清をデカンテーションにより除去し、FastLane Cell SYBR Green Kit(Qiagen社製)に含まれるBuffer FCW(Wash buffer)を25μl添加した。デカンデーションによりBuffer FCWを除去後、同じくKitに含まれるCell Processing Mix(Lysis buffer)を15μl加え、室温で5分間保持し、細胞を溶解した。細胞溶解液を75℃で5分間加熱したものを定量PCRのテンプレートとして用いた。定量PCRではFastLane Cell SYBR Green Kitを使用し、線維化関連遺伝子としてCOL1A1遺伝子を、内部標準としてGAPDH遺伝子およびPPIA遺伝子の発現レベルを測定した。各遺伝子を増幅するためのPCRプライマーはいずれもタカラバイオ社より購入したものを用いた(カタログ番号 ; 5060D-40407, プライマーセットID ; COL1A1,CH000497 ;GAPDH,HA067812;PPIA,HA067810)。384ウェルタイプの定量PCRプレート(Applied Biosystems社製)に、上記テンプレート1.0μlと 2x QuantiTect SYBR Green RT-PCR Master Mix 5.0μl、Forward primer 5 pmol、Reverse primer 5 pmol、QuantiTect RT Mix 0.1μlを含むPCR反応溶液を調製し、7900HT Fast Real Time PCR System(Applied Biosystems社製)を用いて定量PCRを実施した。
 得られた Cycle thresholdの値(Ct値)を基に、GAPDH遺伝子による補正もしくはPPIA遺伝子による補正のいずれかで、同一プレート上の陰性コントロールsiRNAに対してCOL1A1遺伝子の発現が50%以下に低下しているsiRNAをヒット候補siRNAとして抽出した。そして、同一遺伝子に対する3配列のsiRNAのうち2配列以上がヒット候補となっている遺伝子を、一次スクリーニングのヒット候補とし、158遺伝子を得た。
The following experiment was performed as a primary screening. From the human whole gene siRNA library (Silencer Select Human Genome siRNA Library; manufactured by Ambion; 21,585 gene; 64,755 siRNA; 3 siRNA / gene), the focused siRNA library (5,982 siRNAs) of the above 1,994 gene was prepared by cherry picking . Using the focused siRNA library, we searched for a gene whose expression of COL1A1 gene is reduced by suppressing expression in human tubular epithelial cell line HK2. As a screening protocol, first, 1.5 μl (0.375 pmol, final 12.5 nM) of 0.25 μM library siRNA was spotted on a 384 well plate (Corning) coated with Poly-D-Lysine (PDL). In addition, Negative Control # 1 siRNA (NT-1; manufactured by Ambion) was dispensed in the same amount on each plate as a negative control, and used for hit criteria setting.
Meanwhile, Opti-MEM I (Gibco) 3.5 μl and siRNA introduction reagent RNAiMAX (Invitrogen) 0.1 μl were mixed, kept at room temperature for 5 minutes, and then 384-well plate dispensed with the above siRNA Added to. After incubation at room temperature for 20 minutes, add 25 μl of Keratinocyte Serum Free Media (Invitrogen, 0.05 mg / ml bovine pituitary extract and 5 ng / ml human rEGF) containing 5,000 HK2 cells, 37 ° C The cells were cultured for 48 hours in a 5% CO2 environment. Thereafter, 30 μl of Keratinocyte Serum Free Media containing recombinant TGFβ (manufactured by R & D) at a concentration of 20 ng / ml was added and further cultured for 24 hours.
The culture supernatant was removed by decantation, and 25 μl of Buffer FCW (Wash buffer) contained in FastLane Cell SYBR Green Kit (manufactured by Qiagen) was added. After removing Buffer FCW by decantation, 15 μl of Cell Processing Mix (Lysis buffer) also contained in Kit was added and kept at room temperature for 5 minutes to lyse the cells. A cell lysate heated at 75 ° C. for 5 minutes was used as a template for quantitative PCR. For quantitative PCR, FastLane Cell SYBR Green Kit was used, and the expression levels of COL1A1 gene as fibrosis-related gene and GAPDH gene and PPIA gene as internal standards were measured. The PCR primers used to amplify each gene were purchased from Takara Bio Inc. (catalog number; 5060D-40407, primer set ID; COL1A1, CH000497; GAPDH, HA067812; PPIA, HA067810). PCR containing 1.0 μl of the above template and 5.0 μl of 2x QuantiTect SYBR Green RT-PCR Master Mix, Forward primer 5 pmol, Reverse primer 5 pmol, QuantiTect RT Mix 0.1 μl on a 384-well quantitative PCR plate (Applied Biosystems) A reaction solution was prepared, and quantitative PCR was performed using 7900HT Fast Real Time PCR System (Applied Biosystems).
Based on the obtained Cycle threshold value (Ct value), the expression of COL1A1 gene is reduced to 50% or less compared to the negative control siRNA on the same plate by either GAPDH gene correction or PPIA gene correction. SiRNAs were extracted as hit candidate siRNAs. Then, 158 genes were obtained by using, as a primary screening hit candidate, a gene in which two or more of the three sequences of siRNAs for the same gene were hit candidates.
 次に、再現性試験を実施した。上記158遺伝子のフォーカストsiRNAライブラリーを作製し(474 siRNA,3 siRNAs/遺伝子)、一次スクリーニングと同様なプロトコールにより各遺伝子をsiRNAで発現抑制した際のCOL1A1遺伝子の発現低下度合いを評価した。なお、再現性試験はn = 3で行い、ヒットsiRNAのクライテリアは、陰性コントロールに対して、COL1A1遺伝子の発現を40%減少させているものとした。結果、COL8A2、GALNT2、FRYL及びTIMP1遺伝子の4つの遺伝子について再現性を確認し、当該遺伝子群を一次スクリーニングのヒット遺伝子とした。
 一次スクリーニングの再現性試験の結果を、表2に記載する。

Figure JPOXMLDOC01-appb-T000002
Next, a reproducibility test was performed. A focused siRNA library of the above 158 genes was prepared (474 siRNA, 3 siRNAs / gene), and the degree of expression reduction of the COL1A1 gene when each gene was suppressed by siRNA was evaluated using the same protocol as the primary screening. The reproducibility test was performed at n = 3, and the criteria for hit siRNA was that the expression of the COL1A1 gene was reduced by 40% compared to the negative control. As a result, the reproducibility of four genes, COL8A2, GALNT2, FRYL, and TIMP1, was confirmed, and the gene group was used as a primary screening hit gene.
The results of the primary screening reproducibility test are listed in Table 2.

Figure JPOXMLDOC01-appb-T000002
 実施例3で得られた4遺伝子のうち、COL1A1遺伝子発現の抑制データから最も有望なターゲット遺伝子であると思われるGALNT2について、2次以降の評価を行うこととした。
 具体的には、2次評価では、他の線維化マーカーであるalpha-smooth muscle actin(αSMA, ACTA2)タンパクの発現に与える影響をウエスタンブロットにより調べた。なお、siRNAは一次スクリーニングと同じ配列を使用し(3 siRNA/遺伝子)、陰性コントロールにはNegative Control #1 siRNAを用いた。Opti-MEM I 56.75μlとRNAiMAX 1.75μlを混合し、室温で5分間保持した後、0.1μMのsiRNA 25μl(final 5 nM)と混合し、室温でさらに20分間保持した。本溶液を24ウェルプレートに移し、HK2細胞を50,000個含むKeratinocyte Serum Free Mediaを各ウェルに416.5μl添加し、37℃、5%CO2環境下で48時間培養した。次に、培地交換により2 ng/ml のrecombinant TGFβを含むKeratinocyte Serum Free Mediaを500μl添加し、さらに24時間培養した。培養上清を除去後、PBSで洗浄し、70μlのRIPA buffer で細胞を溶解した。本細胞溶解液を遠心分離後、上清のタンパク濃度を測定し、6μgの全タンパク質をSDSポリアクリルアミドゲル電気泳動に供した。泳動後、PVDF膜にトランスファーし、Blocking One(ナカライテスク社製)を用いてブロッキングした後、一次抗体(マウス抗αSMA抗体、Sigma社製)と6.5℃にて終夜で反応させた。TBS-Tで洗浄後、HRP標識抗マウスIgG抗体(二次抗体、GE Healthcare社製)と室温で1時間反応させた。TBS-Tで洗浄した後、ECL Advance Western Blotting Detection Kit(GE Healthcare社製)とLAS-3000(FUJIFILM社製)を用いて抗原抗体反応を検出し、シグナルを定量化した。結果を表3に示す。3配列中、2配列のsiRNAでTGFβ刺激によるαSMAタンパク量の増加が陰性コントロールに比して低下したことから、GALNT2遺伝子の発現抑制はタンパクレベルでも線維化マーカーの増加を抑制することが分かった。

Figure JPOXMLDOC01-appb-T000003
Of the 4 genes obtained in Example 3, GALNT2, which is considered to be the most promising target gene from the suppression data of COL1A1 gene expression, was evaluated from the second order onward.
Specifically, in the secondary evaluation, the influence of other fibrosis markers on the expression of alpha-smooth muscle actin (αSMA, ACTA2) protein was examined by Western blot. In addition, siRNA used the same sequence as the primary screening (3 siRNA / gene), and Negative Control # 1 siRNA was used as a negative control. 56.75 μl of Opti-MEM I and 1.75 μl of RNAiMAX were mixed and kept at room temperature for 5 minutes, then mixed with 25 μl of 0.1 μM siRNA (final 5 nM) and kept at room temperature for another 20 minutes. This solution was transferred to a 24-well plate, 416.5 μl of Keratinocyte Serum Free Media containing 50,000 HK2 cells was added to each well, and cultured for 48 hours at 37 ° C. in a 5% CO 2 environment. Next, 500 μl of Keratinocyte Serum Free Media containing 2 ng / ml recombinant TGFβ was added by medium exchange, and further cultured for 24 hours. After removing the culture supernatant, the cells were washed with PBS, and the cells were lysed with 70 μl of RIPA buffer. After centrifuging the cell lysate, the protein concentration of the supernatant was measured, and 6 μg of total protein was subjected to SDS polyacrylamide gel electrophoresis. After electrophoresis, it was transferred to a PVDF membrane, blocked using Blocking One (manufactured by Nacalai Tesque), and then reacted with a primary antibody (mouse anti-αSMA antibody, manufactured by Sigma) at 6.5 ° C. overnight. After washing with TBS-T, the mixture was reacted with an HRP-labeled anti-mouse IgG antibody (secondary antibody, manufactured by GE Healthcare) at room temperature for 1 hour. After washing with TBS-T, antigen-antibody reaction was detected using ECL Advance Western Blotting Detection Kit (GE Healthcare) and LAS-3000 (FUJIFILM), and the signal was quantified. The results are shown in Table 3. Among 3 sequences, 2 sequences siRNA showed an increase in αSMA protein level by TGFβ stimulation compared to negative control, indicating that suppression of GALNT2 gene expression also suppressed increase in fibrosis markers at the protein level. .

Figure JPOXMLDOC01-appb-T000003
 更に、三次評価として、ヒト・プライマリー尿細管上皮細胞(Renal proximal tubular epithelial cell : RPTEC、LONZA社製)を用い、実施例2と同様にsiRNAで各遺伝子を発現抑制した際にTGFβ刺激によるCOL1A1遺伝子の発現上昇が減弱するかを調べた。なお、siRNAは一次スクリーニングと同じ配列を用い(3 siRNA/遺伝子)、陰性コントロールにはNegative Control #2 siRNA(NT-2;Ambion社製)を使用した。また、RPTECは2種類のロット(Lot0000264325, 0000197809)を使用して評価した。Opti-MEM I 69.3μlとRNAiMAX 0.7μlを混合し、室温で5分間保持した後、0.1μMのsiRNA 30μl(final 5 nM)と混合し、室温でさらに20分間保持した。本溶液を24ウェルプレートに移し、RPTECを75,000個含む REGM Bullet Kit(LONZA社製)を各ウェルに500μl添加し、37℃、5%CO2環境下で培養した。24時間後、培地交換により添加因子を含まないREGM Bullet Kitを加えることでStarvation処理を行い、さらにその24時間後に培地交換により10 ng/ml のrecombinant TGFβを含み添加因子を含まないREGM Bullet Kitを500μl添加し、24時間培養した。培養上清を除去後、実施例2と同様にFastLane Cell SYBR Green Kitと7900HT Fast Real Time PCR Systemを用いて、COL1A1遺伝子とGAPDH遺伝子のCt値を測定し、GAPDH遺伝子を内部標準として各siRNAについてCOL1A1遺伝子の発現量を算出した。細胞Lot0000197809の結果を表4に示す。3配列のsiRNA全てにおいてTGFβ刺激によるCOL1A1遺伝子の発現上昇が陰性コントロールに比して抑制され、またこのフェノタイプはもう一方の細胞ロット(Lot0000264325)でも確認された。このことから、GALNT2遺伝子はヒト・プライマリー尿細管上皮細胞においても線維化マーカーの発現を抑制する有望なターゲットであることが明らかとなった。

Figure JPOXMLDOC01-appb-T000004
Furthermore, as a third evaluation, human primary tubule epithelial cells (Renal proximal tubular epithelial cells: RPTEC, manufactured by LONZA) were used, and when each gene was suppressed with siRNA in the same manner as in Example 2, COL1A1 gene stimulated by TGFβ It was investigated whether the increase in the expression of the gene was attenuated. In addition, siRNA used the same sequence as the primary screening (3 siRNA / gene), and Negative Control # 2 siRNA (NT-2; manufactured by Ambion) was used as a negative control. RPTEC was evaluated using two types of lots (Lot0000264325, 0000197809). 69.3 μl of Opti-MEM I and 0.7 μl of RNAiMAX were mixed and kept at room temperature for 5 minutes, then mixed with 30 μl of 0.1 μM siRNA (final 5 nM) and kept at room temperature for another 20 minutes. This solution was transferred to a 24-well plate, 500 μl of REGM Bullet Kit (manufactured by LONZA) containing 75,000 RPTECs was added to each well, and cultured in a 37 ° C., 5% CO 2 environment. After 24 hours, Starvation treatment is performed by adding a REGM Bullet Kit that does not contain additional factors by changing the medium.After 24 hours, a REGM Bullet Kit that contains 10 ng / ml recombinant TGFβ and does not contain additional factors is added by changing the medium. 500 μl was added and cultured for 24 hours. After removing the culture supernatant, Ct values of COL1A1 gene and GAPDH gene are measured using FastLane Cell SYBR Green Kit and 7900HT Fast Real Time PCR System in the same manner as in Example 2, and for each siRNA using GAPDH gene as an internal standard. The expression level of COL1A1 gene was calculated. The results for cells Lot0000197809 are shown in Table 4. In all three siRNAs, the increase in COL1A1 gene expression by TGFβ stimulation was suppressed as compared to the negative control, and this phenotype was also confirmed in the other cell lot (Lot0000264325). This revealed that the GALNT2 gene is a promising target that suppresses the expression of fibrosis markers in human primary tubular epithelial cells.

Figure JPOXMLDOC01-appb-T000004
 更に、GALNT2の発現抑制が遺伝子発現プロフィールに与える影響を評価した。COL1A1の発現抑制作用が認められたGALNT2に対する2種類のsiRNA(siRNA#2, #3)をHK2細胞に導入し、48時間後にrecombinant TGFβを添加した。さらにその24時間後に細胞から全RNAを回収し、マイクロアレイデータを取得した。陰性コントロールsiRNAに比べ、GALNT2に対する2種類のsiRNAで共通して発現低下する遺伝子群を抽出した結果、COL1A1以外にCOL4A2やP4HA2、SNAI1等が含まれていたことから、GALNT2は複数の線維化関連因子の発現を制御していることが明らかとなった。
Furthermore, the effect of GALNT2 expression suppression on gene expression profiles was evaluated. Two types of siRNA (siRNA # 2, # 3) against GALNT2 in which COL1A1 expression-suppressing action was observed were introduced into HK2 cells, and after 48 hours, recombinant TGFβ was added. Further, 24 hours later, total RNA was collected from the cells, and microarray data was obtained. Compared to the negative control siRNA, GALNT2 was associated with multiple fibrosis because COL4A2, P4HA2, SNAI1, etc. were included in addition to COL1A1 as a result of extracting genes that commonly decrease expression in two types of siRNAs against GALNT2. It became clear that the expression of the factor was controlled.
 本発明は、医薬品の分野、特に慢性腎臓病又は線維化疾患の治療薬の開発および製造の分野において利用可能である。 The present invention can be used in the field of pharmaceuticals, particularly in the field of development and production of therapeutic agents for chronic kidney disease or fibrotic diseases.

Claims (13)

  1. GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質を含む、医薬組成物。 A pharmaceutical composition comprising a substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or a substance that suppresses the activity of a protein encoded by these genes.
  2. GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質が核酸である、請求項1に記載の医薬組成物。 The pharmaceutical composition according to claim 1, wherein the substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or the substance that suppresses the activity of the protein encoded by these genes is a nucleic acid.
  3. 前記核酸が、siRNA若しくはアンチセンスオリゴヌクレオチド又はこれらの発現ベクターである請求項2に記載の医薬組成物。 The pharmaceutical composition according to claim 2, wherein the nucleic acid is siRNA or antisense oligonucleotide or an expression vector thereof.
  4. GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質を含む、COL1A1遺伝子の発現抑制剤。 A COL1A1 gene expression inhibitor comprising a substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene, or a substance that suppresses the activity of a protein encoded by these genes.
  5. GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質が核酸である、請求項4に記載のCOL1A1遺伝子の発現抑制剤。 The COL1A1 gene expression inhibitor according to claim 4, wherein the substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or the substance that suppresses the activity of the protein encoded by these genes is a nucleic acid.
  6. 前記核酸が、siRNA若しくはアンチセンスオリゴヌクレオチド又はこれらの発現ベクターである請求項5に記載のCOL1A1遺伝子の発現抑制剤。 The COL1A1 gene expression inhibitor according to claim 5, wherein the nucleic acid is siRNA, antisense oligonucleotide, or an expression vector thereof.
  7. GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質を含む、COL1A1遺伝子が関連する疾患の治療用医薬組成物。 A pharmaceutical composition for treating a disease associated with the COL1A1 gene, comprising a substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene, or a substance that suppresses the activity of a protein encoded by these genes.
  8. GALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現を抑制する物質又はこれらの遺伝子がコードするタンパク質の活性を抑制する物質が核酸である、請求項7に記載の医薬組成物。 The pharmaceutical composition according to claim 7, wherein the substance that suppresses the expression of GALNT2, COL8A2, FRYL, or TIMP1 gene or the substance that suppresses the activity of the protein encoded by these genes is a nucleic acid.
  9. 前記核酸が、siRNA若しくはアンチセンスオリゴヌクレオチド又はこれらの発現ベクターである請求項8に記載の医薬組成物。 The pharmaceutical composition according to claim 8, wherein the nucleic acid is siRNA or antisense oligonucleotide or an expression vector thereof.
  10. COL1A1遺伝子が関連する疾患が慢性腎臓病又は線維化疾患である請求項7~9のいずれかに記載の医薬組成物。 The pharmaceutical composition according to any one of claims 7 to 9, wherein the disease associated with the COL1A1 gene is chronic kidney disease or fibrosis disease.
  11. (1)細胞と被験物質とを接触させる工程、
    (2)被験物質を接触させた細胞におけるGALNT2、COL8A2、FRYL又はTIMP1遺伝子の発現量を測定し、該発現量を、被験物質を接触させない対照細胞における前記遺伝子の発現量と比較する工程、及び
    (3)被験物質を接触させた細胞における前記遺伝子の発現が、対照細胞における前記遺伝子の発現よりも低下している場合に、被験物質をCOL1A1遺伝子が関連する疾患の治療物質として選択する工程を含む、
    COL1A1遺伝子が関連する疾患の治療物質のスクリーニング方法。
    (1) a step of bringing cells into contact with a test substance;
    (2) measuring the expression level of the GALNT2, COL8A2, FRYL, or TIMP1 gene in a cell contacted with the test substance, and comparing the expression level with the expression level of the gene in a control cell not contacted with the test substance; (3) selecting the test substance as a therapeutic substance for a disease associated with the COL1A1 gene when the expression of the gene in the cell contacted with the test substance is lower than the expression of the gene in a control cell; Including,
    A screening method for a therapeutic agent for a disease associated with the COL1A1 gene.
  12. 配列番号1で表されるアミノ酸配列を有するポリペプチド、又は配列番号1で表されるアミノ酸配列の1又は数個のアミノ酸が欠失、置換、及び/若しくは付加されたアミノ酸配列を有し、N-アセチルガラクトサミン転移活性を有するポリペプチドを用いることを特徴とするCOL1A1遺伝子が関連する疾患の治療物質のスクリーニング方法。 A polypeptide having the amino acid sequence represented by SEQ ID NO: 1, or an amino acid sequence in which one or several amino acids of the amino acid sequence represented by SEQ ID NO: 1 are deleted, substituted, and / or added; A method for screening a therapeutic substance for a disease associated with the COL1A1 gene, which comprises using a polypeptide having acetylgalactosamine transfer activity.
  13. COL1A1遺伝子が関連する疾患が慢性腎臓病又は線維化疾患である請求項11又は12に記載の方法。 The method according to claim 11 or 12, wherein the disease associated with the COL1A1 gene is chronic kidney disease or fibrosis disease.
PCT/JP2013/071109 2012-08-06 2013-08-05 Pharmaceutical composition for treating new chronic kidney disease and method for screening medicine for new chronic kidney disease WO2014024820A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2012173531 2012-08-06
JP2012-173531 2012-08-06

Publications (1)

Publication Number Publication Date
WO2014024820A1 true WO2014024820A1 (en) 2014-02-13

Family

ID=50068046

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2013/071109 WO2014024820A1 (en) 2012-08-06 2013-08-05 Pharmaceutical composition for treating new chronic kidney disease and method for screening medicine for new chronic kidney disease

Country Status (1)

Country Link
WO (1) WO2014024820A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001085215A2 (en) * 2000-05-11 2001-11-15 Glycozym Aps METHODS OF MODULATING FUNCTIONS OF POLYPEPTIDE GalNAc TRANSFERASES AND OF SCREENING SUBSTANCES THEREFOR, PHARMACEUTICAL COMPOSITIONS COMPRISING SUCH AGENTS
JP2011508730A (en) * 2007-12-17 2011-03-17 ノバルティス アーゲー UDP-N-acetyl-alpha-D-galactosamine: modification of high density lipoprotein levels by modulation of the polypeptide N-acetylgalactosaminyltransferase (GALNT)
WO2011029996A1 (en) * 2009-09-08 2011-03-17 Biotie Therapies Corp. Use of vap-1 inhibitors for treating fibrotic conditions
WO2011046204A1 (en) * 2009-10-16 2011-04-21 持田製薬株式会社 Marker associated with non-alcoholic steatohepatitis
JP2012051868A (en) * 2010-08-05 2012-03-15 Nitto Denko Corp Composition for regenerating normal tissue from fibrotic tissue

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001085215A2 (en) * 2000-05-11 2001-11-15 Glycozym Aps METHODS OF MODULATING FUNCTIONS OF POLYPEPTIDE GalNAc TRANSFERASES AND OF SCREENING SUBSTANCES THEREFOR, PHARMACEUTICAL COMPOSITIONS COMPRISING SUCH AGENTS
JP2011508730A (en) * 2007-12-17 2011-03-17 ノバルティス アーゲー UDP-N-acetyl-alpha-D-galactosamine: modification of high density lipoprotein levels by modulation of the polypeptide N-acetylgalactosaminyltransferase (GALNT)
WO2011029996A1 (en) * 2009-09-08 2011-03-17 Biotie Therapies Corp. Use of vap-1 inhibitors for treating fibrotic conditions
WO2011046204A1 (en) * 2009-10-16 2011-04-21 持田製薬株式会社 Marker associated with non-alcoholic steatohepatitis
JP2012051868A (en) * 2010-08-05 2012-03-15 Nitto Denko Corp Composition for regenerating normal tissue from fibrotic tissue

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
ALEXAKIS, CATHERINE ET AL.: "Organ-specific collagen expression: implications for renal disease", NEPHRON EXP NEPHROL, vol. 102, no. 3-4, 2006, pages E71 - E75 *
AOYAMA ISAO ET AL.: "Molecular insights into preventive effects of AST-120 on the progression of renal failure", CLIN EXP NEPHROL, vol. 5, no. 4, 2001, pages 209 - 216 *
ELBASHIR SM ET AL.: "RNA interference is mediated by 21- and 22-nucleotide RNAs", GENES DEV, vol. 15, no. 2, 2001, pages 188 - 200 *
LI BING-XIN ET AL.: "Fluorofenidone attenuates renal interstitial fibrosis in the rat model of obstructive nephropathy", MOL CELL BIOCHEM, vol. 354, no. 1-2, 2011, pages 263 - 273 *

Similar Documents

Publication Publication Date Title
Wang et al. CircUbe3a from M2 macrophage-derived small extracellular vesicles mediates myocardial fibrosis after acute myocardial infarction
JP6414886B2 (en) Long non-coding RNA for anti-cancer therapy
JP2018512373A (en) Methods and compositions for treatment of malignant tumors associated with KRAS mutations
US10280422B2 (en) MiR-92 inhibitors and uses thereof
WO2017043370A1 (en) Exosome secretion inhibitor
US9322015B2 (en) Methods of using microRNA-26a to promote angiogenesis
US20160145615A1 (en) Agonists of ddah1 for treating endothelial dysfunction
KR102142791B1 (en) Use of miR-204 inhibitors for treating osteoarthritis
JP6478416B2 (en) Pharmaceutical composition for the treatment of chronic kidney disease
EP3237619B1 (en) Compositions and methods for inhibiting expression of adamts-5 and adam17
JP6436477B2 (en) Pharmaceutical composition for cancer treatment
WO2014024820A1 (en) Pharmaceutical composition for treating new chronic kidney disease and method for screening medicine for new chronic kidney disease
WO2012020839A1 (en) Pharmaceutical composition for cancer therapy
US20180112218A1 (en) Targeting Hepatitis B Virus (HBV) Host Factors
US20230407297A1 (en) Bioengineered wnt5a therapeutics for advanced cancers
US11952574B2 (en) Compositions and methods for inhibiting transmembrane serine protease 6 (TMPRSS6) expression
US20230287427A1 (en) Inhibition of lncExACT1 to Treat Heart Disease
WO2023234410A1 (en) Inhibitory agent for myocardial cell death, and prophylactic or therapeutic agent for myocardial disorders or heart failure
WO2024048528A1 (en) Prophylactic agent and/or therapeutic agent for heart diseases and laminopathy
TW202300645A (en) Compositions and methods for modulating pnpla3 expression
JP2023548209A (en) ILD, PF - Viral vectors and nucleic acids for ILD and IPF treatment
WO2023244870A1 (en) Compositions and methods for control of transient site-specific copy gains, genomic insertions, and rearrangements associated with mixed lineage leukemia
WO2013154128A1 (en) Novel arteriosclerosis treatment drug composition and method for screening arteriosclerosis treatment drugs
WO2022096563A1 (en) Viral vectors and nucleic acids for use in the treatment of ild, pf-ild and ipf
WO2021062251A2 (en) Targeting rlim to modulate body weight and obesity

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: 13827111

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: 13827111

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: JP