WO2008029871A1 - Nerve fiber degeneration inhibitor - Google Patents

Nerve fiber degeneration inhibitor Download PDF

Info

Publication number
WO2008029871A1
WO2008029871A1 PCT/JP2007/067375 JP2007067375W WO2008029871A1 WO 2008029871 A1 WO2008029871 A1 WO 2008029871A1 JP 2007067375 W JP2007067375 W JP 2007067375W WO 2008029871 A1 WO2008029871 A1 WO 2008029871A1
Authority
WO
WIPO (PCT)
Prior art keywords
chondroitin sulfate
substance
protein
gene
proteodarican
Prior art date
Application number
PCT/JP2007/067375
Other languages
French (fr)
Japanese (ja)
Inventor
Hiroyuki Yoneyama
Kenji Ishida
Jun Koyama
Original Assignee
Stelic Institute Of Regenerative Medicine, Stelic Institute & Co.
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
Priority claimed from PCT/JP2006/323678 external-priority patent/WO2008029493A1/en
Application filed by Stelic Institute Of Regenerative Medicine, Stelic Institute & Co. filed Critical Stelic Institute Of Regenerative Medicine, Stelic Institute & Co.
Publication of WO2008029871A1 publication Critical patent/WO2008029871A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • G01N33/5044Chemical 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 involving specific cell types
    • G01N33/5058Neurological cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/46Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
    • G01N2333/47Assays involving proteins of known structure or function as defined in the subgroups
    • G01N2333/4701Details
    • G01N2333/4722Proteoglycans, e.g. aggreccan
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/988Lyases (4.), e.g. aldolases, heparinase, enolases, fumarase

Definitions

  • the present invention relates to a therapeutic and prophylactic agent for neurofibrotic degenerative diseases based on control of accumulation of chondroitin sulfate proteoglycan (CSPG), a method for inhibiting neurofibrotic degeneration, and Alzheimer based on the method.
  • CSPG chondroitin sulfate proteoglycan
  • Background art on methods of treating or preventing neurofibrotic degenerative diseases including AD Alzheimer's disease, PD (Parkinson's disease), and amyotro phic Lateral Sclerosis (ALS)
  • Cranial nerve fiber degenerative disease in neurofibrotic degenerative diseases is recognized as an intractable disease resulting from a decrease due to neuronal cell death.
  • This neurofibrotic degenerative disease can be roughly divided into two groups representing symptoms related to memory and dementia, and symptoms related to movement.
  • the former is Alzheimer's disease and the latter is Parkinson's disease.
  • Alzheimer's disease is projected to increase to 34 million in 20 years in over 18 million patients worldwide (International Association for Alzheimer's Disease: ADI).
  • ADI International Association for Alzheimer's Disease
  • Alzheimer's disease is due to genetically-onset familial Alzheimer's disease and abnormal metabolic balance (decreased acetylcholine) caused by a defect in the factors involved in the production of amyloid 0 peptide (A / 3) and its degradation.
  • a / 3 amyloid precursor protein
  • APP amyloid precursor protein
  • Non-patent Document 1 genes that express apolipotanic protein (apo ⁇ ) are greatly involved as a risk factor for Alzheimer's disease!.
  • Non-patent document 2 genes that express apolipotanic protein (apo ⁇ ) are greatly involved as a risk factor for Alzheimer's disease!.
  • Alzheimer's disease is the only therapeutic drug for Alzheimer's disease that is still approved for sale in Japan. Symptoms have the effect of delaying the progression of dementia by about 9 months, but do not promote the recovery of cognitive function!
  • This therapeutic drug is intended to increase the concentration of acetylcholine that has been reduced in the brain by inhibiting acetylcholinesterase, an enzyme that degrades acetylcholine, a neurotransmitter that is related to memory and learning.
  • Donepezil hydrochloride does not serve as a curative for Alzheimer's disease and is only given to mild patients.
  • each research institution for Alzheimer's disease has created animal models to discover disease-causing factors, trial and error, and treatments that are still a decisive factor because many factors are involved. No law or treatment has been established.
  • Parkinson's disease which is a disease but is listed as an intractable disease. This is due to the death of dopamine neurons in the part of the brain known as the substantia nigra, which reduces the amount of dopamine (a neurotransmitter made by neurons in the substantia nigra). This is a disease caused by a loss of balance with acetylcholine, another neurotransmitter, and a relative increase in acetylcholine. The symptoms include tremor, muscle stiffness, slow motion and posture maintenance. There are four major symptoms called disability. In Japan, this disease tends to be more common in women with a male-female ratio of 1: 1.5-2, and a wide range of people in their 30s and 80s It has developed in the age group. The current number of patients is around 100 per 100,000 population
  • L-dopa precursor of donomin
  • donomin receptor stimulant increased L-dopa effect
  • anticholinergic agent acetylcholine inhibitor
  • Dopamine release promoters noradrenaline supplements (remedies for freezing), dopamine degradation inhibitors (MAO-B inhibitors, COMT inhibitors), etc. It is used according to the shape.
  • these therapies do not act as curatives and are used to relieve symptoms.
  • long-term administration such as 5 to 10 years can cause wearing off phenomenon (shortening of drug effect), on-off phenomenon (loss of drug effect), dyskinesia (involuntary movement), psychiatric symptoms such as hallucination and delusion. Appears.
  • proteodarican focused on the treatment of the above-mentioned neurofibrotic degenerative disease has a structure in which one or more glycosaminodarlican (GAG) chains are covalently bound to a protein called a core protein. It is thought that the specific sugar chain structure of the GAG chain is responsible for the various functions of proteolidicans.
  • GAG glycosaminodarlican
  • Proteolidicans are based on the type of GAG chains and chondroitin 3 ⁇ 43 ⁇ 4 acid proteoglycans (CSPG: chondroitin) sulfate proteoglycans), denorematanite) 3 ⁇ 4 acid proteoglycans (DSPGs: dermatan sulfate proteoglycans), heparan acid proteoglycans (HSPGs) Is done.
  • CSPG chondroitin 3 ⁇ 43 ⁇ 4 acid proteoglycans
  • DSPGs dermatan sulfate proteoglycans
  • HSPGs heparan acid proteoglycans
  • Non-patent Documents 20 to 22 As a function in the month, HSPGs and co-protein strength GAG chain, which is extended as a side chain, converts non-fibrotic amyloid ⁇ -protein into neurotoxic fibrotic amyloid / 3-protein and amyloid against the proteolytic mechanism
  • 13-protein
  • Non-patent Documents 20 to 22 heparan sulfate proteodarican and glycosaminodarlican are considered to be the most prominent etiology of Parkinson's disease and do not fibrize -synuclein! /, Or a protein that causes Alzheimer's disease Amyloid precursor protein (APP) is also denatured! / Hanare is considered to be a thing (Non-Patent Document 23).
  • APP Amyloid precursor protein
  • CSPG is an essential molecule during the embryonic period and is abundant in each organ.
  • Neural regeneration such as neural stem cell differentiation inducer (Patent Document 1), nerve regeneration using human / bone morphogenetic protein (Patent Document 2), human, nerve regeneration using bone morphogenetic protein (Patent Document 3) It is considered as a molecule that controls the processes involved! /, While treating the damage of the central nervous system (Patent Document 4), materials and methods for promoting the repair of nerve tissue (Patent Document) It is also involved in the inhibition of nerve regeneration at various sites such as 5) and blood vessel smooth muscle cell treatment inhibitory factor (Patent Document 6).
  • chondroitin sulfate proteodaricans which are regenerative inhibitors that are expressed when the central nervous system is damaged
  • chondroitinase ABC There is a report example that degrades CSPG by administration of an enzyme that selectively removes chondroitin sulfate, which is a type of GAG chain, and promotes regeneration of the central nerve
  • Patent Document 1 Patent Publication 2005-278641
  • Patent Document 2 Patent Publication 2005-007196
  • Patent Document 3 Patent Publication No. 09-501932
  • Patent Document 4 Patent Publication 2005-526740
  • Patent Document 5 Patent Publication 2005-500375
  • Patent Document 6 Patent Publication 08-510209
  • Non-patent literature l Iwata et al. J. Neurosci. (2004) 24 (4) 991-998
  • Non-Patent Document 2 Strittmatter, W.J et al. Proc. Natl. Acad. Sci. (1993) 90 8098-8102
  • Non-Patent Document 3 Forman et al. Nat. Med. (2004) 10 (10) 1055-1063
  • Non-Patent Document 4 Matthew, J. F. Nature, review (2006) 7 306-318
  • Non-Patent Document 5 Gsponer, J. et al. Protein Pept Lett. (2006) 13 (3) 287-293
  • Non-Patent Document 6 McNaught, .S.P. Et al. Neurobiol. Aging (2006) 27 530-545
  • Non-Patent Document 7 Hattori, N. et al. Lancet. (2004) 364 (9435) 722-724
  • Non-patent literature 8 Murakami, T. et al. Ann neurol. (2004) 55 (3) 439-442
  • Non-patent literature 9 Hisae Kadowaki et al. Experimental medicine (2006) 24, 10173-180
  • Non-patent literature 10 Muramatsu, S. et al. Rinsho Shinkeigaku. (2005) 45 (11) 902-904
  • Non-patent literature ll Iwata. N. et al. J. Neurosci. (2004) 24 (4) 991- 998
  • Non-patent document 12 Hadaczek P, et al. Hum Gene Ther. (2006) 17 (3) 291-302
  • Non-patent document 13 Lindahl, U et al. (1972) In Glycoproteins (Gottschalk, A. ed) pp. 49
  • Non-Patent Document 14 Oegema, T et al. J. Biol. Chem. (1984) 259 1720-1726
  • Non-patent document 15 Sugahara, et al. J. Biol. Chem. (1988) 263 10168-10174
  • Non-patent document 16 Sugahara, et al. J. Biol. Chem. (1992) 267 6027-6035
  • Non-Patent Document 17 De Waard et al. J. Biol. Chem. (1992) 267 6036-6043
  • Non-Patent Document 18 Moses, J, Oldberg et al. Eur. J. Biol. (1992) 248 521-526
  • Non-Patent Document 19 Yamada, S et al. Trends in Glycoscience and Glycotechnology, (1998
  • Non-patent literature 20 Castillo, GM et al. J. Neurochem. (1997) 69 2452-2465
  • Non-patent literature 21 Cotman, S. et al. Mol. Cell. Neurosci. 15 (2000) 183-198
  • Non-patent Reference 22 Snow, AD et al. Neurobiol. Aging (1989) 10 481-497
  • Non-Patent Document 23 Horsen, J.V. et al. J. Alzheimers Dis. (2004) 6 469-474
  • Non-Patent Document 24 Rhodes KE, Fawcett JW J. Anat. (2004) 204 33-48
  • Non-Patent Document 25 DeWitt, D.A. et al. Brain Res. (1994) 656 205-209
  • An object of the present invention is to provide a neurofibrous degeneration inhibitor, a therapeutic agent for a neurofibrotic degenerative disease containing the drug as an active ingredient, and a screening method for the neurofibrotic degeneration inhibitor. .
  • chondroitinase chondroitinase ABC
  • TH Tyro sine hydroxylase
  • neurofibrous degeneration can be suppressed by inhibiting the accumulation or biosynthesis of chondroitin sulfate proteodalycan, and completed the present invention.
  • a substance that inhibits the production or accumulation of chondroitin sulfate proteodarican is useful as a neurofibrotic degeneration inhibitor.
  • the drug is a drug for treatment or prevention of neurofibrotic degenerative diseases.
  • a gene silencing nucleic acid which is a substance having a “synthesis inhibitory action” of chondroitin sulfate proteodarican and a chondroitinase which is a substance having a “degradation promoting action” of chondroitin sulfate proteoglycan
  • a substance that has the “desulfating action” of chondroitin sulfate proteodarican “inhibits the production or accumulation of chondroitin sulfate proteodarican” in completely different pathways.
  • the present invention relates to a neurofibrous degeneration inhibitor, a therapeutic agent for a neurofibrotic degenerative disease containing the drug as an active ingredient, a screening method for a neurofibrotic degeneration inhibitor, and more specifically, ,
  • a neurofibrotic degeneration inhibitor comprising a substance that inhibits the production or accumulation of chondroitin sulfate proteodarican as an active ingredient
  • [6] Characteristically characterized in that the production or accumulation of chondroitin sulfate proteodarican is inhibited in the brain. Specifically, it is characterized by suppressing chondroitin sulfate proteodarican deposition in brain tissue), [1] A drug according to any one of to [5], [7] a drug for treatment or prevention of neurofibrotic degenerative disease, a drug according to [1] to [6], or any of the above,
  • the neurofibrotic degenerative disease is cerebrospinal nerve or peripheral nerve fibrotic degenerative disease
  • the neurofibrotic degenerative disease is Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, polyglutamine disease, spinal muscular atrophy, Huntington's disease, or multiple sclerosis. Drugs,
  • a dopamine neuron regeneration promoter in brain tissue comprising as an active ingredient a substance that inhibits the production or accumulation of chondroitin sulfate proteodarican;
  • An agent for inhibiting the destruction of brain tissue in inflammatory cells comprising as an active ingredient a substance that inhibits the production or accumulation of chondroitin sulfate proteodarican,
  • a screening method for a neurofibrotic degeneration inhibitor comprising selecting a substance having an action of inhibiting the production or accumulation of chondroitin sulfate proteodarican from a test sample,
  • a screening method for a neurofibrotic degeneration inhibitor comprising the following steps (a) to (c):
  • the present invention further relates to the following.
  • [17] A method for treating a neurofibrotic degenerative disease, comprising a step of administering the drug according to any one of [1] to [9] to an individual (patient or the like).
  • a method for suppressing neurofibrotic degeneration comprising a step of administering a substance that inhibits the production or accumulation of chondroitin sulfate proteodarican to an individual.
  • a method for activating glial astrocytes comprising a step of administering a substance that inhibits the production or accumulation of chondroitin sulfate proteodarican to an individual.
  • a method for preventing the destruction of brain tissue in inflammatory cells including the step of administering a substance that inhibits the production or accumulation of chondroitin sulfate proteodarican (to prevent invasion such as infection or drug-induced encephalitis or trauma) How to protect against).
  • composition comprising the drug according to any one of [1] to [; 11] and a pharmaceutically acceptable carrier.
  • chondroitin sulfate proteodalycan is related to the onset of neurofibrotic degeneration. Inhibition of chondroitin sulfate proteoglycan production and accumulation has been shown to suppress neurofibrotic degeneration. It will be possible to provide a therapeutic agent for neurofibrotic degenerative diseases with a new concept that has never existed before. In particular, neurofibrotic degeneration is closely related to Parkinson's disease, Alzheimer's disease, etc., where the number of patients is increasing in modern society, and a new concept of therapeutic drugs has important medical and industrial significance.
  • FIG. L Untreated group, chondroitinase ABC, GalNAcST siRNA treatment in Parkinson's disease model mice induced by l-methyl-4-pheny ⁇ 1, 2, 3, 6 tetrahydropyridine (MPTP) This is a photograph examining the expression of ⁇ -actin, GalNAc4ST-1 and GALNAC4S-6ST by RT-PCR on the 8th day (final day) in the group.
  • FIG. 2 Photograph showing the deposition of chondroitin sulfate teodarican (CSPG) in MPTP-induced Parkinson's disease model mice in the untreated group, Gal NAcST siRNA-treated group and chondroitinase ABC-treated group .
  • CSPG chondroitin sulfate teodarican
  • FIG. 3 is a photograph showing infiltration of F4 / 80-positive inflammatory macrophage in an untreated group, a GalNAcST siRNA-treated group and a chondroitinase ABC-treated group in Parkinson's disease model mice induced by MPTP.
  • FIG. 4 is a photograph showing fibroblasts in the brain in an untreated group, a Gal NAcST siRNA-treated group and a chondroitinase ABC-treated group in a Parkinson's disease model mouse induced by MPTP.
  • FIG. 5 is a photograph showing glial cell astrocytes in an untreated group, a GalNAcST siRNA-treated group and a chondroitinase ABC-treated group in Parkinson's disease model mice induced by MPTP.
  • Fig.6 Tyrosine hydroxylase (TH) secretion in MPTP-induced Parkinson's disease model mice in the untreated group, GalNAcS T siRNA-treated group and chondroitinase ABC-treated group! It is a photograph.
  • Fig. 8 shows the results of analysis of inflammation and fibrosis-related gene expression by Real-time PCR.
  • the graph shows the relative ratio of TNF_a and TGF- ⁇ in the house keeping gene ( ⁇ -actin).
  • FIG. 9 shows the results of Nurrl gene expression analysis by Real_time PCR method.
  • Nurrl gene expression in brain tissue was performed using SYBR premix kit (Takara Bio) and Real-time PCR thermal cycler DICE (Takara Bio).
  • the graph shows the dimensions of Nurrl and house keeping Gene (/ 3-actin).
  • FIG. 11 is a diagram showing a comparison of survival rates between a treated group and an untreated group.
  • the survival rate of each group was compared over time. The analysis was carried out using day 7 (day 7) as an endpoint, using the force planmeyer survival curve.
  • the treatment group was treated with C4_sulfatase and GalNAc4S_6ST siRNA.
  • Parkinson's disease As a pathological condition associated with Parkinson's disease, which is one of the typical neurofibrotic degenerative diseases, there is a degenerative condition such as fomentation due to infiltration of macrophages, lysen fibroblasts, etc. in brain neurons.
  • the present inventors have focused on the function of chondroitin sulfate proteodarican in order to improve the degenerative state of neurons in the brain as an effective method for treating Parkinson's disease.
  • a state in which the accumulation of chondroitin sulfate proteodarican was suppressed in Parkinson's disease model mice was analyzed in detail, and it was found that the accumulation of chondroitin sulfate proteodarican was improved compared to neurons in the wild-type brain.
  • the present invention relates to a neurofibrotic degeneration inhibitor comprising, as an active ingredient, a substance that inhibits the production or accumulation of chondroitin sulfate proteodalycan.
  • the "chondroitin sulfate proteodarican” of the present invention is one of the proteodaricans, and is a covalently bonded compound of chondroitin sulfate / dermatan sulfate, which is a typical sulfated mucopolysaccharide, and a protein (coprotein). It is a general term.
  • the “chondroitin sulfate proteodarican” in the present invention is preferably a human chondroitin sulfate proteodarican, but the species from which it is derived is not particularly limited. Proteins equivalent to kan (homologs, orthologs, etc.) are also used in the present invention.
  • Konkondoroloyichitin sulphate sulfate proteoteo daridarikankan has a tantalum protein that is equivalent to the human proteotheododalyricacan.
  • the coconut doroleutintin sulfate sulfate sulfate proteoteo odalyrikankan is temporarily affected by inflammatory inflammation and the like.
  • the chain of guriglicosasamininodararirikakan (GGAAGG)) binds to and becomes a proprotetheodaridarikankan, it is so loose.
  • the activity of chondroitin sulfate proteodarican includes, for example, cell adhesion ability or cell growth promotion force S.
  • chondroitin sulfate proteodarican as follows.
  • the proliferation of tumor cells is measured in the presence of a protein having preferably 80% or more, more preferably 90% or more, most preferably 95% or more.
  • Proteins that have the effect of promoting mitotic proliferation can be identified as proteins with chondroitin sulfate proteodarican activity (Int J Exp Pathol. 2005 Aug; 86 (4): 219_29 and Histochem Cell Biol. 2005 Aug; 124 (2 ): 139-49)
  • high homology means 50% or more, preferably 70% or more, more preferably 80% or more, more preferably 90% or more (for example, 95% or more, further 96% , 97%, 98%, or 99% or higher), which is the mBLAST algorithm (Altschul et al. (1990) Proc. Natl. Acad. Sci. USA 8 7: 2264-8; arlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90: 5873-7).
  • Neuron fibrotic degeneration in the present invention refers to an abnormal state in nerve tissue.
  • the state of causing fountain fibrosis, the state of inflammation, the state of infiltrating fountain fibroblasts' inflammatory cells, the loss of specific cell types in nerve tissue, and cell death The power S that can be mentioned is not limited to these.
  • “inhibiting production or accumulation” of chondroitin sulfate proteodarican means, for example, “degradation promotion”, “synthesis inhibition”, “desulfation”, “sulfation” of chondroitin sulfate proteodarican.
  • Powers that include “inhibition” and the like are not limited to these, and it means that the abundance, function, or activity of chondroitin sulfate proteodarican is reduced or eliminated as compared with the comparison target.
  • the “substance that inhibits the production or accumulation” of chondroitin sulfate proteodarican is not particularly limited, but preferably the “substance that has an activity of promoting degradation of chondroitin sulfate proteoglycan” and “the substance has an inhibitory effect on synthesis”. “Substance”, “Substance with desulfurization and oxidation”, or “Substance with sulfation-inhibiting action”.
  • “Degradation promotion” of chondroitin sulfate proteodarican includes, for example, inhibition of expression / reduction of the protein that is the core of chondroitin sulfate proteodarican.
  • the “protein that is the core of chondroitin sulfate proteoglycan” is, for example, matri X type chondroitene sulfate prosuclicans, and examples thereof include core proteins such as aggrican, versican, neurocan, and b revican.
  • core proteins such as / 3 glycan, Decorin, Biglycan, Fibromodulin, and PG_Lb can be mentioned. These are only examples, and are not limited to these, and may be any protein that is widely used as the core of chondroitin sulfate proteodalycan.
  • “Expression” means “transcription” from a gene! /, And includes “translation” into a polypeptide and “degradation inhibition” of a protein. “Expression of the protein that is the core of chondroitin sulfate proteodarican” refers to the transcription and translation of the gene that encodes the protein that is the core of chondroitin sulfate proteodarican, or the chondroitin sulfate proteo This means that the protein that forms the core of Darican is produced.
  • the function of the protein serving as the core of chondroitin sulfate proteodarican includes, for example, the function of the protein binding to chondroitin sulfate and the binding to other components in the cell.
  • the various functions described above can be appropriately evaluated (measured) by those skilled in the art using common techniques. Specifically, the methods described in the examples described later, or the methods can be appropriately modified and carried out.
  • “degradation promotion” of chondroitin sulfate proteodarican is the expression of an enzyme that cleaves or degrades chondroitin sulfate proteodarican or an enzyme related thereto. May be a rise.
  • these enzymes include, but are not limited to, meta-mouth proteinases (eg, AD AMTS-1, ADAMTS-4, ADAMTS-5, etc.) chondroitinase, Calpain I, and the like.
  • “degradation promotion” may be a decrease in the abundance of chondroitin sulfate proteodarican caused by administration of these enzymes or a part of the enzymes.
  • Degradation promotion may be caused by administration of a substance that promotes suppression of chondroitin sulfate proteodarican expression.
  • substances include, but are not limited to, n-butylate, Diethyl carbamazepine, i'umcamycin, non-steroidal estrogen ⁇ and yclofeml deipheno.
  • Preferable embodiments of the "substance having a decomposition promoting action” include, for example, a compound (nucleic acid) selected from the group consisting of the following (a) to (c).
  • nucleic acid having a ribozyme activity that specifically cleaves the transcription product of the gene encoding the core protein of chondroitin sulfate proteodarican
  • examples of the “substance having a decomposition promoting action” include compounds selected from the following groups (a) to (c):
  • Synthetic inhibition of chondroitin sulfate proteodarican includes, for example, inhibition of glycosaminodarlican biosynthesis, inhibition of enzymes involved in chondroitin sulfate proteodarican synthesis, but are not necessarily limited to these. This refers to inhibiting the process of chondroitin sulfate proteodarican synthesis.
  • glycosaminoglyca For example, / 3 -D-xyloside, 2-deoxy-D-glucose (2-DG, ethane-hydroxy-l, l-diphosphonate (b TDP), 5 -hexyl-2-deoxyundine HUdR).
  • 2-DG 2-deoxy-D-glucose
  • b TDP ethane-hydroxy-l
  • b TDP l-diphosphonate
  • 5 -hexyl-2-deoxyundine HUdR 5 -hexyl-2-deoxyundine
  • examples of enzymes involved in chondroitin synthesis include GalNAc4ST-1, GalNAc4 ST-2, GALNAC4S_6ST, UA20ST, GalT_I, GalT_H, GlcAT_I, XylosylT, and the like. By inhibiting these and other enzymes and suppressing their expression, the synthesis of chondroitin sulfate proteodalycan is inhibited.
  • Preferable embodiments of the "substance having a synthesis inhibitory action” include, for example, compounds (nucleic acids) selected from the group consisting of the following (a) to (c).
  • examples of the "substance having a synthesis inhibitory action” include compounds selected from the following groups (a) to (c):
  • Desulfation of chondroitin sulfate proteodarican refers to removal of sulfate groups in chondroitin sulfate proteodarican, for example, desulfation by endogenous or externally administered desulfating enzymes, or sulfation. This includes, but is not limited to, the process by which sulfate groups are removed.
  • Examples of the desulfating enzyme include Chondroitin-4-sulfatase, Chondroitin-6-sulfatase force S.
  • Examples of compounds that inhibit sulfation include Chlorate and EGF rece. ptor antagonist and the like.
  • Preferable embodiments of the "substance having desulfating action” include, for example, a compound (nucleic acid) selected from the group consisting of the following (a) to (c).
  • nucleic acid having a ribozyme activity that specifically cleaves a transcript of a gene encoding a chondroitin sulfate proteodarican desulfase inhibitor protein
  • Examples of the "substance having desulfating action” include compounds selected from the following groups (a) to (c):
  • the “desulfation-inhibiting compound” is not limited to a protein, and includes non-protein compounds such as coenzymes, for example.
  • the “sulfation inhibitory action” of chondroitin sulfate proteodarican is, for example, the force S including inhibition of sulfate group transfer enzyme, but is not limited to this, and occurs in the process of chondroitin sulfate proteodarican synthesis. It refers to inhibition of sulfation.
  • Examples of the sulfotransferase include C4ST-1 (Chondroitin D-N-acetylgalactosamine).
  • a compound (nucleic acid) selected from the group consisting of the following (a) to (c) can be mentioned.
  • a compound (nucleic acid) selected from the group consisting of the following (a) to (c) can be mentioned.
  • examples of the “substance having a sulfation inhibiting action” include compounds selected from the group consisting of the following (a) to (c).
  • the enzymes exemplified above include not only one enzyme corresponding to one gene but also an enzyme group sharing certain characteristics.
  • chondroitinase is a collective term for enzymes such as ABC, AC, and B that share the characteristics of mucopolysaccharide-degrading enzymes but differ in substrate specificity.
  • chondroitinase AC I cleaves the chondroitin sulfates (A, C or E), chondroitin, chondroitin sulfate-dermatan sulfate hybrid type and hyaluronic acid N-acetylhexoxide binding bond.
  • an oligosaccharide having a ⁇ 4-glucuronic acid residue at the non-reducing end is generated.
  • This enzyme does not act on dermatan sulfate (chondroitin sulfate B, which has L-iduronic acid as hexuronic acid), ketalan sulfate, heparan sulfate and heparin.
  • chondroitinase AC II cleaves the N-acetyl hexosaminide bond of chondroitin, chondroitin sulfate A and chondroitin sulfate C in a desorbing reaction to produce ⁇ 4-unsaturated disaccharide ( ⁇ 0 ⁇ 05, A Di-4S and A Di-6S). This enzyme also works well on hyaluronic acid.
  • chondroitin sulfate B dermatan sulfate
  • Chondroitinase B dermatanase
  • Generate oligosaccharides do not contain L-iduronic acid! /, Does not act on chondroitin sulfate A and chondroitin sulfate C! / ,.
  • Dermatan a derivative of dermatan sulfate with the sulfate group removed Is not a substrate for this enzyme.
  • the site where the second position of the L-iduronic acid unit of dermatan sulfate is sulfated is more cleaved by this enzyme.
  • Chondroitinase ABC cleaves the N-acetyl hexosaminide bond of chondroitin sulfate A, chondroitin sulfate C, dermatan sulfate, chondroitin, and hyaluronic acid in a reactive manner, and generates ⁇ 4-hexuronic acid at the non-reducing end. Mainly produces disaccharides with residues.
  • Chondroitinase is a generic name for enzymes that have different properties but have a common property called mucopolysaccharide-degrading enzyme, and always 3 ⁇ 4 Chondroitinase ACI, Chonaroitinase AC II, Cnondrotinase Not limited to B, Chondroitinase ABC.
  • chondroitin-4-sulfatase chondroitin_6_sulfatase is a sequence referenced by multiple accession numbers in the genome database (for example, Gen bank accession number NT_039500 (part of which is accession number CAAA01098429 (sequence No .: 83)), and NT_078575, NT_039353, NW_001030904, NW_0 01030811, NW_001030796, NW_000349) are searched on the public gene database Genbank.
  • aggrican accesion number NM—007424, nucleotide sequence number: 1, amino acid sequence number: 2
  • versican accesion number BC096495, SEQ ID NO: 3 for nucleotide sequence, SEQ ID NO: 4 for amino acid sequence
  • neurocan accession number NM_010875, nucleotide sequence SEQ ID NO: 5, amino acid sequence SEQ ID NO: 6)
  • Biglycan (Accession number BC057185, SEQ ID NO: 13 for nucleotide sequence, SEQ ID NO: 14 for amino acid sequence)
  • Fibromodulin (Accession number NM_021355, nucleotide sequence number: 15, amino acid sequence number: 16)
  • PG-Lb (Accession number NM_007884, nucleotide sequence number: 17, amino acid sequence number: 18)
  • ADAMTS-1 (Accession number NM_009621, nucleotide sequence SEQ ID NO: 19, amino acid sequence SEQ ID NO: 20)
  • ADAMTS_4 (Accession number NM_172845, SEQ ID NO: 21 of nucleotide sequence, SEQ ID NO: 22 of amino acid sequence)
  • ADAMTS_5 (Accession number AF140673, SEQ ID NO: 23, Amino Acid IJ SEQ ID: 24)
  • Calpain I (Accession number NM_007600, nucleotide sequence SEQ ID NO: 25, amino acid sequence SEQ ID NO: 26)
  • GalNAc4ST-l accession number NM_175140, nucleotide sequence number: 27, amino acid sequence number: 28
  • GalNAc4ST-2 (Accession number NM_199055, nucleotide sequence SEQ ID NO: 29, amino acid sequence SEQ ID NO: 30)
  • GALNAC4S-6ST (Accession number NM_029935, nucleotide sequence SEQ ID NO: 31, amino acid sequence SEQ ID NO: 32)
  • UA20ST (Accession number NM—177387, SEQ ID NO: 33 for nucleotide sequence, SEQ ID NO: 34 for amino acid sequence)
  • GalT-I (Accession number NM—016769, nucleotide sequence number: 35, amino acid sequence number: 36)
  • GalT-II (Accession number BC064767, nucleotide sequence number: 37, amino acid sequence number: 38)
  • GlcAT-I accession number BC058082, nucleotide sequence number: 39, amino acid sequence number: 40, or accession number NM_024256, nucleotide sequence number: 41, amino acid sequence number: 42)
  • XylosylT (Accession number NM—145828, nucleotide sequence number: 43, amino acid sequence number: 44)
  • C4ST-1 (Accession number NM_021439, nucleotide sequence SEQ ID NO: 45, amino acid sequence SEQ ID NO: 46)
  • C4ST-2 (Accession number NM_021528, nucleotide sequence number: 47, amino acid sequence number: 48)
  • C4ST-3 (Accession No. XM—355798, nucleotide sequence SEQ ID NO: 49, amino acid sequence SEQ ID NO: 50)
  • D4ST (Accession number NM— 028117, nucleotide sequence number: 51, amino acid sequence number: 52)
  • C6ST-1 (Accession number NM_016803, nucleotide sequence SEQ ID NO: 53, amino acid sequence SEQ ID NO: 54)
  • C6ST-2 (Accession number AB046929, nucleotide sequence number: 55, amino acid sequence number: 56)
  • proteins other than those described above have high homology (usually 70% or more, preferably 80% or more, more preferably 90% or more, most preferably 95% or more) with the sequences described in the sequence listing. And the function of the protein (for example, it binds to the intracellular components)
  • the protein having a function or the like) is included in the protein of the present invention.
  • the above proteins are, for example, the following: IJ numbers: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36 , 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, the amino acid sequence in which one or more amino acids are added, calo, deletion, substitution, or insertion
  • a protein consisting of a sequence, wherein the number of normally changing amino acids is within 30 amino acids, preferably within 10 amino acids, more preferably within 5 amino acids, most preferably within 3 amino acids.
  • Examples of the gene in the present invention include, for example, SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 51, 53, or 55, an endogenous gene in another organism corresponding to the DNA comprising the nucleotide sequence described in any one of the above (a homologue of the above human gene) Etc.).
  • IJ numbers 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31,
  • DNA is generally sequence numbers: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, respectively. 39, 41, 43, 45, 47, 49, 51, 53, and 55.
  • High homology means 50% or more, preferably 70% or more, more preferably 80% or more, more preferably 90% or more (for example, 95% or more, further 96%, 97%, 98% or 99% or more). Means the homology of (above). This homology is determined by the mBLAST algorithm (Altschul et al. (1990) Proc. Natl. Acad. Sci. USA 87: 2264-8; Karlin and Altschul (1993) Proc.
  • Power can be determined by S. Further, when the DNA is isolated from the living body, the IJ numbers: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 51, 53, 55. It is considered that the DNA hybridizes under stringent conditions.
  • stringent conditions for example, “2 X SSC, 0.1% SDS, 50.C”, “2 X SSC, 0.1% SDS, 42 ° C”, “1 X SSC, 0.1% SDS, 37 ° C ”and more stringent conditions“ 2 X SSC, 0.1% SDS, 65.C ”,“ 0.5 X SSC, 0.1% SDS, 42.C ”and“ 0.2 X SSC, 0.1% SDS, 65 ° The ability S to raise the condition of “C”.
  • a person skilled in the art will convert a protein functionally equivalent to the above protein from the above highly homologous proteins into a chondroitin sulfate proteodarican degradation promoting action, synthetic inhibitory action, desulfating action or sulfating action. It can be obtained appropriately by using an inhibitory activity measurement method. A specific activity measuring method will be described in the section of the screening method in the present invention. Moreover, those skilled in the art can appropriately obtain an endogenous gene corresponding to the above gene in another organism based on the base sequence of the above gene.
  • the above-mentioned proteins and genes corresponding to the above-mentioned proteins and genes in organisms other than humans, or the above-mentioned proteins and genes functionally equivalent to the above-mentioned proteins and genes are also simply referred to as It may be described in.
  • the protein of the present invention can be prepared not only as a natural protein but also as a recombinant protein using a gene recombination technique.
  • a natural protein for example, it can be prepared by a method using affinity chromatography using an antibody against the above protein against an extract of a cell (tissue) considered to express the above protein. It is.
  • a recombinant protein can be prepared, for example, by culturing cells transformed with DNA encoding the protein.
  • the above-mentioned protein of the present invention is suitably used, for example, in the screening method described later.
  • Nucleic acid in the present invention means RNA or DNA. Chemically synthesized nucleic acid analogs such as so-called PNA (p-mark tide nucleic acid) are also included in the nucleic acids of the present invention. PNA replaces the pentose / phosphate skeleton, which is the basic skeleton structure of nucleic acid, with a polyamide skeleton with glycine as a unit, and has a three-dimensional structure very similar to nucleic acid.
  • PNA p-mark tide nucleic acid
  • antisense nucleic acids inhibit the expression of target genes by inhibiting various processes such as transcription, splicing or translation (Hirashima and Inoue, Shinsei Kagaku Kogaku Kenkyu 2 Nucleic acid IV gene replication and expression, Japan Biochemical Society, Tokyo Chemical Doujin, 1993, 319-347.).
  • the antisense nucleic acid used in the present invention can be any of the above-mentioned chondroitin sulfate proteodarican core protein, synthase, desulfase-suppressing protein, and sulfotransferase by any of the above-described actions. Expression and / or function of the gene encoding the force may be inhibited.
  • the above-mentioned chondroitin sulfate proteodarican core protein, synthase, desulfase inhibitor protein, or a gene encoding a sulfate transfer enzyme is complementary to the untranslated region near the 5 'end of the mRNA. If an antisense sequence is designed, it would be effective to inhibit gene translation.
  • an IJ complementary to the coding region or the 3 ′ untranslated region can also be used.
  • Nucleic acids containing [I] are also included in the antisense nucleic acids used in the present invention.
  • the antisense nucleic acid used is linked downstream of an appropriate promoter, and preferably a sequence containing a transcription termination signal is linked on the 3 ′ side.
  • the nucleic acid thus prepared can be transformed into a desired animal (cell) using a known method.
  • the sequence of the antisense nucleic acid is the gene or gene encoding the endogenous chondroitin sulfate proteodarican core protein, synthase, desulfase inhibitor protein, or sulfate group of the animal (cell) to be transformed. It is preferably a sequence complementary to a part of it, but it effectively suppresses gene expression. As far as possible, they need not be completely complementary.
  • the transcribed RNA preferably has a complementarity of 90% or more, most preferably 95% or more, to the target gene transcript.
  • the length of the antisense nucleic acid is preferably at least 15 bases and less than 25 bases, but the antisense nucleic acid of the present invention is not necessarily of this length. For example, it may be 100 bases or more, or 500 bases or more.
  • the antisense nucleic acid of the present invention is not particularly limited.
  • the base sequence of the Versican gene (GenBank accession number BC096495, SEQ ID NO: 3), C4ST-1 (GenBank accession number NM_021439, SEQ ID NO: : 45), C4ST-2 (GenBank accession number NM_021528, SEQ ID NO: 47), C4ST-3 (GenBank accession number XM_35579 8, SEQ ID NO: 49), etc. .
  • Ribozyme refers to an RNA molecule that has catalytic activity. The ability of ribozymes to have various activities exists, and in particular, research focusing on ribozymes as enzymes that cleave RNA has made it possible to design ribozymes that cleave RNA in a site-specific manner.
  • Some ribozymes have a size of 400 nucleotides or more, such as group I intron type and Ml RNA contained in RNase P, but some have an active domain of about 40 nucleotides called hammerhead type or hairpin type. (Makoto Koizumi and Eiko Otsuka, Protein Nucleic Acid Enzyme, 1990, 35, 2191 ⁇ ).
  • the self-cleaving domain of the hammerhead ribozyme cleaves 3 ′ of C15 in the sequence G13U14C15, but base pairing between U14 and A9 is important for its activity. Shows that A15 or U15 can also be cleaved (Koizumi, M. et al., FEBS Lett, 1988, 228, 228.).
  • a ribozyme whose substrate binding site is complementary to the RNA sequence in the vicinity of the target site, it is possible to create a restriction RNA-cleaving ribozyme that recognizes the sequence UC, UU or UA in the target RNA (Koizumi , M.
  • Hairpin ribozymes are also useful for the purposes of the present invention. This ribozyme is found, for example, in the minus strand of tobacco ring spot virus satellite RNA (Buzayan, JM., Nature, 1986, 323, 349). It has been shown that hairpin-type ribozymes can also produce target-specific RNA cleavage ribozymes (Kikuchi, Y. & Sasaki, N., Nucl Acids Res, 1991, 19, 6751., Hiroshi Kikuchi, Biology, 1992, 30, 112.).
  • the ribozyme is used to specifically cleave the above-mentioned chondroitin sulfate proteodarican core protein, synthase, desulfase inhibitor protein, or transcript of a gene encoding a sulfotransferase. Gene expression can be inhibited.
  • RNA interference (hereinafter abbreviated as "RNAi") using double-stranded RNA having the same or similar sequence as the target gene sequence. can be fi.
  • RNAi small interfering RNA
  • dsRNA double-stranded RNA
  • RNAi can be induced by using short dsRNA (siRNA). RNAi is more stable, easier to experiment, and less expensive than knockout mice. Has many advantages.
  • RNAi is a short double-stranded RNA (hereinafter abbreviated as “dsRNA”) consisting of a sense RNA consisting of a sequence homologous to the mRNA of the target gene and an antisense RNA consisting of a complementary sequence.
  • dsRNA short double-stranded RNA
  • This is a phenomenon that induces destruction by specifically and selectively binding to the target gene mRNA, and efficiently inhibiting (suppressing) the expression of the target gene by cleaving the target gene. For example, when dsRNA is introduced into a cell, the expression of the gene homologous to the RNA is suppressed (knocked down).
  • RNAi is the target Since gene expression can be suppressed, it has been attracting attention as a simple gene knockout method to replace conventional complicated and low efficiency homologous recombination methods, or as a method applicable to gene therapy. .
  • the RNA used for RNAi must be completely identical to the above-mentioned chondroitin sulfate proteodarican core protein, synthetic enzyme, desulfase inhibitor protein, or gene encoding a sulfotransferase, or a partial region of the gene. None, but preferably has complete homology.
  • the target is not particularly limited as long as it is a gene encoding the above-mentioned chondroitin sulfate proteoglycan core protein, synthetic enzyme, desulfase inhibitor protein, or sulfate transferase. It is possible to make any arbitrary region as a target candidate.
  • the base sequence of Versican gene SEQ ID NO: 3
  • the base sequence of C4ST-1 gene SEQ ID NO: 45
  • the base sequence of C4ST-2 gene SEQ ID NO: 47
  • the base sequence of C4ST-3 gene It can be created based on SEQ ID NO: 49. More specifically, a partial region of the sequence can be a target candidate.
  • a partial region of the base sequence of the Versican gene (SEQ ID NO: 57), the base of the C4ST-1 gene Partial region of sequence (SEQ ID NO: 58), partial region of base sequence of C4ST-2 gene (SEQ ID NO: 59), partial region of base sequence of C4ST-3 gene (SEQ ID NO: 60), C6ST -Partial region of the base sequence of 1 gene (SEQ ID NO: 61), partial region of the base sequence of C6ST-2 gene (SEQ ID NO: 62), partial region of the base sequence of GalNAc4ST-l gene (SEQ ID NO: 6 3), a partial region of the nucleotide sequence of the GalNAc4ST-2 gene (SEQ ID NO: 64), a partial region of the base sequence of GALNAC4S-6ST (SEQ ID NO: 65), and the like. More specifically, siRNA targeting the DNA sequence specifically shown by the present specification (SEQ ID NO: 7;! To 82) can be exemplified.
  • siRNA synthesized in vitro is linked to plasmid DNA and introduced into the cell
  • a method of annealing two RNAs, or the like can be employed.
  • the two RNA molecules may be molecules having a structure in which one end is closed, for example, siRNA (shRNA) having a hairpin structure.
  • shRNA is called short hairpin RNA, and part of a single strand forms a complementary strand with other regions.
  • This is an RNA molecule having a stem-loop structure. That is, a molecule capable of forming a double-stranded RNA structure in the molecule is also included in the siRNA of the present invention.
  • RNA capable of suppressing the expression of Versican, C4ST_1, C4ST_2, C4ST-3, etc. by the RNAi effect, which is specifically shown in the present specification
  • siRNA targeting DNA ligated IJ SEQ ID NO: 7;!
  • siRNA of the present invention may be used as long as it has a function of suppressing the expression of a gene encoding a chondroitin sulfate proteodarican core protein, a synthetic enzyme, a desulfase inhibitor protein, or a sulfotransferase. .
  • RNA used for RNAi need not be completely identical (homologous) to the gene encoding the protein or a partial region of the gene, but is completely identical (similar) ).
  • the double-stranded RNA having the RNAi effect in the present invention includes double-stranded RNA before being digested by DICER as described above. That is, even if a long RNA that does not have the RNAi effect as it is is expected to be degraded into siRNA having the RNAi effect in the cell, the double-stranded RNA in the present invention
  • the length of is not particularly limited.
  • the strand RNA can be decomposed in advance with DICER, and the degradation product can be used as the agent of the present invention.
  • This degradation product is expected to contain double-stranded RNA molecules (siRNA) having the RNAi effect. According to this method, it is not necessary to particularly select a region on mRNA expected to have an RNAi effect. That is, the region on the mRNA of the above-mentioned gene of the present invention having an RNAi effect does not necessarily need to be accurately defined.
  • double-stranded RNA that can be suppressed by the RNAi effect is known to those skilled in the art.
  • the double-stranded RNA of the present invention can be prepared based on the nucleotide sequence set forth in SEQ ID NO: 71.
  • RNA RNA sequence having a stronger // RNAi effect from the mRNA sequence which is a transcription product of the sequence, by a known method. If one strand is known, those skilled in the art can easily know the base sequence of the other strand (complementary strand).
  • a siRNA can be appropriately prepared by those skilled in the art using a commercially available nucleic acid synthesizer. In addition, for synthesis of desired RNA, a general synthetic contract service can be used.
  • the siRNA in the present invention may be a mixture of a plurality of sets of double-stranded RNAs for a region containing a target sequence, which need not necessarily be a set of double-stranded RNAs for the target sequence.
  • siRNA as a nucleic acid mixture corresponding to the target sequence can be appropriately prepared by a person skilled in the art using a commercially available nucleic acid synthesizer and a DICER enzyme. You can use the composite contract service.
  • the siRNA of the present invention includes so-called “cocktail siRNA”.
  • RNA ribonucleotides
  • one or more ribonucleotides constituting siRNA may be a corresponding deoxyribonucleotide.
  • This “corresponding” refers to the same base species (adenine, guanine, cytosine, thymine (uracil)) although the structures of the sugar moieties are different.
  • uracil uracil
  • the “plurality” is not particularly limited, but preferably refers to a small number of about 2 to 5
  • a DNA (vector) capable of expressing the above RNA of the present invention is also the above-mentioned tag of the present invention. It is contained in the preferable aspect of the compound which can suppress the expression of the gene which codes a protein.
  • the DNA (vector) capable of expressing the double-stranded RNA of the present invention is a DNA encoding one strand of the double-stranded RNA and a DNA encoding the other strand of the double-stranded RNA, Each DNA has a structure linked to a promoter so that it can be expressed.
  • the expression vector of the present invention can be prepared by appropriately inserting DNA encoding the RNA of the present invention into various known expression vectors.
  • the expression inhibitory substance of the present invention includes the above-mentioned chondroitin sulfate proteodarican core protein, synthase, desulfase inhibitor protein, or expression regulatory region of a gene encoding a sulfotransferase (for example, Specific examples include the base sequence represented by SEQ ID NO: 66, which is the promoter region of PG-Lb.)
  • SEQ ID NO: 66 which is the promoter region of PG-Lb.
  • the compound is, for example, a promoter DNA fragment of a gene encoding the above chondroitin sulfate proteodarican core protein, synthase, desulfase inhibitor protein, or sulfotransferase, and binding activity to the DNA fragment It can be obtained by a screening method using as an index.
  • those skilled in the art will determine whether or not the desired compound inhibits the expression of the above-mentioned chondroitin sulfate-teododalican core protein, synthase, desulfase-inhibiting protein, or gene encoding sulfotransferase. The determination can be appropriately carried out by a known method such as a reporter assay method.
  • the DNA (vector) capable of expressing the RNA of the present invention is also the core protein, synthase, desulfase-inhibiting protein, or sulfate group of the above-described chondroitin sulfate proteodarican of the present invention.
  • a preferred embodiment of the compound capable of inhibiting the expression of a gene encoding a transferase is included.
  • the DNA (beta) capable of expressing the double-stranded RNA of the present invention is a DNA that encodes one strand of the double-stranded RNA and a DNA force S that encodes the other strand of the double-stranded RNA.
  • the structure linked to the promoter so that each can be expressed has DNA.
  • Those skilled in the art can appropriately prepare the above DNA of the present invention by a general genetic technique. More specifically, the expression vector of the present invention can be prepared by appropriately inserting DNA encoding the RNA of the present invention into various known expression vectors.
  • a preferred embodiment of the vector of the present invention is a vector that expresses RNA (siRNA) capable of suppressing the expression of Versican, C4ST_1, C4ST_2, C4ST-3, and the like by the RNAi effect.
  • siRNA RNA
  • chondroitin sulfate proteodarican core protein synthetic enzyme, desulfating enzyme inhibitory compound, or antibody that binds to sulfotransferase
  • a polyclonal antibody can be obtained as follows. Serum is obtained by immunizing small animals such as rabbits with recombinant (recombinant) protein expressed in microorganisms as a fusion protein with the above-mentioned natural protein or GST, or a partial peptide thereof.
  • ammonium sulfate precipitation protein A, protein G column, DEAE ion exchange chromatography, core protein of the above chondroitin sulfate proteodarican, synthase, desulfase inhibitor compound, or sulfate transferase Or by purification using an affinity column coupled with a synthetic peptide.
  • a monoclonal antibody for example, the above-mentioned chondroitin sulfate proteodarican core protein, synthetic enzyme, desulfase inhibitor compound, or sulfotransferase or its partial peptide is immunized to a small animal such as a mouse.
  • the spleen is removed from the mouse and ground to separate the cells.
  • the cells are fused with mouse myeloma cells using a reagent such as polyethylene glycol. From the above, a clone producing an antibody that binds to the above chondroitin sulfate proteodarican coprotein, synthase, desulfase inhibitor compound, or sulfotransferase is selected.
  • the obtained hyperidoma was transplanted into the abdominal cavity of the mouse, and ascites was collected from the mouse, and the resulting monoclonal antibody was purified using, for example, ammonium sulfate precipitation, protein A, protein G column, DEAE ion exchange chromatography, Coupling of the chondroitin sulfate proteodarican core protein, synthetic enzyme, desulfase inhibitor compound, or sulfotransferase protein or synthetic peptide described above It can be prepared by purification using an affinity column or the like.
  • the antibody of the present invention is not particularly limited as long as it binds to the above-described chondroitin sulfate proteodarican core protein of the present invention, a synthetic enzyme, a desulfase inhibitor compound, or a sulfotransferase.
  • a synthetic enzyme a desulfase inhibitor compound
  • a sulfotransferase a sulfotransferase.
  • human antibodies, humanized antibodies obtained by genetic recombination, and antibody fragments or modified antibodies thereof may also be used.
  • the protein of the present invention used as a sensitizing antigen for obtaining an antibody is not limited with respect to the animal species from which it is derived, but is preferably a protein derived from a mammal such as a mouse, particularly a human-derived protein.
  • a human-derived protein can be appropriately obtained by those skilled in the art using the gene sequence or amino acid sequence disclosed in the present specification.
  • the protein used as the sensitizing antigen may be a complete protein or a partial peptide of the protein.
  • the partial peptide of the protein include an amino group (N) terminal fragment and a carboxy (C) terminal fragment of the protein.
  • antibody means an antibody that reacts with the full length or fragment of a protein.
  • human lymphocytes such as human lymphocytes infected with EB virus are sensitized with proteins, protein-expressing cells or lysates thereof in vitro. And fusion of sensitized lymphocytes with human-derived permanent mitotic cells, such as U266, to produce a hyperidoma that produces the desired human antibody with protein-binding activity. .
  • chondroitin sulfate proteodarican core protein, synthase, desulfase inhibitor compound, or antibody to sulfate group transferase of the present invention binds to the protein, thereby causing expression or function of the protein. An inhibiting effect is expected.
  • a human antibody or a humanized antibody is preferable in order to reduce immunogenicity.
  • the present invention provides a substance capable of inhibiting the function of the above-mentioned chondroitin sulfate proteodarican core protein, synthase, desulfase inhibitor compound, or sulfotransferase.
  • it also contains a low molecular weight substance (low molecular weight compound) that binds to the core protein, synthetic enzyme, desulfurizing oxidase inhibiting compound, or sulfotransferase of the chondroitin sulfate proteodarican described above.
  • the low molecular weight substance may be a natural or artificial compound. Usually, it is a compound that can be produced or obtained by using methods known to those skilled in the art. The compound of the present invention can also be obtained by the screening method described later.
  • the above-mentioned chondroitin sulfate proteodarican core protein synthetic enzyme, desulfase inhibitor protein, or sulfotransferase of the present invention
  • the above-mentioned chondroitin sulfate proteodarican And a mutant having a dominant negative property (dominant negative protein) with respect to a core protein, a synthase, a desulfase inhibitor protein, or a sulfotransferase.
  • the chondroitin sulfate proteodarican core protein, synthase, desulfurase inhibitor protein, or the protein variant having a dominant negative property to sulfate group refers to the core of chondroitin sulfate proteodarican. It refers to a protein having a function of eliminating or reducing the activity of an endogenous wild-type protein by expressing a gene encoding a protein, a synthase, a desulfase-inhibiting protein, or a sulfotransferase.
  • Examples of such dominant negative proteins include Versican core protein mutants that competitively inhibit the binding to chondroitin sulfate with the wild-type Versican core protein.
  • the organ, tissue or cell that inhibits the production or accumulation of chondroitin sulfate proteodarican is not particularly limited, but is preferably an organ or tissue containing nerve cells, more preferably the brain or brain.
  • a compound that inhibits the production or accumulation of chondroitin sulfate proteodarican is expected to be a drug for the treatment or prevention of neurofibrotic degenerative diseases.
  • treatment or prevention refers to a case where it has a partial effect that is not necessarily required to have a complete therapeutic or preventive effect on an organ, tissue, or cell that exhibits neurofibrotic degeneration. It may be.
  • a neurofibrotic degenerative disease is a special disease as long as it is a disease accompanied by neurofibrotic degeneration.
  • a cranial nerve fibrotic degenerative disease more preferably a cerebrospinal nerve fibrotic degenerative disease or a peripheral nerve fibrotic degenerative disease, more preferably Parkinson's disease, Alzheimer's Disease, amyotrophic lateral sclerosis, polyglutamine disease, spinal muscular atrophy, Huntington's disease, or multiple sclerosis.
  • the neurofibrotic degeneration inhibitor of the present invention has an action of suppressing neurofibrotic degeneration by inhibiting the production or accumulation of chondroitin sulfate proteodarican which is the cause of neurofibrotic degeneration.
  • preferred embodiments of the present invention include, for example, a therapeutic agent for cranial nerve fibrotic degenerative disease, a therapeutic agent for cerebrospinal neurofibrotic degenerative disease, a peripheral nerve fiber containing the neurofibrotic degeneration inhibitor of the present invention as an active ingredient.
  • Sexual degenerative disease treatment agent Parkinson's disease treatment agent, Alzheimer's disease treatment agent, amyotrophic lateral sclerosis treatment agent, polyglutamine disease treatment agent, spinal muscular atrophy treatment agent, Huntington's disease treatment agent, or multiple sclerosis Provide therapeutic agents.
  • the “nerve fibrotic degeneration inhibitor” of the present invention is expressed as a “nerve fibrotic degeneration therapeutic agent”, a “nerve fibrotic degeneration inhibitor”, a “nerve fibrotic degeneration improving agent” or the like. Is also possible. Further, in the present invention, the “suppressor” is expressed as “medicine”, “pharmaceutical composition”, “therapeutic drug” or the like.
  • treatment in the present invention includes a prophylactic effect capable of previously suppressing the occurrence of neurofibrotic degeneration.
  • it is not necessarily limited to a case having a complete therapeutic effect on cells (tissues) expressing neurofibrotic degeneration, and may have a partial effect.
  • the drug of the present invention can be mixed with a physiologically acceptable carrier, excipient, certain! /, Diluent, etc. and administered orally or parenterally as a pharmaceutical composition.
  • dosage forms such as granules, powders, tablets, capsules, solvents, emulsions or suspensions can be used.
  • parenteral agents dosage forms such as injections, infusions, drugs for external use, inhalants (nebulizers) or suppositories can be selected.
  • the injection include intracranial injection, intranasal injection, subcutaneous injection, intramuscular injection, and intraperitoneal injection.
  • the medicine for external use include a nasal administration agent or an ointment.
  • a tablet for oral administration can be produced by adding an excipient, a disintegrant, a binder, a lubricant and the like to the drug of the present invention, mixing, and compressing and shaping.
  • an excipient lactose, starch, mannitol or the like is generally used.
  • disintegrant calcium carbonate or carboxymethyl cellulose calcium is generally used.
  • binder gum arabic, carboxymethyl cellulose, or polybulur pyrrolidone is used.
  • talc magnesium stearate and the like are known.
  • the tablet containing the drug of the present invention can be subjected to a known coating for masking or enteric preparation.
  • a coating agent ethyl cellulose, polyoxyethylene glycol or the like can be used.
  • the injection can be obtained by dissolving the agent of the present invention as a main component together with an appropriate dispersant, or dissolving or dispersing in a dispersion medium.
  • aqueous solvent distilled water, physiological saline, Ringer's solution, or the like is used as a dispersion medium.
  • oil-based solvents various vegetable oils such as propylene glycol are used as dispersion media.
  • a preservative such as paraben can be added as necessary.
  • known isotonic agents such as sodium chloride and glucose can be added to the injection.
  • a soothing agent such as benzalkonium chloride pro-hydrochloride can be added.
  • an external preparation can be obtained by making the agent of the present invention into a solid, liquid, or semi-solid composition.
  • a solid or liquid composition it can be set as an external preparation by setting it as the composition similar to what was described previously.
  • a semi-solid composition can be prepared by adding a thickener to an appropriate solvent as required.
  • the solvent water, ethyl alcohol, polyethylene glycol, or the like can be used.
  • the thickener bentonite, polybutyl alcohol, acrylic acid, methacrylic acid, or polyvinyl pyrrolidone is generally used.
  • This composition can be added with a preservative such as benzalkonium chloride.
  • a suppository can also be obtained by combining an oily base material such as cacao butter or an aqueous gel base material such as cellulose derivative as a carrier.
  • a method of administering a vector incorporating a nucleic acid can be mentioned.
  • the above-mentioned vectors include adenovirus vectors, adeno-associated virus vectors, herpes vinores vectors, vaccinia winores betaters, retro winores betaters, and lentivirus vectors. Throw well with the power S.
  • a phospholipid vesicle such as a ribosome
  • the endoplasmic reticulum retaining siRNA or shRNA is introduced into a predetermined cell by the ribofusion method.
  • the obtained cells are then administered systemically, for example, intravenously or intraarterially. It can also be administered locally to neurofibrous degenerated tissue or the like.
  • siRNA it has a very excellent specific post-transcriptional inhibitory effect in vitro. In vivo, it is rapidly degraded by nuclease activity in serum. Therefore, the limited duration in vivo has become a problem, and the development of optimal and effective delivery systems has been demanded.
  • siRNA has been developed for gene therapy for neurofibrotic degenerative diseases by the above method!
  • BAC El Beta-site APP Cleaving Enzyme: ⁇ -secretase siRNA was developed to reduce amyloid formation using in vivo tofence nick mice.
  • a delivery system has been reported in which the BACE1 siRNA is incorporated into a lentiviral vector and expressed in the brain (Singer et al. Nat. Neurosci. (2005) 8 (10) 1343-1349).
  • the necessary amount (effective amount) of the drug of the present invention is administered to mammals including humans within the safe dose range.
  • the dosage of the drug of the present invention is the type of dosage form, In consideration of the administration method, the patient's age and weight, the patient's symptoms, etc., the final decision can be made as appropriate based on the judgment of a doctor or veterinarian. For example, the power varies depending on age, sex, symptoms, administration route, number of administrations, and dosage forms.For example, the dose in the case of adenovirus is about 10 6 to 10 13 per day, 1 week to 8 It is administered at weekly intervals.
  • the application site or the type of the disease is not particularly limited as long as it is a disease that expresses neurofibrotic degeneration.
  • cerebrospinal nerve fibrotic degenerative disease for example, cerebrospinal nerve fibrotic degenerative disease, peripheral nerve fibrotic degeneration Applicable for diseases, Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, polyglutamine disease, spinal muscular atrophy, Huntington's disease, or multiple sclerosis.
  • the above-mentioned diseases may be accompanied with other diseases.
  • the present invention also provides a screening method for a neurofibrotic degeneration inhibitor, which comprises selecting a substance having an action of inhibiting the production or accumulation of chondroitin sulfate proteodarican from a test sample.
  • a screening method for a neurofibrotic degeneration inhibitor which comprises selecting a substance having an action of inhibiting the production or accumulation of chondroitin sulfate proteodarican from a test sample.
  • a preferred embodiment of the screening method of the present invention includes screening a neurofibrotic degeneration inhibitor comprising the step of selecting a substance having the action described in any of the following (a) to (d): Is the method.
  • CSPG chondroitin sulfate proteodarican
  • GAG glycosaminoglycan
  • Test compounds for example, huge compound libraries owned by pharmaceutical companies
  • Method for detecting cut cross section of chondroitin sulfate proteodarican (CSPG) / chondroitin sulfate proteodarican (CSPG) / free glycosaminodarlican (GAG) The above three tools are used. If (1) and (2) are mixed in a test tube or on a culture dish, and the effect is detected easily by (3)!
  • chondroitin sulfate proteodarican, synthase, desulfase inhibitor compound, sulfate transferase, degradation promoting enzyme, and desulfase used are derived from human, mouse, Forces derived from rats and the like are not particularly limited to those derived from these.
  • the part of chondroitin sulfate proteodalycan is a component such as a glycosaminodarican chain, a core protein, or a part thereof, and is not particularly limited.
  • test compound used in the embodiments described below is not particularly limited.
  • natural compounds, organic compounds, inorganic compounds, proteins, peptides and other single compounds, compound libraries examples include gene library expression products, cell extracts, cell culture supernatants, fermented microorganism products, marine organism extracts, plant extracts, and the like.
  • the "contact" to the test compound in the embodiment described below is usually chondroitin sulfate proteodarican, a part thereof, a synthetic enzyme, a desulfase inhibitor compound, a sulfotransferase, a degradation promoting enzyme.
  • the method is not limited to this method, which is performed by mixing a desulfating enzyme with a test compound.
  • the above “contact” can be performed by contacting a cell expressing these proteins or a part thereof with a test compound.
  • the origin of the "cell” in the embodiments described below is not particularly limited to cells derived from humans, mice, rats, etc., and is used in each embodiment. It is also possible to use microbial cells such as Escherichia coli and yeast transformed to express the protein.
  • microbial cells such as Escherichia coli and yeast transformed to express the protein.
  • a cell expressing a chondroitin sulfate proteodarican can be expressed as a cell that expresses an endogenous chondroitin sulfate proteodarican gene or an exogenous chondroitin sulfate proteodarican gene, Cells in which the gene is expressed can be used.
  • Cells expressing the exogenous chondroitin sulfate proteodarican gene are usually created by introducing an expression vector containing the chondroitin sulfate proteodarican gene into the host cell.
  • the power to do S can be produced by general genetic engineering techniques.
  • chondroitin sulfate proteodarican core protein (the matrix is chondroitin sulfate proteoglycan core, and the matrix type is chondroitin sulfate proteoglycan core protein such as aggrican, vers ican, neurocan, brevican, etc.
  • membrane-type chondroitin sulfate proteoglycans are core proteins such as Decorin, Biglycan, Fibromodulin, and PG_Lb
  • synthetic enzymes include, for example, GalNAc4ST_l, GalNAc4ST_2, GALNAC4S-6ST, UA20ST, GalT-I, GalT_H, GlcAT_I, XylosylT, etc.
  • “Sulfyltransferase” includes, for example, C4ST-1 (Chondroitin D—N—acetylgalactosamine—4—0—sulfotransferase 1), 4S ⁇ 2 (Cnonaroitin DN-acetylgalactosamine) -4-0- sulfotransferase 2), C4ST-3 (Chondroitin D_N_acetylgalactosamine_4_0_sulfotransferase 3), D4ST, C6 ST-1, C6ST-2, etc.
  • C4ST-1 Chodroitin D—N—acetylgalactosamine—4—0—sulfotransferase 1
  • 4S ⁇ 2 Nonaroitin DN-acetylgalactosamine
  • C4ST-3 Chodroitin D_N_acetylgalactosamine_4_0_sulfotransferase 3
  • D4ST
  • degradation promoting enzyme are, for example, ADAMTS-1, ADAM TS-4, ADAMTS-5, Chondroitinase ABC (ChABC), Chondroitinase AC, Chondroitinase B, Calpain I, etc.
  • degradation promoting enzyme is, for example, Chondroitin-4- sulfatase, Chondroitm_6_sulmtase (?
  • a method comprising a step of selecting a compound having an action of promoting the degradation of chondroitin sulfate proteodarican.
  • the above method of the present invention includes the following steps, for example.
  • a test compound is brought into contact with chondroitin sulfate proteodarican or a part thereof.
  • the amount of chondroitin sulfate proteodarican or a part thereof is measured.
  • the measurement can be performed by methods known to those skilled in the art.
  • a labeled compound that binds to chondroitin sulfate proteodarican or a part thereof, or It can be detected by measuring the amount of label using an antibody. It can also be detected using a chromatographic method or mass spectrometry.
  • a compound that reduces the abundance of the chondroitin sulfate proteodarican or a part thereof is then selected as compared with the case where the test compound is not contacted (control).
  • the compound that lowers becomes a drug for treating neurofibrotic degeneration.
  • CS-GAG such as chondroitin sulfate A (CS_A), CS_B, CS-C (Seikagaku Corporation, ICN, Sigma, etc.), human-derived proteodalycan (BGN, ISL, etc.), etc.
  • the well plate is coated at a concentration of 10 g / mL (Kawashima H et al .; J. Biol. Chem. 277: 12921-12930, 2002. etc.). Add various test compounds to each well of this plate and detect CS-GAG change after 2 hours reaction at 37 ° C.
  • a WFA lectin (Nodafuji lectin) binding method can be mentioned as a simple method. Since WFA lectin binds to the GalNAc residue of CS-GAG chain, CS-GAG can be easily detected. Chondroitinase ABC is used as a positive control for the test compound. When CS-GAG chain is degraded by chondroitinase ABC addition, WFA lectin cannot be bound, so use that principle.
  • FITC-labeled WFA lectin such as EY
  • the CS-GAG is decomposed to change the FITC fluorescence intensity in the well.
  • the compound with the lowest fluorescence value before and after mixing can be judged as a new treatment candidate compound that satisfies this concept.
  • an anti-CS antibody (clone: CS56, manufactured by Seikagaku Corporation) that directly labels CS-GAG itself can be used.
  • FITC-labeled anti-CS antibody can be added to CS-coated wells so that mass screening can be performed in a very short time and simply if changes in fluorescence values are observed.
  • sGA G Assay Kit manufactured by WIESLAB
  • Sulphanated Glycosaminoglycans ELISA Kit (manufactured by FUNAK OSHI), etc. There is a method to accurately quantify and quantify the GAG content.
  • free GAG is obtained by adding 2-AB (2-aminobenzamide) or 2-AP (2-aminopyridine; V, manufactured by LUD, etc.) to the plate before and after the test compound is mixed. More detailed analysis is possible by simply fluorescently labeling the reducing end of the chain and analyzing each type of sugar chain and the content of each type by HPLC, MALDI-MS, LC-MS, etc. . This is a method for the next stage of screening in which the properties of candidate compounds are examined in detail.
  • a method comprising a step of selecting a substance having an action of inhibiting chondroitin sulfate proteodarican synthesis.
  • the above method of the present invention comprises, for example, the following steps.
  • the synthesis amount of chondroitin sulfate proteodarican or an intermediate in the synthesis process is measured.
  • the measurement can be appropriately carried out by those skilled in the art by a known method, for example, a method using a labeled antibody, mass spectrometry, chromatography, or the like.
  • a compound that reduces (suppresses) the amount of synthesis is selected as compared with the case where the test compound is not contacted (control). Compounds that reduce (suppress) become agents for the treatment of neurofibrotic degeneration. [0127] A simple example of a method and a specific example that can be evaluated (measured) depending on whether the test compound has (b) the activity of inhibiting synthesis! Shown in
  • chondroitin sulfate is produced in 16 hours of cell culture using the standard method of collecting and culturing mononuclear cells after collecting peripheral blood from healthy individuals (Uhlin-Hansen L et al , Blood 82: 2880, 1993, etc.). More simply, known cell lines such as fibroblast cell line MH3T3 (Phillip HA, et al. J. Biol. Chem. 279: 48640, 2004), renal tubular cancer cell line ACHN (Kawashima H et al., J. Biol. Chem.
  • CS-GAG synthase genes such as GalNAc4ST-1 and XylosylT are introduced into CHO cells and L cells in a well-known manner and expressed constantly is created. I can do it.
  • a cell line that constantly synthesizes CS-GAG a therapeutic candidate compound can be determined more clearly.
  • a method including a step of selecting a substance having a desulfating action of chondroitin sulfate proteodarican can be mentioned.
  • the above method of the present invention comprises, for example, the following steps.
  • a test compound is contacted with chondroitin sulfate proteodarican or a part thereof.
  • the amount of chondroitin sulfate proteodarican or a part thereof that has undergone sulfation is measured.
  • the measurement can be performed by methods known to those skilled in the art. For example, it is possible to detect by measuring the amount of label using a labeled compound or antibody that binds to the structure of desulfurization oxidation remaining in chondroitin sulfate proteodarican or a part thereof. It can also be detected using chromatography or mass spectrometry.
  • test compound is not brought into contact with the test compound, compared to the case (control), the abundance of the chondroitin sulfate proteodarican or a part thereof Select a compound that lowers.
  • the compound that lowers becomes a drug for treating neurofibrotic degeneration.
  • human-derived proteodaricans (BGN, ISL, etc.) are prepared and coated on a 96-well plate at a concentration of 10 g / mL (Kawashima H et al.; J. Biol. Chem. 277: 12921-12930, 2002, etc.)). Add various test compounds to each well of this plate and detect CS-GAG change after 2 hours reaction at 37 ° C.
  • the detection method was carried out by desulfating the disaccharide structure of the desulfated fragment remaining on the core protein side of the proteodarican into anti-proteodarican A di4S antibody (clone; 2-B-6, 4 Or the anti-proteodarican ⁇ di6S (clone; 3-B-3, recognizes the part that was sulfated at position 6.
  • the product that has been subjected to desulfation can be easily detected. Therefore, FITC-labeled 2-B-6 and 3-B-3 antibodies can be reacted on the plate before and after mixed culture, and changes in the fluorescence values can be easily detected. Compounds with increased fluorescence intensity before and after reaction are more desulfurized It can be determined that the substance promotes oxidation, and can be easily identified as a novel therapeutic candidate compound that satisfies this concept.
  • a method including a step of selecting a substance having a sulfation inhibitory action of chondroitin sulfate proteodarican can be mentioned.
  • the above method of the present invention comprises, for example, the following steps.
  • a test substance is brought into contact with chondroitin sulfate proteodarican or a part thereof.
  • the amount of chondroitin sulfate proteodarican or a part thereof that has undergone sulfation is measured.
  • the measurement can be performed by methods known to those skilled in the art. For example, it is possible to detect by measuring the amount of labeling using a labeled compound or antibody that binds to chondroitin sulfate proteodarican or a sulfated structure of a part thereof. Moreover, it can also detect using a chromatography method, a mass spectrometry, etc.
  • test compound has the above-mentioned (d) activity of inhibiting sulfation! /, Whether or not! /, And can be evaluated (measured). Show.
  • the cells and cell lines that promote sulfation of chondroitin sulfate are the cells, cells described in (c) above. Consistent with cell lines. In the process of culturing such a cell line for a certain period of time, various test compounds are mixed, and the degree of sulfation before and after culturing is measured, for example, antibody (clone: LY111) for detecting sulfation at position 4 or position 6 An antibody that detects sulfation (clone; MC21C, 1 /, deviation is also available from Seikagaku Corporation) can be easily confirmed. Fluorescence-labeled antibodies may be used to compare fluorescence values before and after culture.
  • detection methods using 2-B-6 and 3-B-3 antibodies may be performed before and after culture. Also good. Compounds that suppress the increase in sulfation after cell culture (LY111 and MC21C fluorescence P), or the progress of desulfation after cell culture (2-B-6 and 3-B-3 fluorescence increase calo) Can be easily determined as a candidate compound that satisfies this concept.
  • a cell line in which a gene of a sulfotransferase such as C4ST-1 or C6ST-1 is introduced into CHO cells or L cells by a well-known method and is expressed constantly. Can be created. By using such a cell line that constantly adds a sulfate group, a therapeutic candidate compound can be determined more clearly.
  • Another preferred embodiment of the present invention is a compound that decreases the expression level of the chondroitin sulfate proteodarican core protein, the synthase, the desulfase inhibitor protein, or the sulfotransferase gene of the present invention.
  • the screening method for a neurofibrotic degeneration inhibitor comprising the following steps (a) to (d): is there.
  • test compound is brought into contact with a cell expressing a gene encoding a chondroitin sulfate proteodlican core protein, a synthetic enzyme, a desulfase inhibitor protein, a sulfotransferase, a degradation promoting enzyme, or a desulfase enzyme.
  • the gene is a chondroitin sulfate proteodarican core protein, synthase, desulfase inhibitor protein, or sulfotransferase
  • the expression level of the gene is reduced compared to the control! /
  • the compound, the gene is chondroitin sulfate
  • a cell expressing a gene encoding a chondroitin sulfate proteodlican core protein, a synthetic enzyme, a desulfurase inhibitor protein, a sulfotransferase, a degradation promoting enzyme, or a desulfurase is expressed.
  • the test compound is brought into contact.
  • chondroitin sulfate proteodarican core protein synthetic enzyme, desulfase inhibitor protein, sulfate transferase, degradation promoting enzyme, or desulfase enzyme
  • gene expression includes both transcription and translation. The gene expression level can be measured by methods known to those skilled in the art.
  • mRNA is extracted from cells expressing any of the above proteins according to a standard method, and the Northern hybridization method, RT-PCR method, DNA array method, or the like using this mRNA as a cage is performed.
  • the amount of transcription of the gene can be measured.
  • a protein fraction is collected from a cell expressing a gene encoding any of the above proteins, and the expression of any of the above! / And any of the proteins is detected by electrophoresis such as SDS-PAGE. It is also possible to measure the translation amount.
  • it is also possible to measure the amount of translation of a gene by detecting the expression of the protein by performing Western blotting using an antibody against any of the above proteins.
  • the antibody used for detecting the protein is not particularly limited as long as it is a detectable antibody. For example, both a monoclonal antibody and a polyclonal antibody can be used.
  • the gene is a chondroitin sulfate proteodarican degradation-promoting enzyme or desulfating enzyme
  • a compound in which the expression level of the gene is increased (enhanced) compared to the control is used.
  • the compound to be increased (enhanced) is a drug for suppressing nerve fiber degeneration or a candidate compound for treating nerve fiber degeneration.
  • the expression level of the core protein, synthase, desulfase inhibitor protein, or sulfotransferase gene of the chondroitin sulfate proteodarican of the present invention is reduced.
  • a method of selecting a compound that increases the expression level of the chondroitin sulfate proteodarican degradation-promoting enzyme or desulfating enzyme gene using the expression of the reporter gene as an index includes, for example, the following steps (a) to (d).
  • the transcriptional regulatory region of a gene encoding a chondroitin sulfate proteodarican core protein, a synthetic enzyme, a desulfase inhibitor protein, a sulfotransferase, a degradation-promoting enzyme, or a desulfase enzyme and the reporter gene are functional.
  • the reporter gene is functionally linked to the core protein, synthase, desulfase inhibitor protein, or sulfotransferase of chondroitin sulfate proteodarican, the expression level of the reporter gene If the reporter gene is functionally linked to a chondroitin sulfate proteodalican degradation-promoting enzyme or desulfating enzyme, the expression level of the reporter gene Selecting a compound that is elevated relative to a control
  • the transcriptional regulatory region and reporter gene of a gene encoding chondroitin sulfate proteodlican core protein, synthase, desulfase inhibitor protein, sulfotransferase, degradation promoting enzyme, or desulfurase A test compound is brought into contact with a cell or cell extract containing DNA having a structure in which and are functionally bound.
  • “functionally bound” refers to chondroitin sulfate proteodalycan core protein, synthase, desulfase inhibitor protein, sulfate transferase, degradation promoting enzyme, Chondroitin sulfate proteolycan core protein, synthase, desulfase inhibitor protein so that the transcription factor binds to the transcriptional regulatory region of the gene encoding desulfase to induce reporter gene expression. It means that a transcriptional regulatory region of a gene encoding a sulfotransferase, a degradation promoting enzyme, or a desulfating enzyme is linked to a reporter gene.
  • the reporter gene is linked to other genes and forms a fusion protein with other gene products, chondroitin sulfate proteodlican core protein, synthase, desulfase inhibitor protein, sulfate If the expression of the fusion protein is induced by binding of a transcription factor to the transcriptional regulatory region of a gene encoding a transferase, a degradation promoting enzyme, or a desulfating enzyme, the above-mentioned "functionally bound" Is included.
  • the reporter gene used in this method is not particularly limited as long as its expression can be detected, for example, CAT gene, lacZ gene, luciferase gene, GFP gene and the like.
  • a structure in which a transcriptional regulatory region of a gene encoding a chondroitin sulfate proteodarican core protein, a synthetic enzyme, a desulfase inhibitor protein, a sulfotransferase, a degradation-promoting enzyme, or a desulfase enzyme and a reporter gene are functionally linked.
  • Examples of the “cell containing a DNA having” include a cell into which a vector having such a structure inserted is introduced. Such vectors can be made by methods well known to those skilled in the art.
  • the introduction of the vector into the cell can be carried out by a general method such as a calcium phosphate precipitation method, an electric pulse perforation method, a lipofussion method, or a microinjection method.
  • a general method such as a calcium phosphate precipitation method, an electric pulse perforation method, a lipofussion method, or a microinjection method.
  • the transcriptional regulatory region of a gene encoding chondroitin sulfate proteodarican core protein, synthase, desulfase inhibitor protein, sulfate transferase, degradation promoting enzyme, or desulfase is functionally associated with the reporter gene.
  • a “cell containing DNA having a linked structure” also includes cells in which the structure is inserted into a chromosome.
  • the introduction of a DNA structure into a chromosome can be performed by a method generally used by those skilled in the art, for example, a gene introduction method using homologous recombination.
  • Transcriptional regulatory region of a gene encoding chondroitin sulfate proteodlican core protein, synthetic enzyme, desulfase inhibitor protein, sulfotransferase, degradation promoting enzyme, or desulfase enzyme and reporter gene are functional.
  • the cell extract containing DNA having a structure bonded to is, for example, a chondroitin sulfate proteodlican core protein, synthase, desulfated enzyme inhibitory protein added to a cell extract contained in a commercially available in vitro transcription translation kit.
  • DNA having a structure in which a transcriptional regulatory region of a gene encoding a sulfotransferase and a reporter gene are functionally linked.
  • contact means "chondroitin sulfate proteodalycan core protein, synthase, desulfase inhibitor protein, sulfotransferase, degradation-promoting enzyme, or transcriptional regulatory region of a gene encoding a desulfase enzyme.
  • the expression level of the reporter gene is measured by! /, And then! /.
  • the expression level of the reporter gene can be measured by methods known to those skilled in the art depending on the type of the reporter gene.
  • the reporter gene is a CAT gene
  • the expression level of the reporter gene can be measured by detecting acetylation of chloramphenicol by the gene product.
  • the reporter gene is the lac Z gene
  • the expression level of the reporter gene can be measured by detecting the fluorescence of the GFP protein.
  • the reporter gene is a gene encoding a chondroitin sulfate proteodalican core protein, a synthetic enzyme, a desulfase inhibitor protein, or a sulfotransferase.
  • a compound that reduces (suppresses) the expression level of the reporter gene compared to the control is selected.
  • a compound that lowers (suppresses) becomes a drug for inhibiting neurofibrotic degeneration or a candidate compound for treating neurofibrotic degeneration.
  • the reporter gene when the reporter gene is functionally linked to a chondroitin sulfate proteodarican degradation-promoting enzyme or a desulfase enzyme, the expression level of the reporter gene is higher than that of the control. Increase (enhance) the compound!
  • the compound to be increased (enhanced) is a drug for suppressing nerve fiber degeneration or a candidate compound for treating nerve fiber degeneration.
  • the neurofibrotic degeneration inhibitor found in the screening method of the present invention is preferably for treating or preventing a neurofibrotic degenerative disease.
  • the present invention also provides a kit containing various drugs, reagents and the like used for carrying out the screening method of the present invention.
  • the kit of the present invention can be appropriately selected from, for example, the above-mentioned various reagents of the present invention according to the screening method to be performed.
  • the kit of the present invention can comprise the chondroitin sulfate proteodarican of the present invention as a constituent element.
  • the kit of the present invention can further contain various reagents, containers and the like used in the method of the present invention.
  • an anti-chondroitin sulfate proteodarican antibody, probe, various reaction reagents, cells, culture solution, control sample, buffer solution, instructions describing the method of use, etc. may be included as appropriate.
  • a screening method for a neurofibrotic degeneration inhibitor comprising a step of detecting whether the production or accumulation of chondroitin sulfate proteodalycan is inhibited. Accordingly, in this screening method, for example, chondroitin sulfate proteolycan that can be used in the detection of chondroitin sulfate proteodarican.
  • Oligonucleotides such as probes for the gene encoding the core protein of darlicans or primers for amplifying any region of the gene, or antibodies that recognize chondroitin sulfate proteodarican (anti-chondroitin sulfate proteodarican antibodies) , And can be included in the components of the screening kit for a neurofibrotic degeneration inhibitor of the present invention.
  • the oligonucleotide specifically hybridizes to, for example, the DNA of the Versican core protein gene of the present invention.
  • “specifically hybridize” means under normal hybridization conditions, preferably under stringent hybridization conditions (for example, Samb Norec et al., Molecular Cloning, Cold Spring Harbor Laboratory Press, New In the conditions described in York, USA, 2nd edition, 1989), it means that cross-hybridization does not occur significantly with DNA encoding other proteins. If specific hybridization is possible, the oligonucleotide does not have to be completely complementary to the base sequence of the Versican core protein gene of the present invention.
  • hybridization conditions include, for example, "2XSSC, 0.1% SDS, 50 ° C”, “2XSSC, 0.1% SDS, 42 ° C", “1XSSC, 0.1% SDS, 37 “C”, more stringent conditions such as “2XSSC, 0.1% SDS, 65.C”, “0.5XSSC, 0.1% SDS, 42 ° C” and “0.2XSSC, 0.1% SDS, 65 ° C” Can be mentioned. More specifically, as a method using Rapid-hy b buffer (Amersham Life Science), pre-hybridization is performed at 68 ° C for 30 minutes or more, then a probe is added, and hybridization is performed at 68 ° C for 1 hour or more.
  • Rapid-hy b buffer Amersham Life Science
  • prehybridization and hybridisation The temperature of the chillon can be set to 60 ° C, and a stringent condition can be set to 68 ° C.
  • a stringent condition can be set to 68 ° C.
  • the oligonucleotide can be used as a probe or primer in the above-described screening kit of the present invention.
  • the length is usually 15 bp to 100 bp, preferably 17 bp to 30 bp.
  • the primer is not particularly limited as long as it can amplify at least a part of the DNA of the gene of the present invention described above, for example, the force described in SEQ ID NO: 69 or 70.
  • the present invention also provides a method for treating or preventing a neurofibrotic degenerative disease, comprising the step of administering the agent of the present invention to an individual (eg, a patient).
  • the individual that is the subject of the prevention or treatment method of the present invention is not particularly limited as long as it is an organism capable of developing a neurofibrotic degenerative disease, but is preferably a human.
  • administration to an individual can be performed by methods known to those skilled in the art, such as intraarterial injection, intravenous injection, and subcutaneous injection.
  • the dose varies depending on the weight and age of the patient, the administration method, etc., but a person skilled in the art (such as a doctor, veterinarian, pharmacist, etc.) can appropriately select an appropriate dose.
  • the present invention relates to the use of the agent of the present invention in the production of a neurofibrotic degeneration inhibitor.
  • Example 1 MPTP Induction Case GalNAcST (siRNA) Treatment with C57BL / 6TcL Parkinson's Disease Model Mice GalNAc4ST-l GALNAC4S-6ST in Chondroitinase ABC Treatment
  • 1-methy ⁇ 4-pheny ⁇ 1,2,3,6 tetrahydropyridine Created a mouse model of Parkinson's disease that selectively denatured min neurons (Amende et al. (2005) Journal of NeuroEngineering and Rehabilitation 2 (20) 1-13) and developed GalNA cST siRNA drug and chondroitinase ABC drug. After administration, the gene expression after treatment and the state of the tissue were compared.
  • MPTP manufactured by Sigma Aldrich Japan
  • BrdU 5 mg / mL ZyMED Laboratory. Inc. 100 1 was administered into the tail vein, and after 1 hour, dissection was performed, the brain was removed, immunostaining sample, gene expression analysis sample Got.
  • RNA-Bee per 1 mg of organ (brain) removed, 1 mL of RNA-Bee (TEL-TEST) is added, and after grinding with an electric homogenizer (DIGITAL HOMOGENIZER, ASONE), chloroform Add 200 ⁇ 1 (Sigma Aldrich Japan) and mix gently, then ice-cool for about 5 minutes and centrifuge using a centrifuge (Centrifbge 5417R, eppendorf) for 15 minutes at 12,000 rpm, 4 ° C. It was.
  • DIGITAL HOMOGENIZER an electric homogenizer
  • RNA concentration in the sample extracted by Otsuka Pharmaceutical Co., Ltd. 50 1 (manufactured by Otsuka Pharmaceutical Co., Ltd.), 100-fold diluted with Otsuka distilled water (manufactured by Otsuka Pharmaceutical Co., Ltd.), and plate reader (POWER Wave XS, BIO-TEK) on a UV plate (Corning Costar) Calculated the RNA concentration in the sample extracted by
  • RNA sample was adjusted to a concentration of 500 rig / 20 ⁇ 1 and set it to 68 ° C for 3 minutes with BLOCK INCUBATOR Heated (ASTEC) and ice-cooled for 10 minutes.
  • ASTEC BLOCK INCUBATOR Heated
  • RT Premix solution (composition: 25 mM MgCl 18.64 ⁇ 1 (Invitrogen)), 5 X Buffer 20 ⁇ 1 (Invitrogen), 0.1 M DTT 6.6 1 (Invitrogen) , 10 mM dNTP mix 10 1 (Invitrogen), RNase Inhibitor 2 ⁇ 1 (Invitrogen), MMLV Reverse transcriptase 1.2 ⁇ 1 (Invitroge ⁇ ), Random primer 2 ⁇ 1 (Invitrogen), sterilization Distilled water 19.56 ⁇ 1 (Otsuka distilled water: Otsuka Pharmaceutical Co., Ltd.) 80 1 was added, BLOCK INCUBATOR (ASTEC Co., Ltd.) was used, and the reaction was heated at 42 ° C for 1 hour.
  • BLOCK INCUBATOR (ASTEC Co., Ltd. was produced) ) was heated at 99 ° C for 5 minutes, and then ice-cooled to prepare cDNA 1001 to be obtained, and the synthesized cDNA was used to carry out PCR reaction with the following composition.
  • PCR Buffer 2 ⁇ 1 Composition: 166 mM (NH 2) SO (Sigma Aldrich Japan), 670 mM Tri
  • Fig. 1 shows the results of gene expression of ⁇ _actin, GalNAc4ST-1 and GALNAC4S-6ST in the untreated group, GalNAcST siRNA-treated group and chondroitize ABC by RT-PCR.
  • the following GalNAcST siRNA (GalNAc4ST-l siRNA cocktail, mixed siRNA of GalNAc4ST_2 siRNA cocktail and GALNAC4S-6ST siRNA cocktail; GeneWorld), Primer (Forward, Reverse) (Hokkaido System Science) Show.
  • [0179] [Primer sequence]
  • Example 2 MPTP-induced C57BL / 6. TcL Parkinson's disease model mouse comparison test of chondroitinase ABC GalNAcST siRNA treatment to suppress the deposition of CSPG in the brain
  • This example uses a brain tissue sample of Parkinson's disease model mouse
  • a comparative study was conducted on the effect of CSPG deposition suppression.
  • the brain tissue obtained in Example 1 was embedded in a freezing embedding agent OCT compound (manufactured by Miles), and a frozen block was prepared with liquid nitrogen. A 10 m thick section was prepared from the frozen block using a cryostat (Microm).
  • the obtained sections were fixed with acetone (manufactured by Sigma Aldrich Japan) for 10 minutes, washed with a phosphate buffer, and further anti-chondroitin sulfate proteodarican (CSPG) antibody (clone CS56, mouse monoclonal antibody, lO as a primary antibody).
  • CSPG anti-chondroitin sulfate proteodarican
  • g / ml manufactured by Seikagaku Corporation
  • a secondary antibody reaction was performed using a histofine mouse stin kit (manufactured by Nichirei; used for mouse monoclonal antibody), and then DAB substrate (manufactured by Nichirei) was added to carry out an enzyme dye reaction.
  • Example 3 Comparison of the suppression of inflammation associated with macrophage infiltration in chondroitinase ABC GalNAcST siRNA treatment using MPTP-induced C57BL / 6.TcL Parkinson's disease model mice
  • the CSPG deposition shown in Example 2 is known to adsorb chemokines, which are in vivo substances that induce inflammatory cells such as macrophages. In addition, it was speculated that the deposition of CSPG would eventually lead to attracting inflammatory cells to induce destruction of brain tissue. Similarly, the effects of GalN AcST administration and chondroitinase ABC on brain macrophage accumulation kinetics were compared using brain tissue samples. Sections obtained in the same manner as in Example 2 were fixed with 4% PFA (Paraformaldehyde) phosphate buffer (Nacalai Testa) for 10 minutes, washed with deionized water, and rat-derived anti-mouse as the primary antibody.
  • PFA Paraformaldehyde
  • Macrophage antibody (clone F4 / 80; 1: 200 dilution; manufactured by BMA) was added, and the mixture was reacted at 4 ° C.
  • the secondary antibody Alexa488-labeled anti-rat IgG goat antibody (1: 200 dilution; Invitrogen) was added and allowed to react at room temperature for 30 minutes.
  • the tissue image obtained by the above method is shown in Fig. 3 (the original image is a power error).
  • the strong positive signal in the untreated group was a result of the accumulation of more macrophages in the tissue margin than in the control group.
  • the results were almost the same as in the control group.
  • Example 4 MPTP induction C57BL / 6TcL Parkinson's disease model mouse chondroitinase ABOGalNAcST siRNA ⁇ i ratio of cellulose JMl force f meter CSPG deposition shown in Example 2 may further promote cell fibrosis
  • tissue samples from brain tissue we examined tissue findings obtained by in vivo administration of GalNAcST and chondroitinase ABC for fibrosis in neurons in the brain. Compared.
  • Sections obtained in the same manner as in Examples 2 and 3 were fixed for 10 minutes with 4% PFA phosphate buffer lysate), washed with deionized water, and fibroblast antibody ( ER-TR7; 1: 100 dilution; manufactured by BMA) was added and allowed to react overnight at 4 ° C.
  • fibroblast antibody ER-TR7; 1: 100 dilution; manufactured by BMA
  • Alexa488-labeled anti-rat IgG goat antibody (1: 200 dilution; Invitrogen) was added and allowed to react at room temperature for 30 minutes.
  • the tissue image obtained by the above method is shown in Fig. 4 (the original image is in color).
  • a strong positive signal in the untreated group was the result of fibroblast infiltration in the brain in the vicinity of the enlarged cortex of the posterior corpus callosum than in the control group. Furthermore, when compared with the findings in the GalNAcST siRNA and chondroitinase ABC treatment group, the positive findings of fibroblasts were unidentifiable as in the control group. From the above results, Parkinson The findings of ER-TR7 positive signal in brain tissue induced by a disease mouse model were found to be significantly suppressed by in vivo administration of Gal NAcST siRNA and chondroitinase ABC.
  • Example 5 Comparative study of the effect of astrocytes on treatment with GalNAcST (siRNA) chondroitinase ABC in MPTP-induced C57BL / 6.TcL Parkinson's disease model mice In addition to numerous neurons in the brain, glial cells There are cells that function to supply nutrients.
  • anti-GFAP antibody was used to stain astrocytes in glial cells, and the tissue findings obtained by in vivo administration of GalNAcST and in vivo administration of chondroitinase ABC were compared.
  • FIG. 5 shows tissue images of the control group, the untreated group, the GalNAcST siRNA, and the chondroitinase ABC treatment group (the original figure is color).
  • Example 6 GalNAcST (by live C57BL / gJgL Parkinson's mouse) Comparative study of the effect of dopamine neurons on siRNA treatment
  • tyrosine hydroxylase is an enzyme that converts dopamine precursors into dopamine. Sections obtained in the same manner as in Examples 2, 3, and 4 were fixed with 4% PFA phosphate buffer (manufactured by Nacalai Tester) for 10 minutes, washed with deionized water, and rabbit polyclonal as a primary antibody. Anti-tyrosine hydroxylase antibody (1:50 dilution; Calbiochem) was added and allowed to react at room temperature for 1 hour.
  • FIG. 6 shows tissue images of the control group, the untreated group, the GalN AcST siRNA, and the chondroitinase ABC treatment group (the original figure is the same). In the vicinity of the upper mesencephalon, it was confirmed that the tyrosine neuroxylase was normally expressed in the control group! /, But it appeared as a negative signal in the untreated group! This suggests that MPTP selectively destroyed dopamine neurons.
  • the present inventor examined the effect on actual CSPG expression by immunostaining using CS-56 antibody (Fig. 7).
  • the photo shows the findings near the dentate gyrus.
  • a CSPG positive signal is strongly expressed.
  • C4-sulfata se treatment decreased the expression of positive signals.
  • the expression of inflammation markers and fountain maintenance markers was examined. As shown in FIG. 8, compared to the untreated group, there was a significant effect of suppressing the difference in the expression of TNF- ⁇ and TGF- / 3 in the treated group!
  • Example 7 MPTP induction C57BL6 TcL Parkinson's disease model mice in C4-sulfatas e ⁇ i CSPG ⁇ ne JMI power ratio f meter
  • a mouse model of Parkinson's disease in which donomin neurons were selectively denatured with 1-methy ⁇ 4-pheny ⁇ 2,3-dihydropyridinium (MPDP) was administered and treated with C4-sulfatase drug. Later, gene expression and tissue appearance were compared.
  • MPDP 1-methy ⁇ 4-pheny ⁇ 2,3-dihydropyridinium
  • CSPG deposition suppression effect was compared!
  • the obtained brain tissue was embedded in a freezing embedding agent OCT compound (manufactured by Miles), and a frozen block was prepared with liquid nitrogen.
  • a section having a thickness of 10 ⁇ m was prepared from the frozen block using a cryostat (manufactured by Microm).
  • the obtained sections were fixed with acetone (manufactured by Sigma Aldrich Japan) for 10 minutes, washed with a phosphate buffer, and further anti-chondroitin sulfate proteodarican (CSPG) antibody (clone CS56, as a primary antibody).
  • CSPG anti-chondroitin sulfate proteodarican
  • Mouse monoclonal antibody (lO ⁇ g / ml; manufactured by Seikagaku Corporation) was added and reacted at room temperature for 1 hour. Subsequently, after performing a secondary antibody reaction using a histofine mouse stin kit (manufactured by Nichirei; used for mouse monoclonal antibody), DAB substrate (manufactured by Nichirei) was added to perform an enzyme dye reaction. It was. This specimen was observed with an optical microscope (Leica). This tissue image is shown in FIG. It was found that the positive signal in the untreated group was stronger than the control group's CSPG accumulation near the dentate gyrus.
  • RNA concentration in the sample extracted by (POWER Wave XS, manufactured by BIO-TEK) was calculated. Next, the following procedure was performed to perform RT reaction (cDNA synthesis).
  • RNA sample was adjusted to a concentration of 500 rig / 20 ⁇ 1, heated at 68 ° C for 3 minutes with BLOCK INCUBATOR (ASTEC), and ice-cooled for 10 minutes.
  • RT Pre Mix solution composition: 25 mM MgCl 18.64 ⁇ 1 (Invitrogen)), 5 X Buffer 20 ⁇ 1 (Invitrogen), 0.1 M DTT 6.6 ⁇ 1 (Invitrogen) ), 10 mM dNTP mix 10 ⁇ 1 (Invitrogen), RNase Inhibitor 2 ⁇ 1 (Invitrogen), MMLV Reverse transcriptase 1.2 ⁇ 1 (Invitrogen), Rando Add m primer 2 ⁇ 1 (Invitrogen), sterile distilled water 19.56 ⁇ 1 (Otsuka distilled water: Otsuka Pharmaceutical Co., Ltd.) 80 1 and heat reaction at 42 ° C for 1 hour using BLOCK INCUBATOR (ASTEC) 1 hour later, heat at 99 ° C for 5 minutes with
  • FIG. 8 shows that TNF-a and TGF- ⁇ gene expression was suppressed by C4_sulfatase treatment.
  • TNF-a Tumor Necrosis Factor
  • TGF- ⁇ Transforming growth factor-beta
  • tissue sample sections were stained with an anti-tyrosine hydroxylase antibody using a dopamine neuron marker, and the tissue findings were compared.
  • This tyrosine hydroxylase (TH) is a dopamine precursor It is an enzyme that converts to dolomine.
  • the section obtained in the same manner as in Example 7 was fixed with 4% PFA phosphate buffer solution (manufactured by Leytester) for 10 minutes, washed with deionized water, and rabbit polyclonal anti-tyrosine hydroxylase as the primary antibody.
  • Antibody (1:50 dilution; Calbiochem) was added and allowed to react for 1 hour at room temperature.
  • the survival rate of C4-sulfatase and GalNAc4S_6ST siRNA treated group and untreated group in Parkinson's disease model induced by MPDP administration! / Using the Kaplan Meier survival curve And compared.
  • the survival rate on day 7 was 16.7% in the untreated group, 50% in the C4-sufatase treated group, and 71.4% in the GalNAc4S-6ST siRNA treated group.
  • treatment with C4-sulfatase or GalNAc4S_6ST siRNA prevents the overexpression and over-deposition of CSPG, thereby preventing the adsorption of chemokines related to the induction of inflammatory cells, and prevents the destruction of brain tissue in the inflammatory cells attracted by the deposition of CSPG.
  • GalNAc4S-6ST siRNA The sequence of GalNAc4S-6ST siRNA is shown below.
  • chondroitin sulfate proteodarican (CSPG) accumulation As an example of examining the effect of chondroitin sulfate proteodarican (CSPG) accumulation according to the present invention, the side chain of chondroitin sulfate proteodarican (acetilgalatatosamine stone) 3 ⁇ 4 acid group was transferred.
  • CSPG chondroitin sulfate proteodarican
  • N-acetylgalactosamine- 4-0- sulfotransferase N-acet ylgalactosamine- 4-0- sulrotransferase-l, N-acetylgalactosamine- 4-0- sulrotransfera se ⁇ 2, N-acetylgactosamine-4-sulfate 6-O Chondroitinase ABC, a degrading enzyme of chondroitin sulfate, which is a side chain of the siRNA of -sulfotransferase (GalNAc4ST-l, GalNAc4 ST-2, GALNAC4S-6ST), is a GAG chain associated with chondroitin sulfate proteodarican in the hypothalamus of the brain It is effective in treating or preventing Parkinson's disease by suppressing the sulfation and accumulation of and inhibiting the cell death of dopamine neurons.
  • neurofibrotic degenerative diseases thought to be caused by the accumulation of abnormal proteins; Alzheimer's disease, polyglutamine disease, amyotrophic lateral sclerosis, spinal muscular atrophy, Huntington's disease and multiple sclerosis Therefore, it is considered that overexpression and accumulation of chondroitin sulfate proteodalycan in the present invention is a factor that reduces brain function. Since the neurofibrotic degeneration inhibitor according to the present invention can effectively improve lesions by an unprecedented mechanism of action and drug therapy, it can be an excellent therapy useful for further improvement of patient QOL and medical treatment.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Neurosurgery (AREA)
  • Animal Behavior & Ethology (AREA)
  • Neurology (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Molecular Biology (AREA)
  • Urology & Nephrology (AREA)
  • Biochemistry (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • Hospice & Palliative Care (AREA)
  • Toxicology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Biotechnology (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Psychiatry (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

It was verified that by an experiment using a gene-silencing nucleic acid (GalNAcST siRNA), which is a substance having an “action of inhibiting the synthesis” of chondroitin sulfate proteoglycan (CSPG), chondroitinase ABC, which is a substance having an “action of promoting the degradation” of CSPG and C4-sulfatase, which is a substance having an “action of desulfating” CSPG, a substance that “inhibits the production or accumulation of CSPG” in a totally different route of a totally different property has an effect of inhibiting nerve fiber degeneration. It was revealed that the substance that “inhibits the production or accumulation of CSPG” which is a feature common to these is useful for inhibiting nerve fiber degeneration. The invention relates to a nerve fiber degeneration inhibitor containing a substance that inhibits the production or accumulation of CSPG as an active ingredient and a screening method for the nerve fiber degeneration inhibitor.

Description

明 細 書  Specification
神経線維性変性抑制剤  Neurofibrotic degeneration inhibitor
技術分野  Technical field
[0001] 本発明は、コンドロイチン硫酸プロテオグリカン(CSPG:Chondroitin Sulfate Proteogl yeans)の蓄積制御に基づく神経線維性変性疾患の治療薬と予防薬、神経線維性変 性の抑制方法、及び該方法に基づくアルツハイマー病(AD Alzheimer's disease)や パーキンソン病(PD:Parkinson's disease)、また筋萎縮性側索硬化症(ALS: Amyotro phic Lateral Sclerosis)等を含む神経線維性変性疾患の治療又は予防方法に関する 背景技術  [0001] The present invention relates to a therapeutic and prophylactic agent for neurofibrotic degenerative diseases based on control of accumulation of chondroitin sulfate proteoglycan (CSPG), a method for inhibiting neurofibrotic degeneration, and Alzheimer based on the method. Background art on methods of treating or preventing neurofibrotic degenerative diseases including AD Alzheimer's disease, PD (Parkinson's disease), and amyotro phic Lateral Sclerosis (ALS)
[0002] 神経線維性変性疾患における脳神経線維性変性疾患はニューロンの細胞死によ る減少から生じる難病として認識されている。そして、この神経線維性変性疾患には 大きく記憶と痴呆に関する症状、また動作に関する症状を表す二つのグループに分 けられる。代表的な例として、前者はアルツハイマー病が挙げられ、後者はパーキン ソン病が挙げられる。アルツハイマー病は、全世界で 1800万人以上の患者で 20年後 では 3400万人へと増加すると予測されている(国際アルツハイマー病協会: ADI)。現 在、 日本全国では少なくとも 30万人の患者であと 4年後の 2010年には 80万人を超す と推測されている。この疾患は遺伝子解析と統計学的では男性より女性に多く発症 することが多ぐ高齢者に多く見られる病気である(丸美屋和漢薬研究所の調査)。  [0002] Cranial nerve fiber degenerative disease in neurofibrotic degenerative diseases is recognized as an intractable disease resulting from a decrease due to neuronal cell death. This neurofibrotic degenerative disease can be roughly divided into two groups representing symptoms related to memory and dementia, and symptoms related to movement. As a typical example, the former is Alzheimer's disease and the latter is Parkinson's disease. Alzheimer's disease is projected to increase to 34 million in 20 years in over 18 million patients worldwide (International Association for Alzheimer's Disease: ADI). Currently, it is estimated that there will be at least 300,000 patients in Japan and more than 800,000 in 2010, four years later. According to genetic analysis and statistical analysis, this disease is more common among elderly people who are more common in women than in men (survey by the Marumaya Japanese Medicine Research Institute).
[0003] アルツハイマー病には遺伝的に発症する家族性アルツハイマー病とアミロイド 0ぺ プチド (A /3 )の産生やその分解に関与する因子の不具合により引き起こされる代謝 バランス(アセチルコリンの減少)の異常による孤発性アルツハイマー病の二種類が 存在する。どちらの因子においても A /3の脳内蓄積の根本原因によるもので、神経細 胞に老人班が現れてくる症状である。 A /3はアミロイド前駆体タンパク質 (APP:Amyloi d precursor protein)からプロテアーゼで切り出されることによって生成され、 β -シー ト構造の割合が増加して不溶、凝集することが報告されている。また、近年の大きな 発見としてアミロイドの蓄積を防ぐ分解酵素であるネプライシンの同定がなされ、この 活性が減少すると脳内に A /3の蓄積が多くなることが実証された。これに付随して、ァ デノウィルスによるネプライシンの脳内発現を起こし脳内の A β量の減少を確認した 報告がある(非特許文献 1)。さらに、アルツハイマー病の危険因子としてアポリポタン ノ ク質(アポ Ε)を発現する遺伝子が大きく関わって!/、ることが報告されて!/、る(非特許 文献 2)。以上の知見以外にも様々な要因に対して精力的に研究されいくつかの原 因タンパク質や遺伝子の変異等が見つかつている。その例として、高度にリン酸化さ れて!/、るタウタンパク質の蓄積による神経原線維変性とアミロイド前駆体タンパク質( APP:Amyloid precursor protein)とプレセ二リン 1および 2をコードする遺伝子の変異 により A 13の蓄積を促進することが報告されて!/、る(非特許文献 3)。 [0003] Alzheimer's disease is due to genetically-onset familial Alzheimer's disease and abnormal metabolic balance (decreased acetylcholine) caused by a defect in the factors involved in the production of amyloid 0 peptide (A / 3) and its degradation. There are two types of sporadic Alzheimer's disease. Both factors are caused by the root cause of the accumulation of A / 3 in the brain, and are symptoms in which senile plaques appear in nerve cells. It has been reported that A / 3 is produced by cleaving from amyloid precursor protein (APP) with protease, and the proportion of β-sheet structure is increased, resulting in insolubility and aggregation. In addition, a major discovery in recent years was the identification of neprisin, a degrading enzyme that prevents amyloid accumulation. It has been demonstrated that accumulation of A / 3 increases in the brain as activity decreases. Along with this, there has been a report confirming a decrease in the amount of Aβ in the brain due to the expression of neprisin in the brain by adenovirus (Non-patent Document 1). Furthermore, it has been reported that genes that express apolipotanic protein (apo Ε) are greatly involved as a risk factor for Alzheimer's disease! (Non-patent document 2). In addition to the above-mentioned findings, research has been conducted energetically for various factors, and several causative proteins and gene mutations have been found. Examples include highly phosphorylated! /, Neurofibrillary degeneration due to accumulation of rutau protein and mutations in genes encoding amyloid precursor protein (APP) and presenilin 1 and 2. It has been reported to promote the accumulation of A13!
[0004] 上記のように多数の研究報告がされている力 まだ日本国内で発売が認められて いるアルツハイマー病の治療薬は塩酸ドネぺジル(商品名:ァリセブト)しかなぐアル ッハイマー病の症状には痴呆の進行を約 9ヶ月遅らせる効果があるものの認知機能 の回復を促す効果はな!/、。この治療薬は記憶や学習に関係してレ、る神経伝達物質 であるアセチルコリンを分解する酵素のアセチルコリンエステラーゼを阻害することに より脳内において減少したアセチルコリン濃度を高める目的のものである。塩酸ドネ ぺジルはアルツハイマー病の根治薬としての役割を持たず軽度の患者にのみ投与さ れるといった薬でしかない。現在、アルツハイマー病に関して各研究機関において病 気の発症因子を発見するため動物モデルを作製し、試行錯誤して!/、るが多種の因 子が関わっているためにまだ決め手となるような治療法や治療薬は確立されていな い。 [0004] The power of many research reports as described above Alzheimer's disease is the only therapeutic drug for Alzheimer's disease that is still approved for sale in Japan. Symptoms have the effect of delaying the progression of dementia by about 9 months, but do not promote the recovery of cognitive function! This therapeutic drug is intended to increase the concentration of acetylcholine that has been reduced in the brain by inhibiting acetylcholinesterase, an enzyme that degrades acetylcholine, a neurotransmitter that is related to memory and learning. Donepezil hydrochloride does not serve as a curative for Alzheimer's disease and is only given to mild patients. Currently, each research institution for Alzheimer's disease has created animal models to discover disease-causing factors, trial and error, and treatments that are still a decisive factor because many factors are involved. No law or treatment has been established.
[0005] 次に、中枢神経線維性変性疾患のうちアルツハイマー病の患者数よりは少な!/、疾 患であるが難病疾患として挙げられているパーキンソン病がある。これは、中脳の黒 質として知られている脳の一部に存在するドーパミンニューロンの細胞死が原因とな りドーパミン(黒質内のニューロンにて作られる神経伝達物質)の量が減少し、もう一 つの神経伝達物質であるアセチルコリンとのバランスが崩れアセチルコリンが相対的 に多くなることにより引き起こされる病気で、その症状としては振戦(ふるえ)、筋肉の 固縮、動作の緩慢および姿勢保持障害と言われる 4大症状がある。 日本において、こ の病気は男女比 1: 1.5〜2でやや女性に多い傾向があり 30代〜 80代といった幅広い 年齢層で発症されている。現在の患者数は、人口 10万人に対して 100人前後の割合[0005] Next, among the central nervous fibrotic degenerative diseases, there are fewer than the number of patients with Alzheimer's disease! / There is Parkinson's disease, which is a disease but is listed as an intractable disease. This is due to the death of dopamine neurons in the part of the brain known as the substantia nigra, which reduces the amount of dopamine (a neurotransmitter made by neurons in the substantia nigra). This is a disease caused by a loss of balance with acetylcholine, another neurotransmitter, and a relative increase in acetylcholine. The symptoms include tremor, muscle stiffness, slow motion and posture maintenance. There are four major symptoms called disability. In Japan, this disease tends to be more common in women with a male-female ratio of 1: 1.5-2, and a wide range of people in their 30s and 80s It has developed in the age group. The current number of patients is around 100 per 100,000 population
(厚生科学審議会疾病対策部会難病対策委員会議事録)で発症しており日本全国 で約 12万人の患者がいると推測されている。また、その全体の 5〜10%が遺伝性パ 一キンソン病として占めている。現在、病因究明の研究が世界各国で盛んに進んで おり、家族性パーキンソン病の患者における遺伝子の解析の結果から、その原因遺 伝子として a -synuclein (タンパク質のフォールデイングの異常と蓄積)、 parkin (タン パク質分解システムとミトコンドリア機能)、 PINK1 (ミトコンドリア機能)、 DJ-1 (酸化スト レス)、 LRRK2 (リン酸化)が同定されている力 これらの遺伝子がどのようにパーキン ソン病に関与しているのかについてはまだ解明されていない。また、脳内におけるミ スフオールデイングや凝集した蛋白質の蛋白分解機構に関わる遺伝子の突然変異 により蛋白質の蓄積が神経線維性変性疾患の要因であるという研究が報告されてい る(非特許文献 4〜8)。 It is estimated that there are about 120,000 patients in Japan throughout the country. In addition, 5 to 10% of all cases account for hereditary Parkinson's disease. Currently, research on the etiology has been actively conducted around the world, and from the results of gene analysis in patients with familial Parkinson's disease, a-synuclein (abnormality and accumulation of protein folding), Parkin (protein degradation system and mitochondrial function), PINK1 (mitochondrial function), DJ-1 (oxidation stress), LRRK2 (phosphorylation) have been identified how these genes are involved in Parkinson's disease It is not yet elucidated whether they are doing it. In addition, studies have been reported that protein accumulation is a factor in neurofibrotic degenerative diseases due to mutations in genes related to the protein folding mechanism in the brain and the protein degradation mechanism of aggregated proteins (Non-Patent Documents 4 to 4). 8).
[0006] 近年、上述の他に報告されている神経線維性変性疾患としてポリグルタミン病、筋 萎縮性側索硬化症、脊髄性筋萎縮症、ハンチントン病そして多発性硬化症などがあ るがこれらも異常タンパク質の蓄積による小胞体ストレスが神経細胞を死に至らせる ことが報告されている。本来、異常タンパク質はタンパク質分解システム(UPR: unfold ed protein response, ERAD: ER associated degrdation,ュビキチン-プロァァソーム) により分解され排除される力 過剰な異常タンパク質に対しては十分な反応ができず 細胞は細胞死のシグナルを発するようになる(非特許文献 9)。このように細胞内では 複雑なシグナル伝達が起こり様々な経路を経て神経線維性変性を誘導させて!/、る。 そこで、各々の疾患においてシグナル伝達等の研究が行われているがどれも複雑で シグナル伝達に対する治療薬の開発は困難を極めている。  [0006] In recent years, other neurofibrotic degenerative diseases that have been reported include polyglutamine disease, amyotrophic lateral sclerosis, spinal muscular atrophy, Huntington's disease, and multiple sclerosis. It has also been reported that endoplasmic reticulum stress due to abnormal protein accumulation leads to death of neurons. Originally, abnormal proteins are decomposed and eliminated by proteolytic system (UPR: unfolded protein response, ERAD: ER associated degrdation, ubiquitin-prosome). A death signal is emitted (Non-patent Document 9). In this way, complex signal transduction occurs in the cell and induces neurofibrotic degeneration through various pathways! / Therefore, studies on signal transduction and the like have been conducted for each disease, but all are complicated and it is extremely difficult to develop a therapeutic agent for signal transduction.
[0007] 現在、多種の神経線維性変性疾患における治療の中ではパーキンソン病における 治療が進んでおり、運動理学療法、外科的治療と薬物療法に分けられているが、薬 物療法が一般に広く用いられている。特に、振戦や他の症状に最も効果がある L-ド ーパ(ドーノ ミンの前駆体)、ドーノ ミン受容体刺激薬 (L-ドーパ効果の上昇)、抗コリ ン薬 (アセチルコリン抑制剤)、ドーパミン放出促進薬、ノルアドレナリン補充薬 (すく み現象の対処薬)、ドーパミン分解阻害剤(MAO-B阻害剤、 COMT阻害剤)などが症 状に合わせて使用されている。し力、しながら、これらの治療薬は根治薬としての働き は無く症状の緩和に対する薬として用いられている。また、長期投与によりさまざまな 問題が現れてくる場合が確認されている。例えば L-ドーパの場合 5-10年といった長 期投与によって、 wearing off現象 (薬効時間の短縮)、 on-off現象 (薬効喪失)、ジス キネジァ (不随意運動)、幻覚 ·妄想などの精神症状が出現する。 [0007] Currently, treatment for Parkinson's disease is advancing among treatments for various neurofibrotic degenerative diseases, which are divided into kinematic physical therapy, surgical treatment, and pharmacotherapy, but drug therapy is generally widely used. It has been. In particular, L-dopa (precursor of donomin), donomin receptor stimulant (increased L-dopa effect), anticholinergic agent (acetylcholine inhibitor), which is most effective for tremor and other symptoms , Dopamine release promoters, noradrenaline supplements (remedies for freezing), dopamine degradation inhibitors (MAO-B inhibitors, COMT inhibitors), etc. It is used according to the shape. However, these therapies do not act as curatives and are used to relieve symptoms. In addition, it has been confirmed that various problems may appear due to long-term administration. For example, in the case of L-Dopa, long-term administration such as 5 to 10 years can cause wearing off phenomenon (shortening of drug effect), on-off phenomenon (loss of drug effect), dyskinesia (involuntary movement), psychiatric symptoms such as hallucination and delusion. Appears.
[0008] これから更なる高齢化社会になりつつある現在、これまでとは全く違った視点、つま り薬物治療による一時的な症状の緩和だけではなぐ病気の根本となる原因を探り阻 止するようなメカニズムで安全性に優れ、有効的に阻止又は副作用を抑制させること が出来る新しい薬剤の開発が切望されている。現在、脳神経疾患は世界中の中枢と なる研究所を中心として研究が進められている。そして様々な創薬が考えられていて 、最近では「脳の遺伝子治療」が根本的な治療法のひとつとして期待を集めている( 非特許文献 10〜12)。 [0008] As we are becoming an aging society in the future, we will try to find and prevent a completely different viewpoint, that is, the root cause of illness that is not merely by temporary relief of symptoms by drug treatment. There is a strong need for the development of new drugs that have excellent safety and can effectively prevent or suppress side effects. At present, cranial nerve disease is being researched mainly by research centers that are central to the world. Various drug discovery has been conceived, and recently, “brain gene therapy” has been expected as one of the fundamental treatment methods (Non-Patent Documents 10 to 12).
[0009] ここで、上述の神経線維性変性疾患治療の為に着目したプロテオダリカンは、コア 蛋白質と呼ばれる蛋白質に、 1本以上のグリコサミノダリカン (GAG)鎖が共有結合し た構造をもつ分子で、 GAG鎖の特異的な糖鎖構造がプロテオダリカンの多彩な機能 を担うと考えられており、プロテオダリカンは GAG鎖の種類に基づいて、コンドロイチ ン¾¾酸プロテオグリカン(CSPG:chondroitin sulfate proteoglycans)、デノレマタン石) ¾酸 プロテオグリカン(DSPGs:dermatan sulfate proteoglycans)、 へパラン 酸プロテオグ リカン(HSPGs:he印 aran sulfate proteoglycans)、ケタラン硫酸プロテオグリカン(KSP Gs:keratan sulfate proteoglycans)と 4つに大別される。 (非特許文献 13〜 19)この中 で、 HSPGsに関しては、多彩なサイト力イン、接着分子、ケモカインと結合し、その機 能を大きく修飾するため幅広く研究されてきた。月 内における働きとして、 HSPGsとコ ァ蛋白質力 側鎖として伸長している GAG鎖は非線維化アミロイド β -プロテインを神 経毒性線維化アミロイド /3 -プロテインに転換、そして蛋白分解機構に逆らいアミロイ ド 13 -プロテイン (Α β )を保護するとレ、う報告がある(非特許文献 20〜22)。同様に、 へパラン硫酸プロテオダリカンとグリコサミノダリカンはパーキンソン病の病因として最 も有力視されてレ、る -synucleinをも線維化するのではな!/、か、またアルツハイマー 病の原因タンパク質であるアミロイド前駆体タンパク質 (APP)をも変性させて!/、るので はなレ、かと考えられてレ、る (非特許文献 23)。 [0009] Here, proteodarican focused on the treatment of the above-mentioned neurofibrotic degenerative disease has a structure in which one or more glycosaminodarlican (GAG) chains are covalently bound to a protein called a core protein. It is thought that the specific sugar chain structure of the GAG chain is responsible for the various functions of proteolidicans. Proteolidicans are based on the type of GAG chains and chondroitin ¾¾ acid proteoglycans (CSPG: chondroitin) sulfate proteoglycans), denorematanite) ¾ acid proteoglycans (DSPGs: dermatan sulfate proteoglycans), heparan acid proteoglycans (HSPGs) Is done. (Non-Patent Documents 13 to 19) Among these, HSPGs have been extensively studied to bind to various site force-ins, adhesion molecules, and chemokines and greatly modify their functions. As a function in the month, HSPGs and co-protein strength GAG chain, which is extended as a side chain, converts non-fibrotic amyloid β-protein into neurotoxic fibrotic amyloid / 3-protein and amyloid against the proteolytic mechanism There is a report that protects 13-protein (Αβ) (Non-patent Documents 20 to 22). Similarly, heparan sulfate proteodarican and glycosaminodarlican are considered to be the most prominent etiology of Parkinson's disease and do not fibrize -synuclein! /, Or a protein that causes Alzheimer's disease Amyloid precursor protein (APP) is also denatured! / Hanare is considered to be a thing (Non-Patent Document 23).
[0010] 一方、 CSPGに関しては、胎生時期には必須分子で、各臓器に豊富に存在する。神 経幹細胞の分化誘導剤(特許文献 1)、ヒト/骨形成蛋白を用いる神経再生 (特許文 献 2)、ヒト、骨形態形成蛋白を用いる神経再生(特許文献 3)のような神経再生に関 わる過程を制御して!/、る分子として考えられて!/、る一方、中枢神経系の損傷の治療( 特許文献 4)、神経組織の修復を促進するための材料および方法(特許文献 5)や血 管平滑筋細胞の治療阻害因子(特許文献 6)などの様々な箇所における神経再生の 阻害にも関与している。その例として、まず中枢神経の損傷時に発現してくる再生阻 害物質であるコンドロイチン硫酸プロテオダリカン群としてニューロカン、ブレビカン、 NG2が同定され(非特許文献 24)、これらにコンドロイチナーゼ ABC (GAG鎖の一種で あるコンドロイチン硫酸を選択的に除去する酵素)投与により CSPGを分解して中枢神 経の再生を促す(特許文献 4)という報告例がある。また、この CSPGの発現において 、パーキンソン病患者の脳内のレビー小体(LB : LeWy body)に蓄積していると報告さ れているがその生体内機能は未だ明ら力、となっていない(非特許文献 25)。また、脳 内での CSPG増加における意義もまだわかって!/、な!/、(非特許文献 23)。 [0010] On the other hand, CSPG is an essential molecule during the embryonic period and is abundant in each organ. Neural regeneration such as neural stem cell differentiation inducer (Patent Document 1), nerve regeneration using human / bone morphogenetic protein (Patent Document 2), human, nerve regeneration using bone morphogenetic protein (Patent Document 3) It is considered as a molecule that controls the processes involved! /, While treating the damage of the central nervous system (Patent Document 4), materials and methods for promoting the repair of nerve tissue (Patent Document) It is also involved in the inhibition of nerve regeneration at various sites such as 5) and blood vessel smooth muscle cell treatment inhibitory factor (Patent Document 6). As an example, neurocan, blebican, and NG2 were first identified as chondroitin sulfate proteodaricans, which are regenerative inhibitors that are expressed when the central nervous system is damaged (Non-patent Document 24), and chondroitinase ABC ( There is a report example that degrades CSPG by administration of an enzyme that selectively removes chondroitin sulfate, which is a type of GAG chain, and promotes regeneration of the central nerve (Patent Document 4). Further, in the expression of this CSPG, Lewy body in the brains of patients with Parkinson's disease: has been reported to be accumulated in (LB Le Wy body) has become its biological function yet Akira et force, and No (Non-patent document 25). In addition, the significance of increasing CSPG in the brain is still known! /, NA! / (Non-patent Document 23).
[0011] 特許文献 1:特許公開 2005-278641  [0011] Patent Document 1: Patent Publication 2005-278641
特許文献 2:特許公開 2005-007196  Patent Document 2: Patent Publication 2005-007196
特許文献 3:特許公表平 09-501932  Patent Document 3: Patent Publication No. 09-501932
特許文献 4:特許公表 2005-526740  Patent Document 4: Patent Publication 2005-526740
特許文献 5:特許公表 2005-500375  Patent Document 5: Patent Publication 2005-500375
特許文献 6:特許公表平 08-510209  Patent Document 6: Patent Publication 08-510209
非特許文献 l : Iwata et al. J.Neurosci. (2004) 24(4) 991-998  Non-patent literature l: Iwata et al. J. Neurosci. (2004) 24 (4) 991-998
非特許文献 2 : Strittmatter, W.J et al. Proc. Natl. Acad. Sci. (1993) 90 8098-8102 非特許文献 3 : Forman et al. Nat. Med. (2004) 10(10) 1055-1063  Non-Patent Document 2: Strittmatter, W.J et al. Proc. Natl. Acad. Sci. (1993) 90 8098-8102 Non-Patent Document 3: Forman et al. Nat. Med. (2004) 10 (10) 1055-1063
非特許文献 4 : Matthew, J. F. Nature, review (2006) 7 306-318  Non-Patent Document 4: Matthew, J. F. Nature, review (2006) 7 306-318
非特許文献 5 : Gsponer, J. et al. Protein Pept Lett. (2006) 13(3) 287-293  Non-Patent Document 5: Gsponer, J. et al. Protein Pept Lett. (2006) 13 (3) 287-293
非特許文献 6 : McNaught, .S.P. et al. Neurobiol. Aging (2006) 27 530-545  Non-Patent Document 6: McNaught, .S.P. Et al. Neurobiol. Aging (2006) 27 530-545
非特許文献 7 : Hattori, N. et al. Lancet. (2004) 364(9435) 722-724 非特許文献 8: Murakami, T. et al. Ann neurol. (2004) 55(3) 439-442 非特許文献 9 :門脇寿枝 et al.実験医学(2006) 24, 10173-180 Non-Patent Document 7: Hattori, N. et al. Lancet. (2004) 364 (9435) 722-724 Non-patent literature 8: Murakami, T. et al. Ann neurol. (2004) 55 (3) 439-442 Non-patent literature 9: Hisae Kadowaki et al. Experimental medicine (2006) 24, 10173-180
非特許文献 10 : Muramatsu、 S. et al. Rinsho Shinkeigaku. (2005) 45 (11) 902-904 非特許文献 l l : Iwata. N. et al. J. Neurosci. (2004) 24(4) 991-998  Non-patent literature 10: Muramatsu, S. et al. Rinsho Shinkeigaku. (2005) 45 (11) 902-904 Non-patent literature ll: Iwata. N. et al. J. Neurosci. (2004) 24 (4) 991- 998
非特許文献 12 : Hadaczek P, et al. Hum Gene Ther. (2006)17(3) 291-302 非特許文献 13 : Lindahl, U et al. (1972) In Glycoproteins (Gottschalk, A. ed) pp. 49 Non-patent document 12: Hadaczek P, et al. Hum Gene Ther. (2006) 17 (3) 291-302 Non-patent document 13: Lindahl, U et al. (1972) In Glycoproteins (Gottschalk, A. ed) pp. 49
1—517, Elsevier, New York. 1-517, Elsevier, New York.
非特許文献 14 : Oegema, T et al. J. Biol. Chem. (1984) 259 1720-1726  Non-Patent Document 14: Oegema, T et al. J. Biol. Chem. (1984) 259 1720-1726
非特許文献 15 : Sugahara, et al. J. Biol. Chem. (1988) 263 10168-10174 非特許文献 16 : Sugahara, et al. J. Biol. Chem. (1992) 267 6027-6035  Non-patent document 15: Sugahara, et al. J. Biol. Chem. (1988) 263 10168-10174 Non-patent document 16: Sugahara, et al. J. Biol. Chem. (1992) 267 6027-6035
非特許文献 17 : De Waard et al. J. Biol. Chem. (1992) 267 6036-6043  Non-Patent Document 17: De Waard et al. J. Biol. Chem. (1992) 267 6036-6043
非特許文献 18 : Moses, J, Oldberg et al. Eur. J.Biol. (1992) 248 521-526  Non-Patent Document 18: Moses, J, Oldberg et al. Eur. J. Biol. (1992) 248 521-526
非特許文献 19 : Yamada, S et al. Trends in Glycoscience and Glycotechnology,(1998 Non-Patent Document 19: Yamada, S et al. Trends in Glycoscience and Glycotechnology, (1998
) 10, 95-123 ) 10, 95-123
非特許文献 20 : Castillo, G. M. et al. J. Neurochem. (1997) 69 2452-2465 非特許文献 21 : Cotman, S.し et al. Mol. Cell. Neurosci. 15 (2000) 183-198 非特許文献 22 : Snow, A.D. et al. Neurobiol. Aging (1989)10 481-497  Non-patent literature 20: Castillo, GM et al. J. Neurochem. (1997) 69 2452-2465 Non-patent literature 21: Cotman, S. et al. Mol. Cell. Neurosci. 15 (2000) 183-198 Non-patent Reference 22: Snow, AD et al. Neurobiol. Aging (1989) 10 481-497
非特許文献 23 : Horsen, J.V. et al. J. Alzheimers Dis. (2004) 6 469-474  Non-Patent Document 23: Horsen, J.V. et al. J. Alzheimers Dis. (2004) 6 469-474
非特許文献 24 : Rhodes KE, Fawcett JW J. Anat. (2004) 204 33-48  Non-Patent Document 24: Rhodes KE, Fawcett JW J. Anat. (2004) 204 33-48
非特許文献 25 : DeWitt, D.A. et al. Brain Res. (1994) 656 205-209  Non-Patent Document 25: DeWitt, D.A. et al. Brain Res. (1994) 656 205-209
発明の開示  Disclosure of the invention
発明が解決しょうとする課題  Problems to be solved by the invention
[0012] 本発明は、神経線維性変性抑制剤、および該薬剤を有効成分とする神経線維性 変性疾患の治療剤、並びに、神経線維性変性抑制剤のスクリ一ユング方法の提供を 課題とする。 [0012] An object of the present invention is to provide a neurofibrous degeneration inhibitor, a therapeutic agent for a neurofibrotic degenerative disease containing the drug as an active ingredient, and a screening method for the neurofibrotic degeneration inhibitor. .
課題を解決するための手段  Means for solving the problem
[0013] 本発明者らは、そのような薬剤を開発するため鋭意研究を重ねるうち、これまで神 経線維性変性疾患の病因とはされてレ、な力、つた CSPGの過剰な蓄積力 ニューロン の死やシナプスの欠損(アルツハイマー病)、ドーパミンニューロンの細胞死(パーキ ンソン病)、また運動ニューロンの変性 (筋萎縮性側索硬化症)を促進させる他にも、 多種の神経線維性変性疾患の因子に働レ、て!/、るのではな!/、かと考えた。本発明者 らは、この考えに基づ!/、て神経線維性変性疾患の一つであるパーキンソン病モデル マウスでの研究を進めた結果、 CSPGの硫酸基転移酵素のうち GalNAc4ST-l、 GalNA c4ST-2、 GALNAC4S-6STの遺伝子らを siRNAによってノックダウンする処置、または 、コンドロイチン硫酸における糖鎖を除去するコンドロイチナーゼ(コンドロイチネース ) ABCによる処置により、ドーパミンニューロンにおける CSPG過剰発現または過剰蓄 積が抑制し、グリア細胞におけるァストロサイトの役割である神経細胞機能制御が活 発化することを見出した。その他にも、 泉維芽細胞の浸潤を抑制し、ドーノ ミンニュー ロンの細胞死を防ぎドーパミン合成酵素であるチロシンハイドロキシラーゼ(TH: Tyro sine hydroxylase)の分泌を顕著にさせ、パーキンソン病の症状を治療することを見出 した。即ち、コンドロイチン硫酸プロテオダリカンの蓄積あるいは生合成を阻害するこ とによって、神経線維性変性を抑制させることができることを実証し、本発明を完成さ せた。コンドロイチン硫酸プロテオダリカンの生成もしくは蓄積を阻害する物質は、神 経線維性変性抑制剤として有用である。また、該薬剤は、神経線維性変性疾患の治 療もしくは予防のための薬剤となる。 [0013] While the present inventors have conducted intensive research to develop such a drug, it has so far been considered as the etiology of neurofibrotic degenerative diseases, and has an excessive accumulation power of CSPG neurons. In addition to promoting cell death, synaptic deficiency (Alzheimer's disease), dopamine neuron cell death (Parkinson's disease), and motor neuron degeneration (amyotrophic lateral sclerosis), various neurofibrotic degenerative diseases I thought that it would work on the factor of the day! /, Why not! / Based on this idea, the present inventors have conducted research on Parkinson's disease model mice, which are one of the neurofibrotic degenerative diseases. As a result, among the CSPG sulfotransferases, GalNAc4ST-l, GalNAc cPGST-2, GALNAC4S-6ST genes are knocked down by siRNA, or chondroitinase (chondroitinase) ABC is used to remove sugar chains in chondroitin sulfate. It was found that the product was suppressed and the neuronal function control, which is the role of astrocytes in glial cells, was activated. In addition, it suppresses infiltration of fountain fibroblasts, prevents cell death of donomin neuron, makes secretion of tyrosine hydroxylase (TH: Tyro sine hydroxylase) prominent, and treats symptoms of Parkinson's disease I found something to do. That is, it was demonstrated that neurofibrous degeneration can be suppressed by inhibiting the accumulation or biosynthesis of chondroitin sulfate proteodalycan, and completed the present invention. A substance that inhibits the production or accumulation of chondroitin sulfate proteodarican is useful as a neurofibrotic degeneration inhibitor. In addition, the drug is a drug for treatment or prevention of neurofibrotic degenerative diseases.
[0014] 上述したコンドロイチン硫酸を分解するコンドロイチナーゼ ABCと硫酸化転移酵素 のノックダウンを概念とした siRNAを用いてプロテオダリカン蓄積を制御し、脳神経線 維性変性疾患における病態変化を観察した実際の臨床治療は報告されていない。 今回、その一例としてコア蛋白質からのらせん状側鎖を伸長するグリコサミノダリカン に対して石) ¾酸化転移酵素である N— acetylgalactosamine— 4— 0-sulfotransferase:N— ace tylga ctosamine_4_0_sulrotransferae_ 1, N_acetylgalactosamine_4_0_smfotransfera se-2, N_acetylgactosamine_4_sulfate 6-O-sulfotransferase (GalNAc4ST-l, GalNAc4 ST-2, GALNAC4S-6ST)の siRNAとコンドロイチン硫酸の分解酵素であるコンドロイチ ナーゼ ABC投与に着目しその実施例を示す。  [0014] The above-mentioned chondroitinase ABC that degrades chondroitin sulfate and siRNA based on the concept of sulfating transferase knockdown were used to control proteodalycan accumulation and to observe pathological changes in neurodegenerative diseases of the brain No actual clinical treatment has been reported. As an example, N-acetylgalactosamine—4—0-sulfotransferase: N—acetylga ctosamine_4_0_sulrotransferae_1, N_acetylgalactosamine_4_0_smfotransfera se -2, N_acetylgactosamine_4_sulfate 6-O-sulfotransferase (GalNAc4ST-l, GalNAc4 ST-2, GALNAC4S-6ST) siRNA and chondroitinase ABC, which is a degrading enzyme of chondroitin sulfate, are shown in the examples.
[0015] さらに本発明者らは、 C4-sulfataseについて線維化抑制効果を検討した。その結果 、これらの物質は!/、ずれも神経線維性変異を抑制する効果を奏することが示された。 [0016] 即ち、コンドロイチン硫酸プロテオダリカンの「合成阻害作用」を有する物質である 遺伝子サイレンシング核酸(GalNAcST siRNA)と、コンドロイチン硫酸プロテオグリカ ンの「分解促進作用」を有する物質であるコンドロイチナーゼ ABC、並びに、コンドロ ィチン硫酸プロテオダリカンの「脱硫酸化作用」を有する物質である C4-sulfataseを用 いた実験によって、全く異なる性質の全く異なる経路で「コンドロイチン硫酸プロテオ ダリカンの生成または蓄積を阻害」する物質が、神経線維性変異を抑制する効果が あることが実証された。つまり、これらに共通する特徴である「コンドロイチン硫酸プロ テオダリカンの生成または蓄積を阻害」する物質は、神経線維性変性を抑制する作 用を有することが明らかとなった。 [0015] Furthermore, the present inventors examined the fibrosis inhibitory effect of C4-sulfatase. As a result, it was shown that these substances have an effect of suppressing neurofibrotic mutation! [0016] That is, a gene silencing nucleic acid (GalNAcST siRNA) which is a substance having a “synthesis inhibitory action” of chondroitin sulfate proteodarican and a chondroitinase which is a substance having a “degradation promoting action” of chondroitin sulfate proteoglycan Through experiments using ABC and C4-sulfatase, a substance that has the “desulfating action” of chondroitin sulfate proteodarican, “inhibits the production or accumulation of chondroitin sulfate proteodarican” in completely different pathways. It has been demonstrated that the substance to suppress the neurofibrotic mutation. In other words, it has been clarified that a substance that “inhibits the production or accumulation of chondroitin sulfate proteodarican”, which is a feature common to these, has an action of suppressing nerve fiber degeneration.
[0017] 本発明は、神経線維性変性抑制剤、および該薬剤を有効成分とする神経線維性 変性疾患の治療剤、並びに、神経線維性変性抑制剤のスクリーニング方法等に関し 、より具体的には、  [0017] The present invention relates to a neurofibrous degeneration inhibitor, a therapeutic agent for a neurofibrotic degenerative disease containing the drug as an active ingredient, a screening method for a neurofibrotic degeneration inhibitor, and more specifically, ,
〔1〕 コンドロイチン硫酸プロテオダリカンの生成もしくは蓄積を阻害する物質を有効 成分として含む、神経線維性変性抑制剤、  [1] a neurofibrotic degeneration inhibitor comprising a substance that inhibits the production or accumulation of chondroitin sulfate proteodarican as an active ingredient,
〔2〕 前記物質が、コンドロイチン硫酸プロテオダリカン分解促進作用を有する物質 である、〔1〕に記載の薬剤、  [2] The drug according to [1], wherein the substance is a substance having a chondroitin sulfate proteodarican degradation promoting action,
〔3〕 前記物質が、コンドロイチン硫酸プロテオダリカン合成阻害作用を有する物質 である、〔1〕に記載の薬剤、  [3] The drug according to [1], wherein the substance is a substance having a chondroitin sulfate proteodarican synthesis inhibitory action,
〔4〕 前記物質が、コンドロイチン硫酸プロテオダリカン脱硫酸化作用を有する物質 である、〔1〕に記載の薬剤、  [4] The drug according to [1], wherein the substance is a substance having a chondroitin sulfate proteodarican desulfation action,
〔5〕 前記物質が、コンドロイチン硫酸プロテオダリカン硫酸化阻害作用を有する物 質である、〔1〕に記載の薬剤、  [5] The drug according to [1], wherein the substance is a substance having an inhibitory action on chondroitin sulfate proteodarican sulfation,
〔6〕 脳においてコンドロイチン硫酸プロテオダリカンの生成もしくは蓄積が阻害され ることを特徴とするはり具体的には、脳組織におけるコンドロイチン硫酸プロテオダリ カン沈着を抑制することを特徴とする)、〔1〕〜〔5〕のいずれかに記載の薬剤、 〔7〕 神経線維性変性疾患の治療用または予防用の、〔1〕〜〔6〕の!/、ずれかに記載 の薬剤、  [6] Characteristically characterized in that the production or accumulation of chondroitin sulfate proteodarican is inhibited in the brain. Specifically, it is characterized by suppressing chondroitin sulfate proteodarican deposition in brain tissue), [1] A drug according to any one of to [5], [7] a drug for treatment or prevention of neurofibrotic degenerative disease, a drug according to [1] to [6], or any of the above,
〔8〕 前記神経線維性変性疾患が脳脊髄神経または末梢神経線維性変性疾患であ る、〔7〕に記載の薬剤、 [8] The neurofibrotic degenerative disease is cerebrospinal nerve or peripheral nerve fibrotic degenerative disease The drug according to [7],
〔9〕 前記神経線維性変性疾患がパーキンソン病、アルツハイマー病、筋萎縮性側 索硬化症、ポリグルタミン病、脊髄性筋萎縮症、ハンチントン病、または多発性硬化 症である、〔7〕に記載の薬剤、  [9] The neurofibrotic degenerative disease is Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, polyglutamine disease, spinal muscular atrophy, Huntington's disease, or multiple sclerosis. Drugs,
〔10〕 コンドロイチン硫酸プロテオダリカンの生成もしくは蓄積を阻害する物質を有 効成分とする、脳組織におけるドーパミンニューロン再生促進剤、  [10] a dopamine neuron regeneration promoter in brain tissue, comprising as an active ingredient a substance that inhibits the production or accumulation of chondroitin sulfate proteodarican;
[11 ] コンドロイチン硫酸プロテオダリカンの生成もしくは蓄積を阻害する物質を有 効成分とする、炎症細胞における脳組織破壊防止剤、  [11] An agent for inhibiting the destruction of brain tissue in inflammatory cells, comprising as an active ingredient a substance that inhibits the production or accumulation of chondroitin sulfate proteodarican,
〔12〕 被検試料から、コンドロイチン硫酸プロテオダリカンの生成もしくは蓄積を阻害 する作用を有する物質を選択することを特徴とする、神経線維性変性抑制剤のスクリ 一ユング方法、  [12] A screening method for a neurofibrotic degeneration inhibitor, comprising selecting a substance having an action of inhibiting the production or accumulation of chondroitin sulfate proteodarican from a test sample,
〔13〕 以下の(a)〜(d)の!/、ずれかに記載の作用を有する物質を選択する工程を含 む、〔12〕に記載のスクリーニング方法、  [13] The screening method according to [12], comprising a step of selecting a substance having the action described in any of the following (a) to (d)! /
(a)コンドロイチン硫酸プロテオダリカンの分解促進作用 ( a ) Promoting the degradation of chondroitin sulfate proteodarican
(b)コンドロイチン硫酸プロテオダリカンの合成阻害作用  (b) Inhibition of chondroitin sulfate proteodarican synthesis
(c)コンドロイチン硫酸プロテオダリカンの脱硫酸化作用  (c) Desulfation effect of chondroitin sulfate proteodarican
(d)コンドロイチン硫酸プロテオダリカンの硫酸化阻害作用  (d) Inhibitory effect of chondroitin sulfate proteodarican on sulfation
〔14〕 以下の工程 (a)〜(c)を含む、神経線維性変性抑制剤のスクリーニング方法、 [14] A screening method for a neurofibrotic degeneration inhibitor comprising the following steps (a) to (c):
(a)コンドロイチン硫酸プロテオダリカンまたはその一部と被検化合物を接触させるェ 程と、 ( a ) contacting the test compound with chondroitin sulfate proteodarican or a part thereof;
(b)コンドロイチン硫酸プロテオダリカンまたはその一部の存在量を測定する工程と、 (b) measuring the abundance of chondroitin sulfate proteodarican or a part thereof;
(c)被検化合物の非存在下において測定した場合と比較して、存在量を低下させる 物質を選択する工程 (c) A step of selecting a substance that reduces the abundance compared to the case where measurement is performed in the absence of the test compound.
〔15〕 前記神経線維性変性抑制剤が、神経線維性変性疾患の治療用または予防 用である、請求項 12〜; 14のいずれかに記載のスクリーニング方法、  [15] The screening method according to any one of claims 12 to 14, wherein the neurofibrotic degeneration inhibitor is for treating or preventing a neurofibrotic degenerative disease,
を、提供するものである。 Is provided.
さらに本発明は、以下に関する。  The present invention further relates to the following.
[16] 〔1〕〜〔9〕の!/、ずれかに記載の薬剤の、神経線維性変性抑制剤の製造にお ける使用。 [16] In the manufacture of a neurofibrotic degeneration inhibitor, the drug according to [1] to [9]! Use.
〔17〕 〔1〕〜〔9〕のいずれかに記載の薬剤を、個体(患者等)へ投与する工程を含 む、神経線維性変性疾患の治療方法。  [17] A method for treating a neurofibrotic degenerative disease, comprising a step of administering the drug according to any one of [1] to [9] to an individual (patient or the like).
〔18〕 コンドロイチン硫酸プロテオダリカンの生成もしくは蓄積を阻害する物質を個 体へ投与する工程を含む、神経線維性変性を抑制する方法。  [18] A method for suppressing neurofibrotic degeneration, comprising a step of administering a substance that inhibits the production or accumulation of chondroitin sulfate proteodarican to an individual.
〔19〕 コンドロイチン硫酸プロテオダリカンの生成もしくは蓄積を阻害する物質を個 体へ投与する工程を含む、グリア細胞のァストロサイトを活性化する方法。  [19] A method for activating glial astrocytes, comprising a step of administering a substance that inhibits the production or accumulation of chondroitin sulfate proteodarican to an individual.
〔20〕 コンドロイチン硫酸プロテオダリカンの生成もしくは蓄積を阻害する物質を個 体 (例えば、ドーパミンニューロンの機能が低下した患者等)へ投与する工程を含む、 脳組織におけるドーノ ミンニューロンの再生を促進する方法。  [20] Promote regeneration of donomin neurons in brain tissue, including administering to individuals (for example, patients with impaired dopamine neuron function) a substance that inhibits the production or accumulation of chondroitin sulfate proteodarican Method.
〔21〕 コンドロイチン硫酸プロテオダリカンの生成もしくは蓄積を阻害する物質を個 体へ投与する工程を含む、炎症細胞における脳組織の破壊を防止する方法 (感染 や薬剤性の脳炎または外傷等の侵襲に対して保護する方法)。  [21] A method for preventing the destruction of brain tissue in inflammatory cells, including the step of administering a substance that inhibits the production or accumulation of chondroitin sulfate proteodarican (to prevent invasion such as infection or drug-induced encephalitis or trauma) How to protect against).
〔22〕 〔1〕〜〔; 11〕のいずれかに記載の薬剤および薬学的に許容された担体を含ん でなる組成物。  [22] A composition comprising the drug according to any one of [1] to [; 11] and a pharmaceutically acceptable carrier.
発明の効果  The invention's effect
[0019] 本発明によって、神経線維性変性の発症にコンドロイチン硫酸プロテオダリカンの 生成や蓄積が関係していることが明らかになった。コンドロイチン硫酸プロテオグリカ ンの生成や蓄積を阻害することにより神経線維性変性が抑制されることが示された。 これまでにない新しいコンセプトの神経線維性変性疾患の治療薬が提供できることに なる。特に神経線維性変性は現代社会で患者数が増大している、パーキンソン病、 アルツハイマー病などと密接に関連しており、新しいコンセプトの治療薬剤は医療上 また産業上も重要な意義を持つ。  [0019] According to the present invention, it has been clarified that the production and accumulation of chondroitin sulfate proteodalycan is related to the onset of neurofibrotic degeneration. Inhibition of chondroitin sulfate proteoglycan production and accumulation has been shown to suppress neurofibrotic degeneration. It will be possible to provide a therapeutic agent for neurofibrotic degenerative diseases with a new concept that has never existed before. In particular, neurofibrotic degeneration is closely related to Parkinson's disease, Alzheimer's disease, etc., where the number of patients is increasing in modern society, and a new concept of therapeutic drugs has important medical and industrial significance.
図面の簡単な説明  Brief Description of Drawings
[0020] [図 l] l-methyl-4-pheny卜 1, 2, 3, 6 tetrahydropyridine (MPTP)によって誘導された パーキンソン病(Parkinson's disease)モデルマウスにおける未治療群、コンドロイチ ナーゼ ABC、 GalNAcST siRNA処置群における 8日目(最終日 )の RT-PCRによる β—a ctin、 GalNAc4ST- lと GALNAC4S-6STの発現を検討した写真である。 [図 2]MPTPによって誘導されたパーキンソン病モデルマウスにおける未治療群、 Gal NAcST siRNA処置群とコンドロイチナーゼ ABC処置群におけるコンドロイチン硫酸プ 口テオダリカン (CSPG)の脳内における沈着について示した写真である。 [0020] [Fig. L] Untreated group, chondroitinase ABC, GalNAcST siRNA treatment in Parkinson's disease model mice induced by l-methyl-4-pheny 卜 1, 2, 3, 6 tetrahydropyridine (MPTP) This is a photograph examining the expression of β-actin, GalNAc4ST-1 and GALNAC4S-6ST by RT-PCR on the 8th day (final day) in the group. [Fig. 2] Photograph showing the deposition of chondroitin sulfate teodarican (CSPG) in MPTP-induced Parkinson's disease model mice in the untreated group, Gal NAcST siRNA-treated group and chondroitinase ABC-treated group .
[図 3]MPTPによって誘導されたパーキンソン病モデルマウスでの未治療群、 GalNAcS T siRNA処置群とコンドロイチナーゼ ABC処置群における F4/80陽性炎症性マクロフ ァージの浸潤について示した写真である。 FIG. 3 is a photograph showing infiltration of F4 / 80-positive inflammatory macrophage in an untreated group, a GalNAcST siRNA-treated group and a chondroitinase ABC-treated group in Parkinson's disease model mice induced by MPTP.
[図 4]MPTPによって誘導されたパーキンソン病モデルマウスにおける未治療群、 Gal NAcST siRNA処置群とコンドロイチナーゼ ABC処置群における脳内の線維芽細胞に ついて示した写真である。  FIG. 4 is a photograph showing fibroblasts in the brain in an untreated group, a Gal NAcST siRNA-treated group and a chondroitinase ABC-treated group in a Parkinson's disease model mouse induced by MPTP.
[図 5]MPTPによって誘導されたパーキンソン病モデルマウスでの未治療群、 GalNAcS T siRNA処置群とコンドロイチナーゼ ABC処置群においてグリア細胞のァストロサイト について示した写真である。  FIG. 5 is a photograph showing glial cell astrocytes in an untreated group, a GalNAcST siRNA-treated group and a chondroitinase ABC-treated group in Parkinson's disease model mice induced by MPTP.
[図 6]MPTPによって誘導されたパーキンソン病モデルマウスでの未治療群、 GalNAcS T siRNA処置群とコンドロイチナーゼ ABC処置群におけるチロシンハイドロキシラーゼ (TH: Tyrosine hydroxylase)の分泌につ!/、て示した写真である。  [Fig.6] Tyrosine hydroxylase (TH) secretion in MPTP-induced Parkinson's disease model mice in the untreated group, GalNAcS T siRNA-treated group and chondroitinase ABC-treated group! It is a photograph.
園 7]脳組織における CSPGの発現を示す写真である。脳組織中における CSPGの発 現動態につ!/、て酵素抗体免疫染色法を用いて解析を行った結果を示す。一次抗体 は CS_56 (生化学工業社製)、マウススティンキットを用いて染色を行った。写真は、 中脳の歯状回付近の様子を示してレ、る。 [Sono 7] This is a photograph showing the expression of CSPG in brain tissue. The results of CSPG expression in brain tissue are analyzed using enzyme antibody immunostaining. The primary antibody was stained using CS_56 (manufactured by Seikagaku Corporation) and a mouse stin kit. The picture shows the midbrain near the dentate gyrus.
園 8]Real-time PCR法による炎症及び線維化関連遺伝子発現解析の結果を示す図 である。線維化マーカーである TGF- /3、マクロファージ浸潤に伴う炎症の指標である TNF- aの発現について Rea卜 time PCRにて検討した結果を示す。グラフは TNF_ aと TGF- βを house keeping Gene ( β -actin)における相対比を示している。 Fig. 8] shows the results of analysis of inflammation and fibrosis-related gene expression by Real-time PCR. The result of examining the expression of TGF- / 3, which is a fibrosis marker, and TNF-a, which is an index of inflammation associated with macrophage infiltration, is shown by rea time PCR. The graph shows the relative ratio of TNF_a and TGF-β in the house keeping gene (β-actin).
[図 9]Real_time PCR法による Nurrl遺伝子発現解析の結果を示す図である。脳組織 中における Nurrlの遺伝子発現について、 SYBR premix kit (タカラバイオ社製)と Real -time PCR thermal cycler DICE (タカラバイオ社製)を用いて行った。グラフは Nurrl と house keeping Gene ( /3—actin)との申目メ寸匕を示してレヽる。 FIG. 9 shows the results of Nurrl gene expression analysis by Real_time PCR method. Nurrl gene expression in brain tissue was performed using SYBR premix kit (Takara Bio) and Real-time PCR thermal cycler DICE (Takara Bio). The graph shows the dimensions of Nurrl and house keeping Gene (/ 3-actin).
園 10]脳組織における DA neuronの発現動態の解析結果を示す写真である。脳組織 中におけるドーパミンニューロン (DA neuron)の発現動態について蛍光免疫染色法 を用いて解析を行った結果を示す。一次抗体は抗 TH抗体(Carbiochem社製)、二次 抗体は Alexa-488標識抗ゥサギ IgG抗体(Invitrogen社製)を用いて行った。 [Sono 10] These are photographs showing the analysis results of DA neuron expression dynamics in brain tissue. Brain tissue The results of an analysis of the expression dynamics of dopamine neurons (DA neuron) using fluorescent immunostaining are shown. The primary antibody was an anti-TH antibody (manufactured by Carbiochem), and the secondary antibody was an Alexa-488 labeled anti-rabbit IgG antibody (manufactured by Invitrogen).
[図 11]治療群、未治療群との生存率の比較を示す図である。各群(治療群、未治療 群)の生存率について時間経過ごとに比較した。解析は力プランマイヤーの生存曲 線を用いて、 day 7 (7日目)をエンドポイントとして行った。治療群として、 C4_sulfatase と GalNAc4S_6ST siRNAにて治療した。  FIG. 11 is a diagram showing a comparison of survival rates between a treated group and an untreated group. The survival rate of each group (treated group, untreated group) was compared over time. The analysis was carried out using day 7 (day 7) as an endpoint, using the force planmeyer survival curve. The treatment group was treated with C4_sulfatase and GalNAc4S_6ST siRNA.
発明を実施するための最良の形態  BEST MODE FOR CARRYING OUT THE INVENTION
[0021] 以下、本発明を詳細に説明する。  Hereinafter, the present invention will be described in detail.
代表的な神経線維性変性疾患の一つであるパーキンソン病に伴う病態として、脳 の神経細胞におけるマクロファージ · ί泉維芽細胞などの浸潤などによる泉維化などの 変性状態がある。本発明者らは、脳の神経細胞における変性状態の改善をパーキン ソン病治療の有効な方法の一つとするため、コンドロイチン硫酸プロテオダリカンの機 能に着目した。そして、コンドロイチン硫酸プロテオダリカンの蓄積を抑制する状態を パーキンソン病モデルマウスにおいて作出し、詳細に解析したところ、野生型の脳の 神経細胞に比してコンドロイチン硫酸プロテオダリカンの蓄積が改善している細胞が 多数観察されるとともに、炎症状態などが改善された。即ち、コンドロイチン硫酸プロ テオダリカンの生成もしくは蓄積を阻害すると、パーキンソン病に深く関与している脳 の神経細胞におけるコンドロイチン硫酸プロテオダリカンの異常蓄積状態の改善が 促進され、神経線維性変性の改善につながることを見出した。  As a pathological condition associated with Parkinson's disease, which is one of the typical neurofibrotic degenerative diseases, there is a degenerative condition such as fomentation due to infiltration of macrophages, lysen fibroblasts, etc. in brain neurons. The present inventors have focused on the function of chondroitin sulfate proteodarican in order to improve the degenerative state of neurons in the brain as an effective method for treating Parkinson's disease. A state in which the accumulation of chondroitin sulfate proteodarican was suppressed in Parkinson's disease model mice was analyzed in detail, and it was found that the accumulation of chondroitin sulfate proteodarican was improved compared to neurons in the wild-type brain. Many cells were observed and the inflammatory condition was improved. In other words, inhibiting the production or accumulation of chondroitin sulfate proteodarican promotes improvement of abnormal accumulation state of chondroitin sulfate proteodarican in brain neurons that are deeply involved in Parkinson's disease, leading to improvement of neurofibrotic degeneration. I found out.
[0022] 本発明は、コンドロイチン硫酸プロテオダリカンの生成もしくは蓄積を阻害する物質 を有効成分として含む、神経線維性変性抑制剤に関する。  [0022] The present invention relates to a neurofibrotic degeneration inhibitor comprising, as an active ingredient, a substance that inhibits the production or accumulation of chondroitin sulfate proteodalycan.
[0023] 本発明の「コンドロイチン硫酸プロテオダリカン」は、プロテオダリカンの一つであり、 代表的な硫酸化ムコ多糖であるコンドロイチン硫酸/デルマタン硫酸とタンパク質(コ ァタンパク質)との共有結合化合物の総称である。本発明における「コンドロイチン硫 酸プロテオダリカン」は、ヒトのコンドロイチン硫酸プロテオダリカンであることが好まし いが、その由来する生物種は特に制限されず、ヒト以外の生物におけるコンドロイチ ン硫酸プロテオダリカンと同等なタンパク質(ホモログ.オルソログ等)も本発明におけ るる「「ココンンドドロロイイチチンン硫硫酸酸ププロロテテオオダダリリカカンン」」にに含含ままれれるる。。例例ええばば、、ヒヒトトココンンドドロロイイチチンン硫硫酸酸 ププロロテテオオダダリリカカンンにに相相当当すするるタタンンパパクク質質をを有有しし、、かかつつ、、ヒヒトトののココンンドドロロイイチチンン硫硫酸酸ププロロテテ ォォググリリカカンンとと同同等等ななタタンンパパクク質質をを有有すするる生生物物ででああれればば、、本本発発明明をを実実施施すするるここととはは可可能能 ででああるる。。ままたた本本発発明明ににおおけけるるココンンドドロロイイチチンン硫硫酸酸ププロロテテオオダダリリカカンンににはは、、炎炎症症ななどどでで一一 時時的的ににググリリココササミミノノダダリリカカンン((GGAAGG))鎖鎖がが結結合合ししププロロテテオオダダリリカカンンににななるるもものの、、いいわわゆゆるる
Figure imgf000014_0001
[0023] The "chondroitin sulfate proteodarican" of the present invention is one of the proteodaricans, and is a covalently bonded compound of chondroitin sulfate / dermatan sulfate, which is a typical sulfated mucopolysaccharide, and a protein (coprotein). It is a general term. The “chondroitin sulfate proteodarican” in the present invention is preferably a human chondroitin sulfate proteodarican, but the species from which it is derived is not particularly limited. Proteins equivalent to kan (homologs, orthologs, etc.) are also used in the present invention. It is included in "" Konkondoroloyichitin sulphate sulfate proteoteo daridarikankan "". . For example, for example, it has a tantalum protein that is equivalent to the human proteotheododalyricacan. As long as it is a living organism that has the same quality as that of coconut doroleutitin sulphate sulfate, it can be used as an inventor of this invention. This is the possibility of actually implementing this. . In addition, in the present invention, the coconut doroleutintin sulfate sulfate sulfate proteoteo odalyrikankan is temporarily affected by inflammatory inflammation and the like. Although the chain of guriglicosasamininodararirikakan ((GGAAGG)) binds to and becomes a proprotetheodaridarikankan, it is so loose.
Figure imgf000014_0001
[[00002244]] 以以下下のの記記載載ににおお!い/、てて、、ココンンドドロロイイチチンン硫硫酸酸プロテオグリカンとして、 aaggggrniccaann、^ vveerrssiiccaa n、 neurocan、 brevican、 β glycan、 Decorin、 Biglycan、 Pibromodulin^ PG- Lbを例不" 5 る。本発明におけるコンドロイチン硫酸プロテオダリカンはこれらに限られず、コンドロ ィチン硫酸プロテオダリカンとしての活性を持つ物質であればよ!/、。ここでコンドロイ チン硫酸プロテオダリカンの活性とは、例えば細胞接着能、または細胞増殖促進など 力 S挙げられる。当業者は次のような方法でコンドロイチン硫酸プロテオダリカンとして の活性を評価することができる。コンドロイチン硫酸プロテオダリカンのアミノ酸配列の 一部の領域を含むタンパク質、または一部の領域と高い相同性(通常 70%以上、好 ましくは 80%以上、より好ましくは 90%以上、最も好ましくは 95%以上)を有するタンパ ク質の存在下で腫瘍細胞(例えば Caco_2、 HT-29細胞など)の分裂増殖を測定する 。分裂増殖を促進する効果を持つタンパク質をコンドロイチン硫酸プロテオダリカン活 性を有するタンパク質として判定できる(Int J Exp Pathol. 2005 Aug;86(4):219_29お よび Histochem Cell Biol. 2005 Aug; 124(2): 139-49)。ここで高い相同性とは、 50%以 上、好ましくは 70%以上、さらに好ましくは 80%以上、より好ましくは 90%以上(例えば 、 95%以上、さらには 96%、 97%、 98%または 99%以上)の相同性を意味する。この 相同性は、 mBLASTアルゴリズム(Altschul et al. (1990) Proc. Natl. Acad. Sci. USA 8 7: 2264-8; arlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90: 5873-7)によ つて決定することができる。  [[00002244]] The following descriptions are as follows: As a conchondroleutintintin sulfate sulfate proteoglycan, aaggggrniccaann, ^ vveerrssiiccan, neurocan, brevican, β glycan, Decorin , Biglycan, Pibromodulin ^ PG-Lb ”5, etc. The chondroitin sulfate proteodarican in the present invention is not limited to these, and any substance having activity as a chondroitin sulfate proteodarican! /, Here The activity of chondroitin sulfate proteodarican includes, for example, cell adhesion ability or cell growth promotion force S. Those skilled in the art can evaluate the activity of chondroitin sulfate proteodarican as follows. A protein that includes a partial region of the amino acid sequence of chondroitin sulfate proteodarican, or a high homology with a partial region (typically 70% or more The proliferation of tumor cells (eg, Caco_2, HT-29 cells, etc.) is measured in the presence of a protein having preferably 80% or more, more preferably 90% or more, most preferably 95% or more. Proteins that have the effect of promoting mitotic proliferation can be identified as proteins with chondroitin sulfate proteodarican activity (Int J Exp Pathol. 2005 Aug; 86 (4): 219_29 and Histochem Cell Biol. 2005 Aug; 124 (2 ): 139-49) Here, high homology means 50% or more, preferably 70% or more, more preferably 80% or more, more preferably 90% or more (for example, 95% or more, further 96% , 97%, 98%, or 99% or higher), which is the mBLAST algorithm (Altschul et al. (1990) Proc. Natl. Acad. Sci. USA 8 7: 2264-8; arlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90: 5873-7).
[0025] 本発明における「神経線維性変性」とは、神経組織における異常状態を指す。例え ば、 泉維化を生じている状態、炎症を生じている状態、 泉維芽細胞'炎症細胞などが 浸潤している状態、神経組織中の特定の細胞種の脱落 ·細胞死を起こしている状態 などが挙げられる力 S、これらには限られない。 [0026] 本発明におけるコンドロイチン硫酸プロテオダリカンの「生成もしくは蓄積を阻害す る」とは、例えば、コンドロイチン硫酸プロテオダリカンの「分解促進」、「合成阻害」、「 脱硫酸化」、「硫酸化阻害」などが挙げられる力 これらには限らず、コンドロイチン硫 酸プロテオダリカンの存在量、機能または活性が比較対象よりも低下または消失させ ることをいう。本発明において、コンドロイチン硫酸プロテオダリカンの「生成もしくは蓄 積を阻害する物質」とは、特に制限されないが、好ましくはコンドロイチン硫酸プロテ ォグリカンの「分解促進作用を有する物質」、「合成阻害作用を有する物質」、「脱硫 酸化作用を有する物質」、または「硫酸化阻害作用を有する物質」である。 [0025] "Neural fibrotic degeneration" in the present invention refers to an abnormal state in nerve tissue. For example, the state of causing fountain fibrosis, the state of inflammation, the state of infiltrating fountain fibroblasts' inflammatory cells, the loss of specific cell types in nerve tissue, and cell death The power S that can be mentioned is not limited to these. In the present invention, “inhibiting production or accumulation” of chondroitin sulfate proteodarican means, for example, “degradation promotion”, “synthesis inhibition”, “desulfation”, “sulfation” of chondroitin sulfate proteodarican. Powers that include “inhibition” and the like are not limited to these, and it means that the abundance, function, or activity of chondroitin sulfate proteodarican is reduced or eliminated as compared with the comparison target. In the present invention, the “substance that inhibits the production or accumulation” of chondroitin sulfate proteodarican is not particularly limited, but preferably the “substance that has an activity of promoting degradation of chondroitin sulfate proteoglycan” and “the substance has an inhibitory effect on synthesis”. “Substance”, “Substance with desulfurization and oxidation”, or “Substance with sulfation-inhibiting action”.
[0027] コンドロイチン硫酸プロテオダリカンの「分解促進」とは、たとえばコンドロイチン硫酸 プロテオダリカンのコアとなるタンパク質の発現の阻害 ·存在の減少が挙げられる。こ こで「コンドロイチン硫酸プロテオグリカンのコアとなるタンパク質」とは、例えば、 matri X typeコンドロイテン硫酸プロァスクリカンで、あれは、 aggrican、 versican、 neurocan、 b revicanなどのコアタンパク質が挙げられる。また膜型コンドロイチン硫酸プロテオダリ カンであれば、例えば /3 glycan、 Decorin、 Biglycan, Fibromodulin, PG_Lbなどのコア タンパク質が挙げられる。これらはいずれも例示であり、これらに限らず広くコンドロイ チン硫酸プロテオダリカンのコアとなるタンパク質であればよい。 “Degradation promotion” of chondroitin sulfate proteodarican includes, for example, inhibition of expression / reduction of the protein that is the core of chondroitin sulfate proteodarican. Here, the “protein that is the core of chondroitin sulfate proteoglycan” is, for example, matri X type chondroitene sulfate prosuclicans, and examples thereof include core proteins such as aggrican, versican, neurocan, and b revican. In the case of membrane-type chondroitin sulfate proteodlicans, for example, core proteins such as / 3 glycan, Decorin, Biglycan, Fibromodulin, and PG_Lb can be mentioned. These are only examples, and are not limited to these, and may be any protein that is widely used as the core of chondroitin sulfate proteodalycan.
[0028] 「発現」とは遺伝子からの「転写」ある!/、はポリペプチドへの「翻訳」及びタンパク質 の「分解抑制」によるもの力含まれる。「コンドロイチン硫酸プロテオダリカンのコアとな るタンパク質の発現」とは、コンドロイチン硫酸プロテオダリカンのコアとなるタンパク質 をコードする遺伝子の転写および翻訳が生じること、またはこれらの転写 ·翻訳により コンドロイチン硫酸プロテオダリカンのコアとなるタンパク質が生成されることを意味す る。また、「コンドロイチン硫酸プロテオダリカンのコアとなるタンパク質の機能」とは、 例えば、該タンパク質がコンドロイチン硫酸と結合する機能や、その他の細胞中の構 成要素との結合等を挙げることができる。上述の各種機能は、当業者においては、一 般的な技術を用いて、適宜、評価 (測定)することが可能である。具体的には、後述の 実施例に記載の方法、あるいは該方法を適宜改変して実施することができる。 [0028] "Expression" means "transcription" from a gene! /, And includes "translation" into a polypeptide and "degradation inhibition" of a protein. “Expression of the protein that is the core of chondroitin sulfate proteodarican” refers to the transcription and translation of the gene that encodes the protein that is the core of chondroitin sulfate proteodarican, or the chondroitin sulfate proteo This means that the protein that forms the core of Darican is produced. In addition, “the function of the protein serving as the core of chondroitin sulfate proteodarican” includes, for example, the function of the protein binding to chondroitin sulfate and the binding to other components in the cell. The various functions described above can be appropriately evaluated (measured) by those skilled in the art using common techniques. Specifically, the methods described in the examples described later, or the methods can be appropriately modified and carried out.
[0029] さらにまた、コンドロイチン硫酸プロテオダリカンの「分解促進」は、コンドロイチン硫 酸プロテオダリカンを切断あるいは分解する酵素またはこれらに関連する酵素の発現 の上昇であってもよい。これらの酵素の例としては、メタ口プロティナーゼ(例えば AD AMTS-1 , ADAMTS-4, ADAMTS-5など)ゃコンドロイチナーゼ、 Calpain Iなどが挙げ られる力 S、これらには限られない。また「分解促進」は、これらの酵素または酵素の一 部の投与により生ずる、コンドロイチン硫酸プロテオダリカンの存在量の減少であって あよい。 [0029] Furthermore, “degradation promotion” of chondroitin sulfate proteodarican is the expression of an enzyme that cleaves or degrades chondroitin sulfate proteodarican or an enzyme related thereto. May be a rise. Examples of these enzymes include, but are not limited to, meta-mouth proteinases (eg, AD AMTS-1, ADAMTS-4, ADAMTS-5, etc.) chondroitinase, Calpain I, and the like. Further, “degradation promotion” may be a decrease in the abundance of chondroitin sulfate proteodarican caused by administration of these enzymes or a part of the enzymes.
[0030] また「分解促進」は、コンドロイチン硫酸プロテオダリカンの発現の抑制を促す物質 の投与により生じるものであってもよい。これらの物質には例えば n-butylate、 Diethyl carbamazepine、 i'umcamycin、 non— steroidal estrogen^ し yclofeml deiphenoレぶど げられる力 S、これらには限られない。  [0030] "Degradation promotion" may be caused by administration of a substance that promotes suppression of chondroitin sulfate proteodarican expression. These substances include, but are not limited to, n-butylate, Diethyl carbamazepine, i'umcamycin, non-steroidal estrogen ^ and yclofeml deipheno.
[0031] 「分解促進作用を有する物質」の好ましい態様としては、例えば以下の(a)〜(c)か らなる群より選択される化合物(核酸)を挙げることができる。  [0031] Preferable embodiments of the "substance having a decomposition promoting action" include, for example, a compound (nucleic acid) selected from the group consisting of the following (a) to (c).
(a)コンドロイチン硫酸プロテオダリカンのコアタンパク質をコードする遺伝子の転写 産物またはその一部に対するアンチセンス核酸 ( a ) An antisense nucleic acid for a transcription product of a gene encoding the core protein of chondroitin sulfate proteodarican or a part thereof
(b)コンドロイチン硫酸プロテオダリカンのコアタンパク質をコードする遺伝子の転写 産物を特異的に開裂するリボザィム活性を有する核酸  (b) a nucleic acid having a ribozyme activity that specifically cleaves the transcription product of the gene encoding the core protein of chondroitin sulfate proteodarican
(c)コンドロイチン硫酸プロテオダリカンのコアタンパク質をコードする遺伝子の発現 を RNAi効果により阻害する作用を有する核酸  (c) Nucleic acid that acts to inhibit the expression of the gene encoding the core protein of chondroitin sulfate proteodarican by the RNAi effect
[0032] また「分解促進作用を有する物質」としては例えば以下の(a)〜(c)力、らなる群より 選択される化合物を挙げることができる。  In addition, examples of the “substance having a decomposition promoting action” include compounds selected from the following groups (a) to (c):
(a)コンドロイチン硫酸プロテオダリカンのコアタンパク質と結合する抗体 ( a ) an antibody that binds to the core protein of chondroitin sulfate proteodarican
(b)コンドロイチン硫酸プロテオダリカンのコアタンパク質に対してドミナントネガティブ の性質を有するコンドロイチン硫酸プロテオダリカン変異体  (b) Chondroitin sulfate proteodarican mutants having dominant negative properties for the core protein of chondroitin sulfate proteodarican
(c)コンドロイチン硫酸プロテオダリカンのコアタンパク質と結合する低分子化合物  (c) Low molecular weight compound that binds to the core protein of chondroitin sulfate proteodalycan
[0033] コンドロイチン硫酸プロテオダリカンの「合成阻害」とは、たとえばグリコサミノダリカン の生合成の阻害、コンドロイチン硫酸プロテオダリカン合成に関わる酵素の阻害など が挙げられるが、必ずしもこれらに限らず、コンドロイチン硫酸プロテオダリカンが合 成される過程のレ、ずれかを阻害することを指す。 [0033] “Synthetic inhibition” of chondroitin sulfate proteodarican includes, for example, inhibition of glycosaminodarlican biosynthesis, inhibition of enzymes involved in chondroitin sulfate proteodarican synthesis, but are not necessarily limited to these. This refers to inhibiting the process of chondroitin sulfate proteodarican synthesis.
[0034] コンドロイチン硫酸プロテオダリカンの合成を阻害する物質として、グリコサミノグリカ ンの生合成を阻害する物質としては、たとえば、 /3 -D-xyloside, 2-deoxy-D-glucose (2-DGノ、 ethane- l-hydroxy-l, l-diphosphonate (b TDP)、 5-hexyl-2-deoxyundine HUdR)などが挙げられる。これらをはじめとした物質によりグリコサミノダリカンの生合 成が阻害され、コンドロイチン硫酸プロテオダリカンの合成が阻害される。 [0034] As a substance that inhibits the synthesis of chondroitin sulfate proteodarican, glycosaminoglyca For example, / 3 -D-xyloside, 2-deoxy-D-glucose (2-DG, ethane-hydroxy-l, l-diphosphonate (b TDP), 5 -hexyl-2-deoxyundine HUdR). These and other substances inhibit the biosynthesis of glycosaminodarlicans and inhibit the synthesis of chondroitin sulfate proteodaricans.
[0035] 一方、コンドロイチン合成に関わる酵素としては、例えば、 GalNAc4ST-l、 GalNAc4 ST- 2、 GALNAC4S_6ST、 UA20ST、 GalT_I、 GalT_H、 GlcAT_I、 XylosylTなどが挙げ られる。これらをはじめとした酵素を阻害、発現の抑制等を行うことにより、コンドロイ チン硫酸プロテオダリカンの合成が阻害される。  On the other hand, examples of enzymes involved in chondroitin synthesis include GalNAc4ST-1, GalNAc4 ST-2, GALNAC4S_6ST, UA20ST, GalT_I, GalT_H, GlcAT_I, XylosylT, and the like. By inhibiting these and other enzymes and suppressing their expression, the synthesis of chondroitin sulfate proteodalycan is inhibited.
[0036] 「合成阻害作用を有する物質」の好ましい態様としては、例えば以下の(a)〜(c)か らなる群より選択される化合物(核酸)を挙げることができる。  [0036] Preferable embodiments of the "substance having a synthesis inhibitory action" include, for example, compounds (nucleic acids) selected from the group consisting of the following (a) to (c).
(a)コンドロイチン硫酸プロテオダリカン合成酵素をコードする遺伝子の転写産物また はその一部に対するアンチセンス核酸 ( a ) An antisense nucleic acid for a transcript or a part of a gene encoding chondroitin sulfate proteodarican synthase
(b)コンドロイチン硫酸プロテオダリカン合成酵素をコードする遺伝子の転写産物を 特異的に開裂するリボザィム活性を有する核酸  (b) Nucleic acid with ribozyme activity that specifically cleaves the transcript of the gene encoding chondroitin sulfate proteodarican synthase
(c)コンドロイチン硫酸プロテオダリカン合成酵素をコードする遺伝子の発現を RNAi 効果により阻害する作用を有する核酸  (c) a nucleic acid having an action of inhibiting the expression of a gene encoding chondroitin sulfate proteodarican synthase by the RNAi effect
[0037] また「合成阻害作用を有する物質」としては例えば以下の(a)〜(c)力、らなる群より 選択される化合物を挙げることができる。  [0037] In addition, examples of the "substance having a synthesis inhibitory action" include compounds selected from the following groups (a) to (c):
(a)コンドロイチン硫酸プロテオダリカン合成酵素と結合する抗体 ( a ) an antibody that binds to chondroitin sulfate proteodalycan synthase
(b)コンドロイチン硫酸プロテオダリカン合成酵素に対してドミナントネガティブの性質 を有するコンドロイチン硫酸プロテオダリカン合成酵素変異体  (b) Chondroitin sulfate proteodarican synthase mutant having dominant negative properties for chondroitin sulfate proteodarican synthase
(c)コンドロイチン硫酸プロテオダリカン合成酵素と結合する低分子化合物  (c) Low molecular weight compound that binds to chondroitin sulfate proteodarican synthase
[0038] コンドロイチン硫酸プロテオダリカンの「脱硫酸化」とは、コンドロイチン硫酸プロテオ ダリカン中の硫酸基が除かれることを指し、例えば内在性あるいは外部から投与され る脱硫酸化酵素による脱硫酸化、または硫酸化を抑制する化合物による硫酸化の抑 制などが挙げられる力 これらに限られず、硫酸基が除去される過程を指す。  [0038] “Desulfation” of chondroitin sulfate proteodarican refers to removal of sulfate groups in chondroitin sulfate proteodarican, for example, desulfation by endogenous or externally administered desulfating enzymes, or sulfation. This includes, but is not limited to, the process by which sulfate groups are removed.
[0039] 脱硫酸化酵素としては、例えば、 Chondroitin-4-sulfatase , Chondroitin-6-sulfatase 力 S挙げられる。また、硫酸化を抑制する化合物としては、たとえば Chlorate、 EGF rece ptor antagonistなどが挙げられる。 [0039] Examples of the desulfating enzyme include Chondroitin-4-sulfatase, Chondroitin-6-sulfatase force S. Examples of compounds that inhibit sulfation include Chlorate and EGF rece. ptor antagonist and the like.
[0040] 「脱硫酸化作用を有する物質」の好ましい態様としては、例えば以下の(a)〜(c)か らなる群より選択される化合物(核酸)を挙げることができる。 [0040] Preferable embodiments of the "substance having desulfating action" include, for example, a compound (nucleic acid) selected from the group consisting of the following (a) to (c).
(a)コンドロイチン硫酸プロテオダリカン脱硫酸化酵素抑制タンパク質をコードする遺 伝子の転写産物またはその一部に対するアンチセンス核酸 ( a ) Antisense nucleic acid for a transcript of a gene encoding a chondroitin sulfate proteodarican desulfase inhibitor protein or a part thereof
(b)コンドロイチン硫酸プロテオダリカン脱硫酸化酵素抑制タンパク質をコードする遺 伝子の転写産物を特異的に開裂するリボザィム活性を有する核酸  (b) a nucleic acid having a ribozyme activity that specifically cleaves a transcript of a gene encoding a chondroitin sulfate proteodarican desulfase inhibitor protein
(c)コンドロイチン硫酸プロテオダリカン脱硫酸化酵素抑制タンパク質をコードする遺 伝子の発現を RNAi効果により阻害する作用を有する核酸  (c) a nucleic acid having an action of inhibiting the expression of a gene encoding a chondroitin sulfate proteodarican desulfase inhibitor protein by the RNAi effect
[0041] また「脱硫酸化作用を有する物質」としては例えば以下の(a)〜(c)力、らなる群より 選択される化合物を挙げることができる。 [0041] Examples of the "substance having desulfating action" include compounds selected from the following groups (a) to (c):
(a)コンドロイチン硫酸プロテオダリカン脱硫酸化酵素抑制化合物と結合する抗体( a ) an antibody that binds to a chondroitin sulfate proteodarican desulfase inhibitor compound
(b)コンドロイチン硫酸プロテオダリカン脱硫酸化酵素抑制タンパク質に対して、ドミ ナントネガティブの性質を有するコンドロイチン硫酸プロテオダリカン脱硫酸化抑制タ ンパク質変異体 (b) Chondroitin sulfate proteodarican desulfating enzyme inhibitory protein against chondroitin sulfate proteodarican desulfating enzyme inhibitory protein
(c)コンドロイチン硫酸プロテオダリカン脱硫酸化酵素抑制化合物と結合する低分子 化合物  (c) a small molecule compound that binds to a chondroitin sulfate proteodarican desulfase inhibitor compound
[0042] ここで「脱硫酸化抑制化合物」は、タンパク質には限られず、例えば補酵素など非タ ンパク質化合物を含む。  Here, the “desulfation-inhibiting compound” is not limited to a protein, and includes non-protein compounds such as coenzymes, for example.
[0043] コンドロイチン硫酸プロテオダリカンの「硫酸化阻害作用」とは、例えば、硫酸基転 移酵素の阻害が挙げられる力 S、これに限らず、コンドロイチン硫酸プロテオダリカンが 合成される過程に生じる硫酸化が阻害されることを指す。 [0043] The “sulfation inhibitory action” of chondroitin sulfate proteodarican is, for example, the force S including inhibition of sulfate group transfer enzyme, but is not limited to this, and occurs in the process of chondroitin sulfate proteodarican synthesis. It refers to inhibition of sulfation.
[0044] 硫酸基転移酵素としては、例えば、 C4ST- 1 (Chondroitin D-N-acetylgalactosamine[0044] Examples of the sulfotransferase include C4ST-1 (Chondroitin D-N-acetylgalactosamine).
-4-O-sulfotransrerase 1)、 u4S ι ~2 (Cnondroitin D-N-acetylgalactosamine-4-O-sulf otransferase 2)、 C4ST-J (Chondroitin D-N-acetylgalactosamine-4-O-sulfotransfera se 3)、 D4ST、 C6ST_ 1、 C6ST-2などが挙げられる。 -4-O-sulfotransrerase 1), u4S ι ~ 2 (Cnondroitin DN-acetylgalactosamine-4-O-sulfotransferase 2), C4ST-J (Chondroitin DN-acetylgalactosamine-4-O-sulfotransfera se 3), D4ST, C6ST_ 1 And C6ST-2.
[0045] 「硫酸化阻害作用を有する物質」の好ましい態様としては、例えば以下の(a)〜(c) からなる群より選択される化合物(核酸)を挙げること力 Sできる。 (a)コンドロイチン硫酸プロテオダリカン硫酸基転移酵素をコードする遺伝子の転写 産物またはその一部に対するアンチセンス核酸 [0045] As a preferred embodiment of the "substance having sulfation inhibitory action", for example, a compound (nucleic acid) selected from the group consisting of the following (a) to (c) can be mentioned. ( a ) An antisense nucleic acid for a transcript or a part of a gene encoding chondroitin sulfate proteodalycan sulfate transferase
(b)コンドロイチン硫酸プロテオダリカン硫酸基転移酵素をコードする遺伝子の転写 産物を特異的に開裂するリボザィム活性を有する核酸  (b) A nucleic acid having a ribozyme activity that specifically cleaves a transcript of a gene encoding chondroitin sulfate proteodarican sulfate transferase.
(c)コンドロイチン硫酸プロテオダリカン硫酸基転移酵素をコードする遺伝子の発現 を RNAi効果により阻害する作用を有する核酸  (c) Nucleic acid having the action of inhibiting the expression of the gene encoding chondroitin sulfate proteodarican sulfate transferase by the RNAi effect
[0046] また「硫酸化阻害作用を有する物質」としては、例えば以下の(a)〜(c)からなる群 より選択される化合物を挙げることができる。  In addition, examples of the “substance having a sulfation inhibiting action” include compounds selected from the group consisting of the following (a) to (c).
(a)コンドロイチン硫酸プロテオダリカン硫酸基転移酵素と結合する抗体 ( a ) an antibody that binds to chondroitin sulfate proteodalycan sulfate transferase
(b)コンドロイチン硫酸プロテオダリカン硫酸基転移酵素変異体  (b) Chondroitin sulfate proteodalycan sulfate transferase mutant
(c)コンドロイチン硫酸プロテオダリカン硫酸基転移酵素と結合する低分子化合物  (c) A low molecular weight compound that binds to chondroitin sulfate proteodalycan sulfate transferase
[0047] 上記例示した酵素は、一遺伝子に対応する一酵素を示すのみならず、ある特徴を 共有する酵素群をも含む。例えばコンドロイチナーゼは、ムコ多糖分解酵素という特 徴は共有するが基質特異性などの異なる ABC、 AC、 Bなどの酵素の総称である。例 えば、コンドロイチナーゼ AC Iは、コンドロイチン硫酸類(A、 Cまたは E)、コンドロイチ ン、コンドロイチン硫酸-デルマタン硫酸ハイブリッド型およびヒアルロン酸の N-ァセチ ルへキソサミニド結合を脱離反応的に切断して、非還元末端に Δ 4-グルクロン酸残 基を持つオリゴ糖を生成する。本酵素はデルマタン硫酸 (コンドロィチン硫酸 B,へキ スロン酸として L-ィズロン酸を持つもの)、ケタラン硫酸、へパラン硫酸およびへパリン には作用しない。また、コンドロイチナーゼ AC IIは、コンドロイチン、コンドロイチン硫 酸 Aおよびコンドロイチン硫酸 Cの N-ァセチルへキソサミニド結合を脱離反応的に切 断して、 厶4-不飽和ニ糖(厶0卜05、 A Di-4Sおよび A Di-6S)を生成する。本酵素は ヒアルロン酸にもよく作用する。デルマタン硫酸 (コンドロイチン硫酸 B)には作用せず 、本酵素の競合的阻害剤となる。コンドロイチナーゼ B (デルマタナーゼ)は、デルマ タン硫酸の L-ィズロン酸に結合した N-ァセチルガラタトサミニド結合を脱離反応的に 切断し、非還元末端に Δ 4-へキスロン酸残基を持つオリゴ糖 (2糖および 4糖)を生成 する。本酵素は L-ィズロン酸を含まな!/、コンドロイチン硫酸 Aおよびコンドロイチン硫 酸 Cには作用しな!/、。デルマタン硫酸の硫酸基を除去した誘導体であるデルマタン は、この酵素の基質とはならない。デルマタン硫酸の L-ィズロン酸単位の第 2位が硫 酸化されている箇所はこの酵素によってよりょく切断される。コンドロイチナーゼ ABC は、コンドロイチン硫酸 A、コンドロイチン硫酸 C、デルマタン硫酸、コンドロイチンおよ びヒアルロン酸の N-ァセチルへキソサミニド結合を脱離反応的に切断して、非還元 末端に Δ 4-へキスロン酸残基を持つ二糖を主に生成する。本酵素はケタラン硫酸、 へパリンおよびへパラン硫酸には作用しない。コンドロイチナーゼはこのような、異な る性質を持っていながらもムコ多糖分解酵素という共通の性質を持つ酵素の総称で めり、必ずし ¾ここで列不した Chondroitinase ACI、 Chonaroitinase AC II、 Cnondrotin ase B、 Chondroitinase ABCには限られない。 [0047] The enzymes exemplified above include not only one enzyme corresponding to one gene but also an enzyme group sharing certain characteristics. For example, chondroitinase is a collective term for enzymes such as ABC, AC, and B that share the characteristics of mucopolysaccharide-degrading enzymes but differ in substrate specificity. For example, chondroitinase AC I cleaves the chondroitin sulfates (A, C or E), chondroitin, chondroitin sulfate-dermatan sulfate hybrid type and hyaluronic acid N-acetylhexoxide binding bond. Thus, an oligosaccharide having a Δ 4-glucuronic acid residue at the non-reducing end is generated. This enzyme does not act on dermatan sulfate (chondroitin sulfate B, which has L-iduronic acid as hexuronic acid), ketalan sulfate, heparan sulfate and heparin. In addition, chondroitinase AC II cleaves the N-acetyl hexosaminide bond of chondroitin, chondroitin sulfate A and chondroitin sulfate C in a desorbing reaction to produce 厶 4-unsaturated disaccharide (厶 0 卜 05, A Di-4S and A Di-6S). This enzyme also works well on hyaluronic acid. It does not act on dermatan sulfate (chondroitin sulfate B) and becomes a competitive inhibitor of this enzyme. Chondroitinase B (dermatanase) cleaves the N-acetyl galatatosaminide bond bound to L-iduronic acid of dermatan sulfate in an elimination reaction, and adds a Δ 4-hexuronic acid residue to the non-reducing end. Generate oligosaccharides (disaccharides and tetrasaccharides). This enzyme does not contain L-iduronic acid! /, Does not act on chondroitin sulfate A and chondroitin sulfate C! / ,. Dermatan, a derivative of dermatan sulfate with the sulfate group removed Is not a substrate for this enzyme. The site where the second position of the L-iduronic acid unit of dermatan sulfate is sulfated is more cleaved by this enzyme. Chondroitinase ABC cleaves the N-acetyl hexosaminide bond of chondroitin sulfate A, chondroitin sulfate C, dermatan sulfate, chondroitin, and hyaluronic acid in a reactive manner, and generates Δ 4-hexuronic acid at the non-reducing end. Mainly produces disaccharides with residues. This enzyme does not act on ketalan sulfate, heparin and heparan sulfate. Chondroitinase is a generic name for enzymes that have different properties but have a common property called mucopolysaccharide-degrading enzyme, and always ¾ Chondroitinase ACI, Chonaroitinase AC II, Cnondrotinase Not limited to B, Chondroitinase ABC.
[0048] また、このような特徴を共有する酵素群は、必ずしもゲノム DNA上の一遺伝子に対 する bのではない。例えは、 chondroitin-4-sulfatase chondroitin_6_sulfataseは、と もにゲノムデータベース上の複数のァクセッション番号で参照される配列(例えば Gen bankァクセッション番号 NT_039500 (その一部はァクセッション番号 CAAA01098429 ( 配列番号: 83)としてあらわされる)、 NT_078575、 NT_039353、 NW_001030904, NW_0 01030811、 NW_001030796, NW_000349)として公共遺伝子データベース Genbank上 で検索される。 [0048] In addition, the group of enzymes sharing such characteristics is not necessarily b for one gene on genomic DNA. For example, chondroitin-4-sulfatase chondroitin_6_sulfatase is a sequence referenced by multiple accession numbers in the genome database (for example, Gen bank accession number NT_039500 (part of which is accession number CAAA01098429 (sequence No .: 83)), and NT_078575, NT_039353, NW_001030904, NW_0 01030811, NW_001030796, NW_000349) are searched on the public gene database Genbank.
[0049] 上記に例示したもののうち個別の遺伝子に対応しているものは下記のように示され る。すなわち、上記のコンドロイチン硫酸プロテオグリカンとして例示した、 aggrican、 v ersican、 neurocan、 brevican、 β glycan、 Decorin、 Biglycan、 ribromoduiin^ PLT—しり、コ ンドロイチン硫酸プロテオダリカンを切断あるいは分解する酵素またはこれらに関連 する酵素として例示した、 ADAMTS_1、 ADAMTS-4, ADAMTS_5、 Calpain I、コンドロ ィチン合成に関わる酵素として例示した、 GalNAc4ST-l、 GalNAc4ST_2、 GALNAC4 S_6ST、 UA20ST、 GalT-I、 GalT_II、 GlcAT_I、 XylosylT,硫酸基転移酵素として例示 した、 C4ST-1、 C4ST-2、 C4ST-3、 D4ST、 C6ST_1、 C6ST-2をそれぞれヒトにおいて コードする遺伝子の公共遺伝子データベース Genbankにおけるァクセッション番号、 塩基配列、アミノ酸配列は、次の通りである。  [0049] Among those exemplified above, those corresponding to individual genes are shown as follows. In other words, aggrican, versican, neurocan, brevican, β glycan, Decorin, Biglycan, ribromoduiin ^ PLT, and the enzymes that cleave or degrade chondroitin sulfate proteoglycan exemplified above as chondroitin sulfate proteoglycans or related thereto ADAMTS_1, ADAMTS-4, ADAMTS_5, Calpain I, examples of enzymes involved in chondroitin synthesis, GalNAc4ST-l, GalNAc4ST_2, GALNAC4 S_6ST, UA20ST, GalT-I, GalT_II, GlcAT_I, XylosylT, sulfate transfer Public gene databases of genes encoding C4ST-1, C4ST-2, C4ST-3, D4ST, C6ST_1, and C6ST-2, which are exemplified as enzymes, in humans The accession numbers, nucleotide sequences, and amino acid sequences in Genbank are as follows: It is as follows.
[0050] aggrican (ァクセッション番号 NM— 007424、塩基配列の配列番号: 1、アミノ酸配列の配 列番号: 2) versican (ァクセッション番号 BC096495、塩基配列の配列番号: 3、アミノ酸配列の配 列番号: 4) [0050] aggrican (accession number NM—007424, nucleotide sequence number: 1, amino acid sequence number: 2) versican (Accession number BC096495, SEQ ID NO: 3 for nucleotide sequence, SEQ ID NO: 4 for amino acid sequence)
neurocan (ァクセッション番号 NM_010875、塩基配列の配列番号: 5、アミノ酸配列の 配列番号: 6) neurocan (accession number NM_010875, nucleotide sequence SEQ ID NO: 5, amino acid sequence SEQ ID NO: 6)
brevican (ァクセッション番号 NM_007529、塩基配列の配列番号: 7、アミノ酸配列の配 列番号: 8) brevican (Accession number NM_007529, nucleotide sequence number: 7, amino acid sequence number: 8)
β glycan (ァクセッション番号 AF039601、塩基配列の配列番号: 9、アミノ酸配列の配 列番号: 10) β glycan (accession number AF039601, nucleotide sequence number: 9, amino acid sequence number: 10)
Decorin (ァクセッション番号 NM_007833、塩基配列の配列番号: 11、アミノ酸配列の 配列番号: 12)  Decorin (accession number NM_007833, nucleotide sequence SEQ ID NO: 11, amino acid sequence SEQ ID NO: 12)
Biglycan (ァクセッション番号 BC057185、塩基配列の配列番号: 13、アミノ酸配列の 配列番号: 14)  Biglycan (Accession number BC057185, SEQ ID NO: 13 for nucleotide sequence, SEQ ID NO: 14 for amino acid sequence)
Fibromodulin (ァクセッション番号 NM_021355、塩基配列の配列番号: 15、アミノ酸配 列の配列番号: 16)  Fibromodulin (Accession number NM_021355, nucleotide sequence number: 15, amino acid sequence number: 16)
PG-Lb (ァクセッション番号 NM_007884、塩基配列の配列番号: 17、アミノ酸配列の配 列番号: 18)  PG-Lb (Accession number NM_007884, nucleotide sequence number: 17, amino acid sequence number: 18)
ADAMTS-1 (ァクセッション番号 NM_009621、塩基配列の配列番号: 19、アミノ酸配 列の配列番号: 20)  ADAMTS-1 (Accession number NM_009621, nucleotide sequence SEQ ID NO: 19, amino acid sequence SEQ ID NO: 20)
ADAMTS_4 (ァクセッション番号 NM_172845、塩基配列の配列番号: 21、アミノ酸配 列の配列番号: 22)  ADAMTS_4 (Accession number NM_172845, SEQ ID NO: 21 of nucleotide sequence, SEQ ID NO: 22 of amino acid sequence)
ADAMTS_5 (ァクセッション番号 AF140673、塩基配列の配列番号: 23、アミノ酸酉己歹 IJ の配列番号: 24)  ADAMTS_5 (Accession number AF140673, SEQ ID NO: 23, Amino Acid IJ SEQ ID: 24)
Calpain I (ァクセッション番号 NM_007600、塩基配列の配列番号: 25、アミノ酸配列の 配列番号: 26)  Calpain I (Accession number NM_007600, nucleotide sequence SEQ ID NO: 25, amino acid sequence SEQ ID NO: 26)
GalNAc4ST-l (ァクセッション番号 NM_175140、塩基配列の配列番号: 27、アミノ酸 配列の配列番号: 28)  GalNAc4ST-l (accession number NM_175140, nucleotide sequence number: 27, amino acid sequence number: 28)
GalNAc4ST-2 (ァクセッション番号 NM_199055、塩基配列の配列番号: 29、アミノ酸 配列の配列番号: 30) GALNAC4S-6ST (ァクセッション番号 NM_029935、塩基配列の配列番号: 31、ァミノ 酸配列の配列番号: 32) GalNAc4ST-2 (Accession number NM_199055, nucleotide sequence SEQ ID NO: 29, amino acid sequence SEQ ID NO: 30) GALNAC4S-6ST (Accession number NM_029935, nucleotide sequence SEQ ID NO: 31, amino acid sequence SEQ ID NO: 32)
UA20ST (ァクセッション番号 NM— 177387、塩基配列の配列番号: 33、アミノ酸配列の 配列番号: 34)  UA20ST (Accession number NM—177387, SEQ ID NO: 33 for nucleotide sequence, SEQ ID NO: 34 for amino acid sequence)
GalT-I (ァクセッション番号 NM— 016769、塩基配列の配列番号: 35、アミノ酸配列の配 列番号: 36)  GalT-I (Accession number NM—016769, nucleotide sequence number: 35, amino acid sequence number: 36)
GalT-II (ァクセッション番号 BC064767、塩基配列の配列番号: 37、アミノ酸配列の配 列番号: 38)  GalT-II (Accession number BC064767, nucleotide sequence number: 37, amino acid sequence number: 38)
GlcAT-I (ァクセッション番号 BC058082、塩基配列の配列番号: 39、アミノ酸配列の 配列番号: 40、またはァクセッション番号 NM_024256、塩基配列の配列番号: 41、ァ ミノ酸配列の配列番号: 42)  GlcAT-I (accession number BC058082, nucleotide sequence number: 39, amino acid sequence number: 40, or accession number NM_024256, nucleotide sequence number: 41, amino acid sequence number: 42) )
XylosylT (ァクセッション番号 NM— 145828、塩基配列の配列番号: 43、アミノ酸配列の 配列番号: 44)  XylosylT (Accession number NM—145828, nucleotide sequence number: 43, amino acid sequence number: 44)
C4ST-1 (ァクセッション番号 NM_021439、塩基配列の配列番号: 45、アミノ酸配列の 配列番号: 46)  C4ST-1 (Accession number NM_021439, nucleotide sequence SEQ ID NO: 45, amino acid sequence SEQ ID NO: 46)
C4ST-2 (ァクセッション番号 NM_021528、塩基配列の配列番号: 47、アミノ酸配列の 配列番号: 48)  C4ST-2 (Accession number NM_021528, nucleotide sequence number: 47, amino acid sequence number: 48)
C4ST-3 (ァクセッション番号 XM— 355798、塩基配列の配列番号: 49、アミノ酸配列の 配列番号: 50)  C4ST-3 (Accession No. XM—355798, nucleotide sequence SEQ ID NO: 49, amino acid sequence SEQ ID NO: 50)
D4ST (ァクセッション番号 NM— 028117、塩基配列の配列番号: 51、アミノ酸配列の配 列番号: 52)  D4ST (Accession number NM— 028117, nucleotide sequence number: 51, amino acid sequence number: 52)
C6ST-1 (ァクセッション番号 NM_016803、塩基配列の配列番号: 53、アミノ酸配列の 配列番号: 54)  C6ST-1 (Accession number NM_016803, nucleotide sequence SEQ ID NO: 53, amino acid sequence SEQ ID NO: 54)
C6ST-2 (ァクセッション番号 AB046929、塩基配列の配列番号: 55、アミノ酸配列の配 列番号: 56)  C6ST-2 (Accession number AB046929, nucleotide sequence number: 55, amino acid sequence number: 56)
上記以外のタンパク質であっても、例えば配列表に記載された配列と高い相同性( 通常 70%以上、好ましくは 80%以上、より好ましくは 90%以上、最も好ましくは 95%以 上)を有し、かつ、上記タンパク質が有する機能 (例えば細胞内の構成成分と結合す る機能等)を持つタンパク質は、本発明の上記タンパク質に含まれる。上記タンパク 質とは、 列えば、酉己歹 IJ番号: 2、 4、 6、 8、 10、 12、 14、 16、 18、 20、 22、 24、 26、 28 、 30、 32、 34、 36、 38、 40、 42、 44、 46、 48、 50、 52、 54、 56のレヽずれ力、に記載 のアミノ酸配列において、 1以上のアミノ酸が付カロ、欠失、置換、揷入されたアミノ酸 配列からなるタンパク質であって、通常変化するアミノ酸数が 30アミノ酸以内、好まし くは 10アミノ酸以内、より好ましくは 5アミノ酸以内、最も好ましくは 3アミノ酸以内であるEven proteins other than those described above have high homology (usually 70% or more, preferably 80% or more, more preferably 90% or more, most preferably 95% or more) with the sequences described in the sequence listing. And the function of the protein (for example, it binds to the intracellular components) The protein having a function or the like) is included in the protein of the present invention. The above proteins are, for example, the following: IJ numbers: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36 , 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, the amino acid sequence in which one or more amino acids are added, calo, deletion, substitution, or insertion A protein consisting of a sequence, wherein the number of normally changing amino acids is within 30 amino acids, preferably within 10 amino acids, more preferably within 5 amino acids, most preferably within 3 amino acids.
Yes
[0052] 本発明における上記遺伝子には、例えば、配列番号: 1、 3、 5、 7、 9、 11、 13、 15 、 17、 19、 21、 23、 25、 27、 29、 31、 33、 35、 37、 39、 41、 43、 45、 47、 49、 51 、 53、 55のいずれかに記載の塩基配列からなる DNAに対応する他の生物における 内在性の遺伝子(ヒトの上記遺伝子のホモログ等)が含まれる。  [0052] Examples of the gene in the present invention include, for example, SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 51, 53, or 55, an endogenous gene in another organism corresponding to the DNA comprising the nucleotide sequence described in any one of the above (a homologue of the above human gene) Etc.).
[0053] また、酉己歹 IJ番号: 1、 3、 5、 7、 9、 11、 13、 15、 17、 19、 21、 23、 25、 27、 29、 31、  [0053] Also, IJ numbers: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31,
33、 35、 37、 39、 41、 43、 45、 47、 49、 51、 53、 55のいずれ力、に記載の塩基酉己歹 IJ 力、らなる DNAに対応する他の生物の内在性の DNAは、一般的に、それぞれ配列番 号: 1、 3、 5、 7、 9、 11、 13、 15、 17、 19、 21、 23、 25、 27、 29、 31、 33、 35、 37、 39、 41、 43、 45、 47、 49、 51、 53、 55のいずれ力、に記載の DNAと高い申目同十生を有 する。高い相同性とは、 50%以上、好ましくは 70%以上、さらに好ましくは 80%以上、 より好ましくは 90%以上(例えば、 95%以上、さらには 96%、 97%、 98%または 99%以 上)の相同性を意味する。この相同性は、 mBLASTアルゴリズム(Altschul et al. (1990 ) Proc. Natl. Acad. Sci. USA 87: 2264—8; Karlin and Altschul (1993) Proc. Natl. Aca d. Sci. USA 90: 5873-7)によって決定すること力 Sできる。また、該 DNAは、生体内から 単離した場合、それぞれ酉己歹 IJ番号: 1、 3、 5、 7、 9、 11、 13、 15、 17、 19、 21、 23、 25、 27、 29、 31、 33、 35、 37、 39、 41、 43、 45、 47、 49、 51、 53、 55に記載の D NAとストリンジェントな条件下でハイブリダィズすると考えられる。ここで「ストリンジェン トな条件」としては、例えば「2 X SSC、 0.1%SDS、 50。C」、「2 X SSC、 0.1%SDS、 42°C」 、「1 X SSC、 0.1%SDS、 37°C」、よりストリンジェントな条件として「2 X SSC、 0.1%SDS、 65。C」、「0.5 X SSC、 0.1%SDS、 42。C」および「0.2 X SSC、 0.1%SDS、 65°C」の条件を 挙げること力 Sでさる。 [0054] 当業者は、上記の高い相同性を持つタンパク質から、上記のタンパク質に機能的 に同等なタンパク質を、コンドロイチン硫酸プロテオダリカンの分解促進作用、合成阻 害作用、脱硫酸化作用または硫酸化阻害作用の活性測定方法を用いることにより適 宜取得すること力できる。具体的な活性測定方法は、後出の本発明におけるスクリー ユング方法の項にて記載される。また当業者においては、他の生物における上記遺 伝子に相当する内在性の遺伝子を、上記遺伝子の塩基配列を基に適宜取得するこ とが可能である。なお、本明細書においては、ヒト以外の生物における上記タンパク 質および遺伝子に相当する上記タンパク質および遺伝子、あるいは、上述のタンパク 質および遺伝子と機能的に同等な上記タンパク質および遺伝子も、単に上記の名称 で記載する場合がある。 33, 35, 37, 39, 41, 43, 45, 47, 49, 51, 53, 55 DNA is generally sequence numbers: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, respectively. 39, 41, 43, 45, 47, 49, 51, 53, and 55. High homology means 50% or more, preferably 70% or more, more preferably 80% or more, more preferably 90% or more (for example, 95% or more, further 96%, 97%, 98% or 99% or more). Means the homology of (above). This homology is determined by the mBLAST algorithm (Altschul et al. (1990) Proc. Natl. Acad. Sci. USA 87: 2264-8; Karlin and Altschul (1993) Proc. Natl. Aca d. Sci. USA 90: 5873- 7) Power can be determined by S. Further, when the DNA is isolated from the living body, the IJ numbers: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 51, 53, 55. It is considered that the DNA hybridizes under stringent conditions. Here, as “stringent conditions”, for example, “2 X SSC, 0.1% SDS, 50.C”, “2 X SSC, 0.1% SDS, 42 ° C”, “1 X SSC, 0.1% SDS, 37 ° C ”and more stringent conditions“ 2 X SSC, 0.1% SDS, 65.C ”,“ 0.5 X SSC, 0.1% SDS, 42.C ”and“ 0.2 X SSC, 0.1% SDS, 65 ° The ability S to raise the condition of “C”. [0054] A person skilled in the art will convert a protein functionally equivalent to the above protein from the above highly homologous proteins into a chondroitin sulfate proteodarican degradation promoting action, synthetic inhibitory action, desulfating action or sulfating action. It can be obtained appropriately by using an inhibitory activity measurement method. A specific activity measuring method will be described in the section of the screening method in the present invention. Moreover, those skilled in the art can appropriately obtain an endogenous gene corresponding to the above gene in another organism based on the base sequence of the above gene. In the present specification, the above-mentioned proteins and genes corresponding to the above-mentioned proteins and genes in organisms other than humans, or the above-mentioned proteins and genes functionally equivalent to the above-mentioned proteins and genes are also simply referred to as It may be described in.
[0055] 本発明の上記タンパク質は、天然のタンパク質としてのほか、遺伝子組み換え技術 を利用した組換えタンパク質として調製することができる。天然のタンパク質としては、 例えば上記タンパク質が発現していると考えられる細胞 (組織)の抽出液に対し、上 記タンパク質に対する抗体を用いたァフィ二ティークロマトグラフィーを用いる方法に より調製することが可能である。一方、組換えタンパク質は、例えば、上記タンパク質 をコードする DNAで形質転換した細胞を培養することにより、調製することが可能であ る。本発明の上記タンパク質は、例えば、後述のスクリーニング方法において好適に 用いられる。  [0055] The protein of the present invention can be prepared not only as a natural protein but also as a recombinant protein using a gene recombination technique. As a natural protein, for example, it can be prepared by a method using affinity chromatography using an antibody against the above protein against an extract of a cell (tissue) considered to express the above protein. It is. On the other hand, a recombinant protein can be prepared, for example, by culturing cells transformed with DNA encoding the protein. The above-mentioned protein of the present invention is suitably used, for example, in the screening method described later.
[0056] 本発明における「核酸」とは RNAまたは DNAを意味する。また所謂 PNA(p印 tide nuc leic acid)等の化学合成核酸アナログも、本発明の核酸に含まれる。 PNAは、核酸の 基本骨格構造である五単糖 ·リン酸骨格を、グリシンを単位とするポリアミド骨格に置 換したもので、核酸によく似た 3次元構造を有する。  [0056] "Nucleic acid" in the present invention means RNA or DNA. Chemically synthesized nucleic acid analogs such as so-called PNA (p-mark tide nucleic acid) are also included in the nucleic acids of the present invention. PNA replaces the pentose / phosphate skeleton, which is the basic skeleton structure of nucleic acid, with a polyamide skeleton with glycine as a unit, and has a three-dimensional structure very similar to nucleic acid.
[0057] 特定の内在性遺伝子の発現を阻害する方法としては、アンチセンス技術を利用す る方法が当業者によく知られている。アンチセンス核酸が標的遺伝子の発現を阻害 する作用としては、以下のような複数の要因が存在する。即ち、三重鎖形成による転 写開始阻害、 RNAポリメラーゼによって局部的に開状ループ構造が作られた部位と のハイブリッド形成による転写阻害、合成の進みつつある RNAとのハイブリッド形成に よる転写阻害、イントロンとエタソンとの接合点におけるハイブリッド形成によるスプラ イシング阻害、スプライソソーム形成部位とのハイブリッド形成によるスプライシング阻 害、 mRNAとのハイブリッド形成による核から細胞質への移行阻害、キヤッビング部位 やポリ (A)付加部位とのハイブリッド形成によるスプライシング阻害、翻訳開始因子結 合部位とのハイブリッド形成による翻訳開始阻害、開始コドン近傍のリボソーム結合 部位とのハイブリッド形成による翻訳阻害、 mRNAの翻訳領域やポリソーム結合部位 とのハイブリッド形成によるペプチド鎖の伸長阻害、および核酸とタンパク質との相互 作用部位とのハイブリッド形成による遺伝子発現阻害などである。このようにアンチセ ンス核酸は、転写、スプライシングまたは翻訳など様々な過程を阻害することで、標 的遺伝子の発現を阻害する(平島および井上,新生化学実験講座 2核酸 IV遺伝子 の複製と発現, 日本生化学会編,東京化学同人, 1993, 319-347.)。 [0057] As a method for inhibiting the expression of a specific endogenous gene, a method using an antisense technique is well known to those skilled in the art. There are a number of factors that cause the antisense nucleic acid to inhibit the expression of the target gene. Inhibition of transcription initiation due to triplex formation, transcription inhibition due to hybridization with a site where an open loop structure was locally created by RNA polymerase, transcription inhibition due to hybridization with RNA undergoing synthesis, intron By splicing at the junction of Etason Inhibiting splicing, splicing inhibition by hybridization with spliceosome formation site, inhibition of translocation from nucleus to cytoplasm by hybridization with mRNA, inhibition of splicing by hybridization with cabbing site and poly (A) addition site, translation initiation factor Inhibition of translation initiation by hybridization with the binding site, inhibition of translation by hybridization with the ribosome binding site near the initiation codon, inhibition of peptide chain elongation by hybridization with the translation region of mRNA and polysome binding site, and nucleic acid and protein Gene expression inhibition by hybridization with the interaction site. Thus, antisense nucleic acids inhibit the expression of target genes by inhibiting various processes such as transcription, splicing or translation (Hirashima and Inoue, Shinsei Kagaku Kogaku Kenkyu 2 Nucleic acid IV gene replication and expression, Japan Biochemical Society, Tokyo Chemical Doujin, 1993, 319-347.).
[0058] 本発明で用いられるアンチセンス核酸は、上記のいずれの作用により、上述のコン ドロイチン硫酸プロテオダリカンのコアタンパク質、合成酵素、脱硫酸化酵素抑制タン ノ ク質、硫酸基転移酵素のいずれ力、をコードする遺伝子の発現および/または機能 を阻害してもよい。一つの態様としては、上述のコンドロイチン硫酸プロテオダリカン のコアタンパク質、合成酵素、脱硫酸化酵素抑制タンパク質、または硫酸基転移酵 素をコードする遺伝子の mRNAの 5'端近傍の非翻訳領域に相補的なアンチセンス配 列を設計すれば、遺伝子の翻訳阻害に効果的と考えられる。また、コード領域もしく は 3'側の非翻訳領域に相補的な配歹 IJも使用することができる。 [0058] The antisense nucleic acid used in the present invention can be any of the above-mentioned chondroitin sulfate proteodarican core protein, synthase, desulfase-suppressing protein, and sulfotransferase by any of the above-described actions. Expression and / or function of the gene encoding the force may be inhibited. In one embodiment, the above-mentioned chondroitin sulfate proteodarican core protein, synthase, desulfase inhibitor protein, or a gene encoding a sulfate transfer enzyme is complementary to the untranslated region near the 5 'end of the mRNA. If an antisense sequence is designed, it would be effective to inhibit gene translation. In addition, an IJ complementary to the coding region or the 3 ′ untranslated region can also be used.
[0059] このように、上述のコンドロイチン硫酸プロテオダリカンのコアタンパク質、合成酵素 、脱硫酸化酵素抑制タンパク質、または硫酸基転移酵素をコードする遺伝子の翻訳 領域だけでなぐ非翻訳領域の配列のアンチセンス配歹 I]を含む核酸も、本発明で利 用されるアンチセンス核酸に含まれる。使用されるアンチセンス核酸は、適当なプロ モーターの下流に連結され、好ましくは 3'側に転写終結シグナルを含む配列が連結 される。このようにして調製された核酸は、公知の方法を用いることで所望の動物(細 胞)に形質転換することができる。アンチセンス核酸の配列は、形質転換される動物( 細胞)が有する内在性のコンドロイチン硫酸プロテオダリカンのコアタンパク質、合成 酵素、脱硫酸化酵素抑制タンパク質、または硫酸基転移酵素をコードする遺伝子ま たはその一部と相補的な配列であることが好ましいが、遺伝子の発現を有効に抑制 できる限りにおいて、完全に相補的でなくてもよい。転写された RNAは標的遺伝子の 転写産物に対して好ましくは 90%以上、最も好ましくは 95%以上の相補性を有する。 アンチセンス核酸を用いて標的遺伝子の発現を効果的に阻害するには、アンチセン ス核酸の長さは少なくとも 15塩基以上 25塩基未満であることが好ましいが、本発明の アンチセンス核酸は必ずしもこの長さに限定されず、例えば 100塩基以上、または 500 塩基以上であってもよい。 [0059] As described above, the antisense of the sequence of the above-mentioned chondroitin sulfate proteodarican core protein, synthase, desulfase inhibitor protein, or untranslated region that includes not only the translation region of the gene encoding the sulfotransferase. Nucleic acids containing [I] are also included in the antisense nucleic acids used in the present invention. The antisense nucleic acid used is linked downstream of an appropriate promoter, and preferably a sequence containing a transcription termination signal is linked on the 3 ′ side. The nucleic acid thus prepared can be transformed into a desired animal (cell) using a known method. The sequence of the antisense nucleic acid is the gene or gene encoding the endogenous chondroitin sulfate proteodarican core protein, synthase, desulfase inhibitor protein, or sulfate group of the animal (cell) to be transformed. It is preferably a sequence complementary to a part of it, but it effectively suppresses gene expression. As far as possible, they need not be completely complementary. The transcribed RNA preferably has a complementarity of 90% or more, most preferably 95% or more, to the target gene transcript. In order to effectively inhibit the expression of a target gene using an antisense nucleic acid, the length of the antisense nucleic acid is preferably at least 15 bases and less than 25 bases, but the antisense nucleic acid of the present invention is not necessarily of this length. For example, it may be 100 bases or more, or 500 bases or more.
[0060] 本発明のアンチセンス核酸は特に制限されないが、例えば Versican遺伝子の塩基 配列(GenBankのァクセッション番号 BC096495、配列番号: 3)、 C4ST-1 (GenBankの ァクセッション番号 NM_021439、配列番号: 45)、 C4ST-2 (GenBankのァクセッション 番号 NM_021528、配列番号: 47)、 C4ST-3 (GenBankのァクセッション番号 XM_35579 8、配列番号: 49)等をもとに作成することができる。  [0060] The antisense nucleic acid of the present invention is not particularly limited. For example, the base sequence of the Versican gene (GenBank accession number BC096495, SEQ ID NO: 3), C4ST-1 (GenBank accession number NM_021439, SEQ ID NO: : 45), C4ST-2 (GenBank accession number NM_021528, SEQ ID NO: 47), C4ST-3 (GenBank accession number XM_35579 8, SEQ ID NO: 49), etc. .
[0061] 上述のコンドロイチン硫酸プロテオダリカンのコアタンパク質、合成酵素、脱硫酸化 酵素抑制タンパク質、または硫酸基転移酵素をコードする遺伝子の発現の阻害は、 リボザィム、またはリボザィムをコードする DNAを利用して行うことも可能である。リボザ ィムとは触媒活性を有する RNA分子を指す。リボザィムには種々の活性を有するもの が存在する力 S、中でも RNAを切断する酵素としてのリボザィムに焦点を当てた研究に より、 RNAを部位特異的に切断するリボザィムの設計が可能となった。リボザィムには 、グループ Iイントロン型や RNase Pに含まれる Ml RNAのように 400ヌクレオチド以上の 大きさのものもあるが、ハンマーヘッド型やヘアピン型と呼ばれる 40ヌクレオチド程度 の活性ドメインを有するものもある(小泉誠および大塚栄子,タンパク質核酸酵素, 19 90, 35, 2191·)。  [0061] Inhibition of the expression of the above-mentioned chondroitin sulfate proteodarican core protein, synthase, desulfation enzyme inhibitory protein, or gene encoding sulfotransferase is performed using ribozyme or DNA encoding ribozyme. It is also possible to do this. Ribozyme refers to an RNA molecule that has catalytic activity. The ability of ribozymes to have various activities exists, and in particular, research focusing on ribozymes as enzymes that cleave RNA has made it possible to design ribozymes that cleave RNA in a site-specific manner. Some ribozymes have a size of 400 nucleotides or more, such as group I intron type and Ml RNA contained in RNase P, but some have an active domain of about 40 nucleotides called hammerhead type or hairpin type. (Makoto Koizumi and Eiko Otsuka, Protein Nucleic Acid Enzyme, 1990, 35, 2191 ·).
[0062] 例えば、ハンマーヘッド型リボザィムの自己切断ドメインは、 G13U14C15という配列 の C15の 3'側を切断するが、その活性には U14と A9との塩基対形成が重要とされ、 C1 5の代わりに A15または U15でも切断され得ることが示されている(Koizumi, M. et al., FEBS Lett, 1988, 228, 228.)。基質結合部位が標的部位近傍の RNA配列と相補的 なリボザィムを設計すれば、標的 RNA中の UC、 UUまたは UAという配列を認識する制 限酵素的な RNA切断リボザィムを作出することができる(Koizumi, M. et al., FEBS Le tt, 1988, 239, 285·、小泉誠および大塚栄子,タンパク質核酸酵素, 1990, 35, 2191. 、 oizumi, M. et al., Nucl Acids Res, 1989, 17, 7059·)。 [0062] For example, the self-cleaving domain of the hammerhead ribozyme cleaves 3 ′ of C15 in the sequence G13U14C15, but base pairing between U14 and A9 is important for its activity. Shows that A15 or U15 can also be cleaved (Koizumi, M. et al., FEBS Lett, 1988, 228, 228.). By designing a ribozyme whose substrate binding site is complementary to the RNA sequence in the vicinity of the target site, it is possible to create a restriction RNA-cleaving ribozyme that recognizes the sequence UC, UU or UA in the target RNA (Koizumi , M. et al., FEBS Le tt, 1988, 239, 285, Makoto Koizumi and Eiko Otsuka, Protein Nucleic Acid Enzymes, 1990, 35, 2191. Oizumi, M. et al., Nucl Acids Res, 1989, 17, 7059.).
[0063] また、ヘアピン型リボザィムも本発明の目的に有用である。このリボザィムは、例え ばタバコリングスポットウィルスのサテライト RNAのマイナス鎖に見出される(Buzayan, JM., Nature, 1986, 323, 349·)。ヘアピン型リボザィム力、らも、標的特異的な RNA切断 リボザィムを作出できることが示されている(Kikuchi, Y. & Sasaki, N., Nucl Acids Res, 1991, 19, 6751.、菊池洋,化学と生物, 1992, 30, 112.)。このように、リボザィムを用い て上述のコンドロイチン硫酸プロテオダリカンのコアタンパク質、合成酵素、脱硫酸化 酵素抑制タンパク質、または硫酸基転移酵素をコードする遺伝子の転写産物を特異 的に切断することで、該遺伝子の発現を阻害することができる。  [0063] Hairpin ribozymes are also useful for the purposes of the present invention. This ribozyme is found, for example, in the minus strand of tobacco ring spot virus satellite RNA (Buzayan, JM., Nature, 1986, 323, 349). It has been shown that hairpin-type ribozymes can also produce target-specific RNA cleavage ribozymes (Kikuchi, Y. & Sasaki, N., Nucl Acids Res, 1991, 19, 6751., Hiroshi Kikuchi, Biology, 1992, 30, 112.). In this way, the ribozyme is used to specifically cleave the above-mentioned chondroitin sulfate proteodarican core protein, synthase, desulfase inhibitor protein, or transcript of a gene encoding a sulfotransferase. Gene expression can be inhibited.
[0064] 内在性遺伝子の発現の抑制は、さらに、標的遺伝子配列と同一もしくは類似した配 列を有する二本鎖 RNAを用いた RNA干渉(RNA interference,以下「RNAi」と略称す る)によっても fiうことができる。  [0064] Inhibition of endogenous gene expression may also be caused by RNA interference (hereinafter abbreviated as "RNAi") using double-stranded RNA having the same or similar sequence as the target gene sequence. can be fi.
[0065] 近年のゲノムプロジェクトの完了によってヒトの全塩基配列が解読され数多くの疾患 関連遺伝子が盛んに同定されている現在、特定の遺伝子を標的とした治療法、創薬 開発が盛んに実施されている。中でも特異的転写後抑制効果を発揮する small interf ering RNA (siRNA)の遺伝子治療への応用が注目されている。 RNAiは、 2本鎖 RNA( dsRNA)が直接細胞内に取り込まれると、この dsRNAと相同な配列を持つ遺伝子の発 現が抑えられ現在注目を浴びている手法である。哺乳類細胞においては、短鎖 dsR NA(siRNA)を用いることにより、 RNAiを誘導する事が可能で、 RNAiは、ノックアウトマ ウスと比較して、効果が安定、実験が容易、費用が安価であるなど、多くの利点を有 している。  [0065] With the completion of recent genome projects, the entire human base sequence has been decoded and many disease-related genes have been actively identified. Currently, therapeutic methods and drug discovery targeting specific genes are being actively implemented. ing. Of particular interest is the application of small interfering RNA (siRNA), which exerts a specific post-transcriptional inhibitory effect, to gene therapy. RNAi is a technique that is currently attracting attention because when double-stranded RNA (dsRNA) is directly taken into cells, the expression of genes with a sequence homologous to this dsRNA is suppressed. In mammalian cells, RNAi can be induced by using short dsRNA (siRNA). RNAi is more stable, easier to experiment, and less expensive than knockout mice. Has many advantages.
[0066] RNAi効果による阻害作用を有する核酸は、一般的に siRNAもしくは shRNAとも呼ば れる。 RNAiは、標的遺伝子の mRNAと相同な配列からなるセンス RNAとこれと相補的 な配列からなるアンチセンス RNAとからなる短鎖二本鎖 RNA (以下、「dsRNA」と略称 する)を細胞等に導入することにより、標的遺伝子 mRNAに特異的かつ選択的に結合 して破壊を誘導し、当該標的遺伝子を切断することにより標的遺伝子の発現を効率 よく阻害する(抑制する)現象である。例えば、 dsRNAを細胞内に導入すると、その RN Aと相同配列の遺伝子の発現が抑制(ノックダウン)される。このように RNAiは、標的 遺伝子の発現を抑制し得ることから、従来の煩雑で効率の低い相同組換えによる遺 伝子破壊方法に代わる簡易な遺伝子ノックアウト方法として、または、遺伝子治療へ の応用可能な方法として注目されている。 RNAiに用いる RNAは、上述のコンドロイチ ン硫酸プロテオダリカンのコアタンパク質、合成酵素、脱硫酸化酵素抑制タンパク質 、または硫酸基転移酵素をコードする遺伝子もしくは該遺伝子の部分領域と必ずしも 完全に同一である必要はないが、完全な相同性を有することが好ましい。 [0066] A nucleic acid having an inhibitory action by the RNAi effect is generally also called siRNA or shRNA. RNAi is a short double-stranded RNA (hereinafter abbreviated as “dsRNA”) consisting of a sense RNA consisting of a sequence homologous to the mRNA of the target gene and an antisense RNA consisting of a complementary sequence. This is a phenomenon that induces destruction by specifically and selectively binding to the target gene mRNA, and efficiently inhibiting (suppressing) the expression of the target gene by cleaving the target gene. For example, when dsRNA is introduced into a cell, the expression of the gene homologous to the RNA is suppressed (knocked down). Thus RNAi is the target Since gene expression can be suppressed, it has been attracting attention as a simple gene knockout method to replace conventional complicated and low efficiency homologous recombination methods, or as a method applicable to gene therapy. . The RNA used for RNAi must be completely identical to the above-mentioned chondroitin sulfate proteodarican core protein, synthetic enzyme, desulfase inhibitor protein, or gene encoding a sulfotransferase, or a partial region of the gene. None, but preferably has complete homology.
[0067] siRNAの設計にあたっては、ターゲットとしては上述のコンドロイチン硫酸プロテオグ リカンのコアタンパク質、合成酵素、脱硫酸化酵素抑制タンパク質、または硫酸基転 移酵素をコードする遺伝子であれば特に限定されるものではなぐ任意の領域を全 てターゲット候補とすることが可能である。例えば、 Versican遺伝子の塩基配列(配列 番号: 3)、 C4ST-1遺伝子の塩基配列(配列番号: 45)、 C4ST-2遺伝子の塩基配列( 配列番号: 47)、 C4ST-3遺伝子の塩基配列(配列番号: 49)等をもとに作成すること 力 Sできる。より具体的には、その配列の一部の領域をターゲット候補とすることが可能 であり、例えば、 Versican遺伝子の塩基配列の一部領域(配列番号: 57)、 C4ST-1遺 伝子の塩基配列の一部領域 (配列番号: 58)、 C4ST-2遺伝子の塩基配列の一部領 域 (配列番号: 59)、 C4ST-3遺伝子の塩基配列の一部領域 (配列番号: 60)、 C6ST- 1遺伝子の塩基配列の一部領域 (配列番号: 61)、 C6ST-2遺伝子の塩基配列の一 部領域 (配列番号: 62)、 GalNAc4ST-l遺伝子の塩基配列の一部領域 (配列番号: 6 3)、 GalNAc4ST-2遺伝子の塩基配列の一部領域(配列番号: 64)、 GALNAC4S-6S Tの塩基配列の一部領域 (配列番号: 65)等をもとに作成することができる。さらに具 体的には、本明細書によって具体的に示された DNA配列(配列番号: 7;!〜 82)を標 的とする siRNAが例示できる。  [0067] In designing siRNA, the target is not particularly limited as long as it is a gene encoding the above-mentioned chondroitin sulfate proteoglycan core protein, synthetic enzyme, desulfase inhibitor protein, or sulfate transferase. It is possible to make any arbitrary region as a target candidate. For example, the base sequence of Versican gene (SEQ ID NO: 3), the base sequence of C4ST-1 gene (SEQ ID NO: 45), the base sequence of C4ST-2 gene (SEQ ID NO: 47), the base sequence of C4ST-3 gene ( It can be created based on SEQ ID NO: 49). More specifically, a partial region of the sequence can be a target candidate. For example, a partial region of the base sequence of the Versican gene (SEQ ID NO: 57), the base of the C4ST-1 gene Partial region of sequence (SEQ ID NO: 58), partial region of base sequence of C4ST-2 gene (SEQ ID NO: 59), partial region of base sequence of C4ST-3 gene (SEQ ID NO: 60), C6ST -Partial region of the base sequence of 1 gene (SEQ ID NO: 61), partial region of the base sequence of C6ST-2 gene (SEQ ID NO: 62), partial region of the base sequence of GalNAc4ST-l gene (SEQ ID NO: 6 3), a partial region of the nucleotide sequence of the GalNAc4ST-2 gene (SEQ ID NO: 64), a partial region of the base sequence of GALNAC4S-6ST (SEQ ID NO: 65), and the like. More specifically, siRNA targeting the DNA sequence specifically shown by the present specification (SEQ ID NO: 7;! To 82) can be exemplified.
[0068] siRNAを細胞に導入するには、 in vitroで合成した siRNAをプラスミド DNAに連結し てこれを細胞に導入する方法、 2本の RNAをァニールする方法などを採用することが できる。  [0068] In order to introduce siRNA into a cell, a method in which siRNA synthesized in vitro is linked to plasmid DNA and introduced into the cell, a method of annealing two RNAs, or the like can be employed.
[0069] また上記 2本の RNA分子は、ここで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 (shRNA)であってもよい。 shRNAとは、ショートヘアピン RNA(sh ort hairpin RNA)と呼ばれ、一本鎖の一部の領域が他の領域と相補鎖を形成するた めにステムループ構造を有する RNA分子である。即ち、分子内において二本鎖 RNA 構造を形成し得る分子もまた本発明の siRNAに含まれる。 [0069] The two RNA molecules may be molecules having a structure in which one end is closed, for example, siRNA (shRNA) having a hairpin structure. shRNA is called short hairpin RNA, and part of a single strand forms a complementary strand with other regions. This is an RNA molecule having a stem-loop structure. That is, a molecule capable of forming a double-stranded RNA structure in the molecule is also included in the siRNA of the present invention.
[0070] また本発明の好ましい態様としては、 Versican、 C4ST_1、 C4ST_2、 C4ST-3等の発 現を RNAi効果により抑制し得る RNA(siRNA)であって、本明細書によって具体的に 示された DNA配歹 IJ (配列番号: 7;!〜 82)を標的とする siRNAにおいて、例えば、 1もし くは少数の RNAが付加もしくは欠失された構造の二本鎖 RNAであっても、上述のコン ドロイチン硫酸プロテオダリカンのコアタンパク質、合成酵素、脱硫酸化酵素抑制タン パク質、または硫酸基転移酵素をコードする遺伝子の発現を抑制する機能を有する ものであれば、本発明の siRNAに含まれる。  [0070] Further, as a preferred embodiment of the present invention, RNA (siRNA) capable of suppressing the expression of Versican, C4ST_1, C4ST_2, C4ST-3, etc. by the RNAi effect, which is specifically shown in the present specification In siRNA targeting DNA ligated IJ (SEQ ID NO: 7;! -82), for example, even if it is a double-stranded RNA having a structure in which one or a few RNAs are added or deleted, the above-mentioned Any siRNA of the present invention may be used as long as it has a function of suppressing the expression of a gene encoding a chondroitin sulfate proteodarican core protein, a synthetic enzyme, a desulfase inhibitor protein, or a sulfotransferase. .
[0071] RNAi (siRNA)のために使用される RNAは、上記タンパク質をコードする遺伝子もし くは該遺伝子の部分領域と完全に同一(相同)である必要はないが、完全な同一(相 同)性を有することが好ましい。  [0071] RNA used for RNAi (siRNA) need not be completely identical (homologous) to the gene encoding the protein or a partial region of the gene, but is completely identical (similar) ).
[0072] RNAi機構の詳細については未だに不明な部分もある力 DICERといわれる酵素(R Nase III核酸分解酵素ファミリーの一種)が二本鎖 RNAと接触し、二本鎖 RNAが small i nterfering RNAまたは siRNAと呼ばれる小さな断片に分解されるものと考えられている 。本発明における RNAi効果を有する二本鎖 RNAには、このように DICERによって分 解される前の二本鎖 RNAも含まれる。即ち、そのままの長さでは RNAi効果を有さない ような長鎖の RNAであっても、細胞において RNAi効果を有する siRNAへ分解されるこ とが期待されるため、本発明における二本鎖 RNAの長さは、特に制限されない。  [0072] The details of the RNAi mechanism are still unclear. An enzyme called DICER (a member of the RNase III nuclease family) comes into contact with double-stranded RNA, and the double-stranded RNA becomes small interfering RNA or It is thought to be broken down into small fragments called siRNAs. The double-stranded RNA having the RNAi effect in the present invention includes double-stranded RNA before being digested by DICER as described above. That is, even if a long RNA that does not have the RNAi effect as it is is expected to be degraded into siRNA having the RNAi effect in the cell, the double-stranded RNA in the present invention The length of is not particularly limited.
[0073] 例えば、上述のコンドロイチン硫酸プロテオダリカンのコアタンパク質、合成酵素、 脱硫酸化酵素抑制タンパク質、または硫酸基転移酵素をコードする遺伝子の mRNA の全長もしくはほぼ全長の領域に対応する長鎖二本鎖 RNAを、例えば、予め DICER で分解させ、その分解産物を本発明の薬剤として利用することが可能である。この分 解産物には、 RNAi効果を有する二本鎖 RNA分子(siRNA)が含まれることが期待され る。この方法によれば、 RNAi効果を有することが期待される mRNA上の領域を、特に 選択しなくともよい。即ち、 RNAi効果を有する本発明の上述の遺伝子の mRNA上の 領域は、必ずしも正確に規定される必要はない。  [0073] For example, the above-mentioned chondroitin sulfate proteodarican core protein, synthase, desulfase inhibitor protein, or two long-chains corresponding to the full-length or almost full-length region of mRNA of the gene encoding sulfate group transferase For example, the strand RNA can be decomposed in advance with DICER, and the degradation product can be used as the agent of the present invention. This degradation product is expected to contain double-stranded RNA molecules (siRNA) having the RNAi effect. According to this method, it is not necessary to particularly select a region on mRNA expected to have an RNAi effect. That is, the region on the mRNA of the above-mentioned gene of the present invention having an RNAi effect does not necessarily need to be accurately defined.
[0074] 本発明の上記「RNAi効果により抑制し得る二本鎖 RNA」は、当業者においては、該 二本鎖 RNAの標的となる上述のコンドロイチン硫酸プロテオダリカンのコアタンパク質 、合成酵素、脱硫酸化酵素抑制タンパク質、または硫酸基転移酵素をコードする遺 伝子の塩基配列を基に、適宜作製することができる。一例を示せば、配列番号: 71 に記載の塩基配列をもとに、本発明の二本鎖 RNAを作製することができる。即ち、配 列番号: 71に記載の塩基配列をもとに、該配列の転写産物である mRNAの任意の連 続する RNA領域を選択し、この領域に対応する二本鎖 RNAを作製することは、当業 者においては、通常の試行の範囲内において適宜行い得ることである。また、該配 列の転写産物である mRNA配列から、より強!/、RNAi効果を有する siRNA配列を選択 することも、当業者においては、公知の方法によって適宜実施することが可能である 。また、一方の鎖が判明していれば、当業者においては容易に他方の鎖 (相補鎖)の 塩基配列を知ることができる。 siRNAは、当業者においては市販の核酸合成機を用 いて適宜作製することが可能である。また、所望の RNAの合成については、一般の合 成受託サービスを利用することができる。 [0074] The above-mentioned "double-stranded RNA that can be suppressed by the RNAi effect" of the present invention is known to those skilled in the art. Produced as appropriate based on the base sequence of the gene encoding the above chondroitin sulfate proteodarican core protein, synthetic enzyme, desulfase inhibitor protein, or sulfotransferase, which is the target of double-stranded RNA. Can do. For example, the double-stranded RNA of the present invention can be prepared based on the nucleotide sequence set forth in SEQ ID NO: 71. That is, based on the nucleotide sequence set forth in SEQ ID NO: 71, an arbitrary continuous RNA region of mRNA that is a transcription product of the sequence is selected, and a double-stranded RNA corresponding to this region is prepared. The person skilled in the art can appropriately carry out within the range of normal trials. Moreover, those skilled in the art can appropriately select a siRNA sequence having a stronger // RNAi effect from the mRNA sequence which is a transcription product of the sequence, by a known method. If one strand is known, those skilled in the art can easily know the base sequence of the other strand (complementary strand). A siRNA can be appropriately prepared by those skilled in the art using a commercially available nucleic acid synthesizer. In addition, for synthesis of desired RNA, a general synthetic contract service can be used.
[0075] また、本発明における siRNAは、必ずしも標的配列に対する一組の 2本鎖 RNAであ る必要はなぐ標的配列を含んだ領域に対する複数組の 2本鎖 RNAの混合物であつ てもよい。ここで標的配列に対応した核酸混合物としての siRNAは、当業者において は市販の核酸合成機および DICER酵素を用いて適宜作成することが可能であり、ま た、所望の RNAの合成については、一般の合成受託サービスを利用することができ る。なお、本発明の siRNAには、所謂「カクテル siRNA」が含まれる。  [0075] In addition, the siRNA in the present invention may be a mixture of a plurality of sets of double-stranded RNAs for a region containing a target sequence, which need not necessarily be a set of double-stranded RNAs for the target sequence. Here, siRNA as a nucleic acid mixture corresponding to the target sequence can be appropriately prepared by a person skilled in the art using a commercially available nucleic acid synthesizer and a DICER enzyme. You can use the composite contract service. The siRNA of the present invention includes so-called “cocktail siRNA”.
[0076] また、本発明における siRNAは、必ずしも全てのヌクレオチドがリボヌクレオチド(RN A)でなくともよい。即ち、本発明において、 siRNAを構成する 1もしくは複数のリボヌク レオチドは、対応するデォキシリボヌクレオチドであってもよい。この「対応する」とは、 糖部分の構造は異なるものの、同一の塩基種(アデニン、グァニン、シトシン、チミン( ゥラシル))であることを指す。例えば、アデニンを有するリボヌクレオチドに対応する う。また、前記「複数」とは特に制限されないが、好ましくは 2〜5個程度の少数を指す [0076] In addition, in the siRNA of the present invention, not all nucleotides are necessarily ribonucleotides (RNA). That is, in the present invention, one or more ribonucleotides constituting siRNA may be a corresponding deoxyribonucleotide. This “corresponding” refers to the same base species (adenine, guanine, cytosine, thymine (uracil)) although the structures of the sugar moieties are different. For example, it corresponds to a ribonucleotide having an adenine. The “plurality” is not particularly limited, but preferably refers to a small number of about 2 to 5
Yes
[0077] さらに、本発明の上記 RNAを発現し得る DNA (ベクター)もまた、本発明の上述のタ ンパク質をコードする遺伝子の発現を抑制し得る化合物の好ましい態様に含まれる。 例えば、本発明の上記二本鎖 RNAを発現し得る DNA (ベクター)は、該ニ本鎖 RNAの 一方の鎖をコードする DNA、および該ニ本鎖 RNAの他方の鎖をコードする DNAが、 それぞれ発現し得るようにプロモーターと連結した構造を有する DNAである。本発明 の上記 DNAは、当業者においては、一般的な遺伝子工学技術により、適宜作製する こと力 Sできる。より具体的には、本発明の RNAをコードする DNAを公知の種々の発現 ベクターへ適宜揷入することによって、本発明の発現ベクターを作製することが可能 である。 [0077] Furthermore, a DNA (vector) capable of expressing the above RNA of the present invention is also the above-mentioned tag of the present invention. It is contained in the preferable aspect of the compound which can suppress the expression of the gene which codes a protein. For example, the DNA (vector) capable of expressing the double-stranded RNA of the present invention is a DNA encoding one strand of the double-stranded RNA and a DNA encoding the other strand of the double-stranded RNA, Each DNA has a structure linked to a promoter so that it can be expressed. Those skilled in the art can appropriately prepare the above DNA of the present invention by general genetic engineering techniques. More specifically, the expression vector of the present invention can be prepared by appropriately inserting DNA encoding the RNA of the present invention into various known expression vectors.
[0078] また、本発明の発現阻害物質には、上述のコンドロイチン硫酸プロテオダリカンのコ ァタンパク質、合成酵素、脱硫酸化酵素抑制タンパク質、または硫酸基転移酵素を コードする遺伝子の発現調節領域 (例えば、プロモーター領域。具体的な例としては 、 PG-Lbのプロモーター領域である配列番号: 66で表される塩基配列が挙げられる。 )と結合することにより、上述のコンドロイチン硫酸プロテオダリカンのコアタンパク質、 合成酵素、脱硫酸化酵素抑制タンパク質、または硫酸基転移酵素をコードする遺伝 子の発現を阻害する化合物が含まれる。該化合物は、例えば上述のコンドロイチン 硫酸プロテオダリカンのコアタンパク質、合成酵素、脱硫酸化酵素抑制タンパク質、 または硫酸基転移酵素をコードする遺伝子のプロモーター DNA断片を用いて、該 D NA断片との結合活性を指標とするスクリーニング方法により、取得することが可能で ある。また当業者においては、所望の化合物について、上述のコンドロイチン硫酸プ 口テオダリカンのコアタンパク質、合成酵素、脱硫酸化酵素抑制タンパク質、または硫 酸基転移酵素をコードする遺伝子の発現を阻害するか否かの判定を公知の方法、 例えばレポーターアツセィ法等により適宜実施することができる。  [0078] In addition, the expression inhibitory substance of the present invention includes the above-mentioned chondroitin sulfate proteodarican core protein, synthase, desulfase inhibitor protein, or expression regulatory region of a gene encoding a sulfotransferase (for example, Specific examples include the base sequence represented by SEQ ID NO: 66, which is the promoter region of PG-Lb.) By binding to the above-mentioned core protein of chondroitin sulfate proteodarican A compound that inhibits the expression of a gene encoding a synthase, a desulfase inhibitor protein, or a sulfotransferase. The compound is, for example, a promoter DNA fragment of a gene encoding the above chondroitin sulfate proteodarican core protein, synthase, desulfase inhibitor protein, or sulfotransferase, and binding activity to the DNA fragment It can be obtained by a screening method using as an index. In addition, those skilled in the art will determine whether or not the desired compound inhibits the expression of the above-mentioned chondroitin sulfate-teododalican core protein, synthase, desulfase-inhibiting protein, or gene encoding sulfotransferase. The determination can be appropriately carried out by a known method such as a reporter assay method.
[0079] さらに、本発明の上記 RNAを発現し得る DNA (ベクター)もまた、本発明の上述のコ ンドロイチン硫酸プロテオダリカンのコアタンパク質、合成酵素、脱硫酸化酵素抑制タ ンパク質、または硫酸基転移酵素をコードする遺伝子の発現を阻害し得る化合物の 好ましい態様に含まれる。例えば本発明の上記二本鎖 RNAを発現し得る DNA (ベタ ター)は、該ニ本鎖 RNAの一方の鎖をコードする DNA、および該ニ本鎖 RNAの他方 の鎖をコードする DNA力 S、それぞれ発現し得るようにプロモーターと連結した構造を 有する DNAである。本発明の上記 DNAは、当業者においては、一般的な遺伝子ェ 学技術により、適宜作製すること力できる。より具体的には、本発明の RNAをコードす る DNAを公知の種々の発現ベクターへ適宜揷入することによって、本発明の発現べ クタ一を作製することが可能である。 [0079] Furthermore, the DNA (vector) capable of expressing the RNA of the present invention is also the core protein, synthase, desulfase-inhibiting protein, or sulfate group of the above-described chondroitin sulfate proteodarican of the present invention. A preferred embodiment of the compound capable of inhibiting the expression of a gene encoding a transferase is included. For example, the DNA (beta) capable of expressing the double-stranded RNA of the present invention is a DNA that encodes one strand of the double-stranded RNA and a DNA force S that encodes the other strand of the double-stranded RNA. , The structure linked to the promoter so that each can be expressed It has DNA. Those skilled in the art can appropriately prepare the above DNA of the present invention by a general genetic technique. More specifically, the expression vector of the present invention can be prepared by appropriately inserting DNA encoding the RNA of the present invention into various known expression vectors.
[0080] 本発明の上記ベクターの好ましい態様としては、 Versican、 C4ST_1、 C4ST_2、 C4S T-3等の発現を RNAi効果により抑制し得る RNA (siRNA)を発現するベクターを挙げる こと力 Sでさる。 [0080] A preferred embodiment of the vector of the present invention is a vector that expresses RNA (siRNA) capable of suppressing the expression of Versican, C4ST_1, C4ST_2, C4ST-3, and the like by the RNAi effect.
[0081] 上述のコンドロイチン硫酸プロテオダリカンのコアタンパク質、合成酵素、脱硫酸化 酵素抑制化合物、または硫酸基転移酵素に結合する抗体は、当業者に公知の方法 により調製することが可能である。ポリクローナル抗体であれば、例えば、次のように して得ること力 Sできる。天然の上述のタンパク質、あるいは GSTとの融合タンパク質とし て微生物において発現させたリコンビナント(組み換え)タンパク質、またはその部分 ペプチドをゥサギ等の小動物に免疫し血清を得る。これを、例えば、硫安沈殿、プロ ティン A、プロテイン Gカラム、 DEAEイオン交換クロマトグラフィー、上述のコンドロイチ ン硫酸プロテオダリカンのコアタンパク質、合成酵素、脱硫酸化酵素抑制化合物、ま たは硫酸基転移酵素や合成ペプチドをカップリングしたァフィ二ティーカラム等により 精製することにより調製する。また、モノクローナル抗体であれば、例えば上述のコン ドロイチン硫酸プロテオダリカンのコアタンパク質、合成酵素、脱硫酸化酵素抑制化 合物、または硫酸基転移酵素やその部分ペプチドをマウスなどの小動物に免疫を行 い、同マウスより脾臓を摘出し、これをすりつぶして細胞を分離し、該細胞とマウスミエ ローマ細胞とをポリエチレングリコール等の試薬を用いて融合させ、これによりできた 融合細胞(ノヽイブリドーマ)の中から、上述のコンドロイチン硫酸プロテオダリカンのコ ァタンパク質、合成酵素、脱硫酸化酵素抑制化合物、または硫酸基転移酵素に結合 する抗体を産生するクローンを選択する。次いで、得られたハイプリドーマをマウス腹 腔内に移植し、同マウスより腹水を回収し、得られたモノクローナル抗体を、例えば、 硫安沈殿、プロテイン A、プロテイン Gカラム、 DEAEイオン交換クロマトグラフィー、上 述のコンドロイチン硫酸プロテオダリカンのコアタンパク質、合成酵素、脱硫酸化酵素 抑制化合物、または硫酸基転移酵素のタンパク質や合成ペプチドをカップリングした ァフィ二ティーカラム等により精製することで、調製することが可能である。 [0081] The above-described chondroitin sulfate proteodarican core protein, synthetic enzyme, desulfating enzyme inhibitory compound, or antibody that binds to sulfotransferase can be prepared by methods known to those skilled in the art. For example, a polyclonal antibody can be obtained as follows. Serum is obtained by immunizing small animals such as rabbits with recombinant (recombinant) protein expressed in microorganisms as a fusion protein with the above-mentioned natural protein or GST, or a partial peptide thereof. For example, ammonium sulfate precipitation, protein A, protein G column, DEAE ion exchange chromatography, core protein of the above chondroitin sulfate proteodarican, synthase, desulfase inhibitor compound, or sulfate transferase Or by purification using an affinity column coupled with a synthetic peptide. In the case of a monoclonal antibody, for example, the above-mentioned chondroitin sulfate proteodarican core protein, synthetic enzyme, desulfase inhibitor compound, or sulfotransferase or its partial peptide is immunized to a small animal such as a mouse. The spleen is removed from the mouse and ground to separate the cells. The cells are fused with mouse myeloma cells using a reagent such as polyethylene glycol. From the above, a clone producing an antibody that binds to the above chondroitin sulfate proteodarican coprotein, synthase, desulfase inhibitor compound, or sulfotransferase is selected. Next, the obtained hyperidoma was transplanted into the abdominal cavity of the mouse, and ascites was collected from the mouse, and the resulting monoclonal antibody was purified using, for example, ammonium sulfate precipitation, protein A, protein G column, DEAE ion exchange chromatography, Coupling of the chondroitin sulfate proteodarican core protein, synthetic enzyme, desulfase inhibitor compound, or sulfotransferase protein or synthetic peptide described above It can be prepared by purification using an affinity column or the like.
[0082] 本発明の抗体は、本発明の上述のコンドロイチン硫酸プロテオダリカンのコアタンパ ク質、合成酵素、脱硫酸化酵素抑制化合物、または硫酸基転移酵素に結合するもの であれば特に制限はなぐ上記ポリクローナル抗体、モノクローナル抗体のほかにヒト 抗体、遺伝子組み換えによるヒト型化抗体、さらにその抗体断片や抗体修飾物であ つてもよい。 [0082] The antibody of the present invention is not particularly limited as long as it binds to the above-described chondroitin sulfate proteodarican core protein of the present invention, a synthetic enzyme, a desulfase inhibitor compound, or a sulfotransferase. In addition to polyclonal antibodies and monoclonal antibodies, human antibodies, humanized antibodies obtained by genetic recombination, and antibody fragments or modified antibodies thereof may also be used.
[0083] 抗体取得の感作抗原として使用される本発明のタンパク質はその由来となる動物 種について制限されないが、哺乳動物、例えばマウスゃヒト由来のタンパク質が好ま しぐ特にヒト由来のタンパク質が好ましい。ヒト由来のタンパク質は、当業者において は本明細書に開示される遺伝子配列またはアミノ酸配列を用いて適宜取得すること ができる。  [0083] The protein of the present invention used as a sensitizing antigen for obtaining an antibody is not limited with respect to the animal species from which it is derived, but is preferably a protein derived from a mammal such as a mouse, particularly a human-derived protein. . A human-derived protein can be appropriately obtained by those skilled in the art using the gene sequence or amino acid sequence disclosed in the present specification.
[0084] 本発明において、感作抗原として使用されるタンパク質は、完全なタンパク質あるい はタンパク質の部分ペプチドであってもよい。タンパク質の部分ペプチドとしては、例 えば、タンパク質のアミノ基 (N)末端断片やカルボキシ (C)末端断片が挙げられる。 本明細書における「抗体」とはタンパク質の全長または断片に反応する抗体を意味す  [0084] In the present invention, the protein used as the sensitizing antigen may be a complete protein or a partial peptide of the protein. Examples of the partial peptide of the protein include an amino group (N) terminal fragment and a carboxy (C) terminal fragment of the protein. As used herein, “antibody” means an antibody that reacts with the full length or fragment of a protein.
[0085] また、ヒト以外の動物に抗原を免疫して上記ハイプリドーマを得る他に、ヒトリンパ球 、例えば EBウィルスに感染したヒトリンパ球を in vitroでタンパク質、タンパク質発現細 胞またはその溶解物で感作し、感作リンパ球をヒト由来の永久分裂能を有するミエ口 一マ細胞、例えば U266と融合させ、タンパク質への結合活性を有する所望のヒト抗 体を産生するハイプリドーマを得ることもできる。 [0085] In addition to immunizing a non-human animal with an antigen to obtain the above hyperidoma, human lymphocytes such as human lymphocytes infected with EB virus are sensitized with proteins, protein-expressing cells or lysates thereof in vitro. And fusion of sensitized lymphocytes with human-derived permanent mitotic cells, such as U266, to produce a hyperidoma that produces the desired human antibody with protein-binding activity. .
[0086] 本発明の上述のコンドロイチン硫酸プロテオダリカンのコアタンパク質、合成酵素、 脱硫酸化酵素抑制化合物、または硫酸基転移酵素に対する抗体は、該タンパク質と 結合することにより、該タンパク質の発現もしくは機能を阻害する効果が期待される。 得られた抗体を人体に投与する目的 (抗体治療)で使用する場合には、免疫原性を 低下させるため、ヒト抗体やヒト型化抗体が好ましい。  [0086] The above-described chondroitin sulfate proteodarican core protein, synthase, desulfase inhibitor compound, or antibody to sulfate group transferase of the present invention binds to the protein, thereby causing expression or function of the protein. An inhibiting effect is expected. When the obtained antibody is used for the purpose of administering it to the human body (antibody treatment), a human antibody or a humanized antibody is preferable in order to reduce immunogenicity.
[0087] さらに本発明は、上述のコンドロイチン硫酸プロテオダリカンのコアタンパク質、合成 酵素、脱硫酸化酵素抑制化合物、または硫酸基転移酵素の機能を阻害し得る物質 として、上述のコンドロイチン硫酸プロテオダリカンのコアタンパク質、合成酵素、脱硫 酸化酵素抑制化合物、または硫酸基転移酵素に結合する低分子量物質 (低分子化 合物)も含有する。該低分子量物質は、天然または人工の化合物であってもよい。通 常、当業者に公知の方法を用いることによって製造または取得可能な化合物である 。また本発明の化合物は、後述のスクリーニング方法によって、取得することも可能で ある。 [0087] Further, the present invention provides a substance capable of inhibiting the function of the above-mentioned chondroitin sulfate proteodarican core protein, synthase, desulfase inhibitor compound, or sulfotransferase. In addition, it also contains a low molecular weight substance (low molecular weight compound) that binds to the core protein, synthetic enzyme, desulfurizing oxidase inhibiting compound, or sulfotransferase of the chondroitin sulfate proteodarican described above. The low molecular weight substance may be a natural or artificial compound. Usually, it is a compound that can be produced or obtained by using methods known to those skilled in the art. The compound of the present invention can also be obtained by the screening method described later.
[0088] さらに本発明の上述のコンドロイチン硫酸プロテオダリカンのコアタンパク質、合成 酵素、脱硫酸化酵素抑制タンパク質、または硫酸基転移酵素の発現もしくは機能を 阻害し得る物質として、上述のコンドロイチン硫酸プロテオダリカンのコアタンパク質、 合成酵素、脱硫酸化酵素抑制タンパク質、または硫酸基転移酵素に対してドミナント ネガティブの性質を有する変異体(ドミナントネガティブタンパク質)を挙げること力 Sで きる。「コンドロイチン硫酸プロテオダリカンのコアタンパク質、合成酵素、脱硫酸化酵 素抑制タンパク質、または硫酸基転移酵素に対してドミナントネガティブの性質を有 する該タンパク質変異体」とは、コンドロイチン硫酸プロテオダリカンのコアタンパク質 、合成酵素、脱硫酸化酵素抑制タンパク質、または硫酸基転移酵素をコードする遺 伝子を発現させることによって、内在性の野生型タンパク質の活性を消失もしくは低 下させる機能を有するタンパク質を指す。このようなドミナントネガティブタンパク質と しては、例えば、コンドロイチン硫酸との結合を野生型 Versicanコアタンパク質と競合 阻害するような Versicanコアタンパク質変異体を挙げることができる。  [0088] Further, as a substance capable of inhibiting the expression or function of the above-mentioned chondroitin sulfate proteodarican core protein, synthetic enzyme, desulfase inhibitor protein, or sulfotransferase of the present invention, the above-mentioned chondroitin sulfate proteodarican And a mutant having a dominant negative property (dominant negative protein) with respect to a core protein, a synthase, a desulfase inhibitor protein, or a sulfotransferase. “The chondroitin sulfate proteodarican core protein, synthase, desulfurase inhibitor protein, or the protein variant having a dominant negative property to sulfate group” refers to the core of chondroitin sulfate proteodarican. It refers to a protein having a function of eliminating or reducing the activity of an endogenous wild-type protein by expressing a gene encoding a protein, a synthase, a desulfase-inhibiting protein, or a sulfotransferase. Examples of such dominant negative proteins include Versican core protein mutants that competitively inhibit the binding to chondroitin sulfate with the wild-type Versican core protein.
[0089] また本発明において、コンドロイチン硫酸プロテオダリカンの生成もしくは蓄積を阻 害する臓器、組織または細胞は特に限定はされないが、好ましくは神経細胞を含む 臓器、組織であり、より好ましくは脳、脳組織または脳組織における神経細胞である。  [0089] In the present invention, the organ, tissue or cell that inhibits the production or accumulation of chondroitin sulfate proteodarican is not particularly limited, but is preferably an organ or tissue containing nerve cells, more preferably the brain or brain. A nerve cell in tissue or brain tissue.
[0090] コンドロイチン硫酸プロテオダリカンの生成もしくは蓄積を阻害する化合物は、神経 線維性変性疾患の治療または予防のための薬剤となることが期待される。ここで「治 療または予防」は、神経線維性変性を呈する臓器、組織、または細胞に対して、必ず しも完全な治療効果または予防効果を有する必要はなぐ部分的な効果を有する場 合であってよい。  [0090] A compound that inhibits the production or accumulation of chondroitin sulfate proteodarican is expected to be a drug for the treatment or prevention of neurofibrotic degenerative diseases. Here, “treatment or prevention” refers to a case where it has a partial effect that is not necessarily required to have a complete therapeutic or preventive effect on an organ, tissue, or cell that exhibits neurofibrotic degeneration. It may be.
[0091] 本発明にお!/、て神経線維性変性疾患は、神経線維性変性を伴う疾患であれば特 に限定はされな!/、が、好ましくは脳神経線維性変性疾患であり、より好ましくは脳脊 髄神経線維性変性疾患または末梢神経線維性変性疾患であり、さらに好ましくはパ 一キンソン病、アルツハイマー病、筋萎縮性側索硬化症、ポリグルタミン病、脊髄性 筋萎縮症、ハンチントン病、または多発性硬化症等を挙げることができる。 [0091] In the present invention, a neurofibrotic degenerative disease is a special disease as long as it is a disease accompanied by neurofibrotic degeneration. Is preferably a cranial nerve fibrotic degenerative disease, more preferably a cerebrospinal nerve fibrotic degenerative disease or a peripheral nerve fibrotic degenerative disease, more preferably Parkinson's disease, Alzheimer's Disease, amyotrophic lateral sclerosis, polyglutamine disease, spinal muscular atrophy, Huntington's disease, or multiple sclerosis.
[0092] 本発明の神経線維性変性抑制剤は、神経線維性変性の原因であるコンドロイチン 硫酸プロテオダリカンの生成もしくは蓄積を阻害することにより神経線維性変性を抑 制する作用を有する。従って、本発明の好ましい態様としては、例えば、本発明の神 経線維性変性抑制剤を有効成分とする、脳神経線維性変性疾患治療剤、脳脊髄神 経線維性変性疾患治療剤、末梢神経線維性変性疾患治療剤、パーキンソン病治療 剤、アルツハイマー病治療剤、筋萎縮性側索硬化症治療剤、ポリグルタミン病治療 剤、脊髄性筋萎縮症治療剤、ハンチントン病治療剤、または多発性硬化症治療剤を 提供する。 [0092] The neurofibrotic degeneration inhibitor of the present invention has an action of suppressing neurofibrotic degeneration by inhibiting the production or accumulation of chondroitin sulfate proteodarican which is the cause of neurofibrotic degeneration. Accordingly, preferred embodiments of the present invention include, for example, a therapeutic agent for cranial nerve fibrotic degenerative disease, a therapeutic agent for cerebrospinal neurofibrotic degenerative disease, a peripheral nerve fiber containing the neurofibrotic degeneration inhibitor of the present invention as an active ingredient. Sexual degenerative disease treatment agent, Parkinson's disease treatment agent, Alzheimer's disease treatment agent, amyotrophic lateral sclerosis treatment agent, polyglutamine disease treatment agent, spinal muscular atrophy treatment agent, Huntington's disease treatment agent, or multiple sclerosis Provide therapeutic agents.
[0093] また本発明の「神経線維性変性抑制剤」は、「神経線維性変性治療剤」、「神経線 維性変性阻害剤」、または「神経線維性変性改善剤」等と表現することも可能である。 また、本発明において「抑制剤」は、「医薬品」、「医薬組成物」、「治療用医薬」等と表 現することあでさる。  In addition, the “nerve fibrotic degeneration inhibitor” of the present invention is expressed as a “nerve fibrotic degeneration therapeutic agent”, a “nerve fibrotic degeneration inhibitor”, a “nerve fibrotic degeneration improving agent” or the like. Is also possible. Further, in the present invention, the “suppressor” is expressed as “medicine”, “pharmaceutical composition”, “therapeutic drug” or the like.
[0094] なお、本発明における「治療」には、神経線維性変性の発生を予め抑制し得る予防 的な効果も含まれる。また、神経線維性変性発現細胞 (組織)に対して、必ずしも、完 全な治療効果を有する場合に限定されず、部分的な効果を有する場合であってもよ い。  It should be noted that “treatment” in the present invention includes a prophylactic effect capable of previously suppressing the occurrence of neurofibrotic degeneration. In addition, it is not necessarily limited to a case having a complete therapeutic effect on cells (tissues) expressing neurofibrotic degeneration, and may have a partial effect.
[0095] 本発明の薬剤は、生理学的に許容される担体、賦形剤、ある!/、は希釈剤等と混合 し、医薬組成物として経口、あるいは非経口的に投与することができる。経口剤として は、顆粒剤、散剤、錠剤、カプセル剤、溶剤、乳剤、あるいは懸濁剤等の剤型とする ことができる。非経口剤としては、注射剤、点滴剤、外用薬剤、吸入剤(ネブライザ一 )あるいは座剤等の剤型を選択することができる。注射剤には、頭蓋内投与注射剤、 鼻腔内投与注射剤、皮下注射剤、筋肉注射剤、あるいは腹腔内注射剤等を示すこと ができる。外用薬剤には、経鼻投与剤、あるいは軟膏剤等を示すことができる。主成 分である本発明の薬剤を含むように、上記の剤型とする製剤技術は公知である。 [0096] 例えば、経口投与用の錠剤は、本発明の薬剤に賦形剤、崩壊剤、結合剤、および 滑沢剤等を加えて混合し、圧縮整形することにより製造することができる。賦形剤に は、乳糖、デンプン、あるいはマンニトール等が一般に用いられる。崩壊剤としては、 炭酸カルシウムやカルボキシメチルセルロースカルシウム等が一般に用いられる。結 合剤には、アラビアゴム、カルボキシメチルセルロース、あるいはポリビュルピロリドン が用いられる。滑沢剤としては、タルクゃステアリン酸マグネシウム等が公知である。 [0095] The drug of the present invention can be mixed with a physiologically acceptable carrier, excipient, certain! /, Diluent, etc. and administered orally or parenterally as a pharmaceutical composition. As oral preparations, dosage forms such as granules, powders, tablets, capsules, solvents, emulsions or suspensions can be used. As parenteral agents, dosage forms such as injections, infusions, drugs for external use, inhalants (nebulizers) or suppositories can be selected. Examples of the injection include intracranial injection, intranasal injection, subcutaneous injection, intramuscular injection, and intraperitoneal injection. Examples of the medicine for external use include a nasal administration agent or an ointment. The preparation technique for the above dosage form so as to include the drug of the present invention which is the main component is known. [0096] For example, a tablet for oral administration can be produced by adding an excipient, a disintegrant, a binder, a lubricant and the like to the drug of the present invention, mixing, and compressing and shaping. As the excipient, lactose, starch, mannitol or the like is generally used. As the disintegrant, calcium carbonate or carboxymethyl cellulose calcium is generally used. As the binder, gum arabic, carboxymethyl cellulose, or polybulur pyrrolidone is used. As the lubricant, talc magnesium stearate and the like are known.
[0097] 本発明の薬剤を含む錠剤は、マスキングや、腸溶性製剤とするために、公知のコー ティングを施すこと力できる。コーティング剤には、ェチルセルロースやポリオキシェ チレングリコール等を用いることができる。  [0097] The tablet containing the drug of the present invention can be subjected to a known coating for masking or enteric preparation. As the coating agent, ethyl cellulose, polyoxyethylene glycol or the like can be used.
[0098] また注射剤は、主成分である本発明の薬剤を適当な分散剤とともに溶解、分散媒 に溶解、あるいは分散させることにより得ること力 Sできる。分散媒の選択により、水性溶 剤と油性溶剤のいずれの剤型とすることもできる。水性溶剤とするには、蒸留水、生 理食塩水、あるいはリンゲル液等を分散媒とする。油性溶剤では、各種植物油ゃプ ロピレングリコール等を分散媒に利用する。このとき、必要に応じてパラベン等の保存 剤を添加することもできる。また注射剤中には、塩化ナトリウムゃブドウ糖等の公知の 等張化剤を加えることができる。更に、塩化ベンザルコニゥムゃ塩酸プロ力インのよう な無痛化剤を添加することができる。  [0098] The injection can be obtained by dissolving the agent of the present invention as a main component together with an appropriate dispersant, or dissolving or dispersing in a dispersion medium. Depending on the choice of the dispersion medium, either aqueous solvent or oil-based solvent can be used. In order to use an aqueous solvent, distilled water, physiological saline, Ringer's solution, or the like is used as a dispersion medium. For oil-based solvents, various vegetable oils such as propylene glycol are used as dispersion media. At this time, a preservative such as paraben can be added as necessary. In addition, known isotonic agents such as sodium chloride and glucose can be added to the injection. Furthermore, a soothing agent such as benzalkonium chloride pro-hydrochloride can be added.
[0099] また、本発明の薬剤を固形、液状、あるいは半固形状の組成物とすることにより外 用剤とすることができる。固形、あるいは液状の組成物については、先に述べたもの と同様の組成物とすることで外用剤とすることができる。半固形状の組成物は、適当 な溶剤に必要に応じて増粘剤を加えて調製することができる。溶剤には、水、ェチル アルコール、あるいはポリエチレングリコール等を用いることができる。増粘剤には、 一般にベントナイト、ポリビュルアルコール、アクリル酸、メタクリル酸、あるいはポリビ ニルピロリドン等が用いられる。この組成物には、塩化ベンザルコニゥム等の保存剤 を加えること力 Sできる。また、担体としてカカオ脂のような油性基材、あるいはセルロー ス誘導体のような水性ゲル基材を組み合わせることにより、座剤とすることもできる。  [0099] Further, an external preparation can be obtained by making the agent of the present invention into a solid, liquid, or semi-solid composition. About a solid or liquid composition, it can be set as an external preparation by setting it as the composition similar to what was described previously. A semi-solid composition can be prepared by adding a thickener to an appropriate solvent as required. As the solvent, water, ethyl alcohol, polyethylene glycol, or the like can be used. As the thickener, bentonite, polybutyl alcohol, acrylic acid, methacrylic acid, or polyvinyl pyrrolidone is generally used. This composition can be added with a preservative such as benzalkonium chloride. A suppository can also be obtained by combining an oily base material such as cacao butter or an aqueous gel base material such as cellulose derivative as a carrier.
[0100] 本発明の薬剤を遺伝子治療剤として使用する場合は、本発明の薬剤を注射により 直接投与する方法のほか、核酸が組込まれたベクターを投与する方法が挙げられる 。上記ベクターとしては、アデノウイルスベクター、アデノ随伴ウィルスベクター、ヘル ぺスゥイノレスベクター、ワクシニアウイノレスベタター、レトロウイノレスベタター、レンチウ ィルスべクタ一等が挙げられ、これらのウィルスベクターを用いることにより効率よく投 与すること力 Sでさる。 [0100] When the agent of the present invention is used as a gene therapy agent, in addition to the method of directly administering the agent of the present invention by injection, a method of administering a vector incorporating a nucleic acid can be mentioned. . Examples of the above-mentioned vectors include adenovirus vectors, adeno-associated virus vectors, herpes vinores vectors, vaccinia winores betaters, retro winores betaters, and lentivirus vectors. Throw well with the power S.
[0101] また、本発明の薬剤をリボソームなどのリン脂質小胞体に導入し、その小胞体を投 与することも可能である。 siRNAや shRNAを保持させた小胞体をリボフヱクシヨン法に より所定の細胞に導入する。そして、得られる細胞を例えば静脈内、動脈内等に全身 投与する。神経線維性変性組織等に局所的に投与することもできる。 siRNAに関して 、 in vitroにおいては非常に優れた特異的転写後抑制効果を示す力 in vivoにおい ては血清中のヌクレアーゼ活性により速やかに分解されてしまう。それゆえ、生体内 にて持続時間が限られることが問題となり最適で効果的なデリバリーシステム開発が 求められてきた。一つの例として、 Ochiya, Tら(Ochiya,T et al. Nat. Med. (1999) 5 7 07-710; Ochiya,T et al. Curr.Gene Ther. (2001) 1 31-52)により生体親和性材料で あるァテロコラーゲンが核酸と混合し複合体を形成させると、生体中の分解酵素から 核酸を保護する作用があり siRNAのキャリア一として非常に適しているキャリアーであ ると報告されている。また別の例として、直接脳内にリボソームやウィルスを用いたデ リバリーシステム等が開発されている(Xia, H et al. Nat. Biotechnol. (2002) 20 1006- 1010; Brummelkamp, T. R. et al. Cancer Cell (2002) 2 243-247; Barton, G.M. and Medzhitov, R. Proc. Natl. Acad. Sci. USA, (2002) 99 14943-14945; Abbas- Terki, T • et al. Hum. Gene. Ther. (2002) 13 2197-2201)。そして、上記の方法等により siRNA が神経線維性変性疾患の遺伝子治療薬に向けて開発されて!/、る。その例として BAC El (Beta-site APP Cleaving Enzyme: β— secretase)の siRNA力、 in vivoのトフンスシェ ニックマウスを用いてアミロイドの形成を減少させるために開発された。デリバリーシス テムとしてはその BACE1 siRNAをレンチウィルスのベクターに組み込み脳内で発現 させる系にて報告されている(Singer et al. Nat. Neurosci. (2005) 8(10) 1343-1349)。 これらはいずれも例示であり、本発明におけるデリバリー方法はこれらに限られない。  [0101] It is also possible to introduce the drug of the present invention into a phospholipid vesicle such as a ribosome and administer the vesicle. The endoplasmic reticulum retaining siRNA or shRNA is introduced into a predetermined cell by the ribofusion method. The obtained cells are then administered systemically, for example, intravenously or intraarterially. It can also be administered locally to neurofibrous degenerated tissue or the like. With respect to siRNA, it has a very excellent specific post-transcriptional inhibitory effect in vitro. In vivo, it is rapidly degraded by nuclease activity in serum. Therefore, the limited duration in vivo has become a problem, and the development of optimal and effective delivery systems has been demanded. As an example, Ochiya, T et al. (Ochiya, T et al. Nat. Med. (1999) 5 7 07-710; Ochiya, T et al. Curr. Gene Ther. (2001) 1 31-52) It has been reported that when atelocollagen, an affinity material, is mixed with nucleic acid to form a complex, it protects the nucleic acid from degrading enzymes in the living body and is a very suitable carrier for siRNA. Yes. As another example, a delivery system using ribosomes and viruses directly in the brain has been developed (Xia, H et al. Nat. Biotechnol. (2002) 20 1006- 1010; Brummelkamp, TR et al. Cancer Cell (2002) 2 243-247; Barton, GM and Medzhitov, R. Proc. Natl. Acad. Sci. USA, (2002) 99 14943-14945; Abbas- Terki, T • et al. Hum. Gene. Ther (2002) 13 2197-2201). Then, siRNA has been developed for gene therapy for neurofibrotic degenerative diseases by the above method! For example, BAC El (Beta-site APP Cleaving Enzyme: β-secretase) siRNA was developed to reduce amyloid formation using in vivo tofence nick mice. A delivery system has been reported in which the BACE1 siRNA is incorporated into a lentiviral vector and expressed in the brain (Singer et al. Nat. Neurosci. (2005) 8 (10) 1343-1349). These are only examples, and the delivery method in the present invention is not limited to these.
[0102] 本発明の薬剤は、安全とされている投与量の範囲内において、ヒトを含む哺乳動物 に対して、必要量 (有効量)が投与される。本発明の薬剤の投与量は、剤型の種類、 投与方法、患者の年齢や体重、患者の症状等を考慮して、最終的には医師または 獣医師の判断により適宜決定することができる。一例を示せば、年齢、性別、症状、 投与経路、投与回数、剤型によって異なる力 例えばアデノウイルスの場合の投与量 は 1日 1回あたり 106〜1013個程度であり、 1週〜 8週間隔で投与される。 [0102] The necessary amount (effective amount) of the drug of the present invention is administered to mammals including humans within the safe dose range. The dosage of the drug of the present invention is the type of dosage form, In consideration of the administration method, the patient's age and weight, the patient's symptoms, etc., the final decision can be made as appropriate based on the judgment of a doctor or veterinarian. For example, the power varies depending on age, sex, symptoms, administration route, number of administrations, and dosage forms.For example, the dose in the case of adenovirus is about 10 6 to 10 13 per day, 1 week to 8 It is administered at weekly intervals.
[0103] また、 siRNAまたは shRNAを目的の組織または器官に導入するために、市販の遺伝 子導入キット(例えばアデノエクスプレス:クローンテック社)を用いることもできる。  [0103] In order to introduce siRNA or shRNA into a target tissue or organ, a commercially available gene introduction kit (for example, Adeno Express: Clontech) can also be used.
[0104] 本発明の薬剤を使用する場合は、神経線維性変性を発現する疾患であれば適用 部位もしくは疾患の種類は特に限定されず、例えば脳脊髄神経線維性変性疾患、 末梢神経線維性変性疾患、パーキンソン病、アルツハイマー病、筋萎縮性側索硬化 症、ポリグルタミン病、脊髄性筋萎縮症、ハンチントン病、または多発性硬化症等を 対象として適用される。上記疾患は、他の疾患と併発したものであってもよい。  [0104] When the agent of the present invention is used, the application site or the type of the disease is not particularly limited as long as it is a disease that expresses neurofibrotic degeneration. For example, cerebrospinal nerve fibrotic degenerative disease, peripheral nerve fibrotic degeneration Applicable for diseases, Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, polyglutamine disease, spinal muscular atrophy, Huntington's disease, or multiple sclerosis. The above-mentioned diseases may be accompanied with other diseases.
[0105] また本発明は、被検試料からコンドロイチン硫酸プロテオダリカンの生成もしくは蓄 積を阻害する作用を有する物質を選択することを特徴とする神経線維性変性抑制剤 のスクリーニング方法を提供する。本発明のスクリーニング方法によって、神経線維 性変性抑制剤もしくは神経線維性変性抑制剤のための候補化合物を効率的に取得 すること力 Sでさる。  [0105] The present invention also provides a screening method for a neurofibrotic degeneration inhibitor, which comprises selecting a substance having an action of inhibiting the production or accumulation of chondroitin sulfate proteodarican from a test sample. With the screening method of the present invention, it is possible to efficiently obtain a neurofibrous degeneration inhibitor or a candidate compound for a neurofibrous degeneration inhibitor.
[0106] 本発明のスクリーニング方法の好ましい態様は、以下の(a)〜(d)のいずれかに記 載の作用を有する物質を選択する工程を含む、神経線維性変性抑制剤のスクリー二 ング方法である。  [0106] A preferred embodiment of the screening method of the present invention includes screening a neurofibrotic degeneration inhibitor comprising the step of selecting a substance having the action described in any of the following (a) to (d): Is the method.
(a)コンドロイチン硫酸プロテオダリカンの分解促進作用 ( a ) Promoting the degradation of chondroitin sulfate proteodarican
(b)コンドロイチン硫酸プロテオダリカンの合成阻害作用  (b) Inhibition of chondroitin sulfate proteodarican synthesis
(c)コンドロイチン硫酸プロテオダリカンの脱硫酸化作用  (c) Desulfation effect of chondroitin sulfate proteodarican
(d)コンドロイチン硫酸プロテオダリカンの硫酸化阻害作用  (d) Inhibitory effect of chondroitin sulfate proteodarican on sulfation
[0107] これらに共通したスクリーニングの基本的な原理として、代表的な例として、下記の 工程を含むものが挙げられる。  [0107] As a basic principle of screening common to these, a typical example includes one including the following steps.
(1)コンドロイチン硫酸プロテオダリカン(CSPG)そのもの/もしくはグリコサミノグリカ ン(GAG)鎖/もしくは CSPGや GAG鎖を合成(生成)する細胞  (1) Cells that synthesize (generate) chondroitin sulfate proteodarican (CSPG) itself / or glycosaminoglycan (GAG) chain / or CSPG or GAG chain
(2)被検化合物 (例えば、製薬企業の有する莫大な化合物ライブラリー) (3)コンドロイチン硫酸プロテオダリカン(CSPG)の切断断面/コンドロイチン硫酸プ 口テオダリカン (CSPG)量/遊離グリコサミノダリカン (GAG)量を検出する方法 上記 3種のツールを用いる。 (1)と(2)を試験管内、もしくは培養皿上で混合させ、そ の効果を(3)により簡便に検出すると!/、う手順が望まし!/、。 (2) Test compounds (for example, huge compound libraries owned by pharmaceutical companies) (3) Method for detecting cut cross section of chondroitin sulfate proteodarican (CSPG) / chondroitin sulfate proteodarican (CSPG) / free glycosaminodarlican (GAG) The above three tools are used. If (1) and (2) are mixed in a test tube or on a culture dish, and the effect is detected easily by (3)!
[0108] 以下、本発明のスクリーニング方法の態様を例示する。なお、以下に記載の態様に おいては、用いられるコンドロイチン硫酸プロテオダリカン、合成酵素、脱硫酸化酵素 抑制化合物、硫酸基転移酵素、分解促進酵素、脱硫酸化酵素の由来としては、ヒト、 マウス、ラット等に由来するものが挙げられる力 これらに由来するものに特に制限さ れない。コンドロイチン硫酸プロテオダリカンの一部とは、グリコサミノダリカン鎖、コア タンパク質などの構成要素またはその一部であり、特に限定されない。  [0108] Hereinafter, embodiments of the screening method of the present invention will be exemplified. In the embodiment described below, the chondroitin sulfate proteodarican, synthase, desulfase inhibitor compound, sulfate transferase, degradation promoting enzyme, and desulfase used are derived from human, mouse, Forces derived from rats and the like are not particularly limited to those derived from these. The part of chondroitin sulfate proteodalycan is a component such as a glycosaminodarican chain, a core protein, or a part thereof, and is not particularly limited.
[0109] また以下に記載の態様に用いる被検化合物としては、特に制限されないが、例え ば、天然化合物、有機化合物、無機化合物、タンパク質、ペプチドなどの単一化合 物、並びに、化合物ライブラリー、遺伝子ライブラリーの発現産物、細胞抽出物、細胞 培養上清、発酵微生物産生物、海洋生物抽出物、植物抽出物等が挙げられる。  [0109] The test compound used in the embodiments described below is not particularly limited. For example, natural compounds, organic compounds, inorganic compounds, proteins, peptides and other single compounds, compound libraries, Examples include gene library expression products, cell extracts, cell culture supernatants, fermented microorganism products, marine organism extracts, plant extracts, and the like.
[0110] また以下に記載の態様における被検化合物への「接触」は、通常、コンドロイチン硫 酸プロテオダリカン、その一部、合成酵素、脱硫酸化酵素抑制化合物、硫酸基転移 酵素、分解促進酵素、または脱硫酸化酵素を被検化合物と混合することによって行う 、この方法に限定されない。例えば、これらのタンパク質またはその一部を発現す る細胞を被検化合物と接触させることにより、上記「接触」を行うことができる。  [0110] In addition, the "contact" to the test compound in the embodiment described below is usually chondroitin sulfate proteodarican, a part thereof, a synthetic enzyme, a desulfase inhibitor compound, a sulfotransferase, a degradation promoting enzyme. However, the method is not limited to this method, which is performed by mixing a desulfating enzyme with a test compound. For example, the above “contact” can be performed by contacting a cell expressing these proteins or a part thereof with a test compound.
[0111] また以下に記載の態様における「細胞」の由来としては、ヒト、マウス、ラット等に由 来する細胞が挙げられる力 これらに由来する細胞に特に制限されず、それぞれの 態様において用いられるタンパク質を発現するように形質転換された大腸菌、酵母 等の微生物細胞を利用することも可能である。例えば、「コンドロイチン硫酸プロテオ ダリカンを発現する細胞」としては、内在性のコンドロイチン硫酸プロテオダリカン遺伝 子を発現して!/、る細胞、または外来性のコンドロイチン硫酸プロテオダリカン遺伝子 が導入され、該遺伝子が発現している細胞を利用することができる。外来性のコンド ロイチン硫酸プロテオダリカン遺伝子が発現した細胞は、通常コンドロイチン硫酸プロ テオダリカン遺伝子が揷入された発現ベクターを宿主細胞へ導入することにより作製 すること力 Sできる。該発現べクタ一は、一般的な遺伝子工学技術によって作製するこ と力 Sできる。 [0111] The origin of the "cell" in the embodiments described below is not particularly limited to cells derived from humans, mice, rats, etc., and is used in each embodiment. It is also possible to use microbial cells such as Escherichia coli and yeast transformed to express the protein. For example, a cell expressing a chondroitin sulfate proteodarican can be expressed as a cell that expresses an endogenous chondroitin sulfate proteodarican gene or an exogenous chondroitin sulfate proteodarican gene, Cells in which the gene is expressed can be used. Cells expressing the exogenous chondroitin sulfate proteodarican gene are usually created by introducing an expression vector containing the chondroitin sulfate proteodarican gene into the host cell. The power to do S. The expression vector can be produced by general genetic engineering techniques.
[0112] また以下の記載において、「コンドロイチン硫酸プロテオダリカンコアタンパク質」と (ま、 列えは、 matrix typeコンドロ チン硫酸プロテオグリカンでめれは、 aggrican、 vers ican、 neurocan, brevicanなどのコアタンパク質、また膜型コンドロイチン硫酸プロテオ グリカンであれば、例えば Decorin、 Biglycan, Fibromodulin、 PG_Lbなどのコアタンパ ク質である。また「合成酵素」は、例えば、 GalNAc4ST_l、 GalNAc4ST_2、 GALNAC4S — 6ST、 UA20ST、 GalT-I、 GalT_H、 GlcAT_I、 XylosylTなどである。また「硫酸基転移 酵素」は、例えば、 C4ST-1 (Chondroitin D—N— acetylgalactosamine— 4—0— sulfotransfe rase 1)、 し 4S丄し2 (Cnonaroitin D-N- acetylgalactosamine- 4-0- sulfotransferase 2)、 C4ST-3 (Chondroitin D_N_acetylgalactosamine_4_0_sulfotransferase 3)、 D4ST、 C6 ST-1、 C6ST-2などである。また、「分解促進酵素」とは、例えば、 ADAMTS-1 , ADAM TS- 4、 ADAMTS- 5、 Chondroitinase ABC (ChABC)、 Chondroitinase AC、 Chondroiti nase B、 Calpain Iなどである。また「脱硫酸化酵素」は、例えば、 Chondroitin-4-sulfat ase、 Chondroitm_6_sulmtaseなと(?める。  [0112] In addition, in the following description, “chondroitin sulfate proteodarican core protein” (the matrix is chondroitin sulfate proteoglycan core, and the matrix type is chondroitin sulfate proteoglycan core protein such as aggrican, vers ican, neurocan, brevican, etc. In addition, membrane-type chondroitin sulfate proteoglycans are core proteins such as Decorin, Biglycan, Fibromodulin, and PG_Lb, and “synthetic enzymes” include, for example, GalNAc4ST_l, GalNAc4ST_2, GALNAC4S-6ST, UA20ST, GalT-I, GalT_H, GlcAT_I, XylosylT, etc. “Sulfyltransferase” includes, for example, C4ST-1 (Chondroitin D—N—acetylgalactosamine—4—0—sulfotransferase 1), 4S 丄 2 (Cnonaroitin DN-acetylgalactosamine) -4-0- sulfotransferase 2), C4ST-3 (Chondroitin D_N_acetylgalactosamine_4_0_sulfotransferase 3), D4ST, C6 ST-1, C6ST-2, etc. Also, "degradation promoting enzyme" Are, for example, ADAMTS-1, ADAM TS-4, ADAMTS-5, Chondroitinase ABC (ChABC), Chondroitinase AC, Chondroitinase B, Calpain I, etc. Also, “desulfating enzyme” is, for example, Chondroitin-4- sulfatase, Chondroitm_6_sulmtase (?
[0113] 本発明のスクリーニング方法の態様として、コンドロイチン硫酸プロテオダリカンの分 解促進作用を有する化合物を選択する工程を含む方法を挙げることができる。本発 明の上記方法は例えば以下の工程からなる。  [0113] As an embodiment of the screening method of the present invention, there can be mentioned a method comprising a step of selecting a compound having an action of promoting the degradation of chondroitin sulfate proteodarican. The above method of the present invention includes the following steps, for example.
(a)コンドロイチン硫酸プロテオダリカンまたはその一部と被検化合物を接触させるェ 程 ( a ) The process of contacting the test compound with chondroitin sulfate proteodarican or a part thereof
(b)コンドロイチン硫酸プロテオダリカンまたはその一部の存在量を測定する工程 (b) a step of measuring the abundance of chondroitin sulfate proteodarican or a part thereof
(c)被検化合物の非存在下において測定した場合と比較して、存在量を低下させる 物質を選択する工程 (c) A step of selecting a substance that reduces the abundance compared to the case where measurement is performed in the absence of the test compound.
[0114] 上記方法においてはまず、コンドロイチン硫酸プロテオダリカンまたはその一部に 被検化合物を接触させる。  [0114] In the above method, first, a test compound is brought into contact with chondroitin sulfate proteodarican or a part thereof.
[0115] 本方法においては次いで、コンドロイチン硫酸プロテオダリカンまたはその一部の 量を測定する。測定は、当業者に公知の方法によって行うことができる。例えば、コン ドロイチン硫酸プロテオダリカンまたはその一部に結合する標識された化合物または 抗体を用い、標識量を測定することにより検出することができる。また、クロマトグラフィ 一法や質量分析法などを用いて検出することもできる。 [0115] Next, in the present method, the amount of chondroitin sulfate proteodarican or a part thereof is measured. The measurement can be performed by methods known to those skilled in the art. For example, a labeled compound that binds to chondroitin sulfate proteodarican or a part thereof, or It can be detected by measuring the amount of label using an antibody. It can also be detected using a chromatographic method or mass spectrometry.
[0116] 本方法においては、次いで、被検化合物を接触させない場合(対照)と比較して、 該コンドロイチン硫酸プロテオダリカンまたはその一部の存在量を低下させる化合物 を選択する。低下させる化合物は神経線維性変性治療のための薬剤となる。 [0116] In this method, a compound that reduces the abundance of the chondroitin sulfate proteodarican or a part thereof is then selected as compared with the case where the test compound is not contacted (control). The compound that lowers becomes a drug for treating neurofibrotic degeneration.
[0117] 被検化合物が上記 (a)分解促進作用の活性を有して!/、るか否かにつ!/、て評価 (測 定)可能な方法、具体例の簡単な一例を以下に示す。 [0117] A simple example of a method and a specific example that can be evaluated (measured) according to whether the test compound has the above activity (a) has the activity of promoting decomposition! Shown in
[0118] 上記 (a)コンドロイチン硫酸プロテオダリカンの分解促進作用に関するスクリーニング 方法態様: [0118] (a) Screening method for promoting the degradation of chondroitin sulfate proteodarican
CS-GAGとして、コンドロイチン硫酸 A(CS_A)、 CS_B、 CS-C (生化学工業社、 ICN 社、 Sigma社など)、ヒト由来プロテオダリカン(BGN社、 ISL社など)などを準備し、 96 穴プレートに 10 g/mLの濃度でコーティングする(Kawashima H et al.; J. Biol. Che m. 277: 12921-12930, 2002.など、既知の方法による)。本プレートの各ゥエルに各種 の被検化合物を添加し、 37°Cで 2時間反応後に CS-GAGの変化を検出する。  Prepare CS-GAG such as chondroitin sulfate A (CS_A), CS_B, CS-C (Seikagaku Corporation, ICN, Sigma, etc.), human-derived proteodalycan (BGN, ISL, etc.), etc. The well plate is coated at a concentration of 10 g / mL (Kawashima H et al .; J. Biol. Chem. 277: 12921-12930, 2002. etc.). Add various test compounds to each well of this plate and detect CS-GAG change after 2 hours reaction at 37 ° C.
[0119] 検出方法としては、例えば WFAレクチン (ノダフジレクチン)結合法が簡便な手法と して挙げられる。 WFAレクチンは CS-GAG鎖の GalNAc残基に結合するため、 CS-GA Gを簡便に検出できる。被検化合物の陽性コントロールとしてはコンドロイチナーゼ A BCを使用する。コンドロイチナーゼ ABC添加により、 CS-GAG鎖が分解されると WFA レクチンが結合できなくなるため、その原理を利用する。より具体的には、 FITC標識 WFAレクチン (EY社など)を、被検化合物混合前後で CSコーティング'ゥエルに添カロ し、 CS-GAGが分解される事により、ゥエル中の FITC蛍光強度の変化を蛍光プレート リーダーあるいは蛍光顕微鏡などの検出機器により極めて簡便に定量 ·数値化でき る。混合前後で最も蛍光数値を減少させた化合物が、本コンセプトを満たす新規の 治療候補化合物として判定できる。  [0119] As a detection method, for example, a WFA lectin (Nodafuji lectin) binding method can be mentioned as a simple method. Since WFA lectin binds to the GalNAc residue of CS-GAG chain, CS-GAG can be easily detected. Chondroitinase ABC is used as a positive control for the test compound. When CS-GAG chain is degraded by chondroitinase ABC addition, WFA lectin cannot be bound, so use that principle. More specifically, FITC-labeled WFA lectin (such as EY) is added to the CS coating well before and after mixing the test compound, and the CS-GAG is decomposed to change the FITC fluorescence intensity in the well. Can be quantified and quantified very easily using a fluorescence plate reader or a fluorescence microscope. The compound with the lowest fluorescence value before and after mixing can be judged as a new treatment candidate compound that satisfies this concept.
[0120] また、他の検出方法として、 CS-GAGそのものを直接的に標識する抗 CS抗体(クロ ーン: CS56、生化学工業社製)を使用する事ができる。 WFAレクチンと同様に、 FITC 標識抗 CS抗体を CSコーティング'ゥエルに添加する事で、蛍光数値の変化を見れば 、極めて短時間かつ簡便に大量スクリーニングができる。 [0121] より詳細な検出方法として、被検化合物混合前後のプレートをそのまま使用し、 sGA G Assay Kit (WIESLAB社製)、 Sulphanated Glycosaminoglycans, ELISA Kit (FUNAK OSHI社製)などを適用することにより、 GAG含有量を正確に定量 ·数値化する方法が ある。 [0120] Further, as another detection method, an anti-CS antibody (clone: CS56, manufactured by Seikagaku Corporation) that directly labels CS-GAG itself can be used. As with the WFA lectin, FITC-labeled anti-CS antibody can be added to CS-coated wells so that mass screening can be performed in a very short time and simply if changes in fluorescence values are observed. [0121] As a more detailed detection method, by using the plate before and after mixing the test compound as it is, and applying sGA G Assay Kit (manufactured by WIESLAB), Sulphanated Glycosaminoglycans, ELISA Kit (manufactured by FUNAK OSHI), etc. There is a method to accurately quantify and quantify the GAG content.
[0122] さらに詳細には、被検化合物混合前後のプレートに 2-AB (2-aminobenzamide)や 2- AP (2-aminopyridine; V、ずれも LUD社製など)を添加することにより、遊離 GAG鎖の 還元末端を簡便に蛍光標識し、糖鎖の各タイプや、各タイプの含有率までを、 HPLC 、 MALDI-MS, LC-MSなどで解析する事により、より詳細な解析が可能である。候補 化合物の特性を詳細に調べるという、スクリーニングの次の段階の方法である。  [0122] More specifically, free GAG is obtained by adding 2-AB (2-aminobenzamide) or 2-AP (2-aminopyridine; V, manufactured by LUD, etc.) to the plate before and after the test compound is mixed. More detailed analysis is possible by simply fluorescently labeling the reducing end of the chain and analyzing each type of sugar chain and the content of each type by HPLC, MALDI-MS, LC-MS, etc. . This is a method for the next stage of screening in which the properties of candidate compounds are examined in detail.
[0123] 本発明のスクリーニング方法の他の態様としては、コンドロイチン硫酸プロテオダリ カンの合成阻害作用を有する物質を選択する工程を含む方法を挙げることができる 。本発明の上記方法は例えば以下の工程からなる。  [0123] As another embodiment of the screening method of the present invention, there may be mentioned a method comprising a step of selecting a substance having an action of inhibiting chondroitin sulfate proteodarican synthesis. The above method of the present invention comprises, for example, the following steps.
(a)コンドロイチン硫酸プロテオダリカンまたはその一部を発現する細胞、該細胞抽 出液、もしくはコンドロイチン硫酸プロテオダリカンの合成過程を構成する酵素および 基質などを含む物質群と被検化合物を接触させる工程  (a) contacting a test compound with a substance group containing a cell expressing chondroitin sulfate proteodarican or a part thereof, the cell extract, or an enzyme and a substrate constituting the synthesis process of chondroitin sulfate proteodarican Process
(b)前記細胞、細胞抽出液または物質群における、コンドロイチン硫酸プロテオグリカ ンまたはその合成過程における中間体の合成量を測定する工程  (b) a step of measuring the amount of chondroitin sulfate proteoglycan or an intermediate in the synthesis process in the cell, cell extract or substance group.
(c)被検化合物を接触させな!/、場合と比較して、前記合成量を低下させる化合物を 選択する工程  (c) Do not contact a test compound! /, a step of selecting a compound that reduces the amount of synthesis compared to the case
[0124] 上記方法においてはまず、コンドロイチン硫酸プロテオダリカンまたはその一部を発 現する細胞、該細胞抽出液、もしくはコンドロイチン硫酸プロテオダリカンの合成過程 を構成する酵素および基質などを含む物質群と被検化合物を接触させる。  [0124] In the above method, first, a cell that expresses chondroitin sulfate proteodarican or a part thereof, a cell extract, or a substance group containing an enzyme and a substrate constituting the synthesis process of chondroitin sulfate proteodarican, and the like. A test compound is brought into contact.
[0125] 次いで、コンドロイチン硫酸プロテオダリカンまたはその合成過程における中間体の 合成量を測定する。測定は当業者においては公知の手法、例えば、標識した抗体に よる方法、質量分析法、クロマトグラフィー法等によって適宜実施することができる。  [0125] Next, the synthesis amount of chondroitin sulfate proteodarican or an intermediate in the synthesis process is measured. The measurement can be appropriately carried out by those skilled in the art by a known method, for example, a method using a labeled antibody, mass spectrometry, chromatography, or the like.
[0126] さらに被検化合物を接触させない場合 (対照)と比較して、合成量を低下 (抑制)さ せる化合物を選択する。低下 (抑制)させる化合物は神経線維性変性治療のための 薬剤となる。 [0127] 被検化合物が上記 (b)合成阻害作用の活性を有して!/、るか否かにつ!/、て評価 (測 定)可能な方法、具体例の簡単な一例を以下に示す。 [0126] A compound that reduces (suppresses) the amount of synthesis is selected as compared with the case where the test compound is not contacted (control). Compounds that reduce (suppress) become agents for the treatment of neurofibrotic degeneration. [0127] A simple example of a method and a specific example that can be evaluated (measured) depending on whether the test compound has (b) the activity of inhibiting synthesis! Shown in
[0128] 上記 (b)コンドロイチン硫酸プロテオダリカンの合成阻害作用に関するスクリーニング 方法態様: [0128] (b) Screening for the synthesis inhibitory action of chondroitin sulfate proteodarican Method aspect:
コンドロイチン硫酸を合成する細胞、細胞株は当該研究者には既知である。ヒトで は例えば、健常人の末梢血を採取後、単核球を分離 '培養するという標準的な方法 により、 16時間の細胞培養でコンドロイチン硫酸を産生してくる(Uhlin-Hansen L et al ·, Blood 82:2880, 1993·など)。また、より簡便には、既知の細胞株、例えば、線維芽 細胞株 MH3T3 (Phillip HA, et al. J. Biol. Chem. 279:48640, 2004など)、腎尿細管 由来癌細胞株 ACHN (Kawashima H et al., J. Biol. Chem. 277: 12921, 2002)、腎遠 位尿細管由来細胞株 MDCK (Borges FT et al., Kidney Int. 68: 1630, 2005.など)、血 管内皮細胞株 HUVEC (Schick BP et al., Blood 97:449, 2001など)など、多数挙げら れる。このような細胞株を一定時間培養する過程にお!/、て各種被検化合物を混合し 、培養前後の CS-GAG量の変化を上記 (a)の方法で簡便に数値化できる。細胞培養 後の CS-GAG量の増加(すなわち、 CS-GAG合成量を反映する)を抑制する化合物 力 本コンセプトを満たす治療候補化合物として、容易に判定できる。  Cells and cell lines that synthesize chondroitin sulfate are known to the investigator. In humans, for example, chondroitin sulfate is produced in 16 hours of cell culture using the standard method of collecting and culturing mononuclear cells after collecting peripheral blood from healthy individuals (Uhlin-Hansen L et al , Blood 82: 2880, 1993, etc.). More simply, known cell lines such as fibroblast cell line MH3T3 (Phillip HA, et al. J. Biol. Chem. 279: 48640, 2004), renal tubular cancer cell line ACHN (Kawashima H et al., J. Biol. Chem. 277: 12921, 2002), distal renal tubular cell line MDCK (Borges FT et al., Kidney Int. 68: 1630, 2005., etc.), vascular endothelial cells Many strains such as HUVEC strains (Schick BP et al., Blood 97: 449, 2001, etc.) are mentioned. In the process of culturing such a cell line for a certain period of time, various test compounds are mixed, and the change in the CS-GAG amount before and after the culture can be easily quantified by the method (a). Compound power that suppresses the increase in CS-GAG amount after cell culture (ie, reflects the amount of CS-GAG synthesis) It can be easily determined as a therapeutic candidate compound that satisfies this concept.
[0129] さらに、より選択的には、例えば GalNAc4ST-lや XylosylTなどの CS-GAG合成酵素 の遺伝子を CHO細胞や L細胞などへ周知の方法で導入、恒常的に発現させた細胞 株を作成する事ができる。このような恒常的に CS-GAGを合成する細胞株を使用する 事により、よりクリア一に治療候補化合物を判定する事ができる。  [0129] Furthermore, more selectively, for example, a cell line in which CS-GAG synthase genes such as GalNAc4ST-1 and XylosylT are introduced into CHO cells and L cells in a well-known manner and expressed constantly is created. I can do it. By using such a cell line that constantly synthesizes CS-GAG, a therapeutic candidate compound can be determined more clearly.
[0130] 本発明のスクリーニング方法の他の態様としては、コンドロイチン硫酸プロテオダリ カンの脱硫酸化作用を有する物質を選択する工程を含む方法を挙げることができる 。本発明の上記方法は例えば以下の工程からなる。  [0130] As another embodiment of the screening method of the present invention, a method including a step of selecting a substance having a desulfating action of chondroitin sulfate proteodarican can be mentioned. The above method of the present invention comprises, for example, the following steps.
(a)コンドロイチン硫酸プロテオダリカンまたはその一部と被検化合物を接触させるェ 程 ( a ) The process of contacting the test compound with chondroitin sulfate proteodarican or a part thereof
(b)コンドロイチン硫酸プロテオダリカンまたはその一部の硫酸化を受けていた量を 測定する工程  (b) a step of measuring the amount of chondroitin sulfate proteodarican or a part thereof that has undergone sulfation
(c)被検化合物の非存在下におレ、て測定した場合と比較して、硫酸化を受けて!/、た 量を低下させる物質を選択する工程 (c) Compared with the measurement in the absence of the test compound, it was subjected to sulfation! /, The process of selecting substances that reduce the amount
[0131] 上記方法においてはまず、コンドロイチン硫酸プロテオダリカンまたはその一部に 被検化合物を接触させる。  [0131] In the above method, first, a test compound is contacted with chondroitin sulfate proteodarican or a part thereof.
[0132] 本方法においては次いで、コンドロイチン硫酸プロテオダリカンまたはその一部の 硫酸化を受けていた量を測定する。測定は、当業者に公知の方法によって行うことが できる。例えば、コンドロイチン硫酸プロテオダリカンまたはその一部に残存する脱硫 酸化の構造に結合する、標識された化合物または抗体を用い、標識量を測定するこ とにより検出すること力 Sできる。また、クロマトグラフィー法や質量分析法などを用いて 検出することあでさる。  [0132] Next, in this method, the amount of chondroitin sulfate proteodarican or a part thereof that has undergone sulfation is measured. The measurement can be performed by methods known to those skilled in the art. For example, it is possible to detect by measuring the amount of label using a labeled compound or antibody that binds to the structure of desulfurization oxidation remaining in chondroitin sulfate proteodarican or a part thereof. It can also be detected using chromatography or mass spectrometry.
[0133] 本方法にお!/、ては、次!/、で、被検化合物を接触させなレ、場合(対照)と比較して、 該コンドロイチン硫酸プロテオダリカンまたはその一部の存在量を低下させる化合物 を選択する。低下させる化合物は神経線維性変性治療のための薬剤となる。  [0133] In this method! /, The following! /, The test compound is not brought into contact with the test compound, compared to the case (control), the abundance of the chondroitin sulfate proteodarican or a part thereof Select a compound that lowers. The compound that lowers becomes a drug for treating neurofibrotic degeneration.
[0134] 被検化合物が上記 (c)脱硫酸化作用の活性を有して!/、るか否かにつ!/、て評価 (測 定)可能な方法、具体例の簡単な一例を以下に示す。 [0134] A simple example of a method and a specific example that can be evaluated (measured) according to whether the test compound has (c) the activity of desulfating action! Shown in
[0135] 上記 (c)コンドロイチン硫酸プロテオダリカンの脱硫酸化作用に関するスクリーニング 方法態様: [0135] (c) Screening for desulfating action of chondroitin sulfate proteodarican Method aspect:
基本的に上記 (a)の方法と同様、ヒト由来プロテオダリカン (BGN社、 ISL社など)など を準備し、 96穴プレートに 10 g/mLの濃度でコーティングする(Kawashima H et al. ; J. Biol. Chem. 277: 12921-12930, 2002·など、既知の方法による)。本プレートの各ゥ エルに各種の被検化合物を添加し、 37°Cで 2時間反応後に CS-GAGの変化を検出 する。  Basically, as in the method (a) above, human-derived proteodaricans (BGN, ISL, etc.) are prepared and coated on a 96-well plate at a concentration of 10 g / mL (Kawashima H et al.; J. Biol. Chem. 277: 12921-12930, 2002, etc.)). Add various test compounds to each well of this plate and detect CS-GAG change after 2 hours reaction at 37 ° C.
[0136] 検出方法は、脱硫酸化する事により、プロテオダリカンのコア蛋白側に残存した脱 硫酸化断片の 2糖構造を、抗プロテオダリカン A di4S抗体(クローン; 2-B-6、 4位に硫 酸化を受けて!/、た部分を認識する)あるいは抗プロテオダリカン Δ di6S (クローン; 3-B -3、 6位に硫酸化を受けていた部分を認識する。ともに生化学工業社製)と反応させ ることで、脱硫酸化を受けた部分の検出が容易にできる。したがって、混合培養前後 のプレートにおいて、 FITC標識した 2-B-6や 3-B-3抗体を反応させ、その蛍光数値 の変化を簡便に検出できる。反応前後の蛍光強度を増加させた化合物が、より脱硫 酸化を促進する物質であると判定でき、本コンセプトを満たす新規治療候補化合物と して簡便に同定できる。 [0136] The detection method was carried out by desulfating the disaccharide structure of the desulfated fragment remaining on the core protein side of the proteodarican into anti-proteodarican A di4S antibody (clone; 2-B-6, 4 Or the anti-proteodarican Δdi6S (clone; 3-B-3, recognizes the part that was sulfated at position 6. The product that has been subjected to desulfation can be easily detected. Therefore, FITC-labeled 2-B-6 and 3-B-3 antibodies can be reacted on the plate before and after mixed culture, and changes in the fluorescence values can be easily detected. Compounds with increased fluorescence intensity before and after reaction are more desulfurized It can be determined that the substance promotes oxidation, and can be easily identified as a novel therapeutic candidate compound that satisfies this concept.
[0137] 本発明のスクリーニング方法の他の態様としては、コンドロイチン硫酸プロテオダリ カンの硫酸化阻害作用を有する物質を選択する工程を含む方法を挙げることができ る。本発明の上記方法は、例えば以下の工程からなる。  [0137] As another embodiment of the screening method of the present invention, a method including a step of selecting a substance having a sulfation inhibitory action of chondroitin sulfate proteodarican can be mentioned. The above method of the present invention comprises, for example, the following steps.
(a)コンドロイチン硫酸プロテオダリカンまたはその一部を発現する細胞もしくは細胞 抽出液あるいはコンドロイチン硫酸プロテオダリカンの硫酸化過程を構成する酵素、 基質などを含む物質群と被検化合物を接触させる工程 ( a ) A step in which a test compound is contacted with a substance or a cell extract expressing chondroitin sulfate proteodarican or a part thereof, or a substance group containing an enzyme, a substrate, or the like that constitutes the sulfation process of chondroitin sulfate proteodarican
(b)前記細胞、細胞抽出液または物質群におけるコンドロイチン硫酸プロテオグリカ ンの硫酸化活性を測定する工程  (b) a step of measuring the sulfation activity of chondroitin sulfate proteoglycan in the cell, cell extract or substance group
(c)被検化合物を接触させな!/、場合と比較して、前記活性を低下させる化合物を選 択する工程  (c) Do not contact a test compound! /, a step of selecting a compound that decreases the activity compared to the case
[0138] 上記方法においてはまず、コンドロイチン硫酸プロテオダリカンまたはその一部に 被検物質を接触させる。  [0138] In the above method, first, a test substance is brought into contact with chondroitin sulfate proteodarican or a part thereof.
[0139] 本方法においては次いで、コンドロイチン硫酸プロテオダリカンまたはその一部の 硫酸化を受けていた量を測定する。測定は、当業者に公知の方法によって行うことが できる。例えば、コンドロイチン硫酸プロテオダリカンまたはその一部の硫酸化構造に 結合する、標識された化合物または抗体を用い、標識量を測定することにより検出す ること力 Sできる。また、クロマトグラフィー法や質量分析法などを用いて検出することも できる。  [0139] Next, in the present method, the amount of chondroitin sulfate proteodarican or a part thereof that has undergone sulfation is measured. The measurement can be performed by methods known to those skilled in the art. For example, it is possible to detect by measuring the amount of labeling using a labeled compound or antibody that binds to chondroitin sulfate proteodarican or a sulfated structure of a part thereof. Moreover, it can also detect using a chromatography method, a mass spectrometry, etc.
[0140] 本方法にお!/、ては、次!/、で、被検化合物を接触させなレ、場合(対照)と比較して、 該コンドロイチン硫酸プロテオダリカンまたはその一部の存在量を低下させる化合物 を選択する。低下させる化合物は神経線維性変性治療のための薬剤となる。  [0140] In this method! /, The following! /, When the test compound is not contacted, compared to the case (control), the abundance of the chondroitin sulfate proteodarican or a part thereof Select a compound that lowers. The compound that lowers becomes a drug for treating neurofibrotic degeneration.
[0141] 被検化合物が上記 (d)硫酸化阻害の活性を有して!/、るか否かにつ!/、て評価 (測定) 可能な方法、具体例の簡単な一例を以下に示す。 [0141] The test compound has the above-mentioned (d) activity of inhibiting sulfation! /, Whether or not! /, And can be evaluated (measured). Show.
[0142] 上記 (d)コンドロイチン硫酸プロテオダリカンの硫酸化阻害作用に関するスクリーニン グ方法態様: [0142] (d) Screening method relating to sulfation inhibitory action of chondroitin sulfate proteodarican:
コンドロイチン硫酸の硫酸化を促進する細胞、細胞株は上記(c)に記載の細胞、細 胞株と一致する。このような細胞株を一定時間培養する過程において各種被検化合 物を混合し、培養前後の硫酸化の程度を、例えば、 4位硫酸化を検出する抗体(クロ ーン; LY111)や 6位硫酸化を検出する抗体(クローン; MC21C、 1/、ずれも生化学工業 社製)で簡便に確認できる。蛍光標識抗体を用いて、培養前後の蛍光数値を比較し ても良いし、上記 (c)同様に、培養前後で 2-B-6や 3-B-3抗体を使用した検出法を行 つても良い。細胞培養後の硫酸化の増加(LY111や MC21Cの蛍光数値増力 P)を抑制 する化合物、もしくは細胞培養後の脱硫酸化の進行(2-B-6や 3-B-3の蛍光数値増 カロ)を促進する化合物が、本コンセプトを満たす治療候補化合物として、容易に判定 できる。 The cells and cell lines that promote sulfation of chondroitin sulfate are the cells, cells described in (c) above. Consistent with cell lines. In the process of culturing such a cell line for a certain period of time, various test compounds are mixed, and the degree of sulfation before and after culturing is measured, for example, antibody (clone: LY111) for detecting sulfation at position 4 or position 6 An antibody that detects sulfation (clone; MC21C, 1 /, deviation is also available from Seikagaku Corporation) can be easily confirmed. Fluorescence-labeled antibodies may be used to compare fluorescence values before and after culture. Similarly to (c) above, detection methods using 2-B-6 and 3-B-3 antibodies may be performed before and after culture. Also good. Compounds that suppress the increase in sulfation after cell culture (LY111 and MC21C fluorescence P), or the progress of desulfation after cell culture (2-B-6 and 3-B-3 fluorescence increase calo) Can be easily determined as a candidate compound that satisfies this concept.
[0143] さらに、より選択的には、例えば C4ST-1や C6ST-1などの硫酸基転移酵素の遺伝 子を CHO細胞や L細胞などへ周知の方法で導入、恒常的に発現させた細胞株を作 成する事ができる。このような恒常的に硫酸基を付加する細胞株を使用する事により 、よりクリア一に治療候補化合物を判定する事ができる。  [0143] Furthermore, more selectively, for example, a cell line in which a gene of a sulfotransferase such as C4ST-1 or C6ST-1 is introduced into CHO cells or L cells by a well-known method and is expressed constantly. Can be created. By using such a cell line that constantly adds a sulfate group, a therapeutic candidate compound can be determined more clearly.
[0144] 本発明の他の好ましい態様は、本発明のコンドロイチン硫酸プロテオダリカンのコア タンパク質、合成酵素、脱硫酸化酵素抑制タンパク質、または硫酸基転移酵素の遺 伝子の発現レベルを低下させる化合物、あるいはコンドロイチン硫酸プロテオグリカ ンの分解促進酵素または脱硫酸化酵素の遺伝子の発現レベルを上昇させる化合物 を選択する、以下の(a)〜(d)の工程を含む神経線維性変性抑制剤のスクリーニング 方法である。  [0144] Another preferred embodiment of the present invention is a compound that decreases the expression level of the chondroitin sulfate proteodarican core protein, the synthase, the desulfase inhibitor protein, or the sulfotransferase gene of the present invention, Alternatively, by selecting a compound that increases the expression level of the chondroitin sulfate proteoglycan degradation-promoting enzyme or desulfating enzyme gene, the screening method for a neurofibrotic degeneration inhibitor comprising the following steps (a) to (d): is there.
(a)コンドロイチン硫酸プロテオダリカンコアタンパク質、合成酵素、脱硫酸化酵素抑 制タンパク質、硫酸基転移酵素、分解促進酵素、または脱硫酸化酵素をコードする 遺伝子を発現する細胞に、被検化合物を接触させる工程 ( a ) A test compound is brought into contact with a cell expressing a gene encoding a chondroitin sulfate proteodlican core protein, a synthetic enzyme, a desulfase inhibitor protein, a sulfotransferase, a degradation promoting enzyme, or a desulfase enzyme. Process
(b)前記細胞における遺伝子の発現量を測定する工程  (b) measuring the gene expression level in the cell
(c)該発現量を被検化合物の非存在下におレ、て測定した場合 (対照)と比較するェ 程  (c) When the expression level is measured in the absence of the test compound and compared with the (control)
(d)前記遺伝子がコンドロイチン硫酸プロテオダリカンのコアタンパク質、合成酵素、 脱硫酸化酵素抑制タンパク質、または硫酸基転移酵素である場合には、前記遺伝子 の発現量が対照と比較して低下して!/、る化合物、前記遺伝子がコンドロイチン硫酸 プロテオダリカンの分解促進酵素、または脱硫酸化酵素である場合には、前記遺伝 子の発現量が対照と比較して上昇している化合物を選択する工程 (d) If the gene is a chondroitin sulfate proteodarican core protein, synthase, desulfase inhibitor protein, or sulfotransferase, the expression level of the gene is reduced compared to the control! /, The compound, the gene is chondroitin sulfate In the case of proteodarican degradation-promoting enzyme or desulfating enzyme, a step of selecting a compound in which the expression level of the gene is increased compared to the control
[0145] 上記方法においてはまず、コンドロイチン硫酸プロテオダリカンコアタンパク質、合 成酵素、脱硫酸化酵素抑制タンパク質、硫酸基転移酵素、分解促進酵素、または脱 硫酸化酵素をコードする遺伝子を発現する細胞に、被検化合物を接触させる。  [0145] In the above method, first, a cell expressing a gene encoding a chondroitin sulfate proteodlican core protein, a synthetic enzyme, a desulfurase inhibitor protein, a sulfotransferase, a degradation promoting enzyme, or a desulfurase is expressed. The test compound is brought into contact.
[0146] 本方法におレ、ては次!/、で、コンドロイチン硫酸プロテオダリカンコアタンパク質、合 成酵素、脱硫酸化酵素抑制タンパク質、硫酸基転移酵素、分解促進酵素、または脱 硫酸化酵素をコードする遺伝子の発現量を測定する。ここで「遺伝子の発現」には、 転写および翻訳の双方が含まれる。遺伝子の発現量の測定は、当業者に公知の方 法によって行うことができる。  [0146] In this method, chondroitin sulfate proteodarican core protein, synthetic enzyme, desulfase inhibitor protein, sulfate transferase, degradation promoting enzyme, or desulfase enzyme The expression level of the encoded gene is measured. Here, “gene expression” includes both transcription and translation. The gene expression level can be measured by methods known to those skilled in the art.
[0147] 例えば、上記いずれかのタンパク質を発現する細胞から mRNAを定法に従って抽 出し、この mRNAを铸型としたノーザンハイブリダィゼーシヨン法、 RT- PCR法、 DNA アレイ法等を実施することによって該遺伝子の転写量の測定を行うことができる。また 、上記いずれかのタンパク質をコードする遺伝子を発現する細胞からタンパク質画分 を回収し、上記!/、ずれかのタンパク質の発現を SDS-PAGE等の電気泳動法で検出す ることにより、遺伝子の翻訳量の測定を行うこともできる。さらに、上記いずれかのタン パク質に対する抗体を用いて、ウェスタンブロッテイング法を実施することにより該タン ノ ク質の発現を検出することにより、遺伝子の翻訳量の測定を行うことも可能である。 該タンパク質の検出に用いる抗体としては、検出可能な抗体であれば特に制限はな いが、例えばモノクローナル抗体、またはポリクローナル抗体の両方を利用すること ができる。  [0147] For example, mRNA is extracted from cells expressing any of the above proteins according to a standard method, and the Northern hybridization method, RT-PCR method, DNA array method, or the like using this mRNA as a cage is performed. Thus, the amount of transcription of the gene can be measured. In addition, a protein fraction is collected from a cell expressing a gene encoding any of the above proteins, and the expression of any of the above! / And any of the proteins is detected by electrophoresis such as SDS-PAGE. It is also possible to measure the translation amount. Furthermore, it is also possible to measure the amount of translation of a gene by detecting the expression of the protein by performing Western blotting using an antibody against any of the above proteins. . The antibody used for detecting the protein is not particularly limited as long as it is a detectable antibody. For example, both a monoclonal antibody and a polyclonal antibody can be used.
[0148] 本方法にお!/、ては、次!/、で、被検化合物を接触させなレ、場合(対照)と該遺伝子の 発現量を比較する。  [0148] In this method, the expression level of the gene is compared with that in the case where the test compound is not brought into contact in the following manner!
[0149] 本方法にお!/、ては、次!/、で、前記遺伝子がコンドロイチン硫酸プロテオダリカンのコ ァタンパク質、合成酵素、脱硫酸化酵素抑制タンパク質、または硫酸基転移酵素で ある場合には、前記遺伝子の発現量が対照と比較して低下 (抑制)させて!/、る化合物 を選択する。低下 (抑制)させる化合物は、神経線維性変性抑制のための薬剤もしく は神経線維性変性治療のための候補化合物となる。 [0150] また、前記遺伝子がコンドロイチン硫酸プロテオダリカンの分解促進酵素、または脱 硫酸化酵素である場合には、前記遺伝子の発現量が対照と比較して上昇 (増強)さ せている化合物を選択する。上昇 (増強)させる化合物は、神経線維性変性抑制の ための薬剤もしくは神経線維性変性治療のための候補化合物となる。 [0149] In the case of this method! /, In the following! /, When the gene is a chondroitin sulfate proteodarican core protein, a synthase, a desulfase inhibitor protein, or a sulfotransferase Select a compound that decreases (suppresses) the expression level of the gene compared to the control! A compound that lowers (suppresses) becomes a drug for suppressing neurofibrotic degeneration or a candidate compound for treating neurofibrotic degeneration. [0150] In addition, when the gene is a chondroitin sulfate proteodarican degradation-promoting enzyme or desulfating enzyme, a compound in which the expression level of the gene is increased (enhanced) compared to the control is used. select. The compound to be increased (enhanced) is a drug for suppressing nerve fiber degeneration or a candidate compound for treating nerve fiber degeneration.
[0151] また、本発明のスクリーニング方法の一態様としては、本発明のコンドロィチン硫酸 プロテオダリカンのコアタンパク質、合成酵素、脱硫酸化酵素抑制タンパク質、または 硫酸基転移酵素の遺伝子の発現レベルを低下させる化合物、ある!/、はコンドロイチ ン硫酸プロテオダリカンの分解促進酵素または脱硫酸化酵素の遺伝子の発現レべ ルを上昇させる化合物を、レポーター遺伝子の発現を指標として選択する方法であ る。本発明の上記方法は例えば以下の(a)〜(d)の工程を含む。  [0151] Further, as one aspect of the screening method of the present invention, the expression level of the core protein, synthase, desulfase inhibitor protein, or sulfotransferase gene of the chondroitin sulfate proteodarican of the present invention is reduced. Is a method of selecting a compound that increases the expression level of the chondroitin sulfate proteodarican degradation-promoting enzyme or desulfating enzyme gene using the expression of the reporter gene as an index. The method of the present invention includes, for example, the following steps (a) to (d).
(a)コンドロイチン硫酸プロテオダリカンコアタンパク質、合成酵素、脱硫酸化酵素抑 制タンパク質、硫酸基転移酵素、分解促進酵素、または脱硫酸化酵素をコードする 遺伝子の転写調節領域とレポーター遺伝子とが機能的に結合した構造を有する DN Aを含む細胞または細胞抽出液と、被検化合物を接触させる工程 ( a ) The transcriptional regulatory region of a gene encoding a chondroitin sulfate proteodarican core protein, a synthetic enzyme, a desulfase inhibitor protein, a sulfotransferase, a degradation-promoting enzyme, or a desulfase enzyme and the reporter gene are functional. A step of contacting a test compound with a cell or cell extract containing DNA having a bound structure
(b)前記レポーター遺伝子の発現レベルを測定する工程  (b) measuring the expression level of the reporter gene
(c)被検化合物を接触させな!/ヽ場合 (対照)と比較する工程  (c) Do not let the test compound come into contact!
(d)前記レポーター遺伝子がコンドロイチン硫酸プロテオダリカンのコアタンパク質、 合成酵素、脱硫酸化酵素抑制タンパク質、または硫酸基転移酵素と機能的に結合し てレ、る場合には、前記レポーター遺伝子の発現レベルが対照と比較して低下してレヽ る化合物、前記レポーター遺伝子がコンドロイチン硫酸プロテオダリカンの分解促進 酵素、または脱硫酸化酵素に機能的に結合している場合には、前記レポーター遺伝 子の発現レベルが対照と比較して上昇している化合物を選択する工程  (d) if the reporter gene is functionally linked to the core protein, synthase, desulfase inhibitor protein, or sulfotransferase of chondroitin sulfate proteodarican, the expression level of the reporter gene If the reporter gene is functionally linked to a chondroitin sulfate proteodalican degradation-promoting enzyme or desulfating enzyme, the expression level of the reporter gene Selecting a compound that is elevated relative to a control
[0152] 本方法においてはまず、コンドロイチン硫酸プロテオダリカンコアタンパク質、合成 酵素、脱硫酸化酵素抑制タンパク質、硫酸基転移酵素、分解促進酵素、または脱硫 酸化酵素をコードする遺伝子の転写調節領域とレポーター遺伝子とが機能的に結合 した構造を有する DNAを含む細胞または細胞抽出液と、被検化合物を接触させる。  [0152] In this method, first, the transcriptional regulatory region and reporter gene of a gene encoding chondroitin sulfate proteodlican core protein, synthase, desulfase inhibitor protein, sulfotransferase, degradation promoting enzyme, or desulfurase A test compound is brought into contact with a cell or cell extract containing DNA having a structure in which and are functionally bound.
[0153] ここで「機能的に結合した」とは、コンドロイチン硫酸プロテオダリカンコアタンパク質 、合成酵素、脱硫酸化酵素抑制タンパク質、硫酸基転移酵素、分解促進酵素、また は脱硫酸化酵素をコードする遺伝子の転写調節領域に転写因子が結合することによ り、レポーター遺伝子の発現が誘導されるように、コンドロイチン硫酸プロテオダリカン コアタンパク質、合成酵素、脱硫酸化酵素抑制タンパク質、硫酸基転移酵素、分解 促進酵素、または脱硫酸化酵素をコードする遺伝子の転写調節領域とレポーター遺 伝子とが結合していることをいう。従って、レポーター遺伝子が他の遺伝子と結合して おり、他の遺伝子産物との融合タンパク質を形成する場合であっても、コンドロイチン 硫酸プロテオダリカンコアタンパク質、合成酵素、脱硫酸化酵素抑制タンパク質、硫 酸基転移酵素、分解促進酵素、または脱硫酸化酵素をコードする遺伝子の転写調 節領域に転写因子が結合することによって、該融合タンパク質の発現が誘導されるも のであれば、上記「機能的に結合した」の意に含まれる。コンドロイチン硫酸プロテオ ダリカンコアタンパク質、合成酵素、脱硫酸化酵素抑制タンパク質、硫酸基転移酵素 、分解促進酵素、または脱硫酸化酵素をコードする遺伝子の cDNA塩基配列に基づ いて、当業者においては、ゲノム中に存在するコンドロイチン硫酸プロテオダリカンコ ァタンパク質、合成酵素、脱硫酸化酵素抑制タンパク質、硫酸基転移酵素、分解促 進酵素、または脱硫酸化酵素をコードする遺伝子の転写調節領域を周知の方法に より取得することが可能である。 [0153] Here, "functionally bound" refers to chondroitin sulfate proteodalycan core protein, synthase, desulfase inhibitor protein, sulfate transferase, degradation promoting enzyme, Chondroitin sulfate proteolycan core protein, synthase, desulfase inhibitor protein so that the transcription factor binds to the transcriptional regulatory region of the gene encoding desulfase to induce reporter gene expression. It means that a transcriptional regulatory region of a gene encoding a sulfotransferase, a degradation promoting enzyme, or a desulfating enzyme is linked to a reporter gene. Therefore, even when the reporter gene is linked to other genes and forms a fusion protein with other gene products, chondroitin sulfate proteodlican core protein, synthase, desulfase inhibitor protein, sulfate If the expression of the fusion protein is induced by binding of a transcription factor to the transcriptional regulatory region of a gene encoding a transferase, a degradation promoting enzyme, or a desulfating enzyme, the above-mentioned "functionally bound" Is included. Based on the cDNA base sequence of the gene encoding chondroitin sulfate proteodalican core protein, synthase, desulfase inhibitor protein, sulfate transferase, degradation promoting enzyme, or desulfase, Obtain the transcriptional regulatory region of the gene encoding chondroitin sulfate proteodalycan core protein, synthetic enzyme, desulfase inhibitor protein, sulfotransferase, degradation-promoting enzyme, or desulfase enzyme existing in It is possible.
本方法に用いるレポーター遺伝子としては、その発現が検出可能であれば特に制 限はなぐ例えば、 CAT遺伝子、 lacZ遺伝子、ルシフェラーゼ遺伝子、および GFP遺 伝子等が挙げられる。「コンドロイチン硫酸プロテオダリカンコアタンパク質、合成酵素 、脱硫酸化酵素抑制タンパク質、硫酸基転移酵素、分解促進酵素、または脱硫酸化 酵素をコードする遺伝子の転写調節領域とレポーター遺伝子とが機能的に結合した 構造を有する DNAを含む細胞」として、例えば、このような構造が揷入されたベクター を導入した細胞が挙げられる。このようなベクターは、当業者に周知の方法により作 製すること力 Sできる。ベクターの細胞への導入は、一般的な方法、例えば、リン酸カル シゥム沈殿法、電気パルス穿孔法、リポフエクシヨン法、マイクロインジェクション法等 によって実施すること力 Sできる。「コンドロイチン硫酸プロテオダリカンコアタンパク質、 合成酵素、脱硫酸化酵素抑制タンパク質、硫酸基転移酵素、分解促進酵素、または 脱硫酸化酵素をコードする遺伝子の転写調節領域とレポーター遺伝子とが機能的に 結合した構造を有する DNAを含む細胞」には、染色体に該構造が揷入された細胞も 含まれる。染色体への DNA構造の揷入は、当業者に一般的に用いられる方法、例え ば、相同組み換えを利用した遺伝子導入法により行うことができる。 The reporter gene used in this method is not particularly limited as long as its expression can be detected, for example, CAT gene, lacZ gene, luciferase gene, GFP gene and the like. “A structure in which a transcriptional regulatory region of a gene encoding a chondroitin sulfate proteodarican core protein, a synthetic enzyme, a desulfase inhibitor protein, a sulfotransferase, a degradation-promoting enzyme, or a desulfase enzyme and a reporter gene are functionally linked. Examples of the “cell containing a DNA having” include a cell into which a vector having such a structure inserted is introduced. Such vectors can be made by methods well known to those skilled in the art. The introduction of the vector into the cell can be carried out by a general method such as a calcium phosphate precipitation method, an electric pulse perforation method, a lipofussion method, or a microinjection method. “The transcriptional regulatory region of a gene encoding chondroitin sulfate proteodarican core protein, synthase, desulfase inhibitor protein, sulfate transferase, degradation promoting enzyme, or desulfase is functionally associated with the reporter gene. A “cell containing DNA having a linked structure” also includes cells in which the structure is inserted into a chromosome. The introduction of a DNA structure into a chromosome can be performed by a method generally used by those skilled in the art, for example, a gene introduction method using homologous recombination.
[0155] 「コンドロイチン硫酸プロテオダリカンコアタンパク質、合成酵素、脱硫酸化酵素抑 制タンパク質、硫酸基転移酵素、分解促進酵素、または脱硫酸化酵素をコードする 遺伝子の転写調節領域とレポーター遺伝子とが機能的に結合した構造を有する DN Aを含む細胞抽出液」とは、例えば、市販の試験管内転写翻訳キットに含まれる細胞 抽出液に、コンドロイチン硫酸プロテオダリカンコアタンパク質、合成酵素、脱硫酸化 酵素抑制タンパク質、または硫酸基転移酵素をコードする遺伝子の転写調節領域と レポーター遺伝子とが機能的に結合した構造を有する DNAを添加したものを挙げる こと力 Sでさる。 [0155] "Transcriptional regulatory region of a gene encoding chondroitin sulfate proteodlican core protein, synthetic enzyme, desulfase inhibitor protein, sulfotransferase, degradation promoting enzyme, or desulfase enzyme and reporter gene are functional. The cell extract containing DNA having a structure bonded to is, for example, a chondroitin sulfate proteodlican core protein, synthase, desulfated enzyme inhibitory protein added to a cell extract contained in a commercially available in vitro transcription translation kit. Or the addition of DNA having a structure in which a transcriptional regulatory region of a gene encoding a sulfotransferase and a reporter gene are functionally linked.
[0156] ここで「接触」は、「コンドロイチン硫酸プロテオダリカンコアタンパク質、合成酵素、 脱硫酸化酵素抑制タンパク質、硫酸基転移酵素、分解促進酵素、または脱硫酸化 酵素をコードする遺伝子の転写調節領域とレポーター遺伝子とが機能的に結合した 構造を有する DNAを含む細胞」の培養液に被検化合物を添加する、または該 DNAを 含む上記の市販された細胞抽出液に被検化合物を添加することにより行うことができ る。被検化合物がタンパク質の場合には、例えば、該タンパク質を発現する DNAベタ ターを、該細胞へ導入することにより行うことも可能である。  [0156] Here, "contact" means "chondroitin sulfate proteodalycan core protein, synthase, desulfase inhibitor protein, sulfotransferase, degradation-promoting enzyme, or transcriptional regulatory region of a gene encoding a desulfase enzyme. By adding a test compound to the culture solution of “cells containing DNA having a structure functionally linked to a reporter gene” or adding the test compound to the above-described commercially available cell extract containing the DNA It can be carried out. In the case where the test compound is a protein, for example, it can be carried out by introducing a DNA vector that expresses the protein into the cell.
[0157] 本方法にお!/、ては、次!/、で、該レポーター遺伝子の発現レベルを測定する。レポ 一ター遺伝子の発現レベルは、該レポーター遺伝子の種類に応じて、当業者に公知 の方法により測定することができる。例えば、レポーター遺伝子が CAT遺伝子である 場合には、該遺伝子産物によるクロラムフエ二コールのァセチル化を検出することに よって、レポーター遺伝子の発現量を測定することができる。レポーター遺伝子が lac Z遺伝子である場合には、該遺伝子発現産物の触媒作用による色素化合物の発色 を検出することにより、また、ルシフェラーゼ遺伝子である場合には、該遺伝子発現 産物の触媒作用による蛍光化合物の蛍光を検出することにより、さらに、 GFP遺伝子 である場合には、 GFPタンパク質による蛍光を検出することにより、レポーター遺伝子 の発現量を測定することができる。 [0158] 本方法にお!/、ては、次!/、で、測定したレポーター遺伝子の発現レベルを被検化合 物の非存在下におレ、て測定した場合 (対照)と比較する。 [0157] In this method, the expression level of the reporter gene is measured by! /, And then! /. The expression level of the reporter gene can be measured by methods known to those skilled in the art depending on the type of the reporter gene. For example, when the reporter gene is a CAT gene, the expression level of the reporter gene can be measured by detecting acetylation of chloramphenicol by the gene product. When the reporter gene is the lac Z gene, by detecting the color development of the dye compound by the catalytic action of the gene expression product, and when it is a luciferase gene, the fluorescent compound by the catalytic action of the gene expression product In the case of the GFP gene, the expression level of the reporter gene can be measured by detecting the fluorescence of the GFP protein. [0158] In this method, compare the expression level of the reporter gene measured in the absence of the test compound (control) with the following! /.
[0159] 本方法にお!/、ては、次!/、で、前記レポーター遺伝子がコンドロイチン硫酸プロテオ ダリカンのコアタンパク質、合成酵素、脱硫酸化酵素抑制タンパク質、または硫酸基 転移酵素をコードする遺伝子と機能的に結合している場合には、前記レポーター遺 伝子の発現レベルが対照と比較して低下 (抑制)させて!/、る化合物を選択する。低下 (抑制)させる化合物は、神経線維性変性抑制のための薬剤もしくは神経線維性変 性治療のための候補化合物となる。  [0159] In this method, the reporter gene is a gene encoding a chondroitin sulfate proteodalican core protein, a synthetic enzyme, a desulfase inhibitor protein, or a sulfotransferase. In the case of functional binding, a compound that reduces (suppresses) the expression level of the reporter gene compared to the control is selected. A compound that lowers (suppresses) becomes a drug for inhibiting neurofibrotic degeneration or a candidate compound for treating neurofibrotic degeneration.
[0160] また、前記レポーター遺伝子がコンドロイチン硫酸プロテオダリカンの分解促進酵 素、または脱硫酸化酵素にと機能的に結合している場合には、前記レポーター遺伝 子の発現レベルが対照と比較して上昇 (増強)させて!/、る化合物を選択する。上昇( 増強)させる化合物は、神経線維性変性抑制のための薬剤もしくは神経線維性変性 治療のための候補化合物となる。  [0160] In addition, when the reporter gene is functionally linked to a chondroitin sulfate proteodarican degradation-promoting enzyme or a desulfase enzyme, the expression level of the reporter gene is higher than that of the control. Increase (enhance) the compound! The compound to be increased (enhanced) is a drug for suppressing nerve fiber degeneration or a candidate compound for treating nerve fiber degeneration.
[0161] 本発明のスクリーニング方法にお!/、て見出される神経線維性変性抑制剤は、好ま しくは、神経線維性変性疾患の治療用または予防用のものである。  [0161] The neurofibrotic degeneration inhibitor found in the screening method of the present invention is preferably for treating or preventing a neurofibrotic degenerative disease.
[0162] また本発明は、本発明のスクリーニング方法を実施するために用いられる、各種薬 剤 ·試薬等を含むキットを提供する。  [0162] The present invention also provides a kit containing various drugs, reagents and the like used for carrying out the screening method of the present invention.
[0163] 本発明のキットは、例えば本発明の上述の各種試薬の中から、実施するスクリー二 ング方法にあわせて適宜選択することができる。例えば本発明のキットは、本発明の コンドロイチン硫酸プロテオダリカンを構成要素とすることができる。本発明のキットに は、さらに、本発明の方法において使用される各種試薬、容器等を含めることができ る。例えば、抗コンドロイチン硫酸プロテオダリカン抗体、プローブ、各種反応試薬、 細胞、培養液、対照サンプル、緩衝液、使用方法を記載した指示書等を適宜含める こと力 Sでさる。  [0163] The kit of the present invention can be appropriately selected from, for example, the above-mentioned various reagents of the present invention according to the screening method to be performed. For example, the kit of the present invention can comprise the chondroitin sulfate proteodarican of the present invention as a constituent element. The kit of the present invention can further contain various reagents, containers and the like used in the method of the present invention. For example, an anti-chondroitin sulfate proteodarican antibody, probe, various reaction reagents, cells, culture solution, control sample, buffer solution, instructions describing the method of use, etc. may be included as appropriate.
[0164] 本発明の好ましい態様においては、コンドロイチン硫酸プロテオダリカンの生成もし くは蓄積が阻害されてレ、るかどうかを検出する工程を含む、神経線維性変性抑制剤 のスクリーニング方法である。従って、該スクリーニング方法において、例えばコンドロ ィチン硫酸プロテオダリカンの検出の際に利用可能な、コンドロイチン硫酸プロテオ ダリカンのコアタンパク質をコードする遺伝子に対するプローブもしくは該遺伝子の任 意の領域を増幅するためのプライマー等のオリゴヌクレオチド、または、コンドロイチン 硫酸プロテオダリカンを認識する抗体 (抗コンドロイチン硫酸プロテオダリカン抗体)も 、本発明の神経線維性変性抑制剤スクリーニング用キットの構成要素に含めることが できる。 [0164] In a preferred embodiment of the present invention, there is provided a screening method for a neurofibrotic degeneration inhibitor comprising a step of detecting whether the production or accumulation of chondroitin sulfate proteodalycan is inhibited. Accordingly, in this screening method, for example, chondroitin sulfate proteolycan that can be used in the detection of chondroitin sulfate proteodarican. Oligonucleotides such as probes for the gene encoding the core protein of darlicans or primers for amplifying any region of the gene, or antibodies that recognize chondroitin sulfate proteodarican (anti-chondroitin sulfate proteodarican antibodies) , And can be included in the components of the screening kit for a neurofibrotic degeneration inhibitor of the present invention.
[0165] 上記オリゴヌクレオチドは、例えば、本発明の Versicanコアタンパク質遺伝子の DNA に特異的にハイブリダィズするものである。ここで「特異的にハイブリダィズする」とは 、通常のハイブリダィゼーシヨン条件下、好ましくはストリンジェントなハイブリダィゼー シヨン条件下 (ί列えば、サムブノレックら, Molecular Cloning, Cold Spring Harbour Lab oratory Press, New York, USA,第2版 1989に記載の条件)において、他のタンパク 質をコードする DNAとクロスハイブリダィゼーシヨンを有意に生じないことを意味する。 特異的なハイブリダィズが可能であれば、該オリゴヌクレオチドは、本発明の Versican コアタンパク質遺伝子の塩基配列に対し、完全に相補的である必要はない。 [0165] The oligonucleotide specifically hybridizes to, for example, the DNA of the Versican core protein gene of the present invention. Here, “specifically hybridize” means under normal hybridization conditions, preferably under stringent hybridization conditions (for example, Samb Norec et al., Molecular Cloning, Cold Spring Harbor Laboratory Press, New In the conditions described in York, USA, 2nd edition, 1989), it means that cross-hybridization does not occur significantly with DNA encoding other proteins. If specific hybridization is possible, the oligonucleotide does not have to be completely complementary to the base sequence of the Versican core protein gene of the present invention.
[0166] 本発明においてハイブリダィゼーシヨンの条件としては、例えば「2XSSC、 0.1%SD S、 50°C」、「2XSSC、 0.1%SDS、 42°C」、「1XSSC、 0.1%SDS、 37°C」、よりストリンジェ ントな条件として「2XSSC、 0.1%SDS、 65。C」、「0.5XSSC、 0.1%SDS、 42°C」および「 0.2XSSC、 0.1%SDS、 65°C」等の条件を挙げることができる。より詳細には、 Rapid-hy b buffer (Amersham Life Science)を用いた方法として、 68°Cで 30分間以上プレハイブ リダイゼーシヨンを行った後、プローブを添加して 1時間以上 68°Cに保ってハイブリッ ド形成させ、その後 2XSSC、 0.1%SDS中、室温で 20分間の洗浄を 3回行い、続いて 1 XSSC、 0.1%SDS中、 37°Cで 20分間の洗浄を 3回行い、最後に 1XSSC、 0.1%SDS中 、 50°Cで 20分間の洗浄を 2回行うことができる。その他、例えば Expresshyb Hybridizat ion Solution (CLONTECH)中、 55°Cで 30分間以上プレハイブリダィゼーシヨンを行つ た後、標識プローブを添加して 37〜55°Cで 1時間以上インキュベートし、 2XSSC、 0.1 %SDS中、室温で 20分間の洗浄を 3回、 1XSSC、 0.1%SDS中、 37°Cで 20分間の洗浄 を 1回行うこともできる。ここで、例えば、プレハイブリダィゼーシヨン、ハイブリダィゼー シヨンや 2度目の洗浄の際の温度をより高く設定することにより、よりストリンジヱントな 条件とすること力できる。例えば、プレハイブリダィゼーシヨンおよびハイブリダィゼー シヨンの温度を 60°C、さらにストリンジヱントな条件としては 68°Cとすることができる。当 業者であれば、このようなバッファーの塩濃度、温度等の条件に加えて、プローブ濃 度、プローブの長さ、プローブの塩基配列構成、反応時間等のその他の条件を加味 し、条件を設定することができる。 [0166] In the present invention, hybridization conditions include, for example, "2XSSC, 0.1% SDS, 50 ° C", "2XSSC, 0.1% SDS, 42 ° C", "1XSSC, 0.1% SDS, 37 “C”, more stringent conditions such as “2XSSC, 0.1% SDS, 65.C”, “0.5XSSC, 0.1% SDS, 42 ° C” and “0.2XSSC, 0.1% SDS, 65 ° C” Can be mentioned. More specifically, as a method using Rapid-hy b buffer (Amersham Life Science), pre-hybridization is performed at 68 ° C for 30 minutes or more, then a probe is added, and hybridization is performed at 68 ° C for 1 hour or more. After that, wash twice in 2XSSC, 0.1% SDS at room temperature for 20 minutes, then wash in 1 XSSC, 0.1% SDS at 37 ° C for 20 minutes three times, and finally 1XSSC, It can be washed twice for 20 minutes at 50 ° C in 0.1% SDS. In addition, for example, in Expresshyb Hybridization Solution (CLONTECH), prehybridize at 55 ° C for 30 minutes or longer, add labeled probe, and incubate at 37-55 ° C for 1 hour or longer. It is also possible to perform 3 washes for 20 minutes at room temperature in 0.1% SDS and 1 wash for 20 minutes at 37 ° C in 1XSSC and 0.1% SDS. Here, for example, by setting the temperature at the time of pre-hybridization, hybridization, and the second washing higher, it is possible to make the conditions more stringent. For example, prehybridization and hybridisation The temperature of the chillon can be set to 60 ° C, and a stringent condition can be set to 68 ° C. Those skilled in the art will consider the conditions such as the salt concentration and temperature of the buffer, as well as other conditions such as the probe concentration, probe length, probe base sequence composition, and reaction time. Can be set.
[0167] 該オリゴヌクレオチドは、上記本発明のスクリーニング用キットにおけるプローブや プライマーとして用いることができる。該オリゴヌクレオチドをプライマーとして用いる場 合、その長さは、通常 15 bp〜100 bpであり、好ましくは 17 bp〜30 bpである。プライマ 一は、例えば配列番号: 69または 70に記載のものである力 上記本発明中の遺伝 子の DNAの少なくとも一部を増幅しうるものであれば、特に制限されない。  [0167] The oligonucleotide can be used as a probe or primer in the above-described screening kit of the present invention. When the oligonucleotide is used as a primer, the length is usually 15 bp to 100 bp, preferably 17 bp to 30 bp. The primer is not particularly limited as long as it can amplify at least a part of the DNA of the gene of the present invention described above, for example, the force described in SEQ ID NO: 69 or 70.
[0168] また本発明は、本発明の薬剤を個体 (例えば、患者等)へ投与する工程を含む、神 経線維性変性疾患の治療もしくは予防方法を提供する。  [0168] The present invention also provides a method for treating or preventing a neurofibrotic degenerative disease, comprising the step of administering the agent of the present invention to an individual (eg, a patient).
[0169] 本発明の予防もしくは治療方法の対象となる個体は、神経線維性変性疾患を発症 し得る生物であれば特に制限されないが、好ましくはヒトである。  [0169] The individual that is the subject of the prevention or treatment method of the present invention is not particularly limited as long as it is an organism capable of developing a neurofibrotic degenerative disease, but is preferably a human.
[0170] 個体への投与は、一般的には、例えば、動脈内注射、静脈内注射、皮下注射など 当業者に公知の方法により行うことができる。投与量は、患者の体重や年齢、投与方 法などにより変動するが、当業者(医師、獣医師、薬剤師等)であれば適当な投与量 を適宜選択することが可能である。  [0170] In general, administration to an individual can be performed by methods known to those skilled in the art, such as intraarterial injection, intravenous injection, and subcutaneous injection. The dose varies depending on the weight and age of the patient, the administration method, etc., but a person skilled in the art (such as a doctor, veterinarian, pharmacist, etc.) can appropriately select an appropriate dose.
[0171] さらに本発明は、本発明の薬剤の、神経線維性変性抑制剤の製造における使用に 関する。  [0171] Furthermore, the present invention relates to the use of the agent of the present invention in the production of a neurofibrotic degeneration inhibitor.
[0172] なお本明細書において引用された全ての先行技術文献は、参照として本明細書に 組み入れられる。  [0172] It should be noted that all prior art documents cited in the present specification are incorporated herein by reference.
実施例  Example
[0173] 以下、実施例を用いて本発明をさらに具体的に説明する。但し、本発明の技術的 範囲はこれらの実施例に限定されるものではない。  [0173] Hereinafter, the present invention will be described more specifically with reference to Examples. However, the technical scope of the present invention is not limited to these examples.
[0174] 実施例 1 MPTP誘発件 C57BL/6TcLパーキンソン病モデルマウスによる GalNAcST ( siRNA)処置 コンドロイチナーゼ ABC処置における GalNAc4ST-l GALNAC4S-6ST ィ云 の f计 Example 1 MPTP Induction Case GalNAcST (siRNA) Treatment with C57BL / 6TcL Parkinson's Disease Model Mice GalNAc4ST-l GALNAC4S-6ST in Chondroitinase ABC Treatment
本実施例では 1 -methy卜 4-pheny卜 1,2,3,6 tetrahydropyridine (MPTP)によりドーノ ミンニューロンを選択的に変性させたパーキンソン病のマウスモデルを作成(Amende et al. (2005) Journal of NeuroEngineering and Rehabilitation 2(20) 1- 13)して GalNA cST siRNA薬とコンドロイチナーゼ ABC薬を投与し、治療後における遺伝子発現と組 織の様子を比較検討した。 In this example, 1-methy 卜 4-pheny 卜 1,2,3,6 tetrahydropyridine (MPTP) Created a mouse model of Parkinson's disease that selectively denatured min neurons (Amende et al. (2005) Journal of NeuroEngineering and Rehabilitation 2 (20) 1-13) and developed GalNA cST siRNA drug and chondroitinase ABC drug. After administration, the gene expression after treatment and the state of the tissue were compared.
[0175] 妊娠 14日目 C57BL/6JcLマウス(日本クレア社製)を飼育、出産させ、 8週令 C57B L/6JcLマウス雌(日本クレア社製)、各々にコンドロイチナーゼ ABC (Sigma Aldrich Ja pan) 4 U/ml 100 1と GalNAcST (GalNAc4ST-l、 GalNAc4ST- 2と GALNAC4S- 6STの カクテル配列を混合したもの) siRNA 1 g (GeneWorld社製)を siRNA媒体である 1% A telocollagen (高研社製)と混合させたもの 200 L腹腔内投与した。その後、二日、三 日そして四日後の三度にわたりドーノ ミンニューロンのみを選択的に破壊する MPTP (Sigma Aldrich Japan社製)を 30 mg/kgで体内へ投与したものを飼育した。実験 8日 目に BrdU 5 mg/mL (ZyMED Laboratory. Inc社製) 100 1を尾静脈内投与処置し、 1 時間後、解剖し、脳を摘出し、免疫染色用サンプル、遺伝子発現解析用サンプルを 得た。以上より摘出した臓器 (脳) 50 mg当たりに対し、 RNA-Bee (TEL-TEST社製) 1 mLを力 Pえ、電動ホモジナイザー(DIGITAL HOMOGENIZER, ASONE社製)にて粉 砕させた後、 chloroform 200 ^ 1 (Sigma Aldrich Japan社製)を加え穏やかに混合後、 約 5分氷冷し、 12,000 rpm、 4°C、 15分間 遠心分離機(Centrifbge 5417R、 eppendorf 社製)を用い遠心分離を行った。遠心分離後の上澄み液 500 Lを別のエツペンドル フチューブに移し、上澄み液と同等量の isopropannol 500 L (Sigma Aldrich Japan社 製)を加え混合後、 1 しの glycogen dnvitrogen社製)を加え、 15分間氷冷した。氷冷 15分後、 12,000 rpm、 4。C、 15分間遠心し、その後、 75% Ethanol 1000 1 (Sigma Aldri ch Japan社製)で 3回洗浄して得られた RNA沈殿物を 30分間〜 1時間、 自然乾燥させ た後、大塚蒸留水 50 1 (大塚製薬社製)に溶解させ、さらに大塚蒸留水 (大塚製薬 社製)にて 100倍希釈し、 UVプレート(Corning Costar社製)上でプレートリーダー(P OWER Wave XS、 BIO-TEK社製)により抽出したサンプル中の RNA濃度を算出した[0175] Day 14 of pregnancy C57BL / 6JcL mice (CLEA Japan, Inc.) were bred and born, 8 weeks old female C57B L / 6JcL mice (CLEA Japan), chondroitinase ABC (Sigma Aldrich Ja pan) ) 4 U / ml 100 1 and GalNAcST (GalNAc4ST-l, GalNAc4ST-2 and GALNAC4S-6ST cocktail sequence mixed) 1% A telocollagen (Kokensha) siRNA 1 g (GeneWorld) 200 L) was mixed intraperitoneally. Thereafter, MPTP (manufactured by Sigma Aldrich Japan), which selectively destroys only donomin neurons, was administered to the body at 30 mg / kg three times on the second, third and fourth days. On the 8th day of the experiment, BrdU 5 mg / mL (ZyMED Laboratory. Inc.) 100 1 was administered into the tail vein, and after 1 hour, dissection was performed, the brain was removed, immunostaining sample, gene expression analysis sample Got. From the above, per 1 mg of organ (brain) removed, 1 mL of RNA-Bee (TEL-TEST) is added, and after grinding with an electric homogenizer (DIGITAL HOMOGENIZER, ASONE), chloroform Add 200 ^ 1 (Sigma Aldrich Japan) and mix gently, then ice-cool for about 5 minutes and centrifuge using a centrifuge (Centrifbge 5417R, eppendorf) for 15 minutes at 12,000 rpm, 4 ° C. It was. Transfer 500 L of the supernatant after centrifugation to another Eppendorf tube, add isopropannol 500 L (manufactured by Sigma Aldrich Japan) in the same amount as the supernatant, mix, and add 1 glycogen dnvitrogen) for 15 minutes. Ice-cooled. 15 minutes after ice cooling, 12,000 rpm, 4. C. Centrifugation for 15 minutes, followed by 3 times washing with 75% Ethanol 1000 1 (Sigma Aldrich Japan), the RNA precipitate was air-dried for 30 minutes to 1 hour, and then Otsuka distilled water. 50 1 (manufactured by Otsuka Pharmaceutical Co., Ltd.), 100-fold diluted with Otsuka distilled water (manufactured by Otsuka Pharmaceutical Co., Ltd.), and plate reader (POWER Wave XS, BIO-TEK) on a UV plate (Corning Costar) Calculated the RNA concentration in the sample extracted by
Yes
[0176] 次に、 RT reaction (cDNA合成)を行うため以下の手技を行った。算出して得られた RNAサンプルを 500 rig/20 ^ 1の濃度に調整し、 68°C、 3分間、 BLOCK INCUBATOR (ASTEC製)にて加温し、 10分間、氷冷した。氷冷後、予め調製していた RT Pre Mix 液(組成: 25 mM MgCl 18.64 μ 1 (Invitrogen社製)、 5 X Buffer 20 μ 1 (Invitrogen社製 )、 0.1 M DTT 6.6 1 (Invitrogen社製)、 10 mM dNTP mix 10 1 (Invitrogen社製)、 R Nase Inhibitor 2 μ 1 (Invitrogen社製)、 MMLV Reverse transcriptase 1.2 μ 1 (Invitroge η社製)、 Random primer 2 μ 1 (Invitrogen社製)、滅菌蒸留水 19.56 μ 1 (大塚蒸留水: 大塚製薬社製)を 80 1加え BLOCK INCUBATOR (ASTEC社製)にて 42°C、 1時間、 加温反応させ、 1時間後、 BLOCK INCUBATOR (ASTEC社製)にて 99°C、 5分間、加 熱した後、氷冷し求める cDNA 100 1を作製し、合成して得られた cDNAを用いて、以 下の組成で PCR反応を行った。 [0176] Next, the following procedure was performed for RT reaction (cDNA synthesis). Adjust the calculated RNA sample to a concentration of 500 rig / 20 ^ 1 and set it to 68 ° C for 3 minutes with BLOCK INCUBATOR Heated (ASTEC) and ice-cooled for 10 minutes. RT Premix solution (composition: 25 mM MgCl 18.64 μ1 (Invitrogen)), 5 X Buffer 20 μ1 (Invitrogen), 0.1 M DTT 6.6 1 (Invitrogen) , 10 mM dNTP mix 10 1 (Invitrogen), RNase Inhibitor 2 μ 1 (Invitrogen), MMLV Reverse transcriptase 1.2 μ 1 (Invitroge η), Random primer 2 μ 1 (Invitrogen), sterilization Distilled water 19.56 μ 1 (Otsuka distilled water: Otsuka Pharmaceutical Co., Ltd.) 80 1 was added, BLOCK INCUBATOR (ASTEC Co., Ltd.) was used, and the reaction was heated at 42 ° C for 1 hour. After 1 hour, BLOCK INCUBATOR (ASTEC Co., Ltd. was produced) ) Was heated at 99 ° C for 5 minutes, and then ice-cooled to prepare cDNA 1001 to be obtained, and the synthesized cDNA was used to carry out PCR reaction with the following composition.
[0177] PCR Buffer 2 μ 1 [組成: 166 mM (NH ) SO (Sigma Aldrich Japan社製)、 670mM Tri [0177] PCR Buffer 2 μ 1 [Composition: 166 mM (NH 2) SO (Sigma Aldrich Japan), 670 mM Tri
4 2 4  4 2 4
s -HC1 pH8.8 (Invitrogen社製)、 67 mM MgCl - 6H O (Sigma Aldrich Japan社製)、 10 0 mM 2-mercaptoethanol] (WAKO社製))、 25 mM dNTP mix 0.8 1 (Invitrogen社製 )、 DMSO 0.6 1 (Sigma Aldrich Japan社製)、 Primer Forward 0.2 l (GeneWorld社 製)、 Primer Reverse 0.2 l (GeneWorld社製)、大塚蒸留水 15.7〃 1 (大塚製薬社製) 、 Taq polymerase 0.1 ^ KPerkin Elmer社製)、上述のようにして得られた cDNA 1 μ 1 を混合させ Authorized Themal Cycler (印 pendorff土製)により 94°C 45second、 55°C 4 5 sec, 72°C 60second 30cycle反応させた。反応終了後、得られた PCR産物に 2 1 L oading Dye (Invitrogen社^ Dを力!];^、 1.5% UltraPure Agarose (Invitrogen†土 )グノレを 作製し、 Mupid-2 plus (AD VANCE社製)により 100 V、 20分間電気泳動を行った。泳 動後、 1 X LoTE [組成: 3 mM Tris- HCl (pH7.5) (Invitrogen社製)、 0.2 mM EDTA (pH 7.5) (Sigma Aldrich Japan社製)]にて 10000倍希釈 Ethydium Bromide (Invitrogen社 製)染色液中で 20分間振とうさせた後、 I-Scope WD (ADVANCE社製)に設置した EX ILIM (CASIO社製)にてゲル撮影し遺伝子発現の確認を行った。  s-HC1 pH8.8 (Invitrogen), 67 mM MgCl-6H 2 O (Sigma Aldrich Japan), 100 mM 2-mercaptoethanol] (WAKO)), 25 mM dNTP mix 0.8 1 (Invitrogen) ), DMSO 0.6 1 (manufactured by Sigma Aldrich Japan), Primer Forward 0.2 l (manufactured by GeneWorld), Primer Reverse 0.2 l (manufactured by GeneWorld), 15.7 〃 1 (manufactured by Otsuka Pharmaceutical), Taq polymerase 0.1 ^ KPerkin Elmer) and 1 μ 1 of cDNA obtained as described above were mixed and reacted at 94 ° C for 45 seconds, 55 ° C for 45 seconds, 72 ° C for 60 seconds for 30 cycles using Authorized Themal Cycler (made by Pendorff Earth). . After completion of the reaction, prepare 21 1 Loading Dye (Invitrogen ^ D power!); ^, 1.5% UltraPure Agarose (Invitrogen † Sat) gnore, and use Mupid-2 plus (AD VANCE) ) For 100 minutes at 100 V. After swimming, 1 X LoTE [Composition: 3 mM Tris-HCl (pH7.5) (Invitrogen), 0.2 mM EDTA (pH 7.5) (Sigma Aldrich Japan) The product was shaken in Ethydium Bromide (Invitrogen) staining solution for 20 minutes and then geled with EX ILIM (CASIO) installed in I-Scope WD (ADVANCE). Photographs were taken to confirm gene expression.
[0178] 図 1に RT-PCRによる未治療群、 GalNAcST siRNA処置群とコンドロイチーゼ ABCに おける β _actin、 GalNAc4ST-lと GALNAC4S-6STの遺伝子発現結果を示した。今回 、用いた GalNAcST siRNA (GalNAc4ST-l siRNAカクテル、 GalNAc4ST_2 siRNAカク テルと GALNAC4S-6ST siRNAカクテルの混合 siRNA; GeneWorld社製)、 Primer (For ward, Reverse) (北海道システム ·サイエンス社製)を以下に示す。 [0179] [Primer配列] [0178] Fig. 1 shows the results of gene expression of β_actin, GalNAc4ST-1 and GALNAC4S-6ST in the untreated group, GalNAcST siRNA-treated group and chondroitize ABC by RT-PCR. The following GalNAcST siRNA (GalNAc4ST-l siRNA cocktail, mixed siRNA of GalNAc4ST_2 siRNA cocktail and GALNAC4S-6ST siRNA cocktail; GeneWorld), Primer (Forward, Reverse) (Hokkaido System Science) Show. [0179] [Primer sequence]
* β -actin (Gene world社製)  * β -actin (Gene world)
Forward: 5,- GACCCAGATCATGTTTGAGAC -3,(配列番号: 67)  Forward: 5,-GACCCAGATCATGTTTGAGAC -3, (SEQ ID NO: 67)
Reverse: 5, - ATGCCTGGGTACATGGTGGTA -3,(配列番号: 68)  Reverse: 5,-ATGCCTGGGTACATGGTGGTA -3, (SEQ ID NO: 68)
[0180] *GalNAc4ST-l (北海道システム ·サイエンス社製)  [0180] * GalNAc4ST-l (Hokkaido System Science)
Forward: 5,- ACGTGCCTTTTACACCCAAG- 3,(配列番号: 69)  Forward: 5,-ACGTGCCTTTTACACCCAAG-3, (SEQ ID NO: 69)
Reverse: 5 ' -GTGTGCCCTTTTCTGTGGAT-3 ' (酉己歹 IJ番号: 70)  Reverse: 5 '-GTGTGCCCTTTTCTGTGGAT-3' (酉 己 歹 IJ number: 70)
[0181] [GalNAc4ST-l siRNAカクテル配歹 IJ] (GenBank accession number NM.175140)  [0181] [GalNAc4ST-l siRNA cocktail IJ] (GenBank accession number NM.175140)
(Gene world社製)  (Gene world)
5 ' -ACCCCCAACTCGGAACGATGCGGCT-3 ' (酉己歹 IJ番号 : 71)  5 '-ACCCCCAACTCGGAACGATGCGGCT-3' (酉 番号 IJ number: 71)
5 ' -TGCATGTTCTCGTCCATCCTGCTG-3 ' (配列番号: 72)  5 '-TGCATGTTCTCGTCCATCCTGCTG-3' (SEQ ID NO: 72)
5 ' -CGCCACCGTGTACTGTACTGTGAAGT-3 ' (配列番号: 73)  5 '-CGCCACCGTGTACTGTACTGTGAAGT-3' (SEQ ID NO: 73)
5,- AGGCT GCTCCAACTG GAAGAGGGTG- 3,(配列番号: 74)  5,-AGGCT GCTCCAACTG GAAGAGGGTG-3 (SEQ ID NO: 74)
[0182] [GalNAc4ST-2 siRNAカクテル配歹 IJ] (GenBank accession number NM— 199055) [0182] [GalNAc4ST-2 siRNA cocktail served IJ] (GenBank accession number NM—199055)
(Gene world社製)  (Gene world)
5 ' -ATATAGTATCTAGGATATATGTAG-3 ' (配列番号: 75)  5 '-ATATAGTATCTAGGATATATGTAG-3' (SEQ ID NO: 75)
5 ' -GAAGTACCAAAAGCTGGCTGCTCTA-3 ' (配列番号: 76)  5 '-GAAGTACCAAAAGCTGGCTGCTCTA-3' (SEQ ID NO: 76)
5 ' -TTCTATCACTTGGACTATTTGATGTT-3 ' (酉己歹 IJ番号 : 77)  5 '-TTCTATCACTTGGACTATTTGATGTT-3' (IJ number: 77)
5 ' -TACACAACTCCACATTTGTAATTTG-3 ' (配列番号: 78)  5 '-TACACAACTCCACATTTGTAATTTG-3' (SEQ ID NO: 78)
[0183] [GALNAC4S-6ST siRNAカクテル配歹 IJ] (GenBank accession number NM.029935) [0183] [GALNAC4S-6ST siRNA cocktail IJ] (GenBank accession number NM.029935)
(Gene world社製)  (Gene world)
5 ' -CCAGAAGCCAAGCTCATTGTTATG-3 ' (配列番号: 79)  5 '-CCAGAAGCCAAGCTCATTGTTATG-3' (SEQ ID NO: 79)
5 ' - CTGTGGAGAGGTTGTACTCAGACTA- 3 ' (配列番号: 80)  5 '-CTGTGGAGAGGTTGTACTCAGACTA- 3' (SEQ ID NO: 80)
5 ' -ATTTGCCTGGAAGACAACGTGAGAGC-3 ' (配列番号:81)  5 '-ATTTGCCTGGAAGACAACGTGAGAGC-3' (SEQ ID NO: 81)
5 ' -GTCCCTTCTGCAGAAGCTGGGCCCACT-3 ' (酉己歹 IJ番号: 82)  5 '-GTCCCTTCTGCAGAAGCTGGGCCCACT-3' (酉 歹 IJ number: 82)
[0184] 図 1の RT-PCRにより内因性コントロールである β -actinの発現が未治療群、 GalNA cST (siRNA)、コンドロイチナーゼ ABC処置群ともに同レベルでの発現で認められた。 これに対して GalNAc4ST-lと GALNAC4S-6STにおいて未治療群と比較し GalNAcST (siRNA)処置群にお!/、て発現減少が認められ Atellocollagen媒体 GalNAcST siRNA 投与による遺伝子発現抑制(減少)が確認された。 [0184] Expression of endogenous control β-actin was observed at the same level in the untreated group, GalNA cST (siRNA), and chondroitinase ABC-treated group by RT-PCR in FIG. In contrast, GalNAc4ST-l and GALNAC4S-6ST compared with the untreated group In the (siRNA) treatment group, a decrease in expression was observed, and suppression of gene expression (reduction) was confirmed by administration of Atellocollagen medium GalNAcST siRNA.
[0185] 実施例 2 MPTP誘発性 C57BL/6.TcLパーキンソン病モデルマウスによるコンドロイ チナーゼ ABC GalNAcST siRNA処置における CSPGの脳内沈着抑制効果の比較検 この実施例ではパーキンソン病モデルマウスの脳組織サンプルを使用して CSPGの 沈着抑制効果にっレ、て比較検討を行った。実施例 1にて得られた脳組織を凍結用 包埋剤 OCTコンパウンド(Miles社製)に包埋して液体窒素にて凍結ブロックを作成し た。その凍結ブロックからクリオスタツト(Microm社製)を用いて厚さ 10 mの切片を作 成した。得られた切片をアセトン(Sigma Aldrich Japan社製)で 10分間固定後、リン酸 緩衝液で洗浄し、さらに一次抗体として抗コンドロイチン硫酸プロテオダリカン (CSPG )抗体(クローン CS56、マウスモノクローナル抗体、 lO g/ml;生化学工業社製)を添 加し、室温で 1時間反応させた。続いて、ヒストファインマウススティンキット(ニチレイ 社製;マウスモノクローナル抗体に対して使用)を用いて二次抗体反応を行った後、 DAB基質 (ニチレイ社製)を添加し酵素色素反応を行った。この標本を光学顕微鏡( ライカ社製)にて観察した。この組織像を図 2として示した (原図はカラーである)。未 治療群における陽性のシグナルは歯状回付近にて対照群の CSPG蓄積より強く示し ていることがわかる。そして、 GalNAcST siRNAとコンドロイチナーゼ ABC治療群にお ける CSPGの蓄積は抑制されていることが見受けられる。つまり、以上の結果を考察す ると、 MPTP誘導型パーキンソン病マウスモデルの脳組織における CSPGの沈着は Gal NAcST siRNAとコンドロイチナーゼ ABCの生体内投与において抑制される所見が明 らかにされた。 [0185] Example 2 MPTP-induced C57BL / 6. TcL Parkinson's disease model mouse comparison test of chondroitinase ABC GalNAcST siRNA treatment to suppress the deposition of CSPG in the brain This example uses a brain tissue sample of Parkinson's disease model mouse Thus, a comparative study was conducted on the effect of CSPG deposition suppression. The brain tissue obtained in Example 1 was embedded in a freezing embedding agent OCT compound (manufactured by Miles), and a frozen block was prepared with liquid nitrogen. A 10 m thick section was prepared from the frozen block using a cryostat (Microm). The obtained sections were fixed with acetone (manufactured by Sigma Aldrich Japan) for 10 minutes, washed with a phosphate buffer, and further anti-chondroitin sulfate proteodarican (CSPG) antibody (clone CS56, mouse monoclonal antibody, lO as a primary antibody). g / ml (manufactured by Seikagaku Corporation) was added and allowed to react at room temperature for 1 hour. Subsequently, a secondary antibody reaction was performed using a histofine mouse stin kit (manufactured by Nichirei; used for mouse monoclonal antibody), and then DAB substrate (manufactured by Nichirei) was added to carry out an enzyme dye reaction. This specimen was observed with an optical microscope (manufactured by Leica). This tissue image is shown in Fig. 2 (the original image is in color). It can be seen that the positive signal in the untreated group is stronger than the CSPG accumulation in the control group near the dentate gyrus. It can be seen that the accumulation of CSPG in the GalNAcST siRNA and chondroitinase ABC treatment group is suppressed. In other words, considering the above results, it was found that CSPG deposition in the brain tissue of an MPTP-induced Parkinson's disease mouse model was suppressed by in vivo administration of Gal NAcST siRNA and chondroitinase ABC. .
[0186] 実施例 3 MPTP誘発性 C57BL/6.TcLパーキンソン病モデルマウスによるコンドロイチ ナーゼ ABC GalNAcST siRNA処置におけるマクロファージ浸潤に伴う炎症抑制効 の比較检討  [0186] Example 3 Comparison of the suppression of inflammation associated with macrophage infiltration in chondroitinase ABC GalNAcST siRNA treatment using MPTP-induced C57BL / 6.TcL Parkinson's disease model mice
実施例 2で示した CSPGの沈着はマクロファージなどの炎症細胞を誘導する生体内 物質であるケモカインを吸着することが知られている。さらに CSPGの沈着が結果的に 炎症細胞を引き寄せて脳組織の破壊を誘発させることにつながると推測し、実施例 2 と同様に脳組織のサンプルを用いて、脳内マクロファージの集積動態に対して GalN AcST投与とコンドロイチナーゼ ABCの効果を比較した。実施例 2と同様の方法で得ら れた切片を 4% PFA (Paraformaldehyde)リン酸緩衝液(ナカライテスタ社製)で 10分間 固定後、脱イオン水で洗浄し、一次抗体としてラット由来抗マウスマクロファージ抗体 (クローン F4/80 ; l : 200希釈; BMA社製)を添加し、 4°Cにてー晚反応させた。次に二 次抗体である Alexa488標識抗ラット IgGャギ抗体(1: 200希釈; Invitrogen社)を添加し 、室温で 30分反応させた。以上の手法にて得られた組織像を図 3に示した (原図は力 ラーである)。未治療群での強い陽性シグナルは組織辺縁部にて対照群より多数の マクロファージの集積を示している結果であった。さらに、 GalNAcST siRNAとコンドロ イチナーゼ ABCによる治療群における染色結果にて比較してみると対照群とほぼ同 程度であった。以上の結果より、パーキンソン病マウスモデルの脳組織におけるマク 口ファージの集積は、 GalNAcST siRNAとコンドロイチナーゼ ABCの生体内投与により 顕著に抑制されることが明らかになった。 (マクロファージ集積は緑色、核は Sytox-Or ange unvitrogen)を用ぃ飞赤色に飞 k不) The CSPG deposition shown in Example 2 is known to adsorb chemokines, which are in vivo substances that induce inflammatory cells such as macrophages. In addition, it was speculated that the deposition of CSPG would eventually lead to attracting inflammatory cells to induce destruction of brain tissue. Similarly, the effects of GalN AcST administration and chondroitinase ABC on brain macrophage accumulation kinetics were compared using brain tissue samples. Sections obtained in the same manner as in Example 2 were fixed with 4% PFA (Paraformaldehyde) phosphate buffer (Nacalai Testa) for 10 minutes, washed with deionized water, and rat-derived anti-mouse as the primary antibody. Macrophage antibody (clone F4 / 80; 1: 200 dilution; manufactured by BMA) was added, and the mixture was reacted at 4 ° C. Next, the secondary antibody Alexa488-labeled anti-rat IgG goat antibody (1: 200 dilution; Invitrogen) was added and allowed to react at room temperature for 30 minutes. The tissue image obtained by the above method is shown in Fig. 3 (the original image is a power error). The strong positive signal in the untreated group was a result of the accumulation of more macrophages in the tissue margin than in the control group. Furthermore, when comparing the staining results in the treatment group with GalNAcST siRNA and chondroitinase ABC, the results were almost the same as in the control group. From the above results, it was revealed that the accumulation of macaque phages in the brain tissue of a Parkinson's disease mouse model was significantly suppressed by in vivo administration of GalNAcST siRNA and chondroitinase ABC. (The macrophage accumulation is green, the nucleus is Sytox-Organge unvitrogen)
実施例 4 MPTP誘発件 C57BL/6TcLパーキンソン病モデルマウスによるコンドロイチ ナーゼ ABOGalNAcST siRNA几 i における 細 閏ネ JMl 力 の比 f计 実施例 2で示した CSPGの沈着はさらに細胞の線維化を促進させる可能性があると 考え、実施例 2、 3と同様に脳組織のサンプルを用いて、脳内の神経細胞における線 維化に対して GalNAcST投与とコンドロイチナーゼ ABCの生体内投与により得られる 組織所見を比較した。実施例 2、 3と同様の方法で得られた切片を 4%PFAリン酸緩 衝液け力ライテスタ社製)で 10分間固定後、脱イオン水で洗浄し、一次抗体として線 維芽細胞抗体 (ER-TR7; 1: 100希釈; BMA社製)を添加し、 4°Cで一晩反応させた。 次に二次抗体である Alexa488標識抗ラット IgGャギ抗体(1: 200希釈; Invitrogen社)を 添加し、室温で 30分反応させた。以上の手法にて得られた組織像を図 4に示した (原 図はカラーである)。未治療群での強い陽性シグナルは脳梁膨大後部顆粒皮質付近 にて対照群より脳内における線維芽細胞の浸潤を示している結果であった。さらに、 GalNAcST siRNAとコンドロイチナーゼ ABC治療群における所見と比較すると対照群 と同様に線維芽細胞の陽性所見は確認できな力、つた。以上の結果より、パーキンソン 病マウスモデルの誘導される脳組織における ER-TR7の陽性シグナルの所見は、 Gal NAcST siRNAとコンドロイチナーゼ ABCの生体内投与により顕著に抑制されることが 明らかになった。また、本実施例と実施例 2と 3にて明らかにされた CSPGの沈着と、そ の沈着により誘導されるマクロファージの浸潤を含めて考察すると CSPGの過剰発現 を抑制することで炎症細胞の誘導、また細胞の線維化を阻止することができた。その 結果、神経線維性変性の抑制につながることが可能であると結論づけられる。 (線維 芽細胞は緑色、核は Sytox-Orange (invitrogen)を用いて赤色にて表示) Example 4 MPTP induction C57BL / 6TcL Parkinson's disease model mouse chondroitinase ABOGalNAcST siRNA 几 i ratio of cellulose JMl force f meter CSPG deposition shown in Example 2 may further promote cell fibrosis In the same manner as in Examples 2 and 3, using tissue samples from brain tissue, we examined tissue findings obtained by in vivo administration of GalNAcST and chondroitinase ABC for fibrosis in neurons in the brain. Compared. Sections obtained in the same manner as in Examples 2 and 3 were fixed for 10 minutes with 4% PFA phosphate buffer lysate), washed with deionized water, and fibroblast antibody ( ER-TR7; 1: 100 dilution; manufactured by BMA) was added and allowed to react overnight at 4 ° C. Next, the secondary antibody Alexa488-labeled anti-rat IgG goat antibody (1: 200 dilution; Invitrogen) was added and allowed to react at room temperature for 30 minutes. The tissue image obtained by the above method is shown in Fig. 4 (the original image is in color). A strong positive signal in the untreated group was the result of fibroblast infiltration in the brain in the vicinity of the enlarged cortex of the posterior corpus callosum than in the control group. Furthermore, when compared with the findings in the GalNAcST siRNA and chondroitinase ABC treatment group, the positive findings of fibroblasts were unidentifiable as in the control group. From the above results, Parkinson The findings of ER-TR7 positive signal in brain tissue induced by a disease mouse model were found to be significantly suppressed by in vivo administration of Gal NAcST siRNA and chondroitinase ABC. In addition, considering the CSPG deposition revealed in this example and Examples 2 and 3, and macrophage infiltration induced by the deposition, the induction of inflammatory cells by suppressing the overexpression of CSPG And also prevented cell fibrosis. As a result, it can be concluded that it can lead to suppression of neurofibrotic degeneration. (Fibroblasts are displayed in green, nuclei are displayed in red using Sytox-Orange (invitrogen))
[0188] 実施例 5 MPTP誘発性 C57BL/6.TcLパーキンソン病モデルマウスによる GalNAcST ( siRNA) コンドロイチナーゼ ABC処置におけるァストロサイトの活性効果の比較検討 脳には多数の神経細胞の他にグリア細胞という栄養素の供給の働きを持つ細胞が 存在している。本実施例では抗 GFAP抗体を用いてグリア細胞におけるァストロサイト の染色を行レ、、 GalNAcST投与とコンドロイチナーゼ ABCの生体内投与により得られ る組織所見を比較した。実施例 2、 3、 4と同様の方法で得られた切片を 4%PFAリン 酸緩衝液け力ライテスタ社製)で 10分間固定後、脱イオン水で洗浄し、一次抗体とし て抗 GFAP抗体(1: 20希釈; Santa Cruz Biotechnology)を添加し、 4°Cでー晚反応さ せた。次に二次抗体である Alexa488標識抗ャギ IgG-ロバ抗体(1 : 200希釈; Invitroge n社)を添加し、室温で 30分反応させた。図 5に対照群、未治療群、 GalNAcST siRNA 、コンドロイチナーゼ ABC処置群各々の組織画像を示した(原図はカラーである)。脳 梁膨大後部顆粒皮質付近において、対照群では正常にァストロサイトが存在してい ることが確認できるが未治療群においては陰性のシグナルとして現れた。これは、 MP TPにより選択的にグリア細胞のァストロサイトが破壊されたことを示唆する所見である 。一方、 GalNAcST siRNAとコンドロイチナーゼ ABC処置を行った処置群におけるシ グナルは未治療群に比べて強く現れていることが確認できた。グリア細胞におけるァ スト口サイトの役割として神経細胞の機能維持を中心とした働きを行うことで知られて いる。この所見では CSPGの抑制によりグリア細胞のァストロサイト働きが回復したこと が示唆され、神経細胞における機能の向上が考えられる。 (ァストロサイトは緑色、核 は Sytox-Orange (invitrogen)を用いて赤色にて 不) [0188] Example 5 Comparative study of the effect of astrocytes on treatment with GalNAcST (siRNA) chondroitinase ABC in MPTP-induced C57BL / 6.TcL Parkinson's disease model mice In addition to numerous neurons in the brain, glial cells There are cells that function to supply nutrients. In this example, anti-GFAP antibody was used to stain astrocytes in glial cells, and the tissue findings obtained by in vivo administration of GalNAcST and in vivo administration of chondroitinase ABC were compared. Sections obtained in the same manner as in Examples 2, 3 and 4 were fixed with 4% PFA phosphate buffer solution (manufactured by Leitesta) for 10 minutes, washed with deionized water, and anti-GFAP antibody as the primary antibody. (1:20 dilution; Santa Cruz Biotechnology) was added and allowed to react at 4 ° C. Next, Alexa488-labeled anti-goat IgG-donkey antibody (1: 200 dilution; Invitrogen), which is a secondary antibody, was added and reacted at room temperature for 30 minutes. FIG. 5 shows tissue images of the control group, the untreated group, the GalNAcST siRNA, and the chondroitinase ABC treatment group (the original figure is color). In the vicinity of the massive corpus callosum granule cortex, it was confirmed that astrocytes were normally present in the control group, but a negative signal appeared in the untreated group. This suggests that MP TP selectively destroyed glial astrocytes. On the other hand, it was confirmed that the signal in the treatment group treated with GalNAcST siRNA and chondroitinase ABC appeared stronger than that in the untreated group. It is known that the role of the first mouth site in glial cells is to play a role mainly in maintaining the function of nerve cells. This finding suggests that the inhibition of CSPG restored the function of astrocytes in glial cells, suggesting an improvement in function in neurons. (Astrocytes are green, nuclei are red using Sytox-Orange (invitrogen))
[0189] 実施例 6 生 C57BL/gJgLパーキンソン痞モ ルマウスによる GalNAcST ( siRNA)処置におけるドーパミンニューロンの活性効果の比較検討 [0189] Example 6 GalNAcST (by live C57BL / gJgL Parkinson's mouse) Comparative study of the effect of dopamine neurons on siRNA treatment
以上の実施例で示された結果を最終的に明らかにするために得られた組織サンプ ル切片をドーパミンニューロンのマーカとする抗チロシンハイドロキシラーゼ抗体にて ドーパミンニューロンを染め、組織所見を比較検討した。このチロシンノヽイド口キシラ ーゼ(TH: thyrosine hydroxylase)はドーパミン前駆体をドーパミンへ転換する酵素で ある。実施例 2、 3、 4と同様の方法で得られた切片を 4%PFAリン酸緩衝液(ナカライ テスタ社製)で 10分間固定後、脱イオン水で洗浄し、一次抗体としてラビットポリクロー ナル抗チロシンハイドロキシラーゼ抗体(1: 50希釈; Calbiochem社)を添加し、室温で 1時間反応させた。次に二次抗体である Alexa488標識抗ラビット-ロバ抗体(1 : 200希 釈; Invitrogen社)を添加し、室温で 30分反応させた。図 6に対照群、未治療群、 GalN AcST siRNA、コンドロイチナーゼ ABC処置群各々の組織画像を示した(原図はカラ 一である)。中脳上丘付近において、対照群では正常にチロシンノヽイドロキシラーゼ が発現して!/、ることが確認できるが未治療群にお!/、ては陰性のシグナルとして現れ た。これは、 MPTPにより選択的にドーパミンニューロンが破壊されたことが示唆される 所見である。一方、 GalNAcST siRNAとコンドロイチナーゼ ABC処置を行った処置群 におけるシグナルは未治療群に比べて強く現れていることが確認できた。つまり、 Gal NAcST siRNAとコンドロイチナーゼ ABCの生体内投与によって実施例 5にて示したァ スト口サイトの活性化に伴いドーパミンニューロンの機能回復が期待できる結論が得 られた。 (THは緑色、核は Sytox-Orange (invitrogen)を用いて赤色にて表示)  In order to finally clarify the results shown in the above Examples, dopamine neurons were stained with an anti-tyrosine hydroxylase antibody using tissue sample sections obtained as markers of dopamine neurons, and the tissue findings were compared. . This tyrosine hydroxylase (TH) is an enzyme that converts dopamine precursors into dopamine. Sections obtained in the same manner as in Examples 2, 3, and 4 were fixed with 4% PFA phosphate buffer (manufactured by Nacalai Tester) for 10 minutes, washed with deionized water, and rabbit polyclonal as a primary antibody. Anti-tyrosine hydroxylase antibody (1:50 dilution; Calbiochem) was added and allowed to react at room temperature for 1 hour. Next, the secondary antibody Alexa488-labeled anti-rabbit-donkey antibody (1: 200 dilution; Invitrogen) was added and reacted at room temperature for 30 minutes. FIG. 6 shows tissue images of the control group, the untreated group, the GalN AcST siRNA, and the chondroitinase ABC treatment group (the original figure is the same). In the vicinity of the upper mesencephalon, it was confirmed that the tyrosine neuroxylase was normally expressed in the control group! /, But it appeared as a negative signal in the untreated group! This suggests that MPTP selectively destroyed dopamine neurons. On the other hand, it was confirmed that the signal in the treatment group treated with GalNAcST siRNA and chondroitinase ABC appeared stronger than that in the untreated group. That is, it was concluded that the functional recovery of dopamine neurons can be expected by in vivo administration of Gal NAcST siRNA and chondroitinase ABC with the activation of the first oral site shown in Example 5. (TH is displayed in green, nucleus is displayed in red using Sytox-Orange (invitrogen))
[0190] さらに本発明者は、実際の CSPG発現についての影響に関して、 CS-56抗体を用い た免疫染色にて検討した(図 7)。写真は、歯状回付近の所見について示している。 未治療群では、 CSPG陽性のシグナルが強く発現している。これに対して、 C4-sulfata se治療では陽性のシグナルの発現は低下していた。次に、 C4-sulfataseの抗炎症、 抗泉維化効果を検討する目的で、炎症マーカー、 泉維化マーカーの発現について 検討した。図 8に示すように、未治療群にくらべ、治療群において、 TNF- αと TGF- /3 の発現にお!/、て有意差の抑制効果があった。  [0190] Furthermore, the present inventor examined the effect on actual CSPG expression by immunostaining using CS-56 antibody (Fig. 7). The photo shows the findings near the dentate gyrus. In the untreated group, a CSPG positive signal is strongly expressed. In contrast, C4-sulfata se treatment decreased the expression of positive signals. Next, in order to investigate the anti-inflammatory and anti-spring maintenance effects of C4-sulfatase, the expression of inflammation markers and fountain maintenance markers was examined. As shown in FIG. 8, compared to the untreated group, there was a significant effect of suppressing the difference in the expression of TNF-α and TGF- / 3 in the treated group!
[0191] 本実験では DA neuronの再生についての検討を試みた。 DA neuronの発現に関与 する遺伝子である Nurrlをマーカーとして C4sulfataseが及ぼす影響について検討し た結果について図 9に示す。 C4sul治療群では、未治療群に比べて Nurrlの発現が 有意(p< 0.001)に増加していた。また、図 10に示すように、脳組織における DA neur onの発現を検討したところ、同様の結果を得た。これらの結果は、 PDモデルに対する 抗 CSPG療法は、抗炎症、抗線維化効果と合わせて、 DA neuronの再生を促進させて いる可能性が示唆された。 [0191] In this experiment, we tried to examine the regeneration of DA neuron. We examined the effect of C4sulfatase using Nurrl, a gene involved in DA neuron expression, as a marker. Figure 9 shows the results. In the C4sul-treated group, Nurrl expression was significantly increased (p <0.001) compared to the untreated group. Further, as shown in FIG. 10, when the expression of DA neuron in brain tissue was examined, the same result was obtained. These results suggest that anti-CSPG therapy for PD model may promote DA neuron regeneration, combined with anti-inflammatory and anti-fibrotic effects.
[0192] 最後に、 MPDP投与によって誘因されるパーキンソン病モデルにおける治療群、未 治療群の生存率についても比較を行った。その結果、 end pointにおける生存率は、 未治療群が 16.7%、 C4sul治療群が 50%、 GalNAc si治療群が 71.4%であった。感染 や薬剤性の脳炎、外傷などの侵襲に対する保護作用が示唆される結果となった。 以下、実験方法および結果について具体的に説明する。  [0192] Finally, we compared the survival rates of treated and untreated groups in Parkinson's disease model induced by MPDP administration. As a result, the survival rate at the end point was 16.7% in the untreated group, 50% in the C4sul treated group, and 71.4% in the GalNAc si treated group. The results suggest a protective effect against invasion such as infection, drug-induced encephalitis, and trauma. Hereinafter, the experimental method and results will be specifically described.
[0193] 実施例 7 MPTP誘発件 C57BL6 TcLパーキンソン病モデルマウスによる C4-sulfatas e几 i におけろ CSPGの ^^ ネ JMI 力 の比 f计  Example 7 MPTP induction C57BL6 TcL Parkinson's disease model mice in C4-sulfatas e 几 i CSPG ^^ ne JMI power ratio f meter
本実施例では 1- methy卜 4- pheny卜 2,3- dihydropyridinium (MPDP)によりドーノ ミン ニューロンを選択的に変性させたパーキンソン病のマウスモデルを作成して C4-sulfa tase薬を投与し、治療後における遺伝子発現と組織の様子を比較検討した。  In this example, a mouse model of Parkinson's disease in which donomin neurons were selectively denatured with 1-methy 卜 4-pheny 卜 2,3-dihydropyridinium (MPDP) was administered and treated with C4-sulfatase drug. Later, gene expression and tissue appearance were compared.
[0194] 妊娠 14日目 C57BL6J/JcLマウス(日本クレア社製)を飼育、出産させ、 8週令 C57B L6J/JcLマウス雌(日本クレア社製)各々に C4-sulfatase (生化学工業) 4 U/mlを 100 1腹腔内投与した。その後、 2日、 3日そして 4日後の 3度にわたりドーパミンニューロ ンのみを選択的に破壊する MPDP (Sigma Aldrich Japan社製)を 30 mg/kgで体内へ 投与したものを飼育した。実験 8日目に BrdU 5 mg/mL(ZyMED Laboratory. Inc社製) 100 1を尾静脈内投与処置し、 1時間後解剖し、脳を摘出し、免疫染色用サンプル、 遺伝子発現解析用サンプルを得た。  [0194] Day 14 of pregnancy C57BL6J / JcL mice (CLEA Japan, Inc.) were bred and given birth, and C4-sulfatase (Seikagaku Corporation) 4 U for each 8 week-old female C57B L6J / JcL mouse (CLEA Japan) 100 ml / ml was administered intraperitoneally. Thereafter, MPDP (manufactured by Sigma Aldrich Japan) that selectively destroys only dopamine neuron at 30 mg / kg was reared for 3 times after 2 days, 3 days and 4 days. On the 8th day of the experiment, BrdU 5 mg / mL (manufactured by ZyMED Laboratory. Inc.) 100 1 was administered into the tail vein, dissected one hour later, the brain was removed, and a sample for immunostaining and a sample for gene expression analysis were prepared. Obtained.
[0195] このパーキンソン病モデルマウスの脳組織サンプルを使用して CSPGの沈着抑制効 果につ!/、て比較検討を行った。得られた脳組織を凍結用包埋剤 OCTコンパウンド( Miles社製)に包埋して液体窒素にて凍結ブロックを作成した。その凍結ブロックから クリオスタツト(Microm社製)を用いて厚さ 10 μ mの切片を作成した。得られた切片を アセトン(Sigma Aldrich Japan社製)で 10分間固定後、リン酸緩衝液で洗浄し、さらに 一次抗体として抗コンドロイチン硫酸プロテオダリカン(CSPG)抗体(クローン CS56、 マウスモノクローナル抗体、 lO ^ g/ml;生化学工業社製)を添加し、室温で 1時間反 応させた。続いて、ヒストファインマウススティンキット(ニチレイ社製;マウスモノクロ一 ナル抗体に対して使用)を用いて二次抗体反応を行った後、 DAB基質 (ニチレイ社 製)を添加し酵素色素反応を行った。この標本を光学顕微鏡 (ライカ社製)にて観察 した。この組織像を図 7として示した。未治療群における陽性のシグナルは歯状回付 近にて対照群の CSPG蓄積より強く示していることがわかった。そして、 C4-sulfatase 治療群における CSPGの蓄積が抑制されていることが見受けられた。つまり、以上の 結果を考察すると、 MPDP誘導型パーキンソン病マウスモデルの脳組織における CSP Gの沈着は C4-sulfataseの生体内投与において抑制される所見が明らかにされた。[0195] Using a brain tissue sample of this Parkinson's disease model mouse, CSPG deposition suppression effect was compared! The obtained brain tissue was embedded in a freezing embedding agent OCT compound (manufactured by Miles), and a frozen block was prepared with liquid nitrogen. A section having a thickness of 10 μm was prepared from the frozen block using a cryostat (manufactured by Microm). The obtained sections were fixed with acetone (manufactured by Sigma Aldrich Japan) for 10 minutes, washed with a phosphate buffer, and further anti-chondroitin sulfate proteodarican (CSPG) antibody (clone CS56, as a primary antibody). Mouse monoclonal antibody (lO ^ g / ml; manufactured by Seikagaku Corporation) was added and reacted at room temperature for 1 hour. Subsequently, after performing a secondary antibody reaction using a histofine mouse stin kit (manufactured by Nichirei; used for mouse monoclonal antibody), DAB substrate (manufactured by Nichirei) was added to perform an enzyme dye reaction. It was. This specimen was observed with an optical microscope (Leica). This tissue image is shown in FIG. It was found that the positive signal in the untreated group was stronger than the control group's CSPG accumulation near the dentate gyrus. It was also found that CSPG accumulation was suppressed in the C4-sulfatase treatment group. In other words, considering the above results, it was found that the deposition of CSP G in the brain tissue of the MPDP-induced Parkinson's disease mouse model was suppressed by in vivo administration of C4-sulfatase.
WMH C4- sulfaase几 i によろ 力 ^ ffl t l 力 の比 討 実施例 7より摘出した臓器 (脳) 50 mg当たりに対し、 RNA-Bee (TEL-TEST社製) 1 mLを力 Pえ、電動ホモジナイザー(DIGITAL HOMOGENIZER, ASONE社製)にて粉 砕させた後、 chloroform 200 μ l(Sigma Aldrich Japan社製)を加え穏やかに混合後、 約 5分氷冷し、 12,000rpm、 4°C、 15分間 遠心分離機(Centrifoge 5417R、 eppendorf 社製)を用い遠心分離を行った。遠心分離後の上澄み液 500 Lを別のエツペンドル フチューブに移し、上澄み液と同等量の isopropannol 500 μ L(Sigma Aldrich Japan社 製)を加え混合後、 l Lの glycogen(Invitrogen社製)を加え、 15分間氷冷した。氷冷 15 分後、 12,000rpm、 4°C、 15分間遠心し、その後、 75% Ethanol 1000 l(Sigma Aldrich Japan社製)で 3回洗浄して得られた RNA沈殿物を 30分間〜 1時間、自然乾燥させた 後、大塚蒸留水 50 大塚製薬社製)に溶解させ、さらに大塚蒸留水 (大塚製薬社製) にて 100倍希釈し、 UVプレート(Corning Costar社製)上でプレートリーダー (POWER Wave XS、 BIO-TEK社製)により抽出したサンプル中の RNA濃度を算出した。次に、 R T reaction(cDNA合成)を行うため以下の手技を行った。算出して得られた RNAサンプ ノレを 500 rig/20 ^ 1の濃度に調整し、 68°C、 3分間、 BLOCK INCUBATOR(ASTEC製) にて加温し、 10分間、氷冷した。氷冷後、予め調製していた RT Pre Mix液 (組成: 25 mM MgCl 18.64 μ 1 (Invitrogen社製)、 5 X Buffer 20 μ 1 (Invitrogen社製)、 0. 1M DTT 6.6 μ 1 (Invitrogen社製)、 10 mM dNTP mix 10 μ 1 (Invitrogen社製)、 RNase Inhibitor 2 μ 1 (Invitrogen社製)、 MMLV Reverse transcriptase 1.2 μ 1 (Invitrogen社 )、 Rando m primer 2 μ 1 (Invitrogen社製)、滅菌蒸留水 19.56 μ 1 (大塚蒸留水:大塚製薬社製) を 80 1加え BLOCK INCUBATOR(ASTEC社製)にて 42°C、 1時間、加温反応させ、 1 時間後、 BLOCK INCUBATOR(ASTEC社製)にて 99°C、 5分間、加熱した後、氷冷し 求める cDNA 100 1を作製し、合成して得られた cDNAを用いて、以下の組成で Red- time PCR反応を行った。 SYBR Premix EX taq 12.5 ^ 1 (TAKARA社製)、滅菌蒸留水 11.3 1 (大塚蒸留水),プライマー 0.1 1 (50 pmol/ μ 1), cDNAl 1を混合させ、 Realtime PCR Dice (TAKARA社製)により 95°C 5秒, 60°C 30秒で 40サイクル反応させた。 反応終了後、 β -actinを補正遺伝子として炎症、線維化マーカーである TNF- aと TG F- βの遺伝子発現を解析した。 Comparison of force ^ ffl tl force with WMH C4-sulfaase 几 i For each 50 mg of organ (brain) extracted from Example 7, force 1 mL of RNA-Bee (TEL-TEST) into electric power. After pulverization with a homogenizer (DIGITAL HOMOGENIZER, ASONE), add 200 μl of chloroform (Sigma Aldrich Japan) and mix gently, then ice-cool for about 5 minutes, 12,000 rpm, 4 ° C, Centrifugation was performed using a centrifuge (Centrifoge 5417R, eppendorf) for 15 minutes. Transfer 500 L of the supernatant after centrifugation to another Eppendorf tube, add 500 μL of isopropannol (Sigma Aldrich Japan) equivalent to the supernatant, mix, and then add l L of glycogen (Invitrogen). Ice-cooled for 15 minutes. After 15 minutes of ice cooling, centrifuge for 15 minutes at 12,000 rpm at 4 ° C, and then wash with 75% Ethanol 1000 l (manufactured by Sigma Aldrich Japan) three times for 30 minutes to 1 After natural drying for a period of time, dissolve in Otsuka distilled water 50 (Otsuka Pharmaceutical Co., Ltd.), further dilute 100 times with Otsuka distilled water (Otsuka Pharmaceutical Co., Ltd.), and plate reader on UV plate (Corning Costar Co., Ltd.) The RNA concentration in the sample extracted by (POWER Wave XS, manufactured by BIO-TEK) was calculated. Next, the following procedure was performed to perform RT reaction (cDNA synthesis). The calculated RNA sample was adjusted to a concentration of 500 rig / 20 ^ 1, heated at 68 ° C for 3 minutes with BLOCK INCUBATOR (ASTEC), and ice-cooled for 10 minutes. RT Pre Mix solution (composition: 25 mM MgCl 18.64 μ 1 (Invitrogen)), 5 X Buffer 20 μ 1 (Invitrogen), 0.1 M DTT 6.6 μ 1 (Invitrogen) ), 10 mM dNTP mix 10 μ1 (Invitrogen), RNase Inhibitor 2 μ1 (Invitrogen), MMLV Reverse transcriptase 1.2 μ1 (Invitrogen), Rando Add m primer 2 μ 1 (Invitrogen), sterile distilled water 19.56 μ 1 (Otsuka distilled water: Otsuka Pharmaceutical Co., Ltd.) 80 1 and heat reaction at 42 ° C for 1 hour using BLOCK INCUBATOR (ASTEC) 1 hour later, heat at 99 ° C for 5 minutes with BLOCK INCUBATOR (ASTEC), then cool on ice to prepare cDNA 1001 to be obtained, and use the cDNA obtained by synthesis to Red-time PCR reaction was performed with the composition. SYBR Premix EX taq 12.5 ^ 1 (TAKARA), sterile distilled water 11.3 1 (Otsuka distilled water), primer 0.1 1 (50 pmol / μ 1), cDNAl 1 were mixed, and Realtime PCR Dice (TAKARA) The reaction was carried out for 40 cycles at 95 ° C for 5 seconds and 60 ° C for 30 seconds. After completion of the reaction, gene expression of TNF-a and TGF-β, which are inflammation and fibrosis markers, was analyzed using β-actin as a correction gene.
[0197] 図 8に、 TNF- aと TGF- βの遺伝子発現が C4_sulfatase治療によって抑制されたこ とを示した。今回用いた β -actin, TNF- a, TGF- β primer (Forward, Reverse) (TA KARA社製)の配列情報を以下に示す。  [0197] Figure 8 shows that TNF-a and TGF-β gene expression was suppressed by C4_sulfatase treatment. The sequence information of β-actin, TNF-a, and TGF-β primer (Forward, Reverse) (TA KARA) used here is shown below.
[0198] β -actin: TAKARA社製  [0198] β-actin: TAKARA
F: CATCCGTAAAGACCTCTATGCCAAC (配列番号: 84)  F: CATCCGTAAAGACCTCTATGCCAAC (SEQ ID NO: 84)
R: ATGGAGCCACCGATCCACA (酉己歹 IJ番号: 85)  R: ATGGAGCCACCGATCCACA (IJ number: 85)
[0199] Tumor Necrosis Factor (TNF- a ): TAKARA社製  [0199] Tumor Necrosis Factor (TNF-a): manufactured by TAKARA
R: CCTGGTTGGCTGCTTGCTT (配歹 IJ番号 : 87) R: CCTGGTTGGCTGCTTGCTT (Distributed IJ number: 87)
[0200] Transforming growth factor-beta (TGF- β ): TAKARA社製 [0200] Transforming growth factor-beta (TGF-β): TAKARA
F: GTGTGGAGCAACATGTGGAACTCTA (配列番号: 88)  F: GTGTGGAGCAACATGTGGAACTCTA (SEQ ID NO: 88)
R: TTGGTTCAGCCACTGCCGTA (配列番号: 89)  R: TTGGTTCAGCCACTGCCGTA (SEQ ID NO: 89)
[0201] 実施例 9 C4-sulfatase処置におけるドーパミンニューロンの活性効果の比較検討 [0201] Example 9 Comparative study of the effect of dopamine neurons on C4-sulfatase treatment
以上の実施例で示された結果を最終的に明らかにするために、まずドーパミンニュ 一ロンの必須遺伝子である Nurrl遺伝子の発現を実施例 8と同様な方法で発現解析 を行った。その結果、図 9に示すように、未治療群と比べて有意のある上昇を示した。  In order to finally clarify the results shown in the above examples, first, expression analysis of Nurrl gene, which is an essential gene of dopamine neuron, was performed in the same manner as in Example 8. As a result, as shown in FIG. 9, there was a significant increase compared to the untreated group.
[0202] 更に、得られた組織サンプル切片をドーパミンニューロンのマーカーとする抗チロシ ンハイドロキシラーゼ抗体にてドーパミンニューロンを染め、組織所見を比較検討し た。このチロシンハイドロキシラーゼ(TH: thyrosine hydroxylase)はドーパミン前駆体 をドーノ ミンへ転換する酵素である。実施例 7と同様の方法で得られた切片を 4%PF Aリン酸緩衝液け力ライテスタ社製)で 10分間固定後、脱イオン水で洗浄し、一次抗 体としてラビットポリクローナル抗チロシンハイドロキシラーゼ抗体(1: 50希釈; Calbioc hem社)を添加し、室温で 1時間反応させた。次に二次抗体である Alexa488標識抗ラ ビット-ロバ抗体(1 : 200希釈; Invitrogen社)を添加し、室温で 30分反応させた。図 10 に未治療群、 C4-sulfatase治療群に対する組織画像を示した。未治療群の黒質付近 においては陰性のシグナルとして現れた。これは、 MPDPにより選択的にドーパミン二 ユーロンが破壊されたことが示唆される所見である。一方、 C4-sulfatase処置を行った 処置群におけるシグナルは強く現れていることが確認できた。つまり、 C4-sulfataseの 生体内投与によって実施例 5にて示したァストロサイトの活性化に伴いドーパミンニュ 一ロンの機能回復が期待できる結論が得られた。 (THは緑色) [0202] Furthermore, the obtained tissue sample sections were stained with an anti-tyrosine hydroxylase antibody using a dopamine neuron marker, and the tissue findings were compared. This tyrosine hydroxylase (TH) is a dopamine precursor It is an enzyme that converts to dolomine. The section obtained in the same manner as in Example 7 was fixed with 4% PFA phosphate buffer solution (manufactured by Leytester) for 10 minutes, washed with deionized water, and rabbit polyclonal anti-tyrosine hydroxylase as the primary antibody. Antibody (1:50 dilution; Calbiochem) was added and allowed to react for 1 hour at room temperature. Next, Alexa488-labeled anti-rabbit-donkey antibody (1: 200 dilution; Invitrogen), which is a secondary antibody, was added and reacted at room temperature for 30 minutes. Untreated group 10 showed tissue image for C4-sulfatas e treatment group. A negative signal appeared near the substantia nigra of the untreated group. This suggests that MPDP selectively destroyed dopamine di eurone. On the other hand, it was confirmed that a strong signal appeared in the treatment group treated with C4-sulfatase. That is, it was concluded that the functional recovery of dopamine neuron can be expected with the activation of astrocytes shown in Example 5 by in vivo administration of C4-sulfatase. (TH is green)
[0203] 以下に、 Nurrlにおけるプライマーの配列を示す。  [0203] The sequence of primers in Nurrl is shown below.
Nuclea receptor subfamily 4 GroupA member2 (Nurrl)  Nuclea receptor subfamily 4 GroupA member2 (Nurrl)
F:CTGCCCTGGCTATGGTCACA (配歹 IJ番号: 90)  F: CTGCCCTGGCTATGGTCACA (Distributed IJ number: 90)
R: AGACAGGTAGTTGGGTCGGTTCA (配列番号: 91)  R: AGACAGGTAGTTGGGTCGGTTCA (SEQ ID NO: 91)
[0204] 実施例 10 GalNAc4S-6ST siRNAにおける外来因子からの保 If作用  [0204] Example 10 Preservative If Effect from Foreign Factors in GalNAc4S-6ST siRNA
実施例 7、 8、 9と同様に MPDP投与によって誘因されるパーキンソン病モデルにお ける C4-sulfataseと GalNAc4S_6ST siRNA治療群と未治療群の生存率につ!/、てカプラ ンマイヤーの生存曲線を用いて比較を行った。図 11に示すように、 7日目における生 存率は、未治療群が 16.7%、 C4-sufatase治療群が 50%、 GalNAc4S-6ST siRNA治療 群が 71.4%であった。つまり C4-sulfataseや GalNAc4S_6ST siRNAによる処置によって CSPGの過剰発現及び過剰沈着を防ぐことで炎症細胞の誘導に関わるケモカインの 吸着を防ぎ、 CSPGの沈着が引き寄せる炎症細胞における脳組織の破壊を防ぐメカ ニズムによって感染や薬剤性の脳炎、外傷などの侵襲に対する保護作用を示唆する ことができる結果となった。  As in Examples 7, 8, and 9, the survival rate of C4-sulfatase and GalNAc4S_6ST siRNA treated group and untreated group in Parkinson's disease model induced by MPDP administration! /, Using the Kaplan Meier survival curve And compared. As shown in FIG. 11, the survival rate on day 7 was 16.7% in the untreated group, 50% in the C4-sufatase treated group, and 71.4% in the GalNAc4S-6ST siRNA treated group. In other words, treatment with C4-sulfatase or GalNAc4S_6ST siRNA prevents the overexpression and over-deposition of CSPG, thereby preventing the adsorption of chemokines related to the induction of inflammatory cells, and prevents the destruction of brain tissue in the inflammatory cells attracted by the deposition of CSPG. The results suggest that it can protect against invasion such as infection, drug-induced encephalitis, and trauma.
[0205] GalNAc4S-6ST siRNAの配列を以下に示す。  [0205] The sequence of GalNAc4S-6ST siRNA is shown below.
[GalNAc4S-6ST siRNA配歹 IJ] iGene社製  [GalNAc4S-6ST siRNA delivery IJ] iGene
5' -GGAGCAGAGCAAGAUGAAUACAAUC—3' (酉己歹 IJ番号: 92) 5 ' -CCUCGUCUCGUUCUACUUAUGUUAG _3 ' (酉己歹 lj番号: 93) 5 '-GGAGCAGAGCAAGAUGAAUACAAUC—3' (酉 己 歹 IJ number: 92) 5 '-CCUCGUCUCGUUCUACUUAUGUUAG _3' (酉 己 歹 lj number: 93)
産業上の利用可能性  Industrial applicability
[0206] 本発明に係る、コンドロイチン硫酸プロテオダリカン (CSPG)の蓄積の影響を検討す る例として、コンドロイチン硫酸プロテオダリカンの側鎖であるァセチルガラタトサミン の石) ¾酸基を転移する酵素である N- acetylgalactosamine- 4-0- sulfotransferase:N-acet ylgalactosamine- 4-0- sulrotransferase-l, N- acetylgalactosamine- 4-0- sulrotransfera se~2, N-acetylgactosamine-4-sulfate 6-O-sulfotransferase (GalNAc4ST-l, GalNAc4 ST-2, GALNAC4S-6ST)の siRNAと側鎖であるコンドロイチン硫酸の分解酵素である コンドロイチナーゼ ABCは脳の視床下部におけるコンドロイチン硫酸プロテオダリカン に付随する GAG鎖の硫酸化とその蓄積を抑え、ドーパミンニューロンの細胞死を抑 制することによりパーキンソン病の治療又は予防に効果を有する。さらに、異常タンパ ク質の蓄積により発症されると考えられる神経線維性変性疾患;アルツハイマー病、 ポリグルタミン病、筋萎縮性側索硬化症、脊髄性筋萎縮症、ハンチントン病そして多 発性硬化症などに対しても本発明におけるコンドロイチン硫酸プロテオダリカンの過 剰発現および蓄積が脳機能を低下させる要因であると考える。本発明に係る神経線 維性変性抑制剤は、これまでにない作用機序、薬剤療法によって病変を有効に改善 出来る事から、患者の QOLのさらなる向上、医療に役立つ優れた療法と成り得る。  [0206] As an example of examining the effect of chondroitin sulfate proteodarican (CSPG) accumulation according to the present invention, the side chain of chondroitin sulfate proteodarican (acetilgalatatosamine stone) ¾ acid group was transferred. N-acetylgalactosamine- 4-0- sulfotransferase: N-acet ylgalactosamine- 4-0- sulrotransferase-l, N-acetylgalactosamine- 4-0- sulrotransfera se ~ 2, N-acetylgactosamine-4-sulfate 6-O Chondroitinase ABC, a degrading enzyme of chondroitin sulfate, which is a side chain of the siRNA of -sulfotransferase (GalNAc4ST-l, GalNAc4 ST-2, GALNAC4S-6ST), is a GAG chain associated with chondroitin sulfate proteodarican in the hypothalamus of the brain It is effective in treating or preventing Parkinson's disease by suppressing the sulfation and accumulation of and inhibiting the cell death of dopamine neurons. In addition, neurofibrotic degenerative diseases thought to be caused by the accumulation of abnormal proteins; Alzheimer's disease, polyglutamine disease, amyotrophic lateral sclerosis, spinal muscular atrophy, Huntington's disease and multiple sclerosis Therefore, it is considered that overexpression and accumulation of chondroitin sulfate proteodalycan in the present invention is a factor that reduces brain function. Since the neurofibrotic degeneration inhibitor according to the present invention can effectively improve lesions by an unprecedented mechanism of action and drug therapy, it can be an excellent therapy useful for further improvement of patient QOL and medical treatment.
[0207] 本明細書中で引用した全ての刊行物、特許及び特許出願はその全体を参照により 本明細書中に組み入れるものとする。  [0207] All publications, patents, and patent applications cited herein are hereby incorporated by reference in their entirety.

Claims

請求の範囲 The scope of the claims
[I] コンドロイチン硫酸プロテオダリカンの生成もしくは蓄積を阻害する物質を有効成分 として含む、神経線維性変性抑制剤。  [I] A neurofibrotic degeneration inhibitor comprising as an active ingredient a substance that inhibits the production or accumulation of chondroitin sulfate proteodarican.
[2] 前記物質が、コンドロイチン硫酸プロテオダリカン分解促進作用を有する物質であ る、請求項 1に記載の薬剤。  [2] The drug according to claim 1, wherein the substance is a substance having a chondroitin sulfate proteodarican degradation promoting action.
[3] 前記物質が、コンドロイチン硫酸プロテオダリカン合成阻害作用を有する物質であ る、請求項 1に記載の薬剤。 [3] The drug according to claim 1, wherein the substance is a substance having a chondroitin sulfate proteodarican synthesis inhibitory action.
[4] 前記物質が、コンドロイチン硫酸プロテオダリカン脱硫酸化作用を有する物質であ る、請求項 1に記載の薬剤。 [4] The drug according to claim 1, wherein the substance is a substance having a chondroitin sulfate proteodarican desulfation effect.
[5] 前記物質が、コンドロイチン硫酸プロテオダリカン硫酸化阻害作用を有する物質で ある、請求項 1に記載の薬剤。 [5] The drug according to claim 1, wherein the substance is a substance having an inhibitory action on chondroitin sulfate proteodarican sulfation.
[6] 脳においてコンドロイチン硫酸プロテオダリカンの生成もしくは蓄積が阻害されるこ とを特徴とする、請求項 1〜5のいずれかに記載の薬剤。 [6] The agent according to any one of [1] to [5], wherein production or accumulation of chondroitin sulfate proteodalycan is inhibited in the brain.
[7] 神経線維性変性疾患の治療用または予防用の、請求項;!〜 6の!/、ずれかに記載の 薬剤。 [7] The drug according to any one of claims;! To 6, which is used for treatment or prevention of neurofibrotic degenerative diseases.
[8] 前記神経線維性変性疾患が脳脊髄神経または末梢神経線維性変性疾患である、 請求項 7に記載の薬剤。  8. The drug according to claim 7, wherein the neurofibrotic degenerative disease is cerebrospinal nerve or peripheral nerve fibrotic degenerative disease.
[9] 前記神経線維性変性疾患がパーキンソン病、アルツハイマー病、筋萎縮性側索硬 化症、ポリグルタミン病、脊髄性筋萎縮症、ハンチントン病、または多発性硬化症であ る、請求項 7に記載の薬剤。 [9] The neurofibrotic degenerative disease is Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, polyglutamine disease, spinal muscular atrophy, Huntington's disease, or multiple sclerosis. The drug described in 1.
[10] コンドロイチン硫酸プロテオダリカンの生成もしくは蓄積を阻害する物質を有効成分 として含む、脳組織におけるドーパミンニューロン再生促進剤。 [10] An agent for promoting the regeneration of dopamine neurons in brain tissue, comprising as an active ingredient a substance that inhibits the production or accumulation of chondroitin sulfate proteodarican.
[I I] コンドロイチン硫酸プロテオダリカンの生成もしくは蓄積を阻害する物質を有効成分 として含む、炎症細胞における脳組織破壊防止剤。  [I I] An agent for preventing destruction of brain tissue in inflammatory cells, containing as an active ingredient a substance that inhibits the production or accumulation of chondroitin sulfate proteodarican.
[12] 被検試料から、コンドロイチン硫酸プロテオダリカンの生成もしくは蓄積を阻害する 作用を有する物質を選択することを特徴とする、神経線維性変性抑制剤のスクリー二 ング方法。  [12] A screening method for a neurofibrotic degeneration inhibitor, which comprises selecting a substance having an action of inhibiting the production or accumulation of chondroitin sulfate proteodarican from a test sample.
[13] 以下の(a)〜(d)の!/、ずれかに記載の作用を有する物質を選択する工程を含む、 請求項 12に記載のスクリーニング方法。 [13] The following (a) to (d) are included, including a step of selecting a substance having the action described in any one of The screening method according to claim 12.
(a)コンドロイチン硫酸プロテオダリカンの分解促進作用 ( a ) Promoting the degradation of chondroitin sulfate proteodarican
(b)コンドロイチン硫酸プロテオダリカンの合成阻害作用  (b) Inhibition of chondroitin sulfate proteodarican synthesis
(c)コンドロイチン硫酸プロテオダリカンの脱硫酸化作用  (c) Desulfation effect of chondroitin sulfate proteodarican
(d)コンドロイチン硫酸プロテオダリカンの硫酸化阻害作用  (d) Inhibitory effect of chondroitin sulfate proteodarican on sulfation
[14] 以下の工程 (a)〜(c)を含む、神経線維性変性抑制剤のスクリーニング方法。  [14] A screening method for a neurofibrotic degeneration inhibitor comprising the following steps (a) to (c).
(a)コンドロイチン硫酸プロテオダリカンまたはその一部と被検化合物を接触させるェ 程 ( a ) The process of contacting the test compound with chondroitin sulfate proteodarican or a part thereof
(b)コンドロイチン硫酸プロテオダリカンまたはその一部の存在量を測定する工程 (b) a step of measuring the abundance of chondroitin sulfate proteodarican or a part thereof
(c)被検化合物の非存在下において測定した場合と比較して、存在量を低下させる 物質を選択する工程 (c) A step of selecting a substance that reduces the abundance compared to the case where measurement is performed in the absence of the test compound.
[15] 前記神経線維性変性抑制剤が、神経線維性変性疾患の治療用または予防用であ る、請求項 12〜; 14のいずれかに記載のスクリーニング方法。  [15] The screening method according to any one of [12] to [14], wherein the neurofibrotic degeneration inhibitor is used for treatment or prevention of a neurofibrotic degenerative disease.
PCT/JP2007/067375 2006-09-08 2007-09-06 Nerve fiber degeneration inhibitor WO2008029871A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
JP2006244532 2006-09-08
JP2006-244532 2006-09-08
JPPCT/JP2006/323678 2006-11-28
PCT/JP2006/323678 WO2008029493A1 (en) 2006-09-08 2006-11-28 Nerve fiber degeneration inhibitor

Publications (1)

Publication Number Publication Date
WO2008029871A1 true WO2008029871A1 (en) 2008-03-13

Family

ID=39157296

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2007/067375 WO2008029871A1 (en) 2006-09-08 2007-09-06 Nerve fiber degeneration inhibitor

Country Status (1)

Country Link
WO (1) WO2008029871A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2876162A4 (en) * 2012-07-17 2016-03-16 Stelic Institute&Co Mucosal healing promoter

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
CHAU C.H. ET AL.: "Chondroitinase ABC enhances axonal regrowth through Schwann cell-seeded guidance channels after spinal cord injury", FASEB JOURNAL, 20 November 2003 (2003-11-20), XP003008297, Retrieved from the Internet <URL:http://www.fasebj.org/cgi/reprint/03-0196fjev1> *
DEWITT D.A. ET AL.: "Chondroitin sulfate proteoglycans are a common component of neuronal inclusions and astrocytic reaction in neurodegenerative diseases", BRAIN RESEARCH, vol. 656, no. 1, 1994, pages 205 - 209, XP003021627 *
MASSEY J.M. ET AL.: "Chondroitinase ABC digestion of the perineuronal net promotes functional collateral sprouting in the cuneate nucleus after cervical spinal cord injury", JOURNAL OF NEUROSCIENCE, vol. 26, no. 16, 2006, pages 4406 - 4414, XP003021629 *
MOON L.D.: "Regeneration of CNS axons back to their target following treatment of adult rat brain with chondroitinase ABC", NATURE NEUROSCIENCE, vol. 4, no. 5, 2001, pages 465 - 466, XP003021628 *
PIZZORUSSO T. ET AL.: "Reactivation of ocular dominance plasticity in the adult visual cortext", SCIENCE, vol. 298, 2002, pages 1248 - 1251, XP002245002 *
ROLLS A. ET AL.: "A sulfated disaccharide derived from chondroitin sulfate proteoglycan protects against inflammation-associated neurodegeneration", FASEB JOURNAL, 5 January 2006 (2006-01-05), XP003021630, Retrieved from the Internet <URL:http://www.fasebj.org/cgi/reprint/05-4540fjev1> *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2876162A4 (en) * 2012-07-17 2016-03-16 Stelic Institute&Co Mucosal healing promoter
EP3121279A1 (en) * 2012-07-17 2017-01-25 Stelic Institute&Co. Mucosal healing promoter
EP3693462A1 (en) * 2012-07-17 2020-08-12 Stelic Institute&Co. Mucosal healing promoter
EP3878962A1 (en) * 2012-07-17 2021-09-15 Stelic Institute & Co. Mucosal healing promoter

Similar Documents

Publication Publication Date Title
US20220340910A1 (en) Sugar chain-related gene and use thereof
JP4147264B2 (en) Neurofibrotic degeneration inhibitor
JP4101276B2 (en) Chronic obstructive pulmonary disease improving agent
WO2008020489A1 (en) Remedy for inflammatory bowel disease
AU2012322762B2 (en) Method of diagnosis, prognostic or treatment of neurodegenerative diseases
JP2007528202A (en) Compositions and methods for nerve regeneration
JP2012527883A (en) Pyroline-5-carboxylate reductase 1 mutein
JPWO2007049424A1 (en) Liver fibrosis inhibitor
WO2008029871A1 (en) Nerve fiber degeneration inhibitor
WO2008026298A1 (en) Remedy for kidney disease
JP2009234916A (en) Insulin resistance reducer based on chondroitin sulfate proteoglycan accumulation regulation
WO2008029868A1 (en) Ocular fibrous neovascularization inhibitor
Sevegnani et al. Parkin R274W mutation affects muscle and mitochondrial physiology
Mirvis Heterogenous Dynamics and Mechanisms of Mammalian Primary Cilia Disassembly
JP2009108033A (en) Hepatic fibrosis-suppressing agent
Alabed The role of RhoA interacting proteins in the Nogo signalling pathway of axon outgrowth inhibition

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: 07806816

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: 07806816

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: JP