WO2020213982A1 - Composition for prevention or treatment of neurodegenerative diseases using cd9 and method for screening therapeutic agent for neurodegenerative diseases - Google Patents

Composition for prevention or treatment of neurodegenerative diseases using cd9 and method for screening therapeutic agent for neurodegenerative diseases Download PDF

Info

Publication number
WO2020213982A1
WO2020213982A1 PCT/KR2020/005155 KR2020005155W WO2020213982A1 WO 2020213982 A1 WO2020213982 A1 WO 2020213982A1 KR 2020005155 W KR2020005155 W KR 2020005155W WO 2020213982 A1 WO2020213982 A1 WO 2020213982A1
Authority
WO
WIPO (PCT)
Prior art keywords
disease
protein
neurodegenerative diseases
inhibitor
activity
Prior art date
Application number
PCT/KR2020/005155
Other languages
French (fr)
Korean (ko)
Inventor
황정후
Original Assignee
황정후
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 황정후 filed Critical 황정후
Publication of WO2020213982A1 publication Critical patent/WO2020213982A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6872Intracellular protein regulatory factors and their receptors, e.g. including ion channels
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • G01N33/6896Neurological disorders, e.g. Alzheimer's disease
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/136Screening for pharmacological compounds
    • 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
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70596Molecules with a "CD"-designation not provided for elsewhere in G01N2333/705
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value

Definitions

  • the present invention relates to a composition for preventing or treating neurodegenerative diseases using CD9 and a screening method for a therapeutic agent for neurodegenerative diseases, and more particularly, to a prevention or treatment of neurodegenerative diseases comprising an inhibitor of CD9 protein expression or activity as an active ingredient
  • a pharmaceutical composition for use and (a) treating a test substance on cells expressing CD9 protein or mRNA; (b) measuring the expression level of CD9 protein or mRNA in the cells and control cells not treated with the test substance; And (c) selecting a test substance that reduces the expression level of CD9 protein or mRNA compared to the control cells as a candidate substance for treating neurodegenerative diseases.
  • Neurodegenerative diseases are associated with symptoms when nerve cells degenerate, lose function, and often die. Patients with neurodegenerative diseases may experience extreme degeneration in cognitive or motor abilities, and because these diseases are primarily progressive, their quality of life and expectations for life may be significantly reduced as a result. .
  • Parkinson's Disease PD
  • AD Alzheimer's disease
  • ALS amyotrophic lateral sclerosis
  • HD Huntington's disease
  • Frontotemporal Dementia cortical-basal ganglia.
  • Degeneration Cortico Basal Degeneration
  • PSP progressive supranuclear palsy
  • dementia is a disease that causes the most extensive cell damage and accompanies degenerative mental disorders.
  • major symptoms such as memory impairment and loss of judgment are well known.
  • Dementia can be broadly divided into vascular dementia caused by stenosis or blockage of cerebrovascular blood vessels, Alzheimer's dementia, which is known to be caused by accumulation of beta-amyloid protein in the brain, and mixed dementia caused by a combination of these two causes.
  • Alzheimer's dementia accounts for the largest proportion of dementia patients, and a recent study reports that 1 in 85 people will have the disease by 2050, of which 43% need special care. (Prabhulkar S, Piatyszek R, et al. J Neurochem., 2012, 122, 374-381).
  • Among neurodegenerative diseases, Alzheimer's disease is largely classified into hereditary Alzheimer's disease and sporadic Alzheimer's disease (SAD).
  • SAD sporadic Alzheimer's disease
  • Hereditary Alzheimer's disease accounts for 5-10% of all Alzheimer's patients.
  • the hyperphosphorylated, aggregated tau is a central neuropathological feature of Alzheimer's disease and many human neurodegenerative diseases.
  • significant reports have been published on the role of tau in recent years.
  • the development of various tau transgenic animal models has made significant progress in tau dependent pathology, including the propagation of tau pathology from neurons to other cells and interactions between A ⁇ and tau. According to this pathology and tau-related biomarkers, new treatments have been obtained. Nevertheless, the etiological factors leading to neurodegeneration and the exact mechanism of tau-mediated neurotoxicity still need to be elucidated, and the exact cause of the onset is not known to date (Ann Transl Med. 2018 May; 6(10). ): 175.)
  • Alzheimer's disease pathological features include senile plaques that accumulate outside of nerve cells, neurofibrilary tangles that look like tangled bundles of threads in the cell body of nerve cells, and neuroal loss. And the like. These pathological features appear in both hereditary Alzheimer's disease and sporadic Alzheimer's disease. Among them, a toxic protein called amyloid ⁇ peptide (A ⁇ ) has been revealed as a major constituent of senile spots. Amyloid beta peptides are insoluble peptides consisting of 40 to 42 amino acids resulting from abnormal cleavage of amyloid proproteins.
  • amyloid beta peptide is regarded as a major pathogenic material for Alzheimer's disease.
  • the overall pathogenesis of Alzheimer's disease is that when mutations in the presenilin 1 and 2 genes (PS 1 and 2) occur, the amyloid proprotein is abnormally cleaved by ⁇ -secretase and amyloid beta peptide is produced. do. It is known that necrosis of cranial nerve cells occurs due to the produced amyloid beta peptide, which causes Alzheimer's disease.
  • CD9 is a tetraspanin-based cell membrane glycoprotein having a molecular weight of 24 to 27 kDa, and there are 34 tetraspanins in mammals, and 33 of them have been identified in humans.
  • Each tetraspanin contains cell adhesion and migration, platelet activity and aggregation, fusion of egg and sperm during fertilization in mammals, development and metastasis of cancer, humoral immune response and allergic response, human immunodeficiency virus-1 (HIV It is known as an antigen that plays an important role in -1) and influenza virus replication.
  • the present inventors have repeatedly conducted extensive research to develop a new treatment for neurodegenerative diseases, and found that inhibition of the expression or activity of the CD9 protein can improve symptoms such as memory loss caused by degenerative brain lesions.
  • the invention was completed.
  • compositions for the prevention or treatment of neurodegenerative diseases consisting of an inhibitor of the expression or activity of the CD9 protein as an active ingredient.
  • compositions for the prevention or treatment of neurodegenerative diseases consisting essentially of an inhibitor of expression or activity of a CD9 protein as an active ingredient.
  • Another object of the present invention is the steps of (a) treating a test substance on cells expressing CD9 protein or mRNA; (b) measuring the expression level of CD9 protein or mRNA in the cells and control cells not treated with the test substance; And (c) selecting a test substance that reduces the expression level of CD9 protein or mRNA compared to the control cells as a candidate substance for treating a neurodegenerative disease.
  • Another object of the present invention is to provide a use of an inhibitor of expression or activity of a CD9 protein for preparing a preparation for preventing or treating neurodegenerative diseases.
  • Another object of the present invention is to provide a method for treating neurodegenerative diseases comprising administering to an individual in need thereof an effective amount of a composition comprising an inhibitor of CD9 protein expression or activity as an active ingredient.
  • the present invention provides a pharmaceutical composition for preventing or treating neurodegenerative diseases comprising an inhibitor of expression or activity of CD9 protein as an active ingredient.
  • the present invention provides a pharmaceutical composition for preventing or treating neurodegenerative diseases comprising an inhibitor of CD9 protein expression or activity as an active ingredient.
  • the present invention provides a pharmaceutical composition for preventing or treating neurodegenerative diseases essentially consisting of an inhibitor of CD9 protein expression or activity as an active ingredient.
  • the present invention comprises the steps of: (a) treating cells expressing CD9 protein or mRNA with a test substance; (b) measuring the expression level of CD9 protein or mRNA in the cells and control cells not treated with the test substance; And (c) selecting a test substance that reduces the expression level of CD9 protein or mRNA compared to the control cells as a candidate substance for treating a neurodegenerative disease.
  • the present invention provides a use of an inhibitor of expression or activity of a CD9 protein to prepare a preparation for preventing or treating neurodegenerative diseases.
  • the present invention provides a method for treating neurodegenerative diseases comprising administering to an individual in need thereof an effective amount of a composition comprising an inhibitor of CD9 protein expression or activity as an active ingredient. to provide.
  • the present invention provides a pharmaceutical composition for preventing or treating neurodegenerative diseases comprising an inhibitor of expression or activity of CD9 protein as an active ingredient.
  • the present invention provides a pharmaceutical composition for preventing or treating neurodegenerative diseases comprising an inhibitor of CD9 protein expression or activity as an active ingredient.
  • the present invention provides a pharmaceutical composition for preventing or treating neurodegenerative diseases essentially consisting of an inhibitor of CD9 protein expression or activity as an active ingredient.
  • treatment refers to suppression of occurrence or recurrence of a disease, alleviation of symptoms, reduction of direct or indirect pathological consequences of a disease, reduction of disease progression rate, improvement of disease state, improvement, alleviation or improved prognosis. do.
  • prevention' as used in the present invention means any action that suppresses the onset or delays the progression of a disease.
  • 'Protein' is used interchangeably with'polypeptide' or'peptide', for example, refers to a polymer of amino acid residues as commonly found in proteins in nature.
  • a'fragment' of the CD9 protein refers to a peptide of a portion of the CD9 protein.
  • DNA deoxyribonucleic acid
  • RNA ribonucleic acid
  • DNA is composed of four bases: adenine (A), guanine (G), cytosine (C), and thymine (T), and RNA is composed of uracil (U) instead of thymine.
  • A forms a hydrogen bond with T or U
  • C forms a hydrogen bond with the G base, and the relationship between these bases is called'complementary'.
  • mRNA messenger RNA or messenger RNA
  • RNA messenger RNA or messenger RNA
  • the neurodegenerative disease refers to a disease related to symptoms that appear when nerve cells degenerate, lose function, and die, and Alzheimer's disease, dementia, mild cognitive impairment, Parkinson's disease, progressive nuclear paralysis, multi-system Atrophy, Olive Nucleus-Peripheral-Cerebral Atrophy (OPCA), Shy-Drager Syndrome, Stroke-Black Matter Degeneration, Huntington's Disease, Amyotrophic Lateral Sclerosis (ALS), Essential Tremors, Cortical-basal Nucleus Degeneration, Diffuse Lewy Body Disease , Parkin's-ALS-dementia complex, Niemann-Pick disease, Pick's disease, cerebral ischemia, and may be selected from the group consisting of cerebral infarction, but is not limited thereto, and most preferably Alzheimer's disease or dementia. have.
  • OPCA Olive Nucleus-Peripheral-Cerebral Atrophy
  • ALS Amyotrophic Lateral Sclerosis
  • Essential Tremors Cortical-basal Nucle
  • CD9 is a tetraspanin-based cell membrane glycoprotein having a molecular weight of 24 to 27 kDa, and there are 34 tetraspanins in mammals, and 33 of them have been identified in humans.
  • Each tetraspanin contains cell adhesion and migration, platelet activity and aggregation, fusion of egg and sperm during fertilization in mammals, development and metastasis of cancer, humoral immune response and allergic response, human immunodeficiency virus-1 (HIV It is known as an antigen that plays an important role in -1) and influenza virus replication.
  • the term'CD9' as used herein is a cell surface glycoprotein receptor of about 24 to 27 kDa belonging to a member of the tetraspanin family, and a signal that plays an important role in regulating cell development, activity, growth and motility. It is known to regulate signal transduction events. In addition, it can regulate cell adhesion and cell migration, and induce platelet activation and aggregation, which are involved in platelet-induced endothelial cell proliferation. In addition, it is involved in various phenomena within cells, such as promoting muscle cell fusion and contributing to myotube maintenance.
  • the CD9 protein in the present invention may be derived from mammals, preferably derived from humans. Most preferably, the CD9 protein in the present invention comprises the amino acid sequence of human CD9 isoform 1 (NP_001760.1) represented by SEQ ID NO: 1 or human CD9 isoform 2 (NP_001317241.1) represented by SEQ ID NO: 2 Characterized in that (NCBI Genbank accession number in parentheses).
  • the CD9 protein expression inhibitor is any one selected from the group consisting of an antisense oligonucleotide complementary to CD9 mRNA, siRNA, shRNA, miRNA, ribozyme, crispr-cas9, DNAzyme, and protein nucleic acid (PNA). It can be.
  • the CD9 mRNA most preferably comprises a nucleotide sequence of human CD9 mRNA transcript variant 1 (NM_001769.3) represented by SEQ ID NO: 3 or human CD9 mRNA transcript variant 2 (NM_001330312.1) represented by SEQ ID NO: 4 (NCBI Genbank accession number in parentheses).
  • SEQ ID NO: 4 NCBI Genbank accession number in parentheses.
  • the CD9 protein expression inhibitor according to the present invention may preferably be a shRNA that binds complementarily to CD9 mRNA and causes degradation of the mRNA.
  • the'siRNA small interfering RNA or short interfering RNA or silencing RNA
  • the'siRNA is artificially introduced into the cell to induce degradation of the mRNA of a specific gene, thereby preventing protein translation, thereby inhibiting gene expression.
  • It is a short double-stranded RNA that causes RNA interference, and is composed of 20 to 25 nucleotides complementary to a specific site of the target mRNA.
  • an antisense strand complementary to the target mRNA binds to the RNA-induced silencing complex (RISC) protein complex and binds to the target mRNA, and the argonaute protein in the RISC complex cleaves and degrades the target mRNA. It suppresses the expression of specific genes through mechanisms such as suppressing the binding of proteins and ribosomes important for protein translation with mRNA.
  • RISC RNA-induced silencing complex
  • the shRNA is a substance that induces RNA interference, and is a molecule having a double-stranded structure in the molecule and a hairpin-like structure by partially containing a palindromic nucleotide sequence in a single-stranded RNA.
  • a shRNA expression plasmid is introduced into a cell and expressed, a 21-23 base pair siRNA is generated by RNase III (ribonuclease III) enzyme called dicer in the cell to induce RNAi.
  • the siRNA or shRNA may be obtained by applying various modifications to improve the stability of the oligonucleotide in vivo, impart resistance to nuclease enzymes, and reduce non-specific immune responses.
  • the modification of the oligonucleotide is the OH group at the 2′ carbon position of the sugar structure in one or more nucleotides -CH 3 (methyl), -OCH 3 (methoxy), -NH 2 , -F, -O-2-methoxyethyl, -O-propyl, -O-2-methylthioethyl, -O-3-aminopropyl, -O-3-dimethylaminopropyl, -ON-methylacetamido or -O-dimethylami Modification by substitution with dooxyethyl; Modification in which oxygen in the sugar structure in the nucleotide is substituted with sulfur; Alternatively, one or more modifications selected from modifications of nucleotide bonds to phosphoroth
  • the CD9 protein activity inhibitor may be any one selected from the group consisting of compounds, peptides, peptide mimetics, aptamers, antibodies, natural extracts, and synthetic compounds that specifically bind to CD9 protein.
  • the pharmaceutical composition according to the present invention can be variously formulated according to the route of administration by a method known in the art together with a pharmaceutically acceptable carrier for the treatment of neurodegenerative diseases.
  • the carrier includes all kinds of solvents, dispersion media, oil-in-water or water-in-oil emulsions, aqueous compositions, liposomes, microbeads and microsomes.
  • it is formulated using a transfection reagent, and can be delivered to cells using subcutaneous, blood, bone marrow, or abdominal cavity.
  • the pharmaceutical composition according to the present invention may be administered to a patient in a pharmaceutically effective amount, that is, in an amount sufficient to prevent or relieve symptoms and treat neurodegenerative diseases.
  • a typical daily dosage may be administered in the range of about 0.01 to 1000 mg/kg, and preferably, it may be administered in the range of about 1 to 100 mg/kg.
  • the pharmaceutical composition of the present invention can be administered in one or several divided doses within a preferred dosage range.
  • the dosage of the pharmaceutical composition according to the present invention may be appropriately selected by a person skilled in the art according to the route of administration, the subject of administration, age, sex, weight, individual differences, and disease conditions.
  • the route of administration may be administered orally or parenterally.
  • Parenteral administration methods include, but are not limited to, intravenous, intramuscular, intraarterial, intramedullary, intrathecal, intracardiac, transdermal, subcutaneous, intraperitoneal, intranasal, intestinal, topical, sublingual, rectal, or pancreatic It may be intravenous administration, but is not limited thereto.
  • powders, granules, tablets, pills, dragees, capsules, solutions, gels, syrups, suspensions, wafers according to a method known in the art together with a suitable carrier for oral administration. It can be formulated in the form of such as.
  • suitable carriers include sugars including lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol and maltitol, and starches including corn starch, wheat starch, rice starch and potato starch, cellulose, Fillers such as celluloses including methyl cellulose, sodium carboxymethylcellulose and hydroxypropylmethylcellulose, gelatin, and polyvinylpyrrolidone may be included. In addition, in some cases, cross-linked polyvinylpyrrolidone, agar, alginic acid or sodium alginate may be added as a disintegrant. Furthermore, the pharmaceutical composition may further include an anti-aggregating agent, a lubricant, a wetting agent, a flavoring agent, an emulsifying agent and a preservative.
  • the pharmaceutical composition of the present invention when administered parenterally, can be formulated according to a method known in the art in the form of injections, transdermal administrations and nasal inhalants together with a suitable parenteral carrier.
  • the pharmaceutical composition can be administered by any device capable of moving the active substance to the target cell.
  • Preferred modes of administration and formulations are intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections or instillations.
  • Injectables include aqueous solvents such as physiological saline or ring gel solution, vegetable oils, higher fatty acid esters (e.g., oleic acid ethyl, etc.), alcohols (e.g., ethanol, benzyl alcohol, propylene glycol, or glycerin), and other non-aqueous solvents.
  • Stabilizers for preventing deterioration e.g., ascorbic acid, sodium hydrogen sulfite, sodium pyrosulfite, BHA, tocopherol, EDTA, etc.
  • emulsifiers e.g., ascorbic acid, sodium hydrogen sulfite, sodium pyrosulfite, BHA, tocopherol, EDTA, etc.
  • buffers for pH control emulsifiers
  • buffers for pH control e.g., buffers for pH control
  • Pharmaceutical carriers such as preservatives (eg, mercuric nitrate, thimerosal, benzalkonium chloride, phenol, cresol, benzyl alcohol, etc.) may be included.
  • suitable carriers for injections include, but are not limited to, water, ethanol, polyol (eg, glycerol, propylene glycol and liquid polyethylene glycol, etc.), a mixture thereof and/or a solvent or dispersion medium containing vegetable oil.
  • suitable carriers include isotonic solutions such as Hanks' solution, Ringer's solution, phosphate buffered saline (PBS) containing triethanolamine or sterile water for injection, 10% ethanol, 40% propylene glycol and 5% dextrose. Etc. can be used.
  • the injection may further include an isotonic agent such as sugar or sodium chloride in most cases.
  • transdermal administration ointments, creams, lotions, gels, external solutions, pasta, liniment, air rolls, and the like are included.
  • transdermal administration means that the active ingredient in an effective amount contained in the pharmaceutical composition is delivered into the skin by topically administering the pharmaceutical composition to the skin.
  • the pharmaceutical composition of the present invention may be prepared in an injectable formulation and administered by lightly pricking the skin with a 30 gauge thin injection needle or applying it directly to the skin. These formulations are described in prescriptions generally known in pharmaceutical chemistry.
  • the compounds used according to the invention can be used in pressurized packs or with suitable propellants, for example dichlorofluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. It can be conveniently delivered from a nebulizer in the form of an aerosol spray.
  • the dosage unit can be determined by providing a valve that delivers a metered amount.
  • gelatin capsules and cartridges for use in an inhaler or insufflator can be formulated to contain a powder mixture of the compound and a suitable powder base such as lactose or starch.
  • compositions according to the present invention include one or more buffers (e.g., saline or PBS), carbohythrate (e.g., glucose, mannose, sucrose or dextran), antioxidants, bacteriostatic agents, chelating agents (e.g. For example, EDTA or glutathione), adjuvants (eg, aluminum hydroxide), suspending agents, thickening agents and/or preservatives may further be included.
  • buffers e.g., saline or PBS
  • carbohythrate e.g., glucose, mannose, sucrose or dextran
  • antioxidants e.g., bacteriostatic agents, chelating agents (e.g. For example, EDTA or glutathione), adjuvants (eg, aluminum hydroxide), suspending agents, thickening agents and/or preservatives may further be included.
  • bacteriostatic agents e.g., EDTA or glutathione
  • adjuvants
  • compositions of the present invention may be formulated using methods known in the art to provide rapid, sustained or delayed release of the active ingredient after administration to a mammal.
  • composition of the present invention may be administered alone or may be administered in combination with a known compound having an effect of treating neurodegenerative diseases.
  • (c) It provides a screening method for a neurodegenerative disease treatment comprising the step of selecting a test substance that reduces the expression level of CD9 protein or mRNA compared to the control cell as a candidate substance for treatment of neurodegenerative disease.
  • the step (a) is a step of contacting cells expressing CD9 protein or mRNA in order to determine whether the test substance to be analyzed has an activity to inhibit the expression of CD9 protein or mRNA.
  • 'contacting' or'treatment' is a general meaning, and two or more agents (e.g., two polypeptides) are combined, or an agent and a cell (e.g., Protein and cell).
  • Contact can occur in vitro. For example, combining two or more agents in a test tube or other container, or combining a test agent with cells or cell lysates and a test agent. Contact can also occur in cells or in situ.
  • two polypeptides are brought into contact in a cell or cell lysate by coexpressing a recombinant polynucleotide encoding two polypeptides in a cell.
  • a protein chip or a protein array in which the protein to be tested is arranged on the surface of a stationary bed may be used.
  • the'test substance' can be used interchangeably with a test agent or agent, and any substance, molecule, element, compound ( Includes a compound, entity, or combination thereof. Examples include proteins, polypeptides, small organic molecules, polysaccharides, and polynucleotides. It may also be a natural product, a synthetic compound or a chemical compound, or a combination of two or more substances.
  • Test formulations can be synthetic or natural. The test formulations can be obtained from a wide variety of sources, including libraries of synthetic or natural compounds. Combinatorial libraries can be produced from several types of compounds that can be synthesized in a step-by-step manner.
  • Compounds of multiple combinatorial libraries can be prepared by the encoded synthetic libraries (ESL) method (WO 95/12608, WO93/06121, WO 94/08051, WO95/395503 and WO 95/30642).
  • Peptide libraries can be prepared by the phage display method (WO91/18980).
  • Libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts can be obtained from commercial sources or collected in the field.
  • Known pharmacological agents can be directed or applied to a random chemical modification, such as acylation, alkylation, esterification, amidification, to prepare structural analogues.
  • the test agent may be a naturally occurring protein or fragment thereof. Such test formulations can be obtained from natural sources, such as cell or tissue lysates. Libraries of polypeptide preparations can be produced, for example, by conventional methods or obtained from commercially available cDNA libraries.
  • the test agent may be a peptide, such as a peptide having about 5 to 30 amino acids, preferably about 5 to 20, more preferably about 7 to 15 amino acids.
  • the peptide may be a naturally occurring protein, a random peptide, or a cut product of a'biased' random peptide.
  • the test agent may be a nucleic acid.
  • the nucleic acid test agent can be a naturally occurring nucleic acid, a random nucleic acid, or a'biased' random nucleic acid.
  • cuts of prokaryotic or eukaryotic genomes can be used analogously to those described above.
  • test agent may be a small molecule (eg, a molecule having a molecular weight of about 1,000 Da or less).
  • a high throughput assay may be preferably applied to a method for screening a small molecule modulating agent. Many assays are useful for this screening.
  • the CD9-expressing cell may be a cell that internally expresses CD9, or may be a cell that overexpresses CD9 by being transformed with a recombinant expression vector containing a polynucleotide encoding CD9.
  • the cells expressing the CD9 gene may be fibroblasts.
  • Step (b) is a step of measuring the gene expression level of CD9 in cells expressing CD9 contacted with the test substance and cells expressing CD9 without contact with the test substance.
  • the measurement of CD9 mRNA expression can be performed without limitation, using conventional methods for determining the expression level in the art, and examples of analysis methods include reverse transcription polymerase chain reaction (RT-PCR), competitive RT-PCR (competitive RT-PCR), real-time RT-PCR, RNase protection assay (RPA), northern blotting, DNA microarray chip, RNA sequencing (RNA) sequencing) and the like, but are not limited thereto.
  • RT-PCR reverse transcription polymerase chain reaction
  • competitive RT-PCR competitive RT-PCR
  • RNase protection assay RNase protection assay
  • northern blotting DNA microarray chip
  • RNA sequencing (RNA) sequencing) and the like but are not limited thereto.
  • methods for measuring the expression level of CD9 protein can be used without limitation, methods known in the art, such as western blotting, dot blotting, and enzyme-linked immunosorbent assay. ), Radioactive Immunoassay (RIA), Radioimmune Diffusion Method, Ouchteroni Immunity Diffusion Method, Rocket Immunoelectrophoresis, Immunohistochemical Staining, Immunoprecipitation, Complement Fixation Assay, Flow Cytometry (FACS) or Protein Chip Method And the like, but are not limited thereto.
  • the step (c) is a step of selecting a test substance that reduces the expression level of CD9 protein or mRNA compared to the control cells as a candidate substance for treating neurodegenerative diseases.
  • the present invention provides a use of a CD9 protein expression or activity inhibitor for preparing a preparation for preventing or treating neurodegenerative diseases.
  • the present invention provides a method for treating neurodegenerative diseases comprising administering an effective amount of a composition comprising an inhibitor of expression or activity of a CD9 protein as an active ingredient to an individual in need thereof.
  • The'effective amount' of the present invention refers to an amount showing an effect of improving, treating, preventing, detecting, diagnosing, or inhibiting or reducing neurodegenerative diseases when administered to an individual, and the'individual' is an animal, preferably For example, it may be an animal including a mammal, especially a human, and may be a cell, tissue, organ, etc. derived from an animal. The individual may be a patient in need of the effect.
  • The'treatment' of the present invention generally refers to improving the symptoms of neurodegenerative diseases or neurodegenerative diseases, which may include curing, substantially preventing, or improving the condition, It includes, but is not limited to, alleviating, curing, or preventing one symptom or most of the symptoms resulting from neurodegenerative disease.
  • Substances that inhibit the expression or activity of CD9 have the effect of improving the memory of patients with neurodegenerative diseases and can be usefully used in the development of treatments for neurodegenerative diseases, and candidates for treatment of neurodegenerative diseases through analysis of the expression level of CD9 Can be screened.
  • Figure 3 shows tau phosphorylation according to treatment with anti-CD9 antibody in AD fibroblasts.
  • FIG. 4A to 4F show the effect of improving memory by administering anti-CD9Ab.
  • Each general motor activity Fig. 4A.
  • the number of alternating repetitions of mice administered with the drug Fig. 4b
  • contextual recall Fig. 4c
  • freezing time Fig. 4d
  • the number of arrivals to the target area in the Morris water maze Fig. 4e
  • the time spent in Fig. 4f was measured.
  • mice Female 3xTg mice (JAX 004807) were used as experimental animals. Experimental animals were provided with sterile feed and water, and were reared with less than 10 animals each. The day and night cycle was set to 12 hours. Animal experiments were conducted in an animal breeding facility at Korea University under the guidance and supervision of the Animal Experimental Ethics Committee, and animal experiments were performed according to the protocol approved by Korea University IACUC.
  • AD fibroblasts purchased from Coriell (Camden, NJ), (AG07374(AD fibroblast 1), AG05770(AD fibroblast 2)) age-control fibroblasts (AG09857), and normal human tonsils extracted in this laboratory. Fibroblasts were used.
  • Mouse control IgG1, anti-CD9 ALB6 (Immunotech) was used as the primary antibody, and goat anti-mouse IgG (Jackson Immunoresearch) labeled with FITC was used as the secondary antibody.
  • Cells were prepared by adding trypsin-EDTA to fibroblasts in culture. Half of the cells were treated with FACS permeabilizing solution 2 purchased from Becton Dickinson Bioscience for 15 minutes, and the other half was left at 4°C. The cells were washed with PBS containing 1% Fetal bovine serum (FBS) and 0.05% sodium azide, transferred to a FACS tube, and reacted with the primary antibody at 4°C for 15 minutes. After washing with FACS wash buffer, secondary antibody was added and reacted at 4° C. for 15 minutes. After washing with FACS buffer, CD9 expression was analyzed with BD FACSCalibur.
  • FBS Fetal bovine serum
  • the Y-maze device consisted of three arms 32 cm (length) x 10 cm (width) with 26 cm walls (San Diego Instruments, San Diego, CA). Each mouse was placed in the center of the Y-maze and allowed to move freely through the maze for 5 minutes. It was considered effective if all four paws of the mouse entered the arm of the maze. Alternation was defined as entering 3 different arms (i.e. 1, 2, 3 or 2, 3, 1, etc.) in 3 succession. The percentage shift score was calculated using the following formula.
  • spontaneous alternation% (number of spontaneous)/(total number of arm entries -2) x 100
  • the total number of arms was used as a measure of the general activity of the animal.
  • the maze was cleaned with 70% ethanol to minimize the odor signal.
  • the device used was a white round plastic tank (122 cm in diameter) with a wall of 76 cm, filled with water held at 22u +/- 22uC, made opaque by adding non-toxic white paint, and a detachable square inside (side 10 cm long) Plexiglas platform.
  • the tank was in a test room containing a number of visible visual cues. Mice were placed on the platform for 10 seconds before the first test of the first session. Mice were trained to swim on a platform submerged 1.5 cm below the surface of the water. The platform was in a fixed position equidistant from the center and wall of the tank. Mice were trained three times a day. During each training period, mice were placed in tanks in random order at one of the four designated starting points.
  • mice The mouse was allowed to find and escape from the submerged platform. If the platform was not found within 60 seconds, it was manually guided to the platform and left there for 10 seconds. Mice were trained to reach a training criterion of 20 seconds (escape waiting time). Mice were evaluated in the probe test for 24 hours after the last training session and consisted of 60 seconds of free swimming in a platformless pool. The performance of each animal acquisition parameters provide data on (waiting to find the platform and distance tracking time), and probe-trial parameters (the number of entries for the target platform area) Any-Maze TM video tracking system (Stoelting Co .) was used.
  • Protein concentration was quantified using Bradford Assay (Bio-Rad Protein Assay 500-0006, Germany, Munchen), and total cell proteins, age-control fibroblasts, and AD-fibroblasts were isolated with 8-10% SDS polyacrylamide gel. .
  • Polyvinylidene difluoride (PVDF) membrane was blocked in Tris buffered saline (5% no fat formula, 0.1% Tween20).
  • As the primary antibody anti PHF-tau clone AT8 (MN1020, Thermo Scientific, Rockford, USA), p Tau pSer422 (T7944, Sigma), anti-tau clone tau-5 (MAB361, Millipore Billerica, Ma), CD9 were used. I did. All smears were standardized with actin (MAB1501, Chemicon-Millipore, Billerica, Ma).
  • CD9 still did not express age-control fibroblast.
  • AD fibroblast 1 which showed relatively lower expression of CD9 than AD fibroblast 2 in cell membranes, was hardly expressed even after permeabilization. It is believed that the CD9 molecules on the cell surface and inside the cell were lost in the permeabilization step. Meanwhile, it was found that CD9, a cell membrane protein, was expressed at a high level in AD fibroblast 2 after permeabilization. It was interpreted that the CD9 protein, which normally exists only in the cell membrane, is also present in the nucleoplasm or the nucleus, depending on the progression of alzheimer's disease.
  • NFTs neurofibrillary tangles
  • tau hyperphosphorylation is associated with changes in microtubule assembly leading to neurofibrillary tangles (NFTs) and neurodegeneration in Alzheimer's disease. have. In addition, it has been linked to many other neurological disorders, including Alzheimer's and Parkinson's.
  • the level (control lane) of phosphorylated tau (p-Ser422) in AD fibroblasts was significantly increased compared to age control fibroblasts (not shown).
  • the antibody was administered, it was confirmed that it was significantly reduced (anti-CD9-Ab panel), but the total amount of tau did not show any significant change (FIG. 3 ).
  • mice Four Tg mice (3xTg+Ab) treated with anti-CD9 Ab in Y-maze and the group treated with placebo (3xTg) had no significant difference in total cancer entry and exit, so there was no difference in general exercise activity. It was found (Fig. 4A). However, the number of alternating repetitions of the mice administered with the drug was higher than that of the control group and the percentage value was higher (Fig. 4B, # p ⁇ 0.05).
  • mice were tested at baseline spatial memory function using the Morris water maze. In this study, we performed a visible platform training, followed by a hidden platform test with four probe attempts per day. All mice in each group were able to reach the training criterion within 4 days and similarly displayed skilled swimming ability. However, in the probe test, 3xTg mice treated with anti-CD9Ab showed a significant increase in the number of arrivals to the target area and time spent in the target area when compared to the control group (Fig. 4E, 4F, # p ⁇ 0.05).
  • Substances that inhibit the expression or activity of CD9 have the effect of improving the memory of patients with neurodegenerative diseases and can be usefully used in the development of treatments for neurodegenerative diseases, and candidates for treatment of neurodegenerative diseases through analysis of the expression level of CD9 It has high industrial applicability because it can screen.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Analytical Chemistry (AREA)
  • Microbiology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Biotechnology (AREA)
  • Neurosurgery (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Neurology (AREA)
  • Pathology (AREA)
  • Cell Biology (AREA)
  • Epidemiology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Mycology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biophysics (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)

Abstract

The present invention relates to a composition for the prevention or treatment of neurodegenerative diseases using CD9, and a method for screening a therapeutic agent for neurodegenerative diseases and, more specifically, to a pharmaceutical composition for the prevention or treatment of neurodegenerative diseases comprising, as an active ingredient, an inhibitor against the expression or activity of CD9 protein, and a method for screening a therapeutic agent for neurodegenerative diseases. A material for inhibiting the expression or activity of CD9 exhibits the effect of improving the memory of patients with neurodegenerative diseases, and thus can be effectively used in the development of a therapeutic agent for neurodegenerative diseases, and through assay of the expression level of CD9, candidate materials for the therapeutic agent for neurodegenerative diseases can be screened.

Description

CD9을 이용한 퇴행성 신경질환의 예방 또는 치료용 조성물과 퇴행성 신경질환 치료제 스크리닝 방법Composition for the prevention or treatment of neurodegenerative diseases and screening method for the treatment of neurodegenerative diseases using CD9
본 출원은 2019년 4월 18일에 출원된 대한민국 특허출원 제10-2019-0045489호를 우선권으로 주장하고, 상기 명세서 전체는 본 출원의 참고문헌이다.This application claims priority to Korean Patent Application No. 10-2019-0045489 filed on April 18, 2019, and the entire specification is a reference of this application.
본 발명은 CD9을 이용한 퇴행성 신경질환의 예방 또는 치료용 조성물과 퇴행성 신경질환 치료제 스크리닝 방법에 관한 것으로, 보다 상세하게는 CD9 단백질의 발현 또는 활성 억제제를 유효성분으로 포함하는 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물과, (a) CD9 단백질 또는 mRNA를 발현하는 세포에 시험물질을 처리하는 단계; (b) 상기 세포와 시험물질을 처리하지 않은 대조군 세포에서 CD9 단백질 또는 mRNA의 발현 수준을 측정하는 단계; 및 (c) 대조군 세포와 비교하여 CD9 단백질 또는 mRNA의 발현 수준을 감소시키는 시험 물질을 퇴행성 신경질환 치료제 후보물질로 선별하는 단계를 포함하는 퇴행성 신경질환 치료제 스크리닝 방법에 관한 것이다. The present invention relates to a composition for preventing or treating neurodegenerative diseases using CD9 and a screening method for a therapeutic agent for neurodegenerative diseases, and more particularly, to a prevention or treatment of neurodegenerative diseases comprising an inhibitor of CD9 protein expression or activity as an active ingredient A pharmaceutical composition for use, and (a) treating a test substance on cells expressing CD9 protein or mRNA; (b) measuring the expression level of CD9 protein or mRNA in the cells and control cells not treated with the test substance; And (c) selecting a test substance that reduces the expression level of CD9 protein or mRNA compared to the control cells as a candidate substance for treating neurodegenerative diseases.
신경퇴행성 질환 (neurodegenerative diseases)은 신경세포가 퇴화하고, 기능을 잃고, 그리고 종종 사멸하는 경우의 증상들과 관련된다. 신경퇴행성 질환을 가진 환자들은 인지 (cognitive) 또는 운동 (motor) 능력에 있어서 극심한 퇴화를 겪을 수 있고, 이들 질환은 주로 진행성이기 때문에 결과적으로 그들의 삶의 질 및 삶에 대한 기대는 현저히 감소될 수 있다.Neurodegenerative diseases are associated with symptoms when nerve cells degenerate, lose function, and often die. Patients with neurodegenerative diseases may experience extreme degeneration in cognitive or motor abilities, and because these diseases are primarily progressive, their quality of life and expectations for life may be significantly reduced as a result. .
이들 질환은 파킨슨병 (Parkinson's Disease; PD), 알츠하이머병 (Alzheimer's disease; AD), 루게릭병(amyotrophic lateral sclerosis; ALS), 헌팅턴병 (Huntington's disease; HD), 전두측두엽 치매(Frontotemporal Dementia), 피질-기저핵 퇴행증 (Cortico Basal Degeneration), 진행성 핵상마비 (progressive supranuclear palsy; PSP) 및 다른 질병들을 포함한다.These diseases include Parkinson's Disease (PD), Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), Frontotemporal Dementia, and cortical-basal ganglia. Degeneration (Cortico Basal Degeneration), progressive supranuclear palsy (PSP) and other diseases.
다양한 신경퇴행성 질환 중에서도 치매 (dementia)는 가장 광범위한 세포 손상을 유발하는 질환으로 퇴행성 정신장애를 동반하며, 특히 기억력 장애, 판단력 상실 등의 주요증상이 잘 알려져 있다. 치매는 뇌혈관의 협착이나 폐색으로 생기는 혈관성 치매와 베타아밀로이드 단백질이 뇌 내에 축적되어 발병하는 것으로 알려진 알츠하이머성 치매, 그리고 이들 두 가지 원인이 복합적으로 작용하여 생기는 혼합형 치매로 크게 나눌 수 있다.Among various neurodegenerative diseases, dementia is a disease that causes the most extensive cell damage and accompanies degenerative mental disorders. In particular, major symptoms such as memory impairment and loss of judgment are well known. Dementia can be broadly divided into vascular dementia caused by stenosis or blockage of cerebrovascular blood vessels, Alzheimer's dementia, which is known to be caused by accumulation of beta-amyloid protein in the brain, and mixed dementia caused by a combination of these two causes.
치매 환자 중 가장 많은 비율을 차지하는 유형은 알츠하이머성 치매이며, 최근 연구에 따르면 2050년에는 85명중 1명의 비율로 이 질환을 가질 것이고, 이중 43 %는 특별한 관리를 받아야한다는 보고가 있다. (PrabhulkarS, Piatyszek R, et al. J Neurochem., 2012, 122, 374-381). 신경퇴행성 질환 중 알츠하이머병은 크게 유전성 알츠하이머병 (familiar Alzheimer's disease)과 산발성 알츠하이머병 (sporadic Alzheimer's disease, SAD)으로 분류된다. 유전성 알츠하이머병은 전체 알츠하이머 병 환자의 5~10 % 정도를 차지한다.Alzheimer's dementia accounts for the largest proportion of dementia patients, and a recent study reports that 1 in 85 people will have the disease by 2050, of which 43% need special care. (Prabhulkar S, Piatyszek R, et al. J Neurochem., 2012, 122, 374-381). Among neurodegenerative diseases, Alzheimer's disease is largely classified into hereditary Alzheimer's disease and sporadic Alzheimer's disease (SAD). Hereditary Alzheimer's disease accounts for 5-10% of all Alzheimer's patients.
hyperphosphorylated, aggregated tau는 알츠하이머병 및 많은 인간 신경 퇴행성 질환의 중심 신경 병리학적 특징이다. MAPT 유전자의 돌연변이 발견 이후, 최근 몇 년간 타우 (tau)의 역할에 대한 상당한 보고서가 발표되었다. 다양한 타우 트랜스 제닉 동물 모델의 개발에 따라 뉴런에서 다른 세포로의 타우 병리학의 전파 및 Aβ와 타우 사이의 상호 작용을 포함하여 타우 의존성 병리학에서 상당한 진전을 보였다. 이러한 병리학 및 타우 관련 생체 표지 물질에 따라 새로운 치료법이 얻어졌다. 그럼에도 불구하고, 신경 변성을 유발하는 병인학적 요인 및 타우 매개 신경 독성의 정확한 기전은 아직 밝혀야 할 필요가 있으며, 현재까지 발병의 정확한 원인은 알려진 것이 없다.(Ann Transl Med. 2018 May; 6(10): 175.)The hyperphosphorylated, aggregated tau is a central neuropathological feature of Alzheimer's disease and many human neurodegenerative diseases. Following the discovery of mutations in the MAPT gene, significant reports have been published on the role of tau in recent years. The development of various tau transgenic animal models has made significant progress in tau dependent pathology, including the propagation of tau pathology from neurons to other cells and interactions between Aβ and tau. According to this pathology and tau-related biomarkers, new treatments have been obtained. Nevertheless, the etiological factors leading to neurodegeneration and the exact mechanism of tau-mediated neurotoxicity still need to be elucidated, and the exact cause of the onset is not known to date (Ann Transl Med. 2018 May; 6(10). ): 175.)
알츠하이머병의 병리학적 특징으로는 신경세포의 외부에 축적되어지는 노인반점 (senile plaques), 신경세포의 세포체 내에 엉켜진 실뭉치처럼 보이는 신경섬유 덩어리 (neurofibrilary tangles) 및 신경세포의 손실(neuronal loss) 등을 들 수 있다. 이러한 병리학적 특징은 유전성 알츠하이머병 및 산발성 알츠하이머병 모두에 나타나며, 이 중 노인반점의 주요 구성 요소로는 응집된 아밀로이드 베타 펩티드 (amyloid β peptide; Aβ)라는 독성단백질이 밝혀져 있다. 아밀로이드 베타 펩티드는 아밀로이드 전구단백질의 비정상적인 절단으로부터 생성된 40 내지 42개의 아미노산으로 이루어진 불용성 펩티드이다. 또한, 아밀로이드 베타 펩티드의 과다 축적은 유전성 알츠하이머병 및 산발성 알츠하이머병 모두에 공통적 현상으로 나타난다고 보고되어 있다. 따라서 아밀로이드 베타 펩티드는 알츠하이머병의 주요 병원성 물질 (pathogenic material)로 간주되고 있다. 알츠하이머병의 전반적인 발병과정은 프레세닐린 1, 2 유전자 (PS 1, 2)의 돌연변이가 발생하면 베타-세크레타제(β-secretase)에 의해 아밀로이드 전구단백질이 비정상적으로 절단되고 아밀로이드 베타 펩티드가 생성된다. 생성된 아밀로이드 베타 펩티드에 의해 뇌신경세포의 괴사가 일어나며, 이로 인해 알츠하이머병이 발병하게 된다고 알려져 있다.Pathological features of Alzheimer's disease include senile plaques that accumulate outside of nerve cells, neurofibrilary tangles that look like tangled bundles of threads in the cell body of nerve cells, and neuroal loss. And the like. These pathological features appear in both hereditary Alzheimer's disease and sporadic Alzheimer's disease. Among them, a toxic protein called amyloid β peptide (Aβ) has been revealed as a major constituent of senile spots. Amyloid beta peptides are insoluble peptides consisting of 40 to 42 amino acids resulting from abnormal cleavage of amyloid proproteins. In addition, it has been reported that excessive accumulation of amyloid beta peptide appears as a phenomenon common to both hereditary Alzheimer's disease and sporadic Alzheimer's disease. Therefore, amyloid beta peptide is regarded as a major pathogenic material for Alzheimer's disease. The overall pathogenesis of Alzheimer's disease is that when mutations in the presenilin 1 and 2 genes (PS 1 and 2) occur, the amyloid proprotein is abnormally cleaved by β-secretase and amyloid beta peptide is produced. do. It is known that necrosis of cranial nerve cells occurs due to the produced amyloid beta peptide, which causes Alzheimer's disease.
현재까지 치매의 원인과 병인론 그리고 그 치료법에 대한 광범위하고 다양한 연구가 진행되었지만, 원인규명이 미비하고 효과적인 치료법의 개발 또한 미진하다. 비록 FDA에서 알츠하이머 치매의 치료제로 타크린 (tacrine), 리바스티그민 (rivastigmine), 갈란타민 (galantamine), 도네페질 (donepezil), 메만틴 (memantine)을 인가하였음에도, 현재 사용되는 치매 치료제 대부분은 퇴행성 치매의 증상을 완화시키는 정신퇴행 완화 물질에 불과하며, 이들 중 대부분은 소염작용을 주로 하는 약물로 간독성과 소화기관의 점막을 손상시키는 등 부작용이 상당하며, 궁극적인 원인치료라기 보다는 대증적인 요법에 국한되어 있다는 한계가 있다.Until now, extensive and diverse studies on the cause, etiology and treatment of dementia have been conducted, but the cause is insufficient and the development of effective treatments is also insufficient. Although the FDA approved tacrine, rivastigmine, galantamine, donepezil, and memantine as treatments for Alzheimer's dementia, most dementia treatments currently used are degenerative. It is only a psychodegenerative substance that relieves the symptoms of dementia. Most of these drugs are mainly anti-inflammatory drugs, and have considerable side effects such as hepatotoxicity and damage to the mucous membrane of the digestive tract, and are suitable for symptomatic therapy rather than ultimate cause treatment. There is a limit to being limited.
한편, CD9은 분자량이 24 내지 27 kDa의 테트라스파닌(tetraspanin)계 세포막 당단백질로 포유 동물에는 34개의 테트라스파닌이 있으며, 그 중 33개가 인간에서 확인되었다. 각각의 테트라스파닌은 세포 부착 및 이동, 혈소판 활성 및 응집, 포유동물의 수정 과정에서 난자와 정자의 융합, 암의 발생 및 전이, 체액성 면역 반응 및 알러지 반응, 인간 면역 결핍 바이러스-1(HIV-1)과 인플루엔자 바이러스 복제 등에 중요한 역할을 하는 항원으로 알려져 있다. 그러나, 인간에서 CD9 항원의 세포 노화 조절이나, 노화 조절 연구는 단순히 노화된 사람 혈관 내피세포에서 발현이 증가되는 것이 보고되었을 뿐, CD9의 발현 및 활성이 퇴행성 신경질환에 미치는 영향은 아직까지 보고된 바 없다.Meanwhile, CD9 is a tetraspanin-based cell membrane glycoprotein having a molecular weight of 24 to 27 kDa, and there are 34 tetraspanins in mammals, and 33 of them have been identified in humans. Each tetraspanin contains cell adhesion and migration, platelet activity and aggregation, fusion of egg and sperm during fertilization in mammals, development and metastasis of cancer, humoral immune response and allergic response, human immunodeficiency virus-1 (HIV It is known as an antigen that plays an important role in -1) and influenza virus replication. However, studies on the regulation of cellular senescence or aging control of the CD9 antigen in humans have only reported that the expression is increased in aging human vascular endothelial cells, and the effect of the expression and activity of CD9 on neurodegenerative diseases has been reported. There is no bar.
이에, 본 발명자는 새로운 퇴행성 신경질환 치료법을 개발하기 위하여 예의 연구를 거듭한 결과, CD9 단백질의 발현 또는 활성을 저해하면 퇴행성 뇌 병변으로 유발되는 기억력 감소 등의 증상이 개선될 수 있음을 발견하고 본 발명을 완성하게 되었다. Accordingly, the present inventors have repeatedly conducted extensive research to develop a new treatment for neurodegenerative diseases, and found that inhibition of the expression or activity of the CD9 protein can improve symptoms such as memory loss caused by degenerative brain lesions. The invention was completed.
따라서, 본 발명의 목적은 CD9 단백질의 발현 또는 활성 억제제를 유효성분으로 포함하는 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물을 제공하는 것이다. Accordingly, it is an object of the present invention to provide a pharmaceutical composition for preventing or treating neurodegenerative diseases comprising an inhibitor of expression or activity of CD9 protein as an active ingredient.
또한 CD9 단백질의 발현 또는 활성 억제제를 유효성분으로 구성되는 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물을 제공하는 것이다.In addition, it is to provide a pharmaceutical composition for the prevention or treatment of neurodegenerative diseases consisting of an inhibitor of the expression or activity of the CD9 protein as an active ingredient.
또한 CD9 단백질의 발현 또는 활성 억제제를 유효성분으로 필수적으로 구성되는 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물을 제공하는 것이다.In addition, it is to provide a pharmaceutical composition for the prevention or treatment of neurodegenerative diseases consisting essentially of an inhibitor of expression or activity of a CD9 protein as an active ingredient.
본 발명의 다른 목적은 (a) CD9 단백질 또는 mRNA를 발현하는 세포에 시험물질을 처리하는 단계; (b) 상기 세포와 시험물질을 처리하지 않은 대조군 세포에서 CD9 단백질 또는 mRNA의 발현 수준을 측정하는 단계; 및 (c) 대조군 세포와 비교하여 CD9 단백질 또는 mRNA의 발현 수준을 감소시키는 시험 물질을 퇴행성 신경질환 치료제 후보물질로 선별하는 단계를 포함하는 퇴행성 신경질환 치료제 스크리닝 방법을 제공하는 것이다. Another object of the present invention is the steps of (a) treating a test substance on cells expressing CD9 protein or mRNA; (b) measuring the expression level of CD9 protein or mRNA in the cells and control cells not treated with the test substance; And (c) selecting a test substance that reduces the expression level of CD9 protein or mRNA compared to the control cells as a candidate substance for treating a neurodegenerative disease.
본 발명의 또 다른 목적은 퇴행성 신경질환의 예방 또는 치료용 제제를 제조하기 위한 CD9 단백질의 발현 또는 활성 억제제의 용도를 제공하는 것이다.Another object of the present invention is to provide a use of an inhibitor of expression or activity of a CD9 protein for preparing a preparation for preventing or treating neurodegenerative diseases.
본 발명의 또 다른 목적은 CD9 단백질의 발현 또는 활성 억제제를 유효성분으로 포함하는 조성물의 유효량을 이를 필요로 하는 개체에 투여하는 단계를 포함하는 퇴행성 신경질환의 치료 방법을 제공하는 것이다.Another object of the present invention is to provide a method for treating neurodegenerative diseases comprising administering to an individual in need thereof an effective amount of a composition comprising an inhibitor of CD9 protein expression or activity as an active ingredient.
상기한 목적을 달성하기 위하여 본 발명은 CD9 단백질의 발현 또는 활성 억제제를 유효성분으로 포함하는 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물을 제공한다. In order to achieve the above object, the present invention provides a pharmaceutical composition for preventing or treating neurodegenerative diseases comprising an inhibitor of expression or activity of CD9 protein as an active ingredient.
또한 본 발명은 CD9 단백질의 발현 또는 활성 억제제를 유효성분으로 구성되는 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물을 제공한다.In addition, the present invention provides a pharmaceutical composition for preventing or treating neurodegenerative diseases comprising an inhibitor of CD9 protein expression or activity as an active ingredient.
또한 본 발명은 CD9 단백질의 발현 또는 활성 억제제를 유효성분으로 필수적으로 구성되는 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물을 제공한다.In addition, the present invention provides a pharmaceutical composition for preventing or treating neurodegenerative diseases essentially consisting of an inhibitor of CD9 protein expression or activity as an active ingredient.
본 발명의 다른 목적을 달성하기 위하여 본 발명은 (a) CD9 단백질 또는 mRNA를 발현하는 세포에 시험물질을 처리하는 단계; (b) 상기 세포와 시험물질을 처리하지 않은 대조군 세포에서 CD9 단백질 또는 mRNA의 발현 수준을 측정하는 단계; 및 (c) 대조군 세포와 비교하여 CD9 단백질 또는 mRNA의 발현 수준을 감소시키는 시험 물질을 퇴행성 신경질환 치료제 후보물질로 선별하는 단계를 포함하는 퇴행성 신경질환 치료제 스크리닝 방법을 제공한다. In order to achieve another object of the present invention, the present invention comprises the steps of: (a) treating cells expressing CD9 protein or mRNA with a test substance; (b) measuring the expression level of CD9 protein or mRNA in the cells and control cells not treated with the test substance; And (c) selecting a test substance that reduces the expression level of CD9 protein or mRNA compared to the control cells as a candidate substance for treating a neurodegenerative disease.
본 발명의 또 다른 목적을 달성하기 위하여, 본 발명은 퇴행성 신경질환의 예방 또는 치료용 제제를 제조하기 위한 CD9 단백질의 발현 또는 활성 억제제의 용도를 제공한다.In order to achieve another object of the present invention, the present invention provides a use of an inhibitor of expression or activity of a CD9 protein to prepare a preparation for preventing or treating neurodegenerative diseases.
본 발명의 또 다른 목적을 달성하기 위하여, 본 발명은 CD9 단백질의 발현 또는 활성 억제제를 유효성분으로 포함하는 조성물의 유효량을 이를 필요로 하는 개체에 투여하는 단계를 포함하는 퇴행성 신경질환의 치료 방법을 제공한다.In order to achieve another object of the present invention, the present invention provides a method for treating neurodegenerative diseases comprising administering to an individual in need thereof an effective amount of a composition comprising an inhibitor of CD9 protein expression or activity as an active ingredient. to provide.
이하 본 발명을 상세히 설명한다. Hereinafter, the present invention will be described in detail.
본 발명은 CD9 단백질의 발현 또는 활성 억제제를 유효성분으로 포함하는 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물을 제공한다. The present invention provides a pharmaceutical composition for preventing or treating neurodegenerative diseases comprising an inhibitor of expression or activity of CD9 protein as an active ingredient.
또한 본 발명은 CD9 단백질의 발현 또는 활성 억제제를 유효성분으로 구성되는 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물을 제공한다.In addition, the present invention provides a pharmaceutical composition for preventing or treating neurodegenerative diseases comprising an inhibitor of CD9 protein expression or activity as an active ingredient.
또한 본 발명은 CD9 단백질의 발현 또는 활성 억제제를 유효성분으로 필수적으로 구성되는 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물을 제공한다.In addition, the present invention provides a pharmaceutical composition for preventing or treating neurodegenerative diseases essentially consisting of an inhibitor of CD9 protein expression or activity as an active ingredient.
본 명세서에서 '치료'는 질환의 발생 또는 재발 억제, 증상의 완화, 질병의 직접 또는 간접적인 병리학적 결과의 감소, 질병 진행 속도의 감소, 질병 상태의 개선, 호전, 완화 또는 개선된 예후를 의미한다. 본 발명에서 사용되는 용어 '예방'은 질환의 발병을 억제시키거나 진행을 지연시키는 모든 행위를 의미한다.In the present specification,'treatment' refers to suppression of occurrence or recurrence of a disease, alleviation of symptoms, reduction of direct or indirect pathological consequences of a disease, reduction of disease progression rate, improvement of disease state, improvement, alleviation or improved prognosis. do. The term'prevention' as used in the present invention means any action that suppresses the onset or delays the progression of a disease.
'단백질'은 '폴리펩타이드(polypeptide)' 또는 '펩타이드(peptide)'와 호환성 있게 사용되며, 예컨대, 자연 상태의 단백질에서 일반적으로 발견되는 바와 같이 아미노산 잔기의 중합체를 말한다. 예를 들어, CD9 단백질의 '단편(fragment)'은 CD9 단백질의 일부분의 펩타이드를 말한다. 'Protein' is used interchangeably with'polypeptide' or'peptide', for example, refers to a polymer of amino acid residues as commonly found in proteins in nature. For example, a'fragment' of the CD9 protein refers to a peptide of a portion of the CD9 protein.
본 발명에서 '폴리뉴클레오티드(polynucleotide)' 또는 '핵산'은 단일 또는 이중 가닥의 형태로 된 데옥시리보핵산(deoxyribonucleic acid, DNA) 또는 리보핵산(ribonucleic acid, RNA)를 말한다. 다른 제한이 없는 한, 자연적으로 생성되는 뉴클레오티드와 비슷한 방법으로 핵산에 혼성화되는 자연적 뉴클레오티드의 공지된 아날로그도 포함된다. 일반적으로 DNA는 아데닌(adenine, A), 구아닌(guanine, G), 시토신(cytosine, C), 티민(thymine, T) 등 네 가지 염기로 구성되어 있으며, RNA는 티민 대신 우라실(Uracil, U)을 가지고 있다. 핵산 이중 가닥에서 A는 T 또는 U, C는 G 염기와 수소결합을 이루는데, 이러한 염기의 관계를 '상보적(complementary)'이라고 한다.In the present invention,'polynucleotide' or'nucleic acid' refers to deoxyribonucleic acid (DNA) or ribonucleic acid (RNA) in the form of single or double strands. Unless otherwise limited, also include known analogs of natural nucleotides that hybridize to nucleic acids in a manner similar to those of naturally occurring nucleotides. In general, DNA is composed of four bases: adenine (A), guanine (G), cytosine (C), and thymine (T), and RNA is composed of uracil (U) instead of thymine. Have. In the double-stranded nucleic acid, A forms a hydrogen bond with T or U, and C forms a hydrogen bond with the G base, and the relationship between these bases is called'complementary'.
한편, 'mRNA(messenger RNA 또는 전령 RNA)'는 단백질 합성 과정에서 특정 유전자의 염기서열의 유전 정보를 리보솜(ribosome)으로 전달하여 폴리펩티드 합성(단백질 번역, translation)의 청사진 역할을 하는 RNA이다. 유전자를 주형(template)으로 하여 단일 가닥의 mRNA가 전사(transcription) 과정을 통하여 합성된다.On the other hand,'mRNA (messenger RNA or messenger RNA)' is an RNA that serves as a blueprint for polypeptide synthesis (protein translation, translation) by transferring the genetic information of the nucleotide sequence of a specific gene to the ribosome during protein synthesis. Using the gene as a template, single-stranded mRNA is synthesized through a transcription process.
본 발명에서 상기 퇴행성 신경질환은 신경세포가 퇴화하고, 기능을 잃고, 그리고 사멸하는 경우 나타나는 증상들과 관련된 질환을 의미하며, 알츠하이머병, 치매, 경도인지장애, 파킨슨병, 진행성 핵상마비, 다계통 위축증, 감람핵-뇌교-소뇌 위축증(OPCA), 샤이-드래거 증후군, 선조체-흑질 퇴행증, 헌팅톤병, 근위축성 측색 경화증(ALS), 본태성 진전증, 피질-기저핵 퇴행증, 미만성 루이 소체 질환, 파킨스-ALS-치매 복합증, 니만픽 병(Niemann-Pick disease), 픽병, 뇌허혈 및 뇌경색으로 이루어진 군에서 선택될 수 있으나, 이에 제한되는 것은 아니며, 가장 바람직하게는 알츠하이머병 또는 치매일 수 있다. In the present invention, the neurodegenerative disease refers to a disease related to symptoms that appear when nerve cells degenerate, lose function, and die, and Alzheimer's disease, dementia, mild cognitive impairment, Parkinson's disease, progressive nuclear paralysis, multi-system Atrophy, Olive Nucleus-Peripheral-Cerebral Atrophy (OPCA), Shy-Drager Syndrome, Stroke-Black Matter Degeneration, Huntington's Disease, Amyotrophic Lateral Sclerosis (ALS), Essential Tremors, Cortical-basal Nucleus Degeneration, Diffuse Lewy Body Disease , Parkin's-ALS-dementia complex, Niemann-Pick disease, Pick's disease, cerebral ischemia, and may be selected from the group consisting of cerebral infarction, but is not limited thereto, and most preferably Alzheimer's disease or dementia. have.
한편, CD9은 분자량이 24 내지 27 kDa의 테트라스파닌(tetraspanin)계 세포막 당단백질로 포유 동물에는 34개의 테트라스파닌이 있으며, 그 중 33개가 인간에서 확인되었다. 각각의 테트라스파닌은 세포 부착 및 이동, 혈소판 활성 및 응집, 포유동물의 수정 과정에서 난자와 정자의 융합, 암의 발생 및 전이, 체액성 면역 반응 및 알러지 반응, 인간 면역 결핍 바이러스-1(HIV-1)과 인플루엔자 바이러스 복제 등에 중요한 역할을 하는 항원으로 알려져 있다.Meanwhile, CD9 is a tetraspanin-based cell membrane glycoprotein having a molecular weight of 24 to 27 kDa, and there are 34 tetraspanins in mammals, and 33 of them have been identified in humans. Each tetraspanin contains cell adhesion and migration, platelet activity and aggregation, fusion of egg and sperm during fertilization in mammals, development and metastasis of cancer, humoral immune response and allergic response, human immunodeficiency virus-1 (HIV It is known as an antigen that plays an important role in -1) and influenza virus replication.
본 발명에서 사용된 용어 'CD9'는 테트라스파닌계(tetraspanin family)의 멤버에 속하는 약 24~27 kDa의 세포표면 당단백질 수용체로서, 세포 발달, 활성, 성장 및 운동성을 조절하는데 중요한 역할을 하는 신호전달 작용(signal transduction events)을 조절한다고 알려져 있다. 또한, 세포 부착 및 세포 이동을 조절할 수 있고, 혈소판-유발 내피세포 증식(platelet-induced endothelial cell proliferation)에 관여하는 혈소판활성화(platelet activation)와 집성(aggregation)을 유도한다. 아울러, 근육세포 융합(muscle cell fusion)을 촉진하고 근관 유지(myotube maintenance)에 기여하는 등 세포 내의 다양한 현상에 관여한다.The term'CD9' as used herein is a cell surface glycoprotein receptor of about 24 to 27 kDa belonging to a member of the tetraspanin family, and a signal that plays an important role in regulating cell development, activity, growth and motility. It is known to regulate signal transduction events. In addition, it can regulate cell adhesion and cell migration, and induce platelet activation and aggregation, which are involved in platelet-induced endothelial cell proliferation. In addition, it is involved in various phenomena within cells, such as promoting muscle cell fusion and contributing to myotube maintenance.
본 발명에서의 CD9 단백질은 포유류에서 유래한 것일 수 있으며, 바람직하게는 인간에서 유래한 것이다. 가장 바람직하게, 본 발명에서의 CD9 단백질은 서열번호 1로 표시되는 인간의 CD9 isoform 1(NP_001760.1) 또는 서열번호 2로 표시되는 인간의 CD9 isoform 2(NP_001317241.1)의 아미노산 서열을 포함하는 것을 특징으로 한다(괄호 안은 NCBI Genbank accession number). The CD9 protein in the present invention may be derived from mammals, preferably derived from humans. Most preferably, the CD9 protein in the present invention comprises the amino acid sequence of human CD9 isoform 1 (NP_001760.1) represented by SEQ ID NO: 1 or human CD9 isoform 2 (NP_001317241.1) represented by SEQ ID NO: 2 Characterized in that (NCBI Genbank accession number in parentheses).
상기 CD9 단백질의 발현 억제제는 CD9 mRNA에 상보적으로 결합하는 안티센스 올리고뉴클레오티드(antisense oligonucleotide), siRNA, shRNA, miRNA, ribozyme, crispr-cas9, DNAzyme 및 PNA(protein nucleic acid)로 이루어진 군에서 선택된 어느 하나인 것일 수 있다. 또한 상기 CD9 mRNA는 가장 바람직하게는 서열번호 3으로 표시되는 인간 CD9 mRNA transcript variant 1(NM_001769.3) 또는 서열번호 4로 표시되는 인간 CD9 mRNA transcript variant 2(NM_001330312.1)의 염기서열을 포함하는 것이다(괄호 안은 NCBI Genbank accession number). 상기한 인간의 CD9 mRNA transcript variant들의 coding region(exon) 등의 특징 사항은 괄호 안에 기재된 Genbank accession number로 NCBI 데이터베이스를 검색하여 나오는 서열정보에서 확인된다.The CD9 protein expression inhibitor is any one selected from the group consisting of an antisense oligonucleotide complementary to CD9 mRNA, siRNA, shRNA, miRNA, ribozyme, crispr-cas9, DNAzyme, and protein nucleic acid (PNA). It can be. In addition, the CD9 mRNA most preferably comprises a nucleotide sequence of human CD9 mRNA transcript variant 1 (NM_001769.3) represented by SEQ ID NO: 3 or human CD9 mRNA transcript variant 2 (NM_001330312.1) represented by SEQ ID NO: 4 (NCBI Genbank accession number in parentheses). Features such as the coding region (exon) of the human CD9 mRNA transcript variants are identified in the sequence information displayed by searching the NCBI database with the Genbank accession number in parentheses.
나아가, 본 발명에 따른 CD9 단백질 발현억제제는 바람직하게는 CD9 mRNA에 상보적으로 결합하여 mRNA의 분해를 유발하는 shRNA일 수 있다. Furthermore, the CD9 protein expression inhibitor according to the present invention may preferably be a shRNA that binds complementarily to CD9 mRNA and causes degradation of the mRNA.
본 발명에서 상기 'siRNA(small interfering RNA 또는 short interfering RNA 또는 silencing RNA)'는 세포 안에 인위적으로 도입되어 특정 유전자의 mRNA의 분해를 유도하여 단백질 번역(translation)이 나지 않도록 하여 유전자 발현을 억제하는 RNA 간섭(RNA interference) 현상을 일으키는 짧은 이중가닥의 RNA로, 표적 mRNA의 특정 부위에 상보적인 20 내지 25개의 뉴클레오티드로 구성되어 있다. 세포 내에서 siRNA의 이중 가닥 중 표적 mRNA에 상보적인 가닥(antisense strand)이 RISC(RNA-induced silencing complex) 단백질 복합체와 결합하여 표적 mRNA에 결합하고, RISC 복합체 안의 argonaute 단백질이 표적 mRNA를 절단하여 분해시키거나, 단백질 번역에 중요한 단백질과 리보솜이 mRNA와 결합하는 것을 억제하는 등의 기작으로 특정 유전자의 발현을 억제한다. In the present invention, the'siRNA (small interfering RNA or short interfering RNA or silencing RNA)' is artificially introduced into the cell to induce degradation of the mRNA of a specific gene, thereby preventing protein translation, thereby inhibiting gene expression. It is a short double-stranded RNA that causes RNA interference, and is composed of 20 to 25 nucleotides complementary to a specific site of the target mRNA. Among the double strands of siRNA in cells, an antisense strand complementary to the target mRNA binds to the RNA-induced silencing complex (RISC) protein complex and binds to the target mRNA, and the argonaute protein in the RISC complex cleaves and degrades the target mRNA. It suppresses the expression of specific genes through mechanisms such as suppressing the binding of proteins and ribosomes important for protein translation with mRNA.
본 발명에서 상기 shRNA는 RNA 간섭을 유도하는 물질로서, 1본쇄 RNA에서 부분적으로 회문상의 염기서열을 포함함으로써, 분자 내에서 2본쇄 구조를 가지고 헤어핀과 같은 구조가 되는 약 20염기 이상의 분자이다. 또한, shRNA 발현용 플라스미드(plasmid)를 세포 내로 도입시켜 발현하게 되면, 세포 내의 다이서(dicer)라는 RNaseⅢ(ribonucleaseⅢ) 효소에 의해 21~23개 염기쌍의 siRNA가 생성되어 RNAi를 유도한다.In the present invention, the shRNA is a substance that induces RNA interference, and is a molecule having a double-stranded structure in the molecule and a hairpin-like structure by partially containing a palindromic nucleotide sequence in a single-stranded RNA. In addition, when a shRNA expression plasmid is introduced into a cell and expressed, a 21-23 base pair siRNA is generated by RNase III (ribonuclease III) enzyme called dicer in the cell to induce RNAi.
본 발명에서 상기 siRNA 또는 shRNA는 올리고뉴클레오티드의 생체 내 안정성 향상, 핵산 분해효소 저항성 부여 및 비특이적 면역반응 감소를 위한 다양한 변형(modification)을 가한 것일 수 있다. 상기 올리고뉴클레오티드의 변형은 하나 이상의 뉴클레오티드 내 당 구조의 2´ 탄소 위치에서 OH기가 -CH 3(메틸), -OCH 3(methoxy), -NH 2, -F, -O-2-메톡시에틸, -O-프로필(propyl), -O-2-메틸티오에틸(methylthioethyl), -O-3-아미노프로필, -O-3-디메틸아미노프로필, -O-N-메틸아세트아미도 또는 -O-디메틸아미도옥시에틸로의 치환에 의한 변형; 뉴클레오티드 내 당(sugar) 구조 내의 산소가 황으로 치환된 변형; 또는 뉴클레오티드결합의 포스포로티오에이트(phosphorothioate) 또는 보라노포스페이트(boranophosphate), 메틸포스포네이트(methyl phosphonate) 결합으로의 변형에서 선택된 하나 이상의 변형이 조합되어 사용될 수 있으며, PNA(peptide nucleic acid), LNA(locked nucleic acid) 또는 UNA(unlocked nucleic acid) 형태로의 변형도 사용이 가능하다.In the present invention, the siRNA or shRNA may be obtained by applying various modifications to improve the stability of the oligonucleotide in vivo, impart resistance to nuclease enzymes, and reduce non-specific immune responses. The modification of the oligonucleotide is the OH group at the 2′ carbon position of the sugar structure in one or more nucleotides -CH 3 (methyl), -OCH 3 (methoxy), -NH 2 , -F, -O-2-methoxyethyl, -O-propyl, -O-2-methylthioethyl, -O-3-aminopropyl, -O-3-dimethylaminopropyl, -ON-methylacetamido or -O-dimethylami Modification by substitution with dooxyethyl; Modification in which oxygen in the sugar structure in the nucleotide is substituted with sulfur; Alternatively, one or more modifications selected from modifications of nucleotide bonds to phosphorothioate, boranophosphate, and methyl phosphonate bonds may be used in combination, and PNA (peptide nucleic acid), Modification into the form of LNA (locked nucleic acid) or UNA (unlocked nucleic acid) can also be used.
또한 상기 CD9 단백질 활성 억제제는 CD9 단백질에 특이적으로 결합하는 화합물, 펩티드, 펩티드 유사체(mimetics), 앱타머, 항체, 천연추출물 및 합성화합물로 이루어진 군에서 선택된 어느 하나일 수도 있다. In addition, the CD9 protein activity inhibitor may be any one selected from the group consisting of compounds, peptides, peptide mimetics, aptamers, antibodies, natural extracts, and synthetic compounds that specifically bind to CD9 protein.
본 발명에 따른 약학적 조성물은 퇴행성 신경질환 치료를 위해 약학적으로 허용되는 담체와 함께 당업계에 공지된 방법으로 투여 경로에 따라 다양하게 제형화될 수 있다. 상기 담체로는 모든 종류의 용매, 분산매질, 수중유 또는 유중수 에멀젼, 수성 조성물, 리포좀, 마이크로비드 및 마이크로좀이 포함된다. 바람직하게는 transfection reagent를 이용하여 제형화하고, 피하, 혈액, 골수, 복강 등을 이용 세포에 전달할 수 있다.The pharmaceutical composition according to the present invention can be variously formulated according to the route of administration by a method known in the art together with a pharmaceutically acceptable carrier for the treatment of neurodegenerative diseases. The carrier includes all kinds of solvents, dispersion media, oil-in-water or water-in-oil emulsions, aqueous compositions, liposomes, microbeads and microsomes. Preferably, it is formulated using a transfection reagent, and can be delivered to cells using subcutaneous, blood, bone marrow, or abdominal cavity.
상기 본 발명에 따른 약학적 조성물은 약학적으로 유효한 양, 즉 퇴행성 신경질환을 예방하거나 증상을 완화하고 치료하기 충분한 양으로 환자에게 투여될 수 있다. 예를 들어 일반적인 1일 투여량으로는 약 0.01 내지 1000㎎/㎏의 범위로 투여될 수 있으며, 바람직하게는, 약 1 내지 100mg/kg의 범위로 투여될 수 있다. 본 발명의 약학적 조성물은 바람직한 투여량 범위 내에서 1회 또는 수회로 분할 투여할 수 있다. 또한 본 발명에 따른 약학적 조성물의 투여량은 투여 경로, 투여 대상, 연령, 성별 체중, 개인차 및 질병 상태에 따라 통상의 기술자가 적절하게 선택할 수 있다. The pharmaceutical composition according to the present invention may be administered to a patient in a pharmaceutically effective amount, that is, in an amount sufficient to prevent or relieve symptoms and treat neurodegenerative diseases. For example, a typical daily dosage may be administered in the range of about 0.01 to 1000 mg/kg, and preferably, it may be administered in the range of about 1 to 100 mg/kg. The pharmaceutical composition of the present invention can be administered in one or several divided doses within a preferred dosage range. In addition, the dosage of the pharmaceutical composition according to the present invention may be appropriately selected by a person skilled in the art according to the route of administration, the subject of administration, age, sex, weight, individual differences, and disease conditions.
투여 경로로는 경구적 또는 비경구적으로 투여될 수 있다. 비경구적인 투여방법으로는 이에 한정되지는 않으나 정맥 내, 근육 내, 동맥 내, 골수 내, 경막 내, 심장 내, 경피, 피하, 복강 내, 비강 내, 장관, 국소, 설하, 직장, 또는 췌장 내 투여일 수 있으나, 이에 제한되지 않는다. The route of administration may be administered orally or parenterally. Parenteral administration methods include, but are not limited to, intravenous, intramuscular, intraarterial, intramedullary, intrathecal, intracardiac, transdermal, subcutaneous, intraperitoneal, intranasal, intestinal, topical, sublingual, rectal, or pancreatic It may be intravenous administration, but is not limited thereto.
본 발명의 약학적 조성물을 경구 투여하는 경우, 적합한 경구 투여용 담체와 함께 당업계에 공지된 방법에 따라 분말, 과립, 정제, 환제, 당의정제, 캡슐제, 액제, 겔제, 시럽제, 현탁액, 웨이퍼 등의 형태로 제형화할 수 있다. 적합한 담체의 예로는 락토즈, 덱스트로즈, 수크로즈, 솔비톨, 만니톨, 자일리톨, 에리스리톨 및 말티톨 등을 포함하는 당류와 옥수수 전분, 밀 전분, 쌀 전분 및 감자 전분 등을 포함하는 전분류, 셀룰로즈, 메틸 셀룰로즈, 나트륨 카르복시메틸셀룰로오즈 및 하이드록시프로필메틸셀룰로즈 등을 포함하는 셀룰로즈류, 젤라틴, 폴리비닐피롤리돈 등과 같은 충전제가 포함될 수 있다. 또한, 경우에 따라 가교결합 폴리비닐피롤리돈, 한천, 알긴산 또는 나트륨 알기네이트 등을 붕해제로 첨가할 수 있다. 나아가, 상기 약학적 조성물은 항응집제, 윤활제, 습윤제, 향료, 유화제 및 방부제 등을 추가로 포함할 수 있다.In the case of oral administration of the pharmaceutical composition of the present invention, powders, granules, tablets, pills, dragees, capsules, solutions, gels, syrups, suspensions, wafers according to a method known in the art together with a suitable carrier for oral administration. It can be formulated in the form of such as. Examples of suitable carriers include sugars including lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol and maltitol, and starches including corn starch, wheat starch, rice starch and potato starch, cellulose, Fillers such as celluloses including methyl cellulose, sodium carboxymethylcellulose and hydroxypropylmethylcellulose, gelatin, and polyvinylpyrrolidone may be included. In addition, in some cases, cross-linked polyvinylpyrrolidone, agar, alginic acid or sodium alginate may be added as a disintegrant. Furthermore, the pharmaceutical composition may further include an anti-aggregating agent, a lubricant, a wetting agent, a flavoring agent, an emulsifying agent and a preservative.
또한, 비경구적으로 투여하는 경우, 본 발명의 약학적 조성물은 적합한 비경구용 담체와 함께 주사제, 경피투여제 및 비강 흡입제의 형태로 당 업계에 공지된 방법에 따라 제형화될 수 있다. In addition, when administered parenterally, the pharmaceutical composition of the present invention can be formulated according to a method known in the art in the form of injections, transdermal administrations and nasal inhalants together with a suitable parenteral carrier.
또한, 약학적 조성물은 활성 물질이 표적 세포로 이동할 수 있는 임의의 장치에 의해 투여될 수 있다. 바람직한 투여방식 및 제제는 정맥 주사제, 피하 주사제, 피내 주사제, 근육 주사제 또는 점적 주사제 등이다. 주사제는 생리식염액 또는 링겔액 등의 수성 용제, 식물유, 고급 지방산 에스테르 (예로, 올레인산에칠 등), 알코올류(예로, 에탄올, 벤질알코올, 프로필렌글리콜 또는 글리세린 등) 등의 비수성 용제 등을 이용하여 제조할 수 있고, 변질 방지를 위한 안정화제 (예로, 아스코르빈산, 아황산수소나트륨, 피로아황산나트륨, BHA, 토코페롤, EDTA 등), 유화제, pH 조절을 위한 완충제, 미생물 발육을 저지하기 위한 보존제 (예로, 질산페닐수은, 치메로살, 염화벤잘코늄, 페놀, 크레솔, 벤질알코올 등) 등의 약제학적 담체를 포함할 수 있다. In addition, the pharmaceutical composition can be administered by any device capable of moving the active substance to the target cell. Preferred modes of administration and formulations are intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections or instillations. Injectables include aqueous solvents such as physiological saline or ring gel solution, vegetable oils, higher fatty acid esters (e.g., oleic acid ethyl, etc.), alcohols (e.g., ethanol, benzyl alcohol, propylene glycol, or glycerin), and other non-aqueous solvents. Stabilizers for preventing deterioration (e.g., ascorbic acid, sodium hydrogen sulfite, sodium pyrosulfite, BHA, tocopherol, EDTA, etc.), emulsifiers, buffers for pH control, and for inhibiting the growth of microorganisms Pharmaceutical carriers such as preservatives (eg, mercuric nitrate, thimerosal, benzalkonium chloride, phenol, cresol, benzyl alcohol, etc.) may be included.
상기 주사제의 경우에는 반드시 멸균되어야 하며 박테리아 및 진균과 같은 미생물의 오염으로부터 보호되어야 한다. 주사제의 경우 적합한 담체의 예로는 이에 한정되지는 않으나, 물, 에탄올, 폴리올(예를 들어, 글리세롤, 프로필렌 글리콜 및 액체 폴리에틸렌 글리콜 등), 이들의 혼합물 및/또는 식물유를 포함하는 용매 또는 분산매질일 수 있다. 보다 바람직하게는, 적합한 담체로는 행크스 용액, 링거 용액, 트리에탄올 아민이 함유된 PBS(phosphate buffered saline) 또는 주사용 멸균수, 10% 에탄올, 40% 프로필렌 글리콜 및 5% 덱스트로즈와 같은 등장 용액 등을 사용할 수 있다. 상기 주사제를 미생물 오염으로부터 보호하기 위해서는 파라벤, 클로로부탄올, 페놀, 소르빈산, 티메로살 등과 같은 다양한 항균제 및 항진균제를 추가로 포함할 수 있다. 또한, 상기 주사제는 대부분의 경우 당 또는 나트륨 클로라이드와 같은 등장화제를 추가로 포함할 수 있다. In the case of such injections, they must be sterilized and protected from contamination by microorganisms such as bacteria and fungi. Examples of suitable carriers for injections include, but are not limited to, water, ethanol, polyol (eg, glycerol, propylene glycol and liquid polyethylene glycol, etc.), a mixture thereof and/or a solvent or dispersion medium containing vegetable oil. I can. More preferably, suitable carriers include isotonic solutions such as Hanks' solution, Ringer's solution, phosphate buffered saline (PBS) containing triethanolamine or sterile water for injection, 10% ethanol, 40% propylene glycol and 5% dextrose. Etc. can be used. In order to protect the injection from microbial contamination, various antibacterial and antifungal agents such as paraben, chlorobutanol, phenol, sorbic acid, thimerosal, and the like may be additionally included. In addition, the injection may further include an isotonic agent such as sugar or sodium chloride in most cases.
경피투여제의 경우, 연고제, 크림제, 로션제, 겔제, 외용액제, 파스타제, 리니멘트제, 에어롤제 등의 형태가 포함된다. 상기에서 '경피투여'는 약학적 조성물을 국소적으로 피부에 투여하여 약학적 조성물에 함유된 유효한 양의 활성성분이 피부 내로 전달되는 것을 의미한다. 예컨대, 본 발명의 약학적 조성물을 주사형 제형으로 제조하여 이를 30 게이지의 가는 주사 바늘로 피부를 가볍게 단자(prick)하거나 피부에 직접적으로 도포하는 방법으로 투여될 수 있다. 이들 제형은 제약 화학에 일반적으로 공지된 처방서에 기술되어 있다. In the case of transdermal administration, ointments, creams, lotions, gels, external solutions, pasta, liniment, air rolls, and the like are included. In the above,'transdermal administration' means that the active ingredient in an effective amount contained in the pharmaceutical composition is delivered into the skin by topically administering the pharmaceutical composition to the skin. For example, the pharmaceutical composition of the present invention may be prepared in an injectable formulation and administered by lightly pricking the skin with a 30 gauge thin injection needle or applying it directly to the skin. These formulations are described in prescriptions generally known in pharmaceutical chemistry.
흡입투여제의 경우, 본 발명에 따라 사용되는 화합물은 적합한 추진제, 예를 들면, 디클로로플루오로메탄, 트리클로로플루오로메탄, 디클로로테트라플루오로에탄, 이산화탄소 또는 다른 적합한 기체를 사용하여, 가압 팩 또는 연무기로부터 에어로졸 스프레이 형태로 편리하게 전달할 수 있다. 가압 에어로졸의 경우, 투약 단위는 계량된 양을 전달하는 밸브를 제공하여 결정할 수 있다. 예를 들면, 흡입기 또는 취입기에 사용되는 젤라틴 캡슐 및 카트리지는 화합물 및 락토오즈 또는 전분과 같은 적합한 분말 기제의 분말 혼합물을 함유하도록 제형화할 수 있다.In the case of inhalation administration, the compounds used according to the invention can be used in pressurized packs or with suitable propellants, for example dichlorofluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. It can be conveniently delivered from a nebulizer in the form of an aerosol spray. In the case of a pressurized aerosol, the dosage unit can be determined by providing a valve that delivers a metered amount. For example, gelatin capsules and cartridges for use in an inhaler or insufflator can be formulated to contain a powder mixture of the compound and a suitable powder base such as lactose or starch.
그 밖의 약학적으로 허용되는 담체로는 당업계에 공지되어 있는 것을 참고로 할 수 있다.Other pharmaceutically acceptable carriers may refer to those known in the art.
본 발명에 따른 약학적 조성물은 하나 이상의 완충제(예를 들어, 식염수 또는 PBS), 카보하이트레이트(예를 들어, 글루코스, 만노즈, 슈크로즈 또는 덱스트란), 항산화제, 정균제, 킬레이트화제(예를 들어, EDTA 또는 글루타치온), 아쥬반트(예를 들어, 알루미늄 하이드록사이드), 현탁제, 농후제 및/또는 보존제를 추가로 포함할 수 있다.Pharmaceutical compositions according to the present invention include one or more buffers (e.g., saline or PBS), carbohythrate (e.g., glucose, mannose, sucrose or dextran), antioxidants, bacteriostatic agents, chelating agents (e.g. For example, EDTA or glutathione), adjuvants (eg, aluminum hydroxide), suspending agents, thickening agents and/or preservatives may further be included.
또한, 본 발명의 약학적 조성물은 포유동물에 투여된 후 활성 성분의 신속, 지속 또는 지연된 방출을 제공할 수 있도록 당업계에 공지된 방법을 사용하여 제형화될 수 있다. In addition, the pharmaceutical compositions of the present invention may be formulated using methods known in the art to provide rapid, sustained or delayed release of the active ingredient after administration to a mammal.
또한, 본 발명의 약학적 조성물은 단독으로 투여하거나, 퇴행성 신경질환 치료의 효과가 있는 공지의 화합물과 병용하여 투여할 수 있다. Further, the pharmaceutical composition of the present invention may be administered alone or may be administered in combination with a known compound having an effect of treating neurodegenerative diseases.
또한 본 발명은 In addition, the present invention
(a) CD9 단백질 또는 mRNA를 발현하는 세포에 시험물질을 처리하는 단계;(a) treating cells expressing CD9 protein or mRNA with a test substance;
(b) 상기 세포와 시험물질을 처리하지 않은 대조군 세포에서 CD9 단백질 또는 mRNA의 발현 수준을 측정하는 단계; 및(b) measuring the expression level of CD9 protein or mRNA in the cells and control cells not treated with the test substance; And
(c) 대조군 세포와 비교하여 CD9 단백질 또는 mRNA의 발현 수준을 감소시키는 시험 물질을 퇴행성 신경질환 치료제 후보물질로 선별하는 단계를 포함하는 퇴행성 신경질환 치료제 스크리닝 방법을 제공한다. (c) It provides a screening method for a neurodegenerative disease treatment comprising the step of selecting a test substance that reduces the expression level of CD9 protein or mRNA compared to the control cell as a candidate substance for treatment of neurodegenerative disease.
상기 (a) 단계는 분석의 대상인 시험 물질이 CD9 단백질 또는 mRNA의 발현을 억제하는 활성이 있는지 확인하기 위하여 CD9 단백질 또는 mRNA를 발현하는 세포에 접촉시키는 단계이다.The step (a) is a step of contacting cells expressing CD9 protein or mRNA in order to determine whether the test substance to be analyzed has an activity to inhibit the expression of CD9 protein or mRNA.
본 발명의 방법에서 '접촉(contacting)' 또는 '처리(treatment)'는 일반적인 의미이며, 2개 이상의 제제(예를 들어, 2개의 리펩티드)를 결합시키거나, 제제와 세포(예를 들어, 단백질과 세포)를 결합시키는 것을 말한다. 접촉은 시험관 내(in vitro)에서 일어날 수 있다. 예컨대, 시험관(test tube) 또는 다른 컨테이너(container)에서 2개 이상의 제제를 결합시키거나 시험 제제와 세포 또는 세포 용해물과 시험 제제를 결합시키는 것이다. 또한 접촉은 세포 또는 인 시투(in situ)에서 일어날 수도 있다. 예컨대, 2개의 폴리펩티드를 암호화하는 재조합 폴리뉴클레오티드를 세포 내에서 공동발현(coexpression)시킴으로써 세포 또는 세포 용해물에서 2개의 폴리펩티드를 접촉시키는 것이다. 또한 테스트하고자 하는 단백질이 고정상의 표면에 배열된 단백질 칩(protein chip)이나 단백질 어레이(protein array)를 이용할 수도 있다.In the method of the present invention,'contacting' or'treatment' is a general meaning, and two or more agents (e.g., two polypeptides) are combined, or an agent and a cell (e.g., Protein and cell). Contact can occur in vitro. For example, combining two or more agents in a test tube or other container, or combining a test agent with cells or cell lysates and a test agent. Contact can also occur in cells or in situ. For example, two polypeptides are brought into contact in a cell or cell lysate by coexpressing a recombinant polynucleotide encoding two polypeptides in a cell. In addition, a protein chip or a protein array in which the protein to be tested is arranged on the surface of a stationary bed may be used.
또한 본 발명의 방법에서 '시험 물질'은 시험 제제(test agent) 또는 제제(agent)와 호환가능하게 사용할 수 있는 것으로, 임의의 물질(substance), 분자(molecule), 원소(element), 화합물(compound), 실재물(entity) 또는 이들의 조합을 포함한다. 예를 들어 단백질, 폴리펩티드, 저분자 유기화합물(small organic molecule), 다당류(polysaccharide), 폴리뉴클레오티드 등을 포함한다. 또한 자연 산물(natural product), 합성 화합물 또는 화학 화합물 또는 2개 이상의 물질의 조합일 수도 있다.In addition, in the method of the present invention, the'test substance' can be used interchangeably with a test agent or agent, and any substance, molecule, element, compound ( Includes a compound, entity, or combination thereof. Examples include proteins, polypeptides, small organic molecules, polysaccharides, and polynucleotides. It may also be a natural product, a synthetic compound or a chemical compound, or a combination of two or more substances.
보다 구체적으로 본 발명의 방법으로 스크리닝할 수 있는 시험 제제는, 폴리펩티드, 베타-턴 유도체(beta-turn mimetics), 다당류, 인지질, 호르몬, 프로스타글란딘, 스테로이드, 방향족 화합물, 헤테로사이클릭 화합물, 벤조디아제핀(benzodiazepines), 올리고머릭 N-치환 글리신(oligomeric N-substituted glycines), 올리고카르바메이트(oligocarbamates), 당류(saccharides), 지방산, 퓨린, 피리미딘 또는 이들의 유도체, 구조적 아날로그 또는 이들의 조합을 포함한다. 시험 제제는 합성 물질 또는 천연물질일 수 있다. 상기 시험 제제는 합성 또는 자연 화합물의 라이브러리를 포함하는 광범위하고 다양한 출처로부터 얻어질 수 있다. 조합(combinatorial) 라 이브러리는 스텝-바이-스텝 방식으로 합성될 수 있는 여러 종류의 화합물로 생산될 수 있다. 다수의 조합 라이브러리의 화합물들은 ESL(encoded synthetic libraries) 방법(WO 95/12608, WO93/06121, WO 94/08051, WO95/395503 및 WO 95/30642)에 의해 제조될 수 있다. 펩티드 라이브러리는 파지 디스플레이 방법(WO91/18980)에 의해 제조될 수 있다. 박테리아, 곰팡이, 식물 및 동물 추출물 형태의 자연 화합물의 라이브러리는 상업적인 출처로부터 얻거나 또는 필드(field)에서 수집될 수 있다. 공지된 약리학적(pharmacological) 제제가 구조적 아날로그를 제조하기 위하여 아실화, 알킬화, 에스테르화 반응(esterification), 아미드화 반응(amidification)과 같이 지시되거나(direct) 무작위한 화학적 수식에 적용될 수 있다.More specifically, test agents that can be screened by the method of the present invention include polypeptides, beta-turn mimetics, polysaccharides, phospholipids, hormones, prostaglandins, steroids, aromatic compounds, heterocyclic compounds, benzodiazepines. ), oligomeric N-substituted glycines, oligocarbamates, saccharides, fatty acids, purines, pyrimidines or derivatives thereof, structural analogues, or combinations thereof. Test formulations can be synthetic or natural. The test formulations can be obtained from a wide variety of sources, including libraries of synthetic or natural compounds. Combinatorial libraries can be produced from several types of compounds that can be synthesized in a step-by-step manner. Compounds of multiple combinatorial libraries can be prepared by the encoded synthetic libraries (ESL) method (WO 95/12608, WO93/06121, WO 94/08051, WO95/395503 and WO 95/30642). Peptide libraries can be prepared by the phage display method (WO91/18980). Libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts can be obtained from commercial sources or collected in the field. Known pharmacological agents can be directed or applied to a random chemical modification, such as acylation, alkylation, esterification, amidification, to prepare structural analogues.
상기 시험 제제는 자연적으로 생성되는 단백질 또는 이의 단편일 수 있다. 이런 시험 제제는 자연 출처(natural source), 예컨대, 세포 또는 조직 용해물로부터 수득될 수 있다. 폴리펩티드 제제의 라이브러리는 예컨대, 통상적인 방법에 의해 생성되거나 상업적으로 입수할 수 있는 cDNA 라이브러리로부터 수득될 수 있다. 상기 시험 제제는 펩티드, 예컨대, 약 5~30개, 바람직하게는 약 5~20개, 보다 바람직하게는 약 7~15개의 아미노산을 가지는 펩티드일 수 있다. 상기 펩티드는 자연적으로 생성되는 단백질, 랜덤 펩티드 또는 '바이어스(biased)' 랜덤 펩티드의 절단물일 수 있다. 또한 상기 시험 제제는 핵산일 수 있다. 핵산 시험 제제는 자연적으로 생성되는 핵산, 랜덤 핵산, 또는 '바이어스(biased)' 랜덤 핵산일 수 있다. 예컨대, 원핵 또는 진핵 게놈의 절단물을 위에서 기재한 바와 유사하게 사용할 수 있다.The test agent may be a naturally occurring protein or fragment thereof. Such test formulations can be obtained from natural sources, such as cell or tissue lysates. Libraries of polypeptide preparations can be produced, for example, by conventional methods or obtained from commercially available cDNA libraries. The test agent may be a peptide, such as a peptide having about 5 to 30 amino acids, preferably about 5 to 20, more preferably about 7 to 15 amino acids. The peptide may be a naturally occurring protein, a random peptide, or a cut product of a'biased' random peptide. In addition, the test agent may be a nucleic acid. The nucleic acid test agent can be a naturally occurring nucleic acid, a random nucleic acid, or a'biased' random nucleic acid. For example, cuts of prokaryotic or eukaryotic genomes can be used analogously to those described above.
또한 상기 시험 제제는 소분자(small molecule; 예를 들어, 약 1,000Da이하의 분자량을 갖는 분자)일 수 있다. 소분자의 조절 제제를 스크리닝하기 위한 방법에는 바람직하게는 고속 분석 어세이(high throughput assay)가 적용될 수 있다. 많은 어세이가 상기 스크리닝에 유용하다.Further, the test agent may be a small molecule (eg, a molecule having a molecular weight of about 1,000 Da or less). A high throughput assay may be preferably applied to a method for screening a small molecule modulating agent. Many assays are useful for this screening.
본 명세서에서 '발현(expression)'이라 함은 세포에서 단백질 또는 핵산이 생성되는 것을 의미한다. 상기 CD9를 발현하는 세포는 CD9를 내재적으로 발현하는 세포일 수도 있고, CD9를 암호화하는 폴리뉴클레오티드를 포함하는 재조합 발현 벡터로 형질전환되어 CD9를 과발현하는 세포일 수도 있다. 바람직하게는 상기 CD9 유전자를 발현하는 세포는 섬유아세포(fibroblast)일 수 있다. In the present specification, "expression" means that a protein or nucleic acid is produced in a cell. The CD9-expressing cell may be a cell that internally expresses CD9, or may be a cell that overexpresses CD9 by being transformed with a recombinant expression vector containing a polynucleotide encoding CD9. Preferably, the cells expressing the CD9 gene may be fibroblasts.
상기 (b) 단계는 시험 물질을 접촉시킨 CD9를 발현하는 세포와 시험 물질을 접촉시키지 않은 CD9를 발현하는 세포에서 CD9의 유전자 발현 수준을 측정하는 단계이다.Step (b) is a step of measuring the gene expression level of CD9 in cells expressing CD9 contacted with the test substance and cells expressing CD9 without contact with the test substance.
CD9 mRNA 발현의 측정은 당업계에서 통상적인 발현 수준 확인 방법을 제한 없이 사용할 수 있으며, 분석 방법의 예로 역전사중합체연쇄반응(reverse transcription polymerase chain reaction, RT-PCR), 경쟁적 RT-PCR(competitive RT-PCR), 실시간 RT-PCR(real-time RT-PCR), RNase 보호 분석법(RNase protection assay, RPA), 노던 블랏팅(northern blotting), DNA 마이크로어레이 칩(microarray chip), RNA 염기서열분석(RNA sequencing) 등이 있으나, 이들에 한정되는 것은 아니다.The measurement of CD9 mRNA expression can be performed without limitation, using conventional methods for determining the expression level in the art, and examples of analysis methods include reverse transcription polymerase chain reaction (RT-PCR), competitive RT-PCR (competitive RT- PCR), real-time RT-PCR, RNase protection assay (RPA), northern blotting, DNA microarray chip, RNA sequencing (RNA) sequencing) and the like, but are not limited thereto.
또한 CD9 단백질의 발현 수준 측정 방법은 당업계에서 공지되어 있는 방법은 제한 없이 사용할 수 있으며, 그 예로 웨스턴 블랏팅(western blotting), 닷 블랏팅(dot blotting), 효소면역분석법(enzyme-linked immunosorbent assay), 방사능 면역분석법(RIA), 방사면역확산법, 오우크테로니 면역 확산법, 로케트 면역 전기영동, 면역조직화학염색, 면역침전법(immunoprecipitation), 보체 고정 분석법, 유세포 분석법(FACS) 또는 단백질 칩 방법 등이 있으나, 이들에 한정되는 것은 아니다.In addition, methods for measuring the expression level of CD9 protein can be used without limitation, methods known in the art, such as western blotting, dot blotting, and enzyme-linked immunosorbent assay. ), Radioactive Immunoassay (RIA), Radioimmune Diffusion Method, Ouchteroni Immunity Diffusion Method, Rocket Immunoelectrophoresis, Immunohistochemical Staining, Immunoprecipitation, Complement Fixation Assay, Flow Cytometry (FACS) or Protein Chip Method And the like, but are not limited thereto.
상기 (c) 단계는 대조군 세포와 비교하여 CD9 단백질 또는 mRNA의 발현 수준을 감소시키는 시험 물질을 퇴행성 신경질환 치료제 후보물질로 선별하는 단계이다. The step (c) is a step of selecting a test substance that reduces the expression level of CD9 protein or mRNA compared to the control cells as a candidate substance for treating neurodegenerative diseases.
본 발명자가 규명한 바와 같이, CD9의 활성을 저해하면 퇴행성 신경질환 동물모델에서 기억력이 향상되는 바와 같이, CD9의 단백질 또는 mRNA의 발현을 저해하는 물질 또한 CD9의 활성을 저해시키는 효과를 발휘하는 바 퇴행성 신경질환 치료제로서 활용될 가능성이 있다는 것이 자명하게 이해될 수 있다. As identified by the present inventors, as inhibiting the activity of CD9 improves memory in an animal model of neurodegenerative disease, substances that inhibit the expression of protein or mRNA of CD9 also exhibit the effect of inhibiting the activity of CD9. It can be clearly understood that it has the potential to be used as a therapeutic agent for neurodegenerative diseases.
본 발명은 퇴행성 신경질환의 예방 또는 치료용 제제를 제조하기 위한 CD9 단백질의 발현 또는 활성 억제제의 용도를 제공한다.The present invention provides a use of a CD9 protein expression or activity inhibitor for preparing a preparation for preventing or treating neurodegenerative diseases.
본 발명은 CD9 단백질의 발현 또는 활성 억제제를 유효성분으로 포함하는 조성물의 유효량을 이를 필요로 하는 개체에 투여하는 단계를 포함하는 퇴행성 신경질환의 치료 방법을 제공한다.The present invention provides a method for treating neurodegenerative diseases comprising administering an effective amount of a composition comprising an inhibitor of expression or activity of a CD9 protein as an active ingredient to an individual in need thereof.
본 발명의 상기 ‘유효량’이란 개체에게 투여하였을 때, 퇴행성 신경질환의 개선, 치료, 예방, 검출, 진단 또는 퇴행성 신경질환의 억제 또는 감소 효과를 나타내는 양을 말하며, 상기‘개체’란 동물, 바람직하게는 포유동물, 특히 인간을 포함하는 동물일 수 있으며, 동물에서 유래한 세포, 조직, 기관 등일 수도 있다. 상기 개체는 상기 효과가 필요한 환자(patient) 일 수 있다.The'effective amount' of the present invention refers to an amount showing an effect of improving, treating, preventing, detecting, diagnosing, or inhibiting or reducing neurodegenerative diseases when administered to an individual, and the'individual' is an animal, preferably For example, it may be an animal including a mammal, especially a human, and may be a cell, tissue, organ, etc. derived from an animal. The individual may be a patient in need of the effect.
본 발명의 상기 ‘치료’는 퇴행성 신경질환 또는 퇴행성 신경질환의 증상을 개선시키는 것을 포괄적으로 지칭하고, 이는 퇴행성 신경질환을 치유하거나, 실질적으로 예방하거나, 또는 상태를 개선시키는 것을 포함할 수 있으며, 퇴행성 신경질환으로부터 비롯된 한 가지 증상 또는 대부분의 증상을 완화시키거나, 치유하거나 예방하는 것을 포함하나, 이에 제한되는 것은 아니다.The'treatment' of the present invention generally refers to improving the symptoms of neurodegenerative diseases or neurodegenerative diseases, which may include curing, substantially preventing, or improving the condition, It includes, but is not limited to, alleviating, curing, or preventing one symptom or most of the symptoms resulting from neurodegenerative disease.
본 발명의 용어 ‘~을 포함하는(comprising)’이란 ‘함유하는’ 또는 ‘특징으로 하는’과 동일하게 사용되며, 조성물 또는 방법에 있어서, 언급되지 않은 추가적인 성분 요소 또는 방법 단계 등을 배제하지 않는다. 용어 ‘~로 구성되는(consisting of)’이란 별도로 기재되지 않은 추가적인 요소, 단계 또는 성분 등을 제외하는 것을 의미한다. 용어 ‘필수적으로 구성되는(essentially consisting of)’이란 조성물 또는 방법의 범위에 있어서, 기재된 성분 요소 또는 단계와 더불어 이의 기본적인 특성에 실질적으로 영향을 미치지 않는 성분 요소 또는 단계 등을 포함하는 것을 의미한다.The term'comprising' in the present invention is used in the same way as'containing' or'as a feature', and does not exclude additional component elements or method steps that are not mentioned in the composition or method. . The term “consisting of” means excluding additional elements, steps, or ingredients that are not separately described. The term “essentially consisting of” means including, in the scope of a composition or method, the component elements or steps described, as well as the component elements or steps that do not substantially affect its basic properties.
CD9의 발현 또는 활성을 저해하는 물질은 퇴행성 신경질환 환자의 기억력을 개선하는 효과를 발휘하여 퇴행성 신경질환 치료제 개발에 유용하게 활용이 될 수 있으며, CD9의 발현 수준 분석을 통해서 퇴행성 신경질환 치료제 후보물질의 스크리닝할 수 있다.Substances that inhibit the expression or activity of CD9 have the effect of improving the memory of patients with neurodegenerative diseases and can be usefully used in the development of treatments for neurodegenerative diseases, and candidates for treatment of neurodegenerative diseases through analysis of the expression level of CD9 Can be screened.
도 1은 permeabilization을 하지 않은 AD 섬유아세포에서 CD9의 발현을 확인한 결과이다.1 is a result of confirming the expression of CD9 in AD fibroblasts not subjected to permeabilization.
도 2는 permeabilization을 한 경우의 AD 섬유아세포에서 CD9의 발현을 확인한 결과이다.2 is a result of confirming the expression of CD9 in AD fibroblasts in the case of permeabilization.
도 3은 AD 섬유아세포에서 항-CD9 항체의 처리에 따른 타우 인산화를 나타낸 것이다.Figure 3 shows tau phosphorylation according to treatment with anti-CD9 antibody in AD fibroblasts.
도 4a 내지 4f는 anti-CD9Ab을 투여에 따른 기억력 개선 효과를 확인한 것이다. 각각 일반적인 운동 활성(도 4a). 약물을 투여 한 마우스의 교대 반복 횟수(도 4b), 문맥 회상(contextual recall)(도 4c), freezing 시간 (도 4d), Morris 물 미로에서의 표적 영역에 대한 도달 수(도 4e) 및 표적 영역에서 소비한 시간(도 4f)을 측정하였다.4A to 4F show the effect of improving memory by administering anti-CD9Ab. Each general motor activity (Fig. 4A). The number of alternating repetitions of mice administered with the drug (Fig. 4b), contextual recall (Fig. 4c), freezing time (Fig. 4d), the number of arrivals to the target area in the Morris water maze (Fig. 4e) and the target area The time spent in (Fig. 4f) was measured.
이하 본 발명을 상세히 설명한다.Hereinafter, the present invention will be described in detail.
단, 하기 실시예는 본 발명을 예시하는 것일 뿐, 본 발명의 내용이 하기 실시예에 한정되는 것은 아니다.However, the following examples are only illustrative of the present invention, and the contents of the present invention are not limited to the following examples.
실험방법Experiment method
1. 세포, 항체 및 실험동물의 준비1. Preparation of cells, antibodies and experimental animals
암컷 3xTg 생쥐 (JAX 004807)를 실험동물로 사용하였다. 실험동물은 멸균 사료와 물을 제공하고 각각 10 마리 미만의 동물로 사육하였다. 밤낮 주기는 12시간으로 하였다. 동물실험은 동물실험윤리위원회의 지도, 감독하에 고려대학교 동물사육 시설 내에서 진행하였고, 동물실험 절차는 고려대학교 IACUC의 승인된 프로토콜에 따라 수행하였다.Female 3xTg mice (JAX 004807) were used as experimental animals. Experimental animals were provided with sterile feed and water, and were reared with less than 10 animals each. The day and night cycle was set to 12 hours. Animal experiments were conducted in an animal breeding facility at Korea University under the guidance and supervision of the Animal Experimental Ethics Committee, and animal experiments were performed according to the protocol approved by Korea University IACUC.
시험 항체접종(CD9)은 PBS(phosphate buffer solution)과 혼합하여 준비하고 8~12주령 생쥐의 피하로 100ul 주입하였다. 실험별 그룹으로 생쥐 (그룹 당 n=4)를 구분하고 대조군으로는 PBS(control)를 사용하였다. 항체는 1주에 한번 씩 총 4회 주사하여 실험을 진행하였다.Test antibody inoculation (CD9) was prepared by mixing with PBS (phosphate buffer solution), and 100ul was injected subcutaneously in 8-12 week old mice. Mice (n=4 per group) were divided into groups for each experiment, and PBS (control) was used as a control group. The experiment was carried out by injecting the antibody a total of 4 times once a week.
실험에는 Coriell사 (Camden, NJ)에서 구입한 AD 섬유아세포 두 종류와 (AG07374(AD fibroblast 1), AG05770(AD fibroblast 2)) age-control 섬유아세포 (AG09857), 그리고 본 실험실에서 추출한 정상인의 편도선 섬유아세포를 사용하였다.In the experiment, two types of AD fibroblasts purchased from Coriell (Camden, NJ), (AG07374(AD fibroblast 1), AG05770(AD fibroblast 2)) age-control fibroblasts (AG09857), and normal human tonsils extracted in this laboratory. Fibroblasts were used.
또한, Mouse control IgG1, anti-CD9 ALB6 (Immunotech)을 1차 항체로 사용하였고, FITC로 표지된 goat anti-mouse IgG(Jackson Immunoresearch)를 2차 항체로 사용하였다. In addition, Mouse control IgG1, anti-CD9 ALB6 (Immunotech) was used as the primary antibody, and goat anti-mouse IgG (Jackson Immunoresearch) labeled with FITC was used as the secondary antibody.
2. FACS 염색2. FACS staining
배양 중인 섬유아세포에 trypsin-EDTA를 첨가하여 세포를 준비하였다. 세포의 절반은 Becton Dickinson Bioscience에서 구입한 FACS permeabilizing solution 2로 15분 동안 처리하였고, 나머지 절반은 4℃에 방치하였다. 세포를 1% FBS (Fetal bovine serum)와 0.05% sodium azide가 포함된 PBS로 세척 후, FACS tube로 옮기고, 1차 항체와 4℃에서 15분 동안 반응시켰다. FACS wash buffer로 세척 후 2차 항체를 첨가하여 4℃에서 15분 동안 반응시킨 다음 FACS buffer로 세척 후 BD FACSCalibur로 CD9 발현을 분석하였다.Cells were prepared by adding trypsin-EDTA to fibroblasts in culture. Half of the cells were treated with FACS permeabilizing solution 2 purchased from Becton Dickinson Bioscience for 15 minutes, and the other half was left at 4°C. The cells were washed with PBS containing 1% Fetal bovine serum (FBS) and 0.05% sodium azide, transferred to a FACS tube, and reacted with the primary antibody at 4°C for 15 minutes. After washing with FACS wash buffer, secondary antibody was added and reacted at 4° C. for 15 minutes. After washing with FACS buffer, CD9 expression was analyzed with BD FACSCalibur.
3. 동물 행동실험3. Animal behavior experiment
3-1. Y-미로3-1. Y-maze
Y-미로 장치는 26cm 벽 (San Diego Instruments, San Diego, CA)을 갖는 32cm (길이) × 10cm (폭)의 3개의 암(arm)으로 구성되었다. 각 마우스를 Y-미로의 중앙에 놓고 5분 동안 미로를 통해 자유롭게 이동할 수 있게 하였다. 마우스 4개의 발이 모두 미로의 암(arm)에 들어가면 유효한 것으로 간주하였다. 교대(alternation)는 3개의 서로 다른 암(즉, 1, 2, 3 또는 2, 3, 1 등)에 3연속으로 들어가는 것으로 정의되었다. 백분율 교대 점수는 다음 공식을 사용하여 계산하였다. The Y-maze device consisted of three arms 32 cm (length) x 10 cm (width) with 26 cm walls (San Diego Instruments, San Diego, CA). Each mouse was placed in the center of the Y-maze and allowed to move freely through the maze for 5 minutes. It was considered effective if all four paws of the mouse entered the arm of the maze. Alternation was defined as entering 3 different arms (i.e. 1, 2, 3 or 2, 3, 1, etc.) in 3 succession. The percentage shift score was calculated using the following formula.
spontaneous alternation % = (number of spontaneous)/(total number of arm entries -2) x 100spontaneous alternation% = (number of spontaneous)/(total number of arm entries -2) x 100
또한, 총 암(arm) 갯수는 동물의 일반적인 활동의 척도로 사용되었다. 미로를 70% 에탄올로 깨끗이 닦아 악취 신호를 최소화했다. In addition, the total number of arms was used as a measure of the general activity of the animal. The maze was cleaned with 70% ethanol to minimize the odor signal.
3-2. Morris Water Maze 3-2. Morris Water Maze
사용된 장치는 벽이 76cm 인 흰색 원형 플라스틱 탱크 (직경 122 cm)로, 22u +/- 22uC로 유지된 물로 채웠으며, 독성이 없는 흰색 페인트를 추가하여 불투명하게 만들었으며 내부에는 분리 가능한 정사각형 (측면 길이 10cm) Plexiglas 플랫폼. 탱크는 여러 가지 눈에 띄는 시각적 신호를 포함하는 시험실에 있었다. 첫 번째 세션의 첫 번째 시험 전에 마우스를 플랫폼에 10초 동안 두었다. 마우스는 물의 표면 아래 1.5cm에 잠긴 플랫폼으로 수영하도록 훈련 받았다. 플랫폼은 탱크의 중앙과 벽에서 등거리에 고정된 위치에 있었다. 마우스는 하루에 3회의 훈련을 받았다. 각 훈련기간 동안, 마우스는 4개의 지정된 시작점 중 하나에서 무작위 순서로 탱크에 넣었다. 마우스는 침수된 플랫폼에서 찾아 탈출할 수 있도록 했다. 60 초 이내에 플랫폼을 찾지 못하면 수동으로 플랫폼으로 안내되어 10초 동안 그대로 있게했다. 마우스는 20초 (탈출 대기 시간)의 훈련 기준에 도달하도록 훈련되었다. 마우스는 마지막 트레이닝 세션 후 24 시간 동안 프로브 시험에서 평가되었으며, 플랫폼이 없는 풀에서 60초의 자유 수영으로 구성되었다. 각 동물의 성능은 획득 매개 변수 (플랫폼 및 거리 추적을 찾는 대기 시간) 및 probe-trial 매개 변수 (대상 플랫폼 영역과 에 대한 항목 수)에 대한 데이터를 제공하는 Any-Maze TM 비디오 추적 시스템 (Stoelting Co.)를 사용하여 모니터링하였다.The device used was a white round plastic tank (122 cm in diameter) with a wall of 76 cm, filled with water held at 22u +/- 22uC, made opaque by adding non-toxic white paint, and a detachable square inside (side 10 cm long) Plexiglas platform. The tank was in a test room containing a number of visible visual cues. Mice were placed on the platform for 10 seconds before the first test of the first session. Mice were trained to swim on a platform submerged 1.5 cm below the surface of the water. The platform was in a fixed position equidistant from the center and wall of the tank. Mice were trained three times a day. During each training period, mice were placed in tanks in random order at one of the four designated starting points. The mouse was allowed to find and escape from the submerged platform. If the platform was not found within 60 seconds, it was manually guided to the platform and left there for 10 seconds. Mice were trained to reach a training criterion of 20 seconds (escape waiting time). Mice were evaluated in the probe test for 24 hours after the last training session and consisted of 60 seconds of free swimming in a platformless pool. The performance of each animal acquisition parameters provide data on (waiting to find the platform and distance tracking time), and probe-trial parameters (the number of entries for the target platform area) Any-Maze TM video tracking system (Stoelting Co .) was used.
4. 웨스턴 블롯 분석4. Western Blot Analysis
단백질 농도는 Bradford Assay(Bio-Rad Protein Assay 500-0006, Germany, Munchen)를 사용하여 정량하였고 전체 세포 단백질 또는 age-control 섬유아세포, AD-섬유아세포를 8-10% SDS 폴리아크릴 아미드 겔로 분리하였다. Polyvinylidene difluoride(PVDF) 막은 트리스 완충식염수(5% 지방 분유 없음, 0.1% Tween20)에서 블로킹되었다. 1차 항체로, anti PHF-tau clone AT8(MN1020, Thermo Scientific, Rockford, USA), p타우 pSer422(T7944, 시그마), anti-tau clone tau-5(MAB361, Millipore Billerica, Ma), CD9를 사용하였다. 모든 도말은 액틴(MAB1501, Chemicon-Millipore, Billerica, Ma)으로 표준화되었다. Protein concentration was quantified using Bradford Assay (Bio-Rad Protein Assay 500-0006, Germany, Munchen), and total cell proteins, age-control fibroblasts, and AD-fibroblasts were isolated with 8-10% SDS polyacrylamide gel. . Polyvinylidene difluoride (PVDF) membrane was blocked in Tris buffered saline (5% no fat formula, 0.1% Tween20). As the primary antibody, anti PHF-tau clone AT8 (MN1020, Thermo Scientific, Rockford, USA), p Tau pSer422 (T7944, Sigma), anti-tau clone tau-5 (MAB361, Millipore Billerica, Ma), CD9 were used. I did. All smears were standardized with actin (MAB1501, Chemicon-Millipore, Billerica, Ma).
실험결과Experiment result
1. AD 섬유아세포에서 CD9의 발현 확인1. Confirmation of CD9 expression in AD fibroblasts
Permeabilization을 하지 않은 세포의 경우, age-control 섬유아세포는 CD9을 거의 발현하지 않았으나 AD 섬유아세포는 두 종류 모두 CD9을 높은 수준으로 발현하였다(도 1). 이 결과는 세포막에 CD9 분자가 위치한다는 이전 자료에 부합하는 것은 물론 두 환자 그룹간의 차이는 알츠하이머 병의 진행정도와 관련 있는 것으로 추정되었다. 또한, 정상인의 편도선(Tonsil) 섬유아세포의 CD9 발현도 높은 수준인 것을 확인하였다.In the case of cells not subjected to permeabilization, age-control fibroblasts hardly expressed CD9, but AD fibroblasts expressed CD9 at high levels in both types (FIG. 1). This result is consistent with previous data indicating the location of the CD9 molecule in the cell membrane, as well as the difference between the two patient groups was estimated to be related to the progression of Alzheimer's disease. In addition, it was confirmed that CD9 expression was also high in Tonsil fibroblasts in normal humans.
또한, 도 2에서 보듯이, Permeabilization 후에는 여전히 CD9은 age-control fibroblast는 발현되지 않았다. 또한, 세포막에서 AD fibroblast 2에 비하여 상대적으로 CD9의 낮은 발현하는 것으로 나타난 AD fibroblast 1는 Permeabilization 후에도 거의 발현되지 않았다. 이는 permeabilization 처리 단계에서 세포 표면 및 세포 내부에 있는 CD9 분자가 소실된 것으로 판단된다. 한편, 세포막 단백질인 CD9이 Permeabilization 후 AD fibroblast 2에서는 높은 수준으로 발현하는 것으로 나타났다. 이것은 alzheimer 병의 진행정도에 따라서는 정상적으로는 세포막에만 존재하는 CD9 단백이 nucleoplasm 또는 핵에도 존재하는 것으로 해석되었다. In addition, as shown in Figure 2, after permeabilization, CD9 still did not express age-control fibroblast. In addition, AD fibroblast 1, which showed relatively lower expression of CD9 than AD fibroblast 2 in cell membranes, was hardly expressed even after permeabilization. It is believed that the CD9 molecules on the cell surface and inside the cell were lost in the permeabilization step. Meanwhile, it was found that CD9, a cell membrane protein, was expressed at a high level in AD fibroblast 2 after permeabilization. It was interpreted that the CD9 protein, which normally exists only in the cell membrane, is also present in the nucleoplasm or the nucleus, depending on the progression of alzheimer's disease.
2. AD 섬유아세포의 타우(Tau) 과인산화 확인2. Confirmation of Tau hyperphosphorylation in AD fibroblasts
타우 과인산화를 통한 신경 퇴행 탈락된 β-아밀로이드는 타우 병증을 자극할 수 있으며 타우의 과인산화는 알츠하이머 병에서 신경 섬유 엉킴 (neurofibrillary tangles, NFTs)과 신경 퇴화를 일으키는 미세 소관 조립의 변화와 관련되어 있다. 뿐만 아니라 알츠하이머 병과 파킨슨 병을 비롯한 많은 다른 신경계 질환과 관련이 있다. 본 발명에서 항-CD9 항체를 투여하여 타우의 인산화를 확인한 결과, AD 섬유아세포에서 인산화된 타우(p-Ser422)의 수준(control lane)은 age control 섬유아세포(미도시)에 비해 현저하게 상승하였으며, 항체를 투여한 경우 현저하게 감소(anti-CD9-Ab panel)되는 것을 확인하였다, 하지만, 전체 타우 양은 의미 있는 변화를 보이지 않았다 (도 3). Neurodegeneration through tau hyperphosphorylation Depleted β-amyloid may stimulate tauopathy, and tau hyperphosphorylation is associated with changes in microtubule assembly leading to neurofibrillary tangles (NFTs) and neurodegeneration in Alzheimer's disease. have. In addition, it has been linked to many other neurological disorders, including Alzheimer's and Parkinson's. In the present invention, as a result of confirming the phosphorylation of tau by administration of anti-CD9 antibody, the level (control lane) of phosphorylated tau (p-Ser422) in AD fibroblasts was significantly increased compared to age control fibroblasts (not shown). , When the antibody was administered, it was confirmed that it was significantly reduced (anti-CD9-Ab panel), but the total amount of tau did not show any significant change (FIG. 3 ).
3. Tg 마우스에서의 기억력 개선 효과 3. The effect of improving memory in Tg mice
Y-maze에서 anti-CD9 Ab을 투여한 4 마리의 Tg 마우스 (3xTg+Ab)와 위약으로 처리한 그룹(3xTg)은 총 암 출입수에 유의한 차이가 없었으므로 일반적인 운동 활성에는 차이가 없음을 알 수 있었다(도 4A). 그러나 약물을 투여 한 마우스의 교대 반복 횟수는 대조군에 비해 높았으며 백분율 값이 높았다(도 4B, #p<0.05). Four Tg mice (3xTg+Ab) treated with anti-CD9 Ab in Y-maze and the group treated with placebo (3xTg) had no significant difference in total cancer entry and exit, so there was no difference in general exercise activity. It was found (Fig. 4A). However, the number of alternating repetitions of the mice administered with the drug was higher than that of the control group and the percentage value was higher (Fig. 4B, # p<0.05).
공포 컨디셔닝 테스트에서 두 그룹 모두 훈련 단계에서 차이가 나타나지 않았다(데이터는 표시되지 않음). 그러나, anti-CD9Ab을 투여받은 3xTg 마우스는 위약을 투여한 동물군보다 문맥 회상(contextual recall)에서는 freezing 시간에서 의미있는 차이를 보이지 않았다(도 4C). anti-CD9Ab을 투여한 3×Tg 마우스는 위약을 투여한 패밀리보다 cued recall 패러다임에서도 유의하게 더 높은 freezing 시간을 보였다 (도 4D, #p<0.05).In the fear conditioning test, there was no difference between the two groups at the training stage (data not shown). However, 3xTg mice receiving anti-CD9Ab did not show a significant difference in freezing time in contextual recall than in the animal group receiving placebo (FIG. 4C). The 3×Tg mice administered with anti-CD9Ab showed significantly higher freezing time even in the cued recall paradigm than the family administered with placebo (Fig. 4D, # p<0.05).
마우스는 Morris 물 미로를 사용하여 기준 공간 기억 기능에서 검사를 받았다. 이 연구에서 우리는 눈에 보이는 플랫폼 교육을 수행한 다음 하루에 네 번의 프로브 시도로 숨겨진 플랫폼 테스트를 수행했다. 각 그룹의 모든 마우스는 4 일 이내에 훈련 기준에 도달할 수 있었고 유사하게 숙련된 수영능력을 나타냈다. 그러나, 탐침 시험에서 anti-CD9Ab으로 치료한 3xTg 마우스는 대조군과 비교할 때 표적 영역에 대한 도달 수 및 표적 영역에서 소비한 시간의 유의한 증가를 보였다 (도 4E, 4F, #p<0.05). Mice were tested at baseline spatial memory function using the Morris water maze. In this study, we performed a visible platform training, followed by a hidden platform test with four probe attempts per day. All mice in each group were able to reach the training criterion within 4 days and similarly displayed skilled swimming ability. However, in the probe test, 3xTg mice treated with anti-CD9Ab showed a significant increase in the number of arrivals to the target area and time spent in the target area when compared to the control group (Fig. 4E, 4F, # p<0.05).
CD9의 발현 또는 활성을 저해하는 물질은 퇴행성 신경질환 환자의 기억력을 개선하는 효과를 발휘하여 퇴행성 신경질환 치료제 개발에 유용하게 활용이 될 수 있으며, CD9의 발현 수준 분석을 통해서 퇴행성 신경질환 치료제 후보물질의 스크리닝할 수 있어 산업상 이용가능성이 높다. Substances that inhibit the expression or activity of CD9 have the effect of improving the memory of patients with neurodegenerative diseases and can be usefully used in the development of treatments for neurodegenerative diseases, and candidates for treatment of neurodegenerative diseases through analysis of the expression level of CD9 It has high industrial applicability because it can screen.

Claims (16)

  1. CD9 단백질의 발현 또는 활성 억제제를 유효성분으로 포함하는 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물.A pharmaceutical composition for the prevention or treatment of neurodegenerative diseases comprising an inhibitor of CD9 protein expression or activity as an active ingredient.
  2. 제1항에 있어서, 상기 퇴행성 신경질환은 알츠하이머병, 치매, 경도인지장애, 파킨슨병, 진행성 핵상마비, 다계통 위축증, 감람핵-뇌교-소뇌 위축증(OPCA), 샤이-드래거 증후군, 선조체-흑질 퇴행증, 헌팅톤병, 근위축성 측색 경화증(ALS), 본태성 진전증, 피질-기저핵 퇴행증, 미만성 루이 소체 질환, 파킨스-ALS-치매 복합증, 니만픽 병(Niemann-Pick disease), 픽병, 뇌허혈 및 뇌경색으로 이루어진 군에서 선택되는 1종 이상인 것을 특징으로 하는 약학적 조성물. The method of claim 1, wherein the neurodegenerative disease is Alzheimer's disease, dementia, mild cognitive impairment, Parkinson's disease, progressive supranuclear palsy, multiple system atrophy, olive nucleus-poni-cerebellar atrophy (OPCA), Shy-Drager syndrome, striatum- Black matter degeneration, Huntington's disease, amyotrophic lateral sclerosis (ALS), essential tremors, cortical-basal ganglia degeneration, diffuse Lewy body disease, Parkin's-ALS-dementia complex, Niemann-Pick disease, Pick's disease , A pharmaceutical composition, characterized in that at least one selected from the group consisting of cerebral ischemia and cerebral infarction.
  3. 제1항에 있어서, 상기 CD9 단백질은 서열번호 1 또는 서열번호 2로 표시되는 아미노선 서열을 포함하는 것을 특징으로 하는 조성물.The composition of claim 1, wherein the CD9 protein comprises an amino line sequence represented by SEQ ID NO: 1 or SEQ ID NO: 2.
  4. 제1항에 있어서, 상기 CD9 단백질 발현 억제제는 CD9 mRNA에 상보적으로 결합하는 안티센스 올리고뉴클레오티드(antisense oligonucleotide), siRNA, shRNA, miRNA, ribozyme, crispr-cas9, DNAzyme 및 PNA(protein nucleic acid)로 이루어진 군에서 선택된 어느 하나인 것을 특징으로 하는 조성물.The method of claim 1, wherein the CD9 protein expression inhibitor is composed of an antisense oligonucleotide complementary to CD9 mRNA, siRNA, shRNA, miRNA, ribozyme, crispr-cas9, DNAzyme, and protein nucleic acid (PNA). Composition, characterized in that any one selected from the group.
  5. 제4항에 있어서, 상기 CD9 mRNA는 서열번호 3 또는 서열번호 4로 표시되는 염기 서열을 포함하는 것을 특징으로 하는 조성물.The composition of claim 4, wherein the CD9 mRNA comprises a nucleotide sequence represented by SEQ ID NO: 3 or SEQ ID NO: 4.
  6. 제1항에 있어서, 상기 CD9 단백질 활성 억제제는 CD9 단백질에 특이적으로 결합하는 화합물, 펩티드, 펩티드 유사체(mimetics), 앱타머, 항체, 천연추출물 및 합성화합물로 이루어진 군에서 선택된 어느 하나인 것을 특징으로 하는 조성물.The method of claim 1, wherein the CD9 protein activity inhibitor is any one selected from the group consisting of compounds, peptides, peptide mimetics, aptamers, antibodies, natural extracts, and synthetic compounds that specifically bind to CD9 protein. Composition made by.
  7. 제1항에 있어서, 상기 CD9 단백질 활성 억제제는 CD9 단백질에 특이적으로 결합하는 항체인 것을 특징으로 하는 조성물.The composition of claim 1, wherein the inhibitor of CD9 protein activity is an antibody that specifically binds to a CD9 protein.
  8. (a) CD9 단백질 또는 mRNA를 발현하는 세포에 시험물질을 처리하는 단계;(a) treating cells expressing CD9 protein or mRNA with a test substance;
    (b) 상기 세포와 시험물질을 처리하지 않은 대조군 세포에서 CD9 단백질 또는 mRNA의 발현 수준을 측정하는 단계; 및(b) measuring the expression level of CD9 protein or mRNA in the cells and control cells not treated with the test substance; And
    (c) 대조군 세포와 비교하여 CD9 단백질 또는 mRNA의 발현 수준을 감소시키는 시험 물질을 퇴행성 신경질환 치료제 후보물질로 선별하는 단계를 포함하는 퇴행성 신경질환 치료제 스크리닝 방법.(c) A method for screening a therapeutic agent for neurodegenerative diseases comprising the step of selecting a test substance that decreases the expression level of CD9 protein or mRNA compared to a control cell as a candidate substance for treating a neurodegenerative disease.
  9. 제8항에 있어서, 상기 CD9의 mRNA 발현 수준은 RT-PCR, 정량적 또는 반정량적 RT-PCR(Quantitative 또는 semi-Quantitative RT-PCR), 정량적 또는 반정량적 리얼 타임 RT-PCR(Quantitative 또는 semi-Quantitative real-time RT-PCR), 노던 블롯(northern blot), DNA 칩(chip) 및 RNA 칩으로 이루어지는 군에서 선택된 어느 하나의 방법을 이용하여 측정되는 것을 특징으로 하는 스크리닝 방법.The method of claim 8, wherein the mRNA expression level of CD9 is RT-PCR, quantitative or semi-quantitative RT-PCR (Quantitative or semi-Quantitative RT-PCR), quantitative or semi-quantitative real-time RT-PCR (Quantitative or semi-Quantitative Real-time RT-PCR), Northern blot (northern blot), a screening method characterized in that it is measured using any one method selected from the group consisting of a DNA chip (chip) and an RNA chip.
  10. 제8항에 있어서, CD9의 단백질 발현 수준은 웨스턴 블롯, ELISA, 방사선면역분석법, 방사면역확산법, 오우크레로니(Ouchterlony) 면역확산법, 로케트 면역전기영동, 면역조직화학염색, 면역침전분석, 보체고정분석, FACS 및 단백질 칩으로 이루어진 군에서 선택된 어느 하나의 방법을 이용하여 측정되는 것을 특징으로 하는 스크리닝 방법.The method of claim 8, wherein the protein expression level of CD9 is Western blot, ELISA, radioimmunoassay, radioimmuno diffusion method, Ouchterlony immunodiffusion method, rocket immunoelectrophoresis, immunohistochemical staining, immunoprecipitation analysis, and fixation of complement. A screening method, characterized in that it is measured using any one method selected from the group consisting of analysis, FACS, and protein chips.
  11. 퇴행성 신경질환의 예방 또는 치료용 제제를 제조하기 위한 CD9 단백질의 발현 또는 활성 억제제의 용도.Use of an inhibitor of expression or activity of a CD9 protein for preparing a preparation for the prevention or treatment of neurodegenerative diseases.
  12. 제11항에 있어서, 상기 퇴행성 신경질환은 알츠하이머병, 치매, 경도인지장애, 파킨슨병, 진행성 핵상마비, 다계통 위축증, 감람핵-뇌교-소뇌 위축증(OPCA), 샤이-드래거 증후군, 선조체-흑질 퇴행증, 헌팅톤병, 근위축성 측색 경화증(ALS), 본태성 진전증, 피질-기저핵 퇴행증, 미만성 루이 소체 질환, 파킨스-ALS-치매 복합증, 니만픽 병(Niemann-Pick disease), 픽병, 뇌허혈 및 뇌경색으로 이루어진 군에서 선택되는 1종 이상인 것을 특징으로 하는 용도.The method of claim 11, wherein the neurodegenerative disease is Alzheimer's disease, dementia, mild cognitive impairment, Parkinson's disease, progressive supranuclear palsy, multiple system atrophy, olive nucleus-poni-cerebellar atrophy (OPCA), Shy-Drager syndrome, striatum- Black matter degeneration, Huntington's disease, amyotrophic lateral sclerosis (ALS), essential tremors, cortical-basal ganglia degeneration, diffuse Lewy body disease, Parkin's-ALS-dementia complex, Niemann-Pick disease, Pick's disease , Use, characterized in that at least one selected from the group consisting of cerebral ischemia and cerebral infarction.
  13. 제11항에 있어서, 상기 CD9 단백질 활성 억제제는 CD9 단백질에 특이적으로 결합하는 항체인 것을 특징으로 하는 용도.The use according to claim 11, wherein the inhibitor of CD9 protein activity is an antibody that specifically binds to the CD9 protein.
  14. CD9 단백질의 발현 또는 활성 억제제를 유효성분으로 포함하는 조성물의 유효량을 이를 필요로 하는 개체에 투여하는 단계를 포함하는 퇴행성 신경질환의 치료 방법.A method for treating neurodegenerative diseases comprising administering to an individual in need thereof an effective amount of a composition comprising an inhibitor of CD9 protein expression or activity as an active ingredient.
  15. 제14항에 있어서, 상기 퇴행성 신경질환은 알츠하이머병, 치매, 경도인지장애, 파킨슨병, 진행성 핵상마비, 다계통 위축증, 감람핵-뇌교-소뇌 위축증(OPCA), 샤이-드래거 증후군, 선조체-흑질 퇴행증, 헌팅톤병, 근위축성 측색 경화증(ALS), 본태성 진전증, 피질-기저핵 퇴행증, 미만성 루이 소체 질환, 파킨스-ALS-치매 복합증, 니만픽 병(Niemann-Pick disease), 픽병, 뇌허혈 및 뇌경색으로 이루어진 군에서 선택되는 1종 이상인 것을 특징으로 하는 치료 방법.The method of claim 14, wherein the neurodegenerative disease is Alzheimer's disease, dementia, mild cognitive impairment, Parkinson's disease, progressive supranuclear palsy, multiple system atrophy, olive nucleus-poni-cerebellar atrophy (OPCA), Shy-Drager syndrome, striatum- Black matter degeneration, Huntington's disease, amyotrophic lateral sclerosis (ALS), essential tremors, cortical-basal ganglia degeneration, diffuse Lewy body disease, Parkin's-ALS-dementia complex, Niemann-Pick disease, Pick's disease , Treatment method, characterized in that at least one selected from the group consisting of cerebral ischemia and cerebral infarction.
  16. 제14항에 있어서, 상기 CD9 단백질 활성 억제제는 CD9 단백질에 특이적으로 결합하는 항체인 것을 특징으로 하는 치료 방법.The method of claim 14, wherein the inhibitor of CD9 protein activity is an antibody that specifically binds to the CD9 protein.
PCT/KR2020/005155 2019-04-18 2020-04-17 Composition for prevention or treatment of neurodegenerative diseases using cd9 and method for screening therapeutic agent for neurodegenerative diseases WO2020213982A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2019-0045489 2019-04-18
KR1020190045489A KR20200122601A (en) 2019-04-18 2019-04-18 Composition for preventing or treating neurodegenerative disease and method for screening agents using CD9

Publications (1)

Publication Number Publication Date
WO2020213982A1 true WO2020213982A1 (en) 2020-10-22

Family

ID=72837530

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2020/005155 WO2020213982A1 (en) 2019-04-18 2020-04-17 Composition for prevention or treatment of neurodegenerative diseases using cd9 and method for screening therapeutic agent for neurodegenerative diseases

Country Status (2)

Country Link
KR (1) KR20200122601A (en)
WO (1) WO2020213982A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070161584A1 (en) * 2003-12-26 2007-07-12 Kenichiro Kosai Drug for preventing or treating heart diseases comprising cd9 gene
KR101227971B1 (en) * 2008-06-25 2013-01-30 성균관대학교산학협력단 CD9―specific human antibody
WO2016021894A1 (en) * 2014-08-04 2016-02-11 영남대학교 산학협력단 Pharmaceutical composition for preventing or treating cellular senescence or senescence-associated diseases, containing cd9 antibody as active ingredient
KR20160068964A (en) * 2013-10-24 2016-06-15 나노소믹스 인코포레이티드 Biomarkers and diagnostic methods for alzheimer's disease and other neurodegenerative disorders

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070161584A1 (en) * 2003-12-26 2007-07-12 Kenichiro Kosai Drug for preventing or treating heart diseases comprising cd9 gene
KR101227971B1 (en) * 2008-06-25 2013-01-30 성균관대학교산학협력단 CD9―specific human antibody
KR20160068964A (en) * 2013-10-24 2016-06-15 나노소믹스 인코포레이티드 Biomarkers and diagnostic methods for alzheimer's disease and other neurodegenerative disorders
WO2016021894A1 (en) * 2014-08-04 2016-02-11 영남대학교 산학협력단 Pharmaceutical composition for preventing or treating cellular senescence or senescence-associated diseases, containing cd9 antibody as active ingredient

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
KIM, TAE WOO ET AL.: "Identification of replicative senescence-associated genes in human umbilical vein endothelial cells by an annealing control primer system", EXPERIMENTAL GERONTOLOGY, vol. 43, 2008, pages 286 - 295, XP022523981, DOI: 10.1016/j.exger.2007.12.010 *

Also Published As

Publication number Publication date
KR20200122601A (en) 2020-10-28

Similar Documents

Publication Publication Date Title
WO2018139819A1 (en) Uses for prevention or treatment of brain diseases using microrna
WO2014182051A1 (en) Composition comprising asm inhibitor as active ingredient for preventing or treating degenerative neurological disorders
US20170095496A1 (en) Galactoside inhibitors for new uses
KR20190110938A (en) Composition for preventing or treating neurodegenerative disorder comprising cyclooxygenase 2 acetylase
WO2019194586A1 (en) Novel target for anti-cancer and immune-enhancing
WO2020213982A1 (en) Composition for prevention or treatment of neurodegenerative diseases using cd9 and method for screening therapeutic agent for neurodegenerative diseases
WO2021177518A1 (en) Pharmaceutical composition for lowering blood cholesterol, preventing or treating cardiovascular diseases and reducing inflammation
WO2020159191A1 (en) Composition comprising mls-stat3 for prevention or treatment of immune disease
WO2019235876A1 (en) Pharmaceutical composition comprising expression or activity inhibitor of c-src for preventing or treating synucleinopathy
WO2009093864A2 (en) Composition for preventing or treating brain diseases
WO2010021516A9 (en) Novel use of lipocalin 2 for treating brain damage
WO2020017788A1 (en) Composition for preventing or treating diabetes by using cd9, and method for screening for antidiabetic agents
WO2018212503A1 (en) Cotl1 protein involved in maintaining homeostasis of hematopoietic stem cell, and use thereof
WO2021246755A1 (en) Composition, for preventing, alleviating or treating metabolic disease, comprising phospholipase d2 inhibitor
WO2022098118A1 (en) Composition for regulating osteoclast differentiation, and use thereof
WO2019066519A1 (en) Pharmaceutical composition for prevention or treatment of age-related macular degeneration containing rna complex targeting connective tissue growth factor
WO2022005214A1 (en) Screening kit for therapeutic agent for coronavirus disease using esterase activity of spike protein of covid-19 virus, and use of substance screened by using kit for prevention, alleviation, or treatment of coronavirus disease
KR101341200B1 (en) Pharmaceutical composition for treating or preventing metabolic syndrome comprising TRAP80 regulator
WO2017023001A1 (en) Non-alcoholic fatty liver regulator 14-3-3 protein
WO2017023002A1 (en) Non-alcoholic fatty liver regulating factor 14-3-3 protein
WO2020197304A1 (en) Composition for preventing, ameliorating, or treating pancreatic cancer, including tetraspanin-2 inhibitor as active ingredient
WO2020197367A1 (en) Composition for preventing or treating diabetes by using st8sia1, and method for screening for antidiabetic agents
WO2015111971A1 (en) Pharmaceutical composition containing gpr119 ligand as active ingredient for preventing or treating non-alcoholic fatty liver disease
WO2009136752A9 (en) Novel peptide for augmenting brain-derived neutrotrophic factor (bdnf) protein expression in hippocampal neurons, hippocampal tissue and cerebral-cortex tissue
WO2016159695A1 (en) Composition for preventing or treating autoimmune disease, containing ssu72 as active ingredient

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: 20791147

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: 20791147

Country of ref document: EP

Kind code of ref document: A1