WO2015108372A1 - Composition for preventing or treating neurological disorders caused by excitotoxicity or synaptic dysfunction, containing osmotin, and method for preventing or treating neurological disorders by using same - Google Patents

Composition for preventing or treating neurological disorders caused by excitotoxicity or synaptic dysfunction, containing osmotin, and method for preventing or treating neurological disorders by using same Download PDF

Info

Publication number
WO2015108372A1
WO2015108372A1 PCT/KR2015/000493 KR2015000493W WO2015108372A1 WO 2015108372 A1 WO2015108372 A1 WO 2015108372A1 KR 2015000493 W KR2015000493 W KR 2015000493W WO 2015108372 A1 WO2015108372 A1 WO 2015108372A1
Authority
WO
WIPO (PCT)
Prior art keywords
osmotin
glutamate
expression
treatment
present
Prior art date
Application number
PCT/KR2015/000493
Other languages
French (fr)
Korean (ko)
Inventor
김명옥
박현영
Original Assignee
경상대학교산학협력단
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 경상대학교산학협력단 filed Critical 경상대학교산학협력단
Publication of WO2015108372A1 publication Critical patent/WO2015108372A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/168Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from plants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • 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

Definitions

  • the present invention relates to a composition for preventing, ameliorating or treating neurological diseases caused by excitatory toxicity or synaptic dysfunction, and a method for preventing or treating neurological diseases using the same.
  • the present invention relates to a pharmaceutical composition or a food composition for preventing, ameliorating or treating a neurological disease caused by excitatory toxicity or synaptic dysfunction, including osmotin as an active ingredient, and using the composition, excitatory toxicity or synapse A method for preventing or treating a neurological disorder caused by a dysfunction.
  • Excitotoxicity refers to a pathological process in which nerve cells are damaged or killed by nerve cells being overstimulated by neurotransmitters such as glutamate or the like. Excitatory toxicity-mediated damage or death of nerve cells has been reported in various central nervous system diseases including ischemia and neurodegenerative diseases.
  • Osmotin is a multifunctional plant protein derived from Nicotiana tabacum and the like and has a molecular weight of about 24 kDa.
  • Osmotin is a protein belonging to the family of pathogenesis related-5 (PR-5) that provides plants with osmotolerance and exhibits antimicrobial activity.
  • PR-5 pathogenesis related-5
  • Osmotin is believed to be a structural and functional homologue with the mammalian hormone adiponectin and can act biologically as an adiponectin counterpart.
  • Korean Patent No. 10-1308232 discloses a composition for treating neurodegenerative diseases by alcohol containing osmotin, but the registered patent discloses an alcohol of osmotin. Efficacy of the neurodegenerative diseases induced by the present invention was only identified, and no effect was suggested for the neurological diseases caused by excitatory toxicity or synaptic dysfunction.
  • the present inventors have diligently tried to determine the effect of osmotin in neurological diseases caused by excitatory toxicity or synaptic dysfunction.
  • the osmotin is an AMPA ( ⁇ -Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) glutamate receptor.
  • AMPA ⁇ -Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
  • An object of the present invention is to provide a pharmaceutical composition for the prevention or treatment of neurological diseases caused by excitatory toxicity or synaptic dysfunction comprising osmotin as an active ingredient.
  • Another object of the present invention to provide a food composition for the prevention or improvement of neurological diseases caused by excitatory toxicity or synaptic dysfunction comprising osmotin as an active ingredient.
  • Still another object of the present invention is to provide a method for preventing or treating neurological diseases caused by excitatory toxicity or synaptic dysfunction using the composition.
  • composition containing the osmotin of the present invention as an active ingredient may be usefully used as a substance for preventing or treating excitatory toxicity or synaptic dysfunction induced by excessive stimulation of a neurotransmitter, and the neurological diseases caused thereby.
  • the effect of inhibiting the expression of the AMPA glutamate receptor is excellent, particularly the effect of preventing or treating excitatory or synaptic dysfunction induced by glutamate, and the neurological diseases caused thereby.
  • FIG. 1A and 1B show changes in expression of AMPA glutamine receptor (AMPAR), p-AMPAR and CaMKII in the cortex and hippocampus of osmotin treatment.
  • AMPAR AMPA glutamine receptor
  • p-AMPAR p-AMPAR
  • CaMKII CaMKII
  • 1C and 1D show changes in expression of p-CREB and synaptopycin in the cortex and hippocampus of the brain following osmotin treatment.
  • 2A and 2B show changes in expression of p53, Bax and Bcl-2 in the cortex and hippocampus of the brain following osmotin treatment.
  • 2C and 2D show changes in the expression of cytochrome-C, caspase-3 and PARP-1 in the cortex and hippocampus of the brain following osmotin treatment.
  • 3A and 3B show changes in the degree of DNA fragmentation in three regions of the hippocampus (CA1, CA3 and DG) following osmotin treatment.
  • 4A and 4B show changes in the expression of p-JNK, p-PI3K and p-Akt in cortex and hippocampus following osmotin treatment.
  • 4C and 4C show changes in expression of p-JNK and p-PI3K in CA1 region of hippocampus following osmotin treatment.
  • 5A and 5B show changes in distribution and colocation of p53 and p-Akt in CA1 region of hippocampus following osmotin treatment.
  • 5C and 5D show changes in the distribution and colocation of p53 and caspase-3 in the CA1 region of the hippocampus following osmotin treatment.
  • FIG. 6 is a simplified schematic of the mechanism of action of osmotin on biomechanisms induced by glutamate.
  • C is a control group
  • Glu is a glutamate treatment group
  • Os is an osmotin treatment group
  • Glu + Os means a glutamate and osmotin treatment group
  • the present invention provides a pharmaceutical composition for the prevention or treatment of neurological diseases caused by excitatory toxicity or synaptic dysfunction comprising osmotin as an active ingredient as one embodiment.
  • osmotin is a multifunctional plant protein derived from Nicotiana tabacum and the like, and is composed of about 150 to 250 amino acids depending on the individual, and has a molecular weight of about 24 kDa. It is known. Osmotin is a member of the PR-5 (Pathogenesis related-5) family that provides plants with osmotolerance and exhibits antimicrobial activity. It is believed that it can act as an adiponectin counterpart.
  • Korean Patent No. 10-1308232 discloses that osmotin exhibits an effect of inhibiting the death of alcohol-induced nerve cells, and by using the neurodegenerative disease caused by alcohol A therapeutic composition is provided.
  • the registered patent is limited to alcohol-induced neurodegenerative diseases, and there is no disclosure about neurological diseases induced by excitatory toxicity or synaptic dysfunction among neurological diseases, and the efficacy of osmotin on such neurological diseases. There was no suggestion about.
  • osmotin has an effect of treating neurological diseases caused by excitatory toxicity or synaptic dysfunction, and particularly, has a turbulent effect in the treatment of excitatory toxicity or synaptic dysfunction caused by glutamate overstimulation. It was confirmed.
  • Neurodegenerative diseases caused by alcohol are neurological diseases in which neuronal cell death is induced by alcohol.
  • the osmotin inhibits neuronal cell death, but the excitatory toxicity or synaptic function of the present invention.
  • Neurological diseases induced by the disorder are induced by excessive stimulation by neurotransmitters (particularly, glutamate).
  • the first clarification of glutathione receptor expression inhibition of osmotin and the like is performed. There is a difference in that it was first disclosed the therapeutic use of neurological diseases caused by excessive stimulation.
  • the osmotin can be used without limitation in its origin or type.
  • the osmotin may be separated from various plants or artificially synthesized according to genetic engineering methods, or may be purchased from domestic and foreign companies.
  • the neurological disease caused by the excitatory toxicity or synaptic dysfunction may be included without limitation to the type or condition of the disease, as long as it is induced by the excitatory toxicity or synaptic dysfunction.
  • the neurological disorder may be a mental neurological disorder, and examples of the mental neurological disorder include, but are not limited to, depression, schizophrenia, OCD, anxiety neurosis, and the like.
  • the neurological diseases include neurological diseases induced by nerve damage caused by excitotoxicity or synaptic dysfunction.
  • prevention means any action that inhibits or delays neurological symptoms caused by excitatory or synaptic dysfunction by administering the pharmaceutical composition of the present invention to a subject.
  • treatment refers to any action by which the pharmaceutical composition of the present invention is administered to a subject to improve or benefit neurological symptoms caused by excitatory toxicity or synaptic dysfunction.
  • the prevention or treatment of neurological diseases caused by the excitatory toxicity or synaptic dysfunction using the pharmaceutical composition comprising the osmotin of the present invention as an active ingredient may be performed by reducing expression of AMPA glutamate receptor or decreasing phosphorylation of AMPA glutamate receptor. Can be.
  • the term “decreased expression” or “increased expression” means that the expression of the protein is reduced or increased at the level of transcription or the activity of the protein as compared to the condition prior to treatment of the composition comprising the osmotin of the invention. It means all cases that are reduced or increased.
  • Said “reduced expression” or “increased expression” is for the purpose of the present invention, more preferably the expression of the protein of interest increased or decreased compared to the normal state induces excitatory or synaptic dysfunction, administration of the composition of the present invention It means all cases where the expression of the protein is reduced or increased than the condition before the administration of the composition of the present invention.
  • AMPA ⁇ -Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid glutamate receptor
  • Prevention or treatment of neurological diseases caused by the excitatory toxicity or synaptic dysfunction using the pharmaceutical composition comprising the osmotin of the present invention as an active ingredient, may be performed by increasing the expression of synaptopycin.
  • synaptophysin is a glycoprotein of about 30 kDa present in the synaptic vesicle of nerve cells or secretory cells in the nerve, also known as synaptic vesicle protein p38. Mean protein.
  • the expression of synaptopycin was significantly reduced when glutamate was treated in the rat at 7 days of age, but the osmotin treatment of the present invention after the glutamate treatment resulted in the reduction of synaptopycin. It was confirmed that the expression level is increased (Experimental Example 2).
  • the prevention or treatment of neurological diseases caused by the excitatory toxicity or synaptic dysfunction using the pharmaceutical composition comprising the osmotin of the present invention as an active ingredient may be performed by reducing the expression of CaMKII.
  • CaMKII (Ca 2+ / calmodulin-depandent protein kinase II) is a kind of serine / threonine-specific protein kinase, and means a kinase regulated by a Ca 2+ / calmodulin complex.
  • the expression of CaMKII was significantly increased when glutamate was treated in the rat at 7 days of age, but as a result of treating the osmotin of the present invention after the glutamate treatment, the expression level of CaMKII was increased. It was confirmed that the decrease by (experimental example 1).
  • JNK / PI3K / Akt pathway is a kind of intracellular signal transduction pathway, and a factor involved in the pathway "JNK” means "c-Jun N-terminal kinase” and " PI3K “means” Phosphoinositide 3-kinase "and” Akt “means” Protein kinase B (PKB) ".
  • p-JNK expression was increased and p-PI3K and p-Akt expression was reduced when glutamate was treated in rats of 7 days of age, but the osmotin of the present invention after the glutamate treatment As a result, it was confirmed that the increased expression level of p-JNK was significantly decreased, and the expression levels of p-PI3K and p-Akt were decreased significantly (Experimental Example 4).
  • the pharmaceutical composition comprising the osmotin of the present invention as an active ingredient also exhibits the effect of inhibiting apoptosis of nerve cells.
  • apoptosis is a form of a cell controlled by genes and kills itself, and is a concept distinguished from necrosis, which is necrosis or pathological death of cells. Also called.
  • the expression changes of p53, Bcl-2, Bax, which are known to be involved in apoptosis, the delivery of mitochondrial cytochromes to the cell substrate and the activation of caspase-3 and PARP-1 were observed. It was.
  • p53 used in the present invention is one of tumor suppressor factors, and is known to play a role in inhibiting abnormal proliferation of cells and inducing cancer cells to die.
  • Bcl-2 is a type of Bcl-2 family protein having a molecular weight of about 26 kDa and is known as an apoptosis inhibitor. Bcl-2 blocks apoptosis mediated by Fas antigen and TNF receptors, and overexpression of Bcl-2 inhibits apoptosis and is known to prolong cell life. Bcl-2 is also known to play a role in inhibiting the action of Bax and inhibiting the release of cytochrome C.
  • Bax is known as an apoptosis inducing factor and belongs to the Bcl-2 family because Bcl-2 and its gene sequence are similar.
  • Bax is activated, apoptosis is promoted and cell death is increased.
  • Bcl-2 is activated, apoptosis is suppressed and cell death is reduced.
  • treatment of glutamate in rats at 7 days of age increased the expression level of p53 protein and the Bax / Bcl-2 ratio, the delivery of mitochondrial cytochrome C and the caspase-3 and PARP-1.
  • Activation was promoted, but as a result of treating the osmotin of the present invention after the glutamate treatment, all of the above aspects were found to be reversed, and it was confirmed that the apoptosis of nerve cells was suppressed (Experimental Example 3).
  • the pharmaceutical composition of the present invention may further include a pharmaceutically acceptable carrier, in addition to including osmotin as an active ingredient.
  • the "pharmaceutically acceptable” means that it is commonly used in the pharmaceutical field that does not impede the biological activity and properties of the compound to be administered without stimulating the organism upon administration thereof.
  • the type of the carrier is not particularly limited and any carrier can be used as long as it is commonly used in the art.
  • Non-limiting examples of the carrier include saline, sterile water, Ringer's solution, buffered saline, albumin injection solution, lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, maltodextrin, glycerol, ethanol, and the like. Can be. These may be used alone or in combination of two or more thereof.
  • composition of the present invention may be used by adding other pharmaceutically acceptable additives, such as excipients, diluents, antioxidants, buffers or bacteriostatic agents, if necessary, fillers, extenders, wetting agents, disintegrants, dispersants, surfactants , Binders or lubricants may be additionally added and used.
  • additives such as excipients, diluents, antioxidants, buffers or bacteriostatic agents, if necessary, fillers, extenders, wetting agents, disintegrants, dispersants, surfactants , Binders or lubricants may be additionally added and used.
  • osmotin may be included in 0.00001% to 99.99% by weight based on the total weight of the pharmaceutical composition, preferably 1% to 99.99% by weight, more preferably 10 It may be included in the weight% to 99.99% by weight, more preferably 50% to 99.99% by weight, and may also be included in the total content of the pharmaceutical composition.
  • compositions of the present invention can be formulated and used in a variety of formulations suitable for oral or parenteral administration.
  • Non-limiting examples of the formulation for oral administration include troches, lozenges, tablets, aqueous suspensions, oily suspensions, prepared powders, granules, emulsions, hard capsules, soft capsules, syrups or elixirs, and the like. Can be mentioned.
  • a binder such as lactose, saccharose, sorbitol, mannitol, starch, amylopectin, cellulose or gelatin and the like; Excipients such as dicalcium phosphate and the like; Disintegrants such as corn starch or sweet potato starch; Lubricants such as magnesium stearate, calcium stearate, sodium stearyl fumarate or polyethylene glycol wax and the like can be used, and sweeteners, fragrances, syrups and the like can also be used.
  • a liquid carrier such as fatty oil may be additionally used in addition to the above-mentioned materials.
  • Non-limiting examples of the parenteral preparations include injection liquids, suppositories, respiratory inhalation powders, spray aerosols, ointments, application powders, oils, creams, and the like.
  • a sterile aqueous solution In order to formulate the pharmaceutical composition for parenteral administration, a sterile aqueous solution, a non-aqueous solvent, a suspension, an emulsion, a freeze-dried preparation, an external preparation, and the like may be used.
  • a non-aqueous solvent and the suspension propylene glycol, polyethylene Glycols, vegetable oils such as olive oil, injectable esters such as ethyloleate and the like can be used.
  • the pharmaceutical composition of the present invention when the pharmaceutical composition of the present invention is formulated as an injection solution, the composition of the present invention is mixed with water with a stabilizer or buffer to prepare a solution or suspension, which may be used as an ampoule or vial. It may be formulated for unit administration.
  • a propellant or the like when the pharmaceutical composition of the present invention is formulated with an aerosol, a propellant or the like may be combined with the additive to disperse the dispersed concentrate or the wet powder.
  • composition of the present invention when formulated into an ointment, a cream, or the like, animal oil, vegetable oil, wax, paraffin, starch, trakant, cellulose derivative, polyethylene glycol, silicone, bentonite, silica, talc, zinc oxide It may be formulated using such as carrier.
  • a pharmaceutically effective amount, effective dosage of the pharmaceutical composition of the present invention may vary depending on the method of formulating the pharmaceutical composition, the mode of administration, the time of administration and / or the route of administration, and the likeness of the reaction to be achieved by the administration of the pharmaceutical composition.
  • the type and extent of the subject, the type, age, weight, general state of health, condition or extent of the disease, sex, diet, excretion, components of the drug or other composition used concurrently or simultaneously with the subject Various factors and similar factors well known in the medical arts can be varied, and one of ordinary skill in the art can easily determine and prescribe a dosage effective for the desired treatment.
  • Administration of the pharmaceutical composition of the present invention may be administered once a day, may be divided into several times.
  • the pharmaceutical compositions of the present invention may be administered as individual therapeutic agents or in combination with other therapeutic agents and may be administered sequentially or simultaneously with conventional therapeutic agents. Taking all of the above factors into consideration, it is important to administer an amount that can obtain the maximum effect in a minimum amount without side effects, and can be easily determined by those skilled in the art.
  • the pharmaceutical composition of the present invention for a more preferable effect, it may be administered in an amount of 0.01 mg / kg / day to 100 mg / kg / day, more preferably from 0.1 mg / kg / day to It may be administered in an amount of 70 mg / kg / day, more preferably 1 mg / kg / day to 50 mg / kg / day.
  • the administration may be administered one time, divided into several times, and the administration cycle may be appropriately selected.
  • the route of administration and mode of administration of the pharmaceutical composition of the present invention may be independent of each other, and are not particularly limited in the way, and may be any route of administration and mode of administration as long as the pharmaceutical composition can reach the desired site. Can follow.
  • the pharmaceutical composition may be administered by oral or parenteral administration.
  • parenteral administration method for example, intravenous administration, intraperitoneal administration, intramuscular administration, transdermal administration or subcutaneous administration may be used, and the method of applying, spraying or inhaling the composition to a diseased site may also be used. May be, but is not limited to these.
  • a food composition for the prevention or improvement of neurological diseases caused by excitatory toxicity or synaptic dysfunction comprising osmotin as an active ingredient.
  • improvement means any action that at least reduces the parameters associated with alleviation or treatment of a condition, such as the degree of symptoms.
  • the food composition of the present invention is not particularly limited and includes a health functional food composition.
  • the health functional food composition of the present invention When used as a food additive, the composition may be added as it is or used with other food or food ingredients, and may be appropriately used according to a conventional method.
  • the kind of the food is not particularly limited, and includes all foods in a general sense.
  • foods that can be added to the material include meat, sausages, bread, chocolate, candy, snacks, confectionery, pizza, ramen, dairy products, including other noodles, gums, ice cream, various soups, drinks, tea , A drink, an alcoholic beverage, and a vitamin complex.
  • the health functional food composition of the present invention is a beverage composition
  • it may contain various flavors or natural carbohydrates and the like as additional ingredients, as in the usual beverage.
  • natural carbohydrates include monosaccharides such as glucose and fructose; Disaccharides such as maltose and sucrose; Natural sweeteners such as dextrin, cyclodextrin; Synthetic sweeteners such as saccharin and aspartame; and the like.
  • the proportion of the additional components added above may be appropriately determined by the choice of those skilled in the art.
  • the health functional food composition of the present invention includes various nutrients, vitamins, electrolytes, flavors, coloring agents, pectic acid and salts thereof, alginic acid and salts thereof, organic acids, protective colloidal thickeners, pH adjusting agents, stabilizers, preservatives, glycerin , Alcohols, carbonation agents used in carbonated beverages, and the like.
  • the health functional food composition of the present invention may contain a flesh for preparing natural fruit juice, fruit drink or vegetable drink. These components can be used independently or can be used in combination of 2 or more. The proportion of such additives may also be appropriately selected by those skilled in the art.
  • a method for preventing or treating neurological diseases caused by excitotoxicity or synaptic dysfunction comprising administering to a subject the pharmaceutical composition comprising osmotin as an active ingredient.
  • a method for preventing or ameliorating neurological diseases caused by excitatory toxicity or synaptic dysfunction comprising administering to a subject the food composition comprising osmotin as an active ingredient.
  • mammals means all animals, including mammals, including rats, domestic animals, humans, and the like.
  • osmotin was purified from salt-adapted cultured Nicotiana tabacum cells.
  • the endotoxin content of osmotin was ⁇ 0.03 EU / mg protein.
  • Brain sections of pups treated with saline, glutamate or glutamate + osmotin in Example 1 were analyzed 12 hours after drug treatment.
  • Transcardial perfusion was performed by treatment with 1 ⁇ phosphate buffered saline (PBS) and 4% ice-cold treated paraformaldehyde.
  • the brain tissue was then post-fixed in 4% paraformaldehyde and transferred to 20% Sukurose until the brain tissue sank to the bottom of the tube.
  • the brain was frozen in OCT (AO, USA) and then the brain was cut into 14 ⁇ m sections in the coronal plane using a CM 3050S cryostat (Leica, Germany). The brain sections were thawed-fixed to probes on positively charged slides (Fisher, USA).
  • a pre-stained protein ladder (GangNam-STAIN TM , iNtRon Biotechnology, Inc. Republic of Korea) containing a wide range of molecular weights from 10 kDa to 245 kDa was operated in parallel. was used. In order to minimize membrane blocking and non-specificity, blocking was performed using 5% (w / v) skim milk.
  • Antibodies used in immunoblotting to detect proteins include rabbit-derived anti-actin, anti-Bcl-2, anti-Bax and anti-caspase-3; Chlorine-derived anti-cytochrome c and anti-p53; And mouse-derived polyclonal antibodies anti-PARP-1 and anti-JNK (Santa Cruz Biotechnology, Santa Cruz, CA, USA).
  • rabbit-derived anti-p-CREB (Ser133), anti-CaMKII, anti-p-Akt (Ser473), anti-p-PI3K (Y458 / Y199), anti-p-JNK (Thr183 / Tyr185), anti -AMPA, anti-p-AMPA (Ser845) and anti-synaptopycin (Cell Signaling Technology, Inc) were used.
  • Membrane-bound secondary antibodies were visualized using ECL detection reagents according to the manufacturer's instructions (Amersham Pharmacia Biotech, Uppsala, Sweden). The optical density of the band was then analyzed by densitometry using a computer-based Sigma Gel program version 1.0 (SPSS, Chicago, IL, USA).
  • the tissue-containing slides according to Example 2 were washed twice for 15 minutes in 0.01 M PBS and proteolytic enzyme K solution was added to the tissues and incubated at 37 ° C. for 5 minutes. The tissues were then incubated for 90 minutes in blocking solution containing 0.3% Triton X-100 and normal pig serum in PBS.
  • the primary antibodies (rabbit polyclonal p-Akt, p-PI3K and caspase-3 (Cell Signaling Technology, Inc), and goat polyclonal IgG p53 and p-JNK (Santa cruz); All present at 1: 100 in PBS) overnight at 4 ° C.
  • TUNEL staining was performed according to the recommendations of In Situ Cell Death Detection Kit Fluorescein (GenScript, NJ, USA). Glass cover slides were encapsulated on glass slides with an encapsulant and FITC filters were used for TUNEL staining (green color) detection. TUNEL-positive (green) staining patterns were obtained with a confocal laser scanning microscope (FluoView FV 1000, Olympus, Japan), and TUNEL-positive cells in different regions of each section were counted with a computer based program.
  • the band of Western blot was scanned and the optical density of the band was analyzed by densitometer using Sigma Gel System (SPSS Inc., Chicago, IL). Density values are expressed as mean ⁇ SEM. One-way analysis (ANOVA) of variables was performed to identify significant differences, followed by Student's t-test to identify the significance of differences among the treatment groups. P values less than 0.05 were considered significant (p ⁇ 0.05).
  • AMPA glutamate receptors are ligand-gated ion channels consisting of a combination of four different subunits, GluR1-4, wherein overexpression of glutamate receptors causes excitatory toxicity and may result in neuronal damage.
  • the rat cortex and hippocampus were observed 4 hours after the first subcutaneous injection of glutamate in rats after 7 days of birth, whereas the AMPA glutamate receptor protein (Glu2 / 3/4) was overexpressed, whereas the glutamate treatment was performed. Later, it was observed that protein expression levels of AMPA receptors were significantly reduced in the animal group treated with osmotin (FIGS. 1A and 1B).
  • Calmodulin-dependent kinase-II (CaMKII) is an important regulatory factor in the biochemical mechanism leading to neuronal cell death due to acute excitatory toxicity and excitatory toxicity when antagonists inhibit the action of CaMKII. By the death of nerve cells is reduced.
  • osmotin is effective in protecting the nerve against acute excitatory toxicity induced by glutamate (FIGS. 1A and 1B).
  • synaptophysin In order to analyze the inhibitory effect of glutamate and the beneficial effect of osmotin on synaptic plasticity, the expression level of synaptophysin was measured using Western blot. Specifically, 4 hours after the first subcutaneous injection of glutamate in rats after 7 days of birth, the brain cortex and hippocampus of the rats were observed, whereas synaptopycin was significantly reduced, but after the glutamate treatment. Osmotin-treated animals were found to have increased expression levels of synaptophycins (FIGS. 1C and 1D). This means that osmotin is effective in repairing synaptic dysfunction induced by glutamate in the cortex and hippocampus of the brain.
  • Excitotoxic damage induced by glutamate can lead to a wide range of p53-mediated neuronal damage. Therefore, the levels of different cellular apoptosis markers were measured to determine whether neuronal damage was induced 4 hours after glutamate (10 mg / kg) in rats 7 days after birth.
  • glutamate is expressed in the level of proapoptotic p53, the ratio of proapoptotic Bax and antiapoptotic Bcl-2, the delivery of mitochondrial cytochrome to cell substrate and caspa in both cortex and hippocampus of brain. Toxicity was observed to be induced by increasing the activation of aze-3 and PARP-1.
  • DNA damage is one of the typical features of cellular apoptosis.
  • TUNEL staining DNA cleavage analysis based on enzyme labeling at the free 3 'DNA end was performed to visualize damaged DNA.
  • P-JNK (c-Jun N-) in the cerebral cortex and hippocampus of animals treated with glutamate alone and rats treated with osmotin after glutamate treatment in rats after 7 days of birth using Western blot Terminal kinase) expression levels were measured.
  • Phosphoinositide 3-kinase PI3-K / Akt pathway is one of the important pro-survival pathways in neurons. Therefore, it was examined whether the pathway was relevant when glutamate was treated in the cortex and hippocampus of the brain.
  • p-Akt was found to co-locate with p53 mostly.
  • the treatment of glutamate alone inhibited the distribution of p-Akt (red) and increased the distribution of p53 (green) in the CA1 region of the hippocampus of the brain, whereas osmotin after glutamate treatment
  • the distribution of p-Akt and p53 induced by glutamate in the CA1 region of the hippocampus was reversed (Figs. 5A and 5B).

Abstract

The present invention relates to: a composition for preventing, alleviating or treating neurological disorders caused by excitotoxicity or synaptic dysfunction; and a method for preventing or treating neurological disorders by using the same. Specifically, the present invention relates to: a pharmaceutical composition or a food composition for preventing, alleviating or treating neurological disorders caused by excitotoxicity or synaptic dysfunction, containing osmotin as an active ingredient; and a method for preventing or treating neurological disorders caused by excitotoxicity or synaptic dysfunction, by using the composition.

Description

오스모틴을 포함하는 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환의 예방 또는 치료용 조성물, 및 이를 이용한 신경 질환의 예방 또는 치료 방법A composition for preventing or treating neurological diseases caused by excitatory toxicity or synaptic dysfunction, including osmotin, and a method for preventing or treating neurological diseases using the same
본 발명은 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환을 예방하거나 개선 또는 치료하기 위한 조성물 및 이를 이용한 신경 질환의 예방 또는 치료 방법에 관한 것이다.The present invention relates to a composition for preventing, ameliorating or treating neurological diseases caused by excitatory toxicity or synaptic dysfunction, and a method for preventing or treating neurological diseases using the same.
구체적으로, 본 발명은 오스모틴을 유효 성분으로 포함하는, 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환의 예방, 개선 또는 치료용 약학 조성물 또는 식품 조성물에 관한 것이며, 상기 조성물을 이용하여 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환을 예방 또는 치료하는 방법에 관한 것이다.Specifically, the present invention relates to a pharmaceutical composition or a food composition for preventing, ameliorating or treating a neurological disease caused by excitatory toxicity or synaptic dysfunction, including osmotin as an active ingredient, and using the composition, excitatory toxicity or synapse A method for preventing or treating a neurological disorder caused by a dysfunction.
흥분독성(excitotoxicity) 이란, 신경 세포가 글루타메이트 또는 이의 유사 물질과 같은 신경 전달 물질에 의해 과잉 자극을 받음으로써 신경 세포가 손상되거나 사멸하는 병리학적 과정을 말한다. 흥분독성으로 매개되는 신경 세포의 손상 또는 사멸은 허혈 및 신경 퇴행성 질환 등을 포함하는 다양한 중추 신경계 질환에서 보고되고 있다.Excitotoxicity refers to a pathological process in which nerve cells are damaged or killed by nerve cells being overstimulated by neurotransmitters such as glutamate or the like. Excitatory toxicity-mediated damage or death of nerve cells has been reported in various central nervous system diseases including ischemia and neurodegenerative diseases.
흥분독성에 관한 생체 외 및 생체 내에서 수행된 종래 연구들을 통하여, NMDA 또는 AMPA 글루타메이트 수용체의 과잉 자극이 신경 세포 아팝토시스를 유도하는 것으로 보고되고 있으며, 일부 신경 질환에서 글루타메이트 및 글루타메이트성 활성화 과잉이 나타나는 것으로 보고되고 있다. 상기 글루타메이트 수용체의 과잉 자극 중, NMDA 글루타메이트 수용체에 관한 종래 연구에서는 NMDA 글루타메이트 수용체의 지속적인 활성화에 의하여 세포 내 칼슘 증가, 분해 효소(catabolic enzyme)와 카스파아제(caspase)의 활성화, 및 활성 산소와 질소 자유 라디칼의 생성을 유발함으로써, 결과적으로 세포 아팝토시스를 일으킨다고 보고되고 있다.Previous studies conducted in vitro and in vivo on excitatory toxicity report that excess stimulation of NMDA or AMPA glutamate receptors induces neuronal apoptosis, and glutamate and glutamate activation excess in some neurological diseases It is reported to appear. During the excessive stimulation of glutamate receptors, conventional studies on NMDA glutamate receptors suggest that intracellular calcium increase, activation of catabolic enzymes and caspases, and free radicals and nitrogen free by the continuous activation of NMDA glutamate receptors. It is reported that by inducing the production of radicals, the resulting cell apoptosis.
그러나 글루타메이트 유도성 흥분독성의 정확한 메커니즘에 대하여는 아직 명확히 밝혀진 바는 없는 실정이다.However, the exact mechanism of glutamate-induced excitatory toxicity has not been elucidated yet.
한편, 오스모틴(Osmotin)은 토바코(Nicotiana tabacum) 등에서 유래된 다기능 식물 단백질로서 24 kDa 가량의 분자량을 갖는다. 오스모틴은 식물에 오스모틴 내성(osmotolerance)을 제공하고 항균 활성을 나타내는 PR-5 (pathogenesis related-5) 패밀리에 속하는 단백질이다. 오스모틴은 포유동물의 호르몬인 아디포넥틴과 구조적 및 기능적 상동체로서, 생물학적으로 아디포넥틴 대항체로서 작용할 수 있는 것으로 여겨지고 있다. 아디포넥틴의 효능과 관련하여 항-염증, 항-당뇨, 항-죽상경화 효능 등이 보고된 바 있으며, 이에 대한 오스모틴의 아디포넥틴 대항체로서의 효과로, 뮤린 대장염 동물 모델에서 아디포넥틴의 항-염증 효과를 감소시킨다는 것이 보고된 바 있다. 그 밖에 오스모틴과 관련하여서는 비만 및 당뇨병과 관련된 효과가 연구되었을 뿐, 오스모틴의 신경 질환과 관련된 효능에 대하여는 연구가 미비한 실정이다.Osmotin is a multifunctional plant protein derived from Nicotiana tabacum and the like and has a molecular weight of about 24 kDa. Osmotin is a protein belonging to the family of pathogenesis related-5 (PR-5) that provides plants with osmotolerance and exhibits antimicrobial activity. Osmotin is believed to be a structural and functional homologue with the mammalian hormone adiponectin and can act biologically as an adiponectin counterpart. The anti-inflammatory, anti-diabetic, and anti-atherogenic effects have been reported in relation to the efficacy of adiponectin, and the effects of osmotin as an adiponectin counterpart on the anti-inflammatory effect of adiponectin have been shown in murine colitis animal model Reduction has been reported. In addition, only the effects related to obesity and diabetes have been studied in relation to osmotin, and studies on the efficacy related to neurological diseases of osmotin are insufficient.
오스모틴의 신경 퇴행성 질환에 대한 효능과 관련하여, 대한민국 등록특허 제10-1308232호에서 오스모틴을 포함하는 알코올에 의한 신경 퇴행성 질환 치료용 조성물에 관하여 개시하고 있으나, 상기 등록특허는 오스모틴의 알코올에 의해 유도된 신경 퇴행성 질환에 대한 효능만을 규명한 것으로, 흥분독성이나 시냅스 기능 장애에 의한 신경 질환에 대하여는 어떠한 효능도 시사하는 바가 없었다.Regarding the efficacy of osmotin against neurodegenerative diseases, Korean Patent No. 10-1308232 discloses a composition for treating neurodegenerative diseases by alcohol containing osmotin, but the registered patent discloses an alcohol of osmotin. Efficacy of the neurodegenerative diseases induced by the present invention was only identified, and no effect was suggested for the neurological diseases caused by excitatory toxicity or synaptic dysfunction.
본 발명자들은 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환에 있어서 오스모틴의 효과를 규명하고자 예의 노력한 결과, 오스모틴이 AMPA (α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) 글루타메이트 수용체의 발현을 감소시키는 등의 작용을 하여, 글루타메이트에 의한 과잉 자극에 따른 흥분독성 또는 시냅스 기능 장애를 예방 또는 치료하는 효과가 있음을 확인하고, 이를 활용한 본 발명을 완성하기에 이르렀다.The present inventors have diligently tried to determine the effect of osmotin in neurological diseases caused by excitatory toxicity or synaptic dysfunction. As a result, the osmotin is an AMPA (α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) glutamate receptor. By reducing the expression of, such as, it was confirmed that there is an effect of preventing or treating excitatory toxicity or synaptic dysfunction due to excessive stimulation by glutamate, and came to complete the present invention utilizing the same.
본 발명은 오스모틴을 유효 성분으로 포함하는 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환의 예방 또는 치료용 약학 조성물을 제공하는 것을 목적으로 한다.An object of the present invention is to provide a pharmaceutical composition for the prevention or treatment of neurological diseases caused by excitatory toxicity or synaptic dysfunction comprising osmotin as an active ingredient.
본 발명의 다른 목적은 오스모틴을 유효 성분으로 포함하는 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환의 예방 또는 개선용 식품 조성물을 제공하는 것이다.Another object of the present invention to provide a food composition for the prevention or improvement of neurological diseases caused by excitatory toxicity or synaptic dysfunction comprising osmotin as an active ingredient.
본 발명의 또 다른 목적은 상기 조성물을 이용하여 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환의 예방 또는 치료하는 방법을 제공하는 것이다.Still another object of the present invention is to provide a method for preventing or treating neurological diseases caused by excitatory toxicity or synaptic dysfunction using the composition.
본 발명의 오스모틴을 유효 성분으로 포함하는 조성물은 신경 전달 물질의 과잉 자극에 의해 유도되는 흥분독성 또는 시냅스 기능 장애, 및 이로 인해 유발되는 신경 질환을 예방 또는 치료하는 물질로 유용하게 이용될 수 있다. 또한, AMPA 글루타메이트 수용체의 발현을 저해하는 효능이 우수하여, 특히 글루타메이트에 의해 유도되는 흥분독성 또는 시냅스 기능 장애, 및 이로 인해 유발되는 신경 질환을 예방 또는 치료하는 효과가 우수하다.The composition containing the osmotin of the present invention as an active ingredient may be usefully used as a substance for preventing or treating excitatory toxicity or synaptic dysfunction induced by excessive stimulation of a neurotransmitter, and the neurological diseases caused thereby. . In addition, the effect of inhibiting the expression of the AMPA glutamate receptor is excellent, particularly the effect of preventing or treating excitatory or synaptic dysfunction induced by glutamate, and the neurological diseases caused thereby.
도 1a 및 1b는 오스모틴 처리에 따른, 뇌의 피질 및 해마에서의 AMPA 글루타민 수용체(AMPAR), p-AMPAR 및 CaMKⅡ의 발현 변화를 관찰한 것이다.1A and 1B show changes in expression of AMPA glutamine receptor (AMPAR), p-AMPAR and CaMKII in the cortex and hippocampus of osmotin treatment.
도 1c 및 1d는 오스모틴 처리에 따른, 뇌의 피질 및 해마에서의 p-CREB 및 시냅토파이신의 발현 변화를 관찰한 것이다.1C and 1D show changes in expression of p-CREB and synaptopycin in the cortex and hippocampus of the brain following osmotin treatment.
도 2a 및 2b는 오스모틴 처리에 따른, 뇌의 피질 및 해마에서의 p53, Bax 및 Bcl-2의 발현 변화를 관찰한 것이다.2A and 2B show changes in expression of p53, Bax and Bcl-2 in the cortex and hippocampus of the brain following osmotin treatment.
도 2c 및 2d는 오스모틴 처리에 따른, 뇌의 피질 및 해마에서의 시토크롬-C, 카스파아제-3 및 PARP-1의 발현 변화를 관찰한 것이다.2C and 2D show changes in the expression of cytochrome-C, caspase-3 and PARP-1 in the cortex and hippocampus of the brain following osmotin treatment.
도 3a 및 3b는 오스모틴 처리에 따른, 해마의 세 영역(CA1, CA3 및 DG)에서의 DNA 단편화 정도의 변화를 관찰한 것이다.3A and 3B show changes in the degree of DNA fragmentation in three regions of the hippocampus (CA1, CA3 and DG) following osmotin treatment.
도 4a 및 4b는 오스모틴 처리에 따른, 뇌의 피질 및 해마에서의 p-JNK, p-PI3K 및 p-Akt의 발현 변화를 관찰한 것이다.4A and 4B show changes in the expression of p-JNK, p-PI3K and p-Akt in cortex and hippocampus following osmotin treatment.
도 4c 및 4c는 오스모틴 처리에 따른, 해마의 CA1 영역에서의 p-JNK 및 p-PI3K의 발현 변화를 관찰한 것이다.4C and 4C show changes in expression of p-JNK and p-PI3K in CA1 region of hippocampus following osmotin treatment.
도 5a 및 5b는 오스모틴 처리에 따른, 해마의 CA1 영역에서의 p53 및 p-Akt의 분포 변화 및 공동위치화를 관찰한 것이다.5A and 5B show changes in distribution and colocation of p53 and p-Akt in CA1 region of hippocampus following osmotin treatment.
도 5c 및 5d는 오스모틴 처리에 따른, 해마의 CA1 영역에서의 p53 및 카스파아제-3의 분포 변화 및 공동위치화를 관찰한 것이다.5C and 5D show changes in the distribution and colocation of p53 and caspase-3 in the CA1 region of the hippocampus following osmotin treatment.
도 6은 글루타메이트에 의해 유도되는 생체 기작에 대한 오스모틴의 작용 메카니즘을 간략히 도식화한 것이다.6 is a simplified schematic of the mechanism of action of osmotin on biomechanisms induced by glutamate.
도 1 내지 도 6에서 "C"는 대조군, "Glu"는 글루타메이트 처리군, "Os"는 오스모틴 처리군, "Glu+Os"는 글루타메이트 및 오스모틴 처리군을 의미한다.1 to 6, "C" is a control group, "Glu" is a glutamate treatment group, "Os" is an osmotin treatment group, and "Glu + Os" means a glutamate and osmotin treatment group.
상기 과제를 해결하기 위하여, 본 발명은 하나의 구현예로서 오스모틴을 유효 성분으로 포함하는 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환의 예방 또는 치료용 약학 조성물을 제공한다.In order to solve the above problems, the present invention provides a pharmaceutical composition for the prevention or treatment of neurological diseases caused by excitatory toxicity or synaptic dysfunction comprising osmotin as an active ingredient as one embodiment.
본 발명에서 사용되는 용어, "오스모틴(Osmotin)"은 토바코(Nicotiana tabacum) 등에서 유래된 다기능 식물 단백질로서, 개체에 따라 약 150개 내지 250개의 아미노산으로 이루어져 있으며, 약 24 kDa 가량의 분자량을 갖는 것으로 알려져 있다. 오스모틴은 식물에 오스모틴 내성(osmotolerance)을 제공하고 항균 활성을 나타내는 PR-5 (Pathogenesis related-5) 패밀리에 속하는 단백질로서, 포유동물의 호르몬인 아디포넥틴(Adiponectin)과 구조적 및 기능적 상동체로, 생물학적으로 아디포넥틴 대항체로서 작용할 수 있는 것으로 여겨지고 있다. 아디포넥틴의 효능과 관련하여 항-염증, 항-당뇨, 항-죽상경화 효능 등이 보고된 바 있으며, 이에 대한 오스모틴의 아디포넥틴 대항체로서의 효과로, 뮤린 대장염 동물 모델에서 아디포넥틴의 항-염증 효과를 감소시킨다는 것이 보고된 바 있다. 그 밖에 오스모틴은 비만이나 당뇨병, 암의 치료 효능이 있는 것으로 알려져 있다.As used herein, the term "osmotin" is a multifunctional plant protein derived from Nicotiana tabacum and the like, and is composed of about 150 to 250 amino acids depending on the individual, and has a molecular weight of about 24 kDa. It is known. Osmotin is a member of the PR-5 (Pathogenesis related-5) family that provides plants with osmotolerance and exhibits antimicrobial activity. It is believed that it can act as an adiponectin counterpart. The anti-inflammatory, anti-diabetic, and anti-atherogenic effects have been reported in relation to the efficacy of adiponectin, and the effects of osmotin as an adiponectin counterpart on the anti-inflammatory effect of adiponectin have been shown in murine colitis animal model Reduction has been reported. Osmotin is also known to be effective in treating obesity, diabetes and cancer.
오스모틴의 신경 질환과 관련된 선행 기술로, 대한민국 등록특허 제10-1308232호에서는 오스모틴이 알코올에 의해 유도되는 신경 세포의 사멸을 억제하는 효능을 나타냄을 규명하고 이를 이용하여 알코올에 의한 신경 퇴행성 질환 치료용 조성물을 제공하고 있다. 그러나 상기 등록특허는 알코올에 의해 유도되는 신경 퇴행성 질환에 국한된 것으로서, 신경 질환 중 흥분독성이나 시냅스 기능 장애에 의해 유도되는 신경 질환에 대하여는 어떠한 개시도 되어있지 않으며, 이러한 신경 질환에 대한 오스모틴의 효능에 대하여는 전혀 시사하는 바가 없었다.In the prior art related to the neuropathy of osmotin, Korean Patent No. 10-1308232 discloses that osmotin exhibits an effect of inhibiting the death of alcohol-induced nerve cells, and by using the neurodegenerative disease caused by alcohol A therapeutic composition is provided. However, the registered patent is limited to alcohol-induced neurodegenerative diseases, and there is no disclosure about neurological diseases induced by excitatory toxicity or synaptic dysfunction among neurological diseases, and the efficacy of osmotin on such neurological diseases. There was no suggestion about.
이에 본 발명자들은 오스모틴이 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환을 치료하는 효과가 있음을 최초로 규명하였으며, 특히 글루타메이트에 의한 과잉 자극에 따른 흥분독성 또는 시냅스 기능 장애의 치료에 탁원한 효과가 있음을 확인하였다.Accordingly, the present inventors first identified that osmotin has an effect of treating neurological diseases caused by excitatory toxicity or synaptic dysfunction, and particularly, has a turbulent effect in the treatment of excitatory toxicity or synaptic dysfunction caused by glutamate overstimulation. It was confirmed.
알코올에 의한 신경 퇴행성 질환은 알코올에 의해 신경 세포의 사멸이 유도되어 나타나는 신경 질환으로, 선행 특허에서는 오스모틴의 신경 세포 사멸 억제 효능에 관하여만 규명한 것인 반면, 본 발명의 흥분독성 또는 시냅스 기능 장애에 의해 유도되는 신경 질환은 신경 전달 물질(특히, 글루타메이트)에 의한 과잉 자극으로 인해 유도되는 것으로서, 본 발명에서는 오스모틴의 글루타메이트 수용체 발현 억제 효능 등을 최초로 규명하여, 신경 전달 물질에 따른 과잉 자극을 억제함으로써 과잉 자극에 따른 신경 질환의 치료 용도를 최초로 밝혔다는 점에서 차이가 있다.Neurodegenerative diseases caused by alcohol are neurological diseases in which neuronal cell death is induced by alcohol. In the prior patent, only the osmotin inhibits neuronal cell death, but the excitatory toxicity or synaptic function of the present invention. Neurological diseases induced by the disorder are induced by excessive stimulation by neurotransmitters (particularly, glutamate). In the present invention, the first clarification of glutathione receptor expression inhibition of osmotin and the like is performed. There is a difference in that it was first disclosed the therapeutic use of neurological diseases caused by excessive stimulation.
본 발명의 구체적인 실시예에서, 생후 7일의 Sqrague-Dawley 랫트에 글루타메이트를 단독 처리한 경우, AMPA 글루타메이트 수용체의 발현이 증가되고 시냅토파이신의 발현이 감소되며 CaMKⅡ의 발현이 증가되는 등 흥분독성 또는 시냅스 기능 장애 증상이 유도되었으나, 상기 글루타메이트 처리 후 오스모틴을 처리한 경우, 상기 증상들이 모두 반대 양상을 보여 오스모틴이 흥분독성 또는 시냅스 기능 장애를 예방 또는 치료하는 효능이 있음을 확인하였다(실험예 1 및 2).In a specific embodiment of the present invention, when treated with glutamate alone in Sqrague-Dawley rats at 7 days of age, the expression of AMPA glutamate receptors is increased, the expression of synaptopycin is decreased, and the expression of CaMKII is increased. Synaptic dysfunction symptoms were induced, but when osmotin was treated after glutamate treatment, all of the above symptoms were reversed, confirming that osmotin is effective in preventing or treating excitatory toxicity or synaptic dysfunction (Experimental Example). 1 and 2).
본 발명에서 상기 오스모틴은 그 기원이나 종류에 제한없이 사용 가능하다. 상기 오스모틴은 다양한 식물체로부터 분리하거나 유전 공학적 방법에 따라 인위적으로 합성하여 사용할 수 있으며, 국내외의 회사로부터 구입하여 사용할 수도 있다.In the present invention, the osmotin can be used without limitation in its origin or type. The osmotin may be separated from various plants or artificially synthesized according to genetic engineering methods, or may be purchased from domestic and foreign companies.
본 발명에서 상기 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환은 흥분독성 또는 시냅스 기능 장애에 의하여 유도되는 것이라면 병증의 종류나 질환명에 제한되지 않고 모두 포함될 수 있다.In the present invention, the neurological disease caused by the excitatory toxicity or synaptic dysfunction may be included without limitation to the type or condition of the disease, as long as it is induced by the excitatory toxicity or synaptic dysfunction.
예를 들어, 상기 신경 질환은 정신 신경 질환일 수 있으며, 상기 정신 신경 질환의 예로 우울증, 정신 분열증, 강박 신경증, 불안 신경증 등을 들 수 있으나, 이에 제한되지 않는다.For example, the neurological disorder may be a mental neurological disorder, and examples of the mental neurological disorder include, but are not limited to, depression, schizophrenia, OCD, anxiety neurosis, and the like.
그밖에, 상기 신경 질환은 흥분독성 또는 시냅스 기능 장애에 의한 신경 손상에 의해 유도되는 신경 질환들을 포함한다.In addition, the neurological diseases include neurological diseases induced by nerve damage caused by excitotoxicity or synaptic dysfunction.
본 발명에서 사용되는 용어, "예방"이란, 본 발명의 상기 약학 조성물을 개체에 투여하여 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환 증상을 억제시키거나 지연시키는 모든 행위를 의미한다.As used herein, the term "prevention" means any action that inhibits or delays neurological symptoms caused by excitatory or synaptic dysfunction by administering the pharmaceutical composition of the present invention to a subject.
본 발명에서 사용되는 용어, "치료"란, 본 발명의 상기 약학 조성물을 개체에 투여하여 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환 증세가 호전되도록 하거나 이롭게 되도록 하는 모든 행위를 의미한다.As used herein, the term "treatment" refers to any action by which the pharmaceutical composition of the present invention is administered to a subject to improve or benefit neurological symptoms caused by excitatory toxicity or synaptic dysfunction.
본 발명의 상기 오스모틴을 유효 성분으로 포함하는 약학 조성물을 이용한 상기 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환의 예방 또는 치료는, AMPA 글루타메이트 수용체의 발현 감소 또는 AMPA 글루타메이트 수용체의 인산화 감소에 의해 수행될 수 있다.The prevention or treatment of neurological diseases caused by the excitatory toxicity or synaptic dysfunction using the pharmaceutical composition comprising the osmotin of the present invention as an active ingredient may be performed by reducing expression of AMPA glutamate receptor or decreasing phosphorylation of AMPA glutamate receptor. Can be.
본 발명에서 사용되는 용어, "발현 감소" 또는 "발현 증가"는 본 발명의 오스모틴을 포함하는 조성물을 처리하기 이전의 상태에 비하여, 해당 단백질의 발현이 전사 수준에서 감소 또는 증가되거나 단백질의 활성이 감소 또는 증가되는 모든 경우를 의미한다. 상기 "발현 감소" 또는 "발현 증가"는 본 발명의 목적상, 보다 바람직하게는 흥분독성 또는 시냅스 기능 장애가 유도되어 정상 상태에 비해 증가 또는 감소된 해당 단백질의 발현이, 본 발명의 상기 조성물의 투여에 의해 정상 상태로 회복되거나, 또는 본 발명의 상기 조성물을 투여하기 이전의 상태보다 단백질의 발현이 감소 또는 증가되는 모든 경우를 의미한다.As used herein, the term “decreased expression” or “increased expression” means that the expression of the protein is reduced or increased at the level of transcription or the activity of the protein as compared to the condition prior to treatment of the composition comprising the osmotin of the invention. It means all cases that are reduced or increased. Said "reduced expression" or "increased expression" is for the purpose of the present invention, more preferably the expression of the protein of interest increased or decreased compared to the normal state induces excitatory or synaptic dysfunction, administration of the composition of the present invention It means all cases where the expression of the protein is reduced or increased than the condition before the administration of the composition of the present invention.
본 발명에서 사용되는 용어, "AMPA (α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) 글루타메이트 수용체"는 채널형 글루타메이트 수용체의 일종을 의미한다.As used herein, the term "AMPA (α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) glutamate receptor" refers to a type of channel glutamate receptor.
본 발명의 구체적인 실시예에서, 생후 7일의 랫트에 글루타메이트를 처리한 경우, AMPA 글루타메이트 수용체 및 p-AMPA 글루타메이트 수용체의 발현이 증가되는 것으로 확인되었으나, 상기 글루타메이트 처리 후 본 발명의 오스모틴을 처리한 경우, 상기 AMPA 글루타메이트 수용체 및 p-AMPA 글루타메이트 수용체의 발현이 유의적으로 감소되는 것으로 확인되었다(실험예 1).In a specific embodiment of the present invention, when treated with glutamate in rats of 7 days of age, it was confirmed that the expression of AMPA glutamate receptor and p-AMPA glutamate receptor is increased, but the osmotin of the present invention after the glutamate treatment In this case, it was confirmed that the expression of the AMPA glutamate receptor and the p-AMPA glutamate receptor was significantly reduced (Experimental Example 1).
본 발명의 상기 오스모틴을 유효 성분으로 포함하는 약학 조성물을 이용한 상기 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환의 예방 또는 치료는, 시냅토파이신의 발현 증가에 의해 수행될 수 있다.Prevention or treatment of neurological diseases caused by the excitatory toxicity or synaptic dysfunction using the pharmaceutical composition comprising the osmotin of the present invention as an active ingredient, may be performed by increasing the expression of synaptopycin.
본 발명에서 사용되는 용어, "시냅토파이신(synaptophysin)"은 신경 세포 또는 신경 내 분비 세포의 시냅스 소포막에 존재하는 약 30 kDa 가량의 당단백질로서, 시냅스 소포 단백질(synaptic vesicle protein) p38로도 알려져 있는 단백질을 의미한다.As used herein, the term "synaptophysin" is a glycoprotein of about 30 kDa present in the synaptic vesicle of nerve cells or secretory cells in the nerve, also known as synaptic vesicle protein p38. Mean protein.
본 발명의 구체적인 실시예에서, 생후 7일의 랫트에 글루타메이트를 처리한 경우 시냅토파이신의 발현이 현저히 감소되었으나, 상기 글루타메이트 처리 후 본 발명의 오스모틴을 처리한 결과, 상기 감소되었던 시냅토파이신의 발현 수준이 증가되는 것으로 확인되었다(실험예 2).In a specific embodiment of the present invention, the expression of synaptopycin was significantly reduced when glutamate was treated in the rat at 7 days of age, but the osmotin treatment of the present invention after the glutamate treatment resulted in the reduction of synaptopycin. It was confirmed that the expression level is increased (Experimental Example 2).
본 발명의 상기 오스모틴을 유효 성분으로 포함하는 약학 조성물을 이용한 상기 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환의 예방 또는 치료는, CaMKⅡ의 발현 감소에 의해 수행될 수 있다.The prevention or treatment of neurological diseases caused by the excitatory toxicity or synaptic dysfunction using the pharmaceutical composition comprising the osmotin of the present invention as an active ingredient may be performed by reducing the expression of CaMKII.
본 발명에서 사용되는 용어, "CaMKⅡ (Ca2+/calmodulin-depandent protein kinase Ⅱ)"는 세린/트레오닌-특이적 단백질 키나아제의 일종으로, Ca2+/calmodulin 복합체에 의하여 조절되는 키나아제를 의미한다.As used herein, the term “CaMKII (Ca 2+ / calmodulin-depandent protein kinase II)” is a kind of serine / threonine-specific protein kinase, and means a kinase regulated by a Ca 2+ / calmodulin complex.
본 발명의 구체적인 실시예에서, 생후 7일의 랫트에 글루타메이트를 처리한 경우 CaMKⅡ의 발현이 현저히 증가되었으나, 상기 글루타메이트 처리 후 본 발명의 오스모틴을 처리한 결과, 상기 증가되었던 CaMKⅡ의 발현 수준이 유의적으로 감소되는 것으로 확인되었다(실험예 1).In a specific embodiment of the present invention, the expression of CaMKII was significantly increased when glutamate was treated in the rat at 7 days of age, but as a result of treating the osmotin of the present invention after the glutamate treatment, the expression level of CaMKII was increased. It was confirmed that the decrease by (experimental example 1).
본 발명의 상기 오스모틴을 유효 성분으로 포함하는 약학 조성물을 이용한 상기 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환의 예방 또는 치료는, JNK/PI3K/Akt 경로에서 p-JNK의 발현 감소, p-PI3K의 발현 증가 또는 p-Akt의 발현 증가에 의해 수행될 수 있다.Prevention or treatment of neurological diseases caused by the excitatory toxicity or synaptic dysfunction using the pharmaceutical composition comprising the osmotin of the present invention, the expression of p-JNK in the JNK / PI3K / Akt pathway, p-PI3K It can be carried out by increasing the expression of or by increasing the expression of p-Akt.
본 발명에서 사용되는 용어, "JNK/PI3K/Akt 경로"는 세포 내 신호 전달 경로의 일종으로, 상기 경로에 관여하는 인자인 "JNK"는 "c-Jun N-terminal kinase"를 의미하고, "PI3K"는 "Phosphoinositide 3-kinase"를 의미하며, "Akt"는 "Protein kinase B (PKB)"를 의미한다. As used herein, the term "JNK / PI3K / Akt pathway" is a kind of intracellular signal transduction pathway, and a factor involved in the pathway "JNK" means "c-Jun N-terminal kinase" and " PI3K "means" Phosphoinositide 3-kinase "and" Akt "means" Protein kinase B (PKB) ".
본 발명의 구체적인 실시예에서, 생후 7일의 랫트에 글루타메이트를 처리한 경우 p-JNK는 발현이 증가되고 p-PI3K 및 p-Akt는 발현이 감소되었으나, 상기 글루타메이트 처리 후 본 발명의 오스모틴을 처리한 결과, 상기 증가되었던 p-JNK의 발현 수준은 유의적으로 감소되고, 상기 감소되었던 p-PI3K 및 p-Akt의 발현 수준은 유의적으로 증가되는 것으로 확인되었다(실험예 4).In a specific embodiment of the present invention, p-JNK expression was increased and p-PI3K and p-Akt expression was reduced when glutamate was treated in rats of 7 days of age, but the osmotin of the present invention after the glutamate treatment As a result, it was confirmed that the increased expression level of p-JNK was significantly decreased, and the expression levels of p-PI3K and p-Akt were decreased significantly (Experimental Example 4).
본 발명의 상기 오스모틴을 유효 성분으로 포함하는 약학 조성물은 또한, 신경 세포의 아팝토시스를 억제하는 효능을 나타낸다.The pharmaceutical composition comprising the osmotin of the present invention as an active ingredient also exhibits the effect of inhibiting apoptosis of nerve cells.
본 발명에서 사용되는 용어, "아팝토시스"는 세포가 유전자에 의해 제어되어 스스로 사멸하는 방식의 한 형태로서, 세포의 괴사나 병적인 죽음인 네크로시스(Necrosis)와는 구별되는 개념으로, 세포 자살이라고도 한다.As used herein, the term "apoptosis" is a form of a cell controlled by genes and kills itself, and is a concept distinguished from necrosis, which is necrosis or pathological death of cells. Also called.
본 발명의 구체적인 실시예에서, 아팝토시스에 관여한다고 알려진 p53, Bcl-2, Bax의 발현 변화 양상, 미토콘드리아 시토크롬의 세포 기질로의 전달 및 카스파아제-3과 PARP-1의 활성화 변화 양상을 관찰하였다.In a specific embodiment of the present invention, the expression changes of p53, Bcl-2, Bax, which are known to be involved in apoptosis, the delivery of mitochondrial cytochromes to the cell substrate and the activation of caspase-3 and PARP-1 were observed. It was.
본 발명에서 사용되는 용어, "p53"은 종양 억제 인자의 하나로, 세포의 이상 증식을 억제하고 암세포가 사멸되도록 유도하는 역할을 담당하는 것으로 알려져 있다.The term "p53" used in the present invention is one of tumor suppressor factors, and is known to play a role in inhibiting abnormal proliferation of cells and inducing cancer cells to die.
본 발명에서 사용되는 용어, "Bcl-2"는 분자량 약 26 kDa인 Bcl-2 패밀리 단백질의 일종으로 아팝토시스 억제 인자로 알려져 있다. Bcl-2는 Fas 항원과 TNF 수용체를 매개로 하는 아팝토시스를 차단하며, Bcl-2를 과발현시키면 아팝토시스를 억제하여 세포 수명이 연장되는 것으로 알려져 있다. 또한 Bcl-2는 Bax의 작용을 억제하고 시토크롬 C의 방출을 저해하는 역할을 수행하는 것으로 알려져 있다.As used herein, the term "Bcl-2" is a type of Bcl-2 family protein having a molecular weight of about 26 kDa and is known as an apoptosis inhibitor. Bcl-2 blocks apoptosis mediated by Fas antigen and TNF receptors, and overexpression of Bcl-2 inhibits apoptosis and is known to prolong cell life. Bcl-2 is also known to play a role in inhibiting the action of Bax and inhibiting the release of cytochrome C.
본 발명에서 사용되는 용어, "Bax"는 아팝토시스 유도 인자로 알려져 있으며, Bcl-2와 유전자 염기 서열이 비슷하여 Bcl-2 패밀리에 속한다. Bax가 활성화되면 아팝토시스를 촉진하여 세포 사멸이 증가하고, 반대로 Bcl-2가 활성화되면 아팝토시스를 억제하여 세포사멸이 감소하게 된다.As used herein, the term "Bax" is known as an apoptosis inducing factor and belongs to the Bcl-2 family because Bcl-2 and its gene sequence are similar. When Bax is activated, apoptosis is promoted and cell death is increased. On the contrary, when Bcl-2 is activated, apoptosis is suppressed and cell death is reduced.
본 발명의 구체적인 실시에에서, 생후 7일의 랫트에 글루타메이트를 처리한 경우 p53 단백질의 발현 수준 및 Bax/Bcl-2 비율이 증가되었으며, 미토콘드리아 시토크롬 C의 전달 및 카스파아제-3와 PARP-1의 활성화가 촉진되었으나, 상기 글루타메이트 처리 후 본 발명의 오스모틴을 처리한 결과, 상기 양상들이 모두 반대로 나타나는 것으로 확인되어, 신경 세포의 아팝토시스를 억제하는 것으로 확인되었다(실험예 3).In a specific embodiment of the present invention, treatment of glutamate in rats at 7 days of age increased the expression level of p53 protein and the Bax / Bcl-2 ratio, the delivery of mitochondrial cytochrome C and the caspase-3 and PARP-1. Activation was promoted, but as a result of treating the osmotin of the present invention after the glutamate treatment, all of the above aspects were found to be reversed, and it was confirmed that the apoptosis of nerve cells was suppressed (Experimental Example 3).
본 발명의 상기 약학 조성물은, 오스모틴을 유효 성분으로 포함하는 것에 더하여, 약학적으로 허용 가능한 담체를 추가로 포함할 수 있다.The pharmaceutical composition of the present invention may further include a pharmaceutically acceptable carrier, in addition to including osmotin as an active ingredient.
본 발명에서, 상기 "약학적으로 허용 가능"하다는 것은, 이를 투여 시 생물체를 자극하지 않으면서, 투여되는 화합물의 생물학적 활성 및 특성을 저해하지 않는, 약학 분야에서 통상적으로 사용되는 것을 의미한다.In the present invention, the "pharmaceutically acceptable" means that it is commonly used in the pharmaceutical field that does not impede the biological activity and properties of the compound to be administered without stimulating the organism upon administration thereof.
본 발명에서, 상기 담체의 종류는 특별히 제한되지 아니하며 당해 기술 분야에서 통상적으로 사용되는 담체라면 어느 것이든 사용할 수 있다. 상기 담체의 비제한적인 예로는, 식염수, 멸균수, 링거액, 완충 식염수, 알부민 주사 용액, 락토오스, 덱스트로오스, 수크로오스, 솔비톨, 만니톨, 자일리톨, 에리스리톨, 말티톨, 말토 덱스트린, 글리세롤, 에탄올 등을 들 수 있다. 이들은 단독으로 사용되거나 2 종 이상을 혼합하여 사용될 수 있다.In the present invention, the type of the carrier is not particularly limited and any carrier can be used as long as it is commonly used in the art. Non-limiting examples of the carrier include saline, sterile water, Ringer's solution, buffered saline, albumin injection solution, lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, maltodextrin, glycerol, ethanol, and the like. Can be. These may be used alone or in combination of two or more thereof.
또한, 본 발명의 상기 약학 조성물은 필요한 경우, 부형제, 희석제, 항산화제, 완충액 또는 정균제 등 기타 약학적으로 허용 가능한 첨가제들을 첨가하여 사용할 수 있으며, 충진제, 증량제, 습윤제, 붕해제, 분산제, 계면 활성제, 결합제 또는 윤활제 등을 부가적으로 첨가하여 사용할 수 있다.In addition, the pharmaceutical composition of the present invention may be used by adding other pharmaceutically acceptable additives, such as excipients, diluents, antioxidants, buffers or bacteriostatic agents, if necessary, fillers, extenders, wetting agents, disintegrants, dispersants, surfactants , Binders or lubricants may be additionally added and used.
본 발명의 상기 약학 조성물에 있어서, 오스모틴은 상기 약학 조성물의 전체의 중량을 기준으로 0.00001 중량% 내지 99.99 중량%로 포함될 수 있으며, 바람직하게는 1 중량% 내지 99.99 중량%, 보다 바람직하게는 10 중량% 내지 99.99 중량%, 더욱 바람직하게는 50 중량% 내지 99.99 중량%로 포함될 수 있으며, 약학 조성물의 전체 함량으로도 포함될 수 있다.In the pharmaceutical composition of the present invention, osmotin may be included in 0.00001% to 99.99% by weight based on the total weight of the pharmaceutical composition, preferably 1% to 99.99% by weight, more preferably 10 It may be included in the weight% to 99.99% by weight, more preferably 50% to 99.99% by weight, and may also be included in the total content of the pharmaceutical composition.
본 발명의 상기 약학 조성물은 경구 투여 또는 비경구 투여를 위한 적합한 다양한 제형으로 제제화되어 사용될 수 있다.The pharmaceutical compositions of the present invention can be formulated and used in a variety of formulations suitable for oral or parenteral administration.
상기 경구 투여용 제제의 비제한적인 예로는, 트로키제(troches), 로젠지(lozenge), 정제, 수용성 현탁액, 유성 현탁액, 조제 분말, 과립, 에멀젼, 하드 캡슐, 소프트 캡슐, 시럽 또는 엘릭시르제 등을 들 수 있다.Non-limiting examples of the formulation for oral administration include troches, lozenges, tablets, aqueous suspensions, oily suspensions, prepared powders, granules, emulsions, hard capsules, soft capsules, syrups or elixirs, and the like. Can be mentioned.
본 발명의 상기 약학 조성물을 경구 투여용으로 제제화하기 위하여, 락토오스, 사카로오스, 솔비톨, 만니톨, 전분, 아밀로펙틴, 셀룰로오스 또는 젤라틴 등과 같은 결합제; 디칼슘 포스페이트 등과 같은 부형제; 옥수수 전분 또는 고구마 전분 등과 같은 붕해제; 스테아르산 마그네슘, 스테아르산 칼슘, 스테아릴 푸마르산 나트륨 또는 폴리에틸렌 글리콜 왁스 등과 같은 윤활유 등을 사용할 수 있으며, 감미제, 방향제, 시럽제 등도 사용할 수 있다.In order to formulate the pharmaceutical composition of the present invention for oral administration, a binder such as lactose, saccharose, sorbitol, mannitol, starch, amylopectin, cellulose or gelatin and the like; Excipients such as dicalcium phosphate and the like; Disintegrants such as corn starch or sweet potato starch; Lubricants such as magnesium stearate, calcium stearate, sodium stearyl fumarate or polyethylene glycol wax and the like can be used, and sweeteners, fragrances, syrups and the like can also be used.
나아가 캡슐제의 경우에는 상기 언급한 물질 외에도 지방유와 같은 액체 담체 등을 추가로 사용할 수 있다.Furthermore, in the case of a capsule, a liquid carrier such as fatty oil may be additionally used in addition to the above-mentioned materials.
상기 비경구용 제제의 비제한적인 예로는, 주사액, 좌제, 호흡기 흡입용 분말, 스프레이용 에어로졸제, 연고, 도포용 파우더, 오일, 크림 등을 들 수 있다.Non-limiting examples of the parenteral preparations include injection liquids, suppositories, respiratory inhalation powders, spray aerosols, ointments, application powders, oils, creams, and the like.
본 발명의 상기 약학 조성물을 비경구 투여용으로 제제화하기 위하여, 멸균된 수용액, 비수성용제, 현탁제, 유제, 동결 건조 제제, 외용제 등을 사용할 수 있으며, 상기 비수성용제, 현탁제로는 프로필렌글리콜, 폴리에틸렌글리콜, 올리브 오일과 같은 식물성 기름, 에틸올레이트와 같은 주사 가능한 에스테르 등이 사용될 수 있다.In order to formulate the pharmaceutical composition for parenteral administration, a sterile aqueous solution, a non-aqueous solvent, a suspension, an emulsion, a freeze-dried preparation, an external preparation, and the like may be used. As the non-aqueous solvent and the suspension, propylene glycol, polyethylene Glycols, vegetable oils such as olive oil, injectable esters such as ethyloleate and the like can be used.
또한, 보다 구체적으로 본 발명의 상기 약학 조성물을 주사액으로 제제화하는 경우, 본 발명의 상기 조성물을 안정제 또는 완충제와 함께 물에서 혼합하여 용액 또는 현탁액으로 제조하고 이를 앰플(ampoule) 또는 바이알(vial)의 단위 투여용으로 제제화할 수 있다. 또한, 본 발명의 상기 약학 조성물을 에어로졸제로 제제화하는 경우, 수분산된 농축물 또는 습윤 분말이 분산되도록 추진제 등이 첨가제와 함께 배합할 수 있다.Further, more specifically, when the pharmaceutical composition of the present invention is formulated as an injection solution, the composition of the present invention is mixed with water with a stabilizer or buffer to prepare a solution or suspension, which may be used as an ampoule or vial. It may be formulated for unit administration. In addition, when the pharmaceutical composition of the present invention is formulated with an aerosol, a propellant or the like may be combined with the additive to disperse the dispersed concentrate or the wet powder.
또한, 본 발명의 상기 약학 조성물을 연고, 크림 등으로 제제화하는 경우에는, 동물성 유, 식물성 유, 왁스, 파라핀, 전분, 트라칸트, 셀룰로오스 유도체, 폴리에틸렌 글리콜, 실리콘, 벤토나이트, 실리카, 탈크, 산화 아연 등을 담체로 사용하여 제제화할 수 있다.In addition, when the pharmaceutical composition of the present invention is formulated into an ointment, a cream, or the like, animal oil, vegetable oil, wax, paraffin, starch, trakant, cellulose derivative, polyethylene glycol, silicone, bentonite, silica, talc, zinc oxide It may be formulated using such as carrier.
본 발명의 상기 약학 조성물의 약학적 유효량, 유효 투여량은 상기 약학 조성물의 제제화 방법, 투여 방식, 투여 시간 및/또는 투여 경로 등에 의해 다양해질 수 있으며, 상기 약학 조성물의 투여로 달성하고자 하는 반응의 종류와 정도, 투여 대상이 되는 개체의 종류, 연령, 체중, 일반적인 건강 상태, 질병의 증세나 정도, 성별, 식이, 배설, 해당 개체에 동시 또는 이시에 함께 사용되는 약물 기타 조성물의 성분 등을 비롯한 여러 인자 및 의약 분야에서 잘 알려진 유사 인자에 따라 다양해질 수 있으며, 당해 기술 분야에서 통상의 지식을 가진 자는 목적하는 치료에 효과적인 투여량을 용이하게 결정하고 처방할 수 있다.A pharmaceutically effective amount, effective dosage of the pharmaceutical composition of the present invention may vary depending on the method of formulating the pharmaceutical composition, the mode of administration, the time of administration and / or the route of administration, and the likeness of the reaction to be achieved by the administration of the pharmaceutical composition. The type and extent of the subject, the type, age, weight, general state of health, condition or extent of the disease, sex, diet, excretion, components of the drug or other composition used concurrently or simultaneously with the subject, Various factors and similar factors well known in the medical arts can be varied, and one of ordinary skill in the art can easily determine and prescribe a dosage effective for the desired treatment.
본 발명의 상기 약학 조성물의 투여는 하루에 1회 투여될 수 있고, 수회에 나누어 투여될 수도 있다. 본 발명의 약학 조성물은 개별 치료제로 투여하거나 다른 치료제와 병용하여 투여될 수 있고 종래의 치료제와는 순차적 또는 동시에 투여될 수 있다. 상기 요소를 모두 고려하여 부작용 없이 최소한의 양으로 최대 효과를 얻을 수 있는 양을 투여하는 것이 중요하며, 당업자에 의해 용이하게 결정될 수 있다.Administration of the pharmaceutical composition of the present invention may be administered once a day, may be divided into several times. The pharmaceutical compositions of the present invention may be administered as individual therapeutic agents or in combination with other therapeutic agents and may be administered sequentially or simultaneously with conventional therapeutic agents. Taking all of the above factors into consideration, it is important to administer an amount that can obtain the maximum effect in a minimum amount without side effects, and can be easily determined by those skilled in the art.
본 발명의 상기 약학 조성물을 주사 투여하는 경우에는 보다 바람직한 효과를 위하여, 0.01 mg/kg/day 내지 100 mg/kg/day의 양으로 투여될 수 있으며, 보다 바람직하게는 0.1 mg/kg/day 내지 70 mg/kg/day, 더욱 바람직하게는 1 mg/kg/day 내지 50 mg/kg/day의 양으로 투여될 수 있다. 상기 투여는 1회에 투여될 수 있고 수회에 나누어 투여될 수 있으며, 투여 주기는 적절히 선택될 수 있다.In the case of injection administration of the pharmaceutical composition of the present invention, for a more preferable effect, it may be administered in an amount of 0.01 mg / kg / day to 100 mg / kg / day, more preferably from 0.1 mg / kg / day to It may be administered in an amount of 70 mg / kg / day, more preferably 1 mg / kg / day to 50 mg / kg / day. The administration may be administered one time, divided into several times, and the administration cycle may be appropriately selected.
본 발명의 상기 약학 조성물의 투여 경로 및 투여 방식은 각각 독립적일 수 있으며, 그 방식에 있어 특별히 제한되지 아니하며, 목적하는 해당 부위에 상기 약학 조성물이 도달할 수 있는 한 임의의 투여 경로 및 투여 방식에 따를 수 있다. 상기 약학 조성물은 경구 투여 또는 비경구 투여 방식으로 투여할 수 있다.The route of administration and mode of administration of the pharmaceutical composition of the present invention may be independent of each other, and are not particularly limited in the way, and may be any route of administration and mode of administration as long as the pharmaceutical composition can reach the desired site. Can follow. The pharmaceutical composition may be administered by oral or parenteral administration.
상기 비경구 투여하는 방법으로는, 예를 들어 정맥 내 투여, 복강 내 투여, 근육 내 투여, 경피 투여 또는 피하 투여 등을 이용할 수 있으며, 상기 조성물을 질환 부위에 도포하거나 분무, 흡입하는 방법 또한 이용할 수 있으나 이들에 제한되지 아니한다.As the parenteral administration method, for example, intravenous administration, intraperitoneal administration, intramuscular administration, transdermal administration or subcutaneous administration may be used, and the method of applying, spraying or inhaling the composition to a diseased site may also be used. May be, but is not limited to these.
본 발명의 또 다른 구현예에 따르면, 오스모틴을 유효 성분으로 포함하는 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환의 에방 또는 개선용 식품 조성물을 제공한다.According to another embodiment of the present invention, there is provided a food composition for the prevention or improvement of neurological diseases caused by excitatory toxicity or synaptic dysfunction comprising osmotin as an active ingredient.
본 발명에서 사용되는 용어, "개선"이란 상태의 완화 또는 치료와 관련된 파라미터, 예를 들면 증상의 정도를 적어도 감소시키는 모든 행위를 의미한다.As used herein, the term " improvement " means any action that at least reduces the parameters associated with alleviation or treatment of a condition, such as the degree of symptoms.
*본 발명의 상기 식품 조성물은 특별히 제한되지 아니하며, 건강 기능 식품 조성물을 포함한다.The food composition of the present invention is not particularly limited and includes a health functional food composition.
본 발명의 상기 건강 기능 식품 조성물을 식품 첨가물로 사용할 경우, 상기 조성물을 그대로 첨가하거나 다른 식품 또는 식품 성분과 함께 사용될 수 있고, 통상적인 방법에 따라 적절하게 사용될 수 있다.When the health functional food composition of the present invention is used as a food additive, the composition may be added as it is or used with other food or food ingredients, and may be appropriately used according to a conventional method.
상기 식품의 종류는 특별히 제한되지 아니하며, 통상적인 의미에서의 식품을 모두 포함한다. 상기 물질을 첨가할 수 있는 식품의 비제한적인 예로는 육류, 소세지, 빵, 초콜릿, 캔디류, 스낵류, 과자류, 피자, 라면, 기타 면류, 껌류, 아이스크림류를 포함한 낙농제품, 각종 스프, 음료수, 차, 드링크제, 알코올 음료 및 비타민 복합제 등을 들 수 있다.The kind of the food is not particularly limited, and includes all foods in a general sense. Non-limiting examples of foods that can be added to the material include meat, sausages, bread, chocolate, candy, snacks, confectionery, pizza, ramen, dairy products, including other noodles, gums, ice cream, various soups, drinks, tea , A drink, an alcoholic beverage, and a vitamin complex.
본 발명의 상기 건강 기능 식품 조성물이 음료 조성물인 경우, 통상의 음료와 같이 여러 가지 향미제 또는 천연 탄수화물 등을 추가 성분으로서 함유할 수 있다. 상기 천연 탄수화물의 비제한적인 예로 포도당, 과당과 같은 모노사카라이드; 말토스, 수크로오스와 같은 디사카라이드; 덱스트린, 사이클로덱스트린과 같은 천연 감미제; 사카린, 아스파르탐과 같은 합성 감미제 등을 들 수 있다. 상기 첨가되는 추가 성분의 비율은 당업자의 선택에 의해 적절하게 결정될 수 있다.When the health functional food composition of the present invention is a beverage composition, it may contain various flavors or natural carbohydrates and the like as additional ingredients, as in the usual beverage. Non-limiting examples of the natural carbohydrates include monosaccharides such as glucose and fructose; Disaccharides such as maltose and sucrose; Natural sweeteners such as dextrin, cyclodextrin; Synthetic sweeteners such as saccharin and aspartame; and the like. The proportion of the additional components added above may be appropriately determined by the choice of those skilled in the art.
상기 외에 본 발명의 건강 기능 식품 조성물은 여러 가지 영양제, 비타민, 전해질, 풍미제, 착색제, 펙트산 및 그의 염, 알긴산 및 그의 염, 유기산, 보호성 콜로이드 증점제, pH 조절제, 안정화제, 방부제, 글리세린, 알코올, 탄산 음료에 사용되는 탄산화제 등을 함유할 수 있다. 그 밖에 본 발명의 건강 기능 식품 조성물은 천연 과일 주스, 과일 음료 또는 야채 음료 등의 제조를 위한 과육을 함유할 수 있다. 이러한 성분은 독립적으로 사용되거나 2 이상을 조합하여 사용할 수 있다. 이러한 첨가물의 비율 또한 당업자에 의해 적절히 선택될 수 있다.In addition to the above, the health functional food composition of the present invention includes various nutrients, vitamins, electrolytes, flavors, coloring agents, pectic acid and salts thereof, alginic acid and salts thereof, organic acids, protective colloidal thickeners, pH adjusting agents, stabilizers, preservatives, glycerin , Alcohols, carbonation agents used in carbonated beverages, and the like. In addition, the health functional food composition of the present invention may contain a flesh for preparing natural fruit juice, fruit drink or vegetable drink. These components can be used independently or can be used in combination of 2 or more. The proportion of such additives may also be appropriately selected by those skilled in the art.
본 발명의 또 다른 구현예에 따르면, 오스모틴을 유효 성분으로 포함하는 상기 약학 조성물을 개체에 투여하는 것을 포함하는, 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환을 예방 또는 치료하는 방법을 제공한다.According to another embodiment of the present invention, there is provided a method for preventing or treating neurological diseases caused by excitotoxicity or synaptic dysfunction, comprising administering to a subject the pharmaceutical composition comprising osmotin as an active ingredient.
본 발명의 또 다른 구현예에 따르면, 오스모틴을 유효 성분으로 포함하는 상기 식품 조성물을 개체에 투여하는 것을 포함하는, 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환을 예방 또는 개선하는 방법을 제공한다.According to another embodiment of the present invention, there is provided a method for preventing or ameliorating neurological diseases caused by excitatory toxicity or synaptic dysfunction, comprising administering to a subject the food composition comprising osmotin as an active ingredient.
본 발명에서 사용되는 용어, "개체"는 쥐, 가축, 인간 등을 포함하는 포유 동물을 비롯한 모든 동물을 의미한다.As used herein, the term "individual" means all animals, including mammals, including rats, domestic animals, humans, and the like.
본 발명에서, 상기 조성물의 투여량, 투여 경로, 투여 방식 등에 관한 설명은, 본 발명의 상기 약학 조성물과 관련하여 설명한 내용으로 갈음한다.In the present invention, the description of the dosage, route of administration, mode of administration and the like of the composition is replaced with the contents described in connection with the pharmaceutical composition of the present invention.
이하, 실시예를 통하여 본 발명을 보다 상세히 설명한다. 다만, 이들 실시예는 본 발명을 예시적으로 설명하기 위한 것에 불과하므로 본 발명의 범위가 이들 실시예에 의해 한정되는 것으로 해석되어서는 아니된다.Hereinafter, the present invention will be described in more detail with reference to Examples. However, these examples are only for illustrating the present invention by way of example, and the scope of the present invention should not be construed as being limited by these examples.
실시예 1: 동물 및 약물 처리Example 1: Animal and Drug Treatment
출생 7일 이후의 Sprague-Dawley 랫트(평균 몸무게 18 g) 20 마리를 1 군당 5 마리씩 총 4개의 군으로 나누고, 상기 각 군에 각각 살린(0.9%)[대조군], 글루타메이트(10 mg/kg), 오스모틴(15 ㎍/g), 및 글루타메이트(10 mg/kg)+오스모틴(15 ㎍/g)을 피하 주사 방법으로 처리하였다. 상기 약물처리에서, 글루타메이트 처리는 오스모틴 보다 1 시간 또는 2 시간 전에 수행되었다. 상기 각 군의 랫트들은 상기 약물 처리 후 4 시간 내지 12 시간 후 희생되었다. 본 실시예의 모든 실험 및 윤리적 절차는 대한민국 경상대학교의 생물학부에서 Division of Applied Life Sciences 지방 동물 윤리 위원회에 따라 이루어졌다.Twenty Sprague-Dawley rats (average weight 18 g) after 7 days of birth were divided into four groups of five animals per group, each of which was added to each group (0.9%) [control], glutamate (10 mg / kg). , Osmotin (15 μg / g), and glutamate (10 mg / kg) + osmotin (15 μg / g) were treated by subcutaneous injection method. In the drug treatment, glutamate treatment was performed 1 hour or 2 hours before osmotin. Rats from each group were sacrificed 4 to 12 hours after the drug treatment. All experimental and ethical procedures of this example were made in accordance with the Division of Applied Life Sciences Local Animal Ethics Committee at the Department of Biology, Gyeongsang National University, Korea.
상기에서 글루타메이트는 Sigma-Aldrich Co. LLC로부터 구입하였고 오스모틴은 염제-적응(salt-adapted)된 배양된 토바코(Nicotiana tabacum) 세포로부터 정제하여 이용하였다. 오스모틴의 내독소 함량은 <0.03 EU/mg 단백질이었다.In the above glutamate is Sigma-Aldrich Co. It was purchased from LLC and osmotin was purified from salt-adapted cultured Nicotiana tabacum cells. The endotoxin content of osmotin was <0.03 EU / mg protein.
실시예 2: 조직 채취 및 샘플 준비Example 2: Tissue Collection and Sample Preparation
상기 실시예 1에서 살린, 글루타메이트 또는 글루타메이트+오스모틴으로 처리된 새끼 랫트의 뇌 섹션을 약물 처리 후 12시간 후에 분석하였다. 1×포스페이트 버퍼 살린(PBS)을 처리하고, 4% 얼음-냉처리된 파라포름알데하이드를 처리하여 경심 관류(transcardial perfusion)를 수행하였다. 그 다음, 뇌 조직을 4% 파라포름알데하이드에서 밤새 후(post)-고정시키고, 상기 뇌 조직이 튜브 바닥에 가라앉을 때까지 20%의 수쿠로오스에 옮겼다. 상기 뇌를 O.C.T (A.O., USA)에서 냉동시킨 다음, 상기 뇌를 CM 3050S 크라이오스탯(Leica, Germany)을 이용하여 관상면(coronal plane)으로 14 ㎛ 섹션으로 절단하였다. 상기 뇌 섹션을 양전하가 부가된 슬라이드에서 프로브에 해동-고정시켰다(Fisher, USA).Brain sections of pups treated with saline, glutamate or glutamate + osmotin in Example 1 were analyzed 12 hours after drug treatment. Transcardial perfusion was performed by treatment with 1 × phosphate buffered saline (PBS) and 4% ice-cold treated paraformaldehyde. The brain tissue was then post-fixed in 4% paraformaldehyde and transferred to 20% Sukurose until the brain tissue sank to the bottom of the tube. The brain was frozen in OCT (AO, USA) and then the brain was cut into 14 μm sections in the coronal plane using a CM 3050S cryostat (Leica, Germany). The brain sections were thawed-fixed to probes on positively charged slides (Fisher, USA).
실시예 3: 웨스턴 블랏 분석Example 3: Western Blot Analysis
여러 단백질의 발현 수준을 측정하기 위하여 웨스턴 블랏 분석을 이용하였다. 상기 실시예 1의 각 군의 랫트를 상기 약물 처리 후 4 시간 후에 희생시키고, 피질 및 해마를 절개하여 드라이 아이스로 냉동시켰다. 그 다음, 상기 피질 및 해마를 프로테아제 억제 인자 칵테일을 포함한 0.2M PBS에 균질화하였다. 상기 호모지네이트의 단백질 농도를 측정하기 위하여 Bio-Rad 단백질 분석 키트(Bio-Rad Laboratories, CA, USA)를 사용하였다. 총 단백질의 일정량(샘플당 30 ㎍)을 10% 내지 15% SDS-PAGE 전기영동에 사용하였다.Western blot analysis was used to measure the expression levels of various proteins. Rats of each group of Example 1 were sacrificed 4 hours after the drug treatment, and the cortex and hippocampus were excised and frozen with dry ice. The cortex and hippocampus were then homogenized in 0.2 M PBS with a protease inhibitory factor cocktail. Bio-Rad Protein Assay Kit (Bio-Rad Laboratories, CA, USA) was used to measure the protein concentration of the homogenate. An amount of total protein (30 μg per sample) was used for 10% to 15% SDS-PAGE electrophoresis.
검출된 단백질의 분자량을 측정하기 위하여, 10 kDa 내지 245 kDa의 광범위한 분자량을 포함하는, 미리 염색된 단백질 사다리(protein ladder)(GangNam-STAINTM, iNtRon Biotechnology, Inc. Republic of Korea)를 병렬로 작동시켜 이용하였다. 멤브레인 블로킹 및 비-특이성을 최소화하기 위하여, 5% (w/v) 탈지유를 이용하여 블로킹을 수행하였다.In order to determine the molecular weight of the detected protein, a pre-stained protein ladder (GangNam-STAIN , iNtRon Biotechnology, Inc. Republic of Korea) containing a wide range of molecular weights from 10 kDa to 245 kDa was operated in parallel. Was used. In order to minimize membrane blocking and non-specificity, blocking was performed using 5% (w / v) skim milk.
단백질을 검출하기 위하여 면역 블랏팅에 사용된 항체로, 래빗-유래 항-액틴, 항-Bcl-2, 항-Bax 및 항-카스파아제-3; 염소-유래 항-시토크롬 c 및 항-p53; 및 마우스-유래 다중클론성 항체 항-PARP-1 및 항-JNK (Santa Cruz Biotechnology, Santa Cruz, CA, USA)를 사용하였다. 또한, 래빗-유래 항-p-CREB (Ser133), 항-CaMKⅡ, 항-p-Akt (Ser473), 항-p-PI3K (Y458/Y199), 항-p-JNK (Thr183/Tyr185), 항-AMPA, 항-p-AMPA (Ser845) 및 항-시냅토파이신(Cell Signaling Technology, Inc)을 사용하였다.Antibodies used in immunoblotting to detect proteins include rabbit-derived anti-actin, anti-Bcl-2, anti-Bax and anti-caspase-3; Chlorine-derived anti-cytochrome c and anti-p53; And mouse-derived polyclonal antibodies anti-PARP-1 and anti-JNK (Santa Cruz Biotechnology, Santa Cruz, CA, USA). In addition, rabbit-derived anti-p-CREB (Ser133), anti-CaMKII, anti-p-Akt (Ser473), anti-p-PI3K (Y458 / Y199), anti-p-JNK (Thr183 / Tyr185), anti -AMPA, anti-p-AMPA (Ser845) and anti-synaptopycin (Cell Signaling Technology, Inc) were used.
제조자(Amersham Pharmacia Biotech, Uppsala, Sweden)의 지시에 따라 ECL 검출 시약을 사용하여 막-결합 이차 항체를 시각화하였다. 그 다음, X-선 필름을 스캔하고 컴퓨터-베이스 Sigma Gel 프로그램 버전 1.0 (SPSS, Chicago, IL, USA)을 이용한 농도계(densitometry)를 통해 밴드의 광학 밀도를 분석하였다.Membrane-bound secondary antibodies were visualized using ECL detection reagents according to the manufacturer's instructions (Amersham Pharmacia Biotech, Uppsala, Sweden). The optical density of the band was then analyzed by densitometry using a computer-based Sigma Gel program version 1.0 (SPSS, Chicago, IL, USA).
실시예 4: 면역 형광 분석Example 4: Immunofluorescence Assay
상기 실시예 2에 따른 조직-함유 슬라이드를 0.01M PBS에서 15분 동안 2번 세척하고 단백질 분해 효소 K 용액을 상기 조직에 첨가하고 37℃에서 5분 동안 배양하였다. 그 후, 상기 조직을 PBS 내 0.3% Triton X-100 및 정상 돼지 혈청을 포함하는 블로킹 용액에서 90분 동안 배양하였다. 그 다음, 1차 항체(래빗 다중클론성 p-Akt, p-PI3K 및 카스파제-3 (Cell Signaling Technology, Inc), 및 염소 다중클론성 IgG p53 및 p-JNK (Santa cruz); 상기 항체들은 모두 PBS 내 1:100으로 존재)를 밤새 4℃에서 적용하였다. 그 다음, 이차 항체(돼지 항-래빗 TRITC (Dako), 및 래빗 항-염소 FITC (Santa cruz); 상기 항체들은 모두 PBS 내 1:50으로 존재)를 90분 동안 상온에서 적용하였다. 그 다음, 슬라이드를 5분 동안 PBS로 2번 세척하였다. 그 다음, 이중 염색을 위해 배양을 병행하여 수행하였다. 유리 커버 슬라이드를 봉입제를 이용하여 유리 슬라이드 상에 봉입하고, 공초점 현미경(FluoView FV 1000 Olympus, Japan)을 사용하여 이미지를 캡쳐하였다.The tissue-containing slides according to Example 2 were washed twice for 15 minutes in 0.01 M PBS and proteolytic enzyme K solution was added to the tissues and incubated at 37 ° C. for 5 minutes. The tissues were then incubated for 90 minutes in blocking solution containing 0.3% Triton X-100 and normal pig serum in PBS. The primary antibodies (rabbit polyclonal p-Akt, p-PI3K and caspase-3 (Cell Signaling Technology, Inc), and goat polyclonal IgG p53 and p-JNK (Santa cruz); All present at 1: 100 in PBS) overnight at 4 ° C. Secondary antibodies (pig anti-rabbit TRITC (Dako), and rabbit anti-goat FITC (Santa cruz); all of these antibodies are present at 1:50 in PBS) were applied at room temperature for 90 minutes. The slides were then washed twice with PBS for 5 minutes. Then, the culture was performed in parallel for double staining. Glass cover slides were encapsulated on glass slides with an encapsulant and images were captured using a confocal microscope (FluoView FV 1000 Olympus, Japan).
실시예 5: 터널(TUNEL) 염색Example 5: Tunnel Staining
글루타메이트에 의해 유도되는 세포 아팝토시스를 검출하기 위하여, 핵 DNA를 TUNEL (GenScript Corporation, USA)로 염색하고 PI를 사용하여 대비 염색을 수행하였다. TUNEL 염색은 In Situ Cell Death Detection kit Fluorescein (GenScript, NJ, USA)의 권장사항에 따라 수행하였다. 유리 커버 슬라이드를 봉입제를 이용하여 유리 슬라이드 상에 봉입하고, TUNEL 염색(그린색) 검출을 위하여 FITC 필터를 이용하였다. 공초점 레이저 스캐닝 현미경(FluoView FV 1000, Olympus, Japan)으로 TUNEL-양성(그린) 염색 패턴을 수득하고, 각 섹션의 상이한 영역의 TUNEL-양성 세포를 컴퓨터 베이스 프로그램으로 카운트하였다.To detect cellular apoptosis induced by glutamate, nuclear DNA was stained with TUNEL (GenScript Corporation, USA) and contrast staining was performed using PI. TUNEL staining was performed according to the recommendations of In Situ Cell Death Detection Kit Fluorescein (GenScript, NJ, USA). Glass cover slides were encapsulated on glass slides with an encapsulant and FITC filters were used for TUNEL staining (green color) detection. TUNEL-positive (green) staining patterns were obtained with a confocal laser scanning microscope (FluoView FV 1000, Olympus, Japan), and TUNEL-positive cells in different regions of each section were counted with a computer based program.
실시예 6: 데이터의 분석 및 통계Example 6: Analysis and Statistics of Data
웨스턴 블랏의 밴드를 스캔하고 Sigma Gel System (SPSS Inc., Chicago, IL)을 이용한 농도계를 통해 밴드의 광학 밀도를 분석하였다. 밀도 값은 평균±SEM으로 표시하였다. 유의한 차이점을 확인하기 위하여 변수의 일-원 분석(one-way analysis, ANOVA)을 수행하였고, 이후 관련 처리 그룹 간의 차이점의 유의성을 확인하기 위한 Student's t-test를 수행하였다. 0.05 미만의 P 값은 유의적인 것으로 고려하였다(p<0.05).The band of Western blot was scanned and the optical density of the band was analyzed by densitometer using Sigma Gel System (SPSS Inc., Chicago, IL). Density values are expressed as mean ± SEM. One-way analysis (ANOVA) of variables was performed to identify significant differences, followed by Student's t-test to identify the significance of differences among the treatment groups. P values less than 0.05 were considered significant (p <0.05).
실험예 1: 글루타메이트에 의해 유도된 흥분독성을 약화시키는 오스모틴의 효능 검증Experimental Example 1: Verification of the efficacy of osmotin to attenuate excitatory toxicity induced by glutamate
(1) AMPA 글루타메이트 수용체의 발현 수준 감소(1) Reduction of expression level of AMPA glutamate receptor
AMPA 글루타메이트 수용체는 4개의 상이한 서브 유닛인 GluR1-4의 조합으로 이루어진 리간드-개폐형(ligand-gated) 이온 채널로서, 글루타메이트 수용체의 과발현은 흥분독성을 유발하며 결과적으로 신경 손상을 유발할 수 있다.AMPA glutamate receptors are ligand-gated ion channels consisting of a combination of four different subunits, GluR1-4, wherein overexpression of glutamate receptors causes excitatory toxicity and may result in neuronal damage.
출생 7일 이후의 랫트에 글루타메이트를 1회 피하주사한 후 4시간 후 상기 랫트의 뇌의 피질 및 해마를 관찰한 결과, AMPA 글루타메이트 수용체 단백질(Glu2/3/4)이 과발현된 반면, 상기 글루타메이트 처리 후, 오스모틴으로 처리한 동물군에서는 AMPA 수용체의 단백질 발현 수준이 현저히 감소되는 것으로 관찰되었다(도 1a 및 1b).The rat cortex and hippocampus were observed 4 hours after the first subcutaneous injection of glutamate in rats after 7 days of birth, whereas the AMPA glutamate receptor protein (Glu2 / 3/4) was overexpressed, whereas the glutamate treatment was performed. Later, it was observed that protein expression levels of AMPA receptors were significantly reduced in the animal group treated with osmotin (FIGS. 1A and 1B).
(2) p-AMPA 글루타메이트 수용체의 발현 수준 감소(2) decreased expression level of p-AMPA glutamate receptor
흥분독성 및 장기 상승 작용(long-term potentiation, LTP)과 연관되어 있는 것으로 알려진, AMPA 글루타메이트 수용체 단백질(Glu1)의 serine 845 위치의 인산화에 대한 실험을 수행한 결과, 글루타메이트를 처리한 동물군에서는 뇌의 피질 및 해마에서 p-AMPA 글루타메이트 수용체의 발현 수준이 증가되는 것으로 관찰된 반면, 글루타메이트 처리 후 오스모틴을 처리한 동물군의 뇌의 피질 및 해마에서 p-AMPA 글루타메이트 수용체의 발현 수준이 감소되는 것으로 관찰되었다.Experiments on phosphorylation of the serine 845 position of AMPA glutamate receptor protein (Glu1), known to be associated with excitatory toxicity and long-term potentiation (LTP), showed that brains in glutamate-treated animals It was observed that the expression level of p-AMPA glutamate receptor is increased in the cortex and hippocampus of, whereas the expression level of p-AMPA glutamate receptor is decreased in the cortex and hippocampus of brains of osmotin treated animals after glutamate treatment. Was observed.
(3) CaMKⅡ의 발현 수준 감소(3) decreased expression level of CaMKII
미토콘드리아 내 Ca+2의 축적은 급성 글루타메이트 흥분독성에서 상당히 중요한 단계에 속하는 것으로, 흥분독성은 세포 내 자유 칼슘 수준의 현저한 증가와 관련이 있다. 칼모둘린-의존성 키나아제-Ⅱ (CaMKⅡ)는 급성 흥분독성으로 인해 신경 세포 사멸에 이르게 되는 생화학적 기작에 있어서 중요한 조절 인자로서, 길항물질(antagonist)을 이용하여 CaMKⅡ의 작용을 억제시키는 경우 흥분독성에 의한 신경 세포의 사멸이 감소된다.Accumulation of Ca +2 in mitochondria is a significant step in acute glutamate excitatory toxicity, which is associated with a significant increase in free calcium levels in cells. Calmodulin-dependent kinase-II (CaMKII) is an important regulatory factor in the biochemical mechanism leading to neuronal cell death due to acute excitatory toxicity and excitatory toxicity when antagonists inhibit the action of CaMKII. By the death of nerve cells is reduced.
출생 7일 이후의 랫트에 글루타메이트를 1회 피하주사한 후 4시간 후 상기 랫트의 뇌의 피질 및 해마를 관찰한 결과, CaMKⅡ의 발현 수준이 증가된 것으로 관찰된 반면, 상기 글루타메이트 처리 후 오스모틴을 처리한 동물군에서는 CaMKⅡ의 발현 수준이 현저히 감소되는 것으로 확인되었다. 이는 오스모틴이 글루타메이트에 의해 유도된 급성 흥분독성에 대하여 신경을 보호하는 역할을 수행하는 효능이 있음을 의미하는 것이다(도 1a 및 1b).After 4 hours of subcutaneous injection of glutamate in rats after 7 days of birth, the cortex and hippocampus of the rats were observed 4 hours later, whereas the expression level of CaMKII was observed to increase, whereas osmotin was treated after glutamate treatment. The treated animals were found to significantly reduce the expression level of CaMKII. This means that osmotin is effective in protecting the nerve against acute excitatory toxicity induced by glutamate (FIGS. 1A and 1B).
실험예 2: 글루타메이트에 의해 유도된 시냅스 기능 장애를 회복시키는 오스모틴의 효능 검증Experimental Example 2: Validation of Osmotin Efficacy in Restoring Synaptic Dysfunction Induced by Glutamate
글루타메이트에 의해 유도된 흥분독성에 의하여 증가된 CaMKⅡ 및 Ca+2의 농도는 신경 시냅스에 있어 부정적인 영향을 미친다.Increased CaMKII and Ca +2 concentrations due to excitatory toxicity induced by glutamate have a negative effect on neuronal synapses.
시냅스 가소성(plasticity)에 있어서 글루타메이트의 억제 효과 및 오스모틴의 유익한 효과를 분석하기 위하여, 웨스턴 블랏을 이용해 시냅토파이신(synaptophysin)의 발현 수준을 측정하였다. 구체적으로, 출생 7일 이후의 랫트에 글루타메이트를 1회 피하주사한 후 4시간 후 상기 랫트의 뇌의 피질 및 해마를 관찰한 결과, 시냅토파이신이 현저히 감소된 것으로 관찰된 반면, 상기 글루타메이트 처리 후 오스모틴을 처리한 동물군에서는 시냅토파이신의 발현 수준이 증가되는 것으로 확인되었다(도 1c 및 1d). 이는 오스모틴이 뇌의 피질 및 해마에서 글루타메이트에 의해 유도된 시냅스 기능 장애를 회복시키는 효능이 있음을 의미하는 것이다.In order to analyze the inhibitory effect of glutamate and the beneficial effect of osmotin on synaptic plasticity, the expression level of synaptophysin was measured using Western blot. Specifically, 4 hours after the first subcutaneous injection of glutamate in rats after 7 days of birth, the brain cortex and hippocampus of the rats were observed, whereas synaptopycin was significantly reduced, but after the glutamate treatment. Osmotin-treated animals were found to have increased expression levels of synaptophycins (FIGS. 1C and 1D). This means that osmotin is effective in repairing synaptic dysfunction induced by glutamate in the cortex and hippocampus of the brain.
또한, 상기 랫트의 뇌의 피질 및 해마에서의 CREB의 인산화에 대한 글루타메이트의 처리 효과를 분석하기 위하여, 웨스턴 블랏을 이용해 p-CREB 단백질의 발현 수준을 측정하였다. 살린 처리 동물군과 비교하여, 글루타메이트 처리 동물군은 뇌의 피질 및 해마에서 p-CREB 단백질의 발현 수준이 현저히 감소되는 것으로 확인된 반면, 상기 글루타메이트 처리 후 오스모틴을 처리한 동물군의 경우, p-CREB 단백질의 발현이 현저히 증가되는 것으로 관찰되었다(도 1c 및 1d). 이는 오스모틴이 뇌의 피질 및 해마에서 장기 상승 작용을 억제하는 효능이 있음을 의미하는 것이다.In addition, in order to analyze the effect of glutamate on the phosphorylation of CREB in the cortex and hippocampus of the rat brain, Western blot was used to measure the expression level of p-CREB protein. Compared to the saline-treated animals, the glutamate-treated animals were found to significantly reduce the expression level of p-CREB protein in the brain cortex and hippocampus, whereas in the animals treated with osmotin after the glutamate treatment, p A significant increase in the expression of CREB protein was observed (FIGS. 1C and 1D). This means that osmotin has the effect of inhibiting long-term synergy in the cortex and hippocampus of the brain.
실험예 3: 글루타메이트에 의해 유도된 신경 손상 및 DNA 단편화를 억제하는 오스모틴의 효능 검증Experimental Example 3: Verification of the efficacy of osmotin to suppress neuronal damage and DNA fragmentation induced by glutamate
(1) 신경 손상에 대한 오스모틴의 효능(1) Effect of Osmotin on Nerve Damage
글루타메이트에 의해 유도된 흥분독성 손상은 p53-매개의 광범위한 신경 손상을 유도할 수 있다. 이에, 출생 7일 이후의 랫트에 글루타메이트(10 mg/kg)를 1회 피하주사한 후 4시간 후에 신경 손상이 유도되었는지 여부를 측정하기 위하여 상이한 세포 아팝토시스 마커의 수준을 측정하였다.Excitotoxic damage induced by glutamate can lead to a wide range of p53-mediated neuronal damage. Therefore, the levels of different cellular apoptosis markers were measured to determine whether neuronal damage was induced 4 hours after glutamate (10 mg / kg) in rats 7 days after birth.
상기 측정 결과, 글루타메이트가 뇌의 피질 및 해마 모두에서 프로아팝토시스성 p53의 발현 수준, 프로아팝토시스성 Bax 및 항아팝토시스성 Bcl-2의 비율, 미토콘드리아 시토크롬의 세포 기질로의 전달 및 카스파아제-3 및 PARP-1의 활성화를 증가시킴으로써 독성이 유도되는 것으로 관찰되었다.As a result of this measurement, glutamate is expressed in the level of proapoptotic p53, the ratio of proapoptotic Bax and antiapoptotic Bcl-2, the delivery of mitochondrial cytochrome to cell substrate and caspa in both cortex and hippocampus of brain. Toxicity was observed to be induced by increasing the activation of aze-3 and PARP-1.
반면, 글루타메이트 처리 후 오스모틴을 처리한 경우, 글루타메이트에 의해 유도된 상기 변화들이 반대로 유도되었다. 즉, 뇌의 피질 및 해마에서 p53 단백질의 발현 수준 및 Bax/Bcl-2 비율이 감소되었으며, 미토콘드리아 시토크롬 C의 전달 및 카스파아제-3과 PARP-1의 활성화를 감소시켰다(도 2a 내지 2d).On the other hand, when osmotin was treated after glutamate treatment, the changes induced by glutamate were reversed. That is, the expression level of p53 protein and Bax / Bcl-2 ratio were decreased in the cortex and hippocampus of the brain, and reduced the delivery of mitochondrial cytochrome C and activation of caspase-3 and PARP-1 (FIGS. 2A-2D).
(2) DNA 단편화에 대한 오스모틴의 효능(2) Effect of Osmotin on DNA Fragmentation
DNA 손상은 세포 아팝토시스의 전형적 특징 중 하나이다. 이에, 손상된 DNA를 시각화하기 위하여 TUNEL 염색(자유 3' DNA 말단의 효소 표지에 기초한 DNA 절단 분석)을 수행하였다.DNA damage is one of the typical features of cellular apoptosis. Thus, TUNEL staining (DNA cleavage analysis based on enzyme labeling at the free 3 'DNA end) was performed to visualize damaged DNA.
대조군과 비교하여, 글루타메이트를 처리한 경우, 해마의 세 영역인 CA1, CA3 및 DG에서 핵 단편화 및 세포 아팝토시스가 유도된 반면, 글루타메이트 처리 후 오스모틴을 처리한 경우에는 상기 해마의 세 영역에서 핵 단편화 및 세포 아팝토시스(TUNEL-양성 세포가 거의 없음)가 현저히 감소되는 것으로 확인되었다(도 3a 및 3b).Compared with the control group, glutamate treatment resulted in nuclear fragmentation and cellular apoptosis in CA1, CA3, and DG of the hippocampus, whereas osmotin treatment after glutamate treatment resulted in three regions of the hippocampus. Nuclear fragmentation and cellular apoptosis (almost no TUNEL-positive cells) were found to be markedly reduced (FIGS. 3A and 3B).
실험예 4: 글루타메이트에 의해 유도된 신경 손상으로부터, JNK/PI3K/Akt 세포 내 신호 전달 경로를 통해 뇌를 보호하는 오스모틴의 효능 검증Experimental Example 4 Verification of the Efficacy of Osmotin to Protect the Brain through Signal Transduction Pathway in JNK / PI3K / Akt Cells from Glutamate-induced Neuronal Injury
(1) p-JNK의 발현 수준 감소(1) decreased expression level of p-JNK
웨스턴 블랏을 이용하여, 출생 7일 이후의 랫트에 글루타메이트를 단독으로 처리한 동물군 및 상기 글루타메이트 처리 후 오스모틴을 처리한 동물군의 뇌의 피질 및 해마에서의 p-JNK (c-Jun N-말단 키나아제) 발현 수준을 측정하였다.P-JNK (c-Jun N-) in the cerebral cortex and hippocampus of animals treated with glutamate alone and rats treated with osmotin after glutamate treatment in rats after 7 days of birth using Western blot Terminal kinase) expression levels were measured.
상기 측정 결과, 글루타메이트 단독 처리 동물군의 경우 p-JNK의 발현 수준이 현저히 증가된 반면, 글루타메이트 처리 후 오스모틴을 처리한 동물군의 경우 p-JNK의 발현 수준이 현저히 감소되는 것으로 확인되었다(도 4a 및 4b).As a result of the measurement, the expression level of p-JNK was significantly increased in the animal group treated with glutamate alone, whereas the expression level of p-JNK was significantly decreased in the animal group treated with osmotin after glutamate treatment (FIG. 4a and 4b).
또한, 해마의 CA1 영역에서의 p-JNK 발현의 면역 블랏 결과를 측정하기 위하여 면역 형광 염색법을 이용하여 측정한 결과, 상기 웨스턴 블랏의 결과와 일관성있는 결과가 도출되었다. 즉, 글루타메이트 처리에 의하여 해마의 CA1 영역에서 p-JNK의 발현 수준이 증가된 반면, 글루타메이트 처리 후 오스모틴을 처리한 결과, 상기 해마의 CA1 영역에서 p-JNK의 발현 수준이 감소되는 것으로 관찰되었다(도 4c 및 4d).In addition, as a result of measuring the immunoblot of p-JNK expression in the CA1 region of the hippocampus by immunofluorescence staining, a result consistent with that of the western blot was obtained. That is, the expression level of p-JNK in the CA1 region of the hippocampus was increased by glutamate treatment, whereas the osmotin treatment after glutamate treatment decreased the expression level of p-JNK in the CA1 region of the hippocampus. (FIGS. 4C and 4D).
(2) JNK/PI3K/Akt 경로의 관련성(2) Relevance of JNK / PI3K / Akt Pathway
포스포이노시티드(phosphoinositide) 3-키나아제 PI3-K/Akt 경로는 신경 세포에서 중요한 프로-생존 경로 중 하나이다. 이에, 뇌의 피질 및 해마에서 글루타메이트를 처리한 경우, 상기 경로가 관련성이 있는지를 조사하였다.Phosphoinositide 3-kinase PI3-K / Akt pathway is one of the important pro-survival pathways in neurons. Therefore, it was examined whether the pathway was relevant when glutamate was treated in the cortex and hippocampus of the brain.
예상한 바와 같이, 글루타메이트를 처리한 경우, 뇌의 피질 및 해마에서 p-PI3K 및 p-Akt의 발현 수준이 현저히 감소된 반면, 글루타메이트 처리 후 오스모틴을 처리한 경우에는 p-PI3K 및 p-Akt의 발현 수준이 증가되는 것으로 확인되었다(도 4c 및 4d).As expected, glutamate treatment significantly reduced the expression levels of p-PI3K and p-Akt in the cortex and hippocampus of the brain, whereas p-PI3K and p-Akt were treated with osmotin after glutamate treatment. The expression level of was found to be increased (FIGS. 4C and 4D).
또한, 뇌의 해마에서의 p-PI3K의 발현을 면역 형광 염색법을 이용하여 관찰한 결과, 공초점 현미경으로 관찰된 이미지에서 오스모틴을 처리하는 경우, 뇌의 해마의 CA1 영역에서 글루타메이트에 의해 유도되었던 p-PI3K 및 p-Akt 발현 수준의 감소가 회복되는 것으로 확인되었다(도 4c 및 4d).In addition, the expression of p-PI3K in the hippocampus of the brain was observed using immunofluorescence staining. When osmotin was treated in the image observed by confocal microscopy, glutamate was induced in the CA1 region of the hippocampus of the brain. Reductions in p-PI3K and p-Akt expression levels were found to recover (FIGS. 4C and 4D).
실험예 5: 글루타메이트에 의해 유도된 p53, p-Akt 및 카스파아제-3의 분포 변화 및 공동위치화(colocalization)에 대한 오스모틴의 억제 효능 검증Experimental Example 5: Validation of Osmotin's Inhibitory Effect on Distribution Changes and Colocalization of p53, p-Akt and Caspase-3 Induced by Glutamate
상기 실험 결과들에 따른 웨스턴 블랏 결과를 확인하고, p-Akt 및 p53을 통하여 글루타메이트에 의해 유도되는 신경 손상에 대한 오스모틴 신경 보호 효과의 메커니즘을 규명하기 위하여, p-Akt 및 p53의 공동위치화 측정 및 뇌의 해마(CA1) 영역에서의 이들의 분포 변화를 관찰하였다.Co-location of p-Akt and p53 to confirm Western blot results according to the experimental results and to elucidate the mechanism of the osmotin neuroprotective effect on neuronal damage induced by glutamate via p-Akt and p53 Measurements and changes in their distribution in the hippocampus (CA1) region of the brain were observed.
상기 측정 결과, p-Akt는 대부분 p53과 공동위치화되는 것으로 나타났다. 구체적으로, 대조군과 비교할 때, 글루타메이트를 단독으로 처리한 경우에는 뇌의 해마의 CA1 영역에서 p-Akt(레드)의 분포가 억제되고 p53(그린)의 분포가 증가된 반면, 글루타메이트 처리 후 오스모틴을 처리한 경우에는 해마의 CA1 영역에서 글루타메이트에 의해 유도된 p-Akt 및 p53의 분포가 반대의 양상으로 변화하였다(도 5a 및 5b).As a result of the measurement, p-Akt was found to co-locate with p53 mostly. Specifically, when compared to the control group, the treatment of glutamate alone inhibited the distribution of p-Akt (red) and increased the distribution of p53 (green) in the CA1 region of the hippocampus of the brain, whereas osmotin after glutamate treatment In the case of treatment, the distribution of p-Akt and p53 induced by glutamate in the CA1 region of the hippocampus was reversed (Figs. 5A and 5B).
또한, 상기와 같은 방법으로 해마에서의 p53 및 카스파아제-3의 분포 변화를 관찰한 결과, 대조군과 비교하여 글루타메이트를 단독으로 처리한 경우에는 해마에서 p53(그린) 및 카스파아제-3(레드)의 발현 수준이 증가된 것으로 관찰되어, 상기 p53 및 카스파아제-3이 공동위치화되는 것으로 확인되었다. 반면, 글루타메이트 처리 후 오스모틴을 처리한 경우에는 해마에서 p53 및 카스파아제-3의 발현 수준이 감소된 것에 비추어, 역시 이들 단백질의 공동위치화가 확인되었다(도 5c 및 5d).In addition, the distribution of p53 and caspase-3 in the hippocampus was observed in the same manner as described above. When glutamate was treated alone compared to the control group, p53 (green) and caspase-3 (red) in the hippocampus. The expression level of was observed to be increased, confirming that p53 and caspase-3 were co-located. On the other hand, in the case of osmotin treatment after glutamate treatment, co-location of these proteins was also confirmed in light of the decreased expression levels of p53 and caspase-3 in the hippocampus (FIGS. 5C and 5D).
본 명세서는 본 발명의 기술 분야에서 통상의 지식을 가진 자이면 충분히 인식하고 유추할 수 있는 내용은 그 상세한 기재를 생략하였으며, 본 명세서에 기재된 구체적인 예시들 이외에 본 발명의 기술적 사상이나 필수적 구성을 변경하지 않는 범위내에서 보다 다양한 변형이 가능하다. 따라서 본 발명은 본 명세서에서 구체적으로 설명하고 예시한 것과 다른 방식으로 실시될 수 있으며, 이는 본 발명의 기술 분야에 통상의 지식을 가진 자이면 이해할 수 있는 사항이다.In the present specification, those skilled in the art of the present invention can fully recognize and infer the details that have been omitted, and the technical spirit or essential configuration of the present invention in addition to the specific examples described in this specification are changed. Many more variations are possible without departing from the scope of the invention. Therefore, the present invention can be implemented in a manner different from that specifically described and illustrated herein, which can be understood by those skilled in the art.

Claims (8)

  1. 오스모틴(Osmotin)을 유효 성분으로 포함하는, 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환의 예방 또는 치료용 약학 조성물.A pharmaceutical composition for preventing or treating neurological diseases caused by excitatory toxicity or synaptic dysfunction, comprising osmotin as an active ingredient.
  2. 제1항에 있어서, 상기 흥분독성 또는 시냅스 기능 장애는 글루타메이트(Glutamate)에 의해 유도되는 것인, 약학 조성물.The pharmaceutical composition of claim 1, wherein the excitatory or synaptic dysfunction is induced by glutamate.
  3. 제1항에 있어서, 상기 신경 질환의 예방 또는 치료는, AMPA (α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) 글루타메이트 수용체의 발현 감소 또는 AMPA 글루타메이트 수용체의 인산화 감소에 의해 수행되는 것인, 약학 조성물.The method of claim 1, wherein the prevention or treatment of the neurological disease is performed by reducing expression of AMPA (α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) glutamate receptor or decreasing phosphorylation of AMPA glutamate receptor. Will, pharmaceutical composition.
  4. 제1항에 있어서, 상기 신경 질환의 예방 또는 치료는, 시냅토파이신의 발현 증가에 의해 수행되는 것인, 약학 조성물.The pharmaceutical composition of claim 1, wherein the prevention or treatment of the neurological disease is performed by increasing expression of synaptophycin.
  5. 제1항에 있어서, 상기 신경 질환의 예방 또는 치료는, CaMKⅡ (Ca2+/calmodulin-depandent protein kinase Ⅱ)의 발현 감소에 의해 수행되는 것인, 약학 조성물.The pharmaceutical composition of claim 1, wherein the prevention or treatment of neurological disease is performed by decreasing the expression of CaMKII (Ca 2+ / calmodulin-depandent protein kinase II).
  6. 제1항에 있어서, 상기 신경 질환의 예방 또는 치료는, JNK/PI3K/Akt 경로에서 p-JNK의 발현 감소, p-PI3K의 발현 증가 또는 p-Akt의 발현 증가에 의해 수행되는 것인, 약학 조성물.The method of claim 1, wherein the prevention or treatment of the neurological disease is performed by decreasing the expression of p-JNK, increasing the expression of p-PI3K or increasing the expression of p-Akt in the JNK / PI3K / Akt pathway. Composition.
  7. 오스모틴(Osmotin)을 유효 성분으로 포함하는, 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환의 예방 또는 개선용 건강기능식품 조성물.Health functional food composition for the prevention or improvement of neurological diseases caused by excitatory toxicity or synaptic dysfunction comprising osmotin (Osmotin) as an active ingredient.
  8. 오스모틴(Osmotin)을 유효 성분으로 포함하는 약학 조성물을 개체에 투여하는 단계를 포함하는, 흥분독성 또는 시냅스 기능 장애에 의한 신경 질환을 예방 또는 치료하는 방법.A method of preventing or treating neurological diseases caused by excitatory or synaptic dysfunction, comprising administering to a subject a pharmaceutical composition comprising Osmotin as an active ingredient.
PCT/KR2015/000493 2014-01-17 2015-01-16 Composition for preventing or treating neurological disorders caused by excitotoxicity or synaptic dysfunction, containing osmotin, and method for preventing or treating neurological disorders by using same WO2015108372A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2014-0006347 2014-01-17
KR1020140006347A KR20150086429A (en) 2014-01-17 2014-01-17 Composition for preventing or treating neurological disorder induced by excitotoxicity or synaptic dysfunction, and method for preventing or treating neurological disorder using the same

Publications (1)

Publication Number Publication Date
WO2015108372A1 true WO2015108372A1 (en) 2015-07-23

Family

ID=53543192

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2015/000493 WO2015108372A1 (en) 2014-01-17 2015-01-16 Composition for preventing or treating neurological disorders caused by excitotoxicity or synaptic dysfunction, containing osmotin, and method for preventing or treating neurological disorders by using same

Country Status (2)

Country Link
KR (1) KR20150086429A (en)
WO (1) WO2015108372A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017136922A1 (en) * 2016-02-08 2017-08-17 Junaxo, Inc. Use of trehalose for treatment of neurological diseases
CN114342879A (en) * 2022-02-14 2022-04-15 北京端点医药研究开发有限公司 Construction method of animal model of neurodegenerative disease

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20110085260A (en) * 2010-01-19 2011-07-27 경상대학교산학협력단 Composition for preventing and treating neurological disorder comprising osmotin

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20110085260A (en) * 2010-01-19 2011-07-27 경상대학교산학협력단 Composition for preventing and treating neurological disorder comprising osmotin

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
NARASIMHAN, M. L. ET AL.: "Osmotin Is a Homolog of Mammalian Adiponectin and Controls Apoptosis in Yeast through a Homolog of Mammalian Adiponectin Receptor", MOLECULAR CELL, vol. 17, 21 January 2005 (2005-01-21), pages 171 - 180, XP002380322 *
NASEER, M. I.: "Neuroprotective effect against ethanol induced modulation of siRNA-mediated GABA(B) receptor and apoptotic neurodegeneration in the developing rat brain", THESIS OF GYEONGSANG NATIONAL UNIVERSIT Y, 2010 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017136922A1 (en) * 2016-02-08 2017-08-17 Junaxo, Inc. Use of trehalose for treatment of neurological diseases
US10736909B2 (en) 2016-02-08 2020-08-11 Junaxo, Inc. Use of trehalose for treatment of neurological diseases
CN114342879A (en) * 2022-02-14 2022-04-15 北京端点医药研究开发有限公司 Construction method of animal model of neurodegenerative disease

Also Published As

Publication number Publication date
KR20150086429A (en) 2015-07-28

Similar Documents

Publication Publication Date Title
WO2019103203A1 (en) Novel peptide and composition comprising same
WO2017078499A2 (en) Composition for prevention or treatment of neuroinflammatory disease, containing protein tyrosine phosphatase inhibitor
WO2018169282A2 (en) Pharmaceutical composition containing atpif1 for treatment of diabetes
WO2018088862A1 (en) Pharmaceutical composition for preventing or treating neurodegenerative diseases which includes flower extract of daphne genkwa or fractions thereof as active ingredient
WO2020218720A1 (en) Composition for preventing or treating muscular disorders or improving muscular functions, containing leonurus japonicus extract or leonurine
WO2018056706A1 (en) Composition comprising thioredoxin-interacting protein-derived peptide or polynucleotide encoding same as effective ingredient for reverse-ageing of aged stem cell and use thereof
WO2015108372A1 (en) Composition for preventing or treating neurological disorders caused by excitotoxicity or synaptic dysfunction, containing osmotin, and method for preventing or treating neurological disorders by using same
WO2015111832A1 (en) Composition for preventing or treating prostate-related diseases, containing poncirus trifoliate extract
WO2013100718A2 (en) Pharmaceutical composition for preventing or treating inflammatory diseases or asthma, containing lagerstroemia ovalifolia extract or fraction thereof as active ingredient
WO2014042392A1 (en) Composition using metformin for preventing or treating immune diseases including lupus
WO2020122391A1 (en) Composition for preventing or treating cellular senescence-associated diseases, containing homoharringtonine as active ingredient
WO2020116810A1 (en) Pharmaceutical composition, comprising inhibitory peptide against fas signaling, for prevention or treatment of obesity, fatty liver, or steatohepatitis
WO2013022139A1 (en) Method for treatment of diseases caused by accumulation of beta-amyloids in brain parenchyma with low concentration thiazolidinedione derivatives
WO2018106009A1 (en) Composition containing maple tree leaf extract or fractions thereof for preventing or treating inflammatory ocular diseases
WO2021033995A1 (en) Composition comprising amomum tsaoko extract for prevention, alleviation, or treatment of sarcopenia-related disease
WO2017105081A1 (en) Pharmaceutical composition for treating lung diseases or alleviating symptoms of lung diseases
WO2020189911A1 (en) Composition including noranhydroicaritin, for preventing or treating cardiovascular metabolic diseases
WO2015072667A1 (en) Pharmaceutical composition for preventing or treating degenerative cranial nerve diseases
WO2020122392A1 (en) Composition for preventing or treating cellular senescence-related diseases comprising zotarolimus as active ingredient
WO2020122393A1 (en) Composition for preventing or treating cellular senescence-related diseases comprising salinomycin as effective component
WO2020106048A1 (en) Pharmaceutical composition for prevention or treatment of neurodegenerative disease
WO2015102205A1 (en) Pharmaceutical composition for preventing or treating cancer, containing proteasome inhibitor and loperamide as active ingredients
WO2019198982A1 (en) Composition for preventing or improving skeletal muscle atrophy containing kukoamine a and kukoamine b as active ingredients
WO2018143615A1 (en) Pharmaceutical composition containing peroxiredoxin 1 protein activity inhibitor as active ingredient for preventing or treating bone disease
WO2019098568A2 (en) Composition for alleviating, preventing or treating pain comprising camellia japonica extract

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: 15736971

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: 15736971

Country of ref document: EP

Kind code of ref document: A1