WO2019212300A1 - Composition comprising aster koraiensis nakai extract or fraction thereof as effective ingredient for prevention or treatment of parkinson's disease - Google Patents

Composition comprising aster koraiensis nakai extract or fraction thereof as effective ingredient for prevention or treatment of parkinson's disease Download PDF

Info

Publication number
WO2019212300A1
WO2019212300A1 PCT/KR2019/005352 KR2019005352W WO2019212300A1 WO 2019212300 A1 WO2019212300 A1 WO 2019212300A1 KR 2019005352 W KR2019005352 W KR 2019005352W WO 2019212300 A1 WO2019212300 A1 WO 2019212300A1
Authority
WO
WIPO (PCT)
Prior art keywords
disease
parkinson
extract
bee
protein
Prior art date
Application number
PCT/KR2019/005352
Other languages
French (fr)
Korean (ko)
Inventor
양현옥
박정윤
장리준
이주영
정상훈
천윤선
Original Assignee
한국과학기술연구원
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 한국과학기술연구원 filed Critical 한국과학기술연구원
Publication of WO2019212300A1 publication Critical patent/WO2019212300A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23KFODDER
    • A23K10/00Animal feeding-stuffs
    • A23K10/30Animal feeding-stuffs from material of plant origin, e.g. roots, seeds or hay; from material of fungal origin, e.g. mushrooms
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • A23L33/105Plant extracts, their artificial duplicates or their derivatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/28Asteraceae or Compositae (Aster or Sunflower family), e.g. chamomile, feverfew, yarrow or echinacea
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2002/00Food compositions, function of food ingredients or processes for food or foodstuffs
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2200/00Function of food ingredients
    • A23V2200/30Foods, ingredients or supplements having a functional effect on health
    • A23V2200/322Foods, ingredients or supplements having a functional effect on health having an effect on the health of the nervous system or on mental function

Definitions

  • the present invention relates to a composition for the prevention or treatment of Parkinson's disease, which comprises the bee herb extract or fractions thereof as an active ingredient.
  • Degenerative brain diseases are disorders in which motor, memory, and cognitive disorders are caused by neurodegenerative diseases caused by various causes.
  • Parkinson's disease is the most common neurodegenerative brain disease after Alzheimer's disease. .
  • Parkinson's disease was first described in 1817 by James Parkinson, a British medical doctor, about 60 years later, the term Parkinson's was used by J.Charout, and by Kinner wilson in 1912, Parkinson's disease It was found that the disease is related to the extrapyramidal system and not the pyramidal system (Habibi et al., 2011).
  • Parkinson's disease is classified as a progressive neurological disorder intractable disease without effective and effective long-term treatment (Pan et al., 2008, Habibi et al., 2011).
  • Parkinson's disease is caused by various pathological factors occurring in Parkinson's disease patients.
  • the abnormal killing of dopamine-releasing neurons among the neurotransmitters distributed in the melanoma of the midbrain is disrupted or destroyed by the substantia nigra par compacta (Camins et al., 2008, Smith). , 2008).
  • Abnormal death of dopamine neurons occurs through a series of processes.
  • mitochondria damaged by various factors such as the environment, heredity, and aging, cause functional degradation of the proteolytic system, which causes abnormal proteins and organelles. It is caused by accumulation and not decomposition. These causes raise the importance of proteolytic systems in the treatment and prevention of Parkinson's disease.
  • L-dopa is a treatment that increases the level of dopamine in the blood by direct injection of Levodopa, a precursor of dopamine neurons.
  • Direct infusion of Levodopa results in substantial symptomatic relief in Parkinson's patients, but increased doses are essential for long-term use and are known to cause a variety of unforeseen side effects as the dose increases (Rock and Peterson, 2006). , McGeer and McGeer, 2008). Therefore, it is urgent to develop more fundamental treatments and drugs that can suppress the protection and death of nerve cells that facilitate dopamine secretion, rather than the direct administration of L-dopa, which increases dopamine blood levels. As one of these attempts, research into the development of therapeutic agents targeting autophagy, one of the proteolytic systems, has recently attracted attention.
  • Autophagy or autophagy, is derived from the Greek word for "self,” meaning “self,” and “eating,” meaning to eat. It not only maintains the metabolic balance of the cell but also affects the fate of the cell. It is known as a mechanism of self-sacrifice that plays an important role (Eisenberg-Lerner et al., 2009).
  • This autophagy acts as a precursor to autophagosomes and is a morphological form of autophagosomes, which are independent bilayers, which gradually extend the edges of cells to completely surround unwanted proteins in the cell, forming vesicles. It has a characteristic structure.
  • autophagosomes move along microtubules to fuse with lysosomes in cells and form autolysosomes, which are reported to degrade proteins by proteolytic enzymes secreted inside lysosomes. It became.
  • Aster koraiensis Nakai is a compositae plant, also called Gymnaster koraiensis (Nakai) Kitamura, and is a native plant of Korea.
  • Beetle odor is a perennial herb distributed mainly in Jeju Island and sub-Gyeonggi province, and is known to grow well in valleys and wetlands with high humidity.
  • the present inventors are conducting research on the development of a prophylactic and therapeutic agent for Parkinson's disease through induction of autophagy
  • the extract of Aster koraiensis Nakai plants induces autophagy and at the same time prevents and prevents Parkinson's disease.
  • the present invention was completed by confirming that it is very effective for treatment.
  • An object of the present invention is to provide a pharmaceutical composition for the prevention or treatment of Parkinson's disease comprising a bee odor extract or fractions thereof as an active ingredient.
  • Another object of the present invention is to provide a method for preventing or treating Parkinson's disease, comprising administering the pharmaceutical composition to a subject.
  • Still another object of the present invention is to provide a food composition for preventing or improving Parkinson's disease, which comprises a bee anesthetist extract or a fraction thereof as an active ingredient.
  • Still another object of the present invention is to provide a feed composition for preventing or improving Parkinson's disease, which comprises a bee anesthetist extract or a fraction thereof as an active ingredient.
  • Still another object of the present invention is to provide a composition for inhibiting brain cell death by in vitro autophagy inducing effect, which comprises a bee stinger extract or a fraction thereof as an active ingredient.
  • Beetle odor extract of the present invention increases the expression of LC3 protein, a membrane protein of autophagosome, induces autophagy through the increase of phosphorylated AMPK and UK1 which induces autophagy, and the reduction of mTOR that inhibits autophagy. Therefore, the composition including the same can be effectively used for the prevention or treatment of Parkinson's disease through induction of autophagy action.
  • Figure 1 shows the results confirmed by Western blotting to increase the expression of the autophagy membrane protein LC3 expression in accordance with the concentration of the bee odor extract in SH-SY5Y cell line (Control: control). ** indicates P ⁇ 0.01 for control and *** indicates P ⁇ 0.001.
  • Figure 2 is a result confirmed by Western blotting to increase the expression of the phosphorylated AMPK protein according to the concentration of the bee hijab extract (Control: control). *** indicates P ⁇ 0.001 for the control.
  • Figure 3 is the result confirmed by Western blotting to increase the expression of phosphorylated ULK1 protein expression according to the concentration of the bee sting odor extract (Control: control). *** indicates P ⁇ 0.001 for control
  • Figure 4 is a result confirmed by Western blotting to reduce the expression of mTOR protein expression according to the concentration of the bee sting odor extract (Control: control). * Indicates P ⁇ 0.05 for control and *** indicates P ⁇ 0.001.
  • Figure 6 shows the results confirmed by Western blotting to increase the expression of the autophagy membrane protein LC3 expression according to the bee odor fraction in SH-SY5Y cell line (Control: control).
  • Figure 7 is a result confirmed by Western blotting to reduce the amount of mTOR protein expression according to the addition concentration of the honey bee odor fraction (Control: control). ** indicates P ⁇ 0.01 for control and *** indicates P ⁇ 0.001.
  • FIG. 8 shows the effect of Western blotting on the expression of Tyrosine hydroxylase (TH) protein according to bee hives extracts and fractions in substantia nigra of C57BL / 6 mice induced Parkinson's disease by MPTP.
  • the result was confirmed (Control: control; MPTP: MPTP-treated group; Ropinirole: ropinirole + MPTP-treated group).
  • *** indicates P ⁇ 0.001 compared to the control group, ## indicates P ⁇ 0.01 for the MPTP treatment group and ### indicates P ⁇ 0.001 for the MPTP treatment group.
  • FIG. 11 shows the results of Western blotting confirming the effect of increasing the amount of phosphorylated AMPK protein expression according to bee hives extracts and fractions in substantia nigra of C57BL / 6 mice induced Parkinson's disease by MPTP.
  • Control control; MPTP: MPTP-only group; rofinirol: rofiniro + MPTP-treated group).
  • # Indicates P ⁇ 0.05 for MPTP alone treatment group.
  • FIG. 12 shows Western blotting effect of increased phosphorylated ULK1 (s555) protein expression levels according to bee hives extract and fraction administration in substantia nigra of C57BL / 6 mice induced Parkinson's disease by MPTP administration. This was confirmed by (Control: control group; MPTP: MPTP treatment group; ropinillol: ropinillol + MPTP treatment group). * Indicates P ⁇ 0.05 compared to the control, ## indicates P ⁇ 0.01 for the MPTP-only group and ### indicates P ⁇ 0.001 for the MPTP-only group.
  • FIG. 13 shows the results of Western blotting confirming the reduction effect of mTOR protein expression according to bee hives extract and fraction administration in substantia nigra of C57BL / 6 mice induced Parkinson's disease by MPTP administration.
  • Control control; MPTP: MPTP alone group; ropinirol: rofiniro + MPTP treated group).
  • One aspect of the present invention for achieving the above object provides a pharmaceutical composition for the prevention or treatment of Parkinson's disease, including Aster koraiensis Nakai extract or a fraction thereof as an active ingredient.
  • the present invention provides a preventive or therapeutic use of Parkinson's disease of the composition comprising an extract of Aster koraiensis Nakai or fractions thereof as an active ingredient.
  • beetle odor is scientific name Aster koraiensis Nakai, but in the present invention can be used to extract the above-ground parts of the plant, more specifically, the leaves and stems of the plant can be used, preventing or treating Parkinson's disease As long as it has an effect, it is not specifically limited to this.
  • the hummingbird odor can be purchased commercially, or may be used collected or cultivated in nature.
  • extract refers to an extract, such as an extract obtained by extracting a bee odor, a diluent or concentrate of the extract, a dried product obtained by drying the extract, a modifier or purified product of the extract, or a mixture thereof. It includes extracts of all formulations that can be formed using extracts.
  • the method of extracting the hummingbird odor is not particularly limited, and may be extracted according to a method commonly used in the art.
  • Non-limiting examples of the extraction method may include hot water extraction, ultrasonic extraction, filtration, reflux extraction, etc., these may be carried out alone or in combination of two or more methods.
  • the type of extraction solvent used to extract the bee odor is not particularly limited, and any solvent known in the art may be used.
  • Non-limiting examples of the extraction solvent may include water, alcohols of C1 to C4, mixed solvents thereof, and the like, which may be used alone or in combination of one or more thereof. Specifically, preferably 95% ethanol may be used, but is not limited thereto.
  • fraction refers to the result obtained by performing fractionation to separate a specific component or a specific group of components from a mixture comprising several various components.
  • the fractionation method of obtaining the fraction in the present invention is not particularly limited, and may be performed according to a method commonly used in the art.
  • Non-limiting examples of the fractionation method include a solvent fractionation method performed by treating various solvents, an ultrafiltration fractionation method performed through an ultrafiltration membrane having a constant molecular weight cut-off value, and various chromatography (size, charge, hydrophobicity). Or chromatographed fractions), and combinations thereof, which are prepared for separation according to affinity.
  • a method of obtaining a fraction from the extract by treating a predetermined solvent with an extract obtained by extracting the bee sting odor of the present invention are prepared for separation according to affinity.
  • the kind of the fractionation solvent used to obtain the fraction in the present invention is not particularly limited, and any solvent known in the art may be used.
  • Non-limiting examples of the fractionation solvents include polar solvents such as water and alcohols having 1 to 4 carbon atoms; Nonpolar solvents such as hexane, ethyl acetate, chloroform and dichloromethane; Or a mixed solvent thereof. These may be used alone or in combination of one or more thereof, but is not limited thereto.
  • hexane, ethyl acetate, butanol or water may be used alone or in combination of one or more thereof, but is not limited thereto. It doesn't happen.
  • extract or fraction may be prepared and used in the form of a dry powder after extraction, but is not limited thereto.
  • Parkinson's disease means a degenerative brain disease of the nervous system caused by the loss of dopamine neurons. Stability, stiffness, slowness of movement (slowness of motion) and postural instability are characteristic, and clinical symptoms usually begin after age 60.
  • prevention means any action that inhibits or delays the progression of Parkinson's disease by the administration of a pharmaceutical composition of the present invention comprising a cane odor extract or a fraction thereof.
  • treatment means any action in which Parkinson's disease is ameliorated or beneficially altered by administration of the pharmaceutical composition.
  • the "pharmaceutical composition” of the present invention may further comprise a pharmaceutically acceptable carrier, excipient or diluent commonly used in the manufacture of a pharmaceutical composition, which carrier is a non-naturally occuring carrier. It may include.
  • the pharmaceutical composition may be used in the form of powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols and the like, oral formulations, external preparations, suppositories, and sterile injectable solutions, respectively, according to conventional methods. .
  • “Pharmaceutically acceptable” means to exhibit properties that are not toxic to cells or humans exposed to the composition.
  • the type of the carrier is not particularly limited, and any carrier can be used as long as it is a conventionally used and pharmaceutically acceptable carrier in the art.
  • Non-limiting examples of the carrier include saline, sterile water, Ringer's solution, buffered saline, albumin injection solution, dextrose solution, maltodextrin solution, glycerol, ethanol and the like. These may be used alone or in combination of two or more thereof.
  • ком ⁇ онентs such as antioxidants, buffers and / or bacteriostatic agents may be added and used, and diluents, dispersants, surfactants, binders, lubricants, and the like may be additionally added to provide a solution such as an aqueous solution, a suspension, an emulsion, or the like. It may be formulated into a use formulation, pills, capsules, granules or tablets.
  • the mode of administration of the pharmaceutical composition for preventing or treating Parkinson's disease according to the present invention is not particularly limited and may be in accordance with a method commonly used in the art.
  • the composition may be administered by oral or parenteral administration.
  • the composition for preventing or treating Parkinson's disease of the present invention can be prepared in various formulations according to the desired mode of administration.
  • Another aspect of the invention provides a method of preventing or treating Parkinson's disease comprising administering the pharmaceutical composition to a subject.
  • Parkinson's disease prevention and treatment is as described above.
  • the term "administration” means the introduction of certain substances into an individual in a suitable manner.
  • the term "individual” means all animals, such as rats, mice, and livestock, including humans who may or may develop Parkinson's disease. As a specific example, it may be a mammal including a human.
  • the "individual” may include a companion animal.
  • the companion animal refers to an animal that lives together with a human, and includes, but is not limited to, a mammal such as a dog, a cat, a hamster, and a guinea pig, a bird such as a parrot, a canary, and the like.
  • the method for preventing or treating Parkinson's disease of the present invention may include administering to the individual a pharmaceutically effective amount of a medicinal composition comprising a bee herb extract or a fraction thereof.
  • the "pharmaceutically effective amount” means an amount sufficient to treat the disease at a reasonable benefit / risk ratio applicable to the medical treatment and not causing side effects, and the effective dose level refers to the sex, age, weight, Well-known in the field and other medical fields, including health status, type of disease, severity, drug activity, drug sensitivity, method of administration, time of administration, route of administration, and rate of release, duration of treatment, combination or drug used simultaneously Depending on the factor, it can be readily determined by one skilled in the art.
  • the pharmaceutical composition of the present invention may be administered at 0.0001 to 500 mg / kg body weight per day, more specifically at 0.01 to 500 mg / kg body weight, based on solids. Dosing may include administering the recommended dose once a day or in several divided doses.
  • 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. It may also be single or multiple doses. Taking all of these factors into consideration it is important to administer an amount that will achieve the maximum effect in a minimum amount without causing side effects, which can be readily determined by one skilled in the art.
  • the route of administration and the mode of administration of the composition are not particularly limited, and any route of administration can be reached as long as the composition comprising the composition can be reached at the desired site.
  • the composition may be administered through various routes, oral or parenteral, and non-limiting examples of the route of administration include oral, rectal, topical, intravenous, intraperitoneal, intramuscular, intraarterial, transdermal, nasal What is administered through intralateral or inhalation etc. are mentioned.
  • Yet another aspect of the present invention provides a food composition for preventing or improving Parkinson's disease, comprising a bee odorous extract or a fraction thereof as an active ingredient.
  • the term " improvement" means any action that at least reduces the parameters associated with the condition to be treated with the administration of the compositions of the present invention, eg the extent of symptoms.
  • the term "food” of the present invention meat, sausage, bread, chocolate, candy, snacks, confectionery, pizza, ramen, other noodles, gum, dairy products including ice cream, various soups, drinks, tea, drinks, alcoholic beverages , Vitamin complexes, nutraceuticals and health foods, and includes all foods in the usual sense.
  • the food composition of the present invention can be consumed on a daily basis, a high Parkinson's disease improvement effect can be expected, and thus can be very useful for health promotion purposes.
  • the functional food is the same term as a food for special health use (Food for special health use, FoSHU), in addition to the nutritional supply, the processed food, so that the bioregulatory function appears efficiently, high medical effect Means food.
  • Food for special health use Food for special health use, FoSHU
  • the term 'function (sex)' refers to obtaining a useful effect for health purposes such as nutrient control or physiological action on the structure and function of the human body.
  • the food of the present invention may be prepared by a method commonly used in the art, and the preparation may be performed by adding raw materials and ingredients commonly added in the art.
  • the formulation of the food may be prepared without limitation as long as the formulation is recognized as a food.
  • Food composition of the present invention can be prepared in a variety of dosage forms, unlike the general medicine has the advantage that there is no side effect that can occur when taking a long-term use of the drug as a natural product, and because the portability is excellent,
  • the food of the invention can be taken as an adjuvant for enhancing the Parkinson's disease improving effect.
  • the health food refers to foods having active health maintenance or promotion effect as compared to general foods
  • the health supplement food refers to foods for health supplementation purposes.
  • nutraceutical, health food, dietary supplement are used interchangeably.
  • the health functional food is a food prepared by adding the extract of the present invention to food materials such as beverages, teas, spices, gums, confections, or the like, encapsulated, powdered, suspensions, etc.
  • food materials such as beverages, teas, spices, gums, confections, or the like, encapsulated, powdered, suspensions, etc.
  • the food composition may further include a physiologically acceptable carrier, and the type of carrier is not particularly limited and may be any carrier that is commonly used in the art.
  • the food composition may include additional ingredients that are commonly used in food compositions to improve the smell, taste, time and the like.
  • additional ingredients may include vitamins A, C, D, E, B1, B2, B6, B12, niacin, biotin, folate, panthotenic acid, and the like.
  • minerals such as zinc (Zn), iron (Fe), calcium (Ca), chromium (Cr), magnesium (Mg), manganese (Mn), copper (Cu) and chromium (Cr); And amino acids such as lysine, tryptophan, cysteine, valine and the like.
  • the food composition is a preservative (potassium sorbate, sodium benzoate, salicylic acid, sodium dehydroacetic acid, etc.), fungicides (bleaching powder and highly bleaching powder, sodium hypochlorite, etc.), antioxidants (butylhydroxyanisol (BHA), butylhydride) Oxytoluene (BHT), etc.), colorants (such as tar pigments), colorants (sodium nitrite, sodium nitrite, etc.), bleach (sodium sulfite), seasonings (such as MSG glutamate), sweeteners (ducin, cyclate, saccharin Foods such as sodium, etc.), fragrances (vanillin, lactones, etc.), swelling agents (alum, potassium D-tartrate, etc.), reinforcing agents, emulsifiers, thickeners (foils), coatings, gum herbicides, foam inhibitors, solvents, modifiers, etc. It may include food additives.
  • the additive may
  • Another aspect of the present invention provides a feed composition for the prevention or improvement of Parkinson's disease comprising a bee anesthetized extract or a fraction thereof as an active ingredient.
  • composition comprising the honey bee odor extract or fractions thereof of the present invention can be used for the prevention or treatment of the development of Parkinson's disease in individuals other than humans, such as livestock or companion animals, can be utilized as a functional feed additive, or a feed composition.
  • the content of the bee-flavored extract or fractions thereof in the feed composition according to the present invention can be appropriately adjusted according to the type and age of the livestock, the type of application, the desired effect, and the like, for example, 1 to 99% by weight, preferably 10 to 90% by weight. %, More preferably 20 to 80% by weight, but is not limited thereto.
  • diluents, dispersants, surfactants, binders or lubricants may be additionally added to formulate injectable formulations, capsules, granules or tablets such as aqueous solutions, suspensions, emulsions and the like.
  • the feed composition of the present invention is a supplementary ingredient such as amino acids, inorganic salts, vitamins, antioxidants, antifungal agents, antibacterial agents, and vegetable protein feed such as milled or crushed wheat, barley, corn, blood meal, meat meal, fish meal as auxiliary ingredients.
  • a supplementary ingredient such as amino acids, inorganic salts, vitamins, antioxidants, antifungal agents, antibacterial agents, and vegetable protein feed such as milled or crushed wheat, barley, corn, blood meal, meat meal, fish meal as auxiliary ingredients.
  • animal protein feed animal fat and vegetable fat, such as can be used with nutritional supplements, growth promoters, digestive absorption accelerators, disease prevention agents.
  • the feed composition of the present invention When the feed composition of the present invention is used as a feed additive, the feed composition may be added as it is or used together with other ingredients, and may be suitably used according to a conventional method.
  • Dosage forms of the feed composition may be prepared in immediate release or sustained release formulation in combination with a nontoxic pharmaceutically acceptable carrier.
  • a nontoxic pharmaceutically acceptable carrier may be corn starch, lactose, sucrose, propylene glycol.
  • a solid carrier it may be a dosage form such as a tablet, a powder, a torokee agent, or the like, and in the case of a liquid carrier, it may be a dosage form of a syrup, a liquid suspension, an emulsion, or a solution.
  • the dosage may contain preservatives, lubricants, solution promoters, stabilizers, and may also contain other inflammatory disease ameliorating agents and substances useful for virus prevention.
  • the feed composition of the present invention is applicable to a number of animal diets, ie feeds, including mammals, poultry, fish and shellfish.
  • animal diets including mammals, poultry, fish and shellfish.
  • Commercially important mammals such as pigs, cows and goats, zoo animals such as elephants and camels, and livestock such as dogs and cats.
  • Commercially important poultry includes chickens, ducks, geese and the like, and may include commercially raised fish and crustaceans such as trout and shrimp.
  • the feed composition according to the present invention may be mixed in a livestock feed in an amount of about 10 to 500 g, preferably 10 to 100 g per kg, on a dry weight basis, and then thoroughly mixed and then fed into a mesh or further processing. Palletization, expansion, extrusion through the process is preferred.
  • the composition of the present invention increases the protein expression level of phosphorylated ULK1 and inhibits mTOR protein expression by increasing the protein expression level of phosphorylated AMPK in dopaminergic cell lines, and autophagy in an animal model induced by Parkinson's disease through MPTP. Through the induction of the brain cell death can be effectively suppressed, it was confirmed that it has an excellent effect on the prevention, treatment and improvement of Parkinson's disease.
  • the bee anesthetized extract of the present invention has an excellent effect of inducing autophagy and promoting the degradation of alpha-synuclein protein, which is known as the ultimate cause of Parkinson's disease, thereby preventing or treating Parkinson's disease. It was confirmed that the composition can be effectively used for improvement.
  • Beetle odor was cultivated in Bukpyeong-myeon, Jeongseon-gu, Gangwon-do, and washed and dried completely to obtain 15 kg of dry weight. The sample was crushed in a grinder and extracted with 95% ethanol twice under reduced pressure for 2 hours at 60 ⁇ 70 °C. kg was obtained.
  • Example 1-2 Effect of Increasing the Expression Level of LC3 Protein by the Beetle Odor Extract
  • the dopaminergic cell line SH-SY5Y cell line was 10% heat-inactivated fetal bovine serum of Gibco (Grand Island, NY, USA) Dulbecco's modified eagle medium nutrient mixture F-12 (Ham) 1 ⁇ containing 1% penicillin / streptomycin (hereinafter referred to as P / S, Gibco); Cultured in DMEM / F12 (Gibco) medium was used.
  • This cell line was purchased from ATCC: The Global Bioresource Center (CRL-2266 TM) (Manassas, Virgina 20108, USA).
  • the bee-flavored extract obtained in Example 1 was added to the cell line (SH-SY5Y) in the amount shown in FIG. 1, then incubated at 5% CO 2 , 37 ° C. for 24 hours, and collected for 6 minutes at 13,000 rpm. Centrifugation was washed twice with Dulbecco's phosphate buffered saline (DPBS). After lysis solution (1X lysis buffer, 1X Protease inhibitor cocktail, 1x Phenylmethyl sulfonyl fluoride) was added to the obtained cells, the cells were reacted at 4 ° C for 30 minutes, and then completely lysed while shaking at the same 4 ° C for 30 minutes. Centrifugation was performed at 13,000 rpm for 20 minutes to obtain the supernatant from which the protein was released. The supernatant was quantified by Bradford method using Protein assay dye Reagent concentrate (Bio-Rad).
  • Example 1-3 Effect of Increasing Phosphorylated AMPK Protein Expression by Beetle Anesthetic Extract
  • Example 2 In order to measure the effect of increasing the expression level of phosphorylated AMPK protein of the bee-flavored extract obtained in Example 1, it was added to the cultured SH-SY5Y cell line in the amount of the bee-flavored extract described in Figure 2, at 5% CO 2 , 37 °C Incubated for 24 hours, and experimented in the same manner as in Example 2.
  • Primary antibodies used in this experiment are AMPK, p-AMPK, GAPDH (Cell signaling technology), GAPDH was used as a loading control.
  • Example 1-4 Effect of Increasing Phosphorylated ULK1 Protein Expression by Beetle Odor Extract
  • Example 1-5 Effect of Increasing Expression of mTOR Protein by Beetle Odor Extract
  • Example 1-6 Effect of Induction of Autophagy Action on Beetle Anesthetic Extracts Through Autophagy Inhibitor Bafilomycin A
  • Example 2 In order to confirm whether the effect of inducing autophagy action of the bee-flavored extract was a non-specific result of false positive or true autophagy induction effect, the cultured SH-SY5Y cell line was obtained in Example 1 Beetle odor extract was treated for 18 hours at 50 ⁇ g / ml concentration as described in Figure 5 and then treated with 100nM bafilomycin A1 (hereinafter, baf A1) and incubated for 6 hours. Thereafter, the amount of LC3 protein expression was measured by Western blotting in the same manner as in Example 2.
  • bafilomycin A1 hereinafter, baf A1
  • Baf A1 is an autophagy inhibitor that inhibits the fusion of autophagosomes and lysosomes, thereby inhibiting the maturation of autophagic vacuoles. Specifically, autophagy flux occurs only at the last stage of the autophagy route, but autophagy induction occurs. However, when baf A1 is treated, LC3 protein expression is increased as autophagy flux is suppressed.
  • the bee-flavored fraction was added to the cell line (SH-SY5Y cell) at 5, 10, 25, 50, and 100 ⁇ g / ml depending on the fraction toxicity. After the addition, 5% CO 2 , and incubated for 24 hours at 37 °C, it was confirmed through the Western blotting experiment of the same method as Example 2.
  • Example 7 In order to determine the effect of reducing mTOR protein expression of the hummingbird BuOH fraction obtained in Example 7 at all, the hummingbird fraction was added to the SH-SY5Y cell line as described in FIG. 7, followed by 24% at 5% CO 2 , 37 ° C. The culture was carried out in the same manner as in Example 2. Primary antibodies used in this experiment are mTOR, GAPDH (Cell signaling technology), GAPDH was used as a loading control.
  • mice C57BL / 6 male mice (8 weeks old) were used and feed and water were freely ingested. Mice were divided into seven groups, six in each group.
  • the control group and the MPTP-treated group were orally administered with saline once 3 days, the bee odor extract (200 mg / kg) group obtained in Example 1-1 and the bee odor fraction obtained in Example 2-1 (25, 50, 100 mg / kg) group was dissolved orally in each saline extract and fractions and administered once orally for 3 days.
  • rofinirol was dissolved in physiological saline and administered once orally for 3 days.
  • MPTP (30 mg / kg) dissolved in physiological saline was administered orally to all groups except the control group 1 hour after drug administration, and to all groups except the control group on the 9th to 10th day of drug administration. The same amount of drug was administered orally once.
  • mice in each group were killed and brain tissues were separated into substantia nigra and striatum.
  • the isolated brain tissue was homogenized with PRO-PREPTM Lysis Buffer (iNtRON, Gyeonggi, Korea) supplemented with Phosphatase Inhibitor Cocktail Set I (Sigma-Aldrich, MO, USA).
  • the homogenate was left at 4 ° C. for 30 minutes and then the supernatant was collected by centrifugation at 13,000 rpm, 4 ° C. for 30 minutes, and the supernatant was quantitated by Bradford method using Protein assay dye Reagent concentrate (Bio-Rad). It was.
  • Example 1-2 The mixture was mixed with a predetermined amount of SDS loading buffer and heated at 99 ° C. for 5 minutes, and protein expression was measured by Western blotting in the same manner as in Example 1-2.
  • Primary antibodies used in this experiment include TH, Beclin-1, p-mTOR, Mtor, p-ULK1 (s555), ULK1, p-AMPK, AMPK, LC3, ⁇ -synuclein, Cell signaling technology (GAPDH). GAPDH was used as loading control.

Abstract

The present invention relates to a pharmaceutical composition comprising an Aster koraiensis Nakai extract or a fraction thereof as an effective ingredient for prevention or treatment of Parkinson's disease, a method for prevention or treatment of Parkinson's disease by using the pharmaceutical composition, and a food composition and an animal feed composition for prevention or alleviation of Parkinson's disease. An Aster koraiensis Nakai extract or a fraction thereof according to the present invention is characterized by specifically increasing the quantity of a protein involved in the autophagy preventive of the death of dopamine neurons, which is a cause of Parkinson's disease. Particularly, the Aster koraiensis Nakai extract or a fraction thereof exhibits excellent effects of upregulating the expression of LC protein, which is a membrane protein of an autophagosome, and the expression of p-AMPK and p-ULK1 proteins, which induce autophagy, and remarkably downregulating the expression of mTOR protein, which inhibits autophagy, thereby finding advantageous applications in the prevention or treatment of Parkinson's disease. In addition, the composition of the present invention is derived from a natural material and thus can be safely used for the prevention, treatment, and alleviation of Parkinson's disease, without side effects.

Description

벌개미취 추출물 또는 이의 분획물을 유효성분으로 포함하는 파킨슨병의 예방 또는 치료용 조성물Parkinson's disease preventive or therapeutic composition comprising a bee odor extract or fractions thereof as an active ingredient
본 발명은 벌개미취 추출물 또는 이의 분획물을 유효성분으로 하는 파킨슨병의 예방 또는 치료용 조성물에 관한 것이다.The present invention relates to a composition for the prevention or treatment of Parkinson's disease, which comprises the bee herb extract or fractions thereof as an active ingredient.
최근 노인 인구 비율이 점차 높아져 가는 고령화 현상이 사회적 문제로 대두됨에 따라, 퇴행성 뇌질환(neurodegenerative disorders)에 대한 관심이 높아지고 있다. 퇴행성 뇌질환이란 다양한 원인에 의해 퇴행성으로 변한 신경 때문에 운동 작용, 기억, 인지 작용 등에 장애가 발생하는 질환이며, 그 중에서 파킨슨병(Parkinson's disease)은 알츠하이머 질환 다음으로 가장 발병률이 높은 대표적인 신경 퇴행성 뇌 질환이다. 파킨슨병은 1817년 영국 의학자인 James Parkinson이 "흔들림성 마비"를 처음으로 기술하였으며, 그로부터 60여년 후에 J.Charout에 의해 파킨슨병이라는 용어가 사용되었고, 1912년 Kinner wilson에 의해 파킨슨병이 척수의 중추 경로 중 추체계(pyramidal system)가 아닌 추체외로계(extrapyramidal system) 관련 질환임을 알게 되었다(Habibi et al., 2011).As the aging phenomena, which are increasing in proportion to the elderly population, have recently emerged as a social problem, there is a growing interest in neurodegenerative disorders. Degenerative brain diseases are disorders in which motor, memory, and cognitive disorders are caused by neurodegenerative diseases caused by various causes. Among them, Parkinson's disease is the most common neurodegenerative brain disease after Alzheimer's disease. . Parkinson's disease was first described in 1817 by James Parkinson, a British medical doctor, about 60 years later, the term Parkinson's was used by J.Charout, and by Kinner wilson in 1912, Parkinson's disease It was found that the disease is related to the extrapyramidal system and not the pyramidal system (Habibi et al., 2011).
파킨슨병의 발병 원인의 규명을 위한 연구가 수십년에 걸쳐 진행되어 왔으나, 그 기전에 대해 정확하게 밝혀진 바가 없다. 그로 인해 파킨슨병은 유효적이고 효과적인 장기 치료방법이 없는 점진적인 신경 장애 난치병으로 분류된다(Pan et al., 2008, Habibi et al., 2011). 다만 파킨슨병 환자에게 발생되는 다양한 병리학적인 요인에 의해서 파킨슨병이 발병되는 것으로 추측되고 있다. 그 요인으로서, 중뇌의 흑색질(substantia nigra par compacta)이 파괴되거나 손상되면서 중뇌의 흑색질에 분포하는 신경전달물질 중 도파민을 분비하는 신경세포의 비정상적인 사멸이 가장 대표적이다(Camins et al., 2008, Smith, 2008). 도파민 신경세포의 비정상적인 사멸은 일련의 과정을 통해 일어나는데, 먼저 환경, 유전, 노화와 같은 다양한 요인에 의해 손상을 입은 미토콘드리아가 단백질 분해 시스템의 기능 장애를 야기시키게 되고, 그로 인해 비정상적인 단백질과 세포소기관들이 분해되지 못하고 축적됨으로써 발생하게 된다. 이러한 원인들로 인해 파킨슨병의 치료 및 예방에 있어서, 단백질 분해 시스템의 중요성이 대두되고 있다. Research into the cause of Parkinson's disease has been undertaken for decades, but the mechanism is not clear. As a result, Parkinson's disease is classified as a progressive neurological disorder intractable disease without effective and effective long-term treatment (Pan et al., 2008, Habibi et al., 2011). However, it is estimated that Parkinson's disease is caused by various pathological factors occurring in Parkinson's disease patients. As a factor, the abnormal killing of dopamine-releasing neurons among the neurotransmitters distributed in the melanoma of the midbrain is disrupted or destroyed by the substantia nigra par compacta (Camins et al., 2008, Smith). , 2008). Abnormal death of dopamine neurons occurs through a series of processes. First, mitochondria damaged by various factors, such as the environment, heredity, and aging, cause functional degradation of the proteolytic system, which causes abnormal proteins and organelles. It is caused by accumulation and not decomposition. These causes raise the importance of proteolytic systems in the treatment and prevention of Parkinson's disease.
현재까지 임상에서 파킨슨병 환자에게 가장 높은 비율로 처방되는 약제는 L-dopa 제제이며, 이는 도파민 신경세포의 전구체인 Levodopa를 직접 주입하여 혈중 도파민 농도를 증가시켜주는 치료법이다. 직접적인 Levodopa의 주입은 파킨슨병 환자들에게 실질적인 증상 완화를 가져오지만, 장기간 사용하였을 경우 용량의 증가가 필수적이며, 용량의 증가에 따라 예측하지 못했던 다양한 부작용을 초래하는 것으로 알려져 있다(Rock and Peterson, 2006, McGeer and McGeer, 2008). 그러므로 도파민 혈중 농도를 높여주는 L-dopa를 직접 투여하는 치료법 보다는, 도파민 분비를 원활하게 해 주는 신경세포의 보호 및 사멸을 억제할 수 있는 좀 더 근본적인 치료법 및 약제의 개발이 시급하다고 할 수 있다. 이러한 시도 중 하나로 단백질 분해 시스템 중 하나인 자가포식작용을 표적으로 한 치료제 개발 연구가 최근 주목을 받고 있다. To date, the highest prescription for Parkinson's disease patients is L-dopa, which is a treatment that increases the level of dopamine in the blood by direct injection of Levodopa, a precursor of dopamine neurons. Direct infusion of Levodopa results in substantial symptomatic relief in Parkinson's patients, but increased doses are essential for long-term use and are known to cause a variety of unforeseen side effects as the dose increases (Rock and Peterson, 2006). , McGeer and McGeer, 2008). Therefore, it is urgent to develop more fundamental treatments and drugs that can suppress the protection and death of nerve cells that facilitate dopamine secretion, rather than the direct administration of L-dopa, which increases dopamine blood levels. As one of these attempts, research into the development of therapeutic agents targeting autophagy, one of the proteolytic systems, has recently attracted attention.
오토파지(Autophagy), 즉 자가포식작용은 자아, 자신이라는 뜻의 “self”와 먹는다는 의미의 “eating”을 칭하는 그리스어에서 파생된 단어로, 세포의 신진대사 균형을 유지할 뿐만 아니라 세포의 운명에 중요한 역할을 하는 자기 희생의 메커니즘으로 알려져 있다(Eisenberg-Lerner et al., 2009). 이러한 자가포식 작용은 자가포식소체(autophagosome)의 전구체이자 독립된 이중막인 파고폴(phagophore)의 가장자리가 점점 연장되어 세포 내 불필요한 단백질을 완전히 에워싸, 소포체인 자가포식소체를 형성할 수 있는 형태학적으로 특징적인 구조를 가지고 있다. 또한 자가포식소체는 미세소관을 따라 이동하여 세포 내 리소좀(lysosome)과 융합하게 되며 자가포식용해소체(autolysosome)를 형성하게 되며 이때, 리소좀 내부에서 분비되는 단백질분해효소에 의해 단백질을 분해시키는 것으로 보고되었다.Autophagy, or autophagy, is derived from the Greek word for "self," meaning "self," and "eating," meaning to eat. It not only maintains the metabolic balance of the cell but also affects the fate of the cell. It is known as a mechanism of self-sacrifice that plays an important role (Eisenberg-Lerner et al., 2009). This autophagy acts as a precursor to autophagosomes and is a morphological form of autophagosomes, which are independent bilayers, which gradually extend the edges of cells to completely surround unwanted proteins in the cell, forming vesicles. It has a characteristic structure. In addition, autophagosomes move along microtubules to fuse with lysosomes in cells and form autolysosomes, which are reported to degrade proteins by proteolytic enzymes secreted inside lysosomes. It became.
한편 벌개미취(Aster koraiensis Nakai)는 국화과(Compositae) 식물로 Gymnaster koraiensis (Nakai) Kitamura 라고도 불리며 우리나라 자생식물이다. 벌개미취는 주로 제주도, 경기도 이남 지역에서 분포하는 여러해살이풀이며, 특히 습도가 높은 계곡이나 습지에서 잘 생육하는 것으로 알려져 있다. 이러한 벌개미취 추출물 및 단일 성분(compound)의 효능에 대해 지금까지 알려진 것으로는 당뇨 망막 병증이 유도된 마우스의 망막 혈관 손상에 대한 방어 효과, 암 예방 효과를 가진 것으로 알려진 해독 효소의 유도 효과, 벌개미취 추출물의 단일 성분에 대한 단백질 당화 및 알도오스 환원 효소 저해 활성, 당뇨병 마우스 모델에 대하여 항-세포자살(anti-apoptosis) 활성을 통해 당뇨병성 신장증의 발달 억제 등을 비롯하여 대장암 억제, 간 보호, 산화스트레스 방어 효과 등이 있다. 그러나 아직까지 벌개미취를 통하여 구체적으로 파킨슨병을 비롯하여 자가포식작용에 관한 연구가 진행된 바가 없다.On the other hand, Aster koraiensis Nakai is a compositae plant, also called Gymnaster koraiensis (Nakai) Kitamura, and is a native plant of Korea. Beetle odor is a perennial herb distributed mainly in Jeju Island and sub-Gyeonggi Province, and is known to grow well in valleys and wetlands with high humidity. Known so far the efficacy of these bee and herb extract and single compound (compound) of diabetic retinopathy induced mouse retinal vascular damage, protective effect of the detoxification enzymes known to have cancer prevention effect, bee anesthetic extract of Protein glycation and aldose reductase inhibitory activity against a single component and anti-apoptosis activity against diabetic mouse models, including inhibition of the development of diabetic nephropathy, colon cancer suppression, liver protection, and oxidative stress Effect. However, there have been no studies on autophagy, including Parkinson's disease, through bee anesthesia.
이에 따라, 본 발명자는 자가포식 작용 유도를 통해 파킨슨병의 예방 및 치료제 개발에 관한 연구를 진행하던 중, 벌개미취(Aster koraiensis Nakai) 식물의 추출물이 자가포식 작용을 유도시킴과 동시에 파킨슨병의 예방 및 치료에 매우 효과적임을 확인하여 본 발명을 완성하였다.Accordingly, while the present inventors are conducting research on the development of a prophylactic and therapeutic agent for Parkinson's disease through induction of autophagy, the extract of Aster koraiensis Nakai plants induces autophagy and at the same time prevents and prevents Parkinson's disease. The present invention was completed by confirming that it is very effective for treatment.
본 발명의 목적은 벌개미취 추출물 또는 이의 분획물을 유효성분으로 포함하는 파킨슨병의 예방 또는 치료용 약학적 조성물을 제공하는 것이다.An object of the present invention is to provide a pharmaceutical composition for the prevention or treatment of Parkinson's disease comprising a bee odor extract or fractions thereof as an active ingredient.
본 발명의 다른 목적은 상기 약학적 조성물을 개체에 투여하는 단계를 포함하는 파킨슨병의 예방 또는 치료 방법을 제공하는 것이다Another object of the present invention is to provide a method for preventing or treating Parkinson's disease, comprising administering the pharmaceutical composition to a subject.
본 발명의 또 다른 목적은 벌개미취 추출물 또는 이의 분획물을 유효성분으로 포함하는 파킨슨병의 예방 또는 개선용 식품 조성물을 제공하는 것이다.Still another object of the present invention is to provide a food composition for preventing or improving Parkinson's disease, which comprises a bee anesthetist extract or a fraction thereof as an active ingredient.
본 발명의 또 다른 목적은 벌개미취 추출물 또는 이의 분획물을 유효성분으로 포함하는 파킨슨병의 예방 또는 개선용 사료 조성물을 제공하는 것이다.Still another object of the present invention is to provide a feed composition for preventing or improving Parkinson's disease, which comprises a bee anesthetist extract or a fraction thereof as an active ingredient.
본 발명의 또 다른 목적은 벌개미취 추출물 또는 이의 분획물을 유효성분으로 포함하는, 인비트로(in vitro) 자가포식 유도 효과에 의한 뇌세포 사멸 억제용 조성물을 제공하는 것이다.Still another object of the present invention is to provide a composition for inhibiting brain cell death by in vitro autophagy inducing effect, which comprises a bee stinger extract or a fraction thereof as an active ingredient.
본 발명의 벌개미취 추출물은 자가포식소체의 막 단백질인 LC3 단백질의 발현을 증가시키고, 자가포식 작용을 유도하는 인산화된 AMPK와 UK1의 증가 및 자가포식 작용을 억제하는 mTOR의 감소 효과를 통해 자가포식 유도하며, 이로 인해 이를 포함하는 조성물은 자가포식 작용 유도를 통한 파킨슨병의 예방 또는 치료 용도로 효과적으로 활용될 수 있다.Beetle odor extract of the present invention increases the expression of LC3 protein, a membrane protein of autophagosome, induces autophagy through the increase of phosphorylated AMPK and UK1 which induces autophagy, and the reduction of mTOR that inhibits autophagy. Therefore, the composition including the same can be effectively used for the prevention or treatment of Parkinson's disease through induction of autophagy action.
도 1은 SH-SY5Y 세포주에서 벌개미취 추출물의 첨가 농도에 따른 자가포식소체 막단백질인 LC3의 발현량의 증가효과를 웨스턴블롯팅으로 확인한 결과이다 (Control: 대조군). **는 대조군에 대해 P<0.01인 것을 나타내며, ***는 P<0.001인 것을 나타낸다.Figure 1 shows the results confirmed by Western blotting to increase the expression of the autophagy membrane protein LC3 expression in accordance with the concentration of the bee odor extract in SH-SY5Y cell line (Control: control). ** indicates P <0.01 for control and *** indicates P <0.001.
도 2는 벌개미취 추출물의 첨가 농도에 따른 인산화된 AMPK 단백질의 발현량의 증가효과를 웨스턴블롯팅으로 확인한 결과이다 (Control: 대조군). ***는 대조군에 대해 P<0.001인 것을 나타낸다.Figure 2 is a result confirmed by Western blotting to increase the expression of the phosphorylated AMPK protein according to the concentration of the bee hijab extract (Control: control). *** indicates P <0.001 for the control.
도 3은 벌개미취 추출물의 첨가농도에 따른 인산화된 ULK1 단백질 발현량의 증가효과를 웨스턴블롯팅으로 확인한 결과이다 (Control: 대조군). ***는 대조군에 대해 P<0.001인 것을 나타낸다Figure 3 is the result confirmed by Western blotting to increase the expression of phosphorylated ULK1 protein expression according to the concentration of the bee sting odor extract (Control: control). *** indicates P <0.001 for control
도 4는 벌개미취 추출물의 첨가 농도에 따른 mTOR단백질 발현량의 감소효과를 웨스턴블롯팅으로 확인한 결과이다 (Control: 대조군). *는 대조군에 대해 P<0.05인 것을 나타내며, ***는 P<0.001인 것을 나타낸다.Figure 4 is a result confirmed by Western blotting to reduce the expression of mTOR protein expression according to the concentration of the bee sting odor extract (Control: control). * Indicates P <0.05 for control and *** indicates P <0.001.
도 5는 자가포식작용의 억제제인 Bafilomycin A1을 투여하였을 때, 벌개미취 추출물 처리에 따른 LC3 단백질 발현량의 변화를 확인한 결과이다 (Control: 대조군). ***는 대조군에 대해 P<0.001인 것을 나타내며, ###은 벌개미취 추출물을 50 μg/ml의 농도로 처리한 군에 대해 P<0.001인 것을 나타낸다.5 is a result of confirming the change in the expression level of LC3 protein according to the bee sting anesthetic extract treatment when Bafilomycin A1, an inhibitor of autophagy, was administered (Control: control). *** indicates P <0.001 for the control group and ### indicates P <0.001 for the group treated with the bee-flavored extract at a concentration of 50 μg / ml.
도 6은 SH-SY5Y 세포주에서 벌개미취 분획물에 따른 자가포식소체 막단백질인 LC3의 발현량의 증가효과를 웨스턴블롯팅으로 확인한 결과이다 (Control: 대조군). Figure 6 shows the results confirmed by Western blotting to increase the expression of the autophagy membrane protein LC3 expression according to the bee odor fraction in SH-SY5Y cell line (Control: control).
도 7은 벌개미취 분획물의 첨가 농도에 따른 mTOR 단백질 발현량의 감소효과를 웨스턴블롯팅으로 확인한 결과이다 (Control: 대조군). **는 대조군에 대해 P<0.01인 것을 나타내며, ***는 P<0.001인 것을 나타낸다.Figure 7 is a result confirmed by Western blotting to reduce the amount of mTOR protein expression according to the addition concentration of the honey bee odor fraction (Control: control). ** indicates P <0.01 for control and *** indicates P <0.001.
도 8은 MPTP 투여에 의해 파킨슨 병(Parkinson’s disease)을 유도시킨 C57BL/6 마우스의 흑질(substantia nigra)에서 벌개미취 추출물 및 분획물 투여에 따른 Tyrosine hydroxylase (TH) 단백질 발현량의 증가효과를 웨스턴블롯팅으로 확인한 결과이다 (Control: 대조군; MPTP: MPTP 단독 처리군; 로피니롤(Ropinirole): 로피니롤+MPTP 처리군). ***는 대조군에 비하여 P<0.001인 것을 나타내며, ##는 MPTP 단독 처리군에 대해 P<0.01, ###는 MPTP 단독 처리군에 대해 P<0.001인 것을 나타낸다.FIG. 8 shows the effect of Western blotting on the expression of Tyrosine hydroxylase (TH) protein according to bee hives extracts and fractions in substantia nigra of C57BL / 6 mice induced Parkinson's disease by MPTP. The result was confirmed (Control: control; MPTP: MPTP-treated group; Ropinirole: ropinirole + MPTP-treated group). *** indicates P <0.001 compared to the control group, ## indicates P <0.01 for the MPTP treatment group and ### indicates P <0.001 for the MPTP treatment group.
도 9는 MPTP 투여에 의해 파킨슨 병(Parkinson’s disease)을 유도시킨 C57BL/6 마우스의 흑질(substantia nigra)에서 벌개미취 추출물 및 분획물 투여에 따른 α-synuclein 단백질 발현량의 감소효과를 웨스턴블롯팅으로 확인한 결과이다 (Control: 대조군; MPTP: MPTP 단독 처리군; 로피니롤: 로피니롤+MPTP 처리군). ***는 대조군에 비하여 P<0.001인 것을 나타내며, ##는 MPTP 단독 처리군에 대해 P<0.01, ###는 MPTP 단독 처리군에 대해 P<0.001인 것을 나타낸다.9 is a Western blotting confirmed the effect of reducing the expression level of α-synuclein protein according to bee hijab extract and fraction administration in the substantia nigra of C57BL / 6 mice induced Parkinson's disease by MPTP administration (Control: control; MPTP: MPTP-only group; rofinirol: rofiniro + MPTP-treated group). *** indicates P <0.001 compared to the control group, ## indicates P <0.01 for the MPTP treatment group and ### indicates P <0.001 for the MPTP treatment group.
도 10은 MPTP 투여에 의해 파킨슨 병(Parkinson’s disease)을 유도시킨 C57BL/6 마우스의 흑질(substantia nigra)에서 벌개미취 추출물 및 분획물 투여에 따른 LC3 단백질 발현량의 증가 효과를 웨스턴블롯팅으로 확인한 결과이다 (Control: 대조군; MPTP: MPTP 단독 처리군; 로피니롤: 로피니롤+MPTP 처리군). #는 MPTP 단독 처리군에 대해 P<0.05인 것을 나타낸다.10 is a result of Western blotting confirming the effect of increasing the level of LC3 protein expression according to the administration of bee sting extract and fraction in the substantia nigra of C57BL / 6 mice induced Parkinson's disease by MPTP administration ( Control: control; MPTP: MPTP alone group; ropinirol: rofiniro + MPTP treated group). # Indicates P <0.05 for MPTP alone treatment group.
도 11은 MPTP 투여에 의해 파킨슨 병(Parkinson’s disease)을 유도시킨 C57BL/6 마우스의 흑질(substantia nigra)에서 벌개미취 추출물 및 분획물 투여에 따른 인산화된 AMPK 단백질 발현량의 증가효과를 웨스턴블롯팅으로 확인한 결과이다 (Control: 대조군; MPTP: MPTP 단독 처리군; 로피니롤: 로피니롤+MPTP 처리군). #는 MPTP 단독 처리군에 대해 P<0.05인 것을 나타낸다.FIG. 11 shows the results of Western blotting confirming the effect of increasing the amount of phosphorylated AMPK protein expression according to bee hives extracts and fractions in substantia nigra of C57BL / 6 mice induced Parkinson's disease by MPTP. (Control: control; MPTP: MPTP-only group; rofinirol: rofiniro + MPTP-treated group). # Indicates P <0.05 for MPTP alone treatment group.
도 12는 MPTP 투여에 의해 파킨슨 병(Parkinson’s disease)을 유도시킨 C57BL/6 마우스의 흑질(substantia nigra)에서 벌개미취 추출물 및 분획물 투여에 따른 인산화된 ULK1 (s555) 단백질 발현량의 증가효과를 웨스턴블롯팅으로 확인한 결과이다 (Control: 대조군; MPTP: MPTP 단독 처리군; 로피니롤: 로피니롤+MPTP 처리군). *는 대조군에 비하여 P<0.05인 것을 나타내며, ##는 MPTP 단독 처리군에 대해 P<0.01, ###는 MPTP 단독 처리군에 대해 P<0.001인 것을 나타낸다.FIG. 12 shows Western blotting effect of increased phosphorylated ULK1 (s555) protein expression levels according to bee hives extract and fraction administration in substantia nigra of C57BL / 6 mice induced Parkinson's disease by MPTP administration. This was confirmed by (Control: control group; MPTP: MPTP treatment group; ropinillol: ropinillol + MPTP treatment group). * Indicates P <0.05 compared to the control, ## indicates P <0.01 for the MPTP-only group and ### indicates P <0.001 for the MPTP-only group.
도 13은 MPTP 투여에 의해 파킨슨 병(Parkinson’s disease)을 유도시킨 C57BL/6 마우스의 흑질(substantia nigra)에서 벌개미취 추출물 및 분획물 투여에 따른 mTOR 단백질 발현량의 감소효과를 웨스턴블롯팅으로 확인한 결과이다 (Control: 대조군; MPTP: MPTP 단독 처리군; 로피니롤: 로피니롤+MPTP 처리군). #는 MPTP 단독 처리군에 대해 P<0.05, ##는 MPTP 단독 처리군에 대해 P<0.01인 것을 나타낸다.FIG. 13 shows the results of Western blotting confirming the reduction effect of mTOR protein expression according to bee hives extract and fraction administration in substantia nigra of C57BL / 6 mice induced Parkinson's disease by MPTP administration. Control: control; MPTP: MPTP alone group; ropinirol: rofiniro + MPTP treated group). # Indicates P <0.05 for the MPTP treatment group and ## indicates P <0.01 for the MPTP treatment group.
도 14는 MPTP 투여에 의해 파킨슨 병(Parkinson’s disease)을 유도시킨 C57BL/6 마우스의 흑질(substantia nigra)에서 벌개미취 추출물 및 분획물 투여에 따른 Beclin-1 단백질 발현량의 증가효과를 웨스턴블롯팅으로 확인한 결과이다 (Control: 대조군; MPTP: MPTP 단독 처리군; 로피니롤: 로피니롤+MPTP 처리군). ##는 MPTP 단독 처리군에 대해 P<0.01, ###는 MPTP 단독 처리군에 대해 P<0.001인 것을 나타낸다.14 shows the results of Western blotting confirming the effect of increasing the expression of Beclin-1 protein according to bee hives extracts and fractions in substantia nigra of C57BL / 6 mice induced Parkinson's disease by MPTP administration. (Control: control; MPTP: MPTP-only group; rofinirol: rofiniro + MPTP-treated group). ## represents P <0.01 for the MPTP-only group and ### represents P <0.001 for the MPTP-only group.
본 발명에서 개시된 각각의 설명 및 실시형태는 각각의 다른 설명 및 실시 형태에도 적용될 수 있다. 즉, 본 발명에서 개시된 다양한 요소들의 모든 조합이 본 발명의 범주에 속한다. 또한, 하기 기술된 구체적인 서술에 의하여 본 발명의 범주가 제한된다고 볼 수 없다.Each description and embodiment disclosed in the present invention may be applied to each other description and embodiment. That is, all combinations of the various elements disclosed in the present invention fall within the scope of the present invention. In addition, the scope of the present invention is not to be limited by the specific description described below.
상기 목적을 달성하기 위한 본 발명의 하나의 양태는 벌개미취(Aster koraiensis Nakai) 추출물 또는 이의 분획물을 유효성분으로 포함하는 파킨슨병의 예방 또는 치료용 약학적 조성물을 제공한다. One aspect of the present invention for achieving the above object provides a pharmaceutical composition for the prevention or treatment of Parkinson's disease, including Aster koraiensis Nakai extract or a fraction thereof as an active ingredient.
또한, 본 발명은 벌개미취(Aster koraiensis Nakai) 추출물 또는 이의 분획물을 유효성분으로 포함하는 조성물의 파킨슨병의 예방 또는 치료용도를 제공한다. In addition, the present invention provides a preventive or therapeutic use of Parkinson's disease of the composition comprising an extract of Aster koraiensis Nakai or fractions thereof as an active ingredient.
본 발명의 용어, "벌개미취"는 학명이 Aster koraiensis Nakai이며, 본 발명에서 상기 식물의 지상부를 추출하여 사용할 수 있으나 보다 구체적으로는 상기 식물의 잎, 줄기를 사용할 수 있으며, 파킨슨병의 예방 또는 치료 효과를 갖는 한, 특별히 이에 제한되지 않는다. As used herein, the term "beetle odor" is scientific name Aster koraiensis Nakai, but in the present invention can be used to extract the above-ground parts of the plant, more specifically, the leaves and stems of the plant can be used, preventing or treating Parkinson's disease As long as it has an effect, it is not specifically limited to this.
상기 벌개미취는 상업적으로 판매되는 것을 구입하거나, 자연에서 채취 또는 재배된 것을 사용할 수 있다.The hummingbird odor can be purchased commercially, or may be used collected or cultivated in nature.
본 발명의 용어, "추출물"은 벌개미취를 추출 처리하여 얻어지는 추출액, 상기 추출액의 희석액이나 농축액, 상기 추출액을 건조하여 얻어지는 건조물, 상기 추출액의 조정제물이나 정제물, 또는 이들의 혼합물 등, 추출액 자체 및 추출액을 이용하여 형성 가능한 모든 제형의 추출물을 포함한다.As used herein, the term "extract" refers to an extract, such as an extract obtained by extracting a bee odor, a diluent or concentrate of the extract, a dried product obtained by drying the extract, a modifier or purified product of the extract, or a mixture thereof. It includes extracts of all formulations that can be formed using extracts.
상기 벌개미취를 추출하는 방법은 특별히 제한되지 않으며, 당해 기술 분야에서 통상적으로 사용하는 방법에 따라 추출할 수 있다. 상기 추출 방법의 비 제한적인 예로는, 열수 추출법, 초음파 추출법, 여과법, 환류 추출법 등을 들 수 있으며, 이들은 단독으로 수행되거나 2종 이상의 방법을 병용하여 수행될 수 있다.The method of extracting the hummingbird odor is not particularly limited, and may be extracted according to a method commonly used in the art. Non-limiting examples of the extraction method may include hot water extraction, ultrasonic extraction, filtration, reflux extraction, etc., these may be carried out alone or in combination of two or more methods.
본 발명에서 상기 벌개미취를 추출하는데 사용되는 추출 용매의 종류는 특별히 제한되지 않으며, 당해 기술 분야에서 공지된 임의의 용매를 사용할 수 있다. 상기 추출 용매의 비제한적인 예로는 물, C1 내지 C4의 알코올 및 이들의 혼합 용매 등을 들 수 있으며, 이들은 단독으로 사용되거나 1종 이상 혼합하여 사용될 수 있다. 구체적으로, 바람직하게는 95% 에탄올이 사용될 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the type of extraction solvent used to extract the bee odor is not particularly limited, and any solvent known in the art may be used. Non-limiting examples of the extraction solvent may include water, alcohols of C1 to C4, mixed solvents thereof, and the like, which may be used alone or in combination of one or more thereof. Specifically, preferably 95% ethanol may be used, but is not limited thereto.
본 발명의 용어, "분획물"은 여러 다양한 구성 성분들을 포함하는 혼합물로부터 특정 성분 또는 특정 성분 그룹을 분리하기 위하여 분획을 수행하여 얻어진 결과물을 의미한다.As used herein, the term "fraction" refers to the result obtained by performing fractionation to separate a specific component or a specific group of components from a mixture comprising several various components.
본 발명에서 상기 분획물을 얻는 분획 방법은 특별히 제한되지 않으며, 당해 기술 분야에서 통상적으로 사용하는 방법에 따라 수행될 수 있다. 상기 분획 방법의 비제한적인 예로는, 다양한 용매를 처리하여 수행하는 용매 분획법, 일정한 분자량 컷-오프 값을 갖는 한외 여과막을 통과시켜 수행하는 한외여과 분획법, 다양한 크로마토그래피(크기, 전하, 소수성 또는 친화성에 따른 분리를 위해 제작된 것)를 수행하는 크로마토그래피 분획법, 및 이들의 조합 등이 있다. 구체적으로, 본 발명의 벌개미취를 추출하여 얻은 추출물에 소정의 용매를 처리하여 상기 추출물로부터 분획물을 얻는 방법을 들 수 있다.The fractionation method of obtaining the fraction in the present invention is not particularly limited, and may be performed according to a method commonly used in the art. Non-limiting examples of the fractionation method include a solvent fractionation method performed by treating various solvents, an ultrafiltration fractionation method performed through an ultrafiltration membrane having a constant molecular weight cut-off value, and various chromatography (size, charge, hydrophobicity). Or chromatographed fractions), and combinations thereof, which are prepared for separation according to affinity. Specifically, a method of obtaining a fraction from the extract by treating a predetermined solvent with an extract obtained by extracting the bee sting odor of the present invention.
본 발명에서 상기 분획물을 얻는 데 사용되는 분획 용매의 종류는 특별히 제한되지 않으며, 당해 기술 분야에서 공지된 임의의 용매를 사용할 수 있다. 상기 분획 용매의 비제한적인 예로는 물, 탄소수 1 내지 4의 알코올 등의 극성 용매; 헥산(Hexan), 에틸 아세테이트(Ethyl acetate), 클로로포름(Chloroform), 디클로로메탄(Dichloromethane) 등의 비극성 용매; 또는 이들의 혼합용매 등을 들 수 있다. 이들은 단독으로 사용되거나 1종 이상 혼합하여 사용될 수 있지만, 이에 제한되는 것은 아니며, 구체적으로, 헥산, 에틸 아세테이트, 부탄올(butanol) 또는 물이 단독으로 사용되거나 1종 이상 혼합하여 사용될 수 있지만, 이에 제한되는 것은 아니다.The kind of the fractionation solvent used to obtain the fraction in the present invention is not particularly limited, and any solvent known in the art may be used. Non-limiting examples of the fractionation solvents include polar solvents such as water and alcohols having 1 to 4 carbon atoms; Nonpolar solvents such as hexane, ethyl acetate, chloroform and dichloromethane; Or a mixed solvent thereof. These may be used alone or in combination of one or more thereof, but is not limited thereto. Specifically, hexane, ethyl acetate, butanol or water may be used alone or in combination of one or more thereof, but is not limited thereto. It doesn't happen.
또한, 상기 추출물 또는 분획물은 추출 후 건조 분말 형태로 제조되어 사용될 수 있지만, 이제 제한되는 것은 아니다.In addition, the extract or fraction may be prepared and used in the form of a dry powder after extraction, but is not limited thereto.
본 발명의 용어, "파킨슨병"은 도파민 신경세포의 소실로 인해 발생하는 신경계의 퇴행성 뇌 질환을 의미한다. 안정떨림, 경직, 운동완만(운동느림) 및 자세 불안정성이 특징적으로 나타나며, 일반적으로 60세 이후에 임상 증상이 나타나기 시작한다. As used herein, the term "Parkinson's disease" means a degenerative brain disease of the nervous system caused by the loss of dopamine neurons. Stability, stiffness, slowness of movement (slowness of motion) and postural instability are characteristic, and clinical symptoms usually begin after age 60.
본 발명의 용어, "예방"은 벌개미취 추출물 또는 이의 분획물을 포함하는 본 발명의 약학적 조성물의 투여에 의해 파킨슨병의 진행을 억제시키거나 또는 지연시키는 모든 행위를 의미한다.As used herein, the term "prevention" means any action that inhibits or delays the progression of Parkinson's disease by the administration of a pharmaceutical composition of the present invention comprising a cane odor extract or a fraction thereof.
본 발명의 용어, "치료"는 상기 약학적 조성물의 투여에 의해 파킨슨병이 호전되거나 이롭게 변경되는 모든 행위를 의미한다.As used herein, the term "treatment" means any action in which Parkinson's disease is ameliorated or beneficially altered by administration of the pharmaceutical composition.
본 발명의 "약학적 조성물"은 약학적 조성물의 제조에 통상적으로 사용하는 약학적으로 허용가능한 담체, 부형제 또는 희석제를 추가로 포함할 수 있고, 상기 담체는 비자연적 담체(non-naturally occuring carrier)를 포함할 수 있다.The "pharmaceutical composition" of the present invention may further comprise a pharmaceutically acceptable carrier, excipient or diluent commonly used in the manufacture of a pharmaceutical composition, which carrier is a non-naturally occuring carrier. It may include.
상기 약학적 조성물은, 각각 통상의 방법에 따라 산제, 과립제, 정제, 캡슐제, 현탁액, 에멀젼, 시럽, 에어로졸 등의 경구형 제형, 외용제, 좌제 및 멸균 주사용액의 형태로 제형화하여 사용될 수 있다.The pharmaceutical composition may be used in the form of powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols and the like, oral formulations, external preparations, suppositories, and sterile injectable solutions, respectively, according to conventional methods. .
상기 "약학적으로 허용가능한"은 상기 조성물에 노출되는 세포나 인간에게 독성이 없는 특성을 나타내는 것을 의미한다."Pharmaceutically acceptable" means to exhibit properties that are not toxic to cells or humans exposed to the composition.
구체적으로, 상기 담체의 종류는 특별히 제한되지 않으며, 당해 기술 분야에서 통상적으로 사용되고 약학적으로 허용되는 담체라면 어느 것이든 사용할 수 있다. 상기 담체의 비제한적인 예로, 식염수, 멸균수, 링거액, 완충 식염수, 알부민 주사 용액, 덱스트로즈 용액, 말토 덱스트린 용액, 글리세롤, 에탄올 등을 들 수 있다. 이들은 단독으로 사용되거나 2종 이상을 혼합하여 사용될 수 있다. 또한, 필요한 경우 항산화제, 완충액 및/또는 정균제 등 다른 통상의 첨가제를 첨가하여 사용할 수 있으며, 희석제, 분산제, 계면 활성제, 결합제, 윤활제 등을 부가적으로 첨가하여 수용액, 현탁액, 유탁액 등과 같은 주사용 제형, 환약, 캡슐, 과립 또는 정제 등으로 제제화하여 사용할 수 있다.Specifically, the type of the carrier is not particularly limited, and any carrier can be used as long as it is a conventionally used and pharmaceutically acceptable carrier in the art. Non-limiting examples of the carrier include saline, sterile water, Ringer's solution, buffered saline, albumin injection solution, dextrose solution, maltodextrin solution, glycerol, ethanol and the like. These may be used alone or in combination of two or more thereof. In addition, if necessary, other conventional additives such as antioxidants, buffers and / or bacteriostatic agents may be added and used, and diluents, dispersants, surfactants, binders, lubricants, and the like may be additionally added to provide a solution such as an aqueous solution, a suspension, an emulsion, or the like. It may be formulated into a use formulation, pills, capsules, granules or tablets.
본 발명에 따른 파킨슨병의 예방 또는 치료용 약학적 조성물의 투여 방식은 특별히 제한되지 않으며, 당해 기술 분야에서 통상적으로 사용하는 방식에 따를 수 있다. 상기 투여 방식의 비제한적인 예로, 조성물을 경구 투여 또는 비경구 투여 방식으로 투여할 수 있다. 또한, 본 발명의 파킨슨병의 예방 또는 치료용 조성물은 목적하는 투여 방식에 따라 다양한 제형으로 제조될 수 있다.The mode of administration of the pharmaceutical composition for preventing or treating Parkinson's disease according to the present invention is not particularly limited and may be in accordance with a method commonly used in the art. As a non-limiting example of the mode of administration, the composition may be administered by oral or parenteral administration. In addition, the composition for preventing or treating Parkinson's disease of the present invention can be prepared in various formulations according to the desired mode of administration.
본 발명의 다른 하나의 양태는 상기 약학적 조성물을 개체에 투여하는 단계를 포함하는 파킨슨병의 예방 또는 치료 방법을 제공한다.Another aspect of the invention provides a method of preventing or treating Parkinson's disease comprising administering the pharmaceutical composition to a subject.
이때, 상기 파킨슨병, 예방 및 치료의 정의는 상기에서 설명한 바와 같다.At this time, the definition of Parkinson's disease, prevention and treatment is as described above.
본 발명의 용어, "투여"는 적절한 방법으로 개체에게 소정의 물질을 도입하는 것을 의미한다.As used herein, the term "administration" means the introduction of certain substances into an individual in a suitable manner.
본 발명의 용어, "개체"는 파킨슨병이 발병하였거나, 발병할 수 있는 인간을 포함한 쥐, 생쥐, 가축 등의 모든 동물을 의미한다. 구체적인 예로, 인간을 포함한 포유동물일 수 있다.As used herein, the term "individual" means all animals, such as rats, mice, and livestock, including humans who may or may develop Parkinson's disease. As a specific example, it may be a mammal including a human.
보다 구체적으로, 상기 “개체”는 반려동물을 포함할 수 있다. 상기 반려동물은 인간과 함께 더불어 사는 동물을 의미하며, 구체적인 종류로서 개, 고양이, 햄스터, 기니피그 등의 포유류, 앵무새, 카나리아 등의 조류 등을 들 수 있으나, 이에 제한되는 것은 아니다.More specifically, the "individual" may include a companion animal. The companion animal refers to an animal that lives together with a human, and includes, but is not limited to, a mammal such as a dog, a cat, a hamster, and a guinea pig, a bird such as a parrot, a canary, and the like.
구체적으로, 본 발명의 파킨슨병의 예방 또는 치료 방법은 개체에 벌개미취 추출물 또는 이의 분획물을 포함하는 약학적 조성물을 약학적으로 유효한 양으로 투여하는 단계를 포함할 수 있다.Specifically, the method for preventing or treating Parkinson's disease of the present invention may include administering to the individual a pharmaceutically effective amount of a medicinal composition comprising a bee herb extract or a fraction thereof.
상기 "약학적으로 유효한 양"은 의학적 치료에 적용 가능한 합리적인 수혜/위험 비율로 질환을 치료하기에 충분하며 부작용을 일으키지 않을 정도의 양을 의미하며, 유효 용량 수준은 환자의 성별, 연령, 체중, 건강상태, 질병의 종류, 중증도, 약물의 활성, 약물에 대한 민감도, 투여 방법, 투여 시간, 투여 경로, 및 배출 비율, 치료 기간, 배합 또는 동시에 사용되는 약물을 포함한 요소 및 기타 의학 분야에 잘 알려진 요소에 따라 당업자에 의해 용이하게 결정될 수 있다.The "pharmaceutically effective amount" means an amount sufficient to treat the disease at a reasonable benefit / risk ratio applicable to the medical treatment and not causing side effects, and the effective dose level refers to the sex, age, weight, Well-known in the field and other medical fields, including health status, type of disease, severity, drug activity, drug sensitivity, method of administration, time of administration, route of administration, and rate of release, duration of treatment, combination or drug used simultaneously Depending on the factor, it can be readily determined by one skilled in the art.
본 발명의 약학적 조성물은 고형분을 기준으로 1일 0.0001 내지 500 mg/체중 kg으로, 더욱 구체적으로 0.01 내지 500 mg/체중kg으로 투여할 수 있다. 투여는 상기 권장 투여량을 하루에 한 번 투여할 수도 있고, 수 회 나누어 투여할 수도 있다.The pharmaceutical composition of the present invention may be administered at 0.0001 to 500 mg / kg body weight per day, more specifically at 0.01 to 500 mg / kg body weight, based on solids. Dosing may include administering the recommended dose once a day or in several divided doses.
본 발명의 약학적 조성물은 개별 치료제로 투여되거나 다른 치료제와 병용하여 투여될 수 있고, 종래의 치료제와는 순차적 또는 동시에 투여될 수 있다. 또한, 단일 또는 다중 투여될 수 있다. 상기 요소를 모두 고려하여 부작용을 유발하지 않으면서 최소한의 양으로 최대 효과를 얻을 수 있는 양을 투여하는 것이 중요하며, 이는 당업자에 의해 용이하게 결정될 수 있다.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. It may also be single or multiple doses. Taking all of these factors into consideration it is important to administer an amount that will achieve the maximum effect in a minimum amount without causing side effects, which can be readily determined by one skilled in the art.
본 발명의 파킨슨병의 예방 또는 치료 방법에서, 상기 조성물을 투여하는 투여 경로 및 투여 방식은 특별히 제한되지 않으며, 목적하는 해당 부위에 상기 조성물을 포함하는 조성물이 도달할 수 있는 한 임의의 투여 경로 및 투여 방식에 따를 수 있다. 구체적으로, 상기 조성물은 경구 또는 비경구의 다양한 경로를 통하여 투여될 수 있으며, 그 투여 경로의 비제한적인 예로는, 구강, 직장, 국소, 정맥내, 복강내, 근육내, 동맥내, 경피, 비측내 또는 흡입 등을 통하여 투여되는 것을 들 수 있다.In the method for preventing or treating Parkinson's disease of the present invention, the route of administration and the mode of administration of the composition are not particularly limited, and any route of administration can be reached as long as the composition comprising the composition can be reached at the desired site. Depending on the mode of administration. Specifically, the composition may be administered through various routes, oral or parenteral, and non-limiting examples of the route of administration include oral, rectal, topical, intravenous, intraperitoneal, intramuscular, intraarterial, transdermal, nasal What is administered through intralateral or inhalation etc. are mentioned.
본 발명의 또 다른 하나의 양태는 벌개미취 추출물 또는 이의 분획물을 유효성분으로 포함하는 파킨슨병의 예방 또는 개선용 식품 조성물을 제공한다.Yet another aspect of the present invention provides a food composition for preventing or improving Parkinson's disease, comprising a bee odorous extract or a fraction thereof as an active ingredient.
이때, 상기 벌개미취, 추출물, 분획물, 파킨슨병 및 예방의 정의는 상기에서 설명한 바와 같다.At this time, the definition of bee odor, extract, fraction, Parkinson's disease and prevention is as described above.
본 발명의 용어, "개선"은 본 발명의 조성물의 투여로 치료되는 상태와 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 the condition to be treated with the administration of the compositions of the present invention, eg the extent of symptoms.
본 발명의 용어, "식품"은 육류, 소시지, 빵, 초콜릿, 캔디류, 스낵류, 과자류, 피자, 라면, 기타 면류, 껌류, 아이스크림류를 포함한 낙농제품, 각종 스프, 음료수, 차, 드링크제, 알코올음료, 비타민 복합제, 건강 기능 식품 및 건강 식품 등이 있으며, 통상적인 의미에서의 식품을 모두 포함한다.The term "food" of the present invention, meat, sausage, bread, chocolate, candy, snacks, confectionery, pizza, ramen, other noodles, gum, dairy products including ice cream, various soups, drinks, tea, drinks, alcoholic beverages , Vitamin complexes, nutraceuticals and health foods, and includes all foods in the usual sense.
본 발명의 식품 조성물은 일상적으로 섭취하는 것이 가능하기 때문에 높은 파킨슨병 개선 효과를 기대할 수 있으므로, 건강 증진 목적으로 매우 유용하게 사용될 수 있다.Since the food composition of the present invention can be consumed on a daily basis, a high Parkinson's disease improvement effect can be expected, and thus can be very useful for health promotion purposes.
상기 건강 기능(성) 식품(functional food)이란, 특정보건용 식품(food for special health use, FoSHU)과 동일한 용어로, 영양 공급 외에도 생체조절기능이 효율적으로 나타나도록 가공된 의학, 의료효과가 높은 식품을 의미한다. 여기서 '기능(성)'이라 함은 인체의 구조 및 기능에 대하여 영양소를 조절하거나 생리학적 작용 등과 같은 보건용도에 유용한 효과를 얻는 것을 의미한다. 본 발명의 식품은 당 업계에서 통상적으로 사용되는 방법에 의하여 제조가능하며, 상기 제조시에는 당 업계에서 통상적으로 첨가하는 원료 및 성분을 첨가하여 제조할 수 있다. 또한, 상기 식품의 제형은 식품으로 인정되는 제형이면 제한 없이 제조될 수 있다. 본 발명의 식품용 조성물은 다양한 형태의 제형으로 제조될 수 있으며, 일반 약품과는 달리 천연물을 원료로 하여 약품의 장기 복용 시 발생할 수 있는 부작용 등이 없는 장점이 있고, 휴대성이 뛰어나므로, 본 발명의 식품은 파킨슨병 개선 효과를 증진시키기 위한 보조제로 섭취가 가능하다.The functional food (functional food) is the same term as a food for special health use (Food for special health use, FoSHU), in addition to the nutritional supply, the processed food, so that the bioregulatory function appears efficiently, high medical effect Means food. Here, the term 'function (sex)' refers to obtaining a useful effect for health purposes such as nutrient control or physiological action on the structure and function of the human body. The food of the present invention may be prepared by a method commonly used in the art, and the preparation may be performed by adding raw materials and ingredients commonly added in the art. In addition, the formulation of the food may be prepared without limitation as long as the formulation is recognized as a food. Food composition of the present invention can be prepared in a variety of dosage forms, unlike the general medicine has the advantage that there is no side effect that can occur when taking a long-term use of the drug as a natural product, and because the portability is excellent, The food of the invention can be taken as an adjuvant for enhancing the Parkinson's disease improving effect.
상기 건강 식품(health food)은 일반식품에 비해 적극적인 건강유지나 증진 효과를 가지는 식품을 의미하고, 건강보조식품(health supplement food)은 건강보조 목적의 식품을 의미한다. 경우에 따라, 건강 기능 식품, 건강식품, 건강보조식품의 용어는 혼용된다.The health food refers to foods having active health maintenance or promotion effect as compared to general foods, and the health supplement food refers to foods for health supplementation purposes. In some cases, the terms nutraceutical, health food, dietary supplement are used interchangeably.
구체적으로, 상기 건강 기능 식품은 본 발명의 추출물을 음료, 차류, 향신료, 껌, 과자류 등의 식품 소재에 첨가하거나, 캡슐화, 분말화, 현탁액 등으로 제조한 식품으로, 이를 섭취할 경우 건강상 특정한 효과를 가져오는 것을 의미하나, 일반 약품과는 달리 식품을 원료로 하여 약품의 장기 복용 시 발생할 수 있는 부작용이 없는 장점이 있다.Specifically, the health functional food is a food prepared by adding the extract of the present invention to food materials such as beverages, teas, spices, gums, confections, or the like, encapsulated, powdered, suspensions, etc. Means to bring effect, but unlike the general medicine has the advantage that there is no side effect that can occur when taking long-term use of the drug as a raw material.
상기 식품 조성물은 생리학적으로 허용 가능한 담체를 추가로 포함할 수 있는데, 담체의 종류는 특별히 제한되지 않으며 당해 기술 분야에서 통상적으로 사용되는 담체라면 어느 것이든 사용할 수 있다.The food composition may further include a physiologically acceptable carrier, and the type of carrier is not particularly limited and may be any carrier that is commonly used in the art.
또한, 상기 식품 조성물은 식품 조성물에 통상 사용되어 냄새, 맛, 시각 등을 향상시킬 수 있는 추가 성분을 포함할 수 있다. 예들 들어, 비타민 A, C, D, E, B1, B2, B6, B12, 니아신(niacin), 비오틴(biotin), 폴레이트(folate), 판토텐산(panthotenic acid) 등을 포함할 수 있다. 또한, 아연(Zn), 철(Fe), 칼슘(Ca), 크롬(Cr), 마그네슘(Mg), 망간(Mn), 구리(Cu), 크륨(Cr) 등의 미네랄; 및 라이신, 트립토판, 시스테인, 발린 등의 아미노산을 포함할 수 있다.In addition, the food composition may include additional ingredients that are commonly used in food compositions to improve the smell, taste, time and the like. For example, it may include vitamins A, C, D, E, B1, B2, B6, B12, niacin, biotin, folate, panthotenic acid, and the like. In addition, minerals such as zinc (Zn), iron (Fe), calcium (Ca), chromium (Cr), magnesium (Mg), manganese (Mn), copper (Cu) and chromium (Cr); And amino acids such as lysine, tryptophan, cysteine, valine and the like.
또한, 상기 식품 조성물은 방부제(소르빈산 칼륨, 벤조산나트륨, 살리실산, 데히드로초산나트륨 등), 살균제(표백분과 고도 표백분, 차아염소산나트륨 등), 산화방지제(부틸히드록시아니졸(BHA), 부틸히드록시톨류엔(BHT) 등), 착색제(타르색소 등), 발색제(아질산 나트륨, 아초산 나트륨 등), 표백제(아황산나트륨), 조미료(MSG 글루타민산나트륨 등), 감미료(둘신, 사이클레메이트, 사카린, 나트륨 등), 향료(바닐린, 락톤류 등), 팽창제(명반, D-주석산수소칼륨 등), 강화제, 유화제, 증점제(호료), 피막제, 검기초제, 거품억제제, 용제, 개량제 등의 식품 첨가물(food additives)을 포함할 수 있다. 상기 첨가물은 식품의 종류에 따라 선별되고 적절한 양으로 사용될 수 있다.In addition, the food composition is a preservative (potassium sorbate, sodium benzoate, salicylic acid, sodium dehydroacetic acid, etc.), fungicides (bleaching powder and highly bleaching powder, sodium hypochlorite, etc.), antioxidants (butylhydroxyanisol (BHA), butylhydride) Oxytoluene (BHT), etc.), colorants (such as tar pigments), colorants (sodium nitrite, sodium nitrite, etc.), bleach (sodium sulfite), seasonings (such as MSG glutamate), sweeteners (ducin, cyclate, saccharin Foods such as sodium, etc.), fragrances (vanillin, lactones, etc.), swelling agents (alum, potassium D-tartrate, etc.), reinforcing agents, emulsifiers, thickeners (foils), coatings, gum herbicides, foam inhibitors, solvents, modifiers, etc. It may include food additives. The additive may be selected according to the type of food and used in an appropriate amount.
본 발명의 또 다른 하나의 양태는 벌개미취 추출물 또는 이의 분획물을 유효성분으로 포함하는 파킨슨병의 예방 또는 개선용 사료 조성물을 제공한다.Another aspect of the present invention provides a feed composition for the prevention or improvement of Parkinson's disease comprising a bee anesthetized extract or a fraction thereof as an active ingredient.
상기 벌개미취, 추출물, 분획물, 예방 및 개선에 대해서는 상기 설명한 바와 같다.The bee odor, extract, fraction, prevention and improvement are as described above.
본 발명의 벌개미취 추출물 또는 이의 분획물을 포함하는 조성물은 가축, 또는 반려동물과 같이 인간 이외의 개체의 파킨슨병 발병 예방 또는 치료를 위하여 사용될 수 있으며, 기능성 사료첨가제, 또는 사료용 조성물로 활용할 수 있다.The composition comprising the honey bee odor extract or fractions thereof of the present invention can be used for the prevention or treatment of the development of Parkinson's disease in individuals other than humans, such as livestock or companion animals, can be utilized as a functional feed additive, or a feed composition.
본 발명에 따른 사료 조성물 내 벌개미취 추출물 또는 이의 분획물의 함량은 적용 가축의 종류 및 연령, 적용 형태, 목적하는 효과 등에 따라서 적절하게 조절 가능하며, 예컨대 1 내지 99 중량%, 바람직하게는 10 내지 90 중량%, 더욱 바람직하게는 20 내지 80 중량%로 사용될 수 있으나, 이에 제한되는 것은 아니다.The content of the bee-flavored extract or fractions thereof in the feed composition according to the present invention can be appropriately adjusted according to the type and age of the livestock, the type of application, the desired effect, and the like, for example, 1 to 99% by weight, preferably 10 to 90% by weight. %, More preferably 20 to 80% by weight, but is not limited thereto.
본 발명의 사료 조성물은 투여를 위해서 벌개미취 추출물 또는 이의 분획물 이외에 추가로 구연산, 푸마르산, 아디프산, 젖산 등의 유기산; 인산칼륨, 인산나트륨, 중합 인산염 등의 인산염; 폴리페놀, 카테친, 토코페롤, 비타민 C, 녹차 추출물, 키토산, 탄니산 등의 천연 항산화제 중 1종 이상을 혼합하여 사용할 수 있다. 또한 희석제, 분산제, 계면활성제, 결합제 또는 윤활제를 부가적으로 첨가하여 수용액, 현탁액, 유탁액 등과 같은 주사용 제형, 캡슐, 과립 또는 정제로 제제화할 수 있다.Feed composition of the present invention for administration, in addition to the bee anesthetic extract or fractions thereof, organic acids such as citric acid, fumaric acid, adipic acid, lactic acid; Phosphates such as potassium phosphate, sodium phosphate and polymerized phosphate; One or more of natural antioxidants such as polyphenol, catechin, tocopherol, vitamin C, green tea extract, chitosan and tannic acid can be mixed and used. In addition, diluents, dispersants, surfactants, binders or lubricants may be additionally added to formulate injectable formulations, capsules, granules or tablets such as aqueous solutions, suspensions, emulsions and the like.
또한, 본 발명의 사료 조성물은 보조성분으로 아미노산, 무기염류, 비타민, 항산화제, 항진균제, 항균제 등과 같은 각종 보조제 및 분쇄 또는 파쇄된 밀, 보리, 옥수수 등의 식물성 단백질 사료, 혈분, 육분, 생선분 등의 동물성 단백질 사료, 동물성 지방 및 식물성 지방 같은 주성분 이외에도 영양 보충제, 성장 촉진제, 소화 흡수 촉진제, 질병 예방제와 함께 사용될 수 있다.In addition, the feed composition of the present invention is a supplementary ingredient such as amino acids, inorganic salts, vitamins, antioxidants, antifungal agents, antibacterial agents, and vegetable protein feed such as milled or crushed wheat, barley, corn, blood meal, meat meal, fish meal as auxiliary ingredients. In addition to the main components such as animal protein feed, animal fat and vegetable fat, such as can be used with nutritional supplements, growth promoters, digestive absorption accelerators, disease prevention agents.
본 발명의 사료 조성물을 사료 첨가물로 사용할 경우, 상기 사료 조성물을 그대로 첨가하거나 다른 성분과 함께 사용될 수 있고, 통상적인 방법에 따라 적절하게 사용될 수 있다. 사료 조성물의 투여 형태는 비독성 제약상 허용 가능한 담체와 조합하여 즉시 방출 또는 서방성 제형으로 제조할 수 있다. 이러한 식용 담체는 옥수수 전분, 락토스, 수크로스, 프로필렌 글리콜일 수 있다. 고체형 담체의 경우에는 정제, 산제, 토로키제 등의 투여 형태일 수 있으며 액체형 담체의 경우에는 시럽제, 액체 현탁액제, 에멀젼제, 용액제 등의 투여 형태일 수 있다. 또한, 투여제는 보존제, 윤화제, 용액 촉진제, 안정화제를 함유할 수 있으며 다른 염증 질환 개선제 및 바이러스 예방상 유용한 물질을 함유할 수도 있다.When the feed composition of the present invention is used as a feed additive, the feed composition may be added as it is or used together with other ingredients, and may be suitably used according to a conventional method. Dosage forms of the feed composition may be prepared in immediate release or sustained release formulation in combination with a nontoxic pharmaceutically acceptable carrier. Such edible carriers may be corn starch, lactose, sucrose, propylene glycol. In the case of a solid carrier, it may be a dosage form such as a tablet, a powder, a torokee agent, or the like, and in the case of a liquid carrier, it may be a dosage form of a syrup, a liquid suspension, an emulsion, or a solution. In addition, the dosage may contain preservatives, lubricants, solution promoters, stabilizers, and may also contain other inflammatory disease ameliorating agents and substances useful for virus prevention.
본 발명의 사료 조성물은 포유류, 가금류, 어류 및 갑각류를 포함하는 다수의 동물 식이 즉, 사료에 적용할 수 있다. 상업용으로 중요한 돼지, 소, 염소 등의 포유류, 코끼리, 낙타 등의 동물원 동물, 개, 고양이 등의 가축에게 사용할 수 있다. 상업적으로 중요한 가금류에는 닭, 오리, 거위 등이 포함되며, 송어와 새우와 같은 상업적으로 사육되는 어류 및 갑각류를 포함 할 수 있다.The feed composition of the present invention is applicable to a number of animal diets, ie feeds, including mammals, poultry, fish and shellfish. Commercially important mammals such as pigs, cows and goats, zoo animals such as elephants and camels, and livestock such as dogs and cats. Commercially important poultry includes chickens, ducks, geese and the like, and may include commercially raised fish and crustaceans such as trout and shrimp.
본 발명에 따른 사료 조성물은 가축사료에 건조 중량 기준으로 1 ㎏ 당 약 10 내지 500 g, 바람직하기로는 10 내지 100 g의 양으로 혼합될 수 있고, 완전히 혼합한 후 매쉬로 공급하거나, 추가 가공 공정을 통하여 팰렛화, 팽창화, 압출 공정을 거치는 것이 바람직하다.The feed composition according to the present invention may be mixed in a livestock feed in an amount of about 10 to 500 g, preferably 10 to 100 g per kg, on a dry weight basis, and then thoroughly mixed and then fed into a mesh or further processing. Palletization, expansion, extrusion through the process is preferred.
본 발명의 구체적인 실시예에서는, 벌개미취의 추출물을 도파민성 세포인 SH-SY5Y 세포주에 처리한 결과, 자가포식소체의 막단백질인 LC3 단백질 발현이 증가됨을 확인하였다(실시예 1-2 참조). 또한 인산화된 AMPK 단백질 발현이 증가됨을 확인하였다(실시예 1-3 참조). In a specific example of the present invention, as a result of treatment of the extract of bee anesthetized with SH-SY5Y cell line, which is a dopaminergic cell, it was confirmed that the expression of LC3 protein, which is a membrane protein of autophagosome, was increased (see Example 1-2). It was also confirmed that the phosphorylated AMPK protein expression was increased (see Example 1-3).
본 발명의 또 다른 구체적인 실시예에서는, 벌개미취의 추출물이 인산화된 ULK1 단백질 발현을 증가하는 것을 세포실험을 통하여 웨스턴블롯팅으로 확인하였다(실시예 1-4 참조). In another specific embodiment of the present invention, it was confirmed by Western blotting through the cell experiment that the extract of the hummingbird odor increased phosphorylated ULK1 protein expression (see Example 1-4).
본 발명의 다른 일 실시예에서는, 벌개미취의 추출물이 mTOR 단백질 발현을 감소하는 것을 세포실험을 통하여 웨스턴블롯팅으로 확인하였다(실시예 1-5 참조)In another embodiment of the present invention, it was confirmed by Western blotting through the cell experiment that the extract of the hummingbird anesthetize reduces mTOR protein expression (see Example 1-5).
본 발명의 다른 일 실시예에서는, 벌개미취 추출물의 자가포식 작용 유도 효과를 확인하기 위해 autophagy inhibitor인 Bafilomycin A1을 통해 autophagic flux를 억제하는 웨스턴블롯팅으로 확인하였다(실시예 1-6 참조).In another embodiment of the present invention, it was confirmed by Western blotting to suppress the autophagic flux through the autophagy inhibitor Bafilomycin A1 in order to confirm the effect of induction of autophagy action of the bee odorant extract (see Example 1-6).
본 발명의 또 다른 일 실시예에서는, 벌개미취의 분획물에 의한 LC3 단백질 발현량을 확인한 결과, BuOH 분획물에 의해 LC3 단백질 발현이 증가되는 것을 확인하였다(실시예 2-1, 2 참조).In another embodiment of the present invention, as a result of confirming the expression level of the LC3 protein by the fraction of the antler odor, it was confirmed that the expression of the LC3 protein is increased by the BuOH fraction (see Examples 2-1 and 2).
본 발명의 또 다른 일 실시예에서는, 벌개미취 BuOH 분획물이 mTOR 단백질 발현을 증가하는 것을 세포실험을 통하여 웨스턴블롯팅으로 확인하였다(실시예 2-3 참조).In another embodiment of the present invention, it was confirmed by Western blotting through the cell experiment that the hummingbird BuOH fraction increased the expression of mTOR protein (see Example 2-3).
본 발명의 또 다른 일 실시예에서는, 벌개미취의 추출물이 MPTP를 통해 파킨슨병이 유도된 동물 모델에 투여한 결과, 자가포식작용 유도를 통해 뇌세포 사멸을 효과적으로 억제할 수 있는 활성을 웨스턴블롯팅으로 확인하였다(실시예 3 참조).In another embodiment of the present invention, as a result of administering the extract of the hummingbird to the Parkinson's disease-induced animal model through MPTP, the activity that can effectively inhibit brain cell death through induction of autophagy by Western blotting It confirmed (refer Example 3).
이로서 본 발명의 조성물은 도파민성 세포주에서 인산화된 AMPK의 단백질 발현량 증가에 의해 인산화된 ULK1의 단백질 발현량이 증가되고 mTOR 단백질 발현을 억제하며, MPTP를 통해 파킨슨병이 유도된 동물 모델에서 자가포식 작용의 유도를 통해 뇌세포 사멸을 효과적으로 억제할 수 있어 파킨슨병의 예방, 치료 및 개선에 탁월한 효과가 있음을 확인하였다.As such, the composition of the present invention increases the protein expression level of phosphorylated ULK1 and inhibits mTOR protein expression by increasing the protein expression level of phosphorylated AMPK in dopaminergic cell lines, and autophagy in an animal model induced by Parkinson's disease through MPTP. Through the induction of the brain cell death can be effectively suppressed, it was confirmed that it has an excellent effect on the prevention, treatment and improvement of Parkinson's disease.
즉, 본 발명의 벌개미취 추출물은 자가포식 작용을 유도하여 파킨슨병의 궁극적인 원인으로 알려진 알파-시누클레인(alpha-synuclein) 단백질의 분해를 촉진하는 효과가 매우 우수하여, 파킨슨병의 예방 또는 치료 및 개선에 효과적으로 사용될 수 있는 조성물임을 확인하였다. In other words, the bee anesthetized extract of the present invention has an excellent effect of inducing autophagy and promoting the degradation of alpha-synuclein protein, which is known as the ultimate cause of Parkinson's disease, thereby preventing or treating Parkinson's disease. It was confirmed that the composition can be effectively used for improvement.
이하 본 발명을 실시예 및 실험예를 통하여 보다 상세하게 설명한다. 그러나 이들 실시예 및 실험예는 본 발명을 예시적으로 설명하기 위한 것으로 본 발명의 범위가 이들 실시예 및 실험예에 한정되는 것은 아니다.Hereinafter, the present invention will be described in more detail with reference to Examples and Experimental Examples. However, these Examples and Experimental Examples are for illustrative purposes only and the scope of the present invention is not limited to these Examples and Experimental Examples.
실시예 1: 벌개미취 추출물의 자가포식 작용 유도 효과Example 1 Induction of Autophagy Action by Honey Bee Extract
실시예 1-1: 벌개미취 추출물의 제조Example 1-1 Preparation of Beetle Anesthetic Extract
벌개미취는 강원도 정선국 북평면에서 재배한 것으로 세척 후 완전 건조하여 건조 중량 15 kg을 얻었고, 시료를 분쇄기에 분쇄한 후 95% 에탄올로 60~70℃에서 3시간 동안 2회 감압 추출하여 95% 에탄올 추출물 1.7kg을 얻었다. Beetle odor was cultivated in Bukpyeong-myeon, Jeongseon-gu, Gangwon-do, and washed and dried completely to obtain 15 kg of dry weight.The sample was crushed in a grinder and extracted with 95% ethanol twice under reduced pressure for 2 hours at 60 ~ 70 ℃. kg was obtained.
실시예 1-2: 벌개미취 추출물에 의한 LC3 단백질 발현량 증가 효과Example 1-2: Effect of Increasing the Expression Level of LC3 Protein by the Beetle Odor Extract
상기 실시예 1에서 수득한 벌개미취 추출물의 LC3 단백질 발현량 증가 효과를 측정하기 위하여, 도파민성 세포주인 SH-SY5Y 세포주를 Gibco사(Grand Island, NY, USA)의 10% heat-inactivated fetal bovine serum (이하 FBS라 함)과 1% penicillin/streptomycin (이하 P/S라 함, Gibco)이 함유된 Dulbecco’s modified eagle medium nutrient mixture F-12 (Ham) 1x; DMEM/F12 (Gibco) 배지에서 배양하여 사용하였다. 이 세포주는 ATCC: The Global Bioresource Center (CRL-2266™)(Manassas, Virgina 20108, USA)로부터 구입하였다.In order to measure the effect of increasing the expression level of the LC3 protein of the honey bee extract obtained in Example 1, the dopaminergic cell line SH-SY5Y cell line was 10% heat-inactivated fetal bovine serum of Gibco (Grand Island, NY, USA) Dulbecco's modified eagle medium nutrient mixture F-12 (Ham) 1 × containing 1% penicillin / streptomycin (hereinafter referred to as P / S, Gibco); Cultured in DMEM / F12 (Gibco) medium was used. This cell line was purchased from ATCC: The Global Bioresource Center (CRL-2266 ™) (Manassas, Virgina 20108, USA).
구체적으로 상기 세포주 (SH-SY5Y)에 실시예 1에서 수득한 벌개미취 추출물을 도 1에 기재된 첨가량으로 첨가한 다음, 5% CO2, 37℃에서 24시간 동안 배양하고, 포집하여 13,000 rpm에서 6분간 원심분리하여 DPBS (Dulbecco’s phosphate buffered saline)로 2회 세척하였다. 얻어진 세포에 lysis solution (1X lysis buffer, 1X Protease inhibitor cocktail, 1x Phenylmethyl sulfonyl fluoride)를 첨가하여 4℃ 조건에서 30분간 반응시키고, 그 후 동일한 4℃ 조건에서 30분간 진탕하면서 완전히 용해(lysis) 시킨 다음 13,000 rpm에서 20분간 원심 분리하여 단백질이 방출된 상층액을 취하였다. 취한 상층액은 Protein assay dye Reagent concentrate (Bio-Rad)를 이용하여 Bradford method로 단백질을 정량하였다.Specifically, the bee-flavored extract obtained in Example 1 was added to the cell line (SH-SY5Y) in the amount shown in FIG. 1, then incubated at 5% CO 2 , 37 ° C. for 24 hours, and collected for 6 minutes at 13,000 rpm. Centrifugation was washed twice with Dulbecco's phosphate buffered saline (DPBS). After lysis solution (1X lysis buffer, 1X Protease inhibitor cocktail, 1x Phenylmethyl sulfonyl fluoride) was added to the obtained cells, the cells were reacted at 4 ° C for 30 minutes, and then completely lysed while shaking at the same 4 ° C for 30 minutes. Centrifugation was performed at 13,000 rpm for 20 minutes to obtain the supernatant from which the protein was released. The supernatant was quantified by Bradford method using Protein assay dye Reagent concentrate (Bio-Rad).
동량의 세포파쇄액은 SDS loading buffer와 혼합하여 99℃에서 5분간 가열한 후에 15% SDS-폴리아크릴아마이드 겔에서 분리를 하고(Bio-Rad), semi dry transfer 방법을 통해 약 45분간 polyvinylidene fluoride membrane (PVDF)(Millipore)에 이동시켰다. Transfer된 PVDF membrane을 blocking buffer (5% skim milk, 3% bovine serum albumin)를 통해 1시간~1시간 30분 동안 반응시켜 blocking 하였다. blocking buffer에 각 항체를 1,000배로 희석한 후, PVDF membrane을 4℃ 조건에서 하룻밤 동안 반응시켰다. 본 실험에서 사용된 1차 항체는 LC3, GAPDH (Cell signaling technology)이 있으며, GAPDH는 loading control로 사용하였다. 그 후 다음날 TBST buffer (Trizma base, sodium chloride, Tween 20)를 사용하여 10분/1회, 3회 세척한 후 blocking buffer에 1:2,000의 비율로 희석한 anti-rabbit lgG secondary항체(Cell signaling technology)를 이용하여 상온에서 2시간 동안 반응시켰으며, 동일한 조건으로 세척하였다. 그 후 ECL 용액(Thermo Fisher Scientific, Bio-Rad, Ab FRONTIRE)을 이용하여 membrane을 형광 발색 시켜 LAS 4000 film (Fujifilm)을 이용해 단백질의 발현 양상을 확인하였다.The same amount of cell lysate was mixed with SDS loading buffer and heated at 99 ° C. for 5 minutes, then separated from 15% SDS-polyacrylamide gel (Bio-Rad), and the polyvinylidene fluoride membrane for about 45 minutes by semi dry transfer method. (PVDF) (Millipore). The transferred PVDF membrane was blocked by reacting with blocking buffer (5% skim milk, 3% bovine serum albumin) for 1 hour to 1 hour 30 minutes. After diluting each antibody 1,000-fold in blocking buffer, the PVDF membrane was reacted overnight at 4 ° C. Primary antibodies used in this experiment were LC3, GAPDH (Cell signaling technology), GAPDH was used as a loading control. The next day, washed with TBST buffer (Trizma base, sodium chloride, Tween 20) for 10 minutes / 1 time, 3 times, and then diluted anti-rabbit lgG secondary antibody (Cell signaling technology) in a ratio of 1: 2,000 in blocking buffer. ) Was reacted at room temperature for 2 hours, and washed under the same conditions. Afterwards, the membrane was fluorescently colored using an ECL solution (Thermo Fisher Scientific, Bio-Rad, Ab FRONTIRE) to confirm the expression of the protein using the LAS 4000 film (Fujifilm).
이후 LC3Ⅰ 단백질에서 LC3Ⅱ 단백질로 전이되는 비율을 확인한 결과를 도 1에 기재하였다. 이때 벌개미취 추출물을 첨가하지 않은 경우를 대조군으로 하였다. 그 결과, 도 1에서 확인할 수 있는 바와 같이, 대조군에 비하여 벌개미취 추출물의 첨가에 의해 LC3 단백질의 발현량이 농도의존적으로 증가되는 것을 확인할 수 있었으며, 특히 추출물을 50 μg/ml 농도로 처리하였을 때, LC3Ⅰ 단백질에서 LC3Ⅱ 단백질로 전이되는 비율이 2.05인 것으로 확인하였다. 이를 통해, 벌개미취 추출물이 도파민성 세포주에 처리되었을 때, 자가포식소폐 막 단백질 중 하나인 LC3 단백질의 발현량을 증가시키고, 자가포식 활성을 나타내는 지표인 LC3 I에서 LC3 II 단백질로의 전이를 증가시켜, 결과적으로 자가포식 유도 효과를 나타냄을 확인할 수 있다.Then, the results of confirming the rate of transition from the LC3I protein to the LC3II protein are described in FIG. 1. In this case, the case where the bee anesthetist extract was not added was used as a control. As a result, as can be seen in Figure 1, it was confirmed that the expression level of the LC3 protein was increased in a concentration-dependent manner by the addition of the beetail odor extract compared to the control, especially when the extract was treated at a concentration of 50 μg / ml, LC3 I The ratio of protein to LC3II protein was found to be 2.05. This resulted in increased expression of LC3 protein, which is one of autophagy membrane proteins, and increased transition from LC3 I to LC3 II protein, which is a marker of autophagy activity, As a result, it can be seen that it exhibits an autophagy induction effect.
실시예 1-3: 벌개미취 추출물에 의한 인산화된 AMPK 단백질 발현량 증가 효과Example 1-3: Effect of Increasing Phosphorylated AMPK Protein Expression by Beetle Anesthetic Extract
상기 실시예 1에서 수득한 벌개미취 추출물의 인산화된 AMPK 단백질 발현량 증가 효과를 측정하기 위하여, 배양한 SH-SY5Y 세포주에 도 2에 기재된 벌개미취 추출물 첨가량으로 첨가한 다음, 5% CO2, 37℃에서 24시간 동안 배양하고, 실시예 2와 동일한 방법으로 실험하였다. 본 실험에서 사용된 1차 항체는 AMPK, p-AMPK, GAPDH (Cell signaling technology)이 있으며, GAPDH는 loading control로 사용하였다. In order to measure the effect of increasing the expression level of phosphorylated AMPK protein of the bee-flavored extract obtained in Example 1, it was added to the cultured SH-SY5Y cell line in the amount of the bee-flavored extract described in Figure 2, at 5% CO 2 , 37 ℃ Incubated for 24 hours, and experimented in the same manner as in Example 2. Primary antibodies used in this experiment are AMPK, p-AMPK, GAPDH (Cell signaling technology), GAPDH was used as a loading control.
이후 전체 AMPK(t-AMPK) 단백질 중 인산화된 AMPK(p-AMPK) 단백질의 상대적 밀도를 나타내는 결과를 도 2에 기재하였고, 벌개미취 추출물을 처리하지 않은 경우를 대조군으로 하였다. 그 결과, 도 2에서 확인할 수 있는 바와 같이, 대조군에 비하여 벌개미취 추출물에 의해 인산화된 AMPK 단백질의 발현량이 농도의존적으로 증가되는 것을 확인할 수 있었다. 이를 통해, 벌개미취 추출물이 도파민성 세포주에 처리되었을 때, 자가포식을 유도하는 인산화된 AMPK 단백질의 발현량을 증가시켜, 결과적으로 자가포식 유도 효과를 나타냄을 확인할 수 있다.Since the results showing the relative density of the phosphorylated AMPK (p-AMPK) protein in the total AMPK (t-AMPK) protein is described in Figure 2, the case was not treated with the bee sting extract was used as a control. As a result, as can be seen in Figure 2, compared with the control group it was confirmed that the expression level of the phosphorylated AMPK protein by the bee anesthetized extract increased concentration-dependently. Through this, it can be confirmed that when the bee odor extract was treated to the dopaminergic cell line, the expression level of the phosphorylated AMPK protein that induces autophagy was increased, resulting in the effect of autophagy induction.
실시예 1-4: 벌개미취 추출물에 의한 인산화된 ULK1 단백질 발현량 증가 효과Example 1-4: Effect of Increasing Phosphorylated ULK1 Protein Expression by Beetle Odor Extract
벌개미취 추출물의 인산화된 ULK1 단백질 발현량 증가 효과를 확인하기 위해, 배양한 SH-SY5Y 세포주에 도 3에 기재된 벌개미취 추출물 첨가량으로 첨가한 다음, 5% CO2, 37℃에서 24시간 동안 배양하여 상시 실시예 2와 같은 방법으로 실험하였다. 본 실험에서 사용된 1차 항체는 ULK1, p-ULK1(s555), GAPDH (Cell signaling technology)이 있으며, GAPDH는 loading control로 사용하였다.In order to confirm the effect of increasing the expression level of phosphorylated ULK1 protein of the bee-flavored extract, it was added to the cultured SH-SY5Y cell line with the amount of the bee-flavored extract described in FIG. 3, followed by culturing for 24 hours at 5% CO 2 , 37 ° C. The experiment was carried out in the same manner as in Example 2. The primary antibodies used in this experiment include ULK1, p-ULK1 (s555), GAPDH (Cell signaling technology), GAPDH was used as a loading control.
이후 전체 ULK1 단백질(t-ULK1) 중 인산화된 ULK1 단백질(p-ULK1)의 상대적 밀도를 나타내는 결과를 도 3에 기재하였고, 벌개미취 추출물을 처리하지 않은 경우를 대조군으로 하였다. 그 결과, 도 3에서 확인할 수 있는 바와 같이, 대조군에 비하여 벌개미취 추출물에 의해 인산화된 ULK1 단백질의 발현량이 농도의존적으로 증가되는 것을 확인할 수 있었다. 이를 통해, 벌개미취 추출물이 도파민성 세포주에 처리되었을 때, 자가포식을 유도하는 인산화된 ULK1 단백질의 발현량을 증가시켜, 결과적으로 자가포식 유도 효과를 나타냄을 확인할 수 있다.Then, the results showing the relative density of the phosphorylated ULK1 protein (p-ULK1) in the total ULK1 protein (t-ULK1) is described in Figure 3, the case was not treated with the bee hijab extract as a control. As a result, as can be seen in Figure 3, compared with the control group it was confirmed that the expression level of the phosphorylated ULK1 protein phosphorylated by the bee sting anesthetic extract increased. Through this, it can be confirmed that when the bee odor extract was treated to the dopaminergic cell line, the expression level of the phosphorylated ULK1 protein that induces autophagy was increased, resulting in an autophagy induction effect.
실시예 1-5: 벌개미취 추출물에 의한 mTOR 단백질 발현 증가 효과Example 1-5: Effect of Increasing Expression of mTOR Protein by Beetle Odor Extract
벌개미취 추출물의 mTOR 단백질 발현량 억제 효과를 확인하기 위해, 배양한 SH-SY5Y 세포주에 도 4에 기재된 벌개미취 에탄올 추출물 첨가량으로 첨가한 다음, 5% CO2, 37℃에서 24시간 동안 배양하여 상시 실시예 2와 같은 방법으로 실험하였다. 본 실험에서 사용된 1차 항체는 mTOR, GAPDH (Cell signaling technology)이 있으며, GAPDH는 loading control로 사용하였다.In order to confirm the inhibitory effect of the mTOR protein expression of the bee odor extract, added to the cultured SH-SY5Y cell line with the addition of the bee odor ethanol extract shown in Figure 4, and then incubated for 24 hours at 5% CO 2 , 37 ℃ always Experiment was carried out in the same manner as 2. Primary antibodies used in this experiment are mTOR, GAPDH (Cell signaling technology), GAPDH was used as a loading control.
이후 mTOR의 상대적 밀도를 나타내는 결과를 도 4에 기재하였고, 벌개미취 추출물을 처리하지 않은 경우를 대조군으로 하였다. 그 결과, 도 4에서 확인할 수 있는 바와 같이, 대조군에 비하여 벌개미취 추출물에 의해 mTOR 단백질의 발현이 농도의존적으로 감소되는 것을 확인할 수 있었다. 이를 통해, 벌개미취 추출물이 도파민성 세포주에 처리되었을 때, 자가포식을 억제하는 mTOR 단백질의 발현량을 감소시켜, 결과적으로 자가포식 유도 효과를 나타냄을 확인할 수 있다.Since the results showing the relative density of mTOR is described in Figure 4, the case was not treated with the bee herb extract was used as a control. As a result, as can be seen in Figure 4, it was confirmed that the expression of mTOR protein is reduced in a concentration-dependent manner by the bee stinger extract compared to the control group. Through this, it can be confirmed that when the bee sting odor extract was treated to the dopaminergic cell line, the expression level of mTOR protein that inhibits autophagy was reduced, resulting in an autophagy induction effect.
실시예 1-6: 자가포식 억제제 Bafilomycin A를 통한 벌개미취 추출물의 자가포식 작용 유도 효과Example 1-6 Effect of Induction of Autophagy Action on Beetle Anesthetic Extracts Through Autophagy Inhibitor Bafilomycin A
벌개미취 추출물의 자가포식 작용 유도 효과가 위양성(false positive)에 의한 비특이적인 결과로 나타난 현상인지, 진정한 자가포식 유도 효과에 의한 것인지를 확인하기 위해, 배양한 SH-SY5Y 세포주에 실시예 1에서 수득한 벌개미취 추출물을 도 5에 기재된 것과 같이 50 μg/ml 농도로 18시간 처리한 뒤 100nM bafilomycin A1 (이하, baf A1)을 처리하고, 6시간 동안 배양하였다. 그 후 실시예 2와 같은 방법으로 웨스턴블롯팅으로 LC3 단백질 발현량을 측정하였다. In order to confirm whether the effect of inducing autophagy action of the bee-flavored extract was a non-specific result of false positive or true autophagy induction effect, the cultured SH-SY5Y cell line was obtained in Example 1 Beetle odor extract was treated for 18 hours at 50 μg / ml concentration as described in Figure 5 and then treated with 100nM bafilomycin A1 (hereinafter, baf A1) and incubated for 6 hours. Thereafter, the amount of LC3 protein expression was measured by Western blotting in the same manner as in Example 2.
Baf A1은 자가포식소체와 리소좀의 융합을 억제함으로써, 자가포식소포(autophagic vacuoles)의 성숙(maturation)을 저해하는 자가포식 억제제(autophagy inhibitor)이다. 구체적으로, 자가포식 경로의 마지막 단계에서 오토파지 플럭스(autophagic flux)가 일어나야 비로소 자가포식 유도가 일어나게 되지만, baf A1을 처리하는 경우에는 오토파지 플럭스가 억제됨에 따라 LC3 단백질 발현이 증가하게 된다. Baf A1 is an autophagy inhibitor that inhibits the fusion of autophagosomes and lysosomes, thereby inhibiting the maturation of autophagic vacuoles. Specifically, autophagy flux occurs only at the last stage of the autophagy route, but autophagy induction occurs. However, when baf A1 is treated, LC3 protein expression is increased as autophagy flux is suppressed.
그 결과, 도 5에서 확인할 수 있는 바와 같이, Baf A1을 처리한 군의 LC3 단백질의 발현이 대조군에 비해 유의적으로 증가하는 것을 통해 오토파지 플럭스가 억제된 것을 확인할 수 있었다. 또한, 벌개미취 추출물과 baf A1을 함께 처리 하였을 때 벌개미취 추출물 단독 처리 군에 비해 LC3 단백질의 발현(LC3 I에 대한 LC3 II의 상대적 밀도)이 유의적으로 증가하는 것을 확인하였다. 이를 통해, 벌개미취 추출물의 자가포식 작용 유도 효과가 오토파지 플럭스의 조절을 통한 것임을 알 수 있었다.As a result, as can be seen in Figure 5, it was confirmed that the autophagy flux was inhibited by the significantly increased expression of the LC3 protein of the Baf A1 treated group compared to the control. In addition, it was confirmed that the expression of LC3 protein (relative density of LC3 II relative to LC3 I) was significantly increased when the bee anesthetist extract and baf A1 were treated together. Through this, it can be seen that the effect of inducing the autophagy action of the bee anesthesia extract through the control of the autophagy flux.
실시예 2: 벌개미취 분획물의 자가포식 작용 유도 효과Example 2: Induction of Autophagy Action by Honey Bee Fractions
실시예 2-1: 벌개미취 분획물의 제조Example 2-1 Preparation of Beetle Odor Fraction
95% 에탄올 추출하여 얻은 벌개미취 에탄올 추출물 1.7 kg 중 1 kg을 H2O로 현탁시킨 다음 동량의 헥산(n-Hexane)을 가하여 진탕 방치하여 헥산 분획(149 g)을 얻고, 수층에 동량의 에틸아세테이트(EtOAc)를 가하여 진탕 방치하여 에틸아세테이트 분획(175 g)을 얻고, 수층에 다시 동량의 부탄올(n-BuOH)을 가하여 진탕 방치하여 부탄올 분획(190 g)을 얻었으며, 수층을 농축하여 물(H2O) 분획을 얻었다. 1 kg of the bee-flavored ethanol extract 1.7 kg obtained by 95% ethanol extraction was suspended with H 2 O, and the mixture was left to shake with the same amount of hexane ( n -Hexane) to obtain a hexane fraction (149 g). The same amount of ethyl acetate was added to the aqueous layer. (EtOAc) was added to the mixture, and the mixture was left to shake to obtain an ethyl acetate fraction (175 g). An equal amount of butanol ( n -BuOH) was added to the aqueous layer and the mixture was left to shake to obtain a butanol fraction (190 g). H 2 O) fractions were obtained.
실시예 2-2: 벌개미취 분획물에 의한 LC3 단백질 발현량 증가 효과Example 2-2: Effect of Increasing LC3 Protein Expression by Honey Bee Fraction
상기 실시예 7에서 수득한 벌개미취 분획물의 LC3 단백질 발현 증가 효과를 측정하기 위하여, 상기 세포주 (SH-SY5Y cell)에 벌개미취 분획물을 분획물 독성에 따라 각각 5, 10, 25, 50, 100 μg/ml로 첨가한 다음, 5% CO2, 37℃에서 24시간 동안 배양하고, 실시예 2와 같은 방법의 웨스턴블롯팅 실험을 통하여 확인하였다. In order to measure the effect of increasing the expression level of LC3 protein of the bee-flavored fraction obtained in Example 7, the bee-flavored fraction was added to the cell line (SH-SY5Y cell) at 5, 10, 25, 50, and 100 μg / ml depending on the fraction toxicity. After the addition, 5% CO 2 , and incubated for 24 hours at 37 ℃, it was confirmed through the Western blotting experiment of the same method as Example 2.
상기 LC3 단백질 발현을 확인한 결과를 도 6에 기재하였고, 벌개미취 분획물을 첨가하지 않은 경우를 대조군으로 하였다. 그 결과, 도 6에서 확인할 수 있는 바와 같이, 벌개미취 분획물에 의해 LC3 II 단백질의 발현이 유의적으로 증가함을 확인할 수 있었다. 이를 통해, 벌개미취 분획물이 도파민성 세포주에 처리되었을 때, 자가포식 활성을 나타내는 지표인 LC3 I에서 LC3 II 단백질로의 전이를 증가시켰는바, 결과적으로 자가포식 유도 효과를 나타냄을 확인할 수 있다.The results of confirming the expression of the LC3 protein are shown in FIG. 6, and the case where no bee-flavored fraction was added was used as a control. As a result, as can be seen in Figure 6, it was confirmed that the expression of the LC3 II protein significantly increased by the bee odor fraction. Through this, it was confirmed that when the hummingbird fraction was treated to the dopaminergic cell line, the transition from LC3 I, which is an indicator of autophagy activity, to LC3 II protein was increased, resulting in an autophagy induction effect.
실시예 2-3: 벌개미취 분획물에 의한 mTOR 단백질 발현 증가 효과Example 2-3: Effect of Increasing mTOR Protein Expression by Honey Bee Fraction
상시 실시예 7에서 수득한 벌개미취 BuOH 분획물의 mTOR 단백질 발현 감소 효과를 측정하기 위하여, SH-SY5Y 세포주에 벌개미취 분획물을 도 7에 기재된 바와 같이 첨가한 다음, 5% CO2, 37℃에서 24시간 동안 배양하고, 실시예 2와 같은 방법으로 실험하였다. 본 실험에서 사용된 1차 항체는 mTOR, GAPDH (Cell signaling technology)이 있으며, GAPDH는 loading control로 사용하였다.In order to determine the effect of reducing mTOR protein expression of the hummingbird BuOH fraction obtained in Example 7 at all, the hummingbird fraction was added to the SH-SY5Y cell line as described in FIG. 7, followed by 24% at 5% CO 2 , 37 ° C. The culture was carried out in the same manner as in Example 2. Primary antibodies used in this experiment are mTOR, GAPDH (Cell signaling technology), GAPDH was used as a loading control.
상기 mTOR의 상대적 밀도를 나타낸 결과를 도 7에 기재하였고, 벌개미취 분획물을 첨가하지 않은 경우를 대조군으로 하였다. 그 결과, 도 7에서 확인할 수 있는 바와 같이, 벌개미취 분획물을 처리한 경우 농도의존적으로 mTOR 단백질의 발현이 유의적으로 감소함을 확인 할 수 있었다. 이를 통해, 벌개미취 분획물이 도파민성 세포주에 처리되었을 때, 자가포식을 억제하는 mTOR 단백질의 발현량을 감소시켜, 결과적으로 자가포식 유도 효과를 나타냄을 확인할 수 있다The results showing the relative density of the mTOR is shown in FIG. 7, and the case where no bee sting fraction was added was used as a control. As a result, as can be seen in Figure 7, it was confirmed that the expression of mTOR protein significantly reduced in the concentration-dependent treatment when the bee odor fraction. Through this, it can be confirmed that when the bee odor fraction was treated in the dopaminergic cell line, the expression level of mTOR protein that inhibited autophagy was reduced, resulting in an autophagy induction effect.
실시예 3: MPTP 투여에 의한 파킨슨병 동물모델에서, 벌개미취 에탄올 추출물에 의한 자가포식 작용을 통하여 뇌세포 사멸에 미치는 효과Example 3: Effect of MPTP on Parkinson's Disease Animal Model on Brain Cell Death through Autophagy by Beetle-odorous Ethanol Extract
C57BL/6 웅성 마우스(8주령)를 사용하고, 사료와 물은 자유 섭취하였다. 마우스를 각 군당 6마리씩 7군으로 나누었다. 대조군과 MPTP 단독처리군은 생리식염수를 3일간 단회 경구 투여하였고, 상기 실시예 1-1에서 수득한 벌개미취 추출물(200 mg/kg)군과 상기 실시예 2-1에서 수득한 벌개미취 분획물(25, 50, 100 mg/kg)군은 생리식염수에 각각의 추출물 및 분획물을 용해시킨 뒤 3일간 단회 경구 투여하였다. 양성대조군은 로피니롤을 생리식염수에 용해하여 3일간 단회 경구 투여하였다. 약물 투여 4일째부터 8일째까지는 대조군을 제외한 모든 군에 약물 투여 1시간 후에 생리식염수에 용해한 MPTP(30 mg/kg)를 단회 경구 투여하였으며, 약물 투여 9일째부터 10일째에는 대조군을 제외한 모든 군에 동일한 양의 약물을 단회 경구 투여하였다. C57BL / 6 male mice (8 weeks old) were used and feed and water were freely ingested. Mice were divided into seven groups, six in each group. The control group and the MPTP-treated group were orally administered with saline once 3 days, the bee odor extract (200 mg / kg) group obtained in Example 1-1 and the bee odor fraction obtained in Example 2-1 (25, 50, 100 mg / kg) group was dissolved orally in each saline extract and fractions and administered once orally for 3 days. In the positive control group, rofinirol was dissolved in physiological saline and administered once orally for 3 days. From 4th to 8th day of drug administration, MPTP (30 mg / kg) dissolved in physiological saline was administered orally to all groups except the control group 1 hour after drug administration, and to all groups except the control group on the 9th to 10th day of drug administration. The same amount of drug was administered orally once.
약물 투여 종료 후 7일째 각 군의 마우스를 치사시킨 후, 뇌 조직을 흑질(substantia nigra)과 선조체(striatum)로 분리하였다. 분리된 뇌 조직을 Phosphatase Inhibitor Cocktail Set I (Sigma-Aldrich, MO, USA)을 보충 한 PRO-PREPTM 용해 완충액(iNtRON, 경기, 한국)으로 균질화하였다. 균질액을 4℃에서 30분 동안 방치한 다음 13,000 rpm, 4℃, 30분간 원심 분리하여 상등액을 취하고, 취한 상층액은 Protein assay dye Reagent concentrate (Bio-Rad)를 이용하여 Bradford method로 단백질을 정량하였다. 그 후 일정량의 SDS loading buffer와 혼합하여 99℃에서 5분간 가열한 후에 상시 실시예 1-2와 같은 방법으로 웨스턴블롯팅을 통해 단백질 발현을 측정하였다. 본 실험에서 사용된 1차 항체는 TH, Beclin-1, p-mTOR, Mtor, p-ULK1(s555), ULK1, p-AMPK, AMPK, LC3, α-synuclein, GAPDH (Cell signaling technology)이 있으며, GAPDH는 loading control로 사용하였다.Seven days after the end of drug administration, mice in each group were killed and brain tissues were separated into substantia nigra and striatum. The isolated brain tissue was homogenized with PRO-PREPTM Lysis Buffer (iNtRON, Gyeonggi, Korea) supplemented with Phosphatase Inhibitor Cocktail Set I (Sigma-Aldrich, MO, USA). The homogenate was left at 4 ° C. for 30 minutes and then the supernatant was collected by centrifugation at 13,000 rpm, 4 ° C. for 30 minutes, and the supernatant was quantitated by Bradford method using Protein assay dye Reagent concentrate (Bio-Rad). It was. Thereafter, the mixture was mixed with a predetermined amount of SDS loading buffer and heated at 99 ° C. for 5 minutes, and protein expression was measured by Western blotting in the same manner as in Example 1-2. Primary antibodies used in this experiment include TH, Beclin-1, p-mTOR, Mtor, p-ULK1 (s555), ULK1, p-AMPK, AMPK, LC3, α-synuclein, Cell signaling technology (GAPDH). GAPDH was used as loading control.
상기 단백질들의 발현을 확인한 결과를 도 8 내지 도14에 기재하였고, 벌개미취 추출물, 분획물 및 MPTP를 첨가하지 않은 경우를 대조군으로 하였으며, 로피니롤과 MPTP를 투여한 경우를 양성 대조군으로 하였다. 로피니롤은 파킨슨병 약물 치료에서 사용되는 물질으로, 도파민 수용체의 작용제(dopamine agonist)이다. MPTP는 파킨슨병 동물 모델에서 사용되는 물질으로, 도파민 길항제(dopamine antagonist)이다.The results of confirming the expression of the proteins are described in Figures 8 to 14, the case of the addition of the bee hijab extract, fractions and MPTP was used as a control, and the case of administration of ropinilol and MPTP as a positive control. Ropinirol is a substance used in the treatment of Parkinson's disease medication and is a dopamine agonist. MPTP is a substance used in animal models of Parkinson's disease and is a dopamine antagonist.
그 결과, 도 8에서 확인할 수 있는 바와 같이, MPTP 단독처리군의 TH 발현이 대조군에 비해 감소된 반면에, 벌개미취 추출물 및 분획물을 투여한 군들에서는 농도의존적으로 도파민의 전구체(L-DOPA)를 조절하는 효소인 TH 단백질 발현이 유의적으로 증가하였음을 확인할 수 있다. As a result, as can be seen in Figure 8, while the THTP expression of the MPTP-treated group was reduced compared to the control group, the group administered the bee-flavored extract and fractions controlled the dopamine precursor (L-DOPA) in a concentration-dependent manner It can be seen that the expression of TH protein, which is an enzyme, increased significantly.
그 결과, 도 9에서 확인할 수 있는 바와 같이, MPTP 단독 처리군에서 α-synuclein의 응집이 대조군에 비해 유의적으로 증가하는 반면에, 벌개미취 추출물 및 분획물을 투여한 군에서는 파킨슨병의 원인 단백질로 알려져 있는 α-synuclein 단백질의 응집이 대조군 수준 또는 그 이하로 감소되었음을 확인할 수 있다. As a result, as can be seen in Figure 9, the aggregation of α-synuclein in the MPTP-treated group significantly increased compared to the control group, while the group that was administered the hive extract and fractions known as the cause of Parkinson's disease protein It can be seen that aggregation of the α-synuclein protein present is reduced to or below the control level.
또한, 도 10에서 확인할 수 있는 바와 같이, 대조군에 비해 벌개미취 추출물 및 분획물을 투여한 군의 자가포식소체 막 단백질인 LC3 단백질 발현이 농도의존적으로 증가함을 확인할 수 있었다. In addition, as can be seen in Figure 10, compared with the control group it was confirmed that the expression of LC3 protein, autophagosome membrane protein of the group that was administered the group of bee anesthetized extract and fraction increased concentration-dependently.
또한, 도 11에서 확인할 수 있는 바와 같이, MPTP 단독처리군의 경우, 자가포식 작용을 유도하는 인산화된 AMPK 단백질 발현이 대조군에 비해 감소한 반면에, 벌개미취 추출물 및 분획물을 투여한 군에서는 농도의존적으로 증가함을 확인할 수 있었다. Also, as can be seen in FIG. 11, in the MPTP-treated group, the expression of phosphorylated AMPK protein that induces autophagy was decreased in comparison with the control group, whereas in the group administered bee anesthetized extract and fraction, the concentration-dependent increase was observed. Could confirm.
그 결과, 도 12에서 확인할 수 있는 바와 같이, MPTP 단독처리군의 경우, 자가포식 작용을 유도하는 인산화된 ULK1 단백질 발현이 대조군에 비해 감소한 반면에, 벌개미취 추출물 및 분획물을 투여한 군에서 농도의존적으로 유의적으로 증가함을 확인할 수 있었다. As a result, As can be seen in FIG. 12, in the MPTP-treated group, the expression of phosphorylated ULK1 protein, which induces autophagy, was reduced compared to the control group, whereas the group receiving the bee-flavored extract and fractions was significantly concentration-dependently. It could be confirmed that the increase.
그 결과, 도 13에서 확인할 수 있는 바와 같이, MPTP 단독처리군의 경우, 자가포식 작용을 억제하는 인산화된 mTOR 단백질의 발현이 대조군에 비해 증가된 반면에, 벌개미취 추출물 및 분획물을 투여한 군에서는 농도 의존적으로 유의적으로 감소함을 확인할 수 있었다. As a result, as can be seen in Figure 13, in the MPTP-treated group, the expression of phosphorylated mTOR protein that inhibits autophagy was increased compared to the control group, while in the group to which the bee-flavored extract and fractions were administered, the concentration was increased. It was confirmed that the decrease significantly.
그 결과, 도 14에서 확인할 수 있는 바와 같이, MPTP 단독처리군의 경우. 자가포식 작용을 유도하는 대표적인 단백질인 Beclin-1 단백질의 발현이 대조군에 비해 감소하는 반면에, 벌개미취 추출물 및 분획물을 투여한 군에서는 농도의존적으로 유의적으로 증가하는 것을 확인할 수 있었다. As a result, as can be seen in Figure 14, in the case of the MPTP single treatment group. While the expression of Beclin-1 protein, a representative protein that induces autophagy, was decreased compared to the control group, it was confirmed that concentrations were significantly increased in the group administered bee anesthetized extract and fractions.
이상의 결과에서, MPTP 독성물질로 파킨슨병을 유도한 동물 모델에서 벌개미취 추출물 및 분획물의 처리는 도파민의 전구체(L-DOPA)를 조절하는 효소인 TH의 양을 촉진시키고(도 8), 파킨슨병에서 비정상적으로 응집된 형태로 나타나는 α-synuclein 단백질의 응집을 억제하고(도 9), 자가포식소페 막 단백질 LC3와 자가포식을 유도하는 p-AMPK, p-ULK1, Beclin-1 단백질의 발현을 증가시키고, 자가포식을 억제하는 p-mTOR 단백질의 발현을 감소시킴(도 10-14)을 확인하였고, 이를 통해, 벌개미취 추출물 및 분획물은 자가포식 작용의 유도를 통해 농도의존적으로 파킨슨병의 치료 및 개선에 활성을 나타냄을 확인할 수 있다. In the above results, the treatment of bee-flavored extracts and fractions in the Parkinson's disease-induced animal model with MPTP toxin promotes the amount of TH, an enzyme that modulates the precursor of dopamine (L-DOPA) (FIG. 8), and in Parkinson's disease. Inhibits aggregation of α-synuclein protein in abnormally aggregated form (FIG. 9), increases expression of autophagy membrane protein LC3 and p-AMPK, p-ULK1, Beclin-1 proteins that induce autophagy; It was confirmed that the expression of p-mTOR protein that inhibits autophagy is reduced (Figs. 10-14). Through this, bee-flavored extracts and fractions can be used to treat and improve Parkinson's disease in a concentration-dependent manner through the induction of autophagy. It can be seen that the activity.
이상의 설명으로부터, 본 발명이 속하는 기술분야의 당업자는 본 발명이 그 기술적 사상이나 필수적 특징을 변경하지 않고서 다른 구체적인 형태로 실시될 수 있다는 것을 이해할 수 있을 것이다. 이와 관련하여, 이상에서 기술한 실시예들은 모든 면에서 예시적인 것이며 한정적인 것이 아닌 것으로 이해해야만 한다. 본 발명의 범위는 상기 상세한 설명보다는 후술하는 특허 청구범위의 의미 및 범위 그리고 그 등가 개념으로부터 도출되는 모든 변경 또는 변형된 형태가 본 발명의 범위에 포함되는 것으로 해석되어야 한다.From the above description, those skilled in the art will appreciate that the present invention can be implemented in other specific forms without changing the technical spirit or essential features. In this regard, it should be understood that the embodiments described above are exemplary in all respects and not restrictive. The scope of the present invention should be construed that all changes or modifications derived from the meaning and scope of the following claims and equivalent concepts rather than the detailed description are included in the scope of the present invention.

Claims (9)

  1. 벌개미취 추출물 또는 이의 분획물을 유효성분으로 포함하는 파킨슨병의 예방 또는 치료용 약학적 조성물.A pharmaceutical composition for the prevention or treatment of Parkinson's disease comprising bee odor extract or fractions thereof as an active ingredient.
  2. 제1항에 있어서, 상기 벌개미취는 벌개미취의 잎 또는 줄기에서 추출된 것인, 약학적 조성물.The pharmaceutical composition of claim 1, wherein the bee odor is extracted from leaves or stems of the bee odor.
  3. 제1항에 있어서, 상기 벌개미취 추출물은 물, 탄소수 1 내지 4의 알코올, 및 이들의 혼합용매로 구성되는 군으로부터 선택되는 용매로 추출하여 제조되는 것인, 약학적 조성물.The pharmaceutical composition of claim 1, wherein the beetail odor extract is prepared by extraction with a solvent selected from the group consisting of water, alcohols having 1 to 4 carbon atoms, and mixed solvents thereof.
  4. 제1항에 있어서, 상기 파킨슨병의 예방 또는 치료는 자가포식 유도작용을 통한 뇌세포 사멸의 억제에 의한 것인, 약학적 조성물.The pharmaceutical composition of claim 1, wherein the prevention or treatment of Parkinson's disease is by inhibition of brain cell death through autophagy induction.
  5. 제1항 내지 제4항 중 어느 한 항에 따른 약학적 조성물을 개체에 투여하는 단계를 포함하는 파킨슨병의 예방 또는 치료 방법.A method of preventing or treating Parkinson's disease, comprising administering to a subject a pharmaceutical composition according to any one of claims 1 to 4.
  6. 벌개미취 추출물 또는 이의 분획물을 유효성분으로 포함하는, 파킨슨병의 예방 또는 개선용 식품 조성물.Beetle odor extract or fractions thereof as an active ingredient, a food composition for preventing or improving Parkinson's disease.
  7. 제6항에 있어서, 상기 파킨슨병의 예방 또는 치료는 자가포식 유도작용을 통한 뇌세포 사멸의 억제에 의한 것인, 식품 조성물.The food composition of claim 6, wherein the prevention or treatment of Parkinson's disease is by inhibition of brain cell death through autophagy induction.
  8. 벌개미취 추출물 또는 분획물을 유효성분으로 포함하는, 파킨슨병의 예방 또는 개선용 사료 조성물.Beetle odor extract or fraction comprising as an active ingredient, Parkinson's disease prevention or improvement of feed composition.
  9. 벌개미취 추출물 또는 분획물을 유효성분으로 포함하는, 인비트로(in vitro) 자가포식 유도에 의한 뇌세포 사멸 억제용 조성물.A composition for inhibiting brain cell death by in vitro autophagy induction, comprising bee anesthetized extract or fraction as an active ingredient.
PCT/KR2019/005352 2018-05-04 2019-05-03 Composition comprising aster koraiensis nakai extract or fraction thereof as effective ingredient for prevention or treatment of parkinson's disease WO2019212300A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2018-0052009 2018-05-04
KR1020180052009A KR102072471B1 (en) 2018-05-04 2018-05-04 Composition for preventing and/or treating a neurodegenerative disease comprising extract or fraction of Aster koraiensis Nakai

Publications (1)

Publication Number Publication Date
WO2019212300A1 true WO2019212300A1 (en) 2019-11-07

Family

ID=68386610

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2019/005352 WO2019212300A1 (en) 2018-05-04 2019-05-03 Composition comprising aster koraiensis nakai extract or fraction thereof as effective ingredient for prevention or treatment of parkinson's disease

Country Status (2)

Country Link
KR (1) KR102072471B1 (en)
WO (1) WO2019212300A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102234494B1 (en) * 2019-10-02 2021-03-31 대한민국 Composition comprising Aster ageratoides Turcz. extract for improving cognitive function
KR102335690B1 (en) * 2020-03-12 2021-12-07 한국과학기술연구원 Novel triterpene saponin derivative and use thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20050007955A (en) * 2003-07-12 2005-01-21 충남대학교산학협력단 An Extract Having an Antioxidative Activity in Brain Tissue
KR20120067183A (en) * 2010-12-15 2012-06-25 상지대학교산학협력단 Anticonvulsive compositions comprising aster glehni extract, fractions thereof, or compounds isolated from thereform
KR20120095251A (en) * 2011-02-18 2012-08-28 한국과학기술연구원 Composition comprising the extracts and fractions of gymnaster koraiensis for prevention or treatment of retinal diseases

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20050007955A (en) * 2003-07-12 2005-01-21 충남대학교산학협력단 An Extract Having an Antioxidative Activity in Brain Tissue
KR20120067183A (en) * 2010-12-15 2012-06-25 상지대학교산학협력단 Anticonvulsive compositions comprising aster glehni extract, fractions thereof, or compounds isolated from thereform
KR20120095251A (en) * 2011-02-18 2012-08-28 한국과학기술연구원 Composition comprising the extracts and fractions of gymnaster koraiensis for prevention or treatment of retinal diseases

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
KIM, KYUNG-A ET AL.: "Edible wild vegetable, Gymnaster koraiensis protects retinal ganglion cells against oxidative stress", FOOD AND CHEMICAL TOXICOLOGY, vol. 49, 2011, pages 2131 - 2143, XP028266613 *
ZHANG, HONG-TAO ET AL.: "Effect of Aster tataricus on production of inflammatory mediators in LPS stimulated rat astrocytoma cell line (C6) and THP-1 cells", SAUDI PHARMACEUTICAL JOURNAL, vol. 25, 2017, pages 370 - 375, XP029945119 *

Also Published As

Publication number Publication date
KR102072471B1 (en) 2020-02-04
KR20190127410A (en) 2019-11-13

Similar Documents

Publication Publication Date Title
WO2018012834A1 (en) Akkermansia muciniphila strain having effect of preventing or treating degenerative brain diseases or metabolic diseases, and use thereof
WO2016060426A1 (en) Composition containing dolichos lablab l. extract as active ingredient for preventing or alleviating non-alcoholic fatty liver diseases
WO2019212300A1 (en) Composition comprising aster koraiensis nakai extract or fraction thereof as effective ingredient for prevention or treatment of parkinson&#39;s disease
WO2016032249A1 (en) Pharmaceutical composition containing vaccinium bracteatum thunb. extract or fraction thereof as active ingredient for preventing or treating neuroinflammation or neuro-degenerative diseases
WO2019088412A1 (en) Composition containing tea extract with increased specific ingredient content for prevention or alleviation of lipid metabolism disorders
WO2020040432A1 (en) Pharmaceutical composition for preventing or treating muscle diseases, containing ginseng berry extract as active ingredient
WO2013047958A1 (en) Composition for inhibiting liver function deterioration, containing citrus peel extract or narirutin as active ingredient, and method for extracting narirutin from citrus peel
WO2020218727A9 (en) Peptide isolated from protein hydrolysate of tenebrio molitor mealworm and composition comprising same as active ingredient for prevention or treatment of liver injury
WO2017082479A1 (en) Pharmaceutical composition for prevention or treatment of obesity comprising bean germinated embryo extract
WO2016093613A2 (en) Composition for preventing or treating abnormal weight loss, containing citrus unshiu peel extract
WO2019231198A1 (en) Composition comprising cimicifuga dahurica extract, fraction thereof, or cimicifuga dahurica-derived compound as effective ingredient for prevention or treatment of degenerative brain disease
WO2018008999A1 (en) Composition comprising aster koraiensis extract or fraction thereof as active ingredient for regulating blood glucose and preventing or treating diabetes
WO2018106009A1 (en) Composition containing maple tree leaf extract or fractions thereof for preventing or treating inflammatory ocular diseases
KR102379612B1 (en) Composition for preventing and treating osteoporosis comprising extracts of fermented tenebrio molitor larva
KR20150051160A (en) Composition for preventing or treating thyroid disorders comprising aloe extracts or fraction thereof
WO2011046411A2 (en) Chestnut extracts with neuro cellular protective activity and acetylcholinesterase inhibiting activity, and use thereof
WO2020204242A1 (en) Antimicrobial composition for inhibiting oral bacteria and orally disintegrating film
KR102282883B1 (en) Composition for preventing and/or treating a degenerative brain disease comprising as an active ingredient an extract of Cimicifuga dahurica, a fraction thereof, or a compound derived from Cimicifuga dahurica
WO2023121314A1 (en) Use of apios americana tuber extract for protection against alcoholic liver damage or alcoholic brain damage
JP6113325B2 (en) Antiallergic oral composition
KR20150051159A (en) Composition for preventing or treating thyroid disorders comprising phytolacca esculenta houttuyn extracts or fraction thereof
WO2020256243A1 (en) Composition for preventing or treating depression comprising mixed extract of dioscorea nipponica makino and prickly pear
WO2022270760A1 (en) Method for treating non-alcoholic steatohepatitis through co-administration of curcumin derivative and tgf-β receptor inhibitor
WO2023191249A1 (en) Composition for ameliorating obesity or lipid metabolism comprising asimina triloba extract
WO2022030719A1 (en) Composition for improving, treating, or preventing muscle disease, or improving muscle function, containing rose hip as acitve ingredient

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19796184

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: 19796184

Country of ref document: EP

Kind code of ref document: A1