WO2018008908A1 - Pharmaceutical composition for preventing or treating degenerative neuronal diseases, containing compound isolated from withania coagulans as active ingredient - Google Patents

Pharmaceutical composition for preventing or treating degenerative neuronal diseases, containing compound isolated from withania coagulans as active ingredient Download PDF

Info

Publication number
WO2018008908A1
WO2018008908A1 PCT/KR2017/006986 KR2017006986W WO2018008908A1 WO 2018008908 A1 WO2018008908 A1 WO 2018008908A1 KR 2017006986 W KR2017006986 W KR 2017006986W WO 2018008908 A1 WO2018008908 A1 WO 2018008908A1
Authority
WO
WIPO (PCT)
Prior art keywords
expression
coa
app
mice
disease
Prior art date
Application number
PCT/KR2017/006986
Other languages
French (fr)
Korean (ko)
Inventor
김명옥
Original Assignee
경상대학교산학협력단
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 경상대학교산학협력단 filed Critical 경상대학교산학협력단
Publication of WO2018008908A1 publication Critical patent/WO2018008908A1/en

Links

Images

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • A61K31/366Lactones having six-membered rings, e.g. delta-lactones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • 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)
    • A61K36/81Solanaceae (Potato family), e.g. tobacco, nightshade, tomato, belladonna, capsicum or jimsonweed

Definitions

  • the present invention relates to a pharmaceutical composition for the prevention or treatment of degenerative neurological diseases containing a compound isolated from Vitania coagulans as an active ingredient.
  • AD Alzheimer's disease
  • a ⁇ amyloid ⁇ petide
  • APP amyloid precursor protein
  • is the protein C-terminal fragment remaining after the N-terminal fragment of APP is cut by the protease enzyme beta secretase (beta-secretase) is again broken down aspartic acid protein called gamma secretase ( ⁇ -secretase)
  • beta secretase beta-secretase
  • gamma secretase gamma secretase
  • HSPs Heat-shock proteins
  • the HSP70 family is known to play an important role in neuroprotection, but its expression is suppressed in the course of AD and other aging-related diseases, preventing them from functioning properly.
  • Withanolides are natural compounds of the steroidal lactones family found in plants belonging to the family Solanaceae .
  • Vitalanides are extracted from the genus Withania , and the plants of this genus include Withania somnifera and Withania coagulans.
  • coagulans ) species is famous.
  • Vitania coagulans are native to Pakistan, Iran, Afghanistan, and India.
  • the name coagulans is derived from the fruit's agglomerate properties, which are used to make cheese. Plants, roots, berries, seeds, leaves and stems have been used as traditional folk medicine, and the fruit is known to have a calming and diuretic effect and an effect on asthma.
  • Coa-A coagulansin-A
  • Coagulus enhances the expression of HSP70 protein and inhibits the expression of BACE1 to inhibit the amyloid production pathway. Confirmed that the efficacy was completed the present invention.
  • One aspect of the present invention to provide a pharmaceutical composition for the prevention or treatment of degenerative neurological diseases containing a compound represented by the following formula (1) or a pharmaceutically acceptable salt thereof as an active ingredient.
  • Another aspect of the present invention to provide a health functional food for the prevention or improvement of degenerative neurological disease containing the compound represented by the formula (1) as an active ingredient.
  • One aspect of the present invention provides a pharmaceutical composition for the prevention or treatment of degenerative neurological diseases containing a compound represented by the following formula (1) or a pharmaceutically acceptable salt thereof as an active ingredient.
  • degenerative neurological disease refers to a disease causing degenerative changes in neurons of the central nervous system and causing various symptoms.
  • Representative degenerative neurological disorders include Alzheimer's disease, Parkinson's disease, Progressive supranuclear palsy, Multiple system strophy, Olive nucleus-People-cerebellar atrophy , Shy-Drager syndrome, Striatonigral degeneration, Huntington's disease, Amyotrophic lateral sclerosis (ALS), Essential tremor, Cortico-basal ganlionic degeneration, Diffuse Lewy body disease, Parkin-ALS-dementia complex of Guam, and Pick's disease. have.
  • Alzheimer's disease is the most common of the many conditions that cause dementia, with neuronal fibers twisted abnormally in elderly spots and nerve cells.
  • neuronal fibers twisted abnormally in elderly spots and nerve cells.
  • brain neurons that are important for maintaining memory and other intellectual abilities, and the amount of specific chemicals coming and going between these brain neurons is separated.
  • the neurodegenerative disease may be Alzheimer's disease or dementia induced by the accumulation of amyloid beta protein in brain cells, but the disease having the therapeutic and prophylactic effect of the compound of Formula 1 is not limited thereto. .
  • amyloid beta is a protein produced by proteolytic enzyme action from an amyloid precursor protein, and is known to cause brain cell death while being deposited in brain tissue.
  • amyloid precursor protein a protein produced by proteolytic enzyme action from an amyloid precursor protein
  • fibrous structures called 'senior spots' and 'nerve fibrous knots', in which A ⁇ is accumulated, are seen in the nerve cells, and nerve cells are killed by apoptosis. It is known.
  • the compound represented by Formula 1 may promote the expression of HSP70 in neurons, inhibit the expression of BACE1, and inhibit the inflammatory response of neurons.
  • composition of the present invention may further comprise a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable refers to a composition that is physiologically acceptable and that, when administered to a human, typically does not cause allergic or similar reactions, such as gastrointestinal disorders, dizziness, and the like.
  • Pharmaceutically acceptable carriers include, for example, carriers for oral administration such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid and the like, and parenteral administration such as water, suitable oils, saline, aqueous glucose and glycols, and the like.
  • the like may further comprise stabilizers and preservatives. Suitable stabilizers include antioxidants such as sodium hydrogen sulfite, sodium sulfite or ascorbic acid.
  • Suitable preservatives include benzalkonium chloride, methyl- or propyl-paraben and chlorobutanol.
  • Other pharmaceutically acceptable carriers may be referred to those described in the following documents (Remington's Pharmaceutical Sciences, 19th ed., Mack Publishing Company, Easton, PA, 1995).
  • the pharmaceutical composition according to the present invention may be formulated in a suitable form according to methods known in the art together with the pharmaceutically acceptable carrier as described above. That is, the pharmaceutical composition of the present invention may be prepared in various parenteral or oral administration forms according to known methods. As representative of formulations for parenteral administration, isotonic aqueous solutions or suspensions are preferred for injectable formulations.
  • Injectable formulations may be prepared according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • each component may be formulated for injection by dissolving in saline or buffer.
  • formulations for oral administration include, but are not limited to, powders, granules, tablets, pills and capsules.
  • compositions formulated in such a manner can be administered in a effective amount via several routes, including oral, transdermal, subcutaneous, intravenous or intramuscular.
  • an 'effective amount' refers to an amount that exhibits a prophylactic or therapeutic effect when administered to a patient.
  • the dosage of the pharmaceutical composition according to the present invention may be appropriately selected depending on the route of administration, subject to administration, age, gender weight, individual difference and disease state.
  • the pharmaceutical composition of the present invention can vary the content of the active ingredient according to the degree of disease, according to one embodiment can be administered in an effective amount of 1 to 100 mg / kg / day in the form of parenteral administration have.
  • Another aspect of the present invention provides a health functional food for the prevention or improvement of degenerative neurological disease containing the compound represented by the formula (1) as an active ingredient.
  • the health food of the present invention may be added as it is, or used with other food or food ingredients, and may be appropriately used according to a conventional method.
  • the kind of food there is no particular limitation on the kind of food.
  • the food include drinks, meat, sausages, breads, biscuits, rice cakes, chocolates, candy, snacks, confectionery, pizza, ramen, other noodles, gums, dairy products including ice cream, various soups, beverages, alcoholic beverages and vitamins. Combinations and the like, and include all of the health foods in the conventional sense.
  • the compound of formula 1 according to the present invention may be added as it is to food or used together with other food or food ingredients, and may be suitably used according to conventional methods.
  • the mixing amount of the active ingredient can be suitably determined according to the purpose of use (prevention or improvement).
  • the amount of the compound of formula 1 in the health food can be added to 0.01 to 15% by weight of the total food weight, the health beverage composition is based on 100 ml in a ratio of 0.02 to 5 g, preferably 0.3 to 1 g Can be added.
  • the amount may be below the above range, and the active ingredient may be used in an amount above the above range because there is no problem in terms of safety.
  • the health functional beverage composition of the present invention contains the compound of Chemical Formula 1 as an essential ingredient in the indicated ratio, and there are no particular limitations on other ingredients, and it may contain various flavors or natural carbohydrates as additional ingredients, such as ordinary drinks. Can be.
  • natural carbohydrates include monosaccharides such as glucose, fructose and the like; Disaccharides such as maltose, sucrose and the like; And conventional sugars such as polysaccharides such as dextrin, cyclodextrin, and sugar alcohols such as xylitol, sorbitol, and erythritol.
  • natural flavoring agents such as, tauumatin, stevia extract (for example, rebaudioside A, glycyrrhizin, etc.) and synthetic flavoring agents (saccharin, aspartame, etc.) can be advantageously used. .
  • the food of the present invention includes various nutrients, vitamins, minerals (electrolytes), flavors such as synthetic flavors and natural flavors, coloring and neutralizing agents (such as cheese and chocolate), pectic acid and salts thereof, alginic acid and its Salts, organic acids, protective colloidal thickeners, pH adjusters, stabilizers, preservatives, glycerin, alcohols, carbonation agents used in carbonated drinks, and the like.
  • these components may be used independently or in combination, and the proportion of such additives is not so critical but is generally selected from the range of 0 to about 20 parts by weight per 100 parts by weight of the compound of the present invention.
  • the compound represented by Formula 1 may promote the expression of HSP70 in neurons and inhibit the expression of BACE1.
  • the compound of Formula 1 of the present invention promotes the expression of HSP70 and has the effect of inhibiting the production and aggregation of A ⁇ by inhibiting the expression of BACE1, it may be used as an active ingredient of a pharmaceutical composition for preventing or treating neurodegenerative diseases. .
  • FIG. 1 is a diagram showing the survival rate of SH-SY5Y cells following coagulansin-A (Coa-A) and amyloid beta oligomer (A ⁇ O) treatment.
  • Figure 2 is a diagram confirming the change in the expression of proteins following Coa-A and A ⁇ O treatment.
  • Figure 3 is a diagram confirming the change in the expression of HSP70 protein by Coa-A and A ⁇ O treatment.
  • FIG. 4 is a diagram confirming the expression change of HSP70 and BACE1 protein according to Coa-A administration in APP / PS1 mice.
  • Figure 5 is a diagram confirming the expression changes of HSP70 protein according to Coa-A administration in the cortex and hippocampus of APP / PS1 mice.
  • FIG. 6 is a diagram confirming the action state of the amyloid production pathway according to Coa-A administration in APP / PS1 mice.
  • Figure 7 is a diagram confirming the action state of the non-amyloid production pathway following Coa-A administration in APP / PS1 mice.
  • FIG. 9 is a view showing the change in size and repeatability of A ⁇ spots according to Coa-A administration in the brain of APP / PS1 mice.
  • FIG. 10 is a diagram confirming the change in the size and repeatability of A ⁇ spots according to Coa-A administration in the brain of APP / PS1 mice.
  • FIG. 11 is a view showing the change in size and repeatability of A ⁇ spots according to Coa-A administration in the brain of APP / PS1 mice.
  • FIG. 12 is a diagram confirming the changes in HSP70 and A ⁇ according to Coa-A administration in the brain of APP / PS1 mice.
  • Figure 13 is a diagram confirming the activity of microglia according to Coa-A administration in the brain of APP / PS1 mice.
  • FIG. 14 is a diagram confirming the activity of astrocytes according to Coa-A administration in the brain of APP / PS1 mice.
  • Figure 15 is a diagram confirming the expression of inflammatory response related molecules according to Coa-A administration in the brain of APP / PS1 mice.
  • 16 is a diagram confirming the expression of the exhibited synaptic protein and postsynaptic protein in the brain of APP / PS1 mice.
  • 17 is a diagram confirming the expression of the exhibited synaptic protein and postsynaptic protein in the brain of APP / PS1 mice.
  • 19 is a diagram confirming the escape time during the training period before the Coa-A administration in Morris water maze experiment.
  • 21 is a view confirming the behavioral change rate according to the Coa -A administration in the Y-maze experiment.
  • Coagulantine A ( Coagulansin -A, Coa -A) and amyloid beta oligomers
  • Coa-A was provided by Professor Bushra Mirza of QUAD-I-Asam University (Islamabad, Pakistan) and the extraction method is disclosed in " J. Nat. Prod. 76, 22.28 (2013)”.
  • Amyloid beta oligomers dissolve A ⁇ peptides (Sigma, USA) in hexafluoro propanol (HFIP), and disperse after sonication and filtering- Stored at 80.
  • a ⁇ O Amyloid beta oligomers
  • SH-SY5Y cells a human neuroblastoma, contain high concentrations of D-glucose, 10% fetal bovine serum (FBS) and 1% antibiotics (penicillin-streptomycin). Dulbecco's modified Eagle medium) was incubated in humidified conditions containing 37 and 5% CO 2 . A ⁇ O was treated at a concentration of 0.5 ⁇ m and Coa-A at 20 ⁇ g / ml.
  • MTT experiments were performed using 3- [4, 5-dimethylthiazol-2-yl] -2 and 5-diphenyl tetrazolium bromide to measure cell viability.
  • SH-SY5Y cells were cultured in a 96-well plate with 200 ⁇ l DMEM medium at 1 ⁇ 10 5 / well concentration. After 48 hours, the cells were further incubated with A ⁇ O (0.5 ⁇ m) alone, Coa-A alone (10, 20 and 30 ⁇ g / ml), or medium containing A ⁇ 1-42 and Coa-A, respectively. After 24 hours, MTT solution dissolved in phosphate buffered saline (PBS) at a concentration of 5 ml / ml was added to each well, and further incubated at 37 to 4 hours. Thereafter, DMSO (dimethyl sulfoxide) was added to dissolve formazone and mixed for 10 to 20 minutes, and then absorbance (570 to 630 nm) was measured using ApoTox-Glo TM (Promega, USA).
  • a ⁇ O 0.5 ⁇ m
  • Coa-A 10, 20 and 30 ⁇ g / ml
  • MTT solution dissolved in phosphate
  • APP / PS1 double transgenic mice are transgenic animals in which a human mutant APP gene and a mutant PS1 (preselin) gene are inserted, and exhibit an neuropathological characteristic of AD due to the significant development of geriatric spots and amyloid beta accumulation.
  • mice Eight-week-old male APP / PS1 double transgenic mice were purchased from Jackson Laboratory (Main, USA), and C57BL / 6J wild-type mice were purchased from Samtako Korea (South Korea) at 25 ° C. Breeding in a 12 hour light cycle and random feeding conditions. Every effort has been made to minimize the sacrifice and suffering of the animals, and the experimental procedure was carried out with the approval of the Animal Ethics Committee of Gyeongsang National University.
  • Protein concentration was measured using a BioRad protein assay kit (BioRad Laboratories, Calif., USA), and then 20 ⁇ g of protein was collected using 4-12% Bolt TM mini gel (Novex, Life Technologies, Kiryat Simona, Israel). Electrophoresis was transferred to the PVDF membrane. The membrane was then blocked with 5% (w / v) scheme milk to reduce nonspecific binding and reacted with the primary antibody at 4 overnight. The membrane was then washed, reacted with secondary antibodies and protein bands were identified according to the manufacturer's instructions using ECL detection reagents (Amersham Pharmacia Biotech, Uppsala, Sweden). All x-ray films were scanned and the intensity of the bands was analyzed using Sigma Gel program, version 1.0, SPSS, Illinois, USA.
  • Mouse brain grinding soluble A ⁇ 1 -42 1 -40, and A ⁇ concentration in the solution (homogenate) using a Human A ⁇ 1 -42, and A ⁇ 1 -40 EILSA kits was determined according to the manufacturer's instructions.
  • ice-cold 4% paraformaldehyde was perfused to the mouse via perfusion, and the brain was post-fixed with 4% paraformaldehyde for 72 hours, followed by 72% in 20% sucrose. Stored for hours. Brains were frozen with O.C.T compound (A.O, USA) and coronal sections were cut to 14- ⁇ m thick using a CM 3050C cryostat (Leica, Germany). The cut coronal plane was attached by melting on a probe-on plus charged slide (Fisher, Illinois, USA).
  • Fluorescent staining was performed to confirm protein expression in brain tissues, and staining was performed in parallel for dual staining.
  • the slides were washed twice with 0.01 M PBS for 15 minutes each and reacted for 37 to 5 minutes by the addition of proteinase-k solution. After reacting with a blocking solution consisting of 5% normal goat serum, 0.3% Triton X-100 and PBS for 1 hour at room temperature, the primary antibody diluted 1: 100 was reacted overnight. The next day TRITC- or FICT-bound secondary antibody (Santa Cruz) was diluted 1:50 in PBS and reacted for 90 minutes, and then the slides were mounted with Prolong Antifade Reagent (Molecular Probe, Eugene, USA) for confocal laser- Observation was made with a scanning microscope (Flouview FV 1000, Olympus, Japan).
  • Thioflavin S staining was performed to identify amyloid plaques in the brain.
  • the slides were washed twice with 0.01 M PBS for 10 minutes, immersed in 1% thioflavin S solution and left at room temperature for 20 minutes. After immersing in 70% ethanol for 5 minutes, washed twice with water, counterstained with propidium iodide (PI) and then covered with a cover slip.
  • PI propidium iodide
  • the density of the protein bands is expressed as mean ⁇ standard error mean (SEM) values in arbitrary units.
  • Statistical analysis was performed by one-way analysis of variance (ANOVA) in all experiments, and further Student's t-test using Prism 5 (graph Pad Software, California, USA).
  • the results of morphology analysis and amyloid spots were analyzed using Image J software (open source software provided by the National Institutes of Health, USA) and expressed as Integral Optical Densities (IODs). The results were considered significant when p ⁇ 0.05, 0.01 and 0.001.
  • MTT assay was performed to confirm cell viability change by Coa-A treatment.
  • the experimental group treated with A ⁇ O significantly reduced the survival rate of SH-SY5Y cells compared to the control group, and the experimental group treated with Coa-A showed cells at all three concentrations (10, 20 and 30 ⁇ g / ml). It was confirmed that no toxicity appeared.
  • the experimental group treated with A ⁇ O and Coa-A was confirmed that the cell survival rate was significantly increased compared to the experimental group treated with only A ⁇ O (FIG. 1).
  • HSP70 and BACE1 proteins were confirmed by A ⁇ O (0.5 ⁇ m) treatment in SH-SY5Y cells.
  • HSP70 was increased in the hippocampus and cortex of APP / PS1 mice administered with Coa-A and decreased in APP / PS1 mice administered with vehicle through immunofluorescence staining (FIG. 5). ).
  • HSP70 protein is known to inhibit protein aggregation and protect neurons. Therefore, the effect of Coa-A on the production and aggregation of A ⁇ in APP / PS1 mice was investigated.
  • Coa-A administration increased expression of ⁇ -secretase (s-APP ⁇ ) genes such as A disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) and ADAM17, resulting in a non-amyloidogenic pathway. ) was confirmed to act (Fig. 7).
  • s-APP ⁇ ⁇ -secretase genes
  • Activated glial cells including microglia and astrocytes, and associated neuroinflammation are one of the important features of the AD brain. Therefore, the effect of Coa-A administration on the activity and neuroinflammatory glial cells was confirmed.
  • microglia marker Iba1 ionized calcium-binding adapter molecule 1
  • astrocyte markers glial fibrillary acidic protein (GFAP) was confirmed by immunofluorescence staining. 'S brain has a significant number of activated glial cells. On the other hand, it was confirmed that glial cell activity was inhibited in the brain of APP / PS1 mice administered with Coa-A (FIGS. 13 and 14).
  • the escape time was measured.
  • the escape time of Coa-A-administered APP / PS1 mice was significantly reduced compared to the vehicle-administered APP / PS1 mice. It was confirmed that the learning ability was improved.
  • the evacuation time of the APP / PS1 mice administered with Coa-A was significantly lower than that of the APP / PS1 mice administered with the vehicle (FIG. 19).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Natural Medicines & Medicinal Plants (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Mycology (AREA)
  • Alternative & Traditional Medicine (AREA)
  • Microbiology (AREA)
  • Medical Informatics (AREA)
  • Botany (AREA)
  • Biotechnology (AREA)
  • Nutrition Science (AREA)
  • Food Science & Technology (AREA)
  • Polymers & Plastics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to a pharmaceutical composition for preventing or treating degenerative neuronal diseases, containing Coagulansin A as an active ingredient. Coagulansin-A has effects of promoting the expression of HSP70 and inhibiting the expression of BACE1 so as to inhibit the formation and aggregation of Aβ, thereby being usable as an active ingredient of a pharmaceutical composition for preventing or treating degenerative neuronal diseases.

Description

비타니아 코아귤란스에서 분리한 화합물을 유효성분으로 함유하는 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물Pharmaceutical composition for the prevention or treatment of degenerative neurological diseases, containing as an active ingredient a compound isolated from vitania coagulans
본 발명은 비타니아 코아귤란스에서 분리한 화합물을 유효성분으로 함유하는 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물에 관한 것이다.The present invention relates to a pharmaceutical composition for the prevention or treatment of degenerative neurological diseases containing a compound isolated from Vitania coagulans as an active ingredient.
알츠하이머병 (Alzheimer's disease; 이하 AD로 표기함)은 나이가 들어감에 따라 뇌에서 선택적으로 신경세포의 사멸이 일어나는 퇴행성 신경질환이다. AD 환자의 뇌에서 나타나는 조직병리학적 특징으로는 뇌 조직 내에서 세포 외부에 침착되는 노인 반점(senile plaque) 및 신경세포 내부에 실타래처럼 뭉쳐서 축적되는 신경섬유 덩어리(neurofibrillary tangle; NFT)가 있다. 노인 반점의 주요 구성성분으로는 아밀로이드 전구단백질(amyloid precursor protein; 이하 APP로 표기함)에서 단백질 분해효소들에 의해 잘려져 나온 아밀로이드 베타(amyloid β petide; 이하 Aβ로 표기함)가 있으며, 최근의 여러 가지 보고들에 의하여 Aβ가 AD의 여러 가지 병리학적 증상들을 일으키는 주요 병인으로 생각되고 있다.Alzheimer's disease (hereinafter referred to as AD) is a degenerative neurological disease in which the brain selectively kills nerve cells as it ages. Histopathological features in the brain of AD patients include senile plaques that are deposited outside the cells in brain tissue and neurofibrillary tangles (NFTs) that accumulate like threads in nerve cells. The main components of the senile plaques are amyloid β petide (hereinafter referred to as Aβ), which is cleaved by proteolytic enzymes from amyloid precursor protein (hereinafter referred to as APP). Branch reports suggest that Αβ is the major cause of various pathological symptoms of AD.
Aβ는 APP의 N-말단 단편이 베타 세크리테아제(β-secretase)라는 단백질 분해효소에 의하여 잘려진 후에 남아있는 APP C-말단 단편이 다시 감마 세크리테아제(γ-secretase)라는 아스파트르산 단백질 분해효소에 의해 막을 관통하는 부분이 잘려서 만들어진다. 40개에서 42개 또는 43개의 아미노산을 가지는 형태로 나타나며, 이 중 Aβ42가 노인 반점의 침착에 있어 중요하게 작용하는 것으로 알려져 있다.Αβ is the protein C-terminal fragment remaining after the N-terminal fragment of APP is cut by the protease enzyme beta secretase (beta-secretase) is again broken down aspartic acid protein called gamma secretase (γ-secretase) The part that penetrates the membrane is cut by enzymes. It appears in the form of 40 to 42 or 43 amino acids, of which Aβ 42 is known to play an important role in the deposition of age spots.
열-충격 단백질(heat-shock proteins; 이하 HSP로 표기함)은 단백질이 입체적 구조를 가지도록 돕는 샤페론(chaperone)의 하나로, 스트레스 및 비정상적인 환경에서 단백질의 응집 또는 잘못 접힘(misfolding)을 방지하는 역할을 한다. 샤페론 분자 중에서 HSP70 패밀리는 신경보호에 있어 중요한 역할을 하는 것으로 알려져 있으나, AD 및 다른 노화 관련 질병이 진행되는 경우 발현이 억제되기 때문에 적절한 기능을 수행하지 못하게 된다.Heat-shock proteins (hereinafter referred to as HSPs) are chaperones that help the proteins have a three-dimensional structure, which prevents aggregation or misfolding of proteins in stress and abnormal environments. Do it. Among the chaperone molecules, the HSP70 family is known to play an important role in neuroprotection, but its expression is suppressed in the course of AD and other aging-related diseases, preventing them from functioning properly.
비타놀리드(Withanolides)는 가지과(Solanaceae)에 속하는 식물에서 발견되는 스테로이드 락톤(steroidal lactones) 계열의 천연 화합물이다. 비타놀리드는 비타니아(Withania) 속에서 추출되며, 이 속의 식물로는 비타니아 솜니페라(Withania somnifera) 및 비타니아 코아귤란스(Withania coagulans) 종이 유명하다. 비타니아 코아귤란스는 파키스탄, 이란, 아프가니스탄 및 인도에 주로 자생하고 있으며, 코아귤란스라는 이름은 이 식물의 열매가 우유를 응집시키는 특성이 있어 치즈 제조에 이용한 것에서 유래하였다. 식물 전체, 뿌리, 열매, 씨앗, 잎 및 줄기 등이 전통 민간 의약으로 사용되어 왔으며, 열매는 진정 효과 및 이뇨 효과가 있고, 천식에 효과가 있는 것으로 알려져 있다. Withanolides are natural compounds of the steroidal lactones family found in plants belonging to the family Solanaceae . Vitalanides are extracted from the genus Withania , and the plants of this genus include Withania somnifera and Withania coagulans. coagulans ) species is famous. Vitania coagulans are native to Pakistan, Iran, Afghanistan, and India. The name coagulans is derived from the fruit's agglomerate properties, which are used to make cheese. Plants, roots, berries, seeds, leaves and stems have been used as traditional folk medicine, and the fruit is known to have a calming and diuretic effect and an effect on asthma.
본 발명자들은 비타니아 코아귤란스에서 추출한 코아귤란신-A(Coagulansin-A; 이하 Coa-A로 표기함) 화합물이 HSP70 단백질의 발현을 상승시키고, BACE1의 발현을 억제하여 아밀로이드 생성 경로를 억제하는 효능이 있음을 확인하여 본 발명을 완성하였다.The present inventors found that the coagulansin-A (Coagulansin-A; hereinafter referred to as Coa-A) compound extracted from Vit. Coagulus enhances the expression of HSP70 protein and inhibits the expression of BACE1 to inhibit the amyloid production pathway. Confirmed that the efficacy was completed the present invention.
본 발명의 일 양상은 하기 화학식 1로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물을 제공하는 것이다.One aspect of the present invention to provide a pharmaceutical composition for the prevention or treatment of degenerative neurological diseases containing a compound represented by the following formula (1) or a pharmaceutically acceptable salt thereof as an active ingredient.
Figure PCTKR2017006986-appb-C000001
Figure PCTKR2017006986-appb-C000001
본 발명의 다른 양상은 상기 화학식 1로 표시되는 화합물을 유효성분으로 함유하는 퇴행성 신경질환의 예방 또는 개선용 건강기능식품을 제공하는 것이다.Another aspect of the present invention to provide a health functional food for the prevention or improvement of degenerative neurological disease containing the compound represented by the formula (1) as an active ingredient.
본 발명의 일 양상은 하기 화학식 1로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물을 제공한다.One aspect of the present invention provides a pharmaceutical composition for the prevention or treatment of degenerative neurological diseases containing a compound represented by the following formula (1) or a pharmaceutically acceptable salt thereof as an active ingredient.
[화학식 1][Formula 1]
Figure PCTKR2017006986-appb-I000001
Figure PCTKR2017006986-appb-I000001
본 발명의 용어, "퇴행성 신경질환"이란 중추신경계의 신경세포에 퇴행성 변화가 나타나면서 여러 가지 증상을 유발하는 질환을 의미하는 것을 의미한다. 대표적인 퇴행성 신경질환에는 알츠하이머병(Alzheimer's disease), 파킨슨병(Parkinson's disease), 진행성 핵상마비(Progressive supranuclear palsy), 다계통 위축증(Multiple system strophy), 감람핵-뇌교-소뇌 위축증(Olivopontocerebellar atrophy: OPCA), 샤이-드래거 증후군(Shy-Drager syndrome), 선조체-흑질 퇴행증(Striatonigral degeneration), 헌팅톤병(Huntington's disease), 근위축성 측색 경화증(Amyotrophic lateral sclerosis;ALS), 본태성 진전증(Essential tremor), 피질-기저핵 퇴행증(Cortico-basal ganlionic degeneration), 미만성 루이 소체 질환(Diffuse Lewy body disease), 파킨스-ALS-치매 복합증(Parkinson-ALS-dementia complex of Guam) 및 픽병(Pick's disease)등이 있다.As used herein, the term "degenerative neurological disease" refers to a disease causing degenerative changes in neurons of the central nervous system and causing various symptoms. Representative degenerative neurological disorders include Alzheimer's disease, Parkinson's disease, Progressive supranuclear palsy, Multiple system strophy, Olive nucleus-People-cerebellar atrophy , Shy-Drager syndrome, Striatonigral degeneration, Huntington's disease, Amyotrophic lateral sclerosis (ALS), Essential tremor, Cortico-basal ganlionic degeneration, Diffuse Lewy body disease, Parkin-ALS-dementia complex of Guam, and Pick's disease. have.
알츠하이머병은 치매를 일으키는 많은 질환들 중에 가장 흔한 것으로, 노인 반점과 신경세포 안에서 신경원 섬유들이 비정상적으로 꼬여 있는 모습을 보인다. 이외에도 기억 및 다른 지적 능력을 유지하는데 중요한 뇌신경세포가 많이 없어지고, 이러한 뇌신경세포 사이에서 오가는 특정 화학물질의 양이 많이 떨어져 있는 모습을 보인다.Alzheimer's disease is the most common of the many conditions that cause dementia, with neuronal fibers twisted abnormally in elderly spots and nerve cells. In addition, there are many brain neurons that are important for maintaining memory and other intellectual abilities, and the amount of specific chemicals coming and going between these brain neurons is separated.
일 구체예에 따르면 상기 퇴행성 신경질환은 뇌세포 내에 아밀로이드 베타 단백질이 축적되어 유도되는 알츠하이머병 또는 치매일 수 있으나, 이에 본 발명의 화학식 1의 화합물의 치료 및 예방 효과가 있는 질병이 제한되는 것은 아니다.According to one embodiment, the neurodegenerative disease may be Alzheimer's disease or dementia induced by the accumulation of amyloid beta protein in brain cells, but the disease having the therapeutic and prophylactic effect of the compound of Formula 1 is not limited thereto. .
본 발명의 용어, "아밀로이드 베타"는 아밀로이드 전구 단백질로부터 단백질 분해효소 작용에 의해 생성된 단백질로, 뇌 조직에 침착되면서 뇌세포의 사멸을 일으키는 것으로 알려져 있다. 대뇌의 신경세포가 변성하여 대량으로 사멸하는 경우 Aβ가 축적된 '노인 반점', '신경 원섬유 매듭'이라고 하는 섬유 모양의 구조가 신경 세포 내에서 보이며, 신경 세포는 세포사멸 기작에 의해 사멸하는 것으로 알려져 있다.As used herein, the term "amyloid beta" is a protein produced by proteolytic enzyme action from an amyloid precursor protein, and is known to cause brain cell death while being deposited in brain tissue. When neurons in the cerebrum degenerate and die in large quantities, fibrous structures called 'senior spots' and 'nerve fibrous knots', in which Aβ is accumulated, are seen in the nerve cells, and nerve cells are killed by apoptosis. It is known.
일 구체예에 따르면 상기 화학식 1로 표시되는 화합물은 신경세포에서 HSP70의 발현을 촉진하고, BACE1의 발현을 억제할 수 있으며, 신경세포의 염증반응을 억제할 수 있다.According to one embodiment, the compound represented by Formula 1 may promote the expression of HSP70 in neurons, inhibit the expression of BACE1, and inhibit the inflammatory response of neurons.
본 발명의 조성물은 약학적으로 허용되는 담체를 추가로 포함할 수 있다. 상기에서 "약학적으로 허용되는"이란 생리학적으로 허용되고 인간에게 투여될 때, 통상적으로 위장 장애, 현기증 등과 같은 알레르기 반응 또는 이와 유사한 반응을 일으키지 않는 조성물을 말한다. 약학적으로 허용되는 담체로는 예를 들면, 락토스, 전분, 셀룰로스 유도체, 마그네슘 스테아레이트, 스테아르산 등과 같은 경구 투여용 담체 및 물, 적합한 오일, 식염수, 수성 글루코스 및 글리콜 등과 같은 비경구 투여용 담체 등이 있으며 안정화제 및 보존제를 추가로 포함할 수 있다. 적합한 안정화제로는 아황산수소나트륨, 아황산나트륨 또는 아스코르 브산과 같은 항산화제가있다. 적합한 보존제로는 벤즈알코늄 클로라이드, 메틸- 또는 프로필-파라벤 및 클로로부탄올이 있다. 그 밖의 약학적으로 허용되는 담체로는 다음의 문헌에 기재되어 있는 것을 참고로 할 수 있다(Remington's Pharmaceutical Sciences, 19th ed., Mack Publishing Company, Easton, PA, 1995). 상기 본 발명에 따른 약학적 조성물은 상술한 바와 같은 약학적으로 허용되는 담체와 함께 당업계에 공지된 방법에 따라 적합한 형태로 제형화 될 수 있다. 즉, 본 발명의 약학적 조성물은 공지의 방법에 따라 다양한 비경구 또는 경구 투여용 형태로 제조될 수 있다. 비경구 투여용 제형의 대표적인 것으로는 주사용 제형으로 등장성 수용액 또는 현탁액이 바람직하다. 주사용 제형은 적합한 분산제 또는 습윤제 및 현탁화제를 사용하여 당업계에 공지된 기술에 따라 제조할 수 있다. 예를 들면, 각 성분을 식염수 또는 완충액에 용해시켜 주사용으로 제형화될 수 있다. 또한, 경구 투여용 제형으로는, 이에 한정되지는 않으나, 분말, 과립, 정제, 환약 및 캡슐 등이 있다.The composition of the present invention may further comprise a pharmaceutically acceptable carrier. As used herein, "pharmaceutically acceptable" refers to a composition that is physiologically acceptable and that, when administered to a human, typically does not cause allergic or similar reactions, such as gastrointestinal disorders, dizziness, and the like. Pharmaceutically acceptable carriers include, for example, carriers for oral administration such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid and the like, and parenteral administration such as water, suitable oils, saline, aqueous glucose and glycols, and the like. And the like may further comprise stabilizers and preservatives. Suitable stabilizers include antioxidants such as sodium hydrogen sulfite, sodium sulfite or ascorbic acid. Suitable preservatives include benzalkonium chloride, methyl- or propyl-paraben and chlorobutanol. Other pharmaceutically acceptable carriers may be referred to those described in the following documents (Remington's Pharmaceutical Sciences, 19th ed., Mack Publishing Company, Easton, PA, 1995). The pharmaceutical composition according to the present invention may be formulated in a suitable form according to methods known in the art together with the pharmaceutically acceptable carrier as described above. That is, the pharmaceutical composition of the present invention may be prepared in various parenteral or oral administration forms according to known methods. As representative of formulations for parenteral administration, isotonic aqueous solutions or suspensions are preferred for injectable formulations. Injectable formulations may be prepared according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. For example, each component may be formulated for injection by dissolving in saline or buffer. In addition, formulations for oral administration include, but are not limited to, powders, granules, tablets, pills and capsules.
상기와 같은 방법으로 제형화된 약학적 조성물은 유효량으로 경구, 경피, 피하, 정맥 또는 근육을 포함한 여러 경로를 통해 투여될 수 있다. 상기에서 '유효량'이란 환자에게 투여하였을 때, 예방 또는 치료 효과를 나타내는 양을 말한다. 본 발명에 따른 약학적 조성물의 투여량은 투여 경로, 투여 대상, 연령, 성별 체중, 개인차 및 질병 상태에 따라 적절히 선택할 수 있다. 바람직하게 본 발명의 약학적 조성물은 질환의 정도에 따라 유효성분의 함량을 달리할 수 있으며, 일 구체예에 따르면 비경구 투여의 형태일 때 1 내지 100 mg/kg/day의 유효량으로 투여할 수 있다.Pharmaceutical compositions formulated in such a manner can be administered in a effective amount via several routes, including oral, transdermal, subcutaneous, intravenous or intramuscular. As used herein, an 'effective amount' refers to an amount that exhibits a prophylactic or therapeutic effect when administered to a patient. The dosage of the pharmaceutical composition according to the present invention may be appropriately selected depending on the route of administration, subject to administration, age, gender weight, individual difference and disease state. Preferably the pharmaceutical composition of the present invention can vary the content of the active ingredient according to the degree of disease, according to one embodiment can be administered in an effective amount of 1 to 100 mg / kg / day in the form of parenteral administration have.
본 발명의 다른 양상은 화학식 1로 표시되는 화합물을 유효성분으로 함유하는 퇴행성 신경질환의 예방 또는 개선용 건강기능식품을 제공한다.Another aspect of the present invention provides a health functional food for the prevention or improvement of degenerative neurological disease containing the compound represented by the formula (1) as an active ingredient.
[화학식 1][Formula 1]
Figure PCTKR2017006986-appb-I000002
Figure PCTKR2017006986-appb-I000002
본 발명의 건강식품은 상기 화학식 1의 화합물을 그대로 첨가하거나 다른 식품 또는 식품 성분과 함께 사용될 수 있고, 통상적인 방법에 따라 적절하게 사용될 수 있다.The health food of the present invention may be added as it is, or used with other food or food ingredients, and may be appropriately used according to a conventional method.
상기 식품의 종류에는 특별한 제한은 없다. 상기 식품의 예로는 드링크제, 육류, 소세지, 빵, 비스켓, 떡, 쵸코렛, 캔디류, 스넥류, 과자류, 피자, 라면, 기타 면류, 껌류, 아이스크림류를 포함한 낙농제품, 각종 스프, 음료, 알콜 음료 및 비타민 복합제 등이 있으며, 통상적인 의미에서의 건강식품을 모두 포함한다.There is no particular limitation on the kind of food. Examples of the food include drinks, meat, sausages, breads, biscuits, rice cakes, chocolates, candy, snacks, confectionery, pizza, ramen, other noodles, gums, dairy products including ice cream, various soups, beverages, alcoholic beverages and vitamins. Combinations and the like, and include all of the health foods in the conventional sense.
본 발명에 따른 화학식 1의 화합물은 식품에 그대로 첨가하거나 다른 식품 또는 식품 성분과 함께 사용될 수 있고, 통상적인 방법에 따라 적절하게 사용될 수 있다. 유효 성분의 혼합량은 그의 사용 목적(예방 또는 개선용)에 따라 적합하게 결정될 수 있다. 일반적으로, 건강식품 중의 상기 화학식 1의 화합물의 양은 전체 식품 중량의 0.01 내지 15 중량으로 가할 수 있으며, 건강 음료 조성물은 100 ㎖를 기준으로 0.02 내지 5 g, 바람직하게는 0.3 내지 1 g의 비율로 가할 수 있다. 그러나 건강 및 위생을 목적으로 하거나 또는 건강 조절을 목적으로 하는 장기간 섭취의 경우에는 상기 양은 상기 범위 이하일 수 있으며, 안전성 면에서 아무런 문제가 없기 때문에 유효성분은 상기 범위 이상의 양으로도 사용될 수 있다.The compound of formula 1 according to the present invention may be added as it is to food or used together with other food or food ingredients, and may be suitably used according to conventional methods. The mixing amount of the active ingredient can be suitably determined according to the purpose of use (prevention or improvement). In general, the amount of the compound of formula 1 in the health food can be added to 0.01 to 15% by weight of the total food weight, the health beverage composition is based on 100 ml in a ratio of 0.02 to 5 g, preferably 0.3 to 1 g Can be added. However, in the case of long-term intake for health and hygiene or health control purposes, the amount may be below the above range, and the active ingredient may be used in an amount above the above range because there is no problem in terms of safety.
본 발명의 건강 기능성 음료 조성물은 지시된 비율로 필수 성분으로서 상기 화학식 1의 화합물을 함유하는 외에 다른 성분에는 특별한 제한이 없으며 통상의 음료와 같이 여러 가지 향미제 또는 천연 탄수화물 등을 추가 성분으로 함유할 수 있다. 상술한 천연 탄수화물의 예는 모노사카라이드, 예를 들어, 포도당, 과당 등; 디사카라이드, 예를 들어 말토스, 슈크로스 등; 및 폴리사카라이드, 예를 들어 덱스트린, 시클로덱스트린 등과 같은 통상적인 당, 및 자일리톨, 소르비톨, 에리트리톨 등의 당알콜이다. 상술한 것 이외의 향미제로서 천연 향미제(타우마틴, 스테비아 추출물(예를 들어 레바우디오시드 A, 글리시르히진등) 및 합성 향미제(사카린, 아스파르탐 등)를 유리하게 사용할 수 있다.The health functional beverage composition of the present invention contains the compound of Chemical Formula 1 as an essential ingredient in the indicated ratio, and there are no particular limitations on other ingredients, and it may contain various flavors or natural carbohydrates as additional ingredients, such as ordinary drinks. Can be. Examples of the above-mentioned natural carbohydrates include monosaccharides such as glucose, fructose and the like; Disaccharides such as maltose, sucrose and the like; And conventional sugars such as polysaccharides such as dextrin, cyclodextrin, and sugar alcohols such as xylitol, sorbitol, and erythritol. As flavoring agents other than those mentioned above, natural flavoring agents (tauumatin, stevia extract (for example, rebaudioside A, glycyrrhizin, etc.) and synthetic flavoring agents (saccharin, aspartame, etc.) can be advantageously used. .
상기 외에 본 발명의 식품은 여러 가지 영양제, 비타민, 광물(전해질), 합성 풍미제 및 천연 풍미제 등의 풍미제, 착색제 및 중진제(치즈, 초콜릿 등), 펙트산 및 그의 염, 알긴산 및 그의 염, 유기산, 보호성 콜로이드 증점제, pH 조절제, 안정화제, 방부제, 글리세린, 알코올, 탄산음료에 사용되는 탄산화제 등을 함유할 수 있다. 이러한 성분은 독립적으로 또는 조합하여 사용할 수 있으며, 이러한 첨가제의 비율은 그렇게 중요하진 않지만 본 발명의 화합물 100 중량부 당 0 내지 약 20 중량부의 범위에서 선택되는 것이 일반적이다.In addition to the above, the food of the present invention includes various nutrients, vitamins, minerals (electrolytes), flavors such as synthetic flavors and natural flavors, coloring and neutralizing agents (such as cheese and chocolate), pectic acid and salts thereof, alginic acid and its Salts, organic acids, protective colloidal thickeners, pH adjusters, stabilizers, preservatives, glycerin, alcohols, carbonation agents used in carbonated drinks, and the like. These components may be used independently or in combination, and the proportion of such additives is not so critical but is generally selected from the range of 0 to about 20 parts by weight per 100 parts by weight of the compound of the present invention.
일 구체예에 따르면 상기 화학식 1로 표시되는 화합물은 신경세포에서 HSP70의 발현을 촉진하고, BACE1의 발현을 억제할 수 있다.According to one embodiment, the compound represented by Formula 1 may promote the expression of HSP70 in neurons and inhibit the expression of BACE1.
본 발명의 화학식 1의 화합물은 HSP70의 발현을 촉진하고, BACE1의 발현을 억제하여 Aβ의 생성 및 응집을 억제하는 효능이 있으므로 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물의 유효성분으로 사용될 수 있다.Since the compound of Formula 1 of the present invention promotes the expression of HSP70 and has the effect of inhibiting the production and aggregation of Aβ by inhibiting the expression of BACE1, it may be used as an active ingredient of a pharmaceutical composition for preventing or treating neurodegenerative diseases. .
도 1은 코아귤란신 A(Coagulansin-A, Coa-A) 및 아밀로이드 베타 올리고머(AβO) 처리에 따른 SH-SY5Y 세포의 생존율을 나타낸 도이다.1 is a diagram showing the survival rate of SH-SY5Y cells following coagulansin-A (Coa-A) and amyloid beta oligomer (AβO) treatment.
도 2는 Coa-A 및 AβO 처리에 따른 단백질의 발현 변화를 확인한 도이다.Figure 2 is a diagram confirming the change in the expression of proteins following Coa-A and AβO treatment.
도 3은 Coa-A 및 AβO 처리에 따른 HSP70 단백질의 발현 변화를 확인한 도이다.Figure 3 is a diagram confirming the change in the expression of HSP70 protein by Coa-A and AβO treatment.
도 4는 APP/PS1 마우스에서 Coa-A 투여에 따른 HSP70 및 BACE1 단백질의 발현 변화를 확인한 도이다.4 is a diagram confirming the expression change of HSP70 and BACE1 protein according to Coa-A administration in APP / PS1 mice.
도 5는 APP/PS1 마우스의 피질 및 해마에서 Coa-A 투여에 따른 HSP70 단백질의 발현 변화를 확인한 도이다.Figure 5 is a diagram confirming the expression changes of HSP70 protein according to Coa-A administration in the cortex and hippocampus of APP / PS1 mice.
도 6은 APP/PS1 마우스에서 Coa-A 투여에 따른 아밀로이드 생성 경로의 작용 상태를 확인한 도이다.6 is a diagram confirming the action state of the amyloid production pathway according to Coa-A administration in APP / PS1 mice.
도 7은 APP/PS1 마우스에서 Coa-A 투여에 따른 비-아밀로이드 생성 경로의 작용 상태를 확인한 도이다.Figure 7 is a diagram confirming the action state of the non-amyloid production pathway following Coa-A administration in APP / PS1 mice.
도 8은 APP/PS1 마우스에서 Coa-A 투여에 따른 수용성 Aβ1 -40 및 Aβ1 -42의 수준을 확인한 도이다.8 is a check for the level of soluble Aβ, and Aβ 1 -40 1 -42 according to the Coa-A therapy in APP / PS1 mice.
도 9는 APP/PS1 마우스의 뇌에서 Coa-A 투여에 따른 Aβ 반점의 크기 및 반복 정도 변화를 확인한 도이다. 9 is a view showing the change in size and repeatability of Aβ spots according to Coa-A administration in the brain of APP / PS1 mice.
도 10은 APP/PS1 마우스의 뇌에서 Coa-A 투여에 따른 Aβ 반점의 크기 및 반복 정도 변화를 확인한 도이다. 10 is a diagram confirming the change in the size and repeatability of Aβ spots according to Coa-A administration in the brain of APP / PS1 mice.
도 11은 APP/PS1 마우스의 뇌에서 Coa-A 투여에 따른 Aβ 반점의 크기 및 반복 정도 변화를 확인한 도이다. 11 is a view showing the change in size and repeatability of Aβ spots according to Coa-A administration in the brain of APP / PS1 mice.
도 12는 APP/PS1 마우스의 뇌에서 Coa-A 투여에 따른 HSP70 및 Aβ 변화를 확인한 도이다.12 is a diagram confirming the changes in HSP70 and Aβ according to Coa-A administration in the brain of APP / PS1 mice.
도 13은 APP/PS1 마우스의 뇌에서 Coa-A 투여에 따른 미세아교세포의 활성 여부를 확인한 도이다.Figure 13 is a diagram confirming the activity of microglia according to Coa-A administration in the brain of APP / PS1 mice.
도 14는 APP/PS1 마우스의 뇌에서 Coa-A 투여에 따른 성상교세포의 활성 여부를 확인한 도이다.14 is a diagram confirming the activity of astrocytes according to Coa-A administration in the brain of APP / PS1 mice.
도 15는 APP/PS1 마우스의 뇌에서 Coa-A 투여에 따른 염증 반응 관련 분자들의 발현을 확인한 도이다.Figure 15 is a diagram confirming the expression of inflammatory response related molecules according to Coa-A administration in the brain of APP / PS1 mice.
도 16은 APP/PS1 마우스의 뇌에서 전시냅스 단백질 및 후시냅스 단백질의 발현을 확인한 도이다.16 is a diagram confirming the expression of the exhibited synaptic protein and postsynaptic protein in the brain of APP / PS1 mice.
도 17 APP/PS1 마우스의 뇌에서 전시냅스 단백질 및 후시냅스 단백질의 발현을 확인한 도이다.17 is a diagram confirming the expression of the exhibited synaptic protein and postsynaptic protein in the brain of APP / PS1 mice.
도 18은 모리스 수중 미로 실험에서 Coa-A 투여 전의 훈련 기간 동안 탈출 시간을 확인한 도이다.18 is a diagram confirming the escape time during the training period before the Coa-A administration in Morris water maze experiment.
도 19는 모리스 수중 미로 실험에서 Coa-A 투여 전의 훈련 기간 동안 탈출 시간을 확인한 도이다.19 is a diagram confirming the escape time during the training period before the Coa-A administration in Morris water maze experiment.
도 20은 모리스 수중 미로 훈련 종료 후 프로브 테스트 결과를 확인한 도이다.20 is a diagram confirming the probe test results after the completion of Morris underwater maze training.
도 21은 Y-미로 실험에서 Coa -A 투여에 따른 행동 변경율을 확인한 도이다.21 is a view confirming the behavioral change rate according to the Coa -A administration in the Y-maze experiment.
이하 하나 이상의 구체예를 실시예를 통하여 보다 상세하게 설명한다. 그러나, 이들 실시예는 하나 이상의 구체예를 예시적으로 설명하기 위한 것으로 본 발명의 범위가 이들 실시예에 한정되는 것은 아니다.Hereinafter, one or more embodiments will be described in more detail with reference to Examples. However, these examples are provided to illustrate one or more embodiments illustratively and the scope of the present invention is not limited to these examples.
실험방법Experiment method
1. One. 코아귤란신Coagulantine A( A ( CoagulansinCoagulansin -A, -A, CoaCoa -A) 및 아밀로이드 베타 올리고머-A) and amyloid beta oligomers
Coa-A는 퀘이드-아이-아잠 대학교(이슬라마바드, 파키스탄)의 Bushra Mirza 교수로부터 제공받았으며, 추출방법은 "J. Nat. Prod. 76, 22.28 (2013)"에 개시되어 있다.Coa-A was provided by Professor Bushra Mirza of QUAD-I-Asam University (Islamabad, Pakistan) and the extraction method is disclosed in " J. Nat. Prod. 76, 22.28 (2013)".
아밀로이드 베타 올리고머(Aβ oligomer, 이하 AβO로 표기함)는 Aβ 펩티드(Sigma, 미국)를 헥사플루오로 프로판올(hexafluoro propanol, HFIP)에 용해시키고, 초음파 분해(sonication) 및 필터레이션을 거친 후에 분주하여 -80에 보관하였다.Amyloid beta oligomers (denoted Aβ oligomers, hereinafter referred to as AβO) dissolve Aβ peptides (Sigma, USA) in hexafluoro propanol (HFIP), and disperse after sonication and filtering- Stored at 80.
2. 세포 실험2. Cell experiment
2-1. 2-1. SHSH -- SY5YSY5Y 세포 배양  Cell culture
인간 신경모세포종(neuroblastoma)인 SH-SY5Y 세포는 높은 농도의 D-글루코스, 10% 소태아혈청(fetal bovine serum, 이하 FBS로 표기함) 및 1% 항생제(penicillin-streptomycin)를 함유하고 있는 DMEM(Dulbecco's modified Eagle medium) 배지를 이용하여 37 및 5% CO2를 포함하는 가습 조건에서 배양하였다. AβO는 0.5 ㎛, Coa-A는 20 ㎍/㎖ 농도로 처리하였다.SH-SY5Y cells, a human neuroblastoma, contain high concentrations of D-glucose, 10% fetal bovine serum (FBS) and 1% antibiotics (penicillin-streptomycin). Dulbecco's modified Eagle medium) was incubated in humidified conditions containing 37 and 5% CO 2 . AβO was treated at a concentration of 0.5 µm and Coa-A at 20 µg / ml.
2-2. 2-2. MTTMTT assay assay
세포의 생존율을 측정하기 위하여 3-[4, 5-dimethylthiazol- 2-yl]-2, 5-diphenyl tetrazolium bromide를 이용하여 MTT 실험을 수행하였다.MTT experiments were performed using 3- [4, 5-dimethylthiazol-2-yl] -2 and 5-diphenyl tetrazolium bromide to measure cell viability.
구체적으로, SH-SY5Y 세포를 96-웰 플레이트에 200 ㎕ DMEM 배지와 함께1X105/웰 농도로 분주하여 배양하였다. 48시간 후에 AβO(0.5 ㎛) 단독, Coa-A 단독(10, 20 및 30 ㎍/㎖), 또는 Aβ1-42 및 Coa-A가 같이 포함되어 있는 배지로 각각 교체하여 추가로 배양하였다. 24시간 후에 5 ㎖/㎖ 농도로 인산완충용액(phosphate buffered saline, PBS)에 용해시킨 MTT 용액을 각 웰에 첨가하고, 37에서 4시간 동안 추가로 배양하였다. 이후, 포르마존을 용해시키기 위하여 DMSO(dimethyl sulfoxide)를 첨가하여 10 내지 20분 동안 혼합시킨 후, ApoTox-GloTM(Promega, 미국)을 이용하여 흡광도(570 내지 630 ㎚)를 측정하였다.Specifically, SH-SY5Y cells were cultured in a 96-well plate with 200 μl DMEM medium at 1 × 10 5 / well concentration. After 48 hours, the cells were further incubated with AβO (0.5 μm) alone, Coa-A alone (10, 20 and 30 μg / ml), or medium containing Aβ 1-42 and Coa-A, respectively. After 24 hours, MTT solution dissolved in phosphate buffered saline (PBS) at a concentration of 5 ml / ml was added to each well, and further incubated at 37 to 4 hours. Thereafter, DMSO (dimethyl sulfoxide) was added to dissolve formazone and mixed for 10 to 20 minutes, and then absorbance (570 to 630 nm) was measured using ApoTox-Glo (Promega, USA).
3. 3. 실험 동물Experimental animals 및 약물 처리 And drug treatment
APP/PS1 이중 형질전환 마우스는 인간의 돌연변이 APP 유전자 및 돌연변이 PS1(preselin) 유전자가 삽입된 형질전환 동물로서, 노인 반점 및 아밀로이드 베타 축적이 유의하게 발생하여 AD의 신경병리학적 특성을 나타내는 동물이다.APP / PS1 double transgenic mice are transgenic animals in which a human mutant APP gene and a mutant PS1 (preselin) gene are inserted, and exhibit an neuropathological characteristic of AD due to the significant development of geriatric spots and amyloid beta accumulation.
8주령의 수컷 APP/PS1 이중 형질전환 마우스는 Jackson Laboratory(메인, 미국)에서, C57BL/6J 야생형 마우스는 샘타코 코리아(SAMTAKO Korea, 대한민국)에서 구입하여 25℃에서 12시간 명암주기 및 임의 섭식 조건으로 사육하였다. 동물의 희생 및 고통을 최소화하기 위하여 모든 노력을 하였으며, 실험 과정은 경상대학교 동물 윤리 위원회의 승인을 받아 수행하였다. Eight-week-old male APP / PS1 double transgenic mice were purchased from Jackson Laboratory (Main, USA), and C57BL / 6J wild-type mice were purchased from Samtako Korea (South Korea) at 25 ° C. Breeding in a 12 hour light cycle and random feeding conditions. Every effort has been made to minimize the sacrifice and suffering of the animals, and the experimental procedure was carried out with the approval of the Animal Ethics Committee of Gyeongsang National University.
마우스의 연령이 12개월이 되었을 때 무작위로 네 그룹(n=10)으로 나누어 실험에 이용하였다: vehicle을 투여한 야생형 마우스, Coa-A를 투여한 야생형 마우스, vehicle을 투여한 APP/PS1 마우스 및 Coa-A를 투여한 APP/PS1 마우스. vehicle(0.9% 식염수) 또는 Coa-A(50 ㎎/㎏)는 2주 동안 날마다 복강 내 (inintraperiton, i.p.)에 투여하였다.When the mice were 12 months old, they were randomly divided into four groups (n = 10) and used in the experiments: wild type mice with vehicle, wild type mice with Coa-A, APP / PS1 mice with vehicle, and APP / PS1 mice administered Coa-A. A vehicle (0.9% saline) or Coa-A (50 mg / kg) was administered intraperitoneally (inintraperiton, i.p.) every day for two weeks.
4. 단백질 분석4. Protein Analysis
4-1. 4-1. 웨스턴Weston 블롯팅Blotting
BioRad protein assay kit(BioRad Laboratories, 캘리포니아, 미국)를 이용하여 단백질 농도를 측정한 후, 20 ㎍의 단백질을 4 내지 12% BoltTM mini gel(Novex, Life Technologies, 키리야트 시모나, 이스라엘)을 이용하여 전기영동하여 PVDF 막으로 트랜스퍼하였다. 이후 비특이적 결합을 줄이기 위하여 막을 5%(w/v) 스킴 밀크로 블록킹하고, 1차 항체와 4에서 하룻밤 동안 반응시켰다. 이후 막을 세척하고, 2차 항체와 반응시킨 후 ECL 탐지 시약(Amersham Pharmacia Biotech, 웁살라, 스웨덴)을 이용하여 제조사의 지시에 따라 단백질 밴드를 확인하였다. 모든 엑스-레이 필름은 스캔한 후 시그마 젤 프로그램 버전 1.0(Sigma Gel program, version 1.0, SPSS, 일리노이주, 미국)을 이용하여 밴드의 강도를 분석하였다. Protein concentration was measured using a BioRad protein assay kit (BioRad Laboratories, Calif., USA), and then 20 μg of protein was collected using 4-12% Bolt TM mini gel (Novex, Life Technologies, Kiryat Simona, Israel). Electrophoresis was transferred to the PVDF membrane. The membrane was then blocked with 5% (w / v) scheme milk to reduce nonspecific binding and reacted with the primary antibody at 4 overnight. The membrane was then washed, reacted with secondary antibodies and protein bands were identified according to the manufacturer's instructions using ECL detection reagents (Amersham Pharmacia Biotech, Uppsala, Sweden). All x-ray films were scanned and the intensity of the bands was analyzed using Sigma Gel program, version 1.0, SPSS, Illinois, USA.
사용한 1차 항체는 항-HSP70, 항-p-IKK(Cell Signaling, 매사추세츠, 미국), 항-β-actin, 항-PSD95, 항-Iba-1, 항-GFAP, 항-amyloid C-terminus, 항-BACE-1, 항-phospho-tau(Ser413), 항-Synaptophysin, 항-ADAM10, 항-p-NF-κB, 항-COX2, 항-iNOS, 항-IL-1β, 항-IDE(Santa Cruz Biotechnology, 캘리포니아, 미국), 항-APP, 항- ADAM17(Abcam technologies, 영국) 및 항-amyloid 6E10(BioLegends, 캘리포니아, 미국)이다.Primary antibodies used were anti-HSP70, anti-p-IKK (Cell Signaling, Massachusetts, USA), anti-β-actin, anti-PSD95, anti-Iba-1, anti-GFAP, anti-amyloid C-terminus, Anti-BACE-1, anti-phospho-tau (Ser413), anti-Synaptophysin, anti-ADAM10, anti-p-NF-κB, anti-COX2, anti-iNOS, anti-IL-1β, anti-IDE (Santa Cruz Biotechnology, California, USA), anti-APP, anti-ADAM17 (Abcam technologies, UK) and anti-amyloid 6E10 (BioLegends, CA, USA).
4-2. 효소면역(Enzyme-linked immunosorbent assay) 분석4-2. Enzyme-linked immunosorbent assay
마우스 뇌 분쇄액(homogenate) 내의 수용성 Aβ1 -42 및 Aβ1 -40 농도는 Human Aβ1 -42 및 Aβ1 -40 EILSA kits(Invitrogen, 캘리포니아, 미국)를 이용하여 제조사의 지시에 따라 측정하였다.Mouse brain grinding soluble Aβ 1 -42 1 -40, and Aβ concentration in the solution (homogenate) using a Human Aβ 1 -42, and Aβ 1 -40 EILSA kits (Invitrogen, CA, USA) was determined according to the manufacturer's instructions.
5. 뇌 조직 분석5. Brain Tissue Analysis
5-1. 뇌 단면 슬라이드 제작5-1. Brain Section Slide Production
조직 분석을 위하여 얼음으로 냉각시킨 4% 파라포름알데하이드(paraformaldehyde)를 마우스에 경심관류로 관류시키고, 뇌를 4% 파라포름알데하이드로 72시간 동안 후고정(post-fix)시킨 후 20% 수크로오스에 72시간 동안 보관하였다. 뇌를 O.C.T compound(A.O, 미국)로 얼리고, CM 3050C 크라이오스탯(Leica, 독일)을 이용하여 14-㎛ 두께로 관상면(coronal sections)을 잘랐다. 자른 관상면은 probe-on plus charged slide(Fisher, 일리노이주, 미국)에 녹이면서 붙였다.For tissue analysis, ice-cold 4% paraformaldehyde was perfused to the mouse via perfusion, and the brain was post-fixed with 4% paraformaldehyde for 72 hours, followed by 72% in 20% sucrose. Stored for hours. Brains were frozen with O.C.T compound (A.O, USA) and coronal sections were cut to 14-μm thick using a CM 3050C cryostat (Leica, Germany). The cut coronal plane was attached by melting on a probe-on plus charged slide (Fisher, Illinois, USA).
5-2. 5-2. 면역형광Immunofluorescence 분석 analysis
뇌 조직 내의 단백질 발현을 확인하기 위하여 형광염색을 실시하였으며, 이중염색을 위하여 염색 과정은 병행하여 진행하였다.Fluorescent staining was performed to confirm protein expression in brain tissues, and staining was performed in parallel for dual staining.
구체적으로, 슬라이드를 0.01 M PBS로 15분씩 두 번 세척하고, 프로테이나아제-k 용액을 첨가하여 37에서 5분 동안 반응시켰다. 이후 5% 정상 염소 혈청, 0.3% 트리톤 X-100 및 PBS로 이루어진 블록킹 용액과 실온에서 1시간 동안 반응시키고, 1:100으로 희석한 1차 항체를 처리하여 하룻밤 동안 반응시켰다. 다음날 TRITC- 또는 FICT가 결합된 2차 항체(Santa Cruz)를 PBS로 1:50으로 희석하여 90분 동안 반응시킨 후 슬라이드를 Prolong Antifade Reagent(Molecular Probe, 유진, 미국)로 마운팅하여 공초점 레이저-스캐닝 현미경(Flouview FV 1000, Olympus, 일본)으로 관찰하였다.Specifically, the slides were washed twice with 0.01 M PBS for 15 minutes each and reacted for 37 to 5 minutes by the addition of proteinase-k solution. After reacting with a blocking solution consisting of 5% normal goat serum, 0.3% Triton X-100 and PBS for 1 hour at room temperature, the primary antibody diluted 1: 100 was reacted overnight. The next day TRITC- or FICT-bound secondary antibody (Santa Cruz) was diluted 1:50 in PBS and reacted for 90 minutes, and then the slides were mounted with Prolong Antifade Reagent (Molecular Probe, Eugene, USA) for confocal laser- Observation was made with a scanning microscope (Flouview FV 1000, Olympus, Japan).
5-3. 5-3. 티오플라빈Thioflavin S 염색( S dyeing thioflavinthioflavin S staining) S staining)
뇌에 있는 아밀로이드 플라크(amyloid plaques)를 확인하기 위하여 티오플라빈 S 염색을 실시하였다.Thioflavin S staining was performed to identify amyloid plaques in the brain.
구체적으로, 슬라이드를 0.01 M PBS로 10분씩 두 번 세척하고, 1% 티오플라빈 S 용액에 담근 후 실온에서 20분 동안 방치하였다. 이후 70% 에탄올에 5분 동안 담근 후 물로 2번 세척하고, 요오드화 프로피디움(propidium iodide, PI)으로 대조염색(counterstaining)한 후 커버슬립을 덮어 관찰하였다.Specifically, the slides were washed twice with 0.01 M PBS for 10 minutes, immersed in 1% thioflavin S solution and left at room temperature for 20 minutes. After immersing in 70% ethanol for 5 minutes, washed twice with water, counterstained with propidium iodide (PI) and then covered with a cover slip.
6. 모리스 수중 미로 실험 및 Y-미로 실험 6. Morris Underwater Maze Experiment and Y-Maze Experiment
6-1. 모리스 수중 미로 실험6-1. Morris Underwater Maze Experiment
해마-의존적(hippocampus-dependent) 학습 및 기억 능력을 평가하기 위하여, 모리스 수중 미로 실험을 실시하였다.In order to assess hippocampus-dependent learning and memory ability, a Morris underwater maze experiment was conducted.
마우스(n=10/그룹)는 연속 3일 동안 하루에 2번씩 훈련받았으며, 모리스 수중 미로에서 60초 이상의 시간 동안 마우스가 숨겨진 도피대를 찾는 탈출 시간을 계산하였다. 마우스가 60초 동안 도피대를 발견하지 못할 경우, 도피대로 안내하여 도피대에서 30초 동안 머무르도록 하였다. 2주 동안 Coa-A 또는 vehicle을 투여한 후에, 연속 5일 동안 훈련을 계속하였고, 탈출 시간을 평가하였다. 마우스는 3일 동안 휴식을 취하게 한 후 숨겨진 도피대 없이 프로브 테스트를 수행하여 도피대가 있던 사분면에서 소비하는 시간을 기록하였다.Mice (n = 10 / group) were trained twice a day for three consecutive days, and the escape time for the mouse to find hidden escapes was calculated for more than 60 seconds in the Morris underwater maze. If the mouse did not find the shelters for 60 seconds, they were guided to the shelters to stay at the shelters for 30 seconds. After administering Coa-A or vehicle for 2 weeks, training was continued for 5 consecutive days and evacuation time was evaluated. Mice were allowed to rest for 3 days and then probe tests without hidden hides were recorded to record the time spent in the quadrant where the hides were.
6-2. Y-미로 실험6-2. Y-maze experiment
해마-의존적 공간 기억 능력을 평가하기 위하여 Y-미로 실험을 실시하였다. 훈련 기간 후에, 마우스를 Y 미로의 중앙에 놓아 8분 동안 무작위로 자유롭게 이동하도록 허용하였으며, 가지에 들어간 총 횟수 및 성공적인 트리플을 행동 소프트웨어 시스템으로 기록하였다. 변경 행동율(alteration behavior percentage, %)은 [성공적인 트리플 세트(/가지에 들어간 전체 횟수-2]*100으로 계산하였다. 변경 행동율의 비율은 공간 작업 기억 능력과 양의 상관관계가 있다.Y-maze experiments were conducted to assess hippocampal-dependent spatial memory capacity. After the training period, the mouse was placed in the center of the maze to allow free movement at random for 8 minutes, and the total number of branches and successful triples were recorded with the behavioral software system. The alteration behavior percentage (%) was calculated as [successful triple set (total number of branches-2) * 100. The percentage of alteration behavior rate is positively correlated with the spatial working memory capacity.
7. 통계 분석7. Statistical Analysis
단백질 밴드의 밀도는 임의 단위(arbitrary units)로 평균±표준 평균 오차(standard error mean, SEM) 수치를 표기하였다. 통계적 분석은 모든 실험에서 일원 분산 분석(one-way analysis of variance, ANOVA)을 수행하였고, Prism 5(graph Pad Software, 캘리포니아, 미국)를 이용하여 Student's t-test를 추가로 실시하였다. 형태 분석 결과 및 아밀로이드 반점에 대한 결과는 Image J software(미국의 국립보건원에서 제공하는 공개 소프트웨어)를 이용하여 분석하였으며, Integral Optical Densities(IODs)로 표시하였다. 결과값이 p<0.05, 0.01 및 0.001일 경우 유의한 것으로 판단하였다.The density of the protein bands is expressed as mean ± standard error mean (SEM) values in arbitrary units. Statistical analysis was performed by one-way analysis of variance (ANOVA) in all experiments, and further Student's t-test using Prism 5 (graph Pad Software, California, USA). The results of morphology analysis and amyloid spots were analyzed using Image J software (open source software provided by the National Institutes of Health, USA) and expressed as Integral Optical Densities (IODs). The results were considered significant when p <0.05, 0.01 and 0.001.
실험결과Experiment result
1. One. CoaCoa -A 처리에 의한 신경세포의 생존율 분석 결과Survival Analysis Results of Neurons by -A Treatment
Coa-A 처리에 의한 세포 생존율 변화를 확인하기 위하여 MTT assay를 수행하였다. 그 결과, AβO(0.5 ㎛)를 처리한 실험군은 대조군과 비교하여 SH-SY5Y 세포 생존율이 현저히 감소하고, Coa-A를 처리한 실험군은 세 농도 모두(10, 20 및 30 ㎍/㎖)에서 세포 독성이 나타나지 않는 것을 확인하였다. 반면에 AβO 및 Coa-A를 같이 처리한 실험군은 AβO만 처리한 실험군과 비교하여 세포 생존율이 현저하게 증가한 것을 확인하였다(도 1).MTT assay was performed to confirm cell viability change by Coa-A treatment. As a result, the experimental group treated with AβO (0.5 μm) significantly reduced the survival rate of SH-SY5Y cells compared to the control group, and the experimental group treated with Coa-A showed cells at all three concentrations (10, 20 and 30 μg / ml). It was confirmed that no toxicity appeared. On the other hand, the experimental group treated with AβO and Coa-A was confirmed that the cell survival rate was significantly increased compared to the experimental group treated with only AβO (FIG. 1).
2. 2. CoaCoa -A 처리에 의한 단백질 발현 변화 분석Analysis of Protein Expression Changes by -A Treatment
SH-SY5Y 세포에서 AβO(0.5 ㎛) 처리에 의한 HSP70 및 BACE1 단백질의 발현 변화를 확인하였다. Expression changes of HSP70 and BACE1 proteins were confirmed by AβO (0.5 μm) treatment in SH-SY5Y cells.
그 결과, AβO를 처리한 실험군의 경우 시간 의존적으로 6, 12 및 24 시간(각각 p<0.05, 0.01 및 0.001) 모두에서 HSP70 단백질의 발현은 현저하게 감소하고, BACE1 단백질의 발현은 증가하는 것을 확인하였다(도 2). 반면, Coa-A를 처리한 실험군은 세 농도 모두(10, 20 및 30 ㎍/㎖)에서 AβO 처리에 의한 HSP70 단백질의 발현 억제를 역전하여 HSP70 단백질의 발현이 증가하는 것을 확인하였다(24 시간에서 가장 유의한 증가를 보임; p<0.001)(도 3).As a result, in the experimental group treated with AβO, the expression of HSP70 protein was markedly decreased and the expression of BACE1 protein was increased at all 6, 12 and 24 hours (p <0.05, 0.01 and 0.001, respectively) time-dependently. (FIG. 2). On the other hand, the experimental group treated with Coa-A reversed the inhibition of expression of HSP70 protein by AβO treatment at all three concentrations (10, 20 and 30 µg / ml) and confirmed that the expression of HSP70 protein increased (at 24 hours). Most significant increase; p <0.001) (FIG. 3).
3. 3. CoaCoa -A 투여에 의한 뇌 조직 내 단백질 발현 변화 분석Analysis of Protein Expression in Brain Tissue by A-A Administration
알츠하이머 동물 모델인 APP/PS1 마우스에서 HSP70의 발현에 Coa-A가 미치는 영향을 확인하였다.The effect of Coa-A on the expression of HSP70 in the Alzheimer's animal model APP / PS1 mice was confirmed.
웨스턴 블롯 결과, 야생형 마우스와 비교하여 vehicle을 투여한 APP/PS1 마우스에서 HSP70 단백질의 발현이 감소하였으며, 대조적으로 Coa-A를 투여한 APP/PS1 마우스에서는 HSP70 단백질의 발현이 유의하게 증가한 것을 확인하였다. 또한, Coa-A를 투여시 APP/PS1 마우스의 뇌에서 BACE1 단백질의 발현이 현저하게 억제된 것을 확인하였다(도 4).Western blot results showed that the expression of HSP70 protein was reduced in APP / PS1 mice treated with vehicle compared to wild-type mice, whereas the expression of HSP70 protein was significantly increased in APP / PS1 mice treated with Coa-A. . In addition, it was confirmed that the expression of BACE1 protein was significantly inhibited in the brain of APP / PS1 mice when Coa-A was administered (FIG. 4).
또한, 면역형광 염색 결과를 통하여 Coa-A를 투여한 APP/PS1 마우스의 해마 및 피질(cortex)에서 HSP70의 발현이 증가하고, vehicle을 투여한 APP/PS1 마우스에는 감소하는 것을 확인하였다(도 5).In addition, the expression of HSP70 was increased in the hippocampus and cortex of APP / PS1 mice administered with Coa-A and decreased in APP / PS1 mice administered with vehicle through immunofluorescence staining (FIG. 5). ).
4. 4. CoaCoa -A 투여에 의한 뇌 조직 내 In brain tissue by -A administration Aβ의Of Aβ 생산 및 응집 개선 효과 분석 Analysis of production and coagulation improvement effects
HSP70 단백질은 단백질의 응집을 억제하고, 뉴런을 보호하는 것으로 알려져 있다. 따라서, APP/PS1 마우스에서 Aβ의 생산 및 응집에 Coa-A가 미치는 영향을 조사하였다.HSP70 protein is known to inhibit protein aggregation and protect neurons. Therefore, the effect of Coa-A on the production and aggregation of Aβ in APP / PS1 mice was investigated.
웨스턴 블롯으로 확인한 결과, 야생형 마우스와 비교하여 APP/PS1 마우스에서 아밀로이드 생성 경로(amyloidogenic pathway)의 활성이 현저하게 증가한 것을 확인하였다. 구체적으로, 아밀로이드 전구체 단백질(amyloid precursor protein, APP), Aβ 올리고머, 섬유성 Aβ(fibrillar Aβ) 및 인산화된 타우(phosphor tau) 단백질의 양이 유의하게 증가하였다. 반면, Coa-A를 2주 동안 투여한 APP/PS1 마우스에서는 상기 단백질들의 발현이 감소하였으며, 아밀로이드 생성 반응이 억제되는 것을 확인하였다(도 6).As a result of Western blot, it was confirmed that the activity of amyloidogenic pathway was significantly increased in APP / PS1 mice compared to wild type mice. Specifically, the amounts of amyloid precursor protein (APP), Aβ oligomer, fibrillar Aβ and phosphorylated tau protein were significantly increased. On the other hand, APP / PS1 mice administered with Coa-A for 2 weeks decreased the expression of the proteins and confirmed that the amyloidogenesis response was suppressed (FIG. 6).
또한, Coa-A를 투여한 경우 ADAM10(A disintegrin and metalloproteinase domain-containing protein 10) 및 ADAM17과 같은 α-세크레타아제(s-APPα) 유전자 발현이 증가하여 비-아밀로이드 생성 경로(non-amyloidogenic pathway)가 작용하는 것을 확인하였다(도 7). In addition, Coa-A administration increased expression of α-secretase (s-APPα) genes such as A disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) and ADAM17, resulting in a non-amyloidogenic pathway. ) Was confirmed to act (Fig. 7).
아울러 마우스의 뇌 분쇄액에서 수용성 Aβ1 -40 및 Aβ1 -42의 수준을 측정한 결과, vehicle을 투여한 APP/PS1 마우스와 비교하여 Coa-A를 투여한 APP/PS1 마우스에서 수용성 Aβ1 -40 및 Aβ1 - 42이 감소한 것을 확인하였다(도 8).In addition, a result of measuring the levels of soluble Aβ, and Aβ 1 -40 1 -42 in the mouse brains grinding liquid, compared to the administration of vehicle APP / PS1 mouse soluble in an APP / PS1 mice administered the Coa-A Aβ 1 - 40 and Aβ 1 - 42 was found to have decreased (Fig. 8).
또한, 뇌 조직의 티오플라빈 S 및 Aβ(C-말단 및 6E10) 염색을 비교한 결과, 야생형 마우스와 비교하여 APP/PS1 마우스의 해마 및 피질에서는 Aβ 반점 및 Aβ의 양이 증가하고, Coa-A를 투여한 APP/PS1 마우스에서는 Aβ의 양이 감소할 뿐만 아니라 Aβ 반점의 크기 및 반복 정도가 감소하는 것을 확인하였다(도 9 내지 도 11).In addition, the comparison of thioflavin S and Aβ (C-terminal and 6E10) staining of brain tissues showed that the amount of Aβ spots and Aβ increased in the hippocampus and cortex of APP / PS1 mice compared to wild-type mice, and Coa- APP / PS1 mice administered A were found not only to reduce the amount of Aβ but also to reduce the size and repeatability of Aβ spots (FIGS. 9 to 11).
또한, 뇌 조직에서 HSP70 및 Aβ를 이중 염색한 결과, Coa-A 투여에 의하여 뇌의 해마 및 피질에서 HSP70 단백질 발현이 현저하게 증가하고, Aβ의 응집이 감소하는 것을 확인하였다(도 12).In addition, as a result of double staining of HSP70 and Aβ in brain tissues, it was confirmed that CoSP-A administration significantly increased HSP70 protein expression in the hippocampus and cortex of the brain and decreased aggregation of Aβ (FIG. 12).
5. 5. CoaCoa -A 투여에 의한 뇌 조직 내 신경염증(Inflammation in brain tissue by -A administration neuroinflammationneuroinflammation ) 억제효과 분석) Inhibitory effect analysis
미세아교세포(microglia) 및 성상교세포(astrocyte)를 포함한 활성화된 교세포(glial cell) 및 연관된 신경염증은 AD 뇌의 중요한 특징 중의 하나이다. 따라서, Coa-A 투여가 교세포의 활성 및 신경염증에 미치는 영향을 확인하였다.Activated glial cells, including microglia and astrocytes, and associated neuroinflammation are one of the important features of the AD brain. Therefore, the effect of Coa-A administration on the activity and neuroinflammatory glial cells was confirmed.
미세아교세포 마커인 Iba1(ionized calcium-binding adapter molecule 1) 및 성상교세포 마커인 아교섬유산성단백질(glial fibrillary acidic protein, GFAP)의 발현을 면역형광 염색으로 확인한 결과, vehicle을 투여한 APP/PS1 마우스의 뇌에는 현저하게 많은 수의 활성화된 교세포가 있는 것을 확인하였다. 반면에 Coa-A를 투여한 APP/PS1 마우스의 뇌에서는 교세포의 활성이 억제된 것을 확인할 수 있었다(도 13 및 도 14).Expression of microglia marker Iba1 (ionized calcium-binding adapter molecule 1) and astrocyte markers glial fibrillary acidic protein (GFAP) was confirmed by immunofluorescence staining. 'S brain has a significant number of activated glial cells. On the other hand, it was confirmed that glial cell activity was inhibited in the brain of APP / PS1 mice administered with Coa-A (FIGS. 13 and 14).
또한 NF-κB 활성화 경로는 면역 반응에서 중요한 역할을 하는 것으로 알려져 있기 때문에, Coa-A 투여에 의한 영향을 확인하였다.In addition, since NF-κB activation pathway is known to play an important role in the immune response, the effect of Coa-A administration was confirmed.
웨스턴 블롯으로 확인한 결과, 야생형 마우스와 비교하여 vehicle을 투여한APP/PS1 마우스에서 인산화된 NF-κB, 인산화된 IκB kinase(IKK), 고리형 산화효소2(cyclooxygenase2, COX2), 유도형 산화질소 합성효소(inducible nitric oxide synthetase), 인터루킨-1 베타(IL-1β)의 수준이 현저하게 증가하는 것을 확인하였다(p<0.001). 반면 vehicle을 투여한 APP/PS1 마우스와 비교하여 Coa-A를 투여한 APP/PS1 마우스에서는 상기 분자들의 발현이 유의하게 감소한 것을 확인할 수 있었다(p<0.001)(도 15). Western blot confirmed that phosphorylated NF-κB, phosphorylated IκB kinase (IKK), cyclooxygenase2 (COX2), and induced nitric oxide synthesis in vehicle-treated APP / PS1 mice compared to wild-type mice. The levels of enzymes (inducible nitric oxide synthetase) and interleukin-1 beta (IL-1β) were significantly increased (p <0.001). On the other hand, it was confirmed that the expression of the molecules was significantly reduced in APP / PS1 mice administered with Coa-A compared to vehicle-administered APP / PS1 mice (p <0.001) (FIG. 15).
6. 6. CoaCoa -A 투여에 의한 전- 및 Pre- and by -A administration 후시냅스Postsynapse 기능 장애(pre- and  Dysfunction) postsynapticpostsynaptic dysfunction) 개선 효과 분석 dysfunction) improvement effect analysis
Coa-A가 전- 및 후시냅스 단백질(pre- and postsynaptic protein) 발현에 미치는 영향을 확인하였다.The effect of Coa-A on pre- and postsynaptic protein expression was confirmed.
뇌 분쇄액에서 웨스턴 블롯으로 확인한 결과, 야생형 마우스와 비교하여 vehicle을 투여한 APP/PS1 마우스에서 시냅토파이신(synaptophysin, SYP) 및 PSD95(postsynaptic density protein 95)의 발현이 감소하고, Coa-A를 투여한 APP/PS1 마우스에서는 발현이 현저하게 증가한 것을 확인하였다(도 16). Western blot analysis of brain pulmonary fluid showed that synaptophysin (SYP) and PSD95 (postsynaptic density protein 95) expression decreased in CoA-A in vehicle-treated APP / PS1 mice compared to wild-type mice. It was confirmed that expression was significantly increased in the APP / PS1 mice administered (Figure 16).
또한, 뇌의 피질 및 해마의 치아 이랑(dentate gyrus, DG)에서 SYP 및 PSD95를 염색한 결과 상기와 유사한 결과를 확인하였다(도 17).In addition, staining of SYP and PSD95 in the cerebral cortex and hippocampus of the teeth (dentate gyrus, DG) confirmed similar results to the above (FIG. 17).
7. 7. CoaCoa -A 투여에 의한 학습 및 기억력 개선 효과 분석-A-Administration of Learning and Memory Improvement Effects
학습 및 공간 기억 능력을 평가하기 위하여 미로 실험을 수행하였다.Maze experiments were conducted to evaluate learning and spatial memory.
Vehicle 또는 Coa-A 투여 전의 훈련에서 1일 차에 야생형 마우스 및 APP/PS1 마우스가 숨겨진 도피대를 찾는 탈출 시간을 비교한 결과, APP/PS1 마우스가 약간 높은 것을 확인하였다. 2일 및 3일 차에는 두 마우스 모두 탈출 시간이 감소한 양상을 나타내었으나, APP/PS1 마우스와 비교하여 야생형 마우스가 현저하게 감소한 탈출 시간을 나타내었다(도 18).In the training before vehicle or Coa-A administration, the escape time for wild-type mice and APP / PS1 mice to find hidden hides was compared to find that APP / PS1 mice were slightly higher. On day 2 and day 3, both mice showed reduced escape time, but significantly reduced escape time of wild-type mice compared to APP / PS1 mice (FIG. 18).
Coa-A를 2주 동안 투여한 이후 탈출 시간을 측정한 결과, 1일 차에서는 vehicle을 투여한 APP/PS1 마우스와 비교하여 Coa-A를 투여한 APP/PS1 마우스의 탈출 시간이 현저한 비율로 감소한 것을 확인하여, 학습 능력이 향상된 것을 알 수 있었다. 또한, 훈련 2일에서 5일 차까지 비교한 결과 vehicle을 투여한 APP/PS1 마우스와 비교하여 Coa-A를 투여한 APP/PS1 마우스의 탈출 시간이 유의하게 낮은 것을 확인하였다(도 19). After estimating Coa-A for two weeks, the escape time was measured. On Day 1, the escape time of Coa-A-administered APP / PS1 mice was significantly reduced compared to the vehicle-administered APP / PS1 mice. It was confirmed that the learning ability was improved. In addition, as a result of the comparison from the second to the fifth day of training, the evacuation time of the APP / PS1 mice administered with Coa-A was significantly lower than that of the APP / PS1 mice administered with the vehicle (FIG. 19).
공간 기억 유지 능력을 확인하기 위하여, 프로브 테스트를 실시하여 도피대를 제거하고, 도피대가 있던 표적 사분면에서 체류하는 시간을 측정하였다. 그 결과 vehicle을 투여한 APP/PS1 마우스와 비교하여 Coa-A를 투여한 APP/PS1 마우스가 표적 사분면에서 더 긴 시간을 체류하는 것을 확인하였다(도 20).In order to confirm the spatial memory retention ability, a probe test was performed to remove the sheath and to measure the time to stay in the target quadrant where the sheath was. As a result, it was confirmed that the APP / PS1 mice administered Coa-A stayed longer in the target quadrant compared to the APP / PS1 mice administered vehicle (FIG. 20).
또한, 단기 기억에 미치는 영향을 확인하기 위하여 Y-미로 실험을 실시한 결과, 야생형 마우스와 비교하여 vehicle을 투여한 APP/PS1 마우스의 변경 행동율이 더 낮았으며, vehicle을 투여한 APP/PS1 마우스와 비교하여 Coa-A를 투여한 APP/PS1 마우스의 변경 행동율이 더 높은 것을 확인하였다(도 21).In addition, the Y-maze experiments were conducted to confirm the effect on short-term memory, and the altered behavior rate of APP / PS1 mice with vehicle was lower than that of wild-type mice. In comparison, it was confirmed that the altered behavior rate of APP / PS1 mice administered Coa-A was higher (FIG. 21).
이제까지 본 발명에 대하여 그 바람직한 실시예들을 중심으로 살펴보았다. 본 발명이 속하는 기술 분야에서 통상의 지식을 가진 자는 본 발명이 본 발명의 본질적인 특성에서 벗어나지 않는 범위에서 변형된 형태로 구현될 수 있음을 이해할 수 있을 것이다. 그러므로 개시된 실시예들은 한정적인 관점이 아니라 설명적인 관점에서 고려되어야 한다. 본 발명의 범위는 전술한 설명이 아니라 특허청구범위에 나타나 있으며, 그와 동등한 범위 내에 있는 모든 차이점은 본 발명에 포함된 것으로 해석되어야 할 것이다.So far I looked at the center of the preferred embodiment for the present invention. Those skilled in the art will appreciate that the present invention can be implemented in a modified form without departing from the essential features of the present invention. Therefore, the disclosed embodiments should be considered in descriptive sense only and not for purposes of limitation. The scope of the present invention is shown in the claims rather than the foregoing description, and all differences within the scope will be construed as being included in the present invention.

Claims (7)

  1. 하기 화학식 1로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물:A pharmaceutical composition for preventing or treating degenerative neurological diseases containing a compound represented by the following Chemical Formula 1 or a pharmaceutically acceptable salt thereof as an active ingredient:
    [화학식 1][Formula 1]
    Figure PCTKR2017006986-appb-I000003
    Figure PCTKR2017006986-appb-I000003
  2. 제1항에 있어서, 상기 화학식 1로 표시되는 화합물은 HSP70의 발현을 촉진하고, BACE1의 발현을 억제하는 것을 특징으로 하는 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물.The pharmaceutical composition for preventing or treating degenerative neurological disease according to claim 1, wherein the compound represented by Chemical Formula 1 promotes expression of HSP70 and inhibits expression of BACE1.
  3. 제1항에 있어서, 상기 퇴행성 신경질환은 뇌졸중, 중풍, 기억력 상실, 기억력 손상, 치매, 파킨슨병 및 알츠하이머 병으로 이루어진 군에서 선택되는 어느 하나인 것을 특징으로 하는 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물.The method of claim 1, wherein the neurodegenerative disease is stroke, stroke, memory loss, memory impairment, dementia, Parkinson's disease and Alzheimer's disease, characterized in that any one selected from the group consisting of prevention or treatment of neurodegenerative diseases Composition.
  4. 제1항에 있어서, 상기 퇴행성 신경질환은 뇌세포 내 아밀로이드 베타 단백질에 의해 유도된 질환인 것을 특징으로 하는 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물.The method of claim 1, wherein the neurodegenerative disease is a pharmaceutical composition for the prevention or treatment of neurodegenerative diseases, characterized in that the disease induced by amyloid beta protein in brain cells.
  5. 제1항에 있어서, 상기 조성물은 신경세포의 염증반응을 억제하는 것을 특징으로 하는 퇴행성 신경질환의 예방 또는 치료용 약학적 조성물.According to claim 1, wherein the composition is a pharmaceutical composition for the prevention or treatment of degenerative neurological diseases, characterized in that to inhibit the inflammatory response of nerve cells.
  6. 하기 화학식 1로 표시되는 화합물을 유효성분으로 함유하는 퇴행성 신경질환의 예방 또는 개선용 건강기능식품:Health functional food for the prevention or improvement of degenerative neurological disease containing the compound represented by the formula (1) as an active ingredient:
    [화학식 1][Formula 1]
    Figure PCTKR2017006986-appb-I000004
    Figure PCTKR2017006986-appb-I000004
  7. 제6항에 있어서, 상기 화학식 1로 표시되는 화합물은 HSP70의 발현을 촉진하고, BACE1의 발현을 억제하는 것을 특징으로 하는 퇴행성 신경질환의 예방 또는 개선용 건강기능식품.The health functional food for preventing or improving degenerative neurological disease according to claim 6, wherein the compound represented by Chemical Formula 1 promotes the expression of HSP70 and inhibits the expression of BACE1.
PCT/KR2017/006986 2016-07-04 2017-06-30 Pharmaceutical composition for preventing or treating degenerative neuronal diseases, containing compound isolated from withania coagulans as active ingredient WO2018008908A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR1020160083944A KR101802659B1 (en) 2016-07-04 2016-07-04 A composition for preventing or protecting neurodegenerative diseases comprising compound purified from Withania coagulans
KR10-2016-0083944 2016-07-04

Publications (1)

Publication Number Publication Date
WO2018008908A1 true WO2018008908A1 (en) 2018-01-11

Family

ID=60811064

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2017/006986 WO2018008908A1 (en) 2016-07-04 2017-06-30 Pharmaceutical composition for preventing or treating degenerative neuronal diseases, containing compound isolated from withania coagulans as active ingredient

Country Status (2)

Country Link
KR (1) KR101802659B1 (en)
WO (1) WO2018008908A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20230321175A1 (en) * 2022-04-08 2023-10-12 Khalifa University of Science and Technology Anti-diabetic steroidal lactones of withania coagulans for treatment of type 2 diabetes

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010053655A2 (en) * 2008-11-07 2010-05-14 University Of Kansas Therapeutic methods with withaferin a and analogs

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010053655A2 (en) * 2008-11-07 2010-05-14 University Of Kansas Therapeutic methods with withaferin a and analogs

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
DUBEY, A.: "Heat shock proteins: a therapeutic target worth to consider", VETERINARY WORLD, vol. 8, no. 1, 2015, pages 46 - 51, XP055601614 *
JAHAN, E.: "Coagulansins A and B, New Withanolides from Withania coagulans Dunal", HELVETICA CHIMICA ACTA, vol. 93, no. 3, 2010, pages 530 - 535 *
KHAN, I.: "Coagulansin-A has beneficial effects on the development of bovine embryos in vitro via HSP70 induction", BIOSCI. REP., vol. 36, no. 2, April 2016 (2016-04-01), pages 1 - 10, XP055601611 *
KHODAEI, M.: "Remedial Use of Withanolides from Withania Coagolans (Stocks) Dunal", ADVANCES IN LIFE SCIENCES, 1 February 2012 (2012-02-01), pages 6 - 19 *

Also Published As

Publication number Publication date
KR101802659B1 (en) 2017-11-28

Similar Documents

Publication Publication Date Title
Chandra et al. Cinnamic acid activates PPARα to stimulate Lysosomal biogenesis and lower Amyloid plaque pathology in an Alzheimer's disease mouse model
WO2016140527A1 (en) Composition for preventing, improving or treating neurological disorders, containing osmotin peptide as active ingredient
Angot et al. Dissecting the potential molecular mechanisms underlying α-synuclein cell-to-cell transfer in Parkinson's disease
WO2011090270A2 (en) Composition containing osmotin for preventing and treating neurological disorders
Toba et al. PPARγ agonist pioglitazone improves cerebellar dysfunction at pre-Aβ deposition stage in APPswe/PS1dE9 Alzheimer's disease model mice
KR20180094556A (en) Composition for the improvement of memory and cognition ability, or prevention, delay, improvement or treatment of Alzheimer&#39;s disease comprising extract or fraction of Aronia Melanocarpa Elliott fruit
CA3047078A1 (en) Pharmaceutical composition for preventing or treating neurodegenerative diseases which includes flower extract of daphne genkwa or fractions thereof as active ingredient
KR102316236B1 (en) Compositons for differentiating oligodendrocytes containing gintonin and compositions for preventing or treating demyelinating diseases
Gooch et al. Molecular basis of Alzheimer’s disease
WO2018008908A1 (en) Pharmaceutical composition for preventing or treating degenerative neuronal diseases, containing compound isolated from withania coagulans as active ingredient
KR20170003395A (en) The pharmaceutical composition for the prevention or treatment of the symptoms in the dementia comprising the extracts from Coriandrum sativum
WO2009093864A2 (en) Composition for preventing or treating brain diseases
WO2013022139A1 (en) Method for treatment of diseases caused by accumulation of beta-amyloids in brain parenchyma with low concentration thiazolidinedione derivatives
WO2020250182A2 (en) Composition for treating neurodegenerative disease comprising gypenoside compound as active ingredient
WO2022114906A1 (en) Novel pharmaceutical composition for treating neurodegenerative diseases
WO2016043354A1 (en) Novel pharmaceutical composition for treating alzheimer&#39;s disease
WO2017131468A1 (en) Composition for preventing, alleviating or treating neurological disorders, containing, as active ingredient, novel peptide activating adiponectin receptors
US20220339230A1 (en) Composition comprising bilberry extract as active ingredient for preventing or treating sensorineural hearing impairment
WO2017105081A1 (en) Pharmaceutical composition for treating lung diseases or alleviating symptoms of lung diseases
KR102241282B1 (en) Composition for hard tissue formation and, dentin or pulp regeneration containing Ixeris dentata extract and its active compound
CN117202911A (en) Pharmaceutical composition for treating neuroinflammation comprising eleutheroside B as active ingredient
US20090131339A1 (en) Compounds having neuroprotective properties
WO2018080158A1 (en) Composition for preventing, improving or treating cognitive impairment, containing elaeagnus glabra extract as active ingredient
KR101906789B1 (en) Pharmaceutical composition for treatment or prevention of degenerative brain disease containing peucedanocoumarin iii
KR20100049768A (en) Compositions for prophylaxis or treatment of cerebrovascular diseases, or for improving memory impairments, containing ethanol extract from aralia cordata, vitis amurensis and glycyrrhizae radix

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: 17824464

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: 17824464

Country of ref document: EP

Kind code of ref document: A1