KR20190120967A - Composition for anti-inflammatory treatment comprising sesquiterpene-based metabolites - Google Patents

Composition for anti-inflammatory treatment comprising sesquiterpene-based metabolites Download PDF

Info

Publication number
KR20190120967A
KR20190120967A KR1020180044378A KR20180044378A KR20190120967A KR 20190120967 A KR20190120967 A KR 20190120967A KR 1020180044378 A KR1020180044378 A KR 1020180044378A KR 20180044378 A KR20180044378 A KR 20180044378A KR 20190120967 A KR20190120967 A KR 20190120967A
Authority
KR
South Korea
Prior art keywords
formula
composition
compound
inflammatory
sesquiterpene
Prior art date
Application number
KR1020180044378A
Other languages
Korean (ko)
Other versions
KR102074575B1 (en
Inventor
오현철
김윤철
윤치수
김관우
이상찬
Original Assignee
원광대학교산학협력단
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 원광대학교산학협력단 filed Critical 원광대학교산학협력단
Priority to KR1020180044378A priority Critical patent/KR102074575B1/en
Publication of KR20190120967A publication Critical patent/KR20190120967A/en
Application granted granted Critical
Publication of KR102074575B1 publication Critical patent/KR102074575B1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • A61K31/122Ketones having the oxygen directly attached to a ring, e.g. quinones, vitamin K1, anthralin
    • 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
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/33Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing oxygen
    • A61K8/35Ketones, e.g. benzophenone
    • A61K8/355Quinones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/49Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds
    • A61K8/4973Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds with oxygen as the only hetero atom
    • A61K8/498Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds with oxygen as the only hetero atom having 6-membered rings or their condensed derivatives, e.g. coumarin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin
    • 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
    • A23V2200/324Foods, ingredients or supplements having a functional effect on health having an effect on the immune system

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Birds (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Rheumatology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Immunology (AREA)
  • Pain & Pain Management (AREA)
  • Mycology (AREA)
  • Nutrition Science (AREA)
  • Food Science & Technology (AREA)
  • Polymers & Plastics (AREA)
  • Emergency Medicine (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Dermatology (AREA)
  • Medicines Containing Plant Substances (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to an anti-inflammatory composition containing sesquiterpene-based metabolites. More specifically, the present invention relates to the anti-inflammatory composition containing sesquiterpene-based metabolites which can be separated from an ethyl acetate fraction of a Nardostachys chinensis methanol extract. A composition of the present invention exhibits an excellent anti-inflammatory effect, thereby being able to be usefully used as a pharmaceutical composition, a cosmetic composition, and a food composition.

Description

세스퀴테르펜 계열 대사체를 포함하는 항염증 조성물{Composition for anti-inflammatory treatment comprising sesquiterpene-based metabolites}Composition for anti-inflammatory treatment comprising sesquiterpene-based metabolites {Composition for anti-inflammatory treatment comprising sesquiterpene-based metabolites}

본 발명은 세스퀴테르펜 계열 대사체를 함유하는 항염증 조성물에 관한 것으로, 보다 상세하게는 감송향 메탄올 추출물의 에틸아세테이트 분획물로부터 분리될 수 있는 세스퀴테르펜 계열 대사체를 함유하는 항염증 약학적 조성물, 화장품 조성물 및 식품 조성물에 관한 것이다.The present invention relates to an anti-inflammatory composition containing a sesquiterpene-based metabolite, and more particularly, to an anti-inflammatory pharmaceutical composition containing a sesquiterpene-based metabolite that can be separated from the ethyl acetate fraction of the methanol extract of saccharin. , Cosmetics compositions and food compositions.

염증성 질환이란 세균의 침입에 의해 형성되는 농양의 병리적 상태를 의미한다. 대표적인 염증성 질환으로는 뇌염, 골관절염, 류마티스 관절염, 통풍, 강직성 척추염, 건염, 건막염, 류마티스 열, 루프스, 섬유근통(Fibromyalgia), 건선 관절염, 천식, 아토피, 크론병, 궤양성 대장염 등이 있다. 염증은 물리적인 외상, 유해한 화학물질, 미생물에 의한 감염이나 생체 내 대사산물 중의 자극성 물질에 의하여 야기되는 조직손상에 대하여 국소적으로 나타나는 정상적이고 보호적인 생체 내 방어기전의 발현이다.Inflammatory disease refers to the pathological condition of an abscess formed by the invasion of bacteria. Representative inflammatory diseases include encephalitis, osteoarthritis, rheumatoid arthritis, gout, ankylosing spondylitis, tendinitis, tendonitis, rheumatic fever, lupus, fibromyalgia, psoriatic arthritis, asthma, atopy, Crohn's disease, ulcerative colitis. Inflammation is a manifestation of normal and protective in vivo defense mechanisms that are localized to tissue damage caused by physical trauma, harmful chemicals, microbial infections or irritants in metabolites in vivo.

생체에 있어서 염증의 발생에는 다양한 생화학적인 현상이 관여하고 있다. 대식세포(Macrophage)는 다양한 기능을 가진 세포로 화학적 자극에 의하여 여러 가지 사이토카인(cytokine)과 질소산화물(NO)을 생성하여 염증반응에서 중요한 역할을 한다. 특히, 대식세포에서 인터페론-감마(Interferons-γ, IFN-γ)와 같은 사이토카인 자극에 의해 발현되는 유도성 질소산화물 합성효소(iNOS)는 장시간 동안 다량의 질소산화물(NO)을 생산한다. 이러한 산화적 스트레스는 IκB에 의하여 억제되어 있는 염증 반응의 전사인자인 NF-κB활성을 촉진시키는 것으로 알려져 있다. 활성화된 NF-κB는 핵으로 이동하여 iNOS, COX-2 및 IL-1β나 TNF-α와 같은 여러 종류의 사이토카인 등 염증반응을 유도하는 유전자 발현을 촉진시키는 것으로 알려져 있으며, 이들 인자들을 저해하면 염증 반응이 억제되는 것으로 알려져 있다(Baeuerle et al., Annu. Rev. Immunol., 12:141-179, 1994).Various biochemical phenomena are involved in the development of inflammation in the living body. Macrophage is a multi-functional cell that plays an important role in the inflammatory response by generating various cytokines and NOx by chemical stimulation. In particular, inducible nitric oxide synthase (iNOS) expressed by cytokine stimulation such as interferons-gamma (IFN-γ) in macrophages produces large amounts of nitric oxide (NO) for a long time. This oxidative stress is known to promote NF-κB activity, a transcription factor of the inflammatory response inhibited by IκB. Activated NF-κB is known to promote the expression of genes that induce inflammatory responses, such as iNOS, COX-2, and various cytokines such as IL-1β or TNF-α. Inflammatory responses are known to be inhibited (Baeuerle et al., Annu. Rev. Immunol., 12: 141-179, 1994).

염증유발물질 중 하나인 NO는 정상상태에서는 내피세포나 대식세포에서 생산되며 세포살상과 살균작용 이외에도 다양한 생리활성을 나타내는데, 특히 혈관 내피세포의 이완작용에 있어서 혈압의 항상성(homeostasis)을 유지하는 역할을 하는 것으로 알려져 있다. LPS(lipopolysaccharide), 염증유발인자 및 방사선조사 등의 자극은 특히, 유도형 산화질소 합성효소인 iNOS의 발현을 유도하여 많은 양의 NO를 4 내지 6 시간 동안 계속적으로 생성시키는데 이와 같이 생성된 과도한 양의 NO는 상기와 같은 염증성 질환들을 유발한다. NO, one of the inflammatory substances, is produced in endothelial cells or macrophages in a normal state and exhibits various physiological activities in addition to cell killing and bactericidal action. In particular, it plays a role in maintaining homeostasis of blood pressure in the relaxation of vascular endothelial cells It is known to do. Stimulation of lipopolysaccharide (LPS), pro-inflammatory factors, and irradiation, in particular, induces the expression of iNOS, an inducible nitric oxide synthase, continuously producing large amounts of NO for 4-6 hours. NO causes such inflammatory diseases.

이러한 염증 질환을 치료하기 위한 가장 일반적인 염증성 질환 예방 또는 치료제는 크게 스테로이드성 및 비스테로이드성 염증성 질환 예방 또는 치료제로 구분되며, 이 중 대부분의 합성 염증성 질환 예방 또는 치료제는 주작용 이외에 여러 가지 부작용을 수반하는 경우가 많으므로 효과가 탁월하며 부작용이 적은 염증성 질환 예방 또는 치료제의 개발이 절실히 요구되고 있는 실정이다. 특히, 효능 및 부작용 측면에서 볼 때, 예로부터 임상적 경험이 풍부하고 안전성 측면에서 탁월한 평가를 받고 있는 천연물 제제가 염증 질환의 예방 및 치료제 개발에 있어 좋은 후보물질이 될 것으로 여겨진다.The most common inflammatory disease prevention or treatment for treating such inflammatory diseases is classified into steroidal and nonsteroidal inflammatory disease prevention or treatment, and most of these synthetic inflammatory disease prevention or treatment has various side effects in addition to the main action. In many cases, excellent effects and development of drugs for preventing or treating inflammatory diseases with fewer side effects are urgently needed. In particular, in terms of efficacy and side effects, natural product formulations, which have a long history of clinical experience and excellent evaluation in terms of safety, are considered to be good candidates for the development of preventive and therapeutic agents for inflammatory diseases.

감송향(Nardostachys jatamansi)은 전통적으로 위통, 위장경련, 흉복장만, 신경성 위장병, 구토, 두통, 각기 등에 사용되며, 몇몇 아시아 국가들에서 강장제, 자극제 및 항경련제로 사용되어 왔다(Bagchi, A., 등, Planta Med., 57, 9697 (1991)). Nardostachys jatamansi has traditionally been used for stomach pain, gastrointestinal spasms, chest glands, neuropathy, vomiting, headaches, and the like, and has been used as tonics, irritants and anticonvulsants in several Asian countries (Bagchi, A., Et al., Planta Med., 57, 9697 (1991)).

감송향의 뿌리에는 야타만시산(Jatamansic acid) 및 야타만손 (Jatamansone)과 같은 다양한 세스퀴테르펜 (sesquiterpenes), 리그난, 및 네오리그난이 존재하는 것으로 알려져 있다(Chatterji, A, 등, National Institute of Science Communication, vol. 5, pp 99-100 (1997); Arora, RB., Indian Council of Medical Research, vol. 51 (1965)).The roots of the nectar are known to contain various sesquiterpenes, lignans, and neolignans such as Jatamansic acid and Jatamansone (Chatterji, A, et al., National Institute of Science Communication, vol. 5, pp 99-100 (1997); Arora, RB., Indian Council of Medical Research, vol. 51 (1965)).

본 발명자들은 항염증 활성이 우수한 신규물질을 천연물 유래의 대사 산물로부터 연구하던 중, 감송향으로부터 항염증 활성이 우수한 세스퀴테르펜 계열 대사체를 분리 및 정제하고 이의 항염증 효과를 확인함으로써 본 발명을 완성하였다.The inventors of the present invention, while studying a novel substance with excellent anti-inflammatory activity from a metabolite derived from natural products, isolates and purifies sesquiterpene-based metabolites having excellent anti-inflammatory activity from sweet persimmon and confirms the anti-inflammatory effect thereof. Completed.

본 발명의 목적은 세스퀴테르펜 계열 대사체를 포함하는 염증성 질환의 치료 또는 예방용 약학적 조성물을 제공하는 것이다.An object of the present invention to provide a pharmaceutical composition for the treatment or prevention of inflammatory diseases comprising sesquiterpene-based metabolites.

또한, 본 발명은 상기 대사체를 포함하는 식품 조성물 및 화장품 조성물을 제공하고자 한다.In addition, the present invention is to provide a food composition and cosmetic composition comprising the metabolite.

본 발명의 다른 목적은 감송향 추출물로부터 분리된 항염증 효과를 갖는 신규한 세스퀴테르펜 계열 대사체를 제공하는 것이다.Another object of the present invention is to provide a novel sesquiterpene-based metabolite having an anti-inflammatory effect isolated from the extract of Persimmon.

본 발명은 신규한 세스퀴테르펜 계열 대사체인, 하기 화학식 I의 화합물 또는 이의 약제학적으로 허용가능한 염을 제공한다.The present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof, which is a novel sesquiterpene family of metabolites.

[화학식 I][Formula I]

Figure pat00001
Figure pat00001

(여기서, R는 α-OH, β-OH 또는 α-메톡시-α-트리플루오로메틸페닐아세테이트기임).Wherein R is an α-OH, β-OH or α-methoxy-α-trifluoromethylphenylacetate group.

본 발명의 상기 화학식 I에서 R이 β-OH 일 수 있으며, 화합물 1 또는 칸숀 J (kanshone J)라고 한다.In the above formula (I) of the present invention, R may be β-OH, and is referred to as compound 1 or kanshone J.

본 발명의 상기 화학식 I에서, R이 α-메톡시-α-트리플루오로메틸페닐아세테이트기(MTPA)일 수 있으며, (S) MTPA인 경우 화합물 1a, (R) MTPA인 경우 화합물 1b라고 한다.In the above formula (I) of the present invention, R may be an α-methoxy-α-trifluoromethylphenylacetate group (MTPA), and (S) MTPA is referred to as Compound 1a, and (R) MTPA is referred to as Compound 1b.

본 발명의 상기 화학식 I에서, R이 α-OH 일 수 있으며, 화합물 2 또는 칸숀 K (kanshone K)라고 한다.In the above formula (I) of the present invention, R may be α-OH, and is referred to as compound 2 or kanshone K.

또한, 본 발명은 하기의 세스퀴테르펜 계열 대사체 또는 이의 약제학적으로 허용가능한 염을 제공한다:The present invention also provides the following sesquiterpene family metabolites or pharmaceutically acceptable salts thereof:

화합물 3 : 데옥소-나치놀 A (desoxo-narchinol A)Compound 3: desoxo-narchinol A

Figure pat00002
Figure pat00002

화합물 4 : 나치놀 B (narchinol B)Compound 4: narchinol B

Figure pat00003
Figure pat00003

화합물 5 : 불라탄트리올 (bullatantriol)Compound 5: Bullatantriol

Figure pat00004
Figure pat00004

화합물 6 : 1β,4β,7α-트리히드록시엔데스만 (1β,4β,7α-trihydroxyeudesmane)Compound 6: 1β, 4β, 7α-trihydroxyendesman (1β, 4β, 7α-trihydroxyeudesmane)

Figure pat00005
Figure pat00005

화합물 7 : 테클라트리올 (teuclatriol)Compound 7: teclalatriol

Figure pat00006
Figure pat00006

화합물 8 : 4β-히드록시-8β메톡시-10-메틸렌-2,9-다이옥사트리사이클로[4.3.1.03,7]데케인 (4β-hydroxy-8β-methoxy-10-methylene-2,9-dioxatricyclo[4.3.1.03,7]decane)Compound 8: 4β-hydroxy-8βmethoxy-10-methylene-2,9-dioxatricyclo [4.3.1.03,7] decane (4β-hydroxy-8β-methoxy-10-methylene-2,9- dioxatricyclo [4.3.1.03,7] decane)

Figure pat00007
Figure pat00007

화합물 9 : 야타마닌 A (jatamanin A)Compound 9: Yatamanin A

Figure pat00008
Figure pat00008

본 발명은 상기 화합물 1 내지 9, 구체적으로 칸숀 J (kanshone J), 칸숀 K(kanshone K), 데옥소-나치놀 A(desoxo-narchinol A), 나치놀 B(narchinol B), 불라탄트리올(bullatantriol), 1β,4β,7α-트리히드록시엔데스만(1β,4β,7α-trihydroxyeudesmane), 테클라트리올(teuclatriol), 4β-히드록시-8β-메톡시-10-메틸렌-2,9-다이옥사트리사이클로[4.3.1.03,7]데케인(4β-hydroxy-8β-methoxy-10-methylene-2,9-dioxatricyclo[4.3.1.03,7]decane), 야타마닌 A(jatamanin A) 및 이들의 약제학적으로 허용가능한 염으로 이루어지는 군으로부터 선택되는 하나 이상의 세스퀴테르펜 계열 대사체를 유효성분으로 포함하는 염증성 질환의 예방 또는 치료용 약학적 조성물을 제공한다.The present invention relates to compounds 1 to 9, specifically Kanshone J, Kanshone K, Desoxo-narchinol A, Nachinol B, Bulatantriol (bullatantriol), 1β, 4β, 7α-trihydroxyendesman, 1β, 4β, 7α-trihydroxyeudesmane, teclalatriol, 4β-hydroxy-8β-methoxy-10-methylene-2,9 Dioxatricyclo [4.3.1.03,7] decane (4β-hydroxy-8β-methoxy-10-methylene-2,9-dioxatricyclo [4.3.1.03,7] decane), yatamanin A And it provides a pharmaceutical composition for the prevention or treatment of inflammatory diseases comprising at least one sesquiterpene-based metabolite selected from the group consisting of pharmaceutically acceptable salts thereof as an active ingredient.

여기서, 칸숀 J는 칸숀 J의 MTPA 에스테르를 포함할 수 있다.Here, Kanshon J may comprise the MTPA ester of Kanshon J.

바람직하게, 상기 조성물은 나치놀 A(desoxo-narchinol A), 나치놀 B(narchinol B) 또는 이의 약제학적으로 허용가능한 염을 유효성분으로 포함할 수 있다.Preferably, the composition may include desoxo-narchinol A, nachinol B, or a pharmaceutically acceptable salt thereof as an active ingredient.

이때 상기 세스퀴테르펜 계열 대사체는 감송향 메탄올 추출물의 에틸아세테이트 분획물로부터 분리될 수 있다.In this case, the sesquiterpene-based metabolites may be separated from the ethyl acetate fraction of the extract of Methanol.

본 발명의 조성물은 NO(Nitric oxide) 또는 PGE2(prostaglandin E2)의 생성을 억제함으로써 항염증 효과를 나타낼 수 있다.The compositions of the invention may also show anti-inflammatory effects by inhibiting the production of NO (Nitric oxide), or PGE 2 (prostaglandin E 2).

또한 본 발명의 조성물은 iNOS(inducible NO synthase) 또는 COX-2(Cyclooxygenase-2)단백질의 발현을 억제할 수 있다.In addition, the composition of the present invention can inhibit the expression of iNOS (inducible NO synthase) or COX-2 (Cyclooxygenase-2) protein.

뿐만 아니라, 본 발명의 조성물은 IL-1β(Interleukin 1β), IL-6(Interleukin 6) 또는 TNF-α(Tumor Necrosis Factor α)의 발현을 억제할 수 있다.In addition, the composition of the present invention can inhibit the expression of IL-1β (Interleukin 1β), IL-6 (Interleukin 6) or TNF-α (Tumor Necrosis Factor α).

이때, 상기 약학적 조성물은 분말, 과립, 정제, 환제, 당의정제, 캡슐제, 액제, 겔제, 시럽제, 슬러리제, 현탁액, 외용산제, 외용정제, 외용액제, 연고제, 크림제, 겔제, 경고제, 드레싱제, 패취제, 스프레이 또는 주사제의 형태로 제형화 될 수 있다.At this time, the pharmaceutical composition is a powder, granules, tablets, pills, dragees, capsules, solutions, gels, syrups, slurries, suspensions, external preparations, external preparations, external preparations, ointments, creams, gels, warnings It may be formulated in the form of a dressing, a patch, a spray or an injection.

또한 본 발명은 상기 세스퀴테르펜 계열 대사체를 유효성분으로 포함하는 화장품 조성물을 제공한다.The present invention also provides a cosmetic composition comprising the sesquiterpene-based metabolite as an active ingredient.

이때, 상기 화장품 조성물은 용액, 현탁액, 에멀전, 페이스트, 겔, 크림, 로션, 파우더, 비누, 계면활성제-함유 클렌징, 오일, 분말 파운데이션, 에멀전 파운데이션, 왁스 파운데이션 또는 스프레이의 형태로 제형화 될 수 있다.The cosmetic composition can be formulated in the form of solutions, suspensions, emulsions, pastes, gels, creams, lotions, powders, soaps, surfactant-containing cleansing, oils, powder foundations, emulsion foundations, wax foundations or sprays. .

또한 본 발명은 상기 세스퀴테르펜 계열 대사체를 유효성분으로 함유하는 식품 조성물을 제공한다.The present invention also provides a food composition containing the sesquiterpene-based metabolite as an active ingredient.

이때, 상기 식품 조성물은 정제, 과립제, 환제, 캅셀제, 액제 또는 산제의 형태로 제형화 될 수 있다.At this time, the food composition may be formulated in the form of tablets, granules, pills, capsules, solutions or powders.

또한 본 발명의 염증성 질환은 뇌염, 골관절염, 류마티스 관절염, 통풍, 강직성 척추염, 건염, 건막염, 류마티스 열, 루프스, 섬유근통(Fibromyalgia), 건선 관절염, 천식, 아토피, 크론병 또는 궤양성 대장염일 수 있으며, 바람직하게는 뇌염일 수 있다.Inflammatory diseases of the present invention may also be encephalitis, osteoarthritis, rheumatoid arthritis, gout, ankylosing spondylitis, tendonitis, tendonitis, rheumatic fever, lupus, fibromyalgia, psoriatic arthritis, asthma, atopy, Crohn's disease or ulcerative colitis , Preferably encephalitis.

본 발명의 세스퀴테르펜 계열 대사체를 유효성분으로 포함하는 조성물은 NO 또는 PGE2의 생성, iNOS 또는 COX-2 단백질 발현과 IL-1β, IL-6 또는 TNF-α의 발현을 억제하여 우수한 항염증 효과를 갖는다.The composition comprising a sesquiterpene-based metabolite of the present invention as an active ingredient is excellent in inhibiting the production of NO or PGE 2 , iNOS or COX-2 protein expression, and IL-1β, IL-6 or TNF-α expression. Has an inflammatory effect.

특히, 미세아교세포에서 우수한 항염증 효과를 나타내는 것으로, 뇌염의 개선, 치료 및 예방에 우수한 효과를 갖는다.In particular, it exhibits an excellent anti-inflammatory effect in microglia, and has an excellent effect on the improvement, treatment and prevention of encephalitis.

따라서 본 발명의 세스퀴테르펜 계열 대사체를 포함하는 항염증 조성물을 유효성분으로 하여 친환경적이며 인체 친화적인 의약품, 화장품 및 식품을 개발할 수 있다.Therefore, by using the anti-inflammatory composition comprising the sesquiterpene-based metabolite of the present invention as an active ingredient, it is possible to develop eco-friendly and human-friendly medicines, cosmetics and foods.

도 1은 감송향 메탄올 추출물의 에틸아세테이트 분획물로부터 세스퀴테르펜 계열 대사체를 분리하는 방법을 나타낸 것이다.
도 2는 신규 화합물 1과 2의 NMR 데이터이다.
도 3은 화합물 1의 NOESY 스펙트럼 데이터 해석과 Mosher's 방법에 의해 생성된 화합물 1a와 1b 의 차를 계산하여 화합물 1의 입체구조를 확인한 것이다.
도 4는 세스퀴테르펜 계열 대사체 9종에 대하여 미세아교세포에서 NO 억제 효과를 IC50 값으로 나타낸 것이다.
도 5는 화합물 3(a) 및 4(b)에 대하여 primary 미세아교세포에서의 NO 생성 억제효과를 나타낸 것이다.
도 6은 화합물 3 및 4에 대하여 미세아교세포 (a 및 b)와 primary 미세아교세포 (c 및 d)에서 염증성 사이토카인인 PGE2의 생성 억제 효과를 나타낸 것이다.
도 7은 화합물 3 및 4의 iNOS 와 COX-2 단백질 발현량 억제 효과를 나타낸 것이다.
도 8은 화합물 3에 대하여 미세아교세포와 primary 미세아교세포에서 염증성 사이토카인인 IL-1β (a 및 d), IL-6 (b 및 e) 및 TNF-α (c 및 f)의 생성 억제 효과를 나타낸 것이다.
도 9는 화합물 4에 대하여 미세아교세포와 primary 미세아교세포에서 염증성 사이토카인인 IL-1β (a 및 d), IL-6 (b 및 e) 및 TNF-α (c 및 f)의 생성 억제 효과를 나타낸 것이다.
FIG. 1 shows a method for separating sesquiterpene-based metabolites from ethyl acetate fractions of methanol extract of Gamsong-hyang.
2 is NMR data of novel compounds 1 and 2. FIG.
FIG. 3 confirms the steric structure of Compound 1 by calculating the difference between Compounds 1a and 1b generated by NOESY spectral data analysis and Mosher's method.
Figure 4 shows the inhibitory effect of NO in microglia cells with IC 50 values for 9 sesquiterpene family metabolites.
Figure 5 shows the effect of inhibiting NO production in primary microglia for compounds 3 (a) and 4 (b).
Figure 6 shows the effect of inhibiting the production of inflammatory cytokine PGE 2 in microglia (a and b) and primary microglia (c and d) for the compounds 3 and 4.
Figure 7 shows the effect of inhibiting the expression of iNOS and COX-2 protein of compounds 3 and 4.
FIG. 8 shows the inhibitory effect of the production of inflammatory cytokines IL-1β (a and d), IL-6 (b and e) and TNF-α (c and f) in microglial cells and primary microglial cells. It is shown.
Figure 9 shows the inhibitory effect of the production of inflammatory cytokines IL-1β (a and d), IL-6 (b and e) and TNF-α (c and f) in microglia and primary microglia for compound 4 It is shown.

이하, 실시예를 통하여 본 발명을 더욱 구체적으로 설명한다. 그러나 이들 실시예는 본 발명에 대한 이해를 돕기 위한 예시의 목적으로 제공된 것일 뿐, 본 발명의 범위가 하기 실시예에 의해 제한되는 것은 아니다.Hereinafter, the present invention will be described in more detail with reference to Examples. However, these examples are provided only for the purpose of illustration to help the understanding of the present invention, the scope of the present invention is not limited by the following examples.

[실시예1]Example 1

감송향 에틸아세테이트 분획물의 제조Preparation of Persimmon Ethyl Acetate Fraction

감송향 5kg을 메탄올 24L에 넣고 2시간 동안 초음파 추출하여 감송향 메탄올 추출물 663g을 수득하였다. 5 kg of persimmon perfume was added to 24 L of methanol, and ultrasonic extraction was performed for 2 hours, thereby obtaining 663 g of a persimmon extract.

수득한 감송향 메탄올 추출물을 물 8L와 메탄올 1L에 녹인 후 헥산 (18L), 클로로포름(18L), 에틸아세테이트(18L), 부탄올(18L) 순으로 파티션을 진행하여 에틸아세테이트 분획물 21.8g을 얻었다.The obtained methanol extract was dissolved in 8 L of water and 1 L of methanol, and partitioned in the order of hexane (18 L), chloroform (18 L), ethyl acetate (18 L) and butanol (18 L) to obtain 21.8 g of ethyl acetate fraction.

[실시예2]Example 2

감송향 에틸아세테이트 분획물로부터 9종 세스퀴테르펜 계열 대사체의 분리Isolation of Nine Sesquiterpene-Based Metabolites from Persimmon Ethyl Acetate Fraction

도 1에 기재된 방법과 같이, 실시예 1에서 얻어진 감송향 에틸아세테이트 분획물로부터 9종의 세스퀴테르펜을 분리하였다.As in the method described in Fig. 1, nine sesquiterpenes were separated from the sweetened ethyl acetate fraction obtained in Example 1.

1 단계Stage 1

실시예 1에서 얻어진 에틸아세테이트 분획물을 실리카겔 (silica gel) 컬럼 크로마토그래피를 이용하여 헥산 에틸아세테이트 혼합액(헥산:에틸아세테이트 = 3.5:1 - 0:1, v/v)을 추출 용매로 하여 13가지 분획물을 얻었다. The ethyl acetate fraction obtained in Example 1 was subjected to silica gel column chromatography using hexane ethyl acetate mixed solution (hexane: ethyl acetate = 3.5: 1-0: 1, v / v) as 13 extraction fractions. Got.

2 단계2 steps

상기 1단계의 13가지 분획물 중 3번째 분획물 [NJ(E)-S3]을 실리카겔 컬럼 크로마토그래피를 이용하여 헥산 에틸아세테이트 혼합액(헥산:에틸아세테이트 = 3.5:1 - 0:1, v/v)을 추출 용매로 하여 9가지 분획물을 얻었다.The hexane ethyl acetate mixed solution (hexane: ethyl acetate = 3.5: 1-0: 1, v / v) of the third fraction [NJ (E) -S3] of the 13 fractions of step 1 was subjected to silica gel column chromatography. Nine fractions were obtained as extraction solvents.

3 단계3 steps

상기 1단계의 13가지 분획물 중 4 및 5번째 분획물 [NJ(E)-S(45)]을 C18 컬럼 크로마토그래피를 이용하여 물 메탄올 혼합액(물:메탄올 = 9:1 - 0:1, v/v)을 추출 용매로 하여 15가지 분획물을 얻었다. The 4th and 5th fractions [NJ (E) -S (45)] of the 13 fractions of the above step 1 were purified using C18 column chromatography (water: methanol = 9: 1-0: 1, v / 15 fractions were obtained using v) as the extraction solvent.

4단계4 steps

상기 3단계의 15가지 분획물 중 3번째 분획물 [NJ(E)-S(45)-3]을 실리카겔 컬럼 크로마토그래피를 이용하여 디클로로메테인 아세톤 혼합액(디클로로메테인: 아세톤 = 5:1 - 0:1, v/v)을 추출 용매로 하여 6가지 분획물을 얻었다.The third fraction [NJ (E) -S (45) -3] of the fifteen fractions of step 3 was subjected to silica gel column chromatography using dichloromethane acetone mixture (dichloromethane: acetone = 5: 1-0: 6 fractions were obtained using 1, v / v) as the extraction solvent.

5단계5 steps

상기 3단계의 15가지 분획물 중 6번째 분획물 [NJ(E)-S(45)-6]을 실리카겔 컬럼 크로마토그래피를 이용하여 헥산 에틸아세테이트 혼합액(헥산:에틸아세테이트 = 1:1 - 0:1, v/v)을 추출 용매로 하여 7가지 분획물을 얻었다.Sixth fraction [NJ (E) -S (45) -6] of the fifteen fractions of the third step was prepared using hexane ethyl acetate mixed solution (hexane: ethyl acetate = 1: 1-0: 1, using silica gel column chromatography). 7 fractions were obtained using v / v) as the extraction solvent.

6단계6 steps

상기 3단계의 15가지 분획물 중 7번째 분획물 [NJ(E)-S(45)-7]을 실리카겔 컬럼 크로마토그래피를 이용하여 헥산 에틸아세테이트 혼합액(헥산:에틸아세테이트 = 3:1 - 0:1, v/v)을 추출 용매로 하여 15가지 분획물을 얻었다. The 7th fraction [NJ (E) -S (45) -7] of the 15 fractions of step 3 was subjected to silica gel column chromatography using hexane ethyl acetate mixture (hexane: ethyl acetate = 3: 1-0: 1, 15 fractions were obtained using v / v) as the extraction solvent.

7단계7 steps

상기 3단계의 15가지 분획물 중 9번째 분획물 [NJ(E)-S(45)-9]을 C18 컬럼 크로마토그래피를 이용하여 물 메탄올 혼합액(물:메탄올 = 6:4 - 4:6, v/v)을 추출 용매로 하여 4가지 분획물을 얻었다. The ninth fraction [NJ (E) -S (45) -9] of the fifteen fractions in step 3 was subjected to C18 column chromatography using a mixture of water methanol (water: methanol = 6: 4-4: 6, v / Four fractions were obtained using v) as the extraction solvent.

화합물 1의 분리Isolation of Compound 1

상기 7단계의 4가지 분획물 중 2 번째 분획물 [NJ(E)-S(45)-9-2]을 실리카겔 컬럼 크로마토그래피를 이용하여 클로로포름 에틸아세테이트 혼합액(클로로포름:에틸아세테이트 = 1.5:1 - 0:1, v/v)을 추출 용매로 하여 8가지 분획물을 얻었다. 그 중 4 번째 분획물 [NJ(E)-S(45)-92-4]을 실리카겔 컬럼 크로마토그래피를 이용하여 헥산 아세톤 혼합액(헥산:아세톤 = 7:3 - 1:1, v/v)을 추출 용매로 하여 화합물 1 (6.6 mg)을 분리하였다. The second fraction of the four fractions of step 7 [NJ (E) -S (45) -9-2] was subjected to silica gel column chromatography using chloroform ethyl acetate mixed solution (chloroform: ethyl acetate = 1.5: 1-0: 8 fractions were obtained using 1, v / v) as the extraction solvent. The fourth fraction [NJ (E) -S (45) -92-4] was extracted using hexane acetone (hexane: acetone = 7: 3-1: 1, v / v) using silica gel column chromatography. Compound 1 (6.6 mg) was isolated as a solvent.

화합물 2의 분리Isolation of Compound 2

상기 6단계의 15가지 분획물 중 5 번째 분획물 [NJ(E)-S(45)-7-5]을 물 메탄올 용매 기울기 조건[물(0.1% 포름산):메탄올 = 65:35 - 40:60, v/v]으로 30분간 역상 HPLC를 실시하여 화합물 2 (5.2 mg)를 분리하였다. The fifth fraction [NJ (E) -S (45) -7-5] of the fifteen fractions in step 6 was replaced with water methanol solvent gradient conditions [water (0.1% formic acid): methanol = 65: 35-40: 60, v / v] was subjected to reverse phase HPLC for 30 minutes to separate compound 2 (5.2 mg).

화합물 3의 분리Isolation of Compound 3

상기 2단계의 9가지 분획물 중 3, 4 및 5 번째 분획물[NJ(E)-S3-A]을 C18 컬럼 크로마토그래피를 물 메탄올 혼합액(물:메탄올 = 8:2 - 0:1, v/v)을 추출 용매로 하여 9가지 분획물을 얻었으며, 그 중 5번째 분획물[NJ(E)-S3A-5]을 물 메탄올 용매 기울기 조건[물(0.1% 포름산):메탄올 = 45:55 - 35:65, v/v]으로 18분간 역상 HPLC를 실시하여 화합물 3 (12 mg)을 분리하였다.  The 3, 4 and 5th fractions [NJ (E) -S3-A] of the 9 fractions of step 2 were subjected to C18 column chromatography using a water methanol mixture (water: methanol = 8: 2-0: 1, v / v). Nine fractions were obtained as the extraction solvent, and the fifth fraction [NJ (E) -S3A-5] was subjected to the water methanol solvent gradient condition [water (0.1% formic acid): methanol = 45:55-35: 65, v / v] was subjected to reverse phase HPLC for 18 minutes to separate compound 3 (12 mg).

화합물 4의 분리Isolation of Compound 4

상기 6단계의 15가지 분획물 중 5 번째 분획물 [NJ(E)-S(45)-7-5]을 물 메탄올 용매 기울기 조건[물(0.1% 포름산):메탄올 = 65:35 - 40:60, v/v]으로 30분간 역상 HPLC를 실시하여 화합물 4 (8.2 mg)를 분리하였다. The fifth fraction [NJ (E) -S (45) -7-5] of the fifteen fractions in step 6 was replaced with water methanol solvent gradient conditions [water (0.1% formic acid): methanol = 65: 35-40: 60, v / v] was subjected to reverse phase HPLC for 30 minutes to separate compound 4 (8.2 mg).

화합물 5의 분리Isolation of Compound 5

상기 7단계의 4가지 분획물 중 3번째 분획물 [NJ(E)-S(45)-9-3]을 실리카겔 컬럼 크로마토그래피를 이용하여 클로로포름 에틸아세테이트 혼합액(클로로포름:에틸아세테이트 = 2:1 - 0:1, v/v)을 추출 용매로 하여 8가지 분획물을 얻었다. 그 중 7 번째 분획물 [NJ(E)-S(45)-93-7]을 C18 컬럼 크로마토그래피를 이용하여 물 메탄올 혼합액(물:메탄올 = 65:35, v/v)을 추출 용매로 하여 화합물 5 (2.8 mg)를 분리하였다.The third fraction [NJ (E) -S (45) -9-3] of the four fractions of step 7 was subjected to silica gel column chromatography using chloroform ethyl acetate mixed solution (chloroform: ethyl acetate = 2: 1-0: 8 fractions were obtained using 1, v / v) as the extraction solvent. The seventh fraction [NJ (E) -S (45) -93-7] was extracted using C18 column chromatography using a mixture of water methanol (water: methanol = 65:35, v / v) as an extraction solvent. 5 (2.8 mg) was isolated.

화합물 6의 분리Isolation of Compound 6

상기 5단계의 7가지 분획물 중 4 및 5번째 분획물 [NJ(E)-S6-(45)]을 실리카겔 컬럼 크로마토그래피를 이용하여 헥산 아세톤 혼합액(헥산:아세톤 = 5:2, v/v)을 추출 용매로 하여 화합물 6 (25 mg)을 분리하였다. Fourth and fifth fractions [NJ (E) -S6- (45)] of the seven fractions of step 5 were subjected to hexane acetone mixture (hexane: acetone = 5: 2, v / v) using silica gel column chromatography. Compound 6 (25 mg) was isolated as an extraction solvent.

화합물 7의 분리Isolation of Compound 7

상기 7단계의 4가지 분획물 중 3번째 분획물 [NJ(E)-S(45)-9-3]을 실리카겔 컬럼 크로마토그래피를 이용하여 클로로포름 에틸아세테이트 혼합액(클로로포름:에틸아세테이트 = 2:1 - 0:1, v/v)을 추출 용매로 하여 8가지 분획물을 얻었다. 그 중 3 번째 분획물 [NJ(E)-S(45)-93-3]을 prep 실리카겔 TLC를 이용하여 헥산 아세톤 혼합액(헥산:아세톤 = 1.8, v/v)을 추출 용매로 하여 화합물 7 (3.5 mg)을 분리하였다. The third fraction [NJ (E) -S (45) -9-3] of the four fractions of step 7 was subjected to silica gel column chromatography using chloroform ethyl acetate mixed solution (chloroform: ethyl acetate = 2: 1-0: 8 fractions were obtained using 1, v / v) as the extraction solvent. The third fraction [NJ (E) -S (45) -93-3] was purified using prep silica gel TLC and extracted with hexane acetone (hexane: acetone = 1.8, v / v) as a solvent. mg) was isolated.

화합물 8의 분리Isolation of Compound 8

상기 3단계의 15가지 분획물 중 5번째 분획물 [NJ(E)-S(45)-5]을 실리카겔 컬럼 크로마토그래피를 이용하여 헥산 에틸아세테이트 혼합액(헥산:에틸아세테이트 = 2:1 - 0:1, v/v)을 추출 용매로 하여 화합물 8 (11.2 mg)을 분리하였다. The fifth fraction [NJ (E) -S (45) -5] of the fifteen fractions of step 3 was subjected to silica gel column chromatography using hexane ethyl acetate mixed solution (hexane: ethyl acetate = 2: 1-0: 1, v / v) was used as an extraction solvent to separate compound 8 (11.2 mg).

화합물 9의 분리Isolation of Compound 9

상기 4 단계의 6가지 분획물 중 첫 번째 분획물 [NJ(E)-S(45)-3-1]을 물 메탄올 용매 기울기 조건[물(0.1% 포름산):메탄올 = 70:30 - 50:50, v/v]으로 30분간 역상 HPLC를 실시하여 화합물 9 (6.9 mg)를 분리하였다.The first fraction [NJ (E) -S (45) -3-1] of the six fractions of step 4 was subjected to water methanol solvent gradient conditions [water (0.1% formic acid): methanol = 70: 30-50: 50, v / v] was subjected to reverse phase HPLC for 30 minutes to separate compound 9 (6.9 mg).

[실시예 3]Example 3

분리된 세스퀴테르펜 계열 대사체의 구조 확인Confirmation of structure of isolated sesquiterpene metabolites

화합물 1과 2의 동정 과정에서, 이들은 이전에 보고되지 않은 신규 물질임을 NMR 데이터와 Mass 데이터를 통하여 확인하였고, Moser's 방법을 이용하여 구조를 규명하였고, 그 결과를 도 2 및 도 3에 나타내었다. 화합물 1과 2는 칸숀 J(kanshone J) 와 칸숀 K(kanshone K) 로 명명하였다.In the process of identifying compounds 1 and 2, they were identified by NMR data and Mass data that were not previously reported, and their structures were identified using Moser's method, and the results are shown in FIGS. 2 and 3. Compounds 1 and 2 were named kanshone J and kanshone K.

화합물 1 내지 9를 1H-NMR, 13C-NMR 및 Mass 데이터로 확인한 결과를 하기 표 1에 나타내었다.Compounds 1 to 9 were identified by the 1 H-NMR, 13 C-NMR and Mass data are shown in Table 1 below.

화합물compound
번호number
명명denomination 화학식Chemical formula 측정값Measures
1One 칸숀 J
(kanshone J)
Kanshon J
(kanshone J)

Figure pat00009
Figure pat00009
brownish oil;
Figure pat00010
-98.3 (c 0.3, MeOH); HRESIMS m/z 273.1463 [M+Na]+, calcd. for C15H22O3Na, 273.1467.
brownish oil;
Figure pat00010
-98.3 (c 0.3, MeOH); HRESIMS m / z 273.1463 [M + Na] + , calcd. for C 15 H 22 O 3 Na, 273.1467.
1a1a MTPA S esterMTPA S ester
Figure pat00011
Figure pat00011
1H NMR (400 MHz, MeOH-d 4 ) data, δ7.14(H-7), 6.82 (H-1), 6.16 (H-8), 5.59 (H-2), 3.01 (H-4), 2.68 (H-6), 1.86 (H-3), 1.78 (H-3), 1.10 (H-12), 1.04 (H-14), 1.01 (H-15), 0.75 (H-13). 1 H NMR (400 MHz, MeOH- d 4 ) data, δ 7.14 (H-7), 6.82 (H-1), 6.16 (H-8), 5.59 (H-2), 3.01 (H-4) , 2.68 (H-6), 1.86 (H-3), 1.78 (H-3), 1.10 (H-12), 1.04 (H-14), 1.01 (H-15), 0.75 (H-13).
1b1b MTPA R esterMTPA R ester
Figure pat00012
Figure pat00012
1H NMR (400 MHz, MeOH-d 4 ) data, δ7.17(H-7), 6.86 (H-1), 6.19 (H-8), 5.61 (H-2), 2.84 (H-4), 2.70 (H-6), 1.81 (H-3), 1.67 (H-3), 1.12 (H-12), 1.05 (H-14), 1.00 (H-13), 0.95 (H-15). 1 H NMR (400 MHz, MeOH- d 4 ) data, δ 7.17 (H-7), 6.86 (H-1), 6.19 (H-8), 5.61 (H-2), 2.84 (H-4) , 2.70 (H-6), 1.81 (H-3), 1.67 (H-3), 1.12 (H-12), 1.05 (H-14), 1.00 (H-13), 0.95 (H-15).
22 칸숀 K(kanshone K)Kanshone K
Figure pat00013
Figure pat00013
yellow oil;
Figure pat00014
-76.2 (c 0.3, MeOH); HRESIMS m/z 273.1457 [M+Na]+, calcd. for C15H22O3Na, 273.1467.
yellow oil;
Figure pat00014
-76.2 (c 0.3, MeOH); HRESIMS m / z 273.1457 [M + Na] + , calcd. for C 15 H 22 O 3 Na, 273.1467.
33 데옥소-나치놀 A(desoxo-narchinol A)Desoxo-narchinol A
Figure pat00015
Figure pat00015
1H NMR (400 MHz, chloroform-d) δ7.07 (H-7), 7.05 (H-1), 6.16 (H-8), 4.07 (H-6), 2.33 (H-4), 2.28 (H-2), 1.56 (H-3), 0.97 (H-11), 0.96 (H-12); 13C NMR (100 MHz, chloroform-d) δ187.5 (C-9), 146.8 (C-8), 140.8 (C-7), 138.5 (C-10), 130.5 (C-1), 68.4 (C-6), 42.5 (C-5), 30.9 (C-4), 26.4 (C-2), 25.6 (C-3), 19.8 (C-11), 15.0 (C-12); HRESIMS m/z 193.1224 [M+H]+, calcd for C12H17O2, 193.1229. 1 H NMR (400 MHz, chloroform- d ) δ7.07 (H-7), 7.05 (H-1), 6.16 (H-8), 4.07 (H-6), 2.33 (H-4), 2.28 ( H-2), 1.56 (H-3), 0.97 (H-11), 0.96 (H-12); 13 C NMR (100 MHz, chloroform- d ) δ 187.5 (C-9), 146.8 (C-8), 140.8 (C-7), 138.5 (C-10), 130.5 (C-1), 68.4 ( C-6), 42.5 (C-5), 30.9 (C-4), 26.4 (C-2), 25.6 (C-3), 19.8 (C-11), 15.0 (C-12); HRESIMS m / z 193.1224 [M + H] + , calcd for C 12 H 17 O 2 , 193.1229.
44 나치놀 B(narchinol B)Narchinol B
Figure pat00016
Figure pat00016
1H NMR (400 MHz, methanol-d 4 ) δ7.14 (H-7), 6.86 (H-1), 6.14 (H-8), 4.21 (H-2), 4.08 (H-6), 2.62 (H-4), 1.74 (H-3), 1.66 (H-3), 0.96 (H-12), 0.91 (H-11); 13C NMR (100 MHz, methanol-d 4 ) δ189.6 (C-9), 149.9 (C-7), 142.7 (C-10), 137.7 (C-1), 130.5 (C-8), 68.6 (C-6), 64.1 (C-2), 43.9 (C-5), 36.1 (C-3), 26.7 (C-4), 18.3 (C-11), 15.0 (C-12); HRESIMS m/z 231.0991 [M+Na]+, calcd for C12H16O3Na, 231.0997. 1 H NMR (400 MHz, methanol- d 4 ) δ7.14 (H-7), 6.86 (H-1), 6.14 (H-8), 4.21 (H-2), 4.08 (H-6), 2.62 (H-4), 1.74 (H-3), 1.66 (H-3), 0.96 (H-12), 0.91 (H-11); 13 C NMR (100 MHz, methanol- d 4 ) δ 189.6 (C-9), 149.9 (C-7), 142.7 (C-10), 137.7 (C-1), 130.5 (C-8), 68.6 (C-6), 64.1 (C-2), 43.9 (C-5), 36.1 (C-3), 26.7 (C-4), 18.3 (C-11), 15.0 (C-12); HRESIMS m / z 231.0991 [M + Na] + , calcd for C 12 H 16 O 3 Na, 231.0997.
55 불라탄트리올(bullatantriol)Bullatantriol
Figure pat00017
Figure pat00017
1H NMR (400 MHz, pyridine-d 5 ) δ3.75 (H-9), 2.85 (H-4), 2.45 (H-8), 2.41 (H-3 and 10), 1.99 (H-2), 1.96 (H-8), 1.91 (H-7), 1.66 (H-3), 1.64 (H-1 and 14), 1.62 (H-7), 1.60 (H-10), 1.59 (H-15), 1.55 (H-2), 1.45 (H-12 and 13), 1.13 (H-5); 13C NMR (100 MHz, pyridine-d 5 ) δ79.7 (C-9), 71.3 (C-6), 70.6 (C-11), 59.8 (C-5), 52.3 (C-10), 48.2 (C-1), 42.4 (C-7), 40.2 (C-2), 33.2 (C-3 and 14), 32.6 (C-15), 32.4 (C-4), 31.0 (C-12), 30.7 (C-13), 29.4 (C-8); HRESIMS m/z 279.1935 [M+Na]+, calcd for C15H28O3Na, 279.1936. 1 H NMR (400 MHz, pyridine- d 5 ) δ 3.75 (H-9), 2.85 (H-4), 2.45 (H-8), 2.41 (H-3 and 10), 1.99 (H-2) , 1.96 (H-8), 1.91 (H-7), 1.66 (H-3), 1.64 (H-1 and 14), 1.62 (H-7), 1.60 (H-10), 1.59 (H-15) ), 1.55 (H-2), 1.45 (H-12 and 13), 1.13 (H-5); 13 C NMR (100 MHz, pyridine- d 5 ) δ 77.9 (C-9), 71.3 (C-6), 70.6 (C-11), 59.8 (C-5), 52.3 (C-10), 48.2 (C-1), 42.4 (C-7), 40.2 (C-2), 33.2 (C-3 and 14), 32.6 (C-15), 32.4 (C-4), 31.0 (C-12), 30.7 (C-13), 29.4 (C-8); HRESIMS m / z 279.1935 [M + Na] + , calcd for C 15 H 28 O 3 Na, 279.1936.
66 1ββαα-트리히드록시엔데스만(1ββαα-trihydroxyeudesmane)1ββαα-trihydroxyeudesmane
Figure pat00018
Figure pat00018
1H NMR (400 MHz, pyridine-d 5 ) δ3.72 (H-1), 2.58 (H-2), 2.29 (H-9), 2.12 (H-6), 2.01 (H-9), 2.00 (H-6), 1.97 (H-3), 1.93 (H-2), 1.80 (H-5 and 11), 1.79 (H-8), 1.68 (H-3), 1.65 (H-14), 1.37 (H-15), 1.13 (H-12 and 13); 13C NMR (100 MHz, pyridine-d 5 ) δ79.6 (C-1), 73.1 (C-7), 70.8 (C-4), 45.8 (C-5), 41.1 (C-3), 40.13 (C-11), 40.10 (C-10), 35.7 (C-9), 30.7 (C-6), 30.5 (C-15), 29.7 (C-8), 28.4 (C-2), 17.6 (C-13), 17.5 (C-12), 12.6 (C-14); HRESIMS m/z 279.1931 [M+Na]+, calcd for C15H28O3Na, 279.1936. 1 H NMR (400 MHz, pyridine- d 5 ) δ3.72 (H-1), 2.58 (H-2), 2.29 (H-9), 2.12 (H-6), 2.01 (H-9), 2.00 (H-6), 1.97 (H-3), 1.93 (H-2), 1.80 (H-5 and 11), 1.79 (H-8), 1.68 (H-3), 1.65 (H-14), 1.37 (H-15), 1.13 (H-12 and 13); 13 C NMR (100 MHz, pyridine- d 5 ) δ79.6 (C-1), 73.1 (C-7), 70.8 (C-4), 45.8 (C-5), 41.1 (C-3), 40.13 (C-11), 40.10 (C-10), 35.7 (C-9), 30.7 (C-6), 30.5 (C-15), 29.7 (C-8), 28.4 (C-2), 17.6 ( C-13), 17.5 (C-12), 12.6 (C-14); HRESIMS m / z 279.1931 [M + Na] + , calcd for C 15 H 28 O 3 Na, 279.1936.
77 테클라트리올(teuclatriol)Teclalatriol
Figure pat00019
Figure pat00019
1H NMR (400 MHz, pyridine-d 5 ) δ4.46 (H-6), 2.29 (H-2), 2.26 (H-1), 2.25 (H-5), 2.23 (H-9), 2.05 (H-11), 2.04 (H-3), 1.82 (H-2 and 3), 1.81 (H-9), 1.79 (H-8), 1.52 (H-8), 1.49 (H-15), 1.44 (H-14), 1.33 (H-7), 1.22 (H-12), 1.11 (H-13); 13C NMR (100 MHz, pyridine-d 5 ) δ80.9 (C-4), 74.4 (C-10), 71.5 (C-6), 56.2 (C-5), 52.6 (C-7), 49.1 (C-9), 46.5 (C-1), 42.0 (C-3), 30.6 (C-11), 24.2 (C-2), 23.6 (C-15), 22.6 (C-14), 22.0 (C-13), 21.5 (C-8), 21.4 (C-12); HRESIMS m/z 279.1936 [M+Na]+, calcd for C15H28O3Na, 279.1936. 1 H NMR (400 MHz, pyridine- d 5 ) δ4.46 (H-6), 2.29 (H-2), 2.26 (H-1), 2.25 (H-5), 2.23 (H-9), 2.05 (H-11), 2.04 (H-3), 1.82 (H-2 and 3), 1.81 (H-9), 1.79 (H-8), 1.52 (H-8), 1.49 (H-15), 1.44 (H-14), 1.33 (H-7), 1.22 (H-12), 1.11 (H-13); 13 C NMR (100 MHz, pyridine- d 5 ) δ 80.9 (C-4), 74.4 (C-10), 71.5 (C-6), 56.2 (C-5), 52.6 (C-7), 49.1 (C-9), 46.5 (C-1), 42.0 (C-3), 30.6 (C-11), 24.2 (C-2), 23.6 (C-15), 22.6 (C-14), 22.0 ( C-13), 21.5 (C-8), 21.4 (C-12); HRESIMS m / z 279.1936 [M + Na] + , calcd for C 15 H 28 O 3 Na, 279.1936.
88 4β히드록시-8β메톡시-10-메틸렌-2,9- 다이옥사트리사이클로[4.3.1.03,7]데케인(4ββdioxatricyclo[4.3.1.03,7]decane)4βhydroxy-8βmethoxy-10-methylene-2,9-dioxatricyclo [4.3.1.03,7] decane (4ββdioxatricyclo [4.3.1.03,7] decane)
Figure pat00020
Figure pat00020
1H NMR (400 MHz, chloroform-d) δ5.01 (H-3), 4.97 (H-1), 4.84 (H-11), 4.77 (H-11), 3.91 (H-7), 3.39 (1-OCH3), 3.09 (H-5), 2.28 (H-9), 2.11 (H-6), 1.82 (H-6), 1.38 (H-10); 13C NMR (100 MHz, chloroform-d) δ149.3 (C-4), 107.2 (C-11), 97.2 (C-1), 93.8 (C-3), 82.1 (C-8), 79.2 (C-7), 55.4 (1-OCH3), 43.2 (C-9), 42.9 (C-6), 36.6 (C-5), 19.0 (C-10); HRESIMS m/z 235.0945 [M+Na]+, calcd for C11H16O4Na, 235.0946. 1 H NMR (400 MHz, chloroform- d ) δ5.01 (H-3), 4.97 (H-1), 4.84 (H-11), 4.77 (H-11), 3.91 (H-7), 3.39 ( 1-OCH3), 3.09 (H-5), 2.28 (H-9), 2.11 (H-6), 1.82 (H-6), 1.38 (H-10); 13 C NMR (100 MHz, chloroform- d ) δ 149.3 (C-4), 107.2 (C-11), 97.2 (C-1), 93.8 (C-3), 82.1 (C-8), 79.2 ( C-7), 55.4 (1-OCH3), 43.2 (C-9), 42.9 (C-6), 36.6 (C-5), 19.0 (C-10); HRESIMS m / z 235.0945 [M + Na] + , calcd for C 11 H 16 O 4 Na, 235.0946.
99 야타마닌 A(jatamanin A)Yatamanin A
Figure pat00021
Figure pat00021
1H NMR (400 MHz, methanol-d 4 ) δ5.09 (H-3 and 11), 5.05 (H-11), 4.41 (H-3), 3.74 (H-7), 3.33 (H-5), 2.98 (H-9), 2.16 (H-6), 1.97 (H-6), 1.46 (H-10); 13C NMR (100 MHz, methanol-d 4 ) δ174.9 (C-1), 144.2 (C-4), 113.6 (C-11), 86.1 (C-8), 81.4 (C-7), 71.0 (C-3), 53.6 (C-9), 41.0 (C-6), 40.7 (C-5), 21.9 (C-10); HRESIMS m/z 221.0789 [M+Na]+, calcd for C10H14O4Na, 221.0790. 1 H NMR (400 MHz, methanol- d 4 ) δ 5.09 (H-3 and 11), 5.05 (H-11), 4.41 (H-3), 3.74 (H-7), 3.33 (H-5) , 2.98 (H-9), 2.16 (H-6), 1.97 (H-6), 1.46 (H-10); 13 C NMR (100 MHz, methanol- d 4 ) δ174.9 (C-1), 144.2 (C-4), 113.6 (C-11), 86.1 (C-8), 81.4 (C-7), 71.0 (C-3), 53.6 (C-9), 41.0 (C-6), 40.7 (C-5), 21.9 (C-10); HRESIMS m / z 221.0789 [M + Na] + , calcd for C 10 H 14 O 4 Na, 221.0790.

[실시예 4] Example 4

마우스 유래 미세아교세포인 BV2 세포의 배양Culture of BV2 Cells, a Mouse-derived Microglial Cell

마우스 유래 미세아교세포인 BV2 세포(2 ××105 cells/well)를 10% heat-inactivated FBS, 페니실린 G (penicillin G, 100 IU/ml, Gibco, 15240062) 및 스트렙토마이신(streptomycin, 100 μμg/ml, Gibco, 15240062)을 함유한 DMEM 배지에 분주하고 5% 이산화탄소 배양기(Sanyo, MCO175) 내에서 37 ℃에서 24시간 배양하였다.Mouse-derived microglia BV2 cells (2 × 10 5 cells / well) were treated with 10% heat-inactivated FBS, penicillin G (penicillin G, 100 IU / ml, Gibco, 15240062), and streptomycin (100 μg / ml, Gibco, 15240062) were dispensed in DMEM medium and incubated for 24 hours at 37 ° C. in a 5% carbon dioxide incubator (Sanyo, MCO175).

[실시예 5] Example 5

세스퀴테르펜 계열 대사체의 NO 생성 억제 효과 확인Inhibition of NO Production by Sesquiterpene Metabolites

세스퀴테르펜 계열 대사체의 BV2 세포에서의 염증 매개 인자인 NO 의 생성 억제 효과를 확인하기 위하여 문헌(Kim et al., European Journal of Pharmacology, 721, pp. 267-276(2013))의 방법을 이용하여 실험을 진행 하였다. The method of Kim et al., European Journal of Pharmacology, 721, pp. 267-276 (2013) was used to identify the inhibitory effect of NO, the inflammation mediator in BV2 cells of sesquiterpene family metabolites. The experiment was carried out using.

BV2 세포에 9종의 세스퀴테르펜 계열 대사체를 세포독성이 나타나지 않는 농도로 처리하고 3시간 배양하여 전처리 하였으며, 리포폴리싸카라이드(LPS)로 염증반응을 유발한 다음, 24 시간 동안 5% 이산화탄소 배양기 내에서 배양하였다.Nine sesquiterpene-based metabolites were treated with BV2 cells at a concentration that showed no cytotoxicity and incubated for 3 hours, followed by inflammatory reaction with lipopolysaccharide (LPS), and then 5% carbon dioxide for 24 hours. The culture was carried out in an incubator.

배양 후 NO 생성의 억제를 확인하였으며, 그 결과를 도 4에 나타내었다. 화합물 1 내지 9는 60 μM 농도 내에서 도 4에 나타낸 바와 같이 NO 생성억제효과를 보였으며, 화합물 3 및 4가 가장 우수한 효과를 나타내는 것을 확인하였다. The inhibition of NO production after the culture was confirmed, and the results are shown in FIG. 4. Compounds 1 to 9 showed NO production inhibitory effect as shown in Figure 4 within 60 μM concentration, it was confirmed that the compound 3 and 4 shows the best effect.

[실시예 6] Example 6

primary 미세아교세포에서 화합물 3 및 4의 NO 생성 억제 효과 확인Inhibition of NO Production by Compounds 3 and 4 in Primary Microglial Cells

화합물 3 및 4에 대하여 BV2 세포와 primary 미세아교세포에서 염증 매개 인자들의 생성과 iNOS 및 COX-2의 단백질 발현 억제를 확인하기 위하여 문헌(Kim et al., European Journal of Pharmacology, 721, pp. 267-276(2013))의 방법을 이용하여 실험을 진행하였다.To confirm the production of inflammatory mediators and inhibition of protein expression of iNOS and COX-2 in BV2 cells and primary microglia for compounds 3 and 4 (Kim et al., European Journal of Pharmacology, 721, pp. 267). -276 (2013)) was conducted using the method.

화합물 3 및 4가 primary 미세아교세포에서 항염증효과를 나타내는지를 다음과 같이 확인하였다. primary 미세아교세포는 생후 1일령 랫트의 대뇌피질을 분리하여 75cm2 티플라스크에 2주간 배양한 뒤, 세포 여과기를 통하여 미세아교세포만을 분리하였다. It was confirmed whether the compounds 3 and 4 show an anti-inflammatory effect in the primary microglia cells. Primary microglia cells were isolated from the cerebral cortex of 1-day-old rats and cultured in a 75cm 2 flask for 2 weeks, and only microglia were isolated through a cell filter.

그 후, B27 첨가물 (Thermo, 17504044)과 함께, 10% heat-inactivated FBS, 페니실린 G와 스트렙토마이신을 함유한 DMEM 배지에서 3일간 배양하고, B27 첨가물이 없는 DMEM 배지로 교체하여 3일간 배양한 뒤 실험에 사용하였다. Then, incubated for 3 days in DMEM medium containing 10% heat-inactivated FBS, penicillin G and streptomycin with B27 additive (Thermo, 17504044), and then incubated for 3 days after replacing with DMEM medium without B27 additive. It was used for the experiment.

상기 배양된 primary 미세아교세포에 화합물3 및 4를 1.3, 2.5, 5.0 및 10.0 μM 농도로 3시간 배양하여 전처리한 후, 리포폴리싸카라이드(LPS)로 염증반응을 유발한 다음 24 시간 동안 5% 이산화탄소 배양기 내에서 배양하였다. 배양 후, NO 생성 억제를 그리스 반응으로 확인하였으며, 그 결과를 도 5에 나타내었다. After pre-treatment of the cultured primary microglial cells 3 and 4 in a concentration of 1.3, 2.5, 5.0 and 10.0 μM for 3 hours, the inflammatory reaction with lipopolysaccharide (LPS) induced 5% for 24 hours The culture was carried out in a carbon dioxide incubator. After incubation, NO production inhibition was confirmed by a grease reaction, and the results are shown in FIG. 5.

도 5A 및 도 5B에 나타낸 바와 같이, 화합물 3 및 4의 농도가 증가함에 따라 NO의 생성이 현저하게 억제되는 것을 확인하였다.As shown in Figures 5A and 5B, it was confirmed that the production of NO is significantly inhibited as the concentration of compounds 3 and 4 increases.

[실시예 7] Example 7

화합물 3 및 4의 PGEPGE of compounds 3 and 4 22 생성 억제 효과 확인 Confirmation of generation suppression

화합물 3 및 4의 함량에 따른 PGE2 생성 억제 효과를 확인하기 위하여, 효소 면역 측정법 (enzyme-linked immunosorbent assay, ELISA) 키트 (R&D System, KGE004B)를 사용하여 수행한 실험 방법은 하기와 같다.In order to confirm the inhibitory effect of PGE2 production according to the content of compounds 3 and 4, the experimental method performed using an enzyme-linked immunosorbent assay (ELISA) kit (R & D System, KGE004B) is as follows.

BV2 세포와 primary 미세아교세포를 60 mm 디쉬에 3 X 106 cells/ml 농도로 12시간 배양한 후, 화합물 3 및 4를 농도별로 (1.3 ~ 10.0 μM) 처리 하고 3시간 동안 배양하는 전처리를 수행한 다음, 리포폴리싸카라이드를 1 및 0.5 μg/ml 각각 처리하고 24시간 동안 5% 이산화탄소 배양기 내에서 염증반응을 유발하였다.After incubating the BV2 cells and the primary microglia for 12 hours at a concentration of 3 X 10 6 cells / ml in a 60 mm dish, the compounds 3 and 4 were treated by concentration (1.3 to 10.0 μM) and pretreated for 3 hours. Then, lipopolysaccharide was treated with 1 and 0.5 μg / ml respectively and induced an inflammatory reaction in a 5% carbon dioxide incubator for 24 hours.

배양 후, 배양액을 모으고, ELISA Kit의 96 웰 플레이트에 150 μL를 넣은 후, 1차 항체 용액 50 μL을 넣고 500 ± 50 rpm 조건으로 1시간 동안 실온에서 반응시켰다. 반응 정지 후, 상기 웰 플레이트에 PGE2 포합 용액 50 μL를 첨가하여 500 ± 50 rpm 조건으로 2시간 동안 실온에서 반응시켰다. 상기 반응 정지 후, 상기 웰 플레이트에 세척 용액 400 μL을 첨가하여 세척하였고, 이를 3번 반복하였다. After incubation, the culture solution was collected, 150 μL was placed in a 96 well plate of an ELISA Kit, and then 50 μL of the primary antibody solution was added and reacted at room temperature for 1 hour at 500 ± 50 rpm. After the reaction was stopped, 50 μL of a PGE 2 conjugated solution was added to the well plate, and reacted at room temperature for 2 hours at 500 ± 50 rpm. After the reaction was stopped, 400 μL of the washing solution was added to the well plate and washed, and this was repeated three times.

이후, 기질 반응 용액 200 μL를 첨가하여 차광된 공간에서 30 분동안 반응시킨 후, 반응 정지 용액 100 μL를 첨가하여 반응을 정지시키고, 마이크로플레이트 리더기를 사용하여 450 nm 파장에서 흡광도를 측정하여 흡광도를 PGE2 농도로 환산하였고, 그 결과를 도 6에 나타내었다.Subsequently, 200 μL of the substrate reaction solution was added to react for 30 minutes in the shaded space, and then 100 μL of the reaction stop solution was added to stop the reaction, and the absorbance was measured by measuring the absorbance at 450 nm using a microplate reader. Converted to PGE 2 concentration, the results are shown in FIG.

도 6에 나타낸 바와 같이, 화합물 3 및 4의 농도가 증가함에 따라, BV2 세포 (도 6A 및 6B) 및 primary 미세아교세포 (도 6C 및 6D)에서 리포폴리싸카라이드에 의한 PGE2의 생성이 농도 의존적으로 억제되는 것을 확인하였다.As shown in FIG. 6, as the concentration of compounds 3 and 4 increases, the production of PGE 2 by lipopolysaccharide in BV2 cells (FIGS. 6A and 6B) and primary microglia (FIGS. 6C and 6D) It was confirmed that it is suppressed dependently.

[실시예 8] Example 8

화합물 3 및 4의 iNOS 및 COX-2 단백질 발현 억제 효과 확인Inhibition of iNOS and COX-2 Protein Expression by Compounds 3 and 4

화합물 3 및 4의 함량에 따른 iNOS 및 COX-2 단백질 발현 억제 효과를 확인하기 위하여, 웨스턴 블럿 분석법(western blot analysis)을 수행한 실험 방법은 다음과 같다.In order to confirm the iNOS and COX-2 protein expression inhibitory effect according to the content of the compounds 3 and 4, the experimental method performed the Western blot analysis is as follows.

먼저, 60 mm 디쉬에 3 X 106 cells/ml 농도로 BV2 세포와 primary 미세아교세포를 12시간 배양하고, 화합물 3 및 4를 농도 별로 처리 후 3시간 동안 배양하는 전처리를 수행한 다음, 리포폴리싸카라이드를 1 및 0.5 μg/ml 각각 처리하고 24시간 동안 5% 이산화탄소 배양기 내에서 염증반응을 유발하였다.First, BV2 cells and primary microglial cells were incubated for 12 hours at a concentration of 3 X 10 6 cells / ml in a 60 mm dish, followed by pretreatment for incubation for 3 hours after treatment of compounds 3 and 4 by concentration, followed by lipopoly The saccharides were treated with 1 and 0.5 μg / ml respectively and induced an inflammatory response in a 5% carbon dioxide incubator for 24 hours.

배양 후, 배양액을 제외한 BV2 세포 및 primary 미세아교세포에 RIPA(89900, Thermo) 버퍼를 첨가하고, 4℃ 및 14,000 × g의 조건에서 25분 동안 원심분리하여, 상등액을 분리하였다. 상기 분리된 상등액에서 단백질 정량은 BSA 단백질 실험 키트(Pierce Biotechnology)를 이용하여 수행하였다.After incubation, RIPA (89900, Thermo) buffer was added to the BV2 cells and the primary microglia except the culture medium, and centrifuged at 4 ° C. and 14,000 × g for 25 minutes to separate the supernatant. Protein quantification in the isolated supernatant was performed using a BSA protein experiment kit (Pierce Biotechnology).

구체적으로, 상기 상등액을 7.5% SDS-폴리아크릴아마이드겔(polyacrylamide gel)를 이용하여 2시간 동안 전기영동한 후, 나이트로셀룰로스 맴브레인(Nitrocellulose membrane, NC membrane)를 이용하여 폴리아크릴아마이드겔로부터 전사하였다. 전사된 나이트로셀룰로스 맴브레인을 5% 무지방유가 포함된 신선한 블로킹 버퍼(blocking buffer, 0.1% Tween 20 in Tris-buffered saline)에서 1시간 동안 반응을 정지시켰다. 반응 정지 후, 상기 멤브레인을 PBST(PBS, 0.1% Tween 20)로 10분에 1회씩 3회 세척한 후에, iNOS(SC-650, Santacruz biotechnology, USA), COX-2(SC-1745, Santacruz biotechnology, USA)의 항체(Ab)를 1:1000으로 희석하여 넣고 3시간 동안 반응을 수행하였다. 반응 후, 상기 PBST로 10분에 1회씩 3회 세척하고, 2차 항체(Anti-rabbit IgG, AP132, Millipore, USA)를 1:1000으로 희석하여 넣고 1시간 동안 반응을 수행하였다.Specifically, the supernatant was electrophoresed for 2 hours using 7.5% SDS-polyacrylamide gel, and then transferred from polyacrylamide gel using a nitrocellulose membrane (NC membrane). . The transferred nitrocellulose membrane was quenched for 1 hour in a fresh blocking buffer containing 0.1% Tween 20 in Tris-buffered saline containing 5% nonfat milk. After stopping the reaction, the membrane was washed three times every 10 minutes with PBST (PBS, 0.1% Tween 20), and then iNOS (SC-650, Santacruz biotechnology, USA), COX-2 (SC-1745, Santacruz biotechnology) , USA) antibody (Ab) was diluted 1: 1000 and the reaction was performed for 3 hours. After the reaction, the plate was washed three times with PBST once every 10 minutes, and the secondary antibody (Anti-rabbit IgG, AP132, Millipore, USA) was diluted 1: 1000 and reacted for 1 hour.

반응 후, PBST로 10분에 1회씩 3회 세척한 다음, ECL(Amersham Pharmacia Biotech) 용액을 1:1로 잘 섞어서 나이트로셀룰로스 맴브레인(Nitrocellulose membrane, NC membrane) 위에 부어서 발광시키고, 암실에서 X선 필름에 감광한 후 현상하였다. 동일한 방법으로 액틴(Actin)에 대한 항체(SC-1616, Santacruz biotechnology, USA)를 이용하여 액틴(Actin)의 함량을 측정하였고, 그 결과를 도 7에 나타내었다.After the reaction, washed three times with PBST once every 10 minutes, and then mixed ECL (Amersham Pharmacia Biotech) solution 1: 1 well, poured onto a nitrocellulose membrane (NC membrane) to emit light, X-ray in the dark room It developed after photosensitive to a film. In the same manner, the content of Actin (Actin) was measured using an antibody to Actin (SC-1616, Santacruz biotechnology, USA), and the results are shown in FIG. 7.

도 7에 나타난 바와 같이, 화합물 3 및 4의 농도가 증가함에 따라 BV2세포와 primary 미세아교세포에서 염증 매개 인자로 알려진 iNOS 및 COX-2의 단백질 발현이 현저하게 억제되는 것을 확인하였다.As shown in Figure 7, as the concentration of compounds 3 and 4 was confirmed that the protein expression of iNOS and COX-2 known as inflammatory mediators in BV2 cells and primary microglia cells is significantly inhibited.

[실시예 9] Example 9

화합물 3 및 4의 사이토카인 생성 억제효과 확인Inhibition of cytokine production of compounds 3 and 4

화합물 3 및 4의 함량에 따른 염증성 사이토카인의 생성 억제 효과를 확인하기 위하여, 효소 면역 측정법 (enzyme-linked immunosorbent assay, ELISA) 키트 (R&D System, MLB00C, M6000B, MTA00B, RLB00, R6000B, RTA00)를 사용하여 수행한 실험 방법은 다음과 같다.Enzyme-linked immunosorbent assay (ELISA) kits (R & D System, MLB00C, M6000B, MTA00B, RLB00, R6000B, RTA00) were used to confirm the inhibitory effect of the production of inflammatory cytokines according to the contents of compounds 3 and 4. The experimental method performed using the following is as follows.

BV2 세포와 primary 미세아교세포를 60 mm 디쉬에 3 X 106 cells/ml 농도로 12시간 배양한 후, 화합물 3과 4를 농도별로 (1.3 ~ 10.0 μM) 처리 하고 3시간 동안 배양하는 전처리를 수행한 다음, 리포폴리싸카라이드를 1 및 0.5 μg/ml 각각 처리하고 24시간 동안 5% 이산화탄소 배양기 내에서 염증반응을 유발하였다. 배양 후, 배양액을 모으고, ELISA Kit의 96 웰 플레이트에 반응 용액 50 μL와 배양액 50 μL를 첨가하여 1시간 동안 상온에서 반응시켰다. 상기 반응 후, 상기 웰 플레이트에 세척 용액 400 μL를 첨가하여 세척하였고, 이를 5회 반복하였다. 이후, 사이토카인 포함 용액을 100 μL 첨가하여 2시간 동안 상온에서 반응시켰다. 상기 반응 후, 세척 용액 400 μL를 첨가하여 세척하였고, 이를 5회 반복하였다. 이후, 기질 반응 용액 100 μL 를 첨가하여 차광된 공간에서 30분 동안 반응시키고, 반응 정지 용액 100 μL를 첨가하여 반응을 정지 시킨 뒤, 마이크로플레이트 리더기를 사용하여 450 nm 파장에서 흡광도를 측정하고, 측정된 흡광도를 농도로 환산하하여, 도 8 및 도 9에 나타내었다.After incubating BV2 cells and primary microglia cells at a concentration of 3 X 10 6 cells / ml in a 60 mm dish for 12 hours, compounds 3 and 4 were treated by concentrations (1.3 to 10.0 μM) and pretreated for 3 hours. Then, lipopolysaccharide was treated with 1 and 0.5 μg / ml respectively and induced an inflammatory reaction in a 5% carbon dioxide incubator for 24 hours. After incubation, the culture solution was collected, and 50 μL of the reaction solution and 50 μL of the culture solution were added to the 96 well plate of the ELISA Kit, followed by reaction at room temperature for 1 hour. After the reaction, 400 μL of the washing solution was added to the well plate and washed, and this was repeated five times. Thereafter, 100 μL of the cytokine-containing solution was added and reacted at room temperature for 2 hours. After the reaction, 400 μL of washing solution was added and washed, which was repeated five times. Then, 100 μL of the substrate reaction solution was added to react for 30 minutes in the shaded space, and 100 μL of the reaction stop solution was added to stop the reaction, and then the absorbance was measured at 450 nm using a microplate reader. The absorbance thus obtained was converted into concentration, and shown in FIGS. 8 and 9.

도 8에 나타난 바와 같이, 화합물 3의 농도가 증가함에 따라 BV2 세포 (도 8A, 8B 및 8C) 및 primary 미세아교세포 (도 8D, 8E 및 8F)에서 리포폴리싸카라이드에 의해 증가한 염증성 싸이토카인인 IL-1β(도 8A 및 8D), IL-6 (도 8B 및 8E) 및 TNF-α (도 8C 및 8F)의 생성이 억제되는 것을 확인하였다.As shown in FIG. 8, IL is an inflammatory cytokine increased by lipopolysaccharide in BV2 cells (FIGS. 8A, 8B and 8C) and primary microglia (FIGS. 8D, 8E and 8F) as the concentration of compound 3 increases. It was confirmed that the production of −1β (FIGS. 8A and 8D), IL-6 (FIGS. 8B and 8E) and TNF-α (FIGS. 8C and 8F) were inhibited.

또한, 도 9에 나타난 바와 같이, 화합물 4의 농도가 증가함에 따라 BV2 세포 (도 9A, 9B 및 9C) 및 primary 미세아교세포 (도 9D, 9E 및 9F)에서 리포폴리싸카라이드에 의해 증가한 염증성 싸이토카인인 IL-1β(도 9A 및 9D), IL-6 (도 9B 및 9E) 및 TNF-α (도 9C 및 9F)의 생성이 억제되는 것을 확인하였다.In addition, as shown in FIG. 9, inflammatory cytokines increased by lipopolysaccharide in BV2 cells (FIGS. 9A, 9B and 9C) and primary microglia (FIGS. 9D, 9E and 9F) as the concentration of compound 4 increased. It was confirmed that the production of phosphorus IL-1β (FIGS. 9A and 9D), IL-6 (FIGS. 9B and 9E) and TNF-α (FIGS. 9C and 9F) were inhibited.

Claims (13)

하기 화학식 1 내지 9의 화합물, 및 이의 약제학적으로 허용가능한 염으로 이루어지는 군으로부터 선택되는 하나 이상의 세스퀴테르펜 계열 대사체를 유효성분으로 함유하는 염증성 질환의 예방 또는 치료용 약학적 조성물.
[화학식 1]
Figure pat00022

(R는 β-OH, 또는 α-메톡시-α-트리플루오로메틸페닐아세테이트기임).
[화학식 2]
Figure pat00023

(R는 α-OH임).
[화학식 3]
Figure pat00024

[화학식 4]
Figure pat00025

[화학식 5]
Figure pat00026

[화학식 6]
Figure pat00027

[화학식 7]
Figure pat00028

[화학식 8]
Figure pat00029

[화학식 9]
Figure pat00030

A pharmaceutical composition for preventing or treating an inflammatory disease containing as an active ingredient at least one sesquiterpene-based metabolite selected from the group consisting of a compound of Formulas 1 to 9, and pharmaceutically acceptable salts thereof.
[Formula 1]
Figure pat00022

(R is a β-OH or α-methoxy-α-trifluoromethylphenylacetate group).
[Formula 2]
Figure pat00023

(R is α-OH).
[Formula 3]
Figure pat00024

[Formula 4]
Figure pat00025

[Formula 5]
Figure pat00026

[Formula 6]
Figure pat00027

[Formula 7]
Figure pat00028

[Formula 8]
Figure pat00029

[Formula 9]
Figure pat00030

제1항에 있어서, 상기 조성물은 화학식 3의 화합물, 화학식 4의 화합물 또는 이의 약제학적으로 허용가능한 염을 유효성분으로 함유하는 것을 특징으로 하는 염증성 질환의 예방 또는 치료용 약학적 조성물.
According to claim 1, wherein the composition is a pharmaceutical composition for the prevention or treatment of inflammatory diseases, characterized in that it contains a compound of formula 3, a compound of formula 4 or a pharmaceutically acceptable salt thereof as an active ingredient.
제1항에 있어서, 상기 염증성 질환은 뇌염, 골관절염, 류마티스 관절염, 통풍, 강직성 척추염, 건염, 건막염, 류마티스 열, 루프스, 섬유근통(Fibromyalgia), 건선 관절염, 천식, 아토피, 크론병 또는 궤양성 대장염인 것을 특징으로 하는 약학적 조성물.
The method of claim 1, wherein the inflammatory disease is encephalitis, osteoarthritis, rheumatoid arthritis, gout, ankylosing spondylitis, tendinitis, tendonitis, rheumatic fever, lupus, fibromyalgia, psoriatic arthritis, asthma, atopy, Crohn's disease or ulcerative colitis Pharmaceutical composition, characterized in that.
제3항에 있어서, 상기 염증성 질환은 뇌염인 것을 특징으로 하는 약학적 조성물.
The pharmaceutical composition of claim 3, wherein the inflammatory disease is encephalitis.
제1항에 있어서, 상기 세스퀴테르펜 계열 대사체는 감송향 메탄올 추출물의 에틸아세테이트 분획물로부터 분리된 것을 특징으로 하는 염증성 질환의 예방 또는 치료용 약학적 조성물.
The method of claim 1, wherein the sesquiterpene-based metabolite is a pharmaceutical composition for the prevention or treatment of inflammatory diseases, characterized in that separated from the ethyl acetate fraction of methanol extract.
제1항에 있어서, 상기 조성물은 NO(Nitric oxide) 또는 PGE2(prostaglandin E2)의 생성을 억제하는 것을 특징으로 하는 염증성 질환의 예방 또는 치료용 약학적 조성물.
According to claim 1, wherein the composition is a pharmaceutical composition for the prevention or treatment of inflammatory diseases, characterized in that to inhibit the production of NO (Nitric oxide) or PGE 2 (prostaglandin E 2 ).
제1항에 있어서, 상기 조성물은 iNOS(inducible NO synthase) 또는 COX-2(Cyclooxygenase-2) 단백질의 발현을 억제하는 것을 특징으로 하는 염증성 질환의 예방 또는 치료용 약학적 조성물.
According to claim 1, wherein the composition is a pharmaceutical composition for the prevention or treatment of inflammatory diseases, characterized in that by inhibiting the expression of inducible NO synthase (iNOS) or COX-2 (Cyclooxygenase-2) protein.
제1항에 있어서, 상기 조성물은 IL-1β(Interleukin 1β), IL-6(Interleukin 6) 또는 TNF-α(Tumor Necrosis Factor α)의 발현을 억제하는 것을 특징으로 하는 염증성 질환의 예방 또는 치료용 약학적 조성물.
According to claim 1, wherein the composition is for the prevention or treatment of inflammatory diseases, characterized in that the suppression of the expression of IL-1β (Interleukin 1β), IL-6 (Interleukin 6) or TNF-α (Tumor Necrosis Factor α) Pharmaceutical compositions.
하기 화학식 1 내지 9의 화합물, 및 이의 약제학적으로 허용가능한 염으로 이루어지는 군으로부터 선택되는 하나 이상의 세스퀴테르펜 계열 대사체를 유효성분으로 함유하는 염증성 질환의 예방 또는 개선용 식품 조성물.
[화학식 1]
Figure pat00031

(R는 β-OH, 또는 α-메톡시-α-트리플루오로메틸페닐아세테이트기임).
[화학식 2]
Figure pat00032

(R는 α-OH임).
[화학식 3]
Figure pat00033

[화학식 4]
Figure pat00034

[화학식 5]
Figure pat00035

[화학식 6]
Figure pat00036

[화학식 7]
Figure pat00037

[화학식 8]
Figure pat00038

[화학식 9]
Figure pat00039

A food composition for preventing or ameliorating an inflammatory disease containing as an active ingredient at least one sesquiterpene-based metabolite selected from the group consisting of a compound of Formulas 1 to 9, and pharmaceutically acceptable salts thereof.
[Formula 1]
Figure pat00031

(R is a β-OH or α-methoxy-α-trifluoromethylphenylacetate group).
[Formula 2]
Figure pat00032

(R is α-OH).
[Formula 3]
Figure pat00033

[Formula 4]
Figure pat00034

[Formula 5]
Figure pat00035

[Formula 6]
Figure pat00036

[Formula 7]
Figure pat00037

[Formula 8]
Figure pat00038

[Formula 9]
Figure pat00039

제9항에 있어서, 상기 식품 조성물은 정제, 과립제, 환제, 캅셀제, 액제 또는 산제의 형태로 제형화될 수 있는 것을 특징으로 하는 식품 조성물.
The food composition of claim 9, wherein the food composition may be formulated in the form of tablets, granules, pills, capsules, liquids or powders.
하기 화학식 1 내지 9의 화합물, 및 이의 약제학적으로 허용가능한 염으로 이루어지는 군으로부터 선택되는 하나 이상의 세스퀴테르펜 계열 대사체를 유효성분으로 함유하는 염증성 질환의 예방 또는 개선용 화장품 조성물.
[화학식 1]
Figure pat00040

(R는 β-OH, 또는 α-메톡시-α-트리플루오로메틸페닐아세테이트기임).
[화학식 2]
Figure pat00041

(R는 α-OH임).
[화학식 3]
Figure pat00042

[화학식 4]
Figure pat00043

[화학식 5]
Figure pat00044

[화학식 6]
Figure pat00045

[화학식 7]
Figure pat00046

[화학식 8]
Figure pat00047

[화학식 9]
Figure pat00048

A cosmetic composition for preventing or ameliorating an inflammatory disease containing as an active ingredient at least one sesquiterpene-based metabolite selected from the group consisting of a compound of Formulas 1 to 9, and pharmaceutically acceptable salts thereof.
[Formula 1]
Figure pat00040

(R is a β-OH or α-methoxy-α-trifluoromethylphenylacetate group).
[Formula 2]
Figure pat00041

(R is α-OH).
[Formula 3]
Figure pat00042

[Formula 4]
Figure pat00043

[Formula 5]
Figure pat00044

[Formula 6]
Figure pat00045

[Formula 7]
Figure pat00046

[Formula 8]
Figure pat00047

[Formula 9]
Figure pat00048

제11항에 있어서, 상기 화장품 조성물은 용액, 현탁액, 에멀전, 페이스트, 겔, 크림, 로션, 파우더, 비누, 계면활성제-함유 클렌징, 오일, 분말 파운데이션, 에멀전 파운데이션, 왁스 파운데이션 또는 스프레이의 형태로 제형화될 수 있는 것을 특징으로 하는 화장품 조성물.
The formulation of claim 11 wherein the cosmetic composition is in the form of a solution, suspension, emulsion, paste, gel, cream, lotion, powder, soap, surfactant-containing cleansing, oil, powder foundation, emulsion foundation, wax foundation or spray Cosmetic composition, characterized in that can be converted.
하기 화학식 I의 화합물 또는 이의 약제학적으로 허용가능한 염:
[화학식 I]
Figure pat00049

(여기서, R는 α-OH, β-OH, 또는 α-메톡시-α-트리플루오로메틸페닐아세테이트기임).
A compound of formula (I) or a pharmaceutically acceptable salt thereof:
[Formula I]
Figure pat00049

Wherein R is an α-OH, β-OH, or α-methoxy-α-trifluoromethylphenylacetate group.
KR1020180044378A 2018-04-17 2018-04-17 Composition for anti-inflammatory treatment comprising sesquiterpene-based metabolites KR102074575B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
KR1020180044378A KR102074575B1 (en) 2018-04-17 2018-04-17 Composition for anti-inflammatory treatment comprising sesquiterpene-based metabolites

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
KR1020180044378A KR102074575B1 (en) 2018-04-17 2018-04-17 Composition for anti-inflammatory treatment comprising sesquiterpene-based metabolites

Publications (2)

Publication Number Publication Date
KR20190120967A true KR20190120967A (en) 2019-10-25
KR102074575B1 KR102074575B1 (en) 2020-02-06

Family

ID=68420557

Family Applications (1)

Application Number Title Priority Date Filing Date
KR1020180044378A KR102074575B1 (en) 2018-04-17 2018-04-17 Composition for anti-inflammatory treatment comprising sesquiterpene-based metabolites

Country Status (1)

Country Link
KR (1) KR102074575B1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115813894A (en) * 2021-09-18 2023-03-21 海南师范大学 Sesquiterpene compound and preparation method and application thereof

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20100132172A (en) * 2009-06-09 2010-12-17 강원대학교산학협력단 Anti-arthritic agent using cyathula officinalis
KR20120020518A (en) * 2010-08-30 2012-03-08 한국생명공학연구원 Eudesman compounds, thereof and compositions for containing the same as an active ingredient
KR20150068338A (en) * 2015-04-22 2015-06-19 주식회사한국전통의학연구소 A PHARMACEUTICAL COMPOSITION FOR PREVENTING AND TREATING SEPSIS COMPRISING DESOXO-NARCHINOL-A AND 8α-HYDROXYPINORESINOL AS ACTIVE INGREDIENTS
KR20150106276A (en) * 2014-03-11 2015-09-21 경희대학교 산학협력단 Anti-inflammatory composition containing diterpenes from the roots of oriza sativa l.
KR20150142154A (en) * 2014-06-10 2015-12-22 대한민국(농촌진흥청장) Anti-inflammatory active composition and pharmaceutical composition comprising the same
KR20160060580A (en) * 2014-11-19 2016-05-30 한국생명공학연구원 Pharmaceutical composition comprising sesquiterpenoid for preventing or treating STAT3 mediated disease and use thereof
KR101849338B1 (en) * 2017-03-02 2018-04-16 이화여자대학교 산학협력단 A pharmaceutical composition comprising Kaempferol-3-O-rhamnopyranoside or pharmaceutically acceptable salt thereof as an active ingredient for prevention or treatment of inflammatory diseases, allergic diseases or malaria

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20100132172A (en) * 2009-06-09 2010-12-17 강원대학교산학협력단 Anti-arthritic agent using cyathula officinalis
KR20120020518A (en) * 2010-08-30 2012-03-08 한국생명공학연구원 Eudesman compounds, thereof and compositions for containing the same as an active ingredient
KR20150106276A (en) * 2014-03-11 2015-09-21 경희대학교 산학협력단 Anti-inflammatory composition containing diterpenes from the roots of oriza sativa l.
KR20150142154A (en) * 2014-06-10 2015-12-22 대한민국(농촌진흥청장) Anti-inflammatory active composition and pharmaceutical composition comprising the same
KR20160060580A (en) * 2014-11-19 2016-05-30 한국생명공학연구원 Pharmaceutical composition comprising sesquiterpenoid for preventing or treating STAT3 mediated disease and use thereof
KR20150068338A (en) * 2015-04-22 2015-06-19 주식회사한국전통의학연구소 A PHARMACEUTICAL COMPOSITION FOR PREVENTING AND TREATING SEPSIS COMPRISING DESOXO-NARCHINOL-A AND 8α-HYDROXYPINORESINOL AS ACTIVE INGREDIENTS
KR101849338B1 (en) * 2017-03-02 2018-04-16 이화여자대학교 산학협력단 A pharmaceutical composition comprising Kaempferol-3-O-rhamnopyranoside or pharmaceutically acceptable salt thereof as an active ingredient for prevention or treatment of inflammatory diseases, allergic diseases or malaria

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Journal of Ethnopharmacology, vol. 160, pp 94~100(2015년) *
석곡과 감송향의 일산화질소 생성 저해 성분(충북대학교 박사학위논문, 황지상, 2010.02.) *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115813894A (en) * 2021-09-18 2023-03-21 海南师范大学 Sesquiterpene compound and preparation method and application thereof

Also Published As

Publication number Publication date
KR102074575B1 (en) 2020-02-06

Similar Documents

Publication Publication Date Title
EP2214790B1 (en) Therapeutic applications of extracts of fraxinus excelsior seeds
Wang et al. Anti-inflammatory eudesmane sesquiterpenoids from Artemisia hedinii
Brahmachari Andrographolide: A molecule of antidiabetic promise
US4046914A (en) Therapeutically active substituted saturated and mono-and polyunsaturated alkyl-glycerylethers
TW202304497A (en) Novel polyphenol compound
KR102074575B1 (en) Composition for anti-inflammatory treatment comprising sesquiterpene-based metabolites
KR102446515B1 (en) Composition for suppressing adipocyte differentiation containing artemisia scoparia extract, fraction thereof or compound separated therefrom as an active ingredient for preventing or improving obesity, health functional food
Hosford et al. Natural antagonists of platelet‐activating factor
KR980008225A (en) Anticancer drugs containing dexacinol angelate as an active ingredient
KR101556773B1 (en) Anti-inflammatory composition containing diterpenes from the roots of oriza sativa l.
CZ288170B6 (en) Pharmaceutical composition and use thereof
KR20120121684A (en) Antipruritic composition containing astragalin and quercetin
Cho et al. Sesquiterpene lactone and its unique proaporphine hybrids from Magnolia grandiflora L. and their anti-inflammatory activity
CN114874213B (en) Preparation of matrine and matrine extract and anti-inflammatory and antipruritic applications
KR102057388B1 (en) Composition for anti-inflammatory treatment comprising nardosinone-based metabolites
TW202313064A (en) Novel isoflavone compound
KR100470158B1 (en) An extract of Sedum kamtschaticum Fisch. and a chemical compound isolated from the extract, and a pharmaceutical composition for anti-inflammatory containing them as an effective constituents
KR101056878B1 (en) Novel Jasmonate Derivatives and Their Pharmaceutical Uses
CN109045012A (en) The application of new skeleton loop coil sesquiterpene dimers compound
KR102525843B1 (en) Anti-inflammatory composition comprising Liriodendron tulipifera extracts or alkamide isolated therefrom
KR101577189B1 (en) - Composition for preventing or treating of allergy comprising -cubebenoate
JP4500951B1 (en) DNA synthase inhibitors
KR102612269B1 (en) Compound extracspted from the leave of Viburnum erosum Thunb and tyrosinase activity inhibitory ability composition comprising the same
KR101038022B1 (en) A novel sugar compound
JP4296312B2 (en) Novel triterpene compound and anticancer / immunity enhancer comprising the compound as an active ingredient

Legal Events

Date Code Title Description
A201 Request for examination
E902 Notification of reason for refusal
AMND Amendment
E601 Decision to refuse application
AMND Amendment
X701 Decision to grant (after re-examination)