KR20100081627A - A composition comprising the compound isolated from the extract of rubiae radix for preventing and treating inflammatory disease - Google Patents

A composition comprising the compound isolated from the extract of rubiae radix for preventing and treating inflammatory disease Download PDF

Info

Publication number
KR20100081627A
KR20100081627A KR1020090000936A KR20090000936A KR20100081627A KR 20100081627 A KR20100081627 A KR 20100081627A KR 1020090000936 A KR1020090000936 A KR 1020090000936A KR 20090000936 A KR20090000936 A KR 20090000936A KR 20100081627 A KR20100081627 A KR 20100081627A
Authority
KR
South Korea
Prior art keywords
extract
methoxy
compound
prenyl
carbomethoxy
Prior art date
Application number
KR1020090000936A
Other languages
Korean (ko)
Other versions
KR101072663B1 (en
Inventor
김영호
전도연
우현주
우미희
양재하
Original Assignee
대구한의대학교산학협력단
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 대구한의대학교산학협력단 filed Critical 대구한의대학교산학협력단
Priority to KR1020090000936A priority Critical patent/KR101072663B1/en
Priority to PCT/KR2009/007837 priority patent/WO2010079914A2/en
Publication of KR20100081627A publication Critical patent/KR20100081627A/en
Application granted granted Critical
Publication of KR101072663B1 publication Critical patent/KR101072663B1/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
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • A23L33/105Plant extracts, their artificial duplicates or their derivatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/74Rubiaceae (Madder family)
    • 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]

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Natural Medicines & Medicinal Plants (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Botany (AREA)
  • Mycology (AREA)
  • Medical Informatics (AREA)
  • Biotechnology (AREA)
  • Alternative & Traditional Medicine (AREA)
  • Microbiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Nutrition Science (AREA)
  • Food Science & Technology (AREA)
  • Polymers & Plastics (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Rheumatology (AREA)
  • Pain & Pain Management (AREA)
  • Medicines Containing Plant Substances (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

PURPOSE: A composition containing a compound isolated from Rubiae Radix is provided to reduce expression of protein which causes inflammation and pro-inflammatory cytokine expression. CONSTITUTION: A pharmaceutical composition for preventing and treating inflammatory disease contains a compound isolated from Rubiae Radix extract or pharmaceutically acceptable salt as an active ingredient. The compound is 2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone, 3'-bis(3,4-dihydro-4-hydroxy-6-methoxy-2H-1-benzopyran), Epoxymollugin, Soranjidiol, Oleanolic acid, 1-acetoxy-3-methoxy-9,10-anthraquinone, Alizarin 2-methyl ether, Furonollugin, or mollugin. The Rubiae Radix extract is crude extract, polar solvent soluble extract, or non-polar solvent soluble extract.

Description

천초근 추출물로부터 분리된 화합물을 유효성분으로 함유하는 염증성 질환의 예방 및 치료용 조성물 {A composition comprising the compound isolated from the extract of Rubiae Radix for preventing and treating inflammatory disease}A composition comprising the compound isolated from the extract of Rubiae Radix for preventing and treating inflammatory disease}

본 발명은 천초근 추출물로부터 분리된 화합물을 유효성분으로 함유하는 염증성 질환의 예방 및 치료용 조성물을 제공한다.The present invention provides a composition for the prophylaxis and treatment of inflammatory diseases containing a compound isolated from Cheoncho root extract as an active ingredient.

[문헌 1] Hawiger J., Innate Immunity and Inflammation: A Transcriptional Paradigm, Immunologic Research, 23, pp.99-109, 2001Hawiger J., Innate Immunity and Inflammation: A Transcriptional Paradigm, Immunologic Research, 23 , pp. 99-109, 2001

[문헌 2] Gallo RL, et al., Biology and clinical relevance of naturally occurring antimicrobial peptides, J. Allergy. Clin. Immunol., 110, pp.823-831. 2002Gallo RL, et al., Biology and clinical relevance of naturally occurring antimicrobial peptides, J. Allergy. Clin. Immunol., 110 , pp. 823-831. 2002

[문헌 3] Graeme B, et al., Acute Inflammation, American Journal of Pathology, 86(1), pp.185-274, 1977Graeme B, et al., Acute Inflammation, American Journal of Pathology, 86 (1) , pp.185-274, 1977

[문헌 4] Yamaki K, et al., Characteristics of histamine-induced leukocyte rolling in the undisturbed microcirculation of the rat mesentry, British J. Pharmacol., 123, pp.390-399, 1998Yamaki K, et al., Characteristics of histamine-induced leukocyte rolling in the undisturbed microcirculation of the rat mesentry, British J. Pharmacol., 123 , pp . 390-399, 1998

[문헌 5] Brocklehurst WE, Role of Kinins and Prostaglandins in Inflammation, Proc. Roy. Soc. Med., 64, pp.4-6, 1971Document 5 Brocklehurst WE, Role of Kinins and Prostaglandins in Inflammation, Proc. Roy. Soc. Med., 64 , pp . 4-6, 1971

[문헌 6] Esmon CT, The interactions between inflammation and coagulation. British Journal of Haematology, 131, pp.417-430, 2005Esmon CT, The interactions between inflammation and coagulation. British Journal of Haematology, 131 , pp. 417-430, 2005

[문헌 7] Huang AL, et al., Effects of Systemic Inflammation on Endothelium-Dependent Vasodilation, Trends Cardiovasc. Med., 16, pp.15-20, 2006Huang AL, et al., Effects of Systemic Inflammation on Endothelium-Dependent Vasodilation, Trends Cardiovasc. Med ., 16 , pp. 15-20, 2006

[문헌 8] Renauld JC, New insights into the role of cytokines in asthma, J. Clin. Pathol, 54, pp.577-589, 2001Renauld JC, New insights into the role of cytokines in asthma, J. Clin. Pathol, 54 , pp. 577-589, 2001

[문헌 9] Blake GJ, et al., Tumour necrosis factor-α, inflammatory biomarkers, and atherogenesis, Eur. Heart J., 23, pp.345-347, 20029 Blake GJ, et al., Tumor necrosis factor-α, inflammatory biomarkers, and atherogenesis, Eur. Heart J., 23 , pp. 345-347, 2002

[문헌 10] Moncada S, et al., Nitric oxide: Physiology, pathophysiology, and pharmacology, Pharmacol. Rev., 43, pp.109-142, 1991Moncada S, et al., Nitric oxide: Physiology, pathophysiology, and pharmacology, Pharmacol. Rev. , 43 , pp.109-142, 1991

[문헌 11] Mordan LJ, et al., Inhibitors of endogenous nitrogen oxide formation block the promotion of neoplastic transformation in C3H10T1/2 fibroblasts, Carcinogenesis, 14, pp.1555-1559, 1993Mordan LJ, et al., Inhibitors of endogenous nitrogen oxide formation block the promotion of neoplastic transformation in C3H10T1 / 2 fibroblasts, Carcinogenesis, 14 , pp. 1555-1559, 1993

[문헌 12] Lirk P, et al., Inducible nitric oxide synthase. Time for reappraisal, Curr. Drug. Targets. Inflamm. Allergy., 1, pp.89-108, 2002Lirk P, et al., Inducible nitric oxide synthase. Time for reappraisal, Curr. Drug. Targets. Inflamm. Allergy., 1 , pp.89-108, 2002

[문헌 13] Hattori Y, et al., Lipopolysaccharide activates Akt in vascular smooth muscle cells resulting in induction of inducible nitric oxide synthase through nuclear factor-kappa B activation, Eur. J. Pharmacol, 481, pp.153-158, 2003Hattori Y, et al., Lipopolysaccharide activates Akt in vascular smooth muscle cells resulting in induction of inducible nitric oxide synthase through nuclear factor-kappa B activation, Eur. J. Pharmacol , 481 , pp. 153-158, 2003

[문헌 14] Kim SH, et al., Selenium attenuates lipopolysaccharide- induced oxidative stress responses through modulation of p38 MAPK and NF-κB signaling pathways, Exp. Biol. Medicine, pp.565-701, 2004[14] Kim SH, et al., Selenium attenuates lipopolysaccharide-induced oxidative stress responses through modulation of p38 MAPK and NF-κB signaling pathways, Exp. Biol. Medicine, pp . 565-701, 2004

[문헌 15] Robinson MJ, et al., Mitogen-activated protein kinase pathways, Cur. Opi. Cell Biol., 9, pp.180-186, 1997Robinson MJ, et al., Mitogen-activated protein kinase pathways, Cur. Opi. Cell Biol., 9 , pp. 180-186, 1997

[문헌 16] 박종희, 한약백과도감(하), 도서출판 신일상사, p.796-797, 2002[Document 16] Park, Hee Park, Encyclopedia of Herbal Medicine (B), Book Publishing Company, Shinil Corporation, p.796-797, 2002

[문헌 17] Jun DY, et al., Apoptogenic activity of Zanthoxylum schinifolium toward human acute leukemia Jurkat T cells is associated with ER stress-mediated caspase-8 activation that stimulates mitochondria- dependent or -independent caspase cascade, Carcinogenesis, 28, pp.1303-1313, 2007Jun DY, et al., Apoptogenic activity of Zanthoxylum schinifolium toward human acute leukemia Jurkat T cells is associated with ER stress-mediated caspase-8 activation that stimulates mitochondria-dependent or -independent caspase cascade, Carcinogenesis, 28 , pp .1303-1313, 2007

[문헌 18] Wang S et al., J. Ethnopharmacol., 114(3), pp.458-462, 200718. Wang S et al., J. Ethnopharmacol. , 114 (3) , pp.458-462, 2007

[문헌 19] Heo et al., J. Immunol., 179(9), pp.6305-6310, 2007Heo et al., J. Immunol., 179 (9) , pp.6305-6310, 2007

[문헌 20] Haiqi He, et al., Molecular Immunology, 43, p.783, 2006[20] Haiqi He, et al., Molecular Immunology, 43 , p.783, 2006

[문헌 21] Lee et al., J. Ethnopharmacol., 97, pp.561-566, 2005[21] Lee et al., J. Ethnopharmacol., 97 , pp.561-566, 2005

염증반응은 병원체에 의한 감염이나 조직의 손상과 같은 다양한 요인에 의해 일어나는 생체방어반응으로 감염부위나 손상부위에만 피해를 국한시키기 위한 초기 보호작용을 수행한다. 대부분의 경우, 이러한 염증반응은 내재면역의 구성 요소를 이용한 병원성 요인의 제거 및 특이적 적응면역의 유도 등으로 이어진다 (Hawiger J., Innate Immunity and Inflammation: A Transcriptional Paradigm, Immunologic Research, 23, pp.99-109, 2001). 염증에 수반되는 특징으로서 알려진 발적 (rubor), 부종 (tumor), 열 (calor), 통증 (dolor) 등은 염증부위에서의 염증 매개체와 사이토카인 등의 작용에 의한 혈관의 확장에 따른 국소혈류의 증가 및 국소혈류속도의 감소, 혈관의 투과성증가에 따른 혈장성분의 혈관외 유출 증가, 혈관내피세포의 순환면역세포에 대한 부착성 증가에 따른 면역세포의 혈관외 유출 증가 및 화학주성에 의한 감염부위로의 이동 증가와 같은 연속적인 면역반응들의 결과이다(Gallo RL, et al., Biology and clinical relevance of naturally occurring antimicrobial peptides, J. Allergy. Clin. Immunol, 110, pp.823-831. 2002; Graeme B, et al., Acute Inflammation, American Journal of Pathology, 86(1), pp.185-274, 1977). 조직손상에 대한 반응으로서 일부 세포들에서 생성되는 히스타민, 그리고 혈액 내에서 비활성화 상태로 존재하다가 조직손상에 의해 활성화되는 저분자 펩티드성 키닌도 염증관련 혈관확장과 혈관의 투과성 증가에 기여한다(Yamaki K, et al., Characteristics of histamine-induced leukocyte rolling in the undisturbed microcirculation of the rat mesentry, British J. Pharmacol, 123, pp.390-399, 1998; Brocklehurst WE, Role of Kinins and Prostaglandins in Inflammation, Proc. Roy. Soc. Med., 64, pp.4-6, 1971). 특히 키닌의 일종인 브라디키닌은 피부의 통점을 자극하는 작용을 한다. 한편, 손상된 조직에서의 혈관의 확장과 혈관의 투과성 증가는 또한 혈액응고효소들이 조직으로 유출되게 하고 뒤이어 이들 효소의 연쇄반응에 의해 혈액응고의 주요 요소인 불용성의 섬유소가닥이 생성되게 한다. 응고된 섬유소 가닥은 손상된 조직부위를 통한 감염이 다른 부위로 확산되는 것을 차단하는 방어벽 역할을 한다 (Esmon CT., The interactions between inflammation and coagulation. British Journal of Haematology. 131, pp.417-430, 2005). 일반적으로 급성염증반응은 빠르게 진행되고 짧은 기간 유지되며, 급성염증에는 급성기 반응이라는 전신성 반응이 동반된다. 한편 감염이나 자가면역질환과 같은 일부 질환과 관련하여 지속적인 면역활성화의 결과로서 만성염증이 유발될 수 있으며, 대식세포 (macrophages)의 축적과 활성화는 만성염증반응의 증거가 된다 (Huang AL, et al., Effects of Systemic Inflammation on Endothelium-Dependent Vasodilation, Trends, Cardiovasc. Med., 16, pp.15-20, 2006). 그러나 지속적인 만성염증반응의 경우, 숙주세포나 조직에 심각한 손상을 유발할 수 있다. Inflammatory reactions are biological defenses caused by various factors, such as infection by tissues or damage to tissues, and perform an initial protective action to limit the damage to only infected or damaged areas. In most cases, this inflammatory response leads to the elimination of pathogenic factors using intrinsic immune components and induction of specific adaptive immunity (Hawiger J., Innate Immunity and Inflammation: A Transcriptional Paradigm, Immunologic Research, 23 , pp. 99-109, 2001). Rubs, tumors, heat, and pain, known as inflammation-associated features, are associated with the expansion of local blood flow due to the expansion of blood vessels by the action of inflammatory mediators and cytokines. Increased extravascular outflow of plasma components due to increased and decreased local blood flow rate, increased permeability of blood vessels, increased extravascular outflow of immune cells due to increased adhesion of vascular endothelial cells to circulating immune cells, and infection due to chemotaxis This is the result of successive immune responses such as increased migration to (Gallo RL, et al., Biology and clinical relevance of naturally occurring antimicrobial peptides, J. Allergy. Clin. Immunol, 110 , pp. 823-831. 2002; Graeme B, et al., Acute Inflammation, American Journal of Pathology, 86 (1) , pp. 185-274, 1977. Histamine produced in some cells as a response to tissue damage, and low molecular peptide kinin, which is inactivated in the blood and is activated by tissue damage, also contributes to inflammation-associated vasodilation and increased blood vessel permeability (Yamaki K, et al., Characteristics of histamine-induced leukocyte rolling in the undisturbed microcirculation of the rat mesentry, British J. Pharmacol, 123 , pp. 390-399, 1998; Brocklehurst WE, Role of Kinins and Prostaglandins in Inflammation, Proc. Roy. Soc. Med., 64 , pp. 4-6, 1971). Especially, bradykinin, a kind of kinin, works to stimulate the pain of the skin. On the other hand, dilation of blood vessels and increased permeability of blood vessels in damaged tissues also cause blood coagulation enzymes to leak into the tissues, followed by the chain reaction of these enzymes to produce insoluble fibrous strands, which is a major component of blood coagulation. Coagulated fibrous strands act as a barrier against the spread of infection through damaged tissue (Esmon CT., The interactions between inflammation and coagulation. British Journal of Haematology. 131 , pp . 417-430, 2005 ). In general, acute inflammatory reactions progress rapidly and are maintained for a short period of time. Acute inflammation is accompanied by a systemic reaction called an acute phase reaction. On the other hand, chronic inflammation can be induced as a result of continuous immune activation associated with some diseases such as infections and autoimmune diseases, and the accumulation and activation of macrophages is evidence of chronic inflammatory response (Huang AL, et al. ., Effects of Systemic Inflammation on Endothelium-Dependent Vasodilation, Trends, Cardiovasc.Med ., 16 , pp. 15-20, 2006). However, persistent chronic inflammatory reactions can cause serious damage to host cells or tissues.

감염부위에서의 염증반응은 병원체에 대한 대식세포의 반응에 의해 개시된다. 병원체에 의해 활성화된 대식세포가 생성하는 반응성 활성산소종 및 활성질소종, 프로스타글란딘, 루코트리엔 등과 같은 염증매개체, 그리고 TNF-α, IL-6 및 IL-1β 등과 같은 염증유발성 사이토카인 등이 염증반응에 관여되는 것으로 알려져 있다 (Renauld JC, New insights into the role of cytokines in asthma, J. Clin. Pathol., 54, pp.577-589, 2001; Blake GJ, et al., Tumour necrosis factor-α, inflammatory biomarkers, and atherogenesis, Eur. Heart J., 23, pp.345-347, 2002). 이러한 대식세포의 염증관련 작용에 있어서는 이들 염증매개체 유전자들의 발현을 유도하는 전사인자인 NF-κB의 활성화가 매우 중요하다. 대식세포 내에서 유도성 산화질소 합성효소 (inducible nitric oxide synthase, iNOS), 시클로옥시게나제 (cyclooxygenase, COX-2), TNF-α, IL-6, IL-1β 등의 염증관련 유전자들이 NF-κB에 의해 전사된다. Inflammatory responses at the site of infection are initiated by the response of macrophages to pathogens. Reactive reactive oxygen species and reactive nitrogen species produced by macrophages activated by pathogens, inflammatory mediators such as prostaglandins, leukotrienes, and inflammatory cytokines such as TNF-α, IL-6 and IL-1β. (Renauld JC, New insights into the role of cytokines in asthma, J. Clin. Pathol ., 54 , pp. 577-589, 2001; Blake GJ, et al., Tumour necrosis factor -α, inflammatory biomarkers, and atherogenesis, Eur. Heart J., 23 , pp.345-347, 2002). In the inflammation-related action of such macrophages, the activation of NF-κB, a transcription factor that induces the expression of these inflammatory mediator genes, is very important. Inflammatory genes such as inducible nitric oxide synthase (iNOS), cyclooxygenase (COX-2), TNF-α, IL-6 and IL-1β in macrophages is transcribed by κB.

산화질소 (nitric oxide, NO)는 지속력이 짧은 자유라디칼로서 산화질소 합성효소 (nitric oxide synthase, NOS)의 촉매작용에 의해 L-아르기닌 (L-arginine)의 산화반응이 일어날 때, L-아르기닌으로부터 L-시트룰린 (L-citrulline)과 함께 산화질소가 생성된다는 사실은 이미 널리 알려져 있다. 생체 내에서 산화질소는 다양한 면역관련 기능을 나타내지만 (Moncada S, et al., Nitric oxide: Physiology, pathophysiology, and pharmacology. Pharmacol. Rev., 43, pp.109-142, 1991), 과도한 산화질소의 생성은 오히려 세포독성을 나타낼 수 있으므로 숙주세포 및 조직의 손상을 초래할 뿐만 아니라 동맥경화, 발암 등 다양한 병리적 현상에 관여할 수도 있다 (Mordan LJ, et al., Inhibitors of endogenous nitrogen oxide formation block the promotion of neoplastic transformation in C3H10T1/2 fibroblasts, Carcinogenesis, 14, pp.1555-1559, 1993; Lirk P, et al., Inducible nitric oxide synthase, Time for reappraisal, Curr. Drug. Targets. Inflamm. Allergy., 1, pp.89-108, 2002). NO 생성을 촉매하는 산화질소 합성효소 (NOS)에는 세 가지 유형이 알려져 있는데, 이들 중 신경 산화질소 합성효소 (neuronal NOS, nNOS, NOS1)와 내피 산화질소 합성효소 (endothelial NOS, eNOS, NOS3)는 구성적으로 항상 발현되는 반면, iNOS는 식세포 (phagocytes)가 세균성 내독소인 지질다당류 (lipopolysaccharide, LPS)에 의해 자극될 때 활성화되는 전사인자 NF-κB에 의해 비로소 전사되는 유도성이다. Nitric oxide (NO) is a short-lasting free radical that is derived from L-arginine when the oxidation reaction of L-arginine occurs by catalysis of nitric oxide synthase (NOS). It is well known that nitric oxide is produced with L-citrulline. Nitric oxide exhibits various immune-related functions in vivo (Moncada S, et al., Nitric oxide: Physiology, pathophysiology, and pharmacology.Pharmacol. Rev. , 43 , pp.109-142, 1991) Production may be cytotoxic and may lead to damage to host cells and tissues, as well as to various pathological phenomena such as atherosclerosis and carcinogenesis (Mordan LJ, et al., Inhibitors of endogenous nitrogen oxide formation block the promotion of neoplastic transformation in C3H10T1 / 2 fibroblasts, Carcinogenesis, 14 , pp. 1555-1559, 1993; Lirk P, et al., Inducible nitric oxide synthase, Time for reappraisal, Curr. Drug.Targets.Inflamm.Allergy., 1 , pp.89-108, 2002). Three types of nitric oxide synthase (NOS) are known to catalyze NO production, including neuronal nitric oxide synthase (neuronal NOS, nNOS, NOS1) and endothelial nitric oxide synthase (endothelial NOS, eNOS, NOS3). While constitutively expressed at all times, iNOS is inducible to be transcribed by the transcription factor NF-κB, which is activated when phagocytes are stimulated by the bacterial endotoxin lipopolysaccharide (LPS).

대식세포 내에서 전사인자 NF-κB는 그 저해제인 IκB와 결합하여 불활성 상태로 세포질에 존재하지만, 세포표면의 Toll-유사 수용체 (Toll-like receptor)에 세균성 LPS가 결합할 때 유도되는 신호전달과정에 의해 IκB가 인산화되어 NF-κB/IκB로부터 해리되고 프로테아좀 분해작용에 의해 제거됨에 따라 NF-κB는 비로소 활성화 상태로 된다. 이어서 NF-κB는 핵으로 이동하여 다양한 염증 관련 유전자들의 전사를 유도한다. 이러한 NF-κB의 활성화에는 Akt 신호전달 (Hattori Y, et al., Lipopolysaccharide activates Akt in vascular smooth muscle cells resulting in induction of inducible nitric oxide synthase through nuclear factor-kappa B activation, Eur. J. Pharmacol, 481, pp.153-158, 2003)과 ERK, c-jun- 및 p38-MAPK 신호전달 경로가 관여하는 것으로 보고되었다 (Kim SH, et al., Selenium attenuates lipopolysaccharide- induced oxidative stress responses through modulation of p38 MAPK and NF-κB signaling pathways, Exp. Biol. Medicine., pp.565-701, 2004; Robinson MJ, et al., Mitogen-activated protein kinase pathways, Cur. Opi. Cell Biol., 9, pp.180-186, 1997). In macrophages, the transcription factor NF-κB is present in the cytoplasm in an inactive state by binding to its inhibitor, IκB, but is a signaling process induced when bacterial LPS binds to Toll-like receptors on the cell surface. NF-κB finally becomes activated as IκB is phosphorylated to dissociate from NF-κB / IκB and removed by proteasome digestion. NF-κB then migrates to the nucleus to induce transcription of various inflammation related genes. Such activation of NF-κB includes Akt signaling (Hattori Y, et al., Lipopolysaccharide activates Akt in vascular smooth muscle cells resulting in induction of inducible nitric oxide synthase through nuclear factor-kappa B activation, Eur. J. Pharmacol , 481 , 153-158, 2003) and ERK, c-jun- and p38-MAPK signaling pathways have been reported to be involved (Kim SH, et al., Selenium attenuates lipopolysaccharide-induced oxidative stress responses through modulation of p38 MAPK and NF-κB signaling pathways, Exp.Biol.Medicine ., Pp.565-701, 2004; Robinson MJ, et al., Mitogen-activated protein kinase pathways, Cur.Opi.Cell Biol., 9 , pp.180-186 , 1997).

염증반응에 있어서 iNOS의 전사단계에서의 발현조절은 NO의 지속시간과 생성정도를 결정하는데 있어서 대단히 중요하다. 그러므로 iNOS의 발현조절기전 및 iNOS의 효소활성은 만성염증관련 질환의 개선을 위한 새로운 항염증 약제를 분리하고자 하는 연구에서 약제작용의 표적으로 이용되고 있다. The expression regulation of iNOS in the transcriptional phase of the inflammatory response is of great importance in determining the duration and production of NO. Therefore, iNOS expression control mechanism and iNOS enzymatic activity have been used as a target of drug action in the study to isolate new anti-inflammatory drugs for the improvement of chronic inflammation-related diseases.

천초근(草根, Rubiae Radix)은 꼭두서니과(Rubiaceae)의 꼭두서니 (Rubia akane Nakai, =R. cordifolia L. var. mangista Miq.) 및 갈퀴꼭두서니 (Rubia cordifolia L.)의 뿌리를 건조한 것으로 다른 이름으로 천초(草), 홍천근(紅根), 지소본(地蘇本), 천근(根), 토숙본초(土宿本草), 혈견수(血見愁), 활혈단(活血丹), 지소목(地蘇木), 천초(草), 모수(茅蒐), 여려(茹), 염비초(染緋草), 풍자초(風車草), 과산용(過山龍), 우만(牛蔓)으로 불리운다. 산지는 한국과 중국이며, 특이한 냄새에 맛은 조금 달며 성질은 찬 것이 특징으로, 성분은 안트라퀴논(anthraquinone)류에 속하는 알리자린(alizarin), 루비에리트릭 산(rubierythric acid, =alizarin-2- primeveroside), 퍼푸린(purpurin), 산토퍼푸린(xanthopurpurin), 문지스틴(munjistin), 프소이도퍼푸린(pseudopurpurin) 이다. 염색 재료로 사용하며, 지혈작용, 생리불순, 혈액순환장애, 어혈의 효과가 있으나(박종희, 한약백과도감(하), 도서출판 신일상사, p.796-797, 2002), 상기 문헌 어디에도 본 발명의 천초근 추출물로부터 분리된 화합물이 LPS의 처리에 의해 유도되는 NO 생성 및 친염증성 사이토카인들의 생성을 억제하여 염증성 질환의 예방 및 치료를 위한 조성물로서 사용가능하다고 교시되거나 개시된 바 없다. Rubiae Radix is the dried root of Rubia akane Nakai (= R. Cordifolia L. var.mangista Miq.) And Rake cordifolia ( Rbia cordifolia L.). (草草), Hongcheongeun, Jisobon, Cheongeun, Tosukboncho, Blood Bran Number, Live Blood Troupes, Jisomok It is called 木, Cheoncho, Parasitic, Yeoryo, Yeombicho, Saengchocho, Wind Jarcho, Persanyong, and Uman. Its origin is Korea and China, and its characteristic odor is a little sweet and cold. Its ingredients are alizarin and rubierythric acid (= alizarin-2- primeveroside) belonging to anthraquinones. ), Purpurin (purpurin), xanthopurpurin (xanthopurpurin), munjistin (munjistin), pseudopurpurin (pseudopurpurin). It is used as a dyeing material and has the effects of hemostatic action, menstrual disorder, blood circulation disorder, and blood bleeding (Park Jong Hee, Encyclopedia of Herbal Medicine (B), Book Publishing Shinil Corp., p.796-797, 2002). The compounds isolated from the myofibrillar root extracts of do not teach or disclose that they can be used as compositions for the prevention and treatment of inflammatory diseases by inhibiting NO production and proinflammatory cytokines production induced by the treatment of LPS.

이에 본 발명자는 마우스 대식세포주인 RAW264.7을 세균성 LPS로 처리할 때 일어나는 NO 생성 및 친염증성 사이토카인의 생성을 저해하는 새로운 약제를 찾기 위하여, 천초 (Rubia cordifolia L.)의 뿌리(천초근)로부터 분리 정제한 단일물질들을 대상으로 RAW264.7 세포의 NO 생성 및 친염증성 사이토카인의 생성에 대한 저해능을 조사한 결과, 천초근으로부터 분리된 화합물인 2-카르보메톡시-2,3-에폭시-3-프레닐-1,4-나프토퀴논(이하‘RA-MC-AA’라 함)등의 천초근 분리 화합물이 RAW264.7 세포에 있어서 LPS의 처리에 의해 유도되는 NO 생성 및 친염증성 사이토카인의 생성을 저해하는 항염증성 기능이 있음을 최초로 확인하였으며, 마우스 대식세포인 RAW264.7 세포에서 LPS로 유도되어진 NO 생성량, p-IkBα, iNOS 및 COX-2의 단백질 발현을 감소시킬 뿐만 아니라 iNOS, COX-2, TNF-α, IL-6 및 IL-1β의 mRNA발현과 같은 친염증성 사이토카인들의 발현 함량을 유의적으로 감소함을 확인함으로써 본 발명을 완성하였다.Therefore, the present inventors found a new drug that inhibits the production of NO and proinflammatory cytokines produced by the treatment of mouse macrophage RAW264.7 with bacterial LPS, the root of Cheoncho ( Rubia cordifolia L.) Inhibition of NO production and pro-inflammatory cytokine production in RAW264.7 cells was investigated in the purified single-substances from 2-carbomethoxy-2,3-epoxy-3. Transdermal isolated compounds, such as prenyl-1,4-naphthoquinone (hereinafter referred to as 'RA-MC-AA'), are NO-producing and proinflammatory cytokines induced by LPS treatment in RAW264.7 cells. For the first time, it has been shown to have an anti-inflammatory function that inhibits the production of LPS-induced NO production, p-IkBα, iNOS and COX-2 protein expression in mouse macrophage RAW264.7 cells as well as iNOS, COX-2, TNF-α, IL-6 and The present invention was completed by confirming that the expression content of proinflammatory cytokines such as mRNA expression of IL-1β was significantly reduced.

상기 목적을 달성하기 위하여, 본 발명은 천초근 추출물로부터 분리된 화합물 또는 이의 약학적으로 허용 가능한 염을 유효성분으로 함유하는 염증성 질환의 예방 및 치료용 약학조성물을 제공한다.In order to achieve the above object, the present invention provides a pharmaceutical composition for the prevention and treatment of inflammatory diseases containing a compound or a pharmaceutically acceptable salt thereof isolated from Cheoncho root extract as an active ingredient.

또한, 본 발명은 천초근 추출물로부터 분리된 화합물 또는 이의 약학적으로 허용 가능한 염을 유효성분으로 함유하는 염증성 질환의 예방 및 개선용 건강기능식품을 제공한다.In addition, the present invention provides a health functional food for the prevention and improvement of inflammatory diseases containing a compound or a pharmaceutically acceptable salt thereof isolated from Cheoncho root extract as an active ingredient.

본원에서 정의되는 천초근으로부터 분리된 화합물은 2-카르보메톡시-2,3-에폭시-3-프레닐-1,4-나프토퀴논 (2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone), 3,3′-비스(3,4-디하이드로-4-하이드록시-6-메톡시-2H-1-벤조피란)(3,3′-bis(3,4-dihydro-4-hydroxy-6-methoxy-2H-1-benzopyran)), 에폭시몰루긴(Epoxymollugin), 소란지디올(Soranjidiol), 올레아놀릭 산(Oleanolic acid), 1-아세톡시-3-메톡시-9,10-안트라퀴논(1-acetoxy-3-methoxy-9,10-anthraquinone), 알리자린 2-메틸 아테르(Alizarin 2-methyl ether), 푸로놀루긴(Furonollugin), 몰루긴(mollugin)으로 구성된 군으로부터 선택된 화합물이며, 바람직하게는 2-카르보메톡시-2,3-에폭시-3-프레닐-1,4-나프토퀴논(2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone) 화합물임을 특징으로 한다.Compounds isolated from the herbaceous root as defined herein may be 2-carbomethoxy-2,3-epoxy-3-prenyl- (2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone). 1,4-naphthoquinone), 3,3'-bis (3,4-dihydro-4-hydroxy-6-methoxy-2H-1-benzopyran) (3,3'-bis (3,4- dihydro-4-hydroxy-6-methoxy-2H-1-benzopyran), Epoxymollugin, Soranjidiol, Oleanolic acid, 1-acetoxy-3-methoxy- Group consisting of 9,10-anthraquinone (1-acetoxy-3-methoxy-9,10-anthraquinone), alizarin 2-methyl ether, furonollugin, mollugin It is a compound selected from, preferably 2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone (2-carbomethoxy-2,3-epoxy-3-prenyl-1,4 -naphthoquinone) compound.

본 발명의 화합물은 당해 기술분야에서 통상적인 방법에 따라 약학적으로 허용 가능한 염 및 용매화물로 제조될 수 있다.The compounds of the present invention can be prepared with pharmaceutically acceptable salts and solvates according to methods conventional in the art.

약학적으로 허용 가능한 염으로는 유리산(free acid)에 의해 형성된 산부가염이 유용하다. 산부가염은 통상의 방법, 예를 들면 화합물을 과량의 산 수용액에 용해시키고, 이 염을 메탄올, 에탄올, 아세톤 또는 아세토니트릴과 같은 수혼화성 유기 용매를 사용하여 침전시켜서 제조한다. 동일한 몰량의 화합물 및 물 중의 산 또는 알코올(예, 글리콜 모노메틸에테르)을 가열하고 이어서 상기 혼합물을 증발시켜서 건조시키거나, 또는 석출된 염을 흡인 여과시킬 수 있다.As the pharmaceutically acceptable salt, acid addition salts formed by free acid are useful. Acid addition salts are prepared by conventional methods, for example by dissolving a compound in an excess of aqueous acid solution and precipitating the salt using a water miscible organic solvent such as methanol, ethanol, acetone or acetonitrile. Equal molar amounts of the compound and acid or alcohol (eg, glycol monomethylether) in water can be heated and the mixture can then be evaporated to dryness or the precipitated salts can be suction filtered.

이 때, 유리산으로는 유기산과 무기산을 사용할 수 있으며, 무기산으로는 염산, 인산, 황산, 질산, 주석산 등을 사용할 수 있고 유기산으로는 메탄술폰산, p- 톨루엔술폰산, 아세트산, 트리플루오로아 세트산, 시트르산, 말레인산(maleic acid), 숙신산, 옥살산, 벤조산, 타르타르산, 푸마르산, 만데르산, 프로피온산(propionic acid), 구연산(citric acid), 젖산(lactic acid), 글리콜산(glycollic acid), 글루콘산(gluconic acid), 갈락투론산, 글루탐산, 글루타르산(glutaric acid), 글루쿠론산(glucuronic acid), 아스파르트산, 아스코르빈산, 카본산, 바닐릭산 및 히드로 아이오딕산 등을 사용할 수 있다.In this case, organic acids and inorganic acids may be used as the free acid, hydrochloric acid, phosphoric acid, sulfuric acid, nitric acid, tartaric acid, etc. may be used as the inorganic acid, and methanesulfonic acid, p -toluenesulfonic acid, acetic acid, trifluoroacetic acid, etc. may be used as the organic acid. , Citric acid, maleic acid, succinic acid, oxalic acid, benzoic acid, tartaric acid, fumaric acid, manderic acid, propionic acid, citric acid, lactic acid, glycolic acid, gluconic acid (gluconic acid), galacturonic acid, glutamic acid, glutaric acid (glutaric acid), glucuronic acid (glucuronic acid), aspartic acid, ascorbic acid, carbonic acid, vanic acid and hydroiodic acid may be used.

또한, 염기를 사용하여 약학적으로 허용 가능한 금속염을 만들 수 있다. 알칼리 금속 또는 알칼리토 금속염은, 예를 들면 화합물을 과량의 알칼리 금속 수산화물 또는 알칼리토 금속 수산화물 용액 중에 용해하고, 비 용해 화합물염을 여과한 후 여액을 증발, 건조시켜 얻는다. 이때, 금속염으로서는 특히 나트륨, 칼륨 또는 칼슘염을 제조하는 것이 제약상 적합하며, 또한 이에 대응하는 은염은 알칼리 금속 또는 알칼리토 금속염을 적당한 은염(예, 질산은)과 반응시켜 얻는다.In addition, bases can be used to make pharmaceutically acceptable metal salts. An alkali metal or alkaline earth metal salt is obtained by, for example, dissolving a compound in an excess alkali metal hydroxide or alkaline earth metal hydroxide solution, filtering the non-soluble compound salt, and then evaporating and drying the filtrate. At this time, as the metal salt, it is particularly suitable to prepare sodium, potassium or calcium salt, and the corresponding silver salt is obtained by reacting an alkali metal or alkaline earth metal salt with a suitable silver salt (for example, silver nitrate).

본 발명의 화합물의 약학적으로 허용 가능한 염은, 달리 지시되지 않는 한, 본 발명의 화합물에 존재할 수 있는 산성 또는 염기성기의 염을 포함한다. 예를 들면, 약학적으로 허용 가능한 염으로는 히드록시기의 나트륨, 칼슘 및 칼륨염이 포함되며, 아미노기의 기타 약학적으로 허용 가능한 염으로는 하이드로브로마이드, 황산염, 수소 황산염, 인산염, 수소 인산염, 이수소 인산염, 아세테이트, 숙시네이트, 시트레이트, 타르트레이트, 락테이트, 만델레이트, 메탄설포 네이트(메실레이트) 및 p-톨루엔설포네이트(토실레이트) 염이 있으며, 당업계에서 알려진 염의 제조 방법이나 제조과정을 통하여 제조될 수 있다. Pharmaceutically acceptable salts of the compounds of the invention include salts of acidic or basic groups which may be present in the compounds of the invention, unless otherwise indicated. For example, pharmaceutically acceptable salts include sodium, calcium and potassium salts of the hydroxy group, and other pharmaceutically acceptable salts of the amino group include hydrobromide, sulfate, hydrogen sulphate, phosphate, hydrogen phosphate, dihydrogen Phosphate, acetate, succinate, citrate, tartrate, lactate, mandelate, methanesulfonate (mesylate) and p -toluenesulfonate (tosylate) salts. It can be prepared through.

이하, 본 발명을 더욱 상세히 설명한다. Hereinafter, the present invention will be described in more detail.

본 발명의 화합물들은 하기와 같은 제조방법으로 수득될 수 있다. The compounds of the present invention can be obtained by the preparation method as follows.

예를 들어, 먼저, 본 발명의 천초근 조추출물을 수득하는 방법을 설명하면, 우선 천초근을 세척하여 음건한 다음 세절하여 마쇄기로 갈아 미세분말화한 후, 천초근 시료 중량의 약 1 내지 20배, 바람직하게는 약 1 내지 7배에 달하는 부피의 물 및 메탄올, 에탄올, 부탄올 등과 같은 C1 내지 C4의 저급알콜 또는 이들의 혼합용매로, 바람직하게는 메탄올, 더욱 바람직하게는 50 내지 100% 메탄올로 10℃ 내지 100℃, 바람직하게는 50℃ 내지 90℃에서 1 내지 20시간, 바람직하게는 5 내지 15시간동안 열수 추출, 냉침 추출, 환류 냉각 추출 또는 초음파 추출 등의 추출방법, 바람직하게는 환류추출방법을 사용하여, 진공여과에 의해 상층액을 회수한 다음, 상기의 과정을 2 내지 7회, 바람직하게는 4회 반복 수행하여 상층액을 모으고 감압농축하여 본 발명의 천초근 메탄올 조추출물 수득할 수 있다.For example, first, the method of obtaining the crude extract of Cheoncho root of the present invention will be described. First, the Cheoncho root is washed and shaded, and then finely pulverized into a crusher and then finely powdered. Pear, preferably about 1 to 7 times the volume of water and C1 to C4 lower alcohols such as methanol, ethanol, butanol and the like or a mixed solvent thereof, preferably methanol, more preferably 50 to 100% methanol Extraction method such as hot water extraction, cold needle extraction, reflux cooling extraction or ultrasonic extraction at 10 ° C. to 100 ° C., preferably 50 ° C. to 90 ° C. for 1 to 20 hours, preferably 5 to 15 hours, preferably reflux. Using the extraction method, the supernatant was recovered by vacuum filtration, and the above procedure was repeated 2 to 7 times, preferably four times, to collect the supernatant and concentrated under reduced pressure to obtain the supernatural root methane of the present invention. Crude extract can be obtained.

상기에서 수득한 메탄올 조추출물을 물 및 메틸렌클로라이드의 혼합용매에 혼합하여 분획한 후, 감압 농축하여 본 발명의 천초근 수 가용성 분획물 및 메틸렌클로라이드 가용성 분획물을 각각 수득하였고, 상기 메틸렌클로라이드 가용성 분획물을 헥산 및 메틸렌클로라이드 혼합용매를 극성을 증가시키는 방법을 이용하여 플래쉬 컬럼크로마토그래피, RP C18 컬럼크로마토그래피 또는 실리카겔 오픈 컬럼크로마토그래피 등의 크로마토그래피를 이용한 정제방법을 선택적으로 수회 반복 수행하여 본 발명의 화합물들 즉, 2-카르보메톡시-2,3-에폭시-3-프레닐-1,4-나프토퀴논 (2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone), 3,3′-비스(3,4-디 하이드로-4-하이드록시-6-메톡시-2H-1-벤조피란)(3,3′-bis(3,4-dihydro-4-hydroxy-6-methoxy-2H-1-benzopyran)), 에폭시몰루긴(Epoxymollugin), 소란지디올(Soranjidiol), 올레아놀릭 산(Oleanolic acid), 1-아세톡시-3-메톡시-9,10-안트라퀴논(1-acetoxy-3-methoxy-9,10-anthraquinone), 알리자린 2-메틸 아테르(Alizarin 2-methyl ether), 푸로놀루긴(Furonollugin), 몰루긴(mollugin)을 수득할 수 있다. The methanol crude extract obtained above was mixed with a mixed solvent of water and methylene chloride, and then fractionated. The mixture was concentrated under reduced pressure to obtain a cheoncho root water soluble fraction and a methylene chloride soluble fraction of the present invention, respectively. And methylene chloride mixed solvent using a method of increasing polarity to selectively purify the chromatography using chromatography, such as flash column chromatography, RP C18 column chromatography, or silica gel open column chromatography, to selectively perform the compounds of the present invention. That is, 2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone (3, carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone), 3, 3′-bis (3,4-dihydro-4-hydroxy-6-methoxy-2H-1-benzopyran) (3,3′-bis (3,4-dihydro-4-hydroxy-6-methoxy -2H-1-benzopyran)), epoxymollugin, small Soranjidiol, Oleanolic acid, 1-acetoxy-3-methoxy-9,10-anthraquinone, alizarin 2-methyl Alizarin 2-methyl ether, furonollugin, mollugin can be obtained.

본 발명은 상기의 제법으로 얻어진 천초근 추출물로부터 분리된 상기 화합물 또는 이의 약학적으로 허용 가능한 염을 유효성분으로 함유하는 염증성 질환의 예방 및 치료용 약학조성물을 제공한다.The present invention provides a pharmaceutical composition for the prevention and treatment of inflammatory diseases containing the compound or a pharmaceutically acceptable salt thereof isolated from the cheoncho root extract obtained by the above method as an active ingredient.

또한, 천초근은 오랫동안 식용되거나 생약으로 사용되어 오던 약재로서 본 발명의 천초근 추출물로부터 분리된 화합물 역시 독성 및 부작용 등의 문제가 없다. In addition, cheoncho root is a medicine that has been used for a long time as an edible or herbal medicine isolated from the cheoncho root extract of the present invention also has no problems such as toxicity and side effects.

본원에서 정의되는 추출물은 천초근의 조추출물, 극성용매 가용 추출물 또는 비극성용매 가용 추출물임을 특징으로 한다.The extract as defined herein is characterized in that crude extract of cheoncho root, polar solvent soluble extract or nonpolar solvent soluble extract.

본원에서 정의되는 상기 조추출물은 정제수를 포함한 물, 메탄올, 에탄올, 부탄올 등의 탄소수 1 내지 4의 저급알코올 또는 이들의 혼합용매로부터 선택된 용매, 바람직하게는 메탄올, 더욱 바람직하게는 50~100% 메탄올에 가용한 추출물을 포함한다.The crude extract as defined herein is a solvent selected from water containing purified water, lower alcohols having 1 to 4 carbon atoms such as methanol, ethanol, butanol, or a mixed solvent thereof, preferably methanol, more preferably 50 to 100% methanol Contains extracts available for use.

본원에서 정의되는 상기 비극성용매 가용 추출물은 메틸렌클로라이드, 헥산, 클로로포름, 디클로로메탄 또는 에틸아세테이트, 바람직하게는 메틸렌클로라이드에 가용한 추출물을 포함한다.The nonpolar solvent soluble extract as defined herein includes extracts soluble in methylene chloride, hexane, chloroform, dichloromethane or ethyl acetate, preferably methylene chloride.

본원에서 정의되는 염증성 질환은 상기 염증성 질환은 아토피피부염, 관절염, 요도염, 방광염, 동맥경화증, 알러지 질환, 비염, 천식, 급성통증, 만성통증, 치주염, 치은염, 염증성 장질환, 통풍, 심근경색, 울혈성 심부전, 고혈압, 협심증, 위궤양, 뇌경색, 다운증후군, 다발성 경화증, 비만, 당뇨, 치매, 우울증, 정신분열증, 결핵, 수면장애, 패혈증, 화상 또는 췌장염 등이며, 바람직하게는 아토피피부염, 관절염, 요도염, 방광염 또는 치주염이며, 더욱 바람직하게는 치주염, 요도염 또는 방광염인 것을 특징으로 한다.Inflammatory diseases defined herein are inflammatory diseases include atopic dermatitis, arthritis, urethritis, cystitis, arteriosclerosis, allergic diseases, rhinitis, asthma, acute pain, chronic pain, periodontitis, gingivitis, inflammatory bowel disease, gout, myocardial infarction, congestion Sexual heart failure, hypertension, angina pectoris, gastric ulcer, cerebral infarction, Down syndrome, multiple sclerosis, obesity, diabetes, dementia, depression, schizophrenia, tuberculosis, sleep disorders, sepsis, burns or pancreatitis, preferably atopic dermatitis, arthritis, urethritis , Cystitis or periodontitis, more preferably periodontitis, urethritis or cystitis.

본 발명의 염증성 질환의 예방 및 치료용 조성물은, 조성물 총 중량에 대하여 상기 화합물을 0.1 내지 50 중량 %로 포함한다. The composition for preventing and treating inflammatory diseases of the present invention comprises 0.1 to 50% by weight of the compound based on the total weight of the composition.

본 발명의 화합물을 포함하는 약학조성물은, 약학적 조성물의 제조에 통상적으로 사용하는 적절한 담체, 부형제 및 희석제를 더 포함할 수 있다.The pharmaceutical composition comprising the compound of the present invention may further include appropriate carriers, excipients and diluents commonly used in the preparation of pharmaceutical compositions.

본 발명의 조성물에 포함될 수 있는 담체, 부형제 및 희석제로는 락토즈, 덱스트로즈, 수크로스, 솔비톨, 만니톨, 자일 리톨, 에리스리톨, 말티톨, 전분, 아카시아 고무, 알지네이트, 젤라틴, 칼슘 포스페이트, 칼슘 실리케이트, 셀룰로즈, 메틸 셀룰로즈, 미정질 셀룰로스, 폴리비닐 피롤리돈, 물, 메틸히드록시벤조에이트, 프로필히드록 시벤조에이트, 탈크, 마그네슘 스테아레이트 및 광물유를 들 수 있다.Carriers, excipients and diluents that may be included in the compositions of the present invention include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia rubber, alginate, gelatin, calcium phosphate, calcium silicate , Cellulose, methyl cellulose, microcrystalline cellulose, polyvinyl pyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil.

본 발명의 화합물의 약학적 투여 형태는 이들의 약학적 허용 가능한 염의 형태로도 사용될 수 있고, 또한 단독으로 또는 타 약학적 활성 화합물과 결합뿐만 아 니라 적당한 집합으로 사용될 수 있다. Pharmaceutical dosage forms of the compounds of the present invention may be used in the form of their pharmaceutically acceptable salts, or may be used alone or in combination with other pharmaceutically active compounds, as well as in a suitable collection.

본 발명의 화합물을 포함하는 조성물은, 각각 통상의 방법에 따라 산제, 과립제, 정제, 캡슐제, 현탁액, 에멀젼, 시럽 및 에어로졸 등의 경구형 제형, 외용제, 좌제 및 멸균 주사용액의 형태로 제형화하여 사용될 수 있다. Compositions comprising the compounds of the invention are each formulated in the form of oral dosage forms, external preparations, suppositories, and sterile injectable solutions, such as powders, granules, tablets, capsules, suspensions, emulsions, syrups and aerosols, according to conventional methods Can be used.

상세하게는, 제제화할 경우에는 보통 사용하는 충진제, 증량제, 결합제, 습윤제, 붕해제, 계면활성제 등의 희석제 또는 부형제를 사용하여 조제될 수 있다. 경구투여를 위한 고형제제에는 정제, 환제, 산제, 과립제 및 캡슐제 등이 포함되며, 이러한 고형제제는 상기 화합물에 적어도 하나 이상의 부형제 예를 들면, 전분, 칼슘카보네이트 (calcium carbonate), 수크로스 (sucrose), 락토오스 (lactose) 및 젤라틴 등을 섞어 조제될 수 있다. 또한, 단순한 부형제 이외에 마그네슘 스테아레이트 및 탈크 같은 윤활제들도 사용될 수 있다. 경구를 위한 액상 제제로는 현탁제, 내용액제, 유제 및 시럽제 등이 해당되는데, 흔히 사용되는 단순 희석제인 물 및 리퀴드 파라핀 이외에 여러 가지 부형제, 예를 들면 습윤제, 감미제, 방향제 및 보존제 등이 포함될 수 있다. 비경구 투여를 위한 제제에는 멸균된 수용액, 비수성용제, 현탁제, 유제, 동결건조 제제 및 좌제가 포함된다. 비수성용제, 현탁제로는 프로필렌글리콜(propylene glycol), 폴리 에틸렌 글리콜 및 올리브 오일과 같은 식물성 기름 및 에틸올레이트와 같은 주사 가능한 에스테르 등이 사용 될 수 있다. 좌제의 기제로는 위텝솔 (witepsol), 마크로골, 트윈 (tween) 61, 카카오지, 라우린지 및 글리 세로젤라틴 등이 사용될 수 있다.More specifically, when formulating the composition, it can be prepared using a diluent or an excipient such as a filler, an extender, a binder, a wetting agent, a disintegrant, a surfactant, and the like. Solid form preparations for oral administration include tablets, pills, powders, granules and capsules, and the like form at least one excipient such as starch, calcium carbonate, sucrose in the compound. ), Lactose, gelatin and the like can be mixed. In addition to the simple excipients, lubricants such as magnesium stearate and talc may also be used. Liquid preparations for oral use include suspensions, solvents, emulsions and syrups, and may include various excipients, such as wetting agents, sweeteners, fragrances and preservatives, in addition to the commonly used simple diluents, water and liquid paraffin. have. Formulations for parenteral administration include sterilized aqueous solutions, non-aqueous solutions, suspensions, emulsions, freeze-dried preparations and suppositories. As the non-aqueous solvent and suspending agent, vegetable oils such as propylene glycol, polyethylene glycol and olive oil, and injectable esters such as ethyl oleate may be used. As the base of the suppository, witepsol, macrogol, tween 61, cacao butter, laurin butter and glycerogelatin may be used.

본 발명의 화합물의 바람직한 투여량은 환자의 상태 및 체중, 질병의 정도, 약물형태, 투여경로 및 기간에 따라 다르지만, 당업자에 의해 적절하게 선택될 수 있다. 그러나 바람직한 효과를 위해서, 본 발명의 화합물은 0.0001 ~ 100 mg/kg으로, 바람직하게는 0.00 1 ~ 100 mg/kg의 양을 일일 1회 내지 수회로 나누어 투여할 수 있다. 조성물에서 본 발명의 화합물은 전체 조성물 총 중량에 대하여 0.0001 ~ 50 중량%의 함량으로 배합될 수 있다.Preferred dosages of the compounds of the present invention depend on the condition and weight of the patient, the extent of the disease, the form of the drug, the route of administration and the duration, but may be appropriately selected by those skilled in the art. However, for the desired effect, the compound of the present invention may be administered in an amount of 0.0001 to 100 mg / kg, preferably in an amount of 0.00 1 to 100 mg / kg once or several times a day. The compound of the present invention in the composition may be formulated in an amount of 0.0001 to 50% by weight based on the total weight of the total composition.

본 발명의 약학 조성물은 쥐, 마우스, 가축, 인간 등의 포유동물에 다양한 경로로 투여될 수 있다. 투여의 모든 방식 은 예상될 수 있는데, 예를 들면, 경구, 직장 또는 정맥, 근육, 피하, 자궁내 경막 및 뇌혈관내 (intracere broventricular) 주사에 의해 투여될 수 있다. The pharmaceutical composition of the present invention can be administered to mammals such as mice, mice, livestock, humans, etc. by various routes. All modes of administration can be expected, for example by oral, rectal or intravenous, intramuscular, subcutaneous, intrauterine dural and intracere broventricular injections.

또한, 본 발명은 천초근 분리 화합물 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 염증성 질환의 예방 및 개선용 건강기능 식품을 제공한다. The present invention also provides a dietary supplement for the prevention and improvement of an inflammatory disease, which contains the mycelial root separating compound or a pharmaceutically acceptable salt thereof as an active ingredient.

본 발명의 화합물은 염증성 질환의 예방 및 치료를 위한 약제, 식품 및 음료 등에 다양하게 이용될 수 있다. 본 발명의 화합물을 첨가할 수 있는 식품으로는, 예를 들어, 각종 식품류, 음료, 껌, 차, 비타민 복합제 및 건강보조 식품류 등이 있고, 분말, 과립, 정제 및 캡슐 또는 음료인 형태로 사용할 수 있다.The compounds of the present invention can be used in various ways, such as drugs, food and beverages for the prevention and treatment of inflammatory diseases. Foods to which the compound of the present invention may be added include, for example, various foods, beverages, gums, teas, vitamin complexes and dietary supplements, and may be used in the form of powders, granules, tablets and capsules or beverages. have.

본 발명의 화합물은 독성 및 부작용은 거의 없으므로 예방 목적으로 장기간 복용 시에도 안심하고 사용할 수 있는 약제이다. Since the compound of the present invention has little toxicity and side effects, it is a drug that can be used safely even for long-term administration for the purpose of prevention.

본 발명의 상기 화합물은 염증성 질환의 예방 및 치료를 목적으로 식품 또는 음료에 첨가될 수 있다. 이 때, 식품 또는 음료 중의 상기 화합물의 양은 일반적으로 본 발명의 건강식품 조성물은 전체 식품 중량의 0.01 내지 15 중량%로 가할 수 있으며, 건강 음료 조성물은 100 ㎖를 기준으로 0.02 내지 30 g, 바람직하게는 0. 3 내지 10 g의 비율로 가할 수 있다. The compounds of the present invention may be added to food or beverages for the purpose of preventing and treating inflammatory diseases. At this time, the amount of the compound in the food or beverage is generally added to the health food composition of the present invention to 0.01 to 15% by weight of the total food weight, the health beverage composition is 0.02 to 30 g based on 100 ml, preferably Can be added at a rate of 0.3 to 10 g.

본 발명의 건강 음료 조성물은 지시된 비율로 필수 성분으로서 상기 화합물을 함유하는 것 외에 액체성분에는 특별한 제한점은 없으며 통상의 음료와 같이 여러 가지 향미제 또는 천연 탄수화물 등을 추가 성분으로서 함유할 수 있다. 상술한 천연 탄수화물의 예는 모노사카라이드, 예를 들어, 포도당, 과당 등의 디사카라이드, 예를 들어 말토스, 슈크로스 등의 폴리 사카라이드, 예를 들어 덱스트린, 시클로덱스트린 등과 같은 통상적인 당, 자일리톨, 소르비톨 및 에리트리톨 등의 당알코올이다. 상술한 것 이외의 향미제로서 천연 향미제(타우마틴, 스테비아 추출물(예를 들어 레바우디오시드 A, 글리시르히진 등) 및 합성 향미제(사카린, 아스파르탐 등)를 유리하게 사용할 수 있다. 상기 천연 탄수화물의 비율은 본 발명의 조성물 100 ㎖당 일반적으로 약 1 내지 20 g, 바람직하게는 약 5 내지 12 g이다.The health beverage composition of the present invention, in addition to containing the compound as an essential ingredient in the indicated proportions, has no particular limitation on the liquid component and may contain various flavors or natural carbohydrates as additional ingredients, such as ordinary drinks. Examples of the above-mentioned natural carbohydrates include monosaccharides such as disaccharides such as glucose and fructose, for example polysaccharides such as maltose and sucrose, and conventional sugars such as dextrin and cyclodextrin. Sugar alcohols such as xylitol, sorbitol and erythritol. As flavoring agents other than those described above, natural flavoring agents (tauumatin, stevia extract (e.g., Rebaudioside A, glycyrrhizin, etc.) and synthetic flavoring agents (saccharin, aspartame, etc.) can be advantageously used. The proportion of said natural carbohydrates is generally about 1-20 g, preferably about 5-12 g per 100 ml of the composition of the present invention.

상기 외에 본 발명의 화합물은 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. 그밖에 본 발명의 화합물은 천연 과일 쥬스 및 과일 쥬스 음료 및 야채 음료의 제조를 위한 과육을 함유할 수 있다. 이러한 성분은 독립적으로 또는 조합하여 사용할 수 있다. 이러한 첨가제의 비율은 그렇게 중요하진 않지만 본 발명의 조성물 100 중량부 당 0 내지 약 20 중량 부의 범위에서 선택되는 것이 일반적이 다.In addition to the above, the compounds of the present invention include 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. In addition, the compounds of the present invention may contain flesh for the production of natural fruit juices and fruit juice beverages and vegetable beverages. These components can be used independently or in combination. The proportion of such additives is not so critical but is usually selected in the range of 0 to about 20 parts by weight per 100 parts by weight of the composition of the present invention.

상기에 언급한 바와 같이, 본 발명의 천초근 추출물로부터 분리된 화합물은 마우스 대식세포인 RAW264.7 세포에서 LPS로 유도되어진 NO 생성량, p-IkBα, iNOS, 및 COX-2의 단백질 발현을 감소시킬 뿐만 아니라 iNOS, COX-2, TNF-α, IL-6, 및 IL-1β의 mRNA 발현과 같은 친염증성 사이토카인들의 발현 함량을 유의적으로 감소함을 확인함으로써, 상기 조성물은 염증성 질환의 예방 및 치료용 약학조성물 또는 건강기능식품으로 유용하게 이용할 수 있다.As mentioned above, the compounds isolated from the mycorrhizal extract of the present invention may reduce LPS-induced NO production, p-IkBα, iNOS, and COX-2 protein expression in RAW264.7 cells, which are mouse macrophages. In addition, by confirming that the expression content of pro-inflammatory cytokines such as mRNA expression of iNOS, COX-2, TNF-α, IL-6, and IL-1β significantly decreased, the composition prevents and prevents inflammatory diseases. It can be usefully used as a therapeutic pharmaceutical composition or health functional food.

이하, 본 발명을 하기 실시예 및 실험예에 의해 상세히 설명한다.Hereinafter, the present invention will be described in detail with reference to the following examples and experimental examples.

단, 하기 실시예 및 실험예는 본 발명을 예시하는 것일 뿐, 본 발명의 내용이 하기 실시예, 참고예 및 실험예에 의해 한정되는 것은 아니다.However, the following Examples and Experimental Examples are only illustrative of the present invention, the contents of the present invention is not limited by the following Examples, Reference Examples and Experimental Examples.

실시예 1. 천초근 조추출물의 제조Example 1. Preparation of Cultivated Root Extract

1-1. 천초근 조추출물의 제조1-1. Preparation of Cheoncho-Geun Crude Extract

대구광역시 중구 봉산동 약령시장에서 구입한 천초근을 세척하여 음건한 다음 마쇄기로 갈아 미세 분말화 한 후, 미세 분말 시료 10 kg에 95% 메탄올(MeOH) 40.5 ℓ를 가하여 80℃에서 8 시간 추출한 다음, 진공 여과하여 상층액을 회수하였 다. 이 과정을 4 회 반복하여 상층액을 모은 후, 감압 농축하여 천초근 메탄올 조추출물 1048.4 g을 수득하였다.After washing Cheoncho root purchased from Yangnyeong Market, Bongsan-dong, Jung-gu, Daegu Metropolitan City, dry it with a grinding machine and finely powder it. The supernatant was recovered by vacuum filtration. This process was repeated four times to collect the supernatant, and then concentrated under reduced pressure to obtain 1048.4 g of crude chondroitin extract.

1-2. 천초근 가용 조추출물의 제조1-2. Preparation of Crude Soluble Extracts

상기 실시예 1-1에서 얻은 천초근 메탄올 조추출물 1048.4g을 물 2.2 ℓ에 현탁시킨 후에, 메틸렌클로라이드 (CH2Cl2)로 용매 분획하였다. 먼저 물 2.2 ℓ에 현탁시킨 천초근 메탄올 가용 추출물에 메틸렌클로라이드 2 ℓ를 첨가하여 용해한 다음, 이를 분획여두에서 메틸렌클로라이드 층에 용해되는 성분만 분리해서 진공 건조하였다. 이 과정을 5 회 반복 수행하여 메틸렌클로라이드 가용 추출물 360 g을 수득하였다.1048.4 g of Cheoncho root methanol crude extract obtained in Example 1-1 was suspended in 2.2 L of water, followed by solvent fractionation with methylene chloride (CH 2 Cl 2 ). First, 2 liters of methylene chloride were added to the solubilized extract of Cheonchogeun methanol suspended in 2.2 liters of water, and then vacuum-dried by separating only the components dissolved in the methylene chloride layer in the fractionated filter. This process was repeated five times to give 360 g of methylene chloride soluble extract.

실시예 2. 천초근 추출물로부터 분리된 화합물의 분리Example 2. Isolation of Compound Isolated from Cheoncho Geun Extract

상기 실시예 1-2에서 수득한 메틸렌클로라이드 가용 추출물을 플래시 컬럼 크로마토그래피 (flash column chromatography; YMC Gel ODS-A, 60A, 220 mesh)로 즉, 길이 75 cm, 직경 9 cm의 컬럼에 230~400 메쉬 (mesh) 실리카켈 (silica gel)을 채우고 100% 헥산 (Hexane)으로 용리 (elution)하여 고정상 (stationary phase)를 균일한 상태로 만든 후, 상기 실시예 1-2에서 수득한 메틸렌클로라이드 가용성 추출물 360g을 실리카겔 (70~230 메쉬) 720g에 흡착시켜 컬럼에 로딩하였다. 이어서, 헥산 : 메틸렌클로라이드 = 98 : 2, 95 : 5, 90 : 10, 85 : 15, 80 : 20, 60 : 40, 50 : 50 순으로 이동상 (mobilie phase)의 극성을 높이다가 메틸렌클로라이드 : 메탄올 = 99 : 1, 98 : 2, 96 : 4로 용리 (elution)시켜 TLC패턴을 기준으로 RA-MC-1 ~ RA-MC-34의 분획물을 분획하였다. The methylene chloride soluble extract obtained in Example 1-2 was subjected to flash column chromatography (YMC Gel ODS-A, 60A, 220 mesh), that is, 230 to 400 in a column having a length of 75 cm and a diameter of 9 cm. After filling the mesh silica gel and eluting with 100% hexane to make the stationary phase uniform, the methylene chloride soluble extract obtained in Example 1-2 was obtained. 360 g was adsorbed onto 720 g of silica gel (70-230 mesh) and loaded onto the column. Then, increase the polarity of the mobile phase in the order of hexane: methylene chloride = 98: 2, 95: 5, 90: 10, 85: 15, 80: 20, 60: 40, 50: 50, and then methylene chloride: methanol Elution was performed at 99: 1, 98: 2, and 96: 4 to fractionate fractions of RA-MC-1 to RA-MC-34 based on the TLC pattern.

상기 분획물 중에 열다섯번째 분획물인 RA-MC-15 분획물을 RP C18 컬럼 크로마토그래피 (RP C18 column chromatography)를 실시하였으며 컬럼 (I.D. 57 cm X 3.5 cm)에 RP C18 (40~63㎛)을 약 22 cm 채우고 CH3CN : H2O = 3 : 7 혼합용액으로 용리하여 균일한 상채로 만든 후 RA-MC-15 분획 6.2 g을 100% CH3CN에 완전히 녹여 로딩하였고, 이어서 CH3CN의 비율을 단계적으로 높이면서 용리시켜서 얻은 일곱 번째 분획물인 RA-MC-15-7 분획물 약 1.2 g을 다시 규소 오픈 컬럼 크로마토그래피 (SiO2 open column chromatography)를 실시하였다. 컬럼 (I.D. 53 cm X 2.3 cm)에 실리카겔 (70 메쉬이하)을 약 15 cm 정도 채우고 100% 헥산으로 용리시켜 균일한 상태로 만들고 샘플을 실리카겔 (70 메쉬 이하) 약 0.72 g에 흡착시켜 컬럼에 로딩하고 Hexane : EtOAc = 18 : 0.3 혼합용액으로 용리시켜 하기 표 1에 나타낸 바와 같이, 본 발명의 화합물 2-카르보메톡시-2,3-에폭시-3-프레닐-1,4-나프토퀴논 (2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone) 754 mg을 분리 정제하여 수득하였다. 또한 상기 분획물 중에 RA-MC 분획물을 사용하여 상기와 동일한 방법으로 본 발명의 화합물들인 3‘-비스(3,4-디하이드로-4-하이드록시-6-메톡시-2H-1-벤조피란)(3,3’-bis(3,4-dihydro-4-hydroxy-6-methoxy-2H-1-benzopyran)) 51 mg, 에폭시몰루긴 (Epoxymollugin) 36.4 mg, 소란지디올 (Soranjidiol) 30 mg, 올레아놀 릭 산(Oleanolic acid) 28 mg, 1-아세톡시-3-메톡시-9,10-안트라퀴논 (1-acetoxy-3-methoxy-9,10-anthraquinone) 31 mg, 알리자린 2-메틸 아테르 (Alizarin 2-methyl ether) 42 mg, 푸로몰루긴 (Furomollugin) 76 mg, 몰루긴 (Mollugin) 670 mg 을 차례로 분리 정제하여 수득하였다 (표 1 참조).The fifteenth fraction, RA-MC-15, was subjected to RP C18 column chromatography and RP C18 (40-63 μm) was added to the column (ID 57 cm × 3.5 cm). After filling in cm, eluting with a mixture of CH 3 CN: H 2 O = 3: 7 to make a uniform solution, 6.2 g of RA-MC-15 fraction was completely dissolved in 100% CH 3 CN and loaded, followed by the ratio of CH 3 CN. a step-by-step to the seventh fraction is RA-MC-15-7 fractions, about 1.2 g of silicon again open column chromatography (SiO 2 open column chromatography) obtained by the elution was carried out while increasing. Fill the column (ID 53 cm x 2.3 cm) with silica gel (less than 70 mesh) about 15 cm, elute with 100% hexane to make a homogeneous state, and sample is adsorbed to about 0.72 g of silica gel (less than 70 mesh) and loaded onto the column And eluting with a Hexane: EtOAc = 18: 0.3 mixed solution, as shown in Table 1 below, the compound 2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone ( 754 mg of 2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone) was obtained by separation and purification. In addition, 3'-bis (3,4-dihydro-4-hydroxy-6-methoxy-2H-1-benzopyran) of the compounds of the present invention in the same manner as described above using the RA-MC fraction in the fraction (3,3'-bis (3,4-dihydro-4-hydroxy-6-methoxy-2H-1-benzopyran)) 51 mg, Epoxymollugin 36.4 mg, Soranjidiol 30 mg, 28 mg oleanolic acid, 31 mg 1-acetoxy-3-methoxy-9,10-anthraquinone, alizarin 2-methyl ether 42 mg of Alizarin 2-methyl ether), 76 mg of furomollugin, and 670 mg of molugin were obtained by sequentially separating and purifying them (see Table 1).

천초근 추출물로부터 분리되는 16가지의 화합물16 Compounds Isolated from Cheoncho Geun Extract PlantPlant FractionFraction Chemical compound nameChemical compound name 천초근
(Rubiae Radix)
Cheoncho Geun
(Rubiae Radix)
RA-MC-AARA-MC-AA 2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone
RA-MC-BBRA-MC-BB 3,3′-bis(3,4-dihydro-4-hydroxy-6-methoxy-2H-1-benzopyran)3,3′-bis (3,4-dihydro-4-hydroxy-6-methoxy-2H-1-benzopyran) RA-MC-CCRA-MC-CC EpoxymolluginEpoxymollugin RA-MC-DDRA-MC-DD SoranjidiolSoranjidiol RA-MC-EERA-MC-EE Oleanolic acidOleanolic acid RA-MC-FFRA-MC-FF 1-acetoxy-3-methoxy-9,10-anthraquinone1-acetoxy-3-methoxy-9,10-anthraquinone RA-MC-GGRA-MC-GG Alizarin 2-methyl etherAlizarin 2-methyl ether RA-MC-HaRA-MC-Ha FuronolluginFuronollugin RA-MC-HbRA-MC-Hb molluginmollugin

참고예 1. 실험재료의 준비Reference Example 1. Preparation of Experimental Materials

DMEM(Dulbecco's Modified Eagle Medium)과 FBS(fetal bovine serum)은 Hyclone사 (Gaithersbug, MD, USA)에서 구입하였으며, LPS (Lipopolysaccharide), Griess reagent, MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide)는 Sigma 사(St. Louis, Mo, USA)에서 구입하였다. BCA 단백질 분석 시약(BCA Protein assay reagent)은 Pierce사 (Rockford, IL, USA)에서 구입하였고, ECL 웨스턴 블롯 시약은 Amersham사 (Arlington Heights, IL, USA)에서 구입하였다. 그밖의 실험에 사용된 1차 항체인 anti-IkBα, anti-p-Ikbα monoclonal antibody는 Cell signaling 사에서 구입하였고, mAb (iNOS monoclonal antibody)는 BD science 사에서, COX-2 antibody는 Santa Cruz Biotechnology사에서 구입하였으며, 2차 항체인 anti-mouse 또는 anti-goat IgG horseradish peroxidase (HRP)-conjugated antibody는 Amersharm사 (Arlington Heights, IL, USA)에서 구입하였다. 실험에 사용된 모든 시약은 분석용 등급이상으로 사용하였다.DMEM (Dulbecco's Modified Eagle Medium) and FBS (fetal bovine serum) were purchased from Hyclone (Gaithersbug, MD, USA), LPS (Lipopolysaccharide), Griess reagent, MTT (3- (4,5-dimethylthiazol-2-yl) ) -2,5-diphenyl-tetrazolium bromide) was purchased from Sigma (St. Louis, Mo, USA). BCA Protein assay reagent was purchased from Pierce (Rockford, IL, USA), and ECL Western Blot reagent was purchased from Amersham (Arlington Heights, IL, USA). Other primary anti-IkBα and anti-p-Ikbα monoclonal antibodies used in other experiments were purchased from Cell signaling, mAb (iNOS monoclonal antibody) from BD Science, and COX-2 antibody from Santa Cruz Biotechnology. A secondary antibody, anti-mouse or anti-goat IgG horseradish peroxidase (HRP) -conjugated antibody, was purchased from Amersharm (Arlington Heights, IL, USA). All reagents used in the experiments were used above the analytical grade.

참고예 2. 세포배양Reference Example 2. Cell Culture

마우스의 대식세포주인 RAW264.7 세포는 ATCC(American Type Culture Collection, NO. TIB-71)에서 분양받아서, 10% FBS과 100 μg/ml의 겐타마이신(gentamycin)을 포함하는 DMEM (Dulbecco's Modified Eagle Medium) 배지에 37℃, 5% CO2의 조건으로 배양하여 하기 실험에 사용하였다. RAW264.7 cells, a macrophage line in mice, were distributed from the American Type Culture Collection (NOCC.TIB-71) and contained Dulbecco's Modified Eagle Medium containing 10% FBS and 100 μg / ml gentamycin. ) Was cultured in a medium at 37 ℃, 5% CO 2 conditions were used in the following experiment.

실험예 1. 세포독성 측정Experimental Example 1. Measurement of cytotoxicity

상기 실시예 2에서 얻은 화합물들의 세포독성을 측정하기 위하여 MTT assay 방법을 이용하여 하기와 같은 실험을 하였다 (Jun DY, et al., Apoptogenic activity of Zanthoxylum schinifolium toward human acute leukemia Jurkat T cells is associated with ER stress-mediated caspase-8 activation that stimulates mitochondria- dependent or -independent caspase cascade, Carcinogenesis, 28, pp.1303-1313, 2007).In order to measure the cytotoxicity of the compounds obtained in Example 2 was performed using the MTT assay method (Jun DY, et al., Apoptogenic activity of Zanthoxylum schinifolium toward human acute leukemia Jurkat T cells is associated with ER stress-mediated caspase-8 activation that stimulates mitochondria-dependent or -independent caspase cascade, Carcinogenesis, 28 , pp.1303-1313, 2007).

세포독성 분석을 위해 96-웰 플레이트(96-well plate)에 세포농도 2 × 105 cells/well로 조절하여 RAW264.7 세포를 분주한 후 16시간 배양하고, 상기 실시예 2의 화합물들을 농도별로 1 시간 전처리하고 LPS 0.1 μg/ml을 처리한 후 12 시간 동안 배양한 후 50 μl의 MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-dip henyl tetrazolium bromide]를 첨가하고 4 시간 반응시켜 포르마잔(formazan)을 형성시킨 후, 2300 rpm에서 15분 원심 분리하여 상등액을 제거하고 DMSO 150 μl/well을 가하여 포르마잔을 녹인 다음 ELISA reader(Variokan, Thermo Electron) 를 이용하여 540 nm 파장에서 측정하였다.For cytotoxicity analysis, the cell concentration was adjusted to a 96-well plate (96-well plate) at 2 × 10 5 cells / well, and the RAW264.7 cells were dispensed and cultured for 16 hours. After pretreatment for 1 hour, treatment with 0.1 μg / ml LPS, incubation for 12 hours, and then 50 μl of MTT [3- (4,5-dimethylthiazol-2-yl) -2,5-dip henyl tetrazolium bromide] was added. After reaction for 4 hours to form formazan (formazan), centrifugation at 2300 rpm for 15 minutes to remove the supernatant, and dissolved formazan by adding 150 μl / well DMSO and 540 using ELISA reader (Variokan, Thermo Electron) Measured at nm wavelength.

실험결과, 표 2 및 도 1에서 나타내는 바와 같이 6.25 μg/ml 농도의 화합물 처리에 의해서는 RAW264.7 세포에 대한 세포생존율이 크게 영향을 받지 않았으나, RA-MC-AA 화합물의 경우, 12.5 μg/ml 농도에서는 세포생존율이 대조구에 비해 23.2% 정도로 감소되는 것으로 나타나 12.5 μg/ml 농도에서는 대식세포 RAW264.7 세포에 대하여 세포독성 효과가 있음을 확인하였다(표 2 및 도 1 참조). As a result, as shown in Table 2 and FIG. 1, the cell viability of RAW264.7 cells was not significantly affected by the compound treatment of 6.25 μg / ml concentration, but in the case of RA-MC-AA compound, 12.5 μg / ml Cell viability was reduced by 23.2% compared to the control at ml concentration, it was confirmed that there is a cytotoxic effect on macrophage RAW264.7 cells at 12.5 μg / ml concentration (see Table 2 and Figure 1).

분획물Fraction 세포 생존율 (Cell viability, %)Cell viability (%) 6.25 μg/ml6.25 μg / ml 12.5 μg/ml12.5 μg / ml RA-MC-AARA-MC-AA 8585 2323 RA-MC-BBRA-MC-BB 9393 112112 RA-MC-CCRA-MC-CC 108108 111111 RA-MC-DDRA-MC-DD 112112 110110 RA-MC-EERA-MC-EE 110110 9393 RA-MC-FFRA-MC-FF 106106 110110 RA-MC-GGRA-MC-GG 108108 102102 RA-MC-HaRA-MC-Ha 108108 109109 RA-MC-HbRA-MC-Hb 120120 115115 LPS 단독 투여군LPS-only group 100100 대조군 (No treatment)Control (No treatment) 114114

실험예 2. NO의 생성량 측정Experimental Example 2 Measurement of NO Production

NO의 생성량을 측정하기 위해 문헌 (Wang S et al., J. Ethnopharmacol., 114(3), pp.458-462, 2007)에 기재되어 있는 방법을 이용하여 하기와 같이 실험하였다.In order to determine the amount of NO produced, experiments were conducted using the method described in Wang S et al., J. Ethnopharmacol. , 114 (3) , pp. 458-462, 2007.

NO생성량 측정을 위해 96-웰 플레이트(96-well plate)에 세포농도 2 × 10 5 cells/well 로 희석하여 RAW264.7 세포를 분주한 후 16 시간 배양하고, 상기 실시예 2의 화합물을 농도별로 1 시간 전처리하고, LPS 0.1 μg/ml을 처리한 후 16 시간 동안 배양한 후 상등액 100 μl를 회수하여 그리스 반응액 (Griess Reagent) 100μl을 넣어주고 15분간 상온에서 빛을 차단한 상태로 반응 시킨 후 Automatic ELISA reader (precision microplate reader, Molecular Devices, BIO-TEX EL800uv-PC, USA)를 이용하여 540 nm에서 흡광도를 측정하였다. 아질산 나트륨 (sodium nitrite)의 농도별 표준곡선을 이용하여 배양액 내의 NO 농도를 결정하였다.In order to measure NO production, the cells were diluted in a 96-well plate with a cell concentration of 2 × 10 5 cells / well, and the RAW264.7 cells were dispensed and incubated for 16 hours, and the compound of Example 2 was prepared by concentration. After pretreatment for 1 hour, incubated for 16 hours after treatment with 0.1 μg / ml LPS, 100 μl of the supernatant was collected, 100 μl of Greases Reagent was added, and reacted with light at room temperature for 15 minutes. Absorbance was measured at 540 nm using an Automatic ELISA reader (precision microplate reader, Molecular Devices, BIO-TEX EL800uv-PC, USA). NO concentration in the culture was determined using a standard curve of concentration of sodium nitrite.

실험결과, 표 3 및 도 2에 나타난 바와 같이 RA-MC-AA 화합물은 6.25 μg/ml 농도조건에서 마우스 대식세포주 RAW264.7의 NO 생성은 거의 99% 정도로 현저히 저해하는 효과를 나타내었으므로(표 3 및 도 2 참조), RAW264.7 세포의 NO 생성을 저해할 수 있는 최소농도를 규명하기 위하여 RA-MC-AA 화합물을 1 μg/ml, 2 μg/ml 또는 4 μg/ml 농도로 첨가하는 경우에도 LPS에 의한 RAW264.7 세포의 NO 생성이 저해되는지를 확인하였다. Experimental results, as shown in Table 3 and Figure 2 RA-MC-AA compound showed a significant inhibitory effect of the NO production of the mouse macrophage RAW264.7 almost 99% at 6.25 μg / ml concentration conditions (Table 3 and FIG. 2), adding RA-MC-AA compounds at a concentration of 1 μg / ml, 2 μg / ml or 4 μg / ml to identify the minimum concentration that can inhibit NO production in RAW264.7 cells. In addition, it was confirmed whether NO production of RAW264.7 cells by LPS was inhibited.

그 결과, 도 3-A에서 나타내는 바와 같이 1 μg/ml, 2 μg/ml 또는 4 μg/ml 농도 조건에서는 LPS에 의한 RAW264.7 세포의 NO 생성이 농도 의존적으로 저해되어 58%, 82% 및 99%로 각각 저해 되는 것으로 확인하였으며(도 3-A 참조), 동일한 농도조건에서 MTT assay로 확인한 RA-MC-AA 화합물은 도 3-B에서 나타내는 바와 같이 세포독성이 나타나지 않음을 확인하였다(도 3-B 참조).As a result, as shown in Fig. 3-A, NO production of RAW264.7 cells by LPS was inhibited at a concentration of 1 μg / ml, 2 μg / ml or 4 μg / ml, resulting in 58%, 82% and It was confirmed that each inhibited by 99% (see Figure 3-A), RA-MC-AA compound confirmed by the MTT assay under the same concentration conditions was confirmed that no cytotoxicity as shown in Figure 3-B (Fig. 3-B).

FractionFraction NO 생성 저해율 (NO synthesis inhibition, %)NO synthesis inhibition (%) 6.25 μg/ml6.25 μg / ml 12.5 μg/ml12.5 μg / ml RA-MC-AARA-MC-AA 9999 100100 RA-MC-BBRA-MC-BB 88 1010 RA-MC-CCRA-MC-CC 33 77 RA-MC-DDRA-MC-DD 66 44 RA-MC-EERA-MC-EE 1919 4040 RA-MC-FFRA-MC-FF 77 55 RA-MC-GGRA-MC-GG 77 66 RA-MC-HaRA-MC-Ha 1010 66 RA-MC-HbRA-MC-Hb 4343 9292 LPS 단독 투여군LPS-only group 00

실험예 3. 웨스턴 블롯 분석법 (Western blot analysis)Experimental Example 3. Western blot analysis

Ik Bα, p-IkBα, COX-2 또는 iNOS 단백질 발현 측정을 하기 위해 허 등의 방법 (Heo et al., J. Immunol., 179(9), pp.6305-6310, 2007)을 이용하여 웨스턴 블롯 분석법을 하기와 같이 수행하였다. To determine the expression of Ik Bα, p-IkBα, COX-2, or iNOS proteins, Western using the method of Hu et al. (Heo et al., J. Immunol., 179 (9) , pp.6305-6310, 2007) Blot analysis was performed as follows.

참고예 2의 방법으로 배양된 4 × 106 세포에 PBS(ice-cold phosphate buffered saline: 2.7 mM KCl, 137 mM NaCl, 10 mM Na2HPO4, 2 mM KH2PO4, pH 7.4)으로 3회 세척한 후, 용해 완충액 (lysis buffer: 137 mM NaCl, 15 mM EGTA, 1 mM sodium orthovanadate, 15 mM MgCl2, 0.1 % Triton X-100, 25mM MOPS, 2.5 μg/ml proteinase inhibitor E64, pH 7.2)으로 현탁하고 초음파 분쇄기로 파쇄한 다음, 4℃에서 30분간 반응시키고 14000rpm에서 20분간 원심 분리하여 상층액을 모았다. 동일한 양의 단백질을 4~12% SDS-PAGE(sodium dodecyl sulfate-polyacrylamide gel electrophoresis)로 분리시킨 후, 단백질을 Immobilon-P 멤브레인(membrane)에 전이 (transfer)하였다. 이 멤브레인을 항체의 비특이적 결합을 차단하기 위하여 블로킹 용액 (blocking buffer: 3% non-fat milk와 0.1% Tween 20을 함유한 TBS 용액)에서 1시간 동안 반응시킨 후, 각 단백질에 대한 항체 (anti-iNOS 및 anti-COX2)를 가하여 1 내지 2 시간 동안 반응시켰다. 이어서 0.1% Tween 20을 함유한 TBST 용액으로 10 분씩 3회 세척한 다음, 2차 항체 (anti-mouse 또는 anti-rabbit IgG horseradish peroxidase-conjugated antibody)로 90 분에서 2 시간 동안 반응시켰다. 이어서 ECL 시스템으로 반응 시킨 후에 X-ray 필름 (AGFA, Belgium) 상에서 단백질을 확인하였다. 각 시료의 단백질 정량은 Micro BCA 단백질 에세이키트 (Micro BCA protein assay kit)를 사용하여 562 nm에서 흡광도를 측정하여 실시하였다.PBS (ice-cold phosphate buffered saline: 2.7 mM KCl, 137 mM NaCl, 10 mM Na 2 HPO 4 , 2 mM KH 2 PO 4 , pH 7.4) was added to 4 × 10 6 cells cultured by the method of Reference Example 2. After washing several times, with lysis buffer (lysis buffer: 137 mM NaCl, 15 mM EGTA, 1 mM sodium orthovanadate, 15 mM MgCl2, 0.1% Triton X-100, 25 mM MOPS, 2.5 μg / ml proteinase inhibitor E64, pH 7.2) The suspension was crushed with an ultrasonic mill, and then reacted at 4 ° C. for 30 minutes and centrifuged at 14000 rpm for 20 minutes to collect the supernatant. The same amount of protein was separated by 4-12% SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis), and then the protein was transferred to an Immobilon-P membrane (membrane). The membrane was reacted for one hour in a blocking buffer (TBS solution containing 3% non-fat milk and 0.1% Tween 20) to block nonspecific binding of the antibody, followed by antibody to each protein (anti- iNOS and anti-COX2) were added and reacted for 1-2 hours. Subsequently, the cells were washed three times for 10 minutes with a TBST solution containing 0.1% Tween 20, and then reacted with a secondary antibody (anti-mouse or anti-rabbit IgG horseradish peroxidase-conjugated antibody) for 90 minutes to 2 hours. The protein was then identified on X-ray film (AGFA, Belgium) after reaction with the ECL system. Protein quantification of each sample was performed by measuring the absorbance at 562 nm using a Micro BCA protein assay kit.

참고로, 대식세포는 염증반응의 개시와 확대에서 중요한 역할을 하는 세포로서, 세균성 LPS에 의해 세포표면의 Toll-유사 수용체 (Toll-like receptor)가 자극되는 대식세포는 세포내 신호전달과정을 통해 전사인자인 NF-κB를 활성화하며, 이어서 활성화된 NF-κB의 작용으로 유도성 산화질소생성효소 (iNOS)의 발현을 증가시킨다. 그 결과, 세포내에서 증가된 iNOS의 촉매작용으로 L-아르기닌(L-arginine)으로부터 염증매개체 중의 하나인 NO의 생성이 증진됨을 알 수 있다(Haiqi He, et al., Molecular Immunology, 43, p.783, 2006). For reference, macrophages play an important role in the initiation and expansion of inflammatory responses. Macrophages that stimulate Toll-like receptors on the cell surface by bacterial LPS are expressed through intracellular signaling. It activates the transcription factor NF-κB, which in turn increases the expression of inducible nitric oxide synthase (iNOS) by the action of activated NF-κB. As a result, it can be seen that the production of NO, one of the inflammatory mediators from L-arginine, is enhanced by catalysis of increased iNOS in cells (Haiqi He, et al., Molecular Immunology, 43 , p. .783, 2006).

실험결과, 도 4에 나타난 바와 같이, LPS의 처리에 의해 전사인자 NF-κB의 활성화와 관련된 iNOS의 발현 및 COX-2의 발현 수준은 RA-MC-AA 화합물에 의해 농도 의존적으로 감소되는 경향을 나타내었으며(도 4 참조), As shown in FIG. 4, the expression of iNOS and the level of COX-2 associated with activation of the transcription factor NF-κB by LPS treatment tended to decrease concentration-dependently by RA-MC-AA compounds. (See FIG. 4),

또한, 도 5에 나타난 바와 같이 동일한 조건하에서 NF-κB의 활성화와 관련된 IκB 단백질의 인산화는 현저히 증가하였으나 RA-MC-AA 화합물의 존재하는 조건에서는 농도 의존적으로 IκB의 인산화가 현저히 감소함을 확인하였다(도 5 참조).In addition, as shown in FIG. 5, phosphorylation of IκB protein associated with NF-κB activation was significantly increased under the same conditions, but it was confirmed that phosphorylation of IκB was significantly reduced in the presence of RA-MC-AA compound. (See Figure 5).

실험예 4. 역전사중합효소연쇄반응(RT-PCR)Experimental Example 4 Reverse Transcriptase Chain Reaction (RT-PCR)

RA-MC-AA 화합물의 NO 생성 억제능이 iNOS mRNA 및 친염증성 사이토카인들의 발현 억제에 의한 것인지 확인하기 위해 기존의 문헌에 기재된 방법 (Lee et al., J. Ethnopharmacol., 97, pp.561-566, 2005)을 이용하여 RT-PCR을 하기와 같이 수행하였다. The method described in the existing literature to determine whether the inhibition of NO production of RA-MC-AA compounds is due to the inhibition of the expression of iNOS mRNA and proinflammatory cytokines (Lee et al., J. Ethnopharmacol., 97 , pp.561- 566, 2005) was performed as follows.

RAW264.7 세포에 실시예 2의 화합물 RA-MC-AA(2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone)을 1~4 μg/ml의 농도로 1 시간 처리하여 반응시킨 후, 0.1 μg/ml의 LPS 4시간 전처리하여 NO 생성을 유도하였다. LPS 처리하고 4시간 경과 후, 세포를 트립신(Trypsin)-EDTA를 처리하여 회수하고 total RNA를 트리졸(TRIzole: Invitrogen, Carlsbad, CA, USA)을 사용하여 분리하였다. 첫 번째 가닥 cDNA는 총 RNA의 2μg으로부터 MMLV-역전사 효소(reverse transcriptase: Gibco BRL)를 사용하여 합성하였다. 프라이머(Primer)의 염기서열은 하기의 표 4에 나타내었다. RAW264.7 cells were treated with compound RA-MC-AA (2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone) of Example 2 at a concentration of 1 to 4 μg / ml for 1 hour. After the reaction, 0.1 μg / ml of LPS was pretreated for 4 hours to induce NO production. After 4 hours of LPS treatment, cells were recovered by treatment with Trypsin-EDTA and total RNA was isolated using Trizole: Invitrogen, Carlsbad, CA, USA. The first strand cDNA was synthesized from 2 μg of total RNA using MMLV-reverse transcriptase (Gibco BRL). The base sequence of the primer (Primer) is shown in Table 4 below.

genesgenes oligonucleotide sequenceoligonucleotide sequence size size iNOSiNOS sensesense 5-ATGTCCGAAGCAAACATCAC-35-ATGTCCGAAGCAAACATCAC-3 449 bp449 bp antisenseantisense 5-TAATGTCCAGGAAGTAGGTG-35-TAATGTCCAGGAAGTAGGTG-3 COX-2COX-2 sensesense 5-CAGCAAATCCTTGCTGTTCC-3′5-CAGCAAATCCTTGCTGTTCC-3 ′ 490 bp490 bp antisenseantisense 5-TGGGCAAAGAATGCAAACATC-35-TGGGCAAAGAATGCAAACATC-3 TNF-αTNF-α sensesense 5-TACTGAACTTCGGGGTGATCGGTCC-35-TACTGAACTTCGGGGTGATCGGTCC-3 295 bp295 bp antisenseantisense 5-CAGCCTTGTCCCTTGAAGAGAACC-35-CAGCCTTGTCCCTTGAAGAGAACC-3 IL-6IL-6 sensesense 5-GAAATGATGGATGCTTCCAAACTGG-35-GAAATGATGGATGCTTCCAAACTGG-3 414 bp414 bp antisenseantisense 5-GGATATATTTTCTGACCACAGTGAGG-35-GGATATATTTTCTGACCACAGTGAGG-3 IL-1βIL-1β sensesense 5-CAAGGAGAACCAAGCAAC-35-CAAGGAGAACCAAGCAAC-3 350 bp350 bp antisenseantisense 5-GGGGAAGGCATTAGAAAC-35-GGGGAAGGCATTAGAAAC-3 GAPDHGAPDH sensesense 5-CCACTGGCGTCTTCACCAC-35-CCACTGGCGTCTTCACCAC-3 349 bp349 bp antisenseantisense 5-CCTGCTTCACCACCTTTTG-35-CCTGCTTCACCACCTTTTG-3

PCR은 98 ℃에서 2 분간 가열한 후 98 ℃에서 10 초, 50 ℃에서 30 초, 72 ℃에서 1 분간 반응하는 순서로 20 회 반복하고, 이후에 72 ℃에서 5 분간 반응하였다. PCR 생성물은 1.2% 아가로스 젤 (agarose gel)에서 전기영동하고 EtBr(ethidium bromide: 0.5 μg/ml)로 염색하여 확인하였다. PCR was repeated 20 times in order of heating at 98 ° C. for 2 minutes and then reacting at 98 ° C. for 10 seconds, at 50 ° C. for 30 seconds, and at 72 ° C. for 1 minute, and then at 5 ° C. for 5 minutes. PCR products were confirmed by electrophoresis on 1.2% agarose gel and stained with EtBr (ethidium bromide: 0.5 μg / ml).

실험결과, 도 6에서 나타내는 바와 같이 LPS를 처리한 RAW264. 7 세포에서는 iNOS mRNA 발현 수준이 현저히 증가하였으나 RA-MC-AA 화합물와 LPS를 함께 처리한 경우에는 RA-MC-AA 화합물의 농도 의존적으로 iNOS와 COX-2 mRNA 수준이 감소하는 것을 확인할 수 있었으며, 친염증성 사이토카인인 TNF-α, IL-6 및 IL-1β mRNA 수준도 LPS를 처리한 RAW264. 7 세포에서 현저히 증가하였으나, RA-MC-AA 화합물와 LPS를 함께 처리한 경우에는 RA-MC-AA 화합물에 의해 농도 의존적으로 감소됨을 확인하였다(도 6 참조). As a result of the experiment, RAW264 processed LPS as shown in FIG. INOS mRNA expression level was significantly increased in 7 cells. However, when RA-MC-AA compound and LPS were treated together, iNOS and COX-2 mRNA levels decreased depending on the concentration of RA-MC-AA compound. Inflammatory cytokines TNF-α, IL-6 and IL-1β mRNA levels were also treated with LPS treated RAW264. Although significantly increased in 7 cells, when the RA-MC-AA compound and LPS were treated together, it was confirmed that the concentration was decreased depending on the RA-MC-AA compound (see FIG. 6).

따라서, 마우스 대식세포주인 RAW264.7 세포에 있어서 LPS 처리에 의해 유도되는 NO 생성에 대한 RA-MC-AA 화합물의 저해효과는 iNOS 유전자의 발현과 관련된 전사인자 NF-κB의 활성화 억제를 통해서 이루어짐을 시사하며, 이러한 NF-κB의 활성화 억제는 NF-κB의 저해인자인 IkB 단백질의 불활성화 기전인 그 인산화를 방해하여 일어나는 것임을 나타낸다. LPS 처리에 의해 대식세포 내에서 유도되는 전사인자 NF-κB의 활성화에 미치는 RA-MC-AA 화합물의 억제 효과는, NF-κB의 전사활성에 의해 유도되는 다양한 친염증성 사이토카인들인 TNF-α, IL-6, IL-1β mRNA 수준의 감소현상으로도 확인되었다. Therefore, the inhibitory effect of RA-MC-AA compound on NO production induced by LPS treatment in RAW264.7 cells, a mouse macrophage, was achieved through the inhibition of activation of the transcription factor NF-κB associated with iNOS gene expression. It is suggested that this inhibition of NF-κB occurs by interfering with its phosphorylation mechanism, which is an inactivation mechanism of IkB protein, an inhibitor of NF-κB. The inhibitory effect of the RA-MC-AA compound on the activation of the transcription factor NF-κB induced in macrophages by LPS treatment is characterized by TNF-α, a variety of proinflammatory cytokines induced by the transcriptional activity of NF-κB. It was also confirmed by the reduction of IL-6 and IL-1β mRNA levels.

이러한 결과는, 도 7에서 나타내는 바와 같이 천초근 추출물로부터 분리되는 단일물질인 2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone (RA-MC-AA)는 IκB의 인산화를 감소시킴으로 LPS에 의해 유도되는 RAW264.7 세포내의 전사인자인 NF-κB 활성화를 저해하며, 이를 통해 LPS에 의해 유도되는 iNOS, COX-2 및 친염증성 사이토카인들인 TNF-α, IL-6, IL-1β의 유전자 전사를 저해함으로써 iNOS, COX-2 및 친염증성 사이토카인들인 TNF-α, IL-6, IL-1β 단백질의 수준을 낮출 수 있고, 따라서 LPS 처리에 의한 NO 생성을 저해하는 효과 즉 항염증 효과를 나타낼 수 있음을 보여준다(도 7 참조). As shown in FIG. 7, 2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone (RA-MC-AA), which is a single substance isolated from Cheoncho root extract, showed phosphorylation of IκB. Reduction inhibits the activation of NF-κB, a transcription factor in RAW264.7 cells induced by LPS, through which iNOS, COX-2 and proinflammatory cytokines, TNF-α, IL-6, IL, are induced by LPS. By inhibiting the gene transcription of -1β, it is possible to lower the levels of iNOS, COX-2 and proinflammatory cytokines TNF-α, IL-6, IL-1β protein, thus inhibiting NO production by LPS treatment, i.e. It can be shown to have an anti-inflammatory effect (see FIG. 7).

본 발명의 화합물을 포함하는 약학조성물의 제제예를 설명하나, 본 발명은 이를 한정하고자 함이 아닌 단지 구체적으로 설명하고자 함이다.Examples of the pharmaceutical compositions containing the compounds of the present invention will be described, but the present invention is not intended to be limited thereto, but is intended to be described in detail.

제제예 1. 산제의 제조Formulation Example 1 Preparation of Powder

RA-MC-AA 화합물 300 mgRA-MC-AA Compound 300 mg

유당 100 mgLactose 100 mg

탈크 10 mgTalc 10 mg

상기의 성분들을 혼합하고 기밀포에 충진하여 산제를 제조한다.The above ingredients are mixed and filled in an airtight cloth to prepare a powder.

제 제예 2. 정제의 제조Example 2 Preparation of Tablet

RA-MC-BB 화합물 300 mgRA-MC-BB Compound 300 mg

옥수수전분 100 mgCorn starch 100 mg

유당 100 mgLactose 100 mg

스테아린산 마그네슘 2 mg2 mg magnesium stearate

상기의 성분들을 혼합한 후 통상의 정제의 제조방법에 따라서 타정하여 정제를 제조한다.After mixing the above components, tablets are prepared by tableting according to a conventional method for preparing tablets.

제제예 3. 캅셀제의 제조Formulation Example 3 Preparation of Capsule

RA-MC-CC 화합물 300 mgRA-MC-CC Compound 300 mg

결정성 셀룰로오스 3 mg3 mg of crystalline cellulose

락토오스 14.8 mgLactose 14.8 mg

마그네슘 스테아레이트 0.2 mgMagnesium Stearate 0.2 mg

통상의 캡슐제 제조방법에 따라 상기의 성분을 혼합하고 젤라틴 캡슐에 충전하여 캡슐제를 제조한다.According to a conventional capsule preparation method, the above ingredients are mixed and filled into gelatin capsules to prepare capsules.

제제예 4. 주사제의 제조Formulation Example 4 Preparation of Injection

RA-MC-DD 화합물 300 mgRA-MC-DD Compound 300 mg

만니톨 180 mgMannitol 180 mg

주사용 멸균 증류수 2974 mgSterile distilled water for injection 2974 mg

Na2HPO412H2O 26 mgNa 2 HPO 4 12H 2 O 26 mg

통상의 주사제의 제조방법에 따라 1 앰플당 (2 ㎖) 상기의 성분 함량으로 제조한다.According to the conventional method for preparing an injection, the amount of the above ingredient is prepared per ampoule (2 ml).

제제예 5. 액제의 제조Formulation Example 5 Preparation of Liquid

RA-MC-EE 화합물 300 mgRA-MC-EE Compound 300 mg

이성화당 10 g10 g of isomerized sugar

만니톨 5 g5 g of mannitol

정제수 적량Purified water

통상의 액제의 제조방법에 따라 정제수에 각각의 성분을 가하여 용해시키고 레몬 향을 적량 가한 다음 상기의 성분을 혼합한 다음 정제수를 가하여 전체를 정제수를 가하여 전체 100 ㎖로 조절한 후 갈색 병에 충진하여 멸균시켜 액제를 제조한다.According to the conventional method of preparing a liquid solution, each component is added to the purified water to dissolve, the lemon flavor is added appropriately, the above components are mixed, purified water is added, the whole is adjusted to 100 ml by the addition of purified water, and then filled into a brown bottle. The solution is prepared by sterilization.

제제예 6. 건강 식품의 제조Formulation Example 6 Preparation of Healthy Food

RA-MC-FF 화합물 1000 ㎎RA-MC-FF Compound 1000 mg

비타민 혼합물 적량Vitamin mixture proper amount

비타민 A 아세테이트 70 ㎍70 μg of Vitamin A Acetate

비타민 E 1.0 ㎎Vitamin E 1.0 mg

비타민 B1 0.13 ㎎Vitamin B 1 0.13 mg

비타민 B2 0.15 ㎎0.15 mg of vitamin B 2

비타민 B6 0.5 ㎎Vitamin B 6 0.5 mg

비타민 B12 0.2 ㎍Vitamin B 12 0.2 g

비타민 C 10 ㎎Vitamin C 10 mg

비오틴 10 ㎍10 μg biotin

니코틴산아미드 1.7 ㎎Nicotinic Acid 1.7 mg

엽산 50 ㎍50 μg folic acid

판토텐산 칼슘 0.5 ㎎Calcium Pantothenate 0.5mg

무기질 혼합물 적량Mineral mixture

황산제1철 1.75 ㎎Ferrous Sulfate 1.75 mg

산화아연 0.82 ㎎Zinc Oxide 0.82 mg

탄산마그네슘 25.3 ㎎Magnesium carbonate 25.3 mg

제1인산칼륨 15 ㎎Potassium monophosphate 15 mg

제2인산칼슘 55 ㎎Dibasic calcium phosphate 55 mg

구연산칼륨 90 ㎎Potassium Citrate 90 mg

탄산칼슘 100 ㎎Calcium Carbonate 100 mg

염화마그네슘 24.8 ㎎Magnesium chloride 24.8 mg

상기의 비타민 및 미네랄 혼합물의 조성비는 비교적 건강식품에 적합한 성분을 바람직한 실시예로 혼합 조성하였지만, 그 배합비를 임의로 변형 실시하여도 무방하며, 통상의 건강식품 제조방법에 따라 상기의 성분을 혼합한 다음, 과립을 제조하고, 통상의 방법에 따라 건강식품 조성물 제조에 사용할 수 있다.Although the composition ratio of the above-mentioned vitamin and mineral mixture is comparatively mixed with a composition suitable for health food as a preferred embodiment, the compounding ratio may be arbitrarily modified, and the above ingredients are mixed according to a conventional method for producing healthy foods , Granules can be prepared and used in the manufacture of health food compositions according to conventional methods.

제제예 7. 건강 음료의 제조Formulation Example 7 Preparation of Healthy Drink

RA-MC-Hb 화합물 300 ㎎RA-MC-Hb Compound 300 mg

비타민 C 15 g15 g of vitamin C

비타민 E(분말) 100 g100 g of vitamin E (powder)

젖산철 19.75 gIron lactate 19.75 g

산화아연 3.5 g3.5 g of zinc oxide

니코틴산아미드 3.5 gNicotinamide 3.5 g

비타민 A 0.2 g0.2 g of vitamin A

비타민 B1 0.25 gVitamin B 1 0.25 g

비타민 B2 0.3 g0.3 g of vitamin B 2

물 정량Water quantification

통상의 건강음료 제조방법에 따라 상기의 성분을 혼합한 다음, 약 1시간 동 안 85℃에서 교반 가열한 후, 만들어진 용액을 여과하여 멸균된 2 ℓ 용기에 취득하여 밀봉 멸균한 뒤 냉장 보관한 다음 본 발명의 건강음료 조성물 제조에 사용한다. After mixing the above components in accordance with the conventional healthy beverage production method, and stirred and heated at 85 ℃ for about 1 hour, the resulting solution is filtered and obtained in a sterilized 2 L container, sealed sterilization and refrigerated It is used to prepare a healthy beverage composition of the present invention.

상기 조성비는 비교적 기호음료에 적합한 성분을 바람직한 실시예로 혼합 조성하였지만 수요계층이나, 수요국가, 사용용도 등 지역적, 민족적 기호도에 따라서 그 배합비를 임의로 변형 실시하여도 무방하다.Although the compositional ratio is relatively mixed with a component suitable for a favorite drink, it is also possible to arbitrarily modify the compounding ratio according to the regional or national preference such as the demand class, the demanding country, and the use purpose.

[이 발명을 지원한 국가연구개발사업][National R & D project supporting this invention]

[과제고유번호] [Task unique number]

B0009-008       B0009-008

[부처명][Name of Buddha]

지식경제부       Ministry of Knowledge Economy

[연구사업명][Name of research project]

지역혁신센터 사업       Regional Innovation Center Project

[연구과제명][Name of Research Project]

한약재 유래 천연물 소재의 면역조절작용 기전규명 및 산업화 기술개발       Mechanism of immunomodulation and development of industrialization technology

[주관기관][Host]

대구한의대학교 산학협력단        Daegu Haany University Industry-Academic Cooperation Foundation

[연구기간][Research period]

2008년 03월 01일 ~ 2010년 02월 28일        March 01, 2008 to February 28, 2010

도 1는 천초근 추출물로부터 분리된 화합물들의 세포독성을 나타낸 도이며(0, 6.25, 12.5 ug/ml),1 is a diagram showing the cytotoxicity of the compounds isolated from Cheoncho root extract (0, 6.25, 12.5 ug / ml),

도 2은 천초근 추출물로부터 분리된 화합물들의 NO 생성감소량을 나타낸 도이고(0, 6.25, 12.5 ug/ml),Figure 2 is a diagram showing the reduced NO production of compounds isolated from Cheoncho root extract (0, 6.25, 12.5 ug / ml),

도 3-A는 RA-MC-AA 화합물의 NO 생성감소량을 나타낸 도이며(0, 1, 2, 4 ug/ml),Figure 3-A is a diagram showing the NO production reduction amount of RA-MC-AA compound (0, 1, 2, 4 ug / ml),

도 3-B는 RA-MC-AA 화합물의 세포독성을 나타낸 도이고(0, 1, 2, 4 ug/ml),Figure 3-B shows cytotoxicity of RA-MC-AA compounds (0, 1, 2, 4 ug / ml),

도 4은 RA-MC-AA 화합물의 염증성 관련 단백질의 생성감소량을 나타낸 도이며(iNOS, COX-2),Figure 4 is a diagram showing the reduced production of inflammatory related proteins of RA-MC-AA compound (iNOS, COX-2),

도 5은 RA-MC-AA 화합물의 염증성 관련 단백질의 생성감소량을 나타낸 도이고(p-IkBα, IkBα),Figure 5 is a diagram showing the reduced production of inflammatory related proteins of RA-MC-AA compound (p-IkBα, IkBα),

도 6은 RA-MC-AA 화합물의 iNOS, COX-2, TNF-α, IL-6, IL-1β mRNA발현 억제를 나타낸 도이며(0, 1, 2, 4 ug/ml),6 is a diagram showing the inhibition of iNOS, COX-2, TNF-α, IL-6, IL-1β mRNA expression of RA-MC-AA compound (0, 1, 2, 4 ug / ml),

도 7는 대식세포주 Raw264.7의 염증성 신호전달을 모식화한 도이다.7 is a diagram schematically illustrating the inflammatory signaling of macrophage line Raw264.7.

Claims (9)

천초근 추출물로부터 분리된 2-카르보메톡시-2,3-에폭시-3-프레닐-1,4-나프토퀴논 (2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone), 3,3′-비스(3,4-디하이드로-4-하이드록시-6-메톡시-2H-1-벤조피란)(3,3′-bis(3,4-dihydro-4-hydroxy-6-methoxy-2H-1-benzopyran)), 에폭시몰루긴(Epoxymollugin), 소란지디올(Soranjidiol), 올레아놀릭 산(Oleanolic acid), 1-아세톡시-3-메톡시-9,10-안트라퀴논(1-acetoxy-3-methoxy-9,10-anthraquinone), 알리자린 2-메틸 아테르(Alizarin 2-methyl ether), 푸로놀루긴(Furonollugin), 및 몰루긴(mollugin) 으로 구성된 군으로부터 선택된 화합물 또는 이의 약학적으로 허용 가능한 염을 유효성분으로 함유하는 염증성 질환의 예방 및 치료용 약학조성물.2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone isolated from Cheoncho Geun extract (2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone ), 3,3′-bis (3,4-dihydro-4-hydroxy-6-methoxy-2H-1-benzopyran) (3,3′-bis (3,4-dihydro-4-hydroxy -6-methoxy-2H-1-benzopyran), Epoxymollugin, Soranjidiol, Oleanolic acid, 1-acetoxy-3-methoxy-9,10-anthra A compound selected from the group consisting of 1-acetoxy-3-methoxy-9,10-anthraquinone, alizarin 2-methyl ether, furonollugin, and mollugin; or A pharmaceutical composition for the prevention and treatment of inflammatory diseases containing a pharmaceutically acceptable salt thereof as an active ingredient. 제 1항에 있어서, 상기 천초근 추출물로부터 분리된 화합물은 2-카르보메톡시-2,3-에폭시-3-프레닐-1,4-나프토퀴논 (2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone) 화합물인 약학 조성물.The compound of claim 1, wherein the compound isolated from the myelin extract is 2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone (2-carbomethoxy-2,3-epoxy- 3-prenyl-1,4-naphthoquinone) compound. 제 1항에 있어서, 상기 추출물은 천초근의 조추출물, 극성용매 가용 추출물 또는 비극성용매 가용 추출물임을 특징으로 하는 약학조성물.According to claim 1, wherein the extract is a pharmaceutical composition, characterized in that the crude extract of cheoncho root, polar solvent soluble extract or non-polar solvent soluble extract. 제 3항에 있어서, 상기 조추출물은 정제수를 포함한 물, 탄소수 1 내지 4의 저급알코올 또는 이들의 혼합용매로부터 선택된 용매에 가용한 추출물을 포함함을 특징으로 하는 약학조성물.The pharmaceutical composition according to claim 3, wherein the crude extract comprises an extract available in a solvent selected from water including purified water, a lower alcohol having 1 to 4 carbon atoms, or a mixed solvent thereof. 제 3항에 있어서, 상기 비극성용매 가용 추출물은 메틸렌클로라이드, 헥산, 클로로포름, 디클로로메탄 또는 에틸아세테이트에 가용한 추출물을 포함함을 특징으로 하는 약학조성물.The pharmaceutical composition according to claim 3, wherein the nonpolar solvent soluble extract comprises an extract soluble in methylene chloride, hexane, chloroform, dichloromethane or ethyl acetate. 제 1항에 있어서, 상기 염증성 질환은 아토피피부염, 관절염, 요도염, 방광염, 동맥경화증, 알러지 질환, 비염, 천식, 급성통증, 만성통증, 치주염, 치은염, 염증성 장질환, 통풍, 심근경색, 울혈성 심부전, 고혈압, 협심증, 위궤양, 뇌경색, 다운증후군, 다발성 경화증, 비만, 당뇨, 치매, 우울증, 정신분열증, 결핵, 수면장애, 패혈증, 화상 또는 췌장염인 것을 특징으로 하는 약학조성물.The method of claim 1, wherein the inflammatory disease is atopic dermatitis, arthritis, urethritis, cystitis, arteriosclerosis, allergic diseases, rhinitis, asthma, acute pain, chronic pain, periodontitis, gingivitis, inflammatory bowel disease, gout, myocardial infarction, congestive A pharmaceutical composition characterized in that heart failure, hypertension, angina pectoris, gastric ulcer, cerebral infarction, Down syndrome, multiple sclerosis, obesity, diabetes, dementia, depression, schizophrenia, tuberculosis, sleep disorders, sepsis, burns or pancreatitis. 제 1항에 있어서, 조성물 총 중량에 대하여 상기 추출물을 0.1 내지 50% 중 량으로 포함됨을 특징으로 하는 약학조성물.The pharmaceutical composition according to claim 1, wherein the extract is included in an amount of 0.1 to 50% by weight based on the total weight of the composition. 천초근 추출물로부터 분리된 2-카르보메톡시-2,3-에폭시-3-프레닐-1,4-나프토퀴논 (2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone), 3,3′-비스(3,4-디하이드로-4-하이드록시-6-메톡시-2H-1-벤조피란)(3,3′-bis(3,4-dihydro-4-hydroxy-6-methoxy-2H-1-benzopyran)), 에폭시몰루긴(Epoxymollugin), 소란지디올(Soranjidiol), 올레아놀릭 산(Oleanolic acid), 1-아세톡시-3-메톡시-9,10-안트라퀴논(1-acetoxy-3-methoxy-9,10-anthraquinone), 알리자린 2-메틸 아테르(Alizarin 2-methyl ether), 푸로놀루긴(Furonollugin), 및 몰루긴(mollugin), 으로 구성된 군으로부터 선택된 화합물 또는 이의 약학적으로 허용 가능한 염을 유효성분으로 함유하는 염증성 질환의 예방 및 개선용 건강기능식품.2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone isolated from Cheoncho Geun extract (2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone ), 3,3′-bis (3,4-dihydro-4-hydroxy-6-methoxy-2H-1-benzopyran) (3,3′-bis (3,4-dihydro-4-hydroxy -6-methoxy-2H-1-benzopyran), Epoxymollugin, Soranjidiol, Oleanolic acid, 1-acetoxy-3-methoxy-9,10-anthra Compound selected from the group consisting of quinone (1-acetoxy-3-methoxy-9,10-anthraquinone), alizarin 2-methyl ether, furonollugin, and mollugin, Or health functional food for the prevention and improvement of inflammatory diseases containing a pharmaceutically acceptable salt thereof as an active ingredient. 제 8항에 있어서, 분말, 과립, 정제, 캡슐 또는 음료인 건강기능식품.The dietary supplement of claim 8 which is a powder, granule, tablet, capsule or beverage.
KR1020090000936A 2009-01-06 2009-01-06 A composition comprising the compound isolated from the extract of Rubiae Radix for preventing and treating inflammatory disease KR101072663B1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
KR1020090000936A KR101072663B1 (en) 2009-01-06 2009-01-06 A composition comprising the compound isolated from the extract of Rubiae Radix for preventing and treating inflammatory disease
PCT/KR2009/007837 WO2010079914A2 (en) 2009-01-06 2009-12-28 A composition comprising the compound isolated from the extract of rubiae radix for preventing and treating inflammatory diseases

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
KR1020090000936A KR101072663B1 (en) 2009-01-06 2009-01-06 A composition comprising the compound isolated from the extract of Rubiae Radix for preventing and treating inflammatory disease

Publications (2)

Publication Number Publication Date
KR20100081627A true KR20100081627A (en) 2010-07-15
KR101072663B1 KR101072663B1 (en) 2011-10-11

Family

ID=42316951

Family Applications (1)

Application Number Title Priority Date Filing Date
KR1020090000936A KR101072663B1 (en) 2009-01-06 2009-01-06 A composition comprising the compound isolated from the extract of Rubiae Radix for preventing and treating inflammatory disease

Country Status (2)

Country Link
KR (1) KR101072663B1 (en)
WO (1) WO2010079914A2 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101103426B1 (en) * 2009-04-10 2012-01-09 영남대학교 산학협력단 Pharmaceutical composition for treating inflammatory bowel disease comprising mollugin or its analogues
KR20160137042A (en) * 2015-05-22 2016-11-30 주식회사 더삼점영 A composition of plant extract for treating acne patients
KR20160142160A (en) 2015-06-02 2016-12-12 경북대학교 산학협력단 Apoptosis inhibitor comprising 2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone and use thereof
KR20170063122A (en) * 2015-11-30 2017-06-08 (주)아모레퍼시픽 Skin external composition containing aralosides
KR20220027642A (en) 2020-08-27 2022-03-08 경희대학교 산학협력단 A composition for prevention or treatment of bone disease comprising anthraquinone

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102875517A (en) * 2012-10-08 2013-01-16 河南大学 Mollugin and preparation method and application thereof
CN102920814B (en) * 2012-11-21 2014-02-26 孙振玲 Traditional Chinese medicine for treating chronic rheumatism
CN113679704B (en) * 2021-10-13 2023-08-25 张惊宇 Application of compound in preparation of medicine for relieving neuropathic pain

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS61268648A (en) * 1985-05-22 1986-11-28 Rooto Seiyaku Kk Production of oleanolic acid
FR2830195B1 (en) * 2001-10-03 2004-10-22 Sederma Sa COSMETIC AND DERMOPHARMACEUTICAL COMPOSITIONS FOR SKINS WITH ACNE TREND
KR100464063B1 (en) * 2001-11-21 2005-01-06 한국생명공학연구원 Triterpenoid compounds with apoptosis-inducing activity on cells
KR100485133B1 (en) * 2001-12-27 2005-04-22 류종훈 Composition containing an extract of rubiae radix for protecting brain cells and treating brain stroke

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101103426B1 (en) * 2009-04-10 2012-01-09 영남대학교 산학협력단 Pharmaceutical composition for treating inflammatory bowel disease comprising mollugin or its analogues
KR20160137042A (en) * 2015-05-22 2016-11-30 주식회사 더삼점영 A composition of plant extract for treating acne patients
KR20160142160A (en) 2015-06-02 2016-12-12 경북대학교 산학협력단 Apoptosis inhibitor comprising 2-carbomethoxy-2,3-epoxy-3-prenyl-1,4-naphthoquinone and use thereof
KR20170063122A (en) * 2015-11-30 2017-06-08 (주)아모레퍼시픽 Skin external composition containing aralosides
WO2017095146A1 (en) * 2015-11-30 2017-06-08 주식회사 아모레퍼시픽 Composition containing araloside for external application to skin
CN108366951A (en) * 2015-11-30 2018-08-03 株式会社爱茉莉太平洋 Composition for preparation for external use on skin containing the wooden saponin(e
US11052098B2 (en) 2015-11-30 2021-07-06 Amorepacific Corporation Composition containing araloside for external application to skin
CN108366951B (en) * 2015-11-30 2021-10-01 株式会社爱茉莉太平洋 Skin external preparation composition containing wood saponin
KR20220027642A (en) 2020-08-27 2022-03-08 경희대학교 산학협력단 A composition for prevention or treatment of bone disease comprising anthraquinone

Also Published As

Publication number Publication date
WO2010079914A2 (en) 2010-07-15
WO2010079914A3 (en) 2010-10-07
KR101072663B1 (en) 2011-10-11

Similar Documents

Publication Publication Date Title
KR101072663B1 (en) A composition comprising the compound isolated from the extract of Rubiae Radix for preventing and treating inflammatory disease
KR101416149B1 (en) Composition comprising an extract of Curcuma longa L. or Curcuma aromatica L. isolated therefrom having IL-6 induced STAT3 inhibitory activity
KR20110087448A (en) A composition comprising the compounds isolated from the inulae flos extract of inula japonica thunberg having anti-inflammatory or anti-allergic activity
KR101010743B1 (en) A composition comprising the extract of complex herbsKRJS as an active ingredient for preventing and treating inflammatory disease
KR20110017062A (en) Composition comprising ginsenoside for preventing and treating thrombotic diseases
KR101281710B1 (en) Composition comprising an extact of cirsium japonicum and the compounds isolated thereform for the treatment and preventation of obesity
KR102187335B1 (en) Composition for improving premenstrual syndrome symptom comprising compounds isolated from Hordeum vulgare extract
KR102080410B1 (en) Composition for Preventing, Treating or Improving of Andropause Syndrome Comprising Elderberry Extracts
KR100991279B1 (en) A composition comprising the novel compound isolated from the extract of Parthenocissus tricuspidata for preventing and treating inflammatory disease
KR20200103518A (en) Composition for improving premenstrual syndrome symptom comprising compounds isolated from Chrysanthemum zawadskii extract
KR100700481B1 (en) Composition comprising the extract of Cinnamomum cassia for preventing and treating fracture
KR101158536B1 (en) Angiogenesis inducing agent comprising the fractions from the extracts of Patrinia villosa Juss. as an active ingredient
KR20090128725A (en) The composition comprising extracts or fractions of magnolia obovata thunb for treating and preventing inflammation disease
KR20170054315A (en) Composition comprising extract of Nelumbo nucifera Gaertn. stamen and ovary for prevention and treatment of allergic diseases or inflammatory diseases
KR101457117B1 (en) Pharmaceutical composition and functional food for prevention or treatment of bone disease comprising the dryopteris crassithizoma extract
KR101403999B1 (en) A method for preparing a purified extract and the composition comprising the same for treating and preventing asthma and allergic disease
KR101392333B1 (en) Pharmaceutical compositions comprising extract or fractions of Rhododendron album Blume for prevention and treatment of inflammatory diseases
KR20150060653A (en) Pharmaceutical compositions for the treatment of cancer metastasis or inhibition of metastasis containing Quassia undulata extracts as active fractions
KR101454336B1 (en) Compositions for preventing and treating arthritis
KR101739425B1 (en) Composition for preventing or improving inflammatory disorders comprising fraction of Oenanthe javanica ethanol extract as effective component
KR20140065184A (en) Composition for preventing or treating liver cancer comprising ethyl acetate fraction from orostachys japonicus
KR102512643B1 (en) Composition for the prevention, improvement or treatment of allergic disease comprising fraction of biosulfur-containing filtrate
KR20060028076A (en) Composition containing yuanhuacine compound having an anti-cancer activity
KR101406126B1 (en) Composition containing Hedyotis diffusa extract for treating or preventing obesity
KR20110109558A (en) A composition comprising the extract of arctium lappa linne for treating or preventing vascular disease

Legal Events

Date Code Title Description
A201 Request for examination
E902 Notification of reason for refusal
E701 Decision to grant or registration of patent right
GRNT Written decision to grant
FPAY Annual fee payment

Payment date: 20140926

Year of fee payment: 4

FPAY Annual fee payment

Payment date: 20150925

Year of fee payment: 5

FPAY Annual fee payment

Payment date: 20160825

Year of fee payment: 6

FPAY Annual fee payment

Payment date: 20170925

Year of fee payment: 7

FPAY Annual fee payment

Payment date: 20180921

Year of fee payment: 8

FPAY Annual fee payment

Payment date: 20190925

Year of fee payment: 9