KR101010741B1 - A composition comprising the extract of complex herbs as an active ingredient for preventing and treating inflammatory disease - Google Patents

A composition comprising the extract of complex herbs as an active ingredient for preventing and treating inflammatory disease Download PDF

Info

Publication number
KR101010741B1
KR101010741B1 KR1020080021032A KR20080021032A KR101010741B1 KR 101010741 B1 KR101010741 B1 KR 101010741B1 KR 1020080021032 A KR1020080021032 A KR 1020080021032A KR 20080021032 A KR20080021032 A KR 20080021032A KR 101010741 B1 KR101010741 B1 KR 101010741B1
Authority
KR
South Korea
Prior art keywords
composition
extract
present
gardenia
active ingredient
Prior art date
Application number
KR1020080021032A
Other languages
Korean (ko)
Other versions
KR20090095844A (en
Inventor
박선동
허숙경
윤현정
김창현
Original Assignee
동국대학교 경주캠퍼스 산학협력단
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 동국대학교 경주캠퍼스 산학협력단 filed Critical 동국대학교 경주캠퍼스 산학협력단
Priority to KR1020080021032A priority Critical patent/KR101010741B1/en
Publication of KR20090095844A publication Critical patent/KR20090095844A/en
Application granted granted Critical
Publication of KR101010741B1 publication Critical patent/KR101010741B1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/28Asteraceae or Compositae (Aster or Sunflower family), e.g. chamomile, feverfew, yarrow or echinacea
    • A61K36/282Artemisia, e.g. wormwood or sagebrush
    • 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)
    • A61K36/70Polygonaceae (Buckwheat family), e.g. spineflower or dock
    • A61K36/708Rheum (rhubarb)
    • 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)
    • A61K36/744Gardenia
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2200/00Function of food ingredients
    • A23V2200/30Foods, ingredients or supplements having a functional effect on health
    • A23V2200/326Foods, ingredients or supplements having a functional effect on health having effect on cardiovascular health
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2200/00Function of food ingredients
    • A23V2200/30Foods, ingredients or supplements having a functional effect on health
    • A23V2200/3262Foods, ingredients or supplements having a functional effect on health having an effect on blood cholesterol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2236/00Isolation or extraction methods of medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicine
    • A61K2236/30Extraction of the material
    • A61K2236/33Extraction of the material involving extraction with hydrophilic solvents, e.g. lower alcohols, esters or ketones

Abstract

본 발명은 인진 (Artemisiae Capillaris Herba), 치자 (Gardeniae Fructus) 및 대황 (Rhei Radix et Rhizoma)의 복합 생약 추출물을 유효성분으로 함유하는 조성물에 관한 것으로, 상세하게는 본 발명의 복합 생약 추출물이 마우스 대식세포인 RAW 264.7 세포에서 LPS-유도된 NO, PGE2, iNOS 발현, COX-2 발현 및 염증성 사이토카인 (proinflammatory cytokines)의 함량을 유의적으로 감소하였으므로, 상기 조성물은 염증성 질환의 예방 및 치료용 약학조성물 또는 건강기능식품으로 유용하게 이용할 수 있다.The present invention ( Artemisiae Capillaris Herba ), Gardenia ( Gardeniae) Fructus ) and rhubarb ( Rhei Radix et Rhizoma ) relates to a composition containing the complex herbal extract of the present invention as an active ingredient, specifically, the complex herbal extract of the present invention LPS-induced NO, PGE 2 , iNOS expression, COX- in RAW 264.7 cells, which are mouse macrophages 2 Significantly reduced the expression and content of inflammatory cytokines (proinflammatory cytokines), the composition can be usefully used as a pharmaceutical composition or health functional food for the prevention and treatment of inflammatory diseases.

인진, 치자, 대황, 염증성 질환 Human, Gardenia, Rhubarb, Inflammatory diseases

Description

복합 생약 추출물을 유효성분으로 함유하는 염증성 질환의 예방 및 치료용 조성물 {A composition comprising the extract of complex herbs as an active ingredient for preventing and treating inflammatory disease} A composition comprising the extract of complex herbs as an active ingredient for preventing and treating inflammatory disease}

본 발명은 인진, 치자 및 대황의 복합 생약 추출물을 유효성분으로 함유하는 염증성 질환의 예방 및 치료용 약학조성물 또는 건강기능식품에 관한 것이다.The present invention relates to a pharmaceutical composition or health functional food for the prevention and treatment of inflammatory diseases containing a complex herbal extract of phosphorus, gardenia and rhubarb as an active ingredient.

[문헌 1] Lee TH et al., Mol. Cells., 23(3), pp398-404, 2007 [1] Lee TH et al., Mol. Cells. , 23 (3), pp 398-404, 2007

[문헌 2] Nishida T et al., Dig. Dis. Sci., 52(8), pp1890-1896, 20072 Nishida T et al., Dig. Dis. Sci. , 52 (8), pp 1890-1896, 2007

[문헌 3] 김호철. 한약약리학, 서울, 집문당, 2004[Reference 3] Kim Ho-cheol. Herbal Pharmacology, Seoul, Moon Mundang, 2004

[문헌 4] 박영순, 한방의 약리해설, 아카데미서적, pp153-154, 2002[Document 4] Park, Young-Soon, Pharmacological Commentary on Oriental Medicine, Academy Books, pp153-154, 2002

[문헌 5] Lee IA et al., Biol . Pharm . Bull ., 28(11), pp2106-2110, 2005[5] Lee IA et al., Biol . Pharm . Bull . , 28 (11), pp 2106-2110, 2005

[문헌 6] Peng CH et al., Curr . Cancer Drug Targets, 5(4) pp299-305, 20056 Peng CH et al., Curr . Cancer Drug Targets , 5 (4) pp 299-305, 2005

[문헌 7] 曾野維喜, 續東西醫學臨床漢方處方學, 南山堂, 1995[Reference 7] 曾 野 維 喜,, 東西 醫學 臨 床 漢 方 處方 學, 南山 堂, 1995

[문헌 8] 김영명 외. 한국식품영양과학회지, 35(7), pp841-846, 2006[Reference 8] Kim Young-myung et al. Korean Journal of Food Science and Nutrition, 35 (7), pp841-846, 2006

[문헌 9] Kim B.H et al., 대한한의학회지, 23(3), pp74-84, 2002 [Reference 9] Kim B.H et al., The Journal of Korean Oriental Medicine, 23 (3), pp74-84, 2002

[문헌 10] Desai A et al., J. Pharm . Sci . [Epub ahead of print], 200710 Desai A et al., J. Pharm . Sci . [Epub ahead of print], 2007

[문헌 11] Wang S et al., J. Ethnopharmacol ., 114(3), pp458-462, 2007[11] Wang S et al., J. Ethnopharmacol . , 114 (3), pp 458-462, 2007

[문헌 12] Heo et al., J. Immunol ., 179(9), pp6305-6310, 2007Heo et al., J. Immunol ., 179 (9), pp6305-6310, 2007

[문헌 13] Heo et al., J. Leukoc Biol ., 79(2), pp330-338, 2006Heo et al., J. Leukoc Biol ., 79 (2), pp 330-338, 2006

염증 (inflammation)은 외부 자극에 대한 생체조직의 방어반응의 하나이다. 염증반응이 일어나면 여러 가지 염증 인자들 (pro-inflammatory mediators)이 만들어지는데 이로 인하여 임상적으로는 발적, 발열, 종창, 동통, 기능장애 등의 증상이 나타난다. 염증 인자에는 유도적 산화질소 합성효소 (inducible nitric oxide synthase, iNOS)에 의해서 만들어지는 산화질소 (nitric oxide, NO)와 시클로옥시게나제-2 (cyclooxygenase-2, COX-2)에 의해서 만들어지는 프로스타글란딘E2 (prostagrandinE2, PGE2) 등이 있다. 이러한 염증 인자는 염증반응의 전사인자인 핵 전사인자-κB (nuclear factor-κB, NF-κB)를 활성화시키며, 그 결과 과량의 NO와 PGE2를 생성하여 염증을 일으킨다 (Lee TH et al., Mol . Cells ., 23(3), pp398-404, 2007; Nishida T et al., Dig . Dis. Sci ., 52(8), pp1890-1896, 2007). 포유동물 세포의 산화질소 합성효소 (nitric oxide synthase, NOS)의 경우, 유사 형태가 3종류 존재하는데, 신경성 산화질소 합성효소 (neuronal NOS, nNOS), 내피세포 산화질소 합성효소 (endothelial NOS, eNOS), 그리고 iNOS이다. 그 중에서 특히 iNOS가 염증반응에 관여한다. nNOS와 eNOS는 항상 발현되어있으며, iNOS의 경우 인터페론-γ (Interferon-γ), 리포폴리사카라이드 (lipopolysaccharide, LPS), 그리고 여러 가지 염증성 사이토카인의 자극이 있을 때 발현된다. 보통 eNOS는 강력한 혈관확장제로서 혈관의 항상성 (vascular homeostasis) 유지에 중요한 역할을 한다. 또한, NO는 급성, 만성 염증반응을 조절하기도 한다. PGE2는 COX에 의해서 아라키돈산 (arachidonic acid)으로부터 생산된다. COX에 대해서는 1990년대 초반에 주로 연구되었는데, 이 또한 유사형태가 2가지 존재한다. COX-1은 거의 모든 조직에 발현되어 있고, 프로스타글란딘을 생산하여 신장의 혈액 흐름을 조절하거나 위장의 세포를 보호하는 등의 생리적인 기능을 조절한다. 반대로, COX-2는 미생물에 의한 감염이나 손상 혹은 여러 요인의 스트레스에 반응한 대식세포 (Macrophage)에서 발현된다. 즉 iNOS와 COX-2의 발현과 NO, PGE2의 생산은 면역세포의 대표적인 염증인자이다. Inflammation is one of the defenses of biological tissues against external stimuli. When inflammatory reactions occur, various inflammatory factors (pro-inflammatory mediators) are produced, which causes clinical symptoms such as redness, fever, swelling, pain and dysfunction. Inflammatory factors include nitric oxide (NO), which is produced by inducible nitric oxide synthase (iNOS), and prostaglandins, which are produced by cyclooxygenase-2 (COX-2). E 2 (prostagrandinE 2 , PGE 2 ). These inflammatory factors activate nuclear transcription factor-κB (NF-κB), a transcription factor of the inflammatory response, resulting in excess NO and PGE 2 , resulting in inflammation (Lee TH et al., Mol . Cells . , 23 (3), pp 398-404, 2007; Nishida T et al., Dig . Dis. Sci . , 52 (8), pp 1890-1896, 2007). In the case of nitric oxide synthase (NOS) in mammalian cells, there are three similar forms: neuronal nitric oxide synthase (neuronal NOS, nNOS) and endothelial nitric oxide synthase (eNOS). And iNOS. In particular, iNOS is involved in the inflammatory response. nNOS and eNOS are always expressed and iNOS is expressed in the presence of stimulation of interferon-γ, lipopolysaccharide (LPS), and various inflammatory cytokines. Normally eNOS is a potent vasodilator that plays an important role in maintaining vascular homeostasis. NO also modulates acute and chronic inflammatory responses. PGE 2 is produced from arachidonic acid by COX. COX was mainly studied in the early 1990s, but there are also two similar forms. COX-1 is expressed in almost all tissues and produces prostaglandins that regulate physiological functions such as regulating blood flow in the kidneys or protecting the cells of the stomach. In contrast, COX-2 is expressed in macrophage in response to infection or damage caused by microorganisms or stress of various factors. In other words, iNOS and COX-2 expression, NO and PGE 2 production are representative inflammatory factors of immune cells.

또다른 염증인자로서는 염증성 사이토카인인 종양 괴사 인자-α (tumor necrosis factor-α, TNF-α), 인터류켄-1β (interleukin-1β, IL-1β), IL-6, 단핵구 화학주성 단백질-1 (monocyte chemoattractant protein-1, MCP-1) 등을 포함한다. Other inflammatory factors include tumor necrosis factor-α (TNF-α), interleuken-1β, IL-6, monocyte chemotactic protein-1, which are inflammatory cytokines. (monocyte chemoattractant protein-1, MCP-1), and the like.

최근, 천연자원은 여러 치료제의 선도 물질로서 개발되어 제약 산업에 소중한 자원으로서 이용되어 왔다. 그러므로 천연자원에서 항염증 질환의 예방 및 치료 물질을 개발하여 인공적인 화학물질보다 부작용을 줄일 수 있을 뿐만 아니라 가격면에서도 경쟁력이 있다. Recently, natural resources have been developed as leading substances in many therapeutic agents and have been used as valuable resources for the pharmaceutical industry. Therefore, it is possible to reduce the side effects than artificial chemicals by developing anti-inflammatory disease prevention and treatment materials from natural resources, and it is also competitive in price.

인진 (Artemisiae Capillaris Herba)은 이담, 간기능 보호 작용과 항균, 항바이러스 작용이 있으며, 또한 혈압강하와 항동맥경화 활성을 갖는다고 알려져 있다 (김호철. 한약약리학, 서울, 집문당. pp239-241, 2004). 인진의 성분 중 에스쿠 레틴 (esculetin)은 담즙의 분비를 촉진하고, 클로로제닉산 (chlorogenic acid) 및 카페탄닉산 (caffetannic acid)는 혈중의 콜레스테롤 (cholesterol)과 β-지단백질 (β-lipoprotein)을 저하시켜 혈관벽에 지방이 침착하는 것을 방지하며, 혈중지질 저하작용, 콜레스테롤 저하작용을 한다. 또한 유제놀 (eugenol) 및 유제닉산 (eugenic acid)은 항혈전작용, 담즙분비 촉진작용, 항산화작용을 한다고 알려져 있다 (박영순, 한방의 약리해설, 아카데미서적, pp153-154, 2002). Injin ( Artemisiae Capillaris Herba ) is known to have edema, hepatic function protection, antibacterial and antiviral effects, and also has an antihypertensive effect on blood pressure and arteriosclerosis (Kim, Ho-Cheol. Among the components of the phosphorus, esculetin promotes the secretion of bile, while chlorogenic acid and caffetannic acid release cholesterol and β-lipoprotein in the blood. Lowering prevents the deposition of fat on the walls of blood vessels, blood lipid lowering, cholesterol lowering action. In addition, eugenol and eugenic acid are known to have anti-thrombotic action, bile secretion promoting action and antioxidant action (Park, Young-Soon, Pharmacological Commentary of Oriental Medicine, Academy Books, pp153-154, 2002).

치자 (Gardeniae Fructus)는 피부진균에 대한 억제작용, 이담작용 및 췌장분비촉진작용이 있으며, 간기능 보호와 심근수축억제작용이 있다 (김호철. 한약약리학, 서울, 집문당. pp125-127, 2004). 치자와 그 구성성분 중 크로신 (crocin)과 크로세틴 (crocetin)은 췌장 리파아제 (lipase)의 활성을 억제함으로써 고지혈증을 개선시킬 수 있다고 보고하였다 (Lee IA et al., Biol . Pharm . Bull ., 28(11), pp2106-2110, 2005). 최근에는 치자의 제니포사이드 (geniposide)가 간, 심장 그리고 신장에 해롭지 않으면서 항종양인자가 될 수 있음이 보고되었다 (Peng CH et al., Curr . Cancer Drug Targets, 5(4) pp299-305, 2005). 치자의 성분으로는 제니포사이드 (geniposide), 제니핀 (genipin), 갈데노사이드 (gardenoside), 샨지사이드 (shanzhiside), 갈데닌 (gardenin), 만니톨 (mannitol), β-시토스테롤 (β-sitosterol), 노나코산 (nonacosane), 펙틴 (pectin), 탄닌 (tannin) 등이 있어 담즙분비 촉진작용, 항고콜레스테롤혈증, 제산작용, 항아세틸콜린작용, 항히스타민작용 등이 알려져 있다 (曾野維喜, 續東西醫學臨床漢方處方學, 南山堂, p98, 1995).Gardenia ( Gardeniae Fructus ) has the effect of inhibiting skin fungi, dipharymia and pancreatic secretion, protecting liver function and inhibiting myocardial contraction (Kim Ho-cheol. Herbal Pharmacology, Seoul, Munmundang. Pp125-127, 2004). Gardenia and its components, crocin and crocetin, have been reported to improve hyperlipidemia by inhibiting pancreatic lipase activity (Lee IA et al., Biol . Pharm . Bull . 28 (11), pp 2106-2110, 2005). Recently, gardenia Jenny Forsythe (geniposide) the liver, heart, and was reported to be a stand Anti yanginja do harm to the kidneys (Peng CH et al., Curr . Cancer Drug Targets , 5 (4) pp 299-305, 2005). Gardenia jasminoides, geniposide, genipin, galdenoside, shanzhiside, galdenin, mannitol, β-sitosterol, Nonnacosane, pectin, tannin, etc. are known to promote bile secretion, antihypercholesterolemia, antacid, antiacetylcholine, and antihistamine. (曾 野 維 喜, 續 東西 醫學臨 床 漢 方 處方 學, 南山 堂, p98, 1995).

대황 (Rhei Radix et Rhizoma)은 사하 성분인 안트라센 글리코사이드 (anthracene glycosides)를 함유하고 각종 세균에 대한 항균작용도 있다. 그 외에 이담, 지혈, 이뇨작용이 있으며 면역조절작용도 있다 (김호철. 한약약리학, 집문당, pp174-177, 2004). 대황에서 라미나란 (laminaran)을 추출한 다음 고지방 식이 흰쥐에게 투여한바 혈청 지질성분의 개선효과가 있으며 (김영명 외. 한국식품영양과학회지, 35(7), pp841-846, 2006), 대황이 Bax 단백질의 발현을 억제하여 상대적으로 Bax/Bcl-2 자연적 세포사를 억제하여 Mogolian gerbil의 가역성 전뇌허혈 모델에서 신경보호효과도 보고되었다 (Kim B.H et al., 대한한의학회지, 23(3), pp74-84, 2002). 대황의 성분은 안트라퀴논 (antraquinone) 유도체인 레인 (rhein), 이모딘 (emodin), 크리소파놀 (chrysophanol), 알로에-이모딘 (aloe-emodin), 피쉬온 (physcion) 등과 그 배당체들로 이루어져 있으며 다양한 약리작용이 밝혀져 있다. 특히 세노사이드 A (sennoside A)는 소화기계에서 사하작용이 강력함이 이미 증명되었으며, 혈액에 대한 작용으로 혈장침투압을 상승시켜, 조직의 수분을 혈관내로 이동시키고, 대출혈에 의한 혈액용량을 보충하며, 혈액점도를 감소시켜 미소순환장애를 개선한다고 알려졌다. 이외에도 항고지혈증, 항감염, 항종양, 이뇨, 그리고 여성 호르몬 양 작용 등이 밝혀져 활용되어온 약물이다 (曾野維喜, 續東西醫學臨床漢方處方學, 南山堂. pp207-208, 1995). Rhubarb (Rhei Radix et Rhizoma ) contains anthracene glycosides, which are a lower component, and has antibacterial activity against various bacteria. In addition, there is edema, hemostasis, diuretic effect and immunomodulatory activity (Ho Chul Kim. Pharmacology of Korean Medicine, Moon Mundang, pp174-177, 2004). After extracting laminaran from rhubarb and administering it to high-fat diet rats, it improves serum lipid composition (Kim Young-Myung et al., The Korean Journal of Food Science and Nutrition, 35 (7), pp841-846, 2006). Inhibition of Bax / Bcl-2 spontaneous cell death was also reported by neuroprotective effect of Mogolian gerbil in reversible whole cerebral ischemia model (Kim BH et al., The Korean Journal of Oriental Medicine, 23 (3), pp74-). 84, 2002). Rhubarb consists of anthraquinone derivatives rhein, emodin, chrysophanol, aloe-emodin, physcion, and glycosides thereof. And various pharmacological actions are revealed. Sennoside A, in particular, has been proved to have potent action in the digestive system, increasing plasma invasive pressure by action on blood, moving tissue moisture into blood vessels, and replenishing blood volume by bleeding blood. It is known to improve microcirculation disorder by reducing blood viscosity. In addition, antihyperlipidemia, anti-infection, anti-tumor, diuresis, and the effects of female hormones have been identified and used (曾 野 維 喜, 續 東西 醫學 臨 床 漢 方 學, 南山 堂. Pp207-208, 1995).

이에 본 발명자들은 마우스 대식세포인 RAW 264.7 세포에 인진, 치자 및 대황의 복합 생약 추출물을 처리하여 MTS 분석법, NO 생성량, PGE2 생성량, iNOS, COX-2 및 염증성 사이토카인 TNF-α, IL-1β, IL-6 및 MCP-1의 생성량 함량을 측정 함으로써 항염증 효과를 확인하여 본 발명을 완성하였다.In this regard, the present inventors treated a complex herbal extract of human, gardenia and rhubarb on RAW 264.7 cells, which are mouse macrophages, by MTS assay, NO production, PGE 2 production, iNOS, COX-2 and inflammatory cytokines TNF-α, IL-1β By measuring the amount of IL-6 and MCP-1 produced, the anti-inflammatory effect was confirmed to complete the present invention.

상기 목적을 달성하기 위하여, 본 발명은 인진, 치자 및 대황의 복합생약 추출물을 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 composite herbal extract of phosphorus, gardenia and rhubarb as an active ingredient.

또한, 본 발명은 인진, 치자 및 대황의 복합생약 추출물을 유효성분으로 함유하는 염증성 질환의 예방 및 개선용 건강기능식품을 제공한다.In another aspect, the present invention provides a health functional food for the prevention and improvement of inflammatory diseases containing a composite herbal extract of phosphorus, gardenia and rhubarb as an active ingredient.

본원에서 정의되는 추출물은 정제수를 포함한 물, 탄소수 1 내지 4의 저급알코올 또는 이들의 혼합용매로부터 선택된 용매, 바람직하게는 메탄올 추출물을 포함한다.Extracts as defined herein include solvents selected from water, including purified water, lower alcohols having 1 to 4 carbon atoms or mixed solvents thereof, preferably methanol extracts.

본원에서 정의되는 복합 생약 추출물은 인진, 치자 및 대황의 건조중량비 (w/w)가 5 내지 15: 1 내지 10: 1 내지 6, 바람직하게는 10: 5: 3을 포함함을 특징으로 한다.The complex herbal extract as defined herein is characterized in that the dry weight ratio (w / w) of phosphorus, gardenia and rhubarb comprises from 5 to 15: 1 to 10: 1 to 6, preferably 10: 5: 3.

본원에서 정의되는 염증성 질환은 동맥경화증, 결막염, 아토피, 관절염, 비염, 궤양성 대장염, 고혈압, 당뇨병, 암 또는 천식, 바람직하게는 동맥경화증, 관절염, 고혈압, 당뇨병 또는 암, 더욱 바람직하게는 동맥경화증 또는 고혈압을 포함한다.Inflammatory diseases as defined herein include atherosclerosis, conjunctivitis, atopic, arthritis, rhinitis, ulcerative colitis, hypertension, diabetes, cancer or asthma, preferably arteriosclerosis, arthritis, hypertension, diabetes or cancer, more preferably arteriosclerosis Or hypertension.

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

본 발명의 생약 추출물은 하기와 같이 제조될 수 있다. 건조된 인진, 치자 및 대황을 각각 1 내지 5배, 바람직하게는 3배의 정제수를 포함한 물, 탄소수 1 내지 4의 저급알코올 또는 이들의 혼합용매로부터 선택된 용매, 바람직하게는 메탄올로 15℃ 내지 100℃, 바람직하게는 20℃ 내지 30℃에서 24시간 내지 60시간, 바람직하게는 48시간 동안 추출하는 제 1단계; 제 1단계를 2회 반복하는 제 2단계; 이를 여과농축 및 동결 건조시키는 제 3단계를 포함하는 제조방법을 통해 본 발명의 추출물들을 수득할 수 있다.The herbal extract of the present invention can be prepared as follows. The dried phosphorus, gardenia and rhubarb are each selected from water containing 1 to 5 times, preferably 3 times purified water, a lower alcohol having 1 to 4 carbon atoms or a mixed solvent thereof, preferably 15 to 100 with methanol. A first step of extraction at 24 ° C., preferably at 20 ° C. to 30 ° C. for 24 hours to 60 hours, preferably for 48 hours; A second step of repeating the first step twice; Extracts of the present invention can be obtained through a manufacturing method including a third step of concentrating the filtration and freeze-drying.

또한 본 발명의 복합 생약 추출물은 하기와 같이 제조될 수 있다. 인진, 치자 및 대황의 건조중량비 (w/w)가 5 내지 15: 1 내지 10: 1 내지 6, 바람직하게는 10: 5: 3으로 혼합하는 제 1단계; 이에 1 내지 5배, 바람직하게는 3배의 정제수를 포함한 물, 탄소수 1 내지 4의 저급알코올 또는 이들의 혼합용매로부터 선택된 용매, 바람직하게는 메탄올로 15℃ 내지 100℃, 바람직하게는 20℃ 내지 30℃에서 24시간 내지 60시간, 바람직하게는 48시간 동안 추출하는 제 2단계; 제 2단계를 2회 반복하는 제 3단계; 이를 여과농축 및 동결 건조시키는 제 4단계를 포함하는 제조방법을 통해 본 발명의 복합 생약 추출물을 수득할 수 있다. In addition, the herbal extract of the present invention can be prepared as follows. A first step of mixing the dry weight ratio (w / w) of phosphorus, gardenia and rhubarb in 5 to 15: 1 to 10: 1 to 6, preferably 10: 5; A solvent selected from water containing 1 to 5 times, preferably 3 times purified water, a lower alcohol having 1 to 4 carbon atoms or a mixed solvent thereof, preferably 15 to 100 ℃, preferably 20 to A second step of extracting at 30 ° C. for 24 to 60 hours, preferably 48 hours; A third step of repeating the second step twice; Through the manufacturing method comprising the fourth step of filtration concentrated and freeze-dried can be obtained the complex herbal extract of the present invention.

본 발명은 상기의 제조방법으로 얻어진 복합 생약의 추출물을 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 extract of the complex herbal medicine obtained by the above method as an active ingredient.

또한, 본 발명은 상기의 제조방법으로 얻어진 복합 생약 추출물을 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 composite herbal extract obtained by the above production method as an active ingredient.

본 발명의 복합 생약 추출물을 함유하는 염증성 질환의 예방 및 치료를 위한 약학조성물은, 조성물 총 중량에 대하여 상기 복합 생약 추출물을 0.1 내지 50 중량% 포함한다.The pharmaceutical composition for the prevention and treatment of inflammatory diseases containing the complex herbal extract of the present invention comprises 0.1 to 50% by weight of the complex herbal extract based on the total weight of the composition.

본 발명의 복합 생약 추출물을 함유하는 약학조성물은 약학적 조성물의 제조에 통상적으로 사용하는 적절한 담체, 부형제 및 희석제를 더 포함할 수 있다. The pharmaceutical composition containing the complex herbal extract of the present invention may further comprise suitable carriers, excipients and diluents commonly used in the preparation of pharmaceutical compositions.

본 발명의 복합 생약 추출물을 함유하는 약학조성물은, 각각 통상의 방법에 따라 산제, 과립제, 정제, 캡슐제, 현탁액, 에멀젼, 시럽, 에어로졸 등의 경구형 제형, 외용제, 좌제 및 멸균 주사용액의 형태로 제형화하여 사용될 수 있다. 화합물을 함유하는 조성물에 함유될 수 있는 담체, 부형제 및 희석제로는 락토즈, 덱스트로즈, 수크로스, 솔비톨, 만니톨, 자일리톨, 에리스리톨, 말티톨, 전분, 아카시아 고무, 알지네이트, 젤라틴, 칼슘 포스페이트, 칼슘 실리케이트, 셀룰로즈, 메틸 셀룰로즈, 미정질 셀룰로스, 폴리비닐 피롤리돈, 물, 메틸히드록시벤조에이트, 프로필히드록시벤조에이트, 탈크, 마그네슘 스테아레이트 및 광물유를 들 수 있다. 제제화할 경우에는 보통 사용하는 충진제, 증량제, 결합제, 습윤제, 붕해제, 계면활성제 등의 희석제 또는 부형제를 사용하여 조제된다. 경구투여를 위한 고형제제에는 정제, 환제, 산제, 과립제, 캡슐제 등이 포함되며, 이러한 고형제제는 상기 화합물에 적어도 하나 이상의 부형제 예를 들면, 전분, 칼슘카보네이트 (calcium carbonate), 수크로스 (sucrose) 또는 락토오스 (lactose), 젤라틴 등을 섞어 조제된다. 또한 단순한 부형제 이외에 마그네슘 스테아레이트, 탈크 같은 윤활제들도 사용된다. 경구를 위한 액상 제제로는 현탁제, 내용액제, 유제, 시럽제 등이 해당되는데 흔히 사용되는 단순희석제인 물, 리퀴드 파라핀 이외에 여러 가지 부형제, 예를 들면 습윤제, 감미제, 방향제, 보존제 등이 포함될 수 있다. 비경구 투여를 위한 제제에는 멸균된 수용액, 비수성용제, 현탁제, 유제, 동결건조 제제, 좌제가 포함된다. 비수성용제, 현탁제로는 프로필렌글리콜 (propylene glycol), 폴리에틸렌 글리콜, 올리브 오일과 같은 식물성 기름, 에틸올레이트와 같은 주사 가능한 에스테르 등이 사용될 수 있다. 좌제의 기제로는 위텝솔 (witepsol), 마크로골 (macrogol), 트윈 (tween) 61, 카카오지, 라우린지, 글리세로제라틴 등이 사용될 수 있다.Pharmaceutical compositions containing the complex herbal extract of the present invention may be in the form of powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols, oral formulations, external preparations, suppositories, and sterile injectable solutions, respectively, according to conventional methods. Can be formulated and used. Carriers, excipients and diluents which may be contained in the composition containing the compound include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia rubber, alginate, gelatin, calcium phosphate, calcium Silicates, cellulose, methyl cellulose, microcrystalline cellulose, polyvinyl pyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil. When formulated, diluents or excipients such as fillers, extenders, binders, wetting agents, disintegrating agents, and surfactants are usually used. Solid form preparations for oral administration include tablets, pills, powders, granules, capsules, and the like, which form at least one excipient such as starch, calcium carbonate, sucrose, etc. Or lactose, gelatin and the like. In addition to simple excipients, lubricants such as magnesium stearate and talc are also used. Oral liquid preparations include suspensions, solvents, emulsions, and syrups, and may include various excipients, such as wetting agents, sweeteners, fragrances, and preservatives, in addition to commonly used simple diluents such as water and liquid paraffin. . Formulations for parenteral administration include sterile aqueous solutions, non-aqueous solvents, suspensions, emulsions, lyophilized preparations, suppositories. As the non-aqueous solvent and suspending agent, propylene glycol, polyethylene glycol, vegetable oil such as olive oil, injectable ester such as ethyl oleate, and the like can be used. As the base of the suppository, witepsol, macrogol, tween 61, cacao butter, laurin butter, glycerogelatin and the like can be used.

본 발명의 복합 생약 추출물의 사용량은 환자의 나이, 성별, 체중에 따라 달라질 수 있으나, 0.1 내지 100 mg/kg을 일일 1회 내지 수회 투여할 수 있다. 또한 그 투여량은 투여경로, 질병의 정도, 성별, 체중, 나이 등에 따라서 증감될 수 있다. 따라서 상기 투여량은 어떠한 면으로든 본 발명의 범위를 한정하는 것은 아니다.The amount of the combined herbal extract of the present invention may vary depending on the age, sex, and weight of the patient, but may be administered from 0.1 to 100 mg / kg once or several times daily. The dosage may also be increased or decreased depending on the route of administration, the severity of the disease, sex, weight, age, and the like. Therefore, the above dosage does not limit the scope of the present invention in any aspect.

상기 약학조성물은 쥐, 생쥐, 가축, 인간 등의 포유동물에 다양한 경로로 투여될 수 있다. 투여의 모든 방식은 예상될 수 있는데, 예를 들면, 경구, 직장 또는 정맥, 근육, 피하, 자궁 내 경막 또는 뇌혈관 내 (intracerebroventricular) 주사에 의해 투여될 수 있다.The pharmaceutical composition may be administered to various mammals such as mice, mice, livestock, humans, and the like. All modes of administration can be expected, for example by oral, rectal or intravenous, intramuscular, subcutaneous, intrauterine dural or intracerebroventricular injection.

본 발명은 복합 생약 추출물을 유효성분으로 포함하는 염증성 질환의 예방 및 개선용 건강기능식품을 제공한다. 이를 첨가할 수 있는 식품으로는 각종 식품류, 분말, 과립, 정제, 캡슐, 시럽제, 음료, 껌, 차, 비타민 복합제, 건강기능성 식품류 등이 있다.The present invention provides a dietary supplement for the prevention and improvement of inflammatory diseases comprising a composite herbal extract as an active ingredient. Foods to which it may be added include various foods, powders, granules, tablets, capsules, syrups, beverages, gums, teas, vitamin complexes, and health functional foods.

또한, 염증성 질환의 예방 효과를 목적으로 식품 또는 음료에 첨가될 수 있다. 이때 식품 또는 음료 중의 복합 생약 추출물의 양은 전체 식품 중량의 0.01 내지 15중량%로 가할 수 있으며, 건강 기능성 음료 조성물은 100 ml 기준으로 0.02 내지 5 g, 바람직하게는 0.3 내지 1 g의 비율로 가할 수 있다.It may also be added to food or beverages for the purpose of preventing inflammatory diseases. In this case, the amount of the composite herbal extract in the food or beverage may be added at 0.01 to 15% by weight of the total food weight, and the health functional beverage composition may be added at a ratio of 0.02 to 5 g, preferably 0.3 to 1 g, based on 100 ml. have.

본 발명의 건강 기능성 음료 조성물은 지시된 비율로 필수 성분으로서 상기 추출물을 함유하는 외에는 다른 성분에는 특별한 제한이 없으며, 통상의 음료와 같이 여러 가지 향미제 또는 천연 탄수화물 등을 추가성분으로서 함유할 수 있다. 상술한 천연 탄수화물의 예는 모노사카라이드, 예를 들어, 포도당, 과당 등; 디사카라이드, 예를 들어 말토스, 슈크로스 등; 및 폴리사카라이드, 예를 들어 덱스트린, 시클로덱스트린 등과 같은 통상적인 당 및 자일리톨, 소르비톨, 에르트리톨 등의 당알콜이다. 상술한 것 이외의 향미제로서 천연 향미제 (타우마틴, 스테비아 추출물) 및 합성 향미제 (사카린, 아스파르탐 등)를 유리하게 사용할 수 있다. 상기 천연 탄수화물의 비율은 본 발명의 조성물 100 ml 당 일반적으로 약 1 내지 20 g, 바람직하게는 약 5 내지 12 g이다.The health functional beverage composition of the present invention is not particularly limited to other ingredients except for containing the extract as an essential ingredient in the indicated ratio, 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 glucose, fructose and the like; Disaccharides such as maltose, sucrose and the like; And sugars such as conventional sugars such as polysaccharides such as dextrin, cyclodextrin and the like and xylitol, sorbitol, erythritol. As flavoring agents other than those mentioned above, natural flavoring agents (tautin, stevia extract) and synthetic flavoring agents (saccharin, aspartame, etc.) can be advantageously used. The proportion of such natural carbohydrates is generally about 1 to 20 g, preferably about 5 to 12 g per 100 ml of the composition of the present invention.

상기 외에 본 발명의 추출물은 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, 알콜, 탄산 음료에 사용되는 탄산화제 등을 함유할 수 있다. 그 밖에 본 발명의 추출물은 천연 과일주스 및 야채 음료의 제조를 위한 과육을 함유할 수 있다. 이러한 성분은 독립적으로 또는 조합하여 사용할 수 있 다. 이러한 첨가제의 비율은 그렇게 중요하지는 않지만 본 발명의 추출물 100 중량부 당 0 내지 약 20 중량부의 범위에서 선택되는 것이 일반적이다.In addition to the above, the extract of the present invention is a variety of nutrients, vitamins, minerals (electrolytes), synthetic flavors and natural flavors, such as flavoring agents, coloring and neutralizing agents (such as cheese, 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 beverages, and the like. In addition, the extract of the present invention may contain a pulp for the production of natural fruit juices and vegetable drinks. 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 extract of the present invention.

상기에서 설명한 바와 같이, 본 발명의 복합 생약 추출물은 RAW 264.7 세포에서 염증 인자의 지표인 NO의 생성량, PGE2의 생성량, iNOS의 발현도, COX-2의 발현도, 염증성 사이토카인인 TNF-α, IL-1β, IL-6 및 MCP-1의 생성량을 유의적으로 감소하는 항염 효과를 보여 염증성 질환의 예방 및 치료용 조성물로서 사용할 수 있다.As described above, the complex herbal extract of the present invention is the production of NO, PGE 2 , iNOS expression, COX-2 expression, inflammatory cytokine TNF-α in RAW 264.7 cells It can be used as a composition for the prevention and treatment of inflammatory diseases by showing an anti-inflammatory effect that significantly reduces the production of IL-1β, IL-6 and MCP-1.

이하, 본 발명을 상세히 설명한다. 단, 하기 실시예, 참고예 및 실험예는 본 발명을 예시하는 것일 뿐, 본 발명의 내용이 이에 의해 한정되는 것은 아니다.Hereinafter, the present invention will be described in detail. However, the following Examples, Reference Examples and Experimental Examples are merely illustrative of the present invention, but the content of the present invention is not limited thereto.

실시예 1. 생약 추출물의 제조Example 1 Preparation of Herbal Extracts

1-1. 인진의 메탄올 가용 추출물의 제조1-1. Preparation of Methanol Soluble Extract of Injin

인진 (Artemisiae Capillaris Herba)은 동국대학교 한의과대학 본초학 교실에서 선별된 것을 정선하여 300 g에 3배량의 100% 메탄올을 가한 다음, 48시간동안 25℃에서 추출하여, 이 과정을 2회 반복하여 여과한 후 농축하고 동결 건조를 통해 본 발명의 시료 (이하 "AC"라 명명함) 16.08 g (수율 5.36%)를 수득하였다. Injin ( Artemisiae Capillaris Herba ) was selected from the Department of Herbal Medicine, College of Oriental Medicine, Dongguk University, added 3 times 100% methanol to 300 g, extracted at 25 ° C for 48 hours, and filtered through this process twice. Concentrated and lyophilized to give 16.08 g (yield 5.36%) of the sample of the present invention (hereinafter referred to as "AC").

1-2. 치자의 메탄올 가용 추출물의 제조1-2. Preparation of Methanol Soluble Extract of Gardenia Gardenia

치자 (Gardeniae Fructus)은 동국대학교 한의과대학 본초학 교실에서 선별된 것을 정선하여 300 g에 3배량의 100% 메탄올을 가한 다음, 48시간동안 25℃에서 추출하여, 이 과정을 2회 반복하여 여과한 후 농축하고 동결 건조를 통해 본 발명의 시료 (이하 “GF"이라 명명함) 76.5 g (수율 25.5%)을 수득하였다. Gardenia ( Gardeniae Fructus ) was selected from the Department of Herbal Medicine, Dongguk University, College of Oriental Medicine, added 3 times 100% methanol to 300 g, extracted at 25 ℃ for 48 hours, this process was filtered twice, concentrated and frozen Drying gave 76.5 g (yield 25.5%) of the sample of the present invention (hereinafter referred to as "GF").

1-3. 대황의 메탄올 가용 추출물의 제조1-3. Preparation of Methanol Soluble Extract of Rhubarb

대황 (Rhei Radix et Rhizoma)은 동국대학교 한의과대학 본초학 교실에서 선별된 것을 정선하여 300 g에 3배량의 100% 메탄올을 가한 다음, 48시간동안 25℃에서 추출하여, 이 과정을 2회 반복하여 여과한 후 농축하고 동결 건조를 통해 본 발명의 시료 (이하 “RR"이라 명명함) 57.9 g (수율 19.3%)을 수득하였다.Rhubarb ( Rhei Radix et Rhizoma ) was selected from the Department of Herbal Medicine, College of Oriental Medicine, Dongguk University, added 3 times 100% methanol to 300 g, extracted at 25 ° C for 48 hours, and filtered through this process twice. After concentrating and freeze-drying, 57.9 g (yield 19.3%) of the sample of the present invention (hereinafter referred to as “RR”) were obtained.

1-4. 복합 생약 추출물의 제조1-4. Preparation of Complex Herbal Extracts

인진, 치자 및 대황 (동국대학교 한의과대학 본초학 교실 제공)을 각각 150 g, 75 g 및 45 g을 혼합하여 총 270 g에 3배량의 100% methanol을 가한 다음 48시간 동안, 25℃에서 추출하고, 이 과정을 2회 반복하여 여과한 후 농축하고 동결 건조를 통해 본 발명의 시료 (이하 “IJHT"이라 명명함)하여 48.06 g (수율 17.8%)을 수득하여 하기 실험에 사용하였다.Injin, Gardenia and Rhubarb (provided with the Department of Herbal Medicine, Dongguk University) were mixed with 150 g, 75 g and 45 g, respectively, and 3 times of 100% methanol was added to a total of 270 g, followed by extraction at 25 ° C. for 48 hours. This process was repeated twice, filtered, concentrated and lyophilized to give a sample of the present invention (hereinafter referred to as “IJHT”) to obtain 48.06 g (yield 17.8%) of the following experiment.

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

Dulbecco's Modifide Eagle Medium (DMEM), fetal bovine serum (FBS), streptomycin-penicillin 등의 세포 배양용 시약들은 Gibco BRL사 (Grand Island, USA)에서 구입하였으며, Sodium Dodesyl Sulfate (SDS), Acrylamide, Bis, Protein assay reagent는 Bio-Rad사 (Hercules, USA)에서 구입하였고, NP-40, CAPS, tween20, protease inhibitors 등은 Sigma사 (St. Louis, USA)에서 구입하였다. 실험에 사용된 1차 항체인 iNOS monoclonal antibody (mAb) 및 2차 항체인 anti-rabbit IgG horseradish peroxidase (HRP)-conjugated antibody는 SantaCruz Biotechnology사 (Santa Cruz, CA)에서, COX-2 polyclonal antibody와 β-actin mAb는 Cell Signaling Technology사 (Beverly, MA)에서 구입하였다. Aqueous One Solution Cell Priliferation Assay (MTS) 킷트와 Griess Reagent System은 Promega사 (Madison, USA)에서 구입하였고, 각종 사이토카인 측정을 위한 ELISA 킷트는 Pierce Biotechnology사 (Rockford, USA)에서 구입하였으며, PGE2 분석 킷트는 R&D사 (Minneapolis, USA)에서 구입하였다. 실험에 사용된 모든 시약은 분석용 등급이상으로 사용하였다.Cell culture reagents such as Dulbecco's Modifide Eagle Medium (DMEM), fetal bovine serum (FBS), and streptomycin-penicillin were purchased from Gibco BRL (Grand Island, USA), Sodium Dodesyl Sulfate (SDS), Acrylamide, Bis, Protein Assay reagents were purchased from Bio-Rad (Hercules, USA), NP-40, CAPS, tween20, protease inhibitors, etc. were purchased from Sigma (St. Louis, USA). The primary antibody iNOS monoclonal antibody (mAb) and the secondary antibody anti-rabbit IgG horseradish peroxidase (HRP) -conjugated antibody were produced by SantaCruz Biotechnology (Santa Cruz, CA), and COX-2 polyclonal antibody and β. -actin mAb was purchased from Cell Signaling Technology (Beverly, MA). Aqueous One Solution Cell Priliferation Assay (MTS) kit and Griess Reagent System were purchased from Promega (Madison, USA) and ELISA kits for various cytokine measurements were purchased from Pierce Biotechnology (Rockford, USA) and PGE 2 analysis The kit was purchased from R & D (Minneapolis, USA). All reagents used in the experiments were used above the analytical grade.

참고예 2. 세포배양Reference Example 2. Cell Culture

마우스의 대식세포주인 RAW 264.7 세포 (한국세포주은행 (KCLB))를 10% FBS과 1% 페니실린-스트렙토마이신 (penicillin-streptomycin)을 포함하는 DMEM (Dulbecco's Modified Eagle Medium) 배지에 37℃, 5% CO2의 조건에서 배양하여 하기 실험에 사용하였다. 또한 모든 실험결과는 평균과 표준 편차로 표시하고 유의성 검증은 시그마 플롯 (Sigma Plot, Window용 version 7.0)을 이용하여 student's t-test (p<0.001)를 실시하여 실험군과 대조군(#) 또는 실험군과 LPS 시험군(*)간의 유의성을 표기하였다.Macrophage lines of mice, RAW 264.7 cells (KCLB), were treated with DMEM (Dulbecco's Modified Eagle Medium) medium containing 10% FBS and 1% penicillin-streptomycin (37%, 5% CO). Cultured under the conditions of 2 was used in the following experiment. In addition, all experimental results are expressed as mean and standard deviation, and significance test is performed by using student's t- test (p <0.001) using sigma plot (Sigma Plot, version 7.0 for Window). Significance between LPS test groups (*) is indicated.

실험예 1. MTS 분석법Experimental Example 1. MTS Analysis

상기 실시예 1에서 수득한 추출물들의 세포에 대한 독성 측정을 위해 문헌 (Desai A et al., J. Pharm . Sci. [Epub ahead of print], 2007)에 기재된 5-(3-caroboxymeth-oxyphenyl)-2H-tetra-zolium inner salt (MTS) 분석 방법을 이용하여 하기와 같이 실험을 수행하였다. 5- (3-caroboxymeth-oxyphenyl) described in Desai A et al., J. Pharm . Sci. [Epub ahead of print], 2007 for measuring the toxicity of the extracts obtained in Example 1 above . The experiment was performed using the -2H-tetra-zolium inner salt (MTS) analysis method as follows.

상기 참고예 2의 방법으로 배양된 RAW 264.7 세포를 96 웰 플레이트에 1×104/웰 분주하고, 상기 실시예 1에서 수득한 추출물들을 각각 농도별 (0, 50, 100, 300, 500, 700 μg/ml)로 18 시간동안 처리하였다. 웰당 20 ㎕의 MTS 용액을 첨가하여 37℃, 5% CO2 배양기에서 4시간 동안 반응시킨 후, 마이크로 플레이트 리더 (microplate reader, DYNEX, Opsys MR, USA)를 이용하여 450 nm에서 흡광도의 변화를 측정하여 추출물을 처리하지 않은 대조군에 대한 세포생존율을 백분율로 표시하였다. 각 농도별 약재가 갖는 흡광도를 보정하기 위하여 세포를 뺀 배지를 같이 배양하여 대조군과 실험군의 흡광도를 비교 보정하여 세포 생존율을 백분율 (평균값±표준편차)로 표시하였다 (하기 수학식 1 참조).RAW 264.7 cells cultured by the method of Reference Example 2 were divided into 1 × 10 4 / well in 96 well plates, and the extracts obtained in Example 1 were prepared by concentration (0, 50, 100, 300, 500, 700, respectively). μg / ml) for 18 hours. After 20 μl MTS solution was added per well for 4 hours in a 37 ° C., 5% CO 2 incubator, the change in absorbance at 450 nm was measured using a microplate reader (DYNEX, Opsys MR, USA). By expressing the cell viability of the control group without the extract was expressed as a percentage. In order to correct the absorbance of the drug by each concentration, the culture medium without the cells were cultured together to compare and correct the absorbance of the control group and the experimental group to express the cell viability as a percentage (mean value ± standard deviation) (see Equation 1 below).

세포생존율 (%) = {(시험군의 흡광도-시료만의 흡광도)/ 대조군의 흡광도} X 100% Cell viability = {(absorbance of test group-absorbance of sample only) / absorbance of control group} X 100

시료sample 농도(㎍/ml)Concentration (㎍ / ml) 세포생존율(%)Cell survival rate (%) ACAC 00 100±0100 ± 0 5050 112±7112 ± 7 100100 105±8105 ± 8 300300 99±499 ± 4 500500 38±238 ± 2 700700 32±332 ± 3 GFGF 00 100±0100 ± 0 5050 123±15123 ± 15 100100 122±14122 ± 14 300300 115±13115 ± 13 500500 112±8112 ± 8 700700 102±5102 ± 5 RRRR 00 100±0100 ± 0 5050 118±11118 ± 11 100100 108±9108 ± 9 300300 100±9100 ± 9 500500 79±279 ± 2 700700 58±458 ± 4 IJHTIJHT 00 100±0100 ± 0 5050 119±14119 ± 14 100100 122±15122 ± 15 300300 123±12123 ± 12 500500 119±11119 ± 11 700700 106±15106 ± 15

실험결과, 표 1에 나타난 바와 같이 IJHT 및 GF는 모든 농도에서 RAW 264.7의 세포 생존율에는 거의 영향을 주지 않은 반면에 AC 및 RR은 고농도에서 세포의 생존율을 감소시킴을 확인할 수 있었다. 따라서 RAW 264.7의 세포 생존율에 영향을 주지 않는 IJHT 300 μg/㎖의 농도로 하기 실험을 수행하였다. As shown in Table 1, IJHT and GF had little effect on the cell viability of RAW 264.7 at all concentrations, whereas AC and RR decreased cell viability at high concentrations. Therefore, the following experiment was carried out at a concentration of 300 μg / ㎖ of IJHT does not affect the cell viability of RAW 264.7.

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

NO의 생성량을 측정하기 위해 문헌 (Wang S et al., J. Ethnopharmacol., 114(3), pp458-462, 2007)에 기재되어 있는 방법을 이용하여 하기와 같이 실험하였다.In order to determine the amount of NO produced, experiments were performed using the method described in Wang S et al., J. Ethnopharmacol. , 114 (3), pp458-462, 2007.

상기 참고예 2의 방법으로 배양된 RAW 264.7 세포에 상기 실시예 1-4에서 수득한 IJHT를 전처리 하고 1시간 후 100 ng/ml의 LPS를 처리하여 18시간 배양하였다. 배양액 50 ㎕와 같은 양의 그리스 반응액 (Griess Reagent)을 넣어주고 10분간 상온에서 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.RAW 264.7 cells cultured by the method of Reference Example 2 were pretreated with the IJHT obtained in Example 1-4 and incubated for 18 hours by treatment with 100 ng / ml LPS after 1 hour. Add 50 g of culture medium with the same amount of Greases Reagent and allow to react at room temperature for 10 minutes, using Automatic ELISA reader (precision microplate reader, Molecular Devices, BIO-TEX EL800uv-PC, USA) at 540 nm. Absorbance was measured. NO concentration in the culture was determined using a standard curve of concentration of sodium nitrite.

실험결과, 도 1A에 나타난 바와 같이 LPS 100 ng/ml의 농도로 처리하였을 때, 아무 처리도 하지 않은 대조군에 비하여 NO의 생성량이 약 10배 증가됨을 확인할 수 있었다. 또한 IJHT 300 μg/㎖의 농도를 1시간 동안 전 처리하였을 때, LPS-유도된 NO의 생성량이 약 74% 감소됨을 확인할 수 있었다.As a result, as shown in FIG. 1A, when the LPS was treated at a concentration of 100 ng / ml, it was confirmed that the amount of NO produced was increased by about 10 times compared to the control which did not have any treatment. In addition, when the concentration of IJHT 300 μg / ㎖ pretreated for 1 hour, it was confirmed that the amount of LPS-induced NO production reduced by about 74%.

실험예 3. PGEExperimental Example 3. PGE 22 의 생성량 측정Measurement of production

PGE2의 양을 측정하기 위해 상용 경쟁적 효소 면역분석 킷트 (commercial competitive enzyme immunoassay kit, R&D systems, Minneapolis)를 이용하여 하기와 같이 실험을 수행하였다. In order to measure the amount of PGE 2 , experiments were performed using a commercial competitive enzyme immunoassay kit (R & D systems, Minneapolis) as follows.

상기 참고예 2의 방법으로 배양된 RAW 264.7 세포에 상기 실시예 1-4에서 수득한 IJHT 300 ㎍/ml을 1시간 동안 전 처리한 후 100 ng/ml의 LPS를 처리하였다. 18 시간 후 세포 배양액을 goat anti-mouse로 코팅 (coating)된 96웰 플레이트에 각각의 배양액을 100 ㎕씩 주입 (loading)한다. 여기에 primary antibody solution 50 ㎕와 PGE2 conjugate 50 ㎕씩 첨가하여 4℃에서 하룻밤 방치시켰다. 세척 완충용액 (washing buffer)로 4회 세척하고 기질용액 (substrate solution)을 200 ㎕씩 처리하여 5-20분간 반응시킨 후, 50 ㎕의 반응정지 용액 (stop solution)을 처리한 후 450 nm에서 흡광도를 측정하였다.RAW 264.7 cells cultured by the method of Reference Example 2 were pretreated with 300 μg / ml of IJHT obtained in Example 1-4 for 1 hour and then treated with 100 ng / ml of LPS. After 18 hours, 100 µl of each culture was loaded into a 96-well plate coated with goat anti-mouse. 50 μl of primary antibody solution and 50 μl of PGE 2 conjugate were added thereto, and the plate was left at 4 ° C. overnight. After washing 4 times with washing buffer and treating with 200 μl of substrate solution for 5-20 minutes, the solution was treated with 50 μl of stop solution and absorbance at 450 nm. Was measured.

실험결과, 도 1B에 나타난 바와 같이, LPS 100 ng/ml의 농도에서 아무 처리도 하지 않은 대조군에 비해 PGE2 생성량이 약 20배 증가됨을 확인 할 수 있었다. 또한, IJHT 300 ㎍/ml 처리한 실험군은 LPS-유도된 PGE2의 생성량을 약 35% 감소됨을 확인할 수 있었다.As a result, as shown in Figure 1B, it was confirmed that the amount of PGE 2 production is increased by about 20 times compared to the control without any treatment at a concentration of 100 ng / ml LPS. In addition, it was confirmed that the experimental group treated with IJHT 300 ㎍ / ml reduced the amount of LPS-induced PGE 2 by about 35%.

실험예 4. 웨스턴 블롯 분석법 (Western blot analysis)Experimental Example 4. Western blot analysis

iNOS 및 COX-2 단백질 발현 측정을 하기 위해 허 등의 방법 (Heo et al., J. Immunol., 179(9), pp6305-6310, 2007)을 이용하여 웨스턴 블롯 분석법을 하기와 같이 수행하였다. Western blot analysis was performed using Hu et al. (Heo et al., J. Immunol., 179 (9), pp6305-6310, 2007) to measure iNOS and COX-2 protein expression.

상기 실시예 1-4에서 수득한 IJHT 300 ㎍/ml을 참고예 2의 방법으로 배양된 세포에 ice-cold Tris buffered saline (TBS: 20 mM Tris-HCl, pH 8.0, 137 mM NaCl)으로 3회 세척한 후, 용해 완충액 (lysis buffer: TBS, 1% NP-40, 1 mM sodium orthovanadata, 10 ㎍/㎖ aprotinin, 10 ㎍/㎖ leupeptin 및 1 mM PMSF)를 넣어 4℃에서 30분간 반응시키고 12,000×g에서 10분간 원심 분리하여 상층액을 모았다. 동일한 양의 단백질을 7.5% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE)로 분리시킨 후, 단백질을 니트로셀룰로스 멤브레인 (nitrocellulose membrane, Whatman, Germany)에 전이 (transfer)하였다. 이 멤브레인을 항체의 비특이적 결합을 차단하기 위하여 블로킹 용액 (blocking buffer: 5% non-fat milk와 0.1% Tween20을 함유한 TBS 용액)에서 1시간 동안 반응시킨 후, 각 단백질에 대한 항체 (anti-iNOS 및 anti-COX2)를 가하여 1 내지 2시간 동안 반응시켰다. 이어서 0.1% Tween20을 함유한 TBST 용액으로 40분간 세척한 다음, 2차 항체 (anti-rabbit IgG horseradish peroxidase-conjugated antibody)로 반응시켰다. 이어서 ECL system으로 반응 시킨 후에 X-ray film (AGFA, Belgium) 상에서 단백질을 확인하였다. 각 시료의 단백질 정량은 Bradford protein assay kit를 사용하여 595 nm에서 흡광도를 측정하여 실시하였다.300 μg / ml of IJHT obtained in Example 1-4 was three times with ice-cold Tris buffered saline (TBS: 20 mM Tris-HCl, pH 8.0, 137 mM NaCl) in cells cultured by the method of Reference Example 2. After washing, add lysis buffer (lysis buffer: TBS, 1% NP-40, 1 mM sodium orthovanadata, 10 μg / ml aprotinin, 10 μg / ml leupeptin and 1 mM PMSF) and react at 30 ° C. for 30 minutes. The supernatant was collected by centrifugation at g for 10 minutes. The same amount of protein was separated by 7.5% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), and then the protein was transferred to a nitrocellulose membrane (Whatman, Germany). The membrane was reacted for 1 hour in a blocking buffer (blocking buffer: TBS solution containing 5% non-fat milk and 0.1% Tween20) to block nonspecific binding of the antibody, followed by antibody to each protein (anti-iNOS). And anti-COX2) were added and reacted for 1 to 2 hours. It was then washed with a TBST solution containing 0.1% Tween20 for 40 minutes and then reacted with a secondary antibody (anti-rabbit IgG horseradish peroxidase-conjugated antibody). Subsequently, the protein was confirmed on the X-ray film (AGFA, Belgium) after the reaction with the ECL system. Protein quantification of each sample was performed by measuring the absorbance at 595 nm using a Bradford protein assay kit.

실험결과, 도 2에 나타난 바와 같이, LPS 100 ng/ml를 처리했을 때, iNOS 와 COX-2의 발현이 대조군에 비해 현저히 증가됨을 확인할 수 있었다. 또한, IJHT 300 μg/㎖의 농도를 1시간 동안 전 처리했을 때, LPS 100 ng/ml에 의해 유도되는 iNOS 및 COX-2의 발현이 각각 52% 및 79% 감소됨을 확인할 수 있었다.As a result, as shown in Figure 2, when treated with LPS 100 ng / ml, it was confirmed that the expression of iNOS and COX-2 significantly increased compared to the control. In addition, when the concentration of IJHT 300 μg / ㎖ pretreated for 1 hour, it was confirmed that the expression of iNOS and COX-2 induced by LPS 100 ng / ml 52% and 79%, respectively.

실험예 5. 세포 배양액 내의 사이토카인 (cytokines) 측정Experimental Example 5. Measurement of cytokines in cell culture

염증을 나타내는 지표로써, 세포 배양액 내의 염증성 사이토카인 (proinflammatory cytokines)의 양을 측정하기 위해 허 등의 방법 (Heo et al., J. Leukoc Biol., 79(2), pp330-338, 2006)을 이용하여 ELISA 분석법 (Enzyme-Linked Immunosorbent Assay)을 하기와 같이 수행하였다. As an indicator of inflammation, Hu et al. (Heo et al., J. Leukoc Biol., 79 (2), pp330-338, 2006) were used to measure the amount of inflammatory cytokines in cell culture. ELISA assay (Enzyme-Linked Immunosorbent Assay) was performed as follows.

상기 참조예 2의 방법으로 배양된 RAW 264.7 세포에 상기 실시예 1-4에서 수득한 IJHT 300 μg/㎖의 농도로 1시간 처리한 후 LPS 100 ng/ml를 처리하여 18 시간 후 세포 배양액을 적절한 농도로 희석한 후, 사이토카인으로 코팅된 96웰 플레이트에 50 ㎕씩 첨가하여 4℃에서 오버나이트시켰다. 워싱 완충액 (washing buffer)로 3회 세척하고 100 ㎕의 비오티닐화 항체 반응액 (biotinylated antibody reagent)를 각각의 웰에 처리하여 1시간 동안 상온에서 반응시킨 후 3회 세척한 다음, 100 ㎕의 스트렙타비딘-HRP 용액 (streptavidine-HRP solution)을 각각의 웰에 처리하여 1시간 동안 상온에서 반응시킨 후 다시 세척 완충용액 (washing buffer)로 3회 세척하였다. 여기에 di(2-ethylhexyl)-2,4,5-trimethoxybenzalmalonate (TMB) 기질을 100 ㎕씩 처리하여 5-30분간 반응시킨 후 100 ㎕의 반응 정지용액 (stop solution)을 처리한 후 450 nm에서 흡광도를 측정하였다. RAW 264.7 cells cultured by the method of Reference Example 2 were treated with IJHT obtained in Example 1-4 at a concentration of 300 μg / ml for 1 hour, and then treated with LPS 100 ng / ml for 18 hours. After dilution to concentration, 50 μl each was added to a cytokine-coated 96 well plate and overnight at 4 ° C. Wash three times with washing buffer and 100 μl of biotinylated antibody reagent to each well, react at room temperature for 1 hour, wash three times, and then 100 μl of strep. Tavidin-HRP solution (streptavidine-HRP solution) was treated in each well and allowed to react at room temperature for 1 hour, and then washed three times with washing buffer again. 100 μl of di (2-ethylhexyl) -2,4,5-trimethoxybenzalmalonate (TMB) substrate was treated for 5-30 minutes, followed by 100 μl reaction stop solution at 450 nm. Absorbance was measured.

실험결과, 도 3에 나타난 바와 같이 RAW 264.7 세포에 LPS 100 ng/ml의 농도로 처리하였을 때, TNF-α는 4.3배, IL-1β는 6.2배, IL-6는 57.3배, MCP-1은 5.2배 증가됨을 확인할 수 있었다. IJHT 300 μg/㎖의 농도를 1시간 동안 전 처리한 후 100 ng/ml LPS를 18시간 동안 처리하여 LPS에 의해 유도되는 TNF-α의 생성량은 44%, IL-1β의 생성량은 67%, IL-6의 생성량은 34%, MCP-1의 생성량은 69%를 효과적으로 감소됨을 확인할 수 있었다.As a result, as shown in Figure 3 when treated with a concentration of LPS 100 ng / ml in RAW 264.7 cells, TNF-α is 4.3 times, IL-1β 6.2 times, IL-6 57.3 times, MCP-1 is 5.2 times increased. After pretreatment with IJHT concentration of 300 μg / ml for 1 hour, 100 ng / ml LPS was treated for 18 hours to produce 44% of TNF-α induced by LPS, 67% of IL-1β, and IL. The production amount of -6 was found to be effectively reduced by 34%, MCP-1 production by 69%.

본 발명의 복합 생약 추출물을 포함하는 조성물의 제제예를 설명하나, 본 발명은 이를 한정하고자 함이 아닌 단지 구체적으로 설명하고자 함이다.Although the formulation example of the composition comprising the complex herbal extract of the present invention will be described, the present invention is not intended to limit it, it is intended to explain in detail only.

제제예Formulation example 1.  One. 산제의Powder 조제 pharmacy

IJHT 20 mgIJHT 20 mg

유당 100 mgLactose 100 mg

탈크 10 mgTalc 10 mg

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

제제예Formulation example 2. 정제의 제조 2. Preparation of Tablets

IJHT 10 mgIJHT 10 mg

옥수수 전분 100 mgCorn starch 100 mg

유당 100 mgLactose 100 mg

스테아린산 마그네슘 2 mg2 mg magnesium stearate

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

제제예Formulation example 3. 캅셀제의 제조 3. Manufacture of capsule

IJHT 10 mgIJHT 10 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 충진하여 캡슐제를 제조한다.According to a conventional capsule preparation method, the above ingredients are mixed and filled into gelatin capsules to prepare capsules.

제제예Formulation example 4. 주사제의 제조 4. Preparation of injections

IJHT 10 mgIJHT 10 mg

만니톨 180 mgMannitol 180 mg

주사용 멸균 증류수 2974 mgSterile distilled water for injection 2974 mg

Na2HPO4·12H2O 26 mg Na 2 HPO 4 · 12H 2 O 26 mg

통상 주사제의 제조방법에 따라 1 앰플당 (2 ml) 상기의 성분 함량으로 제조한다.It is usually prepared in the above ingredient content per ampoules (2 ml) according to the preparation method of the injection.

제제예Formulation example 5.  5. 액제의Liquid 제조 Produce

IJHT 20 mgIJHT 20 mg

이성화당 10 g10 g of isomerized sugar

만니톨 5 g5 g of mannitol

정제수 적정량Purified Water Proper Amount

통상 액제의 제조방법에 따라 정제수에 각각의 성분을 가하여 용해시키고 레몬향을 적량 가한 다음 상기의 성분을 혼합한 다음 정제수를 가하여 전체를 정제수를 가하여 전체 100 ml로 조절한 후 갈색병에 충진하여 멸균시켜 액제를 제조한다.Normally, each component is added to the purified water to dissolve according to the preparation method of the liquid solution, lemon flavor is added, the above ingredients 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 and sterilized. To prepare a liquid solution.

제제예Formulation example 6. 건강기능식품의 제조 6. Preparation of dietary supplements

IJHT 1000 mgIJHT 1000 mg

비타민 혼합물 적량Vitamin mixture proper amount

비타민 A 아세테이트 70 ㎍70 μg of Vitamin A Acetate

비타민 E 1.0 mg Vitamin E 1.0 mg

비타민 B1 0.13 mgVitamin B1 0.13 mg

비타민 B2 0.15 mgVitamin B2 0.15 mg

비타민 B6 0.5 mgVitamin B6 0.5 mg

비타민 B12 0.2 ㎍0.2 μg of vitamin B12

비타민 C 10 mgVitamin C 10 mg

비오틴 10 ㎍10 μg biotin

니코틴산아미드 1.7 mgNicotinamide 1.7 mg

엽산 50 ㎍50 μg folic acid

판토텐산 칼슘 0.5 mgCalcium Pantothenate 0.5 mg

무기질 혼합물 적량Mineral mixture

황산제1철 1.75 mgFerrous Sulfate 1.75 mg

산화아연 0.82 mgZinc Oxide 0.82 mg

탄산마그네슘 25.3 mgMagnesium carbonate 25.3 mg

제1인산칼륨 15 mg15 mg potassium monophosphate

제2인산칼슘 55 mgDicalcium Phosphate 55 mg

구연산칼륨 90 mgPotassium Citrate 90 mg

탄산칼슘 100 mgCalcium Carbonate 100 mg

염화마그네슘 24.8 mgMagnesium chloride 24.8 mg

상기의 비타민 및 미네랄 혼합물의 조성비는 비교적 건강기능식품에 적합한 성분을 바람직한 실시예로 혼합 조성하였지만, 그 배합비를 임의로 변형 실시하여도 무방하며, 통상의 건강기능식품 제조방법에 따라 상기의 성분을 혼합한 다음, 과립을 제조하고, 통상의 방법에 따라 건강기능식품 조성물 제조에 사용할 수 있다.Although the composition ratio of the above-mentioned vitamin and mineral mixtures is a composition that is relatively suitable for the health functional food, the composition is mixed in a preferred embodiment, but the compounding ratio may be arbitrarily modified, and the above ingredients are mixed according to a conventional health functional food manufacturing method. Then, the granules may be prepared and used for preparing the nutraceutical composition according to a conventional method.

제제예Formulation example 7. 건강 음료의 제조 7. Manufacture of health drinks

IJHT 1000 mg IJHT 1000 mg

구연산 1000 mgCitric acid 1000 mg

올리고당 100 g100 g oligosaccharides

매실농축액 2 gPlum concentrate 2 g

타우린 1 g1 g of taurine

정제수 전체 900 ml900 ml of purified water whole

통상의 건강음료 제조방법에 따라 상기의 성분을 혼합한 다음, 약 1시간동안 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 according to 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 then refrigerated and stored Used to prepare the healthy beverage composition of the invention.

상기 조성비는 비교적 기호음료에 적합한 성분을 바람직한 실시예로 혼합 조성하였지만, 수요계층, 수요국가, 사용용도 등 지역적, 민족적 기호도에 따라서 그 배합비를 임의로 변형 실시하여도 무방하다.Although the composition ratio is a mixture of the components suitable for the preferred beverage as a preferred embodiment, the blending ratio may be arbitrarily varied according to the regional and national preferences such as the demand level, the demanding country, and the intended use.

도 1은 복합 생약 추출물의 LPS-유도된 NO (A) 및 PGE2 생성량 (B)에 미치는 효과를 측정한 도이며, 1 is a diagram measuring the effect on the LPS-induced NO (A) and PGE 2 production (B) of the complex herbal extracts,

도 2는 복합 생약 추출물의 LPS-유도된 iNOS 및 COX-2 발현에 미치는 효과를 측정한 도이고,Figure 2 is a measure of the effect on the expression of LPS-induced iNOS and COX-2 of the complex herbal extracts,

도 3은 복합 생약 추출물의 LPS-유도된 TNF-α (A), IL-1β (B), IL-6 (C) 및 MCP-1의 생성량 (D)에 미치는 효과를 측정한 도이다.Figure 3 is a measure of the effect on the production (D) of LPS-induced TNF-α (A), IL-1β (B), IL-6 (C) and MCP-1 of the complex herbal extracts.

Claims (8)

인진, 치자 및 대황의 건조중량비 (w/w)가 10 : 5 : 3인 복합 생약 메탄올 추출물을 유효성분으로 함유하는 동맥경화증의 예방 및 치료용 약학조성물.A pharmaceutical composition for the prevention and treatment of atherosclerosis, which contains a complex herbal methanol extract having a dry weight ratio (w / w) of phosphorus, gardenia and rhubarb (w / w) of 10: 5: 3 as an active ingredient. 삭제delete 삭제delete 삭제delete 삭제delete 삭제delete 인진, 치자 및 대황의 건조중량비 (w/w)가 10 : 5 : 3인 복합 생약 메탄올 추출물을 유효성분으로 함유하는 동맥경화증의 예방 및 개선용 건강기능식품.Health functional food for the prevention and improvement of atherosclerosis, which contains a compound herbal methanol extract having a dry weight ratio (w / w) of phosphorus, gardenia and rhubarb as 10: 5: 3 as an active ingredient. 삭제delete
KR1020080021032A 2008-03-06 2008-03-06 A composition comprising the extract of complex herbs as an active ingredient for preventing and treating inflammatory disease KR101010741B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
KR1020080021032A KR101010741B1 (en) 2008-03-06 2008-03-06 A composition comprising the extract of complex herbs as an active ingredient for preventing and treating inflammatory disease

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
KR1020080021032A KR101010741B1 (en) 2008-03-06 2008-03-06 A composition comprising the extract of complex herbs as an active ingredient for preventing and treating inflammatory disease

Publications (2)

Publication Number Publication Date
KR20090095844A KR20090095844A (en) 2009-09-10
KR101010741B1 true KR101010741B1 (en) 2011-01-25

Family

ID=41296118

Family Applications (1)

Application Number Title Priority Date Filing Date
KR1020080021032A KR101010741B1 (en) 2008-03-06 2008-03-06 A composition comprising the extract of complex herbs as an active ingredient for preventing and treating inflammatory disease

Country Status (1)

Country Link
KR (1) KR101010741B1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20050094278A (en) * 2004-03-22 2005-09-27 광동제약 주식회사 A composition for preventing or treating adult diseases containing rei rhizoma
KR20070042405A (en) * 2005-10-18 2007-04-23 인제대학교 산학협력단 The water-soluble fractions from artemisia species having anti-inflammatory effect

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20050094278A (en) * 2004-03-22 2005-09-27 광동제약 주식회사 A composition for preventing or treating adult diseases containing rei rhizoma
KR20070042405A (en) * 2005-10-18 2007-04-23 인제대학교 산학협력단 The water-soluble fractions from artemisia species having anti-inflammatory effect

Also Published As

Publication number Publication date
KR20090095844A (en) 2009-09-10

Similar Documents

Publication Publication Date Title
KR101010743B1 (en) A composition comprising the extract of complex herbsKRJS as an active ingredient for preventing and treating inflammatory disease
KR101150643B1 (en) A composition comprising the compounds isolated from the Inulae Flos extract of Inula japonica Thunberg having anti-inflammatory or anti-allergic activity
KR20100011453A (en) A composition comprising the extract of melia toosendan sied. et zucc. as an active ingredient for preventing and treating oxidative stress-induced diseases and inflammatory disease
KR101072663B1 (en) A composition comprising the compound isolated from the extract of Rubiae Radix for preventing and treating inflammatory disease
KR100765416B1 (en) Composition comprising the extract of Siegesbeckiae Herba for preventing and treating arthritis
KR20090105098A (en) A composition comprising the extract of complex herbs(ims) as an active ingredient for preventing and treating inflammatory disease
KR20110017062A (en) Composition comprising ginsenoside for preventing and treating thrombotic diseases
KR20090116547A (en) A composition comprising the extract of inula flos as an active ingredient for preventing and treating inflammatory, allergy and asthmatic diseases
KR100825432B1 (en) Composition comprising the extract of Aralia cordata Thunb for the prevention or treatment of inflammation and allergic disease
KR102032110B1 (en) Composition for prevention or treatment of allergic skin diseases comprising fermented extract of ginseng
JP7117030B2 (en) A composition for preventing, treating, or ameliorating prostatic disease, containing extracts of ragweed, lokon, and red pine as active ingredients
KR100700481B1 (en) Composition comprising the extract of Cinnamomum cassia for preventing and treating fracture
KR101010741B1 (en) A composition comprising the extract of complex herbs as an active ingredient for preventing and treating inflammatory disease
KR20090128725A (en) The composition comprising extracts or fractions of magnolia obovata thunb for treating and preventing inflammation disease
KR101027573B1 (en) A new compound ponciol isolated from the mixed herbal extract and the use thereof
KR101403999B1 (en) A method for preparing a purified extract and the composition comprising the same for treating and preventing asthma and allergic disease
KR20080023570A (en) Composition comprising the extract of salvia miltiorrhiza bunge for the prevention and treatment of asthma and allergic disease
KR100547560B1 (en) Pharmaceutical composition comprising the extract of Kalopanax pictus, Pueraria thunbergiana and Rhus verniciflua having anti-inflammatory activity.
KR101448355B1 (en) Composition comprising an extract of Juncus effusus L. var. decipiens Buchen. for preventing and treating inflammatory disease or allergic disease
KR100629624B1 (en) Composition comprising the leaf extract of Rubus Coreanus having anti-inflammatory activity
KR20090114022A (en) A composition comprising the extract of complex herbs(ims) as an active ingredient for preventing and treating atherosclerosis
KR101454336B1 (en) Compositions for preventing and treating arthritis
KR100818363B1 (en) Health food comprising the extract of Aralia cordata Thunb for the prevention or improvement of inflammation and allergic disease
KR100736702B1 (en) Composition comprising an extract of Cassia mimosoides var. nomame MAKINO for the prevention and treatment of ischemic diseases
KR100665503B1 (en) Composition comprising an extract of fraxini cortex for the prevention and treatment of ischemic diseases and degenerative brain diseases

Legal Events

Date Code Title Description
A201 Request for examination
E902 Notification of reason for refusal
N231 Notification of change of applicant
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: 20140110

Year of fee payment: 4

FPAY Annual fee payment

Payment date: 20141216

Year of fee payment: 5

FPAY Annual fee payment

Payment date: 20151224

Year of fee payment: 6

FPAY Annual fee payment

Payment date: 20170104

Year of fee payment: 7

FPAY Annual fee payment

Payment date: 20180103

Year of fee payment: 8

FPAY Annual fee payment

Payment date: 20181226

Year of fee payment: 9

FPAY Annual fee payment

Payment date: 20200120

Year of fee payment: 10

R401 Registration of restoration