TW201532610A - Novel use of a dimethyl sulphoxide (DMSO) extract or fraction from graptopetalum sp - Google Patents

Novel use of a dimethyl sulphoxide (DMSO) extract or fraction from graptopetalum sp Download PDF

Info

Publication number
TW201532610A
TW201532610A TW103138926A TW103138926A TW201532610A TW 201532610 A TW201532610 A TW 201532610A TW 103138926 A TW103138926 A TW 103138926A TW 103138926 A TW103138926 A TW 103138926A TW 201532610 A TW201532610 A TW 201532610A
Authority
TW
Taiwan
Prior art keywords
disease
fraction
extract
dmso
rhodiola
Prior art date
Application number
TW103138926A
Other languages
Chinese (zh)
Other versions
TWI681774B (en
Inventor
Chi-Ying Huang
Wei-Hsiang Hsu
Hong-Jhih Jhuang
yan-xi Chen
Original Assignee
Chi-Ying Huang
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 Chi-Ying Huang filed Critical Chi-Ying Huang
Publication of TW201532610A publication Critical patent/TW201532610A/en
Application granted granted Critical
Publication of TWI681774B publication Critical patent/TWI681774B/en

Links

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)
    • A61K36/41Crassulaceae (Stonecrop family)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Natural Medicines & Medicinal Plants (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Hematology (AREA)
  • Biotechnology (AREA)
  • Psychiatry (AREA)
  • Obesity (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Hospice & Palliative Care (AREA)
  • Alternative & Traditional Medicine (AREA)
  • Diabetes (AREA)
  • Botany (AREA)
  • Medical Informatics (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Medicines Containing Plant Substances (AREA)

Abstract

The preset invention relates to a new approach for treating a metabolic disease, such as diabetes, obesity or fatty liver disease using a dimethyl sulfoxide (DMSO) extract or fraction from Graptopetalum sp. or Rhodiola sp., or an active compound isolated from the DMSO extract or fraction. Also provided in the present invention is the use of the dimethyl sulfoxide (DMSO) extract or fraction from Graptopetalum sp. or Rhodiola sp., or the active compound isolated from the DMSO extract or fraction in activating AMPK pathway and autophagy pathway, which can be used in prevention or treatment of a neurodegenerative disease or amyloid-related disease, such as Alzheimer's disease.

Description

一種縞辦屬二甲亞碸萃取物或級分之新穎用途 Novel use of dimethyl sulfoxide extract or fraction

本發明係關於一種用於治療代謝性疾病或神經退化性疾病的新方法和組合物。具體而言,本發明係關於一種使用來自縞辦屬(Graptopetalum sp.)的萃取物或級分用於治療代謝性疾病或神經退化性疾病的方法和組合物。 The present invention relates to a novel method and composition for treating a metabolic disease or a neurodegenerative disease. In particular, the present invention relates to a method and composition for treating a metabolic disease or a neurodegenerative disease using an extract or fraction derived from Graptopetalum sp.

石蓮花(Graptopetalum paraguayense,GP)是一種中國傳統草藥,具有多種保健功效。根據古老的中國藥方,GP可緩和肝的病症、降低血壓、美白肌膚、緩解疼痛和感染、抑制發炎、以及改善腦部的功能而被認為具有潛在的有益功效。 Graptopetalum paraguayense (GP) is a traditional Chinese herbal medicine with a variety of health benefits. According to the ancient Chinese prescription, GP is considered to have potential beneficial effects in alleviating liver disease, lowering blood pressure, whitening skin, relieving pain and infection, inhibiting inflammation, and improving brain function.

研究顯示該GP葉片萃取物在體外試驗中可以抑制酪胺酸酶(tyrosinase)和血管收縮素轉化酶(angiotensin-converting enzyme)的活性並清除自由基(Chen,等人,Studies on the inhibitory effect of Graptopetalum paraguayense E.Walther extracts on the angiotensin converting enzyme.Food Chemistry 100:1032-1036,2007;Chung,等人,Studies on the antioxidative activity of Graptopetalum paraguayense E.Walther.Food Chemistry 91:419-424,2005;以及Huang,等人,Studies on the inhibitory effect of Graptopetalum paraguayense E.Walther extracts on mushroom tyrosinase.Food Chemistry 89:583-587,2005)。過去已發現的是GP莖部的水萃取物和50%乙醇萃取物及95%乙醇的莖部萃取物具有抗氧化活性,其被證實對於人類肝細胞癌(Hepatocellular carcinoma,HCC)細胞株(HepG2)的增生具有抑製效果(Chen等人,In vitro antioxidant and antiproliferative activity of the stem extracts from Graptopetalum paraguayense.Am J Chin Med 2008;36:369-383)。體內的試驗研究證實GP的葉片萃取物可抑制小神經膠質細胞的活化、氧化壓力及iNOS的表現,以減少缺血性腦損傷(Kao等人,Graptopetalum paraguayense E.Walther leaf extracts protect against brain injury in ischemic rats.Am J Chin Med 2010;38:495-516)。 Studies have shown that the GP leaf extract can inhibit the activity of tyrosinase and angiotensin-converting enzyme and scavenge free radicals in vitro (Chen, et al., Studies on the inhibitory effect of Graptopetalum paraguayense E. Walther extracts on the angiotensin converting enzyme. Food Chemistry 100: 1032-1036, 2007; Chung, et al, Studies on the antioxidative activity of Graptopetalum paraguayense E. Walther. Food Chemistry 91: 419-424, 2005; Huang, et al, Studies on the inhibitory effect of Graptopetalum paraguayense E. Walther extracts on mushroom tyrosinase. Food Chemistry 89: 583-587, 2005). It has been found in the past that the water extract of GP stem and the stem extract of 50% ethanol extract and 95% ethanol have antioxidant activity, which has been confirmed for human hepatocellular carcinoma (HCC) cell line (HepG2). Proliferation has an inhibitory effect (Chen et al, In vitro antioxidant and antiproliferative activity of the stem extracts from Graptopetalum paraguayense. Am J Chin Med 2008; 36: 369-383). In vivo studies have confirmed that GP leaf extract can inhibit microglia activation, oxidative stress and iNOS expression to reduce ischemic brain damage (Kao et al, Graptopetalum paraguayense E. Walther leaf extracts protect against brain injury in Ischemic rats. Am J Chin Med 2010; 38: 495-516).

在2004年由Hsu提出申請並在2008年獲證的美國專利號7,364,758中已揭示來自縞辦屬的乙醇萃取物具有體內和體外抗肝纖維化和抗發炎功效。然後,其部分連續申請案美國專利號7,588,776在2008年被提出申請並在2009年獲證,其指出縞辦屬的水溶性級分係有效於治療肝的疾病或症狀,例如發炎、脂肪變性及纖維化。 It has been disclosed in U.S. Patent No. 7,364,758, issued to Hsu in 2004, which is hereby incorporated by reference in its entirety in U.S. Patent No. 7,364,758, which is incorporated herein by reference. Then, part of its continuous application US Patent No. 7,588,776 was filed in 2008 and certified in 2009, which states that the water-soluble fraction of the genus is effective in treating diseases or symptoms of the liver, such as inflammation and steatosis. Fibrosis.

在2011年提出申請並在2012年10月11日公開的美國專利公開第20120259004號中亦揭示藉由使用二甲亞碸(DMSO)萃取選自於由縞辦屬、紅景天屬(Rhodiola sp.)及石蓮花屬(Echeveria sp.)所組成之群組的植物所製備的新萃取物和級分、以及分離自該新萃取物的新化合物可有效治療癌症或纖維化,例如肝癌或肝纖維化。 Filed in 2011 and US Patent October 11, 2012 Publication No. 20120259004 also revealed by the use of dimethyl sulfoxide (DMSO) extract is selected from the stripe do genus Rhodiola (Rhodiola sp New extracts and fractions prepared from plants of the group consisting of Echeveria sp., and new compounds isolated from the new extract are effective in treating cancer or fibrosis, such as liver cancer or liver Fibrosis.

本發明係關於一種縞辦屬(Graptopetalum sp.)或紅景天屬(Rhodiola sp.)之二甲亞碸(DMSO)萃取物或級分及分離自該DMSO萃取 物或級分的活性化合物用於治療代謝性疾病,例如糖尿病或脂肪肝疾病,或神經退化性疾病的新用途。 The present invention relates to a dimethyl sulfoxide (DMSO) extract or fraction of Graptopetalum sp. or Rhodiola sp. and an active compound isolated from the DMSO extract or fraction. For the treatment of metabolic diseases, such as diabetes or fatty liver disease, or new uses of neurodegenerative diseases.

在一方面,本發明提供一種用於治療代謝性疾病的方法,包含投予一組合物於所需個體,該組合物包含一治療有效量來自縞辦屬或紅景天屬的二甲亞碸(DMSO)萃取物或級分、或分離自該DMSO萃取物或級分的活性化合物。 In one aspect, the invention provides a method for treating a metabolic disease comprising administering a composition to a subject in need thereof, the composition comprising a therapeutically effective amount of dimethyl hydrazine from the genus Rhododendron or Rhodiola (DMSO) extract or fraction, or active compound isolated from the DMSO extract or fraction.

在另一方面,本發明提供一種組合物用於製備治療代謝性疾病的藥物之用途;其中該組合物包含來自縞辦屬或紅景天屬的二甲亞碸(DMSO)萃取物或級分、或分離自該DMSO萃取物或級分的活性化合物。 In another aspect, the invention provides the use of a composition for the manufacture of a medicament for the treatment of a metabolic disorder; wherein the composition comprises a dimethyl hydrazine (DMSO) extract or fraction from the genus Rhododendron or Rhodiola Or an active compound isolated from the DMSO extract or fraction.

在再一方面,本發明提供一種用於治療代謝性疾病的醫藥組合物;其中該組合物包含來自縞辦屬或紅景天屬的二甲亞碸(DMSO)萃取物或級分、或分離自該DMSO萃取物或級分的活性化合物。 In still another aspect, the present invention provides a pharmaceutical composition for treating a metabolic disease; wherein the composition comprises a dimethyl hydrazine (DMSO) extract or fraction, or a separation from the genus Rhododendron or Rhodiola The active compound from the DMSO extract or fraction.

在本發明之一具體實施例中,該代謝性疾病為糖尿病。 In a specific embodiment of the invention, the metabolic disease is diabetes.

在本發明之另一具體實施例中,該代謝性疾病為肥胖。 In another embodiment of the invention, the metabolic disease is obesity.

在本發明之第三具體實施例中,該代謝性疾病為脂肪肝疾病,包括酒精性脂肪肝疾病(alcoholic fatty liver disease,AFLD)或非酒精性脂肪肝疾病(nonalcoholic fatty liver disease,NAFLD)。 In a third embodiment of the present invention, the metabolic disease is a fatty liver disease, including alcoholic fatty liver disease (AFLD) or nonalcoholic fatty liver disease (NAFLD).

在再一方面,本發明提供一種使用組合物治療B型肝炎病毒(Hepatitis B virus,HBV)相關的肝相關疾病(例如HCC、肝纖維化或肝硬化)的方法、用途或組合物,該組合物包含來自縞辦屬或紅景天屬的二甲亞碸(DMSO)萃取物或級分、或分離自該DMSO萃取物或級分的活性化合物。 In still another aspect, the present invention provides a method, use, or composition for treating a hepatitis B virus (HBV)-related liver-related disease (eg, HCC, liver fibrosis, or cirrhosis) using a composition, the combination The composition comprises a dimethyl hydrazine (DMSO) extract or fraction from the genus Rhodiola or Rhodiola, or an active compound isolated from the DMSO extract or fraction.

在又一方面,本發明提供一種使用組合物治療糖尿病相關的肝相關疾病(例如HCC)的方法、用途或組合物,該組合物包含來自縞辦屬或紅景天屬的二甲亞碸(DMSO)萃取物或級分、或分離自該DMSO萃取物或級分的活性化合物。 In yet another aspect, the present invention provides a method, use, or composition for treating a diabetes-related liver-related disease (eg, HCC) using a composition comprising dimethyl hydrazine from the genus Rhododendron or Rhodiola ( DMSO) extract or fraction, or active compound isolated from the DMSO extract or fraction.

在又再一方面,本發明提供一種使用組合物治療肥胖相關的肝相關疾病的方法、用途或組合物,該組合物包含來自縞辦屬或紅景天屬的二甲亞碸(DMSO)萃取物或級分、或分離自該DMSO萃取物或級分的活性化合物。 In still another aspect, the present invention provides a method, use or composition for treating obesity-related liver-related diseases using a composition comprising dimethyl sulfoxide (DMSO) extraction from the genus Rhododendron or Rhodiola Or fraction, or an active compound isolated from the DMSO extract or fraction.

在再一方面,本發明提供一種使用組合物治療肥胖引起的高血膽固醇或高量三酸甘油酯的方法、用途或組合物,該組合物包含來自縞辦屬或紅景天屬的二甲亞碸(DMSO)萃取物或級分、或分離自該DMSO萃取物或級分的活性化合物。 In still another aspect, the present invention provides a method, use or composition for treating obesity-induced hypercholesterolemia or high triglyceride using a composition comprising dimethyl or erythromycin from the genus Rhodiola A guanidine (DMSO) extract or fraction, or an active compound isolated from the DMSO extract or fraction.

在再更一方面,本發明提供一種使用組合物預防或治療代謝症候群的方法、用途或組合物,該組合物包含來自縞辦屬或紅景天屬的二甲亞碸(DMSO)萃取物或級分、或分離自該DMSO萃取物或級分的活性化合物。 In still a further aspect, the present invention provides a method, use or composition for preventing or treating a metabolic syndrome using a composition comprising a dimethyl hydrazine (DMSO) extract from the genus Rhodiola or Rhodiola or Fractions, or active compounds isolated from the DMSO extract or fraction.

在再更一方面,本發明提供一種使用醫藥組合物預防或治療癌症的協同方法或組合物,該醫藥組合物包含來自縞辦屬或紅景天屬的二甲亞碸(DMSO)萃取物或級分、或分離自該DMSO萃取物或級分的活性化合物,並與一抗癌藥物組合。 In still a further aspect, the present invention provides a synergistic method or composition for preventing or treating cancer using a pharmaceutical composition comprising dimethyl hydrazine (DMSO) extract from the genus Rhododendron or Rhodiola or Fractions, or active compounds isolated from the DMSO extract or fraction, are combined with an anti-cancer drug.

在本發明之一具體實施例中,該癌症為肝癌,例如肝細胞癌(Hepatocellular carcinoma,HCC),而且該抗癌藥物為抗肝癌藥物,例如蕾 莎瓦(Sorafenib)。 In a specific embodiment of the present invention, the cancer is liver cancer, such as Hepatocellular carcinoma (HCC), and the anticancer drug is an anti-hepatocarcinoma drug, such as bud Sorafenib.

本發明亦提供一種活化AMPK途徑或自噬途徑的方法、用途或組合物,其中該組合物為來自縞辦屬或紅景天屬的二甲亞碸(DMSO)萃取物或級分、或分離自該DMSO萃取物或級分的活性化合物。 The invention also provides a method, use or composition for activating an AMPK pathway or an autophagy pathway, wherein the composition is a dimethyl hydrazine (DMSO) extract or fraction, or isolated from the genus Rhodiola or Rhodiola The active compound from the DMSO extract or fraction.

此外,本發明亦提供一種使用組合物預防或治療神經退化性疾病的方法或組合物,該組合物包含來自縞辦屬或紅景天屬的二甲亞碸(DMSO)萃取物或級分、或分離自該DMSO萃取物或級分的活性化合物。 Further, the present invention also provides a method or composition for preventing or treating a neurodegenerative disease using a composition comprising a dimethyl hydrazine (DMSO) extract or fraction derived from the genus Rhododendron or Rhodiola, Or an active compound isolated from the DMSO extract or fraction.

在本發明之一具體實施例中,該神經退化性疾病為帕金森氏症(Parkinson’s disease)、阿茲海默症(Alzheimer’s disease)、或杭丁頓氏症(Huntington’s disease)。 In a specific embodiment of the invention, the neurodegenerative disease is Parkinson's disease, Alzheimer's disease, or Huntington's disease.

此外,本發明亦提供一種使用組合物預防或治療澱粉樣蛋白相關疾病的方法、用途或組合物,該組合物包含來自縞辦屬或紅景天屬的二甲亞碸(DMSO)萃取物或級分、或分離自該DMSO萃取物或級分的活性化合物。 Further, the present invention provides a method, use or composition for preventing or treating an amyloid-related disease using a composition comprising dimethyl hydrazine (DMSO) extract from the genus Rhododendron or Rhodiola or Fractions, or active compounds isolated from the DMSO extract or fraction.

在本發明之一具體實施例中,該澱粉樣蛋白相關疾病為阿茲海默症、第2型糖尿病、帕金森氏症、杭丁頓氏症、致死性家族失眠症(Fatal Familial Insomnia)、或類風濕性關節炎(Rheumatoid arthritis)。 In a specific embodiment of the present invention, the amyloid-related diseases are Alzheimer's disease, type 2 diabetes, Parkinson's disease, Huntington's disease, Fatal Familial Insomnia, Or rheumatoid arthritis (Rheumatoid arthritis).

此外,本發明亦提供一種使用組合物抗老化的方法、用途或組合物,該組合物包含來自縞辦屬或紅景天屬的二甲亞碸(DMSO)萃取物或級分、或分離自該DMSO萃取物或級分的活性化合物。 Further, the present invention also provides a method, use or composition for using the composition against aging, comprising dimethyl hydrazine (DMSO) extract or fraction from genus Rhodiola or Rhodiola, or isolated from The DMSO extract or fraction of the active compound.

在本發明之一具體實施例中,縞辦屬為石蓮花(Graptopetalum paraguayense)。 In one embodiment of the invention, the genus is Graptopetalum paraguayense .

在本發明之一具體實施例中,紅景天屬為紅景天(Rhodiola rosea)。 In a specific embodiment of the invention, the Rhodiola rosea is Rhodiola rosea .

當結合附圖閱讀時,將更好地理解前面的概述、以及以下本發明的實施方式。為了說明本發明的目的,在圖式中顯示目前較佳的具體實施例。然而,應當理解的是,本發明並不限於圖式中顯示的具體實施例。 The foregoing summary, as well as the following embodiments of the present invention, For the purpose of illustrating the invention, the present preferred embodiments are shown in the drawings. However, it should be understood that the invention is not limited to the specific embodiments shown in the drawings.

在圖式中:圖1A-1B顯示HH-F3在Hep3B/T2細胞中抑制8-Br-cAMP/迪皮質醇(dexamethasone)誘導的醣異生酶(gluconeogenic enzyme)基因表現的效果。圖1A提供以8-溴-cAMP(8-Br-cAMP)、迪皮質醇(Dex)單獨或同時處理經培養的人類肝腫瘤Hep3B/T2細胞30分鐘的結果;其中藉由即時定量PCR量測醣異生基因、葡萄糖-6-磷酸酶(glucose-6-phosphataseG6Pase)及磷酸烯醇丙酮酸羧激酶(phosphoenol pyruvate carboxykinasePEPCK)的mRNAs並使用β-肌動蛋白當作標準。使用胰島素作為正對照組。圖1B提供以8-Br-cAMP(8-Br-cAMP)、迪皮質醇(Dex)單獨或同時處理經培養的人類肝腫瘤Hep3B/T2細胞30分鐘,然後使用不同濃度的HH-F3和胰島素處理的結果;其中藉由即時定量PCR量測醣異生基因、葡萄糖-6-磷酸酶(G6Pase)及磷酸烯醇丙酮酸羧激酶(PEPCK)的mRNAs並使用β-肌動蛋白當作標準。 In the drawings: Figures 1A-1B show the effect of HH-F3 on inhibition of 8-Br-cAMP/dexamethasone-induced gluconeogenic enzyme gene expression in Hep3B/T2 cells. Figure 1A provides the results of treatment of cultured human liver tumor Hep3B/T2 cells with 8-bromo-cAMP (8-Br-cAMP), decortisol (Dex) alone or simultaneously for 30 minutes; mRNAs of gluconeogenesis genes, glucose-6-phosphatase ( G6Pase ) and phosphoenol pyruvate carboxykinase ( PEPCK ) were determined using β-actin as a standard. Insulin was used as a positive control group. Figure 1B provides treatment of cultured human liver tumor Hep3B/T2 cells with 8-Br-cAMP (8-Br-cAMP), decortisol (Dex) alone or simultaneously for 30 minutes, followed by different concentrations of HH-F3 and insulin. The results of the treatment; wherein mRNAs of the gluconeogenesis gene, glucose-6-phosphatase ( G6Pase ) and phosphoenolpyruvate carboxykinase ( PEPCK ) were measured by real-time quantitative PCR and β-actin was used as a standard.

圖2A-2C顯示HH-F3在Hep3B/T2細胞中抑制8-Br-cAMP/Dex誘導的醣異生共活化劑PGC-1α表現活性的效果。圖2A提供在無血清的DMEM培養基中以8-Br-cAMP、迪皮質醇(Dex)單獨或同時處理、並與HH-F3 組合處理經培養的人類肝腫瘤Hep3B/T2細胞24小時的結果,其中藉由即時定量PCR量測PGC-1α mRNAs並標準化為β-肌動蛋白,以及使用胰島素作為正對照組。圖2B顯示使用核萃取物處理的Hep3B/T2細胞中表現的PGC-1α和HNF-4α蛋白水平,其係藉由西方墨漬法分析測定,其中B23係使用做為標準;Dex:迪皮質醇(Dexamethasone);其中此等是來自3組獨立實驗的代表性數據。圖2C提供以HH-F3處理Hep3B/T2細胞、然後以抗PGC1-α、HNF-4α、及FoxO1的抗體進行西方墨漬法分析的結果;其中此等是來自3組獨立實驗的代表性數據。 2A-2C show the effect of HH-F3 in inhibiting the expression activity of 8-Br-cAMP/Dex-induced gluconeogenesis co-activator PGC-1α in Hep3B/T2 cells. Figure 2A provides the results of treatment of cultured human liver tumor Hep3B/T2 cells with 8-Br-cAMP, decortisol (Dex) alone or simultaneously in serum-free DMEM medium, and 24 hours, in combination with HH-F3, Among them, PGC-1α mRNAs were measured by real-time quantitative PCR and normalized to β-actin, and insulin was used as a positive control group. Figure 2B shows the levels of PGC-1α and HNF-4α protein expressed in Hep3B/T2 cells treated with nuclear extracts as determined by Western blotting analysis, in which B23 is used as a standard; Dex: dicortisol (Dexamethasone); these are representative data from 3 independent experiments. Figure 2C provides the results of Western blot analysis of Hep3B/T2 cells treated with HH-F3 followed by antibodies against PGC1-α, HNF-4α, and FoxO1; these are representative data from 3 independent experiments. .

圖3A-3B顯示HH-F3在經培養的人類肝腫瘤Hep3B/T2細胞中通過活化AMPK來抑制醣異生酶基因表現的效果。圖3A係以8-Br-cAMP加上迪皮質醇預處理經培養的人類肝腫瘤Hep3B/T2細胞30分鐘的結果。圖3B顯示以8-Br-cAMP加上迪皮質醇預處理經培養的人類肝腫瘤Hep3B/T2細胞30分鐘,然後在無血清的DMEM中使用不同濃度的HH-F3處理24小時的結果;其中以葡萄糖-6-磷酸酶啟動子驅動的螢光素酶報告質體轉染Hep3B/T2細胞,而且該將螢光素酶的活性係由與對照組相比的共轉染pCMV-β-半乳糖苷酶質體以β-半乳糖苷酶的活性標準化。圖3C顯示環狀AMP/DEX刺激G6Pase啟動子活性的結果,其係在HH-F3或HH-F3與AMPK抑製劑化合物C存在下進行檢驗;其中在一天的處理之後,製備細胞溶解產物以用於螢光素酶活性分析;其中使用胰島素作為正對照;以及使用二甲雙胍作為AMPK正對照。 3A-3B show the effect of HH-F3 on the inhibition of gluconeogenesis gene expression by activating AMPK in cultured human liver tumor Hep3B/T2 cells. Figure 3A shows the results of pre-treatment of cultured human liver tumor Hep3B/T2 cells with 8-Br-cAMP plus dicortisol for 30 minutes. Figure 3B shows the results of pretreatment of cultured human liver tumor Hep3B/T2 cells with 8-Br-cAMP plus dicorticol for 30 minutes, followed by treatment with different concentrations of HH-F3 for 24 hours in serum-free DMEM; The luciferase reporter plastid driven by the glucose-6-phosphatase promoter was transfected into Hep3B/T2 cells, and the luciferase activity was co-transfected with pCMV-β-half compared to the control group. The lactobionase plastid is normalized by the activity of β-galactosidase. Figure 3C shows the results of cyclic AMP/DEX-stimulated G6Pase promoter activity, which was tested in the presence of HH-F3 or HH-F3 and AMPK inhibitor Compound C; wherein after one day of treatment, cell lysates were prepared for use. Analysis of luciferase activity; in which insulin was used as a positive control; and metformin was used as a positive control for AMPK.

圖4A-4B顯示HH-F3在Hep3B/T2細胞中抑制8-Br-cAMP/Dex誘導的HBV核心啟動子活性的效果。圖4A顯示啟動子活性檢驗的結果;其 中以HBV核心啟動子(core promoter,CP)和HBV X啟動子(X promoter,XP)驅動的螢光素酶報告質體轉染Hep3B/T2細胞。圖4B顯示在無血清的DMEM中、在不存在或存在不同濃度的HH-F3下以8-Br-cAMP/Dex處理1天之後的結果;其中製備細胞溶解產物係用於螢光素酶活性分析;其中該螢光素酶的活性係由共轉染的pCMV-β-半乳糖苷酶質體以β-半乳糖苷酶活性標準化;且胰島素係使用為正對照組。 4A-4B show the effect of HH-F3 in inhibiting 8-Br-cAMP/Dex-induced HBV core promoter activity in Hep3B/T2 cells. Figure 4A shows the results of a promoter activity assay; The luciferase reporter plastids driven by the HBV core promoter (CP) and the HBV X promoter (X promoter, XP) were transfected into Hep3B/T2 cells. Figure 4B shows the results after treatment with 8-Br-cAMP/Dex for 1 day in serum-free DMEM in the absence or presence of different concentrations of HH-F3; wherein cell lysates were prepared for luciferase activity Analysis; wherein the activity of the luciferase was normalized by β-galactosidase activity from the co-transfected pCMV-β-galactosidase plastid; and the insulin system was used as a positive control group.

圖5A-5E顯示HH-F3在Hep3B/T2或1.3ES2細胞中抑制HBV表面抗原、基因表現、醣異生酶表現、及HBV DNA水平的效果。圖5A提供之影像顯示使用不同濃度的HH-F3在無血清的DMEM培養基中處理48小時經培養的人類肝腫瘤Hep3B/T2細胞;其中藉由ELISA測定HBV表面抗原並以MTT檢驗標準化;其中使用胰島素作為正對照組。圖5B提供以不同濃度的GP和HH-F3在無血清的DMEM培養基中處理48小時經培養的1.3ES2細胞之結果,其中藉由即時定量PCR量測醣異生基因、G6Pase、PEPCK、及PGC-1α mRNAs的表現,並標準化為β-肌動蛋白;使用HE-145作為正對照組。圖5C提供以不同濃度的GP和HH-F3在無血清的DMEM培養基中處理48小時經培養的1.3ES2細胞之結果,其中藉由即時定量PCR量測HBV mRNA的表現,並使用β-肌動蛋白當作標準;使用HE-145作為正對照組。圖5D提供以HH-F3處理1.3ES2細胞,然後使用對抗核蛋白的抗體進行西方墨漬法分析的結果;其中此等是來自3組獨立實驗的代表性數據。使用HE-145作為正對照組。圖5E提供以不同濃度的GP和HH-F3在無血清的DMEM培養基中處理48小時經培養的1.3ES2細胞之結果,其中藉由即時定量PCR量測培養基中的野生型HBV DNA並使用β-肌動蛋白當作標準;使用HE-145作為正對照 組。 Figures 5A-5E show the effect of HH-F3 on inhibition of HBV surface antigen, gene expression, gluconeogenesis performance, and HBV DNA levels in Hep3B/T2 or 1.3 ES2 cells. Figure 5A provides images showing cultured human liver tumor Hep3B/T2 cells treated with different concentrations of HH-F3 in serum-free DMEM medium for 48 hours; HBV surface antigen was determined by ELISA and normalized by MTT assay; Insulin was used as a positive control group. Figure 5B provides the results of treatment of cultured 1.3 ES2 cells with different concentrations of GP and HH-F3 in serum-free DMEM medium for 48 hours, wherein gluconeogenesis genes, G6Pase, PEPCK , and PGC were measured by real-time quantitative PCR. The expression of -1α mRNAs was normalized to β-actin; HE-145 was used as a positive control group. Figure 5C provides the results of treatment of cultured 1.3 ES2 cells with different concentrations of GP and HH-F3 in serum-free DMEM medium for 48 hours, in which the performance of HBV mRNA was measured by real-time quantitative PCR and β-muscle movement was used. Protein was used as a standard; HE-145 was used as a positive control group. Figure 5D provides the results of Western blot analysis using 1.3H2 cells treated with HH-F3 followed by antibodies against nuclear proteins; these are representative data from 3 independent experiments. HE-145 was used as a positive control group. Figure 5E provides the results of treatment of cultured 1.3 ES2 cells with different concentrations of GP and HH-F3 in serum-free DMEM medium for 48 hours, wherein wild-type HBV DNA in the medium was measured by real-time quantitative PCR and β- was used. Actin was used as a standard; HE-145 was used as a positive control group.

圖6A-6B顯示GP萃取物和HH-F3在HCC中降低脂肪酸合成活性的效果,其顯示GP萃取物和HH-F3的處理降低了HCC中的脂肪酸合成活性。圖6A顯示以GP萃取物和HH-F3處理Huh7和Mahlavu細胞、然後使用抗磷酸化-AMPK(P-AMPK)、AMPK、SREBP2、磷酸化-ACC(Ser79)(P-ACC)、ACC、FASN及GAPDH的抗體進行西方墨漬法分析(n=3)的結果。圖6B顯示以HH-F3處理細胞然後使用抗P-AMPK、AMPK、SREBP2、P-ACC、ACC、FASN、GAPD及β-肌動蛋白的抗體進行西方墨漬法分析的結果,其中以BNL細胞來產生皮下腫瘤,並將小鼠分成兩組:一組作為未處理的對照組(3隻不同的小鼠),而其他四隻小鼠每天經由口服(P.O.)途徑使用HH-F3(20mg/天)處理3週;3週之後,犧牲小鼠並均勻化腫瘤,隨後進行西方墨漬法及定量。 6A-6B show the effect of GP extract and HH-F3 in reducing fatty acid synthesis activity in HCC, which shows that treatment of GP extract and HH-F3 reduces fatty acid synthesis activity in HCC. Figure 6A shows treatment of Huh7 and Mahlavu cells with GP extract and HH-F3, followed by anti-phospho-AMPK (P-AMPK), AMPK, SREBP2, phosphorylation-ACC (Ser 79 ) (P-ACC), ACC, The antibodies of FASN and GAPDH were analyzed by Western blotting analysis (n=3). Figure 6B shows the results of Western blot analysis using cells treated with HH-F3 followed by antibodies against P-AMPK, AMPK, SREBP2, P-ACC, ACC, FASN, GAPD and β-actin, with BNL cells To produce subcutaneous tumors, the mice were divided into two groups: one group as an untreated control group (3 different mice), and the other four mice used HH-F3 (20 mg/day) via the oral (PO) route every day. Days) 3 weeks; after 3 weeks, the mice were sacrificed and the tumors were homogenized, followed by Western blotting and quantification.

圖7A提供之影像顯示以GP/HH-F3處理以降低油酸(Oleic Acid,OA)和棕櫚酸(Palmitic acid,PA)誘導的脂質累積的Huh7細胞;其中GP/HH-F3在Huh7中降低了由油酸(OA)和棕櫚酸(PA)誘導的脂質累積;使用油酸(1mM)和棕櫚酸(2mM)培育Huh7細胞24小時和48小時,產生了藉由油紅O(oil red O)染色可見的明顯細胞內脂質累積及由MTT檢驗的細胞存活率;其中該原始放大倍率為400倍,其係使用GP/HH-F3處理48小時(OA+PA/GP:油酸和棕櫚酸與GP共處理;OA/HH-F3:油酸和棕櫚酸與HH-F3共處理)。 Figure 7A provides images showing Huh7 cells treated with GP/HH-F3 to reduce lipid accumulation induced by oleic acid (OA) and palmitic acid (PA); wherein GP/HH-F3 is reduced in Huh7 Lipid accumulation induced by oleic acid (OA) and palmitic acid (PA); Huh7 cells were incubated with oleic acid (1 mM) and palmitic acid (2 mM) for 24 hours and 48 hours, resulting in oil red O (oil red O) Significant intracellular lipid accumulation visible by staining and cell viability by MTT assay; wherein the original magnification was 400-fold, which was treated with GP/HH-F3 for 48 hours (OA+PA/GP: oleic acid and palmitic acid) Co-treatment with GP; OA/HH-F3: oleic acid and palmitic acid co-treated with HH-F3).

圖7B提供Huh7細胞之計數(油酸和棕櫚酸)的結果,其中使用油酸和棕櫚酸(OA+PA)處理對照組,並將細胞分別使用油酸和棕櫚 酸與GP萃取物(GP)或HH-F3共處理24小時或48小時,以減少脂肪負荷,並藉由油紅O將細胞內脂質液滴染色來定量(OA+PA/GP:油酸和棕櫚酸與GP共處理;OA/HH-F3:油酸和棕櫚酸與HH-F3共處理)。 Figure 7B provides the results of Huh7 cell counts (oleic acid and palmitic acid) in which the control group was treated with oleic acid and palmitic acid (OA + PA) and the cells were separately treated with oleic acid and palm The acid is co-treated with GP extract (GP) or HH-F3 for 24 or 48 hours to reduce fat load and quantify by staining intracellular lipid droplets with Oil Red O (OA+PA/GP: oleic acid and Palmitic acid was co-treated with GP; OA/HH-F3: oleic acid and palmitic acid co-treated with HH-F3).

圖8A-8D顯示GP/HH-F3和蕾莎瓦(Sorafenib)的組合對Huh7之細胞存活率的協同作用。圖8A提供分別在蕾莎瓦(5μM和10μM)存在或不存在下使用各種濃度的GP萃取物處理Huh7細胞的結果;其中藉由SRB檢驗測定細胞存活率;並將數據表示為平均值±三個獨立實驗的標準差(SD)。圖8B提供分別在蕾莎瓦(5μM和10μM)存在或不存在下使用各種濃度的HH-F3萃取物處理Huh7細胞的結果;其中藉由SRB檢驗測定細胞存活率;並將數據表示為平均值±三個獨立實驗的標準差(SD)。圖8C提供等效線圖分析的結果,證明在Huh7細胞中72小時,蕾莎瓦和各種濃度的GP萃取物之間有協同相互作用。圖8D提供等效線圖分析的結果,證明在Huh7細胞中72小時,蕾莎瓦和各種濃度的HH-F3之間有協同相互作用。 Figures 8A-8D show the synergistic effect of the combination of GP/HH-F3 and Sorafenib on cell viability of Huh7. Figure 8A provides the results of treatment of Huh7 cells with various concentrations of GP extract in the presence or absence of Lysawa (5 μM and 10 μM); cell viability was determined by SRB assay; data were expressed as mean ± three The standard deviation (SD) of an independent experiment. Figure 8B provides the results of treatment of Huh7 cells with various concentrations of HH-F3 extract in the presence or absence of Lysawa (5 μM and 10 μM); cell viability was determined by SRB assay; data was expressed as mean ± standard deviation (SD) of three independent experiments. Figure 8C provides the results of an isobologram analysis demonstrating a synergistic interaction between Lysawa and various concentrations of GP extract at 72 hours in Huh7 cells. Figure 8D provides the results of an isobologram analysis demonstrating a synergistic interaction between Lysawa and various concentrations of HH-F3 at 72 hours in Huh7 cells.

圖9顯示HH-F3在初代皮層神經元中對Aβ25-35誘導的神經毒性之影響;其中使用載體(0.1%的DMSO)或HH-F3預處理初代皮層神經元2小時,隨後曝露於10μM的Aβ25-3540小時;以及培育之後,藉由MTT檢驗檢測細胞存活率;其中數據為來自至少三組獨立實驗的平均±SD,並以相對於對照細胞表示,而且以*指出使用Aβ和HH-F3加上Aβ處理的細胞之間的顯著差異(p<0.05)。 Figure 9 shows the effect of HH-F3 on Aβ 25-35- induced neurotoxicity in primary cortical neurons; pre-treatment of primary cortical neurons with vehicle (0.1% DMSO) or HH-F3 for 2 hours followed by exposure to 10 μM Aβ 25-35 for 40 hours; and after incubation, cell viability was determined by MTT assay; data were mean ± SD from at least three independent experiments and expressed relative to control cells, with * indicating use of Aβ and Significant differences between HH-F3 plus A[beta] treated cells (p<0.05).

圖10A-10C顯示HH-F3在swAPP695-SH-SY5Y細胞培養中對細胞外Aβ40和Aβ42水平的影響;其中使用載體或HH-F3培育APP695-SHSY-5Y-(使用人類APP695轉染的SHSY-5Y神經細胞瘤細胞)24 小時。圖10A顯示細胞的細胞存活率,其係藉由MTT檢驗檢測。圖10B顯示細胞外Aβ40的水平,其係藉由ELISA測定。圖10C顯示細胞外Aβ42的水平,其係藉由ELISA測定。 Figures 10A-10C show the effect of HH-F3 on extracellular Aβ 40 and Aβ 42 levels in swAPP 695 -SH-SY5Y cell culture; in which APP 695 -SHSY-5Y- was incubated with vector or HH-F3 (using human APP695) Dyeed SHSY-5Y neuroblastoma cells) for 24 hours. Figure 10A shows cell viability of cells as determined by MTT assay. Figure 10B shows the level of extracellular A[beta] 40 as determined by ELISA. Figure 1OC shows the level of extracellular A[beta] 42 as determined by ELISA.

圖11A-11D顯示HH-F3對APP處理和Aβ清除的影響。圖11A提供以HH-F3或γSI處理SH-SY5Y-APP695細胞24小時的結果。藉由免疫轉印法量測APP(抗Aβ1-16抗體)、APPX-F(抗Aβ1-40抗體)、及APP-CTF(抗APPct抗體)與細胞內Aβ降解酶的水平並顯示APP-CTF/肌動蛋白的相對水平。圖11B提供以HH-F3處理SH-SY5Y-APP695細胞24小時、然後使用抗胰島素降解酶(insulin-degrading enzyme,IDE)和腦啡肽酶(Neprilysin,NEP)的抗體進行西方墨漬法分析的結果;其中此等是來自3組獨立實驗的代表性數據。圖11C提供以指定濃度的HH-F3培育SH-SY5Y-APP695條件培養基24小時的結果。該剩餘的Aβ1-40係藉由ELISA量測。圖11D提供以指定濃度的HH-F3培育SH-SY5Y-APP695條件培養基24小時的結果。該條件培養基中的NEP和IDE水平係藉由免疫轉印法量測的。 Figures 11A-11D show the effect of HH-F3 on APP treatment and Aβ clearance. Figure 11A provides the results of treating SH-SY5Y-APP695 cells with HH-F3 or γSI for 24 hours. The levels of APP (anti-Aβ1-16 antibody), APPX-F (anti-Aβ1-40 antibody), APP-CTF (anti-APPct antibody) and intracellular Aβ degrading enzyme were measured by immunotransfer method and APP-CTF was displayed. / Relative level of actin. Figure 11B provides treatment of SH-SY5Y-APP695 cells with HH-F3 for 24 hours, followed by Western blot analysis using antibodies against insulin-degrading enzyme (IDE) and enkephalinase (Neprilysin, NEP) Results; these are representative data from 3 independent experiments. Figure 11C provides the results of culturing SH-SY5Y-APP695 conditioned medium for 24 hours at the indicated concentrations of HH-F3. The remaining Aβ1-40 was measured by ELISA. Figure 11D provides the results of culturing SH-SY5Y-APP695 conditioned medium for 24 hours at the indicated concentrations of HH-F3. The levels of NEP and IDE in the conditioned medium were measured by immunotransfer.

圖12顯示HH-F3在swAPP695-SH-SY5Y細胞中對自噬活性的影響;其中以載體或HH-F3培育APP695-SHSY-5Y-24小時。藉由西方墨漬法檢測自噬標記LC3。 Figure 12 shows the effect of HH-F3 on autophagy activity in swAPP 695 -SH-SY5Y cells; APP 695 -SHSY-5Y was incubated with vehicle or HH-F3 for 24 hours. Autophagy marker LC3 was detected by Western blotting.

圖13A-13B顯示HH-F3在抑制Aβ凝聚上的效果。圖13A提供未以Aβ培育24小時的HH-F3之結果。檢測Th-T的螢光(Ex 440/Em 482)。圖13B提供以Aβ1-40培育24小時的HH-F3之結果。檢測Th-T的螢光(Ex 440/Em 482)。C=PBS;V=載體(Aβ1-40)。圖13C提供以Aβ1-42培育24小時的HH-F3之結果。檢測Th-T的螢光(Ex 440/Em 482)。C=PBS;V=載 體(Aβ1-42)。 Figures 13A-13B show the effect of HH-F3 on inhibition of A[beta] aggregation. Figure 13A provides the results of HH-F3 not incubated with Aβ for 24 hours. Fluorescence of Th-T was detected (Ex 440/Em 482). Figure 13B provides the results of HH-F3 incubated with Aβ1-40 for 24 hours. Fluorescence of Th-T was detected (Ex 440/Em 482). C = PBS; V = vector (Aβ1-40). Figure 13C provides the results of HH-F3 incubated with A? 1-42 for 24 hours. Fluorescence of Th-T was detected (Ex 440/Em 482). C=PBS; V=Load Body (Aβ 1-42).

圖14顯示HH-F3在Aβ25-35誘導的細胞毒性上的保護效果。以指定濃度的HH-F3預處理初代皮層神經元2小時。隨後,使用10μM的Aβ25-35刺激細胞46小時。細胞存活率係藉由MTT測試量測。 Figure 14 shows the protective effect of HH-F3 on Aβ25-35-induced cytotoxicity. Primary cortical neurons were pretreated with the indicated concentrations of HH-F3 for 2 hours. Subsequently, cells were stimulated with 10 μM of Aβ 25-35 for 46 hours. Cell viability was measured by MTT assay.

圖15顯示在APP/PS1小鼠中HH-F3對於Aβ斑塊負荷的影響。該APP/PS1轉基因小鼠在120天大時於大腦皮層中發展出Aβ斑塊(Radde等人,Abeta42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology.EMBO Rep 2006;7:940-946)。對140天大的APP/PS1小鼠口服投予HH-F3(300mg/kg/天)一個月。該代表性影像係藉由在APP/PS1小鼠的大腦半球中的Aβ斑塊負荷之BSB染色予以評估,然後手動計數Aβ斑塊數量。 Figure 15 shows the effect of HH-F3 on A[beta] plaque load in APP/PS1 mice. The APP/PS1 transgenic mice developed Aβ plaques in the cerebral cortex at 120 days of age (Radde et al., Abeta42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology. EMBO Rep 2006; 7: 940-946) . HH-F3 (300 mg/kg/day) was orally administered to 140-day-old APP/PS1 mice for one month. This representative image was evaluated by BSB staining of A[beta] plaque load in the cerebral hemisphere of APP/PS1 mice, and then the number of A[beta] plaques was manually counted.

圖16A-16B顯示HH-F3對APP/PS1小鼠之大腦皮層中的Aβ之影響。圖16A提供以ELISA測定的大腦皮層均質物中SDS可溶的Aβ1-40和Aβ1-42濃度之定量分析結果(載體n=3,HH-F3n=6)。圖16B提供以ELISA測定的大腦皮層均質物中不可溶級分(甲酸可溶的)的Aβ1-40和Aβ1-42濃度之定量分析結果(載體n=3,HH-F3 n=6)。 Figures 16A-16B show the effect of HH-F3 on A[beta] in the cerebral cortex of APP/PS1 mice. Figure 16A provides quantitative analysis of SDS soluble A?1-40 and A? 1-42 concentrations in cerebral cortex homogenates as determined by ELISA (vector n = 3, HH-F3n = 6). Figure 16B provides quantitative analysis of the concentration of Aβ1-40 and Aβ 1-42 of the insoluble fraction (formic acid soluble) in the cerebral cortex homogenate as determined by ELISA (vector n=3, HH-F3 n=6).

圖17A-17B顯示HH-F3向上調控APP/PS1小鼠之大腦皮層中的Aβ清除相關蛋白。圖17A提供藉由免疫轉印法檢測皮層溶解產物之Aβ清除相關蛋白的結果。圖17B提供測定的WT與APP/PS1或載體與APP/PS1之間的蛋白定量分析比之結果。 Figures 17A-17B show that HH-F3 up-regulates Aβ clearance-associated proteins in the cerebral cortex of APP/PS1 mice. Figure 17A provides the results of detecting Aβ clearance-related proteins of cortical lysates by immunotransfer. Figure 17B provides the results of a quantitative analysis of the protein between the determined WT and APP/PS1 or vector and APP/PS1.

圖18顯示以不同方式萃取的GP在抑制HBV表面抗原中的效果。其提供顯示在無血清的DMEM培養基中使用以不同方式萃取的GP萃取 物(200μg/ml)處理48小時經培養的人類肝腫瘤Hep3B/T2細胞之影像;其中藉由ELISA測定HBV表面抗原並標準化為MTT檢驗;其中使用胰島素作為正對照組。 Figure 18 shows the effect of GP extracted in different ways in inhibiting HBV surface antigen. It provides GP extraction that is shown to be extracted in different ways using serum-free DMEM medium. Images of cultured human liver tumor Hep3B/T2 cells were treated for 48 hours; (HBV surface antigen was determined by ELISA and normalized to MTT assay; insulin was used as a positive control group.

除非另有定義,否則本文中使用的所有技術和科學術語具有與本發明所屬技術領域中具有通常知識之人士一般理解的相同的含義。 Unless otherwise defined, all technical and scientific terms used herein have the same meaning meaning meaning meaning

在下表中列出本文中使用的縮寫之全名: The full names of the abbreviations used in this article are listed in the following table:

本文所使用的單數形「一」及「該」包括複數的參照物,除非上下文另有明確說明。因此,舉例來說,提及「一樣品」包括複數個該樣品及其為所屬技術領域中具有通常知識者習知的等同物。 As used herein, the singular and " Thus, for example, reference to "a sample" includes a plurality of such samples and their equivalents as those of ordinary skill in the art.

本發明提供一種藉由使用DMSO萃取植物所製備的、縞辦屬或紅景天屬之DMSO萃取物的新用途,指的是在代謝性疾病治療的萃取物。 The present invention provides a novel use of a DMSO extract of the genus Rhododendron or Rhodiola prepared by extracting a plant using DMSO, and refers to an extract treated in a metabolic disease.

根據本發明,該萃取物可使用本技術領域中常用的或標準的方法藉由二甲亞碸(DMSO)萃取植物予以製備。在本發明之一實施例中,該植物的葉子係研磨並凍乾成為粉末,並與DMSO進行劇烈震盪,較佳為30%DMSO。在以DMSO萃取之前,可包括使用甲醇(MeOH)進一步萃取。 According to the present invention, the extract can be prepared by extracting plants with dimethyl hydrazine (DMSO) using methods commonly used in the art or standard methods. In one embodiment of the invention, the leaves of the plant are ground and lyophilized to a powder and violently shaken with DMSO, preferably 30% DMSO. Further extraction with methanol (MeOH) may be included prior to extraction with DMSO.

本文所使用的「縞辦屬(Graptopetalum sp.)」乙詞是指縞辦屬中的任何植物,或其中的一部分或多個部分。縞辦屬或其部分中超過一個種的組合也可以考慮。縞辦屬較佳為石蓮花(Graptopetalum paraguayense.)。 As used herein, the term " Graptopetalum sp." refers to any plant in the genus, or part or parts of it. Combinations of more than one species of the genus or part thereof may also be considered. It is better to use Graptopetalum paraguayense .

在本發明之一較佳具體實施例中,該縞辦屬為石蓮花。 In a preferred embodiment of the invention, the genus is a stone lotus.

本文所使用的「紅景天屬(Rhodiola sp.)」乙詞是指紅景天屬中的任何植物,或其中的一部分或多個部分。紅景天屬或其部分中超過一個種的組合也可以考慮。 As used herein, the term " Rhodiola sp." refers to any plant in the genus Rhodiola, or a part or parts thereof. Combinations of more than one species of Rhodiola or parts thereof are also contemplated.

在本發明之一較佳具體實施例中,該紅景天屬為紅景天(Rhodiola rosea.)。 In a preferred embodiment of the invention, the Rhodiola rosea is Rhodiola rosea .

本文所使用的「萃取物」乙詞是指藉由使用溶劑浸泡或混合待萃取物質所得到的溶液。在本發明中,該萃取物為DMSO萃取物。 As used herein, the term "extract" refers to a solution obtained by soaking or mixing a substance to be extracted with a solvent. In the present invention, the extract is a DMSO extract.

本發明亦提供富含來自植物的抗癌成分之級分,該植物係選自於由縞辦屬所組成之群組,該級分之製備是藉由使用二甲亞碸(DMSO)萃取植物,然後藉由色層層析法分離以獲得級分。在本發明之一實施例中,該植物為石蓮花。該級分是藉由使用DMSO萃取植物並藉由色層層析法分離以獲得級分而得到。在本發明之一實施例中,將Sephadex LH-20管柱用於色層層析法,稱為HH-F3。因此,該級分HH-F3是一種用於代謝性疾病預防或治療的潛在治療劑。 The present invention also provides a fraction enriched in plant-derived anti-cancer components selected from the group consisting of genus, which is prepared by extracting plants using dimethyl hydrazine (DMSO). Then, it was separated by chromatography to obtain a fraction. In one embodiment of the invention, the plant is a stone lotus. This fraction was obtained by extracting plants using DMSO and separating them by chromatography to obtain fractions. In one embodiment of the invention, a Sephadex LH-20 column is used for chromatography, referred to as HH-F3. Therefore, this fraction HH-F3 is a potential therapeutic agent for the prevention or treatment of metabolic diseases.

根據本發明之一實施例,石蓮花(GP)萃取物和HH-F3級分可藉由美國專利公開號第20120259004號中指示的方法來製備,該專利案之揭示內容係以引用方式全部併入本文中。 According to an embodiment of the present invention, the stone lotus (GP) extract and the HH-F3 fraction can be prepared by the method indicated in U.S. Patent Publication No. 20120259004, the disclosure of which is incorporated herein by reference. Into this article.

亦已知的是該HH-F3級分含有一富含3,4,5-三羥基芐基(3,4,5-trihydroxy benzylic)成分的原花青素(proanthocyanidin),該3,4,5-三羥基芐基成分具有以下式I結構, It is also known that the HH-F3 fraction contains a proanthocyanidin rich in 3,4,5-trihydroxybenzylic components, the 3,4,5-three The hydroxybenzyl component has the structure of the following formula I,

其中一個R為H或前花青素(prucyanidin,PC)單元;而另一個R為OH或前飛燕草素(prodelphindine,PD)單元;n為範圍從21至38的數字;以及PC單元:PD單元<1:20。前花青素(PC)單元的結構為 One of R is H or a procucyanidin (PC) unit; the other R is OH or a prodelphindine (PD) unit; n is a number ranging from 21 to 38; and PC unit: PD Unit <1:20. The structure of the proanthocyanidin (PC) unit is

並且前飛燕草素(PD)的結構為 And the structure of the former delphinidin (PD) is

本文所使用的「代謝性疾病」乙詞是指任何擾亂正常代謝的疾病或失調,代謝是在細胞層次上將食物轉換為能量的過程。代謝性疾病影響細胞進行關鍵生化反應的能力,其涉及蛋白質(胺基酸)、碳水化合物(糖和澱粉)、或脂質(脂肪酸)的處理或輸送。例如,代謝性疾病包括糖尿病、肥胖及脂肪肝疾病,無論是酒精性脂肪肝疾病(AFLD)或非酒精性 脂肪肝疾病(NAFLD)。 As used herein, the term "metabolic disease" refers to any disease or disorder that disrupts normal metabolism. Metabolism is the process of converting food into energy at the cellular level. Metabolic diseases affect the ability of cells to undergo key biochemical reactions involving the processing or delivery of proteins (amino acids), carbohydrates (sugars and starches), or lipids (fatty acids). For example, metabolic diseases include diabetes, obesity, and fatty liver disease, whether it is alcoholic fatty liver disease (AFLD) or nonalcoholic Fatty liver disease (NAFLD).

本文所使用的「代謝症候群」乙詞是指醫療病症的組合,當該等醫療病症同時發生時會提高罹患心血管疾病和糖尿病的風險。 As used herein, the term "metabolic syndrome" refers to a combination of medical conditions that increase the risk of cardiovascular disease and diabetes when these medical conditions occur simultaneously.

本文所使用的「AMPK途徑」乙詞是指其中AMP活化蛋白激酶(AMP-activated protein kinase,AMPK)在作為細胞能量恆定的主調控者上發揮關鍵作用的途徑。AMPK係活化以回應耗盡細胞ATP供應的壓力,例如低血糖、缺氧、局部缺血及熱休克。作為回應低ATP水平的細胞能量感應物,AMPK活化正調控補充細胞ATP供應的訊號傳導途徑,包括脂肪酸氧化和自噬。AMPK負調控消耗ATP的生物合成過程,包括糖質新生、脂質及蛋白合成。AMPK通過若干直接參與此等過程的酵素之直接磷酸化以及通過藉由磷酸化轉錄因子、共活化劑、及共抑制劑來轉錄控制代謝而完成之。由於其作為脂質和葡萄糖代謝兩者之中央調控者的角色,AMPK被認為是第II型糖尿病、肥胖及癌症治療的潛在治療標靶。AMPK亦通過其與mTOR和去乙醯化酶(sirtuins)的相互作用而被涉及在若干作為老化的關鍵調製劑的物種中。 As used herein, the term "AMPK pathway" refers to a pathway in which AMP-activated protein kinase (AMPK) plays a key role as a major regulator of cellular energy. The AMPK system is activated in response to stresses that deplete the cellular ATP supply, such as hypoglycemia, hypoxia, ischemia, and heat shock. As a cellular energy sensor that responds to low ATP levels, AMPK activation is regulating signaling pathways that complement cellular ATP supply, including fatty acid oxidation and autophagy. AMPK negatively regulates the biosynthesis processes that consume ATP, including gluconeogenesis, lipids, and protein synthesis. AMPK is accomplished by direct phosphorylation of several enzymes directly involved in these processes and by transcriptional control of metabolism by phosphorylation of transcription factors, coactivators, and co-inhibitors. Because of its role as a central regulator of both lipid and glucose metabolism, AMPK is considered a potential therapeutic target for type II diabetes, obesity, and cancer treatment. AMPK is also involved in several species that are key modulators of aging through its interaction with mTOR and sirtuins.

本文所使用的「自噬」或「自噬途徑」,亦被稱為自體吞噬作用,其是指通過溶酶體的作用涉及不必要或不正常細胞成分之細胞降解的基本分解代謝機制。該細胞成分的分解可以藉由保持細胞能量水平以確保細胞在飢餓期間存活。若經調控,自噬可確保細胞成分的合成、降解及再循環。在此過程中,標靶的細胞質組分係被從自噬小體(autophagosomes)內的其餘細胞分離,其接著與溶酶體融合並降解或再循環。 As used herein, "autophagy" or "autophagy pathway", also known as autophagy, refers to a basic catabolic mechanism involved in the degradation of cells of unwanted or abnormal cellular components by the action of lysosomes. Decomposition of this cellular component can be achieved by maintaining cellular energy levels to ensure that cells survive during starvation. If regulated, autophagy ensures the synthesis, degradation and recycling of cellular components. During this process, the cytoplasmic component of the target is isolated from the remaining cells within the autophagosomes, which are then fused to the lysosome and degraded or recycled.

本文所使用的「神經退化性疾病」乙詞是指主要影響人腦中 神經元的病症範圍。因為神經元是包括大腦和脊髓神經的神經系統之建構單元,而且神經元通常不會複製或自身替換,所以當它們損壞或死亡時,它們無法被人體替換。神經退化性疾病的一些典型實例包括帕金森氏症、阿茲海默症、及杭丁頓氏症。 The term "neurodegenerative disease" as used herein refers to the main influence on the human brain. The range of conditions of a neuron. Because neurons are the building blocks of the nervous system that include the brain and spinal nerves, and neurons are usually not copied or replaced by themselves, they cannot be replaced by the body when they are damaged or die. Some typical examples of neurodegenerative diseases include Parkinson's disease, Alzheimer's disease, and Huntington's disease.

本文所使用的「澱粉樣蛋白相關疾病」乙詞是指涉及澱粉樣蛋白的疾病類別。澱粉樣蛋白是共享特定結構特徵的不溶性纖維蛋白凝聚體,其出現在天然存在於體內的蛋白和多肽之各種不當折疊變體。該錯誤折疊的結構會改變它們的合適配置,使他們彼此或與其他的細胞成分錯誤地交互作用而生成不溶性纖絲。已知澱粉樣蛋白與嚴重的人類疾病之病理相關,於器官中澱粉樣蛋白纖絲的異常積聚可能導致澱粉樣變性症,而且可能在各種神經退化性疾病中產生作用。該澱粉樣蛋白相關疾病包括但不限於阿茲海默症、第2型糖尿病、帕金森氏症、杭丁頓氏症、致死性家族失眠症、及類風濕性關節炎。 As used herein, the term "amyloid-related disease" refers to a disease class involving amyloid. Amyloid is an insoluble fibrin agglomerate that shares specific structural features that occur in various improperly folded variants of proteins and polypeptides naturally present in the body. The misfolded structure changes their proper configuration, causing them to erroneously interact with each other or with other cellular components to produce insoluble filaments. It is known that amyloid is associated with pathology of severe human diseases, and abnormal accumulation of amyloid fibrils in organs may cause amyloidosis and may play a role in various neurodegenerative diseases. The amyloid-related diseases include, but are not limited to, Alzheimer's disease, type 2 diabetes, Parkinson's disease, Huntington's disease, lethal family insomnia, and rheumatoid arthritis.

據信,具有抑制Aβ累積(或凝聚)的能力之試劑可能適用於澱粉樣蛋白相關疾病。另外,蛋白凝聚被認為是諸如阿茲海默症、帕金森氏症、及杭丁頓氏症等神經退化性疾病的常見病狀。自噬是溶酶體降解的過程,其回收細胞廢物並消除受損的胞器和蛋白凝聚體,而且自噬途徑中的障礙對於神經退化性疾病的發病機制有所貢獻。因此,自噬誘導劑可為神經退化性疾病的治療標靶。 It is believed that agents having the ability to inhibit A[beta] accumulation (or aggregation) may be suitable for amyloid-related diseases. In addition, protein aggregation is considered to be a common condition of neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. Autophagy is a process of lysosomal degradation that recovers cellular waste and eliminates damaged organelles and protein aggregates, and obstacles in the autophagy pathway contribute to the pathogenesis of neurodegenerative diseases. Therefore, an autophagy inducing agent can be a therapeutic target for neurodegenerative diseases.

本發明證實的是GP萃取物或HH-F3級分係能夠抑制Hep3B/T2細胞中的環狀AMP/迪皮質醇誘導的醣異生酶基因表現和HBV核心啟動子的表現活性。實施例亦闡釋GP萃取物或HH-F3級分在Hep3B/T2或 1.3ES2細胞中抑制了HBV表面抗原和醣異生酶的基因表現;然而,該GP萃取物/HH-F3級分不影響糖分解,而是將癌細胞的代謝從糖分解移到葡萄糖氧化並誘導粒腺體相依的細胞凋亡。其闡釋GP萃取物/HH-F3級分經由AMPK相依的方式調控了脂質生成相關蛋白的表現和醣異生酶基因的表現;因此,該GP萃取物/HH-F3級分可以改善異常脂質累積並減少腫瘤發展的進展,具體而言是在患有第2型糖尿病而需要胰島素或二甲雙胍(Metformin)藥物以在體內抑制肝臟糖質新生的患者身上。 The present invention demonstrates that the GP extract or the HH-F3 fraction can inhibit the expression of the cyclic AMP/digocortisol-induced gluconeogenesis gene and the activity of the HBV core promoter in Hep3B/T2 cells. The examples also illustrate that the GP extract or HH-F3 fraction is in Hep3B/T2 or The gene expression of HBV surface antigen and gluconeogenesis enzyme was inhibited in 1.3ES2 cells; however, the GP extract/HH-F3 fraction did not affect the sugar decomposition, but shifted the metabolism of cancer cells from sugar decomposition to glucose oxidation. Inducing granulocyte-dependent apoptosis. It clarifies that the GP extract/HH-F3 fraction regulates the expression of lipid-related proteins and the expression of gluconeogenesis genes via AMPK-dependent manner; therefore, the GP extract/HH-F3 fraction can improve abnormal lipid accumulation. It also reduces the progression of tumor development, specifically in patients with type 2 diabetes who require insulin or metformin (Metformin) to inhibit liver gluconeogenesis in the body.

實施例7和圖8中亦證實GP/HH-F3和蕾莎瓦(Sorafenib)的組合在Huh7的細胞存活率上提供了協同效用。因此,該預防性的發明提供了用於預防或治療癌症的協同方法或組合物。 It was also demonstrated in Example 7 and Figure 8 that the combination of GP/HH-F3 and Sorafenib provided a synergistic effect on the cell viability of Huh7. Accordingly, the prophylactic invention provides a synergistic method or composition for preventing or treating cancer.

老化是會影響所有器官的普遍過程。在細胞穩態中與年齡相關的破壞導致對生理壓力和器官功能障礙的回應度降低。能量代謝的有效調控是細胞穩態的關鍵要求。AMPK是對ATP水平降低的細胞回應之主要調控者,並作為感測者以在細胞內維持能量平衡。已知AMPK在壓力期間由葡萄糖和脂肪酸刺激能量產生,並抑制蛋白、膽固醇及糖原合成的能量消耗。一般來說,AMPK的活化會向下調控合成途徑,例如蛋白、脂肪酸及膽固醇的生物合成,但會開啟產生ATP的分解代謝途徑,例如脂肪酸氧化、葡萄糖攝取及糖分解。其不僅通過多種關鍵代謝酶的直接磷酸化、而且還以組織特異的方式改變基因的表現來實現。目前,AMPK被視為重要的分子標靶,因為據信AMPK活化劑可被用於代謝和神經退化性疾病的治療,並且作為抗衰老藥物。 Aging is a universal process that affects all organs. Age-related disruption in cellular homeostasis results in reduced response to physiological stress and organ dysfunction. Effective regulation of energy metabolism is a key requirement for cell homeostasis. AMPK is a major regulator of cellular responses to reduced ATP levels and acts as a sensor to maintain energy balance within the cell. AMPK is known to produce energy from glucose and fatty acid stimulation during stress and inhibits the energy expenditure of protein, cholesterol and glycogen synthesis. In general, activation of AMPK down-regulates synthetic pathways, such as protein, fatty acid, and cholesterol biosynthesis, but opens the catabolic pathways that produce ATP, such as fatty acid oxidation, glucose uptake, and sugar breakdown. It is achieved not only by direct phosphorylation of a variety of key metabolic enzymes, but also by altering the expression of genes in a tissue-specific manner. Currently, AMPK is considered an important molecular target because AMPK activators are believed to be useful in the treatment of metabolic and neurodegenerative diseases, and as anti-aging drugs.

因此,鑑於本發明中顯示GP/HH-F3為AMPK活化劑的結 果,其建議GP/HH-F3藉由調控AMPK途徑而具有抗衰老能力。 Therefore, in view of the present invention, it is shown that GP/HH-F3 is a AMPK activator. As a result, it is suggested that GP/HH-F3 has anti-aging ability by regulating the AMPK pathway.

本發明亦提供一種使用本發明之萃取物、級分或式I化合物的用途、方法或組合物。 The invention also provides a use, method or composition of the use of the extract, fraction or compound of formula I of the invention.

本文所使用的「治療有效量」乙詞是指足以實現所欲之治療目的的藥劑量。該給予藥劑的治療有效量將會隨著不同因素而有所不同,例如藥劑的本質、投予之方式、接收藥劑的動物之大小和種類、以及投予之目的。每一個體案例之治療有效量可由技藝人士根據本文之揭示及現有技藝所建立之方法以經驗決定。 As used herein, the term "therapeutically effective amount" refers to an amount of a medicament sufficient to achieve the desired therapeutic purpose. The therapeutically effective amount of the administered agent will vary with different factors, such as the nature of the agent, the mode of administration, the size and type of the animal receiving the agent, and the purpose for which it is administered. The therapeutically effective amount of each individual case can be determined empirically by the skilled artisan in light of the teachings herein and the methods established in the prior art.

本發明之組合物可藉由任何合適的途徑投予,包括但不限於非經口服的或經口服的投予方式。該用於非經口服投予的組合物包括溶液、懸浮液、乳化液、及固態之可注射組合物,其在使用前可用溶劑立刻溶解或懸浮。該注射可藉由將一種或多種之活性成分溶解、懸浮、或乳化於稀釋劑中以製得。所述稀釋劑之實施例為用於注射之蒸餾水、生理食鹽水、植物油、酒精及其組合。再者,該注射可能包含穩定劑、助溶劑、懸浮劑、乳化劑、緩和劑、緩衝液、保存劑等。該注射於最後之製備步驟係經滅菌的、或以無菌程序製得。本發明之組合物亦可製備成無菌之固態製劑,例如藉由冷凍乾燥,且可在滅菌後使用、或於使用前立即溶於無菌之可注射水或其他無菌之稀釋劑中使用。 The compositions of the present invention can be administered by any suitable route including, but not limited to, parenteral or oral administration. The compositions for parenteral administration include solutions, suspensions, emulsions, and solid injectable compositions which can be dissolved or suspended in a solvent immediately before use. The injection can be made by dissolving, suspending, or emulsifying one or more active ingredients in a diluent. Examples of the diluent are distilled water for injection, physiological saline, vegetable oil, alcohol, and combinations thereof. Furthermore, the injection may contain stabilizers, solubilizers, suspending agents, emulsifiers, demulcents, buffers, preservatives and the like. The final preparation step of the injection is sterilized or prepared in a sterile procedure. The compositions of the present invention may also be prepared as sterile solid preparations, for example by lyophilization, and may be employed after sterilization or dissolved in sterile injectable water or other sterile diluent immediately prior to use.

根據本發明,該組合物亦可透過口服路徑投予,其中該組合物可處於固體或液體形式。該固體組合物包括片劑、丸劑、囊劑、分散的粉劑、顆粒及其類似物。該口服組合物亦包括可被黏貼於口腔的漱口藥及舌下片劑。該囊劑包括硬膠囊及軟膠囊。於該等口服用固體組合物中,一 或多種之活性化合物可被單獨地混合或與稀釋劑、黏結劑、粉碎劑、潤滑劑、穩定劑、助溶劑混合,並於之後被以傳統方式配製成製劑。必要時,該等製劑可以包覆劑進行包覆,或可以兩種或多種包覆層包覆。另一方面,該口服用液體組合物包括醫藥上可接受的水溶液、懸浮液、乳化液、糖漿、酏劑及其類似物。於該等組合物中,一或多種活性化合物可被溶解、懸浮或乳化於常用之稀釋劑中(如純化水、乙醇或其混合物等)。除了該等稀釋劑外,該組合物亦可含有濕潤劑、懸浮劑、乳化劑、甜味劑、調味劑、芳香劑、保存劑和緩衝液及其類似物。 According to the invention, the composition can also be administered by oral route, wherein the composition can be in solid or liquid form. The solid compositions include tablets, pills, sachets, dispersed powders, granules, and the like. The oral compositions also include mouthwashes and sublingual tablets that can be applied to the oral cavity. The capsules include hard capsules and soft capsules. In such oral solid compositions, one The active compound or compounds may be mixed separately or mixed with a diluent, a binder, a pulverizer, a lubricant, a stabilizer, a co-solvent, and then formulated in a conventional manner. If necessary, the preparations may be coated with a coating agent or may be coated with two or more coating layers. In another aspect, the oral liquid composition comprises a pharmaceutically acceptable aqueous solution, suspension, emulsion, syrup, elixir, and the like. In such compositions, one or more of the active compounds can be dissolved, suspended or emulsified in conventional diluents (such as purified water, ethanol or mixtures thereof, etc.). In addition to such diluents, the compositions may contain wetting agents, suspending agents, emulsifying agents, sweetening agents, flavoring agents, perfuming agents, preservatives and buffers, and the like.

另外,本發明提供了本發明之GP萃取物、HH-F3級分或式I化合物在製造用於治療代謝性疾病的藥物之用途,具體而言是糖尿病、肥胖或脂肪肝疾病。另一方面,本發明提供一種用於預防或治療代謝性疾病的方法,具體而言是糖尿病、肥胖或脂肪肝疾病,包含對需要該預防或治療之個體投予治療有效量的本發明萃取物、級分或化合物,具體而言是糖尿病、肥胖或脂肪肝疾病。 Further, the present invention provides the use of the GP extract of the present invention, the HH-F3 fraction or the compound of the formula I in the manufacture of a medicament for the treatment of a metabolic disease, in particular diabetes, obesity or fatty liver disease. In another aspect, the present invention provides a method for preventing or treating a metabolic disease, in particular a diabetes, obesity or fatty liver disease, comprising administering to a subject in need of such prevention or treatment a therapeutically effective amount of an extract of the present invention , fractions or compounds, in particular diabetes, obesity or fatty liver disease.

提供以下的實施例來說明本發明,而且以下的實施例不會被以任何方式解讀為限制本發明之範圍。 The following examples are provided to illustrate the invention, and the following examples are not to be construed as limiting the scope of the invention.

實施例 Example

實施例1 GP萃取物和級分之製備 Example 1 Preparation of GP extracts and fractions

將石蓮花(GP)的葉子磨碎且於-20℃冷凍乾燥成粉末,並於萃取前儲存於25℃之防潮箱中。首先,將1.5g的GP粉末與10ml 100%的甲醇(MeOH)劇烈震盪5分鐘,接著以1,500g離心5分鐘。將該上清液移除後,將10ml的30% DMSO加入各個離心沈澱物以使再懸浮各個萃取物。該 懸浮物係藉由劇烈震盪5分鐘予以混合,並以1,500g離心5分鐘兩次,再以9,300g離心5分鐘,並在室溫下使用0.45μm濾紙過濾。將30% DMSO上清液存放於-20℃作為150mg/ml的原液(稱為GP萃取物)或藉由一Sephadex LH-20管柱分離成四個級分(F1-F4)。透過西方墨漬法,使用AURKA、AURKB、及FLJ10540蛋白程度的分析,活性分子係在級分3中獲得,並稱為HH-F3級分(數據未顯示)。該HH-F3級分進一步以具有UV檢測器(Shimadzu SPD-M10A)與正相HPLC管柱(Phenomenex Luna 5u Silica 100A,4.6×250mm)的HPLC及1H-和13C-NMR光譜分析以辨識該活性分子之結構(數據未顯示)。該GP萃取物及/或HH-F3亦以透析膜(MWCO 12-14,000)(Spectrum Laboratories,Rancho Dominguez,CA)進行對水透析,以取得活性化合物。 The leaves of Stone Lotus (GP) were ground and lyophilized to a powder at -20 ° C and stored in a moisture barrier at 25 ° C prior to extraction. First, 1.5 g of GP powder was shaken vigorously with 10 ml of 100% methanol (MeOH) for 5 minutes, followed by centrifugation at 1,500 g for 5 minutes. After the supernatant was removed, 10 ml of 30% DMSO was added to each of the centrifuged precipitates to resuspend the respective extracts. The suspension was mixed by vigorous shaking for 5 minutes, centrifuged twice at 1,500 g for 5 minutes, centrifuged at 9,300 g for 5 minutes, and filtered at room temperature using a 0.45 μm filter paper. The 30% DMSO supernatant was stored at -20 ° C as a 150 mg/ml stock solution (referred to as GP extract) or separated into four fractions (F1-F4) by a Sephadex LH-20 column. The Western blotting method was used to analyze the degree of AURKA, AURKB, and FLJ10540 protein, and the active molecule was obtained in Fraction 3 and was referred to as HH-F3 fraction (data not shown). The HH-F3 fraction was further characterized by HPLC and 1 H- and 13 C-NMR spectral analysis with a UV detector (Shimadzu SPD-M10A) and a normal phase HPLC column (Phenomenex Luna 5u Silica 100A, 4.6 x 250 mm). The structure of the active molecule (data not shown). The GP extract and/or HH-F3 was also dialyzed against water using a dialysis membrane (MWCO 12-14,000) (Spectrum Laboratories, Rancho Dominguez, CA) to obtain the active compound.

實施例2 HH-F3在Hep3B/T2細胞中8-Br-cAMP/迪皮質醇誘導的醣異生酶基因表現上的影響 Example 2 Effect of HH-F3 on the expression of 8-Br-cAMP/dicortisol-induced gluconeogenesis gene in Hep3B/T2 cells

培養人類肝腫瘤Hep3B/T2細胞並使用0.5mM的8-Br-8-Br-cAMP(8-Br-cAMP)和0.5μM的迪皮質醇(Dex)任一者或兩者處理30分鐘。然後,將不同濃度(5μg/ml、10μg/ml、及20μg/ml)的HH-F3級分加入無血清的DMEM中24小時。藉由即時定量PCR量測醣異生基因、葡萄糖-6-磷酸酶(G6Pase)及磷酸烯醇丙酮酸羧激脢(PEPCK)的mRNA,並標準化為β-肌動蛋白。使用胰島素作為正對照。 Human liver tumor Hep3B/T2 cells were cultured and treated with either 0.5 mM 8-Br-8-Br-cAMP (8-Br-cAMP) and 0.5 μM dicortisol (Dex) or both for 30 minutes. Then, different concentrations (5 μg/ml, 10 μg/ml, and 20 μg/ml) of the HH-F3 fraction were added to serum-free DMEM for 24 hours. The mRNA of the gluconeogenesis gene, glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxypyrene (PEPCK) was measured by real-time quantitative PCR and normalized to β-actin. Insulin was used as a positive control.

環狀AMP及迪皮質醇(8-Br-cAMP/Dex)係使用於此測試以協同活化關鍵醣異生基因、葡萄糖-6-磷酸酶(G6Pase)及磷酸烯醇丙酮酸羧激脢(PEPCK)在人類肝腫瘤Hep3B/T2細胞中的基因表現,該結果顯示 於圖1A。該HH-F3級分(HH-F3)的處理結果顯示於圖1B,其指出HH-F3級分的處理可以在Hep3B/T2細胞中抑制8-Br-cAMP/DEX誘導的G6PasePEPCK基因表現。 Cyclic AMP and decortisol (8-Br-cAMP/Dex) were used in this assay to synergistically activate key gluconeogenesis genes, glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxypyrrol (PEPCK). ) gene expression in human liver tumors Hep3B / T2 cells, which results are shown in Figure 1A. The results of the HH-F3 fraction (HH-F3) treatment are shown in Figure 1B, which indicates that treatment of the HH-F3 fraction can inhibit 8-Br-cAMP/DEX-induced G6Pase and PEPCK gene expression in Hep3B/T2 cells. .

實施例3 HH-F3級分在8-Br-cAMP/Dex誘導的醣異生共活化劑PGC-1α表現活性上的影響。 Example 3 Effect of HH-F3 fraction on 8-Br-cAMP/Dex-induced gluconeogenesis co-activator PGC-1α expression activity.

培養人類肝腫瘤Hep3B/T2細胞並使用0.5mM的8-Br-8-Br-cAMP(8-Br-cAMP)和0.5μM的迪皮質醇(Dex)任一者或兩者處理30分鐘,以及使用10nM的胰島素或20μg/ml的HH-F3級分(HH-F3)在無血清的DMEM培養基中共處理24小時。藉由即時定量PCR量測PGC-1αmRNAs,並標準化為β-肌動蛋白。使用胰島素作為正對照組。藉由西方墨漬法分析測定PGC-1α的核萃取物和HNF-4α蛋白水平。使用B23作為負荷控制。Dex:迪皮質醇。 Human liver tumor Hep3B/T2 cells were cultured and treated with either 0.5 mM 8-Br-8-Br-cAMP (8-Br-cAMP) and 0.5 μM dicortisol (Dex) for 30 minutes, and Co-treatment with 10 nM insulin or 20 μg/ml HH-F3 fraction (HH-F3) in serum-free DMEM medium for 24 hours. PGC-1α mRNAs were measured by real-time quantitative PCR and normalized to β-actin. Insulin was used as a positive control group. The nuclear extract and HNF-4α protein levels of PGC-1α were determined by Western blot analysis. Use B23 as load control. Dex: Decortisol.

Mahlavu細胞係以不同濃度(0μg/ml、5μg/ml、25μg/ml、及50μg/ml)的HH-F3處理,然後使用抗PGC1-α、SIRT1、PPARγ、及FoxO1的抗體進行西方墨漬法分析。此等是來自3個獨立實驗的代表性數據。 The Mahlavu cell line was treated with different concentrations (0 μg/ml, 5 μg/ml, 25 μg/ml, and 50 μg/ml) of HH-F3, and then Western blotting was performed using antibodies against PGC1-α, SIRT1, PPARγ, and FoxO1. analysis. These are representative data from 3 independent experiments.

已知共活化因子PGC1-α的表現驅動關鍵醣異生酶例如PEPCK和G6Pase的轉錄,並與轉錄因子HNF4-α和FoxO1相關,而且G6PasePEPCK係藉由PGC1-α/FoxO1/Sirt1的途徑進行調控。該Hep3B/T2細胞係以8-Br-cAMP/Dex處理以誘導醣異生基因的表現,然後以HH-F3處理,而且8-Br-cAMP和迪皮質醇協同活化基因和醣異生共活化劑PGC1-α基因表現的蛋白表現。該結果顯示於圖2A和圖2B,其指出HH-F3影響PGC1-α/FoxO1/Sirt1的途徑;其中該HH-F3的處理在Hep3B/T2細胞中抑制了 8-Br-cAMP/Dex誘導的PGC1-α表現水平(見圖2A);然而,在人類肝腫瘤Hep3B/T2細胞中對HNF4-α蛋白的表現沒有影響(參見圖2B)。事實上,已知的是,關鍵醣異生基因的主要代謝調控劑和共活化劑PGC1-α已被證明會強烈地共活化FoxO1和HNF4-α用於HBV轉錄。圖2C亦指出HH-F3的處理在Hep3B/T2細胞中抑制了PGC1-α、HNF4-α、及FoxO1的表現。 It is known that the expression of the co-activator PGC1-α drives transcription of key gluconeogenesis enzymes such as PEPCK and G6Pase, and is associated with the transcription factors HNF4-α and FoxO1, and that G6Pase and PEPCK are pathways by PGC1-α/FoxO1/Sirt1. Carry out regulation. The Hep3B/T2 cell line was treated with 8-Br-cAMP/Dex to induce the expression of gluconeogenesis genes, then treated with HH-F3, and 8-Br-cAMP and dicortisol synergistically activate genes and gluconeogenesis Protein expression of the agent PGC1-α gene. The results are shown in Figures 2A and 2B, which indicate that HH-F3 affects the pathway of PGC1-α/FoxO1/Sirt1; wherein treatment of HH-F3 inhibits 8-Br-cAMP/Dex induction in Hep3B/T2 cells PGC1-α expression levels (see Figure 2A); however, there was no effect on the expression of HNF4-α protein in human liver tumor Hep3B/T2 cells (see Figure 2B). In fact, it is known that the major metabolic regulator of the key gluconeogenesis gene and the coactivator PGC1-α have been shown to strongly co-activate FoxO1 and HNF4-α for HBV transcription. Figure 2C also indicates that treatment with HH-F3 inhibits the expression of PGC1-α, HNF4-α, and FoxO1 in Hep3B/T2 cells.

實施例4 透過AMPK的活化來抑制醣異生酶基因表現之HH-F3作用。 Example 4 The HH-F3 action of the gluconeogenesis gene expression was inhibited by activation of AMPK.

培養人類肝腫瘤Hep3B/T2細胞並使用0.5mM的8-Br-cAMP和0.5μM的迪皮質醇(Dex)預處理30分鐘。然後,將不同濃度(0μg/ml、5μg/ml、10μg/ml、及20μg/ml)的HH-F3加入無血清的DMEM中24小時。對於啟動子活性的檢驗,使用由葡萄糖-6-磷酸酶啟動子驅動的螢光素酶報告質體轉染Hep3B/T2細胞。與對照相比將螢光素酶的活性從共轉染的pCMV-β-半乳糖苷酶質體標準化為β-半乳糖苷酶的活性。在HH-F3或HH-F3與AMPK抑製劑化合物C存在下檢驗環狀AMP/DEX刺激的G6Pase啟動子活性。處理一天後,製備細胞溶解產物以進行螢光素酶活性分析。使用胰島素作為正對照組。使用二甲雙胍作為AMPK正對照。 Human liver tumor Hep3B/T2 cells were cultured and pretreated with 0.5 mM 8-Br-cAMP and 0.5 μM dicortisol (Dex) for 30 minutes. Then, different concentrations (0 μg/ml, 5 μg/ml, 10 μg/ml, and 20 μg/ml) of HH-F3 were added to serum-free DMEM for 24 hours. For the assay of promoter activity, Hep3B/T2 cells were transfected with luciferase reporter plastids driven by the glucose-6-phosphatase promoter. Luciferase activity was normalized from co-transfected pCMV-[beta]-galactosidase plastids to [beta]-galactosidase activity compared to control. Cyclic AMP/DEX stimulated G6Pase promoter activity was tested in the presence of HH-F3 or HH-F3 and AMPK inhibitor Compound C. After one day of treatment, cell lysates were prepared for luciferase activity assay. Insulin was used as a positive control group. Metformin was used as a positive control for AMPK.

該Hep3B/T2細胞係以8-Br-cAMP/Dex處理以誘導G6Pase啟動子的活性,然後使用HH-F3處理。該結果顯示於圖3A和圖3B,其顯示HH-F3可以以劑量依賴的方式降低G6Pase啟動子的活性。先前報告的是,AMP活化蛋白激酶(AMPK)係一在能量耗盡情況下抑制ATP消耗並刺激ATP產生的細胞能量感測劑。已知AMPK的活化可抑制G6Pase和PEPCK的基因表現,並抑制肝葡萄糖產生。因此,測試HH-F3經由AMPK途徑調控G6Pase 的效果。該結果顯示AMPK抑製劑化合物C可部分逆轉HH-F3抑制的G6Pase啟動子活性(見圖3C),其表示HH-F3可透過AMPK的活化來作用以抑制醣異生酶的基因表現。 The Hep3B/T2 cell line was treated with 8-Br-cAMP/Dex to induce activity of the G6Pase promoter and then treated with HH-F3. The results are shown in Figures 3A and 3B, which show that HH-F3 can reduce the activity of the G6 Pase promoter in a dose-dependent manner. It was previously reported that AMP-activated protein kinase (AMPK) is a cellular energy sensor that inhibits ATP depletion and stimulates ATP production in the event of energy depletion. Activation of AMPK is known to inhibit gene expression of G6Pase and PEPCK and inhibit hepatic glucose production. Therefore, testing HH-F3 regulates G6Pase via the AMPK pathway Effect. This result shows that the AMPK inhibitor compound C partially reverses the H6-F3 inhibited G6Pase promoter activity (see Fig. 3C), which indicates that HH-F3 can act through the activation of AMPK to inhibit the gene expression of gluconeogenesis.

實施例5 HH-F3在Hep3B/T2細胞中對8-Br-cAMP/Dex誘導的HBV核心啟動子活性之影響。 Example 5 Effect of HH-F3 on 8-Br-cAMP/Dex-induced HBV core promoter activity in Hep3B/T2 cells.

藉由HBV核心啟動子(CP)和HBV X啟動子(XP)驅動的螢光素酶報告質體轉染Hep3B/T2細胞以進行啟動子活性檢驗。然後,在無血清的DMEM中、在不存在或存在不同濃度(0μg/ml、5μg/ml、10μg/ml、及20μg/ml)的HH-F3下使用8-Br-cAMP/Dex處理細胞1天,製備細胞溶解產物來進行螢光素酶活性分析。將螢光素酶的活性從共轉染的pCMV-β-半乳糖苷酶質體標準化為β-半乳糖苷酶活性。使用胰島素作為正對照。 The luciferase reporter plastids driven by the HBV core promoter (CP) and the HBV X promoter (XP) were transfected into Hep3B/T2 cells for promoter activity assay. Then, cells were treated with 8-Br-cAMP/Dex in serum-free DMEM in the absence or presence of different concentrations (0 μg/ml, 5 μg/ml, 10 μg/ml, and 20 μg/ml) of HH-F3. Days, cell lysates were prepared for luciferase activity assays. Luciferase activity was normalized from co-transfected pCMV-[beta]-galactosidase plastids to [beta]-galactosidase activity. Insulin was used as a positive control.

Hep3B/T2係一表現HBV的肝細胞株。HBV基因組包括聚合酶、表面、核心、及HBx。該X基因編碼X蛋白(HBx),其具有反活化性質,而且在肝的致癌作用中可能是重要的。該核心基因編碼核心的核鞘蛋白(在病毒包裝中是重要的)。先前的體外研究中建議核心啟動子的突變增加了HBV的複製。如圖4A的結果所示,環狀AMP和迪皮質醇協同地活化了B肝病毒核心啟動子的活性,但是對X啟動子沒有影響。如圖4B所示,HH-F3的劑量依賴性抑制8-Br-cAMP/Dex誘導的HBV核心啟動子的活性。該測試結果可支持HBV感染與糖尿病和HCC相關,並且為B型肝炎病毒(HBV)感染與糖尿病和HCC的關聯提供了可能的解釋。 Hep3B/T2 is a hepatocyte cell line that expresses HBV. The HBV genome includes polymerase, surface, core, and HBx. This X gene encodes the X protein (HBx), which has anti-activating properties and may be important in the carcinogenesis of the liver. This core gene encodes the core nuclear sheath protein (important in viral packaging). Previous in vitro studies have suggested that mutations in the core promoter increase HBV replication. As shown in the results of Figure 4A, cyclic AMP and dicortisol synergistically activated the activity of the B liver viral core promoter, but had no effect on the X promoter. As shown in Figure 4B, the dose-dependent inhibition of 8-H-FAMP/Dex-induced HBV core promoter activity of HH-F3. This test results support that HBV infection is associated with diabetes and HCC, and provides a possible explanation for the association of hepatitis B virus (HBV) infection with diabetes and HCC.

實施例6 HH-F3在Hep3B/T2或1.3ES2細胞中對HBV表面抗原、基因表現及醣異生酶表現的影響。 Example 6 Effect of HH-F3 on HBV surface antigen, gene expression and gluconeogenesis enzyme expression in Hep3B/T2 or 1.3 ES2 cells.

培養人類肝腫瘤Hep3B/T2細胞並使用不同濃度(0μg/ml、5μg/ml、10μg/ml、及20μg/ml)的HH-F3在無血清的DMEM培養基中處理48小時。藉由ELISA測定HBV表面抗原,並利用MTT檢驗進行標準化。胰島素係使用作為正對照。使用不同濃度的HH-F3在無血清的DMEM培養基中處理1.3ES2細胞48小時。藉由即時定量PCR量測醣異生基因、G6PasePEPCK、及PGC-1α mRNAs和HBV mRNA的表現,並標準化為β-肌動蛋白。HE-145係使用作為正對照(SF(Serum free):無血清)。 Human liver tumor Hep3B/T2 cells were cultured and treated with different concentrations (0 μg/ml, 5 μg/ml, 10 μg/ml, and 20 μg/ml) of HH-F3 in serum-free DMEM medium for 48 hours. HBV surface antigen was determined by ELISA and normalized using the MTT assay. Insulin was used as a positive control. 1.3 ES2 cells were treated in serum-free DMEM medium for 48 hours using different concentrations of HH-F3. The expression of gluconeogenesis genes, G6Pase , PEPCK, and PGC-1α mRNAs and HBV mRNA was measured by real-time quantitative PCR and normalized to β-actin. HE-145 was used as a positive control (SF (Serum free): serum free).

如圖5A所示,HH-F3劑量依賴性在Hep3B/T2細胞中抑制了HBV表面抗原的表現。 As shown in Figure 5A, HH-F3 dose-dependent inhibition of HBV surface antigen expression in Hep3B/T2 cells.

此外,衍生自HepG2細胞並含有一個整合複製的HBV基因組的1.3ES2細胞株係使用來商討HH-F3是否可以經由調控PGC1-α來抑制HBV基因的表現。如圖5B-5E所示,HH-F3抑制關鍵的醣異生基因、G6PasePEPCK的基因表現、以及過氧化體增殖物活化受體-γ共活化劑1α(PGC-1α)基因、在1.3ES2細胞中的HBV基因表現。 In addition, a 1.3 ES2 cell line derived from HepG2 cells and containing an integrated replication HBV genome was used to discuss whether HH-F3 can inhibit the expression of the HBV gene via regulation of PGC1-α. As shown in Figures 5B-5E, HH-F3 inhibits key gluconeogenesis genes, G6Pase and PEPCK gene expression, and peroxisome proliferator-activated receptor-gamma coactivator 1α (PGC-1α) gene, at 1.3 HBV gene expression in ES2 cells.

實施例7 GP和HH-F3對HCC中脂肪酸合成之活性的影響 Example 7 Effect of GP and HH-F3 on the Activity of Fatty Acid Synthesis in HCC

Huh7和Mahlavu細胞係以GP和HH-F3進行處理,然後使用抗磷酸化-AMPK、磷酸化-AKT、磷酸化-ACC(Ser79)、AMPK、SREBP2、AKT、ACC、及FASN的抗體進行西方墨漬法分析(n=3)。使用BNL細胞來產生皮下腫瘤。將小鼠分成兩組。一組作為未處理的對照組(3隻不同的小鼠),而其他四隻小鼠每天經由口服(P.O.)途徑以HH-F3(20mg/天)處理3週。3週後,犧牲小鼠並均勻化腫瘤,隨後進行西方墨漬法及定量。 The Huh7 and Mahlavu cell lines were treated with GP and HH-F3, and then Western anti-phospho-AMPK, phosphorylated-AKT, phosphorylated-ACC (Ser 79 ), AMPK, SREBP2, AKT, ACC, and FASN antibodies were used. Ink stain analysis (n=3). BNL cells are used to produce subcutaneous tumors. The mice were divided into two groups. One group served as an untreated control group (3 different mice), while the other four mice were treated with HH-F3 (20 mg/day) for 3 weeks per day via the oral (PO) route. After 3 weeks, the mice were sacrificed and the tumors were homogenized, followed by Western blotting and quantification.

由最近發現可知與異常脂質生成相關的是癌症,其顯示主要 脂質生成酶的抑制可以減少腫瘤形成。脂質係與許多病理過程有關聯,包含肝脂肪變性(脂肪肝),其表現是脂質在肝細胞中過量累積。異常的脂質累積係與能量代謝中的多基因遺傳缺陷和肝臟惡性腫瘤有關。肝癌細胞需要更多的能量透過失調的脂質重新生成來存活,其可能有助於肝腫瘤形成和人類HCC預後。在HCC中,該異常脂質生成的程度與臨床侵略性、AKT-mTOR訊號傳遞途徑的活化、以及AMPK的抑制相關聯。肝AMPK的活化,一種代謝物感測激酶,在代謝壓力的狀況下扮演保護的角色,刺激脂肪酸氧化以及分別經由增加的磷酸化與乙醯CoA羧化酶(acetyl CoA carboxylase,ACC)以及HMG-CoA還原酶(HMG-CoA reductase,HMGCR)、用於重新合成脂肪酸和膽固醇的限速酶之去活化而敏銳地關閉脂肪酸的合成。此外,該脂肪酸合成酶(FASN)的蛋白表現已知被AMPK負調控。事實上,由於其組織分佈及不尋常的酶活性,標靶FASN,其為脂質生成的關鍵酶並在HCC中有過度表現,提供了治療性應用的機會。 It has been recently discovered that cancer associated with abnormal lipid production is known, which shows mainly Inhibition of lipogenic enzymes can reduce tumor formation. Lipid lines are associated with a number of pathological processes, including hepatic steatosis (fatty liver), which is manifested by excessive accumulation of lipids in hepatocytes. Abnormal lipid accumulation is associated with polygenic genetic defects and hepatic malignancies in energy metabolism. Liver cancer cells require more energy to survive through the regeneration of dysregulated lipids, which may contribute to liver tumor formation and human HCC prognosis. In HCC, the extent of this abnormal lipid production is associated with clinical aggressiveness, activation of the AKT-mTOR signaling pathway, and inhibition of AMPK. Activation of hepatic AMPK, a metabolite-sensing kinase that plays a protective role in metabolic stress, stimulates fatty acid oxidation and increases phosphorylation with acetyl CoA carboxylase (ACC) and HMG-, respectively. The deactivation of CoA reductase (HMG-CoA reductase, HMGCR), a rate-limiting enzyme for the re-synthesis of fatty acids and cholesterol, sharply shuts down the synthesis of fatty acids. Furthermore, the protein expression of this fatty acid synthase (FASN) is known to be negatively regulated by AMPK. In fact, due to its tissue distribution and unusual enzymatic activity, the target FASN, which is a key enzyme in lipid production and overexpressed in HCC, offers an opportunity for therapeutic applications.

在此測試中,其發現HH-F3可以透過AMPK途徑和PGC1-α抑制8-Br-cAMP/Dex誘導的醣異生酶基因和HBV核心促進劑的活性。此外,已知的是AMPK途徑和PGC1-α也調控脂質生成。因此,進行測試來檢查GP/HH-F3在脂肪生成是否有作用。如圖6所示,在HCC細胞株和在BALB/c小鼠中使用GP或HH-F3處理藉由活化AMPK(增加AMPK磷酸化)並抑制活化的AKT(減少AKT磷酸化)而降低了脂肪酸(fatty acid,FA)合成的活性,BALB/c小鼠被皮下注射在具有BNL細胞的側腹上。此外,GP/HH-F3的處理導致磷酸化增加及AMPK下游標靶ACC因此失活、以及FASN的蛋白表現水平以濃度依賴方式降低。此等結果指出GP/HH-F3處理可以在HCC中 降低脂肪酸合成的活性。 In this test, it was found that HH-F3 can inhibit the activity of 8-Br-cAMP/Dex-induced gluconeogenesis gene and HBV core promoter through the AMPK pathway and PGC1-α. Furthermore, it is known that the AMPK pathway and PGC1-alpha also regulate lipid production. Therefore, a test was performed to check whether GP/HH-F3 has a role in lipogenesis. As shown in Figure 6, treatment with GP or HH-F3 in HCC cell lines and in BALB/c mice reduced fatty acids by activating AMPK (increasing AMPK phosphorylation) and inhibiting activated AKT (reducing AKT phosphorylation) (fatty acid, FA) Synthetic activity, BALB/c mice were injected subcutaneously on the flank of BNL cells. Furthermore, treatment with GP/HH-F3 resulted in increased phosphorylation and inactivation of the AMPK downstream target ACC, as well as a decrease in FASN protein expression levels in a concentration-dependent manner. These results indicate that GP/HH-F3 processing can be in HCC Reduce the activity of fatty acid synthesis.

實施例8 GP/HH-F3在Huh7中降低油酸(OA)和棕櫚酸(PA)誘導的脂質累積之影響。 Example 8 GP/HH-F3 reduced the effects of oleic acid (OA) and palmitic acid (PA) induced lipid accumulation in Huh7.

Huh7細胞係與油酸(1mM)和棕櫚酸(2mM)分別培養24小時和48小時,其造成一藉由油紅O染色可見的明顯細胞內脂質累積及由MTT檢驗的細胞存活率。圖7A顯示原始放大倍率(×400),其顯示對照組(無處理)、使用OA和PA(OA+PA)處理的組、使用OA和PA與GP處理48小時的組、以及使用OA和PA與HH-F3處理48小時的組之結果。在圖7中發現藉由油紅O將細胞內脂滴染色所定量,分別使用GP和HH-F3處理的Huh7細胞可以減少脂肪負荷(OA+PA/GP:油酸和棕櫚酸與GP共處理;OA/HH-F3:油酸和棕櫚酸與HH-F3共處理)。 The Huh7 cell line was incubated with oleic acid (1 mM) and palmitic acid (2 mM) for 24 hours and 48 hours, respectively, which resulted in a significant intracellular lipid accumulation visible by Oil Red O staining and cell viability as determined by MTT assay. Figure 7A shows the original magnification (x400) showing the control (no treatment), the group treated with OA and PA (OA + PA), the group treated with OA and PA and GP for 48 hours, and the use of OA and PA Results of the group treated with HH-F3 for 48 hours. In Figure 7, it was found that Huh7 cells treated with GP and HH-F3, respectively, were able to reduce fat load by oil red O staining of intracellular lipid droplets (OA+PA/GP: oleic acid and palmitic acid co-treated with GP ; OA/HH-F3: oleic acid and palmitic acid co-processed with HH-F3).

其可得出的結論是GP/HH-F3可活化AMPK並抑制活化的AKT,從而減少脂質合成(見圖7),雖然仍不清楚GP/HH-F3是否直接或間接調控AMPK和AKT。此外,其已知的是肝AMPK的活化和AKT的抑制可以經由增加的磷酸化和ACC的失活、以及減少的SREBP-1c和SREBP-2轉錄活化而敏銳地關閉脂肪酸的合成。另外,SREBP-1c在肝臟中調控脂質生成相關的基因,包括ACCFAS,而SREBP-2可活化膽固醇生物合成的兩種關鍵酶,HmgcrHmgcs1,其在腫瘤形成中具有重要的作用。 It can be concluded that GP/HH-F3 activates AMPK and inhibits activated AKT, thereby reducing lipid synthesis (see Figure 7), although it is still unclear whether GP/HH-F3 directly or indirectly regulates AMPK and AKT. Furthermore, it is known that activation of hepatic AMPK and inhibition of AKT can sharply shut down the synthesis of fatty acids via increased phosphorylation and inactivation of ACC, as well as reduced transcriptional activation of SREBP-1c and SREBP-2. In addition, SREBP-1c regulates lipid-related genes in the liver, including ACC and FAS , while SREBP-2 activates two key enzymes for cholesterol biosynthesis, Hmgcr and Hmgcs1 , which play important roles in tumor formation.

實施例9 GP/HH-F3和蕾莎瓦(Sorafenib)的組合對Huh7之細胞存活率的協同作用。 Example 9 Synergistic effect of the combination of GP/HH-F3 and Sorafenib on cell viability of Huh7.

分別在蕾莎瓦(5μM和10μM)存在或不存在下,分別使用各種濃度的GP(0μg/ml、50μg/ml、150μg/ml、250μg/ml、及500μg/ml) 和HH-F3(0μg/ml、5μg/ml、15μg/ml、25μg/ml、及50μg/ml)處理Huh7細胞。藉由該SRB檢驗測定細胞存活率。數據係表示為平均值±三個獨立實驗的標準差(SD)。此外,等效線圖分析證明在Huh7細胞中72小時,蕾莎瓦和各種濃度的GP/HH-F3之間有協同相互作用。 Various concentrations of GP (0 μg/ml, 50 μg/ml, 150 μg/ml, 250 μg/ml, and 500 μg/ml) were used in the presence or absence of Lysa (5 μM and 10 μM), respectively. Huh7 cells were treated with HH-F3 (0 μg/ml, 5 μg/ml, 15 μg/ml, 25 μg/ml, and 50 μg/ml). Cell viability was determined by this SRB test. Data are expressed as mean ± standard deviation (SD) of three independent experiments. In addition, isobologram analysis demonstrated a synergistic interaction between Lysawa and various concentrations of GP/HH-F3 at 72 hours in Huh7 cells.

在Huh7中分別組合各種濃度的GP和HH-F3與蕾莎瓦(唯一被FDA核准用於預先HCC患者的藥物),隨後在72小時進行細胞增殖檢驗的結果顯示於圖8A和圖8B。該組合指數(combination index,CI)分析亦顯示於圖8C和圖8D,其使用Chou-Talalay法來評估此等潛在療法之間的相互作用為協同的(CI<1)、加成的(CI=1)或是拮抗的(CI>1)。如結果所示,GP和HH-H3皆可與蕾莎瓦協同抑制Huh7的增殖。 Various concentrations of GP and HH-F3 and Lysawa (the only drugs approved by the FDA for pre-HCC patients) were combined in Huh7, and the results of the cell proliferation test performed at 72 hours are shown in Figures 8A and 8B. The combination index (CI) analysis is also shown in Figures 8C and 8D, which uses the Chou-Talalay method to assess the interaction between these potential therapies as synergistic (CI < 1), additive (CI) =1) or antagonistic (CI>1). As shown by the results, both GP and HH-H3 synergistically inhibited the proliferation of Huh7 with Lysawa.

實施例10 HH-F3在治療徹底破壞初代皮層神經元中的Aβ-誘導神經毒性和在swAPP695-SH-SY5Y的細胞培養中減弱Aβ生產之影響 Example 10 HH-F3 completely destroys Aβ-induced neurotoxicity in primary cortical neurons and attenuates Aβ production in cell culture of swAPP 695 -SH-SY5Y

該初代皮層神經元係以載體(0.1%的DMSO)或HH-F3預處理2小時,隨後曝露於10μM的Aβ25-3540小時。培育後,藉由MTT檢驗檢測細胞存活率。如圖9所示,由MTT還原測得10μM的Aβ25-35纖絲導致細胞存活率明顯降低39.4±7.5%,並且Aβ25-35誘導的細胞死亡被HH-F3處理以劑量依賴的方式消除。 The primary cortical neuron was pretreated with vehicle (0.1% DMSO) or HH-F3 for 2 hours and then exposed to 10 μM Aβ 25-35 for 40 hours. After incubation, cell viability was determined by MTT assay. As shown in Figure 9, 10 μM of Aβ 25-35 fibrils detected by MTT reduction resulted in a significant decrease in cell viability by 39.4 ± 7.5%, and Aβ 25-35 induced cell death was attenuated by HH-F3 in a dose-dependent manner. .

目前,用於阿茲海默症的單一療法策略的失敗表示有超過一種途徑貢獻於阿茲海默症的發病機制。具有超過一種標靶的藥物可能是更有希望的。因此,HH-F3亦測試來檢查其在swAPP695-SH-SY5Y細胞培養中可否減少Aβ產生。該SH-SY5Y細胞中的細胞外Aβ水平低於可檢測極限,但swAPP695-SH-SY5Y細胞中的Aβ40和Aβ42水平分別為1.2ng/ml和57.5pg/ml。 HH-F3的處理之後,在SH-SY5Y細胞上沒有發現毒性(見圖10A)。藉由ELISA測定細胞外的Aβ40和Aβ42水平。該結果顯示於圖10B和圖10C,其指出50μg/ml的HH-F3具有能力將Aβ40和Aβ42的產生分別減少30.0±11.2%和36.8±16.0%。 At present, the failure of monotherapy strategies for Alzheimer's disease indicates that there is more than one way to contribute to the pathogenesis of Alzheimer's disease. Drugs with more than one target may be more promising. Therefore, HH-F3 was also tested to see if it could reduce Aβ production in swAPP 695 -SH-SY5Y cell culture. The level of extracellular Aβ in the SH-SY5Y cells was below the detectable limit, but the levels of Aβ 40 and Aβ 42 in swAPP 695 -SH-SY5Y cells were 1.2 ng/ml and 57.5 pg/ml, respectively. After treatment with HH-F3, no toxicity was found on SH-SY5Y cells (see Figure 10A). Extracellular Aβ 40 and Aβ 42 levels were determined by ELISA. The results are shown in Figures 10B and 10C, which indicate that 50 μg/ml of HH-F3 has the ability to reduce the production of Aβ 40 and Aβ 42 by 30.0 ± 11.2% and 36.8 ± 16.0%, respectively.

細胞外Aβ累積可藉由抑制澱粉樣蛋白變性途徑或增加Aβ清除來減少,包含增強Aβ降解酶的活性和自噬。因此,進行免疫轉印法來檢測APP和APP-CTF的水平(圖11A)。在以指定濃度的HH-F3處理24小時後,總體APP的水平沒有明顯改變。出乎意料的是,當APP係以抗Aβ 1-40抗體免疫轉印時,檢測到一個未驗明的APP種類(APP-XF),而且該種類被HH-F3減少。基本上,該APP-CTF的水平係增加(圖11A)。該γ-分泌酶抑製劑係使用來驗證γ-分泌酶對APP-CTF水平的涉入(圖11A)。另一方面,主要的Aβ降解酶,例如細胞中的胰島素降解酶(insulin degradation enzyme,IDE)和腦啡肽酶(neprilysin,NEP)的表現並沒有受到HH-F3影響(圖11B)。藉由24小時的培育後檢查其中剩餘的Aβ1-40以檢測SH-SY5Y-APP695細胞的條件培養基中Aβ降解酶的活性(圖11C)。結果顯示,HH-F3活化了Aβ降解酶的活性以促進條件培養基中Aβ1-40的清除,因為條件培養基中IDE和NEP的水平並未受到HH-F3影響(圖11D)。 Extracellular Aβ accumulation can be reduced by inhibiting the amyloid degeneration pathway or increasing Aβ clearance, including enhancing Aβ degrading enzyme activity and autophagy. Therefore, an immunotransfer method was performed to detect the levels of APP and APP-CTF (Fig. 11A). There was no significant change in the level of overall APP after 24 hours of treatment with the indicated concentrations of HH-F3. Unexpectedly, when APP was immunotransferred with an anti-Aβ 1-40 antibody, an unidentified APP species (APP-XF) was detected and the species was reduced by HH-F3. Basically, the level of the APP-CTF is increased (Fig. 11A). This γ-secretase inhibitor was used to verify the involvement of γ-secretase in APP-CTF levels (Fig. 11A). On the other hand, major Aβ degrading enzymes such as insulin degradation enzyme (IDE) and enprilysin (NEP) in cells were not affected by HH-F3 (Fig. 11B). The remaining Aβ1-40 was examined by 24 hours of incubation to detect the activity of Aβ degrading enzyme in the conditioned medium of SH-SY5Y-APP695 cells (Fig. 11C). The results showed that HH-F3 activated the activity of Aβ degrading enzyme to promote the clearance of Aβ1-40 in the conditioned medium because the levels of IDE and NEP in the conditioned medium were not affected by HH-F3 (Fig. 11D).

自噬係涉及Aβ的清除。最近的報告指出在AD患者的腦中beclin-1(一種早期自噬調控器)向下調控(Pickford等人,The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice,J Clin Invest 2008;118:2190-2199)。亦測試了HH-F3對swAPP695-SH-SY5Y細胞中自噬活 化的影響。該APP695-SHSY-5Y細胞係與載體或HH-F3培育24小時,並藉由西方墨漬法檢測LC3自噬標記。該結果記載於圖12。 Autophagy is involved in the clearance of Aβ. Recent reports indicate that beclin-1 (an early autophagy regulator) is down-regulated in the brains of AD patients (Pickford et al, The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice) , J Clin Invest 2008; 118: 2190-2199). The effect of HH-F3 on autophagy activation in swAPP 695 -SH-SY5Y cells was also tested. The APP 695 -SHSY-5Y cell line was incubated with vehicle or HH-F3 for 24 hours and the LC3 autophagy marker was detected by Western blotting. This result is shown in FIG.

HH-F3含有多酚化合物,例如沒食子酸(gallic acid)和單寧酸(tannic acid)。多酚化合物已被報告可抑制Aβ凝聚。在此專利中,使用硫代黃素T(Thioflavin T,TH-T),一種可以插入Aβ的β-摺疊片結構的分子來檢測Aβ凝聚。當與澱粉樣蛋白纖絲結合時,Th-T的發射產生偏移,從而在其螢光光譜中引發密集的特定發射帶(482nm)。該結果顯示單寧酸(10μM)和剛果紅(10μM)抑制了Aβ1-40和Aβ1-42纖絲兩者的形成。HH-F3以濃度依賴的方式抑制Aβ1-40和Aβ1-42兩者的凝聚(圖13)。 HH-F3 contains polyphenolic compounds such as gallic acid and tannic acid. Polyphenolic compounds have been reported to inhibit Aβ aggregation. In this patent, thioflavin T (TH-T), a molecule that can be inserted into the β-sheet structure of Aβ, is used to detect Aβ aggregation. When combined with amyloid fibrils, the emission of Th-T shifts, causing a dense specific emission band (482 nm) in its fluorescence spectrum. The results show that tannic acid (10 μM) and Congo red (10 μM) inhibited the formation of both Aβ 1-40 and Aβ 1-42 fibrils. HH-F3 inhibited aggregation of both Aβ 1-40 and Aβ 1-42 in a concentration-dependent manner ( FIG. 13 ).

預防Aβ介導的神經毒性已成為AD治療的重要治療策略。Aβ25-35是APP的膜間域中11個胺基酸的合成肽。其明顯的是Aβ25-35無法透過正常的APP處理產生。然而,Aβ25-35係選擇作為全長Aβ的模型,因其保留了其物理和生物性質兩者,同時其短的長度可容易允許衍生物被合成和研究(Hughes等人,Inhibition of toxicity in the beta-amyloid peptide fragment beta-(25-35)using N-methylated derivatives:a general strategy to prevent amyloid formation.J Biol Chem 2000;275:25109-25115)。雖然其不存在生物系統中,但Aβ25-35片段被許多科學家廣泛地與內生片段Aβ42一起使用或取而代之,而且被發現至少與全長片段一樣毒(Yankner等人,Neurotoxicity of a fragment of the amyloid precursor associated with Alzheimer's disease.Science 1989;245:417-420)。皮層神經元被HH-F3預處理2小時。隨後,使用10μM的Aβ25-35刺激細胞46小時。該Aβ25-35-介導的細胞存活率損失被HH-F3明顯恢復。20和50μg/ml的HH-F3分別明顯恢復了19%和33%的細胞存活率。發生了該HH-F3對細胞存活率的濃度依賴恢復效應,而該HH-F3的處理是在Aβ 處理前而非處理後(圖14)。 Prevention of Aβ-mediated neurotoxicity has become an important therapeutic strategy for AD therapy. Aβ 25-35 is a synthetic peptide of 11 amino acids in the intermembrane domain of APP. It is obvious that Aβ 25-35 cannot be produced by normal APP treatment. However, the Aβ 25-35 line was selected as a model for full-length Aβ because it retains both its physical and biological properties, while its short length can easily allow derivatives to be synthesized and studied (Hughes et al., Inhibition of toxicity in the Beta-amyloid peptide fragment beta-(25-35) using N-methylated derivatives: a general strategy to prevent amyloid formation. J Biol Chem 2000;275:25109-25115). Although it is not present in biological systems, the Aβ 25-35 fragment has been widely used or replaced by many scientists with the endogenous fragment Aβ 42 and has been found to be at least as toxic as the full-length fragment (Yankner et al., Neurotoxicity of a fragment of the Amyloid precursor associated with Alzheimer's disease. Science 1989; 245: 417-420). Cortical neurons were pretreated with HH-F3 for 2 hours. Subsequently, cells were stimulated with 10 μM of Aβ 25-35 for 46 hours. This Aβ 25-35 -mediated loss of cell viability was significantly restored by HH-F3. 20 and 50 μg/ml of HH-F3 significantly restored cell survival rates of 19% and 33%, respectively. The concentration-dependent recovery effect of the HH-F3 on cell viability occurred, and the HH-F3 treatment was before Aβ treatment but not after treatment (Fig. 14).

HH-F3在體外的AD病理學中擁有多種功能。因此,該動物模型的試驗係執行以調查HH-F3的體內效果。該APP/PS1轉基因小鼠在120天大時在大腦皮層中發展出Aβ斑塊(Radde等人,2006)。140天大的APP/PS1小鼠係口服投予HH-F3(300mg/kg/天)一個月。在體外檢驗中,本發明已經證實HH-F3可以抑制Aβ凝聚。因此,將大腦半球中的澱粉樣蛋白斑塊染色(圖15)。由於與Aβ的高親和力,染料1-溴-2,5-雙(3-羧基-4-羥苯乙烯基)苯(1-Bromo-2,5-bis(3-carboxy-4-hydroxystyryl)benzene,BSB)一直被用來檢測澱粉樣蛋白斑塊。該結果顯示HH-F3僅輕微減少Aβ斑塊的負荷。載體組和HH-F3組之間的Aβ斑塊負荷數為43.3±23.1和22.3±7.5(圖15)。 HH-F3 has multiple functions in AD pathology in vitro. Therefore, the test of this animal model was performed to investigate the in vivo effect of HH-F3. The APP/PS1 transgenic mice developed A[beta] plaques in the cerebral cortex at 120 days of age (Radde et al., 2006). The 140-day-old APP/PS1 mouse was orally administered HH-F3 (300 mg/kg/day) for one month. In an in vitro assay, the present inventors have demonstrated that HH-F3 can inhibit A[beta] coagulation. Therefore, amyloid plaques in the cerebral hemisphere were stained (Fig. 15). Due to its high affinity with Aβ, the dye 1-bromo-2,5-bis(3-carboxy-4-hydroxystyryl)benzene , BSB) has been used to detect amyloid plaques. This result shows that HH-F3 only slightly reduces the load of Aβ plaques. The number of Aβ plaque loads between the vehicle group and the HH-F3 group was 43.3 ± 23.1 and 22.3 ± 7.5 (Fig. 15).

本發明使用兩個步驟來分離可溶和不可溶Aβs。該可溶Aβs係溶於可含有單體、二聚物和寡聚物的2% SDS級分中。該不可溶Aβs係溶於70%的甲酸級分中。然後藉由ELISA量測可溶和不可溶Aβs。HH-F3減少了APP/PS1小鼠的大腦皮層中的可溶和不可溶Aβ(圖16)。 The present invention uses two steps to separate soluble and insoluble A[beta]s. The soluble A[beta]s are soluble in a 2% SDS fraction which may contain monomers, dimers and oligomers. The insoluble A[beta]s are soluble in the 70% formic acid fraction. Soluble and insoluble Aβs were then measured by ELISA. HH-F3 reduced soluble and insoluble Aβ in the cerebral cortex of APP/PS1 mice (Fig. 16).

HH-F3影響SH-SY5Y-APP695細胞中的Aβ清除。因此,藉由免疫轉印法檢測APP/PS1小鼠的大腦皮層中與Aβ清除有關的蛋白(圖17A)。與野生型小鼠相比,該轉基因小鼠的IDE水平被向下調控,而NEP和APOE的水平被HH-F3向上調控。此外,與野生型小鼠相比,該轉基因小鼠的PPARγ和pAMPK水平被向下調控,而PPARγ、pAKT及pAMPK明顯地被HH-F3向上調控(圖17B)。 HH-F3 affects Aβ clearance in SH-SY5Y-APP695 cells. Therefore, proteins associated with Aβ clearance in the cerebral cortex of APP/PS1 mice were detected by immunotransfer method (Fig. 17A). The IDE levels of the transgenic mice were down-regulated compared to wild-type mice, while the levels of NEP and APOE were up-regulated by HH-F3. Furthermore, PPARγ and pAMPK levels were down-regulated in this transgenic mouse compared to wild-type mice, while PPARγ, pAKT and pAMPK were significantly up-regulated by HH-F3 (Fig. 17B).

GP的粗萃取物(H2O、甲醇、100%乙醇、70%乙醇、或50%乙醇)沒有明顯影響Hep3B/T2中的HBsAg水平。DMSO、30% DMSO及80% 丙酮的GP萃取物可以部分降低Hep3B/T2中的HBsAg水平(圖18)。 The crude extract of GP (H 2 O, methanol, 100% ethanol, 70% ethanol, or 50% ethanol) did not significantly affect HBsAg levels in Hep3B/T2. GP extracts in DMSO, 30% DMSO, and 80% acetone partially reduced HBsAg levels in Hep3B/T2 (Figure 18).

鑑於以上的結果,HH-F3有望用於阿茲海默症的治療。 In view of the above results, HH-F3 is expected to be used for the treatment of Alzheimer's disease.

總而言之,該GP萃取物或HH-F3級分係具有潛力作為一種新穎療法用於患有代謝性疾病的患者,該代謝性疾病例如糖尿病、肥胖、脂肪肝疾病,包括酒精性脂肪肝疾病(AFLD)、非酒精性脂肪肝疾病(NAFLD)、HBV相關的肝相關疾病(包括HCC、肝纖維化或肝硬化)、糖尿病相關的肝相關疾病、肥胖相關的肝相關疾病、及肥胖引起的高血膽固醇或高三酸甘油酯水平。其建議的是可使用GP萃取物或HH-F3級分來治療或預防代謝症候群。 In conclusion, the GP extract or HH-F3 fraction has potential as a novel therapy for patients with metabolic diseases such as diabetes, obesity, fatty liver disease, including alcoholic fatty liver disease (AFLD). ), nonalcoholic fatty liver disease (NAFLD), HBV-related liver-related diseases (including HCC, liver fibrosis, or cirrhosis), diabetes-related liver-related diseases, obesity-related liver-related diseases, and high blood caused by obesity Cholesterol or high triglyceride levels. It is suggested that GP extract or HH-F3 fraction can be used to treat or prevent metabolic syndrome.

此外,本發明中指出的是該GP萃取物或HH-F3級分能夠活化AMPK途徑和自噬途徑。因此,建議可以將GP萃取物或HH-F3級分發展成為用於預防或治療神經退化性疾病或澱粉樣蛋白相關疾病的藥物,例如帕金森氏症、阿茲海默症、及杭丁頓氏症、而且甚至是抗衰老的藥物。 Furthermore, it is indicated in the present invention that the GP extract or HH-F3 fraction is capable of activating the AMPK pathway and the autophagy pathway. Therefore, it is recommended that the GP extract or the HH-F3 fraction be developed into a drug for preventing or treating a neurodegenerative disease or an amyloid-related disease, such as Parkinson's disease, Alzheimer's disease, and Huntington. And even anti-aging drugs.

據信,本發明所屬技術領域中具有通常知識之人士可以基於本文中的描述而將本發明利用到最寬的範圍,並不需要進一步的說明。因此,所提供的描述和申請專利範圍應被理解為用於演示的目的,而不是要以任何方式限制本發明的範圍。 It is believed that one of ordinary skill in the art to which the present invention pertains can utilize the invention to the broadest scope and no further description. The scope of the invention is to be construed as being limited by the scope of the invention.

Claims (29)

一種組合物用於製備治療代謝性疾病的藥物之用途,其中該組合物包含一來自縞辦屬(Graptopetalum sp.)或紅景天屬(Rhodiola sp.)的二甲亞碸(dimethyl sulfoxide,DMSO)萃取物或級分、或一分離自該DMSO萃取物或級分的活性化合物。 Use of a composition for the preparation of a medicament for treating a metabolic disease, wherein the composition comprises a dimethyl sulfoxide (DMSO) from Graptopetalum sp. or Rhodiola sp. An extract or fraction, or an active compound isolated from the DMSO extract or fraction. 如申請專利範圍第1項之用途,其中該代謝性疾病為糖尿病、肥胖、脂肪肝疾病、B型肝炎病毒(Hepatitis B virus,HBV)有關的肝相關疾病、糖尿病有關的肝相關疾病、肥胖有關的肝相關疾病、或肥胖誘發的高血膽固醇或高三酸甘油酯含量。 For example, the use of the patent scope of the first aspect, wherein the metabolic disease is diabetes, obesity, fatty liver disease, hepatitis B virus (HBV) related liver related diseases, diabetes related liver related diseases, obesity-related Hepatic-related disease, or obesity-induced high blood cholesterol or high triglyceride levels. 如申請專利範圍第2項之用途,其中該代謝性疾病為糖尿病。 The use of the second aspect of the patent application, wherein the metabolic disease is diabetes. 如申請專利範圍第2項之用途,其中該代謝性疾病為肥胖。 The use of the second aspect of the patent application, wherein the metabolic disease is obesity. 如申請專利範圍第2項之用途,其中該代謝性疾病為脂肪肝疾病。 The use of the second aspect of the patent application, wherein the metabolic disease is fatty liver disease. 如申請專利範圍第5項之用途,其中該脂肪肝疾病為酒精性脂肪肝疾病(alcoholic fatty liver disease,AFLD)或非酒精性脂肪肝疾病(nonalcoholic fatty liver disease,NAFLD)。 The use of the fifth aspect of the patent application, wherein the fatty liver disease is alcoholic fatty liver disease (AFLD) or nonalcoholic fatty liver disease (NAFLD). 如申請專利範圍第2項之用途,其中該代謝性疾病為B型肝炎病毒(HBV)有關的肝相關疾病。 The use of the second aspect of the patent application, wherein the metabolic disease is a hepatitis B virus (HBV) related liver related disease. 如申請專利範圍第2項之用途,其中該代謝性疾病為糖尿病有關的肝相關疾病。 The use of the second aspect of the patent application, wherein the metabolic disease is a liver-related disease associated with diabetes. 如申請專利範圍第2項之用途,其中該代謝性疾病為肥胖有關的肝相關疾病。 The use of the second aspect of the patent application, wherein the metabolic disease is a liver-related disease associated with obesity. 如申請專利範圍第7-9項中任一項之用途,其中該肝相關疾病為肝細胞 癌(Hepatocellular carcinoma,HCC)、肝纖維化或肝硬化。 The use of any one of the claims 7-9, wherein the liver related disease is hepatocyte Hepatocellular carcinoma (HCC), liver fibrosis or cirrhosis. 如申請專利範圍第10項之用途,其中該肝相關疾病為肝細胞癌(HCC)。 The use of the scope of claim 10, wherein the liver related disease is hepatocellular carcinoma (HCC). 如申請專利範圍第7項之用途,其中該B型肝炎病毒(HBV)有關的肝相關疾病為HBV誘發的肝纖維化和肝硬化。 For example, the use of the hepatitis B virus (HBV)-related liver-related disease is HBV-induced liver fibrosis and cirrhosis. 一種組合物用於製備預防或治療代謝症候群的藥物之用途,其中該組合物包含一來自縞辦屬或紅景天屬的二甲亞碸(DMSO)萃取物或級分、或一分離自該DMSO萃取物或級分的活性化合物。 Use of a composition for the preparation of a medicament for preventing or treating metabolic syndrome, wherein the composition comprises a dimethyl hydrazine (DMSO) extract or fraction from the genus Rhodiola or Rhodiola, or a DMSO extract or fraction of active compound. 一種組合物用於製備預防或治療癌症的藥物之用途,其中該組合物包含一來自縞辦屬或紅景天屬的二甲亞碸(DMSO)萃取物或級分、或一分離自該DMSO萃取物或級分的活性化合物,與一抗癌藥物在預防或治療癌症上具有協同作用之比例結合。 Use of a composition for the preparation of a medicament for preventing or treating cancer, wherein the composition comprises a dimethyl hydrazine (DMSO) extract or fraction from the genus Rhodiola or Rhodiola, or an isolated from the DMSO The active compound of the extract or fraction is combined with a synergistic ratio of an anticancer drug in preventing or treating cancer. 如申請專利範圍第14項之用途,其中該癌症為肝癌,且該抗癌藥物為一抗肝癌藥物。 For example, the application of the scope of claim 14 wherein the cancer is liver cancer, and the anticancer drug is an anti-hepatocarcinoma drug. 如申請專利範圍第15項之用途,其中該抗癌藥物為蕾莎瓦(Sorafenib)。 For example, the application of the scope of claim 15 wherein the anticancer drug is Sorafenib. 如申請專利範圍第15項之用途,其中該癌症為HCC,並且該抗癌藥物為蕾莎瓦。 The use of the scope of claim 15 wherein the cancer is HCC and the anticancer drug is Lysawa. 一種組合物用於製備活化AMPK途徑或自噬途徑的藥物之用途,其中該組合物包含一來自縞辦屬或紅景天屬的二甲亞碸(DMSO)萃取物或級分、或一分離自該DMSO萃取物或級分的活性化合物。 Use of a composition for the manufacture of a medicament for activating an AMPK pathway or an autophagy pathway, wherein the composition comprises a dimethyl hydrazine (DMSO) extract or fraction, or a separation from the genus Rhodiola or Rhodiola The active compound from the DMSO extract or fraction. 一種組合物用於製備預防或治療神經退化性疾病的藥物之用途,其中該組合物包含一來自縞辦屬或紅景天屬的二甲亞碸(DMSO)萃取物或級分、或分離自該DMSO萃取物或級分的活性化合物。 Use of a composition for the preparation of a medicament for preventing or treating a neurodegenerative disease, wherein the composition comprises a dimethyl hydrazine (DMSO) extract or fraction from the genus Rhodiola or Rhodiola, or isolated from The DMSO extract or fraction of the active compound. 如申請專利範圍第19項之用途,其中該神經退化性疾病為帕金森氏症、阿茲海默症、及杭丁頓氏症。 The use of claim 19, wherein the neurodegenerative disease is Parkinson's disease, Alzheimer's disease, and Huntington's disease. 一種組合物用於製備預防或治療澱粉樣蛋白(amyloid)相關疾病的藥物之用途,其中該組合物包含一來自縞辦屬或紅景天屬的二甲亞碸(DMSO)萃取物或級分、或一分離自該DMSO萃取物或級分的活性化合物。 Use of a composition for the preparation of a medicament for preventing or treating amyloid-related diseases, wherein the composition comprises a dimethyl hydrazine (DMSO) extract or fraction from the genus Rhododendron or Rhodiola Or an active compound isolated from the DMSO extract or fraction. 如申請專利範圍第21項之用途,其中該澱粉樣蛋白相關疾病為阿茲海默症、第2型糖尿病、帕金森氏症、杭丁頓氏症、致死性家族失眠症(Fatal Familial Insomnia)、或類風濕性關節炎。 The use of the scope of claim 21, wherein the amyloid-related diseases are Alzheimer's disease, type 2 diabetes, Parkinson's disease, Huntington's disease, and fatal family insomnia (Fatal Familial Insomnia) Or rheumatoid arthritis. 一種組合物用於製備抗老化藥物之用途,其中該組合物包含一來自縞辦屬或紅景天屬的二甲亞碸(DMSO)萃取物或級分、或分離自該DMSO萃取物或級分的活性化合物。 Use of a composition for the preparation of an anti-aging agent, wherein the composition comprises a dimethyl hydrazine (DMSO) extract or fraction from the genus Rhodiola or Rhodiola, or is isolated from the DMSO extract or grade The active compound of the fraction. 如申請專利範圍第1-23項中任一項之用途,其中縞辦屬為石蓮花(Graptopetalum paraguayense)。 The use of any one of the claims 1 to 23 , wherein the genus is Graptopetalum paraguayense . 如申請專利範圍第1-23項中任一項之用途,其中紅景天屬為紅景天(Rhodiola rosea)。 The use of any one of claims 1 to 23, wherein the Rhodiola rosea is Rhodiola rosea . 如申請專利範圍第1-23項中任一項之用途,其中該級分富含活性成分,其係藉由以二甲亞碸(DMSO)萃取該植物、接著藉由層析分離所製備以獲得一稱為HH-F3的級分。 The use of any one of the preceding claims, wherein the fraction is enriched in an active ingredient by extracting the plant with dimethyl hydrazine (DMSO), followed by chromatographic separation. A fraction called HH-F3 was obtained. 如申請專利範圍第26項之用途,其中Sephadex LH-20管柱被用於層析。 For example, the use of the Sephadex LH-20 column is used for chromatography. 如申請專利範圍第1-23項中任一項之用途,其中該活性化合物為具有一式I結構的化合物 其中R中之一為H或原花青素(prucyanidin,PC)單元,而另一個R為OH或原飛燕草素(prodelphindine,PD)單元;n為一範圍從21至38的數字;以及PC單元對PD單元之比例不超過1:20。 The use of any one of the preceding claims, wherein the active compound is a compound having the structure of Formula I Wherein one of R is H or a procucyanidin (PC) unit, and the other R is OH or a prodelphindine (PD) unit; n is a number ranging from 21 to 38; and the PC unit is PD The ratio of units does not exceed 1:20. 一種如申請專利範圍第28項所提出的化合物用於製備治療代謝性疾病、預防或治療代謝症候群、癌症、或神經退化性疾病、澱粉樣蛋白相關疾病;以及活化AMPK途徑或自噬途徑的藥物;或抗老化藥物之用途。 A compound as claimed in claim 28 for the preparation of a medicament for the treatment of a metabolic disease, prevention or treatment of metabolic syndrome, cancer, or neurodegenerative diseases, amyloid-related diseases; and activating an AMPK pathway or an autophagy pathway Or the use of anti-aging drugs.
TW103138926A 2013-11-08 2014-11-10 Novel use of a dimethyl sulphoxide (dmso) extract or fraction from graptopetalum sp TWI681774B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361901742P 2013-11-08 2013-11-08
US61/901,742 2013-11-08

Publications (2)

Publication Number Publication Date
TW201532610A true TW201532610A (en) 2015-09-01
TWI681774B TWI681774B (en) 2020-01-11

Family

ID=53040927

Family Applications (1)

Application Number Title Priority Date Filing Date
TW103138926A TWI681774B (en) 2013-11-08 2014-11-10 Novel use of a dimethyl sulphoxide (dmso) extract or fraction from graptopetalum sp

Country Status (3)

Country Link
CN (1) CN105813646B (en)
TW (1) TWI681774B (en)
WO (1) WO2015067216A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105342904A (en) * 2015-11-02 2016-02-24 嘉文丽(福建)化妆品有限公司 Essence with skin anti-wrinkle effect

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7364758B2 (en) * 2004-08-31 2008-04-29 Shih-Lan Hsu Pharmaceutical use of Graptopetalum and related plants
CN101428054A (en) * 2007-11-08 2009-05-13 中国医学科学院药物研究所 Uses of rhodiola rosea in preventing and treating insulin resistance, and correlated metabolism diseases
CN103269706B (en) * 2010-09-30 2015-06-24 黄奇英 Anti-cancer extract and compounds
CN103857400B (en) * 2011-08-08 2019-07-12 香港科技大学 Rose gadol extract and isolated compound and its application in treatment neurodegenerative disease

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105342904A (en) * 2015-11-02 2016-02-24 嘉文丽(福建)化妆品有限公司 Essence with skin anti-wrinkle effect
CN105342904B (en) * 2015-11-02 2018-04-10 嘉文丽(福建)化妆品有限公司 A kind of Essence with skin wrinkle resisting effect

Also Published As

Publication number Publication date
TWI681774B (en) 2020-01-11
WO2015067216A1 (en) 2015-05-14
CN105813646B (en) 2020-07-28
CN105813646A (en) 2016-07-27

Similar Documents

Publication Publication Date Title
Shao et al. Nurr1: A vital participant in the TLR4-NF-κB signal pathway stimulated by α-synuclein in BV-2 cells
AU2015254309B2 (en) Muscle atrophy inhibitor containing quercetin glycoside
Yang et al. Defective hepatic autophagy in obesity promotes ER stress and causes insulin resistance
AU2018202291A1 (en) Compositions and methods for increasing lifespan and health span
Wang et al. Dexmedetomidine alleviates LPS-induced apoptosis and inflammation in macrophages by eliminating damaged mitochondria via PINK1 mediated mitophagy
AU2021269396B2 (en) Very-long-chain polyunsaturated fatty acids, elovanoid hydroxylated derivatives, and methods of use
Wu et al. Mechanisms and therapeutic targets of depression after intracerebral hemorrhage
Zhou et al. Polysaccharides from Lycium barbarum ameliorate amyloid pathology and cognitive functions in APP/PS1 transgenic mice
Kang et al. Sappanone A protects mice against cisplatin-induced kidney injury
KR20110115589A (en) Treatment of neurotrophic factor mediated disorders
Baskaran et al. Hypocholesterolemic and antiatherosclerotic potential of Basella alba leaf extract in hypercholesterolemia-induced rabbits
Antony et al. A Pilot clinical study to evaluate the effect of Emblica officinalis extract (Amlamax™) on markers of systemic inflammation and dyslipidemia
JPWO2009028605A1 (en) Preventive or alleviating peripheral neuropathy with anticancer agents
Jiang et al. Icariin Ameliorates Amyloid Pathologies by Maintaining Homeostasis of Autophagic Systems in Aβ 1–42-Injected Rats
Yi et al. Genistein exerts a cell-protective effect via nrf2/ho-1//pi3k signaling in ab25-35-induced alzheimer’s disease models in vitro
JP2019534273A (en) Composition for preventing or treating hepatitis containing monoacetyl diacylglycerol compound
Shao et al. Pinocembrin promotes OPC differentiation and remyelination via the mTOR signaling pathway
Zhou et al. Daphnoretin relieves IL-1β-mediated chondrocytes apoptosis via repressing endoplasmic reticulum stress and NLRP3 inflammasome
Zhang et al. Effects of Chinese medicine Tong xinluo on diabetic nephropathy via inhibiting TGF-β1-induced epithelial-to-mesenchymal transition
Paidi et al. Medha Plus–A novel polyherbal formulation ameliorates cognitive behaviors and disease pathology in models of Alzheimer’s disease
JP7411175B2 (en) Lipocalin-type prostaglandin D2 synthase production promoter
TWI681774B (en) Novel use of a dimethyl sulphoxide (dmso) extract or fraction from graptopetalum sp
Zheng et al. The effect of geniste on Aβ25–35-induced PC12 cell apoptosis through the JNK-dependent Fas pathway
Yang et al. Polygonatum kingianum saponins delay cellular senescence through SASP down-regulation and prolong the healthy lifespan of Caenorhabditis elegans by activating sir-2.1/autophagy
Li et al. A natural small molecule aspidosperma-type alkaloid, hecubine, as a new TREM2 activator for alleviating lipopolysaccharide-induced neuroinflammation in vitro and in vivo