芍芪多苷的研制及其对大鼠化学性肝纤维化的作用和促进胶原代谢的分子机制
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
肝纤维化(hepatic fibrosis,HF)是大多数慢性肝病所共有的病理特征,是肝脏细胞外基质(extracellular matrix,ECM)合成和降解失衡、导致其过度沉积的结果,也是慢性肝炎进一步向肝硬化、肝癌发展的重要环节。近年来的研究证实HF在未进入肝硬化之前,尚有逆转可能,因此预防和逆转HF对慢性肝病、肝硬化等治疗有积极意义。但HF的形成是一个复杂的过程,目前用于治疗或逆转HF的方法大多效果不理想或长期应用不良反应较大。因此,探究HF的发病机制,并在中医药领域寻找高效低毒、能控制HF病情的新药是当前研究的重要方向。白芍和黄芪是中医临床常用的保肝药物,尤其是益气活血、养血柔肝方剂的主要组份。本实验采用均匀设计法优选出白芍和黄芪的最佳剂量配比后,运用现代提取分离技术制得白芍和黄芪复方的芍芪多苷有效部位群(shaoqiduogan,SQDG);在大鼠化学性HF模型上,探讨SQDG对大鼠HF的治疗作用及其特点;并从细胞因子、胶原代谢相关酶的蛋白表达水平等方面研究SQDG治疗大鼠HF的可能机制,从而为抗HF中药新药——SQDG制剂的进一步开发奠定基础。
     目的:研究SQDG有效部位群的制备工艺,并建立相应的质量控制方法;从大鼠肝功能的改变、反映胶原水平的血清和肝脏指标及肝脏病理形态学等的改变,明确SQDG对大鼠HF的治疗作用;从细胞因子、胶原代谢相关酶等方面研究SQDG
Hepatic fibrosis (HF) is the hallmark of most chronic liver diseases. Its essense is excessive deposition of extracellular matrix (ECM) components in liver, which occurs due to an imbalance between the production and degradation of matrix. Collagen is the most important part of ECM. Its final stage is cirrhosis. HF is generally considered reversible conditions. Thus, to prevent or reverse HF has positive significance for therapeutics on chronic liver diseases and cirrhosis. But HF is a complex process, and current therapies targeting at arresting or revering HF are largely ineffective and some has unacceptable side effects in longterm therapy. Therefore, one of important trends focuses on investigation the mechanisms of HF and exploration new effective drugs without ill effects. Radix Paeonia Pall and Radix Astragali are two common traditional herbs in treatment for liver diseases. The present study was designed to optimize the proportion of the two herbs by uniform design and produce the effective compound------shaoqiduogan (SQDG) by modern techniques of extract, purity and separation, andto investigate the effects of SQDG on chemical HF of rat and its possible mechanisms. AIM To study on preparation process of SQDG and establish its quality standards. From the changes of liver function indices, liver index, spleen index, ECM levels on
    serum and liver, and histological morphological of HF rats induced by CCI4, we observed the effects of SQDG on HF of rat. We explored its possible mechanisms on HF from cytokines and collagen metabolism. At last, we observed the effect on promotion HSC-T6 collagen degradation of SQDG and the changes of the expressions of G a i and G a s of HSC-T6 stimulated with TGF-pM by Western - blot analysis. METHODS The CCI4 -treated mice acute liver injury model was used to observe the protective effects of total glucosides of paeony (TGP) and astragalosides (ASTs). Uniform design was used to optimize the best proportion of Radix Paeonia Pall and Radix Astragali. The optimum extraction was investigated by orthogonal design. Macroporous resin was used to separate SQDG. High performance liquid chromatography(HPLC), high performance liquid chromatography-evaporative light scattering detector ( HPLC-ELSD) and colormetric method were used to determine paeoniflorin(Pae), astragaloside IV and ASTs, respectively. Experimental rat models of chemical HF induced by CCI4 was used to investigated intervention effects of SQDG. SQDG (42.5, 85, 170 mg-kg-l-d-1, 6d-w-l) were given by intragastric administration. Colchicine (Col), TPG, ASTs were given as positive control.The alanine aminotransferase (ALT), aspartate aminotransferase (AST) levels, A/G in plasma and hydroxyproline (Hyp) contents in liver tissue were assayed by spectrophotometry. The body weight, liver and spleen weight were assayed. The hyaluronic acid (HA), procollagen III (PCIII) and collagen I (C- I ) were assessed by radioimmunoassay. The transforming growth factor betal (TGF-pi) level in plasma was determined by enzyme-linked immunosorbent assay (ELISA).TGF-pi, C- I , matrix metalloproteinasel3 (MMP-13) and tissue inhibitors of metalloproteinasel (TIMP-1) levels in liver were assayed by immunohistochemistry. Hepatic tissue sections were stained with hematoxylin-eosin (HE), Masson, and Gomori examined under a light microscope, and scored.In vitro, the collagen production of HSC-T6 cells stimulated with TGF-pi was
    measured with 3H-Proline incorporation. MMP-13 and TIMP-1 levels in culture supernatants of HSC-T6 cells stimulated with TGF-pl were determined by ELISA.Western-blot analysis was used to detect the changes of the expression of G-protein on HSC-T6 cell membrane stimulated by TGF-0 Qug-L"1) and effects on its of SQDG.RESULTS1. Preparation process and quality standards of SQDG were established.According to the extraction ratio of TGP and ASTs, the optimum proportion of Radix Paeonia Pall and Radix Astragali was decided to be 4 I 1 by uniform design. The optimum preparation process of SQDG was investrgated by orthogonal design and macroporous resin column separation as the paeoniflorin content index by HPLC. The optimum alcohol-extracting factors was as follows: 6 times of 70% alcohol, refluxing and extracting for 2 times, 1.5h per times. The extracts water solution was separated by D101 macroporous resin. The optimal separation conditions were as follows: The ratio of herbs and resin was 1 '. 2. The extracts water solution was absorbed by resin with a flow rate of 4 BV/h. Water, 20% alcohol, 40% alcohol and 70% alcohol were used to elute the resin column, respectively. Portions of 20% ethanol and 70 % ethanol were collected, and evaporated to dryness under reduced pressure, which yielded yellow dry powder......SQDG.Scientific method of quality control was established. TLC indentification of Radix Paeonia Pall and Radix Astragali were carried out. HPLC, HPLC-ELSD and colormetric method were used to determine Pae, AST IV and ASTs, respectively. The purity of Pae, ASTs and AST IVin SQDG were 51.23%, 5.03% and 0.686%, respectively.All these results showed that the process of extraction and purity was feasible, and the methods of quality control were scientific and reliable.2, SQDG could intervene in chemical hepatic fibrosis of ratsThe HF models in rats were induced by CCU. The histological morphological
    examination of rat livers showed that SQDG (85, 170 mg-kg"1, ig) significantly reduced the scores of liver fibrosis of HF rats. SQDG (85, 170 mg-kg"1, ig) treatment markedly alleviated the degree of liver fibrogenesis and formation of pseudolobulus.Administration with SQDG (85, 170 mg-kg"1, ig) significantly reduced the elevated serum ALT, AST activities and elevated the reduced A/G of HF rats. Administration with SQDG (85, 170 mg-kg"1, ig) significantly reduced the elevated liver index and administration with SQDG (170 mg-kg"1, ig) significantly reduced the elevated spleen index. The results illustrated that SQDG has protective effects on liver of HF, and suggested that its effects on HF was related to improving liver functions.Administration with SQDG (85, 170 mg-kg"1, ig) significantly reduced the elevated serum HA, PCIII, C- I levels and Hyp content in liver homogenate. It suggested that SQDG could prevent HF via reducing collagen deposition.3. Inhibiting secretion of TGF-pi was one of important characteristics of SQDG preventing HFTGF-J31 is one of the most powerful profibrofenic activity cytokines. The results of ELISA and immunohistochemistry demonstrated that SQDG significantly reduced the elevated serum TGF-pi level, and significantly decreased TGF-pl expression in liver. The results showed that SQDG could inhibit HSC activation and decrease collagen production by reducing secretion of TGF-pi.4. Controlling the levels of MMP-13 and TIMP-1 to promote collagen degradation SQDG is one of important mechanisms of SQDG preventing HFThe results immunohistochemistry demonstrated that SQDG significantly increased MMP-13 expression and significantly decreased TIMP-1 expression in liver. In vitro, SQDG (20~160mg-L'1) significantly raised MMP-13 level and reduced the elevated TIMP-1 level in culture supernatants of HSC-T6 cells stimulated by TGF-pl. Therefore, SQDG could facilitate MMP-13 expression and inhibit TIMP-1 expression. The results suggested that SQDG intervene in rat HF via elevating the decreased
引文
1. Alcolado R, Arthur MJ, Iredale JP. Pathogenesis of liver fibrosis. Clin Sci(Lond). 1997; 92(2): 103~12
    2. Rockey DC. The cell and molecular biology of hepatic fibrogenesis. Clinical and therapeutic implications. Clin Liver Dis, 2000; 4(2): 319~55
    3. Friedman SL. Molecular regulation of hepatic fibrosis, an integrated cellular response to tissue injury, J Biol Chem, 2000; 275(4): 2247~50
    4. Lotersztajn S, Julien B, Teixeira-Clerc F, Lotersztajn S, Julien B, Teixeira-Clerc F, Grenard P, Mallat A. Hepatic fibrosis: molecular mechanisms and drug targets. Annu Rev Pharmacol Toxicol, 2005; 45: 605~28
    5. Beljaars L, Meijer DKF, Poelstra K. Targeting hepatic stellate cells for cell-specific treatment of liver fibrosis. Front-Biosci, 2002; 7: 214~22
    6. Pinzani M. Therapies for hepatic fibrosis: real hope or just academic exercise? Dig Liver Dis, 2004; 36(11): 714~6
    7.何德华,詹镕洲.肝胆病理学.第二军医大学出版社.1997;12:序
    8. Brenner DA. Signal transduction during liver regeneration, J Gastroenterol Hepatol, 1998; 13 (Suppl): S93~5
    9. Zhang LJ, Yu JP, Li D, Huang YH, Chen ZX, Wang XZ. Effects of cytokines on carbon tetrachloride-induced hepatic fibrogenesis in rats. World J Gastroenterol, 2004; 10(1): 77~81
    10.蒋毅萍.中草药抗肝纤维化的研究.中药材,2001;24(3):212~4
    11. Li CX, Li L, Lou J, Yang WX, Lei TW, Li YH, Liu J, Cheng ML, Huang LH. The protective effects of traditional Chinese medicine prescription, Han-Dan-Gan-Le. On CCl_4- induceds liver fibrosis in rats. Am J Chin Med, 1998; 26(3-4): 325~32
    12. Geerts A, Rogiers V. Sho-saiko-To: the right blend of traditional Oriental medicine and liver cell biology [editorial; comment]. Hepatology, 1999; 29(1): 282~4
    13.王伯祥主编.中医肝胆病学.第1版,北京:中国医药科技出版社,1997:1~19
    14. Friedman SL, Lalazar A, Crong L. HSC-T6 cells, an immortalized rat hepatic stellate cell line. Hepatology, 1997; 26(4Pt): 338A
    15. Igarash S, Hatahara T, Nagai Y, Hori H, S akakibara K, Katoh M, Sakai A, Sugimoto T. Anti-fibrotic effect of malotilate on liver fibrosis induced by carbon tetrachloride in rats. Jpn J Exp Med, 1986; 56(5): 235~45
    16.韩德惠.肝羟脯氨酸测定方法.见:陈奇主编.中药药理研究方法学.北京:人民卫生出版社.1993:846~7
    17.全国病毒性肝炎防治方案(试行).中华传染病杂志,1995;13 (4):241~4
    18. Friedman SL. Liver fibrosis - from bench to bedside, J Hepatol, 2003; 38(S1): S38~53
    19. Gressner AM. Transdifferentiation of hepatic setllate cell (Ito cells) to myofibroblasts: a key event in hepatic fibrogenesis. Kidney Int, 1996; 46(54): 39~44
    20. Arthur MJ, Mann DA, Iredale JP. Tissue inhibitors of metalloproteinases, hepatic stellate cells and livr fibrosis. J Gastro Hepatol, 1998; 13(S1): S33~8
    21. Bissell DM. Hepatic fibrosis as wound repair: a progress report. J Castroenteral, 1998; 33(2): 295~302
    22.李颖,王静艳,王玉梅,刘沛.4种血清学指标对各型肝病患者肝纤维化的诊断意义.中国医科大学学报,2003;32(2):138~9,142
    23.周朝晖,佘为民,倪燕君.慢性肝炎患者血清纤维化指标与肝组织纤维化之间的关系.中国临床医学,2003;10(5):655~6
    24.蔡卫民,张彬彬,翁红雷,胡中荣,闾军,郑敏,刘荣华.八项肝纤维化血清标志物比较研究.中华肝脏病杂志,2004;12(4):219~22
    25. Jiang Z, You DY, Chen XC, Wu J. Monitoring of serum markers of fibrosis during CCL_4-induced liver damage. Effects of antifibrotic agents. J Hepatol, 1992; 16(3): 282~9
    26. Shi J, Aisaki K, Ikawa Y, Wake K. Evidence of hepatocyte apoptosis in rat liver after the administration of carbon tetrachloride. Am J Pathol, 1998; 153(2): 515~25
    27.孔宪高,高峰.肝纤维化的实验与临床研究.第二军医大学学报,1996;17(1):1
    28. Boobis AR, Fawthrop DJ, Davis DS. Mechanisms of cell death. Trends Pharmacol Sci, 1989; 10(7): 275~80
    29. Parola M, Leonarduzzi G, Bisai F, Albano E, Biocca ME, Poli G, Dianzani MU. Vitamin E dietary supplementation protects against carbon tetrachloride-induced chronic liver damage and cirrhosis. Hepatology, 1992; 16(4): 1014~21.
    30. Burk RF, Lane JM, Patel K. Relationship of oxygen and glutathion in protection against carbon tetrachloride-induced hepatic microsomal lipip peroxidation and covalent binding in the rat. J Clin invest, 1984; 74 (6): 1996~2001
    31.徐叔云,陈敏珠,魏伟等,白芍总苷免疫药理与临床。见:周金黄,刘干中主编:中药药理与临床研究进展(第一册),北京:中国科学技术出版社,1992:49~59
    32.魏伟,刘家骏,刘家琴,李旭,徐叔云.白芍总苷对乙型肝炎的治疗作用及其前景.中国药理学通报,2000;16(5):597~8
    33.郭浩,魏伟,陈敏珠,等.白芍总苷对免疫调节细胞的作用.中国药理学与毒理学杂志,1993,7(3):191~5
    34.魏文树,汪文琦,王玉山,等.白芍总苷对免疫应答的调控作用.中国药理学通报,1987;3(3):148~50
    35.周玲玲,魏伟,沈玉先,徐叔云.白芍总苷对小鼠系统性红斑狼疮样改变的保护作用.中国药理学通报,2002;18(2):175~7
    36.王华,魏伟,岳莉,汪倪萍,桂双英,吴丽,孙妩弋,徐叔云.白芍总苷对卡介苗加脂多糖引起的小鼠免疫性肝损伤的保护作用.中国药理学通报,2004;20(8):875~8
    37.詹可顺.白芍总苷对急性肝损伤的保护作用及其可能机制.安徽医科大学硕士研究生毕业论文,2003
    38. Wang H, Wei W, Wang NP, Wu CY, Yan SX, Yue L, Zhang LL, Xu SY. Therapeutic effects of total glucosides of paeony on rats with immunological hepatic fibrosis. World J Gastroenterol. 2005; 11(14): 2124~9
    39.来季华.丹参合并黄芪对慢性肝炎、肝硬化抗纤维化治疗疗效与预后分析.中成药,2003;25(8):690,附1~2
    40.马红,王宝恩,等.黄芪对免疫损伤性肝纤维化大鼠的治疗作用.中西医结合肝病杂志,1998;7 (1):32~5
    41.吴强,杨雁,薛绍礼,张宵翔,邹宇宏,陈敏珠.黄芪总苷对肝星状细胞增殖和合成胶原的抑制作用.中国药理学通报,2003;19 (8):892~5
    42.韩涛,王宝恩,钱绍诚,马雪梅,王爱民,郑丽娜,高英堂.黄芪对肝星状细胞胶原降解基因表达的影响.天津医科大学学报,2003;9(2):159~61
    43.宋少刚,杨雁,陈敏珠.黄芪总提物对大鼠肝星状细胞增殖及其产生胶原的影响,中国临床药理学与治疗学,2001;6 (2):111~3
    44. Gui SY, Wei W, Wang H, Wu L, Sun WY, Wu CY. Protective effect of fufanghuangqiduogan against acute liver injury in mice. World J Gasiroenterol. 2005; 11(19): 2984~9
    45.陈岳祥.肝纤维化药物治疗研究现状.国外医学消化系疾病分册
    46. Cedilo A, Mourelle M, Muriel P. Effect of colchicines and trimethylcolchicinic acid on CCl_4-induced cirrhosis in the rat. Pharmacology Toxicology. 1996; 79(4): 241~6
    47. Li D, Friedman SL. Liver fibrogenesis and the role of hepatic stellate cells: New insights and prospects for therapy. J Gastroenterol Hepatol, 1999; 14(7): 618~33
    48. Vawada N. The hepatic prerisinusoidal stellate cell. Histol Histopathol, 1997; 12(4): 1069~80
    49. George J, Pera N, Phung N, Leclercq I, Yun Hou J, Farrell G. Lipid peroxidation, stellate cell activation and hepatic fibrogenesis in a rat model of chronic steatohepatitis. J Hepatol, 2003; 39 (5): 756~64
    50. Lu GM, Shimizu I, Cui XZ, Itonaga M, Tamaki K, Fukuno H, Inoue H, Honda H, Ito S. Antioxidant and antiapoptotic activities of idoxifene and estradiol in hepatic fibrosis in rats. Life Sciences, 2004; 74(7): 897~907
    51.王红.脂质过氧化与肝纤维化.国外医学临床生物化学与检验学分册.1998;19(3):138~9
    52. Bissell DM, Friedman SL, Maher JJ, Roll FJ. Connective tissue biology and hepatic fibrosis: report of a conference. Hepatology, 1990; 11 (3): 488~98
    53. Plebani M, Burlina A. Biochemical makers of hepatic fibrosis. Chin Biochem, 1991; 24(3): 219~39
    54.李继强,曾民德,范建高.病毒性肝炎患者细胞外基底成分的检测及其与肝纤维化的关系.上海医学,1996;19(3):273~5
    55.张世兰,杨永平,陈德永,宋喜秀,杨守纯,王成武.血清透明质酸、Ⅲ型胶原氨基肽、Ⅳ型胶原及层连蛋白与PGA指数对肝纤维化的诊断意义.中华传染病杂志,1999;17:261~2
    56.姚树坤,殷飞.肝纤维化的早期诊断.世界华人消化杂志,2000;8(6):681~3
    57. Du WD, Zhang YE, Zhai WR, Zhou XM. Dynamic changes of type Ⅰ, Ⅲ and Ⅳ collagen synthesis and distribution Of collagen-producing cells in carbon tetrachloride -induced rat liver fibrosis. Word J Gastroenterol, 1999; 5(5): 397~403
    58. Loreal O, Clement B, Schuppan D, Rescan PY, Rissel M, Guillouzo A. Distribution and cellular origin of collagen Ⅳ during development and in cirrhosis. Gastroenterology, 1992; 102(3): 980~7
    59. Ramadori G, Knittel T, Saile B. Fibrosis and altered matrix synthesis. Digestion, 1998; 59(4): 372~5
    60.姜慧卿,张晓岚.肝纤维化的发生机制.世界华人消化杂志,2000,8(6):687~9
    61.殷蔚荑,白宁,李杰.大鼠免疫性肝纤维化模型及血清检测指标的研究.中华消化杂志,1989;9 (2):95~7
    62.方步武,刘平,刘成,胡义扬,徐列明,李风华,顾宏图,周丽利.虫草多糖抗免疫损伤性大鼠肝纤维化的作用及其机理研究.上海中医药杂志,2000; 9: 37~40
    63. Boesen CC, Radaev S, Motyka SA, Patamawenu A, Sun PD. The 1.1 A crystal structure of human TGF-beta type II receptor ligand binding domain. Structure, 2002; 10(7): 913~9
    64. Gressner AM. The cell biology of liver fibrogenesis-animbalance of proliferation, growth arrest and apoptosis of myofiblasts. Cell Tissue Res,1998; 292(3):447~52
    65. Casini A, Pinzani M, Milani S, Grappone C, Galli G, Jezequel AM, Schuppan D, Rotella CM, Surrenti C. Regulation of extracellular matrixs synthesis by transforming growth factor betal in human fat-storing cells. Gastroenterology, 1993; 105(1): 245~53
    66. Yamashita H, ten Dijke P, Franzen P, Miyazono K, Heldin CH. Formation of heterooligomeric complexes of type I and type II receptors for transforming growthin factor beta. J Bio Chem, 1994; 269(31): 20172~8
    67. Benyon BC, Arthur MJ. Mechanisms of hepatic fibrosis. J Pediatr Gastroenterol Nutr. 1998; 27(1): 75-85
    68. Knittel T, Janneck T, Muller L, Fellmer P, Ramadori G. Transforming growth factor betal regulated gene expression of ito cells. Hepatology, 1996; 24(2): 352 ~ 60
    69. Gressner AM, Polzar B, Lahme B, Mannherz HG. Induction of rat liver parenchymal cell apoptosis by hepatic myofiboblasts via trasforming growth factor beta.Hepatology, 1996; 23(3): 571 ~ 81
    70. De Bleser PJ, Niki T, Rogiers V, Geerts A. Transforming growth factor-beta gene expression in normal and fibrotic rat liver. J Hepatol, 1997; 26(4): 886 ~ 93
    71. Bissel DM, Wang SS, Jarnagin WR, Roll FJ. Cell-specific expression of transforming growth factors-beta in rat liver. Evidence for autocrine regulation of hepatocyte proliferation. J Clin Invest, 1995; 96(1): 447~55
    72. Iredale JP. Review: Tissue inhibitors of metalloproteinases in liver fibrosis. Int J Biochem Cell Biol, 1997; 29(1): 43 ~ 54
    73. Nagase H, Woessner JF Jr. Matrix metalloproteinases. J Biol Chem, 1999; 274(31): 21491~4
    74. Murphy G, Docherty AJP, Hembry RM, Reynolds JJ. Metalloproteinases and tissue damage. Br J Rheumatol, 1991; 30( Suppl, 1): 25~31
    75. Leyland H, Gentry J, Arthur MJP, Benyon RC. The plasminogen activating system in hepatic stellate cells. Hepatology, 1996; 24(5): 1172~8
    76. Iredale JP, Benyon RC, Arthur MJ, Ferris WF, Alcolado R, Winwood PJ, Clark N, Murphy G. Tissue inhibitor of metalloproteinase-1 messenger RNA expression is enhanced relative to interstitial collagenase messenger RNA in experimental liver injury and fibrosis. Hepatology, 1996; 24(1): 176~84
    77. Edwards DR, Murphy G, Reynolds JJ, Whitham SE, Docherty AJ, Angel P, Heath JK. Transforming growth factor-beta modulates the expression of collagenase and metalloproteinase inhibitor. EMBO J, 1987; 6(7): 1899~904
    78. McCrudden R, Iredale JP. Liver fibrosis, the hepatic stellate cell and tissue inhibitors of metalloproteinase. Hiatol Histopathol, 2000; 15(4): 1159~68
    79. Vogel S, Piantedosi R, Frank J, Lalazar A, Rockey DC, Friedman SL, Blaner WS. An immortalized rat liver stellate cell line (HSC-T6): a new cell mode for the study of retinoid metaboism in vitro. J Lipid Res, 2000; 41(6): 882~93
    80. Senoo H, Hata RI, Nagai Y. Stellate cells (vitamin A-storing cells) are the primary site of collagen synthesis In nonparenchymal cells in the liver. Bio med Res,1984; 5 (6): 451~8
    81. Isbrucker RA, Peterson TC. Platelet-derived growth factor and pentoxifylline modulate of collagen synthesis in myofibroblasts. Toxicol and Appl Pharmacol, 1998; 149(1): 120—6
    82. Schuppan D, Porov Y. Hepatic fibrosis: from bench to bedside. J Gastroenterol Hepatol, 2002; 17(suppl 3): S300~5
    83. Safadi R, Friedman SL. Hepatic fibrosis-role of hepatic stellate cell activation. Med

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700