过氧化物酶体增殖物激活受体-α在酒精性肝损伤中的动态表达及中药酒肝颗粒的干预作用
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
作为一种多功能的核转录因子,PPAR-α可以在细胞水平抑制NF-κB的转录活性,调控炎性细胞因子的表达,从而抑制局部的炎症反应;可以调控脂肪酸氧化相关基因的表达,在肝脏脂肪细胞分化、脂质的储存、转运及脂肪酸氧化过程中发挥重要作用。
     本课题研究应用白酒灌胃制备酒精性肝损伤大鼠模型,动态观察了酒精性肝损伤进程中PPAR-α的基因表达变化及中药酒肝颗粒的干预作用,并对PPAR-α的基因表达与炎症因子、氧化应激指标之间进行了相关性分析,旨在研究探讨PPAR-α在酒精性肝损伤中可能的作用及酒肝颗粒的作用机理。研究结果提示:PPAR-α参与了酒精性肝损伤,并在其发生、发展进程中具有关键作用,有望成为治疗酒精性肝损伤新的靶点;中药酒肝颗粒可以上调PPAR-α的基因表达,抑制肝脏炎症反应,提高其抗氧化能力,对酒精性肝损伤的具有较好的防治作用。
     本课题研究应用Real-time PCR、RT-PCR及免疫组化等实验方法动态观察了PPAR-α在酒精性肝损伤形成过程中不同时间段的表达变化,结果初步证实PPAR-α在酒精性肝损伤中具有重要作用;同时,从基因及蛋白水平探讨了中药酒肝颗粒治疗酒精性肝损伤的作用机理,为其今后的临床推广应用提供了理论基础。
Insobriety is becoming one severe social problem in the world. Long-term excessive drinking can lead to disorders of enzymatic activities and result in alcoholic liver injury (ALI). A series of pathologic physiology variations will be presented in the liver of ALI, including initial fatty degeneration, following alcoholic hepatitis, alcoholic hepatic fibrosis and final alcoholic liver cirrhosis. And some severe complications, such as ascitic fluid, upper gastrointestinal bleeding, hepatic encephalopathy and so on, might appear at last. The incidence of ALI is increasing in China during the recent years, which results in its second position just under virus hepatitis in liver diseases. ALI has already influenced people quality of life and health severely. Studies manifest that early ALI may be completely recovered if it could be diagnosed and treated in time. On the contrary, it could develop into alcoholic liver cirrhosis,liver cancer and even severe complications, where the pathological changes are too difficult to recover. Thus, it is very significant for the prognosis of patients with ALI to discover, stop drinking and treat positively at the initial stage.
     It is clear that excessive drinking is the causative agent of ALI. While its nosogenesis is very complicated, including toxic effects of alcohol and its metabolin, immuno-modulation, inflammatory factors, apoptosis, endotoxin, genetic polymorphism, additive effects of virus, etc. Moreover, ALI is also correlated with nutritional-state and genetic predisposition. Presently, it is generally accepted that inflammatory factors and oxidative stress play an important role in ALI. Alcohol intake can make some cell factors and mediators of inflammation increase massively, which may lead to inflammation, necrosis, apoptosis, fibrosis in liver and result in cell damage. Besides, long-term drinking can make prooxidants increase and antioxygens decrease, which may aggravate the oxidative stress and lead to liver cells death or apoptosis.
     Recently studies discovered that Peroxisome Proliferator Activated Receptor-α(PPAR-α), as a multifunctional nuclear factor, has many biological effects. PPAR-αcan inhibit the transcription and signal transduction of NF-κB (P65/ReLA subunit) and adjust the expression of inflammatory factors on cellular level, which might inhibit local inflammatory reaction. PPAR-αalso can control the expression of genes related with fatty acid oxidation in liver, which may play an important role in cytodifferentiation of adipose cells, storage and transport of lipids, fatty acid oxidation. Excitomotors of PPAR-αcan ameliorate fatty-degeneration of liver cells, reverse inflammatory reaction, and up-regulate the gene expression of fatty acid-binding proteins in liver. Studies hint that PPAR-αmight play a significant role in the pathologic mechanism of ALI, and might become one new therapeutic target for ALI.
     Hereto, there are less really therapeutical remedies for ALI yet. Chinese traditional medicine, with relatively less side effects, can regulate physiological reactions into balance by more targets. And it has a broad future of application and development in prevention and cure of ALI. Recently, there are numerous studies and reports on Chinese traditional medicine against ALI. The most are confined to observations of therapeutic effect, but less investigations on the mechanism of action. While, reports about studying effects of Chinese traditional medicine on expression of PPAR-αare rare. Jiugan Keli (JGKL) is a formula designed by Prof. Shi-zhong Yang on basis of his years of clinical experience. Prophase pharmacodynamics studies and clinical observations confirm that JGKL can alleviate hangover, decrease blood-fat level,protect liver function and lower activities of aminopherase efficiently. But its exact mechanism of action is still not clear now.
     In our study, we established ALI animal models successfully by lavage with distillate spirit. During the different stages of ALI (4w、8w、12w、16w), we observed the expression of PPAR-αand NF-κB in hepatic tissue of rats through molecular biology technique continuously. Besides, we also detected the level of TNF-α, IL-6 in serum, the content of MDA and the activity of SOD in hepatic tissue, ect. Then we analyzed the dependability between the expression of PPAR-αand other indexes, in order to identify the possible roles of PPAR-αin the development of ALI. Moreover, we treated ALI animals with JGKL and stop-drinking (spontaneous recovery) for 8w. Then we observed the expression of PPAR-αand NF-κB in hepatic tissue, the level of TNF-α, IL-6, FFA in serum, the content of MDA and the activity of SOD in hepatic tissue, ect. Moreover, In order to approach its mechanism of action against ALI further, we observed the effects of JGKL on the expression of PPAR-αand studied its mechanism of action initially. The following is the study outcomes:
     1) In the study, we established ALI animal models successfully by lavage with distillate spirit. A series of pathologic physiology variations were presented in hepatic tissue of rats with ALI, including adipose degeneration, inflammation, necrosis and slight fibrosis. And we found that the hepatic injury aggravated steadily with the increase of alcohol intake.
     2) During different stages of ALI, the gene expression of PPAR-αin hepatic tissue decreased obviously with the increase of alcohol intake.
     3) The expression of NF-κB in hepatic tissue of ALI rats increase steadily with the increase of alcohol intake.
     4) The level of TNF-α, IL-6, FFA in serum and the content of MDA in hepatic tissue of ALI increased with the increase of alcohol intake. While the activity of SOD in hepatic tissue decreased steadily with the increase of alcohol intake.
     5) Analysis of dependability indicates that the gene expression of PPAR-αwas correlated with the degree of liver injury, expression of inflammatory factors (NF-κB, TNF-α, IL-6) negatively. Meanwhile, there were certain inverse correlations between the gene expression of PPAR-αand the content of MDA, the level of FFA respectively. On the contrary, the gene expression of PPAR-αwas correlated with the activity of SOD in hepatic tissue positively.
     6) JGKL could against the further liver injury caused by alcohol efficiently. It might enhance the gene expression of PPAR-αand lower the expression of NF-κB, TNF-α, IL-6 in hepatic tissue. JGKL also could ameliorate the impaired hepatic function and lighten the oxidative stress. Besides, JGKL might protect liver cells and recover the impaired hepatic tissue.
     7) The impaired hepatic function, the level of blood-fat, the indexes of oxidation/anti-oxidation and histopathological changes might improve gradually after stop-drinking. The expression of inflammatory factors (NF-κB, TNF-α, IL-6) decreased obviously, while the expression of PPAR-αincreased gradually.
     8) Combining stop-drinking and JGKL could treat ALI more significantly, which might enhance the expression of PPAR-α, weaken the expression of inflammatory factors (NF-κB, TNF-α, IL-6) in hepatic tissue, improve the indexes of oxidation/anti-oxidation and others obviously.
     The study results suggested that:
     1) PPAR-αmight play a significant role in the nosogenesis of ALI. The decreased expression of PPAR-αcan enhance the expression of inflammatory factors, degrade the power of removing free-radical and aggravate the oxidative stress. Thus, to increase the expression of PPAR-αmight be anticipated to become one new therapeutic target for ALI.
     2) JGKL has good preventive and therapeutic effects on ALI. PPAR-αshould be a target of its effects, and to increase the expression of PPAR-αmight be one of main mechanism of action of JGKL.
     3) Stop-drinking (spontaneous recovery) is a chief method for curing ALI. While, we found that combining stop-drinking and JGKL might be more efficiently to treat ALI.
     Significance and innovation of the study:
     1) The study is the first of its kind using real-time PCR and RT-PCR to observe the expression of PPAR-αin the different stages of ALI. The study demonstrates that the gene expression of PPAR-αdecreases obviously during different stages of ALI with the increase of alcohol intake.
     2) The study is the first of its kind indicating through dependablity analysis that the gene expression of PPAR-αis correlated with the degree of liver injury, the expression of inflammatory factors (NF-κB, TNF-α, IL-6) negatively in ALI. Meanwhile, there are certain inverse correlations between the gene expression of PPAR-αand the indexes of oxidative stress (the content of MDA, the level of FFA) respectively. On the contrary, the gene expression of PPAR-αis correlated with the activity of SOD in hepatic tissue positively.
     3) The study is the first of its kind explaining mechanism of action agaist ALI of JGKL from gene and protein level. The study indicates that JGKL has good preventive and therapeutic effects on ALI. JGKL can enhance the expression of PPAR-α, which might decrease inflammatory reaction in hepatic tissue, improve the power of antioxygen and inhibit the lipid peroxidation efficiently.
引文
[1] 中华医学会肝脏病学分会脂肪肝和酒精性肝病学组. 酒精性肝病诊断标准[J]. 中华肝脏病杂志, 2003, 11(2):72.
    [2] 凌奇荷, 卿笃信. 重视对酒精性肝病的研究[J].中华消化杂志, 2001, 21(9):517-518.
    [3] Mann RE, Smart RG, Govoni R. The epidemiology of alcoholic liver disease[J]. Alcohol Res Health, 2003, 27(3):209-219.
    [4] Diehl AM. Liver disease in alcohol abusers: clinical perspective[J]. Alcohol, 2002, 27(1):721.
    [5] O’shea RS, McCullough AJ. Treatment of alcoholic hepatitis[J]. Clin Liver Dis, 2005, 9(1):103-134.
    [6] Neuberger J, Schulz KH, Day C, et al. Transplantation for alcoholic liver disease[J]. J Hepatol, 2002, 36(1):130-137.
    [7] 庄辉. 酒精性肝病的流行病学[M]. 见:江正辉, 王泰龄主编, 酒精性肝病. 北京:中国医药科技出版社, 2001:3-11.
    [8] 鲁晓岚, 陶明, 罗金燕, 等. 饮酒与肝病流行病学调查[J]. 中华肝脏病杂志, 2002, 10(6):467-468.
    [9] 鲁晓岚, 陶明, 罗金燕, 等. 西安酒精性肝病流行病学[J]. 世界华人消化杂志, 2003, 11(6):719-722.
    [10] 厉有名, 陈卫星, 虞朝辉, 等. 浙江省酒精性肝病流行病学调查概况[J]. 中华肝脏病杂志, 2003, 11(11):647-649.
    [11] 戴水奇, 黄顺玲, 张雪红, 等. 湖南省酒精性肝病流行病学分析[J]. 中西医结合肝病杂志, 2005, 15(2):105-106.
    [12] James M, Ryder S. Alcohol-related liver disease [J]. Practitioner, 2003, 247(1646):414.
    [13] 孙艳, 吴阳, 刘兵, 等. 酒精性肝病的研究进展[J]. 吉林大学学报 (医学版), 2006, 32(4):733-736.
    [14] 齐锦生, 赵京山, 李恩. 解酒醒神方药对酒精中毒小鼠模型作用的实验研究[J]. 中国中医基础医学杂志, 2001, 7(11):47-49.
    [15] 陈子謦, 王泰龄. 酒精性大鼠肝损伤中酶组织化学反应的观察[J]. 中日友好医院学报, 1996, 10(3):197-200.
    [16] 大西久仁房. 酒精与肝病[J]. 日本医学介绍, 1997, 18(1):6-7.
    [17] Tuma DJ, Casey CA. Dangerous byproducts of alcohol breakdown focus on adducts[J]. Alcohol Res Health, 2003, 27(4):285-290.
    [18] Niemela O, Parkila S, Hettuala SY, et al. Sequential acetaldehyde production, lipid peroxidation, and fibrogenesis in micropig model of alcohol-induced liver diease[J]. Hepatology, 1995, 22(4):1208-1214.
    [19] 陈曦, 牛俊奇, 王峰. 酒精性肝病患者血清抗乙醛蛋白加合物抗体滴度的检测[J]. 中华肝脏病杂志, 2002, 10(5):350-353.
    [20] Hoek JB, Pastorino JG. Ethanol oxidative stress and cytokine-induced liver cell injury [J]. Alcohol, 2002, 27(1):63-68.
    [21] Wu D, Cederbaum AI. Alcohol, oxidative stress, and free radical damage[J]. Alcohol Res Health, 2003, 27(4):277-284.
    [22] Jones BE, Liu H, Lo CR, et a1. Cytochrome P4502E1 expression induces hepatoeyte resistance to cell death from oxidative stress [J]. Antioxid Redox Signal, 2002,4(5):701.
    [23] Loguercio C, Federico A. Oxidative Stress in Viraland Alcoholic Hepatitis[J]. Free Radic Biol Med, 2003, 34(1):1.
    [24] 孙艳, 孟祥伟, 迟宝荣, 等. 酒精性肝病发病机制的研究进展[J]. 吉林医学, 2006, 27(3):230-231.
    [25] 王吉耀, 涂传涛. 酒精性肝病的发病机制[J]. 临床内科杂志, 2004, 21(2):73-74.
    [26] Lin SC, Chung CY, Chiang CL, et al. The influence of propolis ethanol extract on liver microsomal enzymes and glutathione after chronic alcohol administration[J]. Am J Chin Med, 1999, 27:83-93.
    [27] 吕学儒, 宋晓风, 王长庚. 醒酒冲剂作用的研究[J]. 职业与健康, 2002, 18(1):136 -137.
    [28] Itoga S, Nakai T. Mutaions in the exons and exon-intron junction regions of human cytochrome P4502E1 gene and alcoholism[J]. Alcohol Clin Exp Res, 2003, (Supp14):S13-S16.
    [29] Jerrells TR. Role of activated CD8+ T Cells in the initiation of hepatic damage[J]. Alcohol, 2002, 27(1):47-52.
    [30] Yin M, Bradford BU, Wheeler MD, et al. Reduced early alcohol-induced liver injury in CD14-deficient mice[J]. J Immunal, 2001, 166(7):4737-4742.
    [31] Kamimuea S, Tsukamoto H. Cytokine gene expression by Kupffer cells in the experimental alcoholic liver disease[J]. Hepatology, 1995, 22(4):1304-1309.
    [32] Grove J, Daly AK, Bassendine MF, et al. Interleukin-10 promoter region polymor- phisms and susceptibility to advanced alcoholic liver disease[J]. Gut, 2000, 46(4):540-545.
    [33] Colombat M, Charlotte F, Ratziu V. Portal lymphocytic infiltrate in alcoholic liver disease[J]. Hum Pathol, 2002, 23(12):1170.
    [34] 赵世义, 王永健, 石礼, 等. TNF-α, TGF-β, IL-8 在酒精性肝病中发病机制的相关研究[J]. 中华名医论坛, 2005, 17(3):11-12.
    [35] Keshavarzian A, Holmes EW, Patel M, et al. Leaky gut in alcoholic cirrhosis. A possible mechanism for alcoholic-induced liver damage [J]. Am J Gastroenterol, 1999, 94(1):200-207.
    [36] Mathurin P, Deng QC, Keshavarzian A, et al. Exacerbaction of alcoholic liver injury by enteral endotoxin in rats [J]. Hepatology, 2000, 32(5):1008-1017.
    [37] Wheeler MD. Endotoxin and Kupffer cell activation inalcoholic liver disease[J]. Alcohol Res Health, 2003, 27(4):300-306.
    [38] Neuman MG. Cytokines central factors in alcoholic liver disease [J]. Alcohol Res Health, 2003, 27 (4):307-316.
    [39] 王瑞科, 刘秀珍, 张谷运, 等. 酒精性肝病白细胞介素-6 的检测及意义[J]. 临床肝胆病杂志, 2005, 21(2):100-101.
    [40] Uesugi T, Froh M, Arteel GE, et al. Toll-like receptor-4 is involved in the mechanism of early alcohol-induced liver injury in mice[J]. Hepatology, 2001, 34(1):101-108.
    [41] Monzoni A, Masutti F, Saccoccio G, et al. Genetic determinants of ethanol-induced liver damage[J]. J Mol Med, 2001, 7(4):255-262.
    [42] Vidal F, Lorrnzo A, Auguet T, et al. Genetic polymorphisms of ADH2, ADH3, CYP4502E1 Dra-Ⅰand Pst-Ⅰand ALDH2 in Spanish men: lack of association with alcoholism and alcoholic liver disease[J]. J Hepatol, 2004, 41(5):744.
    [43] Bataller R, North KE, Brenner DA. Genetic polymorphisms and the prograssion of liver fibrosis: a critical appraisal[J]. Hepatology, 2003, 37(3):493-503.
    [44] 孟祥伟, 迟宝荣, 高沿航, 等. 细胞色素 P4502E1 基因多态性在酒精性肝病发病中的意义[J]. 吉林大学学报(医学版), 2003, 29(4):451.
    [45] Lieber CS. Hepatitis C and alcohol[J]. J Clin Gastroenterl, 2003, 36(2):100.
    [46] 陈建, 赵卫东, 王敏, 等. 乙型肝炎病毒对酒精性肝病易感性的影响[J]. 中华内科杂志, 2005, 44(8):611-612.
    [47] 陆伦根. 酒精性肝病与乙型和丙型病毒性肝炎[J]. 现代医药卫生, 2004, 20(6):393-394.
    [48] Rouault TA, Klausner RD. Iron-sulfur clusters as biosensors of oxidants and iron[J]. Trends Biochem Sci, 1996, 21(5):174-177.
    [49] Pietrangelo A. Iron-induced oxidant stress in alcoholic liver fibro- genesis[J]. Alcohol, 2003, 30(2):121-129.
    [50] Li H, Hou S, Wang W, et al. Invitro effects of metal ions on lipid peroxidation induced by alcohol in mice liver homogenate[J]. J Environ Biol, 2003, 24(4):423-428.
    [51] 孟文高. 酒精性肝病辨治述要[J]. 湖南中医杂志, 2004,20(4):53.
    [52] 赵益梅, 韩涛, 于学美. 试从血瘀论酒精性肝病[J]. 吉林中医药, 2004, 24(9):1-2.
    [53] 高学清, 王正君. 消脂益肝汤治疗酒精性脂肪肝的临床观察[J]. 湖北中医杂志, 2005, 27(5):38-39.
    [54] Walsh K, Alexander G. alcoholic liver disease[J]. Postgrad Med J, 2000, 76(895):280-286.
    [56] Campollo O, Martinez MD, Valencia JJ, et al. Drinking patterns and beverage preferences of liver cirrhosis patients in Mexico[J]. Subst Use Misuse, 2001, 36(3):387-398.
    [59] Gordon H. Detection of alcoholic liver disease[J]. World J Gastroentero1, 2001, 7(3):297-302.
    [57] 厉有名, 陈卫星, 虞朝晖, 等. 浙江省酒精性肝病流行病学调查概况[J]. 中华肝脏病杂志, 2003, 11(11):647-649.
    [58] 黎宏章, 林慧佳, 乐敏, 等. 嗜酒与肝功能损害危险因素相关性分析[J]. 浙江预防医学, 2005, 17(3):19.
    [59] 戴水奇, 黄顺玲, 张雪红, 等. 湖南省酒精性肝病流行病学分析[J]. 中西医结合肝病杂志, 2005, 15(2):105-106.
    [60] Becker U, Gronbaek M, Johansen D, et al. Lower risk for alcohol-induced cirrhosis in wine dringkers[J]. Hepatology, 2002, 35(4):868-875.
    [61] Tsukamoto H, Lin M, Ohata M, et al. Iron primes hepatic macrophages for NF-kappaB activation in alcoholic liver injury[J]. Am J Physiol, 1999,277 (6Pt 1):G1240-G1250.
    [62] Bunout D. Nutritional and metabolic effects of alcoholism: their relationship with alcoholic liver disease[J]. Nutrition, 1999, 15(7-8):583-589.
    [63] 鲁晓岚, 罗金燕, 陶明, 等. 酒精性肝病的危险因素分析[J]. 胃肠病学和肝病学杂志, 2004,13(2):2-5.
    [64] Isseinalln I. Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators[J]. Nature, 1900, 347:645.
    [65] Desvergne B, Wahli W. Peroxisome proliferator-activated receptors : nuclear control of metabolism[J]. Endocr Rev, 1999, 20(5):649-688.
    [66] Andersen O, Eijsink VG, Thomassen M. Multiple variants of the peroxisome proliferator activated receptor (PPAR)γ are expressed in the liver of Atlantic salmon (Salmosalar)[J]. Gene, 2000, 255:411-418.
    [67] Heinlein CA, Ting HJ, Yeh S, et al. Identification of ARA70 as a lingand enhanced coactivator for the peroxisome proliferator activated receptor gamma[J]. J Biol Chem, 1999, 274:16147-16152.
    [68] Zafrani ES. Non-alcoholic fatty liver disease, an emerging pathological spectrum[J]. Virchows Arch, 2004, 444(1):3-12.
    [69] Reddy JK. Ⅲ Nonalcoholic steatosis and steatohepatitis. Peroxisomal beta-oxidation,PPAR alpha,and steato-hepatitis[J].Am J Physiol Gastrointest Liver Physiol, 2001,28l(6):Gl333-G1339.
    [70] Hashimoto T, Cook WS, Qi C, et al. Defect in peroxisome proliferator-activated receptor alpha-inducible fatty acid oxidation determines the severity of hepatic steatosis in response to fasting[J].J Biol Chem, 2000, 275:28918-28928.
    [71] Ip E, Farrell GC, Robertso G, et al. Gentral role of PPAR alpha dependent hepatic lipid turnover in dietary steatohepatitis in mice[J]. Hepatology, 2003, 38(1):123-132.
    [72] Lee SS, Chan WY, Lo CK, et al. Requirement of PPAR alpha in maintaining phospholipid and triacylglycerol homeostasis during energy deprivation[J]. J Lipid Res, 2004, 45(11):2025-2037.
    [73] 戚晓红, 张昭萍, 李晓宇, 等. 过氧化物酶体增殖物激活受体在实验性大鼠脂肪肝中的表达[J]. 中国病理生理杂志, 2003, 19(9):1206-1209.
    [74] YE Ping,WANG Zhao-jun, et al.Age related decrease in expression of peroxisome proliferator activated receptor α and its effects on development of dyslipidemia[J]. Chinese Medical Journal, 2005, 118(13):1093-1098.
    [75] 石如玲, 姜玲玲. 胆汁酸对大鼠肝脏过氧化物酶体增殖剂激活受体-αmRNA 表达的影响[J]. 河北医科大学学报, 2005, 26(5):368-369.
    [76] Chitturi S, Farrell GC. Etiopathogenesis of nonalcoholic steato-hepatitis[J]. Semin Liver Dis, 2001, 21(1):27-41.
    [77] Nakajima T, Kamijo Y, Tanaka N, et al. Peroxisome proliferator activated receptor alpha protects against alcohol-induced liver damage[J]. Hepatology, 2004, 40(4):783-786.
    [78] Nanji AA, Dannenberg AJ, Jokelainen K, et al. Alcoholic liver injury in the rat is associated with reduced expression of Peroxisome proliferator alpha (PPAR alpha) regulated genes and is ameliorated by PPAR alpha activation[J]. J Pharmacol Exp Ther, 2004, 310(1):417-424.
    [79] Neuschwander-Tetri BA, Caldwell SH. Nonalcoholic steato-hepatitis: summary of an AASLD single topic conference[J]. Hepatology, 2003, 37(5):1202-1219.
    [80] Staels B, Koening W, Habib A, et al. Activation of human aortic smooth muscle cell is inhibited by PPAR-α but not by PPAR-γ activators[J]. Nature, 1998, 393(6687):790-793.
    [81] Delerive P, Gervios P, Fruchart JC, et al. Induction of kappa B-alpha expression as a mechanism contributing to the anti-inflammatory activities of peroxisome proliferator activated receptor-alpha activators[J]. J Biol Chem, 2000, 275(47):36703-36707.
    [82] Kon K, Ikejima K, Hirose M, et a1. Pioglitazone prevents early-phase hepatic fibrogenesis caused by carbon retrachloride[J]. Biochem Bilphys Res Commun, 2002, 29l(1):55-61.
    [83] Tang H, McLachlan A. Transcriptional regulation of hepatitis B virus by nuclear hormone receptors is a critical determinant of viral tropism[J]. Proc Natl Acad Sci USA, 2001, 98(4):l841-1846.
    [84] Guidotti LG, Eggers CM, Raney AK, et a1. In vivo regulation of hepatitis B virus replication by peroxisome proliferators[J]. J Virol, l999, 73:10377-l0386.
    [85] Christopher CJ, Paula JL, Frank JG. Central role of PPAR-αin the mechanism of action of hepatocarcinogenic peroxisome proliferators[J]. Mutation Research, 2000, 448(2):139-151.
    [86] Rusyn I, Rose ML, Bojes HK,et a1. Novel role of oxidants in the molecular mechanism of action of peroxisome proliferators[J]. Antixid Redox Signal, 2000, 2(3):607-611.
    [87] Miller RT, Scappina LA, Long SM, et al. Role of thyroid hormones in hepatic effects of peroxisome protiferators[J]. Toxicd Pathol, 2001, 29(1):149-155.
    [88] Paul A Grirnaldi.The roles of PPARs in adipocyte different- tiation[J]. Progress in Lipid Research, 2001, 40(4):269-281.
    [89] 李舒丹, 厉有名, 虞朝辉. 大鼠慢性酒精性肝损伤观察[J]. 中国公共卫生, 2004, 20(2):195-196.
    [90] 中华医学会肝病学分会脂肪肝和酒精性肝病学组. 酒精性肝病诊疗指南[J]. 中华肝脏病杂志, 2006, 14(3):164-166.
    [91] 晏春根, 周霞跃. 细胞因子与脂肪性肝炎[J].中华肝病杂志, 2001, 8(3):184-185.
    [92] 邢全台, 郝洪升. 慢性乙醇中毒大鼠组织内细胞因子的变化[J]. 中华肝病杂志, 2002, 10(6):463-464.
    [93] 李有田, 薛浩, 许丹, 等. 自拟解酒护肝饮治疗酒精性肝病 32 例[J]. 吉大学学报(医学版), 2004, 30(6):967-969.
    [94] 阎明, 孟繁立, 董春峡, 等. 非诺贝特和辛伐他汀对酒精性脂肪肝血清游离脂肪酸谱的影响[J]. 中国药理学通报, 2003, 19(11):1290-1294.
    [95] 赵冬梅, 刘耕陶. 肝细胞损伤机制进展的研究及其对研究治肝病新药的启示[J]. 中国药理学通报, 2001, 17(6):605-609.
    [96] 郑仲谨, 李磊. 核转录因子κB 在全身炎症反应综合征中的作用[J]. 免疫学杂志, 2001, 17(4):314-317.
    [97] Altavilla D, Saitta A, Squadrito G, et al. Evidence for a role of nuclear factor-kappa B in acute hypovolemic hemorrhagic shock[J]. Surgery, 2002, 131(1):50-58.
    [98] Altavilla D, Saitta A, Guarini S, et al. Oxidatice stress causes nuclear factor-kappa B activation in acute hypovolemic hemorrhagic shock[J]. Free Radic Biol Med, 2001, 30(10):1055-1066.
    [99] Tsukamoto H, Rippe R, Niemela O, et a1.Roles of oxidative in activation of Kupffer and Ito cells in liver fibrogenesis[J]. J Gstroenterol Hepatol, 1995, 10(suppl1):s50-s53.
    [100] Nanji AA, Jokelaine K, Rahemtulla A, et a1. Activation of nuclear factor kappa B and cytokine imbalance in experimental alcoholic liver disease in the rat[J], Hepatology, 1999, 30(4):934-943.
    [101] Kono H, Wheeler MD, Rusyn, et a1. Gender differences in early alchohol-induced liver injury:a role of CD14, NF-kappa B and TNF-alpha[J]. Am J Physiol Gastrointest Liver Physiol, 2000, 278(4):G652-G661.
    [102] Iimuro Y, Bradford BU, Yanashina S, et a1.The glutathion precursor L-2-oxothii- azolidine-4-carboxylic acid protects against liver injury due to chronic enteral exposure in the rat[J]. Hepatology, 2000, 31(2):391.
    [103] 赵彩彦. 核因子-κB 与酒精性肝病[J]. 国外医学, 消化系疾病分册, 2002, 22(4):230-232.
    [104] 刘铁军, 杨世忠, 邓厚波, 等. 酒肝颗粒治疗酒精性肝炎新药前期研究[J]. 长春中医学院学报, 2005,21(1): 48-50.
    [105] 张尚文.高热量饮食减少酒精性肝炎病死率[J].国外医学情报, 1994, 15(12):6.
    [106] Mezey E, Potter JJ, Rennie-Tankersley L, et al. A randomized placebo controlledtrial of vitamin E for alcoholic hepatitis[J]. J Hepatol, 2004, 40(1):40-46.
    [107] 于乐成, 顾长海. 水飞蓟宾素与酒精性肝病[J]. 肝脏, 2000, 5(3):182.
    [108] 邢全台, 袁孟彪, 等. 思美泰治疗酒精性肝病疗效观察[J]. 胃肠病学和肝病学杂志, 2002, 11(3):3.
    [109] 覃佳成. 阿拓莫兰治疗酒精性肝病疗效观察[J]. 广西医药杂志, 2004, 26(1):128.
    [110] Cao Q, Mak KM, Lieber CS. Dilinoleoylphosphatidylcholine decreases acetaldehyde induced TNF-alpha gene ration in Kupffercells of ethanol-fed rats[J]. Biochem Biophys Res Commun, 2002, 299(3):459-464.
    [111] 汤银红, 丁斌如. 枳椇子水提物对乙醇脱氢酶活性的影响[J]. 中药药理与临床. 2004, 20(2):24.
    [112] 嵇扬, 李俊, 杨平. 枳椇子对急性酒精中毒的作用[J]. 中药材, 2001, 24(2):126-128.
    [113] Hase K, Ohsugi M, Xiong Q, et al. Hepatoprotective effect of Hovenia dulcis THUNB. On experimental liver injuries induced by carbon tetrachloride or D- galactosamin/lipopolysaccharide[J]. Biol Pharm Bull, 1997, 20(4):381-387.
    [114] 林冬静, 金政. 决明子提取物对急性肝损伤保护作用的实验研究[J]. 时珍国医国药. 2006, 17(2):2-3.
    [115] 刘素丽, 冯志杰, 吕艳春, 等. 泽泻对肝硬化门脉高压血流动力学影响的临床研究[J]. 临床肝胆病杂志, 2006, 22(2):119-120.
    [116] 赖凌燕. 丹参对实验性酒精性肝损伤保护作用的初步研究[J]. 广西医科大学学报, 1999, 16(4):480-481.
    [117] 林秋实, 陈吉棣. 山楂及山楂黄酮预防大鼠脂质代谢紊乱的分子机制研究[J]. 营养学报, 2000, 22(2):131-136.
    [118] 黄智芬, 黎汉忠, 施智严, 等. 健脾消积Ⅱ服液结合西药治疗Ⅱ、Ⅲ期肝癌 40 例临床观察[J]. 中国中西医结合杂志, 2003, 23(3):193.

© 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