N-乙酰半胱氨酸对急性酒精性肝损伤大鼠保护作用的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的:观察N-乙酰半胱氨酸(N-acetylcysteine,NAC)对急性酒精性肝损伤大鼠的保护作用。
     方法:采用酒精灌胃法建立急性肝损伤大鼠模型。50只雄性Wistar大鼠随机分为5组(n=10):正常对照组、模型组、乙酰半胱氨酸低剂量组(150mg/kg)、中剂量组(300mg/kg)、高剂量组(600mg/kg)。正常对照组用等量的生理盐水代替酒精灌胃和腹腔注射;模型组以56°白酒按14ml/kg体重灌胃,每日一次,连续10d;各治疗组在酒精灌胃前30min分别腹腔注射不同剂量乙酰半胱氨酸,每日一次,连续10d。治疗结束后测定各组大鼠肝脏指数,留取腹主动脉血及肝脏标本,分离血清测丙氨酸氨基转移酶(ALT)、天门冬氨酸氨基转移酶(AST)活性及肿瘤坏死因子-α(TNF-α)的含量,制备肝组织匀浆测超氧化物歧化酶(SOD)、谷胱甘肽过氧化物酶(GSH-Px)活性与丙二醛(MDA)含量,肝组织行HE染色光镜下观察病理学改变,并行免疫组织化学染色检测肝脏中核转录因子-κB(NF-κB)与半胱氨酸蛋白酶-3(Caspase-3)的表达。
     结果:①与正常对照组比较,模型组大鼠肝脏指数显著增加,血清ALT、AST活性及TNF-α含量明显升高,肝组织MDA含量及NF-κB、Caspase-3的表达增强,SOD、GSH-Px活性减低(P<0.01);光镜下肝组织结构紊乱,肝细胞脂肪变性严重,伴炎症细胞浸润。②治疗结束后,乙酰半胱氨酸各剂量组ALT、AST活性均明显降低,肝脏指数减小,肝组织中MDA含量降低,SOD、GSH-Px活性明显升高,与模型组比较有统计学意义(P<0.01或P<0.05)。与低剂量组比较,中、高剂量组MDA含量降低,SOD、GSH-Px活性升高,(P<0.01或P<0.05)。与中剂量组比较,高剂量组SOD活性明显升高(P<0.01)。③光镜下各治疗组肝组织脂肪变性和炎症程度轻于模型组,高剂量组与正常对照组无明显差异,中剂量组轻微脂变,低剂量组脂变略重。④各治疗组肝组织中NF-κB和Caspase-3的表达均明显减少,血清TNF-α含量降低,与模型组比较差别有统计学意义(P<0.01或P<0.05)。与低剂量组比较,中、高剂量组NF-κB和Caspase-3的表达减少,高剂量组TNF-α含量降低(P<0.01或P<0.05)。与中剂量组比较,高剂量组NF-κB和Caspase-3的表达减少,TNF-α含量降低(P<0.01或P<0.05)。
     结论:乙酰半胱氨酸不仅可以减少脂质过氧化产物MDA的含量,而且能提高肝组织中抗氧化酶SOD、GSH-Px活性,发挥抗氧化作用,进而改善肝功能,减轻肝细胞的脂肪堆积。同时还可以通过抑制NF-κB的激活,减少炎症因子的释放,抑制凋亡信号的传导,发挥抗炎及抗凋亡作用,从而有效防治酒精对肝脏的损伤,且其治疗作用具有一定的量效关系。
Objective:To investigate the protective effect of N-acetylcysteine(NAC) on acute alcoholic liver injury in rats.
     Methods:The liver injury rat models were induced by infusing alcohol into stomach of rats. Fifty male Wistar rats were randomly divided into 5 groups(n=10):control group, acute liver injury model group, and low-(150mg/kg), middle-(300mg/kg), high-(600mg/kg) dose group of N-acetylcysteine. The control group were given the same amount of salime instead of alcohol through intraperitpneal injection and gastric infusion. The model group were infused stomach with 56°liqueur once a day for 10d. Different doses of N-acetylcysteine groups were injected NAC intraperitoneally at 30 min before infusing liqueur once a day for 10d. After treatment, the liver indices were measured and aorta blood and liver tissue were collected. Serum was isolated for measurement of alanine transaminase(ALT), aspartate transaminase(AST) activity and tumor necrosis factor-a(TNF-a) content. Liver tissue homogenate was prepared to assay superoxide dismutase(SOD), glutathione perioxidase(GSH-Px) activities and malondialdehyde (MDA) content. The liver histopathologies were explored with HE staining. The expression of nuclear factor-kappa B(NF-κB) and Caspase-3 in liver were detected by immunohistochemistry.
     Results:①Compared with control group, liver indices, serum ALT, AST level and TNF-αcontent increased, liver tissue MDA content and the expression of NF-κB and Caspase-3 increased, SOD, GSH-Px activities decreased(P<0.01) significantly in model group. Under light microscope, the structure of the liver tissue was disorded, with a serious liver steatosis, accompanied by the infiltration of inflammatory cells.②After treatment, serum ALT, AST level and liver indices decreased, and liver tissues MDA content decreased, SOD, GSH-Px activities increased in difference doses of NAC groups(P<0.01 or P<0.05), the difference was significant compared with model group. Compared with low-dose group, MDA content decreased, SOD, GSH-Px activity increased in middle and high-dose groups(P<0.01 or P<0.05).Compared with the middle dose group, SOD activity was significantly higher in high-dose group(P<0.01).③Under light microscope, the treatment groups were lighter in liver steatosis and the degree of inflammation than the model group. High-dose group and control group were not significantly different. The middle-dose group had a slight steatosis and the low-dose group have a more serious steatosis.④In three different doses of NAC groups, the expression of NF-κB and Caspase-3 were significantly reduced, serum TNF-a content were decreased(P<0.01 or P<0.05). Compared with model group, the difference was significant. Compared with low-dose group, the expression of NF-κB and Caspase-3 reduced in middle and high-dose group, serum TNF-a decreased in the high-dose group(P<0.01 or P<0.05). Compared with the middle-dose group, the expression of NF-κB and Caspase-3 were reduced, serumTNF-a content decreased in the high-dose group(P<0.01 or P<0.05).
     Conclusions:The acute alcoholic liver injury models in rats were made successfully. N-acetylcysteine can not only reduce the lipid peroxidation product MDA content, but also improve the liver tissue antioxidant enzymes SOD, GSH-Px activity, to improve liver function, reduce the accumulation of lipid in liver cells. Also, it can reduce the release of inflammatory cytokines by inhibiting NF-κB activating, and inhibit apoptosis signal transduction, having anti-inflammatory and anti-apoptotic effects. Thus NAC might have effect of alleviating the liver injury induced by alcohol and its therapeutic effect has a certain dose-effect relationship.
引文
[1]苏俊,廖彩仙,周杰,等.酒精性肝病105例临床分析.实用医学杂志,2007,23(5):704-705.
    [2]刘克洲.第四讲酒精性肝病发病机制若干进展.中华消化杂志,2002,22(4):230-232.
    [3]夏丽娟,陈飞虎.核因子-κB的分子生物学特性及其在炎症反应中的作用.安徽医药,2005,9(3):162-164.
    [4]Song M, Kellum JA, Kaldas H, et al. Evidence that glutathione depletion is a mechanism responsible for the anti-inflammatory effects of ethyl pyruvate in cultured lipopolysaccharide-stimulated RAW 264.7 cells. J Pharmacol Exp Ther,2004,308(1):307-316.
    [5]Patriarca S, Furfaro AL, Domenicotti C, et al. Supplementation with N-acetylcysteine and taurine failed to restore glutathione content in liver of streptozotocin-induced diabetics rats but protected from oxidative stress. Biochim Biophys Acta,2005,1741(1-2):48-54.
    [6]Harrison P, Wendon J, Williams R,et al. Evidence of increased guanylate cyclase activation by acetylcysteine in fulminant hepatic failure. Hepatology,1996,23(5):1067-1072.
    [7]Paterson RL, Galley HF, Webster NR. The effect of N-acetylcysteine on nuclear factor-kappa B activation, interleukin-6, interleukin-8 and intercellular adhesion molecule-1 expression in patients with sepsis. Crit Care Med,2003,31(11):2574-2578.
    [8]孙俊,王玉霞,孙曼雯.乙酰半胱氨酸及还原型谷胱甘肽对抗硫芥引起的细胞凋亡及坏死.中国药理与毒理学杂志,2000,14(1):26-30.
    [9]吴刚,郭树华.N-乙酰半胱氨酸在肝病治疗中的应用.中华肝脏病杂志,2004,12(3):185-186.
    [10]李毅敏,赵树仪.N-乙酰半胱氨酸的研究进展.天津药学.2003,15(2):50-53.
    [11]王福根,俞佳.席建军乙酰半胱氨酸对大鼠酒精性肝损伤的保护作用.中国临床药理学与治疗学,2008,13(5):517-520.
    [12]Cutrin C, Menino MJ, Otero X, et al. Effect of nifedipine and S-Adneosylmethionine in the liver of rats treated with CCl4 and ehtanol for one month. Life Sci,1992,51(10):113-118.
    [13]Harrison DJ, Burt AD. Pathology of alcoholic liver disease. Baillieres Clin Gastroenterol.1993,7(3):641-662.
    [14]Lieber CS, Decarli LM, The feeding of ehtnaol in liquid diets:1986 update. Alcohol Clin Exp Res,1986, 10(5):550-553.
    [15]Tsukamoto H, Reideblerger RD, French SW, et al. Long-term cannulation model for blood sampling and intragastric infusion in the rat. Am J Physiol,1984,247(3):595-599.
    [16]赵静波,王泰玲,张晶,等.大鼠急性酒精性肝损伤模型分析.中日友好医院学报,1996,10(1):17-19.
    [17]童英,姚小曼,吴少平.乙醇诱发急性肝损伤生物标记物的探讨.中国食品卫生杂志,1999,11:12-14.
    [18]厉有名.酒精性肝病的发病机制.中华肝脏病杂志,2003,11(11):690-691.
    [19]Wu D, Cederbaum AI. Oxidative stress and alcoholic liver disease. Semin Liver Dis,2009,29(2):141-154.
    [20]Britton RS, Bacon BR. Role of free radicals in liver diseases and hepatic fibrosis. Hepatogastroenterology, 1994,41:343-348.
    [21]Tsukamoto H.Oxidative stress, antioxidants, and alcoholic liver fibrogenesis. Alcohol,1993,10(6):465-467.
    [22]Grove J, Daly AK, Bassendine MF, et al. Association of a tumor necrosis factor promoter polymorphism with susceptibility to alcoholic steatohepatitis. Hepatology,1997,26(1):143-146.
    [23]Tomita K, Azuma T, Kitamura N, et al. Pioglitazone prevents alcohol-induced fatty liver in rats through up-regulation of c-Met. Gastroenterology,2004,126:873-885.
    [24]陈韶华,厉有名.酒精性肝病肝细胞凋亡与HSC凋亡.国外医学·消化系疾病分册,2003,23(1):40-43.
    [25]崔巍,傅宝玉.实验性酒精性肝病大鼠中脂质过氧化产物的动态变化.辽宁医学杂志,2001,15(3):131-132.
    [26]Wu D, Cederbaum AI. Alcohol, oxidative srtess, and free radical damage. Alcohol Res Health,2003, 27(4):277-284.
    [27]裴凌鹏.顺式白藜芦醇对小鼠急性酒精肝损伤的预防研究.上海中医药杂志,2008,42(9):67-69.
    [28]王春妍,范玉强,迟宝荣,等.核因子-κB及其下游因子TNF-α、Bcl-2在急性肝损伤中的作用及机制.世界华人消化杂志,2008,16(25):2804-2808.
    [29]Watanabe T, Jono H, Han JH, et al. Synergistic activation of NF-kappa B by nontypeable Haemophilus influenzae and tumor necrosis factor-a. PNAS,2004,101(10):3563-3568.
    [30]莫成林,李燕.细胞因子在酒精性肝炎中的作用.国外医学·药学分册,2003,30(3):171-175.
    [31]颜天华,袁伯俊.肿瘤坏死因子肝损伤机理研究进展.国外医学·药学分册,1998,25(6):330-334.
    [32]Zhang Y, Goodyer C, Le Blanc A. Selective and protracted apoptosis in human primary neurons microinjection with active caspase-3,-6,-7and-8. J Neurosci,2000,20(22):8384-8389.
    [33]Yin XM, Ding WX. Death receptor activation-induced hepatocyte apoptosis and liver injury. Curr Mol Med.2003,3(6):491-508.
    [34]Mathurin P, Mendenhall CL, Carithers RL Jr, et al. Corticosteroids improve short-term survival in patients with severe alcoholic hepatitis (AH):individual data analysis of the last three randomized placebo controlled double blind trials of corticosteroids in severe AH. J Hepatol,2002,36(4):480-487.
    [35]Arteel GE. Oxidants and antioxidants in alcohol-induced liver disease. Gastroenterology,2003,124(3): 778-790.
    [36]Kelly GS. Clinical applications of N-acetylcysteine. Alten Med Rev,1998,3(2):114-127.
    [37]Fusai G, Glantzounis GK, Hafez T, et al. N-acetylcysteine ameliorates the late phase of liver ischaemia/reperfusion injury in the rabbit with hepatic steatosis. Clin Sci,2005,109(5):465-473.
    [38]De Flora S, Izzotti A, D'Agostini F, et al. Mechanisms of N-acetylcysteine in the prevention of DNA
    [39]Ceconi C, Currello S, Cargnoni A, et al. The role of glutathione status in the protection against. ischemic and reperfusion damage:effects of N-acetylcesteine. J Mol Cell Cardiol,1988,20(1):5-13.
    [40]江正辉,王泰龄.酒精性肝病.中国医药科技出版社,2001:57.
    [41]李旭,孟莹,杨希山,等.N-乙酰半胱氨酸对肝星状细胞核因子KB的影响.中华消化杂志,2005,25(2):83-86.
    [42]Pajonk F, Riess K, Sommer A, et al. N-acety-L-cysteine inhibits 26s proteasome function:implications for effects on NF-kappaB activation. Free Radic Biol Med,2002,32(6):536-543.
    [43]Oka S, Kamata K, Yagisawa H, et al. N-acetylcysteine suppresses TNF-induced NF-kappa B activation through inhibition of 1 kappa B kinases. FEBS Lett,2000,472(2-3):196-202.
    [44]Vendemiale G, Grattagliano I, Caruso ML, et al. Increased oxidative stress in dimethylnitrosamine-induced liver fibrosis in the rat:effect of N-acetylcysteine and interferon-alpha. Toxicol Appl Pharmacol,2001, 175(2):130-139.
    [45]Smikstein MJ, Knapp GL, Kulig KW, et al. Efficacy of oral N-acetylcysteine in the treatment of acetaminophen overdose. J Med,1988,319(24):1557-1562.
    [46]Rust C, Gores GJ, Apoptosis and liver disease. Am Med,2000,108:567-574.
    [47]黄瑞健,孙培吾,林乌拉,等.生长激素对大鼠心肌缺血/再灌注后心肌细胞凋亡及凋亡相关基因NF-κB蛋白表达的影响.中国急救医学,2004,24(6):420-422.
    [1]苏俊,廖彩仙,周杰,等.酒精性肝病105例临床分析.实用医学杂志,2007,23(5):704-705.
    [2]厉有名.酒精性肝病的发病机制.中华肝脏病杂志,2003,11(11):690-691.
    [3]Lim SP, Batta K, Tan BB. Calciphylaxis in a patient with alcoholic liver disease in the absence of renal failure. Clin Exp Dermatol,2003,28(1):341.
    [4]Preedy VR, Reilly ME, Patel VB, et al. Protein metabolism in alcoholism:effects on specific tissues and the whole body. Nutrition,1999,15(7-8):604-608.
    [5]Porta EA. Dietary modulation of oxidative stress in alcoholic liver disease in rats. J Nutr,1997,127(5): 912-915.
    [6]Eaton S, Record CO, Bartlett K. Multiple biochemical effects in the pathogenesis of alcoholic fatty liver. Eur J Clin Invest,1997,27(9):719-722.
    [7]丁哗,高志安.酒精性肝病的研究进展.锦州医学院学报.2006,27(2):48-49.
    [8]卿笃信,凌奇荷.酒精代谢酶与酒精性肝病的关系研究进展.国外医学·生理、病理科学与临床分册.2003,23(3):310-313.
    [9]Oneta C, Simanowski U, Martinez M, et al. First pass metabolism of ethanol is strikingly influenced by speed of gastric emptying. Gut,1998,43(5):612-619.
    [10]陈曦,吕文芝,牛俊奇,等.乙醛蛋白加合物在酒精性肝病中的致病作用.临床肝胆病杂志.2003,19(5):268-270.
    [11]Robertson G, Leclercq I, Farrell GC. Nonalcoholic steatosis and steatohepatitis. Ⅱ. Cytochrome P-450 enzymes and oxidative stress. Am J Physiol Gastrointest Liver Physiol,2001,281(5):1135-1139.
    [12]曾民德.脂肪肝发病机制“二次打击”的假设.肝脏,2001,6:145.
    [13]刘克洲.酒精性肝病发病机制若干进展.中华消化杂志,2002,22(4):230-232.
    [14]周俊英,姚树坤.NADPH氧化酶在酒精性肝病发病机制中的作用.中华肝脏病杂志,2005,13(8):633-635
    [15]白晓斌,徐晓云,刘浩.铁在酒精性肝病中的作用.国外医学·医学地理分册,2005,26(3),125-127,142.
    [16]Kishore R, Hill JR, McMullen MR, et al. ERK1/2 and Egr-1 contribute to increased TNF-alpha production in rat Kupffer cells after chronic ethanol feeding. Am J Physiol Gastrointest Liver Physiol, 2002,282(1):6-15.
    [17]莫成林,李燕.细胞因子在酒精性肝炎中的作用.国外医学·药学分册,2003,30(3):171-174.
    [18]Ohkubo K, Masumoto T, Horiike N, et al. Induction of CINC (interleukin-8) production in rat liver by non-parenchymal cells. J Gastroenterol Hepatol,1998,13(7):696-702.
    [19]O'Neill LA, Bowie AG. The family of five:TIR-domain-containing adaptors in Toll-like receptor signalling. Nat Rev Immunol,2007,7:353-364.
    [20]Takeda K, Akira S. TLR signaling pathways. Semin Immunol.2004,16(1):3-9.
    [21]Medvedev AE, Sabroe I, Hasday JD, et al. Tolerance to microbial TLR ligands:molecular mechanisms and relevance to disease. J Endotoxin Res,2006,12(3):133-150.
    [22]Yee SB, Hanumegowda UM, Hotchkiss JA, et al. Role of neutrophils in the synergistic liver injury from monocrotaline and bacterial lipopolysaccharide exposure. Toxicol Sci,2003,7(2):43-56.
    [23]Yee SB, Harkema JR, Ganey PE, Roth RA. The coagulation system contributes to synergistic liver injury from exposure to monocrotaline and bacterial lipopolysaccharide.Toxicol Sci,2003,74:457-469.
    [24]Hirose M, Nishikawa M, Qian W, et al. Mannose-conjugated alendronate selectively depletes Kupffer cells and inhibits endotoxemic shock in the mice.Hepatol Res,2006,36(1):3-10.
    [25]Scott MJ, Liu S, Su GL, et al. Hepatocytes enhance effects of lipopolysaccharide on liver nonparenchymal cells through close cell interactions. Shock,2005,23(5):453-458
    [26]Higuchi Y, Kawakami S, Yamashita F, et al. The potential role of fucosylated cationic liposome/NF kappa B decoy complexes in the treatment of cytokine-related liver disease. Biomaterials 2007,28(26): 532-539.
    [27]Nanji AA. Apoptosis and alcoholic liver disease. Semin Liver Dis 1998,18(2):187-190.
    [28]Neuman MG, Shear NH, Bellentani S, et al. Role of cytokines in ethanol-induced cytotoxicity in vitro in Hep G2 cells. Gastroenterology,1998,115(1):157-166.
    [29]Naveau S, Emilie D, Balian A, et al. Plasma levels of soluble tumor necrosis factor receptors p55 and p75 in patients with alcoholic liver disease of increasing severity. J Hepatol 1998,28(5):778-784.
    [30]Ziol M, Tepper M, Lohez M, et al. Clinical and biological relevance of hepatocyte apoptosis in alcoholic hepatitis. J Hepatol,2001,34(2):254-260.
    [31]Deaciuc IV, Fortunato F, D'Souza NB, et al. Modulation of caspase-3 activity and Fas ligand mRNA expression in rat liver cells in vivo by alcohol and lipopolysaccharide. Alcohol Clin Exp Res,1999,23(2): 349-356.
    [32]Cederbaum AI. Ethanol-related cytotoxicity catalyzed by CYP2E1-dependent generation of reactive oxygen intermediates in transduced HepG2 cells. BioFactors,1998,8(1-2):93-96.
    [33]Chandel NS, Schumacker PT, Arch RH. Reactive oxygen species are downstream produets of TRAF-mediated signal transduction. J Biol Chem 2001,276(46):42728-42736.
    [34]Minana JB, Gomez-Cambronero L, Lloret A, et al. Mitochondrial oxidative stress and CD95 ligand:a dual mechanism for hepatocyte apoptosis in chronic alcoholism. Hepatology,2002,35(5):1205-1214.
    [35]Rust C, Gores GJ. Apoptosis and liver disease. Am J Med,2000,108(7):567-574.
    [36]刘厚钰.我国酒精性肝病研究现状与展望.中华消化杂志,2002,22(7):389-390.

© 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