高迁移率族蛋白B1在肝脏慢性炎症与纤维化中的作用
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
研究背景:
     肝纤维化(Hepaticfibrosis)这一病理过程是各种慢性肝病进展为肝硬化的共同途径。肝纤维化最终发展为肝硬化和肝癌的比率甚高,高达25%~40%,而肝硬化的并发症和肝癌的死亡率是严重危害人们健康的世界性问题。肝纤维化是一个具有可逆性的病理过程,因此,阻止肝纤维化的进展是改善肝病预后的重要策略。目前研究认为,慢性炎症反应是肝纤维化发生发展的重要因素。近期的研究表明,高迁移率族蛋白B1(Highmobilitygroupproteinbox1,HMGB1)作为典型的内源性危险相关分子(Danger-associatedmolecularpatterns,DAMPs),在肝损伤引发的炎症反应中起着重要的作用。针对HMGB1在肝纤维化中作用的研究,引发了学者们极大的兴趣。因此,我们旨在观察HMGB1对肝脏慢性炎症反应和纤维化反应的作用。
     目的:
     通过观察高迁移率组蛋白1(Highmobilitygroupboxprotein1,HMGB1)对肝纤维化小鼠的相关炎症因子和纤维化因子的影响,探讨HMGB1在肝脏慢性炎症和纤维化中的作用。
     方法:
     第一部分实验:昆明雄性小鼠40只,采用CCL-4灌胃法制备肝纤维化模型,随机分为正常对照组(1ul/g生理盐水灌胃,每5天一次)、2周模型组、4周模型组、6周模型组(1ul/gCCL4油溶液灌胃,每5天一次)。各组动物处理前12小时禁食,摘除眼球法取血,ELISA法检测血清中透明质酸(HA)、Ⅰ型胶原蛋白(Col-Ⅰ)、Ⅲ型前胶原蛋白(PⅢP)和肿瘤坏死因子α(TNF-α)、白介素6(IL-6)的含量;分别对肝组织病理切片进行HE染色和天狼猩红染色;Westernblot法检测肝组织HMGB1的表达水平。
     第二部分实验:昆明雄性小鼠20只,采用CCL4灌胃法制备肝纤维化模型,随机分为模型组(1ul/gCCL4油溶液灌胃,每5天一次)、干预组(1ul/gCCL4油溶液灌胃,50ug/mouseHMGB1-Ab和5ug/g丙酮酸乙酯腹腔注射,每5天一次)。各组动物处理前12小时禁食,摘除眼球法取血,ELISA法检测血清中HA、Col-Ⅰ、PⅢP和IL-6、TNF-α的含量;分别对肝组织病理切片进行HE染色和天狼猩红染色;半定量RT-PCR法检测小鼠肝组织中TGF-β、α-SMA和MMP-9mRNA的表达。
     结果:
     1、建立小鼠肝纤维化模型。6周模型组病理切片显示肝细胞变性坏死比较严重,伴有炎症细胞浸润,并出现肝纤维组织增生;ELISA法检测血清胶原蛋白含量结果显示,正常对照组血清中HA、Col-Ⅰ、PⅢP含量较少;2周、4周、6周模型组血清中HA、Col-Ⅰ、PⅢP含量均较正常对照组明显增加(P<0.05);6周模型组血清中HA、Col-Ⅰ、PⅢP含量均较4周模型组明显增加(P<0.05)。
     2、CCL4致肝纤维化模型IL-6、TNF-α和HMGB1的变化。ELISA法检测正常对照组血清中IL-6、TNF-α含量较少;2周、4周、6周模型组血清中IL-6、TNF-α含量均较对照组明显增加(P<0.05);6周组血清中IL-6、TNF-α含量均较4周组明显增加(P<0.05)。Western-blot结果表明正常对照组肝组织内无HMGB1表达,肝纤维化过程中肝组织HMGB1的表达随着CCL4损伤周期的延长而逐渐升高,与正常对照组比较,2周、4周、6周模型组的HMGB1表达量明显增加,有统计学意义(P<0.05);与4周模型组比较,6周HMGB1的表达量显著增加(P<0.05)。
     3、拮抗HMGB1可降低血清中IL-6、TNF-α的含量。ELISA法检测各组动物血清中炎症因子含量结果显示,与模型组比较,干预组动物血清中IL-6、TNF-α的含量明显减少(P<0.05)。
     4、拮抗HMGB1可降低肝纤维化程度。HE染色显示,模型组肝细胞弥漫性脂肪变性,中央静脉周围肝细胞灶状坏死,炎症细胞浸润,汇管区周围纤维化,纤维间隔形成。干预组肝细胞少量的脂肪变性,炎症细胞浸润减少,汇管区有纤维产生,但无间隔形成。天狼猩红染色结果显示模型组肝组织内胶原纤维向肝小叶延伸,并形成纤维间隔,与模型组相比较,干预组肝组织内胶原纤维显著下降(0.57%±0.21%vs1.02%±0.33%;P<0.05)。ELISA法检测各组动物血清中胶原蛋白含量结果显示,与模型组比较,干预组动物血清中HA、Col-Ⅰ、PⅢP的含量明显减少(P<0.05)。RT-PCR结果显示与模型组小鼠肝脏组织TGF-β、α-SMA的表达量相比较,干预组小鼠肝脏组织这些因子的表达量均明显降低(P<0.05),同时MMP-9mRNA的表达量增高。
     结论:1.CCl4致肝纤维化动物模型中,HMGB1和相关炎症因子IL-6、TNF-α的表达增高.2.HMGB1的促纤维化作用与促炎作用有关。拮抗HMGB1后,相关炎症因子的浓度降低,致纤维化因子减少,肝纤维化程度减轻。
Background:
     Hepatic fibrosis is the common pathological process of a variety of chronic liver disease to cirrhosis, and is an important part of influencing the prognosis of chronic liver disease. The rate of liver fibrosis to cirrhosis and liver cancer is very high(25%~40%), and these diseases are becoming serious threats to people's health worldwide. Hepatic fibrosis is a reversible pathological process, so holding back the progress of hepatic fibrosis is an important strategy to improve the liver disease prognosis. Recognized chronic inflammatory response is an important factor in hepatic fibrosis. Recent studies show that the High mobility group protein box1(HMGB1), as a typical endogenous Danger-associated molecular patterns(DAMPs), plays an important role in the inflammatory response of the liver damage. The study of the role of HMGB1in liver fibrosis, led to great interest of scholars. Therefore, we aim to observe the role of HMGB1in chronic hepatic inflammatory response and fibrosis reaction.
     Objective:
     To investigate the role of High Mobility Group Box Protein1in the chronic hepatic inflammation and fibrosis reaction through observe the expression of inflammatory cytokines and fibrosis factor.
     Methods:
     The first part of the experiment:Forty male Kunming mice, the ccl4gavage preparation of liver fibrosis were randomly divided into three groups:the normal control group,1ul/g saline was administered every five days;2weeks model group,4weeks model group,6weeks model group, ccl4(1ul/g,diluted with corn oil) was administered every five days. Each mice was sacrificed by removal of the eye method. The parameters of plasma'HA, Col-Ⅰ, PⅢP and IL-6, TNF-α were measured by ELISA. The morphological changes of the liver tissue were observed with HE staining and Sirius red staining. The level of HMGB1in liver tissue were measured by western blot.
     The second part of the experiment:Twenty male Kunming mice,the ccl4gavage preparation of liver fibrosis were randomly divided into two groups:the model group, ccl4(1ul/g,diluted with corn oil) was administered every five days; the intervention group, ccl4(1ul/g, diluted with corn oil),50ug/mouse HMGB1-Ab,5ug/g ethyl pyruvate were administered by gavage or intraperitoneal injection every five days. Each mice was sacrificed by removal of the eye method. The parameters of plasma'HA, Col-Ⅰ, PⅢP and IL-6, TNF-α were measured by ELISA. The morphological changes of the liver tissue were observed with HE staining and Sirius red staining. The level of HMGB1in liver tissue were measured by western blot. The level of TGF-β,α-SMA和MMP-9mRNA were detected by RT-PCR.
     Results:
     1. Successfully established liver fibrosis model. In6weeks model group, liver cells degenerate and necrosis, inflammatory cells infiltrate and liver fibrous tissue proliferate. The levels of HA, Col-Ⅰ, PⅢP in the normal control group was very low in serum measured by ELISA. Compared with the normal control group, the levels of them in2weeks model group,4weeks model group and6weeks model group were remarkably increased (P<0.05), meanwhile, they in6weeks group were significantly higher than4weeks group(P<0.05).
     2. The changes of IL-6, TNF-α and HMGB1in ccl4induced liver fibrosis model. The levels of IL-6, TNF-α in the normal control group was very low in serum measured by ELISA. Compared with the normal control group, the levels of them in4weeks model group and6weeks model group were remarkably increased (P<0.05), meanwhile, they in6weeks group were significantly higher than4weeks group(P<0.05). Wester-blot results show that no HMGB1expression in the liver tissue of normal control group, and in the process of liver fibrosis, the higher HMGB1is expressed in liver tissue,the longer ccl4is administered. Compared with the normal control group, the levels of HMGB1in4weeks model group and6weeks model group were significantly increased (P<0.05), meanwhile, they in6weeks group were much higher than4weeks group(P<0.05).
     3. Antagonistic HMGB1can reduce the degree of liver fibrosis. In model group, liver cells steatosis and necrosis, inflammation cells infiltrate, liver fibrous tissue proliferate. Liver cells of intervention group have a small amount of steatosis,inflammatory cells infiltration,but no liver fibrosis formation in tissue. Antagonistic HMGB1can cause the marked decrease of liver fibrosis tissue by Sirius red staining (0.57%±0.21%vs1.02%±0.33%; P<0.05) and significantly reversed liver fibrosis. Compared with model group, the levels of HA, Col-Ⅰ, PⅢP in intervention group were remarkably decreased (P<0.05). Compared with model group, The mRNA expression of TGF-β, α-SMA in intervention group were significantly downregulated (P<0.05), however, the mRNA expression of MMP-9was upregulated(P<0.05).
     4. Antagonistic HMGB1can reduce the content of IL-6, TNF-α. Compared with model group, the levels of IL-6, TNF-α in intervention group were remarkably decreased (P<0.05).
     Conclusion:
     1. In the CC14induced liver fibrosis model, the levels of HMGB1and inflammatory factors IL-6,TNF-a are significantly increased.
     2. There are relationship between fibrogenic role and proinflammatory role in HMGB1. HMGB1antibody can reduce the lever of the inflammatory factors and fibro-genic cytokines in mice with liver fibrosis.
引文
[1]R. Bataller and D. A. Brenner. Liver fi brosis. Journal of Clinical Investigation [J].2005,115(2):209-218[2]Michal Cohen-Naftaly, Scott L. Friedman. Current status of novel antifibrotic therapies in patients with chronic liver disease Therapeutic Advances in Gastroenterology[J].2011,4(6):391-417[3]Novo E,Marra F, Zamara E. Overexp ression of Bc122by activatedhuman hepatic stellatecells:resistance to apoptosis as amechanismof p rogressive hepatic fibrogenesis in humans[J]. Gut,2006,55(8):1174-1182[4]S. L. Friedman. Mechanisms of hepatic fi brogenesis. Gas-troenterology [J].2008,134(6):1655-1669[5]Albanis E, Friedman SL. Hepatic fibrosis Pathogenesis and princinples of therapy[J]. Clin Liver Dis,2001,5(2):315-334[6]Kaczynski J, Cook T, Urrutia R. Spl-and Kriappel-like transcription factors. Genome Biol,2003,4(2):206[7]Yang H, Tracey KJ. Targeting HMGB1in inflammation[J]. Biochim Biophys Acta,2010,1799(1-2):49-156[8]Ajit Varki. Since there are PAMPs and DAMPs, there must be SAMPs? Glycan " self-associated molecular patterns " dampen innate immunity, but pathogens can mimic them. Glycobiology [J].2011,21(9):1121-1124[9]Alessandra Castiglioni, Valentina Canti, Patrizia Rovere-Querini, et al. High-mobility group box1(HMGB1) as a master regulator of innate immunity Cell[J]. Tissue Res.2011343:189-199[10]Goodwin GH, Sanders C, Johns EW. A new group of chromatin-associated proteins with a high content of acidic and basic amino acids[J]. Eur. J. Biochem.1973.38:14-19.[11]Bustin M. Regulation of DNA-dependent activities by the functional motifs of the high-mobility-group chromosomal proteins[J]. Mol. Cell Biol.1999.19:5237-5246.[12]Agresti A, Bianchi M E. HMGB proteins and gene expression [J]. Curr. Opin. Genet. Dev.2003.13:170-178.[13]Zhao ZF, Han DW, Liu MS, et al. Expression of HMGB-1and its extracellular release of cultured primary hepatic parenchymal cells and Kupffer cells induced by LPS.中华肝脏病杂志[J].2007,15(9):676-680[14]Allan Tsung, Rohit Sahai, Hiroyuki Tanaka, et al. The nuclear factor HMGB1mediates hepatic injury after murine liver ischemia-reperfusion. Journal of experimental medicine[J].2005,201(7):1135-1143[15]J. H.Youn, Y. J. Oh, E. S. Kim, et al. High mobility group box1protein binding to lipopolysaccharide facilitates transfer of lipopoly-saccharide to CD14and enhances lipopolysaccharide-mediated TNF a production in human monocytes. Immunol[J].2008,180:5067-5074[16]E. Seki and D. A. Brenner. Toll-like receptors and adaptor molecules in liver disease:update. Hepatology[J].2008,48(1):322-335[17]Nanji AA, Khettry U, Sadrzadeh SM, et al. Severity of liver injury in experimental alcoholic liver disease. Correlation with plasma endotoxin, prostaglandin E2, leukotriene B4, and thromboxane B2. Am J Pathol[J]1993,142:367-373[18]Su GL, Klein RD, Aminlari A, et al. Kupffer cell activation by lipopolysaccharide in rats: role for lipopolysaccharide binding protein and toll-like receptor4. Hepatology[J].2000,31:932-936[19]Hitoshi Matsumoto, Shinji Tamura, Yoshihiro Kamada, et al. Adiponectin deficiency exacerbates lipopolysaccharide/D-galactosamine-induced liver injury in mice.World Journal of Gastroenterology [J].2006,12(21):3352-3358[20]Sitia G, Iannacone M, M ii ller S, et al. Treatment with HMGB1inhibitors diminishes CTL-induced liver disease in HBV transgenic mice. Journal of leukocyte biology [J].2007,81(1):100-107[21]Ayse Albayrak,Muhammet H. Uyanik,Serkan Cerrah, et al. Is HMGB1a New Indirect Marker for Revealing Fibrosis in Chronic Hepatitis and a New Therapeutic Target in Treatment? Viral Immunology [J].2010,23(6):633-638[22]Liu HB, Fan XG, Huang JJ, et al. Serum level of HMGB1in patients with hepatitis B and its clinical significance.中华肝脏病杂志[J].2007,15(11):812-815[23]Liu CH, Hu w, Wang XL, Liu P, Xu LM. Effects of salvianolic acid-A on NIH/3T3fibroblast proliferation, collagen synthesis and gene expression[J]. World J Gastroenter01.2000,6:361—364[24]Shimlzu I.MizobueK Y, Ysuda M, et al. Inhibitory Effect Oes-tradiol on Activation of Rat Hepatic Stellaee Cells in Vivo andin Vitro[J]. Gut.1999,44:127.[25]Youn, J. H., Y. J. Oh, E. S. Kim, J. E. Choi, and J. S. Shin. High mobility group box1protein binding to lipopolysaccharide facilitates transfer of lipopoly-saccharide to CD14and enhances lipopolysaccharide-mediated TNF a production in human monocytes [J]. Immunol.2008,180:5067-5074.[26]Yong gang Sha, Jaroslaw Zmijewski, Zhiwei, et al. HMGB1Develops Enhanced Proinammatory Activity by Binding to Cytokines [J]. The Journal of Immunology,2008,180;2531-2537[27]Yang-Hua Qin, Sheng-Ming Dai,Gu-Sheng Tang, et al. HMGB1Enhances the Proin fl ammatory Activity of Lipopolysaccharide by Promoting the Phosphorylation of MAPK p38through Receptor for Advanced Glycation End Products [J]. The Journal of Immunology,2009,183;6244-6250.[28]Ekihivo S, Samuele DM,etc. TLR4enhances TGF-β signaling and hepatic fibrosis[J].Nature medicine,2007,13(11):1324-1332[29]Cui X,Shimizu I,Lu G,et al.Inhibitory effect of a soluble transforming growth factor beta type Ⅱ activation of rat hepatic stellate cells in primary culture[J]. J Hepatol,2003;39(5):731-737.[30]Issa R,Zhou XY, Christothea M.Spontaneous recovery from micronoular cirrhosis:evidence for incomplete resolution associated with matrix cross-linking [J]. Gastroenterology,2004,126(7):1795-1808.[31]Jang JH,Kang KJ,Kim YH,et al.Reevaluation of experimental model of hepatic fibrosis induced by hepatotoxic drugs:an easy,applicable,and reproducible model[J]. Transplant Proc,2008,40(8):2700-2703.[32]Wu XL,Zang WZ,Jiang MD,et al. Effect of oxymatrine on the TGF-beta-smad signaling pathway in rats with CCl4-induced hepatic fibrosis[J]. World J Gastroenterol,2008,14(13):2100-2105.[33]叶霖财,肖智勇等。四氯化碳只肝纤维化动物模型实验条件的优化[J].军事医学科学院院报,2010,43(4):340-344.[34]郑敏.ROC曲线评价血清肝纤维化指标在诊断肝纤维化的价值[J].中华传染病杂志,2002,20:225-227.[35]Eghbali-Fatourechi G, Sieck GC, Prakash YS, et al. Type Ⅰ procollagen production and cell proliferation is mediated by transforming growth factor-beta in a model of hepatic fibrosis[J]. Endocrinology,1996,137(5):1894-1903[36]中华肝脏病学会肝纤维化学组.肝纤维化诊断及疗效评估共识[J].中华肝脏病杂志,2002,10(5):327.328[37]姚希贤,徐克成,主编.肝纤维化的基础与临床[M].上海:上海科技教育出版社,[38]Friedman SL. Molecular mechanisms of hepatic fibrosis and principle of therapy[J].Gastroenterology,1997,32:424-430[39]Friedman SL. Stellate cells:a moving target in hepatic fibrogenesis[J]. Hepatology,2004,40(5):1041-1043[40]Guyot C, Lepreux S, Combe C. et al. Hepatic fibrosis and cirrhosis:the (myo)fibroblastic cell subpopulations involved[J].Int J Biochem Cell Biol,2006,38(2):135-151[41]Rippe RA, Brenner DA. From quiescence to activation:Gene regulation in hepatic stellate??cells[J]. Gastroenterology,2004,127(4):1260-1262[42]赵中夫,韩德五,张芸.高迁移率族蛋白-1在实验性急性肝衰竭中的作用.中华肝病杂志.2006;14(5):393-394.[43]Sass G, Heinlein S, Agli A, et al. Cytokine expression in three mouse models of experimental hepatitis[J]. Cytokine2002,19(3):115-120.[44]Y. Li, M. Chang, O. Abar, et al. Multiple variants in toll-likereceptor4gene modulate risk of liver fibrosis in Caucasians with chronic hepatitis C infection [J]. Journal of Hepatology,2009,51(4):750-757[45]WP Pulskens,E Rampanelli,et al.TLR4Promotes Fibrosis but Attenuates Tubular Damage in Progressive Renal Injury[J]J Am Soc Nephrol,2010,21:1299-1308.[46]Yu M, Wang H, Ding A, et al. HMGB1signals through toll-like receptor (TLR)4and TLR2[J]. Shock,2006,26:174-179[47]Fink MP,MDFCCM. Ethyl pyruvate:A novel anti—inflammatory agent[J]. Critical Care Medicine,2003,3(1Supplement):S51—S56[48]Rahman I,MacNee W. Regulation redox glutathione levels and gene transcription in lung inflammation:therapeutic approaches [J]. Free Radic Biol Med,2000,28:1405-1420,[49]姚咏明,姚胜,陈劲松,等.NF.r,B抑制剂对烫伤脓毒症大鼠致炎/抗炎细胞因子表达的影晌[J].解放军医学杂志,2004,29:33-35.[50]陈晓红.细胞因子对肝纤维化形成的影响[J].国外医学内科学分册,2001,28(10)121-22.[51]S chrode r D, Hege r J, Pipe r HM, Eul e r G. Angiotensin II stimulates apoptosis via TGF-betal signaling in ventricular cardiomyocytes of rat[J]. J Mol Med2006;84:975-983[52]Liu J, Gong H, Zhang ZT, Wang Y. Effect of angiotensin II and angiotensin Ⅱ type1receptor antagonist on the proliferation, contraction and collagen synthesis in rat hepatic stellate cells[J]. Chin Med J (Engl)2008;121:161-165[53]Gressner AM, Weiskirchen R. Modern pathogenetic concepts of liver fibrosis suggest stellate cells and TGF-beta as major players and therapeutic target[J]s. J Cell Mol Med,2006,10(1):76-79[54]Pan CH, Wen CH, Lin CS. Interplay of angiotensin II and angiotensin(1-7) in the regulation of matrix metalloproteinases of human cardiocytes[J]. Exp Physiol2008;93:599-612[55]Milan S, Milani, H. Herbst, D. et al.Trasforming growth factors be tagene experession innormal and fibrotic rat liver [J]. J Hepatol,1997,26(4):886-893[56]Anne TM, Paakkop,Sequeiros GB,et al.Matrix metallproteinases immunor ellctive protein:a maker of agressiveness in brest car cinoma[J].Cancer,1998,83:1153-1162[57]张勤,张文PCNA,MMP9预计肝细胞肝癌预后的研究分析[J].临床肝胆病杂志,2003,19(2):113-114[1]Sass G, Heinlein S,Agli A, et al. Cytokine expression in three mouse modles of experitmental hepatitis. Cytokine,2002,19:115-120.[2]Tsung A, Sahai R, Tanaka H et al. The nuclear factor HMGB1mediates hepatic injury after murine liver ischemia-reperfusion. JEM,2005,201:1135-1143.[3]赵中夫,韩德五,张芸,等.高迁移率族蛋白1在实验性急性肝衰竭中的作用。中华肝脏病杂志,2006,14(5):388-389.[4]R. Bataller and D. A. Brenner, Liver fibrosis, Journal of Clinical Investigation [J].2005,115(2)209-218.[5]S. L. Friedman, Mechanisms of hepatic fibrogenesis, Gas-troenterology [J].2008,134(6):1655-1669,[6]Novo E, Marra F, Zamara E. Overexp ression of Bc122by activatedhuman hepatic stellate cells:resistance to apoptosis as amechanismof p rogressive hepatic fibrogenesis in humans [J]. Gut,2006,55(8):11741182[7]Kojiro Taura, Kouichi Miura, Keiko Iwaisako, et al. Hepatocytes Do Not Undergo Epithelial-Mesenchymal Transition in Liver Fibrosis in Mice [J]. Hepatology,2010,51(3):1027-1036[8]Asahina K, Tsai SY, Li P, et al. Mesenchymal orgin of hepatics stellate cells, submesothelial cells, andprivascular mesenchymal cells during mouse liver develupment [J]. Hepatology,2009,49(3):998-1011[9]Pinzani M, Marra F, Garloni V. Cytokine receptors and signaling in hepatic stellate cells [J].Semin Liver Dis,2001,21(3):397-416[10]Cassiman D,Libbrecht L, Dsemet V, et al.Hepatic stellate cell/myofibroblast subpopulation in fibrotic human and rat livers [J].J Hepatol,2002,36(2):200-209[11]Nanji AA, Khettry U, Sadrzadeh SM, et al. Severity of liver injury in experimental alcoholic liver disease. Correlation with plasma endotoxin, prostaglandin E2, leukotriene B4, and thromboxane B2[J]. Am J Pathol1993,142:367-373.[12]Su GL, Klein RD, Aminlari A, et al. Kupffer cell activation by lipopolysaccharide in rats:role for lipopolysaccharide binding protein and toll-like receptor4[J]. Hepatology2000;31:932-936.[13]Hitoshi Matsumoto, Shinji Tamura, Yoshihiro Kamada, et al. Adiponectin deficiency exacerbates lipopolysaccharide/D-galactosamine-induced liver injury in mice [J]. World Journal of Gastroenterology,2006,12(21):3352-3358.[14]Ayse Albayrak, Muhammet H. Uyanik, Serkan Cerrah, et al. Is HMGB1a New Indirect Marker for Revealing Fibrosis in Chronic Hepatitis and a New Therapeutic Target in Treatment Viral Immunology [J].2010,23(6):633-638.[15]Iwamoto K, Kanno K, Hyogo H, et al. Chayama K. Advanced glycation end products enhance the proliferation and activation of hepatic stellate cells [J]. Journal of Gastroenterology,2008;43(4):298-304.[16]Christina Lohwasser, Daniel Neureiter, Yury Popov, et al. Role of the receptor foradvanced glycation??end products in hepatic fibrosis [J].World Journal of Gastroenterology,2009,15(46):5789-5798[17]Y. Kao, B. Jawan, S. Goto, et al. High-Mobility Group Box1Protein Activates Hepatic Stellate Cells In Vitro [J]. Transplantation Proceedings2008,40(8):2704-2705.[18]Youn, J. H., Y. J. Oh, E. S. Kim, J. E. Choi, and J. S. Shin. High mobility group box1protein binding to lipopolysaccharide facilitates transfer of lipopoly-saccharide to CD14and enhances lipopolysaccharide-mediated TNF α production in human monocytes [J]. Immunol.2008,180:5067-5074.[19]Guo J.Friedman SL. Hepaticfibrogensis. Semin Liver Dis,2007,27(4):413-426.[20]DvidKS, Alan BW. Liver fibrosis and inflammation. A REVIEW. annals of HEPATOL,2003,2:159-163.[21]Vaillant B, Chiaramonte MG, Cheever AW, Soloway PD, Wynn TA. Regulation of hepatic fibrosis and extracellular matrix gengs by the thresponse:new insight into the role of tissue inhibitors of matrix metalloproteinnases, JImmunol2001:167:7017-7026.[22]Arthur MJP, Mann DA, Iredale JP. Tissue inhibitors of metalloproteinases, Hepatic stellate cells and liver fibrosis. J Gastroenterol Hepatol,1998,(suppl) S33-S38.[23]Iredale JP, Benyon RC, Pickering J, et al. Mechanisms of spontaneoution resolution ofrat liver fibrosis. Hepatic stellate cell apoptosis and reduced hepatic expression of metalloprotainnase inhibitors.J Clin Invest,1998,102:538-539.[24]E. Seki, S. De Minicis, C. H. Osterreicher et al. TLR4enhances TGF-β signaling and hepatic fibrosis[J]. Nature Medicine,2007,13(11):1324-1332[25]D Onichtchouk, YG Chen, R. Dosch, et al. Silencing of TGF-β signalling by the pseudoreceptor BAMBI [J]. Nature,1999,401(6752):480-485[26]X. Yan, Z. Lin, F. Chen et al. Human BAMBI cooperates with Smad7to inhibit transforming growth factor-β signaling [J]. Journal of Biological Chemistry,2009,284(44):30097-30104[27]Y. Li, M. Chang,0. Abar, et al. Multiple variants in toll-likereceptor4gene modulate risk of liver fibrosis in Caucasians with chronic hepatitis C infection [J]. Journal of Hepatology,2009,51(4):750-757[28]E. Seki and D. A. Brenner, Toll-like receptors and adaptor molecules in liver disease:update [J]. Hepatology,2008,48(1):322-335[29]Yu M, Wang H, Ding A, et al. HMGB1signals through toll-like receptor (TLR)4and TLR2. Shock,2006,26:174-179

© 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