HBV相关慢加亚急性肝衰竭患者外周血辅助性T淋巴细胞及相关炎症因子的表达和临床意义的初步探讨
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
背景在我国,慢性乙型病毒性肝炎(CHB)是引起慢加亚急性肝衰竭(SACLF)的主要病因,有报道称CHB占SACLF所有病因的80%。目前认为,HBV感染患者病情进展和转归受诸多因素影响,其中宿主的免疫功能发挥了重要作用,细胞因子在调节免疫应答中扮演了关键角色。辅助性T细胞(Th细胞)根据分泌的细胞因子不同分为:Th0、Th1、Th2、Th3,目前研究最多的是Th1和Th2两个亚型。研究认为在病毒性肝炎感染中,Th1细胞有利于病毒的清除,而Th2细胞主要与组织损伤及慢性化有关。最近,CD4+Th细胞家族新型成员-Th17细胞亚群的出现改写了经典的Th1/Th2细胞反应模式。该细胞以高分泌IL-17为特征,具有很强的促炎症作用。2005年Kim等人发现了IL-32,该因子可由T细胞、NK细胞、单核细胞、上皮细胞产生,进一步研究表明其有促炎症因子的特性。IL-10是机体一种强有力的免疫和炎症抑制因子,由Th2细胞、巨噬细胞、树突状细胞(Dendritic cells,DC)和B细胞等产生,它可以减低T细胞活性,从而降低免疫反应的效应,导致机体清除病毒能力减弱。上述细胞及细胞因子在CHB中的变化已有报道,但其是否参与了HBV相关慢加亚急性肝衰竭(HBV-SACLF)的发病过程以及在肝衰竭病程中的动态变化趋势、与ALT的相关性研究未见报道。
     目的探讨HBV-SACLF患者外周血Th1、Th2、Th17及血清中IL-32、IL-10的动态变化趋势及其临床意义。
     方法采用流式细胞技术检测38例HBV-SACLF患者、46例肝衰竭前期患者、20例CHB患者、20例健康体检者(对照组),外周全血标本中Th1、Th2、Th17的表达水平,比较四组之间上述指标的差异;动态观察HBV-SACLF患者早、中、晚期Th1、Th2、Th17的变化趋势;比较感染组与非感染组、存活组与非存活组Th1、Th2、Th17表达水平的变化;分析Th1、Th2、Th17与ALT的相关性。同时采用ELISA检测上述四组血清中IL-32、IL-10的表达水平,并观察IL-32、IL-10在HBV-SACLF患者早、中、晚期的动态变化。
     结果1.HBV-SACLF组、肝衰竭前期组、CHB组、对照组Th1、Th2、Th17的表达水平分别为:①T h1:(22.58±3.86)%、(20.80±3.32)%、(5.23±1.51)%、(6.45±1.27)%;Th1比例在HBV-SACLF组与CHB组及对照组比较,均明显升高,差异具有显著性(P_2=0.04,P3=0.032)。②Th2:(1.73±0.64)%、(1.03±0.34)%、(6.15±0.97)%、(1.98±0.46)%;Th2比例在HBV-SACLF组与CHB组及对照组比较,均明显下降,差异具有显著性(P_2=0.007,P3=0.041)。③T h17:(3.48±0.80)%、(3.61±1.20)%、(1.09±0.4)%、(0.72±0.11)%;Th17比例在HBV-SACLF组与CHB组及对照组比较,均明显升高,差异具有显著性(P_2=0.003,P3<0.001)。
     2.HBV-SACLF早期、中期、晚期Th1、Th2、Th17的表达水平分别为:①Th1:(26.11±5.19)%、(22.45±4.06)%、(18.33±3.21)%;Th1比例在HBV-SACLF早期与中期,中期与晚期相比逐渐降低,两组之间比较差异均具有显著性(P1=0.001,P_2=0.004)。②T h2:(0.95±0.20)%、(1.66±0.41)%、(2.54±1.03)%;Th2比例在HBV-SACLF早期与中期,中期与晚期相比逐渐升高,两组之间比较差异均具有显著性(P1=0.003,P_2=0.048)。③Th17:(2.71±0.34)%、(3.39±0.73)%、(4.45±1.22)%;Th17比例在HBV-SACLF早期与中期,中期与晚期相比逐渐升高,两组之间比较差异均具有显著性(P1=0.002,P_2=0.007)。
     3.HBV-SACLF感染组与非感染组Th1、Th2、Th17的表达水平分别为:①Th1:(23.59±3.91)%、(21.44±3.51)%;两组比较差异无显著意义。②T h2:(1.92±0.70)%、(1.76±0.55)%;两组比较差异无显著意义。③T h17:(4.65±1.33)%、(3.92±1.27)%,两组比较差异具有显著性(P=0.032)。
     4.HBV-SACLF存活组与非存活组Th1、Th2、Th17的表达水平分别为:①Th1:(20.78±3.37)%、(25.61±4.05)%;两组比较差异无显著意义。②T h2:(1.65±0.59)%、(1.88±0.63)%;两组比较差异无显著意义。③T h17:(3.21±1.02)%、(4.53±1.29)%,两组比较差异具有显著性(P=0.029)。
     5.对HBV-SACLF患者Th17、Th1、Th2与ALT进行Pearson相关性分析,结果发现Th17与ALT呈正相关(r=0.616,P=0.044),差异具有统计学意义;Th1、Th2与ALT无明显相关性。
     6. HBV-SACLF组、肝衰竭前期组、CHB组、对照组IL-32、IL-10的表达水平分别为:①IL-32:(500.98±152.33)pg/ml、(486.45±129.06)pg/ml、(281.72±99.28)pg/ml、(178.16±50.54)pg/ml;IL-32在HBV-SACLF组与CHB组及对照组比较,均显著升高,差异具有显著性(P_2=0.021,P3=0.033)。②I L-10:(2.82±1.03)pg/ml、(3.05±1.83)pg/ml、(13.15±3.37)pg/ml、(7.62±2.17)pg/ml;IL-10在HBV-SACLF组与CHB组及对照组比较,均显著下降,差异具有显著性(P_2=0.024,P3=0.019)。
     7. HBV-SACLF早期、中期、晚期IL-32、IL-10的表达水平分别为:①IL-32:(540.69±155.71)pg/ml、(498.43±135.56)pg/ml、(450.77±102.33)pg/ml;IL-32在HBV-SACLF早期与中期,中期与晚期相比逐渐降低,两组之间比较差异均具有显著性(P1=0.046,P_2=0.001)。②I L-10:(1.94±0.44)pg/ml、(2.83±0.97)pg/ml、(3.69±1.23)pg/ml;IL-10在HBV-SACLF早期与中期,中期与晚期相比逐渐升高,两组之间比较差异均具有显著性(P1=0.004,P_2=0.032)。
     结论Th1细胞和IL-32在HBV-SACLF患者中明显升高,随着病情发展呈逐渐下降趋势;Th2细胞和IL-10在HBV-SACLF患者中明显降低,其在HBV-SACLF早、中、晚期逐渐升高;Th17细胞在HBV-SACLF患者中明显升高,随着病情发展呈逐渐升高趋势;继发感染组Th17细胞较非感染组明显升高;存活组Th17细胞低于非存活组;且Th17细胞与肝细胞损害呈正相关。本文研究辅助性T细胞1、2、17及细胞因子IL-32、IL-10的紊乱是导致HBV-SACLF发生、发展的关键因素,通过检测其在肝衰竭早、中、晚期的动态变化可以判断机体的免疫状态,为临床的免疫调控治疗提供理论依据。
Background In China, as a major cause of Subacute-on-chronic liver failure(SACLF), CHB accounted for80%of all SACLF etiology. Currently it considered that theoutcome of HBV infection is influenced by many factors, and the host's immune played asignificant role, and the cytokines also played a key role in regulating the immune response.According to the secretion of cytokines, T helper cells were divided into: Th0, Th1, Th2,Th3, Studies suggested that in viral hepatitis infection, Th1cells favored the removal of thevirus while Th2cells were primarily associated with tissue damage and chronic. Recently,a new effecter Th17belonging to CD4+subsets rewritted the classic Th1/Th2cell reactionmode, which was a strong pro-inflammatory factor characterized by secretion ofinterleukin17. In2005,Kim et al found IL-32,which produced by T cells, NK cells,monocytes, epithelial cells.Further studies showed it could promote the characteristics ofinflammatory cytokines. IL-10as a strong immune and inflammatory inhibitor wasproduced by Th2cells, macrophages, dendritic cells (Dendritic cells, DC), and B cells, etc.It could reduce the activity of T cells and then reduced the immune response, making thebody to get rid of the virus diminished capacity. Above cells and cytokines had beenreported t in CHB, whether the dynamic changes of the above cells and cytokines in thehepatitis B-related SACLF (HBV-SACLF)in different stages and the correlation of theirbetween ALT had not been reported.
     Objective To investigate the expression,dynamic change and clinical significance ofTh1、Th2、Th17in peripheral blood and IL-32、IL-10in serum of the patients with SACLF during different periods.
     Methods Detected the ratio of Th1、Th2、Th17from peripheral blood cells in38HBV-SACLF patients,46pre-liver failure patients,20CHB patients and20health adultsby fluorescence-activated cell sorter; compared the above indicators among the four groups;observed dynamic changes of Th1、Th2and Th17during the nonage, metaphase andadvanced stage of HBV-SACLF; studied the expression level changes of Th1, Th2, ofTh17in infection group and non-infected group, survival group and non-survival group;analyzed the correlation of Th1, Th2,Th17and ALT. Detected the expression ofIL-32,IL-10from the serum of38HBV-SACLF patients,46Pre-liver failure patients,20CHB patients and20health adults by ELISA; observed dynamic changes of IL-32,IL-10during the nonage, metaphase and advanced stage of HBV-SACLF.
     Results
     1. The ratios of Th1, Th2,Th17on peripheral blood cells in38HBV-SACLF patients,46Pre-liver failure patients,20CHB patients and20health adults were as follows:①Th1:(22.58±3.86)%、(20.80±3.32)%、(5.23±1.51)%、(6.45±1.27)%;the ratios of Th1in HBV-SACLF group were significantly higher than CHB group and control group(P_2=0.04,P3=0.032).②T h2:(1.73±0.64)%、(1.03±0.34)%、(6.15±0.97)%、(1.98±0.46)%; the ratios of Th2in HBV-SACLF group were significantly lower than CHB group andcontrol group(P_2=0.007,P3=0.041)③Th17:(3.48±0.80)%、(3.61±1.20)%、(1.09±0.4)%、(0.72±0.11)%,;the ratios of Th17in HBV-SACLF group were significantly higherthan CHB group and control group(P_2=0.003,P3<0.001).
     2. The ratios of Th1, Th2and Th17cells in the nonage, metaphase and advanced stageof38HBV-SACLF patients were as follows:①T h1:(26.11±5.19)%、(22.45±4.06)%、(18.33±3.21)%;The ratios of Th1decreased gradually, There were also significantdifferences among the three periods(P1=0.001, P_2=0.004).②Th2:(0.95±0.20)%、(1.66±0.41)%、(2.54±1.03)%;The ratios of Th2increased gradually, There were alsosignificant differences among the three periods(P1=0.003, P_2=0.048).③T h17:(2.71±0.34)%、(3.39±0.73)%、(4.45±1.22)%; The ratio of Th17increased gradually, There were also significant differences among the three periods(P1=0.002, P_2=0.007).
     3.The ratios of Th1,Th2,Th17cells between the infection group and the non-infectedgroup in HBV-SACLF patients were as follows:①T h1:(23.59±3.91)%、(21.44±3.51)%; The difference was not statistically significant between the two groups.②Th2:(1.92±0.70)%、(1.76±0.55)%; The difference was not statistically significant betweenthe two groups.③Th17:(4.65±1.33)%、(3.92±1.27)%; There was significantly differencebetween the two groups (P=0.032).
     4. The ratios of Th1,Th2,Th17cells between the survival group and the death group inHBV-SACLF patients were as follows:①Th1:(20.78±3.37)%、(25.61±4.05)%; Thedifference was not statistically significant between the two groups.②T h2:(1.65±0.59)%、(1.88±0.63)%; The difference was not statistically significant between the two groups.③T h17:(3.21±1.02)%、(4.53±1.29)%; There was significantly difference between thetwo groups (P=0.029).
     5. Correlation analysis of Th1,Th2,Th17and ALT of HBV-SACLF patients: Th17hadsignificant positive correlation with ALT(r=0.616, P=0.044). Th1and Th2hadnon-significant correlation with ALT.
     6. The expression of IL-32, IL-10in38HBV-SACLF patients,46Pre-liver failurepatients,20CHB patients and20health adults were as follows:①IL-32:(500.98±152.33)pg/ml、(486.45±129.06)pg/ml、(281.72±99.28)pg/ml、(178.16±50.54)pg/ml; The ratiosof IL-32in HBV-SACLF group were significantly higher than CHB group and controlgroup(P_2=0.021,P3=0.033).②IL-10:(2.82±1.03)pg/ml、(3.05±1.83)pg/ml、(13.15±3.37)pg/ml、(7.62±2.17)pg/ml; The ratios of IL-10in HBV-SACLF group weresignificantly lower than CHB group and control group (P_2=0.024,P3=0.019).
     7. The ratios of IL-32, IL-10in the nonage, metaphase and advanced stage of38HBV-SACLF patients were as follows:①I L-32:(540.69±155.71) pg/ml、(498.43±135.56)pg/ml、(450.77±102.33)pg/ml; The ratios of IL-32decreased gradually,There were also significant differences among the three periods(P1=0.046, P_2=0.001).②I L-10:(1.94±0.44)pg/ml、(2.83±0.97)pg/ml、(3.69±1.23)pg/ml; The ratios of IL-10 increased gradually, There were also significant differences among the three periods(P1=0.004, P_2=0.032).
     Conclusion The Th1and IL-32in HBV-SACLF patients were significantly increasedand there was a gradual downward trend with the progression of the disease. Th2,Th17andIL-10were significantly higher and upward gradually with the progression of SACLF. Theratios of Th17in infection group were significantly higher than non-infected group;however, it was lower in survival group than death group and had significant positivecorrelation with ALT. In our studies, the disorders of Th1,Th2,Th17and IL-32, IL-10werekey factors in the development of HBV-SACLF. By detecting their dynamic changes inHBV-SACLF, we can judge patients’ immune status and offer theoretical basis for theclinical treatment of immune regulation.
引文
1.中华医学会肝病学分会,感染病学分会.慢性乙型肝炎防治指南[S].中华肝脏病杂志,2005,13(12):881-891.
    2.中华医学会肝病学分会,中华医学会感染病学分会,慢性乙型肝炎防治指南[J].中华流行病学杂志,2006,27(1):79-88.
    3.Zou Z,Chen J,Xin S, et al.Features of onset of chronic severe hepatitis in520cases[J].Zhonghua Shi Yan He Lin Chuang Bing Du Xue ZaZhi,2002,16(4):322-325.
    4.Rehermann B,Nascimbeni M.Immunology of hepatitis B virus and hepatitis C virusinfection[J].Nat Rev lmmunol,2005,5(3):215-229.
    5.Miller SA,Weinmann AS.Common themes emerge in the transcriptional control of Thelper and developmental cell fate decisions regulated by the T-box,GATA and RORfamilies[J].Immunology,2009,126(3):306-315.
    6.Liblau RS,Singer SM,McDevitt HO.Th l and Th2CD4+T cells in the pathogenesis oforgan-specific autoimmune diseases[J].Trends in Immunology,1995,16(1):34-8.
    7.Romagnani S.Thl and Th2in human diseases[J].Clin Immunol Immunopathol,1996,80(1):225-35.
    8.Harrington LE,et a1.Interleukin-17-producing CD4+effector T cells develop via alineage distinct from the T helper type l and2lineages[J].Naturelmmunology,2005,6(11),1123-1132.
    9.Park H,et a1.A distinct lineage of CD4T cells regulates tissue inflammation byproducing Interleukin17[J].Nat Immunol,2005,6(11),1133-1141.
    10.Steinman L. A brief history of T(H)17,the first major revision in the T(H)1/T(H)2hypothesis of T cell-mediated tissue damage[J]. Nat Med,2007,13(2):139-45.
    11.Afzail B, Lombardi G, Lechler RI, Lord GM. The role of T helper17(Th17)andRegulatory T cells(Treg) in human organ transplantation and autoimmunedisease[J]. Clin Exp Immunol,2007,148(1):32-46.
    12.Khader SA, Bell GK, Pearl JE, et al. IL-23and IL-17in the establishment of protectivepulmonary CD4+T cell responses after vaccination and during Mycobacteriumtuberculosis challenge[J]. Nat Immunol,2007,8(4):369-77.
    13.游晶,庄林,马永良等.慢性乙型肝炎的Th细胞亚群及相关细胞因子网络失衡[J].世界华人消化杂志,2007,15(8):791-799.
    14.Kim SH, Han SY, Azam TA, et al. Interleukin-32: a cytokine and inducer of TNFalpha[J]. Immunity,2005,22(1):131-142.
    15.Netea MG,Azam T,Ferwerda G,et a1.IL-32synergizes with nucleotide oligomerizationdomain(NOD)l and(NOD)2ligands for IL-1β and IL-6production through a caspase1-dependent mechanism[J].Proc Natl Acad Sci USA,2005,102(45):16309-16314.
    16.Goda C, Kanaji T, Kanaji S,et al. Involvement of IL-32in activation-induced cell deathin T cells[J]. Int Immunol,2006,18(2):233-240.
    17.Allain JP. Occult hepatitis B virus infection: implications in transfusion[J].VoxSang,2004,86(2):83-91.
    18.Kaminski G,Alnaqdy A,A1-Belushi I,et a1.Evidence of occult hepatitis B virusinfection among omani blood donors:a preliminary study [J].Med PrincPract,2006,15(5):368-372.
    19.Hyodo N,Tajimi M,Ugajin T,Nakamura I,Imawari M. Frequencies ofInterferon-gammaand interleukin-10secreting cells in peripheral blood mononuclear cells and liverinfiltrating lymphocytes in chronic hepatitis B virus infection[J]. Hepatologyresearch,2003,27(2):109-16.
    20.Tülek N,Saglam SK,Saglam M,et al. Soluble interleukin-2receptor and interleukin-10levels in Patienis with chronic hepatitis B infection[J]. Hepatogastroenterology,2000,May-Jun (33):828-31.
    1.中华医学会感染病学分会肝衰竭与人工肝学组中华医学会肝病学分会重型肝病与人工肝学组,肝衰竭诊治指南.中华临床感染病杂志,2012,12(5):321-327.
    2.中华医学会肝病学分会,中华医学会感染病学分会,慢性乙型肝炎防治指南[J].中华流行病学杂志,2006,27(1):79-88.
    3.Sarin SK,Kumar A, Almeida JA,et a1.Acute-on-chronic liver failure:consensusrecommendations of the Asian Pacific Association for the study of theliver(APASL)[J].Hepatol Int,2009,3(1):269-282.
    4.叶一龙,高志良.肝衰竭发生机制中的三重打击学说—免疫损伤,缺血缺氧和内毒素血症.国际肝病,2009,22:13.
    5.Rehermann B,Nascimbeni M.Immunology of hepatitis B virus and hepatitis C virusinfection[J].Nat Rev lmmunol,2005,5(3):215-229.
    6.Miller SA,Weinmann AS.Common themes emerge in the transcriptional control of Thelper and developmental cell fate decisions regulated by the T-box,GATA and RORfamilies[J].Immunology,2009,126(3):306-315.
    7.杨婉凤金树根,汤小青等.肝病患者外周血CD4+、CD8+和CD16+56+细胞亚群变化[J].世界肿瘤杂志,2002,1(2):119-122.
    8.游晶,庄林,马永良,唐宝璋.慢性乙型肝炎的Th细胞亚群及相关细胞因子网络失衡[J].世界华人消化杂志,2007,15(8):791-799.
    9.Liblau RS,Singer SM,McDevitt HO.Th l and Th2CD4+T cells in the pathogenesis oforgan-specific autoimmune diseases[J].Trends in Immunology,1995,16(1):34-8.
    10.Romagnani S.Thl and Th2in human diseases[J].Clin Immunol Immunopathol,1996,80(1):225-35.
    11.Akpolat N,Yahsi S,Godekmerdan A,et a1.Relationship between serum cytokine levelsand histopathological changes of liver in patients with hepatitis B[J].World JGastroenterol,2005,11(21):3260-3.
    12.Milich DR,Jones JE,Hughes JL,et a1.Is a function of the secreted hepatitis B e antigento induce immunologic tolerance in utero?[J]. Proc Natl Acad Sci USA,1990,87(17):6599-603.
    13.Jiang R,Feng X,Guo Y, et a1.T helper cells in patients with chronic hepatitis B virusinfection [J].Chinese Medical Journal,2002,115(3):422-424.
    14.Szkaradkiewicz A,Jopek A,Wysocki J,et a1.HBcAg-specific cytokine production byCD4T lymphocytes of children with acute and chronic hepatitis B[J].VirusResearch,2003,97(2):127-133.
    15.Miyamoto M, Prause O, Sj strand M, et al.Endogenous IL-17as a mediator ofneutrophil recruitment caused by endotoxin exposure in mouse airways [J]. J Immunol,2003,170(9):4665-4672.
    16.Ye P,Garvey PB, Zhang P, et al.Interleukin-17and lung host defense against Klebsiellapneumoniae infection [J].Am J Respir Cell Mol Biol,2001,25(3):335-340.
    17.Weaver CT, Hatton RD, Mangan PR, et al. IL-17family cytokines and the expandingdiversity of effector T cell lineages[J]. Annu Rev Immunol,2007,25(1):821-852.
    18.Liang SC,Tan XY, Luxenberg DP, et al.Interleukin (IL)-22and IL-17are coexpressedby Th17cells and cooperatively enhance expression of antimicrobial peptides [J]. J ExpMed,2006,203(10):2271-2279.
    19.Ouyang W, Kolls JK, Zheng Y.The biological functions of T helper17cell effectorcytokines in inflammation [J]. Immunity,2008,28(4):454-467.
    20.Starnes T,Broxmeyer H E, Robertson MJ, et al. Cutting edge: IL-17D, a novel memberof the IL-17family, stimulates cytokine production and inhibits hemopoiesis [J]. JImmunol,2002,169(2):642-646.
    21.Witowski J, Pawlaczyk K, Breborowicz A, et al. IL-17stimulates intraperitonealneutrophil infiltration through the release of GRO alpha chemokine from mesothelialcells[J]. J Immunol,2000,165(10):5814-5821.
    22.Nowak EC,Weaver CT,Turner H,et a1.IL-9as a mediator of Th l7-driven inflammatorydisease[J].J Exp Med,2009,206(8):1653-1660.
    23.Yamada H.Th17cells in human rheumatoid arthritis[J].Nihon Rinsho Meneki GakkaiKaishi,2009,32(4):249-255.
    24.Kryczek I,Banerjee M,Cheng P,et a1.Phenotype,distribution,generation,and functionaland clinical relevance of Th17cells in the human tumor environments[J].Blood,2009,114(6):1141-1149.
    25.Luzza F,Parrello T,Monteleone G,et a1.Up-regulation of IL-17is associated withbioactive IL-8expression in Helicobacter pylori-infected human gastric mucosa[J].JImmunol,2000,l65(9):5332-5337.
    26.Johnson RB,Wood N,Serio FG.Interleukin-11and IL-17and the pathogenesis ofperiodontal disease[J].J Periodontol,2004,75(1):37-43.
    27.Chabaud M,Durand JM,Buchs N,et a1.Human interleukin-17:A T cell-derivedproinflammatory cytokine produced by the rheumatoid synovium[J].ArthritisRheum,1999,42(5):963-970.
    28.郭宁红,石庆之.Th17细胞与自身免疫性血液病[J].重庆医学,2010,39(18):2533-2535.
    29.Zhang B,Rong G,Wei H,et a1.The prevalence of Thl7cells in patients with gastriccancer[J].Biochem Biophys Res Commun,2008,374(3):533-537.
    30.Ge J,Wang K,Meng QH,et al.Implication of Thl7and Thl cells in patients with chronicactive hepatitis B[J].J Clin Immunol,2010,30(1):60-67.
    31.Zhang JY,Zhang Z,Lin F,et a1.Interleukin-17-producing CD4(+) T cells increase withseverity of liver damage in patients with chronic hepatitis B[J].Hepatology,2010,51(1):81-91.
    32.Zenewicz LA,Yancopoulos GD,Valenzuela DM,etAl.Innate and adaptive interleukin-22protects mice from inflammatory bowel disease[J].Immunity,2008,29(6):947-57.
    33.马劲夫,黎檀实等.Th17细胞在脓毒症中的作用机制研究[J].军医进修学院学报,2010,31(7):718-720.
    34.游晶,庄林,马永良等.慢性乙型肝炎的Th细胞亚群及相关细胞因子网络失衡[J].世界华人消化杂志,2007,15(8):791-799.
    35.Leon An,Wang XL, et al.Cytokines in alcoholic liver disease[J].Arch Toxicol,2012,86:1337-1348.
    36.周卫敏,许青田.慢性乙型肝炎患者血清HBV DNA与病毒复制水平相关性分析[J].临床研究,2008,5(12):79-82.
    37.Goda C, Kanaji T, Kanaji S,et al. Involvement of IL-32in activation-induced cell deathin T cells[J]. Int Immunol,2006,18(2):233-240.
    38.Allain JP. Occult hepatitis B virus infection: implications in transfusion[J].VoxSang,2004,86(2):83-91.
    39.Kaminski G,Alnaqdy A,A1-Belushi I,et a1.Evidence of occult hepatitis B virus infectionamong Omani blood donors:a preliminary study [J].Med Princ Pract,2006,15(5):368-372.
    40.许春梅,左维泽,沈兰超等.白介素-32在HBV感染者的变化及其临床意义[J].现代生物医学进展,2010,10(2):298-301.
    41.Hyodo N,Tajimi M,Ugajin T,et al.Frequencies of Interferon-gamma and interleukin-10secreting cells in peripheral blood mononuclear cells and liver infiltrating lymphocytesin chronic hepatitis B virus infection[J].HePatol Res,2003,27(2):109-16.
    42.Tülek N,Saglam SK,Saglam M,et al. Soluble interleukin-2receptor and interleukin-10levels in patienis with chronic hepatitis B infection[J]. Hepatogastroenterology,2000,47(33):828-31.
    1.Abbas AK, Murphy KM, Sher A. Functional diversity of helper T lymphocytes [J].Nature,1996,383(6603):787-793.
    2.Langrish CL, Chen Y, Blumenschein WM, et al.IL-23drives a pathogenic T cellpopulation that induces autoimmune inflammation [J]. J Exp Med,2005,201(2):233-240.
    3.Harrington LE, Hatton RD, Mangan PR, et al. Interleukin17-producing CD4+effector Tcells develop via a lineage distinct from the T helper type1and2lineages [J].NatImmunol,2005,6(11):1123-1132.
    4.Park H, Li Z, Yang XO, et al. A distinct lineage of CD4T cells regulates tissueinflammation by producing interleukin17[J]. Nat Immunol,2005,6(11):1133-1141.
    5.Infante-Duarte,Horton HF,Byrne MC,et a1.Microbial lipopeptides induce the productionof IL-17in Th cells[J].J Immunol,2000,165(11):6107-6115.
    6.Bettelli E, Carrier Y, Gao W, et al. Reciprocal developmental pathways for the generationof pathogenic effector TH17and regulatory T cells[J]. Nature,2006,441(7090):235-
    238.
    7.Veldhoen M, Hocking RJ, Atkins CJ, et al. TGF-beta in the context of an inflammatorycytokine milieu supports de novo differentiation of IL-17-producing T cells[J].Immunity,2006,24(2):179-189.
    8.Mangan PR, Harrington LE, O'Quinn DB, et al. Transforming growth factor-beta inducesdevelopment of the T (H)17lineage[J].Nature,2006,441(7090):231-234.
    9.Wilson NJ, Boniface K, Chan JR, et al. Development, cytokine profile and function ofhuman interleukin17-producing helper T cells [J].Nat Immunol,2007,8(9):950-957.
    10.Zhou L, Lopes JE, Chong MM, et al.TGF-β-induced Foxp3inhibits TH17celldifferentiation by antagonizing RORγt function[J] Nature,2008,453(7192),236-240.
    11.Ivanov II,McKenzie BS,Zhou L,et al.The orphan nuclear receptor RORgammat directsthe differentiation program of proinflammatory IL-17+Thelper cells[J]. Cell,2006,126(6):1121-1133.
    12.郑钰涵,吴晓东,孙兵.Th17细胞分化和功能的研究进展[J].生命科学,2010,22(6):534-538.
    13.Cruz A,Khader SA,Torrado E,et al.Cutting edge:IFN-gamma regulates the inductionand expansion of IL-17-producing CD4T cells during mycobacterial infection[J].JImmunol,2006,177(3):1416-1420.
    14.Stumhofer JS,Laurence A,Wilson EH,et al.Interleukin27negatively regulates thedevelopment of interleukin17-producing T helper cells during chronic inflammation ofthe central nervous systerm[J].Nat Immunol,2006,7(9):937-945.
    15.Kolls JK, Lindén A.Interleukin-17family members and inflammation[J]. Immunity,2004,21(4):467-476.
    16.Aggarwal S,Gurney AL.IL-17:prototype member of an emerging cytokine family[J].JLeukoc Bio1,2002,71(1):l-8.
    17.Moseley TA,Haudenschild DR,Rose L,et al.Interleukin-17family and IL-17receptors[J].Cytokine Growth Factor Rev,2003,14(2):155-74.
    18.Silva WA Jr,Covas DT,Panepucci RA,et al.The profile of gene expression of humanmarrow mesenchymal stem cells[J].Stem Cell,2003,21(6):661-9.
    19.Zenewicz LA,Yancopoulos GD,Valenzuela DM,et a1.Interleukin-22but notinterleukin-l7provides protection to hepatocytes during acute liverinflammation[J].Immunity,2007,27(4):647-659.
    20.Ouyang W,Kolls JK,Zheng Y.The biological functions of T helper17cell effectorcytokines in inflammation[J].Immunity,2008,28(4):454-467.
    21.Chen MT,Billaud JN,S llberg M,et a1.A function of the hepatitis B virus precoreprotein is to regulate the immune response to the core antigen[J].Proc Natl Acad SciUSA,2004,101(41):14913-8.
    22.Akpolat N,Yahsi S,Godekmerdan A,et a1.Relationship between serum cytokine levelsand histopathological changes of liver in patients with hepatitis B[J].World JGastroenterol,2005,11(21:3260-3.
    23.Wahl C,Wegenka UM,Leith user F,et a1.IL-22-dependent attenuation of Tcell-dependent(ConA)hepatitis in herpes virus entry mediator deficiency[J].Jlmmunol,2009,182(8):4521-4528.
    24.Svetlana Radaeva,Rui Sun,Hong-na pan,et al. Interleukin22(IL-22)plays a protectiverole in T cell–mediated murine hepatitis:IL-22is a survival factor for hepatocytes viaSTAT3activation[J]. Hepatology,2004,39(5):1332-1342.
    25.Aggarwal S,Ghilardi N,Xie MH,et a1.Interleukin-23promotes a distinct CD4T cellactivation state characterized by the production of interleukin-17[J].J BiolChem,2003,278(3):1910-1914.
    26.Iwakura Y,Ishigame H.The IL-23/IL-17axis in inflammation[J].J ClinInvest,2006,116(5):1218-1222.
    27.Caruso R,Pallone F,Monteleone G. Emerging role of IL-23/IL-17axis in Hpylori-associated pathology[J].World J Gastroenterol,2007,13(42):5547-5551.
    28. Zelante T,De Luca A,Bonifazi P,et a1.IL-23and the Thl7pathway promoteinflammation and impair antifungal immune resistance[J].Eur JImmunol,2007,37(10):2695-2706.
    29. Hurst SD, Muchamuel T, Gorman DM, et al. New IL-17family members promote Th1or Th2responses in the lung: in vivo function of the novel cytokine IL-25[J].JImmunol,2002,169(1):443-453.
    30.Kim M R, Manoukian R, Yeh R, et al. Transgenic overexpression of human IL-17Eresults in eosinophilia, B-lymphocyte hyperplasia, and altered antibodyproduction[J].Blood,2002,100(7):2330-2340.
    31.Weir C,Bernard CC,B ckst rm BT.IL-5-deficient mice are susceptible to experimentalautoimmune encephalomyelitis[J].Int Immunol,2003,15(11):1283-1289.
    32.Fujino S, Andoh A, Bamba S, et al. Increased expression of interleukin17ininflammatory bowel disease[J]. Gut,2003,52(1):65-70.
    33.Lan RY, Salunga TL, Tsuneyama K, e t al. Hepatic IL-17responses in human andmurine primary biliary cirrhosis [J].J Autoimmun,2009,32(1):43-51.
    34.Rong G, Zhou Y, Xiong Y, et al. Imbalance between T helper type17and T regulatorycells in patients with primary biliary cirrhosis: the serum cytokine profile and peripheralcell population [J].Clin Exp Immunol,2009,156(2):217-225.
    35.Harada K,Shimoda S,Sato Y,et al.Periductal interleukin-17production in associationwith biliary innate immunity contributes to the pathogenesis of cholangiopathy inprimary biliary cirrhosis[J]. Clin Exp Immunol,2009,157(2):261-270.
    36.Shimada N,Yamamoto K,Kuroda MJ, et al.HBcAg-specific CD8T cells play animportant role in virus suppression,and acute flare-up is associated with the expansion ofactivated memory T cells[J].J Clin Immunol,2003,23(3):223-232.
    37.Tsai SL,Sheen IS,Chien RN,et al.Activation of Th1immunity is a common immunemechanism for the successful treatment of hepatitis B and C: tetramer assay andtherapeutic implications[J].J Biomed Sci,2003,10(1):120-135.
    38.Ge J,Wang K,Meng QH,et al.Implication of Thl7and Thl Cells in Patients with chronicactive hepatitis B[J].J Clin Immunol,2010,30(1):60-7.39Zhang JY,Zhang Z,Lin F,et a1.Interleukin-17-producing CD4(+) T cells increase withseverity of liver damage in patients with chronic hepatitis B[J].Hepatology,2010,51(1):81-91.
    40.Marc Veldhoen,Richard J,Hocking,et a1.TGF beta in the context of an inflammatorycytokine milieu supports de novo differentiation of IL-17-producing T cells[J].Imunity,2006,24(2):179-189.
    41.Wu Y, Zhang J, Chen S, et al. Frequencies of epitope-specific cytotoxic T lymphocytesin active chronic viral hepatitis B infection by using MHC class I peptidetetramers[J].Immunol Lett,2004,92(3):253-258.
    42.Yasumi Y,Takikawa Y,Endo R,Suzuki K.Interleukin-17as a new marker of severity ofacute hepatic injury[J].Hepatol Res,2007,37(4):248-54.
    43. Estelle Bettelli,Thomas Korn,Mohamed Oukka, et a1.Induction and effector functionsof TH17cells[J].Nature,2008,453(7198):1051-1057.
    44.Diaz JA,Booth AJ,Lu G,et a1.Critical role for IL-6in hypertrophy and fibrosis inchronic cardiac allograft rejection[J].Am J Transplant,2009,9(8):1773-1783.
    45.Faust SM,Lu G,Marini BL,et a1.Role of T cell TGFbeta signaling and IL-I7in allograftacceptance and fibrosis associated with chronic rejection [J].J Immunol,2009,183(11):7297-7306.
    46.秦来英,杜文军,刘葵花等.白细胞介素17的肝内表达与慢性乙型肝炎肝纤维化的相关性[J].中华肝脏病杂志,2009,17(3):221-222.
    47.Mantovani A,Allavena P,Sica A,et al. Cancer-related inflammation[J].Nature,2008,454(7203):436-444.
    48.Zhang JP, Yan J, Xu J, et al. Increased intratumoral IL-17-producing cells correlatewith poor survival in hepatolcellular carcinom a patients[J]. J Hepatol,2009,50(5):980-989.
    49.Numasaki M, Fukushi J, Ono M, et al. Interleukin-17promotes angiogenesis andtumor growth[J]. Blood,2003,101(7):2620-2627.
    50.Numasaki M, Watanabe M, Suzuki T, et al. IL-17enhances the net angiogenicactivity and in vivo growth of human non-small cell lung cancer in SCID mice throughpromoting CXCR-2-dependent angiogenesis[J]. J Immunol,2005,175(9):6177-6189.
    51. Chabaud M, Lubberts E, Joosten L, et al. IL-17derived from Juxta-articular boneand synovium contributes to joint degradation in rheumatoid arthritis[J]. Arthritis Res,2001,3(3):168-177.
    52.Caldwell CC,Okaya T,Martignoni A,et al. Divergent functions of CD4+T lymphocytesin acute liver inflammation and injury after ischemia-reperfusion [J].Am J PhysiolGastrointest Liver Physiol,2005,289(5):969-976.
    53.Maher JJ. Treatment of alcoholic hepatitis[J].J Gastroenterol Hepatol,2002,17(4):448-55.
    54.Lemmers A, Moreno C, Gustot T, et al. The interleukin-17pathway is involved inhuman alcoholic liver disease[J]. Hepatology,2009,49(2):646-657.
    55.Fábrega E,López-Hoyos M,San Segundo D,et al. Effect of immunosuppressant bloodlevels on serum concentration of interleukin-17and-23in stable liver transplantrecipients[J]. Transplant Proc,2009,41(3):1025-1027.

© 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