术后腹腔细菌感染对原位肝移植大鼠细胞免疫功能的影响及胸腺肽α1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
肝移植已成为治疗终末期肝病最有效的手段,然而,感染仍是肝移植术后严重影响受体生存率和移植物存活率的主要并发症之一。由于受体术前基础疾病的严重性以及肝移植手术的特殊性,术后感染表现为病原菌种类复杂、病情凶险、死亡率极高。并且,感染与移植后排斥反应关系密切,病毒感染已被证实可促进移植后排斥反应的发生,而细菌感染与术后排斥反应的关系目前尚未完全清楚。重症感染时,机体免疫系统功能受到抑制,T细胞亚群比例失调、功能受损,呈现一种“免疫麻痹”现象,这一概念已得到越来越多证据的支持。在众多因素中,DCs的表达变化引起了广泛观注,它可通过异质性改变,影响效应性T细胞的活化,发挥免疫抑制效应。相对于此,免疫调理对脓毒症的治疗作用也越来越明确。但是,由于担心诱发排斥反应的发生,器官移植术后感染免疫调理治疗仍有争议。本研究采用大肠杆菌诱导近交系大鼠肝移植术后腹腔感染,在此基础上,给予胸腺肽α1免疫调理治疗,通过检测受体细胞免疫功能的变化,从而为临床治疗提供一定的基础理论依据。
     目的
     建立近郊系大鼠原位肝移植术后腹腔细菌感染模型,研究细菌感染对移植肝急性排斥反应的影响,进而研究其对受体T细胞免疫功能的影响及脾脏DCs的增殖情况和其生物学效应,并探讨胸腺肽α1对其的免疫调理作用。
     方法
    
     1.建立封闭群大鼠原位全肝移植模型:以Kamada经典二袖套法为基础,采用快速切取供肝,一针法连续缝合肝上下腔静脉,进行120例大鼠原位肝移植,观察各手术步骤操作时间、术后并发症及术后生存期。
     2.建立大鼠原位肝移植急性排斥反应模型:分3种移植组合:SD→SD组(同基因对照组),SD→Wistar组及DA→Lewis组,每组32例。分别于术后3、5、7、10天各随机处死4只,观察外周血肝功能变化(ALT和TBIL)与肝脏病理改变,将余大鼠留作生存分析。
     3.建立大鼠原位肝移植急性排斥反应模型并留置腹腔导管,于术后3天、5天两个时间点灌注ATCC25922大肠杆菌活菌,每个时间点分为5个感染组:2×108cfu/ml、2×107cfu/ml、5×106cfu/ml、1×106cfu/ml、5×105cfu/ml,1个对照组:腹腔灌注灭菌生理盐水2ml。于灌注前1d、灌注后1d、3d、5d、7d共5个时间点,每个时间点4只大鼠,另设8只大鼠观察感染后一般情况及生存时间。观察受体一般情况、腹水细菌培养及白细胞计数、外周血白细胞计数、TCO2、HCO3-、PH、ALT、TB以及肝肾肺HE染色,评价感染的状态及严重程度。
     4.以DA或LEW大鼠为供体,LEW大鼠为受体,建立大鼠肝移植模型,根据有无诱导感染分为4组:G1组为LEW→LEW,无感染组;G2组为LEW→LEW,感染组;G3组为DA→LEW,无感染组;G4组为DA→LEW,感染组。注菌时间为术后第三天,浓度1×106cfu/mL。每组于感染前1d、感染后1、3、5d,即术后第2、4、6、8d,共4个时间点,每个时间点4只大鼠。通过观察肝脏病理改变、免疫组化法及荧光定量PCR检测移植肝内CXCR3、CXCL10变化及TUNEL法检测肝脏淋巴细胞凋亡情况,评价细菌感染对移植肝免疫排斥反应的影响。
     5.实验动物分组、处置及取材时间同步骤4,应用流式细胞术检测外周血T淋巴细胞亚群比例,单相混合淋巴细胞培养鉴定其功能,ELISA法检测血清细胞因子改变,观察细菌感染对受体T淋巴细胞亚群、功能和分化的影响。
     6.实验动物分组、处置及取材时间同步骤4,通过免疫磁珠分选术分离受体脾脏DCs,流式细胞术检测其表型、单相混合淋巴细胞培养鉴定其功能的改变,观察细菌感染对受体脾脏DCs的影响。
     7.以DA大鼠为供体,LEW大鼠为受体,建立大鼠肝移植模型,根据有无诱导感染和/或药物干预分为4组:G1组为移植对照组;G2组在G1组基础上药物干预;G3组为感染组;G4组感染后药物干预组。感染诱导方法同步骤4,胸腺肽α1一次性2mg于术后6天或感染后72h注入腹腔。每组于感染前1d、胸腺肽α1处理后2d,即术后第8d,共2个时间点,每个时间点4只大鼠,每组另设6只大鼠观察生存期。观察大鼠一般情况、生存期、移植肝病理变化、外周血T细胞亚群比例及功能变化,探讨免疫调理对大鼠肝移植术后腹腔感染的治疗作用。
     8.统计学处理:应用SPSS16.0软件进行单因素方差分析、t检验、秩和检验、生存分析等,P<0.05判定为有统计学意义。
     结果
     1.大鼠原位肝移植至操作稳定阶段,各主要步骤的时间为:供体手术30.2±2.5min,修肝5.7±1.6 min,肝上下腔静脉吻合9.1±0.8 min,门静脉重建1.6±0.5 min,肝下下腔静脉重建1.5±0.4 min,无肝期15.4±1.1 min,胆道重建1.1±0.3 min,受体手术39.5±1.4 min。手术成功率100%,1周存活率97.7%,晚期并发症如胆道梗阻发生率为3.3%。
     2.SD→SD组均生存超过120d;SD→Wistar组中位生存时间为100d,95%可信区间为(83.369~116.631)d;两组差异无统计学意义(P=0.317);DA→Lewis组中位生存时间为12d,95%可信区间为(10.125~13.848)d,生存时间明显较SD→Wistar组缩短,差异有统计学意义(P=0.000)。SD→SD组血清ALT、TBIL浓度术后随时间的延长,逐渐下降,肝功能好转;SD→Wistar组血清ALT、TBIL浓度随时间延长,于术后5d下降相对较慢,随后逐步下降;DA→Lewis组血清ALT、TBIL浓度术后随时间的延长,呈进行性升高。至术后10d,ALT:SD-SD< SD-Wistar     3.生存期的比较,术后5d腹腔注菌组感染后存活时间最短(大部分在1~3d)。术后3d注菌,G1、G2、G3组大多数在感染后2~3d死亡;G4组存活时间在感染后5~9d;G5组存活时间在感染后9~10d;Log-Rank:G4组与G5组比较,χ2=13.322,ν=1,P = 0.000,差异有显著统计学意义;G4组与G0组比较,χ2=11.829,ν=1,P = 0.001,差异有显著统计学意义;G5组与G0组比较,χ2=0.013,ν=1,P = 0.909,差异无统计学意义。腹水白细胞计数比较,G0组术后早期轻度升高,后期降至诊断标准以下;G4组持续显著升高;G5组先升高后下降,感染后7d与G4组比较差异有显著统计学意义(P<0.01)。G4组腹水细菌培养持续阳性,G5组后期腹水细菌培养阴性。随着时间推移,G4外周血WBC、ALT、TBIL均持续升高,而PH值、HCO3-、TCO2持续下降,表现为较重的酸碱平衡紊乱、肝功能异常,至观察终点与G0组比较均有统计学差异(P<0.05);G5感染后早期亦有不同程度异常,但后期均好转,至观察终点,与G4组比较均有统计学差异(P<0.05)。HE染色结果显示,各组至观察终点肾肺均未见明显异常病理改变。
     4.HE结果显示,感染后第5d(术后第8d),G3组4只大鼠病理表现均呈重度急性排斥反应,G4组仅有1只受体大鼠移植肝呈重度急性排斥反应表现,而肝实质损害加重,RAI评为为(8.0±0.8 vs 6.5±1.3,P<0.05)。CXCL-10及其配体CXCR-3免疫组化结果及mRNA水平的相对表达量,G3组呈持续上升趋势;而G4组观察时间内早中期升高,感染后5d下降,此时与G3组的比较差异有统计学意义(P<0.05)。各组移植肝内凋亡细胞均持续增多,但G3组凋亡细胞主要分布在肝实质,随时间推移,汇管区凋亡细胞增多;G4组凋亡细胞主要分布在肝实质,随感染加重,汇管区凋亡细胞明显增多,术后8d与G3组的比较差异有统计学意义(P<0.05)。
     5.混合淋巴细胞培养结果显示:G1组T淋巴细胞功能术后轻度升高,G2组淋巴细胞功能呈上升趋势,G3组淋巴细胞功能明显升高,G4组淋巴细胞功能持续升高,G2、G4组淋巴细胞功能上升幅度均不及G3组,且在感染后上升趋势明显变缓。G1、G2、G3组术后第8d淋巴细胞功能比较差别均有统计学意义(0.3727±0.05479 vs 0.7795±0.00995 vs 0.9550±0.03594,P<0.05);G4组感染后第5d(术后第8d),淋巴细胞功能低于G3组(0.7560±0.00787 vs 0.9550±0.03594,P<0.05)。流式细胞术结果显示:G1组T淋巴细胞亚群及其比值均变化不大;G2组感染后5d,T细胞亚群比值降低;G3组CD4+和CD8+T细胞在术后前三个时间点有所下降,至术后8d方回升,CD8+/CD4+T细胞改变升高,与术后2d比较差异有统计学意义(P<0.05);G4组随着感染的加重,CD4+和CD8+T细胞均较快下降,两者比值亦持续下降,至感染后5d,其比值在1.43~1.46之间,较之感染前差异有统计学意义(P<0.05)。感染后5d(术后8d),三者差异G4组与G1、G3组差异均有统计学意义。外周血IL-10水平的变化:G1组术后比较稳定,变化不大;G2组感染后呈低水平升高;G3组术后持续降低;G4组感染后持续显著升高;术后第8d,G3     6.经免疫磁珠新鲜分离的大鼠脾脏DCs具有典型的树突状特征,每个脾脏可得到约1.5~3×106个OX62+细胞。代表DCs成熟状态的表面分子MHC-Ⅱ、CD80、CD86三者的变化趋势在各组术后各时间点基本保持一致。G1组术后高表达;G2组在感染后表达降低;G3组术后表达上升;G4组随着感染加重,表达水平明显降低,DCs趋于幼稚化。G2组三者表达率术后2d与术后8d差异均有统计学意义(P<0.05);G3组MHC-Ⅱ表达率术后2d与术后8d差异有统计学意义(P=0.017),CD80表达率虽不具统计学差异,但还是有所升高,CD86表达率有统计学差异;G4组三者表达率,自感染后第一天起,各时间点比较差异均有统计学意义(P<0.01);术后8d,三者表达率在各组之间均有明显差异(P<0.01)。DCs刺激同种异体T细胞增殖的能力,G4组术后8d明显下降,G3组上升,(0.27±0.05 vs 0.93±0.02,P<0.01)。
     7.G1组与G2组中位生存时间均为12d,95%CI分别为(10.400~13.600)和(10.868~13.132),两者比较差异无统计学意义(P = 0.416);G3组与G4中位生存时间分别为9d和10d,95%CI分别为(7.400~10.600)和(8.868~11.132),两者比较差异无统计学意义(P = 0.671)。至观察终点,G1、G2组RAI评分均为8~9分,属于重度急性排斥反应;G3、G4组术后第8d移植肝RAI评分6~8分,介于中到重度排斥反应之间;G1与G2、G3与G4组比较差异均无统计学意义(P>0.05)。处理后,G2组、G4组淋巴细胞亚群数量及比值均升高,T淋巴细胞功能升高,G2组与G1组及G4组与G3组比较,差异均有统计学意义(P<0.05)。
     结论
     1.本实验建立了稳定的封闭群大鼠原位肝移植模型,手术时间短、成功率高、稳定可靠。在此基础上,成功建立了稳定的近交系大鼠原位肝移植急性排斥模型,结果稳定、可靠、重复性好,可用于相关领域的基础研究。
     2.本实验采用腹腔内大肠杆菌灌注建立肝移植术后腹腔细菌感染的模型可行、可靠、可重复:1)可比较确切的证实腹腔内感染的存在,2)无需二次手术诱导感染,减少创伤对受体大鼠的打击,可满足实验需要。
     3.随着感染加重,一定程度上加重了肝实质损害,但部分缓解了肝移植排斥反应病理表现程度。趋化因子CXCL-10及其受体CXCR-3在感染后期表达减少以及移植肝汇管区及小叶中央静脉周围浸润淋巴细胞凋亡增多是移植肝排斥反应减轻的原因之一。
     4.排斥反应和感染早期T淋巴细胞功能均增加,移植术后感染后期淋巴细胞功能严重降低;排斥反应使机体CD4+/CD8+T细胞比值升高;感染使CD4+/CD8+比值早期增加,晚期降低;二者的协同作用则使CD4+/CD8+T细胞比值显著下降。排斥反应发生时,血清IFN-γ水平升高;感染作用下,IL-10水平升高;两者共同作用下,促进Th1细胞向Th2细胞转变。机体抗炎反应占优势,表现为一种局部和全身免疫抑制状态。
     5.细菌感染早期或急性免疫排斥反应均可促进脾脏DCs快速成熟,促进其刺激同种异体T细胞增殖的功能;至感染后期,尤其是排斥反应和感染同时作用时,DCs表现为未成熟状态,刺激同种异体T细胞增殖的功能明显下降。
     6.大鼠肝移植术后合并腹腔细菌感染,短时间内,给予免疫调理治疗未明显延长受体生存时间,但亦未发现加重移植肝急性排斥反应的程度,却能提高肝移植术后受体T淋巴细胞亚群、比值及淋巴细胞功能。
Liver transplantation(LT) has become the most effective treatment for patients with end-staged liver diseases, however, infections is still common after LT and seriously decreases the recepients and graft survival. As the severity of illness of recepients pretransplant, as well as the particularity of LT, posttransplantive infections charactered various of pathogen species, severity, and high morbidity and mortality. Moreover, infection related with rejection posttransplant. Viral infection has been shown to promote chronic rejection posttransplant, whilethe relationship between bacterial infection and rejection are not entirely definited. Severe infections, accompanied with imbalance and functional impairment of T cell subsets, showed "immune paralysis" of the immune system. DCs play an immunosuppressive effect role during this course via their heterogeneity. There has been growing evidences to support this. The effection of immune regulation treatments to sepsis has been more explicit. However, it is controversial in organ transplant due to fear of rejection. In this study, the intraabdominal infection was induced by E. coli after orthotopic liver transplantation in rats(ROLT), and thymosin-alpha1 was injected intraperitoneally then, the changes of cellular immune response was monitored.
     Objective
    
     To momitor the immume function changes of T cells and the proliferation and biological effect of dendritic cells derived from spleen and to explore the role of thymosinα1 in the immune conditioning of recepients by established a model of intraabdominal bacterial infection following orthotopic liver transplantation in rat.
     Methods
     1. Established the orthotopic liver transplantation model of closed colony rats: based on the classical two-cuff technique by Kamada, 120 cases of rat orthotopic liver transplantation were performed with fast resecting the donor liver, continuous sutureing suprahepatic inferior vena cavum(SHVC) to observe the time of surgical procedures, postoperative complications and recepients survival rate.
     2. Establishment of rats orthotopic liver transplantation with acute rejection models: according to the different donor-recipient combinations,the research animals were divided into three groups: SD→SD group (Isograft group), SD→Wistar group and DA→Lewis groups, each group with 32 cases. Six rats killed randomly to observe the changes in blood liver function (ALT and TBIL) and liver pathological changes in 3, 5, 7, 10 days respectively after transplantation. The residual rats were retained for survival analysis.
     3. Peritoneal catheter was retented after rats orthotopic liver transplantation with acute rejection model. E. coli ATCC25922 was perfused through the tube 3 days and 5 days posttransplant. The research groups were randomly divided into five infection groups, including 2×108cfu/ml, 2×107cfu/ml, 5×106cfu/ml, 1×106cfu/ml, 5×105cfu/ml, and one control group (perfused sterile saline 2ml intraperitoneally) at each time point. Rats were sacrificed in 1 day before perfusion, 1 day, 3 days, 5 days, and 7 days after perfusion resceptively, each time point with four rats, another eight rats involved in each group was retained for survival analysin. To observed recepients’general condition, ascites bacterial culture and leukocyte count, peripheral blood white blood cell count, TCO2, HCO3-, PH, ALT, TB, as well as liver and kidney lung HE staining to evaluate the status and severity of infection.
     4. Used DA or LEW rats as donors and LEW rats as recepients, established rat liver transplantation model. Based on whether induction of infection or no, the experimental animals were divided into four groups: G1 group (LEW→LEW, non-infection group), G2 group (LEW→LEW, infection group), G3 group (DA→LEW, non-infection group), G4 group (DA→LEW, infection group). The bacteria were perfused through peritoneal catheter with concentration of 1×106cfu/mL three day after transplantation. Rats were sacrificed in 1 day before perfusion, 1 day, 3 days, and 5 days after perfusion resceptively, each time point with four rats. Through three means: liver pathology, CXCR3 and its ligand CXCL10 changes detected by immunohistochemistry and fluorescence quantitative PCR, and apoptosis of liver cells assayed by TUNEL to evaluate the change of the graft immune rejection responses.
     5. Experimental animal groups, disposition, and time of specimen collection were similar with step 4. The proportion of peripheral blood T lymphocyte subsets and its function were identified by flow cytometry (FCM) and one-way mixed lymphocyte culture respectively. Serum cytokine changes were detected by ELISA to evaluated the T lymphocyte subsets, function and differentiation after bacterial infection.
     6. Experimental animal groups, disposition, and time of specimen collection were similar with step 4. The spleen dendritic cells(DCs) of recepients were isolated freshly by magnetic cell sorting technique. Their phenotype and function were detected by flow cytometry and one-way mixed lymphocyte culture to evaluate the effects on spleen DCs by bacterial infection on recepients.
     7. Used DA rats as donors and LEW rats as recepients to established rat liver transplantation model. According to whether induction of infection and/or drug intervention or no, rats were divided into four groups: G1 group was the control group, G2 group was drug intervention based on G1, G3 group was infected group, G4 group was drug intervention based on G3. Infection-induced method was similar to the step4. Thymosinα1 with a dose of 2mg was injected into the abdominal cavity in a one-time six days after transplantataion (72h after infection). Rats were sacrificed 1 day pre-infection and 2 days after drug intervention (eight days posttransplantation) respectively. Each time point and each group included four rats. Six rats were retained to survival analysis in each group. The general conditions and survival rate were observed. The pathological changes in liver grafts, the proportion of peripheral blood T lymphocyte subsets and their functional changes were inditified to evaluate the effection of immune regulation on rat liver transplantation with intra-abdominal bacteria infection.
     8. The data were analyzed through SPSS16.0 software. One-way ANOVA, t test, rank sum test methods and Kaplan-Meier survival analysis were applied. P<0.05 was considered statistically significant.
     Results
     1. In the stabile stage of rat orthotopic liver transplantation, the time of the main steps of operation were as follow: donor operation 30.2±2.5min, donator liver trimming 5.7±1.6 min, suprahepatic inferior vena cava(SHVC) anastomosis 9.1±0.8 min, portal vein (PV) reconstruction 1.6±0.5 min, infrahepatic vena cava(IHVC) reconstruction of 1.5±0.4 min, anhepatic period 15.4±1.1min, biliary duct reconstruction 1.1±0.3min, recepients operation 39.5±1.4 min. Surgical success rate was 100%, one-week survival rate was 97.7%, late complications such as biliary obstruction incidence was 3.3%.
     2. The survival time of SD→SD group was more than 120d; the median survival time of SD→Wistar group was 100d, its 95%CI was (83.369~116.631)d, no statistical difference was found between the two groups(P=0.317). The median survival time of DA→Lewis Group was 12d, its 95%CI was (10.125 ~13.848)d, the difference had remarkable statistical significance (vs SD→Wistar group, P=0.000). In regard to serum ALT and TBIL levels, downward trend was found in SD→SD group gradually, the same was seen in SD→Wistar group but slower during the first five days posttransplant, while in DA→Lewis Group they increased progressively. At 10 days after transplantation, ALT and TBIL presented SD-SD < SD-Wistar < DA-Lewis simultaneously, (58.38±12.44 vs 100.96±16.50 vs 1162.25±84.34) and (15.99±5.92 vs 76.65±9.45 vs 175.06±16.67) respectively, the difference all had statistical significance (DA→Lewis Group vs SD→SD group and SD→Wistar group, P<0.05). There was no acute rejection episodes in SD→SD group with rejection activity index (RAI) score 1 to 2 score. The same representations was found in SD→Wistar group except that 1 rat showed severe acute rejection with RAI 8 score at 10 days postoperation. In DA→Lewis group, acute rejection episodes appeared in some rats at three days after operation and the pathological grading of damages all intensified quickly with RAI score 8 to 9 at 10 days posttransplant. And the result of Kruskal-Wallis H test in day 10 showed:χ2=20.107,ν=2, P=0.000, obviously statistical significance was found between three groups.
     3. The survival time were extremely short in all intra-abdominal injection groups induced in 5d postoperative (mostly of the 1~3 days after infection). Among the intra-abdominal injection groups induced in 3d posttransplant, the survival time after infection were most of 2~3 days in G1, G2 and G3 group, 5~9 days in G4, 9~10 days in G5. The result of Log-Rank analysis were follow: (χ2=13.322,ν=1,P = 0.000, G4 vs G5), (χ2=11.829,ν=1,P = 0.001,G4 vs G0) and (χ2=0.013,ν=1,P = 0.909,G5 vs G0). In regard to ascites white blood cell count, a continuously increased trend was found in G4, while G5 showed increased firstly and then decreased. Remarkly statistical difference was found with P<0.01 in 7 days after infection (G4 vs G5). Ascites bacterial culture was negative in G5 group during the late phase of infection, it might be that the body resisted bacterial spontaneously. While in G4, it was continuously positvie. As time goes by, peripheral WBC, ALT and TBIL increased continuously, PH, HCO3- and TCO2 value decreased, showing serious acid-base balance disturbances and abnormal liver function in G4 group. Statistical difference was found with P<0.05 in end point of study (G4 vs G0). There were abnormalities during earlier phase of infection in G5, but normalities during later phase, statistical difference was also found with P<0.05 in end point of study (G4 vs G5). HE staining showed that kidneys and lung were normal in pathology in each group at the end of the observation.
     4. HE staining showed that all the four rats presented severe acute rejection in G3 group. While only one recepient showed severe acute rejection pathologically in G4 group, but parenchyma of liver graft was damaged more seriously than G3. The RAI score of this two groups were (8.0±0.8 vs 6.5±1.3,P<0.05). In regard to CXCL-10 and its ligand CXCR-3 either in immunohistochemistry results or the relative expression of mRNA levels, a continuously increased trend was found in G3 group, while G4 showed increased firstly and then decreased. statistical difference was found with P<0.05 in end point of study (G4 vs G3). The amount of apoptotic cells in liver graft increased continuously in all groups, they distributed mainly in the hepatic parenchyma in G3 but mainly in portal area in G4. Statistical difference was found with P<0.05 in terminal point of study (G4 vs G3).
     5. The result of one way MLR showed that T lympholeukocyte function elevated slightly in G1, increased in G2, elevated remarkably in G3, increased constituously in G4. The increased magnitude of G2 and G4 were lower than G3. Statistical differences were found between G1, G2 and G3 (0.3727±0.05479 vs 0.7795±0.00995 vs 0.9550±0.03594, P<0.05) at day 5 after infection. The difference was seen between G3 and G4 (0.7560±0.00787 vs 0.9550±0.03594, P<0.05). Correspondingly, the trends of T lympholeukocyte subpopulation and CD4+/CD8+ ratio in various group was different. No obvious changes were found in G1 group. Rising trend was found in G2 group except day 5 postinfection. The subpopulations of T lympholeukocyte decreased in the first three time point in G3 except day 8, the CD4+/CD8+ ratio elevated continuously ( day 8 vs day 2, P<0.05). Obvious decreased trend of CD4+ and CD8+ T cells and CD4+/CD8+ were seen in G4 with a ratio of 1.43~1.46 at day 5 after infection (P <0.05, vs G1 and G3). Regarding to serum cytokines, the trends of IL-10 level were unchanged in G1, mildly elevated in G2, significantly reduced in G3, and remarkably elevated in G4. Pairwise comparisons between groups differences were obviously statistical significant (P <0.01) in day 5 after infection. Contrastively, IFN-γlevel was significantly increased in G3 group (and acute rejection occurred synchronously), but reduced notedly in G2 and G4 at day 5 after infection (vs day 3, P<0.01). Pairwise comparisons between groups differences were remarkably statistical significant (P<0.01) in day 5 after infection. Another serum cytokines we examined was IL-12, the trends of it in all experimental groups were similar to IFN-γlevel, but the level decreased more significantly in day 5 post-infection in G4 than in G2 (P<0.05).
     6. Rats spleen DCs were with typical dendritic features isolated using immunomagnetic beads freshly. About 1.5~3×106 cells were available from each spleen. The expression trends of MHC-Ⅱ, CD80 and CD86 remained almost coherence at all time points in all group. G1 was high expression postoperative, G2 group decreased after infection, G3 increased continuously during the whole process, the expression levels in G4 were significantly lower, DCs oriented toward immature. Pairwise comparisons between groups differences were significantly statistical significant (P<0.01) in day 8 after surgery. Differences were all statistical significant on three surface molecules in G2 and G4 groups at day 8 vs day 2 posttransplant (P<0.05). The same results were seen in G3 except CD80. In regard to the function (to stimulate allogeneic T cell proliferation capacity) of DCs at day 8 after operation, decreased in G4 group but increased in G3 group (0.27±0.05 vs 0.93±0.02, P<0.01).
     7. The median survival time were 12 days in both G1 and G2 group, 95% CI was (10.400~13.600) and (10.868 ~13.132), respectively, the difference was not statistical significant (P>0.05). The median survival time were 9 days (95% CI, 7.400~10.600) in G3 and 10 days (95% CI, 8.868~11.132) in G4, the difference was not statistical significant (P>0.05). At the terminal of research, HE staining result showed severe acute rejection with RAI score of 8~9 in G1 and G2, and moderate to severe acute rejection with RAI score of 6~8 in G3 and G4. Difference was not statistical significant (P>0.05) between G3 and G4. The subsets of T cells and CD4+/CD8+ ratio both increased in treatment groups G2 (vs G1) and G4 (vs G3) (P<0.05).
     Conclusions
     1. A high survival rate, stable and reliable closed colony rat orthotopic liver transplantation was established in this study. Futhermore, a inbred rat orthotopic liver transplantation with acute rejection model was successful established too.The results were stable, reliable, reproducible, can be used for basic research in related fields.
     2. The model of intraabdominal infection induced by E. coli after liver transplantation in the inbred rat was feasible and reliable and repeatable with following advantages e.g. more precise confirmation of the existence of intraabdominal infection and reducement of trauma to the rats. The model could meet the needs for experimental study.
     3. Intraperitoneal bacterial infection after liver transplantation in the rats worsened the hepatic parenchymal damage in some extent, but released the pathological manifestations of acute rejection of allograft partly. The decreased expression of chemokine CXCL-10 and its receptor CXCR-3 in the late stage of infection, as well as the augmentation of apoptosis of infiltrating lymphocytes around the centrilobular vein and periportal space of allograft may be one of the causes contributed to the improvement of liver acute rejection.
     4. The T lymphocyte function increased during the early stage of acute rejection and/or infection. It reduced significantly in the late phase of acute rejection accompanied with infection. The ratio of CD4+/CD8+ T cells increased in the acute rejection model, and increased during the early stage of infection model but decreased in the later period, and it dropped dramatically in the infection folling acute rejection model. The levels of serum IFN-γelevated when acute rejection occurred; the serum levels of IL-10 increased in the infection model. The synergistic action of these two factors promoted the change from Th1 cells to Th2 cells. Serum IL-12 secretion reduction may have contributed to this change. Anti-inflammatory response and systemic immunosuppression took the place during this time.
     5. The spleen DCs presented rapid mature and full function to stimulate allogeneic T cell proliferation during early stage of acute rejection and/or infection. But it was immaturing and lost the function during the late phase of infection, in particular under the comorbid state of infection and rejection, which may be involved in local and systemic immune suppression of the recepients.
     6. Immune regulating treatment did not significantly prolong the survival time of recepients and not augmented acute rejection during the short-term of rats liver transplantation complicated with abdominal bacterial infection. But it improved the subproportions, ratio and function of T lymphocyte according to our small size samples. A large scale of sample size was needed to enhance the reliability of this results.
引文
[1] Safdar N, Said A, Lucey MR. The role of selective digestive decontamination for reducing infection in patients undergoing liver transplantation: a systematic review and meta-analysis [J]. Liver Transpl, 2004, 10(7): 817-827.
    [2] Echaniz A, Pita S, Otero A, et al. [Incidence, risk factors and influence on survival of infectious complications in liver transplantation] [J]. Enferm Infecc Microbiol Clin, 2003, 21(5): 224-231.
    [3] Hollenbeak CS, Alfrey EJ, Souba WW. The effect of surgical site infections on outcomes and resource utilization after liver transplantation [J]. Surgery, 2001, 130(2): 388-395.
    [4] Toniutto P FE, Caldato M. Favourable outcome of adefovir-dipivoxil treatment in acute de novo hepatitis B after liven ransplantation [J]. Transplantation, 2004, 77(472-473.
    [5]钟林,彭志海.肝移植术后细菌感染的防治[J].肝胆胰外科杂志, 2007, 19(1): 4.
    [6]张栋,张忠涛,刘建,等.肝移植术后的细菌感染[J].中华肝胆外科杂志, 2004, 6(5): 297-299.
    [7]汪雅萍,应春妹,张翻曼,等.肝移植患者术后感染细菌分布及耐药性比较[J].检验医学, 2007, 22(33): 351.
    [8] Lautenschlager I, Hockerstedt K, Jalanko H, et al. Persistent cytomegalovirus in liver allografts with chronic rejection [J]. Hepatology, 1997, 25(1): 190-194.
    [9] Toupance O, Bouedjoro-Camus MC, Carquin J, et al. Cytomegalovirus-related disease and risk of acute rejection in renal transplant recipients: a cohort study with case-control analyses [J]. Transpl Int, 2000, 13(6): 413-419.
    [10] Li Y,Yan H XW. Allograft rejection-related gene expression in the endothelial cells of renal transplantation recipients after cytomegalovirus infection. [J]. J Zhejiang Univ Sci B, 2009, 10(11): 820-828.
    [11] Tamura K, Oka T, Ohsawa K, et al. Allogeneic cell stimulation enhances cytomegalovirus replication in the early period of primary infection in an experimental rat model [J]. J Heart Lung Transplant, 2003, 22(4): 452-459.
    [12] Jazrawi SF, Zaman A, Muhammad Z, et al. Tumor necrosis factor-alpha promoter polymorphisms and the risk of rejection after liver transplantation: a case control analysis of 210 donor-recipient pairs [J]. Liver Transpl, 2003, 9(4): 377-382.
    [13]丁隆,杨宇,董家鸿.大鼠肝移植术后腹腔细菌感染对移植肝免疫排斥反应的影响[J].第三军医大学学报, 2007, 29(11): 1072-1075.
    [14] American College of Chest Physicians/Society of Critical Care Medicine Consensus Conference: definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis [J]. Crit Care Med, 1992, 20(864-874.
    [15] Bone RC. Sir Isaac Newton, sepsis, SIRS, and CARS [J]. Crit Care Med, 1996, 24(7): 1125-1128.
    [16] Bone RC. Immunologic dissonance: a continuing evolution in our understanding of the systemic inflammatory response syndrome (SIRS) and the multiple organ dysfunction syndrome (MODS) [J]. Ann Intern Med, 1996, 125(8): 680-687.
    [17] Friedman G, Jankowski S, Marchant A, et al. Blood interleukin 10 levels parallel the severity of septic shock [J]. J Crit Care, 1997, 12(4): 183-187.
    [18] Giamarellos-Bourboulis EJ, Tsaganos T, Spyridaki E, et al. Early changes of CD4-positive lymphocytes and NK cells in patients with severe Gram-negative sepsis [J]. Crit Care, 2006, 10(6): R166.
    [19] Bach FH AH. Transplantation Immunology, 1995, Wiley & Sons
    [20] Mosmann TR, Schumacher JH, Fiorentino DF, et al. Isolation of monoclonal antibodies specific for IL-4, IL-5, IL-6, and a new Th2-specific cytokine (IL-10), cytokine synthesis inhibitory factor, by using a solid phase radioimmunoadsorbent assay [J]. J Immunol, 1990, 145(9): 2938-2945.
    [21] Kubin M, Kamoun M, Trinchieri G. Interleukin 12 synergizes with B7/CD28 interaction in inducing efficient proliferation and cytokine production of human T cells [J]. J Exp Med, 1994, 180(1): 211-222.
    [22] Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity [J]. Nat Rev Immunol, 2003, 3(2): 133-146.
    [23]邱文洪,王国华,朱慧芬,等. hIL-10修饰树突状细胞对实验动物淋巴细胞增殖及胞毒效应的影响[J].中国免疫学杂志, 2003, 19(8): 527-529.
    [24] Roelen DL, Schuurhuis DH, van den Boogaardt DE, et al. Prolongation of skin graft survival by modulation of the alloimmune response with alternatively activated dendritic cells [J]. Transplantation, 2003, 76(11): 1608-1615.
    [25] Thai NL, Fu F, Qian S, et al. Cytokine mRNA profiles in mouse orthotopic liver transplantation. Graft rejection is associated with augmented TH1 function [J]. Transplantation, 1995, 59(2): 274-281.
    [26] Coito AJ, Shaw GD, Li J, et al. Selectin-mediated interactions regulate cytokine networks and macrophage heme oxygenase-1 induction in cardiac allograft recipients [J]. Lab Invest, 2002, 82(1): 61-70.
    [27] Lin ML, Zhan Y, Nutt SL, et al. NK cells promote peritoneal xenograft rejection through an IFN-gamma-dependent mechanism [J]. Xenotransplantation, 2006, 13(6): 536-546.
    [28] Zhang XG, Lu Y, Wang B, et al. Cytokine production during the inhibition of acute vascular rejection in a concordant hamster-to-rat cardiac xenotransplantation model [J]. Chin Med J (Engl), 2007, 120(2): 145-149.
    [29] Trinchieri G. Interleukin-12: a proinflammatory cytokine with immunoregulatory functions that bridge innate resistance and antigen-specific adaptive immunity [J]. Annu Rev Immunol, 1995, 13(251-276.
    [30] Szabo SJ, Dighe AS, Gubler U, et al. Regulation of the interleukin (IL)-12R beta 2 subunit expression in developing T helper 1 (Th1) and Th2 cells [J]. J Exp Med, 1997, 185(5): 817-824.
    [31] Steinman RM. Dendritic cells and the control of immunity: enhancing the efficiency of antigen presentation [J]. Mt Sinai J Med, 2001, 68(3): 160-166.
    [32] Banchereau J, Steinman RM. Dendritic cells and the control of immunity [J]. Nature, 1998, 392(6673): 245-252.
    [33] Rifle G, Mousson C. Dendritic cells and second signal blockade: a step toward allograft tolerance? [J]. Transplantation, 2002, 73(1 Suppl): S1-2.
    [34] Morelli AE, Thomson AW. Dendritic cells: regulators of alloimmunity and opportunities for tolerance induction [J]. Immunol Rev, 2003, 196(125-146.
    [35] Hotchkiss RS, Tinsley KW, Swanson PE, et al. Depletion of dendritic cells, but not macrophages, in patients with sepsis [J]. J Immunol, 2002, 168(5): 2493-2500.
    [36] Itano AA, McSorley SJ, Reinhardt RL, et al. Distinct dendritic cell populations sequentially present antigen to CD4 T cells and stimulate different aspects of cell-mediated immunity [J]. Immunity, 2003, 19(1): 47-57.
    [37] Mazariegos GV, Zahorchak AF, Reyes J, et al. Dendritic cell subset ratio in peripheral blood correlates with successful withdrawal of immunosuppression in liver transplant patients [J]. Am J Transplant, 2003, 3(6): 689-696.
    [38] Jain A, Reyes J, Kashyap R, et al. Long-term survival after liver transplantation in 4,000 consecutive patients at a single center [J]. Ann Surg, 2000, 232(4): 490-500.
    [39] Casas-Melley AT, Falkenstein KP, Flynn LM, et al. Improvement in renal function and rejection control in pediatric liver transplant recipients with the introduction of sirolimus [J]. Pediatr Transplant, 2004, 8(4): 362-366.
    [40]蔡志仕江艺,蔡秋程,等.肝移植术后停用免疫抑制剂59天一例报道[J].中国现代普通外科进展2008, 11(4): 367-368.
    [41] Boehnert MU, Armbruster FP, Hilbig H. Relaxin as a protective substance in preservation solutions for organ transplantation, as shown in an isolated perfused rat liver model [J]. Transplant Proc, 2008, 40(4): 978-980.
    [42]王宇周,蒋晓青.丹参对原位肝移植大鼠供肝冷保存再灌注损伤的保护作用[J].中国组织工程研究与临床康复, 2008, 12(5): 801-804.
    [43] Cao H, Liu H, Wu ZY. Effects of combined immune therapy on survival and Th1/Th2 cytokine balance in rat orthotopic liver transplantation [J]. Chin Med J (Engl), 2007, 120(20): 1809-1812.
    [44] Chen Y, Chen J, Liu Z, et al. Relationship between TH1/TH2 cytokines and immune tolerance in liver transplantation in rats [J]. Transplant Proc, 2008, 40(8): 2691-2695.
    [45] Lee S, Charters AC, Chandler JG, et al. A technique for orthotopic liver transplantation in the rat [J]. Transplantation, 1973, 16(6): 664-669.
    [46] Miyata M, Fischer JH, Fuhs M, et al. A simple method for orthotopic liver transplantation in the rat. Cuff technique for three vascular anastomoses [J].Transplantation, 1980, 30(5): 335-338.
    [47] Kamada N, Calne RY. Orthotopic liver transplantation in the rat. Technique using cuff for portal vein anastomosis and biliary drainage [J]. Transplantation, 1979, 28(1): 47-50.
    [48] Kamada N, Calne RY. A surgical experience with five hundred thirty liver transplants in the rat [J]. Surgery, 1983, 93(1 Pt 1): 64-69.
    [49] Kashfi A, Mehrabi A, Pahlavan PS, et al. A review of various techniques of orthotopic liver transplantation in the rat [J]. Transplant Proc, 2005, 37(1): 185-188.
    [50] Miko I, Brath E, Furka I. Basic teaching in microsurgery [J]. Microsurgery, 2001, 21(4): 121-123.
    [51] Krishnan KG, Dramm P, Schackert G. Simple and viable in vitro perfusion model for training microvascular anastomoses [J]. Microsurgery, 2004, 24(4): 335-338.
    [52] Klein I, Steger U, Timmermann W, et al. Microsurgical training course for clinicians and scientists at a German University hospital: a 10-year experience [J]. Microsurgery, 2003, 23(5): 461-465.
    [53] Schemmer P, Schoonhoven R, Swenberg JA, et al. Gentle in situ liver manipulation during organ harvest decreases survival after rat liver transplantation: role of Kupffer cells [J]. Transplantation, 1998, 65(8): 1015-1020.
    [54] Kebis A KM, Grancic P, et al. A novel way of liver preservation improves rat iver viability upon reperfusion [J]. J Zhejiang Univ Sci B, 2007, 8(5): 289-295.
    [55] Guo H, Wu YJ, Zheng SS, et al. Application of modified two-cuff technique and multiglycosides tripterygium wilfordii in hamster-to-rat liver xenotransplant model [J]. World J Gastroenterol, 2003, 9(7): 1550-1553.
    [56] Tokunaga Y, Ozaki N, Wakashiro S, et al. Effects of perfusion pressure during flushing on the viability of the procured liver using noninvasive fluorometry [J]. Transplantation, 1988, 45(6): 1031-1035.
    [57] Tan F, Chen Z, Zhao Y, et al. Novel technique for suprahepatic vena cava reconstruction in rat orthotopic liver transplantation [J]. Microsurgery, 2005, 25(7): 556-560.
    [58] Jia C, Wang W, Zhu Y, et al. Suprahepatic vena cava manipulative bleedingalleviates hepatic ischemia-reperfusion injury in rats [J]. Dig Liver Dis, 2008, 40(4): 285-292.
    [59] Pan TL, Wang PW, Huang CC, et al. Expression, by functional proteomics, of spontaneous tolerance in rat orthotopic liver transplantation [J]. Immunology, 2004, 113(1): 57-64.
    [60] Suzuki A, Kudoh S, Mori K, et al. Expression of nitric oxide and inducible nitric oxide synthase in acute renal allograft rejection in the rat [J]. Int J Urol, 2004, 11(10): 837-844.
    [61] Banff schema for grading liver allograft rejection: an international consensus document [J]. Hepatology, 1997, 25(3): 658-663.
    [62]陈忠华.同种肝脏移植免疫特惠器官现象及其临床意义[J].中华肝脏病杂志, 2005, 13(3): 221.
    [63] Bell D, Young JW, Banchereau J. Dendritic cells [J]. Adv Immunol, 1999, 72(255-324.
    [64] Bilezikci B, Demirhan B, Kocbiyik A, et al. Relevant histopathologic findings that distinguish acute cellular rejection from cholangitis in hepatic allograft biopsy specimens [J]. Transplant Proc, 2008, 40(1): 248-250.
    [65] Zimmermann FA, Butcher GW, Davies HS, et al. Techniques for orthotopic liver transplantation in the rat and some studies of the immunologic responses to fully allogeneic liver grafts [J]. Transplant Proc, 1979, 11(1): 571-577.
    [66] Adachi K, Fujino M, Kitazawa Y, et al. Exogenous expression of Fas-ligand or CrmA prolongs the survival in rat liver transplantation [J]. Transplant Proc, 2006, 38(8): 2710-2713.
    [67] Yamamoto S, Okuda T, Yamasaki K, et al. FK778 controls acute rejection after rat liver allotransplantation showing positive interaction with FK506 [J]. Transplant Proc, 2005, 37(1): 126-129.
    [68] Cordoba SP, Wang C, Williams R, et al. Gene array analysis of a rat model of liver transplant tolerance identifies increased complement C3 and the STAT-1/IRF-1 pathway during tolerance induction [J]. Liver Transpl, 2006, 12(4): 636-643.
    [69] Jia C, Zheng S, Zhu Y. Intrathymic inoculation of liver specific antigen alleviatesliver transplant rejection [J]. Chin Med Sci J, 2004, 19(1): 38-43.
    [70] Jiang GP, Hu ZH, Zheng SS, et al. Adenovirus mediated CTLA4Ig gene inhibits infiltration of immune cells and cell apoptosis in rats after liver transplantation [J]. World J Gastroenterol, 2005, 11(7): 1065-1069.
    [71] Nakano T, Lai CY, Goto S, et al. Role of antinuclear antibodies in experimental and clinical liver transplantation [J]. Transplant Proc, 2006, 38(10): 3605-3606.
    [72]姜明山韩.不同品系大鼠之间原位肝移植的实验观察[J].中华实验外科杂志, 2004, 21(4): 428-429.
    [73]浦立勇姚,李相成,等. DA到Lewis大鼠肝脏移植急性排斥模型的建立[J].第四军医大学学报, 2006, 27(12): 1071-1073.
    [74]冉崇福窦,等. DA→LEW大鼠肝脏急性排斥反应模型的技术改进与评价[J].西南国防医药, 2007, 17(3): 278-280.
    [75]姚坤厚高,等.大鼠原位肝移植急性排斥模型的建立[J].实用医学杂志, 2008, 24(8): 1305-1307.
    [76]周建党朱,郭建军,等.肝移植术后感染病原菌耐药性分析[J].中华器官移植杂志, 2005, 26(8): 455-457.
    [77] Singh N, Paterson DL, Chang FY, et al. Methicillin-resistant Staphylococcus aureus: the other emerging resistant gram-positive coccus among liver transplant recipients [J]. Clin Infect Dis, 2000, 30(2): 322-327.
    [78]鲍扬,李幼生,黎介寿.一种新型腹腔感染动物模型的建立[J].中华实验外科杂志, 2004, 21(1): 99-100.
    [79] Ikuta S, Ono S, Kinoshita M, et al. Enhanced interferon-gamma production and bacterial clearance in the liver of splenectomized mice in the models of Escherichia coli injection or intestinal obstruction [J]. Shock, 2004, 21(5): 452-457.
    [80] Suzuki K, Khanna R, Nolph KD, et al. Expected white blood cell counts and differentials in a rat model of peritoneal dialysis [J]. Perit Dial Int, 1995, 15(2): 142-146.
    [81] Singh N. Infectious diseases in the liver transplant recipient [J]. Semin Gastrointest Dis, 1998, 9(3): 136-146.
    [82]张栋,张忠涛,刘建等.肝移植术后的细菌感染[J].中华肝胆外科杂志, 2004, 10(5): 297-300.
    [83] Hubscher S. Diagnosis and grading of liver allograft rejection: a European perspective [J]. Transplant Proc, 1996, 28(1): 504-507.
    [84]孙强朱,何晓顺,等.暂停免疫抑制剂对肝移植术后严重感染疗效的探讨[J].中华普通外科杂志2007, 22(9): 650-652.
    [85] Hetts SW. To die or not to die: an overview of apoptosis and its role in disease [J]. JAMA, 1998, 279(4): 300-307.
    [86] Qian S, Lu L, Fu F, et al. Apoptosis within spontaneously accepted mouse liver allografts: evidence for deletion of cytotoxic T cells and implications for tolerance induction [J]. J Immunol, 1997, 158(10): 4654-4661.
    [87] Qian S, Lu L, Li Y, et al. Apoptosis of graft-infiltrating cytotoxic T cells: a mechanism underlying "split tolerance" in mouse liver transplantation [J]. Transplant Proc, 1997, 29(1-2): 1168-1169.
    [88] Lazzeri E, Rotondi M, Mazzinghi B, et al. High CXCL10 expression in rejected kidneys and predictive role of pretransplant serum CXCL10 for acute rejection and chronic allograft nephropathy [J]. Transplantation, 2005, 79(9): 1215-1220.
    [89]陈国栋,刘玉兰.趋化因子I P-10和M I P-3α在肝移植大鼠肝脏中的表达研究[J].中国现代医学杂志, 2005, 15(17): 2594-2996.
    [90] Hall BM. Cells mediating allograft rejection [J]. Transplantation, 1991, 51(6): 1141-1151.
    [91] Charo IF, Ransohoff RM. The many roles of chemokines and chemokine receptors in inflammation [J]. N Engl J Med, 2006, 354(6): 610-621.
    [92] Kuschert GS, Coulin F, Power CA, et al. Glycosaminoglycans interact selectively with chemokines and modulate receptor binding and cellular responses [J]. Biochemistry, 1999, 38(39): 12959-12968.
    [93] Gattass CR, King LB, Luster AD, et al. Constitutive expression of interferon gamma-inducible protein 10 in lymphoid organs and inducible expression in T cells and thymocytes [J]. J Exp Med, 1994, 179(4): 1373-1378.
    [94] Luster AD, Ravetch JV. Biochemical characterization of a gammainterferon-inducible cytokine (IP-10) [J]. J Exp Med, 1987, 166(4): 1084-1097.
    [95] Vanguri P, Farber JM. Identification of CRG-2. An interferon-inducible mRNA predicted to encode a murine monokine [J]. J Biol Chem, 1990, 265(25): 15049-15057.
    [96] Ohmori Y, Hamilton TA. A macrophage LPS-inducible early gene encodes the murine homologue of IP-10 [J]. Biochem Biophys Res Commun, 1990, 168(3): 1261-1267.
    [97] Melter M, Exeni A, Reinders ME, et al. Expression of the chemokine receptor CXCR3 and its ligand IP-10 during human cardiac allograft rejection [J]. Circulation, 2001, 104(21): 2558-2564.
    [98] Agostini C, Calabrese F, Rea F, et al. Cxcr3 and its ligand CXCL10 are expressed by inflammatory cells infiltrating lung allografts and mediate chemotaxis of T cells at sites of rejection [J]. Am J Pathol, 2001, 158(5): 1703-1711.
    [99] Hancock WW, Gao W, Csizmadia V, et al. Donor-derived IP-10 initiates development of acute allograft rejection [J]. J Exp Med, 2001, 193(8): 975-980.
    [100] Loetscher M, Gerber B, Loetscher P, et al. Chemokine receptor specific for IP10 and mig: structure, function, and expression in activated T-lymphocytes [J]. J Exp Med, 1996, 184(3): 963-969.
    [101] Zhai Y, Shen XD, Hancock WW, et al. CXCR3+CD4+ T cells mediate innate immune function in the pathophysiology of liver ischemia/reperfusion injury [J]. J Immunol, 2006, 176(10): 6313-6322.
    [102] Tsuchihashi S, Zhai Y, Bo Q, et al. Heme oxygenase-1 mediated cytoprotection against liver ischemia and reperfusion injury: inhibition of type-1 interferon signaling [J]. Transplantation, 2007, 83(12): 1628-1634.
    [103] Spanaus KS, Nadal D, Pfister HW, et al. C-X-C and C-C chemokines are expressed in the cerebrospinal fluid in bacterial meningitis and mediate chemotactic activity on peripheral blood-derived polymorphonuclear and mononuclear cells in vitro [J]. J Immunol, 1997, 158(4): 1956-1964.
    [104] McLoughlin RM, Solinga RM, Rich J, et al. CD4+ T cells and CXC chemokines modulate the pathogenesis of Staphylococcus aureus wound infections [J].Proc Natl Acad Sci U S A, 2006, 103(27): 10408-10413.
    [105] Krams SM, Egawa H, Quinn MB, et al. Apoptosis as a mechanism of cell death in liver allograft rejection [J]. Transplantation, 1995, 59(4): 621-625.
    [106] Chiffoleau E, Walsh PT, Turka L. Apoptosis and transplantation tolerance [J]. Immunol Rev, 2003, 193(124-145.
    [107] Van Parijs L, Abbas AK. Homeostasis and self-tolerance in the immune system: turning lymphocytes off [J]. Science, 1998, 280(5361): 243-248.
    [108] Meyer D, Thorwarth W, Otto C, et al. Early T-cell inactivation and apoptosis-critical events for tolerance induction after allogeneic liver transplantation [J]. Transplant Proc, 2001, 33(1-2): 256-258.
    [109] Wells AD, Li XC, Li Y, et al. Requirement for T-cell apoptosis in the induction of peripheral transplantation tolerance [J]. Nat Med, 1999, 5(11): 1303-1307.
    [110] Mottram PL, Han WR, Purcell LJ, et al. Increased expression of IL-4 and IL-10 and decreased expression of IL-2 and interferon-gamma in long-surviving mouse heart allografts after brief CD4-monoclonal antibody therapy [J]. Transplantation, 1995, 59(4): 559-565.
    [111] Neuberger J. Incidence, timing, and risk factors for acute and chronic rejection [J]. Liver Transpl Surg, 1999, 5(4 Suppl 1): S30-36.
    [112]施红顾,朱玲,等..大剂量胸腺肽对恶性血液病化疗患者T细胞亚群的影响[J].临床血液学杂志, 2000, 13(3): 112-115.
    [113] Adams D. Mechanisms of liver allograft rejection in man [J]. Clin Sci (Lond), 1990, 78(4): 343-350.
    [114] Romagnani S. Human TH1 and TH2 subsets: doubt no more [J]. Immunol Today, 1991, 12(8): 256-257.
    [115] Schmitt E, Hoehn P, Huels C, et al. T helper type 1 development of naive CD4+ T cells requires the coordinate action of interleukin-12 and interferon-gamma and is inhibited by transforming growth factor-beta [J]. Eur J Immunol, 1994, 24(4): 793-798.
    [116] Weiner HL. Oral tolerance: immune mechanisms and the generation of Th3-type TGF-beta-secreting regulatory cells [J]. Microbes Infect, 2001, 3(11):947-954.
    [117] Josien R, Douillard P, Guillot C, et al. A critical role for transforming growth factor-beta in donor transfusion-induced allograft tolerance [J]. J Clin Invest, 1998, 102(11): 1920-1926.
    [118] Yang JS, Xu LY, Xiao BG, et al. Laquinimod (ABR-215062) suppresses the development of experimental autoimmune encephalomyelitis, modulates the Th1/Th2 balance and induces the Th3 cytokine TGF-beta in Lewis rats [J]. J Neuroimmunol, 2004, 156(1-2): 3-9.
    [119] Wang YL, Tang ZQ, Gao W, et al. Influence of Th1, Th2, and Th3 cytokines during the early phase after liver transplantation [J]. Transplant Proc, 2003, 35(8): 3024-3025.
    [120] Minguela A, Torio A, Marin L, et al. Implication of Th1, Th2, and Th3 cytokines in liver graft acceptance [J]. Transplant Proc, 1999, 31(1-2): 519-520.
    [121]金伯泉.白细胞分化抗原的应用.细胞和分子免疫学[M], 2001,第二版,科学出版社(29-31.
    [122] Ji SM, Li LS, Sun QQ, et al. Immunoregulation of thymosin alpha 1 treatment of cytomegalovirus infection accompanied with acute respiratory distress syndrome after renal transplantation [J]. Transplant Proc, 2007, 39(1): 115-119.
    [123] Bradley JA, Bolton EM, Pettigrew G. Monitoring T cell alloreactivity after organ transplantation [J]. Clin Exp Immunol, 2005, 142(2): 229-232.
    [124]尤鹏,刘玉兰,陈国栋.大鼠肝移植自发免疫耐受模型T淋巴细胞亚群的变化[J].中华肝脏病杂志, 2005, 13(9): 698-699.
    [125] Calne RY, Sells RA, Pena JR, et al. Induction of immunological tolerance by porcine liver allografts [J]. Nature, 1969, 223(5205): 472-476.
    [126] Kamada N, Shinomiya T. Clonal deletion as the mechanism of abrogation of immunological memory following liver grafting in rats [J]. Immunology, 1985, 55(1): 85-90.
    [127] Mathew JM, Marsh JW, Susskind B, et al. Analysis of T cell responses in liver allograft recipients. Evidence for deletion of donor-specific cytotoxic T cells in the peripheral circulation [J]. J Clin Invest, 1993, 91(3): 900-906.
    [128] Meyer D, Baumgardt S, Loeffeler S, et al. Apoptosis of T lymphocytes in liver and/or small bowel allografts during tolerance induction [J]. Transplantation, 1998, 66(11): 1530-1536.
    [129] Wang SD, Huang KJ, Lin YS, et al. Sepsis-induced apoptosis of the thymocytes in mice [J]. J Immunol, 1994, 152(10): 5014-5021.
    [130] Ayala A, Herdon CD, Lehman DL, et al. The induction of accelerated thymic programmed cell death during polymicrobial sepsis: control by corticosteroids but not tumor necrosis factor [J]. Shock, 1995, 3(4): 259-267.
    [131] Hotchkiss RS, Tinsley KW, Swanson PE, et al. Prevention of lymphocyte cell death in sepsis improves survival in mice [J]. Proc Natl Acad Sci U S A, 1999, 96(25): 14541-14546.
    [132] de Waal Malefyt R, Haanen J, Spits H, et al. Interleukin 10 (IL-10) and viral IL-10 strongly reduce antigen-specific human T cell proliferation by diminishing the antigen-presenting capacity of monocytes via downregulation of class II major histocompatibility complex expression [J]. J Exp Med, 1991, 174(4): 915-924.
    [133] Moore KW, de Waal Malefyt R, Coffman RL, et al. Interleukin-10 and the interleukin-10 receptor [J]. Annu Rev Immunol, 2001, 19(683-765.
    [134] Maloy KJ, Salaun L, Cahill R, et al. CD4+CD25+ T(R) cells suppress innate immune pathology through cytokine-dependent mechanisms [J]. J Exp Med, 2003, 197(1): 111-119.
    [135] Banchereau J, Briere F, Caux C, et al. Immunobiology of dendritic cells [J]. Annu Rev Immunol, 2000, 18(767-811.
    [136] Lechler R, Ng WF, Steinman RM. Dendritic cells in transplantation--friend or foe? [J]. Immunity, 2001, 14(4): 357-368.
    [137] Voisine C, Hubert FX, Trinite B, et al. Two phenotypically distinct subsets of spleen dendritic cells in rats exhibit different cytokine production and T cell stimulatory activity [J]. J Immunol, 2002, 169(5): 2284-2291.
    [138] Mellman I, Steinman RM. Dendritic cells: specialized and regulated antigen processing machines [J]. Cell, 2001, 106(3): 255-258.
    [139] Liu YJ. Dendritic cell subsets and lineages, and their functions in innate andadaptive immunity [J]. Cell, 2001, 106(3): 259-262.
    [140] Shortman K. Burnet oration: dendritic cells: multiple subtypes, multiple origins, multiple functions [J]. Immunol Cell Biol, 2000, 78(2): 161-165.
    [141] Jonuleit H, Schmitt E, Schuler G, et al. Induction of interleukin 10-producing, nonproliferating CD4(+) T cells with regulatory properties by repetitive stimulation with allogeneic immature human dendritic cells [J]. J Exp Med, 2000, 192(9): 1213-1222.
    [142] Inaba K, Turley S, Iyoda T, et al. The formation of immunogenic major histocompatibility complex class II-peptide ligands in lysosomal compartments of dendritic cells is regulated by inflammatory stimuli [J]. J Exp Med, 2000, 191(6): 927-936.
    [143] Fossum S. The life history of dendritic leukocytes (DL) [J]. Curr Top Pathol, 1989, 79(101-124.
    [144] Sallusto F, Lanzavecchia A. Understanding dendritic cell and T-lymphocyte traffic through the analysis of chemokine receptor expression [J]. Immunol Rev, 2000, 177(134-140.
    [145] Mueller DL, Jenkins MK, Schwartz RH. Clonal expansion versus functional clonal inactivation: a costimulatory signalling pathway determines the outcome of T cell antigen receptor occupancy [J]. Annu Rev Immunol, 1989, 7(445-480.
    [146] Robey E, Allison JP. T-cell activation: integration of signals from the antigen receptor and costimulatory molecules [J]. Immunol Today, 1995, 16(7): 306-310.
    [147] Hathcock KS, Laszlo G, Pucillo C, et al. Comparative analysis of B7-1 and B7-2 costimulatory ligands: expression and function [J]. J Exp Med, 1994, 180(2): 631-640.
    [148] Inaba K, Witmer-Pack M, Inaba M, et al. The tissue distribution of the B7-2 costimulator in mice: abundant expression on dendritic cells in situ and during maturation in vitro [J]. J Exp Med, 1994, 180(5): 1849-1860.
    [149] de la Mata M, Riera CM, Iribarren P. Identification of a CD8alpha(+) dendritic cell subpopulation in rat spleen and evaluation of its OX-62 expression [J]. Clin Immunol, 2001, 101(3): 371-378.
    [150] Flohe SB, Agrawal H, Schmitz D, et al. Dendritic cells during polymicrobial sepsis rapidly mature but fail to initiate a protective Th1-type immune response [J]. J Leukoc Biol, 2006, 79(3): 473-481.
    [151] Fridenshtein A. [Stromal bone marrow cells and the hematopoietic microenvironment] [J]. Arkh Patol, 1982, 44(10): 3-11.
    [152] Nair R, Nyamweya N, Gonen S, et al. Influence of various drugs on the glass transition temperature of poly(vinylpyrrolidone): a thermodynamic and spectroscopic investigation [J]. Int J Pharm, 2001, 225(1-2): 83-96.
    [153] Kobayashi H, Miura S, Nagata H, et al. In situ demonstration of dendritic cell migration from rat intestine to mesenteric lymph nodes: relationships to maturation and role of chemokines [J]. J Leukoc Biol, 2004, 75(3): 434-442.
    [154] Brenan M, Puklavec M. The MRC OX-62 antigen: a useful marker in the purification of rat veiled cells with the biochemical properties of an integrin [J]. J Exp Med, 1992, 175(6): 1457-1465.
    [155] Brenan M, Rees DJ. Sequence analysis of rat integrin alpha E1 and alpha E2 subunits: tissue expression reveals phenotypic similarities between intraepithelial lymphocytes and dendritic cells in lymph [J]. Eur J Immunol, 1997, 27(11): 3070-3079.
    [156] Huang FP, Platt N, Wykes M, et al. A discrete subpopulation of dendritic cells transports apoptotic intestinal epithelial cells to T cell areas of mesenteric lymph nodes [J]. J Exp Med, 2000, 191(3): 435-444.
    [157] Allaerts W, Jeucken PH, Debets R, et al. Heterogeneity of pituitary folliculo-stellate cells: implications for interleukin-6 production and accessory function in vitro [J]. J Neuroendocrinol, 1997, 9(1): 43-53.
    [158] Rossetti M, Piccoli GB, Burdese M, et al. Tailored immunosuppression and steroid withdrawal in pancreas-kidney transplantation [J]. Rev Diabet Stud, 2004, 1(3): 129-136.
    [159] Pugin J. Sepsis and the immune response [J]. Intensive Care Med, 1999, 25(9): 1027-1028.
    [160] Kox WJ, Volk T, Kox SN, et al. Immunomodulatory therapies in sepsis [J].Intensive Care Med, 2000, 26 Suppl 1(S124-128.
    [161] Perl M, Chung CS, Garber M, et al. Contribution of anti-inflammatory/immune suppressive processes to the pathology of sepsis [J]. Front Biosci, 2006, 11(272-299.
    [162] Chen W, Frank ME, Jin W, et al. TGF-beta released by apoptotic T cells contributes to an immunosuppressive milieu [J]. Immunity, 2001, 14(6): 715-725.
    [163] Song GY, Chung CS, Schwacha MG, et al. Splenic immune suppression in sepsis: A role for IL-10-induced changes in P38 MAPK signaling [J]. J Surg Res, 1999, 83(1): 36-43.
    [164] McHugh RS, Whitters MJ, Piccirillo CA, et al. CD4(+)CD25(+) immunoregulatory T cells: gene expression analysis reveals a functional role for the glucocorticoid-induced TNF receptor [J]. Immunity, 2002, 16(2): 311-323.
    [165] June CH, Bluestone JA, Nadler LM, et al. The B7 and CD28 receptor families [J]. Immunol Today, 1994, 15(7): 321-331.
    [166] Hotchkiss RS, Osmon SB, Chang KC, et al. Accelerated lymphocyte death in sepsis occurs by both the death receptor and mitochondrial pathways [J]. J Immunol, 2005, 174(8): 5110-5118.
    [167] Li Y, Chen H, Li X, et al. A new immunomodulatory therapy for severe sepsis: Ulinastatin Plus Thymosin {alpha} 1 [J]. J Intensive Care Med, 2009, 24(1): 47-53.
    [168] Chen XM, Jiang HL, Zhou LF, et al. Immunomodulatory function of orally administered thymosin alpha1 [J]. J Zhejiang Univ Sci B, 2005, 6(9): 873-876.
    [169] Zavaglia C, Airoldi A, Pinzello G. Antiviral therapy of HBV- and HCV-induced liver cirrhosis [J]. J Clin Gastroenterol, 2000, 30(3): 234-241.
    [170] Saruc M, Ozden N, Turkel N, et al. Long-term outcomes of thymosin-alpha 1 and interferon alpha-2b combination therapy in patients with hepatitis B e antigen (HBeAg) negative chronic hepatitis B [J]. J Pharm Sci, 2003, 92(7): 1386-1395.
    [171] Rustgi V. Combination therapy of thymalfasin (thymosin-alpha 1) and peginterferon alfa-2a in patients with chronic hepatitis C virus infection who are non-responders to standard treatment [J]. J Gastroenterol Hepatol, 2004, 19(12): S76-78.
    [172]黄顺伟管,陈娟,等.免疫调理对脓毒症免疫功能和预后的作用[J].中国普通外科杂志, 2009, 18(9): 926-931.
    [173]姜军李,黎介寿.胸腺肽α1对严重腹腔感染大鼠蛋白质代谢和细胞因子的影响[J].肠外与肠内营养, 2005, 12(3): 165-169.
    [174]房林薛,周以明.胸腺肽对肠梗阻细菌移位的影响[J].中国现代医学杂志, 2000, 10(7): 27-31.
    [175] Rustgi V. Combination therapy of thymalfasin (thymosin-alpha 1) and peginterferon alfa-2a in patients with chronic hepatitis C virus infection who are non-responders to standard treatment [J]. J Gastroenterol Hepatol, 2004, 19 Suppl 6(S76-78.
    [176] Dallman MJ, Wood KJ, Hamano K, et al. Cytokines and peripheral tolerance to alloantigen [J]. Immunol Rev, 1993, 133(5-18.
    [177] Kusaka S, Grailer AP, Fechner JH, Jr., et al. Evidence for a possible Th2 bias in human renal transplant tolerance [J]. Transplant Proc, 1995, 27(1): 225-226.
    [178] Hara M, Kingsley CI, Niimi M, et al. IL-10 is required for regulatory T cells to mediate tolerance to alloantigens in vivo [J]. J Immunol, 2001, 166(6): 3789-3796.
    [179] Wisnoski N, Chung CS, Chen Y, et al. The contribution of CD4+CD25+ T-regulatory-cells to immune suppression in sepsis [J]. Shock, 2007, 27(3): 251-257.
    [1] Wieers G, Gras J, Bourdeaux C, et al. Monitoring tolerance after human liver transplantation [J]. Transpl Immunol, 2007, 17(2): 83-93.
    [2] Calne RY, Sells RA, Pena JR, et al. Induction of immunological tolerance by porcine liver allografts [J]. Nature, 1969, 223(5205): 472-476.
    [3] Knechtle SJ, Kolbeck P, Tsuchimoto S, et al. Hyperacute rejection of liver transplants in rats [J]. Curr Surg, 1986, 43(4): 303-305.
    [4] Knechtle SJ, Kolbeck PC, Tsuchimoto S, et al. Hepatic transplantation into sensitized recipients. Demonstration of hyperacute rejection [J]. Transplantation, 1987, 43(1): 8-12.
    [5] van Twuyver E, de Hoop J, ten Berge RJ, et al. Comparison of T cell responses in patients with a long-term surviving renal allograft versus a long-term surviving liver allograft. It's a different world [J]. Transplantation, 1996, 61(9): 1392-1397.
    [6] Knechtle SJ, Kwun J. Unique aspects of rejection and tolerance in liver transplantation [J]. Semin Liver Dis, 2009, 29(1): 91-101.
    [7] Zimmermann FA, Davies HS, Knoll PP, et al. Orthotopic liver allografts in the rat. The influence of strain combination on the fate of the graft [J]. Transplantation, 1984, 37(4): 406-410.
    [8] Calne R, Friend P, Moffatt S, et al. Prope tolerance, perioperative campath 1H,and low-dose cyclosporin monotherapy in renal allograft recipients [J]. Lancet, 1998, 351(9117): 1701-1702.
    [9] Calne RY. Prope tolerance--the future of organ transplantation from the laboratory to the clinic [J]. Transpl Immunol, 2004, 13(2): 83-86.
    [10] Riordan SM, Williams R. Tolerance after liver transplantation: does it exist and can immunosuppression be withdrawn? [J]. J Hepatol, 1999, 31(6): 1106-1119.
    [11] Jugie M, Canioni D, Le Bihan C, et al. Study of the impact of liver transplantation on the outcome of intestinal grafts in children [J]. Transplantation, 2006, 81(7): 992-997.
    [12] Buchman AL, Iyer K, Fryer J. Parenteral nutrition-associated liver disease and the role for isolated intestine and intestine/liver transplantation [J]. Hepatology, 2006, 43(1): 9-19.
    [13] Sun J, Sheil AG, Wang C, et al. Tolerance to rat liver allografts: IV. Acceptance depends on the quantity of donor tissue and on donor leukocytes [J]. Transplantation, 1996, 62(12): 1725-1730.
    [14] Davies HS, Pollard SG, Calne RY. Soluble HLA antigens in the circulation of liver graft recipients [J]. Transplantation, 1989, 47(3): 524-527.
    [15] Lord R, Kamada N, Kobayashi E, et al. Isolation of a 40 kDa immunoinhibitory protein induced by rat liver transplantation [J]. Transpl Immunol, 1995, 3(2): 174-179.
    [16] Sharland A, Yan Y, Wang C, et al. Evidence that apoptosis of activated T cells occurs in spontaneous tolerance of liver allografts and is blocked by manipulations which break tolerance [J]. Transplantation, 1999, 68(11): 1736-1745.
    [17] Starzl TE. Chimerism and tolerance in transplantation [J]. Proc Natl Acad Sci U S A, 2004, 101 Suppl 2(14607-14614.
    [18] Adams D. Mechanisms of liver allograft rejection in man [J]. Clin Sci (Lond), 1990, 78(4): 343-350.
    [19] Loveland B, Ceredig R, Hogarth M, et al. The key role of Lyt-1+ cells in skin graft rejection in the mouse [J]. Transplant Proc, 1981, 13(1 Pt 2): 1079-1081.
    [20] Dumble LJ, MacDonald IM, Kincaid-Smith P. Human renal allograft rejection is predicted by serial determinations of antibody-dependent cellular cytotoxicity [J].Transplantation, 1980, 29(1): 30-34.
    [21] Thai NL, Fu F, Qian S, et al. Cytokine mRNA profiles in mouse orthotopic liver transplantation. Graft rejection is associated with augmented TH1 function [J]. Transplantation, 1995, 59(2): 274-281.
    [22] Lun A, Cho MY, Muller C, et al. Diagnostic value of peripheral blood T-cell activation and soluble IL-2 receptor for acute rejection in liver transplantation [J]. Clin Chim Acta, 2002, 320(1-2): 69-78.
    [23] Boleslawski E, Conti F, Sanquer S, et al. Defective inhibition of peripheral CD8+ T cell IL-2 production by anti-calcineurin drugs during acute liver allograft rejection [J]. Transplantation, 2004, 77(12): 1815-1820.
    [24] Gibelli NE, Pinho-Apezzato ML, Miyatani HT, et al. Basiliximab-chimeric anti-IL2-R monoclonal antibody in pediatric liver transplantation: comparative study [J]. Transplant Proc, 2004, 36(4): 956-957.
    [25] Warle MC, Metselaar HJ, Hop WC, et al. Early differentiation between rejection and infection in liver transplant patients by serum and biliary cytokine patterns [J]. Transplantation, 2003, 75(1): 146-151.
    [26] Wang YL, Tang ZQ, Gao W, et al. Influence of Th1, Th2, and Th3 cytokines during the early phase after liver transplantation [J]. Transplant Proc, 2003, 35(8): 3024-3025.
    [27] Chen Y, Chen J, Liu Z, et al. Relationship between TH1/TH2 cytokines and immune tolerance in liver transplantation in rats [J]. Transplant Proc, 2008, 40(8): 2691-2695.
    [28] Coito AJ, Shaw GD, Li J, et al. Selectin-mediated interactions regulate cytokine networks and macrophage heme oxygenase-1 induction in cardiac allograft recipients [J]. Lab Invest, 2002, 82(1): 61-70.
    [29] Lin ML, Zhan Y, Nutt SL, et al. NK cells promote peritoneal xenograft rejection through an IFN-gamma-dependent mechanism [J]. Xenotransplantation, 2006, 13(6): 536-546.
    [30] Zhang XG, Lu Y, Wang B, et al. Cytokine production during the inhibition of acute vascular rejection in a concordant hamster-to-rat cardiac xenotransplantationmodel [J]. Chin Med J (Engl), 2007, 120(2): 145-149.
    [31] Mosmann TR, Schumacher JH, Fiorentino DF, et al. Isolation of monoclonal antibodies specific for IL-4, IL-5, IL-6, and a new Th2-specific cytokine (IL-10), cytokine synthesis inhibitory factor, by using a solid phase radioimmunoadsorbent assay [J]. J Immunol, 1990, 145(9): 2938-2945.
    [32] Takayama T, Kaneko K, Morelli AE, et al. Retroviral delivery of transforming growth factor-beta1 to myeloid dendritic cells: inhibition of T-cell priming ability and influence on allograft survival [J]. Transplantation, 2002, 74(1): 112-119.
    [33] Hua N, Yamashita K, Hashimoto T, et al. Gene therapy-mediated CD40L and CD28 co-stimulatory signaling blockade plus transient anti-xenograft antibody suppression induces long-term acceptance of cardiac xenografts [J]. Transplantation, 2004, 78(10): 1463-1470.
    [34] Watanabe T, Miyatake T, Kumamoto H, et al. Adenovirus-mediated CTLA4 immunoglobulin G gene therapy in cardiac xenotransplantation [J]. Transplant Proc, 2004, 36(8): 2478-2479.
    [35] Miao G, Ito T, Uchikoshi F, et al. Development of donor-specific immunoregulatory T-cells after local CTLA4Ig gene transfer to pancreatic allograft [J]. Transplantation, 2004, 78(1): 59-64.
    [36] Zhang M, Wang Q, Liu Y, et al. Effective induction of immune tolerance by portal venous infusion with IL-10 gene-modified immature dendritic cells leading to prolongation of allograft survival [J]. J Mol Med, 2004, 82(4): 240-249.
    [37] Thomas JM, Contreras JL, Jiang XL, et al. Peritransplant tolerance induction in macaques: early events reflecting the unique synergy between immunotoxin and deoxyspergualin [J]. Transplantation, 1999, 68(11): 1660-1673.
    [38] Tashiro H, Shinozaki K, Yahata H, et al. Prolongation of liver allograft survival after interleukin-10 gene transduction 24-48 hours before donation [J]. Transplantation, 2000, 70(2): 336-339.
    [39] Shinozaki K, Yahata H, Tanji H, et al. Allograft transduction of IL-10 prolongs survival following orthotopic liver transplantation [J]. Gene Ther, 1999, 6(5): 816-822.
    [40] Dhodapkar MV, Steinman RM. Antigen-bearing immature dendritic cells induce peptide-specific CD8(+) regulatory T cells in vivo in humans [J]. Blood, 2002, 100(1): 174-177.
    [41] Lang T, Krams SM, Martinez OM. Production of IL-4 and IL-10 does not lead to immune quiescence in vascularized human organ grafts [J]. Transplantation, 1996, 62(6): 776-780.
    [42] Mottram PL, Han WR, Purcell LJ, et al. Increased expression of IL-4 and IL-10 and decreased expression of IL-2 and interferon-gamma in long-surviving mouse heart allografts after brief CD4-monoclonal antibody therapy [J]. Transplantation, 1995, 59(4): 559-565.
    [43] Otto C, Kauczok J, Martens N, et al. Mechanisms of tolerance induction after rat liver transplantation: intrahepatic CD4(+) T cells produce different cytokines during rejection and tolerance in response to stimulation [J]. J Gastrointest Surg, 2002, 6(3): 455-463.
    [44] Josien R, Douillard P, Guillot C, et al. A critical role for transforming growth factor-beta in donor transfusion-induced allograft tolerance [J]. J Clin Invest, 1998, 102(11): 1920-1926.
    [45] Wahl SM. Transforming growth factor beta (TGF-beta) in inflammation: a cause and a cure [J]. J Clin Immunol, 1992, 12(2): 61-74.
    [46] Dauer M, Obermaier B, Herten J, et al. Mature dendritic cells derived from human monocytes within 48 hours: a novel strategy for dendritic cell differentiation from blood precursors [J]. J Immunol, 2003, 170(8): 4069-4076.
    [47] Castriconi R, Cantoni C, Della Chiesa M, et al. Transforming growth factor beta 1 inhibits expression of NKp30 and NKG2D receptors: consequences for the NK-mediated killing of dendritic cells [J]. Proc Natl Acad Sci U S A, 2003, 100(7): 4120-4125.
    [48] Verrecchia F, Tacheau C, Wagner EF, et al. A central role for the JNK pathway in mediating the antagonistic activity of pro-inflammatory cytokines against transforming growth factor-beta-driven SMAD3/4-specific gene expression [J]. J Biol Chem, 2003, 278(3): 1585-1593.
    [49] Bonham CA, Lu L, Banas RA, et al. TGF-beta 1 pretreatment impairs the allostimulatory function of human bone marrow-derived antigen-presenting cells for both naive and primed T cells [J]. Transpl Immunol, 1996, 4(3): 186-191.
    [50] Witowski J, Ksiazek K, Jorres A. Interleukin-17: a mediator of inflammatory responses [J]. Cell Mol Life Sci, 2004, 61(5): 567-579.
    [51] Antonysamy MA, Fanslow WC, Fu F, et al. Evidence for a role of IL-17 in organ allograft rejection: IL-17 promotes the functional differentiation of dendritic cell progenitors [J]. J Immunol, 1999, 162(1): 577-584.
    [52] Szabo SJ, Dighe AS, Gubler U, et al. Regulation of the interleukin (IL)-12R beta 2 subunit expression in developing T helper 1 (Th1) and Th2 cells [J]. J Exp Med, 1997, 185(5): 817-824.
    [53] Schmitt E, Hoehn P, Huels C, et al. T helper type 1 development of naive CD4+ T cells requires the coordinate action of interleukin-12 and interferon-gamma and is inhibited by transforming growth factor-beta [J]. Eur J Immunol, 1994, 24(4): 793-798.
    [54] Weiner HL. Oral tolerance: immune mechanisms and the generation of Th3-type TGF-beta-secreting regulatory cells [J]. Microbes Infect, 2001, 3(11): 947-954.
    [55] Yang JS, Xu LY, Xiao BG, et al. Laquinimod (ABR-215062) suppresses the development of experimental autoimmune encephalomyelitis, modulates the Th1/Th2 balance and induces the Th3 cytokine TGF-beta in Lewis rats [J]. J Neuroimmunol, 2004, 156(1-2): 3-9.
    [56] Minguela A, Torio A, Marin L, et al. Implication of Th1, Th2, and Th3 cytokines in liver graft acceptance [J]. Transplant Proc, 1999, 31(1-2): 519-520.
    [57] Lowin B, Hahne M, Mattmann C, et al. Cytolytic T-cell cytotoxicity is mediated through perforin and Fas lytic pathways [J]. Nature, 1994, 370(6491): 650-652.
    [58] McDiarmid SV, Farmer DG, Kuniyoshi JS, et al. Perforin and granzyme B. Cytolytic proteins up-regulated during rejection of rat small intestine allografts [J]. Transplantation, 1995, 59(5): 762-766.
    [59] Tannapfel A, Kohlhaw K, Ebelt J, et al. Apoptosis and the expression of Fas and Fas ligand (FasL) antigen in rejection and reinfection in liver allograft specimens [J].Transplantation, 1999, 67(7): 1079-1083.
    [60] Nagata S, Golstein P. The Fas death factor [J]. Science, 1995, 267(5203): 1449-1456.
    [61] Martinez OM, Rosen HR. Basic concepts in transplant immunology [J]. Liver Transpl, 2005, 11(4): 370-381.
    [62] Griffith TS, Ferguson TA. The role of FasL-induced apoptosis in immune privilege [J]. Immunol Today, 1997, 18(5): 240-244.
    [63] Krams SM, Egawa H, Quinn MB, et al. Apoptosis as a mechanism of cell death in liver allograft rejection [J]. Transplantation, 1995, 59(4): 621-625.
    [64] Meyer D, Thorwarth W, Otto C, et al. Early T-cell inactivation and apoptosis-critical events for tolerance induction after allogeneic liver transplantation [J]. Transplant Proc, 2001, 33(1-2): 256-258.
    [65] Wells AD, Li XC, Li Y, et al. Requirement for T-cell apoptosis in the induction of peripheral transplantation tolerance [J]. Nat Med, 1999, 5(11): 1303-1307.
    [66] Qian S, Lu L, Li Y, et al. Apoptosis of graft-infiltrating cytotoxic T cells: a mechanism underlying "split tolerance" in mouse liver transplantation [J]. Transplant Proc, 1997, 29(1-2): 1168-1169.
    [67] Qian S, Lu L, Fu F, et al. Apoptosis within spontaneously accepted mouse liver allografts: evidence for deletion of cytotoxic T cells and implications for tolerance induction [J]. J Immunol, 1997, 158(10): 4654-4661.
    [68] Sakaguchi S, Sakaguchi N, Asano M, et al. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases [J]. J Immunol, 1995, 155(3): 1151-1164.
    [69] Bolton EM. Regulatory T cells in transplantation: natural or induced? [J]. Transplantation, 2005, 79(6): 643-645.
    [70] Oluwole SF, Oluwole OO, DePaz HA, et al. CD4+CD25+ regulatory T cells mediate acquired transplant tolerance [J]. Transpl Immunol, 2003, 11(3-4): 287-293.
    [71] Salama AD, Najafian N, Clarkson MR, et al. Regulatory CD25+ T cells in human kidney transplant recipients [J]. J Am Soc Nephrol, 2003, 14(6): 1643-1651.
    [72] Watanabe N, Wang YH, Lee HK, et al. Hassall's corpuscles instruct dendritic cells to induce CD4+CD25+ regulatory T cells in human thymus [J]. Nature, 2005, 436(7054): 1181-1185.
    [73] Toda A, Piccirillo CA. Development and function of naturally occurring CD4+CD25+ regulatory T cells [J]. J Leukoc Biol, 2006, 80(3): 458-470.
    [74] von Boehmer H. Mechanisms of suppression by suppressor T cells [J]. Nat Immunol, 2005, 6(4): 338-344.
    [75] Walsh PT, Taylor DK, Turka LA. Tregs and transplantation tolerance [J]. J Clin Invest, 2004, 114(10): 1398-1403.
    [76] Jiang H, Chess L. An integrated view of suppressor T cell subsets in immunoregulation [J]. J Clin Invest, 2004, 114(9): 1198-1208.
    [77] Lan RY, Ansari AA, Lian ZX, et al. Regulatory T cells: development, function and role in autoimmunity [J]. Autoimmun Rev, 2005, 4(6): 351-363.
    [78] Kim-Schulze S, Scotto L, Vlad G, et al. Recombinant Ig-like transcript 3-Fc modulates T cell responses via induction of Th anergy and differentiation of CD8+ T suppressor cells [J]. J Immunol, 2006, 176(5): 2790-2798.
    [79] Liu J, Liu Z, Witkowski P, et al. Rat CD8+ FOXP3+ T suppressor cells mediate tolerance to allogeneic heart transplants, inducing PIR-B in APC and rendering the graft invulnerable to rejection [J]. Transpl Immunol, 2004, 13(4): 239-247.
    [80] Chiffoleau E, Heslan JM, Heslan M, et al. TLR9 ligand enhances proliferation of rat CD4+ T cell and modulates suppressive activity mediated by CD4+ CD25+ T cell [J]. Int Immunol, 2007, 19(2): 193-201.
    [81] Chai JG, Xue SA, Coe D, et al. Regulatory T cells, derived from naive CD4+CD25- T cells by in vitro Foxp3 gene transfer, can induce transplantation tolerance [J]. Transplantation, 2005, 79(10): 1310-1316.
    [82] Battaglia M, Stabilini A, Draghici E, et al. Rapamycin and interleukin-10 treatment induces T regulatory type 1 cells that mediate antigen-specific transplantation tolerance [J]. Diabetes, 2006, 55(1): 40-49.
    [83] Albert MH, Liu Y, Anasetti C, et al. Antigen-dependent suppression of alloresponses by Foxp3-induced regulatory T cells in transplantation [J]. Eur JImmunol, 2005, 35(9): 2598-2607.
    [84] Battaglia M, Gregori S, Bacchetta R, et al. Tr1 cells: from discovery to their clinical application [J]. Semin Immunol, 2006, 18(2): 120-127.
    [85] Bone RC. Sir Isaac Newton, sepsis, SIRS, and CARS [J]. Crit Care Med, 1996, 24(7): 1125-1128.
    [86] Bone RC. Immunologic dissonance: a continuing evolution in our understanding of the systemic inflammatory response syndrome (SIRS) and the multiple organ dysfunction syndrome (MODS) [J]. Ann Intern Med, 1996, 125(8): 680-687.
    [87] Perl M, Chung CS, Garber M, et al. Contribution of anti-inflammatory/immune suppressive processes to the pathology of sepsis [J]. Front Biosci, 2006, 11(272-299.
    [88] Mahidhara R, Billiar TR. Apoptosis in sepsis [J]. Crit Care Med, 2000, 28(4 Suppl): N105-113.
    [89] Ding Y, Chung CS, Newton S, et al. Polymicrobial sepsis induces divergent effects on splenic and peritoneal dendritic cell function in mice [J]. Shock, 2004, 22(2): 137-144.
    [90] Efron PA, Martins A, Minnich D, et al. Characterization of the systemic loss of dendritic cells in murine lymph nodes during polymicrobial sepsis [J]. J Immunol, 2004, 173(5): 3035-3043.
    [91] Razonable RR, Paya CV. Infections and allograft rejection - intertwined complications of organ transplantation [J]. Swiss Med Wkly, 2005, 135(39-40): 571-573.
    [92] Simon CO, Seckert CK, Dreis D, et al. Role for tumor necrosis factor alpha in murine cytomegalovirus transcriptional reactivation in latently infected lungs [J]. J Virol, 2005, 79(1): 326-340.
    [93] Garbino J, Romand JA, Pittet D, et al. Infection and rejection in liver transplant patients: a 10-year Swiss single-centre experience [J]. Swiss Med Wkly, 2005, 135(39-40): 587-593.