首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 93 毫秒
1.
目的用抗CD-40L单抗加小剂量CsA联合免疫治疗肝移植大鼠受体,观察其生存时间?移植肝组织学和Th1/Th2细胞因子谱的变化。方法建立大鼠肝移植模型后,将动物随机分为4组。A组为同基因移植组,SD→SD;B组为同种异体移植组,SD→Wistar,不用任何免疫抑制治疗措施;C组,SD→Wistar,采用CsA处理;D组,SD→Wistar,采用CsA加抗CD-40L(CD-154)单抗处理。观察各组肝移植受体生存期和移植肝病理变化;用ELISA法检测外周血细胞因子水平。结果A,D组均可长期存活,B,C组生存时间分别为(13.8±2.4)d,(29.8±4.1)d;B,C组病理组织切片见中/重度急性排斥反应,D组移植肝组织损伤程度显著减轻,A组基本无排斥反应。B组血清IL-2和IFN-γ高于其余各组(P<0.05),C,D组IL-4,IL-10水平较B组有所升高(但P>0.05),尤其D组IL-10表达水平显著高于B组(P<0.05)。结论联合免疫治疗可有效抑制其急性排斥反应,延长大鼠肝移植受体的生存时间。Th2类细胞因子IL-4和IL-10的高水平表达与诱导移植耐受、抑制排斥反应有重要关系,它有助于大鼠肝移植受体和移植肝的长期存活。  相似文献   

2.
目的 观察抗CD-40L单抗加小剂量CsA联合免疫治疗对肝移植大鼠受体免疫耐受诱导的作用.方法 在建立稳定大鼠肝移植模型的基础上,将肝移植模型分为5组.A组为SD→SD对照组;B组为SD→Wistar对照组,A,B组术后不用任何治疗措施;C组为SD→Wistar,术后用CsA1~5 d;D组为SD→Wistar,术后用CsA 1~5 d加抗CD-40L(CD-154)单抗0~2d;E组为D组+术前供体特异性输血(DSBT).观察受体存活时间、移植肝病理改变以及术后外周血中细胞因子的变化.结果 A,D,E组受体大鼠存活时点(均>60 d)均明显长于B组和C组.D,E组移植肝急性排斥反应明显减轻.B组IL-2和IFN-γ的血清水平显著高于其余各组(P<0.05).B,C,D,E 4组IL-4和IL-10较A组均有明显增加,尤其D,E组的IL-10表达较B组显著增高(P<0.05). 结论 抗CD-40L单抗加小剂量CsA(伴或不伴DSBT)联合免疫治疗,可有效延长肝移植大鼠受体生存时间、减轻急性排斥反应并诱导Th2类细胞因子的高水平表达,有助于受体和移植肝的长期存活.  相似文献   

3.
目的:研究大鼠肝移植术后移植肝和外周血中细胞因子的表达,并分析其在急性排异反应和耐受诱导中的作用。方法:Kamada法建立大鼠肝移植模型,并随机分为4组,每组8只。A组:同品系组;B组:免疫排异组;C组:B组+免疫抑制剂;D组:C组+骨髓输注。RT-PCR和ELISA技术分别检测移植肝和外周血中细胞因子IL-2、INF-γ、IL-4、IL-10、TGF-β1的表达,并观察受体存活率。结果:在B组中Th1型细胞因子(IL-2、INF-γ)表达高于A、C和D组(P<0.05);而Th2型细胞因子(IL-4、IL-10)和TGF-β1表达低于C和D组(P<0.05);细胞因子在C组与D组大鼠中的表达无统计学差异。A、C、D组大鼠术后近、远期存活率明显优于B组。结论:Th1型细胞因子(IL-2、INF-γ)参与肝移植急性排异反应的发生,Th2型细胞因子(IL-4、IL-10)和TGF-β1可能参与移植物免疫耐受的诱导,延长移植物存活。  相似文献   

4.
细胞因子在心脏移植急性排斥反应中的表达及其作用机理   总被引:4,自引:0,他引:4  
目的观察同种大鼠心脏移植急性排斥反应中,局部细胞因子网络的变化及其机理.方法健康雄性Wistar大鼠(受体)和SD大鼠各48只,将接受移植心脏的Wistar大鼠分4组,每组12只.A组对照组;B组抗白介素2单克隆抗体(IL-2Mab)组;C组环孢菌素A(CsA)组;D组IL-2Mab加CsA组.4组大鼠分别静脉给予生理盐水、抗白介素2单克隆抗体及口服CsA、静脉给予抗白介素2单克隆抗体加口服CsA,采用改良的移植术式建立移植模型.常规监测排斥反应发生情况.应用RT-PCR的方法于术后第1、3、5、7、9、11、14天动态检测移植物局部细胞因子IL-1β、IL-2、CD25、IL-4、IL-5、IL-6、IL-10、TNFα、IFNγ的表达水平.结果移植心脏存活时间,A组为(8.3±1.7)d;B组为(29.2±7.1)d;C组为(26.4±5.7)d;D组为(55.0±11.6)d.B、C、D组移植心脏存活时间显著延长,与A组相比,差异有极显著性意义(P<0.01).存活时间较长的移植心脏的淋巴细胞浸润和心肌坏死的程度比存活时间较短的心脏明显减轻.IL-1β的表达在各组均较高;IL-4、IL-5、IL-6、IL-10的表达水平在移植心脏存活时间较长的组较高;而IL-2、CD25、IFN-γ、TNFα的表达则相对较低;4组相比差异有显著性意义(P<0.05).结论细胞因子网络在心脏移植排斥反应中发生了相应的变化,并与干预的因素及移植物存活时间有密切的关系.IL-2Mab、CsA联合用药促使TH1类细胞因子向TH2类细胞因子整体偏离,这种免疫偏离使移植心脏存活时间显著延长.  相似文献   

5.
目的比较不同品系大鼠之间肝移植排斥反应特点。方法依据大鼠不同品系分成对照组SD→SD(A组),实验组Wistar→SD(B组)、Lewis→BN(C组)以及DA→Lewis(D组)4个组,采用Kamada双袖套法建立大鼠原位肝移植模型。比较各组受体大鼠术后10 d血清肝功能指标[丙氨酸转氨酶(ALT)、总胆红素(TB)和白蛋白(Alb)]、血清白细胞介素(IL)-2和IL-10水平、急性排斥反应组织病理学分级和术后平均生存时间。结果与A、B组比较,C组和D组的血清ALT和TB明显升高,Alb明显降低,差异均有统计学意义(均为P0.05)。与C组比较,D组的TB明显升高,Alb明显降低,差异均有统计学意义(均为P0.05)。与A组比较,B、C、D组大鼠血清IL-2和IL-10水平均明显升高,C组和D组的IL-2/IL-10亦明显升高,差异有统计学意义(均为P0.05)。与B组比较,C组和D组大鼠血清IL-2水平明显升高,D组的IL-2/IL-10亦明显升高,差异有统计学意义(均为P0.05)。与C组比较,D组大鼠血清IL-2水平明显升高,D组的IL-2/IL-10亦明显升高,差异有统计学意义(均为P0.05)。肝组织病理学检查显示,A组大鼠肝组织未见明显排斥反应,排斥活动性指数(RAI)评分(1.8±0.7)分;B组RAI评分(3.1±1.3)分,属轻度或不明确性排斥反应;C组RAI评分(6.9±0.8)分,属中~重度排斥反应;D组RAI评分(8.8±0.5)分,属重度排斥反应。各组间RAI评分比较以及组间两两比较,差异均有统计学意义(均为P0.05)。A组、B组、C组、D组受体大鼠术后平均生存时间分别为(119.3±1.9)d、(116.9±8.3)d、(53.4±6.1)d、(12.1±2.4)d,A组与B组的生存时间比较差异无统计学意义,余各组两两比较差异均有统计学意义(均为P0.05)。结论 4种组合中,大鼠DA→Lewis模型排斥反应最剧烈,Lewis→BN次之,而Wistar→SD最轻,接近于免疫耐受。  相似文献   

6.
目的 研究大鼠异体肢体移植术后急性排斥反应阶段,移植肢体血管内皮细胞细胞间黏附分子-1(intercellular adhesion molecule-1,ICAM-1)动态变化及环孢素A(cyclosporine A,CsA)抗免疫排斥的作用.方法 建立大鼠肢体移植动物模型,随机分为对照组(Wistar大鼠→Wistar大鼠)、排斥组(SD大鼠→ Wistar大鼠)和CsA治疗组(SD大鼠→Wistar大鼠),术后1、4、7 d获取移植肢体股动脉行病理学观察,采用免疫组化法检测移植肢体血管ICAM-1表达的变化.结果 对照组供体移植肢体股动脉血管内皮细胞仅出现轻微肿胀与ICAM-1表达微弱;排斥组血管内皮细胞肿胀明显,淋巴细胞大量浸润,ICAM-1的表达强度和数量明显增加;CsA治疗组移植肢体血管内皮细胞仅有少量淋巴细胞浸润,ICAM-1表达较弱.结论 大鼠异体肢体移植术后急性排斥反应阶段,血管内皮细胞ICAM-1表达水平与排斥反应的发生和发展有关,CsA可降低移植肢体血管内皮细胞ICAM-1表达,抑制复合组织移植术后急性排斥反应.  相似文献   

7.
目的 探讨肝癌肝移植后应用活化供体脾脏来源的淋巴细胞,对受体实施过继性免疫治疗以预防肝癌复发的可行性.方法 以培养黏附法体外分离培养Wistar大鼠骨髓树突状细胞(DCs)前体细胞,经诱导后与Wistar大鼠肝癌细胞CBRH-7919裂解物抗原共同孵育,形成负载癌抗原的DCs.以此DCs分别诱导受体(Wistar大鼠)、供体(SD大鼠)来源的脾淋巴细胞作为两个实验组C(受体活化组)、D(供体活化组),以未经抗原诱导的受、供体脾淋巴细胞作为对照组A(受体对照组)、B(供体对照组).对比活化、非活化以及供体来源、受体来源的脾淋巴细胞在体外对受体肿瘤细胞的杀伤活性.以上述不同来源和处理的脾淋巴细胞对SD→Wistar大鼠肝移植后肿瘤复发模型行过继性免疫治疗,以盐水治疗作为对照组E,每组6只大鼠,观察各组免疫治疗对于肿瘤浸润淋巴细胞、受体肝脏成瘤率和供肝的排斥反应的影响.结果 DCs诱导脾细胞过程中,培养上清液γ干扰素(IFN-γ)分泌C、D组较A、B组有明显的升高.C、D组对肝癌细胞的杀伤活性较A、B组显著增强,D组较C组显著增强.D组脾细胞回输体内后,在移植后大鼠体内观察到了肿瘤浸润淋巴细胞增多,肿瘤组织坏死和成瘤率下降.免疫治疗前后供肝未见严重排斥反应发生.结论 活化的供体脾细胞较受体脾细胞有更强的肿瘤杀伤效果.应用供体脾淋巴细胞对肝移植受体进行过继性免疫治疗可以在不增加对移植肝排斥反应的同时,为预防肝癌肝移植术后复发、延长生存提供一种可能的方法.  相似文献   

8.
不同品系大鼠之间原位肝移植的实验观察   总被引:10,自引:2,他引:10  
目的 探讨不同品系大鼠之间原位肝移植耐受或排斥关系。方法 采用KamadaN等双袖套法进行原位肝移植 ;采用OnoK等改良腹腔内吻合法进行异位心脏移植。结果 Wistar→SD、SD→Wistar以及SD→DA大鼠原位肝移植 ,受体鼠存活均超过 180d ;同种组合方式的异位心脏移植供心平均存活 6.3d。给原位肝移植受体大鼠再移植供体源心脏 ,移植心脏存活均超过 15 0d。给原位肝移植受体大鼠再移植第 3品系大鼠心脏 ,移植心脏平均存活 6.8d。结论 Wistar→SD、SD→Wistar以及SD→DA大鼠的移植组合是分离耐受关系。  相似文献   

9.
目的 通过建立同种异体大鼠骨髓及肝联合移植动物模型,探讨提高大鼠肝移植模型的稳定性和存活率的方法及可行性.方法 将SD大鼠(♂)、Wistar大鼠(♀)分成三组:Ⅰ组大鼠Kamada"二袖套法"行SD→Wistar大鼠肝移植;Ⅱ组Wistar大鼠(♀)TBI(11 Gy),4 h后输人SD大鼠(♂)BMC(8×107),28 d后Kamada"二袖套法"行SD→Wistar大鼠肝移植;Ⅲ组Wistar大鼠(♀)TBI(7 Gy),4h后输入SD大鼠(♂)BMC(8×107),2 d后CTX(50 mg/kg体重)腹腔注射,28 d后Kamada"二袖套法"行SD→Wistar大鼠肝移植.分别于BMT后10、20 d通过PCR方法检测Ⅱ组和Ⅲ组Wistar大鼠体内的SD大鼠源性Y染色体特异性片段.并比较3组大鼠肝移植术后1周生存率、生存状况及生存时间.结果 Ⅱ组和Ⅲ组大鼠外周血均检测出SD大鼠源性嵌合体.DTH检查显示对SD大鼠产生免疫耐受.肝移植结果显示,Ⅰ组大鼠均在4~5 d死亡,而Ⅱ组和Ⅲ组大鼠1周存活率为80.0%和84.2%,但Ⅱ组的生存状况不如Ⅲ组.结论 应用7GyTBI+CTX+供体BMT可成功建立同种异体大鼠嵌合体模型,诱导特异性免疫耐受,大大提高肝移植术后大鼠的生存状况及生存时间.  相似文献   

10.
目的 探讨CD28、CD40通路共刺激后淋巴细胞产生Th1(IL-2、IFN-γ、IL-12)及Th2细胞因子(IL-4、IL-10)的变化及免疫抑制剂环孢素(CsA)、雷帕霉素(RPM)及霉酚酸(MPA)对共刺激通路激活后淋巴细胞产生Th1及Th2细胞因子的影响.方法采用单克隆抗体(mAb)与淋巴细胞表面CD3、CD28及CD40L分子结合产生相应刺激信号,单刺激及共刺激组分为:a组,CD3 mAb单刺激;b组,CD3 mAb加CD28 mAb共刺激;c组,CD3 mAb加CD28 mAb加CD40 L mAb共刺激;d组,CD3 mAb加CD28 mAb加CTLA4 mAb共刺激.各mAb的终浓度均为100 ng/ml.干预组分别将终浓度为300 ng/ml的CsA、RPM、MPA加入上述4组.ELISA法测定上述细胞培养上清中的细胞因子值.结果 a、b、c 3组IFN-γ分别为(248.91±11.20)、(555.08±24.42)、(548.19±33.06)ng/ml,IL-2分别为(29.48±8.61)、(1100.82±99.29)、(842.23±29.31)ng/ml,IL-4分别为(32.29±6.76)、(116.02±15.03)、(147.28±18.07)ng/ml,IL-10分别为(147.01±10.47)、(291.79±12.47)、(302.52±35.18)ng/ml.b、c组与a组比较,差异均有统计学意义(P<0.01);b、c组IL-2、IL-12、IL-4比较,差异均有统计学意义(P<0.05).d组IFN-γ、IL-2及IL-10分别为(497.42±29.03)、(739.77±18.58)及(120.33±13.21)ng/ml,与b组相比,差异均有统计学意义(P<0.05).CsA、RPM及MPA对共刺激后Th1/Th2细胞因子的产生均有抑制作用,CsA对4种细胞因子产生的抑制作用强于RPM和MPA,其中对IL-2及IL-4的抑制作用更为明显.CsA与CTLA4 mAb有协同作用.共刺激后IL-12产生升高,MPA可抑制单刺激和共刺激后IL-12的产生,CsA和RPM对IL-12的产生无明显抑制作用.结论 CD28、CD40共刺激通路在淋巴细胞活化中起关键作用.CsA、RPM、MPA及CTLA4 mAb对共刺激后Th1/Th2细胞因子的产生均有抑制作用,CsA的抑制作用更为明显.CD40 L mAb使Th1细胞因子及IL-12水平下降,又促进Th2细胞因子(以IL-4为主)产生,该作用可能是抗CD40 L抗体诱导移植物存活期延长的机制之一.  相似文献   

11.
Th1 effector CD4+ cells contribute to the pathogenesis of proliferative and crescentic glomerulonephritis, but whether effector Th17 cells also contribute is unknown. We compared the involvement of Th1 and Th17 cells in a mouse model of antigen-specific glomerulonephritis in which effector CD4+ cells are the only components of adaptive immunity that induce injury. We planted the antigen ovalbumin on the glomerular basement membrane of Rag1−/− mice using an ovalbumin-conjugated non-nephritogenic IgG1 monoclonal antibody against α3(IV) collagen. Subsequent injection of either Th1- or Th17-polarized ovalbumin-specific CD4+ effector cells induced proliferative glomerulonephritis. Mice injected with Th1 cells developed progressive albuminuria over 21 d, histologic injury including 5.5 ± 0.9% crescent formation/segmental necrosis, elevated urinary nitrate, and increased renal NOS2, CCL2, and CCL5 mRNA. Mice injected with Th17 cells developed albuminuria by 3 d; compared with Th1-injected mice, their glomeruli contained more neutrophils and greater expression of renal CXCL1 mRNA. In conclusion, Th1 and Th17 effector cells can induce glomerular injury. Understanding how these two subsets mediate proliferative forms of glomerulonephritis may lead to targeted therapies.Although proliferative and crescentic glomerulonephritides occur in different primary renal diseases and are an important component of several systemic diseases, features of human renal biopsies suggest some common effector pathways. In most cases of rapidly progressive GN there is evidence for an important role for cellular immune effectors: T cells, macrophages, and neutrophils,13 a role confirmed in animal models.47 CD4+ T cells are key components of renal injury.4,8 When activated, CD4+ cells tend to differentiate into subsets (T helper cells—Th1, Th2 and Th17) that engage immune effectors in different ways. In proliferative forms of GN, T cells direct adaptive immune responses that drive glomerular disease, but also, in rapidly progressive GN, CD4+ cells themselves accumulate in glomeruli as effectors. These effector T helper cells activate innate effector cells, predominantly neutrophils and macrophages, and activate and damage intrinsic renal cells.In GN, the variable Th1-Th2 predominance of responses influences the histologic patterns and severity of GN.9 Th1 cells, which secrete IFNγ and activate macrophages, are important in some forms of experimental proliferative GN. Th2 cells, characterized by IL-4 production, promote humoral immunity and are important in several forms of GN, but there is little evidence that Th2 cells play primary roles as effector cells within glomeruli in rapidly progressive GN. A binary Th1/Th2 model explains many of the differences in the patterns of immune responses in GN. However, there are discrepancies10 that might be explained by defining a role for a third major subset, Th17 cells, characterized by the production of IL-17A. Its biology has recently been comprehensively reviewed.11 Th17 subset effects potentially relevant to rapidly progressive GN include direct effects on neutrophils and stimulating the production of neutrophil chemoattractants by tissue cells. Thus, in most rapidly progressive types of GN, cell-mediated injury, a key component of injury, may be directed by the Th17 subset, the Th1 subset, or both subsets.Although antigen-specific T cells are critical to adaptive immune responses, the cells themselves are relatively infrequent. T cell receptor (TcR) transgenic mice help define the contributions of different antigen-specific T helper cell subsets in organ-specific disease.1214 In the studies presented here we have established a new antigen-specific model of GN. Ovalbumin (OVA)-specific OT-II TcR transgenic CD4+ cells15 are polarized ex vivo under Th1- or Th17-inducing conditions. Effector cells are transferred into recombination activating gene-1 deficient (Rag1−/−) mice with OVA planted in their glomeruli by injecting an OVA conjugate. OVA is conjugated to a mouse IgG1 mAb binding to α3(IV) collagen in murine glomerular basement membrane (GBM). The mAb-OVA conjugate dose is capable of planting significant OVA in glomeruli as an antigen to induce effector responses, but is insufficient to induce significant histologic or functional injury as an antibody. This model allows us to understand effector CD4+ T cell and Th subset-induced injury, with no effects from CD8+ cells or B cells.CD4+ cells, isolated from OVA-specific TcR transgenic (OT-II) mice and cultured under Th1 or Th17 priming conditions (see Concise Methods), were confirmed to be Th1 or Th17 by cytokine production before transfer. Th1 polarized cells expressed IFNγ, whereas no IL-17A or IL-4 production was detected (Figure 1A). Th17 polarized cells were strong IL-17A producers, showing only weak IFNγ production, with >20% of cells producing IL-17A, but few IFNγ or double-positive cells (Figure 1B). To plant OVA in glomeruli, the mAb 8D1, a non-nephritogenic, murine IgG1 binding to mouse α3(IV)NC1,16 was conjugated to OVA and purified by size-exclusion chromatography so that no free OVA or unconjugated 8D1 mAb remained, confirmed by Western blotting (Figure 1C). Antigen-specific CD4+ cells recognized OVA bound to the 8D1 anti-GBM mAb. Culture of CFSE-labeled naive OT-II cells incubated with the 8D1-OVA conjugate enhanced their survival (30% to 40% survival after 72 h versus 5% to 6% with unconjugated antibody) and induced OT-II cell proliferation (72 h: 27% of cells, up to 4 cycles, Figure 1D). OT-II cells incubated with unconjugated 8D1 did not proliferate (Figure 1E). After intravenous injection, 8D1-OVA conjugates bound to the GBM in a linear manner; no fluorescence signal was observed after transfer of Th1 cells without 8D1-OVA (Figure 1G). Western blotting showed OVA in the kidney after injection of 8D1-OVA conjugate (Figure 1H). Lungs from 8D1-OVA-injected mice were weakly positive for mouse IgG, whereas no IgG was detected in the spleen or liver (detecting antibody titer 1:100) 3 or 21 d after injection. Mouse IgG was not detected in sera (ELISA, dilution 1:100) at day 3 or day 21. As expected (given the transfer of only CD4+ cells to Rag1−/− mice), no anti-OVA antibodies in sera were detected in recipient mice (data not shown).Open in a separate windowFigure 1.Differentiation of OVA-specific OT-II Th1 and Th17 cells, antibody-OVA conjugation, glomerular IgG and intrarenal OVA detection, and recipient immune responses after cell transfer. (A) After stimulating naive OT-II cells with OVA in a Th1 environment, IFNγ was produced and intracellular cytokine staining of CD4+ cells demonstrated strong IFNγ staining with minimal IL-17A or IL-4. (B) Culturing cells in a Th17-stimulating environment led to strong IL-17A production, whereas cells stained positive for IL-17A but not IL-4, and only 2% of cells produced IFNγ. (C) Chromatographic profile of 8D1-OVA conjugation. The numbers 1 to 7 represent fractions collected for analyses by Western blotting, which confirmed that all OVA-conjugated fractions contained OVA and IgG (lanes 1–6), whereas unconjugated fractions (represented as “Un”) contained IgG alone. The lane labeled “M” contained molecular weight markers. (D and E): 8D1-OVA was recognized by OT-II cells because multiple cycles of proliferation of cultured naive OT-II cells (D) were seen with 8D1-OVA conjugate and (E) not seen with unconjugated antibody. Strong linear IgG staining of glomeruli was seen after (F) the administration of 8D1-OVA to Rag1−/− mice, but not after (G) the injection of Th1 cells without antibody. Western blotting of homogenized kidney (H) 24 h after the administration of 8D1-OVA demonstrated OVA in the kidneys (labeled as OVA-Ab); this was not seen after the administration of unconjugated antibody (labeled as Un Ab). (I) Systemic immune responses of recipient Rag1−/− mice at 21 d assessed by splenic cytokine production demonstrated enhanced IFNγ production in mice given 8D1-OVA and Th1 cells, with enhanced IL-17A production by mice receiving 8D1-OVA and Th17 cells. (J) DTH to OVA (at 21 d) was induced only in mice given 8D1-OVA and Th1 cells. *P < 0.05, ***P < 0.001.To determine whether transfer of either Th1 or Th17 antigen-specific effector cells induces glomerular injury, 8D1-OVA conjugate was administered intravenously to Rag1−/− mice (lacking adaptive immunity). Three hours later, 5 × 106 Th1 or Th17 cells were injected intravenously. Groups of mice injected with 8D1-OVA alone (without cells) or Th1 cells alone (without 8D1-OVA) served as controls. At 21 d, the injected T cells largely maintained their initial phenotype, because host splenocytes from mice given Th1 cells showed enhanced OVA-stimulated IFNγ production whereas IL-17A production was enhanced in mice given Th17 cells (Figure 1I). Dermal-delayed-type hypersensitivity (DTH) was induced by footpad injection of OVA and measured after 24 h. Only mice that received the 8D1-OVA conjugate and Th1 polarized cells developed dermal DTH (Figure 1J), a classical Th1 response.17After planting OVA in glomeruli, administration of Th1 or Th17 cells induced glomerular disease. Urinary albumin excretion was not increased in mice given 8D1-OVA conjugate alone or Th1 cells alone, but Th1 or Th17 cells with 8D1-OVA induced significant albuminuria (Figure 2A). Albuminuria was consistent throughout the time course of the study in the Th17 group, whereas in the Th1 group there was a progressive increase in albuminuria until day 21 (Figure 2B). Control mice given Th1 cells alone or the 8D1-OVA conjugate alone exhibited only mild histologic changes (no crescent formation, fibrinoid necrosis, or hyalinosis). Analysis of histologic injury demonstrated substantially more abnormal glomeruli in the mice given 8D1-OVA conjugate with Th1 or Th17 cells compared with control groups (Figure 2C). Th1 and Th17 (+8D1-OVA) recipients developed proliferative GN, [glomerular hypercellularity: 8D1-OVA and Th1 cells: 32.1 ± 1.0 cells/glomerular cross section (c/gcs), 8D1-OVA and Th17 cells: 29.8 ± 1.1 c/gcs, 8D1-OVA alone: 21.3 ± 0.2 c/gcs, Th1 cells alone: 18.9 ± 2.0 c/gcs; P < 0.001]. Representative kidney sections from each group are shown (Figure 2, D through G). Crescent formation and fibrinoid necrosis, although seen in only a few glomeruli, was observed exclusively in mice given 8D1-OVA conjugate and Th1 cells (5.5 ± 0.9% at day 21; Figure 2, H and I). No crescent formation was observed in mice receiving 8D1-OVA conjugate and Th17 cells. Mice did not develop significant renal impairment (measured by BUN; data not shown).Open in a separate windowFigure 2.Renal injury in mice injected with 8D1-OVA conjugate, then either Th1 or Th17 cells. (A) Mice given 8D1-OVA conjugate or Th1 cells alone did not develop albuminuria above values for noninjected Rag1−/− mice (dotted line). At 21 d, albuminuria was increased in mice given 8D1-OVA and Th1 cells or 8D1-OVA and Th17 cells. (B) In mice given 8D1-OVA and Th17 cells, albuminuria had plateaued by day 7 and did not progress. In mice given 8D1-OVA and Th1 cells there was a progressive rise in albuminuria. (C) Histologic injury was significant in mice given 8D1-OVA and either Th1 or Th17 cells. Representative glomeruli from mice given (D) 8D1-OVA alone, (E) Th1 cells alone, (F) 8D1-OVA and Th1 cells, and (G) 8D1-OVA and Th17 cells are shown. (H and I) Crescentic injury and fibrinoid necrosis were only seen in mice given 8D1-OVA and Th1 cells. ***P < 0.001Recruitment and activation of leukocyte subpopulations differed in mice administered Th1 or Th17 cells (Figure 3A). Although glomerular CD4+ cell and macrophage numbers were similarly increased in mice given 8D1-OVA conjugate and either Th1 or Th17 cells at day 21, more neutrophils were found in mice given 8D1-OVA and Th17 cells compared with mice given 8D1-OVA and Th1 cells. Interstitial leukocyte infiltrates followed a similar pattern (Figure 3B). Consistent with the finding of increased neutrophils in kidneys of mice receiving Th17 cells, renal mRNA expression of the primary neutrophil attracting chemokine CXCL1 was elevated (Figure 3C). Th17 cells attract neutrophils18 and in vitro studies have shown that neutrophil recruitment is achieved via production of CXCL8, the human homologue of CXCL1, by Th17 cells.19 It is therefore likely that at least some of the Th17-induced renal injury is mediated by neutrophils. In mice receiving 8D1-OVA and Th1 cells, macrophages were likely to be more activated; only these mice developed dermal DTH and increased expression of mRNA for the macrophage chemoattractants CCL2 and CCL5 (Figure 3, D and E), which have been associated with experimental crescentic GN.20 Furthermore, type 2 nitric oxide synthase (NOS2/iNOS) mRNA, a marker of macrophage activation21 and urinary nitrate, a marker of intrarenal macrophage NOS2 production, were increased in this group (Figure 3, F and G).Open in a separate windowFigure 3.Leukocytes in kidneys of mice with either Th1- or Th17-induced injury 21 d after cell transfer. (A) Glomerular CD4+T cells, neutrophils, and macrophages were increased in mice given 8D1-OVA and Th1/Th17 cells. Neutrophil recruitment was incrementally increased in mice given 8D1-OVA and Th17 cells compared with 8D1-OVA and Th1 cells. (B) A similar pattern of recruitment was seen in the cortical interstitium. Renal chemokine mRNA expression demonstrated (C) enhanced CXCL1 mRNA in mice given 8D1-OVA and Th17 cells, whereas (D) CCL2 and (E) CCL5 were increased in mice given 8D1-OVA and Th1 cells. (F and G) NOS2 and urinary nitrate, markers of macrophage activation, were increased in mice receiving 8D1-OVA and Th1 cells. For mRNA, values for the 8D1-OVA alone group are presented as 1. *P < 0.05, **P < 0.01, ***P < 0.001.Further studies were performed 3 d after cell transfer. At this time point, albuminuria was present in mice receiving 8D1-OVA conjugate and Th17 cells, but not 8D1-OVA conjugate and Th1 cells (Figure 4A), and a higher proportion of glomeruli were abnormal in mice that had received Th17 cells (Figure 4B). Therefore, Th17-induced glomerular injury occurred earlier than Th1-induced injury. Leukocytes were present in glomeruli (Figure 4C) with increased numbers of neutrophils in glomeruli of mice receiving Th17 cells (compared with Th1 cell recipients), whereas Th1 cell recipients exhibited more macrophages. At day 3, these findings were glomerulo-specific; differences between Th1 and Th17 cell recipients were not seen in the interstitium (Figure 4D).Open in a separate windowFigure 4.Renal disease in mice 3 d after injection with 8D1-OVA and either Th1 or Th17 cells. (A) Pathologic albuminuria (dotted line represents values for noninjected Rag1−/− mice) and (B) increased numbers of abnormal glomeruli were evident in mice that received 8D1-OVA and Th17 cells. (C) Leukocyte recruitment to glomeruli demonstrated CD4+ cells (more in mice receiving Th1 cells), with comparatively more neutrophils in glomeruli of Th17 cell recipients and more macrophages in glomeruli of Th1 cell recipients. (D) Interstitial leukocytes were similar in Th1 and Th17 cell recipients 3 d after cell transfer. *P < 0.05, **P < 0.01, ***P < 0.001.These studies used Rag1−/− mice as recipients of effector antigen-specific Th1 or Th17 cells. Because these mice do not possess T or B cells, OVA planted in glomeruli cannot induce CD8+ or B cell responses, and regulatory T cells are unable to influence the pattern of injury. A major advantage of this strategy is that Th1- and Th17-mediated injury can be assessed in a pure experimental system. However, T cells transferred into Rag1−/− mice can undergo homeostatic expansion, and it is possible that the transferred Th1 cells might have expanded more rapidly than Th17 cells. Recently, studies in experimental type 1 diabetes induced by transfer of cells from a TcR transgenic mouse specific for an islet autoantigen showed conversion of Th17 cells to a Th1 phenotype after transfer.22,23 Although our Th17 polarized OT-II cells, specific for a foreign antigen, showed some IFNγ production after 21 d, they were still capable of producing IL-17A. Furthermore, dermal DTH and renal disease were different in Th1 recipients compared with the Th17 recipients at 21 d, supporting the maintenance of separate phenotypes after transfer. Although the studies presented here are the first to demonstrate a role for Th17 and Th1 cells in the same experimental system, other studies2426 have used genetically deficient mice to implicate Th17 cells in experimental renal disease.These studies describe a novel model of cell-mediated proliferative GN for which adaptive components are only effector antigen-specific CD4+ T cells. They demonstrate that both Th1 and Th17 cells can induce proliferative GN. Th17 cells induce albuminuria early, with persistent accumulation of leukocytes. Administration of Th1 cells lead to a slower rise in albuminuria, but more macrophage activation and DTH-like injury, including, in some glomeruli, crescent formation and fibrinoid necrosis. It is likely that Th1 and Th17 responses play a role in proliferative forms of GN and both represent potential therapeutic targets.  相似文献   

12.
Th1/Th2细胞与肿瘤复发   总被引:6,自引:0,他引:6  
目的探讨T辅助淋巴细胞Ⅰ型(Th1)/T辅助淋巴细胞Ⅱ型(Th2)在肿瘤复发中的研究进展。方法复习国内、外相关文献并进行综述。结果肿瘤治疗后体内出现Th1向Th2漂移,使肿瘤细胞逃避机体的免疫监视,导致肿瘤的复发。结论Th1向Th2型漂移与肿瘤治疗后的复发有关,促使肿瘤治疗后的机体细胞因子由Th2向Th1逆转,重新达到平衡,成为肿瘤免疫治疗的新思路。  相似文献   

13.
14.
15.
In the pathogenesis of anti-neutrophil cytoplasm antibodies (ANCA)-associated vasculitis, T cell contribution is indicated by T cell-dependent ANCA production combined with the presence of T cells in inflammatory infiltrates. However, the exact pathogenic role of T cells in ANCA-associated vasculitis remains to be determined. The Th1/Th2 concept is useful for understanding T cell involvement in pathological processes. This review focuses on T cells and particularly the Th1/Th2 paradigm in ANCA-associated vasculitis. Most research has been done in Wegener's granulomatosis, where a shift in T cell response, from a Th1 pattern in localized disease towards a Th0/Th2 pattern in generalized disease, appears to occur. Although less thoroughly studied, data in Churg-Strauss syndrome and microscopic polyangiitis indicate that these diseases are predominantly associated with Th2 patterns. Further studies elucidating the true nature of the polarization towards Th1 or Th2 in ANCA-associated vasculitis are clearly needed.  相似文献   

16.
Th1/Th2 balance in childhood idiopathic nephrotic syndrome   总被引:5,自引:0,他引:5  
AIMS: In view of the conflicting evidence of helper T cell type 1 (Th1) or type 2 (Th2) pattern of cytokine synthesis in childhood idiopathic nephrotic syndrome (INS) this study examined the balance of Th1 and Th2 which are characterized by intracellular cytokine production of interferon-gamma (IFNgamma) and interleukin-4 (IL-4), respectively. SUBJECTS AND METHODS: Sixteen children with steroid-sensitive INS (mean age 9.0 years) were included in this study, together with 15 healthy normal children (mean age 7.9 years) for the control group. Intracellular production of both IFNgamma and IL-4 in helper T cell (CD4+ cell) was investigated by a 3-color flow cytometry. RESULTS: The cross-sectional data showed no significant differences of percentages of Th0 (IFNgamma+ IL-4+ CD4+ cell), Th1 (IFNgamma+ lL-4- CD4+ cell) and Th2 (IFNgamma- IL-4+ CD4+ cell) in CD4+ cells (p > 0.05). The Th1/Th2 ratio during nephrotic relapse did not differ from those during nephrotic remission and in normal healthy children (p > 0.05). CONCLUSION: We conclude that there is no significant skew of Th1/Th2 balance in childhood INS and that the cardinal immunological abnormality does not lie in helper T cells but in other cells, such as suppressor/cytotoxic T cells, natural killer cells or monocytes/macrophage. To clarify the pathogenesis of INS, comprehensive studies for these cells would be worthwhile.  相似文献   

17.
目的:研究Th1/Th2细胞因子对同种异系小鼠心脏移植存活时间的影响.方法:使用野生型BALB/c小鼠作为供体,野生型B6小鼠、IL-4基因去除B6小鼠及IFN-γ基因去除B6小鼠作为受体,行腹部异位小鼠心脏移植.部分IL-4基因去除小鼠、IFN-γ基因去除小鼠联合应用α-半乳糖神经酰胺(α-galactosylceramide,α-GalCer),以获得更强的Th1/Th2偏移.比较移植物存活时间.向野生型、IL-4基因去除及IFN-γ基因去除B6小鼠腹腔内注射供体小鼠脾细胞,提取受体小鼠脾脏CD8<'8>T细胞行淋巴细胞毒试验.结果:IFN-γ基因去除组小鼠的移植物存活时间为(6.40±0.55)d,联合应用α-GalCer组移植物存活时间为(8.00±1.15)d.IL-4基因去除组小鼠的移植物存活时间为(8.00±1.00)d,联合应用α-GalCer组移植物存活时间为(8.60±1.34)d.淋巴细胞毒试验显示IFN-γ基因去除小鼠的CD8+T细胞毒性明显增强.结论:Th1/Th2细胞因子与排斥反应之间并不存在简单的对应关系.  相似文献   

18.
To investigate the change of Th1, Th2, Th3 cytokines during early liver transplantation. IFN-r, IL-4, TGF-beta production by CD4+ T cells were determined by fluorescence activated cell sorter analysis. Comparing the acute rejection with the non-acute rejection groups on the 7th, 14th, and 28th day showed that high interferon-gamma production associated with acute rejection in the early posttransplant period. There was no evidence of significant changes in interleukin-4 and transforming growth factor beta (TGF-beta) levels between non-acute rejection groups with acute rejection groups. Th1 cytokine high production is significantly associated with acute rejection in liver transplant recipients.  相似文献   

19.
20.
儿童原发性肾病综合征与Th1/Th2细胞失衡   总被引:8,自引:0,他引:8  
目的 探讨Th1/Th2细胞失衡在原发性肾病综合征(PNS)患儿发病中的作用。方法运用三色荧光标记法流式细胞术检测21例初发和16例缓解期NS患儿外周血Th1和Th2细胞百分率(%),并以10例正常儿童作对照。结果 正常儿童外周血Th1、Th2和Th0细胞百分率分别为(13.42±4.36)%,(2.53±1.97)%和(1.25±0.92)%。初发的PNS患儿均明显减低,分别为(2.34±2.09)%,(1.02±0.96)%和(0.40±0.38)%(P<0.05)。缓解期PNS患儿,分别为(11.96±4.75)%,(2.87±2.46)%和(1.31±0.87)%,与正常儿童比,差异均无显著性意义。由于初发的PNS患儿Th1细胞减低相对于Th2细胞而言更显著,因而导致Th1/Th2比值也较缓解期PNS患儿和正常儿童明显下降(2.43±2.65比4.17±2.32和5.41±2.77,P均<0.05)。结论 儿童PNS是一种以Th2细胞占优势的免疫介导的肾小球疾病,源于Th1细胞减低所致的Th1/Th2细胞失衡在原发性肾病综合征发病过程中可能起着重要作用。  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号