节点文献

Tim-1-Fc抑制同种移植物排斥反应的作用与机制

Effect and Mechanism of Tim-1-Fc in the Suppression of Allograft Rejection

【作者】 肖亮

【导师】 丁国善;

【作者基本信息】 第二军医大学 , 外科学, 2011, 博士

【摘要】 CD4T辅助细胞的活化、增殖和分化是抗原特异性免疫反应产生的关键环节。该反应的产生除了需要抗原递呈细胞(Antigen presenting cells, APCs)提供抗原肽MHC复合物与T细胞受体特异性结合外,还必须有APCs提供协同刺激分子如免疫球蛋白家族成员(CD80、ICOSL等)、TNF家族成员(CD40、OX40、4-1BB、CD27、LIGHT等)和多种细胞因子的参与。随着研究的深入,不断有新的分子被发现参与、调控了T细胞的活化过程。因此,T细胞的活化与调节仍然是基础免疫学研究的重点,也是移植免疫学关注的热点。T细胞免疫球蛋白粘蛋白(T cell immunoglobulin mucin,Tim)是2001年发现的表达在T细胞和DCs表面与细胞活化相关的新分子。小鼠TIM基因家族位于11B1.1,目前共发现8个基因,编码蛋白Tim-l~4和4个假想蛋白Tim-5~8。人类TIM基因家族只有3个成员,定位于染色体5q33.2,分别编码蛋白Tim-l, 3, 4(没有Tim-2)。Tim是一类具有共同基序的跨膜糖蛋白,其结构包括免疫球蛋白(Ig)样区、粘蛋白样区、跨膜区和胞内区。除Tim-4外,Tim-1,2,3的胞内区均含有酪氨酸激酶磷酸化位点,直接参与信号转导。早期研究发现,Tim-1表达于所有活化的CD4+ T细胞,并在其分化后仍高表达于Th2细胞,而低表达于Th1,Th17,CD11c+骨髓源性树突状细胞(Dendritic cells,DCs)和CD19+ B细胞。Tim-1的配体为Tim-4,与其它Tim蛋白不同,小鼠Tim-4只表达于APCs。它作为Tim-1的配体参与T细胞的活化和增殖,并能减少其凋亡。Tim-主要表达于Th1细胞和DCs,它与DCs上的Galectin-9结合后能导致Th1细胞的凋亡。然而,随着研究的深入,人们发现Tim-1和Tim-4在免疫调节中的作用比预想的更为复杂。最近的研究显示,小鼠Tim-4与人IgG1 Fc段融合蛋白(Tim-4-Ig)在不同的终浓度下能刺激或抑制培养体系中T细胞的活化。另外, Tim-4-Ig可以通过与小鼠初始CD4+ T细胞上的某种配体结合抑制其活化,经证实,这种配体并非Tim-1。然而,Tim-4-Ig的这种作用只局限于不表达Tim-1的小鼠初始T细胞,它不能抑制预先激活的T细胞增殖。上述研究提示Tim-4-Ig可以与T细胞上的不同配体结合,介导不同的免疫效应。此外,Tim-1的不同单克隆抗体(3B3和RMT1-10)也可以对T细胞活化产生相反的作用,这可能取决于它们与Tim-1结合的亲和力不同或对细胞骨架结构的影响不同。因此,进一步阐明Tim-1调节T细胞反应的机制,对于开发治疗自身免疫性疾病和移植物排斥反应的新药物,具有重要的意义。基于上述研究,本课题拟进一步:(1)合成人源化Tim-1胞外段和IgG1 Fc段融合蛋白(Tim-1-Fc),检测其与小鼠不同种类细胞的结合情况,验证其是否具有交叉活性,并判定这种可能的结合是否是Tim-4依赖性的;(2)体外研究Tim-1-Fc对anti-CD3, anti-CD28 mAbs介导的小鼠CD4+ T细胞活化,增殖,凋亡,细胞周期和对同种DCs介导的混合淋巴细胞反应(Allogeneic mixed lymphocyte response, allo-MLR)的影响,探讨其胞内信号转导途径;观察其对调节性T细胞(Regulatory T cells, Tregs)增殖和叉头状转录因子p3(Forkhead transcription factor p3, Foxp3)表达的影响;(3)通过腹腔注射Tim-1-Fc(对照组注射PBS和人IgG1),阻断Tim-4-Tim-1介导的T细胞活化,观察其对小鼠颈部异位心脏移植物生存期的影响,初步揭示其抑制移植排斥反应的作用和体内机制;通过本课题研究,有望进一步认识Tim-1-Fc分子在T细胞介导的免疫反应和抑制同种移植物排斥反应中的作用,为寻找临床免疫抑制治疗的新靶点提供实验依据。目的:合成人源化Tim-1胞外段和IgG1 Fc段融合蛋白(Tim-1-Fc),检测其与小鼠不同细胞结合情况,验证其是否具有交叉活性,并判定这种可能的结合是否是Tim-4依赖性的。方法:从基因库中搜索人Tim-1胞外段和IgG1 Fc段的cDNA序列,克隆到哺乳动物细胞表达载体pcDNA3.1上,利用转染试剂将重组质粒转入中国仓鼠卵巢细胞(Chinese hamster ovary, CHO)中,通过Western-blot检测Tim-1-Fc表达情况,经亲和、离子交换等多种层析方法纯化蛋白。采用流式细胞仪(Flow cytometry, FCM)检测Tim-1-Fc与未接受刺激的或刺激3天后的小鼠DCs,CD4+CD25- T细胞,CD4+CD25+ T细胞(Natural regulatory T cells, nTregs)结合情况,加入Tim-4单抗,阻断可能的Tim-4-Tim-1-Fc结合。结果:经Western-blot检测,合成的Tim-1-Fc分子量约62KD,纯度>92%。Tim-1-Fc不结合DCs,nTregs和未活化的CD4+CD25- T细胞,但能选择性结合活化后的CD4+CD25- T细胞(Effector T cells, Teffs),且该作用不被anti-Tim-4 mAb阻断。结论:Tim-1-Fc构建成功。小鼠Teffs上存在人Tim-1的新型配体。目的:观察Tim-1-Fc在体外对CD4+ T细胞活化,增殖,凋亡和细胞周期,胞内信号转导,allo-MLR以及nTregs增殖、Fxop3表达的影响。方法:在CD4+ T细胞培养体系中加入不同终浓度的Tim-1-Fc(1,5,10μg/ml),按不同比例(1:40,1:20,1:10)将DC与CD4+ T细胞混合培养,采用3H掺入法检测细胞增殖,CSFE检测细胞分裂,ELISA检测培养上清液中Th1/Th2细胞因子分泌情况,PI/Annexin V双染色法检测各组CD4+ T细胞凋亡的比例,RT-PCR法检测Tim-1-Fc对nTregs内Foxp3基因转录的影响,Western-blot检测Tim-1-Fc对细胞周期蛋白Cdk2,Cdk4,p27kip表达及anti-CD3(2μg/ml), anti-CD28(1μg/ml)mAbs刺激后不同时间点(5, 15, 30min)CD4+ T细胞内信号分子AKT,ERK1/2磷酸化情况的影响。结果:Tim-1-Fc不但能抑制CD4+ T细胞表面活化标志CD25,CD69表达、抑制细胞分裂增殖,该作用不依赖于Tim-4(RT-PCR和FCM检测也未发现小鼠T细胞表达Tim-4);还能显著下调Cdk2,Cdk4而增加p27kip表达,使细胞停滞在G1期;抑制CD4+ T细胞内AKT,ERK1/2磷酸化;抑制allo-MLR,诱导T细胞对同种抗原的低反应性,减少IL-2,IFN-γ而促进IL-10的分泌。但Tim-1-Fc对CD4+ T细胞凋亡,nTregs增殖和Foxp3表达无明显影响。结论:Tim-1-Fc在体外能通过与T细胞表面新型受体结合抑制CD4+ T细胞活化、增殖和Th1源性细胞因子表达,使细胞停滞在G0/G1期,并下调T细胞内信号分子AKT,ERK1/2磷酸化水平。还能抑制小鼠allo-MLR,诱导T细胞对同种抗原的低反应性;但它不影响CD4+ T细胞凋亡,nTregs增殖和Foxp3表达。目的:观察Tim-1-Fc对小鼠心脏移植物存活期的影响并探讨其抑制排斥反应的体内机制。方法:采用BALB/c→C57BL/6小鼠颈部异位心脏移植模型,将受体分为3组(每组n=10):组1,心脏移植+PBS治疗组;组2,心脏移植+ hIgG1(10μg/g/天术后06天)治疗组;组3,心脏移植+Tim-1-Fc(10μg/g/天术后06天)腹腔注射治疗组。专人记录心脏移植物存活时间。于术后第7天处死小鼠,收集血液、心脏、脾脏标本。Real-time PCR检测心脏移植物内淋巴细胞表面分子CD3, CD11b和炎性细胞因子IL-2, IFN-γ, IL-4, IL-10和转录因子Foxp3表达情况,病理检查心脏移植物内炎性细胞浸润和组织破坏情况。磁珠分选受体小鼠脾脏CD4+ T细胞,用BALB/c小鼠DCs刺激,3H掺入法检测其增殖情况,ELISA检测Th1,Th2细胞因子水平。FCM检测受体小鼠脾脏中CD4+Foxp3+调节性T细胞占总CD4+T细胞比例,以及CD4+CD25- T细胞培养体系中加入Tim-1-Fc后Foxp3的表达变化。结果:三组小鼠心脏移植物存活时间分别为6.5±0.5,6.7±0.4和17.1±1.1天。Tim-1-Fc治疗组移植物内IL-2、IFN-γ、CD11b、CD3 mRNA表达水平显著较低,病理损害亦明显减轻,但IL-10水平升高(P<0.05);Tim-1-Fc组受体脾脏来源CD4+ T细胞接受同种异基因DCs刺激后增殖水平显著较低(P<0.05);受体小鼠脾脏中CD4+Foxp3+调节性T细胞占总CD4+T细胞比例显著升高(P<0.05),但Tim-1-Fc并不能直接诱导CD4+CD25- T细胞表达Foxp3。结论:Tim-1-Fc能减轻小鼠心脏移植物Th1细胞因子表达和炎性细胞浸润,促进Th2细胞因子表达,降低受体脾脏内供体反应性T细胞对同种抗原的反应能力,显著延长移植物存活期;Tim-1-Fc虽不能直接诱导Tregs,但它通过调节Th1/Th2平衡向Th2发展,提高脾脏内CD4+Foxp3+ T细胞比例,调节Teffs/Tregs平衡向Tregs偏移,成功抑制了小鼠心脏移植物排斥反应,使其有望成为抗排斥治疗的新药物。

【Abstract】 Activation, proliferation and differentiation of CD4+ T helper cell are crucial elementsof antigen-specific immune response. This process needs not only the specific binding of Tcellreceptors with antigenic peptide/MHC complexes provided by antigen-presenting cells(APCs), but also needs co-stimulatory molecules such as immunoglobulin family members(CD80, ICOSL, etc.), TNF family members (CD40, OX40, 4-1BB, CD27, LIGHT, etc.)and a variety of inflammatory cytokines. With the in-depth research, new co-stimulatorymolecules have being discovered to be involved in T cells activation. Thus, the activationand regulation of T cells remain major scientific issues to basic immunology research andhot spots to transplantation immunology research.T cell immunoglobulin mucin (Tim) is a newly discovered molecular expressed on thesurface of T cells and APCs and is associated to T-cell activation. Mouse TIM genes arelocated on chromosome 11B1.1, encoding protein Tim-14, and four hypothetical proteinTim-58. Human TIM genes are located on chromosome 5q33.2, encoding protein Tim-l, 3,4, but not Tim-2. Tim represents a group of transmembrane glycoproteins with commonmotifs: immunoglobulin (Ig)-like domain, mucin-like domain, transmembrane region andintracellular domain. In addition to Tim-4, Tim-1, 2 and 3 all contain an intracellulartyrosine kinase phosphorylation site, which is directly involved in the intracellular signaltransduction.Tim-1 is expressed on all activated CD4+ T cells, and highly expressed on Th2 cellsafter differentiation, while lowly expressed on Th1, Th17, CD11c+ bone marrow-deriveddendritic cells (DCs) and CD19+ B cells. Tim-1 is the natural ligand for Tim-4, which isonly expressed on APCs and is involved in T cell activation, proliferation, and apoptosisinhibition. Tim-3 is mainly expressed on Th1 cells and DCs, and after cross-linking withGalectin-9, it can lead to the apoptosis of Th1 cells.However, the mechanisms for immune regulation by Tim-1 and Tim-4 are morecomplex than initially expected. Recent studies showed that Tim-4-Ig may either stimulateor inhibit T cell proliferation depending on its concentration. Moreover, Tim-4-Ig wasdemonstrated to inhibit naive mouse CD4+ T cell activation through a novel ligand otherthan Tim-1, as such an inhibitory effect of Tim-4-Ig was specific to naive T cells that do notexpress Tim-1, and the effect disappeared in pre-activated T cells. This suggests thepossibility that the opposite effect of Tim-4-Ig on T cell activation observed in the previous studies could be resulted from its engagement with different receptors expressed on T cells.On the other hand, anti-Tim-1 mAbs (3B3 and IRM-10) were also found to mediate either astimulatory or an inhibitory effect on T cell activation depending on their binding affinity toTim-1 or their influence on the cytoskeleton. Thus, further elucidation of the role of Tim-1in regulating T cell responses is highly important for developing novel therapeuticstrategies targeting Tim-1 for the treatment of autoimmune diseases and allograft rejection.Based on the above studies, further efforts of this study include: (1) Synthesization ofhuman Tim-1 excellular domain and IgG1 Fc domain fusion protein (Tim-1-Fc). To analyzethe biding activity of Tim-1-Fc with different kinds of mouse cell types such asunstimulated or stimulated dendritic cells (DCs), CD4+CD25- T cells and CD4+Foxp3+regulatory T cells (nTregs) to identify the cross-reactive, and to test whether this bindingwas Tim-4 dependent; (2) Exploration of the influence of Tim-1-Fc on allogeneic mixedlymphocyte response, activation and proliferation of CD4+ T cells, cells cycle and apoptosisevoked by anti-CD3 and anti-CD28 mAbs. Investigation of the intracellular signalingpathways in CD4+ T cells after ligation with Tim-1-Fc. Observation of its effect on theproliferation and Foxp3 expression of nTregs. (3) Intraperitoneal injection of Tim-1-Fc tomice that receive heterotopic cardiac transplantation, while PBS and hIgG1 treatment wereused as controls. The mechanisms of Tim-1-Fc in suppressing allograft acute rejection areto be explored. Through this research, we could get a better understanding of human Tim-1in mouse T cell-mediated immune responses and allograft rejection suppression, which mayprovide a new target for clinical immunosuppressive therapy.Objective: To synthesize humanized excellular domain of Tim-1 and IgG1 Fc domainfusion protein (Tim-1-Fc). To analyze the biding activity of Tim-1-Fc with different kindsof mouse cell types such as DCs, CD4+CD25- T cells and nTregs, and to test whether thisbinding activity was Tim-4 dependent. Methods: cDNA sequences of human Tim-1extracellular domain and IgG1 Fc fragment were got from the gene bank and cloned intothe mammalian cell expression vector pcDNA3.1, the recombinant plasmids weretransformed into CHO cells using transfection reagent. Tim-1-Fc expression was detected by SDS-PAGE and Western-blot, and purified by affinity column. The binding activity ofTim-1-Fc (5μg/ml) with fresh isolated or activated DCs, CD4+CD25- T cells and nTregswas labeled using mouse anti-human IgG1(Fc)-FITC and measured by FCM. Anti-Tim-4mAb (10μg/ml) was added to block the possible Tim-4-Tim-1-Fc engagement. ResultsResults:Western-blot test revealed that Tim-1-Fc molecular weight was about 62KD, purity was>92%. Tim-1-Fc could not bind to unstimulated or stimulated DCs or nTregs, but couldselectively bind to activated CD4+CD25- T cells (Teffs), and this effect could not bereversed by anti-Tim-4 mAb. Conclusion: Tim-1-Fc was constructed successfully. Theremust be a novel ligand for human Tim-1 expressed on mouse Teffs other than Tim-4.Objective: To test the influence of Tim-1-Fc on allo-MLR and on the activation,proliferation, cell cycles, apoptosis and Fxop3 expression of CD4+ T cells; to explore theTim-1-Fc-triggered intracellular signaling pathways in CD4+ T cells. Methods: Differentconcentrations of Tim-1-Fc (1, 5, 10μg/ml) and different proportions (1:40, 1:20, 1:10) ofDC:T-cell were added to test the influence of Tim-1-Fc on allo-MLR. 3H-TdR, CFSE andELISA were used to assay the proliferation and cytokines production of CD4+ T cells coculturedwith Tim-1-Fc. FCM was used to test the influence of Tim-1-Fc on the apoptosis(PI/Annexin V) and Foxp3 expression of CD4+ T cells. Western blot was used to explore thephosphorylation of intracellular signal moleculars. Results: Tim-1-Fc significantlyinhibited CD4+ T cells activation, proliferation and allo-MLR but not the apoptosis. Theseeffects were Tim-4-independent. Also, Tim-1-Fc reduced IL-2,IFN-γwhile promoted IL-10production, and it negatively regulated Cdk2 and Cdk4 expression while increased p27kipexpression. Moreover, the phosphorylation of AKT and ERK1/2 in CD4+ T cells was alsoinhibited, but the proliferation of nTregs or Foxp3 expression were not inhibited by Tim-1-Fc. Conclusion: Tim-1-Fc was able to inhibit the activation, proliferation, differentiation,Th1 cytokines production and allo-MLR of CD4+ T cells, and to suppress thephosphorylation of AKT, ERK1/2 in CD4+ T cells, but has not influence on the proliferation and Foxp3 expression of nTregs in vitro.Objective: To observe the influence of Tim-1-Fc on cardiac allograft survival andallograft acute rejection and to explore the in vivo mechanisms. Methods:BALB/c→C57BL/6 mouse model of cervical heterotopic heart transplantation wasperformed, the recipients were divided into 3 groups according to the different treatments(for each group, n=10): Group 1, transplantation + PBS treatment group; Group 2,transplantation + human IgG1 treatment group (10μg/g/day, intraperitoneal injection, 0-6days post-operatively); Group 3, transplantation+Tim-1-Fc treatment group (10μg/g/day,intraperitoneal injection, 0-6 days post-operatively). The survival time of cardiac allograftwas recorded. At the 7th day after transplant, the mice were killed, the bloods, hearts andspleens were collected. ELISA test for blood cytokines (IL-2, IFN-γ, IL-4 and IL-10) levelsand intragraft RT-PCR detection for inflammatory cytokines production, lymphocytessurface molecules (CD3 and CD11b) and Foxp3 expression were conducted. Recipients’splenic CD4+ T cells were isolated by magnetic activated cell sorting and stimulated withallogenetic DCs, the proliferation was detected by using 3H thymidine incorporation using aliquid scintillation counter. The ratio of Tregs in recipient mice spleens was also assessed.CD4+CD25- T cells were isolated and cultured with Tim-1-Fc and/or recombinant humanTGF-β1,then the expression of Foxp3 were determined by FCM. Results: Recipientstreated with Tim-1-Fc showed significantly prolonged allograft survival (17.1±1.1 days)compared to those treated with PBS (6.5±0.5 days; P<0.01) or control hIgG1 (6.7±0.4 days;P<0.01). Histological analysis of heart grafts at POD 7 revealed severe mononuclear cellsinfiltration and myocardial necrosis in the control groups, whereas almost normal tissuestructure with minimal mononuclear cell infiltration was seen in Tim-1-Fc-treated mice.Tim-1-Fc significantly reduced the intragraft transcription for IL-2, IFN-γ, CD3 and CD11b. The proliferation of CD4+ T cells in response to allogenetic DCs was significantlyinhibited in Tim-1-Fc treated group. Moreover, the proportion of CD4+Foxp3+ Tregs insplenic CD4+ T cells was significantly increased, although Tim-1-Fc did not induce Foxp3expression in CD4+CD25- T cells. ConclusionConclusion: Tim-1-Fc was able to prolong mouse heartallograft survival though reducing the inflammatory cells infiltration and Th1 cytokinesproduction in the graft and decreasing the proliferative capability of donor reactive T cellsto allogenetic antigens, while increasing the proportion of Tregs in the spleen, thus shiftingthe balance of Th1/Th2 towards Th2 and the balance of Tregs/Teffs towards regulators,although it did not induce Foxp3 expression in CD4+CD25- T cells directly. These madeTim-1-Fc become a promising drug for anti-rejection therapy.

节点文献中: 

本文链接的文献网络图示:

本文的引文网络