Tim3和Tim4在小鼠心脏移植模型中的免疫调节作用机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的建立简便而高效的分离小鼠心脏移植物内浸润淋巴细胞的方法,使用流式细胞仪检测移植心内浸润淋巴细胞的亚型。
     方法建立小鼠颈部心脏移植模型,分实验组(同种异体移植组:Balb/c小鼠为供体,C57BL/6小鼠为受体)和对照组(同系移植组:供受体均为C57BL/6小鼠)。移植术后3天,5天和7天获取移植心,将其中一部分移植物进行病理切片观察排斥反应情况,剩余移植物剪碎并采用改进的Ⅱ型胶原酶消化法(250 U/ml,30-40 min)消化心脏,然后用Ficoll密度梯度离心法(800g×20 min)分离单个核细胞,以钙离子荧光染料Indo-1分析细胞活性,最后以荧光标记抗体CD4, CD8, CD44, CD62L, Foxp3进行流式细胞染色,流式细胞仪检测移植物内浸润淋巴细胞亚群组成。
     结果移植物内分离的单个核细胞数量稳定在1×106以上,术后第7天数量达到最高峰,分离所得的单个核细胞数量与排斥反应严重程度相关。淋巴细胞占分离所得单个核细胞的比例为(31.9±2.3)%,活性为(95.1±2.1)%。移植物内浸润的T细胞大部分为效应性T细胞,CD4+/CD8+比值随着排斥反应的进行逐渐降低。
     结论本法单用胶原酶消化移植心,采用Ficoll密度梯度离心法,减少了对心脏移植物内浸润淋巴细胞的损伤,获得的细胞可进一步用于流式分析其表型,为直接检测浸润到移植物内的淋巴细胞亚型提供一种高效和可靠的方法,是一种值得推广的免疫学研究手段。
     目的检测Tim3 mRNA在移植后移植物内的表达变化及Tim3在受者体内不同部位T淋巴细胞上表达,探讨其与急性排斥反应的关系。
     方法建立小鼠心脏移植模型,分实验组(同种异体移植组:Balb/c小鼠为供体,C57BL/6小鼠为受体)和对照组(同系移植组:供受体均为C57BL/6小鼠)。移植术后3天,5天,7天和9天获取移植物,采用实时定量RT-PCR检测Tim3 mRNA在移植物内的表达变化。术后3天和6天分离受者外周血、脾脏、引流淋巴结、移植物内淋巴细胞,流式细胞仪检测Tim3阳性细胞在CD4+和CD8+T细胞中的比例。
     结果同基因组移植物内Tim3表达很低。异基因组移植术后第3天移植物内Tim3mRNA的表达明显升高,并且Tim3 mRNA的表达随着排斥反应的进行而显著升高(P<0.01),于移植物完全排斥前达到最高峰,随后表达逐渐降低。移植术后受者外周血和脾脏内Tim3阳性细胞比例无明显变化(P>0.05),引流淋巴结内Tim3+/CD4+比值轻度升高(P<0.05),移植物内Tim3+/CD4+和Tim3+/CD8+比值显著升高(P<0.01)。移植术后第3天和第6天引流淋巴结内Tim3+/CD4+比值差异无统计学意义(P>0.05),而术后第6天移植物内Tim3+/CD4+和Tim3+/CD8+比值均显著高于第3天(P<0.01)。
     结论移植物内Tim3 mRNA的表达与小鼠完全异基因心脏移植排斥反应的进展动态相关。异基因组受者移植物引流淋巴结和移植物内Tim3+细胞比例升高,以移植物内升高更为显著。
     目的观察Galectin-9对同种异体心脏移植物存活时间的影响,探讨Galectin-9激活Tim3-Tim3L通路在小鼠心脏移植模型中的免疫调节作用。
     方法建立小鼠同种异体心脏移植模型,分实验组和对照组。实验组受者术后连续7天给予稳定的Galectin-9蛋白,对照组给予PBS。观察心脏移植物存活时间。术后第7天获取实验组和对照组心脏移植物,病理切片观察排斥情况,免疫组化观察移植物内浸润CD4+和CD8+T细胞数量。实时定量RT-PCR检测移植物内Tim3,IFN-γ和IL-17mRNA的表达。流式细胞仪检测引流淋巴结和移植物内Tim3+细胞比例及外周血中Th1和Th17细胞比例。
     结果Galectin-9蛋白处理组心脏移植物平均存活时间为22.7天,对照组仅为7.2天,Galectin-9治疗组移植物存活时间显著延长(P<0.05)。病理切片显示Galectin-9治疗组移植物排斥反应明显减轻,浸润的CD4+和CD8+T细胞明显减少。Galectin-9治疗后移植物内Tim3、IFN-γ和IL-17mRNA表达减少。引流淋巴结和移植物内Tim3+细胞比例降低,外周血中Th1和Th17细胞比例也降低。
     结论短期Galectin-9治疗能显著延长同种异体心脏移植物存活时间,其机制是减少移植物内淋巴细胞浸润,并通过触发Tim3+细胞程序性死亡而降低受者体内Th1和Th17细胞反应。
     目的观察阻断Tim4对小鼠心脏移植物存活时间的影响,探讨Tim4在Tim4-Timl通路诱导免疫耐受中的作用。
     方法建立小鼠同种异体心脏移植模型,给予受者抗Tim4抗体,观察其对移植物存活时间的影响,体外混合淋巴细胞培养检测抗Tim4抗体对同种反应性T细胞增殖的作用。比较Tim4基因缺陷和野生型心脏移植物在受体体内的存活时间,病理切片判断排斥情况,免疫荧光染色观察移植物内浸润淋巴细胞。实时定量RT-PCR检测移植物内免疫相关基因表达,流式细胞仪检测移植物浸润淋巴细胞亚型的变化。过继输注受者体内效应性T细胞和调节性T细胞给Babl/c SCID小鼠,观察其对皮肤移植物存活时间的影响。给予小剂量雷帕霉素观察其对Tim4基因缺陷的移植物存活时间的影响,并通过术前给予抗CD25抗体清除受者体内Treg明确Treg在本实验中的作用。
     结果抗Tim4抗体能抑制混合淋巴细胞培养体系中同种反应性T细胞增殖,延长移植物存活时间。供体Tim4基因缺陷明显减轻排斥反应,减少移植物内效应性T细胞浸润,增加调节性T细胞数量,从而显著延长移植物存活时间(P<0.01)。Tim4基因缺陷能减少活化的效应性T细胞而增加Treg的数量,但是对效应性T细胞和Treg的功能无明显影响。联合小剂量雷帕霉素能诱导Tim4基因缺陷移植物长期存活。Tim4基因缺陷或/和小剂量雷帕霉素延长移植物存活时间依赖Treg的存在。
     结论Tim4在同种异体免疫反应中发挥重要的免疫调节作用,阻断Tim4-Timl通路能明显延长心脏移植物存活时间。
Objective To introduce a simple and efficient method for isolation of intragraft infiltrating lymphocytes and examine the subsets of lymphocytes by immunofluorescent staining and flow cytometer analysis.
     Methods The murine cardiac transplant model was built. Experimental group (Balb/c to C57BL/6) and control group (C57BL/6 to C57BL/6) were designed in this study. Cardiac grafts harvested on 3,5 and 7 days after operation respectively were cut into small pieces. These pieces were then digested with modified enzyme digestion singly using collagenaseⅡ(250 U/ml,30-40 min). Mononuclear cells were isolated by Ficoll density gradient centrifugation (800g×20 min). Ca2+fluorescence dye Indo-1 was used to analyze cell viability. Histological slices were made to observe the rejection. Subsets of lymphocytes were identified by flow cytometry after surface antigen and intracellular cytokine immunofluorescent staining.
     Results The harvested mononuclear cells were more than 1×106, which contained (31.9±2.3)%lymphocytes with viability of (95.1±2.1)%. The number of the mononuclear cells from the graft reached a peak on 7 days after transplantation and was closely correlated with the severity of rejection. Flow cytometry can analyze surface and intracellular antigens of the intragraft infiltrating lymphocytes at the same time. Most of the intragraft infiltrating lymphocytes were effector T cells. The ratio of CD4+/CD8+decreased gradually as the progression of rejection.
     Conclusion The modified method of isolating intragraft infiltrating lymphocytes with high viability is simple, reliable and efficient. It can be used widely in transplant immunology research.
     Objective To detect the changes of Tim3 expression in cardiac graft and T lymphocytes from different sites in mice heart-transplant recipients and explore the relationship of Tim3 expression level with acute rejection.
     Methods The murine cardiac transplant model was built. Experimental group (Balb/c to C57BL/6) and control group (C57BL/6 to C57BL/6) were designed in this study. Cardiac grafts were harvested on 3,5,7 and 9 days after operation respectively. Real-time quantitative RT-PCR was used to detect the local expression of Tim3 mRNA in graft. Lymphocytes were isolated from peripheral blood, spleen, draining lymph nodes and allograft on 3 days and 6 days after transplantation. The expression of Tim3 on T lymphocyte subsets CD4+and CD8+T cells was detected by flow cytometry.
     Results The expression of Tim3 was very low in isograft group. In allograft group, as the acute rejection occurred, the Tim3 expression increased significantly on 3 days after transplantation, which was related to the severity of rejection (P<0.01). It reached its peak on 7 days and then decreased gradually after the completely rejection. No significant increase of Tim3+cells was observed in peripheral blood and spleen after transplantation (P > 0.05). The expression of Tim3 on CD4+and CD8+of graft infiltrating T cells was obviously increased (P<0.01). While the ratio of Tim3+/CD4+had a slight elevation in draining lymph node (P<0.05). There was no obvious difference in Tim3+/CD4+ratio in draining lymph node between 3 days and 6 days (P> 0.05). But the ratios of Tim3+/CD4+ and Tim3+/CD8+in graft infiltrating T cells on 6 days were significantly (P<0.01) higher than that on 3 days.
     Conclusion The expression of Tim3 incresed in cardiac graft and T lymphocytes from draining lymph node and allograft after transplantation, which was correlated with the progression of acute rejection in mice.
     Objective To investigate the effect of Tim3 ligand (Galectin-9) on the survival of allograft and explore its immunomodulating role in an allogeneic heart transplant model.
     Methods Murine cardiac transplant model from BALB/c to C57BL/6 was built. The study was divided into experimental group and control group. Experimental group were administered with Galectin-9 for 7 days since day 1 posttransplant while control group with PBS. The complete cessation of heart beating was defined as the observation endpoint. The survival of allograft was observed in two groups. Histology and immunohistochemistry were performed to estimate the severity of rejection on 7 days posttransplant. Real-time quantitative RT-PCR was utilized to analyze the Tim3, IFN-γand IL-17 mRNA expression in allograft. The phenotype and cytokine profile of lymphocytes from peripheral blood, draining lymph nodes and allograft was analyzed by flow cytometry.
     Results The survival of allograft treated with Galectin-9 (mean survival time, MST= 22.7 days) was significantly prolonged compared with the control group (MST= 7.2 days), which was associated with reduced infiltration of CD4 and CD8 lymphocytes in allograft. Galectin-9 decreased the expression of Tim3, IFN-γand IL-17 mRNA in allograft notably. The proportion of Tim3 positive cells in draining lymph nodes and allograft and Thl and Th17 positive cell in peripheral blood was obviously lower than the control group.
     Conclusion A short-course administration of Galectin-9 significantly prolonged the survival of fully allogeneic cardiac allografts, which was associated with the suppression of Th1 and Th17 immune responses.
     Objective To study the effect of Tim4 blockade on the survival of cardiac allograft and explore the role of Tim4 in Tim4-Tim1 pathway in inducing immune tolerance.
     Methods Murine cardiac transplant model from BALB/c to C57BL/6 was built. The survival was compared between the recipients treated with anti-Tim4 Ab and PBS. MLC was used to assess the effect of anti-Tim4 Ab on cell proliferation. The survival of Tim4-/-and wild-type graft in recipients was observed. Histology was performed to estimate the severity of rejection. Immunofluorescent staining was carried out for detecting lymphocytes in allograft. The expression of immune-related genes was determined by Real-time quantitative RT-PCR. The subsets of intragraft infiltrating lymphocytes were analyzed by flow cytometry. Effector T cells and Tregs from the recipients were sorted by FACS. These cells were then transfused into Balb/c SCID mice followed by implantation of skin allograft. The rejection and survival of the allograft were observed. The effect of low dose rapamycin on the survival of Tim4-/- allograft was studied. The role of Tregs in the model was evaluated by administering anti-CD25 mAb before transplantation.
     Results Recipients conditioned with anti-Tim4 mAb exhibited prolonged cardiac allograft survival with suppressing the T cell proliferation in MLC. The survival of Tim4-/- allograft was significantly prolonged compared with wild-type group. The prolonged survival time was associated with reduced lymphocytes and increased Treg in allografts. Strikingly, when combined with a low dose of rapamycin, Tim4-/- heart allografts survived indefinitely. The absence of Tim4 led to the decreased recruitment, but not decreased effect, in the effector T cells compartment. Similarly, it increased recruitment, but not increased function, in the Treg compartment. Moreover, depletion of Treg with anti-CD25 Ab completely abrogated the tolerogenic effects of Tim4 deficiency alone or when combined with rapamycin.
     Conclusion A blockade of the Tim4-Tim1 axis improves allograft survival as a monotherapy and synergizes with low-dose rapamycin to induce complete transplantation tolerance by enhancing immunoregulation.
引文
1. Wasowska B, Baldwin WM, Howell DN, et al. The effects of donor-specific blood transfusion enhancement of rat renal allografts on cytotoxic activity and phenotypes of peripheral blood lymphocytes, splenocytes, and graft-infiltrating cells. Transplantation, 1991,51:451-459.
    2.2. Beissert S, Schwarz A, Schwarz T. Regulatory T cells. J Invest Dermatol,2006,126 (1):15-24.
    3. Wang Y, Afanasyeva M, Hill SL, et al. Characterization of murine autoimmune myocarditis induced by self and foreign cardiac myosin. Autoimmunity,1999,31: 151-162.
    4. Penninger JM, Neu N, Timms E, et al. The induction of experimental autoimmune myocarditis in mice lacking CD4 or CD8 molecules. J Exp Med,1993,178:1837-1842.
    5. Hirasawa M, Deguchi H, Ukimura A, et al. Immunologic interaction between infiltrating eosinophils and T lymphocytes in murine spontaneous eosinophilic myocarditis. Int Arch Allergy Immunol,2003,130:73-81.
    6. Braylan RC. Attributes and applications of flow cytometry. Ann.Clin. Laboratory Sci, 1983,13,379-384.
    7. Afanasyeva M, Georgakopoulos D, Belardi DF, et al. Quantitative analysis of myocardial inflammation by flow cytometry in murine autoimmune myocarditis: correlation with cardiac function. Am J Pathol,2004,164(3):807-815.
    8. Chen ZH. A technique of cervical heterotopic heart transplantation in mice. Transplantation,1991,52:1099.
    9. Stinn JL, Taylor MK, Becker G, et al. Interferon-gamma-secreting T-cell populations in rejecting murine cardiac allografts:assessment by flow cytometry. Am J Pathol 1998, 153:1383-1392.
    10.林清泉,李懿,涂意辉,等.胶原酶阶梯消化法体外培养成骨细胞的研究.中华实验外科杂志,2001,18:359-360.
    11.于聪慧,冷希圣,魏玉华,等.胶原酶及灌流时间对大鼠肝细胞分离效果的影响.中华实验外科杂志,2000,17:541-542.
    12. Egorova MV, Afanaspev SA, Popov SV. A simple method for isolation of cardiomyocytes from adult rat heart. Bull Exp Biol Med,2005,140 (3):370-373.
    13.李温斌,陈室田.心肌细胞的分离、培养及其应用.心肺血管病杂志,1994,13(4):239-242.
    1. Monney L, Sabatos CA, Gaglia JL, et al. Thl-specific cell surface protein Tim3 regulates macrophage activation and severity of an autoimmune disease. Nature,2002, 415:536-541.
    2. Sanchez Fueyo A, Tian J, Picatella D, et al. Tim3 inhibits T helper type-2 mediated auto and alloimmune responses and promotes immunological tolerance. Nat Immunol,2003,4 (11):1093-1101.
    3. Sabatos CA, Sumone CK, Cha E, et al. Interaction of Tim3 and Tim3 ligand regulates T helper type 1 responses and induction of peripheral tolerance. Nat Immunol,2003, 4(11):1102-1110.
    4. Anderson, A. C. et al. Promotion of tissue inflammation by the immune receptor Tim-3 expressed on innate immune cells. Science,2007,318:1141-1143.
    5. Dardalhon V, Korn T, Kuchroo VK, et al. New roles for Tim family members in immune regulation. Nat Rev Immunol,2008,8(8):577-80.
    6. Field EH, Gao Q, Chen N, et al. Balancing the immune system for tolerance:A case for regulatory cells. Transplantation,1997,64:1-7.
    7. Renesto PG, Ponciano VC, Cenedeze MA, et al. High expression of Tim3 mRNA in urinary cells from kidney transplant recipients with acute rejection. Am J Transplant, 2007; 7:1661.
    8. Chen ZH. A technique of cervical heterotopic heart transplantation in mice. Transplantation,1991,52:1099.
    9. Su EW, Lin JY, Kane LP. Timl and Tim3 proteins in immune regulation. Cytokine, 2008,44(1):9-13.
    10. Roselynn R, Rosemarie D; Kuchroo VK, et al. The costimulatory role of Tim molecules. Immunological reviews,2009,229(1):259-70.
    11. He W, Fang Z, Wang F, et al. Galectin-9 Significantly Prolongs the Survival of Fully Mismatched Cardiac Allografts in Mice. Transplantation,88(6):782-790.
    1. Renesto PG, Ponciano VC, Cenedeze MA, et al. High expression of Tim3 mRNA in urinary cells from kidney transplant recipients with acute rejection. Am J Transplant, 2007,7:1661.
    2. Nakae S, Iwakura Y, Suto H, et al. Phenotypic differences between Th1 and Th17 cells and negative regulation of Thl cell differentiation by IL-17. J Leukoc Biol,2007,81: 1258.
    3. Khademi M, Illes Z, Gielen AW, et al. T Cell Ig and mucin domain containing molecule-3 (Tim3) and Tim1 molecules are differentially expressed on human Thl and Th2 cells and in cerebrospinal fluidderived mononuclear cells in multiple sclerosis. J Immunol,2004,172:7169.
    4. Sanchez-Fueyo A, Tian J, Picarella D, et al. Tim3 inhibits T helper type 1-mediated auto-and alloimmune responses and promotes immunological tolerance. Nat Immunol, 2003,4:1093.
    5.5 Meyers J H, Sabatos C A, Chakravarti S, et al.The Tim gene family regulates autoimmune and allergic diseases. Trends Mol Med,2005, 11(8):362-369.
    6. Seki M, Sakata KM, Oomizu S, et al. Beneficial effect of galectin-9 on rheumatoid arthritis by induction of apoptosis of synovial fibroblasts. Arthritis Rheum,2007,56: 3968.
    7. Tsuboi Y, Abe H, Nakagawa R, et al. Galectin-9 protects mice from the Shwartzman reaction by attracting prostaglandin E2-producing polymorphonuclear leukocytes. Clin Immunol,2007,124:221.
    8. Katoh S, Ishii N, Nobumoto A, et al. Galectin-9 inhibits CD44 hyaluronan interaction and suppresses a murine model of allergic asthma. Am J Respir Crit Care Med,2007, 176:27.
    9. Zhu C, Anderson AC, Schubart A, et al. The Tim3 ligand Galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol,2005,6:1245.
    10. Dardalhon V, Korn T, Kuchroo VK, et al. New roles for Tim family members in immuneregulation. Nat Rev Immunology,2008,8(8):577-80.
    11. Seki M, Oomizu S, Sakata KM, et al. Galectin-9 suppresses the generation of Th17, promotes the induction of regulatory T cells, and regulates experimental autoimmune arthritis. Clin Immunol,2008,127:78.
    12. Wang F, He W, Yuan J, et al. Activation of Tim3-Galectin-9 pathway improves survival of fully allogeneic skin grafts. Transpl Immunol,2008,19:12.
    13. Wang F, He H, Zhou H, et al. The Tim3 ligand Galectin-9 negatively regulates CD8+ alloreactive T cell and prolongs survival of skin graft. Cellular Immunol,2007, 250:68-74
    14. Chen ZH. A technique of cervical heterotopic heart transplantation in mice. Transplantation,1991,52:1099.
    15. Van Loosdregt J, van Oosterhout MF, Bruggink AH, et al. The chemokine and chemokine receptor profile of infiltrating cells in the wall of arteries with cardiac allograft vasculopathy is indicative of a memory T-helper 1 response. Circulation,2006, 114:1599.
    16. Piccotti JR, Chan SY, VanBuskirk AM, et al. Are Th2 helper T lymphocytes beneficial, deleterious, or irrelevant in promoting allograft survival? Transplantation,1997,63: 619.
    17. Ueno T, Habicht A, Clarkson MR, et al. The emerging role of T cell Ig mucin 1 in alloimmune responses in an experimental mouse transplant model. J Clin Invest,2008, 118:742.
    18. Degauque N, Mariat C, Kenny J, et al. Immunostimulatory Timl specific antibody deprograms Tregs and prevents transplant tolerance in mice. J Clin Invest,2008,118: 735.
    19. Abdi R, Means TK, Ito T, et al. Differential role of CCR2 in islet and heart allograft rejection:Tissue specificity of chemokine/chemokine receptor function in vivo. J Immunol,2004,172:767.
    20. Kearley J, McMillan SJ, Lloyd CM. Th2-driven allergen-induced airway inflammation is reduced after treatment with anti-Tim3 antibody in vivo. J Exp Med,2007,204: 1289.
    21. Li J, Simeoni E, Fleury S, et al. Gene transfer of soluble interleukin-17 receptor prolongs cardiac allograft survival in a rat model. Eur J Cardiothorac Surg,2006,29: 779.
    22. Yuan X, Paez-Cortez J, Schmitt-Knosalla I, et al. A novel role of CD4+Th17 cells in mediating cardiac allograft rejection and vasculopathy. J Exp Med,2008,205:3133.
    23. Serada S, Fujimoto M, Mihara M, et al. IL-6 blockade inhibits the induction of myelin antigen-specific Th17 cells and Th1 cells in experimental autoimmune encephalomyelitis. Proc Natl Acad Sci USA,2008,105:9041.
    24. Tureci O, Schmitt H, Fadle N, et al. Molecular definition of a novel human galectin which is immunogenic in patients with Hodgkin's disease. J Biol Chem,1997,272: 6416.
    25. Keryer-Bibens C, Pioche-Durieu C, Villemant C, et al. Exosomes released by EBV-infected nasopharyngeal carcinoma cells convey the viral latent membrane protein 1 and the immunomodulatory protein galectin 9. BMC Cancer,2006,6:283.
    26. Klibi J, Niki T, Riedel A, et al. Blood diffusion and Th1-suppressive effects of Galectin-9-containing exosomes released by Epstein-Barr virus-infected nasopharyngeal carcinoma cells. Blood,2008,113:1957.
    27. Wada J, Ota K, Kumar A, et al. Developmental regulation, expression, and apoptotic potential of Galectin-9, a beta-galactoside binding lectin. J Clin Invest,1997,99:2452.
    28. Shimizu Y, Kabir-Salmani M, Azadbakht M, et al. Expression and localization of Galectin-9 in the human uterodome. Endocr J,2008,55:879.
    1. Meyers JH, Chakravarti S, Schlesinger D, et al. Tim4 is the ligand for Timl, and the Timl-Tim4 interaction regulates T cell proliferation. Nat Immunol,2005,6,455-464.
    2. Rodriguez-Manzanet R, Meyers JH, Balasubramanian S, et al. Tim4 expressed on APCs induces T cell expansion and survival. J. Immunol,2008,180:4706-4713.
    3. de Souza A J, Oriss T B, O'malley KJ, et al. T cell Ig and mucin 1 (Timl) is expressed on in vivo activated T cells and provides a costimulatory signal for T cell activation. Proc Natl Acad Sci USA,2005,102 (47):17113-17118.
    4. Umetsu SE, Lee WL, McIntire JJ, et al. Timl induces T cell activation and inhibits the development of peripheral tolerance. Nat Immunol,2005,6 (5):447-454.
    5. Degauque N, Mariat C, Kenny J, et al. Immunostimulatory Timl-specific antibody deprograms Tregs and prevents transplant tolerance in mice. J Clin Invest,2008, 118:735-741
    6. Ueno T, Habicht A, Clarkson MR, et al. The emerging role of T cell Ig mucin 1 in alloimmune responses in an experimental mouse transplant model. J Clin Invest,2008, 118:742-751
    7. Yuan X, Ansari J, Addio F, et al. Targeting Timl to overcome resistance to transplantation tolerance mediated by CD8+T17 cells. Proc Natl Acad Sci USA,2009, 106(26):10734-9.
    8. Chen ZH. A technique of cervical heterotopic heart transplantation in mice. Transplantation,1991,52:1099.
    9. Gill R, Tsung A, Billiar T, et al. Linking oxidative stress to inflammation:Toll-like receptors. Free Radic Biol Med,2010,48(9):1121-32.
    10. Kim IK, Bedi DS, Denecke C, et al. Impact of innate and adaptive immunity on rejection and tolerance. Transplantation,2008,86(7):889-94.
    11. Miller DM, Thornley TB, Pearson T, et al., TLR agonists abrogate co-stimulation blockade induced mixed chimerism and transplantation tolerance. Ann N Y Acad Sci, 2008,1150:149-51.
    12. Miller DM, Thornley TB, Greiner DL, et al. Viral infection:a potent barrier to transplantation tolerance. Clin Dev Immunol,2008,2008:742810.
    13. Palmer SM, Burch LH, Davis RD, et al. The role of innate immunity in acute allograft rejection after lung transplantation. Am J Respir Crit Care Med,2003,168(6):628-32.
    14. Palmer SM, Burch LH, Davis RD, et al. Innate immunity influences long-term outcomes after human lung transplant. Am J Respir Crit Care Med,2005,171(7): 780-5.
    15. Ducloux D, Deschamps M, Yannaraki M, et al. Relevance of Toll-like receptor-4 polymorphisms in renal transplantation. Kidney Int,2005,67(6):2454-61.
    16. Palmer SM, Burch LH, Mir S, et al. Donor polymorphisms in Toll-like receptor-4 influence the development of rejection after renal transplantation. Clin Transplant, 2006,20(1):30-6.
    17. Mariat C, Degauque N, Balasubramanian S, et al., Timl signaling substitutes for conventional signal 1 and requires costimulation to induce T cell proliferation. J Immunol,2009,182(3):1379-85.
    18. van Besouw NM, Zui jderwijk JM, Vaessen LM, et al. The direct and indirect allogeneic presentation pathway during acute rejection after human cardiac transplantation. Clin Exp Immunol,2005,141(3):534-40.
    19. Roncarolo MG and Battaglia M. Regulatory T-cell immunotherapy for tolerance to self antigens and alloantigens in humans. Nat Rev Immunol,2007,7(8):585-98.
    20. Xiao S, Najafian N, Reddy J, et al. Differential engagement of Timl during activation can positively or negatively costimulate T cell expansion and effector function. J Exp Med,2007,204(7):1691-702.
    21. Piccirillo CA and Shevach EM. Naturally-occurring CD4+CD25+immunoregulatory T cells:central players in the arena of peripheral tolerance. Semin Immunol,2004, 2(16):81-88.
    1. McIntire JJ, Umetsu SE, Akbari O, et al. Identification of Tapr (an airway hyperreactivity regulatory locus) and the linked Tim gene family. Nat Immunol,2001, 2:11092-1116.
    2. Kuchroo VK, Umet su DT, DeKruyff RH, et al. The Tim gene family:emerging roles in immunity and disease. Nat Rev Immunol,2003,3:454-462.
    3. Kuchroo VK, Meyers JH, Umetsu DT, et al. Tim family of genes in immunity and tolerance. Adv Immunol,2006,91:227-49.
    4. Kuchroo VK, Dardalhon V, Xiao S, et al. New roles for Tim family members in immune regulation. Nat Rev Immunol,2008,8:577-580.
    5. Degauque N, Mariat C, Kenny J, et al. Immunostimulatory Timl-specific antibody deprograms Tregs and prevents transplant tolerance in mice. J Clin Invest,2008, 118:735-741.
    6. Santiago C, Ballesteros A, Martinez-MuIoz L, et al. Structures of T cell immunoglobulin mucin protein 4 show a metal-Ion-dependent ligand binding site where phosphatidylserine binds. Immunity,2007,27(6):941-51.
    7. Hafler DA, Kuchroo VJ. Tims:central regulators of immune responses. J. Exp. Med, 2008,205(12),2699-2701.
    8. Ichimura T, Bonventre JV, Sanicola M, et al. Kidney injury molecule-1 (KIM-1), a putative epithelial cell adhesion molecule containing a novel immunoglobulin domain, is up-regulated in renal cells after injury. J Biol Chem,1998,273 (7):4135-4142.
    9. Kaplan G, Totsuka A, Thompson P, et al. Identification of a surface glycoprotein on African green monkey kidney cells as a receptor for hepatitis A virus. EMBO J,1996, 15(16):4282-4296.
    lO.Umetsu SE, Lee WL, McIntire JJ, et al. Tim-1 induces T cell activation and inhibits the development of peripheral tolerance. Nature Immunol,2005,6:447-454.
    11.Meyers JH, Chakravarti S, Schlesinger D, et al. Tim-4 is the ligand for Timl and the Tim-1-Tim-4 interaction regulates T cell proliferation. Nat Immunol,2005,6(5): 455-64.
    12. Nakae S, Iwakura Y, Suto H, et al. Phenotypic differences between Th1 and Th17 cells and negative regulation of Th1 cell differentiation by IL-17. J Leukoc Biol,2007, 81(5):1258-68.
    13. Khademi M, Illes Z, Gielen A, et al. T Cell Ig-and mucin-domain-containing molecule-3 (Tim3) and Timl molecules are differentially expressed on human Thl and Th2 cells and in cerebrospinal fluid-derived mononuclear cells in multiple sclerosis. J Immunol,2004,172(11):7169-76.
    14. Kim HS, Kim HS, Lee CW, et al. T cell Ig domain and mucin domain 1 engagement on invariant NKT cells in the presence of TCR stimulation enhances IL-4 production but inhibits IFN-gamma production. J Immunol,2010,184(8):4095-106.
    15. Sizing ID, Bailly V, McCoon P, et al. Epitope-dependent effect of anti-murine Tim1 monoclonal antibodies on T cell activity and lung immune responses. J Immunol,2007, 178:2249-2261.
    16. de Souza A, Oak J, Jordanhazy R, et al. Timl-Mediated T Cell Activation Requires Recruitment and Activation of PI3-Kinase. J Immunol,2008,80(10):6518-6526.
    17. de Souza AJ, Oriss TB, O'Malley KJ, Ray, A.& Kane, L. P. T cell Ig and mucin 1 (Tim1) is expressed on in vivo-activated T cells and provides a costimulatory signal for T cell activation. Proc. Natl. Acad. Sci. USA,2005,102,17113-17118.
    18. Li XC, Rothstein DM, Sayegh MH. Costimulatory pathways in transplantation: challenges and new developments. Immunol Rev,2009,229(1):271-93.
    19. Mariat C, Degauque N, Strom TB. Timl:a new player in transplant immunity. Transplantation,2009,87(9):84-6.
    20. Xiao S, Najafian N, Reddy J, et al. Differential engagement of Tim-1 during activation can positively or negatively costimulate T cell expansion and effector function. J. Exp. Med,2007,204,1691-1702.
    21. Ueno T, Habicht A, Clarkson MR, et al. The emerging role of T cell Ig mucin 1 in alloimmune responses in an experimental mouse transplant model. J. Clin. Invest,2008, 118,742-751.
    22. Yuan X, Ansari MJ, D'Addio F, et al. Targeting Timl to overcome resistance to transplantation tolerance mediated by CD8+T17 cells. Proc Natl Acad Sci USA,2009, 106(26):10734-9.
    23. Binne L, Scott ML, Rennert PD. Human Timl associates with the TCR complex and up-regulates T cell activation signals. J. Immunol,2007,178,4342-4350.
    24. Chakravarti S, Sabatos CA, Xiao S, et al. Tim2 regulates T helper type 2 responses and autoimmunity. J Exp Med,2005,202:437-444
    25. Chen TT, Li L, Chung DH, et al. Tim2 is expressed on B cells and in liver and kidney and is a receptor for H-ferritin endocytosis. J Exp Med,2005,202 (7):955-965.
    26. Chakravarti S, Sabatos CA, Xiao S, et al. Tim2 regulates T helper type 2 responses and autoimmunity. J Exp Med,2005,202 (3):437-444.
    27. Kumanogoh A, Marukawa S, Suzuki K, et al. Class Ⅳ semaphorin Sema4A enhances T cell activation and interacts with Tim2. Nature,2002,419,629-633.
    28. Kumanogoh A, Shikina T, Suzuki K, et al. Nonredundant roles of Sema4A in the immune system:defective T cell priming and Thl/Th2 regulation in Sema4A-deficient mice. Immunity,2005,22,305-316.
    29. Fukushima A, Sumi T, Fukuda K, et al. T-cell Ig and mucin domain-containing protein (Tim)-2 regulates murine allergic conjunctivitis during the effector phase. Immunol Lett,2007,110(2):133-8.
    30. Rennert PD, Ichimura T, Sizing ID, et al. T Cell, Ig Domain, Mucin Domain-2 Gene-Deficient Mice Reveal a Novel Mechanism for the Regulation of Th2 Immune Responses and Airway Inflammation. J Immunol,2006,177,4311-4321.
    31. Monney L, Sabatos CA, Gaglia JL, et al. Thl-specific cell surface protein Tim3 regulates macrophage activation and severity of an autoimmune disease. Nature, 2002,415,536-541.
    32. Monney L, Sabato S, Catherine A, et al. Th2 specific cell surface protein Tim3 regulates macrophage activation and severity of an autoimmune disease. Nature,2002, 415(6871):536-541.
    33. Sanchez-Fueyo A, Tian J, Picatella D, et al. Tim3 inhibits T helper type 1-mediated auto-and alloimmune responses and promotes immunological tolerance. Nat Immunol, 2003,4(11):1093-1101.
    34. Sabatos CA, Sumone CK, Cha E, et al. Interaction of Tim3 and Tim3 ligand regulates T helper type 1 responses and induction of peripheral tolerance. Nat Immunol,2003, 4(11):1102-1110.
    35. Anderson AC, Anderson DE, Bregoli Lisa, et al. Promotion of tissue inflammation by the immune receptor Tim3 expressed on innate immune cells. Science,2007,318, 1141-1143.
    36. Koguchi K, Anderson DE, Yang L, et al. Dysregulated T cell expression of Tim3 in multiple sclerosis. J Exp Med,2006,203,1413-1418.
    37. Yang L, Anderson DE, Kuchroo J, et al. Lack of Tim3 immunoregulation in multiple sclerosis. J. Immunol,2008,180,4409-4414.
    38. Zhu C, Anderson AC, Schubart A, et al. The Tim3 ligand Galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol,2005,6:1245-1252
    39. Su EW, Lin JY, Kane LP, et al. Timl and Tim3 proteins in immune regulation. Cytokine,2008,44(1):9-13.
    40. Wada J, Kanwar YS. Identification and characterization of Galectin-9, a novel P-galactoside-binding mammalian lectin. J Bio Chem,1997,272:6078-6086.
    41.41. Rabinovich GA, Baum LG, Tinari N, et al. Galectins and their ligands:amplifiers, silencers or tuners of the inflammatory response. Trends Immunol,2002,23 (6):3132320.
    42. Seki M, Oomizu S, Sakata KM, et al. Galectin-9 suppresses the generation of Th17, promotes the induction of regulatory T cells, and regulates experimental autoimmune arthritis. Clin Immunol,2008,127:78.
    43. Meyers JH, Sabatos CA, Chakravarti S, et al. The Tim gene family regulates autoimmune and allergic diseases. Trends Mol Med,2005, 11(8):362-369.
    44. Anderson AC, Anderson DE, Bregoli L, et al. Promotion of Tissue Inflammation by the Immune Receptor Tim3 Expressed on Innate Immune Cells. Science,2007, 318:1141-1143.
    45. Nagahara K, Arikawa T, Oomizu S, et al. Galectin-9 Increases Tim3+Dendritic Cells and CD8+T Cells and Enhances Antitumor Immunity via Galectin-9-Tim3 Interactions. J Immunol,2008,181:7660-7669.
    46. Rodriguez-Manzanet R, Meyers JH, Balasubramanian S, et al. Tim4 expressed on APCs induces T cell expansion and survival. J Immunol,2008,180:4706-4713.
    47. Meyers JH, Chakravarti S, Schlesinger D, et al. Tim4 is the ligand for Tim1, and the Timl-Tim4 interaction regulates T cell proliferation. Nature Immunol,2005,6, 455-464.
    48. de Souza AJ, Oriss TB, O'malley KJ, et al. T cell Ig and mucin 1 (Tim1) is expressed on in vivo activated T cells and provides a costimulatory signal for T cell activation. Proc Natl Acad Sci USA,2005,102 (47):17113-17118.
    49. Cao E, Zang X, Ramagopal U, et al. T cell immunoglobulin mucin-3 crystal structure reveals a novel ligand binding surface. Immunity,2007,26,311-321.
    50. Santiago C, Ballesteros A, Tami C, et al. Structures of T cell immunoglobulin mucin receptors 1 and 2 reveal mechanisms for regulation of immune responses by the Tim receptor family. Immunity,2007,26,299-310.
    51. Miyanishi M, Tada K, Koike M, et al. Identification of Tim4 as a phosphatidylserine receptor. Nature,2007,450,435-439.
    52. Kobayashi N, Karisola P, Pena-Cruz V, et al. Timl and Tim4 glycoproteins bind phosphatidylserine and mediate uptake of apoptotic cells. Immunity,2007,27(6): 927-40.
    53. Savill J, Gregory C. Apoptotic PS to phagocyte Tim4:Eat me. Immunity,2007,27 (6): 830-832.
    54. Rodriguez-Manzanet R, Sanjuan MA, Wu HY, et al. T and B cell hyperactivity and autoimmunity associated with niche-specific defects in apoptotic body clearance in Tim4-deficient mice. Proc Natl Acad Sci USA,2010, Apr 5. [Epub ahead of print]

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700